1
|
Yang Q, Zheng Y, Dong J, Xu Q, Li S, Li A, Xiong X, Zhang Y. Serum interleukin-33 combined with FEF75% z-score and FeNO improves the diagnostic accuracy of asthma in children. J Pediatr (Rio J) 2024; 100:81-87. [PMID: 37741632 PMCID: PMC10751693 DOI: 10.1016/j.jped.2023.08.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/18/2023] [Revised: 08/15/2023] [Accepted: 08/17/2023] [Indexed: 09/25/2023] Open
Abstract
OBJECTIVE To investigate the diagnostic efficacy of serum IL-33 single indicator and combined indicators for asthma in children. METHODS 132 children were initially diagnosed with asthma during acute exacerbation and 100 healthy children were included. Serum IL-33 concentration differences were compared between asthmatic and normal children. Correlations between IL-33 with pulmonary function parameters, FeNO, peripheral blood EOS counts and serum total IgE were analyzed in asthmatic children. ROC curves were used to assess IL-33 diagnostic efficacy and its combined indicators. To prevent overfitting of the predictive model, the hold-out cross-validation method was used. RESULTS (1) Serum IL-33 concentrations were significantly higher in children with asthma than in normal children (p < 0.001). (2) IL-33 concentration was negatively correlated with FVC z-score, FEV1 z-score and FEF75% z-score in asthmatic children (p < 0.05). (3) The area under the ROC curve of IL-33 was 0.821, which was higher than those of FeNO, FVC z-score, and FEV1 z-score. (4) Cross-validation of the combined indicators showed that IL-33 significantly improved asthma diagnostic efficacy. The combination of IL-33, FEF75% z-score, and FeNO showed the highest diagnostic efficacy, with the AUC, sensitivity, and specificity of the combined indicator being 0.954, 90.1%, and 89. 0%, respectively, and good extrapolation of the predictive model. CONCLUSION Serum IL-33 is higher in children with asthma and increases with the severity of pulmonary ventilation obstruction. A single indicator of serum IL-33 demonstrates moderate diagnostic accuracy, and its combination with FEF75% z-score and FeNO significantly improves the diagnostic accuracy in childhood asthma.
Collapse
Affiliation(s)
- Qiuyan Yang
- Third Affiliated Hospital of Zhengzhou University, Department of Pediatrics, Zhengzhou, China; Henan Pediatric Clinical Research Center, Zhengzhou, China; Henan Key Laboratory of Child Brain Injury, Zhengzhou, China; Institute of Neuroscience of Zhengzhou University, Zhengzhou, China
| | - Yuehong Zheng
- Third Affiliated Hospital of Zhengzhou University, Department of Pediatrics, Zhengzhou, China
| | - Junjun Dong
- Third Affiliated Hospital of Zhengzhou University, Department of Pediatrics, Zhengzhou, China
| | - Qingrong Xu
- Third Affiliated Hospital of Zhengzhou University, Department of Pediatrics, Zhengzhou, China
| | - Shufang Li
- Third Affiliated Hospital of Zhengzhou University, Department of Pediatrics, Zhengzhou, China
| | - Aijun Li
- Third Affiliated Hospital of Zhengzhou University, Department of Pediatrics, Zhengzhou, China
| | - Xiaoman Xiong
- Third Affiliated Hospital of Zhengzhou University, Department of Pediatrics, Zhengzhou, China
| | - Yanli Zhang
- Third Affiliated Hospital of Zhengzhou University, Department of Pediatrics, Zhengzhou, China; Henan Pediatric Clinical Research Center, Zhengzhou, China; Henan Key Laboratory of Child Brain Injury, Zhengzhou, China; Institute of Neuroscience of Zhengzhou University, Zhengzhou, China.
| |
Collapse
|
2
|
Duan S, Wang J, Lou X, Chen D, Shi P, Jiang H, Wang Z, Li W, Qian F. A novel anti-IL-33 antibody recognizes an epitope FVLHN of IL-33 and has a therapeutic effect on inflammatory diseases. Int Immunopharmacol 2023; 122:110578. [PMID: 37423158 DOI: 10.1016/j.intimp.2023.110578] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2023] [Revised: 06/19/2023] [Accepted: 06/24/2023] [Indexed: 07/11/2023]
Abstract
As a crucial member of the Interleukin-1 (IL-1) family, IL-33 plays an indispensable role in modulating inflammatory responses. Here, we developed an effective anti-human IL-33 monoclonal antibody (mAb) named 5H8. Importantly, we have identified an epitope (FVLHN) of IL-33 protein as a recognition sequence for 5H8, which plays an important role in mediating the biological activity of IL-33. We observed that 5H8 significantly suppressed IL-33-induced IL-6 expression in bone marrow cells and mast cells in a dose-dependent manner in vitro. Furthermore, 5H8 effectively relievedHDM-induced asthma and PR8-induced acute lung injury in vivo. These findings indicate that targeting the FVLHN epitope is critical for inhibiting IL-33 function. In addition, wedetected that the Tm value of 5H8 was 66.47℃ and the KD value was 173.0 pM, which reflected that 5H8 had good thermal stability and high affinity. Taken together, our data suggest that our newly developed 5H8 antibody has potential as a therapeutic antibody for treating inflammatory diseases.
Collapse
Affiliation(s)
- Shixin Duan
- Shanghai Frontiers Science Center for Drug Target Identification and Delivery, School of Pharmacy, Shanghai Jiao Tong University, Shanghai 200240, PR China; Engineering Research Center of Cell & Therapeutic Antibody, Ministry of Education, Pharm-X Center, School of Pharmacy, Shanghai Jiao Tong University, Shanghai 200240, PR China
| | - Jun Wang
- Shanghai Frontiers Science Center for Drug Target Identification and Delivery, School of Pharmacy, Shanghai Jiao Tong University, Shanghai 200240, PR China; Xiamen Innovax Biotech Co, Xiamen, Fujian 361005, PR China
| | - Xinyi Lou
- Shanghai Frontiers Science Center for Drug Target Identification and Delivery, School of Pharmacy, Shanghai Jiao Tong University, Shanghai 200240, PR China
| | - Dongxin Chen
- Shanghai Frontiers Science Center for Drug Target Identification and Delivery, School of Pharmacy, Shanghai Jiao Tong University, Shanghai 200240, PR China
| | - Peiyunfeng Shi
- Shanghai Frontiers Science Center for Drug Target Identification and Delivery, School of Pharmacy, Shanghai Jiao Tong University, Shanghai 200240, PR China
| | - Hongchao Jiang
- Shanghai Frontiers Science Center for Drug Target Identification and Delivery, School of Pharmacy, Shanghai Jiao Tong University, Shanghai 200240, PR China
| | - Zhiming Wang
- Shanghai Frontiers Science Center for Drug Target Identification and Delivery, School of Pharmacy, Shanghai Jiao Tong University, Shanghai 200240, PR China
| | - Wen Li
- Shanghai Frontiers Science Center for Drug Target Identification and Delivery, School of Pharmacy, Shanghai Jiao Tong University, Shanghai 200240, PR China
| | - Feng Qian
- Shanghai Frontiers Science Center for Drug Target Identification and Delivery, School of Pharmacy, Shanghai Jiao Tong University, Shanghai 200240, PR China; Engineering Research Center of Cell & Therapeutic Antibody, Ministry of Education, Pharm-X Center, School of Pharmacy, Shanghai Jiao Tong University, Shanghai 200240, PR China.
| |
Collapse
|
3
|
Cazzola M, Page CP, Matera MG, Rogliani P, Hanania NA. Revisiting asthma pharmacotherapy: where do we stand and where do we want to go? Eur Respir J 2023; 62:2300700. [PMID: 37474159 DOI: 10.1183/13993003.00700-2023] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2023] [Accepted: 06/25/2023] [Indexed: 07/22/2023]
Abstract
Several current guidelines/strategies outline a treatment approach to asthma, which primarily consider the goals of improving lung function and quality of life and reducing symptoms and exacerbations. They suggest a strategy of stepping up or down treatment, depending on the patient's overall current asthma symptom control and future risk of exacerbation. While this stepwise approach is undeniably practical for daily practice, it does not always address the underlying mechanisms of this heterogeneous disease. In the last decade, there have been attempts to improve the treatment of severe asthma, such as the addition of a long-acting antimuscarinic agent to the traditional inhaled corticosteroid/long-acting β2-agonist treatment and the introduction of therapies targeting key cytokines. However, despite such strategies several unmet needs in this population remain, motivating research to identify novel targets and develop improved therapeutic and/or preventative asthma treatments. Pending the availability of such therapies, it is essential to re-evaluate the current conventional "one-size-fits-all" approach to a more precise asthma management. Although challenging, identifying "treatable traits" that contribute to respiratory symptoms in individual patients with asthma may allow a more pragmatic approach to establish more personalised therapeutic goals.
Collapse
Affiliation(s)
- Mario Cazzola
- Unit of Respiratory Medicine, Department of Experimental Medicine, University of Rome "Tor Vergata", Rome, Italy
| | - Clive P Page
- Sackler Institute of Pulmonary Pharmacology, Institute of Pharmaceutical Science, King's College London, London, UK
| | - Maria Gabriella Matera
- Unit of Pharmacology, Department of Experimental Medicine, University of Campania "Luigi Vanvitelli", Naples, Italy
| | - Paola Rogliani
- Unit of Respiratory Medicine, Department of Experimental Medicine, University of Rome "Tor Vergata", Rome, Italy
| | - Nicola A Hanania
- Section of Pulmonary and Critical Care Medicine, Baylor College of Medicine, Houston, TX, USA
| |
Collapse
|
4
|
Zhu S, Zhong S, Cheng K, Zhang LS, Bai JW, Cao Z, Wang S, Chen W, Cheng S, Ma L, Ling Z, Huang Y, Gu W, Sun X, Yi C, Zhao M, Liang S, Xu JF, Sun B, Zhang Y. Vitamin B6 regulates IL-33 homeostasis to alleviate type 2 inflammation. Cell Mol Immunol 2023; 20:794-807. [PMID: 37217797 PMCID: PMC10310729 DOI: 10.1038/s41423-023-01029-6] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2022] [Accepted: 04/18/2023] [Indexed: 05/24/2023] Open
Abstract
Interleukin-33 (IL-33) is a crucial nuclear cytokine that induces the type 2 immune response and maintains immune homeostasis. The fine-tuned regulation of IL-33 in tissue cells is critical to control of the type 2 immune response in airway inflammation, but the mechanism is still unclear. Here, we found that healthy individuals had higher phosphate-pyridoxal (PLP, an active form of vitamin B6) concentrations in the serum than asthma patients. Lower serum PLP concentrations in asthma patients were strongly associated with worse lung function and inflammation. In a mouse model of lung inflammation, we revealed that PLP alleviated the type 2 immune response and that this inhibitory effect relied on the activity of IL-33. A mechanistic study showed that in vivo, pyridoxal (PL) needed to be converted into PLP, which inhibited the type 2 response by regulating IL-33 stability. In mice heterozygous for pyridoxal kinase (PDXK), the conversion of PL to PLP was limited, and IL-33 levels were increased in the lungs, aggravating type 2 inflammation. Furthermore, we found that the mouse double minute 2 homolog (MDM2) protein, an E3 ubiquitin-protein ligase, could ubiquitinate the N-terminus of IL-33 and sustain IL-33 stability in epithelial cells. PLP reduced MDM2-mediated IL-33 polyubiquitination and decreased the level of IL-33 through the proteasome pathway. In addition, inhalation of PLP alleviated asthma-related effects in mouse models. In summary, our data indicate that vitamin B6 regulates MDM2-mediated IL-33 stability to constrain the type 2 response, which might help develop a potential preventive and therapeutic agent for allergy-related diseases.
Collapse
Affiliation(s)
- Songling Zhu
- Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, 230022, China
| | - Shufen Zhong
- State Key Laboratory of Cell Biology, CAS Center for Excellence in Molecular Cell Science, Shanghai Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, University of Chinese Academy of Sciences, 320 Yueyang Road, Shanghai, 200031, China
| | - Kebin Cheng
- Department of Respiratory and Critical Care Medicine, Shanghai Pulmonary Hospital, Institute of Respiratory Medicine, School of Medicine, Tongji University, 507 Zhengmin Road, Shanghai, 200433, China
| | - Li-Sha Zhang
- Department of Respiratory and Critical Care Medicine, Shanghai Pulmonary Hospital, Institute of Respiratory Medicine, School of Medicine, Tongji University, 507 Zhengmin Road, Shanghai, 200433, China
| | - Jiu-Wu Bai
- Department of Respiratory and Critical Care Medicine, Shanghai Pulmonary Hospital, Institute of Respiratory Medicine, School of Medicine, Tongji University, 507 Zhengmin Road, Shanghai, 200433, China
| | - Zu Cao
- Department of Respiratory and Critical Care Medicine, Shanghai Pulmonary Hospital, Institute of Respiratory Medicine, School of Medicine, Tongji University, 507 Zhengmin Road, Shanghai, 200433, China
| | - Su Wang
- Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, 230022, China
| | - Wen Chen
- State Key Laboratory of Cell Biology, CAS Center for Excellence in Molecular Cell Science, Shanghai Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, University of Chinese Academy of Sciences, 320 Yueyang Road, Shanghai, 200031, China
| | - Shipeng Cheng
- State Key Laboratory of Cell Biology, CAS Center for Excellence in Molecular Cell Science, Shanghai Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, University of Chinese Academy of Sciences, 320 Yueyang Road, Shanghai, 200031, China
| | - Liyan Ma
- State Key Laboratory of Cell Biology, CAS Center for Excellence in Molecular Cell Science, Shanghai Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, University of Chinese Academy of Sciences, 320 Yueyang Road, Shanghai, 200031, China
| | - Zhiyang Ling
- State Key Laboratory of Cell Biology, CAS Center for Excellence in Molecular Cell Science, Shanghai Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, University of Chinese Academy of Sciences, 320 Yueyang Road, Shanghai, 200031, China
| | - Yuying Huang
- State Key Laboratory of Cell Biology, CAS Center for Excellence in Molecular Cell Science, Shanghai Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, University of Chinese Academy of Sciences, 320 Yueyang Road, Shanghai, 200031, China
| | - Wangpeng Gu
- Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, 230022, China
| | - Xiaoyu Sun
- State Key Laboratory of Cell Biology, CAS Center for Excellence in Molecular Cell Science, Shanghai Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, University of Chinese Academy of Sciences, 320 Yueyang Road, Shanghai, 200031, China
| | - Chunyan Yi
- State Key Laboratory of Cell Biology, CAS Center for Excellence in Molecular Cell Science, Shanghai Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, University of Chinese Academy of Sciences, 320 Yueyang Road, Shanghai, 200031, China
| | - Meng Zhao
- State Key Laboratory of Cell Biology, CAS Center for Excellence in Molecular Cell Science, Shanghai Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, University of Chinese Academy of Sciences, 320 Yueyang Road, Shanghai, 200031, China
| | - Shuo Liang
- Department of Respiratory and Critical Care Medicine, Shanghai Pulmonary Hospital, Institute of Respiratory Medicine, School of Medicine, Tongji University, 507 Zhengmin Road, Shanghai, 200433, China.
| | - Jin-Fu Xu
- Department of Respiratory and Critical Care Medicine, Shanghai Pulmonary Hospital, Institute of Respiratory Medicine, School of Medicine, Tongji University, 507 Zhengmin Road, Shanghai, 200433, China.
| | - Bing Sun
- Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, 230022, China.
- State Key Laboratory of Cell Biology, CAS Center for Excellence in Molecular Cell Science, Shanghai Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, University of Chinese Academy of Sciences, 320 Yueyang Road, Shanghai, 200031, China.
| | - Yaguang Zhang
- State Key Laboratory of Cell Biology, CAS Center for Excellence in Molecular Cell Science, Shanghai Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, University of Chinese Academy of Sciences, 320 Yueyang Road, Shanghai, 200031, China.
| |
Collapse
|
5
|
Jrad AIS, Trad M, Bzeih W, El Hasbani G, Uthman I. Role of pro-inflammatory interleukins in osteoarthritis: a narrative review. Connect Tissue Res 2022; 64:238-247. [PMID: 36541851 DOI: 10.1080/03008207.2022.2157270] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
PURPOSE This manuscript will summarize the role of pro-inflammatory cytokines and tackle newly discussed ones within the scope of OA pathogenesis as mentioned in the recent literature. This will allow for a better understanding of the mechanisms behind such a complicated disease. MATERIAL AND METHODS Relevant articles were obtained by searching key terms including "pro-inflammatory cytokines," "inflammation," "pathophysiology," "cartilage damage," and "OA" in PubMed and Google Scholar databases. The year ranges set for the selection of the articles was between 2015 -2021. Inclusion criteria was based on the relevance and contribution to the field of the study. RESULTS Osteoarthritis (OA) has a complex multifactorial pathophysiology which is attributed to molecular and biomechanical changes that disrupt the normal balance of synthesis and degradation of articular cartilage and subchondral bone. Pro-inflammatory cytokines, with their wide range of action and intricate signaling pathways, are the constant subject of new discoveries revolving around this inflammatory disease. The available literature indicates that some of these cytokines such as IL-33, IL-17, IL-6, and IL-22 have a direct relation to cartilage degradation, while others like IL-15, IL-1, IL-7, and IL-34 have an indirect one. CONCLUSIONS Inflammation has an essential role in the manifestation of osteoarthritis clinical events. Specifically, certain cytokines exhibit pro-inflammatory properties that are markedly activated during the course of the disease and notably alter the homeostasis of the joint environment. However, clinical trials and observational studies remain insufficient to navigate the varying nature of this disease in humans.
