1
|
Liu S, Wang M, Xu L, Deng D, Lu L, Tian J, Zhou D, Rui K. New insight into the role of SOCS family in immune regulation and autoimmune pathogenesis. J Adv Res 2025:S2090-1232(25)00313-3. [PMID: 40349956 DOI: 10.1016/j.jare.2025.05.020] [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: 11/20/2024] [Revised: 04/07/2025] [Accepted: 05/08/2025] [Indexed: 05/14/2025] Open
Abstract
BACKGROUND Suppressor of cytokine signaling (SOCS) proteins regulate signal transduction by interacting with cytokine receptors and signaling proteins and targeting associated proteins for degradation. Recent studies have demonstrated that the SOCS proteins serve as crucial inhibitors in cytokine signaling networks and play a pivotal role in both innate and adaptive immune responses. AIM OF REVIEW In this review, we aim to discuss recent advancements in understanding the complex functions of SOCS proteins in various immune cells, as well as the effects of SOCS proteins in human health and diseases. Increasing evidence indicates that SOCS proteins are frequently dysregulated in developing autoimmune diseases, suggesting that therapeutic targeting of SOCS proteins could provide clinical benefit. KEY SCIENTIFIC CONCEPTS OF REVIEW This review provides a comprehensive understanding of SOCS proteins in immune regulation and autoimmune pathogenesis, it also highlights the role of SOCS-related mimetic peptides in immunotherapy.
Collapse
Affiliation(s)
- Shiyi Liu
- Department of Laboratory Medicine, Affiliated Hospital of Jiangsu University, Zhenjiang, China; Department of Immunology, Jiangsu Key Laboratory of Laboratory Medicine, School of Medicine, Jiangsu University, Zhenjiang, China
| | - Mingwei Wang
- Department of Emergency, Affiliated People's Hospital of Jiangsu University, Zhenjiang, China
| | - Liangjie Xu
- Department of Cardiology, Affiliated Hospital of Jiangsu University, Zhenjiang, China
| | - Daihua Deng
- Department of Rheumatology, Mianyang Central Hospital, School of Medicine, University of Electronic Science and Technology of China, Mianyang, China
| | - Liwei Lu
- Department of Pathology and Shenzhen Institute of Research and Innovation, The University of Hong Kong, Chongqing International Institute for Immunology, China
| | - Jie Tian
- Department of Immunology, Jiangsu Key Laboratory of Laboratory Medicine, School of Medicine, Jiangsu University, Zhenjiang, China.
| | - Dongmei Zhou
- Department of Rheumatology and Immunology, Affiliated Hospital of Xuzhou Medical University, Xuzhou, China.
| | - Ke Rui
- Department of Laboratory Medicine, Affiliated Hospital of Jiangsu University, Zhenjiang, China.
| |
Collapse
|
2
|
Lin TL, Fan YH, Fan KS, Juan CK, Chen YJ, Wu CY. Cardiovascular disease risk in patients with psoriasis receiving biologics targeting TNF-α, IL-12/23, IL-17, and IL-23: A population-based retrospective cohort study. PLoS Med 2025; 22:e1004591. [PMID: 40245096 PMCID: PMC12052210 DOI: 10.1371/journal.pmed.1004591] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/05/2024] [Revised: 05/05/2025] [Accepted: 03/25/2025] [Indexed: 04/19/2025] Open
Abstract
BACKGROUND Psoriasis is associated with various cardiovascular diseases (CVDs). The aim of this study was to compare the risk of CVD in patients with psoriasis who were prescribed biologics or oral therapies, and to assess the association between different classes of biologics and CVD risk. METHODS AND FINDINGS This retrospective cohort study utilized the TriNetX Global Collaborative Network (2014-2025). Patients with psoriasis newly prescribed biologics (BIO-cohort) and those newly initiating oral anti-psoriatic drugs without biologic exposure (Non-BIO-cohort) were enrolled. A propensity score-matched analysis was conducted, accounting for age, sex, race, comorbidities, body mass index, serum lipid profile, and inflammatory marker levels. Cardiovascular risk was compared between the BIO- and Non-BIO-cohorts using Cox regression to calculate hazard ratios (HRs) with 95% confidence intervals (CIs). After matching, each cohort comprised 12,732 patients, with approximately 50% being female, a mean age of 57 years, and 55% identifying as White. The 5-year cumulative incidence of any CVDs was significantly lower in the BIO-cohort (10.68%; 95% CI [10.03%, 11.36%]) than in the Non-BIO-cohort (16.17%; 95% CI: [15.34%, 17.05%]) (p < 0.001). The BIO-cohort had attenuated risks of any CVDs (HR 0.621; 95% CI [0.571, 0.676]), cerebrovascular diseases (HR 0.616; 95% CI [0.519, 0.731]), arrhythmias (HR 0.632; 95% CI [0.565, 0.706]), inflammatory heart diseases (HR 0.566; 95% CI [0.360, 0.891]), ischemic heart diseases (HR 0.579; 95% CI [0.465, 0.721]), heart failure (HR 0.637; 95% CI [0.521, 0.780]), non-ischemic cardiomyopathy (HR 0.654; 95% CI [0.466, 0.918]), thrombotic disorders (HR 0.570; 95% CI [0.444, 0.733]), peripheral arterial occlusive diseases (HR 0.501; 95% CI [0.383, 0.656]), and major adverse cardiac events (HR 0.697; 95% CI [0.614, 0.792]). Receiving only anti-tumor necrosis factor (TNF)-α (HR 0.886; 95% CI [0.807, 0.973]), anti-interleukin (IL)-17 (HR 0.724; 95% CI [0.599, 0.875]), or anti-IL-23 (HR 0.739; 95% CI [0.598, 0.914]) was associated with reduced risks of any CVDs, whereas no significant association was observed for only anti-IL-12/23 (HR 0.915; 95% CI [0.742, 1.128]). This risk reduction remained consistent across various subgroups, including age (≤45 or >45 years), sex (male or female), regions of research data (the United States, Europe, Middle East and Africa, and Asia-Pacific), and comorbidities (psoriatic arthritis, hypertension, diabetes, hyperlipidemia, overweight or obesity). Eight sensitivity analyses, such as extending the washout period or tightening medication definitions, validated our findings. The main limitation of our study is the observational design, which can only establish associations, not causation. CONCLUSIONS Patients with psoriasis prescribed biologics exhibited a lower risk of CVDs versus those on oral therapy. Anti-TNF-α, anti-IL-17, and anti-IL-23 were associated with decreased cardiovascular hazards, while anti-IL-12/23 was not.
Collapse
Affiliation(s)
- Teng-Li Lin
- Department of Dermatology, Dalin Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, Chiayi, Taiwan
- Ph.D. Program of Interdisciplinary Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan
| | - Yi-Hsuan Fan
- Department of Pediatrics, Chung Shan Medical University Hospital, Taichung, Taiwan
| | - Kuo-Sheng Fan
- Department of Internal Medicine, Division of Chest Medicine, Dalin Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, Chiayi, Taiwan
| | - Chao-Kuei Juan
- Department of Dermatology, Taichung Veterans General Hospital, Taichung, Taiwan
| | - Yi-Ju Chen
- Department of Dermatology, Taichung Veterans General Hospital, Taichung, Taiwan
- Department of Post-Baccalaureate Medicine, College of Medicine, National Chung Hsing University, Taichung, Taiwan
- Faculty of Medicine and Institute of Clinical Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan
| | - Chun-Ying Wu
- Faculty of Medicine and Institute of Clinical Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan
- Institute of Biomedical Informatics, National Yang Ming Chiao Tung University, Taipei, Taiwan
- Division of Translational Research, Taipei Veterans General Hospital, Taipei, Taiwan
- College of Public Health, China Medical University, Taichung, Taiwan
| |
Collapse
|
3
|
Bartoloni E, Cacciapaglia F, Erre GL, Gremese E, Manfredi A, Piga M, Sakellariou G, Spinelli FR, Viapiana O, Atzeni F. Immunomodulation for accelerated atherosclerosis in rheumatoid arthritis and systemic lupus erythematosus. Autoimmun Rev 2025; 24:103760. [PMID: 39894242 DOI: 10.1016/j.autrev.2025.103760] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2024] [Revised: 01/26/2025] [Accepted: 01/27/2025] [Indexed: 02/04/2025]
Abstract
In the last decades, consisting evidence supported a close relationship between both innate and adaptive immune systems and the accelerated cardiovascular (CV) disease characterizing autoimmune diseases, such as systemic lupus erythematosus (SLE) and rheumatoid arthritis (RA). Indeed, several cell lines involved in the pathogenesis of these autoimmune diseases, such as macrophages and dendritic cells, as well as different T and B lymphocyte subsets, and inflammatory cytokines, have been demonstrated to be directly involved in the mechanisms underlying early atherosclerotic arterial wall damage. Traditional CV risk factors play a concomitant role but do not sufficiently account for the increased prevalence of CV disease in these patients. Indeed, the pathophysiological link between RA and SLE and atherosclerosis is based on complex inflammatory pathways that interconnect these conditions and may explain the significant morbidity and mortality rates demonstrated in these patients, with consequent significant negative effects on quality of life and long-term survival. Consequently, it is intriguing to hypothesize that immunosuppressive drugs commonly used in the treatment of these pathologies may also exert an immunomodulatory and anti-inflammatory effect in mitigating the atherosclerotic damage that has been demonstrated to occur early in the initial stages of the disease. Recognizing risk factors, predicting occurrences and early intervention to prevent CV disease development have emerged as critical objectives in RA and SLE treatment. In this review, we aimed to provide an updated overview of the atherogenic effects exerted by the immune and inflammatory pathways involved in the pathogenesis of RA and SLE. Moreover, we examined the available evidence which may support the potential effects of immunosuppressive therapies in reducing CV damage and, consequently, CV disease risk in these patients.
Collapse
Affiliation(s)
- Elena Bartoloni
- Rheumatology Unit, Department of Medicine and Surgery, University of Perugia, Italy
| | - Fabio Cacciapaglia
- Rheumatology Unit, Department of Precision and Regenerative Medicine and Ionian Area (DePReMeI), University of Bari, Bari, Italy; Department of Medicine and Surgery, LUM University "Giuseppe De Gennaro" Casamassima & Rheumatology Service "Miulli" General Hospital Acquaviva delle Fonti, Bari, Italy
| | - Gian Luca Erre
- Dipartimento di Scienze Mediche, Chirurgiche e Sperimentali, Università degli Studi di Sassari, Italy
| | - Elisa Gremese
- Rheumatology and Clinical Immunology, IRCCS Humanitas Research Hospital, Rozzano, Milan, Italy; Department of Biomedical Sciences, Humanitas University, Pieve Emanuele, Milan, Italy
| | - Andreina Manfredi
- University of Modena and Reggio Emilia,AUSL-IRCCS Reggio Emilia, Reggio Emilia, Italy
| | - Matteo Piga
- Rheumatology Unit, AOU Cagliari, Department of Medical Sciences and Public Health, University of Cagliari, Italy
| | - Garifallia Sakellariou
- Department of Internal Medicine and Therapeutics, University of Pavia, Istituti Clinici Scientifici Maugeri, Pavia, Italy
| | - Francesca Romana Spinelli
- Reumatology Unit, Department of Internal, Anesthesiological, and Cardiovascular Clinical Sciences, Sapienza University of Rome, Rome, Italy
| | - Ombretta Viapiana
- Rheumatology Unit, Department of Medicine, University and Azienda Ospedaliera Universitaria Integrata of Verona, Italy
| | - Fabiola Atzeni
- Rheumatology Unit, Department of Experimental and Internal Medicine, University of Messina, Italy.
| |
Collapse
|
4
|
Suur BE, Karadimou G, Willems CJJM, Bergman O, Lengquist M, Kronqvist M, Baumgartner R, Malin S, Gisterå A, Hansson GK, Mälarstig A, Hedin U, Ketelhuth DFJ, Matic L. PCSK6 ablation in blood circulating cells increases atherosclerotic burden, but improves plaque stability by activating Th17-smooth muscle cell modulatory axis. Vascul Pharmacol 2025; 159:107490. [PMID: 40097084 DOI: 10.1016/j.vph.2025.107490] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2024] [Revised: 03/02/2025] [Accepted: 03/14/2025] [Indexed: 03/19/2025]
Abstract
BACKGROUND Proprotein convertase subtilisins/kexins (PCSKs) have been implicated in cancers and cardiovascular disease. We have shown that PCSK6 is a key protease regulating smooth muscle cell (SMC)-mediated vascular remodeling, but also that it can be expressed by T cells and macrophages in atherosclerotic plaques. Whether PCSK6 regulates innate and adaptive immune responses in the context of vascular inflammation is still unknown. METHODS In this study, detailed immunophenotyping of constitutive Pcsk6-/- mice was performed. Bone marrow transplantation into high-cholesterol diet fed Ldlr-/- mice was used to investigate PCSK6-mediated immune effects in atherogenesis and plaque stability. RESULTS Compared to controls, Pcsk6-/- mice showed higher plasma levels of the chemoattractants CCL2 and CCCL3, and Th17 cytokines IL-17 A and IL-17F. Pcsk6 ablation led to increased naïve and effector-memory CD4+ and CD8+ cell numbers in the spleen, and increased release of IL-17 A, IFN-γ and IL-10 as well as proliferation by spleenocytes in vitro. Lack of Pcsk6 also affected innate immunity as macrophages from Pcsk6-/- mice secreted more cytokines, including TNF-α, CCL2, IL-6 and IL-10 upon LPS stimulation in vitro, and were more prone to oxLDL uptake. In line with a pro-inflammatory phenotype, Pcsk6-/-➔Ldlr-/- transplanted mice presented a higher atherosclerotic plaque burden compared to Ldlr-/- receiving control bone marrow. Although larger, Pcsk6-/-➔Ldlr-/- plaques showed increased stability features, including collagen deposition and SMC presence coinciding with significantly increased local levels of the fibrogenic cytokine IL-17. CONCLUSIONS Global Pcsk6 ablation leads to the activation of both adaptive and innate immune systems. Interestingly, Pcsk6-/- ablation in bone marrow of hyperlipidemic mice revealed its dual role in atherogenesis, activating a Th17-SMC modulatory axis that promotes plaque stability, despite increased atherosclerotic burden.
Collapse
Affiliation(s)
- Bianca E Suur
- Department of Molecular Medicine and Surgery, Karolinska Institutet and Karolinska University Hospital, Sweden; Steno Diabetes Center Aarhus, Aarhus University Hospital, Aarhus, Denmark; Department of Biomedicine, Aarhus University, Aarhus, Denmark
| | - Glykeria Karadimou
- Department of Molecular Medicine and Surgery, Karolinska Institutet and Karolinska University Hospital, Sweden
| | - Colin J J M Willems
- Department of Molecular Medicine and Surgery, Karolinska Institutet and Karolinska University Hospital, Sweden
| | - Otto Bergman
- Department of Molecular Medicine and Surgery, Karolinska Institutet and Karolinska University Hospital, Sweden; Department of Medicine, Center for Molecular Medicine, Karolinska Institutet, Sweden
| | - Mariette Lengquist
- Department of Molecular Medicine and Surgery, Karolinska Institutet and Karolinska University Hospital, Sweden
| | - Malin Kronqvist
- Department of Molecular Medicine and Surgery, Karolinska Institutet and Karolinska University Hospital, Sweden
| | - Roland Baumgartner
- Department of Medicine, Center for Molecular Medicine, Karolinska Institutet, Sweden
| | - Stephen Malin
- Department of Medicine, Center for Molecular Medicine, Karolinska Institutet, Sweden
| | - Anton Gisterå
- Department of Medicine, Center for Molecular Medicine, Karolinska Institutet, Sweden
| | - Göran K Hansson
- Department of Medicine, Center for Molecular Medicine, Karolinska Institutet, Sweden
| | - Anders Mälarstig
- Department of Medicine, Center for Molecular Medicine, Karolinska Institutet, Sweden
| | - Ulf Hedin
- Department of Molecular Medicine and Surgery, Karolinska Institutet and Karolinska University Hospital, Sweden
| | - Daniel F J Ketelhuth
- Department of Medicine, Center for Molecular Medicine, Karolinska Institutet, Sweden; Department of Molecular Medicine, University of Southern Denmark, Denmark
| | - Ljubica Matic
- Department of Molecular Medicine and Surgery, Karolinska Institutet and Karolinska University Hospital, Sweden.
| |
Collapse
|
5
|
Liu Q, Yang R, Wang D, Liu Q. Role of low-density cholesterol and Interleukin-17 interaction in breast cancer pathogenesis and treatment. Cell Biol Int 2025; 49:139-153. [PMID: 39318044 DOI: 10.1002/cbin.12250] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2024] [Revised: 09/02/2024] [Accepted: 09/17/2024] [Indexed: 09/26/2024]
Abstract
Breast cancer (BC) has become the most prevalent cancer worldwide, and further research is being conducted to deepen our understanding of its pathogenesis and treatment. Lipid metabolism disorder is a significant alteration in cancer cells, and the investigation into the role of Interleukin-17 (IL-17) in malignant tumors has emerged as a research focus in recent years. Thus, exploring changes in lipid metabolism and inflammatory factors in BC cells is crucial in identifying potential therapeutic targets. This article summarizes the progress made in the research on the main low-density cholesterol (LDL) transporter and IL-17 in lipid metabolism, and their potential involvement in the development of BC. The article aims to establish a theoretical foundation for the development of BC-related therapies.
Collapse
Affiliation(s)
- Qingqing Liu
- Guangdong Provincial Hospital of Chinese Medicine, Guangdong Provincial Hospital of Chinese Medicine-Zhuhai Hospital, The 2nd Clinical School of Medicine, Guangzhou University of Chinese Medicine, Guangdong, 510120, China
| | - Rongyuan Yang
- Guangdong Provincial Hospital of Chinese Medicine, Guangdong Provincial Hospital of Chinese Medicine-Zhuhai Hospital, The 2nd Clinical School of Medicine, Guangzhou University of Chinese Medicine, Guangdong, 510120, China
| | - Dawei Wang
- Guangdong Provincial Hospital of Chinese Medicine, Guangdong Provincial Hospital of Chinese Medicine-Zhuhai Hospital, The 2nd Clinical School of Medicine, Guangzhou University of Chinese Medicine, Guangdong, 510120, China
- The 1st Affiliated Hospital of Guangzhou University of Traditional Chinese Medicine, Guangdong, 510405, China
| | - Qing Liu
- Guangdong Provincial Hospital of Chinese Medicine, Guangdong Provincial Hospital of Chinese Medicine-Zhuhai Hospital, The 2nd Clinical School of Medicine, Guangzhou University of Chinese Medicine, Guangdong, 510120, China
| |
Collapse
|
6
|
Cheng WH, Wang Y. Inflammatory Pathways in Coronary Artery Disease: Which Ones to Target for Secondary Prevention? Cells 2025; 14:153. [PMID: 39936945 PMCID: PMC11817712 DOI: 10.3390/cells14030153] [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: 01/02/2025] [Revised: 01/14/2025] [Accepted: 01/17/2025] [Indexed: 02/13/2025] Open
Abstract
Coronary artery disease (CAD), the build-up of atherosclerotic plaques on the wall of blood vessels, causes adverse cardiovascular events. Secondary prevention focuses on treating patients with existing plaques to prevent disease progression. Recent studies have shown that inflammation is an independent risk factor that drives disease progression, and targeting inflammation could be an effective therapeutic strategy for secondary prevention. In this review, we highlighted the roles of several inflammatory pathways in rupture and erosion, two major processes through which established plaques lead to adverse cardiovascular events. In the past 15 years, numerous clinical trials have tested the therapeutic potential of targeting these pathways, including neutralizing inflammatory cytokines and blocking signaling transduction of the inflammatory pathways. Only colchicine was approved for clinical use in patients with CAD. This is primarily due to the multifaceted roles of inflammatory pathways in disease progression. Commonly used pre-clinical models provided robust information for the onset of early disease but limited understanding of the inflammatory network in established plaques. This review will summarize lessons learned from successful and failed clinical trials to advocate for assessing inflammation in established plaques before designing therapeutics for secondary prevention.
