51
|
The unique role of innate lymphoid cells in cancer and the hepatic microenvironment. Cell Mol Immunol 2022; 19:1012-1029. [PMID: 35962192 PMCID: PMC9424527 DOI: 10.1038/s41423-022-00901-1] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2022] [Accepted: 06/22/2022] [Indexed: 11/17/2022] Open
Abstract
Cancer is a complex disease, and despite incredible progress over the last decade, it remains the leading cause of death worldwide. Liver cancers, including hepatocellular carcinoma (HCC), and liver metastases are distinct from other cancers in that they typically emerge as a consequence of long-term low-grade inflammation. Understanding the mechanisms that underpin inflammation-driven tissue remodeling of the hepatic immune environment is likely to provide new insights into much needed treatments for this devastating disease. Group 1 innate lymphoid cells (ILCs), which include natural killer (NK) cells and ILC1s, are particularly enriched in the liver and thought to contribute to the pathogenesis of a number of liver diseases, including cancer. NK cells are an attractive, but underexplored, therapeutic target in hepatic disease due to their role in immunosurveillance and their ability to recognize and eliminate malignant cells. ILC1s are closely related to and share many phenotypic features with NK cells but are less well studied. Thus, their utility in immunotherapeutic approaches is not yet well understood. Here, we review our current understanding of ILCs in cancer with a particular focus on liver and liver-related diseases.
Collapse
|
52
|
Marques RF, de Melo FM, Novais JT, Soares IS, Bargieri DY, Gimenez AM. Immune System Modulation by the Adjuvants Poly (I:C) and Montanide ISA 720. Front Immunol 2022; 13:910022. [PMID: 35844531 PMCID: PMC9278660 DOI: 10.3389/fimmu.2022.910022] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2022] [Accepted: 05/31/2022] [Indexed: 11/13/2022] Open
Abstract
Adjuvants are essential for vaccine development, especially subunit-based vaccines such as those containing recombinant proteins. Increasing the knowledge of the immune response mechanisms generated by adjuvants should facilitate the formulation of vaccines in the future. The present work describes the immune phenotypes induced by Poly (I:C) and Montanide ISA 720 in the context of mice immunization with a recombinant protein based on the Plasmodium vivax circumsporozoite protein (PvCSP) sequence. Mice immunized with the recombinant protein plus Montanide ISA 720 showed an overall more robust humoral response, inducing antibodies with greater avidity to the antigen. A general trend for mixed Th1/Th2 inflammatory cytokine profile was increased in Montanide-adjuvanted mice, while a balanced profile was observed in Poly (I:C)-adjuvanted mice. Montanide ISA 720 induced a gene signature in B lymphocytes characteristic of heme biosynthesis, suggesting increased differentiation to Plasma Cells. On the other hand, Poly (I:C) provoked more perturbations in T cell transcriptome. These results extend the understanding of the modulation of specific immune responses induced by different classes of adjuvants, and could support the optimization of subunit-based vaccines.
Collapse
Affiliation(s)
- Rodolfo F. Marques
- Department of Parasitology, Institute of Biomedical Sciences, University of São Paulo, São Paulo, Brazil
| | - Filipe Menegatti de Melo
- Department of Parasitology, Institute of Biomedical Sciences, University of São Paulo, São Paulo, Brazil
| | - Janaina Tenório Novais
- Department of Clinical and Toxicological Analyses, School of Pharmaceutical Sciences, University of São Paulo, São Paulo, Brazil
| | - Irene S. Soares
- Department of Clinical and Toxicological Analyses, School of Pharmaceutical Sciences, University of São Paulo, São Paulo, Brazil
| | - Daniel Youssef Bargieri
- Department of Parasitology, Institute of Biomedical Sciences, University of São Paulo, São Paulo, Brazil
| | - Alba Marina Gimenez
- Department of Parasitology, Institute of Biomedical Sciences, University of São Paulo, São Paulo, Brazil
| |
Collapse
|
53
|
Favaro RR, Phillips K, Delaunay-Danguy R, Ujčič K, Markert UR. Emerging Concepts in Innate Lymphoid Cells, Memory, and Reproduction. Front Immunol 2022; 13:824263. [PMID: 35774779 PMCID: PMC9237338 DOI: 10.3389/fimmu.2022.824263] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2021] [Accepted: 04/15/2022] [Indexed: 12/28/2022] Open
Abstract
Members of the innate immune system, innate lymphoid cells (ILCs), encompass five major populations (Natural Killer (NK) cells, ILC1s, ILC2s, ILC3s, and lymphoid tissue inducer cells) whose functions include defense against pathogens, surveillance of tumorigenesis, and regulation of tissue homeostasis and remodeling. ILCs are present in the uterine environment of humans and mice and are dynamically regulated during the reproductive cycle and pregnancy. These cells have been repurposed to support pregnancy promoting maternal immune tolerance and placental development. To accomplish their tasks, immune cells employ several cellular and molecular mechanisms. They have the capacity to remember a previously encountered antigen and mount a more effective response to succeeding events. Memory responses are not an exclusive feature of the adaptive immune system, but also occur in innate immune cells. Innate immune memory has already been demonstrated in monocytes/macrophages, neutrophils, dendritic cells, and ILCs. A population of decidual NK cells characterized by elevated expression of NKG2C and LILRB1 as well as a distinctive transcriptional and epigenetic profile was found to expand during subsequent pregnancies in humans. These cells secrete high amounts of interferon-γ and vascular endothelial growth factor likely favoring placentation. Similarly, uterine ILC1s in mice upregulate CXCR6 and expand in second pregnancies. These data provide evidence on the development of immunological memory of pregnancy. In this article, the characteristics, functions, and localization of ILCs are reviewed, emphasizing available data on the uterine environment. Following, the concept of innate immune memory and its mechanisms, which include epigenetic changes and metabolic rewiring, are presented. Finally, the emerging role of innate immune memory on reproduction is discussed. Advances in the comprehension of ILC functions and innate immune memory may contribute to uncovering the immunological mechanisms underlying female fertility/infertility, placental development, and distinct outcomes in second pregnancies related to higher birth weight and lower incidence of complications.
Collapse
|
54
|
Sunaga S, Tsunoda J, Teratani T, Mikami Y, Kanai T. Heterogeneity of ILC2s in the Intestine; Homeostasis and Pathology. Front Immunol 2022; 13:867351. [PMID: 35707544 PMCID: PMC9190760 DOI: 10.3389/fimmu.2022.867351] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2022] [Accepted: 05/05/2022] [Indexed: 12/12/2022] Open
Abstract
Group 2 innate lymphoid cells (ILC2s) were identified in 2010 as a novel lymphocyte subset lacking antigen receptors, such as T-cell or B-cell receptors. ILC2s induce local immune responses characterized by producing type 2 cytokines and play essential roles for maintaining tissue homeostasis. ILC2s are distributed across various organs, including the intestine where immune cells are continuously exposed to external antigens. Followed by luminal antigen stimulation, intestinal epithelial cells produce alarmins, such as IL-25, IL-33, and thymic stromal lymphopoietin, and activate ILC2s to expand and produce cytokines. In the context of parasite infection, the tuft cell lining in the epithelium has been revealed as a dominant source of intestinal IL-25 and possesses the capability to regulate ILC2 homeostasis. Neuronal systems also regulate ILC2s through neuropeptides and neurotransmitters, and interact with ILC2s bidirectionally, a process termed “neuro-immune crosstalk”. Activated ILC2s produce type 2 cytokines, which contribute to epithelial barrier function, clearance of luminal antigens and tissue repair, while ILC2s are also involved in chronic inflammation and tissue fibrosis. Recent studies have shed light on the contribution of ILC2s to inflammatory bowel diseases, mainly comprising ulcerative colitis and Crohn’s disease, as defined by chronic immune activation and inflammation. Modern single-cell analysis techniques provide a tissue-specific picture of ILC2s and their roles in regulating homeostasis in each organ. Particularly, single-cell analysis helps our understanding of the uniqueness and commonness of ILC2s across tissues and opens the novel research area of ILC2 heterogeneity. ILC2s are classified into different phenotypes depending on tissue and phase of inflammation, mainly inflammatory and natural ILC2 cells. ILC2s can also switch phenotype to ILC1- or ILC3-like subsets. Hence, recent studies have revealed the heterogeneity and plasticity of ILC2, which indicate dynamicity of inflammation and the immune system. In this review, we describe the regulatory mechanisms, function, and pathological roles of ILC2s in the intestine.
Collapse
Affiliation(s)
- Shogo Sunaga
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, Keio University School of Medicine, Shinjuku-ku, Tokyo, Japan
| | - Junya Tsunoda
- Department of Surgery, Keio University School of Medicine, Tokyo, Japan
| | - Toshiaki Teratani
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, Keio University School of Medicine, Shinjuku-ku, Tokyo, Japan
| | - Yohei Mikami
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, Keio University School of Medicine, Shinjuku-ku, Tokyo, Japan
- *Correspondence: Yohei Mikami, ; Takanori Kanai,
| | - Takanori Kanai
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, Keio University School of Medicine, Shinjuku-ku, Tokyo, Japan
- AMED-CREST, Japan Agency for Medical Research and Development, Tokyo, Japan
- *Correspondence: Yohei Mikami, ; Takanori Kanai,
| |
Collapse
|
55
|
Olguín-Martínez E, Muñoz-Paleta O, Ruiz-Medina BE, Ramos-Balderas JL, Licona-Limón I, Licona-Limón P. IL-33 and the PKA Pathway Regulate ILC2 Populations Expressing IL-9 and ST2. Front Immunol 2022; 13:787713. [PMID: 35711429 PMCID: PMC9197159 DOI: 10.3389/fimmu.2022.787713] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2021] [Accepted: 04/12/2022] [Indexed: 11/13/2022] Open
Abstract
Type 2 Innate lymphoid cells (ILC2s) are tissue-resident immune cells activated by epithelial-derived alarmins upon tissue damage. They regulate immunity against helminth parasites and allergies by expressing type 2 immune response cytokines including IL-9, known to be critical for inducing and potentiating the immune response in such context. Although ILC2s are reported to be the main source of IL-9 in mice during N. brasiliensis infection, the mechanisms that regulate the expression of IL-9 in these cells are yet to be described. Recent studies have shown that in addition to cytokines, multiple molecules can differentially modulate the functions of ILC2s in various contexts both in vitro and in vivo. Among these stimuli are lipid mediators and neuropeptides, which activate the PKA pathway and have been associated with the regulation of type 2 immune cytokines. In this work we found that ILC2s in mice infected with N. brasiliensis can be classified into different groups based on the expression of IL-9 and ST2. These distinct populations were distributed in the lung and the small intestine. Through the development of an in vitro culture system, we sought to determine the stimuli that regulate the expression of these markers in ILC2s. We identified the alarmin IL-33 as being a key player for increased IL-9 expression. Additionally, we found the PKA pathway to be a dual regulator of ILC2 cells, working synergistically with IL-33 to enhance IL-9 production and capable of modulating proliferation and the expression of ILC2 markers. These data provide further evidence of a high heterogeneity between ILC2 subsets in a context dependent manner and calls for careful consideration when choosing the markers to identify these cells in vivo. Distinguishing ILC2 subsets and dissecting their mechanisms of activation is critical for a deeper understanding of the biology of these cells, allowing their manipulation for therapeutic purposes.
Collapse
Affiliation(s)
- Enrique Olguín-Martínez
- Departamento de Biología Celular y del Desarrollo, Instituto de Fisiología Celular, Universidad Nacional Autónoma de México, México City, Mexico
| | - Ofelia Muñoz-Paleta
- Departamento de Biología Celular y del Desarrollo, Instituto de Fisiología Celular, Universidad Nacional Autónoma de México, México City, Mexico
| | - Blanca E. Ruiz-Medina
- Departamento de Biología Celular y del Desarrollo, Instituto de Fisiología Celular, Universidad Nacional Autónoma de México, México City, Mexico
| | - Jose Luis Ramos-Balderas
- Departamento de Biología Celular y del Desarrollo, Instituto de Fisiología Celular, Universidad Nacional Autónoma de México, México City, Mexico
| | | | - Paula Licona-Limón
- Departamento de Biología Celular y del Desarrollo, Instituto de Fisiología Celular, Universidad Nacional Autónoma de México, México City, Mexico
- *Correspondence: Paula Licona-Limón,
| |
Collapse
|
56
|
Singh SS, Chauhan SB, Ng SSS, Corvino D, de Labastida Rivera F, Engel JA, Waddell N, Mukhopadhay P, Johnston RL, Koufariotis LT, Nylen S, Prakash Singh O, Engwerda CR, Kumar R, Sundar S. Increased amphiregulin expression by CD4 + T cells from individuals with asymptomatic Leishmania donovani infection. Clin Transl Immunology 2022; 11:e1396. [PMID: 35663920 PMCID: PMC9136704 DOI: 10.1002/cti2.1396] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2021] [Revised: 04/08/2022] [Accepted: 05/12/2022] [Indexed: 11/10/2022] Open
Abstract
Objectives There is an urgent need to be able to identify individuals with asymptomatic Leishmania donovani infection, so their risk of progressing to VL and transmitting parasites can be managed. This study examined transcriptional markers expressed by CD4+ T cells that could distinguish asymptomatic individuals from endemic controls and visceral leishmaniasis (VL) patients. Methods CD4+ T cells were isolated from individuals with asymptomatic L. donovani infection, endemic controls and VL patients. RNA was extracted and RNAseq employed to identify differentially expressed genes. The expression of one gene and its protein product during asymptomatic infection were evaluated. Results Amphiregulin (AREG) was identified as a distinguishing gene product in CD4+ T cells from individuals with asymptomatic L. donovani infection, compared to VL patients and healthy endemic control individuals. AREG levels in plasma and antigen-stimulated whole-blood assay cell culture supernatants were significantly elevated in asymptomatic individuals, compared to endemic controls and VL patients. Regulatory T (Treg) cells were identified as an important source of AREG amongst CD4+ T-cell subsets in asymptomatic individuals. Conclusion Increased Treg cell AREG expression was identified in individuals with asymptomatic L. donovani infection, suggesting the presence of an ongoing inflammatory response in these individuals required for controlling infection and that AREG may play an important role in preventing inflammation-induced tissue damage and subsequent disease in asymptomatic individuals.
Collapse
Affiliation(s)
- Siddharth Sankar Singh
- Department of Medicine, Institute of Medical SciencesBanaras Hindu UniversityVaranasiIndia
| | - Shashi Bhushan Chauhan
- Department of Medicine, Institute of Medical SciencesBanaras Hindu UniversityVaranasiIndia
| | - Susanna SS Ng
- QIMR Berghofer Medical Research InstituteBrisbaneAustralia
- Institute for Experimental OncologyUniversity of BonnBonnGermany
| | - Dillon Corvino
- QIMR Berghofer Medical Research InstituteBrisbaneAustralia
- Institute for Experimental OncologyUniversity of BonnBonnGermany
| | | | | | - Nic Waddell
- Department of Medicine, Institute of Medical SciencesBanaras Hindu UniversityVaranasiIndia
| | - Pamela Mukhopadhay
- Department of Medicine, Institute of Medical SciencesBanaras Hindu UniversityVaranasiIndia
| | - Rebecca L Johnston
- Department of Medicine, Institute of Medical SciencesBanaras Hindu UniversityVaranasiIndia
| | - Lambros T Koufariotis
- Department of Medicine, Institute of Medical SciencesBanaras Hindu UniversityVaranasiIndia
| | - Susanne Nylen
- Department of Microbiology, Tumor and Cell BiologyKarolinska InstituteStockholmSweden
| | | | | | - Rajiv Kumar
- Centre of Experimental Medicine and SurgeryInstitute of Medical SciencesBanaras Hindu UniversityVaranasiIndia
| | - Shyam Sundar
- Department of Medicine, Institute of Medical SciencesBanaras Hindu UniversityVaranasiIndia
| |
Collapse
|
57
|
Li Y, Lan F, Yang Y, Xu Y, Chen Y, Qin X, Lv Z, Wang W, Ying S, Zhang L. The absence of IL-9 reduces allergic airway inflammation by reducing ILC2, Th2 and mast cells in murine model of asthma. BMC Pulm Med 2022; 22:180. [PMID: 35524325 PMCID: PMC9074312 DOI: 10.1186/s12890-022-01976-2] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2022] [Accepted: 04/18/2022] [Indexed: 11/16/2022] Open
Abstract
Allergic asthma is an allergic inflammatory disease of the airways, in which numerous cell types and cytokines have been shown to contribute to pathogenesis of the disease. Although increased expression of IL-9 has been shown to influence the activity of structural as well as eosinophils and mast cells in asthma, the influence of IL-9 on function of ILC2 and Th2 cells remains unclear. This study therefore aimed to elucidate the role of IL-9 on ILC2 and Th2 cells using a murine model of asthma. A murine model of asthma was established using wild type (WT) and IL-9-deficient (Il9−/−) transgenic mice sensitized to house dust mite (HDM). Bronchoalveolar lavage fluid (BALF) and lung tissues were collected, and analysed for inflammatory cells (eosinophils, mast cells, Th2 cells and ILC2 cells), histopathological changes, and several cytokines. HDM challenge significantly increased accumulation of ILC2 cells, Th2 cells and mast cells, as well as goblet cell hyperplasia, and the expression of cytokines IL-4, IL-5 and IL-13, but not IFN-γ, in WT mice compared to saline-challenged control group. In contrast, all pathological changes, including infiltration of ILC2 cells, Th2 cells and mast cells, were significantly attenuated in HDM-challenged Il9−/− mice. Furthermore, the number of Ki67+ILC2 cells, Ki67+Th2 cells and Ki67+mast cells were significantly reduced in the absence of IL-9 signalling. These data suggest that IL-9 promotes the proliferation and type 2 cytokine production of type 2 cells in the murine models of asthma, and therefore might be a potential therapeutic target for asthma treatment.
