1
|
Guo Y, Deng F, Jiang Y, Cao G, Zhang Y, Liu G, Alimujiang M, Ayati M, Chen Y, Chen L, Lv S, Dou X. IL-37 Alleviates Sepsis-Induced Lung Injury by Inhibiting Inflammatory Response Through the TGF-β/Smad3 Pathway. Immunol Invest 2025:1-15. [PMID: 40270428 DOI: 10.1080/08820139.2025.2495958] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/25/2025]
Abstract
Introduction: Sepsis is caused by an uncontrolled inflammatory response and immune dysfunction, with lung injury being the most common complication and one of the leading causes of death in clinically ill patients. Interleukin 37 (IL-37) is a multifunctional cytokine that plays a vital role in various pathophysiological processes, including inflammation, infection, and immunity.Methods: The study involved both clinical and animal experiments (establishing an animal model of sepsis-induced lung injury). Firstly, 50 patients with sepsis-induced lung injury and 50 healthy controls were included. In addition, a more in-depth study was conducted using animal models.Results: IL-37, IL-6, PCT, and CRP levels were significantly higher in the sepsis-induced lung injury group. Correlation analysis revealed that IL-37 significantly correlated with IL-6, PCT, and CRP levels. In animal experiments, IL-37 significantly attenuated CLP-induced pulmonary edema and cellular injury while reducing the levels of inflammatory factors IL-6 and TNF-α, as well as sepsis-related inflammatory markers PCT and CRP. Moreover, IL-37 significantly downregulated the expression levels of genes and proteins of apoptosis-related molecules Caspase-3 and Bax and pathway molecules TGF-β and Smad3. Discussion: The TGF-β/Smad3 pathway is involved in the process of IL-37 inhibiting inflammatory response and ameliorating sepsis-induced lung injury.
Collapse
Affiliation(s)
- Yufang Guo
- Department of Critical Care Medicine, The Friendship Hospital of Ili Kazakh Autonomous Prefecture, Yili, Xinjiang, P.R. China
- State Key Laboratory of Pathogenesis, Prevention and Treatment of High Incidence Diseases in Central Asia, Xinjiang Medical University, Urumqi, Xinjiang, P.R. China
| | - Feifei Deng
- Department of Critical Care Medicine, The Friendship Hospital of Ili Kazakh Autonomous Prefecture, Yili, Xinjiang, P.R. China
| | - Yali Jiang
- Department of Critical Care Medicine, The Friendship Hospital of Ili Kazakh Autonomous Prefecture, Yili, Xinjiang, P.R. China
- Ili & Jiangsu Joint Institute of Health, Yili, Xinjiang, P.R. China
| | - Guodong Cao
- Department of Critical Care Medicine, The Friendship Hospital of Ili Kazakh Autonomous Prefecture, Yili, Xinjiang, P.R. China
- State Key Laboratory of Pathogenesis, Prevention and Treatment of High Incidence Diseases in Central Asia, Xinjiang Medical University, Urumqi, Xinjiang, P.R. China
- Ili & Jiangsu Joint Institute of Health, Yili, Xinjiang, P.R. China
| | - Yixin Zhang
- Department of Critical Care Medicine, Medical School of Shihezi University, Shihezi, Xinjiang, P.R. China
| | - Gaowu Liu
- Department of Critical Care Medicine, The Friendship Hospital of Ili Kazakh Autonomous Prefecture, Yili, Xinjiang, P.R. China
| | - Mayinur Alimujiang
- Department of Critical Care Medicine, The Friendship Hospital of Ili Kazakh Autonomous Prefecture, Yili, Xinjiang, P.R. China
| | - Mairhaba Ayati
- Department of Critical Care Medicine, The Friendship Hospital of Ili Kazakh Autonomous Prefecture, Yili, Xinjiang, P.R. China
| | - Yufeng Chen
- Department of Critical Care Medicine, The Friendship Hospital of Ili Kazakh Autonomous Prefecture, Yili, Xinjiang, P.R. China
| | - Lili Chen
- Department of Critical Care Medicine, The Friendship Hospital of Ili Kazakh Autonomous Prefecture, Yili, Xinjiang, P.R. China
| | - Su Lv
- Department of Critical Care Medicine, The Friendship Hospital of Ili Kazakh Autonomous Prefecture, Yili, Xinjiang, P.R. China
| | - Xueqin Dou
- Department of Critical Care Medicine, The Friendship Hospital of Ili Kazakh Autonomous Prefecture, Yili, Xinjiang, P.R. China
| |
Collapse
|
2
|
Wu Q, Zeng Y, Geng K, Guo M, Teng FY, Yan PJ, Lei Y, Long Y, Jiang ZZ, Law BYK, Xu Y. The role of IL-1 family cytokines in diabetic cardiomyopathy. Metabolism 2025; 163:156083. [PMID: 39603339 DOI: 10.1016/j.metabol.2024.156083] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/30/2024] [Revised: 11/18/2024] [Accepted: 11/21/2024] [Indexed: 11/29/2024]
Abstract
Diabetic cardiomyopathy (DCM) is the primary cause of heart failure in patients with diabetes and is characterised by contractile dysfunction and left ventricular hypertrophy. The complex pathological and physiological mechanisms underlying DCM have contributed to a limited number of available treatment options. A substantial body of evidence has established that DCM is a low-grade inflammatory cardiovascular disorder, with the interleukin-1 (IL-1) family of cytokines playing crucial roles in initiating inflammatory responses and shaping innate and adaptive immunity. In this review, we aim to provide an overview of the underlying mechanisms of the IL-1 family and their relevance in DCM of various aetiologies. Furthermore, we highlighted potential therapeutic targets within the IL-1 family for the management of DCM.
Collapse
Affiliation(s)
- Qi Wu
- State Key Laboratory of Quality Research in Chinese Medicine, Dr. Neher's Biophysics Laboratory for Innovative Drug Discovery, Faculty of Chinese Medicine, Macau University of Science and Technology, Taipa, Macao 999078, China; Department of Pathology, and Luzhou Key Laboratory of Precision Pathology Diagnosis for Serious Diseases, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan 646000, China
| | - Yan Zeng
- State Key Laboratory of Quality Research in Chinese Medicine, Dr. Neher's Biophysics Laboratory for Innovative Drug Discovery, Faculty of Chinese Medicine, Macau University of Science and Technology, Taipa, Macao 999078, China; Department of Endocrinology and Metabolism, and Metabolic Vascular Disease Key Laboratory of Sichuan Province, the Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan 646000, China
| | - Kang Geng
- Department of Endocrinology and Metabolism, and Metabolic Vascular Disease Key Laboratory of Sichuan Province, the Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan 646000, China; Department of Plastic and burns surgery, National Key Clinical Construction Specialty, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan 646000, China
| | - Man Guo
- Department of Endocrinology and Metabolism, and Metabolic Vascular Disease Key Laboratory of Sichuan Province, the Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan 646000, China
| | - Fang-Yuan Teng
- Department of Endocrinology and Metabolism, and Metabolic Vascular Disease Key Laboratory of Sichuan Province, the Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan 646000, China
| | - Pi-Jun Yan
- Department of Endocrinology and Metabolism, and Metabolic Vascular Disease Key Laboratory of Sichuan Province, the Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan 646000, China
| | - Yi Lei
- State Key Laboratory of Quality Research in Chinese Medicine, Dr. Neher's Biophysics Laboratory for Innovative Drug Discovery, Faculty of Chinese Medicine, Macau University of Science and Technology, Taipa, Macao 999078, China; Department of Endocrinology and Metabolism, and Metabolic Vascular Disease Key Laboratory of Sichuan Province, the Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan 646000, China
| | - Yang Long
- Department of Endocrinology and Metabolism, and Metabolic Vascular Disease Key Laboratory of Sichuan Province, the Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan 646000, China
| | - Zong-Zhe Jiang
- Department of Endocrinology and Metabolism, and Metabolic Vascular Disease Key Laboratory of Sichuan Province, the Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan 646000, China.
| | - Betty Yuen-Kwan Law
- State Key Laboratory of Quality Research in Chinese Medicine, Dr. Neher's Biophysics Laboratory for Innovative Drug Discovery, Faculty of Chinese Medicine, Macau University of Science and Technology, Taipa, Macao 999078, China.
| | - Yong Xu
- State Key Laboratory of Quality Research in Chinese Medicine, Dr. Neher's Biophysics Laboratory for Innovative Drug Discovery, Faculty of Chinese Medicine, Macau University of Science and Technology, Taipa, Macao 999078, China; Department of Endocrinology and Metabolism, and Metabolic Vascular Disease Key Laboratory of Sichuan Province, the Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan 646000, China.
