1
|
Bento LC, Bacal NS, Marti LC. Overview of the development, characterization, and function of human types 1, 2, and 3 innate lymphoid cells. EINSTEIN-SAO PAULO 2024; 22:eRW1042. [PMID: 39630753 PMCID: PMC11634355 DOI: 10.31744/einstein_journal/2024rw1042] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2024] [Accepted: 07/31/2024] [Indexed: 12/07/2024] Open
Abstract
Hematopoiesis is characterized by the differentiation and maturation of multipotent stem cells into hematopoietic cells. Common lymphoid progenitor cells differentiate into B and T lymphocytes; natural killer cells can also originate from common lymphoid progenitors. In recent years, a cellular subtype of lymphocytes, called innate lymphocytes, has been described. Innate lymphoid cells (ILCs) play an important effector and regulatory role in innate immunity, and similar to natural killer cells, depend on the γc and Id2 chains for their development. These cells are divided into three main subtypes according to their characteristics, namely type 1 innate lymphocytes (ILC1), type 2 (ILC2), and type 3 (ILC3); the production of cytokines and transcription factors is essential for this classification. Furthermore, these cells have high plasticity, which allows them to change their phenotype in response to the environment. ILCs have recently been characterized further and emerged as a family of effectors and regulators of innate immune responses. Uncontrolled activation of these cells can contribute to inflammatory, autoimmune diseases and cancer. The current review aimed to describe their main characteristics, immunophenotypes, and plasticity, and based on the existing literature, suggested a phenotypic analysis to differentiate innate lymphocytes from natural killer cells, and across the subsets.
Collapse
Affiliation(s)
- Laiz Cameirão Bento
- Hospital Israelita Albert EinsteinClinical Pathology LaboratorySão PauloSPBrazilClinical Pathology Laboratory, Hospital Israelita Albert Einstein, São Paulo, SP, Brazil.
| | - Nydia Strachman Bacal
- Hospital Israelita Albert EinsteinClinical Pathology LaboratorySão PauloSPBrazilClinical Pathology Laboratory, Hospital Israelita Albert Einstein, São Paulo, SP, Brazil.
| | - Luciana Cavalheiro Marti
- Hospital Israelita Albert EinsteinExperimental Biology Laboratory Prof. Dr Geraldo Antonio de Medeiros NetoSão PauloSPBrazilExperimental Biology Laboratory Prof. Dr Geraldo Antonio de Medeiros Neto, Hospital Israelita Albert Einstein, São Paulo, SP, Brazil.
| |
Collapse
|
2
|
Tian C, Liu Q, Zhang X, Li Z. Blocking group 2 innate lymphoid cell activation and macrophage M2 polarization: potential therapeutic mechanisms in ovalbumin-induced allergic asthma by calycosin. BMC Pharmacol Toxicol 2024; 25:30. [PMID: 38650035 PMCID: PMC11036756 DOI: 10.1186/s40360-024-00751-9] [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: 06/28/2023] [Accepted: 04/10/2024] [Indexed: 04/25/2024] Open
Abstract
BACKGROUND Calycosin, a flavonoid compound extracted from Astragalus membranaceus, has shown anti-asthma benefits in house dust mite-induced asthma. Recent studies have suggested that innate-type cells, including group 2 innate lymphoid cells (ILC2s) and macrophages, serve as incentives for type 2 immunity and targets for drug development in asthma. This work focuses on the effects of calycosin on the dysregulated ILC2s and macrophages in allergic asthma. METHODS In vivo, the asthmatic mouse model was established with ovalbumin (OVA) sensitization and challenge, and calycosin was intraperitoneally administered at doses of 20 and 40 mg/kg. In vivo, mouse primary ILC2s were stimulated with interleukin (IL)-33 and mouse RAW264.7 macrophages were stimulated with IL-4 and IL-13 to establish the cell models. Cells were treated with calycosin at doses of 5 and 10 µM. RESULTS In vivo, we observed significantly reduced numbers of eosinophils, neutrophils, monocyte macrophages and lymphocytes in the bronchoalveolar lavage fluid (BALF) of OVA-exposed mice with 40 mg/kg calycosin. Histopathological assessment showed that calycosin inhibited the airway inflammation and remodeling caused by OVA. Calycosin markedly decreased the up-regulated IL-4, IL-5, IL-13, IL-33, and suppression tumorigenicity 2 (ST2) induced by OVA in BALF and/or lung tissues of asthmatic mice. Calycosin repressed the augment of arginase 1 (ARG1), IL-10, chitinase-like 3 (YM1) and mannose receptor C-type 1 (MRC1) levels in the lung tissues of asthmatic mice. In vivo, calycosin inhibited the IL-33-induced activation as well as the increase of IL-4, IL-5, IL-13 and ST2 in ILC2s. Calycosin also repressed the increase of ARG1, IL-10, YM1 and MRC1 induced by IL-4 and IL-13 in RAW264.7 macrophages. In addition, we found that these changes were more significant in 40 mg/kg calycosin treatment than 20 mg/kg calycosin. CONCLUSIONS Collectively, this study showed that calycosin might attenuate OVA-induced airway inflammation and remodeling in asthmatic mice via preventing ILC2 activation and macrophage M2 polarization. Our study might contribute to further study of asthmatic therapy.
Collapse
Affiliation(s)
- Chunyan Tian
- Department of Respiratory Medicine, First Affiliated Hospital, Heilongjiang University of Chinese Medicine, Harbin, China
- Department of Graduate, Heilongjiang University of Chinese Medicine, Harbin, China
| | - Qi Liu
- Department of Respiratory Medicine, First Affiliated Hospital, Heilongjiang University of Chinese Medicine, Harbin, China
| | - Xiaoyu Zhang
- Department of Respiratory Medicine, First Affiliated Hospital, Heilongjiang University of Chinese Medicine, Harbin, China
| | - Zhuying Li
- Department of Respiratory Medicine, First Affiliated Hospital, Heilongjiang University of Chinese Medicine, Harbin, China.
| |
Collapse
|
3
|
Kim J, Ham J, Kang HR, Bae YS, Kim T, Kim HY. JAK3 inhibitor suppresses multipotent ILC2s and attenuates steroid-resistant asthma. SCIENCE ADVANCES 2023; 9:eadi3770. [PMID: 38117887 PMCID: PMC10732531 DOI: 10.1126/sciadv.adi3770] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/21/2023] [Accepted: 11/17/2023] [Indexed: 12/22/2023]
Abstract
Steroids are the standard treatment for allergic airway inflammation in asthma, but steroid-refractory asthma poses a challenge. Group 2 innate lymphoid cells (ILC2s), such as T helper 2 (TH2) cells, produce key asthma-related type 2 cytokines. Recent insights from mouse and human studies indicate a potential connection between ILC2s and steroid-resistant asthma. Here, we highlight that lung ILC2s, rather than TH2 cells, can develop steroid resistance, allowing them to persist and maintain their disease-driving activity even during steroid treatment. The emergence of multipotent IL-5+IL-13+IL-17A+ ILC2s is associated with steroid-resistant ILC2s. The Janus kinase 3 (JAK3)/signal transducer and activator of transcription (STAT) 3, 5, and 6 pathways contribute to the acquisition of steroid-resistant ILC2s. The JAK3 inhibitor reduces ILC2 survival, proliferation, and cytokine production in vitro and ameliorates ILC2-driven Alternaria-induced asthma. Furthermore, combining a JAK3 inhibitor with steroids results in the inhibition of steroid-resistant asthma. These findings suggest a potential therapeutic approach for addressing this challenging condition in chronic asthma.
Collapse
Affiliation(s)
- Jihyun Kim
- Laboratory of Mucosal Immunology, Department of Biomedical Sciences, Seoul National University College of Medicine, Seoul, South Korea
- Institute of Allergy and Clinical Immunology, Seoul National University Medical Research Center, Seoul, South Korea
| | - Jongho Ham
- Laboratory of Mucosal Immunology, Department of Biomedical Sciences, Seoul National University College of Medicine, Seoul, South Korea
- Institute of Allergy and Clinical Immunology, Seoul National University Medical Research Center, Seoul, South Korea
- Department of Biological Sciences, SRC Center for Immune Research on Non-lymphoid Organs, Sungkyunkwan University, Suwon, South Korea
| | - Hye Ryun Kang
- Institute of Allergy and Clinical Immunology, Seoul National University Medical Research Center, Seoul, South Korea
- Department of Internal Medicine, Seoul National University Hospital, Seoul, South Korea
| | - Yong-Soo Bae
- Department of Biological Sciences, SRC Center for Immune Research on Non-lymphoid Organs, Sungkyunkwan University, Suwon, South Korea
- Department of Biological Sciences, Sungkyunkwan University, Suwon, South Korea
| | - TaeSoo Kim
- Department of Life Science, Multitasking Macrophage Research Center, Ewha Womans University, Seoul, South Korea
| | - Hye Young Kim
- Laboratory of Mucosal Immunology, Department of Biomedical Sciences, Seoul National University College of Medicine, Seoul, South Korea
- Institute of Allergy and Clinical Immunology, Seoul National University Medical Research Center, Seoul, South Korea
- Department of Biological Sciences, SRC Center for Immune Research on Non-lymphoid Organs, Sungkyunkwan University, Suwon, South Korea
| |
Collapse
|
4
|
Xu S, Zhang Y, Liu X, Liu H, Zou X, Zhang L, Wang J, Zhang Z, Xu X, Li M, Li K, Shi S, Zhang Y, Miao Z, Zha J, Yu Y. Nr4a1 marks a distinctive ILC2 activation subset in the mouse inflammatory lung. BMC Biol 2023; 21:218. [PMID: 37833706 PMCID: PMC10576290 DOI: 10.1186/s12915-023-01690-3] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2022] [Accepted: 08/25/2023] [Indexed: 10/15/2023] Open
Abstract
BACKGROUND Group 2 innate lymphoid cells (ILC2s) are critical sources of type 2 cytokines and represent one of the major tissue-resident lymphoid cells in the mouse lung. However, the molecular mechanisms underlying ILC2 activation under challenges are not fully understood. RESULTS Here, using single-cell transcriptomics, genetic reporters, and gene knockouts, we identify four ILC2 subsets, including two non-activation subsets and two activation subsets, in the mouse acute inflammatory lung. Of note, a distinct activation subset, marked by the transcription factor Nr4a1, paradoxically expresses both tissue-resident memory T cell (Trm), and effector/central memory T cell (Tem/Tcm) signature genes, as well as higher scores of proliferation, activation, and wound healing, all driven by its particular regulons. Furthermore, we demonstrate that the Nr4a1+ILC2s are restrained from activating by the programmed cell death protein-1 (PD-1), which negatively modulates their activation-related regulons. PD-1 deficiency places the non-activation ILC2s in a state that is prone to activation, resulting in Nr4a1+ILC2 differentiation through different activation trajectories. Loss of PD-1 also leads to the expansion of Nr4a1+ILC2s by the increase of their proliferation ability. CONCLUSIONS The findings show that activated ILC2s are a heterogenous population encompassing distinct subsets that have different propensities, and therefore provide an opportunity to explore PD-1's role in modulating the activity of ILC2s for disease prevention and therapy.
Collapse
Affiliation(s)
- Shasha Xu
- Department of Hematology, Tongji Hospital, Frontier Science Center for Stem Cell Research, School of Life Sciences and Technology, Tongji University, 1239 Siping Road, Shanghai, 200092, China
| | - Yu Zhang
- Department of Hematology, Tongji Hospital, Frontier Science Center for Stem Cell Research, School of Life Sciences and Technology, Tongji University, 1239 Siping Road, Shanghai, 200092, China
| | - Xingjie Liu
- Department of Hematology, Tongji Hospital, Frontier Science Center for Stem Cell Research, School of Life Sciences and Technology, Tongji University, 1239 Siping Road, Shanghai, 200092, China
| | - Huisheng Liu
- Department of Hematology, Tongji Hospital, Frontier Science Center for Stem Cell Research, School of Life Sciences and Technology, Tongji University, 1239 Siping Road, Shanghai, 200092, China
| | - Xinya Zou
- Department of Hematology, Tongji Hospital, Frontier Science Center for Stem Cell Research, School of Life Sciences and Technology, Tongji University, 1239 Siping Road, Shanghai, 200092, China
| | - Linlin Zhang
- Department of Hematology, Tongji Hospital, Frontier Science Center for Stem Cell Research, School of Life Sciences and Technology, Tongji University, 1239 Siping Road, Shanghai, 200092, China
| | - Jing Wang
- Department of Hematology, Tongji Hospital, Frontier Science Center for Stem Cell Research, School of Life Sciences and Technology, Tongji University, 1239 Siping Road, Shanghai, 200092, China
| | - Zhiwei Zhang
- Department of Hematology, Tongji Hospital, Frontier Science Center for Stem Cell Research, School of Life Sciences and Technology, Tongji University, 1239 Siping Road, Shanghai, 200092, China
| | - Xiang Xu
- Department of Hematology, Tongji Hospital, Frontier Science Center for Stem Cell Research, School of Life Sciences and Technology, Tongji University, 1239 Siping Road, Shanghai, 200092, China
| | - Mingxia Li
- Department of Hematology, Tongji Hospital, Frontier Science Center for Stem Cell Research, School of Life Sciences and Technology, Tongji University, 1239 Siping Road, Shanghai, 200092, China
| | - Kairui Li
- Department of Hematology, Tongji Hospital, Frontier Science Center for Stem Cell Research, School of Life Sciences and Technology, Tongji University, 1239 Siping Road, Shanghai, 200092, China
| | - Shuyue Shi
- Department of Hematology, Tongji Hospital, Frontier Science Center for Stem Cell Research, School of Life Sciences and Technology, Tongji University, 1239 Siping Road, Shanghai, 200092, China
| | - Ying Zhang
- Department of Hematology, Tongji Hospital, Frontier Science Center for Stem Cell Research, School of Life Sciences and Technology, Tongji University, 1239 Siping Road, Shanghai, 200092, China
| | - Zhichao Miao
- Translational Research Institute of Brain and Brain-Like Intelligence and Department of Anesthesiology, Shanghai Fourth People's Hospital Affiliated to Tongji University School of Medicine, Shanghai, 200081, China
- GMU-GIBH Joint School of Life Sciences, The Guangdong-Hong Kong-Macau Joint Laboratory for Cell Fate Regulation and Diseases, Guangzhou Laboratory, Guangzhou Medical University, Guangzhou, China
| | - Jie Zha
- Department of Hematology, The First Affiliated Hospital of Xiamen University and Institute of Hematology, School of Medicine, Xiamen University, Xiamen, China.
- Key Laboratory of Xiamen for Diagnosis and Treatment of Hematological Malignancy, Xiamen, China.
| | - Yong Yu
- Department of Hematology, Tongji Hospital, Frontier Science Center for Stem Cell Research, School of Life Sciences and Technology, Tongji University, 1239 Siping Road, Shanghai, 200092, China.