Collapse
Affiliation(s)
- Yan Li
- Department of Otorhinolaryngology Head and Neck Surgery, Beijing Tongren Hospital, Capital Medical University, Beijing Institute of Otorhinolaryngology, Key Laboratory of Otorhinolaryngology Head and Neck Surgery, Ministry of Education, Beijing Key Laboratory of Nasal Diseases, No. 17, HouGouHuTong, DongCheng District, Beijing, 100730, China
| | - Feng Lan
- Department of Otorhinolaryngology Head and Neck Surgery, Beijing Tongren Hospital, Capital Medical University, Beijing Institute of Otorhinolaryngology, Key Laboratory of Otorhinolaryngology Head and Neck Surgery, Ministry of Education, Beijing Key Laboratory of Nasal Diseases, No. 17, HouGouHuTong, DongCheng District, Beijing, 100730, China
| | - Yiran Yang
- Department of Immunology, School of Basic Medical Sciences, Capital Medical University, 10 Xi Tou Tiao, You An Men Wai, Fengtai District, Beijing, 100069, China
| | - Yingjie Xu
- Department of Immunology, School of Basic Medical Sciences, Capital Medical University, 10 Xi Tou Tiao, You An Men Wai, Fengtai District, Beijing, 100069, China
| | - Yalin Chen
- Department of Thyroid Head and Neck Surgery, Qingdao Central Hospital, Qingdao University, Qingdao, China
| | - Xiaofeng Qin
- Department of Immunology, School of Basic Medical Sciences, Capital Medical University, 10 Xi Tou Tiao, You An Men Wai, Fengtai District, Beijing, 100069, China
| | - Zhe Lv
- Department of Immunology, School of Basic Medical Sciences, Capital Medical University, 10 Xi Tou Tiao, You An Men Wai, Fengtai District, Beijing, 100069, China
| | - Wei Wang
- Department of Immunology, School of Basic Medical Sciences, Capital Medical University, 10 Xi Tou Tiao, You An Men Wai, Fengtai District, Beijing, 100069, China
| | - Sun Ying
- Department of Immunology, School of Basic Medical Sciences, Capital Medical University, 10 Xi Tou Tiao, You An Men Wai, Fengtai District, Beijing, 100069, China.
| | - Luo Zhang
- Department of Otorhinolaryngology Head and Neck Surgery, Beijing Tongren Hospital, Capital Medical University, Beijing Institute of Otorhinolaryngology, Key Laboratory of Otorhinolaryngology Head and Neck Surgery, Ministry of Education, Beijing Key Laboratory of Nasal Diseases, No. 17, HouGouHuTong, DongCheng District, Beijing, 100730, China.
| |
Collapse
|
58
|
Sharma A, Noon JB, Kontodimas K, Garo LP, Platten J, Quinton LJ, Urban JF, Reinhardt C, Bosmann M. IL-27 Enhances γδ T Cell–Mediated Innate Resistance to Primary Hookworm Infection in the Lungs. THE JOURNAL OF IMMUNOLOGY 2022; 208:2008-2018. [PMID: 35354611 PMCID: PMC9012701 DOI: 10.4049/jimmunol.2000945] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/12/2020] [Accepted: 02/09/2022] [Indexed: 11/19/2022]
Abstract
IL-27 is a heterodimeric IL-12 family cytokine formed by noncovalent association of the promiscuous EBI3 subunit and selective p28 subunit. IL-27 is produced by mononuclear phagocytes and unfolds pleiotropic immune-modulatory functions through ligation to IL-27 receptor α (IL-27RA). Although IL-27 is known to contribute to immunity and to limit inflammation after various infections, its relevance for host defense against multicellular parasites is still poorly defined. Here, we investigated the role of IL-27 during infection with the soil-transmitted hookworm, Nippostrongylus brasiliensis, in its early host intrapulmonary life cycle. IL-27(p28) was detectable in bronchoalveolar lavage fluid of C57BL/6J wild-type mice on day 1 after s.c. inoculation. IL-27RA expression was most abundant on lung-invading γδ T cells. Il27ra-/- mice showed increased lung parasite burden together with aggravated pulmonary hemorrhage and higher alveolar total protein leakage as a surrogate for epithelial-vascular barrier disruption. Conversely, injections of recombinant mouse (rm)IL-27 into wild-type mice reduced lung injury and parasite burden. In multiplex screens, higher airway accumulations of IL-6, TNF-α, and MCP-3 (CCL7) were observed in Il27ra-/- mice, whereas rmIL-27 treatment showed a reciprocal effect. Importantly, γδ T cell numbers in airways were enhanced by endogenous or administered IL-27. Further analysis revealed a direct antihelminthic function of IL-27 on γδ T cells as adoptive intratracheal transfer of rmIL-27-treated γδ T cells during primary N. brasiliensis lung infection conferred protection in mice. In summary, this report demonstrates protective functions of IL-27 to control the early lung larval stage of hookworm infection.
Collapse
Affiliation(s)
- Arjun Sharma
- Pulmonary Center, Department of Medicine, Boston University School of Medicine, Boston, Massachusetts
- Center for Thrombosis and Hemostasis, University Medical Center of the Johannes Gutenberg University Mainz, Mainz, Germany
| | - Jason B Noon
- Pulmonary Center, Department of Medicine, Boston University School of Medicine, Boston, Massachusetts
| | - Konstantinos Kontodimas
- Pulmonary Center, Department of Medicine, Boston University School of Medicine, Boston, Massachusetts
| | - Lucien P Garo
- Pulmonary Center, Department of Medicine, Boston University School of Medicine, Boston, Massachusetts
| | - Johannes Platten
- Center for Thrombosis and Hemostasis, University Medical Center of the Johannes Gutenberg University Mainz, Mainz, Germany
| | - Lee J Quinton
- Pulmonary Center, Department of Medicine, Boston University School of Medicine, Boston, Massachusetts
- Division of Infectious Disease and Immunology, Department of Medicine, University of Massachusetts Chan Medical School, Worcester, MA
| | - Joseph F Urban
- Agricultural Research Service, Beltsville Agricultural Research Center, Animal Parasitic Diseases Laboratory and Beltsville Human Nutrition Research Center, Diet, Genomics, and Immunology Laboratory, U.S. Department of Agriculture, Beltsville, MD; and
| | - Christoph Reinhardt
- Center for Thrombosis and Hemostasis, University Medical Center of the Johannes Gutenberg University Mainz, Mainz, Germany
| | - Markus Bosmann
- Pulmonary Center, Department of Medicine, Boston University School of Medicine, Boston, Massachusetts;
- Center for Thrombosis and Hemostasis, University Medical Center of the Johannes Gutenberg University Mainz, Mainz, Germany
- Research Center for Immunotherapy, University Medical Center of the Johannes Gutenberg-University Mainz, Mainz, Germany
| |
Collapse
|
59
|
Fu Y, Wang J, Zhou B, Pajulas A, Gao H, Ramdas B, Koh B, Ulrich BJ, Yang S, Kapur R, Renauld JC, Paczesny S, Liu Y, Tighe RM, Licona-Limón P, Flavell RA, Takatsuka S, Kitamura D, Tepper RS, Sun J, Kaplan MH. An IL-9-pulmonary macrophage axis defines the allergic lung inflammatory environment. Sci Immunol 2022; 7:eabi9768. [PMID: 35179949 PMCID: PMC8991419 DOI: 10.1126/sciimmunol.abi9768] [Citation(s) in RCA: 48] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Despite IL-9 functioning as a pleiotropic cytokine in mucosal environments, the IL-9-responsive cell repertoire is still not well defined. Here, we found that IL-9 mediates proallergic activities in the lungs by targeting lung macrophages. IL-9 inhibits alveolar macrophage expansion and promotes recruitment of monocytes that develop into CD11c+ and CD11c- interstitial macrophage populations. Interstitial macrophages were required for IL-9-dependent allergic responses. Mechanistically, IL-9 affected the function of lung macrophages by inducing Arg1 activity. Compared with Arg1-deficient lung macrophages, Arg1-expressing macrophages expressed greater amounts of CCL5. Adoptive transfer of Arg1+ lung macrophages but not Arg1- lung macrophages promoted allergic inflammation that Il9r-/- mice were protected against. In parallel, the elevated expression of IL-9, IL-9R, Arg1, and CCL5 was correlated with disease in patients with asthma. Thus, our study uncovers an IL-9/macrophage/Arg1 axis as a potential therapeutic target for allergic airway inflammation.
Collapse
Affiliation(s)
- Yongyao Fu
- Department of Microbiology and Immunology, Indiana University School of Medicine, Indianapolis, IN, 46202, USA
| | - Jocelyn Wang
- Department of Microbiology and Immunology, Indiana University School of Medicine, Indianapolis, IN, 46202, USA
| | - Baohua Zhou
- Department of Pediatrics and Herman B Wells Center for Pediatric Research, Indiana University School of Medicine, Indianapolis, IN, 46202, USA
| | - Abigail Pajulas
- Department of Microbiology and Immunology, Indiana University School of Medicine, Indianapolis, IN, 46202, USA
| | - Hongyu Gao
- Department of Medical and Molecular Genetics, Indiana University School of Medicine, Indianapolis, IN, 46202, USA
| | - Baskar Ramdas
- Department of Pediatrics and Herman B Wells Center for Pediatric Research, Indiana University School of Medicine, Indianapolis, IN, 46202, USA
| | - Byunghee Koh
- Department of Microbiology and Immunology, Indiana University School of Medicine, Indianapolis, IN, 46202, USA
| | - Benjamin J Ulrich
- Department of Microbiology and Immunology, Indiana University School of Medicine, Indianapolis, IN, 46202, USA
| | - Shuangshuang Yang
- Department of Microbiology and Immunology, Indiana University School of Medicine, Indianapolis, IN, 46202, USA
| | - Reuben Kapur
- Department of Pediatrics and Herman B Wells Center for Pediatric Research, Indiana University School of Medicine, Indianapolis, IN, 46202, USA
| | - Jean-Christophe Renauld
- Ludwig Institute for Cancer Research, Experimental Medicine Unit, Université Catholique de Louvain, Brussels, 1200 Belgium
| | - Sophie Paczesny
- Department of Microbiology and Immunology, Medical University of South Carolina, 171 Ashley Avenue, Charleston, SC 29425
| | - Yunlong Liu
- Department of Medical and Molecular Genetics, Indiana University School of Medicine, Indianapolis, IN, 46202, USA
| | - Robert M Tighe
- Division of Pulmonary, Allergy, and Critical Care Medicine, Duke University Medical Center, Durham, NC 27710, United States
| | - Paula Licona-Limón
- Departamento de Biologia Celular y del Desarrollo, Instituto de Fisiologia Celular, Universidad Nacional Autónoma de México, 04510, Mexico City, Mexico
| | - Richard A. Flavell
- Department of Immunobiology, Yale University School of Medicine, New Haven, CT 06510, USA
| | - Shogo Takatsuka
- Division of Molecular Biology, Research Institute for Biomedical Sciences (RIBS), Tokyo University of Science, Noda, Japan
| | - Daisuke Kitamura
- Division of Molecular Biology, Research Institute for Biomedical Sciences (RIBS), Tokyo University of Science, Noda, Japan
| | - Robert S. Tepper
- Department of Pediatrics and Herman B Wells Center for Pediatric Research, Indiana University School of Medicine, Indianapolis, IN, 46202, USA
| | - Jie Sun
- Department of Medicine, Mayo Clinic, Rochester, MN 55905, USA
| | - Mark H Kaplan
- Department of Microbiology and Immunology, Indiana University School of Medicine, Indianapolis, IN, 46202, USA
| |
Collapse
|
60
|
Cairo C, Webb TJ. Effective Barriers: The Role of NKT Cells and Innate Lymphoid Cells in the Gut. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2022; 208:235-246. [PMID: 35017213 DOI: 10.4049/jimmunol.2100799] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/13/2021] [Accepted: 10/19/2021] [Indexed: 06/14/2023]
Abstract
The critical role of commensal microbiota in regulating the host immune response has been established. In addition, it is known that host-microbial interactions are bidirectional, and this interplay is tightly regulated to prevent chronic inflammatory disease. Although many studies have focused on the role of classic T cell subsets, unconventional lymphocytes such as NKT cells and innate lymphoid cells also contribute to the regulation of homeostasis at mucosal surfaces and influence the composition of the intestinal microbiota. In this review, we discuss the mechanisms involved in the cross-regulation between NKT cells, innate lymphoid cells, and the gut microbiota. Moreover, we highlight how disruptions in homeostasis can lead to immune-mediated disorders.
Collapse
Affiliation(s)
- Cristiana Cairo
- Department of Medicine, University of Maryland School of Medicine, Baltimore, MD;
- Institute of Human Virology, University of Maryland School of Medicine, Baltimore, MD
| | - Tonya J Webb
- Department of Microbiology and Immunology, University of Maryland School of Medicine, Baltimore, MD; and
- Marlene and Stewart Greenebaum Comprehensive Cancer Center, University of Maryland School of Medicine, Baltimore, MD
| |
Collapse
|
61
|
Wu X, Kasmani MY, Zheng S, Khatun A, Chen Y, Winkler W, Zander R, Burns R, Taparowsky EJ, Sun J, Cui W. BATF promotes group 2 innate lymphoid cell-mediated lung tissue protection during acute respiratory virus infection. Sci Immunol 2022; 7:eabc9934. [PMID: 35030033 DOI: 10.1126/sciimmunol.abc9934] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023]
Abstract
[Figure: see text].
