1
|
Cortez VS, Viragova S, Koga S, Liu M, O'Leary CE, Ricardo-Gonzalez RR, Schroeder AW, Kochhar N, Klein OD, Diamond MS, Liang HE, Locksley RM. IL-25-induced memory ILC2s mediate long-term small intestinal adaptation. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.03.25.645270. [PMID: 40196473 PMCID: PMC11974837 DOI: 10.1101/2025.03.25.645270] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/09/2025]
Abstract
The adaptation of intestinal helminths to vertebrates evolved strategies to attenuate host tissue damage to support reproductive needs of parasites necessary to disseminate offspring to the environment. Helminths initiate the IL-25-mediated tuft cell-ILC2 circuit that enhances barrier protection of the host although viable parasites can target and limit the pathway. We used IL-25 to create small intestinal adaptation marked by anatomic, cell compositional and immunologic changes that persisted months after induction. Small intestinal adaptation was associated with heightened resistance to barrier pathogens, including in the lung, and sustained by transcriptionally and epigenetically modified, tissue-resident, memory-effector ILC2s distinct from those described by innate 'training'; epithelial stem cells remained unaltered. Despite requiring IL-25 for induction, memory ILC2s maintained an activated state in the absence of multiple alarmins and supported mucosal resilience while avoiding adverse sensitization to chronic inflammation, revealing a pathway for deploying innate immune cells to coordinate a distributed mucosal defense.
Collapse
|
2
|
Liu H, Wu M, Wang Q, Gao L, Jiang H, Shi K, Lin Y, Zhou J, Huang J, Qu S, Zhang Y, Zheng F, Huang Y, Han J. IL-33 released during challenge phase regulates allergic asthma in an age-dependent way. Cell Mol Immunol 2025; 22:191-207. [PMID: 39134801 PMCID: PMC11782506 DOI: 10.1038/s41423-024-01205-2] [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: 01/31/2024] [Accepted: 07/12/2024] [Indexed: 02/01/2025] Open
Abstract
Epithelial-derived cytokines, especially type 2 alarmins (TSLP, IL-25, and IL-33), have emerged as critical mediators of type 2 inflammation. IL-33 attracts more interest for its strong association with allergic asthma, especially in childhood asthma. However, the age-dependent role of IL-33 to the development of allergic asthma remains elusive. Here, using OVA-induced allergic asthma model in neonatal and adult mice, we report that IL-33 is the most important alarmin in neonatal lung both at steady state or inflammation. The deficiency of IL-33/ST2 abrogated the development of allergic asthma only in neonates, whereas in adults the effect was limited. Interestingly, the deficiency of IL-33/ST2 equally dampened the ILC2 responses in both neonatal and adult models. However, the effect of IL-33/ST2 deficiency on Th2 responses is age-dependent, which is only blocked in neonates. Furthermore, IL-33/ST2 signaling is dispensable for OVA sensitization. Following OVA challenge in adults, the deficiency of IL-33/ST2 results in compensational more TSLP, which in turn recruits and activates lung DCs and boosts Th2 responses. The enriched γδ T17 cells in IL-33/ST2 deficient neonatal lung suppress the expression of type 2 alarmins, CCL20 and GM-CSF via IL-17A, thus might confer the inhibition of allergic asthma. Finally, on the basis of IL-33 deficiency, the additive protective effects of TSLP blocking is much more pronounced than IL-25 blocking in adults. Our studies demonstrate that the role of IL-33 for ILC2 and Th2 responses varies among ages in OVA models and indicate that the factor of age should be considered for intervention of asthma.
Collapse
Affiliation(s)
- Hangyu Liu
- Department of Immunology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Department of Pathogen Biology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Min Wu
- Department of Gynecologic Oncology, Women's Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Qiangqiang Wang
- Department of Immunology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Liuchuang Gao
- Department of Clinical Laboratory, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, China
- Key Clinical Laboratory of Henan Province, Zhengzhou, 450052, China
| | - Han Jiang
- Department of Pathogen Biology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Ketai Shi
- Department of Immunology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yawen Lin
- Department of Immunology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Junyi Zhou
- Department of Immunology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Ju Huang
- Department of Immunology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Shen Qu
- Department of Pathogen Biology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yuwei Zhang
- Department of Pathogen Biology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Fang Zheng
- Department of Immunology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yafei Huang
- Department of Pathogen Biology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.
- State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious Diseases, Huazhong University of Science and Technology, Wuhan, China.
| | - Junyan Han
- Department of Immunology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.
- Key Laboratory of Vascular Aging, Ministry of Education, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China.
| |
Collapse
|
3
|
Donahue KL, Watkoske HR, Kadiyala P, Du W, Brown K, Scales MK, Elhossiny AM, Espinoza CE, Lasse Opsahl EL, Griffith BD, Wen Y, Sun L, Velez-Delgado A, Renollet NM, Morales J, Nedzesky NM, Baliira RK, Menjivar RE, Medina-Cabrera PI, Rao A, Allen B, Shi J, Frankel TL, Carpenter ES, Bednar F, Zhang Y, Pasca di Magliano M. Oncogenic KRAS-Dependent Stromal Interleukin-33 Directs the Pancreatic Microenvironment to Promote Tumor Growth. Cancer Discov 2024; 14:1964-1989. [PMID: 38958646 PMCID: PMC11450371 DOI: 10.1158/2159-8290.cd-24-0100] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2024] [Revised: 05/18/2024] [Accepted: 07/01/2024] [Indexed: 07/04/2024]
Abstract
Pancreatic cancer is characterized by an extensive fibroinflammatory microenvironment. During carcinogenesis, normal stromal cells are converted to cytokine-high cancer-associated fibroblasts (CAF). The mechanisms underlying this conversion, including the regulation and function of fibroblast-derived cytokines, are poorly understood. Thus, efforts to therapeutically target CAFs have so far failed. Herein, we show that signals from epithelial cells expressing oncogenic KRAS-a hallmark pancreatic cancer mutation-activate fibroblast autocrine signaling, which drives the expression of the cytokine IL33. Stromal IL33 expression remains high and dependent on epithelial KRAS throughout carcinogenesis; in turn, environmental stress induces interleukin-33 (IL33) secretion. Using compartment-specific IL33 knockout mice, we observed that lack of stromal IL33 leads to profound reprogramming of multiple components of the pancreatic tumor microenvironment, including CAFs, myeloid cells, and lymphocytes. Notably, loss of stromal IL33 leads to an increase in CD8+ T-cell infiltration and activation and, ultimately, reduced tumor growth. Significance: This study provides new insights into the mechanisms underlying the programming of CAFs and shows that during this process, expression of the cytokine IL33 is induced. CAF-derived IL33 has pleiotropic effects on the tumor microenvironment, supporting its potential as a therapeutic target.
Collapse
Affiliation(s)
| | - Hannah R. Watkoske
- Department of Surgery, University of Michigan, Ann Arbor, Michigan.
- College of Literature, Science, and the Arts, University of Michigan, Ann Arbor, Michigan.
| | - Padma Kadiyala
- Immunology Graduate Program, University of Michigan, Ann Arbor, Michigan.
| | - Wenting Du
- Department of Surgery, University of Michigan, Ann Arbor, Michigan.
| | - Kristee Brown
- Department of Surgery, University of Michigan, Ann Arbor, Michigan.
| | - Michael K. Scales
- Department of Cell and Developmental Biology, University of Michigan, Ann Arbor, Michigan.
| | - Ahmed M. Elhossiny
- Department of Computational Medicine and Bioinformatics, University of Michigan, Ann Arbor, Michigan.
| | | | | | | | - Yukang Wen
- Department of Surgery, University of Michigan, Ann Arbor, Michigan.
| | - Lei Sun
- Department of Surgery, University of Michigan, Ann Arbor, Michigan.
| | - Ashley Velez-Delgado
- Department of Cell and Developmental Biology, University of Michigan, Ann Arbor, Michigan.
| | - Nur M. Renollet
- College of Literature, Science, and the Arts, University of Michigan, Ann Arbor, Michigan.
| | - Jacqueline Morales
- Department of Cell and Developmental Biology, University of Michigan, Ann Arbor, Michigan.
| | - Nicholas M. Nedzesky
- College of Literature, Science, and the Arts, University of Michigan, Ann Arbor, Michigan.
| | | | - Rosa E. Menjivar
- Department of Surgery, University of Michigan, Ann Arbor, Michigan.
- Cellular and Molecular Biology Program, University of Michigan, Ann Arbor, Michigan.
| | | | - Arvind Rao
- Department of Computational Medicine and Bioinformatics, University of Michigan, Ann Arbor, Michigan.
- Rogel Cancer Center, University of Michigan, Ann Arbor, Michigan.
- Cancer Data Science Resource, University of Michigan, Ann Arbor, Michigan.
- Department of Radiation Oncology, University of Michigan, Ann Arbor, Michigan.
- Department of Biostatistics, University of Michigan, Ann Arbor, Michigan.
| | - Benjamin Allen
- Department of Cell and Developmental Biology, University of Michigan, Ann Arbor, Michigan.
- Rogel Cancer Center, University of Michigan, Ann Arbor, Michigan.
| | - Jiaqi Shi
- Rogel Cancer Center, University of Michigan, Ann Arbor, Michigan.
- Department of Pathology and Clinical Labs, University of Michigan, Ann Arbor, Michigan.
| | - Timothy L. Frankel
- Department of Surgery, University of Michigan, Ann Arbor, Michigan.
- Rogel Cancer Center, University of Michigan, Ann Arbor, Michigan.
| | - Eileen S. Carpenter
- Rogel Cancer Center, University of Michigan, Ann Arbor, Michigan.
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan.
| | - Filip Bednar
- Cancer Biology Program, University of Michigan, Ann Arbor, Michigan.
- Department of Surgery, University of Michigan, Ann Arbor, Michigan.
- Rogel Cancer Center, University of Michigan, Ann Arbor, Michigan.
| | - Yaqing Zhang
- Department of Surgery, University of Michigan, Ann Arbor, Michigan.
- Rogel Cancer Center, University of Michigan, Ann Arbor, Michigan.
| | - Marina Pasca di Magliano
- Department of Surgery, University of Michigan, Ann Arbor, Michigan.
- Department of Cell and Developmental Biology, University of Michigan, Ann Arbor, Michigan.
- Rogel Cancer Center, University of Michigan, Ann Arbor, Michigan.
| |
Collapse
|
4
|
Liu A, Hayashi M, Ohsugi Y, Katagiri S, Akira S, Iwata T, Nakashima T. The IL-33/ST2 axis is protective against acute inflammation during the course of periodontitis. Nat Commun 2024; 15:2707. [PMID: 38548743 PMCID: PMC10978877 DOI: 10.1038/s41467-024-46746-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2023] [Accepted: 03/07/2024] [Indexed: 04/01/2024] Open
Abstract
Periodontitis, which is induced by repeated bacterial invasion and the ensuing immune reactions that follow, is the leading cause of tooth loss. Periodontal tissue is comprised of four different components, each with potential role in pathogenesis, however, most studies on immune responses focus on gingival tissue. Here, we present a modified ligature-induced periodontitis model in male mice to analyze the pathogenesis, which captures the complexity of periodontal tissue. We find that the inflammatory response in the peri-root tissues and the expression of IL-6 and RANKL by Thy-1.2- fibroblasts/stromal cells are prominent throughout the bone destruction phase, and present already at an early stage. The initiation phase is characterized by high levels of ST2 (encoded by Il1rl1) expression in the peri-root tissue, suggesting that the IL-33/ST2 axis is involved in the pathogenesis. Both Il1rl1- and Il33-deficient mice exhibit exacerbated bone loss in the acute phase of periodontitis, along with macrophage polarization towards a classically activated phenotype and increased neutrophil infiltration, indicating a protective role of the IL-33/ST2 axis in acute inflammation. Thus, our findings highlight the hidden role of the peri-root tissue and simultaneously advance our understanding of the etiology of periodontitis via implicating the IL-33/ST2 axis.
Collapse
Affiliation(s)
- Anhao Liu
- Department of Cell Signaling, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, 1-5-45, Yushima, Bunkyo-ku, Tokyo, 113-8549, Japan
- Department of Periodontology, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, 1-5-45, Yushima, Bunkyo-ku, Tokyo, 113-8549, Japan
| | - Mikihito Hayashi
- Department of Cell Signaling, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, 1-5-45, Yushima, Bunkyo-ku, Tokyo, 113-8549, Japan
| | - Yujin Ohsugi
- Department of Periodontology, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, 1-5-45, Yushima, Bunkyo-ku, Tokyo, 113-8549, Japan
| | - Sayaka Katagiri
- Department of Periodontology, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, 1-5-45, Yushima, Bunkyo-ku, Tokyo, 113-8549, Japan
| | - Shizuo Akira
- Laboratory of Host Defense, IFReC,Osaka University, 3-1 Yamadaoka, Suita, Osaka, 565-0871, Japan
| | - Takanori Iwata
- Department of Periodontology, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, 1-5-45, Yushima, Bunkyo-ku, Tokyo, 113-8549, Japan
| | - Tomoki Nakashima
- Faculty of Dentistry, Tokyo Medical and Dental University, 1-5-45, Yushima, Bunkyo-ku, Tokyo, 113-8549, Japan.
| |
Collapse
|
5
|
Kobayashi T, Iijima K, Matsumoto K, Lama JK, Kita H. Lung-resident CD69 +ST2 + T H2 cells mediate long-term type 2 memory to inhaled antigen in mice. J Allergy Clin Immunol 2023; 152:167-181.e6. [PMID: 36720287 PMCID: PMC10330297 DOI: 10.1016/j.jaci.2023.01.016] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2022] [Revised: 01/16/2023] [Accepted: 01/19/2023] [Indexed: 01/30/2023]
Abstract
BACKGROUND Chronic airway diseases such as asthma are characterized by persistent type 2 immunity in the airways. We know little about the mechanisms that explain why type 2 inflammation continues in these diseases. OBJECTIVE We used mouse models to investigate the mechanisms involved in long-lasting immune memory. METHODS Naive mice were exposed intranasally to ovalbumin (OVA) antigen with Alternaria extract as an adjuvant. Type 2 memory was analyzed by parabiosis model, flow cytometry with in vivo antibody labeling, and intranasal OVA recall challenge. Gene-deficient mice were used to analyze the mechanisms. RESULTS In the parabiosis model, mice previously exposed intranasally to OVA with Alternaria showed more robust antigen-specific immune responses and airway inflammation than mice with circulating OVA-specific T cells. After a single airway exposure to OVA with Alternaria, CD69+ST2+ TH2-type T cells, which highly express type 2 cytokine messenger RNA and lack CD62L expression, appeared in lung tissue within 5 days and persisted for at least 84 days. When exposed again to OVA in vivo, these cells produced type 2 cytokines quickly without involving circulating T cells. Development of tissue-resident CD69+ST2+ TH2 cells and long-term memory to an inhaled antigen were abrogated in mice deficient in ST2 or IL-33, but not TSLP receptor. CONCLUSION CD69+ST2+ TH2 memory cells develop quickly in lung tissue after initial allergen exposure and persist for a prolonged period. The ST2/IL-33 pathway may play a role in the development of immune memory in lung to certain allergens.
Collapse
Affiliation(s)
- Takao Kobayashi
- Division of Allergic Diseases, Asthma and Clinical Immunology, and Department of Medicine, Mayo Clinic, Scottsdale, Ariz
| | - Koji Iijima
- Division of Allergic Diseases, Asthma and Clinical Immunology, and Department of Medicine, Mayo Clinic, Scottsdale, Ariz
| | - Koji Matsumoto
- Division of Allergic Diseases, Asthma and Clinical Immunology, and Department of Medicine, Mayo Clinic, Scottsdale, Ariz
| | - Jyoti K Lama
- Immunology Program, Mayo Clinic Graduate School of Biomedical Sciences, Rochester and Scottsdale, Rochester, Minn
| | - Hirohito Kita
- Division of Allergic Diseases, Asthma and Clinical Immunology, and Department of Medicine, Mayo Clinic, Scottsdale, Ariz; Department of Immunology, Mayo Clinic, Rochester, and Mayo Clinic, Scottsdale, Ariz.
| |
Collapse
|
6
|
Hou J, Zhou Y, Cai Z, Terekhova M, Swain A, Andhey PS, Guimaraes RM, Ulezko Antonova A, Qiu T, Sviben S, Strout G, Fitzpatrick JAJ, Chen Y, Gilfillan S, Kim DH, Van Dyken SJ, Artyomov MN, Colonna M. Transcriptomic atlas and interaction networks of brain cells in mouse CNS demyelination and remyelination. Cell Rep 2023; 42:112293. [PMID: 36952346 PMCID: PMC10511667 DOI: 10.1016/j.celrep.2023.112293] [Citation(s) in RCA: 39] [Impact Index Per Article: 19.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2022] [Revised: 10/04/2022] [Accepted: 03/06/2023] [Indexed: 03/24/2023] Open
Abstract
Demyelination is a hallmark of multiple sclerosis, leukoencephalopathies, cerebral vasculopathies, and several neurodegenerative diseases. The cuprizone mouse model is widely used to simulate demyelination and remyelination occurring in these diseases. Here, we present a high-resolution single-nucleus RNA sequencing (snRNA-seq) analysis of gene expression changes across all brain cells in this model. We define demyelination-associated oligodendrocytes (DOLs) and remyelination-associated MAFBhi microglia, as well as astrocytes and vascular cells with signatures of altered metabolism, oxidative stress, and interferon response. Furthermore, snRNA-seq provides insights into how brain cell types connect and interact, defining complex circuitries that impact demyelination and remyelination. As an explicative example, perturbation of microglia caused by TREM2 deficiency indirectly impairs the induction of DOLs. Altogether, this study provides a rich resource for future studies investigating mechanisms underlying demyelinating diseases.