Collapse
Affiliation(s)
| | - Maha Trad
- Faculty of Medical Sciences, Lebanese University, Hadath, Lebanon
| | - Wafaa Bzeih
- Faculty of Medical Sciences, Lebanese University, Hadath, Lebanon
| | - Georges El Hasbani
- Department of Internal Medicine, St. Vincent's Medical Center, Bridgeport, CT, USA
| | - Imad Uthman
- Department of Internal Medicine, American University of Beirut Medical Center, Beirut, Lebanon
| |
Collapse
|
6
|
Muacevic A, Adler JR, Tahlawi R, Aljabri HA. Immunotherapy in the Treatment of Allergic Rhinitis in Children. Cureus 2022; 14:e32464. [PMID: 36644088 PMCID: PMC9834958 DOI: 10.7759/cureus.32464] [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] [Accepted: 12/13/2022] [Indexed: 12/14/2022] Open
Abstract
Allergic rhinitis (AR) is an inflammation of the nasal membranes characterized by multiple allergic symptoms. It is a widespread health problem that affects patients' ability to engage in social and physical activity, which lowers their quality of life. The pathophysiology of AR is complex and requires sensitization and the development of a specific immune response to the allergen. Allergen-specific immunotherapy (AIT) is a therapeutic method that induces specific immune tolerance to allergens. The objectives of this review are to demonstrate the mechanism of action of immunotherapy, explain how it alleviates clinical symptoms of allergic rhinitis, list the indications and contraindications of immunotherapy in the treatment of allergic rhinitis, and identify different modalities of allergen immunotherapy, their disease-modifying effects, as well as their potential risks and benefits. The review of the literature highlights that T-cell and B-cell responses to inhaled allergens are altered by AIT, which decreases both early and late reactions to allergen exposure. To induce clinical and immunologic tolerance, especially in the pediatric age, escalating dosages of the causing allergen are administered subcutaneously or sublingually. AIT is indicated for severe persistent AR when avoidance measures and medications are inadequate to control the symptoms. To conclude, AIT is a disease-modifying therapy that is safe and effective for the treatment of allergic rhinitis. It is indicated when the symptoms are uncontrolled or when there are undesirable effects from pharmacotherapy.
Collapse
|
7
|
Farne H, Glanville N, Johnson N, Kebadze T, Aniscenko J, Regis E, Zhu J, Trujillo-Torralbo MB, Kon OM, Mallia P, Prevost AT, Edwards MR, Johnston SL, Singanayagam A, Jackson DJ. Effect of CRTH2 antagonism on the response to experimental rhinovirus infection in asthma: a pilot randomised controlled trial. Thorax 2022; 77:950-959. [PMID: 34716281 PMCID: PMC9510426 DOI: 10.1136/thoraxjnl-2021-217429] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2021] [Accepted: 09/24/2021] [Indexed: 11/03/2022]
Abstract
BACKGROUND AND AIMS The chemoattractant receptor-homologous molecule expressed on T helper type 2 cells (CRTH2) antagonist timapiprant improved lung function and asthma control in a phase 2 study, with evidence suggesting reduced exacerbations. We aimed to assess whether timapiprant attenuated or prevented asthma exacerbations induced by experimental rhinovirus (RV) infection. We furthermore hypothesised that timapiprant would dampen RV-induced type 2 inflammation and consequently improve antiviral immune responses. METHODS Atopic patients with partially controlled asthma on maintenance inhaled corticosteroids were randomised to timapiprant (n=22) or placebo (n=22) and challenged with RV-A16 3 weeks later. The primary endpoint was the cumulative lower respiratory symptom score over the 14 days post infection. Upper respiratory symptoms, spirometry, airway hyperresponsiveness, exhaled nitric oxide, RV-A16 virus load and soluble mediators in upper and lower airways samples, and CRTH2 staining in bronchial biopsies were additionally assessed before and during RV-A16 infection. RESULTS Six subjects discontinued the study and eight were not infected; outcomes were assessed in 16 timapiprant-treated and 14 placebo-treated, successfully infected subjects. There were no differences between treatment groups in clinical exacerbation severity including cumulative lower respiratory symptom score day 0-14 (difference 3.0 (95% CI -29.0 to 17.0), p=0.78), virus load, antiviral immune responses, or RV-A16-induced airway inflammation other than in the bronchial biopsies, where CRTH2 staining was increased during RV-A16 infection in the placebo-treated but not the timapiprant-treated group. Timapiprant had a favourable safety profile, with no deaths, serious adverse events or drug-related withdrawals. CONCLUSION Timapiprant treatment had little impact on the clinicopathological changes induced by RV-A16 infection in partially controlled asthma.
Collapse
Affiliation(s)
- Hugo Farne
- National Heart and Lung Institute, Imperial College London, London, UK
| | | | - Nicholas Johnson
- Imperial Clinical Trials Unit, Imperial College London, London, UK
| | - Tata Kebadze
- National Heart and Lung Institute, Imperial College London, London, UK
| | - Julia Aniscenko
- National Heart and Lung Institute, Imperial College London, London, UK
| | - Eteri Regis
- National Heart and Lung Institute, Imperial College London, London, UK
| | - Jie Zhu
- National Heart and Lung Institute, Imperial College London, London, UK
| | | | - Onn Min Kon
- National Heart and Lung Institute, Imperial College London, London, UK
| | - Patrick Mallia
- National Heart and Lung Institute, Imperial College London, London, UK
| | - A Toby Prevost
- Imperial Clinical Trials Unit, Imperial College London, London, UK
| | - Michael R Edwards
- National Heart and Lung Institute, Imperial College London, London, UK
| | | | - Aran Singanayagam
- National Heart and Lung Institute, Imperial College London, London, UK
| | - David J Jackson
- Guy’s Severe Asthma Centre, Guy’s and St Thomas’ NHS Foundation Trust, London, UK,MRC & Asthma UK Centre in Allergic Mechanisms of Asthma, King’s College London, London, UK
| |
Collapse
|
8
|
Xu C, Du L, Chen F, Tang K, Tang L, Shi J, Xiao L, Zeng Z, Liang Y, Guo Y. Increased expression of IL1-RL1 is associated with type 2 and type 1 immune pathways in asthma. BMC Immunol 2022; 23:23. [PMID: 35578178 PMCID: PMC9112580 DOI: 10.1186/s12865-022-00499-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2021] [Accepted: 05/10/2022] [Indexed: 11/10/2022] Open
Abstract
Background Asthma is a common chronic airway disease in the world. The purpose of this study was to explore the expression of IL1-RL1 in sputum and its correlation with Th1 and Th2 cytokines in asthma. Methods We recruited 132 subjects, detected IL1-RL1 protein level in sputum supernatant by ELISA, and analyzed the correlation between the expression level of IL1-RL1 and fraction of exhaled nitric oxide (FeNO), IgE, peripheral blood eosinophil count (EOS#), and Th2 cytokines (IL-4, IL-5, IL-10, IL-13, IL-33 and TSLP) and Th1 cytokines (IFN-γ, IL-2, IL-8). The diagnostic value of IL1-RL1 was evaluated by ROC curve. The expression of IL1-RL1 was further confirmed by BEAS-2B cell in vitro. Results Compared with the healthy control group, the expression of IL1-RL1 in sputum supernatant, sputum cells and serum of patients with asthma increased. The AUC of ROC curve of IL1-RL1 in sputum supernatant and serum were 0.6840 (p = 0.0034), and 0.7009 (p = 0.0233), respectively. IL1-RL1 was positively correlated with FeNO, IgE, EOS#, Th2 cytokines (IL-4, IL-5, IL-10, IL-13, IL-33 and TSLP) and Th1 cytokines (IFN-γ, IL-2, IL-8) in induced sputum supernatant. Four weeks after inhaled glucocorticoids (ICS) treatment, the expression of IL1-RL1 in sputum supernatant and serum was increased. In vitro, the expression of IL1-RL1 in BEAS-2B was increased after stimulated by IL-4 or IL-13 for 24 h. Conclusion The expression of IL1-RL1 in sputum supernatant, sputum cells and serum of patients with asthma was increased, and was positively correlated with some inflammatory markers in patients with asthma. IL1-RL1 may be used as a potential biomarker for the diagnosis and treatment of asthma. Supplementary Information The online version contains supplementary material available at 10.1186/s12865-022-00499-z.
Collapse
Affiliation(s)
- Changyi Xu
- Department of Pulmonary and Critical Care Medicine, The First Affiliated Hospital of Sun Yat-Sen University, No. 58 Zhongshan 2nd Road, Guangzhou, 510080, Guangdong, China.,Institute of Respiratory Diseases of Sun Yat-Sen University, No. 58 Zhongshan 2nd Road, Guangzhou, 510080, Guangdong, China
| | - Lijuan Du
- Department of Pulmonary and Critical Care Medicine, The First Affiliated Hospital of Sun Yat-Sen University, No. 58 Zhongshan 2nd Road, Guangzhou, 510080, Guangdong, China.,Institute of Respiratory Diseases of Sun Yat-Sen University, No. 58 Zhongshan 2nd Road, Guangzhou, 510080, Guangdong, China
| | - Fengjia Chen
- Department of Pulmonary and Critical Care Medicine, The First Affiliated Hospital of Sun Yat-Sen University, No. 58 Zhongshan 2nd Road, Guangzhou, 510080, Guangdong, China.,Institute of Respiratory Diseases of Sun Yat-Sen University, No. 58 Zhongshan 2nd Road, Guangzhou, 510080, Guangdong, China
| | - Kun Tang
- Department of Pulmonary and Critical Care Medicine, The First Affiliated Hospital of Sun Yat-Sen University, No. 58 Zhongshan 2nd Road, Guangzhou, 510080, Guangdong, China.,Institute of Respiratory Diseases of Sun Yat-Sen University, No. 58 Zhongshan 2nd Road, Guangzhou, 510080, Guangdong, China
| | - Lu Tang
- Department of Pulmonary and Critical Care Medicine, The First Affiliated Hospital of Sun Yat-Sen University, No. 58 Zhongshan 2nd Road, Guangzhou, 510080, Guangdong, China.,Institute of Respiratory Diseases of Sun Yat-Sen University, No. 58 Zhongshan 2nd Road, Guangzhou, 510080, Guangdong, China
| | - Jia Shi
- Department of Pulmonary and Critical Care Medicine, The First Affiliated Hospital of Sun Yat-Sen University, No. 58 Zhongshan 2nd Road, Guangzhou, 510080, Guangdong, China.,Institute of Respiratory Diseases of Sun Yat-Sen University, No. 58 Zhongshan 2nd Road, Guangzhou, 510080, Guangdong, China
| | - Lisha Xiao
- Department of Pulmonary and Critical Care Medicine, The First Affiliated Hospital of Sun Yat-Sen University, No. 58 Zhongshan 2nd Road, Guangzhou, 510080, Guangdong, China.,Institute of Respiratory Diseases of Sun Yat-Sen University, No. 58 Zhongshan 2nd Road, Guangzhou, 510080, Guangdong, China
| | - Zhimin Zeng
- Department of Pulmonary and Critical Care Medicine, The First Affiliated Hospital of Sun Yat-Sen University, No. 58 Zhongshan 2nd Road, Guangzhou, 510080, Guangdong, China. .,Institute of Respiratory Diseases of Sun Yat-Sen University, No. 58 Zhongshan 2nd Road, Guangzhou, 510080, Guangdong, China.
| | - Yuxia Liang
- Department of Pulmonary and Critical Care Medicine, The First Affiliated Hospital of Sun Yat-Sen University, No. 58 Zhongshan 2nd Road, Guangzhou, 510080, Guangdong, China. .,Institute of Respiratory Diseases of Sun Yat-Sen University, No. 58 Zhongshan 2nd Road, Guangzhou, 510080, Guangdong, China.
| | - Yubiao Guo
- Department of Pulmonary and Critical Care Medicine, The First Affiliated Hospital of Sun Yat-Sen University, No. 58 Zhongshan 2nd Road, Guangzhou, 510080, Guangdong, China. .,Institute of Respiratory Diseases of Sun Yat-Sen University, No. 58 Zhongshan 2nd Road, Guangzhou, 510080, Guangdong, China.
| |
Collapse
|
9
|
General Rehabilitation Program after Knee or Hip Replacement Significantly Influences Erythrocytes Oxidative Stress Markers and Serum ST2 Levels. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2022; 2022:1358858. [PMID: 35401921 PMCID: PMC8986427 DOI: 10.1155/2022/1358858] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 12/31/2021] [Revised: 02/12/2022] [Accepted: 03/14/2022] [Indexed: 12/12/2022]
Abstract
The survival of erythrocytes in the circulating blood depends on their membranes' structural and functional integrity. One of the mechanisms that may underlie the process of joint degeneration is the imbalance of prooxidants and antioxidants, promoting cellular oxidative stress. The study is aimed at observing the effects of the 21-day general rehabilitation program on the erythrocytes redox status and serum ST2 marker in patients after knee or hip replacement in the course of osteoarthritis. Erythrocytes and serum samples were collected from 36 patients. We analyzed the selected markers of the antioxidant system in the erythrocytes: catalase (CAT), glutathione reductase (glutathione disulfide reductase (GR, GSR)), total superoxide dismutase activity (SOD), glutathione peroxidase (GPx), glutathione transferase (GST) activity, and cholesterol and lipofuscin (LPS) concentration. In serum, we analyzed the concentration of the suppression of tumorigenicity 2 (ST2) marker. After the 21-day general rehabilitation program, the total SOD and GPx activity, measured in the hemolysates, significantly increased (p < 0.001) while LPS, cholesterol, and ST2 levels in serum significantly decreased (p < 0.001). General rehabilitation reduces oxidative stress in patients after knee or hip replacement in the course of osteoarthritis. Individually designed, regular physical activity is the essential element of the postoperative protocol, which improves the redox balance helping patients recover after the s4urgery effectively.
Collapse
|
10
|
Reciprocal regulation of IL-33 receptor-mediated inflammatory response and pulmonary fibrosis by TRAF6 and USP38. Proc Natl Acad Sci U S A 2022; 119:e2116279119. [PMID: 35238669 PMCID: PMC8917384 DOI: 10.1073/pnas.2116279119] [Citation(s) in RCA: 35] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Abstract
Significance IL-33R mediates local inflammatory responses and plays crucial roles in the pathogenesis of immune diseases. In this study, we identified USP38, which negatively regulates IL-33-triggered signaling by mediating K27-linked deubiquitination of IL-33R at K511 and its autophagic degradation. USP38 deficiency aggravates IL-33-induced lung inflammatory response and bleomycin-induced pulmonary fibrosis. We further show that the E3 ubiquitin ligase TRAF6 catalyzes K27-linked polyubiquitination of IL-33R at K511, and that deficiency of TRAF6 inhibits IL-33-mediated signaling. Our findings reveal an important mechanism regarding how IL-33R is precisely regulated to ensure its inactivation in rest cells and proper activation following IL-33 stimulation.
Collapse
|
11
|
Ham J, Shin JW, Ko BC, Kim HY. Targeting the Epithelium-Derived Innate Cytokines: From Bench to Bedside. Immune Netw 2022; 22:e11. [PMID: 35291657 PMCID: PMC8901708 DOI: 10.4110/in.2022.22.e11] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2021] [Revised: 02/15/2022] [Accepted: 02/15/2022] [Indexed: 02/05/2023] Open
Abstract
When epithelial cells are exposed to potentially threatening external stimuli such as allergens, bacteria, viruses, and helminths, they instantly produce "alarmin" cytokines, namely, IL-33, IL-25, and TSLP. These alarmins alert the immune system about these threats, thereby mobilizing host immune defense mechanisms. Specifically, the alarmins strongly stimulate type-2 immune cells, including eosinophils, mast cells, dendritic cells, type-2 helper T cells, and type-2 innate lymphoid cells. Given that the alarm-raising role of IL-33, IL-25, and TSLP was first detected in allergic and infectious diseases, most studies on alarmins focus on their role in these diseases. However, recent studies suggest that alarmins also have a broad range of effector functions in other pathological conditions, including psoriasis, multiple sclerosis, and cancer. Therefore, this review provides an update on the epithelium-derived cytokines in both allergic and non-allergic diseases. We also review the progress of clinical trials on biological agents that target the alarmins and discuss the therapeutic potential of these agents in non-allergic diseases.
Collapse
Affiliation(s)
- Jongho Ham
- Laboratory of Mucosal Immunology, Department of Biomedical Sciences, Seoul National University College of Medicine, Seoul 03080, Korea.,Department of Biomedical Sciences, BK21 Plus Biomedical Science Project, Seoul National University College of Medicine, Seoul 03080, Korea.,CIRNO, Sungkyunkwan University, Suwon 16419, Korea
| | - Jae Woo Shin
- Laboratory of Mucosal Immunology, Department of Biomedical Sciences, Seoul National University College of Medicine, Seoul 03080, Korea.,CIRNO, Sungkyunkwan University, Suwon 16419, Korea.,Institute of Allergy and Clinical Immunology, Seoul National University Medical Research Center, Seoul 03080, Korea
| | - Byeong Cheol Ko
- Laboratory of Mucosal Immunology, Department of Biomedical Sciences, Seoul National University College of Medicine, Seoul 03080, Korea.,Department of Medicine, Seoul National University College of Medicine, Seoul 03080, Korea
| | - Hye Young Kim
- Laboratory of Mucosal Immunology, Department of Biomedical Sciences, Seoul National University College of Medicine, Seoul 03080, Korea.,Department of Biomedical Sciences, BK21 Plus Biomedical Science Project, Seoul National University College of Medicine, Seoul 03080, Korea.,CIRNO, Sungkyunkwan University, Suwon 16419, Korea.,Institute of Allergy and Clinical Immunology, Seoul National University Medical Research Center, Seoul 03080, Korea
| |
Collapse
|
12
|
Peluzzo AM, Autieri MV. Challenging the Paradigm: Anti-Inflammatory Interleukins and Angiogenesis. Cells 2022; 11:cells11030587. [PMID: 35159396 PMCID: PMC8834461 DOI: 10.3390/cells11030587] [Citation(s) in RCA: 33] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2021] [Revised: 01/31/2022] [Accepted: 02/04/2022] [Indexed: 02/01/2023] Open
Abstract
Angiogenesis is a vital biological process, and neovascularization is essential for the development, wound repair, and perfusion of ischemic tissue. Neovascularization and inflammation are independent biological processes that are linked in response to injury and ischemia. While clear that pro-inflammatory factors drive angiogenesis, the role of anti-inflammatory interleukins in angiogenesis remains less defined. An interleukin with anti-inflammatory yet pro-angiogenic effects would hold great promise as a therapeutic modality to treat many disease states where inflammation needs to be limited, but revascularization and reperfusion still need to be supported. As immune modulators, interleukins can polarize macrophages to a pro-angiogenic and reparative phenotype, which indirectly influences angiogenesis. Interleukins could also potentially directly induce angiogenesis by binding and activating its receptor on endothelial cells. Although a great deal of attention is given to the negative effects of pro-inflammatory interleukins, less is described concerning the potential protective effects of anti-inflammatory interleukins on various disease processes. To focus this review, we will consider IL-4, IL-10, IL-13, IL-19, and IL-33 to be anti-inflammatory interleukins, all of which have recognized immunomodulatory effects. This review will summarize current research concerning anti-inflammatory interleukins as potential drivers of direct and indirect angiogenesis, emphasizing their role in future therapeutics.
