1
|
Bennstein SB, Uhrberg M. Circulating innate lymphoid cells (cILCs): Unconventional lymphocytes with hidden talents. J Allergy Clin Immunol 2024; 154:523-536. [PMID: 39046403 DOI: 10.1016/j.jaci.2024.06.016] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2024] [Revised: 05/16/2024] [Accepted: 06/28/2024] [Indexed: 07/25/2024]
Abstract
Innate lymphoid cells (ILCs) are a group of lymphocytes that are devoid of antigen-specific receptors and are mainly found in tissues. The subtypes ILC1, 2, and 3 mirror T-cell functionality in terms of cytokine production and expression of key transcription factors. Although the majority of ILCs are found in tissue (tILCs), they have also been described within the circulation (cILCs). As a result of their better accessibility and putative prognostic value, human cILCs are getting more and more attention in clinical research. However, cILCs are in many aspects functionally distinct from their tILC counterparts. In fact, from the 3 ILC subsets found within the circulation, only for cILC2s could a clear functional correspondence to their tissue counterparts be established. Indeed, cILC2s are emerging as a major driver of allergic reactions with a particular role in asthma. In contrast, recent studies revealed that cILC1s and cILC3s are predominantly in an immature state and constitute progenitors for natural killer cells and ILCs, respectively. We provide an overview about the phenotype and function of the different cILC subtypes compared to tILCs in health and disease, including transcriptomic signatures, frequency dynamics, and potential clinical value. Furthermore, we will highlight the dynamics of the NKp44+ ILC3 subset, which emerges as prognostic marker in peripheral blood for inflammatory bowel disease and leukemia.
Collapse
Affiliation(s)
- Sabrina B Bennstein
- Institute for Transplantation Diagnostics and Cell Therapeutics, Medical Faculty and University Hospital Düsseldorf, Heinrich Heine University Düsseldorf, Düsseldorf, Germany; Institute of Immunology, Faculty of Medicine, RWTH Aachen University, Aachen, Germany.
| | - Markus Uhrberg
- Institute for Transplantation Diagnostics and Cell Therapeutics, Medical Faculty and University Hospital Düsseldorf, Heinrich Heine University Düsseldorf, Düsseldorf, Germany.
| |
Collapse
|
2
|
Qin M, Fang Y, Zheng Q, Peng M, Wang L, Sang X, Cao G. Tissue microenvironment induces tissue specificity of ILC2. Cell Death Discov 2024; 10:324. [PMID: 39013890 PMCID: PMC11252336 DOI: 10.1038/s41420-024-02096-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2024] [Revised: 07/04/2024] [Accepted: 07/09/2024] [Indexed: 07/18/2024] Open
Abstract
Type 2 innate lymphoid cells were found to be members of the innate immune cell family, which is involved in innate and adaptive immunity to resist the invasion of foreign antigens and induce allergic reactions caused by allergens. The advancement of ILC2 research has pointed out that ILC2s have a high degree of diversity, challenging the notion of their homogeneity as a cellular population. An increasing number of studies indicate that ILC2 is a cell population with tissue specificity which can be induced by the tissue microenvironment. In addition, crosstalk between tissues can change ILC2 functions of migration and activation. Here, we emphasize that ILC2 undergoes adaptive changes under the regulation of the tissue microenvironment and distant tissues, thereby coordinating the organization's operation. In addition, ILC2 alterations induced by the tissue microenvironment are not limited to the ILC2 cell population, and ILC2 can also transdifferentiate into another class of ILC cell population (ILC1 or ILC3). In this review, we summarized the tissue-specific effects of ILC2 by tissue microenvironment and focused on the function of ILC2 in inter-tissue crosstalk. Lastly, we discussed the transdifferentiations of ILC2 caused by the abnormal change in tissue environment.
Collapse
Affiliation(s)
- Minjing Qin
- School of Pharmacy, Zhejiang Chinese Medical University, Hangzhou, China
| | - Yuanyuan Fang
- School of Pharmacy, Zhejiang Chinese Medical University, Hangzhou, China
| | - Qitong Zheng
- School of Pharmacy, Zhejiang Chinese Medical University, Hangzhou, China
| | - Mengyun Peng
- School of Pharmacy, Zhejiang Chinese Medical University, Hangzhou, China
| | - Lu Wang
- School of Pharmacy, Zhejiang Chinese Medical University, Hangzhou, China
| | - Xia'nan Sang
- School of Pharmacy, Zhejiang Chinese Medical University, Hangzhou, China.
| | - Gang Cao
- School of Pharmacy, Zhejiang Chinese Medical University, Hangzhou, China.
| |
Collapse
|
3
|
García M, Carrasco García A, Weigel W, Christ W, Lira-Junior R, Wirth L, Tauriainen J, Maleki K, Vanoni G, Vaheri A, Mäkelä S, Mustonen J, Nordgren J, Smed-Sörensen A, Strandin T, Mjösberg J, Klingström J. Innate lymphoid cells are activated in HFRS, and their function can be modulated by hantavirus-induced type I interferons. PLoS Pathog 2024; 20:e1012390. [PMID: 39038044 PMCID: PMC11293681 DOI: 10.1371/journal.ppat.1012390] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2024] [Revised: 08/01/2024] [Accepted: 07/03/2024] [Indexed: 07/24/2024] Open
Abstract
Hantaviruses cause the acute zoonotic diseases hemorrhagic fever with renal syndrome (HFRS) and hantavirus pulmonary syndrome (HPS). Infected patients show strong systemic inflammation and immune cell activation. NK cells are highly activated in HFRS, suggesting that also other innate lymphoid cells (ILCs) might be responding to infection. Here, we characterized peripheral ILC responses, and measured plasma levels of soluble factors and plasma viral load, in 17 Puumala virus (PUUV)-infected HFRS patients. This revealed an increased frequency of ILC2 in patients, in particular the ILC2 lineage-committed c-Kitlo ILC2 subset. Patients' ILCs showed an activated profile with increased proliferation and displayed altered expression of several homing markers. How ILCs are activated during viral infection is largely unknown. When analyzing PUUV-mediated activation of ILCs in vitro we observed that this was dependent on type I interferons, suggesting a role for type I interferons-produced in response to virus infection-in the activation of ILCs. Further, stimulation of naïve ILC2s with IFN-β affected ILC2 cytokine responses in vitro, causing decreased IL-5 and IL-13, and increased IL-10, CXCL10, and GM-CSF secretion. These results show that ILCs are activated in HFRS patients and suggest that the classical antiviral type I IFNs are involved in shaping ILC functions.
Collapse
Affiliation(s)
- Marina García
- Center for Infectious Medicine, Department of Medicine Huddinge, Karolinska Institutet, Stockholm, Sweden
| | - Anna Carrasco García
- Center for Infectious Medicine, Department of Medicine Huddinge, Karolinska Institutet, Stockholm, Sweden
| | - Whitney Weigel
- Center for Infectious Medicine, Department of Medicine Huddinge, Karolinska Institutet, Stockholm, Sweden
| | - Wanda Christ
- Center for Infectious Medicine, Department of Medicine Huddinge, Karolinska Institutet, Stockholm, Sweden
| | - Ronaldo Lira-Junior
- Section of Oral Diagnostics and Surgery, Division of Oral Diagnostics and Rehabilitation, Department of Dental Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Lorenz Wirth
- Center for Infectious Medicine, Department of Medicine Huddinge, Karolinska Institutet, Stockholm, Sweden
- Mechanistic & Structural Biology, Discovery Sciences, R&D, AstraZeneca, Gothenburg, Sweden
| | - Johanna Tauriainen
- Center for Infectious Medicine, Department of Medicine Huddinge, Karolinska Institutet, Stockholm, Sweden
| | - Kimia Maleki
- Center for Infectious Medicine, Department of Medicine Huddinge, Karolinska Institutet, Stockholm, Sweden
| | - Giulia Vanoni
- Institut Curie, PSL University, Inserm, Immunity and Cancer, Paris, France
| | - Antti Vaheri
- Department of Virology, Faculty of Medicine, University of Helsinki, Helsinki, Finland
| | - Satu Mäkelä
- Department of Internal Medicine, Tampere University Hospital, Tampere, Finland
- Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland
| | - Jukka Mustonen
- Department of Internal Medicine, Tampere University Hospital, Tampere, Finland
- Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland
| | - Johan Nordgren
- Division of Molecular Medicine and Virology, Department of Biomedical and Clinical Sciences, Linköping University, Linköping, Sweden
| | - Anna Smed-Sörensen
- Division of Immunology and Allergy, Department of Medicine Solna, Karolinska Institutet, Karolinska University Hospital, Stockholm, Sweden
| | - Tomas Strandin
- Department of Virology, Faculty of Medicine, University of Helsinki, Helsinki, Finland
- Division of Immunology and Allergy, Department of Medicine Solna, Karolinska Institutet, Karolinska University Hospital, Stockholm, Sweden
| | - Jenny Mjösberg
- Center for Infectious Medicine, Department of Medicine Huddinge, Karolinska Institutet, Stockholm, Sweden
| | - Jonas Klingström
- Center for Infectious Medicine, Department of Medicine Huddinge, Karolinska Institutet, Stockholm, Sweden
- Division of Molecular Medicine and Virology, Department of Biomedical and Clinical Sciences, Linköping University, Linköping, Sweden
| |
Collapse
|
4
|
Lahire S, Fichel C, Rubaszewski O, Lerévérend C, Audonnet S, Visneux V, Perotin JM, Deslée G, Le Jan S, Potteaux S, Le Naour R, Pommier A. Elastin-derived peptides favor type 2 innate lymphoid cells in chronic obstructive pulmonary disease. Am J Physiol Lung Cell Mol Physiol 2024; 326:L812-L820. [PMID: 38712445 DOI: 10.1152/ajplung.00306.2023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2023] [Revised: 03/22/2024] [Accepted: 04/22/2024] [Indexed: 05/08/2024] Open
Abstract
Chronic obstructive pulmonary disease (COPD) is a condition characterized by chronic airway inflammation and obstruction, primarily caused by tobacco smoking. Although the involvement of immune cells in COPD pathogenesis is well established, the contribution of innate lymphoid cells (ILCs) remains poorly understood. ILCs are a type of innate immune cells that participate in tissue remodeling processes, but their specific role in COPD has not been fully elucidated. During COPD, the breakdown of pulmonary elastin generates elastin peptides that elicit biological activities on immune cells. This study aimed to investigate the presence of ILC in patients with COPD and examine the impact of elastin peptides on their functionality. Our findings revealed an elevated proportion of ILC2 in the peripheral blood of patients with COPD, and a general activation of ILC as indicated by an increase in their cytokine secretion capacity. Notably, our study demonstrated that serum from patients with COPD promotes ILC2 phenotype, likely due to the elevated concentration of IL-5, a cytokine known to favor ILC2 activation. Furthermore, we uncovered that this increase in IL-5 secretion is partially attributed to its secretion by macrophages upon stimulation by elastin peptides, suggesting an indirect role of elastin peptides on ILC in COPD. These findings shed light on the involvement of ILC in COPD and provide insights into the potential interplay between elastin breakdown, immune cells, and disease progression. Further understanding of the mechanisms underlying ILC activation and their interaction with elastin peptides could contribute to the development of novel therapeutic strategies for COPD management.NEW & NOTEWORTHY Elastin-derived peptides, generated following alveolar degradation during emphysema in patients with COPD, are able to influence the response of type 2 innate lymphoid cells. We show that the orientation of innate lymphoid cells in patients with COPD is shifted toward a type 2 profile and that elastin peptides are indirectly participating in that shift through their influence of macrophages, which in turn impact innate lymphoid cells.
Collapse
Affiliation(s)
- Sarah Lahire
- Université de Reims Champagne Ardenne, Immuno-Régulation dans les Maladies Auto-Immunes, Inflammatoires et le Cancer (IRMAIC), Reims, France
| | - Caroline Fichel
- Université de Reims Champagne Ardenne, Immuno-Régulation dans les Maladies Auto-Immunes, Inflammatoires et le Cancer (IRMAIC), Reims, France
| | - Océane Rubaszewski
- Université de Reims Champagne Ardenne, Immuno-Régulation dans les Maladies Auto-Immunes, Inflammatoires et le Cancer (IRMAIC), Reims, France
| | - Cédric Lerévérend
- Université de Reims Champagne Ardenne, Immuno-Régulation dans les Maladies Auto-Immunes, Inflammatoires et le Cancer (IRMAIC), Reims, France
- Institut Godinot, Unicancer, Reims, France
| | - Sandra Audonnet
- Université de Reims Champagne Ardenne, Plateforme de cytométrie en flux, URCACyt, Reims, France
| | - Vincent Visneux
- CHU de Reims, Service des maladies respiratoires, Reims, France
| | - Jeanne-Marie Perotin
- CHU de Reims, Service des maladies respiratoires, Reims, France
- Université de Reims Champagne Ardenne, Inserm UMR-S 1250 Pathologies Pulmonaires et Plasticité Cellulaire (P3Cell), Reims, France
| | - Gaëtan Deslée
- CHU de Reims, Service des maladies respiratoires, Reims, France
- Université de Reims Champagne Ardenne, Inserm UMR-S 1250 Pathologies Pulmonaires et Plasticité Cellulaire (P3Cell), Reims, France
| | - Sébastien Le Jan
- Université de Reims Champagne Ardenne, Immuno-Régulation dans les Maladies Auto-Immunes, Inflammatoires et le Cancer (IRMAIC), Reims, France
| | - Stéphane Potteaux
- Université de Reims Champagne Ardenne, Immuno-Régulation dans les Maladies Auto-Immunes, Inflammatoires et le Cancer (IRMAIC), Reims, France
- Institut Godinot, Unicancer, Reims, France
- Délégation régionale Inserm Paris Ile-de France Centre Nord, Paris, France
| | - Richard Le Naour
- Université de Reims Champagne Ardenne, Immuno-Régulation dans les Maladies Auto-Immunes, Inflammatoires et le Cancer (IRMAIC), Reims, France
| | - Arnaud Pommier
- Université de Reims Champagne Ardenne, Immuno-Régulation dans les Maladies Auto-Immunes, Inflammatoires et le Cancer (IRMAIC), Reims, France
| |
Collapse
|
5
|
Lu HF, Zhou YC, Luo DD, Yang DH, Wang XJ, Cheng BH, Zeng XH. ILC2s: Unraveling the innate immune orchestrators in allergic inflammation. Int Immunopharmacol 2024; 131:111899. [PMID: 38513576 DOI: 10.1016/j.intimp.2024.111899] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/01/2024] [Revised: 03/05/2024] [Accepted: 03/17/2024] [Indexed: 03/23/2024]
Abstract
The prevalence rate of allergic diseases including asthma, atopic rhinitis (AR) and atopic dermatitis (AD) has been significantly increasing in recent decades due to environmental changes and social developments. With the study of innate lymphoid cells, the crucial role played by type 2 innate lymphoid cells (ILC2s) have been progressively unveiled in allergic diseases. ILC2s, which are a subset of innate lymphocytes initiate allergic responses. They respond swiftly during the onset of allergic reactions and produce type 2 cytokines, working in conjunction with T helper type 2 (Th2) cells to induce and sustain type 2 immune responses. The role of ILC2s represents an intriguing frontier in immunology; however, the intricate immune mechanisms of ILC2s in allergic responses remain relatively poorly understood. To gain a comphrehensive understanding of the research progress of ILC2, we summarize recent advances in ILC2s biology in pathologic allergic inflammation to inspire novel approaches for managing allergic diseases.