Collapse
Affiliation(s)
- Jinchao Hou
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Yingyue Zhou
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Zhangying Cai
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Marina Terekhova
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Amanda Swain
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Prabhakar S Andhey
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Rafaela M Guimaraes
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO 63110, USA; Ribeirão Preto Medical School, University of São Paulo - Ribeirão Preto, São Paulo 14049-900, Brazil
| | - Alina Ulezko Antonova
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Tian Qiu
- Department of Genetics, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Sanja Sviben
- Washington University Center for Cellular Imaging, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Gregory Strout
- Washington University Center for Cellular Imaging, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - James A J Fitzpatrick
- Washington University Center for Cellular Imaging, Washington University School of Medicine, St. Louis, MO 63110, USA; Departments of Cell Biology and Physiology and Neuroscience, Washington University School of Medicine, St. Louis, MO 63110, USA; Department of Biomedical Engineering, Washington University in St. Louis, St. Louis, MO 63110, USA
| | - Yun Chen
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO 63110, USA; Department of Neurology, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Susan Gilfillan
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Do-Hyun Kim
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Steven J Van Dyken
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Maxim N Artyomov
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Marco Colonna
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO 63110, USA.
| |
Collapse
|
7
|
Ricardo-Gonzalez RR, Kotas ME, O'Leary CE, Singh K, Damsky W, Liao C, Arouge E, Tenvooren I, Marquez DM, Schroeder AW, Cohen JN, Fassett MS, Lee J, Daniel SG, Bittinger K, Díaz RE, Fraser JS, Ali N, Ansel KM, Spitzer MH, Liang HE, Locksley RM. Innate type 2 immunity controls hair follicle commensalism by Demodex mites. Immunity 2022; 55:1891-1908.e12. [PMID: 36044899 PMCID: PMC9561030 DOI: 10.1016/j.immuni.2022.08.001] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2022] [Revised: 06/27/2022] [Accepted: 08/02/2022] [Indexed: 01/05/2023]
Abstract
Demodex mites are commensal parasites of hair follicles (HFs). Normally asymptomatic, inflammatory outgrowth of mites can accompany malnutrition, immune dysfunction, and aging, but mechanisms restricting Demodex outgrowth are not defined. Here, we show that control of mite HF colonization in mice required group 2 innate lymphoid cells (ILC2s), interleukin-13 (IL-13), and its receptor, IL-4Ra-IL-13Ra1. HF-associated ILC2s elaborated IL-13 that attenuated HFs and epithelial proliferation at anagen onset; in their absence, Demodex colonization led to increased epithelial proliferation and replacement of gene programs for repair by aberrant inflammation, leading to the loss of barrier function and HF exhaustion. Humans with rhinophymatous acne rosacea, an inflammatory condition associated with Demodex, had increased HF inflammation with decreased type 2 cytokines, consistent with the inverse relationship seen in mice. Our studies uncover a key role for skin ILC2s and IL-13, which comprise an immune checkpoint that sustains cutaneous integrity and restricts pathologic infestation by colonizing HF mites.
Collapse
Affiliation(s)
- Roberto R Ricardo-Gonzalez
- Department of Dermatology, University of California, San Francisco, San Francisco, CA, USA; Department of Microbiology & Immunology, University of California, San Francisco, San Francisco, CA, USA; Chan Zuckerberg Biohub, San Francisco, CA, USA.
| | - Maya E Kotas
- Department of Medicine, University of California, San Francisco, San Francisco, CA, USA
| | - Claire E O'Leary
- Department of Medicine, University of California, San Francisco, San Francisco, CA, USA
| | - Katelyn Singh
- Department of Dermatology, Yale School of Medicine, New Haven, CT, USA
| | - William Damsky
- Department of Dermatology, Yale School of Medicine, New Haven, CT, USA; Department of Pathology, Yale School of Medicine, New Haven, CT, USA
| | - Chang Liao
- Department of Medicine, University of California, San Francisco, San Francisco, CA, USA
| | - Elizabeth Arouge
- Department of Dermatology, University of California, San Francisco, San Francisco, CA, USA
| | - Iliana Tenvooren
- Department of Otolaryngology and Microbiology & Immunology, University of California, San Francisco, San Francisco, CA, USA
| | - Diana M Marquez
- Department of Otolaryngology and Microbiology & Immunology, University of California, San Francisco, San Francisco, CA, USA
| | - Andrew W Schroeder
- Department of Medicine, University of California, San Francisco, San Francisco, CA, USA
| | - Jarish N Cohen
- Department of Pathology, University of California, San Francisco, San Francisco, CA, USA
| | - Marlys S Fassett
- Department of Dermatology, University of California, San Francisco, San Francisco, CA, USA; Department of Microbiology & Immunology, University of California, San Francisco, San Francisco, CA, USA
| | - Jinwoo Lee
- Department of Medicine, University of California, San Francisco, San Francisco, CA, USA
| | - Scott G Daniel
- Division of Gastroenterology, Hepatology, and Nutrition, The Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Kyle Bittinger
- Division of Gastroenterology, Hepatology, and Nutrition, The Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Roberto Efraín Díaz
- Department of Bioengineering and Therapeutic Sciences, University of California, San Francisco, San Francisco, CA, USA; Tetrad Graduate Program, University of California, San Francisco, San Francisco, CA, USA
| | - James S Fraser
- Department of Bioengineering and Therapeutic Sciences, University of California, San Francisco, San Francisco, CA, USA
| | - Niwa Ali
- Department of Dermatology, University of California, San Francisco, San Francisco, CA, USA
| | - K Mark Ansel
- Department of Microbiology & Immunology, University of California, San Francisco, San Francisco, CA, USA
| | - Matthew H Spitzer
- Department of Otolaryngology and Microbiology & Immunology, University of California, San Francisco, San Francisco, CA, USA; Parker Institute for Cancer Immunotherapy, San Francisco, CA, USA; Chan Zuckerberg Biohub, San Francisco, CA, USA
| | - Hong-Erh Liang
- Department of Medicine, University of California, San Francisco, San Francisco, CA, USA
| | - Richard M Locksley
- Department of Microbiology & Immunology, University of California, San Francisco, San Francisco, CA, USA; Department of Medicine, University of California, San Francisco, San Francisco, CA, USA; Howard Hughes Medical Institute, University of California, San Francisco, San Francisco, CA, USA.
| |
Collapse
|
8
|
Indralingam CS, Gutierrez-Gonzalez AK, Johns SC, Tsui T, Cannon DT, Fuster MM, Bigby TD, Jennings PA, Breen EC. IL-33/ST2 receptor-dependent signaling in the development of pulmonary hypertension in Sugen/hypoxia mice. Physiol Rep 2022; 10:e15185. [PMID: 35150208 PMCID: PMC8839421 DOI: 10.14814/phy2.15185] [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: 10/29/2021] [Revised: 01/10/2022] [Accepted: 01/11/2022] [Indexed: 11/24/2022] Open
Abstract
Pulmonary arterial hypertension (PAH) is associated with significant morbidity and mortality. PAH is characterized by pulmonary artery remodeling, elevated right ventricular pressure (RVP) and, ultimately, cardiac failure. Pulmonary endothelial cells can sense danger or damage caused by mechanical injury or pathogens through alarmin cytokines. These cytokines can signal proliferation to restore barrier integrity or aberrant hyperproliferation and remodeling. We hypothesized that IL‐33 signals pulmonary artery endothelial cells to proliferate under hypertensive conditions during the remodeling response and rise in RVP. To test this hypothesis, pulmonary hypertension (PH) was induced in C57Bl/6J, IL‐33 receptor gene deleted (ST2−/−) and MYD88 gene deleted (MYD88−/−) mice by exposure to 10% O2 and SU5416 injections (SUHX). RVP, arterial wall thickness, endothelial cell proliferation and IL‐33 levels and signaling were evaluated. In response to SUHX. RVP increased in C57Bl/6J mice in response to SUHX (49% male and 70% female; p < 0.0001) and this SUHX response was attenuated in ST2−/− mice (29% male p = 0.003; 30% female p = 0.001) and absent in MYD88−/− mice. Wall thickness was increased in SUHX C57Bl/6J mice (p = 0.005), but not in ST2−/− or MYD88−/− mice. Proliferating cells were detected in C57Bl/6J mice by flow cytometry (CD31+/BrDU+; p = 0.02) and immunofluorescence methods (Ki‐67+). IL‐33 was increased by SUHX (p = 0.03) but a genotype effect was not observed (p = 0.76). We observed that in hPAECs, IL‐33 expression is regulated by both IL‐33 and DLL4. These data suggest IL‐33/ST2 signaling is essential for the endothelial cell proliferative response in PH.
Collapse
Affiliation(s)
| | | | - Scott C Johns
- VA San Diego Healthcare System, La Jolla, California, USA
| | - Tzuhan Tsui
- Medicine, University of California, San Diego, La Jolla, California, USA
| | - Daniel T Cannon
- School of Exercise and Nutritional Sciences, San Diego State University, San Diego, California, USA
| | - Mark M Fuster
- VA San Diego Healthcare System, La Jolla, California, USA.,Division of Pulmonary & Critical Care, University of California, San Diego, La Jolla, California, USA
| | - Timothy D Bigby
- VA San Diego Healthcare System, La Jolla, California, USA.,Division of Pulmonary & Critical Care, University of California, San Diego, La Jolla, California, USA
| | - Patricia A Jennings
- Chemistry and Biochemistry, University of California, San Diego, La Jolla, California, USA
| | - Ellen C Breen
- Medicine, University of California, San Diego, La Jolla, California, USA
| |
Collapse
|
9
|
Westermann S, Dietschmann A, Doehler D, Castiglione K, Bochner BS, Voehringer D, Radtke D. Siglec-F Promotes IL-33-Induced Cytokine Release from Bone Marrow-Derived Eosinophils Independently of the ITIM and ITIM-like Motif Phosphorylation. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2022; 208:732-744. [PMID: 34996839 DOI: 10.4049/jimmunol.2100184] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/24/2021] [Accepted: 11/19/2021] [Indexed: 12/15/2022]
Abstract
Eosinophils are potent innate effector cells associated mainly with type 2 immune responses elicited by helminths and allergens. Their activity needs to be tightly controlled to prevent severe inflammation and tissue damage. Eosinophil degranulation and secretion of inflammatory effector molecules, including cytokines, chemokines, and lipid mediators, can be regulated by activating and inhibitory receptors on the cell surface. In this study, we investigated the modulation of proliferation, apoptosis, gene expression, and cytokine/chemokine secretion from IL-33-activated Mus musculus eosinophils on cross-linking of the transmembrane receptor Sialic acid-binding Ig-like lectin F (Siglec-F). Siglec-F contains an ITIM plus an ITIM-like motif in its intracellular tail and is mainly regarded as an inhibitory and apoptosis-inducing receptor. In vitro costimulation of bone marrow-derived eosinophils with anti-Siglec-F and IL-33 compared with treatment with either alone led to enhanced STAT6 phosphorylation, stronger induction of hypoxia/glycolysis-related proinflammatory genes, and elevated secretion of type 2 cytokines (IL-4, IL-13) and chemokines (CCL3, CCL4) with only minor effects on proliferation and apoptosis. Using a competitive mixed bone marrow chimera approach with wild-type and Siglec-F-deficient eosinophils, we observed no evidence for Siglec-F-regulated inhibition of Aspergillus fumigatus-elicited lung eosinophilia. Truncation of the Siglec-F cytoplasmic tail, but not mutation of the ITIM and ITIM-like motifs, ablated the effect of enhanced cytokine/chemokine secretion. This provides evidence for an ITIM phosphorylation-independent signaling pathway from the cytoplasmic tail of the Siglec-F receptor that enhances effector molecule release from activated eosinophils.
Collapse
Affiliation(s)
- Stefanie Westermann
- Department of Infection Biology, University Hospital Erlangen and Friedrich-Alexander University Erlangen-Nuremberg, Erlangen, Germany; and
| | - Axel Dietschmann
- Department of Infection Biology, University Hospital Erlangen and Friedrich-Alexander University Erlangen-Nuremberg, Erlangen, Germany; and
| | - Daniela Doehler
- Department of Infection Biology, University Hospital Erlangen and Friedrich-Alexander University Erlangen-Nuremberg, Erlangen, Germany; and
| | - Kirstin Castiglione
- Department of Infection Biology, University Hospital Erlangen and Friedrich-Alexander University Erlangen-Nuremberg, Erlangen, Germany; and
| | - Bruce S Bochner
- Division of Allergy and Immunology, Department of Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL
| | - David Voehringer
- Department of Infection Biology, University Hospital Erlangen and Friedrich-Alexander University Erlangen-Nuremberg, Erlangen, Germany; and
| | - Daniel Radtke
- Department of Infection Biology, University Hospital Erlangen and Friedrich-Alexander University Erlangen-Nuremberg, Erlangen, Germany; and
| |
Collapse
|
10
|
IL-33: A central cytokine in helminth infections. Semin Immunol 2021; 53:101532. [PMID: 34823996 DOI: 10.1016/j.smim.2021.101532] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/13/2021] [Revised: 11/10/2021] [Accepted: 11/12/2021] [Indexed: 02/06/2023]
Abstract
IL-33 is an alarmin cytokine which has been implicated in allergy, fibrosis, inflammation, tumorigenesis, metabolism, and homeostasis. However, amongst its strongest roles are in helminth infections, where IL-33 usually (but not always) is central to induction of an effective anti-parasitic immune response. In this review, we will summarise the literature around this fascinating cytokine, its activity on immune and non-immune cells, the unique (and sometimes counterintuitive) responses it induces, and how it can coordinate the immune response during infections by parasitic helminths. Finally, we will summarise some of the ways that parasites have developed to modulate the IL-33 pathway for their own benefit.
Collapse
|
11
|
Cannon DT, Nogueira L, Gutierrez-Gonzalez AK, Gilmore NK, Bigby TD, Breen EC. Role of IL-33 receptor (ST2) deletion in diaphragm contractile and mitochondrial function in the Sugen5416/hypoxia model of pulmonary hypertension. Respir Physiol Neurobiol 2021; 295:103783. [PMID: 34508866 DOI: 10.1016/j.resp.2021.103783] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2021] [Revised: 08/14/2021] [Accepted: 09/06/2021] [Indexed: 12/22/2022]
Abstract
Pulmonary arterial hypertension (PAH) is a progressive disease of the pulmonary vasculature that leads to right ventricular failure. Skeletal muscle maladaptations limit physical activity and may contribute to disease progression. The role of alarmin/inflammatory signaling in PAH respiratory muscle dysfunction is unknown. We hypothesized that diaphragm mitochondrial and contractile functions are impaired in SU5416/hypoxia-induced pulmonary hypertension due to increased systemic IL-33 signaling. We induced pulmonary hypertension in adult C57Bl/6 J (WT) and ST2 (IL1RL1) gene ablated mice by SU5416/hypoxia (SuHx). We measured diaphragm fiber mitochondrial respiration, inflammatory markers, and contractile function ex vivo. SuHx reduced coupled and uncoupled permeabilized myofiber respiration by ∼40 %. During coupled respiration with complex I substrates, ST2-/- attenuated SuHx inhibition of mitochondrial respiration (genotype × treatment interaction F[1,67] = 3.3, p = 0.07, η2 = 0.04). Flux control ratio and coupling efficiency were not affected by SuHx or genotype. A higher substrate control ratio for succinate was observed in SuHx fibers and attenuated in ST2-/- fibers (F[1,67] = 5.3, p < 0.05, η2 = 0.07). Diaphragm TNFα, but not IL-33 or NFkB, was increased in SuHx vs. DMSO in both genotypes (F[1,43] = 4.7, p < 0.05, η2 = 0.1). Diaphragm force-frequency relationships were right-shifted in SuHx vs. WT (F[3,440] = 8.4, p < 0.05, η2 = 0.0025). There was no effect of ST2-/- on the force-frequency relationship. Force decay during a fatigue protocol at 100 Hz, but not at 40 Hz, was attenuated by SuHx vs. DMSO in both genotypes (F[1,41] = 5.6, p < 0.05, η2 = 0.11). SuHx mice exhibit a modest compensation in diaphragm contractility and mitochondrial dysfunction during coupled respiration; the latter partially regulated through ST2 signaling.
Collapse
Affiliation(s)
- Daniel T Cannon
- School of Exercise & Nutritional Sciences, San Diego State University, United States.
| | - Leonardo Nogueira
- Department of Medicine, University of California, San Diego, United States; Instituto de Bioquímica Médica Leopoldo de Meis, Federal University of Rio de Janeiro, Brazil
| | | | - Natalie K Gilmore
- Department of Medicine, University of California, San Diego, United States
| | - Timothy D Bigby
- Department of Medicine, University of California, San Diego, United States
| | - Ellen C Breen
- Department of Medicine, University of California, San Diego, United States
| |
Collapse
|
12
|
Pivniouk V, Gimenes Junior JA, Honeker LK, Vercelli D. The role of innate immunity in asthma development and protection: Lessons from the environment. Clin Exp Allergy 2021; 50:282-290. [PMID: 31581343 DOI: 10.1111/cea.13508] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2019] [Revised: 08/27/2019] [Accepted: 09/21/2019] [Indexed: 12/24/2022]
Abstract
Asthma, a complex, chronic disease characterized by airway inflammation, hyperresponsiveness and remodelling, affects over 300 million people worldwide. While the disease is typically associated with exaggerated allergen-induced type 2 immune responses, these responses are strongly influenced by environmental exposures that stimulate innate immune pathways capable of promoting or protecting from asthma. The dual role played by innate immunity in asthma pathogenesis offers multiple opportunities for both research and clinical interventions and is the subject of this review.