| |
Collapse
|
3
|
Wang C, Zhong J, Hu J, Cao C, Qi S, Ma R, Fu W, Zhang X, Akdis CA, Gao Y. IL-37 protects against house dust mite-induced airway inflammation and airway epithelial barrier dysfunction via inhibiting store-operated calcium entry. Int Immunopharmacol 2024; 138:112525. [PMID: 38941668 DOI: 10.1016/j.intimp.2024.112525] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2024] [Revised: 06/03/2024] [Accepted: 06/17/2024] [Indexed: 06/30/2024]
Abstract
BACKGROUND Airway epithelial barrier dysfunction has been proved to contribute to the development of type 2 inflammation of asthma. Interleukin (IL)-37 is a negative regulator of immune responses and allergic airway inflammation. However, whether IL-37 has any effect on airway epithelial barrier has been unknown. METHODS We evaluated the role of IL-37 in both mouse model and cultured 16HBE cells. Histology and ELISA assays were used to evaluate airway inflammation. FITC-dextran permeability assay was used to evaluate the airway epithelial barrier function. Immunofluorescence, western blot and quantitative Real-Time PCR (RT-PCR) were used to evaluate the distribution and expression of tight junction proteins. RT-PCR and Ca2+ fluorescence measurement were used to evaluate the mRNA expression and activity of store-operated calcium entry (SOCE). RESULTS IL-37 inhibited house dust mite (HDM)-induced airway inflammation and decreased the levels of IgE in serum and type 2 cytokines in bronchoalveolar lavage fluid (BALF) compared to asthmatic mice. IL-37 protected against HDM-induced airway epithelial barrier dysfunction, including reduced leakage of FITC-dextran, enhanced expression of TJ proteins, and restored the membrane distribution of TJ proteins. Moreover, IL-37 decreased the level of IL-33 in the BALF of asthmatic mice and the supernatants of HDM-treated 16HBE cells. IL-37 decreased the peak level of Ca2+ fluorescence induced by thapsigargin and HDM, and inhibited the mRNA expression of Orai1, suggesting an inhibiting effect of IL-37 on SOCE in airway epithelial cells. CONCLUSION IL-37 plays a protective role in airway inflammation and HDM-induced airway epithelial barrier dysfunction by inhibiting SOCE.
Collapse
Affiliation(s)
- Changchang Wang
- Department of Allergology, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Jian Zhong
- Department of Allergology, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Jiaqian Hu
- Department of Allergology, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Can Cao
- Department of Allergology, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Shiquan Qi
- Department of Allergology, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Ruxue Ma
- Department of Allergology, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Wei Fu
- Department of Allergology, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Xiaolian Zhang
- Department of Allergology, Zhongnan Hospital of Wuhan University, Wuhan, China; Department of Immunology, School of Basic Medical Sciences, Wuhan University, Wuhan, China
| | - Cezmi A Akdis
- Swiss Institute of Allergy and Asthma Research (SIAF), University of Zurich, Davos, Switzerland
| | - Yadong Gao
- Department of Allergology, Zhongnan Hospital of Wuhan University, Wuhan, China; Department of Allergy, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China.
| |
Collapse
|
4
|
Wang Q, Zhang G, An C, Hambly BD, Bao S. The role of IL-37 in gastrointestinal diseases. Front Immunol 2024; 15:1431495. [PMID: 39206201 PMCID: PMC11349528 DOI: 10.3389/fimmu.2024.1431495] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2024] [Accepted: 07/31/2024] [Indexed: 09/04/2024] Open
Abstract
Gastrointestinal mucosal surface is frequently under challenge due to it's the large surface area and most common entry of microbes. IL-37, an anti-inflammatory cytokine, regulates local and systemic host immunity. H. pylori infection leads to the inhibition of IL-37 in the gastric mucosa, contributing to heightened mucosal inflammation and destruction, thereby facilitating increased proliferation of H. pylori. Food allergy, due to immune dysregulation, also contribute to GI injury. On the other hand, elevated levels of IL-37 observed in gastric cancer patients align with reduced host immunity at the cellular and humoral levels, indicating that IL-37 may contribute to the development of gastric cancer via suppressing pro-inflammatory responses. While IL-37 provides protection in an IBD animal model, the detection of highly produced IL-37 in IBD patients suggests a stage-dependent role, being protective in acute inflammation but potentially exacerbates the development of IBD in chronic conditions. Moreover, elevated colonic IL-37 in CRC correlates with overall survival time and disease time, indicating a protective role for IL-37 in CRC. The differential regulation and expression of IL-37 between upper- and lower-GI organs may be attributed to variations in the microbial flora. This information suggests that IL-37 could be a potential therapeutic agent, depending on the stage and location.
Collapse
Affiliation(s)
- Qiang Wang
- Department of Anatomy, School of Basic Medicine, Gansu University of Chinese Medicine, Lanzhou, China
| | - Guangrun Zhang
- School of Traditional Chinese and Western Medicine, Gansu University of Chinese Medicine, Lanzhou, China
| | - Caiping An
- Department of Nephrology, Gansu Provincial Hospital, Lanzhou, China
| | - Brett D. Hambly
- Centre for Healthy Futures, Torrens University Australia, Sydney, NSW, Australia
| | - Shisan Bao
- Foreign Affairs Office, The Third Affiliated Hospital of Gansu University of Chinese Medicine, Baiyin, China
- Foreign Affairs Office, The First People’s Hospital of Baiyin, Baiyin, China
| |
Collapse
|
5
|
Rusiñol L, Puig L. A Narrative Review of the IL-18 and IL-37 Implications in the Pathogenesis of Atopic Dermatitis and Psoriasis: Prospective Treatment Targets. Int J Mol Sci 2024; 25:8437. [PMID: 39126010 PMCID: PMC11312859 DOI: 10.3390/ijms25158437] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2024] [Revised: 07/30/2024] [Accepted: 08/01/2024] [Indexed: 08/12/2024] Open
Abstract
Atopic dermatitis and psoriasis are prevalent inflammatory skin conditions that significantly impact the quality of life of patients, with diverse treatment options available. Despite advances in understanding their underlying mechanisms, recent research highlights the significance of interleukins IL-18 and IL-37, in Th1, Th2, and Th17 inflammatory responses, closely associated with the pathogenesis of psoriasis and atopic dermatitis. Hence, IL-18 and IL-37 could potentially become therapeutic targets. This narrative review synthesizes knowledge on these interleukins, their roles in atopic dermatitis and psoriasis, and emerging treatment strategies. Findings of a literature search up to 30 May 2024, underscore a research gap in IL-37-targeted therapies. Conversely, IL-18-focused treatments have demonstrated promise in adult-onset Still's Disease, warranting further exploration for their potential efficacy in psoriasis and atopic dermatitis.
Collapse
Affiliation(s)
- Lluís Rusiñol
- Dermatology Department, Hospital de la Santa Creu i Sant Pau, 08025 Barcelona, Spain;
- Institut de Recerca Sant Pau (IR Sant Pau), Sant Quintí 77-79, 08041 Barcelona, Spain
- Unitat Docent Hospital Universitari Sant Pau, Universitat Autònoma de Barcelona, 08193 Bellaterra, Spain
| | - Lluís Puig
- Dermatology Department, Hospital de la Santa Creu i Sant Pau, 08025 Barcelona, Spain;
- Institut de Recerca Sant Pau (IR Sant Pau), Sant Quintí 77-79, 08041 Barcelona, Spain
- Unitat Docent Hospital Universitari Sant Pau, Universitat Autònoma de Barcelona, 08193 Bellaterra, Spain
| |
Collapse
|
6
|
Jiang L, Lunding LP, Webber WS, Beckmann K, Azam T, Falkesgaard Højen J, Amo-Aparicio J, Dinarello A, Nguyen TT, Pessara U, Parera D, Orlicky DJ, Fischer S, Wegmann M, Dinarello CA, Li S. An antibody to IL-1 receptor 7 protects mice from LPS-induced tissue and systemic inflammation. Front Immunol 2024; 15:1427100. [PMID: 38983847 PMCID: PMC11231367 DOI: 10.3389/fimmu.2024.1427100] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2024] [Accepted: 06/10/2024] [Indexed: 07/11/2024] Open
Abstract
Introduction Interleukin-18 (IL-18), a pro-inflammatory cytokine belonging to the IL-1 Family, is a key mediator ofautoinflammatory diseases associated with the development of macrophage activation syndrome (MAS).High levels of IL-18 correlate with MAS and COVID-19 severity and mortality, particularly in COVID-19patients with MAS. As an inflammation inducer, IL-18 binds its receptor IL-1 Receptor 5 (IL-1R5), leadingto the recruitment of the co-receptor, IL-1 Receptor 7 (IL-1R7). This heterotrimeric complex subsequentlyinitiates downstream signaling, resulting in local and systemic inflammation. Methods We reported earlier the development of a novel humanized monoclonal anti-human IL-1R7 antibody whichspecifically blocks the activity of human IL-18 and its inflammatory signaling in human cell and wholeblood cultures. In the current study, we further explored the strategy of blocking IL-1R7 inhyperinflammation in vivo using animal models. Results We first identified an anti-mouse IL-1R7 antibody that significantly suppressed mouse IL-18 andlipopolysaccharide (LPS)-induced IFNg production in mouse splenocyte and peritoneal cell cultures. Whenapplied in vivo, the antibody reduced Propionibacterium acnes and LPS-induced liver injury and protectedmice from tissue and systemic hyperinflammation. Importantly, anti-IL-1R7 significantly inhibited plasma,liver cell and spleen cell IFNg production. Also, anti-IL-1R7 downregulated plasma TNFa, IL-6, IL-1b,MIP-2 production and the production of the liver enzyme ALT. In parallel, anti-IL-1R7 suppressed LPSinducedinflammatory cell infiltration in lungs and inhibited the subsequent IFNg production andinflammation in mice when assessed using an acute lung injury model. Discussion Altogether, our data suggest that blocking IL-1R7 represents a potential therapeutic strategy to specificallymodulate IL-18-mediated hyperinflammation, warranting further investigation of its clinical application intreating IL-18-mediated diseases, including MAS and COVID-19.