Collapse
Affiliation(s)
- Hui-Fei Lu
- Department of Graduate and Scientific Research, Zhuhai Campus of Zunyi Medical University, Zhuhai 519041, China; Department of Otolaryngology, Shenzhen Key Laboratory of Otolaryngology, Shenzhen Institute of Otolaryngology, Shenzhen Longgang Otolaryngology Hospital, Shenzhen, 518172, China
| | - Yi-Chi Zhou
- Department of Gastroenterology, Beijing University of Chinese Medicine Shenzhen Hospital (Longgang), Shenzhen 518172, China
| | - Dan-Dan Luo
- Department of Graduate and Scientific Research, Zhuhai Campus of Zunyi Medical University, Zhuhai 519041, China
| | - Dun-Hui Yang
- Department of Otolaryngology, Shenzhen Key Laboratory of Otolaryngology, Shenzhen Institute of Otolaryngology, Shenzhen Longgang Otolaryngology Hospital, Shenzhen, 518172, China
| | - Xi-Jia Wang
- Department of Graduate and Scientific Research, Zhuhai Campus of Zunyi Medical University, Zhuhai 519041, China
| | - Bao-Hui Cheng
- Department of Otolaryngology, Shenzhen Key Laboratory of Otolaryngology, Shenzhen Institute of Otolaryngology, Shenzhen Longgang Otolaryngology Hospital, Shenzhen, 518172, China.
| | - Xian-Hai Zeng
- Department of Graduate and Scientific Research, Zhuhai Campus of Zunyi Medical University, Zhuhai 519041, China; Department of Otolaryngology, Shenzhen Key Laboratory of Otolaryngology, Shenzhen Institute of Otolaryngology, Shenzhen Longgang Otolaryngology Hospital, Shenzhen, 518172, China.
| |
Collapse
|
6
|
van Zelst CM, in ’t Veen JC, Krabbendam L, de Boer GM, de Bruijn MJ, van Nimwegen M, van der Ploeg EK, van Uden D, Stadhouders R, Tramper-Stranders GA, Hendriks RW, Braunstahl GJ. Aberrant characteristics of peripheral blood innate lymphoid cells in COPD, independent of smoking history. ERJ Open Res 2024; 10:00652-2023. [PMID: 38375427 PMCID: PMC10875467 DOI: 10.1183/23120541.00652-2023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2023] [Accepted: 12/19/2023] [Indexed: 02/21/2024] Open
Abstract
Background Distinguishing asthma and COPD can pose challenges in clinical practice. Increased group 1 innate lymphoid cells (ILC1s) have been found in the lungs and peripheral blood of COPD patients, while asthma is associated with elevated levels of ILC2s. However, it is unclear whether the inflammatory characteristics of ILC1s and ILC2s differ between COPD and asthma. This study aims to compare peripheral blood ILC subsets and their expression of inflammatory markers in COPD patients, asthma patients and controls. Methods The study utilised multi-colour flow cytometry to analyse peripheral blood ILC populations in clinically stable COPD patients (n=38), asthma patients (n=37), and smoking (n=19) and non-smoking (n=16) controls. Results Proportions of peripheral blood inflammatory CD4+ ILC1s were significantly higher in COPD patients than in asthma. Proportions of CD4- ILC1s were increased in COPD patients compared to asthma patients and smoking controls. Frequencies of CD117- ILC2s were significantly reduced in COPD patients compared with asthma patients. In contrast, the fraction of inflammatory CD45RO+ cells within the CD117- ILC2 population was significantly increased. Principal component analyses showed that combined features of the circulating ILC compartment separated COPD patients from asthma patients and both control groups. Conclusion Our in-depth characterisation of ILC1 and ILC2 populations in peripheral blood revealed significant differences in their phenotypes between COPD and asthma patients and smoking or non-smoking controls. These findings suggest a role for both ILC subsets in COPD disease pathology, independent of smoking history, and may have implications for patient stratification and therapy development.
Collapse
Affiliation(s)
- Cathelijne M. van Zelst
- Department of Pulmonology, Franciscus Gasthuis and Vlietland, Rotterdam, the Netherlands
- Department of Pulmonary Medicine, Erasmus MC, University Medical Center, Rotterdam, the Netherlands
| | - Johannes C.C.M. in ’t Veen
- Department of Pulmonology, Franciscus Gasthuis and Vlietland, Rotterdam, the Netherlands
- Department of Pulmonary Medicine, Erasmus MC, University Medical Center, Rotterdam, the Netherlands
| | - Lisette Krabbendam
- Department of Pulmonary Medicine, Erasmus MC, University Medical Center, Rotterdam, the Netherlands
| | - Geertje M. de Boer
- Department of Pulmonology, Franciscus Gasthuis and Vlietland, Rotterdam, the Netherlands
- Department of Pulmonary Medicine, Erasmus MC, University Medical Center, Rotterdam, the Netherlands
| | - Marjolein J.W. de Bruijn
- Department of Pulmonary Medicine, Erasmus MC, University Medical Center, Rotterdam, the Netherlands
| | - Menno van Nimwegen
- Department of Pulmonary Medicine, Erasmus MC, University Medical Center, Rotterdam, the Netherlands
| | - Esmee K. van der Ploeg
- Department of Pulmonary Medicine, Erasmus MC, University Medical Center, Rotterdam, the Netherlands
| | - Denise van Uden
- Department of Pulmonary Medicine, Erasmus MC, University Medical Center, Rotterdam, the Netherlands
| | - Ralph Stadhouders
- Department of Pulmonary Medicine, Erasmus MC, University Medical Center, Rotterdam, the Netherlands
| | - Gerdien A. Tramper-Stranders
- Department of Pulmonology, Franciscus Gasthuis and Vlietland, Rotterdam, the Netherlands
- Department of Pulmonary Medicine, Erasmus MC, University Medical Center, Rotterdam, the Netherlands
| | - Rudi W. Hendriks
- Department of Pulmonary Medicine, Erasmus MC, University Medical Center, Rotterdam, the Netherlands
- Shared senior authors
| | - Gert-Jan Braunstahl
- Department of Pulmonology, Franciscus Gasthuis and Vlietland, Rotterdam, the Netherlands
- Department of Pulmonary Medicine, Erasmus MC, University Medical Center, Rotterdam, the Netherlands
- Shared senior authors
| |
Collapse
|
7
|
Hegewisch-Solloa E, Nalin AP, Freud AG, Mace EM. Deciphering the localization and trajectory of human natural killer cell development. J Leukoc Biol 2023; 114:487-506. [PMID: 36869821 DOI: 10.1093/jleuko/qiad027] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2022] [Revised: 02/07/2023] [Accepted: 02/12/2023] [Indexed: 03/05/2023] Open
Abstract
Innate immune cells represent the first line of cellular immunity, comprised of both circulating and tissue-resident natural killer cells and innate lymphoid cells. These innate lymphocytes arise from a common CD34+ progenitor that differentiates into mature natural killer cells and innate lymphoid cells. The successive stages in natural killer cell maturation are characterized by increased lineage restriction and changes to phenotype and function. Mechanisms of human natural killer cell development have not been fully elucidated, especially the role of signals that drive the spatial localization and maturation of natural killer cells. Cytokines, extracellular matrix components, and chemokines provide maturation signals and influence the trafficking of natural killer cell progenitors to peripheral sites of differentiation. Here we present the latest advances in our understanding of natural killer and innate lymphoid cell development in peripheral sites, including secondary lymphoid tissues (i.e. tonsil). Recent work in the field has provided a model for the spatial distribution of natural killer cell and innate lymphoid cell developmental intermediates in tissue and generated further insights into the developmental niche. In support of this model, future studies using multifaceted approaches seek to fully map the developmental trajectory of human natural killer cells and innate lymphoid cells in secondary lymphoid tissues.
Collapse
Affiliation(s)
- Everardo Hegewisch-Solloa
- Department of Pediatrics, Vagelos College of Physicians and Surgeons, Columbia University Irving Medical Center, 630 W 168th St. New York, NY 10032, USA
| | - Ansel P Nalin
- Biomedical Sciences Graduate Program, Medical Scientist Training Program, Comprehensive Cancer Center and The James Cancer Hospital and Solove Research Institute, The Ohio State University, 460 W 10th Ave. Columbus, OH 43210, USA
| | - Aharon G Freud
- Department of Pathology, Comprehensive Cancer Center and The James Cancer Hospital and Solove Research Institute, The Ohio State University, 460 W 12th Ave. Columbus, OH 43210, USA
| | - Emily M Mace
- Department of Pediatrics, Vagelos College of Physicians and Surgeons, Columbia University Irving Medical Center, 630 W 168th St. New York, NY 10032, USA
| |
Collapse
|
8
|
Xu G(B, Pan YX, Mei W, Chen H. Single-Cell RNA Sequencing (scRNA-seq) Identifies L1CAM as a Key Mediator between Epithelial Tuft Cell and Innate Lymphoid Cell in the Colon of Hnrnp I Knockout Mice. Biomedicines 2023; 11:2734. [PMID: 37893107 PMCID: PMC10604312 DOI: 10.3390/biomedicines11102734] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2023] [Revised: 09/26/2023] [Accepted: 10/05/2023] [Indexed: 10/29/2023] Open
Abstract
(1) Background: Knockout (KO) of heterogeneous nuclear ribonucleoprotein I (Hnrnp I) in mouse intestinal epithelial cells (IECs) induced a severe inflammatory response in the colon, followed by hyperproliferation. This study aimed to investigate the epithelial lineage dynamics and cell-cell communications that underlie inflammation and colitis. (2) Methods: Single cells were isolated from the colons of wildtype (WT) and KO mice and used in scRNA-seq. Whole colons were collected for immunofluorescence staining and cytokine assays. (3) Results: from scRNA-seq, the number of DCLK1 + colonic tuft cells was significantly higher in the Hnrnp I KO mice compared to the WT mice. This was confirmed by immunofluorescent staining of DCLK1. The DCLK1 + colonic tuft cells in KO mice developed unique communications with lymphocytes via interactions between surface L1 cell adhesion molecule (L1CAM) and integrins. In the KO mice colons, a significantly elevated level of inflammatory cytokines IL4, IL6, and IL13 were observed, which marks type-2 immune responses directed by group 2 innate lymphoid cells (ILC2s). (4) Conclusions: This study demonstrates one critical cellular function of colonic tuft cells, which facilitates type-2 immune responses by communicating with ILC2s via the L1CAM-integrins interaction. This communication promotes pro-inflammatory signaling pathways in ILC2, leading to the increased secretion of inflammatory cytokines.
Collapse
Affiliation(s)
- Guanying (Bianca) Xu
- Department of Food Science and Human Nutrition, College of Agricultural, Consumer, and Environmental Sciences, University of Illinois at Urbana-Champaign, Urbana, IL 61801, USA; (G.X.); (Y.-X.P.)
| | - Yuan-Xiang Pan
- Department of Food Science and Human Nutrition, College of Agricultural, Consumer, and Environmental Sciences, University of Illinois at Urbana-Champaign, Urbana, IL 61801, USA; (G.X.); (Y.-X.P.)
- Division of Nutritional Sciences, College of Agricultural, Consumer, and Environmental Sciences, University of Illinois at Urbana-Champaign, Urbana, IL 61801, USA
| | - Wenyan Mei
- Division of Nutritional Sciences, College of Agricultural, Consumer, and Environmental Sciences, University of Illinois at Urbana-Champaign, Urbana, IL 61801, USA
- Department of Comparative Biosciences, College of Veterinary Medicine, University of Illinois at Urbana-Champaign, Urbana, IL 61801, USA
| | - Hong Chen
- Department of Food Science and Human Nutrition, College of Agricultural, Consumer, and Environmental Sciences, University of Illinois at Urbana-Champaign, Urbana, IL 61801, USA; (G.X.); (Y.-X.P.)
- Division of Nutritional Sciences, College of Agricultural, Consumer, and Environmental Sciences, University of Illinois at Urbana-Champaign, Urbana, IL 61801, USA
| |
Collapse
|
9
|
Falquet M, Su Z, Wyss T, Ercolano G, Trabanelli S, Jandus C. Dynamic single-cell regulomes characterize human peripheral blood innate lymphoid cell subpopulations. iScience 2023; 26:107728. [PMID: 37694139 PMCID: PMC10483052 DOI: 10.1016/j.isci.2023.107728] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2023] [Revised: 07/25/2023] [Accepted: 08/21/2023] [Indexed: 09/12/2023] Open
Abstract
Innate lymphoid cells (ILCs) are plastic immune cells divided into 3 main subsets, characterized by distinct phenotypic and functional profiles. Using single cell approaches, heightened heterogeneity of mouse ILCs has been appreciated, imprinted by tissue signals that shape their transcriptome and epigenome. Intra-subset diversity has also been observed in human ILCs. However, combined transcriptomic and epigenetic analyses of single ILCs in humans are lacking. Here, we show high transcriptional and epigenetic heterogeneity among human circulating ILCs in healthy individuals. We describe phenotypically distinct subclusters and diverse chromatin accessibility within main ILC populations, compatible with differentially poised states. We validate the use of this healthy donor-based analysis as resource dataset to help inferring ILC changes occurring in disease conditions. Overall, our work provides insights in the complex human ILC biology. We anticipate it to facilitate hypothesis-driven studies in patients, without the need to perform single cell OMICs using precious patients' material.
Collapse
Affiliation(s)
- Maryline Falquet
- Department of Pathology and Immunology, Faculty of Medicine, University of Geneva, Geneva, Switzerland
- Ludwig Institute for Cancer Research, Lausanne Branch, Lausanne, Switzerland
- Geneva Center for Inflammation Research, Geneva, Switzerland
- Translational Research Center for Oncohematology, Department of Medicine, Faculty of Medicine, University of Geneva, Geneva, Switzerland
| | - Ziyang Su
- Department of Pathology and Immunology, Faculty of Medicine, University of Geneva, Geneva, Switzerland
- Ludwig Institute for Cancer Research, Lausanne Branch, Lausanne, Switzerland
- Geneva Center for Inflammation Research, Geneva, Switzerland
- Translational Research Center for Oncohematology, Department of Medicine, Faculty of Medicine, University of Geneva, Geneva, Switzerland
| | - Tania Wyss
- Department of Pathology and Immunology, Faculty of Medicine, University of Geneva, Geneva, Switzerland
- Ludwig Institute for Cancer Research, Lausanne Branch, Lausanne, Switzerland
- Geneva Center for Inflammation Research, Geneva, Switzerland
- Translational Data Science Facility, AGORA Cancer Research Center, SIB Swiss Institute of Bioinformatics, Lausanne, Switzerland
- Translational Research Center for Oncohematology, Department of Medicine, Faculty of Medicine, University of Geneva, Geneva, Switzerland
| | - Giuseppe Ercolano
- Department of Experimental Pharmacology, University of Naples Federico II, Naples, Italy
| | - Sara Trabanelli
- Department of Pathology and Immunology, Faculty of Medicine, University of Geneva, Geneva, Switzerland
- Ludwig Institute for Cancer Research, Lausanne Branch, Lausanne, Switzerland
- Geneva Center for Inflammation Research, Geneva, Switzerland
- Translational Research Center for Oncohematology, Department of Medicine, Faculty of Medicine, University of Geneva, Geneva, Switzerland
| | - Camilla Jandus
- Department of Pathology and Immunology, Faculty of Medicine, University of Geneva, Geneva, Switzerland
- Ludwig Institute for Cancer Research, Lausanne Branch, Lausanne, Switzerland
- Geneva Center for Inflammation Research, Geneva, Switzerland
- Translational Research Center for Oncohematology, Department of Medicine, Faculty of Medicine, University of Geneva, Geneva, Switzerland
| |
Collapse
|
10
|
Thio CLP, Chang YJ. The modulation of pulmonary group 2 innate lymphoid cell function in asthma: from inflammatory mediators to environmental and metabolic factors. Exp Mol Med 2023; 55:1872-1884. [PMID: 37696890 PMCID: PMC10545775 DOI: 10.1038/s12276-023-01021-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2022] [Revised: 03/21/2023] [Accepted: 03/29/2023] [Indexed: 09/13/2023] Open
Abstract
A dysregulated type 2 immune response is one of the fundamental causes of allergic asthma. Although Th2 cells are undoubtedly central to the pathogenesis of allergic asthma, the discovery of group 2 innate lymphoid cells (ILC2s) has added another layer of complexity to the etiology of this chronic disease. Through their inherent innate type 2 responses, ILC2s not only contribute to the initiation of airway inflammation but also orchestrate the recruitment and activation of other members of innate and adaptive immunity, further amplifying the inflammatory response. Moreover, ILC2s exhibit substantial cytokine plasticity, as evidenced by their ability to produce type 1- or type 17-associated cytokines under appropriate conditions, underscoring their potential contribution to nonallergic, neutrophilic asthma. Thus, understanding the mechanisms of ILC2 functions is pertinent. In this review, we present an overview of the current knowledge on ILC2s in asthma and the regulatory factors that modulate lung ILC2 functions in various experimental mouse models of asthma and in humans.
Collapse
Affiliation(s)
| | - Ya-Jen Chang
- Institute of Biomedical Sciences, Academia Sinica, Taipei City, 115, Taiwan.
- Institute of Translational Medicine and New Drug Development, China Medical University, Taichung City, 404, Taiwan.
| |
Collapse
|
11
|
Del Zotto G, Vacca P, Moretta L, Quatrini L. CPHEN-15: Comprehensive phenotyping of human peripheral blood helper-ILCs by flow cytometry. Cytometry A 2023; 103:378-382. [PMID: 36708139 DOI: 10.1002/cyto.a.24717] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2022] [Revised: 01/16/2023] [Accepted: 01/18/2023] [Indexed: 01/29/2023]
Abstract
Innate lymphoid cells (ILCs) comprise cytotoxic NK cells and helper-ILCs, which are further divided in ILC1, ILC2, and ILC3. Helper-ILCs mirror the effector functions of helper T-cell subsets and contribute to host immune defense, tissue homeostasis and repair through cytokine secretion. Although they are mainly tissue-resident, helper ILCs are also found in the peripheral blood (PB). In the human setting, it may be needed to analyze circulating helper ILCs to compare pathological to physiological conditions. In this review, we provide simple guidelines and a list of markers useful to study human PB helper ILCs phenotype and function by flow cytometry.