Collapse
Affiliation(s)
- Vadim Pivniouk
- Asthma and Airway Disease Research Center, University of Arizona, Tucson, AZ, USA.,Department of Cellular and Molecular Medicine, University of Arizona, Tucson, AZ, USA
| | | | - Linnea K Honeker
- Asthma and Airway Disease Research Center, University of Arizona, Tucson, AZ, USA
| | - Donata Vercelli
- Asthma and Airway Disease Research Center, University of Arizona, Tucson, AZ, USA.,Department of Cellular and Molecular Medicine, University of Arizona, Tucson, AZ, USA
| |
Collapse
|
13
|
Wu Y, Shi W, Wang H, Yue J, Mao Y, Zhou W, Kong X, Guo Q, Zhang L, Xu P, Wang Y. Anti-ST2 Nanoparticle Alleviates Lung Inflammation by Targeting ILC2s-CD4 +T Response. Int J Nanomedicine 2020; 15:9745-9758. [PMID: 33299314 PMCID: PMC7721292 DOI: 10.2147/ijn.s268282] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2020] [Accepted: 11/04/2020] [Indexed: 11/23/2022] Open
Abstract
Background Asthma has been regarded as an inflammatory disease, and group 2 innate lymphoid cells (ILC2s) are implicated in asthma pathogenesis. However, no strategy is available to block ILC2s function. Efficiency is also limited due to the use of systemic or subcutaneous routes of administration. The purpose of this study was to investigate the effects of nanoparticles targeting suppression of tumorigenicity 2 (ST2), which is the ILC2 receptor, to alleviate lung inflammation in the murine model of asthma. Methods The ultra-small SPIO nanoparticles were firstly synthesized, OVA-induced mice were administered by anti-ST2-conjugated nanoparticles. The inflammatory degree of the lung was investigated by H&E. The percentages of ILC2s and CD4+T cells in bronchoalveolar lavage fluid (BALF) and lung tissue were determined by FACS. Th2-cytokine and OVA-IgE levels were detected by real-time PCR and ELISA, respectively. Results Treatment with anti-ST2-conjugated nanoparticles significantly alleviated airway inflammation, IL-33 and IL-13 levels and the percentage of CD4+T cells. The percentage of ILC2s was increased, whereas the levels of IL-13 and IL-5 expressed by ILC2s were reduced. Conclusion In the present study, we demonstrated that anti-ST2-conjugated nanoparticles can efficiently control lung inflammation in OVA-induced mice by reducing the ability of ILC2s to produce IL-5 and IL-13, thereby reducing CD4+T cells. Our study also demonstrated that the nanoparticle delivery system could improve the performance of anti-ST2, which may be used as a strategic tool to expand the current drug market.
Collapse
Affiliation(s)
- Yumin Wu
- Department of Laboratory Medicine, The Third Affiliated Hospital of Soochow University, Changzhou, Jiangsu 213003, People's Republic of China
| | - Weifeng Shi
- Department of Laboratory Medicine, The Third Affiliated Hospital of Soochow University, Changzhou, Jiangsu 213003, People's Republic of China
| | - Honghai Wang
- Department of Orthopaedics, The Third Affiliated Hospital of Soochow University, Changzhou, Jiangsu 213003, People's Republic of China
| | - Jiawei Yue
- Department of Orthopaedics, The Third Affiliated Hospital of Soochow University, Changzhou, Jiangsu 213003, People's Republic of China
| | - Yijie Mao
- Department of Laboratory Medicine, The Third Affiliated Hospital of Soochow University, Changzhou, Jiangsu 213003, People's Republic of China
| | - Wei Zhou
- Department of Laboratory Medicine, The Third Affiliated Hospital of Soochow University, Changzhou, Jiangsu 213003, People's Republic of China
| | - Xinagmin Kong
- Department of Laboratory Medicine, The Third Affiliated Hospital of Soochow University, Changzhou, Jiangsu 213003, People's Republic of China
| | - Qiqiong Guo
- Department of Laboratory Medicine, The Third Affiliated Hospital of Soochow University, Changzhou, Jiangsu 213003, People's Republic of China
| | - Lirong Zhang
- Department of Laboratory Medicine, The Third Affiliated Hospital of Soochow University, Changzhou, Jiangsu 213003, People's Republic of China
| | - Pengxiao Xu
- Department of Laboratory Medicine, The Third Affiliated Hospital of Soochow University, Changzhou, Jiangsu 213003, People's Republic of China
| | - Yuyue Wang
- Department of Laboratory Medicine, The Third Affiliated Hospital of Soochow University, Changzhou, Jiangsu 213003, People's Republic of China
| |
Collapse
|
14
|
Proenkephalin + regulatory T cells expanded by ultraviolet B exposure maintain skin homeostasis with a healing function. Proc Natl Acad Sci U S A 2020; 117:20696-20705. [PMID: 32769209 DOI: 10.1073/pnas.2000372117] [Citation(s) in RCA: 42] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Regulatory T (Treg) cells, expressing CD25 (interleukin-2 receptor α chain) and Foxp3 transcription factor, maintain immunological self-tolerance and suppress various immune responses. Here we report a feature of skin Treg cells expanded by ultraviolet B (UVB) exposure. We found that skin Treg cells possessing a healing function are expanded by UVB exposure with the expression of an endogenous opioid precursor, proenkephalin (PENK). Upon UVB exposure, skin Treg cells were expanded with a unique TCR repertoire. Also, they highly expressed a distinctive set of genes enriched in "wound healing involved in inflammatory responses" and the "neuropeptide signaling pathway," as indicated by the high expression of Penk. We found that not only was PENK expression at the protein level detected in the UVB-expanded skin Treg (UVB-skin Treg) cells, but that a PENK-derived neuropeptide, methionine enkephalin (Met-ENK), from Treg cells promoted the outgrowth of epidermal keratinocytes in an ex vivo skin explant assay. Notably, UVB-skin Treg cells also promoted wound healing in an in vivo wound closure assay. In addition, UVB-skin Treg cells produced amphiregulin (AREG), which plays a key role in Treg-mediated tissue repair. Identification of a unique function of PENK+ UVB-skin Treg cells provides a mechanism for maintaining skin homeostasis.
Collapse
|
15
|
Zhang S, Wu S, Shen Y, Xiao Y, Gao L, Shi S. Cytotoxicity studies of Fe 3O 4 nanoparticles in chicken macrophage cells. ROYAL SOCIETY OPEN SCIENCE 2020; 7:191561. [PMID: 32431865 PMCID: PMC7211854 DOI: 10.1098/rsos.191561] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/28/2019] [Accepted: 03/06/2020] [Indexed: 05/06/2023]
Abstract
Magnetic Fe3O4 nanoparticles (Fe3O4-NPs) have been widely investigated for their biomedical applications. The main purpose of this study was to evaluate the cytotoxic effects of different sizes of Fe3O4-NPs in chicken macrophage cells (HD11). Experimental groups based on three sizes of Fe3O4-NPs (60, 120 and 250 nm) were created, and the Fe3O4-NPs were added to the cells at different doses according to the experimental group. The cell activity, oxidative index (malondialdehyde (MDA), superoxide dismutase (SOD) and reactive oxygen species (ROS)), apoptosis and pro-inflammatory cytokine secretion level were detected to analyse the cytotoxic effects of Fe3O4-NPs of different sizes in HD11 cells. The results revealed that the cell viability of the 60 nm Fe3O4-NPs group was lower than those of the 120 and 250 nm groups when the same concentration of Fe3O4-NPs was added. No significant difference in MDA was observed among the three Fe3O4-NP groups. The SOD level and ROS production of the 60 nm group were significantly greater than those of the 120 and 250 nm groups. Furthermore, the highest levels of apoptosis and pro-inflammatory cytokine secretion were caused by the 60 nm Fe3O4-NPs. In conclusion, the smaller Fe3O4-NPs produced stronger cytotoxicity in chicken macrophage cells, and the cytotoxic effects may be related to the oxidative stress and apoptosis induced by increased ROS production as well as the increased expression of pro-inflammatory cytokines.
Collapse
Affiliation(s)
- Shan Zhang
- Poultry Institute, Chinese Academy of Agricultural Sciences, Yangzhou, Jiangsu 225125, People's Republic of China
| | - Shu Wu
- Poultry Institute, Chinese Academy of Agricultural Sciences, Yangzhou, Jiangsu 225125, People's Republic of China
| | - Yiru Shen
- Poultry Institute, Chinese Academy of Agricultural Sciences, Yangzhou, Jiangsu 225125, People's Republic of China
| | - Yunqi Xiao
- Poultry Institute, Chinese Academy of Agricultural Sciences, Yangzhou, Jiangsu 225125, People's Republic of China
| | - Lizeng Gao
- Institute of Biophysics, Chinese Academy of Science, CAS Engineering Laboratory for Nanozyme, Institute of Biophysics, CAS, Beijing 100101, China
| | - Shourong Shi
- Poultry Institute, Chinese Academy of Agricultural Sciences, Yangzhou, Jiangsu 225125, People's Republic of China
- Jiangsu Co-innovation Centre for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou, Jiangsu 225000, People's Republic of China
- Author for correspondence: Shourong Shi e-mail:
| |
Collapse
|
16
|
Ozasa K, Temizoz B, Kusakabe T, Kobari S, Momota M, Coban C, Ito S, Kobiyama K, Kuroda E, Ishii KJ. Cyclic GMP-AMP Triggers Asthma in an IL-33-Dependent Manner That Is Blocked by Amlexanox, a TBK1 Inhibitor. Front Immunol 2019; 10:2212. [PMID: 31616416 PMCID: PMC6775192 DOI: 10.3389/fimmu.2019.02212] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2019] [Accepted: 09/02/2019] [Indexed: 01/01/2023] Open
Abstract
Extracellular host-derived DNA, as one of damage associated molecular patterns (DAMPs), is associated with allergic type 2 immune responses. Immune recognition of such DNA generates the second messenger cyclic GMP-AMP (cGAMP) and induces type-2 immune responses; however, its role in allergic diseases, such as asthma, has not been fully elucidated. This study aimed to determine whether cGAMP could induce asthma when used as an adjuvant. We intranasally sensitized mice with cGAMP together with house dust mite antigen (HDM), followed by airway challenge with HDM. We then assessed the levels of eosinophils in the broncho-alveolar lavage fluid (BALF) and serum HDM-specific antibodies. cGAMP promoted HDM specific allergic asthma, characterized by significantly increased HDM specific IgG1 and total IgE in the serum and infiltration of eosinophils in the BALF. cGAMP stimulated lung fibroblast cells to produce IL-33 in vitro, and mice deficient for IL-33 or IL-33 receptor (ST2) failed to develop asthma enhancement by cGAMP. Not only Il-33 -/- mice, but also Sting -/-, Tbk1 -/-, and Irf3 -/- Irf7 -/- mice which lack the cGAMP-mediated innate immune activation failed to increase eosinophils in the BALF than that from wild type mice. Consistently, intranasal and oral administration of amlexanox, a TBK1 inhibitor, decreased cGAMP-induced lung allergic inflammation. Thus, cGAMP functions as a type 2 adjuvant in the lung and can promote allergic asthma in manners that dependent on the intracellular STING/TBK1/IRF3/7 signaling pathway and the resultant intercellular signaling pathway via IL-33 and ST2 might be a novel therapeutic target for allergic asthma.
Collapse
Affiliation(s)
- Koji Ozasa
- Laboratory of Adjuvant Innovation, Center for Vaccine and Adjuvant Research, National Institutes of Biomedical Innovation, Health and Nutrition, Osaka, Japan.,Department of Pediatrics, Yokohama City University Graduate School of Medicine, Yokohama, Japan
| | - Burcu Temizoz
- Laboratory of Adjuvant Innovation, Center for Vaccine and Adjuvant Research, National Institutes of Biomedical Innovation, Health and Nutrition, Osaka, Japan.,Laboratory of Vaccine Science, WPI Immunology Frontier Research Center, Osaka University, Osaka, Japan.,Division of Vaccine Science, Department of Microbiology and Immunology, The Institute of Medical Science, The University of Tokyo, Tokyo, Japan.,International Research and Development Center for Mucosal Vaccines, The Institute of Medical Science, The University of Tokyo, Tokyo, Japan
| | - Takato Kusakabe
- Laboratory of Adjuvant Innovation, Center for Vaccine and Adjuvant Research, National Institutes of Biomedical Innovation, Health and Nutrition, Osaka, Japan.,Laboratory of Vaccine Science, WPI Immunology Frontier Research Center, Osaka University, Osaka, Japan.,Laboratory of Mock-Up Vaccine Project, Center for Vaccine and Adjuvant Research, National Institutes of Biomedical Innovation, Health and Nutrition, Osaka, Japan
| | - Shingo Kobari
- Laboratory of Adjuvant Innovation, Center for Vaccine and Adjuvant Research, National Institutes of Biomedical Innovation, Health and Nutrition, Osaka, Japan.,Department of Pediatrics, Yokohama City University Graduate School of Medicine, Yokohama, Japan
| | - Masatoshi Momota
- Laboratory of Adjuvant Innovation, Center for Vaccine and Adjuvant Research, National Institutes of Biomedical Innovation, Health and Nutrition, Osaka, Japan.,Laboratory of Mock-Up Vaccine Project, Center for Vaccine and Adjuvant Research, National Institutes of Biomedical Innovation, Health and Nutrition, Osaka, Japan
| | - Cevayir Coban
- International Research and Development Center for Mucosal Vaccines, The Institute of Medical Science, The University of Tokyo, Tokyo, Japan.,Laboratory of Malaria Immunology, WPI Immunology Frontier Research Center, Osaka University, Osaka, Japan.,Division of Malaria Immunology, Department of Microbiology and Immunology, The Institute of Medical Science, The University of Tokyo, Tokyo, Japan
| | - Shuichi Ito
- Department of Pediatrics, Yokohama City University Graduate School of Medicine, Yokohama, Japan
| | - Kouji Kobiyama
- Laboratory of Adjuvant Innovation, Center for Vaccine and Adjuvant Research, National Institutes of Biomedical Innovation, Health and Nutrition, Osaka, Japan.,Laboratory of Vaccine Science, WPI Immunology Frontier Research Center, Osaka University, Osaka, Japan.,Division of Vaccine Science, Department of Microbiology and Immunology, The Institute of Medical Science, The University of Tokyo, Tokyo, Japan.,International Research and Development Center for Mucosal Vaccines, The Institute of Medical Science, The University of Tokyo, Tokyo, Japan
| | - Etsushi Kuroda
- Laboratory of Adjuvant Innovation, Center for Vaccine and Adjuvant Research, National Institutes of Biomedical Innovation, Health and Nutrition, Osaka, Japan.,Laboratory of Vaccine Science, WPI Immunology Frontier Research Center, Osaka University, Osaka, Japan.,Department of Immunology, Hyogo College of Medicine, Nishinomiya, Japan
| | - Ken J Ishii
- Laboratory of Adjuvant Innovation, Center for Vaccine and Adjuvant Research, National Institutes of Biomedical Innovation, Health and Nutrition, Osaka, Japan.,Laboratory of Vaccine Science, WPI Immunology Frontier Research Center, Osaka University, Osaka, Japan.,Division of Vaccine Science, Department of Microbiology and Immunology, The Institute of Medical Science, The University of Tokyo, Tokyo, Japan.,International Research and Development Center for Mucosal Vaccines, The Institute of Medical Science, The University of Tokyo, Tokyo, Japan.,Laboratory of Mock-Up Vaccine Project, Center for Vaccine and Adjuvant Research, National Institutes of Biomedical Innovation, Health and Nutrition, Osaka, Japan
| |
Collapse
|
17
|
Takemura N, Kurashima Y, Mori Y, Okada K, Ogino T, Osawa H, Matsuno H, Aayam L, Kaneto S, Park EJ, Sato S, Matsunaga K, Tamura Y, Ouchi Y, Kumagai Y, Kobayashi D, Suzuki Y, Yoshioka Y, Nishimura J, Mori M, Ishii KJ, Rothenberg ME, Kiyono H, Akira S, Uematsu S. Eosinophil depletion suppresses radiation-induced small intestinal fibrosis. Sci Transl Med 2019; 10:10/429/eaan0333. [PMID: 29467297 DOI: 10.1126/scitranslmed.aan0333] [Citation(s) in RCA: 55] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2017] [Revised: 10/05/2017] [Accepted: 01/17/2018] [Indexed: 12/14/2022]
Abstract
Radiation-induced intestinal fibrosis (RIF) is a serious complication after abdominal radiotherapy for pelvic tumor or peritoneal metastasis. Herein, we show that RIF is mediated by eosinophil interactions with α-smooth muscle actin-positive (α-SMA+) stromal cells. Abdominal irradiation caused RIF especially in the submucosa (SM) of the small intestine, which was associated with the excessive accumulation of eosinophils in both human and mouse. Eosinophil-deficient mice showed markedly ameliorated RIF, suggesting the importance of eosinophils. After abdominal irradiation, chronic crypt cell death caused elevation of extracellular adenosine triphosphate, which in turn activated expression of C-C motif chemokine 11 (CCL11) by pericryptal α-SMA+ cells in the SM to attract eosinophils in mice. Inhibition of C-C chemokine receptor 3 (CCR3) by genetic deficiency or neutralizing antibody (Ab) treatment suppressed eosinophil accumulation in the SM after irradiation in mice, suggesting a critical role of the CCL11/CCR3 axis in the eosinophil recruitment. Activated α-SMA+ cells also expressed granulocyte-macrophage colony-stimulating factor (GM-CSF) to activate eosinophils. Transforming growth factor-β1 from GM-CSF-stimulated eosinophils promoted collagen expression by α-SMA+ cells. In translational studies, treatment with a newly developed interleukin-5 receptor α-targeting Ab, analogous to the human agent benralizumab, depleted intestinal eosinophils and suppressed RIF in mice. Collectively, we identified eosinophils as a crucial factor in the pathogenesis of RIF and showed potential therapeutic strategies for RIF by targeting eosinophils.