- Wellcome Trust Sanger Institute, Hinxton, Cambridge, CB10 1HH, UK.
| |
Collapse
|
5
|
Kim S, Cho S, Kim JH. CD1-mediated immune responses in mucosal tissues: molecular mechanisms underlying lipid antigen presentation system. Exp Mol Med 2023; 55:1858-1871. [PMID: 37696897 PMCID: PMC10545705 DOI: 10.1038/s12276-023-01053-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2023] [Revised: 05/02/2023] [Accepted: 05/07/2023] [Indexed: 09/13/2023] Open
Abstract
The cluster of differentiation 1 (CD1) molecule differs from major histocompatibility complex class I and II because it presents glycolipid/lipid antigens. Moreover, the CD1-restricted T cells that recognize these self and foreign antigens participate in both innate and adaptive immune responses. CD1s are constitutively expressed by professional and nonprofessional antigen-presenting cells in mucosal tissues, namely, the skin, lung, and intestine. This suggests that CD1-reactive T cells are involved in the immune responses of these tissues. Indeed, evidence suggests that these cells play important roles in diverse diseases, such as inflammation, autoimmune disease, and infection. Recent studies elucidating the molecular mechanisms by which CD1 presents lipid antigens suggest that defects in these mechanisms could contribute to the activities of CD1-reactive T cells. Thus, improving our understanding of these mechanisms could lead to new and effective therapeutic approaches to CD1-associated diseases. In this review, we discuss the CD1-mediated antigen presentation system and its roles in mucosal tissue immunity.
Collapse
Affiliation(s)
- Seohyun Kim
- Department of Biotechnology, College of Life Sciences and Biotechnology, Korea University, Seoul, 02841, Republic of Korea
| | - Sumin Cho
- Department of Biotechnology, College of Life Sciences and Biotechnology, Korea University, Seoul, 02841, Republic of Korea
| | - Ji Hyung Kim
- Department of Biotechnology, College of Life Sciences and Biotechnology, Korea University, Seoul, 02841, Republic of Korea.
| |
Collapse
|
6
|
Ike E, Kawano T, Takahashi K, Miyasaka T, Takahashi T. Calcitonin Gene-Related peptide receptor antagonist suppresses allergic asthma responses via downregulation of group 2 innate lymphoid cells in mice. Int Immunopharmacol 2023; 122:110608. [PMID: 37441811 DOI: 10.1016/j.intimp.2023.110608] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2023] [Revised: 06/20/2023] [Accepted: 07/03/2023] [Indexed: 07/15/2023]
Abstract
Allergic asthma is caused by chronic inflammation and hyper-responsiveness of the airway and is thought to be mediated by adaptive T helper type 2 (Th2)-driven immunity. However, recent studies have demonstrated that neuropeptide calcitonin gene-related peptide (CGRP)-mediated activation of group 2 innate lymphoid cells (ILC2s) may contribute to the development of asthma pathogenesis. Here, we investigated the therapeutic effects of the systemic administration of rimegepant, a CGRP receptor antagonist, on allergic asthma. Hyperplasia of CGRP-immunoreactive pulmonary neuroendocrine cells (PNECs) was observed in ovalbumin (OVA)-induced asthmatic mice. Concomitant with this, we observed an increase in the content of total lung CGRP. Upon antigen challenge, the concentration of plasma CGRP was transiently upregulated, whereas CGRP immunoreactivity within PNECs was intensively downregulated, suggesting that PNECs were the most likely source of CGRP. When rimegepant was administered according to CGRP kinetics, it suppressed asthma phenotypes, including airway hyper-responsiveness, infiltration of inflammatory cells in bronchoalveolar lavage fluid (BALF), hyperplasia of mucus-producing cells, and production of the Th2 cytokine IL-5. Moreover, we observed a decrease in the number of ILC2s and their capacity for IL-5 release in the presence of IL-33 in rimegepant-treated mice. In the allergic asthma model, rimegepant suppressed the activation of ILC2s mediated by PNEC-derived CGRP and subsequently impaired adaptive Th2-driven immunity, which ameliorated asthmatic phenotypes. Thus, an anti-CGRP signal strategy to target ILC2 will be a novel and attractive approach for treating allergic asthma that is refractory to other treatments.
Collapse
Affiliation(s)
- Erina Ike
- Division of Pathophysiology, Department of Pharmaceutical Sciences, Faculty of Pharmaceutical Sciences, Tohoku Medical and Pharmaceutical University, 4-4-1 Komatsushima, Aoba-ku, Sendai, Miyagi 981-8558 Japan
| | - Tasuku Kawano
- Division of Pathophysiology, Department of Pharmaceutical Sciences, Faculty of Pharmaceutical Sciences, Tohoku Medical and Pharmaceutical University, 4-4-1 Komatsushima, Aoba-ku, Sendai, Miyagi 981-8558 Japan
| | - Kento Takahashi
- Division of Pathophysiology, Department of Pharmaceutical Sciences, Faculty of Pharmaceutical Sciences, Tohoku Medical and Pharmaceutical University, 4-4-1 Komatsushima, Aoba-ku, Sendai, Miyagi 981-8558 Japan
| | - Tomomitsu Miyasaka
- Division of Pathophysiology, Department of Pharmaceutical Sciences, Faculty of Pharmaceutical Sciences, Tohoku Medical and Pharmaceutical University, 4-4-1 Komatsushima, Aoba-ku, Sendai, Miyagi 981-8558 Japan
| | - Tomoko Takahashi
- Division of Pathophysiology, Department of Pharmaceutical Sciences, Faculty of Pharmaceutical Sciences, Tohoku Medical and Pharmaceutical University, 4-4-1 Komatsushima, Aoba-ku, Sendai, Miyagi 981-8558 Japan.
| |
Collapse
|
7
|
Shimizu Y, Horigane-Konakai Y, Ishii Y, Sugimoto C, Wakao H. Mucosal-associated invariant T cells repress group 2 innate lymphoid cells in Alternaria alternata-induced model of allergic airway inflammation. Front Immunol 2022; 13:1005226. [PMID: 36458017 PMCID: PMC9706205 DOI: 10.3389/fimmu.2022.1005226] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2022] [Accepted: 10/27/2022] [Indexed: 08/13/2023] Open
Abstract
Mucosal-associated invariant T (MAIT) cells, a blossoming member of the innate-like T cells, play a pivotal role in host defense through engaging the mucosal immunity. Although it has been suggested that MAIT cells are somehow implicated in the allergic airway inflammation mediated by group 2 innate lymphoid cells (ILC2s) such as asthma, the precise role(s) of MAIT cells in such inflammation has remained elusive. To explore the possible roles of MAIT cells in the inflammation, we examined whether MAIT cells suppressed the production of T helper (Th) 2 and inflammatory cytokines from ILC2s, and constrained the proliferation of ILC2s, both of which are prerequisite for airway inflammation. Given that laboratory mice are poor at MAIT cells, a novel mouse line rich in MAIT cells was used. We found that mice rich in MAIT cells showed alleviated airway inflammation as evidenced by reduced infiltration of the immune cells and hyperplasia in goblet cells in the lung concomitant with compromised production of Th2 and inflammatory cytokines, while wild type mice exhibited severe inflammation upon challenge with the fungal extracts. In vitro coculture experiments using purified ILC2s and MAIT cells unrevealed that cytokine-stimulated MAIT cells suppressed ILC2s to produce the cytokines as well as to proliferate most likely via production of IFN-γ. Furthermore, reconstitution of the allergic airway inflammation in the highly immunocompromised mice showed that ILC2-mediated inflammation was alleviated in mice that received MAIT cells along with ILC2s. We concluded that MAIT cells played a crucial role in suppressing the cytokine-producing capacity of ILC2s and ILC2 proliferation, that ultimately led to decrease in the allergic airway inflammation. The results open up a novel therapeutic horizon in ILC2-mediated inflammatory diseases by modulating MAIT cell activity.
Collapse
Affiliation(s)
- Yasuo Shimizu
- Department of Pulmonary Medicine and Clinical Immunology, Dokkyo Medical University, Mibu, Tochigi, Japan
- Respiratory Endoscopy Center, Dokkyo Medical University Hospital, Mibu, Tochigi, Japan
| | - Yukiko Horigane-Konakai
- Department of Pulmonary Medicine and Clinical Immunology, Dokkyo Medical University, Mibu, Tochigi, Japan
| | - Yoshii Ishii
- Department of Pulmonary Medicine and Clinical Immunology, Dokkyo Medical University, Mibu, Tochigi, Japan
| | - Chie Sugimoto
- Host Defense Division, Research Center for Advanced Medical Science, Dokkyo Medical University, Mibu, Tochigi, Japan
| | - Hiroshi Wakao
- Host Defense Division, Research Center for Advanced Medical Science, Dokkyo Medical University, Mibu, Tochigi, Japan
| |
Collapse
|
8
|
Emerging Effects of IL-33 on COVID-19. Int J Mol Sci 2022; 23:ijms232113656. [PMID: 36362440 PMCID: PMC9658128 DOI: 10.3390/ijms232113656] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2022] [Revised: 10/27/2022] [Accepted: 11/03/2022] [Indexed: 11/09/2022] Open
Abstract
Since the start of COVID-19 pandemic caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), more than 6 million people have lost their lives worldwide directly or indirectly. Despite intensified efforts to clarify the immunopathology of COVID-19, the key factors and processes that trigger an inflammatory storm and lead to severe clinical outcomes in patients remain unclear. As an inflammatory storm factor, IL-33 is an alarmin cytokine, which plays an important role in cell damage or infection. Recent studies have shown that serum IL-33 is upregulated in COVID-19 patients and is strongly associated with poor outcomes. Increased IL-33 levels in severe infections may result from an inflammatory storm caused by strong interactions between activated immune cells. However, the effects of IL-33 in COVID-19 and the underlying mechanisms remain to be fully elucidated. In this review, we systematically discuss the biological properties of IL-33 under pathophysiological conditions and its regulation of immune cells, including neutrophils, innate lymphocytes (ILCs), dendritic cells, macrophages, CD4+ T cells, Th17/Treg cells, and CD8+ T cells, in COVID-19 phagocytosis. The aim of this review is to explore the potential value of the IL-33/immune cell pathway as a new target for early diagnosis, monitoring of severe cases, and clinical treatment of COVID-19.
Collapse
|
9
|
Maggi E, Parronchi P, Azzarone BG, Moretta L. A pathogenic integrated view explaining the different endotypes of asthma and allergic disorders. Allergy 2022; 77:3267-3292. [PMID: 35842745 DOI: 10.1111/all.15445] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2022] [Revised: 07/08/2022] [Accepted: 07/13/2022] [Indexed: 01/28/2023]
Abstract
The inflammation of allergic diseases is characterized by a complex interaction between type 2 and type 3 immune responses, explaining clinical symptoms and histopathological patterns. Airborne stimuli activate the mucosal epithelium to release a number of molecules impacting the activity of resident immune and environmental cells. Signals from the mucosal barrier, regulatory cells, and the inflamed tissue are crucial conditions able to modify innate and adaptive effector cells providing the selective homing of eosinophils or neutrophils. The high plasticity of resident T- and innate lymphoid cells responding to external signals is the prerequisite to explain the multiplicity of endotypes of allergic diseases. This notion paved the way for the huge use of specific biologic drugs interfering with pathogenic mechanisms of inflammation. Based on the response of the epithelial barrier, the activity of resident regulatory cells, and functions of structural non-lymphoid environmental cells, this review proposes some immunopathogenic scenarios characterizing the principal endotypes which can be associated with a precise phenotype of asthma. Recent literature indicates that similar concepts can also be applied to the inflammation of other non-respiratory allergic disorders. The next challenges will consist in defining specific biomarker(s) of each endotype allowing for a quick diagnosis and the most effective personalized therapy.
Collapse
Affiliation(s)
- Enrico Maggi
- Department of Immunology, Bambino Gesù Children's Hospital, IRCCS, Rome, Italy
| | - Paola Parronchi
- Department of Clinical and Experimental Medicine, University of Florence, Florence, Italy
| | | | - Lorenzo Moretta
- Department of Immunology, Bambino Gesù Children's Hospital, IRCCS, Rome, Italy
| |
Collapse
|
10
|
Huang Z, Chu M, Chen X, Wang Z, Jiang L, Ma Y, Wang Y. Th2A cells: The pathogenic players in allergic diseases. Front Immunol 2022; 13:916778. [PMID: 36003397 PMCID: PMC9393262 DOI: 10.3389/fimmu.2022.916778] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2022] [Accepted: 07/12/2022] [Indexed: 11/13/2022] Open
Abstract
Proallergic type 2 helper T (Th2A) cells are a subset of memory Th2 cells confined to atopic individuals, and they include all the allergen-specific Th2 cells. Recently, many studies have shown that Th2A cells characterized by CD3+ CD4+ HPGDS+ CRTH2+ CD161high ST2high CD49dhigh CD27low play a crucial role in allergic diseases, such as atopic dermatitis (AD), food allergy (FA), allergic rhinitis (AR), asthma, and eosinophilic esophagitis (EoE). In this review, we summarize the discovery, biomarkers, and biological properties of Th2A cells to gain new insights into the pathogenesis of allergic diseases.