Collapse
Affiliation(s)
- Xiaopeng Wu
- Blood Research Institute, Versiti Wisconsin, Milwaukee, WI 53213, USA
| | - Moujtaba Y Kasmani
- Blood Research Institute, Versiti Wisconsin, Milwaukee, WI 53213, USA.,Department of Microbiology and Immunology, Medical College of Wisconsin, Milwaukee, WI 53226, USA
| | - Shikan Zheng
- Blood Research Institute, Versiti Wisconsin, Milwaukee, WI 53213, USA
| | - Achia Khatun
- Blood Research Institute, Versiti Wisconsin, Milwaukee, WI 53213, USA.,Department of Microbiology and Immunology, Medical College of Wisconsin, Milwaukee, WI 53226, USA
| | - Yao Chen
- Blood Research Institute, Versiti Wisconsin, Milwaukee, WI 53213, USA.,Department of Microbiology and Immunology, Medical College of Wisconsin, Milwaukee, WI 53226, USA
| | - Wendy Winkler
- Blood Research Institute, Versiti Wisconsin, Milwaukee, WI 53213, USA
| | - Ryan Zander
- Blood Research Institute, Versiti Wisconsin, Milwaukee, WI 53213, USA
| | - Robert Burns
- Blood Research Institute, Versiti Wisconsin, Milwaukee, WI 53213, USA
| | - Elizabeth J Taparowsky
- Department of Biological Sciences, Purdue University Center for Cancer Research, Purdue University, West Lafayette, IN 47907, USA
| | - Jie Sun
- Department of Medicine, Mayo Clinic, Rochester, MN 55905, USA.,Department of Immunology, Mayo Clinic, Rochester, MN 55905, USA
| | - Weiguo Cui
- Blood Research Institute, Versiti Wisconsin, Milwaukee, WI 53213, USA.,Department of Microbiology and Immunology, Medical College of Wisconsin, Milwaukee, WI 53226, USA
| |
Collapse
|
62
|
Michla M, Wilhelm C. Food for thought - ILC metabolism in the context of helminth infections. Mucosal Immunol 2022; 15:1234-1242. [PMID: 36045216 PMCID: PMC9705246 DOI: 10.1038/s41385-022-00559-y] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2022] [Revised: 08/09/2022] [Accepted: 08/09/2022] [Indexed: 02/04/2023]
Abstract
Helminths are multicellular ancient organisms residing as parasites at mucosal surfaces of their host. Through adaptation and co-evolution with their hosts, helminths have been able to develop tolerance mechanisms to limit inflammation and avoid expulsion. The study of helminth infections as an integral part of tissue immunology allowed us to understand fundamental aspects of mucosal and barrier immunology, which led to the discovery of a new group of tissue-resident immune cells, innate lymphoid cells (ILC), over a decade ago. Here, we review the intricate interplay between helminth infections and type 2 ILC (ILC2) biology, discuss the host metabolic adaptation to helminth infections and the metabolic pathways fueling ILC2 responses. We hypothesize that nutrient competition between host and helminths may have prevented chronic inflammation in the past and argue that a detailed understanding of the metabolic restraints imposed by helminth infections may offer new therapeutic avenues in the future.
Collapse
Affiliation(s)
- Marcel Michla
- grid.10388.320000 0001 2240 3300Unit for Immunopathology, Department of Clinical Chemistry and Clinical Pharmacology, University Hospital Bonn, University of Bonn, 53127 Bonn, Germany
| | - Christoph Wilhelm
- grid.10388.320000 0001 2240 3300Unit for Immunopathology, Department of Clinical Chemistry and Clinical Pharmacology, University Hospital Bonn, University of Bonn, 53127 Bonn, Germany
| |
Collapse
|
63
|
Abstract
Synthetic receptor signalling has the potential to endow adoptively transferred T cells with new functions that overcome major barriers in the treatment of solid tumours, including the need for conditioning chemotherapy1,2. Here we designed chimeric receptors that have an orthogonal IL-2 receptor extracellular domain (ECD) fused with the intracellular domain (ICD) of receptors for common γ-chain (γc) cytokines IL-4, IL-7, IL-9 and IL-21 such that the orthogonal IL-2 cytokine elicits the corresponding γc cytokine signal. Of these, T cells that signal through the chimeric orthogonal IL-2Rβ-ECD-IL-9R-ICD (o9R) are distinguished by the concomitant activation of STAT1, STAT3 and STAT5 and assume characteristics of stem cell memory and effector T cells. Compared to o2R T cells, o9R T cells have superior anti-tumour efficacy in two recalcitrant syngeneic mouse solid tumour models of melanoma and pancreatic cancer and are effective even in the absence of conditioning lymphodepletion. Therefore, by repurposing IL-9R signalling using a chimeric orthogonal cytokine receptor, T cells gain new functions, and this results in improved anti-tumour activity for hard-to-treat solid tumours.
Collapse
|
64
|
IL-33: A central cytokine in helminth infections. Semin Immunol 2021; 53:101532. [PMID: 34823996 DOI: 10.1016/j.smim.2021.101532] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/13/2021] [Revised: 11/10/2021] [Accepted: 11/12/2021] [Indexed: 02/06/2023]
Abstract
IL-33 is an alarmin cytokine which has been implicated in allergy, fibrosis, inflammation, tumorigenesis, metabolism, and homeostasis. However, amongst its strongest roles are in helminth infections, where IL-33 usually (but not always) is central to induction of an effective anti-parasitic immune response. In this review, we will summarise the literature around this fascinating cytokine, its activity on immune and non-immune cells, the unique (and sometimes counterintuitive) responses it induces, and how it can coordinate the immune response during infections by parasitic helminths. Finally, we will summarise some of the ways that parasites have developed to modulate the IL-33 pathway for their own benefit.
Collapse
|
65
|
Popple SJ, Burrows K, Mortha A, Osborne LC. Remote regulation of type 2 immunity by intestinal parasites. Semin Immunol 2021; 53:101530. [PMID: 34802872 DOI: 10.1016/j.smim.2021.101530] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/24/2021] [Revised: 11/10/2021] [Accepted: 11/12/2021] [Indexed: 02/06/2023]
Abstract
The intestinal tract is the target organ of most parasitic infections, including those by helminths and protozoa. These parasites elicit prototypical type 2 immune activation in the host's immune system with striking impact on the local tissue microenvironment. Despite local containment of these parasites within the intestinal tract, parasitic infections also mediate immune adaptation in peripheral organs. In this review, we summarize the current knowledge on how such gut-tissue axes influence important immune-mediated resistance and disease tolerance in the context of coinfections, and elaborate on the implications of parasite-regulated gut-lung and gut-brain axes on the development and severity of airway inflammation and central nervous system diseases.
Collapse
Affiliation(s)
- S J Popple
- Department of Microbiology & Immunology, Life Sciences Institute, University of British Columbia, Vancouver, BC, Canada
| | - K Burrows
- Department of Immunology, University of Toronto, Toronto, ON, Canada
| | - A Mortha
- Department of Immunology, University of Toronto, Toronto, ON, Canada
| | - L C Osborne
- Department of Microbiology & Immunology, Life Sciences Institute, University of British Columbia, Vancouver, BC, Canada.
| |
Collapse
|
66
|
Mindt BC, Krisna SS, Duerr CU, Mancini M, Richer L, Vidal SM, Gerondakis S, Langlais D, Fritz JH. The NF-κB Transcription Factor c-Rel Modulates Group 2 Innate Lymphoid Cell Effector Functions and Drives Allergic Airway Inflammation. Front Immunol 2021; 12:664218. [PMID: 34867937 PMCID: PMC8635195 DOI: 10.3389/fimmu.2021.664218] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2021] [Accepted: 08/27/2021] [Indexed: 01/03/2023] Open
Abstract
Group 2 innate lymphoid cells (ILC2s) play a key role in the initiation and orchestration of early type 2 immune responses. Upon tissue damage, ILC2s are activated by alarmins such as IL-33 and rapidly secrete large amounts of type 2 signature cytokines. ILC2 activation is governed by a network of transcriptional regulators including nuclear factor (NF)-κB family transcription factors. While it is known that activating IL-33 receptor signaling results in downstream NF-κB activation, the underlying molecular mechanisms remain elusive. Here, we found that the NF-κB subunit c-Rel is required to mount effective innate pulmonary type 2 immune responses. IL-33-mediated activation of ILC2s in vitro as well as in vivo was found to induce c-Rel mRNA and protein expression. In addition, we demonstrate that IL-33-mediated activation of ILC2s leads to nuclear translocation of c-Rel in pulmonary ILC2s. Although c-Rel was found to be a critical mediator of innate pulmonary type 2 immune responses, ILC2-intrinsic deficiency of c-Rel did not have an impact on the developmental capacity of ILC2s nor affected homeostatic numbers of lung-resident ILC2s at steady state. Moreover, we demonstrate that ILC2-intrinsic deficiency of c-Rel alters the capacity of ILC2s to upregulate the expression of ICOSL and OX40L, key stimulatory receptors, and the expression of type 2 signature cytokines IL-5, IL-9, IL-13, and granulocyte-macrophage colony-stimulating factor (GM-CSF). Collectively, our data using Rel−/− mice suggest that c-Rel promotes acute ILC2-driven allergic airway inflammation and suggest that c-Rel may contribute to the pathophysiology of ILC2-mediated allergic airway disease. It thereby represents a promising target for the treatment of allergic asthma, and evaluating the effect of established c-Rel inhibitors in this context would be of great clinical interest.
Collapse
Affiliation(s)
- Barbara C. Mindt
- McGill University Research Centre on Complex Traits (MRCCT), Montréal, QC, Canada
- Department of Microbiology and Immunology, McGill University, Montréal, QC, Canada
| | - Sai Sakktee Krisna
- McGill University Research Centre on Complex Traits (MRCCT), Montréal, QC, Canada
- Department of Physiology, McGill University, Montréal, QC, Canada
| | - Claudia U. Duerr
- Charité – Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Department of Microbiology, Infectious Diseases and Immunology, Berlin, Germany
| | - Mathieu Mancini
- McGill University Research Centre on Complex Traits (MRCCT), Montréal, QC, Canada
- Department of Human Genetics, McGill University, Montréal, QC, Canada
| | - Lara Richer
- Department of Pathology, McGill University, Montréal, QC, Canada
| | - Silvia M. Vidal
- McGill University Research Centre on Complex Traits (MRCCT), Montréal, QC, Canada
- Department of Human Genetics, McGill University, Montréal, QC, Canada
| | - Steven Gerondakis
- Biomedicine Discovery Institute and Department of Biochemistry and Molecular Biology, Monash University, Clayton, VIC, Australia
| | - David Langlais
- McGill University Research Centre on Complex Traits (MRCCT), Montréal, QC, Canada
- Department of Human Genetics, McGill University, Montréal, QC, Canada
- McGill University Genome Centre, Montreal, QC, Canada
| | - Jörg H. Fritz
- McGill University Research Centre on Complex Traits (MRCCT), Montréal, QC, Canada
- Department of Microbiology and Immunology, McGill University, Montréal, QC, Canada
- Department of Physiology, McGill University, Montréal, QC, Canada
- FOCiS Centre of Excellence in Translational Immunology (CETI), Montréal, QC, Canada
- *Correspondence: Jörg H. Fritz,
| |
Collapse
|
67
|
Olguín-Martínez E, Ruiz-Medina BE, Licona-Limón P. Tissue-Specific Molecular Markers and Heterogeneity in Type 2 Innate Lymphoid Cells. Front Immunol 2021; 12:757967. [PMID: 34759931 PMCID: PMC8573327 DOI: 10.3389/fimmu.2021.757967] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2021] [Accepted: 09/21/2021] [Indexed: 12/25/2022] Open
Abstract
Innate lymphoid cells (ILCs) are the most recently described group of lymphoid subpopulations. These tissue-resident cells display a heterogeneity resembling that observed on different groups of T cells, hence their categorization as cytotoxic NK cells and helper ILCs type 1, 2 and 3. Each one of these groups is highly diverse and expresses different markers in a context-dependent manner. Type 2 innate lymphoid cells (ILC2s) are activated in response to helminth parasites and regulate the immune response. They are involved in the etiology of diseases associated with allergic responses as well as in the maintenance of tissue homeostasis. Markers associated with their identification differ depending on the tissue and model used, making the study and understanding of these cells a cumbersome task. This review compiles evidence for the heterogeneity of ILC2s as well as discussion and analyses of molecular markers associated with their identity, function, tissue-dependent expression, and how these markers contribute to the interaction of ILC2s with specific microenvironments to maintain homeostasis or respond to pathogenic challenges.
Collapse
Affiliation(s)
- Enrique Olguín-Martínez
- Departamento de Biología Celular y del Desarrollo, Instituto de Fisiología Celular, Universidad Nacional Autónoma de México, México City, Mexico
| | - Blanca E Ruiz-Medina
- Departamento de Biología Celular y del Desarrollo, Instituto de Fisiología Celular, Universidad Nacional Autónoma de México, México City, Mexico
| | - Paula Licona-Limón
- Departamento de Biología Celular y del Desarrollo, Instituto de Fisiología Celular, Universidad Nacional Autónoma de México, México City, Mexico
| |
Collapse
|
68
|
Orimo K, Tamari M, Saito H, Matsumoto K, Nakae S, Morita H. Characteristics of tissue-resident ILCs and their potential as therapeutic targets in mucosal and skin inflammatory diseases. Allergy 2021; 76:3332-3348. [PMID: 33866593 DOI: 10.1111/all.14863] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2020] [Revised: 03/30/2021] [Accepted: 04/12/2021] [Indexed: 12/11/2022]
Abstract
Discovery of innate lymphoid cells (ILCs), which are non-T and non-B lymphocytes that have no antigen-specific receptors, changed the classical concept of the mechanism of allergy, which had been explained mainly as antigen-specific acquired immunity based on IgE and Th2 cells. The discovery led to dramatic improvement in our understanding of the mechanism of non-IgE-mediated allergic inflammation. Numerous studies conducted in the past decade have elucidated the characteristics of each ILC subset in various organs and tissues and their ontogeny. We now know that each ILC subset exhibits heterogeneity. Moreover, the functions and activating/suppressing factors of each ILC subset were found to differ among both organs and types of tissue. Therefore, in this review, we summarize our current knowledge of ILCs by focusing on the organ/tissue-specific features of each subset to understand their roles in various organs. We also discuss ILCs' involvement in human inflammatory diseases in various organs and potential therapeutic/preventive strategies that target ILCs.
Collapse
Affiliation(s)
- Keisuke Orimo
- Department of Allergy and Clinical Immunology National Research Institute for Child Health and Development Tokyo Japan
| | - Masato Tamari
- Department of Allergy and Clinical Immunology National Research Institute for Child Health and Development Tokyo Japan
| | - Hirohisa Saito
- Department of Allergy and Clinical Immunology National Research Institute for Child Health and Development Tokyo Japan
| | - Kenji Matsumoto
- Department of Allergy and Clinical Immunology National Research Institute for Child Health and Development Tokyo Japan
| | - Susumu Nakae
- Graduate School of Integrated Sciences for Life Hiroshima University Hiroshima Japan
- Precursory Research for Embryonic Science and Technology Japan Science and Technology Agency Saitama Japan
| | - Hideaki Morita
- Department of Allergy and Clinical Immunology National Research Institute for Child Health and Development Tokyo Japan
| |
Collapse
|
69
|
Singh SS, Chauhan SB, Kumar A, Kumar S, Engwerda CR, Sundar S, Kumar R. Amphiregulin in cellular physiology, health, and disease: Potential use as a biomarker and therapeutic target. J Cell Physiol 2021; 237:1143-1156. [PMID: 34698381 DOI: 10.1002/jcp.30615] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2021] [Revised: 10/02/2021] [Accepted: 10/06/2021] [Indexed: 12/18/2022]
Abstract
Amphiregulin (AREG), which acts as one of the ligands for epidermal receptor growth factor receptor (EGFR), plays a crucial role in tissue repair, inflammation, and immunity. AREG is synthesized as membrane-anchored pre-protein, and is excreted after proteolytic cleavage, and serves as an autocrine or paracrine factor. After engagement with the EGFR, AREG triggers a cascade of signaling events required for many cellular physiological processes including metabolism, cell cycle, and proliferation. Under different inflammatory and pathogenic conditions, AREG is expressed by various activated immune cells that orchestrate both tolerance and host resistance mechanisms. Several factors including xenobiotics, cytokines, and inflammatory lipids have been shown to trigger AREG gene expression and release. In this review, we discuss the structure, function, and regulation of AREG, its role in tissue repair, inflammation, and homeostasis as well as the potential of AREG as a biomarker and therapeutic target.
