1
|
Woolley C, Princip M, Hackl-Zuccarella C, Auschra B, Holzgang SA, Pazhenkottil A, Gomez Vieito D, von Känel R. Reduced interleukin-6 stress reactivity in male physicians with occupational burnout. J Neural Transm (Vienna) 2025:10.1007/s00702-025-02945-9. [PMID: 40418270 DOI: 10.1007/s00702-025-02945-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2024] [Accepted: 05/08/2025] [Indexed: 05/27/2025]
Abstract
Occupational burnout is associated with an elevated risk of cardiovascular events, but the mechanisms underlying this connection have rarely been studied. Low grade inflammation with interleukin (IL)-6 as a key player has been postulated as a possible link between low-grade inflammation and cardiovascular events. Another possibility could be attenuated inflammatory responses to stress reflecting a dysregulated innate immune response. The aim of this study was to examine the relationship between burnout and IL-6 reactivity to acute stress in physicians with burnout. Sixty male physicians were recruited, 30 with burnout, assessed with the Maslach Burnout Inventory, and 30 without burnout. Participants underwent the Trier Social Stress Test, inflicting uncontrollability, and social evaluative threat. Blood samples for IL-6 measurements were taken five times at predefined intervals during stress. Repeated measures analysis of covariance was conducted across the five time points, adjusting for age. IL-6 reactivity over time was significantly lower in physicians with burnout compared to their matched healthy counterparts (F(1.90, 108.10) = 4.88, p = 0.010) during stress. Confirming these results, there was also a significantly lower area under the curve with respect to increase in physicians with burnout compared to those without burnout (F(1, 57) = 4.99, p = 0.029). This study suggests that burnout is associated with lowered IL-6 reactivity in male physicians with burnout. An attenuated innate immune response in burnout complies with increased allostatic load such that dysfunctional stress responses might increase the risk of cardiovascular events in the long term.
Collapse
Affiliation(s)
- Christoph Woolley
- Department of Consultation-Liaison Psychiatry and Psychosomatic Medicine, University Hospital Zurich, University of Zurich, Haldenbachstrasse 16/18, 8091, Zurich, Switzerland.
| | - Mary Princip
- Department of Consultation-Liaison Psychiatry and Psychosomatic Medicine, University Hospital Zurich, University of Zurich, Haldenbachstrasse 16/18, 8091, Zurich, Switzerland
| | - Claudia Hackl-Zuccarella
- Department of Consultation-Liaison Psychiatry and Psychosomatic Medicine, University Hospital Zurich, University of Zurich, Haldenbachstrasse 16/18, 8091, Zurich, Switzerland
| | - Bianca Auschra
- Department of Consultation-Liaison Psychiatry and Psychosomatic Medicine, University Hospital Zurich, University of Zurich, Haldenbachstrasse 16/18, 8091, Zurich, Switzerland
| | - Sarah Andrea Holzgang
- Department of Consultation-Liaison Psychiatry and Psychosomatic Medicine, University Hospital Zurich, University of Zurich, Haldenbachstrasse 16/18, 8091, Zurich, Switzerland
| | - Aju Pazhenkottil
- Department of Consultation-Liaison Psychiatry and Psychosomatic Medicine, University Hospital Zurich, University of Zurich, Haldenbachstrasse 16/18, 8091, Zurich, Switzerland
- Department of Cardiology, University Hospital Zurich, Zurich, Switzerland
- Cardiac Imaging, Department of Nuclear Medicine, University Hospital Zurich, Zurich, Switzerland
| | - Diego Gomez Vieito
- Department of Consultation-Liaison Psychiatry and Psychosomatic Medicine, University Hospital Zurich, University of Zurich, Haldenbachstrasse 16/18, 8091, Zurich, Switzerland
- Institute of Molecular Cancer Research, University of Zurich, Zurich, Switzerland
| | - Roland von Känel
- Department of Consultation-Liaison Psychiatry and Psychosomatic Medicine, University Hospital Zurich, University of Zurich, Haldenbachstrasse 16/18, 8091, Zurich, Switzerland
| |
Collapse
|
2
|
Li Y, Xue L, Feng J, Wang Z, Long Y, Liu W, Zhang S, Zhi X, Hao H, Wang X, Liu H, Wang L. Insufficient BK channel function enhances NF-κB nuclear translocation and promotes IL-6 synthesis in vascular smooth muscle cells induced by AT1-AA. Biochem Pharmacol 2025:117000. [PMID: 40414513 DOI: 10.1016/j.bcp.2025.117000] [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: 11/06/2024] [Revised: 04/06/2025] [Accepted: 05/22/2025] [Indexed: 05/27/2025]
Abstract
The inflammatory phenotype of vascular smooth muscle cells (VSMCs) is an important factor in triggering vascular disease, and interleukin-6 (IL-6) is one of the earliest inflammatory cytokines upregulated in many inflammatory contexts. Angiotensin II-1 receptor autoantibody (AT1-AA) can promote the phenotypic transformation of VSMCs into macrophage-like cells, then synthesize abundant IL-6 to induce vascular inflammation. Previous studies suggested that abnormal BK channel function on the surface of VSMCs played an important role in the synthesis of IL-6, but the mechanism of abnormal BK channel involvement in AT1-AA-induced IL-6 synthesis in VSMCs was unclear. In this study, the agonist NS1619 of the BK channel and the inhibitor Paxilline were used to reverse or exacerbate IL-6 synthesis in AT1-AA-induced VSMCs. It is known that NF-κB can enter the nucleus due to increased calcium ion concentration caused by BK channel dysfunction, thereby increasing IL-6 transcription. This study observed that Paxilline pretreatment significantly increased the residence time of AT1-AA-induced NF-κB in the nucleus, while NS1619 pretreatment showed the opposite trend. JSH-23 inhibiting NF-κB nuclear entry reversed the increase in IL-6 expression in VSMCs induced by AT1-AA. This study found that AT1-AA enhanced NF-κB nuclear translocation by inhibiting BK channel function, which in turn promoted IL-6 transcription in VSMCs, increased IL-6 synthesis, and induced vascular inflammation. This study revealed the importance of BK channel dysfunction in the process of AT1-AA increasing IL-6 synthesis and promoting vascular inflammation, and provided a new idea for alleviating vascular inflammatory diseases from the perspective of improving potassium channel function.
Collapse
Affiliation(s)
- Yang Li
- School of Basic Medical Sciences, Shanxi Medical University, Taiyuan, PR China
| | - Lingxia Xue
- School of Basic Medical Sciences, Shanxi Medical University, Taiyuan, PR China
| | - Jiayan Feng
- School of Basic Medical Sciences, Shanxi Medical University, Taiyuan, PR China
| | - Zhuoxi Wang
- School of Basic Medical Sciences, Shanxi Medical University, Taiyuan, PR China
| | - Yaolin Long
- School of Basic Medical Sciences, Shanxi Medical University, Taiyuan, PR China
| | - Weiqian Liu
- School of Basic Medical Sciences, Shanxi Medical University, Taiyuan, PR China
| | - Suli Zhang
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Capital Medical University, Beijing, PR China
| | - Xiaoyan Zhi
- School of Basic Medical Sciences, Shanxi Medical University, Taiyuan, PR China
| | - Haihu Hao
- Department of Orthopaedics, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Third Hospital of Shanxi Medical University, Taiyuan, PR China
| | - Xiaohui Wang
- School of Basic Medical Sciences, Shanxi Medical University, Taiyuan, PR China
| | - Huirong Liu
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Capital Medical University, Beijing, PR China
| | - Li Wang
- School of Basic Medical Sciences, Shanxi Medical University, Taiyuan, PR China.
| |
Collapse
|
3
|
Wang X, Chen L, Wei J, Zheng H, Zhou N, Xu X, Deng X, Liu T, Zou Y. The immune system in cardiovascular diseases: from basic mechanisms to therapeutic implications. Signal Transduct Target Ther 2025; 10:166. [PMID: 40404619 PMCID: PMC12098830 DOI: 10.1038/s41392-025-02220-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2024] [Revised: 12/22/2024] [Accepted: 03/20/2025] [Indexed: 05/24/2025] Open
Abstract
Immune system plays a crucial role in the physiological and pathological regulation of the cardiovascular system. The exploration history and milestones of immune system in cardiovascular diseases (CVDs) have evolved from the initial discovery of chronic inflammation in atherosclerosis to large-scale clinical studies confirming the importance of anti-inflammatory therapy in treating CVDs. This progress has been facilitated by advancements in various technological approaches, including multi-omics analysis (single-cell sequencing, spatial transcriptome et al.) and significant improvements in immunotherapy techniques such as chimeric antigen receptor (CAR)-T cell therapy. Both innate and adaptive immunity holds a pivotal role in CVDs, involving Toll-like receptor (TLR) signaling pathway, nucleotide-binding oligomerization domain-containing proteins 1 and 2 (NOD1/2) signaling pathway, inflammasome signaling pathway, RNA and DNA sensing signaling pathway, as well as antibody-mediated and complement-dependent systems. Meanwhile, immune responses are simultaneously regulated by multi-level regulations in CVDs, including epigenetics (DNA, RNA, protein) and other key signaling pathways in CVDs, interactions among immune cells, and interactions between immune and cardiac or vascular cells. Remarkably, based on the progress in basic research on immune responses in the cardiovascular system, significant advancements have also been made in pre-clinical and clinical studies of immunotherapy. This review provides an overview of the role of immune system in the cardiovascular system, providing in-depth insights into the physiological and pathological regulation of immune responses in various CVDs, highlighting the impact of multi-level regulation of immune responses in CVDs. Finally, we also discuss pre-clinical and clinical strategies targeting the immune system and translational implications in CVDs.
Collapse
Affiliation(s)
- Xiaoyan Wang
- Shanghai Institute of Cardiovascular Diseases, Zhongshan Hospital and Institutes of Biomedical Sciences, Fudan University, Shanghai, China.
- State Key Laboratory of Cardiovascular Diseases, Zhongshan Hospital, Fudan University, Shanghai, China.
| | - Liming Chen
- Shanghai Institute of Cardiovascular Diseases, Zhongshan Hospital and Institutes of Biomedical Sciences, Fudan University, Shanghai, China
- State Key Laboratory of Cardiovascular Diseases, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Jianming Wei
- Central Diagnostics Laboratory, University Medical Center Utrecht, University Utrecht, Utrecht, The Netherlands
| | - Hao Zheng
- Jiangsu Provincial Key Laboratory of Critical Care Medicine and Department of Critical Care Medicine, School of Medicine, Zhongda Hospital, Southeast University, Nanjing, China
| | - Ning Zhou
- Department of Cardiovascular Medicine, Anzhen Hospital Affiliated to Capital Medical University, Beijing, China
| | - Xinjie Xu
- State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Xin Deng
- Shanghai Institute of Cardiovascular Diseases, Zhongshan Hospital and Institutes of Biomedical Sciences, Fudan University, Shanghai, China
- State Key Laboratory of Cardiovascular Diseases, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Tao Liu
- Jiangsu Provincial Key Laboratory of Critical Care Medicine and Department of Critical Care Medicine, School of Medicine, Zhongda Hospital, Southeast University, Nanjing, China.
- Department of Biochemistry and Molecular Biology, School of Medicine, Southeast University, Jiangsu, Nanjing, China.
- State Key Laboratory of Respiratory Disease, Joint International Research Laboratory of Respiratory Health, National Clinical Research Center for Respiratory Disease, National Center for Respiratory Medicine, Department of Allergy and Clinical Immunology, Guangzhou Institute of Respiratory Health, the First Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong, China.
| | - Yunzeng Zou
- Shanghai Institute of Cardiovascular Diseases, Zhongshan Hospital and Institutes of Biomedical Sciences, Fudan University, Shanghai, China.
- State Key Laboratory of Cardiovascular Diseases, Zhongshan Hospital, Fudan University, Shanghai, China.
- Institutes of Advanced Medical Sciences and Huaihe Hospital, Henan University, Kaifeng, Henan, China.
| |
Collapse
|
4
|
Sun J, Zhou L, Li Z, He G, Mao H, Zhao J, Hunt JA, Chen X. Perovskite-Graphene Heterostructure Biosensor Integrated with Biotunable Nanoplasmonic Ternary Logic Gate for Ultrasensitive Cytokine Detection. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025:e03124. [PMID: 40397015 DOI: 10.1002/advs.202503124] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/19/2025] [Revised: 05/01/2025] [Indexed: 05/22/2025]
Abstract
The integration of 2D-materials and optoelectronic devices has attracted great attention for advanced applications. We propose the first perovskite/graphene heterostructure-based FET biosensor with uniquely biotunable ternary logic gating functionality. The biosensor integrates a lateral perovskite-on-graphene heterostructure phototransistor with a vertical bio-nano-photonic filter, with a decoupled construction inset. In the phototransistor, photoactive perovskite quantum dots (PQDs) serve as sensitizers to absorb light while a high mobility single-layer graphene (SLG) acts as an expressway for carrier transport. In the bio-nano-photonic filter, a localized surface plasmon resonance (LSPR) is induced by gold nanoparticles (AuNPs) in conjunction with antigen-antibody binding, tuning the delivery of light passing through the filter and facilitating biotunable functionality with ternary modes. The biosensor is set up to detect human interleukin-6 (IL6) in order to determine and achieve ultrahigh sensitivity with a limit of detection (LOD) of 0.9 fg mL-1 (43 aM), which is 4 orders of magnitude greater than graphene-FET biosensors. This ultrahigh sensitivity is achieved due to the synergistic effect of PQDs/SLG heterostructure, exhibiting superior electrical, optical, and physicochemical properties, consequently providing significantly high performance of the biosensor in terms of label-free, ultrahigh sensitivity (attomolar level), rapid responsivity (5 min), excellent stability, and selectivity. This heterostructure-based biotunable configuration could open a new avenue for 2D materials in the realm of next-generation bio-nano-photonic platforms for applications in healthcare, early diagnosis, and rapid detection.
Collapse
Affiliation(s)
- Jiaxing Sun
- School of Science and Technology, Nottingham Trent University, Nottingham, NG11 8NS, UK
| | - Lin Zhou
- Key Laboratory of Transducer Technology, Shanghai Institute of Microsystem and Information Technology, Chinese Academy of Sciences, Shanghai, 200050, China
| | - Zening Li
- Key Laboratory of Transducer Technology, Shanghai Institute of Microsystem and Information Technology, Chinese Academy of Sciences, Shanghai, 200050, China
| | - Guolin He
- General Surgery Center, Department of Hepatobiliary Surgery II, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong, 510280, China
| | - Hongju Mao
- Key Laboratory of Transducer Technology, Shanghai Institute of Microsystem and Information Technology, Chinese Academy of Sciences, Shanghai, 200050, China
| | - Jianlong Zhao
- Key Laboratory of Transducer Technology, Shanghai Institute of Microsystem and Information Technology, Chinese Academy of Sciences, Shanghai, 200050, China
| | - John A Hunt
- Medical Technologies Innovation Facility, Nottingham Trent University, Nottingham, NG11 8NS, UK
| | - Xianfeng Chen
- School of Science and Technology, Nottingham Trent University, Nottingham, NG11 8NS, UK
| |
Collapse
|
5
|
Oberther TJ, Moore AR, Kohler AA, Holland-Winkler AM. Relationship Between Systemic Inflammation and Glycemic Control in Firefighters. J Funct Morphol Kinesiol 2025; 10:148. [PMID: 40407432 PMCID: PMC12101145 DOI: 10.3390/jfmk10020148] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/28/2025] [Revised: 04/21/2025] [Accepted: 04/25/2025] [Indexed: 05/26/2025] Open
Abstract
Background: Firefighters are at risk for developing metabolic diseases such as type 2 diabetes due to occupational-related stress and poor health behaviors. Firefighters often experience chronic inflammatory responses that may contribute to the development of insulin resistance. This study examined the relationship between systemic inflammation markers and glycemic control markers in firefighters. Methods: Blood samples were collected from twenty full-time male firefighters to assess HbA1c, fasting glucose, and insulin to estimate the Homeostatic Model of Assessment of Insulin Resistance (HOMA-IR), C-reactive protein (CRP), and homocysteine. Body composition and cardiovascular metrics were also recorded. Pearson partial correlation analyses were performed to evaluate relationships between homocysteine and CRP and the variables HOMA-IR and HbA1c while controlling for age and body fat percentage (BF%). SPSS version 29 was used for all analyses (α = 0.05). Data transformation was used where appropriate to ensure the normal distribution of each variable. Results: A significant positive correlation was found between homocysteine and HbA1c before (p = 0.006, r = 0.605) and after controlling for age and BF% (ppartial = 0.013, rpartial = 0.588), indicating that homocysteine levels are associated with impaired glycemic control in firefighters. No other relationships were found to be significant. Conclusions: The findings support a potential link between systemic inflammation and poor glycemic control in firefighters. Due to the occupational hazards that contribute to chronic inflammation, targeted interventions such as dietary modifications may help decrease the risk of diabetes and cardiovascular disease in this high-risk population.
Collapse
|
6
|
Fan J, Yang Y, Yang Y, Rui D, Guo S, Li M, Wang Y, Feng Y, Zhao C. Predictive role of the dietary inflammatory index on stroke risk among hypertensive patients. Sci Rep 2025; 15:13602. [PMID: 40253411 PMCID: PMC12009329 DOI: 10.1038/s41598-025-96908-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2024] [Accepted: 04/01/2025] [Indexed: 04/21/2025] Open
Abstract
This study aims to evaluate the association between the dietary inflammatory index (DII) and stroke risk in hypertensive patients. Data were sourced from the National Health and Nutrition Examination Survey (NHANES) spanning 1999-2020, including 23,712 hypertensive patients. DII scores were calculated based on dietary intake data, and stroke diagnoses were determined through self-reported physician diagnoses. The relationship between DII and stroke risk was assessed using multivariable logistic regression models. Dose-response relationships and subgroup differences were explored through stratified analysis and restricted cubic spline (RCS) methods. Key dietary factors associated with stroke were identified using least absolute shrinkage and selection operator (LASSO) regression and incorporated into a risk prediction nomogram model. The model's discriminatory ability for stroke was evaluated using receiver operating characteristic (ROC) curves. After adjusting for confounding factors, the highest DII quartile was associated with an adjusted odds ratio (OR) of 1.44 (95% CI 1.19, 1.74) for stroke compared to the lowest quartile, and each unit increase in DII was associated with an OR of 1.08 (95% CI 1.04-1.13) for stroke prevalence. The RCS curve demonstrated a nonlinear relationship between DII and stroke, with a turning point at 0.29. The nomogram model based on key dietary factors identified by LASSO regression had an area under the curve (AUC) of 70.93% (95% CI 69.81%-72.06%). There is a nonlinear relationship between DII and stroke risk in hypertensive patients. Given the inherent limitations of a cross-sectional study design, further research is needed to establish causality.
Collapse
Affiliation(s)
- Jingru Fan
- Jinan University, No.601.West of Huangpu Avenue, Guangzhou, 510632, China
- Shantou Central Hospital, Shantou, Guangdong, China
| | - Yane Yang
- Department of Cardiology, The Second People's Hospital of Baoshan, Baoshan, Yunnan, China
| | - Yunjie Yang
- Kunming City Maternal and Child Health Hospital, Yunnan, China
| | - Dewei Rui
- Shantou Central Hospital, Shantou, Guangdong, China
| | - Shunqi Guo
- Shantou Central Hospital, Shantou, Guangdong, China
| | - Meiju Li
- Department of Cardiology, The Second People's Hospital of Baoshan, Baoshan, Yunnan, China
| | - Yibing Wang
- Department of Cardiology, The Second People's Hospital of Baoshan, Baoshan, Yunnan, China
| | - Yingqing Feng
- Jinan University, No.601.West of Huangpu Avenue, Guangzhou, 510632, China.
- Department of Cardiology, Guangdong Cardiovascular Institute, Guangdong Provincial People's Hospital, Guangzhou, China.
| | - Chuanwei Zhao
- Department of Cardiology, The Second People's Hospital of Baoshan, Baoshan, Yunnan, China.
| |
Collapse
|
7
|
Steffens J, Kuth K, Kraus T, Dott W, Michael S, Baumann R. Inflammatory Responses to Zn/Cu-Containing Welding Fume in Human Alveolar Epithelial and Macrophage Cell Lines, with MIP-1β/CCL4 as a Much More Sensitive Macrophage Activation Marker than IL-8 and TNF-α. Int J Mol Sci 2025; 26:3843. [PMID: 40332485 PMCID: PMC12027897 DOI: 10.3390/ijms26083843] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2025] [Revised: 04/10/2025] [Accepted: 04/15/2025] [Indexed: 05/08/2025] Open
Abstract
Zinc (Zn)- and copper (Cu)-containing welding fumes elevate inflammatory markers (CRP, TNF-α, IL-6, IL-8) in healthy individuals and welders. Zn- and Cu-containing nanoparticles are toxic to human macrophages. Therefore, ZnO exposure limits are under discussion. In this study, the effects of Zn/Cu-containing welding fume suspensions on A549 alveolar epithelial cells (exposure concentrations: 0.01/0.1/1/10/100 µg/mL) and THP-1 macrophages (additionally 0.001 µg/mL) were investigated over a period of 48 h. Effects on apoptosis, cytotoxicity, genotoxicity, superoxide dismutase (SOD) activity, and cytokine levels (IL-6, IL-8, MIP-1β/CCL4, TNF-α) were evaluated. Welding fume exposure increased SOD activity, and it increased Annexin-V binding and cytotoxicity effects starting at 10 µg/mL in A549 cells and particularly in THP-1 macrophages. A549 cells showed increased IL-6 at 10 and 100 µg/mL, and significant IL-8 release occurred at 10 µg/mL for A549 and 0.1 µg/mL for macrophages. Exposed macrophages released TNF-α at 1 µg/mL after 24 and 48 h and MIP-1β/CCL4 at 0.01 µg/mL after 6 h and at 0.001 µg/mL after 48 h. No genotoxic effects were detected. MIP-1β/CCL4 is a sensitive new biomarker for human macrophages exposed to Zn/Cu-containing welding fumes. The findings suggest that Zn/Cu particles affect lung cells already at doses below current occupational thresholds.
