51
|
Jia Z, Guo M, Ge X, Chen F, Lei P. IL-33/ST2 Axis: A Potential Therapeutic Target in Neurodegenerative Diseases. Biomolecules 2023; 13:1494. [PMID: 37892176 PMCID: PMC10605306 DOI: 10.3390/biom13101494] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2023] [Revised: 10/01/2023] [Accepted: 10/03/2023] [Indexed: 10/29/2023] Open
Abstract
Interleukin 33 (IL-33) belongs to the IL-1 family and is localized in the nucleus. IL-33 is primarily composed of three distinct domains, namely the N-terminal domain responsible for nuclear localization, the intermediate sense protease domain, and the C-terminal cytokine domain. Its specific receptor is the suppression of tumorigenicity 2 (ST2), which is detected in serum-stimulated fibroblasts and oncogenes. While most other cytokines are actively produced in cells, IL-33 is passively produced in response to tissue damage or cell necrosis, thereby suggesting its role as an alarm following cell infection, stress, or trauma. IL-33 plays a crucial role in congenital and acquired immunity, which assists in the response to environmental stress and maintains tissue homeostasis. IL-33/ST2 interaction further produces many pro-inflammatory cytokines. Moreover, IL-33 is crucial for central nervous system (CNS) homeostasis and the pathogenic mechanisms underlying CNS degenerative disorders. The present work summarizes the structure of IL-33, its fundamental activities, and its role in immunoregulation and neurodegenerative diseases. Therefore, this work proposes that IL-33 may play a role in the pathogenic mechanism of diseases and can be used in the development of treatment strategies.
Collapse
Affiliation(s)
- Zexi Jia
- Department of Geriatrics, Tianjin Medical University General Hospital, Tianjin 300052, China; (Z.J.); (X.G.)
- Tianjin Geriatrics Institute, Tianjin Medical University General Hospital, Tianjin 300052, China
| | - Mengtian Guo
- Department of Internal Medicine, Beijing Chao-Yang Hospital, Capital Medical University, Beijing 100054, China;
| | - Xintong Ge
- Department of Geriatrics, Tianjin Medical University General Hospital, Tianjin 300052, China; (Z.J.); (X.G.)
- Tianjin Geriatrics Institute, Tianjin Medical University General Hospital, Tianjin 300052, China
| | - Fanglian Chen
- Tianjin Neurological Institute, Tianjin 300052, China
| | - Ping Lei
- Department of Geriatrics, Tianjin Medical University General Hospital, Tianjin 300052, China; (Z.J.); (X.G.)
- Tianjin Geriatrics Institute, Tianjin Medical University General Hospital, Tianjin 300052, China
| |
Collapse
|
52
|
Yarsky E, Banzon TM, Phipatanakul W. Effects of Allergen Exposure and Environmental Risk Factors in Schools on Childhood Asthma. Curr Allergy Asthma Rep 2023; 23:613-620. [PMID: 37651001 PMCID: PMC11262705 DOI: 10.1007/s11882-023-01108-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/16/2023] [Indexed: 09/01/2023]
Abstract
PURPOSE OF REVIEW This review aims to assess the prevalence of common allergen exposures and environmental risk factors for asthma in schools, examine the underlying mechanisms of these environmental risk factors, and explore possible prevention strategies. RECENT FINDINGS Cockroach, mouse, dust mites, fungi, viral infections, ozone pollution, and cleaning products are common allergen exposures and environmental risk factors in schools which may affect asthma morbidity. Novel modifiable environmental risk factors in schools are also being investigated to identify potential associations with increased asthma morbidity. While several studies have investigated the benefit of environmental remediation strategies in schools and their impact on asthma morbidity, future studies are warranted to further define the effects of modifiable risk factors in schools and determine whether school mitigation strategies may help improve asthma symptoms in students with asthma.
Collapse
Affiliation(s)
- Eva Yarsky
- Rutgers New Jersey Medical School, Newark, NJ, USA
| | - Tina M Banzon
- Division of Allergy and Immunology, Boston Children's Hospital, Harvard Medical School, Boston, MA, USA
| | - Wanda Phipatanakul
- Division of Allergy and Immunology, Boston Children's Hospital, Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
53
|
Zhang B, Zhu G, Liu J, Zhang C, Yao K, Huang X, Cen X, Zhao Z. Single-cell transcriptional profiling reveals immunomodulatory properties of stromal and epithelial cells in periodontal immune milieu with diabetes in rats. Int Immunopharmacol 2023; 123:110715. [PMID: 37562294 DOI: 10.1016/j.intimp.2023.110715] [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: 03/08/2023] [Revised: 07/01/2023] [Accepted: 07/24/2023] [Indexed: 08/12/2023]
Abstract
Periodontitis is the sixth major complication of diabetes. Gingiva, as an important component of periodontal tissues, serves as the first defense barrier against infectious stimuli. However, relatively little is known about cellular heterogeneity and cell-specific changes in gingiva in response to diabetes-associated periodontitis. To characterize molecular changes linking diabetes with periodontitis, we profiled single-cell transcriptome analyses of a total of 45,259 cells from rat gingiva with periodontitis under normoglycemic and diabetic condition. The single-cell profiling revealed that stromal and epithelial cells of gingiva contained inflammation-related subclusters enriched in functions of immune cell recruitment. Compared to normoglycemic condition, diabetes led to a reduction in epithelial basal cells, fibroblasts and smooth muscle cells in gingiva with periodontitis. Analysis of differentially expressed genes indicated that stromal and epithelial populations were reprogrammed towards pro-inflammatory phenotypes promoting immune cell recruitment in diabetes-related periodontitis. In aspect of immune cells, diabetes prominently enhanced neutrophil and M1 macrophage infiltration in periodontitis lesions. Cell-cell communications revealed enhanced crosstalk between stromal/epithelial cells and immune cells mediating by chemokine/chemokine receptor interplay in diabetes-associated periodontitis. Our findings deconvolved cellular heterogeneity of rat gingiva associated with periodontitis and diabetes, uncovered altered immune milieu caused by the disease, and revealed immunomodulatory functions of stromal and epithelial cells in gingival immune niche. The present study improves the understanding of the link between the diabetes and periodontitis and helps in formulating precise therapeutic strategies for diabetes-enhanced periodontitis.
Collapse
Affiliation(s)
- Bo Zhang
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases & Department of Orthodontics, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, China
| | - Guanyin Zhu
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases & Department of Orthodontics, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, China
| | - Junqi Liu
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases & Department of Orthodontics, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, China
| | - Chenghao Zhang
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases & Department of Orthodontics, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, China
| | - Ke Yao
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases & Department of Orthodontics, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, China
| | - Xinqi Huang
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases & Department of Orthodontics, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, China
| | - Xiao Cen
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases & Department of Orthodontics, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, China
| | - Zhihe Zhao
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases & Department of Orthodontics, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, China.
| |
Collapse
|
54
|
Tsuji G, Yamamura K, Kawamura K, Kido-Nakahara M, Ito T, Nakahara T. Regulatory Mechanism of the IL-33-IL-37 Axis via Aryl Hydrocarbon Receptor in Atopic Dermatitis and Psoriasis. Int J Mol Sci 2023; 24:14633. [PMID: 37834081 PMCID: PMC10572928 DOI: 10.3390/ijms241914633] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2023] [Revised: 09/20/2023] [Accepted: 09/26/2023] [Indexed: 10/15/2023] Open
Abstract
Interleukin (IL)-33 and IL-37 have been identified as novel cytokines involved in various inflammatory diseases. However, their specific roles remain largely unknown. Recent studies have shown that IL-33, which triggers inflammation, and IL-37, which suppresses it, cooperatively regulate the balance between inflammation and anti-inflammation. IL-33 and IL-37 are also deeply involved in the pathogenesis of inflammatory skin diseases such as atopic dermatitis (AD) and psoriasis. Furthermore, a signaling pathway by which aryl hydrocarbon receptor (AHR), a receptor for dioxins, regulates the expression of IL-33 and IL-37 has been revealed. Here, we outline recent findings on the mechanisms regulating IL-33 and IL-37 expression in AD and psoriasis. IL-33 expression is partially dependent on mitogen-activated protein kinase (MAPK) activation, and IL-37 has a role in suppressing MAPK in human keratinocytes. Furthermore, IL-33 downregulates skin barrier function proteins including filaggrin and loricrin, thereby downregulating the expression of IL-37, which colocalizes with these proteins. This leads to an imbalance of the IL-33-IL-37 axis, involving increased IL-33 and decreased IL-37, which may be associated with the pathogenesis of AD and psoriasis. Therefore, AHR-mediated regulation of the IL-33-IL-37 axis may lead to new therapeutic strategies for the treatment of AD and psoriasis.
Collapse
Affiliation(s)
- Gaku Tsuji
- Research and Clinical Center for Yusho and Dioxin, Kyushu University Hospital, 3-1-1, Maidashi, Higashi-ku, Fukuoka 812-8582, Japan; (K.Y.); (T.N.)
- Department of Dermatology, Graduate School of Medical Sciences, Kyushu University, 3-1-1, Maidashi, Higashi-ku, Fukuoka 812-8582, Japan; (K.K.); (M.K.-N.); (T.I.)
| | - Kazuhiko Yamamura
- Research and Clinical Center for Yusho and Dioxin, Kyushu University Hospital, 3-1-1, Maidashi, Higashi-ku, Fukuoka 812-8582, Japan; (K.Y.); (T.N.)
- Department of Dermatology, Graduate School of Medical Sciences, Kyushu University, 3-1-1, Maidashi, Higashi-ku, Fukuoka 812-8582, Japan; (K.K.); (M.K.-N.); (T.I.)
| | - Koji Kawamura
- Department of Dermatology, Graduate School of Medical Sciences, Kyushu University, 3-1-1, Maidashi, Higashi-ku, Fukuoka 812-8582, Japan; (K.K.); (M.K.-N.); (T.I.)
| | - Makiko Kido-Nakahara
- Department of Dermatology, Graduate School of Medical Sciences, Kyushu University, 3-1-1, Maidashi, Higashi-ku, Fukuoka 812-8582, Japan; (K.K.); (M.K.-N.); (T.I.)
| | - Takamichi Ito
- Department of Dermatology, Graduate School of Medical Sciences, Kyushu University, 3-1-1, Maidashi, Higashi-ku, Fukuoka 812-8582, Japan; (K.K.); (M.K.-N.); (T.I.)
| | - Takeshi Nakahara
- Research and Clinical Center for Yusho and Dioxin, Kyushu University Hospital, 3-1-1, Maidashi, Higashi-ku, Fukuoka 812-8582, Japan; (K.Y.); (T.N.)
- Department of Dermatology, Graduate School of Medical Sciences, Kyushu University, 3-1-1, Maidashi, Higashi-ku, Fukuoka 812-8582, Japan; (K.K.); (M.K.-N.); (T.I.)
| |
Collapse
|
55
|
Chua AJ, Jafar A, Luong AU. Update on allergic fungal rhinosinusitis. Ann Allergy Asthma Immunol 2023; 131:300-306. [PMID: 36854353 DOI: 10.1016/j.anai.2023.02.018] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2023] [Revised: 02/14/2023] [Accepted: 02/15/2023] [Indexed: 02/27/2023]
Abstract
Allergic fungal rhinosinusitis (AFRS) is a unique clinical entity that falls under the broader umbrella of chronic rhinosinusitis with nasal polyps with type 2 inflammation. It is characterized by nasal polyposis, production of characteristic thick eosinophilic mucin, and expansile change of involved sinus cavities. The diagnosis is classically made using the Bent and Kuhn criteria. However, recent studies have indicated the lack of specificity of some major criteria. The need to fulfill all 5 criteria before diagnosing AFRS partially mitigates this but renders the criteria cumbersome to use, and highlights the need to develop more specific criteria. Our understanding of AFRS pathophysiology has advanced significantly and has helped elucidate the lack of histatins contributing to the inability to clear fungal spores, consequently leading to fungi-induced disruption of the epithelial barrier and stimulation of sinonasal epithelial cells. These trigger a cascade of type 2 inflammatory cytokines driven by both the adaptive and innate immune system. Although more research is needed, these findings could hypothetically point to a limited type 3 immune response at the sinus mucosa, resulting in a compensatory overstimulation of type 2 inflammatory processes. Treatment for AFRS remains centered on surgery and topical corticosteroids. Short courses of systemic corticosteroids may be used with caution, and fungal-specific immunotherapy and systemic antifungals are options in recalcitrant disease. Biologics show early promise, as we await data from randomized controlled trials under way. Finally, new insights into AFRS pathology provide opportunities for novel therapeutic strategies.
Collapse
Affiliation(s)
- Andy J Chua
- Department of Otorhinolaryngology-Head and Neck Surgery, Sengkang General Hospital, Singapore Health Services, Singapore
| | - Ali Jafar
- Department of Otorhinolaryngology-Head and Neck Surgery, University of Texas Health Science Center, Houston, Texas
| | - Amber U Luong
- Department of Otorhinolaryngology-Head and Neck Surgery, University of Texas Health Science Center, Houston, Texas; Center for Immunology and Autoimmune Diseases, Institute of Molecular Medicine, McGovern Medical School at The University of Texas Health Science Center, Houston, Texas.
| |
Collapse
|
56
|
Cassano A, Chong AS, Alegre ML. Tregs in transplantation tolerance: role and therapeutic potential. FRONTIERS IN TRANSPLANTATION 2023; 2:1217065. [PMID: 38993904 PMCID: PMC11235334 DOI: 10.3389/frtra.2023.1217065] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 05/04/2023] [Accepted: 08/14/2023] [Indexed: 07/13/2024]
Abstract
CD4+ Foxp3+ regulatory T cells (Tregs) are indispensable for preventing autoimmunity, and they play a role in cancer and transplantation settings by restraining immune responses. In this review, we describe evidence for the importance of Tregs in the induction versus maintenance of transplantation tolerance, discussing insights into mechanisms of Treg control of the alloimmune response. Further, we address the therapeutic potential of Tregs as a clinical intervention after transplantation, highlighting engineered CAR-Tregs as well as expansion of donor and host Tregs.
Collapse
Affiliation(s)
- Alexandra Cassano
- Department of Medicine, University of Chicago, Chicago, IL, United States
| | - Anita S. Chong
- Department of Surgery, University of Chicago, Chicago, IL, United States
| | - Maria-Luisa Alegre
- Department of Medicine, University of Chicago, Chicago, IL, United States
| |
Collapse
|
57
|
Wang Y, Cheng D, Li Z, Sun W, Zhou S, Peng L, Xiong H, Jia X, Li W, Han L, Liu Y, Ni C. IL33-mediated NPM1 promotes fibroblast-to-myofibroblast transition via ERK/AP-1 signaling in silica-induced pulmonary fibrosis. Toxicol Sci 2023; 195:71-86. [PMID: 37399107 DOI: 10.1093/toxsci/kfad061] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/05/2023] Open
Abstract
Silicosis is a global occupational pulmonary disease due to the accumulation of silica dust in the lung. Lacking effective clinical drugs makes the treatment of this disease quite challenging in clinics largely because the pathogenic mechanisms remain obscure. Interleukin 33 (IL33), a pleiotropic cytokine, could promote wound healing and tissue repair via the receptor ST2. However, the mechanisms governing the involvement of IL33 in silicosis progression remain to be further explored. Here, we demonstrated that the IL33 levels in the lung sections were significantly overexpressed after bleomycin and silica treatment. Chromatin immunoprecipitation assay, knockdown, and reverse experiments were performed in lung fibroblasts to prove gene interaction following exogenous IL33 treatment or cocultured with silica-treated lung epithelial cells. Mechanistically, we illustrated that silica-stimulated lung epithelial cells secreted IL33 and further promoted the activation, proliferation, and migration of pulmonary fibroblasts by activating the ERK/AP-1/NPM1 signaling pathway in vitro. And more, treatment with NPM1 siRNA-loaded liposomes markedly protected mice from silica-induced pulmonary fibrosis in vivo. In conclusion, the involvement of NPM1 in the progression of silicosis is regulated by the IL33/ERK/AP-1 signaling axis, which is the potential therapeutic target candidate in developing novel antifibrotic strategies for pulmonary fibrosis.
