1
|
Zavidić T, Babarović E, Drvar V, Ćurko-Cofek B, Laškarin G. Patients with Higher Pulse Wave Velocity Are More Likely to Develop a More Severe Form of Knee Osteoarthritis: Implications for Cardiovascular Risk. Biomedicines 2025; 13:1208. [PMID: 40427035 PMCID: PMC12109211 DOI: 10.3390/biomedicines13051208] [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: 03/23/2025] [Revised: 05/09/2025] [Accepted: 05/10/2025] [Indexed: 05/29/2025] Open
Abstract
Background/Objectives: Knee osteoarthritis (KOA) is a progressive degenerative joint disease characterised by low-grade inflammation and is associated with increased cardiovascular (CV) risk and arterial stiffness. Pulse wave velocity (PWV) is a quantitative measure of arterial stiffness and an important tool for detecting subclinical arterial calcification and CV risk. This study aimed to determine whether PWV can distinguish radiographically mild KOA (Kellgren-Lawrence grades 1-2) from severe KAO (Kellgren-Lawrence grades 3-4) in terms of CV risk factors. Methods: A total of 223 postmenopausal women with KOA participated in this cross-sectional study. Assessments included anthropometry, laboratory analyses, blood pressure and PWV measurements, a 6 min walk test, pain evaluation using a visual analogue scale (VAS), and completion of the International Physical Activity Questionnaire (IPAQ). Results: PWV was significantly higher in the severe KOA group (10.53 m/s vs. 8.78 m/s, p < 0.001). A cut-off value of 8.4 m/s effectively distinguished between severe and mild forms of KOA (AUC = 0.798, p = 0.001). OA grade, pain, age, waist circumference, WHR, SCORE 2/SCORE 2OP, systolic blood pressure, serum glucose, HbA1c, uric acid, creatinine, and erythrocyte sedimentation rate were increased in the group with PWV > 8.4 m/s, compared to the group with PWV ≤ 8.4 m/s. Conversely, eGFR, the 6 min walk test and physical activity of patients were reduced in the group with PWV > 8.4 m/s. A patient with a PWV > 8.4 m/s has a 1.77 times higher chance of developing a more severe form of the disease than a patient with a lower PWV. Conclusions: Patients with a higher PWV are more likely to develop a more severe form of KOA, which is associated with increased cardiovascular risk.
Collapse
Affiliation(s)
- Tina Zavidić
- Department of Family Medicine, Faculty of Medicine, University of Rijeka, B. Branchetta 20, 51000 Rijeka, Croatia
- Istrian Helath Centers, J. Dobrile 1, 52000 Pazin, Croatia
| | - Emina Babarović
- Department of Pathology, Faculty of Medicine, University of Rijeka, B. Branchetta 20, 51000 Rijeka, Croatia;
| | - Vedrana Drvar
- Clinical Institute of Laboratory Diagnostics, University Hospital Centre Rijeka, V. Dukića 7, 51000 Rijeka, Croatia;
| | - Božena Ćurko-Cofek
- Department of Physiology, Immunology and Pathophysiology, Faculty of Medicine, University of Rijeka, B. Branchetta 20, 51000 Rijeka, Croatia; (B.Ć.-C.); (G.L.)
| | - Gordana Laškarin
- Department of Physiology, Immunology and Pathophysiology, Faculty of Medicine, University of Rijeka, B. Branchetta 20, 51000 Rijeka, Croatia; (B.Ć.-C.); (G.L.)
- Hospital for Medical Rehabilitation of the Heart and Lung Diseases and Rheumatism “Thalassotherapia Opatija”, M. Tita 188, 51412 Opatija, Croatia
| |
Collapse
|
2
|
An T, Guo M, Wang Z, Liu K. Tissue-Resident Macrophages in Cardiovascular Diseases: Heterogeneity and Therapeutic Potential. Int J Mol Sci 2025; 26:4524. [PMID: 40429668 PMCID: PMC12111180 DOI: 10.3390/ijms26104524] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2025] [Revised: 05/05/2025] [Accepted: 05/07/2025] [Indexed: 05/29/2025] Open
Abstract
Tissue-resident macrophages (TRMs) play a crucial role in maintaining tissue homeostasis and regulating immune responses. In recent years, an increasing number of studies have highlighted their central role in cardiovascular diseases. This review provides a comprehensive overview of TRMs, with a particular emphasis on cardiac resident macrophages (CRMs), discussing their origin, heterogeneity, and functions in various cardiovascular diseases. We conduct an in-depth analysis of macrophage subpopulations based on C-C Chemokine Receptor Type 2 (CCR2) receptor expression, elucidating the role of CCR2+ macrophages in promoting fibrosis and cardiac remodeling, while highlighting the protective functions of CCR2- macrophages in suppressing inflammation and promoting tissue repair. In atherosclerosis, we focus on the role of metabolic reprogramming in regulating macrophage polarization, revealing how metabolic pathways influence the balance between pro-inflammatory M1 and anti-inflammatory M2 macrophages, thereby affecting plaque stability and disease progression. By summarizing the roles of these macrophage subpopulations in myocardial infarction, heart failure, and other diseases, we propose potential therapeutic strategies aimed at modulating different macrophage subtypes. These include targeting the CCR2 signaling pathway to mitigate inflammation and fibrosis, and metabolic reprogramming to restore the balance between M1 and M2 macrophages. Finally, we highlight the need for future research to focus on the functional diversity and molecular mechanisms of human TRMs to develop novel immunotherapeutic strategies and improve the prognosis of cardiovascular diseases.
Collapse
Affiliation(s)
- Tianhui An
- Department of Geriatrics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China;
| | - Mengyuan Guo
- Department of Cardiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China;
| | - Zhaohui Wang
- Department of Geriatrics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China;
| | - Kun Liu
- Department of Cardiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China;
| |
Collapse
|
3
|
Hao D, McBride MA, Bohannon JK, Hernandez A, Klein B, Williams DL, Sherwood ER. Metabolic adaptations driving innate immune memory: mechanisms and therapeutic implications. J Leukoc Biol 2025; 117:qiaf037. [PMID: 40138361 DOI: 10.1093/jleuko/qiaf037] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2024] [Revised: 03/12/2025] [Accepted: 03/24/2025] [Indexed: 03/29/2025] Open
Abstract
Immune memory is a hallmark of the adaptive immune system. However, recent research reveals that innate immune cells also retain memory of prior pathogen exposure that prompts enhanced responses to subsequent infections. This phenomenon is termed "innate immune memory" or "trained immunity." Notably, remodeling of cellular metabolism, which closely links to epigenetic reprograming, is a prominent feature of innate immune memory. Adaptations in glycolysis, the tricarboxylic acid cycle, oxidative phosphorylation, glutaminolysis, and lipid synthesis pathways are critical for establishing innate immune memory. This review provides an overview of the current understanding of how metabolic adaptations drive innate immune memory. This understanding is fundamental to understanding innate immune system functions and advancing therapies against infectious diseases.
Collapse
Affiliation(s)
- Dan Hao
- Department of Anesthesiology, Vanderbilt University Medical Center, 1211 Medical Center Drive, Nashville, TN 37232, United States
| | - Margaret A McBride
- Department of Pathology, Microbiology and Immunology, Vanderbilt University Medical Center, 1211 Medical Center Drive, Nashville, TN 37232, United States
| | - Julia K Bohannon
- Department of Anesthesiology, Vanderbilt University Medical Center, 1211 Medical Center Drive, Nashville, TN 37232, United States
- Department of Pathology, Microbiology and Immunology, Vanderbilt University Medical Center, 1211 Medical Center Drive, Nashville, TN 37232, United States
| | - Antonio Hernandez
- Department of Anesthesiology, Vanderbilt University Medical Center, 1211 Medical Center Drive, Nashville, TN 37232, United States
| | - Benjamin Klein
- Department of Anesthesiology, Vanderbilt University Medical Center, 1211 Medical Center Drive, Nashville, TN 37232, United States
| | - David L Williams
- Department of Surgery, East Tennessee State University, Quillen College of Medicine, P.O. Box 70575, Johnson City, TN 37614, United States
- Center for Inflammation, Infectious Disease and Immunology, Quillen College of Medicine, 1276 Gilbreath Drive, Johnson City, TN 37614, United States
| | - Edward R Sherwood
- Department of Anesthesiology, Vanderbilt University Medical Center, 1211 Medical Center Drive, Nashville, TN 37232, United States
- Department of Pathology, Microbiology and Immunology, Vanderbilt University Medical Center, 1211 Medical Center Drive, Nashville, TN 37232, United States
- Department of Surgery, East Tennessee State University, Quillen College of Medicine, P.O. Box 70575, Johnson City, TN 37614, United States
- Center for Inflammation, Infectious Disease and Immunology, Quillen College of Medicine, 1276 Gilbreath Drive, Johnson City, TN 37614, United States
| |
Collapse
|
4
|
Liu D, Fan X, Wang J, Weng R, Tu J, Wang J, Ning X, Zhao Y. Association Between NLR, MLR and Stroke Incidence, All-Cause Mortality Among Low-Income Aging Populations: A Prospective Cohort Study. J Inflamm Res 2025; 18:5715-5726. [PMID: 40322532 PMCID: PMC12048293 DOI: 10.2147/jir.s513811] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2024] [Accepted: 04/16/2025] [Indexed: 05/08/2025] Open
Abstract
Objective This study aimed to assess the association of the Neutrophil-to-Lymphocyte Ratio (NLR) and Monocyte-to-Lymphocyte Ratio (MLR) in predicting stroke incidence and all-cause mortality in low-income elderly populations. Methods This prospective cohort study included participants who were middle-aged or elderly individuals from a low-income population in China. Participants were selected into the cohort and complete baseline assessments, which included questionnaire surveys, physical examinations, blood tests, and carotid artery ultrasound evaluations. Cox proportional hazards regression analysis was used to assess the associations of the NLR and MLR with the incidence of stroke and all-cause mortality. The predictive performance of the model was evaluated using the area under the receiver operating characteristic curve (AUC-ROC). Results A total of 3948 participants were enrolled in the study. Over a median follow-up period of 7 years, 262 participants experienced stroke events and 227 participants died. After adjusting for potential confounding variables, the final model revealed that a higher NLR was significantly associated with an increased risk of stroke (HR: 1.776, 95% CI: 1.250-2.254, P = 0.001) and all-cause mortality (HR: 1.558, 95% CI: 1.148-2.116, P = 0.004). Furthermore, a higher MLR was found to be associated with an increased risk of all-cause mortality (HR: 1.397, 95% CI: 1.054-1.852, P = 0.020), but no significant association was observed between MLR and stroke incidence. ROC analysis revealed that the AUC for NLR in predicting stroke was 0.55 (95% CI: 0.52-0.59, P=0.005), while the AUC for MLR was 0.58 (95% CI: 0.54-0.62, P<0.001). Similarly, the AUC for NLR in predicting all-cause mortality was 0.57 (95% CI: 0.53-0.61, P<0.001), and the AUC for MLR was 0.61 (95% CI: 0.57-0.65, P<0.001). Conclusion These findings indicate that NLR is associated with an increased risk of stroke and all-cause mortality, while higher MLR is associated with all-cause mortality but not with stroke incidence. However, the modest predictive performance of both markers suggests that their clinical utility remains limited. Further research is needed to validate these associations and explore their potential role in comprehensive risk assessment models.
Collapse
Affiliation(s)
- Dongjing Liu
- Department of Science and Education, Shenzhen Third People’s Hospital and The Second Hospital Affiliated with The Southern University of Science and Technology, Shenzhen, Guangdong, People’s Republic of China
| | - Xiaonan Fan
- National Clinical Research Center for Infectious Disease, Shenzhen Third People’s Hospital and The Second Hospital Affiliated with The Southern University of Science and Technology, Shenzhen, Guangdong, People’s Republic of China
| | - Junwei Wang
- Department of Cardiology, Shenzhen Third People’s Hospital and The Second Hospital Affiliated with The Southern University of Science and Technology, Shenzhen, Guangdong, People’s Republic of China
| | - Ruihui Weng
- Department of Neurology, Shenzhen Third People’s Hospital and The Second Hospital Affiliated with The Southern University of Science and Technology, Shenzhen, Guangdong, People’s Republic of China
| | - Jun Tu
- Department of Neurology, Tianjin Medical University General Hospital, Tianjin, 300052, People’s Republic of China
- Laboratory of Epidemiology, Tianjin Neurological Institute, Tianjin, 300052, People’s Republic of China
- Tianjin Neurological Institute, Key Laboratory of Post-Neuroinjury Neuro-Repair and Regeneration in Central Nervous System, Ministry of Education and Tianjin City, Tianjin, 300052, People’s Republic of China
| | - Jinghua Wang
- Department of Neurology, Tianjin Medical University General Hospital, Tianjin, 300052, People’s Republic of China
- Laboratory of Epidemiology, Tianjin Neurological Institute, Tianjin, 300052, People’s Republic of China
- Tianjin Neurological Institute, Key Laboratory of Post-Neuroinjury Neuro-Repair and Regeneration in Central Nervous System, Ministry of Education and Tianjin City, Tianjin, 300052, People’s Republic of China
- Center of Clinical Epidemiology, Shenzhen Third People’s Hospital and The Second Hospital Affiliated with The Southern University of Science and Technology, Shenzhen, Guangdong, People’s Republic of China
| | - Xianjia Ning
- Department of Neurology, Tianjin Medical University General Hospital, Tianjin, 300052, People’s Republic of China
- Laboratory of Epidemiology, Tianjin Neurological Institute, Tianjin, 300052, People’s Republic of China
- Tianjin Neurological Institute, Key Laboratory of Post-Neuroinjury Neuro-Repair and Regeneration in Central Nervous System, Ministry of Education and Tianjin City, Tianjin, 300052, People’s Republic of China
- Center of Clinical Epidemiology, Shenzhen Third People’s Hospital and The Second Hospital Affiliated with The Southern University of Science and Technology, Shenzhen, Guangdong, People’s Republic of China
| | - Yu Zhao
- Department of Neurology, Shenzhen Third People’s Hospital and The Second Hospital Affiliated with The Southern University of Science and Technology, Shenzhen, Guangdong, People’s Republic of China
| |
Collapse
|
5
|
Zhang J, Xu Y, Han Z, Liu B, Wang M, Bao L, He Y. LACC1 Enhances Polyamine Immunometabolism in Inflammatory Macrophages to Inhibit Atherosclerosis Progression. J Cardiovasc Transl Res 2025:10.1007/s12265-024-10585-9. [PMID: 40293654 DOI: 10.1007/s12265-024-10585-9] [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: 04/15/2024] [Accepted: 11/25/2024] [Indexed: 04/30/2025]
Abstract
To explore the function and potential mechanism of laccase domain-containing 1 (LACC1) on atherosclerosis (AS). ApoE-/- mice feed with high-fat diet (HFD) were injected with adenovirus shLACC1 (Ad-shLACC1) or Ad-shNC via tail vein. LACC1 was highly expressed in macrophages of atherosclerotic plaque in ApoE-/- mice and ox-LDL-treated Raw264.7 macrophages. LACC1 silencing enhanced AS development and facilitated inflammation in mice. Then, we found that LACC1 silencing facilitated inflammation but repressed polyamine immunometabolism in ox-LDL-treated Raw264.7 macrophages. Through rescue experiments using ornithine or ODC1 inhibitor (DFMO), we further confirmed that LACC1 promoted polyamine immunometabolism to inhibit inflammation in ox-LDL-treated Raw264.7 macrophages. In addition, the observed LACC1 function was dependent on NOS2. In conclusion, we proved that the downregulation of LACC1 promoted AS progression via inhibiting polyamine immunometabolism in inflammatory macrophages, suggesting LACC1 may be a potential therapeutic target for AS.
Collapse
Affiliation(s)
- Jingyong Zhang
- Department of Vascular Surgery, Shandong Provincial Hospital Affiliated to Shandong First Medical University, No.324, Jing five and Wei seven Road, Jinan, 250021, Shandong, China
| | - Yuan Xu
- Department of Traditional Chinese Medicine, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, 250021, Shandong, China
| | - Zonglin Han
- Department of Vascular Surgery, Shandong Provincial Hospital Affiliated to Shandong First Medical University, No.324, Jing five and Wei seven Road, Jinan, 250021, Shandong, China
| | - Bingqi Liu
- Department of Vascular Surgery, Shandong Provincial Hospital Affiliated to Shandong First Medical University, No.324, Jing five and Wei seven Road, Jinan, 250021, Shandong, China
| | - Maohua Wang
- Department of Vascular Surgery, Shandong Provincial Hospital Affiliated to Shandong First Medical University, No.324, Jing five and Wei seven Road, Jinan, 250021, Shandong, China
| | - Lili Bao
- Department of Vascular Surgery, Shandong Provincial Hospital Affiliated to Shandong First Medical University, No.324, Jing five and Wei seven Road, Jinan, 250021, Shandong, China
| | - Yuxiang He
- Department of Vascular Surgery, Shandong Provincial Hospital Affiliated to Shandong First Medical University, No.324, Jing five and Wei seven Road, Jinan, 250021, Shandong, China.
| |
Collapse
|
6
|
Jiang F, Tu J, Luo W, Jia Y, Luo Q, Ye J. Identification of circulating metabolites associated with chronic rhinosinusitis using Mendelian randomization analysis. Braz J Otorhinolaryngol 2025; 91:101626. [PMID: 40286593 PMCID: PMC12056403 DOI: 10.1016/j.bjorl.2025.101626] [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: 09/18/2024] [Revised: 02/17/2025] [Accepted: 03/10/2025] [Indexed: 04/29/2025] Open
Abstract
OBJECTIVE This study aims to employ Mendelian Randomization (MR) analysis to investigate causal relationships between serum metabolites and CRS, identifying key pathogenic and protective factors and analyzing their mechanisms of action. METHODS Utilizing data from the Genome-Wide Association Studies (GWAS) database, employing two-sample MR analysis to investigate the potential causal relationship between 233 circulating metabolites with the occurrence of CRS. Inverse Variance Weighted (IVW) model, MR-Egger method, Weighted Median, and Weighted model were employed. Sensitivity analyses were conducted with Bonferroni correction. This research aims to elucidate the impact of metabolites on the development and progression of CRS, providing valuable insights into the underlying mechanisms. RESULTS Following MR analysis, two metabolites were significantly associated with CRS: Tyrosine (OR = 1.223; 95% CI 1.115-1.341; p = 1.96E-05) and Creatinine (OR = 1.208; 95% CI 1.103-1.322; p = 4.11E-05). These two key risk factors may be further studied for their pathogenesis and could be targeted for modulation in the treatment of CRS. However, there are several protective factors also worth exploring, among which the correlation is more significant: Ratio of conjugated linoleic acid to total fatty acids (OR = 0.809; 95% CI 0.708‒0.923; p = 1.73E-03), Albumin (OR = 0.787; 95% CI 0.670‒0.926; p = 3.76E-03),Conjugated linoleic acid (OR = 0.664; 95% CI 0.491‒0.898; p = 7.85E-03), Diacylglycerol (OR = 0.804; 95% CI 0.654‒0.989; p = 3.87E-02), Apolipoprotein A-I (OR = 0.915; 95% CI 0.845‒0.991; p = 2.89E-02). CONCLUSION In our MR study, we discovered 28 circulating metabolites linked to CRS. Importantly, tyrosine and creatinine were identified as the most significant contributors to the pathogenesis of CRS, highlighting their potential as therapeutic targets. Additionally, several protective factors may offer new avenues for preventive strategies and therapeutic interventions. These findings underscore the clinical relevance of targeting these metabolites to modulate CRS progression and improve patient outcomes. LEVEL OF EVIDENCE Level 2*.1.
Collapse
Affiliation(s)
- Fan Jiang
- Nanchang University, Jiangxi Medical College, The First Affiliated Hospital, Department of Otorhinolaryngology, Head and Neck Surgery, Nanchang, Jiangxi Province, China
| | - Junhao Tu
- Nanchang University, Jiangxi Medical College, The First Affiliated Hospital, Department of Otorhinolaryngology, Head and Neck Surgery, Nanchang, Jiangxi Province, China
| | - Wenqi Luo
- Nanchang University, Jiangxi Medical College, The First Affiliated Hospital, Department of Otorhinolaryngology, Head and Neck Surgery, Nanchang, Jiangxi Province, China
| | - Yizhen Jia
- Nanchang University, Jiangxi Medical College, The First Affiliated Hospital, Department of Otorhinolaryngology, Head and Neck Surgery, Nanchang, Jiangxi Province, China
| | - Qing Luo
- Nanchang University, Jiangxi Medical College, The First Affiliated Hospital, Department of Otorhinolaryngology, Head and Neck Surgery, Nanchang, Jiangxi Province, China; Nanchang University, Jiangxi Medical College, The First Affiliated Hospital, Department of Allergy, Nanchang, Jiangxi Province, China
| | - Jing Ye
- Nanchang University, Jiangxi Medical College, The First Affiliated Hospital, Department of Otorhinolaryngology, Head and Neck Surgery, Nanchang, Jiangxi Province, China; Jiangxi Medicine Academy of Nutrition and Health Management, Nanchang, Jiangxi Province, China; Nanchang University, Jiangxi Medical College, The First Affiliated Hospital, Department of Allergy, Nanchang, Jiangxi Province, China; Nanchang University, Jiangxi Medical College, The First Affiliated Hospital, Institute of Otorhinolaryngology, Nanchang, Jiangxi Province, China.
| |
Collapse
|
7
|
Araya-Sapag MJ, Lara-Barba E, García-Guerrero C, Herrera-Luna Y, Flores-Elías Y, Bustamante-Barrientos FA, Albornoz GG, Contreras-Fuentes C, Yantén-Fuentes L, Luque-Campos N, Vega-Letter AM, Toledo J, Luz-Crawford P. New mesenchymal stem/stromal cell-based strategies for osteoarthritis treatment: targeting macrophage-mediated inflammation to restore joint homeostasis. J Mol Med (Berl) 2025:10.1007/s00109-025-02547-8. [PMID: 40272537 DOI: 10.1007/s00109-025-02547-8] [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: 05/27/2024] [Revised: 04/10/2025] [Accepted: 04/14/2025] [Indexed: 04/25/2025]
Abstract
Macrophages are pivotal in osteoarthritis (OA) pathogenesis, as their dysregulated polarization can contribute to chronic inflammatory processes. This review explores the molecular and metabolic mechanisms that influence macrophage polarization and identifies potential strategies for OA treatment. Currently, non-surgical treatments for OA focus only on symptom management, and their efficacy is limited; thus, mesenchymal stem/stromal cells (MSCs) have gained attention for their anti-inflammatory and immunomodulatory capabilities. Emerging evidence suggests that small extracellular vesicles (sEVs) derived from MSCs can modulate macrophage function, thus offering potential therapeutic benefits in OA. Additionally, the transfer of mitochondria from MSCs to macrophages has shown promise in enhancing mitochondrial functionality and steering macrophages toward an anti-inflammatory M2-like phenotype. While further research is needed to confirm these findings, MSC-based strategies, including the use of sEVs and mitochondrial transfer, hold great promise for the treatment of OA and other chronic inflammatory diseases.