Collapse
Affiliation(s)
- Jan Steffens
- Institute for Occupational, Social and Environmental Medicine, Medical Faculty, RWTH Aachen University, Pauwelsstrasse 30, 52074 Aachen, Germany
- Institute for Translational Medicine, Medical Faculty, Medical School Hamburg (MSH), Am Kaiserkai 1, 20457 Hamburg, Germany
| | - Katharina Kuth
- Institute for Occupational, Social and Environmental Medicine, Medical Faculty, RWTH Aachen University, Pauwelsstrasse 30, 52074 Aachen, Germany
- Institute of Hygiene and Environmental Medicine, Medical Faculty, RWTH Aachen University, Pauwelsstrasse 30, 52074 Aachen, Germany
| | - Thomas Kraus
- Institute for Occupational, Social and Environmental Medicine, Medical Faculty, RWTH Aachen University, Pauwelsstrasse 30, 52074 Aachen, Germany
| | - Wolfgang Dott
- Institute of Hygiene and Environmental Medicine, Medical Faculty, RWTH Aachen University, Pauwelsstrasse 30, 52074 Aachen, Germany
| | - Sabrina Michael
- Institute of Hygiene and Environmental Medicine, Medical Faculty, RWTH Aachen University, Pauwelsstrasse 30, 52074 Aachen, Germany
| | - Ralf Baumann
- Institute for Occupational, Social and Environmental Medicine, Medical Faculty, RWTH Aachen University, Pauwelsstrasse 30, 52074 Aachen, Germany
- Institute for Translational Medicine, Medical Faculty, Medical School Hamburg (MSH), Am Kaiserkai 1, 20457 Hamburg, Germany
| |
Collapse
|
8
|
Mageswaran N, Zainal SH, Hassan NI, Abd Karim NH, Ismail NAS. Emerging Biomarkers and Electrochemical Biosensors for Early Detection of Premature Coronary Artery Disease. Diagnostics (Basel) 2025; 15:940. [PMID: 40218291 PMCID: PMC11988804 DOI: 10.3390/diagnostics15070940] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2025] [Revised: 04/04/2025] [Accepted: 04/05/2025] [Indexed: 04/14/2025] Open
Abstract
Coronary artery disease (CAD) is one of the primary causes of morbidity and death worldwide. Premature CAD (pCAD) is the term used to describe the 3-10% of CAD occurrences that occur in people under 45 worldwide. Diagnostic difficulties arise from the different risk factor profiles of pCAD and late-onset CAD. Better cardiovascular risk prediction in younger populations has been made possible by the development of biomarker detection tools. This can be applied to a diagnostic tool, including electrochemical biosensors, which have been predicted to be instrumental because of their adaptability for point-of-care applications for quicker diagnoses. These biosensors provide efficient, scalable, and reasonably priced solutions for the quick identification and tracking of CAD. Multiplex biomarker detection has been adopted as a viable approach for early diagnosis and risk assessment due to the constraints of using a single biomarker for pCAD diagnosis. Thus, this study looks at current developments in biosensing technology and discusses established and new cardiac biomarker panels for pCAD identification.
Collapse
Affiliation(s)
- Nanthini Mageswaran
- Department of Medicine, Faculty of Medicine, Universiti Kebangsaan Malaysia, Kuala Lumpur 56000, Malaysia;
| | - Sarah Husnaini Zainal
- Department of Biochemistry, Faculty of Medicine, Universiti Kebangsaan Malaysia, Kuala Lumpur 56000, Malaysia;
| | - Nurul Izzaty Hassan
- Department of Chemical Sciences, Faculty of Science & Technology, Universiti Kebangsaan Malaysia, Bangi 43600, Malaysia; (N.I.H.); (N.H.A.K.)
| | - Nurul Huda Abd Karim
- Department of Chemical Sciences, Faculty of Science & Technology, Universiti Kebangsaan Malaysia, Bangi 43600, Malaysia; (N.I.H.); (N.H.A.K.)
| | - Noor Akmal Shareela Ismail
- Department of Biochemistry, Faculty of Medicine, Universiti Kebangsaan Malaysia, Kuala Lumpur 56000, Malaysia;
| |
Collapse
|
9
|
Grytten E, Laupsa-Borge J, Cetin K, Bohov P, Nordrehaug JE, Skorve J, Berge RK, Strand E, Bjørndal B, Nygård OK, Rostrup E, Mellgren G, Dankel SN. Inflammatory markers after supplementation with marine n-3 or plant n-6 PUFAs: A randomized double-blind crossover study. J Lipid Res 2025; 66:100770. [PMID: 40058591 PMCID: PMC11999210 DOI: 10.1016/j.jlr.2025.100770] [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: 10/06/2022] [Revised: 03/03/2025] [Accepted: 03/06/2025] [Indexed: 04/04/2025] Open
Abstract
Omega-3 (n-3) (e.g., EPA/DHA) and omega-6 (n-6) (e.g., linoleic acid [LA]) FAs are suggested to have opposite effects on inflammation, but results are inconsistent and direct comparisons of n-3 and n-6 are lacking. In a double-blind, randomized, and crossover study, females (n = 16) and males (n = 23) aged 30-70 years with abdominal obesity were supplemented with 3-4 g/d EPA/DHA (fish oil) or 15-20 g/d LA (safflower oil) for 7 weeks, with a 9-week washout phase. Cytokines and chemokines (multiplex assay), acute-phase proteins (MALDI-TOF mass spectrometry), endothelial function (vascular reaction index), blood pressure, FA composition (red blood cell membranes/serum/adipose tissue, GC-MS/MS), and adipose gene expression (microarrays, quantitative PCR) were measured. While significant differences between treatments in relative change scores were found for systolic blood pressure (n-3 vs. n-6: -1.81% vs. 2.61%, P = 0.003), no differences between n-3 and n-6 were found for any circulatory inflammatory markers. However, compared with baseline, n-3 was followed by reductions in circulating TNF (-24.9%, P < 0.001), regulated upon activation, normal T cell expressed and secreted (-12.1%, P < 0.001), and macrophage inflammatory protein 1-beta (-12.5%, P = 0.014), and n-6 by lowered TNF (-18.8%, P < 0.001), regulated upon activation, normal T cell expressed and secreted (-7.37%, P = 0.027), monocyte chemoattractant protein-1 (-7.81%, P = 0.020), and macrophage inflammatory protein 1-beta (-14.2%, P = 0.010). Adipose tissue showed significant treatment differences in weight percent of EPA (n-3 vs. n-6: 50.2%∗ vs. -1.38%, P < 0.001, ∗: significant within-treatment change score), DHA (16.0%∗ vs. -3.67%, P < 0.001), and LA (-0.033 vs. 4.91%∗, P < 0.001). Adipose transcriptomics revealed overall downregulation of genes related to inflammatory processes after n-3 and upregulation after n-6, partly correlating with changes in circulatory markers. These data point to tissue-specific proinflammatory effects of high n-6 intake, but a net systemic anti-inflammatory effect as for n-3.
Collapse
Affiliation(s)
- Elise Grytten
- Hormone Laboratory, Department of Medical Biochemistry and Pharmacology, Haukeland University Hospital, Bergen, Norway; Mohn Nutrition Research Laboratory, Department of Clinical Science, University of Bergen, Bergen, Norway
| | - Johnny Laupsa-Borge
- Hormone Laboratory, Department of Medical Biochemistry and Pharmacology, Haukeland University Hospital, Bergen, Norway; Mohn Nutrition Research Laboratory, Department of Clinical Science, University of Bergen, Bergen, Norway; Bevital AS, Bergen, Norway
| | - Kaya Cetin
- Hormone Laboratory, Department of Medical Biochemistry and Pharmacology, Haukeland University Hospital, Bergen, Norway; Mohn Nutrition Research Laboratory, Department of Clinical Science, University of Bergen, Bergen, Norway
| | - Pavol Bohov
- Department of Clinical Science, University of Bergen, Bergen, Norway
| | - Jan Erik Nordrehaug
- Department of Clinical Science, University of Bergen, Bergen, Norway; Department of Heart Disease, Haukeland University Hospital, Bergen, Norway
| | - Jon Skorve
- Department of Clinical Science, University of Bergen, Bergen, Norway
| | - Rolf K Berge
- Department of Clinical Science, University of Bergen, Bergen, Norway
| | - Elin Strand
- Department of Clinical Science, University of Bergen, Bergen, Norway
| | - Bodil Bjørndal
- Department of Clinical Science, University of Bergen, Bergen, Norway
| | - Ottar K Nygård
- Department of Clinical Science, University of Bergen, Bergen, Norway; Department of Heart Disease, Haukeland University Hospital, Bergen, Norway
| | - Espen Rostrup
- Department of Heart Disease, Haukeland University Hospital, Bergen, Norway
| | - Gunnar Mellgren
- Hormone Laboratory, Department of Medical Biochemistry and Pharmacology, Haukeland University Hospital, Bergen, Norway; Mohn Nutrition Research Laboratory, Department of Clinical Science, University of Bergen, Bergen, Norway
| | - Simon N Dankel
- Hormone Laboratory, Department of Medical Biochemistry and Pharmacology, Haukeland University Hospital, Bergen, Norway; Mohn Nutrition Research Laboratory, Department of Clinical Science, University of Bergen, Bergen, Norway.
| |
Collapse
|
10
|
Thakur M, Philip ST, Desai KR, Thakur K. Effect of Heartfulness Meditation on Stress Biomarkers, Burnout and Well-Being: A Randomized Controlled Study. Stress Health 2025; 41:e70034. [PMID: 40271908 DOI: 10.1002/smi.70034] [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: 10/29/2024] [Revised: 03/29/2025] [Accepted: 04/06/2025] [Indexed: 04/25/2025]
Abstract
The rise in mental health conditions and stress has attracted global attention. Non-pharmacological and traditional approaches like meditative practices are showing promising results. Therefore, the aim of the study was to evaluate the effect of Heartfulness meditation on stress biomarkers, burnout, and well-being. A double-arm prospective randomized controlled study was carried out on 100 participants aged between 18 and 24 years, experiencing moderate to high perceived stress as assessed by the Perceived Stress Scale (PSS-10). These participants were randomized into a study group (meditation) and control group (sham meditation). An intervention of Heartfulness meditation was carried out daily for 12 weeks. Psychometric analysis was carried out to study burnout (exhaustion, cynicism and professional efficacy) and well-being using the standard validated questionnaires- Maslach Burnout Inventory (MBI) and WHO-Well-being Index (WHO-WBI). Biochemical analysis was also carried out to study psychological stress (serum cortisol), and oxidative stress (serum nitrate/nitrite and serum malondialdehyde (MDA) at baseline and post-intervention. Statistical analysis was carried out using the IBM SPSS software version 26.0. p values < 0.05 were considered statistically significant. A significant increase in serum nitrate/nitrite levels, professional efficacy, and well-being, and a significant decrease in measures such as exhaustion, cynicism, and cortisol levels were observed in the study group than the control group post-intervention. A significant negative correlation was also observed between serum MDA and well-being, whereas a positive correlation was observed between MDA and cortisol. The findings from this research suggest the role of Heartfulness meditation in reducing stress and burnout along with improving the well-being of an individual. Therefore, Heartfulness meditation can be used as a potential tool to improve mental health and well-being. However, future studies with a larger number of samples are needed to strengthen our findings. Trial Registration: Clinical Trial Registry of India: CTRI/2023/10/058,423 (https://ctri.nic.in/Clinicaltrials/pmaindet2.php?EncHid=ODIyOTA=&Enc=&userName=CTRI/2023/10/058423).
Collapse
Affiliation(s)
- Mansee Thakur
- Department of Medical Biotechnology, Central Research Laboratory, Mahatma Gandhi Mission School of Biomedical Sciences, Mahatma Gandhi Mission Institute of Health Sciences, Kamothe, Navi Mumbai, Maharashtra, India
| | - Sanjana T Philip
- Department of Medical Biotechnology, Central Research Laboratory, Mahatma Gandhi Mission School of Biomedical Sciences, Mahatma Gandhi Mission Institute of Health Sciences, Kamothe, Navi Mumbai, Maharashtra, India
| | - Kunal R Desai
- Department of Internal Medicine, Boonshoft School of Medicine, Wright State University, Dayton, Ohio, USA
| | - Kapil Thakur
- Central Research Lab, MGM Medical College and Hospital, Mahatma Gandhi Mission Institute of Health Sciences, Nerul, Navi Mumbai, Maharashtra, India
| |
Collapse
|
11
|
Chaiwong W, Liwsrisakun C, Inchai J, Duangjit P, Bumroongkit C, Deesomchok A, Theerakittikul T, Limsukon A, Tajarernmuang P, Niyatiwatchanchai N, Trongtrakul K, Chitchun C, Chattipakorn N, Chattipakorn SC, Apaijai N, Pothirat C. Biomarkers of Oxidative Stress, Systemic Inflammation and Thrombosis in Adult Asthmatic Patients Treated with Inhaled Corticosteroids During Exposure to Fine Particulate Matter. J Clin Med 2025; 14:2360. [PMID: 40217808 PMCID: PMC11989988 DOI: 10.3390/jcm14072360] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2025] [Revised: 03/25/2025] [Accepted: 03/27/2025] [Indexed: 04/14/2025] Open
Abstract
Background/Objectives: Inhaled corticosteroids (ICS) affect oxidative stress and systemic inflammation, which might modify the risk of thrombosis in asthmatic patients exposed to particulate matter with an aerodynamic diameter smaller than 2.5 microns (PM2.5). Therefore, we aim to know the systemic biomarkers of oxidative stress, inflammation, and coagulation in ICS-treated, well-controlled adult asthmatic patients after exposure to PM2.5. Methods: This study was conducted to compare urinary biomarkers of oxidative stress, i.e., 8-hydroxydeoxyguanosine (8-OHdG), and blood biomarkers of inflammation and hypercoagulation, i.e., complete blood count (CBC), high-sensitivity C-reactive protein (hsCRP), fibrinogen, D-dimer, tumor necrosis factor-alpha (TNF-α), and interleukins (IL-6 and IL-8), between well-controlled adult asthmatic patients and healthy controls in low and high-pollution periods. Results: Forty-one ICS-controlled asthmatic patients and twenty controls were included. Urinary 8-OHdG, white blood cells and differential counts, platelets count, hsCRP, IL-6, and IL-8 in the asthma group were not significantly higher than controls during the same period. The D-dimer level of the asthma patients was significantly higher than the controls (p < 0.05). The median level of TNF-α levels during the pollution period in asthma patients was significantly higher than the non-pollution period with levels of 14.3 (9.3, 27.4) and 11.3 (7.8, 21.1) pg/mL, p = 0.041, respectively. Conclusions: During exposure to PM2.5, serum TNF-α was increased while the other markers of oxidative stress and inflammation were not high in ICS-treated asthma. ICS might mitigate PM2.5-induced systemic oxidative stress, inflammation, and hypercoagulation in asthma.
Collapse
Affiliation(s)
- Warawut Chaiwong
- Division of Pulmonary, Critical Care, and Allergy, Department of Internal Medicine, Faculty of Medicine, Chiang Mai University, Chiang Mai 50200, Thailand; (W.C.); (J.I.); (P.D.); (C.B.); (A.D.); (T.T.); (A.L.); (P.T.); (N.N.); (K.T.); (C.C.); (C.P.)
| | - Chalerm Liwsrisakun
- Division of Pulmonary, Critical Care, and Allergy, Department of Internal Medicine, Faculty of Medicine, Chiang Mai University, Chiang Mai 50200, Thailand; (W.C.); (J.I.); (P.D.); (C.B.); (A.D.); (T.T.); (A.L.); (P.T.); (N.N.); (K.T.); (C.C.); (C.P.)
| | - Juthamas Inchai
- Division of Pulmonary, Critical Care, and Allergy, Department of Internal Medicine, Faculty of Medicine, Chiang Mai University, Chiang Mai 50200, Thailand; (W.C.); (J.I.); (P.D.); (C.B.); (A.D.); (T.T.); (A.L.); (P.T.); (N.N.); (K.T.); (C.C.); (C.P.)
| | - Pilaiporn Duangjit
- Division of Pulmonary, Critical Care, and Allergy, Department of Internal Medicine, Faculty of Medicine, Chiang Mai University, Chiang Mai 50200, Thailand; (W.C.); (J.I.); (P.D.); (C.B.); (A.D.); (T.T.); (A.L.); (P.T.); (N.N.); (K.T.); (C.C.); (C.P.)
| | - Chaiwat Bumroongkit
- Division of Pulmonary, Critical Care, and Allergy, Department of Internal Medicine, Faculty of Medicine, Chiang Mai University, Chiang Mai 50200, Thailand; (W.C.); (J.I.); (P.D.); (C.B.); (A.D.); (T.T.); (A.L.); (P.T.); (N.N.); (K.T.); (C.C.); (C.P.)
| | - Athavudh Deesomchok
- Division of Pulmonary, Critical Care, and Allergy, Department of Internal Medicine, Faculty of Medicine, Chiang Mai University, Chiang Mai 50200, Thailand; (W.C.); (J.I.); (P.D.); (C.B.); (A.D.); (T.T.); (A.L.); (P.T.); (N.N.); (K.T.); (C.C.); (C.P.)
| | - Theerakorn Theerakittikul
- Division of Pulmonary, Critical Care, and Allergy, Department of Internal Medicine, Faculty of Medicine, Chiang Mai University, Chiang Mai 50200, Thailand; (W.C.); (J.I.); (P.D.); (C.B.); (A.D.); (T.T.); (A.L.); (P.T.); (N.N.); (K.T.); (C.C.); (C.P.)
| | - Atikun Limsukon
- Division of Pulmonary, Critical Care, and Allergy, Department of Internal Medicine, Faculty of Medicine, Chiang Mai University, Chiang Mai 50200, Thailand; (W.C.); (J.I.); (P.D.); (C.B.); (A.D.); (T.T.); (A.L.); (P.T.); (N.N.); (K.T.); (C.C.); (C.P.)
| | - Pattraporn Tajarernmuang
- Division of Pulmonary, Critical Care, and Allergy, Department of Internal Medicine, Faculty of Medicine, Chiang Mai University, Chiang Mai 50200, Thailand; (W.C.); (J.I.); (P.D.); (C.B.); (A.D.); (T.T.); (A.L.); (P.T.); (N.N.); (K.T.); (C.C.); (C.P.)
| | - Nutchanok Niyatiwatchanchai
- Division of Pulmonary, Critical Care, and Allergy, Department of Internal Medicine, Faculty of Medicine, Chiang Mai University, Chiang Mai 50200, Thailand; (W.C.); (J.I.); (P.D.); (C.B.); (A.D.); (T.T.); (A.L.); (P.T.); (N.N.); (K.T.); (C.C.); (C.P.)
| | - Konlawij Trongtrakul
- Division of Pulmonary, Critical Care, and Allergy, Department of Internal Medicine, Faculty of Medicine, Chiang Mai University, Chiang Mai 50200, Thailand; (W.C.); (J.I.); (P.D.); (C.B.); (A.D.); (T.T.); (A.L.); (P.T.); (N.N.); (K.T.); (C.C.); (C.P.)
| | - Chittrawadee Chitchun
- Division of Pulmonary, Critical Care, and Allergy, Department of Internal Medicine, Faculty of Medicine, Chiang Mai University, Chiang Mai 50200, Thailand; (W.C.); (J.I.); (P.D.); (C.B.); (A.D.); (T.T.); (A.L.); (P.T.); (N.N.); (K.T.); (C.C.); (C.P.)
| | - Nipon Chattipakorn
- Neurophysiology Unit, Cardiac Electrophysiology Research and Training Center, Faculty of Medicine, Chiang Mai University, Chiang Mai 50200, Thailand; (N.C.); (S.C.C.); (N.A.)
- Cardiac Electrophysiology Unit, Department of Physiology, Faculty of Medicine, Chiang Mai University, Chiang Mai 50200, Thailand
- Center of Excellence in Cardiac Electrophysiology, Chiang Mai University, Chiang Mai 50200, Thailand
| | - Siriporn C. Chattipakorn
- Neurophysiology Unit, Cardiac Electrophysiology Research and Training Center, Faculty of Medicine, Chiang Mai University, Chiang Mai 50200, Thailand; (N.C.); (S.C.C.); (N.A.)
- Center of Excellence in Cardiac Electrophysiology, Chiang Mai University, Chiang Mai 50200, Thailand
- Department of Oral Biology and Diagnostic Sciences, Faculty of Dentistry, Chiang Mai University, Chiang Mai 50200, Thailand
| | - Nattayaporn Apaijai
- Neurophysiology Unit, Cardiac Electrophysiology Research and Training Center, Faculty of Medicine, Chiang Mai University, Chiang Mai 50200, Thailand; (N.C.); (S.C.C.); (N.A.)