Collapse
Affiliation(s)
- Yue Wang
- Department of Occupational Medical and Environmental Health, Key Laboratory of Modern Toxicology of Ministry of Education, School of Public Health, Nanjing Medical University, Nanjing 211166, China
| | - Demin Cheng
- Department of Occupational Medical and Environmental Health, Key Laboratory of Modern Toxicology of Ministry of Education, School of Public Health, Nanjing Medical University, Nanjing 211166, China
| | - Ziwei Li
- Department of Occupational Medical and Environmental Health, Key Laboratory of Modern Toxicology of Ministry of Education, School of Public Health, Nanjing Medical University, Nanjing 211166, China
| | - Wenqing Sun
- Department of Occupational Medical and Environmental Health, Key Laboratory of Modern Toxicology of Ministry of Education, School of Public Health, Nanjing Medical University, Nanjing 211166, China
| | - Siyun Zhou
- Department of Occupational Medical and Environmental Health, Key Laboratory of Modern Toxicology of Ministry of Education, School of Public Health, Nanjing Medical University, Nanjing 211166, China
| | - Lan Peng
- Department of Occupational Medical and Environmental Health, Key Laboratory of Modern Toxicology of Ministry of Education, School of Public Health, Nanjing Medical University, Nanjing 211166, China
| | - Haojie Xiong
- Department of Occupational Medical and Environmental Health, Key Laboratory of Modern Toxicology of Ministry of Education, School of Public Health, Nanjing Medical University, Nanjing 211166, China
| | - Xinying Jia
- Department of Occupational Medical and Environmental Health, Key Laboratory of Modern Toxicology of Ministry of Education, School of Public Health, Nanjing Medical University, Nanjing 211166, China
| | - Wei Li
- Department of Occupational Medical and Environmental Health, Key Laboratory of Modern Toxicology of Ministry of Education, School of Public Health, Nanjing Medical University, Nanjing 211166, China
- Institute of Occupational Disease Prevention, Jiangsu Provincial Center for Disease Control and Prevention, Nanjing 210028, China
| | - Lei Han
- Institute of Occupational Disease Prevention, Jiangsu Provincial Center for Disease Control and Prevention, Nanjing 210028, China
| | - Yi Liu
- Department of Occupational Medical and Environmental Health, Key Laboratory of Modern Toxicology of Ministry of Education, School of Public Health, Nanjing Medical University, Nanjing 211166, China
- Gusu School, Nanjing Medical University, Nanjing 211166, China
| | - Chunhui Ni
- Department of Occupational Medical and Environmental Health, Key Laboratory of Modern Toxicology of Ministry of Education, School of Public Health, Nanjing Medical University, Nanjing 211166, China
| |
Collapse
|
58
|
Grund JC, Krammer S, Yang Z, Mitländer H, Rauh M, Zirlik S, Kiefer A, Zimmermann T, Rieker RJ, Geppert CI, Papadopoulos NG, Finotto S. Vitamin D 3 resolved human and experimental asthma via B lymphocyte-induced maturation protein 1 in T cells and innate lymphoid cells. THE JOURNAL OF ALLERGY AND CLINICAL IMMUNOLOGY. GLOBAL 2023; 2:100099. [PMID: 37779516 PMCID: PMC10510005 DOI: 10.1016/j.jacig.2023.100099] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/07/2022] [Revised: 02/15/2023] [Accepted: 03/04/2023] [Indexed: 10/03/2023]
Abstract
Background Vitamin D3 (VitD3) is known to have immunomodulatory functions, and VitD3 deficiency is associated with more severe asthma. Objective We aimed to assess the immunoregulatory effects of VitD3 food supplementation on asthma manifestation, with particular focus on T cells and type 2 innate lymphoid cells. Methods Preschool children and adult asthmatic cohorts were analyzed in the context of VitD3 supplementation and serum levels. In a murine model of ovalbumin-induced asthma, effects of diet VitD3 sufficiency and deficiency on T cells and type 2 innate lymphoid cells immune mechanisms were investigated. Results We found less severe and better-controlled asthma phenotypes along with reduced need for steroid medication in preschool children and asthmatic adults with VitD3 supplementation. VitD3 serum levels correlated with B lymphocyte-induced maturation protein 1 (Blimp-1) expression in blood peripheral mononuclear cells. VitD3-supplement-fed mice showed decreased asthmatic traits, with a decrease in IgE serum levels, reduced airway mucus, and increased IL-10 production by lung cells. Furthermore, we discovered an upregulation of effector T cells and Blimp-1+ lung tissue-resident memory T cells as well as induction of anti-inflammatory Blimp-1+ lung innate lymphoid cells producing IL-10. Conclusion Supplementing VitD3 resulted in amelioration of clinical asthma manifestations in human studies as well as in experimental allergic asthma, indicating that VitD3 shifts proinflammatory immune responses to anti-inflammatory immune responses via upregulating Blimp-1 in lung innate lymphoid cells and tissue-resident memory cells.
Collapse
Affiliation(s)
- Janina C. Grund
- Department of Molecular Pneumology, Universitätsklinikum Erlangen, Erlangen, Germany
| | - Susanne Krammer
- Department of Molecular Pneumology, Universitätsklinikum Erlangen, Erlangen, Germany
| | - Zuqin Yang
- Department of Molecular Pneumology, Universitätsklinikum Erlangen, Erlangen, Germany
| | - Hannah Mitländer
- Department of Molecular Pneumology, Universitätsklinikum Erlangen, Erlangen, Germany
| | - Manfred Rauh
- Department of Clinical Laboratories, Universitätsklinikum Erlangen, Erlangen, Germany
| | - Sabine Zirlik
- I Medical Clinic, Universitätsklinikum Erlangen, Erlangen, Germany
| | - Alexander Kiefer
- Department of Allergy and Pneumology of the Children’s Hospital, Universitätsklinikum Erlangen, Erlangen, Germany
| | - Theodor Zimmermann
- Department of Allergy and Pneumology of the Children’s Hospital, Universitätsklinikum Erlangen, Erlangen, Germany
| | - Ralf J. Rieker
- Institute of Pathology, Friedrich-Alexander-University Erlangen-Nürnberg, Universitätsklinikum Erlangen, Erlangen, Germany
- Comprehensive Cancer Center Erlangen-EMN (CCC ER-EMN), Erlangen
| | - Carol I. Geppert
- Institute of Pathology, Friedrich-Alexander-University Erlangen-Nürnberg, Universitätsklinikum Erlangen, Erlangen, Germany
- Comprehensive Cancer Center Erlangen-EMN (CCC ER-EMN), Erlangen
| | - Nikolaos G. Papadopoulos
- Allergy and Clinical Immunology Unit, 2nd Pediatric Clinic, National and Kapodistrian University of Athens, Athens, Greece
- Centre for Respiratory Medicine & Allergy, Division of Infection, Immunity & Respiratory Medicine, University of Manchester, Manchester, United Kingdom
| | - Susetta Finotto
- Department of Molecular Pneumology, Universitätsklinikum Erlangen, Erlangen, Germany
- Comprehensive Cancer Center Erlangen-EMN (CCC ER-EMN), Erlangen
| |
Collapse
|
59
|
Kang MH, Hong J, Lee J, Cha MS, Lee S, Kim HY, Ha SJ, Lim YT, Bae YS. Discovery of highly immunogenic spleen-resident FCGR3 +CD103 + cDC1s differentiated by IL-33-primed ST2 + basophils. Cell Mol Immunol 2023; 20:820-834. [PMID: 37246159 PMCID: PMC10310784 DOI: 10.1038/s41423-023-01035-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2022] [Accepted: 04/25/2023] [Indexed: 05/30/2023] Open
Abstract
Recombinant interleukin-33 (IL-33) inhibits tumor growth, but the detailed immunological mechanism is still unknown. IL-33-mediated tumor suppression did not occur in Batf3-/- mice, indicating that conventional type 1 dendritic cells (cDC1s) play a key role in IL-33-mediated antitumor immunity. A population of CD103+ cDC1s, which were barely detectable in the spleens of normal mice, increased significantly in the spleens of IL-33-treated mice. The newly emerged splenic CD103+ cDC1s were distinct from conventional splenic cDC1s based on their spleen residency, robust effector T-cell priming ability, and surface expression of FCGR3. DCs and DC precursors did not express Suppressor of Tumorigenicity 2 (ST2). However, recombinant IL-33 induced spleen-resident FCGR3+CD103+ cDC1s, which were found to be differentiated from DC precursors by bystander ST2+ immune cells. Through immune cell fractionation and depletion assays, we found that IL-33-primed ST2+ basophils play a crucial role in the development of FCGR3+CD103+ cDC1s by secreting IL-33-driven extrinsic factors. Recombinant GM-CSF also induced the population of CD103+ cDC1s, but the population neither expressed FCGR3 nor induced any discernable antitumor immunity. The population of FCGR3+CD103+ cDC1s was also generated in vitro culture of Flt3L-mediated bone marrow-derived DCs (FL-BMDCs) when IL-33 was added in a pre-DC stage of culture. FL-BMDCs generated in the presence of IL-33 (FL-33-DCs) offered more potent tumor immunotherapy than control Flt3L-BMDCs (FL-DCs). Human monocyte-derived DCs were also more immunogenic when exposed to IL-33-induced factors. Our findings suggest that recombinant IL-33 or an IL-33-mediated DC vaccine could be an attractive protocol for better tumor immunotherapy.
Collapse
Affiliation(s)
- Myeong-Ho Kang
- Department of Biological Sciences, Sungkyunkwan University, 2066 Seobu-ro, Jangan-gu, Suwon-si, Gyeonggi-do, 16419, Republic of Korea
- Center for Immune Research on Non-Lymphoid Organs, Sungkyunkwan University, 2066 Seobu-ro, Jangan-gu, Suwon-si, Gyounggi-do, 16419, Republic of Korea
| | - JungHyub Hong
- Department of Biological Sciences, Sungkyunkwan University, 2066 Seobu-ro, Jangan-gu, Suwon-si, Gyeonggi-do, 16419, Republic of Korea
- Center for Immune Research on Non-Lymphoid Organs, Sungkyunkwan University, 2066 Seobu-ro, Jangan-gu, Suwon-si, Gyounggi-do, 16419, Republic of Korea
| | - Jinjoo Lee
- Department of Biological Sciences, Sungkyunkwan University, 2066 Seobu-ro, Jangan-gu, Suwon-si, Gyeonggi-do, 16419, Republic of Korea
- Center for Immune Research on Non-Lymphoid Organs, Sungkyunkwan University, 2066 Seobu-ro, Jangan-gu, Suwon-si, Gyounggi-do, 16419, Republic of Korea
| | - Min-Suk Cha
- Department of Biological Sciences, Sungkyunkwan University, 2066 Seobu-ro, Jangan-gu, Suwon-si, Gyeonggi-do, 16419, Republic of Korea
- Center for Immune Research on Non-Lymphoid Organs, Sungkyunkwan University, 2066 Seobu-ro, Jangan-gu, Suwon-si, Gyounggi-do, 16419, Republic of Korea
| | - Sangho Lee
- Department of Biological Sciences, Sungkyunkwan University, 2066 Seobu-ro, Jangan-gu, Suwon-si, Gyeonggi-do, 16419, Republic of Korea
- Center for Immune Research on Non-Lymphoid Organs, Sungkyunkwan University, 2066 Seobu-ro, Jangan-gu, Suwon-si, Gyounggi-do, 16419, Republic of Korea
| | - Hye-Young Kim
- Center for Immune Research on Non-Lymphoid Organs, Sungkyunkwan University, 2066 Seobu-ro, Jangan-gu, Suwon-si, Gyounggi-do, 16419, Republic of Korea
- Laboratory of Mucosal Immunology, Department of Biomedical Sciences, Seoul National University College of Medicine, 103 Daehak-ro, Jongno-gu, Seoul, 03080, Republic of Korea
- Institute of Allergy and Clinical Immunology, Seoul National University Medical Research Center, Seoul, Republic of Korea
| | - Sang-Jun Ha
- Center for Immune Research on Non-Lymphoid Organs, Sungkyunkwan University, 2066 Seobu-ro, Jangan-gu, Suwon-si, Gyounggi-do, 16419, Republic of Korea
- Department of Biochemistry, College of Life Science and Biotechnology, Yonsei University, Seoul, 03722, Republic of Korea
| | - Yong Taik Lim
- Center for Immune Research on Non-Lymphoid Organs, Sungkyunkwan University, 2066 Seobu-ro, Jangan-gu, Suwon-si, Gyounggi-do, 16419, Republic of Korea
- Department of Nano Engineering and School of Chemical Engineering, Sungkyunkwan University, 2066 Seobu-ro, Jangan-gu, Suwon-si, Gyeonggi-do, 16419, Republic of Korea
| | - Yong-Soo Bae
- Department of Biological Sciences, Sungkyunkwan University, 2066 Seobu-ro, Jangan-gu, Suwon-si, Gyeonggi-do, 16419, Republic of Korea.
- Center for Immune Research on Non-Lymphoid Organs, Sungkyunkwan University, 2066 Seobu-ro, Jangan-gu, Suwon-si, Gyounggi-do, 16419, Republic of Korea.
| |
Collapse
|
60
|
Holgado A, Liu Z, Aidarova A, Mueller C, Haegman M, Driege Y, Kreike M, Scott CL, Afonina IS, Beyaert R. A20 is a master switch of IL-33 signaling in macrophages and determines IL-33-induced lung immunity. J Allergy Clin Immunol 2023; 152:244-256.e4. [PMID: 36898482 DOI: 10.1016/j.jaci.2023.02.026] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2022] [Revised: 01/17/2023] [Accepted: 02/06/2023] [Indexed: 03/11/2023]
Abstract
BACKGROUND IL-33 plays a major role in the pathogenesis of allergic diseases such as asthma and atopic dermatitis. On its release from lung epithelial cells, IL-33 primarily drives type 2 immune responses, accompanied by eosinophilia and robust production of IL-4, IL-5, and IL-13. However, several studies show that IL-33 can also drive a type 1 immune response. OBJECTIVE We sought to determine the role of A20 in the regulation of IL-33 signaling in macrophages and IL-33-induced lung immunity. METHODS We studied the immunologic response in lungs of IL-33-treated mice that specifically lack A20 in myeloid cells. We also analyzed IL-33 signaling in A20-deficient bone marrow-derived macrophages. RESULTS IL-33-induced lung innate lymphoid cell type 2 expansion, type 2 cytokine production, and eosinophilia were drastically reduced in the absence of macrophage A20 expression, whereas neutrophils and interstitial macrophages in lungs were increased. In vitro, IL-33-mediated nuclear factor kappa B activation was only weakly affected in A20-deficient macrophages. However, in the absence of A20, IL-33 gained the ability to activate signal transducer and activator of transcription 1 (STAT1) signaling and STAT1-dependent gene expression. Surprisingly, A20-deficient macrophages produced IFN-γ in response to IL-33, which was fully STAT1-dependent. Furthermore, STAT1 deficiency partially restored the ability of IL-33 to induce ILC2 expansion and eosinophilia in myeloid cell-specific A20 knockout mice. CONCLUSIONS We reveal a novel role for A20 as a negative regulator of IL-33-induced STAT1 signaling and IFN-γ production in macrophages, which determines lung immune responses.
Collapse
Affiliation(s)
- Aurora Holgado
- Unit of Molecular Signal Transduction in Inflammation, VIB-UGent Center for Inflammation Research, Ghent, Belgium; Department of Biomedical Molecular Biology, Ghent University, Ghent, Belgium
| | - Zhuangzhuang Liu
- Department of Biomedical Molecular Biology, Ghent University, Ghent, Belgium; Laboratory of Myeloid Cell Biology in Tissue Damage and Inflammation, VIB-UGent Center for Inflammation Research, Ghent, Belgium
| | - Aigerim Aidarova
- Unit of Molecular Signal Transduction in Inflammation, VIB-UGent Center for Inflammation Research, Ghent, Belgium; Department of Biomedical Molecular Biology, Ghent University, Ghent, Belgium
| | - Christina Mueller
- Unit of Molecular Signal Transduction in Inflammation, VIB-UGent Center for Inflammation Research, Ghent, Belgium; Department of Biomedical Molecular Biology, Ghent University, Ghent, Belgium
| | - Mira Haegman
- Unit of Molecular Signal Transduction in Inflammation, VIB-UGent Center for Inflammation Research, Ghent, Belgium; Department of Biomedical Molecular Biology, Ghent University, Ghent, Belgium
| | - Yasmine Driege
- Unit of Molecular Signal Transduction in Inflammation, VIB-UGent Center for Inflammation Research, Ghent, Belgium; Department of Biomedical Molecular Biology, Ghent University, Ghent, Belgium
| | - Marja Kreike
- Unit of Molecular Signal Transduction in Inflammation, VIB-UGent Center for Inflammation Research, Ghent, Belgium; Department of Biomedical Molecular Biology, Ghent University, Ghent, Belgium
| | - Charlotte L Scott
- Department of Biomedical Molecular Biology, Ghent University, Ghent, Belgium; Laboratory of Myeloid Cell Biology in Tissue Damage and Inflammation, VIB-UGent Center for Inflammation Research, Ghent, Belgium
| | - Inna S Afonina
- Unit of Molecular Signal Transduction in Inflammation, VIB-UGent Center for Inflammation Research, Ghent, Belgium; Department of Biomedical Molecular Biology, Ghent University, Ghent, Belgium
| | - Rudi Beyaert
- Unit of Molecular Signal Transduction in Inflammation, VIB-UGent Center for Inflammation Research, Ghent, Belgium; Department of Biomedical Molecular Biology, Ghent University, Ghent, Belgium.
| |
Collapse
|
61
|
Wiley MB, Mehrotra K, Bauer J, Yazici C, Bialkowska AB, Jung B. Acute Pancreatitis: Current Clinical Approaches, Molecular Pathophysiology, and Potential Therapeutics. Pancreas 2023; 52:e335-e343. [PMID: 38127317 PMCID: PMC11913250 DOI: 10.1097/mpa.0000000000002259] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/12/2022] [Accepted: 06/28/2023] [Indexed: 12/23/2023]
Abstract
OBJECTIVE Severe acute pancreatitis (SAP), pancreatic inflammation leading to multiorgan failure, is associated with high morbidity and mortality. There is a critical need to identify novel therapeutic strategies to improve clinical outcomes for SAP patients. MATERIALS AND METHODS A comprehensive literature review was performed to identify current clinical strategies, known molecular pathophysiology, and potential therapeutic targets for SAP. RESULTS Current clinical approaches focus on determining which patients will likely develop SAP. However, therapeutic options are limited to supportive care and fluid resuscitation. The application of a novel 5-cytokine panel accurately predicting disease outcomes in SAP suggests that molecular approaches will improve impact of future clinical trials in AP. CONCLUSIONS Inflammatory outcomes in acute pancreatitis are driven by several unique molecular signals, which compound to promote both local and systemic inflammation. The identification of master cytokine regulators is critical to developing therapeutics, which reduce inflammation through several mechanisms.