Collapse
Affiliation(s)
- Wan-Hei Cheng
- Department of Pathology and Laboratory Medicine, Faculty of Medicine, University of British Columbia, Vancouver, BC V6T 1Z7, Canada;
- Centre for Heart Lung Innovation, University of British Columbia, Vancouver, BC V6Z 1Y6, Canada
| | - Ying Wang
- Department of Pathology and Laboratory Medicine, Faculty of Medicine, University of British Columbia, Vancouver, BC V6T 1Z7, Canada;
- Centre for Heart Lung Innovation, University of British Columbia, Vancouver, BC V6Z 1Y6, Canada
| |
Collapse
|
7
|
Mapuskar KA, London B, Zacharias ZR, Houtman JC, Allen BG. Immunometabolism in the Aging Heart. J Am Heart Assoc 2025; 14:e039216. [PMID: 39719411 PMCID: PMC12054428 DOI: 10.1161/jaha.124.039216] [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: 10/02/2024] [Accepted: 11/19/2024] [Indexed: 12/26/2024]
Abstract
Structural, functional, and molecular-level changes in the aging heart are influenced by a dynamic interplay between immune signaling and cellular metabolism that is referred to as immunometabolism. This review explores the crosstalk between cellular metabolic pathways including glycolysis, oxidative phosphorylation, fatty acid metabolism, and the immune processes that govern cardiac aging. With a rapidly aging population that coincides with increased cardiovascular risk and cancer incidence rates, understanding the immunometabolic underpinnings of cardiac aging provides a foundation for identifying therapeutic targets to mitigate cardiac dysfunction. Aging alters the immune environment of the heart by concomitantly driving the changes in immune cell metabolism, mitochondrial dysfunction, and redox signaling. Shifts in these metabolic pathways exacerbate inflammation and impair tissue repair, creating a vicious cycle that accelerates cardiac functional decline. Treatment with cancer therapy further complicates this landscape, as aging-associated immunometabolic disruptions augment the susceptibility to cardiotoxicity. The current review highlights therapeutic strategies that target the immunometabolic axis to alleviate cardiac aging pathologies. Interventions include modulating metabolic intermediates, improving mitochondrial function, and leveraging immune signaling pathways to restore cardiac health. Advances in immunometabolism thus hold significant potential for translating preclinical findings into therapies that improve the quality of life for the aging population and underscore the need for approaches that address the immunometabolic mechanisms of cardiac aging, providing a framework for future research.
Collapse
Affiliation(s)
- Kranti A. Mapuskar
- Department of Radiation OncologyUniversity of Iowa Hospitals and Clinic, University of Iowa HealthcareIowa CityIAUSA
- Holden Comprehensive Cancer Center, Carver College of MedicineUniversity of Iowa Hospitals and Clinic, University of Iowa HealthcareIowa CityIAUSA
| | - Barry London
- Holden Comprehensive Cancer Center, Carver College of MedicineUniversity of Iowa Hospitals and Clinic, University of Iowa HealthcareIowa CityIAUSA
- Department of Internal MedicineUniversity of Iowa Hospitals and Clinic, University of Iowa HealthcareIowa CityIAUSA
| | - Zeb R. Zacharias
- Holden Comprehensive Cancer Center, Carver College of MedicineUniversity of Iowa Hospitals and Clinic, University of Iowa HealthcareIowa CityIAUSA
- Human Immunology CoreUniversity of Iowa Hospitals and Clinic, University of Iowa HealthcareIowa CityIAUSA
| | - Jon C.D. Houtman
- Holden Comprehensive Cancer Center, Carver College of MedicineUniversity of Iowa Hospitals and Clinic, University of Iowa HealthcareIowa CityIAUSA
- Human Immunology CoreUniversity of Iowa Hospitals and Clinic, University of Iowa HealthcareIowa CityIAUSA
- Department of Microbiology and ImmunologyUniversity of Iowa Hospitals and Clinic, University of Iowa HealthcareIowa CityIAUSA
| | - Bryan G. Allen
- Department of Radiation OncologyUniversity of Iowa Hospitals and Clinic, University of Iowa HealthcareIowa CityIAUSA
- Holden Comprehensive Cancer Center, Carver College of MedicineUniversity of Iowa Hospitals and Clinic, University of Iowa HealthcareIowa CityIAUSA
| |
Collapse
|
8
|
Cho H, Kim YJ, Moon IJ, Lee WJ, Won CH, Lee MW, Chang SE, Jung JM. Risk of major adverse cardiovascular events and all-cause mortality among patients with psoriatic disease treated with tumor necrosis factor-α and interleukin-12/23 inhibitors: a nationwide population-based cohort study in Korea. J DERMATOL TREAT 2024; 35:2321194. [PMID: 38403279 DOI: 10.1080/09546634.2024.2321194] [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: 12/17/2023] [Accepted: 02/13/2024] [Indexed: 02/27/2024]
Abstract
PURPOSE Few studies have investigated the impact of biologics on the risk of major adverse cardiovascular events (MACEs) among Korean patients with psoriatic diseases. We compared the risk of MACEs and all-cause mortality among patients with psoriatic disease treated with tumor necrosis factor (TNF)-α and interleukin (IL)-12/23 inhibitors in Korea. METHODS Patients with psoriatic disease prescribed with TNF-α and IL-12/23 inhibitors since 2016 were selected from the Korean National Health Insurance Service (NHIS) Database. Follow-up data for MACEs and all-cause mortality between 2016 and 2020 were collected. A total of 2886 individuals were included, including 1987 IL-12/23 inhibitor users and 899 TNF-α inhibitor users. RESULTS Compared with IL-12/23 inhibitor users, TNF-α inhibitor users had a higher prevalence of dyslipidemia and a significantly higher risk of all-cause mortality but not MACE. After controlling for age, female TNF-α inhibitor users had a significantly increased risk of all-cause mortality. Meanwhile, after controlling for sex, TNF-α inhibitor users aged 60 years or older demonstrated a significantly elevated risk of all-cause mortality. In conclusion, No statistically significant difference in MACE risk was observed between patients who used TNF-α and IL-12/23 inhibitors. Nevertheless, the use of IL-12/23 inhibitors, especially among older and female patients, resulted in a lower overall mortality.
Collapse
Affiliation(s)
- Hyesoo Cho
- Department of Dermatology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea
| | - Ye-Jee Kim
- Department of Clinical Epidemiology and Biostatistics, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea
| | - Ik Jun Moon
- Department of Dermatology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea
| | - Woo Jin Lee
- Department of Dermatology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea
| | - Chong Hyun Won
- Department of Dermatology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea
| | - Mi Woo Lee
- Department of Dermatology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea
| | - Sung Eun Chang
- Department of Dermatology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea
| | - Joon Min Jung
- Department of Dermatology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea
| |
Collapse
|
9
|
Juhasz V, Charlier FT, Zhao TX, Tsiantoulas D. Targeting the adaptive immune continuum in atherosclerosis and post-MI injury. Atherosclerosis 2024; 399:118616. [PMID: 39546915 DOI: 10.1016/j.atherosclerosis.2024.118616] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/22/2024] [Revised: 09/04/2024] [Accepted: 09/24/2024] [Indexed: 11/17/2024]
Abstract
Atherosclerotic disease is a cholesterol-rich lipoprotein particle-driven disease resulting in the formation of atherosclerotic plaques in large and medium size arteries. Rupture or erosion of atherosclerotic plaques can trigger the formation of a thrombus causing the obstruction of the blood flow in the coronary artery and thereby leading to myocardial infarction (MI). Inflammation is a crucial pillar of the mechanisms leading to atherosclerosis and governing the cardiac repair post-MI. Dissecting the complex and sophisticated networks of the immune responses underlying the formation of atherosclerotic plaques and affecting the healing of the heart after MI will allow the designing of highly precise immunomodulatory therapies for these settings. Notably, MI also accelerates atherosclerosis via modulating the response of the immune system. Therefore, for the identification of effective and safe therapeutic targets, it is critical to consider the inflammatory continuum that interconnects the two pathologies and identify immunomodulatory strategies that confer a protective effect in both settings or at least, affect each pathology independently. Adaptive immunity, which consists of B and T lymphocytes, is a major regulator of atherosclerosis and post-MI cardiac repair. Here, we review and discuss the effect of potential adaptive immunity-targeting therapies, such as cell-depleting therapies, in atherosclerosis and post-MI cardiac injury.
Collapse
Affiliation(s)
- Viktoria Juhasz
- Department of Laboratory Medicine, Medical University of Vienna, Vienna, Austria
| | - Fiona T Charlier
- Division of Cardiovascular Medicine, Department of Medicine, University of Cambridge, Cambridge, United Kingdom
| | - Tian X Zhao
- Division of Cardiovascular Medicine, Department of Medicine, University of Cambridge, Cambridge, United Kingdom; Department of Cardiology, Royal Papworth Hospital NHS Trust, Cambridge, United Kingdom
| | | |
Collapse
|
10
|
Buch MH, Mallat Z, Dweck MR, Tarkin JM, O'Regan DP, Ferreira V, Youngstein T, Plein S. Current understanding and management of cardiovascular involvement in rheumatic immune-mediated inflammatory diseases. Nat Rev Rheumatol 2024; 20:614-634. [PMID: 39232242 DOI: 10.1038/s41584-024-01149-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/22/2024] [Indexed: 09/06/2024]
Abstract
Immune-mediated inflammatory diseases (IMIDs) are a spectrum of disorders of overlapping immunopathogenesis, with a prevalence of up to 10% in Western populations and increasing incidence in developing countries. Although targeted treatments have revolutionized the management of rheumatic IMIDs, cardiovascular involvement confers an increased risk of mortality and remains clinically under-recognized. Cardiovascular pathology is diverse across rheumatic IMIDs, ranging from premature atherosclerotic cardiovascular disease (ASCVD) to inflammatory cardiomyopathy, which comprises myocardial microvascular dysfunction, vasculitis, myocarditis and pericarditis, and heart failure. Epidemiological and clinical data imply that rheumatic IMIDs and associated cardiovascular disease share common inflammatory mechanisms. This concept is strengthened by emergent trials that indicate improved cardiovascular outcomes with immune modulators in the general population with ASCVD. However, not all disease-modifying therapies that reduce inflammation in IMIDs such as rheumatoid arthritis demonstrate equally beneficial cardiovascular effects, and the evidence base for treatment of inflammatory cardiomyopathy in patients with rheumatic IMIDs is lacking. Specific diagnostic protocols for the early detection and monitoring of cardiovascular involvement in patients with IMIDs are emerging but are in need of ongoing development. This Review summarizes current concepts on the potentially targetable inflammatory mechanisms of cardiovascular pathology in rheumatic IMIDs and discusses how these concepts can be considered for the diagnosis and management of cardiovascular involvement across rheumatic IMIDs, with an emphasis on the potential of cardiovascular imaging for risk stratification, early detection and prognostication.
Collapse
Affiliation(s)
- Maya H Buch
- Centre for Musculoskeletal Research, Division of Musculoskeletal & Dermatological Sciences, Faculty of Biology, Medicine & Health, University of Manchester, Manchester, UK.
- NIHR Manchester Biomedical Research Centre, Manchester University NHS Foundation Trust, Manchester Academic Health Science Centre, Manchester, UK.
| | - Ziad Mallat
- Section of Cardiorespiratory Medicine, Victor Phillip Dahdaleh Heart & Lung Research Institute, University of Cambridge, Cambridge, UK
| | - Marc R Dweck
- Centre for Cardiovascular Science, Chancellors Building, Little France Crescent, University of Edinburgh, Edinburgh, UK
| | - Jason M Tarkin
- Section of Cardiorespiratory Medicine, Victor Phillip Dahdaleh Heart & Lung Research Institute, University of Cambridge, Cambridge, UK
| | - Declan P O'Regan
- MRC Laboratory of Medical Sciences, Imperial College London, London, UK
| | - Vanessa Ferreira
- Division of Cardiovascular Medicine, Radcliffe Department of Medicine, University of Oxford, Oxford, UK
| | - Taryn Youngstein
- National Heart & Lung Institute, Imperial College London, London, UK
- Department of Rheumatology, Hammersmith Hospital, Imperial College Healthcare NHS Trust, London, UK
| | - Sven Plein
- Biomedical Imaging Science Department, Leeds Institute of Cardiovascular and Metabolic Medicine, University of Leeds, Leeds, UK
- School of Biomedical Engineering and Imaging Sciences, Kings College London, London, UK
| |
Collapse
|
11
|
de Oliveira WV, Giarola LTP, Ferreira LGR, Schettini IVG, Turani SD, de Oliveira AR, Marinho MAS, Pinto SWL, Barros-Pinheiro M, de Figueiredo RC, Rios DRA. Inflammation and all-cause mortality in patients undergoing peritoneal dialysis. EINSTEIN-SAO PAULO 2024; 22:eAO0627. [PMID: 39140572 PMCID: PMC11323835 DOI: 10.31744/einstein_journal/2024ao0627] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2023] [Accepted: 10/08/2023] [Indexed: 08/15/2024] Open
Abstract
OBJECTIVE This study aimed to evaluate inflammatory biomarkers in patients undergoing peritoneal dialysis and investigate their association with all-cause mortality or transfer to hemodialysis. METHODS This prospective cohort study included 43 patients undergoing peritoneal dialysis. Plasma levels of cytokines were measured using flow cytometry and capture enzyme-linked immunosorbent assay. Biomarkers were categorized based on their respective median values. Survival analysis was conducted using the Kaplan-Meier estimator, considering two outcomes: all-cause mortality and transfer to hemodialysis. RESULTS After adjusting for confounding factors, plasma levels above the median of the levels of CCL2 and plasma, as well as below the median of TNF-α, and the median of dialysate IL-17 levels, were associated with an increased risk of experiencing the specified outcomes after approximately 16 months of follow-up. CONCLUSION These findings suggest that inflammatory biomarkers may be a valuable tool for predicting all-cause mortality and transfer to hemodialysis in patients undergoing peritoneal dialysis.
Collapse
Affiliation(s)
| | - Luciane Teixeira Passos Giarola
- Department of Mathematics and StatisticsUniversidade de São João del-ReiSão João del-ReiMGBrazil Department of Mathematics and Statistics, Universidade de São João del-Rei, São João del-Rei, MG, Brazil.
| | | | | | - Sylvia Dias Turani
- Universidade de São João del-ReiDivinópolisMGBrazil Universidade de São João del-Rei, Divinópolis, MG, Brazil.
| | - Arlindo Ribeiro de Oliveira
- Nephrology CenterComplexo de Saúde São João de DeusDivinópolisMGBrazil Nephrology Center, Complexo de Saúde São João de Deus, Divinópolis, MG, Brazil.
| | - Maria Aparecida Silva Marinho
- Nephrology CenterComplexo de Saúde São João de DeusDivinópolisMGBrazil Nephrology Center, Complexo de Saúde São João de Deus, Divinópolis, MG, Brazil.
| | - Sérgio Wyton Lima Pinto
- Nephrology CenterComplexo de Saúde São João de DeusDivinópolisMGBrazil Nephrology Center, Complexo de Saúde São João de Deus, Divinópolis, MG, Brazil.
| | - Melina Barros-Pinheiro
- Universidade de São João del-ReiDivinópolisMGBrazil Universidade de São João del-Rei, Divinópolis, MG, Brazil.
| | | | - Danyelle Romana Alves Rios
- Universidade de São João del-ReiDivinópolisMGBrazil Universidade de São João del-Rei, Divinópolis, MG, Brazil.
| |
Collapse
|
12
|
Chajadine M, Laurans L, Radecke T, Mouttoulingam N, Al-Rifai R, Bacquer E, Delaroque C, Rytter H, Bredon M, Knosp C, Vilar J, Fontaine C, Suffee N, Vandestienne M, Esposito B, Dairou J, Launay JM, Callebert J, Tedgui A, Ait-Oufella H, Sokol H, Chassaing B, Taleb S. Harnessing intestinal tryptophan catabolism to relieve atherosclerosis in mice. Nat Commun 2024; 15:6390. [PMID: 39080345 PMCID: PMC11289133 DOI: 10.1038/s41467-024-50807-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2023] [Accepted: 07/22/2024] [Indexed: 08/02/2024] Open
Abstract
Tryptophan (Trp) is an essential amino acid, whose metabolism is a key gatekeeper of intestinal homeostasis. Yet, its systemic effects, particularly on atherosclerosis, remain unknown. Here we show that high-fat diet (HFD) increases the activity of intestinal indoleamine 2, 3-dioxygenase 1 (IDO), which shifts Trp metabolism from the production of microbiota-derived indole metabolites towards kynurenine production. Under HFD, the specific deletion of IDO in intestinal epithelial cells leads to intestinal inflammation, impaired intestinal barrier, augmented lesional T lymphocytes and atherosclerosis. This is associated with an increase in serotonin production and a decrease in indole metabolites, thus hijacking Trp for the serotonin pathway. Inhibition of intestinal serotonin production or supplementation with indole derivatives alleviates plaque inflammation and atherosclerosis. In summary, we uncover a pivotal role of intestinal IDO in the fine-tuning of Trp metabolism with systemic effects on atherosclerosis, paving the way for new therapeutic strategies to relieve gut-associated inflammatory diseases.