- Cardiac Electrophysiology Unit, Department of Physiology, Faculty of Medicine, Chiang Mai University, Chiang Mai 50200, Thailand
- Center of Excellence in Cardiac Electrophysiology, Chiang Mai University, Chiang Mai 50200, Thailand
| | - Chaicharn Pothirat
- Division of Pulmonary, Critical Care, and Allergy, Department of Internal Medicine, Faculty of Medicine, Chiang Mai University, Chiang Mai 50200, Thailand; (W.C.); (J.I.); (P.D.); (C.B.); (A.D.); (T.T.); (A.L.); (P.T.); (N.N.); (K.T.); (C.C.); (C.P.)
| |
Collapse
|
12
|
Annink ME, Kraaijenhof JM, Beverloo CYY, Oostveen RF, Verberne HJ, Stroes ESG, Nurmohamed NS. Estimating inflammatory risk in atherosclerotic cardiovascular disease: plaque over plasma? Eur Heart J Cardiovasc Imaging 2025; 26:444-460. [PMID: 39657321 PMCID: PMC11879196 DOI: 10.1093/ehjci/jeae314] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/28/2024] [Revised: 11/04/2024] [Accepted: 11/29/2024] [Indexed: 12/12/2024] Open
Abstract
Inflammation is an important driver of disease in the context of atherosclerosis, and several landmark trials have shown that targeting inflammatory pathways can reduce cardiovascular event rates. However, the high cost and potentially serious adverse effects of anti-inflammatory therapies necessitate more precise patient selection. Traditional biomarkers of inflammation, such as high-sensitivity C-reactive protein, show an association with cardiovascular risk on a population level but do not have specificity for local plaque inflammation. Nowadays, advancements in non-invasive imaging of the vasculature enable direct assessment of vascular inflammation. Positron emission tomography (PET) tracers such as 18F-fluorodeoxyglucose enable detection of metabolic activity of inflammatory cells but are limited by low specificity and myocardial spillover effects. 18F-sodium fluoride is a tracer that identifies active micro-calcification in plaques, indicating vulnerable plaques. Gallium-68 DOTATATE targets pro-inflammatory macrophages by binding to somatostatin receptors, which enhances specificity for plaque inflammation. Coronary computed tomography angiography (CCTA) provides high-resolution images of coronary arteries, identifying high-risk plaque features. Measuring pericoronary adipose tissue attenuation on CCTA represents a novel marker of vascular inflammation. This review examines both established and emerging methods for assessing atherosclerosis-related inflammation, emphasizing the role of advanced imaging in refining risk stratification and guiding personalized therapies. Integrating these imaging modalities with measurements of systemic and molecular biomarkers could shift atherosclerotic cardiovascular disease management towards a more personalized approach.
Collapse
Affiliation(s)
- Maxim E Annink
- Department of Vascular Medicine, Amsterdam UMC, University of Amsterdam, Meibergdreef 9, 1105AZ Amsterdam, The Netherlands
| | - Jordan M Kraaijenhof
- Department of Vascular Medicine, Amsterdam UMC, University of Amsterdam, Meibergdreef 9, 1105AZ Amsterdam, The Netherlands
| | - Cheyenne Y Y Beverloo
- Department of Vascular Medicine, Amsterdam UMC, University of Amsterdam, Meibergdreef 9, 1105AZ Amsterdam, The Netherlands
| | - Reindert F Oostveen
- Department of Vascular Medicine, Amsterdam UMC, University of Amsterdam, Meibergdreef 9, 1105AZ Amsterdam, The Netherlands
| | - Hein J Verberne
- Department of Radiology & Nuclear Medicine, Amsterdam UMC, University of Amsterdam, Meibergdreef 9, 1105AZ Amsterdam, The Netherlands
| | - Erik S G Stroes
- Department of Vascular Medicine, Amsterdam UMC, University of Amsterdam, Meibergdreef 9, 1105AZ Amsterdam, The Netherlands
| | - Nick S Nurmohamed
- Department of Vascular Medicine, Amsterdam UMC, University of Amsterdam, Meibergdreef 9, 1105AZ Amsterdam, The Netherlands
- Department of Cardiology, Amsterdam UMC, Vrije Universiteit Amsterdam, De Boelelaan 1117, 1081HV Amsterdam, The Netherlands
| |
Collapse
|
13
|
Rajput S, Malviya R, Srivastava S, Ahmad I, Rab SO, Uniyal P. Cardiovascular disease and thrombosis: Intersections with the immune system, inflammation, and the coagulation system. ANNALES PHARMACEUTIQUES FRANÇAISES 2025; 83:228-250. [PMID: 39159826 DOI: 10.1016/j.pharma.2024.08.005] [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: 01/11/2024] [Revised: 08/06/2024] [Accepted: 08/13/2024] [Indexed: 08/21/2024]
Abstract
The coagulation and immune system, both essential physiological systems in the human body, are intricately interconnected and play a critical role in determining the overall health of patients. These systems collaborate via various shared regulatory pathways, such as the Tissue Factor (TF) Pathway. Immunological cells that express TF and generate pro-inflammatory cytokines have the ability to affect coagulation. Conversely, coagulation factors and processes have a reciprocal effect on immunological responses by stimulating immune cells and regulating their functions. These interconnected pathways play a role in both preserving well-being and contributing to a range of pathological disorders. The close relationship between blood clotting and inflammation in the development of vascular disease has become a central focus of clinical study. This research specifically examines the crucial elements of this interaction within the contexts of cardiovascular disease and acute coronary syndrome. Tissue factor, the primary trigger of the extrinsic coagulation pathway, has a crucial function by inducing a proinflammatory reaction through the activation of coagulation factors. This, in turn, initiates coagulation and subsequent cellular signalling pathways. Protease-activated receptors establish the molecular connection between coagulation and inflammation by interacting with activated clotting factors II, X, and VII. Thrombosis, a condition characterised by the formation of blood clots, is the most dreaded consequence of cardiovascular disorders and a leading cause of death globally. Consequently, it poses a significant challenge to healthcare systems. Antithrombotic treatments efficiently target platelets and the coagulation cascade, but they come with the inherent danger of causing bleeding. Furthermore, antithrombotics are unable to fully eliminate thrombotic events, highlighting a treatment deficiency caused by a third mechanism that has not yet been sufficiently addressed, namely inflammation. Understanding these connections may aid in the development of novel approaches to mitigate the harmful mutual exacerbation of inflammation and coagulation. Gaining a comprehensive understanding of the intricate interaction among these systems is crucial for the management of diseases and the creation of efficacious remedies. Through the examination of these prevalent regulatory systems, we can discover novel therapeutic approaches that specifically target these complex illnesses. This paper provides a thorough examination of the reciprocal relationship between the coagulation and immune systems, emphasising its importance in maintaining health and understanding disease processes. This review examines the interplay between inflammation and thrombosis and its role in the development of thrombotic disorders.
Collapse
Affiliation(s)
- Shivam Rajput
- Department of Pharmacy, School of Medical and Allied Sciences, Galgotias University, Greater Noida, U.P., India
| | - Rishabha Malviya
- Department of Pharmacy, School of Medical and Allied Sciences, Galgotias University, Greater Noida, U.P., India.
| | - Saurabh Srivastava
- School of Pharmacy, KPJ Healthcare University College (KPJUC), Nilai, Malaysia
| | - Irfan Ahmad
- Department of Clinical Laboratory Sciences, College of Applied Medical Science, King Khalid University, Abha, Saudi Arabia
| | - Safia Obaidur Rab
- Department of Clinical Laboratory Sciences, College of Applied Medical Science, King Khalid University, Abha, Saudi Arabia
| | - Prerna Uniyal
- School of Pharmacy, Graphic Era Hill University, Dehradun, India
| |
Collapse
|
14
|
Blokhina T, Kirichenko T, Markina Y, Khovantseva U, Melnikov I, Guseva O, Bazanovich S, Kozlov S, Orekhov A. Features of the monocyte inflammatory response in patients with premature coronary artery disease. BIOPHYSICS REPORTS 2025; 11:12-17. [PMID: 40070664 PMCID: PMC11891073 DOI: 10.52601/bpr.2024.240030] [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: 07/11/2024] [Accepted: 08/19/2024] [Indexed: 03/14/2025] Open
Abstract
The purpose of this study was to examine the secretion of inflammatory cytokines by cultured monocytes/macrophages in patients with premature coronary artery disease (CAD). The study included 38 patients with premature CAD and 35 patients without CAD. A primary culture of CD14+ monocytes was obtained by immunomagnetic separation. The inflammatory response was induced by incubation of a cell culture with lipopolysaccharide (LPS) for 24 hours on Days 1 and 6. Basal and LPS-stimulated secretion of the cytokines, tumor necrosis factor-α (TNF-α), interleukin-1β (IL-1β), interleukin-6 (IL-6), interleukin-8 (IL-8) and monocyte chemotactic protein-1 (MCP-1) was assessed by enzyme immunoassay on Days 2 and 7 of cultivation. The level of basal secretion of TNF-α, IL-1β, IL-6, MCP-1 was higher in patients with CAD compared to patients in the control group. The levels of re-stimulated TNF-α secretion and the levels of LPS-stimulated and re-stimulated IL-1β secretion on the second and sixth days were also higher in patients with CAD. LPS-stimulated MCP-1 secretion on the second day did not differ in patients of both groups, but re-stimulated MCP-1 secretion was higher in patients with CAD. The results of logistic regression analysis showed that the basal secretion levels of IL-1β and IL-6 were independently associated with premature CAD, along with smoking, body mass index and serum HDL-cholesterol levels.
Collapse
Affiliation(s)
- Tatiana Blokhina
- Department of problems of atherosclerosis, Chazov National Medical Research Center of Cardiology of the Ministry of Health of the Russian Federation, 121552 Moscow, Russia
| | - Tatiana Kirichenko
- Laboratory of medical genetics, Chazov National Medical Research Center of Cardiology of the Ministry of Health of the Russian Federation, 121552 Moscow, Russia
- Laboratory of cellular and molecular pathology of cardiovascular system, State Scientific Center of the Russian Federation Petrovsky National Research Center of Surgery, Moscow 119991, Russia
| | - Yuliya Markina
- Laboratory of cellular and molecular pathology of cardiovascular system, State Scientific Center of the Russian Federation Petrovsky National Research Center of Surgery, Moscow 119991, Russia
| | - Ulyana Khovantseva
- Laboratory of cellular and molecular pathology of cardiovascular system, State Scientific Center of the Russian Federation Petrovsky National Research Center of Surgery, Moscow 119991, Russia
| | - Ivan Melnikov
- Laboratory of cell hemostasis, Chazov National Medical Research Center of Cardiology of the Ministry of Health of the Russian Federation, 121552 Moscow, Russia
- Laboratory of Gas Exchange, Biomechanics and Barophysiology, State Scientific Center of the Russian Federation, The Institute of Biomedical Problems of the Russian Academy of Sciences, Moscow 123007, Russia
| | - Olga Guseva
- Laboratory of cell hemostasis, Chazov National Medical Research Center of Cardiology of the Ministry of Health of the Russian Federation, 121552 Moscow, Russia
| | - Sergey Bazanovich
- Laboratory of stem cells, Chazov National Medical Research Center of Cardiology of the Ministry of Health of the Russian Federation, 121552 Moscow, Russia
| | - Sergey Kozlov
- Department of problems of atherosclerosis, Chazov National Medical Research Center of Cardiology of the Ministry of Health of the Russian Federation, 121552 Moscow, Russia
| | - Alexander Orekhov
- Laboratory of cellular and molecular pathology of cardiovascular system, State Scientific Center of the Russian Federation Petrovsky National Research Center of Surgery, Moscow 119991, Russia
| |
Collapse
|
15
|
Azami P, Mohammadzadeh S, Seirafi S, Razeghian-Jahromi I. A review of cutting-edge biomarkers for diagnosing coronary artery disease. Medicine (Baltimore) 2025; 104:e41377. [PMID: 39854741 PMCID: PMC11771658 DOI: 10.1097/md.0000000000041377] [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: 07/06/2024] [Revised: 12/23/2024] [Accepted: 01/10/2025] [Indexed: 01/26/2025] Open
Abstract
Chronic coronary artery disease (CAD) remains a significant global healthcare burden. Current risk assessment methods have notable limitations in early detection and risk stratification. Hence, there is an urgent need for innovative biomarkers that facilitate the premature CAD diagnosis, ultimately leading to reduction in associated morbidity and mortality rates. This review comprehensively examines recent advances in emerging biomarkers for CAD detection. Our analysis delves into various aspects of these biomarkers such as their mechanisms of action, roles in the pathophysiology of the disease, and different measurement techniques employed in clinical practice. Comparative assessment of biomarker performance between CAD patients and control groups was also presented relying on their sensitivity, specificity, and area under the curve at specific cutoff points. In this regard, prominent biomarkers including Tenascin-C, IL-37, PTX3, transthyretin, soluble interleukin-6 receptor α, and miR-15a are identified as having high diagnostic potential for chronic CAD that indeed showcase promising performance metrics. These findings underscore the role of novel biomarkers in enhancing CAD risk stratification and improving patient outcomes through early intervention. However, the pursuit of an ideal and inclusive biomarker continues due to the multifaceted nature of CAD. Future randomized controlled trials are essential to bridge the gap between research findings and clinical practice in order to augment the practical application of these biomarkers in routine healthcare settings.
Collapse
Affiliation(s)
- Pouria Azami
- Cardiovascular Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | | | - Soroush Seirafi
- Department of Cardiology, Shiraz University of Medical Sciences, Shiraz, Iran
| | | |
Collapse
|
16
|
Wang L, Wang Y, Jiao T, Xu L, Ji E, Tapu SR, Liu Y, Li J. Effects of continuous positive airway pressure treatment on arterial stiffness and inflammatory factors in patients with coronary heart disease complicated with obstructive sleep apnea. J Cardiothorac Surg 2025; 20:59. [PMID: 39799348 PMCID: PMC11724610 DOI: 10.1186/s13019-024-03252-2] [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: 09/09/2024] [Accepted: 12/24/2024] [Indexed: 01/15/2025] Open
Abstract
BACKGROUND Continuous Positive Airway Pressure (CPAP) treatment brings more benefits than risks to most coronary heart disease (CHD) patients with obstructive sleep apnea (OSA). However, the pathophysiological mechanism by which CPAP treatment improves the prognosis of patients with CHD and OSA remains unclear. The purpose of this study was to clarify whether CPAP can improve arterial stiffness and inflammatory factor levels in CHD patients with OSA, and to further improve prognosis. METHOD 59 patients with coronary heart disease complicated by moderate to severe sleep apnea were divided into a CPAP treatment group (CPAP + coronary heart disease standard treatment) and a control group (only coronary heart disease standard treatment). Peripheral blood test reports were collected and pulse wave velocity (PWV) measurements were performed for each patient at the beginning, 3 months, and 6 months of treatment. RESULTS After 6 months of treatment, the CPAP group showed more significant improvement in the levels of inflammatory factors such as white blood cell (WBC), neutrophil (N), C-reactive protein (CRP), interleukin-6 (IL-6), procalcitonin (PCT), and PWV than the control group. CONCLUSION After active treatment with CPAP, arterial stiffness and inflammatory cytokine levels in patients with coronary heart disease and OSA improved. This association should be given more attention in clinical practice, and sleep apnea should be actively treated.
Collapse
Affiliation(s)
- Liang Wang
- Department of Cardiology, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Yuanqi Wang
- School of Medicine, Tongji University, Shanghai, China
| | - Tiantian Jiao
- School of Medicine, Tongji University, Shanghai, China
| | - Linghao Xu
- Department of Cardiology, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Endong Ji
- Department of Emergency and Critical Care, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | | | - Yehong Liu
- Department of Cardiology, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Jiming Li
- Department of Cardiology, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, China.
- Department of Cardiology, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, 200092, China.
| |
Collapse
|
17
|
Moriyama M, Murakawa Y, Kondo M, Taira M, Sumita Y, Honda M, Ichinose K. Tocilizumab Increases Serum Lipids but Does Not Increase Arteriosclerosis, As Measured by Intima-Media Thickness, in Patients With Rheumatoid Arthritis. Cureus 2025; 17:e76782. [PMID: 39897309 PMCID: PMC11786537 DOI: 10.7759/cureus.76782] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/01/2025] [Indexed: 02/04/2025] Open
Abstract
Objectives Patients with rheumatoid arthritis (RA) have a high incidence of arteriosclerotic disease. These are partly attributed to high levels of C-reactive protein (CRP) and inflammatory cytokines, such as tumor necrosis factor-alpha (TNF-α) and interleukin (IL)-6. Tocilizumab (TCZ) is an IL-6 receptor antagonist that lowers CRP levels by directly blocking IL-6 signaling. Tocilizumab has been reported to increase serum lipid levels. However, its effect on arteriosclerosis remains unclear. Therefore, we investigated the effect of TCZ on arteriosclerosis in patients with RA. Methods Eighteen Japanese patients with RA who were administered TCZ were included and assessed at baseline and six and 12 months. The Disease Activity Score-28 with erythrocyte sedimentation rate (DAS28-ESR), CRP, total cholesterol (TC), low-density lipoprotein cholesterol (LDL-C), high-density lipoprotein cholesterol (HDL-C), and carotid intima-media thickness (IMT) were measured and analyzed using high-resolution B-mode ultrasonography. Additionally, the effects of concurrent statin administration on any changes in IMT were evaluated. Results From baseline to six or 12 months, TCZ decreased CRP (P < 0.0005) and DAS28-ESR (P < 0.0005) significantly, whereas TC, LDL-C, and HDL-C levels increased significantly (P < 0.005). However, there was no change in the LDL-C/HDL-C ratio (P = 0.821 at six months and P = 0.168 at 12 months), and carotid IMT (P = 0.6874 at six months and P = 0.6951 at 12 months). Comparison of percentage changes in mean IMT revealed no statistical differences between the patient groups with or without statin administration (P = 0.7208 at six months, P = 0.5928 at 12 months). Conclusions According to the 12-month observation data, no significant association was detected between TCZ use and IMT changes in patients with RA, despite its effects on serum lipids. Further long-term studies are needed to confirm that IL-6 receptor blockers have cardiovascular effects.
Collapse
Affiliation(s)
- Mayuko Moriyama
- Department of Rheumatology, Shimane University Faculty of Medicine, Izumo, JPN
| | - Yohko Murakawa
- Department of Rheumatology, Shimane University Faculty of Medicine, Izumo, JPN
- Department of Internal Medicine III, Shimane University Faculty of Medicine, Izumo, JPN
| | - Masahiro Kondo
- Center of Community Medical Policy, Shimane University Faculty of Medicine, Izumo, JPN
- Department of Rheumatology, Shimane University Faculty of Medicine, Izumo, JPN
| | - Mariko Taira
- Department of Rheumatology, Shimene University Faculty of Medicine, Izumo, JPN
| | - Yoshiko Sumita
- Department of Rheumatology, Shimane University Faculty of Medicine, Izumo, JPN
- Department of Internal Medicine, Izumo Citizens' Rehabilitation Hospital, Izumo, JPN
| | - Manabu Honda
- Department of Rheumatology, Shimane University Faculty of Medicine, Izumo, JPN
| | - Kunihiro Ichinose
- Department of Rheumatology, Shimane University Faculty of Medicine, Izumo, JPN
| |
Collapse
|
18
|
Myrmel GMS, Wereski R, Karaji I, Saeed N, Aakre KM, Mills NL, Pedersen ER. Cardiac biomarkers and CT coronary angiography for the assessment of coronary heart disease. Clin Biochem 2025; 135:110857. [PMID: 39586418 DOI: 10.1016/j.clinbiochem.2024.110857] [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: 09/23/2024] [Revised: 10/23/2024] [Accepted: 11/20/2024] [Indexed: 11/27/2024]
Abstract
Over the last 30 years, the widespread use of cardiac biomarkers has transformed the diagnostic evaluation of patients with coronary heart disease. Cardiac troponin is integral to the definition of acute myocardial infarction. High-sensitivity cardiac troponin (hs-cTn) assays can improve risk stratification to facilitate both the rapid rule out of myocardial infarction and prediction of future cardiovascular events. Numerous circulating biomarkers representing different pathological pathways improve prediction of atherosclerotic cardiovascular disease (ACVD) and coronary artery disease (CAD). In parallel, coronary computed tomography angiography (CCTA) has become the most widely used imaging modality for the evaluation of patients with possible angina. CCTA now allows for the quantification of coronary calcification, atherosclerotic plaque volume and different plaque characteristics, enabling the identification high-risk features and inflammation. In the future, the use of CCTA is likely to extend to risk stratification for the prevention of ACVD. As such, how to integrate these diagnostic and prognostic circulating and imaging biomarkers is a topic of considerable interest. This review aims to describe current status and future possibilities for the integration of CCTA and cardiac biomarker testing to improve the identification and treatment of individuals with coronary heart disease and heightened cardiovascular risk.