Collapse
Affiliation(s)
| | - Paola Vacca
- Bambino Gesù Children's Hospital, IRCCS, Rome, Italy
| | | | | |
Collapse
|
12
|
Woodrow JS, Hines M, Sommardahl C, Flatland B, Lo Y, Wang Z, Sheats MK, Lennon EM. Initial investigation of molecular phenotypes of airway mast cells and cytokine profiles in equine asthma. Front Vet Sci 2023; 9:997139. [PMID: 36713876 PMCID: PMC9875299 DOI: 10.3389/fvets.2022.997139] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2022] [Accepted: 12/23/2022] [Indexed: 01/13/2023] Open
Abstract
Equine asthma is a naturally occurring lung disease characterized by chronic, partially reversible airway obstruction, pulmonary remodeling, and lower airway inflammation. Asthma is currently divided into two major groups, mild to moderate asthma (mEA) and severe asthma (sEA), but further subtyping by phenotype (i.e., clinical presentation) and/or endotype (i.e., cellular mechanisms) may be warranted. For this study, we were interested in further investigation of cellular and inflammatory characteristics of EA, including airway mast cells. The purpose of this study was to: (1) compare mast cell protease mRNA expression between healthy and asthmatic horses, (2) analyze the cytokine profile present in BALF of currently defined equine asthma groups, and (3) use these data to evaluate potential biomarkers of defined asthma groups. We hypothesized that there would be significant differences in the cellular mast cell phenotypes (i.e., mucosal vs. connective tissue) and cytokine profiles in the BALF of asthmatic vs. healthy horses and across asthma groups. We assert these characteristics may inform additional subtypes of equine asthma. Adult horses were recruited from the institution's teaching herd and clinical caseload. Mast cell protease gene expression of the BALF cellular component and multiplex bead immunoassay for cytokine concentrations in the BALF supernatant were investigated. Airway mast cells primarily expressed tryptase, with low levels of chymase. No significant changes in protease expression were detected across groups. Horses with severe asthma had increased TNF-α, CXCL-8, and IFN-γ concentrations in BALF supernatant. Multidimensional analysis demonstrated healthy and mEA horses have overlapping characteristics, with sEA separating from the other groups. This difference was primarily due to BALF neutrophil and lymphocyte concentrations. These study results further inform understanding of EA immunopathology, and future studies designed to investigate asthma phenotypes and endotypes. Ultimately, a better understanding of these groups could help identify novel therapeutic strategies.
Collapse
Affiliation(s)
- Jane S. Woodrow
- Department of Comparative and Experimental Medicine, College of Veterinary Medicine, University of Tennessee, Knoxville, Knoxville, TN, United States,Department of Clinical Sciences and Advanced Medicine, College of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA, United States
| | - Melissa Hines
- Department of Large Animal Clinical Sciences, College of Veterinary Medicine, University of Tennessee, Knoxville, Knoxville, TN, United States
| | - Carla Sommardahl
- Department of Large Animal Clinical Sciences, College of Veterinary Medicine, University of Tennessee, Knoxville, Knoxville, TN, United States
| | - Bente Flatland
- Department of Biomedical and Diagnostic Sciences, College of Veterinary Medicine, University of Tennessee, Knoxville, Knoxville, TN, United States
| | - Yancy Lo
- Bioinformatics Core, Institute for Biomedical Informatics, University of Pennsylvania, Philadelphia, PA, United States
| | - Zhiping Wang
- Bioinformatics Core, Institute for Biomedical Informatics, University of Pennsylvania, Philadelphia, PA, United States
| | - Mary Katie Sheats
- Department of Clinical Sciences, Comparative Medicine Institute, College of Veterinary Medicine, North Carolina State University, Raleigh, NC, United States
| | - Elizabeth M. Lennon
- Department of Clinical Sciences and Advanced Medicine, College of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA, United States,*Correspondence: Elizabeth M. Lennon ✉
| |
Collapse
|
13
|
Forte D, Pellegrino RM, Trabanelli S, Tonetti T, Ricci F, Cenerenti M, Comai G, Tazzari P, Lazzarotto T, Buratta S, Urbanelli L, Narimanfar G, Alabed HBR, Mecucci C, La Manna G, Emiliani C, Jandus C, Ranieri VM, Cavo M, Catani L, Palandri F. Circulating extracellular particles from severe COVID-19 patients show altered profiling and innate lymphoid cell-modulating ability. Front Immunol 2023; 14:1085610. [PMID: 37207201 PMCID: PMC10189636 DOI: 10.3389/fimmu.2023.1085610] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2022] [Accepted: 04/11/2023] [Indexed: 05/21/2023] Open
Abstract
Introduction Extracellular vesicles (EVs) and particles (EPs) represent reliable biomarkers for disease detection. Their role in the inflammatory microenvironment of severe COVID-19 patients is not well determined. Here, we characterized the immunophenotype, the lipidomic cargo and the functional activity of circulating EPs from severe COVID-19 patients (Co-19-EPs) and healthy controls (HC-EPs) correlating the data with the clinical parameters including the partial pressure of oxygen to fraction of inspired oxygen ratio (PaO2/FiO2) and the sequential organ failure assessment (SOFA) score. Methods Peripheral blood (PB) was collected from COVID-19 patients (n=10) and HC (n=10). EPs were purified from platelet-poor plasma by size exclusion chromatography (SEC) and ultrafiltration. Plasma cytokines and EPs were characterized by multiplex bead-based assay. Quantitative lipidomic profiling of EPs was performed by liquid chromatography/mass spectrometry combined with quadrupole time-of-flight (LC/MS Q-TOF). Innate lymphoid cells (ILC) were characterized by flow cytometry after co-cultures with HC-EPs or Co-19-EPs. Results We observed that EPs from severe COVID-19 patients: 1) display an altered surface signature as assessed by multiplex protein analysis; 2) are characterized by distinct lipidomic profiling; 3) show correlations between lipidomic profiling and disease aggressiveness scores; 4) fail to dampen type 2 innate lymphoid cells (ILC2) cytokine secretion. As a consequence, ILC2 from severe COVID-19 patients show a more activated phenotype due to the presence of Co-19-EPs. Discussion In summary, these data highlight that abnormal circulating EPs promote ILC2-driven inflammatory signals in severe COVID-19 patients and support further exploration to unravel the role of EPs (and EVs) in COVID-19 pathogenesis.
Collapse
Affiliation(s)
- Dorian Forte
- Department of Medical and Surgical Sciences (DIMEC), Institute of Hematology ‘Seràgnoli’, University of Bologna, Bologna, Italy
| | - Roberto Maria Pellegrino
- Department of Chemistry, Biology and Biotechnology, Biochemistry and Molecular Biology Section, University of Perugia, Perugia, Italy
| | - Sara Trabanelli
- Department of Pathology and Immunology, Faculty of Medicine, University of Geneva, Geneva, Switzerland
- Ludwig Institute for Cancer Research, Lausanne Branch, Lausanne, Switzerland
| | - Tommaso Tonetti
- Department of Medical and Surgical Sciences (DIMEC), University of Bologna, Bologna, Italy
- Anesthesia and Intensive Care Medicine, IRCCS Azienda Ospealiero-Universitaria di Bologna, Bologna, Italy
| | - Francesca Ricci
- Immunohematology and blood bank, IRCCS Azienda Ospedaliero-Universitaria di Bologna, Bologna, Italy
| | - Mara Cenerenti
- Department of Pathology and Immunology, Faculty of Medicine, University of Geneva, Geneva, Switzerland
- Ludwig Institute for Cancer Research, Lausanne Branch, Lausanne, Switzerland
| | - Giorgia Comai
- Nephrology, Dialysis and Renal Transplant Unit, IRCCS Azienda Ospedaliero-Universitaria di Bologna, Bologna, Italy
| | - Pierluigi Tazzari
- Immunohematology and blood bank, IRCCS Azienda Ospedaliero-Universitaria di Bologna, Bologna, Italy
| | - Tiziana Lazzarotto
- Department of Medical and Surgical Sciences (DIMEC), University of Bologna, Bologna, Italy
- Microbiology Unit, IRCCS Azienda Ospedaliero-Universitaria di Bologna, Bologna, Italy
| | - Sandra Buratta
- Department of Chemistry, Biology and Biotechnology, Biochemistry and Molecular Biology Section, University of Perugia, Perugia, Italy
| | - Lorena Urbanelli
- Department of Chemistry, Biology and Biotechnology, Biochemistry and Molecular Biology Section, University of Perugia, Perugia, Italy
| | - Ghazal Narimanfar
- Department of Medical and Surgical Sciences (DIMEC), Institute of Hematology ‘Seràgnoli’, University of Bologna, Bologna, Italy
| | - Husam B. R. Alabed
- Department of Chemistry, Biology and Biotechnology, Biochemistry and Molecular Biology Section, University of Perugia, Perugia, Italy
| | - Cristina Mecucci
- Department of Medicine and Surgery, Center for Hemato-Oncology Research (C.R.E.O.), University of Perugia, Perugia, Italy
| | - Gaetano La Manna
- Nephrology, Dialysis and Renal Transplant Unit, IRCCS Azienda Ospedaliero-Universitaria di Bologna, Bologna, Italy
| | - Carla Emiliani
- Department of Chemistry, Biology and Biotechnology, Biochemistry and Molecular Biology Section, University of Perugia, Perugia, Italy
| | - Camilla Jandus
- Department of Pathology and Immunology, Faculty of Medicine, University of Geneva, Geneva, Switzerland
- Ludwig Institute for Cancer Research, Lausanne Branch, Lausanne, Switzerland
| | - Vito Marco Ranieri
- Department of Medical and Surgical Sciences (DIMEC), University of Bologna, Bologna, Italy
- Anesthesia and Intensive Care Medicine, IRCCS Azienda Ospealiero-Universitaria di Bologna, Bologna, Italy
| | - Michele Cavo
- Department of Medical and Surgical Sciences (DIMEC), Institute of Hematology ‘Seràgnoli’, University of Bologna, Bologna, Italy
- Istituto di Ematologia “Seràgnoli”, IRCCS Azienda Ospedaliero-Universitaria di Bologna, Bologna, Italy
| | - Lucia Catani
- Department of Medical and Surgical Sciences (DIMEC), Institute of Hematology ‘Seràgnoli’, University of Bologna, Bologna, Italy
- Istituto di Ematologia “Seràgnoli”, IRCCS Azienda Ospedaliero-Universitaria di Bologna, Bologna, Italy
- *Correspondence: Lucia Catani,
| | - Francesca Palandri
- Istituto di Ematologia “Seràgnoli”, IRCCS Azienda Ospedaliero-Universitaria di Bologna, Bologna, Italy
| |
Collapse
|
14
|
Roma S, Camisaschi C, Mancuso P, Trabanelli S, Vanazzi A, Villa S, Prati D, Fiori S, Lorenzini D, Tabanelli V, Pileri S, Tarella C, Jandus C, Bertolini F. Dampening of cytotoxic innate lymphoid cells: A new tumour immune escape mechanism in B cell non-Hodgkin's lymphoma. Cell Immunol 2022; 382:104615. [PMID: 36228388 DOI: 10.1016/j.cellimm.2022.104615] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2022] [Revised: 09/05/2022] [Accepted: 09/22/2022] [Indexed: 01/13/2023]
Abstract
The role and regulation of innate immune cells is poorly understood in B-cell non-Hodgkin lymphoma (NHL). As natural killer (NK) cells, helper innate lymphoid cells (ILCs) are lymphocytes endowed with either anti- or pro-tumour activity and involved in inflammatory processes. In our ex vivo analysis of NK cells and ILCs from NHL patients, we observed that, in comparison to healthy donors (HD), the frequency of the cytotoxic subset of NK cells, the CD16+ NK, decreased in patients' peripheral blood. In general, circulating NK cells showed a pro-tumorigenic phenotype, while ILCs displayed a more activated/cytotoxic phenotype. Conversely, at the tumour site, in patients' lymph nodes, ILCs showed a low expression of granzyme.In vitromixed lymphocyte-tumour cell cultures with HD PBMCs and NHL cell lines demonstrated that ILC cytotoxic potential was lowered by the presence of tumour cells but, in the absence of T regulatory cells (Tregs), their cytolytic potential was recovered. Our data shed novel light on dysfunctional innate immunity in NHL. We suggest a new mechanism of tumour immuno-escape based on the reduction of cell cytotoxicity involving ILCs and likely controlled by Tregs.
Collapse
Affiliation(s)
- Stefania Roma
- Laboratory of Hematology- Oncology, IRCCS European Institute of Oncology, Milan, Italy
| | - Chiara Camisaschi
- Laboratory of Hematology- Oncology, IRCCS European Institute of Oncology, Milan, Italy
| | - Patrizia Mancuso
- Laboratory of Hematology- Oncology, IRCCS European Institute of Oncology, Milan, Italy
| | - Sara Trabanelli
- Dept. of Pathology and Immunology, Faculty of Medicine, University of Geneva, Switzerland; Ludwig Institute for Cancer Research, Lausanne Branch, Lausanne, Switzerland
| | - Anna Vanazzi
- Division of Clinical Hematology- Oncology, IRCCS European Institute of Oncology, Milan, Italy
| | - Stefania Villa
- Department of Transfusion Medicine and Hematology Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy
| | - Daniele Prati
- Department of Transfusion Medicine and Hematology Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy
| | - Stefano Fiori
- Division of Haematopathology, IRCCS European Institute of Oncology, Milan, Italy
| | - Daniele Lorenzini
- Division of Haematopathology, IRCCS European Institute of Oncology, Milan, Italy
| | - Valentina Tabanelli
- Division of Haematopathology, IRCCS European Institute of Oncology, Milan, Italy
| | - Stefano Pileri
- Division of Haematopathology, IRCCS European Institute of Oncology, Milan, Italy
| | - Corrado Tarella
- Division of Clinical Hematology- Oncology, IRCCS European Institute of Oncology, Milan, Italy
| | - Camilla Jandus
- Dept. of Pathology and Immunology, Faculty of Medicine, University of Geneva, Switzerland; Ludwig Institute for Cancer Research, Lausanne Branch, Lausanne, Switzerland
| | - Francesco Bertolini
- Laboratory of Hematology- Oncology, IRCCS European Institute of Oncology, Milan, Italy; Onco-Tech Lab, European Institute of Oncology IRCCS and Politecnico di Milano, Milan, Italy.
| |
Collapse
|
15
|
Jowett GM, Read E, Roberts LB, Coman D, Vilà González M, Zabinski T, Niazi U, Reis R, Trieu TJ, Danovi D, Gentleman E, Vallier L, Curtis MA, Lord GM, Neves JF. Organoids capture tissue-specific innate lymphoid cell development in mice and humans. Cell Rep 2022; 40:111281. [PMID: 36044863 PMCID: PMC9638027 DOI: 10.1016/j.celrep.2022.111281] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2021] [Revised: 01/06/2022] [Accepted: 08/05/2022] [Indexed: 12/21/2022] Open
Abstract
Organoid-based models of murine and human innate lymphoid cell precursor (ILCP) maturation are presented. First, murine intestinal and pulmonary organoids are harnessed to demonstrate that the epithelial niche is sufficient to drive tissue-specific maturation of all innate lymphoid cell (ILC) groups in parallel, without requiring subset-specific cytokine supplementation. Then, more complex human induced pluripotent stem cell (hiPSC)-based gut and lung organoid models are used to demonstrate that human epithelial cells recapitulate maturation of ILC from a stringent systemic human ILCP population, but only when the organoid-associated stromal cells are depleted. These systems offer versatile and reductionist models to dissect the impact of environmental and mucosal niche cues on ILC maturation. In the future, these could provide insight into how ILC activity and development might become dysregulated in chronic inflammatory diseases.