Collapse
Affiliation(s)
- Mark B Wiley
- From the Department of Medicine, University of Washington, Seattle, WA
| | - Kunaal Mehrotra
- From the Department of Medicine, University of Washington, Seattle, WA
| | - Jessica Bauer
- From the Department of Medicine, University of Washington, Seattle, WA
| | - Cemal Yazici
- Department of Medicine, University of Illinois Chicago, Chicago, IL
| | - Agnieszka B Bialkowska
- Department of Medicine, Renaissance School of Medicine at Stony Brook University, Stony Brook, NY
| | - Barbara Jung
- From the Department of Medicine, University of Washington, Seattle, WA
| |
Collapse
|
62
|
Kaur H, Kaur G, Ali SA. IL-33's role in the gut immune system: A comprehensive review of its crosstalk and regulation. Life Sci 2023; 327:121868. [PMID: 37330043 DOI: 10.1016/j.lfs.2023.121868] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2023] [Revised: 06/02/2023] [Accepted: 06/14/2023] [Indexed: 06/19/2023]
Abstract
The intestinal tract is the largest immune organ in the human body, comprising a complex network of immune cells and epithelial cells that perform a variety of functions such as nutrient absorption, digestion, and waste excretion. Maintenance of homeostasis and effective responses to injury in the colonic epithelium are crucial for maintaining homeostasis between these two cell types. The onset and perpetuation of gut inflammation, characterizing inflammatory bowel diseases (IBD), are triggered by constitutive dysregulation of cytokine production. IL-33 is a newly characterized cytokine that has emerged as a critical modulator of inflammatory disorders. IL-33 is constitutively expressed in the nuclei of different cell types such as endothelial, epithelial, and fibroblast-like cells. Upon tissue damage or pathogen encounter, IL-33 is released as an alarmin and signals through a heterodimer receptor that consists of serum Stimulation-2 (ST2) and IL-1 receptor accessory protein (IL-1RAcP). IL-33 has the ability to induce Th2 cytokine production and enhance both Th1 and Th2, as well as Th17 immune responses. Exogenous administration of IL-33 in mice caused pathological changes in most mucosal tissues such as the lung and the gastrointestinal (GI) tract associated with increased production of type 2 cytokines and chemokines. In vivo and in vitro, primary studies have exhibited that IL-33 can activate Th2 cells, mast cells, or basophils to produce type 2 cytokines such as IL-4, IL-5, and IL-13. Moreover, several novel cell populations, collectively referred to as "type 2 innate lymphoid cells," were identified as being IL-33 responsive and are thought to be important for initiating type 2 immunity. Nevertheless, the underlying mechanisms by which IL-33 promotes type 2 immunity in the GI tract remain to be fully understood. Recently, it has been discovered that IL-33 plays important roles in regulatory immune responses. Highly suppressive ST2 + FoxP3+ Tregs subsets regulated by IL-33 were identified in several tissues, including lymphoid organs, gut, lung, and adipose tissues. This review aims to comprehensively summarize the current knowledge on IL-33's role in the gut immune system, its crosstalk, and regulation. The article will provide insights into the potential applications of IL-33-based therapies in the treatment of gut inflammatory disorders.
Collapse
Affiliation(s)
- Harpreet Kaur
- Department of Pediatrics, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Gurjeet Kaur
- Centre for Healthy Brain Ageing, School of Psychiatry, University of New South Wales, Sydney, NSW 2052, Australia; Mark Wainwright Analytical Centre, Bioanalytical Mass Spectrometry Facility, University of New South Wales, Sydney, NSW 2052, Australia
| | - Syed Azmal Ali
- Division Proteomics of Stem Cells and Cancer, German Cancer Research Center, 69120 Heidelberg, Germany.
| |
Collapse
|
63
|
Murphy RC, Lai Y, Liu M, Al-Shaikhly T, Altman MC, Altemeier WA, Frevert CW, Debley JS, Piliponsky AM, Ziegler SF, Gharib SA, Hallstrand TS. Distinct Epithelial-Innate Immune Cell Transcriptional Circuits Underlie Airway Hyperresponsiveness in Asthma. Am J Respir Crit Care Med 2023; 207:1565-1575. [PMID: 37212596 PMCID: PMC10273121 DOI: 10.1164/rccm.202209-1707oc] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2022] [Accepted: 03/02/2023] [Indexed: 05/23/2023] Open
Abstract
Rationale: Indirect airway hyperresponsiveness (AHR) is a highly specific feature of asthma, but the underlying mechanisms responsible for driving indirect AHR remain incompletely understood. Objectives: To identify differences in gene expression in epithelial brushings obtained from individuals with asthma who were characterized for indirect AHR in the form of exercise-induced bronchoconstriction (EIB). Methods: RNA-sequencing analysis was performed on epithelial brushings obtained from individuals with asthma with EIB (n = 11) and without EIB (n = 9). Differentially expressed genes (DEGs) between the groups were correlated with measures of airway physiology, sputum inflammatory markers, and airway wall immunopathology. On the basis of these relationships, we examined the effects of primary airway epithelial cells (AECs) and specific epithelial cell-derived cytokines on both mast cells (MCs) and eosinophils (EOS). Measurements and Main Results: We identified 120 DEGs in individuals with and without EIB. Network analyses suggested critical roles for IL-33-, IL-18-, and IFN-γ-related signaling among these DEGs. IL1RL1 expression was positively correlated with the density of MCs in the epithelial compartment, and IL1RL1, IL18R1, and IFNG were positively correlated with the density of intraepithelial EOS. Subsequent ex vivo modeling demonstrated that AECs promote sustained type 2 (T2) inflammation in MCs and enhance IL-33-induced T2 gene expression. Furthermore, EOS increase the expression of IFNG and IL13 in response to both IL-18 and IL-33 as well as exposure to AECs. Conclusions: Circuits involving epithelial interactions with MCs and EOS are closely associated with indirect AHR. Ex vivo modeling indicates that epithelial-dependent regulation of these innate cells may be critical in indirect AHR and modulating T2 and non-T2 inflammation in asthma.
Collapse
Affiliation(s)
- Ryan C. Murphy
- Division of Pulmonary, Critical Care and Sleep
- Center for Lung Biology
| | - Ying Lai
- Division of Pulmonary, Critical Care and Sleep
- Center for Lung Biology
| | - Matthew Liu
- Division of Pulmonary, Critical Care and Sleep
- Center for Lung Biology
| | - Taha Al-Shaikhly
- Division of Allergy and Infectious Diseases, Department of Medicine
- Center for Lung Biology
| | - Matthew C. Altman
- Division of Allergy and Infectious Diseases, Department of Medicine
- Immunology Program, Benaroya Research Institute, Seattle, Washington
| | | | | | - Jason S. Debley
- Division of Pulmonary and Sleep Medicine, Department of Pediatrics, Seattle Children’s Hospital, University of Washington, Seattle, Washington
- Center for Immunity and Immunotherapies, Seattle Children’s Research Institute, Seattle, Washington
| | - Adrian M. Piliponsky
- Center for Immunity and Immunotherapies, Seattle Children’s Research Institute, Seattle, Washington
| | - Steven F. Ziegler
- Immunology Program, Benaroya Research Institute, Seattle, Washington
| | - Sina A. Gharib
- Division of Pulmonary, Critical Care and Sleep
- Center for Lung Biology
| | | |
Collapse
|
64
|
Cook DP, Thomas CM, Wu AY, Rusznak M, Zhang J, Zhou W, Cephus JY, Gibson-Corley KN, Polosukhin VV, Norlander AE, Newcomb DC, Stoltz DA, Peebles RS. Cystic Fibrosis Reprograms Airway Epithelial IL-33 Release and Licenses IL-33-Dependent Inflammation. Am J Respir Crit Care Med 2023; 207:1486-1497. [PMID: 36952660 PMCID: PMC10263140 DOI: 10.1164/rccm.202211-2096oc] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2022] [Accepted: 03/23/2023] [Indexed: 03/25/2023] Open
Abstract
Rationale: Type 2 inflammation has been described in people with cystic fibrosis (CF). Whether loss of CFTR (cystic fibrosis transmembrane conductance regulator) function contributes directly to a type 2 inflammatory response has not been fully defined. Objectives: The potent alarmin IL-33 has emerged as a critical regulator of type 2 inflammation. We tested the hypothesis that CFTR deficiency increases IL-33 expression and/or release and deletion of IL-33 reduces allergen-induced inflammation in the CF lung. Methods: Human airway epithelial cells (AECs) grown from non-CF and CF cell lines and Cftr+/+ and Cftr-/- mice were used in this study. Pulmonary inflammation in Cftr+/+ and Cftr-/- mice with and without IL-33 or ST2 (IL-1 receptor-like 1) germline deletion was determined by histological analysis, BAL, and cytokine analysis. Measurements and Main Results: After allergen challenge, both CF human AECs and Cftr-/- mice had increased IL-33 expression compared with control AECs and Cftr+/+ mice, respectively. DUOX1 (dual oxidase 1) expression was increased in CF human AECs and Cftr-/- mouse lungs compared with control AECs and lungs from Cftr+/+ mice and was necessary for the increased IL-33 release in Cftr-/- mice compared with Cftr+/+ mice. IL-33 stimulation of Cftr-/- CD4+ T cells resulted in increased type 2 cytokine production compared with Cftr+/+ CD4+ T cells. Deletion of IL-33 or ST2 decreased both type 2 inflammation and neutrophil recruitment in Cftr-/- mice compared with Cftr+/+ mice. Conclusions: Absence of CFTR reprograms airway epithelial IL-33 release and licenses IL-33-dependent inflammation. Modulation of the IL-33/ST2 axis represents a novel therapeutic target in CF type 2-high and neutrophilic inflammation.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - Katherine N. Gibson-Corley
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, Nashville, Tennessee
| | | | | | - Dawn C. Newcomb
- Department of Internal Medicine and
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, Nashville, Tennessee
| | - David A. Stoltz
- Department of Internal Medicine and
- Department of Molecular Physiology and Biophysics, University of Iowa, Iowa City, Iowa; and
| | - R. Stokes Peebles
- Department of Internal Medicine and
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, Nashville, Tennessee
- Tennessee Valley Healthcare System, U.S. Department of Veterans Affairs, Nashville, Tennessee
| |
Collapse
|
65
|
Jeon YJ, Gil CH, Won J, Jo A, Kim HJ. Symbiotic microbiome Staphylococcus epidermidis restricts IL-33 production in allergic nasal epithelium via limiting the cellular necroptosis. BMC Microbiol 2023; 23:154. [PMID: 37237381 DOI: 10.1186/s12866-023-02898-7] [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: 08/26/2022] [Accepted: 05/17/2023] [Indexed: 05/28/2023] Open
Abstract
BACKGROUND Allergic rhinitis (AR) is characterized by airway inflammation in nasal mucosa from inhaled allergens and interleukin (IL)-33 is the potent inducer of Th2 inflammation in allergic nasal epithelium. Staphylococcus epidermidis is one of the most abundant colonizers of the healthy human nasal mucosa and might impact the allergen-induced inflammatory responses in the nasal epithelium. Thus, we sought to characterize the mechanism of S. epidermidis regulating Th2 inflammation and IL-33 production in AR nasal mucosa. RESULTS The AR symptoms were alleviated and eosinophilic infiltration, serum IgE levels, and Th2 cytokines were significantly decreased in OVA-sensitized AR mice in response to human nasal commensal S. epidermidis. The inoculation of S. epidermidis to normal human nasal epithelial cells reduced IL-33 and GATA3 transcriptions and also reduced IL-33 and GATA3 expression in AR nasal epithelial (ARNE) cells and the nasal mucosa of AR mice. Our data exhibited that the cellular necroptosis of ARNE cells might be involved in IL-33 production and inoculation of S. epidermidis decreased the phosphorylation of necroptosis enzymes in ARNE cells, which was related to the reduction of IL-33 production. CONCLUSIONS We present that human nasal commensal S. epidermidis reduces allergic inflammation by suppressing IL-33 production in nasal epithelium. Our findings indicate that S. epidermidis serves a role in blocking allergen-induced cellular necroptosis in allergic nasal epithelium which might be a key mechanism of reduction of IL-33 and Th2 inflammation.
Collapse
Affiliation(s)
- Yung Jin Jeon
- Department of Otorhinolaryngology, Gyeongsang National University Hospital, Jinju, Republic of Korea
| | - Chan Hee Gil
- Department of Otorhinolaryngology, Seoul National University College of Medicine, Seoul, Republic of Korea
| | - Jina Won
- Department of Otorhinolaryngology, Seoul National University College of Medicine, Seoul, Republic of Korea
| | - Ara Jo
- Department of Otorhinolaryngology, Seoul National University College of Medicine, Seoul, Republic of Korea
| | - Hyun Jik Kim
- Department of Otorhinolaryngology, Seoul National University College of Medicine, Seoul, Republic of Korea.
- Sensory Organ Research Institute, Seoul National University Medical Research Center, Seoul, Republic of Korea.
- Department of Otorhinolaryngology-Head and Neck Surgery, Seoul National University College of Medicine, 103, Daehak-ro, Jongno-gu, Seoul, 03080, Republic of Korea.
| |
Collapse
|
66
|
Alkubaisi NA, Aziz IM, Alsaleh AN, Alhetheel AF, Almajhdi FN. Molecular Profiling of Inflammatory Mediators in Human Respiratory Syncytial Virus and Human Bocavirus Infection. Genes (Basel) 2023; 14:genes14051101. [PMID: 37239461 DOI: 10.3390/genes14051101] [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: 04/11/2023] [Revised: 05/13/2023] [Accepted: 05/14/2023] [Indexed: 05/28/2023] Open
Abstract
Infections due to human respiratory syncytial virus (HRSV) and human bocavirus (HBoV) can mediate the release of several pro-inflammatory cytokines such as IL-6, IL-8, and TNF-α, which are usually associated with disease severity in children. In this study, the change in the expression profile of cytokines and chemokines were determined during HRSV, HBoV, and HRSV coinfection with HBoV in 75 nasopharyngeal aspirates (NPAs) samples, positive real-time reverse transcriptase PCR Assay (rRT-PCR) for HRSV (n = 36), HBoV (n = 23) infection alone or HRSV coinfection with HBoV (n = 16). The samples were collected from hospitalized children. qPCR-based detection revealed that the levels of IL-6, IL-8, IL-10, IL-13, IL-33, and G-CSF were significantly (p < 0.05) greater in patients than in controls. IL-4, IL-17, GM-CSF, and CCL-5 were significantly elevated in children with HRSV coinfection with HBoV than in other groups (p < 0.05). TNF-α, IL-6, IL-8, IL-10, IL-13, and IL-33 in children with HRSV were significantly increased in severe infections compared to mild infections. Whereas, IL-10, IL-13, and IL-33 were significantly increased in severe infection in compared a mild infection in children with HBoV. Further large-scale investigations involving isolates are needed to enhance our knowledge of the association between viral infections and cytokine expression patterns during the different stages of HRSV and HBoV infection.
Collapse
Affiliation(s)
- Noorah A Alkubaisi
- Department of Botany and Microbiology, College of Science, King Saud University, Riyadh 11451, Saudi Arabia
| | - Ibrahim M Aziz
- Department of Botany and Microbiology, College of Science, King Saud University, Riyadh 11451, Saudi Arabia
| | - Asma N Alsaleh
- Department of Botany and Microbiology, College of Science, King Saud University, Riyadh 11451, Saudi Arabia
| | - Abdulkarim F Alhetheel
- Department of Pathology and Laboratory Medicine, College of Medicine, King Saud University, Riyadh 11451, Saudi Arabia
| | - Fahad N Almajhdi
- Department of Botany and Microbiology, College of Science, King Saud University, Riyadh 11451, Saudi Arabia
| |
Collapse
|
67
|
Soh WT, Zhang J, Hollenberg MD, Vliagoftis H, Rothenberg ME, Sokol CL, Robinson C, Jacquet A. Protease allergens as initiators-regulators of allergic inflammation. Allergy 2023; 78:1148-1168. [PMID: 36794967 PMCID: PMC10159943 DOI: 10.1111/all.15678] [Citation(s) in RCA: 32] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2022] [Revised: 02/05/2023] [Accepted: 02/12/2023] [Indexed: 02/17/2023]
Abstract
Tremendous progress in the last few years has been made to explain how seemingly harmless environmental proteins from different origins can induce potent Th2-biased inflammatory responses. Convergent findings have shown the key roles of allergens displaying proteolytic activity in the initiation and progression of the allergic response. Through their propensity to activate IgE-independent inflammatory pathways, certain allergenic proteases are now considered as initiators for sensitization to themselves and to non-protease allergens. The protease allergens degrade junctional proteins of keratinocytes or airway epithelium to facilitate allergen delivery across the epithelial barrier and their subsequent uptake by antigen-presenting cells. Epithelial injuries mediated by these proteases together with their sensing by protease-activated receptors (PARs) elicit potent inflammatory responses resulting in the release of pro-Th2 cytokines (IL-6, IL-25, IL-1β, TSLP) and danger-associated molecular patterns (DAMPs; IL-33, ATP, uric acid). Recently, protease allergens were shown to cleave the protease sensor domain of IL-33 to produce a super-active form of the alarmin. At the same time, proteolytic cleavage of fibrinogen can trigger TLR4 signaling, and cleavage of various cell surface receptors further shape the Th2 polarization. Remarkably, the sensing of protease allergens by nociceptive neurons can represent a primary step in the development of the allergic response. The goal of this review is to highlight the multiple innate immune mechanisms triggered by protease allergens that converge to initiate the allergic response.