Collapse
Affiliation(s)
- Ziyu Huang
- Department of Immunology, School of Basic Medical Sciences, Peking University, NHC Key Laboratory of Medical Immunology (Peking University), Beijing, China
- Department of Clinical Medicine, Mudanjiang Medical University, Mudanjiang, China
| | - Ming Chu
- Department of Immunology, School of Basic Medical Sciences, Peking University, NHC Key Laboratory of Medical Immunology (Peking University), Beijing, China
| | - Xi Chen
- Department of Immunology, School of Basic Medical Sciences, Peking University, NHC Key Laboratory of Medical Immunology (Peking University), Beijing, China
| | - Ziyuan Wang
- Department of Immunology, School of Basic Medical Sciences, Peking University, NHC Key Laboratory of Medical Immunology (Peking University), Beijing, China
| | - Lin Jiang
- Department of Immunology, School of Basic Medical Sciences, Peking University, NHC Key Laboratory of Medical Immunology (Peking University), Beijing, China
| | - Yinchao Ma
- Department of Immunology, School of Basic Medical Sciences, Peking University, NHC Key Laboratory of Medical Immunology (Peking University), Beijing, China
| | - Yuedan Wang
- Department of Immunology, School of Basic Medical Sciences, Peking University, NHC Key Laboratory of Medical Immunology (Peking University), Beijing, China
| |
Collapse
|
11
|
Nong C, Guan P, Li L, Zhang H, Hu H. Tumor immunotherapy: Mechanisms and clinical applications. MEDCOMM – ONCOLOGY 2022. [DOI: 10.1002/mog2.8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Affiliation(s)
- Cheng Nong
- Center for Immunology and Hematology, National Clinical Research Center for Geriatrics State Key Laboratory of Biotherapy, West China Hospital Sichuan University Chengdu China
| | - Pengbo Guan
- Center for Immunology and Hematology, National Clinical Research Center for Geriatrics State Key Laboratory of Biotherapy, West China Hospital Sichuan University Chengdu China
| | - Li Li
- Center for Immunology and Hematology, National Clinical Research Center for Geriatrics State Key Laboratory of Biotherapy, West China Hospital Sichuan University Chengdu China
| | - Huiyuan Zhang
- Center for Immunology and Hematology, National Clinical Research Center for Geriatrics State Key Laboratory of Biotherapy, West China Hospital Sichuan University Chengdu China
| | - Hongbo Hu
- Center for Immunology and Hematology, National Clinical Research Center for Geriatrics State Key Laboratory of Biotherapy, West China Hospital Sichuan University Chengdu China
- Chongqing International Institution for Immunology Chongqing China
| |
Collapse
|
12
|
Mathä L, Takei F, Martinez-Gonzalez I. Tissue Resident and Migratory Group 2 Innate Lymphoid Cells. Front Immunol 2022; 13:877005. [PMID: 35572538 PMCID: PMC9099002 DOI: 10.3389/fimmu.2022.877005] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2022] [Accepted: 04/08/2022] [Indexed: 11/13/2022] Open
Abstract
Group 2 innate lymphoid cells (ILC2s) are present in both mouse and human mucosal and non-mucosal tissues and implicated in initiating type 2 inflammation. ILC2s are considered to be tissue resident cells that develop in the perinatal period and persist throughout life with minimal turning over in adulthood. However, recent studies in animal models have shown their ability to circulate between different organs during inflammation and their potential functions in the destined organs, suggesting their roles in mediating multiple type 2 diseases. Here, we review recent findings on ILC2 migration, including migration within, into and out of tissues during inflammation.
Collapse
Affiliation(s)
- Laura Mathä
- Department of Microbiology, Tumor and Cell Biology, Karolinska Institutet, Solna, Sweden
| | - Fumio Takei
- Terry Fox Laboratory, British Columbia Cancer, Vancouver, BC, Canada.,Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, BC, Canada
| | | |
Collapse
|
13
|
Hasegawa T, Oka T, Demehri S. Alarmin Cytokines as Central Regulators of Cutaneous Immunity. Front Immunol 2022; 13:876515. [PMID: 35432341 PMCID: PMC9005840 DOI: 10.3389/fimmu.2022.876515] [Citation(s) in RCA: 30] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2022] [Accepted: 03/08/2022] [Indexed: 12/13/2022] Open
Abstract
Skin acts as the primary interface between the body and the environment. The skin immune system is composed of a complex network of immune cells and factors that provide the first line of defense against microbial pathogens and environmental insults. Alarmin cytokines mediate an intricate intercellular communication between keratinocytes and immune cells to regulate cutaneous immune responses. Proper functions of the type 2 alarmin cytokines, thymic stromal lymphopoietin (TSLP), interleukin (IL)-25, and IL-33, are paramount to the maintenance of skin homeostasis, and their dysregulation is commonly associated with allergic inflammation. In this review, we discuss recent findings on the complex regulatory network of type 2 alarmin cytokines that control skin immunity and highlight the mechanisms by which these cytokines regulate skin immune responses in host defense, chronic inflammation, and cancer.
Collapse
Affiliation(s)
| | - Tomonori Oka
- Center for Cancer Immunology and Cutaneous Biology Research Center, Department of Dermatology and Center for Cancer Research, Massachusetts General Hospital and Harvard Medical School, Boston, MA, United States
| | - Shadmehr Demehri
- Center for Cancer Immunology and Cutaneous Biology Research Center, Department of Dermatology and Center for Cancer Research, Massachusetts General Hospital and Harvard Medical School, Boston, MA, United States
| |
Collapse
|
14
|
Onoda A, Okamoto S, Shimizu R, El-Sayed YS, Watanabe S, Ogawa S, Abe R, Kamimura M, Soga K, Tachibana K, Takeda K, Umezawa M. Effect of Carbon Black Nanoparticle on Neonatal Lymphoid Tissues Depending on the Gestational Period of Exposure in Mice. FRONTIERS IN TOXICOLOGY 2022; 3:700392. [PMID: 35295157 PMCID: PMC8915855 DOI: 10.3389/ftox.2021.700392] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2021] [Accepted: 07/27/2021] [Indexed: 12/16/2022] Open
Abstract
Introduction: Particulate air pollution, containing nanoparticles, enhances the risk of pediatric allergic diseases that is potentially associated with disruption of neonatal immune system. Previous studies have revealed that maternal exposure to carbon black nanoparticles (CB-NP) disturbs the development of the lymphoid tissues in newborns. Interestingly, the CB-NP-induced immune profiles were observed to be different depending on the gestational period of exposure. It is important to identify the critical exposure period to prevent toxic effects of nanoparticles on the development of the immune system. Therefore, the present study was aimed to investigate the effect of CB-NP on the development of neonatal lymphoid tissues in mice, depending on the gestational period of exposure. Methods: Pregnant ICR mice were treated with a suspension of CB-NP (95 μg/kg body weight) by intranasal instillation; the suspension was administered twice during each gestational period as follows: the pre-implantation period (gestational days 4 and 5), organogenesis period (gestational days 8 and 9), and fetal developmental period (gestational days 15 and 16). The spleen and thymus were collected from offspring mice at 1, 3, and 5-days post-partum. Splenocyte and thymocyte phenotypes were examined by flow cytometry. Gene expression in the spleen was examined by quantitative reverse transcription-polymerase chain reaction. Results: The numbers of total splenocytes and splenic CD3−B220− phenotype (non-T/non-B lymphocytes) in offspring on postnatal day 5 were significantly increased after exposure to CB-NP during the organogenesis period compared with other gestational periods of exposure and control (no exposure). In contrast, expression levels of mRNA associated with chemotaxis and differentiation of immune cells in the spleen were not affected by CB-NP exposure during any gestational period. Conclusion: The organogenesis period was the most susceptible period to CB-NP exposure with respect to lymphoid tissue development. Moreover, the findings of the present and previous studies suggested that long-term exposure to CB-NP across multiple gestational periods including the organogenesis period, rather than acute exposure only organogenesis period, may more severely affect the development of the immune system.
Collapse
Affiliation(s)
- Atsuto Onoda
- The Center for Environmental Health Science for the Next Generation, Research Institute for Science and Technology, Tokyo University of Science, Noda, Japan.,Faculty of Pharmaceutical Sciences, Tokyo University of Science, Noda, Japan.,Faculty of Pharmaceutical Sciences, Sanyo-Onoda City University, Sanyoonoda, Japan
| | - Saki Okamoto
- Faculty of Pharmaceutical Sciences, Tokyo University of Science, Noda, Japan
| | - Ryuhei Shimizu
- Faculty of Pharmaceutical Sciences, Tokyo University of Science, Noda, Japan
| | - Yasser S El-Sayed
- Faculty of Veterinary Medicine, Damanhour University, Damanhour, Egypt
| | - Shiho Watanabe
- Research Institute for Biomedical Sciences, Tokyo University of Science, Noda, Japan
| | - Shuhei Ogawa
- The Center for Environmental Health Science for the Next Generation, Research Institute for Science and Technology, Tokyo University of Science, Noda, Japan.,Research Institute for Biomedical Sciences, Tokyo University of Science, Noda, Japan
| | - Ryo Abe
- Research Institute for Biomedical Sciences, Tokyo University of Science, Noda, Japan.,Advanced Comprehensive Research Center, Teikyo University, Hachioji, Japan
| | - Masao Kamimura
- Department of Materials Science and Technology, Faculty of Advanced Engineering, Tokyo University of Science, Katsushika, Japan
| | - Kohei Soga
- Department of Materials Science and Technology, Faculty of Advanced Engineering, Tokyo University of Science, Katsushika, Japan
| | - Ken Tachibana
- The Center for Environmental Health Science for the Next Generation, Research Institute for Science and Technology, Tokyo University of Science, Noda, Japan.,Faculty of Pharmaceutical Sciences, Tokyo University of Science, Noda, Japan.,Faculty of Pharmaceutical Sciences, Sanyo-Onoda City University, Sanyoonoda, Japan
| | - Ken Takeda
- The Center for Environmental Health Science for the Next Generation, Research Institute for Science and Technology, Tokyo University of Science, Noda, Japan.,Faculty of Pharmaceutical Sciences, Tokyo University of Science, Noda, Japan.,Faculty of Pharmaceutical Sciences, Sanyo-Onoda City University, Sanyoonoda, Japan
| | - Masakazu Umezawa
- The Center for Environmental Health Science for the Next Generation, Research Institute for Science and Technology, Tokyo University of Science, Noda, Japan.,Faculty of Pharmaceutical Sciences, Tokyo University of Science, Noda, Japan.,Department of Materials Science and Technology, Faculty of Advanced Engineering, Tokyo University of Science, Katsushika, Japan
| |
Collapse
|
15
|
Huang M, Wu J, Dong J. Modified BuShenYiQi formula alleviates experimental allergic asthma in mice by negative regulation of type 2 innate lymphoid cells and CD4 + type 9 helper T cells and the VIP-VPAC2 signalling pathway. PHARMACEUTICAL BIOLOGY 2021; 59:1216-1232. [PMID: 34493162 PMCID: PMC8425750 DOI: 10.1080/13880209.2021.1970198] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/04/2021] [Revised: 06/23/2021] [Accepted: 08/14/2021] [Indexed: 06/01/2023]
Abstract
CONTEXT Modified BuShenYiQi formula (M-BYF) is derived from BuShenYiQi formula, used for the treatment of allergic asthma. The exact effect and mechanism of M-BYF on the improvement of asthma remain unclear. OBJECTIVE We investigated the mechanism underlying the therapeutic effect of M-BYF on allergic asthma. MATERIALS AND METHODS The asthma model was established in female BALB/c mice that were sensitized and challenged with ovalbumin (OVA). Mice in the treated groups were orally treated once a day with M-BYF (7, 14 and 28 g/kg/d) or dexamethasone before OVA challenge. Control and Model group received saline. Pathophysiological abnormalities and percentages of lung type 2 innate lymphoid cells (ILC2s) and Th9 cells were measured. Expression levels of type 2 cytokines and transcription factors required for these cells function and differentiation were analysed. Expression of vasoactive intestinal polypeptide (VIP)-VPAC2 signalling pathway-related proteins, and percentages of VIP expressing (VIP+) cells and VPAC2, CD90 co-expressing (VPAC2+CD90+) cells were detected. RESULTS M-BYF alleviated airway hyperresponsiveness, inflammation, mucus hypersecretion and collagen deposition in asthmatic mice. M-BYF down-regulated percentages of ILC2s and Th9 cells with lower expression of GATA3, PU.1 and IRF4, reduced IL-5, IL-13, IL-9 and VIP production. The decrease in the expression of VIP-VPAC2 signalling pathway and percentages of VIP+ cells, VPAC2+CD90+ cells were observed after M-BYF treatment. The LD50 value of M-BYF was higher than 90 g/kg. DISCUSSION AND CONCLUSIONS M-BYF alleviated experimental asthma by negatively regulating ILC2s and Th9 cells and the VIP-VPAC2 signalling pathway. These findings provide the theoretical basis for future research of M-BYF in asthma patient population.
Collapse
Affiliation(s)
- Muhua Huang
- Department of Integrative Medicine, Huashan Hospital, Fudan University, Shanghai, China
| | - Jinfeng Wu
- Department of Integrative Medicine, Huashan Hospital, Fudan University, Shanghai, China
| | - Jingcheng Dong
- Department of Integrative Medicine, Huashan Hospital, Fudan University, Shanghai, China
- Institute of Integrative Medicine, Fudan University, Shanghai, China
| |
Collapse
|
16
|
Xu X, Ye L, Zhang Q, Shen H, Li S, Zhang X, Ye M, Liang T. Group-2 Innate Lymphoid Cells Promote HCC Progression Through CXCL2-Neutrophil-Induced Immunosuppression. Hepatology 2021; 74:2526-2543. [PMID: 33829508 PMCID: PMC8597094 DOI: 10.1002/hep.31855] [Citation(s) in RCA: 51] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/20/2020] [Revised: 02/27/2021] [Accepted: 03/28/2021] [Indexed: 12/28/2022]
Abstract
BACKGROUND AND AIMS Due to their inherent characteristics, the function of group-2 innate lymphoid cells (ILC2s) varies in a context-dependent manner. ILC2s are involved in certain liver diseases; however, their involvement in HCC is unknown. In the present study, we assessed the role of an HCC-derived ILC2 population in tumor progression. APPROACH AND RESULTS Through FACS and single-cell RNA sequencing, we discovered that ILC2s were highly enriched in human HCC and correlated significantly with tumor recurrence and worse progression-free survival as well as overall survival in patients. Mass cytometry identified a subset of HCC-derived ILC2s that had lost the expression of killer cell lectin-like receptor subfamily G, member 1 (KLRG1). Distinct from their circulating counterparts, these hepatic ILC2s highly expressed CD69 and an array of tissue resident-related genes. Furthermore, reduction of E-cadherin in tumor cells caused the loss of KLRG1 expression in ILC2s, leading to their increased proliferation and subsequent accumulation in HCC sites. The KLRG1- ILC2 subset showed elevated production of chemotaxis factors, including C-X-C motif chemokine (C-X-C motif) ligand (CXCL)-2 and CXCL8, which in turn recruited neutrophils to form an immunosuppressive microenvironment, leading to tumor progression. Accordingly, restoring KLRG1 in ILC2s, inhibiting CXCL2 in ILC2s, or depleting neutrophils inhibited tumor progression in a murine HCC model. CONCLUSIONS We identified HCC-associated ILC2s as an immune regulatory cell type that promotes tumor development, suggesting that targeting these ILC2s might lead to new treatments for HCC.