Collapse
Affiliation(s)
- Naoki Takemura
- Department of Mucosal Immunology, Graduate School of Medicine, Chiba University, 1-8-1 Inohana, Chuo-ku, Chiba 260-8670, Japan.,Division of Innate Immune Regulation, International Research and Development Center for Mucosal Vaccines, Institute of Medical Science, The University of Tokyo, 4-6-1 Shirokanedai, Minato-ku, Tokyo 108-8639, Japan
| | - Yosuke Kurashima
- Department of Mucosal Immunology, Graduate School of Medicine, Chiba University, 1-8-1 Inohana, Chuo-ku, Chiba 260-8670, Japan.,Division of Mucosal Immunology, Department of Microbiology and Immunology, Institute of Medical Science, The University of Tokyo, Tokyo 108-8639, Japan.,Institute for Global Prominent Research, Chiba University, Chiba 260-8670, Japan.,Department of Innovative Medicine, Graduate School of Medicine, Chiba University, Chiba 260-8670, Japan.,Division of Clinical Vaccinology, International Research and Development Center for Mucosal Vaccines, The Institute of Medical Science, The University of Tokyo, Tokyo 108-8639, Japan
| | - Yuki Mori
- Laboratory of Biofunctional Imaging, World Premier Institute (WPI) Immunology Frontier Research Center, Osaka University, 3-1 Yamada-oka, Suita, Osaka 565-0871, Japan
| | - Kazuki Okada
- Immunology and Allergy R&D Unit, R&D Division, Kyowa Hakko Kirin Co. Ltd., 3-6-6 Asahi-machi, Machida-shi, Tokyo 194-8533, Japan
| | - Takayuki Ogino
- Department of Gastroenterological Surgery, Graduate School of Medicine, Osaka University, 2-2 Yamada-oka, Suita, Osaka 565-0871, Japan.,Institute of Medical Microbiology and Hygiene, University of Mainz Medical Centre, Obere Zahlbacher Strasse 67, Mainz 55131, Germany
| | - Hideki Osawa
- Department of Gastroenterological Surgery, Graduate School of Medicine, Osaka University, 2-2 Yamada-oka, Suita, Osaka 565-0871, Japan
| | - Hirosih Matsuno
- Department of Gastroenterological Surgery, Graduate School of Medicine, Osaka University, 2-2 Yamada-oka, Suita, Osaka 565-0871, Japan
| | - Lamichhane Aayam
- Division of Mucosal Immunology, Department of Microbiology and Immunology, Institute of Medical Science, The University of Tokyo, Tokyo 108-8639, Japan
| | - Satoshi Kaneto
- Division of Mucosal Immunology, Department of Microbiology and Immunology, Institute of Medical Science, The University of Tokyo, Tokyo 108-8639, Japan
| | - Eun Jeong Park
- Division of Mucosal Immunology, Department of Microbiology and Immunology, Institute of Medical Science, The University of Tokyo, Tokyo 108-8639, Japan.,Department of Molecular Pathobiology and Cell Adhesion Biology, Basic Medical Sciences, Mie University Graduate School of Medicine, 2-174 Edobashi, Tsu, Mie 514-8507, Japan
| | - Shintaro Sato
- Division of Mucosal Immunology, Department of Microbiology and Immunology, Institute of Medical Science, The University of Tokyo, Tokyo 108-8639, Japan.,Mucosal Vaccine Project, BIKEN Innovative Vaccine Research Alliance Laboratories, Research Institute for Microbial Diseases, Osaka University, Osaka 565-0871, Japan
| | - Kouta Matsunaga
- Division of Innate Immune Regulation, International Research and Development Center for Mucosal Vaccines, Institute of Medical Science, The University of Tokyo, 4-6-1 Shirokanedai, Minato-ku, Tokyo 108-8639, Japan
| | - Yusuke Tamura
- Division of Innate Immune Regulation, International Research and Development Center for Mucosal Vaccines, Institute of Medical Science, The University of Tokyo, 4-6-1 Shirokanedai, Minato-ku, Tokyo 108-8639, Japan
| | - Yasuo Ouchi
- Department of Mucosal Immunology, Graduate School of Medicine, Chiba University, 1-8-1 Inohana, Chuo-ku, Chiba 260-8670, Japan
| | - Yutaro Kumagai
- Quantitative Immunology Research Unit, WPI Immunology Frontier Research Center, Osaka University, Osaka 565-0871, Japan
| | - Daichi Kobayashi
- Laboratory of Immune Regulation, Department of Microbiology and Immunology, Graduate School of Medicine, Osaka University, Osaka 565-0871, Japan.,Laboratory of Mucosal Immunology, WPI Immunology Frontier Research Center, Osaka University, Osaka 565-0871, Japan
| | - Yutaka Suzuki
- Department of Medical Genome Science, Graduate School of Frontier Sciences, The University of Tokyo, 5-1-5 Kashiwanoha, Kashiwa, Chiba 277-8562, Japan
| | - Yoshichika Yoshioka
- Laboratory of Biofunctional Imaging, World Premier Institute (WPI) Immunology Frontier Research Center, Osaka University, 3-1 Yamada-oka, Suita, Osaka 565-0871, Japan
| | - Junichi Nishimura
- Department of Gastroenterological Surgery, Graduate School of Medicine, Osaka University, 2-2 Yamada-oka, Suita, Osaka 565-0871, Japan
| | - Masaki Mori
- Department of Gastroenterological Surgery, Graduate School of Medicine, Osaka University, 2-2 Yamada-oka, Suita, Osaka 565-0871, Japan
| | - Ken J Ishii
- Laboratory of Adjuvant Innovation, National Institute of Biomedical Innovation, 7-6-8 Asagi Saito, Ibaraki, Osaka 567-0085, Japan.,Laboratory of Vaccine Science, WPI Immunology Frontier Research Center, Osaka University, Osaka 565-0871, Japan
| | - Mark E Rothenberg
- Division of Allergy and Immunology, Department of Pediatrics, Cincinnati Children's Hospital Medical Center, University of Cincinnati College of Medicine, Cincinnati, OH 45229, USA
| | - Hiroshi Kiyono
- Division of Mucosal Immunology, Department of Microbiology and Immunology, Institute of Medical Science, The University of Tokyo, Tokyo 108-8639, Japan.,Division of Clinical Vaccinology, International Research and Development Center for Mucosal Vaccines, The Institute of Medical Science, The University of Tokyo, Tokyo 108-8639, Japan.,Department of Immunology, Graduate School of Medicine, Chiba University, 1-8-1 Inohana, Chuo-ku, Chiba 260-8670, Japan
| | - Shizuo Akira
- Laboratory of Host Defense, WPI Immunology Frontier Research Center, Osaka University, Osaka 565-0871, Japan.,Department of Host Defense, Research Institute for Microbial Diseases, Osaka University, Osaka 565-0871, Japan
| | - Satoshi Uematsu
- Department of Mucosal Immunology, Graduate School of Medicine, Chiba University, 1-8-1 Inohana, Chuo-ku, Chiba 260-8670, Japan. .,Division of Innate Immune Regulation, International Research and Development Center for Mucosal Vaccines, Institute of Medical Science, The University of Tokyo, 4-6-1 Shirokanedai, Minato-ku, Tokyo 108-8639, Japan
| |
Collapse
|
18
|
IL-33-mediated mast cell activation promotes gastric cancer through macrophage mobilization. Nat Commun 2019; 10:2735. [PMID: 31227713 PMCID: PMC6588585 DOI: 10.1038/s41467-019-10676-1] [Citation(s) in RCA: 155] [Impact Index Per Article: 25.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2018] [Accepted: 05/22/2019] [Indexed: 12/31/2022] Open
Abstract
The contribution of mast cells in the microenvironment of solid malignancies remains controversial. Here we functionally assess the impact of tumor-adjacent, submucosal mast cell accumulation in murine and human intestinal-type gastric cancer. We find that genetic ablation or therapeutic inactivation of mast cells suppresses accumulation of tumor-associated macrophages, reduces tumor cell proliferation and angiogenesis, and diminishes tumor burden. Mast cells are activated by interleukin (IL)-33, an alarmin produced by the tumor epithelium in response to the inflammatory cytokine IL-11, which is required for the growth of gastric cancers in mice. Accordingly, ablation of the cognate IL-33 receptor St2 limits tumor growth, and reduces mast cell-dependent production and release of the macrophage-attracting factors Csf2, Ccl3, and Il6. Conversely, genetic or therapeutic macrophage depletion reduces tumor burden without affecting mast cell abundance. Therefore, tumor-derived IL-33 sustains a mast cell and macrophage-dependent signaling cascade that is amenable for the treatment of gastric cancer. Mast cells within the tumor microenvironment have controversial roles. Here, the authors show, using genetic mouse models, that in gastric cancer, mast cells at the periphery of the tumors are activated via cancer cell produced-IL33 and promote tumorigenesis by recruiting macrophages within the tumors.
Collapse
|
19
|
Guram K, Kim SS, Wu V, Sanders PD, Patel S, Schoenberger SP, Cohen EEW, Chen SY, Sharabi AB. A Threshold Model for T-Cell Activation in the Era of Checkpoint Blockade Immunotherapy. Front Immunol 2019; 10:491. [PMID: 30936880 PMCID: PMC6431643 DOI: 10.3389/fimmu.2019.00491] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2018] [Accepted: 02/22/2019] [Indexed: 12/13/2022] Open
Abstract
Continued discoveries of negative regulators of inflammatory signaling provide detailed molecular insights into peripheral tolerance and anti-tumor immunity. Accumulating evidence indicates that peripheral tolerance is maintained at multiple levels of immune responses by negative regulators of proinflammatory signaling, soluble anti-inflammatory factors, inhibitory surface receptors & ligands, and regulatory cell subsets. This review provides a global overview of these regulatory machineries that work in concert to maintain peripheral tolerance at cellular and host levels, focusing on the direct and indirect regulation of T cells. The recent success of checkpoint blockade immunotherapy (CBI) has initiated a dramatic shift in the paradigm of cancer treatment. Unprecedented responses to CBI have highlighted the central role of T cells in both anti-tumor immunity and peripheral tolerance and underscored the importance of T cell exhaustion in cancer. We discuss the therapeutic implications of modulating the negative regulators of T cell function for tumor immunotherapy with an emphasis on inhibitory surface receptors & ligands—central players in T cell exhaustion and targets of checkpoint blockade immunotherapies. We then introduce a Threshold Model for Immune Activation—the concept that these regulatory mechanisms contribute to defining a set threshold of immunogenic (proinflammatory) signaling required to elicit an anti-tumor or autoimmune response. We demonstrate the value of the Threshold Model in understanding clinical responses and immune related adverse events in the context of peripheral tolerance, tumor immunity, and the era of Checkpoint Blockade Immunotherapy.
Collapse
Affiliation(s)
- Kripa Guram
- Department of Radiation Medicine and Applied Sciences, San Diego Moores Cancer Center, University of California, San Diego, San Diego, CA, United States
| | - Sangwoo S Kim
- Department of Radiation Medicine and Applied Sciences, San Diego Moores Cancer Center, University of California, San Diego, San Diego, CA, United States
| | - Victoria Wu
- Moores Comprehensive Cancer Center, University of California, San Diego, San Diego, CA, United States
| | - P Dominick Sanders
- Department of Radiation Medicine and Applied Sciences, San Diego Moores Cancer Center, University of California, San Diego, San Diego, CA, United States
| | - Sandip Patel
- Division of Hematology and Oncology, Center for Personalized Cancer Therapy, San Diego Moores Cancer Center, University of California, San Diego, San Diego, CA, United States
| | - Stephen P Schoenberger
- Division of Hematology and Oncology, Center for Personalized Cancer Therapy, San Diego Moores Cancer Center, University of California, San Diego, San Diego, CA, United States.,Laboratory of Cellular Immunology, La Jolla Institute for Allergy and Immunology, La Jolla, CA, United States
| | - Ezra E W Cohen
- Moores Comprehensive Cancer Center, University of California, San Diego, San Diego, CA, United States
| | - Si-Yi Chen
- Department of Molecular Microbiology and Immunology, Norris Comprehensive Cancer Center, University of Southern California, Los Angeles, CA, United States
| | - Andrew B Sharabi
- Department of Radiation Medicine and Applied Sciences, San Diego Moores Cancer Center, University of California, San Diego, San Diego, CA, United States.,Moores Comprehensive Cancer Center, University of California, San Diego, San Diego, CA, United States
| |
Collapse
|
20
|
Vasanthakumar A, Kallies A. Interleukin (IL)-33 and the IL-1 Family of Cytokines-Regulators of Inflammation and Tissue Homeostasis. Cold Spring Harb Perspect Biol 2019; 11:cshperspect.a028506. [PMID: 29101106 DOI: 10.1101/cshperspect.a028506] [Citation(s) in RCA: 31] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
Cytokines play an integral role in shaping innate and adaptive immune responses. Members of the interleukin (IL)-1 family regulate a plethora of immune-cell-mediated processes, which include pathogen defense and tissue homeostasis. Notably, the IL-1 family cytokine IL-33 promotes adaptive and innate type 2 immune responses, confers viral protection and facilitates glucose metabolism and tissue repair. At the cellular level, IL-33 stimulates differentiation, maintenance, and function of various immune cell types, including regulatory T cells, effector CD4+ and CD8+ T cells, macrophages, and type 2 innate lymphoid cells (ILC2s). Other IL-1 family members, such as IL-1β and IL-18 promote type 1 responses, while IL-37 limits immune activation. Although IL-1 cytokines play critical roles in immunity and tissue repair, their deregulated expression is often linked to autoimmune and inflammatory diseases. Therefore, IL-1 cytokines are regulated tightly by posttranscriptional mechanisms and decoy receptors. In this review, we discuss the biology and function of IL-1 family cytokines, with a specific focus on regulation and function of IL-33 in immune and tissue homeostasis.
Collapse
Affiliation(s)
- Ajithkumar Vasanthakumar
- Department of Medical Biology, University of Melbourne, Melbourne, Victoria 3052, Australia.,The Walter and Eliza Hall Institute of Medical Research, Melbourne, Victoria 3052, Australia
| | - Axel Kallies
- Department of Medical Biology, University of Melbourne, Melbourne, Victoria 3052, Australia.,The Walter and Eliza Hall Institute of Medical Research, Melbourne, Victoria 3052, Australia.,The Peter Doherty Institute for Infection and Immunity, University of Melbourne, Melbourne, Victoria 3000, Australia
| |
Collapse
|
21
|
Brain regulatory T cells suppress astrogliosis and potentiate neurological recovery. Nature 2019; 565:246-250. [PMID: 30602786 DOI: 10.1038/s41586-018-0824-5] [Citation(s) in RCA: 493] [Impact Index Per Article: 82.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2018] [Accepted: 11/05/2018] [Indexed: 11/08/2022]
Abstract
In addition to maintaining immune tolerance, FOXP3+ regulatory T (Treg) cells perform specialized functions in tissue homeostasis and remodelling1,2. However, the characteristics and functions of brain Treg cells are not well understood because there is a low number of Treg cells in the brain under normal conditions. Here we show that there is massive accumulation of Treg cells in the mouse brain after ischaemic stroke, and this potentiates neurological recovery during the chronic phase of ischaemic brain injury. Although brain Treg cells are similar to Treg cells in other tissues such as visceral adipose tissue and muscle3-5, they are apparently distinct and express unique genes related to the nervous system including Htr7, which encodes the serotonin receptor 5-HT7. The amplification of brain Treg cells is dependent on interleukin (IL)-2, IL-33, serotonin and T cell receptor recognition, and infiltration into the brain is driven by the chemokines CCL1 and CCL20. Brain Treg cells suppress neurotoxic astrogliosis by producing amphiregulin, a low-affinity epidermal growth factor receptor (EGFR) ligand. Stroke is a leading cause of neurological disability, and there are currently few effective recovery methods other than rehabilitation during the chronic phase. Our findings suggest that Treg cells and their products may provide therapeutic opportunities for neuronal protection against stroke and neuroinflammatory diseases.