Collapse
|
13
|
Mai TT, Nguyen PG, Le MT, Tran TD, Huynh PNH, Trinh DTT, Nguyen QT, Thai KM. Discovery of small molecular inhibitors for interleukin-33/ST2 protein-protein interaction: a virtual screening, molecular dynamics simulations and binding free energy calculations. Mol Divers 2022; 26:2659-2678. [PMID: 35031934 PMCID: PMC8760117 DOI: 10.1007/s11030-021-10359-4] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2021] [Accepted: 11/26/2021] [Indexed: 01/13/2023]
Abstract
The interleukin-1 receptor like ST2 has emerged as a potential drug discovery target since it was identified as the receptor of the novel cytokine IL-33, which is involved in many inflammatory and autoimmune diseases. For the treatment of such IL-33-related disorders, efforts have been made to discover molecules that can inhibit the protein–protein interactions (PPIs) between IL-33 and ST2, but to date no drug has been approved. Although several anti-ST2 antibodies have entered clinical trials, the exploration of small molecular inhibitors is highly sought-after because of its advantages in terms of oral bioavailability and manufacturing cost. The aim of this study was to discover ST2 receptor inhibitors based on its PPIs with IL-33 in crystal structure (PDB ID: 4KC3) using virtual screening tools with pharmacophore modeling and molecular docking. From an enormous chemical space ZINC, a potential series of compounds has been discovered with stronger binding affinities than the control compound from a previous study. Among them, four compounds strongly interacted with the key residues of the receptor and had a binding free energy < − 20 kcal/mol. By intensive calculations using data from molecular dynamics simulations, ZINC59514725 was identified as the most potential candidate for ST2 receptor inhibitor in this study.
Collapse
Affiliation(s)
- Tan Thanh Mai
- Department of Medicinal Chemistry, Faculty of Pharmacy, University of Medicine and Pharmacy at Ho Chi Minh City, Ho Chi Minh City, 700000, Vietnam
| | - Phuc Gia Nguyen
- Department of Medicinal Chemistry, Faculty of Pharmacy, University of Medicine and Pharmacy at Ho Chi Minh City, Ho Chi Minh City, 700000, Vietnam
| | - Minh-Tri Le
- Department of Medicinal Chemistry, Faculty of Pharmacy, University of Medicine and Pharmacy at Ho Chi Minh City, Ho Chi Minh City, 700000, Vietnam.,School of Medicine, Vietnam National University Ho Chi Minh City, Ho Chi Minh City, 700000, Vietnam
| | - Thanh-Dao Tran
- Department of Medicinal Chemistry, Faculty of Pharmacy, University of Medicine and Pharmacy at Ho Chi Minh City, Ho Chi Minh City, 700000, Vietnam
| | - Phuong Nguyen Hoai Huynh
- Department of Medicinal Chemistry, Faculty of Pharmacy, University of Medicine and Pharmacy at Ho Chi Minh City, Ho Chi Minh City, 700000, Vietnam
| | - Dieu-Thuong Thi Trinh
- Faculty of Traditional Medicine, University of Medicine and Pharmacy at Ho Chi Minh City, Ho Chi Minh City, 700000, Vietnam
| | - Quoc-Thai Nguyen
- Department of Biochemistry, Faculty of Pharmacy, University of Medicine and Pharmacy at Ho Chi Minh City, Ho Chi Minh City, 700000, Vietnam.
| | - Khac-Minh Thai
- Department of Medicinal Chemistry, Faculty of Pharmacy, University of Medicine and Pharmacy at Ho Chi Minh City, Ho Chi Minh City, 700000, Vietnam.
| |
Collapse
|
14
|
Jovanovic MZ, Geller DA, Gajovic NM, Jurisevic MM, Arsenijevic NN, Jovanovic MM, Supic GM, Vojvodic DV, Jovanovic IP. Dual blockage of PD-L/PD-1 and IL33/ST2 axes slows tumor growth and improves antitumor immunity by boosting NK cells. Life Sci 2022; 289:120214. [PMID: 34890591 DOI: 10.1016/j.lfs.2021.120214] [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: 09/17/2021] [Revised: 11/11/2021] [Accepted: 12/01/2021] [Indexed: 12/09/2022]
Abstract
AIMS Although separate blockage of either IL33/ST2 or PD-L/PD-1 axes has been shown to be beneficial in many tumors, co-blockage of IL33/ST2 and PD-L/PD-1 hasn't been studied yet. MAIN METHODS 4T1 breast cancer and CT26 colon cancer were inducted in BALB/C wild type (WT) and BALB/C ST2 knockout mice, after which mice underwent anti PD-1 and anti IL-33 treatment. KEY FINDINGS Co-blockage of IL33/ST2 and PD-L/PD-1 delayed tumor appearance and slowed tumor growth. Enhanced NK cell cytotoxicity against 4T1 tumor cells in ST2 knockout anti-PD-1 treated mice was associated with overexpression of miRNA-150 and miRNA-155, upregulation of NFκB and STAT3, increased expression of activation markers and decreased expression of immunosuppressive markers in splenic and primary tumor derived NK cells. NK cells from ST2 knockout anti-PD-1 treated mice tend to proliferate more and are less prone to apoptosis. Accumulation of immunosuppressive myeloid derived suppressor cells and regulatory T cells was significantly impaired in spleen and primary tumor of ST2 knockout anti-PD-1 treated mice. SIGNIFICANCE Co-blockage of IL3/ST2 and PD-L/PD-1 axes impedes tumor progression more efficiently than single blockage of either axes, thus offering potential new approach to immunotherapy of tumors.
Collapse
Affiliation(s)
- Marina Z Jovanovic
- Center for Molecular Medicine and Stem Cell Research, Faculty of Medical Sciences, University of Kragujevac, Svetozara Markovica 69, 34000 Kragujevac, Serbia
| | - David A Geller
- Department of Surgery, University of Pittsburgh, 3459 Fifth Avenue, UPMC Montefiore, 7 South Pittsburgh, PA 15213 2582, USA.
| | - Nevena M Gajovic
- Center for Molecular Medicine and Stem Cell Research, Faculty of Medical Sciences, University of Kragujevac, Svetozara Markovica 69, 34000 Kragujevac, Serbia
| | - Milena M Jurisevic
- Department of Pharmacy, Faculty of Medical Sciences, University of Kragujevac, SvetozaraMarkovica 69, 34000 Kragujevac, Serbia.
| | - Nebojsa N Arsenijevic
- Center for Molecular Medicine and Stem Cell Research, Faculty of Medical Sciences, University of Kragujevac, Svetozara Markovica 69, 34000 Kragujevac, Serbia
| | - Milan M Jovanovic
- Department of Abdominal Surgery, Military Medical Academy, Crnotravska 17, 11000 Belgrade, Serbia.
| | - Gordana M Supic
- Institute for Medical Research, Military Medical Academy, Crnotravska 17, 11000 Belgrade, Serbia; Medical Faculty of Military Medical Academy, University of Defense, Crnotravska 17, 11000 Belgrade, Serbia.
| | - Danilo V Vojvodic
- Institute for Medical Research, Military Medical Academy, Crnotravska 17, 11000 Belgrade, Serbia; Medical Faculty of Military Medical Academy, University of Defense, Crnotravska 17, 11000 Belgrade, Serbia.
| | - Ivan P Jovanovic
- Center for Molecular Medicine and Stem Cell Research, Faculty of Medical Sciences, University of Kragujevac, Svetozara Markovica 69, 34000 Kragujevac, Serbia.
| |
Collapse
|
15
|
Straus DB, Pryor D, Haque TT, Kee SA, Dailey JM, Jackson KG, Barnstein BO, Ryan JJ. IL-33 priming amplifies ATP-mediated mast cell cytokine production. Cell Immunol 2022; 371:104470. [PMID: 34942481 DOI: 10.1016/j.cellimm.2021.104470] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2021] [Revised: 11/22/2021] [Accepted: 12/06/2021] [Indexed: 12/28/2022]
Abstract
Inflammatory responses are required to block pathogen infection but can also lead to hypersensitivity and chronic inflammation. Barrier tissues actively release IL-33, ATP, and other alarmins during cell stress, helping identify pathogenic stimuli. However, it is unclear how these signals are integrated. Mast cells are critical initiators of allergic inflammation and respond to IL-33 and ATP. We found that mouse mast cells had a 3-6-fold increase in ATP-induced cytokine production when pre-treated with IL-33. This effect was observed at ATP concentrations < 100 µM and required < 30-minute IL-33 exposure. ATP-induced degranulation was not enhanced by pretreatment nor was the response to several pathogen molecules. Mechanistic studies implicated the P2X7 receptor and calcineurin/NFAT pathway in the enhanced ATP response. Finally, we found that IL-33 + ATP co-stimulation enhanced peritoneal eosinophil and macrophage recruitment. These results support the hypothesis that alarmins collaborate to surpass a threshold necessary to initiate an inflammatory response.
Collapse
Affiliation(s)
- David B Straus
- Department of Biology, Virginia Commonwealth University, Richmond, VA 23284, USA.
| | - Destiny Pryor
- Department of Microbiology and Immunology, Virginia Commonwealth University, Richmond, VA 23298, USA
| | - Tamara T Haque
- Department of Microbiology and Immunology, Virginia Commonwealth University, Richmond, VA 23298, USA
| | - Sydney A Kee
- Department of Biology, Virginia Commonwealth University, Richmond, VA 23284, USA
| | - Jordan M Dailey
- Department of Microbiology and Immunology, Virginia Commonwealth University, Richmond, VA 23298, USA
| | - Kaitlyn G Jackson
- Department of Microbiology and Immunology, Virginia Commonwealth University, Richmond, VA 23298, USA
| | - Brian O Barnstein
- Department of Biology, Virginia Commonwealth University, Richmond, VA 23284, USA
| | - John J Ryan
- Department of Biology, Virginia Commonwealth University, Richmond, VA 23284, USA
| |
Collapse
|
16
|
Turck D, Bohn T, Castenmiller J, De Henauw S, Hirsch‐Ernst KI, Maciuk A, Mangelsdorf I, McArdle HJ, Naska A, Pelaez C, Pentieva K, Siani A, Thies F, Tsabouri S, Vinceti M, Cubadda F, Frenzel T, Heinonen M, Marchelli R, Neuhäuser‐Berthold M, Poulsen M, Prieto Maradona M, Schlatter JR, van Loveren H, Goumperis T, Knutsen HK. Safety of frozen and dried formulations from whole house crickets (Acheta domesticus) as a Novel food pursuant to Regulation (EU) 2015/2283. EFSA J 2021; 19:e06779. [PMID: 34429777 PMCID: PMC8369844 DOI: 10.2903/j.efsa.2021.6779] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022] Open
Abstract
Following a request from the European Commission, the EFSA Panel on Nutrition, Novel Food and Food Allergens (NDA) was asked to deliver an opinion on the safety of frozen and dried formulations from house crickets (Acheta domesticus) as a novel food pursuant to Regulation (EU) 2015/2283. The NF is proposed in three formulations: (i) frozen, (ii) dried, (iii) ground. The main components of the NF are protein, fat and fibre (chitin) in the dried form of the NF, and water, protein, fat and fibre (chitin) in the frozen form of the NF. The Panel notes that the concentrations of contaminants in the NF depend on the occurrence levels of these substances in the insect feed. The Panel further notes that there are no safety concerns regarding the stability of the NF if the NF complies with the proposed specification limits during its entire shelf-life. The NF has a high-protein content, although the true protein levels in the NF are overestimated when using the nitrogen-to-protein conversion factor of 6.25, due to the presence of non-protein nitrogen from chitin. The applicant proposed to use the NF in the form of a snack, and as a food ingredient in a number of food products. The target population proposed by the applicant is the general population. The Panel notes that, considering the composition of the NF and the proposed conditions of use, the consumption of the NF is not nutritionally disadvantageous. The Panel notes that no genotoxicity and no subchronic toxicity studies with the NF were provided by the applicant. Considering that no safety concerns arise from the history of use of A. domesticus or from the compositional data of the NF, the Panel identified no other safety concerns than allergenicity. The Panel considers that the consumption of the NF might trigger primary sensitisation to A. domesticus proteins and may cause allergic reactions in subjects allergic to crustaceans, mites and molluscs. Additionally, allergens from the feed may end up in the NF. The Panel concludes that the NF is safe under the proposed uses and use levels.
Collapse
|
17
|
Turck D, Castenmiller J, De Henauw S, Hirsch‐Ernst KI, Kearney J, Maciuk A, Mangelsdorf I, McArdle HJ, Naska A, Pelaez C, Pentieva K, Siani A, Thies F, Tsabouri S, Vinceti M, Cubadda F, Frenzel T, Heinonen M, Marchelli R, Neuhäuser‐Berthold M, Poulsen M, Maradona MP, Schlatter JR, van Loveren H, Azzollini D, Knutsen HK. Safety of frozen and dried formulations from migratory locust ( Locusta migratoria) as a Novel food pursuant to Regulation (EU) 2015/2283. EFSA J 2021; 19:e06667. [PMID: 34249158 PMCID: PMC8251647 DOI: 10.2903/j.efsa.2021.6667] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Following a request from the European Commission, the EFSA Panel on Nutrition, Novel Food and Food Allergens (NDA) was asked to deliver an opinion on the safety of frozen and dried formulations from migratory locust (Locusta migratoria) as a novel food pursuant to Regulation (EU) 2015/2283. The term migratory locust refers to the adult of the insect species Locusta migratoria. The NF is proposed in three formulations i) frozen without legs and wings; ii) dried without legs and wings; iii) ground with legs and wings. The main components of the NF are protein, fat and fibre (chitin) in the dried form of the NF, and water, protein, fat and fibre (chitin) in the frozen form of the NF. The Panel notes that the concentration of contaminants in the NF depends on the occurrence levels of these substances in the insect feed. The Panel notes that there are no safety concerns regarding the stability of the NF if the NF complies with the proposed specification limits during its entire shelf-life. The NF has a high protein content, although the true protein levels in the NF are overestimated when using the nitrogen-to-protein conversion factor of 6.25, due to the presence of non-protein nitrogen from chitin. The applicant proposed to use the NF as frozen, dried and ground in the form of snack, and as a food ingredient in a number of food products. The target population proposed by the applicant is the general population. The Panel notes that considering the composition of the NF and the proposed conditions of use, the consumption of the NF is not nutritionally disadvantageous. The submitted history of use and toxicity studies from literature did not raise safety concerns. The Panel considers that the consumption of the NF might trigger primary sensitisation to L. migratoria proteins and may cause allergic reactions in subjects with allergy to crustaceans, mites and molluscs. Additionally, allergens from the feed may end up in the NF. The Panel concludes that the NF is safe under the proposed uses and use levels.
Collapse
|
18
|
Gubernatorova EO, Namakanova OA, Gorshkova EA, Medvedovskaya AD, Nedospasov SA, Drutskaya MS. Novel Anti-Cytokine Strategies for Prevention and Treatment of Respiratory Allergic Diseases. Front Immunol 2021; 12:601842. [PMID: 34084159 PMCID: PMC8167041 DOI: 10.3389/fimmu.2021.601842] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2020] [Accepted: 04/26/2021] [Indexed: 12/12/2022] Open
Abstract
Asthma is a heterogeneous inflammatory disease characterized by airflow obstruction, wheezing, eosinophilia and neutrophilia of the airways. Identification of distinct inflammatory patterns characterizing asthma endotypes led to the development of novel therapeutic approaches. Cytokine or cytokine receptor targeting by therapeutic antibodies, such as anti-IL-4 and anti-IL-5, is now approved for severe asthma treatment. However, the complexity of cytokine networks in asthma should not be underestimated. Inhibition of one pro-inflammatory cytokine may lead to perturbed expression of another pro-inflammatory cytokine. Without understanding of the underlying mechanisms and defining the molecular predictors it may be difficult to control cytokine release that accompanies certain disease manifestations. Accumulating evidence suggests that in some cases a combined pharmacological inhibition of pathogenic cytokines, such as simultaneous blockade of IL-4 and IL-13 signaling, or blockade of upstream cytokines, such as TSLP, are more effective than single cytokine targeting. IL-6 and TNF are the important inflammatory mediators in the pathogenesis of asthma. Preliminary data suggests that combined pharmacological inhibition of TNF and IL-6 during asthma may be more efficient as compared to individual neutralization of these cytokines. Here we summarize recent findings in the field of anti-cytokine therapy of asthma and discuss immunological mechanisms by which simultaneous targeting of multiple cytokines as opposed to targeting of a single cytokine may improve disease outcomes.
