1
|
Naeini MK, Cecelja M, Freidin MB, Smith IG, Hysi P, Nielsen CS, Williams FMK. Chronic widespread musculoskeletal pain shares a highly heritable latent pathway with atherosclerosis and arterial stiffness. Pain 2025; 166:1425-1435. [PMID: 39620366 PMCID: PMC12067610 DOI: 10.1097/j.pain.0000000000003486] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2024] [Revised: 09/27/2024] [Accepted: 10/10/2024] [Indexed: 05/14/2025]
Abstract
ABSTRACT Chronic widespread pain (CWP) is prevalent and associated with reduced life expectancy. Cardiovascular disease is one possible mechanism for this. The purpose of this study was to examine the association of CWP with arterial stiffness and carotid plaque measured using ultrasound to determine if shared environmental or genetic factors might account for any observed association. Around 3000 participants from the TwinsUK with CWP information and measures of carotid-femoral pulse wave velocity (cfPWV), carotid intima-media thickness (cIMT), and plaque were considered. The relationship between CWP and cfPWV, cIMT, and plaque was determined. UK Biobank data were used to replicate the association. Cholesky decomposition and multivariate pathway twin models were examined. Using a 2-sample Mendelian randomisation approach, the causal association between CWP and coronary artery disease was assessed. TwinsUK participants demonstrated a significant association between CWP and increased cfPWV consistent with arterial stiffening (OR = 1.35, P -value = 0.012), as well as the presence of carotid plaque (OR = 1.45, P -value = 0.8e-5). The twin modelling showed a common latent component and pathway underlying CWP, cfPWV, and carotid plaque, with genetic factors accounting for 68% and 90% of the latent factor variation, respectively. The 2-sample MR revealed a potential causal association between CWP and coronary artery disease. This study found that those with CWP have increased the risk of arterial stiffness and atherosclerosis and suggests that CWP leads to an increased risk of cardiovascular disease through genetic factors.
Collapse
Affiliation(s)
- Maryam Kazemi Naeini
- Department of Twin Research and Genetic Epidemiology, School of Life Course and Population Sciences, King's College London, London, United Kingdom
| | - Marina Cecelja
- Department of Twin Research and Genetic Epidemiology, School of Life Course and Population Sciences, King's College London, London, United Kingdom
| | - Maxim B. Freidin
- Department of Twin Research and Genetic Epidemiology, School of Life Course and Population Sciences, King's College London, London, United Kingdom
- Department of Biology, School of Biological and Behavioural Sciences, Queen Mary University of London, London, United Kingdom
| | - Isabelle Granville Smith
- Department of Twin Research and Genetic Epidemiology, School of Life Course and Population Sciences, King's College London, London, United Kingdom
| | - Pirro Hysi
- Department of Twin Research and Genetic Epidemiology, School of Life Course and Population Sciences, King's College London, London, United Kingdom
| | - Christopher Sivert Nielsen
- Division of Mental and Physical Health, Norwegian Institute of Public Health, Oslo, Norway
- Division of Emergencies and Critical Care, Department of Pain Management and Research, Oslo University Hospital, Oslo, Norway
| | - Frances M. K. Williams
- Department of Twin Research and Genetic Epidemiology, School of Life Course and Population Sciences, King's College London, London, United Kingdom
| |
Collapse
|
2
|
Langhoff AF, Modin D, Dons M, Skaarup KG, Sengeløv M, Borchsenius J, Gislason G, Wisborg FD, Hove J, Lindhardt TB, Davidsen U, Jørgensen T, Kristensen AB, Olesen JB, Hansen ML, Henningsen K, Yafasov K, Pareek M, Johansen ND, Biering-Sørensen T. Inflammation and risk of cardiovascular outcomes in atrial fibrillation and flutter patients: A Nationwide Registry Study. Heart Rhythm 2025:S1547-5271(25)02511-1. [PMID: 40449818 DOI: 10.1016/j.hrthm.2025.05.051] [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: 05/12/2025] [Revised: 05/19/2025] [Accepted: 05/23/2025] [Indexed: 06/03/2025]
Abstract
BACKGROUND Atrial fibrillation (AF) is associated with development of heart failure (HF). Inflammation is increasingly recognized as a driver for cardiovascular disease, but the impact of inflammation on the risk of cardiovascular outcomes in AF requires further investigation. OBJECTIVE To establish the prognostic value of C-reactive protein (CRP) in relation to CV outcomes in patients with AF treated with first-time direct current (DC) cardioversion. METHODS A nationwide study of 8,691 first-time DC cardioverted patients with AF from 2011 to 2018 was conducted using the Danish National Health Registries for baseline comorbidities and prescribed medications. New-onset cardiovascular diagnoses were the primary outcomes, registered either at the outpatient clinic or upon admission. Incidence rates were reported, and multivariable adjusted Cox proportional-hazard models presented the hazard ratios for outcomes. RESULTS During a median follow-up time of 719 days (IQR: 328-1168 days), 568 (8.2%) developed HF. Higher CRP was associated with an increased risk of incident HF in univariable analysis (HR 1.07 per 1 mg/L increase in CRP (95%CI: 1.05-1.08), (p<0.001) and multivariable (HR 1.06 (95%CI: 1.04-1.08), (p<0.001) analysis. Likewise, patients with CRP > 3mg/L had an increased risk of incident HF (HR 1.78 (95%CI: 1.51-2.10), (p<0.001) compared with patients with CRP ≤ 3mg/L. CONCLUSION In patients with AF, increased CRP was associated with an increased long-term risk of HF, IHD, and CV death, even after multivariable adjustments. These results suggest that CRP may be a valuable risk marker in patients with AF.
Collapse
Affiliation(s)
- Adam Femerling Langhoff
- Department of Cardiology, Copenhagen University Hospital - Herlev and Gentofte, Copenhagen, Denmark; Center for Translational Cardiology and Pragmatic Randomized Trials, Department of Biomedical Sciences, Faculty of Health and Medical Sciences, Copenhagen, Denmark.
| | - Daniel Modin
- Department of Cardiology, Copenhagen University Hospital - Herlev and Gentofte, Copenhagen, Denmark
| | - Maria Dons
- Department of Cardiology, Copenhagen University Hospital - Herlev and Gentofte, Copenhagen, Denmark
| | - Kristoffer Grundtvig Skaarup
- Department of Cardiology, Copenhagen University Hospital - Herlev and Gentofte, Copenhagen, Denmark; Center for Translational Cardiology and Pragmatic Randomized Trials, Department of Biomedical Sciences, Faculty of Health and Medical Sciences, Copenhagen, Denmark
| | - Morten Sengeløv
- Department of Cardiology, Copenhagen University Hospital - Herlev and Gentofte, Copenhagen, Denmark; Center for Translational Cardiology and Pragmatic Randomized Trials, Department of Biomedical Sciences, Faculty of Health and Medical Sciences, Copenhagen, Denmark
| | - Julie Borchsenius
- Department of Cardiology, Copenhagen University Hospital - Herlev and Gentofte, Copenhagen, Denmark
| | - Gunnar Gislason
- Department of Cardiology, Copenhagen University Hospital - Herlev and Gentofte, Copenhagen, Denmark; Department of Clinical Medicine, Faculty of Health and Medical Sciences, Copenhagen, Denmark
| | | | - Jens Hove
- Department of Cardiology, Copenhagen University Hospital - Hvidovre, Copenhagen, Denmark
| | - Tommi Bo Lindhardt
- Department of Cardiology, Copenhagen University Hospital - North Zealand, Hillerod-Frederikssund, Denmark
| | - Ulla Davidsen
- Department of Cardiology, Copenhagen University Hospital - Bispebjerg, Copenhagen, Denmark
| | - Tami Jørgensen
- Department of Cardiology, Copenhagen University Hospital - Bispebjerg, Copenhagen, Denmark
| | - Alex Berg Kristensen
- Department of Cardiology, Copenhagen University Hospital - Bispebjerg, Copenhagen, Denmark
| | - Jonas Bjerring Olesen
- Department of Cardiology, Copenhagen University Hospital - Herlev and Gentofte, Copenhagen, Denmark
| | - Morten Lock Hansen
- Department of Cardiology, Copenhagen University Hospital - Herlev and Gentofte, Copenhagen, Denmark
| | - Kristoffer Henningsen
- Department of Cardiology, Copenhagen University Hospital - Herlev and Gentofte, Copenhagen, Denmark
| | - Karim Yafasov
- Department of Cardiology, Copenhagen University Hospital - North Zealand, Hillerod-Frederikssund, Denmark
| | - Manan Pareek
- Department of Cardiology, Copenhagen University Hospital - Herlev and Gentofte, Copenhagen, Denmark; Center for Translational Cardiology and Pragmatic Randomized Trials, Department of Biomedical Sciences, Faculty of Health and Medical Sciences, Copenhagen, Denmark; Department of Cardiology, Copenhagen University Hospital - Rigshospitalet, Copenhagen, Denmark
| | - Niklas Dyrby Johansen
- Department of Cardiology, Copenhagen University Hospital - Herlev and Gentofte, Copenhagen, Denmark; Center for Translational Cardiology and Pragmatic Randomized Trials, Department of Biomedical Sciences, Faculty of Health and Medical Sciences, Copenhagen, Denmark
| | - Tor Biering-Sørensen
- Department of Cardiology, Copenhagen University Hospital - Herlev and Gentofte, Copenhagen, Denmark; Center for Translational Cardiology and Pragmatic Randomized Trials, Department of Biomedical Sciences, Faculty of Health and Medical Sciences, Copenhagen, Denmark; Department of Cardiology, Copenhagen University Hospital - Rigshospitalet, Copenhagen, Denmark; Steno Diabetes Center Copenhagen, Copenhagen, Denmark