Collapse
Affiliation(s)
- María Jesús Araya-Sapag
- Programa de Doctorado en Biomedicina, Facultad de Medicina, Universidad de los Andes, Santiago, Chile
- Laboratorio de Inmunología Celular y Molecular, Centro de Investigación e Innovación Biomédica, Facultad de Medicina, Universidad de los Andes, Santiago, Chile
- IMPACT, Center of Interventional Medicine for Precision and Advanced Cellular Therapy, Santiago, Chile
| | - Eliana Lara-Barba
- Programa de Doctorado en Biomedicina, Facultad de Medicina, Universidad de los Andes, Santiago, Chile
- Laboratorio de Inmunología Celular y Molecular, Centro de Investigación e Innovación Biomédica, Facultad de Medicina, Universidad de los Andes, Santiago, Chile
- IMPACT, Center of Interventional Medicine for Precision and Advanced Cellular Therapy, Santiago, Chile
| | - Cynthia García-Guerrero
- Programa de Doctorado en Biomedicina, Facultad de Medicina, Universidad de los Andes, Santiago, Chile
- Laboratorio de Inmunología Celular y Molecular, Centro de Investigación e Innovación Biomédica, Facultad de Medicina, Universidad de los Andes, Santiago, Chile
- IMPACT, Center of Interventional Medicine for Precision and Advanced Cellular Therapy, Santiago, Chile
| | - Yeimi Herrera-Luna
- Laboratorio de Inmunología Celular y Molecular, Centro de Investigación e Innovación Biomédica, Facultad de Medicina, Universidad de los Andes, Santiago, Chile
- IMPACT, Center of Interventional Medicine for Precision and Advanced Cellular Therapy, Santiago, Chile
| | - Yesenia Flores-Elías
- Laboratorio de Inmunología Celular y Molecular, Centro de Investigación e Innovación Biomédica, Facultad de Medicina, Universidad de los Andes, Santiago, Chile
- IMPACT, Center of Interventional Medicine for Precision and Advanced Cellular Therapy, Santiago, Chile
| | - Felipe A Bustamante-Barrientos
- Laboratorio de Inmunología Celular y Molecular, Centro de Investigación e Innovación Biomédica, Facultad de Medicina, Universidad de los Andes, Santiago, Chile
- IMPACT, Center of Interventional Medicine for Precision and Advanced Cellular Therapy, Santiago, Chile
| | - Guillermo G Albornoz
- Laboratorio de Inmunología Celular y Molecular, Centro de Investigación e Innovación Biomédica, Facultad de Medicina, Universidad de los Andes, Santiago, Chile
- IMPACT, Center of Interventional Medicine for Precision and Advanced Cellular Therapy, Santiago, Chile
| | - Consuelo Contreras-Fuentes
- Laboratorio de Inmunología Celular y Molecular, Centro de Investigación e Innovación Biomédica, Facultad de Medicina, Universidad de los Andes, Santiago, Chile
- IMPACT, Center of Interventional Medicine for Precision and Advanced Cellular Therapy, Santiago, Chile
| | - Liliana Yantén-Fuentes
- Programa de Doctorado en Biomedicina, Facultad de Medicina, Universidad de los Andes, Santiago, Chile
- Laboratorio de Inmunología Celular y Molecular, Centro de Investigación e Innovación Biomédica, Facultad de Medicina, Universidad de los Andes, Santiago, Chile
- IMPACT, Center of Interventional Medicine for Precision and Advanced Cellular Therapy, Santiago, Chile
- Red de Equipamiento Científico Avanzado (REDECA), Facultad de Medicina, Universidad de Chile, Santiago, Chile
| | - Noymar Luque-Campos
- Laboratorio de Inmunología Celular y Molecular, Centro de Investigación e Innovación Biomédica, Facultad de Medicina, Universidad de los Andes, Santiago, Chile
- IMPACT, Center of Interventional Medicine for Precision and Advanced Cellular Therapy, Santiago, Chile
| | - Ana María Vega-Letter
- Escuela de Ingeniería Bioquímica, Pontificia Universidad Católica de Valparaíso, Valparaíso, Chile
| | - Jorge Toledo
- Red de Equipamiento Científico Avanzado (REDECA), Facultad de Medicina, Universidad de Chile, Santiago, Chile.
- Centro de Investigación Clínica Avanzada (CICA), Hospital Clínico Universidad de Chile, Santiago, Chile.
| | - Patricia Luz-Crawford
- Laboratorio de Inmunología Celular y Molecular, Centro de Investigación e Innovación Biomédica, Facultad de Medicina, Universidad de los Andes, Santiago, Chile.
- IMPACT, Center of Interventional Medicine for Precision and Advanced Cellular Therapy, Santiago, Chile.
| |
Collapse
|
8
|
Li M, Sun X, Zeng L, Sun A, Ge J. Metabolic Homeostasis of Immune Cells Modulates Cardiovascular Diseases. RESEARCH (WASHINGTON, D.C.) 2025; 8:0679. [PMID: 40270694 PMCID: PMC12015101 DOI: 10.34133/research.0679] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 02/15/2025] [Revised: 03/20/2025] [Accepted: 03/28/2025] [Indexed: 04/25/2025]
Abstract
Recent investigations into the mechanisms underlying inflammation have highlighted the pivotal role of immune cells in regulating cardiac pathophysiology. Notably, these immune cells modulate cardiac processes through alternations in intracellular metabolism, including glycolysis and oxidative phosphorylation, whereas the extracellular metabolic environment is changed during cardiovascular disease, influencing function of immune cells. This dynamic interaction between immune cells and their metabolic environment has given rise to the novel concept of "immune metabolism". Consequently, both the extracellular and intracellular metabolic environment modulate the equilibrium between anti- and pro-inflammatory responses. This regulatory mechanism subsequently influences the processes of myocardial ischemia, cardiac fibrosis, and cardiac remodeling, ultimately leading to a series of cardiovascular events. This review examines how local microenvironmental and systemic environmental changes induce metabolic reprogramming in immune cells and explores the subsequent effects of aberrant activation or polarization of immune cells in the progression of cardiovascular disease. Finally, we discuss potential therapeutic strategies targeting metabolism to counteract abnormal immune activation.
Collapse
Affiliation(s)
- Mohan Li
- Department of Cardiology, Zhongshan Hospital,
Fudan University, Shanghai Institute of Cardiovascular Diseases, Shanghai 200032, China
- State Key Laboratory of Cardiology, Zhongshan Hospital,
Fudan University, Shanghai 200032, China
- Key Laboratory of Viral Heart Diseases, National Health Commission, Shanghai 200032, China
- Key Laboratory of Viral Heart Diseases,
Chinese Academy of Medical Sciences, Shanghai 200032, China
- National Clinical Research Center for Interventional Medicine, Shanghai 200032, China
| | - Xiaolei Sun
- Department of Cardiology, Zhongshan Hospital,
Fudan University, Shanghai Institute of Cardiovascular Diseases, Shanghai 200032, China
- State Key Laboratory of Cardiology, Zhongshan Hospital,
Fudan University, Shanghai 200032, China
- Key Laboratory of Viral Heart Diseases, National Health Commission, Shanghai 200032, China
- Key Laboratory of Viral Heart Diseases,
Chinese Academy of Medical Sciences, Shanghai 200032, China
- National Clinical Research Center for Interventional Medicine, Shanghai 200032, China
| | - Linqi Zeng
- Department of Cardiology, Zhongshan Hospital,
Fudan University, Shanghai Institute of Cardiovascular Diseases, Shanghai 200032, China
- State Key Laboratory of Cardiology, Zhongshan Hospital,
Fudan University, Shanghai 200032, China
- Key Laboratory of Viral Heart Diseases, National Health Commission, Shanghai 200032, China
- Key Laboratory of Viral Heart Diseases,
Chinese Academy of Medical Sciences, Shanghai 200032, China
- National Clinical Research Center for Interventional Medicine, Shanghai 200032, China
| | - Aijun Sun
- Department of Cardiology, Zhongshan Hospital,
Fudan University, Shanghai Institute of Cardiovascular Diseases, Shanghai 200032, China
- State Key Laboratory of Cardiology, Zhongshan Hospital,
Fudan University, Shanghai 200032, China
- Key Laboratory of Viral Heart Diseases, National Health Commission, Shanghai 200032, China
- Key Laboratory of Viral Heart Diseases,
Chinese Academy of Medical Sciences, Shanghai 200032, China
- National Clinical Research Center for Interventional Medicine, Shanghai 200032, China
- Institutes of Biomedical Sciences,
Fudan University, Shanghai 200032, China
| | - Junbo Ge
- Department of Cardiology, Zhongshan Hospital,
Fudan University, Shanghai Institute of Cardiovascular Diseases, Shanghai 200032, China
- State Key Laboratory of Cardiology, Zhongshan Hospital,
Fudan University, Shanghai 200032, China
- Key Laboratory of Viral Heart Diseases, National Health Commission, Shanghai 200032, China
- Key Laboratory of Viral Heart Diseases,
Chinese Academy of Medical Sciences, Shanghai 200032, China
- National Clinical Research Center for Interventional Medicine, Shanghai 200032, China
- Institutes of Biomedical Sciences,
Fudan University, Shanghai 200032, China
| |
Collapse
|
9
|
Liu R, Dai L, Jia S, Geng S, Niu Y, Chen J, Dong C, Li C, Shi Y, Wang X, Zhang J, Zhao N, Gao Z, Yang X, Gao S. Fut8 regulated Unc5b hyperfucosylation reduces macrophage emigration and accelerates atherosclerosis development via the ferroptosis pathway. Free Radic Biol Med 2025; 235:1-14. [PMID: 40262667 DOI: 10.1016/j.freeradbiomed.2025.04.025] [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: 09/27/2024] [Revised: 04/16/2025] [Accepted: 04/17/2025] [Indexed: 04/24/2025]
Abstract
The accumulation of foam cells in the arterial walls is a defining characteristic of atherosclerosis. Enhancing their migration from plaques may represent a key strategy for slowing disease progression. Recent studies suggest that fucosyltransferase 8 (Fut8) impairs macrophage migration from the intima by modifying the Unc5b membrane receptor, thereby influencing the development of atherosclerosis. This study investigated the roles of Fut8 and Unc5b in foam cell migration using ApoE-/- mouse and foam cell models, employing techniques such as western blotting, mitochondrial function assays, wound healing experiments, and immunofluorescence staining. The findings indicate that Fut8 upregulation increases P53 expression and reduces SLC7A11 and GPX4 levels, leading to altered intracellular concentrations of GSH and Fe2+, impaired mitochondrial function, and reduced migration capacity, all of which promote atherosclerosis. These mechanisms are closely associated with ferroptosis. Intervention with N-acetylcysteine (NAC) and buthionine sulfoximine (BSO) demonstrated that NAC mitigates oxidative stress and migration inhibition, induced by oxidized low-density lipoprotein (ox-LDL). Additionally, inhibiting ferroptosis slowed the progression of atherosclerosis in ApoE-/- mice. Together, these results highlight that Fut8 exacerbates atherosclerosis through a P53/SLC7A11-mediated enhancement of ferroptosis in foam cells, offering a novel perspective on the pathophysiology of atherosclerosis.
Collapse
Affiliation(s)
- Rujin Liu
- Graduate School of Inner Mongolia Medical University, Inner Mongolia Medical University, Hohhot, 010110, PR China; College of Basic Medicine, Inner Mongolia Medical University, Hohhot, 010110, PR China.
| | - Lina Dai
- College of Basic Medicine, Inner Mongolia Medical University, Hohhot, 010110, PR China; Medical Experiments Center, Inner Mongolia Medical University, Hohhot, 010110, PR China.
| | - Sihui Jia
- The Affiliated Hospital of Inner Mongolia Medical University, Hohhot, 010110, PR China; Department of Blood Transfusion, The Affiliated Hospital of Qingdao University, Qingdao, 266000, Shandong, PR China.
| | - Shijia Geng
- College of Basic Medicine, Inner Mongolia Medical University, Hohhot, 010110, PR China; Medical Experiments Center, Inner Mongolia Medical University, Hohhot, 010110, PR China.
| | - Yan Niu
- Medical Experiments Center, Inner Mongolia Medical University, Hohhot, 010110, PR China.
| | - Jie Chen
- College of Basic Medicine, Inner Mongolia Medical University, Hohhot, 010110, PR China; Medical Experiments Center, Inner Mongolia Medical University, Hohhot, 010110, PR China.
| | - Chongyang Dong
- Medical Experiments Center, Inner Mongolia Medical University, Hohhot, 010110, PR China; College of Traditional Chinese Medicine, Inner Mongolia Medical University, Hohhot, 010110, PR China.
| | - Chenlei Li
- Graduate School of Inner Mongolia Medical University, Inner Mongolia Medical University, Hohhot, 010110, PR China; College of Basic Medicine, Inner Mongolia Medical University, Hohhot, 010110, PR China.
| | - Yuanjia Shi
- Graduate School of Inner Mongolia Medical University, Inner Mongolia Medical University, Hohhot, 010110, PR China; College of Basic Medicine, Inner Mongolia Medical University, Hohhot, 010110, PR China.
| | - Xiaomeng Wang
- Graduate School of Inner Mongolia Medical University, Inner Mongolia Medical University, Hohhot, 010110, PR China; College of Basic Medicine, Inner Mongolia Medical University, Hohhot, 010110, PR China.
| | - Jing Zhang
- Graduate School of Inner Mongolia Medical University, Inner Mongolia Medical University, Hohhot, 010110, PR China; College of Basic Medicine, Inner Mongolia Medical University, Hohhot, 010110, PR China.
| | - Ningxia Zhao
- Graduate School of Inner Mongolia Medical University, Inner Mongolia Medical University, Hohhot, 010110, PR China; College of Basic Medicine, Inner Mongolia Medical University, Hohhot, 010110, PR China.
| | - Zhanfeng Gao
- The Affiliated Hospital of Inner Mongolia Medical University, Hohhot, 010110, PR China.
| | - Xi Yang
- College of Basic Medicine, Inner Mongolia Medical University, Hohhot, 010110, PR China; Medical Experiments Center, Inner Mongolia Medical University, Hohhot, 010110, PR China.
| | - Shang Gao
- College of Basic Medicine, Inner Mongolia Medical University, Hohhot, 010110, PR China; Medical Experiments Center, Inner Mongolia Medical University, Hohhot, 010110, PR China.
| |
Collapse
|
10
|
Liu Q, Liu P, Li C, Zhao Z, Wang D, Liu Q, Yang H. Effects of Chinese Medicine on modulating interleukin-17-regulated macrophages in coronary heart disease. Front Pharmacol 2025; 16:1499786. [PMID: 40276600 PMCID: PMC12018881 DOI: 10.3389/fphar.2025.1499786] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2024] [Accepted: 02/10/2025] [Indexed: 04/26/2025] Open
Abstract
Coronary atherosclerotic heart disease (CHD) is one of the leading causes of death from cardiovascular disease worldwide and has significant inflammatory features. Macrophages play an important role in atherosclerotic plaque formation and inflammation. IL-17, as a pro-inflammatory cytokine, further exacerbates the development of CHD by interacting with macrophages. In recent years, there has been increasing evidence that traditional Chinese medicine (CM) has a wide range of applications in regulating the immune system and treating CHD. This article reviewed the role of CM in the regulation of IL-17-regulated macrophages, discussed the core components and targets of CM in the treatment of CHD, and laid a theoretical foundation for its clinical application. The results show that CM can effectively inhibit the formation of foam cells, stabilize vulnerable plaque and delay the progression of atherosclerosis by inhibiting inflammation, regulating the polarization of macrophages and promoting cholesterol outflow. In addition, CM can also regulate the expression and signaling pathway of IL-17, further inhibit inflammatory response and improve the symptoms of CHD, providing a new idea and method for the prevention and treatment of CHD.
Collapse
Affiliation(s)
- Qingqing Liu
- Guangdong Provincial Hospital of Chinese Medicine‐Zhuhai Hospital, State Key Laboratory of Traditional Chinese Medicine Syndrome, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangdong Provincial Hospital of Chinese Medicine, Guangzhou, Guangdong, China
| | - Peizhong Liu
- Guangdong Provincial Hospital of Chinese Medicine‐Zhuhai Hospital, State Key Laboratory of Traditional Chinese Medicine Syndrome, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangdong Provincial Hospital of Chinese Medicine, Guangzhou, Guangdong, China
| | - Chuangpeng Li
- Guangdong Provincial Hospital of Chinese Medicine‐Zhuhai Hospital, State Key Laboratory of Traditional Chinese Medicine Syndrome, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangdong Provincial Hospital of Chinese Medicine, Guangzhou, Guangdong, China
| | - Zhen Zhao
- Guangdong Provincial Hospital of Chinese Medicine‐Zhuhai Hospital, State Key Laboratory of Traditional Chinese Medicine Syndrome, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangdong Provincial Hospital of Chinese Medicine, Guangzhou, Guangdong, China
| | - Dawei Wang
- Guangdong Provincial Hospital of Chinese Medicine‐Zhuhai Hospital, State Key Laboratory of Traditional Chinese Medicine Syndrome, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangdong Provincial Hospital of Chinese Medicine, Guangzhou, Guangdong, China
- State Key Laboratory of Traditional Chinese Medicine Syndrome, The First Affiliated Hospital of Guangzhou University of Traditional Chinese Medicine, Guangzhou, Guangdong, China
| | - Qing Liu
- Guangdong Provincial Hospital of Chinese Medicine‐Zhuhai Hospital, State Key Laboratory of Traditional Chinese Medicine Syndrome, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangdong Provincial Hospital of Chinese Medicine, Guangzhou, Guangdong, China
| | - Huawei Yang
- Guangdong Provincial Hospital of Chinese Medicine‐Zhuhai Hospital, State Key Laboratory of Traditional Chinese Medicine Syndrome, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangdong Provincial Hospital of Chinese Medicine, Guangzhou, Guangdong, China
| |
Collapse
|
11
|
Li H, Ye B, Tian J, Wang B, Zha Y, Zheng S, Ma T, Zhuang W, Park WS, Liang J. Monotropein resists atherosclerosis by reducing inflammation, oxidative stress, and abnormal proliferation and migration of vascular smooth muscle cells. THE KOREAN JOURNAL OF PHYSIOLOGY & PHARMACOLOGY : OFFICIAL JOURNAL OF THE KOREAN PHYSIOLOGICAL SOCIETY AND THE KOREAN SOCIETY OF PHARMACOLOGY 2025; 29:245-255. [PMID: 39972674 PMCID: PMC11842295 DOI: 10.4196/kjpp.24.352] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/29/2024] [Revised: 11/10/2024] [Accepted: 11/13/2024] [Indexed: 02/21/2025]
Abstract
Monotropein is a compound classified into iridoid which is found in herbaceous plants Morindae officinalis. It possesses anti-inflammatory, antioxidant, and anti-osteoarthritic activities. Previous study indicates that monotropein may have the potential to combat cardiovascular disease, although the related mechanism remains unclear. In this study, we constructed the model of atherosclerosis by oxidized low density lipoprotein-induced vascular smooth muscle cells and LDLR-/- mice given high-fat diet to investigate the effects of monotropein on atherosclerosis. Our results showed that monotropein treatment significantly reduced the area of atherosclerotic plaques and necrotic cores in mice, inhibited the proliferation and migration of vascular smooth muscle cells, and reduced inflammatory responses and oxidative stress, which in turn alleviated atherosclerosis. In addition, we found that monotropein reduced the expression levels of P-NF-κB and P-AP-1. In conclusion, our data suggest that monotropein inhibited the proliferation and migration of vascular smooth muscle cells by mediating the activity of NF-κB, AP-1, reducing the level of inflammation and oxidative stress, and thus resisting the development of atherosclerosis. These findings demonstrate the efficacious therapeutic impact of monotropein on atherosclerosis and elucidate its specific target.
Collapse
Affiliation(s)
- Hongliang Li
- Institute of Translational Medicine, Medical College, Yangzhou University, Yangzhou 225001, Jiangsu, China
- Jiangsu Key Laboratory of Experimental & Translational Non-coding RNA Research, Yangzhou University, Yangzhou 225001, Jiangsu, China
| | - Bingqian Ye
- Institute of Translational Medicine, Medical College, Yangzhou University, Yangzhou 225001, Jiangsu, China
| | - Jiping Tian
- Institute of Translational Medicine, Medical College, Yangzhou University, Yangzhou 225001, Jiangsu, China
| | - Bofan Wang
- Institute of Translational Medicine, Medical College, Yangzhou University, Yangzhou 225001, Jiangsu, China
| | - Yiwen Zha
- Institute of Translational Medicine, Medical College, Yangzhou University, Yangzhou 225001, Jiangsu, China
| | - Shuying Zheng
- Institute of Translational Medicine, Medical College, Yangzhou University, Yangzhou 225001, Jiangsu, China
| | - Tan Ma
- Institute of Translational Medicine, Medical College, Yangzhou University, Yangzhou 225001, Jiangsu, China
- Jiangsu Key Laboratory of Experimental & Translational Non-coding RNA Research, Yangzhou University, Yangzhou 225001, Jiangsu, China
| | - Wenwen Zhuang
- Department of Physiology, Kangwon National University School of Medicine, Chuncheon 24341, Korea
| | - Won Sun Park
- Institute of Translational Medicine, Medical College, Yangzhou University, Yangzhou 225001, Jiangsu, China
| | - Jingyan Liang
- Institute of Translational Medicine, Medical College, Yangzhou University, Yangzhou 225001, Jiangsu, China
- Jiangsu Key Laboratory of Experimental & Translational Non-coding RNA Research, Yangzhou University, Yangzhou 225001, Jiangsu, China
| |
Collapse
|
12
|
Qin YS, Yi J, Chen YJ, Zhang W, Tang SF. Recent Advances in Micro/Nanomotor for the Therapy and Diagnosis of Atherosclerosis. ACS APPLIED MATERIALS & INTERFACES 2025; 17:11443-11468. [PMID: 39648908 DOI: 10.1021/acsami.4c15165] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/10/2024]
Abstract
Atherosclerotic cardiovascular disease poses a significant global public health threat with a high incidence that can result in severe mortality and disability. The lack of targeted effects from traditional therapeutic drugs on atherosclerosis may cause damage to other organs and tissues, necessitating the need for a more focused approach to address this dilemma. Micro/nanomotors are self-propelled micro/nanoscale devices capable of converting external energy into autonomous movement, which offers advantages in enhancing penetration depth and retention while increasing contact area with abnormal sites, such as atherosclerotic plaque, inflammation, and thrombosis, within blood vessel walls. Recent studies have demonstrated the crucial role micro/nanomotors play in treating atherosclerotic cardiovascular disease. Hence, this review highlights the recent progress of micro/nanomotor technology in atherosclerotic cardiovascular disease, including the effective promotion of micro/nanomotors in the circulatory system, overcoming hemorheological barriers, targeting the atherosclerotic plaque microenvironment, and targeting intracellular drug delivery, to facilitate atherosclerotic plaque localization and therapy. Furthermore, we also describe the potential application of micro/nanomotors in the imaging of vulnerable plaque. Finally, we discuss key challenges and prospects for treating atherosclerotic cardiovascular disease while emphasizing the importance of designing individualized management strategies specific to its causes and microenvironmental factors.
Collapse
Affiliation(s)
- Yu-Sheng Qin
- Department of Laboratory Medicine, Liuzhou Key Laboratory of Precision Medicine for Viral Diseases, Guangxi Health Commission Key Laboratory of Clinical Biotechnology (Liuzhou People's Hospital), Liuzhou People's Hospital, Liuzhou 545006, China
| | - Juan Yi
- Department of Laboratory Medicine, Liuzhou Traditional Chinese Medical Hospital, The Third Affiliated Hospital of Guangxi University of Chinese Medicine, Liuzhou 545006, China
| | - Yan-Jun Chen
- Department of Pathology, Southwest Hospital, Third Military Medical University, Chongqing 400038, China
| | - Wei Zhang
- Department of Radiology, Liuzhou People's Hospital, Liuzhou 545006, China
| | - Shi-Fu Tang
- Department of Laboratory Medicine, Liuzhou Key Laboratory of Precision Medicine for Viral Diseases, Guangxi Health Commission Key Laboratory of Clinical Biotechnology (Liuzhou People's Hospital), Liuzhou People's Hospital, Liuzhou 545006, China
| |
Collapse
|
13
|
Zhang S, Ji Y, Xu B, Hu D, Zhang X, Song Y, Chen K, Wen Y, He X, Chen Y, Zheng T. Study on the use of black phosphorus quantum dots in the treatment of atherosclerosis. Aging (Albany NY) 2025; 17:563-587. [PMID: 39998897 PMCID: PMC11892921 DOI: 10.18632/aging.206205] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2023] [Accepted: 01/01/2025] [Indexed: 02/27/2025]
Abstract
Atherosclerosis is the pathological basis of cardiovascular disease, and there are no clinical drugs that can safely and efficiently remove atherosclerotic plaques. In this study, black phosphorus quantum dots (BPQDs) were applied to the treatment of atherosclerosis in high fat diet ApoE-/- model mice that BPQDs were given every other day for 3 weeks without changing the high-fat diet. 45.3% atherosclerotic plaque was cleared efficiently within 3 weeks in BPQDs intravenous administration way every other day. The treatment was more effective than traditional statins. The findings suggest that BPQDs have great potential to be applied for clinical prevention and treatment of AS that does not require dietary changes.
