1
|
Zhang Y, Zhang W, Ma M, Zhang X, Li C, Deng T, Gao J, Gao C, Wang N. Corydalis yanhusuo extract and its pharmacological substances alleviate food allergy by inhibiting mast cells activation via PLC/PKC/STAT3 pathway. JOURNAL OF ETHNOPHARMACOLOGY 2025; 337:118809. [PMID: 39251152 DOI: 10.1016/j.jep.2024.118809] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/30/2024] [Revised: 09/03/2024] [Accepted: 09/07/2024] [Indexed: 09/11/2024]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Food allergies have increasingly become a disease that affects global health and need for corresponding therapeutic drugs urgently. As a traditional Chinses medicine with a wide range of pharmacological effects, however, there was no clear research confirming therapeutic effect and pharmacological substances of Corydalis yanhusuo (YHS) on food allergies. Mast cells (MCs) are the main effector cells which mediate allergic and pseudo-allergic reactions. MATERIALS AND METHODS In this study, we investigated the effect of YHS extract on treating food allergy and its underlying mechanism. The inhibitory effect of YHS on MCs activation in vitro was evaluated by Ca2+ influx, degranulation, and cytokine release detection. The in vivo effect was investigated using the passive cutaneous anaphylaxis (PCA), active systemic allergy as well as OVA-induced food allergy mice. Western blot was performed to reveal the signaling pathway. RESULTS YHS extract showed an inhibitory effect on MCs activation and food allergy both in vitro and in vivo. PLC/PKC/STAT3 signaling pathway was suppressed by YHS extract in the disease. HPLC analysis revealed YHS extract contains corydaline and tetrahydropalmatine, and both compounds inhibited MCs activation induced by C48/80 in vitro. CONCLUSION YHS extract inhibited the MCs activation and food allergy via PLC/PKC/STAT3 pathway.
Collapse
Affiliation(s)
- Yongjing Zhang
- Department of Medical Oncology, The First Affiliated Hospital, Xi'an Jiaotong University, Xi'an, China; School of Pharmacy, Xi'an Jiaotong University, Xi'an, China
| | - Wen Zhang
- School of Pharmacy, Xi'an Jiaotong University, Xi'an, China
| | - Mengyang Ma
- School of Pharmacy, Xi'an Jiaotong University, Xi'an, China
| | - Xinping Zhang
- School of Pharmacy, Xi'an Jiaotong University, Xi'an, China
| | - Chenjia Li
- School of Pharmacy, Xi'an Jiaotong University, Xi'an, China
| | - Tingting Deng
- School of Pharmacy, Xi'an Jiaotong University, Xi'an, China
| | - Jie Gao
- School of Pharmacy, Xi'an Jiaotong University, Xi'an, China
| | - Chang Gao
- School of Pharmacy, Xi'an Jiaotong University, Xi'an, China
| | - Nan Wang
- School of Pharmacy, Xi'an Jiaotong University, Xi'an, China.
| |
Collapse
|
2
|
Xie H, Zhang H, Chen D, Cheng L, Gu F, Wang S, Liu M, Li L, Zeng Q, He S. Increased CD123 + HLA-DR - Granulocytes in Allergic Rhinitis and Influence of Allergens on Expression of Cell Membrane Markers. Am J Rhinol Allergy 2024; 38:294-305. [PMID: 38715340 DOI: 10.1177/19458924241252456] [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] [Indexed: 09/06/2024]
Abstract
BACKGROUND It is reported that CD123 + HLA-DR- cells in PBMC are basophils, and CD203c, CD63, and FcεRI molecules are activation markers of basophils. However, little is known of CD123 + HLA-DR-cells in blood granulocytes. OBJECTIVE To investigate the presence of CD123 + HLA-DR- cells in the blood granulocytes and peripheral PBMC of patients with allergic rhinitis (AR), as well as the impact of allergens on the cell membrane markers of basophils. METHODS Flow cytometry was used to detect the expression of the membrane molecules. RESULTS While CD123 + HLA-DR- PBMCs are representative of basophils, their presence did not significantly change in patients with AR. In contrast, both the percentage and number of CD123 + HLA-DR- granulocytes, which make up only up to 50% of basophils, were significantly increased in patients with seasonal (sAR) and perennial AR (pAR). CD63+, CD203c+, and FcεRIα+ cells within CD123 + HLA-DR- granulocytes also showed enhanced activity in patients with AR. Allergen extracts from house dust mite allergen extract (HDME) and Artemisia sieversiana wild extract further increased the number of CD123 + HLA-DR- cells in granulocytes of sAR and pAR patients, as well as in PBMCs of pAR patients. CONCLUSIONS The use of CD123 + HLA-DR- granulocytes and PBMC may not be sufficient for diagnosing AR. Allergens could potentially contribute to the development of AR by influencing the number of CD123 + HLA-DR- cells, as well as the expression of CD63, CD203c, and FcεRIαin these cells.
Collapse
Affiliation(s)
- Hua Xie
- The Respiratory and Allergic Disease Diagnosing Management Center of the PLA. General Hospital of Northern Theater Command, Shenyang, Liaoning, China
| | - Huiyun Zhang
- Translational Medicine Research Centre, Shenyang Medical College, Shenyang, Liaoning, China
| | - Dong Chen
- Allergy and Clinical Immunology Research Centre, the First Affiliated Hospital of Jinzhou Medical University, Jinzhou, Liaoning, China
| | - Lei Cheng
- Department of Otorhinolaryngology & Clinical Allergy Center, the First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, China
| | - Fangqiu Gu
- Allergy and Clinical Immunology Research Centre, the First Affiliated Hospital of Jinzhou Medical University, Jinzhou, Liaoning, China
| | - Shunlan Wang
- Central Laboratory, Affiliated Haikou Hospital of Xiangya Medical College, Central South University, Haikou, Hainan, China
| | - Meicen Liu
- The Respiratory and Allergic Disease Diagnosing Management Center of the PLA. General Hospital of Northern Theater Command, Shenyang, Liaoning, China
| | - Li Li
- The Respiratory and Allergic Disease Diagnosing Management Center of the PLA. General Hospital of Northern Theater Command, Shenyang, Liaoning, China
| | - Qingwei Zeng
- The Respiratory and Allergic Disease Diagnosing Management Center of the PLA. General Hospital of Northern Theater Command, Shenyang, Liaoning, China
| | - Shaoheng He
- The Respiratory and Allergic Disease Diagnosing Management Center of the PLA. General Hospital of Northern Theater Command, Shenyang, Liaoning, China
- Translational Medicine Research Centre, Shenyang Medical College, Shenyang, Liaoning, China
| |
Collapse
|
3
|
Van der Borght K, Brimnes J, Haspeslagh E, Brand S, Neyt K, Gupta S, Knudsen NPH, Hammad H, Andersen PS, Lambrecht BN. Sublingual allergen immunotherapy prevents house dust mite inhalant type 2 immunity through dendritic cell-mediated induction of Foxp3 + regulatory T cells. Mucosal Immunol 2024; 17:618-632. [PMID: 38570140 DOI: 10.1016/j.mucimm.2024.03.012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2023] [Revised: 03/03/2024] [Accepted: 03/22/2024] [Indexed: 04/05/2024]
Abstract
Sublingual allergen immunotherapy (SLIT) is an emerging treatment option for allergic asthma and a potential disease-modifying strategy for asthma prevention. The key cellular events leading to such long-term tolerance remain to be fully elucidated. We administered prophylactic SLIT in a mouse model of house dust mite (HDM)-driven allergic asthma. HDM extract was sublingually administered over 3 weeks followed by intratracheal sensitization and intranasal challenges with HDM. Prophylactic SLIT prevented allergic airway inflammation and hyperreactivity with a low lab-to-lab variation. The HDM-specific T helper (Th)2 (cluster of differentiation 4 Th) response was shifted by SLIT toward a regulatory and Th17 response in the lung and mediastinal lymph node. By using Derp1-specific cluster of differentiation 4+ T cells (1-DER), we found that SLIT blocked 1-DER T cell recruitment to the mediastinal lymph node and dampened IL-4 secretion following intratracheal HDM sensitization. Sublingually administered Derp1 protein activated 1-DER T cells in the cervical lymph node via chemokine receptor7+ migratory dendritic cells (DC). DCs migrating from the oral submucosa to the cervical lymph node after SLIT-induced Foxp3+ regulatory T cells. When mice were sensitized with HDM, prior prophylactic SLIT increased Derp1 specific regulatory T cells (Tregs) and lowered Th2 recruitment in the lung. By using Foxp3-diphtheria toxin receptor mice, Tregs were found to contribute to the immunoregulatory prophylactic effect of SLIT on type 2 immunity. These findings in a mouse model suggest that DC-mediated functional Treg induction in oral mucosa draining lymph nodes is one of the driving mechanisms behind the disease-modifying effect of prophylactic SLIT.
Collapse
Affiliation(s)
- Katrien Van der Borght
- Laboratory of Immunoregulation and Mucosal Immunology, VIB-UGent Center for Inflammation Research, Ghent, Belgium; Department of Internal Medicine and Pediatrics, Ghent University, Ghent, Belgium
| | - Jens Brimnes
- Immunology Department, In vivo Biology Team, ALK Abelló A/S, Hørsholm, Denmark
| | - Eline Haspeslagh
- Laboratory of Immunoregulation and Mucosal Immunology, VIB-UGent Center for Inflammation Research, Ghent, Belgium; Department of Internal Medicine and Pediatrics, Ghent University, Ghent, Belgium
| | - Stephanie Brand
- Immunology Department, In vivo Biology Team, ALK Abelló A/S, Hørsholm, Denmark
| | - Katrijn Neyt
- Laboratory of Immunoregulation and Mucosal Immunology, VIB-UGent Center for Inflammation Research, Ghent, Belgium; Department of Internal Medicine and Pediatrics, Ghent University, Ghent, Belgium
| | - Shashank Gupta
- Immunology Department, In vivo Biology Team, ALK Abelló A/S, Hørsholm, Denmark
| | | | - Hamida Hammad
- Laboratory of Immunoregulation and Mucosal Immunology, VIB-UGent Center for Inflammation Research, Ghent, Belgium; Department of Internal Medicine and Pediatrics, Ghent University, Ghent, Belgium
| | - Peter S Andersen
- Immunology Department, In vivo Biology Team, ALK Abelló A/S, Hørsholm, Denmark
| | - Bart N Lambrecht
- Laboratory of Immunoregulation and Mucosal Immunology, VIB-UGent Center for Inflammation Research, Ghent, Belgium; Department of Internal Medicine and Pediatrics, Ghent University, Ghent, Belgium; Department of Pulmonary Medicine, Erasmus MC, Rotterdam, The Netherlands.