| |
Collapse
|
3
|
Famurewa AC, Akhigbe RE, George MY, Adekunle YA, Oyedokun PA, Akhigbe TM, Fatokun AA. Mechanisms of ferroptotic and non-ferroptotic organ toxicity of chemotherapy: protective and therapeutic effects of ginger, 6-gingerol and zingerone in preclinical studies. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2025; 398:4747-4778. [PMID: 39636404 PMCID: PMC11985630 DOI: 10.1007/s00210-024-03623-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/04/2024] [Accepted: 11/08/2024] [Indexed: 12/07/2024]
Abstract
Chemotherapy (CT) is one of the flagship options for the treatment of cancers worldwide. It involves the use of cytotoxic anticancer agents to kill or inhibit the proliferation of cancer cells. However, despite its clinical efficacy, CT triggers side effect toxicities in several organs, which may impact cancer patient's quality of life and treatment outcomes. While the side effect toxicity is consistent with non-ferroptotic mechanisms involving oxidative stress, inflammation, mitochondrial impairment and other aberrant signalling leading to apoptosis and necroptosis, recent studies show that ferroptosis, a non-apoptotic, iron-dependent cell death pathway, is also involved in the pathophysiology of CT organ toxicity. CT provokes organ ferroptosis via system Xc-/GPX-4/GSH/SLC7A11 axis depletion, ferritinophagy, iron overload, lipid peroxidation and upregulation of ferritin-related proteins. Cisplatin (CP) and doxorubicin (DOX) are common CT drugs indicated to induce ferroptosis in vitro and in vivo. Studies have explored natural preventive and therapeutic strategies using ginger rhizome and its major bioactive compounds, 6-gingerol (6G) and zingerone (ZG), to combat mechanisms of CT side effect toxicity. Ginger extract, 6G and ZG mitigate non-ferroptotic oxidative inflammation, apoptosis and mitochondrial dysfunction mechanisms of CT side effect toxicity, but their effects on CT-induced ferroptosis remain unclear. Systematic investigations are, therefore, needed to unfold the roles of ginger, 6G and ZG on ferroptosis involved in CT side effect toxicity, as they are potential natural agents for the prevention of CT toxicity. This review reveals the ferroptotic and non-ferroptotic toxicity mechanisms of CT and the protective mechanisms of ginger, 6G and ZG against CT-induced, ferroptotic and non-ferroptotic organ toxicities.
Collapse
Affiliation(s)
- Ademola C Famurewa
- Department of Medical Biochemistry, Faculty of Basic Medical Sciences, College of Medical Sciences, Alex Ekwueme Federal University Ndufu-Alike, Ikwo, Nigeria.
- Centre for Natural Products Discovery, School of Pharmacy and Biomolecular Sciences, Faculty of Science, Liverpool John Moores University, Byrom Street, Liverpool, L3 3AF, UK.
| | - Roland E Akhigbe
- Department of Physiology, Ladoke Akintola University of Technology, Ogbomoso, Oyo State, Nigeria
- Reproductive Biology and Toxicology Research Laboratory, Oasis of Grace Hospital, Osogbo, Nigeria
| | - Mina Y George
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Ain Shams University, Cairo, 11566, Egypt
| | - Yemi A Adekunle
- Department of Pharmaceutical and Medicinal Chemistry, College of Pharmacy, Afe Babalola University, Ado-Ekiti, Nigeria
| | - Precious A Oyedokun
- Department of Physiology, Ladoke Akintola University of Technology, Ogbomoso, Oyo State, Nigeria
- Reproductive Biology and Toxicology Research Laboratory, Oasis of Grace Hospital, Osogbo, Nigeria
| | - Tunmise M Akhigbe
- Reproductive Biology and Toxicology Research Laboratory, Oasis of Grace Hospital, Osogbo, Nigeria
- Breeding and Genetics Unit, Department of Agronomy, Osun State University, Osogbo, Osun State, Nigeria
| | - Amos A Fatokun
- Centre for Natural Products Discovery, School of Pharmacy and Biomolecular Sciences, Faculty of Science, Liverpool John Moores University, Byrom Street, Liverpool, L3 3AF, UK
| |
Collapse
|
4
|
Potashnikova D, Fasler-Kan E. Special Issue "Functional Role of Cytokines in Cancer and Chronic Inflammation". Int J Mol Sci 2025; 26:4048. [PMID: 40362288 PMCID: PMC12072130 DOI: 10.3390/ijms26094048] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2025] [Accepted: 04/22/2025] [Indexed: 05/15/2025] Open
Abstract
Cytokines are a diverse group of signaling proteins that are secreted by a wide range of cell types, including, but not limited to, immune cells [...].
Collapse
Affiliation(s)
- Daria Potashnikova
- Department of Cell Biology and Histology, School of Biology, Lomonosov Moscow State University, 119234 Moscow, Russia
- I.V. Davydovsky City Clinical Hospital, Moscow Department of Healthcare, 109240 Moscow, Russia
| | - Elizaveta Fasler-Kan
- Department of Pediatric Surgery, Children’s Hospital, Inselspital Bern, CH-3010 Bern, Switzerland
- Department of Biomedical Research, University of Bern, CH-3010 Bern, Switzerland
| |
Collapse
|
5
|
Lachowicz JI, Gać P. Short-Term and Long-Term Effects of Inhaled Ultrafine Particles on Blood Markers of Cardiovascular Diseases: A Systematic Review and Meta-Analysis. J Clin Med 2025; 14:2846. [PMID: 40283676 PMCID: PMC12028172 DOI: 10.3390/jcm14082846] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2025] [Revised: 04/03/2025] [Accepted: 04/14/2025] [Indexed: 04/29/2025] Open
Abstract
Background/Objectives: Air pollution is the highest environmental risk factor of mortality and morbidity worldwide, leading to over 4 million deaths each year. Among different air pollutants, ultrafine particles (UFPs) constitute the highest risk factor of cardiovascular diseases (CVDs). Epidemiological studies have associated UFPs with the short- and long-term imbalance of numerous blood markers. Our objective was to systematically review the short-term and long-term impact of UFP exposure on blood markers of CVDs. Methods: We prepared the systematic review of CVD blood markers and meta-analyses of the short- and long-term effects of UFP exposure on high-sensitivity C-reactive protein (hsCRP) concentration. The eligibility criteria were established with the use of the Provider, Enrollment, Chain, and Ownership System (PECOS) model, and the literature search was conducted in Web of Science, PubMed, and Scopus databases from 1 January 2013 to 9 January 2025. The risk of bias (RoB) was prepared according to a World Health Organization (WHO) template. Results: The results showed an increase in hsCRP as a result of both short-term and long-term UFPs. Moreover, IL-6 and IL-1β together with other inflammatory markers increased after short-term UFP exposure. In addition, different nucleic acids, among which were miR-24-3p and let-7d-5p, were differentially expressed (DE) as a result of short-term UFP exposure. Chronic exposure to UFPs could lead to a persistent increase in hsCRP and other blood markers of CVDs. Conclusions: Our findings underline that UFPs may lead to the development and/or worsening of cardiovascular outcomes in fragile populations living in air-polluted areas.
Collapse
Affiliation(s)
- Joanna Izabela Lachowicz
- Department of Environmental Health, Occupational Medicine and Epidemiology, Wroclaw Medical University, Mikulicza-Radeckiego 7, 50-368 Wroclaw, Poland;
| | | |
Collapse
|
6
|
Altyar AE, Bhardwaj S, Ghaboura N, Kaushik P, Alenezi SK, Mantargi MJS, Afzal M. Role of IL-2, IL-6, and TNF-α as Potential Biomarkers in Ischemic Heart Disease: A Comparative Study of Patients with CAD and Non-CAD. Med Sci (Basel) 2025; 13:40. [PMID: 40265387 PMCID: PMC12015866 DOI: 10.3390/medsci13020040] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2025] [Revised: 03/20/2025] [Accepted: 03/28/2025] [Indexed: 04/24/2025] Open
Abstract
Background: Ischemic heart disease (CAD), a leading global health burden, arises primarily from atherosclerosis, an inflammatory condition characterized by lipid accumulation and metabolic dysregulation. The precise contribution of inflammatory cytokines (IL-2, IL-6, and TNF-α) to CAD pathogenesis remains an area of significant research. Aim: The primary aim of this study is to examine the IL-2, IL-6, and TNF-α in patients with coronary artery disease (CAD) and compare them with Non-CAD individuals to evaluate their potential as diagnostic biomarkers for CAD. Methodology: A prospective observational study was conducted over 3 years, involving 100 participants divided into CAD and non-CAD groups. Blood samples were isolated and analyzed for IL-2, IL-6, and TNF-α levels utilizing ELISA kits. Biochemical parameters, including lipid profiles, were also assessed. Results: This study observed significantly elevated IL-6 in patients with CAD compared with controls, while IL-2 and TNF-α levels did not reach statistical significance. The CAD group exhibited dyslipidemia characterized by elevated triglycerides and reduced HDL. Furthermore, the CAD group demonstrated alterations in biochemical parameters, including lower albumin and calcium levels, higher urea and uric acid levels, and an elevated erythrocyte sedimentation rate. These findings suggest a systemic inflammatory state and metabolic disturbances in patients with CAD. Conclusions: This study highlights IL-6 as a potential biomarker and key player in CAD pathogenesis. These findings warrant further investigation into the therapeutic potential of targeting inflammatory pathways for cardiovascular risk reduction.