Collapse
Affiliation(s)
- Shengwei Zhang
- Shenzhen Key Laboratory for Drug Addiction and Medication Safety, Department of Ultrasound, Institute of Ultrasonic Medicine, Peking University Shenzhen Hospital, Shenzhen Peking University, The Hong Kong University of Science and Technology Medical Center, Shenzhen 518036, Guangdong, P.R. China
- Department of Ultrasound, Xiaolan People’s Hospital of Zhongshan, Zhongshan 528415, Guangdong, P.R. China
| | - Yiran Ji
- Shenzhen Key Laboratory for Drug Addiction and Medication Safety, Department of Ultrasound, Institute of Ultrasonic Medicine, Peking University Shenzhen Hospital, Shenzhen Peking University, The Hong Kong University of Science and Technology Medical Center, Shenzhen 518036, Guangdong, P.R. China
| | - Bingxuan Xu
- Shenzhen Key Laboratory for Drug Addiction and Medication Safety, Department of Ultrasound, Institute of Ultrasonic Medicine, Peking University Shenzhen Hospital, Shenzhen Peking University, The Hong Kong University of Science and Technology Medical Center, Shenzhen 518036, Guangdong, P.R. China
| | - Die Hu
- Shenzhen Key Laboratory for Drug Addiction and Medication Safety, Department of Ultrasound, Institute of Ultrasonic Medicine, Peking University Shenzhen Hospital, Shenzhen Peking University, The Hong Kong University of Science and Technology Medical Center, Shenzhen 518036, Guangdong, P.R. China
| | - Xue Zhang
- Shenzhen Key Laboratory for Drug Addiction and Medication Safety, Department of Ultrasound, Institute of Ultrasonic Medicine, Peking University Shenzhen Hospital, Shenzhen Peking University, The Hong Kong University of Science and Technology Medical Center, Shenzhen 518036, Guangdong, P.R. China
| | - Yujian Song
- Shenzhen Key Laboratory for Drug Addiction and Medication Safety, Department of Ultrasound, Institute of Ultrasonic Medicine, Peking University Shenzhen Hospital, Shenzhen Peking University, The Hong Kong University of Science and Technology Medical Center, Shenzhen 518036, Guangdong, P.R. China
- Ultrasound Diagnosis and Treatment Center of the First People’s Hospital of Foshan, Foshan 528000, Guangdong, P.R. China
| | - Keke Chen
- Shenzhen Key Laboratory for Drug Addiction and Medication Safety, Department of Ultrasound, Institute of Ultrasonic Medicine, Peking University Shenzhen Hospital, Shenzhen Peking University, The Hong Kong University of Science and Technology Medical Center, Shenzhen 518036, Guangdong, P.R. China
- Department of Ultrasound, Nanjing Drum Tower Hospital, Nanjing 210000, Jiangsu, P.R. China
| | - Yilin Wen
- Shenzhen Key Laboratory for Drug Addiction and Medication Safety, Department of Ultrasound, Institute of Ultrasonic Medicine, Peking University Shenzhen Hospital, Shenzhen Peking University, The Hong Kong University of Science and Technology Medical Center, Shenzhen 518036, Guangdong, P.R. China
| | - Xiaoxin He
- Shenzhen Key Laboratory for Drug Addiction and Medication Safety, Department of Ultrasound, Institute of Ultrasonic Medicine, Peking University Shenzhen Hospital, Shenzhen Peking University, The Hong Kong University of Science and Technology Medical Center, Shenzhen 518036, Guangdong, P.R. China
| | - Yun Chen
- Shenzhen Key Laboratory for Drug Addiction and Medication Safety, Department of Ultrasound, Institute of Ultrasonic Medicine, Peking University Shenzhen Hospital, Shenzhen Peking University, The Hong Kong University of Science and Technology Medical Center, Shenzhen 518036, Guangdong, P.R. China
| | - Tingting Zheng
- Shenzhen Key Laboratory for Drug Addiction and Medication Safety, Department of Ultrasound, Institute of Ultrasonic Medicine, Peking University Shenzhen Hospital, Shenzhen Peking University, The Hong Kong University of Science and Technology Medical Center, Shenzhen 518036, Guangdong, P.R. China
| |
Collapse
|
14
|
Wei J, Peng MY, Lu HX. Functional transformation of macrophage mitochondria in cardiovascular diseases. Mol Cell Biochem 2025; 480:747-757. [PMID: 38884847 DOI: 10.1007/s11010-024-05049-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2024] [Accepted: 06/09/2024] [Indexed: 06/18/2024]
Abstract
Mitochondria are pivotal in the modulation of macrophage activation, differentiation, and survival. Furthermore, macrophages are instrumental in the onset and progression of cardiovascular diseases. Hence, it is imperative to investigate the role of mitochondria within macrophages in the context of cardiovascular disease. In this review, we provide an updated description of the origin and classification of cardiac macrophages and also focused on the relationship between macrophages and mitochondria in cardiovascular diseases with respect to (1) proinflammatory or anti-inflammatory macrophages, (2) macrophage apoptosis, (3) macrophage pyroptosis, and (4) macrophage efferocytosis. Clarifying the relationship between mitochondria and macrophages can aid the exploration of novel therapeutic strategies for cardiovascular disease.
Collapse
Affiliation(s)
- Jing Wei
- Department of Laboratory Medicine, Nanjing First Hospital, Nanjng Medical University, Nanjing, 211100, China
| | - Ming-Yu Peng
- Department of Laboratory Medicine, Jiangning Hospital Affiliated to Nanjng Medical University, Nanjing, 211100, China
| | - Hong-Xiang Lu
- Department of Laboratory Medicine, Jiangning Hospital Affiliated to Nanjng Medical University, Nanjing, 211100, China.
- Department of Laboratory Medicine, Nanjing First Hospital, Nanjng Medical University, Nanjing, 211100, China.
| |
Collapse
|
15
|
Wang Y, Wang C, Li J. Neutrophil extracellular traps: a catalyst for atherosclerosis. Mol Cell Biochem 2024; 479:3213-3227. [PMID: 38401035 DOI: 10.1007/s11010-024-04931-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2023] [Accepted: 01/05/2024] [Indexed: 02/26/2024]
Abstract
Neutrophil extracellular traps (NETs) are network-like structures released by activated neutrophils. They consist mainly of double-stranded DNA, histones, and neutrophil granule proteins. Continuous release of NETs in response to external stimuli leads to activation of surrounding platelets and monocytes/macrophages, resulting in damage to endothelial cells (EC) and vascular smooth muscle cells (VSMC). Some clinical trials have demonstrated the association between NETs and the severity and prognosis of atherosclerosis. Furthermore, experimental findings have shed light on the molecular mechanisms by which NETs contribute to atherogenesis. NETs play a significant role in the formation of atherosclerotic plaques. This review focuses on recent advancements in the understanding of the relationship between NETs and atherosclerosis. It explores various aspects, including the formation of NETs in atherosclerosis, clinical trials investigating NET-induced atherosclerosis, the mechanisms by which NETs promote atherogenesis, and the translational implications of NETs. Ultimately, we aim to propose new research directions for the diagnosis and treatment of atherosclerosis.
Collapse
Affiliation(s)
- Yinyu Wang
- Department of Cardiology, Affiliated Hospital of Jiangsu University, Zhenjiang, China
| | - Cuiping Wang
- Department of Cardiology, Affiliated Hospital of Jiangsu University, Zhenjiang, China.
| | - Jiayan Li
- Department of Cardiology, Affiliated Hospital of Jiangsu University, Zhenjiang, China
| |
Collapse
|
16
|
Li W, Huang Y, Liu J, Zhou Y, Sun H, Fan Y, Liu F. Defective macrophage efferocytosis in advanced atherosclerotic plaque and mitochondrial therapy. Life Sci 2024; 359:123204. [PMID: 39491771 DOI: 10.1016/j.lfs.2024.123204] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2024] [Revised: 10/02/2024] [Accepted: 10/30/2024] [Indexed: 11/05/2024]
Abstract
Atherosclerosis (AS) is a chronic inflammatory disease primarily affecting large and medium-sized arterial vessels, characterized by lipoprotein disorders, intimal thickening, smooth muscle cell proliferation, and the formation of vulnerable plaques. Macrophages (MΦs) play a vital role in the inflammatory response throughout all stages of atherosclerotic development and are considered significant therapeutic targets. In early lesions, macrophage efferocytosis rapidly eliminates harmful cells. However, impaired efferocytosis in advanced plaques perpetuates the inflammatory microenvironment of AS. Defective efferocytosis has emerged as a key factor in atherosclerotic pathogenesis and the progression to severe cardiovascular disease. Herein, this review probes into investigate the potential mechanisms at the cellular, molecular, and organelle levels underlying defective macrophage efferocytosis in advanced lesion plaques. In the inflammatory microenvironments of AS with interactions among diverse inflammatory immune cells, impaired macrophage efferocytosis is strongly linked to multiple factors, such as a lower absolute number of phagocytes, the aberrant expression of crucial molecules, and impaired mitochondrial energy provision in phagocytes. Thus, focusing on molecular targets to enhance macrophage efferocytosis or targeting mitochondrial therapy to restore macrophage metabolism homeostasis has emerged as a potential strategy to mitigate the progression of advanced atherosclerotic plaque, providing various treatment options.
Collapse
Affiliation(s)
- Wanling Li
- School of Pharmacy and Bioengineering, Chongqing University of Technology, Chongqing 400054, China; The General Hospital of Western Theater Command, Chengdu 610083, China
| | - Yaqing Huang
- School of Pharmacy and Bioengineering, Chongqing University of Technology, Chongqing 400054, China; The General Hospital of Western Theater Command, Chengdu 610083, China
| | - Jun Liu
- School of Pharmacy and Bioengineering, Chongqing University of Technology, Chongqing 400054, China
| | - Yue Zhou
- School of Pharmacy and Bioengineering, Chongqing University of Technology, Chongqing 400054, China
| | - Hongyu Sun
- The General Hospital of Western Theater Command, Chengdu 610083, China
| | - Yonghong Fan
- The General Hospital of Western Theater Command, Chengdu 610083, China.
| | - Feila Liu
- School of Pharmacy and Bioengineering, Chongqing University of Technology, Chongqing 400054, China.
| |
Collapse
|
17
|
Zhang H, Wang Y, Liu M, Qi Y, Shen S, Gang Q, Jiang H, Lun Y, Zhang J. Deep Learning and Single-Cell Sequencing Analyses Unveiling Key Molecular Features in the Progression of Carotid Atherosclerotic Plaque. J Cell Mol Med 2024; 28:e70220. [PMID: 39586797 PMCID: PMC11588433 DOI: 10.1111/jcmm.70220] [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: 05/31/2024] [Revised: 10/30/2024] [Accepted: 11/07/2024] [Indexed: 11/27/2024] Open
Abstract
Rupture of advanced carotid atherosclerotic plaques increases the risk of ischaemic stroke, which has significant global morbidity and mortality rates. However, the specific characteristics of immune cells with dysregulated function and proven biomarkers for the diagnosis of atherosclerotic plaque progression remain poorly characterised. Our study elucidated the role of immune cells and explored diagnostic biomarkers in advanced plaque progression using single-cell RNA sequencing and high-dimensional weighted gene co-expression network analysis. We identified a subcluster of monocytes with significantly increased infiltration in the advanced plaques. Based on the monocyte signature and machine-learning approaches, we accurately distinguished advanced plaques from early plaques, with an area under the curve (AUC) of 0.899 in independent external testing. Using microenvironment cell populations (MCP) counter and non-negative matrix factorisation, we determined the association between monocyte signatures and immune cell infiltration as well as the heterogeneity of the patient. Finally, we constructed a convolutional neural network deep learning model based on gene-immune correlation, which achieved an AUC of 0.933, a sensitivity of 92.3%, and a specificity of 87.5% in independent external testing for diagnosing advanced plaques. Our findings on unique subpopulations of monocytes that contribute to carotid plaque progression are crucial for the development of diagnostic tools for clinical diseases.
Collapse
Affiliation(s)
- Han Zhang
- Department of Vascular SurgeryThe First Hospital of China Medical UniversityShenyangLiaoningChina
| | - Yixian Wang
- Department of Vascular SurgeryThe First Hospital of China Medical UniversityShenyangLiaoningChina
| | - Mingyu Liu
- Department of Vascular SurgeryThe First Hospital of China Medical UniversityShenyangLiaoningChina
| | - Yao Qi
- Department of Vascular SurgeryThe First Hospital of China Medical UniversityShenyangLiaoningChina
| | - Shikai Shen
- Department of Vascular SurgeryThe First Hospital of China Medical UniversityShenyangLiaoningChina
| | - Qingwei Gang
- Department of Vascular SurgeryThe First Hospital of China Medical UniversityShenyangLiaoningChina
| | - Han Jiang
- Department of Vascular SurgeryThe First Hospital of China Medical UniversityShenyangLiaoningChina
| | - Yu Lun
- Department of Vascular SurgeryThe First Hospital of China Medical UniversityShenyangLiaoningChina
| | - Jian Zhang
- Department of Vascular SurgeryThe First Hospital of China Medical UniversityShenyangLiaoningChina
| |
Collapse
|
18
|
Pereira-Neves A, Dias L, Fragão-Marques M, Vidoedo J, Ribeiro H, Andrade JP, Rocha-Neves J. Monocyte Count as a Predictor of Major Adverse Limb Events in Aortoiliac Revascularization. J Clin Med 2024; 13:6412. [PMID: 39518551 PMCID: PMC11546730 DOI: 10.3390/jcm13216412] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2024] [Revised: 10/20/2024] [Accepted: 10/24/2024] [Indexed: 11/16/2024] Open
Abstract
Background/Objectives: Atherosclerosis is a leading cause of death, especially in the developed world, and is marked by chronic arterial inflammation and lipid accumulation. As key players in its progression, monocytes contribute to plaque formation, inflammation, and tissue repair. Understanding monocyte involvement is crucial for developing better therapeutic approaches. The objective of this study is to assess the prognostic value of monocytes for limb-related outcomes following revascularization for complex aortoiliac lesions, thereby emphasizing the central role of monocytes in atherosclerosis. Methods: This prospective cohort study-enrolled patients who had undergone elective aortoiliac revascularization at two hospitals between January 2013 and December 2023. Patients with TASC II type D lesions were included, excluding those with aneurysmal disease. Demographic, clinical, and procedural data were gathered, and patients were monitored for limb-related outcomes. Preoperative complete blood counts were analyzed, and statistical analyses, including multivariable Cox regression, were conducted to identify predictors of major adverse limb events (MALE). Results: The study included 135 patients with a mean age of 62.4 ± 9.20 years and predominantly male (93%). Patients were followed for a median of 61 IQR [55.4-66.6] months. Smoking history (91%) was the most prevalent cardiovascular risk factor. Preoperative monocyte count >0.720 × 109/L was associated with worse 30-day limb-related outcomes (MALE: OR 7.138 95% CI: 1.509-33.764, p = 0.013) and long-term outcomes, including secondary patency (p = 0.03), major amputation (p = 0.04), and MALE (p = 0.039). Cox regression analysis confirmed an elevated monocyte count as an independent predictor of MALE (adjusted hazard ratio 2.149, 95% CI: 1.115-4.144, p = 0.022). Conclusions: This study demonstrated that patients with a higher absolute monocyte count may be more exposed to the risk of MALE in patients with aortoiliac TASC II type D lesions undergoing revascularization, with predictive accuracy in both the short and long term. Additionally, it was an independent predictor of major amputation. This new marker has the potential to serve as a cost-effective and easily available tool for risk stratification, helping identify patients at higher risk of MALE.
Collapse
Affiliation(s)
- António Pereira-Neves
- Unit of Anatomy, Department of Biomedicine, Faculty of Medicine, University of Porto, 4050-513 Porto, Portugal; (J.P.A.); (J.R.-N.)
- Department of Angiology and Vascular Surgery, Unidade Local de Saúde de São João, 4200-319 Porto, Portugal;
| | - Lara Dias
- Department of Angiology and Vascular Surgery, Unidade Local de Saúde de São João, 4200-319 Porto, Portugal;
- Department of Surgery and Physiology, Faculty of Medicine, University of Porto, 4050-513 Porto, Portugal
| | - Mariana Fragão-Marques
- Cardiovascular R&D Unit, Faculty of Medicine, University of Porto, 4050-513 Porto, Portugal;
| | - José Vidoedo
- Department of Angiology and Vascular Surgery, Unidade Local de Saúde entre o Tâmega e o Sousa, 4560-136 Penafiel, Portugal;
| | - Hugo Ribeiro
- Community Palliative Care Support Team Gaia, 4430-043 Vila Nova de Gaia, Portugal;
- Faculty of Medicine, University of Coimbra, 3004-535 Coimbra, Portugal
- Centre for Innovative Biomedicine and Biotechnology, 3004-504 Coimbra, Portugal
- MEDCIDS—Faculty of Medicine, University of Porto, 4050-513 Porto, Portugal
| | - José Paulo Andrade
- Unit of Anatomy, Department of Biomedicine, Faculty of Medicine, University of Porto, 4050-513 Porto, Portugal; (J.P.A.); (J.R.-N.)
- Rise@Health, Rua Dr. Plácido da Costa, s/n, 4200-450 Porto, Portugal
| | - João Rocha-Neves
- Unit of Anatomy, Department of Biomedicine, Faculty of Medicine, University of Porto, 4050-513 Porto, Portugal; (J.P.A.); (J.R.-N.)
- Rise@Health, Rua Dr. Plácido da Costa, s/n, 4200-450 Porto, Portugal
| |
Collapse
|
19
|
Wang J, Wu Q, Wang X, Liu H, Chen M, Xu L, Zhang Z, Li K, Li W, Zhong J. Targeting Macrophage Phenotypes and Metabolism as Novel Therapeutic Approaches in Atherosclerosis and Related Cardiovascular Diseases. Curr Atheroscler Rep 2024; 26:573-588. [PMID: 39133247 PMCID: PMC11392985 DOI: 10.1007/s11883-024-01229-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/22/2024] [Indexed: 08/13/2024]
Abstract
PURPOSE OF THE REVIEW Macrophage accumulation and activation function as hallmarks of atherosclerosis and have complex and intricate dynamics throughout all components and stages of atherosclerotic plaques. In this review, we focus on the regulatory roles and underlying mechanisms of macrophage phenotypes and metabolism in atherosclerosis. We highlight the diverse range of macrophage phenotypes present in atherosclerosis and their potential roles in progression and regression of atherosclerotic plaque. Furthermore, we discuss the challenges and opportunities in developing therapeutic strategies for preventing and treating atherosclerotic cardiovascular disease. RECENT FINDINGS Dysregulation of macrophage polarization between the proinflammatory M1 and anti-inflammatory M2 phenotypealters the immuno-inflammatory response during atherosclerosis progression, leading to plaque initiation, growth, and ultimately rupture. Altered metabolism of macrophage is a key feature for their function and the subsequent progression of atherosclerotic cardiovascular disease. The immunometabolism of macrophage has been implicated to macrophage activation and metabolic rewiring of macrophages within atherosclerotic lesions, thereby shifting altered macrophage immune-effector and tissue-reparative function. Targeting macrophage phenotypes and metabolism are potential therapeutic strategies in the prevention and treatment of atherosclerosis and atherosclerotic cardiovascular diseases. Understanding the precise function and metabolism of specific macrophage subsets and their contributions to the composition and growth of atherosclerotic plaques could reveal novel strategies to delay or halt development of atherosclerotic cardiovascular diseases and their associated pathophysiological consequences. Identifying biological stimuli capable of modulating macrophage phenotypes and metabolism may lead to the development of innovative therapeutic approaches for treating patients with atherosclerosis and coronary artery diseases.
Collapse
Affiliation(s)
- Juan Wang
- Beijing Key Laboratory of Hypertension, Heart Center of Beijing Chao-Yang Hospital, Capital Medical University, Beijing, China.
| | - Qiang Wu
- Senior Department of Cardiology, the Sixth Medical Center, Chinese PLA General Hospital, Beijing, China
- Journal of Geriatric Cardiology Editorial Office, Chinese PLA General Hospital, Beijing, China
| | - Xinyu Wang
- Beijing Key Laboratory of Hypertension, Heart Center of Beijing Chao-Yang Hospital, Capital Medical University, Beijing, China
| | - Hongbin Liu
- Department of Cardiology, the Second Medical Center, National Clinical Research Center for Geriatric Diseases, Chinese PLA General Hospital, Beijing, China
| | - Mulei Chen
- Beijing Key Laboratory of Hypertension, Heart Center of Beijing Chao-Yang Hospital, Capital Medical University, Beijing, China
| | - Li Xu
- Beijing Key Laboratory of Hypertension, Heart Center of Beijing Chao-Yang Hospital, Capital Medical University, Beijing, China
| | - Ze Zhang
- National Institute of Biological Sciences, Beijing, China
| | - Kuibao Li
- Beijing Key Laboratory of Hypertension, Heart Center of Beijing Chao-Yang Hospital, Capital Medical University, Beijing, China
| | - Weiming Li
- Beijing Key Laboratory of Hypertension, Heart Center of Beijing Chao-Yang Hospital, Capital Medical University, Beijing, China.
| | - Jiuchang Zhong
- Beijing Key Laboratory of Hypertension, Heart Center of Beijing Chao-Yang Hospital, Capital Medical University, Beijing, China.
| |
Collapse
|
20
|
Kang N, Kim J, Kwon M, Son Y, Eo SK, Baryawno N, Kim BS, Yoon S, Oh SO, Lee D, Kim K. Blockade of mTORC1 via Rapamycin Suppresses 27-Hydroxycholestrol-Induced Inflammatory Responses. Int J Mol Sci 2024; 25:10381. [PMID: 39408711 PMCID: PMC11477202 DOI: 10.3390/ijms251910381] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2024] [Revised: 09/15/2024] [Accepted: 09/24/2024] [Indexed: 10/20/2024] Open
Abstract
Atherosclerosis is characterized by the deposition and accumulation of extracellular cholesterol and inflammatory cells in the arterial blood vessel walls, and 27-hydroxycholesterol (27OHChol) is the most abundant cholesterol metabolite. 27OHChol is an oxysterol that induces immune responses, including immune cell activation and chemokine secretion, although the underlying mechanisms are not fully understood. In this study, we investigated the roles of the mechanistic target of rapamycin (mTOR) in 27HChol-induced inflammation using rapamycin. Treating monocytic cells with rapamycin effectively reduced the expression of CCL2 and CD14, which was involved with the increased immune response by 27OHChol. Rapamycin also suppressed the phosphorylation of S6 and 4EBP1, which are downstream of mTORC1. Additionally, it also alleviates the increase in differentiation markers into macrophage. These results suggest that 27OHChol induces inflammation by activating the mTORC1 signaling pathway, and rapamycin may be useful for the treatment of atherosclerosis-related inflammation involving 27OHchol.
Collapse
Affiliation(s)
- Nakyung Kang
- Department of Pharmacology, School of Medicine, Pusan National University, Yangsan 50612, Republic of Korea; (N.K.); (J.K.); (Y.S.)
| | - Jaesung Kim
- Department of Pharmacology, School of Medicine, Pusan National University, Yangsan 50612, Republic of Korea; (N.K.); (J.K.); (Y.S.)
| | - Munju Kwon
- Department of Convergence Medicine, School of Medicine, Pusan National University, Yangsan 50612, Republic of Korea;
| | - Yonghae Son
- Department of Pharmacology, School of Medicine, Pusan National University, Yangsan 50612, Republic of Korea; (N.K.); (J.K.); (Y.S.)
| | - Seong-Kug Eo
- College of Veterinary Medicine and Bio-Safety Research Institute, Jeonbuk National University, Iksan 54596, Republic of Korea;
| | - Ninib Baryawno
- Childhood Cancer Research Unit, Department of Women’s and Children’s Health, Karolinska Institute, 17177 Stockholm, Sweden;
| | - Byoung Soo Kim
- School of Biomedical Convergence Engineering, Pusan National University, Yangsan 50612, Republic of Korea;
| | - Sik Yoon
- Department of Anatomy, School of Medicine, Pusan National University, Yangsan 50612, Republic of Korea; (S.Y.); (S.-O.O.)
| | - Sae-Ock Oh
- Department of Anatomy, School of Medicine, Pusan National University, Yangsan 50612, Republic of Korea; (S.Y.); (S.-O.O.)
| | - Dongjun Lee
- Department of Convergence Medicine, School of Medicine, Pusan National University, Yangsan 50612, Republic of Korea;
- Transplantation Research Center, Research Institute for Convergence of Biomedical Science and Technology, Pusan National University Yangsan Hospital, Yangsan 50612, Republic of Korea
| | - Koanhoi Kim
- Department of Pharmacology, School of Medicine, Pusan National University, Yangsan 50612, Republic of Korea; (N.K.); (J.K.); (Y.S.)
| |
Collapse
|
21
|
Li X, Li Y, Hao Q, Jin J, Wang Y. Metabolic mechanisms orchestrated by Sirtuin family to modulate inflammatory responses. Front Immunol 2024; 15:1448535. [PMID: 39372420 PMCID: PMC11449768 DOI: 10.3389/fimmu.2024.1448535] [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: 06/13/2024] [Accepted: 09/03/2024] [Indexed: 10/08/2024] Open
Abstract
Maintaining metabolic homeostasis is crucial for cellular and organismal health throughout their lifespans. The intricate link between metabolism and inflammation through immunometabolism is pivotal in maintaining overall health and disease progression. The multifactorial nature of metabolic and inflammatory processes makes study of the relationship between them challenging. Homologs of Saccharomyces cerevisiae silent information regulator 2 protein, known as Sirtuins (SIRTs), have been demonstrated to promote longevity in various organisms. As nicotinamide adenine dinucleotide-dependent deacetylases, members of the Sirtuin family (SIRT1-7) regulate energy metabolism and inflammation. In this review, we provide an extensive analysis of SIRTs involved in regulating key metabolic pathways, including glucose, lipid, and amino acid metabolism. Furthermore, we systematically describe how the SIRTs influence inflammatory responses by modulating metabolic pathways, as well as inflammatory cells, mediators, and pathways. Current research findings on the preferential roles of different SIRTs in metabolic disorders and inflammation underscore the potential of SIRTs as viable pharmacological and therapeutic targets. Future research should focus on the development of promising compounds that target SIRTs, with the aim of enhancing their anti-inflammatory activity by influencing metabolic pathways within inflammatory cells.