| |
Collapse
|
4
|
Park SY, Lee YY, Kim MH, Kim CE. Deciphering the Systemic Impact of Herbal Medicines on Allergic Rhinitis: A Network Pharmacological Approach. Life (Basel) 2024; 14:553. [PMID: 38792575 PMCID: PMC11122645 DOI: 10.3390/life14050553] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2024] [Revised: 04/18/2024] [Accepted: 04/23/2024] [Indexed: 05/26/2024] Open
Abstract
Allergic rhinitis (AR) is a systemic allergic disease that has a considerable impact on patients' quality of life. Current treatments include antihistamines and nasal steroids; however, their long-term use often causes undesirable side effects. In this context, traditional Asian medicine (TAM), with its multi-compound, multi-target herbal medicines (medicinal plants), offers a promising alternative. However, the complexity of these multi-compound traits poses challenges in understanding the overall mechanisms and efficacy of herbal medicines. Here, we demonstrate the efficacy and underlying mechanisms of these multi-compound herbal medicines specifically used for AR at a systemic level. We utilized a modified term frequency-inverse document frequency method to select AR-specific herbs and constructed an herb-compound-target network using reliable databases and computational methods, such as the Quantitative Estimate of Drug-likeness for compound filtering, STITCH database for compound-target interaction prediction (with a high confidence score threshold of 0.7), and DisGeNET and CTD databases for disease-gene association analysis. Through this network, we conducted AR-related targets and pathway analyses, as well as clustering analysis based on target-level information of the herbs. Gene ontology enrichment analysis was conducted using a protein-protein interaction network. Our research identified 14 AR-specific herbs and analyzed whether AR-specific herbs are highly related to previously known AR-related genes and pathways. AR-specific herbs were found to target several genes related to inflammation and AR pathogenesis, such as PTGS2, HRH1, and TBXA2R. Pathway analysis revealed that AR-specific herbs were associated with multiple AR-related pathways, including cytokine signaling, immune response, and allergic inflammation. Additionally, clustering analysis based on target similarity identified three distinct subgroups of AR-specific herbs, corroborated by a protein-protein interaction network. Group 1 herbs were associated with the regulation of inflammatory responses to antigenic stimuli, while Group 2 herbs were related to the detection of chemical stimuli involved in the sensory perception of bitter taste. Group 3 herbs were distinctly associated with antigen processing and presentation and NIK/NF-kappa B signaling. This study decodes the principles of TAM herbal configurations for AR using a network pharmacological approach, providing a holistic understanding of drug effects beyond specific pathways.
Collapse
Affiliation(s)
- Sa-Yoon Park
- Department of Physiology, College of Korean Medicine, Gachon University, Seongnam 13120, Republic of Korea
- Biomedical Research Institute, Seoul National University Hospital, Seoul 03080, Republic of Korea
| | - Yoon Yeol Lee
- Department of Physiology, College of Korean Medicine, Gachon University, Seongnam 13120, Republic of Korea
| | - Min Hee Kim
- Department of Ophthalmology, Otolaryngology, and Dermatology, Kyung Hee University College of Korean Medicine, Kyung Hee University Hospital at Gangdong, Seoul 05278, Republic of Korea
| | - Chang-Eop Kim
- Department of Physiology, College of Korean Medicine, Gachon University, Seongnam 13120, Republic of Korea
| |
Collapse
|
5
|
Xie H, Chen L, Zhang H, Wang J, Zang Y, Zhan M, Gu F, Wang S, He S. Increased expressions of CD123, CD63, CD203c, and Fc epsilon receptor I on blood leukocytes of allergic asthma. Front Mol Biosci 2022; 9:907092. [PMID: 36032674 PMCID: PMC9403487 DOI: 10.3389/fmolb.2022.907092] [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: 03/29/2022] [Accepted: 06/27/2022] [Indexed: 11/28/2022] Open
Abstract
Background : Altered basophil identification markers have been discovered to associate with allergic asthma (AA) in recent years. However, little is known about the expression of basophil markers in blood granulocytes. Aim: To parallel test blood basophils in peripheral blood mononuclear cell (PBMC) and granulocyte populations of patients with AA and AA combined with allergic rhinitis (ARA) Methods: The expressions of surface molecules were determined via flow cytometry. CD123 expressing cells in blood were isolated using a cell sorting technique, and mouse AA models were employed for in vivo study. Results: The numbers of CD123+HLA-DR- cells in the granulocytes of AA and ARA patients markedly increased. However, only 49.7% of CD123+HLA-DR- cells in granulocytes and 99.0% of CD123+HLA-DR- cells in PBMCs were basophils. Almost all CD123+HLA-DR- cells expressed CD63 regardless in granulocytes or PBMC. The numbers of CD63, Fc epsilon receptor I (FcεRI), and CD203c expressing cells markedly enhanced in CD123+HLA-DR- granulocytes of AA and ARA patients. Mean fluorescence intensity (MFI) of CD63 and CD203c expressions on CD123+HLA-DR- PBMC and granulocytes of AA and ARA patients dramatically elevated. House dust mite extract (HDME) and Artemisia sieversiana wild allergen extract (ASWE) enhanced the numbers of CD63+CD123+HLA-DR- granulocytes and PBMC and the MFI of CD203c expression on CD123+HLA-DR- granulocyte of AA and ARA patients. Histamine, tryptase, and PGD2 enhanced proportions of CD123+ KU812 cells. ASWE- and HDME-induced AA mice showed upregulated CD63 expression on basophils. In conclusion, upregulated expressions of CD123, CD203c, CD63, and FcεRIα in PBMC and granulocytes of patients with AA and ARA suggest that CD123+HLA-DR- cells may contribute to the development of AA and ARA.
Collapse
Affiliation(s)
- Hua Xie
- The PLA Center of Respiratory and Allergic Disease Diagnosing Management, General Hospital of Northern Theater Command, Shenyang, China
- Allergy and Clinical Immunology Research Centre, The First Affiliated Hospital of Jinzhou Medical University, Jinzhou, China
| | - Liping Chen
- Translational Medicine Institute, Shenyang Medical College, Shenyang, China
- Department of Respiratory and Critical Care Medicine, The Second Hospital of Senyang Medical College, Shenyang, China
| | - Huiyun Zhang
- Translational Medicine Institute, Shenyang Medical College, Shenyang, China
| | - Junling Wang
- Allergy and Clinical Immunology Research Centre, The First Affiliated Hospital of Jinzhou Medical University, Jinzhou, China
| | - Yanyan Zang
- Allergy and Clinical Immunology Research Centre, The First Affiliated Hospital of Jinzhou Medical University, Jinzhou, China
| | - Mengmeng Zhan
- Translational Medicine Institute, Shenyang Medical College, Shenyang, China
| | - Fangqiu Gu
- Allergy and Clinical Immunology Research Centre, The First Affiliated Hospital of Jinzhou Medical University, Jinzhou, China
| | - Shunlan Wang
- Central Laboratory, Affiliated Haikou Hospital of Xiangya Medical College, Central South University, Haikou, China
| | - Shaoheng He
- The PLA Center of Respiratory and Allergic Disease Diagnosing Management, General Hospital of Northern Theater Command, Shenyang, China
- Allergy and Clinical Immunology Research Centre, The First Affiliated Hospital of Jinzhou Medical University, Jinzhou, China
| |
Collapse
|
6
|
Nur Husna SM, Tan HTT, Md Shukri N, Mohd Ashari NS, Wong KK. Allergic Rhinitis: A Clinical and Pathophysiological Overview. Front Med (Lausanne) 2022; 9:874114. [PMID: 35463011 PMCID: PMC9021509 DOI: 10.3389/fmed.2022.874114] [Citation(s) in RCA: 76] [Impact Index Per Article: 25.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2022] [Accepted: 03/15/2022] [Indexed: 01/25/2023] Open
Abstract
Allergic rhinitis (AR) represents a global health concern where it affects approximately 400 million people worldwide. The prevalence of AR has increased over the years along with increased urbanization and environmental pollutants thought to be some of the leading causes of the disease. Understanding the pathophysiology of AR is crucial in the development of novel therapies to treat this incurable disease that often comorbids with other airway diseases. Hence in this mini review, we summarize the well-established yet vital aspects of AR. These include the epidemiology, clinical and laboratory diagnostic criteria, AR in pediatrics, pathophysiology of AR, Th2 responses in the disease, as well as pharmacological and immunomodulating therapies for AR patients.
Collapse
Affiliation(s)
- Siti Muhamad Nur Husna
- Department of Immunology, School of Medical Sciences, Universiti Sains Malaysia, Kubang Kerian, Malaysia
| | - Hern-Tze Tina Tan
- Department of Immunology, School of Medical Sciences, Universiti Sains Malaysia, Kubang Kerian, Malaysia
| | - Norasnieda Md Shukri
- Department of Otorhinolaryngology, Head and Neck Surgery, School of Medical Sciences, Universiti Sains Malaysia, Kubang Kerian, Malaysia
- Hospital Universiti Sains Malaysia, Kubang Kerian, Malaysia
| | - Noor Suryani Mohd Ashari
- Department of Immunology, School of Medical Sciences, Universiti Sains Malaysia, Kubang Kerian, Malaysia
| | - Kah Keng Wong
- Department of Immunology, School of Medical Sciences, Universiti Sains Malaysia, Kubang Kerian, Malaysia
| |
Collapse
|
7
|
Li Y, Liu C, Li H, Wang X. Exploring the role of basophil activation test in diagnosis of Dermatophagoides farinae sensitization and evaluation of therapeutic efficacy of subcutaneous immunotherapy in children. Scand J Immunol 2022; 96:e13168. [PMID: 35366340 DOI: 10.1111/sji.13168] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2021] [Revised: 03/28/2022] [Accepted: 03/29/2022] [Indexed: 11/29/2022]
Abstract
OBJECTIVE It aims to detect basophil activation ratio (%CD63+ ) in peripheral blood of children with allergic asthma and rhinitis by using Flow cytometry (FCM) , so as to analyze the application values and clinical relevance of the Basophil Activation Test (BAT) in diagnosis of Dermatophagoides farinae (Derf) sensitization and monitoring therapeutic efficacy of subcutaneous immunotherapy (SCIT). METHODS It was a prospective study. From the newly diagnosed children with asthma and rhinitis in our pediatric clinic, 39 patients diagnosed Derf sensitization and 15 patients not allergic to Derf were enrolled; another 4 healthy children were taken as control group. Using Derf extracts in concentration of 1 μg/ml, 10 μg/ml and 100 μg/ml as the stimulus, BAT results were expressed as %CD63+ in diagnosis of Derf sensitization and its correlation with skin prick tests (SPT), serum total IgE (tIgE), specific IgE (sIgE), sIgE/tIgE, specific IgG4 (sIgG4), FEV1%pred in pulmonary ventilation function, exhaled nitric oxide (FeNO), children asthma control test (C-ACT) and visual analogue scale (VAS) were observed. In sensitization group, %CD63+ , sIgG4 and clinical indicators were detected again from patients who had received SCIT to analyze their internal connections. RESULTS The average levels of %CD63+ in three concentrations showed an increasing concentration-dependent trend overall. %CD63+ in sensitization group was significantly higher than that in the other two groups. The analysis of ROC for Derf sensitization showed the area under the curve (AUC) for BAT in three concentrations were higher than that for sIgE whose AUC is 0.893. %CD63+ was positively correlated with SPT grade, sIgE, sIgE/tIgE and VAS, and negatively correlated with C-ACT. In patients receiving SCIT, %CD63+ became lower and sIgG4 level became higher than pretreatment. There was no obvious change in sIgG4 in those who hadn't received SCIT. CONCLUSIONS BAT is a reliable and non-invasive tool for diagnosis of Derf sensitization in children with asthma and rhinitis. CD63-based BAT has clinical value to monitor outcome of SCIT, and the change of basophil activation is inherently related to induction of sIgG4.