Collapse
Affiliation(s)
- Ahmed E. Altyar
- Department of Pharmacy Practice, Faculty of Pharmacy, King Abdulaziz University, P.O. Box 80260, Jeddah 21589, Saudi Arabia;
- Pharmacy Program, Department of Pharmacy Practice, Batterjee Medical College, P.O. Box 6231, Jeddah 21442, Saudi Arabia
| | - Shilpa Bhardwaj
- Department of Neurochemistry, Institute of Human Behaviour and Allied Sciences Hospital, Delhi 110095, India
| | - Nehmat Ghaboura
- Pharmacy Program, Department of Pharmacy Practice, Batterjee Medical College, P.O. Box 6231, Jeddah 21442, Saudi Arabia
| | - Priya Kaushik
- Pacific Institute of Medical Sciences, Udaipur 313015, India
| | - Sattam Khulaif Alenezi
- Department of Pharmacology and Toxicology, College of Pharmacy, Qassim University, Buraydah 51452, Saudi Arabia
| | - Mohammed Jaffar Sadiq Mantargi
- Pharmacy Program, Department of Pharmaceutical Sciences, Batterjee Medical College, P.O. Box 6231, Jeddah 21442, Saudi Arabia
| | - Muhammad Afzal
- Pharmacy Program, Department of Pharmaceutical Sciences, Batterjee Medical College, P.O. Box 6231, Jeddah 21442, Saudi Arabia
| |
Collapse
|
7
|
Ong LT, Sia CH. Pathophysiological Links Between Myocardial Infarction and Anxiety Disorder, Major Depressive Disorder, Bipolar Disorder and Schizophrenia. BIOLOGY 2025; 14:336. [PMID: 40282201 PMCID: PMC12025231 DOI: 10.3390/biology14040336] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/06/2025] [Revised: 03/09/2025] [Accepted: 03/21/2025] [Indexed: 04/29/2025]
Abstract
There is increasing evidence demonstrating that psychiatric conditions elevate the risk of developing accelerated atherosclerosis and early-onset cardiovascular disease (CVD), including myocardial infarction (MI). Several mechanisms contribute to this observation. The dysfunction of the autonomic nervous system and hyperactivity of the hypothalamic-pituitary-adrenal axis in these patients contribute to the development of MI. Additionally, patients with underlying psychiatric disorders often have abnormal levels of anti-inflammatory and pro-inflammatory cytokines, which can lead to early vascular damage and subsequent atherosclerosis. Elevated PAI-1 levels, reduced tPA activity, and the decreased brain-derived neurotrophic factor (BDNF), influenced by coagulation and inflammation, may contribute to depression and its link to MI. Oxidative stress, marked by increased reactive species and impaired antioxidant defenses, is associated with cellular damage and has been consistently implicated in schizophrenia and bipolar disorder, potentially contributing to myocardial infarction. Finally, molecular genetic studies have indicated that psychiatric disorders and myocardial infarction may share potential pleiotropic genes. The interplay between the psychiatric conditions and myocardial infarction underscores the importance of integrated care approaches to manage both mental and physical health.
Collapse
Affiliation(s)
| | - Ching-Hui Sia
- Department of Cardiology, National University Heart Centre, Singapore 119228, Singapore;
| |
Collapse
|
8
|
Christa M, Dennis F, Philip R, Jakob L, Timo S. Admission glucose, HbA1c levels and inflammatory cytokines in patients with acute ST-elevation myocardial infarction. Clin Proteomics 2025; 22:8. [PMID: 39962379 PMCID: PMC11834255 DOI: 10.1186/s12014-025-09530-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2024] [Accepted: 02/04/2025] [Indexed: 02/20/2025] Open
Abstract
BACKGROUND To investigate the association between admission glucose and HbA1c values and inflammatory plasma proteins in hospitalized patients with acute ST-elevation myocardial infarction (STEMI). METHODS This analysis was based on 345 STEMI patients recorded by the population-based Myocardial Infarction Registry Augsburg between 2009 and 2013. Using the OLINK inflammatory panel, a total of 92 protein biomarkers were measured in arterial blood samples, which were obtained within the scope of cardiac catheterization immediately after admission. The associations between admission glucose and HbA1c levels and the 92 protein markers were investigated using multivariable linear regression models. RESULTS Admission glucose showed significantly positive associations with the inflammatory markers IL-10, IL-8, IL-6, FGF-21, IL-7, ST1A1, MCP-1, 4E-BP1, SIRT2, STAMBP and IL-18R1 after Bonferroni adjustment. HbA1c values were only significantly associated with IL-18R1. In stratified analyses, admission glucose was not significantly associated with any plasma protein in the diabetes subgroup, while there were several protein markers that showed significantly positive associations with admission glucose in STEMI patients without known diabetes, namely IL-10, CCL20, IL-8, MCP-1 and IL-6. CONCLUSIONS Admission glucose in patients hospitalized due to an acute STEMI seems to be related to an inflammatory and immune-related response, expressed by an increase in inflammation-related plasma proteins in particular in non-diabetic patients with stress hyperglycemia. The present results may open new avenues for the development of biomarkers suitable as potential diagnostic or prognostic clinical markers.
Collapse
Affiliation(s)
- Meisinger Christa
- Epidemiology, Medical Faculty, University of Augsburg, University Hospital Augsburg, Stenglinstraße 2, 86156, Augsburg, Germany.
| | - Freuer Dennis
- Epidemiology, Medical Faculty, University of Augsburg, University Hospital Augsburg, Stenglinstraße 2, 86156, Augsburg, Germany
| | - Raake Philip
- Department of Cardiology, Respiratory Medicine and Intensive Care, University Hospital Augsburg, Stenglinstraße 2, 86156, Augsburg, Germany
| | - Linseisen Jakob
- Epidemiology, Medical Faculty, University of Augsburg, University Hospital Augsburg, Stenglinstraße 2, 86156, Augsburg, Germany
| | - Schmitz Timo
- Epidemiology, Medical Faculty, University of Augsburg, University Hospital Augsburg, Stenglinstraße 2, 86156, Augsburg, Germany
| |
Collapse
|
9
|
Fraile-Martinez O, García-Montero C, Gomez-Lahoz AM, Sainz F, Bujan J, Barrena-Blázquez S, López-González L, Díaz-Pedrero R, Álvarez-Mon M, García-Honduvilla N, Saez MA, Monserrat J, Ortega MA. Evidence of Inflammatory Network Disruption in Chronic Venous Disease: An Analysis of Circulating Cytokines and Chemokines. Biomedicines 2025; 13:150. [PMID: 39857734 PMCID: PMC11763091 DOI: 10.3390/biomedicines13010150] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2024] [Revised: 01/05/2025] [Accepted: 01/07/2025] [Indexed: 01/27/2025] Open
Abstract
Background: Chronic venous disease (CVD) comprises a set of vascular disorders that affect the venous system with important local and systemic repercussions. A growing body of evidence displays the relationship between suffering from CVD and a marked deregulation of the immune inflammatory system. In this sense, the previous literature has reported some significant changes in the level of various circulating inflammatory parameters in these patients. However, more research is required to detail and deepen this complex relationship. Methods: In this work, we studied, using a multiplex technique, the levels of circulating cytokines and chemokines detectable in the serum of 40 patients with CVD and compared it with 38 healthy controls (HCs). In parallel, we performed Spearman's correlation analysis to explore potential inflammatory networks in CVD. Results: In this study, we measured circulating cytokines and chemokines in CVD patients using a multiplex assay. Results showed increased levels of several pro-inflammatory mediators (IL-1β, IL-2, IL-5, IL-6, IL-7, IL-8, IL-12, IL-17A, IL-23, TNF-α, IFN-γ, fractalkine, ITAC, and GM-CSF) and a decrease in IL-13, with no significant changes in IL-4, IL-10, IL-21, MIP-1α, MIP-1β, or MIP-3α. The Spearman correlation analysis revealed strong, positive correlations among several inflammatory mediators in HC, particularly between TNF-alpha, IL-1β, IL-17A, and IL-23, forming a highly interconnected cytokine network. In contrast, CVD patients showed fewer, weaker, and distinct correlations, with new associations such as IFN-γ with IL-1β and IL-23, suggesting a disrupted inflammatory profile. Conclusions: The distinct inflammatory profile in CVD patients, characterized by altered cytokine and chemokine levels and a less coordinated cytokine network, underscores the reconfiguration of inflammatory pathways in this condition. These findings highlight potential therapeutic targets aimed at restoring immune balance and mitigating chronic inflammation in CVD.
