1
|
Yang K, Liu Q, Fan A, Lin H, Wang X, Cui T, Fan G, Li L. Th17 Cells in Cardiovascular Disease. Cell Biochem Funct 2025; 43:e70069. [PMID: 40181529 DOI: 10.1002/cbf.70069] [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: 08/26/2024] [Revised: 01/12/2025] [Accepted: 03/06/2025] [Indexed: 04/05/2025]
Abstract
Recent research has shown a strong link between Th17 cells and their cytokine IL-17, and various cardiovascular diseases such as atherosclerosis, myocardial infarction, myocarditis, and arrhythmia. Moreover, Th17 cell signalling is likely to be a key factor in cardiovascular disease. Here, we summarize recent advances in the source, function, regulation, and the effects of Th17 signaling in cardiovascular disease. Research on Th17 will suggest more specific strategies to manipulate these functions. Thus, effective treatment of cardiovascular disease and future clinical treatment will be possible.
Collapse
Affiliation(s)
- Ke Yang
- State Key Laboratory of Modern Chinese Medicine, Key Laboratory of Pharmacology of Traditional Chinese Medical Formulae, Ministry of Education, Tianjin University of Traditional Chinese Medicine, Tianjin, China
- National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, State Key Laboratory of Component-based Chinese Medicine, First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Qianqian Liu
- State Key Laboratory of Modern Chinese Medicine, Key Laboratory of Pharmacology of Traditional Chinese Medical Formulae, Ministry of Education, Tianjin University of Traditional Chinese Medicine, Tianjin, China
- National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, State Key Laboratory of Component-based Chinese Medicine, First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Aodi Fan
- State Key Laboratory of Modern Chinese Medicine, Key Laboratory of Pharmacology of Traditional Chinese Medical Formulae, Ministry of Education, Tianjin University of Traditional Chinese Medicine, Tianjin, China
- National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, State Key Laboratory of Component-based Chinese Medicine, First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Hanqing Lin
- State Key Laboratory of Modern Chinese Medicine, Key Laboratory of Pharmacology of Traditional Chinese Medical Formulae, Ministry of Education, Tianjin University of Traditional Chinese Medicine, Tianjin, China
- National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, State Key Laboratory of Component-based Chinese Medicine, First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Xizheng Wang
- State Key Laboratory of Modern Chinese Medicine, Key Laboratory of Pharmacology of Traditional Chinese Medical Formulae, Ministry of Education, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Tianyi Cui
- State Key Laboratory of Modern Chinese Medicine, Key Laboratory of Pharmacology of Traditional Chinese Medical Formulae, Ministry of Education, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Guanwei Fan
- State Key Laboratory of Modern Chinese Medicine, Key Laboratory of Pharmacology of Traditional Chinese Medical Formulae, Ministry of Education, Tianjin University of Traditional Chinese Medicine, Tianjin, China
- National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, State Key Laboratory of Component-based Chinese Medicine, First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Lan Li
- State Key Laboratory of Modern Chinese Medicine, Key Laboratory of Pharmacology of Traditional Chinese Medical Formulae, Ministry of Education, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| |
Collapse
|
2
|
Kuang S, Ma X, Sun L, Wang C, Li Y, Wang G, Sun J, Zhou F, Zhang C. Exploring the Association Between Immune Cell Phenotypes and Osteoporosis Mediated by Inflammatory Cytokines: Insights from GWAS and Single-Cell Transcriptomics. Immunotargets Ther 2025; 14:227-246. [PMID: 40125424 PMCID: PMC11927574 DOI: 10.2147/itt.s510102] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2024] [Accepted: 03/07/2025] [Indexed: 03/25/2025] Open
Abstract
BACKGROUND Patients with osteoporosis experience increased fracture risk and decreased quality of life, which pose significant health burdens and financial challenges. Despite established links between immune cell phenotypes and inflammatory cytokines and osteoporosis, the exact mechanism involved remains unclear, and further understanding is needed for effective prevention and treatment. METHODS Here, we performed a two-sample Mendelian randomization (MR) study to estimate the causal effects between 731 immune cell types, 91 and 41 inflammatory factors (which may have some overlap), and 5 types of osteoporosis. In subsequent mediation MR analysis, we assessed whether these inflammatory cytokines mediate the causal relationship between immune cell phenotypes and osteoporosis. Additionally, colo- calization analysis was performed using Bayesian colocalization. Single-cell transcriptomic analysis was performed using datasets from osteoporosis patients available in the Gene Expression Omnibus (GEO) database. Subsequently, single-cell sequencing analysis was performed, including dimensionality reduction, clustering, and pathway enrichment, to investigate the underlying mechanisms. Finally, to confirm the critical role of IgD⁺CD24⁺ B cells and IL-17C in osteoporosis, we established vivo dexamethasone-induced osteoporosis model. Micro-CT was used to assess the effectiveness of model establishment. Flow cytometry was performed to determine the proportion of IgD⁺CD24⁺ B cells within lymphocytes in the blood. ELISA and Western blotting were used to measure IL-17C levels in serum and bone tissue. Immunohistochemistry was conducted to evaluate the expression of IL-17C in bone tissue. RESULTS This study found that 32 immune cell phenotypes and 38 inflammatory cytokines were significantly associated with osteoporosis. Mediation analysis indicated that IgD+ CD24+ B cells exacerbated the risk of osteoporosis by influencing the levels of interleukin-17C (IL-17C). The mediated effect was 0.07837, accounting for 15.5% of the total effect. Single-cell transcriptome analysis supported that IgD+ CD24+ B cells play a key role in musculoskeletal-related pathways in osteoporosis patients. Additionally, we have demonstrated the significant involvement of IgD⁺CD24⁺ B cells and IL-17C in the osteoporosis disease model. CONCLUSION Inflammatory cytokines play a crucial role in the pathogenesis of immunity-related osteoporosis. In particular, IgD+ CD24+ B cell %lymphocyte increase the risk of osteoporosis by modulating the levels of interleukin-17C. Our results provide evidence to support the link between immunity and osteoporosis and offer new therapeutic strategies for targeting inflammatory pathways in immune-mediated osteoporosis.
Collapse
Affiliation(s)
- Shouxiang Kuang
- Department of Orthopaedics, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Shandong, People’s Republic of China
| | - Xiaoqing Ma
- Minimally Invasive Therapy Oncology Department, The Second Affiliated Hospital of Shandong First Medical University, Taian, Shandong, People’s Republic of China
| | - Lipeng Sun
- Department of Orthopaedics, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Shandong, People’s Republic of China
| | - Chang Wang
- Department of Orthopaedics, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Shandong, People’s Republic of China
| | - Yang Li
- Department of Orthopaedics, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Shandong, People’s Republic of China
| | - Guodong Wang
- Department of Orthopaedics, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Shandong, People’s Republic of China
| | - Jianmin Sun
- Department of Orthopaedics, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Shandong, People’s Republic of China
| | - Fengge Zhou
- Tumor Research and Therapy Center, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Shandong, People’s Republic of China
| | - Chenggui Zhang
- Department of Orthopaedics, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Shandong, People’s Republic of China
| |
Collapse
|
3
|
Yang J, Yang L, Wang Y, Huai L, Shi B, Zhang D, Xu W, Cui D. Interleukin-6 related signaling pathways as the intersection between chronic diseases and sepsis. Mol Med 2025; 31:34. [PMID: 39891057 PMCID: PMC11783753 DOI: 10.1186/s10020-025-01089-6] [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: 08/28/2024] [Accepted: 01/16/2025] [Indexed: 02/03/2025] Open
Abstract
Sepsis is associated with immune dysregulated and organ dysfunction due to severe infection. Clinicians aim to restore organ function, rather than prevent diseases that are prone to sepsis, resulting in high mortality and a heavy public health burden. Some chronic diseases can induce sepsis through inflammation cascade reaction and Cytokine Storm (CS). Interleukin (IL)-6, the core of CS, and its related signaling pathways have been considered as contributors to sepsis. Therefore, it is important to study the relationship between IL-6 and its related pathways in sepsis-related chronic diseases. This review generalized the mechanism of sepsis-related chronic diseases via IL-6 related pathways with the purpose to take rational management for these diseases. IL-6 related signaling pathways were sought in Kyoto Encyclopedia of Genes and Genomes (KEGG), and retrieved protein-protein interaction in the Search for Interaction Genes tool (STRING). In PubMed and Google Scholar, the studies were searched out, which correlating to IL-6 related pathways and associating with the pathological process of sepsis. Focused on the interactions of sepsis and IL-6 related pathways, some chronic diseases have been studied for association with sepsis, containing insulin resistance, Alcoholic liver disease (ALD), Alzheimer disease (AD), and atherosclerosis. This article summarized the inflammatory mechanisms of IL-6 cross-talked with other mediators of some chronic diseases in vitro, animal models, and human experiments, leading to the activation of pathways and accelerating the progression of sepsis. The clinicians should be highlight to this kind of diseases and more clinical trials are needed to provide more reliable theoretical basis for health policy formulation.
Collapse
Affiliation(s)
- Jie Yang
- Department of Emergency, the People's Hospital of Liaoning Province, 33 Wenyi Road, Shenhe District, Shenyang, 110016, China.
| | - Lin Yang
- Department of Emergency, the People's Hospital of Liaoning Province, 33 Wenyi Road, Shenhe District, Shenyang, 110016, China
| | - Yanjiao Wang
- Department of Emergency, the People's Hospital of Liaoning Province, 33 Wenyi Road, Shenhe District, Shenyang, 110016, China
| | - Lu Huai
- Department of Emergency, the People's Hospital of Liaoning Province, 33 Wenyi Road, Shenhe District, Shenyang, 110016, China
| | - Bohan Shi
- Department of Emergency, the People's Hospital of Liaoning Province, 33 Wenyi Road, Shenhe District, Shenyang, 110016, China
| | - Di Zhang
- Department of Emergency, the People's Hospital of Liaoning Province, 33 Wenyi Road, Shenhe District, Shenyang, 110016, China
| | - Wei Xu
- Department of Emergency, the People's Hospital of Liaoning Province, 33 Wenyi Road, Shenhe District, Shenyang, 110016, China
| | - Di Cui
- Department of Emergency, the People's Hospital of Liaoning Province, 33 Wenyi Road, Shenhe District, Shenyang, 110016, China
| |
Collapse
|
4
|
Yu J, Liang P. A Mendelian randomization study on associations between plasma lipidome, circulating inflammatory proteins, and erectile dysfunction. Transl Androl Urol 2024; 13:2724-2734. [PMID: 39816232 PMCID: PMC11732301 DOI: 10.21037/tau-24-378] [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: 07/30/2024] [Accepted: 12/03/2024] [Indexed: 01/18/2025] Open
Abstract
Background Some studies suggest a potential association between plasma lipidome and erectile dysfunction (ED), but the underlying mechanism and whether circulating inflammatory proteins act as mediators remain unclear. The purpose of this study was to investigate the potential causal relationships between plasma lipidome, inflammatory proteins, and ED. Methods Plasma lipidome, circulating inflammatory proteins, and ED cases were identified based on the summary data from several large-scale genome-wide association studies (GWAS). The causal relationships of plasma lipidome and circulating inflammatory proteins with ED were explored by a bidirectional two-sample, two-sample Mendelian randomization (MR) method. The inverse variance weighted (IVW) method was used as the primary analytical method. MR-Egger and the weighted median (IVW) methods were utilized as supplementary analytical techniques. Sensitivity analyses including MR-Pleiotropy RESidual Sum and Outlier method (PRESSO), Cochran's Q test, and MR-Egger intercept test were conducted to address heterogeneity and horizontal pleiotropy. Results Ceramide (d42:2) and triacylglycerol (56:3) were found to be associated with an increased risk of ED, while phosphatidylethanolamine (O-18:1_18:2) and phosphatidylinositol (18:1_18:1) were linked to a decreased risk of ED. Interleukin-1α (IL-1α), IL-7, IL-17C, and the tumor necrosis factor (TNF) receptor superfamily member 9 (TNFRSF9) positively affected ED. Conversely, leukemia inhibitory factor and urokinase-type plasminogen activator (uPA) showed a negative impact. Mediation analysis indicated that the uPA mediated between Triacylglycerol (56:3) and ED, accounting for a mediation proportion of -14.71%. Conclusions There was a causal relationship between plasma lipidome and circulating inflammatory proteins with ED. Circulating inflammatory proteins appeared to mediate between triacylglycerol (56:3) levels and ED.
Collapse
Affiliation(s)
- Jiacheng Yu
- Department of Urology, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Peihe Liang
- Department of Urology, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| |
Collapse
|
5
|
Gu X, Weng R, Deng Q, Rao J, Zhao J, Hou J, Liu S. Interleukin-17D accelerates atherosclerosis through promoting endothelial cells ferroptosis via CD93/miR-181a-5p/SLC7A11 signaling. Int Immunopharmacol 2024; 143:113558. [PMID: 39510035 DOI: 10.1016/j.intimp.2024.113558] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2024] [Revised: 10/30/2024] [Accepted: 10/30/2024] [Indexed: 11/15/2024]
Abstract
IL-17D has been found to induce inflammatory cytokines in endothelial cells, but its exact role in atherosclerosis (AS) is unclear. This study aims to explore IL-17D' function in AS development. The expression of IL-17D was examined in AS patients and mice, and its clinical significance was evaluated in patients with acute coronary syndrome (ACS). Apolipoprotein E and IL-17D deficient mice (ApoE-/-IL-17D-/-) were generated for this study. The inflammation response and ferroptosis status in vascular endothelial cells were assessed following IL-17D treatment. Flow cytometry was used to identify the functional receptor of IL-17D. Additionally, RNA-seq was utilized to analyze the miRNA expression profiles induced by IL-17D. Plasma levels of IL-17D were elevated in both AS patients and mice, and were correlated with an increased incidence of major adverse cardiovascular events (MACEs). ApoE-/-IL-17D-/- mice displayed reduced inflammation and fewer atherosclerotic lesions. Treatment with IL-17D resulted in elevated levels of IL-6, IL-8, and ROS, as well as impaired cell viability and GSH production in endothelial cells. Ferroptosis inhibitor (Fer-1) suppressed the proinflammatory effects by IL-17D. Furthermore, CD93 was identified as the functional receptor for IL-17D in endothelial cells. The inhibition of miR-181a-5p led to a significant increase in cell viability and GSH levels, alongside a reduction in ROS and IL-6/IL-8 levels, while the suppression of SLC7A11 abolished these effects. Our findings suggest that IL-17D promotes endothelial inflammation by causing ferroptosis via CD93/miR-181a-5p/SLC7A11 signaling pathway. These insights advance our understanding of the pathophysiology of AS and identify a potential target for therapeutic intervention.