Collapse
Affiliation(s)
- Mouna Chajadine
- Université Paris Cité, Inserm, PARCC, F-75015, Paris, France
| | | | - Tobias Radecke
- Université Paris Cité, Inserm, PARCC, F-75015, Paris, France
| | | | - Rida Al-Rifai
- Université Paris Cité, Inserm, PARCC, F-75015, Paris, France
| | - Emilie Bacquer
- Université Paris Cité, Inserm, PARCC, F-75015, Paris, France
| | - Clara Delaroque
- Microbiome-Host interactions, Institut Pasteur, Université Paris Cité, INSERM U1306, Paris, France
- INSERM U1016, Team "Mucosal microbiota in chronic inflammatory diseases", CNRS UMR10 8104, Université Paris Cité, Paris, France
| | - Héloïse Rytter
- Microbiome-Host interactions, Institut Pasteur, Université Paris Cité, INSERM U1306, Paris, France
- INSERM U1016, Team "Mucosal microbiota in chronic inflammatory diseases", CNRS UMR10 8104, Université Paris Cité, Paris, France
| | - Marius Bredon
- Sorbonne Université, INSERM, Centre de Recherche Saint-Antoine, CRSA, AP-HP, Saint Antoine Hospital, Gastroenterology Department, F-75012, Paris, France
- Paris Centre for Microbiome Medicine (PaCeMM) FHU, Paris, France
| | - Camille Knosp
- Université Paris Cité, Inserm, PARCC, F-75015, Paris, France
| | - José Vilar
- Université Paris Cité, Inserm, PARCC, F-75015, Paris, France
| | - Coralie Fontaine
- Inserm U1297, Institut des Maladies Métaboliques et Cardiovasculaires, Université de Toulouse, BP 84225, 31 432 Toulouse cedex 04, cedex, France
| | - Nadine Suffee
- Université Paris Cité, Inserm, PARCC, F-75015, Paris, France
| | | | - Bruno Esposito
- Université Paris Cité, Inserm, PARCC, F-75015, Paris, France
| | - Julien Dairou
- Université Paris cité, CNRS, Laboratoire de Chimie et de Biochimie Pharmacologiques et Toxicologiques, F-75006 Paris, France. 45 rue des Saints Pères, 75006, Paris, France
| | - Jean Marie Launay
- Assistance Publique Hôpitaux de Paris, Service de Biochimie and INSERM U942, Hôpital Lariboisière, Paris, France
| | - Jacques Callebert
- Assistance Publique Hôpitaux de Paris, Service de Biochimie and INSERM U942, Hôpital Lariboisière, Paris, France
| | - Alain Tedgui
- Université Paris Cité, Inserm, PARCC, F-75015, Paris, France
| | | | - Harry Sokol
- Sorbonne Université, INSERM, Centre de Recherche Saint-Antoine, CRSA, AP-HP, Saint Antoine Hospital, Gastroenterology Department, F-75012, Paris, France
- Paris Centre for Microbiome Medicine (PaCeMM) FHU, Paris, France
- Université Paris-Saclay, INRAe, AgroParisTech, Micalis institute, Jouy-en-Josas, France Université Paris-Saclay, INRAe, AgroParisTech, Micalis institute, Jouy-en-Josas, France
| | - Benoit Chassaing
- Microbiome-Host interactions, Institut Pasteur, Université Paris Cité, INSERM U1306, Paris, France
- INSERM U1016, Team "Mucosal microbiota in chronic inflammatory diseases", CNRS UMR10 8104, Université Paris Cité, Paris, France
| | - Soraya Taleb
- Université Paris Cité, Inserm, PARCC, F-75015, Paris, France.
| |
Collapse
|
13
|
Kobayashi R, Hashida N. Overview of Cytomegalovirus Ocular Diseases: Retinitis, Corneal Endotheliitis, and Iridocyclitis. Viruses 2024; 16:1110. [PMID: 39066272 PMCID: PMC11281654 DOI: 10.3390/v16071110] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2024] [Revised: 06/24/2024] [Accepted: 06/29/2024] [Indexed: 07/28/2024] Open
Abstract
Cytomegalovirus (CMV) infection is a significant clinical concern in newborns, immunocompromised patients with acquired immunodeficiency syndrome (AIDS), and patients undergoing immunosuppressive therapy or chemotherapy. CMV infection affects many organs, such as the lungs, digestive organs, the central nerve system, and eyes. In addition, CMV infection sometimes occurs in immunocompetent individuals. CMV ocular diseases includes retinitis, corneal endotheliitis, and iridocyclitis. CMV retinitis often develops in infected newborns and immunocompromised patients. CMV corneal endotheliitis and iridocyclitis sometimes develop in immunocompetent individuals. Systemic infections and CMV ocular diseases often require systemic treatment in addition to topical treatment.
Collapse
Affiliation(s)
| | - Noriyasu Hashida
- Department of Ophthalmology, Graduate School of Medicine, Osaka University, Osaka 565-0871, Japan
| |
Collapse
|
14
|
Miao Y, Yan T, Liu J, Zhang C, Yan J, Xu L, Zhang N, Zhang X. Meta-analysis of the association between interleukin-17 and ischemic cardiovascular disease. BMC Cardiovasc Disord 2024; 24:252. [PMID: 38750443 PMCID: PMC11097571 DOI: 10.1186/s12872-024-03897-w] [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: 11/16/2023] [Accepted: 04/22/2024] [Indexed: 05/18/2024] Open
Abstract
BACKGROUND Interleukin-17 (IL-17) has been hypothesized to be involved in ischemic cardiovascular disease (ICVD). However, the association of IL-17 with ICVD remained unclear. The aim of this study was to systematically analyze the available evidence regarding the association between IL-17 and ICVD. METHODS We searched the PubMed, Web of Science, Cochrane Library, and Embase databases up to October 2023 to identify publications on the association between IL-17 and ICVD. The merged results were analyzed using a random effects model for meta-analysis and subgroup analysis. RESULTS A total of 955 publications were initially identified in our search and screened; six studies were eventually included in the analysis. The average age of study participants was 60.3 ± 12.6 years and 65.5% were men. There was a high degree of heterogeneity among studies. The results showed that IL-17 level were higher in the case group than those in the control group (standardized mean difference, SMD = 1.60, 95% confidence interval (95% CI): 0.53-2.66, P = 0.003). In sensitivity analysis, the merged results showed good robustness. Additionally, subgroup analysis showed that race and ethnicity, sample size, and detection methods were significant factors influencing heterogeneity in the published studies. CONCLUSION Our finding revealed that increased IL-17 level contributed to the development of ICVD, suggesting IL-17 as a potential risk marker. Further research is needed to establish IL-17 as a therapeutic biomarker of ICVD.
Collapse
Affiliation(s)
- Yu Miao
- Department of Health Statistics, Public Health College, Inner Mongolia Medical University, Hohhot, Inner Mongolia Province, 010000, China
| | - Tao Yan
- Department of Health Statistics, Public Health College, Inner Mongolia Medical University, Hohhot, Inner Mongolia Province, 010000, China
| | - Jia Liu
- Department of Health Statistics, Public Health College, Inner Mongolia Medical University, Hohhot, Inner Mongolia Province, 010000, China
| | - Chunfa Zhang
- Department of Health Statistics, Public Health College, Inner Mongolia Medical University, Hohhot, Inner Mongolia Province, 010000, China
| | - Jinli Yan
- Department of Health Statistics, Public Health College, Inner Mongolia Medical University, Hohhot, Inner Mongolia Province, 010000, China
| | - Lei Xu
- Department of Health Statistics, Public Health College, Inner Mongolia Medical University, Hohhot, Inner Mongolia Province, 010000, China
| | - Nan Zhang
- Department of Health Statistics, Public Health College, Inner Mongolia Medical University, Hohhot, Inner Mongolia Province, 010000, China.
| | - Xingguang Zhang
- Department of Health Statistics, Public Health College, Inner Mongolia Medical University, Hohhot, Inner Mongolia Province, 010000, China.
| |
Collapse
|
15
|
Zhang X, Chen S, Yin G, Liang P, Feng Y, Yu W, Meng D, Liu H, Zhang F. The Role of JAK/STAT Signaling Pathway and Its Downstream Influencing Factors in the Treatment of Atherosclerosis. J Cardiovasc Pharmacol Ther 2024; 29:10742484241248046. [PMID: 38656132 DOI: 10.1177/10742484241248046] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 04/26/2024]
Abstract
Atherosclerosis is now widely considered to be a chronic inflammatory disease, with increasing evidence suggesting that lipid alone is not the main factor contributing to its development. Rather, atherosclerotic plaques contain a significant amount of inflammatory cells, characterized by the accumulation of monocytes and lymphocytes on the vessel wall. This suggests that inflammation may play a crucial role in the occurrence and progression of atherosclerosis. As research deepens, other pathological factors have also been found to influence the development of the disease. The Janus kinase/signal transducer and activator of transcription (JAK/STAT) pathway is a recently discovered target of inflammation that has gained attention in recent years. Numerous studies have provided evidence for the causal role of this pathway in atherosclerosis, and its downstream signaling factors play a significant role in this process. This brief review aims to explore the crucial role of the JAK/STAT pathway and its representative downstream signaling factors in the development of atherosclerosis. It provides a new theoretical basis for clinically affecting the development of atherosclerosis by interfering with the JAK/STAT signaling pathway.
Collapse
Affiliation(s)
- Xin Zhang
- The First Clinical Medical College of Shandong University of Traditional Chinese Medicine, Jinan City, Shandong Province, China
| | - Suwen Chen
- The First Clinical Medical College of Shandong University of Traditional Chinese Medicine, Jinan City, Shandong Province, China
| | - Guoliang Yin
- The First Clinical Medical College of Shandong University of Traditional Chinese Medicine, Jinan City, Shandong Province, China
| | - Pengpeng Liang
- The First Clinical Medical College of Shandong University of Traditional Chinese Medicine, Jinan City, Shandong Province, China
| | - Yanan Feng
- The First Clinical Medical College of Shandong University of Traditional Chinese Medicine, Jinan City, Shandong Province, China
| | - Wenfei Yu
- The First Clinical Medical College of Shandong University of Traditional Chinese Medicine, Jinan City, Shandong Province, China
| | - Decheng Meng
- The First Clinical Medical College of Shandong University of Traditional Chinese Medicine, Jinan City, Shandong Province, China
| | - Hongshuai Liu
- The First Clinical Medical College of Shandong University of Traditional Chinese Medicine, Jinan City, Shandong Province, China
| | - Fengxia Zhang
- Hospital of Shandong University of Traditional Chinese Medicine, Jinan City, Shandong Province, China
| |
Collapse
|
16
|
Laurans L, Mouttoulingam N, Chajadine M, Lavelle A, Diedisheim M, Bacquer E, Creusot L, Suffee N, Esposito B, Melhem NJ, Le Goff W, Haddad Y, Paul JL, Rainteau D, Tedgui A, Ait-Oufella H, Zitvogel L, Sokol H, Taleb S. An obesogenic diet increases atherosclerosis through promoting microbiota dysbiosis-induced gut lymphocyte trafficking into the periphery. Cell Rep 2023; 42:113350. [PMID: 37897726 DOI: 10.1016/j.celrep.2023.113350] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2022] [Revised: 09/13/2023] [Accepted: 10/11/2023] [Indexed: 10/30/2023] Open
Abstract
Although high-fat diet (HFD)-induced gut microbiota dysbiosis is known to affect atherosclerosis, the underlying mechanisms remain to be fully explored. Here, we show that the progression of atherosclerosis depends on a gut microbiota shaped by an HFD but not a high-cholesterol (HC) diet and, more particularly, on low fiber (LF) intake. Mechanistically, gut lymphoid cells impacted by HFD- or LF-induced microbiota dysbiosis highly proliferate in mesenteric lymph nodes (MLNs) and migrate from MLNs to the periphery, which fuels T cell accumulation within atherosclerotic plaques. This is associated with the induction of mucosal addressin cell adhesion molecule 1 (MAdCAM-1) within plaques and the presence of enterotropic lymphocytes expressing β7 integrin. MLN resection or lymphocyte deficiency abrogates the pro-atherogenic effects of a microbiota shaped by LF. Our study shows a pathological link between a diet-shaped microbiota, gut immune cells, and atherosclerosis, suggesting that a diet-modulated microbiome might be a suitable therapeutic target to prevent atherosclerosis.
Collapse
Affiliation(s)
- Ludivine Laurans
- Université Paris Cité, INSERM, Paris Cardiovascular Research Center, 75015 Paris, France
| | - Nirmala Mouttoulingam
- Université Paris Cité, INSERM, Paris Cardiovascular Research Center, 75015 Paris, France
| | - Mouna Chajadine
- Université Paris Cité, INSERM, Paris Cardiovascular Research Center, 75015 Paris, France
| | - Aonghus Lavelle
- Sorbonne Université, INSERM, Centre de Recherche Saint-Antoine, CRSA, AP-HP, Saint Antoine Hospital, Gastroenterology Department, 75012 Paris, France; Paris Centre for Microbiome Medicine (PaCeMM) FHU, Paris, France
| | - Marc Diedisheim
- Clinique Saint Gatien Alliance (NCT+), 37540 Saint-Cyr-sur-Loire, France; Institut Necker-Enfants Malades (INEM), Université Paris Cité, INSERM UMR-S1151, CNRS UMR-S8253, 75015 Paris, France
| | - Emilie Bacquer
- Université Paris Cité, INSERM, Paris Cardiovascular Research Center, 75015 Paris, France
| | - Laura Creusot
- Sorbonne Université, INSERM, Centre de Recherche Saint-Antoine, CRSA, AP-HP, Saint Antoine Hospital, Gastroenterology Department, 75012 Paris, France; Paris Centre for Microbiome Medicine (PaCeMM) FHU, Paris, France
| | - Nadine Suffee
- Université Paris Cité, INSERM, Paris Cardiovascular Research Center, 75015 Paris, France; INSERM UMRS1166, ICAN-Institute of Cardiometabolism and Nutrition, Sorbonne University, 75013 Paris, France
| | - Bruno Esposito
- Université Paris Cité, INSERM, Paris Cardiovascular Research Center, 75015 Paris, France
| | - Nada Joe Melhem
- Université Paris Cité, INSERM, Paris Cardiovascular Research Center, 75015 Paris, France
| | - Wilfried Le Goff
- INSERM UMRS1166, ICAN-Institute of Cardiometabolism and Nutrition, Sorbonne University, 75013 Paris, France
| | - Yacine Haddad
- Université Paris Cité, INSERM, Paris Cardiovascular Research Center, 75015 Paris, France; Gustave Roussy, Villejuif, France; Institut National de la Santé et de la Recherche Médicale, Gustave Roussy, UMR1015, Villejuif, France
| | - Jean-Louis Paul
- Université Paris-Sud, Equipe d'Accueil 4529, UFR de Pharmacie, Chatenay-Malabry, France and Assistance Publique Hôpitaux de Paris, Hôpital Européen Georges Pompidou, Paris, France
| | - Dominique Rainteau
- Paris Centre for Microbiome Medicine (PaCeMM) FHU, Paris, France; Sorbonne Université, INSERM, Centre de Recherche Saint-Antoine, CRSA, AP-HP, Saint Antoine Hospital, Clinical Metabolomics Department, 75012 Paris, France
| | - Alain Tedgui
- Université Paris Cité, INSERM, Paris Cardiovascular Research Center, 75015 Paris, France
| | - Hafid Ait-Oufella
- Université Paris Cité, INSERM, Paris Cardiovascular Research Center, 75015 Paris, France
| | - Laurence Zitvogel
- Gustave Roussy, Villejuif, France; Institut National de la Santé et de la Recherche Médicale, Gustave Roussy, UMR1015, Villejuif, France; Université Paris-Saclay, Faculté de Médecine, Le Kremlin Bicêtre, France; Center of Clinical Investigations BIOTHERIS, INSERM CIC1428, Gustave Roussy, Villejuif, France
| | - Harry Sokol
- Sorbonne Université, INSERM, Centre de Recherche Saint-Antoine, CRSA, AP-HP, Saint Antoine Hospital, Gastroenterology Department, 75012 Paris, France; Paris Centre for Microbiome Medicine (PaCeMM) FHU, Paris, France; INRAe, Micalis & AgroParisTech, Jouy en Josas, France
| | - Soraya Taleb
- Université Paris Cité, INSERM, Paris Cardiovascular Research Center, 75015 Paris, France.
| |
Collapse
|
17
|
Appleton BD, Palmer SA, Smith HP, Stephens LE, Major AS. Oxidized Phospholipid oxPAPC Alters Regulatory T-Cell Differentiation and Decreases Their Protective Function in Atherosclerosis in Mice. Arterioscler Thromb Vasc Biol 2023; 43:2119-2132. [PMID: 37675632 PMCID: PMC10720352 DOI: 10.1161/atvbaha.123.319674] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2023] [Accepted: 08/22/2023] [Indexed: 09/08/2023]
Abstract
BACKGROUND Regulatory T cells (Tregs) are protective in atherosclerosis but reduced during disease progression due to cell death and loss of stability. However, the mechanisms of Treg dysfunction remain unknown. Oxidized phospholipids are abundant in atherosclerosis and can activate innate immune cells, but little is known regarding their impact on T cells. Given Treg loss during atherosclerosis progression and oxidized phospholipid levels in the plaque microenvironment, we investigated whether oxidized 1-palmitoyl-2-arachidonoyl-sn-glycero-3-phosphocholine (oxPAPC), an oxidized phospholipid associated with atherosclerotic plaques, alters Treg differentiation and function. METHODS CD4+ T cells were polarized to Treg, T helper (Th) 1, and Th17 cells with or without oxPAPC and assessed by flow cytometry. Gene expression in oxPAPC-treated Tregs was analyzed by bulk RNA sequencing. Functional studies of oxPAPC-induced Tregs were performed by coculturing Tregs with CellTrace Violet-labeled cells in vitro, and by adoptively transferring Tregs to hyperlipidemic Ldlr-/- mice to measure atherosclerosis progression. RESULTS Compared with controls, oxPAPC-treated Tregs were less viable, but surviving cells expressed higher levels of the Th1-associated markers T-bet, CXCR3, and IFN (interferon)-γ. Th1 and Th17 skewing cultures were unaltered by oxPAPC. IFN-γ is linked to Treg instability, thus Treg polarization experiments were repeated using Ifngr1-/- CD4+ T cells. IFNγR1 (INF gamma receptor 1) deficiency did not improve cell viability in oxPAPC-treated Tregs; however, T-bet and IFN-γ expression was not increased in surviving cells suggesting a role for IFN-γsignaling. OxPAPC-treated Tregs were less suppressive in vitro, and adoptive transfer studies in hyperlipidemic Ldlr-/- mice showed that oxPAPC-induced Tregs possessed altered tissue homing and were insufficient to inhibit atherosclerosis progression. CONCLUSIONS OxPAPC elicits Treg-specific changes altering Treg differentiation and inducing a Th1-like phenotype in surviving cells partially through IFN-γ signaling. This is biologically relevant as oxPAPC-treated Tregs do not reduce atherosclerosis progression in Ldlr-/- mice. This study supports the role of oxidized phospholipids in negatively impacting Treg differentiation and atheroprotective function.