Collapse
Affiliation(s)
- Xiaoqing Li
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, China
- Key Laboratory of Immune Response and Immunotherapy, University of Science and Technology of China, Hefei, Anhui, China
| | - Yunjia Li
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, China
- Key Laboratory of Immune Response and Immunotherapy, University of Science and Technology of China, Hefei, Anhui, China
| | - Quan Hao
- China Spallation Neutron Source, Dongguan, Guangdong, China
| | - Jing Jin
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, China
- Key Laboratory of Immune Response and Immunotherapy, University of Science and Technology of China, Hefei, Anhui, China
| | - Yi Wang
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, China
- Key Laboratory of Immune Response and Immunotherapy, University of Science and Technology of China, Hefei, Anhui, China
| |
Collapse
|
22
|
Santarsiero A, Todisco S, Convertini P, De Leonibus C, Infantino V. Transcriptional Regulation and Function of Malic Enzyme 1 in Human Macrophage Activation. Biomedicines 2024; 12:2089. [PMID: 39335602 PMCID: PMC11428690 DOI: 10.3390/biomedicines12092089] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2024] [Revised: 09/04/2024] [Accepted: 09/10/2024] [Indexed: 09/30/2024] Open
Abstract
Macrophages represent primary players of the innate immune system. Macrophage activation triggers several signaling pathways and is tightly associated with metabolic changes, which drive different immune subsets. Recent studies unveil the role of various metabolic enzymes in macrophage activation. Here, we show that malic enzyme 1 (ME1) is overexpressed in LPS-induced macrophages. Through chromatin immunoprecipitation, we demonstrate that ME1 transcriptional regulation is under control of NF-κB. Furthermore, ME1 activity is also increased in activated human PBMC-derived macrophages. Notably, ME1 gene silencing decreases nitric oxide as well as reactive oxygen species and prostaglandin E2 inflammatory mediators. Therefore, modulating ME1 provides a potential approach for immunometabolic regulation and in turn macrophage function.
Collapse
Affiliation(s)
- Anna Santarsiero
- Department of Science, University of Basilicata, 85100 Potenza, Italy; (A.S.); (S.T.); (P.C.)
| | - Simona Todisco
- Department of Science, University of Basilicata, 85100 Potenza, Italy; (A.S.); (S.T.); (P.C.)
| | - Paolo Convertini
- Department of Science, University of Basilicata, 85100 Potenza, Italy; (A.S.); (S.T.); (P.C.)
| | - Chiara De Leonibus
- Department of Health Sciences, University of Basilicata, 85100 Potenza, Italy;
- Telethon Institute of Genetics and Medicine (TIGEM), 80078 Pozzuoli, Italy
| | - Vittoria Infantino
- Department of Health Sciences, University of Basilicata, 85100 Potenza, Italy;
| |
Collapse
|
23
|
Wang J, Wang J, Zhong J, Liu H, Li W, Chen M, Xu L, Zhang W, Zhang Z, Wei Z, Guo J, Wang X, Sui J, Liu X, Zhang S, Wang X. LRG1 promotes atherosclerosis by inducing macrophage M1-like polarization. Proc Natl Acad Sci U S A 2024; 121:e2405845121. [PMID: 39178231 PMCID: PMC11363312 DOI: 10.1073/pnas.2405845121] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2024] [Accepted: 06/05/2024] [Indexed: 08/25/2024] Open
Abstract
Atherosclerosis is a chronic inflammatory disease of the arterial wall characterized by the accumulation of cholesterol-rich lipoproteins in macrophages. How macrophages commit to proinflammatory polarization under atherosclerosis conditions is not clear. Report here that the level of a circulating protein, leucine-rich alpha-2 glycoprotein 1 (LRG1), is elevated in the atherosclerotic tissue and serum samples from patients with coronary artery disease (CAD). LRG1 stimulated macrophages to proinflammatory M1-like polarization through the activation of extracellular signal-regulated kinase 1/2 (ERK1/2) and c-Jun N-terminal kinase (JNK) pathways. The LRG1 knockout mice showed significantly delayed atherogenesis progression and reduced levels of macrophage-related proinflammatory cytokines in a high-fat diet-induced Apoe-/- mouse atherosclerosis model. An anti-LRG1 neutralizing antibody also effectively blocked LRG1-induced macrophage M1-like polarization in vitro and conferred therapeutic benefits to animals with ApoE deficiency-induced atherosclerosis. LRG1 may therefore serve as an additional biomarker for CAD and targeting LRG1 could offer a potential therapeutic strategy for CAD patients by mitigating the proinflammatory response of macrophages.
Collapse
Affiliation(s)
- Juan Wang
- Heart-Center of Beijing Chao-Yang hospital, Capital Medical University, Beijing Key Laboratory of Hypertension, Beijing100020, China
| | - Jing Wang
- National Institute of Biological Sciences, Beijing102206, China
| | - Jiuchang Zhong
- Heart-Center of Beijing Chao-Yang hospital, Capital Medical University, Beijing Key Laboratory of Hypertension, Beijing100020, China
| | - Hongbin Liu
- Department of Cardiology, The Second Medical Center, Beijing 301 Hospital, Beijing100853, China
| | - Weiming Li
- Heart-Center of Beijing Chao-Yang hospital, Capital Medical University, Beijing Key Laboratory of Hypertension, Beijing100020, China
| | - Mulei Chen
- Heart-Center of Beijing Chao-Yang hospital, Capital Medical University, Beijing Key Laboratory of Hypertension, Beijing100020, China
| | - Li Xu
- Heart-Center of Beijing Chao-Yang hospital, Capital Medical University, Beijing Key Laboratory of Hypertension, Beijing100020, China
| | - Wenbin Zhang
- National Institute of Biological Sciences, Beijing102206, China
- Tsinghua Institute of Multidisciplinary Biomedical Research, Tsinghua University, Beijing102206, China
| | - Ze Zhang
- National Institute of Biological Sciences, Beijing102206, China
- Tsinghua Institute of Multidisciplinary Biomedical Research, Tsinghua University, Beijing102206, China
| | - Zhizhong Wei
- National Institute of Biological Sciences, Beijing102206, China
| | - Jia Guo
- National Institute of Biological Sciences, Beijing102206, China
- Tsinghua Institute of Multidisciplinary Biomedical Research, Tsinghua University, Beijing102206, China
| | - Xinyu Wang
- Heart-Center of Beijing Chao-Yang hospital, Capital Medical University, Beijing Key Laboratory of Hypertension, Beijing100020, China
| | - Jianhua Sui
- National Institute of Biological Sciences, Beijing102206, China
- Tsinghua Institute of Multidisciplinary Biomedical Research, Tsinghua University, Beijing102206, China
| | - Xingpeng Liu
- Heart-Center of Beijing Chao-Yang hospital, Capital Medical University, Beijing Key Laboratory of Hypertension, Beijing100020, China
| | - Sitao Zhang
- National Institute of Biological Sciences, Beijing102206, China
- Tsinghua Institute of Multidisciplinary Biomedical Research, Tsinghua University, Beijing102206, China
| | - Xiaodong Wang
- National Institute of Biological Sciences, Beijing102206, China
- Tsinghua Institute of Multidisciplinary Biomedical Research, Tsinghua University, Beijing102206, China
| |
Collapse
|
24
|
Liu Z, Zheng L. Associations between SII, SIRI, and cardiovascular disease in obese individuals: a nationwide cross-sectional analysis. Front Cardiovasc Med 2024; 11:1361088. [PMID: 39238504 PMCID: PMC11374596 DOI: 10.3389/fcvm.2024.1361088] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2023] [Accepted: 07/30/2024] [Indexed: 09/07/2024] Open
Abstract
Background Systemic immune-inflammation index (SII) and systemic inflammation response index (SIRI) are comprehensive markers of inflammatory status. However, the correlation between SII and SIRI and the prevalence of cardiovascular disease (CVD) in populations with obesity remains unknown. Methods This is a cross-sectional study with data obtained from the National Health and Nutrition Examination Survey from 1999 to 2018. SII and SIRI were calculated using the following equations: SII = (platelet count × neutrophil count)/lymphocyte count. SIRI = (neutrophil count × monocyte count)/lymphocyte count. Spearman's rank correlation coefficient was used to assess the relationship between SII and SIRI and baseline variables. Logistic regression models and generalized additive model (GAM) with a spline smoothing function were used to evaluate the association between SIRI and CVD prevalence. Nomogram and receiver operating characteristic curve (ROC) analysis were used to assess the value of the risk prediction model. Results A total of 17,261 participants with obesity and SII and SIRI publicly available data were used for this study. Multivariate logistic regression analysis revealed that SIRI, rather than SII, was an independent risk factor for CVD prevalence. For every standard deviation increase in SIRI, there was a 13%, 15%, and 28% increase in the odds ratios of CVD prevalence (OR = 1.13, 95% CI: 1.04-1.22, P = 0.01), coronary heart disease (OR = 1.15, 95% CI: 1.05-1.26, P = 0.002), and congestive heart failure (OR = 1.28, 95% CI: 1.16-1.41, P < 0.001). ROC results demonstrated that SIRI had a certain accuracy in predicting CVD prevalence (AUC = 0.604), especially when combined with other variables used in the nomogram (AUC = 0.828). The smooth curve fitting regression analysis demonstrated a significant linear association between the risk of SIRI and the odds ratio of CVD prevalence (P for nonlinear = 0.275). Conclusions SIRI is a relatively stable indicator of inflammation and is independently associated with the prevalence of CVD. It may serve as a novel inflammatory indicator to estimate CVD prevalence in populations with obesity.
Collapse
Affiliation(s)
- Zhou Liu
- Department of Cardiology, Huai'an Hospital Affiliated to Yangzhou University (The Fifth People's Hospital of Huai'an), Huai'an, China
| | - Longxuan Zheng
- Department of Cardiology, Huai'an Hospital Affiliated to Yangzhou University (The Fifth People's Hospital of Huai'an), Huai'an, China
| |
Collapse
|
25
|
Lei S, Liu C, Zheng TX, Fu W, Huang MZ. The relationship of redox signaling with the risk for atherosclerosis. Front Pharmacol 2024; 15:1430293. [PMID: 39148537 PMCID: PMC11324460 DOI: 10.3389/fphar.2024.1430293] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2024] [Accepted: 07/09/2024] [Indexed: 08/17/2024] Open
Abstract
Oxidative balance plays a pivotal role in physiological homeostasis, and many diseases, particularly age-related conditions, are closely associated with oxidative imbalance. While the strategic role of oxidative regulation in various diseases is well-established, the specific involvement of oxidative stress in atherosclerosis remains elusive. Atherosclerosis is a chronic inflammatory disorder characterized by plaque formation within the arteries. Alterations in the oxidative status of vascular tissues are linked to the onset, progression, and outcome of atherosclerosis. This review examines the role of redox signaling in atherosclerosis, including its impact on risk factors such as dyslipidemia, hyperglycemia, inflammation, and unhealthy lifestyle, along with dysregulation, vascular homeostasis, immune system interaction, and therapeutic considerations. Understanding redox signal transduction and the regulation of redox signaling will offer valuable insights into the pathogenesis of atherosclerosis and guide the development of novel therapeutic strategies.
Collapse
Affiliation(s)
- Sujuan Lei
- Department of General Surgery (Hepatopancreatobiliary Surgery), The Affiliated Hospital, Southwest Medical University, Luzhou, China
| | - Chen Liu
- Department of General Surgery (Hepatopancreatobiliary Surgery), The Affiliated Hospital, Southwest Medical University, Luzhou, China
| | - Tian-Xiang Zheng
- Metabolic Hepatobiliary and Pancreatic Diseases Key Laboratory of Luzhou City, Academician (Expert) Workstation of Sichuan Province, Department of General Surgery (Hepatopancreatobiliary Surgery), Chongqing, Sichuan, China
| | - Wenguang Fu
- Department of General Surgery (Hepatopancreatobiliary Surgery), The Affiliated Hospital, Southwest Medical University, Luzhou, China
- Metabolic Hepatobiliary and Pancreatic Diseases Key Laboratory of Luzhou City, Academician (Expert) Workstation of Sichuan Province, Department of General Surgery (Hepatopancreatobiliary Surgery), Chongqing, Sichuan, China
| | - Mei-Zhou Huang
- Department of General Surgery (Hepatopancreatobiliary Surgery), The Affiliated Hospital, Southwest Medical University, Luzhou, China
- Metabolic Hepatobiliary and Pancreatic Diseases Key Laboratory of Luzhou City, Academician (Expert) Workstation of Sichuan Province, Department of General Surgery (Hepatopancreatobiliary Surgery), Chongqing, Sichuan, China
| |
Collapse
|
26
|
Shimizu Y, Kawashiri SY, Noguchi Y, Sasaki N, Matsuyama M, Nakamichi S, Arima K, Nagata Y, Maeda T, Hayashida N. Association between eating speed and atherosclerosis in relation to growth differentiation factor-15 levels in older individuals in a cross-sectional study. Sci Rep 2024; 14:16492. [PMID: 39019981 PMCID: PMC11255208 DOI: 10.1038/s41598-024-67187-3] [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: 08/22/2023] [Accepted: 07/09/2024] [Indexed: 07/19/2024] Open
Abstract
Although fast eating speed has been associated with cardiovascular risk factors, no studies have reported an association between fast eating speed and atherosclerosis as evaluated by carotid intima-media thickness (CIMT). Rapid glucose ingestion is known to cause glucose spikes, which may accelerate atherogenesis and increase levels of growth differentiation factor 15 (GDF-15). Therefore, GDF-15 levels may influence the association between fast eating speed and atherosclerosis. To evaluate the association between eating speed and atherosclerosis in relation to GDF-15, this cross-sectional study analyzed 742 Japanese aged 60-69 years. They were required to have normal thyroid hormone levels, because both GDF-15 levels and atherosclerosis (CIMT ≥ 1.1 mm) can be influenced by thyroid dysfunction. Participants were stratified by the median GDF-15 level. A significant positive association was observed between fast eating speed and atherosclerosis, but only among participants with a high GDF-15 level: the sex- and age-adjusted odds ratios (95% confidence intervals) were 1.95 (1.09, 3.48) in participants with a high GDF-15 level, and 0.83 (0.37, 1.88) in those with a low GDF-15 level. This association remained even after further adjustment for thyroid function and metabolic factors. Serum concentrations of GDF-15 may mediate the association between fast eating speed and atherosclerosis.
Collapse
Affiliation(s)
- Yuji Shimizu
- Epidemiology Section, Division of Public Health, Osaka Institute of Public Health, Osaka, 537-0025, Japan.
- Department of General Medicine, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki, 852-8523, Japan.
| | - Shin-Ya Kawashiri
- Department of Community Medicine, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki, 852-8523, Japan
- Leading Medical Research Core Unit, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki, 852-8523, Japan
| | - Yuko Noguchi
- Department of Community Medicine, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki, 852-8523, Japan
| | - Nagisa Sasaki
- Epidemiology Section, Division of Public Health, Osaka Institute of Public Health, Osaka, 537-0025, Japan
| | - Mutsumi Matsuyama
- Division of Strategic Collaborative Research, Atomic Bomb Disease Institute, Nagasaki University, Nagasaki, 852-8523, Japan
| | - Seiko Nakamichi
- Department of General Medicine, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki, 852-8523, Japan
- Nagasaki University Health Center, Nagasaki, 852-8523, Japan
| | - Kazuhiko Arima
- Leading Medical Research Core Unit, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki, 852-8523, Japan
- Department of Public Health, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki, 852-8523, Japan
| | - Yasuhiro Nagata
- Department of Community Medicine, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki, 852-8523, Japan
- Leading Medical Research Core Unit, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki, 852-8523, Japan
| | - Takahiro Maeda
- Department of General Medicine, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki, 852-8523, Japan
- Leading Medical Research Core Unit, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki, 852-8523, Japan
- Department of Island and Community Medicine, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki, 853-0031, Japan
| | - Naomi Hayashida
- Leading Medical Research Core Unit, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki, 852-8523, Japan
- Division of Strategic Collaborative Research, Atomic Bomb Disease Institute, Nagasaki University, Nagasaki, 852-8523, Japan
| |
Collapse
|
27
|
Yang B, Hang S, Xu S, Gao Y, Yu W, Zang G, Zhang L, Wang Z. Macrophage polarisation and inflammatory mechanisms in atherosclerosis: Implications for prevention and treatment. Heliyon 2024; 10:e32073. [PMID: 38873669 PMCID: PMC11170185 DOI: 10.1016/j.heliyon.2024.e32073] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2023] [Revised: 05/11/2024] [Accepted: 05/28/2024] [Indexed: 06/15/2024] Open
Abstract
Atherosclerosis is a chronic inflammatory disease characterised by plaque accumulation in the arteries. Macrophages are immune cells that are crucial in the development of atherosclerosis. Macrophages can adopt different phenotypes, with the M1 phenotype promoting inflammation while the M2 phenotype counteracting it. This review focuses on the factors that drive the polarisation of M1 macrophages towards a pro-inflammatory phenotype during AS. Additionally, we explored metabolic reprogramming mechanisms and cytokines secretion by M1 macrophages. Hyperlipidaemia is widely recognised as a major risk factor for atherosclerosis. Modified lipoproteins released in the presence of hyperlipidaemia can trigger the release of cytokines and recruit circulating monocytes, which adhere to the damaged endothelium and differentiate into macrophages. Macrophages engulf lipids, leading to the formation of foam cells. As atherosclerosis progresses, foam cells become the necrotic core within the atherosclerotic plaques, destabilising them and triggering ischaemic disease. Furthermore, we discuss recent research focusing on targeting macrophages or inflammatory pathways for preventive or therapeutic purposes. These include statins, PCSK9 inhibitors, and promising nanotargeted drugs. These new developments hold the potential for the prevention and treatment of atherosclerosis and its related complications.
Collapse
Affiliation(s)
- Bo Yang
- Department of Cardiology, Affiliated Hospital of Jiangsu University, Zhenjiang, 212001, China
| | - Sanhua Hang
- Department of Hematology, Affiliated Danyang Hospital of Nantong University, Danyang, 212300, China
| | - Siting Xu
- Department of Cardiology, Affiliated Hospital of Jiangsu University, Zhenjiang, 212001, China
| | - Yun Gao
- Department of Pathology, Affiliated Hospital of Jiangsu University, Zhenjiang, 212001, China
| | - Wenhua Yu
- Department of Cardiology, Affiliated Hospital of Jiangsu University, Zhenjiang, 212001, China
| | - Guangyao Zang
- Department of Cardiology, Affiliated Hospital of Jiangsu University, Zhenjiang, 212001, China
| | - Lili Zhang
- Department of Cardiology, Affiliated Hospital of Jiangsu University, Zhenjiang, 212001, China
| | - Zhongqun Wang
- Department of Cardiology, Affiliated Hospital of Jiangsu University, Zhenjiang, 212001, China
| |
Collapse
|
28
|
Guo Y, Luo L, Zhu J, Li C. Advance in Multi-omics Research Strategies on Cholesterol Metabolism in Psoriasis. Inflammation 2024; 47:839-852. [PMID: 38244176 DOI: 10.1007/s10753-023-01961-9] [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: 09/24/2023] [Revised: 11/29/2023] [Accepted: 12/25/2023] [Indexed: 01/22/2024]
Abstract
The skin is a complex and dynamic organ where homeostasis is maintained through the intricate interplay between the immune system and metabolism, particularly cholesterol metabolism. Various factors such as cytokines, inflammatory mediators, cholesterol metabolites, and metabolic enzymes play crucial roles in facilitating these interactions. Dysregulation of this delicate balance contributes to the pathogenic pathways of inflammatory skin conditions, notably psoriasis. In this article, we provide an overview of omics biomarkers associated with psoriasis in relation to cholesterol metabolism. We explore multi-omics approaches that reveal the communication between immunometabolism and psoriatic inflammation. Additionally, we summarize the use of multi-omics strategies to uncover the complexities of multifactorial and heterogeneous inflammatory diseases. Finally, we highlight potential future perspectives related to targeted drug therapies and research areas that can advance precise medicine. This review aims to serve as a valuable resource for those investigating the role of cholesterol metabolism in psoriasis.
Collapse
Affiliation(s)
- Youming Guo
- Hospital for Skin Diseases, Institute of Dermatology, Chinese Academy of Medical Sciences & Peking Union Medical College, Nanjing, Jiangsu, China
- Jiangsu Key Laboratory of Molecular Biology for Skin Diseases and STIs, Nanjing, Jiangsu, China
| | - Lingling Luo
- Hospital for Skin Diseases, Institute of Dermatology, Chinese Academy of Medical Sciences & Peking Union Medical College, Nanjing, Jiangsu, China
| | - Jing Zhu
- Hospital for Skin Diseases, Institute of Dermatology, Chinese Academy of Medical Sciences & Peking Union Medical College, Nanjing, Jiangsu, China
| | - Chengrang Li
- Hospital for Skin Diseases, Institute of Dermatology, Chinese Academy of Medical Sciences & Peking Union Medical College, Nanjing, Jiangsu, China.
- Jiangsu Key Laboratory of Molecular Biology for Skin Diseases and STIs, Nanjing, Jiangsu, China.
| |
Collapse
|
29
|
Derler M, Teubenbacher T, Carapuig A, Nieswandt B, Fessler J, Kolb D, Mussbacher M. Platelets induce endoplasmic reticulum stress in macrophages in vitro. J Thromb Haemost 2024; 22:1475-1488. [PMID: 38278417 DOI: 10.1016/j.jtha.2024.01.009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2023] [Revised: 12/23/2023] [Accepted: 01/15/2024] [Indexed: 01/28/2024]
Abstract
BACKGROUND Endoplasmic reticulum (ER) stress is a key feature of lipid-laden macrophages and contributes to the development of atherosclerotic plaques. Blood platelets are known to interact with macrophages and fine-tune effector functions such as inflammasome activation and phagocytosis. However, the effect of platelets on ER stress induction is unknown. OBJECTIVES The objective of this study is to elucidate the potential of platelets in regulating ER stress in macrophages in vitro. METHODS Bone marrow-derived macrophages and RAW 264.7 cells were incubated with isolated murine platelets, and ER stress and inflammation markers were determined by reverse transcription-quantitative polymerase chain reaction, Western blotting, and enzyme-linked immunosorbent assay. ER morphology was investigated by electron microscopy. Cell viability, lipid accumulation, and activation were measured by flow cytometry. To gain mechanistic insights, coincubation experiments were performed with platelet decoys/releasates as well as lipopolysaccharide, blocking antibodies, and TLR4 inhibitors. RESULTS Coincubation of platelets and macrophages led to elevated levels of ER stress markers (BIP, IRE1α, CHOP, and XBP1 splicing) in murine and human macrophages, which led to a pronounced enlargement of the ER. Macrophage ER stress was accompanied by increased release of proinflammatory cytokines and intracellular lipid accumulation, but not cell death. Platelet decoys, but not platelet releasates or lysate from other cells, phenocopied the effect of platelets. Blocking TLR4 inhibited inflammatory activation of macrophages but did not affect ER stress induction by platelet coincubation. CONCLUSION To our knowledge, this study is the first to demonstrate that platelets induce ER stress and unfolded protein response in macrophages by heat-sensitive membrane proteins, independent of inflammatory activation of macrophages.