Collapse
Affiliation(s)
- Xiaodong Gu
- Meizhou Clinical Institute, Shantou University Medical College, Meizhou 514000, PR China; Institute of Basic Medical Sciences, Meizhou People's Hospital (Huangtang Hospital), Meizhou 514000, China; Guangdong Engineering Technology Research Center of Molecular Diagnostics for Cardiovascular Diseases, Meizhou 514000, PR China
| | - Ruiqiang Weng
- Meizhou Clinical Institute, Shantou University Medical College, Meizhou 514000, PR China; Institute of Basic Medical Sciences, Meizhou People's Hospital (Huangtang Hospital), Meizhou 514000, China; Guangdong Engineering Technology Research Center of Molecular Diagnostics for Cardiovascular Diseases, Meizhou 514000, PR China
| | - Qiaoting Deng
- Meizhou Clinical Institute, Shantou University Medical College, Meizhou 514000, PR China; Institute of Basic Medical Sciences, Meizhou People's Hospital (Huangtang Hospital), Meizhou 514000, China; Guangdong Engineering Technology Research Center of Molecular Diagnostics for Cardiovascular Diseases, Meizhou 514000, PR China
| | - Jiawei Rao
- Meizhou Clinical Medical School, Guangdong Medical University, Meizhou 514000, PR China
| | - Junli Zhao
- Meizhou Clinical Institute, Shantou University Medical College, Meizhou 514000, PR China
| | - Jingyuan Hou
- Meizhou Clinical Institute, Shantou University Medical College, Meizhou 514000, PR China; Guangdong Engineering Technology Research Center of Molecular Diagnostics for Cardiovascular Diseases, Meizhou 514000, PR China; Cardiovascular Disease Research Institute, Meizhou People's Hospital (Huangtang Hospital), Meizhou 514000, China.
| | - Sudong Liu
- Meizhou Clinical Institute, Shantou University Medical College, Meizhou 514000, PR China; Institute of Basic Medical Sciences, Meizhou People's Hospital (Huangtang Hospital), Meizhou 514000, China; Guangdong Engineering Technology Research Center of Molecular Diagnostics for Cardiovascular Diseases, Meizhou 514000, PR China.
| |
Collapse
|
6
|
Wen Y, Chen Q, Wang H, Xie S, Chen H, Yao W, Zhang L, Sun W, Wen J, Yang X, Chung KF, Zhang Q, Tao A, Yan J. Contribution of IL-17C-mediated macrophage polarization to Type 17 inflammation in neutrophilic asthma. Cell Commun Signal 2024; 22:557. [PMID: 39568050 PMCID: PMC11580697 DOI: 10.1186/s12964-024-01937-8] [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: 08/26/2024] [Accepted: 11/09/2024] [Indexed: 11/22/2024] Open
Abstract
BACKGROUND IL-17C has been described in a variety of inflammatory diseases driven by neutrophils. However, the role of IL-17C in neutrophilic asthma has not been completely characterized. METHODS The level of IL-17C in asthmatic patients and mice was assessed. Il-17c-deficient mice or mice treated with exogenous rmIL-17C were performed for OVA/CFA-induced asthmatic mice model. Pulmonary inflammation was evaluated by histological analysis, flow cytometry and cytokine analysis. Il-17re-overexpressed Raw264.7 were used in vitro to investigate the role of IL-17C in macrophage polarization. RESULTS Here, we show IL-17C were increased in serum or plasma from asthmatic patients and OVA/CFA-induced asthma mice. In the OVA/CFA-induced model, exogenous rmIL-17C aggravated neutrophil- and Type 17-dominated inflammation and promoted M1 macrophage differentiation, whereas deficiency of Il-17c reversed the pro-inflammatory phenotypes and inhibited the expansion of M1 macrophages. In vitro, IL-17C in synergy with IFN-γ induced STAT1 activation in Il-17re overexpressed Raw264.7 to upregulate M1-related genes expression, and promoted pro-inflammatory M1 polymerization, whereas IL-17C in contrast to the effect of IL-4 inhibited STAT6 activation, to reduce Raw264.7 differentiation to M2 macrophage and functional M2-related genes expression. CONCLUSIONS IL-17C promotes allergic inflammation via M1 polarization of pulmonary macrophages in neutrophilic asthma. Modulation of the IL-17C/IL-17RE axis represents a novel therapeutic target in neutrophilic asthma.
Collapse
Affiliation(s)
- Yuhuan Wen
- The State Key Laboratory of Respiratory Disease, Guangdong Provincial Key Laboratory of Allergy & Clinical Immunology, The Second Affiliated Hospital, Sino-French Hoffmann Institute, Guangzhou Medical University, Guangzhou, 510260, China
| | - Qile Chen
- The State Key Laboratory of Respiratory Disease, Guangdong Provincial Key Laboratory of Allergy & Clinical Immunology, The Second Affiliated Hospital, Sino-French Hoffmann Institute, Guangzhou Medical University, Guangzhou, 510260, China
| | - Hao Wang
- Department of Hematology, Guangzhou First People's HospitalInstitute of Blood Transfusion and Hematology, Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Shiyun Xie
- The State Key Laboratory of Respiratory Disease, Guangdong Provincial Key Laboratory of Allergy & Clinical Immunology, The Second Affiliated Hospital, Sino-French Hoffmann Institute, Guangzhou Medical University, Guangzhou, 510260, China
| | - Honglv Chen
- The State Key Laboratory of Respiratory Disease, Guangdong Provincial Key Laboratory of Allergy & Clinical Immunology, The Second Affiliated Hospital, Sino-French Hoffmann Institute, Guangzhou Medical University, Guangzhou, 510260, China
| | - Wenruo Yao
- The State Key Laboratory of Respiratory Disease, Guangdong Provincial Key Laboratory of Allergy & Clinical Immunology, The Second Affiliated Hospital, Sino-French Hoffmann Institute, Guangzhou Medical University, Guangzhou, 510260, China
| | - Le Zhang
- The State Key Laboratory of Respiratory Disease, Guangdong Provincial Key Laboratory of Allergy & Clinical Immunology, The Second Affiliated Hospital, Sino-French Hoffmann Institute, Guangzhou Medical University, Guangzhou, 510260, China
| | - Weimin Sun
- Department of Laboratory Medicine, The Seventh Affiliated Hospital of Southern Medical University, Foshan, China
| | - Junjie Wen
- Pulmonary and Critical Care Medicine, National Clinical Research Center for Respiratory Disease, National Center for Respiratory Medicine, State Key Laboratory of Respiratory Diseases, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510120, China
| | - Xiaojing Yang
- Pulmonary and Critical Care Medicine, National Clinical Research Center for Respiratory Disease, National Center for Respiratory Medicine, State Key Laboratory of Respiratory Diseases, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510120, China
| | - Kian Fan Chung
- National Heart and Lung Institute, Imperial College & Biomedical Research Unit, Royal Brompton & Harefield NHS Trust, London, UK
| | - Qingling Zhang
- Pulmonary and Critical Care Medicine, National Clinical Research Center for Respiratory Disease, National Center for Respiratory Medicine, State Key Laboratory of Respiratory Diseases, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510120, China.
| | - Ailin Tao
- The State Key Laboratory of Respiratory Disease, Guangdong Provincial Key Laboratory of Allergy & Clinical Immunology, The Second Affiliated Hospital, Sino-French Hoffmann Institute, Guangzhou Medical University, Guangzhou, 510260, China.
| | - Jie Yan
- The State Key Laboratory of Respiratory Disease, Guangdong Provincial Key Laboratory of Allergy & Clinical Immunology, The Second Affiliated Hospital, Sino-French Hoffmann Institute, Guangzhou Medical University, Guangzhou, 510260, China.
| |
Collapse
|
7
|
Döring Y, van der Vorst EPC, Weber C. Targeting immune cell recruitment in atherosclerosis. Nat Rev Cardiol 2024; 21:824-840. [PMID: 38664575 DOI: 10.1038/s41569-024-01023-z] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 03/26/2024] [Indexed: 10/17/2024]
Abstract
Atherosclerosis is the primary underlying cause of myocardial infarction and stroke. Atherosclerotic cardiovascular disease is characterized by a chronic inflammatory reaction in medium-to-large-sized arteries, with its onset and perpetuation driven by leukocytes infiltrating the subendothelial space. Activation of endothelial cells triggered by hyperlipidaemia and lipoprotein retention in the arterial intima initiates the accumulation of pro-inflammatory leukocytes in the arterial wall, fostering the progression of atherosclerosis. This inflammatory response is coordinated by an array of soluble mediators, namely cytokines and chemokines, that amplify inflammation both locally and systemically and are complemented by tissue-specific molecules that regulate the homing, adhesion and transmigration of leukocytes. Despite abundant evidence from mouse models, only a few therapies targeting leukocytes in atherosclerosis have been assessed in humans. The major challenges for the clinical translation of these therapies include the lack of tissue specificity and insufficient selectivity of inhibition strategies. In this Review, we discuss the latest research on receptor-ligand pairs and interactors that regulate leukocyte influx into the inflamed artery wall, primarily focusing on studies that used pharmacological interventions. We also discuss mechanisms that promote the resolution of inflammation and highlight how major findings from these research areas hold promise as potential therapeutic strategies for atherosclerotic cardiovascular disease.
Collapse
Affiliation(s)
- Yvonne Döring
- Department of Angiology, Swiss Cardiovascular Center, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland.
- Institute for Cardiovascular Prevention (IPEK), Ludwig-Maximilians-University Munich (LMU), Munich, Germany.
- DZHK (German Centre for Cardiovascular Research), Partner Site Munich Heart Alliance, Munich, Germany.
- Department for BioMedical Research (DBMR), University of Bern, Bern, Switzerland.
| | - Emiel P C van der Vorst
- Institute for Cardiovascular Prevention (IPEK), Ludwig-Maximilians-University Munich (LMU), Munich, Germany.
- Institute for Molecular Cardiovascular Research (IMCAR), RWTH Aachen University, Aachen, Germany.
- Aachen-Maastricht Institute for CardioRenal Disease (AMICARE), RWTH Aachen University, Aachen, Germany.
- Interdisciplinary Center for Clinical Research (IZKF), RWTH Aachen University, Aachen, Germany.
| | - Christian Weber
- Institute for Cardiovascular Prevention (IPEK), Ludwig-Maximilians-University Munich (LMU), Munich, Germany.
- DZHK (German Centre for Cardiovascular Research), Partner Site Munich Heart Alliance, Munich, Germany.
- Department of Biochemistry, Cardiovascular Research Institute Maastricht (CARIM), Maastricht University, Maastricht, Netherlands.
- Munich Cluster for Systems Neurology (SyNergy), Munich, Germany.
| |
Collapse
|
8
|
Shi S, Zhu C, Hu Y, Jiang P, Zhao J, Xu Q. ENG is a Biomarker of Prognosis and Angiogenesis in Liver Cancer, and Promotes the Differentiation of Tumor Cells into Vascular ECs. FRONT BIOSCI-LANDMRK 2024; 29:315. [PMID: 39344331 DOI: 10.31083/j.fbl2909315] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2024] [Revised: 08/08/2024] [Accepted: 08/16/2024] [Indexed: 10/01/2024]
Abstract
BACKGROUND Liver cancer is a highly lethal malignancy with frequent recurrence, widespread metastasis, and low survival rates. The aim of this study was to explore the role of Endoglin (ENG) in liver cancer progression, as well as its impacts on angiogenesis, immune cell infiltration, and the therapeutic efficacy of sorafenib. METHODS A comprehensive evaluation was conducted using online databases Gene Expression Omnibus (GEO) and The Cancer Genome Atlas (TCGA), 76 pairs of clinical specimens of tumor and adjacent non-tumor liver tissue, and tissue samples from 32 hepatocellular carcinoma (HCC) patients treated with sorafenib. ENG expression levels were evaluated using quantitative Reverse Transcription Polymerase Chain Reaction (qRT-PCR), Western blot, and immunohistochemical analysis. Cox regression analysis, Spearman rank correlation analysis, and survival analysis were used to assess the results. Functional experiments included Transwell migration assays and tube formation assays with Human Umbilical Vein Endothelial Cells (HUVECs). RESULTS Tumor cells exhibited retro-differentiation into endothelial-like cells, with a significant increase in ENG expression in these tumor-derived endothelial cells (TDECs). High expression of ENG was associated with more aggressive cancer characteristics and worse patient prognosis. Pathway enrichment and functional analyses identified ENG as a key regulator of immune responses and angiogenesis in liver cancer. Further studies confirmed that ENG increases the expression of Collagen type Iα1 (COL1A1), thereby promoting angiogenesis in liver cancer. Additionally, HCC patients with elevated ENG levels responded well to sorafenib treatment. CONCLUSIONS This study found that ENG is an important biomarker of prognosis in liver cancer. Moreover, ENG is associated with endothelial cell differentiation in liver cancer and plays a crucial role in formation of the tumor vasculature. The assessment of ENG expression could be a promising strategy to identify liver cancer patients who might benefit from targeted immunotherapies.