Collapse
Affiliation(s)
- Brenna D. Appleton
- Department of Pathology, Microbiology and Immunology, Vanderbilt University
| | | | | | | | - Amy S. Major
- Department of Pathology, Microbiology and Immunology, Vanderbilt University
- Department of Medicine, Vanderbilt University Medical Center
- Tennessee Valley Health System, Department of Veterans Affairs
| |
Collapse
|
18
|
Şenol N, Şahin M, Şahin U. The protective role of 5-hydroxy-1,4-naphthoquinone against the harmful effects of 50 Hz electric field in rat lung tissue. Electromagn Biol Med 2023; 42:133-143. [PMID: 37811636 DOI: 10.1080/15368378.2023.2265935] [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: 12/02/2022] [Accepted: 09/16/2023] [Indexed: 10/10/2023]
Abstract
There is strong scientific evidence that the electric field is harmful to life. Exposure to an electric field (EF) can cause lung toxicity and respiratory disorders. In addition, the electric field has been shown to cause tissue damage through inflammation and apoptosis. Juglone (JUG) is one of the powerful antioxidants with anti-apoptotic and anti-inflammatory, various pharmacological properties in the biological system. In this study, we evaluated the efficacy of JUG against the potential adverse effects of electric field on the lung. Twenty-four Wistar albino rats were randomly divided into three groups; control group (Cont), EF group, and EF exposure+JUG-treated group (EJUG). After routine histological procedures, sections stained with hematoxylin-eosin (H&E) showed significant changes in lung tissues in the EF group compared to the Cont group. Significant protective effects were observed in the building volumes and histopathology in the EJUG group. Our immunohistochemical and gene expression results increased the expression of caspase-3 and tumor necrosis factor alpha (TNF-α) in the EF group (p < 0.05). Juglon increased cytokine signal suppressor (SOCS) expression (p < 0.001). These findings were consistent with the antioxidant effect of JUG treatment. We reasoned that exposure to EF damaged rat lung tissues and administration of JUG alleviated the complications caused by 50 Hz EF.
Collapse
Affiliation(s)
- Nurgül Şenol
- Department of Nutrition and Dietetics, Faculty of Health Sciences, Süleyman Demirel University, Isparta, Türkiye
| | - Melda Şahin
- Department of Bioengineering, Institute of Science, Süleyman Demirel University, Isparta, Türkiye
| | - Uğur Şahin
- Department of Chemistry, Faculty of Art and Science, University of Süleyman Demirel, Isparta, Türkiye
- Genetic Research Unit, Innovative Technologies Application and Research Center, Süleyman Demirel University, Isparta, Türkiye
| |
Collapse
|
19
|
Dolade N, Rayego-Mateos S, Garcia-Carrasco A, Guerin M, Martín-Ventura JL, Ruiz-Ortega M, Tharaux PL, Valdivielso JM. B- and T-lymphocyte attenuator could be a new player in accelerated atherosclerosis associated with chronic kidney disease. Clin Sci (Lond) 2023; 137:1409-1429. [PMID: 37655751 DOI: 10.1042/cs20230399] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2023] [Revised: 07/31/2023] [Accepted: 09/01/2023] [Indexed: 09/02/2023]
Abstract
BACKGROUND In chronic kidney disease (CKD), cardiovascular morbi-mortality is higher than in general population. Atherosclerotic cardiovascular disease is accelerated in CKD, but specific CKD-related risk factors for atherosclerosis are unknown. METHODS CKD patients from the NEFRONA study were used. We performed mRNA array from blood of patients free from atheroma plaque at baseline, with (n=10) and without (n=10) de novo atherosclerotic plaque development 2 years later. Selected mRNA candidates were validated in a bigger sample (n=148). Validated candidates were investigated in vivo in an experimental model of CKD-accelerated atherosclerosis, and in vitro in murine macrophages. RESULTS mRNA array analysis showed 92 up-regulated and 67 down-regulated mRNAs in samples from CKD patients with de novo plaque development. The functional analysis pointed to a paramount role of the immune response. The validation in a bigger sample confirmed that B- and T-lymphocyte co-inhibitory molecule (BTLA) down-regulation was associated with de novo plaque presence after 2 years. However, BTLA down-regulation was not found to be associated with atherosclerotic progression in patients with plaque already present at baseline. In a model of CKD-accelerated atherosclerosis, mRNA and protein expression levels of BTLA were significantly decreased in blood samples and atheroma plaques. Plaques from animals with CKD were bigger, had more infiltration of inflammatory cells, higher expression of IL6 and IL17 and less presence of collagen than plaques from control animals. Incubation of macrophages with rat uremic serum decreased BTLA expression. CONCLUSIONS BTLA could be a potential biomarker or therapeutic target for atherosclerosis incidence in CKD patients.
Collapse
Affiliation(s)
- Nuria Dolade
- Red de Investigación Renal (REDinREN), Ricords2040, Spain
- Vascular and Renal Translational Research Group, Institut de Recerca Biomèdica de Lleida IRBLleida, Lleida 25198, Spain
| | - Sandra Rayego-Mateos
- Red de Investigación Renal (REDinREN), Ricords2040, Spain
- Vascular and Renal Translational Research Group, Institut de Recerca Biomèdica de Lleida IRBLleida, Lleida 25198, Spain
| | - Alicia Garcia-Carrasco
- Red de Investigación Renal (REDinREN), Ricords2040, Spain
- Vascular and Renal Translational Research Group, Institut de Recerca Biomèdica de Lleida IRBLleida, Lleida 25198, Spain
| | - Maryse Guerin
- INSERM UMR_S 1166 - ICAN. Faculté de Médecine Pitié-Salpêtrière: 75013 Paris, France
| | - Jose-Luis Martín-Ventura
- Vascular Research Laboratory, IIS-Fundacion Jimenez Diaz, Universidad Autonoma, Avda Reyes Catolicos 2, 28040 Madrid, Spain and CIBER de Enfermedades Cardiovasculares (CIBERCV), Madrid, Spain
| | - Marta Ruiz-Ortega
- Cellular and Molecular Biology in Renal and Vascular Pathology Laboratory, IIS-Fundación Jiménez Díaz-Universidad Autónoma Madrid, Madrid, Spain
| | - Pierre-Louis Tharaux
- Paris Cardiovascular research Centre (PARCC), Institut National de la Santé et de la Recherche Médicale, INSERM, Université Paris Cité, Paris, France
| | - Jose Manuel Valdivielso
- Red de Investigación Renal (REDinREN), Ricords2040, Spain
- Vascular and Renal Translational Research Group, Institut de Recerca Biomèdica de Lleida IRBLleida, Lleida 25198, Spain
| |
Collapse
|
20
|
Gong W, Tian Y, Li L. T cells in abdominal aortic aneurysm: immunomodulation and clinical application. Front Immunol 2023; 14:1240132. [PMID: 37662948 PMCID: PMC10471798 DOI: 10.3389/fimmu.2023.1240132] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2023] [Accepted: 08/07/2023] [Indexed: 09/05/2023] Open
Abstract
Abdominal aortic aneurysm (AAA) is characterized by inflammatory cell infiltration, extracellular matrix (ECM) degradation, and vascular smooth muscle cell (SMC) dysfunction. The inflammatory cells involved in AAA mainly include immune cells including macrophages, neutrophils, T-lymphocytes and B lymphocytes and endothelial cells. As the blood vessel wall expands, more and more lymphocytes infiltrate into the outer membrane. It was found that more than 50% of lymphocytes in AAA tissues were CD3+ T cells, including CD4+, CD8+T cells, γδ T cells and regulatory T cells (Tregs). Due to the important role of T cells in inflammatory response, an increasing number of researchers have paid attention to the role of T cells in AAA and dug into the relevant mechanism. Therefore, this paper focuses on reviewing the immunoregulatory role of T cells in AAA and their role in immunotherapy, seeking potential targets for immunotherapy and putting forward future research directions.
Collapse
Affiliation(s)
| | | | - Lei Li
- Department of Vascular Surgery, The Second Affiliated Hospital of Dalian Medical University, Dalian, Liaoning, China
| |
Collapse
|
21
|
Yazman S, Depboylu BC, Saruhan E, Cenikli U, Harmandar B, Arslan K, İlhan G, İştar H. New Biomarkers (Endocan, Interleukin-17, and Thrombospondin-4) for the Diagnosis, Assessment of Severity, and Follow-Up of Peripheral Arterial Disease. Angiology 2023; 74:631-639. [PMID: 37010303 DOI: 10.1177/00033197231166492] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/04/2023]
Abstract
The present study evaluated the use of endocan, interleukin-17 (IL-17), and thrombospondin-4 (TSP-4) blood levels as potential biomarkers for the diagnosis and follow-up of peripheral arterial disease (PAD). Patients with PAD (Rutherford categories I, II, and III) who were admitted between March 2020 and March 2022 for cardiovascular surgery or outpatient clinic follow-up were included. The patients (n = 60) were divided into 2 groups: medical treatment (n = 30) and surgical treatment (n = 30). In addition, a control group (n = 30) was created for comparison. Endocan, IL-17, and TSP-4 blood levels were measured at the time of diagnosis and at the first month after treatment. Endocan and IL-17 values were found to be significantly higher in both groups that underwent medical (259.7 ± 46 pg/mL, 63.7 ± 16.6 pg/mL) and surgical (290.3 ± 84.5 pg/mL, 66.4 ± 19.6 pg/mL) treatment than the control group (187.4 ± 34.5 pg/mL, 56.5 ± 7.2 pg/mL P < .001). Tsp-4 value was found to be significantly higher only in the surgical treatment group (15 ± 4.3 ng/mL) than the control group (12.9 ± 1.4 ng/mL P < .05). The decreases in endocan, IL-17, and TSP-4 levels at the first month of treatment in both groups were also significant (P < .001). A combination of classical and these new biomarkers could be included in PAD screening, early diagnosis, severity determination, and follow-up protocols in order to provide effective assessment in clinical practice.
Collapse
Affiliation(s)
- Serkan Yazman
- Department of Cardiovascular Surgery, Muğla Sıtkı Koçman University Faculty of Medicine, Mugla, Turkey
| | - Burak Can Depboylu
- Department of Cardiovascular Surgery, Muğla Sıtkı Koçman University Faculty of Medicine, Mugla, Turkey
| | - Ercan Saruhan
- Department of Medical Biochemistry, Muğla Sıtkı Koçman University Faculty of Medicine, Mugla, Turkey
| | - Utku Cenikli
- Department of Neurology, Muğla Sıtkı Koçman University Faculty of Medicine, Mugla, Turkey
| | - Buğra Harmandar
- Department of Cardiovascular Surgery, Muğla Sıtkı Koçman University Faculty of Medicine, Mugla, Turkey
| | - Kadir Arslan
- Department of Cardiovascular Surgery, Muğla Sıtkı Koçman University Faculty of Medicine, Mugla, Turkey
| | - Gökhan İlhan
- Department of Cardiovascular Surgery, Muğla Sıtkı Koçman University Faculty of Medicine, Mugla, Turkey
| | - Hande İştar
- Department of Cardiovascular Surgery, Muğla Sıtkı Koçman University Faculty of Medicine, Mugla, Turkey
| |
Collapse
|
22
|
Chi CC, Wu YW, Chao TH, Chen CC, Chen YJ, Cheng HM, Chiu HY, Chiu YW, Chung WH, Hsieh TY, Huang PH, Huang YH, Lin SH, Lin TH, Ueng KC, Wang CC, Wang YC, Wu NL, Jia-Yin Hou C, Tsai TF. 2022 Taiwanese Dermatological Association (TDA), Taiwanese Association for Psoriasis and Skin Immunology (TAPSI), and Taiwan Society of cardiology (TSOC) joint consensus recommendations for the management of psoriatic disease with attention to cardiovascular comorbidities. J Formos Med Assoc 2023; 122:442-457. [PMID: 36347733 DOI: 10.1016/j.jfma.2022.10.010] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2022] [Revised: 08/09/2022] [Accepted: 10/20/2022] [Indexed: 11/08/2022] Open
Abstract
Psoriatic disease is a chronic inflammatory disorder with skin and joint manifestations. Due to the persistent inflammatory state exhibited by patients with psoriasis, multiple systemic comorbidities occur more frequently in patients with psoriasis than in the general population, and the risk of cardiovascular (CV) diseases is significantly increased. As the pathophysiology of psoriatic disease is becoming better understood, the sharing of underlying pathogenic mechanisms between psoriatic and CV diseases is becoming increasingly apparent. Consequently, careful attention to CV comorbidities that already exist or may potentially develop is needed in the management of patients with psoriasis, particularly in the screening and primary prevention of CV disease and in treatment selection due to potential drug-drug and drug-disease interactions. Furthermore, as the use of effective biologic therapy and more aggressive oral systemic treatment for psoriatic disease is increasing, consideration of the potential positive and negative effects of oral and biologic treatment on CV disease is warranted. To improve outcomes and quality of care for patients with psoriasis, the Taiwanese Dermatological Association, the Taiwanese Association for Psoriasis and Skin Immunology, and the Taiwan Society of Cardiology established a Task Force of 20 clinicians from the fields of dermatology, cardiology, and rheumatology to jointly develop consensus expert recommendations for the management of patients with psoriatic disease with attention to CV comorbidities.
Collapse
Affiliation(s)
- Ching-Chi Chi
- Department of Dermatology, Chang Gung Memorial Hospital-Linkou, Taoyuan, Taiwan; College of Medicine, Chang Gung University, Taoyuan, Taiwan
| | - Yen-Wen Wu
- Division of Cardiology, Cardiovascular Medical Center, Far Eastern Memorial Hospital, New Taipei City, Taiwan; School of Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan
| | - Ting-Hsing Chao
- Division of Cardiology, Department of Internal Medicine, National Cheng Kung University Hospital, Tainan, Taiwan; College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | - Chih-Chiang Chen
- School of Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan; Department of Dermatology, Taipei Veterans General Hospital, Taipei, Taiwan
| | - Yi-Ju Chen
- Department of Dermatology, Taichung Veterans General Hospital, Taichung, Taiwan; College of Medicine, National Chung Hsing University, Taichung, Taiwan
| | - Hao-Min Cheng
- School of Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan; Center for Evidence-Based Medicine, Taipei Veterans General Hospital, Taipei, Taiwan
| | - Hsien-Yi Chiu
- Department of Dermatology, National Taiwan University Hospital Hsin-Chu Branch, Hsinchu, Taiwan; College of Medicine, National Taiwan University, Taipei, Taiwan
| | - Yu-Wei Chiu
- Division of Cardiology, Cardiovascular Medical Center, Far Eastern Memorial Hospital, New Taipei City, Taiwan; Department of Computer Science and Engineering, Yuan Ze University, Taoyuan, Taiwan
| | - Wen-Hung Chung
- Department of Dermatology, Chang Gung Memorial Hospital-Linkou, Taoyuan, Taiwan; College of Medicine, Chang Gung University, Taoyuan, Taiwan
| | - Tsu-Yi Hsieh
- Division of Allergy, Immunology and Rheumatology, Department of Internal Medicine, Taichung Veterans General Hospital, Taichung, Taiwan
| | - Po-Hsun Huang
- Division of Cardiology, Department of Internal Medicine, Taipei Veterans General Hospital, Taipei, Taiwan; Cardiovascular Research Center, National Yang Ming Chiao Tung University, Taipei, Taiwan
| | - Yu-Huei Huang
- Department of Dermatology, Chang Gung Memorial Hospital-Linkou, Taoyuan, Taiwan; College of Medicine, Chang Gung University, Taoyuan, Taiwan
| | - Shang-Hung Lin
- College of Medicine, Chang Gung University, Taoyuan, Taiwan; Department of Dermatology, Kaohsiung Chang Gung Memorial Hospital, Kaohsiung, Taiwan
| | - Tsung-Hsien Lin
- Division of Cardiology, Department of Internal Medicine, Kaohsiung Medical University Hospital, Kaohsiung, Taiwan; College of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Kwo-Chang Ueng
- Division of Cardiology, Department of Internal Medicine, Chung-Shan Medical University Hospital, Taichung, Taiwan; College of Medicine, Chung-Shan Medical University, Taichung, Taiwan
| | - Chun-Chieh Wang
- College of Medicine, Chang Gung University, Taoyuan, Taiwan; Division of Cardiology, Department of Internal Medicine, Chang Gung Memorial Hospital-Linkou, Taoyuan, Taiwan
| | - Yu-Chen Wang
- Division of Cardiology, Department of Internal Medicine, Asia University Hospital, Taichung, Taiwan; Department of Medical Laboratory Science and Biotechnology, Asia University, Taichung, Taiwan
| | - Nan-Lin Wu
- Department of Dermatology, MacKay Memorial Hospital, Taipei, Taiwan; Department of Medicine, MacKay Medical College, New Taipei City, Taiwan
| | - Charles Jia-Yin Hou
- Department of Medicine, MacKay Medical College, New Taipei City, Taiwan; Division of Cardiology, Department of Internal Medicine, MacKay Memorial Hospital, Taipei, Taiwan.
| | - Tsen-Fang Tsai
- College of Medicine, National Taiwan University, Taipei, Taiwan; Department of Dermatology, National Taiwan University Hospital, Taipei, Taiwan.
| |
Collapse
|
23
|
Kommoss KS, Enk A, Heikenwälder M, Waisman A, Karbach S, Wild J. Cardiovascular comorbidity in psoriasis - psoriatic inflammation is more than just skin deep. J Dtsch Dermatol Ges 2023. [PMID: 37186503 DOI: 10.1111/ddg.15071] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2022] [Accepted: 02/24/2023] [Indexed: 05/17/2023]
Abstract
BACKGROUND There is a growing understanding of inflammation in psoriasis beyond its dermatological manifestation, towards systemic inflammation. Management of possible comorbidities encompassing psychological, metabolic and cardiovascular disease is recommended in national and international dermatology guidelines for treatment of psoriasis patients. Vice versa, psoriasis is being recognized as a new risk factor for cardiovascular inflammation within the cardiological community. METHODS A review of the literature was conducted. Key points regarding epidemiological, mechanistic and management aspects were summarized and put into context for physicians treating psoriasis patients. RESULTS Efforts are currently being made to better understand the mechanistic underpinnings of systemic inflammation within psoriatic inflammation. Studies looking to "hit two birds with one stone" regarding specifically cardiovascular comorbidities of psoriasis patients using established systemic dermatological therapies have so far provided heterogeneous data. The diagnosis of psoriasis entails preventive and therapeutic consequences regarding concomitant diseases for the individual patient. CONCLUSIONS The knowledge of comorbidities in psoriasis calls for pronounced interdisciplinary care of psoriasis patients, to which this article highlights efforts regarding vascular inflammation and cardiovascular disease.