Collapse
|
22
|
Hong J, Kim S, Lin PC. Interleukin-33 and ST2 Signaling in Tumor Microenvironment. J Interferon Cytokine Res 2018; 39:61-71. [PMID: 30256696 DOI: 10.1089/jir.2018.0044] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Interleukin-33 (IL-33) is one of the members of the IL-1 family of cytokines and a ligand of ST2 and IL-1 receptor accessory protein (IL-1RAcP) that is known to affect Th2 inflammatory response with partial effects on Th1 responses. This cytokine is released by epithelial and smooth muscle cells of the airway system during their injury by several environmental stimuli, such as allergens, viruses, helminths, and pollutants. IL-33 is an alarmin that acts as an endogenous danger signal, and it has been known to affect various types of cells, such as mast cells, basophils, eosinophils, T cells, and specific subsets of innate lymphoid cells (ILCs). In recent findings, this cytokine is believed to have a critical role in several types of cancers, such as lung cancer, liver cancer, and head and neck squamous cell cancer. The expression of IL-33/ST2 in cancer tissues shows a close association with tumor growth and tumor progression in several types of cancer, suggesting the IL-33/ST2 pathway as a potential target for therapy.
Collapse
Affiliation(s)
- Jaewoo Hong
- 1 Cancer and Inflammation Program, Center for Cancer Research, National Cancer Institute, Frederick, Maryland
| | - Soohyun Kim
- 2 Laboratory of Cytokine Immunology, Department of Biomedical Science and Technology, Konkuk University, Seoul, South Korea
| | - P Charles Lin
- 1 Cancer and Inflammation Program, Center for Cancer Research, National Cancer Institute, Frederick, Maryland
| |
Collapse
|
23
|
De Grove KC, Provoost S, Braun H, Blomme EE, Teufelberger AR, Krysko O, Beyaert R, Brusselle GG, Joos GF, Maes T. IL-33 signalling contributes to pollutant-induced allergic airway inflammation. Clin Exp Allergy 2018; 48:1665-1675. [PMID: 30159930 DOI: 10.1111/cea.13261] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2018] [Revised: 07/27/2018] [Accepted: 08/20/2018] [Indexed: 12/11/2022]
Abstract
BACKGROUND Clinical and experimental studies have identified a crucial role for IL-33 and its receptor ST2 in allergic asthma. Inhalation of traffic-related pollutants, such as diesel exhaust particles (DEP), facilitates the development of asthma and can cause exacerbations of asthma. However, it is unknown whether IL-33/ST2 signalling contributes to the enhancing effects of air pollutants on allergic airway responses. OBJECTIVE We aim to investigate the functional role of IL-33/ST2 signalling in DEP-enhanced allergic airway responses, using an established murine model. METHODS C57BL/6J mice were exposed to saline, DEP alone, house dust mite (HDM) alone or combined DEP+HDM. To inhibit IL-33 signalling, recombinant soluble ST2 (r-sST2) was given prophylactically (ie, during the whole experimental protocol) or therapeutically (ie, at the end of the experimental protocol). Airway hyperresponsiveness and the airway inflammatory responses were assessed in bronchoalveolar lavage fluid (BALF) and lung. RESULTS Combined exposure to DEP+HDM increased IL-33 and ST2 expression in lung, elevated inflammatory responses and bronchial hyperresponsiveness compared to saline, sole DEP or sole HDM exposure. Prophylactic interference with the IL-33/ST2 signalling pathway impaired the DEP-enhanced allergic airway inflammation in the BALF, whereas effects on lung inflammation and airway hyperresponsiveness were minimal. Treatment with r-sST2 at the end of the experimental protocol did not modulate the DEP-enhanced allergic airway responses. CONCLUSION Our data suggest that the IL-33/ST2 pathway contributes to the onset of DEP-enhanced allergic airway inflammation.
Collapse
Affiliation(s)
- Katrien C De Grove
- Department of Respiratory Medicine, Laboratory for Translational Research in Obstructive Pulmonary Diseases, Ghent University Hospital, Ghent, Belgium
| | - Sharen Provoost
- Department of Respiratory Medicine, Laboratory for Translational Research in Obstructive Pulmonary Diseases, Ghent University Hospital, Ghent, Belgium
| | - Harald Braun
- Center for Inflammation Research, Unit of Molecular Signal Transduction in Inflammation, VIB, Ghent, Belgium.,Department of Biomedical Molecular Biology, Ghent University, Ghent, Belgium
| | - Evy E Blomme
- Department of Respiratory Medicine, Laboratory for Translational Research in Obstructive Pulmonary Diseases, Ghent University Hospital, Ghent, Belgium
| | - Andrea R Teufelberger
- Upper Airway Research Laboratory, Department of Otorhinolaryngology, Ghent University, Ghent, Belgium
| | - Olga Krysko
- Upper Airway Research Laboratory, Department of Otorhinolaryngology, Ghent University, Ghent, Belgium
| | - Rudi Beyaert
- Center for Inflammation Research, Unit of Molecular Signal Transduction in Inflammation, VIB, Ghent, Belgium.,Department of Biomedical Molecular Biology, Ghent University, Ghent, Belgium
| | - Guy G Brusselle
- Department of Respiratory Medicine, Laboratory for Translational Research in Obstructive Pulmonary Diseases, Ghent University Hospital, Ghent, Belgium
| | - Guy F Joos
- Department of Respiratory Medicine, Laboratory for Translational Research in Obstructive Pulmonary Diseases, Ghent University Hospital, Ghent, Belgium
| | - Tania Maes
- Department of Respiratory Medicine, Laboratory for Translational Research in Obstructive Pulmonary Diseases, Ghent University Hospital, Ghent, Belgium
| |
Collapse
|
24
|
Reeder KM, Dunaway CW, Blackburn JP, Yu Z, Matalon S, Hastie AT, Ampleford EJ, Meyers DA, Steele C. The common γ-chain cytokine IL-7 promotes immunopathogenesis during fungal asthma. Mucosal Immunol 2018; 11:1352-1362. [PMID: 29907867 PMCID: PMC6319622 DOI: 10.1038/s41385-018-0028-1] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2017] [Revised: 01/06/2018] [Accepted: 02/03/2018] [Indexed: 02/04/2023]
Abstract
Asthmatics sensitized to fungi are reported to have more severe asthma, yet the immunopathogenic pathways contributing to this severity have not been identified. In a pilot assessment of human asthmatics, those subjects sensitized to fungi demonstrated elevated levels of the common γ-chain cytokine IL-7 in lung lavage fluid, which negatively correlated with the lung function measurement PC20. Subsequently, we show that IL-7 administration during experimental fungal asthma worsened lung function and increased the levels of type 2 cytokines (IL-4, IL-5, IL-13), proallergic chemokines (CCL17, CCL22) and proinflammatory cytokines (IL-1α, IL-1β). Intriguingly, IL-7 administration also increased IL-22, which we have previously reported to drive immunopathogenic responses in experimental fungal asthma. Employing IL22CreR26ReYFP reporter mice, we identified γδ T cells, iNKT cells, CD4 T cells and ILC3s as sources of IL-22 during fungal asthma; however, only iNKT cells were significantly increased after IL-7 administration. IL-7-induced immunopathogenesis required both type 2 and IL-22 responses. Blockade of IL-7Rα in vivo resulted in attenuated IL-22 production, lower CCL22 levels, decreased iNKT cell, CD4 T-cell and eosinophil recruitment, yet paradoxically increased dynamic lung resistance. Collectively, these results suggest a complex role for IL-7 signaling in allergic fungal asthma.
Collapse
Affiliation(s)
- Kristen M Reeder
- Department of Medicine, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Chad W Dunaway
- Department of Medicine, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Jonathan P Blackburn
- Department of Medicine, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Zhihong Yu
- Department of Anesthesiology, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Sadis Matalon
- Department of Anesthesiology, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Annette T Hastie
- Department of Medicine, Wake Forest University, Winston-Salem, NC, USA
| | | | | | - Chad Steele
- Department of Medicine, University of Alabama at Birmingham, Birmingham, AL, USA.
| |
Collapse
|
25
|
Arae K, Morita H, Unno H, Motomura K, Toyama S, Okada N, Ohno T, Tamari M, Orimo K, Mishima Y, Suto H, Okumura K, Sudo K, Miyazawa H, Taguchi H, Saito H, Matsumoto K, Nakae S. Chitin promotes antigen-specific Th2 cell-mediated murine asthma through induction of IL-33-mediated IL-1β production by DCs. Sci Rep 2018; 8:11721. [PMID: 30082755 PMCID: PMC6079063 DOI: 10.1038/s41598-018-30259-2] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2018] [Accepted: 07/26/2018] [Indexed: 12/15/2022] Open
Abstract
Chitin, which is a major component of house dust mites (HDM), fungi, crustaceans, etc., can activate immune cells, suggesting that it contributes to development of allergic disorders such as asthma. Although the pathophysiological sensitization route of asthmatic patients to allergens is considered via the respiratory tract, the roles of intranasally-administered chitin in development of asthma remain unclear. After ovalbumin (OVA) challenge, development of airway inflammation was profoundly exacerbated in mice sensitized with OVA in the presence of chitin. The exacerbation was dependent on IL-33, but not IL-25, thymic stromal lymphopoietin or IL-17A. Chitin enhanced IL-33-dependent IL-1β production by dendritic cells (DCs). Furthermore, chitin- and IL-33-stimulated DC-derived IL-1β promoted OVA-specific Th2 cell activation, resulting in aggravation of OVA-induced airway inflammation. These findings indicate the adjuvant activity of chitin via a new mechanism and provide important clues for development of therapeutics for allergic disorders caused by HDM, fungi and crustaceans.
Collapse
Grants
- Grants-in-Aid for Young Scientists (22790941 and 24791005) and Grants-in-Aid for Scientific Research (26461491) from the Ministry of Education, Culture, Sports, Science and Technology, Japan. The Grant for Joint Research Project of the Institute of Medical Science, the University of Tokyo (2024)
- Grants-in-Aid for Young Scientists (25860822) from the Ministry of Education, Culture, Sports, Science and Technology, Japan
- Grants-in-Aid for Challenging Exploratory Research (15K15377 and 16K15515) from the Ministry of Education, Culture, Sports, Science and Technology, Japan.A Health Labour Sciences Research Grant from the Ministry of Health, Labour and Welfare, Japan.
- Grants-in-Aid for Young Scientists (21790942 and 24688029) and the Program for Improvement of Research Environment for Young Researchers, The Special Coordination Funds for Promoting Science and Technology from the Ministry of Education, Culture, Sports, Science and Technology, Japan. Precursory Research for Embryonic Science and Technology, Japan Science and Technology Agency. A Health Labour Sciences Research Grant from the Ministry of Health, Labour and Welfare, Japan
Collapse
Affiliation(s)
- Ken Arae
- Department of Immunology, Faculty of Health Sciences, Kyorin University, Tokyo, 181-8612, Japan
- Department of Allergy and Clinical Immunology, National Research Institute for Child Health and Development, Tokyo, 157-8535, Japan
| | - Hideaki Morita
- Department of Allergy and Clinical Immunology, National Research Institute for Child Health and Development, Tokyo, 157-8535, Japan
| | - Hirotoshi Unno
- Department of Allergy and Clinical Immunology, National Research Institute for Child Health and Development, Tokyo, 157-8535, Japan
| | - Kenichiro Motomura
- Department of Allergy and Clinical Immunology, National Research Institute for Child Health and Development, Tokyo, 157-8535, Japan
| | - Sumika Toyama
- Department of Allergy and Clinical Immunology, National Research Institute for Child Health and Development, Tokyo, 157-8535, Japan
| | - Naoko Okada
- Department of Allergy and Clinical Immunology, National Research Institute for Child Health and Development, Tokyo, 157-8535, Japan
| | - Tatsukuni Ohno
- Department of Molecular Immunology, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, Tokyo, 113-8510, Japan
| | - Masato Tamari
- Department of Allergy and Clinical Immunology, National Research Institute for Child Health and Development, Tokyo, 157-8535, Japan
| | - Keisuke Orimo
- Department of Allergy and Clinical Immunology, National Research Institute for Child Health and Development, Tokyo, 157-8535, Japan
| | - Yuko Mishima
- Department of Immunology, Faculty of Health Sciences, Kyorin University, Tokyo, 181-8612, Japan
| | - Hajime Suto
- Atopy Research Center, Juntendo University, Tokyo, 113-0033, Japan
| | - Ko Okumura
- Atopy Research Center, Juntendo University, Tokyo, 113-0033, Japan
| | - Katsuko Sudo
- Animal Research Center, Tokyo Medical University, Tokyo, 160-8402, Japan
| | - Hiroshi Miyazawa
- Department of Medical technology, Faculty of Health Sciences, Kyorin University, Tokyo, 181-8612, Japan
| | - Haruhiko Taguchi
- Department of Immunology, Faculty of Health Sciences, Kyorin University, Tokyo, 181-8612, Japan
| | - Hirohisa Saito
- Department of Allergy and Clinical Immunology, National Research Institute for Child Health and Development, Tokyo, 157-8535, Japan
| | - Kenji Matsumoto
- Department of Allergy and Clinical Immunology, National Research Institute for Child Health and Development, Tokyo, 157-8535, Japan
| | - Susumu Nakae
- Laboratory of Systems Biology, Center for Experimental Medicine and Systems Biology, The Institute of Medical Science, The University of Tokyo, Tokyo, 108-8639, Japan.
- Precursory Research for Embryonic Science and Technology (PREST), Japan Science and Technology Agency, Saitama, Japan.
| |
Collapse
|
26
|
Abstract
The extracellular forms of the IL-1 cytokines are active through binding to specific receptors on the surface of target cells. IL-1 ligands bind to the extracellular portion of their ligand-binding receptor chain. For signaling to take place, a non-binding accessory chain is recruited into a heterotrimeric complex. The intracellular approximation of the Toll-IL-1-receptor (TIR) domains of the 2 receptor chains is the event that initiates signaling. The family of IL-1 receptors (IL-1R) includes 10 structurally related members, and the distantly related soluble protein IL-18BP that acts as inhibitor of the cytokine IL-18. Over the years the receptors of the IL-1 family have been known with many different names, with significant confusion. Thus, we will use here a recently proposed unifying nomenclature. The family includes several ligand-binding chains (IL-1R1, IL-1R2, IL-1R4, IL-1R5, and IL-1R6), 2 types of accessory chains (IL-1R3, IL-1R7), molecules that act as inhibitors of signaling (IL-1R2, IL-1R8, IL-18BP), and 2 orphan receptors (IL-1R9, IL-1R10). In this review, we will examine how the receptors of the IL-1 family regulate the inflammatory and anti-inflammatory functions of the IL-1 cytokines and are, more at large, involved in modulating defensive and pathological innate immunity and inflammation. Regulation of the IL-1/IL-1R system in the brain will be also described, as an example of the peculiarities of organ-specific modulation of inflammation.
Collapse
Affiliation(s)
- Diana Boraschi
- Institute of Protein Biochemistry, National Research Council, Naples, Italy
| | - Paola Italiani
- Institute of Protein Biochemistry, National Research Council, Naples, Italy
| | - Sabrina Weil
- Immunology FB08, Justus-Liebig-Universitat Giessen, Giessen, Germany
| | - Michael U Martin
- Immunology FB08, Justus-Liebig-Universitat Giessen, Giessen, Germany
| |
Collapse
|
27
|
Zhao Y, De Los Santos FG, Wu Z, Liu T, Phan SH. An ST2-dependent role of bone marrow-derived group 2 innate lymphoid cells in pulmonary fibrosis. J Pathol 2018; 245:399-409. [DOI: 10.1002/path.5092] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2018] [Revised: 04/17/2018] [Accepted: 04/26/2018] [Indexed: 12/20/2022]
Affiliation(s)
- Yuyue Zhao
- Department of Respiratory Medicine, Peking Union Medical College Hospital; Chinese Academy of Medical Sciences and Peking Union Medical College; Beijing China
| | | | - Zhe Wu
- Department of Pathology; University of Michigan Medical School; Ann Arbor Michigan USA
| | - Tianju Liu
- Department of Pathology; University of Michigan Medical School; Ann Arbor Michigan USA
| | - Sem H Phan
- Department of Pathology; University of Michigan Medical School; Ann Arbor Michigan USA
| |
Collapse
|
28
|
Nakanishi K. Unique Action of Interleukin-18 on T Cells and Other Immune Cells. Front Immunol 2018; 9:763. [PMID: 29731751 PMCID: PMC5920033 DOI: 10.3389/fimmu.2018.00763] [Citation(s) in RCA: 167] [Impact Index Per Article: 23.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2018] [Accepted: 03/27/2018] [Indexed: 12/26/2022] Open
Abstract
Interleukin (IL)-18 was originally discovered as a factor that enhances interferon (IFN)-γ production by anti-CD3-stimulated Th1 cells, particularly in association with IL-12. IL-12 is a cytokine that induces development of Th1 cells. IL-18 cannot induce Th1 cell development, but has the capacity to activate established Th1 cells to produce IFN-γ in the presence of IL-12. Thus, IL-18 is regarded as a proinflammatory cytokine that facilitates type 1 responses. However, in the absence of IL-12 but presence of IL-2, IL-18 stimulates natural killer cells, NKT cells, and even established Th1 cells to produce IL-3, IL-9, and IL-13. Thus, IL-18 also facilitates type 2 responses. This unique function of IL-18 contributes to infection-associated allergic diseases. Together with IL-3, IL-18 stimulates mast cells and basophils to produce IL-4, IL-13, and chemical mediators such as histamine. Thus, IL-18 also induces innate-type allergic inflammation. IL-18 belongs to the IL-1 family of cytokines, which share similar molecular structures, receptors structures, and signal transduction pathways. Nevertheless, IL-18 shows a unique function by binding to a specific receptor expressed on distinct types of cells. In this review article, I will focus on the unique features of IL-18 in lymphocytes, basophils, and mast cells, particularly in comparison with IL-33.