Collapse
MESH Headings
- Humans
- Liver Neoplasms/genetics
- Liver Neoplasms/metabolism
- Liver Neoplasms/pathology
- Liver Neoplasms/blood supply
- Liver Neoplasms/drug therapy
- Neovascularization, Pathologic/genetics
- Neovascularization, Pathologic/metabolism
- Biomarkers, Tumor/genetics
- Biomarkers, Tumor/metabolism
- Prognosis
- Carcinoma, Hepatocellular/genetics
- Carcinoma, Hepatocellular/metabolism
- Carcinoma, Hepatocellular/pathology
- Carcinoma, Hepatocellular/blood supply
- Carcinoma, Hepatocellular/drug therapy
- Sorafenib/pharmacology
- Sorafenib/therapeutic use
- Cell Differentiation
- Endoglin/metabolism
- Endoglin/genetics
- Male
- Female
- Middle Aged
- Cell Line, Tumor
- Phenylurea Compounds/pharmacology
- Human Umbilical Vein Endothelial Cells/metabolism
- Endothelial Cells/metabolism
- Endothelial Cells/pathology
- Niacinamide/analogs & derivatives
- Niacinamide/pharmacology
- Antineoplastic Agents/pharmacology
- Antineoplastic Agents/therapeutic use
- Angiogenesis
Collapse
Affiliation(s)
- Shangheng Shi
- Organ Transplantation Center, The Affiliated Hospital of Qingdao University, 266003 Qingdao, Shandong, China
- The Institute of Transplantation Science, Qingdao University, 266003 Qingdao, Shandong, China
| | - Cunle Zhu
- Organ Transplantation Center, The Affiliated Hospital of Qingdao University, 266003 Qingdao, Shandong, China
- The Institute of Transplantation Science, Qingdao University, 266003 Qingdao, Shandong, China
| | - Yue Hu
- Hepatobiliary and Pancreatic Surgery Department, Affiliated First Hospital of Ningbo University, 315000 Ningbo, Zhejiang, China
| | - Peng Jiang
- Organ Transplantation Center, The Affiliated Hospital of Qingdao University, 266003 Qingdao, Shandong, China
- The Institute of Transplantation Science, Qingdao University, 266003 Qingdao, Shandong, China
| | - Jinxin Zhao
- Organ Transplantation Center, The Affiliated Hospital of Qingdao University, 266003 Qingdao, Shandong, China
- The Institute of Transplantation Science, Qingdao University, 266003 Qingdao, Shandong, China
| | - Qingguo Xu
- Organ Transplantation Center, The Affiliated Hospital of Qingdao University, 266003 Qingdao, Shandong, China
- The Institute of Transplantation Science, Qingdao University, 266003 Qingdao, Shandong, China
| |
Collapse
|
9
|
Qin X, Zhu L, Zhong Y, Wang Y, Luo X, Li J, Yan F, Wu G, Qiu J, Wang G, Qu K, Zhang K, Wu W. Universal cell membrane camouflaged nano-prodrugs with right-side-out orientation adapting for positive pathological vascular remodeling in atherosclerosis. Chem Sci 2024; 15:7524-7544. [PMID: 38784734 PMCID: PMC11110172 DOI: 10.1039/d4sc00761a] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2024] [Accepted: 03/27/2024] [Indexed: 05/25/2024] Open
Abstract
A right-side-out orientated self-assembly of cell membrane-camouflaged nanotherapeutics is crucial for ensuring their biological functionality inherited from the source cells. In this study, a universal and spontaneous right-side-out coupling-driven ROS-responsive nanotherapeutic approach, based on the intrinsic affinity between phosphatidylserine (PS) on the inner leaflet and PS-targeted peptide modified nanoparticles, has been developed to target foam cells in atherosclerotic plaques. Considering the increased osteopontin (OPN) secretion from foam cells in plaques, a bioengineered cell membrane (OEM) with an overexpression of integrin α9β1 is integrated with ROS-cleavable prodrugs, OEM-coated ETBNPs (OEM-ETBNPs), to enhance targeted drug delivery and on-demand drug release in the local lesion of atherosclerosis. Both in vitro and in vivo experimental results confirm that OEM-ETBNPs are able to inhibit cellular lipid uptake and simultaneously promote intracellular lipid efflux, regulating the positive cellular phenotypic conversion. This finding offers a versatile platform for the biomedical applications of universal cell membrane camouflaging biomimetic nanotechnology.
Collapse
Affiliation(s)
- Xian Qin
- Key Laboratory for Biorheological Science and Technology of Ministry of Education, State and Local Joint Engineering Laboratory for Vascular Implants, Bioengineering College of Chongqing University Chongqing 400030 China
- Chongqing University Three Gorges Hospital, Chongqing Municipality Clinical Research Center for Endocrine and Metabolic Diseases Chongqing 404000 China
- School of Medicine, Chongqing University Chongqing 404010 China
| | - Li Zhu
- Key Laboratory for Biorheological Science and Technology of Ministry of Education, State and Local Joint Engineering Laboratory for Vascular Implants, Bioengineering College of Chongqing University Chongqing 400030 China
| | - Yuan Zhong
- Key Laboratory for Biorheological Science and Technology of Ministry of Education, State and Local Joint Engineering Laboratory for Vascular Implants, Bioengineering College of Chongqing University Chongqing 400030 China
| | - Yi Wang
- College of Basic Medical Sciences, Chongqing Medical University Chongqing 400016 China
| | - Xiaoshan Luo
- Guizhou Information Engineering University Bijie 551700 China
| | - Jiawei Li
- Chongqing University Three Gorges Hospital, Chongqing Municipality Clinical Research Center for Endocrine and Metabolic Diseases Chongqing 404000 China
- School of Medicine, Chongqing University Chongqing 404010 China
| | - Fei Yan
- Chongqing University Three Gorges Hospital, Chongqing Municipality Clinical Research Center for Endocrine and Metabolic Diseases Chongqing 404000 China
| | - Guicheng Wu
- Chongqing University Three Gorges Hospital, Chongqing Municipality Clinical Research Center for Endocrine and Metabolic Diseases Chongqing 404000 China
| | - Juhui Qiu
- Key Laboratory for Biorheological Science and Technology of Ministry of Education, State and Local Joint Engineering Laboratory for Vascular Implants, Bioengineering College of Chongqing University Chongqing 400030 China
| | - Guixue Wang
- Key Laboratory for Biorheological Science and Technology of Ministry of Education, State and Local Joint Engineering Laboratory for Vascular Implants, Bioengineering College of Chongqing University Chongqing 400030 China
- JinFeng Laboratory Chongqing 401329 China
| | - Kai Qu
- Key Laboratory for Biorheological Science and Technology of Ministry of Education, State and Local Joint Engineering Laboratory for Vascular Implants, Bioengineering College of Chongqing University Chongqing 400030 China
- Chongqing University Three Gorges Hospital, Chongqing Municipality Clinical Research Center for Endocrine and Metabolic Diseases Chongqing 404000 China
- School of Medicine, Chongqing University Chongqing 404010 China
| | - Kun Zhang
- Key Laboratory for Biorheological Science and Technology of Ministry of Education, State and Local Joint Engineering Laboratory for Vascular Implants, Bioengineering College of Chongqing University Chongqing 400030 China
- Chongqing University Three Gorges Hospital, Chongqing Municipality Clinical Research Center for Endocrine and Metabolic Diseases Chongqing 404000 China
- School of Medicine, Chongqing University Chongqing 404010 China
| | - Wei Wu
- Key Laboratory for Biorheological Science and Technology of Ministry of Education, State and Local Joint Engineering Laboratory for Vascular Implants, Bioengineering College of Chongqing University Chongqing 400030 China
- JinFeng Laboratory Chongqing 401329 China
| |
Collapse
|
10
|
Wang C, Liu S, Yang Y, Kamronbek R, Ni S, Cheng Y, Zhou C, Yan H, Li L, Liu H, Wang Y, Qin Y, Yin C, Zhang M. Interleukin-4 and Interleukin-17 are associated with coronary artery disease. Clin Cardiol 2024; 47:e24188. [PMID: 38146141 PMCID: PMC10823557 DOI: 10.1002/clc.24188] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/13/2023] [Revised: 10/12/2023] [Accepted: 10/30/2023] [Indexed: 12/27/2023] Open
Abstract
INTRODUCTION The present study aimed to examine the correlation between serum cytokine levels and the incidence of coronary artery disease (CAD), a leading cause of mortality globally, which is known to have a strong association with inflammatory factors. The study further sought to determine the predictors of CAD to distinguish patients with coronary artery lesions from those suspected of having CAD. METHODS AND RESULTS In this study, 487 patients who underwent coronary angiography as a result of suspected CAD but without acute myocardial infarction (AMI) were recruited. The serum levels of the cytokines interleukin (IL)-1β, IL-2, IL-4, IL-5, IL-6, IL-8, IL-10, IL-12p70, IL-17, tumor necrosis factor-α, interferon (IFN)-α, and IFN-γ were measured using a multiplexed particle-based flow cytometric assay technique. The results of the study revealed that the levels of IL-4, IL-12p70, IL-17, IFN-α, and IFN-γ in the CAD group were significantly lower compared to those in the non-CAD group. Multivariate logistic regression analysis indicated that two serum cytokines (IL-4 and IL-17), one protective factor (high-density lipoprotein cholesterol [HDL-C]), and three risk factors (sex, smoking, and diabetes) were independently predictive of CAD. The receiver operating characteristic curve analysis showed that the combined use of these predictors in a multivariate model demonstrated good predictive performance for CAD, as evidenced by an area under the curve value of 0.826. CONCLUSION The results of the study indicated that serum IL-4 and IL-17 levels serve as independent predictors of CAD. The risk prediction model established in the research, which integrates these serum cytokines (IL-4 and IL-17) with relevant clinical risk factors (gender, smoking, and diabetes) and the protective factor HDL-C, holds the potential to differentiate patients with CAD from those suspected of having CAD but without AMI.
Collapse
Affiliation(s)
- Chenyang Wang
- Center for Coronary Heart Disease, Beijing Anzhen HospitalCapital Medical UniversityBeijingChina
| | - Sheng Liu
- Center for Coronary Heart Disease, Beijing Anzhen HospitalCapital Medical UniversityBeijingChina
| | - Yunxiao Yang
- Center for Coronary Heart Disease, Beijing Anzhen HospitalCapital Medical UniversityBeijingChina
| | - Raimov Kamronbek
- Center for Coronary Heart Disease, Beijing Anzhen HospitalCapital Medical UniversityBeijingChina
| | - Siyao Ni
- Center for Coronary Heart Disease, Beijing Anzhen HospitalCapital Medical UniversityBeijingChina
| | - Yunjiu Cheng
- Department of Cardiology, Key Laboratory on Assisted Circulation, Ministry of Health, The First Affiliated HospitalSun Yat‐Sen UniversityGuangzhouChina
| | - Can Zhou
- Center for Coronary Heart Disease, Beijing Anzhen HospitalCapital Medical UniversityBeijingChina
| | - Huiyuan Yan
- Department of CardiologyHangjinqi People's HospitalInner MongoliaChina
| | - Li Li
- Liver Research Center, Beijing Friendship HospitalCapital Medical UniversityBeijingChina
| | - Hao Liu
- Department of OrthopedicsThe First Affiliated Hospital of Chongqing Medical UniversityChongqingChina
| | - Yu Wang
- Key Laboratory of Upper Airway Dysfunction‐Related Cardiovascular Diseases, Beijing Anzhen Hospital, Beijing Institute of Heart, Lung and Blood Vessel DiseaseCapital Medical UniversityBeijingChina
| | - Yanwen Qin
- Key Laboratory of Upper Airway Dysfunction‐Related Cardiovascular Diseases, Beijing Anzhen Hospital, Beijing Institute of Heart, Lung and Blood Vessel DiseaseCapital Medical UniversityBeijingChina
| | - Chengqian Yin
- Center for Coronary Heart Disease, Beijing Anzhen HospitalCapital Medical UniversityBeijingChina
| | - Ming Zhang
- Center for Coronary Heart Disease, Beijing Anzhen HospitalCapital Medical UniversityBeijingChina
| |
Collapse
|
11
|
Rao L, Peng B, Li T. Nonnegative matrix factorization analysis and multiple machine learning methods identified IL17C and ACOXL as novel diagnostic biomarkers for atherosclerosis. BMC Bioinformatics 2023; 24:196. [PMID: 37173646 PMCID: PMC10176911 DOI: 10.1186/s12859-023-05244-w] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2023] [Accepted: 03/21/2023] [Indexed: 05/15/2023] Open
Abstract
BACKGROUND Atherosclerosis is the common pathological basis for many cardiovascular and cerebrovascular diseases. The purpose of this study is to identify the diagnostic biomarkers related to atherosclerosis through machine learning algorithm. METHODS Clinicopathological parameters and transcriptomics data were obtained from 4 datasets (GSE21545, GSE20129, GSE43292, GSE100927). A nonnegative matrix factorization algorithm was used to classify arteriosclerosis patients in GSE21545 dataset. Then, we identified prognosis-related differentially expressed genes (DEGs) between the subtypes. Multiple machine learning methods to detect pivotal markers. Discrimination, calibration and clinical usefulness of the predicting model were assessed using area under curve, calibration plot and decision curve analysis respectively. The expression level of the feature genes was validated in GSE20129, GSE43292, GSE100927. RESULTS 2 molecular subtypes of atherosclerosis was identified, and 223 prognosis-related DEGs between the 2 subtypes were identified. These genes are not only related to epithelial cell proliferation, mitochondrial dysfunction, but also to immune related pathways. Least absolute shrinkage and selection operator, random forest, support vector machine- recursive feature elimination show that IL17C and ACOXL were identified as diagnostic markers of atherosclerosis. The prediction model displayed good discrimination and good calibration. Decision curve analysis showed that this model was clinically useful. Moreover, IL17C and ACOXL were verified in other 3 GEO datasets, and also have good predictive performance. CONCLUSION IL17C and ACOXL were diagnostic genes of atherosclerosis and associated with higher incidence of ischemic events.
Collapse
Affiliation(s)
- Li Rao
- Department of Geriatrics, Renmin Hospital of Wuhan University, Wuhan, 430060, Hubei, China
| | - Bo Peng
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, 430060, Hubei, China
- Cardiovascular Research Institute of Wuhan University, Wuhan, 430060, Hubei, China
- Hubei Key Laboratory of Cardiology, Wuhan, 430060, Hubei, China
- Department of Neurology, Renmin Hospital of Wuhan University, Wuhan, 430060, Hubei, China
| | - Tao Li
- Department of Neurology, Renmin Hospital of Wuhan University, Wuhan, 430060, Hubei, China.
| |
Collapse
|
12
|
Ling S, You Z, Li Y, Zhang J, Zhao S, He Y, Chen X. The role of γδ T17 cells in cardiovascular disease. J Leukoc Biol 2022; 112:1649-1661. [PMID: 36073777 DOI: 10.1002/jlb.3mr0822-761rr] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2022] [Revised: 06/23/2022] [Accepted: 08/03/2022] [Indexed: 01/04/2023] Open
Abstract
Due to the ability of γδ T cells to bridge adaptive and innate immunity, γδ T cells can respond to a variety of molecular cues and acquire the ability to induce a variety of cytokines such as IL-17 family, IFN-γ, IL-4, and IL-10. IL-17+ γδ T cells (γδ T17 cells) populations have recently received considerable interest as they are the major early source of IL-17A in many immune response models. However, the exact mechanism of γδ T17 cells is still poorly understood, especially in the context of cardiovascular disease (CVD). CVD is the leading cause of death in the world, and it tends to be younger. Here, we offer a review of the cardiovascular inflammatory and immune functions of γδ T17 cells in order to understand their role in CVD, which may be the key to developing new clinical applications.