Collapse
Affiliation(s)
- Katharina S Kommoss
- Department of Dermatology, University Hospital Heidelberg, Heidelberg, Germany
- Division of Chronic Inflammation and Cancer, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Alexander Enk
- Department of Dermatology, University Hospital Heidelberg, Heidelberg, Germany
| | - Mathias Heikenwälder
- Division of Chronic Inflammation and Cancer, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Ari Waisman
- Institute for Molecular Medicine, University Medical Center of Mainz, Mainz, Germany
- Research Center for Immunotherapy, University Medical Center of the Johannes Gutenberg-University Mainz, Mainz, Germany
| | - Susanne Karbach
- Center for Cardiology - Cardiology I, University Medical Center Mainz, Mainz, Germany
- Center for Thrombosis and Hemostasis, University Medical Center Mainz, Mainz, Germany
- German Center for Cardiovascular Research (DZHK) - Partner site RheinMain, Germany
| | - Johannes Wild
- Center for Cardiology - Cardiology I, University Medical Center Mainz, Mainz, Germany
- Center for Thrombosis and Hemostasis, University Medical Center Mainz, Mainz, Germany
- German Center for Cardiovascular Research (DZHK) - Partner site RheinMain, Germany
| |
Collapse
|
24
|
Cicmil S, Cicmil A, Pavlic V, Krunić J, Sladoje Puhalo D, Bokonjić D, Čolić M. Periodontal Disease in Young Adults as a Risk Factor for Subclinical Atherosclerosis: A Clinical, Biochemical and Immunological Study. J Clin Med 2023; 12:jcm12062197. [PMID: 36983201 PMCID: PMC10051366 DOI: 10.3390/jcm12062197] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2023] [Revised: 03/08/2023] [Accepted: 03/09/2023] [Indexed: 03/16/2023] Open
Abstract
Although a strong relationship between periodontal disease (PD) and atherosclerosis was shown in adults, little data are published in younger PD patients. Therefore, this study aimed to investigate and correlate clinical parameters of PD, pro- and immunoregulatory cytokines in gingival crevicular fluid (GCF) and serum, biochemical and hematological parameters associated with atherosclerosis risk, and carotid intima-media thickness (IMT) in our younger study participants (n = 78) (mean age 35.92 ± 3.36 years) who were divided into two equal groups: subjects with and without PD. PD patients had higher values of IMT, hs-CRP, triglycerides, total cholesterol, and LDL; most proinflammatory and Th1/Th17-associated cytokines in GCF; and IL-8, IL-12, IL-18, and IL-17A in serum compared to subjects without PD. These cytokines in GCF positively correlated with most clinical periodontal parameters. Clinical periodontal parameters, TNF-α and IL-8 in GCF and IL-17A, hs-CRP, and LDL in serum, had more significant predictive roles in developing subclinical atherosclerosis (IMT ≥ 0.75 mm) in comparison with other cytokines, fibrinogen, and other lipid status parameters. Hs-CRP correlated better with the proinflammatory cytokines than the parameters of lipid status. Except for serum IL-17A, there was no significant association of clinical and immunological PD parameters with lipid status. Overall, these results suggest that dyslipidemia and PD status seem to be independent risk factors for subclinical atherosclerosis in our younger PD population.
Collapse
Affiliation(s)
- Smiljka Cicmil
- Department of Oral Rehabilitation, Faculty of Medicine Foca, University of East Sarajevo, 73300 Foca, Bosnia and Herzegovina
- Correspondence:
| | - Ana Cicmil
- Department of Oral Rehabilitation, Faculty of Medicine Foca, University of East Sarajevo, 73300 Foca, Bosnia and Herzegovina
| | - Verica Pavlic
- Department of Periodontology and Oral Medicine, Faculty of Medicine, University of Banja Luka, 78000 Banja Luka, Bosnia and Herzegovina
- Department of Periodontology and Oral Medicine, The Republic of Srpska, Institute of Dentistry, 78000 Banja Luka, Bosnia and Herzegovina
| | - Jelena Krunić
- Department of Dental Pathology, Faculty of Medicine Foca, University of East Sarajevo, 73300 Foca, Bosnia and Herzegovina
| | - Dragana Sladoje Puhalo
- Department of Biochemistry, Faculty of Medicine Foca, University of East Sarajevo, 73300 Foca, Bosnia and Herzegovina
| | - Dejan Bokonjić
- Department of Pediatrics, Faculty of Medicine Foca, University of East Sarajevo, 73300 Foca, Bosnia and Herzegovina
| | - Miodrag Čolić
- Center for Biomedical Sciences, Faculty of Medicine Foca, University of East Sarajevo, 73300 Foca, Bosnia and Herzegovina
- Serbian Academy of Sciences and Arts, 11000 Belgrade, Serbia
| |
Collapse
|
25
|
Bahmani L, Baghi M, Peymani M, Javeri A, Ghaedi K. The PBX1/miR-141-miR-200a/EGR2/SOCS3 Axis; Integrative Analysis of Interaction Networks to Discover the Possible Mechanism of MiR-141 and MiR-200a-Mediated Th17 Cell Differentiation. IRANIAN JOURNAL OF BIOTECHNOLOGY 2023; 21:e3211. [PMID: 36811100 PMCID: PMC9938929 DOI: 10.30498/ijb.2022.317078.3211] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/28/2021] [Accepted: 03/06/2022] [Indexed: 02/24/2023]
Abstract
Background Overexpression of miR-141 and miR-200a is known to be associated with the differentiation of T helper 17 (Th17) cells, which are key players in the pathophysiology of autoimmune disorders. However, the function and governing mechanism of these two microRNAs (miRNAs) in Th17 cell skewing are poorly defined. Objectives The aim of the present study was to identify the common upstream transcription factors and downstream target genes of miR-141 and miR-200a to obtain a better insight into the possible dysregulated molecular regulatory networks driving miR-141/miR-200a-mediated Th17 cell development. Materials and Methods A consensus-based prediction strategy was applied for in-silico identification of potential transcription factors and putative gene targets of miR-141 and miR-200a. Thereafter, we analyzed the expression patterns of candidate transcription factors and target genes during human Th17 cell differentiation by quantitative real-time PCR and examined the direct interaction between both miRNAs and their potential target sequences using dual-luciferase reporter assays. Results According to our miRNA-based and gene-based interaction network analyses, pre-B cell leukemia homeobox (PBX1) and early growth response 2 (EGR2) were respectively taken into account as the potential upstream transcription factor and downstream target gene of miR-141 and miR-200a. There was a significant overexpression of the PBX1 gene during the Th17 cell induction period. Furthermore, both miRNAs could directly target EGR2 and inhibit its expression. As a downstream gene of EGR2, the suppressor of cytokine signaling 3 (SOCS3) was also downregulated during the differentiation process. Conclusions These results indicate that activation of the PBX1/miR-141-miR-200a/EGR2/SOCS3 axis may promote Th17 cell development and, therefore, trigger or exacerbate Th17-mediated autoimmunity.
Collapse
Affiliation(s)
- Leila Bahmani
- Department of Stem Cells and Regenerative Medicine, Institute for Medical Biotechnology, National Institute of Genetic Engineering and Biotechnology (NIGEB), Tehran, Iran
| | - Masoud Baghi
- Department of Cell and Molecular Biology and Microbiology, Faculty of Biological Science and Technology, University of Isfahan, Isfahan, Iran,
Department of Animal Biotechnology, Cell Science Research Center, Royan Institute for Biotechnology, ACECR, Isfahan, Iran
| | - Maryam Peymani
- Department of Biology, Faculty of Basic Sciences, Shahrekord Branch, Islamic Azad University, Shahrekord, Iran
| | - Arash Javeri
- Department of Stem Cells and Regenerative Medicine, Institute for Medical Biotechnology, National Institute of Genetic Engineering and Biotechnology (NIGEB), Tehran, Iran
| | - Kamran Ghaedi
- Department of Cell and Molecular Biology and Microbiology, Faculty of Biological Science and Technology, University of Isfahan, Isfahan, Iran
| |
Collapse
|
26
|
Fernández-Gallego N, Castillo-González R, Méndez-Barbero N, López-Sanz C, Obeso D, Villaseñor A, Escribese MM, López-Melgar B, Salamanca J, Benedicto-Buendía A, Jiménez-Borreguero LJ, Ibañez B, Sastre J, Belver MT, Vega F, Blanco C, Barber D, Sánchez-Madrid F, de la Fuente H, Martín P, Esteban V, Jiménez-Saiz R. The impact of type 2 immunity and allergic diseases in atherosclerosis. Allergy 2022; 77:3249-3266. [PMID: 35781885 DOI: 10.1111/all.15426] [Citation(s) in RCA: 32] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2022] [Revised: 06/22/2022] [Accepted: 06/28/2022] [Indexed: 01/28/2023]
Abstract
Allergic diseases are allergen-induced immunological disorders characterized by the development of type 2 immunity and IgE responses. The prevalence of allergic diseases has been on the rise alike cardiovascular disease (CVD), which affects arteries of different organs such as the heart, the kidney and the brain. The underlying cause of CVD is often atherosclerosis, a disease distinguished by endothelial dysfunction, fibrofatty material accumulation in the intima of the artery wall, smooth muscle cell proliferation, and Th1 inflammation. The opposed T-cell identity of allergy and atherosclerosis implies an atheroprotective role for Th2 cells by counteracting Th1 responses. Yet, the clinical association between allergic disease and CVD argues against it. Within, we review different phases of allergic pathology, basic immunological mechanisms of atherosclerosis and the clinical association between allergic diseases (particularly asthma, atopic dermatitis, allergic rhinitis and food allergy) and CVD. Then, we discuss putative atherogenic mechanisms of type 2 immunity and allergic inflammation including acute allergic reactions (IgE, IgG1, mast cells, macrophages and allergic mediators such as vasoactive components, growth factors and those derived from the complement, contact and coagulation systems) and late phase inflammation (Th2 cells, eosinophils, type 2 innate-like lymphoid cells, alarmins, IL-4, IL-5, IL-9, IL-13 and IL-17).
Collapse
Affiliation(s)
- Nieves Fernández-Gallego
- Vascular Pathophysiology Area, Centro Nacional de Investigaciones Cardiovasculares (CNIC), Madrid, Spain
- Department of Immunology, Instituto de Investigación Sanitaria Hospital Universitario de La Princesa (IIS-Princesa), Universidad Autónoma de Madrid (UAM), Madrid, Spain
| | - Raquel Castillo-González
- Vascular Pathophysiology Area, Centro Nacional de Investigaciones Cardiovasculares (CNIC), Madrid, Spain
- Department of Immunology, Instituto de Investigación Sanitaria Hospital Universitario de La Princesa (IIS-Princesa), Universidad Autónoma de Madrid (UAM), Madrid, Spain
- Department of Pathology, Hospital 12 de Octubre, Madrid, Spain
| | - Nerea Méndez-Barbero
- Vascular Research Laboratory, Instituto de Investigación Sanitaria Fundación Jiménez Díaz (IIS-FJD), Madrid, Spain
- CIBER de Enfermedades Cardiovasculares (CIBERCV), Instituto de Salud Carlos III, Madrid, Spain
| | - Celia López-Sanz
- Department of Immunology, Instituto de Investigación Sanitaria Hospital Universitario de La Princesa (IIS-Princesa), Universidad Autónoma de Madrid (UAM), Madrid, Spain
| | - David Obeso
- Department of Basic Medical Sciences, Faculty of Medicine, Institute of Applied Molecular Medicine Nemesio Díez (IMMA), Universidad San Pablo-CEU, CEU Universities, Madrid, Spain
- Department of Chemistry and Biochemistry, Faculty of Pharmacy, Centre for Metabolomics and Bioanalysis (CEMBIO), Universidad San Pablo-CEU, CEU Universities, Madrid, Spain
| | - Alma Villaseñor
- Department of Basic Medical Sciences, Faculty of Medicine, Institute of Applied Molecular Medicine Nemesio Díez (IMMA), Universidad San Pablo-CEU, CEU Universities, Madrid, Spain
- Department of Chemistry and Biochemistry, Faculty of Pharmacy, Centre for Metabolomics and Bioanalysis (CEMBIO), Universidad San Pablo-CEU, CEU Universities, Madrid, Spain
| | - María M Escribese
- Department of Basic Medical Sciences, Faculty of Medicine, Institute of Applied Molecular Medicine Nemesio Díez (IMMA), Universidad San Pablo-CEU, CEU Universities, Madrid, Spain
| | - Beatriz López-Melgar
- Department of Cardiology, Instituto de Investigación Sanitaria Hospital Universitario de La Princesa (IIS-Princesa), Madrid, Spain
| | - Jorge Salamanca
- Department of Cardiology, Instituto de Investigación Sanitaria Hospital Universitario de La Princesa (IIS-Princesa), Madrid, Spain
| | - Amparo Benedicto-Buendía
- Department of Cardiology, Instituto de Investigación Sanitaria Hospital Universitario de La Princesa (IIS-Princesa), Madrid, Spain
| | - Luis Jesús Jiménez-Borreguero
- CIBER de Enfermedades Cardiovasculares (CIBERCV), Instituto de Salud Carlos III, Madrid, Spain
- Department of Cardiology, Instituto de Investigación Sanitaria Hospital Universitario de La Princesa (IIS-Princesa), Madrid, Spain
| | - Borja Ibañez
- CIBER de Enfermedades Cardiovasculares (CIBERCV), Instituto de Salud Carlos III, Madrid, Spain
- Myocardial Pathophysiology Area, Centro Nacional de Investigaciones Cardiovasculares (CNIC), Madrid, Spain
- Department of Cardiology, Instituto de Investigación Sanitaria Fundación Jiménez Díaz (IIS-FJD), Madrid, Spain
| | - Joaquín Sastre
- Department of Allergy and Immunology, Instituto de Investigación Sanitaria Fundación Jiménez Díaz (IIS-FJD), Universidad Autónoma de Madrid (UAM), Madrid, Spain
- CIBER de Enfermedades Respiratorias (CIBERES), Instituto de Salud Carlos III, Madrid, Spain
| | - María Teresa Belver
- Department of Allergy, Instituto de Investigación Sanitaria Hospital Universitario de La Princesa (IIS-Princesa), Madrid, Spain
| | - Francisco Vega
- Department of Allergy, Instituto de Investigación Sanitaria Hospital Universitario de La Princesa (IIS-Princesa), Madrid, Spain
| | - Carlos Blanco
- Department of Allergy, Instituto de Investigación Sanitaria Hospital Universitario de La Princesa (IIS-Princesa), Madrid, Spain
| | - Domingo Barber
- Department of Basic Medical Sciences, Faculty of Medicine, Institute of Applied Molecular Medicine Nemesio Díez (IMMA), Universidad San Pablo-CEU, CEU Universities, Madrid, Spain
| | - Francisco Sánchez-Madrid
- Vascular Pathophysiology Area, Centro Nacional de Investigaciones Cardiovasculares (CNIC), Madrid, Spain
- Department of Immunology, Instituto de Investigación Sanitaria Hospital Universitario de La Princesa (IIS-Princesa), Universidad Autónoma de Madrid (UAM), Madrid, Spain
- CIBER de Enfermedades Cardiovasculares (CIBERCV), Instituto de Salud Carlos III, Madrid, Spain
| | - Hortensia de la Fuente
- Department of Immunology, Instituto de Investigación Sanitaria Hospital Universitario de La Princesa (IIS-Princesa), Universidad Autónoma de Madrid (UAM), Madrid, Spain
- CIBER de Enfermedades Cardiovasculares (CIBERCV), Instituto de Salud Carlos III, Madrid, Spain
| | - Pilar Martín
- Vascular Pathophysiology Area, Centro Nacional de Investigaciones Cardiovasculares (CNIC), Madrid, Spain
- CIBER de Enfermedades Cardiovasculares (CIBERCV), Instituto de Salud Carlos III, Madrid, Spain
| | - Vanesa Esteban
- Department of Allergy and Immunology, Instituto de Investigación Sanitaria Fundación Jiménez Díaz (IIS-FJD), Universidad Autónoma de Madrid (UAM), Madrid, Spain
- Faculty of Medicine and Biomedicine, Universidad Alfonso X El Sabio, Madrid, Spain
| | - Rodrigo Jiménez-Saiz
- Department of Immunology, Instituto de Investigación Sanitaria Hospital Universitario de La Princesa (IIS-Princesa), Universidad Autónoma de Madrid (UAM), Madrid, Spain
- Department of Immunology and Oncology, Centro Nacional de Biotecnología (CNB)-CSIC, Madrid, Spain
- Faculty of Experimental Sciences, Universidad Francisco de Vitoria (UFV), Madrid, Spain
- Department of Medicine, McMaster Immunology Research Centre (MIRC), McMaster University, Hamilton, Ontario, Canada
| |
Collapse
|
27
|
Márquez-Sánchez AC, Koltsova EK. Immune and inflammatory mechanisms of abdominal aortic aneurysm. Front Immunol 2022; 13:989933. [PMID: 36275758 PMCID: PMC9583679 DOI: 10.3389/fimmu.2022.989933] [Citation(s) in RCA: 55] [Impact Index Per Article: 18.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2022] [Accepted: 08/23/2022] [Indexed: 11/13/2022] Open
Abstract
Abdominal aortic aneurysm (AAA) is a life-threatening cardiovascular disease. Immune-mediated infiltration and a destruction of the aortic wall during AAA development plays significant role in the pathogenesis of this disease. While various immune cells had been found in AAA, the mechanisms of their activation and function are still far from being understood. A better understanding of mechanisms regulating the development of aberrant immune cell activation in AAA is essential for the development of novel preventive and therapeutic approaches. In this review we summarize current knowledge about the role of immune cells in AAA and discuss how pathogenic immune cell activation is regulated in this disease.
Collapse
|
28
|
Wang B, Hou X, Sun Y, Lei C, Yang S, Zhu Y, Jiang Y, Song L. Interleukin-17A influences the vulnerability rather than the size of established atherosclerotic plaques in apolipoprotein E-deficient mice. Open Life Sci 2022; 17:1104-1115. [PMID: 36133421 PMCID: PMC9462543 DOI: 10.1515/biol-2022-0072] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2021] [Revised: 03/08/2022] [Accepted: 03/15/2022] [Indexed: 11/15/2022] Open
Abstract
Interleukin (IL)-17A plays a role in the development of atherosclerotic plaques; however, the mechanism remains unclear. In this study, apolipoprotein E-deficient (ApoE–/–) mice were fed a high-fat diet to induce atherosclerosis, followed by the treatment with exogenous recombinant IL-17A or the neutralizing antibody to confirm the impact of IL-17A on the established atherosclerotic plaques. We found that both the stimulation of IL-17A and blockage of endogenous IL-17 via antibody did not affect the size of the established plaques. However, IL-17A significantly increased the vulnerability of plaques characterized by the accumulation of lipids and T cells with a concurrent decrease in the number of smooth muscle cells. In addition, the blockage by IL-17 neutralizing antibody attenuated plaque vulnerability. Furthermore, we found that although IL-17A did not affect the efferocytosis of macrophages to apoptotic cells, it promoted the apoptosis of macrophages in the presence of oxidized low-density lipoprotein in vitro. Also, IL-17A upregulated chemokines MCP-1 and CXCL-10 expression in the plaques. Our data indicated that IL-17A controlled both SMC and macrophage accumulation and the apoptosis within the plaque, which may further weaken the aorta wall. This study suggests that IL-17A may be a potential therapeutic target for cardiovascular diseases.