Collapse
Affiliation(s)
- Ekaterina O Gubernatorova
- Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, Moscow, Russia.,Lomonosov Moscow State University, Moscow, Russia
| | - Olga A Namakanova
- Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, Moscow, Russia.,Lomonosov Moscow State University, Moscow, Russia
| | - Ekaterina A Gorshkova
- Lomonosov Moscow State University, Moscow, Russia.,Center for Precision Genome Editing and Genetic Technologies for Biomedicine, Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, Moscow, Russia
| | - Alexandra D Medvedovskaya
- Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, Moscow, Russia.,Lomonosov Moscow State University, Moscow, Russia
| | - Sergei A Nedospasov
- Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, Moscow, Russia.,Lomonosov Moscow State University, Moscow, Russia.,Center for Precision Genome Editing and Genetic Technologies for Biomedicine, Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, Moscow, Russia.,Department of Immunobiology and Biomedicine, Sirius University of Science and Technology, Sochi, Russia
| | - Marina S Drutskaya
- Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, Moscow, Russia.,Center for Precision Genome Editing and Genetic Technologies for Biomedicine, Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, Moscow, Russia
| |
Collapse
|
19
|
Rodriguez-Rodriguez N, Gogoi M, McKenzie AN. Group 2 Innate Lymphoid Cells: Team Players in Regulating Asthma. Annu Rev Immunol 2021; 39:167-198. [PMID: 33534604 PMCID: PMC7614118 DOI: 10.1146/annurev-immunol-110119-091711] [Citation(s) in RCA: 49] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2023]
Abstract
Type 2 immunity helps protect the host from infection, but it also plays key roles in tissue homeostasis, metabolism, and repair. Unfortunately, inappropriate type 2 immune reactions may lead to allergy and asthma. Group 2 innate lymphoid cells (ILC2s) in the lungs respond rapidly to local environmental cues, such as the release of epithelium-derived type 2 initiator cytokines/alarmins, producing type 2 effector cytokines such as IL-4, IL-5, and IL-13 in response to tissue damage and infection. ILC2s are associated with the severity of allergic asthma, and experimental models of lung inflammation have shown how they act as playmakers, receiving signals variously from stromal and immune cells as well as the nervous system and then distributing cytokine cues to elicit type 2 immune effector functions and potentiate CD4+ T helper cell activation, both of which characterize the pathology of allergic asthma. Recent breakthroughs identifying stromal- and neuronal-derived microenvironmental cues that regulate ILC2s, along with studies recognizing the potential plasticity of ILC2s, have improved our understanding of the immunoregulation of asthma and opened new avenues for drug discovery.
Collapse
Affiliation(s)
- Noe Rodriguez-Rodriguez
- Medical Research Council, Laboratory of Molecular Biology, Cambridge, Cambridgeshire, CB2 0QH. UK
| | - Mayuri Gogoi
- Medical Research Council, Laboratory of Molecular Biology, Cambridge, Cambridgeshire, CB2 0QH. UK
| | - Andrew N.J. McKenzie
- Medical Research Council, Laboratory of Molecular Biology, Cambridge, Cambridgeshire, CB2 0QH. UK,Corresponding author:
| |
Collapse
|
20
|
Scott LM, Vincent EE, Hudson N, Neal C, Jones N, Lavelle EC, Campbell M, Halestrap AP, Dick AD, Theodoropoulou S. Interleukin-33 regulates metabolic reprogramming of the retinal pigment epithelium in response to immune stressors. JCI Insight 2021; 6:129429. [PMID: 33884963 PMCID: PMC8119202 DOI: 10.1172/jci.insight.129429] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2019] [Accepted: 03/17/2021] [Indexed: 11/17/2022] Open
Abstract
It remains unresolved how retinal pigment epithelial cell metabolism is regulated following immune activation to maintain retinal homeostasis and retinal function. We exposed retinal pigment epithelium (RPE) to several stress signals, particularly Toll-like receptor stimulation, and uncovered an ability of RPE to adapt their metabolic preference on aerobic glycolysis or oxidative glucose metabolism in response to different immune stimuli. We have identified interleukin-33 (IL-33) as a key metabolic checkpoint that antagonizes the Warburg effect to ensure the functional stability of the RPE. The identification of IL-33 as a key regulator of mitochondrial metabolism suggests roles for the cytokine that go beyond its extracellular “alarmin” activities. IL-33 exerts control over mitochondrial respiration in RPE by facilitating oxidative pyruvate catabolism. We have also revealed that in the absence of IL-33, mitochondrial function declined and resultant bioenergetic switching was aligned with altered mitochondrial morphology. Our data not only shed new light on the molecular pathway of activation of mitochondrial respiration in RPE in response to immune stressors but also uncover a potentially novel role of nuclear intrinsic IL-33 as a metabolic checkpoint regulator.
Collapse
Affiliation(s)
- Louis M Scott
- Academic Unit of Ophthalmology, Translational Health Sciences, Bristol Medical School
| | - Emma E Vincent
- School of Cellular and Molecular Medicine, and.,Medical Research Council Integrative Epidemiology Unit, Population Health Sciences, Bristol Medical School, University of Bristol, Bristol, United Kingdom
| | - Natalie Hudson
- Smurfit Institute of Genetics, School of Genetics and Microbiology, Trinity College Dublin, Dublin, Ireland
| | - Chris Neal
- Wolfson Bioimaging Facility, University of Bristol, Bristol, United Kingdom
| | - Nicholas Jones
- Institute of Life Science, Swansea University Medical School, Swansea, United Kingdom
| | - Ed C Lavelle
- Adjuvant Research Group, School of Biochemistry and Immunology, and Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin, Ireland
| | - Matthew Campbell
- Smurfit Institute of Genetics, School of Genetics and Microbiology, Trinity College Dublin, Dublin, Ireland
| | - Andrew P Halestrap
- Department of Biochemistry, Bristol Medical School, University of Bristol, Bristol, United Kingdom
| | - Andrew D Dick
- Academic Unit of Ophthalmology, Translational Health Sciences, Bristol Medical School.,School of Cellular and Molecular Medicine, and.,UCL Institute of Ophthalmology, University College London, London, United Kingdom
| | - Sofia Theodoropoulou
- Academic Unit of Ophthalmology, Translational Health Sciences, Bristol Medical School
| |
Collapse
|
21
|
Passanisi S, Caminiti L, Zirilli G, Lombardo F, Crisafulli G, Aversa T, Pajno GB. Biologics in food allergy: up-to-date. Expert Opin Biol Ther 2021; 21:1227-1235. [PMID: 33733975 DOI: 10.1080/14712598.2021.1904888] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Introduction: In recent years, the advent of immunotherapy has remarkably improved the management of IgE-mediated food allergy. However, some barriers still exist. Therefore, the effort of researchers aims to investigate new perspectives in the field of non-allergen specific therapy, also based on the current knowledge of the pathogenesis of this disease.Areas covered: This review aims to focus on the role of biologics as a treatment option in patients with IgE-mediated food allergy. These agents are characterized by their ability to inactivate the Th2 pro-inflammatory pathways. Biologics can be used both alone and in association with immunotherapy. Monoclonal antibodies targeting IgE, the IL-4/IL-13 axis, IL-5, and alarmins have been proposed and investigated for treating food allergy.Expert opinion: The clinical efficacy and safety of biologics have been demonstrated in several preclinical studies and randomized controlled trials. Future studies are still required to address current unmet needs, including the identification of the optimal dose to be used by ensuring the effectiveness of therapy.
Collapse
Affiliation(s)
- Stefano Passanisi
- Department of Human Pathology in Adult and Developmental Age "Gaetano Barresi, University of Messin, Messina (Italy)
| | - Lucia Caminiti
- Department of Human Pathology in Adult and Developmental Age "Gaetano Barresi, University of Messin, Messina (Italy)
| | - Giuseppina Zirilli
- Department of Human Pathology in Adult and Developmental Age "Gaetano Barresi, University of Messin, Messina (Italy)
| | - Fortunato Lombardo
- Department of Human Pathology in Adult and Developmental Age "Gaetano Barresi, University of Messin, Messina (Italy)
| | - Giuseppe Crisafulli
- Department of Human Pathology in Adult and Developmental Age "Gaetano Barresi, University of Messin, Messina (Italy)
| | - Tommaso Aversa
- Department of Human Pathology in Adult and Developmental Age "Gaetano Barresi, University of Messin, Messina (Italy)
| | - Giovanni B Pajno
- Department of Human Pathology in Adult and Developmental Age "Gaetano Barresi, University of Messin, Messina (Italy)
| |
Collapse
|
22
|
Jovanovic IP, Pejnovic NN, Radosavljevic GD, Arsenijevic NN, Lukic ML. IL-33/ST2 axis in innate and acquired immunity to tumors. Oncoimmunology 2021; 1:229-231. [PMID: 22720252 PMCID: PMC3376988 DOI: 10.4161/onci.1.2.18131] [Citation(s) in RCA: 41] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023] Open
Abstract
Interleukin-33, a ligand for ST2/T1, has an important role in allergy, autoimmunity and inflammation. The role of IL-33/ST2 axis in cancer is not elucidated. Using metastatic breast cancer model we provide evidence that lack of ST2 signaling led to reduced tumor growth and metastasis and enhanced anti-tumor immunity.
Collapse
Affiliation(s)
- Ivan P Jovanovic
- Center for Molecular Medicine and Stem Cell Research; Faculty of Medicine; University of Kragujevac; Kragulevac, Serbia
| | | | | | | | | |
Collapse
|
23
|
Mattei F, Andreone S, Marone G, Gambardella AR, Loffredo S, Varricchi G, Schiavoni G. Eosinophils in the Tumor Microenvironment. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2021; 1273:1-28. [PMID: 33119873 DOI: 10.1007/978-3-030-49270-0_1] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Eosinophils are rare blood-circulating and tissue-infiltrating immune cells studied for decades in the context of allergic diseases and parasitic infections. Eosinophils can secrete a wide array of soluble mediators and effector molecules, with potential immunoregulatory activities in the tumor microenvironment (TME). These findings imply that these cells may play a role in cancer immunity. Despite these cells were known to infiltrate tumors since many years ago, their role in TME is gaining attention only recently. In this chapter, we will review the main biological functions of eosinophils that can be relevant within the TME. We will discuss how these cells may undergo phenotypic changes acquiring pro- or antitumoricidal properties according to the surrounding stimuli. Moreover, we will analyze canonical (i.e., degranulation) and unconventional mechanisms (i.e., DNA traps, exosome secretion) employed by eosinophils in inflammatory contexts, which can be relevant for tumor immune responses. Finally, we will review the available preclinical models that could be employed for the study of the role in vivo of eosinophils in cancer.
Collapse
Affiliation(s)
- Fabrizio Mattei
- Department of Oncology and Molecular Medicine, Istituto Superiore di Sanità, Rome, Italy
| | - Sara Andreone
- Department of Oncology and Molecular Medicine, Istituto Superiore di Sanità, Rome, Italy
| | - Giancarlo Marone
- Department of Public Health, University of Naples Federico II, Naples, Italy.,Azienda Ospedaliera Ospedali dei Colli - Monaldi Hospital Pharmacy, Naples, Italy
| | | | - Stefania Loffredo
- Department of Translational Medical Sciences and Center for Basic and Clinical Immunology Research (CISI), University of Naples Federico II, Naples, Italy.,WAO Center of Excellence, Naples, Italy.,Institute of Experimental Endocrinology and Oncology "G. Salvatore" (IEOS), National Research Council (CNR), Naples, Italy
| | - Gilda Varricchi
- Department of Translational Medical Sciences and Center for Basic and Clinical Immunology Research (CISI), University of Naples Federico II, Naples, Italy. .,WAO Center of Excellence, Naples, Italy. .,Institute of Experimental Endocrinology and Oncology "G. Salvatore" (IEOS), National Research Council (CNR), Naples, Italy.
| | - Giovanna Schiavoni
- Department of Oncology and Molecular Medicine, Istituto Superiore di Sanità, Rome, Italy.
| |
Collapse
|
24
|
Turck D, Castenmiller J, De Henauw S, Hirsch‐Ernst KI, Kearney J, Maciuk A, Mangelsdorf I, McArdle HJ, Naska A, Pelaez C, Pentieva K, Siani A, Thies F, Tsabouri S, Vinceti M, Cubadda F, Frenzel T, Heinonen M, Marchelli R, Neuhäuser‐Berthold M, Poulsen M, Prieto Maradona M, Schlatter JR, van Loveren H, Ververis E, Knutsen HK. Safety of dried yellow mealworm ( Tenebrio molitor larva) as a novel food pursuant to Regulation (EU) 2015/2283. EFSA J 2021; 19:e06343. [PMID: 33488808 PMCID: PMC7805300 DOI: 10.2903/j.efsa.2021.6343] [Citation(s) in RCA: 85] [Impact Index Per Article: 21.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022] Open
Abstract
Following a request from the European Commission, the EFSA Panel on Nutrition, Novel Foods and Food Allergens (NDA) was asked to deliver an opinion on dried yellow mealworm (Tenebrio molitor larva) as a novel food (NF) pursuant to Regulation (EU) 2015/2283. The term yellow mealworm refers to the larval form of the insect species Tenebrio molitor. The NF is the thermally dried yellow mealworm, either as whole dried insect or in the form of powder. The main components of the NF are protein, fat and fibre (chitin). The Panel notes that the levels of contaminants in the NF depend on the occurrence levels of these substances in the insect feed. The Panel notes that there are no safety concerns regarding the stability of the NF if the NF complies with the proposed specification limits during its entire shelf life. The NF has a high protein content, although the true protein levels in the NF are overestimated when using the nitrogen-to-protein conversion factor of 6.25, due to the presence of non-protein nitrogen from chitin. The applicant proposed to use the NF as whole, dried insect in the form of snacks, and as a food ingredient in a number of food products. The target population proposed by the applicant is the general population. The Panel notes that considering the composition of the NF and the proposed conditions of use, the consumption of the NF is not nutritionally disadvantageous. The submitted toxicity studies from the literature did not raise safety concerns. The Panel considers that the consumption of the NF may induce primary sensitisation and allergic reactions to yellow mealworm proteins and may cause allergic reactions in subjects with allergy to crustaceans and dust mites. Additionally, allergens from the feed may end up in the NF. The Panel concludes that the NF is safe under the proposed uses and use levels.
Collapse
|
25
|
Chen Z, Hu Q, Huo Y, Zhang R, Fu Q, Qin X. Serum Interleukin-33 is a Novel Predictive Biomarker of Hemorrhage Transformation and Outcome in Acute Ischemic Stroke. J Stroke Cerebrovasc Dis 2020; 30:105506. [PMID: 33307292 DOI: 10.1016/j.jstrokecerebrovasdis.2020.105506] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2020] [Revised: 11/21/2020] [Accepted: 11/23/2020] [Indexed: 01/20/2023] Open
Abstract
INTRODUCTION Hemorrhage Transformation (HT) in acute ischemic stroke (AIS) depends on multiple factors. Some studies have shown that serum interleukin-33 (IL-33) is of central significance as a neuroprotective factor. However, the relationship between serum IL-33 and HT in AIS has not been evaluated. OBJECTIVE To investigate the relationship between serum IL-33 concentration and HT in AIS. METHODS We recruited 151 consecutive non-thrombolytic patients with AIS clinically diagnosed in The First Affiliated Hospital of Chongqing Medical University from December 2018 to October 2019. If the patients showed radiographic presentation of HT within two weeks following admission, they were assigned to the HT group; others were assigned to the non-HT group. There were 40 healthy control subjects recruited during the same period. Serum IL-33 concentration was detected by ELISA and the independent risk value of HT in AIS was predicted by multivariate logistic regression. The accuracy was analyzed by receiver operating characteristic (ROC) curves. In three months after admission, the functional outcome was measured by modified Rankin scale (mRS). RESULTS ROC curve showed that the area under the curve (AUC) of serum IL-33 was 0.739 (95% CI: 0.657-0.821, P < .001) in predicting HT in AIS. When serum IL-33 concentration was ≤ 67.66 ng/L, the sensitivity and specificity of the prediction were 81.3% and 63%, respectively. Multivariate logistic regression analysis showed that serum IL-33 concentration ≤ 67.66 ng/L was an independent predictor of HT in AIS (OR = 5.773, 95% CI: 1.685-19.792, P = .005). The follow-up results of mRS showed a higher probability of an unfavorable outcome in those with HT compared to those without HT (OR = 6.520, 95% CI: 2.530-16.803, P < .001). CONCLUSIONS HT in AIS is negatively correlated with outcome. Furthermore, serum IL-33 is an independent predictive biomarker of HT and outcome in AIS.
Collapse
Affiliation(s)
- Zhenlei Chen
- Department of Neurology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China and Chongqing Key Laboratory of Neurobiology, No. 1, Youyi Road, Yuanjiagang, Yuzhong District, Chongqing, China.
| | - Qingzhe Hu
- Department of Neurology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China and Chongqing Key Laboratory of Neurobiology, No. 1, Youyi Road, Yuanjiagang, Yuzhong District, Chongqing, China.
| | - Yingchao Huo
- Department of Neurology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China and Chongqing Key Laboratory of Neurobiology, No. 1, Youyi Road, Yuanjiagang, Yuzhong District, Chongqing, China.
| | - Rongrong Zhang
- Department of Neurology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China and Chongqing Key Laboratory of Neurobiology, No. 1, Youyi Road, Yuanjiagang, Yuzhong District, Chongqing, China.
| | - Qing Fu
- Department of Neurology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China and Chongqing Key Laboratory of Neurobiology, No. 1, Youyi Road, Yuanjiagang, Yuzhong District, Chongqing, China.
| | - Xinyue Qin
- Department of Neurology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China and Chongqing Key Laboratory of Neurobiology, No. 1, Youyi Road, Yuanjiagang, Yuzhong District, Chongqing, China.
| |
Collapse
|
26
|
Le MT, Mai TT, Huynh PNH, Tran TD, Thai KM, Nguyen QT. Structure-based discovery of interleukin-33 inhibitors: a pharmacophore modelling, molecular docking, and molecular dynamics simulation approach. SAR AND QSAR IN ENVIRONMENTAL RESEARCH 2020; 31:883-904. [PMID: 33191795 DOI: 10.1080/1062936x.2020.1837239] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/20/2020] [Accepted: 10/12/2020] [Indexed: 06/11/2023]
Abstract
Interleukin (IL)-33 is a new cytokine of the IL-1 family that is related to several inflammatory and autoimmune diseases. IL-33 binds to its ST2 receptor and leads to biological responses thereof. Currently, no drugs have been approved for the treatment of IL-33 related diseases. The aim of this study was to search for small molecules that inhibit the protein-protein interaction between IL-33 and ST2. A virtual screening was first performed to identify potential molecules that can bind IL-33. By analysing the interactions between key residues in the complex of IL-33/ST2, two pharmacophore hypotheses were then generated based on the 'mimicry' and 'pair-rule' principles. From a database of 62,074 compounds, 60 molecules satisfying the pharmacophore models were identified and docked to IL-33. Among 35 compounds successfully docked into the protein, 9 potential ligands in complex with IL-33 were selected for further analysis by molecular dynamics simulations. Based on the stability of the complexes and the interactions of each ligand with the key residues of IL-33, two compounds DB00158 and DB00642 were identified as the most potential inhibitors that can be further investigated as promising novel IL-33 inhibitory drugs.