Collapse
Affiliation(s)
- Xingyuan Xu
- Department of Hepatobiliary and Pancreatic Surgerythe First Affiliated HospitalZhejiang University School of MedicineHangzhouChina,Zhejiang Provincial Key Laboratory of Pancreatic DiseaseHangzhouChina
| | - Longyun Ye
- Department of Hepatobiliary and Pancreatic Surgerythe First Affiliated HospitalZhejiang University School of MedicineHangzhouChina,Zhejiang Provincial Key Laboratory of Pancreatic DiseaseHangzhouChina
| | - Qi Zhang
- Department of Hepatobiliary and Pancreatic Surgerythe First Affiliated HospitalZhejiang University School of MedicineHangzhouChina,Zhejiang Provincial Key Laboratory of Pancreatic DiseaseHangzhouChina
| | - Hang Shen
- Department of Hepatobiliary and Pancreatic Surgerythe First Affiliated HospitalZhejiang University School of MedicineHangzhouChina,Zhejiang Provincial Key Laboratory of Pancreatic DiseaseHangzhouChina
| | - Shanshan Li
- Department of Hepatobiliary and Pancreatic Surgerythe First Affiliated HospitalZhejiang University School of MedicineHangzhouChina,Zhejiang Provincial Key Laboratory of Pancreatic DiseaseHangzhouChina
| | - Xiaoyu Zhang
- Department of Hepatobiliary and Pancreatic Surgerythe First Affiliated HospitalZhejiang University School of MedicineHangzhouChina,Zhejiang Provincial Key Laboratory of Pancreatic DiseaseHangzhouChina
| | - Mao Ye
- Department of Hepatobiliary and Pancreatic Surgerythe First Affiliated HospitalZhejiang University School of MedicineHangzhouChina,Zhejiang Provincial Key Laboratory of Pancreatic DiseaseHangzhouChina
| | - Tingbo Liang
- Department of Hepatobiliary and Pancreatic Surgerythe First Affiliated HospitalZhejiang University School of MedicineHangzhouChina,Zhejiang Provincial Key Laboratory of Pancreatic DiseaseHangzhouChina,Innovation Center for the Study of Pancreatic Disease of Zhejiang ProvinceHangzhouChina
| |
Collapse
|
17
|
Mathä L, Romera-Hernández M, Steer CA, Yin YH, Orangi M, Shim H, Chang C, Rossi FM, Takei F. Migration of Lung Resident Group 2 Innate Lymphoid Cells Link Allergic Lung Inflammation and Liver Immunity. Front Immunol 2021; 12:679509. [PMID: 34305911 PMCID: PMC8299566 DOI: 10.3389/fimmu.2021.679509] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2021] [Accepted: 06/24/2021] [Indexed: 11/29/2022] Open
Abstract
Group 2 innate lymphoid cells (ILC2s) are tissue resident in the lung and activated by inhaled allergens via epithelial-derived alarmins including IL-33. Activated ILC2s proliferate, produce IL-5 and IL-13, and induce eosinophilic inflammation. Here, we report that intranasal IL-33 or the protease allergen papain administration resulted in increased numbers of ILC2s not only in the lung but also in peripheral blood and liver. Analyses of IL-33 treated parabiosis mice showed that the increase in lung ILC2s was due to proliferation of lung resident ILC2s, whereas the increase in liver ILC2s was due to the migration of activated lung ILC2s. Lung-derived ILC2s induced eosinophilic hepatitis and expression of fibrosis-related genes. Intranasal IL-33 pre-treatment also attenuated concanavalin A-induced acute hepatitis and cirrhosis. These results suggest that activated lung resident ILC2s emigrate from the lung, circulate, settle in the liver and promote type 2 inflammation and attenuate type 1 inflammation.
Collapse
Affiliation(s)
- Laura Mathä
- Terry Fox Laboratory, British Columbia Cancer, Vancouver, BC, Canada.,Interdisciplinary Oncology Program, University of British Columbia, Vancouver, BC, Canada
| | - Mónica Romera-Hernández
- Terry Fox Laboratory, British Columbia Cancer, Vancouver, BC, Canada.,Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, BC, Canada
| | - Catherine A Steer
- Terry Fox Laboratory, British Columbia Cancer, Vancouver, BC, Canada
| | - Yi Han Yin
- Terry Fox Laboratory, British Columbia Cancer, Vancouver, BC, Canada
| | - Mona Orangi
- Terry Fox Laboratory, British Columbia Cancer, Vancouver, BC, Canada.,Interdisciplinary Oncology Program, University of British Columbia, Vancouver, BC, Canada
| | - Hanjoo Shim
- Terry Fox Laboratory, British Columbia Cancer, Vancouver, BC, Canada
| | - ChihKai Chang
- Biomedical Research Centre, University of British Columbia, Vancouver, BC, Canada
| | - Fabio M Rossi
- Biomedical Research Centre, University of British Columbia, Vancouver, BC, Canada.,Department of Medical Genetics, University of British Columbia, Vancouver, BC, Canada
| | - Fumio Takei
- Terry Fox Laboratory, British Columbia Cancer, Vancouver, BC, Canada.,Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, BC, Canada
| |
Collapse
|
18
|
Haim-Vilmovsky L, Henriksson J, Walker JA, Miao Z, Natan E, Kar G, Clare S, Barlow JL, Charidemou E, Mamanova L, Chen X, Proserpio V, Pramanik J, Woodhouse S, Protasio AV, Efremova M, Griffin JL, Berriman M, Dougan G, Fisher J, Marioni JC, McKenzie ANJ, Teichmann SA. Mapping Rora expression in resting and activated CD4+ T cells. PLoS One 2021; 16:e0251233. [PMID: 34003838 PMCID: PMC8130942 DOI: 10.1371/journal.pone.0251233] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2021] [Accepted: 04/22/2021] [Indexed: 11/19/2022] Open
Abstract
The transcription factor Rora has been shown to be important for the development of ILC2 and the regulation of ILC3, macrophages and Treg cells. Here we investigate the role of Rora across CD4+ T cells in general, but with an emphasis on Th2 cells, both in vitro as well as in the context of several in vivo type 2 infection models. We dissect the function of Rora using overexpression and a CD4-conditional Rora-knockout mouse, as well as a RORA-reporter mouse. We establish the importance of Rora in CD4+ T cells for controlling lung inflammation induced by Nippostrongylus brasiliensis infection, and have measured the effect on downstream genes using RNA-seq. Using a systematic stimulation screen of CD4+ T cells, coupled with RNA-seq, we identify upstream regulators of Rora, most importantly IL-33 and CCL7. Our data suggest that Rora is a negative regulator of the immune system, possibly through several downstream pathways, and is under control of the local microenvironment.
Collapse
MESH Headings
- Animals
- Antigens, Helminth/immunology
- Antigens, Helminth/metabolism
- CD4-Positive T-Lymphocytes/immunology
- Cells, Cultured
- Cytokines/metabolism
- Disease Models, Animal
- Female
- Gene Expression Regulation/immunology
- Lymphocyte Activation
- Macrophages/immunology
- Mice
- Mice, Inbred C57BL
- Mice, Knockout
- Mice, Transgenic
- Nippostrongylus/immunology
- Nuclear Receptor Subfamily 1, Group F, Member 1/immunology
- Nuclear Receptor Subfamily 1, Group F, Member 1/metabolism
- Pneumonia/immunology
- Pneumonia/parasitology
- Pneumonia/pathology
- Strongylida Infections/immunology
- Strongylida Infections/parasitology
- Th2 Cells/immunology
Collapse
Affiliation(s)
- Liora Haim-Vilmovsky
- EMBL-European Bioinformatics Institute, Wellcome Genome Campus, Hinxton, Cambridge, United Kingdom
- Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton, Cambridge, United Kingdom
| | - Johan Henriksson
- EMBL-European Bioinformatics Institute, Wellcome Genome Campus, Hinxton, Cambridge, United Kingdom
- Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton, Cambridge, United Kingdom
| | - Jennifer A. Walker
- MRC Laboratory of Molecular Biology, Cambridge Biomedical Campus, Cambridge, United Kingdom
| | - Zhichao Miao
- EMBL-European Bioinformatics Institute, Wellcome Genome Campus, Hinxton, Cambridge, United Kingdom
- Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton, Cambridge, United Kingdom
| | - Eviatar Natan
- EMBL-European Bioinformatics Institute, Wellcome Genome Campus, Hinxton, Cambridge, United Kingdom
| | - Gozde Kar
- EMBL-European Bioinformatics Institute, Wellcome Genome Campus, Hinxton, Cambridge, United Kingdom
- Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton, Cambridge, United Kingdom
| | - Simon Clare
- Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton, Cambridge, United Kingdom
| | - Jillian L. Barlow
- MRC Laboratory of Molecular Biology, Cambridge Biomedical Campus, Cambridge, United Kingdom
| | - Evelina Charidemou
- Department of Biochemistry, University of Cambridge, Cambridge, United Kingdom
| | - Lira Mamanova
- Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton, Cambridge, United Kingdom
| | - Xi Chen
- Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton, Cambridge, United Kingdom
| | - Valentina Proserpio
- EMBL-European Bioinformatics Institute, Wellcome Genome Campus, Hinxton, Cambridge, United Kingdom
- Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton, Cambridge, United Kingdom
| | - Jhuma Pramanik
- Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton, Cambridge, United Kingdom
| | - Steven Woodhouse
- Department of Haematology, Cambridge Institute for Medical Research, University of Cambridge, Cambridge, United Kingdom
- Wellcome Trust—Medical Research Council Cambridge Stem Cell Institute, University of Cambridge, Cambridge, United Kingdom
| | - Anna V. Protasio
- Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton, Cambridge, United Kingdom
| | - Mirjana Efremova
- Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton, Cambridge, United Kingdom
| | - Julian L. Griffin
- Department of Biochemistry, University of Cambridge, Cambridge, United Kingdom
- Department of Metabolism, Digestion and Reproduction, Biomolecular Medicine, Imperial College London, London, United Kingdom
| | - Matt Berriman
- Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton, Cambridge, United Kingdom
| | - Gordon Dougan
- Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton, Cambridge, United Kingdom
| | | | - John C. Marioni
- EMBL-European Bioinformatics Institute, Wellcome Genome Campus, Hinxton, Cambridge, United Kingdom
- Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton, Cambridge, United Kingdom
- Cancer Research UK Cambridge Institute, University of Cambridge, Cambridge, United Kingdom
| | - Andrew N. J. McKenzie
- MRC Laboratory of Molecular Biology, Cambridge Biomedical Campus, Cambridge, United Kingdom
| | - Sarah A. Teichmann
- EMBL-European Bioinformatics Institute, Wellcome Genome Campus, Hinxton, Cambridge, United Kingdom
- Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton, Cambridge, United Kingdom
- Theory of Condensed Matter, Cavendish Laboratory, Cambridge, United Kingdom
| |
Collapse
|
19
|
Asghar Pasha M, Yang Q. Innate Lymphoid Cells in Airway Inflammation. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2021; 1303:183-191. [PMID: 33788194 DOI: 10.1007/978-3-030-63046-1_11] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Airways are constantly exposed to antigens and various pathogens. Immune cells in the airways act as first line defense system against these pathogens, involving both innate and acquired immunity. There is accumulating evidence that innate lymphoid cells, newly identified lymphoid lineage cells, play a critical role in regulating tissue homeostasis and in the pathogenesis of inflammation. Cytokines produced by other cells activate innate lymphoid cells, which in turn produce large amount of cytokines that result in inflammation. Type 2 innate lymphoid cells (ILC2s) are recognized as key component of T helper type 2 (Th2) inflammation, and are known to be elevated in type 2 (T2) human airway diseases (asthma). Th2 cytokines produced by ILC2s amplify inflammation via activation of eosinophils, B cells, mast cell, and macrophages. "T2 high asthma" has an increased Th2 response triggered by elevation of IL-4, IL-5 and IL-13 and other inflammatory mediators, leading to increased eosinophilic inflammation. The growing evidence of ILC2 contribution in the induction and maintenance of allergic inflammation suggests that targeting upstream mediators may affect both the innate and adaptive immune responses and all disease phenotypes. Blocking ILC2 activators, activation of inhibitory pathways of ILC2, or suppression of ILC2-mediated pathways may be therapeutic strategies for the type 2 airway diseases.
Collapse
Affiliation(s)
- M Asghar Pasha
- Division of Allergy and Immunology, Department of Medicine, Albany Medical College, Albany, NY, USA.
| | - Qi Yang
- Department of Microbial Disease & Immunology, Albany Medical College, Albany, NY, USA
| |
Collapse
|
20
|
Mattei F, Andreone S, Marone G, Gambardella AR, Loffredo S, Varricchi G, Schiavoni G. Eosinophils in the Tumor Microenvironment. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2021; 1273:1-28. [PMID: 33119873 DOI: 10.1007/978-3-030-49270-0_1] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Eosinophils are rare blood-circulating and tissue-infiltrating immune cells studied for decades in the context of allergic diseases and parasitic infections. Eosinophils can secrete a wide array of soluble mediators and effector molecules, with potential immunoregulatory activities in the tumor microenvironment (TME). These findings imply that these cells may play a role in cancer immunity. Despite these cells were known to infiltrate tumors since many years ago, their role in TME is gaining attention only recently. In this chapter, we will review the main biological functions of eosinophils that can be relevant within the TME. We will discuss how these cells may undergo phenotypic changes acquiring pro- or antitumoricidal properties according to the surrounding stimuli. Moreover, we will analyze canonical (i.e., degranulation) and unconventional mechanisms (i.e., DNA traps, exosome secretion) employed by eosinophils in inflammatory contexts, which can be relevant for tumor immune responses. Finally, we will review the available preclinical models that could be employed for the study of the role in vivo of eosinophils in cancer.
Collapse
Affiliation(s)
- Fabrizio Mattei
- Department of Oncology and Molecular Medicine, Istituto Superiore di Sanità, Rome, Italy
| | - Sara Andreone
- Department of Oncology and Molecular Medicine, Istituto Superiore di Sanità, Rome, Italy
| | - Giancarlo Marone
- Department of Public Health, University of Naples Federico II, Naples, Italy.,Azienda Ospedaliera Ospedali dei Colli - Monaldi Hospital Pharmacy, Naples, Italy
| | | | - Stefania Loffredo
- Department of Translational Medical Sciences and Center for Basic and Clinical Immunology Research (CISI), University of Naples Federico II, Naples, Italy.,WAO Center of Excellence, Naples, Italy.,Institute of Experimental Endocrinology and Oncology "G. Salvatore" (IEOS), National Research Council (CNR), Naples, Italy
| | - Gilda Varricchi
- Department of Translational Medical Sciences and Center for Basic and Clinical Immunology Research (CISI), University of Naples Federico II, Naples, Italy. .,WAO Center of Excellence, Naples, Italy. .,Institute of Experimental Endocrinology and Oncology "G. Salvatore" (IEOS), National Research Council (CNR), Naples, Italy.
| | - Giovanna Schiavoni
- Department of Oncology and Molecular Medicine, Istituto Superiore di Sanità, Rome, Italy.
| |
Collapse
|
21
|
Naserian S, Leclerc M, Shamdani S, Uzan G. Current Preventions and Treatments of aGVHD: From Pharmacological Prophylaxis to Innovative Therapies. Front Immunol 2020; 11:607030. [PMID: 33391276 PMCID: PMC7773902 DOI: 10.3389/fimmu.2020.607030] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2020] [Accepted: 11/16/2020] [Indexed: 12/16/2022] Open
Abstract
Graft versus host disease (GVHD) is one of the main causes of mortality and the reason for up to 50% of morbidity after hematopoietic stem cell transplantations (HSCT) which is the treatment of choice for many blood malignancies. Thanks to years of research and exploration, we have acquired a profound understanding of the pathophysiology and immunopathology of these disorders. This led to the proposition and development of many therapeutic approaches during the last decades, some of them with very promising results. In this review, we have focused on the recent GVHD treatments from classical chemical and pharmacological prophylaxis to more innovative treatments including gene therapy and cell therapy, most commonly based on the application of a variety of immunomodulatory cells. Furthermore, we have discussed the advantages and potentials of cell-free therapy as a newly emerging approach to treat GVHD. Among them, we have particularly focused on the implication of the TNFα-TNFR2 axis as a new immune checkpoint signaling pathway controlling different aspects of many immunoregulatory cells.