Collapse
Affiliation(s)
- Bo Wang
- Institute of Forensic Medicine and Laboratory Medicine, Jining Medical University, Jining City, 272067, Shandong Province, China
| | - Xitan Hou
- Institute of Forensic Medicine and Laboratory Medicine, Jining Medical University, Jining City, 272067, Shandong Province, China
| | - Yaning Sun
- Institute of Forensic Medicine and Laboratory Medicine, Jining Medical University, Jining City, 272067, Shandong Province, China
| | - Chao Lei
- Institute of Forensic Medicine and Laboratory Medicine, Jining Medical University, Jining City, 272067, Shandong Province, China
| | - Sha Yang
- Institute of Forensic Medicine and Laboratory Medicine, Jining Medical University, Jining City, 272067, Shandong Province, China
| | - Yao Zhu
- Institute of Forensic Medicine and Laboratory Medicine, Jining Medical University, Jining City, 272067, Shandong Province, China
| | - Yingming Jiang
- Institute of Forensic Medicine and Laboratory Medicine, Jining Medical University, Jining City, 272067, Shandong Province, China
| | - Li Song
- Department of Neurology, Affiliated Hospital of Jining Medical University, Jining City, 272067, Shandong Province, China
| |
Collapse
|
29
|
Wang Q, Wang Y, Xu D. Research progress on Th17 and T regulatory cells and their cytokines in regulating atherosclerosis. Front Cardiovasc Med 2022; 9:929078. [PMID: 36211578 PMCID: PMC9534355 DOI: 10.3389/fcvm.2022.929078] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2022] [Accepted: 08/17/2022] [Indexed: 11/13/2022] Open
Abstract
Background Coronary heart disease due to atherosclerosis is the leading cause of death worldwide. Atherosclerosis is considered a chronic inflammatory state in the arterial wall that promotes disease progression and outcome, and immune cells play an important role in the inflammatory process. Purpose We review the mechanisms of CD4+ T subsets, i.e., helper T17 (Th17) cells and regulatory T cells (Tregs), in regulating atherosclerosis, focusing on the role of interleukin (IL)-17, IL-10, and other cytokines in this disease and the factors influencing the effects of these cytokines. Results IL-17 secreted by Th17 cells can promote atherosclerosis, but few studies have reported that IL-17 can also stabilize atherosclerotic plaques. Tregs play a protective role in atherosclerosis, and Th17/Treg imbalance also plays an important role in atherosclerosis. Conclusion The immune response is important in regulating atherosclerosis, and studying the mechanism of action of each immune cell on atherosclerosis presents directions for the treatment of atherosclerosis. Nevertheless, the current studies are insufficient for elucidating the mechanism of action, and further in-depth studies are needed to provide a theoretical basis for clinical drug development.
Collapse
Affiliation(s)
- Qiong Wang
- Department of Cardiovascular Medicine, The Second Xiangya Hospital, Central South University, Changsha, China
| | - Yurong Wang
- Department of Cardiovascular Medicine, The Second Xiangya Hospital, Central South University, Changsha, China
| | - Danyan Xu
- Department of Internal Cardiovascular Medicine, Second Xiangya Hospital, Central South University, Changsha, China
| |
Collapse
|
30
|
Looking beyond the Skin: Pathophysiology of Cardiovascular Comorbidity in Psoriasis and the Protective Role of Biologics. Pharmaceuticals (Basel) 2022; 15:ph15091101. [PMID: 36145322 PMCID: PMC9503011 DOI: 10.3390/ph15091101] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2022] [Revised: 08/23/2022] [Accepted: 08/25/2022] [Indexed: 11/16/2022] Open
Abstract
Psoriasis is a chronic systemic inflammatory disease associated with a higher incidence of cardiovascular disease, especially in patients with moderate to severe psoriasis. It has been estimated that severe psoriasis confers a 25% increase in relative risk of cardiovascular disease, regardless of traditional risk factors. Although the underlying pathogenic mechanisms relating psoriasis to increased cardiovascular risk are not clear, atherosclerosis is emerging as a possible link between skin and vascular affection. The hypothesis that the inflammatory cascade activated in psoriasis contributes to the atherosclerotic process provides the underlying basis to suggest that an anti-inflammatory therapy that improved atherosclerosis would also reduce the risk of MACEs. In this sense, the introduction of biological drugs which specifically target cytokines implicated in the inflammatory cascade have increased the expectations of control over the cardiovascular comorbidity present in psoriasis patients, however, their role in vascular damage processes remains controversial. The aim of this paper is to review the mechanistic link between psoriasis and cardiovascular disease development, as well as analyzing which of the biological treatments could also reduce the cardiovascular risk in these patients, fueling a growing debate on the modification of the general algorithm of treatment.
Collapse
|
31
|
Li X, Hu H, Guo D, Hu Y, Zhou H, Chen Y, Fang X. Imbalance of Pro- and Anti-inflammatory Cytokines Induced Different Types of Recurrent Atrial Arrhythmias after Drug Eluting Coronary Stent Implantation. Curr Vasc Pharmacol 2022; 20:447-456. [PMID: 36045517 DOI: 10.2174/1570161120666220831094507] [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: 03/07/2022] [Revised: 06/08/2022] [Accepted: 07/27/2022] [Indexed: 01/25/2023]
Abstract
BACKGROUND Atrial arrhythmias are associated with an increased risk of stroke and death in the elderly. The risk and predictive factors of recurrent atrial arrhythmias in elderly patients after coronary stenting are not well known. OBJECTIVE This research sought to investigate the roles of pro- and anti-inflammatory cytokine imbalances in different types of recurrent atrial arrhythmias in elderly patients defined as individuals aged 65 years or older after sirolimus eluting stent (Cordis, Warren, New Jersey) implantation. METHODS We measured interleukin-1β (IL-1β), interleukin-6 (IL-6), interleukin-8 (IL-8), tumor necrosis factor-α (TNF-α), interleukin-10 (IL-10), interleukin-17 (IL-17), interleukin-13 (IL-13) and interleukin- 37 (IL-37) in elderly patients with recurrent atrial arrhythmias and assessed the impact of pro- and antiinflammatory cytokine imbalances on recurrent atrial arrhythmias in elderly patients after coronary stenting. RESULTS Levels of IL-1 β, IL-6, IL-8, and TNF-α were remarkably increased (p<0.001), and IL-10, IL- 17, IL-13, and IL-37 were remarkably lowered (p<0.001) in elderly patients with recurrent atrial arrhythmias after coronary stent implantation. Imbalance of pro- and anti-inflammatory cytokines induced recurrent atrial arrhythmias after coronary stenting. Pro- and anti-inflammatory cytokine imbalances may be used to identify elderly patients who have an increased risk of developing recurrent atrial arrhythmias after coronary stenting. CONCLUSION The imbalance of pro- and anti-inflammatory cytokines was associated with recurrent atrial arrhythmias in elderly patients after coronary stenting. Pro- and anti-inflammatory cytokines may be clinically useful biomarkers for predicting recurrent atrial arrhythmias in elderly patients after coronary stent implantation.
Collapse
Affiliation(s)
- Xia Li
- Xiamen Road Branch Hospital, The Affiliated Huaian Hospital of Xuzhou Medical University, Huaian 223002, China
| | - Haibo Hu
- Xiamen Road Branch Hospital, The Affiliated Huaian Hospital of Xuzhou Medical University, Huaian 223002, China
| | - Dianxuan Guo
- Department of Geriatrics, The Affiliated Huaian Hospital of Xuzhou Medical University, Huaian 223002, China
| | - Youdong Hu
- Department of Geriatrics, The Affiliated Huaian Hospital of Xuzhou Medical University, Huaian 223002, China
| | - Hualan Zhou
- Department of Geriatrics, The Affiliated Huaian Hospital of Xuzhou Medical University, Huaian 223002, China
| | - Ying Chen
- Department of Geriatrics, The Affiliated Huaian Hospital of Xuzhou Medical University, Huaian 223002, China
| | - Xiang Fang
- Department of Geriatrics, The Affiliated Huaian Hospital of Xuzhou Medical University, Huaian 223002, China
| |
Collapse
|
32
|
Piaserico S, Orlando G, Messina F. Psoriasis and Cardiometabolic Diseases: Shared Genetic and Molecular Pathways. Int J Mol Sci 2022; 23:9063. [PMID: 36012327 PMCID: PMC9409274 DOI: 10.3390/ijms23169063] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2022] [Revised: 08/09/2022] [Accepted: 08/10/2022] [Indexed: 11/26/2022] Open
Abstract
A convincing deal of evidence supports the fact that severe psoriasis is associated with cardiovascular diseases. However, the precise underlying mechanisms linking psoriasis and cardiovascular diseases are not well defined. Psoriasis shares common pathophysiologic mechanisms with atherosclerosis and cardiovascular (CV) risk factors. In particular, polymorphism in the IL-23R and IL-23 genes, as well as other genes involved in lipid and fatty-acid metabolism, renin-angiotensin system and endothelial function, have been described in patients with psoriasis and with cardiovascular risk factors. Moreover, systemic inflammation in patients with psoriasis, including elevated serum proinflammatory cytokines (e.g., TNF-α, IL-17, and IL-23) may contribute to an increased risk of atherosclerosis, hypertension, alteration of serum lipid composition, and insulin resistance. The nonlinear and intricate interplay among various factors, impacting the molecular pathways in different cell types, probably contributes to the development of psoriasis and cardiovascular disease (CVD). Future research should, therefore, aim to fully unravel shared and differential molecular pathways underpinning the association between psoriasis and CVD.
Collapse
Affiliation(s)
- Stefano Piaserico
- Unit of Dermatology, Department of Medicine, University of Padua, Via V. Gallucci 4, 35128 Padua, Italy
| | | | | |
Collapse
|
33
|
Tan HB, Zheng YQ, Zhuang YP. IL-17A in diabetic kidney disease: protection or damage. Int Immunopharmacol 2022; 108:108707. [PMID: 35344813 DOI: 10.1016/j.intimp.2022.108707] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2022] [Revised: 03/05/2022] [Accepted: 03/12/2022] [Indexed: 12/12/2022]
Abstract
The effect of IL-17A in diabetic kidney disease (DKD) has received increasing attention. Interleukin (IL)-17A promotes renal inflammation and the progression of DKD, and IL-17A deficiency improves experimental DKD. However, recent studies have found that the effect of IL-17A on DKD is more complicated than the negative impact. IL-17A alleviates renal inflammation and fibrosis via regulating autophagy or the macrophage phenotype. Moreover, paradoxical expression of IL-17A has been reported in human DKD. This review focuses on how IL-17A affects the progression of DKD and the resulting opportunities and challenges.
Collapse
Affiliation(s)
- Hai-Bo Tan
- Institute of Chinese Medicine, Guangdong Pharmaceutical University, Guangzhou 510006, PR China.
| | - Yan-Qiu Zheng
- Pi-Wei Institute, Science and Technology Innovation Center, Guangzhou University of Chinese Medicine, Guangzhou 510006, PR China
| | - Yu-Pei Zhuang
- Department of Gastroenterology, The First Affiliated Hospital of Guangdong Pharmaceutical University, Guangzhou 510006, PR China
| |
Collapse
|
34
|
Cimmino G, Di Serafino L, Cirillo P. Pathophysiology and mechanisms of Acute Coronary Syndromes: atherothrombosis, immune-inflammation, and beyond. Expert Rev Cardiovasc Ther 2022; 20:351-362. [PMID: 35510629 DOI: 10.1080/14779072.2022.2074836] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/26/2022] [Accepted: 05/04/2022] [Indexed: 12/24/2022]
Abstract
INTRODUCTION The pathophysiology of atherosclerosis and its acute complications, such as the Acute Coronary Syndromes (ACS), is continuously under investigation. Immunity and inflammation seem to play a pivotal role in promoting formation and grow of atherosclerotic plaques. At the same time, plaque rupture followed by both platelets' activation and coagulation cascade induction lead to intracoronary thrombus formation. Although these phenomena might be considered responsible of about 90% of ACS, in up to 5-10% of acute syndromes, a non-obstructive coronary artery disease (MINOCA) might be documented. This paper gives an overview on atherothrombosis and immuno-inflammation processes involved in ACS pathophysiology, also emphasizing the pathological mechanisms potentially involved in MINOCA. AREAS COVERED The relationship between immuno-inflammation and atherothrombosis is continuously updated by recent findings. At the same time, pathophysiology of MINOCA still remains a partially unexplored field, stimulating the research of potential links between these two aspects of ACS pathophysiology. EXPERT OPINION Pathophysiology of ACS has been extensively investigated; however, several gray areas still remain. MINOCA represents one of these areas. At the same time, many aspects of immune-inflammation processes are still unknown. Thus, research should be continued to shed a brighter light on both these sides of "ACS" moon.
Collapse
Affiliation(s)
- Giovanni Cimmino
- Department of Translational Medical Sciences, Section of Cardiology, University of Campania "Luigi Vanvitelli", Naples, Italy
| | - Luigi Di Serafino
- Department of Advanced Biomedical Sciences, University of Naples Federico II, Naples, Italy
| | - Plinio Cirillo
- Department of Advanced Biomedical Sciences, University of Naples Federico II, Naples, Italy
| |
Collapse
|
35
|
Kong P, Cui ZY, Huang XF, Zhang DD, Guo RJ, Han M. Inflammation and atherosclerosis: signaling pathways and therapeutic intervention. Signal Transduct Target Ther 2022; 7:131. [PMID: 35459215 PMCID: PMC9033871 DOI: 10.1038/s41392-022-00955-7] [Citation(s) in RCA: 475] [Impact Index Per Article: 158.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2021] [Revised: 03/01/2022] [Accepted: 03/02/2022] [Indexed: 02/08/2023] Open
Abstract
Atherosclerosis is a chronic inflammatory vascular disease driven by traditional and nontraditional risk factors. Genome-wide association combined with clonal lineage tracing and clinical trials have demonstrated that innate and adaptive immune responses can promote or quell atherosclerosis. Several signaling pathways, that are associated with the inflammatory response, have been implicated within atherosclerosis such as NLRP3 inflammasome, toll-like receptors, proprotein convertase subtilisin/kexin type 9, Notch and Wnt signaling pathways, which are of importance for atherosclerosis development and regression. Targeting inflammatory pathways, especially the NLRP3 inflammasome pathway and its regulated inflammatory cytokine interleukin-1β, could represent an attractive new route for the treatment of atherosclerotic diseases. Herein, we summarize the knowledge on cellular participants and key inflammatory signaling pathways in atherosclerosis, and discuss the preclinical studies targeting these key pathways for atherosclerosis, the clinical trials that are going to target some of these processes, and the effects of quelling inflammation and atherosclerosis in the clinic.
Collapse
Affiliation(s)
- Peng Kong
- Department of Biochemistry and Molecular Biology, College of Basic Medicine, Key Laboratory of Medical Biotechnology of Hebei Province, Key Laboratory of Neural and Vascular Biology of Ministry of Education, Hebei Medical University, Shijiazhuang, 050017, PR China
| | - Zi-Yang Cui
- Department of Biochemistry and Molecular Biology, College of Basic Medicine, Key Laboratory of Medical Biotechnology of Hebei Province, Key Laboratory of Neural and Vascular Biology of Ministry of Education, Hebei Medical University, Shijiazhuang, 050017, PR China
| | - Xiao-Fu Huang
- Department of Biochemistry and Molecular Biology, College of Basic Medicine, Key Laboratory of Medical Biotechnology of Hebei Province, Key Laboratory of Neural and Vascular Biology of Ministry of Education, Hebei Medical University, Shijiazhuang, 050017, PR China
| | - Dan-Dan Zhang
- Department of Biochemistry and Molecular Biology, College of Basic Medicine, Key Laboratory of Medical Biotechnology of Hebei Province, Key Laboratory of Neural and Vascular Biology of Ministry of Education, Hebei Medical University, Shijiazhuang, 050017, PR China
| | - Rui-Juan Guo
- Department of Biochemistry and Molecular Biology, College of Basic Medicine, Key Laboratory of Medical Biotechnology of Hebei Province, Key Laboratory of Neural and Vascular Biology of Ministry of Education, Hebei Medical University, Shijiazhuang, 050017, PR China
| | - Mei Han
- Department of Biochemistry and Molecular Biology, College of Basic Medicine, Key Laboratory of Medical Biotechnology of Hebei Province, Key Laboratory of Neural and Vascular Biology of Ministry of Education, Hebei Medical University, Shijiazhuang, 050017, PR China.
| |
Collapse
|
36
|
Moeinafshar A, Razi S, Rezaei N. Interleukin 17, the double-edged sword in atherosclerosis. Immunobiology 2022; 227:152220. [PMID: 35452921 DOI: 10.1016/j.imbio.2022.152220] [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: 06/04/2021] [Revised: 04/07/2022] [Accepted: 04/14/2022] [Indexed: 11/05/2022]
Abstract
Cardiovascular diseases, including atherosclerosis, are the number one cause of death worldwide. These diseases have taken the place of pneumonia and other infectious diseases in the epidemiological charts. Thus, their importance should not be underestimated. Atherosclerosis is an inflammatory disease. Therefore, immunological signaling molecules and immune cells carry out a central role in its etiology. One of these signaling molecules is interleukin (IL)-17. This relatively newly discovered signaling molecule might have a dual role as acting both pro-atherogenic and anti-atherogenic depending on the situation. The majority of articles have discussed IL-17 and its action in atherosclerosis, and it may be a new target for the treatment of patients with this disease. In this review, the immunological basis of atherosclerosis with an emphasis on the role of IL-17 and a brief explanation of the role of IL-17 on atherosclerogenic disorders will be discussed.
Collapse
Affiliation(s)
- Aysan Moeinafshar
- School of Medicine, Tehran University of Medical Sciences, Tehran, Iran; Cancer Immunology Project (CIP), Universal Scientific Education and Research Network (USERN), Tehran, Iran
| | - Sepideh Razi
- Cancer Immunology Project (CIP), Universal Scientific Education and Research Network (USERN), Tehran, Iran; School of Medicine, Iran University of Medical Sciences, Tehran, Iran; Network of Immunity in Infection, Malignancy and Autoimmunity (NIIMA), Universal Scientific Education and Research Network (USERN), Tehran, Iran
| | - Nima Rezaei
- Network of Immunity in Infection, Malignancy and Autoimmunity (NIIMA), Universal Scientific Education and Research Network (USERN), Tehran, Iran; Research Center for Immunodeficiencies, Children's Medical Center, Tehran University of Medical Sciences, Tehran, Iran; Department of Immunology, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
37
|
Wang J, He L, Li W, Lv S. A Role of IL-17 in Rheumatoid Arthritis Patients Complicated With Atherosclerosis. Front Pharmacol 2022; 13:828933. [PMID: 35211020 PMCID: PMC8861488 DOI: 10.3389/fphar.2022.828933] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2021] [Accepted: 01/19/2022] [Indexed: 12/14/2022] Open
Abstract
Rheumatoid arthritis (RA) is mainly caused by joint inflammation. RA significantly increases the probability of cardiovascular disease. Although the progress of RA has been well controlled recently, the mortality of patients with RA complicated with cardiovascular disease is 1.5–3 times higher than that of patients with RA alone. The number of people with atherosclerosis in patients with RA is much higher than that in the general population, and atherosclerotic lesions develop more rapidly in patients with RA, which has become one of the primary factors resulting in the death of patients with RA. The rapid development of atherosclerosis in RA is induced by inflammation-related factors. Recent studies have reported that the expression of IL-17 is significantly upregulated in patients with RA and atherosclerosis. Simultaneously, there is evidence that IL-17 can regulate the proliferation, migration, and apoptosis of vascular endothelial cells and vascular smooth muscle cells through various ways and promote the secretion of several cytokines leading to the occurrence and development of atherosclerosis. Presently, there is no clear prevention or treatment plan for atherosclerosis in patients with RA. Therefore, this paper explores the mechanism of IL-17 in RA complicated with atherosclerosis and shows the reasons for the high incidence of atherosclerosis in patients with RA. It is hoped that the occurrence and development of atherosclerosis in patients with RA can be diagnosed or prevented in time in the early stage of lesions, and the prevention and treatment of cardiovascular complications in patients with RA can be enhanced to reduce mortality.