Collapse
Affiliation(s)
- Shaoxue Ling
- College of Pharmaceutical Engineering of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, 10 Poyanghu Road, Tianjin, 301617, China
| | - Zonghao You
- College of Pharmaceutical Engineering of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, 10 Poyanghu Road, Tianjin, 301617, China
| | - Yang Li
- School of Chinese Materia Medica, Tianjin University of Traditional Chinese Medicine, 10 Poyanghu Road, Tianjin, 301617, China
| | - Jian Zhang
- School of Chinese Materia Medica, Tianjin University of Traditional Chinese Medicine, 10 Poyanghu Road, Tianjin, 301617, China
| | - Shuwu Zhao
- School of Intergrative Medicine, Tianjin University of Traditional Chinese Medicine, 10 Poyanghu Road, Tianjin, 301617, China
| | - Yongzhi He
- College of Pharmaceutical Engineering of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, 10 Poyanghu Road, Tianjin, 301617, China
| | - Xi Chen
- College of Pharmaceutical Engineering of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, 10 Poyanghu Road, Tianjin, 301617, China
| |
Collapse
|
13
|
Wang Y, Zang J, Liu C, Yan Z, Shi D. Interleukin-17 Links Inflammatory Cross-Talks Between Comorbid Psoriasis and Atherosclerosis. Front Immunol 2022; 13:835671. [PMID: 35514987 PMCID: PMC9063001 DOI: 10.3389/fimmu.2022.835671] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2021] [Accepted: 03/23/2022] [Indexed: 11/13/2022] Open
Abstract
Psoriasis is a chronic, systemic, immune-mediated inflammatory disorder that is associated with a significantly increased risk of cardiovascular disease (CVD). Studies have shown that psoriasis often coexists with atherosclerosis, a chronic inflammatory disease of large and medium-sized arteries, which is a major cause of CVD. Although the molecular mechanisms underlying this comorbidity are not fully understood, clinical studies have shown that when interleukin (IL)-17A inhibitors effectively improve psoriatic lesions, atherosclerotic symptoms are also ameliorated in patients with both psoriasis and atherosclerosis. Also, IL-17A levels are highly expressed in the psoriatic lesions and atherosclerotic plaques. These clinical observations implicit that IL-17A could be a crucial link for psoriasis and atherosclerosis and IL-17A-induced inflammatory responses are the major contribution to the pathogenesis of comorbid psoriasis and atherosclerosis. In this review, the current literature related to epidemiology, genetic predisposition, and inflammatory mechanisms of comorbidity of psoriasis and atherosclerosis is summarized. We focus on the immunopathological effects of IL-17A in both diseases. The goal of this review is to provide the theoretical base for future preventing or treating psoriasis patients with atherosclerosis comorbidity. The current evidence support the notion that treatments targeting IL-17 seem to be hold some promise to reduce cardiovascular risk in patients with psoriasis.
Collapse
Affiliation(s)
- Yan Wang
- College of Clinical Medicine, Jining Medical University, Jining, China
| | - Jinxin Zang
- Department of Neurology, Jining No.1 People's Hospital, Jining, China
| | - Chen Liu
- Laboratory of Medical Mycology, Jining No.1 People's Hospital, Jining, China
| | - Zhongrui Yan
- Department of Neurology, Jining No.1 People's Hospital, Jining, China
| | - Dongmei Shi
- Laboratory of Medical Mycology, Jining No.1 People's Hospital, Jining, China.,Department of Dermatology, Jining No.1 People's Hospital, Jining, China
| |
Collapse
|
14
|
Verdú E, Homs J, Boadas-Vaello P. Physiological Changes and Pathological Pain Associated with Sedentary Lifestyle-Induced Body Systems Fat Accumulation and Their Modulation by Physical Exercise. INTERNATIONAL JOURNAL OF ENVIRONMENTAL RESEARCH AND PUBLIC HEALTH 2021; 18:13333. [PMID: 34948944 PMCID: PMC8705491 DOI: 10.3390/ijerph182413333] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/20/2021] [Revised: 12/02/2021] [Accepted: 12/10/2021] [Indexed: 12/11/2022]
Abstract
A sedentary lifestyle is associated with overweight/obesity, which involves excessive fat body accumulation, triggering structural and functional changes in tissues, organs, and body systems. Research shows that this fat accumulation is responsible for several comorbidities, including cardiovascular, gastrointestinal, and metabolic dysfunctions, as well as pathological pain behaviors. These health concerns are related to the crosstalk between adipose tissue and body systems, leading to pathophysiological changes to the latter. To deal with these health issues, it has been suggested that physical exercise may reverse part of these obesity-related pathologies by modulating the cross talk between the adipose tissue and body systems. In this context, this review was carried out to provide knowledge about (i) the structural and functional changes in tissues, organs, and body systems from accumulation of fat in obesity, emphasizing the crosstalk between fat and body tissues; (ii) the crosstalk between fat and body tissues triggering pain; and (iii) the effects of physical exercise on body tissues and organs in obese and non-obese subjects, and their impact on pathological pain. This information may help one to better understand this crosstalk and the factors involved, and it could be useful in designing more specific training interventions (according to the nature of the comorbidity).
Collapse
Affiliation(s)
- Enrique Verdú
- Research Group of Clinical Anatomy, Embryology and Neuroscience (NEOMA), Department of Medical Sciences, University of Girona, 17003 Girona, Spain;
| | - Judit Homs
- Research Group of Clinical Anatomy, Embryology and Neuroscience (NEOMA), Department of Medical Sciences, University of Girona, 17003 Girona, Spain;
- Department of Physical Therapy, EUSES-University of Girona, 17190 Salt, Spain
| | - Pere Boadas-Vaello
- Research Group of Clinical Anatomy, Embryology and Neuroscience (NEOMA), Department of Medical Sciences, University of Girona, 17003 Girona, Spain;
| |
Collapse
|
15
|
Mishchenko EL, Mishchenko AM, Ivanisenko VA. Mechanosensitive molecular interactions in atherogenic regions of the arteries: development of atherosclerosis. Vavilovskii Zhurnal Genet Selektsii 2021; 25:552-561. [PMID: 34595377 PMCID: PMC8453358 DOI: 10.18699/vj21.062] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2021] [Revised: 03/26/2021] [Accepted: 04/08/2021] [Indexed: 11/30/2022] Open
Abstract
A terrible disease of the cardiovascular system, atherosclerosis, develops in the areas of bends and
branches of arteries, where the direction and modulus of the blood flow velocity vector change, and consequently
so does the mechanical effect on endothelial cells in contact with the blood flow. The review focuses on topical
research studies on the development of atherosclerosis – mechanobiochemical events that transform the proatherogenic
mechanical stimulus of blood flow – low and low/oscillatory arterial wall shear stress in the chains of biochemical
reactions in endothelial cells, leading to the expression of specific proteins that cause the progression
of the pathological process. The stages of atherogenesis, systemic risk factors for atherogenesis and its important
hemodynamic factor, low and low/oscillatory wall shear stress exerted by blood flow on the endothelial cells lining
the arterial walls, have been described. The interactions of cell adhesion molecules responsible for the development
of atherosclerosis under low and low/oscillating shear stress conditions have been demonstrated. The activation
of the regulator of the expression of cell adhesion molecules, the transcription factor NF-κB, and the factors
regulating its activation under these conditions have been described. Mechanosensitive signaling pathways leading
to the expression of NF-κB in endothelial cells have been described. Studies of the mechanobiochemical signaling
pathways and interactions involved in the progression of atherosclerosis provide valuable information for the
development of approaches that delay or block the development of this disease.
Key words: atherogenesis; shear stress; transcription factor NF-κB; RelA expression; mechanosensitive receptors;
cell adhesion molecules; signaling pathways; mechanotransduction.
Collapse
Affiliation(s)
- E L Mishchenko
- Institute of Cytology and Genetics of the Siberian Branch of the Russian Academy of Sciences, Novosibirsk, Russia
| | | | - V A Ivanisenko
- Institute of Cytology and Genetics of the Siberian Branch of the Russian Academy of Sciences, Novosibirsk, Russia
| |
Collapse
|
16
|
Vidal S, Puig L, Carrascosa-Carrillo JM, González-Cantero Á, Ruiz-Carrascosa JC, Velasco-Pastor AM. From Messengers to Receptors in Psoriasis: The Role of IL-17RA in Disease and Treatment. Int J Mol Sci 2021; 22:6740. [PMID: 34201664 PMCID: PMC8268646 DOI: 10.3390/ijms22136740] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2021] [Revised: 06/14/2021] [Accepted: 06/15/2021] [Indexed: 12/16/2022] Open
Abstract
The paradigm of psoriasis as a Th17-driven disease has evolved in the last years towards a much deeper knowledge of the complex pathways, mechanisms, cells, and messengers involved, highlighting the crucial role played by the IL-17 family of cytokines. All IL-17 isoforms signal through IL-17R. Five subunits of IL-17R have been described to date, which couple to form a homo- or hetero-receptor complex. Characteristically, IL-17RA is a common subunit in all hetero-receptors. IL-17RA has unique structural-containing a SEFIR/TILL domain-and functional-requiring ACT-1 for signaling-properties, enabling Th17 cells to act as a bridge between innate and adaptive immune cells. In psoriasis, IL-17RA plays a key role in pathogenesis based on: (a) IL-17A, IL-17F, and other IL-17 isoforms are involved in disease development; and (b) IL-17RA is essential for signaling of all IL-17 cytokines but IL-17D, whose receptor has not been identified to date. This article reviews current evidence on the biology and role of the IL-17 family of cytokines and receptors, with focus on IL-17RA, in psoriasis and some related comorbidities, and puts them in context with current and upcoming treatments.
Collapse
Affiliation(s)
- Silvia Vidal
- Institute of Research, Hospital de la Santa Creu i Sant Pau, 08041 Barcelona, Spain;
| | - Lluís Puig
- Institute of Research, Hospital de la Santa Creu i Sant Pau, 08041 Barcelona, Spain;
| | | | - Álvaro González-Cantero
- Department of Dermatology, Hospital Universitario Ramón y Cajal, M-607, km. 9, 100, 28034 Madrid, Spain;
- Facultad de Medicina, Universidad Francisco de Vitoria, Ctra. Pozuelo-Majadahonda KM 1.800, 28223 Pozuelo de Alarcón, Spain
| | | | | |
Collapse
|
17
|
Kurilenko N, Fatkhullina AR, Mazitova A, Koltsova EK. Act Locally, Act Globally-Microbiota, Barriers, and Cytokines in Atherosclerosis. Cells 2021; 10:cells10020348. [PMID: 33562334 PMCID: PMC7915371 DOI: 10.3390/cells10020348] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2021] [Revised: 01/30/2021] [Accepted: 02/02/2021] [Indexed: 12/12/2022] Open
Abstract
Atherosclerosis is a lipid-driven chronic inflammatory disease that is characterized by the formation and progressive growth of atherosclerotic plaques in the wall of arteries. Atherosclerosis is a major predisposing factor for stroke and heart attack. Various immune-mediated mechanisms are implicated in the disease initiation and progression. Cytokines are key mediators of the crosstalk between innate and adaptive immune cells as well as non-hematopoietic cells in the aortic wall and are emerging players in the regulation of atherosclerosis. Progression of atherosclerosis is always associated with increased local and systemic levels of pro-inflammatory cytokines. The role of cytokines within atherosclerotic plaque has been extensively investigated; however, the cell-specific role of cytokine signaling, particularly the role of cytokines in the regulation of barrier tissues tightly associated with microbiota in the context of cardiovascular diseases has only recently come to light. Here, we summarize the knowledge about the function of cytokines at mucosal barriers and the interplay between cytokines, barriers, and microbiota and discuss their known and potential implications for atherosclerosis development.
Collapse
Affiliation(s)
- Natalia Kurilenko
- Department of Medicine and Department of Biomedical Sciences, Cedars-Sinai Medical Center, 8700 Beverly Blvd, Los Angeles, CA 90048, USA; (N.K.); (A.M.)
| | | | - Aleksandra Mazitova
- Department of Medicine and Department of Biomedical Sciences, Cedars-Sinai Medical Center, 8700 Beverly Blvd, Los Angeles, CA 90048, USA; (N.K.); (A.M.)
| | - Ekaterina K. Koltsova
- Department of Medicine and Department of Biomedical Sciences, Cedars-Sinai Medical Center, 8700 Beverly Blvd, Los Angeles, CA 90048, USA; (N.K.); (A.M.)
- Correspondence:
| |
Collapse
|
18
|
Vella G, Ritzmann F, Wolf L, Kamyschnikov A, Stodden H, Herr C, Slevogt H, Bals R, Beisswenger C. IL-17C contributes to NTHi-induced inflammation and lung damage in experimental COPD and is present in sputum during acute exacerbations. PLoS One 2021; 16:e0243484. [PMID: 33411748 PMCID: PMC7790230 DOI: 10.1371/journal.pone.0243484] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2020] [Accepted: 11/21/2020] [Indexed: 12/20/2022] Open
Abstract
Neutrophilic inflammation results in loss of lung function in chronic obstructive pulmonary disease (COPD). Gram-negative bacteria, such as nontypeable Haemophilus influenzae (NTHi), trigger acute exacerbations of COPD (AECOPD) and contribute to chronic lung inflammation. The pro-inflammatory cytokine interleukin-17C (IL-17C) is expressed by airway epithelial cells and regulates neutrophilic chemotaxis. Here, we explored the function of IL-17C in NTHi- and cigarette smoke (CS)-induced models of COPD. Neutrophilic inflammation and tissue destruction were decreased in lungs of IL-17C-deficient mice (Il-17c-/-) chronically exposed to NTHi. Numbers of pulmonary neutrophils were decreased in Il-17c-/- mice after acute exposure to the combination of NTHi and CS. However, Il-17c-/- mice were not protected from CS-induced lung inflammation. In a preliminary patient study, we show that IL-17C is present in sputum samples obtained during AECOPD and associates with disease severity. Concentrations of IL-17C were significantly increased during advanced COPD (GOLD III/IV) compared to moderate COPD (GOLD I/II). Concentrations of IL-17A and IL-17E did not associate with disease severity. Our data suggest that IL-17C promotes harmful pulmonary inflammation triggered by bacteria in COPD.