Collapse
Affiliation(s)
- Yifan Li
- Department of pediatrics, Second Hospital of Tianjin Medical University, Tianjin, China, 300211
| | - Changshan Liu
- Department of pediatrics, Second Hospital of Tianjin Medical University, Tianjin, China, 300211
| | - Huiqiang Li
- College of Medical Laboratory Science, Tianjin Medical University, Tianjin, China, 300203
| | - Xueyan Wang
- Department of pediatrics, Second Hospital of Tianjin Medical University, Tianjin, China, 300211
| |
Collapse
|
8
|
Lu Y, Peng B, Lin Y, Lin Q, Xia X, Zhong S, Luo L, Huang R. Spirulina polysaccharide induces the metabolic shifts and gut microbiota change of lung cancer in mice. Curr Res Food Sci 2022; 5:1313-1319. [PMID: 36065196 PMCID: PMC9440268 DOI: 10.1016/j.crfs.2022.08.010] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2022] [Revised: 08/15/2022] [Accepted: 08/18/2022] [Indexed: 02/07/2023] Open
|
9
|
Yang T, Li C, Xue W, Huang L, Wang Z. Natural immunomodulating substances used for alleviating food allergy. Crit Rev Food Sci Nutr 2021; 63:2407-2425. [PMID: 34494479 DOI: 10.1080/10408398.2021.1975257] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
Abstract
Food allergy is a serious health problem affecting more than 10% of the human population worldwide. Medical treatments for food allergy remain limited because immune therapy is risky and costly, and anti-allergic drugs have many harmful side effects and can cause drug dependence. In this paper, we review natural bioactive substances capable of alleviating food allergy. The sources of the anti-allergic substances reviewed include plants, animals, and microbes, and the types of substances include polysaccharides, oligosaccharides, polyphenols, phycocyanin, polyunsaturated fatty acids, flavonoids, terpenoids, quinones, alkaloids, phenylpropanoids, and probiotics. We describe five mechanisms involved in anti-allergic activities, including binding with epitopes located in allergens, affecting the gut microbiota, influencing intestinal epithelial cells, altering antigen presentation and T cell differentiation, and inhibiting the degranulation of effector cells. In the discussion, we present the limitations of existing researches as well as promising advances in the development of anti-allergic foods and/or immunomodulating food ingredients that can effectively prevent or alleviate food allergy. This review provides a reference for further research on anti-allergic materials and their hyposensitizing mechanisms.
Collapse
Affiliation(s)
- Tian Yang
- Key Laboratory of Glycobiology and Glycoengineering of Xi'an, College of Food Science and Technology, Northwest University, Xi'an, China
| | - Cheng Li
- Key Laboratory of Glycobiology and Glycoengineering of Xi'an, College of Food Science and Technology, Northwest University, Xi'an, China
| | - Wentong Xue
- College of Food Science and Nutritional Engineering, China Agricultural University, Beijing, People's Republic of China
| | - Linjuan Huang
- Key Laboratory of Glycobiology and Glycoengineering of Xi'an, College of Food Science and Technology, Northwest University, Xi'an, China
| | - Zhongfu Wang
- Key Laboratory of Glycobiology and Glycoengineering of Xi'an, College of Food Science and Technology, Northwest University, Xi'an, China
| |
Collapse
|
10
|
Xue Z, Zhang Y, Zeng Y, Hu S, Bai H, Wang J, Jing H, Wang N. Licochalcone A inhibits MAS-related GPR family member X2-induced pseudo-allergic reaction by suppressing nuclear migration of nuclear factor-κB. Phytother Res 2021; 35:6270-6280. [PMID: 34486187 DOI: 10.1002/ptr.7272] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2021] [Revised: 07/02/2021] [Accepted: 08/24/2021] [Indexed: 12/19/2022]
Abstract
Licochalcone A (Lico A) is a natural flavonoid belonging to the class of substituted chalcone that has various biological effects. Mast cells (MCs) are innate immune cells that mediate hypersensitivity and pseudo-allergic reactions. MAS-related GPR family member X2 (MRGPRX2) on MCs has been recognized as the main receptor for pseudo-allergic reactions. In this study, we investigated the anti-pseudo-allergy effect of Lico A and its underlying mechanism. Substance P (SP), as an MC activator, was used to establish an in vitro and in vivo model of pseudo-allergy. The in vivo effect of Lico A was investigated using passive cutaneous anaphylaxis (PCA) and active systemic allergy, along with degranulation, Ca2+ influx in vitro. SP-induced laboratory of allergic disease 2 (LAD2) cell mRNA expression was explored using RNA-seq, and Lico A inhibited LAD2 cell activation by reverse transcription polymerase chain reaction (RT-PCR), western blotting, and immunofluorescence staining. Lico A showed an inhibitory effect on SP-induced MC activation and pseudo-allergy both in vitro and in vivo. The nuclear factor (NF)-κB pathway is involved in MRGPRX2 induced MC activation, which is inhibited by Lico A. In conclusion, Lico A inhibited the pseudo-allergic reaction mediated by MRGPRX2 by blocking NF-κB nuclear migration.
Collapse
Affiliation(s)
- Zhuoyin Xue
- School of Pharmacy, Xi'an Jiaotong University, Xi'an, China
| | - Yongjing Zhang
- School of Pharmacy, Xi'an Jiaotong University, Xi'an, China
| | - Yingnan Zeng
- School of Pharmacy, Xi'an Jiaotong University, Xi'an, China
| | - Shiling Hu
- School of Pharmacy, Xi'an Jiaotong University, Xi'an, China
| | - Haoyun Bai
- School of Pharmacy, Xi'an Jiaotong University, Xi'an, China
| | - Jue Wang
- School of Pharmacy, Xi'an Jiaotong University, Xi'an, China
| | - Huiling Jing
- Xi'an Hospital of Traditional Chinese Medicine, Xi'an, China
| | - Nan Wang
- School of Pharmacy, Xi'an Jiaotong University, Xi'an, China.,Pazhou Lab, Guangzhou, China
| |
Collapse
|
11
|
Palikhe NS, Gandhi VD, Wu Y, Sinnatamby T, Rowe BH, Mayers I, Cameron L, Vliagoftis H. Peripheral blood intermediate monocyte protease-activated receptor-2 expression increases during asthma exacerbations and after inhalation allergen challenge. Ann Allergy Asthma Immunol 2021; 127:249-256.e2. [PMID: 33895420 DOI: 10.1016/j.anai.2021.04.016] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2020] [Revised: 04/13/2021] [Accepted: 04/16/2021] [Indexed: 10/21/2022]
Abstract
BACKGROUND Myeloid cells, especially dendritic cells and macrophages, play important roles in asthma pathophysiology. Monocytes (Mo) and macrophages express protease-activated receptor-2 (PAR-2), a proinflammatory serine protease receptor implicated in the pathophysiology of allergic airway inflammation. We have revealed that patients with severe asthma and those with a history of frequent asthma exacerbations exhibit increased PAR-2 expression on peripheral blood monocytes. OBJECTIVE To determine PAR-2 expression on peripheral blood intermediate monocytes (IMMo) in subjects with increased airway inflammation, either as a result of an asthma exacerbation or after an inhalation allergen challenge. METHODS A total of 16 adults who presented to the emergency department with asthma exacerbations were recruited after giving an informed consent. After 2 weeks, 10 patients returned for follow-up. A total of 11 patients with mild asthma treated only with as-needed bronchodilators were recruited and underwent inhalation allergen challenge after providing an informed consent. Immune cell profiling was performed by whole blood flow cytometry in both groups of patients. RESULTS PAR-2 expression in peripheral blood IMMo increased in patients with an asthma exacerbation compared with those with stable disease, but this expression decreased after treatment of the asthma exacerbation. Subjects with mild asthma had an increase in percentages of IMMo expressing PAR-2 after an allergen challenge. Patients who presented to the emergency department had lower dendritic cell and dendritic cell subset numbers in peripheral blood during exacerbation compared with after treatment. CONCLUSION Increased PAR-2 expression on Mo during periods of increased airway inflammation may initiate a positive feedback loop leading to systemic inflammatory changes.