Collapse
Affiliation(s)
| | - Ryan Wereski
- British Heart Foundation Centre for Cardiovascular Science, University of Edinburgh, Edinburgh, United Kingdom
| | - Iman Karaji
- Department of Heart Disease, Haukeland University Hospital, Bergen, Norway
| | - Nasir Saeed
- Department of Clinical Science, University of Bergen, Bergen, Norway
| | - Kristin Moberg Aakre
- Department of Heart Disease, Haukeland University Hospital, Bergen, Norway; Department of Clinical Science, University of Bergen, Bergen, Norway; Department of Medical Biochemistry and Pharmacology, Haukeland University Hospital, Bergen, Norway
| | - Nicholas L Mills
- British Heart Foundation Centre for Cardiovascular Science, University of Edinburgh, Edinburgh, United Kingdom; Usher Institute, University of Edinburgh, Edinburgh, United Kingdom
| | - Eva Ringdal Pedersen
- Department of Heart Disease, Haukeland University Hospital, Bergen, Norway; Department of Clinical Science, University of Bergen, Bergen, Norway
| |
Collapse
|
19
|
Celeski M, Segreti A, Crisci F, Cricco R, Piscione M, Di Gioia G, Nusca A, Fossati C, Pigozzi F, Ussia GP, Solaro RJ, Grigioni F. The Role of Cardiac Troponin and Other Emerging Biomarkers Among Athletes and Beyond: Underlying Mechanisms, Differential Diagnosis, and Guide for Interpretation. Biomolecules 2024; 14:1630. [PMID: 39766337 PMCID: PMC11727179 DOI: 10.3390/biom14121630] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2024] [Revised: 12/11/2024] [Accepted: 12/17/2024] [Indexed: 01/15/2025] Open
Abstract
Cardiovascular (CV) disease remains the leading cause of morbidity and mortality worldwide, highlighting the necessity of understanding its underlying molecular and pathophysiological pathways. Conversely, physical activity (PA) and exercise are key strategies in reducing CV event risks. Detecting latent CV conditions in apparently healthy individuals, such as athletes, presents a unique challenge. The early identification and treatment of CV disorders are vital for long-term health and patient survival. Cardiac troponin is currently the most commonly used biomarker for assessing CV changes in both athletes and the general population. However, there remains considerable debate surrounding the mechanisms underlying exercise-induced troponin elevations and its release in non-ischemic contexts. Thus, there is a pressing need to identify and implement more sensitive and specific biomarkers for CV disorders in clinical practice. Indeed, research continues to explore reliable biomarkers for evaluating the health of athletes and the effectiveness of physical exercise. It is essential to analyze current evidence on troponin release in non-ischemic conditions, post-strenuous exercise, and the complex biological pathways that influence its detection. Furthermore, this study summarizes current research on cytokines and exosomes, including their physiological roles and their relevance in various CV conditions, especially in athletes. In addition, this paper gives special attention to underlying mechanisms, potential biomarkers, and future perspectives.
Collapse
Affiliation(s)
- Mihail Celeski
- Fondazione Policlinico Universitario Campus Bio-Medico, Via Alvaro del Portillo, 200, 00128 Roma, Italy (R.C.)
- Unit of Cardiovascular Sciences, Department of Medicine and Surgery, Università Campus Bio-Medico di Roma, Via Alvaro del Portillo, 21, 00128 Roma, Italy
| | - Andrea Segreti
- Fondazione Policlinico Universitario Campus Bio-Medico, Via Alvaro del Portillo, 200, 00128 Roma, Italy (R.C.)
- Unit of Cardiovascular Sciences, Department of Medicine and Surgery, Università Campus Bio-Medico di Roma, Via Alvaro del Portillo, 21, 00128 Roma, Italy
- Department of Movement, Human and Health Sciences, University of Rome “Foro Italico”, Piazza Lauro de Bosis 6, 00135 Roma, Italy
| | - Filippo Crisci
- Fondazione Policlinico Universitario Campus Bio-Medico, Via Alvaro del Portillo, 200, 00128 Roma, Italy (R.C.)
- Unit of Cardiovascular Sciences, Department of Medicine and Surgery, Università Campus Bio-Medico di Roma, Via Alvaro del Portillo, 21, 00128 Roma, Italy
| | - Riccardo Cricco
- Fondazione Policlinico Universitario Campus Bio-Medico, Via Alvaro del Portillo, 200, 00128 Roma, Italy (R.C.)
- Unit of Cardiovascular Sciences, Department of Medicine and Surgery, Università Campus Bio-Medico di Roma, Via Alvaro del Portillo, 21, 00128 Roma, Italy
| | - Mariagrazia Piscione
- Fondazione Policlinico Universitario Campus Bio-Medico, Via Alvaro del Portillo, 200, 00128 Roma, Italy (R.C.)
- Unit of Cardiovascular Sciences, Department of Medicine and Surgery, Università Campus Bio-Medico di Roma, Via Alvaro del Portillo, 21, 00128 Roma, Italy
| | - Giuseppe Di Gioia
- Department of Movement, Human and Health Sciences, University of Rome “Foro Italico”, Piazza Lauro de Bosis 6, 00135 Roma, Italy
- Institute of Sports Medicine and Science, Italian National Olympic Committee, Largo Piero Gabrielli 1, 00197 Roma, Italy
| | - Annunziata Nusca
- Fondazione Policlinico Universitario Campus Bio-Medico, Via Alvaro del Portillo, 200, 00128 Roma, Italy (R.C.)
- Unit of Cardiovascular Sciences, Department of Medicine and Surgery, Università Campus Bio-Medico di Roma, Via Alvaro del Portillo, 21, 00128 Roma, Italy
| | - Chiara Fossati
- Department of Movement, Human and Health Sciences, University of Rome “Foro Italico”, Piazza Lauro de Bosis 6, 00135 Roma, Italy
| | - Fabio Pigozzi
- Department of Movement, Human and Health Sciences, University of Rome “Foro Italico”, Piazza Lauro de Bosis 6, 00135 Roma, Italy
| | - Gian Paolo Ussia
- Fondazione Policlinico Universitario Campus Bio-Medico, Via Alvaro del Portillo, 200, 00128 Roma, Italy (R.C.)
- Unit of Cardiovascular Sciences, Department of Medicine and Surgery, Università Campus Bio-Medico di Roma, Via Alvaro del Portillo, 21, 00128 Roma, Italy
| | - Ross John Solaro
- Department of Physiology and Biophysics and Center for Cardiovascular Research, College of Medicine, University of Illinois at Chicago, Chicago, IL 60612, USA;
| | - Francesco Grigioni
- Fondazione Policlinico Universitario Campus Bio-Medico, Via Alvaro del Portillo, 200, 00128 Roma, Italy (R.C.)
- Unit of Cardiovascular Sciences, Department of Medicine and Surgery, Università Campus Bio-Medico di Roma, Via Alvaro del Portillo, 21, 00128 Roma, Italy
| |
Collapse
|
20
|
Wang J, Kockx M, Pennings GJ, Lambert T, Chow V, Kritharides L. Discordance Between Triglycerides, Remnant Cholesterol and Systemic Inflammation in Patients with Schizophrenia. Biomedicines 2024; 12:2884. [PMID: 39767790 PMCID: PMC11673878 DOI: 10.3390/biomedicines12122884] [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: 11/27/2024] [Revised: 12/13/2024] [Accepted: 12/16/2024] [Indexed: 01/11/2025] Open
Abstract
BACKGROUND/OBJECTIVES Hypertriglyceridaemia and systemic inflammation are prevalent in patients with schizophrenia and contribute to an increased risk of cardiovascular disease. Although elevated triglycerides (TGs) and remnant cholesterol are linked to inflammation in the general population and individuals with metabolic syndrome, whether they are associated in patients with schizophrenia remains unclear. METHODS Fasting levels of TG, cholesterol (total cholesterol (TC), low-density lipoprotein cholesterol (LDL-C), high-density lipoprotein cholesterol (HDL-C) and remnant cholesterol)), and markers of systemic inflammation including high-sensitivity C-reactive protein (hsCRP), leukocyte counts and their differentials (neutrophils, monocytes and lymphocytes) were determined in 147 patients diagnosed with schizophrenia on long-term antipsychotic regimens and compared with 56 age- and sex-matched healthy controls. Apolipoprotein B and glycosylation of acute phase reactant (GlycA) signatures were assessed by NMR. Circulating cytokine levels were measured by a cytokine/chemokine multiplex assay. RESULTS Patients with schizophrenia had markedly elevated TG and remnant cholesterol relative to controls and had evidence of systemic inflammation with increased circulating hsCRP, GlycA, leukocyte, neutrophil counts and neutrophil-to-lymphocyte ratio (NLR). Unexpectedly TG and remnant cholesterol did not correlate with systemic inflammatory markers in patients with schizophrenia, and differences in inflammatory markers between controls and patients persisted after adjusting for the lipid profile. Interleukin (IL)-10 levels were increased in patients with schizophrenia, suggesting an anti-inflammatory signature. CONCLUSIONS The discordance between TG, remnant cholesterol and systemic inflammation in patients with schizophrenia suggests these are likely independent contributors to cardiovascular risk in this population.
Collapse
Affiliation(s)
- Jeffrey Wang
- Atherosclerosis and Vascular Biology Laboratory, The ANZAC Research Institute, Concord Repatriation General Hospital, University of Sydney, Concord 2138, Australia; (J.W.); (G.J.P.); (L.K.)
| | - Maaike Kockx
- Atherosclerosis and Vascular Biology Laboratory, The ANZAC Research Institute, Concord Repatriation General Hospital, University of Sydney, Concord 2138, Australia; (J.W.); (G.J.P.); (L.K.)
| | - Gabrielle J. Pennings
- Atherosclerosis and Vascular Biology Laboratory, The ANZAC Research Institute, Concord Repatriation General Hospital, University of Sydney, Concord 2138, Australia; (J.W.); (G.J.P.); (L.K.)
| | - Tim Lambert
- Concord Clinical School, Faculty of Medicine and Health, University of Sydney, Camperdown 2050, Australia; (T.L.); (V.C.)
- Collaborative Centre for Cardiometabolic Health, Charles Perkins Centre, University of Sydney, Camperdown 2050, Australia
| | - Vincent Chow
- Concord Clinical School, Faculty of Medicine and Health, University of Sydney, Camperdown 2050, Australia; (T.L.); (V.C.)
- Collaborative Centre for Cardiometabolic Health, Charles Perkins Centre, University of Sydney, Camperdown 2050, Australia
- Department of Cardiology, Concord Repatriation General Hospital, Sydney Local Health District, Concord 2138, Australia
| | - Leonard Kritharides
- Atherosclerosis and Vascular Biology Laboratory, The ANZAC Research Institute, Concord Repatriation General Hospital, University of Sydney, Concord 2138, Australia; (J.W.); (G.J.P.); (L.K.)
- Concord Clinical School, Faculty of Medicine and Health, University of Sydney, Camperdown 2050, Australia; (T.L.); (V.C.)
- Department of Cardiology, Concord Repatriation General Hospital, Sydney Local Health District, Concord 2138, Australia
| |
Collapse
|
21
|
Salman M, Cicin J, Abdul Jabbar AB, El-shaer A, Tauseef A, Asghar N, Mirza M, Aboeata A. Trends in sepsis-associated cardiovascular disease mortality in the United States, 1999 to 2022. Front Cardiovasc Med 2024; 11:1505905. [PMID: 39717445 PMCID: PMC11663846 DOI: 10.3389/fcvm.2024.1505905] [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: 10/03/2024] [Accepted: 11/19/2024] [Indexed: 12/25/2024] Open
Abstract
Purpose Cardiovascular disease (CVD) is the leading cause of death in the United States, and sepsis significantly contributes to hospitalization and mortality. This study aims to assess the trends of sepsis-associated CVD mortality rates and variations in mortality based on demographics and regions in the US. Methods The Centers for Disease Control and Prevention Wide-ranging Online Data for Epidemiologic Research (CDC WONDER) database was used to identify CVD and sepsis-related deaths from 1999 to 2022. Data on gender, race and ethnicity, age groups, region, and state classification were statistically analyzed to obtain crude and age-adjusted mortality rates (AAMR). The Joinpoint Regression Program was used to determine trends in mortality within the study period. Results During the study period, there were a total of 1,842,641 deaths with both CVD and sepsis listed as a cause of death. Sepsis-associated CVD mortality decreased between 1999 and 2013, from AAMR of 65.7 in 1999 to 58.8 in 2013 (APC -1.06*%, 95% CI: -2.12% to -0.26%), then rose to 74.3 in 2022 (APC 3.23*%, 95% CI: 2.18%-5.40%). Throughout the study period, mortality rates were highest in men, NH Black adults, and elderly adults (65+ years old). The Northeast region, which had the highest mortality rate in the initial part of the study period, was the only region to see a decline in mortality, while the Northwest, Midwest, and Southern regions experienced significant increases in mortality rates. Conclusion Sepsis-associated CVD mortality has increased in the US over the past decade, and both this general trend and the demographic disparities have worsened since the onset of the COVID-19 pandemic.
Collapse
Affiliation(s)
- Malik Salman
- School of Medicine, Creighton University, Omaha, NE, United States
| | - Jack Cicin
- School of Medicine, Creighton University, Omaha, NE, United States
| | | | - Ahmed El-shaer
- School of Medicine, Creighton University, Omaha, NE, United States
| | - Abubakar Tauseef
- School of Medicine, Creighton University, Omaha, NE, United States
| | - Noureen Asghar
- School of Medicine, Creighton University, Omaha, NE, United States
| | - Mohsin Mirza
- School of Medicine, Creighton University, Omaha, NE, United States
| | - Ahmed Aboeata
- Department of Cardiology, Creighton University Medical Center, Omaha, NE, United States
| |
Collapse
|
22
|
Amin T, Rasool MHU, Ozkan BI, Swaminathan G, Rauf F, Patrizi S, Sethi A, Frishman WH, Aronow WS, Ahmed MS. Leukocytosis as a Risk Factor for Coronary Artery Disease: Pathophysiology and Epidemiology. Cardiol Rev 2024. [DOI: 10.1097/crd.0000000000000824] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/11/2025]
Abstract
Coronary artery disease (CAD) is a significant health concern characterized by reduced blood flow to the heart muscle, primarily due to the buildup of atherosclerotic plaques in the coronary arteries. This process begins with endothelial injury, leading to a cascade of biological responses contributing to plaque formation. Endothelial injury attracts the migration of monocytes which differentiate into macrophages upon uptake of oxidized low-density lipoproteins, changing into lipid-laden macrophage or “foam cells.” The process of plaque formation is influenced by many factors which have been studied extensively in literature such as smoking, hypertension, and diabetes mellitus. Chronic inflammatory illnesses are often associated with a high prevalence of coronary artery syndromes, prompting the evaluation of markers of inflammation such as white blood cell count and inflammatory markers as independent risk factors for CAD. White blood cells play a remarkable role in the pathophysiology of disease formation and progression. The article below aims to discuss the pathophysiology and epidemiology of leukocytosis as a risk factor for CAD.
Collapse
Affiliation(s)
- Toka Amin
- Department of Medicine, Icahn School of Medicine at Mount Sinai Medical Center, New York, NY
| | | | - Bike Ilyada Ozkan
- Department of Medicine, Icahn School of Medicine at Mount Sinai Medical Center, New York, NY
| | - Gowri Swaminathan
- Department of Medicine, Icahn School of Medicine at Mount Sinai Medical Center, New York, NY
| | - Faateh Rauf
- Department of Medicine, Icahn School of Medicine at Mount Sinai Medical Center, New York, NY
| | - Santino Patrizi
- Department of Medicine, Icahn School of Medicine at Mount Sinai Medical Center, New York, NY
| | - Arshia Sethi
- Department of Medicine, Icahn School of Medicine at Mount Sinai Medical Center, New York, NY
| | | | - Wilbert S. Aronow
- Departments of Cardiology and Medicine, Westchester Medical Center and New York Medical College, Valhalla, NY
| | - Mahmoud Samy Ahmed
- Departments of Cardiology and Medicine, Westchester Medical Center and New York Medical College, Valhalla, NY
| |
Collapse
|
23
|
Arosio B, Picca A. The biological roots of the sex-frailty paradox. Exp Gerontol 2024; 198:112619. [PMID: 39490699 DOI: 10.1016/j.exger.2024.112619] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2024] [Revised: 10/09/2024] [Accepted: 10/23/2024] [Indexed: 11/05/2024]
Abstract
Aging is a dynamic process that requires a continuous response and adaptation to internal and external stimuli over the life course. This eventually results in people aging differently and women aging differently than men. The "gender paradox" describes how women experience greater longevity than men, although linked with higher rates of disability and poor health status. Recently, the concept of frailty has been incorporated into this paradox giving rise to the "sex-frailty paradox" which describes how women are frailer because they manifest worse health status but, at the same time, appear less susceptible to death than men of the same age. However, very little is known about the biological roots of this sex-related difference in frailty. Inflamm-aging, the chronic low-grade inflammatory state associated with age, plays a key pathophysiological role in several age-related diseases/conditions, including Alzheimer's disease (AD), for which women have a higher lifetime risk than men. Interestingly, inflamm-aging develops at a different rate in women compared to men, with features that could play a critical role in the development of AD in women. According to this view, a continuum between aging and age-related diseases that probably lacks clear boundaries can be envisioned in which several shared biological mechanisms that progress at different pace may lead to different aging trajectories in women than in men. It, therefore, becomes urgent to consider a holistic approach in the study of aging, and decline it from a gender medicine perspective also considering the biological roots of the sex-frailty paradox.
Collapse
Affiliation(s)
- Beatrice Arosio
- Department of Clinical Sciences and Community Health, University of Milan, Milan, Italy.
| | - Anna Picca
- Department of Medicine and Surgery, LUM University, Casamassima, Italy; Fondazione Policlinico Universitario "A. Gemelli" IRCCS, Rome, Italy
| |
Collapse
|
24
|
Zhang K, Li Y, Lin X, Daneshar M, Karamian F, Li M. Effect of cinnamon supplementation on blood pressure, oxidative stress, and inflammatory biomarkers in adults: An umbrella review of the meta-analyses of randomized controlled trials. Nutr Metab Cardiovasc Dis 2024; 34:2659-2668. [PMID: 39299867 DOI: 10.1016/j.numecd.2024.08.009] [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: 04/29/2024] [Revised: 08/08/2024] [Accepted: 08/19/2024] [Indexed: 09/22/2024]
Abstract
AIMS Cinnamon has positive effects on markers of cardiometabolic health, including blood pressure (BP), oxidative stress, and inflammation. Despite the evidence accumulated from meta-analysis studies on the effects of cinnamon on these markers, the reported findings are still controversial. This umbrella review was conducted to evaluate the evidence and provide a definitive clarification. DATA SYNTHESIS We conducted a systematic search in four scientific databases, including PubMed, Scopus, Web of Science, and Embase electronic databases, up to March 2024 to identify systematic reviews and meta-analyses of randomized clinical trials investigating the impact of cinnamon on blood pressure, oxidative stress, and inflammation. The findings revealed that cinnamon might exert favorable effects on systolic blood pressure (SBP) (ES = -2.36 mmHg; 95% CI: 3.86, -1.40), diastolic blood pressure (DBP) (ES = -1.65 mmHg; 95% CI: 2.41, -0.90), total antioxidant capacity (TAC) (WMD = 0.34; 95% CI: 0.04, 0.64), and interleukin-6 (IL-6) (WMD = -1.48; 95% CI: 2.96, -0.01). However, the results did not show any significant effect of cinnamon on malondialdehyde (MDA) (WMD = -0.47; 95% CI: 0.99, 0.05), C-reactive protein (CRP) (WMD = -1.33; 95% CI: 2.66, 0.00), and intercellular adhesion molecule 1 (ICAM-1) (WMD= 1.53, 95% CI: 12.03, 15.10). CONCLUSIONS The results of the studies included in this umbrella review support the usefulness of cinnamon consumption in modulating BP as well as improving TAC and IL-6 in metabolic disorders. Due to the limited number of studies, clinical diversity, and other limitations, more high-quality studies must be conducted to provide more precise and comprehensive recommendations. REGISTRATION NUMBER PROSPERO, CRD42023487350.
Collapse
Affiliation(s)
- Kaitian Zhang
- Department of Cardiovascular Surgery, Gaozhou People's Hospital, Gaozhou, 525200, China
| | - Yang Li
- Department of Cardiovascular Medicine, First Affiliated Hospital of Baotou Medical College, Baotou, Inner Mongolia, 014010, China
| | - Xuefeng Lin
- Department of Cardiovascular Medicine, First Affiliated Hospital of Baotou Medical College, Baotou, Inner Mongolia, 014010, China
| | - Mazar Daneshar
- Science and Research Branch, School of Nutrition, Islamic Azad University, Tehran, Iran
| | - Fatemeh Karamian
- Science and Research Branch, School of Nutrition, Islamic Azad University, Tehran, Iran
| | - Mingzhu Li
- Department of Pharmacy, Gaozhou People's Hospital, Gaozhou, 525200, China.
| |
Collapse
|
25
|
Kose MP, Piskinpasa ME, Hacioglu Y, Karabag T. The relationship of visceral adiposity with endothelial functions and subclinical atherosclerosis in obese individuals. ROMANIAN JOURNAL OF INTERNAL MEDICINE = REVUE ROUMAINE DE MEDECINE INTERNE 2024; 62:404-413. [PMID: 38981454 DOI: 10.2478/rjim-2024-0024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/31/2024] [Indexed: 07/11/2024]
Abstract
INTRODUCTION On the cardiovascular system, obesity accelerates atherosclerosis progression, inducing pathophysiological changes that are detectable already from young adults. Endothelial dysfunction is one of the earliest vascular alterations observed in obesity. In this study, we aimed to determine endothelial functions and carotid intima-media thickness in patients with obesity without overt cardiovascular disease. METHODS The study was conducted with 112 individuals with obesity without overt cardiovascular disease and any chronical diseases (BMI>30 kg/m2) (84 female, 28 male, mean age: 46.3±11.2 years) and 49 healthy individuals with no diseases (33 female, 16 male, mean age: 44.6±10.2 years). All patients were examined for endothelial functions by the flow-mediated dilatation (FMD) method and carotid intima-media thicknesses (CIMT). All measurements were performed by the same imaging specialist, averaging 3 different measurements. In addition to the body mass index and waist circumference visceral adiposity index (VAI) and triponderal mass index (TPI) also calculated. RESULTS The percentage of FMD obtained by brachial artery ultrasound was significantly lower, visceral adipose tissue, perirenal adipose tissue thicknesses measured by abdominal ultrasound and CIMT were significantly thicker in Group 1 compared to Group 2. FMD had a negative significant correlation with body mass index, visceral adipose tissue thickness, perirenal adipose tissue thickness, and waist and hip circumferences, and carotid intima-media thickness and CIMT had a significant correlation with visceral adipose tissue thickness, perirenal adipose tissue thickness, VAI, TPI and waist, hip circumferences. CONCLUSION Individuals with obesity have impaired endothelial functions and greater carotid intima-media thicknesses compared to healthy individuals. This impairment in endothelial functions is proportional to the amount of visceral and perirenal fat accumulation. Parameterss reflecting visceral fat distribution such as VAI and TPI are also related with these impairment.