Collapse
Affiliation(s)
- Giovanna Vella
- Department of Internal Medicine V – Pulmonology, Allergology and Respiratory Critical Care Medicine, Saarland University, Homburg, Germany
| | - Felix Ritzmann
- Department of Internal Medicine V – Pulmonology, Allergology and Respiratory Critical Care Medicine, Saarland University, Homburg, Germany
| | - Lisa Wolf
- Department of Internal Medicine V – Pulmonology, Allergology and Respiratory Critical Care Medicine, Saarland University, Homburg, Germany
| | - Andreas Kamyschnikov
- Department of Internal Medicine V – Pulmonology, Allergology and Respiratory Critical Care Medicine, Saarland University, Homburg, Germany
| | - Hannah Stodden
- Department of Internal Medicine V – Pulmonology, Allergology and Respiratory Critical Care Medicine, Saarland University, Homburg, Germany
| | - Christian Herr
- Department of Internal Medicine V – Pulmonology, Allergology and Respiratory Critical Care Medicine, Saarland University, Homburg, Germany
| | - Hortense Slevogt
- Septomics Research Center, Jena University Hospital, Jena, Germany
| | - Robert Bals
- Department of Internal Medicine V – Pulmonology, Allergology and Respiratory Critical Care Medicine, Saarland University, Homburg, Germany
| | - Christoph Beisswenger
- Department of Internal Medicine V – Pulmonology, Allergology and Respiratory Critical Care Medicine, Saarland University, Homburg, Germany
| |
Collapse
|
19
|
Yuan H, Jiao L, Yu N, Duan H, Yu Y, Bai Y. Histone Deacetylase 3-Mediated Inhibition of microRNA-19a-3p Facilitates the Development of Rheumatoid Arthritis-Associated Interstitial Lung Disease. Front Physiol 2020; 11:549656. [PMID: 33343379 PMCID: PMC7746846 DOI: 10.3389/fphys.2020.549656] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2020] [Accepted: 10/02/2020] [Indexed: 11/22/2022] Open
Abstract
Histone deacetylase (HDAC) has been implicated in rheumatoid arthritis (RA) progression. We investigated the roles of histone deacetylase 3 (HDAC3) involved in RA-associated interstitial lung disease (ILD) fibrosis. Firstly, we measured the expression of HDAC3 and interleukin 17 receptor A (IL17RA) in lung tissue samples from normal controls, idiopathic pulmonary fibrosis (IPF) patients, and RA-ILD patients. Next, chromatin immunoprecipitation (ChIP) and dual luciferase reporter assay were employed to detect the interaction between HDAC3 and microRNA-19a-3p (miR-19a-3p) and between miR-19a-3p and IL17RA. Further, immunohistochemistry was used to localize HDAC3 and IL17RA expression in lung tissues. Additionally, functional assays were conducted followed by expression determination of HDAC3, miR-19a-3p, and IL17RA with reverse transcription quantitative PCR (RT-qPCR) and Western blot analysis. The effect of HDAC3 on RA-ILD in the constructed RA-ILD mouse model was also studied based on arthritis assessment. We found overexpressed HDAC3 and IL17RA as well as silenced miR-19a-3p in RA-ILD mouse model and RA-ILD patients. In the mouse model, HDAC3 downregulated miR-19a-3p in lung fibroblasts to promote the progression of RA-ILD fibrosis. In lung fibroblasts of RA-ILD mice, IL17RA was a target gene of miR-19a-3p. miR-19a-3p negatively regulated IL17RA, thereby increasing the expression of fibrosis markers, COL1A1, COL3A1, and FN, in lung fibroblasts of mice. Taken together, HDAC3 upregulated IL17RA expression by targeting miR-19a-3p to facilitate the RA-ILD fibrosis development, which sheds light on a new HDAC3/miR-19a-3p/IL17RA axis functioning in RA-ILD fibrosis.
Collapse
Affiliation(s)
- Hui Yuan
- Department of Rheumatic Nephropathy, Affiliated Hospital of Shaanxi University of Chinese Medicine, Xianyang, China
| | - Li Jiao
- Yanching Institute of Technology, Langfang, China
| | - Nan Yu
- Shaanxi University of Chinese Medicine, Xianyang, China
| | - Haifeng Duan
- Department of Imaging, Affiliated Hospital of Shaanxi University of Chinese Medicine, Xianyang, China
| | - Yong Yu
- Department of Imaging, Affiliated Hospital of Shaanxi University of Chinese Medicine, Xianyang, China
| | - Yanrong Bai
- Department of Rheumatic Nephropathy, Affiliated Hospital of Shaanxi University of Chinese Medicine, Xianyang, China
| |
Collapse
|
20
|
Brevi A, Cogrossi LL, Grazia G, Masciovecchio D, Impellizzieri D, Lacanfora L, Grioni M, Bellone M. Much More Than IL-17A: Cytokines of the IL-17 Family Between Microbiota and Cancer. Front Immunol 2020; 11:565470. [PMID: 33244315 PMCID: PMC7683804 DOI: 10.3389/fimmu.2020.565470] [Citation(s) in RCA: 72] [Impact Index Per Article: 14.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2020] [Accepted: 10/15/2020] [Indexed: 12/30/2022] Open
Abstract
The interleukin-(IL-)17 family of cytokines is composed of six members named IL-17A, IL-17B, IL-17C, IL-17D, IL-17E, and IL-17F. IL-17A is the prototype of this family, and it was the first to be discovered and targeted in the clinic. IL-17A is essential for modulating the interplay between commensal microbes and epithelial cells at our borders (i.e., skin and mucosae), and yet, for protecting us from microbial invaders, thus preserving mucosal and skin integrity. Interactions between the microbiota and cells producing IL-17A have also been implicated in the pathogenesis of immune mediated inflammatory diseases and cancer. While interactions between microbiota and IL-17B-to-F have only partially been investigated, they are by no means less relevant. The cellular source of IL-17B-to-F, their main targets, and their function in homeostasis and disease distinguish IL-17B-to-F from IL-17A. Here, we intentionally overlook IL-17A, and we focus instead on the role of the other cytokines of the IL-17 family in the interplay between microbiota and epithelial cells that may contribute to cancer pathogenesis and immune surveillance. We also underscore differences and similarities between IL-17A and IL-17B-to-F in the microbiota-immunity-cancer axis, and we highlight therapeutic strategies that directly or indirectly target IL-17 cytokines in diseases.
Collapse
Affiliation(s)
- Arianna Brevi
- Cellular Immunology Unit, Division of Immunology, Transplantation and Infectious Diseases, I.R.C.C.S. Ospedale San Raffaele, Milan, Italy
| | - Laura Lucia Cogrossi
- Cellular Immunology Unit, Division of Immunology, Transplantation and Infectious Diseases, I.R.C.C.S. Ospedale San Raffaele, Milan, Italy.,Department of Medicine and Surgery, Vita-Salute San Raffaele University, Milan, Italy
| | - Giulia Grazia
- Cellular Immunology Unit, Division of Immunology, Transplantation and Infectious Diseases, I.R.C.C.S. Ospedale San Raffaele, Milan, Italy
| | - Desirée Masciovecchio
- Cellular Immunology Unit, Division of Immunology, Transplantation and Infectious Diseases, I.R.C.C.S. Ospedale San Raffaele, Milan, Italy
| | - Daniela Impellizzieri
- Cellular Immunology Unit, Division of Immunology, Transplantation and Infectious Diseases, I.R.C.C.S. Ospedale San Raffaele, Milan, Italy
| | - Lucrezia Lacanfora
- Cellular Immunology Unit, Division of Immunology, Transplantation and Infectious Diseases, I.R.C.C.S. Ospedale San Raffaele, Milan, Italy
| | - Matteo Grioni
- Cellular Immunology Unit, Division of Immunology, Transplantation and Infectious Diseases, I.R.C.C.S. Ospedale San Raffaele, Milan, Italy
| | - Matteo Bellone
- Cellular Immunology Unit, Division of Immunology, Transplantation and Infectious Diseases, I.R.C.C.S. Ospedale San Raffaele, Milan, Italy
| |
Collapse
|
21
|
Yu J, Zhu R, Yu K, Wang Y, Ding Y, Zhong Y, Zeng Q. Galectin-9: A Suppressor of Atherosclerosis? Front Immunol 2020; 11:604265. [PMID: 33250901 PMCID: PMC7672040 DOI: 10.3389/fimmu.2020.604265] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2020] [Accepted: 10/14/2020] [Indexed: 01/29/2023] Open
Abstract
It is no longer controversial that atherosclerosis is a vascular wall chronic inflammatory disease mediated by cells of innate and adaptive immunity. Galectin-9 (Gal-9) seems to be a crucial regulator of T-cell immunity by inducing apoptosis in specific T-cell subpopulations associated with autoimmunity and inflammatory disease. Accumulating evidence showed that galectin-9 signaling via T-cell immunoglobulin mucin 3 (TIM-3) is concerned with different regulatory functions in autoimmunity, including direct depletion of pro-inflammatory T-cells, expanding the number of regulatory T cells, altering macrophages to an anti-inflammatory state and the induction of repressive myeloid-derived suppressor cells. In addition, anti-Tim-3-Ab administration increased atherosclerotic plaque formation by blocking Tim-3–galectin-9 interaction. Hence, we hypothesize that galectin-9 may be a novel therapy for atherosclerotic disease. Further researches are needed to investigate the precise effect of galectin-9 in the process of atherosclerosis.
Collapse
Affiliation(s)
- Jian Yu
- Department of Cardiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Ruirui Zhu
- Department of Cardiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Kuwu Yu
- Department of Cardiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yue Wang
- Department of Cardiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yan Ding
- Department of Cardiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yucheng Zhong
- Department of Cardiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Qiutang Zeng
- Department of Cardiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
22
|
Vella G, Lunding L, Ritzmann F, Honecker A, Herr C, Wegmann M, Bals R, Beisswenger C. The IL-17 receptor IL-17RE mediates polyIC-induced exacerbation of experimental allergic asthma. Respir Res 2020; 21:176. [PMID: 32641167 PMCID: PMC7346407 DOI: 10.1186/s12931-020-01434-9] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2020] [Accepted: 06/23/2020] [Indexed: 01/08/2023] Open
Abstract
BACKGROUND The interleukin 17 receptor E (IL-17RE) is specific for the epithelial cytokine interleukin-17C (IL-17C). Asthma exacerbations are frequently caused by viral infections. Polyinosinic:polycytidylic acid (pIC) mimics viral infections through binding to pattern recognition receptors (e.g. TLR-3). We and others have shown that pIC induces the expression of IL-17C in airway epithelial cells. Using different mouse models, we aimed to investigate the function of IL-17RE in the development of experimental allergic asthma and acute exacerbation thereof. METHODS Wild-type (WT) and IL-17RE deficient (Il-17re-/-) mice were sensitized and challenged with OVA to induce allergic airway inflammation. pIC or PBS were applied intranasally when allergic airway inflammation had been established. Pulmonary expression of inflammatory mediators, numbers of inflammatory cells, and airway hyperresponsiveness (AHR) were analyzed. RESULTS Ablation of IL-17RE did not affect the development of OVA-induced allergic airway inflammation and AHR. pIC induced inflammation independent of IL-17RE in the absence of allergic airway inflammation. Treatment of mice with pIC exacerbated pulmonary inflammation in sensitized and OVA-challenged mice in an IL-17RE-dependent manner. The pIC-induced expression of cytokines (e.g. keratinocyte-derived chemokine (KC), granulocyte-colony stimulating factor (G-CSF)) and recruitment of neutrophils were decreased in Il-17re-/- mice. pIC-exacerbated AHR was partially decreased in Il-17re-/- mice. CONCLUSIONS Our results indicate that IL-17RE mediates virus-triggered exacerbations but does not have a function in the development of allergic lung disease.
Collapse
Affiliation(s)
- Giovanna Vella
- Department of Internal Medicine V – Pulmonology, Allergology and Critical Care Medicine, Saarland University, D-66421 Homburg, Germany
| | - Lars Lunding
- Division of Asthma Exacerbation & Regulation, Priority Area Asthma and Allergy, Leibniz Lung Center Borstel, Airway Research Center North (ARCN), Member of the German Center for Lung Research (DZL), Borstel, Germany
| | - Felix Ritzmann
- Department of Internal Medicine V – Pulmonology, Allergology and Critical Care Medicine, Saarland University, D-66421 Homburg, Germany
| | - Anja Honecker
- Department of Internal Medicine V – Pulmonology, Allergology and Critical Care Medicine, Saarland University, D-66421 Homburg, Germany
| | - Christian Herr
- Department of Internal Medicine V – Pulmonology, Allergology and Critical Care Medicine, Saarland University, D-66421 Homburg, Germany
| | - Michael Wegmann
- Division of Asthma Exacerbation & Regulation, Priority Area Asthma and Allergy, Leibniz Lung Center Borstel, Airway Research Center North (ARCN), Member of the German Center for Lung Research (DZL), Borstel, Germany
| | - Robert Bals
- Department of Internal Medicine V – Pulmonology, Allergology and Critical Care Medicine, Saarland University, D-66421 Homburg, Germany
| | - Christoph Beisswenger
- Department of Internal Medicine V – Pulmonology, Allergology and Critical Care Medicine, Saarland University, D-66421 Homburg, Germany
| |
Collapse
|
23
|
Wang F, Yin J, Lin Y, Zhang F, Liu X, Zhang G, Kong Y, Lu Z, Wu R, Wang N, Xing T, Qian Y. IL-17C has a pathogenic role in kidney ischemia/reperfusion injury. Kidney Int 2020; 97:1219-1229. [DOI: 10.1016/j.kint.2020.01.015] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2019] [Revised: 12/18/2019] [Accepted: 01/03/2020] [Indexed: 01/14/2023]
|
24
|
Nies JF, Panzer U. IL-17C/IL-17RE: Emergence of a Unique Axis in T H17 Biology. Front Immunol 2020; 11:341. [PMID: 32174926 PMCID: PMC7054382 DOI: 10.3389/fimmu.2020.00341] [Citation(s) in RCA: 56] [Impact Index Per Article: 11.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2019] [Accepted: 02/12/2020] [Indexed: 12/11/2022] Open
Abstract
Therapeutic targeting of IL-17A and its receptor IL-17RA with antibodies has turned out to be a tremendous success in the treatment of several autoimmune conditions. As the IL-17 cytokine family consists of six members (IL-17A to F), it is intriguing to elucidate the biological function of these five other molecules to identify more potential targets. In the past decade, IL-17C has emerged as quite a unique member of this pro-inflammatory cytokine group. In contrast to the well-described IL-17A and IL-17F, IL-17C is upregulated at very early timepoints of several disease settings. Also, the cellular source of the homodimeric cytokine differs from the other members of the family: Epithelial rather than hematopoietic cells were identified as the producers of IL-17C, while its receptor IL-17RE is expressed on TH17 cells as well as the epithelial cells themselves. Numerous investigations led to the current understanding that IL-17C (a) maintains an autocrine loop in the epithelium reinforcing innate immune barriers and (b) stimulates highly inflammatory TH17 cells. Functionally, the IL-17C/RE axis has been described to be involved in the pathogenesis of several diseases ranging from infectious and autoimmune conditions to cancer development and progression. This body of evidence has paved the way for the first clinical trials attempting to neutralize IL-17C in patients. Here, we review the latest knowledge about identification, regulation, and function of the IL-17C/IL-17receptor E pathway in inflammation and immunity, with a focus on the mechanisms underlying tissue injury. We also discuss the rationale for the translation of these findings into new therapeutic approaches in patients with immune-mediated disease.