Collapse
Affiliation(s)
- Liqiong Jiang
- Department of Medicine, University of Colorado Denver, Aurora, CO, United States
| | - Lars P. Lunding
- Division of Lung Immunology, Priority Area of Chronic Lung Diseases, Research Center Borstel-Leibniz Lung Center, Borstel, Germany
- Airway Research Center North, Member of the German Center for Lung Research (DZL), Giessen, Germany
| | - William S. Webber
- Department of Medicine, University of Colorado Denver, Aurora, CO, United States
| | | | - Tania Azam
- Department of Medicine, University of Colorado Denver, Aurora, CO, United States
| | - Jesper Falkesgaard Højen
- Department of Medicine, University of Colorado Denver, Aurora, CO, United States
- Department of Infectious Diseases, Aarhus University Hospital, Aarhus, Denmark
| | - Jesus Amo-Aparicio
- Department of Medicine, University of Colorado Denver, Aurora, CO, United States
| | - Alberto Dinarello
- Department of Medicine, University of Colorado Denver, Aurora, CO, United States
| | - Tom T. Nguyen
- Mucosal Inflammation Program and Division of Gastroenterology, Hepatology, and Nutrition, Department of Pediatrics, Children’s Hospital Colorado, University of Colorado, Aurora, CO, United States
| | - Ulrich Pessara
- MAB Discovery GmbH, Polling, Germany
- IcanoMAB GmbH, Polling, Germany
| | - Daniel Parera
- MAB Discovery GmbH, Polling, Germany
- IcanoMAB GmbH, Polling, Germany
| | - David J. Orlicky
- Department of Pathology, University of Colorado Denver Anschutz Medical Campus, Aurora, CO, United States
| | - Stephan Fischer
- MAB Discovery GmbH, Polling, Germany
- IcanoMAB GmbH, Polling, Germany
| | - Michael Wegmann
- Division of Lung Immunology, Priority Area of Chronic Lung Diseases, Research Center Borstel-Leibniz Lung Center, Borstel, Germany
- Airway Research Center North, Member of the German Center for Lung Research (DZL), Giessen, Germany
| | - Charles A. Dinarello
- Department of Medicine, University of Colorado Denver, Aurora, CO, United States
| | - Suzhao Li
- Department of Medicine, University of Colorado Denver, Aurora, CO, United States
| |
Collapse
|
7
|
Tong Y, Wang L, Wang L, Song J, Fan J, Lai C, Bao J, Weng C, Wang Y, Shuai J, Zhang H, Zhang W. Allergen immunotherapy combined with Notch pathway inhibitors improves HDM-induced allergic airway inflammation and inhibits ILC2 activation. Front Immunol 2024; 14:1264071. [PMID: 38371944 PMCID: PMC10869474 DOI: 10.3389/fimmu.2023.1264071] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2023] [Accepted: 12/29/2023] [Indexed: 02/20/2024] Open
Abstract
Introduction Group 2 innate lymphoid cells (ILC2s) play a crucial role in house dust mite (HDM)-induced allergic inflammation, and allergen immunotherapy (AIT) holds promise for treating the disease by reducing the frequency of ILC2s. Despite significant progress in AIT for allergic diseases, there remains a need to improve the control of allergic symptoms. Methods We investigated the synergistic effect of the Notch signaling pathway and subcutaneous immunotherapy (SCIT) in treating allergic airway inflammation in mice and their impact on the ratio of ILC2s in lung tissues. This was achieved by establishing the HDM-induced airway allergic disorders (HAAD) model and SCIT model. Additionally, we conducted in vitro investigations into the effect of the Notch signaling pathway on the secretory function of activated ILC2s using fluorescence-activated cell sorting. Furthermore, we explored the coactivation of the Notch signaling pathway with SCIT in vitro by sorting ILC2s from the lung tissues of mice after SCIT modeling. Results Previously, our group demonstrated that Notch signaling pathway inhibitors can reduce allergic airway inflammation in mice. Notch signaling induces lineage plasticity of mature ILC2s. In this study, we showed that AIT alleviates allergic airway inflammation and suppresses the frequency of ILC2s induced by HDM. Interestingly, AIT combined with a γ-secretase inhibitor (GSI), an inhibitor of the Notch signaling pathway, significantly inhibited the frequency of ILC2s, reduced airway inflammation, and suppressed Th2-type responses in a mouse model. Furthermore, lung ILC2s from HDM-challenged mice with or without AIT were treated with GSI in vitro, and we found that GSI dramatically reduced the secretion of type 2 inflammatory factors in ILC2s. Discussion These findings suggest that Notch signaling pathway inhibitors can be used as adjuvant therapy for AIT and may hold potential treatment value in the cooperative control of allergic airway inflammation during early AIT.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | | | - Hui Zhang
- Department of Pediatric Allergy and Immunology, The Second Hospital and Yuying Children’s Hospital of Wenzhou Medical University, Wenzhou, China
| | - Weixi Zhang
- Department of Pediatric Allergy and Immunology, The Second Hospital and Yuying Children’s Hospital of Wenzhou Medical University, Wenzhou, China
| |
Collapse
|
8
|
Zhou Y, Xu Z, Liu Z. Role of IL-33-ST2 pathway in regulating inflammation: current evidence and future perspectives. J Transl Med 2023; 21:902. [PMID: 38082335 PMCID: PMC10714644 DOI: 10.1186/s12967-023-04782-4] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2023] [Accepted: 12/01/2023] [Indexed: 12/18/2023] Open
Abstract
Interleukin (IL)-33 is an alarmin of the IL-1 superfamily localized to the nucleus of expressing cells, such as endothelial cells, epithelial cells, and fibroblasts. In response to cellular damage or stress, IL-33 is released and activates innate immune responses in some immune and structural cells via its receptor interleukin-1 receptor like-1 (IL-1RL1 or ST2). Recently, IL-33 has become a hot topic of research because of its role in pulmonary inflammation. The IL-33-ST2 signaling pathway plays a pro-inflammatory role by activating the type 2 inflammatory response, producing type 2 cytokines and chemokines. Elevated levels of IL-33 and ST2 have been observed in chronic pulmonary obstructive disease (COPD). Notably, IL-33 is present in COPD induced by cigarette smoke or acute inflammations. The role of IL-33 in sepsis is becoming increasingly prominent, and understanding its significance in the treatment of sepsis associated with high mortality is critical. In addition to its pro-inflammatory effects, the IL-33-ST2 axis appears to play a role in bacterial clearance and tissue repair. In this review, we focused on the role of the IL-33-ST2 axis in sepsis, asthma, and COPD and summarized the therapeutic targets associated with this axis, providing a basis for future treatment.
Collapse
Affiliation(s)
- Yilu Zhou
- Department of Anesthesiology, Shanghai First Maternity and Infant Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Zhendong Xu
- Department of Anesthesiology, Shanghai First Maternity and Infant Hospital, School of Medicine, Tongji University, Shanghai, China.
| | - Zhiqiang Liu
- Department of Anesthesiology, Shanghai First Maternity and Infant Hospital, School of Medicine, Tongji University, Shanghai, China.
| |
Collapse
|
9
|
Mesjasz A, Trzeciak M, Gleń J, Jaskulak M. Potential Role of IL-37 in Atopic Dermatitis. Cells 2023; 12:2766. [PMID: 38067193 PMCID: PMC10706414 DOI: 10.3390/cells12232766] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2023] [Revised: 11/24/2023] [Accepted: 11/28/2023] [Indexed: 12/18/2023] Open
Abstract
Interleukin 37 (IL-37) is a recently discovered member of the IL-1 cytokine family that appears to have anti-inflammatory and immunosuppressive effects in various diseases. IL-37 acts as a dual-function cytokine, exerting its effect extracellularly by forming a complex with the receptors IL-18 α (IL-18Rα) and IL-1R8 and transmitting anti-inflammatory signals, as well as intracellularly by interacting with Smad3, entering the nucleus, and inhibiting the transcription of pro-inflammatory genes. Consequently, IL-37 is linked to IL-18, which plays a role in the pathogenesis of atopic dermatitis (AD), consistent with our studies. Some isoforms of IL-37 are expressed by keratinocytes, monocytes, and other skin immune cells. IL-37 has been found to modulate the skewed T helper 2 (Th2) inflammation that is fundamental to the pathogenesis of AD. This review provides an up-to-date summary of the function of IL-37 in modulating the immune system and analyses its potential role in the pathogenesis of AD. Moreover, it speculates on IL-37's hypothetical value as a therapeutic target in the treatment of AD.