Collapse
Affiliation(s)
- Oscar Fraile-Martinez
- Department of Medicine and Medical Specialities (CIBEREHD), Faculty of Medicine and Health Sciences, University of Alcalá, 28801 Alcala de Henares, Spain; (O.F.-M.); (C.G.-M.); (A.M.G.-L.); (F.S.); (J.B.); (M.Á.-M.); (N.G.-H.); (M.A.S.); (J.M.)
- Ramón y Cajal Institute of Sanitary Research (IRYCIS), 28034 Madrid, Spain; (S.B.-B.); (L.L.-G.); (R.D.-P.)
| | - Cielo García-Montero
- Department of Medicine and Medical Specialities (CIBEREHD), Faculty of Medicine and Health Sciences, University of Alcalá, 28801 Alcala de Henares, Spain; (O.F.-M.); (C.G.-M.); (A.M.G.-L.); (F.S.); (J.B.); (M.Á.-M.); (N.G.-H.); (M.A.S.); (J.M.)
- Ramón y Cajal Institute of Sanitary Research (IRYCIS), 28034 Madrid, Spain; (S.B.-B.); (L.L.-G.); (R.D.-P.)
| | - Ana María Gomez-Lahoz
- Department of Medicine and Medical Specialities (CIBEREHD), Faculty of Medicine and Health Sciences, University of Alcalá, 28801 Alcala de Henares, Spain; (O.F.-M.); (C.G.-M.); (A.M.G.-L.); (F.S.); (J.B.); (M.Á.-M.); (N.G.-H.); (M.A.S.); (J.M.)
- Ramón y Cajal Institute of Sanitary Research (IRYCIS), 28034 Madrid, Spain; (S.B.-B.); (L.L.-G.); (R.D.-P.)
| | - Felipe Sainz
- Department of Medicine and Medical Specialities (CIBEREHD), Faculty of Medicine and Health Sciences, University of Alcalá, 28801 Alcala de Henares, Spain; (O.F.-M.); (C.G.-M.); (A.M.G.-L.); (F.S.); (J.B.); (M.Á.-M.); (N.G.-H.); (M.A.S.); (J.M.)
- Ramón y Cajal Institute of Sanitary Research (IRYCIS), 28034 Madrid, Spain; (S.B.-B.); (L.L.-G.); (R.D.-P.)
- Angiology and Vascular Surgery Service, Central University Hospital of Defence—UAH, 28047 Madrid, Spain
| | - Julia Bujan
- Department of Medicine and Medical Specialities (CIBEREHD), Faculty of Medicine and Health Sciences, University of Alcalá, 28801 Alcala de Henares, Spain; (O.F.-M.); (C.G.-M.); (A.M.G.-L.); (F.S.); (J.B.); (M.Á.-M.); (N.G.-H.); (M.A.S.); (J.M.)
- Ramón y Cajal Institute of Sanitary Research (IRYCIS), 28034 Madrid, Spain; (S.B.-B.); (L.L.-G.); (R.D.-P.)
| | - Silvestra Barrena-Blázquez
- Ramón y Cajal Institute of Sanitary Research (IRYCIS), 28034 Madrid, Spain; (S.B.-B.); (L.L.-G.); (R.D.-P.)
- Department of Nursing and Physiotherapy, Faculty of Medicine and Health Sciences, University of Alcalá, 28801 Alcala de Henares, Spain
| | - Laura López-González
- Ramón y Cajal Institute of Sanitary Research (IRYCIS), 28034 Madrid, Spain; (S.B.-B.); (L.L.-G.); (R.D.-P.)
- Department of Surgery, Medical and Social Sciences, Faculty of Medicine and Health Sciences, University of Alcalá, 28801 Alcala de Henares, Spain
| | - Raul Díaz-Pedrero
- Ramón y Cajal Institute of Sanitary Research (IRYCIS), 28034 Madrid, Spain; (S.B.-B.); (L.L.-G.); (R.D.-P.)
- Department of Surgery, Medical and Social Sciences, Faculty of Medicine and Health Sciences, University of Alcalá, 28801 Alcala de Henares, Spain
| | - Melchor Álvarez-Mon
- Department of Medicine and Medical Specialities (CIBEREHD), Faculty of Medicine and Health Sciences, University of Alcalá, 28801 Alcala de Henares, Spain; (O.F.-M.); (C.G.-M.); (A.M.G.-L.); (F.S.); (J.B.); (M.Á.-M.); (N.G.-H.); (M.A.S.); (J.M.)
- Ramón y Cajal Institute of Sanitary Research (IRYCIS), 28034 Madrid, Spain; (S.B.-B.); (L.L.-G.); (R.D.-P.)
- Immune System Diseases-Rheumatology and Internal Medicine Service, University Hospital Prince of Asturias, Networking Research Center on for Liver and Digestive Diseases (CIBEREHD), 28806 Alcala de Henares, Spain
| | - Natalio García-Honduvilla
- Department of Medicine and Medical Specialities (CIBEREHD), Faculty of Medicine and Health Sciences, University of Alcalá, 28801 Alcala de Henares, Spain; (O.F.-M.); (C.G.-M.); (A.M.G.-L.); (F.S.); (J.B.); (M.Á.-M.); (N.G.-H.); (M.A.S.); (J.M.)
- Ramón y Cajal Institute of Sanitary Research (IRYCIS), 28034 Madrid, Spain; (S.B.-B.); (L.L.-G.); (R.D.-P.)
| | - Miguel A. Saez
- Department of Medicine and Medical Specialities (CIBEREHD), Faculty of Medicine and Health Sciences, University of Alcalá, 28801 Alcala de Henares, Spain; (O.F.-M.); (C.G.-M.); (A.M.G.-L.); (F.S.); (J.B.); (M.Á.-M.); (N.G.-H.); (M.A.S.); (J.M.)
- Ramón y Cajal Institute of Sanitary Research (IRYCIS), 28034 Madrid, Spain; (S.B.-B.); (L.L.-G.); (R.D.-P.)
- Pathological Anatomy Service, University Hospital Gómez-Ulla, 28806 Alcala de Henares, Spain
| | - Jorge Monserrat
- Department of Medicine and Medical Specialities (CIBEREHD), Faculty of Medicine and Health Sciences, University of Alcalá, 28801 Alcala de Henares, Spain; (O.F.-M.); (C.G.-M.); (A.M.G.-L.); (F.S.); (J.B.); (M.Á.-M.); (N.G.-H.); (M.A.S.); (J.M.)
- Ramón y Cajal Institute of Sanitary Research (IRYCIS), 28034 Madrid, Spain; (S.B.-B.); (L.L.-G.); (R.D.-P.)
| | - Miguel A. Ortega
- Department of Medicine and Medical Specialities (CIBEREHD), Faculty of Medicine and Health Sciences, University of Alcalá, 28801 Alcala de Henares, Spain; (O.F.-M.); (C.G.-M.); (A.M.G.-L.); (F.S.); (J.B.); (M.Á.-M.); (N.G.-H.); (M.A.S.); (J.M.)
- Ramón y Cajal Institute of Sanitary Research (IRYCIS), 28034 Madrid, Spain; (S.B.-B.); (L.L.-G.); (R.D.-P.)
| |
Collapse
|
10
|
Zhou W, Li X, Zhou H, Hu Y, Chen Y, Guo D. TNF-α/IL-1β/IL-1α/IL-12 inflammatory cytokine axes coupled with TLR1/TLR3/TLR5/MYD88 immune signaling pathway over-activation contribute to simultaneous carotid and coronary artery and occlusion in elderly patients. Cytokine 2025; 185:156808. [PMID: 39556941 DOI: 10.1016/j.cyto.2024.156808] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2024] [Revised: 10/22/2024] [Accepted: 11/10/2024] [Indexed: 11/20/2024]
Abstract
BACKGROUND It remains difficult to evaluate the risk factors for concomitant carotid artery as well as coronary artery diseases in elderly patients. The aim of this research was to determine the TNF-α/IL-1β/IL-1α/IL-12 axes-TLR1/TLR3/TLR5/MYD88 immune signaling pathway interactions in coexistent carotid artery occlusion and coronary artery occlusion in elderly patients. METHODS Elderly patients, who underwent carotid ultrasonography and coronary computed tomography angiography, were consecutively included in this research. The analyzed groups consisted of those with coexistent carotid artery occlusion and coronary artery occlusion as well as healthy individuals were enrolled as control group. The circulating levels of tumor necrosis factor-α (TNF-α), interleukin-1β (IL-1β), interleukin-1α (IL-1α), interleukin-12 (IL-12), toll-like receptor 1 (TLR1), toll-like receptor 3 (TLR3), toll-like receptor 5 (TLR5) and myeloid differentiation factor 88 (MYD88) were measured. RESULTS The biomarkers (TNF-α, IL-1β, IL-1α, IL-12, TLR1, TLR3, TLR5 and MYD88) were significantly increased in carotid artery occlusion + left circumflex coronary artery occlusion group when compared with control group and carotid artery occlusion + right coronary artery occlusion group, respectively (P < 0.001), and were further elevated in carotid artery occlusion + left anterior descending coronary artery occlusion group when compared to carotid artery occlusion + right coronary artery occlusion group and carotid artery occlusion + left circumflex coronary artery occlusion group, respectively (P < 0.001). CONCLUSION This research demonstrated that the TNF-α/IL-1β/IL-1α/IL-12 axes and TLR1/TLR3/TLR5/MYD88 immune signaling pathway implicated in the pathogenesis of carotid artery occlusion with coronary artery occlusion in elderly patients.