Collapse
Affiliation(s)
- Wai Tuck Soh
- Max-Planck-Institute for Multidisciplinary Sciences, Göttingen, Germany
| | - Jihui Zhang
- State Key Laboratory of Microbial Resources, Institute of Microbiology, Chinese Academy of Sciences, Beijing, China
| | - Morley D. Hollenberg
- Department of Physiology & Pharmacology, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
- Department of Medicine, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
| | - Harissios Vliagoftis
- Division of Pulmonary Medicine, Department of Medicine, Faculty of Medicine & Dentistry, and Alberta Respiratory Centre, University of Alberta, Edmonton, Alberta, Canada
| | - Marc E. Rothenberg
- Division of Allergy and Immunology, Department of Pediatrics, Cincinnati Children’s Hospital Medical Center, University of Cincinnati College of Medicine, Cincinnati, Ohio, USA
| | - Caroline L. Sokol
- Division of Rheumatology, Allergy and Immunology, Center for Immunology and Inflammatory Diseases, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Clive Robinson
- Institute for Infection and Immunity, St George’s University of London, London, UK
| | - Alain Jacquet
- Department of Biochemistry, Faculty of Medicine, Chulalongkorn University, Bangkok, Thailand
| |
Collapse
|
68
|
Rosenberg L, Liu C, Sharma R, Wood C, Vyhlidal CA, Gaedigk R, Kho AT, Ziniti JP, Celedón JC, Tantisira KG, Weiss ST, McGeachie MJ, Kechris K, Sharma S. Intrauterine Smoke Exposure, microRNA Expression during Human Lung Development, and Childhood Asthma. Int J Mol Sci 2023; 24:7727. [PMID: 37175432 PMCID: PMC10178351 DOI: 10.3390/ijms24097727] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2023] [Revised: 04/14/2023] [Accepted: 04/20/2023] [Indexed: 05/15/2023] Open
Abstract
Intrauterine smoke (IUS) exposure during early childhood has been associated with a number of negative health consequences, including reduced lung function and asthma susceptibility. The biological mechanisms underlying these associations have not been established. MicroRNAs regulate the expression of numerous genes involved in lung development. Thus, investigation of the impact of IUS on miRNA expression during human lung development may elucidate the impact of IUS on post-natal respiratory outcomes. We sought to investigate the effect of IUS exposure on miRNA expression during early lung development. We hypothesized that miRNA-mRNA networks are dysregulated by IUS during human lung development and that these miRNAs may be associated with future risk of asthma and allergy. Human fetal lung samples from a prenatal tissue retrieval program were tested for differential miRNA expression with IUS exposure (measured using placental cotinine concentration). RNA was extracted and miRNA-sequencing was performed. We performed differential expression using IUS exposure, with covariate adjustment. We also considered the above model with an additional sex-by-IUS interaction term, allowing IUS effects to differ by male and female samples. Using paired gene expression profiles, we created sex-stratified miRNA-mRNA correlation networks predictive of IUS using DIABLO. We additionally evaluated whether miRNAs were associated with asthma and allergy outcomes in a cohort of childhood asthma. We profiled pseudoglandular lung miRNA in n = 298 samples, 139 (47%) of which had evidence of IUS exposure. Of 515 miRNAs, 25 were significantly associated with intrauterine smoke exposure (q-value < 0.10). The IUS associated miRNAs were correlated with well-known asthma genes (e.g., ORM1-Like Protein 3, ORDML3) and enriched in disease-relevant pathways (oxidative stress). Eleven IUS-miRNAs were also correlated with clinical measures (e.g., Immunoglobulin E andlungfunction) in children with asthma, further supporting their likely disease relevance. Lastly, we found substantial differences in IUS effects by sex, finding 95 significant IUS-miRNAs in male samples, but only four miRNAs in female samples. The miRNA-mRNA correlation networks were predictive of IUS (AUC = 0.78 in males and 0.86 in females) and suggested that IUS-miRNAs are involved in regulation of disease-relevant genes (e.g., A disintegrin and metalloproteinase domain 19 (ADAM19), LBH regulator of WNT signaling (LBH)) and sex hormone signaling (Coactivator associated methyltransferase 1(CARM1)). Our study demonstrated differential expression of miRNAs by IUS during early prenatal human lung development, which may be modified by sex. Based on their gene targets and correlation to clinical asthma and atopy outcomes, these IUS-miRNAs may be relevant for subsequent allergy and asthma risk. Our study provides insight into the impact of IUS in human fetal lung transcriptional networks and on the developmental origins of asthma and allergic disorders.
Collapse
Affiliation(s)
- Lynne Rosenberg
- Department of Pediatrics and Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA
| | - Cuining Liu
- Department of Biostatistics and Informatics, Colorado School of Public Health, University of Colorado-Anschutz Medical Campus, Aurora, CO 80045, USA
| | - Rinku Sharma
- Channing Division of Network Medicine, Brigham and Women’s Hospital and Harvard Medical School, Boston, MA 02115, USA
| | - Cheyret Wood
- Department of Biostatistics and Informatics, Colorado School of Public Health, University of Colorado-Anschutz Medical Campus, Aurora, CO 80045, USA
| | | | - Roger Gaedigk
- Children’s Mercy Hospital and Clinics, Kansas City, MO 64108, USA
| | - Alvin T. Kho
- Channing Division of Network Medicine, Brigham and Women’s Hospital and Harvard Medical School, Boston, MA 02115, USA
| | - John P. Ziniti
- Channing Division of Network Medicine, Brigham and Women’s Hospital and Harvard Medical School, Boston, MA 02115, USA
| | - Juan C. Celedón
- Division of Pediatric Pulmonary Medicine, University of Pittsburgh School of Medicine, Pittsburgh, PA 15261, USA
| | - Kelan G. Tantisira
- Division of Pediatric Respiratory Medicine, Rady Children’s Hospital, University of California, San Diego, CA 92123, USA
| | - Scott T. Weiss
- Channing Division of Network Medicine, Brigham and Women’s Hospital and Harvard Medical School, Boston, MA 02115, USA
| | - Michael J. McGeachie
- Channing Division of Network Medicine, Brigham and Women’s Hospital and Harvard Medical School, Boston, MA 02115, USA
| | - Katerina Kechris
- Department of Biostatistics and Informatics, Colorado School of Public Health, University of Colorado-Anschutz Medical Campus, Aurora, CO 80045, USA
| | - Sunita Sharma
- Division of Pulmonary Sciences and Critical Care Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA
| |
Collapse
|
69
|
Chen LH, Lo WC, Huang HY, Wu HM. A Lifelong Impact on Endometriosis: Pathophysiology and Pharmacological Treatment. Int J Mol Sci 2023; 24:7503. [PMID: 37108664 PMCID: PMC10139092 DOI: 10.3390/ijms24087503] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2023] [Revised: 04/06/2023] [Accepted: 04/16/2023] [Indexed: 04/29/2023] Open
Abstract
Endometriosis is a chronic inflammatory disease associated with bothersome symptoms in premenopausal women and is complicated with long-term systemic impacts in the post-menopausal stage. It is generally defined by the presence of endometrial-like tissue outside the uterine cavity, which causes menstrual disorders, chronic pelvic pain, and infertility. Endometriotic lesions can also spread and grow in extra-pelvic sites; the chronic inflammatory status can cause systemic effects, including metabolic disorder, immune dysregulation, and cardiovascular diseases. The uncertain etiologies of endometriosis and their diverse presentations limit the treatment efficacy. High recurrence risk and intolerable side effects result in poor compliance. Current studies for endometriosis have paid attention to the advances in hormonal, neurological, and immunological approaches to the pathophysiology and their potential pharmacological intervention. Here we provide an overview of the lifelong impacts of endometriosis and summarize the updated consensus on therapeutic strategies.
Collapse
Affiliation(s)
- Liang-Hsuan Chen
- Department of Obstetrics and Gynecology, Linkou Medical Center, Chang Gung Memorial Hospital, College of Medicine, Chang Gung University, Taoyuan 33305, Taiwan
- Graduate Institute of Clinical Medical Sciences, College of Medicine, Chang Gung University, Taoyuan 33302, Taiwan
| | - Wei-Che Lo
- Department of Obstetrics and Gynecology, Linkou Medical Center, Chang Gung Memorial Hospital, College of Medicine, Chang Gung University, Taoyuan 33305, Taiwan
| | - Hong-Yuan Huang
- Department of Obstetrics and Gynecology, Linkou Medical Center, Chang Gung Memorial Hospital, College of Medicine, Chang Gung University, Taoyuan 33305, Taiwan
| | - Hsien-Ming Wu
- Department of Obstetrics and Gynecology, Linkou Medical Center, Chang Gung Memorial Hospital, College of Medicine, Chang Gung University, Taoyuan 33305, Taiwan
- Graduate Institute of Clinical Medical Sciences, College of Medicine, Chang Gung University, Taoyuan 33302, Taiwan
| |
Collapse
|
70
|
Bochner BS, O'Sullivan JA, Chang AT, Youngblood BA. Siglecs in allergy and asthma. Mol Aspects Med 2023; 90:101104. [PMID: 35835621 PMCID: PMC10757266 DOI: 10.1016/j.mam.2022.101104] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2022] [Revised: 06/28/2022] [Accepted: 07/03/2022] [Indexed: 01/21/2023]
Abstract
The term "allergic diseases" encompasses several common, IgE-mediated conditions that range from being annoying to those that are life-threatening. Available treatments include active avoidance of the instigating allergen and the use of a variety of oral, inhaled, intranasal, intraocular and injected agents. While most individuals with allergies do well with existing therapies, there are still unmet therapeutic needs. Siglecs (sialic acid-binding, immunoglobulin-like lectins) are a family of single-pass transmembrane I-type lectins found on various subsets of cells, especially those of the immune system. All Siglecs have extracellular domains recognizing sialoside ligands, and most contain cytoplasmic domains with inhibitory signaling activity. This review focuses on Siglecs that likely play a role in regulating allergic and asthmatic responses, and how specific Siglecs, expressed on cells such as eosinophils and mast cells, are being targeted for therapeutic benefit.
Collapse
Affiliation(s)
- Bruce S Bochner
- Division of Allergy and Immunology, Department of Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL, USA.
| | - Jeremy A O'Sullivan
- Division of Allergy and Immunology, Department of Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | | | | |
Collapse
|
71
|
Zhao R, Shi Y, Liu N, Li B. Elevated levels of interleukin-33 are associated with asthma: A meta-analysis. Immun Inflamm Dis 2023; 11:e842. [PMID: 37102668 PMCID: PMC10116908 DOI: 10.1002/iid3.842] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2023] [Revised: 03/27/2023] [Accepted: 04/07/2023] [Indexed: 04/28/2023] Open
Abstract
BACKGROUND Previous studies reported that patients with asthma showed higher levels of interleukin (IL)-33 in peripheral blood, compared to healthy control (HCs). However, we also noticed that there were no significant differences of IL-33 levels between controls and asthma patients in a recent study. We aim to conduct this meta-analysis and evaluate the feasibility of IL-33 in peripheral blood that may act as a promising biomarker in asthma. METHODS Articles published before December 2022 were searched in these databases (PubMed, Web of Science, EMBASE, and Google Scholar). We used STATA 12.0 software to compute the results. RESULTS The study showed that asthmatics showed higher IL-33 level in serum and plasma, compared to HCs (serum: standard mean difference [SMD] 2.06, 95% confidence interval [CI] 1.12-3.00, I2 = 98.4%, p < .001; plasma: SMD 3.67, 95% CI 2.32-5.03, I2 = 86.0%, p < .001). Subgroup analysis indicated that asthma adults showed higher IL-33 level in serum, compared to HCs, whereas no significant difference in IL-33 level in serum was showed between asthma children and HCs (adults: SMD 2.17, 95% CI 1.09-3.25; children: SMD 1.81, 95% CI -0.11 to 3.74). The study indicated that moderate and severe asthmatics showed higher IL-33 level in serum, compared to mild asthmatics (SMD 0.78, 95% CI 0.41-1.16, I2 = 66.2%, p = .011). CONCLUSIONS In conclusion, the main findings of present meta-analysis suggested that there was a significant correlation between IL-33 levels and the severity of asthma. Therefore, IL-33 levels of either serum or plasma may be regarded as a useful biomarker of asthma or the degree of disease.
Collapse
Affiliation(s)
- Ranran Zhao
- Department of Respiratory MedicineCapital Medical University Affiliated Beijing Friendship HospitalBeijingChina
| | - Yun Shi
- Medical and Health CenterCapital Medical University Affiliated Beijing Friendship HospitalBeijingChina
| | - Na Liu
- Department of Respiratory MedicineBeijing Hepingli hospitalBeijingChina
| | - Bin Li
- Department of Respiratory MedicineCapital Medical University Affiliated Beijing Friendship HospitalBeijingChina
| |
Collapse
|
72
|
Faiz A, Pavlidis S, Kuo CH, Rowe A, Hiemstra PS, Timens W, Berg M, Wisman M, Guo YK, Djukanović R, Sterk P, Meyer KB, Nawijn MC, Adcock I, Chung KF, van den Berge M. Th2 high and mast cell gene signatures are associated with corticosteroid sensitivity in COPD. Thorax 2023; 78:335-343. [PMID: 36598042 PMCID: PMC10086461 DOI: 10.1136/thorax-2021-217736] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2021] [Accepted: 10/27/2022] [Indexed: 12/12/2022]
Abstract
RATIONALE Severe asthma and chronic obstructive pulmonary disease (COPD) share common pathophysiological traits such as relative corticosteroid insensitivity. We recently published three transcriptome-associated clusters (TACs) using hierarchical analysis of the sputum transcriptome in asthmatics from the Unbiased Biomarkers for the Prediction of Respiratory Disease Outcomes (U-BIOPRED) cohort comprising one Th2-high inflammatory signature (TAC1) and two Th2-low signatures (TAC2 and TAC3). OBJECTIVE We examined whether gene expression signatures obtained in asthma can be used to identify the subgroup of patients with COPD with steroid sensitivity. METHODS Using gene set variation analysis, we examined the distribution and enrichment scores (ES) of the 3 TACs in the transcriptome of bronchial biopsies from 46 patients who participated in the Groningen Leiden Universities Corticosteroids in Obstructive Lung Disease COPD study that received 30 months of treatment with inhaled corticosteroids (ICS) with and without an added long-acting β-agonist (LABA). The identified signatures were then associated with longitudinal clinical variables after treatment. Differential gene expression and cellular convolution were used to define key regulated genes and cell types. MEASUREMENTS AND MAIN RESULTS Bronchial biopsies in patients with COPD at baseline showed a wide range of expression of the 3 TAC signatures. After ICS±LABA treatment, the ES of TAC1 was significantly reduced at 30 months, but those of TAC2 and TAC3 were unaffected. A corticosteroid-sensitive TAC1 signature was developed from the TAC1 ICS-responsive genes. This signature consisted of mast cell-specific genes identified by single-cell RNA-sequencing and positively correlated with bronchial biopsy mast cell numbers following ICS±LABA. Baseline levels of gene transcription correlated with the change in RV/TLC %predicted following 30-month ICS±LABA. CONCLUSION Sputum-derived transcriptomic signatures from an asthma cohort can be recapitulated in bronchial biopsies of patients with COPD and identified a signature of airway mast cells as a predictor of corticosteroid responsiveness.
Collapse
Affiliation(s)
- Alen Faiz
- Respiratory Bioinformatics and Molecular Biology, University of Technology Sydney, Ultimo, New South Wales, Australia
- Pulmonary Diseases, UMCG, Groningen, The Netherlands
- GRAIC, University of Groningen, Groningen, The Netherlands
| | - Stelios Pavlidis
- Department of Computing and Data Science Institute, Imperial College London, London, UK
| | - Chih-Hsi Kuo
- Department of Computing and Data Science Institute, Imperial College London, London, UK
- Airways Disease, Respiratory Cell & Molecular Biology, Airways Disease Section, National Heart and Lung Institute, Faculty of Medicine, Imperial College London, London, UK
| | - Anthony Rowe
- Discovery IT, Janssen Research and Development LLC, High Wycombe, UK
| | - Pieter S Hiemstra
- Pulmonology, Leiden University Medical Center, Leiden, The Netherlands
| | - Wim Timens
- GRAIC, University of Groningen, Groningen, The Netherlands
- Pathology and Medical Biology, University Medical Center Groningen, Groningen, The Netherlands
| | - Marijn Berg
- GRAIC, University of Groningen, Groningen, The Netherlands
- Pathology and Medical Biology, University Medical Center Groningen, Groningen, The Netherlands
| | - Marissa Wisman
- GRAIC, University of Groningen, Groningen, The Netherlands
- Pathology and Medical Biology, University Medical Center Groningen, Groningen, The Netherlands
| | - Yi-Ke Guo
- Department of Computing and Data Science Institute, Imperial College London, London, UK
| | - Ratko Djukanović
- Academic Unit of Clinical and Experimental Sciences, Southampton University Faculty of Medicine, Southampton, UK
| | - Peter Sterk
- Respiratory Medicine, Amsterdam UMC-Locatie AMC, Amsterdam, The Netherlands
| | - Kerstin B Meyer
- Gene expression genomics, Wellcome Sanger Institute, Hinxton, UK
| | - Martijn C Nawijn
- GRAIC, University of Groningen, Groningen, The Netherlands
- Pathology and Medical Biology, University Medical Center Groningen, Groningen, The Netherlands
| | - Ian Adcock
- Department of Computing and Data Science Institute, Imperial College London, London, UK
- Airways Disease, Respiratory Cell & Molecular Biology, Airways Disease Section, National Heart and Lung Institute, Faculty of Medicine, Imperial College London, London, UK
| | - Kian Fan Chung
- Department of Computing and Data Science Institute, Imperial College London, London, UK
- Airways Disease, Respiratory Cell & Molecular Biology, Airways Disease Section, National Heart and Lung Institute, Faculty of Medicine, Imperial College London, London, UK
| | - Maarten van den Berge
- Pulmonary Diseases, UMCG, Groningen, The Netherlands
- GRAIC, University of Groningen, Groningen, The Netherlands
| |
Collapse
|
73
|
Andre AB, Rees KP, O’Connor S, Severson GW, Newbern JM, Wilson-Rawls J, Plaisier CL, Rawls A. Single cell analysis reveals satellite cell heterogeneity for proinflammatory chemokine expression. Front Cell Dev Biol 2023; 11:1084068. [PMID: 37051469 PMCID: PMC10083252 DOI: 10.3389/fcell.2023.1084068] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2022] [Accepted: 03/16/2023] [Indexed: 03/28/2023] Open
Abstract
Background: The expression of proinflammatory signals at the site of muscle injury are essential for efficient tissue repair and their dysregulation can lead to inflammatory myopathies. Macrophages, neutrophils, and fibroadipogenic progenitor cells residing in the muscle are significant sources of proinflammatory cytokines and chemokines. However, the inducibility of the myogenic satellite cell population and their contribution to proinflammatory signaling is less understood.Methods: Mouse satellite cells were isolated and exposed to lipopolysaccharide (LPS) to mimic sterile skeletal muscle injury and changes in the expression of proinflammatory genes was examined by RT-qPCR and single cell RNA sequencing. Expression patterns were validated in skeletal muscle injured with cardiotoxin by RT-qPCR and immunofluorescence.Results: Satellite cells in culture were able to express Tnfa, Ccl2, and Il6, within 2 h of treatment with LPS. Single cell RNA-Seq revealed seven cell clusters representing the continuum from activation to differentiation. LPS treatment led to a heterogeneous pattern of induction of C-C and C-X-C chemokines (e.g., Ccl2, Ccl5, and Cxcl0) and cytokines (e.g., Tgfb1, Bmp2, Il18, and Il33) associated with innate immune cell recruitment and satellite cell proliferation. One cell cluster was enriched for expression of the antiviral interferon pathway genes under control conditions and LPS treatment. Activation of this pathway in satellite cells was also detectable at the site of cardiotoxin induced muscle injury.Conclusion: These data demonstrate that satellite cells respond to inflammatory signals and secrete chemokines and cytokines. Further, we identified a previously unrecognized subset of satellite cells that may act as sensors for muscle infection or injury using the antiviral interferon pathway.