Collapse
Affiliation(s)
- Siddharth S Singh
- Department of Medicine, Institute of Medical Sciences, Banaras Hindu University, Varanasi, India
| | - Shashi B Chauhan
- Department of Medicine, Institute of Medical Sciences, Banaras Hindu University, Varanasi, India
| | - Awnish Kumar
- Centre of Experimental Medicine & Surgery, Institute of Medical Sciences, Banaras Hindu University, Varanasi, India
| | - Shashi Kumar
- Department of Medicine, Institute of Medical Sciences, Banaras Hindu University, Varanasi, India
| | - Christian R Engwerda
- Department of Immunology, QIMR Berghofer Medical Research Institute, Brisbane, Australia
| | - Shyam Sundar
- Department of Medicine, Institute of Medical Sciences, Banaras Hindu University, Varanasi, India
| | - Rajiv Kumar
- Centre of Experimental Medicine & Surgery, Institute of Medical Sciences, Banaras Hindu University, Varanasi, India
| |
Collapse
|
70
|
Daniels JR, Ma JZ, Cao Z, Beger RD, Sun J, Schnackenberg L, Pence L, Choudhury D, Palevsky PM, Portilla D, Yu LR. Discovery of Novel Proteomic Biomarkers for the Prediction of Kidney Recovery from Dialysis-Dependent AKI Patients. KIDNEY360 2021; 2:1716-1727. [PMID: 34913041 PMCID: PMC8670726 DOI: 10.34067/kid.0002642021] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
BACKGROUND AKI requiring dialysis (AKI-D) is associated with prolonged hospitalization, mortality, and progressive CKD among survivors. Previous studies have examined only select urine or serum biomarkers for predicting kidney recovery from AKI. METHODS Serum samples collected on day 8 of randomized RRT from 72 patients enrolled in the Veteran's Affairs/National Institutes of Health Acute Renal Failure Trial Network study were analyzed by the SOMAscan proteomic platform to profile 1305 proteins in each sample. Of these patients, 38 recovered kidney function and dialysis was discontinued, whereas another 34 patients remained on dialysis by day 28. RESULTS Differential serum levels of 119 proteins, with 53 higher and 66 lower, were detected in samples from patients who discontinued dialysis, compared with patients who remained on dialysis by day 28. Patients were classified into tertiles on the basis of SOMAscan protein measurements for the 25 proteins most differentially expressed. The association of serum levels of each protein with kidney recovery was further evaluated using logistic regression analysis. Higher serum levels of CXCL11, CXCL2/CXCL3, CD86, Wnt-7a, BTK, c-Myc, TIMP-3, CCL5, ghrelin, PDGF-C, survivin, CA2, IL-9, EGF, and neuregulin-1, and lower levels of soluble CXCL16, IL1RL1, stanniocalcin-1, IL-6, and FGF23 when classified in tertiles were significantly associated with better kidney recovery. This significant association persisted for each of these proteins after adjusting for potential confounding risk factors including age, sex, cardiovascular SOFA score, congestive heart failure, diabetes, modality of intensive dialysis treatment, cause of AKI, baseline serum creatinine, day 8 urine volume, and estimated 60-day mortality risk. CONCLUSIONS These results suggest concerted changes between survival-related proteins and immune-regulatory chemokines in regulating angiogenesis, endothelial and epithelial remodeling, and kidney cell regeneration, illustrating potential mechanisms of kidney recovery. Thus, this study identifies potential novel predictive biomarkers of kidney recovery in patients with AKI-D.
Collapse
Affiliation(s)
- Jaclyn R. Daniels
- Division of Systems Biology, National Center for Toxicological Research, US Food and Drug Administration, Jefferson, Arkansas
| | - Jennie Z. Ma
- Division of Biostatistics, Department of Public Health Sciences, University of Virginia, Charlottesville, Virginia,Division of Nephrology, Center for Immunity, Inflammation and Regenerative Medicine, University of Virginia, Charlottesville, Virginia
| | - Zhijun Cao
- Division of Systems Biology, National Center for Toxicological Research, US Food and Drug Administration, Jefferson, Arkansas
| | - Richard D. Beger
- Division of Systems Biology, National Center for Toxicological Research, US Food and Drug Administration, Jefferson, Arkansas
| | - Jinchun Sun
- Division of Systems Biology, National Center for Toxicological Research, US Food and Drug Administration, Jefferson, Arkansas
| | - Laura Schnackenberg
- Division of Systems Biology, National Center for Toxicological Research, US Food and Drug Administration, Jefferson, Arkansas
| | - Lisa Pence
- Division of Systems Biology, National Center for Toxicological Research, US Food and Drug Administration, Jefferson, Arkansas
| | - Devasmita Choudhury
- Division of Nephrology, Center for Immunity, Inflammation and Regenerative Medicine, University of Virginia, Charlottesville, Virginia,Salem Veterans Affairs Medical Center, Salem, Virginia
| | - Paul M. Palevsky
- Veterans Affairs Pittsburgh Healthcare System, Pittsburgh, Pennsylvania,Renal-Electrolye Division, Department of Medicine, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
| | - Didier Portilla
- Division of Nephrology, Center for Immunity, Inflammation and Regenerative Medicine, University of Virginia, Charlottesville, Virginia
| | - Li-Rong Yu
- Division of Systems Biology, National Center for Toxicological Research, US Food and Drug Administration, Jefferson, Arkansas
| |
Collapse
|
71
|
Saxena Y, Routh S, Mukhopadhaya A. Immunoporosis: Role of Innate Immune Cells in Osteoporosis. Front Immunol 2021; 12:687037. [PMID: 34421899 PMCID: PMC8374941 DOI: 10.3389/fimmu.2021.687037] [Citation(s) in RCA: 114] [Impact Index Per Article: 28.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2021] [Accepted: 07/22/2021] [Indexed: 12/11/2022] Open
Abstract
Osteoporosis or porous bone disorder is the result of an imbalance in an otherwise highly balanced physiological process known as 'bone remodeling'. The immune system is intricately involved in bone physiology as well as pathologies. Inflammatory diseases are often correlated with osteoporosis. Inflammatory mediators such as reactive oxygen species (ROS), and pro-inflammatory cytokines and chemokines directly or indirectly act on the bone cells and play a role in the pathogenesis of osteoporosis. Recently, Srivastava et al. (Srivastava RK, Dar HY, Mishra PK. Immunoporosis: Immunology of Osteoporosis-Role of T Cells. Frontiers in immunology. 2018;9:657) have coined the term "immunoporosis" to emphasize the role of immune cells in the pathology of osteoporosis. Accumulated pieces of evidence suggest both innate and adaptive immune cells contribute to osteoporosis. However, innate cells are the major effectors of inflammation. They sense various triggers to inflammation such as pathogen-associated molecular patterns (PAMPs), damage-associated molecular patterns (DAMPs), cellular stress, etc., thus producing pro-inflammatory mediators that play a critical role in the pathogenesis of osteoporosis. In this review, we have discussed the role of the innate immune cells in great detail and divided these cells into different sections in a systemic manner. In the beginning, we talked about cells of the myeloid lineage, including macrophages, monocytes, and dendritic cells. This group of cells explicitly influences the skeletal system by the action of production of pro-inflammatory cytokines and can transdifferentiate into osteoclast. Other cells of the myeloid lineage, such as neutrophils, eosinophils, and mast cells, largely impact osteoporosis via the production of pro-inflammatory cytokines. Further, we talked about the cells of the lymphoid lineage, including natural killer cells and innate lymphoid cells, which share innate-like properties and play a role in osteoporosis. In addition to various innate immune cells, we also discussed the impact of classical pro-inflammatory cytokines on osteoporosis. We also highlighted the studies regarding the impact of physiological and metabolic changes in the body, which results in chronic inflammatory conditions such as ageing, ultimately triggering osteoporosis.
Collapse
Affiliation(s)
- Yogesh Saxena
- Department of Biological Sciences, Indian Institute of Science Education and Research Mohali, Mohali, India
| | - Sanjeev Routh
- Department of Biological Sciences, Indian Institute of Science Education and Research Mohali, Mohali, India
| | - Arunika Mukhopadhaya
- Department of Biological Sciences, Indian Institute of Science Education and Research Mohali, Mohali, India
| |
Collapse
|
72
|
Duan Z, Liu M, Yuan L, Du X, Wu M, Yang Y, Wang L, Zhou K, Yang M, Zou Y, Xiang Y, Qu X, Liu H, Qin X, Liu C. Innate lymphoid cells are double-edged swords under the mucosal barrier. J Cell Mol Med 2021; 25:8579-8587. [PMID: 34378306 PMCID: PMC8435454 DOI: 10.1111/jcmm.16856] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2021] [Accepted: 07/20/2021] [Indexed: 11/28/2022] Open
Abstract
As the direct contacting site for pathogens and allergens, the mucosal barrier plays a vital role in the lungs and intestines. Innate lymphoid cells (ILCs) are particularly resident in the mucosal barrier and participate in several pathophysiological processes, such as maintaining or disrupting barrier integrity, preventing various pathogenic invasions. In the pulmonary mucosae, ILCs sometimes aggravate inflammation and mucus hypersecretion but restore airway epithelial integrity and maintain lung tissue homeostasis at other times. In the intestinal mucosae, ILCs can increase epithelial permeability, leading to severe intestinal inflammation on the one hand, and assist mucosal barrier in resisting bacterial invasion on the other hand. In this review, we will illustrate the positive and negative roles of ILCs in mucosal barrier immunity.
Collapse
Affiliation(s)
- Zhen Duan
- Department of Physiology, School of Basic Medicine Science, Central South University, Changsha, China
| | - Mandie Liu
- Department of Physiology, School of Basic Medicine Science, Central South University, Changsha, China
| | - Lin Yuan
- Department of Physiology, School of Basic Medicine Science, Central South University, Changsha, China
| | - Xizi Du
- Department of Physiology, School of Basic Medicine Science, Central South University, Changsha, China
| | - Mengping Wu
- Department of Physiology, School of Basic Medicine Science, Central South University, Changsha, China
| | - Yu Yang
- Department of Physiology, School of Basic Medicine Science, Central South University, Changsha, China
| | - Leyuan Wang
- Department of Physiology, School of Basic Medicine Science, Central South University, Changsha, China
| | - Kai Zhou
- Department of Physiology, School of Basic Medicine Science, Central South University, Changsha, China
| | - Ming Yang
- Centre for Asthma and Respiratory Disease, School of Biomedical Sciences and Pharmacy, Faculty of Health and Medicine, University of Newcastle and Hunter Medical Research Institute, Callaghan, NSW, Australia
| | - Yizhou Zou
- Department of Immunology, School of Basic Medicine Science, Central South University, Changsha, China
| | - Yang Xiang
- Department of Physiology, School of Basic Medicine Science, Central South University, Changsha, China
| | - Xiangping Qu
- Department of Physiology, School of Basic Medicine Science, Central South University, Changsha, China
| | - Huijun Liu
- Department of Physiology, School of Basic Medicine Science, Central South University, Changsha, China
| | - Xiaoqun Qin
- Department of Physiology, School of Basic Medicine Science, Central South University, Changsha, China
| | - Chi Liu
- Department of Physiology, School of Basic Medicine Science, Central South University, Changsha, China.,China-Africa Infectious Diseases Research Center, Xiangya School of Medicine, Central South University, Changsha, China
| |
Collapse
|
73
|
Schanz O, Cornez I, Yajnanarayana SP, David FS, Peer S, Gruber T, Krawitz P, Brossart P, Heine A, Landsberg J, Baier G, Wolf D. Tumor rejection in Cblb -/- mice depends on IL-9 and Th9 cells. J Immunother Cancer 2021; 9:jitc-2021-002889. [PMID: 34272310 PMCID: PMC8287598 DOI: 10.1136/jitc-2021-002889] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/30/2021] [Indexed: 12/22/2022] Open
Abstract
BACKGROUND Casitas B lymphoma-b (Cbl-b) is a central negative regulator of cytotoxic T and natural killer (NK) cells and functions as an intracellular checkpoint in cancer. In particular, Th9 cells support mast cell activation, promote dendritic cell recruitment, enhance the cytolytic function of cytotoxic T lymphocytes and NK cells, and directly kill tumor cells, thereby contributing to tumor immunity. However, the role of Cbl-b in the differentiation and antitumor function of Th9 cells is not sufficiently resolved. METHODS Using Cblb -/- mice, we investigated the effect of knocking out Cblb on the differentiation process and function of different T helper cell subsets, focusing on regulatory T cell (Treg) and Th9 cells. We applied single-cell RNA (scRNA) sequencing of in vitro differentiated Th9 cells to understand how Cbl-b shapes the transcriptome and regulates the differentiation and function of Th9 cells. We transferred tumor-model antigen-specific Cblb -/- Th9 cells into melanoma-bearing mice and assessed tumor control in vivo. In addition, we blocked interleukin (IL)-9 in melanoma cell-exposed Cblb -/- mice to investigate the role of IL-9 in tumor immunity. RESULTS Here, we provide experimental evidence that Cbl-b acts as a rheostat favoring Tregs at the expense of Th9 cell differentiation. Cblb -/- Th9 cells exert superior antitumor activity leading to improved melanoma control in vivo. Accordingly, blocking IL-9 in melanoma cell-exposed Cblb -/- mice reversed their tumor rejection phenotype. Furthermore, scRNA sequencing of in vitro differentiated Th9 cells from naïve T cells isolated from wildtype and Cblb -/- animals revealed a transcriptomic basis for increased Th9 cell differentiation. CONCLUSION We established IL-9 and Th9 cells as key antitumor executers in Cblb -/- animals. This knowledge may be helpful for the future improvement of adoptive T cell therapies in cancer.
Collapse
Affiliation(s)
- Oliver Schanz
- Oncology, Hematology, Immunoncology and Rheumatology, University Hospital Bonn, Bonn, Germany
| | - Isabelle Cornez
- Oncology, Hematology, Immunoncology and Rheumatology, University Hospital Bonn, Bonn, Germany
| | | | - Friederike Sophie David
- Institute for Genomic Statistics and Bioinformatics, University Hospital Bonn, Bonn, Germany.,Institute of Human Genetics, University Hospital Bonn, Bonn, Germany
| | - Sebastian Peer
- Institute of Translational Cell Genetics, Medical University of Innsbruck, Innsbruck, Austria
| | - Thomas Gruber
- Institute of Translational Cell Genetics, Medical University of Innsbruck, Innsbruck, Austria
| | - Peter Krawitz
- Institute for Genomic Statistics and Bioinformatics, University Hospital Bonn, Bonn, Germany
| | - Peter Brossart
- Oncology, Hematology, Immunoncology and Rheumatology, University Hospital Bonn, Bonn, Germany
| | - Annkristin Heine
- Oncology, Hematology, Immunoncology and Rheumatology, University Hospital Bonn, Bonn, Germany
| | | | - Gottfried Baier
- Institute of Translational Cell Genetics, Medical University of Innsbruck, Innsbruck, Austria
| | - Dominik Wolf
- Oncology, Hematology, Immunoncology and Rheumatology, University Hospital Bonn, Bonn, Germany .,Department of Internal Medicine V, Hematology and Oncology, and Tyrolean Cancer Research Institute (TKFI), Medical University of Innsbruck, Innsbruck, Austria
| |
Collapse
|
74
|
Pinto CM, Horta LS, Soares AP, Carvalho BA, Ferreira E, Lages EB, Ferreira LAM, Faraco AAG, Santiago HC, Goulart GAC. Nanoencapsulated Doxorubicin Prevents Mucositis Development in Mice. Pharmaceutics 2021; 13:1021. [PMID: 34371713 PMCID: PMC8329927 DOI: 10.3390/pharmaceutics13071021] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2021] [Revised: 06/27/2021] [Accepted: 06/30/2021] [Indexed: 12/26/2022] Open
Abstract
Doxorubicin (DOX), a chemotherapy drug successfully used in the therapy of various types of cancer, is currently associated with the mucositis development, an inflammation that can cause ulcerative lesions in the mucosa of the gastrointestinal tract, abdominal pain and secondary infections. To increase the safety of the chemotherapy, we loaded DOX into nanostructured lipid carriers (NLCs). The NLC-DOX was characterized by HPLC, DLS, NTA, Zeta potential, FTIR, DSC, TEM and cryogenic-TEM. The ability of NLC-DOX to control the DOX release was evaluated through in vitro release studies. Moreover, the effect of NLC-DOX on intestinal mucosa was compared to a free DOX solution in C57BL/6 mice. The NLC-DOX showed spherical shape, high drug encapsulation efficiency (84.8 ± 4.6%), high drug loading (55.2 ± 3.4 mg/g) and low average diameter (66.0-78.8 nm). The DSC and FTIR analyses showed high interaction between the NLC components, resulting in controlled drug release. Treatment with NLC-DOX attenuated DOX-induced mucositis in mice, improving shortening on villus height and crypt depth, decreased inflammatory parameters, preserved intestinal permeability and increased expression of tight junctions (ZO-1 and Ocludin). These results indicated that encapsulation of DOX in NLCs is viable and reduces the drug toxicity to mucosal structures.