Collapse
Affiliation(s)
- M-T Le
- School of Medicine, Vietnam National University Ho Chi Minh City , Ho Chi Minh City, Vietnam
- Faculty of Pharmacy, University of Medicine and Pharmacy at Ho Chi Minh City , Ho Chi Minh City, Vietnam
| | - T T Mai
- Faculty of Pharmacy, University of Medicine and Pharmacy at Ho Chi Minh City , Ho Chi Minh City, Vietnam
- Saigon Pharmaceutical Science and Technology Center - Sapharcen, University of Medicine and Pharmacy at Ho Chi Minh City , Ho Chi Minh City, Vietnam
| | - P N H Huynh
- Faculty of Pharmacy, University of Medicine and Pharmacy at Ho Chi Minh City , Ho Chi Minh City, Vietnam
- Saigon Pharmaceutical Science and Technology Center - Sapharcen, University of Medicine and Pharmacy at Ho Chi Minh City , Ho Chi Minh City, Vietnam
| | - T-D Tran
- Faculty of Pharmacy, University of Medicine and Pharmacy at Ho Chi Minh City , Ho Chi Minh City, Vietnam
| | - K-M Thai
- Faculty of Pharmacy, University of Medicine and Pharmacy at Ho Chi Minh City , Ho Chi Minh City, Vietnam
- Saigon Pharmaceutical Science and Technology Center - Sapharcen, University of Medicine and Pharmacy at Ho Chi Minh City , Ho Chi Minh City, Vietnam
| | - Q-T Nguyen
- Faculty of Pharmacy, University of Medicine and Pharmacy at Ho Chi Minh City , Ho Chi Minh City, Vietnam
- Saigon Pharmaceutical Science and Technology Center - Sapharcen, University of Medicine and Pharmacy at Ho Chi Minh City , Ho Chi Minh City, Vietnam
| |
Collapse
|
27
|
Kianian F, Karimian SM, Kadkhodaee M, Takzaree N, Seifi B, Sadeghipour HR. Protective effects of ascorbic acid and calcitriol combination on airway remodelling in ovalbumin-induced chronic asthma. PHARMACEUTICAL BIOLOGY 2020; 58:107-115. [PMID: 31942829 PMCID: PMC7006747 DOI: 10.1080/13880209.2019.1710218] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/29/2019] [Revised: 11/12/2019] [Accepted: 12/23/2019] [Indexed: 05/03/2023]
Abstract
Context: Airway remodelling is one of the most refractory problems in asthma. According to the critical roles of oxidative stress and inflammation in airway remodelling, it is supposed that ascorbic acid and calcitriol have beneficial effects. However, a combination of antioxidants may be more effective for asthma therapy.Objective: This study investigated the protective effects of ascorbic acid in combination with calcitriol on airway remodelling in ovalbumin (OVA)-induced chronic asthma.Materials and methods: BALB/c mice were assigned into seven groups: (1) Control; (2) Asthma; (3) Ineffective C (orally 39 mg/kg ascorbic acid); (4) Ineffective D (intraperitoneally 1.5 μg/kg calcitriol); (5) Effective C (orally 130 mg/kg ascorbic acid); (6) Effective D (intraperitoneally 5 μg/kg calcitriol); (7) Combination (orally 39 mg/kg ascorbic acid + intraperitoneally 1.5 μg/kg calcitriol). All animals were sensitized and challenged with OVA except in the control group (normal saline). In all treatment groups, mice were administrated vitamins 30 min before each challenge (three times per week for 8 consecutive weeks).Results: In comparison with the asthma group, co-administration of ineffective doses of ascorbic acid and calcitriol led to the decreased levels of IL-13 (50.5 ± 1.85 vs. 42.13 ± 0.37 pg/mL, p = 0.02) and IgE (58.74 ± 0.43 vs. 45.78 ± 2.05 ng/mL, p = 0.003) as well as the reduction of goblet hyperplasia and subepithelial fibrosis (5 vs. 1 score, p = 0.001 and 5 vs. 2 score, p = 0.001, respectively).Discussion and conclusions: Combination of ascorbic acid with calcitriol in ineffective doses improves airway remodelling due to additive effects possibly through reduction of oxidative stress and inflammation. This study provides a scientific basis for further research and clinical applications of ascorbic acid and calcitriol and can be generalized to the broader pharmacological studies.
Collapse
Affiliation(s)
- Farzaneh Kianian
- Department of Physiology, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Seyed Morteza Karimian
- Department of Physiology, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Mehri Kadkhodaee
- Department of Physiology, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Nasrin Takzaree
- Department of Anatomy and Histology, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Behjat Seifi
- Department of Physiology, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Hamid Reza Sadeghipour
- Department of Physiology, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
28
|
Blocking Interleukin-33 Alleviates the Joint Inflammation and Inhibits the Development of Collagen-Induced Arthritis in Mice. J Immunol Res 2020; 2020:4297354. [PMID: 33490289 PMCID: PMC7801941 DOI: 10.1155/2020/4297354] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2020] [Revised: 10/08/2020] [Accepted: 10/19/2020] [Indexed: 01/02/2023] Open
Abstract
Rheumatoid arthritis (RA) is considered a systemic chronic inflammatory joint disease characterized by chronic synovitis and cartilage and bone destruction. Interleukin-33 (IL-33) is a proinflammatory cytokine which is highly expressed in the synovium of RA patients and the joints of mice with collagen-induced arthritis (CIA) and exacerbates CIA in mice. However, the role of the IL-33-neutralizing antibody in the murine model of CIA remains unclear. In the present study, CIA mice were given intraperitoneally with polyclonal rabbit anti-murine IL-33 antibody (anti-IL-33) or normal rabbit IgG control after the first signs of arthritis. Administration of anti-IL-33 after the onset of disease significantly reduced the severity of CIA and joint damage compared with controls treated with normal rabbit IgG. Anti-IL-33 treatment also significantly decreased the serum levels of interferon-γ(IFN-γ),IL-6, IL-12, IL-33, and tumor necrosis factor-α (TNF-α). Moreover, anti-IL-33 treatment significantly downregulated the production of IFN-γ, IL-6, IL-12, IL-33, and TNF-α in ex vivo-stimulated spleen cells. Together, our results indicate that the IL-33-neutralizing antibody may provide a therapeutic strategy for RA by inhibiting the release of proinflammatory cytokines.
Collapse
|
29
|
Duez C, Gross B, Marquillies P, Ledroit V, Ryffel B, Glineur C. Regulation of IL (Interleukin)-33 Production in Endothelial Cells via Kinase Activation and Fas/CD95 Upregulation. Arterioscler Thromb Vasc Biol 2020; 40:2619-2631. [PMID: 32907372 DOI: 10.1161/atvbaha.120.314832] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
OBJECTIVE The occurrence of new blood vessel formation in the lungs of asthmatic patients suggests a critical role for airway endothelial cells (ECs) in the disease. IL-33 (Interleukin-33)-a cytokine abundantly expressed in human lung ECs-recently emerged as a key factor in the development of allergic diseases, including asthma. In the present study, we evaluated whether mouse and human ECs exposed to the common Dermatophagoides farinae allergen produce IL-33 and characterized the activated signaling pathways. Approach and Results: Mouse primary lung ECs were exposed in vitro to D farinae extract or rmIL-33 (recombinant murine IL-33). Both D farinae and rmIL-33 induced Il-33 transcription without increasing the IL-33 production and upregulated the expression of its receptor, as well as genes involved in angiogenesis and the regulation of immune responses. In particular, D farinae and rmIL-33 upregulated Fas/Cd95 transcript level, yet without promoting apoptosis. Inhibition of caspases involved in the Fas signaling pathway, increased IL-33 protein level in ECs, suggesting that Fas may decrease IL-33 level through caspase-8-dependent mechanisms. Our data also showed that the NF-κB (nuclear factor-κB), PI3K/Akt, and Wnt/β-catenin pathways regulate Il-33 transcription in both mouse and human primary ECs. CONCLUSIONS Herein, we described a new mechanism involved in the control of IL-33 production in lung ECs exposed to allergens.
Collapse
Affiliation(s)
- Catherine Duez
- CNRS UMR 9017, Inserm U1019, CIIL-Center for Infection and Immunity of Lille (C.D., P.M., V.L., C.G.), CHU Lille, Institut Pasteur de Lille, University Lille, France
| | - Barbara Gross
- Inserm U1011-EGID (B.G.), CHU Lille, Institut Pasteur de Lille, University Lille, France
| | - Philippe Marquillies
- CNRS UMR 9017, Inserm U1019, CIIL-Center for Infection and Immunity of Lille (C.D., P.M., V.L., C.G.), CHU Lille, Institut Pasteur de Lille, University Lille, France
| | - Valérie Ledroit
- CNRS UMR 9017, Inserm U1019, CIIL-Center for Infection and Immunity of Lille (C.D., P.M., V.L., C.G.), CHU Lille, Institut Pasteur de Lille, University Lille, France
| | - Bernhard Ryffel
- Laboratory of Molecular and Experimental Immunology and Neurogenetics, CNRS UMR 7355, University of Orleans, France (B.R.)
| | - Corine Glineur
- CNRS UMR 9017, Inserm U1019, CIIL-Center for Infection and Immunity of Lille (C.D., P.M., V.L., C.G.), CHU Lille, Institut Pasteur de Lille, University Lille, France
| |
Collapse
|
30
|
Respiratory syncytial virus upregulates IL-33 expression in mouse model of virus-induced inflammation exacerbation in OVA-sensitized mice and in asthmatic subjects. Cytokine 2020; 138:155349. [PMID: 33132030 DOI: 10.1016/j.cyto.2020.155349] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2020] [Revised: 10/13/2020] [Accepted: 10/14/2020] [Indexed: 12/26/2022]
Abstract
BACKGROUND Bronchial asthma (BA) is a chronic disease of the airways. The great majority of BA exacerbations are associated with respiratory viral infections. Recent findings point out a possible role of proinflammatory cytokine interleukin-33 (IL-33) in the development of atopic diseases. Although, little is known about the role of IL-33 in virus-induced BA exacerbations. METHODS We used mouse models of RSV (respiratory syncytial virus)-induced inflammation exacerbation in OVA-sensitized mice and RSV infection alone in adult animals to characterize expression of il33 in the mouse lungs. Moreover, we studied the influence of il33 knockdown with intranasally administrated siRNA on the development of RSV-induced inflammation exacerbation. In addition, we evaluated the expression of IL33 in the ex vivo stimulated PBMCs from allergic asthma patients and healthy subjects with and without confirmed acute respiratory viral infection. RESULTS Using mouse models, we found that infection with RSV drives enhanced il33 mRNA expression in the mouse lung. Treatment with anti-il33 siRNA diminishes airway inflammation in the lungs (we found a decrease in the number of inflammatory cells in the lungs and in the severity of histopathological alterations) of mice with RSV-induced inflammation exacerbation, but do not influence viral load. Elevated level of the IL33 mRNA was detected in ex vivo stimulated blood lymphocytes of allergic asthmatics infected with respiratory viruses. RSV and rhinovirus were the most detected viruses in volunteers with symptoms of respiratory infection. CONCLUSION The present study provides additional evidence of the crucial role of the IL-33 in pathogenesis of RSV infection and virus-induced allergic bronchial asthma exacerbations.
Collapse
|
31
|
He Z, Song Y, Yi Y, Qiu F, Wang J, Li J, Jin Q, Sacitharan PK. Blockade of IL-33 signalling attenuates osteoarthritis. Clin Transl Immunology 2020; 9:e1185. [PMID: 33133598 PMCID: PMC7587452 DOI: 10.1002/cti2.1187] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2020] [Revised: 09/09/2020] [Accepted: 09/09/2020] [Indexed: 12/20/2022] Open
Abstract
Objectives Osteoarthritis (OA) is the most common form of arthritis characterised by cartilage degradation, synovitis and pain. Disease modifying treatments for OA are not available. The critical unmet need is to find therapeutic targets to reduce both disease progression and pain. The cytokine IL‐33 and its receptor ST2 have been shown to play a role in immune and inflammatory diseases, but their role in osteoarthritis is unknown. Methods Non‐OA and OA human chondrocytes samples were examined for IL‐33 and ST2 expression. Novel inducible cartilage specific knockout mice (IL‐33Acan CreERT2) and inducible fibroblast‐like synoviocyte knockout mice (IL‐33Col1a2 CreERT2) were generated and subjected to an experimental OA model. In addition, wild‐type mice were intra‐articularly administered with either IL‐33‐ or ST2‐neutralising antibodies during experimental OA studies. Results IL‐33 and its receptor ST2 have increased expression in OA patients and a murine disease model. Administering recombinant IL‐33 increased OA and pain in vivo. Synovial fibroblast‐specific deletion of IL‐33 decreased synovitis but did not impact disease outcomes, whilst cartilage‐specific deletion of IL‐33 improved disease outcomes in vivo. Blocking IL‐33 signalling also reduced the release of cartilage‐degrading enzymes in human and mouse chondrocytes. Most importantly, we show the use of monoclonal antibodies against IL‐33 and ST2 attenuates both OA and pain in vivo. Conclusion Overall, our data reveal blockade of IL‐33 signalling as a viable therapeutic target for OA.
Collapse
Affiliation(s)
- Zengliang He
- Department of Orthopedics The Second Hospital of Nanjing The Affiliated Hospital of Nanjing University of Chinese Medicine Nanjing China
| | - Yan Song
- Department of Orthopedics The Second Hospital of Nanjing The Affiliated Hospital of Nanjing University of Chinese Medicine Nanjing China
| | - Yongxiang Yi
- Department of General Surgery The Second Hospital of Nanjing The Affiliated Hospital of Nanjing University of Chinese Medicine Nanjing China
| | - Fengzhuo Qiu
- Department of Neurology The Sir Run Run Hospital Nanjing Medical University Nanjing China
| | - Junhua Wang
- College of Veterinary Medicine Qingdao Agricultural University Qingdao China
| | - Junwei Li
- College of Veterinary Medicine Qingdao Agricultural University Qingdao China
| | - Qingwen Jin
- Department of Neurology The Sir Run Run Hospital Nanjing Medical University Nanjing China
| | - Pradeep Kumar Sacitharan
- The Institute of Ageing and Chronic Disease University of Liverpool Liverpool UK.,Department of Biological Sciences Xi'an Jiaotong-Liverpool University Suzhou Industrial Park Suzhou China
| |
Collapse
|
32
|
IL-33 induces type-2-cytokine phenotype but exacerbates cardiac remodeling post-myocardial infarction with eosinophil recruitment, worsened systolic dysfunction, and ventricular wall rupture. Clin Sci (Lond) 2020; 134:1191-1218. [PMID: 32432676 DOI: 10.1042/cs20200402] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2020] [Revised: 05/02/2020] [Accepted: 05/20/2020] [Indexed: 02/06/2023]
Abstract
Myocardial infarction (MI) is the leading cause of mortality worldwide. Interleukin (IL)-33 (IL-33) is a cytokine present in most cardiac cells and is secreted on necrosis where it acts as a functional ligand for the ST2 receptor. Although IL-33/ST2 axis is protective against various forms of cardiovascular diseases, some studies suggest potential detrimental roles for IL-33 signaling. The aim of the present study was to examine the effect of IL-33 administration on cardiac function post-MI in mice. MI was induced by coronary artery ligation. Mice were treated with IL-33 (1 μg/day) or vehicle for 4 and 7 days. Functional and molecular changes of the left ventricle (LV) were assessed. Single cell suspensions were obtained from bone marrow, heart, spleen, and peripheral blood to assess the immune cells using flow cytometry at 1, 3, and 7 days post-MI in IL-33 or vehicle-treated animals. The results of the present study suggest that IL-33 is effective in activating a type 2 cytokine milieu in the damaged heart, consistent with reduced early inflammatory and pro-fibrotic response. However, IL-33 administration was associated with worsened cardiac function and adverse cardiac remodeling in the MI mouse model. IL-33 administration increased infarct size, LV hypertrophy, cardiomyocyte death, and overall mortality rate due to cardiac rupture. Moreover, IL-33-treated MI mice displayed a significant myocardial eosinophil infiltration at 7 days post-MI when compared with vehicle-treated MI mice. The present study reveals that although IL-33 administration is associated with a reparative phenotype following MI, it worsens cardiac remodeling and promotes heart failure.
Collapse
|
33
|
Berman R, Kopf KW, Min E, Huang J, Downey GP, Alam R, Chu HW, Day BJ. IL-33/ST2 signaling modulates Afghanistan particulate matter induced airway hyperresponsiveness in mice. Toxicol Appl Pharmacol 2020; 404:115186. [PMID: 32777237 DOI: 10.1016/j.taap.2020.115186] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2020] [Revised: 08/03/2020] [Accepted: 08/05/2020] [Indexed: 12/17/2022]
Abstract
Increased symptoms of asthma-like respiratory illnesses have been reported in soldiers returning from tours of duty in Afghanistan. Inhalation of desert particulate matter (PM) may contribute to this deployment-related lung disease (DRLD), but little is known about disease mechanisms. The IL-33 signaling pathway, including its receptor ST2, has been implicated in the pathogenesis of lung diseases including asthma, but its role in PM-mediated airway dysfunction has not been studied. The goal of this study was to investigate whether IL-33/ST2 signaling contributes to airway dysfunction in preclinical models of lung exposure to Afghanistan PM (APM). Wild-type (WT) and ST2 knockout (KO) mice on the BALB/C background were oropharyngeally instilled with a single dose of saline or 50 μg of APM in saline. Airway hyperresponsiveness (AHR) and inflammation were assessed after 24 h. In WT mice, a single APM exposure induced AHR and neutrophilic inflammation. Unlike the WT mice, ST2 KO mice that lack the receptor for IL-33 did not demonstrate AHR although airway neutrophilic inflammation was comparable to the WT mice. Oropharyngeal delivery of a soluble ST2 decoy receptor in APM-exposed WT mice significantly blocked AHR. Additional data in mouse tracheal epithelial cell and lung macrophage cultures demonstrated a role of APM-induced IL-33/ST2 signaling in suppression of regulator of G protein signaling 2 (RGS2), a gene known to protect against bronchoconstriction. We present for the first time that APM may increase AHR, one of the features of asthma, in part through the IL-33/ST2/RGS2 pathway.