Collapse
Affiliation(s)
- Geraldine M Jowett
- School for Immunology and Microbial Sciences, King's College London, London SE1 9RT, UK; Centre for Host Microbiome Interactions, King's College London, London SE1 9RT, UK; Centre for Craniofacial and Regenerative Biology, King's College London, London SE1 9RT, UK; Centre for Gene Therapy & Regenerative Medicine, King's College London, London SE1 9RT, UK; Wellcome Trust Cell Therapies and Regenerative Medicine Ph.D. Programme, London SE1 9RT, UK
| | - Emily Read
- School for Immunology and Microbial Sciences, King's College London, London SE1 9RT, UK; Centre for Host Microbiome Interactions, King's College London, London SE1 9RT, UK; Centre for Gene Therapy & Regenerative Medicine, King's College London, London SE1 9RT, UK
| | - Luke B Roberts
- School for Immunology and Microbial Sciences, King's College London, London SE1 9RT, UK
| | - Diana Coman
- Centre for Host Microbiome Interactions, King's College London, London SE1 9RT, UK
| | - Marta Vilà González
- Wellcome and MRC Cambridge Stem Cell Institute, Puddicombe Way, Cambridge CB2 0AW, UK; Department of Surgery, University of Cambridge and NIHR Cambridge Biomedical Research Centre, Hills Road, Cambridge CB2 0QQ, UK
| | - Tomasz Zabinski
- School for Immunology and Microbial Sciences, King's College London, London SE1 9RT, UK
| | - Umar Niazi
- Guy's and St. Thomas' National Health Service Foundation Trust and King's College London National Institute for Health and Care Research Biomedical Research Centre Translational Bioinformatics Platform, Guy's Hospital, London SE1 9RT, UK
| | - Rita Reis
- School for Immunology and Microbial Sciences, King's College London, London SE1 9RT, UK
| | - Tung-Jui Trieu
- Centre for Host Microbiome Interactions, King's College London, London SE1 9RT, UK; Centre for Craniofacial and Regenerative Biology, King's College London, London SE1 9RT, UK
| | - Davide Danovi
- Centre for Craniofacial and Regenerative Biology, King's College London, London SE1 9RT, UK; bit.bio, Babraham Research Campus, The Dorothy Hodgkin Building, Cambridge CB22 3FH, UK
| | - Eileen Gentleman
- Centre for Craniofacial and Regenerative Biology, King's College London, London SE1 9RT, UK
| | - Ludovic Vallier
- Wellcome and MRC Cambridge Stem Cell Institute, Puddicombe Way, Cambridge CB2 0AW, UK; Department of Surgery, University of Cambridge and NIHR Cambridge Biomedical Research Centre, Hills Road, Cambridge CB2 0QQ, UK
| | - Michael A Curtis
- Centre for Host Microbiome Interactions, King's College London, London SE1 9RT, UK
| | - Graham M Lord
- School for Immunology and Microbial Sciences, King's College London, London SE1 9RT, UK; Faculty of Biology, Medicine and Health, University of Manchester, Oxford Road, Manchester M13 9PL, UK
| | - Joana F Neves
- School for Immunology and Microbial Sciences, King's College London, London SE1 9RT, UK; Centre for Host Microbiome Interactions, King's College London, London SE1 9RT, UK.
| |
Collapse
|
16
|
Calvi M, Di Vito C, Frigo A, Trabanelli S, Jandus C, Mavilio D. Development of Human ILCs and Impact of Unconventional Cytotoxic Subsets in the Pathophysiology of Inflammatory Diseases and Cancer. Front Immunol 2022; 13:914266. [PMID: 35720280 PMCID: PMC9204637 DOI: 10.3389/fimmu.2022.914266] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2022] [Accepted: 04/28/2022] [Indexed: 11/13/2022] Open
Abstract
Innate lymphoid cells (ILCs) were firstly described by different independent laboratories in 2008 as tissue-resident innate lymphocytes mirroring the phenotype and function of T helper cells. ILCs have been subdivided into three distinct subgroups, ILC1, ILC2 and ILC3, according to their cytokine and transcriptional profiles. Subsequently, also Natural Killer (NK) cells, that are considered the innate counterpart of cytotoxic CD8 T cells, were attributed to ILC1 subfamily, while lymphoid tissue inducer (LTi) cells were attributed to ILC3 subgroup. Starting from their discovery, significant advances have been made in our understanding of ILC impact in the maintenance of tissue homeostasis, in the protection against pathogens and in tumor immune-surveillance. However, there is still much to learn about ILC ontogenesis especially in humans. In this regard, NK cell developmental intermediates which have been well studied and characterized prior to the discovery of helper ILCs, have been used to shape a model of ILC ontogenesis. Herein, we will provide an overview of the current knowledge about NK cells and helper ILC ontogenesis in humans. We will also focus on the newly disclosed circulating ILC subsets with killing properties, namely unconventional CD56dim NK cells and cytotoxic helper ILCs, by discussing their possible role in ILC ontogenesis and their contribution in both physiological and pathological conditions.
Collapse
Affiliation(s)
- Michela Calvi
- Department of Medical Biotechnologies and Translational Medicine (BioMeTra), University of Milan, Milan, Italy
| | - Clara Di Vito
- Unit of Clinical and Experimental Immunology, IRCCS Humanitas Research Hospital, Milan, Italy
| | - Alessandro Frigo
- Department of Medical Biotechnologies and Translational Medicine (BioMeTra), University of Milan, Milan, Italy
| | - Sara Trabanelli
- Department of Pathology and Immunology, Faculty of Medicine, University of Geneva, Geneva, Switzerland.,Ludwig Institute for Cancer Research, Lausanne, Switzerland
| | - Camilla Jandus
- Department of Pathology and Immunology, Faculty of Medicine, University of Geneva, Geneva, Switzerland.,Ludwig Institute for Cancer Research, Lausanne, Switzerland
| | - Domenico Mavilio
- Department of Medical Biotechnologies and Translational Medicine (BioMeTra), University of Milan, Milan, Italy.,Unit of Clinical and Experimental Immunology, IRCCS Humanitas Research Hospital, Milan, Italy
| |
Collapse
|
17
|
Heinrich B, Korangy F. Plasticity of Innate Lymphoid Cells in Cancer. Front Immunol 2022; 13:886520. [PMID: 35663967 PMCID: PMC9160464 DOI: 10.3389/fimmu-13-886520] [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: 02/28/2022] [Accepted: 04/20/2022] [Indexed: 11/23/2022] Open
Abstract
Innate lymphoid cells (ILCs) are a heterogenous population of the innate immune system, enriched at mucosal surfaces and are pivotal regulators of immune homeostasis. ILCs are the innate counterpart of T cells. Like T cells, ILC subsets are highly plastic with their composition and function controlled by alterations in their microenvironment. This plasticity allows for the trans-differentiation between the subsets to rapidly respond to their immune environment. The tumor microenvironment (TME) is a heterogeneous milieu characterized by different cytokines and growth factors. Through interaction with the tumor microenvironment, ILCs can transdifferentiate into different subsets resulting in pro or anti-tumor immunity. Thus, studying ILC plasticity might result in new therapeutic approaches for cancer therapy. In this review, we summarize current findings of the functional and plastic heterogeneity of ILCs in homeostasis as well as disease settings with a specific focus on cancer. We specifically highlight tumor-driven plasticity and how ILC-induced inflammation can impact the tumor microenvironment and anti-tumor immunity.
Collapse
Affiliation(s)
- Bernd Heinrich
- Thoracic and Gastrointestinal Malignancies Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, United States
- Department of Gastroenterology, Hepatology and Endocrinology, Hannover Medical School, Hannover, Germany
| | - Firouzeh Korangy
- Thoracic and Gastrointestinal Malignancies Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, United States
- *Correspondence: Firouzeh Korangy,
| |
Collapse
|
18
|
Trabanelli S, Ercolano G, Wyss T, Gomez-Cadena A, Falquet M, Cropp D, Imbratta C, Leblond MM, Salvestrini V, Curti A, Adotevi O, Jandus C, Verdeil G. c-Maf enforces cytokine production and promotes memory-like responses in mouse and human type 2 innate lymphoid cells. EMBO J 2022; 41:e109300. [PMID: 35467036 PMCID: PMC9194744 DOI: 10.15252/embj.2021109300] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2021] [Revised: 03/31/2022] [Accepted: 04/01/2022] [Indexed: 12/13/2022] Open
Abstract
Group‐2 innate lymphoid cells (ILC2s), which are involved in type 2 inflammatory diseases such as allergy, can exhibit immunological memory, but the basis of this ILC2 "trained immunity" has remained unclear. Here, we found that stimulation with IL‐33/IL‐25 or exposure to the allergen papain induces the expression of the transcription factor c‐Maf in mouse ILC2s. Chronic papain exposure results in high production of IL‐5 and IL‐13 cytokines and lung eosinophil recruitment, effects that are blocked by c‐Maf deletion in ILCs. Transcriptomic analysis revealed that knockdown of c‐Maf in ILC2s suppresses expression of type 2 cytokine genes, as well as of genes linked to a memory‐like phenotype. Consistently, c‐Maf was found highly expressed in human adult ILC2s but absent in cord blood and required for cytokine production in isolated human ILC2s. Furthermore, c‐Maf‐deficient mouse or human ILC2s failed to exhibit strengthened (“trained”) responses upon repeated challenge. Thus, the expression of c‐Maf is indispensable for optimal type 2 cytokine production and proper memory‐like responses in group‐2 innate lymphoid cells.
Collapse
Affiliation(s)
- Sara Trabanelli
- Departement of Oncology, UNIL-CHUV, Ludwig Institute for Cancer Research Lausanne, University of Lausanne, Lausanne, Switzerland
| | - Giuseppe Ercolano
- Departement of Oncology, UNIL-CHUV, Ludwig Institute for Cancer Research Lausanne, University of Lausanne, Lausanne, Switzerland
| | - Tania Wyss
- Departement of Oncology, UNIL-CHUV, Ludwig Institute for Cancer Research Lausanne, University of Lausanne, Lausanne, Switzerland
| | - Alejandra Gomez-Cadena
- Departement of Oncology, UNIL-CHUV, Ludwig Institute for Cancer Research Lausanne, University of Lausanne, Lausanne, Switzerland
| | - Maryline Falquet
- Departement of Oncology, UNIL-CHUV, Ludwig Institute for Cancer Research Lausanne, University of Lausanne, Lausanne, Switzerland
| | - Daniela Cropp
- Departement of Oncology, UNIL-CHUV, University of Lausanne, Lausanne, Switzerland
| | - Claire Imbratta
- Departement of Oncology, UNIL-CHUV, University of Lausanne, Lausanne, Switzerland
| | - Marine M Leblond
- Departement of Oncology, UNIL-CHUV, University of Lausanne, Lausanne, Switzerland
| | - Valentina Salvestrini
- IRCCS Azienda Ospedaliero-Universitaria di Bologna, Istituto di Ematologia "Seràgnoli", Bologna, Italy
| | - Antonio Curti
- IRCCS Azienda Ospedaliero-Universitaria di Bologna, Istituto di Ematologia "Seràgnoli", Bologna, Italy
| | - Olivier Adotevi
- INSERM, UMR1098 RIGHT, EFS-BFC, University of Bourgogne Franche-Comté, Besançon, France
| | - Camilla Jandus
- Departement of Oncology, UNIL-CHUV, Ludwig Institute for Cancer Research Lausanne, University of Lausanne, Lausanne, Switzerland
| | - Grégory Verdeil
- Departement of Oncology, UNIL-CHUV, University of Lausanne, Lausanne, Switzerland
| |
Collapse
|
19
|
Abstract
More than a decade ago, type 2 innate lymphoid cells (ILC2s) were discovered to be members of a family of innate immune cells consisting of five subsets that form a first line of defence against infections before the recruitment of adaptive immune cells. Initially, ILC2s were implicated in the early immune response to parasitic infections, but it is now clear that ILC2s are highly diverse and have crucial roles in the regulation of tissue homeostasis and repair. ILC2s can also regulate the functions of other type 2 immune cells, including T helper 2 cells, type 2 macrophages and eosinophils. Dysregulation of ILC2s contributes to type 2-mediated pathology in a wide variety of diseases, potentially making ILC2s attractive targets for therapeutic interventions. In this Review, we focus on the spectrum of ILC2 phenotypes that have been described across different tissues and disease states with an emphasis on human ILC2s. We discuss recent insights in ILC2 biology and suggest how this knowledge might be used for novel disease treatments and improved human health. Type 2 innate lymphoid cells (ILC2s) have diverse phenotypes across different tissues and disease states. Recent insights into ILC2 biology raise new possibilities for the improved treatment of cancer and of metabolic, infectious and chronic inflammatory diseases.
Collapse
Affiliation(s)
- Hergen Spits
- Department of Experimental Immunology, Amsterdam UMC, Location AMC, University of Amsterdam, Amsterdam, Netherlands.
| | - Jenny Mjösberg
- Center for Infectious Medicine, Department of Medicine Huddinge, Karolinska Institutet, Karolinska University Hospital Huddinge, Stockholm, Sweden.
| |
Collapse
|
20
|
Murphy JM, Ngai L, Mortha A, Crome SQ. Tissue-Dependent Adaptations and Functions of Innate Lymphoid Cells. Front Immunol 2022; 13:836999. [PMID: 35359972 PMCID: PMC8960279 DOI: 10.3389/fimmu.2022.836999] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2021] [Accepted: 02/11/2022] [Indexed: 12/21/2022] Open
Abstract
Tissue-resident immune cells reside in distinct niches across organs, where they contribute to tissue homeostasis and rapidly respond to perturbations in the local microenvironment. Innate lymphoid cells (ILCs) are a family of innate immune cells that regulate immune and tissue homeostasis. Across anatomical locations throughout the body, ILCs adopt tissue-specific fates, differing from circulating ILC populations. Adaptations of ILCs to microenvironmental changes have been documented in several inflammatory contexts, including obesity, asthma, and inflammatory bowel disease. While our understanding of ILC functions within tissues have predominantly been based on mouse studies, development of advanced single cell platforms to study tissue-resident ILCs in humans and emerging patient-based data is providing new insights into this lymphocyte family. Within this review, we discuss current concepts of ILC fate and function, exploring tissue-specific functions of ILCs and their contribution to health and disease across organ systems.
Collapse
Affiliation(s)
- Julia M. Murphy
- Department of Immunology, University of Toronto, Toronto, ON, Canada
- Toronto General Hospital Research Institute, University Health Network, Toronto, ON, Canada
- Ajmera Transplant Centre, University Health Network, Toronto, ON, Canada
| | - Louis Ngai
- Department of Immunology, University of Toronto, Toronto, ON, Canada
| | - Arthur Mortha
- Department of Immunology, University of Toronto, Toronto, ON, Canada
| | - Sarah Q. Crome
- Department of Immunology, University of Toronto, Toronto, ON, Canada
- Toronto General Hospital Research Institute, University Health Network, Toronto, ON, Canada
- Ajmera Transplant Centre, University Health Network, Toronto, ON, Canada
| |
Collapse
|
21
|
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: 4.7] [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
|
22
|
Grimaldi A, Pietropaolo G, Stabile H, Kosta A, Capuano C, Gismondi A, Santoni A, Sciumè G, Fionda C. The Regulatory Activity of Noncoding RNAs in ILCs. Cells 2021; 10:cells10102742. [PMID: 34685721 PMCID: PMC8534545 DOI: 10.3390/cells10102742] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2021] [Revised: 10/01/2021] [Accepted: 10/11/2021] [Indexed: 12/12/2022] Open
Abstract
Innate lymphoid cells (ILCs) are innate lymphocytes playing essential functions in protection against microbial infections and participate in both homeostatic and pathological contexts, including tissue remodeling, cancer, and inflammatory disorders. A number of lineage-defining transcription factors concurs to establish transcriptional networks which determine the identity and the activity of the distinct ILC subsets. However, the contribution of other regulatory molecules in controlling ILC development and function is also recently emerging. In this regard, noncoding RNAs (ncRNAs) represent key elements of the complex regulatory network of ILC biology and host protection. ncRNAs mostly lack protein-coding potential, but they are endowed with a relevant regulatory activity in immune and nonimmune cells because of their ability to control chromatin structure, RNA stability, and/or protein synthesis. Herein, we summarize recent studies describing how distinct types of ncRNAs, mainly microRNAs, long ncRNAs, and circular RNAs, act in the context of ILC biology. In particular, we comment on how ncRNAs can exert key effects in ILCs by controlling gene expression in a cell- or state-specific manner and how this tunes distinct functional outputs in ILCs.