Collapse
Affiliation(s)
- Wenhang Zhou
- Xiamen Road Branch Hospital, The Affiliated Huaian Hospital of Xuzhou Medical University, Huaian 223005, China
| | - Xia Li
- Xiamen Road Branch Hospital, The Affiliated Huaian Hospital of Xuzhou Medical University, Huaian 223005, China
| | - Hualan Zhou
- Department of Geriatrics, The Affiliated Huaian Hospital of Xuzhou Medical University, Huaian 223002, China
| | - Youdong Hu
- Department of Geriatrics, The Affiliated Huaian Hospital of Xuzhou Medical University, Huaian 223002, China
| | - Ying Chen
- Department of Geriatrics, The Affiliated Huaian Hospital of Xuzhou Medical University, Huaian 223002, China
| | - Dianxuan Guo
- Xiamen Road Branch Hospital, The Affiliated Huaian Hospital of Xuzhou Medical University, Huaian 223005, China.
| |
Collapse
|
11
|
Zhang XF, Min RX, Wang Z, Qi Y, Li RN, Fan JM. Effects of Ginseng Consumption on Cardiovascular Health Biomarkers in Adults: A Systematic Review and Meta-Analysis of Randomized Controlled Trials. Phytother Res 2024; 38:5873-5900. [PMID: 39387709 DOI: 10.1002/ptr.8339] [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: 01/18/2024] [Revised: 07/21/2024] [Accepted: 09/01/2024] [Indexed: 10/15/2024]
Abstract
Ginseng, with various pharmacological activities, has received increasing attention to improve cardiovascular health (CVH). Therefore, this meta-analysis synthesized the effect of ginseng consumption on biomarkers of CVH in adults. A systematic search was performed in the databases of PubMed, Scopus, Web of Science, Embase, and the Cochrane Library through July 24, 2023 to screen out English-language randomized controlled trials (RCTs) evaluating the effects of ginseng consumption on body composition, blood pressure, vascular stiffness, lipid metabolism, glucose metabolism, insulin resistance, inflammatory cytokines, and adipocytokines in adults. The weighted mean difference (WMD) and 95% confidence interval (CI) were used to evaluate the overall effect size, and STATA 12.0 was used for comprehensive analysis. Forty-five studies were included in the meta-analysis. Ginseng consumption significantly reduced systolic blood pressure (SBP) (WMD = -2.57 mmHg, 95% CI = -4.99 to -0.14, p = 0.038), total cholesterol (TC) (WMD = -4.40 mg/dL, 95% CI = -8.67 to -0.132, p = 0.043), low density lipoprotein cholesterol (LDL-C) (WMD = -2.81 mg/dL, 95% CI = -4.89 to -0.72, p = 0.008), C-reactive protein (CRP) (WMD = -0.41 mg/L, 95% CI = -0.73 to -0.10, p = 0.010), and interleukin-6 (IL-6) (WMD = -2.82 pg./mL, 95% CI = -4.31 to -1.32, p < 0.001). Subgroup analyses suggested that supplementation with ginseng for less than 12 weeks significantly reduced SBP, but 12 weeks or more improved TC and CRP. Ginseng consumption on SBP, TC, and CRP seemed to be more effective on unhealthy participants. The meta-analysis showed that ginseng consumption might have the potential to improve SBP, TC, LDL-C, CRP, and IL-6. These findings suggest that ginseng is a potential candidate for the maintenance of CVH. However, our results had high heterogeneity. Future high-quality studies are needed to firmly establish the clinical efficacy of ginseng consumption.
Collapse
Affiliation(s)
- Xiao-Feng Zhang
- Department of Nutrition and Food Hygiene, College of Public Health, Zhengzhou University, Zhengzhou, Henan, China
| | - Rui-Xue Min
- Department of Nutrition and Food Hygiene, College of Public Health, Zhengzhou University, Zhengzhou, Henan, China
| | - Zhen Wang
- Department of Nutrition and Food Hygiene, College of Public Health, Zhengzhou University, Zhengzhou, Henan, China
| | - Yue Qi
- Department of Nutrition and Food Hygiene, College of Public Health, Zhengzhou University, Zhengzhou, Henan, China
| | - Ruo-Nan Li
- Department of Nutrition and Food Hygiene, College of Public Health, Zhengzhou University, Zhengzhou, Henan, China
| | - Jian-Ming Fan
- Department of Nutrition and Food Hygiene, College of Public Health, Zhengzhou University, Zhengzhou, Henan, China
| |
Collapse
|
12
|
Vitali-Silva A, Bello VA, Poli RC, de Oliveira CEC, Lopes MV, Silveira DN, Bossa BB, Espinosa BR, Ahrens TM, Reiche EMV, Simão ANC. IL18 rs360717 and rs187238 genetic variants are associated with migraine diagnosis. Eur J Pain 2024; 28:1685-1700. [PMID: 38922725 DOI: 10.1002/ejp.2302] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2024] [Revised: 05/06/2024] [Accepted: 06/03/2024] [Indexed: 06/28/2024]
Abstract
BACKGROUND Migraine is a genetically determined disorder that predisposes to recurrent episodes of headache. Interleukin (IL)-18 is a pro-inflammatory cytokine that seems to play a role in migraine pathophysiology, and its genetic variants could potentially impact susceptibility to migraine. OBJECTIVE To investigate the association between IL18 rs360717 and rs187238 genetic variants with migraine diagnosis and its clinical characteristics. METHODS A case-control study was conducted with 152 people with migraine and 155 healthy controls, matched by sex, age, ethnicity, and body mass index. Clinical characteristics of migraine, as well as validated questionnaires regarding disability and impact of migraine, presence of allodynia, anxiety, depression, and hyperacusis were collected. Genotyping for IL18 rs360717 and rs187238 variants was performed using real-time polymerase chain reaction (qPCR) and TaqMan™ method. RESULTS The IL18 rs360717A and rs187238G alleles were associated with increased chance of being diagnosed with migraine (OR = 1.53, 95%CI 1.05-2.24, p = 0.028 and OR = 1.46, 95%CI 1.00-2.14, p = 0.049, respectively). In the dominant model, the rs360717GA + AA genotypes were also associated with a higher chance of migraine than the GG genotype (OR = 1.69, 95%CI 1.05-2.73, p = 0.030). In women, in addition to the previous associations, there was also an effect of the variants on the chance of migraine in the codominant models and dominant models. Furthermore, among women, there was an influence on the prevalence of postdrome perception with rs360717GA + AA (OR = 3.04, 95%CI 1.10-8.42, p = 0.032) and rs187238CG + GG (OR = 2.97, 95%CI 1.08-8.21, p = 0.035). CONCLUSION IL18 rs360717 and rs187238 variants were associated with migraine diagnosis and postdrome symptoms, especially in women. SIGNIFICANCE This study has demonstrated that IL18 rs360717 and rs187238 variants play a role in migraine, influencing the chance of being diagnosed with migraine, particularly among women. There are prospects that IL18 variants could be considered potential genetic biomarkers for migraine.
Collapse
Affiliation(s)
- Aline Vitali-Silva
- Escola de Medicina, Pontifícia Universidade Católica Do Paraná, Londrina, Brazil
- Universidade Estadual de Londrina, Londrina, Brazil
| | | | - Regina Célia Poli
- Escola de Medicina, Pontifícia Universidade Católica Do Paraná, Londrina, Brazil
- Universidade Norte Do Paraná, Londrina, Brazil
| | - Carlos Eduardo Coral de Oliveira
- Escola de Medicina, Pontifícia Universidade Católica Do Paraná, Londrina, Brazil
- Universidade Estadual de Londrina, Londrina, Brazil
| | - Milene Valéria Lopes
- Escola de Medicina, Pontifícia Universidade Católica Do Paraná, Londrina, Brazil
| | | | | | | | | | - Edna Maria Vissoci Reiche
- Escola de Medicina, Pontifícia Universidade Católica Do Paraná, Londrina, Brazil
- Universidade Estadual de Londrina, Londrina, Brazil
| | | |
Collapse
|
13
|
Rantasalo V, Laukka D, Nikulainen V, Jalkanen J, Gunn J, Kiviniemi T, Hakovirta H. Association between aortic calcification and cytokine levels in patients with peripheral artery disease. Clin Transl Sci 2024; 17:e70036. [PMID: 39344403 PMCID: PMC11440032 DOI: 10.1111/cts.70036] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2024] [Revised: 08/10/2024] [Accepted: 09/03/2024] [Indexed: 10/01/2024] Open
Abstract
Aortic calcification-a marker of advanced atherosclerosis in large arteries-associates with cardiovascular mortality and morbidity. Little is known about the soluble inflamJarmatory profiles involved in large artery atherosclerosis. We investigated the correlation between aortic calcification in the abdominal aorta and cytokine levels in a cohort of peripheral artery disease patients. Aortic calcification index was measured from computed tomography exams and circulating cytokine levels were analyzed from blood serum samples of 156 consecutive patients prior to invasive treatment of peripheral artery disease. The study included 156 patients (mean age 70.7 years, 64 (41.0%) women). The mean ankle-brachial index (ABI) was 0.64 and the mean aortic calcification index (ACI) was 52.3. ACI was associated with cytokines cutaneous T-cell-attracting chemokine CTACK (β 23.08, SE 5.22, p < 0.001) and monokine induced by gamma-interferon MIG (β 9.40, SE 2.82, p 0.001) in univariate linear regression. After adjustment with cardiovascular risk factors, CTACK and MIG were independently associated with ACI, β 17.9 (SE 5.22, p < 0.001) for CTACK and β 6.80 (SE 3.33, p 0.043) for MIG. CTACK was significantly higher in the patients representing the highest ACI tertile (highest vs. middle, 7.53 vs. 7.34 Tukeys HSD p-value 0.023 and highest vs. lowest tertile 7.53 vs. 7.29, Tukeys HSD p-value 0.002). MIG was significantly higher in the highest tertile versus lowest (7.65 vs. 7.30, Tukeys HSD p-value 0.027). Cytokines CTACK and MIG are associated with higher ACI, suggesting that CTACK and MIG reflect atherosclerotic disease burden of the aorta. This might further suggest the possible association with other cardiovascular morbidities.