Collapse
Affiliation(s)
- Nami Shrestha Palikhe
- Division of Pulmonary Medicine, Department of Medicine, Faculty of Medicine & Dentistry, University of Alberta, Edmonton, Alberta, Canada; Alberta Respiratory Centre, University of Alberta, Edmonton, Alberta, Canada
| | - Vivek Dipak Gandhi
- Division of Pulmonary Medicine, Department of Medicine, Faculty of Medicine & Dentistry, University of Alberta, Edmonton, Alberta, Canada; Division of Allergy, Pulmonary and Critical Care Medicine, Vanderbilt University, Nashville, Tennessee
| | - Yingqi Wu
- Division of Pulmonary Medicine, Department of Medicine, Faculty of Medicine & Dentistry, University of Alberta, Edmonton, Alberta, Canada; Alberta Respiratory Centre, University of Alberta, Edmonton, Alberta, Canada
| | - Tristan Sinnatamby
- Division of Pulmonary Medicine, Department of Medicine, Faculty of Medicine & Dentistry, University of Alberta, Edmonton, Alberta, Canada
| | - Brian H Rowe
- Alberta Respiratory Centre, University of Alberta, Edmonton, Alberta, Canada; Department of Emergency Medicine, Faculty of Medicine & Dentistry, University of Alberta, Edmonton, Alberta, Canada; School of Public Health, University of Alberta, Edmonton, Alberta, Canada
| | - Irvin Mayers
- Division of Pulmonary Medicine, Department of Medicine, Faculty of Medicine & Dentistry, University of Alberta, Edmonton, Alberta, Canada; Alberta Respiratory Centre, University of Alberta, Edmonton, Alberta, Canada
| | - Lisa Cameron
- Division of Pulmonary Medicine, Department of Medicine, Faculty of Medicine & Dentistry, University of Alberta, Edmonton, Alberta, Canada; Department of Pathology and Laboratory Medicine, Schulich School of Medicine & Dentistry, Western University, London, Ontario, Canada
| | - Harissios Vliagoftis
- Division of Pulmonary Medicine, Department of Medicine, Faculty of Medicine & Dentistry, University of Alberta, Edmonton, Alberta, Canada; Alberta Respiratory Centre, University of Alberta, Edmonton, Alberta, Canada.
| |
Collapse
|
12
|
Bachert C, Maurer M, Palomares O, Busse WW. What is the contribution of IgE to nasal polyposis? J Allergy Clin Immunol 2021; 147:1997-2008. [PMID: 33757720 DOI: 10.1016/j.jaci.2021.03.016] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2020] [Revised: 03/10/2021] [Accepted: 03/17/2021] [Indexed: 02/07/2023]
Abstract
Taking a novel approach, this narrative review collates knowledge about nasal polyposis and the biological functions of IgE in several diseases (allergic rhinitis, allergic asthma, nonsteroidal anti-inflammatory drugs-exacerbated respiratory disease, and chronic spontaneous urticaria) to consider which IgE-mediated mechanisms are relevant to nasal polyposis pathology. A type 2 eosinophil-dominated inflammatory signature is typical in nasal polyp tissue of European patients with nasal polyposis, with a shift toward this endotype observed in Asian populations in recent years. Elevated polyclonal IgE is present in the nasal tissue of patients with and without allergy. It is derived from many different B-cell clones and, importantly, is functional (proinflammatory). Staphylococcus aureus enterotoxins are thought to act as superantigens, inducing production of polyclonal IgE via B-cell and T-cell activation, and triggering release of inflammatory mediators. In some patients, exposure to antigens/triggers leads to production of high levels of antigen-specific IgE, which mediates cross-linking of the high-affinity IgE receptor on various cells, causing release of inflammatory mediators. The efficacy of omalizumab confirms IgE as an important inflammatory mediator in nasal polyposis. By blocking IgE, omalizumab targets the T2 inflammation in nasal polyposis, reduces nasal polyp score and improves symptoms.
Collapse
Affiliation(s)
- Claus Bachert
- Upper Airways Research Laboratory, Ghent University, Ghent, Belgium; Division of ENT Diseases, CLINTEC, Karolinska Institute, Stockholm, Sweden.
| | - Marcus Maurer
- Dermatological Allergology, Allergie-Centrum-Charité, Department of Dermatology and Allergy, Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Oscar Palomares
- Department of Biochemistry and Molecular Biology, School of Chemistry, Complutense University of Madrid, Madrid, Spain
| | - William W Busse
- Department of Medicine, Division of Allergy, Pulmonary and Critical Care Medicine, University of Wisconsin School of Medicine and Public Health, Madison, Wis
| |
Collapse
|
13
|
Lamri Y, Vibhushan S, Pacreau E, Boedec E, Saidoune F, Mailleux A, Crestani B, Blank U, Benhamou M, Papo T, Daugas E, Sacré K, Charles N. Basophils and IgE contribute to mixed connective tissue disease development. J Allergy Clin Immunol 2020; 147:1478-1489.e11. [PMID: 33338538 DOI: 10.1016/j.jaci.2020.12.622] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2020] [Revised: 10/20/2020] [Accepted: 12/01/2020] [Indexed: 12/18/2022]
Abstract
BACKGROUND Mixed connective tissue disease (MCTD) is a rare and complex autoimmune disease that presents mixed features with other connective tissue diseases, such as systemic lupus erythematosus, systemic sclerosis, and myositis. It is characterized by high levels of anti-U1 small nuclear ribonucleoprotein 70k autoantibodies and a high incidence of life-threatening pulmonary involvement. The pathophysiology of MCTD is not well understood, and no specific treatment is yet available for the patients. Basophils and IgE play a role in the development of systemic lupus erythematosus and thus represent new therapeutic targets for systemic lupus erythematosus and other diseases involving basophils and IgE in their pathogenesis. OBJECTIVE We sought to investigate the role of basophils and IgE in the pathophysiology of MCTD. METHODS Basophil activation status and the presence of autoreactive IgE were assessed in peripheral blood of a cohort of patients with MCTD and in an MCTD-like mouse model. Basophil depletion and IgE-deficient animals were used to investigate the contribution of basophils and IgE in the lung pathology development of this mouse model. RESULTS Patients with MCTD have a peripheral basopenia and activated blood basophils overexpressing C-C chemokine receptor 3. Autoreactive IgE raised against the main MCTD autoantigen U1 small nuclear ribonucleoprotein 70k were found in nearly 80% of the patients from the cohort. Basophil activation and IgE anti-U1 small nuclear ribonucleoprotein 70k were also observed in the MCTD-like mouse model along with basophil accumulation in lymph nodes and lungs. Basophil depletion dampened lung pathology, and IgE deficiency prevented its development. CONCLUSIONS Basophils and IgE contribute to MCTD pathophysiology and represent new candidate therapeutic targets for patients with MCTD.
Collapse
Affiliation(s)
- Yasmine Lamri
- Université de Paris, Centre de Recherche sur l'Inflammation, INSERM UMR1149, CNRS ERL8252, Faculté de Médecine site Bichat, Paris, France; Université de Paris, Laboratoire d'Excellence Inflamex, Paris, France
| | - Shamila Vibhushan
- Université de Paris, Centre de Recherche sur l'Inflammation, INSERM UMR1149, CNRS ERL8252, Faculté de Médecine site Bichat, Paris, France; Université de Paris, Laboratoire d'Excellence Inflamex, Paris, France
| | - Emeline Pacreau
- Université de Paris, Centre de Recherche sur l'Inflammation, INSERM UMR1149, CNRS ERL8252, Faculté de Médecine site Bichat, Paris, France; Université de Paris, Laboratoire d'Excellence Inflamex, Paris, France
| | - Erwan Boedec
- Université de Paris, Centre de Recherche sur l'Inflammation, INSERM UMR1149, CNRS ERL8252, Faculté de Médecine site Bichat, Paris, France; Université de Paris, Laboratoire d'Excellence Inflamex, Paris, France
| | - Fanny Saidoune
- Université de Paris, Centre de Recherche sur l'Inflammation, INSERM UMR1149, CNRS ERL8252, Faculté de Médecine site Bichat, Paris, France; Université de Paris, Laboratoire d'Excellence Inflamex, Paris, France
| | - Arnaud Mailleux
- Université de Paris, Laboratoire d'Excellence Inflamex, Paris, France; Université de Paris, INSERM UMR1152, Faculté de Médecine site Bichat, Paris, France
| | - Bruno Crestani
- Université de Paris, Laboratoire d'Excellence Inflamex, Paris, France; Université de Paris, INSERM UMR1152, Faculté de Médecine site Bichat, Paris, France; Department of Pulmonology, Hôpital Bichat, Assistance Publique-Hôpitaux de Paris, Université de Paris, Faculté de Médecine site Bichat, DHU FIRE, Paris, France
| | - Ulrich Blank
- Université de Paris, Centre de Recherche sur l'Inflammation, INSERM UMR1149, CNRS ERL8252, Faculté de Médecine site Bichat, Paris, France; Université de Paris, Laboratoire d'Excellence Inflamex, Paris, France
| | - Marc Benhamou
- Université de Paris, Centre de Recherche sur l'Inflammation, INSERM UMR1149, CNRS ERL8252, Faculté de Médecine site Bichat, Paris, France; Université de Paris, Laboratoire d'Excellence Inflamex, Paris, France
| | - Thomas Papo
- Université de Paris, Centre de Recherche sur l'Inflammation, INSERM UMR1149, CNRS ERL8252, Faculté de Médecine site Bichat, Paris, France; Université de Paris, Laboratoire d'Excellence Inflamex, Paris, France; Department of Internal Medicine, Hôpital Bichat, Assistance Publique-Hôpitaux de Paris, Université de Paris, Faculté de Médecine site Bichat, DHU FIRE, Paris, France
| | - Eric Daugas
- Université de Paris, Centre de Recherche sur l'Inflammation, INSERM UMR1149, CNRS ERL8252, Faculté de Médecine site Bichat, Paris, France; Université de Paris, Laboratoire d'Excellence Inflamex, Paris, France; Department of Nephrology, Hôpital Bichat, Assistance Publique-Hôpitaux de Paris, Université de Paris, Faculté de Médecine site Bichat, DHU FIRE, Paris, France
| | - Karim Sacré
- Université de Paris, Centre de Recherche sur l'Inflammation, INSERM UMR1149, CNRS ERL8252, Faculté de Médecine site Bichat, Paris, France; Université de Paris, Laboratoire d'Excellence Inflamex, Paris, France; Department of Internal Medicine, Hôpital Bichat, Assistance Publique-Hôpitaux de Paris, Université de Paris, Faculté de Médecine site Bichat, DHU FIRE, Paris, France
| | - Nicolas Charles
- Université de Paris, Centre de Recherche sur l'Inflammation, INSERM UMR1149, CNRS ERL8252, Faculté de Médecine site Bichat, Paris, France; Université de Paris, Laboratoire d'Excellence Inflamex, Paris, France.
| |
Collapse
|
14
|
Drazdauskaitė G, Layhadi JA, Shamji MH. Mechanisms of Allergen Immunotherapy in Allergic Rhinitis. Curr Allergy Asthma Rep 2020; 21:2. [PMID: 33313967 PMCID: PMC7733588 DOI: 10.1007/s11882-020-00977-7] [Citation(s) in RCA: 85] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/24/2020] [Indexed: 12/28/2022]
Abstract
PURPOSE OF REVIEW Allergic rhinitis (AR) is a chronic inflammatory immunoglobulin (Ig) E-mediated disease of the nasal mucosa that can be triggered by the inhalation of seasonal or perennial allergens. Typical symptoms include sneezing, rhinorrhea, nasal itching, nasal congestion and symptoms of allergic conjunctivitis. AR affects a quarter of the population in the United States of America and Europe. RECENT FINDINGS AR has been shown to reduce work productivity in 36-59% of the patients with 20% reporting deteriorated job attendance. Moreover, 42% of children with AR report reduced at-school productivity and lower grades. Most importantly, AR impacts the patient's quality of life, due to sleep deprivation. However, a proportion of patients fails to respond to conventional medication and opts for the allergen immunotherapy (AIT), which currently is the only disease-modifying therapeutic option. AIT can be administered by either subcutaneous (SCIT) or sublingual (SLIT) route. Both routes of administration are safe, effective, and can lead to tolerance lasting years after treatment cessation. Both innate and adaptive immune responses that contribute to allergic inflammation are suppressed by AIT. Innate responses are ameliorated by reducing local mast cell, basophil, eosinophil, and circulating group 2 innate lymphoid cell frequencies which is accompanied by decreased basophil sensitivity. Induction of allergen-specific blocking antibodies, immunosuppressive cytokines, and regulatory T and B cell phenotypes are key pro-tolerogenic adaptive immune responses. CONCLUSION A comprehensive understanding of these mechanisms is necessary for optimal selection of AIT-responsive patients and monitoring treatment efficacy. Moreover, it could inspire novel and more efficient AIT approaches.