Collapse
Affiliation(s)
- Sina Naserian
- INSERM UMR-S-MD 1197, Hôpital Paul Brousse, Villejuif, France
- Paris-Saclay University, Villejuif, France
- CellMedEx, Saint Maur Des Fossés, France
| | - Mathieu Leclerc
- Service d’Hématologie Clinique et de Thérapie Cellulaire, Hôpital Henri Mondor, Créteil, France
- INSERM U955, Institut Mondor de Recherche Biomédicale, Créteil, France
- Faculté de Médecine de Créteil, Université Paris-Est, Créteil, France
| | - Sara Shamdani
- INSERM UMR-S-MD 1197, Hôpital Paul Brousse, Villejuif, France
- Paris-Saclay University, Villejuif, France
- CellMedEx, Saint Maur Des Fossés, France
| | - Georges Uzan
- INSERM UMR-S-MD 1197, Hôpital Paul Brousse, Villejuif, France
- Paris-Saclay University, Villejuif, France
| |
Collapse
|
22
|
Maggi E, Veneziani I, Moretta L, Cosmi L, Annunziato F. Group 2 Innate Lymphoid Cells: A Double-Edged Sword in Cancer? Cancers (Basel) 2020; 12:cancers12113452. [PMID: 33233582 PMCID: PMC7699723 DOI: 10.3390/cancers12113452] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2020] [Revised: 11/16/2020] [Accepted: 11/17/2020] [Indexed: 12/14/2022] Open
Abstract
Group 2 Innate Lymphoid Cells (ILC2s) belong to the family of helper ILCs which provide host defense against infectious agents, participate in inflammatory responses and mediate lymphoid organogenesis and tissue repair, mainly at the skin and mucosal level. Based on their transcriptional, phenotypic and functional profile, ILC2s mirror the features of the adaptive CD4+ Th2 cell subset, both contributing to the so-called type 2 immune response. Similar to other ILCs, ILC2s are rapidly activated by signals deriving from tissue and/or other tissue-resident immune cells. The biologic activity of ILCs needs to be tightly regulated in order to prevent them from contributing to severe inflammation and damage in several organs. Indeed, ILC2s display both enhancing and regulatory roles in several pathophysiological conditions, including tumors. In this review, we summarize the actual knowledge about ILC2s ability to induce or impair a protective immune response, their pro- or antitumor activity in murine models, human (children and adults) pathologies and the potential strategies to improve cancer immunotherapy by exploiting the features of ILC2s.
Collapse
Affiliation(s)
- Enrico Maggi
- Immunology Department, Bambino Gesù Children Hospital, 00165 Rome, Italy; (I.V.); (L.M.)
- Correspondence: ; Tel.: +39-06-6859-3617
| | - Irene Veneziani
- Immunology Department, Bambino Gesù Children Hospital, 00165 Rome, Italy; (I.V.); (L.M.)
| | - Lorenzo Moretta
- Immunology Department, Bambino Gesù Children Hospital, 00165 Rome, Italy; (I.V.); (L.M.)
| | - Lorenzo Cosmi
- Department of Experimental and Clinical Medicine, University of Florence, 50134 Florence, Italy; (L.C.); (F.A.)
| | - Francesco Annunziato
- Department of Experimental and Clinical Medicine, University of Florence, 50134 Florence, Italy; (L.C.); (F.A.)
| |
Collapse
|
23
|
Figueroa-Vega N, Marín-Aragón CI, López-Aguilar I, Ibarra-Reynoso L, Pérez-Luque E, Malacara JM. Analysis of the percentages of monocyte subsets and ILC2s, their relationships with metabolic variables and response to hypocaloric restriction in obesity. PLoS One 2020; 15:e0228637. [PMID: 32074122 PMCID: PMC7029876 DOI: 10.1371/journal.pone.0228637] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2019] [Accepted: 01/21/2020] [Indexed: 01/17/2023] Open
Abstract
PURPOSE Obesity results from excess energy intake over expenditure and is characterized by chronic low-grade inflammation involving circulating monocytes (Mo) and group 2 innate lymphoid cells (ILC2s) imbalance. We analyzed circulating Mo subsets and ILC2s percentages and β2-adrenergic receptor (β2AR) expression in lean and obese subjects, and the possible effect of hypocaloric restriction on these innate immune cells. METHODS In 139 individuals aged 45 to 57 years, classified in 74 lean individuals (>18.9kg/m2 BMI <24.9kg/m2) and 65 with obesity (n = 65), we collected fasting blood samples to detect Mo subsets, ILC2s number, and β2AR expression by flow cytometry. Lipids, insulin, leptin, and acylated-ghrelin concentrations were quantified. Resting energy expenditure (REE) was estimated by indirect calorimetry. These measurements were repeated in obese subjects after 7-weeks of hypocaloric restriction. RESULTS Non-classical monocytes (NCM) and β2AR expression on intermediate Mo (IM) were increased in obese individuals (p<0.001, in both cases), whereas the percent of ILC2s was decreased (p<0.0001). Stepwise regression analysis showed significantly negative associations of ILC2s with caloric intake, β2AR expression on IM with REE, but a positive relationship between NCM and HOMA-IR. Caloric restriction allowed a significant diminution of NCM and the β2AR expression on IM, as well as, an increase in the percent of classical Mo (CM), and ILC2s. ΔREE was related to ΔCD16+/CD16- ratio. CONCLUSIONS These findings show that in obesity occur changes in NCM, ILC2s and β2AR expression, which contribute to the low-grade inflammation linked to obesity and might revert with caloric restriction.
Collapse
Affiliation(s)
- Nicté Figueroa-Vega
- Department of Medical Sciences, University of Guanajuato, León Campus, León, Gto., México
| | | | - Itzel López-Aguilar
- Department of Medical Sciences, University of Guanajuato, León Campus, León, Gto., México
| | - Lorena Ibarra-Reynoso
- Department of Medical Sciences, University of Guanajuato, León Campus, León, Gto., México
| | - Elva Pérez-Luque
- Department of Medical Sciences, University of Guanajuato, León Campus, León, Gto., México
| | - Juan Manuel Malacara
- Department of Medical Sciences, University of Guanajuato, León Campus, León, Gto., México
| |
Collapse
|
24
|
Ting HA, von Moltke J. The Immune Function of Tuft Cells at Gut Mucosal Surfaces and Beyond. THE JOURNAL OF IMMUNOLOGY 2019; 202:1321-1329. [PMID: 30782851 DOI: 10.4049/jimmunol.1801069] [Citation(s) in RCA: 70] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/01/2018] [Accepted: 09/21/2018] [Indexed: 12/25/2022]
Abstract
Tuft cells were first discovered in epithelial barriers decades ago, but their function remained unclear until recently. In the last 2 years, a series of studies has provided important advances that link tuft cells to infectious diseases and the host immune responses. Broadly, a model has emerged in which tuft cells use chemosensing to monitor their surroundings and translate environmental signals into effector functions that regulate immune responses in the underlying tissue. In this article, we review the current understanding of tuft cell immune function in the intestines, airways, and thymus. In particular, we discuss the role of tuft cells in type 2 immunity, norovirus infection, and thymocyte development. Despite recent advances, many fundamental questions about the function of tuft cells in immunity remain to be answered.
Collapse
Affiliation(s)
- Hung-An Ting
- Department of Immunology, University of Washington School of Medicine, Seattle, WA 98109
| | - Jakob von Moltke
- Department of Immunology, University of Washington School of Medicine, Seattle, WA 98109
| |
Collapse
|
25
|
Kabata H, Moro K, Koyasu S. The group 2 innate lymphoid cell (ILC2) regulatory network and its underlying mechanisms. Immunol Rev 2019; 286:37-52. [PMID: 30294963 DOI: 10.1111/imr.12706] [Citation(s) in RCA: 210] [Impact Index Per Article: 35.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2018] [Accepted: 08/16/2018] [Indexed: 12/12/2022]
Abstract
Group 2 innate lymphoid cells (ILC2s) play critical roles in the induction of type 2 inflammation, response to parasite infection, metabolic homeostasis, and tissue repair. These multifunctional roles of ILC2s are tightly controlled by complex regulatory systems in the local microenvironment, the disruption of which may cause various health problems. This review summarizes up-to-date knowledge regarding positive and negative regulators for ILC2s based on their function and signaling pathways, including activating cytokines (IL-33, IL-25; MAPK, NF-κB pathways), co-stimulatory cytokines (IL-2, IL-7, IL-9, TSLP; STAT5, IL-4; STAT6, TNF superfamily; MAPK, NF-κB pathways), suppressive cytokines (type1 IFNs, IFN-γ, IL-27; STAT1, IL-10, TGF-β), transdifferentiation cytokines (IL-12; STAT4, IL-1β, IL-18), lipid mediators (LTC4, LTD4, LTE4, PGD2; Ca2+ -NFAT pathways, PGE2, PGI2; AC/cAMP/PKA pathways, LXA4, LTB4), neuropeptides (NMU; Ca2+ -NFAT, MAPK pathways, VIP, CGRP, catecholamine, acetylcholine), sex hormones (androgen, estrogen), nutrients (butyrate; HDAC inhibitors, vitamins), and cell-to-cell interactions (ICOSL-ICOS; STAT5, B7-H6-NKp30, E-cadherin-KLRG1). This comprehensive review affords a better understanding of the regulatory network system for ILC2s, providing impetus to develop new treatment strategies for ILC2-related health problems.
Collapse
Affiliation(s)
- Hiroki Kabata
- Laboratory for Immune Cell Systems, RIKEN Center for Integrative Medical Sciences (IMS), Yokohama, Japan.,Laboratory for Innate Immune Systems, RIKEN Center for Integrative Medical Sciences (IMS), Yokohama, Japan.,Jill Roberts Institute for Research in Inflammatory Bowel Disease, Joan and Sanford I. Weill Department of Medicine, Department of Microbiology and Immunology, Weill Cornell Medicine, Cornell University, New York, New York, USA.,Division of Pulmonary Medicine, Department of Medicine, Keio University School of Medicine, Tokyo, Japan
| | - Kazuyo Moro
- Laboratory for Innate Immune Systems, RIKEN Center for Integrative Medical Sciences (IMS), Yokohama, Japan.,Division of Immunobiology, Department of Medical Life Science, Graduate School of Medical Life Science, Yokohama City University, Yokohama, Japan
| | - Shigeo Koyasu
- Laboratory for Immune Cell Systems, RIKEN Center for Integrative Medical Sciences (IMS), Yokohama, Japan.,Department of Microbiology and Immunology, Keio University School of Medicine, Tokyo, Japan
| |
Collapse
|
26
|
Dzopalić T, Božić-Nedeljković B, Jurišić V. Function of innate lymphoid cells in the immune-related disorders. Hum Cell 2019; 32:231-239. [DOI: 10.1007/s13577-019-00257-1] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2019] [Accepted: 05/05/2019] [Indexed: 12/12/2022]
|
27
|
Peters AL, Luo Z, Li J, Mourya R, Wang Y, Dexheimer P, Shivakumar P, Aronow B, Bezerra JA. Single cell RNA sequencing reveals regional heterogeneity of hepatobiliary innate lymphoid cells in a tissue-enriched fashion. PLoS One 2019; 14:e0215481. [PMID: 31022195 PMCID: PMC6483339 DOI: 10.1371/journal.pone.0215481] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2018] [Accepted: 04/02/2019] [Indexed: 12/29/2022] Open
Abstract
IL-33 promotes type 2 immunity, epithelial repair, and tissue fibrosis by activating group 2 innate lymphoid cells (ILC2). ILC2 lack all known surface markers of mature T, B, NK, and myeloid cell lineages (Linneg), express the IL-33 receptor ST2, and release type 2 cytokines which contribute to cholangiocyte proliferation and activation of hepatic stellate cells. This pathway results in massive proliferation of the extrahepatic bile duct (EHBD) but also exacerbates liver fibrosis, suggesting that there may be tissue-specific subpopulations of IL-33-induced ILC. To determine the tissue-specific subsets of ILC in the hepatobiliary system, we analyzed CD45+Linneg mononuclear cells from IL-33 treated adult Balb/c mouse liver or EHBD by single cell RNA sequencing. Principal component analysis identified 6 major CD45+Linneg cell classes, two of which were restricted to the EHBD. One of these classes, biliary immature myeloid (BIM) cells, was predicted to interact with ILC2 by a network of shared receptor-ligand pairs. BIM highly expressed Gp49 and ST2 receptors on the cell surface while lacking surface expression of markers for mature myeloid cells. In conclusion, single cell RNA sequencing identified IL-33 responsive cell groups regionally confined to the liver or extrahepatic bile duct, including a novel population of CD45+Linneg Gp49-expressing mononuclear cells.