Collapse
Affiliation(s)
- Merve Polat Kose
- Saglik Bilimleri University, Istanbul Education and Research Hospital, Department of Internal Medicine
| | - Mehmet Emin Piskinpasa
- Saglik Bilimleri University, Istanbul Education and Research Hospital, Department of Internal Medicine
| | - Yalcin Hacioglu
- Saglik Bilimleri University, Istanbul Education and Research Hospital, Department of Family Medicine
| | - Turgut Karabag
- Saglik Bilimleri University, Istanbul Education and Research Hospital, Department of Cardiology, Istanbul, Turkey
| |
Collapse
|
26
|
Liñares A, Dopico J, Blanco C, Pico A, Sobrino T, Blanco J, Leira Y. The systemic impact of non-surgical treatment of peri-implantitis with or without adjunctive systemic metronidazole: Secondary analysis of a randomized clinical trial. Clin Oral Implants Res 2024; 35:1519-1530. [PMID: 39093380 PMCID: PMC11629454 DOI: 10.1111/clr.14339] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2023] [Revised: 05/14/2024] [Accepted: 07/14/2024] [Indexed: 08/04/2024]
Abstract
OBJECTIVES The aim of this study was to evaluate the systemic effect of non-surgical peri-implantitis treatment (NSPIT) with or without the administration of systemic metronidazole. METHODS In this secondary analysis from a previously published clinical trial (NCT03564301), peri-implantitis patients were randomized into two groups: test, receiving NSPIT plus 500 mg of oral systemic metronidazole three times a day for 7 days (n = 10); and control group, receiving NSPIT plus placebo (n = 11). Serum samples were obtained at baseline, 3 and 6 months after therapy to determine levels of inflammatory biomarkers, lipid fractions and complete blood counts. RESULTS Both treatment modalities produced improvements in clinical and radiographic parameters. After 6 months from NSPIT, a substantial reduction in C-reactive protein (6.9 mg/dL; 95% CI: 3.7 to 9.9, p < .001) and low-density lipoprotein cholesterol (21.8 mg/dL; 95% CI: -6.9 to 50.5, p = .013) as well as a modest increase in neutrophils counts (0.4 × 103/μL; 95% CI: -0.4 to 1.1, p = .010) was observed in the control group while the test group showed a significant reduction of TNF-α (110.1; 95% CI: 38.9 to 181.4, p = .004). CONCLUSIONS NSPIT showed a short-term beneficial systemic effect regardless of adjunctive use of systemic metronidazole.
Collapse
Affiliation(s)
- Antonio Liñares
- Periodontology Unit, Faculty of OdontologyUniversity of Santiago de CompostelaSantiago de CompostelaSpain
| | - Jose Dopico
- Periodontology Unit, Faculty of OdontologyUniversity of Santiago de CompostelaSantiago de CompostelaSpain
| | - Carlota Blanco
- Periodontology Unit, Faculty of OdontologyUniversity of Santiago de CompostelaSantiago de CompostelaSpain
| | - Alex Pico
- Periodontology Unit, Faculty of OdontologyUniversity of Santiago de CompostelaSantiago de CompostelaSpain
| | - Tomás Sobrino
- NeuroAging Research Group, Clinical Neurosciences Research LaboratoriesHealth Research Institute of Santiago de CompostelaSantiago de CompostelaSpain
| | - Juan Blanco
- Periodontology Unit, Faculty of OdontologyUniversity of Santiago de CompostelaSantiago de CompostelaSpain
| | - Yago Leira
- Periodontology Unit, Faculty of OdontologyUniversity of Santiago de CompostelaSantiago de CompostelaSpain
- NeuroAging Research Group, Clinical Neurosciences Research LaboratoriesHealth Research Institute of Santiago de CompostelaSantiago de CompostelaSpain
| |
Collapse
|
27
|
Prasad K. Role of C-Reactive Protein, An Inflammatory Biomarker in The Development of Atherosclerosis and Its Treatment. Int J Angiol 2024; 33:271-281. [PMID: 39502349 PMCID: PMC11534478 DOI: 10.1055/s-0044-1788296] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2024] Open
Abstract
This article deals with the role of c-reactive protein (CRP) in the development of atherosclerosis and its treatment. CRP has a predictive value in ischemic heart disease, restenosis, coronary artery disease, aortic atherosclerosis, and cerebrovascular disease. This article deals with the synthesis and mechanism of CRP-induced atherosclerosis and its treatment. CRP increases the formation of numerous atherogenic biomolecules such as reactive oxygen species (ROS), cytokines (interleukin [IL]-1β and IL-6), cell adhesion molecules (intercellular adhesion molecule-1, vascular cell adhesion molecule-1, monocyte chemoattractant protein-1, activated complement C 5 , monocyte colony-stimulating factor, and numerous growth factors [insulin-like growth factor, platelet-derived growth factor, and transforming growth factor-β]). ROS mildly oxidizes low-density lipoprotein (LDL)-cholesterol to form minimally modified LDL which is further oxidized to form oxidized LDL. The above atherogenic biomolecules are involved in the development of atherosclerosis and has been described in detail in the text. This paper also deals with the treatment modalities for CRP-induced atherosclerosis which includes lipid-lowering drugs, antihypertensive drugs, antioxidants, aspirin, antidiabetic drugs, angiotensin-converting enzyme inhibitors, angiotensin II receptor blockers, regular physical activity, weight reduction, and stoppage of cigarette smoking. In conclusion, CRP induces atherosclerosis through increases in atherogenic biomolecules and the treatment modalities would prevent, regress, and slow the progression of CRP-induced atherosclerosis.
Collapse
Affiliation(s)
- Kailash Prasad
- Department of Physiology, College of Medicine, University of Saskatchewan, Saskatoon, Saskatchewan, Canada
| |
Collapse
|
28
|
Mehta NN, deGoma E, Shapiro MD. IL-6 and Cardiovascular Risk: A Narrative Review. Curr Atheroscler Rep 2024; 27:12. [PMID: 39589436 PMCID: PMC11599326 DOI: 10.1007/s11883-024-01259-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/04/2024] [Indexed: 11/27/2024]
Abstract
PURPOSE OF REVIEW The objective of this narrative review is to summarize data from recently published prospective observational studies that analyze the association between circulating interleukin-6 (IL-6) levels and cardiovascular clinical or imaging endpoints. RECENT FINDINGS Higher levels of IL-6 are associated with a higher risk of cardiovascular death, major adverse cardiovascular events, myocardial infarction, stroke, peripheral artery disease, and heart failure. Imaging studies have also shown an association between IL-6 and carotid intima-media thickness progression, carotid plaque progression, severity, and vulnerability. These observations have been consistent across a wide range of study populations and after adjusting for traditional and emerging risk factors including high-sensitivity C-reactive protein. Robust epidemiologic evidence supports IL-6 as a central mediator of cardiovascular risk along with human genetic studies and mechanistic experiments. Ongoing clinical studies are testing the therapeutic hypothesis of IL-6 inhibition in patients with atherosclerotic cardiovascular disease or heart failure.
Collapse
Affiliation(s)
- Nehal N Mehta
- The George Washington University School of Medicine, Washington, DC, USA
| | | | | |
Collapse
|
29
|
Napiórkowska-Baran K, Doligalska A, Drozd M, Czarnowska M, Łaszczych D, Dolina M, Szymczak B, Schmidt O, Bartuzi Z. Management of a Patient with Cardiovascular Disease Should Include Assessment of Primary and Secondary Immunodeficiencies: Part 2-Secondary Immunodeficiencies. Healthcare (Basel) 2024; 12:1977. [PMID: 39408157 PMCID: PMC11477378 DOI: 10.3390/healthcare12191977] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2024] [Revised: 09/19/2024] [Accepted: 09/24/2024] [Indexed: 10/20/2024] Open
Abstract
BACKGROUND Cardiovascular diseases are among the most common chronic diseases, generating high social and economic costs. Secondary immunodeficiencies occur more often than primary ones and may result from the co-occurrence of specific diseases, treatment, nutrient deficiencies and non-nutritive bio-active compounds that result from the industrial nutrient practices. OBJECTIVES The aim of this article is to present selected secondary immunodeficiencies and their impact on the cardiovascular system. RESULTS The treatment of a patient with cardiovascular disease should include an assess-ment for immunodeficiencies, because the immune and cardiovascular systems are closely linked. CONCLUSIONS Immune system dysfunctions can significantly affect the course of cardiovascular diseases and their treatment. For this reason, comprehensive care for a patient with cardiovascular disease requires taking into account potential immunodeficiencies, which can have a significant impact on the patient's health.
Collapse
Affiliation(s)
- Katarzyna Napiórkowska-Baran
- Department of Allergology, Clinical Immunology and Internal Diseases, Collegium Medicum Bydgoszcz, Nicolaus Copernicus University Torun, 85-067 Bydgoszcz, Poland;
| | - Agata Doligalska
- Student Research Club of Clinical Immunology, Department of Allergology, Clinical Immunology and Internal Diseases, Collegium Medicum Bydgoszcz, Nicolaus Copernicus University Torun, 85-067 Bydgoszcz, Poland; (A.D.); (M.D.); (M.C.); (D.Ł.); (M.D.); (B.S.); (O.S.)
| | - Magdalena Drozd
- Student Research Club of Clinical Immunology, Department of Allergology, Clinical Immunology and Internal Diseases, Collegium Medicum Bydgoszcz, Nicolaus Copernicus University Torun, 85-067 Bydgoszcz, Poland; (A.D.); (M.D.); (M.C.); (D.Ł.); (M.D.); (B.S.); (O.S.)
| | - Marta Czarnowska
- Student Research Club of Clinical Immunology, Department of Allergology, Clinical Immunology and Internal Diseases, Collegium Medicum Bydgoszcz, Nicolaus Copernicus University Torun, 85-067 Bydgoszcz, Poland; (A.D.); (M.D.); (M.C.); (D.Ł.); (M.D.); (B.S.); (O.S.)
| | - Dariusz Łaszczych
- Student Research Club of Clinical Immunology, Department of Allergology, Clinical Immunology and Internal Diseases, Collegium Medicum Bydgoszcz, Nicolaus Copernicus University Torun, 85-067 Bydgoszcz, Poland; (A.D.); (M.D.); (M.C.); (D.Ł.); (M.D.); (B.S.); (O.S.)
| | - Marcin Dolina
- Student Research Club of Clinical Immunology, Department of Allergology, Clinical Immunology and Internal Diseases, Collegium Medicum Bydgoszcz, Nicolaus Copernicus University Torun, 85-067 Bydgoszcz, Poland; (A.D.); (M.D.); (M.C.); (D.Ł.); (M.D.); (B.S.); (O.S.)
| | - Bartłomiej Szymczak
- Student Research Club of Clinical Immunology, Department of Allergology, Clinical Immunology and Internal Diseases, Collegium Medicum Bydgoszcz, Nicolaus Copernicus University Torun, 85-067 Bydgoszcz, Poland; (A.D.); (M.D.); (M.C.); (D.Ł.); (M.D.); (B.S.); (O.S.)
| | - Oskar Schmidt
- Student Research Club of Clinical Immunology, Department of Allergology, Clinical Immunology and Internal Diseases, Collegium Medicum Bydgoszcz, Nicolaus Copernicus University Torun, 85-067 Bydgoszcz, Poland; (A.D.); (M.D.); (M.C.); (D.Ł.); (M.D.); (B.S.); (O.S.)
| | - Zbigniew Bartuzi
- Department of Allergology, Clinical Immunology and Internal Diseases, Collegium Medicum Bydgoszcz, Nicolaus Copernicus University Torun, 85-067 Bydgoszcz, Poland;
| |
Collapse
|
30
|
Al-Rashed F, AlSaeed H, Almansour N, Al-Mulla F, Hannun YA, Ahmad R. IL-6R (trans-signaling) is a key regulator of reverse cholesterol transport in lipid-laden macrophages. Clin Immunol 2024; 267:110351. [PMID: 39216780 PMCID: PMC11402558 DOI: 10.1016/j.clim.2024.110351] [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: 08/14/2024] [Accepted: 08/21/2024] [Indexed: 09/04/2024]
Abstract
Atherosclerosis is a cardiovascular disease caused by cholesterol-laden arterial plaques. This study evaluated the correlation between interleukin-6 (IL-6), its receptors (IL6R/CD126), and glycoprotein 130 (gp130) alongside atherosclerosis biomarkers in a cohort of 142 subjects, equally divided between lean and obese individuals. Subsequent analyses used THP-1-derived macrophages to assess the biochemical impact of inhibiting IL-6 receptors. IL-6 secretion increased with atherosclerosis in obese subjects, while IL6R/CD126 and gp130 on monocytes decreased. Pharmacological gp130 inhibition altered lipid metabolism, increasing LDLR gene expression and cholesterol synthesis via SREBF2 and mevalonate kinase, along with HMG-CoA reductase at protein levels. gp130-deficient cells produced more cholesterol and had lower ABCA1 levels, suggesting hindered cholesterol efflux. Filipin III staining confirmed cholesterol retention in gp130-inhibited cells. Ex-vivo investigation on lean PBMCs further defined the impact of gp130 inhibition on the reduction of cholesterol efflux. Our results indicates gp130 is crucial for macrophage reverse cholesterol transport and may be a target for atherosclerosis treatments.
Collapse
Affiliation(s)
- Fatema Al-Rashed
- Immunology and Microbiology Department, Dasman Diabetes Institute, Al-Soor Street, Dasman, Kuwait, PO BOX 1180, Dasman 15462, Kuwait.
| | - Halemah AlSaeed
- Immunology and Microbiology Department, Dasman Diabetes Institute, Al-Soor Street, Dasman, Kuwait, PO BOX 1180, Dasman 15462, Kuwait
| | - Nourah Almansour
- Immunology and Microbiology Department, Dasman Diabetes Institute, Al-Soor Street, Dasman, Kuwait, PO BOX 1180, Dasman 15462, Kuwait
| | - Fahd Al-Mulla
- Genetics and Bioinformatics Department, Dasman Diabetes Institute, Kuwait, Kuwait
| | - Yusuf A Hannun
- Stony Brook Cancer Center, Stony Brook University, Stony Brook, NY 11794, USA
| | - Rasheed Ahmad
- Immunology and Microbiology Department, Dasman Diabetes Institute, Al-Soor Street, Dasman, Kuwait, PO BOX 1180, Dasman 15462, Kuwait
| |
Collapse
|
31
|
Cederström S, Jernberg T, Samnegård A, Johansson F, Silveira A, Tornvall P, Lundman P. Inflammatory biomarkers and long-term outcome in young patients three months after a first myocardial infarction. Cytokine 2024; 182:156696. [PMID: 39059290 DOI: 10.1016/j.cyto.2024.156696] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2024] [Revised: 06/18/2024] [Accepted: 07/12/2024] [Indexed: 07/28/2024]
Abstract
BACKGROUND Studies on predictive value of circulating inflammatory biomarkers after myocardial infarction (MI) have often been limited by blood sampling only in an acute setting and short follow-up time. We aimed to compare the long-term predictive value of nine inflammatory biomarkers, known to be involved in atherosclerosis, in young patients investigated three months after a first-time MI. METHODS Nine biomarkers (high-sensitivity C-reactive protein, interleukin (IL)-6, IL-18, monocyte chemoattractant protein-1, matrix metalloproteinase (MMP)-1, MMP-3, MMP-9, serum amyloid A and tumor necrosis factor-alfa) were sampled in 382 young (<60 years) patients and in age and sex-matched controls, three months after a first-time MI between 1996 and 2000. Swedish national patient registers were used to determine cardiovascular (CV) outcomes during 20 years of follow-up. RESULTS In cases, random forest models identified IL-6 as the most important predictor of the primary composite endpoint of death, heart failure (HF) or MI hospitalization, and the separate endpoints death and HF hospitalization. IL-18 was the most important predictor of MI hospitalization. In a Cox regression, the highest tertile of IL-6 was associated with the composite endpoint (HR (95% CI) 1.91 (1.31-2.79)), death (2.38 (1.42-3.98)) and HF hospitalization (2.70 (1.32-5.50)), when adjusting for age, sex and CV risk factors. The highest tertile of IL-18 was associated with MI hospitalization (2.31 (1.08-4.91)) when severity of coronary atherosclerosis was added to the same type of model. CONCLUSIONS When nine inflammatory markers involved in atherosclerosis were analyzed three months after the acute event in young MI patients, IL-6 and IL-18 were the most important biomarkers to predict long-term CV outcomes during 20 years of follow-up.
Collapse
Affiliation(s)
- Sofia Cederström
- Department of Clinical Sciences, Danderyd Hospital, Karolinska Institutet, Sweden.
| | - Tomas Jernberg
- Department of Clinical Sciences, Danderyd Hospital, Karolinska Institutet, Sweden
| | - Ann Samnegård
- Department of Clinical Sciences, Danderyd Hospital, Karolinska Institutet, Sweden
| | - Fredrik Johansson
- Department of Clinical Sciences, Danderyd Hospital, Karolinska Institutet, Sweden
| | - Angela Silveira
- Department of Medicine Solna K2, Karolinska Institutet and Karolinska University Hospital Solna
| | - Per Tornvall
- Department of Clinical Science and Education, Södersjukhuset, Karolinska Institutet, Stockholm, Sweden
| | - Pia Lundman
- Department of Clinical Sciences, Danderyd Hospital, Karolinska Institutet, Sweden
| |
Collapse
|
32
|
Offer S, Di Bucchianico S, Czech H, Pardo M, Pantzke J, Bisig C, Schneider E, Bauer S, Zimmermann EJ, Oeder S, Hartner E, Gröger T, Alsaleh R, Kersch C, Ziehm T, Hohaus T, Rüger CP, Schmitz-Spanke S, Schnelle-Kreis J, Sklorz M, Kiendler-Scharr A, Rudich Y, Zimmermann R. The chemical composition of secondary organic aerosols regulates transcriptomic and metabolomic signaling in an epithelial-endothelial in vitro coculture. Part Fibre Toxicol 2024; 21:38. [PMID: 39300536 DOI: 10.1186/s12989-024-00600-x] [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: 02/02/2024] [Accepted: 09/10/2024] [Indexed: 09/22/2024] Open
Abstract
BACKGROUND The formation of secondary organic aerosols (SOA) by atmospheric oxidation reactions substantially contributes to the burden of fine particulate matter (PM2.5), which has been associated with adverse health effects (e.g., cardiovascular diseases). However, the molecular and cellular effects of atmospheric aging on aerosol toxicity have not been fully elucidated, especially in model systems that enable cell-to-cell signaling. METHODS In this study, we aimed to elucidate the complexity of atmospheric aerosol toxicology by exposing a coculture model system consisting of an alveolar (A549) and an endothelial (EA.hy926) cell line seeded in a 3D orientation at the air‒liquid interface for 4 h to model aerosols. Simulation of atmospheric aging was performed on volatile biogenic (β-pinene) or anthropogenic (naphthalene) precursors of SOA condensing on soot particles. The similar physical properties for both SOA, but distinct differences in chemical composition (e.g., aromatic compounds, oxidation state, unsaturated carbonyls) enabled to determine specifically induced toxic effects of SOA. RESULTS In A549 cells, exposure to naphthalene-derived SOA induced stress-related airway remodeling and an early type I immune response to a greater extent. Transcriptomic analysis of EA.hy926 cells not directly exposed to aerosol and integration with metabolome data indicated generalized systemic effects resulting from the activation of early response genes and the involvement of cardiovascular disease (CVD) -related pathways, such as the intracellular signal transduction pathway (PI3K/AKT) and pathways associated with endothelial dysfunction (iNOS; PDGF). Greater induction following anthropogenic SOA exposure might be causative for the observed secondary genotoxicity. CONCLUSION Our findings revealed that the specific effects of SOA on directly exposed epithelial cells are highly dependent on the chemical identity, whereas non directly exposed endothelial cells exhibit more generalized systemic effects with the activation of early stress response genes and the involvement of CVD-related pathways. However, a greater correlation was made between the exposure to the anthropogenic SOA compared to the biogenic SOA. In summary, our study highlights the importance of chemical aerosol composition and the use of cell systems with cell-to-cell interplay on toxicological outcomes.
Collapse
Affiliation(s)
- Svenja Offer
- Joint Mass Spectrometry Center (JMSC) at Comprehensive Molecular Analytics (CMA), Helmholtz Zentrum München, Ingolstädter Landstr. 1, D-85764, Neuherberg, Germany
- Joint Mass Spectrometry Center (JMSC) at Analytical Chemistry, Institute of Chemistry, University of Rostock, Albert-Einstein-Str. 27, D-18059, Rostock, Germany
| | - Sebastiano Di Bucchianico
- Joint Mass Spectrometry Center (JMSC) at Comprehensive Molecular Analytics (CMA), Helmholtz Zentrum München, Ingolstädter Landstr. 1, D-85764, Neuherberg, Germany.
- Joint Mass Spectrometry Center (JMSC) at Analytical Chemistry, Institute of Chemistry, University of Rostock, Albert-Einstein-Str. 27, D-18059, Rostock, Germany.