Collapse
Affiliation(s)
- Jasper F Nies
- Translational Immunology, III. Department of Medicine, University Medical Center Hamburg-Eppendorf Hamburg, Hamburg, Germany
| | - Ulf Panzer
- Translational Immunology, III. Department of Medicine, University Medical Center Hamburg-Eppendorf Hamburg, Hamburg, Germany.,Hamburg Center of Translational Immunology (HCTI), University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| |
Collapse
|
25
|
Tian D, Hong H, Shang W, Ho CC, Dong J, Tian XY. Deletion of Ppard in CD11c + cells attenuates atherosclerosis in ApoE knockout mice. FASEB J 2020; 34:3367-3378. [PMID: 31919912 DOI: 10.1096/fj.201902069r] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2019] [Revised: 12/13/2019] [Accepted: 12/27/2019] [Indexed: 12/14/2022]
Abstract
Ppardδ, one of the lipid-activated nuclear receptor expressed in many cell types to activate gene transcription, also regulates cellular functions other than lipid metabolism. The mechanism regulating the function of antigen-presenting cells during the development of atherosclerosis is not fully understood. Here we aimed to study the involvement of PPARδ in CD11c+ cells in atherosclerosis. We used the Cre-loxP approach to make conditional deletion of Ppard in CD11c+ cells in mice on Apoe-/- background, which were fed with high cholesterol diet to develop atherosclerosis. Ppard deficiency in CD11c+ cells attenuated atherosclerotic plaque formation and infiltration of myeloid-derived dendritic cells (DCs) and T lymphocytes. Reduced lesion was accompanied by reduced activation of dendritic cells, and also a reduction of activation and differentiation of T cells to Th1 cells. In addition, DC migration to lymph node was also attenuated with Ppard deletion. In bone marrow-derived DCs, Ppard deficiency reduced palmitic acid-induced upregulation of co-stimulatory molecules and pro-inflammatory cytokine IL12 and TNFα. Our results indicated PPARδ activation by fatty acid resulted in the activation of myeloid DCs and subsequent polarization of T lymphocytes, which contributed to atherosclerosis in Apoe-/- mice. These findings also reveal the potential regulatory role of PPARδ in antigen presentation to orchestrate the immune responses during atherosclerosis.
Collapse
Affiliation(s)
- Danyang Tian
- School of Biomedical Sciences, Institute of Vascular Medicine, the Chinese University of Hong Kong, Shatin, Hong Kong.,Department of Physiology, Hebei Medical University, Shijiazhuang, China
| | - Huiling Hong
- School of Biomedical Sciences, Institute of Vascular Medicine, the Chinese University of Hong Kong, Shatin, Hong Kong
| | - Wenbin Shang
- School of Biomedical Sciences, Institute of Vascular Medicine, the Chinese University of Hong Kong, Shatin, Hong Kong
| | - Chin Chung Ho
- School of Biomedical Sciences, Institute of Vascular Medicine, the Chinese University of Hong Kong, Shatin, Hong Kong
| | - Jinghui Dong
- Department of Physiology, Hebei Medical University, Shijiazhuang, China
| | - Xiao Yu Tian
- School of Biomedical Sciences, Institute of Vascular Medicine, the Chinese University of Hong Kong, Shatin, Hong Kong
| |
Collapse
|
26
|
Herrero-Fernandez B, Gomez-Bris R, Somovilla-Crespo B, Gonzalez-Granado JM. Immunobiology of Atherosclerosis: A Complex Net of Interactions. Int J Mol Sci 2019; 20:E5293. [PMID: 31653058 PMCID: PMC6862594 DOI: 10.3390/ijms20215293] [Citation(s) in RCA: 85] [Impact Index Per Article: 14.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2019] [Revised: 10/21/2019] [Accepted: 10/22/2019] [Indexed: 02/07/2023] Open
Abstract
Cardiovascular disease is the leading cause of mortality worldwide, and atherosclerosis the principal factor underlying cardiovascular events. Atherosclerosis is a chronic inflammatory disease characterized by endothelial dysfunction, intimal lipid deposition, smooth muscle cell proliferation, cell apoptosis and necrosis, and local and systemic inflammation, involving key contributions to from innate and adaptive immunity. The balance between proatherogenic inflammatory and atheroprotective anti-inflammatory responses is modulated by a complex network of interactions among vascular components and immune cells, including monocytes, macrophages, dendritic cells, and T, B, and foam cells; these interactions modulate the further progression and stability of the atherosclerotic lesion. In this review, we take a global perspective on existing knowledge about the pathogenesis of immune responses in the atherosclerotic microenvironment and the interplay between the major innate and adaptive immune factors in atherosclerosis. Studies such as this are the basis for the development of new therapies against atherosclerosis.
Collapse
Affiliation(s)
- Beatriz Herrero-Fernandez
- LamImSys Lab. Instituto de Investigación Hospital 12 de Octubre (imas12), 28041 Madrid, Spain.
- Departamento de Fisiología. Facultad de Medicina. Universidad Autónoma de Madrid (UAM), 28029 Madrid, Spain.
| | - Raquel Gomez-Bris
- LamImSys Lab. Instituto de Investigación Hospital 12 de Octubre (imas12), 28041 Madrid, Spain.
| | | | - Jose Maria Gonzalez-Granado
- LamImSys Lab. Instituto de Investigación Hospital 12 de Octubre (imas12), 28041 Madrid, Spain.
- Departamento de Fisiología. Facultad de Medicina. Universidad Autónoma de Madrid (UAM), 28029 Madrid, Spain.
- Centro Nacional de Investigaciones Cardiovasculares Carlos III (CNIC), 28029 Madrid, Spain.
- CIBER de Enfermedades Cardiovasculares, 28029 Madrid, Spain.
| |
Collapse
|
27
|
Zheng N, Zhang L, Wang B, Wang G, Liu J, Miao G, Zhao X, Liu C, Zhang L. Chlamydia pneumoniae infection promotes vascular smooth muscle cell migration via c-Fos/interleukin-17C signaling. Int J Med Microbiol 2019; 309:151340. [PMID: 31494039 DOI: 10.1016/j.ijmm.2019.151340] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2019] [Revised: 07/08/2019] [Accepted: 08/18/2019] [Indexed: 01/01/2023] Open
Abstract
Chlamydia pneumoniae (C. pneumoniae) infection is associated with the initiation and progression of atherosclerosis. The migration of vascular smooth muscle cell (VSMC) from the media to the intima is a key event in the development of atherosclerosis. Interleukin-17C (IL-17C) could enhance cell migration ability. The aim of our study is to investigate the role of IL-17C in C. pneumoniae infection-promoted VSMC migration, thereby possibly accelerating atherosclerosis. We firstly demonstrated that C. pneumoniae infection significantly increased IL-17C expression in VSMCs in the atherosclerotic lesion area from ApoE deficient mice. Our in vitro study further showed that IL-17C is required for C. pneumoniae infection-promoted VSMC migration, and its expression could be regulated by c-Fos through phosphorylating extracellular signal-regulated kinase (ERK). Unexpectedly, in the present study, we also found that IL-17C is critical for C. pneumoniae infection-induced c-Fos activation. c-Fos expression and activation induced by the exposure to recombinant IL-17C were markedly suppressed in the presence of the ERK inhibitor PD98059. These results suggest a possible positive feedback between c-Fos and IL-17C after C. pneumoniae infection. Taken together, our results indicate that C. pneumoniae infection promotes VSMC migration via c-Fos/IL-17C signaling.
Collapse
Affiliation(s)
- Ningbo Zheng
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Tianjin Medical University, Tianjin, 300070, China
| | - Lijun Zhang
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Tianjin Medical University, Tianjin, 300070, China
| | - Beibei Wang
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Tianjin Medical University, Tianjin, 300070, China
| | - Guangyan Wang
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Tianjin Medical University, Tianjin, 300070, China
| | - Jingya Liu
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Tianjin Medical University, Tianjin, 300070, China
| | - Guolin Miao
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Tianjin Medical University, Tianjin, 300070, China
| | - Xi Zhao
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Tianjin Medical University, Tianjin, 300070, China
| | - Changle Liu
- Department of Cardiology, The Second Hospital of Tianjin Medical University, Tianjin, 300211, China
| | - Lijun Zhang
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Tianjin Medical University, Tianjin, 300070, China.
| |
Collapse
|
28
|
Lu HS, Schmidt AM, Hegele RA, Mackman N, Rader DJ, Weber C, Daugherty A. Reporting Sex and Sex Differences in Preclinical Studies. Arterioscler Thromb Vasc Biol 2019; 38:e171-e184. [PMID: 30354222 DOI: 10.1161/atvbaha.118.311717] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Affiliation(s)
- Hong S Lu
- From the Department of Physiology, Saha Cardiovascular Research Center, University of Kentucky, Lexington (H.S.L., A.D.)
| | - Ann Marie Schmidt
- Diabetes Research Program, Division of Endocrinology, Diabetes, and Metabolism, Department of Medicine, New York University Langone Medical Center, New York, NY (A.M.S.)
| | - Robert A Hegele
- Department of Medicine and Robarts Research Institute, Schulich School of Medicine and Dentistry, Western University, London, Ontario, Canada (R.A.H.)
| | - Nigel Mackman
- Department of Medicine, University of North Carolina at Chapel Hill (N.M.)
| | - Daniel J Rader
- Department of Medicine (D.J.R.), Perelman School of Medicine, University of Pennsylvania, Philadelphia.,Department of Genetics (D.J.R.), Perelman School of Medicine, University of Pennsylvania, Philadelphia
| | - Christian Weber
- Department of Medicine, Institute for Cardiovascular Prevention, Ludwig-Maximilians-Universität, Munich, Germany (C.W.).,German Centre for Cardiovascular Research, Partner Site Munich Heart Alliance, Munich, Germany (C.W.)
| | - Alan Daugherty
- From the Department of Physiology, Saha Cardiovascular Research Center, University of Kentucky, Lexington (H.S.L., A.D.)
| |
Collapse
|
29
|
Bryche B, Dewaele A, Saint-Albin A, Le Poupon Schlegel C, Congar P, Meunier N. IL-17c is involved in olfactory mucosa responses to Poly(I:C) mimicking virus presence. Brain Behav Immun 2019; 79:274-283. [PMID: 30776474 DOI: 10.1016/j.bbi.2019.02.012] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/30/2018] [Revised: 02/06/2019] [Accepted: 02/14/2019] [Indexed: 01/21/2023] Open
Abstract
At the interface of the environment and the nervous system, the olfactory mucosa (OM) is a privileged pathway for environmental toxicants and pathogens towards the central nervous system. The OM is known to produce antimicrobial and immunological components but the mechanisms of action of the immune system on the OM remain poorly explored. IL-17c is a potent mediator of respiratory epithelial innate immune responses, whose receptors are highly expressed in the OM of mice. We first characterized the presence of the IL-17c and its receptors in the OM. While IL-17c was weakly expressed in the control condition, it was strongly expressed in vivo after intranasal administration of polyinosinic-polycytidylic (Poly I:C), a Toll Like Receptor 3 agonist, mimicking a viral infection. Using calcium imaging and electrophysiological recordings, we found that IL-17c can effectively activate OM cells through the release of ATP. In the longer term, intranasal chronic instillations of IL-17c increased the cellular dynamics of the epithelium and promoted immune cells infiltrations. Finally, IL-17c decreased cell death induced by Poly(I:C) in an OM primary culture. The OM is thus a tissue highly responsive to immune mediators, proving its central role as a barrier against airway pathogens.