Collapse
Affiliation(s)
- Gabija Drazdauskaitė
- Immunomodulation and Tolerance Group, Allergy & Clinical Immunology, Inflammation, Repair and Development, National Heart & Lung Institute, Imperial College London, 1st Floor, Room 111, Sir Alexander Fleming Building, South Kensington Campus, London, SW7 2AZ, UK
| | - Janice A Layhadi
- Immunomodulation and Tolerance Group, Allergy & Clinical Immunology, Inflammation, Repair and Development, National Heart & Lung Institute, Imperial College London, 1st Floor, Room 111, Sir Alexander Fleming Building, South Kensington Campus, London, SW7 2AZ, UK
| | - Mohamed H Shamji
- Immunomodulation and Tolerance Group, Allergy & Clinical Immunology, Inflammation, Repair and Development, National Heart & Lung Institute, Imperial College London, 1st Floor, Room 111, Sir Alexander Fleming Building, South Kensington Campus, London, SW7 2AZ, UK.
| |
Collapse
|
15
|
Yang R, Wang G, Li L, He H, Zheng M, Lu L, Wu S. Tespa1 plays a role in the modulation of airway hyperreactivity through the IL-4/STAT6 pathway. J Transl Med 2020; 18:444. [PMID: 33228696 PMCID: PMC7685668 DOI: 10.1186/s12967-020-02621-4] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2020] [Accepted: 11/18/2020] [Indexed: 01/11/2023] Open
Abstract
BACKGROUND Thymocyte-expressed, positive selection-associated 1 (Tespa1) is a critical signaling molecule in thymocyte development. This study aimed to investigate the regulatory effect of Tespa1 on mast cells in the pathogenesis of asthma and its relationship with the interleukin (IL)-4/signal transducers and activators of transcription 6 (STAT6) signaling pathway. METHODS Tespa1 mRNA expression analysis and IgE levels were carried out using the induced sputum of 33 adults with stable asthma and 36 healthy controls. Tespa1-knockout mice (Tespa1-/-, KO) and C57BL/6 background (wild-type, WT) mice were sensitized and treated with ovalbumin (OVA) to establish an asthma model. Pathological changes, number and activity of mast cells, and changes in activation of the IL-4/STAT6 pathway in lung tissue were detected. The changes of tryptase expression and STAT6 activation after mast cell gene knockout were analyzed in vitro. The changes of enzyme expression and STAT6 activation after mast cell gene knockout were analyzed in vitro. The association between the Tespa1 and p-STAT6 was analyzed by co-immunoprecipitation method. RESULTS Compared with the healthy controls, Tespa1 expression was decreased, and IgE levels were elevated in the sputum of asthmatic patients. Animal experiments showed that Tespa1-/- mice exhibited more severe inflammation, higher quantity of goblet cells and mast cells in the bronchium, and greater expression of mast cell tryptase, which is induced by ovalbumin, than WT mice. And IL-4, IL-13, phospho-Janus kinase 1, and p-STAT6 expressions presented a higher increase in the Tespa1-/- mouse model than in the WT mouse model. Further in vitro studies confirmed that IL-4 could more significantly promote tryptase and p-STAT6 activities in Tespa1-/- mast cells than their WT counterparts. Correlation analysis results showed a negative correlation between Tespa1 and p-STAT6. Co-immunoprecipitation results demonstrated an association between Tespa1 and p-STAT6. CONCLUSIONS Altogether, our results indicate that Tespa1 can negatively regulate mast cell activity, and this event is related to the mast cell IL-4/STAT6 signaling pathway and could be therapeutically exploited to treat asthma attacks.
Collapse
Affiliation(s)
- Ruhui Yang
- Department of Pharmacology, College of Medicine and Health, Lishui University, No. 1 Xueyuan Road, Liandu District, Lishui, 323000, China
| | - Guangli Wang
- College of Medicine and Health, Lishui University, No. 1 Xueyuan Road, Liandu District, Lishui, 323000, China
| | - Lingyun Li
- Clinical Laboratory, Lishui People's Hospital, Lishui, 323000, China
| | - Hanjiang He
- College of Medicine and Health, Lishui University, No. 1 Xueyuan Road, Liandu District, Lishui, 323000, China
| | - Mingzhu Zheng
- Program in Molecular and Cellular Biology, Zhejiang University School of Medicine, Hangzhou, 310058, China
| | - Linrong Lu
- Program in Molecular and Cellular Biology, Zhejiang University School of Medicine, Hangzhou, 310058, China
| | - Songquan Wu
- College of Medicine and Health, Lishui University, No. 1 Xueyuan Road, Liandu District, Lishui, 323000, China.
| |
Collapse
|
16
|
Rowe RK, Pyle DM, Farrar JD, Gill MA. IgE-mediated regulation of IL-10 and type I IFN enhances rhinovirus-induced Th2 differentiation by primary human monocytes. Eur J Immunol 2020; 50:1550-1559. [PMID: 32383224 DOI: 10.1002/eji.201948396] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2019] [Revised: 03/18/2020] [Accepted: 05/04/2020] [Indexed: 11/08/2022]
Abstract
Rhinovirus (RV) infections are linked to the development and exacerbation of allergic diseases including allergic asthma. IgE, another contributor to atopic disease pathogenesis, has been shown to regulate DC antiviral functions and influence T cell priming by monocytes. We previously demonstrated that IgE-mediated stimulation of monocytes alters multiple cellular functions including cytokine secretion, phagocytosis, and influenza-induced Th1 development. In this study, we investigate the effects of IgE-mediated stimulation on monocyte-driven, RV-induced T cell development utilizing primary human monocyte-T cell co-cultures. We demonstrate that IgE crosslinking of RV-exposed monocytes enhances monocyte-driven Th2 differentiation. This increase in RV-induced Th2 development was regulated by IgE-mediated inhibition of virus-induced type I IFN and induction of IL-10. These findings suggest an additional mechanism by which two clinically significant risk factors for allergic disease exacerbations-IgE-mediated stimulation and rhinovirus infection-may synergistically promote Th2 differentiation and allergic inflammation.
Collapse
Affiliation(s)
- Regina K Rowe
- Departments of Pediatrics, University of Texas Southwestern Medical Center, Dallas, Texas
- Departments of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, Texas
- Departments of Immunology, University of Texas Southwestern Medical Center, Dallas, Texas
- Department of Pediatrics, University of Rochester Medical Center, Rochester, New York
| | - David M Pyle
- Departments of Pediatrics, University of Texas Southwestern Medical Center, Dallas, Texas
- Departments of Immunology, University of Texas Southwestern Medical Center, Dallas, Texas
| | - J David Farrar
- Departments of Pediatrics, University of Texas Southwestern Medical Center, Dallas, Texas
| | - Michelle A Gill
- Departments of Pediatrics, University of Texas Southwestern Medical Center, Dallas, Texas
- Departments of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, Texas
- Departments of Immunology, University of Texas Southwestern Medical Center, Dallas, Texas
| |
Collapse
|
17
|
Leffler J, Gamez C, Jones AP, Rueter K, Read JF, Siafarikas A, Lim EM, Noakes PS, Prescott SL, Stumbles PA, Palmer DJ, Strickland DH. In infants with sufficient vitamin D status at birth, vitamin D supplementation does not impact immune development. Pediatr Allergy Immunol 2020; 31:686-694. [PMID: 32248591 DOI: 10.1111/pai.13250] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/05/2019] [Revised: 03/15/2020] [Accepted: 03/21/2020] [Indexed: 12/01/2022]
Abstract
BACKGROUND Low vitamin D levels have been associated with allergic diseases. Vitamin D has potent immunomodulatory properties, but the mechanisms remain unclear. We have investigated the effect of oral vitamin D supplementation on circulating immune cell phenotypes in infants. METHOD A double-blinded randomised controlled trial was conducted to investigate the effect of oral vitamin D supplementation (400 IU/d) on eczema and immune development. A subset of 78 infants was included in this analysis. Phenotypic analysis of immune cell subsets was performed using flow cytometry. RESULTS Vitamin D supplementation resulted in median 25(OH)D levels of 80.5 vs 59.5 nmol/L in the placebo group at 3 months of age (P = .002) and 87.5 vs 77 nmol/L at 6 months of age (P = .08). We observed significant changes in immune cell composition from birth (cord blood) to 6 months of age. Vitamin D supplementation did not impact these changes, nor did immune cell composition correlate with plasma 25(OH)D levels. Through exploratory analysis, we identified possible associations with eczema development and increased abundance of naïve CD4- T cells at birth, as well as associations with basophils, iNKT and central memory CD4+ T cells, and altered expression patterns of IgE receptor (FcεR1) on monocytes and dendritic cells with eczema at 6 months. CONCLUSIONS Vitamin D supplementation in infants who were vitamin D sufficient at birth did not affect developmental changes in immune cells during the first 6 months of life. However, immune cell profiles at birth and at 6 months of age were associated with early life eczema.