Collapse
Affiliation(s)
- Ville Rantasalo
- Department of Surgery, University of Turku, Turku, Finland
- Division of Gastroenterology and Urology, Department of Vascular Surgery, Turku University Hospital, Turku, Finland
| | - Dan Laukka
- Department of Clinical Neurosciences, University of Turku, Turku, Finland
- Department of Neurosurgery, Neurocenter, Turku University Hospital, Turku, Finland
| | - Veikko Nikulainen
- Division of Gastroenterology and Urology, Department of Vascular Surgery, Turku University Hospital, Turku, Finland
| | - Juho Jalkanen
- Division of Gastroenterology and Urology, Department of Vascular Surgery, Turku University Hospital, Turku, Finland
| | - Jarmo Gunn
- Department of Surgery, University of Turku, Turku, Finland
| | | | - Harri Hakovirta
- Department of Surgery, University of Turku, Turku, Finland
- Satasairaala, Pori, Finland
- King Faisal Specialist Hospital and Research Center, Medina, Saudi Arabia
| |
Collapse
|
14
|
Chen Y, Zhong A. Causal effects of inflammatory cytokines on cardiovascular diseases: Insights from genetic evidence. Heliyon 2024; 10:e35447. [PMID: 39165962 PMCID: PMC11334864 DOI: 10.1016/j.heliyon.2024.e35447] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2024] [Revised: 07/26/2024] [Accepted: 07/29/2024] [Indexed: 08/22/2024] Open
Abstract
Background The causal relationship between inflammatory cytokines and cardiovascular diseases (CVDs) has not been fully elucidated. Exploring this relationship between circulating inflammatory cytokines and CVDs is crucial for early clinical diagnosis and effective treatment. Methods and Results This study investigated the causal relationships between 41 inflammatory cytokines and six CVDs: heart failure (HF), myocardial infarction (MI), unstable angina pectoris (UAP), stable angina pectoris (SAP), valvular heart disease (VHD), and aortic aneurysm (AA), using the Mendelian Randomization (MR) method. The primary analysis employed the inverse-variance weighted (IVW) method within MR. Heterogeneity and pleiotropy were assessed through MR-Egger regression and the Q statistic. Strong evidence supported the effect of macrophage inflammatory protein-1β (MIP-1β) on MI (OR = 1.062, P < 0.001, FDR <0.001). Suggestive evidence showed that the Beta nerve growth factor increased the risk of MI (OR = 1.145, P = 0.025), but the stem cell factor (SCF) demonstrated a potential protective effect against MI (OR = 0.910, P = 0.04). SCF and hepatocyte growth factor (HGF) exhibited potential protective effects against SAP. No inflammatory cytokine was associated with UAP. Monocyte chemotactic protein-1 was linked to an increased risk of VHD (OR = 1.056, P = 0.049). Higher levels of interleukin-13 (IL-13), interferon gamma-induced protein 10 (IP-10), and growth-regulated oncogene-alpha were associated with increased susceptibility to HF. Elevated basic fibroblast growth factor (bFGF) levels exhibited protective effects against AA (OR = 0.751, P = 0.038). Reverse MR analyses revealed that AA significantly decreased circulating TNF-related apoptosis-inducing ligand (TRAIL) levels (OR = 0.907, P < 0.001, FDR = 0.01). MI significantly increased circulating IL-12-p70 levels (OR = 1.146, P < 0.001, FDR = 0.014). Suggestive evidence indicated the Causal effects of six CVDs on 17 circulating inflammatory cytokines. Conclusions This study clarified the causal relationships between specific inflammatory cytokines and six CVDs, providing novel insights and evidence into the genetic involvement of inflammatory cytokines in CVDs. These inflammatory cytokines may be potential biomarkers for early disease diagnosis and treatment evaluation.
Collapse
Affiliation(s)
- Yuxiu Chen
- Department of Emergency Medicine, Guizhou Provincial People's Hospital, Guiyang, 550002, Guizhou, China
| | - Aifang Zhong
- Department of Emergency Medicine, The Second Xiangya Hospital, Central South University, Changsha, 410011, Hunan, China
| |
Collapse
|
15
|
Aguayo-Morales H, Poblano J, Berlanga L, Castillo-Tobías I, Silva-Belmares SY, Cobos-Puc LE. Plant Antioxidants: Therapeutic Potential in Cardiovascular Diseases. COMPOUNDS 2024; 4:479-502. [DOI: 10.3390/compounds4030029] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/03/2025]
Abstract
Cardiovascular diseases (CVDs) are a global health problem. The mortality associated with them is one of the highest. Essentially, CVDs occur when the heart or blood vessels are damaged. Oxidative stress is an imbalance between the production of reactive oxygen species (free radicals) and antioxidant defenses. Increased production of reactive oxygen species can cause cardiac and vascular injuries, leading to CVDs. Antioxidant therapy has been shown to have beneficial effects on CVDs. Plants are a rich source of bioactive antioxidants on our planet. Several classes of these compounds have been identified. Among them, carotenoids and phenolic compounds are the most potent antioxidants. This review summarizes the role of some carotenoids (a/β-carotene, lycopene and lutein), polyphenols such as phenolic acids (caffeic, p-coumaric, ferulic and chlorogenic acids), flavonoids (quercetin, kaempferol and epigallocatechin gallate), and hydroxytyrosol in mitigating CVDs by studying their biological antioxidant mechanisms. Through detailed analysis, we aim to provide a deeper understanding of how these natural compounds can be integrated into cardiovascular health strategies to help reduce the overall burden of CVD.
Collapse
Affiliation(s)
- Hilda Aguayo-Morales
- Facultad de Ciencias Químicas, Unidad Saltillo, Universidad Autónoma de Coahuila, Boulevard Venustiano Carranza S/N Esquina Con Ing, José Cárdenas Valdés, República Oriente, Saltillo 25290, Mexico
| | - Joan Poblano
- Facultad de Ciencias Químicas, Unidad Saltillo, Universidad Autónoma de Coahuila, Boulevard Venustiano Carranza S/N Esquina Con Ing, José Cárdenas Valdés, República Oriente, Saltillo 25290, Mexico
| | - Lia Berlanga
- Facultad de Ciencias Químicas, Unidad Saltillo, Universidad Autónoma de Coahuila, Boulevard Venustiano Carranza S/N Esquina Con Ing, José Cárdenas Valdés, República Oriente, Saltillo 25290, Mexico
| | - Ileana Castillo-Tobías
- Facultad de Ciencias Químicas, Unidad Saltillo, Universidad Autónoma de Coahuila, Boulevard Venustiano Carranza S/N Esquina Con Ing, José Cárdenas Valdés, República Oriente, Saltillo 25290, Mexico
| | - Sonia Yesenia Silva-Belmares
- Facultad de Ciencias Químicas, Unidad Saltillo, Universidad Autónoma de Coahuila, Boulevard Venustiano Carranza S/N Esquina Con Ing, José Cárdenas Valdés, República Oriente, Saltillo 25290, Mexico
| | - Luis E. Cobos-Puc
- Facultad de Ciencias Químicas, Unidad Saltillo, Universidad Autónoma de Coahuila, Boulevard Venustiano Carranza S/N Esquina Con Ing, José Cárdenas Valdés, República Oriente, Saltillo 25290, Mexico
| |
Collapse
|
16
|
Schulz S, Reuter L, Navarrete Santos A, Bitter K, Rehm S, Schlitt A, Reichert S. Major Adverse Cardiovascular Events: The Importance of Serum Levels and Haplotypes of the Anti-Inflammatory Cytokine Interleukin 10. Biomolecules 2024; 14:979. [PMID: 39199367 PMCID: PMC11353162 DOI: 10.3390/biom14080979] [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: 06/10/2024] [Revised: 07/30/2024] [Accepted: 08/07/2024] [Indexed: 09/01/2024] Open
Abstract
BACKGROUND Cardiovascular diseases (CVDs) represent major medical and socio-economic challenges worldwide. There is substantial evidence that CVD is closely linked to inflammatory changes triggered by a complex cytokine network. In this context, interleukin 10 (IL-10) plays an important role as a pleiotropic cytokine with an anti-inflammatory capacity. In this study (a substudy of ClinTrials.gov, identifier: NCT01045070), the prognostic relevance of IL-10 levels and IL-10 haplotypes (rs1800896/rs1800871/rs1800872) was assessed regarding adverse cardiovascular outcomes (combined endpoint: myocardial infarction, stroke/transient ischemic attack, cardiac death and death according to stroke) within a 10-year follow-up. PATIENTS AND METHODS At baseline, 1002 in-patients with CVD were enrolled. Serum levels of IL-10 were evaluated utilizing flow cytometry (BD™ Cytometric Bead Array). Haplotype analyses were carried out by polymerase chain reactions with sequence-specific primers (PCR-SSP). RESULTS In a survival analysis, IL-10 haplotypes were not proven to be cardiovascular prognostic factors in a 10-year follow-up (Breslow test: p = 0.423). However, a higher IL-10 level was associated with adverse cardiovascular outcomes (Breslow test: p = 0.047). A survival analysis considering adjusted hazard ratios (HRs) could not confirm this correlation (Cox regression: adjusted HR = 1.26, p = 0.168). CONCLUSION In the present study, an elevated IL-10 level but not IL-10 haplotypes was linked to adverse cardiovascular outcomes (10-year follow-up) in a cohort of CVD patients.