Collapse
Affiliation(s)
| | | | | | | | - Patrice Congar
- NBO, INRA, Univ Paris-Saclay, 78350 Jouy-en-Josas, France
| | - Nicolas Meunier
- NBO, INRA, Univ Paris-Saclay, 78350 Jouy-en-Josas, France; Université de Versailles Saint-Quentin en Yvelines, 78000 Versailles, France.
| |
Collapse
|
30
|
Demir E, Harmankaya NÖ, Kıraç Utku İ, Açıksarı G, Uygun T, Özkan H, Demir B. The Relationship between Epicardial Adipose Tissue Thickness and Serum Interleukin-17a Level in Patients with Isolated Metabolic Syndrome. Biomolecules 2019; 9:biom9030097. [PMID: 30862094 PMCID: PMC6468684 DOI: 10.3390/biom9030097] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2019] [Revised: 02/26/2019] [Accepted: 03/05/2019] [Indexed: 12/25/2022] Open
Abstract
In this study, it was aimed to investigate the relationship between the epicardial adipose tissue thickness (EATT) and serum IL-17A level insulin resistance in metabolic syndrome patients. This study enrolled a total of 160 subjects, of whom 80 were consecutive patients who applied to our outpatient clinic and were diagnosed with metabolic syndrome, and the other 80 were consecutive patients who were part of the control group with similar age and demographics in whom the metabolic syndrome was excluded. The metabolic syndrome diagnosis was made according to International Diabetes Federation (IDF)-2005 criteria. EATT was measured with transthoracic echocardiography (TTE) in the subjects. IL-17A serum levels were determined using the ELISA method. Fasting blood glucose, HDL, triglyceride, and fasting insulin levels were significantly higher in the metabolic syndrome group compared to the control group. In addition, the metabolic syndrome group had significantly higher high-sensitivity C-reactive protein (hs-CRP) and Homeostatic Model Assessment Insulin Resistance (HOMA-IR) levels than the control group. Similarly, serum IL-17A levels were significantly elevated in the metabolic syndrome group compared to the control group statistically (p < 0.001). As well, EATT was higher in the metabolic syndrome than the control group. Conclusion: By virtue of their proinflammatory properties, EATT and IL-17 may play an important role in the pathogenesis of the metabolic syndrome.
Collapse
Affiliation(s)
- Esra Demir
- Department of İnternal Medicine, Kanuni Sultan Süleyman Education and Research Hospital, 34103 İstanbul, Turkey.
| | - Nazmiye Özlem Harmankaya
- Department of İnternal Medicine, Bakırköy Dr Sadi Konuk Education and Research Hospital, 34147 İstanbul, Turkey.
| | - İrem Kıraç Utku
- Department of İnternal Medicine, Kanuni Sultan Süleyman Education and Research Hospital, 34103 İstanbul, Turkey.
| | - Gönül Açıksarı
- Department of Cardiology, Medeniyet University Medical Faculty, 34722 İstanbul, Turkey.
| | - Turgut Uygun
- Department of Cardiology, Konya Education and Research Hospital, 42040 Konya, Turkey.
| | - Hanise Özkan
- Department of İnternal Medicine, Kanuni Sultan Süleyman Education and Research Hospital, 34103 İstanbul, Turkey.
| | - Bülent Demir
- Department of Cardiology, Bakırköy Dr Sadi Konuk Education and Research Hospital, 34147 İstanbul, Turkey.
| |
Collapse
|
31
|
Pandit AA, Gandham RK, Mukhopadhyay CS, Verma R, Sethi RS. Transcriptome analysis reveals the role of the PCP pathway in fipronil and endotoxin-induced lung damage. Respir Res 2019; 20:24. [PMID: 30709343 PMCID: PMC6359862 DOI: 10.1186/s12931-019-0986-1] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2018] [Accepted: 01/16/2019] [Indexed: 12/21/2022] Open
Affiliation(s)
- Arif Ahmad Pandit
- Department of Animal Biotechnology, School of Animal Biotechnology, Guru Angad Dev Veterinary and Animals Sciences University, Ludhiana, Punjab, 141004, India
| | - Ravi Kumar Gandham
- Division of Veterinary Biotechnology, ICAR-Indian Veterinary Research Institute [Deemed University], Izatnagar, Bareilly, UP, India. National Institute of Animal Biotechnology, Hyderabad, India
| | - C S Mukhopadhyay
- Department of Animal Biotechnology, School of Animal Biotechnology, Guru Angad Dev Veterinary and Animals Sciences University, Ludhiana, Punjab, 141004, India
| | - Ramneek Verma
- Department of Animal Biotechnology, School of Animal Biotechnology, Guru Angad Dev Veterinary and Animals Sciences University, Ludhiana, Punjab, 141004, India
| | - R S Sethi
- Department of Animal Biotechnology, School of Animal Biotechnology, Guru Angad Dev Veterinary and Animals Sciences University, Ludhiana, Punjab, 141004, India.
| |
Collapse
|
32
|
Almén MS, Björk J, Nyman U, Lindström V, Jonsson M, Abrahamson M, Vestergren AS, Lindhe Ö, Franklin G, Christensson A, Grubb A. Shrunken Pore Syndrome Is Associated With Increased Levels of Atherosclerosis-Promoting Proteins. Kidney Int Rep 2019; 4:67-79. [PMID: 30596170 PMCID: PMC6308389 DOI: 10.1016/j.ekir.2018.09.002] [Citation(s) in RCA: 53] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2018] [Revised: 08/30/2018] [Accepted: 09/04/2018] [Indexed: 11/30/2022] Open
Abstract
INTRODUCTION Shrunken pore syndrome (SPS), originally defined by cystatin C-based estimated glomerular filtration rate (eGFRcystatin C) being less than 60% of creatinine-based estimated glomerular filtration rate (eGFRcreatinine) in the absence of extrarenal influences on the plasma levels of cystatin C or creatinine, is associated with a high increase in mortality, even in the absence of reduced glomerular filtration rate (GFR). The objective of the present study was to determine whether the proteome of patients with SPS shows differences from that of patients with normal or reduced measured GFR (mGFR) without SPS. METHODS Four patient cohorts were included: 1 cohort with normal mGFR without SPS, 1 with normal mGFR with SPS, 1 with reduced mGFR without SPS, and 1 with reduced mGFR with SPS. The plasma levels of 177 selected proteins were analyzed. RESULTS Differences in the levels of 30 proteins were specific for SPS; 31 differences were specific for patients with both SPS and reduced mGFR; and 27 were specific for reduced mGFR. Eighteen of the differences specific for SPS concerned proteins described as promoting, or being associated with, atherosclerosis. Twelve of the differences specific for patients with both SPS and reduced mGFR and 10 of the differences specific for reduced mGFR also concerned proteins described as promoting, or being associated with, atherosclerosis. Almost all (82 of 88) of the concentration differences represented increased levels. For SPS, but not for reduced mGFR, a correlation between protein size and increase in level was observed, with smaller proteins being associated with higher levels. CONCLUSION The high mortality in shrunken pore syndrome might be caused by the accumulation of atherosclerosis-promoting proteins in this condition.
Collapse
Affiliation(s)
| | - Jonas Björk
- Department of Occupational and Environmental Medicine, Lund University, Lund, Sweden
| | - Ulf Nyman
- Department of Clinical Sciences, Lund University, Malmö, Sweden
| | - Veronica Lindström
- Department of Clinical Chemistry, Skåne University Hospital, Lund, Lund University, Sweden
| | - Magnus Jonsson
- Department of Clinical Chemistry, Skåne University Hospital, Malmö, Sweden
| | | | | | | | | | - Anders Christensson
- Department of Nephrology, Skåne University Hospital, Malmö, Lund University, Sweden
| | - Anders Grubb
- Department of Clinical Chemistry, Skåne University Hospital, Lund, Lund University, Sweden
| |
Collapse
|
33
|
Zheng Y, Li T. Interleukin-22, a potent target for treatment of non-autoimmune diseases. Hum Vaccin Immunother 2018; 14:2811-2819. [PMID: 30335564 DOI: 10.1080/21645515.2018.1509649] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Interleukin -22 (IL-22) is a member of interleukin-10 (IL-10) family cytokines that is produced by different types of lymphocytes included in both innate and adaptive immune systems. These lymphocytes include activated T cells, most notably Th17 and Th22 cells, as well as NK cells, γδ T cells, etc. IL-22 mediate its effects via the IL-22-IL-22R complex and subsequent Janus Kinase-signal transduces and activators transcription (JAK-STAT) signaling pathway. According to recent evidence, IL-22 played a critical role in the pathogenesis of many non-autoimmune diseases. In this review, we mainly discussed the recent findings and advancements of the role of IL-22 in several non-autoimmune diseases, such as acute lung injury, atherosclerosis and some bacterial infections, suggesting that IL-22 may have therapeutic potential for treating non-autoimmune diseases.
Collapse
Affiliation(s)
- Yue Zheng
- a Cardiology , The Third Central Clinical College of Tianjin Medical University , Tianjin , China.,b Cardiology , Tianjin Key Laboratory of Artificial Cell.,c Artificial Cell Engineering Technology Research Center of Public Health Ministry , Tianjin , China.,d Tianjin Institute of Hepatobiliary Disease , Tianjin , China
| | - Tong Li
- b Cardiology , Tianjin Key Laboratory of Artificial Cell.,c Artificial Cell Engineering Technology Research Center of Public Health Ministry , Tianjin , China.,d Tianjin Institute of Hepatobiliary Disease , Tianjin , China.,e The Third Central Hospital of Tianjin , Tianjin , China
| |
Collapse
|
34
|
The roles of IL-17C in T cell-dependent and -independent inflammatory diseases. Sci Rep 2018; 8:15750. [PMID: 30356086 PMCID: PMC6200819 DOI: 10.1038/s41598-018-34054-x] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2018] [Accepted: 10/11/2018] [Indexed: 02/06/2023] Open
Abstract
IL-17C, which is a member of the IL-17 family of cytokines, is preferentially produced by epithelial cells in the lung, skin and colon, suggesting that IL-17C may be involved in not only host defense but also inflammatory diseases in those tissues. In support of that, IL-17C was demonstrated to contribute to development of T cell-dependent imiquimod-induced psoriatic dermatitis and T cell-independent dextran sodium sulfate-induced acute colitis using mice deficient in IL-17C and/or IL-17RE, which is a component of the receptor for IL-17C. However, the roles of IL-17C in other inflammatory diseases remain poorly understood. Therefore, we investigated the contributions of IL-17C to development of certain disease models using Il17c−/− mice, which we newly generated. Those mice showed normal development of T cell-dependent inflammatory diseases such as FITC- and DNFB-induced contact dermatitis/contact hypersensitivity (CHS) and concanavalin A-induced hepatitis, and T cell-independent inflammatory diseases such as bleomycin-induced pulmonary fibrosis, papain-induced airway eosinophilia and LPS-induced airway neutrophilia. On the other hand, those mice were highly resistant to LPS-induced endotoxin shock, indicating that IL-17C is crucial for protection against that immunological reaction. Therefore, IL-17C neutralization may represent a novel therapeutic approach for sepsis, in addition to psoriasis and acute colitis.
Collapse
|
35
|
Brembilla NC, Senra L, Boehncke WH. The IL-17 Family of Cytokines in Psoriasis: IL-17A and Beyond. Front Immunol 2018; 9:1682. [PMID: 30127781 PMCID: PMC6088173 DOI: 10.3389/fimmu.2018.01682] [Citation(s) in RCA: 325] [Impact Index Per Article: 46.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2018] [Accepted: 07/09/2018] [Indexed: 12/14/2022] Open
Abstract
Psoriasis is a frequent chronic inflammatory skin disease, nowadays considered a major global health problem. Several new drugs, targeting the IL-23/IL-17A pathway, have been recently licensed or are in clinical development. These therapies represent a major improvement of the way in which psoriasis is managed, since they show an unprecedented efficacy on skin symptoms of psoriasis. This has been made possible, thanks to an increasingly more accurate pathogenic view of psoriasis. Today, the belief that Th17 cells mediate psoriasis is moving to the concept of psoriasis as an IL-17A-driven disease. New questions arise at the horizon, given that IL-17A is part of a newly described family of cytokines, which has five distinct homologous: IL-17B, IL-17C, IL-17D, IL-17E, also known as IL-25 and IL-17F. IL-17 family cytokines elicit similar effects in target cells, but simultaneously trigger different and sometimes opposite functions in a tissue-specific manner. This is complicated by the fact that IL-17 cytokines show a high capacity of synergisms with other inflammatory stimuli. In this review, we will summarize the current knowledge around the cytokines belonging to the IL-17 family in relation to skin inflammation in general and psoriasis in particular, and discuss possible clinical implications. A comprehensive understanding of the different roles played by the IL-17 cytokines is crucial to appreciate current and developing therapies and to allow an effective pathogenesis- and mechanisms-driven drug design.
Collapse
Affiliation(s)
| | - Luisa Senra
- Department of Pathology and Immunology, Faculty of Medicine, University of Geneva, Geneva, Switzerland
| | - Wolf-Henning Boehncke
- Department of Pathology and Immunology, Faculty of Medicine, University of Geneva, Geneva, Switzerland.,Division of Dermatology and Venereology, Geneva University Hospitals, Geneva, Switzerland
| |
Collapse
|
36
|
Wall VZ, Barnhart S, Kanter JE, Kramer F, Shimizu-Albergine M, Adhikari N, Wight TN, Hall JL, Bornfeldt KE. Smooth muscle glucose metabolism promotes monocyte recruitment and atherosclerosis in a mouse model of metabolic syndrome. JCI Insight 2018; 3:96544. [PMID: 29875324 DOI: 10.1172/jci.insight.96544] [Citation(s) in RCA: 31] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2017] [Accepted: 05/01/2018] [Indexed: 12/21/2022] Open
Abstract
Metabolic syndrome contributes to cardiovascular disease partly through systemic risk factors. However, local processes in the artery wall are becoming increasingly recognized to exacerbate atherosclerosis both in mice and humans. We show that arterial smooth muscle cell (SMC) glucose metabolism markedly synergizes with metabolic syndrome in accelerating atherosclerosis progression, using a low-density lipoprotein receptor-deficient mouse model. SMCs in proximity to atherosclerotic lesions express increased levels of the glucose transporter GLUT1. Cytokines, such as TNF-α produced by lesioned arteries, promote GLUT1 expression in SMCs, which in turn increases expression of the chemokine CCL2 through increased glycolysis and the polyol pathway. Furthermore, overexpression of GLUT1 in SMCs, but not in myeloid cells, accelerates development of larger, more advanced lesions in a mouse model of metabolic syndrome, which also exhibits elevated levels of circulating Ly6Chi monocytes expressing the CCL2 receptor CCR2. Accordingly, monocyte tracing experiments demonstrate that targeted SMC GLUT1 overexpression promotes Ly6Chi monocyte recruitment to lesions. Strikingly, SMC-targeted GLUT1 overexpression fails to accelerate atherosclerosis in mice that do not exhibit the metabolic syndrome phenotype or monocytosis. These results reveal a potentially novel mechanism whereby arterial smooth muscle glucose metabolism synergizes with metabolic syndrome to accelerate monocyte recruitment and atherosclerosis progression.