Collapse
Affiliation(s)
- Jiexin Wang
- Basic Medical College, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Linxi He
- Basic Medical College, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Weihong Li
- Basic Medical College, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Shangbin Lv
- Basic Medical College, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| |
Collapse
|
38
|
Bonek K, Kuca-Warnawin E, Kornatka A, Zielińska A, Wisłowska M, Kontny E, Głuszko P. Associations of IL-18 with Altered Cardiovascular Risk Profile in Psoriatic Arthritis and Ankylosing Spondylitis. J Clin Med 2022; 11:jcm11030766. [PMID: 35160217 PMCID: PMC8836492 DOI: 10.3390/jcm11030766] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2021] [Revised: 01/27/2022] [Accepted: 01/28/2022] [Indexed: 12/02/2022] Open
Abstract
Objective: To investigate the associations of IL-18 serum levels with serum lipids, cardiovascular risk, and disease activity in patients with ankylosing spondylitis (AS) and psoriatic arthritis (PsA) with axial (axPsA) and peripheral (perPsA) joint involvement. Methods: 155 adult patients (PsA 61/AS 94) were enrolled in the study. Standard disease activity indices, BASDAI, and ASDAS, were calculated for AS and PsA and DAPSA for PsA. Sera from peripheral blood samples were obtained after night fasting. Serum concentrations of cytokines (IL-18, IL-17) were measured by ELISA, while lipid profile with total cholesterol (TC), triglycerides (TG), low-density cholesterol-(LDL), high-density cholesterol (HDL), and C-reactive protein (CRP) concentrations were determined using routine procedures. The atherogenic index was calculated using the standard formula AI = TC/HDL. Results: Patients with PsA and peripheral joint involvement (perPsA) had significantly higher IL-18 serum levels than axial PsA and AS patients (medians 160 vs. 116 vs. 80 pg/mL). In patients with PsA and in the subgroup with PsA+ ischemic heart disease (IHD), IL-18 positively correlated with atherogenic index (AI) (rho = 0.46 and rho = 0.67, respectively) and TG serum concentrations (rho = 0.4 and rho = 0.675), while negatively with HDL levels (rho = −0.37 and rho = −0.608). In PsA + IHD subgroup IL-18 serum levels correlated positively also with disease activity (DAPSA) (rho = 0.613). Importantly, in patients with perPsA, characterized by the highest IL-18 serum levels, cardiovascular risk, and frequency of both hypertriglyceridemia and IHD, positive correlations between IL-18 and IL-17 (rho = 0.47, p = 0.002), TG (rho = 0.45 p = 0.01) levels and AI (rho = 0.63 p = 0.021) were found. Whereas linear regression models revealed that IL-17, TG concentrations and the tender joint count had an impact on IL-18 Conclusions: We confirmed that patients with perPsA are characterized by a more pronounced proinflammatory and proatherogenic cardiovascular risk profile than patients with axPsA and AS. Importantly our study indicates that in PsA, but not in AS, elevated serum concentration of IL-18 is associated with higher disease activity and proatherogenic lipid profile, leading to a higher cardiovascular risk. Thus, our results point out IL-18 as a critical contributor in these pathological processes and possible therapeutic targets.
Collapse
Affiliation(s)
- Krzysztof Bonek
- Department of Rheumatology, National Institute of Geriatrics, Rheumatology and Rehabilitation, 02-637 Warsaw, Poland; (A.Z.); (M.W.); (P.G.)
- Correspondence:
| | - Ewa Kuca-Warnawin
- Department of Pathophysiology and Immunology, National Institute of Geriatrics, Rheumatology and Rehabilitation, 02-637 Warsaw, Poland; (E.K.-W.); (A.K.); (E.K.)
| | - Anna Kornatka
- Department of Pathophysiology and Immunology, National Institute of Geriatrics, Rheumatology and Rehabilitation, 02-637 Warsaw, Poland; (E.K.-W.); (A.K.); (E.K.)
| | - Agnieszka Zielińska
- Department of Rheumatology, National Institute of Geriatrics, Rheumatology and Rehabilitation, 02-637 Warsaw, Poland; (A.Z.); (M.W.); (P.G.)
| | - Małgorzata Wisłowska
- Department of Rheumatology, National Institute of Geriatrics, Rheumatology and Rehabilitation, 02-637 Warsaw, Poland; (A.Z.); (M.W.); (P.G.)
| | - Ewa Kontny
- Department of Pathophysiology and Immunology, National Institute of Geriatrics, Rheumatology and Rehabilitation, 02-637 Warsaw, Poland; (E.K.-W.); (A.K.); (E.K.)
| | - Piotr Głuszko
- Department of Rheumatology, National Institute of Geriatrics, Rheumatology and Rehabilitation, 02-637 Warsaw, Poland; (A.Z.); (M.W.); (P.G.)
| |
Collapse
|
39
|
Inflammatory Cells in Atherosclerosis. Antioxidants (Basel) 2022; 11:antiox11020233. [PMID: 35204116 PMCID: PMC8868126 DOI: 10.3390/antiox11020233] [Citation(s) in RCA: 49] [Impact Index Per Article: 16.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2021] [Revised: 01/21/2022] [Accepted: 01/24/2022] [Indexed: 02/06/2023] Open
Abstract
Atherosclerosis is a chronic progressive disease that involves damage to the intima, inflammatory cell recruitment and the accumulation of lipids followed by calcification and plaque rupture. Inflammation is considered a key mediator of many events during the development and progression of the disease. Various types of inflammatory cells are reported to be involved in atherosclerosis. In the present paper, we discuss the involved inflammatory cells, their characteristic and functional significance in the development and progression of atherosclerosis. The detailed understanding of the role of all these cells in disease progression at different stages sheds more light on the subject and provides valuable insights as to where and when therapy should be targeted.
Collapse
|
40
|
Affi R, Gabillard D, Kouame GM, Ntakpe JB, Moh R, Badje A, Danel C, Inwoley A, Eholié SP, Anglaret X, Weiss L. Plasma sVCAM-1, antiretroviral therapy and mortality in HIV-1-infected West African adults. HIV Med 2022; 23:717-726. [PMID: 35023284 DOI: 10.1111/hiv.13230] [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: 06/18/2021] [Revised: 12/06/2021] [Accepted: 12/23/2021] [Indexed: 11/30/2022]
Abstract
OBJECTIVES We report the association between pre-antiretroviral therapy (pre-ART) soluble vascular cell adhesion molecule-1 (sVCAM-1) levels and long-term mortality in HIV-infected West African adults participating in a trial of early ART in West Africa (Temprano ANRS 12136 trial). METHODS The ART-naïve HIV-infected adults were randomly assigned to start ART immediately or defer ART until the WHO criteria were met. Participants who completed the trial follow-up were invited to participate in a post-trial phase (PTP). The PTP end-point was all-cause death. We used multivariable Cox proportional models to analyse the association between baseline sVCAM-1 and all-cause death, adjusting for ART strategy, sex, CD4 count, plasma HIV-1 RNA and peripheral blood mononuclear cell HIV-1 DNA levels. RESULTS In all, 954 adults (77% women, median CD4 count of 387 cells/μL) were randomly assigned to start ART immediately (n = 477) or to defer initiation of ART (n = 477). They were followed for a median of 5.8 years [interquartile range (IQR): 5.2-6.3]. In multivariable analysis, the risk of death was significantly associated with baseline sVCAM-1 [≥1458 vs. < 1458 ng/mL; adjusted hazard ratio = 2.86, 95% confidence interval (CI): 1.60-5.11]. The 6-year probability of death rates were 14.4% (95%CI: 9.1-22.6) and 9.4% (5.4-16.1) in patients with baseline sVCAM-1 ≥ 1458 ng/mL randomized to deferred and immediate ART, respectively, and 3.8% (2.2-6.5) and 3.5% (1.9-6.3) in patients with baseline sVCAM-1 < 1458 ng/mL randomized to deferred and immediate ART. The median difference between pre-ART and 12-month sVCAM-1 levels in patients randomized to immediate ART was -252 (IQR: -587 to -61). CONCLUSIONS Pre-ART sVCAM-1 levels were significantly associated with mortality, independently of whether ART was started immediately or deferred, but they significantly decreased after 12 months of ART.
Collapse
Affiliation(s)
- Roseline Affi
- CeDReS, CHU de Treichville, Abidjan, Côte d'Ivoire.,PACCI/ANRS Research site in Côte d'Ivoire, Abidjan, Côte d'Ivoire.,Université Felix Houphouët Boigny, Abidjan, Côte d'Ivoire
| | - Delphine Gabillard
- PACCI/ANRS Research site in Côte d'Ivoire, Abidjan, Côte d'Ivoire.,INSERM U1219, University of Bordeaux, IRD, Bordeaux, France
| | - Gérard Menan Kouame
- PACCI/ANRS Research site in Côte d'Ivoire, Abidjan, Côte d'Ivoire.,INSERM U1219, University of Bordeaux, IRD, Bordeaux, France
| | - Jean Baptiste Ntakpe
- PACCI/ANRS Research site in Côte d'Ivoire, Abidjan, Côte d'Ivoire.,INSERM U1219, University of Bordeaux, IRD, Bordeaux, France
| | - Raoul Moh
- PACCI/ANRS Research site in Côte d'Ivoire, Abidjan, Côte d'Ivoire.,Université Felix Houphouët Boigny, Abidjan, Côte d'Ivoire
| | - Anani Badje
- PACCI/ANRS Research site in Côte d'Ivoire, Abidjan, Côte d'Ivoire.,Université Felix Houphouët Boigny, Abidjan, Côte d'Ivoire
| | - Christine Danel
- PACCI/ANRS Research site in Côte d'Ivoire, Abidjan, Côte d'Ivoire.,INSERM U1219, University of Bordeaux, IRD, Bordeaux, France
| | - André Inwoley
- CeDReS, CHU de Treichville, Abidjan, Côte d'Ivoire.,PACCI/ANRS Research site in Côte d'Ivoire, Abidjan, Côte d'Ivoire.,Université Felix Houphouët Boigny, Abidjan, Côte d'Ivoire
| | - Serge P Eholié
- PACCI/ANRS Research site in Côte d'Ivoire, Abidjan, Côte d'Ivoire.,Université Felix Houphouët Boigny, Abidjan, Côte d'Ivoire
| | - Xavier Anglaret
- PACCI/ANRS Research site in Côte d'Ivoire, Abidjan, Côte d'Ivoire.,INSERM U1219, University of Bordeaux, IRD, Bordeaux, France
| | - Laurence Weiss
- Université de Paris, Faculté de Santé, UFR de Médecine, Paris, France.,Service d'Immunologie Clinique, Hôpital Hôtel Dieu, AP-HP, Paris, France.,Institut de Recherche Saint Louis, INSERM U976, Paris, France
| |
Collapse
|
41
|
La Manna S, De Benedictis I, Marasco D. Proteomimetics of Natural Regulators of JAK-STAT Pathway: Novel Therapeutic Perspectives. Front Mol Biosci 2022; 8:792546. [PMID: 35047557 PMCID: PMC8762217 DOI: 10.3389/fmolb.2021.792546] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2021] [Accepted: 11/29/2021] [Indexed: 12/16/2022] Open
Abstract
The JAK-STAT pathway is a crucial cellular signaling cascade, including an intricate network of Protein-protein interactions (PPIs) responsible for its regulation. It mediates the activities of several cytokines, interferons, and growth factors and transduces extracellular signals into transcriptional programs to regulate cell growth and differentiation. It is essential for the development and function of both innate and adaptive immunities, and its aberrant deregulation was highlighted in neuroinflammatory diseases and in crucial mechanisms for tumor cell recognition and tumor-induced immune escape. For its involvement in a multitude of biological processes, it can be considered a valuable target for the development of drugs even if a specific focus on possible side effects associated with its inhibition is required. Herein, we review the possibilities to target JAK-STAT by focusing on its natural inhibitors as the suppressor of cytokine signaling (SOCS) proteins. This protein family is a crucial checkpoint inhibitor in immune homeostasis and a valuable target in immunotherapeutic approaches to cancer and immune deficiency disorders.
Collapse
Affiliation(s)
| | | | - Daniela Marasco
- Department of Pharmacy, University of Naples “Federico II”, Naples, Italy
| |
Collapse
|
42
|
Abstract
Inflammation is intimately involved at all stages of atherosclerosis and remains a substantial residual cardiovascular risk factor in optimally treated patients. The proof of concept that targeting inflammation reduces cardiovascular events in patients with a history of myocardial infarction has highlighted the urgent need to identify new immunotherapies to treat patients with atherosclerotic cardiovascular disease. Importantly, emerging data from new clinical trials show that successful immunotherapies for atherosclerosis need to be tailored to the specific immune alterations in distinct groups of patients. In this Review, we discuss how single-cell technologies - such as single-cell mass cytometry, single-cell RNA sequencing and cellular indexing of transcriptomes and epitopes by sequencing - are ideal for mapping the cellular and molecular composition of human atherosclerotic plaques and how these data can aid in the discovery of new precise immunotherapies. We also argue that single-cell data from studies in humans need to be rigorously validated in relevant experimental models, including rapidly emerging single-cell CRISPR screening technologies and mouse models of atherosclerosis. Finally, we discuss the importance of implementing single-cell immune monitoring tools in early phases of drug development to aid in the precise selection of the target patient population for data-driven translation into randomized clinical trials and the successful translation of new immunotherapies into the clinic.
Collapse
Affiliation(s)
- Dawn M Fernandez
- Department of Medicine, Division of Cardiology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Chiara Giannarelli
- Department of Medicine, Division of Cardiology, Icahn School of Medicine at Mount Sinai, New York, NY, USA.
- New York University Cardiovascular Research Center, New York University Langone Health, New York, NY, USA.
- Department of Pathology, New York University Grossman School of Medicine, New York University Langone Health, New York, NY, USA.
| |
Collapse
|
43
|
van Duijn J, de Jong MJM, Benne N, Leboux RJT, van Ooijen ME, Kruit N, Foks AC, Jiskoot W, Bot I, Kuiper J, Slütter B. Tc17 CD8+ T cells accumulate in murine atherosclerotic lesions, but do not contribute to early atherosclerosis development. Cardiovasc Res 2021; 117:2755-2766. [PMID: 33063097 PMCID: PMC8683708 DOI: 10.1093/cvr/cvaa286] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/11/2020] [Revised: 09/04/2020] [Accepted: 09/30/2020] [Indexed: 01/09/2023] Open
Abstract
AIMS CD8+ T cells can differentiate into subpopulations that are characterized by a specific cytokine profile, such as the Tc17 population that produces interleukin-17. The role of this CD8+ T-cell subset in atherosclerosis remains elusive. In this study, we therefore investigated the contribution of Tc17 cells to the development of atherosclerosis. METHODS AND RESULTS Flow cytometry analysis of atherosclerotic lesions from apolipoprotein E-deficient mice revealed a pronounced increase in RORγt+CD8+ T cells compared to the spleen, indicating a lesion-specific increase in Tc17 cells. To study whether and how the Tc17 subset affects atherosclerosis, we performed an adoptive transfer of Tc17 cells or undifferentiated Tc0 cells into CD8-/- low-density lipoprotein receptor-deficient mice fed a Western-type diet. Using flow cytometry, we showed that Tc17 cells retained a high level of interleukin-17A production in vivo. Moreover, Tc17 cells produced lower levels of interferon-γ than their Tc0 counterparts. Analysis of the aortic root revealed that the transfer of Tc17 cells did not increase atherosclerotic lesion size, in contrast to Tc0-treated mice. CONCLUSION These findings demonstrate a lesion-localized increase in Tc17 cells in an atherosclerotic mouse model. Tc17 cells appeared to be non-atherogenic, in contrast to their Tc0 counterpart.