Collapse
Affiliation(s)
- Alexander B. Andre
- School of Life Sciences, Arizona State University, Tempe, AZ, United States
- Molecular and Cellular Biology Graduate Program, Arizona State University, Tempe, AZ, United States
| | - Katherina P. Rees
- School of Life Sciences, Arizona State University, Tempe, AZ, United States
- Molecular and Cellular Biology Graduate Program, Arizona State University, Tempe, AZ, United States
| | - Samantha O’Connor
- School of Biological and Health Systems Engineering, Arizona State University, Tempe, AZ, United States
- Biomedical Engineering Graduate Program, Arizona State University, Tempe, AZ, United States
| | - Grant W. Severson
- School of Life Sciences, Arizona State University, Tempe, AZ, United States
- Molecular and Cellular Biology Graduate Program, Arizona State University, Tempe, AZ, United States
| | - Jason M. Newbern
- School of Life Sciences, Arizona State University, Tempe, AZ, United States
| | | | - Christopher L. Plaisier
- School of Biological and Health Systems Engineering, Arizona State University, Tempe, AZ, United States
| | - Alan Rawls
- School of Life Sciences, Arizona State University, Tempe, AZ, United States
- *Correspondence: Alan Rawls,
| |
Collapse
|
74
|
Yee AJ, Kandasamy J, Ambalavanan N, Ren C, Halloran B, Olave N, Nicola T, Jilling T. Platelet Activating Factor Activity Modulates Hyperoxic Neonatal Lung Injury Severity. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.03.14.532697. [PMID: 36993203 PMCID: PMC10055044 DOI: 10.1101/2023.03.14.532697] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/19/2023]
Abstract
Hyperoxia-induced inflammation contributes significantly to developmental lung injury and bronchopulmonary dysplasia (BPD) in preterm infants. Platelet activating factor (PAF) is known to be a major driver of inflammation in lung diseases such as asthma and pulmonary fibrosis, but its role in BPD has not been previously investigated. Therefore, to determine whether PAF signaling independently modulates neonatal hyperoxic lung injury and BPD pathogenesis, lung structure was assessed in 14 day-old C57BL/6 wild-type (WT) and PAF receptor knockout (PTAFR KO) mice that were exposed to 21% (normoxia) or 85% O 2 (hyperoxia) from postnatal day 4. Lung morphometry showed that PTAFR KO mice had attenuated hyperoxia-induced alveolar simplification when compared to WT mice. Functional analysis of gene expression data from hyperoxia-exposed vs. normoxia-exposed lungs of WT and PTAFR KO showed that the most upregulated pathways were the hypercytokinemia/hyperchemokinemia pathway in WT mice, NAD signaling pathway in PTAFR KO mice, and agranulocyte adhesion and diapedesis as well as other pro-fibrotic pathways such as tumor microenvironment and oncostatin-M signaling in both mice strains, indicating that PAF signaling may contribute to inflammation but may not be a significant mediator of fibrotic processes during hyperoxic neonatal lung injury. Gene expression analysis also indicated increased expression of pro-inflammatory genes such as CXCL1, CCL2 and IL-6 in the lungs of hyperoxia-exposed WT mice and metabolic regulators such as HMGCS2 and SIRT3 in the lungs of PTAFR KO mice, suggesting that PAF signaling may modulate BPD risk through changes in pulmonary inflammation and/or metabolic reprogramming in preterm infants.
Collapse
|
75
|
Pepper AN, Casale TB. Is it time to set the alarmins as potential targets in food allergy? Ann Allergy Asthma Immunol 2023:S1081-1206(23)00167-9. [PMID: 36906261 DOI: 10.1016/j.anai.2023.03.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2023] [Revised: 02/15/2023] [Accepted: 03/02/2023] [Indexed: 03/11/2023]
Affiliation(s)
- Amber N Pepper
- Division of Allergy and Immunology, Department of Internal Medicine, University of South Florida Morsani College of Medicine, Tampa, Florida
| | - Thomas B Casale
- Division of Allergy and Immunology, Department of Internal Medicine, University of South Florida Morsani College of Medicine, Tampa, Florida.
| |
Collapse
|
76
|
Carney AS, Smith PK. Current Understanding of the Role of Eosinophils in CRSwNP and Implications for Treatment with Mepolizumab and Benralizumab. Am J Rhinol Allergy 2023; 37:175-181. [PMID: 36848284 DOI: 10.1177/19458924221149270] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/01/2023]
Abstract
BACKGROUND International consensus statements now subdivide chronic rhinosinusitis (CRS) into several phenotypes and endotypes, including the presence of polyps (CRSwNP) and eosinophilia (eCRSwNP). Biological treatments aimed at blocking eosinophilic inflammation in CRSwNP via interleukin 5 (IL5) or the interleukin 5 receptor (IL5R) have demonstrated limited efficacy thus far. OBJECTIVE To review the pathophysiology of eCRSwNP, the evidence for mepolizumab (anti-IL5) and benralizumab (anti-IL5R) in CRSwNP, and to highlight areas for future research and therapeutic intervention. METHODS Primary and secondary literature search. RESULTS Clinical trials on mepolizumab and benralizumab in CRSwNP are limited and restricted by trial design which prevents direct comparison with other interventions, including surgery. Both agents would appear to provide some benefit in reducing nasal polyp size but limited clinical patient benefit. Molecular biological research highlights that eCRSwNP can occur in the absence of IL5 and that other cells/cytokines play an important part in the disease's pathophysiology. CONCLUSION Blockade of IL5/IL5R alone would appear to provide limited "real life" clinical benefit in patients with CRSwNP due to the complexities of the pathophysiology of the condition. Therapy aimed at several simultaneous cytokine targets has logic but well-designed trials are unlikely to be forthcoming in the short term due to the financial cost and commercial conflicts of interest.
Collapse
Affiliation(s)
- A Simon Carney
- College of Medicine and Public Health, 1065Flinders University, Adelaide, South Australia, Australia
| | - Peter K Smith
- School of Medicine and Dentistry, Griffith University, Southport, Queensland, Australia
| |
Collapse
|
77
|
Zhang R, Yin Z, Pan J, Zhai C, Athari SS, Dong L. Effect of transfected induced pluripotent stem cells with Decorin gene on control of lung remodeling in allergic asthma. J Investig Med 2023; 71:235-243. [PMID: 36803044 DOI: 10.1177/10815589221140590] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/20/2023]
Abstract
Asthma is a complex respiratory disease, which is controlled by genetic and environmental factors. Type 2-dominant immune response is responsible for asthma. Decorin (Dcn) and stem cells have modulatory effect on immune system and may control tissue remodeling and asthma pathophysiology. In this study, immunomodulatory effect of transduced induced pluripotent stem cells (iPSCs) with expression of Dcn gene on allergic asthma pathophysiology was evaluated. After transduction of iPSCs with Dcn gene, allergic asthma mice were treated with iPSCs and transduced iPSCs via intrabronchial. Then, airway hyperresponsiveness (AHR), levels of interleukin (IL)-4, IL-5, IL-13, IL-33, total IgE, leukotrienes (LTs) B4, C4, hydroxyproline (HP) content, and transforming growth factor-beta (TGF-β) were measured. Also, lung histopathology study was done. AHR, levels of IL-4, IL-5, IL-13, IL-33, total IgE, LTs B4, C4, TGF-β, HP content, mucus secretion, goblet cell hyperplasia, and eosinophilic inflammation were controlled by iPSCs and transduced iPSCs treatment. Therapeutic effect of iPSCs could control main allergic asthma symptoms and related pathophysiologic mechanisms and the effect can be increased when applied with Dcn expression gene.
Collapse
Affiliation(s)
- Ronghua Zhang
- Department of Respiratory, Jinan Central Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China.,Department of Respiratory, Central Hospital Affiliated to Shandong First Medical University, Jinan, China
| | - Zongxiu Yin
- Department of Respiratory, Central Hospital Affiliated to Shandong First Medical University, Jinan, China
| | - Jing Pan
- Department of Respiratory, Central Hospital Affiliated to Shandong First Medical University, Jinan, China
| | - Congying Zhai
- Department of Pulmonary and Critical Care Medicine, Zibo First Hospital, Zibo, Shandong, China
| | - Seyyed Shamsadin Athari
- Department of Immunology, School of Medicine, Zanjan University of Medical Sciences, Zanjan, Iran
| | - Liang Dong
- Department of Respiratory, Shandong Provincial Qianfoshan Hospital, Shandong University, The First Affiliated Hospital of Shandong First Medical University, Shandong Institute of Respiratory Diseases, Jinan, China
| |
Collapse
|
78
|
Rush RE, Blackwood CB, Lemons AR, Dannemiller KC, Green BJ, Croston TL. Persisting Cryptococcus yeast species Vishniacozyma victoriae and Cryptococcus neoformans elicit unique airway inflammation in mice following repeated exposure. Front Cell Infect Microbiol 2023; 13:1067475. [PMID: 36864880 PMCID: PMC9971225 DOI: 10.3389/fcimb.2023.1067475] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2022] [Accepted: 01/31/2023] [Indexed: 02/16/2023] Open
Abstract
Background Allergic airway disease (AAD) is a growing concern in industrialized nations and can be influenced by fungal exposures. Basidiomycota yeast species such as Cryptococcus neoformans are known to exacerbate allergic airway disease; however, recent indoor assessments have identified other Basidiomycota yeasts, including Vishniacozyma victoriae (syn. Cryptococcus victoriae), to be prevalent and potentially associated with asthma. Until now, the murine pulmonary immune response to repeated V. victoriae exposure was previously unexplored. Objective This study aimed to compare the immunological impact of repeated pulmonary exposure to Cryptococcus yeasts. Methods Mice were repeatedly exposed to an immunogenic dose of C. neoformans or V. victoriae via oropharyngeal aspiration. Bronchoalveolar lavage fluid (BALF) and lungs were collected to examine airway remodeling, inflammation, mucous production, cellular influx, and cytokine responses at 1 day and 21 days post final exposure. The responses to C. neoformans and V. victoriae were analyzed and compared. Results Following repeated exposure, both C. neoformans and V. victoriae cells were still detectable in the lungs 21 days post final exposure. Repeated C. neoformans exposure initiated myeloid and lymphoid cellular infiltration into the lung that worsened over time, as well as an IL-4 and IL-5 response compared to PBS-exposed controls. In contrast, repeated V. victoriae exposure induced a strong CD4+ T cell-driven lymphoid response that started to resolve by 21 days post final exposure. Discussion C. neoformans remained in the lungs and exacerbated the pulmonary immune responses as expected following repeated exposure. The persistence of V. victoriae in the lung and strong lymphoid response following repeated exposure were unexpected given its lack of reported involvement in AAD. Given the abundance in indoor environments and industrial utilization of V. victoriae, these results highlight the importance to investigate the impact of frequently detected fungal organisms on the pulmonary response following inhalational exposure. Moreover, it is important to continue to address the knowledge gap involving Basidiomycota yeasts and their impact on AAD.
Collapse
Affiliation(s)
- Rachael E. Rush
- Department of Microbiology, Immunology and Cell Biology, West Virginia University, Morgantown, WV, United States
- Health Effects Laboratory Division, National Institute for Occupational Safety and Health, Centers for Disease Control and Prevention, Morgantown, WV, United States
| | - Catherine B. Blackwood
- Health Effects Laboratory Division, National Institute for Occupational Safety and Health, Centers for Disease Control and Prevention, Morgantown, WV, United States
| | - Angela R. Lemons
- Health Effects Laboratory Division, National Institute for Occupational Safety and Health, Centers for Disease Control and Prevention, Morgantown, WV, United States
| | - Karen C. Dannemiller
- Department of Civil, Environmental & Geodetic Engineering, College of Engineering, Ohio State University, Columbus, OH, United States
- Division of Environmental Health Sciences, College of Public Health, Ohio State University, Columbus, OH, United States
| | - Brett J. Green
- Health Effects Laboratory Division, National Institute for Occupational Safety and Health, Centers for Disease Control and Prevention, Morgantown, WV, United States
| | - Tara L. Croston
- Health Effects Laboratory Division, National Institute for Occupational Safety and Health, Centers for Disease Control and Prevention, Morgantown, WV, United States
| |
Collapse
|
79
|
Calderon AA, Dimond C, Choy DF, Pappu R, Grimbaldeston MA, Mohan D, Chung KF. Targeting interleukin-33 and thymic stromal lymphopoietin pathways for novel pulmonary therapeutics in asthma and COPD. Eur Respir Rev 2023; 32:32/167/220144. [PMID: 36697211 PMCID: PMC9879340 DOI: 10.1183/16000617.0144-2022] [Citation(s) in RCA: 44] [Impact Index Per Article: 22.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2022] [Accepted: 11/15/2022] [Indexed: 01/27/2023] Open
Abstract
Interleukin-33 (IL-33) and thymic stromal lymphopoietin (TSLP) are alarmins that are released upon airway epithelial injury from insults such as viruses and cigarette smoke, and play critical roles in the activation of immune cell populations such as mast cells, eosinophils and group 2 innate lymphoid cells. Both cytokines were previously understood to primarily drive type 2 (T2) inflammation, but there is emerging evidence for a role for these alarmins to additionally mediate non-T2 inflammation, with recent clinical trial data in asthma and COPD cohorts with non-T2 inflammation providing support. Currently available treatments for both COPD and asthma provide symptomatic relief with disease control, improving lung function and reducing exacerbation rates; however, there still remains an unmet need for further improving lung function and reducing exacerbations, particularly for those not responsive to currently available treatments. The epithelial cytokines/alarmins are involved in exacerbations; biologics targeting TSLP and IL-33 have been shown to reduce exacerbations in moderate-to-severe asthma, either in a broad population or in specific subgroups, respectively. For COPD, while there is clinical evidence for IL-33 blockade impacting exacerbations in COPD, clinical data from anti-TSLP therapies is awaited. Clinical data to date support an acceptable safety profile for patients with airway diseases for both anti-IL-33 and anti-TSLP antibodies in development. We examine the roles of IL-33 and TSLP, their potential use as drug targets, and the evidence for target patient populations for COPD and asthma, together with ongoing and future trials focused on these targets.
Collapse
Affiliation(s)
| | | | | | | | | | - Divya Mohan
- Genentench, Inc., San Francisco, CA, USA,Corresponding author: Divya Mohan ()
| | - Kian Fan Chung
- National Heart and Lung institute, Imperial College London, London, UK
| |
Collapse
|
80
|
Hsieh A, Assadinia N, Hackett TL. Airway remodeling heterogeneity in asthma and its relationship to disease outcomes. Front Physiol 2023; 14:1113100. [PMID: 36744026 PMCID: PMC9892557 DOI: 10.3389/fphys.2023.1113100] [Citation(s) in RCA: 28] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2022] [Accepted: 01/05/2023] [Indexed: 01/20/2023] Open
Abstract
Asthma affects an estimated 262 million people worldwide and caused over 461,000 deaths in 2019. The disease is characterized by chronic airway inflammation, reversible bronchoconstriction, and airway remodeling. Longitudinal studies have shown that current treatments for asthma (inhaled bronchodilators and corticosteroids) can reduce the frequency of exacerbations, but do not modify disease outcomes over time. Further, longitudinal studies in children to adulthood have shown that these treatments do not improve asthma severity or fixed airflow obstruction over time. In asthma, fixed airflow obstruction is caused by remodeling of the airway wall, but such airway remodeling also significantly contributes to airway closure during bronchoconstriction in acute asthmatic episodes. The goal of the current review is to understand what is known about the heterogeneity of airway remodeling in asthma and how this contributes to the disease process. We provide an overview of the existing knowledge on airway remodeling features observed in asthma, including loss of epithelial integrity, mucous cell metaplasia, extracellular matrix remodeling in both the airways and vessels, angiogenesis, and increased smooth muscle mass. While such studies have provided extensive knowledge on different aspects of airway remodeling, they have relied on biopsy sampling or pathological assessment of lungs from fatal asthma patients, which have limitations for understanding airway heterogeneity and the entire asthma syndrome. To further understand the heterogeneity of airway remodeling in asthma, we highlight the potential of in vivo imaging tools such as computed tomography and magnetic resonance imaging. Such volumetric imaging tools provide the opportunity to assess the heterogeneity of airway remodeling within the whole lung and have led to the novel identification of heterogenous gas trapping and mucus plugging as important predictors of patient outcomes. Lastly, we summarize the current knowledge of modification of airway remodeling with available asthma therapeutics to highlight the need for future studies that use in vivo imaging tools to assess airway remodeling outcomes.