Collapse
Affiliation(s)
- Reena Berman
- Department of Medicine, Basic Science Section, National Jewish Health, Denver, CO, United States of America
| | - Katrina W Kopf
- Biological Resource Center, National Jewish Health, Denver, CO, United States of America
| | - Elysia Min
- Department of Medicine, Medicine Office of Research, National Jewish Health, Denver, CO, United States of America
| | - Jie Huang
- Department of Medicine, Medicine Office of Research, National Jewish Health, Denver, CO, United States of America
| | - Gregory P Downey
- Department of Medicine, Division of Pulmonary, Critical Care & Sleep Medicine, National Jewish Health, Denver, CO, United States of America
| | - Rafeul Alam
- Department of Medicine, Division of Allergy & Clinical Immunology, National Jewish Health, Denver, CO, United States of America
| | - Hong Wei Chu
- Department of Medicine, Basic Science Section, National Jewish Health, Denver, CO, United States of America.
| | - Brian J Day
- Department of Medicine, Medicine Office of Research, National Jewish Health, Denver, CO, United States of America.
| |
Collapse
|
34
|
Blocking of the IL-33/ST2 Signaling Axis by a Single-Chain Antibody Variable Fragment (scFv) Specific to IL-33 with a Defined Epitope. Int J Mol Sci 2020; 21:ijms21186953. [PMID: 32971846 PMCID: PMC7554688 DOI: 10.3390/ijms21186953] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2020] [Revised: 09/13/2020] [Accepted: 09/16/2020] [Indexed: 12/03/2022] Open
Abstract
Interleukin 33 (IL-33) is an IL-1 family cytokine that plays a central role in immune system by regulating and initiating inflammatory responses. The binding of IL-33 to the suppressor of tumorigenicity 2 (ST2) receptor induces mitogen-activated protein kinases (MAPK) and nuclear factor κB (NF-κB) pathways, thereby leading to inflammatory cytokines production in type 2 helper T cells and type 2 innate lymphoid cells. To develop an antibody specific to IL-33 with a defined epitope, we characterized a single-chain antibody variable fragments (scFvs) clone specific to IL-33, C2_2E12, which was selected from a human synthetic library of scFvs using phage display. Affinity (Kd) of C2_2E12 was determined to be 38 nM using enzyme-linked immunosorbent assay. C2_2E12 did not show cross-reactivity toward other interleukin cytokines, including closely related IL-1 family cytokines and unrelated proteins. Mutational scanning analysis revealed that the epitope of IL-33 consisted of residues 149–158 with key residues being L150 and K151 of IL-33. Structural modeling suggested that L150 and K151 residues are important for the interaction of IL-33 with C2_2E12, implicating that C2_2E12 could block the binding of ST2 to IL-33. Pull-down and in-cell assays supported that C2_2E12 can inhibit the IL-33/ST2 signaling axis. These results suggest that the scFv clone characterized here can function as a neutralizing antibody.
Collapse
|
35
|
Naclerio R, Ansotegui IJ, Bousquet J, Canonica GW, D'Amato G, Rosario N, Pawankar R, Peden D, Bergmann KC, Bielory L, Caraballo L, Cecchi L, Cepeda SAM, Chong Neto HJ, Galán C, Gonzalez Diaz SN, Idriss S, Popov T, Ramon GD, Ridolo E, Rottem M, Songnuan W, Rouadi P. International expert consensus on the management of allergic rhinitis (AR) aggravated by air pollutants: Impact of air pollution on patients with AR: Current knowledge and future strategies. World Allergy Organ J 2020; 13:100106. [PMID: 32256939 PMCID: PMC7132263 DOI: 10.1016/j.waojou.2020.100106] [Citation(s) in RCA: 101] [Impact Index Per Article: 20.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2019] [Revised: 01/17/2020] [Accepted: 01/23/2020] [Indexed: 02/07/2023] Open
Abstract
Allergic rhinitis affects the quality of life of millions of people worldwide. Air pollution not only causes morbidity, but nearly 3 million people per year die from unhealthy indoor air exposure. Furthermore, allergic rhinitis and air pollution interact. This report summarizes the discussion of an International Expert Consensus on the management of allergic rhinitis aggravated by air pollution. The report begins with a review of indoor and outdoor air pollutants followed by epidemiologic evidence showing the impact of air pollution and climate change on the upper airway and allergic rhinitis. Mechanisms, particularly oxidative stress, potentially explaining the interactions between air pollution and allergic rhinitis are discussed. Treatment for the management of allergic rhinitis aggravated by air pollution primarily involves treating allergic rhinitis by guidelines and reducing exposure to pollutants. Fexofenadine a non-sedating oral antihistamine improves AR symptoms aggravated by air pollution. However, more efficacy studies on other pharmacological therapy of coexisting AR and air pollution are currently lacking.
Collapse
Key Words
- AER, Allergic eosinophilic rhinitis
- AP, Activator protein
- AR, Allergic rhinitis
- ARE, Antioxidant response element
- Air pollutants
- Air pollution
- Allergic rhinitis
- Antioxidant enzymes
- CFS, Chronic fatigue syndrome
- CO, Carbon monoxide
- COPD, Chronic obstructive pulmonary disease
- Climate change
- DAMP, Damage-associated molecular patterns
- DEP, Diesel exhaust particles
- ECAT, Elemental carbon attributable to traffic
- ECP, Eosinophil cationic protein
- GSH-Px, Glutathione peroxidase
- HDM, House dust mites
- HEPA, High efficiency particulate air
- HO, Hemeoxygenase
- HVAC, Heating, ventilation and air conditioning
- IAP, Indoor air pollution
- IAQ, Indoor air quality
- INS, Intranasal steroids
- Indoor air quality
- LDH, Lactate dehydrogenase
- MCP, Monocyte chemotactic protein
- MSQPCR, Mold specific quantitative PCR
- NAR, Non allergic rhinitis
- NF-κβ, Nuclear factor kappa β
- NO2, Nitrogen dioxide
- NOx, Nitric oxides
- Nrf2, Nuclear factor erythroid-2 related factor
- O3, Ozone
- OAP, Outdoor air pollution
- Occupational rhinitis
- Oxidative stress
- PAMP, Pathogen-associated molecular patterns
- PM, Particulate matter
- PON, Paraoxonase
- RNS, Reactive nitrosative species
- ROS, Reactive oxygen species
- SO2, Sulphur dioxide
- SOD, Superoxide dismutase
- TLR, Toll like receptor
- TNF, Tumor necrosis factor
- TOS, Total oxidative status
- TRAP, Traffic related air pollutants
- UFP, Ultra-fine particles
- VOCs, Volatile organic compound
Collapse
Affiliation(s)
| | | | - Jean Bousquet
- INSERM U 1168, VIMA: Ageing and Chronic Diseases Epidemiological and Public Health Approaches, Villejuif, France
- University Versailles St-Quentin-en-Yvelines, France
- Allergy-Centre-Charité, Charité–Universita¨tsmedizin Berlin, Berlin, Germany
| | | | - Gennaro D'Amato
- Division of Respiratory and Allergic Diseases, High Specialty Hospital A. Cardarelli, Napoli, Italy; School of Specialization in Respiratory Diseases University Federico II Naples, Italy
| | - Nelson Rosario
- Pediatric Respiratory Medicine Division, Complexo Hospital de Clinicas, UFPR, Curitiba, Brazil
| | - Ruby Pawankar
- Dept. of Pediatrics, Nippon Medical School, Tokyo, Japan
| | - David Peden
- UNC Center for Environmental Medicine, Asthma, and Lung Biology; Division of Allergy, Immunology and Rheumatology, Dpt. of Pediatrics UNS School of Medicine, USA
| | | | - Leonard Bielory
- Medicine & Ophthalmology Hackensack Meridian School of Medicine at Seton Hall University Nutley, New Jersey, USA
| | - Luis Caraballo
- Institute for Immunological Research, University of Cartagena, Cartagena de Indias, Colombia
| | - Lorenzo Cecchi
- Centre de Bioclimatology, University de Florence, Florence, Italy
- SOS Allergy and Immunology, Prato - USL Toscana Centro, Italy
| | - S. Alfonso M. Cepeda
- Fundación Hospital Universitario Metropolitano de Barranquilla, Barranquilla, Colombia
| | | | - Carmen Galán
- Department of Botany, Ecology and Plant Physiology, University of Córdoba, Spain
| | | | - Samar Idriss
- Department of Otolaryngology- Head and Neck Surgery, Eye and Ear University Hospital, Beirut, Lebanon
| | - Todor Popov
- Alexander's University Hospital Clinic of Allergy & Asthma, Bulgaria
| | - German D. Ramon
- Alergia e Inmunología, Hospital Italiano Regional del Sur, Bahía Blanca-Buenos Aires, Argentina
| | - Erminia Ridolo
- Department of Clinical and Experimental Medicine, Università; di Parma, Parma, Italy
| | - Menachem Rottem
- Allergy Asthma and Immunology, Emek Medical Center, Afula, Israel
- Rappaport Faculty of Medicine Technion, Israel Institute of Technology, Haifa, Israel
| | - Wisuwat Songnuan
- Department of Plant Science, Faculty of Science, Mahidol University, Bangkok, Thailand
- Systems Biology of Diseases Research Unit, Faculty of Science, Mahidol University, Bangkok, Thailand
| | - Philip Rouadi
- Department of Otolaryngology- Head and Neck Surgery, Eye and Ear University Hospital, Beirut, Lebanon
| |
Collapse
|
36
|
Long A, Borro M, Sampath V, Chinthrajah RS. New Developments in Non-allergen-specific Therapy for the Treatment of Food Allergy. Curr Allergy Asthma Rep 2020; 20:3. [PMID: 31950290 DOI: 10.1007/s11882-020-0897-8] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
PURPOSE OF REVIEW The prevalence of food allergy is increasing. At the current time, there are no approved treatments for food allergy. Major limitations of immunotherapy are long treatment periods (months or years), frequent clinic visits, high costs, increased risk of adverse events during treatment, and lack of durability of desensitization. Additionally, it is allergen-specific, and in those allergic to multiple allergens, the length and cost of treatment are further increased. In this review, we summarize recent developments in novel non-allergen-specific treatments for food allergy. RECENT FINDINGS A number of monoclonal antibodies that block IgE or specific pro-allergenic cytokines or their receptors have shown promise in clinical trials for food allergy. The insight we have gained through the use of one drug for the treatment of an atopic disease is quickly being translated to other atopic diseases, including food allergy. The future for food allergy treatment with biologics looks bright.
Collapse
Affiliation(s)
- Andrew Long
- Sean N. Parker Center for Allergy and Asthma Research at Stanford University, Stanford University, Grant Building, S093, 300 Pasteur Dr., Stanford, CA, 94305-5101, USA.,Division of Pulmonary and Critical Care Medicine, Stanford University, Stanford, CA, USA
| | - Matteo Borro
- Sean N. Parker Center for Allergy and Asthma Research at Stanford University, Stanford University, Grant Building, S093, 300 Pasteur Dr., Stanford, CA, 94305-5101, USA.,Division of Pulmonary and Critical Care Medicine, Stanford University, Stanford, CA, USA.,Department of Internal Medicine, Clinical Immunology Unit, University of Genoa and Policlinico San Martino, Genoa, Italy
| | - Vanitha Sampath
- Sean N. Parker Center for Allergy and Asthma Research at Stanford University, Stanford University, Grant Building, S093, 300 Pasteur Dr., Stanford, CA, 94305-5101, USA.,Division of Pulmonary and Critical Care Medicine, Stanford University, Stanford, CA, USA
| | - R Sharon Chinthrajah
- Sean N. Parker Center for Allergy and Asthma Research at Stanford University, Stanford University, Grant Building, S093, 300 Pasteur Dr., Stanford, CA, 94305-5101, USA. .,Division of Pulmonary and Critical Care Medicine, Stanford University, Stanford, CA, USA.
| |
Collapse
|
37
|
Krysko O, Teufelberger A, Van Nevel S, Krysko DV, Bachert C. Protease/antiprotease network in allergy: The role of Staphylococcus aureus protease-like proteins. Allergy 2019; 74:2077-2086. [PMID: 30888697 DOI: 10.1111/all.13783] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2018] [Revised: 02/10/2019] [Accepted: 02/22/2019] [Indexed: 12/18/2022]
Abstract
Staphylococcus aureus is being recognized as a major cofactor in atopic diseases such as atopic dermatitis, chronic rhinosinusitis with nasal polyps, and asthma. The understanding of the relationship between S aureus virulence factors and the immune system is continuously improving. Although the precise mechanism of the host's immune response adaptation to the variable secretion profile of S aureus strains continues to be a matter of debate, an increasing number of studies have reported on central effects of S aureus secretome in allergy. In this review, we discuss how colonization of S aureus modulates the innate and adaptive immune response, thereby predisposing the organism to allergic sensitization and disrupting immune tolerance in the airways of patients with asthma and chronic rhinosinusitis with nasal polyps. Next, we provide a critical overview of novel concepts dealing with S aureus in the initiation and persistence of chronic rhinosinusitis with nasal polyps and asthma. The role of the S aureus serine protease-like proteins in the initiation of a type 2 response and the contribution of the IL-33/ST2 signaling axis in allergic responses induced by bacterial allergens are discussed.
Collapse
Affiliation(s)
- Olga Krysko
- Upper Airways Research Laboratory, Department Head and Skin Ghent University Ghent Belgium
| | - Andrea Teufelberger
- Upper Airways Research Laboratory, Department Head and Skin Ghent University Ghent Belgium
| | - Sharon Van Nevel
- Upper Airways Research Laboratory, Department Head and Skin Ghent University Ghent Belgium
| | - Dmitri V. Krysko
- Institute of Biology and Biomedicine National Research Lobachevsky State University of Nizhny Novgorod Nizhny Novgorod Russian Federation
- Cell Death Investigation and Therapy Laboratory, Department of Human Structure and Repair Ghent University Ghent Belgium
- Cancer Research Institute Ghent Ghent Belgium
| | - Claus Bachert
- Upper Airways Research Laboratory, Department Head and Skin Ghent University Ghent Belgium
| |
Collapse
|
38
|
Chen YL, Gutowska-Owsiak D, Hardman CS, Westmoreland M, MacKenzie T, Cifuentes L, Waithe D, Lloyd-Lavery A, Marquette A, Londei M, Ogg G. Proof-of-concept clinical trial of etokimab shows a key role for IL-33 in atopic dermatitis pathogenesis. Sci Transl Med 2019; 11:eaax2945. [PMID: 31645451 DOI: 10.1126/scitranslmed.aax2945] [Citation(s) in RCA: 179] [Impact Index Per Article: 29.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2019] [Revised: 06/15/2019] [Accepted: 09/25/2019] [Indexed: 12/15/2022]
Abstract
Targeted inhibition of cytokine pathways provides opportunities to understand fundamental biology in vivo in humans. The IL-33 pathway has been implicated in the pathogenesis of atopy through genetic and functional associations. We investigated the role of IL-33 inhibition in a first-in-class phase 2a study of etokimab (ANB020), an IgG1 anti-IL-33 monoclonal antibody, in patients with atopic dermatitis (AD). Twelve adult patients with moderate to severe AD received a single systemic administration of etokimab. Rapid and sustained clinical benefit was observed, with 83% achieving Eczema Area and Severity Index 50 (EASI50), and 33% EASI75, with reduction in peripheral eosinophils at day 29 after administration. We noted significant reduction in skin neutrophil infiltration after etokimab compared with placebo upon skin challenge with house dust mite, reactivity to which has been implicated in the pathogenesis of AD. We showed that etokimab also inhibited neutrophil migration to skin interstitial fluid in vitro. Besides direct effects on neutrophil migration, etokimab revealed additional unexpected CXCR1-dependent effects on IL-8-induced neutrophil migration. These human in vivo findings confirm an IL-33 upstream role in modulating skin inflammatory cascades and define the therapeutic potential for IL-33 inhibition in human diseases, including AD.
Collapse
Affiliation(s)
- Yi-Ling Chen
- MRC Human Immunology Unit, NIHR Biomedical Research Centre, Radcliffe Department of Medicine, Weatherall Institute of Molecular Medicine, University of Oxford, Oxford OX3 9DS, UK
| | - Danuta Gutowska-Owsiak
- MRC Human Immunology Unit, NIHR Biomedical Research Centre, Radcliffe Department of Medicine, Weatherall Institute of Molecular Medicine, University of Oxford, Oxford OX3 9DS, UK
- Institute of Biotechnology UG, Intercollegiate Faculty of Biotechnology of University of Gdańsk and Medical University of Gdańsk, 80-307 Gdańsk, Poland
| | - Clare S Hardman
- MRC Human Immunology Unit, NIHR Biomedical Research Centre, Radcliffe Department of Medicine, Weatherall Institute of Molecular Medicine, University of Oxford, Oxford OX3 9DS, UK
| | | | | | | | - Dominic Waithe
- MRC Human Immunology Unit, NIHR Biomedical Research Centre, Radcliffe Department of Medicine, Weatherall Institute of Molecular Medicine, University of Oxford, Oxford OX3 9DS, UK
| | | | | | | | - Graham Ogg
- MRC Human Immunology Unit, NIHR Biomedical Research Centre, Radcliffe Department of Medicine, Weatherall Institute of Molecular Medicine, University of Oxford, Oxford OX3 9DS, UK.