Collapse
Affiliation(s)
- Valerie Z Wall
- Department of Medicine, University of Washington Medicine Diabetes Institute, University of Washington School of Medicine, Seattle, Washington, USA
| | - Shelley Barnhart
- Department of Medicine, University of Washington Medicine Diabetes Institute, University of Washington School of Medicine, Seattle, Washington, USA
| | - Jenny E Kanter
- Department of Medicine, University of Washington Medicine Diabetes Institute, University of Washington School of Medicine, Seattle, Washington, USA
| | - Farah Kramer
- Department of Medicine, University of Washington Medicine Diabetes Institute, University of Washington School of Medicine, Seattle, Washington, USA
| | - Masami Shimizu-Albergine
- Department of Medicine, University of Washington Medicine Diabetes Institute, University of Washington School of Medicine, Seattle, Washington, USA
| | - Neeta Adhikari
- Lillehei Heart Institute, University of Minnesota, Minneapolis, Minnesota, USA
| | - Thomas N Wight
- Benaroya Research Institute, Matrix Biology Program, Seattle, Washington, USA
| | - Jennifer L Hall
- Lillehei Heart Institute, University of Minnesota, Minneapolis, Minnesota, USA.,American Heart Association Institute for Precision Cardiovascular Medicine, Dallas, Texas USA
| | - Karin E Bornfeldt
- Department of Medicine, University of Washington Medicine Diabetes Institute, University of Washington School of Medicine, Seattle, Washington, USA.,Department of Pathology, University of Washington Medicine Diabetes Institute, University of Washington School of Medicine, Seattle, Washington, USA
| |
Collapse
|
37
|
Nosalski R, Guzik TJ. Perivascular adipose tissue inflammation in vascular disease. Br J Pharmacol 2017; 174:3496-3513. [PMID: 28063251 PMCID: PMC5610164 DOI: 10.1111/bph.13705] [Citation(s) in RCA: 271] [Impact Index Per Article: 33.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2016] [Revised: 12/29/2016] [Accepted: 01/04/2017] [Indexed: 12/11/2022] Open
Abstract
Perivascular adipose tissue (PVAT) plays a critical role in the pathogenesis of cardiovascular disease. In vascular pathologies, perivascular adipose tissue increases in volume and becomes dysfunctional, with altered cellular composition and molecular characteristics. PVAT dysfunction is characterized by its inflammatory character, oxidative stress, diminished production of vaso-protective adipocyte-derived relaxing factors and increased production of paracrine factors such as resistin, leptin, cytokines (IL-6 and TNF-α) and chemokines [RANTES (CCL5) and MCP-1 (CCL2)]. These adipocyte-derived factors initiate and orchestrate inflammatory cell infiltration including primarily T cells, macrophages, dendritic cells, B cells and NK cells. Protective factors such as adiponectin can reduce NADPH oxidase superoxide production and increase NO bioavailability in the vessel wall, while inflammation (e.g. IFN-γ or IL-17) induces vascular oxidases and eNOS dysfunction in the endothelium, vascular smooth muscle cells and adventitial fibroblasts. All of these events link the dysfunctional perivascular fat to vascular dysfunction. These mechanisms are important in the context of a number of cardiovascular disorders including atherosclerosis, hypertension, diabetes and obesity. Inflammatory changes in PVAT's molecular and cellular responses are uniquely different from classical visceral or subcutaneous adipose tissue or from adventitia, emphasizing the unique structural and functional features of this adipose tissue compartment. Therefore, it is essential to develop techniques for monitoring the characteristics of PVAT and assessing its inflammation. This will lead to a better understanding of the early stages of vascular pathologies and the development of new therapeutic strategies focusing on perivascular adipose tissue. LINKED ARTICLES This article is part of a themed section on Molecular Mechanisms Regulating Perivascular Adipose Tissue - Potential Pharmacological Targets? To view the other articles in this section visit http://onlinelibrary.wiley.com/doi/10.1111/bph.v174.20/issuetoc.
Collapse
Affiliation(s)
- Ryszard Nosalski
- Institute of Cardiovascular and Medical SciencesUniversity of GlasgowScotlandUK
- Department of Internal and Agricultural MedicineJagiellonian University, Collegium MedicumKrakowPoland
| | - Tomasz J Guzik
- Institute of Cardiovascular and Medical SciencesUniversity of GlasgowScotlandUK
- Department of Internal and Agricultural MedicineJagiellonian University, Collegium MedicumKrakowPoland
| |
Collapse
|
38
|
The atopic dermatitis blood signature is characterized by increases in inflammatory and cardiovascular risk proteins. Sci Rep 2017; 7:8707. [PMID: 28821884 PMCID: PMC5562859 DOI: 10.1038/s41598-017-09207-z] [Citation(s) in RCA: 194] [Impact Index Per Article: 24.3] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2017] [Accepted: 07/24/2017] [Indexed: 02/08/2023] Open
Abstract
Beyond classic “allergic”/atopic comorbidities, atopic dermatitis (AD) emerges as systemic disease with increased cardiovascular risk. To better define serum inflammatory and cardiovascular risk proteins, we used an OLINK high-throughput proteomic assay to analyze moderate-to-severe AD (n = 59) compared to psoriasis (n = 22) and healthy controls (n = 18). Compared to controls, 10 proteins were increased in serum of both diseases, including Th1 (IFN-γ, CXCL9, TNF-β) and Th17 (CCL20) markers. 48 proteins each were uniquely upregulated in AD and psoriasis. Consistent with skin expression, AD serum showed up-regulation of Th2 (IL-13, CCL17, eotaxin-1/CCL11, CCL13, CCL4, IL-10), Th1 (CXCL10, CXCL11) and Th1/Th17/Th22 (IL-12/IL-23p40) responses. Surprisingly, some markers of atherosclerosis (fractalkine/CX3CL1, CCL8, M-CSF, HGF), T-cell development/activation (CD40L, IL-7, CCL25, IL-2RB, IL-15RA, CD6) and angiogenesis (VEGF-A) were significantly increased only in AD. Multiple inflammatory pathways showed stronger enrichment in AD than psoriasis. Several atherosclerosis mediators in serum (e.g. E-selectin, PI3/elafin, CCL7, IL-16) correlated with SCORAD, but not BMI. Also, AD inflammatory mediators (e.g. MMP12, IL-12/IL-23p40, CXCL9, CCL22, PI3/Elafin) correlated between blood and lesional as well as non-lesional skin. Overall, the AD blood signature was largely different compared to psoriasis, with dysregulation of inflammatory and cardiovascular risk markers, strongly supporting its systemic nature beyond atopic/allergic association.
Collapse
|
39
|
Robert M, Miossec P. Effects of Interleukin 17 on the cardiovascular system. Autoimmun Rev 2017; 16:984-991. [PMID: 28705781 DOI: 10.1016/j.autrev.2017.07.009] [Citation(s) in RCA: 103] [Impact Index Per Article: 12.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2017] [Accepted: 06/17/2017] [Indexed: 12/25/2022]
Abstract
Cardiovascular diseases remain the leading cause of death worldwide and account for most of the premature mortality observed in chronic inflammatory diseases. Common mechanisms underlie these two types of disorders, where the contribution of Interleukin (IL)-17A, the founding member of the IL-17 family, is highly suspected. While the local effects of IL-17A in inflammatory disorders have been well described, those on the cardiovascular system remain less studied. This review focuses on the effects of IL-17 on the cardiovascular system both on isolated cells and in vivo. IL-17A acts on vessel and cardiac cells, leading to inflammation, coagulation and thrombosis. In vivo and clinical studies have shown its involvement in the pathogenesis of cardiovascular diseases including atherosclerosis and myocardial infarction that occur prematurely in chronic inflammatory disorders. As new therapeutic approaches are targeting the IL-17 pathway, this review should help to better understand their positive and negative outcomes on the cardio-vascular system.
Collapse
Affiliation(s)
- Marie Robert
- Immunogenomics and Inflammation Research Unit, EA 4130, Department of Immunology and Rheumatology, University of Lyon, Lyon, France
| | - Pierre Miossec
- Immunogenomics and Inflammation Research Unit, EA 4130, Department of Immunology and Rheumatology, University of Lyon, Lyon, France.
| |
Collapse
|
40
|
Nordlohne J, von Vietinghoff S. Interleukin 17A in atherosclerosis - Regulation and pathophysiologic effector function. Cytokine 2017; 122:154089. [PMID: 28663097 DOI: 10.1016/j.cyto.2017.06.016] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2017] [Revised: 06/01/2017] [Accepted: 06/21/2017] [Indexed: 12/20/2022]
Abstract
This review summarizes the current data on the interleukin (IL)-17A pathway in experimental atherosclerosis and clinical data. IL-17A is a prominent cytokine for early T cell response produced by both innate and adaptive leukocytes. In atherosclerosis, increased total IL-17A levels and expression in CD4+ T helper and γδ T cells have been demonstrated. Cytokines including IL-6 and TGFβ that increase IL-17A expression are elevated. Many other factors such as lipids, glucose and sodium chloride concentrations as well as vitamins and arylhydrocarbon receptor agonists that promote IL-17A expression are closely associated with cardiovascular risk in the human population. In acute inflammation models, IL-17A mediates innate leukocyte recruitment of both neutrophils and monocytes. In atherosclerosis, IL-17A increased aortic macrophage and T cell infiltration in most models. Secondary recruitment effects via the endothelium and according to recent data also pericytes have been demonstrated. IL-17 receptor A is highly expressed on monocytes and direct effects have been reported as well. Beyond leukocyte accumulation, IL-17A may affect other factors of plaque formation such as endothelial function, and according to some reports, fibrous cap formation and vascular relaxation with an increase in blood pressure. Anti-IL-17A agents are now available for clinical use. Cardiovascular side effect profiles are benign at this point. IL-17A appears to be a differential regulator of atherosclerosis and its effects in mouse models suggest that its modulation may have contradictory effects on plaque size and possibly stability in different patient populations.
Collapse
Affiliation(s)
- Johannes Nordlohne
- Department of Internal Medicine, Division of Nephrology and Hypertension, Hannover Medical School, Hannover, Germany
| | - Sibylle von Vietinghoff
- Department of Internal Medicine, Division of Nephrology and Hypertension, Hannover Medical School, Hannover, Germany.
| |
Collapse
|
41
|
Wolf L, Sapich S, Honecker A, Jungnickel C, Seiler F, Bischoff M, Wonnenberg B, Herr C, Schneider-Daum N, Lehr CM, Bals R, Beisswenger C. IL-17A-mediated expression of epithelial IL-17C promotes inflammation during acute Pseudomonas aeruginosa pneumonia. Am J Physiol Lung Cell Mol Physiol 2016; 311:L1015-L1022. [PMID: 27694471 DOI: 10.1152/ajplung.00158.2016] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2016] [Accepted: 09/26/2016] [Indexed: 12/31/2022] Open
Abstract
Lung epithelial cells are suggested to promote pathogen-induced pulmonary inflammation by the release of chemokines, resulting in enhanced recruitment of circulating leukocytes. Recent studies have shown that the interleukin-17C (IL-17C) regulates innate immune functions of epithelial cells in an autocrine manner. The aim of this study was to investigate the contribution of IL-17C to pulmonary inflammation in a mouse model of acute Pseudomonas aeruginosa pneumonia. Infection with P. aeruginosa resulted in an increased expression of IL-17C in lung tissue of wild-type mice. Numbers of neutrophils and the expression of the neutrophil-recruiting chemokines keratinocyte-derived chemokine and macrophage inflammatory protein 2 were significantly decreased in lungs of IL-17C-deficient (IL-17C-/-) mice infected with P. aeruginosa at 24 h. Systemic concentrations of interleukin-6 (IL-6) were significantly decreased in infected IL-17C-/- mice at 24 h and the survival of IL-17C-/- mice was significantly increased at 48 h. The expression of IL-17C was reduced in infected mice deficient for interleukin-17A (IL-17A), whereas pulmonary concentrations of IL-17A were not affected by the deficiency for IL-17C. Stimulation of primary alveolar epithelial cells with IL-17A resulted in a significantly increased expression of IL-17C in vitro. Our data suggest that IL-17A-mediated expression of epithelial IL-17C amplifies the release of chemokines by epithelial cells and thereby contributes to the recruitment of neutrophils and systemic inflammation during acute P. aeruginosa pneumonia.
Collapse
Affiliation(s)
- Lisa Wolf
- Department of Internal Medicine V-Pulmonology, Allergology and Respiratory Critical Care Medicine, Saarland University, Homburg, Germany
| | - Sandra Sapich
- Department of Drug Delivery, Helmholtz Institute for Pharmaceutical Research Saarland, Saarbrücken, Germany; and
| | - Anja Honecker
- Department of Internal Medicine V-Pulmonology, Allergology and Respiratory Critical Care Medicine, Saarland University, Homburg, Germany
| | - Christopher Jungnickel
- Department of Internal Medicine V-Pulmonology, Allergology and Respiratory Critical Care Medicine, Saarland University, Homburg, Germany
| | - Frederik Seiler
- Department of Internal Medicine V-Pulmonology, Allergology and Respiratory Critical Care Medicine, Saarland University, Homburg, Germany
| | - Markus Bischoff
- Institute of Medical Microbiology and Hygiene, Saarland University, Homburg, Germany
| | - Bodo Wonnenberg
- Department of Internal Medicine V-Pulmonology, Allergology and Respiratory Critical Care Medicine, Saarland University, Homburg, Germany
| | - Christian Herr
- Department of Internal Medicine V-Pulmonology, Allergology and Respiratory Critical Care Medicine, Saarland University, Homburg, Germany
| | - Nicole Schneider-Daum
- Department of Drug Delivery, Helmholtz Institute for Pharmaceutical Research Saarland, Saarbrücken, Germany; and
| | - Claus-Michael Lehr
- Department of Drug Delivery, Helmholtz Institute for Pharmaceutical Research Saarland, Saarbrücken, Germany; and.,Biopharmaceutics and Pharmaceutical Technology, Department of Pharmacy, Saarland University, Saarbrücken, Germany
| | - Robert Bals
- Department of Internal Medicine V-Pulmonology, Allergology and Respiratory Critical Care Medicine, Saarland University, Homburg, Germany
| | - Christoph Beisswenger
- Department of Internal Medicine V-Pulmonology, Allergology and Respiratory Critical Care Medicine, Saarland University, Homburg, Germany;
| |
Collapse
|