Collapse
Affiliation(s)
- Cristiane M. Pinto
- Department of Pharmaceutics, Faculty of Pharmacy, Universidade Federal de Minas Gerais, Belo Horizonte 31270-901, MG, Brazil; (C.M.P.); (A.P.S.); (E.B.L.); (L.A.M.F.); (A.A.G.F.)
| | - Laila S. Horta
- Department of Biochemistry and Immunology, Universidade Federal de Minas Gerais, Belo Horizonte 31270-901, MG, Brazil; (L.S.H.); (H.C.S.)
| | - Amanda P. Soares
- Department of Pharmaceutics, Faculty of Pharmacy, Universidade Federal de Minas Gerais, Belo Horizonte 31270-901, MG, Brazil; (C.M.P.); (A.P.S.); (E.B.L.); (L.A.M.F.); (A.A.G.F.)
- Department of Biochemistry and Immunology, Universidade Federal de Minas Gerais, Belo Horizonte 31270-901, MG, Brazil; (L.S.H.); (H.C.S.)
| | - Bárbara A. Carvalho
- Department of General Pathology, Biological Science Institute, Universidade Federal de Minas Gerais, Belo Horizonte 31270-901, MG, Brazil; (B.A.C.); (E.F.)
| | - Enio Ferreira
- Department of General Pathology, Biological Science Institute, Universidade Federal de Minas Gerais, Belo Horizonte 31270-901, MG, Brazil; (B.A.C.); (E.F.)
| | - Eduardo B. Lages
- Department of Pharmaceutics, Faculty of Pharmacy, Universidade Federal de Minas Gerais, Belo Horizonte 31270-901, MG, Brazil; (C.M.P.); (A.P.S.); (E.B.L.); (L.A.M.F.); (A.A.G.F.)
| | - Lucas A. M. Ferreira
- Department of Pharmaceutics, Faculty of Pharmacy, Universidade Federal de Minas Gerais, Belo Horizonte 31270-901, MG, Brazil; (C.M.P.); (A.P.S.); (E.B.L.); (L.A.M.F.); (A.A.G.F.)
| | - André A. G. Faraco
- Department of Pharmaceutics, Faculty of Pharmacy, Universidade Federal de Minas Gerais, Belo Horizonte 31270-901, MG, Brazil; (C.M.P.); (A.P.S.); (E.B.L.); (L.A.M.F.); (A.A.G.F.)
| | - Helton C. Santiago
- Department of Biochemistry and Immunology, Universidade Federal de Minas Gerais, Belo Horizonte 31270-901, MG, Brazil; (L.S.H.); (H.C.S.)
| | - Gisele A. C. Goulart
- Department of Pharmaceutics, Faculty of Pharmacy, Universidade Federal de Minas Gerais, Belo Horizonte 31270-901, MG, Brazil; (C.M.P.); (A.P.S.); (E.B.L.); (L.A.M.F.); (A.A.G.F.)
| |
Collapse
|
75
|
Falquet M, Ercolano G, Jandus P, Jandus C, Trabanelli S. Healthy and Patient Type 2 Innate Lymphoid Cells are Differently Affected by in vitro Culture Conditions. J Asthma Allergy 2021; 14:773-783. [PMID: 34239308 PMCID: PMC8259735 DOI: 10.2147/jaa.s304126] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2021] [Accepted: 04/29/2021] [Indexed: 12/20/2022] Open
Abstract
Background Type 2 innate lymphoid cells (ILC2s) have emerged as key players in the development of type 2 driven diseases such as allergy and asthma. Due to their low number in the circulation, in vitro expansion is needed to unravel their mechanisms of action. Purpose The aim of this study is to assess the impact of different culture conditions and address whether the method of expansion may distinctly affect healthy donor or patient-derived ILC2s. Methods Here, we described the impact of six different culture conditions on the proliferation, phenotype and function of human ILC2s freshly obtained from healthy donors (healthy ILC2s) and allergic patients (patient ILC2s). Results We showed that the cytokine cocktail or the PHA induced the highest proliferation of healthy ILC2s and patient ILC2s, respectively. We observed that the stromal cells OP9, used as ILC2 feeders, did not boost their proliferation, but impaired the activation marker expression and the function of patient ILC2s. Furthermore, we demonstrated that the culture conditions differently impacted the activation state of c-Kithigh and c-Kitlow ILC2s, in both healthy donors and allergic patients. Last, we also observed that ILC2s expanded only with IL-2 and IL-7 were the most prone to secrete IL-5 and IL-13 upon IL-33 stimulation. In contrast, in patients, the addition of OP9 cells during the expansion restrained their type 2 cytokine secretory functions. Conclusion This report highlights that culture conditions distinctly impacted on the healthy or patient ILC2 behavior, with important consequences for their study in disease settings.
Collapse
Affiliation(s)
- Maryline Falquet
- Ludwig Institute for Cancer Research, Lausanne Branch, Lausanne, Switzerland.,Department of Pathology and Immunology, Faculty of Medicine, University of Geneva, Geneva, Switzerland
| | - Giuseppe Ercolano
- Ludwig Institute for Cancer Research, Lausanne Branch, Lausanne, Switzerland.,Department of Pathology and Immunology, Faculty of Medicine, University of Geneva, Geneva, Switzerland
| | - Peter Jandus
- Division of Immunology and Allergology, University Hospital and Medical Faculty, Geneva, Switzerland
| | - Camilla Jandus
- Ludwig Institute for Cancer Research, Lausanne Branch, Lausanne, Switzerland.,Department of Pathology and Immunology, Faculty of Medicine, University of Geneva, Geneva, Switzerland
| | - Sara Trabanelli
- Ludwig Institute for Cancer Research, Lausanne Branch, Lausanne, Switzerland.,Department of Pathology and Immunology, Faculty of Medicine, University of Geneva, Geneva, Switzerland
| |
Collapse
|
76
|
Lin Q, Menon MC, He JC. IL-9: a novel pro-podocyte survival cytokine in FSGS. Kidney Int 2021; 98:541-543. [PMID: 32828233 DOI: 10.1016/j.kint.2020.05.045] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2020] [Revised: 05/06/2020] [Accepted: 05/11/2020] [Indexed: 12/26/2022]
Abstract
Progressive focal segmental glomerulosclerosis, characterized by podocyte loss, is often refractory to treatment and leads to progressive proteinuric chronic kidney disease. Interleukin-9 (IL-9) is reported to play important roles in innate and adaptive immunity in extrarenal inflammatory diseases. By using an IL-9 knockout mouse model, Xiong et al. demonstrate IL-9 as a novel pro-podocyte survival cytokine in the adriamycin nephropathy model of focal segmental glomerulosclerosis. Sequential in vitro and in vivo data corroborate a direct protective role, rather than an immunologic role, for IL-9 on podocyte survival. This commentary highlights these novel data and discusses the necessary steps for developing IL-9 as a potential novel therapeutic for focal segmental glomerulosclerosis.
Collapse
Affiliation(s)
- Qisheng Lin
- Division of Nephrology, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, USA; Department of Nephrology, Renji Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai, China
| | - Madhav C Menon
- Division of Nephrology, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, USA
| | - John Cijiang He
- Division of Nephrology, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, USA.
| |
Collapse
|
77
|
Zheng H, Zhang Y, Pan J, Liu N, Qin Y, Qiu L, Liu M, Wang T. The Role of Type 2 Innate Lymphoid Cells in Allergic Diseases. Front Immunol 2021; 12:586078. [PMID: 34177881 PMCID: PMC8220221 DOI: 10.3389/fimmu.2021.586078] [Citation(s) in RCA: 49] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2020] [Accepted: 05/10/2021] [Indexed: 12/22/2022] Open
Abstract
Allergic diseases are significant diseases that affect many patients worldwide. In the past few decades, the incidence of allergic diseases has increased significantly due to environmental changes and social development, which has posed a substantial public health burden and even led to premature death. The understanding of the mechanism underlying allergic diseases has been substantially advanced, and the occurrence of allergic diseases and changes in the immune system state are known to be correlated. With the identification and in-depth understanding of innate lymphoid cells, researchers have gradually revealed that type 2 innate lymphoid cells (ILC2s) play important roles in many allergic diseases. However, our current studies of ILC2s are limited, and their status in allergic diseases remains unclear. This article provides an overview of the common phenotypes and activation pathways of ILC2s in different allergic diseases as well as potential research directions to improve the understanding of their roles in different allergic diseases and ultimately find new treatments for these diseases.
Collapse
Affiliation(s)
- Haocheng Zheng
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China
| | - Yi Zhang
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China
| | - Jiachuang Pan
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China
| | - Nannan Liu
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China
| | - Yu Qin
- Beijing Research Institute of Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China
| | - Linghui Qiu
- Journal Press of Global Traditional Chinese Medicine, Beijing, China
| | - Min Liu
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China
| | - Tieshan Wang
- Beijing Research Institute of Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China
| |
Collapse
|
78
|
Sandonà M, Di Pietro L, Esposito F, Ventura A, Silini AR, Parolini O, Saccone V. Mesenchymal Stromal Cells and Their Secretome: New Therapeutic Perspectives for Skeletal Muscle Regeneration. Front Bioeng Biotechnol 2021; 9:652970. [PMID: 34095095 PMCID: PMC8172230 DOI: 10.3389/fbioe.2021.652970] [Citation(s) in RCA: 64] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2021] [Accepted: 04/01/2021] [Indexed: 12/14/2022] Open
Abstract
Mesenchymal stromal cells (MSCs) are multipotent cells found in different tissues: bone marrow, peripheral blood, adipose tissues, skeletal muscle, perinatal tissues, and dental pulp. MSCs are able to self-renew and to differentiate into multiple lineages, and they have been extensively used for cell therapy mostly owing to their anti-fibrotic and immunoregulatory properties that have been suggested to be at the basis for their regenerative capability. MSCs exert their effects by releasing a variety of biologically active molecules such as growth factors, chemokines, and cytokines, either as soluble proteins or enclosed in extracellular vesicles (EVs). Analyses of MSC-derived secretome and in particular studies on EVs are attracting great attention from a medical point of view due to their ability to mimic all the therapeutic effects produced by the MSCs (i.e., endogenous tissue repair and regulation of the immune system). MSC-EVs could be advantageous compared with the parental cells because of their specific cargo containing mRNAs, miRNAs, and proteins that can be biologically transferred to recipient cells. MSC-EV storage, transfer, and production are easier; and their administration is also safer than MSC therapy. The skeletal muscle is a very adaptive tissue, but its regenerative potential is altered during acute and chronic conditions. Recent works demonstrate that both MSCs and their secretome are able to help myofiber regeneration enhancing myogenesis and, interestingly, can be manipulated as a novel strategy for therapeutic interventions in muscular diseases like muscular dystrophies or atrophy. In particular, MSC-EVs represent promising candidates for cell free-based muscle regeneration. In this review, we aim to give a complete picture of the therapeutic properties and advantages of MSCs and their products (MSC-derived EVs and secreted factors) relevant for skeletal muscle regeneration in main muscular diseases.
Collapse
Affiliation(s)
- Martina Sandonà
- Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS), Fondazione Santa Lucia, Rome, Italy
| | - Lorena Di Pietro
- Dipartimento di Scienze della Vita e Sanità Pubblica, Università Cattolica del Sacro Cuore, Rome, Italy
| | - Federica Esposito
- Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS), Fondazione Santa Lucia, Rome, Italy
| | - Alessia Ventura
- Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS), Fondazione Santa Lucia, Rome, Italy
| | - Antonietta Rosa Silini
- Centro di Ricerca "E. Menni", Fondazione Poliambulanza - Istituto Ospedaliero, Brescia, Italy
| | - Ornella Parolini
- Dipartimento di Scienze della Vita e Sanità Pubblica, Università Cattolica del Sacro Cuore, Rome, Italy.,Fondazione Policlinico Universitario "Agostino Gemelli" IRCCS, Rome, Italy
| | - Valentina Saccone
- Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS), Fondazione Santa Lucia, Rome, Italy.,Dipartimento di Scienze della Vita e Sanità Pubblica, Università Cattolica del Sacro Cuore, Rome, Italy
| |
Collapse
|
79
|
Mathä L, Martinez-Gonzalez I, Steer CA, Takei F. The Fate of Activated Group 2 Innate Lymphoid Cells. Front Immunol 2021; 12:671966. [PMID: 33968080 PMCID: PMC8100346 DOI: 10.3389/fimmu.2021.671966] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2021] [Accepted: 04/07/2021] [Indexed: 12/20/2022] Open
Abstract
Group 2 innate lymphoid cells (ILC2s) reside in both mucosal and non-mucosal tissues and play critical roles in the first line of defense against parasites and irritants such as allergens. Upon activation by cytokines released from epithelial and stromal cells during tissue damage or stimulation, ILC2s produce copious amounts of IL-5 and IL-13, leading to type 2 inflammation. Over the past 10 years, ILC2 involvement in a variety of human diseases has been unveiled. However, questions remain as to the fate of ILC2s after activation and how that might impact their role in chronic inflammatory diseases such as asthma and fibrosis. Here, we review studies that have revealed novel properties of post-activation ILC2s including the generation of immunological memory, exhausted-like phenotype, transdifferentiation and activation-induced migration.
Collapse
Affiliation(s)
- Laura Mathä
- Terry Fox Laboratory, British Columbia Cancer, Vancouver, BC, Canada
| | | | - Catherine A Steer
- Terry Fox Laboratory, British Columbia Cancer, 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
|
80
|
Cox JR, Cruickshank SM, Saunders AE. Maintenance of Barrier Tissue Integrity by Unconventional Lymphocytes. Front Immunol 2021; 12:670471. [PMID: 33936115 PMCID: PMC8079635 DOI: 10.3389/fimmu.2021.670471] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2021] [Accepted: 03/26/2021] [Indexed: 12/12/2022] Open
Abstract
Mucosal surfaces, as a first barrier with the environment are especially susceptible to damage from both pathogens and physical trauma. Thus, these sites require tightly regulated repair programs to maintain barrier function in the face of such insults. Barrier sites are also enriched for unconventional lymphocytes, which lack rearranged antigen receptors or express only a limited range of such receptors, such as ILCs (Innate Lymphoid Cells), γδ T Cells and MAIT (Mucosal-Associated Invariant T Cells). Recent studies have uncovered critical roles for unconventional lymphocytes in regulating mucosal barrier function, and, in particular, have highlighted their important involvement in barrier repair. The production of growth factors such as amphiregulin by ILC2, and fibroblast growth factors by γδ T cells have been shown to promote tissue repair at multiple barrier sites. Additionally, MAIT cells have been shown to exhibit pro-repair phenotypes and demonstrate microbiota-dependent promotion of murine skin healing. In this review we will discuss how immune responses at mucosal sites are controlled by unconventional lymphocytes and the ways in which these cells promote tissue repair to maintain barrier integrity in the skin, gut and lungs.