Collapse
Affiliation(s)
- Alicja Mesjasz
- Dermatological Students Scientific Association, Department of Dermatology, Venereology and Allergology, Faculty of Medicine, Medical University of Gdansk, 80-214 Gdansk, Poland;
| | - Magdalena Trzeciak
- Department of Dermatology, Venereology and Allergology, Faculty of Medicine, Medical University of Gdansk, 80-214 Gdansk, Poland;
| | - Jolanta Gleń
- Department of Dermatology, Venereology and Allergology, Faculty of Medicine, Medical University of Gdansk, 80-214 Gdansk, Poland;
| | - Marta Jaskulak
- Department of Immunobiology and Environmental Microbiology, Faculty of Health Sciences, Medical University of Gdansk, 80-214 Gdansk, Poland;
| |
Collapse
|
10
|
Wang Q, Guo L, Zeng Z, Huang Y, Tang H, Hu H, Yuan X, Deng J, Qin G, Wang X, Zhang Y. Neferine Attenuates HDM-Induced Allergic Inflammation by Inhibiting the Activation of Dendritic Cell. Inflammation 2023; 46:2433-2448. [PMID: 37702907 DOI: 10.1007/s10753-023-01891-6] [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: 04/05/2023] [Revised: 06/05/2023] [Accepted: 08/15/2023] [Indexed: 09/14/2023]
Abstract
House dust mite (HDM) acts as an environmental antigen that might cause chronic allergic diseases. Neferine (NEF) shows anti-inflammation therapeutic effects. This study is to explore the protection role of NEF against HDM-induced allergic inflammation. HDM-induced allergic asthmatic C57BL/6J mice models were established. Differential histological staining was used to analyze lung tissue pathological scores. Flow cytometry was used to analyze subtypes and biomarker expression of immune cells. RT-PCR and ELISA were used to test cytokines-related gene and/or protein expression levels. Western blot was performed to investigate the signaling pathway that mediates allergic inflammation from mice lung tissue and bone marrow-derived dendritic cells (BMDCs). H&E and PAS staining results indicate NEF significantly attenuated inflammatory index and the percentage of goblet cells in the lung tissue induced by HDM. The HDM-elevated TH2 and TH17 cells were significantly decreased by NEF; inflammatory cytokines Il-4, Il-13 and Il-17 were dramatically downregulated in the NEF plus HDM group compared with HDM alone. CD40+ and CD86+ DCs, eosinophils and mast cells, and ILC2 cells were decreased by NEF which was elevated under HDM stimulation. In vivo and ex vivo investigations indicated NEF can attenuate the activated NF-κB signaling induced by HDM is involved in allergic inflammatory immune response and regulates cytokines-related gene expression. HDM-activated DCs promoted differentiation of TH2 and TH17 cells but were attenuated by NEF. This study suggests NEF interrupts the overexpression of some cytokines released by DCs, TH2, and TH17 cells; NEF attenuates HDM-induced allergic inflammation via inhibiting NF-κB signaling of DCs.
Collapse
Affiliation(s)
- Qiao Wang
- Inflammation and Allergic Diseases Research Unit, The Affiliated Hospital of Southwest Medical University, Luzhou, 646000, China
- Department of Respiratory and Critical Care Medicine, The Affiliated Hospital of Southwest Medical University, Luzhou, 646000, China
| | - Linlin Guo
- Department of Obstetrics and Gynecology, The Ohio State University Wexner Medical Center, The Ohio State University, Columbus, OH, USA.
- Department of Microbiology and Immunology, Indiana University School of Medicine, Indianapolis, USA.
| | - Ziling Zeng
- Inflammation and Allergic Diseases Research Unit, The Affiliated Hospital of Southwest Medical University, Luzhou, 646000, China
- Department of Respiratory and Critical Care Medicine, The Affiliated Hospital of Southwest Medical University, Luzhou, 646000, China
| | - Yueru Huang
- Jiangsu Provincial Key Laboratory of Critical Care Medicine, Department of Critical Care Medicine, School of Medicine, Zhongda Hospital, Southeast University, Nanjing, 210009, China
| | - Hongmei Tang
- Inflammation and Allergic Diseases Research Unit, The Affiliated Hospital of Southwest Medical University, Luzhou, 646000, China
| | - Hang Hu
- Inflammation and Allergic Diseases Research Unit, The Affiliated Hospital of Southwest Medical University, Luzhou, 646000, China
- Department of Respiratory and Critical Care Medicine, The Affiliated Hospital of Southwest Medical University, Luzhou, 646000, China
| | - Xiefang Yuan
- Inflammation and Allergic Diseases Research Unit, The Affiliated Hospital of Southwest Medical University, Luzhou, 646000, China
| | - Jun Deng
- Department of Respiratory and Critical Care Medicine, The Affiliated Hospital of Southwest Medical University, Luzhou, 646000, China
| | - Gang Qin
- Department of Otolaryngology Head and Neck Surgery, the Affiliated Hospital of Southwest Medical University, Luzhou, 646000, China
| | - Xing Wang
- Inflammation and Allergic Diseases Research Unit, The Affiliated Hospital of Southwest Medical University, Luzhou, 646000, China.
| | - Yun Zhang
- Inflammation and Allergic Diseases Research Unit, The Affiliated Hospital of Southwest Medical University, Luzhou, 646000, China.
- Department of Respiratory and Critical Care Medicine, The Affiliated Hospital of Southwest Medical University, Luzhou, 646000, China.
| |
Collapse
|
11
|
Tsuji G, Yamamura K, Kawamura K, Kido-Nakahara M, Ito T, Nakahara T. Regulatory Mechanism of the IL-33-IL-37 Axis via Aryl Hydrocarbon Receptor in Atopic Dermatitis and Psoriasis. Int J Mol Sci 2023; 24:14633. [PMID: 37834081 PMCID: PMC10572928 DOI: 10.3390/ijms241914633] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2023] [Revised: 09/20/2023] [Accepted: 09/26/2023] [Indexed: 10/15/2023] Open
Abstract
Interleukin (IL)-33 and IL-37 have been identified as novel cytokines involved in various inflammatory diseases. However, their specific roles remain largely unknown. Recent studies have shown that IL-33, which triggers inflammation, and IL-37, which suppresses it, cooperatively regulate the balance between inflammation and anti-inflammation. IL-33 and IL-37 are also deeply involved in the pathogenesis of inflammatory skin diseases such as atopic dermatitis (AD) and psoriasis. Furthermore, a signaling pathway by which aryl hydrocarbon receptor (AHR), a receptor for dioxins, regulates the expression of IL-33 and IL-37 has been revealed. Here, we outline recent findings on the mechanisms regulating IL-33 and IL-37 expression in AD and psoriasis. IL-33 expression is partially dependent on mitogen-activated protein kinase (MAPK) activation, and IL-37 has a role in suppressing MAPK in human keratinocytes. Furthermore, IL-33 downregulates skin barrier function proteins including filaggrin and loricrin, thereby downregulating the expression of IL-37, which colocalizes with these proteins. This leads to an imbalance of the IL-33-IL-37 axis, involving increased IL-33 and decreased IL-37, which may be associated with the pathogenesis of AD and psoriasis. Therefore, AHR-mediated regulation of the IL-33-IL-37 axis may lead to new therapeutic strategies for the treatment of AD and psoriasis.
Collapse
Affiliation(s)
- Gaku Tsuji
- Research and Clinical Center for Yusho and Dioxin, Kyushu University Hospital, 3-1-1, Maidashi, Higashi-ku, Fukuoka 812-8582, Japan; (K.Y.); (T.N.)
- Department of Dermatology, Graduate School of Medical Sciences, Kyushu University, 3-1-1, Maidashi, Higashi-ku, Fukuoka 812-8582, Japan; (K.K.); (M.K.-N.); (T.I.)
| | - Kazuhiko Yamamura
- Research and Clinical Center for Yusho and Dioxin, Kyushu University Hospital, 3-1-1, Maidashi, Higashi-ku, Fukuoka 812-8582, Japan; (K.Y.); (T.N.)
- Department of Dermatology, Graduate School of Medical Sciences, Kyushu University, 3-1-1, Maidashi, Higashi-ku, Fukuoka 812-8582, Japan; (K.K.); (M.K.-N.); (T.I.)
| | - Koji Kawamura
- Department of Dermatology, Graduate School of Medical Sciences, Kyushu University, 3-1-1, Maidashi, Higashi-ku, Fukuoka 812-8582, Japan; (K.K.); (M.K.-N.); (T.I.)
| | - Makiko Kido-Nakahara
- Department of Dermatology, Graduate School of Medical Sciences, Kyushu University, 3-1-1, Maidashi, Higashi-ku, Fukuoka 812-8582, Japan; (K.K.); (M.K.-N.); (T.I.)
| | - Takamichi Ito
- Department of Dermatology, Graduate School of Medical Sciences, Kyushu University, 3-1-1, Maidashi, Higashi-ku, Fukuoka 812-8582, Japan; (K.K.); (M.K.-N.); (T.I.)
| | - Takeshi Nakahara
- Research and Clinical Center for Yusho and Dioxin, Kyushu University Hospital, 3-1-1, Maidashi, Higashi-ku, Fukuoka 812-8582, Japan; (K.Y.); (T.N.)