Collapse
Affiliation(s)
- Anna L. Peters
- Department of Pediatrics, Division of Gastroenterology, Hepatology and Nutrition, Cincinnati Children’s Hospital Medical Center, Cincinnati, Ohio, United States of America
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, Ohio, United States of America
| | - Zhenhua Luo
- Department of Pediatrics, Division of Gastroenterology, Hepatology and Nutrition, Cincinnati Children’s Hospital Medical Center, Cincinnati, Ohio, United States of America
| | - Jun Li
- Department of Pediatrics, Division of Gastroenterology, Hepatology and Nutrition, Cincinnati Children’s Hospital Medical Center, Cincinnati, Ohio, United States of America
| | - Reena Mourya
- Department of Pediatrics, Division of Gastroenterology, Hepatology and Nutrition, Cincinnati Children’s Hospital Medical Center, Cincinnati, Ohio, United States of America
| | - Yunguan Wang
- Department of Pediatrics, Division of Bioinformatics, Cincinnati Children’s Hospital Medical Center, Cincinnati, Ohio, United States of America
| | - Phillip Dexheimer
- Department of Pediatrics, Division of Bioinformatics, Cincinnati Children’s Hospital Medical Center, Cincinnati, Ohio, United States of America
| | - Pranav Shivakumar
- Department of Pediatrics, Division of Gastroenterology, Hepatology and Nutrition, Cincinnati Children’s Hospital Medical Center, Cincinnati, Ohio, United States of America
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, Ohio, United States of America
| | - Bruce Aronow
- Department of Pediatrics, Division of Bioinformatics, Cincinnati Children’s Hospital Medical Center, Cincinnati, Ohio, United States of America
| | - Jorge A. Bezerra
- Department of Pediatrics, Division of Gastroenterology, Hepatology and Nutrition, Cincinnati Children’s Hospital Medical Center, Cincinnati, Ohio, United States of America
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, Ohio, United States of America
| |
Collapse
|
28
|
Ebihara T, Taniuchi I. Transcription Factors in the Development and Function of Group 2 Innate Lymphoid Cells. Int J Mol Sci 2019; 20:ijms20061377. [PMID: 30893794 PMCID: PMC6470746 DOI: 10.3390/ijms20061377] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2019] [Revised: 03/15/2019] [Accepted: 03/18/2019] [Indexed: 12/18/2022] Open
Abstract
Group 2 innate lymphoid cells (ILC2s) are tissue-resident cells and are a major source of innate TH2 cytokine secretion upon allergen exposure or parasitic-worm infection. Accumulating studies have revealed that transcription factors, including GATA-3, Bcl11b, Gfi1, RORα, and Ets-1, play a role in ILC2 differentiation. Recent reports have further revealed that the characteristics and functions of ILC2 are influenced by the physiological state of the tissues. Specifically, the type of inflammation strongly affects the ILC2 phenotype in tissues. Inhibitory ILC2s, memory-like ILC2s, and ex-ILC2s with ILC1 features acquire their characteristic properties following exposure to their specific inflammatory environment. We have recently reported a new ILC2 population, designated as exhausted-like ILC2s, which emerges after a severe allergic inflammation. Exhausted-like ILC2s are featured with low reactivity and high expression of inhibitory receptors. Therefore, for a more comprehensive understanding of ILC2 function and differentiation, we review the recent knowledge of transcriptional regulation of ILC2 differentiation and discuss the roles of the Runx transcription factor in controlling the emergence of exhausted-like ILC2s. The concept of exhausted-like ILC2s sheds a light on a new aspect of ILC2 biology in allergic diseases.
Collapse
Affiliation(s)
- Takashi Ebihara
- Laboratory for Transcriptional Regulation, RIKEN Center for Integrative Medical Sciences (IMS), 1-7-22 Suehiro-cho, Tsurumi-ku, Yokohama 230-0045, Japan.
| | - Ichiro Taniuchi
- Laboratory for Transcriptional Regulation, RIKEN Center for Integrative Medical Sciences (IMS), 1-7-22 Suehiro-cho, Tsurumi-ku, Yokohama 230-0045, Japan.
| |
Collapse
|
29
|
Esvelt MA, Freeman ZT, Pearson AT, Harkema JR, Clines GA, Clines KL, Dyson MC, Hoenerhoff MJ. The Endothelin-A Receptor Antagonist Zibotentan Induces Damage to the Nasal Olfactory Epithelium Possibly Mediated in Part through Type 2 Innate Lymphoid Cells. Toxicol Pathol 2019; 47:150-164. [PMID: 30595110 PMCID: PMC7357205 DOI: 10.1177/0192623318816295] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
Zibotentan, an endothelin-A receptor antagonist, has been used in the treatment of various cardiovascular disorders and neoplasia. Castrated athymic nude mice receiving zibotentan for a preclinical xenograft efficacy study experienced weight loss, gastrointestinal bloat, and the presence of an audible respiratory click. Human side effects have been reported in the nasal cavity, so we hypothesized that the nasal cavity is a target for toxicity in mice receiving zibotentan. Lesions in the nasal cavity predominantly targeted olfactory epithelium in treated mice and were more pronounced in castrated animals. Minimal lesions were present in vehicle control animals, which suggested possible gavage-related reflux injury. The incidence, distribution, and morphology of lesions suggested direct exposure to the nasal mucosa and a possible systemic effect targeting the olfactory epithelium, driven by a type 2 immune response, with group 2 innate lymphoid cell involvement. Severe nasal lesions may have resulted in recurrent upper airway obstruction, leading to aerophagia and associated clinical morbidity. These data show the nasal cavity is a target of zibotentan when given by gavage in athymic nude mice, and such unanticipated and off-target effects could impact interpretation of research results and animal health in preclinical studies.
Collapse
Affiliation(s)
- Marian A Esvelt
- Unit for Laboratory Animal Medicine, University of Michigan, Ann Arbor, MI 48109
- Animal Resource Center, University of Texas Southwestern Medical Center, Dallas, TX 75390
| | - Zachary T Freeman
- Unit for Laboratory Animal Medicine, University of Michigan, Ann Arbor, MI 48109
| | - Alexander T Pearson
- Section of Hematology/Oncology, The University of Chicago Medicine & Biological Sciences, Chicago, IL, 60637
| | - Jack R Harkema
- Department of Pathobiology and Diagnostic Investigation, College of Veterinary Medicine, Michigan State University, East Lansing, MI 48824
| | - Gregory A Clines
- Division of Metabolism, Endocrinology, and Diabetes, University of Michigan, Ann Arbor, MI 48109
- Endocrinology Section, Ann Arbor VA Medical Center, Ann Arbor, Michigan 48105
| | - Katrina L Clines
- Division of Metabolism, Endocrinology, and Diabetes, University of Michigan, Ann Arbor, MI 48109
| | - Melissa C Dyson
- Unit for Laboratory Animal Medicine, University of Michigan, Ann Arbor, MI 48109
| | - Mark J Hoenerhoff
- Unit for Laboratory Animal Medicine, University of Michigan, Ann Arbor, MI 48109
- In Vivo Animal Core, University of Michigan, Ann Arbor, MI 48109
| |
Collapse
|
30
|
Miyamoto C, Kojo S, Yamashita M, Moro K, Lacaud G, Shiroguchi K, Taniuchi I, Ebihara T. Runx/Cbfβ complexes protect group 2 innate lymphoid cells from exhausted-like hyporesponsiveness during allergic airway inflammation. Nat Commun 2019; 10:447. [PMID: 30683858 PMCID: PMC6347616 DOI: 10.1038/s41467-019-08365-0] [Citation(s) in RCA: 57] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2018] [Accepted: 01/08/2019] [Indexed: 01/01/2023] Open
Abstract
Group 2 innate lymphoid cells (ILC2s) have tissue-resident competence and contribute to the pathogenesis of allergic diseases. However, the mechanisms regulating prolonged ILC2-mediated TH2 cytokine production under chronic inflammatory conditions are unclear. Here we show that, at homeostasis, Runx deficiency induces excessive ILC2 activation due to overly active GATA-3 functions. By contrast, during allergic inflammation, the absence of Runx impairs the ability of ILC2s to proliferate and produce effector TH2 cytokines and chemokines. Instead, functional deletion of Runx induces the expression of exhaustion markers, such as IL-10 and TIGIT, on ILC2s. Finally, these 'exhausted-like' ILC2s are unable to induce type 2 immune responses to repeated allergen exposures. Thus, Runx confers competence for sustained ILC2 activity at the mucosa, and contributes to allergic pathogenesis.
Collapse
Affiliation(s)
- Chizuko Miyamoto
- Laboratory for Transcriptional Regulation, RIKEN Center for Integrative Medical Sciences (IMS), 1-7-22 Suehiro-cho, Tsurumi-ku, Yokohama, 230-0045, Japan
| | - Satoshi Kojo
- Laboratory for Transcriptional Regulation, RIKEN Center for Integrative Medical Sciences (IMS), 1-7-22 Suehiro-cho, Tsurumi-ku, Yokohama, 230-0045, Japan
| | - Motoi Yamashita
- Laboratory for Transcriptional Regulation, RIKEN Center for Integrative Medical Sciences (IMS), 1-7-22 Suehiro-cho, Tsurumi-ku, Yokohama, 230-0045, Japan
| | - Kazuyo Moro
- Laboratory for Innate Immune Systems, RIKEN Center for Integrative Medical Sciences (IMS), 1-7-22 Suehiro-cho, Tsurumi-ku, Yokohama, 230-0045, Japan
| | - Georges Lacaud
- Stem Cell Biology Group, Cancer Research UK Manchester Institute, The University of Manchester, Wilmslow Road, Manchester, M20 4BX, UK
| | - Katsuyuki Shiroguchi
- Laboratory for Immunogenetics, RIKEN Center for Integrative Medical Sciences (IMS), 1-7-22 Suehiro-cho, Tsurumi-ku, Yokohama, 230-0045, Japan
- Laboratory for Prediction of Cell Systems Dynamics, RIKEN Center for Biosystems Dynamics Research (BDR), Suita, Osaka, 565-0874, Japan
- JST PRESTO, Kawaguchi, 332-0012, Japan
| | - Ichiro Taniuchi
- Laboratory for Transcriptional Regulation, RIKEN Center for Integrative Medical Sciences (IMS), 1-7-22 Suehiro-cho, Tsurumi-ku, Yokohama, 230-0045, Japan
| | - Takashi Ebihara
- Laboratory for Transcriptional Regulation, RIKEN Center for Integrative Medical Sciences (IMS), 1-7-22 Suehiro-cho, Tsurumi-ku, Yokohama, 230-0045, Japan.
| |
Collapse
|
31
|
Sex-associated TSLP-induced immune alterations following early-life RSV infection leads to enhanced allergic disease. Mucosal Immunol 2019; 12:969-979. [PMID: 31076663 PMCID: PMC6599479 DOI: 10.1038/s41385-019-0171-3] [Citation(s) in RCA: 60] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2018] [Revised: 03/23/2019] [Accepted: 04/22/2019] [Indexed: 02/04/2023]
Abstract
Many studies have linked severe RSV infection during early-life with an enhanced likelihood of developing childhood asthma, showing a greater susceptibility in boys. Our studies show that early-life RSV infection leads to differential long-term effects based upon the sex of the neonate; leaving male mice prone to exacerbation upon secondary allergen exposure while overall protecting female mice. During initial viral infection, we observed better viral control in the female mice with correlative expression of interferon-β that was not observed in male mice. Additionally, we observed persistent immune alterations in male mice at 4 weeks post infection. These alterations include Th2 and Th17-skewing, innate cytokine expression (Tslp and Il33), and infiltration of innate immune cells (DC and ILC2). Upon exposure to allergen, beginning at 4 weeks following early-life RSV-infection, male mice show severe allergic exacerbation while female mice appear to be protected. Due to persistent expression of TSLP following early-life RSV infection in male mice, genetically modified TSLPR-/- mice were evaluated and demonstrated an abrogation of allergen exacerbation in male mice. These data indicate that TSLP is involved in the altered immune environment following neonatal RSV-infection that leads to more severe responses in males during allergy exposure, later in life. Thus, TSLP may be a clinically relevant therapeutic target early in life.
Collapse
|
32
|
Abstract
Innate lymphoid cells (ILC) are a recently identified group of innate lymphocytes that are preferentially located at barrier surfaces. Barrier surfaces are in direct contact with complex microbial ecosystems, collectively referred to as the microbiota. It is now believed that the interplay of the microbiota with host components (i.e. epithelial cells and immune cells) promotes host fitness by regulating organ homeostasis, metabolism, and host defense against pathogens. In this review, we will give an overview of this multifaceted interplay between ILC and components of the microbiota.
Collapse
Affiliation(s)
- Liudmila Britanova
- Research Centre Immunotherapy and Institute of Microbiology and Hygiene, Mainz, Germany
| | - Andreas Diefenbach
- Department of Microbiology, Charité - Universitätsmedizin Berlin, and Berlin Institute of Health, Berlin, Germany
| |
Collapse
|
33
|
Huang YC, Weng CM, Lee MJ, Lin SM, Wang CH, Kuo HP. Endotypes of severe allergic asthma patients who clinically benefit from anti-IgE therapy. Clin Exp Allergy 2018; 49:44-53. [PMID: 30107059 DOI: 10.1111/cea.13248] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2018] [Revised: 08/02/2018] [Accepted: 08/07/2018] [Indexed: 01/06/2023]
Abstract
BACKGROUND Omalizumab, a recombinant monoclonal anti-IgE antibody, was developed for the treatment of severe allergic asthma. Not all these patients respond to omalizumab. OBJECTIVE This study aimed to evaluate whether the proinflammatory cytokine profiles in the severe allergic asthma patients were different between who responded and nonresponded to omalizumab therapy. METHODS A prospective study was conducted to examine type 2 cytokines and epithelium-derived cytokines in the bronchial tissues by immunohistochemistry, Western blot and PCR analysis among patients with severe allergic asthma before and after omalizumab therapy. RESULTS Fourteen of 23 patients with unstable severe allergic asthma improved their asthma control after 4 months of omalizumab treatment (Responders), while nine failed to improve (Non-Responders). Most of Responders were type 2-high endotype (12/14) with upregulated expression of IL-33, IL-25 and TSLP in their bronchial tissues, while most of Non-Responders were type 2-low endotype (8/9). Repeated bronchoscopic biopsy was done in nine responders after omalizumab treatment and showed a decline in IL-13, IL-33, IL-25 and TSLP expression in the bronchial tissues. Among 14 Responders who continued omalizuamb treatments to a total 12 months, six patients achieved a well control of asthma (ACT ≥ 23), while eight patients required additional treatment for asthma symptoms and had more rhinosinusitis comorbidities and a mixed eosinophilic and neutrophilic inflammation in their bronchial tissues. CONCLUSION Most of the severe allergic asthma patients who benefited from omalizumab treatment were IL-33, IL-25 and TSLP aggravated type 2-high endotype. Rhinosinusitis or with a mixed eosinophilic and neutrophilic airway inflammation should be evaluated in patients who partially responded to omalizumab treatment.
Collapse
Affiliation(s)
- Yu-Chen Huang
- Department of Thoracic Medicine, Chang Gung Memorial Hospital, Taipei, Taiwan
| | - Chih-Ming Weng
- College of Medicine, Taipei Medical University, Taipei, Taiwan
| | - Meng-Jung Lee
- College of Medicine, Taipei Medical University, Taipei, Taiwan
| | - Shu-Min Lin
- Department of Thoracic Medicine, Chang Gung Memorial Hospital, Taipei, Taiwan
| | - Chun-Hua Wang
- Department of Thoracic Medicine, Chang Gung Memorial Hospital, Taipei, Taiwan
| | - Han-Pin Kuo
- College of Medicine, Taipei Medical University, Taipei, Taiwan
| |
Collapse
|
34
|
Lloyd CM, Snelgrove RJ. Type 2 immunity: Expanding our view. Sci Immunol 2018; 3:eaat1604. [PMID: 29980619 DOI: 10.1126/sciimmunol.aat1604] [Citation(s) in RCA: 196] [Impact Index Per Article: 28.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2018] [Accepted: 05/30/2018] [Indexed: 12/14/2022]
Abstract
The classical vision of type 2 immune reactions is that they are characterized by a distinct cellular and cytokine repertoire that is critical for host resistance against helminthic worm infections but, when dysregulated, may cause atopic reactions that result in conditions such as asthma, rhinitis, dermatitis, and anaphylaxis. In this traditional view, the type 2 response is categorized as an adaptive immune response with differentiated T helper cells taking center stage, driving eosinophil recruitment and immunoglobulin production via the secretion of a distinct repertoire of cytokines that include interleukin-4 (IL-4), IL-5, and IL-13. The recent discovery of a group of innate cells that has the capacity to secrete copious amounts of type 2 cytokines, potentially in the absence of adaptive immunity, has reignited interest in type 2 biology. The discovery that these innate lymphoid cells and type 2 cytokines are involved in diverse biological processes-including wound healing, control of metabolic homeostasis, and temperature-has considerably changed our view of type 2 responses and the cytokines, chemokines, and receptors that regulate these responses.