Collapse
Affiliation(s)
- Joshua R Cox
- Manchester Collaborative Centre for Inflammation Research, Division of Infection, Immunity and Respiratory Medicine, School of Biological Science, Faculty of Biology, Medicine and Health, University of Manchester, Manchester Academic Health Science Centre, Manchester, United Kingdom.,Lydia Becker Institute of Immunology and Inflammation, Division of Infection, Immunity and Respiratory Medicine, School of Biological Science, Faculty of Biology, Medicine and Health, University of Manchester, Manchester Academic Health Science Centre, Manchester, United Kingdom
| | - Sheena M Cruickshank
- Lydia Becker Institute of Immunology and Inflammation, Division of Infection, Immunity and Respiratory Medicine, School of Biological Science, Faculty of Biology, Medicine and Health, University of Manchester, Manchester Academic Health Science Centre, Manchester, United Kingdom
| | - Amy E Saunders
- Manchester Collaborative Centre for Inflammation Research, Division of Infection, Immunity and Respiratory Medicine, School of Biological Science, Faculty of Biology, Medicine and Health, University of Manchester, Manchester Academic Health Science Centre, Manchester, United Kingdom.,Lydia Becker Institute of Immunology and Inflammation, Division of Infection, Immunity and Respiratory Medicine, School of Biological Science, Faculty of Biology, Medicine and Health, University of Manchester, Manchester Academic Health Science Centre, Manchester, United Kingdom
| |
Collapse
|
81
|
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
|
82
|
De Pasquale C, Campana S, Bonaccorsi I, Carrega P, Ferlazzo G. ILC in chronic inflammation, cancer and targeting with biologicals. Mol Aspects Med 2021; 80:100963. [PMID: 33726947 DOI: 10.1016/j.mam.2021.100963] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2020] [Revised: 11/11/2020] [Accepted: 03/04/2021] [Indexed: 12/14/2022]
Abstract
Since their discovery, Innate Lymphoid Cells (ILC) have emerged as important effector cells, serving multiple roles in maintaining tissue homeostasis and responding to tissue insults. As such, dysregulations of their function and distribution have been observed in a variety of immune-mediated diseases, suggesting a specific role for ILC in the pathophysiology of several disorders including chronic inflammation and cancer. Here, we provide an updated view on ILC biology dissecting their pathological or protective contribution in chronic inflammatory diseases such as multiple sclerosis, inflammatory bowel diseases, psoriasis, rheumatoid arthritis, asthma and COPD, atherosclerosis, also exploring ILC role in tumor surveillance and progression. Throughout the review, we will also highlight how the potential dual role of these cells for protective or pathogenic immunity in many inflammatory diseases makes them interesting targets for the development of novel therapeutic strategies, particularly promising.
Collapse
Affiliation(s)
- Claudia De Pasquale
- Laboratory of Immunology and Biotherapy, Department of Human Pathology, University of Messina, Messina, Italy
| | - Stefania Campana
- Laboratory of Immunology and Biotherapy, Department of Human Pathology, University of Messina, Messina, Italy
| | - Irene Bonaccorsi
- Laboratory of Immunology and Biotherapy, Department of Human Pathology, University of Messina, Messina, Italy; Cell Factory Center and Division of Clinical Pathology, University Hospital Policlinico G.Martino, Messina, Italy
| | - Paolo Carrega
- Laboratory of Immunology and Biotherapy, Department of Human Pathology, University of Messina, Messina, Italy
| | - Guido Ferlazzo
- Laboratory of Immunology and Biotherapy, Department of Human Pathology, University of Messina, Messina, Italy; Cell Factory Center and Division of Clinical Pathology, University Hospital Policlinico G.Martino, Messina, Italy.
| |
Collapse
|
83
|
Pasvenskaite A, Liutkeviciene R, Gedvilaite G, Vilkeviciute A, Liutkevicius V, Uloza V. The Role of IL-9 Polymorphisms and Serum IL-9 Levels in Carcinogenesis and Survival Rate for Laryngeal Squamous Cell Carcinoma. Cells 2021; 10:cells10030601. [PMID: 33803218 PMCID: PMC8001846 DOI: 10.3390/cells10030601] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2021] [Revised: 03/02/2021] [Accepted: 03/05/2021] [Indexed: 11/16/2022] Open
Abstract
Recent studies have described the dichotomous function of IL-9 in various cancer diseases. However, its function has still not been analysed in laryngeal squamous cell carcinoma (LSCC). In the present study, we evaluated five single nucleotide polymorphisms (SNPs) of IL-9 (rs1859430, rs2069870, rs11741137, rs2069885, and rs2069884) and determined their associations with the patients' five-year survival rate. Additionally, we analysed serum IL-9 levels using an enzyme-linked immunosorbent assay. Three hundred LSCC patients and 533 control subjects were included in this study. A significant association between the patients' survival rate and distribution of IL-9 rs1859430 variants was revealed: patients carrying AA genotype had a higher risk of dying (p = 0.005). Haplotypes A-G-C-G-G of IL-9 (rs1859430, rs2069870, rs11741137, rs2069885, and rs2069884) were associated with 47% lower odds of LSCC occurrence (p = 0.035). Serum IL-9 levels were found detectable in three control group subjects (8.99 ± 12.03 pg/mL). In summary, these findings indicate that the genotypic distribution of IL-9 rs1859430 negatively influences the five-year survival rate of LSCC patients. The haplotypes A-G-C-G-G of IL-9 (rs1859430, rs2069870, rs11741137, rs2069885, and rs2069884) are associated with the lower odds of LSCC development.
Collapse
Affiliation(s)
- Agne Pasvenskaite
- Department of Otorhinolaryngology, Lithuanian University of Health Sciences (LUHS), LT-50161 Kaunas, Lithuania; (V.L.); (V.U.)
- Correspondence: ; Tel.: +370-6532-3034
| | - Rasa Liutkeviciene
- Neuroscience Institute, Lithuanian University of Health Sciences (LUHS), LT-50161 Kaunas, Lithuania; (R.L.); (G.G.); (A.V.)
| | - Greta Gedvilaite
- Neuroscience Institute, Lithuanian University of Health Sciences (LUHS), LT-50161 Kaunas, Lithuania; (R.L.); (G.G.); (A.V.)
| | - Alvita Vilkeviciute
- Neuroscience Institute, Lithuanian University of Health Sciences (LUHS), LT-50161 Kaunas, Lithuania; (R.L.); (G.G.); (A.V.)
| | - Vykintas Liutkevicius
- Department of Otorhinolaryngology, Lithuanian University of Health Sciences (LUHS), LT-50161 Kaunas, Lithuania; (V.L.); (V.U.)
| | - Virgilijus Uloza
- Department of Otorhinolaryngology, Lithuanian University of Health Sciences (LUHS), LT-50161 Kaunas, Lithuania; (V.L.); (V.U.)
| |
Collapse
|
84
|
Roberts LB, Schnoeller C, Berkachy R, Darby M, Pillaye J, Oudhoff MJ, Parmar N, Mackowiak C, Sedda D, Quesniaux V, Ryffel B, Vaux R, Gounaris K, Berrard S, Withers DR, Horsnell WGC, Selkirk ME. Acetylcholine production by group 2 innate lymphoid cells promotes mucosal immunity to helminths. Sci Immunol 2021; 6:6/57/eabd0359. [PMID: 33674321 DOI: 10.1126/sciimmunol.abd0359] [Citation(s) in RCA: 54] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2020] [Accepted: 12/11/2020] [Indexed: 12/13/2022]
Abstract
Innate lymphoid cells (ILCs) are critical mediators of immunological and physiological responses at mucosal barrier sites. Whereas neurotransmitters can stimulate ILCs, the synthesis of small-molecule neurotransmitters by these cells has only recently been appreciated. Group 2 ILCs (ILC2s) are shown here to synthesize and release acetylcholine (ACh) during parasitic nematode infection. The cholinergic phenotype of pulmonary ILC2s was associated with their activation state, could be induced by in vivo exposure to extracts of Alternaria alternata or the alarmin cytokines interleukin-33 (IL-33) and IL-25, and was augmented by IL-2 in vitro. Genetic disruption of ACh synthesis by murine ILC2s resulted in increased parasite burdens, lower numbers of ILC2s, and reduced lung and gut barrier responses to Nippostrongylus brasiliensis infection. These data demonstrate a functional role for ILC2-derived ACh in the expansion of ILC2s for maximal induction of type 2 immunity.
Collapse
Affiliation(s)
- Luke B Roberts
- Department of Life Sciences, Imperial College London, London, UK. .,School of Immunology and Microbial Sciences, King's College London, Great Maze Pond, London SE1 9RT, UK
| | | | - Rita Berkachy
- Department of Life Sciences, Imperial College London, London, UK
| | - Matthew Darby
- Wellcome Centre for Infectious Diseases Research in Africa, Institute of Infectious Disease and Molecular Medicine, University of Cape Town, Cape Town, South Africa
| | - Jamie Pillaye
- Wellcome Centre for Infectious Diseases Research in Africa, Institute of Infectious Disease and Molecular Medicine, University of Cape Town, Cape Town, South Africa.,College of Medical and Dental Sciences, University of Birmingham, Birmingham, UK
| | - Menno J Oudhoff
- Centre of Molecular Inflammation Research (CEMIR), Department of Clinical and Molecular Medicine, Norwegian University of Science and Technology (NTNU), 7491 Trondheim, Norway
| | - Naveen Parmar
- Centre of Molecular Inflammation Research (CEMIR), Department of Clinical and Molecular Medicine, Norwegian University of Science and Technology (NTNU), 7491 Trondheim, Norway
| | - Claire Mackowiak
- Wellcome Centre for Infectious Diseases Research in Africa, Institute of Infectious Disease and Molecular Medicine, University of Cape Town, Cape Town, South Africa
| | - Delphine Sedda
- Laboratory of Molecular and Experimental Immunology and Neurogenetics, UMR 7355, CNRS-University of Orleans and Le Studium Institute for Advanced Studies, Rue Dupanloup, 45000 Orléans, France
| | - Valerie Quesniaux
- Laboratory of Molecular and Experimental Immunology and Neurogenetics, UMR 7355, CNRS-University of Orleans and Le Studium Institute for Advanced Studies, Rue Dupanloup, 45000 Orléans, France
| | - Bernhard Ryffel
- Laboratory of Molecular and Experimental Immunology and Neurogenetics, UMR 7355, CNRS-University of Orleans and Le Studium Institute for Advanced Studies, Rue Dupanloup, 45000 Orléans, France
| | - Rachel Vaux
- Department of Life Sciences, Imperial College London, London, UK
| | | | - Sylvie Berrard
- Université de Paris, NeuroDiderot, Inserm, 75019 Paris, France
| | - David R Withers
- College of Medical and Dental Sciences, University of Birmingham, Birmingham, UK
| | - William G C Horsnell
- Wellcome Centre for Infectious Diseases Research in Africa, Institute of Infectious Disease and Molecular Medicine, University of Cape Town, Cape Town, South Africa. .,College of Medical and Dental Sciences, University of Birmingham, Birmingham, UK.,Laboratory of Molecular and Experimental Immunology and Neurogenetics, UMR 7355, CNRS-University of Orleans and Le Studium Institute for Advanced Studies, Rue Dupanloup, 45000 Orléans, France
| | - Murray E Selkirk
- Department of Life Sciences, Imperial College London, London, UK.
| |
Collapse
|
85
|
Yang D, Guo X, Huang T, Liu C. The Role of Group 3 Innate Lymphoid Cells in Lung Infection and Immunity. Front Cell Infect Microbiol 2021; 11:586471. [PMID: 33718260 PMCID: PMC7947361 DOI: 10.3389/fcimb.2021.586471] [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: 07/23/2020] [Accepted: 01/19/2021] [Indexed: 02/05/2023] Open
Abstract
The lung is constantly exposed to environmental particulates such as aeroallergens, pollutants, or microorganisms and is protected by a poised immune response. Innate lymphoid cells (ILCs) are a population of immune cells found in a variety of tissue sites, particularly barrier surfaces such as the lung and the intestine. ILCs play a crucial role in the innate immune system, and they are involved in the maintenance of mucosal homeostasis, inflammation regulation, tissue remodeling, and pathogen clearance. In recent years, group 3 innate lymphoid cells (ILC3s) have emerged as key mediators of mucosal protection and repair during infection, mainly through IL-17 and IL-22 production. Although research on ILC3s has become focused on the intestinal immunity, the biology and function of pulmonary ILC3s in the pathogenesis of respiratory infections and in the development of chronic pulmonary inflammatory diseases remain elusive. In this review, we will mainly discuss how pulmonary ILC3s act on protection against pathogen challenge and pulmonary inflammation, as well as the underlying mechanisms.
Collapse
Affiliation(s)
- Dan Yang
- Department of Respiratory and Critical Care Medicine, West China School of Medicine and West China Hospital, Sichuan University, Chengdu, China
| | - Xinning Guo
- Department of Respiratory and Critical Care Medicine, West China School of Medicine and West China Hospital, Sichuan University, Chengdu, China
| | - Tingxuan Huang
- Department of Respiratory and Critical Care Medicine, West China School of Medicine and West China Hospital, Sichuan University, Chengdu, China
| | - Chuntao Liu
- Department of Respiratory and Critical Care Medicine, West China School of Medicine and West China Hospital, Sichuan University, Chengdu, China
| |
Collapse
|
86
|
Gouyou B, Ongaro T, Cazzamalli S, De Luca R, Kerschenmeyer A, Valet P, Villa A, Neri D, Matasci M. Antibody-based delivery of interleukin-9 to neovascular structures: Therapeutic evaluation in cancer and arthritis. Exp Biol Med (Maywood) 2021; 246:940-951. [PMID: 33475433 DOI: 10.1177/1535370220981578] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Interleukin-9 is a cytokine with multiple functions, including the ability to activate group 2 innate lymphoid cells, which has been postulated to be therapeutically active in mouse models of arthritis. Similarly, interleukin-9 has been suggested to play an important role in tumor immunity. Here, we describe the cloning, expression, and characterization of three fusion proteins based on murine interleukin-9 and the F8 antibody, specific to the alternatively spliced EDA domain of fibronectin. EDA is strongly expressed in cancer and in various arthritic conditions, while being undetectable in the majority of healthy organs. Interleukin-9-based fusion proteins with an irrelevant antibody specific to hen egg lysozyme served as negative control in our study. The fusion proteins were characterized by quantitative biodistribution analysis in tumor-bearing mice using radioiodinated protein preparations. The highest tumor uptake and best tumor:organ ratios were observed for a format, in which the interleukin-9 moiety was flanked by two units of the F8 antibody in single-chain Fv format. Biological activity of interleukin-9 was retained when the payload was fused to antibodies. However, the targeted delivery of interleukin-9 to the disease site resulted in a modest anti-tumor activity in three different murine models of cancer (K1735M2, CT26, and F9), while no therapeutic benefit was observed in a collagen induced model of arthritis. Collectively, these results confirm the possibility to deliver interleukin-9 to the site of disease but cast doubts about the alleged therapeutic activity of this cytokine in cancer and arthritis, which has been postulated in previous publications.
Collapse
Affiliation(s)
| | - Tiziano Ongaro
- Philochem AG, Libernstrasse 3, Otelfingen 8112, Switzerland
| | | | | | | | - Philippe Valet
- Institut des Maladies Métaboliques et Cardiovasculaires, INSERM U1048, Université de Toulouse, UPS, Cedex 4, Toulouse 31432, France
| | | | - Dario Neri
- Department of Chemistry and Applied Biosciences, Swiss Federal Institute of Technology, Zurich 8093, Switzerland
| | - Mattia Matasci
- Philochem AG, Libernstrasse 3, Otelfingen 8112, Switzerland
| |
Collapse
|
87
|
Wan J, Wu Y, Huang L, Tian Y, Ji X, Abdelaziz MH, Cai W, Dineshkumar K, Lei Y, Yao S, Sun C, Su Z, Wang S, Xu H. ILC2-derived IL-9 inhibits colorectal cancer progression by activating CD8 + T cells. Cancer Lett 2021; 502:34-43. [PMID: 33429004 DOI: 10.1016/j.canlet.2021.01.002] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2020] [Revised: 12/20/2020] [Accepted: 01/04/2021] [Indexed: 12/30/2022]
Abstract
Group 2 innate lymphoid cells (ILC2s), characterized by secretion of type 2 cytokines, regulate multiple immune responses. ILC2s are found in different tumor tissues, and ILC2-derived interleukin (IL)-4, IL-5, and IL-13 act on the cells in tumor microenvironment to participate in tumor progression. ILC2s are abundant in colorectal cancer (CRC) tissue, but the role of ILC2s in CRC remains unclear. In this study, we found that the percentage of ILC2s was higher in CRC tissue than in the adjacent normal tissue and that these ILC2s were the dominant IL-9-secreting cell-subsets in CRC tissue, as shown by flow cytometry analysis. ILC2s-derived IL-9 could activate CD8+ T cells to inhibit tumor growth, while anti-IL-9 reversed this effect. In vivo experiments showed that neutralizing ILC2s promoted tumor growth, while tumor inhibition occurred by intravenous injection of IL-9. In conclusion, our results demonstrated that ILC2-derived IL-9 could activate CD8+ T cells to promote anti-tumor effects in CRC.