- Department of Dermatology, Graduate School of Medical Sciences, Kyushu University, 3-1-1, Maidashi, Higashi-ku, Fukuoka 812-8582, Japan; (K.K.); (M.K.-N.); (T.I.)
| |
Collapse
|
12
|
Frey A, Lunding LP, Wegmann M. The Dual Role of the Airway Epithelium in Asthma: Active Barrier and Regulator of Inflammation. Cells 2023; 12:2208. [PMID: 37759430 PMCID: PMC10526792 DOI: 10.3390/cells12182208] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2023] [Revised: 09/01/2023] [Accepted: 09/03/2023] [Indexed: 09/29/2023] Open
Abstract
Chronic airway inflammation is the cornerstone on which bronchial asthma arises, and in turn, chronic inflammation arises from a complex interplay between environmental factors such as allergens and pathogens and immune cells as well as structural cells constituting the airway mucosa. Airway epithelial cells (AECs) are at the center of these processes. On the one hand, they represent the borderline separating the body from its environment in order to keep inner homeostasis. The airway epithelium forms a multi-tiered, self-cleaning barrier that involves an unstirred, discontinuous mucous layer, the dense and rigid mesh of the glycocalyx, and the cellular layer itself, consisting of multiple, densely interconnected cell types. On the other hand, the airway epithelium represents an immunologically highly active tissue once its barrier has been penetrated: AECs play a pivotal role in releasing protective immunoglobulin A. They express a broad spectrum of pattern recognition receptors, enabling them to react to environmental stressors that overcome the mucosal barrier. By releasing alarmins-proinflammatory and regulatory cytokines-AECs play an active role in the formation, strategic orientation, and control of the subsequent defense reaction. Consequently, the airway epithelium is of vital importance to chronic inflammatory diseases, such as asthma.
Collapse
Affiliation(s)
- Andreas Frey
- Division of Mucosal Immunology and Diagnostics, Research Center Borstel, 23845 Borstel, Germany;
- Airway Research Center North (ARCN), German Center for Lung Research (DZL), 22927 Großhansdorf, Germany;
| | - Lars P. Lunding
- Airway Research Center North (ARCN), German Center for Lung Research (DZL), 22927 Großhansdorf, Germany;
- Division of Lung Immunology, Research Center Borstel, 23845 Borstel, Germany
| | - Michael Wegmann
- Airway Research Center North (ARCN), German Center for Lung Research (DZL), 22927 Großhansdorf, Germany;
- Division of Lung Immunology, Research Center Borstel, 23845 Borstel, Germany
| |
Collapse
|
13
|
Cui L, Qin X, Fu T, Li C, Wang D, Hu Y, Li Y, Chen Y, Cui Y, Wang J, Yuan H, Lv Z, Liu J, Xu D, Wei R, Ying S, Wang W. Attenuated airways inflammation and remodeling in IL-37a and IL-37b transgenic mice with an ovalbumin-induced chronic asthma. Cell Immunol 2023; 391-392:104759. [PMID: 37689011 DOI: 10.1016/j.cellimm.2023.104759] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2023] [Revised: 08/22/2023] [Accepted: 08/28/2023] [Indexed: 09/11/2023]
Abstract
BACKGROUND Asthma is a common chronic respiratory disease characterized by airways inflammation, hyperresponsiveness and remodeling. IL-37, an anti-inflammatory cytokine, consists of five splice isoforms, that is, a-e. Although it has been previously shown that recombinant human IL-37b is able to inhibit airway inflammation and hyperresponsiveness in animal models of asthma, the effects and difference of other IL-37 isoforms, such as IL-37a on features of asthma are unknown. METHODS Animal models of chronic asthma were established using IL-37a and IL-37b transgenic mice with C57BL/6J background and wild-type (WT) mice sensitized and nasally challenged with ovalbumin (OVA). Airway hyperresponsiveness was measured using FlexiVent apparatus, while histological and immunohistological stainings were employed to measure airways inflammation and remodeling indexes, including goblet cell metaplasia, mucus production, deposition of collagen, hypertrophy of airway smooth muscles and pulmonary angiogenesis. RESULTS Compared to WT mice, both IL-37a and IL-37b transgenic mice had significant reduced airway hyperresponsiveness and the declined total numbers of inflammatory cells, predominant eosinophils into airways and lung tissues. Furthermore, all features of airways remodeling, including degrees of mucus expression, collagen deposition, hypertrophy of smooth muscles, thickness of airways and neovascularization markedly decreased in IL-37 transgenic mice compared with OVA-treated WT mice. CONCLUSION Our data suggest that both IL-37a and IL-37b isoforms are able to not only ameliorate airways inflammation and airways hyperresponsiveness, but also greatly reduce airways structural changes of animal models of chronic asthma.
Collapse
Affiliation(s)
- Lele Cui
- Department of Immunology, School of Basic Medical Sciences, Capital Medical University, Beijing, China
| | - Xiaofeng Qin
- Department of Immunology, School of Basic Medical Sciences, Capital Medical University, Beijing, China
| | - Tingting Fu
- Department of Respiratory and Critical Care Medicine, Beijing Chao-Yang Hospital, Capital Medical University & Beijing Institute of Respiratory Medicine, Beijing, China
| | - Chenduo Li
- Department of Immunology, School of Basic Medical Sciences, Capital Medical University, Beijing, China
| | - Dan Wang
- Department of Immunology, School of Basic Medical Sciences, Capital Medical University, Beijing, China
| | - Yue Hu
- Department of Immunology, School of Basic Medical Sciences, Capital Medical University, Beijing, China
| | - Yan Li
- Department of Otorhinolaryngology Head and Neck Surgery, Beijing Tongren Hospital, Capital Medical University, Beijing Institute of Otorhinolaryngology, Key Laboratory of Otorhinolaryngology Head and Neck Surgery, Ministry of Education, Beijing Key Laboratory of Nasal Diseases, Beijing, China
| | - Yan Chen
- Department of Immunology, School of Basic Medical Sciences, Capital Medical University, Beijing, China
| | - Ye Cui
- Department of Immunology, School of Basic Medical Sciences, Capital Medical University, Beijing, China
| | - Jingjing Wang
- Department of Laboratory Animal Sciences, Capital Medical University, Beijing, China
| | - Huihui Yuan
- Department of Immunology, School of Basic Medical Sciences, Capital Medical University, Beijing, China
| | - Zhe Lv
- Department of Immunology, School of Basic Medical Sciences, Capital Medical University, Beijing, China
| | - Jie Liu
- Department of Immunology, School of Basic Medical Sciences, Capital Medical University, Beijing, China
| | - Damo Xu
- School of Medicine, Shenzhen University, Shenzhen, China; Department of General Practice Medicine, Third Affiliated Hospital of Shenzhen University, China
| | - Rongfei Wei
- National Laboratory of Biomacromolecules, CAS Center for Excellence in Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing, China
| | - Sun Ying
- Department of Immunology, School of Basic Medical Sciences, Capital Medical University, Beijing, China
| | - Wei Wang
- Department of Immunology, School of Basic Medical Sciences, Capital Medical University, Beijing, China.
| |
Collapse
|
14
|
Jiang B, Zhou Y, Liu Y, He S, Liao B, Peng T, Yao L, Qi L. Research Progress on the Role and Mechanism of IL-37 in Liver Diseases. Semin Liver Dis 2023; 43:336-350. [PMID: 37582401 PMCID: PMC10620037 DOI: 10.1055/a-2153-8836] [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] [Indexed: 08/17/2023]
Abstract
Cytokines are important components of the immune system that can predict or influence the development of liver diseases. IL-37, a new member of the IL-1 cytokine family, exerts potent anti-inflammatory and immunosuppressive effects inside and outside cells. IL-37 expression differs before and after liver lesions, suggesting that it is associated with liver disease; however, its mechanism of action remains unclear. This article mainly reviews the biological characteristics of IL-37, which inhibits hepatitis, liver injury, and liver fibrosis by inhibiting inflammation, and inhibits the development of hepatocellular carcinoma (HCC) by regulating the immune microenvironment. Based on additional evidence, combining IL-37 with liver disease markers for diagnosis and treatment can achieve more significant effects, suggesting that IL-37 can be developed into a powerful tool for the clinical adjuvant treatment of liver diseases, especially HCC.