Collapse
Affiliation(s)
- Jonatan Leffler
- Telethon Kids Institute, University of Western Australia, Nedlands, WA, Australia
| | - Cristina Gamez
- Telethon Kids Institute, University of Western Australia, Nedlands, WA, Australia.,School of Medicine, The University of Western Australia, Crawley, WA, Australia
| | - Anderson P Jones
- Telethon Kids Institute, University of Western Australia, Nedlands, WA, Australia.,School of Medicine, The University of Western Australia, Crawley, WA, Australia
| | - Kristina Rueter
- School of Medicine, The University of Western Australia, Crawley, WA, Australia.,Immunology Department, Perth Children's Hospital, Nedlands, WA, Australia
| | - James F Read
- Telethon Kids Institute, University of Western Australia, Nedlands, WA, Australia
| | - Aris Siafarikas
- Telethon Kids Institute, University of Western Australia, Nedlands, WA, Australia.,School of Medicine, The University of Western Australia, Crawley, WA, Australia.,Department of Endocrinology and Diabetes, Perth Children's Hospital, Nedlands, WA, Australia
| | - Ee-Mun Lim
- Department of Endocrinology and Diabetes, Sir Charles Gairdner Hospital, Nedlands, WA, Australia.,Department of Clinical Biochemistry, PathWest Laboratory Medicine, Queen Elizabeth II Medical Centre, Nedlands, WA, Australia
| | - Paul S Noakes
- Telethon Kids Institute, University of Western Australia, Nedlands, WA, Australia.,School of Medicine, The University of Western Australia, Crawley, WA, Australia.,School of Medicine, The University of Notre Dame, Fremantle, WA, Australia
| | - Susan L Prescott
- Telethon Kids Institute, University of Western Australia, Nedlands, WA, Australia.,School of Medicine, The University of Western Australia, Crawley, WA, Australia.,Immunology Department, Perth Children's Hospital, Nedlands, WA, Australia
| | - Philip A Stumbles
- Telethon Kids Institute, University of Western Australia, Nedlands, WA, Australia.,College of Science, Health, Engineering and Education, Murdoch University, Murdoch, WA, Australia
| | - Debra J Palmer
- Telethon Kids Institute, University of Western Australia, Nedlands, WA, Australia.,School of Medicine, The University of Western Australia, Crawley, WA, Australia
| | - Deborah H Strickland
- Telethon Kids Institute, University of Western Australia, Nedlands, WA, Australia
| |
Collapse
|
18
|
Rodríguez-Domínguez A, Berings M, Rohrbach A, Huang HJ, Curin M, Gevaert P, Matricardi PM, Valenta R, Vrtala S. Molecular profiling of allergen-specific antibody responses may enhance success of specific immunotherapy. J Allergy Clin Immunol 2020; 146:1097-1108. [PMID: 32298697 DOI: 10.1016/j.jaci.2020.03.029] [Citation(s) in RCA: 55] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2019] [Revised: 02/28/2020] [Accepted: 03/26/2020] [Indexed: 02/06/2023]
Abstract
BACKGROUND House dust mites (HDMs) are among the most important allergen sources containing many different allergenic molecules. Analysis of patients from a double-blind, placebo-controlled allergen-specific immunotherapy (AIT) study indicated that patients may benefit from AIT to different extents depending on their molecular sensitization profiles. OBJECTIVE Our aim was to investigate in a real-life setting whether stratification of patients with HDM allergy according to molecular analysis may enhance AIT success. METHODS Serum and nasal secretion samples from patients with HDM allergy (n = 24) (at baseline, 7, 15, 33, and 52 weeks) who had received 1 year of treatment with a well-defined subcutaneous AIT form (Alutard SQ 510) were tested for IgE and IgG reactivity to 15 microarrayed HDM allergen molecules with ImmunoCAP Immuno-solid-phase Allergen Chip technology. IgG subclass levels to allergens and peptides were determined by ELISA, and IgG blocking was assessed by basophil activation. In vitro parameters were related to reduction of symptoms determined by combined symptom medication score and visual analog scale score. RESULTS Alutard SQ 510 induced protective IgG mainly against Dermatophagoides pteronyssinus (Der p) 1 and Der p 2 and to a lesser extent to Der p 23, but not to the other important allergens such as Der p 5, Der p 7, and Der p 21, showing better clinical efficacy in patients sensitized only to Der p 1 and/or Der p 2 as compared with patients having additional IgE specificities. CONCLUSION Stratification of patients with HDM allergy according to molecular sensitization profiles and molecular monitoring of AIT-induced IgG responses may enhance the success of AIT.
Collapse
Affiliation(s)
- Azahara Rodríguez-Domínguez
- Department of Pathophysiology and Allergy Research, Division of Immunopathology, Center of Pathophysiology, Infectiology and Immunology, Medical University of Vienna, Vienna and Krems, Austria
| | - Margot Berings
- Upper Airways Research Laboratory, Ghent University, Ghent, Belgium
| | - Alexander Rohrbach
- Department of Pediatric Pulmonology, Immunology and Intensive Care Medicine, Charité-University Medicine Berlin, Berlin, Germany
| | - Huey-Jy Huang
- Department of Pathophysiology and Allergy Research, Division of Immunopathology, Center of Pathophysiology, Infectiology and Immunology, Medical University of Vienna, Vienna and Krems, Austria
| | - Mirela Curin
- Department of Pathophysiology and Allergy Research, Division of Immunopathology, Center of Pathophysiology, Infectiology and Immunology, Medical University of Vienna, Vienna and Krems, Austria
| | - Philippe Gevaert
- Upper Airways Research Laboratory, Ghent University, Ghent, Belgium
| | - Paolo M Matricardi
- Department of Pediatric Pulmonology, Immunology and Intensive Care Medicine, Charité-University Medicine Berlin, Berlin, Germany
| | - Rudolf Valenta
- Department of Pathophysiology and Allergy Research, Division of Immunopathology, Center of Pathophysiology, Infectiology and Immunology, Medical University of Vienna, Vienna and Krems, Austria; NRC Institute of Immunology, Federal Biomedical Agency of Russia, Moscow; Department of Clinical Immunology and Allergy, Sechenov First State Medical University, Moscow, Russia; Karl Landsteiner University of Health Sciences, Krems, Austria
| | - Susanne Vrtala
- Department of Pathophysiology and Allergy Research, Division of Immunopathology, Center of Pathophysiology, Infectiology and Immunology, Medical University of Vienna, Vienna and Krems, Austria.
| |
Collapse
|
19
|
Wang C, Li L, Jiang J, Li L, Li J, Xu C, Jin S, Zhu L, Yan G. Pterostilbene Inhibits FcεRI Signaling through Activation of the LKB1/AMPK Pathway in Allergic Response. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2020; 68:3456-3465. [PMID: 32096633 DOI: 10.1021/acs.jafc.9b07126] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/10/2023]
Abstract
In this study, the role and mechanism of pterostilbene (Pts) in mast cell degranulation in vitro and in vivo were investigated. The results showed that Pts inhibited mast cell-mediated local passive allergic reactions in mice. In addition, treatment with Pts reduced both histamine release and calcium influx in rat peritoneal mast cells and RBL-2H3 cells and reduced IgE-mediated mast cell activation. Furthermore, the mechanism underlying Pts inhibition of mast cell signaling was probed via studying the effects of Pts on liver kinase B1 (LKB1), including the use of the LKB1 activator metformin and siRNA knockdown of LKB1. The data showed that Pts reduced the release of inflammatory mediators such as tumor necrosis factor-α, interleukin-6, leukotriene C4, and prostaglandin D2 in mast cells by activating the LKB1/adenosine monophosphate (AMP)-activated protein kinase (AMPK) signaling pathway. Furthermore, Pts inhibited phosphorylation of FcεRI and FcεRI-mediated degranulation in RBL-2H3 cells. These effects were attenuated after LKB1 knockdown. Taken together, Pts could inhibit FcεRI signaling through activation of the LKB1/AMPK signaling pathway in IgE-mediated mast cell activation. Thus, Pts might be an effective therapeutic agent for mast cell-mediated allergic diseases.
Collapse
Affiliation(s)
- Chongyang Wang
- Department of Anatomy, Histology and Embryology, Medical College, Yanbian University, Yanji 133002, P. R. China
- Jilin Key Laboratory of Anaphylactic Disease, Yanbian University, Yanji 133000, P. R. China
| | - Liangchang Li
- Department of Anatomy, Histology and Embryology, Medical College, Yanbian University, Yanji 133002, P. R. China
- Jilin Key Laboratory of Anaphylactic Disease, Yanbian University, Yanji 133000, P. R. China
| | - Jingzhi Jiang
- Department of Anatomy, Histology and Embryology, Medical College, Yanbian University, Yanji 133002, P. R. China
- Jilin Key Laboratory of Anaphylactic Disease, Yanbian University, Yanji 133000, P. R. China
| | - Li Li
- Department of Anatomy, Histology and Embryology, Medical College, Yanbian University, Yanji 133002, P. R. China
- Jilin Key Laboratory of Anaphylactic Disease, Yanbian University, Yanji 133000, P. R. China
| | - Junfeng Li
- Department of Anatomy, Histology and Embryology, Medical College, Yanbian University, Yanji 133002, P. R. China
- Jilin Key Laboratory of Anaphylactic Disease, Yanbian University, Yanji 133000, P. R. China
| | - Chang Xu
- Department of Anatomy, Histology and Embryology, Medical College, Yanbian University, Yanji 133002, P. R. China
- Jilin Key Laboratory of Anaphylactic Disease, Yanbian University, Yanji 133000, P. R. China
| | - Shan Jin
- Jilin Key Laboratory of Anaphylactic Disease, Yanbian University, Yanji 133000, P. R. China
- Department of Dermatology, Yanbian University Hospital, Yanji 133002, P. R. China
| | - Lianhua Zhu
- Jilin Key Laboratory of Anaphylactic Disease, Yanbian University, Yanji 133000, P. R. China
- Department of Dermatology, Yanbian University Hospital, Yanji 133002, P. R. China
| | - Guanghai Yan
- Department of Anatomy, Histology and Embryology, Medical College, Yanbian University, Yanji 133002, P. R. China
- Jilin Key Laboratory of Anaphylactic Disease, Yanbian University, Yanji 133000, P. R. China
| |
Collapse
|
20
|
Wang W, Yin J, Wang X, Ma T, Lan T, Song Q, Guo Y. Relationship between serum inhibitory activity for IgE and efficacy of Artemisia pollen subcutaneous immunotherapy for allergic rhinitis: a preliminary self-controlled study. Allergy Asthma Clin Immunol 2020; 16:18. [PMID: 32158477 PMCID: PMC7057474 DOI: 10.1186/s13223-020-0416-4] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2019] [Accepted: 02/25/2020] [Indexed: 01/18/2023] Open
Abstract
Background Biomarkers of clinical efficacy for subcutaneous immunotherapy (SCIT) on allergic rhinitis (AR) have not been identified yet. This study aims to assess the clinical relevance of serum inhibitory activity for IgE by the method of enzyme-linked immunosorbent facilitated antigen binding (ELIFAB) during SCIT for Artemisia-sensitized AR patients. Methods 19 AR patients were studied who had undergone Artemisia-specific SCIT for more than 8 months (19.68 months on average, ranging from 9 to 33 months). Peripheral bloods were collected before and after treatment. The serum inhibitory activity for IgE was tested by ELIFAB and the level of Artemisia-specific IgG4 (Artemisia-sIgG4) was determined by ELISA. Clinical improvement was evaluated based on the symptom scores and rescue medication use (SMS). The 2-tailed Wilcoxon signed-rank test and the Spearman rank test (two-tailed) were used to analyze data by using SPSS 20.0, with P values of less than 0.05 considered as significant. Results The SMS decreased significantly after SCIT (before: 12.79 ± 4.250, after: 6.11 ± 3.828, P = 0.000 < 0.01), the treatment was remarkably effective for 6 patients, effective for 10 and ineffective for 3, along with a total effective rate 84.21%. The serum inhibitory activity for IgE increased significantly after SCIT (P < 0.05) and was correlated with the levels of Artemisia-sIgG4 (r = − 0.501, P = 0.002 < 0.01). The levels of Artemisia-sIgG4 elevated dramatically after treatment (P < 0.01) and were related with the duration of treatment (r = 0.558, P = 0.000 < 0.01). But there was no relationship between clinical improvements and the serum inhibitory activity for IgE. Conclusions The serum inhibitory activity for IgE increased significantly after SCIT, however, there was no correlation between it and clinical improvements by statistics analysis. So whether the serum inhibitory activity for IgE can act as biomarker of efficacy for SCIT or not needs to be studied further.