Collapse
Affiliation(s)
- Alessio Grimaldi
- Department of Molecular Medicine, Istituto Pasteur-Fondazione Cenci Bolognetti, Sapienza University of Rome, 00161 Rome, Italy; (A.G.); (G.P.); (H.S.); (A.K.); (A.G.); (A.S.); (G.S.)
| | - Giuseppe Pietropaolo
- Department of Molecular Medicine, Istituto Pasteur-Fondazione Cenci Bolognetti, Sapienza University of Rome, 00161 Rome, Italy; (A.G.); (G.P.); (H.S.); (A.K.); (A.G.); (A.S.); (G.S.)
| | - Helena Stabile
- Department of Molecular Medicine, Istituto Pasteur-Fondazione Cenci Bolognetti, Sapienza University of Rome, 00161 Rome, Italy; (A.G.); (G.P.); (H.S.); (A.K.); (A.G.); (A.S.); (G.S.)
| | - Andrea Kosta
- Department of Molecular Medicine, Istituto Pasteur-Fondazione Cenci Bolognetti, Sapienza University of Rome, 00161 Rome, Italy; (A.G.); (G.P.); (H.S.); (A.K.); (A.G.); (A.S.); (G.S.)
| | - Cristina Capuano
- Department of Experimental Medicine, Sapienza University of Rome, 00161 Rome, Italy;
| | - Angela Gismondi
- Department of Molecular Medicine, Istituto Pasteur-Fondazione Cenci Bolognetti, Sapienza University of Rome, 00161 Rome, Italy; (A.G.); (G.P.); (H.S.); (A.K.); (A.G.); (A.S.); (G.S.)
| | - Angela Santoni
- Department of Molecular Medicine, Istituto Pasteur-Fondazione Cenci Bolognetti, Sapienza University of Rome, 00161 Rome, Italy; (A.G.); (G.P.); (H.S.); (A.K.); (A.G.); (A.S.); (G.S.)
- IRCCS (Istituto di Ricovero e Cura a Carattere Scientifico) Neuromed, 86077 Pozzilli, Italy
| | - Giuseppe Sciumè
- Department of Molecular Medicine, Istituto Pasteur-Fondazione Cenci Bolognetti, Sapienza University of Rome, 00161 Rome, Italy; (A.G.); (G.P.); (H.S.); (A.K.); (A.G.); (A.S.); (G.S.)
| | - Cinzia Fionda
- Department of Molecular Medicine, Istituto Pasteur-Fondazione Cenci Bolognetti, Sapienza University of Rome, 00161 Rome, Italy; (A.G.); (G.P.); (H.S.); (A.K.); (A.G.); (A.S.); (G.S.)
- Correspondence: ; Tel.: +39-0649255118; Fax: +39-0644340632
| |
Collapse
|
23
|
An in vitro platform supports generation of human innate lymphoid cells from CD34 + hematopoietic progenitors that recapitulate ex vivo identity. Immunity 2021; 54:2417-2432.e5. [PMID: 34453879 DOI: 10.1016/j.immuni.2021.07.019] [Citation(s) in RCA: 35] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2019] [Revised: 04/12/2021] [Accepted: 07/29/2021] [Indexed: 12/19/2022]
Abstract
Innate lymphoid cells (ILCs) are critical effectors of innate immunity and inflammation, whose development and activation pathways make for attractive therapeutic targets. However, human ILC generation has not been systematically explored, and previous in vitro investigations relied on the analysis of few markers or cytokines, which are suboptimal to assign lineage identity. Here, we developed a platform that reliably generated human ILC lineages from CD34+ hematopoietic progenitors derived from cord blood and bone marrow. We showed that one culture condition is insufficient to generate all ILC subsets, and instead, distinct combination of cytokines and Notch signaling are essential. The identity of natural killer (NK)/ILC1s, ILC2s, and ILC3s generated in vitro was validated by protein expression, functional assays, and both global and single-cell transcriptome analysis, recapitulating the signatures and functions of their ex vivo ILC counterparts. These data represent a resource to aid in clarifying ILC biology and differentiation.
Collapse
|
24
|
Pan L, Chen X, Liu X, Qiu W, Liu Y, Jiang W, Zheng Y, Mou Y, Xu W, Li X, Ge H, Zhu H. Innate lymphoid cells exhibited IL-17-expressing phenotype in active tuberculosis disease. BMC Pulm Med 2021; 21:318. [PMID: 34641843 PMCID: PMC8513179 DOI: 10.1186/s12890-021-01678-1] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2021] [Accepted: 09/21/2021] [Indexed: 12/22/2022] Open
Abstract
BACKGROUND Innate lymphoid cells (ILCs), as an important group of innate immunity, could respond rapidly to Mycobacterium tuberculosis (Mtb) infection. In this research, we studied the phenotypic changes of circulatory ILCs in active tuberculosis (TB) disease. METHODS We recruited 40 patients with active Mtb infection (TB group) and 41 healthy subjects (NC group), and collected their clinical information and peripheral blood. Circulating ILCs, ILC subsets, dendritic cells (DCs), macrophages, and the production of cytokines in ILCs were tested by flow cytometry (FCM). Enzyme-linked immunosorbent assay (ELISA) was used to detect plasma IL-23. RESULTS Compared with healthy control, total ILCs (0.73% vs. 0.42%, P = 0.0019), ILC1 (0.55% vs. 0.31%, P = 0.0024) and CD117+ ILC2 (0.02% vs. 0.01%, P = 0.0267) were upregulated in TB group. The total IL-17+ lymphocytes were elevated (3.83% vs. 1.76%, P = 0.0006) while the IL-22+ lymphocytes remained unchanged. Within ILC subsets, ILC3, CD117+ ILC2 and ILC1 in TB group all expressed increased IL-17 (15.15% vs. 4.55%, 19.01% vs. 4.57%, 8.79% vs. 3.87%, P < 0.0001) but similar IL-22 comparing with healthy control. TB group had more plasma IL-23 than NC group (7.551 vs. 5.564 pg/mL, P = 0.0557). Plasma IL-23 in TB group was positively correlated to IL-17+ ILC3 (r = 0.4435, P = 0.0141), IL-17+CD117+ ILC2 (r = 0.5385, P = 0.0021) and IL-17+ ILC1(r = 0.3719, P = 0.0430). TB group also had elevated DCs (9.35% vs. 6.49%, P < 0.0001) while macrophages remained unchanged. Within TB group, higher proportion of IL-17+ ILCs was related to severer inflammatory status and poorer clinical condition. CONCLUSIONS In active TB disease, circulatory ILCs were upregulated and exhibited IL-17-expressing phenotype. This may expand the understanding of immune reaction to Mtb infection.
Collapse
Affiliation(s)
- Linyue Pan
- Department of Respiratory and Critical Care Medicine, The Affiliated Huadong Hospital of Fudan University, 221 West Yan'an Road, Shanghai, 200040, China.,Department of Respiratory and Critical Care Medicine, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Xiaoli Chen
- Department of Tuberculosis, the Sixth People's Hospital of Nantong, Jiangsu, China
| | - Xuanqi Liu
- Department of Respiratory and Critical Care Medicine, The Affiliated Huadong Hospital of Fudan University, 221 West Yan'an Road, Shanghai, 200040, China
| | - Wenjia Qiu
- Department of Respiratory and Critical Care Medicine, The Affiliated Huadong Hospital of Fudan University, 221 West Yan'an Road, Shanghai, 200040, China
| | - Yunhuan Liu
- Department of Respiratory and Critical Care Medicine, The Affiliated Huadong Hospital of Fudan University, 221 West Yan'an Road, Shanghai, 200040, China
| | - Weiping Jiang
- Department of Respiratory and Critical Care Medicine, The Affiliated Huadong Hospital of Fudan University, 221 West Yan'an Road, Shanghai, 200040, China
| | - Yang Zheng
- Department of Respiratory and Critical Care Medicine, The Affiliated Huadong Hospital of Fudan University, 221 West Yan'an Road, Shanghai, 200040, China
| | - Yan Mou
- Department of Respiratory and Critical Care Medicine, The Affiliated Huadong Hospital of Fudan University, 221 West Yan'an Road, Shanghai, 200040, China
| | - Wei Xu
- Department of Geriatrics, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Xiangyang Li
- Department of Respiratory and Critical Care Medicine, The Affiliated Huadong Hospital of Fudan University, 221 West Yan'an Road, Shanghai, 200040, China
| | - Haiyan Ge
- Department of Respiratory and Critical Care Medicine, The Affiliated Huadong Hospital of Fudan University, 221 West Yan'an Road, Shanghai, 200040, China.
| | - Huili Zhu
- Department of Respiratory and Critical Care Medicine, The Affiliated Huadong Hospital of Fudan University, 221 West Yan'an Road, Shanghai, 200040, China
| |
Collapse
|
25
|
Roma S, Carpen L, Raveane A, Bertolini F. The Dual Role of Innate Lymphoid and Natural Killer Cells in Cancer. from Phenotype to Single-Cell Transcriptomics, Functions and Clinical Uses. Cancers (Basel) 2021; 13:cancers13205042. [PMID: 34680190 PMCID: PMC8533946 DOI: 10.3390/cancers13205042] [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: 08/30/2021] [Revised: 10/05/2021] [Accepted: 10/06/2021] [Indexed: 02/07/2023] Open
Abstract
Simple Summary Innate lymphoid cells (ILCs), a family of innate immune cells including natural killers (NKs), play a multitude of roles in first-line cancer control, in escape from immunity and in cancer progression. In this review, we summarize preclinical and clinical data on ILCs and NK cells concerning their phenotype, function and clinical applications in cellular therapy trials. We also describe how single-cell transcriptome sequencing has been used and forecast how it will be used to better understand ILC and NK involvement in cancer control and progression as well as their therapeutic potential. Abstract The role of innate lymphoid cells (ILCs), including natural killer (NK) cells, is pivotal in inflammatory modulation and cancer. Natural killer cell activity and count have been demonstrated to be regulated by the expression of activating and inhibitory receptors together with and as a consequence of different stimuli. The great majority of NK cell populations have an anti-tumor activity due to their cytotoxicity, and for this reason have been used for cellular therapies in cancer patients. On the other hand, the recently classified helper ILCs are fundamentally involved in inflammation and they can be either helpful or harmful in cancer development and progression. Tissue niche seems to play an important role in modulating ILC function and conversion, as observed at the transcriptional level. In the past, these cell populations have been classified by the presence of specific cellular receptor markers; more recently, due to the advent of single-cell RNA sequencing (scRNA-seq), it has been possible to also explore them at the transcriptomic level. In this article we review studies on ILC (and NK cell) classification, function and their involvement in cancer. We also summarize the potential application of NK cells in cancer therapy and give an overview of the most recent studies involving ILCs and NKs at scRNA-seq, focusing on cancer. Finally, we provide a resource for those who wish to start single-cell transcriptomic analysis on the context of these innate lymphoid cell populations.
Collapse
|
26
|
Liu C, Gong Y, Zhang H, Yang H, Zeng Y, Bian Z, Xin Q, Bai Z, Zhang M, He J, Yan J, Zhou J, Li Z, Ni Y, Wen A, Lan Y, Hu H, Liu B. Delineating spatiotemporal and hierarchical development of human fetal innate lymphoid cells. Cell Res 2021; 31:1106-1122. [PMID: 34239074 PMCID: PMC8486758 DOI: 10.1038/s41422-021-00529-2] [Citation(s) in RCA: 31] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2021] [Accepted: 06/08/2021] [Indexed: 02/07/2023] Open
Abstract
Whereas the critical roles of innate lymphoid cells (ILCs) in adult are increasingly appreciated, their developmental hierarchy in early human fetus remains largely elusive. In this study, we sorted human hematopoietic stem/progenitor cells, lymphoid progenitors, putative ILC progenitor/precursors and mature ILCs in the fetal hematopoietic, lymphoid and non-lymphoid tissues, from 8 to 12 post-conception weeks, for single-cell RNA-sequencing, followed by computational analysis and functional validation at bulk and single-cell levels. We delineated the early phase of ILC lineage commitment from hematopoietic stem/progenitor cells, which mainly occurred in fetal liver and intestine. We further unveiled interleukin-3 receptor as a surface marker for the lymphoid progenitors in fetal liver with T, B, ILC and myeloid potentials, while IL-3RA- lymphoid progenitors were predominantly B-lineage committed. Notably, we determined the heterogeneity and tissue distribution of each ILC subpopulation, revealing the proliferating characteristics shared by the precursors of each ILC subtype. Additionally, a novel unconventional ILC2 subpopulation (CRTH2- CCR9+ ILC2) was identified in fetal thymus. Taken together, our study illuminates the precise cellular and molecular features underlying the stepwise formation of human fetal ILC hierarchy with remarkable spatiotemporal heterogeneity.
Collapse
Affiliation(s)
- Chen Liu
- State Key Laboratory of Proteomics, Academy of Military Medical Sciences, Academy of Military Sciences, Beijing, China
| | - Yandong Gong
- State Key Laboratory of Experimental Hematology, Institute of Hematology, Fifth Medical Center of Chinese PLA General Hospital, Beijing, China
| | - Han Zhang
- Department of Blood Transfusion, Daping Hospital, Army Military Medical University, Chongqing, China
| | - Hua Yang
- Tianjin Central Hospital of Gynecology Obstetrics, Tianjin, China
| | - Yang Zeng
- State Key Laboratory of Experimental Hematology, Institute of Hematology, Fifth Medical Center of Chinese PLA General Hospital, Beijing, China
| | - Zhilei Bian
- Key Laboratory for Regenerative Medicine of Ministry of Education, Institute of Hematology, School of Medicine, Jinan University, Guangzhou, China
| | - Qian Xin
- State Key Laboratory of Proteomics, Academy of Military Medical Sciences, Academy of Military Sciences, Beijing, China
| | - Zhijie Bai
- State Key Laboratory of Proteomics, Academy of Military Medical Sciences, Academy of Military Sciences, Beijing, China
| | - Man Zhang
- State Key Laboratory of Proteomics, Academy of Military Medical Sciences, Academy of Military Sciences, Beijing, China
| | - Jian He
- State Key Laboratory of Proteomics, Academy of Military Medical Sciences, Academy of Military Sciences, Beijing, China
| | - Jing Yan
- State Key Laboratory of Proteomics, Academy of Military Medical Sciences, Academy of Military Sciences, Beijing, China
| | - Jie Zhou
- State Key Laboratory of Experimental Hematology, Institute of Hematology, Fifth Medical Center of Chinese PLA General Hospital, Beijing, China
| | - Zongcheng Li
- State Key Laboratory of Experimental Hematology, Institute of Hematology, Fifth Medical Center of Chinese PLA General Hospital, Beijing, China
| | - Yanli Ni
- State Key Laboratory of Experimental Hematology, Institute of Hematology, Fifth Medical Center of Chinese PLA General Hospital, Beijing, China
| | - Aiqing Wen
- Department of Blood Transfusion, Daping Hospital, Army Military Medical University, Chongqing, China.
| | - Yu Lan
- Key Laboratory for Regenerative Medicine of Ministry of Education, Institute of Hematology, School of Medicine, Jinan University, Guangzhou, China.
| | - Hongbo Hu
- Center for Immunology and Hematology, the State Key Laboratory of Biotherapy, West China Hospital, Sichuan University. Collaboration and Innovation Center for Biotherapy, Chengdu, China.
| | - Bing Liu
- State Key Laboratory of Proteomics, Academy of Military Medical Sciences, Academy of Military Sciences, Beijing, China.
- State Key Laboratory of Experimental Hematology, Institute of Hematology, Fifth Medical Center of Chinese PLA General Hospital, Beijing, China.
- Key Laboratory for Regenerative Medicine of Ministry of Education, Institute of Hematology, School of Medicine, Jinan University, Guangzhou, China.
| |
Collapse
|
27
|
Long X, Wang N, Zhang X. Roles of Clara cell 10-kD protein and type 2 innate lymphoid cells in allergic rhinitis. Cell Cycle 2021; 20:1923-1934. [PMID: 34437822 DOI: 10.1080/15384101.2021.1966961] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022] Open
Abstract
This study examined the potential roles of CC10 (Clara cell 10-kD protein) and ILC2s (type 2 innate lymphoid cells) in allergic rhinitis (AR). After ovalbumin was used to construct the AR model, microarray analysis was performed to reveal the key differentially expressed genes. The phenotypic changes of nasal mucosa were examined by H&E staining. Western blot analysis, qRT-PCR, ELISA and immunohistochemistry were performed to identify the levels of cytokines. The lineage markers (CD127 and CD117) of ILC2s were detected using immunofluorescence. The microarray analysis and qRT-PCR results showed that CC10 overexpression inhibited the expression of A20, BAFF, and IL-4 R in vivo. Also, CC10 overexpression was found to ameliorate the damage of nasal mucosa in AR mice. Investigations revealed that the ILC2s were activated in AR mice and AR patients with high levels of IgE, IgG1, IL-4, IL-5, IL-13, IL-25, and IL-33. Moreover, CD127+ was found to activate ILC2s. However, CC10 overexpression suppressed the activation of ILC2s. In conclusion, this research suggested that CC10 could suppress the activation of ILC2s to attenuate the damage of nasal mucosa and that CD127+ may be a biomarker of the activation of ILC2s in AR mice and AR patients.