Collapse
Affiliation(s)
- Aileen Hsieh
- Centre for Heart Lung Innovation, St. Paul’s Hospital, Vancouver, BC, Canada,Department of Anesthesiology, Pharmacology and Therapeutics, University of British Columbia, Vancouver, BC, Canada
| | - Najmeh Assadinia
- Centre for Heart Lung Innovation, St. Paul’s Hospital, Vancouver, BC, Canada,Department of Anesthesiology, Pharmacology and Therapeutics, University of British Columbia, Vancouver, BC, Canada
| | - Tillie-Louise Hackett
- Centre for Heart Lung Innovation, St. Paul’s Hospital, Vancouver, BC, Canada,Department of Anesthesiology, Pharmacology and Therapeutics, University of British Columbia, Vancouver, BC, Canada,*Correspondence: Tillie-Louise Hackett,
| |
Collapse
|
81
|
Blomberg OS, Spagnuolo L, Garner H, Voorwerk L, Isaeva OI, van Dyk E, Bakker N, Chalabi M, Klaver C, Duijst M, Kersten K, Brüggemann M, Pastoors D, Hau CS, Vrijland K, Raeven EAM, Kaldenbach D, Kos K, Afonina IS, Kaptein P, Hoes L, Theelen WSME, Baas P, Voest EE, Beyaert R, Thommen DS, Wessels LFA, de Visser KE, Kok M. IL-5-producing CD4 + T cells and eosinophils cooperate to enhance response to immune checkpoint blockade in breast cancer. Cancer Cell 2023; 41:106-123.e10. [PMID: 36525971 DOI: 10.1016/j.ccell.2022.11.014] [Citation(s) in RCA: 107] [Impact Index Per Article: 53.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/01/2022] [Revised: 09/30/2022] [Accepted: 11/23/2022] [Indexed: 12/23/2022]
Abstract
Immune checkpoint blockade (ICB) has heralded a new era in cancer therapy. Research into the mechanisms underlying response to ICB has predominantly focused on T cells; however, effective immune responses require tightly regulated crosstalk between innate and adaptive immune cells. Here, we combine unbiased analysis of blood and tumors from metastatic breast cancer patients treated with ICB with mechanistic studies in mouse models of breast cancer. We observe an increase in systemic and intratumoral eosinophils in patients and mice responding to ICB treatment. Mechanistically, ICB increased IL-5 production by CD4+ T cells, stimulating elevated eosinophil production from the bone marrow, leading to systemic eosinophil expansion. Additional induction of IL-33 by ICB-cisplatin combination or recombinant IL-33 promotes intratumoral eosinophil infiltration and eosinophil-dependent CD8+ T cell activation to enhance ICB response. This work demonstrates the critical role of eosinophils in ICB response and provides proof-of-principle for eosinophil engagement to enhance ICB efficacy.
Collapse
Affiliation(s)
- Olga S Blomberg
- Division of Tumor Biology & Immunology, The Netherlands Cancer Institute, Amsterdam, the Netherlands; Oncode Institute, Utrecht, the Netherlands; Department of Immunology, Leiden University Medical Centre, Leiden, the Netherlands
| | - Lorenzo Spagnuolo
- Division of Tumor Biology & Immunology, The Netherlands Cancer Institute, Amsterdam, the Netherlands; Oncode Institute, Utrecht, the Netherlands
| | - Hannah Garner
- Division of Tumor Biology & Immunology, The Netherlands Cancer Institute, Amsterdam, the Netherlands; Oncode Institute, Utrecht, the Netherlands
| | - Leonie Voorwerk
- Division of Tumor Biology & Immunology, The Netherlands Cancer Institute, Amsterdam, the Netherlands
| | - Olga I Isaeva
- Division of Tumor Biology & Immunology, The Netherlands Cancer Institute, Amsterdam, the Netherlands; Division of Molecular Carcinogenesis, The Netherlands Cancer Institute, Amsterdam, the Netherlands
| | - Ewald van Dyk
- Division of Tumor Biology & Immunology, The Netherlands Cancer Institute, Amsterdam, the Netherlands; Oncode Institute, Utrecht, the Netherlands; Division of Molecular Carcinogenesis, The Netherlands Cancer Institute, Amsterdam, the Netherlands
| | - Noor Bakker
- Division of Tumor Biology & Immunology, The Netherlands Cancer Institute, Amsterdam, the Netherlands; Oncode Institute, Utrecht, the Netherlands
| | - Myriam Chalabi
- Division of Molecular Oncology & Immunology, The Netherlands Cancer Institute, Amsterdam, the Netherlands; Department of Gastrointestinal Oncology, The Netherlands Cancer Institute, Amsterdam, the Netherlands; Department of Medical Oncology, The Netherlands Cancer Institute, Amsterdam, the Netherlands
| | - Chris Klaver
- Division of Tumor Biology & Immunology, The Netherlands Cancer Institute, Amsterdam, the Netherlands
| | - Maxime Duijst
- Division of Tumor Biology & Immunology, The Netherlands Cancer Institute, Amsterdam, the Netherlands
| | - Kelly Kersten
- Division of Tumor Biology & Immunology, The Netherlands Cancer Institute, Amsterdam, the Netherlands
| | - Marieke Brüggemann
- Division of Tumor Biology & Immunology, The Netherlands Cancer Institute, Amsterdam, the Netherlands
| | - Dorien Pastoors
- Division of Tumor Biology & Immunology, The Netherlands Cancer Institute, Amsterdam, the Netherlands; Oncode Institute, Utrecht, the Netherlands
| | - Cheei-Sing Hau
- Division of Tumor Biology & Immunology, The Netherlands Cancer Institute, Amsterdam, the Netherlands; Oncode Institute, Utrecht, the Netherlands
| | - Kim Vrijland
- Division of Tumor Biology & Immunology, The Netherlands Cancer Institute, Amsterdam, the Netherlands; Oncode Institute, Utrecht, the Netherlands
| | - Elisabeth A M Raeven
- Division of Tumor Biology & Immunology, The Netherlands Cancer Institute, Amsterdam, the Netherlands; Oncode Institute, Utrecht, the Netherlands
| | - Daphne Kaldenbach
- Division of Tumor Biology & Immunology, The Netherlands Cancer Institute, Amsterdam, the Netherlands; Oncode Institute, Utrecht, the Netherlands
| | - Kevin Kos
- Division of Tumor Biology & Immunology, The Netherlands Cancer Institute, Amsterdam, the Netherlands; Oncode Institute, Utrecht, the Netherlands; Department of Immunology, Leiden University Medical Centre, Leiden, the Netherlands
| | - Inna S Afonina
- VIB-UGent Center for Inflammation Research, Ghent University, Ghent, Belgium; Department of Biomedical Molecular Biology, Ghent University, Ghent, Belgium
| | - Paulien Kaptein
- Division of Molecular Oncology & Immunology, The Netherlands Cancer Institute, Amsterdam, the Netherlands
| | - Louisa Hoes
- Oncode Institute, Utrecht, the Netherlands; Division of Molecular Oncology & Immunology, The Netherlands Cancer Institute, Amsterdam, the Netherlands; Department of Medical Oncology, The Netherlands Cancer Institute, Amsterdam, the Netherlands
| | - Willemijn S M E Theelen
- Department of Thoracic Oncology, The Netherlands Cancer Institute, Amsterdam, the Netherlands
| | - Paul Baas
- Department of Thoracic Oncology, The Netherlands Cancer Institute, Amsterdam, the Netherlands
| | - Emile E Voest
- Oncode Institute, Utrecht, the Netherlands; Division of Molecular Oncology & Immunology, The Netherlands Cancer Institute, Amsterdam, the Netherlands; Department of Medical Oncology, The Netherlands Cancer Institute, Amsterdam, the Netherlands
| | - Rudi Beyaert
- VIB-UGent Center for Inflammation Research, Ghent University, Ghent, Belgium; Department of Biomedical Molecular Biology, Ghent University, Ghent, Belgium
| | - Daniela S Thommen
- Division of Molecular Oncology & Immunology, The Netherlands Cancer Institute, Amsterdam, the Netherlands
| | - Lodewyk F A Wessels
- Oncode Institute, Utrecht, the Netherlands; Division of Molecular Carcinogenesis, The Netherlands Cancer Institute, Amsterdam, the Netherlands
| | - Karin E de Visser
- Division of Tumor Biology & Immunology, The Netherlands Cancer Institute, Amsterdam, the Netherlands; Oncode Institute, Utrecht, the Netherlands; Department of Immunology, Leiden University Medical Centre, Leiden, the Netherlands.
| | - Marleen Kok
- Division of Tumor Biology & Immunology, The Netherlands Cancer Institute, Amsterdam, the Netherlands; Department of Medical Oncology, The Netherlands Cancer Institute, Amsterdam, the Netherlands.
| |
Collapse
|
82
|
Jiang Q, Sherlock DN, Elolimy AA, Vailati-Riboni M, Yoon I, Loor JJ. Impact of a Saccharomyces cerevisiae fermentation product during an intestinal barrier challenge in lactating Holstein cows on ileal microbiota and markers of tissue structure and immunity. J Anim Sci 2023; 101:skad309. [PMID: 37721866 PMCID: PMC10630188 DOI: 10.1093/jas/skad309] [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: 05/19/2023] [Accepted: 09/16/2023] [Indexed: 09/20/2023] Open
Abstract
Feeding a Saccharomyces cerevisiae fermentation product (SCFP; NutriTek, Diamond V, Cedar Rapids, IA) during periods of metabolic stress is beneficial to the health of dairy cows partially through its effect on the gut microbiota. Whether SCFP alters the ileal microbiota in lactating cows during intestinal challenges induced by feed restriction (FR) is not known. We used 16S rRNA sequencing to assess if feeding SCFP during FR to induce gut barrier dysfunction alters microbiota profiles in the ileum. The mRNA abundance of key genes associated with tissue structures and immunity was also detected. Multiparous cows (97.1 ± 7.6 days in milk (DIM); n = 7 per treatment) fed a control diet or the control plus 19 g/d NutriTek for 9 wk were subjected to an FR challenge for 5 d, during which they were fed 40% of their ad libitum intake from the 7 d before FR. All cows were slaughtered at the end of FR. DNA extracted from ileal digesta was subjected to PacBio Full-Length 16S rRNA gene sequencing. High-quality amplicon sequence analyses were performed with Targeted Amplicon Diversity Analysis and MicrobiomeAnalyst. Functional analysis was performed and analyzed using PICRUSt and STAMP. Feeding SCFP did not (P > 0.05) alter dry matter intake, milk yield, or milk components during FR. In addition, SCFP supplementation tended (P = 0.07) to increase the relative abundance of Proteobacteria and Bifidobacterium animalis. Compared with controls, feeding SCFP increased the relative abundance of Lactobacillales (P = 0.03). Gluconokinase, oligosaccharide reducing-end xylanase, and 3-hydroxy acid dehydrogenase were among the enzymes overrepresented (P < 0.05) in response to feeding SCFP. Cows fed SCFP had a lower representation of adenosylcobalamin biosynthesis I (early cobalt insertion) and pyrimidine deoxyribonucleotides de novo biosynthesis III (P < 0.05). Subsets of the Firmicutes genus, Bacteroidota phylum, and Treponema genus were correlated with the mRNA abundance of genes associated with ileal integrity (GCNT3, GALNT5, B3GNT3, FN1, ITGA2, LAMB2) and inflammation (AOX1, GPX8, CXCL12, CXCL14, CCL4, SAA3). Our data indicated that the moderate FR induced dysfunction of the ileal microbiome, but feeding SCFP increased the abundance of some beneficial gut probiotic bacteria and other species related to tissue structures and immunity.
Collapse
Affiliation(s)
- Qianming Jiang
- Department of Animal Sciences, University of Illinois, Urbana, IL 61801, USA
| | - Danielle N Sherlock
- Department of Animal Sciences, University of Illinois, Urbana, IL 61801, USA
| | - Ahmed A Elolimy
- Animal Production Department, National Research Center, Dokki, Giza 12622, Egypt
| | | | | | - Juan J Loor
- Department of Animal Sciences, University of Illinois, Urbana, IL 61801, USA
| |
Collapse
|
83
|
Chauvin C, Retnakumar SV, Bayry J. Gasdermin D as a cellular switch to orientate immune responses via IL-33 or IL-1β. Cell Mol Immunol 2023; 20:8-10. [PMID: 36380096 PMCID: PMC9664042 DOI: 10.1038/s41423-022-00950-6] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2022] [Accepted: 10/29/2022] [Indexed: 11/16/2022] Open
Affiliation(s)
- Camille Chauvin
- Institut National de la Santé et de la Recherche Médicale, Centre de Recherche des Cordeliers, Sorbonne Université, Université de Paris, 75006, Paris, France
| | - Sruthi Vijaya Retnakumar
- Institut National de la Santé et de la Recherche Médicale, Centre de Recherche des Cordeliers, Sorbonne Université, Université de Paris, 75006, Paris, France
| | - Jagadeesh Bayry
- Institut National de la Santé et de la Recherche Médicale, Centre de Recherche des Cordeliers, Sorbonne Université, Université de Paris, 75006, Paris, France.
- Department of Biological Sciences & Engineering, Indian Institute of Technology Palakkad, Palakkad, 678623, India.
| |
Collapse
|
84
|
Watson MM, van der Giezen M, Søreide K. Gut Microbiome Influence on Human Epigenetics, Health, and Disease. HANDBOOK OF EPIGENETICS 2023:669-686. [DOI: 10.1016/b978-0-323-91909-8.00012-8] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/02/2025]
|
85
|
Banday MM, Rao SB, Shankar S, Khanday MA, Finan J, O'Neill E, Coppolino A, Seyfang A, Kumar A, Rinewalt DE, Goldberg HJ, Woolley A, Mallidi HR, Visner G, Gaggar A, Patel KN, Sharma NS. IL-33 mediates Pseudomonas induced airway fibrogenesis and is associated with CLAD. J Heart Lung Transplant 2023; 42:53-63. [PMID: 37014805 PMCID: PMC10260236 DOI: 10.1016/j.healun.2022.09.018] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2021] [Revised: 09/21/2022] [Accepted: 09/22/2022] [Indexed: 12/23/2022] Open
Abstract
BACKGROUND Long term outcomes of lung transplantation are impacted by the occurrence of chronic lung allograft dysfunction (CLAD). Recent evidence suggests a role for the lung microbiome in the occurrence of CLAD, but the exact mechanisms are not well defined. We hypothesize that the lung microbiome inhibits epithelial autophagic clearance of pro-fibrotic proteins in an IL-33 dependent manner, thereby augmenting fibrogenesis and risk for CLAD. METHODS Autopsy derived CLAD and non-CLAD lungs were collected. IL-33, P62 and LC3 immunofluorescence was performed and assessed using confocal microscopy. Pseudomonas aeruginosa (PsA), Streptococcus Pneumoniae (SP), Prevotella Melaninogenica (PM), recombinant IL-33 or PsA-lipopolysaccharide was co-cultured with primary human bronchial epithelial cells (PBEC) and lung fibroblasts in the presence or absence of IL-33 blockade. Western blot analysis and quantitative reverse transcription (qRT) PCR was performed to evaluate IL-33 expression, autophagy, cytokines and fibroblast differentiation markers. These experiments were repeated after siRNA silencing and upregulation (plasmid vector) of Beclin-1. RESULTS Human CLAD lungs demonstrated markedly increased expression of IL-33 and reduced basal autophagy compared to non-CLAD lungs. Exposure of co-cultured PBECs to PsA, SP induced IL-33, and inhibited PBEC autophagy, while PM elicited no significant response. Further, PsA exposure increased myofibroblast differentiation and collagen formation. IL-33 blockade in these co-cultures recovered Beclin-1, cellular autophagy and attenuated myofibroblast activation in a Beclin-1 dependent manner. CONCLUSION CLAD is associated with increased airway IL-33 expression and reduced basal autophagy. PsA induces a fibrogenic response by inhibiting airway epithelial autophagy in an IL-33 dependent manner.