Collapse
Affiliation(s)
- Martina Derler
- Department of Pharmacology and Toxicology, Institute of Pharmaceutical Sciences, University of Graz, Graz, Austria
| | - Theresa Teubenbacher
- Department of Pharmacology and Toxicology, Institute of Pharmaceutical Sciences, University of Graz, Graz, Austria
| | - Anna Carapuig
- Department of Pharmacology and Toxicology, Institute of Pharmaceutical Sciences, University of Graz, Graz, Austria
| | - Bernhard Nieswandt
- Institute of Experimental Biomedicine, Chair of Experimental Biomedicine I, University Hospital Würzburg, Würzburg, Germany; Rudolf Virchow Center for Integrative and Translational Bioimaging, Julius-Maximilians-Universität Würzburg, Würzburg, Germany
| | - Johannes Fessler
- Division of Immunology and Pathophysiology, Medical University of Graz, Graz, Austria
| | - Dagmar Kolb
- Center for Medical Research, Core Facility Ultrastructure Analysis, Medical University of Graz, Graz, Austria; Division of Cell Biology, Histology, and Embryology, Gottfried Schatz Research Center, Medical University of Graz, Graz, Austria; BioTechMed-Graz, Graz, Austria
| | - Marion Mussbacher
- Department of Pharmacology and Toxicology, Institute of Pharmaceutical Sciences, University of Graz, Graz, Austria; BioTechMed-Graz, Graz, Austria.
| |
Collapse
|
30
|
Xie L, Chen J, Hu H, Zhu Y, Wang X, Zhou S, Wang F, Xiang M. Engineered M2 macrophage-derived extracellular vesicles with platelet membrane fusion for targeted therapy of atherosclerosis. Bioact Mater 2024; 35:447-460. [PMID: 38390527 PMCID: PMC10881364 DOI: 10.1016/j.bioactmat.2024.02.015] [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: 09/27/2023] [Revised: 01/04/2024] [Accepted: 02/11/2024] [Indexed: 02/24/2024] Open
Abstract
Atherosclerosis is featured as chronic low-grade inflammation in the arteries, which leads to the formation of plaques rich in lipids. M2 macrophage-derived extracellular vesicles (M2EV) have significant potential for anti-atherosclerotic therapy. However, their therapeutic effectiveness has been hindered by their limited targeting capability in vivo. The objective of this study was to create the P-M2EV (platelet membrane-modified M2EV) using the membrane fusion technique in order to imitate the interaction between platelets and macrophages. P-M2EV exhibited excellent physicochemical properties, and microRNA (miRNA)-sequencing revealed that the extrusion process had no detrimental effects on miRNAs carried by the nanocarriers. Remarkably, miR-99a-5p was identified as the miRNA with the highest expression level, which targeted the mRNA of Homeobox A1 (HOXA1) and effectively suppressed the formation of foam cells in vitro. In an atherosclerotic low-density lipoprotein receptor-deficient (Ldlr-/-) mouse model, the intravenous injection of P-M2EV showed enhanced targeting and greater infiltration into atherosclerotic plaques compared to regular extracellular vesicles. Crucially, P-M2EV successfully suppressed the progression of atherosclerosis without causing systemic toxicity. The findings demonstrated a biomimetic platelet-mimic system that holds great promise for the treatment of atherosclerosis in clinical settings.
Collapse
Affiliation(s)
- Lan Xie
- Department of Cardiology, The Second Affiliated Hospital, Zhejiang University School of Medicine, 88 Jiefang Road, Hangzhou, 310009, China
- State Key Laboratory of Transvascular Implantation Devices, Hangzhou, 310009, China
- Cardiovascular Key Laboratory of Zhejiang Province, Hangzhou, 310009, China
- Department of Rheumatology, The Second Affiliated Hospital, Zhejiang University School of Medicine, 88 Jiefang Road, Hangzhou, 310009, China
| | - Jinyong Chen
- Department of Cardiology, The Second Affiliated Hospital, Zhejiang University School of Medicine, 88 Jiefang Road, Hangzhou, 310009, China
- State Key Laboratory of Transvascular Implantation Devices, Hangzhou, 310009, China
- Cardiovascular Key Laboratory of Zhejiang Province, Hangzhou, 310009, China
| | - Haochang Hu
- Department of Cardiology, The Second Affiliated Hospital, Zhejiang University School of Medicine, 88 Jiefang Road, Hangzhou, 310009, China
- State Key Laboratory of Transvascular Implantation Devices, Hangzhou, 310009, China
- Cardiovascular Key Laboratory of Zhejiang Province, Hangzhou, 310009, China
| | - Yuan Zhu
- Department of Cardiology, The Second Affiliated Hospital, Zhejiang University School of Medicine, 88 Jiefang Road, Hangzhou, 310009, China
- State Key Laboratory of Transvascular Implantation Devices, Hangzhou, 310009, China
- Cardiovascular Key Laboratory of Zhejiang Province, Hangzhou, 310009, China
| | - Xiying Wang
- Department of Gastroenterology, The Second Affiliated Hospital, Zhejiang University School of Medicine, 88 Jiefang Road, Hangzhou, 310009, China
| | - Siyu Zhou
- Department of Cardiology, The Second Affiliated Hospital, Zhejiang University School of Medicine, 88 Jiefang Road, Hangzhou, 310009, China
- State Key Laboratory of Transvascular Implantation Devices, Hangzhou, 310009, China
- Cardiovascular Key Laboratory of Zhejiang Province, Hangzhou, 310009, China
| | - Feifan Wang
- Department of Cardiology, The Second Affiliated Hospital, Zhejiang University School of Medicine, 88 Jiefang Road, Hangzhou, 310009, China
- State Key Laboratory of Transvascular Implantation Devices, Hangzhou, 310009, China
- Cardiovascular Key Laboratory of Zhejiang Province, Hangzhou, 310009, China
| | - Meixiang Xiang
- Department of Cardiology, The Second Affiliated Hospital, Zhejiang University School of Medicine, 88 Jiefang Road, Hangzhou, 310009, China
- State Key Laboratory of Transvascular Implantation Devices, Hangzhou, 310009, China
- Cardiovascular Key Laboratory of Zhejiang Province, Hangzhou, 310009, China
| |
Collapse
|
31
|
Liu Y, Tang X, Yuan H, Gao R. Naringin Inhibits Macrophage Foam Cell Formation by Regulating Lipid Homeostasis and Metabolic Phenotype. Nutrients 2024; 16:1321. [PMID: 38732567 PMCID: PMC11085135 DOI: 10.3390/nu16091321] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2024] [Revised: 04/18/2024] [Accepted: 04/23/2024] [Indexed: 05/13/2024] Open
Abstract
Imbalances in lipid uptake and efflux and inflammation are major contributors to foam cell formation, which is considered a therapeutic target to protect against atherosclerosis. Naringin, a citrus flavonoid abundant in citrus fruits, has been reported to exert an antiatherogenic function, but its pharmacological mechanism is unclear. Naringin treatment effectively inhibits foam cell formation in THP-1 and RAW264.7 macrophages. In this study, mechanically, naringin maintained lipid homeostasis within macrophages through downregulation of the key genes for lipid uptake (MSR1 and CD36) and the upregulation of ABCA1, ABCG1 and SR-B1, which are responsible for cholesterol efflux. Meanwhile, naringin significantly decreased the cholesterol synthesis-related genes and increased the genes involved in cholesterol metabolism. Subsequently, the results showed that ox-LDL-induced macrophage inflammatory responses were inhibited by naringin by reducing the proinflammatory cytokines IL-1β, IL-6 and TNF-α, and increasing the anti- inflammatory cytokine IL-10, which was further verified by the downregulation of pro-inflammatory and chemokine-related genes. Additionally, we found that naringin reprogrammed the metabolic phenotypes of macrophages by suppressing glycolysis and promoting lipid oxidation metabolism to restore macrophage phenotypes and functions. These results suggest that naringin is a potential drug for the treatment of AS as it inhibits macrophage foam cell formation by regulating metabolic phenotypes and inflammation.
Collapse
Affiliation(s)
- Yan Liu
- Department of Pharmacy, Air Force Medical Center, PLA, Beijing 100142, China; (Y.L.); (X.T.); (H.Y.)
- School of Pharmacy, Anhui Medical University, Hefei 230032, China
| | - Xiaohan Tang
- Department of Pharmacy, Air Force Medical Center, PLA, Beijing 100142, China; (Y.L.); (X.T.); (H.Y.)
| | - Hailong Yuan
- Department of Pharmacy, Air Force Medical Center, PLA, Beijing 100142, China; (Y.L.); (X.T.); (H.Y.)
| | - Rong Gao
- Department of Pharmacy, Air Force Medical Center, PLA, Beijing 100142, China; (Y.L.); (X.T.); (H.Y.)
| |
Collapse
|
32
|
Cao L, Wu C, Liu M, Zhang W, Chen H, Wang R, He Z. The association between monocyte-to-high-density lipoprotein ratio and hyperuricemia: Results from 2009 to 2018. Medicine (Baltimore) 2024; 103:e37713. [PMID: 38669360 PMCID: PMC11049789 DOI: 10.1097/md.0000000000037713] [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: 01/05/2024] [Accepted: 03/04/2024] [Indexed: 04/28/2024] Open
Abstract
Previous research has suggested that the monocyte-to-high-density lipoprotein ratio (MHR), an emerging inflammatory biomarker, holds promise in predicting the prevalence of various cardiovascular and metabolic diseases. However, earlier investigations were constrained by the relatively modest sample sizes. This study endeavored to expand the sample size and conduct a more comprehensive exploration of the potential relationship between MHR and hyperuricemia. This cross-sectional study incorporated data from participants of the 2009 to 2018 National Health and Nutrition Examination Survey (NHANES) with complete and qualifying information. MHR was determined by calculating the ratio between monocyte count and high-density lipoprotein levels. Various statistical methodologies such as weighted multivariate logistic regression, subgroup analysis, smoothed curve fitting, and threshold analysis, have been used to explore the correlation between hyperuricemia and MHR. The study included a cohort of 17,694 participants, of whom 3512 were diagnosed with hyperuricemia. MHR levels were notably higher in the hyperuricemia group than in the normal group, aligning with an elevated body mass index (BMI). A comprehensive multivariate logistic analysis, accounting for all relevant adjustments, revealed a notable positive correlation between MHR and hyperuricemia (P < .001, OR = 1.98, 95% CI: 1.54-2.54). Subgroup analysis indicated that the MHR exhibited an enhanced predictive capacity for identifying hyperuricemia risk, particularly in females (P < .05). Curvilinear and threshold analyses revealed a nonlinear association between MHR and hyperuricemia prevalence, with a notable inflection point at 0.826. In the US population, a clear positive correlation was observed between the MHR and prevalence of hyperuricemia. Importantly, the MHR is a more robust predictor of hyperuricemia risk in females. Further investigations are required to confirm these findings.
Collapse
Affiliation(s)
- Lei Cao
- College of Chinese Medicine, Changchun University of Chinese Medicine, Changchun, Jilin, China
| | - Chunwei Wu
- Department of Endocrinology and Metabolism, Affiliated Hospital of Changchun University of Chinese Medicine, Changchun, Jilin, China
| | - Miao Liu
- College of Chinese Medicine, Changchun University of Chinese Medicine, Changchun, Jilin, China
| | - Wenlong Zhang
- Department of Endocrinology and Metabolism, Affiliated Hospital of Changchun University of Chinese Medicine, Changchun, Jilin, China
| | - Hailong Chen
- College of Chinese Medicine, Changchun University of Chinese Medicine, Changchun, Jilin, China
| | - Ruolin Wang
- College of Chinese Medicine, Changchun University of Chinese Medicine, Changchun, Jilin, China
| | - Ze He
- Department of Endocrinology and Metabolism, Affiliated Hospital of Changchun University of Chinese Medicine, Changchun, Jilin, China
| |
Collapse
|
33
|
Harber KJ, Neele AE, van Roomen CP, Gijbels MJ, Beckers L, Toom MD, Schomakers BV, Heister DA, Willemsen L, Griffith GR, de Goede KE, van Dierendonck XA, Reiche ME, Poli A, L-H Mogensen F, Michelucci A, Verberk SG, de Vries H, van Weeghel M, Van den Bossche J, de Winther MP. Targeting the ACOD1-itaconate axis stabilizes atherosclerotic plaques. Redox Biol 2024; 70:103054. [PMID: 38309122 PMCID: PMC10848031 DOI: 10.1016/j.redox.2024.103054] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2023] [Revised: 01/16/2024] [Accepted: 01/20/2024] [Indexed: 02/05/2024] Open
Abstract
Inflammatory macrophages are key drivers of atherosclerosis that can induce rupture-prone vulnerable plaques. Skewing the plaque macrophage population towards a more protective phenotype and reducing the occurrence of clinical events is thought to be a promising method of treating atherosclerotic patients. In the current study, we investigate the immunomodulatory properties of itaconate, an immunometabolite derived from the TCA cycle intermediate cis-aconitate and synthesised by the enzyme Aconitate Decarboxylase 1 (ACOD1, also known as IRG1), in the context of atherosclerosis. Ldlr-/- atherogenic mice transplanted with Acod1-/- bone marrow displayed a more stable plaque phenotype with smaller necrotic cores and showed increased recruitment of monocytes to the vessel intima. Macrophages from Acod1-/- mice contained more lipids whilst also displaying reduced induction of apoptosis. Using multi-omics approaches, we identify a metabolic shift towards purine metabolism, in addition to an altered glycolytic flux towards production of glycerol for triglyceride synthesis. Overall, our data highlight the potential of therapeutically blocking ACOD1 with the aim of stabilizing atherosclerotic plaques.
Collapse
Affiliation(s)
- Karl J Harber
- Department of Medical Biochemistry, Amsterdam UMC, University of Amsterdam, 1105 AZ, Amsterdam, the Netherlands; Amsterdam Cardiovascular Sciences (ACS), Atherosclerosis & Ischemic Syndromes, Amsterdam UMC, the Netherlands; Amsterdam Institute for Infection and Immunity (AII), Inflammatory Diseases, Amsterdam UMC, the Netherlands; Department of Molecular Cell Biology and Immunology, Amsterdam UMC, Vrije Universiteit Amsterdam, 1081 HV, Amsterdam, the Netherlands
| | - Annette E Neele
- Department of Medical Biochemistry, Amsterdam UMC, University of Amsterdam, 1105 AZ, Amsterdam, the Netherlands; Amsterdam Cardiovascular Sciences (ACS), Atherosclerosis & Ischemic Syndromes, Amsterdam UMC, the Netherlands; Amsterdam Institute for Infection and Immunity (AII), Inflammatory Diseases, Amsterdam UMC, the Netherlands
| | - Cindy Paa van Roomen
- Department of Medical Biochemistry, Amsterdam UMC, University of Amsterdam, 1105 AZ, Amsterdam, the Netherlands
| | - Marion Jj Gijbels
- Department of Medical Biochemistry, Amsterdam UMC, University of Amsterdam, 1105 AZ, Amsterdam, the Netherlands; Department of Pathology, CARIM, Cardiovascular Research Institute Maastricht, GROW-School for Oncology and Developmental Biology, Maastricht UMC, University of Maastricht, 6229 HX, Maastricht, the Netherlands
| | - Linda Beckers
- Department of Medical Biochemistry, Amsterdam UMC, University of Amsterdam, 1105 AZ, Amsterdam, the Netherlands
| | - Myrthe den Toom
- Department of Medical Biochemistry, Amsterdam UMC, University of Amsterdam, 1105 AZ, Amsterdam, the Netherlands
| | - Bauke V Schomakers
- Department of Genetic Metabolic Diseases, Amsterdam UMC, University of Amsterdam, 1105 AZ, Amsterdam, the Netherlands
| | - Daan Af Heister
- Department of Molecular Cell Biology and Immunology, Amsterdam UMC, Vrije Universiteit Amsterdam, 1081 HV, Amsterdam, the Netherlands
| | - Lisa Willemsen
- Department of Medical Biochemistry, Amsterdam UMC, University of Amsterdam, 1105 AZ, Amsterdam, the Netherlands; Amsterdam Cardiovascular Sciences (ACS), Atherosclerosis & Ischemic Syndromes, Amsterdam UMC, the Netherlands; Amsterdam Institute for Infection and Immunity (AII), Inflammatory Diseases, Amsterdam UMC, the Netherlands
| | - Guillermo R Griffith
- Department of Medical Biochemistry, Amsterdam UMC, University of Amsterdam, 1105 AZ, Amsterdam, the Netherlands
| | - Kyra E de Goede
- Amsterdam Institute for Infection and Immunity (AII), Inflammatory Diseases, Amsterdam UMC, the Netherlands; Department of Molecular Cell Biology and Immunology, Amsterdam UMC, Vrije Universiteit Amsterdam, 1081 HV, Amsterdam, the Netherlands; Amsterdam Gastroenterology Endocrinology Metabolism (AGEM), Amsterdam UMC, the Netherlands
| | - Xanthe Amh van Dierendonck
- Amsterdam Institute for Infection and Immunity (AII), Inflammatory Diseases, Amsterdam UMC, the Netherlands; Department of Molecular Cell Biology and Immunology, Amsterdam UMC, Vrije Universiteit Amsterdam, 1081 HV, Amsterdam, the Netherlands; Amsterdam Gastroenterology Endocrinology Metabolism (AGEM), Amsterdam UMC, the Netherlands
| | - Myrthe E Reiche
- Department of Medical Biochemistry, Amsterdam UMC, University of Amsterdam, 1105 AZ, Amsterdam, the Netherlands; Amsterdam Cardiovascular Sciences (ACS), Atherosclerosis & Ischemic Syndromes, Amsterdam UMC, the Netherlands; Department of Medical Cell Biology, Uppsala University, 75236, Uppsala, Sweden
| | - Aurélie Poli
- Neuro-Immunology Group, Department of Cancer Research, Luxembourg Institute of Health, 6A Rue Nicolas-Ernest Barblé, L-1210, Luxembourg, Luxembourg
| | - Frida L-H Mogensen
- Neuro-Immunology Group, Department of Cancer Research, Luxembourg Institute of Health, 6A Rue Nicolas-Ernest Barblé, L-1210, Luxembourg, Luxembourg
| | - Alessandro Michelucci
- Neuro-Immunology Group, Department of Cancer Research, Luxembourg Institute of Health, 6A Rue Nicolas-Ernest Barblé, L-1210, Luxembourg, Luxembourg
| | - Sanne Gs Verberk
- Amsterdam Cardiovascular Sciences (ACS), Atherosclerosis & Ischemic Syndromes, Amsterdam UMC, the Netherlands; Amsterdam Institute for Infection and Immunity (AII), Inflammatory Diseases, Amsterdam UMC, the Netherlands; Department of Molecular Cell Biology and Immunology, Amsterdam UMC, Vrije Universiteit Amsterdam, 1081 HV, Amsterdam, the Netherlands
| | - Helga de Vries
- Department of Molecular Cell Biology and Immunology, Amsterdam UMC, Vrije Universiteit Amsterdam, 1081 HV, Amsterdam, the Netherlands; Amsterdam Neuroscience, Amsterdam, the Netherlands
| | - Michel van Weeghel
- Department of Genetic Metabolic Diseases, Amsterdam UMC, University of Amsterdam, 1105 AZ, Amsterdam, the Netherlands; Core Facility Metabolomics, Amsterdam UMC, University of Amsterdam, 1105 AZ Amsterdam, the Netherlands
| | - Jan Van den Bossche
- Amsterdam Cardiovascular Sciences (ACS), Atherosclerosis & Ischemic Syndromes, Amsterdam UMC, the Netherlands; Amsterdam Institute for Infection and Immunity (AII), Inflammatory Diseases, Amsterdam UMC, the Netherlands; Department of Molecular Cell Biology and Immunology, Amsterdam UMC, Vrije Universiteit Amsterdam, 1081 HV, Amsterdam, the Netherlands; Amsterdam Gastroenterology Endocrinology Metabolism (AGEM), Amsterdam UMC, the Netherlands.
| | - Menno Pj de Winther
- Department of Medical Biochemistry, Amsterdam UMC, University of Amsterdam, 1105 AZ, Amsterdam, the Netherlands; Amsterdam Cardiovascular Sciences (ACS), Atherosclerosis & Ischemic Syndromes, Amsterdam UMC, the Netherlands; Amsterdam Institute for Infection and Immunity (AII), Inflammatory Diseases, Amsterdam UMC, the Netherlands.
| |
Collapse
|
34
|
Groh LA, Willems LH, Fintelman P, Reijnen MMPJ, El Messaoudi S, Warlé MC. Dual-Pathway Inhibition with Rivaroxaban and Low-Dose Aspirin Does Not Alter Immune Cell Responsiveness and Distribution in Patients with Coronary Artery Disease. Cardiol Ther 2024; 13:233-242. [PMID: 38055176 PMCID: PMC10899137 DOI: 10.1007/s40119-023-00342-5] [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] [Accepted: 11/08/2023] [Indexed: 12/07/2023] Open
Abstract
INTRODUCTION Cardiovascular diseases (CVD) are the leading cause of death globally. Inflammation is an important driver of CVD where tissue damage may lead to the formation of deadly thrombi. Therefore, antithrombotic drugs, such as platelet inhibitors, are crucial for secondary risk prevention in coronary artery disease (CAD) and peripheral artery disease (PAD). For severe forms of the disease, dual-pathway inhibition (DPI) where low-dose aspirin is combined with rivaroxaban has shown improved efficacy in reducing cardiovascular mortality. METHODS Given this greater improvement in mortality, and the importance of inflammation in driving atherosclerosis, the potential for off-target inflammation-lowering effects of these drugs was evaluated by looking at the change in immune cell distribution and responsiveness to ex vivo lipopolysaccharide (LPS) stimulation after 3 months of DPI in patients with CAD. RESULTS We observed no changes in whole blood or peripheral blood mononuclear cell (PBMC) immune cell responsiveness to LPS after 3 months of DPI. Additionally, we did not observe any changes in the distribution of total white blood cells, monocytes, neutrophils, lymphocytes, or platelets during the study course. Signs of systemic inflammation were studied using Olink proteomics in 33 patients with PAD after 3 months of DPI. No changes were observed in any of the inflammatory proteins measured after the treatment period, suggesting that the state of chronic inflammation was not altered in these subjects. CONCLUSION Three months of DPI does not result in any meaningful change in immune cell responsiveness and distribution in patients with CAD or PAD. TRIAL REGISTRATION ClinicalTrials.gov ID: NCT05210725.
Collapse
Affiliation(s)
- Laszlo A Groh
- Department of Surgery (Internal Address 618), Radboud University Medical Center, Postal Address 9101, 6500 HB, Nijmegen, The Netherlands
- Department of Molecular Cell Biology and Immunology, Amsterdam UMC, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
| | - Loes H Willems
- Department of Surgery (Internal Address 618), Radboud University Medical Center, Postal Address 9101, 6500 HB, Nijmegen, The Netherlands.
| | - Paula Fintelman
- Department of Surgery (Internal Address 618), Radboud University Medical Center, Postal Address 9101, 6500 HB, Nijmegen, The Netherlands
| | - Michel M P J Reijnen
- Department of Surgery, Rijnstate Hospital, Arnhem, The Netherlands
- Multi-Modality Medical Imaging Group, University of Twente, Enschede, The Netherlands
| | - Saloua El Messaoudi
- Department of Cardiology, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Michiel C Warlé
- Department of Surgery (Internal Address 618), Radboud University Medical Center, Postal Address 9101, 6500 HB, Nijmegen, The Netherlands
| |
Collapse
|
35
|
Ciarambino T, Crispino P, Minervini G, Giordano M. Role of Helicobacter pylori Infection in Pathogenesis, Evolution, and Complication of Atherosclerotic Plaque. Biomedicines 2024; 12:400. [PMID: 38398002 PMCID: PMC10886498 DOI: 10.3390/biomedicines12020400] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2023] [Revised: 09/11/2023] [Accepted: 02/07/2024] [Indexed: 02/25/2024] Open
Abstract
The therapeutic management of atherosclerosis focuses almost exclusively on the reduction of plasma cholesterol levels. An important role in the genesis and evolution of atherosclerosis is played by chronic inflammation in promoting thrombosis phenomena after atheroma rupture. This review aims to take stock of the knowledge so far accumulated on the role of endemic HP infection in atherosclerosis. The studies produced so far have demonstrated a causal relationship between Helicobacter pylori (HP) and CVD. In a previous study, we demonstrated in HP-positive patients that thrombin and plasma fragment 1 + 2 production was proportionally related to tumor necrosis factor-alpha levels and that eradication of the infection resulted in a reduction of inflammation. At the end of our review, we can state that HP slightly affects the risk of CVD, particularly if the infection is associated with cytotoxic damage, and HP screening could have a clinically significant role in patients with a high risk of CVD. Considering the high prevalence of HP infection, an infection screening could be of great clinical utility in patients at high risk of CVD.