- Oxford University Hospitals, Oxford OX3 7LE, UK
| |
Collapse
|
39
|
An G, Wang W, Zhang X, Huang Q, Li Q, Chen S, Du X, Corrigan CJ, Huang K, Wang W, Chen Y, Ying S. Combined blockade of IL-25, IL-33 and TSLP mediates amplified inhibition of airway inflammation and remodelling in a murine model of asthma. Respirology 2019; 25:603-612. [PMID: 31610614 DOI: 10.1111/resp.13711] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2018] [Revised: 07/12/2019] [Accepted: 09/23/2019] [Indexed: 12/23/2022]
Abstract
BACKGROUND AND OBJECTIVE Isolated blockade of IL-25, IL-33 and thymic stromal lymphopoietin (TSLP) has been shown to reduce airways inflammation and hyperresponsiveness in murine asthma model. The hypothesis that combined blockade of all three cytokines can accomplish this more effectively has never been addressed. METHODS We studied a murine asthma model employing sensitization and challenge with ovalbumin (OVA) or saline control. To discern the effects of IL-33 blockade, we compared outcomes in strain identical, wild-type and IL-33 receptor (St2 -/- ) gene-deleted mice. We then examined, in the St2 -/- animals, the effects of additional, single or combined blockade of IL-25 and TSLP with blocking antibodies. Outcomes included airways reactivity, inflammatory cellular infiltration, epithelial cell metaplasia, deposition of fibrosis-related proteins, local Th2-type cytokine expression and total and specific serum IgE concentrations measured by ELISA and quantitative immunohistochemistry. RESULTS St2 -/- gene deletion significantly reduced airways reactivity, inflammatory cellular infiltration, lung tissue expression of Th2 cytokines and fibrosis related proteins and serum total IgE in response to OVA sensitization and challenge. Additional administration of anti-IL-25 and anti-TSLP blocking antibodies to the St2 -/- mice further significantly reduced inflammation, Th2 cytokine expression, airways fibrosis and IgE production, while anti-TSLP alone reduced eosinophil infiltration and local IL-4 expression. The airways inflammatory cellular infiltrate and lung tissue expression of Th2 cytokine, but not fibrosis-related proteins were also reduced in the presence of isotype identical, control antibodies. CONCLUSION Combined blockade of these three cytokines may better ameliorate airways pathological changes in this murine asthma model, with implications for human asthma.
Collapse
Affiliation(s)
- Gao An
- Department of Immunology, School of Basic Medical Sciences, Capital Medical University, Beijing, China
| | - Wenjun Wang
- Department of Respiratory and Critical Care Medicine, Beijing Chao-Yang Hospital, Capital Medical University and Beijing Institute of Respiratory Medicine, Beijing, China
| | - Xin Zhang
- Department of Immunology, School of Basic Medical Sciences, Capital Medical University, Beijing, China
| | - Qiong Huang
- Department of Immunology, School of Basic Medical Sciences, Capital Medical University, Beijing, China
| | - Qin Li
- Department of Immunology, School of Basic Medical Sciences, Capital Medical University, Beijing, China
| | - Shihao Chen
- Department of Immunology, School of Basic Medical Sciences, Capital Medical University, Beijing, China
| | - Xiaonan Du
- Department of Immunology, School of Basic Medical Sciences, Capital Medical University, Beijing, China
| | - Chris J Corrigan
- Asthma UK Centre in Allergic Mechanisms of Asthma, School of Immunology and Microbial Sciences, Faculty of Life Sciences and Medicine, King's College London, London, UK
| | - Kewu Huang
- Department of Respiratory and Critical Care Medicine, Beijing Chao-Yang Hospital, Capital Medical University and Beijing Institute of Respiratory Medicine, Beijing, China
| | - Wei Wang
- Department of Immunology, School of Basic Medical Sciences, Capital Medical University, Beijing, China
| | - Yan Chen
- Department of Immunology, School of Basic Medical Sciences, Capital Medical University, Beijing, China
| | - Sun Ying
- Department of Immunology, School of Basic Medical Sciences, Capital Medical University, Beijing, China
| |
Collapse
|
40
|
Lan F, Zhang N, Holtappels G, De Ruyck N, Krysko O, Van Crombruggen K, Braun H, Johnston SL, Papadopoulos NG, Zhang L, Bachert C. Staphylococcus aureus Induces a Mucosal Type 2 Immune Response via Epithelial Cell-derived Cytokines. Am J Respir Crit Care Med 2019; 198:452-463. [PMID: 29768034 DOI: 10.1164/rccm.201710-2112oc] [Citation(s) in RCA: 87] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023] Open
Abstract
RATIONALE Chronic rhinosinusitis with nasal polyps is characterized by a T-helper cell type 2-skewed upper airway inflammation. Mucosal Staphylococcus aureus colonization is found in the majority of patients with nasal polyps. S. aureus is known to induce type 2 cytokine release via enterotoxins. OBJECTIVES To investigate the impact of non-enterotoxin-producing S. aureus on type 2 cytokine release. METHODS TSLP (thymic stromal lymphopoietin), IL-33, and type 2 cytokines were assessed in a human mucosal tissue model upon S. aureus infection. MEASUREMENTS AND MAIN RESULTS S. aureus exposure increased the expression of IL-33, TSLP, IL-5, and IL-13 in nasal polyp tissue, accompanied by elevated expression levels of TSLP and IL-33 receptors, predominantly on CD3+ T cells. S. aureus infection led to the release of TSLP, but not IL-33, IL-5, or IL-13, from healthy inferior turbinate tissue. In contrast, S. epidermidis did not induce any epithelial cell-derived cytokines in nasal polyp or healthy tissue. S. aureus infection also increased the release of IL-33 and TSLP in BEAS-2B epithelial cells, accompanied by activation of NF-κB (nuclear factor κB) pathways. Incubation with CU-CPT22, a specific Toll-like receptor 2 antagonist, significantly reduced the S. aureus-induced release of TSLP and IL-33, and the activity of the NF-κB signal in BEAS-2B cells. CONCLUSIONS This study demonstrates for the first time that S. aureus can directly induce epithelial cell-derived cytokine release via binding to Toll-like receptor 2, and may thereby propagate type 2 cytokine expression in nasal polyp tissue.
Collapse
Affiliation(s)
- Feng Lan
- 1 Department of Otolaryngology Head and Neck Surgery, Beijing Institute of Otolaryngology, Beijing TongRen Hospital, Capital Medical University, Beijing, China.,2 Upper Airways Research Laboratory, Department of Otorhinolaryngology, Ghent University Hospital, Ghent, Belgium
| | - Nan Zhang
- 2 Upper Airways Research Laboratory, Department of Otorhinolaryngology, Ghent University Hospital, Ghent, Belgium
| | - Gabriele Holtappels
- 2 Upper Airways Research Laboratory, Department of Otorhinolaryngology, Ghent University Hospital, Ghent, Belgium
| | - Natalie De Ruyck
- 2 Upper Airways Research Laboratory, Department of Otorhinolaryngology, Ghent University Hospital, Ghent, Belgium
| | - Olga Krysko
- 2 Upper Airways Research Laboratory, Department of Otorhinolaryngology, Ghent University Hospital, Ghent, Belgium
| | - Koen Van Crombruggen
- 2 Upper Airways Research Laboratory, Department of Otorhinolaryngology, Ghent University Hospital, Ghent, Belgium
| | - Harald Braun
- 3 VIB Inflammation Research Center, Ghent University, Ghent, Belgium
| | - Sebastian L Johnston
- 4 Airway Disease Infection Section, National Heart and Lung Institute, Imperial College London, London, United Kingdom
| | - Nikos G Papadopoulos
- 5 Centre for Pediatrics and Child Health, Institute of Human Development, University of Manchester, Manchester, United Kingdom; and
| | - Luo Zhang
- 1 Department of Otolaryngology Head and Neck Surgery, Beijing Institute of Otolaryngology, Beijing TongRen Hospital, Capital Medical University, Beijing, China
| | - Claus Bachert
- 2 Upper Airways Research Laboratory, Department of Otorhinolaryngology, Ghent University Hospital, Ghent, Belgium.,6 Division of ENT Diseases, Clintec, Karolinska Institute, Stockholm, Sweden
| |
Collapse
|
41
|
Innate lymphoid cells in asthma: pathophysiological insights from murine models to human asthma phenotypes. Curr Opin Allergy Clin Immunol 2019; 19:53-60. [PMID: 30516548 DOI: 10.1097/aci.0000000000000497] [Citation(s) in RCA: 33] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
PURPOSE OF REVIEW The current review describes the role of different types of innate lymphoid cells (ILCs) in the pathogenesis of asthma inflammatory phenotypes by linking findings from murine asthma models with human studies. Novel treatment options are needed for patients with steroid-insensitive asthma. Strategies targeting ILCs, or their upstream or downstream molecules are emerging and discussed in this review. RECENT FINDINGS In eosinophilic asthma, ILCs, and especially type 2 ILCs (ILC2s), are activated by alarmins such as IL-33 upon allergen triggering of the airway epithelium. This initiates IL-5 and IL-13 production by ILC2, resulting in eosinophilic inflammation and airway hyperreactivity. Type 3 ILCs (ILC3s) have been shown to be implicated in obesity-induced asthma, via IL-1β production by macrophages, leading ILC3 and release of IL-17. ILC1s might play a role in severe asthma, but its role is currently less investigated. SUMMARY Several studies have revealed that ILC2s play a role in the induction of eosinophilic inflammation in allergic and nonallergic asthmatic patients mainly via IL-5, IL-13, IL-33 and thymic stromal lymphopoietin. Knowledge on the role of ILC3s and ILC1s in asthmatic patients is lagging behind. Further studies are needed to support the hypothesis that these other types of ILCs contribute to asthma pathogenesis, presumably in nonallergic asthma phenotypes.
Collapse
|
42
|
Zhang Y, Li S, Huang S, Cao L, Liu T, Zhao J, Wu J, Wang J, Cao L, Xu J, Dong L. IL33/ST2 contributes to airway remodeling via p-JNK MAPK/STAT3 signaling pathway in OVA-induced allergic airway inflammation in mice. Exp Lung Res 2019; 45:65-75. [PMID: 31112061 DOI: 10.1080/01902148.2019.1611972] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Aim of this study: Airway remodeling, which encompasses structural changes in airway is a main feature of asthma. Interleukin-33 (IL-33) has been reported to be a vital cytokine in airway remodeling in asthma, but the underlying mechanisms are not clear yet. This study focused on discussing the role of IL-33 in airway remodeling in asthma. Material and methods: Female BALB/c mice were divided into a control group, an OVA induced allergic airway disease group and an anti-ST2 antibody intervention group. Immunohistochemistry and western blot were performed to detect IL-33, ST2 expression in addition to airway remodeling markers a-smooth muscle actin (a-SMA) and type 1 collagen in OVA-induced mice model. Levels of p-JNK and p-STAT3 activation in mice were detected by western blot. Human lung fibroblast (HLF) were stimulated with rhIL-33, anti-ST2 antibody and JNK inhibitor sp600125 and levels of JNK and STAT3 activation were determined via western blot and immunofluorescence staining. Results: Anti-ST2 treatment inhibited JNK/STAT3 phosphorylation and airway remodeling in OVA-induced mouse model. IL-33 induced a-SMA and collagen 1 expression was inhibited by anti-ST2 antibody and sp600125 treatment via decreased JNK/STAT3 phosphorylation in human lung fibroblast. Conclusions: IL-33 promoted airway remodeling by interacting with ST2 to activate the JNK/STAT3 signaling pathway in asthma.
Collapse
Affiliation(s)
- Yuanyuan Zhang
- a Department of Pulmonary Diseases , Qilu Hospital, Shandong University , Jinan , Shandong , People's Republic of China
| | - Shuo Li
- a Department of Pulmonary Diseases , Qilu Hospital, Shandong University , Jinan , Shandong , People's Republic of China
| | - Siyuan Huang
- a Department of Pulmonary Diseases , Qilu Hospital, Shandong University , Jinan , Shandong , People's Republic of China
| | - Liuzhao Cao
- a Department of Pulmonary Diseases , Qilu Hospital, Shandong University , Jinan , Shandong , People's Republic of China
| | - Tian Liu
- a Department of Pulmonary Diseases , Qilu Hospital, Shandong University , Jinan , Shandong , People's Republic of China
| | - Jiping Zhao
- a Department of Pulmonary Diseases , Qilu Hospital, Shandong University , Jinan , Shandong , People's Republic of China
| | - Jinxiang Wu
- a Department of Pulmonary Diseases , Qilu Hospital, Shandong University , Jinan , Shandong , People's Republic of China
| | - Junfei Wang
- a Department of Pulmonary Diseases , Qilu Hospital, Shandong University , Jinan , Shandong , People's Republic of China
| | - Lili Cao
- a Department of Pulmonary Diseases , Qilu Hospital, Shandong University , Jinan , Shandong , People's Republic of China
| | - Jiawei Xu
- a Department of Pulmonary Diseases , Qilu Hospital, Shandong University , Jinan , Shandong , People's Republic of China
| | - Liang Dong
- a Department of Pulmonary Diseases , Qilu Hospital, Shandong University , Jinan , Shandong , People's Republic of China
| |
Collapse
|
43
|
Shang K, Wei Y, Su Q, Yu B, Tao Y, He Y, Wang Y, Shi G, Duan L. IL-33 Ameliorates the Development of MSU-Induced Inflammation Through Expanding MDSCs-Like Cells. Front Endocrinol (Lausanne) 2019; 10:36. [PMID: 30863362 PMCID: PMC6399133 DOI: 10.3389/fendo.2019.00036] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/20/2018] [Accepted: 01/16/2019] [Indexed: 11/15/2022] Open
Abstract
Interleukin-33 (IL-33), a member of the IL-1 superfamily, has been shown to play a critical role in many diseases through regulating the immune cell responses, including myeloid-derived suppressor cells (MDSCs). Our previous study demonstrated that IL-33 might play a protective role in kidney injury in gout patients by regulating the lipid metabolism. However, the role of IL-33in the development of MSU-induced inflammation remains elusive. In this study, an increased IL-33 expression was observed in gout patients, which was positively correlated with inflammatory marker CRP. To explore the effects and mechanisms of the increased IL-33 expression in the gout patients, the anti-ST2 antibody and exogenous recombinant IL-33 were used in MSU-induced peritonitis animal model that mimics human gout. Compared with control group, mice with exogenous recombinant IL-33 significantly ameliorated the inflammatory cells infiltration, while blockage of IL-33 signaling by anti-ST2 had no effect on the development of MSU-induced peritonitis. Furthermore, the crucial inflammatory cytokine IL-1β was markedly decreased in IL-33-treated mice. Besides that, a large number of anti-inflammatory MDSCs with CD11b+Gr1intF4/80+ phenotype was observed in the IL-33-treated mice, and adoptive transfer of IL-33-induced MDSCs (CD11b+Gr1intF4/80+) markedly inhibited the IL-1β production in MSU-induced peritonitis. In conclusion, our data provide clear evidences that the increased expression of IL-33 in the gout patients might be due to a cause of self-negative regulation, which inhibits the development of MSU-induced inflammation through expanding MDSCs. Thus, IL-33 might serve as a promising therapeutic target for gout.