Collapse
Affiliation(s)
- Kenji Nakanishi
- Department of Immunology, Hyogo College of Medicine, Hyogo, Japan
| |
Collapse
|
29
|
Khaitov MR, Gaisina AR, Shilovskiy IP, Smirnov VV, Ramenskaia GV, Nikonova AA, Khaitov RM. The Role of Interleukin-33 in Pathogenesis of Bronchial Asthma. New Experimental Data. BIOCHEMISTRY (MOSCOW) 2018. [PMID: 29534664 DOI: 10.1134/s0006297918010029] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
Interleukin-33 (IL-33) belongs to the IL-1 cytokine family and plays an important role in modulating immune system by inducing Th2 immune response via the ST2 membrane receptor. Epithelial cells are the major producers of IL-33. However, IL-33 is also secreted by other cells, e.g., bone marrow cells, dendritic cells, macrophages, and mast cells. IL-33 targets a broad range of cell types bearing the ST2 surface receptor. Many ST2-positive cells, such as Th2 cells, mast cells, basophils, and eosinophils, are involved in the development of allergic bronchial asthma (BA). This suggests that IL-33 directly participates in BA pathogenesis. Currently, the role of IL-33 in pathogenesis of inflammatory disorders, including BA, has been extensively investigated using clinical samples collected from patients, as well as asthma animal models. In particular, numerous studies on blocking IL-33 and its receptor by monoclonal antibodies in asthma mouse model have been performed over the last several years; IL-33- and ST2-deficient transgenic mice have also been generated. In this review, we summarized and analyzed the data on the role of IL-33 in BA pathogenesis and the prospects for creating new treatments for BA.
Collapse
Affiliation(s)
- M R Khaitov
- Institute of Immunology, FMBA of Russia, Moscow, 115478, Russia.
| | | | | | | | | | | | | |
Collapse
|
30
|
Abstract
Allergic inflammation is a type 2 immune disorder classically characterized by high levels of immunoglobulin E (IgE) and the development of Th2 cells. Asthma is a pulmonary allergic inflammatory disease resulting in bronchial hyper-reactivity. Atopic asthma is defined by IgE antibody-mediated mast cell degranulation, while in non-atopic asthma there is no allergen-specific IgE and more involvement of innate immune cells, such as basophils, group 2 innate lymphoid cells (ILC2), and eosinophils. Recently, protease allergens were shown to cause asthmatic responses in the absence of Th2 cells, suggesting that an innate cell network (IL-33/TSLP-basophil-ILC2-IL-5/IL-13 axis) can facilitate the sensitization phase of type 2 inflammatory responses. Recent evidence also indicates that in the chronic phase, these innate immune cells directly or indirectly contribute to the adaptive Th2 cell responses. In this review, we discuss the role of Th2 cytokines (IL-4 and IL-13) and innate immune cells (mast cells, basophils, ILC2s, and dendritic cells) in the cross-talk between innate and adaptive inflammatory responses.
Collapse
Affiliation(s)
- Masato Kubo
- Division of Molecular Pathology, Research Institute for Biomedical Science, Tokyo University of Science, Noda, Japan.,Laboratory for Cytokine Regulation, Research Center for Integrative Medical Science (IMS), RIKEN Yokohama Institute, Yokohama, Kanagawa, Japan
| |
Collapse
|
31
|
Tumes DJ, Papadopoulos M, Endo Y, Onodera A, Hirahara K, Nakayama T. Epigenetic regulation of T-helper cell differentiation, memory, and plasticity in allergic asthma. Immunol Rev 2018; 278:8-19. [PMID: 28658556 DOI: 10.1111/imr.12560] [Citation(s) in RCA: 67] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
An estimated 300 million people currently suffer from asthma, which causes approximately 250 000 deaths a year. Allergen-specific T-helper (Th) cells produce cytokines that induce many of the hallmark features of asthma including airways hyperreactivity, eosinophilic and neutrophilic inflammation, mucus hypersecretion, and airway remodeling. Cytokine-producing Th subsets including Th1 (IFN-γ), Th2 (IL-4, IL-5, IL-13), Th9 (IL-9), Th17 (IL-17), Th22 (IL-22), and T regulatory (IL-10) cells have all been suggested to play a role in the development of asthma. Th differentiation involves genetic regulation of gene expression through the concerted action of cytokines, transcription factors, and epigenetic regulators. We describe how Th differentiation and plasticity is regulated by epigenetic histone and DNA modifications, with a focus on the regulation of histone methylation by members of the polycomb and trithorax complexes. In addition, we outline environmental influences that could influence epigenetic regulation of Th cells and discuss the potential to regulate Th plasticity and function through drugs targeting the epigenetic machinery. It is also becoming apparent that epigenetic regulation of allergen-specific memory Th cells may be important in the development and persistence of chronic allergies. Finally, we describe how epigenetic modifiers regulate cytokine memory in Th cells and describe recently identified hybrid, plastic, and pathogenic memory Th subsets the context of allergic asthma.
Collapse
Affiliation(s)
- Damon J Tumes
- Department of Immunology, Graduate School of Medicine, Chiba University, Chiba, Japan.,South Australian Health and Medical Research Institute, Adelaide, South Australia, Australia
| | | | - Yusuke Endo
- Department of Immunology, Graduate School of Medicine, Chiba University, Chiba, Japan
| | - Atsushi Onodera
- Department of Immunology, Graduate School of Medicine, Chiba University, Chiba, Japan
| | - Kiyoshi Hirahara
- Department of Immunology, Graduate School of Medicine, Chiba University, Chiba, Japan
| | - Toshinori Nakayama
- Department of Immunology, Graduate School of Medicine, Chiba University, Chiba, Japan.,AMED-CREST, AMED, Chiba, Japan
| |
Collapse
|
32
|
Eissmann MF, Dijkstra C, Wouters MA, Baloyan D, Mouradov D, Nguyen PM, Davalos-Salas M, Putoczki TL, Sieber OM, Mariadason JM, Ernst M, Masson F. Interleukin 33 Signaling Restrains Sporadic Colon Cancer in an Interferon-γ-Dependent Manner. Cancer Immunol Res 2018; 6:409-421. [PMID: 29463593 DOI: 10.1158/2326-6066.cir-17-0218] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2017] [Revised: 10/13/2017] [Accepted: 01/30/2018] [Indexed: 11/16/2022]
Abstract
Interleukin 33 (IL33) is an inflammatory cytokine released during necrotic cell death. The epithelium and stroma of the intestine express large amounts of IL33 and its receptor St2. IL33 is therefore continuously released during homeostatic turnover of the intestinal mucosa. Although IL33 can prevent colon cancer associated with inflammatory colitis, the contribution of IL33 signaling to sporadic colon cancer remains unknown. Here, we utilized a mouse model of sporadic colon cancer to investigate the contribution of IL33 signaling to tumorigenesis in the absence of preexisting inflammation. We demonstrated that genetic ablation of St2 enhanced colon tumor development. Conversely, administration of recombinant IL33 reduced growth of colon cancer cell allografts. In reciprocal bone marrow chimeras, the concurrent loss of IL33 signaling within radioresistant nonhematopoietic, and the radiosensitive hematopoietic, compartments was associated with increased tumor burden. We detected St2 expression within the radioresistant mesenchymal cell compartment of the colon whose stimulation with IL33 induced expression of bona fide NF-κB target genes. Mechanistically, we discovered that St2 deficiency within the nonhematopoietic compartment coincided with increased abundance of regulatory T cells and suppression of an IFNγ gene expression signature, whereas IL33 administration triggered IFNγ expression by tumor allograft-infiltrating T cells. The decrease of this IFNγ gene expression signature was associated with more aggressive disease in human colon cancer patients, suggesting that lack of IL33 signaling impaired the generation of a potent IFNγ-mediated antitumor immune response. Collectively, our data reveal that IL33 functions as a tumor suppressor in sporadic colon cancer. Cancer Immunol Res; 6(4); 409-21. ©2018 AACR.
Collapse
Affiliation(s)
- Moritz F Eissmann
- Cancer and Inflammation Laboratory, Olivia Newton-John Cancer Research Institute and La Trobe University School of Cancer Medicine, Heidelberg, Victoria, Australia
| | - Christine Dijkstra
- Cancer and Inflammation Laboratory, Olivia Newton-John Cancer Research Institute and La Trobe University School of Cancer Medicine, Heidelberg, Victoria, Australia
| | - Merridee A Wouters
- Cancer and Inflammation Laboratory, Olivia Newton-John Cancer Research Institute and La Trobe University School of Cancer Medicine, Heidelberg, Victoria, Australia
| | - David Baloyan
- Cancer and Inflammation Laboratory, Olivia Newton-John Cancer Research Institute and La Trobe University School of Cancer Medicine, Heidelberg, Victoria, Australia
| | - Dmitri Mouradov
- Systems Biology and Personalised Medicine Division, The Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria, Australia.,Department of Medical Biology, The University of Melbourne, Parkville, Victoria, Australia
| | - Paul M Nguyen
- Department of Medical Biology, The University of Melbourne, Parkville, Victoria, Australia.,Inflammation Division, The Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria, Australia
| | - Mercedes Davalos-Salas
- Oncogenic Transcription Laboratory, Olivia Newton-John Cancer Research Institute and La Trobe University School of Cancer Medicine, Heidelberg, Victoria, Australia
| | - Tracy L Putoczki
- Department of Medical Biology, The University of Melbourne, Parkville, Victoria, Australia.,Inflammation Division, The Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria, Australia
| | - Oliver M Sieber
- Systems Biology and Personalised Medicine Division, The Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria, Australia.,Department of Medical Biology, The University of Melbourne, Parkville, Victoria, Australia.,Department of Surgery, The University of Melbourne, Parkville, Victoria, Australia.,School of Biomedical Sciences, Monash University, Clayton, Victoria, Australia
| | - John M Mariadason
- Oncogenic Transcription Laboratory, Olivia Newton-John Cancer Research Institute and La Trobe University School of Cancer Medicine, Heidelberg, Victoria, Australia
| | - Matthias Ernst
- Cancer and Inflammation Laboratory, Olivia Newton-John Cancer Research Institute and La Trobe University School of Cancer Medicine, Heidelberg, Victoria, Australia.
| | - Frederick Masson
- Cancer and Inflammation Laboratory, Olivia Newton-John Cancer Research Institute and La Trobe University School of Cancer Medicine, Heidelberg, Victoria, Australia.
| |
Collapse
|
33
|
Vainchtein ID, Chin G, Cho FS, Kelley KW, Miller JG, Chien EC, Liddelow SA, Nguyen PT, Nakao-Inoue H, Dorman LC, Akil O, Joshita S, Barres BA, Paz JT, Molofsky AB, Molofsky AV. Astrocyte-derived interleukin-33 promotes microglial synapse engulfment and neural circuit development. Science 2018; 359:1269-1273. [PMID: 29420261 DOI: 10.1126/science.aal3589] [Citation(s) in RCA: 449] [Impact Index Per Article: 64.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2016] [Revised: 12/04/2017] [Accepted: 01/17/2018] [Indexed: 12/14/2022]
Abstract
Neuronal synapse formation and remodeling are essential to central nervous system (CNS) development and are dysfunctional in neurodevelopmental diseases. Innate immune signals regulate tissue remodeling in the periphery, but how this affects CNS synapses is largely unknown. Here, we show that the interleukin-1 family cytokine interleukin-33 (IL-33) is produced by developing astrocytes and is developmentally required for normal synapse numbers and neural circuit function in the spinal cord and thalamus. We find that IL-33 signals primarily to microglia under physiologic conditions, that it promotes microglial synapse engulfment, and that it can drive microglial-dependent synapse depletion in vivo. These data reveal a cytokine-mediated mechanism required to maintain synapse homeostasis during CNS development.
Collapse
Affiliation(s)
- Ilia D Vainchtein
- Department of Psychiatry/Weill Institute for Neurosciences, University of California, San Francisco, San Francisco, CA, USA
| | - Gregory Chin
- Department of Psychiatry/Weill Institute for Neurosciences, University of California, San Francisco, San Francisco, CA, USA
| | - Frances S Cho
- Neuroscience Graduate Program, University of California, San Francisco, San Francisco, CA, USA.,Gladstone Institute of Neurological Disease, San Francisco, CA 94158, USA
| | - Kevin W Kelley
- Department of Psychiatry/Weill Institute for Neurosciences, University of California, San Francisco, San Francisco, CA, USA
| | - John G Miller
- Department of Psychiatry/Weill Institute for Neurosciences, University of California, San Francisco, San Francisco, CA, USA
| | - Elliott C Chien
- Department of Psychiatry/Weill Institute for Neurosciences, University of California, San Francisco, San Francisco, CA, USA
| | - Shane A Liddelow
- Department of Neurobiology, Stanford University, Palo Alto, CA, USA
| | - Phi T Nguyen
- Department of Psychiatry/Weill Institute for Neurosciences, University of California, San Francisco, San Francisco, CA, USA.,Biomedical Sciences Graduate Program, University of California, San Francisco, San Francisco, CA, USA
| | - Hiromi Nakao-Inoue
- Department of Psychiatry/Weill Institute for Neurosciences, University of California, San Francisco, San Francisco, CA, USA
| | - Leah C Dorman
- Department of Psychiatry/Weill Institute for Neurosciences, University of California, San Francisco, San Francisco, CA, USA.,Neuroscience Graduate Program, University of California, San Francisco, San Francisco, CA, USA
| | - Omar Akil
- Department of Otolaryngology, University of California, San Francisco, San Francisco, CA, USA
| | - Satoru Joshita
- Department of Medicine, Division of Gastroenterology and Hepatology, Shinshu University School of Medicine, Matsumoto, Japan.,Research Center for Next Generation Medicine, Shinshu University, Matsumoto, Japan
| | - Ben A Barres
- Department of Neurobiology, Stanford University, Palo Alto, CA, USA
| | - Jeanne T Paz
- Department of Neurology, University of California, San Francisco, San Francisco, CA, USA.,Gladstone Institute of Neurological Disease, San Francisco, CA 94158, USA
| | - Ari B Molofsky
- Department of Laboratory Medicine, University of California, San Francisco, San Francisco, CA, USA.
| | - Anna V Molofsky
- Department of Psychiatry/Weill Institute for Neurosciences, University of California, San Francisco, San Francisco, CA, USA.
| |
Collapse
|
34
|
Zhao J, Zhao Y. Interleukin-33 and its Receptor in Pulmonary Inflammatory Diseases. Crit Rev Immunol 2018; 35:451-61. [PMID: 27279043 DOI: 10.1615/critrevimmunol.2016015865] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Interleukin-33 (IL-33) is a member of the IL-1 cytokine family. It modulates immune responses and biological functions through binding to its membrane receptor, ST2L. ST2L is a member of the Toll-like/IL-1 (TIR)-receptor superfamily, and its isoform, soluble ST2 (sST2), functions as an inhibitor of the IL-33/ST2L pathway. Levels of IL-33 and sST2 in serum and bronchoalveolar lavage fluid (BAL) are known biomarkers for a variety of disorders such as heart failure, non-small-cell lung cancer, and pulmonary inflammatory diseases. IL-33 also exists in the nuclei, and nuclear IL-33 seems to regulate cytokine gene expression. In this review, we focus on the role of IL-33/ST2 in the pathogenesis of pulmonary inflammatory diseases including asthma, chronic obstructive pulmonary disease (COPD), and lung injury.
Collapse
Affiliation(s)
- Jing Zhao
- Department of Medicine, University of Pittsburgh School of Medicine, Acute Lung Injury Center of Excellence, and Vascular Medical Institute, University of Pittsburgh, Pittsburgh, PA, United States
| | - Yutong Zhao
- Department of Medicine, University of Pittsburgh School of Medicine, Acute Lung Injury Center of Excellence, and Vascular Medical Institute, University of Pittsburgh, Pittsburgh, PA, United States
| |
Collapse
|
35
|
Bonyadi Rad E, Musumeci G, Pichler K, Heidary M, Szychlinska MA, Castrogiovanni P, Marth E, Böhm C, Srinivasaiah S, Krönke G, Weinberg A, Schäfer U. Runx2 mediated Induction of Novel Targets ST2 and Runx3 Leads to Cooperative Regulation of Hypertrophic Differentiation in ATDC5 Chondrocytes. Sci Rep 2017; 7:17947. [PMID: 29263341 PMCID: PMC5738421 DOI: 10.1038/s41598-017-18044-z] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2017] [Accepted: 12/04/2017] [Indexed: 11/17/2022] Open
Abstract
Knowledge concerning expression and function of Suppression of Tumorigenicity 2 (ST2) in chondrocytes is at present, limited. Analysis of murine growth plates and ATDC5 chondrocytes indicated peak expression of the ST2 transmembrane receptor (ST2L) and soluble (sST2) isoforms during the hypertrophic differentiation concomitant with the expression of the hypertrophic markers Collagen X (Col X), Runx2 and MMP-13. Gain- and loss-of-function experiments in ATDC5 and primary human growth plate chondrocytes (PHCs), confirmed regulation of ST2 by the key transcription factor Runx2, indicating ST2 to be a novel Runx2 target. ST2 knock-out mice (ST2−/−) exhibited noticeable hypertrophic zone (HZ) reduction in murine growth plates, accompanied by lower expression of Col X and Osteocalcin (OSC) compared to wild-type (WT) mice. Likewise, ST2 knockdown resulted in decreased Col X expression and downregulation of OSC and Vascular Endothelial Growth Factor (VEGF) in ATDC5 cells. The ST2 suppression was also associated with upregulation of the proliferative stage markers Sox9 and Collagen II (Col II), indicating ST2 to be a new regulator of ATDC5 chondrocyte differentiation. Runx3 was, furthermore, identified as a novel Runx2 target in chondrocytes. This study suggests that Runx2 mediates ST2 and Runx3 induction to cooperatively regulate hypertrophic differentiation of ATDC5 chondrocytes.