Collapse
Affiliation(s)
- Baoyi Jiang
- Institute of Digestive Disease, The Sixth Affiliated Hospital of Guangzhou Medical University, Qingyuan People's Hospital, Qingyuan, China
| | - Yulin Zhou
- Department of Clinical Laboratory, Shunde New Rongqi Hospital, Foshan, China
| | - Yanting Liu
- Institute of Digestive Disease, The Sixth Affiliated Hospital of Guangzhou Medical University, Qingyuan People's Hospital, Qingyuan, China
| | - Siqi He
- Institute of Digestive Disease, The Sixth Affiliated Hospital of Guangzhou Medical University, Qingyuan People's Hospital, Qingyuan, China
| | - Baojian Liao
- Institute of Digestive Disease, The Sixth Affiliated Hospital of Guangzhou Medical University, Qingyuan People's Hospital, Qingyuan, China
| | - Tieli Peng
- Institute of Digestive Disease, The Sixth Affiliated Hospital of Guangzhou Medical University, Qingyuan People's Hospital, Qingyuan, China
| | - Leyi Yao
- Institute of Digestive Disease, The Sixth Affiliated Hospital of Guangzhou Medical University, Qingyuan People's Hospital, Qingyuan, China
| | - Ling Qi
- Institute of Digestive Disease, The Sixth Affiliated Hospital of Guangzhou Medical University, Qingyuan People's Hospital, Qingyuan, China
| |
Collapse
|
15
|
Ji T, Li H. T-helper cells and their cytokines in pathogenesis and treatment of asthma. Front Immunol 2023; 14:1149203. [PMID: 37377958 PMCID: PMC10291091 DOI: 10.3389/fimmu.2023.1149203] [Citation(s) in RCA: 27] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2023] [Accepted: 05/30/2023] [Indexed: 06/29/2023] Open
Abstract
Prosperous advances in understanding the cellular and molecular mechanisms of chronic inflammation and airway remodeling in asthma have been made over the past several decades. Asthma is a chronic inflammatory disease of the airways characterized by reversible airway obstruction that is self-resolving or remits with treatment. Around half of asthma patients are "Type-2-high" asthma with overexpression of type 2 inflammatory pathways and elevated type 2 cytokines. When stimulated by allergens, airway epithelial cells secrete IL-25, IL-33, and TSLP to derive a Th2 immune response. First ILC2 followed by Th2 cells produces a series of cytokines such as IL-4, IL-5, and IL-13. TFH cells control IgE synthesis by secreting IL-4 to allergen-specific B cells. IL-5 promotes eosinophil inflammation, while IL-13 and IL-4 are involved in goblet cell metaplasia and bronchial hyperresponsiveness. Currently, "Type-2 low" asthma is defined as asthma with low levels of T2 biomarkers due to the lack of reliable biomarkers, which is associated with other Th cells. Th1 and Th17 are capable of producing cytokines that recruit neutrophils, such as IFN-γ and IL-17, to participate in the development of "Type-2-low" asthma. Precision medicine targeting Th cells and related cytokines is essential in the management of asthma aiming at the more appropriate patient selection and better treatment response. In this review, we sort out the pathogenesis of Th cells in asthma and summarize the therapeutic approaches involved as well as potential research directions.
Collapse
Affiliation(s)
| | - Hequan Li
- Department of Respiratory and Critical Care Medicine, the First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, China
| |
Collapse
|
16
|
Striz I, Golebski K, Strizova Z, Loukides S, Bakakos P, Hanania N, Jesenak M, Diamant Z. New insights into the pathophysiology and therapeutic targets of asthma and comorbid chronic rhinosinusitis with or without nasal polyposis. Clin Sci (Lond) 2023; 137:727-753. [PMID: 37199256 PMCID: PMC10195992 DOI: 10.1042/cs20190281] [Citation(s) in RCA: 19] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2023] [Revised: 04/22/2023] [Accepted: 04/28/2023] [Indexed: 05/19/2023]
Abstract
Asthma and chronic rhinosinusitis with nasal polyps (CRSwNP) or without (CRSsNP) are chronic respiratory diseases. These two disorders often co-exist based on common anatomical, immunological, histopathological, and pathophysiological basis. Usually, asthma with comorbid CRSwNP is driven by type 2 (T2) inflammation which predisposes to more severe, often intractable, disease. In the past two decades, innovative technologies and detection techniques in combination with newly introduced targeted therapies helped shape our understanding of the immunological pathways underlying inflammatory airway diseases and to further identify several distinct clinical and inflammatory subsets to enhance the development of more effective personalized treatments. Presently, a number of targeted biologics has shown clinical efficacy in patients with refractory T2 airway inflammation, including anti-IgE (omalizumab), anti-IL-5 (mepolizumab, reslizumab)/anti-IL5R (benralizumab), anti-IL-4R-α (anti-IL-4/IL-13, dupilumab), and anti-TSLP (tezepelumab). In non-type-2 endotypes, no targeted biologics have consistently shown clinical efficacy so far. Presently, multiple therapeutical targets are being explored including cytokines, membrane molecules and intracellular signalling pathways to further expand current treatment options for severe asthma with and without comorbid CRSwNP. In this review, we discuss existing biologics, those under development and share some views on new horizons.
Collapse
Affiliation(s)
- Ilja Striz
- Department of Clinical and Transplant Immunology, Institute for Clinical and Experimental Medicine, Prague, Czech Republic
- Institute of Immunology and Microbiology, First Faculty of Medicine, Charles University, Prague, Czech Republic
- Subdivision of Allergology and Clinical Immunology, Institute for Postgraduate Education in Medicine, Prague, Czech Republic
| | - Kornel Golebski
- Department of Pulmonary Medicine, Amsterdam University Medical Centers, University of Amsterdam, the Netherlands
| | - Zuzana Strizova
- Institute of Immunology, Second Faculty of Medicine, Charles University and Motol University Hospital, Prague, Czech Republic
| | - Stelios Loukides
- Department of Respiratory Medicine, National and Kapodistrian University of Athens, Athens, Greece
| | - Petros Bakakos
- First Respiratory Medicine Department, National and Kapodistrian University of Athens, Athens, Greece
| | - Nicola A. Hanania
- Section of Pulmonary and Critical Care Medicine, Baylor College of Medicine, Houston, TX, USA
| | - Milos Jesenak
- Department of Pulmonology and Phthisiology, Jessenius Faculty of Medicine in Martin, Comenius University in Bratislava, University Hospital in Martin, Slovakia
- Department of Pediatrics, Jessenius Faculty of Medicine in Martin, Comenius University in Bratislava, University Hospital in Martin, Slovakia
- Department of Clinical Immunology and Allergology, University Hospital in Martin, Slovakia
| | - Zuzana Diamant
- Department of Microbiology Immunology and Transplantation, KU Leuven, Catholic University of Leuven, Belgium
- Department of Respiratory Medicine and Allergology, Institute for Clinical Science, Skane University Hospital, Lund University, Lund, Sweden
- Department of Respiratory Medicine, First Faculty of Medicine, Charles University and Thomayer Hospital, Prague, Czech Republic
- Department of Clinical Pharmacy and Pharmacology, University of Groningen, University Medical Center Groningen, Groningen, Netherlands
| |
Collapse
|
17
|
Ren C, Chen J, Che Q, Jia Q, Lu H, Qi X, Zhang X, Shu Q. IL-37 alleviates TNF-α-induced pyroptosis of rheumatoid arthritis fibroblast-like synoviocytes by inhibiting the NF-κB/GSDMD signaling pathway. Immunobiology 2023; 228:152382. [PMID: 37075579 DOI: 10.1016/j.imbio.2023.152382] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2023] [Revised: 03/09/2023] [Accepted: 03/31/2023] [Indexed: 04/07/2023]
Abstract
OBJECTIVE Pyroptosis is crucial to rheumatoid arthritis (RA) by inducing and aggravating inflammation. TNF-α is abundant in fibroblast-like synoviocytes of RA (RA-FLSs) and plays a key role in pyroptosis by inducing nuclear factor (NF)-κB activation. Additionally, interleukin (IL)-37 is involved in autoimmune diseases as an anti-inflammatory cytokine and innate and acquired immune response inhibitor. However, the effect of IL-37 on pyroptosis in RA-FLSs remains unclear. Therefore, this study investigated the effects and mechanism of IL-37 on RA-FLS pyroptosis induced by TNF-α. METHODS In this study, the serum cytokines in patients with RA and healthy controls were detected using ELISA. The RA-FLSs were then cultured with TNF-α, with or without various IL-37 concentrations, to test the cytokine levels in the cell supernatant. 5-Ethynyl-2'-Deoxyuridine (EdU) assay assessed the effects of IL-37 on RA FLS proliferation. RA-FLS apoptosis was assessed using flow cytometry and mitochondrial membrane potential (MMP) measurement. In addition, transmission electron microscopy (TEM) was used to examine cell pyroptosis. We selected the optimal concentration for the following experiments and detected the signal pathway of IL-37 on pyroptosis of RA FLSs by quantitative reverse-transcription polymerase chain reaction (qRT-PCR) and Western blotting. Finally, we validated the therapeutic effects of IL-37 on CIA rat model in vivo. RESULTS IL-37 inhibited inflammation in vitro and in vivo and reduced pyroptosis-related protein expression in RA FLSs. Furthermore, we determined that nuclear factor κB (NF-κB) signaling is required for GSDMD-mediated pyroptosis in RA FLSs. CONCLUSION IL-37 alleviates TNF-α-induced pyroptosis of RA FLSs by inhibiting NF-κB/GSDMD signaling. Additionally, our data revealed a novel mechanism for IL-37 in RA FLSs, suggesting a new potential therapy for IL-37 to treat RA.