Collapse
Affiliation(s)
- Mudassir M Banday
- Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts
| | | | - Shruthi Shankar
- University of South Florida, Morsani College of Medicine/Tampa General Hospital
| | | | - Jon Finan
- University of South Florida, Morsani College of Medicine/Tampa General Hospital
| | - Edward O'Neill
- University of South Florida, Morsani College of Medicine/Tampa General Hospital
| | - Antonio Coppolino
- Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts
| | - Andreas Seyfang
- University of South Florida, Morsani College of Medicine/Tampa General Hospital
| | - Archit Kumar
- Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts
| | - Daniel E Rinewalt
- Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts
| | - Hilary J Goldberg
- Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts
| | - Ann Woolley
- Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts
| | - Hari Reddy Mallidi
- Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts
| | - Gary Visner
- Boston Children's Hospital. Harvard Medical School
| | | | - Kapil N Patel
- University of South Florida, Morsani College of Medicine/Tampa General Hospital
| | - Nirmal S Sharma
- Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts; Boston VA Medical Center.
| |
Collapse
|
86
|
Badi YE, Salcman B, Taylor A, Rana B, Kermani NZ, Riley JH, Worsley S, Mumby S, Dahlen S, Cousins D, Bulfone‐Paus S, Affleck K, Chung KF, Bates S, Adcock IM. IL1RAP expression and the enrichment of IL-33 activation signatures in severe neutrophilic asthma. Allergy 2023; 78:156-167. [PMID: 35986608 PMCID: PMC10086999 DOI: 10.1111/all.15487] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2022] [Revised: 07/06/2022] [Accepted: 07/14/2022] [Indexed: 01/09/2023]
Abstract
BACKGROUND Interleukin (IL)-33 is an upstream regulator of type 2 (T2) eosinophilic inflammation and has been proposed as a key driver of some asthma phenotypes. OBJECTIVE To derive gene signatures from in vitro studies of IL-33-stimulated cells and use these to determine IL-33-associated enrichment patterns in asthma. METHODS Signatures downstream of IL-33 stimulation were derived from our in vitro study of human mast cells and from public datasets of in vitro stimulated human basophils, type 2 innate lymphoid cells (ILC2), regulatory T cells (Treg) and endothelial cells. Gene Set Variation Analysis (GSVA) was used to probe U-BIOPRED and ADEPT sputum transcriptomics to determine enrichment scores (ES) for each signature according to asthma severity, sputum granulocyte status and previously defined molecular phenotypes. RESULTS IL-33-activated gene signatures were cell-specific with little gene overlap. Individual signatures, however, were associated with similar signalling pathways (TNF, NF-κB, IL-17 and JAK/STAT signalling) and immune cell differentiation pathways (Th17, Th1 and Th2 differentiation). ES for IL-33-activated gene signatures were significantly enriched in asthmatic sputum, particularly in patients with neutrophilic and mixed granulocytic phenotypes. IL-33 mRNA expression was not elevated in asthma whereas the expression of mRNA for IL1RL1, the IL-33 receptor, was up-regulated in the sputum of severe eosinophilic asthma. The mRNA expression for IL1RAP, the IL1RL1 co-receptor, was greatest in severe neutrophilic and mixed granulocytic asthma. CONCLUSIONS IL-33-activated gene signatures are elevated in neutrophilic and mixed granulocytic asthma corresponding with IL1RAP co-receptor expression. This suggests incorporating T2-low asthma in anti-IL-33 trials.
Collapse
Affiliation(s)
- Yusef Eamon Badi
- National Heart and Lung Institute, Imperial College LondonLondonUK
- Data Science Institute, Imperial College LondonLondonUK
- BenevolentAILondonUK
| | - Barbora Salcman
- School of Biological Sciences, University of ManchesterManchesterUK
| | - Adam Taylor
- GSK Respiratory Therapeutic Area UnitStevenageUK
| | | | | | - John H. Riley
- School of Biological Sciences, University of ManchesterManchesterUK
| | - Sally Worsley
- GSK Value Evidence and Outcomes, GSK HouseBrentfordUK
| | - Sharon Mumby
- National Heart and Lung Institute, Imperial College LondonLondonUK
| | - Sven‐Eric Dahlen
- Institute of Environmental Medicine, Karolinska InstituteStockholmSweden
| | - David Cousins
- Department of Respiratory SciencesUniversity of LeicesterLeicesterUK
| | | | | | - Kian Fan Chung
- National Heart and Lung Institute, Imperial College LondonLondonUK
| | - Stewart Bates
- School of Biological Sciences, University of ManchesterManchesterUK
| | - Ian M. Adcock
- National Heart and Lung Institute, Imperial College LondonLondonUK
| |
Collapse
|
87
|
Teräsjärvi JT, Toivonen L, Mertsola J, Peltola V, He Q. Low serum soluble ST2 level in early childhood is associated with the risk for asthma at 7 years of age. Allergol Int 2023; 72:173-175. [PMID: 35965191 DOI: 10.1016/j.alit.2022.07.002] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2022] [Revised: 06/27/2022] [Accepted: 07/08/2022] [Indexed: 01/25/2023] Open
Affiliation(s)
- Johanna T Teräsjärvi
- Institute of Biomedicine, Research Centre for Infections and Immunity, University of Turku, Turku, Finland
| | - Laura Toivonen
- Department of Paediatrics and Adolescent Medicine, Turku University Hospital and University of Turku, Turku, Finland
| | - Jussi Mertsola
- Department of Paediatrics and Adolescent Medicine, Turku University Hospital and University of Turku, Turku, Finland
| | - Ville Peltola
- Department of Paediatrics and Adolescent Medicine, Turku University Hospital and University of Turku, Turku, Finland
| | - Qiushui He
- Institute of Biomedicine, Research Centre for Infections and Immunity, University of Turku, Turku, Finland; InFLAMES Research Flagship Centre, University of Turku, Turku, Finland.
| |
Collapse
|
88
|
Renjith A, Rajan NS, Shaila SN. Protein and mRNA expression of interleukin-33 in periodontally diseased and healthy individuals and impact of nonsurgical periodontal therapy in salivary IL-33 levels. J Indian Soc Periodontol 2023; 27:45-50. [PMID: 36873980 PMCID: PMC9979825 DOI: 10.4103/jisp.jisp_390_21] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2021] [Revised: 01/12/2022] [Accepted: 02/05/2022] [Indexed: 03/07/2023] Open
Abstract
Background Interleukin (IL)-33 is a member of IL-1 beta family of cytokines having a pivotal role in bone destruction. However, its role in periodontal disease is not clearly established. The objective of the present study was to evaluate salivary and gingival IL-33 expression in periodontally healthy and diseased individuals. The change in salivary IL-33 after nonsurgical therapy was also analyzed. Materials and Methods Salivary IL-33 concentration was estimated using enzyme-linked immunosorbent assay in periodontally healthy and diseased individuals (30 in each group). Re-evaluation was done in periodontitis patients after 6 weeks of nonsurgical therapy. Further, the messenger ribonucleic acid expression of IL-33 in healthy and diseased gingival tissues was also examined using reverse transcriptase-polymerase chain reaction and correlated with IL-1 beta messenger ribonucleic acid. Results Salivary IL-33 concentration in periodontitis patients was 1.65 fold higher than healthy controls (P < 0.0001), and 16% reduction was noticed after nonsurgical treatment. Salivary IL-33 concentration could be used to differentiate periodontitis from health at a cutoff value of 543.16 ng/mL with 93.33% sensitivity and 90% specificity (area under the curve 0.92). Upregulated gingival expression of IL-33 was also noted in periodontitis patients, and it was positively correlated with IL-1 beta (r = 0.7). Conclusion The study reconfirms the role of IL-33 in periodontal disease, proposed a threshold value of differentiating healthy and periodontitis patients, and suggests IL-33 as a potential diagnostic biomarker for periodontal disease and to evaluate the response to periodontal therapy.
Collapse
Affiliation(s)
- Ambili Renjith
- Department of Periodontics, PMS College of Dental Sciences and Research, Thiruvananthapuram, Kerala, India
| | - Nisha Sushama Rajan
- Department of Periodontics, PMS College of Dental Sciences and Research, Thiruvananthapuram, Kerala, India
| | - Shabnas Nazar Shaila
- Department of Periodontics, PMS College of Dental Sciences and Research, Thiruvananthapuram, Kerala, India
| |
Collapse
|
89
|
Lorentz A, Bilotta S, Civelek M. Molecular links between allergy and cancer. Trends Mol Med 2022; 28:1070-1081. [PMID: 35794030 DOI: 10.1016/j.molmed.2022.06.003] [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: 04/21/2022] [Revised: 06/03/2022] [Accepted: 06/03/2022] [Indexed: 01/21/2023]
Abstract
Epidemiologic studies show both positive and negative associations between allergies and cancer. Allergic diseases may protect against tumorigenesis by promoting the immune surveillance, while carcinogenesis may be promoted through inflammatory responses from allergies. Histamine receptor antagonists are the focus of recent cancer studies because of their promising beneficial effect on tumor development. Also, cytokines, particularly IL-4 or IL-33, IgE as well as allergy-related immune cells such as eosinophils can contribute to tumor growth suppression. Depending on cancer types, cancer therapy may be more beneficial when considering combinatorial immunotherapy. In this review, we give an overview on molecular links between allergies and cancer.
Collapse
Affiliation(s)
- Axel Lorentz
- Institute of Nutritional Medicine, University of Hohenheim, 70599 Stuttgart, Germany.
| | - Sabrina Bilotta
- Institute of Nutritional Medicine, University of Hohenheim, 70599 Stuttgart, Germany
| | - Mehtap Civelek
- Institute of Nutritional Medicine, University of Hohenheim, 70599 Stuttgart, Germany
| |
Collapse
|
90
|
Furci F, Murdaca G, Allegra A, Gammeri L, Senna G, Gangemi S. IL-33 and the Cytokine Storm in COVID-19: From a Potential Immunological Relationship towards Precision Medicine. Int J Mol Sci 2022; 23:14532. [PMID: 36498859 PMCID: PMC9740753 DOI: 10.3390/ijms232314532] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2022] [Revised: 11/18/2022] [Accepted: 11/20/2022] [Indexed: 11/23/2022] Open
Abstract
Coronavirus SARS-CoV-2 has represented, and still represents, a real challenge from a clinical, diagnostic and therapeutic point of view. During acute infection, the increased levels of pro-inflammatory cytokines, which are involved in the pathology of disease and the development of SARS-CoV-2-induced acute respiratory disease syndrome, the life-threatening form of this infection, are correlated with patient survival and disease severity. IL-33, a key cytokine involved in both innate and adaptive immune responses in mucosal organs, can increase airway inflammation, mucus secretion and Th2 cytokine synthesis in the lungs, following respiratory infections. Similar to cases of exposure to known respiratory virus infections, exposure to SARS-CoV-2 induces the expression of IL-33, correlating with T-cell activation and lung disease severity. In this work, we analyse current evidence regarding the immunological role of IL-33 in patients affected by COVID-19, to evaluate not only the clinical impact correlated to its production but also to identify possible future immunological therapies that can block the most expressed inflammatory molecules, preventing worsening of the disease and saving patient lives.
Collapse
Affiliation(s)
- Fabiana Furci
- Asthma Centre and Allergy Unit, University of Verona and Verona University Hospital, 37124 Verona, Italy
- Department of Clinical and Experimental Medicine, School and Operative Unit of Allergy and Clinical Immunology, Policlinico G. Martino, University of Messina, 98100 Messina, Italy
| | - Giuseppe Murdaca
- Department of Internal Medicine, University of Genoa, 16126 Genoa, Italy
| | - Alessandro Allegra
- Division of Hematology, Department of Human Pathology in Adulthood and Childhood “Gaetano Barresi”, University of Messina, 98122 Messina, Italy
| | - Luca Gammeri
- Department of Clinical and Experimental Medicine, School and Operative Unit of Allergy and Clinical Immunology, Policlinico G. Martino, University of Messina, 98100 Messina, Italy
| | - Gianenrico Senna
- Asthma Centre and Allergy Unit, University of Verona and Verona University Hospital, 37124 Verona, Italy
- Department of Medicine, University of Verona and Verona University Hospital, 37124 Verona, Italy
| | - Sebastiano Gangemi
- Department of Clinical and Experimental Medicine, School and Operative Unit of Allergy and Clinical Immunology, Policlinico G. Martino, University of Messina, 98100 Messina, Italy
| |
Collapse
|
91
|
Zhang K, Hu Y, Li R, Li T. Single-cell atlas of murine adrenal glands reveals immune-adrenal crosstalk during systemic <i>Candida albicans</i> infection. Front Immunol 2022; 13:966814. [PMID: 36389688 PMCID: PMC9664004 DOI: 10.3389/fimmu.2022.966814] [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: 06/11/2022] [Accepted: 10/10/2022] [Indexed: 11/25/2022] Open
Abstract
Fungal sepsis remains a major health threat with high mortality, where the adrenal gland stress response has been rarely reported. <i>Candida albicans</i> (<i>C.albicans</i>) is the most common opportunistic fungal pathogen of life-threatening disseminated candidiasis and fungal sepsis. In the present study, we performed single-cell RNA sequencing (scRNA-Seq) using the 10x Genomics platform to analyze the changes in murine adrenal transcriptome following systemic <i>C.albicans</i> infection. A total of 16 021 cells were categorized into 18 transcriptionally distinct clusters, representing adrenocortical cells, endothelial cells, various immune cells, mesenchymal cells, smooth muscle cells, adrenal capsule, chromaffin cells, neurons and glials. As the main cell component in the adrenal gland responsible for steroidogenesis, the adrenocortical cells dramatically diminished and were further grouped into 10 subclusters, which differently distributed in the infected and uninfected samples. Pseudo-time analysis revealed transitions of the adrenocortical cells from the initial normal states to active or dysfunctional states following systemic <i>C.albicans</i> infection <i>via</i> two trajectory paths. Endothelial cells in the highly vascularized organ of adrenal gland further proliferated following infection, with the upregulation of genes positively regulating angiogenesis and downregulation of protective genes of endothelial cells. Immune cells were also excessively infiltrated in adrenal glands of <i>C.albicans</i>-infected mice. Macrophages dominated the immune microenvironments in murine adrenal glands both before and after <i>C.albicans</i> infection, mediating the crosstalk among the steroid-producing cells, endothelial cells and immune cells within the adrenal gland. NLR family, pyrin domain containing 3 (NLRP3, encoded by <i>Nlrp3</i>) and complement receptor 3 (CR3, encoded by <i>Itgam</i>) were found to be significantly upregulated on the adrenal macrophages upon systemic <i>C.albicans</i> infection and might play critical roles in mediating the myeloid response. Meanwhile, the number and strength of the interactions between the infiltrating immune cells and adrenal resident cells were unveiled by cell-cell communication analysis to be dramatically increased after systemic <i>C.albicans</i> infection, indicating that the immune-adrenal crosstalk might contribute to the compromised functions of adrenal cells. Overall, our comprehensive picture of the murine adrenal gland microenvironment in systemic <i>C.albicans</i> infection provides deeper insights into the immune-adrenal cell communications during fungal sepsis.
Collapse
Affiliation(s)
- Kai Zhang
- Department of Dermatology and Venerology, Peking University First Hospital, Beijing, China,National Clinical Research Center for Skin and Immune Diseases, Beijing, China,Research Center for Medical Mycology, Peking University, Beijing, China,Beijing Key Laboratory of Molecular Diagnosis on Dermatoses, Beijing, China
| | - Yuzhe Hu
- Department of Immunology, School of Basic Medical Sciences, Peking University Health Science Center, Beijing, China,Key Laboratory of Medical Immunology, National Health Commission of the People's Republic of China, Beijing, China,Peking University Center for Human Disease Genomics, Beijing, China
| | - Ruoyu Li
- Department of Dermatology and Venerology, Peking University First Hospital, Beijing, China,National Clinical Research Center for Skin and Immune Diseases, Beijing, China,Research Center for Medical Mycology, Peking University, Beijing, China,Beijing Key Laboratory of Molecular Diagnosis on Dermatoses, Beijing, China,*Correspondence: Ting Li, ; Ruoyu Li,
| | - Ting Li
- Department of Immunology, School of Basic Medical Sciences, Peking University Health Science Center, Beijing, China,Key Laboratory of Medical Immunology, National Health Commission of the People's Republic of China, Beijing, China,Peking University Center for Human Disease Genomics, Beijing, China,*Correspondence: Ting Li, ; Ruoyu Li,
| |
Collapse
|
92
|
Ghosh S, Rihan M, Ahmed S, Pande AH, Sharma SS. Immunomodulatory potential of apolipoproteins and their mimetic peptides in asthma: Current perspective. Respir Med 2022; 204:107007. [DOI: 10.1016/j.rmed.2022.107007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/01/2022] [Accepted: 10/03/2022] [Indexed: 10/31/2022]
|
93
|
Zhang G, Luo W, Yang W, Li S, Li D, Zeng Y, Li Y. The importance of the
IL
‐1 family of cytokines in nanoimmunosafety and nanotoxicology. WIRES NANOMEDICINE AND NANOBIOTECHNOLOGY 2022; 14:e1850. [DOI: 10.1002/wnan.1850] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/03/2022] [Revised: 08/03/2022] [Accepted: 08/11/2022] [Indexed: 11/24/2022]
Affiliation(s)
- Guofang Zhang
- Laboratory of Immunology and Nanomedicine Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences Shenzhen China
| | - Wenhe Luo
- Laboratory of Immunology and Nanomedicine Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences Shenzhen China
| | - Wenjie Yang
- Laboratory of Immunology and Nanomedicine Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences Shenzhen China
| | - Su Li
- Laboratory of Immunology and Nanomedicine Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences Shenzhen China
| | - Dongjie Li
- Laboratory of Immunology and Nanomedicine Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences Shenzhen China
| | - Yanqiao Zeng
- Laboratory of Immunology and Nanomedicine Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences Shenzhen China
| | - Yang Li
- Laboratory of Immunology and Nanomedicine Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences Shenzhen China
| |
Collapse
|
94
|
Szczepanik-Kułak P, Michalska-Jakubus M, Kowal M, Krasowska D. Serum Levels of Selected IL-1 Family Cytokines in Patients with Morphea. J Clin Med 2022; 11:6375. [PMID: 36362603 PMCID: PMC9655385 DOI: 10.3390/jcm11216375] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2022] [Revised: 09/22/2022] [Accepted: 10/27/2022] [Indexed: 07/27/2023] Open
Abstract
Morphea/localized scleroderma (LoS) represents an inflammatory-sclerotic skin disease, the pathogenesis of which is not fully understood. Given the important role of IL-1 family cytokines in the development and therapy of inflammatory diseases, including systemic sclerosis, we analyzed the clinical significance of serum levels of selected IL-1 family cytokines (IL-1α, IL-1β, IL-18, IL-33, IL-37 and IL-38) in LoS patients (n = 30) using the standardized disease assessment tools and comparison to healthy controls (n = 28). We also compared the pre- and post-treatment concentrations, i.e., before and after systemic (glucocorticosteroids and/or methotrexate) and/or topical (topical glucocorticosteroids and/or calcineurin inhibitors). Our findings did not reveal significant differences in baseline IL-1α, IL-1β, IL-18, IL-33, IL-37 and IL-38 levels between LoS group and HCs; however, after treatment, there were marked changes in concentrations of IL-1α and IL-33 within LoS group as well as in comparison to HCs. We also found significant negative correlations between PGA-A and IL-1α concentration as well as between mLoSSI and IL-1α after treatment. Furthermore, we showed an inverse correlation of baseline IL-1β levels with mLoSSI scores of borderline significance. We believe that IL-1α and IL-33, as well as Il-1β, may be potential mediators and targets of interest in LoS.