- Department Life, Light & Matter (LLM), University of Rostock, D-18051, Rostock, Germany.
| | - Hendryk Czech
- Joint Mass Spectrometry Center (JMSC) at Comprehensive Molecular Analytics (CMA), Helmholtz Zentrum München, Ingolstädter Landstr. 1, D-85764, Neuherberg, Germany
- Joint Mass Spectrometry Center (JMSC) at Analytical Chemistry, Institute of Chemistry, University of Rostock, Albert-Einstein-Str. 27, D-18059, Rostock, Germany
| | - Michal Pardo
- Department of Earth and Planetary Sciences, Faculty of Chemistry, Weizmann Institute of Science, 234 Herzl Street, POB 26, Rehovot, ISR-7610001, Israel
| | - Jana Pantzke
- Joint Mass Spectrometry Center (JMSC) at Comprehensive Molecular Analytics (CMA), Helmholtz Zentrum München, Ingolstädter Landstr. 1, D-85764, Neuherberg, Germany
- Joint Mass Spectrometry Center (JMSC) at Analytical Chemistry, Institute of Chemistry, University of Rostock, Albert-Einstein-Str. 27, D-18059, Rostock, Germany
| | - Christoph Bisig
- Joint Mass Spectrometry Center (JMSC) at Comprehensive Molecular Analytics (CMA), Helmholtz Zentrum München, Ingolstädter Landstr. 1, D-85764, Neuherberg, Germany
| | - Eric Schneider
- Joint Mass Spectrometry Center (JMSC) at Analytical Chemistry, Institute of Chemistry, University of Rostock, Albert-Einstein-Str. 27, D-18059, Rostock, Germany
- Department Life, Light & Matter (LLM), University of Rostock, D-18051, Rostock, Germany
| | - Stefanie Bauer
- Joint Mass Spectrometry Center (JMSC) at Comprehensive Molecular Analytics (CMA), Helmholtz Zentrum München, Ingolstädter Landstr. 1, D-85764, Neuherberg, Germany
| | - Elias J Zimmermann
- Joint Mass Spectrometry Center (JMSC) at Comprehensive Molecular Analytics (CMA), Helmholtz Zentrum München, Ingolstädter Landstr. 1, D-85764, Neuherberg, Germany
- Joint Mass Spectrometry Center (JMSC) at Analytical Chemistry, Institute of Chemistry, University of Rostock, Albert-Einstein-Str. 27, D-18059, Rostock, Germany
| | - Sebastian Oeder
- Joint Mass Spectrometry Center (JMSC) at Comprehensive Molecular Analytics (CMA), Helmholtz Zentrum München, Ingolstädter Landstr. 1, D-85764, Neuherberg, Germany
| | - Elena Hartner
- Joint Mass Spectrometry Center (JMSC) at Comprehensive Molecular Analytics (CMA), Helmholtz Zentrum München, Ingolstädter Landstr. 1, D-85764, Neuherberg, Germany
- Joint Mass Spectrometry Center (JMSC) at Analytical Chemistry, Institute of Chemistry, University of Rostock, Albert-Einstein-Str. 27, D-18059, Rostock, Germany
| | - Thomas Gröger
- Joint Mass Spectrometry Center (JMSC) at Comprehensive Molecular Analytics (CMA), Helmholtz Zentrum München, Ingolstädter Landstr. 1, D-85764, Neuherberg, Germany
- Joint Mass Spectrometry Center (JMSC) at Analytical Chemistry, Institute of Chemistry, University of Rostock, Albert-Einstein-Str. 27, D-18059, Rostock, Germany
| | - Rasha Alsaleh
- Institute and Outpatient Clinic of Occupational, Social and Environmental Medicine, Friedrich-Alexander University of Erlangen-Nuremberg, Henkestr. 9-11, D-91054, Erlangen, Germany
| | - Christian Kersch
- Institute and Outpatient Clinic of Occupational, Social and Environmental Medicine, Friedrich-Alexander University of Erlangen-Nuremberg, Henkestr. 9-11, D-91054, Erlangen, Germany
| | - Till Ziehm
- Institute of Energy and Climate Research, Forschungszentrum Jülich GmbH, Troposphere (IEK-8), Wilhelm- Johen-Str, D-52428, Jülich, Germany
| | - Thorsten Hohaus
- Institute of Energy and Climate Research, Forschungszentrum Jülich GmbH, Troposphere (IEK-8), Wilhelm- Johen-Str, D-52428, Jülich, Germany
| | - Christopher P Rüger
- Joint Mass Spectrometry Center (JMSC) at Analytical Chemistry, Institute of Chemistry, University of Rostock, Albert-Einstein-Str. 27, D-18059, Rostock, Germany
- Department Life, Light & Matter (LLM), University of Rostock, D-18051, Rostock, Germany
| | - Simone Schmitz-Spanke
- Institute and Outpatient Clinic of Occupational, Social and Environmental Medicine, Friedrich-Alexander University of Erlangen-Nuremberg, Henkestr. 9-11, D-91054, Erlangen, Germany
| | - Jürgen Schnelle-Kreis
- Joint Mass Spectrometry Center (JMSC) at Comprehensive Molecular Analytics (CMA), Helmholtz Zentrum München, Ingolstädter Landstr. 1, D-85764, Neuherberg, Germany
| | - Martin Sklorz
- Joint Mass Spectrometry Center (JMSC) at Comprehensive Molecular Analytics (CMA), Helmholtz Zentrum München, Ingolstädter Landstr. 1, D-85764, Neuherberg, Germany
| | - Astrid Kiendler-Scharr
- Institute of Energy and Climate Research, Forschungszentrum Jülich GmbH, Troposphere (IEK-8), Wilhelm- Johen-Str, D-52428, Jülich, Germany
| | - Yinon Rudich
- Department of Earth and Planetary Sciences, Faculty of Chemistry, Weizmann Institute of Science, 234 Herzl Street, POB 26, Rehovot, ISR-7610001, Israel
| | - Ralf Zimmermann
- Joint Mass Spectrometry Center (JMSC) at Comprehensive Molecular Analytics (CMA), Helmholtz Zentrum München, Ingolstädter Landstr. 1, D-85764, Neuherberg, Germany
- Joint Mass Spectrometry Center (JMSC) at Analytical Chemistry, Institute of Chemistry, University of Rostock, Albert-Einstein-Str. 27, D-18059, Rostock, Germany
- Department Life, Light & Matter (LLM), University of Rostock, D-18051, Rostock, Germany
| |
Collapse
|
33
|
Lin Z, Xie Y, Lin Y, Chen X. Association between composite dietary antioxidant index and atherosclerosis cardiovascular disease in adults: A cross-sectional study. Nutr Metab Cardiovasc Dis 2024; 34:2165-2172. [PMID: 39003133 DOI: 10.1016/j.numecd.2024.06.002] [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: 11/04/2023] [Revised: 05/29/2024] [Accepted: 06/04/2024] [Indexed: 07/15/2024]
Abstract
BACKGROUND AND AIM The objective of our study was to examine the association between composite dietary antioxidant index (CDAI) and atherosclerotic cardiovascular disease (ASCVD) in adults. METHODS AND RESULTS Data was gathered from the National Health and Nutrition Examination Survey (NHANES) between 2001 and 2018. To examine the connection between CDAI and ASCVD, multiple logistic regression analyses were performed. Restricted cubic splines were utilized to examine non-linear correlations, and the inflection point was identified using a two-piecewise linear regression approach. Subgroup analyses were performed to demonstrate stability of results. A total of 44,494 individuals were included in the study. The multivariate logistic regression model was fully adjusted and revealed an odds ratio of 0.968 (95% CI: 0.959-0.978; P < 0.001) for the correlation between CDAI and ASCVD. Furthermore, individuals in the highest quartile of CDAI exhibited a decreased risk of ASCVD compared to those in the lowest quartile [0.716 (0.652-0.787); P < 0.001]. Moreover, restricted cubic spline (RCS) analysis revealed non-linear relationship between CDAI and ASCVD, with inflection point at -0.387. The analysis of subgroups showed that the importance of CDAI remained consistent among various age, sex, race, body mass index (BMI), and physical activity. CONCLUSIONS Our research revealed an inverse and non-linear relationship between CDAI and ASCVD in adults. The implications of these findings are significant for future studies and the formulation of dietary guidelines.
Collapse
Affiliation(s)
- Zhiyong Lin
- Department of Cardiovascular and Thoracic Surgery, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou 325027, China
| | - Yuanyuan Xie
- Department of General Practitioner, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325027, China
| | - Yi Lin
- Department of Endocrinology, The Third Affiliated Hospital of Shanghai University, Wenzhou People's Hospital, Wenzhou, 325027, Zhejiang, China
| | - Xiyi Chen
- Department of Cardiovascular and Thoracic Surgery, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou 325027, China.
| |
Collapse
|
34
|
Ko DS, Kim YH. Mendelian Randomization Studies in Atherosclerotic Cardiovascular Diseases. J Lipid Atheroscler 2024; 13:280-291. [PMID: 39355404 PMCID: PMC11439750 DOI: 10.12997/jla.2024.13.3.280] [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: 10/27/2023] [Revised: 01/07/2024] [Accepted: 02/05/2024] [Indexed: 10/03/2024] Open
Abstract
This review aimed to highlight the pivotal role of Mendelian randomization (MR) in advancing atherosclerotic cardiovascular disease (ASCVD) research-a field often hindered by the complexities and limitations of traditional studies. MR, which uses genetic variants as instrumental variables, provides a robust mechanism for inferring causality, offering insights untainted by the confounding factors and biases often prevalent in observational and randomized controlled trials. We explored the significant contributions of MR for elucidating the causal relationship between low-density lipoprotein cholesterol and ASCVD, and analyzed its assumptions and methodological nuances. We discussed issues surrounding instrumental variable selection, pleiotropy, and ethical considerations, in an effort to offer a balanced and insightful analysis. We highlighted the promising integration of MR with emerging technologies and global data sharing, as well as its potential to drive personalized medicine. This review provided a concise yet comprehensive journey into MR's transformative impact on ASCVD research, offering a blend of current insights and challenges, in addition to future prospects. We aimed to serve a valuable resource for those seeking to navigate the intricate pathways of causality and intervention in ASCVD, to aid the development of enhanced understanding and targeted treatment strategies in the future.
Collapse
Affiliation(s)
- Dai Sik Ko
- Division of Vascular Surgery, Department of General Surgery, Gachon University College of Medicine, Gil Medical Center, Incheon, Korea
| | - Yun Hak Kim
- Department of Biomedical Informatics, School of Medicine, Pusan National University, Yangsan, Korea
- Department of Anatomy, School of Medicine, Pusan National University, Yangsan, Korea
| |
Collapse
|
35
|
Evans WS, Pena GS, Gelman B, Kuzmiak‐Glancy S, Prior SJ. Unilateral hindlimb ischaemia-induced systemic inflammation is associated with non-ischaemic skeletal muscle inflammation. Exp Physiol 2024; 109:1604-1613. [PMID: 38888281 PMCID: PMC11363109 DOI: 10.1113/ep091901] [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: 06/04/2024] [Indexed: 06/20/2024]
Abstract
Skeletal muscle atrophy and dysfunction commonly accompany cardiovascular diseases such as peripheral arterial disease and may be partially attributable to systemic inflammation. We sought to determine whether acute systemic inflammation in a model of hindlimb ischaemia (HLI) could affect skeletal muscle macrophage infiltration, fibre size, or capillarization, independent of the ischaemia. Eight-week-old C57BL/6 male mice underwent either Sham or HLI surgery, and were killed 1, 3, or 7 days post-surgery. Circulating inflammatory cytokine concentrations were measured, as well as immune cell infiltration and morphology of skeletal muscle from both limbs of HLI and Sham mice. In HLI compared with Sham mice at day 1, plasma interleukin-1β levels were 216% higher (0.48 ± 0.10 vs. 0.15 ± 0.01 pg/μL, P = 0.005) and decreased by day 3. This was followed by increased macrophage presence in muscle from both ischaemic and non-ischaemic limbs of HLI mice by day 7 (7.3- and 2.3-fold greater than Sham, respectively, P < 0.0001). In HLI mice, muscle from the ischaemic limb had 21% lower fibre cross-sectional area than the non-ischaemic limb (724 ± 28 vs. 916 ± 46 μm2, P = 0.01), but the non-ischaemic limb of HLI mice was no different from Sham. This shows that HLI induces acute systemic inflammation accompanied by immune infiltration in both ischaemic and remote skeletal muscle; however, this did not induce skeletal muscle atrophy in remote muscle within the 7-day time course of this study. This effect of local skeletal muscle ischaemia on the inflammatory status of remote skeletal muscle may signal a priming of muscle for subsequent atrophy over a longer time course. HIGHLIGHTS: What is the central question of this study? Does hindlimb ischaemia-induced inflammation cause acute immune, inflammatory and morphological alterations in remote non-ischaemic skeletal muscle? What is the main finding and its importance? Hindlimb ischaemia induced systemic inflammation with subsequent neutrophil and macrophage infiltration in both ischaemic and non-ischaemic skeletal muscle; however, morphological changes did not occur in non-ischaemic muscle within 7 days. These immune alterations may have functional implications that take longer than 7 days to manifest, and subsequent or prolonged systemic inflammation and immune infiltration of muscle could lead to morphological changes and functional decline.
Collapse
Affiliation(s)
- William S. Evans
- Department of KinesiologyUniversity of Maryland School of Public HealthCollege ParkMarylandUSA
| | - Gabriel S. Pena
- Department of KinesiologyUniversity of Maryland School of Public HealthCollege ParkMarylandUSA
| | - Beata Gelman
- Department of KinesiologyUniversity of Maryland School of Public HealthCollege ParkMarylandUSA
| | - Sarah Kuzmiak‐Glancy
- Department of KinesiologyUniversity of Maryland School of Public HealthCollege ParkMarylandUSA
| | - Steven J. Prior
- Department of KinesiologyUniversity of Maryland School of Public HealthCollege ParkMarylandUSA
- Baltimore Veterans Affairs Geriatric ResearchEducation and Clinical Center and Research and Development ServiceBaltimoreMarylandUSA
| |
Collapse
|
36
|
Bertero E, Carmisciano L, Jonasson C, Butler J, Maack C, Ameri P. Association of inflammatory markers with incident heart failure or cancer in the HUNT3 and Health ABC population studies. Eur J Prev Cardiol 2024; 31:1400-1407. [PMID: 38429011 DOI: 10.1093/eurjpc/zwae089] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/05/2023] [Revised: 02/04/2024] [Accepted: 02/21/2024] [Indexed: 03/03/2024]
Abstract
AIMS To investigate the relationship between chronic low-grade inflammation, as measured by high-sensitivity C-reactive protein (hsCRP) levels, and incident heart failure (HF) or cancer. METHODS AND RESULTS We assessed the relationship between baseline hsCRP concentrations and subsequent HF or cancer in two community-based cohorts, the Trøndelag Health Study (HUNT3) and the Health, Aging and Body Composition (ABC) study. In the latter, the analysis was replicated with interleukin (IL)-1, IL-6, or tumour necrosis factor (TNF)-α instead of hsCRP. In HUNT3, hsCRP was measured in 47 163 subjects (mean age 52.3 ± 15.8 years). During a median follow-up of 12.1 years, 2034 (4.3%) individuals developed HF and 5024 (10.7%) cancer, with 442 (0.9%) being diagnosed with both. After adjusting for age, male sex, diabetes, obesity, previous or current smoking, and comorbidities, elevated baseline hsCRP was associated with a higher risk of HF or cancer [hazard ratio (HR) 1.09; 95% confidence interval (CI), 1.07-1.10]. In the Health ABC study, hsCRP levels were assessed in 2803 participants, who had a mean age of 72.6 ± 2.9 years and a higher burden of comorbidities than in HUNT3. During a median follow-up of 8.2 years, HF and cancer were diagnosed in 346 (12.3%) and 776 (27.7%) subjects, respectively, with 77 (2.7%) having both conditions. After adjusting for the same variables used for the HUNT3 cohort, hsCRP remained significantly associated with incident HF or cancer (HR 1.11; 95% CI, 1.05-1.18), as were IL-1 (HR 1.15; 1.07-1.24), IL-6 (HR 1.09; 1.02-1.17), and TNF-α (HR 1.15; 1.07-1.24). CONCLUSION A state of chronic, low-grade inflammation captured by an increase in hsCRP levels is associated with an increased risk of developing HF or cancer, with potential implications for clinical trials with anti-inflammatory therapies.
Collapse
Affiliation(s)
- Edoardo Bertero
- Department of Internal Medicine, University of Genova, Viale Benedetto XV, 6, 16132 Genova, Italy
- Cardiovascular Disease Unit, Cardiac, Thoracic and Vascular Department, IRCCS Ospedale Policlinico San Martino, Largo Rosanna Benzi 10, 16132 Genova, Italy
- Comprehensive Heart Failure Center (CHFC), University Clinic Würzburg, 97078 Würzburg, Germany
| | - Luca Carmisciano
- Department of Health Sciences, Section of Biostatistics, University of Genova, Via Antonio Pastore 1, 16132 Genova, Italy
| | - Christian Jonasson
- K.G. Jebsen Center for Genetic Epidemiology, Department of Public Health and Nursing, Faculty of Medicine and Health Science, Norwegian University of Science and Technology, Trondheim, Norway
| | - Javed Butler
- Baylor Scott and White Research Institute, Dallas, TX, USA
- Department of Medicine, University of Mississippi, Jackson, MS, USA
| | - Christoph Maack
- Comprehensive Heart Failure Center (CHFC), University Clinic Würzburg, 97078 Würzburg, Germany
| | - Pietro Ameri
- Department of Internal Medicine, University of Genova, Viale Benedetto XV, 6, 16132 Genova, Italy
- Cardiovascular Disease Unit, Cardiac, Thoracic and Vascular Department, IRCCS Ospedale Policlinico San Martino, Largo Rosanna Benzi 10, 16132 Genova, Italy
| |
Collapse
|
37
|
Nsor OO, Alabi BA, Badejo JA, Afolabi F, Nku-Ekpang OA, Iwalewa EO. Soursop leaf extract and fractions protects against L-NAME-induced hypertension and hyperlipidemia. Front Nutr 2024; 11:1437101. [PMID: 39171117 PMCID: PMC11337232 DOI: 10.3389/fnut.2024.1437101] [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: 05/23/2024] [Accepted: 07/16/2024] [Indexed: 08/23/2024] Open
Abstract
Introduction Despite the high phenolic content of Annona muricata, little is known about its anti-hypertensive and antihyperlipidemic properties. This study evaluated the anti-hypertensive and antihyperlipidemic potential of A. muricata leaf extracts. Materials and methods Forty-two male Wistar rats were divided into seven groups of six animals each. N-nitro-L-arginine methyl ester (L-NAME) was used to induce hypertension and hyperlipidemia. Results Phytochemical screening of Annona muricata leaf extracts (AMLE) revealed the presence of saponins, alkaloids, flavonoids, tannins, coumarins, steroids, terpenoids, and phenols. Comparing the methanol extract with the ethyl acetate fraction, quantification revealed that the methanol extract contained more phenolics, flavonoids, and alkaloids. The AMLE rats significantly reduced triglycerides, total cholesterol, LDL, VLDL, atherogenic index, coronary risk index, and blood pressure. The significant decrease in GSH, catalase, SOD, GST, and oxidative stress markers (MDA, nitrites, and MPO) was reversed by AMLE in a dose-dependent manner. Also, the elevated serum levels of TNF-α and IL-1β in the hypertensive rats were attenuated in the treatment groups. Discussion This study suggests the potential ameliorative effects of Annona muricata leaf extracts against L-NAME-induced hypertension in rats. Notably, the study showed the antioxidant and anti-inflammatory properties of A. muricata leaf extracts, which is seen in its ability to attenuate oxidative stress and inflammatory cytokines in L-NAME-induced hypertensive rats. A. muricata extracts also decreased atherogenic risk and improved lipid profiles.