Collapse
Affiliation(s)
- Susanne Schulz
- Department of Operative Dentistry and Periodontology, Medical Faculty, Martin-Luther-University Halle-Wittenberg, 06108 Halle, Germany
| | - Leonie Reuter
- Department of Operative Dentistry and Periodontology, Medical Faculty, Martin-Luther-University Halle-Wittenberg, 06108 Halle, Germany
| | - Alexander Navarrete Santos
- Center for Medical Basic Research, Medical Faculty, Martin-Luther-University Halle-Wittenberg, 06120 Halle, Germany
| | - Kerstin Bitter
- Department of Operative Dentistry and Periodontology, Medical Faculty, Martin-Luther-University Halle-Wittenberg, 06108 Halle, Germany
| | - Selina Rehm
- Department of Operative Dentistry and Periodontology, Medical Faculty, Martin-Luther-University Halle-Wittenberg, 06108 Halle, Germany
| | - Axel Schlitt
- Department of Cardiology, Paracelsus-Harz-Clinic Bad Suderode, 06485 Quedlinburg, Germany;
- Department of Medicine III, Medical Faculty, Martin-Luther-University Halle-Wittenberg, 06120 Halle, Germany
| | - Stefan Reichert
- Department of Operative Dentistry and Periodontology, Medical Faculty, Martin-Luther-University Halle-Wittenberg, 06108 Halle, Germany
| |
Collapse
|
17
|
Cheng X, Zhang R, Qi X, Wang H, Gao T, Zheng L, Qiao M, Li Y, Gao S, Chen J, Chang R, Zheng G, Dong H. Metabolomics and network pharmacology exploration of the effects of bile acids on carotid atherosclerosis and potential underlying mechanisms. Front Endocrinol (Lausanne) 2024; 15:1430720. [PMID: 39076513 PMCID: PMC11284041 DOI: 10.3389/fendo.2024.1430720] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/10/2024] [Accepted: 06/07/2024] [Indexed: 07/31/2024] Open
Abstract
BACKGROUND Bile acids (BAs), products of gut microbiota metabolism, have long been implicated in atherosclerotic disease pathogenesis. Characterizing the serum bile acid profile and exploring its potential role in carotid atherosclerosis (CAS) development are crucial tasks. METHODS In this study, we recruited 73 patients with CAS as the disease group and 77 healthy individuals as the control group. We systematically measured the serum concentrations of 15 bile acids using ultrahigh-performance liquid chromatography-mass spectrometry (UPLC-MS/MS). Multivariate logistic regression and least absolute shrinkage and selection operator (LASSO) regression were applied to analyze the impact of bile acids on the disease and select the key BAs. The possible molecular mechanism was elucidated by network pharmacology. RESULTS (1) The BA profile of patients with CAS significantly differed. (2) Multifactorial logistic regression analysis identified elevated levels of GCDCA (OR: 1.01, P < 0.001), DCA (OR: 1.01, P = 0.005), and TDCA (OR: 1.05, P = 0.002) as independent risk factors for CAS development. Conversely, GCA (OR: 0.99, P = 0.020), LCA (OR: 0.83, P = 0.002), and GUDCA (OR: 0.99, P = 0.003) were associated with protective effects against the disease. GCA, DCA, LCA, and TDCA were identified as the four key BAs. (3) TNF, FXR, GPBAR1, ESR1 and ACE were predicted to be targets of BAs against AS. These four BAs potentially impact AS progression by triggering signaling pathways, including cAMP, PPAR, and PI3K-AKT pathways, via their targets. CONCLUSION This study offers valuable insights into potential therapeutic strategies for atherosclerosis that target bile acids.
Collapse
Affiliation(s)
- Xing Cheng
- Department of Vascular Surgery, The Second Hospital of Shanxi Medical University, Taiyuan, China
| | - Ruijing Zhang
- Department of Nephrology, The Second Hospital of Shanxi Medical University, Taiyuan, China
| | - Xiaotong Qi
- Department of Vascular Surgery, The Second Hospital of Shanxi Medical University, Taiyuan, China
| | - Heng Wang
- Department of Vascular Surgery, The Second Hospital of Shanxi Medical University, Taiyuan, China
| | - Tingting Gao
- Department of Vascular Surgery, The Second Hospital of Shanxi Medical University, Taiyuan, China
| | - Lin Zheng
- Department of Vascular Surgery, The Second Hospital of Shanxi Medical University, Taiyuan, China
| | - Maolin Qiao
- Department of Vascular Surgery, The Second Hospital of Shanxi Medical University, Taiyuan, China
| | - Yaling Li
- Department of Vascular Surgery, The Second Hospital of Shanxi Medical University, Taiyuan, China
| | - Siqi Gao
- Department of Vascular Surgery, The Second Hospital of Shanxi Medical University, Taiyuan, China
| | - Jinshan Chen
- Department of Vascular Surgery, The Second Hospital of Shanxi Medical University, Taiyuan, China
| | - Runze Chang
- Department of Vascular Surgery, The Second Hospital of Shanxi Medical University, Taiyuan, China
| | - Guoping Zheng
- Centre for Transplantation and Renal Research, University of Sydney at Westmead Millennium Institute, Westmead, NSW, Australia
| | - Honglin Dong
- Department of Vascular Surgery, The Second Hospital of Shanxi Medical University, Taiyuan, China
| |
Collapse
|
18
|
Katkenov N, Mukhatayev Z, Kozhakhmetov S, Sailybayeva A, Bekbossynova M, Kushugulova A. Systematic Review on the Role of IL-6 and IL-1β in Cardiovascular Diseases. J Cardiovasc Dev Dis 2024; 11:206. [PMID: 39057626 PMCID: PMC11277031 DOI: 10.3390/jcdd11070206] [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: 05/22/2024] [Revised: 06/19/2024] [Accepted: 06/25/2024] [Indexed: 07/28/2024] Open
Abstract
Cardiovascular diseases (CVDs) are a leading cause of global morbidity and mortality, significantly driven by chronic inflammation. Interleukin-6 (IL-6) and interleukin-1β (IL-1β) are critical inflammatory cytokines implicated in CVD progression. This systematic review evaluates the roles of IL-6 and IL-1β in CVDs by synthesizing data from relevant studies to understand their impact on cardiovascular outcomes and identify potential therapeutic interventions. A comprehensive literature search was conducted using PubMed and Embase, covering studies from January 2014 to December 2024. Inclusion criteria encompassed studies investigating IL-6 and/or IL-1β in CVDs, including human and relevant animal models, and reporting clinical outcomes, molecular mechanisms, or therapeutic interventions. Data extraction and quality assessment were performed independently by two reviewers. Our review included 12 studies focusing on the roles of IL-6 and IL-1β in various CVDs. Elevated IL-6 levels were significantly associated with peripheral artery disease, myocardial infarction, and heart failure, while IL-1β levels were linked to worse outcomes in coronary artery disease and heart failure. Meta-analyses indicated a significant association between higher IL-6 and IL-1β levels and increased risk of adverse cardiovascular events. These findings suggest that targeting IL-6 and IL-1β could offer promising therapeutic strategies for reducing inflammation and improving cardiovascular outcomes.
Collapse
Affiliation(s)
- Nurlubek Katkenov
- Laboratory of Microbiome, National Laboratory Astana, Nazarbayev University, Astana 010000, Kazakhstan; (N.K.); (Z.M.); (S.K.)
| | - Zhussipbek Mukhatayev
- Laboratory of Microbiome, National Laboratory Astana, Nazarbayev University, Astana 010000, Kazakhstan; (N.K.); (Z.M.); (S.K.)
| | - Samat Kozhakhmetov
- Laboratory of Microbiome, National Laboratory Astana, Nazarbayev University, Astana 010000, Kazakhstan; (N.K.); (Z.M.); (S.K.)
| | - Aliya Sailybayeva
- Heart Center, CF “University Medical Center”, Astana 010000, Kazakhstan; (A.S.); (M.B.)
| | | | - Almagul Kushugulova
- Laboratory of Microbiome, National Laboratory Astana, Nazarbayev University, Astana 010000, Kazakhstan; (N.K.); (Z.M.); (S.K.)