Collapse
Affiliation(s)
- Clare M Lloyd
- Imperial College London, Sir Alexander Fleming Building, South Kensington NHLI, Campus, London SW7 2AZ, UK.
| | - Robert J Snelgrove
- Imperial College London, Sir Alexander Fleming Building, South Kensington NHLI, Campus, London SW7 2AZ, UK
| |
Collapse
|
35
|
Schuijs MJ, Halim TYF. Group 2 innate lymphocytes at the interface between innate and adaptive immunity. Ann N Y Acad Sci 2018; 1417:87-103. [PMID: 29492980 DOI: 10.1111/nyas.13604] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2017] [Revised: 12/22/2017] [Accepted: 12/31/2017] [Indexed: 12/23/2022]
Abstract
Group 2 innate lymphoid cells (ILC2) are innate immune cells that respond rapidly to their environment through soluble inflammatory mediators and cell-to-cell interactions. As tissue-resident sentinels, ILC2 help orchestrate localized type 2 immune responses. These ILC2-driven type 2 responses are now recognized in diverse immune processes, different anatomical locations, and homeostatic or pathological settings. ILC2-derived cytokines and cell surface signaling molecules function as key regulators of innate and adaptive immunity. Conversely, ILC2 are governed by their environment. As such, ILC2 form an important nexus of the immune system and may present an attractive target for immune modulation in disease.
Collapse
|
36
|
Martin NM, Griffin DE. Interleukin-10 Modulation of Virus Clearance and Disease in Mice with Alphaviral Encephalomyelitis. J Virol 2018; 92:e01517-17. [PMID: 29263262 PMCID: PMC5827374 DOI: 10.1128/jvi.01517-17] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2017] [Accepted: 12/11/2017] [Indexed: 02/07/2023] Open
Abstract
Alphaviruses are an important cause of mosquito-borne outbreaks of arthritis, rash, and encephalomyelitis. Previous studies in mice with a virulent strain (neuroadapted SINV [NSV]) of the alphavirus Sindbis virus (SINV) identified a role for Th17 cells and regulation by interleukin-10 (IL-10) in the pathogenesis of fatal encephalomyelitis (K. A. Kulcsar, V. K. Baxter, I. P. Greene, and D. E. Griffin, Proc Natl Acad Sci U S A 111:16053-16058, 2014, https://doi.org/10.1073/pnas.1418966111). To determine the role of virus virulence in generation of immune responses, we analyzed the modulatory effects of IL-10 on disease severity, virus clearance, and the CD4+ T cell response to infection with a recombinant strain of SINV of intermediate virulence (TE12). The absence of IL-10 during TE12 infection led to longer morbidity, more weight loss, higher mortality, and slower viral clearance than in wild-type mice. More severe disease and impaired virus clearance in IL-10-/- mice were associated with more Th1 cells, fewer Th2 cells, innate lymphoid type 2 cells, regulatory cells, and B cells, and delayed production of antiviral antibody in the central nervous system (CNS) without an effect on Th17 cells. Therefore, IL-10 deficiency led to more severe disease in TE12-infected mice by increasing Th1 cells and by hampering development of the local B cell responses necessary for rapid production of antiviral antibody and virus clearance from the CNS. In addition, the shift from Th17 to Th1 responses with decreased virus virulence indicates that the effects of IL-10 deficiency on immunopathologic responses in the CNS during alphavirus infection are influenced by virus strain.IMPORTANCE Alphaviruses cause mosquito-borne outbreaks of encephalomyelitis, but determinants of outcome are incompletely understood. We analyzed the effects of the anti-inflammatory cytokine IL-10 on disease severity and virus clearance after infection with an alphavirus strain of intermediate virulence. The absence of IL-10 led to longer illness, more weight loss, more death, and slower viral clearance than in mice that produced IL-10. IL-10 influenced development of disease-causing T cells and entry into the brain of B cells producing antiviral antibody. The Th1 pathogenic cell subtype that developed in IL-10-deficient mice infected with a less virulent virus was distinct from the Th17 subtype that developed in response to a more virulent virus, indicating a role for virus strain in determining the immune response. Slow production of antibody in the nervous system led to delayed virus clearance. Therefore, both the virus strain and the host response to infection are important determinants of outcome.
Collapse
Affiliation(s)
- Nina M Martin
- W. Harry Feinstone Department of Molecular Microbiology and Immunology, Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland, USA
| | - Diane E Griffin
- W. Harry Feinstone Department of Molecular Microbiology and Immunology, Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland, USA
| |
Collapse
|
37
|
Soriani A, Stabile H, Gismondi A, Santoni A, Bernardini G. Chemokine regulation of innate lymphoid cell tissue distribution and function. Cytokine Growth Factor Rev 2018; 42:47-55. [PMID: 29472011 DOI: 10.1016/j.cytogfr.2018.02.003] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2018] [Revised: 02/09/2018] [Accepted: 02/09/2018] [Indexed: 12/14/2022]
Abstract
Three groups of innate lymphoid cells (ILCs) can be defined based on transcription factor requirements, cytokine production profiles, and roles in immunity. Given their strategic anatomical location into barrier tissues and the ability to rapidly produce cytokines and to cross-talk with other immune and non-immune cells, ILCs play fundamental functions in tissue homeostasis and regulation of immune responses. Several members of the chemokine family influence ILC tissue localization in the correct microenvironment by regulating their release from the bone marrow as well as their homing and retention in the tissues. In this review, we discuss the recent advances on how chemokine regulation of ILC tissue-positioning and functional interaction with other cells play essential roles in tissue-specific regulation of innate and adaptive immune responses.
Collapse
Affiliation(s)
- Alessandra Soriani
- Department of Molecular Medicine, Sapienza University of Rome, 00161-Rome, Italy
| | - Helena Stabile
- Department of Molecular Medicine, Sapienza University of Rome, 00161-Rome, Italy
| | - Angela Gismondi
- Department of Molecular Medicine, Sapienza University of Rome, 00161-Rome, Italy
| | - Angela Santoni
- Department of Molecular Medicine, Sapienza University of Rome, Laboratory Affiliated to Institute Pasteur -Italia, 00161-Rome, Italy; IRCCS, Neuromed, Pozzilli, 86077 IS, Italy
| | - Giovanni Bernardini
- Department of Molecular Medicine, Sapienza University of Rome, Laboratory Affiliated to Institute Pasteur -Italia, 00161-Rome, Italy; IRCCS, Neuromed, Pozzilli, 86077 IS, Italy.
| |
Collapse
|
38
|
Nausch N, Mutapi F. Group 2 ILCs: A way of enhancing immune protection against human helminths? Parasite Immunol 2018; 40:e12450. [PMID: 28626924 PMCID: PMC5811928 DOI: 10.1111/pim.12450] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2017] [Accepted: 06/12/2017] [Indexed: 12/13/2022]
Abstract
Group 2 innate lymphoid cells (ILC2s) play crucial roles in type 2 immune responses associated with allergic and autoimmune diseases, viral and helminth infections and tissue homoeostasis. Experimental models show that in helminth infections ILC2s provide an early source of type 2 cytokines and therefore are essential for the induction of potentially protective type 2 responses. Much of our knowledge of ILC2s in helminth infections has come from experimental mouse models with very few studies analysing ILC2s in natural human infections. In attempts to harness knowledge from paradigms of the development of protective immunity in human helminth infections for vaccine development, the role of ILC2 cells could be pivotal. So far, potential vaccines against human helminth infections have failed to provide effective protection when evaluated in human studies. In addition to appropriate antigen selection, it is apparent that more detailed knowledge on mechanisms of induction and maintenance of protective immune responses is required. Therefore, there is need to understand how ILC2 cells induce type 2 responses and subsequently support the development of a protective immune response in the context of immunizations. Within this review, we summarize the current knowledge of the biology of ILC2s, discuss the importance of ILC2s in human helminth infections and explore how ILC2 responses could be boosted to efficiently induce protective immunity.
Collapse
Affiliation(s)
- N. Nausch
- Pediatric Pneumology and Infectious Diseases Group, Department of General Pediatrics, Neonatology and Pediatric CardiologyUniversity Children's Hospital, Heinrich‐Heine‐University DuesseldorfDuesseldorfGermany
| | - F. Mutapi
- Institute of Immunology and Infection Research, Centre for Immunity, Infection and EvolutionSchool of Biological Sciences, University of EdinburghEdinburghUK
| |
Collapse
|
39
|
von Moltke J, Pepper M. Sentinels of the Type 2 Immune Response. Trends Immunol 2018; 39:99-111. [PMID: 29122456 PMCID: PMC6181126 DOI: 10.1016/j.it.2017.10.004] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2017] [Revised: 10/09/2017] [Accepted: 10/10/2017] [Indexed: 12/25/2022]
Abstract
Type 2 immune responses have evolved to sense and respond to large, non-replicating infections or non-microbial noxious compounds in tissues. The development of these responses therefore depends upon highly coordinated and tightly regulated tissue-residing cellular sensors and responders. Multiple exposure to type 2 helper T cell (Th2)-inducing stimuli further enhances both the diversity and potency of the response. This review discusses advances in our understanding of the interacting cellular subsets that comprise both primary and secondary type 2 responses. Current knowledge regarding type 2 immune responses in the lung are initially presented and are then contrasted with what is known about the small intestine. The studies described portray an immune response that depends upon well-organized tissue structures, and suggest their modulation as a therapeutic strategy.
Collapse
Affiliation(s)
- Jakob von Moltke
- Department of Immunology, University of Washington School of Medicine, 750 Republican Street, Seattle, WA 98109, USA
| | - Marion Pepper
- Department of Immunology, University of Washington School of Medicine, 750 Republican Street, Seattle, WA 98109, USA.
| |
Collapse
|
40
|
Male-specific IL-33 expression regulates sex-dimorphic EAE susceptibility. Proc Natl Acad Sci U S A 2018; 115:E1520-E1529. [PMID: 29378942 DOI: 10.1073/pnas.1710401115] [Citation(s) in RCA: 75] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
Abstract
The cellular and molecular basis of sex-dimorphic autoimmune diseases, such as the CNS demyelinating disease multiple sclerosis (MS), remains unclear. Our studies in the SJL mouse model of MS, experimental autoimmune encephalomyelitis (EAE), reveal that sex-determined differences in Il33 expression by innate immune cells in response to myelin peptide immunization regulate EAE susceptibility. IL-33 is selectively induced in PLP139-151-immunized males and activates type 2 innate lymphoid cells (ILC2s), cells that promote and sustain a nonpathogenic Th2 myelin-specific response. Without this attenuating IL-33 response, females generate an encephalitogenic Th17-dominant response, which can be reversed by IL-33 treatment. Mast cells are one source of IL-33 and we provide evidence that testosterone directly induces Il33 gene expression and also exerts effects on the potential for Il33 gene expression during mast cell development. Thus, in contrast to their pathogenic role in allergy, we propose a sex-specific role for both mast cells and ILC2s as attenuators of the pathogenic Th response in CNS inflammatory disease.
Collapse
|
41
|
Cosmi L, Annunziato F. Group 2 Innate Lymphoid Cells Are the Earliest Recruiters of Eosinophils in Lungs of Patients with Allergic Asthma. Am J Respir Crit Care Med 2017; 196:666-668. [PMID: 28475847 DOI: 10.1164/rccm.201704-0799ed] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Affiliation(s)
- Lorenzo Cosmi
- 1 Department of Experimental and Clinical Medicine University of Florence Florence, Italy
| | - Francesco Annunziato
- 1 Department of Experimental and Clinical Medicine University of Florence Florence, Italy
| |
Collapse
|
42
|
Li BWS, Stadhouders R, de Bruijn MJW, Lukkes M, Beerens DMJM, Brem MD, KleinJan A, Bergen I, Vroman H, Kool M, van IJcken WFJ, Rao TN, Fehling HJ, Hendriks RW. Group 2 Innate Lymphoid Cells Exhibit a Dynamic Phenotype in Allergic Airway Inflammation. Front Immunol 2017; 8:1684. [PMID: 29250067 PMCID: PMC5716969 DOI: 10.3389/fimmu.2017.01684] [Citation(s) in RCA: 46] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2017] [Accepted: 11/16/2017] [Indexed: 12/26/2022] Open
Abstract
Group 2 innate lymphoid cells (ILC2) are implicated in allergic asthma as an early innate source of the type 2 cytokines IL-5 and IL-13. However, their induction in house dust mite (HDM)-mediated airway inflammation additionally requires T cell activation. It is currently unknown whether phenotypic differences exist between ILC2s that are activated in a T cell-dependent or T cell-independent fashion. Here, we compared ILC2s in IL-33- and HDM-driven airway inflammation. Using flow cytometry, we found that surface expression levels of various markers frequently used to identify ILC2s were dependent on their mode of activation, highly variable over time, and differed between tissue compartments, including bronchoalveolar lavage (BAL) fluid, lung, draining lymph nodes, and spleen. Whereas in vivo IL-33-activated BAL fluid ILC2s exhibited an almost uniform CD25+CD127+T1/ST2+ICOS+KLRG1+ phenotype, at a comparable time point after HDM exposure BAL fluid ILC2s had a very heterogeneous surface marker phenotype. A major fraction of HDM-activated ILC2s were CD25lowCD127+T1/ST2low ICOSlowKLRG1low, but nevertheless had the capacity to produce large amounts of type 2 cytokines. HDM-activated CD25low ILC2s in BAL fluid and lung rapidly reverted to CD25high ILC2s upon in vivo stimulation with IL-33. Genome-wide transcriptional profiling of BAL ILC2s revealed ~1,600 differentially expressed genes: HDM-stimulated ILC2s specifically expressed genes involved in the regulation of adaptive immunity through B and T cell interactions, whereas IL-33-stimulated ILC2s expressed high levels of proliferation-related and cytokine genes. In both airway inflammation models ILC2s were present in the lung submucosa close to epithelial cells, as identified by confocal microscopy. In chronic HDM-driven airway inflammation ILC2s were also found inside organized cellular infiltrates near T cells. Collectively, our findings show that ILC2s are phenotypically more heterogeneous than previously thought, whereby their surface marker and gene expression profile are highly dynamic.