Collapse
Affiliation(s)
- Paulina Szczepanik-Kułak
- Chair and Department of Dermatology, Venerology and Paediatric Dermatology, Medical University of Lublin, 20-081 Lublin, Poland
| | | | | | | |
Collapse
|
95
|
Jonckheere AC, Steelant B, Seys SF, Cremer J, Dilissen E, Boon L, Liston A, Schrijvers R, Breynaert C, Vanoirbeek JAJ, Ceuppens JL, Bullens DMA. Peribronchial Inflammation Resulting from Regulatory T Cell Deficiency Damages the Respiratory Epithelium and Disturbs Barrier Function. THE JOURNAL OF IMMUNOLOGY 2022; 209:1595-1605. [DOI: 10.4049/jimmunol.2200416] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/08/2022] [Accepted: 08/10/2022] [Indexed: 01/04/2023]
Abstract
Abstract
Regulatory T cells (Tregs) that express the transcription factor Foxp3 have a critical role in limiting inflammatory processes and tissue damage. Whether Tregs are functional in maintaining epithelial barriers and in control of tight junction expression has not yet been explored. In this study, we investigated the effect of Treg deficiency on the airway epithelial barrier in an experimental murine model in which diphtheria toxin was repeatedly injected in Foxp3-diphtheria toxin receptor (DTR) mice to deplete Tregs. This resulted in spontaneous peribronchial inflammation and led to a systemic and local increase of IL-4, IL-5, CCL3, IFN-γ, and IL-10 and a local (lung) increase of IL-6 and IL-33 and decreased amphiregulin levels. Moreover, Treg depletion increased airway permeability and decreased epithelial tight junction (protein and mRNA) expression. CTLA4-Ig treatment of Treg-depleted mice almost completely prevented barrier dysfunction together with suppression of lung inflammation and cytokine secretion. Treatment with anti–IL-4 partly reversed the effects of Treg depletion on tight junction expression, whereas neutralization of IL-6 of IFN-γ had either no effect or only a limited effect. We conclude that Tregs are essential to protect the epithelial barrier at the level of tight junctions by restricting spontaneous T cell activation and uncontrolled secretion of cytokines, in particular IL-4, in the bronchi.
Collapse
Affiliation(s)
- Anne-Charlotte Jonckheere
- *KU Leuven, Department of Microbiology, Immunology and Transplantation, Allergy and Clinical Immunology Research Group, Leuven, Belgium
| | - Brecht Steelant
- *KU Leuven, Department of Microbiology, Immunology and Transplantation, Allergy and Clinical Immunology Research Group, Leuven, Belgium
| | - Sven F. Seys
- *KU Leuven, Department of Microbiology, Immunology and Transplantation, Allergy and Clinical Immunology Research Group, Leuven, Belgium
| | - Jonathan Cremer
- *KU Leuven, Department of Microbiology, Immunology and Transplantation, Allergy and Clinical Immunology Research Group, Leuven, Belgium
| | - Ellen Dilissen
- *KU Leuven, Department of Microbiology, Immunology and Transplantation, Allergy and Clinical Immunology Research Group, Leuven, Belgium
| | - Louis Boon
- †Polpharma Biologics, Utrecht, the Netherlands
| | - Adrian Liston
- ‡KU Leuven, Department of Microbiology, Immunology and Transplantation, Laboratory of Adaptive Immunity, Leuven, Belgium
| | - Rik Schrijvers
- *KU Leuven, Department of Microbiology, Immunology and Transplantation, Allergy and Clinical Immunology Research Group, Leuven, Belgium
| | - Christine Breynaert
- *KU Leuven, Department of Microbiology, Immunology and Transplantation, Allergy and Clinical Immunology Research Group, Leuven, Belgium
| | - Jeroen A. J. Vanoirbeek
- §KU Leuven, Department of Public Health and Primary Care, Centre for Environment and Health, Leuven, Belgium; and
| | - Jan L. Ceuppens
- *KU Leuven, Department of Microbiology, Immunology and Transplantation, Allergy and Clinical Immunology Research Group, Leuven, Belgium
| | - Dominique M. A. Bullens
- *KU Leuven, Department of Microbiology, Immunology and Transplantation, Allergy and Clinical Immunology Research Group, Leuven, Belgium
- ¶UZ Leuven, Clinical Division of Pediatrics, Leuven, Belgium
| |
Collapse
|
96
|
The Genetic Factors of the Airway Epithelium Associated with the Pathology of Asthma. Genes (Basel) 2022; 13:genes13101870. [PMID: 36292755 PMCID: PMC9601469 DOI: 10.3390/genes13101870] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2022] [Revised: 10/12/2022] [Accepted: 10/14/2022] [Indexed: 11/04/2022] Open
Abstract
Asthma is a chronic disease of the airways characterized by inflammation, tightened muscles, and thickened airway walls leading to symptoms such as shortness of breath, chest tightness, and cough in patients. The increased risk of asthma in children of asthmatics parents supports the existence of genetic factors involved in the pathogenesis of this disease. Genome-wide association studies have discovered several single nucleotide polymorphisms associated with asthma. These polymorphisms occur within several genes and can contribute to different asthma phenotypes, affect disease severity, and clinical response to different therapies. The complexity in the etiology of asthma also results from interactions between environmental and genetic factors. Environmental exposures have been shown to increase the prevalence of asthma in individuals who are genetically susceptible. This review summarizes what is currently known about the genetics of asthma in relation to risk, response to common treatments, and gene-environmental interactions.
Collapse
|
97
|
Jou E, Rodriguez-Rodriguez N, McKenzie ANJ. Emerging roles for IL-25 and IL-33 in colorectal cancer tumorigenesis. Front Immunol 2022; 13:981479. [PMID: 36263033 PMCID: PMC9573978 DOI: 10.3389/fimmu.2022.981479] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2022] [Accepted: 09/15/2022] [Indexed: 12/31/2022] Open
Abstract
Colorectal cancer (CRC) is the second leading cause of cancer-related death worldwide, and is largely refractory to current immunotherapeutic interventions. The lack of efficacy of existing cancer immunotherapies in CRC reflects the complex nature of the unique intestinal immune environment, which serves to maintain barrier integrity against pathogens and harmful environmental stimuli while sustaining host-microbe symbiosis during homeostasis. With their expression by barrier epithelial cells, the cytokines interleukin-25 (IL-25) and IL-33 play key roles in intestinal immune responses, and have been associated with inappropriate allergic reactions, autoimmune diseases and cancer pathology. Studies in the past decade have begun to uncover the important roles of IL-25 and IL-33 in shaping the CRC tumour immune microenvironment, where they may promote or inhibit tumorigenesis depending on the specific CRC subtype. Notably, both IL-25 and IL-33 have been shown to act on group 2 innate lymphoid cells (ILC2s), but can also stimulate an array of other innate and adaptive immune cell types. Though sometimes their functions can overlap they can also produce distinct phenotypes dependent on the differential distribution of their receptor expression. Furthermore, both IL-25 and IL-33 modulate pathways previously known to contribute to CRC tumorigenesis, including angiogenesis, tumour stemness, invasion and metastasis. Here, we review our current understanding of IL-25 and IL-33 in CRC tumorigenesis, with specific focus on dissecting their individual function in the context of distinct subtypes of CRC, and the potential prospects for targeting these pathways in CRC immunotherapy.
Collapse
Affiliation(s)
- Eric Jou
- MRC Laboratory of Molecular Biology, Cambridge, United Kingdom
| | | | | |
Collapse
|
98
|
Jiang W, Chu H, Li Z, Ge J, Wang X, Jiang J, Xiao Q, Meng Q, Lou Y, Hao W, Wei X. Integrated proteomic analysis to explore the molecular regulation mechanism of IL-33 mRNA increased by black carbon in the human endothelial cell line EA.hy926. ENVIRONMENTAL TOXICOLOGY 2022; 37:2434-2444. [PMID: 35776887 DOI: 10.1002/tox.23608] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/15/2022] [Revised: 06/08/2022] [Accepted: 06/15/2022] [Indexed: 06/15/2023]
Abstract
Black carbon (BC) correlates with the occurrence and progression of atherosclerosis and other cardiovascular diseases. Increasing evidence has demonstrated that BC could impair vascular endothelial cells, but the underlying mechanisms remain obscure. It is known that IL-33 exerts a significant biological role in cardiovascular disease, but little is known about the molecular regulation of IL-33 expression at present. We first found that BC significantly increased IL-33 mRNA in EA.hy926 cells in a concentration and time-dependent manner, and we conducted this study to explore its underlying mechanism. We identified that BC induced mitochondrial damage and suppressed autophagy function in EA.hy926 cells, as evidenced by elevation of the aspartate aminotransferase (GOT2), reactive oxygen species (ROS) and p62, and the reduction of mitochondrial membrane potential (ΔΨm). However, ROS cannot induce IL-33 mRNA-production in BC-exposed EA.hy926 cells. Further, experiments revealed that BC could promote IL-33 mRNA production through the PI3K/Akt/AP-1 and p38/AP-1 signaling pathways. It is concluded that BC could induce oxidative stress and suppress autophagy function in endothelial cells. This study also provided evidence that the pro-cardiovascular-diseases properties of BC may be due to its ability to stimulate the PI3K/AKT/AP-1 and p38/AP-1 pathway, further activate IL-33 and ultimately result in a local vascular inflammation.
Collapse
Affiliation(s)
- Wanyu Jiang
- Department of Toxicology, School of Public Health, Peking University, Beijing, People's Republic of China
- Beijing Key Laboratory of Toxicological Research and Risk Assessment for Food Safety, Beijing, People's Republic of China
| | - Hongqian Chu
- Translational Medicine Center, Beijing Chest Hospital, Capital Medical University, Beijing, People's Republic of China
- Beijing Key Laboratory in Drug Resistant Tuberculosis Research, Beijing Tuberculosis and Thoracic Tumor Research Institute, Beijing, People's Republic of China
| | - Zekang Li
- Department of Toxicology, School of Public Health, Peking University, Beijing, People's Republic of China
- Beijing Key Laboratory of Toxicological Research and Risk Assessment for Food Safety, Beijing, People's Republic of China
| | - Jianhong Ge
- Department of Toxicology, School of Public Health, Peking University, Beijing, People's Republic of China
- Beijing Key Laboratory of Toxicological Research and Risk Assessment for Food Safety, Beijing, People's Republic of China
| | - Xiaoyun Wang
- Department of Toxicology, School of Public Health, Peking University, Beijing, People's Republic of China
- Beijing Key Laboratory of Toxicological Research and Risk Assessment for Food Safety, Beijing, People's Republic of China
| | - Jianjun Jiang
- Department of Toxicology, School of Public Health, Peking University, Beijing, People's Republic of China
- Beijing Key Laboratory of Toxicological Research and Risk Assessment for Food Safety, Beijing, People's Republic of China
| | - Qianqian Xiao
- Department of Toxicology, School of Public Health, Peking University, Beijing, People's Republic of China
- Beijing Key Laboratory of Toxicological Research and Risk Assessment for Food Safety, Beijing, People's Republic of China
| | - Qinghe Meng
- Department of Toxicology, School of Public Health, Peking University, Beijing, People's Republic of China
- Beijing Key Laboratory of Toxicological Research and Risk Assessment for Food Safety, Beijing, People's Republic of China
| | - Yaxin Lou
- Medical and Health Analytical Center of Peking University, Beijing, People's Republic of China
| | - Weidong Hao
- Department of Toxicology, School of Public Health, Peking University, Beijing, People's Republic of China
- Beijing Key Laboratory of Toxicological Research and Risk Assessment for Food Safety, Beijing, People's Republic of China
| | - Xuetao Wei
- Department of Toxicology, School of Public Health, Peking University, Beijing, People's Republic of China
- Beijing Key Laboratory of Toxicological Research and Risk Assessment for Food Safety, Beijing, People's Republic of China
| |
Collapse
|
99
|
Comparison of cytokine mediators in type 2 inflammatory conditions on the skin and ocular surface. Curr Opin Allergy Clin Immunol 2022; 22:319-327. [DOI: 10.1097/aci.0000000000000842] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
|
100
|
Tatu AL, Nadasdy T, Arbune A, Chioncel V, Bobeica C, Niculet E, Iancu AV, Dumitru C, Popa VT, Kluger N, Clatici VG, Vasile CI, Onisor C, Nechifor A. Interrelationship and Sequencing of Interleukins4, 13, 31, and 33 - An Integrated Systematic Review: Dermatological and Multidisciplinary Perspectives. J Inflamm Res 2022; 15:5163-5184. [PMID: 36110506 PMCID: PMC9468867 DOI: 10.2147/jir.s374060] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2022] [Accepted: 08/13/2022] [Indexed: 11/23/2022] Open
Abstract
The interrelations and sequencing of interleukins are complex (inter)actions where each interleukin can stimulate the secretion of its preceding interleukin. In this paper, we attempt to summarize the currently known roles of IL-4, IL-13, IL-31, and IL-33 from a multi-disciplinary perspective. In order to conduct a comprehensive review of the current literature, a search was conducted using PubMed, Google Scholar, Medscape, UpToDate, and Key Elsevier for keywords. The results were compiled from case reports, case series, letters, and literature review papers, and analyzed by a panel of multi-disciplinary specialist physicians for relevance. Based on 173 results, we compiled the following review of interleukin signaling and its clinical significance across a multitude of medical specialties. Interleukins are at the bed rock of a multitude of pathologies across different organ systems and understanding their role will likely lead to novel treatments and better outcomes for our patients. New interleukins are being described, and the role of this inflammatory cascade is still coming to light. We hope this multi-discipline review on the role interleukins play in current pathology assists in this scope.
Collapse
Affiliation(s)
- Alin Laurentiu Tatu
- Dermatology Department, "Sf. Cuvioasa Parascheva" Clinical Hospital of Infectious Diseases, Galati, Romania.,Clinical Medical Department, Faculty of Medicine and Pharmacy, "Dunarea de Jos" University, Galati, Romania.,Multidisciplinary Integrated Center of Dermatological Interface Research (MIC-DIR) [Centrul Integrat Multi disciplinar de Cercetare de Interfata Dermatologica (CIM-CID)], Galați, Romania
| | - Thomas Nadasdy
- Multidisciplinary Integrated Center of Dermatological Interface Research (MIC-DIR) [Centrul Integrat Multi disciplinar de Cercetare de Interfata Dermatologica (CIM-CID)], Galați, Romania.,Dermatology Department, Municipal Emergency Hospital, Timişoara, Romania
| | - Anca Arbune
- Neurology Department, Fundeni Clinical Institute, Bucharest, Romania
| | - Valentin Chioncel
- Neurology Department, "Bagdasar-Arseni" Emergency Clinical Hospital, Bucharest, Romania
| | - Carmen Bobeica
- Department of Morphological and Functional Sciences, Faculty of Medicine and Pharmacy, "Dunărea de Jos" University, Galați, Romania
| | - Elena Niculet
- Multidisciplinary Integrated Center of Dermatological Interface Research (MIC-DIR) [Centrul Integrat Multi disciplinar de Cercetare de Interfata Dermatologica (CIM-CID)], Galați, Romania
| | - Alina Viorica Iancu
- Department of Morphological and Functional Sciences, Faculty of Medicine and Pharmacy, "Dunărea de Jos" University, Galați, Romania
| | - Caterina Dumitru
- Pharmaceutical Sciences Department, Faculty of Medicine and Pharmacy, "Dunarea de Jos" University, Galati, Romania
| | - Valentin Tudor Popa
- Multidisciplinary Integrated Center of Dermatological Interface Research (MIC-DIR) [Centrul Integrat Multi disciplinar de Cercetare de Interfata Dermatologica (CIM-CID)], Galați, Romania.,Dermatology Department, Center for the Morphologic Study of the Skin MORPHODERM, "Victor Babeș" University of Medicine and Pharmacy, Timișoara, Romania
| | - Nicolas Kluger
- Department of Dermatology, Allergology and Venereology, Helsinki University Central Hospital and University of Helsinki, Helsinki, Finland.,Apolo Medical Center, Bucharest, Romania
| | | | - Claudiu Ionut Vasile
- Clinical Medical Department, Faculty of Medicine and Pharmacy, "Dunarea de Jos" University, Galati, Romania
| | - Cristian Onisor
- Department of Morphological and Functional Sciences, Faculty of Medicine and Pharmacy, "Dunărea de Jos" University, Galați, Romania
| | - Alexandru Nechifor
- Clinical Medical Department, Faculty of Medicine and Pharmacy, "Dunarea de Jos" University, Galati, Romania
| |
Collapse
|