Collapse
Affiliation(s)
- Xiaobo Long
- Department of Otolaryngology-Head and Neck Surgery, Tongji Hospital Affiliated to Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Nan Wang
- Department of Otolaryngology-Head and Neck Surgery, Tongji Hospital Affiliated to Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Xinhao Zhang
- Department of Otolaryngology-Head and Neck Surgery, Tongji Hospital Affiliated to Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| |
Collapse
|
28
|
Marega LF, Sabino JS, Pedroni MV, Teocchi M, Lanaro C, de Albuquerque DM, Dos Santos IP, Costa FF, Dos Santos Vilela MM. Phenotypes of STAT3 gain-of-function variant related to disruptive regulation of CXCL8/STAT3, KIT/STAT3, and IL-2/CD25/Treg axes. Immunol Res 2021; 69:445-456. [PMID: 34390446 DOI: 10.1007/s12026-021-09225-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2021] [Accepted: 07/27/2021] [Indexed: 12/01/2022]
Abstract
STAT3 is a cytokine-signaling transcription factor critical for gene regulation. Gain-of-function (GOF) mutations in STAT3 are associated with lymphoproliferation, autoimmune cytopenias, increased susceptibility to infection, early-onset solid-organ autoimmunity, short stature, and eczema. We studied the JAK/STAT signaling pathway gene expression and the cytokine profile in two families carrying STAT3-GOF variants to shed light on the STAT3-GOF-associated variable expressivity, including the identification of disease markers. Considering 92 target genes, KIT and IL2RA were downregulated only in patients with high clinical penetrance, while CXCL8 was markedly downregulated for all of them. Unlike previous studies, SOCS3-a STAT3 inhibitor-was not upregulated in patients. In addition, low levels of IL-2 and a reduced numbers of Tregs cells were strikingly prevalent in patients. This study shows a disruptive role of STAT3-GOF variants in the regulatory axis activities CXCL8/STAT3, KIT/STAT3, IL2/CD25/Treg, which, by slightly different mechanisms, underlie the broad clinical spectrum seen in the studied patients. In addition, we suggest the investigation of CXCL8 as a biomarker for identifying STAT3-GOF mutation.
Collapse
Affiliation(s)
- Lia Furlaneto Marega
- Laboratory of Pediatric Immunology, Center for Investigation in Pediatrics, School of Medical Sciences, University of Campinas (UNICAMP), Rua Tessália Vieira de Camargo, 126, Cidade Universitária "Zeferino Vaz", São Paulo, CEP 13083-887, Campinas, Brazil
| | - Janine Schincariol Sabino
- Laboratory of Pediatric Immunology, Center for Investigation in Pediatrics, School of Medical Sciences, University of Campinas (UNICAMP), Rua Tessália Vieira de Camargo, 126, Cidade Universitária "Zeferino Vaz", São Paulo, CEP 13083-887, Campinas, Brazil
| | - Marcus Vinicius Pedroni
- Laboratory of Pediatric Immunology, Center for Investigation in Pediatrics, School of Medical Sciences, University of Campinas (UNICAMP), Rua Tessália Vieira de Camargo, 126, Cidade Universitária "Zeferino Vaz", São Paulo, CEP 13083-887, Campinas, Brazil
| | - Marcelo Teocchi
- Laboratory of Pediatric Immunology, Center for Investigation in Pediatrics, School of Medical Sciences, University of Campinas (UNICAMP), Rua Tessália Vieira de Camargo, 126, Cidade Universitária "Zeferino Vaz", São Paulo, CEP 13083-887, Campinas, Brazil
| | - Carolina Lanaro
- Hematology and Hemotherapy Center - Hemocentro Campinas, University of Campinas (UNICAMP), SP, Campinas, Brazil
| | | | - Irene Pereira Dos Santos
- Flow Cytometry Laboratory, Hematology and Hemotherapy Center, University of Campinas (UNICAMP), SP, Campinas, Brazil
| | - Fernando Ferreira Costa
- Hematology and Hemotherapy Center - Hemocentro Campinas, University of Campinas (UNICAMP), SP, Campinas, Brazil
| | - Maria Marluce Dos Santos Vilela
- Laboratory of Pediatric Immunology, Center for Investigation in Pediatrics, School of Medical Sciences, University of Campinas (UNICAMP), Rua Tessália Vieira de Camargo, 126, Cidade Universitária "Zeferino Vaz", São Paulo, CEP 13083-887, Campinas, Brazil.
| |
Collapse
|
29
|
Kumar A, Cao W, Endrias K, Kuchipudi SV, Mittal SK, Sambhara S. Innate lymphoid cells (ILC) in SARS-CoV-2 infection. Mol Aspects Med 2021; 80:101008. [PMID: 34399986 PMCID: PMC8361007 DOI: 10.1016/j.mam.2021.101008] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2021] [Accepted: 08/10/2021] [Indexed: 12/21/2022]
Abstract
Innate Lymphoid Cells (ILCs) are a class of innate immune cells that form the first line of defense against internal or external abiotic and biotic challenges in the mammalian hosts. As they reside in both the lymphoid and non-lymphoid tissues, they are involved in clearing the pathogens through direct killing or by secretion of cytokines that modulate the adaptive immune responses. There is burgeoning evidence that these cells are important in clearing viral infections; therefore, it is critical to understand their role in the resolution or exacerbation of the disease caused by severe acute respiratory syndrome coronavirus (SARS-CoV-2). In this review, we summarize the recent findings related to ILCs in response to SARS-CoV-2 infections.
Collapse
Affiliation(s)
- Amrita Kumar
- Immunology and Pathogenesis Branch, Influenza Division, National Center for Immunization and Respiratory Diseases, Centers for Disease Control and Prevention, Atlanta, GA, USA
| | - Weiping Cao
- Immunology and Pathogenesis Branch, Influenza Division, National Center for Immunization and Respiratory Diseases, Centers for Disease Control and Prevention, Atlanta, GA, USA
| | - Kedan Endrias
- College of Arts and Sciences, Georgia State University, Atlanta, GA, USA
| | - Suresh V Kuchipudi
- Department of Veterinary and Biomedical Sciences and the HUCJ Institutes of Life Sciences, Penn State University, University Park, PA, USA
| | - Suresh K Mittal
- Department of Comparative Pathobiology, Purdue Institute for Inflammation, Immunology and Infectious Disease, and Purdue University Center for Cancer Research, College of Veterinary Medicine, Purdue University, West Lafayette, IN, USA
| | - Suryaprakash Sambhara
- Immunology and Pathogenesis Branch, Influenza Division, National Center for Immunization and Respiratory Diseases, Centers for Disease Control and Prevention, Atlanta, GA, USA.
| |
Collapse
|
30
|
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: 1] [Impact Index Per Article: 0.3] [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
|
31
|
Bartemes KR, Kita H. Roles of innate lymphoid cells (ILCs) in allergic diseases: The 10-year anniversary for ILC2s. J Allergy Clin Immunol 2021; 147:1531-1547. [PMID: 33965091 DOI: 10.1016/j.jaci.2021.03.015] [Citation(s) in RCA: 44] [Impact Index Per Article: 14.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2021] [Revised: 03/01/2021] [Accepted: 03/03/2021] [Indexed: 12/12/2022]
Abstract
In the 12 years since the discovery of innate lymphoid cells (ILCs), our knowledge of their immunobiology has expanded rapidly. Group 2 ILCs (ILC2s) respond rapidly to allergen exposure and environmental insults in mucosal organs, producing type 2 cytokines. Early studies showed that epithelium-derived cytokines activate ILC2s, resulting in eosinophilia, mucus hypersecretion, and remodeling of mucosal tissues. We now know that ILC2s are regulated by other cytokines, eicosanoids, and neuropeptides as well, and interact with both immune and stromal cells. Furthermore, ILC2s exhibit plasticity by adjusting their functions depending on their tissue environment and may consist of several heterogeneous subpopulations. Clinical studies show that ILC2s are involved in asthma, allergic rhinitis, chronic rhinosinusitis, food allergy, and eosinophilic esophagitis. However, much remains unknown about the immunologic mechanisms involved. Beneficial functions of ILCs in maintenance or restoration of tissue well-being and human health also need to be clarified. As our understanding of the crucial functions ILCs play in both homeostasis and disease pathology expands, we are poised to make tremendous strides in diagnostic and therapeutic options for patients with allergic diseases. This review summarizes discoveries in immunobiology of ILCs and their roles in allergic diseases in the past 5 years, discusses controversies and gaps in our knowledge, and suggests future research directions.
Collapse
Affiliation(s)
- Kathleen R Bartemes
- Division of Allergic Diseases and Department of Medicine, Mayo Clinic, Rochester, Minn; Department of Otolaryngology - Head and Neck Surgery, Mayo Clinic, Rochester, Minn
| | - Hirohito Kita
- Department of Immunology, Mayo Clinic, Rochester, Minn; Division of Allergy, Asthma, and Immunology and Department of Medicine, Mayo Clinic, Scottsdale, Ariz.
| |
Collapse
|
32
|
Ghaedi M, Takei F. Innate lymphoid cell development. J Allergy Clin Immunol 2021; 147:1549-1560. [PMID: 33965092 DOI: 10.1016/j.jaci.2021.03.009] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2020] [Revised: 02/26/2021] [Accepted: 03/03/2021] [Indexed: 12/25/2022]
Abstract
Innate lymphoid cells (ILCs) mainly reside at barrier surfaces and regulate tissue homeostasis and immunity. ILCs are divided into 3 groups, group 1 ILCs, group 2 ILCs, and group 3 ILC3, on the basis of their similar effector programs to T cells. The development of ILCs from lymphoid progenitors in adult mouse bone marrow has been studied in detail, and multiple ILC progenitors have been characterized. ILCs are mostly tissue-resident cells that develop in the perinatal period. More recently, ILC progenitors have also been identified in peripheral tissues. In this review, we discuss the stepwise transcription factor-directed differentiation of mouse ILC progenitors into mature ILCs, the critical time windows in ILC development, and the contribution of bone marrow versus tissue ILC progenitors to the pool of mature ILCs in tissues.
Collapse
Affiliation(s)
- Maryam Ghaedi
- Princess Margaret Cancer Centre, University Health Network, Toronto, Ontario, Canada
| | - Fumio Takei
- the Department of Pathology and Laboratory Medicine, University of British Columbia (UBC), Vancouver, British Columbia, Canada; Terry Fox Laboratory, B.C. Cancer, Vancouver, British Columbia, Canada.
| |
Collapse
|
33
|
NK cell and ILC heterogeneity in colorectal cancer. New perspectives from high dimensional data. Mol Aspects Med 2021; 80:100967. [PMID: 33941383 DOI: 10.1016/j.mam.2021.100967] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2021] [Revised: 04/20/2021] [Accepted: 04/23/2021] [Indexed: 12/14/2022]
Abstract
Innate lymphoid cells (ILCs) and tissue-resident natural killer (NK) cells ensure immunity at environmental interfaces and help maintain barrier integrity of the intestinal tract. This wide range of innate lymphocytes is able to provide fast and potent inflammatory responses that, when deregulated, have been associated with pathogenesis of inflammatory bowel disease (IBD) and colorectal cancer (CRC). While the presence of tumor-infiltrating NK cells is generally associated with a favorable outcome in CRC patients, emerging evidence reveals distinct roles for ILCs in regulating CRC pathogenesis and progression. Advances in next generation sequencing technology, and in particular of single-cell RNA-seq approaches, along with multidimensional flow cytometry analysis, have helped to deconvolute the complexity and heterogeneity of the ILC system both in homeostatic and pathological contexts. In this review, we discuss the protective and detrimental roles of NK cells and ILCs in the pathogenesis of CRC, focusing on the phenotypic and transcriptional modifications these cells undergo during CRC development and progression.
Collapse
|
34
|
Piperoglou C, Larid G, Vallentin B, Balligand L, Crinier A, Banzet N, Farnarier C, Gomez-Massa E, Adalia AC, Michel G, Galambrun C, Barlogis V, Vivier E, Vély F. Innate lymphoid cell recovery and occurrence of GvHD after hematopoietic stem cell transplantation. J Leukoc Biol 2021; 111:161-172. [PMID: 33847423 DOI: 10.1002/jlb.5a1019-522rr] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2019] [Revised: 03/01/2021] [Accepted: 03/03/2021] [Indexed: 11/09/2022] Open
Abstract
Lymphocytes are essential for microbial immunity, tumor surveillance, and tissue homeostasis. However, the in vivo development and function of helper-like innate lymphoid cells (ILCs) in humans remain much less well understood than those of T, B, and NK cells. We monitored hematopoietic stem cell transplantation (HSCT) to determine the kinetics of ILC development in both children and adults. It was found that, unlike NK cells, helper-like ILCs recovered slowly, mirroring the pattern observed for T cells, with normalization achieved at 1 year. The type of graft and the proportion of CD34+ cells in the graft did not significantly affect ILC reconstitution. As HSCT is often complicated by acute or chronic graft-versus-host disease (GVHD), the potential role of ILC subsets in maintaining tissue integrity in these conditions was also analyzed. It was found that GVHD was associated with lower levels of activated and gut-homing NKp44+ ILCP, consistent with a non-redundant role of this ILC subset in preventing this life-threatening disorder in lymphopenic conditions.
Collapse
Affiliation(s)
- Christelle Piperoglou
- APHM, Hôpital de la Timone, Service d'Immunologie, Marseille-Immunopole, Marseille, France
| | - Guillaume Larid
- APHM, Hôpital de la Timone, Service d'Immunologie, Marseille-Immunopole, Marseille, France
| | - Blandine Vallentin
- APHM, Hôpital de la Timone, Service d'Hématologie et Oncologie Pédiatrique, Marseille, France
| | - Laura Balligand
- APHM, Hôpital de la Timone, Service d'Hématologie et Oncologie Pédiatrique, Marseille, France
| | | | - Nathalie Banzet
- APHM, Hôpital de la Timone, Service d'Immunologie, Marseille-Immunopole, Marseille, France
| | - Catherine Farnarier
- APHM, Hôpital de la Timone, Service d'Immunologie, Marseille-Immunopole, Marseille, France
| | | | | | -
- APHM, Hôpital de la Timone, Service d'Immunologie, Marseille-Immunopole, Marseille, France
| | - Gérard Michel
- APHM, Hôpital de la Timone, Service d'Hématologie et Oncologie Pédiatrique, Marseille, France
| | - Claire Galambrun
- APHM, Hôpital de la Timone, Service d'Hématologie et Oncologie Pédiatrique, Marseille, France
| | - Vincent Barlogis
- APHM, Hôpital de la Timone, Service d'Hématologie et Oncologie Pédiatrique, Marseille, France
| | - Eric Vivier
- APHM, Hôpital de la Timone, Service d'Immunologie, Marseille-Immunopole, Marseille, France.,Aix Marseille Univ, CNRS, INSERM, CIML, Marseille, France.,Innate Pharma Research Labs, Innate Pharma, Marseille, France
| | - Frédéric Vély
- APHM, Hôpital de la Timone, Service d'Immunologie, Marseille-Immunopole, Marseille, France.,Aix Marseille Univ, CNRS, INSERM, CIML, Marseille, France
| |
Collapse
|
35
|
Campana S, De Pasquale C, Barberi C, Oliveri D, Sidoti Migliore G, Galletti B, Guarneri C, Cannavò SP, Ferlazzo G. Circulating ILC precursors expressing CD62L exhibit a type 2 signature distinctly decreased in psoriatic patients. Eur J Immunol 2021; 51:1792-1798. [PMID: 33728641 PMCID: PMC8360187 DOI: 10.1002/eji.202048893] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2020] [Revised: 12/23/2020] [Accepted: 03/10/2021] [Indexed: 11/21/2022]
Abstract
Human CD117+CRTH2neg innate lymphoid cells (ILC) comprise multipotent precursors (ILCp), which are able to differentiate into subtypes in response to different signals received in peripheral tissues. NKp46+ ILCp have been reported to associate with ILC3 whereas KLRG1+ILCp with ILC2, although the latter can also generate other ILC subsets, thus, maintaining a substantial plasticity. We here showed that CD62L is expressed by ILCp exclusively within KLRG1+ population and its expression marks a loss of their broad differentiation potential. Analysis of cytokine production and relevant markers demonstrated that CD62L+ILCp mainly differentiate into ILC2 whereas CD62Lneg counterpart can also differentiate into other ILC subsets depending on the signals they receive. Remarkably, in peripheral blood of psoriatic patients, where ILC3 are usually enriched, CD62L+ILC were drastically reduced, whereas CD62LnegILC2 upregulated both RORγt and NKp46, thus, suggesting an ongoing conversion to ILC3. Therefore, CD62L now emerges as a potential marker to identify a skewing toward type 2 among ILCp.