Collapse
|
18
|
Li Y, Jiang Q. Uncoupled pyroptosis and IL-1β secretion downstream of inflammasome signaling. Front Immunol 2023; 14:1128358. [PMID: 37090724 PMCID: PMC10117957 DOI: 10.3389/fimmu.2023.1128358] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2022] [Accepted: 03/24/2023] [Indexed: 04/25/2023] Open
Abstract
Inflammasomes are supramolecular platforms that organize in response to various damage-associated molecular patterns and pathogen-associated molecular patterns. Upon activation, inflammasome sensors (with or without the help of ASC) activate caspase-1 and other inflammatory caspases that cleave gasdermin D and pro-IL-1β/pro-IL-18, leading to pyroptosis and mature cytokine secretion. Pyroptosis enables intracellular pathogen niche disruption and intracellular content release at the cost of cell death, inducing pro-inflammatory responses in the neighboring cells. IL-1β is a potent pro-inflammatory regulator for neutrophil recruitment, macrophage activation, and T-cell expansion. Thus, pyroptosis and cytokine secretion are the two main mechanisms that occur downstream of inflammasome signaling; they maintain homeostasis, drive the innate immune response, and shape adaptive immunity. This review aims to discuss the possible mechanisms, timing, consequences, and significance of the two uncoupling preferences downstream of inflammasome signaling. While pyroptosis and cytokine secretion may be usually coupled, pyroptosis-predominant and cytokine-predominant uncoupling are also observed in a stimulus-, cell type-, or context-dependent manner, contributing to the pathogenesis and development of numerous pathological conditions such as cryopyrin-associated periodic syndromes, LPS-induced sepsis, and Salmonella enterica serovar Typhimurium infection. Hyperactive cells consistently release IL-1β without LDH leakage and pyroptotic death, thereby leading to prolonged inflammation, expanding the lifespans of pyroptosis-resistant neutrophils, and hyperactivating stimuli-challenged macrophages, dendritic cells, monocytes, and specific nonimmune cells. Death inflammasome activation also induces GSDMD-mediated pyroptosis with no IL-1β secretion, which may increase lethality in vivo. The sublytic GSDMD pore formation associated with lower expressions of pyroptotic components, GSDMD-mediated extracellular vesicles, or other GSDMD-independent pathways that involve unconventional secretion could contribute to the cytokine-predominant uncoupling; the regulation of caspase-1 dynamics, which may generate various active species with different activities in terms of GSDMD or pro-IL-1β, could lead to pyroptosis-predominant uncoupling. These uncoupling preferences enable precise reactions to different stimuli of different intensities under specific conditions at the single-cell level, promoting cooperative cell and host fate decisions and participating in the pathogen "game". Appropriate decisions in terms of coupling and uncoupling are required to heal tissues and eliminate threats, and further studies exploring the inflammasome tilt toward pyroptosis or cytokine secretion may be helpful.
Collapse
|
19
|
Kanannejad Z, Soleimanian S, Ghahramani Z, Sepahi N, Mohkam M, Alyasin S, Kheshtchin N. Immune checkpoint molecules in prevention and development of asthma. Front Immunol 2023; 14:1070779. [PMID: 36865540 PMCID: PMC9972681 DOI: 10.3389/fimmu.2023.1070779] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2022] [Accepted: 01/30/2023] [Indexed: 02/16/2023] Open
Abstract
Allergic asthma is a respiratory disease initiated by type-2 immune responses characterized by secretion of alarmins, interleukin-4 (IL-4), IL-5, and IL-13, eosinophilic inflammation, and airway hyperresponsiveness (AHR). Immune checkpoints (ICPs) are inhibitory or stimulatory molecules expressed on different immune cells, tumor cells, or other cell types that regulate immune system activation and maintain immune homeostasis. Compelling evidence indicates a key role for ICPs in both the progression and prevention of asthma. There is also evidence of asthma development or exacerbation in some cancer patients receiving ICP therapy. The aim of this review is to provide an updated overview of ICPs and their roles in asthma pathogenesis, and to assess their implications as therapeutic targets in asthma.
Collapse
Affiliation(s)
- Zahra Kanannejad
- Allergy Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Saeede Soleimanian
- Allergy Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Zahra Ghahramani
- Hematology Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Najmeh Sepahi
- Allergy Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Milad Mohkam
- Allergy Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Soheila Alyasin
- Allergy Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Nasim Kheshtchin
- Allergy Research Center, Shiraz University of Medical Sciences, Shiraz, Iran.,Department of Immunology, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran
| |
Collapse
|
20
|
Borgia F, Custurone P, Li Pomi F, Vaccaro M, Alessandrello C, Gangemi S. IL-33 and IL-37: A Possible Axis in Skin and Allergic Diseases. Int J Mol Sci 2022; 24:ijms24010372. [PMID: 36613827 PMCID: PMC9820694 DOI: 10.3390/ijms24010372] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2022] [Revised: 12/21/2022] [Accepted: 12/23/2022] [Indexed: 12/28/2022] Open
Abstract
Interleukin (IL)-37 and IL-33 are among the latest cytokines identified, playing a role in several inflammatory conditions, spanning from systemic conditions to tumors to localized diseases. As newly discovered interleukins, their role is still scarcely understood, but their potential role as therapeutic targets or disease activity markers suggests the need to reorganize the current data for a better interpretation. The aim of this review is to collect and organize data produced by several studies to create a complete picture. The research was conducted on the PubMed database, and the resulting articles were sorted by title, abstract, English language, and content. Several studies have been assessed, mostly related to atopic dermatitis and immunologic pathways. Collective data demonstrates a pro-inflammatory role of IL-33 and an anti-inflammatory one for IL-37, possibly related to each other in an IL-33/IL-37 axis. Although further studies are needed to assess the safety and plausibility of targeting these two interleukins for patients affected by skin conditions, the early results indicate that both IL-33 and IL-37 represent markers of disease activity.
Collapse
Affiliation(s)
- Francesco Borgia
- Department of Clinical and Experimental Medicine, Section of Dermatology, University of Messina, 98125 Messina, Italy
- Correspondence:
| | - Paolo Custurone
- Department of Clinical and Experimental Medicine, Section of Dermatology, University of Messina, 98125 Messina, Italy
| | - Federica Li Pomi
- Department of Clinical and Experimental Medicine, Section of Dermatology, University of Messina, 98125 Messina, Italy
| | - Mario Vaccaro
- Department of Clinical and Experimental Medicine, Section of Dermatology, University of Messina, 98125 Messina, Italy
| | - Clara Alessandrello
- Department of Clinical and Experimental Medicine, School and Operative Unit of Allergy and Clinical Immunology, University of Messina, 98125 Messina, Italy
| | - Sebastiano Gangemi
- Department of Clinical and Experimental Medicine, School and Operative Unit of Allergy and Clinical Immunology, University of Messina, 98125 Messina, Italy
| |
Collapse
|
21
|
Nold-Petry CA, Nold MF. Rationale for IL-37 as a novel therapeutic agent in inflammation. Expert Rev Clin Immunol 2022; 18:1203-1206. [PMID: 35916240 DOI: 10.1080/1744666x.2022.2108792] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Affiliation(s)
- Claudia A Nold-Petry
- Ritchie Centre, Hudson Institute of Medical Research, Melbourne, Australia.,Department of Paediatrics, School of Clinical Sciences, Monash University, Melbourne, Australia
| | - Marcel F Nold
- Ritchie Centre, Hudson Institute of Medical Research, Melbourne, Australia.,Department of Paediatrics, School of Clinical Sciences, Monash University, Melbourne, Australia.,Monash Newborn, Monash Children's Hospital, Melbourne, Australia
| |
Collapse
|
22
|
Adjuvant role of probiotics in allergen-specific immunotherapy. Clin Immunol 2022; 245:109164. [DOI: 10.1016/j.clim.2022.109164] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2022] [Revised: 10/04/2022] [Accepted: 10/06/2022] [Indexed: 11/19/2022]
|
23
|
Wypych-Ślusarska A, Grot M, Kujawińska M, Nigowski M, Krupa-Kotara K, Oleksiuk K, Głogowska-Ligus J, Grajek M. Respiratory Symptoms, Allergies, and Environmental Exposures in Children with and without Asthma. INTERNATIONAL JOURNAL OF ENVIRONMENTAL RESEARCH AND PUBLIC HEALTH 2022; 19:11180. [PMID: 36141448 PMCID: PMC9517590 DOI: 10.3390/ijerph191811180] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/14/2022] [Revised: 09/02/2022] [Accepted: 09/03/2022] [Indexed: 06/16/2023]
Abstract
BACKGROUND Epidemiological data concerning the level of asthma morbidity indicate that in Poland, asthma is diagnosed in 5-10% of the pediatric population. Aim The purpose of this study was to compare the prevalence of respiratory symptoms and allergies in a group of children with and without asthma and to evaluate the association between exposure to environmental factors and the prevalence of bronchial asthma in a pediatric population. MATERIAL AND METHODS A cross-sectional study was conducted on a group of 995 children attending primary schools in the province of Silesia in 2018-2019. The research tool was an anonymous questionnaire developed based on the form used in The International Study of Asthma and Allergies in Childhood (ISAAC). Children's health status, the prevalence of bronchial asthma, and the performance of allergic skin tests were assessed based on parents' indications in a questionnaire. Environmental exposures such as mold and dampness in apartments or ETS were similarly assessed. Analyses were performed using Statistica 13.0; p < 0.05. RESULTS A total of 88 subjects (8.8%) suffered from bronchial asthma. Parents of children with asthma, compared to parents of children without the disease, were more likely to rate their children's health as rather good (43.2% vs. 38.0%) or average (21.6% vs. 3.1%). All analyzed respiratory symptoms, as well as allergies, were statistically more frequent in children with bronchial asthma. CONCLUSIONS The parent's subjective assessment of the child's health varied significantly according to the asthma diagnosis. Asthma is also associated with other diseases: allergic reactions to pollen, house dust, hay fever, and AD (atopic dermatitis) were statistically significantly more frequent among children diagnosed with bronchial asthma.