Collapse
Affiliation(s)
- Wenping Wang
- 1Department of Otolaryngology, Head and Neck Surgery, Peking University Ninth School of Clinical Medicine, Beijing, China
| | - Jinshu Yin
- 1Department of Otolaryngology, Head and Neck Surgery, Peking University Ninth School of Clinical Medicine, Beijing, China.,2Department of Otolaryngology, Head and Neck Surgery, Capital Medical University Affiliated Beijing Shijitan Hospital, Beijing, China
| | - Xueyan Wang
- 3Department of Allergy, Capital Medical University Affiliated Beijing Shijitan Hospital, Beijing, China
| | - Tingting Ma
- 3Department of Allergy, Capital Medical University Affiliated Beijing Shijitan Hospital, Beijing, China
| | - Tianfei Lan
- 3Department of Allergy, Capital Medical University Affiliated Beijing Shijitan Hospital, Beijing, China
| | - Qingkun Song
- 4Department of Science and Technology, Capital Medical University Affiliated Beijing Shijitan Hospital, Beijing, China
| | - Yifan Guo
- 5Department of General Surgery, Peking University Ninth School of Clinical Medicine, Beijing, China
| |
Collapse
|
21
|
Voskamp AL, Kormelink TG, van Wijk RG, Hiemstra PS, Taube C, de Jong EC, Smits HH. Modulating local airway immune responses to treat allergic asthma: lessons from experimental models and human studies. Semin Immunopathol 2020; 42:95-110. [PMID: 32020335 PMCID: PMC7066288 DOI: 10.1007/s00281-020-00782-4] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2019] [Accepted: 01/14/2020] [Indexed: 12/17/2022]
Abstract
With asthma affecting over 300 million individuals world-wide and estimated to affect 400 million by 2025, developing effective, long-lasting therapeutics is essential. Allergic asthma, where Th2-type immunity plays a central role, represents 90% of child and 50% of adult asthma cases. Research based largely on animal models of allergic disease have led to the generation of a novel class of drugs, so-called biologicals, that target essential components of Th2-type inflammation. Although highly efficient in subclasses of patients, these biologicals and other existing medication only target the symptomatic stage of asthma and when therapy is ceased, a flare-up of the disease is often observed. Therefore, it is suggested to target earlier stages in the inflammatory cascade underlying allergic airway inflammation and to focus on changing and redirecting the initiation of type 2 inflammatory responses against allergens and certain viral agents. This focus on upstream aspects of innate immunity that drive development of Th2-type immunity is expected to have longer-lasting and disease-modifying effects, and may potentially lead to a cure for asthma. This review highlights the current understanding of the contribution of local innate immune elements in the development and maintenance of inflammatory airway responses and discusses available leads for successful targeting of those pathways for future therapeutics.
Collapse
Affiliation(s)
- A L Voskamp
- Department of Parasitology, Leiden University Medical Center, Albinusdreef 2 2333 ZA, Leiden, The Netherlands
| | - T Groot Kormelink
- Department of Experimental Immunology, Amsterdam University Medical Centers, AMC, Amsterdam, The Netherlands
| | - R Gerth van Wijk
- Department of Internal Medicine, Section Allergology, Erasmus University Medical Center, Rotterdam, The Netherlands
| | - P S Hiemstra
- Department of Pulmonology, Leiden University Medical Center, Leiden, The Netherlands
| | - C Taube
- Department of Pulmonary Medicine, University Hospital Essen - Ruhrklinik, Essen, Germany
| | - E C de Jong
- Department of Experimental Immunology, Amsterdam University Medical Centers, AMC, Amsterdam, The Netherlands
| | - Hermelijn H Smits
- Department of Parasitology, Leiden University Medical Center, Albinusdreef 2 2333 ZA, Leiden, The Netherlands.
| |
Collapse
|
22
|
|
23
|
Wang W, Yin J. Is it worthy to take full-course immunotherapy for allergic rhinitis? About efficacy biomarker of allergen immunotherapy. Scand J Immunol 2019; 91:e12817. [PMID: 31650620 DOI: 10.1111/sji.12817] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2019] [Revised: 08/10/2019] [Accepted: 08/20/2019] [Indexed: 01/15/2023]
Abstract
Nowadays, people pay more attention to biomarkers that can predict clinical efficacy of immunotherapy for allergic rhinitis. As the only recognized aetiological treatment, the efficacy of allergen immunotherapy (AIT) has been proved by many studies. However, treatment success depends on compliance and persistence greatly, which can be impaired by the lengthy duration of AIT and socioeconomic status of patients. Besides, ineffectiveness is another factor that accounts for non-adherence. If the clinical efficacy can be predicted in the early stage of immunotherapy, it can help patients choose appropriate treatment plans, increase patient compliance and optimize the allocation of medical resources. This paper mainly focuses on five candidate biomarkers, the sIgE/tIgE ratio before treatment, serum inhibitory activity for IgE, decreased basophil activation, upregulation of Tregs and tolerogenic DCs, reviews the time when potential biomarkers can predict or monitor the efficacy of AIT, discusses the reason why these indicators could serve as efficacy biomarkers and interactions among potential biomarkers.
Collapse
Affiliation(s)
- Wenping Wang
- Department of otolaryngology, head and neck surgery, Peking University Ninth School of Clinical Medicine, Beijing, China
| | - Jinshu Yin
- Department of otolaryngology, head and neck surgery, Peking University Ninth School of Clinical Medicine, Capital Medical University affiliated Beijing Shijitan Hospital, Beijing, China
| |
Collapse
|
24
|
Leffler J, Read JF, Jones AC, Mok D, Hollams EM, Laing IA, Le Souef PN, Sly PD, Kusel MMH, de Klerk NH, Bosco A, Holt PG, Strickland DH. Progressive increase of FcεRI expression across several PBMC subsets is associated with atopy and atopic asthma within school-aged children. Pediatr Allergy Immunol 2019; 30:646-653. [PMID: 30985951 DOI: 10.1111/pai.13063] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/20/2018] [Revised: 04/02/2019] [Accepted: 04/03/2019] [Indexed: 12/27/2022]
Abstract
BACKGROUND Antigen-specific IgE binds the Fcε receptor I (FcεRI) expressed on several types of immune cells, including dendritic cells (DCs). Activation of FcεRI on DCs in atopics has been shown to modulate immune responses that potentially contribute to asthma development. However, the extent to which DC subsets differ in FcεRI expression between atopic children with or without asthma is currently not clear. This study aimed to analyse the expression of FcεRI on peripheral blood mononuclear cells (PBMCs) from atopic children with and without asthma, and non-atopic/non-asthmatic age-matched healthy controls. METHODS We performed multiparameter flow cytometry on PBMC from 391 children across three community cohorts and one clinical cohort based in Western Australia. RESULTS We confirmed expression of FcεRI on basophils, monocytes, plasmacytoid and conventional DCs, with higher proportions of all cell populations expressing FcεRI in atopic compared to non-atopic children. Further, we observed that levels of FcεRI expression were elevated across plasmacytoid and conventional DC as well as basophils in atopic asthmatic compared to atopic non-asthmatic children also after adjusting for serum IgE levels. CONCLUSION Our data suggest that the expression pattern of FcεRI on DC and basophils differentiates asthmatic from non-asthmatic atopic children. Given the significant immune modulatory effects observed as a consequence of FcεRI expression, this altered expression pattern is likely to contribute to asthma pathology in children.