Collapse
MESH Headings
- Adoptive Transfer
- Animals
- Aorta/immunology
- Aorta/metabolism
- Aorta/pathology
- Aortic Diseases/genetics
- Aortic Diseases/immunology
- Aortic Diseases/metabolism
- Aortic Diseases/pathology
- Atherosclerosis/genetics
- Atherosclerosis/immunology
- Atherosclerosis/metabolism
- Atherosclerosis/pathology
- CD4-Positive T-Lymphocytes/immunology
- CD4-Positive T-Lymphocytes/metabolism
- CD8-Positive T-Lymphocytes/immunology
- CD8-Positive T-Lymphocytes/metabolism
- CD8-Positive T-Lymphocytes/transplantation
- Cell Differentiation
- Cells, Cultured
- Disease Models, Animal
- Interferon-gamma/metabolism
- Interleukin-17/immunology
- Interleukin-17/metabolism
- Mice, Inbred C57BL
- Mice, Knockout, ApoE
- Nuclear Receptor Subfamily 1, Group F, Member 3/immunology
- Nuclear Receptor Subfamily 1, Group F, Member 3/metabolism
- Phenotype
- Plaque, Atherosclerotic
- Signal Transduction
- Mice
Collapse
Affiliation(s)
- Janine van Duijn
- Division of BioTherapeutics, Leiden Academic Centre for Drug Research, Leiden University, Einsteinweg 55, Room EE1.17, 2333 CC Leiden, The Netherlands
| | - Maaike J M de Jong
- Division of BioTherapeutics, Leiden Academic Centre for Drug Research, Leiden University, Einsteinweg 55, Room EE1.17, 2333 CC Leiden, The Netherlands
| | - Naomi Benne
- Division of BioTherapeutics, Leiden Academic Centre for Drug Research, Leiden University, Einsteinweg 55, Room EE1.17, 2333 CC Leiden, The Netherlands
| | - Romain J T Leboux
- Division of BioTherapeutics, Leiden Academic Centre for Drug Research, Leiden University, Einsteinweg 55, Room EE1.17, 2333 CC Leiden, The Netherlands
| | - Marieke E van Ooijen
- Division of BioTherapeutics, Leiden Academic Centre for Drug Research, Leiden University, Einsteinweg 55, Room EE1.17, 2333 CC Leiden, The Netherlands
| | - Nicky Kruit
- Division of BioTherapeutics, Leiden Academic Centre for Drug Research, Leiden University, Einsteinweg 55, Room EE1.17, 2333 CC Leiden, The Netherlands
| | - Amanda C Foks
- Division of BioTherapeutics, Leiden Academic Centre for Drug Research, Leiden University, Einsteinweg 55, Room EE1.17, 2333 CC Leiden, The Netherlands
| | - Wim Jiskoot
- Division of BioTherapeutics, Leiden Academic Centre for Drug Research, Leiden University, Einsteinweg 55, Room EE1.17, 2333 CC Leiden, The Netherlands
| | - Ilze Bot
- Division of BioTherapeutics, Leiden Academic Centre for Drug Research, Leiden University, Einsteinweg 55, Room EE1.17, 2333 CC Leiden, The Netherlands
| | - Johan Kuiper
- Division of BioTherapeutics, Leiden Academic Centre for Drug Research, Leiden University, Einsteinweg 55, Room EE1.17, 2333 CC Leiden, The Netherlands
| | - Bram Slütter
- Division of BioTherapeutics, Leiden Academic Centre for Drug Research, Leiden University, Einsteinweg 55, Room EE1.17, 2333 CC Leiden, The Netherlands
| |
Collapse
|
44
|
Oliveira DC, Oliveira CGC, Mendes EB, Silveira MM, Cabral JV, Ferreira E. Circulating interleukin-17A in patients with acute and chronic coronary syndromes. AMERICAN JOURNAL OF CARDIOVASCULAR DISEASE 2021; 11:704-709. [PMID: 35116182 PMCID: PMC8784676] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Received: 10/09/2021] [Accepted: 11/25/2021] [Indexed: 06/14/2023]
Abstract
INTRODUCTION Interleukin 17 (IL-17) is produced by Th17 and other cells. It is debatable whether IL-17 is atherogenic or atheroprotective. The role of this interleukin in the development and progression of coronary artery disease is unknown. Our aim was to evaluate if there were differences in serum IL-17A levels according to to clinical presentation of coronary artery disease. METHODS This cross-sectional study enrolled 101 patients with acute coronary syndrome (ACS), 100 patients with chronic coronary syndrome (CCS), and 70 healthy volunteers. Blood samples were collected from patients and controls (within 48 h) to analyze IL-17A levels. Clinical characteristics were recorded using questionnaires. This study was approved by the Ethics Committee. RESULTS Comparisons of the clinical characteristics between patients with ACS and CCS revealed the following: mean age (62 ± 12.4 years vs. 63.3 ± 9.8 years, P = 0.4), male (63.4% vs. 58%, P = 0.4), hypertension (85.1% vs. 79%, P = 0.1), dyslipidemia (48% vs. 31%, P = 0.01), diabetes mellitus (47.5% vs. 41%, P = 0.3), previous myocardial infarction (57.4% vs. 40%, P = 0.01), and smoking (29.7% vs. 38%, P = 1). The peripheral concentrations of IL-17A in ACS, CCS and controls were 5.36 ± 8.83, 6.69 ± 17.92, and 6.26 ± 11.13, respectively, with P = 0.6. In addition, the comparison between ACS and CCS showed: 5.36 ± 8.83 vs. 6.69 ± 17.92%, P = 0.3. CONCLUSION The main finding of this study was that circulating IL-17 levels were similar in patients with ACS, CCS, and healthy volunteers. In addition, there was no difference between patients with ACS and those with CCS. Therefore, in patients with ACS and CCS, circulating IL-17A concentrations are low and there were no differences between patients with coronary artery disease and healthy individuals.
Collapse
Affiliation(s)
- Dinaldo C Oliveira
- Hospital das Clínicas, Federal University of PernambucoPernambuco, Brazil
| | | | - Edivaldo B Mendes
- Hospital das Clínicas, Federal University of PernambucoPernambuco, Brazil
| | - Maria M Silveira
- Hospital das Clínicas, Federal University of PernambucoPernambuco, Brazil
| | - João V Cabral
- Hospital das Clínicas, Federal University of PernambucoPernambuco, Brazil
| | | |
Collapse
|
45
|
Cai J, Cui L, Wang Y, Li Y, Zhang X, Shi Y. Cardiometabolic Comorbidities in Patients With Psoriasis: Focusing on Risk, Biological Therapy, and Pathogenesis. Front Pharmacol 2021; 12:774808. [PMID: 34803716 PMCID: PMC8600112 DOI: 10.3389/fphar.2021.774808] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2021] [Accepted: 10/14/2021] [Indexed: 12/23/2022] Open
Abstract
Psoriasis is a chronic inflammatory disease characterized by erythematous scaly plaques, accompanied by systemic damage that leads to the development of multiple comorbidities. In particular, the association between psoriasis and cardiometabolic comorbidities, including cardiovascular diseases (CVDs), obesity, diabetes mellitus, and metabolic syndrome, has been verified in a considerable number of clinical trials. Moreover, the increased risk of cardiometabolic comorbidities positively correlates with psoriasis severity. Biologic therapy targeting inflammatory pathways or cytokines substantially improves the life quality of psoriasis patients and may affect cardiometabolic comorbidities by reducing their incidences. In this review, we focus on exploring the association between cardiometabolic comorbidities and psoriasis, and emphasize the benefits and precautions of biologic therapy in the management of psoriasis with cardiometabolic comorbidities. The pathogenic mechanisms of cardiometabolic comorbidities in psoriasis patients involve common genetic factors, lipid metabolism, insulin resistance, and shared inflammatory pathways such as tumor necrosis factor-α and interleukin-23/Th-17 pathways.
Collapse
Affiliation(s)
- Jiangluyi Cai
- Department of Dermatology, Shanghai Skin Disease Hospital, Tongji University School of Medicine, Shanghai, China.,Institute of Psoriasis, Tongji University School of Medicine, Shanghai, China
| | - Lian Cui
- Institute of Psoriasis, Tongji University School of Medicine, Shanghai, China.,Department of Dermatology, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, China
| | - Yu Wang
- Department of Dermatology, Shanghai Skin Disease Hospital, Tongji University School of Medicine, Shanghai, China.,Institute of Psoriasis, Tongji University School of Medicine, Shanghai, China
| | - Ying Li
- Department of Dermatology, Shanghai Skin Disease Hospital, Tongji University School of Medicine, Shanghai, China.,Institute of Psoriasis, Tongji University School of Medicine, Shanghai, China
| | - Xilin Zhang
- Department of Dermatology, Shanghai Skin Disease Hospital, Tongji University School of Medicine, Shanghai, China.,Institute of Psoriasis, Tongji University School of Medicine, Shanghai, China
| | - Yuling Shi
- Department of Dermatology, Shanghai Skin Disease Hospital, Tongji University School of Medicine, Shanghai, China.,Institute of Psoriasis, Tongji University School of Medicine, Shanghai, China
| |
Collapse
|
46
|
Campanati A, Marani A, Martina E, Diotallevi F, Radi G, Offidani A. Psoriasis as an Immune-Mediated and Inflammatory Systemic Disease: From Pathophysiology to Novel Therapeutic Approaches. Biomedicines 2021; 9:biomedicines9111511. [PMID: 34829740 PMCID: PMC8615182 DOI: 10.3390/biomedicines9111511] [Citation(s) in RCA: 53] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2021] [Revised: 10/10/2021] [Accepted: 10/17/2021] [Indexed: 12/17/2022] Open
Abstract
Psoriasis is an immune-mediated inflammatory disease, with a chronic relapsing-remitting course, which affects 2–3% of the worldwide population. The progressive acquisitions of the inflammatory pathways involved in the development of psoriasis have led to the identification of the key molecules of the psoriatic inflammatory cascade. At the same time, psoriasis therapy has radically evolved with the introduction of target molecules able to modify the natural history of the disease, acting specifically on these inflammatory pathways. For these reasons, biologics have been demonstrated to be drugs able to change the disease’s natural history, as they reduce the inflammatory background to avoid irreversible organ damage and prevent systemic complications. However, several issues related to the use of biologics in patients with systemic comorbidities, remain open. All these data reflect the extraordinary potentiality of biologics, but also the unmet medical need to improve our knowledge on the long-term risk related to continuous use of these drugs, and their administration in special populations. This narrative review aims to highlight both the efficacy and safety profile of biologics in psoriasis, starting from pathophysiology and moving towards their clinical application.
Collapse
|
47
|
Lutgens E, Joffre J, van Os B, Ait-Oufella H. Targeting cytokines and immune checkpoints in atherosclerosis with monoclonal antibodies. Atherosclerosis 2021; 335:98-109. [PMID: 34593238 DOI: 10.1016/j.atherosclerosis.2021.09.024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/12/2021] [Revised: 08/24/2021] [Accepted: 09/21/2021] [Indexed: 11/15/2022]
Abstract
Over the past fifteen years, treatments using monoclonal antibodies specifically targeting cytokines have been developed to treat chronic inflammatory diseases, including rheumatoid arthritis or psoriasis, both associated with increased cardiovascular risk. The cardiovascular impact of these therapies allows us to validate the clinical relevance of the knowledge acquired from experimental studies about the role of cytokines in atherosclerosis. Several clinical studies have confirmed the protective effects of anti-TNFα and anti-IL-6R monoclonal antibodies against athero-thrombotic cardiovascular risk in patients with chronic inflammatory diseases. Yet, caution is needed since anti-TNFα treatment can aggravate chronic heart failure. More recently, the CANTOS study showed for the first time that an anti-inflammatory treatment using anti-IL-1β monoclonal antibody in coronary artery disease patients significantly reduced cardiovascular events. The effects of IL-23/IL-17 axis blockade on cardiovascular risk in patients with psoriasis or arthritis remain controversial. Several monoclonal antibodies targeting costimulatory molecules have also been developed, a direct way to confirm their involvement in atherothrombotic cardiovascular diseases. Blocking the CD28-CD80/86 axis with Abatacept has been shown to reduce cardiovascular risk. In contrast, the treatment of cancer patients with antibodies blocking immune checkpoint inhibitory receptors, such as CTLA-4, PD1, or PDL1, could worsen the risk of atherothrombotic events. In the future, cardiologists will be increasingly solicited to assess the cardiovascular risk of patients suffering from chronic inflammatory diseases or cancer and participate in choosing the most appropriate treatment. At the same time, immunomodulatory approaches directly targeting cardiovascular diseases will be developed as a complement to the usual treatment strategies.
Collapse
Affiliation(s)
- Esther Lutgens
- Department of Medical Biochemistry Experimental Vascular Biology, Amsterdam, the Netherlands; Institute for Cardiovascular Prevention (IPEK), Ludwig-Maximilians-Universität, Pettenkoferstraße 8a & 9, 80336, Munich, Germany; German Centre for Cardiovascular Research (DZHK), Partner Site Munich Heart Alliance, Pettenkoferstraße 8a & 9, 80336, Munich, Germany.
| | - Jeremie Joffre
- Service de Réanimation Médicale, Hôpital Saint-Antoine, AP-HP, Sorbonne Université, Paris, France; Université de Paris, Inserm U970, Paris Cardiovascular Research Center, Paris, France; Department of Anesthesia and Perioperative Care, UCSF School of Medicine, San Francisco, CA, USA
| | - Bram van Os
- Department of Medical Biochemistry Experimental Vascular Biology, Amsterdam, the Netherlands
| | - Hafid Ait-Oufella
- Service de Réanimation Médicale, Hôpital Saint-Antoine, AP-HP, Sorbonne Université, Paris, France; Université de Paris, Inserm U970, Paris Cardiovascular Research Center, Paris, France.
| |
Collapse
|
48
|
Yoshimura A, Ito M, Mise-Omata S, Ando M. SOCS: negative regulators of cytokine signaling for immune tolerance. Int Immunol 2021; 33:711-716. [PMID: 34415326 DOI: 10.1093/intimm/dxab055] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2021] [Accepted: 08/18/2021] [Indexed: 11/14/2022] Open
Abstract
Cytokines are important intercellular communication tools for immunity. Many cytokines promote gene transcription and proliferation through the JAK/STAT (Janus kinase / signal transducers and activators of transcription) and the Ras/ERK (GDP/GTP-binding rat sarcoma protein / extracellular signal-regulated kinase) pathways, and these signaling pathways are tightly regulated. The SOCS (suppressor of cytokine signaling) family are representative negative regulators of JAK/STAT-mediated cytokine signaling and regulate the differentiation and function of T cells, thus being involved in immune tolerance. Human genetic analysis has shown that SOCS family members are strongly associated with autoimmune diseases, allergy and tumorigenesis. SOCS family proteins also function as immune-checkpoint molecules that contribute to the unresponsiveness of T cells to cytokines.
Collapse
Affiliation(s)
- Akihiko Yoshimura
- Department of Microbiology and Immunology, Keio University School of Medicine, Shinanomachi, Shinjyuku-ku, Tokyo, Japan
| | - Minako Ito
- Medical Institute of Bioregulation Kyushu University, Maidashi, Higashi-ku, Fukuoka, Japan
| | - Setsuko Mise-Omata
- Department of Microbiology and Immunology, Keio University School of Medicine, Shinanomachi, Shinjyuku-ku, Tokyo, Japan
| | - Makoto Ando
- Department of Microbiology and Immunology, Keio University School of Medicine, Shinanomachi, Shinjyuku-ku, Tokyo, Japan
| |
Collapse
|
49
|
Gatsiou A, Sopova K, Tselepis A, Stellos K. Interleukin-17A Triggers the Release of Platelet-Derived Factors Driving Vascular Endothelial Cells toward a Pro-Angiogenic State. Cells 2021; 10:1855. [PMID: 34440624 PMCID: PMC8392697 DOI: 10.3390/cells10081855] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2021] [Revised: 07/16/2021] [Accepted: 07/20/2021] [Indexed: 01/26/2023] Open
Abstract
Platelets comprise a highly interactive immune cell subset of the circulatory system traditionally known for their unique haemostatic properties. Although platelets are considered as a vault of growth factors, cytokines and chemokines with pivotal role in vascular regeneration and angiogenesis, the exact mechanisms by which they influence vascular endothelial cells (ECs) function remain underappreciated. In the present study, we examined the role of human IL-17A/IL-17RA axis in platelet-mediated pro-angiogenic responses. We reveal that IL-17A receptor (IL-17RA) mRNA is present in platelets transcriptome and a profound increase is documented on the surface of activated platelets. By quantifying the protein levels of several factors, involved in angiogenesis, we identified that IL-17A/IL17RA axis selectively induces the release of vascular endothelial growth factor, interleukin -2 and -4, as well as monocyte chemoattractant protein -1 from treated platelets. However, IL-17A exerted no effect on the release of IL-10, an anti-inflammatory factor with potentially anti-angiogenic properties, from platelets. Treatment of human endothelial cell two-dimensional tubule networks or three-dimensional spheroid and mouse aortic ring structures with IL-17A-induced platelet releasate evoked pro-angiogenic responses of ECs. Our findings suggest that IL-17A may critically affect platelet release of pro-angiogenic factors driving ECs towards a pro-angiogenic state.
Collapse
Affiliation(s)
- Aikaterini Gatsiou
- RNA Metabolism and Vascular Inflammation Group, Center of Molecular Medicine, Institute of Cardiovascular Regeneration, Johann Wolfgang Goethe University, 60596 Frankfurt am Main, Germany; (A.G.); (K.S.)
- Laboratory of Clinical Biochemistry, Atherothrombosis Research Center, University of Ioannina, 45110 Ioannina, Greece;
- Biosciences Institute, Vascular Biology and Medicine Theme, Faculty of Medical Sciences, Newcastle University, Newcastle Upon Tyne NE1 3BZ, UK
| | - Kateryna Sopova
- RNA Metabolism and Vascular Inflammation Group, Center of Molecular Medicine, Institute of Cardiovascular Regeneration, Johann Wolfgang Goethe University, 60596 Frankfurt am Main, Germany; (A.G.); (K.S.)
- Biosciences Institute, Vascular Biology and Medicine Theme, Faculty of Medical Sciences, Newcastle University, Newcastle Upon Tyne NE1 3BZ, UK
- Department of Cardiology, Freeman Hospital, Newcastle Hospitals NHS Foundation Trust, Newcastle upon Tyne NE7 7DN, UK
| | - Alexandros Tselepis
- Laboratory of Clinical Biochemistry, Atherothrombosis Research Center, University of Ioannina, 45110 Ioannina, Greece;
| | - Konstantinos Stellos
- RNA Metabolism and Vascular Inflammation Group, Center of Molecular Medicine, Institute of Cardiovascular Regeneration, Johann Wolfgang Goethe University, 60596 Frankfurt am Main, Germany; (A.G.); (K.S.)
- Biosciences Institute, Vascular Biology and Medicine Theme, Faculty of Medical Sciences, Newcastle University, Newcastle Upon Tyne NE1 3BZ, UK
- Department of Cardiology, Freeman Hospital, Newcastle Hospitals NHS Foundation Trust, Newcastle upon Tyne NE7 7DN, UK
| |
Collapse
|
50
|
Abstract
Atherosclerosis is the leading cause of acute cardiovascular events, and vascular calcification is an important pathological phenomenon in atherosclerosis. Recently, many studies have shown that immune cells are closely associated with the development of atherosclerosis and calcification, but there are many conflicting viewpoints because of immune system complications, such as the pro-atherosclerotic and atheroprotective effects of regulatory B cells (Bregs), T helper type 2 (Th2) cells and T helper type 17 (Th17) cells. In this review, we summarize the studies on the roles of immune cells, especially lymphocytes and macrophages, in atherosclerotic calcification. Furthermore, we prepared graphs showing the relationship between T cells, B cells and macrophages and atherosclerotic calcification. Finally, we highlight some potential issues that are closely associated with the function of immune cells in atherosclerotic calcification. Based on current research results, this review summarizes the relationship between immune cells and atherosclerotic calcification, and it will be beneficial to understand the relationship of immune cells and atherosclerotic calcification.
Collapse
Affiliation(s)
- Jingsong Cao
- Clinical Medicine Research Center, 574417The First Affiliated Hospital of University of South China, Hengyang, China.,Department of Endocrinology and Metabolism, 574417The First Affiliated Hospital of University of South China, Hengyang, Hunan, China
| | - Xuyu Zu
- Clinical Medicine Research Center, 574417The First Affiliated Hospital of University of South China, Hengyang, China
| | - Jianghua Liu
- Clinical Medicine Research Center, 574417The First Affiliated Hospital of University of South China, Hengyang, China.,Department of Endocrinology and Metabolism, 574417The First Affiliated Hospital of University of South China, Hengyang, Hunan, China.,Department of Metabolism and Endocrinology, 574417The First Affiliated Hospital of University of South China, Hengyang, China
| |
Collapse
|