Collapse
Affiliation(s)
- Agata Wypych-Ślusarska
- Department of Epidemiology, Faculty of Health Sciences in Bytom, Medical University of Silesia in Katowice, 40-055 Katowice, Poland
| | - Martina Grot
- Department of Epidemiology, Faculty of Health Sciences in Bytom, Medical University of Silesia in Katowice, 40-055 Katowice, Poland
| | - Maria Kujawińska
- Department of Epidemiology, Faculty of Health Sciences in Bytom, Medical University of Silesia in Katowice, 40-055 Katowice, Poland
| | - Maciej Nigowski
- Department of Epidemiology, Faculty of Health Sciences in Bytom, Medical University of Silesia in Katowice, 40-055 Katowice, Poland
| | - Karolina Krupa-Kotara
- Department of Epidemiology, Faculty of Health Sciences in Bytom, Medical University of Silesia in Katowice, 40-055 Katowice, Poland
| | - Klaudia Oleksiuk
- Department of Epidemiology, Faculty of Health Sciences in Bytom, Medical University of Silesia in Katowice, 40-055 Katowice, Poland
| | - Joanna Głogowska-Ligus
- Department of Epidemiology, Faculty of Health Sciences in Bytom, Medical University of Silesia in Katowice, 40-055 Katowice, Poland
| | - Mateusz Grajek
- Department of Public Health, Department of Public Health Policy, Faculty of Health Sciences in Bytom, Medical University of Silesia in Katowice, 40-055 Katowice, Poland
| |
Collapse
|
24
|
Luo W, Hu J, Xu W, Dong J. Distinct spatial and temporal roles for Th1, Th2, and Th17 cells in asthma. Front Immunol 2022; 13:974066. [PMID: 36032162 PMCID: PMC9411752 DOI: 10.3389/fimmu.2022.974066] [Citation(s) in RCA: 38] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2022] [Accepted: 07/28/2022] [Indexed: 12/24/2022] Open
Abstract
Immune response in the asthmatic respiratory tract is mainly driven by CD4+ T helper (Th) cells, represented by Th1, Th2, and Th17 cells, especially Th2 cells. Asthma is a heterogeneous and progressive disease, reflected by distinct phenotypes orchestrated by τh2 or non-Th2 (Th1 and Th17) immune responses at different stages of the disease course. Heterogeneous cytokine expression within the same Th effector state in response to changing conditions in vivo and interlineage relationship among CD4+ T cells shape the complex immune networks of the inflammatory airway, making it difficult to find one panacea for all asthmatics. Here, we review the role of three T helper subsets in the pathogenesis of asthma from different stages, highlighting timing is everything in the immune system. We also discuss the dynamic topography of Th subsets and pathogenetic memory Th cells in asthma.
Collapse
Affiliation(s)
- Weihang Luo
- Department of Integrative Medicine, Huashan Hospital, Fudan University, Shanghai, China
- Institutes of Integrative Medicine, Fudan University, Shanghai, China
| | - Jindong Hu
- Department of Integrative Medicine, Huashan Hospital, Fudan University, Shanghai, China
| | - Weifang Xu
- Shenzhen Hospital of Guangzhou University of Chinese Medicine (Futian), Shenzhen, China
- *Correspondence: Jingcheng Dong, ; Weifang Xu,
| | - Jingcheng Dong
- Department of Integrative Medicine, Huashan Hospital, Fudan University, Shanghai, China
- Institutes of Integrative Medicine, Fudan University, Shanghai, China
- *Correspondence: Jingcheng Dong, ; Weifang Xu,
| |
Collapse
|
25
|
Zissler UM, Thron A, Eckrich J, Bakhtiar S, Schubert R, Zielen S. Bronchial inflammation biomarker patterns link humoral immunodeficiency with bronchiectasis-related small airway dysfunction. Clin Exp Allergy 2022; 52:760-773. [PMID: 35353925 DOI: 10.1111/cea.14140] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2021] [Revised: 03/09/2022] [Accepted: 03/27/2022] [Indexed: 12/16/2022]
Abstract
BACKGROUND The progression of chronic destructive lung disease in patients with humoral immunodeficiency (ID) and concomitant development of bronchiectasis is difficult to prevent. Lung function tests in these patients typically show bronchial obstruction of the small airways in combination with increased air trapping in the distal airways, which is consistent with small airway dysfunction. OBJECTIVE The objective was to assess the grade of chronic lower airway inflammation and small airway dysfunction from induced sputum and the corresponding local pro-inflammatory mediator pattern to discriminate patients affected by bronchiectasis-related Small Airway Dysfunction (SAD). METHODS In a prospective design, 22 patients with ID (14 CVID, 3 XLA, 3 hyper-IgM syndrome, 1 hyper-IgE syndrome and low IgG levels due to treatment with rituximab and 1 SCID after BMT and persistent humoral defect) and 21 healthy controls were examined. Lung function, Fraction Expiratory Nitric Oxide (FeNO) and pro-inflammatory cytokine levels were compared in subsets of patients with (ID + BE) and without bronchiectasis (ID) pre-stratified using high-resolution computed tomography (HRCT) scans and control subjects. RESULTS Analysis of induced sputum showed significantly increased total cell counts and severe neutrophilic inflammation in ID. The concomitant SAD revealed higher total cell numbers compared to ID. Bronchial inflammation in ID is clearly mirrored by pro-inflammatory mediators IL-1β, IL-6 and CXCL-8, whilst TNF-α revealed a correlation with lung function parameters altered in the context of bronchiectasis-related Small Airway Dysfunction. CONCLUSIONS In spite of immunoglobulin substitution, bronchial inflammation was dominated by neutrophils and was highly increased in patients with ID + BE. Notably, the pro-inflammatory cytokines in patients with ID were significantly increased in induced sputum. The context-dependent cytokine pattern in relation to the presence of concomitant bronchiectasis associated with SAD in ID patients could be helpful in delimiting ID patient subgroups and individualizing therapeutic approaches.
Collapse
Affiliation(s)
- Ulrich M Zissler
- Center of Allergy & Environment (ZAUM), Technical University of Munich and Helmholtz Center Munich, German Research Center for Environmental Health, Germany, Member of the German Center for Lung Research (DZL), Munich, Germany
| | - Aljoscha Thron
- Division of Allergology, Pulmonology and Cystic Fibrosis, Department for Children and Adolescents, Goethe University, Frankfurt, Germany
| | - Jonas Eckrich
- Department of Otorhinolaryngology, University Medical Center Bonn (UKB), Bonn, Germany
| | - Shahrzad Bakhtiar
- Division for Stem Cell Transplantation, Immunology and Intensive Care Unit, Department for Children and Adolescents, Goethe University, Frankfurt, Germany
| | - Ralf Schubert
- Division of Allergology, Pulmonology and Cystic Fibrosis, Department for Children and Adolescents, Goethe University, Frankfurt, Germany
| | - Stefan Zielen
- Division of Allergology, Pulmonology and Cystic Fibrosis, Department for Children and Adolescents, Goethe University, Frankfurt, Germany
| |
Collapse
|
26
|
Ruysseveldt E, Martens K, Steelant B. Airway Basal Cells, Protectors of Epithelial Walls in Health and Respiratory Diseases. FRONTIERS IN ALLERGY 2021; 2:787128. [PMID: 35387001 PMCID: PMC8974818 DOI: 10.3389/falgy.2021.787128] [Citation(s) in RCA: 43] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2021] [Accepted: 10/25/2021] [Indexed: 01/02/2023] Open
Abstract
The airway epithelium provides a critical barrier to the outside environment. When its integrity is impaired, epithelial cells and residing immune cells collaborate to exclude pathogens and to heal tissue damage. Healing is achieved through tissue-specific stem cells: the airway basal cells. Positioned near the basal membrane, airway basal cells sense and respond to changes in tissue health by initiating a pro-inflammatory response and tissue repair via complex crosstalks with nearby fibroblasts and specialized immune cells. In addition, basal cells have the capacity to learn from previous encounters with the environment. Inflammation can indeed imprint a certain memory on basal cells by epigenetic changes so that sensitized tissues may respond differently to future assaults and the epithelium becomes better equipped to respond faster and more robustly to barrier defects. This memory can, however, be lost in diseased states. In this review, we discuss airway basal cells in respiratory diseases, the communication network between airway basal cells and tissue-resident and/or recruited immune cells, and how basal cell adaptation to environmental triggers occurs.
Collapse
Affiliation(s)
- Emma Ruysseveldt
- Allergy and Clinical Immunology Research Unit, Department of Microbiology, Immunology and Transplantation, KU Leuven, Leuven, Belgium
| | - Katleen Martens
- Allergy and Clinical Immunology Research Unit, Department of Microbiology, Immunology and Transplantation, KU Leuven, Leuven, Belgium
- Department of Bioscience Engineering, University of Antwerp, Antwerp, Belgium
| | - Brecht Steelant
- Allergy and Clinical Immunology Research Unit, Department of Microbiology, Immunology and Transplantation, KU Leuven, Leuven, Belgium
- Head and Neck Surgery, Department of Otorhinolaryngology, University of Crete School of Medicine, Heraklion, Greece
| |
Collapse
|