Collapse
Affiliation(s)
- Okim Okim Nsor
- Neuropharmacology Unit, Department of Pharmacology and Therapeutics, Faculty of Basic Medical Sciences, College of Medicine, University of Ibadan, Ibadan, Nigeria
| | - Babatunde Adebola Alabi
- Department of Pharmacology and Therapeutics, Faculty of Basic Clinical Sciences, Bowen University, Iwo, Nigeria
- Department of Pharmacology and Therapeutics, Faculty of Medicine and Pharmacy, Kampala International University in Tanzania, Dar es Salaam, Tanzania
| | - Joseph Ayo Badejo
- Neuropharmacology Unit, Department of Pharmacology and Therapeutics, Faculty of Basic Medical Sciences, College of Medicine, University of Ibadan, Ibadan, Nigeria
| | - Faith Afolabi
- Neuropharmacology Unit, Department of Pharmacology and Therapeutics, Faculty of Basic Medical Sciences, College of Medicine, University of Ibadan, Ibadan, Nigeria
| | - Okot-Asi Nku-Ekpang
- Department of Physiology, Faculty of Basic Medical Sciences, University of Calabar, Calabar, Nigeria
| | - Ezekiel Olugbenga Iwalewa
- Neuropharmacology Unit, Department of Pharmacology and Therapeutics, Faculty of Basic Medical Sciences, College of Medicine, University of Ibadan, Ibadan, Nigeria
| |
Collapse
|
38
|
Khan MS, Talha KM, Maqsood MH, Rymer JA, Borlaug BA, Docherty KF, Pandey A, Kahles F, Cikes M, Lam CS, Ducharme A, Voors AA, Hernandez AF, Lincoff AM, Petrie MC, Ridker PM, Fudim M. Interleukin-6 and Cardiovascular Events in Healthy Adults: MESA. JACC. ADVANCES 2024; 3:101063. [PMID: 39077632 PMCID: PMC11284704 DOI: 10.1016/j.jacadv.2024.101063] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 12/06/2023] [Revised: 05/13/2024] [Accepted: 05/20/2024] [Indexed: 07/31/2024]
Abstract
Background Elevated interleukin (IL)-6 levels have been linked to adverse outcomes in patients with and without baseline cardiovascular disease (CVD). Objectives The purpose of this study was to examine the association between circulating IL-6 levels and CVD events without baseline CVD across racial and ethnic groups. Methods We conducted an observational analysis utilizing the MESA (Multi-Ethnic Study of Atherosclerosis), a multicenter, prospective community-based study of CVD at baseline from four racial and ethnic groups. IL-6 levels were measured at the time of enrollment (visit 1) and were divided into 3 terciles. Patient baseline characteristics and outcomes, including all-cause mortality, CV mortality, heart failure, and non-CV mortality, were included. Cox proportional hazard regression models were used to assess associations between IL-6 levels and study outcomes with IL-6 tercile 1 as reference. Results Of 6,622 individuals, over half were women (53%) with a median age of 62 (IQR: 53-70) years. Racial and ethnic composition was non-Hispanic White (39%) followed by African American (27%), Hispanic (22%), and Chinese American (12%). Compared to tercile 1, participants with IL-6 tercile 3 had a higher adjusted risk of and all-cause mortality (HR: 1.98 [95% CI: 1.67-2.36]), CV mortality (HR: 1.55 [95% CI: 1.05-2.30]), non-CV mortality (HR: 2.05 [95% CI: 1.65-2.56]), and heart failure (HR: 1.48 [95% CI: 0.99-2.19]). When tested as a continuous variable, higher levels of IL-6 were associated with an increased risk of all individual outcomes. Compared to non-Hispanic White participants, the unadjusted and adjusted risk of all outcomes across all races and ethnicities was similar across all IL-6 terciles. Conclusions High levels of circulating IL-6 are associated with worse CV outcomes and increased all-cause mortality consistently across all racial and ethnic groups.
Collapse
Affiliation(s)
- Muhammad Shahzeb Khan
- Duke University Medical Center, Duke Clinical Research Institute, Durham, North Carolina, USA
| | - Khawaja M. Talha
- Department of Medicine, University of Mississippi Medical Center, Jackson, Mississippi, USA
| | - Muhammad Haisum Maqsood
- Department of Cardiology, DeBakey Heart and Vascular Center, Houston Methodist Hospital, Houston, Texas, USA
| | - Jennifer A. Rymer
- Duke University Medical Center, Duke Clinical Research Institute, Durham, North Carolina, USA
| | - Barry A. Borlaug
- Department of Cardiovascular Medicine, Mayo Clinic, Rochester, Minnesota, USA
| | - Kieran F. Docherty
- Institute of Cardiovascular and Medical Sciences, University of Glasgow, Glasgow, United Kingdom
| | - Ambarish Pandey
- Division of Cardiology, Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, Texas, USA
| | - Florian Kahles
- Department of Internal Medicine I-Cardiology, University Hospital Aachen, Aachen, Germany
| | - Maja Cikes
- University of Zagreb School of Medicine, University Hospital Centre, Zagreb, Croatia
| | | | - Anique Ducharme
- Institut de Cardiologie, de Montréal, Université de Montréal, Montréal, Québec, Canada
| | - Adrian A. Voors
- Department of Cardiology, University Medical Center of Groningen, University of Groningen, Groningen, the Netherlands
| | - Adrian F. Hernandez
- Duke University Medical Center, Duke Clinical Research Institute, Durham, North Carolina, USA
| | - A. Michael Lincoff
- Heart, Vascular, and Thoracic Institute, Cleveland Clinic, Cleveland, Ohio, USA
| | - Mark C. Petrie
- Institute of Cardiovascular and Medical Sciences, University of Glasgow, Glasgow, United Kingdom
| | - Paul M. Ridker
- Center for Cardiovascular Disease Prevention, Division of Preventive Medicine and the Division of Cardiovascular Medicine, Brigham and Women's Hospital, Boston, Massachusetts, USA
| | - Marat Fudim
- Duke University Medical Center, Duke Clinical Research Institute, Durham, North Carolina, USA
| |
Collapse
|
39
|
Lake JE, Feng H, Hyatt AN, Miao H, Debroy P, Funderburg N, Ailstock K, Dobs A, Haberlen S, Magnani JW, Margolick JB, McGowan K, Palella FJ, Witt MD, Bhasin S, Budoff MJ, Post WS, Brown TT. Transgender Women With Suppressed Testosterone Display Lower Burden of Coronary Disease Than Matched Cisgender Men. J Endocr Soc 2024; 8:bvae120. [PMID: 38974987 PMCID: PMC11223995 DOI: 10.1210/jendso/bvae120] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/29/2024] [Indexed: 07/09/2024] Open
Abstract
Context Cardiovascular disease (CVD) in transgender women (TW) may be affected by gender-affirming hormone therapy (GAHT) and HIV, but few data compare TW on contemporary GAHT to well-matched controls. Objective We compared CVD burden and biomarker profiles between TW and matched cisgender men (CM). Methods Adult TW on GAHT (n = 29) were recruited for a cross-sectional study (2018-2020). CM (n = 48) from the former Multicenter AIDS Cohort Study were matched 2:1 to TW on HIV serostatus, age ±5 years, race/ethnicity, BMI category and antiretroviral therapy (ART) type. Cardiac parameters were measured by CT and coronary atherosclerosis by coronary CT angiography; sex hormone and biomarker concentrations were measured centrally from stored samples. Results Overall, median age was 53 years and BMI 29 kg/m2; 69% were non-white. All participants with HIV (71%) had viral suppression on ART. Only 31% of TW had testosterone suppression (<50 ng/dL, TW-S). Traditional CVD risk factors were similar between groups, except that TW-S had higher BMI than TW with non-suppressed testosterone (TW-T). TW-S had no evidence of non-calcified coronary plaque or advanced coronary stenosis, whereas TW-T and CM had similar burden. TW had lower prevalence of any coronary plaque, calcified plaque and mixed plaque than CM, regardless of testosterone concentrations and HIV serostatus. Estradiol but not testosterone concentrations moderately and negatively correlated with the presence of coronary plaque and stenosis. Small sample size limited statistical power. Conclusion Older TW with suppressed total testosterone on GAHT had no CT evidence of non-calcified coronary plaque or advanced coronary stenosis. Longitudinal studies to understand relationships between GAHT and CVD risk in TW are needed.
Collapse
Affiliation(s)
- Jordan E Lake
- Department of Medicine, UTHealth Houston, Houston, TX 77030, USA
| | - Han Feng
- Tulane Research and Innovation for Arrhythmia Discoveries-TRIAD Center, Tulane University School of Medicine, New Orleans, LA 70112, USA
| | - Ana N Hyatt
- Department of Medicine, UTHealth Houston, Houston, TX 77030, USA
| | - Hongyu Miao
- Department of Statistics and College of Nursing, Florida State University, Tallahassee, FL 32306, USA
| | - Paula Debroy
- Department of Medicine, UTHealth Houston, Houston, TX 77030, USA
| | - Nicholas Funderburg
- Division of Medical Laboratory Science, The Ohio State University, Columbus, OH 43210, USA
| | - Kate Ailstock
- Division of Medical Laboratory Science, The Ohio State University, Columbus, OH 43210, USA
| | - Adrian Dobs
- Department of Medicine, Johns Hopkins University, Baltimore, MD 21218, USA
| | - Sabina Haberlen
- Department of Medicine, Johns Hopkins University, Baltimore, MD 21218, USA
| | - Jared W Magnani
- Department of Medicine, University of Pittsburgh, Pittsburgh, PA 15261, USA
| | - Joseph B Margolick
- Department of Medicine, Johns Hopkins University, Baltimore, MD 21218, USA
| | - Kate McGowan
- Department of Medicine, Johns Hopkins University, Baltimore, MD 21218, USA
| | - Frank J Palella
- Department of Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA
| | - Mallory D Witt
- Department of Medicine, Lundquist Institute, Torrance, CA 90502, USA
| | - Shalender Bhasin
- Department of Medicine, Brigham and Women's Hospital, Boston, MA 02115, USA
| | - Matthew J Budoff
- Department of Medicine, Lundquist Institute, Torrance, CA 90502, USA
| | - Wendy S Post
- Department of Medicine, Johns Hopkins University, Baltimore, MD 21218, USA
| | - Todd T Brown
- Department of Medicine, Johns Hopkins University, Baltimore, MD 21218, USA
| |
Collapse
|
40
|
Maia LA, de Souza JR, da Silva LDFR, Magnani M, de Souza EL, de Brito Alves JL. Effects of Probiotics on Inflammatory Biomarkers and Its Associations With Cardiac Autonomic Function in Women With Arterial Hypertension: A Secondary Analysis of a Randomized Clinical Trial. Probiotics Antimicrob Proteins 2024:10.1007/s12602-024-10303-6. [PMID: 38842655 DOI: 10.1007/s12602-024-10303-6] [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] [Accepted: 05/31/2024] [Indexed: 06/07/2024]
Abstract
Preclinical evidence suggests that probiotic administration may exert an anti-inflammatory effect and reduce autonomic dysfunction and blood pressure. This study evaluated the effects of probiotic therapy on inflammatory biomarkers and characterized the correlations between inflammation and cardiac autonomic function in women with arterial hypertension. Women were randomized into probiotics (n = 20) or placebo (n = 20). The probiotic group received 109 CFU/day of Lactobacillus (L.) paracasei LPC-37, L. rhamnosus HN001, L. acidophilus NCFM, and Bifidobacterium lactis HN019, and the placebo group received polydextrose. Clinical, electrocardiogram, heart rate variability (HRV) analysis, and cytokine levels were assessed at baseline and after 8 weeks. Women who received probiotics for 8 weeks had increased serum levels of IL-17A (p = 0.02) and decreased INF-γ (p = 0.02) compared to baseline. Probiotic supplementation increased serum levels of IL-10 compared to the placebo group (p = 0.03). Probiotic or placebo administration did not change serum levels of TNFα and IL-6. Serum levels of IL-2 (p = 0.001, and p = 0.001) and IL-4 (p = 0.001, and p = 0.001) were reduced in women receiving placebo or probiotics, respectively. Correlations between HRV indices and inflammatory variables showed that INF-γ was positively correlated with heart rate (HR) and sympathetic HRV indices and negatively correlated with vagal HRV indices. IL-10 was negatively correlated with HR and sympathetic HRV indices. IL-6 was negatively correlated with parasympathetic HRV indices and positively correlated with SD2/SD1 ratio. Probiotic therapy has a discreet anti-inflammatory effect in hypertensive women, and pro-inflammatory cytokines were negatively correlated with vagal modulation and positively correlated with sympathetic modulation of HRV. The clinical trial was registered in the Brazilian Registry of Clinical Trials (ReBEC) with the identification RBR-9mj2dt.
Collapse
Affiliation(s)
- Larissa Araújo Maia
- Department of Nutrition, Health Sciences Center, Federal University of Paraiba, Joao Pessoa, PB, Brazil
| | | | | | - Marciane Magnani
- Department of Food Engineering, Technology Center, Federal University of Paraiba, João Pessoa, PB, Brazil
| | - Evandro Leite de Souza
- Department of Nutrition, Health Sciences Center, Federal University of Paraiba, Joao Pessoa, PB, Brazil
| | - José Luiz de Brito Alves
- Department of Nutrition, Health Sciences Center, Federal University of Paraiba, Joao Pessoa, PB, Brazil.
| |
Collapse
|
41
|
Krajewski PK, Matusiak Ł, Ständer S, Thaçi D, Szepietowski JC, Zirpel H. Risk of cardiovascular disorders in hidradenitis suppurativa patients: a large-scale, propensity-matched global retrospective cohort study. Int J Dermatol 2024; 63:799-805. [PMID: 38644522 DOI: 10.1111/ijd.17186] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/06/2024] [Revised: 03/23/2024] [Accepted: 03/25/2024] [Indexed: 04/23/2024]
Abstract
BACKGROUND Patients with hidradenitis suppurativa (HS) often suffer from comorbid diabetes, metabolic syndrome, and hyperlipidemia and, therefore, are susceptible to the development of cardiovascular diseases (CVDs). Moreover, systemic inflammation plays a vital role in the development of atherosclerosis. The creation of atherosclerotic plaque is characterized by endothelial dysfunction driven by elevated concentrations of interleukin (IL)-1, IL-6, and IL-18 among others, as well as tumor necrosis factor (TNF) alpha. METHODS This study aimed to assess the risk of HS patients developing CVDs. We performed a large-scale, propensity-matched global retrospective cohort study analyzing the risk of development of CVDs in patients suffering from HS. The analysis included 144,100 HS patients with 144,100 healthy controls (HC). The cohorts were matched regarding demographics and history of diseases relevant to CVDs, e.g., diabetes, obesity, and nicotine dependence. A total of 90 cardiovascular disorders were identified. The identification of cardiovascular disorders was based on ≥1% appearance of the event, based on absolute numbers, in both cohorts. RESULTS Before the matching, HS patients displayed a higher frequency in excess weight or obesity (25 vs. 14.4%, respectively), nicotine dependence, and diabetes mellitus, but lower odds of primary hypertension in comparison to healthy controls. A total of 47 CVDs are associated with an increased risk of onset in HS patients. Although the highest hazard ratio (HR; 2.1; 95% CI: 1.95-2.269) was found for unspecified heart failure, the HS cohort was exceptionally predisposed to developing myocardial infarction (HR: 2.06; 95% CI: 1.88-2.27) and an acute embolism and deep vein thrombosis of the lower extremity (HR: 1.93; 95% CI: 1.74-2.14). CONCLUSIONS This is the most extensive study on the association of HS with CVDs. We demonstrated that HS patients are at significantly greater risk of developing various CVDs compared to matched controls, with heart failure being the most common one.
Collapse
Affiliation(s)
- Piotr K Krajewski
- Department of Dermatology, Venereology and Allergology, Wroclaw Medical University, Wroclaw, Poland
| | - Łukasz Matusiak
- Department of Dermatology, Venereology and Allergology, Wroclaw Medical University, Wroclaw, Poland
| | - Sascha Ständer
- Comprehensive Center for Inflammation Medicine, University-Hospital Schleswig-Holstein, Lübeck, Germany
| | - Diamant Thaçi
- Comprehensive Center for Inflammation Medicine, University-Hospital Schleswig-Holstein, Lübeck, Germany
| | - Jacek C Szepietowski
- Department of Dermatology, Venereology and Allergology, Wroclaw Medical University, Wroclaw, Poland
| | - Henner Zirpel
- Comprehensive Center for Inflammation Medicine, University-Hospital Schleswig-Holstein, Lübeck, Germany
| |
Collapse
|
42
|
Tsai CY, Liao WL, Wu HM, Chang CW, Chen WL, Hsieh CL. Acupuncture improves neurological function and anti-inflammatory effect in patients with acute ischemic stroke: A double-blinded randomized controlled trial. Complement Ther Med 2024; 82:103049. [PMID: 38729273 DOI: 10.1016/j.ctim.2024.103049] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2023] [Revised: 03/31/2024] [Accepted: 05/06/2024] [Indexed: 05/12/2024] Open
Abstract
BACKGROUND AND PURPOSE Acupuncture exerts an anti-inflammatory effect and is recommended by the World Health Organization as a complementary therapy for stroke. This study investigated the improvement in neurological function outcome in acute-stage intervention of acute ischemic stroke (AIS), and the anti-inflammatory effect of early acupuncture. METHODS Fifty patients with AIS were randomly assigned to either a control group (CG, 25 patients, received sham acupuncture) or treatment group (TG, 25 patients, received acupuncture treatment). Acupuncture intervention was administered twice a week for a total of 8 sessions over 4 consecutive weeks. The primary outcome was the changes in the National Institutes of Health Stroke Scale (NIHSS), modified Rankin Scale (mRS), and Barthel Index (BI) scores. The secondary outcome was the changes in serum inflammation-related biomarker levels.(ANAIS trial) RESULTS: A total of 35 patients (18 patients in the CG and 17 patients in the TG) completed the trial. The reduction in NIHSS scores was greater in the TG than in the CG between V2 (second assessment administered after acupuncture intervention) and V1 (first assessment administered before acupuncture intervention; 4.33 ± 1.91 vs. 2.68 ± 1.42, p = 0.005) and between V3 (third assessment administered 28 days after last acupuncture intervention) and V1 (6.00 ± 2.53 vs. 3.83 ± 2.31, p = 0.012). The increase in BI scores was greater in the TG than in the CG between V2 and V1 (28.89 ± 15.39 vs. 14.21 ± 19.38, p = 0.016) and between V3 and V1 (39.41 ± 20.98 vs. 25.00 ± 18.47, p = 0.038). Among participants with high inflammation, the increase in serum IL-12p70 level between V2 and V1 was greater in the TG than in the CG (0.20 ± 0.19 vs. -0.14 ± 0.30, pg/mL p = 0.006). CONCLUSIONS Acupuncture improved the neurological function of patients with AIS, and the relationship between acupuncture improving neurological function and anti-inflammatory effect needs further study. In addition, studies with larger sample sizes and longer follow-ups as well as multicenter clinical trials are expected in the future.
Collapse
Affiliation(s)
- Chueh-Yi Tsai
- Department of Neurology, Chung Shan Medical University Hospital, Taichung, Taiwan; Graduate Institute of Integrated Medicine, China Medical University, Taichung, Taiwan; Department of Neurology, Nantou Hospital, Ministry of Health and Welfare, Nantou, Taiwan; School of Medicine, Chung Shan Medical University, Taichung, Taiwan.
| | - Wen-Ling Liao
- Graduate Institute of Integrated Medicine, China Medical University, Taichung, Taiwan; Center for Personalized Medicine, China Medical University Hospital, Taichung, Taiwan
| | - Hung-Ming Wu
- Department of Neurology, Chung Shan Medical University Hospital, Taichung, Taiwan; Department of Neurology, Changhua Christian Hospital, Changhua, Taiwan
| | - Chia-Wei Chang
- Department of Neurology, Nantou Hospital, Ministry of Health and Welfare, Nantou, Taiwan
| | - Wei-Liang Chen
- Center for the Neuroscience and Behavioral Medicine, Children's National Research Institute, Children's National Medical Center, Washington D.C, USA; George Washington University, Washington D.C, USA
| | - Ching-Liang Hsieh
- Graduate Institute of Acupuncture Science, China Medical University, Taichung, Taiwan; Department of Chinese Medicine, China Medical University Hospital, Taichung, Taiwan; Chinese Medicine Research Center, China Medical University, Taichung, Taiwan.
| |
Collapse
|
43
|
Zhang CL, Maccarone JR, Grady ST, Collins CM, Moy ML, Hart JE, Kang CM, Coull BA, Schwartz JD, Koutrakis P, Garshick E. Indoor and ambient black carbon and fine particulate matter associations with blood biomarkers in COPD patients. THE SCIENCE OF THE TOTAL ENVIRONMENT 2024; 927:171897. [PMID: 38522542 PMCID: PMC11090036 DOI: 10.1016/j.scitotenv.2024.171897] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/21/2024] [Revised: 03/20/2024] [Accepted: 03/20/2024] [Indexed: 03/26/2024]
Abstract
BACKGROUND Systemic inflammation contributes to cardiovascular risk and chronic obstructive pulmonary disease (COPD) pathophysiology. Associations between systemic inflammation and exposure to ambient fine particulate matter (PM ≤ 2.5 μm diameter; PM2.5), and black carbon (BC), a PM2.5 component attributable to traffic and other sources of combustion, infiltrating indoors are not well described. METHODS Between 2012 and 2017, COPD patients completed in-home air sampling over one-week intervals, up to four times (seasonally), followed by measurement of plasma biomarkers of systemic inflammation, C-reactive protein (CRP) and interleukin-6 (IL-6), and endothelial activation, soluble vascular adhesion molecule-1 (sVCAM-1). Ambient PM2.5, BC and sulfur were measured at a central site. The ratio of indoor/ambient sulfur in PM2.5, a surrogate for fine particle infiltration, was used to estimate indoor BC and PM2.5 of ambient origin. Linear mixed effects regression with a random intercept for each participant was used to assess associations between indoor and indoor of ambient origin PM2.5 and BC with each biomarker. RESULTS 144 participants resulting in 482 observations were included in the analysis. There were significant positive associations between indoor BC and indoor BC of ambient origin with CRP [%-increase per interquartile range (IQR);95 % CI (13.2 %;5.2-21.8 and 11.4 %;1.7-22.1, respectively)]. Associations with indoor PM2.5 and indoor PM2.5 of ambient origin were weaker. There were no associations with IL-6 or sVCAM-1. CONCLUSIONS In homes of patients with COPD without major sources of combustion, indoor BC is mainly attributable to the infiltration of ambient sources of combustion indoors. Indoor BC of ambient origin is associated with increases in systemic inflammation in patients with COPD, even when staying indoors.