Collapse
Affiliation(s)
- Stefania Campana
- Department of Human Pathology, Laboratory of Immunology and Biotherapy, University of Messina, Messina, Italy
| | - Claudia De Pasquale
- Department of Human Pathology, Laboratory of Immunology and Biotherapy, University of Messina, Messina, Italy
| | - Chiara Barberi
- Department of Experimental Medicine (DIMES), University of Genoa, Genoa, Italy
| | - Daniela Oliveri
- Cell Factory Center and Division of Clinical Pathology, University Hospital Policlinico G. Martino, Messina, Italy
| | | | - Bruno Galletti
- Unit of Otorhinolaryngology, University Hospital Policlinico G. Martino, Messina, Italy
| | - Claudio Guarneri
- Unit of Dermatology, University Hospital Policlinico G. Martino, Messina, Italy
| | | | - Guido Ferlazzo
- Department of Human Pathology, Laboratory of Immunology and Biotherapy, University of Messina, Messina, Italy.,Cell Factory Center and Division of Clinical Pathology, University Hospital Policlinico G. Martino, Messina, Italy
| |
Collapse
|
36
|
van der Ploeg EK, Golebski K, van Nimwegen M, Fergusson JR, Heesters BA, Martinez-Gonzalez I, Kradolfer CMA, van Tol S, Scicluna BP, de Bruijn MJW, de Boer GM, Tramper-Stranders GA, Braunstahl GJ, van IJcken WFJ, Nagtegaal AP, van Drunen CM, Fokkens WJ, Huylebroeck D, Spits H, Hendriks RW, Stadhouders R, Bal SM. Steroid-resistant human inflammatory ILC2s are marked by CD45RO and elevated in type 2 respiratory diseases. Sci Immunol 2021; 6:6/55/eabd3489. [PMID: 33514640 DOI: 10.1126/sciimmunol.abd3489] [Citation(s) in RCA: 65] [Impact Index Per Article: 21.7] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2020] [Accepted: 12/23/2020] [Indexed: 12/13/2022]
Abstract
Group 2 innate lymphoid cells (ILC2s) orchestrate protective type 2 immunity and have been implicated in various immune disorders. In the mouse, circulatory inflammatory ILC2s (iILC2s) were identified as a major source of type 2 cytokines. The human equivalent of the iILC2 subset remains unknown. Here, we identify a human inflammatory ILC2 population that resides in inflamed mucosal tissue and is specifically marked by surface CD45RO expression. CD45RO+ ILC2s are derived from resting CD45RA+ ILC2s upon activation by epithelial alarmins such as IL-33 and TSLP, which is tightly linked to STAT5 activation and up-regulation of the IRF4/BATF transcription factors. Transcriptome analysis reveals marked similarities between human CD45RO+ ILC2s and mouse iILC2s. Frequencies of CD45RO+ inflammatory ILC2 are increased in inflamed mucosal tissue and in the circulation of patients with chronic rhinosinusitis or asthma, correlating with disease severity and resistance to corticosteroid therapy. CD45RA-to-CD45RO ILC2 conversion is suppressed by corticosteroids via induction of differentiation toward an immunomodulatory ILC2 phenotype characterized by low type 2 cytokine and high amphiregulin expression. Once converted, however, CD45RO+ ILC2s are resistant to corticosteroids, which is associated with metabolic reprogramming resulting in the activation of detoxification pathways. Our combined data identify CD45RO+ inflammatory ILC2s as a human analog of mouse iILC2s linked to severe type 2 inflammatory disease and therapy resistance.
Collapse
Affiliation(s)
- Esmee K van der Ploeg
- Department of Pulmonary Medicine, Erasmus MC, Rotterdam, Netherlands.,Department of Cell Biology, Erasmus MC, Rotterdam, Netherlands
| | - Korneliusz Golebski
- Department of Experimental Immunology, Amsterdam University Medical Centers, University of Amsterdam, Amsterdam, Netherlands.,Department of Respiratory Medicine, Amsterdam University Medical Centers, University of Amsterdam, Amsterdam, Netherlands
| | | | - Joannah R Fergusson
- Department of Experimental Immunology, Amsterdam University Medical Centers, University of Amsterdam, Amsterdam, Netherlands
| | - Balthasar A Heesters
- Department of Experimental Immunology, Amsterdam University Medical Centers, University of Amsterdam, Amsterdam, Netherlands
| | - Itziar Martinez-Gonzalez
- Department of Experimental Immunology, Amsterdam University Medical Centers, University of Amsterdam, Amsterdam, Netherlands
| | - Chantal M A Kradolfer
- Department of Experimental Immunology, Amsterdam University Medical Centers, University of Amsterdam, Amsterdam, Netherlands
| | - Sophie van Tol
- Department of Experimental Immunology, Amsterdam University Medical Centers, University of Amsterdam, Amsterdam, Netherlands
| | - Brendon P Scicluna
- Center of Experimental and Molecular Medicine, Amsterdam University Medical Centers, University of Amsterdam, Amsterdam, Netherlands.,Department of Clinical Epidemiology and Biostatistics, Amsterdam University Medical Centers, University of Amsterdam, Amsterdam, Netherlands
| | | | - Geertje M de Boer
- Department of Pulmonary Medicine, Erasmus MC, Rotterdam, Netherlands.,Department of Respiratory Medicine, Franciscus Gasthuis and Vlietland, Rotterdam, Netherlands
| | - Gerdien A Tramper-Stranders
- Department of Pediatric Medicine, Franciscus Gasthuis and Vlietland, Rotterdam, Netherlands.,Department of Neonatology, Sophia Children's Hospital, Erasmus MC, Rotterdam, Netherlands
| | - Gert-Jan Braunstahl
- Department of Pulmonary Medicine, Erasmus MC, Rotterdam, Netherlands.,Department of Respiratory Medicine, Franciscus Gasthuis and Vlietland, Rotterdam, Netherlands
| | - Wilfred F J van IJcken
- Department of Cell Biology, Erasmus MC, Rotterdam, Netherlands.,Center for Biomics, Erasmus MC, Rotterdam, Netherlands
| | - A Paul Nagtegaal
- Department of Otorhinolaryngology and Head and Neck Surgery, Erasmus MC, Rotterdam, Netherlands
| | - Cornelis M van Drunen
- Department of Otorhinolaryngology, Amsterdam University Medical Centers, University of Amsterdam, Amsterdam, Netherlands
| | - Wytske J Fokkens
- Department of Otorhinolaryngology, Amsterdam University Medical Centers, University of Amsterdam, Amsterdam, Netherlands
| | | | - Hergen Spits
- Department of Experimental Immunology, Amsterdam University Medical Centers, University of Amsterdam, Amsterdam, Netherlands.
| | - Rudi W Hendriks
- Department of Pulmonary Medicine, Erasmus MC, Rotterdam, Netherlands
| | - Ralph Stadhouders
- Department of Pulmonary Medicine, Erasmus MC, Rotterdam, Netherlands. .,Department of Cell Biology, Erasmus MC, Rotterdam, Netherlands
| | - Suzanne M Bal
- Department of Pulmonary Medicine, Erasmus MC, Rotterdam, Netherlands.,Department of Experimental Immunology, Amsterdam University Medical Centers, University of Amsterdam, Amsterdam, Netherlands
| |
Collapse
|
37
|
De Salvo C, Buela KA, Pizarro TT. Cytokine-Mediated Regulation of Innate Lymphoid Cell Plasticity in Gut Mucosal Immunity. Front Immunol 2020; 11:585319. [PMID: 33424837 PMCID: PMC7794016 DOI: 10.3389/fimmu.2020.585319] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2020] [Accepted: 11/03/2020] [Indexed: 12/23/2022] Open
Abstract
Mucosal barriers are active sites that encounter a bombardment of antigenic stimuli derived from both the commensal flora and a variety of pathogens, as well as from environmental insults. As such, the ability to mount appropriate innate immune responses is an important first line of defense that confers protection to the host. Central to innate immunity are innate lymphoid cells (ILCs), which were first described a decade ago, and represent a family of heterogeneous cells driven by specific transcription factors and exhibit distinct cytokine profiles that are shared with their CD4+ T-helper cell counterparts. ILCs are particularly enriched at mucosal surfaces, and the tissue microenvironment and cytokine milieu in which ILCs reside are critical factors that drive the behavior and overall function of these cells. In fact, ILCs situated at mucosal barriers must be able to temper their response to a constant exposure of environmental antigens, but also promptly react to pathogens or signals that are potentially harmful to the host. In this context, the ability of ILCs to readily transdifferentiate in response to their dynamic surroundings has become a vigorous area of research, and defining specific mechanism(s) of ILC plasticity is at the advent of discovery. This review will summarize what is currently known regarding the network of cytokines and regulatory elements that enable ILCs to readily transform, based on the range of diverse signals and signal gradients they encounter that lead to either protective or pathogenic function(s), with focus on the gut mucosal immune system.
Collapse
Affiliation(s)
| | | | - Theresa T. Pizarro
- Department of Pathology, Case Western Reserve University School of Medicine, Cleveland, OH, United States
| |
Collapse
|
38
|
Darboe A, Nielsen CM, Wolf AS, Wildfire J, Danso E, Sonko B, Bottomley C, Moore SE, Riley EM, Goodier MR. Age-Related Dynamics of Circulating Innate Lymphoid Cells in an African Population. Front Immunol 2020; 11:594107. [PMID: 33343571 PMCID: PMC7738635 DOI: 10.3389/fimmu.2020.594107] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2020] [Accepted: 10/28/2020] [Indexed: 12/15/2022] Open
Abstract
Innate lymphoid cell (ILC) lineages mirror those of CD4+ T helper cell subsets, producing type 1, 2 and 3 cytokines respectively. Studies in adult human populations have shown contributions of non-cytotoxic ILC to immune regulation or pathogenesis in a wide range of diseases and have prompted investigations of potential functional redundancy between ILC and T helper cell compartments in neonates and children. To investigate the potential for ILC to contribute to immune responses across the human lifespan, we examined the numbers and frequencies of peripheral blood ILC subsets in a cohort of Gambians aged between 5 and 73 years of age. ILC2 were the most abundant peripheral blood ILC subset in this Gambian cohort, while ILC1 were the rarest at all ages. Moreover, the frequency of ILC1s (as a proportion of all lymphocytes) was remarkably stable over the life course whereas ILC3 cell frequencies and absolute numbers declined steadily across the life course and ILC2 frequencies and absolute numbers declined from childhood until the age of approx. 30 years of age. Age-related reductions in ILC2 cell numbers appeared to be partially offset by increasing numbers of total and GATA3+ central memory (CD45RA-CCR7+) CD4+ T cells, although there was also a gradual decline in numbers of total and GATA3+ effector memory (CD45RA-CCR7-) CD4+ T cells. Despite reduced overall abundance of ILC2 cells, we observed a coincident increase in the proportion of CD117+ ILC2, indicating potential for age-related adaptation of these cells in childhood and early adulthood. While both CD117+ and CD117- ILC2 cells produced IL-13, these responses occurred predominantly within CD117- cells. Furthermore, comparison of ILC frequencies between aged-matched Gambian and UK young adults (25–29 years) revealed an overall higher proportion of ILC1 and ILC2, but not ILC3 in Gambians. Thus, these data indicate ongoing age-related changes in ILC2 cells throughout life, which retain the capacity to differentiate into potent type 2 cytokine producing cells, consistent with an ongoing role in immune modulation.
Collapse
Affiliation(s)
- Alansana Darboe
- Department of Infection Biology, London School of Hygiene and Tropical Medicine, London, United Kingdom.,Vaccines & Immunity Theme, Infant Immunology, MRC Unit The Gambia at London School of Hygiene and Tropical Medicine, Fajara, Gambia
| | - Carolyn M Nielsen
- Nuffield Department of Medicine, University of Oxford, Oxford, United Kingdom
| | - Asia-Sophia Wolf
- Division of Infection and Immunity, University College London, London, United Kingdom
| | - Jacob Wildfire
- Department of Infection Biology, London School of Hygiene and Tropical Medicine, London, United Kingdom
| | - Ebrima Danso
- Nutrition Theme, MRC International Group, MRC Unit The Gambia at London School of Hygiene and Tropical Medicine, Keneba, Gambia
| | - Bakary Sonko
- Nutrition Theme, MRC International Group, MRC Unit The Gambia at London School of Hygiene and Tropical Medicine, Keneba, Gambia
| | - Christian Bottomley
- Department of Infectious Disease Epidemiology, London School of Hygiene and Tropical Medicine, London, United Kingdom
| | - Sophie E Moore
- Nutrition Theme, MRC International Group, MRC Unit The Gambia at London School of Hygiene and Tropical Medicine, Keneba, Gambia.,Women & Children's Health, Kings College London, London, United Kingdom
| | - Eleanor M Riley
- Institute of Immunology and Infection Research, School of Biological Sciences, University of Edinburgh, Edinburgh, United Kingdom
| | - Martin R Goodier
- Department of Infection Biology, London School of Hygiene and Tropical Medicine, London, United Kingdom
| |
Collapse
|
39
|
Maggi E, Veneziani I, Moretta L, Cosmi L, Annunziato F. Group 2 Innate Lymphoid Cells: A Double-Edged Sword in Cancer? Cancers (Basel) 2020; 12:cancers12113452. [PMID: 33233582 PMCID: PMC7699723 DOI: 10.3390/cancers12113452] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2020] [Revised: 11/16/2020] [Accepted: 11/17/2020] [Indexed: 12/14/2022] Open
Abstract
Group 2 Innate Lymphoid Cells (ILC2s) belong to the family of helper ILCs which provide host defense against infectious agents, participate in inflammatory responses and mediate lymphoid organogenesis and tissue repair, mainly at the skin and mucosal level. Based on their transcriptional, phenotypic and functional profile, ILC2s mirror the features of the adaptive CD4+ Th2 cell subset, both contributing to the so-called type 2 immune response. Similar to other ILCs, ILC2s are rapidly activated by signals deriving from tissue and/or other tissue-resident immune cells. The biologic activity of ILCs needs to be tightly regulated in order to prevent them from contributing to severe inflammation and damage in several organs. Indeed, ILC2s display both enhancing and regulatory roles in several pathophysiological conditions, including tumors. In this review, we summarize the actual knowledge about ILC2s ability to induce or impair a protective immune response, their pro- or antitumor activity in murine models, human (children and adults) pathologies and the potential strategies to improve cancer immunotherapy by exploiting the features of ILC2s.
Collapse
Affiliation(s)
- Enrico Maggi
- Immunology Department, Bambino Gesù Children Hospital, 00165 Rome, Italy; (I.V.); (L.M.)
- Correspondence: ; Tel.: +39-06-6859-3617
| | - Irene Veneziani
- Immunology Department, Bambino Gesù Children Hospital, 00165 Rome, Italy; (I.V.); (L.M.)
| | - Lorenzo Moretta
- Immunology Department, Bambino Gesù Children Hospital, 00165 Rome, Italy; (I.V.); (L.M.)
| | - Lorenzo Cosmi
- Department of Experimental and Clinical Medicine, University of Florence, 50134 Florence, Italy; (L.C.); (F.A.)
| | - Francesco Annunziato
- Department of Experimental and Clinical Medicine, University of Florence, 50134 Florence, Italy; (L.C.); (F.A.)
| |
Collapse
|
40
|
Tufa DM, Yingst AM, Trahan GD, Shank T, Jones D, Shim S, Lake J, Winkler K, Cobb L, Woods R, Jones K, Verneris MR. Human innate lymphoid cell precursors express CD48 that modulates ILC differentiation through 2B4 signaling. Sci Immunol 2020; 5:eaay4218. [PMID: 33219153 PMCID: PMC8294935 DOI: 10.1126/sciimmunol.aay4218] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2019] [Revised: 06/15/2020] [Accepted: 10/28/2020] [Indexed: 12/11/2022]
Abstract
Innate lymphoid cells (ILCs) develop from common lymphoid progenitors (CLPs), which further differentiate into the common ILC progenitor (CILP) that can give rise to both ILCs and natural killer (NK) cells. Murine ILC intermediates have recently been characterized, but the human counterparts and their developmental trajectories have not yet been identified, largely due to the lack of homologous surface receptors in both organisms. Here, we show that human CILPs (CD34+CD117+α4β7+Lin-) acquire CD48 and CD52, which define NK progenitors (NKPs) and ILC precursors (ILCPs). Two distinct NK cell subsets were generated in vitro from CD34+CD117+α4β7+Lin-CD48-CD52+ and CD34+CD117+α4β7+Lin-CD48+CD52+ NKPs, respectively. Independent of NKPs, ILCPs exist in the CD34+CD117+α4β7+Lin-CD48+CD52+ subset and give rise to ILC1s, ILC2s, and NCR+ ILC3s, whereas CD34+CD117+α4β7+Lin-CD48+CD52- ILCPs give rise to a distinct subset of ILC3s that have lymphoid tissue inducer (LTi)-like properties. In addition, CD48-expressing CD34+CD117+α4β7+Lin- precursors give rise to tissue-associated ILCs in vivo. We also observed that the interaction of 2B4 with CD48 induced differentiation of ILC2s, and together, these findings show that expression of CD48 by human ILCPs modulates ILC differentiation.