Collapse
Affiliation(s)
- Ke Shang
- Department of Rheumatology and Clinical Immunology, Jiangxi Provincial People's Hospital, Nanchang, China
| | - Yingying Wei
- Department of Rheumatology and Clinical Immunology, The First Affiliated Hospital of Xiamen University, Xiamen, China
| | - Qun Su
- Department of Rheumatology and Clinical Immunology, The First Affiliated Hospital of Xiamen University, Xiamen, China
| | - Bing Yu
- Department of Rheumatology and Clinical Immunology, The First Affiliated Hospital of Xiamen University, Xiamen, China
| | - Ying Tao
- Department of Rheumatology and Clinical Immunology, The First Affiliated Hospital of Xiamen University, Xiamen, China
| | - Yan He
- Department of Rheumatology and Clinical Immunology, The First Affiliated Hospital of Xiamen University, Xiamen, China
| | - Youlian Wang
- Department of Rheumatology and Clinical Immunology, Jiangxi Provincial People's Hospital, Nanchang, China
| | - Guixiu Shi
- Department of Rheumatology and Clinical Immunology, The First Affiliated Hospital of Xiamen University, Xiamen, China
- Guixiu Shi
| | - Lihua Duan
- Department of Rheumatology and Clinical Immunology, Jiangxi Provincial People's Hospital, Nanchang, China
- Department of Rheumatology and Clinical Immunology, The First Affiliated Hospital of Xiamen University, Xiamen, China
- *Correspondence: Lihua Duan
| |
Collapse
|
44
|
Abukabda AB, McBride CR, Batchelor TP, Goldsmith WT, Bowdridge EC, Garner KL, Friend S, Nurkiewicz TR. Group II innate lymphoid cells and microvascular dysfunction from pulmonary titanium dioxide nanoparticle exposure. Part Fibre Toxicol 2018; 15:43. [PMID: 30413212 PMCID: PMC6230229 DOI: 10.1186/s12989-018-0280-2] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2018] [Accepted: 10/24/2018] [Indexed: 01/16/2023] Open
Abstract
Background The cardiovascular effects of pulmonary exposure to engineered nanomaterials (ENM) are poorly understood, and the reproductive consequences are even less understood. Inflammation remains the most frequently explored mechanism of ENM toxicity. However, the key mediators and steps between lung exposure and uterine health remain to be fully defined. The purpose of this study was to determine the uterine inflammatory and vascular effects of pulmonary exposure to titanium dioxide nanoparticles (nano-TiO2). We hypothesized that pulmonary nano-TiO2 exposure initiates a Th2 inflammatory response mediated by Group II innate lymphoid cells (ILC2), which may be associated with an impairment in uterine microvascular reactivity. Methods Female, virgin, Sprague-Dawley rats (8–12 weeks) were exposed to 100 μg of nano-TiO2 via intratracheal instillation 24 h prior to microvascular assessments. Serial blood samples were obtained at 0, 1, 2 and 4 h post-exposure for multiplex cytokine analysis. ILC2 numbers in the lungs were determined. ILC2s were isolated and phosphorylated nuclear factor kappa-light-chain-enhancer of activated B cells (NF-ĸB) levels were measured. Pressure myography was used to assess vascular reactivity of isolated radial arterioles. Results Pulmonary nano-TiO2 exposure was associated with an increase in IL-1ß, 4, 5 and 13 and TNF- α 4 h post-exposure, indicative of an innate Th2 inflammatory response. ILC2 numbers were significantly increased in lungs from exposed animals (1.66 ± 0.19%) compared to controls (0.19 ± 0.22%). Phosphorylation of the transactivation domain (Ser-468) of NF-κB in isolated ILC2 and IL-33 in lung epithelial cells were significantly increased (126.8 ± 4.3% and 137 ± 11% of controls respectively) by nano-TiO2 exposure. Lastly, radial endothelium-dependent arteriolar reactivity was significantly impaired (27 ± 12%), while endothelium-independent dilation (7 ± 14%) and α-adrenergic sensitivity (8 ± 2%) were not altered compared to control levels. Treatment with an anti- IL-33 antibody (1 mg/kg) 30 min prior to nano-TiO2 exposure resulted in a significant improvement in endothelium-dependent dilation and a decreased level of IL-33 in both plasma and bronchoalveolar lavage fluid. Conclusions These results provide evidence that the uterine microvascular dysfunction that follows pulmonary ENM exposure may be initiated via activation of lung-resident ILC2 and subsequent systemic Th2-dependent inflammation. Electronic supplementary material The online version of this article (10.1186/s12989-018-0280-2) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Alaeddin Bashir Abukabda
- Department of Physiology and Pharmacology, West Virginia University School of Medicine, 64 Medical Center Drive, Robert C. Byrd Health Sciences Center - West Virginia University, Morgantown, WV, 26505-9229, USA.,Toxicology Working Group, West Virginia University School of Medicine, Morgantown, WV, USA
| | - Carroll Rolland McBride
- Department of Physiology and Pharmacology, West Virginia University School of Medicine, 64 Medical Center Drive, Robert C. Byrd Health Sciences Center - West Virginia University, Morgantown, WV, 26505-9229, USA.,Toxicology Working Group, West Virginia University School of Medicine, Morgantown, WV, USA
| | - Thomas Paul Batchelor
- Department of Physiology and Pharmacology, West Virginia University School of Medicine, 64 Medical Center Drive, Robert C. Byrd Health Sciences Center - West Virginia University, Morgantown, WV, 26505-9229, USA.,Toxicology Working Group, West Virginia University School of Medicine, Morgantown, WV, USA
| | - William Travis Goldsmith
- Department of Physiology and Pharmacology, West Virginia University School of Medicine, 64 Medical Center Drive, Robert C. Byrd Health Sciences Center - West Virginia University, Morgantown, WV, 26505-9229, USA.,Toxicology Working Group, West Virginia University School of Medicine, Morgantown, WV, USA
| | - Elizabeth Compton Bowdridge
- Department of Physiology and Pharmacology, West Virginia University School of Medicine, 64 Medical Center Drive, Robert C. Byrd Health Sciences Center - West Virginia University, Morgantown, WV, 26505-9229, USA.,Toxicology Working Group, West Virginia University School of Medicine, Morgantown, WV, USA
| | - Krista Lee Garner
- Department of Physiology and Pharmacology, West Virginia University School of Medicine, 64 Medical Center Drive, Robert C. Byrd Health Sciences Center - West Virginia University, Morgantown, WV, 26505-9229, USA.,Toxicology Working Group, West Virginia University School of Medicine, Morgantown, WV, USA
| | - Sherri Friend
- National Institute for Occupational Safety and Health, Morgantown, WV, USA
| | - Timothy Robert Nurkiewicz
- Department of Physiology and Pharmacology, West Virginia University School of Medicine, 64 Medical Center Drive, Robert C. Byrd Health Sciences Center - West Virginia University, Morgantown, WV, 26505-9229, USA. .,Toxicology Working Group, West Virginia University School of Medicine, Morgantown, WV, USA. .,National Institute for Occupational Safety and Health, Morgantown, WV, USA.
| |
Collapse
|
45
|
Wang Y, Li C, Xu Y, Xu D, Yang G, Liao F, Luo X. Sublingual Immunotherapy Decreases Expression of Interleukin-33 in Children with Allergic Rhinitis. Indian J Pediatr 2018; 85:872-876. [PMID: 29790007 DOI: 10.1007/s12098-018-2703-3] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/27/2017] [Accepted: 05/03/2018] [Indexed: 01/09/2023]
Abstract
OBJECTIVES To identify the expression of IL-33 during SLIT (Sublingual immunotherapy) in AR (Allergic rhinitis) children. METHODS Thirty children received house dust mite (HDM) allergen extract for SLIT and thirty children received placebo in this study. Serum and nasal lavage samples of cases and controls were collected at different time points during SLIT. Interleukin (IL)-33 and other cytokines were estimated in these samples by enzyme-linked immuno sorbent assay (ELISA). Peripheral blood mononuclear cells (PBMC) were prepared and stimulated with rhIL-33 (with or without other stimulators) at different time points during SLIT. RESULTS The present results showed that both serum and nasal lavage of IL-33 levels decreased significantly after 12 mo treatment and this trend maintained at least until 24 mo. The decreased nasal IL-33 level was positively correlated to local Th2 cytokines and increased IL-10 expression at 2 y post SLIT treatment. In vitro experiments showed that IL-33 promotes IL-4 and IL-5 and inhibits IL-10 expression by peripheral blood mononuclear cells (PBMCs) in AR. CONCLUSIONS Decreased IL-33 expression during SLIT may contribute to low Th2 response and enhanced Regulatory T cell cytokines expression. Thus, IL-33 maybe an important predictor during SLIT.
Collapse
Affiliation(s)
- Yuanming Wang
- Department of Otolaryngology, Dongguan Women and Children's Hospital, No 99. Zhenxing Road, Dongguan, 523120, China
| | - Chuling Li
- Department of Otolaryngology, Dongguan Women and Children's Hospital, No 99. Zhenxing Road, Dongguan, 523120, China.
| | - Yaxiong Xu
- Department of Otolaryngology, Dongguan Women and Children's Hospital, No 99. Zhenxing Road, Dongguan, 523120, China
| | - Deyu Xu
- Department of Otolaryngology, Dongguan Women and Children's Hospital, No 99. Zhenxing Road, Dongguan, 523120, China
| | - Gang Yang
- Department of Otolaryngology, Dongguan Women and Children's Hospital, No 99. Zhenxing Road, Dongguan, 523120, China
| | - Fang Liao
- Department of Otolaryngology, Dongguan Women and Children's Hospital, No 99. Zhenxing Road, Dongguan, 523120, China
| | - Xianglin Luo
- Department of Otolaryngology, Dongguan Women and Children's Hospital, No 99. Zhenxing Road, Dongguan, 523120, China
| |
Collapse
|
46
|
The Risk G Allele of the Single-Nucleotide Polymorphism rs928413 Creates a CREB1-Binding Site That Activates IL33 Promoter in Lung Epithelial Cells. Int J Mol Sci 2018; 19:ijms19102911. [PMID: 30257479 PMCID: PMC6212888 DOI: 10.3390/ijms19102911] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2018] [Revised: 09/20/2018] [Accepted: 09/21/2018] [Indexed: 12/22/2022] Open
Abstract
Cytokine interleukin 33 (IL-33) is constitutively expressed by epithelial barrier cells, and promotes the development of humoral immune responses. Along with other proinflammatory mediators released by the epithelium of airways and lungs, it plays an important role in a number of respiratory pathologies. In particular, IL-33 significantly contributes to pathogenesis of allergy and asthma; genetic variations in the IL33 locus are associated with increased susceptibility to asthma. Large-scale genome-wide association studies have identified minor “G” allele of the single-nucleotide polymorphism rs928413, located in the IL33 promoter area, as a susceptible variant for early childhood and atopic asthma development. Here, we demonstrate that the rs928413(G) allele creates a binding site for the cAMP response element-binding protein 1 (CREB1) transcription factor. In a pulmonary epithelial cell line, activation of CREB1, presumably via the p38 mitogen-activated protein kinases (MAPK) cascade, activates the IL33 promoter containing the rs928413(G) allele specifically and in a CREB1-dependent manner. This mechanism may explain the negative effect of the rs928413 minor “G” allele on asthma development.
Collapse
|
47
|
Patel NN, Kohanski MA, Maina IW, Workman AD, Herbert DR, Cohen NA. Sentinels at the wall: epithelial-derived cytokines serve as triggers of upper airway type 2 inflammation. Int Forum Allergy Rhinol 2018; 9:93-99. [PMID: 30260580 DOI: 10.1002/alr.22206] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2018] [Revised: 08/19/2018] [Accepted: 08/21/2018] [Indexed: 12/28/2022]
Abstract
Recent evidence has demonstrated an expanding role of respiratory epithelial cells in immune surveillance and modulation. Studies have been focusing on the earliest events that link epithelial injury to downstream inflammatory responses. Cytokines produced by and released from respiratory epithelial cells are among these early trigger signals. Epithelial-derived cytokines, namely thymic stromal lymphopoietin (TSLP), interleukin (IL)-25, and IL-33, have come to the forefront of recent investigations. Each of these 3 cytokines has been implicated in chronic rhinosinusitis (CRS), asthma, and atopy. Herein we review studies elucidating the roles of epithelial-derived cytokines in the pathobiology of upper airway disease, with particular emphasis on type 2 inflammatory conditions.
Collapse
Affiliation(s)
- Neil N Patel
- Department of Otorhinolaryngology-Head and Neck Surgery, Division of Rhinology, University of Pennsylvania, Perelman School of Medicine, Philadelphia, PA
| | - Michael A Kohanski
- Department of Otorhinolaryngology-Head and Neck Surgery, Division of Rhinology, University of Pennsylvania, Perelman School of Medicine, Philadelphia, PA
| | - Ivy W Maina
- Department of Otorhinolaryngology-Head and Neck Surgery, Division of Rhinology, University of Pennsylvania, Perelman School of Medicine, Philadelphia, PA
| | - Alan D Workman
- Department of Otorhinolaryngology-Head and Neck Surgery, Division of Rhinology, University of Pennsylvania, Perelman School of Medicine, Philadelphia, PA
| | - De'Broski R Herbert
- Department of Pathobiology, University of Pennsylvania School of Veterinary Medicine, Philadelphia, PA
| | - Noam A Cohen
- Department of Otorhinolaryngology-Head and Neck Surgery, Division of Rhinology, University of Pennsylvania, Perelman School of Medicine, Philadelphia, PA.,Philadelphia Veterans Affairs Medical Center, Philadelphia, PA.,Monell Chemical Senses Center, Philadelphia, PA
| |
Collapse
|
48
|
Li J, Saruta K, Dumouchel JP, Magat JM, Thomas JL, Ajami D, Rebek M, Rebek J, Bigby TD. Small Molecule Mimetics of α-Helical Domain of IRAK2 Attenuate the Proinflammatory Effects of IL-33 in Asthma-like Mouse Models. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2018; 200:4036-4043. [PMID: 29728508 PMCID: PMC5988972 DOI: 10.4049/jimmunol.1700693] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/11/2017] [Accepted: 04/10/2018] [Indexed: 01/04/2023]
Abstract
IL-33 and its receptor ST2 play important roles in airway inflammation and contribute to asthma onset and exacerbation. The IL-33/ST2 signaling pathway recruits adapter protein myeloid differentiation primary response 88 (MyD88) to transduce intracellular signaling. MyD88 forms a complex with IL-R-associated kinases (IRAKs), IRAK4 and IRAK2, called the Myddosome (MyD88-IRAK4-IRAK2). The myddosome subsequently activates downstream NF-κB and MAPKs p38 and JNK. We established an asthma-like mouse model by intratracheal administration of IL-33. The IL-33 model has a very similar phenotype compared with the OVA-induced mouse asthma model. The importance of MyD88 in the IL-33/ST2 signaling transduction was demonstrated by the MyD88 knockout mice, which were protected from the IL-33-induced asthma. We synthesized small molecule mimetics of the α-helical domain of IRAK2 with drug-like characteristics based on the recent advances in the designing of α-helix compounds. The mimetics can competitively interfere in the protein-protein interaction between IRAK2 and IRAK4, leading to disruption of Myddosome formation. A series of small molecules were screened using an NF-κB promoter assay in vitro. The lead compound, 7004, was further studied in the IL-33-induced and OVA-induced asthma mouse models in vivo. Compound 7004 can inhibit the IL-33-induced NF-κB activity, disrupt Myddosome formation, and attenuate the proinflammatory effects in asthma-like models. Our data indicate that the Myddosome may represent a novel intracellular therapeutic target for diseases in which IL-33/ST2 plays important roles, such as asthma and other inflammatory diseases.
Collapse
Affiliation(s)
- Jinghong Li
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, University of California, San Diego, La Jolla, CA 92093;
- Veterans Affairs San Diego Healthcare System, San Diego, CA 92161; and
| | - Kunio Saruta
- The Scripps Research Institute, La Jolla, CA 92037
| | - Justin P Dumouchel
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, University of California, San Diego, La Jolla, CA 92093
- Veterans Affairs San Diego Healthcare System, San Diego, CA 92161; and
| | - Jenna M Magat
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, University of California, San Diego, La Jolla, CA 92093
- Veterans Affairs San Diego Healthcare System, San Diego, CA 92161; and
| | - Joanna L Thomas
- Veterans Affairs San Diego Healthcare System, San Diego, CA 92161; and
| | | | - Mitra Rebek
- The Scripps Research Institute, La Jolla, CA 92037
| | - Julius Rebek
- The Scripps Research Institute, La Jolla, CA 92037
| | - Timothy D Bigby
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, University of California, San Diego, La Jolla, CA 92093
- Veterans Affairs San Diego Healthcare System, San Diego, CA 92161; and
| |
Collapse
|
49
|
Mitchell PD, Salter BM, Oliveria JP, El-Gammal A, Tworek D, Smith SG, Sehmi R, Gauvreau GM, O Apos Byrne PM. IL-33 and Its Receptor ST2 after Inhaled Allergen Challenge in Allergic Asthmatics. Int Arch Allergy Immunol 2018; 176:133-142. [PMID: 29694974 DOI: 10.1159/000488015] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2017] [Accepted: 02/12/2018] [Indexed: 12/18/2022] Open
Abstract
BACKGROUND Previous murine models have demonstrated interleukin (IL)-33 to be an important mediator of type-2 inflammation and to promote airway hyperresponsiveness in allergic asthma. A number of inflammatory cells produce IL-33 and eosinophils express ST2 mRNA. The relationship between IL-33 and eosinophils in allergic asthma, however, remains unclear. OBJECTIVE The aim of this work was to evaluate in vitro the effect of allergen inhalation on IL-33 levels and expression of its receptor (ST2L) on eosinophils in allergic asthmatics, and the effect of IL-33 stimulation on eosinophil activity. METHODS Plasma and sputum IL-33, soluble ST2 (sST2) levels, and ST2L expression on eosinophils were measured in 10 healthy controls and 10 allergic asthmatics. Asthmatics underwent allergen and diluent inhalation challenges. Blood and sputum samples were collected to measure IL-33, sST2, and ST2L eosinophil expression before and 24 h after allergen inhalation. Purified blood eosinophils from allergic asthmatics were incubated overnight with IL-33 to assess ST2 and intracellular IL-5 expression. RESULTS Baseline levels of IL-33 in sputum and sST2 in plasma and sputum were similar in allergic asthmatics compared to healthy controls. In addition, there was no difference in blood or sputum eosinophil ST2L expression in healthy controls versus allergic asthmatics. Eosinophil ST2L expression was significantly increased 24 h postallergen inhalation in allergic asthmatics. In vitro stimulation of human eosinophils with IL-33 and LPS significantly increased eosinophil ST2L expression and IL-33 stimulation increased intracellular IL-5 expression, which was attenuated by treatment with sST2 and ST2 blockade. CONCLUSION AND CLINICAL RELEVANCE In mild asthmatics, there was a significant upregulation of ST2 surface expression on eosinophils from blood and sputum following allergen inhalation challenge. In vitro, IL-33 stimulation of eosinophils increases both ST2 membrane expression and IL-5 production. These results support a role for IL-33 in causing allergen-induced eosinophilia. Blockade of IL-33 and ST2 signaling may present a novel therapeutic avenue for asthma treatment.
Collapse
|
50
|
Khaitov MR, Gaisina AR, Shilovskiy IP, Smirnov VV, Ramenskaia GV, Nikonova AA, Khaitov RM. The Role of Interleukin-33 in Pathogenesis of Bronchial Asthma. New Experimental Data. BIOCHEMISTRY (MOSCOW) 2018. [PMID: 29534664 DOI: 10.1134/s0006297918010029] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
Interleukin-33 (IL-33) belongs to the IL-1 cytokine family and plays an important role in modulating immune system by inducing Th2 immune response via the ST2 membrane receptor. Epithelial cells are the major producers of IL-33. However, IL-33 is also secreted by other cells, e.g., bone marrow cells, dendritic cells, macrophages, and mast cells. IL-33 targets a broad range of cell types bearing the ST2 surface receptor. Many ST2-positive cells, such as Th2 cells, mast cells, basophils, and eosinophils, are involved in the development of allergic bronchial asthma (BA). This suggests that IL-33 directly participates in BA pathogenesis. Currently, the role of IL-33 in pathogenesis of inflammatory disorders, including BA, has been extensively investigated using clinical samples collected from patients, as well as asthma animal models. In particular, numerous studies on blocking IL-33 and its receptor by monoclonal antibodies in asthma mouse model have been performed over the last several years; IL-33- and ST2-deficient transgenic mice have also been generated. In this review, we summarized and analyzed the data on the role of IL-33 in BA pathogenesis and the prospects for creating new treatments for BA.
Collapse
Affiliation(s)
- M R Khaitov
- Institute of Immunology, FMBA of Russia, Moscow, 115478, Russia.
| | | | | | | | | | | | | |
Collapse
|