Collapse
Affiliation(s)
- Cathy L Zhang
- Research and Development Service, VA Boston Healthcare System, 1400 VFW Parkway, West Roxbury, MA 02132, USA
| | - Jennifer R Maccarone
- Pulmonary, Allergy, Sleep, and Critical Care Medicine Section, Medical Service, VA Boston Healthcare System, 1400 VFW Parkway, West Roxbury, Boston, MA 02132, USA; The Pulmonary Center, Boston University School of Medicine, 72 East Concord Street, Boston, MA 02118, USA
| | - Stephanie T Grady
- Boston University School of Public Health, 715 Albany St, Boston, MA 02118, USA
| | - Christina M Collins
- Research and Development Service, VA Boston Healthcare System, 1400 VFW Parkway, West Roxbury, MA 02132, USA
| | - Marilyn L Moy
- Pulmonary, Allergy, Sleep, and Critical Care Medicine Section, Medical Service, VA Boston Healthcare System, 1400 VFW Parkway, West Roxbury, Boston, MA 02132, USA; Harvard Medical School, 25 Shattuck Street, Boston, MA 02115, USA
| | - Jaime E Hart
- Department of Environmental Health, Harvard T.H. Chan School of Public Health, 655 Huntington Ave, Boston, MA 02115, USA; Channing Division of Network Medicine, Department of Medicine, Brigham and Women's Hospital, 181 Longwood Avenue, Boston, MA 02115, USA; Harvard Medical School, 25 Shattuck Street, Boston, MA 02115, USA
| | - Choong-Min Kang
- Department of Environmental Health, Harvard T.H. Chan School of Public Health, 655 Huntington Ave, Boston, MA 02115, USA
| | - Brent A Coull
- Department of Environmental Health, Harvard T.H. Chan School of Public Health, 655 Huntington Ave, Boston, MA 02115, USA; Department of Biostatistics, Harvard T.H. Chan School of Public Health, 677 Huntington Ave, Boston, MA 02115, USA
| | - Joel D Schwartz
- Department of Environmental Health, Harvard T.H. Chan School of Public Health, 655 Huntington Ave, Boston, MA 02115, USA; Department of Epidemiology, Harvard T.H. Chan School of Public Health, 677 Huntington Ave, Boston, MA 02115, USA
| | - Petros Koutrakis
- Department of Environmental Health, Harvard T.H. Chan School of Public Health, 655 Huntington Ave, Boston, MA 02115, USA
| | - Eric Garshick
- Pulmonary, Allergy, Sleep, and Critical Care Medicine Section, Medical Service, VA Boston Healthcare System, 1400 VFW Parkway, West Roxbury, Boston, MA 02132, USA; Harvard Medical School, 25 Shattuck Street, Boston, MA 02115, USA.
| |
Collapse
|
44
|
Guzik TJ, Nosalski R, Maffia P, Drummond GR. Immune and inflammatory mechanisms in hypertension. Nat Rev Cardiol 2024; 21:396-416. [PMID: 38172242 DOI: 10.1038/s41569-023-00964-1] [Citation(s) in RCA: 82] [Impact Index Per Article: 82.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 11/20/2023] [Indexed: 01/05/2024]
Abstract
Hypertension is a global health problem, with >1.3 billion individuals with high blood pressure worldwide. In this Review, we present an inflammatory paradigm for hypertension, emphasizing the crucial roles of immune cells, cytokines and chemokines in disease initiation and progression. T cells, monocytes, macrophages, dendritic cells, B cells and natural killer cells are all implicated in hypertension. Neoantigens, the NLRP3 inflammasome and increased sympathetic outflow, as well as cytokines (including IL-6, IL-7, IL-15, IL-18 and IL-21) and a high-salt environment, can contribute to immune activation in hypertension. The activated immune cells migrate to target organs such as arteries (especially the perivascular fat and adventitia), kidneys, the heart and the brain, where they release effector cytokines that elevate blood pressure and cause vascular remodelling, renal damage, cardiac hypertrophy, cognitive impairment and dementia. IL-17 secreted by CD4+ T helper 17 cells and γδ T cells, and interferon-γ and tumour necrosis factor secreted by immunosenescent CD8+ T cells, exert crucial effector roles in hypertension, whereas IL-10 and regulatory T cells are protective. Effector mediators impair nitric oxide bioavailability, leading to endothelial dysfunction and increased vascular contractility. Inflammatory effector mediators also alter renal sodium and water balance and promote renal fibrosis. These mechanisms link hypertension with obesity, autoimmunity, periodontitis and COVID-19. A comprehensive understanding of the immune and inflammatory mechanisms of hypertension is crucial for safely and effectively translating the findings to clinical practice.
Collapse
Affiliation(s)
- Tomasz J Guzik
- Centre for Cardiovascular Sciences, University of Edinburgh, Edinburgh, UK.
- Department of Medicine and Omicron Medical Genomics Laboratory, Jagiellonian University, Collegium Medicum, Kraków, Poland.
- Africa-Europe Cluster of Research Excellence (CoRE) in Non-Communicable Diseases & Multimorbidity, African Research Universities Alliance ARUA & The Guild, Glasgow, UK.
| | - Ryszard Nosalski
- Centre for Cardiovascular Sciences, University of Edinburgh, Edinburgh, UK
| | - Pasquale Maffia
- Africa-Europe Cluster of Research Excellence (CoRE) in Non-Communicable Diseases & Multimorbidity, African Research Universities Alliance ARUA & The Guild, Glasgow, UK
- School of Infection & Immunity, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow, UK
- Department of Pharmacy, School of Medicine and Surgery, University of Naples Federico II, Naples, Italy
| | - Grant R Drummond
- Department of Microbiology, Anatomy, Physiology and Pharmacology, La Trobe University, Melbourne, Victoria, Australia
- Centre for Cardiovascular Biology and Disease Research, La Trobe University, Melbourne, Victoria, Australia
| |
Collapse
|
45
|
Branigan P, Duong YV, Abdulfattah AY, Sabu J, Mallappallil M, John S. Towards Optimal Cardiovascular Health: A Comprehensive Review of Preventive Strategies. Cureus 2024; 16:e60877. [PMID: 38910676 PMCID: PMC11192625 DOI: 10.7759/cureus.60877] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/22/2024] [Indexed: 06/25/2024] Open
Abstract
Heart disease remains a prominent global health concern, with cardiovascular disease (CVD) standing as a leading cause of death worldwide. Preventing heart disease not only decreases the risk of premature death but also mitigates complications like heart attacks, strokes, and arrhythmias, thereby enhancing overall health and quality of life. The economic burden of heart disease treatment highlights the importance of implementing preventive measures, such as lifestyle changes and early interventions, which can alleviate healthcare costs. These strategies, targeting risk factors like hypertension (HTN), diabetes mellitus (DM), dyslipidemia, and obesity, not only prevent heart disease but also reduce the risk of other health issues. Herein, this review covers various preventive measures, including dietary interventions, exercise, controlling HTN, DM, cholesterol, and weight, smoking cessation, and pharmacological interventions. By critically analyzing the guidelines and leveraging robust data alongside variations in recommendations, this review aims to elucidate effective primary prevention strategies for CVD.
Collapse
Affiliation(s)
- Philip Branigan
- Department of Cardiology, State University of New York Downstate Medical Center, Brooklyn, USA
| | - Y V Duong
- Department of Cardiology, University of Debrecen Medical School, Debrecen, HUN
| | - Ammar Y Abdulfattah
- Department of Internal Medicine, State University of New York Downstate Medical Center, Brooklyn, USA
| | - Jacob Sabu
- Department of Cardiology, State University of New York Downstate Health Sciences University, Brooklyn, USA
| | - Mary Mallappallil
- Department of Nephrology, State University of New York Downstate Medical Center, Brooklyn, USA
| | - Sabu John
- Department of Cardiology, State University of New York Downstate Medical Center, Brooklyn, USA
| |
Collapse
|
46
|
Sun H, Ma X, Ma H, Li S, Xia Y, Yao L, Wang Y, Pang X, Zhong J, Yao G, Liu X, Zhang M. High glucose levels accelerate atherosclerosis via NLRP3-IL/ MAPK/NF-κB-related inflammation pathways. Biochem Biophys Res Commun 2024; 704:149702. [PMID: 38422898 DOI: 10.1016/j.bbrc.2024.149702] [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: 02/03/2024] [Revised: 02/19/2024] [Accepted: 02/20/2024] [Indexed: 03/02/2024]
Abstract
BACKGROUND As a chronic inflammatory disease, diabetes mellitus (DM) contributes to the development of atherosclerosis (AS). However, how the NLRP3 inflammasome participates in diabetes-related AS remains unclear. Therefore, this study aimed to elucidate the mechanism through which NLRP3 uses high glucose (HG) levels to promote AS. METHODS Serum and coronary artery tissues were collected from coronary artery disease (CAD) patients with and without DM, respectively. The expression of NLRP3 was detected, and the effects of this inflammasome on diabetes-associated AS were evaluated using streptozotocin (STZ)-induced diabetic apoE-/- mice injected with Adenovirus-mediated NLRP3 interference (Ad-NLRP3i). To elucidate the potential mechanism involved, ox-LDL-irritated human aortic smooth muscle cells were divided into the control, high-glucose, Si-NC, and Si-NLRP3 groups to observe the changes induced by downregulating NLRP3 expression. For up-regulating NLRP3, control and plasmid contained NLRP3 were used. TNF-α, IL-1β, IL-6, IL-18, phosphorylated and total p38, JNK, p65, and IκBα expression levels were detected following the downregulation or upregulation of NLRP3 expression. RESULTS Patients with comorbid CAD and DM showed higher serum levels and expression of NLRP3 in the coronary artery than those with only CAD. Moreover, mice in the Ad-NLRP3i group showed markedly smaller and more stable atherosclerotic lesions compared to those in other DM groups. These mice had decreased inflammatory cytokine production and improved glucose tolerance, which demonstrated the substantial effects of NLRP3 in the progression of diabetes-associated AS. Furthermore, using the siRNA or plasmid to downregulate or upregulate NLRP3 expression in vitro altered cytokines and the MAPK/NF-κB pathway. CONCLUSIONS NLRP3 expression was significantly increased under hyperglycemia. Additionally, it accelerated AS by promoting inflammation via the IL/MAPK/NF-κB pathway.
Collapse
Affiliation(s)
- Hui Sun
- National Key Laboratory for Innovation and Transformation of Luobing Theory, The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education, Chinese National Health Commission and Chinese Academy of Medical Sciences, Department of Cardiology, Qilu Hospital of Shandong University, Jinan, China; Department of Cardiology, Qilu Hospital (Qingdao), Cheeloo College of Medicine, Shandong University, Qingdao, China
| | - Xiaotian Ma
- Department of Medicine Experimental Center, Qilu Hospital (Qingdao), Cheeloo College of Medicine, Shandong University, Qingdao, China
| | - Hong Ma
- Qingdao Branch of Shandong Public Health Clinical Center, Qingdao, China
| | - Shuen Li
- Department of Pathology, Qilu Hospital (Qingdao), Cheeloo College of Medicine, Shandong University, Qingdao, China
| | - Yan Xia
- Department of Pathology, The Affiliated Hospital of Qingdao University, Qingdao, 266000, Shandong, China
| | - Lijie Yao
- Department of Cardiology, Qilu Hospital (Qingdao), Cheeloo College of Medicine, Shandong University, Qingdao, China
| | - Yingcui Wang
- Department of Cardiology, Qilu Hospital (Qingdao), Cheeloo College of Medicine, Shandong University, Qingdao, China
| | - Xuelian Pang
- Department of Cardiology, Qilu Hospital (Qingdao), Cheeloo College of Medicine, Shandong University, Qingdao, China
| | - Jingquan Zhong
- National Key Laboratory for Innovation and Transformation of Luobing Theory, The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education, Chinese National Health Commission and Chinese Academy of Medical Sciences, Department of Cardiology, Qilu Hospital of Shandong University, Jinan, China; Department of Cardiology, Qilu Hospital (Qingdao), Cheeloo College of Medicine, Shandong University, Qingdao, China
| | - Guihua Yao
- National Key Laboratory for Innovation and Transformation of Luobing Theory, The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education, Chinese National Health Commission and Chinese Academy of Medical Sciences, Department of Cardiology, Qilu Hospital of Shandong University, Jinan, China; Department of Cardiology, Qilu Hospital (Qingdao), Cheeloo College of Medicine, Shandong University, Qingdao, China
| | - Xiaoling Liu
- National Key Laboratory for Innovation and Transformation of Luobing Theory, The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education, Chinese National Health Commission and Chinese Academy of Medical Sciences, Department of Cardiology, Qilu Hospital of Shandong University, Jinan, China.
| | - Mei Zhang
- National Key Laboratory for Innovation and Transformation of Luobing Theory, The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education, Chinese National Health Commission and Chinese Academy of Medical Sciences, Department of Cardiology, Qilu Hospital of Shandong University, Jinan, China.
| |
Collapse
|
47
|
Popescu AI, Rata AL, Barac S, Popescu R, Onofrei RR, Vlad C, Vlad D. Narrative Review of Biological Markers in Chronic Limb-Threatening Ischemia. Biomedicines 2024; 12:798. [PMID: 38672153 PMCID: PMC11047884 DOI: 10.3390/biomedicines12040798] [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: 03/10/2024] [Revised: 03/30/2024] [Accepted: 04/01/2024] [Indexed: 04/28/2024] Open
Abstract
BACKGROUND Chronic limb-threatening ischemia (CLTI), the advanced stage of peripheral arterial disease, is diagnosed in the presence of ischemic rest pain, non-healing ulcers, or gangrene. Several studies have demonstrated that inflammation and endothelial dysfunction are some of the main substrates of CLTI. METHODS A narrative review was conducted and reported according to PRISMA guidelines. Three databases were searched-Web of Science, Medline, and EMBASE-for the studies assessing CLTI and the biological markers related to it. RESULTS We included 22 studies, and all the markers identified (C-reactive protein, D-dimers, fibrinogen, cytokines, IL-6, TNF-α, ICAM-1 (Intracellular Adhesion Molecule-1), VCAM-1 (Vascular Cell Adhesion Molecule-1), neutrophile-to-lymphocytes ratio (NLR), IL-8, Pentraxin-3, neutrophil gelatinase-associated lipocalin (NGAL), calprotectin, E-selectin, P-selectin, neopterin, High-Mobility Group Box-1 protein (HGMB-1), Osteoprotegerin (OPG) and Sortilin) were positively associated with advanced CLTI, with major limb or major cardiovascular events in these patients. CONCLUSIONS All the studied markers had increased values in patients with CLTI, especially when associated with diabetes mellitus, proving a very important association between diabetes and major limb or cardiovascular events in these patients. There is a need for more studies to validate these markers in terms of diagnosis or prognosis in CLTI patients and in trying to find new medical strategies that target inflammation or endothelial dysfunction in these patients.
Collapse
Affiliation(s)
- Alexandra Ioana Popescu
- Pharmacology Department, Doctoral School, “Victor Babes” University of Medicine and Pharmacy, 300041 Timisoara, Romania;
| | - Andreea Luciana Rata
- Surgical Emergencies Department, “Victor Babes” University of Medicine and Pharmacy, 300041 Timisoara, Romania;
| | - Sorin Barac
- Vascular Surgery Department, “Victor Babes” University of Medicine and Pharmacy, 300041 Timisoara, Romania
| | - Roxana Popescu
- Cell and Molecular Biology Department, ”Victor Babes” University of Medicine and Pharmacy, 300041 Timisoara, Romania;
| | - Roxana Ramona Onofrei
- Department of Rehabilitation, Physical Medicine and Rheumatology, Research Center for Assessment of Human Motion, Functionality and Disability, ”Victor Babes” University of Medicine and Pharmacy, 300041 Timisoara, Romania;
| | - Cristian Vlad
- Pharmacology Department, “Victor Babes” University of Medicine and Pharmacy, 300041 Timisoara, Romania; (C.V.); (D.V.)
| | - Daliborca Vlad
- Pharmacology Department, “Victor Babes” University of Medicine and Pharmacy, 300041 Timisoara, Romania; (C.V.); (D.V.)
| |
Collapse
|
48
|
Tattersall MC, Jarjour NN, Busse PJ. Systemic Inflammation in Asthma: What Are the Risks and Impacts Outside the Airway? THE JOURNAL OF ALLERGY AND CLINICAL IMMUNOLOGY. IN PRACTICE 2024; 12:849-862. [PMID: 38355013 PMCID: PMC11219096 DOI: 10.1016/j.jaip.2024.02.004] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/23/2023] [Revised: 01/19/2024] [Accepted: 02/02/2024] [Indexed: 02/16/2024]
Abstract
Airway inflammation in asthma has been well recognized for several decades, with general agreement on its role in asthma pathogenesis, symptoms, propensity toward exacerbation, and decline in lung function. This has led to universal recommendation in asthma management guidelines to incorporate the use of inhaled corticosteroid as an anti-inflammatory therapy for all patients with persistent asthma symptoms. However, there has been limited attention paid to the presence and potential impact of systemic inflammation in asthma. Accumulating evidence from epidemiological observations and cohort studies points to a host of downstream organ dysfunction in asthma especially among patients with longstanding or more severe disease, frequent exacerbations, and underlying risk factors for organ dysfunction. Most studies to date have focused on cognitive impairment, depression/anxiety, metabolic syndrome, and cardiovascular abnormalities. In this review, we summarize some of the evidence demonstrating these abnormalities and highlight the proposed mechanisms and potential benefits of treatment in limiting these extrapulmonary abnormalities in patients with asthma. The goal of this commentary is to raise awareness of the importance of recognizing potential extrapulmonary conditions associated with systemic inflammation of asthma. This area of treatment of patients with asthma is a large unmet need.
Collapse
Affiliation(s)
- Matthew C Tattersall
- Department of Medicine, University of Wisconsin School of Medicine and Public Health, Madison, Wis.
| | - Nizar N Jarjour
- Department of Medicine, University of Wisconsin School of Medicine and Public Health, Madison, Wis
| | - Paula J Busse
- Department of Medicine, Division of Clinical Immunology, Icahn School of Medicine at Mount Sinai School of Medicine, New York, NY
| |
Collapse
|
49
|
Tattersall MC, Gangnon RE, Jarjour NN. Asthma and cardiovascular disease: embracing disease heterogeneity is required. Eur Respir J 2024; 63:2400469. [PMID: 38663972 DOI: 10.1183/13993003.00469-2024] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2024] [Accepted: 03/08/2024] [Indexed: 05/21/2025]
Affiliation(s)
- Matthew C Tattersall
- Department of Medicine, Division of Cardiology, University of Wisconsin School of Medicine and Public Health, Madison, WI, USA
| | - Ronald E Gangnon
- Department of Population Health Sciences, University of Wisconsin School of Medicine and Public Health, Madison, WI, USA
- Department of Biostatistics and Medical Informatics, University of Wisconsin-Madison, Madison, WI, USA
| | - Nizar N Jarjour
- Department of Biostatistics and Medical Informatics, University of Wisconsin-Madison, Madison, WI, USA
| |
Collapse
|
50
|
Posadas-Sánchez R, López-Uribe ÁR, Fragoso JM, Vargas-Alarcón G. Interleukin 6 polymorphisms are associated with cardiovascular risk factors in premature coronary artery disease patients and healthy controls of the GEA Mexican study. Exp Mol Pathol 2024; 136:104886. [PMID: 38290570 DOI: 10.1016/j.yexmp.2024.104886] [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: 02/11/2023] [Revised: 01/04/2024] [Accepted: 01/26/2024] [Indexed: 02/01/2024]
Abstract
BACKGROUND AND AIMS Interleukin-6 (IL-6) is an acute-phase protein that plays an important role in the inflammatory response, vascular inflammation, and atherosclerosis process. The study aimed to establish whether IL-6 gene polymorphisms and IL-6 concentrations are associated with premature coronary artery disease (pCAD) and cardiovascular risk factors. METHODS The IL-6 concentrations and the rs2069827, rs1800796, and rs1800795 IL-6 polymorphisms were determined in 1150 pCAD patients and 1083 healthy controls (coronary artery calcium equal to zero determined by tomography). RESULTS The IL-6 polymorphisms studied were not associated with pCAD, but they were associated with cardiovascular risk factors in patients and controls. In controls, under the dominant model, the rs1800795 C allele and the rs2069827 T allele were associated with a low risk of central obesity (OR = 0.401, p = 0.017 and OR = 0.577, p = 0.031, respectively), hypoalphalipoproteinemia (OR = 0.581, p = 0.027 and OR = 0.700, p = 0.014, respectively) and hypertriglyceridemia (OR = 0.575, p = 0.030 and OR = 0.728, p = 0.033, respectively). In pCAD, the rs1800795 C allele was associated with an increased risk of hypoalphalipoproteinemia (OR = 1.370, padditive = 0.025) and increased C-reactive protein (CRP) concentrations (OR = 1.491, padditive = 0.007). pCAD patients had significantly higher serum IL-6 concentrations compared to controls (p = 0.002). In the total population, individuals carrying the rs1800795 GC + CC genotypes had higher levels of IL-6 than carriers of the GG genotype (p = 0.025). In control individuals carrying the C allele (CG + CC), an inverse correlation was observed between IL-6 and HDL-cholesterol levels (p = 0.003). CONCLUSIONS In summary, the IL-6 polymorphisms were not associated with pCAD, however, they were associated with cardiovascular risk factors in pCAD patients and healthy controls. Individuals carrying the rs1800795 GC + CC genotypes had higher levels of IL-6 than carriers of the GG genotype.
Collapse
Affiliation(s)
| | - Ángel Rene López-Uribe
- Department of Endocrinology, Instituto Nacional de Cardiología Ignacio Chávez, Mexico City, Mexico
| | - José Manuel Fragoso
- Department of Molecular Biology, Instituto Nacional de Cardiología Ignacio Chávez, Mexico City, Mexico
| | - Gilberto Vargas-Alarcón
- Department of Molecular Biology, Instituto Nacional de Cardiología Ignacio Chávez, Mexico City, Mexico; Research Direction, Instituto Nacional de Cardiología Ignacio Chávez, Mexico City, Mexico.
| |
Collapse
|