- Heart Center, CF “University Medical Center”, Astana 010000, Kazakhstan; (A.S.); (M.B.)
| |
Collapse
|
19
|
Yin J, Fu X, Luo Y, Leng Y, Ao L, Xie C. A Narrative Review of Diabetic Macroangiopathy: From Molecular Mechanism to Therapeutic Approaches. Diabetes Ther 2024; 15:585-609. [PMID: 38302838 PMCID: PMC10942953 DOI: 10.1007/s13300-024-01532-7] [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: 11/21/2023] [Accepted: 01/11/2024] [Indexed: 02/03/2024] Open
Abstract
Diabetic macroangiopathy, a prevalent and severe complication of diabetes mellitus, significantly contributes to the increased morbidity and mortality rates among affected individuals. This complex disorder involves multifaceted molecular mechanisms that lead to the dysfunction and damage of large blood vessels, including atherosclerosis (AS) and peripheral arterial disease. Understanding the intricate pathways underlying the development and progression of diabetic macroangiopathy is crucial for the development of effective therapeutic interventions. This review aims to shed light on the molecular mechanism implicated in the pathogenesis of diabetic macroangiopathy. We delve into the intricate interplay of chronic inflammation, oxidative stress, endothelial dysfunction, and dysregulated angiogenesis, all of which contribute to the vascular complications observed in this disorder. By exploring the molecular mechanism involved in the disease we provide insight into potential therapeutic targets and strategies. Moreover, we discuss the current therapeutic approaches used for treating diabetic macroangiopathy, including glycemic control, lipid-lowering agents, and vascular interventions.
Collapse
Affiliation(s)
- Jiacheng Yin
- Hospital of Chengdu University of Traditional Chinese Medicine No, 39 Shi-er-Qiao Road, Chengdu, 610072, Sichuan Province, People's Republic of China
| | - Xiaoxu Fu
- Hospital of Chengdu University of Traditional Chinese Medicine No, 39 Shi-er-Qiao Road, Chengdu, 610072, Sichuan Province, People's Republic of China
- TCM Regulating Metabolic Diseases Key Laboratory of Sichuan Province, No. 39 Shi-er-Qiao Road, Chengdu, 610072, Sichuan Province, People's Republic of China
- Department of Endocrinology, Hospital of Chengdu University of Traditional Chinese Medicine, No. 39 Shi-er-Qiao Road, Chengdu, 610072, Sichuan Province, People's Republic of China
| | - Yue Luo
- Hospital of Chengdu University of Traditional Chinese Medicine No, 39 Shi-er-Qiao Road, Chengdu, 610072, Sichuan Province, People's Republic of China
| | - Yuling Leng
- Hospital of Chengdu University of Traditional Chinese Medicine No, 39 Shi-er-Qiao Road, Chengdu, 610072, Sichuan Province, People's Republic of China
| | - Lianjun Ao
- Hospital of Chengdu University of Traditional Chinese Medicine No, 39 Shi-er-Qiao Road, Chengdu, 610072, Sichuan Province, People's Republic of China
| | - Chunguang Xie
- Hospital of Chengdu University of Traditional Chinese Medicine No, 39 Shi-er-Qiao Road, Chengdu, 610072, Sichuan Province, People's Republic of China.
- TCM Regulating Metabolic Diseases Key Laboratory of Sichuan Province, No. 39 Shi-er-Qiao Road, Chengdu, 610072, Sichuan Province, People's Republic of China.
- Department of Endocrinology, Hospital of Chengdu University of Traditional Chinese Medicine, No. 39 Shi-er-Qiao Road, Chengdu, 610072, Sichuan Province, People's Republic of China.
| |
Collapse
|
20
|
Ke D, Ni J, Yuan Y, Cao M, Chen S, Zhou H. Identification and Validation of Hub Genes Related to Neutrophil Extracellular Traps-Mediated Cell Damage During Myocardial Infarction. J Inflamm Res 2024; 17:617-637. [PMID: 38323113 PMCID: PMC10844013 DOI: 10.2147/jir.s444975] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2023] [Accepted: 01/23/2024] [Indexed: 02/08/2024] Open
Abstract
Purpose Studies have shown that neutrophil-mediated formation of neutrophil extracellular traps (NETs) leads to increased inflammatory response and cellular tissue damage during myocardial infarction (MI). We aimed to identify and validate possible hub genes in the process of NETs-mediated cell damage. Methods We performed an immune cell infiltration analysis of the MI transcriptome dataset based on CIBERSORT and ssGSEA algorithms. Gene expression profiles of NETs formation (GSE178883) were used to analyze the physiological processes of peripheral blood neutrophils after phorbol myristate acetate (PMA) stimulation. Bioinformatics and machine learning algorithms were utilized to find candidate hub genes based on NETs-related genes and transcriptome datasets (GSE66360 and GSE179828). We generated the receiver operating curve (ROC) to evaluate the diagnostic value of hub genes. Next, the correlation between hub genes and immune cells was analyzed using CIBERSORT, ssGSEA and xCell algorithms. Finally, we used quantitative real-time PCR (qRT-PCR) and immunohistochemistry to verify gene expression. Results Immune cell infiltration analysis revealed that inflammatory cells such as neutrophils were highly expressed in the peripheral blood of patients with MI. Functional analysis of differentially expressed genes (DEGs) in GSE178883 indicated that the potential pathogenesis lies in immune terms. Using weighted gene co-expression network analysis (WGCNA) and machine learning algorithms, we finally identified the seven hub genes (FCAR, IL1B, MMP9, NFIL3, CXCL2, ICAM1, and ZFP36). The qRT-PCR results showed that IL-1B, MMP9, and NFIL3 mRNA expression was up-regulated in the MI group compared to the control. Immunohistochemical results showed high MMP9, IL-1B, and NFIL3 expression in the infarcted area compared to the non-infarcted area and sham-operated groups. Conclusion We identified seven hub genes associated with NETs-mediated cellular damage during MI. Our results may provide insights into the mechanisms of neutrophil-mediated cell injury during MI.
Collapse
Affiliation(s)
- Da Ke
- Department of Cardiology, Renmin Hospital of Wuhan University, Hubei Key Laboratory of Metabolic and Chronic Diseases, Wuhan, 430060, People’s Republic of China
| | - Jian Ni
- Department of Cardiology, Renmin Hospital of Wuhan University, Hubei Key Laboratory of Metabolic and Chronic Diseases, Wuhan, 430060, People’s Republic of China
| | - Yuan Yuan
- Department of Cardiology, Renmin Hospital of Wuhan University, Hubei Key Laboratory of Metabolic and Chronic Diseases, Wuhan, 430060, People’s Republic of China
| | - Mingzhen Cao
- Department of Cardiology, Renmin Hospital of Wuhan University, Hubei Key Laboratory of Metabolic and Chronic Diseases, Wuhan, 430060, People’s Republic of China
| | - Si Chen
- Hubei Key Laboratory of Metabolic and Chronic Diseases, Wuhan, 430060, People’s Republic of China
| | - Heng Zhou
- Department of Cardiology, Renmin Hospital of Wuhan University, Hubei Key Laboratory of Metabolic and Chronic Diseases, Wuhan, 430060, People’s Republic of China
| |
Collapse
|
21
|
Cai YW, Wu MX, Gao QY, Wang JF, Huang YL, Hu YZ, Qiu RF, Mai WY, Zhang HF. Editorial: Cytokines, novel cell death models and pathways in cardiovascular diseases. Front Cardiovasc Med 2023; 10:1270320. [PMID: 37692034 PMCID: PMC10484615 DOI: 10.3389/fcvm.2023.1270320] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2023] [Accepted: 08/11/2023] [Indexed: 09/12/2023] Open
Affiliation(s)
- Yang-Wei Cai
- Department of Cardiology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
- Guangdong Province Key Laboratory of Arrhythmia and Electrophysiology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
| | - Mao-Xiong Wu
- Department of Cardiology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
- Guangdong Province Key Laboratory of Arrhythmia and Electrophysiology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
| | - Qing-Yuan Gao
- Department of Cardiology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
- Guangdong Province Key Laboratory of Arrhythmia and Electrophysiology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
| | - Jing-Feng Wang
- Department of Cardiology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
- Guangdong Province Key Laboratory of Arrhythmia and Electrophysiology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
| | - Yu-Li Huang
- Department of Cardiology, Shunde Hospital, Southern Medical University, Foshan, China
| | - Yun-Zhao Hu
- Department of Cardiology, Shunde Hospital, Southern Medical University, Foshan, China
| | - Ruo-Feng Qiu
- Capital Health System, Trenton, NJ, United States
| | - Wei-Yi Mai
- Department of Cardiology, First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Hai-Feng Zhang
- Department of Cardiology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
- Guangdong Province Key Laboratory of Arrhythmia and Electrophysiology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
| |
Collapse
|