Collapse
Affiliation(s)
- Bobby W S Li
- Department of Pulmonary Medicine, Rotterdam, Netherlands
| | | | | | - Melanie Lukkes
- Department of Pulmonary Medicine, Rotterdam, Netherlands
| | | | - Maarten D Brem
- Department of Pulmonary Medicine, Rotterdam, Netherlands
| | - Alex KleinJan
- Department of Pulmonary Medicine, Rotterdam, Netherlands
| | - Ingrid Bergen
- Department of Pulmonary Medicine, Rotterdam, Netherlands
| | - Heleen Vroman
- Department of Pulmonary Medicine, Rotterdam, Netherlands
| | - Mirjam Kool
- Department of Pulmonary Medicine, Rotterdam, Netherlands
| | | | | | | | | |
Collapse
|
43
|
Arachidonic acid: Physiological roles and potential health benefits - A review. J Adv Res 2017; 11:33-41. [PMID: 30034874 PMCID: PMC6052655 DOI: 10.1016/j.jare.2017.11.004] [Citation(s) in RCA: 385] [Impact Index Per Article: 48.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2017] [Revised: 11/16/2017] [Accepted: 11/17/2017] [Indexed: 12/17/2022] Open
Abstract
It is time to shift the arachidonic acid (ARA) paradigm from a harm-generating molecule to its status of polyunsaturated fatty acid essential for normal health. ARA is an integral constituent of biological cell membrane, conferring it with fluidity and flexibility, so necessary for the function of all cells, especially in nervous system, skeletal muscle, and immune system. Arachidonic acid is obtained from food or by desaturation and chain elongation of the plant-rich essential fatty acid, linoleic acid. Free ARA modulates the function of ion channels, several receptors and enzymes, via activation as well as inhibition. That explains its fundamental role in the proper function of the brain and muscles and its protective potential against Schistosoma mansoni and S. haematobium infection and tumor initiation, development, and metastasis. Arachidonic acid in cell membranes undergoes reacylation/deacylation cycles, which keep the concentration of free ARA in cells at a very low level and limit ARA availability to oxidation. Metabolites derived from ARA oxidation do not initiate but contribute to inflammation and most importantly lead to the generation of mediators responsible for resolving inflammation and wound healing. Endocannabinoids are oxidation-independent ARA derivatives, critically important for brain reward signaling, motivational processes, emotion, stress responses, pain, and energy balance. Free ARA and metabolites promote and modulate type 2 immune responses, which are critically important in resistance to parasites and allergens insult, directly via action on eosinophils, basophils, and mast cells and indirectly by binding to specific receptors on innate lymphoid cells. In conclusion, the present review advocates the innumerable ARA roles and considerable importance for normal health.
Collapse
|
44
|
Pelaia C, Vatrella A, Lombardo N, Terracciano R, Navalesi P, Savino R, Pelaia G. Biological mechanisms underlying the clinical effects of allergen-specific immunotherapy in asthmatic children. Expert Opin Biol Ther 2017; 18:197-204. [PMID: 29113525 DOI: 10.1080/14712598.2018.1402003] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
INTRODUCTION Allergen-specific immunotherapy (AIT) is indicated for patients with allergic asthma and/or allergic rhinitis, and can be implemented by either subcutaneous injection (SCIT) or sublingual administration (SLIT). AIT reduces asthma symptoms, lowers the use of pharmacologic controller therapy, and decreases the need for rescue medications. SLIT appears to be safer than SCIT, but SCIT seems to be more efficacious and acts earlier in allergic asthmatic children. AREAS COVERED This review looks at the pathobiology of allergic asthma as well as the role of regulatory T and B cells in allergen tolerance. It also reviews the immunological mechanisms underlying the clinical effects induced by AIT in allergic asthmatic children. EXPERT OPINION AIT is very effective in allergic asthmatic children, who can significantly benefit from this particular type of immunotherapy in order to achieve a better control of their disease. AIT is also capable of modifying the natural history of allergic asthma. Furthermore, AIT can potentially represent a valuable therapeutic tool within the context of precision medicine, as recombinant allergen technology might allow the creation of targeted extracts able to be effective against specific proteins to which individual asthmatic children are allergic, thus helping to implement a personalized approach to treatment.
Collapse
Affiliation(s)
- Corrado Pelaia
- a Department of Medical and Surgical Sciences , University "Magna Græcia" of Catanzaro , Catanzaro , Italy
| | - Alessandro Vatrella
- b Department of Medicine, Surgery and Dentistry , University of Salerno , Salerno , Italy
| | - Nicola Lombardo
- a Department of Medical and Surgical Sciences , University "Magna Græcia" of Catanzaro , Catanzaro , Italy
| | - Rosa Terracciano
- c Department of Health Science , University "Magna Græcia" of Catanzaro , Catanzaro , Italy
| | - Paolo Navalesi
- a Department of Medical and Surgical Sciences , University "Magna Græcia" of Catanzaro , Catanzaro , Italy
| | - Rocco Savino
- c Department of Health Science , University "Magna Græcia" of Catanzaro , Catanzaro , Italy
| | - Girolamo Pelaia
- a Department of Medical and Surgical Sciences , University "Magna Græcia" of Catanzaro , Catanzaro , Italy
| |
Collapse
|
45
|
Symowski C, Voehringer D. Interactions between Innate Lymphoid Cells and Cells of the Innate and Adaptive Immune System. Front Immunol 2017; 8:1422. [PMID: 29163497 PMCID: PMC5670097 DOI: 10.3389/fimmu.2017.01422] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2017] [Accepted: 10/12/2017] [Indexed: 12/12/2022] Open
Abstract
Type 2 innate lymphoid cells (ILC2s) are a major source of cytokines, which are also produced by Th2 cells and several cell types of the innate immune system. Work over the past few years indicates that ILC2s play a central role in regulating type 2 immune responses against allergens and helminths. ILC2s can interact with a variety of cells types of the innate and adaptive immune system by cell–cell contacts or by communication via soluble factors. In this review, we provide an overview about recent advances in our understanding how ILC2s orchestrate type 2 immune responses with focus on direct interactions between ILC2s and other cells of the immune system.
Collapse
Affiliation(s)
- Cornelia Symowski
- Department of Infection Biology, University Hospital Erlangen and Friedrich-Alexander University Erlangen-Nuremberg (FAU), Erlangen, Germany
| | - David Voehringer
- Department of Infection Biology, University Hospital Erlangen and Friedrich-Alexander University Erlangen-Nuremberg (FAU), Erlangen, Germany
| |
Collapse
|
46
|
Kudo F, Ikutani M, Iseki M, Takaki S. Cyclosporin A indirectly attenuates activation of group 2 innate lymphoid cells in papain-induced lung inflammation. Cell Immunol 2017; 323:33-40. [PMID: 29108648 DOI: 10.1016/j.cellimm.2017.10.010] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2017] [Revised: 09/17/2017] [Accepted: 10/26/2017] [Indexed: 12/22/2022]
Abstract
Cyclosporin A (CsA) is a well-known immunosuppressant that is used against steroid-resistant asthma. Group 2 innate lymphoid cells (ILC2s) and type 2 helper T (Th2) cells produce Th2 cytokines including IL-5 and play important roles in asthma pathogenesis. Here, we studied the effects of CsA in allergen-induced lung inflammation in mice and found that CsA decreased the number of lung ILC2s and attenuated papain-induced activation of ILC2s accompanied with IL-5 expression. The ILC2 suppression mediated by CsA was not observed in culture or in lymphocyte-deficient Rag2-/- mice. Thus, we propose a new suppressive effect of CsA, i.e., administration of CsA indirectly suppresses maintenance and activation of lung ILC2s in addition to direct suppression of T-cell activation and cytokine production.
Collapse
Affiliation(s)
- Fujimi Kudo
- Department of Immune Regulation, Research Center for Hepatitis and Immunology, Research Institute, National Center for Global Health and Medicine, Chiba, Japan
| | - Masashi Ikutani
- Department of Immune Regulation, Research Center for Hepatitis and Immunology, Research Institute, National Center for Global Health and Medicine, Chiba, Japan
| | - Masanori Iseki
- Department of Immune Regulation, Research Center for Hepatitis and Immunology, Research Institute, National Center for Global Health and Medicine, Chiba, Japan
| | - Satoshi Takaki
- Department of Immune Regulation, Research Center for Hepatitis and Immunology, Research Institute, National Center for Global Health and Medicine, Chiba, Japan.
| |
Collapse
|
47
|
Kim J, Kim G, Min H. Pathological and therapeutic roles of innate lymphoid cells in diverse diseases. Arch Pharm Res 2017; 40:1249-1264. [PMID: 29032487 DOI: 10.1007/s12272-017-0974-2] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2017] [Accepted: 10/12/2017] [Indexed: 12/14/2022]
Abstract
Innate lymphoid cells (ILCs) are a recently defined type of innate-immunity cells that belong to the lymphoid lineage and have lymphoid morphology but do not express an antigen-specific B cell or T-cell receptor. ILCs regulate immune functions prior to the formation of adaptive immunity and exert effector functions through a cytokine release. ILCs have been classified into three groups according to the transcription factors that regulate their development and function and the effector cytokines they produce. Of note, ILCs resemble T helper (Th) cells, such as Th1, Th2, and Th17 cells, and show a similar dependence on transcription factors and distinct cytokine production. Despite their short history in immunology, ILCs have received much attention, and numerous studies have revealed biological functions of ILCs including host defense against pathogens, inflammation, tissue repair, and metabolic homeostasis. Here, we describe recent findings about the roles of ILCs in the pathogenesis of various diseases and potential therapeutic targets.
Collapse
Affiliation(s)
- Jisu Kim
- College of Pharmacy, Chung-Ang University, 84 Heukseokro, Dongjakgu, Seoul, 06974, Korea
| | - Geon Kim
- College of Pharmacy, Chung-Ang University, 84 Heukseokro, Dongjakgu, Seoul, 06974, Korea
| | - Hyeyoung Min
- College of Pharmacy, Chung-Ang University, 84 Heukseokro, Dongjakgu, Seoul, 06974, Korea.
| |
Collapse
|
48
|
Abstract
PURPOSE OF REVIEW The adaptive immune response orchestrated by type 2 T helper (Th2) lymphocytes, strictly cooperates with the innate response of group 2 innate lymphoid cells (ILC2), in the protection from helminths infection, as well as in the pathogenesis of allergic disease. The aim of this review is to explore the pathogenic role of ILC2 in different type 2-mediated disorders. RECENT FINDINGS Recent studies have shown that epithelial cell-derived cytokines and their responding cells, ILC2, play a pathogenic role in bronchial asthma, chronic rhinosinusitis, and atopic dermatitis. The growing evidences of the contribution of ILC2 in the induction and maintenance of allergic inflammation in such disease suggest the possibility to target them in therapy. Biological therapies blocking ILC2 activation or neutralizing their effector cytokines are currently under evaluation to be used in patients with type 2-dominated diseases.
Collapse
|
49
|
Innate lymphoid cells as regulators of immunity, inflammation and tissue homeostasis. Nat Immunol 2017; 17:765-74. [PMID: 27328006 DOI: 10.1038/ni.3489] [Citation(s) in RCA: 707] [Impact Index Per Article: 88.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2016] [Accepted: 05/10/2016] [Indexed: 12/14/2022]
Abstract
Research over the last 7 years has led to the formal identification of innate lymphoid cells (ILCs), increased the understanding of their tissue distribution and has established essential functions of ILCs in diverse physiological processes. These include resistance to pathogens, the regulation of autoimmune inflammation, tissue remodeling, cancer and metabolic homeostasis. Notably, many ILC functions appear to be regulated by mechanisms distinct from those of other innate and adaptive immune cells. In this Review, we focus on how group 2 ILC (ILC2) and group 3 ILC (ILC3) responses are regulated and how these cells interact with other immune and non-immune cells to mediate their functions. We highlight experimental evidence from mouse models and patient-based studies that have elucidated the effects of ILCs on the maintenance of tissue homeostasis and the consequences for health and disease.
Collapse
|
50
|
Vogel P, Janke L, Gravano DM, Lu M, Sawant DV, Bush D, Shuyu E, Vignali DAA, Pillai A, Rehg JE. Globule Leukocytes and Other Mast Cells in the Mouse Intestine. Vet Pathol 2017; 55:76-97. [PMID: 28494703 DOI: 10.1177/0300985817705174] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Only 2 major mast cell (MC) subtypes are commonly recognized in the mouse: the large connective tissue mast cells (CTMCs) and the mucosal mast cells (MMCs). Interepithelial mucosal inflammatory cells, most commonly identified as globule leukocytes (GLs), represent a third MC subtype in mice, which we term interepithelial mucosal mast cells (ieMMCs). This term clearly distinguishes ieMMCs from lamina proprial MMCs (lpMMCs) while clearly communicating their common MC lineage. Both lpMMCs and ieMMCs are rare in normal mouse intestinal mucosa, but increased numbers of ieMMCs are seen as part of type 2 immune responses to intestinal helminth infections and in food allergies. Interestingly, we found that increased ieMMCs were consistently associated with decreased mucosal inflammation and damage, suggesting that they might have a role in controlling helminth-induced immunopathology. We also found that ieMMC hyperplasia can develop in the absence of helminth infections, for example, in Treg-deficient mice, Arf null mice, some nude mice, and certain graft-vs-host responses. Since tuft cell hyperplasia plays a critical role in type 2 immune responses to intestinal helminths, we looked for (but did not find) any direct relationship between ieMMC and tuft cell numbers in the intestinal mucosa. Much remains to be learned about the differing functions of ieMMCs and lpMMCs in the intestinal mucosa, but an essential step in deciphering their roles in mucosal immune responses will be to apply immunohistochemistry methods to consistently and accurately identify them in tissue sections.
Collapse
Affiliation(s)
- Peter Vogel
- 1 Department of Pathology, St Jude Children's Research Hospital, Memphis, TN, USA
| | - Laura Janke
- 1 Department of Pathology, St Jude Children's Research Hospital, Memphis, TN, USA
| | | | - Meifen Lu
- 1 Department of Pathology, St Jude Children's Research Hospital, Memphis, TN, USA
| | - Deepali V Sawant
- 3 Department of Immunology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Dorothy Bush
- 1 Department of Pathology, St Jude Children's Research Hospital, Memphis, TN, USA
| | - E Shuyu
- 4 University of Miami School of Medicine, Miami, FL, USA
| | - Dario A A Vignali
- 3 Department of Immunology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Asha Pillai
- 4 University of Miami School of Medicine, Miami, FL, USA
| | - Jerold E Rehg
- 1 Department of Pathology, St Jude Children's Research Hospital, Memphis, TN, USA
| |
Collapse
|