Collapse
Affiliation(s)
- Tiziana Ciarambino
- Internal Medicine Department, Hospital of Marcianise, ASL Caserta, 81037 Caserta, Italy
| | - Pietro Crispino
- Internal Medicine Department, Hospital of Latina, ASL Latina, 04100 Latina, Italy;
| | - Giovanni Minervini
- Internal Medicine Department, Hospital of Lagonegro, AOR San Carlo, 85042 Lagonegro, Italy;
| | - Mauro Giordano
- Department of Advanced Medical and Surgical Sciences, University of Campania “L. Vanvitelli”, 81100 Naples, Italy;
| |
Collapse
|
36
|
Yin M, Zhang Y, Li X, Liu S, Huang J, Yu H, Li X. Adverse effects of gestational diabetes mellitus on fetal monocytes revealed by single-cell RNA sequencing. iScience 2024; 27:108637. [PMID: 38188508 PMCID: PMC10770529 DOI: 10.1016/j.isci.2023.108637] [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: 08/10/2023] [Revised: 10/18/2023] [Accepted: 12/01/2023] [Indexed: 01/09/2024] Open
Abstract
Gestational diabetes mellitus (GDM), the most prevalent metabolic disorder during pregnancy, has long-term risks of metabolic diseases in offspring. However, the underlying mechanisms remain unclear. Here, we analyzed single-cell transcriptional data of cord blood mononuclear cells (CBMCs) from fetuses of healthy and GDM mothers, peripheral blood mononuclear cells from children and adolescents, and coronary plaques myeloid cells from atherosclerosis. Our results demonstrated that monocytes in cord blood were characterized with down-regulated proinflammatory-related pathways and up-regulated proliferation-related pathways. And monocytes in cord blood from GDM mothers were featured with expanded CXCL8+IL1B+ subclusters, enhanced crosstalk with neutrophil granulocytes and augmented adhesive and phagocytic abilities. Interestingly, CXCL8+IL1B+ monocytes influenced by GDM had transcriptome similarity with those of coronary plaques myeloid cells from individuals with atherosclerotic cardiovascular disease. Collectively, our data reveal adverse impact of maternal GDM environment on fetal monocytes and propose potential mechanisms between maternal GDM and offspring atherosclerosis.
Collapse
Affiliation(s)
- Min Yin
- National Clinical Research Center for Metabolic Diseases, Key Laboratory of Diabetes Immunology, Ministry of Education, and Department of Metabolism and Endocrinology, The Second Xiangya Hospital of Central South University, Changsha, China
| | - Yan Zhang
- National Clinical Research Center for Metabolic Diseases, Key Laboratory of Diabetes Immunology, Ministry of Education, and Department of Metabolism and Endocrinology, The Second Xiangya Hospital of Central South University, Changsha, China
| | - Xinyu Li
- National Clinical Research Center for Metabolic Diseases, Key Laboratory of Diabetes Immunology, Ministry of Education, and Department of Metabolism and Endocrinology, The Second Xiangya Hospital of Central South University, Changsha, China
| | - Shanshan Liu
- National Clinical Research Center for Metabolic Diseases, Key Laboratory of Diabetes Immunology, Ministry of Education, and Department of Metabolism and Endocrinology, The Second Xiangya Hospital of Central South University, Changsha, China
| | - Juan Huang
- National Clinical Research Center for Metabolic Diseases, Key Laboratory of Diabetes Immunology, Ministry of Education, and Department of Metabolism and Endocrinology, The Second Xiangya Hospital of Central South University, Changsha, China
- Section of Endocrinology, Department of Internal Medicine, School of Medicine, Yale University, New Haven, CT, USA
| | - Haibo Yu
- National Clinical Research Center for Metabolic Diseases, Key Laboratory of Diabetes Immunology, Ministry of Education, and Department of Metabolism and Endocrinology, The Second Xiangya Hospital of Central South University, Changsha, China
- Hunan Engineering Research Center of Cell Therapy for Diabetes, Changsha, China
| | - Xia Li
- National Clinical Research Center for Metabolic Diseases, Key Laboratory of Diabetes Immunology, Ministry of Education, and Department of Metabolism and Endocrinology, The Second Xiangya Hospital of Central South University, Changsha, China
| |
Collapse
|
37
|
Julla JB, Girard D, Diedisheim M, Saulnier PJ, Tran Vuong B, Blériot C, Carcarino E, De Keizer J, Orliaguet L, Nemazanyy I, Potier C, Khider K, Tonui DC, Ejlalmanesh T, Ballaire R, Mambu Mambueni H, Germain S, Gaborit B, Vidal-Trécan T, Riveline JP, Garchon HJ, Fenaille F, Lemoine S, Carlier A, Castelli F, Potier L, Masson D, Roussel R, Vandiedonck C, Hadjadj S, Alzaid F, Gautier JF, Venteclef N. Blood Monocyte Phenotype Is A Marker of Cardiovascular Risk in Type 2 Diabetes. Circ Res 2024; 134:189-202. [PMID: 38152893 DOI: 10.1161/circresaha.123.322757] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/04/2023] [Accepted: 12/13/2023] [Indexed: 12/29/2023]
Abstract
BACKGROUND Diabetes is a major risk factor for atherosclerotic cardiovascular diseases with a 2-fold higher risk of cardiovascular events in people with diabetes compared with those without. Circulating monocytes are inflammatory effector cells involved in both type 2 diabetes (T2D) and atherogenesis. METHODS We investigated the relationship between circulating monocytes and cardiovascular risk progression in people with T2D, using phenotypic, transcriptomic, and metabolomic analyses. cardiovascular risk progression was estimated with coronary artery calcium score in a cohort of 672 people with T2D. RESULTS Coronary artery calcium score was positively correlated with blood monocyte count and frequency of the classical monocyte subtype. Unsupervised k-means clustering based on monocyte subtype profiles revealed 3 main endotypes of people with T2D at varying risk of cardiovascular events. These observations were confirmed in a validation cohort of 279 T2D participants. The predictive association between monocyte count and major adverse cardiovascular events was validated through an independent prospective cohort of 757 patients with T2D. Integration of monocyte transcriptome analyses and plasma metabolomes showed a disruption of mitochondrial pathways (tricarboxylic acid cycle, oxidative phosphorylation pathway) that underlined a proatherogenic phenotype. CONCLUSIONS In this study, we provide evidence that frequency and monocyte phenotypic profile are closely linked to cardiovascular risk in patients with T2D. The assessment of monocyte frequency and count is a valuable predictive marker for risk of cardiovascular events in patients with T2D. REGISTRATION URL: https://www.clinicaltrials.gov; Unique identifier: NCT04353869.
Collapse
Affiliation(s)
- Jean-Baptiste Julla
- INSERM, Necker Enfants Malades (INEM), INSERM U1151, CNRS UMR 8253, IMMEDIAB Laboratory (J.-B.J., D.G., M.D., B.T.V., C.B., E.C., L.O., I.N., C.P., K.K., D.C.T., T.E., R.B., J.-P.R., L.P., R.R., C.V., F.A., J.-F.G., N.V.), Université Paris Cité, France
- Cordeliers Research Centre, INSERM, IMMEDIAB Laboratory, Sorbonne Université (J.-B.J., D.G., M.D., B.T.V., C.B., E.C., L.O., C.P., K.K., D.C.T., T.E., R.B., J.-P.R., L.P., R.R., C.V., F.A., J.-F.G., N.V.), Université Paris Cité, France
- Diabetes Institute (J.-B.J., D.G., M.D., B.T.V., C.B., E.C., L.O., C.P., K.K., D.C.T., T.E., R.B., T.V.-T., J.-P.R., L.P., R.R., C.V., F.A., J.-F.G., N.V.), Université Paris Cité, France
- Diabetology, Endocrinology and Nutrition Department, Lariboisière Hospital, Fédération de Diabétologie, France (J.-B.J., T.V.-T., J.-P.R., J.-F.G.)
| | - Diane Girard
- INSERM, Necker Enfants Malades (INEM), INSERM U1151, CNRS UMR 8253, IMMEDIAB Laboratory (J.-B.J., D.G., M.D., B.T.V., C.B., E.C., L.O., I.N., C.P., K.K., D.C.T., T.E., R.B., J.-P.R., L.P., R.R., C.V., F.A., J.-F.G., N.V.), Université Paris Cité, France
- Cordeliers Research Centre, INSERM, IMMEDIAB Laboratory, Sorbonne Université (J.-B.J., D.G., M.D., B.T.V., C.B., E.C., L.O., C.P., K.K., D.C.T., T.E., R.B., J.-P.R., L.P., R.R., C.V., F.A., J.-F.G., N.V.), Université Paris Cité, France
- Diabetes Institute (J.-B.J., D.G., M.D., B.T.V., C.B., E.C., L.O., C.P., K.K., D.C.T., T.E., R.B., T.V.-T., J.-P.R., L.P., R.R., C.V., F.A., J.-F.G., N.V.), Université Paris Cité, France
| | - Marc Diedisheim
- INSERM, Necker Enfants Malades (INEM), INSERM U1151, CNRS UMR 8253, IMMEDIAB Laboratory (J.-B.J., D.G., M.D., B.T.V., C.B., E.C., L.O., I.N., C.P., K.K., D.C.T., T.E., R.B., J.-P.R., L.P., R.R., C.V., F.A., J.-F.G., N.V.), Université Paris Cité, France
- Cordeliers Research Centre, INSERM, IMMEDIAB Laboratory, Sorbonne Université (J.-B.J., D.G., M.D., B.T.V., C.B., E.C., L.O., C.P., K.K., D.C.T., T.E., R.B., J.-P.R., L.P., R.R., C.V., F.A., J.-F.G., N.V.), Université Paris Cité, France
- Diabetes Institute (J.-B.J., D.G., M.D., B.T.V., C.B., E.C., L.O., C.P., K.K., D.C.T., T.E., R.B., T.V.-T., J.-P.R., L.P., R.R., C.V., F.A., J.-F.G., N.V.), Université Paris Cité, France
- Clinique Saint Gatien Alliance (NCT+), Saint-Cyr-sur-Loire, France (M.D.)
| | - Pierre-Jean Saulnier
- Poitiers Université, CHU Poitiers, INSERM, Centre d'Investigation Clinique CIC1402, Poitiers, France (P.-J.S.)
| | - Bao Tran Vuong
- INSERM, Necker Enfants Malades (INEM), INSERM U1151, CNRS UMR 8253, IMMEDIAB Laboratory (J.-B.J., D.G., M.D., B.T.V., C.B., E.C., L.O., I.N., C.P., K.K., D.C.T., T.E., R.B., J.-P.R., L.P., R.R., C.V., F.A., J.-F.G., N.V.), Université Paris Cité, France
- Cordeliers Research Centre, INSERM, IMMEDIAB Laboratory, Sorbonne Université (J.-B.J., D.G., M.D., B.T.V., C.B., E.C., L.O., C.P., K.K., D.C.T., T.E., R.B., J.-P.R., L.P., R.R., C.V., F.A., J.-F.G., N.V.), Université Paris Cité, France
- Diabetes Institute (J.-B.J., D.G., M.D., B.T.V., C.B., E.C., L.O., C.P., K.K., D.C.T., T.E., R.B., T.V.-T., J.-P.R., L.P., R.R., C.V., F.A., J.-F.G., N.V.), Université Paris Cité, France
| | - Camille Blériot
- INSERM, Necker Enfants Malades (INEM), INSERM U1151, CNRS UMR 8253, IMMEDIAB Laboratory (J.-B.J., D.G., M.D., B.T.V., C.B., E.C., L.O., I.N., C.P., K.K., D.C.T., T.E., R.B., J.-P.R., L.P., R.R., C.V., F.A., J.-F.G., N.V.), Université Paris Cité, France
- Cordeliers Research Centre, INSERM, IMMEDIAB Laboratory, Sorbonne Université (J.-B.J., D.G., M.D., B.T.V., C.B., E.C., L.O., C.P., K.K., D.C.T., T.E., R.B., J.-P.R., L.P., R.R., C.V., F.A., J.-F.G., N.V.), Université Paris Cité, France
- Diabetes Institute (J.-B.J., D.G., M.D., B.T.V., C.B., E.C., L.O., C.P., K.K., D.C.T., T.E., R.B., T.V.-T., J.-P.R., L.P., R.R., C.V., F.A., J.-F.G., N.V.), Université Paris Cité, France
| | - Elena Carcarino
- INSERM, Necker Enfants Malades (INEM), INSERM U1151, CNRS UMR 8253, IMMEDIAB Laboratory (J.-B.J., D.G., M.D., B.T.V., C.B., E.C., L.O., I.N., C.P., K.K., D.C.T., T.E., R.B., J.-P.R., L.P., R.R., C.V., F.A., J.-F.G., N.V.), Université Paris Cité, France
- Cordeliers Research Centre, INSERM, IMMEDIAB Laboratory, Sorbonne Université (J.-B.J., D.G., M.D., B.T.V., C.B., E.C., L.O., C.P., K.K., D.C.T., T.E., R.B., J.-P.R., L.P., R.R., C.V., F.A., J.-F.G., N.V.), Université Paris Cité, France
- Diabetes Institute (J.-B.J., D.G., M.D., B.T.V., C.B., E.C., L.O., C.P., K.K., D.C.T., T.E., R.B., T.V.-T., J.-P.R., L.P., R.R., C.V., F.A., J.-F.G., N.V.), Université Paris Cité, France
| | - Joe De Keizer
- Nantes Université, CHU Nantes, CNRS, INSERM, l'institut du thorax, Nantes, France (J.D.K., S.H.)
| | - Lucie Orliaguet
- INSERM, Necker Enfants Malades (INEM), INSERM U1151, CNRS UMR 8253, IMMEDIAB Laboratory (J.-B.J., D.G., M.D., B.T.V., C.B., E.C., L.O., I.N., C.P., K.K., D.C.T., T.E., R.B., J.-P.R., L.P., R.R., C.V., F.A., J.-F.G., N.V.), Université Paris Cité, France
- Cordeliers Research Centre, INSERM, IMMEDIAB Laboratory, Sorbonne Université (J.-B.J., D.G., M.D., B.T.V., C.B., E.C., L.O., C.P., K.K., D.C.T., T.E., R.B., J.-P.R., L.P., R.R., C.V., F.A., J.-F.G., N.V.), Université Paris Cité, France
- Diabetes Institute (J.-B.J., D.G., M.D., B.T.V., C.B., E.C., L.O., C.P., K.K., D.C.T., T.E., R.B., T.V.-T., J.-P.R., L.P., R.R., C.V., F.A., J.-F.G., N.V.), Université Paris Cité, France
| | - Ivan Nemazanyy
- INSERM, Necker Enfants Malades (INEM), INSERM U1151, CNRS UMR 8253, IMMEDIAB Laboratory (J.-B.J., D.G., M.D., B.T.V., C.B., E.C., L.O., I.N., C.P., K.K., D.C.T., T.E., R.B., J.-P.R., L.P., R.R., C.V., F.A., J.-F.G., N.V.), Université Paris Cité, France
| | - Charline Potier
- INSERM, Necker Enfants Malades (INEM), INSERM U1151, CNRS UMR 8253, IMMEDIAB Laboratory (J.-B.J., D.G., M.D., B.T.V., C.B., E.C., L.O., I.N., C.P., K.K., D.C.T., T.E., R.B., J.-P.R., L.P., R.R., C.V., F.A., J.-F.G., N.V.), Université Paris Cité, France
- Cordeliers Research Centre, INSERM, IMMEDIAB Laboratory, Sorbonne Université (J.-B.J., D.G., M.D., B.T.V., C.B., E.C., L.O., C.P., K.K., D.C.T., T.E., R.B., J.-P.R., L.P., R.R., C.V., F.A., J.-F.G., N.V.), Université Paris Cité, France
- Diabetes Institute (J.-B.J., D.G., M.D., B.T.V., C.B., E.C., L.O., C.P., K.K., D.C.T., T.E., R.B., T.V.-T., J.-P.R., L.P., R.R., C.V., F.A., J.-F.G., N.V.), Université Paris Cité, France
| | - Kennan Khider
- INSERM, Necker Enfants Malades (INEM), INSERM U1151, CNRS UMR 8253, IMMEDIAB Laboratory (J.-B.J., D.G., M.D., B.T.V., C.B., E.C., L.O., I.N., C.P., K.K., D.C.T., T.E., R.B., J.-P.R., L.P., R.R., C.V., F.A., J.-F.G., N.V.), Université Paris Cité, France
- Cordeliers Research Centre, INSERM, IMMEDIAB Laboratory, Sorbonne Université (J.-B.J., D.G., M.D., B.T.V., C.B., E.C., L.O., C.P., K.K., D.C.T., T.E., R.B., J.-P.R., L.P., R.R., C.V., F.A., J.-F.G., N.V.), Université Paris Cité, France
- Diabetes Institute (J.-B.J., D.G., M.D., B.T.V., C.B., E.C., L.O., C.P., K.K., D.C.T., T.E., R.B., T.V.-T., J.-P.R., L.P., R.R., C.V., F.A., J.-F.G., N.V.), Université Paris Cité, France
| | - Dorothy Chepngenoh Tonui
- INSERM, Necker Enfants Malades (INEM), INSERM U1151, CNRS UMR 8253, IMMEDIAB Laboratory (J.-B.J., D.G., M.D., B.T.V., C.B., E.C., L.O., I.N., C.P., K.K., D.C.T., T.E., R.B., J.-P.R., L.P., R.R., C.V., F.A., J.-F.G., N.V.), Université Paris Cité, France
- Cordeliers Research Centre, INSERM, IMMEDIAB Laboratory, Sorbonne Université (J.-B.J., D.G., M.D., B.T.V., C.B., E.C., L.O., C.P., K.K., D.C.T., T.E., R.B., J.-P.R., L.P., R.R., C.V., F.A., J.-F.G., N.V.), Université Paris Cité, France
- Diabetes Institute (J.-B.J., D.G., M.D., B.T.V., C.B., E.C., L.O., C.P., K.K., D.C.T., T.E., R.B., T.V.-T., J.-P.R., L.P., R.R., C.V., F.A., J.-F.G., N.V.), Université Paris Cité, France
| | - Tina Ejlalmanesh
- INSERM, Necker Enfants Malades (INEM), INSERM U1151, CNRS UMR 8253, IMMEDIAB Laboratory (J.-B.J., D.G., M.D., B.T.V., C.B., E.C., L.O., I.N., C.P., K.K., D.C.T., T.E., R.B., J.-P.R., L.P., R.R., C.V., F.A., J.-F.G., N.V.), Université Paris Cité, France
- Cordeliers Research Centre, INSERM, IMMEDIAB Laboratory, Sorbonne Université (J.-B.J., D.G., M.D., B.T.V., C.B., E.C., L.O., C.P., K.K., D.C.T., T.E., R.B., J.-P.R., L.P., R.R., C.V., F.A., J.-F.G., N.V.), Université Paris Cité, France
- Diabetes Institute (J.-B.J., D.G., M.D., B.T.V., C.B., E.C., L.O., C.P., K.K., D.C.T., T.E., R.B., T.V.-T., J.-P.R., L.P., R.R., C.V., F.A., J.-F.G., N.V.), Université Paris Cité, France
| | - Raphaelle Ballaire
- INSERM, Necker Enfants Malades (INEM), INSERM U1151, CNRS UMR 8253, IMMEDIAB Laboratory (J.-B.J., D.G., M.D., B.T.V., C.B., E.C., L.O., I.N., C.P., K.K., D.C.T., T.E., R.B., J.-P.R., L.P., R.R., C.V., F.A., J.-F.G., N.V.), Université Paris Cité, France
- Cordeliers Research Centre, INSERM, IMMEDIAB Laboratory, Sorbonne Université (J.-B.J., D.G., M.D., B.T.V., C.B., E.C., L.O., C.P., K.K., D.C.T., T.E., R.B., J.-P.R., L.P., R.R., C.V., F.A., J.-F.G., N.V.), Université Paris Cité, France
- Diabetes Institute (J.-B.J., D.G., M.D., B.T.V., C.B., E.C., L.O., C.P., K.K., D.C.T., T.E., R.B., T.V.-T., J.-P.R., L.P., R.R., C.V., F.A., J.-F.G., N.V.), Université Paris Cité, France
| | - Hendrick Mambu Mambueni
- Genomics platform UFR Simone Veil 1173; U, University of Versailles Paris-Saclay; Inserm UMR 1173 (H.M.M., H.-J.G.)
| | - Stéphane Germain
- Center for Interdisciplinary Research in Biology (CIRB), College de France, CNRS, INSERM, Université PSL, Paris, France (S.G.)
| | - Bénédicte Gaborit
- C2VN, INRAE, INSERM, Aix Marseille University, Marseille, France (B.G.)
- Department of Endocrinology, Metabolic Diseases and Nutrition, Pôle ENDO, AP-HM, Marseille, France (B.G.)
| | - Tiphaine Vidal-Trécan
- Diabetes Institute (J.-B.J., D.G., M.D., B.T.V., C.B., E.C., L.O., C.P., K.K., D.C.T., T.E., R.B., T.V.-T., J.-P.R., L.P., R.R., C.V., F.A., J.-F.G., N.V.), Université Paris Cité, France
- Diabetology, Endocrinology and Nutrition Department, Lariboisière Hospital, Fédération de Diabétologie, France (J.-B.J., T.V.-T., J.-P.R., J.-F.G.)
| | - Jean-Pierre Riveline
- INSERM, Necker Enfants Malades (INEM), INSERM U1151, CNRS UMR 8253, IMMEDIAB Laboratory (J.-B.J., D.G., M.D., B.T.V., C.B., E.C., L.O., I.N., C.P., K.K., D.C.T., T.E., R.B., J.-P.R., L.P., R.R., C.V., F.A., J.-F.G., N.V.), Université Paris Cité, France
- Cordeliers Research Centre, INSERM, IMMEDIAB Laboratory, Sorbonne Université (J.-B.J., D.G., M.D., B.T.V., C.B., E.C., L.O., C.P., K.K., D.C.T., T.E., R.B., J.-P.R., L.P., R.R., C.V., F.A., J.-F.G., N.V.), Université Paris Cité, France
- Diabetes Institute (J.-B.J., D.G., M.D., B.T.V., C.B., E.C., L.O., C.P., K.K., D.C.T., T.E., R.B., T.V.-T., J.-P.R., L.P., R.R., C.V., F.A., J.-F.G., N.V.), Université Paris Cité, France
- Diabetology, Endocrinology and Nutrition Department, Lariboisière Hospital, Fédération de Diabétologie, France (J.-B.J., T.V.-T., J.-P.R., J.-F.G.)
| | - Henri-Jean Garchon
- Genomics platform UFR Simone Veil 1173; U, University of Versailles Paris-Saclay; Inserm UMR 1173 (H.M.M., H.-J.G.)
| | - François Fenaille
- Université Paris-Saclay, CEA, INRAE, Département Médicaments et Technologies pour la Santé (MTS), MetaboHUB, France (F.F., F.C.)
| | - Sophie Lemoine
- Genomics core facility, Institut de Biologie de l'ENS (IBENS), Département de biologie, École Normale Supérieure, CNRS, INSERM, Université PSL, Paris, France (S.L.)
| | - Aurélie Carlier
- Diabetology and Endocrinology Department, Bichat Hospital, Fédération de Diabétologie, France (L.P., A.C., R.R.)
| | - Florence Castelli
- Université Paris-Saclay, CEA, INRAE, Département Médicaments et Technologies pour la Santé (MTS), MetaboHUB, France (F.F., F.C.)
| | - Louis Potier
- INSERM, Necker Enfants Malades (INEM), INSERM U1151, CNRS UMR 8253, IMMEDIAB Laboratory (J.-B.J., D.G., M.D., B.T.V., C.B., E.C., L.O., I.N., C.P., K.K., D.C.T., T.E., R.B., J.-P.R., L.P., R.R., C.V., F.A., J.-F.G., N.V.), Université Paris Cité, France
- Cordeliers Research Centre, INSERM, IMMEDIAB Laboratory, Sorbonne Université (J.-B.J., D.G., M.D., B.T.V., C.B., E.C., L.O., C.P., K.K., D.C.T., T.E., R.B., J.-P.R., L.P., R.R., C.V., F.A., J.-F.G., N.V.), Université Paris Cité, France
- Diabetes Institute (J.-B.J., D.G., M.D., B.T.V., C.B., E.C., L.O., C.P., K.K., D.C.T., T.E., R.B., T.V.-T., J.-P.R., L.P., R.R., C.V., F.A., J.-F.G., N.V.), Université Paris Cité, France
- Diabetology and Endocrinology Department, Bichat Hospital, Fédération de Diabétologie, France (L.P., A.C., R.R.)
| | - David Masson
- INSERM, LNC UMR1231, Dijon, France (D.M.)