Collapse
Affiliation(s)
- Ehsan Bonyadi Rad
- Department of Orthopedics and Trauma Surgery, Medical University Graz, Graz, Austria.
| | - Giuseppe Musumeci
- Department of Biomedical and Biotechnological Sciences, Human Anatomy and Histology Section, School of Medicine, University of Catania, Catania, Italy
| | - Karin Pichler
- Department of Children and Adolescent Medicine, Pediatrics I, Medical University of Innsbruck, Innsbruck, Austria.,Division of Neonatology, Pediatric Intensive Care and Neuropediatrics, Department of Pediatrics, Medical University of Vienna, Vienna, Austria
| | - Maryam Heidary
- Translational Research Department, Institute Curie, Paris, France
| | - Marta Anna Szychlinska
- Department of Biomedical and Biotechnological Sciences, Human Anatomy and Histology Section, School of Medicine, University of Catania, Catania, Italy
| | - Paola Castrogiovanni
- Department of Biomedical and Biotechnological Sciences, Human Anatomy and Histology Section, School of Medicine, University of Catania, Catania, Italy
| | - Egon Marth
- Institute of Hygiene, Microbiology and Environmental Medicine, Medical University Graz, Graz, Austria
| | - Christina Böhm
- Friedrich-Alexander-University Erlangen-Nürnberg (FAU), Department of Internal Medicine 3 - Rheumatology and Immunology, Universitätsklinikum Erlangen, Erlangen, Germany
| | - Sriveena Srinivasaiah
- Department of Orthopedics and Trauma Surgery, Medical University Graz, Graz, Austria
| | - Gerhard Krönke
- Friedrich-Alexander-University Erlangen-Nürnberg (FAU), Department of Internal Medicine 3 - Rheumatology and Immunology, Universitätsklinikum Erlangen, Erlangen, Germany
| | - Annelie Weinberg
- Department of Orthopedics and Trauma Surgery, Medical University Graz, Graz, Austria
| | - Ute Schäfer
- Department of Neurosurgery, Medical University Graz, Graz, Austria
| |
Collapse
|
36
|
Verma M, Liu S, Michalec L, Sripada A, Gorska MM, Alam R. Experimental asthma persists in IL-33 receptor knockout mice because of the emergence of thymic stromal lymphopoietin-driven IL-9 + and IL-13 + type 2 innate lymphoid cell subpopulations. J Allergy Clin Immunol 2017; 142:793-803.e8. [PMID: 29132961 DOI: 10.1016/j.jaci.2017.10.020] [Citation(s) in RCA: 45] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2017] [Revised: 09/07/2017] [Accepted: 10/10/2017] [Indexed: 01/17/2023]
Abstract
BACKGROUND IL-33 plays an important role in the development of experimental asthma. OBJECTIVE We sought to study the role of the IL-33 receptor suppressor of tumorigenicity 2 (ST2) in the persistence of asthma in a mouse model. METHODS We studied allergen-induced experimental asthma in ST2 knockout (KO) and wild-type control mice. We measured airway hyperresponsiveness by using flexiVent; inflammatory indices by using ELISA, histology, and real-time PCR; and type 2 innate lymphoid cells (ILC2s) in lung single-cell preparations by using flow cytometry. RESULTS Airway hyperresponsiveness was increased in allergen-treated ST2 KO mice and comparable with that in allergen-treated wild-type control mice. Peribronchial and perivascular inflammation and mucus production were largely similar in both groups. Persistence of experimental asthma in ST2 KO mice was associated with an increase in levels of thymic stromal lymphopoietin (TSLP), IL-9, and IL-13, but not IL-5, in bronchoalveolar lavage fluid. Expectedly, ST2 deletion caused a reduction in IL-13+ CD4 T cells, forkhead box P3-positive regulatory T cells, and IL-5+ ILC2s. Unexpectedly, ST2 deletion led to an overall increase in innate lymphoid cells (CD45+lin-CD25+ cells) and IL-13+ ILC2s, emergence of a TSLP receptor-positive IL-9+ ILC2 population, and an increase in intraepithelial mast cell numbers in the lung. An anti-TSLP antibody abrogated airway hyperresponsiveness, inflammation, and mucus production in allergen-treated ST2 KO mice. It also caused a reduction in innate lymphoid cell, ILC2, and IL-9+ and IL-13+ ILC2 numbers in the lung. CONCLUSIONS Genetic deletion of the IL-33 receptor paradoxically increases TSLP production, which stimulates the emergence of IL-9+ and IL-13+ ILC2s and mast cells and leads to development of chronic experimental asthma. An anti-TSLP antibody abrogates all pathologic features of asthma in this model.
Collapse
Affiliation(s)
- Mukesh Verma
- Department of Medicine, Division of Allergy & Immunology, National Jewish Health, Denver, Colo
| | - Sucai Liu
- Department of Medicine, Division of Allergy & Immunology, National Jewish Health, Denver, Colo
| | - Lidia Michalec
- Department of Medicine, Division of Allergy & Immunology, National Jewish Health, Denver, Colo; Department of Cytobiology and Proteomics, Medical University of Lodz, Lodz, Poland
| | - Anand Sripada
- Department of Medicine, Division of Allergy & Immunology, National Jewish Health, Denver, Colo
| | - Magdalena M Gorska
- Department of Medicine, Division of Allergy & Immunology, National Jewish Health, Denver, Colo; School of Medicine, University of Colorado Denver, Denver, Colo
| | - Rafeul Alam
- Department of Medicine, Division of Allergy & Immunology, National Jewish Health, Denver, Colo; School of Medicine, University of Colorado Denver, Denver, Colo.
| |
Collapse
|
37
|
Angkasekwinai P, Sodthawon W, Jeerawattanawart S, Hansakon A, Pattanapanyasat K, Wang YH. ILC2s activated by IL-25 promote antigen-specific Th2 and Th9 functions that contribute to the control of Trichinella spiralis infection. PLoS One 2017; 12:e0184684. [PMID: 28898280 PMCID: PMC5595335 DOI: 10.1371/journal.pone.0184684] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2017] [Accepted: 08/29/2017] [Indexed: 01/21/2023] Open
Abstract
IL-25, an IL-17 family cytokine, derived from epithelial cells was shown to regulate Th2- and Th9-type immune responses. We previously reported that IL-25 was important in promoting efficient protective immunity against T. spiralis infection; however, the cellular targets of IL-25 to elicit type-2 immunity during infection have not yet been addressed. Here, we investigated IL-25-responding cells and their involvement in mediating type-2 immune response during T. spiralis infection. ILC2 and CD4+ Th2 cells residing in the gastrointestinal tract of T. spiralis infected mice were found to express high levels of surface interleukin-17 receptor B (IL-17RB), a component of the IL-25 receptor. Following T. spiralis infection, activated ILC2s upregulated surface MHCII expression and enhanced capacity of effector T helper cell in producing antigen-specific Th2 and Th9 cytokines through MHCII-dependent interactions. Reciprocally, lack of CD4+ T helper cells impaired ILC2 function to produce type 2-associated cytokines in responding to IL-25 during T. spiralis infection. Furthermore, mice deficient in IL-17RB showed markedly reduced ILC2 numbers and antigen-specific Th2 and Th9 cytokine production during T. spiralis infection. The Il17rb-/- mice failed to mount effective antigen specific Th2 and Th9 functions resulting in diminished goblet cell and mast cell responses, leading to delayed worm expulsion in the intestines and muscles. Thus, our data indicated that ILC2s and CD4+ Th2 cells are the predominant cellular targets of IL-25 following T. spiralis infection and their collaborative interactions may play a key role in mounting effective antigen-specific Th2 and Th9 cytokine responses against T. spiralis infection.
Collapse
Affiliation(s)
- Pornpimon Angkasekwinai
- Department of Medical Technology, Faculty of Allied Health Sciences, Thammasat University, Pathumthani Thailand.,Graduate Program in Biomedical Sciences, Faculty of Allied Health Sciences, Thammasat University, Pathumthani Thailand
| | - Wichuda Sodthawon
- Department of Medical Technology, Faculty of Allied Health Sciences, Thammasat University, Pathumthani Thailand
| | - Siranart Jeerawattanawart
- Department of Medical Technology, Faculty of Allied Health Sciences, Thammasat University, Pathumthani Thailand.,Graduate Program in Biomedical Sciences, Faculty of Allied Health Sciences, Thammasat University, Pathumthani Thailand
| | - Adithap Hansakon
- Department of Medical Technology, Faculty of Allied Health Sciences, Thammasat University, Pathumthani Thailand.,Graduate Program in Biomedical Sciences, Faculty of Allied Health Sciences, Thammasat University, Pathumthani Thailand
| | - Kovit Pattanapanyasat
- Center of Excellence for Flow Cytometry, Office for Research and Development, Faculty of Medicine, Siriraj Hospital, Mahidol University, Bangkok, Thailand
| | - Yui-Hsi Wang
- Division of Allergy and Immunology, University of Cincinnati, Cincinnati Children's Hospital Medical Center, Cincinnati, OH United States of America
| |
Collapse
|
38
|
Finlay CM, Stefanska AM, Coleman MM, Jahns H, Cassidy JP, McLoughlin RM, Mills KHG. Secreted products of Fasciola hepatica inhibit the induction of T cell responses that mediate allergy. Parasite Immunol 2017; 39. [PMID: 28815724 DOI: 10.1111/pim.12460] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2017] [Accepted: 08/11/2017] [Indexed: 12/24/2022]
Abstract
There is evidence from epidemiology studies of a negative association between infection with helminth parasites and the development of allergy and asthma. Here, we demonstrate that the excretory/secretory products of the helminth Fasciola hepatica (FHES) protected mice against ovalbumin (OVA)-induced allergic asthma when administered at time of allergen sensitization. FHES reduced the accumulation of mucus, eosinophils and lymphocytes into the airways of allergen-challenged mice. Furthermore, FHES treatment suppressed Th2 responses in the airways. Interestingly, systemic administration of FHES at allergen challenge had no effect on airway inflammation, demonstrating that alum-induced Th2 response is set following initial allergen sensitization. Our findings highlight the immunomodulatory potential of molecules secreted by F. hepatica.
Collapse
Affiliation(s)
- C M Finlay
- Immune Regulation Research Group, School of Biochemistry and Immunology, Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin 2, Ireland
| | - A M Stefanska
- Immune Regulation Research Group, School of Biochemistry and Immunology, Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin 2, Ireland
| | - M M Coleman
- Immune Regulation Research Group, School of Biochemistry and Immunology, Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin 2, Ireland
| | - H Jahns
- School of Veterinary Medicine, Veterinary Science Centre, University College Dublin, Dublin 4, Ireland
| | - J P Cassidy
- School of Veterinary Medicine, Veterinary Science Centre, University College Dublin, Dublin 4, Ireland
| | - R M McLoughlin
- Host-Pathogen Interactions Group School of Biochemistry and Immunology, Trinity Biomedical Sciences Institute, Dublin 2, Ireland
| | - K H G Mills
- Immune Regulation Research Group, School of Biochemistry and Immunology, Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin 2, Ireland
| |
Collapse
|
39
|
Morikawa T, Fukuoka A, Matsushita K, Yasuda K, Iwasaki N, Akasaki S, Fujieda S, Yoshimoto T. Activation of group 2 innate lymphoid cells exacerbates and confers corticosteroid resistance to mouse nasal type 2 inflammation. Int Immunol 2017; 29:221-233. [DOI: 10.1093/intimm/dxx030] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2017] [Accepted: 05/21/2017] [Indexed: 12/13/2022] Open
|
40
|
Griesenauer B, Paczesny S. The ST2/IL-33 Axis in Immune Cells during Inflammatory Diseases. Front Immunol 2017; 8:475. [PMID: 28484466 PMCID: PMC5402045 DOI: 10.3389/fimmu.2017.00475] [Citation(s) in RCA: 436] [Impact Index Per Article: 54.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2016] [Accepted: 04/05/2017] [Indexed: 12/16/2022] Open
Abstract
Il1rl1 (also known as ST2) is a member of the IL-1 superfamily, and its only known ligand is IL-33. ST2 exists in two forms as splice variants: a soluble form (sST2), which acts as a decoy receptor, sequesters free IL-33, and does not signal, and a membrane-bound form (ST2), which activates the MyD88/NF-κB signaling pathway to enhance mast cell, Th2, regulatory T cell (Treg), and innate lymphoid cell type 2 functions. sST2 levels are increased in patients with active inflammatory bowel disease, acute cardiac and small bowel transplant allograft rejection, colon and gastric cancers, gut mucosal damage during viral infection, pulmonary disease, heart disease, and graft-versus-host disease. Recently, sST2 has been shown to be secreted by intestinal pro-inflammatory T cells during gut inflammation; on the contrary, protective ST2-expressing Tregs are decreased, implicating that ST2/IL-33 signaling may play an important role in intestinal disease. This review will focus on what is known on its signaling during various inflammatory disease states and highlight potential avenues to intervene in ST2/IL-33 signaling as treatment options.
Collapse
Affiliation(s)
- Brad Griesenauer
- Department of Pediatrics, Indiana University, Indianapolis, IN, USA
- Department of Microbiology Immunology, Indiana University, Indianapolis, IN, USA
- Melvin and Bren Simon Cancer Center, Indiana University, Indianapolis, IN, USA
| | - Sophie Paczesny
- Department of Pediatrics, Indiana University, Indianapolis, IN, USA
- Department of Microbiology Immunology, Indiana University, Indianapolis, IN, USA
- Melvin and Bren Simon Cancer Center, Indiana University, Indianapolis, IN, USA
| |
Collapse
|
41
|
Seki T, Obata-Ninomiya K, Shimogawara-Furushima R, Arai T, Akao N, Hoshino T, Ohta N. IL-33/ST2 contributes to severe symptoms in Plasmodium chabaudi-infected BALB/c mice. Parasitol Int 2017; 67:64-69. [PMID: 28359899 DOI: 10.1016/j.parint.2017.03.008] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2016] [Revised: 03/22/2017] [Accepted: 03/27/2017] [Indexed: 02/07/2023]
Abstract
It has been reported that IL-33 contributes to potentiation of Th2 inflammatory diseases and protection against helminth infection. Increased plasma IL-33 levels have been observed in patients with severe falciparum malaria, however, the role of IL-33 in malaria remains unclear. Here we report that IL-33 enhances inflammatory responses in malaria infection. ST2-deficiency altered severity of inflammation in the liver and serum levels of pro-inflammatory cytokines such as TNF-α and IL-6, and IL-13 that is a Th2 cytokine during Plasmodium chabaudi infection. IL-13-deficient mice have similar phenotype with ST2-deficient mice during P. chabaudi infection. Furthermore, ST2- and IL-13-deficiency reduced mortality from P. chabaudi infection. These results indicate that IL-33/ST2 can induce production of proinflammatory cytokines, such as TNF-α and IL-6, through production of IL-13 in P. chabaudi-infected BALB/c mice, suggesting that IL-33/ST2 play a critical role in inflammatory responses to malaria infection. Thus, these findings may define a novel therapeutic target for patients with severe malaria.
Collapse
Affiliation(s)
- Takenori Seki
- Department of Environmental Parasitology, Tokyo Medical and Dental University, Tokyo, Japan
| | - Kazushige Obata-Ninomiya
- Department of Environmental Parasitology, Tokyo Medical and Dental University, Tokyo, Japan; Department of Immune Regulation, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, Tokyo, Japan
| | | | - Toshio Arai
- Department of Environmental Parasitology, Tokyo Medical and Dental University, Tokyo, Japan
| | - Nobuaki Akao
- Department of Environmental Parasitology, Tokyo Medical and Dental University, Tokyo, Japan
| | - Tomoaki Hoshino
- Division of Respirology, Neurology, and Rheumatology, Department of Medicine 1, Kurume University School of Medicine, Fukuoka, Japan
| | - Nobuo Ohta
- Department of Environmental Parasitology, Tokyo Medical and Dental University, Tokyo, Japan.
| |
Collapse
|
42
|
Nakayama T, Hirahara K, Onodera A, Endo Y, Hosokawa H, Shinoda K, Tumes DJ, Okamoto Y. Th2 Cells in Health and Disease. Annu Rev Immunol 2016; 35:53-84. [PMID: 27912316 DOI: 10.1146/annurev-immunol-051116-052350] [Citation(s) in RCA: 274] [Impact Index Per Article: 30.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Helper T (Th) cell subsets direct immune responses by producing signature cytokines. Th2 cells produce IL-4, IL-5, and IL-13, which are important in humoral immunity and protection from helminth infection and are central to the pathogenesis of many allergic inflammatory diseases. Molecular analysis of Th2 cell differentiation and maintenance of function has led to recent discoveries that have refined our understanding of Th2 cell biology. Epigenetic regulation of Gata3 expression by chromatin remodeling complexes such as Polycomb and Trithorax is crucial for maintaining Th2 cell identity. In the context of allergic diseases, memory-type pathogenic Th2 cells have been identified in both mice and humans. To better understand these disease-driving cell populations, we have developed a model called the pathogenic Th population disease induction model. The concept of defined subsets of pathogenic Th cells may spur new, effective strategies for treating intractable chronic inflammatory disorders.