Collapse
Affiliation(s)
- Jonatan Leffler
- Telethon Kids Institute, University of Western Australia, Nedlands, Western Australia, Australia
| | - James F Read
- Telethon Kids Institute, University of Western Australia, Nedlands, Western Australia, Australia.,School of Medicine, University of Western Australia, Nedlands, Western Australia, Australia
| | - Anya C Jones
- Telethon Kids Institute, University of Western Australia, Nedlands, Western Australia, Australia.,School of Medicine, University of Western Australia, Nedlands, Western Australia, Australia
| | - Danny Mok
- Telethon Kids Institute, University of Western Australia, Nedlands, Western Australia, Australia
| | - Elysia M Hollams
- Telethon Kids Institute, University of Western Australia, Nedlands, Western Australia, Australia
| | - Ingrid A Laing
- Telethon Kids Institute, University of Western Australia, Nedlands, Western Australia, Australia.,School of Medicine, University of Western Australia, Nedlands, Western Australia, Australia
| | - Peter N Le Souef
- Telethon Kids Institute, University of Western Australia, Nedlands, Western Australia, Australia.,School of Medicine, University of Western Australia, Nedlands, Western Australia, Australia
| | - Peter D Sly
- Child Health Research Centre, University of Queensland, Brisbane, Queensland, Australia
| | - Merci M H Kusel
- Telethon Kids Institute, University of Western Australia, Nedlands, Western Australia, Australia
| | - Nicholas H de Klerk
- Telethon Kids Institute, University of Western Australia, Nedlands, Western Australia, Australia
| | - Anthony Bosco
- Telethon Kids Institute, University of Western Australia, Nedlands, Western Australia, Australia
| | - Patrick G Holt
- Telethon Kids Institute, University of Western Australia, Nedlands, Western Australia, Australia.,Child Health Research Centre, University of Queensland, Brisbane, Queensland, Australia
| | - Deborah H Strickland
- Telethon Kids Institute, University of Western Australia, Nedlands, Western Australia, Australia
| |
Collapse
|
25
|
Li X, Wang N, Zhao Y, Zhang Y, Liu Z. Species Specificity on Interaction between IgE and FcεRI. Curr Pharm Biotechnol 2019; 20:690-695. [PMID: 31258078 DOI: 10.2174/1389201020666190619122325] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2019] [Revised: 04/30/2019] [Accepted: 05/01/2019] [Indexed: 11/22/2022]
Abstract
Allergic diseases are one of the most prevalent diseases at present, it is imperative to understanding the pathophysiology and treatment strategies for allergic diseases. In this process, the binding of IgE and FcεRI on effector cells plays a critical role in triggering allergic reactions. However, the species specificity of the interaction between IgE and FcεRI has not been clearly explained. This review described the characteristics and the interaction mechanism in the allergic reaction of IgE and FcεRI and summarized the species specificity between IgE and FcεRI.
Collapse
Affiliation(s)
- Xiangsheng Li
- College of Pharmaceutical Sciences, Hebei University, Baoding 071002, China.,Key Laboratory of Pharmaceutical Quality Control of Hebei Province, Baoding 071002, China
| | - Nannan Wang
- College of Pharmaceutical Sciences, Hebei University, Baoding 071002, China.,Key Laboratory of Pharmaceutical Quality Control of Hebei Province, Baoding 071002, China
| | - Yang Zhao
- College of Pharmaceutical Sciences, Hebei University, Baoding 071002, China.,Key Laboratory of Pharmaceutical Quality Control of Hebei Province, Baoding 071002, China
| | - Yanfen Zhang
- Key Laboratory of Pharmaceutical Quality Control of Hebei Province, Baoding 071002, China.,Offices of Science and Technology, Hebei University, Baoding 071002, China
| | - Zhongcheng Liu
- College of Pharmaceutical Sciences, Hebei University, Baoding 071002, China.,Key Laboratory of Pharmaceutical Quality Control of Hebei Province, Baoding 071002, China
| |
Collapse
|
26
|
Jannat A, Khan M, Shabbir M, Badshah Y. Expression of suppressor of cytokine signaling 3 (SOCS3) and interleukin-6 (-174-G/C) polymorphism in atopic conditions. PLoS One 2019; 14:e0219084. [PMID: 31251775 PMCID: PMC6599118 DOI: 10.1371/journal.pone.0219084] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2019] [Accepted: 06/15/2019] [Indexed: 11/24/2022] Open
Abstract
Hypersensitivity of the immune system is caused by elevated immunoglobulin E (IgE) levels in the serum, in response to a discrete allergen leading to allergic reactions. IgE-mediated inflammation is regulated by the cascade of defense related signaling molecules including interleukin-6 (IL-6) that plays pivotal role in the survival and maturation of mast cells during an allergic reaction. IL-6 mediated defense responses are tightly regulated by Suppressor of Cytokine Signaling 3 (SOCS3), an inhibitory molecules of Janus Kinase-Signal Transducers and Activators of Transcription (JAK-STAT) signaling, in a negative feedback mechanism. The given study focuses on the assessment of crosstalk between SOCS3 and IL-6 to unravel the molecular significance of SOCS3 and IL-6 in the diagnosis and prognosis of allergy. The expression study of SOCS3 through real-time PCR analysis revealed, a 5.9 mean fold increase in SOCS3 expression in atopic cases in comparison to control cases. Moreover, IL-6 has, also, been found significantly enhanced in the serum level of atopic cases (26.4 pg/ml) as compared to control cases (3.686 pg/ml). Female population was found to be at a higher risk to develop atopic condition than male population as females exhibited higher expression of both SOCS3 and IL-6 than males. Furthermore, the polymorphic study of IL-6 promoter region (IL-6 174-G/C) in atopic population has reasserted the importance of SOCS3 and IL-6 in the diagnosis and prognosis of allergy. Expression of SOCS3 and IL-6 serum levels were found to be highly correlated. Therefore establishing the role of IL-6 (-174-G/C) polymorphism on the expression of SOCS3 and IL-6 in atopic cases. Notably, the study established SOCS3 and IL-6 as potential targets for the diagnosis/prognosis of allergy and for the development of reliable therapeutic strategies to control atopic conditions in the near future.
Collapse
Affiliation(s)
- Arooma Jannat
- Atta-ur-Rahman School of Applied Biosciences (ASAB), National University of Sciences and Technology (NUST), Islamabad, Pakistan
| | - Maryam Khan
- Atta-ur-Rahman School of Applied Biosciences (ASAB), National University of Sciences and Technology (NUST), Islamabad, Pakistan
| | - Maria Shabbir
- Atta-ur-Rahman School of Applied Biosciences (ASAB), National University of Sciences and Technology (NUST), Islamabad, Pakistan
| | - Yasmin Badshah
- Atta-ur-Rahman School of Applied Biosciences (ASAB), National University of Sciences and Technology (NUST), Islamabad, Pakistan
- * E-mail:
| |
Collapse
|
27
|
León MA, Palma C, Hernández C, Sandoval M, Cofre C, Perez-Mateluna G, Borzutzky A, Harris PR, Serrano CA. Helicobacter pylori pediatric infection changes FcεRI expression in dendritic cells and Treg profile in vivo and in vitro. Microbes Infect 2019; 21:449-455. [PMID: 31128278 DOI: 10.1016/j.micinf.2019.05.001] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2018] [Revised: 04/15/2019] [Accepted: 05/13/2019] [Indexed: 02/06/2023]
Abstract
H. pylori infection shows an inverse relationship with allergies. Dendritic cells regulate mucosal immune responses including the induction of T regulatory cells which are fundamental in Helicobacter pylori-induced dampening of allergies. In this respect expression of high-affinity IgE receptor (FcεRI) has been associated with a regulatory dendritic cell profile. Therefore we aimed to evaluate possible mechanisms by which H. pylori infection might modify atopy in pediatric patients. Here we show that H. pylori-infected children exhibited both increased expression of FcεRI on peripheral myeloid and plasmacytoid dendritic cells and higher levels of Foxp3 and Latency Associated Peptide on T regulatory cells. Moreover, exposure to H. pylori drove increased FcεRI expression and IL-10 secretion by both pediatric H. pylori-exposed monocyte derived dendritic cells and T cells. Finally, we show a positive correlation between expression of FcεRI in circulating myeloid DCs and total Treg cells, suggesting that in children, H. pylori infection may have a modulating role in atopy, mediated by both altered surface expression of FcεRI on children's DC and an increased T regulatory cell profile.
Collapse
Affiliation(s)
- Miguel A León
- Department of Pediatric Gastroenterology and Nutrition, School of Medicine, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Camila Palma
- Department of Pediatric Gastroenterology and Nutrition, School of Medicine, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Caroll Hernández
- Department of Pediatric Gastroenterology and Nutrition, School of Medicine, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Mauricio Sandoval
- Department of Pediatric Gastroenterology and Nutrition, School of Medicine, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Colomba Cofre
- Department of Pediatric Gastroenterology and Nutrition, School of Medicine, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Guillermo Perez-Mateluna
- Department of Pediatric Infectious Diseases and Immunology, Millennium Institute on Immunology and Immunotherapy, School of Medicine, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Arturo Borzutzky
- Department of Pediatric Infectious Diseases and Immunology, Millennium Institute on Immunology and Immunotherapy, School of Medicine, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Paul R Harris
- Department of Pediatric Gastroenterology and Nutrition, School of Medicine, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Carolina A Serrano
- Department of Pediatric Gastroenterology and Nutrition, School of Medicine, Pontificia Universidad Católica de Chile, Santiago, Chile.
| |
Collapse
|
28
|
Rondón C, Eguíluz-Gracia I, Shamji MH, Layhadi JA, Salas M, Torres MJ, Campo P. IgE Test in Secretions of Patients with Respiratory Allergy. Curr Allergy Asthma Rep 2018; 18:67. [PMID: 30317418 DOI: 10.1007/s11882-018-0821-7] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
PURPOSE OF REVIEW IgE is a key player in multiple inflammatory airway diseases. Ample literature demonstrates its presence in mucosa of patients with allergic rhinitis (AR), local allergic rhinitis (LAR), asthma, or chronic rhinosinusitis with nasal polyposis (CRSwNP). RECENT FINDINGS Current evidence shows that high-affinity IgE in blood stream of allergic individuals derives mainly from the mucosae. Also, mucosal synthesis of IgE can occur in the absence of systemic atopy, and may be relevant in atopic and non-atopic phenotypes of rhinitis as demonstrated in LAR. Specific IgE (sIgE) detection varies depending on technique used for sample collection and its measurement. sIgE detection is highly specific for diagnosis of LAR. Moreover, measurement of sIgE in secretions could be useful in monitoring response to allergen-specific immunotherapy in both AR and LAR phenotypes. This review will focus on recent developments in the role of IgE in respiratory diseases, and the clinical implications of its measurement in secretions.
Collapse
Affiliation(s)
- Carmen Rondón
- Allergy Unit, IBIMA-Regional University Hospital of Málaga, Málaga, Spain
| | | | - Mohamed H Shamji
- Immunomodulation and Tolerance Group, Allergy and Clinical Immunology, Inflammation, Repair and Development, MRC Asthma UK Centre Imperial College London, London, UK
| | - Janice A Layhadi
- Immunomodulation and Tolerance Group, Allergy and Clinical Immunology, Inflammation, Repair and Development, MRC Asthma UK Centre Imperial College London, London, UK
| | - María Salas
- Allergy Unit, IBIMA-Regional University Hospital of Málaga, Málaga, Spain
| | - María José Torres
- Allergy Unit, IBIMA-Regional University Hospital of Málaga, Málaga, Spain
| | - Paloma Campo
- Allergy Unit, IBIMA-Regional University Hospital of Málaga, Málaga, Spain.
- Plaza Hospital Civil, 29009, Málaga, Spain.
| |
Collapse
|