- University of Bourgogne and Franche-Comté, LNC UMR1231, Dijon, France (D.M.)
- FCS Bourgogne-Franche Comté, LipSTIC LabEx, Dijon, France (D.M.)
- Plateau Automatisé de Biochimie, Dijon University Hospital, France (D.M.)
| | - Ronan Roussel
- INSERM, Necker Enfants Malades (INEM), INSERM U1151, CNRS UMR 8253, IMMEDIAB Laboratory (J.-B.J., D.G., M.D., B.T.V., C.B., E.C., L.O., I.N., C.P., K.K., D.C.T., T.E., R.B., J.-P.R., L.P., R.R., C.V., F.A., J.-F.G., N.V.), Université Paris Cité, France
- Cordeliers Research Centre, INSERM, IMMEDIAB Laboratory, Sorbonne Université (J.-B.J., D.G., M.D., B.T.V., C.B., E.C., L.O., C.P., K.K., D.C.T., T.E., R.B., J.-P.R., L.P., R.R., C.V., F.A., J.-F.G., N.V.), Université Paris Cité, France
- Diabetes Institute (J.-B.J., D.G., M.D., B.T.V., C.B., E.C., L.O., C.P., K.K., D.C.T., T.E., R.B., T.V.-T., J.-P.R., L.P., R.R., C.V., F.A., J.-F.G., N.V.), Université Paris Cité, France
- Diabetology and Endocrinology Department, Bichat Hospital, Fédération de Diabétologie, France (L.P., A.C., R.R.)
| | - Claire Vandiedonck
- INSERM, Necker Enfants Malades (INEM), INSERM U1151, CNRS UMR 8253, IMMEDIAB Laboratory (J.-B.J., D.G., M.D., B.T.V., C.B., E.C., L.O., I.N., C.P., K.K., D.C.T., T.E., R.B., J.-P.R., L.P., R.R., C.V., F.A., J.-F.G., N.V.), Université Paris Cité, France
- Cordeliers Research Centre, INSERM, IMMEDIAB Laboratory, Sorbonne Université (J.-B.J., D.G., M.D., B.T.V., C.B., E.C., L.O., C.P., K.K., D.C.T., T.E., R.B., J.-P.R., L.P., R.R., C.V., F.A., J.-F.G., N.V.), Université Paris Cité, France
- Diabetes Institute (J.-B.J., D.G., M.D., B.T.V., C.B., E.C., L.O., C.P., K.K., D.C.T., T.E., R.B., T.V.-T., J.-P.R., L.P., R.R., C.V., F.A., J.-F.G., N.V.), Université Paris Cité, France
| | - Samy Hadjadj
- Nantes Université, CHU Nantes, CNRS, INSERM, l'institut du thorax, Nantes, France (J.D.K., S.H.)
| | - Fawaz Alzaid
- INSERM, Necker Enfants Malades (INEM), INSERM U1151, CNRS UMR 8253, IMMEDIAB Laboratory (J.-B.J., D.G., M.D., B.T.V., C.B., E.C., L.O., I.N., C.P., K.K., D.C.T., T.E., R.B., J.-P.R., L.P., R.R., C.V., F.A., J.-F.G., N.V.), Université Paris Cité, France
- Cordeliers Research Centre, INSERM, IMMEDIAB Laboratory, Sorbonne Université (J.-B.J., D.G., M.D., B.T.V., C.B., E.C., L.O., C.P., K.K., D.C.T., T.E., R.B., J.-P.R., L.P., R.R., C.V., F.A., J.-F.G., N.V.), Université Paris Cité, France
- Diabetes Institute (J.-B.J., D.G., M.D., B.T.V., C.B., E.C., L.O., C.P., K.K., D.C.T., T.E., R.B., T.V.-T., J.-P.R., L.P., R.R., C.V., F.A., J.-F.G., N.V.), Université Paris Cité, France
- Dasman Diabetes Institute, Kuwait (F.A.)
| | - Jean-François Gautier
- INSERM, Necker Enfants Malades (INEM), INSERM U1151, CNRS UMR 8253, IMMEDIAB Laboratory (J.-B.J., D.G., M.D., B.T.V., C.B., E.C., L.O., I.N., C.P., K.K., D.C.T., T.E., R.B., J.-P.R., L.P., R.R., C.V., F.A., J.-F.G., N.V.), Université Paris Cité, France
- Cordeliers Research Centre, INSERM, IMMEDIAB Laboratory, Sorbonne Université (J.-B.J., D.G., M.D., B.T.V., C.B., E.C., L.O., C.P., K.K., D.C.T., T.E., R.B., J.-P.R., L.P., R.R., C.V., F.A., J.-F.G., N.V.), Université Paris Cité, France
- Diabetes Institute (J.-B.J., D.G., M.D., B.T.V., C.B., E.C., L.O., C.P., K.K., D.C.T., T.E., R.B., T.V.-T., J.-P.R., L.P., R.R., C.V., F.A., J.-F.G., N.V.), Université Paris Cité, France
- Diabetology, Endocrinology and Nutrition Department, Lariboisière Hospital, Fédération de Diabétologie, France (J.-B.J., T.V.-T., J.-P.R., J.-F.G.)
| | - Nicolas Venteclef
- INSERM, Necker Enfants Malades (INEM), INSERM U1151, CNRS UMR 8253, IMMEDIAB Laboratory (J.-B.J., D.G., M.D., B.T.V., C.B., E.C., L.O., I.N., C.P., K.K., D.C.T., T.E., R.B., J.-P.R., L.P., R.R., C.V., F.A., J.-F.G., N.V.), Université Paris Cité, France
- Cordeliers Research Centre, INSERM, IMMEDIAB Laboratory, Sorbonne Université (J.-B.J., D.G., M.D., B.T.V., C.B., E.C., L.O., C.P., K.K., D.C.T., T.E., R.B., J.-P.R., L.P., R.R., C.V., F.A., J.-F.G., N.V.), Université Paris Cité, France
- Diabetes Institute (J.-B.J., D.G., M.D., B.T.V., C.B., E.C., L.O., C.P., K.K., D.C.T., T.E., R.B., T.V.-T., J.-P.R., L.P., R.R., C.V., F.A., J.-F.G., N.V.), Université Paris Cité, France
| |
Collapse
|
38
|
Zheng C, Chen S, Deng YY, Qian XP, Chen YY, Hong CZ, Zeng YF, Li QM, Pan LH, Luo JP, Li XY, Zha XQ. Purification, structural characteristics and anti-atherosclerosis activity of a novel green tea polysaccharide. Int J Biol Macromol 2024; 254:127705. [PMID: 37913884 DOI: 10.1016/j.ijbiomac.2023.127705] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2023] [Revised: 10/01/2023] [Accepted: 10/19/2023] [Indexed: 11/03/2023]
Abstract
A new homogeneous polysaccharide (TPS3A) was isolated and purified from Tianzhu Xianyue fried green tea by DEAE-52 cellulose and Sephacryl S-500 column chromatography. Structural characterization indicated that TPS3A mainly consisted of arabinose, galactose, galacturonic acid and rhamnose in a molar ratio of 5.84: 4.15: 2.06: 1, with an average molecular weight of 1.596 × 104 kDa. The structure of TPS3A was characterized as a repeating unit consisting of 1,3-Galp, 1,4-Galp, 1,3,6-Galp, 1,3-Araf, 1,5-Araf, 1,2,4-Rhap and 1-GalpA, with two branches on the C6 of 1,3,6-Galp and C2 of 1,2,4-Rhap, respectively. To investigate the preventive effects of TPS3A on atherosclerosis, TPS3A was administered orally to ApoE-deficient (ApoE-/-) mice. Results revealed that TPS3A intervention could effectively delay the atherosclerotic plaque progression, modulate dyslipidemia, and reduce the transformation of vascular smooth muscle cells (VSMCs) from contractile phenotype to synthetic phenotype by activating the expression of contractile marker alpha-smooth muscle actin (α-SMA) and inhibiting the expression of synthetic marker osteopontin (OPN) in high-fat diet-induced ApoE-/- mice. Our findings suggested that TPS3A markedly alleviated atherosclerosis by regulating dyslipidemia and phenotypic transition of VSMCs, and might be used as a novel functional ingredient to promote cardiovascular health.
Collapse
Affiliation(s)
- Chao Zheng
- Engineering Research Centre of Bioprocess of Ministry of Education, School of Food and Biological Engineering, Hefei University of Technology, No. 193 Tunxi Road, Hefei 230009, People's Republic of China; School of Food and Biological Engineering, Hefei University of Technology, No. 193 Tunxi Road, Hefei 230009, People's Republic of China
| | - Shun Chen
- Engineering Research Centre of Bioprocess of Ministry of Education, School of Food and Biological Engineering, Hefei University of Technology, No. 193 Tunxi Road, Hefei 230009, People's Republic of China; School of Food and Biological Engineering, Hefei University of Technology, No. 193 Tunxi Road, Hefei 230009, People's Republic of China
| | - Yuan-Yuan Deng
- Sericultural and Agri-Food Research Institute, Guangdong Academy of Agricultural Sciences, Guangzhou 510610, People's Republic of China
| | - Xin-Ping Qian
- Engineering Research Centre of Bioprocess of Ministry of Education, School of Food and Biological Engineering, Hefei University of Technology, No. 193 Tunxi Road, Hefei 230009, People's Republic of China; School of Food and Biological Engineering, Hefei University of Technology, No. 193 Tunxi Road, Hefei 230009, People's Republic of China
| | - Ying-Ying Chen
- Engineering Research Centre of Bioprocess of Ministry of Education, School of Food and Biological Engineering, Hefei University of Technology, No. 193 Tunxi Road, Hefei 230009, People's Republic of China; School of Food and Biological Engineering, Hefei University of Technology, No. 193 Tunxi Road, Hefei 230009, People's Republic of China
| | - Cheng-Zhi Hong
- Engineering Research Centre of Bioprocess of Ministry of Education, School of Food and Biological Engineering, Hefei University of Technology, No. 193 Tunxi Road, Hefei 230009, People's Republic of China; School of Food and Biological Engineering, Hefei University of Technology, No. 193 Tunxi Road, Hefei 230009, People's Republic of China
| | - Ya-Fan Zeng
- Engineering Research Centre of Bioprocess of Ministry of Education, School of Food and Biological Engineering, Hefei University of Technology, No. 193 Tunxi Road, Hefei 230009, People's Republic of China; School of Food and Biological Engineering, Hefei University of Technology, No. 193 Tunxi Road, Hefei 230009, People's Republic of China
| | - Qiang-Ming Li
- Engineering Research Centre of Bioprocess of Ministry of Education, School of Food and Biological Engineering, Hefei University of Technology, No. 193 Tunxi Road, Hefei 230009, People's Republic of China; School of Food and Biological Engineering, Hefei University of Technology, No. 193 Tunxi Road, Hefei 230009, People's Republic of China
| | - Li-Hua Pan
- Engineering Research Centre of Bioprocess of Ministry of Education, School of Food and Biological Engineering, Hefei University of Technology, No. 193 Tunxi Road, Hefei 230009, People's Republic of China; School of Food and Biological Engineering, Hefei University of Technology, No. 193 Tunxi Road, Hefei 230009, People's Republic of China
| | - Jian-Ping Luo
- Engineering Research Centre of Bioprocess of Ministry of Education, School of Food and Biological Engineering, Hefei University of Technology, No. 193 Tunxi Road, Hefei 230009, People's Republic of China; School of Food and Biological Engineering, Hefei University of Technology, No. 193 Tunxi Road, Hefei 230009, People's Republic of China
| | - Xue-Ying Li
- Engineering Research Centre of Bioprocess of Ministry of Education, School of Food and Biological Engineering, Hefei University of Technology, No. 193 Tunxi Road, Hefei 230009, People's Republic of China; School of Food and Biological Engineering, Hefei University of Technology, No. 193 Tunxi Road, Hefei 230009, People's Republic of China.
| | - Xue-Qiang Zha
- Engineering Research Centre of Bioprocess of Ministry of Education, School of Food and Biological Engineering, Hefei University of Technology, No. 193 Tunxi Road, Hefei 230009, People's Republic of China; School of Food and Biological Engineering, Hefei University of Technology, No. 193 Tunxi Road, Hefei 230009, People's Republic of China.
| |
Collapse
|
39
|
Zhang Y, Xing Z, Deng A. Unveiling the predictive capacity of inflammatory and platelet markers for central retinal artery occlusion. Thromb Res 2023; 232:108-112. [PMID: 37976730 DOI: 10.1016/j.thromres.2023.11.004] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2023] [Revised: 10/19/2023] [Accepted: 11/02/2023] [Indexed: 11/19/2023]
Abstract
OBJECTIVES Cell counting in peripheral blood samples and their combinations have gained wide usage in clinical research due to their convenient and minimally invasive sampling method. This study aims to evaluate the predictive value of neutrophil-to-lymphocyte ratio (NLR), systemic Immunoinflammatory Index (SII), and systemic Inflammatory Response Index (SIRI), and platelet distribution width (PDW) for the occurrence of non-arteritic Central retinal artery occlusion (NA-CRAO). METHODS We included 123 patients diagnosed with NA-CRAO and 120 age-, sex- and blood pressure-matched individuals in this study. All participants underwent a comprehensive ophthalmic assessment. Peripheral blood samples were retrospectively analysed to obtain patients' blood counts and platelet-related indices, and further NLR, SII and SIRI were calculated. RESULTS NLR, SII, SIRI, and PDW were all found to be elevated and significantly different in NA-CRAO patients compared to controls (p < 0.05). Notably, elevated NLR and PDW were identified as independent influences on the development of NA-CRAO, with a combined predicted AUC of 0.876. CONCLUSION The utilization of NLR and PDW in NA-CRAO prediction may prove to be more effective compared to SII and SIRI.
Collapse
Affiliation(s)
- Yupeng Zhang
- Department of Ophthalmology, Affiliated Hospital of Weifang Medical University, School of Clinical Medicine, Weifang Medical University, 261000 Weifang, Shandong, China
| | - Zhen Xing
- Department of Ophthalmology, Affiliated Hospital of Weifang Medical University, School of Clinical Medicine, Weifang Medical University, 261000 Weifang, Shandong, China
| | - Aijun Deng
- Department of Ophthalmology, Affiliated Hospital of Weifang Medical University, 261000 Weifang, Shandong, China.
| |
Collapse
|
40
|
Separham A, Aslan-abadi N, Sedigh H, Javan-ajdadi R, Mehravani K. Assessment of the Prognostic Value of Monocyte-to-HDL Ratio in ST-Elevation Myocardial Infarction Patients Undergoing Primary Percutaneous Coronary Intervention. Galen Med J 2023; 12:1-9. [PMID: 39553411 PMCID: PMC11568425 DOI: 10.31661/gmj.v12i0.3126] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2023] [Revised: 08/04/2023] [Accepted: 08/27/2023] [Indexed: 11/19/2024] Open
Abstract
BACKGROUND The purpose of this study was to assess the prognostic value of the monocyte-to-high-density lipoprotein ratio (MHR) as a marker of inflammation in patients diagnosed with ST-segment elevation myocardial infarction (STEMI) who underwent primary percutaneous coronary intervention (PCI). MATERIALS AND METHODS This retrospective cross-sectional study was conducted on patients with a diagnosis of STEMI who underwent PCI between March 2021 and March 2022 at Madani Training and Research Hospital in Tabriz, Iran. Data regarding clinical and demographic properties, and laboratory parameters were obtained from medical records. Patients were categorized into two groups according to the median of admission MHR. RESULTS The study population consisted of 652 patients, 378 males (58%), and 275 females (42%), with a median age of 68 years (interquartile range: 57-77). Results showed that groups with higher MHR (15.59) had higher rates of in-hospital mortality and higher major adverse cardiovascular events (MACEs) in comparison with the group featuring lower MHR (15.59). Receiver operating characteristic (ROC) curves demonstrated that MHR could predict in-hospital mortality with a 75.7% sensitivity and 53.5% specificity, as well as predict MACE with 60.2% sensitivity and 59.7% specificity. Multivariate analyses indicated that MHR is an independent predictor of both in-hospital mortality (OR 1.05, 95% CI 1.02-1.08, P=0.002) and MACE (OR 1.05, 95% CI 1.02-1.08, P0.001). CONCLUSION This research indicated that the rise in MHR was independently associated with a higher risk of MACE and in-hospital mortality in STEMI patients undergoing primary PCI.
Collapse
Affiliation(s)
- Ahmad Separham
- Cardiovascular Research Center, Faculty of Medicine, Tabriz University of Medical
Sciences, Tabriz, Iran
| | - Naser Aslan-abadi
- Cardiovascular Research Center, Faculty of Medicine, Tabriz University of Medical
Sciences, Tabriz, Iran
| | - Hamid Sedigh
- Cardiovascular Research Center, Faculty of Medicine, Tabriz University of Medical
Sciences, Tabriz, Iran
| | - Reza Javan-ajdadi
- Cardiovascular Research Center, Faculty of Medicine, Tabriz University of Medical
Sciences, Tabriz, Iran
| | - Kazem Mehravani
- Cardiovascular Research Center, Faculty of Medicine, Tabriz University of Medical
Sciences, Tabriz, Iran
| |
Collapse
|
41
|
Separham A, Aslan-abadi N, Sedigh H, Javan-ajdadi R, Mehravani K. Assessment of the Prognostic Value of Monocyte-to-HDL Ratio in ST-Elevation Myocardial Infarction Patients Undergoing Primary Percutaneous Coronary Intervention. Galen Med J 2023; 12:e3126. [PMID: 39553411 PMCID: PMC11568425 DOI: 10.31661/gmj.v12i.3126] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2023] [Revised: 08/04/2023] [Accepted: 08/27/2023] [Indexed: 03/04/2025] Open
Abstract
BACKGROUND The purpose of this study was to assess the prognostic value of the monocyte-to-high-density lipoprotein ratio (MHR) as a marker of inflammation in patients diagnosed with ST-segment elevation myocardial infarction (STEMI) who underwent primary percutaneous coronary intervention (PCI). MATERIALS AND METHODS This retrospective cross-sectional study was conducted on patients with a diagnosis of STEMI who underwent PCI between March 2021 and March 2022 at Madani Training and Research Hospital in Tabriz, Iran. Data regarding clinical and demographic properties, and laboratory parameters were obtained from medical records. Patients were categorized into two groups according to the median of admission MHR. RESULTS The study population consisted of 652 patients, 378 males (58%), and 275 females (42%), with a median age of 68 years (interquartile range: 57-77). Results showed that groups with higher MHR (15.59) had higher rates of in-hospital mortality and higher major adverse cardiovascular events (MACEs) in comparison with the group featuring lower MHR (15.59). Receiver operating characteristic (ROC) curves demonstrated that MHR could predict in-hospital mortality with a 75.7% sensitivity and 53.5% specificity, as well as predict MACE with 60.2% sensitivity and 59.7% specificity. Multivariate analyses indicated that MHR is an independent predictor of both in-hospital mortality (OR 1.05, 95% CI 1.02-1.08, P=0.002) and MACE (OR 1.05, 95% CI 1.02-1.08, P0.001). CONCLUSION This research indicated that the rise in MHR was independently associated with a higher risk of MACE and in-hospital mortality in STEMI patients undergoing primary PCI.
Collapse
Affiliation(s)
- Ahmad Separham
- Cardiovascular Research Center, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Naser Aslan-abadi
- Cardiovascular Research Center, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Hamid Sedigh
- Cardiovascular Research Center, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Reza Javan-ajdadi
- Cardiovascular Research Center, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Kazem Mehravani
- Cardiovascular Research Center, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| |
Collapse
|
42
|
Li Y, Ma JQ, Wang CC, Zhou J, Sun YD, Wei XL, Zhao ZQ. Ferroptosis: A potential target of macrophages in plaque vulnerability. Open Life Sci 2023; 18:20220722. [PMID: 37791060 PMCID: PMC10543703 DOI: 10.1515/biol-2022-0722] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2023] [Revised: 08/04/2023] [Accepted: 08/16/2023] [Indexed: 10/05/2023] Open
Abstract
Plaque vulnerability has been the subject of several recent studies aimed at reducing the risk of stroke and carotid artery stenosis. Atherosclerotic plaque development is a complex process involving inflammation mediated by macrophages. Plaques become more vulnerable when the equilibrium between macrophage recruitment and clearance is disturbed. Lipoperoxides, which are affected by iron levels in cells, are responsible for the cell death seen in ferroptosis. Ferroptosis results from lipoperoxide-induced mitochondrial membrane toxicity. Atherosclerosis in ApoE(-/-) mice is reduced when ferroptosis is inhibited and iron intake is limited. Single-cell sequencing revealed that a ferroptosis-related gene was substantially expressed in atherosclerosis-modeled macrophages. Since ferroptosis can be regulated, it offers hope as a non-invasive method of treating carotid plaque. In this study, we discuss the role of ferroptosis in atherosclerotic plaque vulnerability, including its mechanism, regulation, and potential future research directions.
Collapse
Affiliation(s)
- Yu Li
- Department of Vascular Surgery, Changhai Hospital, The PLA Naval Medical University, 168 Changhai Road, Shanghai200433, China
| | - Ji-Qing Ma
- Department of Vascular Surgery, Changhai Hospital, The PLA Naval Medical University, 168 Changhai Road, Shanghai200433, China
| | - Chao-Chen Wang
- Department of Vascular Surgery, Changhai Hospital, The PLA Naval Medical University, 168 Changhai Road, Shanghai200433, China
| | - Jian Zhou
- Department of Vascular Surgery, Changhai Hospital, The PLA Naval Medical University, 168 Changhai Road, Shanghai200433, China
| | - Yu-Dong Sun
- Department of General Surgery, Jinling Hospital, Medical School of Nanjing University,
Nanjing201411, China
| | - Xiao-Long Wei
- Department of Vascular Surgery, Changhai Hospital, The PLA Naval Medical University, 168 Changhai Road, Shanghai200433, China
| | - Zhi-Qing Zhao
- Department of Vascular Surgery, Changhai Hospital, The PLA Naval Medical University, 168 Changhai Road, Shanghai200433, China
| |
Collapse
|
43
|
Yang XF, Shang DJ. The role of peroxisome proliferator-activated receptor γ in lipid metabolism and inflammation in atherosclerosis. Cell Biol Int 2023; 47:1469-1487. [PMID: 37369936 DOI: 10.1002/cbin.12065] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2022] [Revised: 05/09/2023] [Accepted: 06/18/2023] [Indexed: 06/29/2023]
Abstract
Cardiovascular disease events are the result of functional and structural abnormalities in the arteries and heart. Atherosclerosis is the main cause and pathological basis of cardiovascular diseases. Atherosclerosis is a multifactorial disease associated with dyslipidemia, inflammation, and oxidative stress, among which dyslipidemia and chronic inflammation occur in all processes. Under the influence of lipoproteins, the arterial intima causes inflammation, necrosis, fibrosis, and calcification, leading to plaque formation in specific parts of the artery, which further develops into plaque rupture and secondary thrombosis. Foam cell formation from macrophages is an early event in the development of atherosclerosis. Lipid uptake causes a vascular inflammatory response, and persistent inflammatory infiltration in the lesion area further promotes the development of the disease. Inhibition of macrophage differentiation into foam cell and reduction of the level of proinflammatory factors in macrophages can effectively alleviate the occurrence and development of atherosclerosis. Peroxisome proliferator-activated receptor γ (PPARγ) is a ligand-activated nuclear receptor that plays an important antiatherosclerotic role by regulating triglyceride metabolism, lipid uptake, cholesterol efflux, macrophage polarity, and inhibiting inflammatory signaling pathways. In addition, PPARγ shifts its binding to ligands and co-activators or co-repressors of transcription of target genes through posttranslational modification, thereby affecting the regulation of its downstream target genes. Many ligand agonists have also been developed targeting PPARγ. In this review, we summarized the role of PPARγ in lipid metabolism and inflammation in development of atherosclerosis, the posttranslational regulatory mechanism of PPARγ, and further discusses the value of PPARγ as an antiatherosclerosis target.