Collapse
Affiliation(s)
- Jie Wan
- The Central Laboratory, The Fourth Affiliated Hospital of Jiangsu University, Zhenjiang, 212001, China; Department of Immunology, Jiangsu University, Zhenjiang, 212013, China
| | - Yinqiu Wu
- The Central Laboratory, The Fourth Affiliated Hospital of Jiangsu University, Zhenjiang, 212001, China
| | - Lan Huang
- Department of Immunology, Jiangsu University, Zhenjiang, 212013, China
| | - Yu Tian
- The Central Laboratory, The Fourth Affiliated Hospital of Jiangsu University, Zhenjiang, 212001, China
| | - Xiaoyun Ji
- Department of Immunology, Jiangsu University, Zhenjiang, 212013, China
| | | | - Wei Cai
- The Central Laboratory, The Fourth Affiliated Hospital of Jiangsu University, Zhenjiang, 212001, China
| | - Kesavan Dineshkumar
- The Central Laboratory, The Fourth Affiliated Hospital of Jiangsu University, Zhenjiang, 212001, China
| | - Yuqing Lei
- The Central Laboratory, The Fourth Affiliated Hospital of Jiangsu University, Zhenjiang, 212001, China
| | - Shun Yao
- Center for Pituitary Tumor Surgery, Department of Neurosurgery, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, 510080, China
| | - Caixia Sun
- Department of Immunology, Jiangsu University, Zhenjiang, 212013, China
| | - Zhaoliang Su
- The Central Laboratory, The Fourth Affiliated Hospital of Jiangsu University, Zhenjiang, 212001, China; Department of Immunology, Jiangsu University, Zhenjiang, 212013, China
| | - Shengjun Wang
- Department of Immunology, Jiangsu University, Zhenjiang, 212013, China; Department of Laboratory Medicine, The Affiliated People's Hospital, Jiangsu University, Zhenjiang, 212001, China
| | - Huaxi Xu
- The Central Laboratory, The Fourth Affiliated Hospital of Jiangsu University, Zhenjiang, 212001, China; Department of Immunology, Jiangsu University, Zhenjiang, 212013, China.
| |
Collapse
|
88
|
Poniewierska-Baran A, Tokarz-Deptuła B, Deptuła W. The role of innate lymphoid cells in selected disease states - cancer formation, metabolic disorder and inflammation. Arch Med Sci 2021; 17:196-206. [PMID: 33488872 PMCID: PMC7811321 DOI: 10.5114/aoms.2019.89835] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/23/2017] [Accepted: 10/13/2017] [Indexed: 12/22/2022] Open
Abstract
Innate lymphoid cells (ILCs) are a recently described group of immune cells that can regulate homeostasis and protect mammalian organisms, including humans, from infections and diseases. Considering this, ILC research is still ongoing to better understand the biology of these cells and their roles in the human body. ILCs are a multifunctional group of immune cells, making it important for the medical community to be familiar with the latest research about the ILC families and their functions in selected disease states, such as cancer formation, metabolic disorders and inflammation. By discovering the roles of ILC populations and their participation in many disorders, we can improve disease diagnostics and patient healthcare.
Collapse
Affiliation(s)
| | - Beata Tokarz-Deptuła
- Department of Immunology, Faculty of Biology, University of Szczecin, Szczecin, Poland
| | - Wiesław Deptuła
- Veterinary Center of the Nicolaus Copernicus University, Torun, Poland
| |
Collapse
|
89
|
Do-Thi VA, Lee JO, Lee H, Kim YS. Crosstalk between the Producers and Immune Targets of IL-9. Immune Netw 2020; 20:e45. [PMID: 33425430 PMCID: PMC7779872 DOI: 10.4110/in.2020.20.e45] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2020] [Revised: 10/19/2020] [Accepted: 10/29/2020] [Indexed: 12/18/2022] Open
Abstract
IL-9 has been reported to play dual roles in the pathogenesis of autoimmune disorders and cancers. The collaboration of IL-9 with microenvironmental factors including the broader cytokine milieu and other cellular components may provide important keys to explain its conflicting effects in chronic conditions. In this review, we summarize recent findings on the cellular sources of, and immunological responders to IL-9, in order to interpret the role of IL-9 in the regulation of immune responses. This knowledge will provide new perspectives to improve clinical benefits and limit adverse effects of IL-9 when treating pathologic conditions.
Collapse
Affiliation(s)
- Van Anh Do-Thi
- Department of Life Sciences, Pohang University of Science and Technology (POSTECH), Pohang 37673, Korea
| | - Jie-Oh Lee
- Department of Life Sciences, Pohang University of Science and Technology (POSTECH), Pohang 37673, Korea
| | - Hayyoung Lee
- Institute of Biotechnology, Chungnam National University, Daejeon 34134, Korea
| | - Young Sang Kim
- Department of Biochemistry, College of Natural Sciences, Chungnam National University, Daejeon 34134, Korea
| |
Collapse
|
90
|
Chen W, Shu Q, Fan J. Neural Regulation of Interactions Between Group 2 Innate Lymphoid Cells and Pulmonary Immune Cells. Front Immunol 2020; 11:576929. [PMID: 33193374 PMCID: PMC7658006 DOI: 10.3389/fimmu.2020.576929] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2020] [Accepted: 10/05/2020] [Indexed: 12/18/2022] Open
Abstract
Emerging evidence supports the involvement of nervous system in the regulation of immune responses. Group 2 innate lymphoid cells (ILC2), which function as a crucial bridge between innate and adaptive immunity, are present in large numbers in barrier tissues. Neuropeptides and neurotransmitters have been found to participate in the regulation of ILC2, adding a new dimension to neuroimmunity. However, a comprehensive and detailed overview of the mechanisms of neural regulation of ILC2, associated with previous findings and prospects for future research, is still lacking. In this review, we compile existing information that supports neurons as yet poorly understood regulators of ILC2 in the field of lung innate and adaptive immunity, focusing on neural regulation of the interaction between ILC2 and pulmonary immune cells.
Collapse
Affiliation(s)
- Weiwei Chen
- Department of Surgery, University of Pittsburgh School of Medicine, Pittsburgh, PA, United States.,The Children's Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child Health, Hangzhou, China
| | - Qiang Shu
- The Children's Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child Health, Hangzhou, China
| | - Jie Fan
- Department of Surgery, University of Pittsburgh School of Medicine, Pittsburgh, PA, United States.,Research and Development, Veterans Affairs Pittsburgh Healthcare System, Pittsburgh, PA, United States.,McGowan Institute for Regenerative Medicine, University of Pittsburgh, Pittsburgh, PA, United States
| |
Collapse
|
91
|
Akama Y, Satoh-Takayama N, Kawamoto E, Ito A, Gaowa A, Park EJ, Imai H, Shimaoka M. The Role of Innate Lymphoid Cells in the Regulation of Immune Homeostasis in Sepsis-Mediated Lung Inflammation. Diagnostics (Basel) 2020; 10:diagnostics10100808. [PMID: 33053762 PMCID: PMC7600279 DOI: 10.3390/diagnostics10100808] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2020] [Revised: 10/06/2020] [Accepted: 10/09/2020] [Indexed: 12/11/2022] Open
Abstract
Septic shock/severe sepsis is a deregulated host immune system response to infection that leads to life-threatening organ dysfunction. Lung inflammation as a form of acute lung injury (ALI) is often induced in septic shock. Whereas macrophages and neutrophils have been implicated as the principal immune cells regulating lung inflammation, group two innate lymphoid cells (ILC2s) have recently been identified as a new player regulating immune homeostasis. ILC2 is one of the three major ILC subsets (ILC1s, ILC2s, and ILC3s) comprised of newly identified innate immune cells. These cells are characterized by their ability to rapidly produce type 2 cytokines. ILC2s are predominant resident ILCs and, thereby, have the ability to respond to signals from damaged tissues. ILC2s regulate the immune response, and ILC2-derived type 2 cytokines may exert protective roles against sepsis-induced lung injury. This focused review not only provides readers with new insights into the signaling mechanisms by which ILC2s modulate sepsis-induced lung inflammation, but also proposes ILC2 as a novel therapeutic target for sepsis-induced ALI.
Collapse
Affiliation(s)
- Yuichi Akama
- Department of Molecular Pathobiology and Cell Adhesion Biology, Graduate School of Medicine, Mie University, 2-174 Edobashi, Tsu City, Mie 514-8507, Japan; (E.K.); (A.I.); (A.G.); (E.J.P.)
- Department of Emergency and Disaster Medicine, Faculty, Graduate School of Medicine, Mie University, 2-174 Edobashi, Tsu City, Mie 514-8507, Japan;
- Correspondence: (Y.A.); (M.S.)
| | - Naoko Satoh-Takayama
- Laboratory for Intestinal Ecosystem, Center for Integrative Medical Sciences, RIKEN, 1-7-22 Suehiro-cho, Tsurumi-ku, Yokohama, Kanagawa 230-0045, Japan;
- Immunobiology Laboratory, Graduate School of Medical Life Science, Yokohama City University, 1-7-29 Suehiro-cho, Tsurumi-ku, Yokohama, Kanagawa 230-0045, Japan
| | - Eiji Kawamoto
- Department of Molecular Pathobiology and Cell Adhesion Biology, Graduate School of Medicine, Mie University, 2-174 Edobashi, Tsu City, Mie 514-8507, Japan; (E.K.); (A.I.); (A.G.); (E.J.P.)
- Department of Emergency and Disaster Medicine, Faculty, Graduate School of Medicine, Mie University, 2-174 Edobashi, Tsu City, Mie 514-8507, Japan;
| | - Atsushi Ito
- Department of Molecular Pathobiology and Cell Adhesion Biology, Graduate School of Medicine, Mie University, 2-174 Edobashi, Tsu City, Mie 514-8507, Japan; (E.K.); (A.I.); (A.G.); (E.J.P.)
- Department of Thoracic and Cardiovascular Surgery, Faculty, Graduate School of Medicine, Mie University, 2-174 Edobashi, Tsu City, Mie 514-8507, Japan
| | - Arong Gaowa
- Department of Molecular Pathobiology and Cell Adhesion Biology, Graduate School of Medicine, Mie University, 2-174 Edobashi, Tsu City, Mie 514-8507, Japan; (E.K.); (A.I.); (A.G.); (E.J.P.)
| | - Eun Jeong Park
- Department of Molecular Pathobiology and Cell Adhesion Biology, Graduate School of Medicine, Mie University, 2-174 Edobashi, Tsu City, Mie 514-8507, Japan; (E.K.); (A.I.); (A.G.); (E.J.P.)
| | - Hiroshi Imai
- Department of Emergency and Disaster Medicine, Faculty, Graduate School of Medicine, Mie University, 2-174 Edobashi, Tsu City, Mie 514-8507, Japan;
| | - Motomu Shimaoka
- Department of Molecular Pathobiology and Cell Adhesion Biology, Graduate School of Medicine, Mie University, 2-174 Edobashi, Tsu City, Mie 514-8507, Japan; (E.K.); (A.I.); (A.G.); (E.J.P.)
- Correspondence: (Y.A.); (M.S.)
| |
Collapse
|
92
|
Gong X, Xia L, Su Z. Friend or foe of innate lymphoid cells in inflammation-associated cardiovascular disease. Immunology 2020; 162:368-376. [PMID: 32967038 DOI: 10.1111/imm.13271] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2020] [Revised: 08/11/2020] [Accepted: 09/04/2020] [Indexed: 12/17/2022] Open
Abstract
As a distinctive population of leucocytes, innate lymphoid cells (ILCs) participate in immune-mediated diseases and play crucial roles in tissue remodelling after injury. ILC lineages can be divided into helper ILCs and cytotoxic ILCs. Most helper ILCs are integrated into the fabric of tissues and produce different types of cytokines involving in the pathogenesis of many kinds of cardiovascular disease and form intricate response circuits with adaptive immune cells. However, the specific phenotype and function of helper ILC subsets in cardiovascular diseases are still poorly understood. In this review, we firstly highlight the distribution of helper ILCs in cardiovascular system and further discuss the potential contribution of helper ILCs in inflammation-associated cardiovascular disease.
Collapse
Affiliation(s)
- Xiangmei Gong
- International Genome Center, Jiangsu University, Zhenjiang, China.,Department of Immunology, Jiangsu University, Zhenjiang, China
| | - Lin Xia
- International Genome Center, Jiangsu University, Zhenjiang, China.,Department of Laboratory Medicine, Affiliated Hospital of Jiangsu University, Zhenjiang, China
| | - Zhaoliang Su
- International Genome Center, Jiangsu University, Zhenjiang, China.,Department of Immunology, Jiangsu University, Zhenjiang, China.,Laboratory Center, The Fourth Affiliated Hospital of Jiangsu University, Zhenjiang, China
| |
Collapse
|
93
|
IL-9-producing T cells: potential players in allergy and cancer. Nat Rev Immunol 2020; 21:37-48. [PMID: 32788707 DOI: 10.1038/s41577-020-0396-0] [Citation(s) in RCA: 77] [Impact Index Per Article: 15.4] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/02/2020] [Indexed: 01/03/2023]
Abstract
IL-9-producing CD4+ T cells have been considered to represent a distinct T helper cell (TH cell) subset owing to their unique developmental programme in vitro, their expression of distinct transcription factors (including PU.1) and their copious production of IL-9. It remains debatable whether these cells represent a truly unique TH cell subset in vivo, but they are closely related to the T helper 2 (TH2) cells that are detected in allergic diseases. In recent years, increasing evidence has also indicated that IL-9-producing T cells may have potent abilities in eradicating advanced tumours, particularly melanomas. Here, we review the latest literature on the development of IL-9-producing T cells and their functions in disease settings, with a particular focus on allergy and cancer. We also discuss recent ideas concerning the therapeutic targeting of these cells in patients with chronic allergic diseases and their potential use in cancer immunotherapy.
Collapse
|
94
|
Miller MM, Reinhardt RL. The Heterogeneity, Origins, and Impact of Migratory iILC2 Cells in Anti-helminth Immunity. Front Immunol 2020; 11:1594. [PMID: 32793230 PMCID: PMC7390839 DOI: 10.3389/fimmu.2020.01594] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2020] [Accepted: 06/16/2020] [Indexed: 12/12/2022] Open
Abstract
Soil-transmitted helminths represent a major global health burden with infections and infection-related comorbidities causing significant reductions in the quality of life for individuals living in endemic areas. Repeated infections and chronic colonization by these large extracellular worms in mammals led to the evolution of type-2 immunity characterized by the production of the type-2 cytokines interleukin (IL)-4, IL-5, and IL-13. Although a number of adaptive and innate immune cells produce type-2 cytokines, a key cellular source in the context of helminth infection is group 2 innate lymphoid cells (ILC2s). ILC2s promote mucosal barrier homeostasis, integrity, and repair by rapidly responding to epithelial cues in mucosal tissues. Though tissue-resident ILC2s (nILC2s) have been studied in detail over the last decade, considerably less is known with regard to a subset of inflammatory ILC2s (iILC2s) that migrate to the lungs of mice early after Nippostrongylus brasiliensis infection and are potent early producers of type-2 cytokines. This review will discuss the relationship and differences between nILC2s and iILC2s that establish their unique roles in anti-helminth immunity. We have placed particular emphasis on studies investigating iILC2 origin, function, and their potential long-term contribution to tissue-resident ILC2 reservoirs in settings of helminth infection.