Collapse
Affiliation(s)
- Toshinori Nakayama
- Department of Immunology, Graduate School of Medicine, Chiba University, Chiba 260-8670, Japan; , , , , , , , .,AMED-CREST, AMED, Chiba 260-8670, Japan
| | - Kiyoshi Hirahara
- Department of Immunology, Graduate School of Medicine, Chiba University, Chiba 260-8670, Japan; , , , , , , ,
| | - Atsushi Onodera
- Department of Immunology, Graduate School of Medicine, Chiba University, Chiba 260-8670, Japan; , , , , , , , .,Institute for Global Prominent Research, Chiba University, Chiba 260-8670, Japan
| | - Yusuke Endo
- Department of Immunology, Graduate School of Medicine, Chiba University, Chiba 260-8670, Japan; , , , , , , ,
| | - Hiroyuki Hosokawa
- Department of Immunology, Graduate School of Medicine, Chiba University, Chiba 260-8670, Japan; , , , , , , ,
| | - Kenta Shinoda
- Department of Immunology, Graduate School of Medicine, Chiba University, Chiba 260-8670, Japan; , , , , , , ,
| | - Damon J Tumes
- Department of Immunology, Graduate School of Medicine, Chiba University, Chiba 260-8670, Japan; , , , , , , , .,South Australian Health and Medical Research Institute, North Terrace, Adelaide SA 5000, Australia
| | - Yoshitaka Okamoto
- Department of Otorhinolaryngology, Graduate School of Medicine, Chiba University, Chiba 260-8670, Japan
| |
Collapse
|
43
|
IL-33-Induced Cytokine Secretion and Survival of Mouse Eosinophils Is Promoted by Autocrine GM-CSF. PLoS One 2016; 11:e0163751. [PMID: 27690378 PMCID: PMC5045177 DOI: 10.1371/journal.pone.0163751] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2016] [Accepted: 09/13/2016] [Indexed: 01/21/2023] Open
Abstract
Eosinophils are major effector cells during allergic responses and helminth infections. Recent studies further highlight eosinophils as important players in many other biological processes. Therefore it is important to understand how these cells can be modulated in terms of survival and effector function. In the present study we investigated how eosinophils respond to the alarmin IL-33. We show that IL-33 promotes eosinophil survival in a ST2- and MyD88-dependent manner. IL-33-mediated protection from apoptosis was dependent on autocrine GM-CSF release. In addition, GM-CSF increased the IL-33-induced secretion of IL-4 and IL-13 from eosinophils. Unexpectedly, this effect was further enhanced by cross-linking of Siglec-F, a proposed inhibitory and apopotosis-inducing receptor on eosinophils. Co-culture experiments with eosinophils and macrophages revealed that the IL-33-induced release of IL-4 and IL-13 from eosinophils was required for differentiation of alternatively activated macrophages (AAMs). The differentiation of AAMs could be further increased in the presence of GM-CSF. These results indicate that cross-talk between Siglec-F and the receptors for IL-33, LPS and GM-CSF plays an important role for efficient secretion of IL-4 and IL-13. Deciphering the molecular details of this cross-talk could lead to the development of new therapeutic option to treat eosinophil-associated diseases.
Collapse
|
44
|
Odegaard JI, Lee MW, Sogawa Y, Bertholet AM, Locksley RM, Weinberg DE, Kirichok Y, Deo RC, Chawla A. Perinatal Licensing of Thermogenesis by IL-33 and ST2. Cell 2016; 166:841-854. [PMID: 27453471 PMCID: PMC4985267 DOI: 10.1016/j.cell.2016.06.040] [Citation(s) in RCA: 93] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2016] [Revised: 06/08/2016] [Accepted: 06/20/2016] [Indexed: 01/09/2023]
Abstract
For placental mammals, the transition from the in utero maternal environment to postnatal life requires the activation of thermogenesis to maintain their core temperature. This is primarily accomplished by induction of uncoupling protein 1 (UCP1) in brown and beige adipocytes, the principal sites for uncoupled respiration. Despite its importance, how placental mammals license their thermogenic adipocytes to participate in postnatal uncoupled respiration is not known. Here, we provide evidence that the "alarmin" IL-33, a nuclear cytokine that activates type 2 immune responses, licenses brown and beige adipocytes for uncoupled respiration. We find that, in absence of IL-33 or ST2, beige and brown adipocytes develop normally but fail to express an appropriately spliced form of Ucp1 mRNA, resulting in absence of UCP1 protein and impairment in uncoupled respiration and thermoregulation. Together, these data suggest that IL-33 and ST2 function as a developmental switch to license thermogenesis during the perinatal period. PAPERCLIP.
Collapse
Affiliation(s)
- Justin I Odegaard
- Cardiovascular Research Institute, University of California, San Francisco, San Francisco, CA 94143-0795, USA
| | - Min-Woo Lee
- Cardiovascular Research Institute, University of California, San Francisco, San Francisco, CA 94143-0795, USA
| | - Yoshitaka Sogawa
- Cardiovascular Research Institute, University of California, San Francisco, San Francisco, CA 94143-0795, USA
| | - Ambre M Bertholet
- Department of Physiology, University of California, San Francisco, San Francisco, CA 94143-0795, USA
| | - Richard M Locksley
- Department of Medicine, University of California, San Francisco, San Francisco, CA 94143-0795, USA; Department Microbiology and Immunology, University of California, San Francisco, San Francisco, CA 94143-0795, USA; Howard Hughes Medical Institute, University of California, San Francisco, San Francisco, CA 94143-0795, USA
| | - David E Weinberg
- Department of Cellular and Molecular Pharmacology, University of California, San Francisco, San Francisco, CA 94143-0795, USA
| | - Yuriy Kirichok
- Department of Physiology, University of California, San Francisco, San Francisco, CA 94143-0795, USA
| | - Rahul C Deo
- Cardiovascular Research Institute, University of California, San Francisco, San Francisco, CA 94143-0795, USA; Department of Medicine, University of California, San Francisco, San Francisco, CA 94143-0795, USA
| | - Ajay Chawla
- Cardiovascular Research Institute, University of California, San Francisco, San Francisco, CA 94143-0795, USA; Department of Physiology, University of California, San Francisco, San Francisco, CA 94143-0795, USA; Department of Medicine, University of California, San Francisco, San Francisco, CA 94143-0795, USA.
| |
Collapse
|
45
|
Papazian D, Hansen S, Würtzen PA. Airway responses towards allergens - from the airway epithelium to T cells. Clin Exp Allergy 2016; 45:1268-87. [PMID: 25394747 DOI: 10.1111/cea.12451] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
The prevalence of allergic diseases such as allergic rhinitis is increasing, affecting up to 30% of the human population worldwide. Allergic sensitization arises from complex interactions between environmental exposures and genetic susceptibility, resulting in inflammatory T helper 2 (Th2) cell-derived immune responses towards environmental allergens. Emerging evidence now suggests that an epithelial dysfunction, coupled with inherent properties of environmental allergens, can be responsible for the inflammatory responses towards allergens. Several epithelial-derived cytokines, such as thymic stromal lymphopoietin (TSLP), IL-25 and IL-33, influence tissue-resident dendritic cells (DCs) as well as Th2 effector cells. Exposure to environmental allergens does not elicit Th2 inflammatory responses or any clinical symptoms in nonatopic individuals, and recent findings suggest that a nondamaged, healthy epithelium lowers the DCs' ability to induce inflammatory T-cell responses towards allergens. The purpose of this review was to summarize the current knowledge on which signals from the airway epithelium, from first contact with inhaled allergens all the way to the ensuing Th2-cell responses, influence the pathology of allergic diseases.
Collapse
Affiliation(s)
- D Papazian
- Department of Cancer & Inflammation Research, Institute of Molecular Medicine, University of Southern Denmark, Odense, Denmark.,ALK, Hørsholm, Denmark
| | - S Hansen
- Department of Cancer & Inflammation Research, Institute of Molecular Medicine, University of Southern Denmark, Odense, Denmark
| | | |
Collapse
|
46
|
Martin NT, Martin MU. Interleukin 33 is a guardian of barriers and a local alarmin. Nat Immunol 2016; 17:122-31. [DOI: 10.1038/ni.3370] [Citation(s) in RCA: 287] [Impact Index Per Article: 31.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2015] [Accepted: 12/01/2015] [Indexed: 12/12/2022]
|
47
|
Group 2 innate lymphoid cells utilize the IRF4-IL-9 module to coordinate epithelial cell maintenance of lung homeostasis. Mucosal Immunol 2016; 9:275-86. [PMID: 26129648 PMCID: PMC4698110 DOI: 10.1038/mi.2015.59] [Citation(s) in RCA: 161] [Impact Index Per Article: 17.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2015] [Accepted: 05/27/2015] [Indexed: 02/04/2023]
Abstract
Group 2 innate lymphoid cells (ILC2s) have an important role in acute allergic lung inflammation. Given their distribution and function, lung ILC2s are hypothesized to coordinate epithelial responses to the external environment; however, how barrier surveillance is linked to ILC2 activation remains unclear. Here, we demonstrate that alveolar type II cells are the main source of interleukin (IL)-33 and thymic stromal lymphopoietin (TSLP) generated in response to chitin or migratory helminths. IL-33 and TSLP synergistically induce an interferon regulatory factor 4 (IRF4)-IL-9 program in ILC2s, and autocrine IL-9 promotes rapid IL-5 and IL-13 production required for optimal epithelial responses in the conducting airways. Thus, ILC2s link alveolar function to regulation of airway flow, revealing a key interaction between resident lymphoid and structural cells that might underlie similar organizational hierarchies in other organs.
Collapse
|
48
|
Abstract
BACKGROUND IL-33, a member of the IL-1 cytokine family that signals through ST2, is upregulated in ulcerative colitis (UC); however, the role of IL-33 in colitis remains unclear. IL-33 augments type 2 immune responses, which have been implicated in UC pathogenesis. We sought to determine the role of IL-33 signaling in oxazolone (OXA) colitis, a type 2 cytokine-mediated murine model of UC. METHODS Colon mucosal IL-33 expression was compared between pediatric and adult UC and non-IBD patients using immunohistochemistry and real-time PCR. OXA colitis was induced in WT, IL-33, and ST2 mice, and histopathology, cytokine levels, and goblet cells were assessed. Transepithelial resistance was measured across IL-33-treated T84 cell monolayers. RESULTS Colon mucosal IL-33 was increased in pediatric patients with active UC and in OXA colitis. IL-33 and ST2 OXA mice exhibited increased disease severity compared with WT OXA mice. OXA induced a mixed mucosal cytokine response, but few differences were observed between OXA WT and IL-33 or ST2 mice. Goblet cells were significantly decreased in IL-33 and ST2 OXA compared with WT OXA mice. IL-33 augmented transepithelial resistance in T84 cells, and this effect was blocked by the ERK1/2 inhibitor PD98,059. CONCLUSIONS OXA colitis is exacerbated in IL-33 and ST2 mice. Increased mucosal IL-33 in human UC and murine colitis may be a homeostatic response to limit inflammation, potentially through effects on epithelial barrier function. Further investigation of IL-33 protective mechanisms would inform the development of novel therapeutic approaches.
Collapse
|
49
|
Akasaki S, Matsushita K, Kato Y, Fukuoka A, Iwasaki N, Nakahira M, Fujieda S, Yasuda K, Yoshimoto T. Murine allergic rhinitis and nasal Th2 activation are mediated via TSLP- and IL-33-signaling pathways. Int Immunol 2015; 28:65-76. [PMID: 26428949 DOI: 10.1093/intimm/dxv055] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2015] [Accepted: 09/25/2015] [Indexed: 12/27/2022] Open
Abstract
Thymic stromal lymphopoietin (TSLP) and IL-33 are epithelium-derived proallergic cytokines that contribute to allergic diseases. Although the involvement of TSLP in allergic rhinitis (AR) is suggested, the exact role of TSLP in AR is poorly understood. Furthermore, the relative contribution of TSLP and IL-33 in nasal allergic responses has not been described. In this study, we examined the roles of TSLP and IL-33 in AR by analyzing acute and chronic AR models. Acute AR mice were intraperitoneally immunized with ragweed, then intranasally challenged with ragweed pollen for four consecutive days. Chronic AR mice were nasally administrated ragweed pollen on consecutive days for 3 weeks. In both models, TSLP receptor (TSLPR)-deficient mice showed defective sneezing responses and reduced serum ragweed-specific IgE levels compared with wild-type (WT) mice. Analyses of bone-marrow chimeric mice demonstrated that hematopoietic cells were responsible for defective sneezing in TSLPR-deficient mice. In addition, FcεRI(+)-cell-specific TSLPR-deficient mice showed partial but significant reduction in sneezing responses. Of note, Th2 activation and nasal eosinophilia were comparable between WT and TSLPR-deficient mice. ST2- and IL-33-deficient mice showed defective Th2 activation and nasal eosinophilia to acute, but not chronic, ragweed exposure. TSLPR and ST2 double-deficient mice showed defective Th2 activation and nasal eosinophilia even after chronic ragweed exposure. These results demonstrate that TSLPR signaling is critical for the early phase response of AR by controlling the IgE-mast-cell/basophil pathway. The IL-33/ST2 pathway is central to nasal Th2 activation during acute allergen exposure, but both TSLPR and ST2 contribute to Th2 responses in chronically allergen-exposed mice.
Collapse
Affiliation(s)
- Shoko Akasaki
- Laboratory of Allergic Diseases, Institute for Advanced Medical Sciences, Hyogo College of Medicine, Nishinomiya, Hyogo 663-8501, Japan
| | - Kazufumi Matsushita
- Laboratory of Allergic Diseases, Institute for Advanced Medical Sciences, Hyogo College of Medicine, Nishinomiya, Hyogo 663-8501, Japan
| | - Yukinori Kato
- Laboratory of Allergic Diseases, Institute for Advanced Medical Sciences, Hyogo College of Medicine, Nishinomiya, Hyogo 663-8501, Japan Department of Otorhinolaryngology-Head and Neck Surgery, Faculty of Medical Science, University of Fukui, Fukui 910-1193, Japan
| | - Ayumi Fukuoka
- Laboratory of Allergic Diseases, Institute for Advanced Medical Sciences, Hyogo College of Medicine, Nishinomiya, Hyogo 663-8501, Japan
| | - Naruhito Iwasaki
- Laboratory of Allergic Diseases, Institute for Advanced Medical Sciences, Hyogo College of Medicine, Nishinomiya, Hyogo 663-8501, Japan Department of Otorhinolaryngology, Shiga University of Medical Science, Otsu, Shiga 520-2121, Japan
| | - Masakiyo Nakahira
- Department of Immunology, Hyogo College of Medicine, Nishinomiya, Hyogo 663-8501, Japan
| | - Shigeharu Fujieda
- Department of Otorhinolaryngology-Head and Neck Surgery, Faculty of Medical Science, University of Fukui, Fukui 910-1193, Japan
| | - Koubun Yasuda
- Department of Immunology, Hyogo College of Medicine, Nishinomiya, Hyogo 663-8501, Japan
| | - Tomohiro Yoshimoto
- Laboratory of Allergic Diseases, Institute for Advanced Medical Sciences, Hyogo College of Medicine, Nishinomiya, Hyogo 663-8501, Japan Department of Immunology, Hyogo College of Medicine, Nishinomiya, Hyogo 663-8501, Japan
| |
Collapse
|
50
|
Molofsky AB, Savage AK, Locksley RM. Interleukin-33 in Tissue Homeostasis, Injury, and Inflammation. Immunity 2015; 42:1005-19. [PMID: 26084021 DOI: 10.1016/j.immuni.2015.06.006] [Citation(s) in RCA: 487] [Impact Index Per Article: 48.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2015] [Indexed: 12/12/2022]
Abstract
Interleukin-33 (IL-33) is a nuclear-associated cytokine of the IL-1 family originally described as a potent inducer of allergic type 2 immunity. IL-33 signals via the receptor ST2, which is highly expressed on group 2 innate lymphoid cells (ILC2s) and T helper 2 (Th2) cells, thus underpinning its association with helminth infection and allergic pathology. Recent studies have revealed ST2 expression on subsets of regulatory T cells, and for a role for IL-33 in tissue homeostasis and repair that suggests previously unrecognized interactions within these cellular networks. IL-33 can participate in pathologic fibrotic reactions, or, in the setting of microbial invasion, can cooperate with inflammatory cytokines to promote responses by cytotoxic NK cells, Th1 cells, and CD8(+) T cells. Here, we highlight the regulation and function of IL-33 and ST2 and review their roles in homeostasis, damage, and inflammation, suggesting a conceptual framework for future studies.
Collapse
Affiliation(s)
- Ari B Molofsky
- Department of Microbiology & Immunology, University of California, San Francisco, 94143-0795, USA; Department of Laboratory Medicine, University of California, San Francisco, 94143-0795, USA
| | - Adam K Savage
- Howard Hughes Medical Institute, University of California, San Francisco, 94143-0795, USA; Department of Microbiology & Immunology, University of California, San Francisco, 94143-0795, USA
| | - Richard M Locksley
- Howard Hughes Medical Institute, University of California, San Francisco, 94143-0795, USA; Department of Medicine, University of California, San Francisco, 94143-0795, USA; Department of Microbiology & Immunology, University of California, San Francisco, 94143-0795, USA.
| |
Collapse
|