Collapse
Affiliation(s)
- Xue-Feng Yang
- School of Life Science, Liaoning Provincial Key Laboratory of Biotechnology and Drug Discovery, Liaoning Normal University, Dalian, China
- Department of Physiology, School of Basic Medical Sciences, Jinzhou Medical University, Jinzhou, China
| | - De-Jing Shang
- School of Life Science, Liaoning Provincial Key Laboratory of Biotechnology and Drug Discovery, Liaoning Normal University, Dalian, China
| |
Collapse
|
44
|
Song B, Zeng Y, Cao Y, Zhang J, Xu C, Pan Y, Zhao X, Liu J. Emerging role of METTL3 in inflammatory diseases: mechanisms and therapeutic applications. Front Immunol 2023; 14:1221609. [PMID: 37671161 PMCID: PMC10475571 DOI: 10.3389/fimmu.2023.1221609] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2023] [Accepted: 07/27/2023] [Indexed: 09/07/2023] Open
Abstract
Despite improvements in modern medical therapies, inflammatory diseases, such as atherosclerosis, diabetes, non-alcoholic fatty liver, chronic kidney diseases, and autoimmune diseases have high incidence rates, still threaten human health, and represent a huge financial burden. N6-methyladenosine (m6A) modification of RNA contributes to the pathogenesis of various diseases. As the most widely discussed m6A methyltransferase, the pathogenic role of METTL3 in inflammatory diseases has become a research hotspot, but there has been no comprehensive review of the topic. Here, we summarize the expression changes, modified target genes, and pathogenesis related to METTL3 in cardiovascular, metabolic, degenerative, immune, and infectious diseases, as well as tumors. In addition to epithelial cells, endothelial cells, and fibroblasts, METTL3 also regulates the function of inflammation-related immune cells, including macrophages, neutrophils, dendritic cells, Th17 cells, and NK cells. Regarding therapeutic applications, METTL3 serves as a target for the treatment of inflammatory diseases with natural plant drug components, such as emodin, cinnamaldehyde, total flavonoids of Abelmoschus manihot, and resveratrol. This review focuses on recent advances in the initiation, development, and therapeutic application of METTL3 in inflammatory diseases. Knowledge of the specific regulatory mechanisms involving METTL3 can help to deepen understanding of inflammatory diseases and lay the foundation for the development of precisely targeted drugs to address inflammatory processes.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - Jingbo Liu
- Department of Periodontics, School of Stomatology, China Medical University, Shenyang, China
| |
Collapse
|
45
|
Antunes MDSM, Sugiyama FHC, Gravina HD, Castro RC, Mercado FJR, de Lima JO, Fontanari C, Frantz FG. COVID-19 inactivated and non-replicating viral vector vaccines induce regulatory training phenotype in human monocytes under epigenetic control. Front Cell Infect Microbiol 2023; 13:1200789. [PMID: 37520439 PMCID: PMC10382685 DOI: 10.3389/fcimb.2023.1200789] [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: 04/05/2023] [Accepted: 05/26/2023] [Indexed: 08/01/2023] Open
Abstract
Background Trained immunity is the enhanced innate immune response resulting from exposure to pathogens or vaccines against an unrelated pathogen stimulus. Certain vaccines induce a memory like response in monocytes and NK cells, leading to modulation in cytokine production, metabolic changes, and modifications in histone patterns. Here, we hypothesized that vaccination against SARS-CoV-2 could induce the training of monocytes in addition to stimulating the adaptive immune response. Methods Therefore, we aimed to investigate the immunophenotyping, cytokine and metabolic profile of monocytes from individuals who were completely immunized with two doses of inactivated COVID-19 vaccine or non-replicating viral vector vaccine. Subsequently, we investigated the epigenetic mechanisms underlying monocyte immune training. As a model of inflammatorychallenge, to understand if the monocytes were trained by vaccination and how they were trained, cells were stimulated in vitro with the endotoxin LPS, an unrelated stimulus that would provoke the effects of training. Results When challenged in vitro, monocytes from vaccinated individuals produced less TNF-α and those who received inactivated vaccine produced less IL-6, whereas vaccination with non-replicating viral vector vaccine induced more IL-10. Inactivated vaccine increased classical monocyte frequency, and both groups showed higher CD163 expression, a hallmark of trained immunity. We observed increased expression of genes involved in glycolysis and reduced IRG1 expression in vaccinated subjects, a gene associated with the tolerance phenotype in monocytes. We observed that both vaccines reduced the chromatin accessibility of genes associated with the inflammatory response, the inactivated COVID-19 vaccine trained monocytes to a regulatory phenotype mediated by histone modifications in the IL6 and IL10 genes, while the non-replicating viral vector COVID-19 vaccine trained monocytes to a regulatory phenotype, mediated by histone modifications in the IL6, IL10, TNF, and CCL2 genes. Conclusions Our findings support the recognized importance of adopting vaccination against SARS CoV-2, which has been shown to be effective in enhancing the adaptive immune response against the virus and reducing mortality and morbidity rates. Here, we provide evidence that vaccination also modulates the innate immune response by controlling the detrimental inflammatory response to unrelated pathogen stimulation.
Collapse
|
46
|
Tian Z, Yang S. Integrating the characteristic genes of macrophage pseudotime analysis in single-cell RNA-seq to construct a prediction model of atherosclerosis. Aging (Albany NY) 2023; 15:6361-6379. [PMID: 37421595 PMCID: PMC10373969 DOI: 10.18632/aging.204856] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2023] [Accepted: 06/19/2023] [Indexed: 07/10/2023]
Abstract
BACKGROUND Macrophages play an important role in the occurrence and development of atherosclerosis. However, few existing studies have deliberately analyzed the changes in characteristic genes in the process of macrophage phenotype transformation. METHOD Carotid atherosclerotic plaque single-cell RNA (scRNA) sequencing data were analyzed to define the cells involved and determine their transcriptomic characteristics. KEGG enrichment analysis, CIBERSORT, ESTIMATE, support vector machine (SVM), random forest (RF), and weighted correlation network analysis (WGCNA) were applied to bulk sequencing data. All data were downloaded from Gene Expression Omnibus (GEO). RESULT Nine cell clusters were identified. M1 macrophages, M2 macrophages, and M2/M1 macrophages were identified as three clusters within the macrophages. According to pseudotime analysis, M2/M1 macrophages and M2 macrophages can be transformed into M1 macrophages. The ROC curve values of the six genes in the test group were statistically significant (AUC (IL1RN): 0.899, 95% CI: 0.764-0.990; AUC (NRP1): 0.817, 95% CI: 0.620-0.971; AUC (TAGLN): 0.846, 95% CI: 0.678-0.971; AUC (SPARCL1): 0.825, 95% CI: 0.620-0.988; AUC (EMP2): 0.808, 95% CI: 0.630-0.947; AUC (ACTA2): 0.784, 95% CI: 0.591-0.938). The atherosclerosis prediction model showed significant statistical significance in both the train group (AUC: 0.909, 95% CI: 0.842-0.967) and the test group (AUC: 0.812, 95% CI: 0.630-0.966). CONCLUSIONS IL1RNHigh M1, NRP1High M2, ACTA2High M2/M1, EMP2High M1/M1, SPACL1High M2/M1 and TAGLNHigh M2/M1 macrophages play key roles in the occurrence and development of arterial atherosclerosis. These marker genes of macrophage phenotypic transformation can also be used to establish a model to predict the occurrence of atherosclerosis.
Collapse
Affiliation(s)
- Zemin Tian
- Department of Vascular and Thyroid Surgery, The First Affiliated Hospital of China Medical University, Shenyang 110001, Liaoning, China
| | - Shize Yang
- Department of Thoracic Surgery, The First Affiliated Hospital of China Medical University, Shenyang 110001, Liaoning, China
| |
Collapse
|
47
|
Xiang D, Jiang L, Yuan Q, Yu Y, Liu R, Chen M, Kuai Z, Zhang W, Yang F, Wu T, He Z, Ke Z, Hong W, He P, Tan N, Sun Y, Shi Z, Wei X, Luo J, Tan X, Huo Y, Qin G, Zhang C. Leukocyte-Specific Morrbid Promotes Leukocyte Differentiation and Atherogenesis. RESEARCH (WASHINGTON, D.C.) 2023; 6:0187. [PMID: 37426471 PMCID: PMC10325668 DOI: 10.34133/research.0187] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 01/23/2023] [Accepted: 06/14/2023] [Indexed: 07/11/2023]
Abstract
Monocyte-to-M0/M1 macrophage differentiation with unclear molecular mechanisms is a pivotal cellular event in many cardiovascular diseases including atherosclerosis. Long non-coding RNAs (lncRNAs) are a group of protein expression regulators; however, the roles of monocyte-lncRNAs in macrophage differentiation and its related vascular diseases are still unclear. The study aims to investigate whether the novel leukocyte-specific lncRNA Morrbid could regulate macrophage differentiation and atherogenesis. We identified that Morrbid was increased in monocytes and arterial walls from atherosclerotic mouse and from patients with atherosclerosis. In cultured monocytes, Morrbid expression was markedly increased during monocyte to M0 macrophage differentiation with an additional increase during M0 macrophage-to-M1 macrophage differentiation. The differentiation stimuli-induced monocyte-macrophage differentiation and the macrophage activity were inhibited by Morrbid knockdown. Moreover, overexpression of Morrbid alone was sufficient to elicit the monocyte-macrophage differentiation. The role of Morrbid in monocyte-macrophage differentiation was also identified in vivo in atherosclerotic mice and was verified in Morrbid knockout mice. We identified that PI3-kinase/Akt was involved in the up-regulation of Morrbid expression, whereas s100a10 was involved in Morrbid-mediated effect on macrophage differentiation. To provide a proof of concept of Morrbid in pathogenesis of monocyte/macrophage-related vascular disease, we applied an acute atherosclerosis model in mice. The results revealed that overexpression of Morrbid enhanced but monocyte/macrophage-specific Morrbid knockout inhibited the monocytes/macrophages recruitment and atherosclerotic lesion formation in mice. The results suggest that Morrbid is a novel biomarker and a modulator of monocyte-macrophage phenotypes, which is involved in atherogenesis.
Collapse
Affiliation(s)
- Di Xiang
- Department of Cardiology, Key Laboratory of Medical Electrophysiology, Ministry of Education, Institute of Cardiovascular Research, The Affiliated Hospital of Southwest Medical University, Southwest Medical University, Luzhou, Sichuan 646000, China
- Department of Biomedical Engineering, School of Medicine,
The University of Alabama at Birmingham, Birmingham, AL 35233, USA
| | - Lei Jiang
- Department of Cardiology, Guangdong Cardiovascular Institute, Guangdong Provincial Key Laboratory of Coronary Heart Disease Prevention, Guangdong Provincial Institute of Geriatric Medicine, Guangdong General Hospital,
Guangdong Academy of Medical Sciences, Guangzhou, Guangdong 510100, China
| | - Qiong Yuan
- Department of Biomedical Engineering, School of Medicine,
The University of Alabama at Birmingham, Birmingham, AL 35233, USA
| | - Yang Yu
- Department of Cardiology, Key Laboratory of Medical Electrophysiology, Ministry of Education, Institute of Cardiovascular Research, The Affiliated Hospital of Southwest Medical University, Southwest Medical University, Luzhou, Sichuan 646000, China
| | - Ruiming Liu
- Department of Biomedical Engineering, School of Medicine,
The University of Alabama at Birmingham, Birmingham, AL 35233, USA
| | - Meiting Chen
- Department of Biomedical Engineering, School of Medicine,
The University of Alabama at Birmingham, Birmingham, AL 35233, USA
| | - Zheng Kuai
- Department of Biomedical Engineering, School of Medicine,
The University of Alabama at Birmingham, Birmingham, AL 35233, USA
| | - Wendy Zhang
- Department of Biomedical Engineering, School of Medicine,
The University of Alabama at Birmingham, Birmingham, AL 35233, USA
| | - Fan Yang
- Department of Cardiology, Guangdong Cardiovascular Institute, Guangdong Provincial Key Laboratory of Coronary Heart Disease Prevention, Guangdong Provincial Institute of Geriatric Medicine, Guangdong General Hospital,
Guangdong Academy of Medical Sciences, Guangzhou, Guangdong 510100, China
| | - Tingting Wu
- Department of Biomedical Engineering, School of Medicine,
The University of Alabama at Birmingham, Birmingham, AL 35233, USA
| | - Zhiyu He
- Department of Biomedical Engineering, School of Medicine,
The University of Alabama at Birmingham, Birmingham, AL 35233, USA
| | - Zuhui Ke
- Department of Cardiology, Guangdong Cardiovascular Institute, Guangdong Provincial Key Laboratory of Coronary Heart Disease Prevention, Guangdong Provincial Institute of Geriatric Medicine, Guangdong General Hospital,
Guangdong Academy of Medical Sciences, Guangzhou, Guangdong 510100, China
| | - Wanzi Hong
- Department of Cardiology, Guangdong Cardiovascular Institute, Guangdong Provincial Key Laboratory of Coronary Heart Disease Prevention, Guangdong Provincial Institute of Geriatric Medicine, Guangdong General Hospital,
Guangdong Academy of Medical Sciences, Guangzhou, Guangdong 510100, China
| | - Pengcheng He
- Department of Cardiology, Guangdong Cardiovascular Institute, Guangdong Provincial Key Laboratory of Coronary Heart Disease Prevention, Guangdong Provincial Institute of Geriatric Medicine, Guangdong General Hospital,
Guangdong Academy of Medical Sciences, Guangzhou, Guangdong 510100, China
| | - Ning Tan
- Department of Cardiology, Guangdong Cardiovascular Institute, Guangdong Provincial Key Laboratory of Coronary Heart Disease Prevention, Guangdong Provincial Institute of Geriatric Medicine, Guangdong General Hospital,
Guangdong Academy of Medical Sciences, Guangzhou, Guangdong 510100, China
| | - Yeying Sun
- Department of Cardiology, Key Laboratory of Medical Electrophysiology, Ministry of Education, Institute of Cardiovascular Research, The Affiliated Hospital of Southwest Medical University, Southwest Medical University, Luzhou, Sichuan 646000, China
| | - Zhen Shi
- Department of Biomedical Engineering, School of Medicine,
The University of Alabama at Birmingham, Birmingham, AL 35233, USA
| | - Xuebiao Wei
- Department of Cardiology, Guangdong Cardiovascular Institute, Guangdong Provincial Key Laboratory of Coronary Heart Disease Prevention, Guangdong Provincial Institute of Geriatric Medicine, Guangdong General Hospital,
Guangdong Academy of Medical Sciences, Guangzhou, Guangdong 510100, China
| | - Jianfang Luo
- Department of Cardiology, Guangdong Cardiovascular Institute, Guangdong Provincial Key Laboratory of Coronary Heart Disease Prevention, Guangdong Provincial Institute of Geriatric Medicine, Guangdong General Hospital,
Guangdong Academy of Medical Sciences, Guangzhou, Guangdong 510100, China
| | - Xiaoqiu Tan
- Department of Cardiology, Key Laboratory of Medical Electrophysiology, Ministry of Education, Institute of Cardiovascular Research, The Affiliated Hospital of Southwest Medical University, Southwest Medical University, Luzhou, Sichuan 646000, China
| | - Yuqing Huo
- Vascular Biology Center, Medical College of Georgia,
Augusta University, Augusta, GA 30912, USA
| | - Gangjian Qin
- Department of Biomedical Engineering, School of Medicine,
The University of Alabama at Birmingham, Birmingham, AL 35233, USA
| | - Chunxiang Zhang
- Department of Cardiology, Key Laboratory of Medical Electrophysiology, Ministry of Education, Institute of Cardiovascular Research, The Affiliated Hospital of Southwest Medical University, Southwest Medical University, Luzhou, Sichuan 646000, China
| |
Collapse
|
48
|
Su ZDZ, Li CQ, Wang HW, Zheng MM, Chen QW. Inhibition of DRP1-dependent mitochondrial fission by Mdivi-1 alleviates atherosclerosis through the modulation of M1 polarization. J Transl Med 2023; 21:427. [PMID: 37386574 PMCID: PMC10311781 DOI: 10.1186/s12967-023-04270-9] [Citation(s) in RCA: 36] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2023] [Accepted: 06/13/2023] [Indexed: 07/01/2023] Open
Abstract
BACKGROUND Inflammation and immune dysfunction with classically activated macrophages(M1) infiltration are important mechanisms in the progression of atherosclerosis (AS). Dynamin-related protein 1 (DRP1)-dependent mitochondrial fission is a novel target for alleviating inflammatory diseases. This study aimed to investigate the effects of DRP1 inhibitor Mdivi-1 on AS. METHODS ApoE-/- mice were fed with a high-fat diet supplemented with or without Mdivi-1. RAW264.7 cells were stimulated by ox-LDL, pretreated with or without MCC950, Mito-TEMPO, or Mdivi-1. The burden of plaques and foam cell formation were determined using ORO staining. The blood lipid profles and inflammatory cytokines in serum were detected by commercial kits and ELISA, respectively. The mRNA expression of macrophage polarization markers, activation of NLRP3 and the phosphorylation state of DRP1 were detected. Mitochondrial reactive oxygen species (mito-ROS), mitochondrial staining, ATP level and mitochondrial membrane potential were detected by mito-SOX, MitoTracker, ATP determination kit and JC-1 staining, respectively. RESULTS In vivo, Mdivi-1 reduced the plaque areas, M1 polarization, NLRP3 activation and DRP1 phosphorylation at Ser616. In vitro, oxidized low-density lipoprotein (ox-LDL) triggered M1 polarization, NLRP3 activation and abnormal accumulation of mito-ROS. MCC950 and Mito-TEMPO suppressed M1 polarization mediated foam cell formation. Mito-TEMPO significantly inhibited NLRP3 activation. In addition, Mdivi-1 reduced foam cells by inhibiting M1 polarization. The possible mechanisms responsible for the anti-atherosclerotic effects of Mdivi-1 on reducing M1 polarization were associated with suppressing mito-ROS/NLRP3 pathway by inhibiting DRP1 mediated mitochondrial fission. In vitro, similar results were observed by DRP1 knockdown. CONCLUSION Inhibition of DRP1-dependent mitochondrial fission by Mdivi-1 alleviated atherogenesis via suppressing mito-ROS/NLRP3-mediated M1 polarization, indicating DRP1-dependent mitochondrial fission as a potential therapeutic target for AS.
Collapse
Affiliation(s)
- Ze-da-Zhong Su
- Department of General Practice, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Chun-Qiu Li
- Department of General Practice, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Hua-Wei Wang
- Department of General Practice, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Min-Ming Zheng
- Department of Ophthalmology, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China.
| | - Qing-Wei Chen
- Department of General Practice, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China.
| |
Collapse
|
49
|
Gou T, Hu M, Xu M, Chen Y, Chen R, Zhou T, Liu J, Guo L, Ao H, Ye Q. Novel wine in an old bottle: Preventive and therapeutic potentials of andrographolide in atherosclerotic cardiovascular diseases. J Pharm Anal 2023; 13:563-589. [PMID: 37440909 PMCID: PMC10334359 DOI: 10.1016/j.jpha.2023.05.010] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2022] [Revised: 05/13/2023] [Accepted: 05/16/2023] [Indexed: 07/15/2023] Open
Abstract
Atherosclerotic cardiovascular disease (ASCVD) frequently results in sudden death and poses a serious threat to public health worldwide. The drugs approved for the prevention and treatment of ASCVD are usually used in combination but are inefficient owing to their side effects and single therapeutic targets. Therefore, the use of natural products in developing drugs for the prevention and treatment of ASCVD has received great scholarly attention. Andrographolide (AG) is a diterpenoid lactone compound extracted from Andrographis paniculata. In addition to its use in conditions such as sore throat, AG can be used to prevent and treat ASCVD. It is different from drugs that are commonly used in the prevention and treatment of ASCVD and can not only treat obesity, diabetes, hyperlipidaemia and ASCVD but also inhibit the pathological process of atherosclerosis (AS) including lipid accumulation, inflammation, oxidative stress and cellular abnormalities by regulating various targets and pathways. However, the pharmacological mechanisms of AG underlying the prevention and treatment of ASCVD have not been corroborated, which may hinder its clinical development and application. Therefore, this review summarizes the physiological and pathological mechanisms underlying the development of ASCVD and the in vivo and in vitro pharmacological effects of AG on the relative risk factors of AS and ASCVD. The findings support the use of the old pharmacological compound ('old bottle') as a novel drug ('novel wine') for the prevention and treatment of ASCVD. Additionally, this review summarizes studies on the availability as well as pharmaceutical and pharmacokinetic properties of AG, aiming to provide more information regarding the clinical application and further research and development of AG.
Collapse
Affiliation(s)
- Tingting Gou
- State Key Laboratory of Southwestern Chinese Medicine Resources, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, China
- School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, China
| | - Minghao Hu
- State Key Laboratory of Southwestern Chinese Medicine Resources, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, China
- School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, China
| | - Min Xu
- State Key Laboratory of Southwestern Chinese Medicine Resources, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, China
- School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, China
| | - Yuchen Chen
- State Key Laboratory of Southwestern Chinese Medicine Resources, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, China
- School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, China
| | - Rong Chen
- State Key Laboratory of Southwestern Chinese Medicine Resources, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, China
- School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, China
| | - Tao Zhou
- State Key Laboratory of Southwestern Chinese Medicine Resources, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, China
- School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, China
| | - Junjing Liu
- State Key Laboratory of Southwestern Chinese Medicine Resources, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, China
- School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, China
| | - Li Guo
- State Key Laboratory of Southwestern Chinese Medicine Resources, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, China
- School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, China
| | - Hui Ao
- State Key Laboratory of Southwestern Chinese Medicine Resources, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, China
- Innovative Institute of Chinese Medicine and Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, China
| | - Qiang Ye
- State Key Laboratory of Southwestern Chinese Medicine Resources, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, China
- School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, China
| |
Collapse
|
50
|
Yao Y, Zhang P. Novel ultrasound techniques in the identification of vulnerable plaques-an updated review of the literature. Front Cardiovasc Med 2023; 10:1069745. [PMID: 37293284 PMCID: PMC10244552 DOI: 10.3389/fcvm.2023.1069745] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2022] [Accepted: 05/08/2023] [Indexed: 06/10/2023] Open
Abstract
Atherosclerosis is an inflammatory disease partly mediated by lipoproteins. The rupture of vulnerable atherosclerotic plaques and thrombosis are major contributors to the development of acute cardiovascular events. Despite various advances in the treatment of atherosclerosis, there has been no satisfaction in the prevention and assessment of atherosclerotic vascular disease. The identification and classification of vulnerable plaques at an early stage as well as research of new treatments remain a challenge and the ultimate goal in the management of atherosclerosis and cardiovascular disease. The specific morphological features of vulnerable plaques, including intraplaque hemorrhage, large lipid necrotic cores, thin fibrous caps, inflammation, and neovascularisation, make it possible to identify and characterize plaques with a variety of invasive and non-invasive imaging techniques. Notably, the development of novel ultrasound techniques has introduced the traditional assessment of plaque echogenicity and luminal stenosis to a deeper assessment of plaque composition and the molecular field. This review will discuss the advantages and limitations of five currently available ultrasound imaging modalities for assessing plaque vulnerability, based on the biological characteristics of the vulnerable plaque, and their value in terms of clinical diagnosis, prognosis, and treatment efficacy assessment.
Collapse
|