Collapse
Affiliation(s)
- Mindy M Miller
- Department of Biomedical Research, National Jewish Health, Denver, CO, United States
| | - R Lee Reinhardt
- Department of Biomedical Research, National Jewish Health, Denver, CO, United States.,Department of Immunology and Microbiology, University of Colorado-Anschutz Medical, Aurora, CO, United States
| |
Collapse
|
95
|
Loh Z, Simpson J, Ullah A, Zhang V, Gan WJ, Lynch JP, Werder RB, Sikder AA, Lane K, Sim CB, Porrello E, Mazzone SB, Sly PD, Steptoe RJ, Spann KM, Sukkar MB, Upham JW, Phipps S. HMGB1 amplifies ILC2-induced type-2 inflammation and airway smooth muscle remodelling. PLoS Pathog 2020; 16:e1008651. [PMID: 32658914 PMCID: PMC7377495 DOI: 10.1371/journal.ppat.1008651] [Citation(s) in RCA: 36] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2020] [Revised: 07/23/2020] [Accepted: 05/24/2020] [Indexed: 12/16/2022] Open
Abstract
Type-2 immunity elicits tissue repair and homeostasis, however dysregulated type-2 responses cause aberrant tissue remodelling, as observed in asthma. Severe respiratory viral infections in infancy predispose to later asthma, however, the processes that mediate tissue damage-induced type-2 inflammation and the origins of airway remodelling remain ill-defined. Here, using a preclinical mouse model of viral bronchiolitis, we find that increased epithelial and mesenchymal high-mobility group box 1 (HMGB1) expression is associated with increased numbers of IL-13-producing type-2 innate lymphoid cell (ILC2s) and the expansion of the airway smooth muscle (ASM) layer. Anti-HMGB1 ablated lung ILC2 numbers and ASM growth in vivo, and inhibited ILC2-mediated ASM cell proliferation in a co-culture model. Furthermore, we identified that HMGB1/RAGE (receptor for advanced glycation endproducts) signalling mediates an ILC2-intrinsic IL-13 auto-amplification loop. In summary, therapeutic targeting of the HMGB1/RAGE signalling axis may act as a novel asthma preventative by dampening ILC2-mediated type-2 inflammation and associated ASM remodelling. Asthma can start at any time in life, although most often begins in early childhood. Wheezy viral bronchiolitis is a major independent risk factor for subsequent asthma. However, key knowledge gaps exist in relation to the sequelae of severe viral bronchiolitis and the pathogenic processes that promote type-2 inflammation and airway wall remodelling, cardinal features of asthma. Our study addresses this gap by identifying high-mobility group box 1 as a pathogenic cytokine that contributes to group 2 innate lymphoid cell-induced airway smooth muscle growth.
Collapse
Affiliation(s)
- Zhixuan Loh
- School of Biomedical Sciences, The University of Queensland, Queensland, Australia
- Institute for Molecular Bioscience, The University of Queensland, Queensland, Australia
| | - Jennifer Simpson
- School of Biomedical Sciences, The University of Queensland, Queensland, Australia
- QIMR Berghofer Medical Research Institute, Queensland, Australia
| | - Ashik Ullah
- School of Biomedical Sciences, The University of Queensland, Queensland, Australia
- QIMR Berghofer Medical Research Institute, Queensland, Australia
| | - Vivian Zhang
- QIMR Berghofer Medical Research Institute, Queensland, Australia
| | - Wan J. Gan
- School of Biomedical Sciences, The University of Queensland, Queensland, Australia
| | - Jason P. Lynch
- School of Biomedical Sciences, The University of Queensland, Queensland, Australia
- QIMR Berghofer Medical Research Institute, Queensland, Australia
| | - Rhiannon B. Werder
- School of Biomedical Sciences, The University of Queensland, Queensland, Australia
- QIMR Berghofer Medical Research Institute, Queensland, Australia
| | - Al Amin Sikder
- School of Biomedical Sciences, The University of Queensland, Queensland, Australia
- QIMR Berghofer Medical Research Institute, Queensland, Australia
| | - Katie Lane
- School of Biomedical Sciences, The University of Queensland, Queensland, Australia
| | - Choon Boon Sim
- Murdoch Children’s Research Institute, The Royal Children’s Hospital, Melbourne, Australia
| | - Enzo Porrello
- Murdoch Children’s Research Institute, The Royal Children’s Hospital, Melbourne, Australia
| | - Stuart B. Mazzone
- School of Biomedical Sciences, The University of Queensland, Queensland, Australia
- Department of Anatomy and Neuroscience, University of Melbourne, Victoria, Australia
| | - Peter D. Sly
- Children’s Health and Environment Program, Child Health Research Centre, University of Queensland, Queensland, Australia
- Australian Infectious Diseases Research Centre, The University of Queensland, Queensland, Australia
| | - Raymond J. Steptoe
- UQ Diamantina Institute, The University of Queensland, Queensland, Australia
| | - Kirsten M. Spann
- School of Biomedical Sciences, Queensland University of Technology, Queensland, Australia
| | - Maria B. Sukkar
- Graduate School of Health, Faculty of Health, University of Technology Sydney, Ultimo, NSW, Australia
| | - John W. Upham
- UQ Diamantina Institute, The University of Queensland, Queensland, Australia
| | - Simon Phipps
- School of Biomedical Sciences, The University of Queensland, Queensland, Australia
- QIMR Berghofer Medical Research Institute, Queensland, Australia
- Australian Infectious Diseases Research Centre, The University of Queensland, Queensland, Australia
- * E-mail:
| |
Collapse
|
96
|
Abstract
Although, as the major organ of gas exchange, the lung is considered a nonlymphoid organ, an interconnected network of lung-resident innate cells, including epithelial cells, dendritic cells, macrophages, and natural killer cells is crucial for its protection. These cells provide defense against a daily assault by airborne bacteria, viruses, and fungi, as well as prevent the development of cancer, allergy, and the outgrowth of commensals. Our understanding of this innate immune environment has recently changed with the discovery of a family of innate lymphoid cells (ILCs): ILC1s, ILC2s, and ILC3s. All lack adaptive antigen receptors but can provide a substantial and rapid source of IFN-γ, IL-5 and IL-13, and IL-17A or IL-22, respectively. Their ability to afford immediate protection to the lung and to influence subsequent adaptive immune responses highlights the importance of understanding ILC-regulated immunity for the design of future therapeutic interventions.
Collapse
Affiliation(s)
- Jillian L Barlow
- Medical Research Council, Laboratory of Molecular Biology, Cambridge University, Cambridgeshire CB2 0QH, United Kingdom;
| | - Andrew N J McKenzie
- Medical Research Council, Laboratory of Molecular Biology, Cambridge University, Cambridgeshire CB2 0QH, United Kingdom;
| |
Collapse
|
97
|
Schulz-Kuhnt A, Wirtz S, Neurath MF, Atreya I. Regulation of Human Innate Lymphoid Cells in the Context of Mucosal Inflammation. Front Immunol 2020; 11:1062. [PMID: 32655549 PMCID: PMC7324478 DOI: 10.3389/fimmu.2020.01062] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2020] [Accepted: 05/04/2020] [Indexed: 12/13/2022] Open
Abstract
Since their identification as a unique cell population, innate lymphoid cells (ILCs) have revolutionized our understanding of immune responses, leaving their impact on multiple inflammatory and fibrotic pathologies without doubt. Thus, a tightly controlled regulation of local ILC numbers and their activity is of crucial importance. Even though this has been extensively studied in murine ILCs in the last few years, our knowledge of human ILCs is still lagging behind. Our review article will therefore summarize recent insights into the function of human ILCs and will particularly focus on their regulation under inflammatory conditions. The quality and intensity of ILC involvement into local immune responses at mucosal sites of the human body can potentially be modulated via three different axes: (1) activation of tissue-resident mature ILCs, (2) plasticity and local transdifferentiation of specific ILC subsets, and (3) tissue migration and accumulation of peripheral ILCs. Despite a still ongoing scientific effort in this field, already existing data on the fate of human ILCs under different pathologic conditions clearly indicate that all three of these mechanisms are of relevance for the clinical course of chronic inflammatory and autoimmune diseases and might likewise provide new target structures for future therapeutic strategies.
Collapse
Affiliation(s)
| | | | | | - Imke Atreya
- Department of Medicine 1, University Hospital of Erlangen, Erlangen, Germany
| |
Collapse
|
98
|
Xiong T, Attar M, Gnirck AC, Wunderlich M, Becker M, Rickassel C, Puelles VG, Meyer-Schwesinger C, Wiech T, Nies JF, Divivier M, Fuchs T, Schulze Zur Wiesch J, Taipaleenmäki H, Hoxha E, Wirtz S, Huber TB, Panzer U, Turner JE. Interleukin-9 protects from early podocyte injury and progressive glomerulosclerosis in Adriamycin-induced nephropathy. Kidney Int 2020; 98:615-629. [PMID: 32446933 DOI: 10.1016/j.kint.2020.04.036] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2019] [Revised: 03/29/2020] [Accepted: 04/01/2020] [Indexed: 02/08/2023]
Abstract
A wide spectrum of immunological functions has been attributed to Interleukin 9 (IL-9), including effects on the survival and proliferation of immune and parenchymal cells. In recent years, emerging evidence suggests that IL-9 expression can promote tissue repair in inflammatory conditions. However, data about the involvement of IL-9 in kidney tissue protection is very limited. Here, we investigated the role of IL-9 in Adriamycin-induced nephropathy (AN), a mouse model for proteinuric chronic kidney disease. Compared to wild type mice, IL-9 knockout (Il9-/-) mice with AN displayed accelerated development of proteinuria, aggravated glomerulosclerosis and deterioration of kidney function. At an early stage of disease, the Il9-/- mice already displayed a higher extent of glomerular podocyte injury and loss of podocyte number compared to wild type mice. In the kidney, T cells and innate lymphoid cells produced IL-9. However, selective deficiency of IL-9 in the innate immune system in Il9-/-Rag2-/- mice that lack T and B cells did not alter the outcome of AN, indicating that IL-9 derived from the adaptive immune system was the major driver of tissue protection in this model. Mechanistically, we could show that podocytes expressed the IL-9 receptor in vivo and that IL-9 signaling protects podocytes from Adriamycin-induced apoptosis in vitro. Finally, in vivo treatment with IL-9 effectively protected wild type mice from glomerulosclerosis and kidney failure in the AN model. The detection of increased serum IL-9 levels in patients with primary focal and segmental glomerulosclerosis further suggests that IL-9 production is induced by glomerular injury in humans. Thus, IL-9 confers protection against experimental glomerulosclerosis, identifying the IL-9 pathway as a potential therapeutic target in proteinuric chronic kidney disease.
Collapse
Affiliation(s)
- Tingting Xiong
- III. Department of Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Madena Attar
- III. Department of Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Ann-Christin Gnirck
- III. Department of Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Malte Wunderlich
- III. Department of Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Martina Becker
- III. Department of Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Constantin Rickassel
- III. Department of Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Victor G Puelles
- III. Department of Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Catherine Meyer-Schwesinger
- Department of Cellular and Integrative Physiology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Thorsten Wiech
- Institute of Pathology, Nephropathology Section, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Jasper F Nies
- III. Department of Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Mylène Divivier
- Institute of Clinical Chemistry and Laboratory Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Tobias Fuchs
- Institute of Clinical Chemistry and Laboratory Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | | | - Hanna Taipaleenmäki
- Department of Trauma, Hand and Reconstructive Surgery, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Elion Hoxha
- III. Department of Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Stefan Wirtz
- Department of Internal Medicine 1, Friedrich Alexander University Erlangen-Nürnberg, University Medical Center Erlangen, Erlangen, Germany
| | - Tobias B Huber
- III. Department of Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Ulf Panzer
- III. Department of Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Jan-Eric Turner
- III. Department of Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany.
| |
Collapse
|
99
|
Kharwadkar R, Ulrich BJ, Abdul Qayum A, Koh B, Licona-Limón P, Flavell RA, Kaplan MH. Expression Efficiency of Multiple Il9 Reporter Alleles Is Determined by Cell Lineage. Immunohorizons 2020; 4:282-291. [PMID: 32439753 DOI: 10.4049/immunohorizons.1900082] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2019] [Accepted: 05/07/2020] [Indexed: 11/19/2022] Open
Abstract
Generation of allelic gene reporter mice has provided a powerful tool to study gene function in vivo. In conjunction with imaging technologies, reporter mouse models facilitate studies of cell lineage tracing, live cell imaging, and gene expression in the context of diseases. Although there are several advantages to using reporter mice, caution is important to ensure the fidelity of the reporter protein representing the gene of interest. In this study, we compared the efficiency of two Il9 reporter strains Il9citrine and Il9GFP in representing IL-9-producing CD4+ TH9 cells. Although both alleles show high specificity in IL-9-expressing populations, we observed that the Il9GFP allele visualized a much larger proportion of the IL-9-producing cells in culture than the Il9citrine reporter allele. In defining the mechanistic basis for these differences, chromatin immunoprecipitation and chromatin accessibility assay showed that the Il9citrine allele was transcriptionally less active in TH9 cells compared with the wild-type allele. The Il9citrine allele also only captured a fraction of IL-9-expressing bone marrow-derived mast cells. In contrast, the Il9 citrine reporter detected Il9 expression in type 2 innate lymphoid cells at a greater percentage than could be identified by IL-9 intracellular cytokine staining. Taken together, our findings demonstrate that the accuracy of IL-9 reporter mouse models may vary with the cell type being examined. These studies demonstrate the importance of choosing appropriate reporter mouse models that are optimal for detecting the cell type of interest as well as the accuracy of conclusions.
Collapse
Affiliation(s)
- Rakshin Kharwadkar
- Herman B Wells Center for Pediatric Research, Department of Pediatrics, Indiana University School of Medicine, Indianapolis, IN 46202.,Department of Biochemistry and Molecular Biology, Indiana University School of Medicine, Indianapolis, IN 46202
| | - Benjamin J Ulrich
- Herman B Wells Center for Pediatric Research, Department of Pediatrics, Indiana University School of Medicine, Indianapolis, IN 46202.,Department of Microbiology and Immunology, Indiana University School of Medicine, Indianapolis, IN 46202
| | - Amina Abdul Qayum
- Herman B Wells Center for Pediatric Research, Department of Pediatrics, Indiana University School of Medicine, Indianapolis, IN 46202
| | - Byunghee Koh
- Herman B Wells Center for Pediatric Research, Department of Pediatrics, Indiana University School of Medicine, Indianapolis, IN 46202.,Department of Microbiology and Immunology, Indiana University School of Medicine, Indianapolis, IN 46202
| | - Paula Licona-Limón
- Departamento de Biologia Celular y del Desarrollo, Instituto de Fisiologia Celular, Universidad Nacional Autónoma de México, 04020 Mexico City, Mexico; and
| | - Richard A Flavell
- Department of Immunobiology, Yale University School of Medicine, New Haven, CT 06510
| | - Mark H Kaplan
- Herman B Wells Center for Pediatric Research, Department of Pediatrics, Indiana University School of Medicine, Indianapolis, IN 46202; .,Department of Biochemistry and Molecular Biology, Indiana University School of Medicine, Indianapolis, IN 46202.,Department of Microbiology and Immunology, Indiana University School of Medicine, Indianapolis, IN 46202
| |
Collapse
|
100
|
Ebihara T. Dichotomous Regulation of Acquired Immunity by Innate Lymphoid Cells. Cells 2020; 9:cells9051193. [PMID: 32403291 PMCID: PMC7290502 DOI: 10.3390/cells9051193] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2020] [Revised: 05/04/2020] [Accepted: 05/08/2020] [Indexed: 12/12/2022] Open
Abstract
The concept of innate lymphoid cells (ILCs) includes both conventional natural killer (NK) cells and helper ILCs, which resemble CD8+ killer T cells and CD4+ helper T cells in acquired immunity, respectively. Conventional NK cells are migratory cytotoxic cells that find tumor cells or cells infected with microbes. Helper ILCs are localized at peripheral tissue and are responsible for innate helper-cytokine production. Helper ILCs are classified into three subpopulations: TH1-like ILC1s, TH2-like ILC2s, and TH17/TH22-like ILC3s. Because of the functional similarities between ILCs and T cells, ILCs can serve as an innate component that augments each corresponding type of acquired immunity. However, the physiological functions of ILCs are more plastic and complicated than expected and are affected by environmental cues and types of inflammation. Here, we review recent advances in understanding the interaction between ILCs and acquired immunity, including T- and B-cell responses at various conditions. Immune suppressive activities by ILCs in particular are discussed in comparison to their immune stimulatory effects to gain precise knowledge of ILC biology and the physiological relevance of ILCs in human diseases.
Collapse
Affiliation(s)
- Takashi Ebihara
- Department of Medical Biology, Akita University Graduate School of Medicine Affiliation, 1-1-1 Hondo, Akita 010-8543, Japan
| |
Collapse
|