Collapse
Affiliation(s)
- Dejene M Tufa
- Department of Pediatric, Division of Children's Cancer and Blood Disorders, University of Colorado and Children's Hospital of Colorado, Research Complex 1, North Tower, 12800 E. 19th Ave., Mail Stop 8302, Room P18-4108, Aurora, CO 80045, USA
| | - Ashley M Yingst
- Department of Pediatric, Division of Children's Cancer and Blood Disorders, University of Colorado and Children's Hospital of Colorado, Research Complex 1, North Tower, 12800 E. 19th Ave., Mail Stop 8302, Room P18-4108, Aurora, CO 80045, USA
| | - George Devon Trahan
- Department of Pediatric, Division of Children's Cancer and Blood Disorders, University of Colorado and Children's Hospital of Colorado, Research Complex 1, North Tower, 12800 E. 19th Ave., Mail Stop 8302, Room P18-4108, Aurora, CO 80045, USA
| | - Tyler Shank
- Department of Pediatric, Division of Children's Cancer and Blood Disorders, University of Colorado and Children's Hospital of Colorado, Research Complex 1, North Tower, 12800 E. 19th Ave., Mail Stop 8302, Room P18-4108, Aurora, CO 80045, USA
| | - Dallas Jones
- Department of Pediatric, Division of Children's Cancer and Blood Disorders, University of Colorado and Children's Hospital of Colorado, Research Complex 1, North Tower, 12800 E. 19th Ave., Mail Stop 8302, Room P18-4108, Aurora, CO 80045, USA
| | - Seonhui Shim
- Department of Pediatric, Division of Children's Cancer and Blood Disorders, University of Colorado and Children's Hospital of Colorado, Research Complex 1, North Tower, 12800 E. 19th Ave., Mail Stop 8302, Room P18-4108, Aurora, CO 80045, USA
| | - Jessica Lake
- Department of Pediatric, Division of Children's Cancer and Blood Disorders, University of Colorado and Children's Hospital of Colorado, Research Complex 1, North Tower, 12800 E. 19th Ave., Mail Stop 8302, Room P18-4108, Aurora, CO 80045, USA
| | - Kevin Winkler
- Department of Pediatric, Division of Children's Cancer and Blood Disorders, University of Colorado and Children's Hospital of Colorado, Research Complex 1, North Tower, 12800 E. 19th Ave., Mail Stop 8302, Room P18-4108, Aurora, CO 80045, USA
| | - Laura Cobb
- Department of Pediatric, Division of Children's Cancer and Blood Disorders, University of Colorado and Children's Hospital of Colorado, Research Complex 1, North Tower, 12800 E. 19th Ave., Mail Stop 8302, Room P18-4108, Aurora, CO 80045, USA
| | - Renee Woods
- Department of Pediatric, Division of Children's Cancer and Blood Disorders, University of Colorado and Children's Hospital of Colorado, Research Complex 1, North Tower, 12800 E. 19th Ave., Mail Stop 8302, Room P18-4108, Aurora, CO 80045, USA
| | - Kenneth Jones
- Department of Pediatric, Division of Children's Cancer and Blood Disorders, University of Colorado and Children's Hospital of Colorado, Research Complex 1, North Tower, 12800 E. 19th Ave., Mail Stop 8302, Room P18-4108, Aurora, CO 80045, USA
| | - Michael R Verneris
- Department of Pediatric, Division of Children's Cancer and Blood Disorders, University of Colorado and Children's Hospital of Colorado, Research Complex 1, North Tower, 12800 E. 19th Ave., Mail Stop 8302, Room P18-4108, Aurora, CO 80045, USA.
| |
Collapse
|
41
|
van der Ploeg EK, Hermans MAW, van der Velden VHJ, Dik WA, van Daele PLA, Stadhouders R. Increased group 2 innate lymphoid cells in peripheral blood of adults with mastocytosis. J Allergy Clin Immunol 2020; 147:1490-1496.e2. [PMID: 33091410 DOI: 10.1016/j.jaci.2020.09.037] [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: 04/23/2020] [Revised: 08/24/2020] [Accepted: 09/04/2020] [Indexed: 01/24/2023]
Abstract
BACKGROUND Systemic mastocytosis is a hematological disease in which aberrant mast cells accumulate because of gain-of-function mutations in the KIT receptor. Group 2 innate lymphoid cells (ILC2s) are effector cells of type 2 immune responses that also express KIT and colocalize with mast cells at barrier tissue sites. In mouse models, mast cell-ILC2 crosstalk can drive local inflammation. However, a possible role for ILC2s in the pathophysiology of mastocytosis remains unexplored. OBJECTIVE We sought to characterize circulating ILC2s in a clinically diverse cohort of patients with mastocytosis. METHODS We included 21 adults with systemic mastocytosis and 18 healthy controls. Peripheral blood ILC2 abundance and phenotype were analyzed by flow cytometry and correlated to clinical characteristics, including the presence of the D816V KIT mutation. RESULTS ILC2 levels were significantly higher in D816V+ patients with mastocytosis compared with D816V- patients or healthy controls. We observed increased proportions of KIT+ ILC2s among patients with mastocytosis, regardless of D816V status. Patients with skin involvement and itch showed the highest levels of ILC2s, which was independent from atopy or serum tryptase levels. Allele-specific quantitative PCR showed that the vast majority of ILC2s did not carry the D816V mutation. CONCLUSIONS Our findings suggest a role for ILC2s and pathogenic ILC2-mast cell crosstalk in mastocytosis. We hypothesize that a high cutaneous D816V+ mast cell burden alters the skin microenvironment to induce chronic local ILC2 activation and their dissemination into the circulation. Activated ILC2s could contribute to skin symptoms by producing inflammatory mediators and by further augmenting mast cell mediator release.
Collapse
Affiliation(s)
- Esmee K van der Ploeg
- Department of Pulmonary Medicine, Erasmus MC, Rotterdam, The Netherlands; Department of Cell Biology, Erasmus MC, Rotterdam, The Netherlands
| | - Maud A W Hermans
- Section of Clinical Immunology, Department of Internal Medicine, Erasmus MC, Rotterdam, The Netherlands; Section of Allergy, Department of Internal Medicine, Erasmus MC, Rotterdam, The Netherlands
| | | | - Willem A Dik
- Laboratory of Medical Immunology, Department of Immunology, Erasmus MC, Rotterdam, The Netherlands
| | - Paul L A van Daele
- Section of Clinical Immunology, Department of Internal Medicine, Erasmus MC, Rotterdam, The Netherlands; Section of Allergy, Department of Internal Medicine, Erasmus MC, Rotterdam, The Netherlands; Laboratory of Medical Immunology, Department of Immunology, Erasmus MC, Rotterdam, The Netherlands
| | - Ralph Stadhouders
- Department of Pulmonary Medicine, Erasmus MC, Rotterdam, The Netherlands; Department of Cell Biology, Erasmus MC, Rotterdam, The Netherlands.
| |
Collapse
|
42
|
Trained immunity and tolerance in innate lymphoid cells, monocytes, and dendritic cells during allergen-specific immunotherapy. J Allergy Clin Immunol 2020; 147:1865-1877. [PMID: 33039478 DOI: 10.1016/j.jaci.2020.08.042] [Citation(s) in RCA: 50] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2020] [Revised: 08/07/2020] [Accepted: 08/17/2020] [Indexed: 12/11/2022]
Abstract
BACKGROUND Despite the efficacy of allergen-specific immunotherapy (AIT), the role of trained immunity and tolerance in this process has not been elucidated. OBJECTIVE Here, we have performed a comprehensive longitudinal analysis of the systemic innate immune cell repertoire during the course of AIT. METHODS Patients with allergy received standard preseasonal subcutaneous AIT with allergoids to birch and/or grass. Healthy controls were monitored without any intervention. Flow cytometry of innate lymphoid cell (ILC), natural killer cell, monocyte cell, and dendritic cell (DC) subsets was performed at baseline, 3 months (birch season), 6 months (grass seasons), and 12 months after the therapy in patients or at similar seasonal time points in controls. Additional analyses were performed in the third-year birch and grass season. RESULTS We observed a durable decrease in group 2 ILCs and an increase of group 1 ILCs after AIT, with dynamic changes in their composition. We found that an expansion of CD127+CD25++ clusters caused observed shifts in the heterogeneity of group 1 ILCs. In addition, we observed development of CD127+CD25++c-Kit+ group 3 ILC clusters. Moreover, we found an increase in the number of intermediate monocytes in parallel with a reduction in nonclassical monocytes during the first year after AIT. Classical and intermediate monocytes presented significant heterogeneity in patients with allergy, but AIT reduced the HLA-DR++ clusters. Finally, an increase in plasmacytoid DCs and CD141+ myeloid DCs was observed in individuals with allergy, whereas the number of CD1c+ myeloid DCs was reduced during the first year of AIT. CONCLUSION AIT induces changes in the composition and heterogeneity of circulating innate immune cells and brings them to the level observed in healthy individuals. Monitoring of ILCs, monocytes, and DCs during AIT might serve as a novel biomarker strategy.
Collapse
|
43
|
Nakamura R, Yoshizawa A, Moriyasu T, Deloer S, Senba M, Kikuchi M, Koyasu S, Moro K, Hamano S. Group 2 Innate Lymphoid Cells Exacerbate Amebic Liver Abscess in Mice. iScience 2020; 23:101544. [PMID: 33083770 PMCID: PMC7509001 DOI: 10.1016/j.isci.2020.101544] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2019] [Revised: 07/24/2020] [Accepted: 09/04/2020] [Indexed: 01/21/2023] Open
Abstract
Entamoeba histolytica, a protozoan parasite in the lumen of the human large intestine, occasionally spreads to the liver and induces amebic liver abscesses (ALAs). Upon infection with E. histolytica, high levels of type 2 cytokines are induced in the liver early after infection. However, the sources and functions of these initial type 2 cytokines in ALA formation remain unclear. In this study, we examined the roles of group 2 innate lymphoid cells (ILC2s) in ALA formation. Hepatic ILC2 numbers were significantly increased and they produced robust levels of IL-5. The in vivo transfer of ILC2s into Rag2−/−common γ chain (γc)−/− KO mice aggravated ALA formation accompanied by eosinophilia and neutrophilia. Furthermore, IL-33-deficient mice and IL-5-neutralized mice had less ALA formations. These results suggest that ILC2s contribute to exacerbating the pathogenesis of ALA by producing early type 2 cytokines and promoting the accumulation of eosinophils and neutrophils in the liver. ILC2s exacerbate ALA by promoting the accumulation of eosinophils and neutrophils Hepatic ILC2s are increased and the main source of IL-5 in the early phase of ALA Hepatic ILC2s localize with IL-33+ cells in the inflammatory areas of ALA IL-33 is a trigger of ILC2-mediated ALA formation
Collapse
Affiliation(s)
- Risa Nakamura
- Department of Parasitology, Institute of Tropical Medicine (NEKKEN), Nagasaki University, Nagasaki, Japan.,Graduate School of Biomedical Sciences Doctoral Leadership Program, Nagasaki University, Nagasaki, Japan.,The Joint Usage/Research Center on Tropical Disease, Institute of Tropical Medicine (NEKKEN), Nagasaki University, Japan
| | - Akihiro Yoshizawa
- Laboratory for Innate Immune Systems, RIKEN Center for Integrative Medical Sciences (IMS), Yokohama, Japan.,Department of Cardiovascular Medicine, International University of Health and Welfare (IUHW), School of Medicine, Chiba, Japan
| | - Taeko Moriyasu
- Graduate School of Biomedical Sciences Doctoral Leadership Program, Nagasaki University, Nagasaki, Japan.,The Joint Usage/Research Center on Tropical Disease, Institute of Tropical Medicine (NEKKEN), Nagasaki University, Japan.,Kenya Research Station, Institute of Tropical Medicine (NEKKEN), Nagasaki University, Nagasaki, Japan
| | - Sharmina Deloer
- Department of Parasitology, Institute of Tropical Medicine (NEKKEN), Nagasaki University, Nagasaki, Japan.,Graduate School of Biomedical Sciences Doctoral Leadership Program, Nagasaki University, Nagasaki, Japan.,The Joint Usage/Research Center on Tropical Disease, Institute of Tropical Medicine (NEKKEN), Nagasaki University, Japan.,Mucosal Immunity Section, Laboratory of Clinical Immunology and Microbiology (LCIM), National Institute of Allergy and infectious Diseases (NIAID), NIH, Maryland, USA
| | - Masachika Senba
- The Joint Usage/Research Center on Tropical Disease, Institute of Tropical Medicine (NEKKEN), Nagasaki University, Japan.,Department of Pathology, Institute of Tropical Medicine (NEKKEN), Nagasaki University, Nagasaki, Japan
| | - Mihoko Kikuchi
- The Joint Usage/Research Center on Tropical Disease, Institute of Tropical Medicine (NEKKEN), Nagasaki University, Japan.,Department of Immunogenetics, Institute of Tropical Medicine (NEKKEN), Nagasaki University, Nagasaki, Japan
| | - Shigeo Koyasu
- Laboratory for Immune Cell Systems, RIKEN Center for Integrative Medical Sciences (IMS), Yokohama, Japan.,Department of Microbiology and Immunology, Keio University School of Medicine, Tokyo, Japan
| | - Kazuyo Moro
- Laboratory for Innate Immune Systems, RIKEN Center for Integrative Medical Sciences (IMS), Yokohama, Japan.,Laboratory for Innate Immune Systems, Osaka University Graduate School of Medicine, Osaka, Japan
| | - Shinjiro Hamano
- Department of Parasitology, Institute of Tropical Medicine (NEKKEN), Nagasaki University, Nagasaki, Japan.,Graduate School of Biomedical Sciences Doctoral Leadership Program, Nagasaki University, Nagasaki, Japan.,The Joint Usage/Research Center on Tropical Disease, Institute of Tropical Medicine (NEKKEN), Nagasaki University, Japan
| |
Collapse
|
44
|
Meininger I, Carrasco A, Rao A, Soini T, Kokkinou E, Mjösberg J. Tissue-Specific Features of Innate Lymphoid Cells. Trends Immunol 2020; 41:902-917. [PMID: 32917510 DOI: 10.1016/j.it.2020.08.009] [Citation(s) in RCA: 105] [Impact Index Per Article: 26.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2020] [Revised: 08/07/2020] [Accepted: 08/07/2020] [Indexed: 02/06/2023]
Abstract
Although the function of the circulating immune cell compartment has been studied in detail for decades, limitations in terms of access and cell yields from peripheral tissues have restricted our understanding of tissue-based immunity, particularly in humans. Recent advances in high-throughput protein analyses, transcriptional profiling, and epigenetics have partially overcome these obstacles. Innate lymphoid cells (ILCs) are predominantly tissue-resident, and accumulating data indicate that they have significant tissue-specific functions. We summarize current knowledge of ILC phenotypes in various tissues in mice and humans, aiming to clarify ILC immunity in distinct anatomical locations.
Collapse
Affiliation(s)
- Isabel Meininger
- Center for Infectious Medicine, Department of Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Anna Carrasco
- Center for Infectious Medicine, Department of Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Anna Rao
- Center for Infectious Medicine, Department of Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Tea Soini
- Center for Infectious Medicine, Department of Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Efthymia Kokkinou
- Center for Infectious Medicine, Department of Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Jenny Mjösberg
- Center for Infectious Medicine, Department of Medicine, Karolinska Institutet, Stockholm, Sweden.
| |
Collapse
|
45
|
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: 2.5] [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
|
46
|
Ohne Y. OMIP-066: Identification of Novel Subpopulations of Human Group 2 Innate Lymphoid Cells in Peripheral Blood. Cytometry A 2020; 97:1028-1031. [PMID: 32583618 DOI: 10.1002/cyto.a.24046] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2019] [Revised: 04/14/2020] [Accepted: 05/11/2020] [Indexed: 01/13/2023]
Abstract
This 14-color flow cytometry panel was designed to identify newly described subpopulations within human group 2 innate lymphoid cells (ILC2s) and other ILC subsets. This panel also allowed to identify recently reported subpopulations of peripheral blood CRTH2- c-Kit+ ILCs. We validated this panel mostly in human peripheral blood but also confirmed that the same panel and gating strategy works well in human tonsillar cells. The panel contains a few markers indicating the activation status of ILCs. In addition, phycoerythrin (PE) channel is available for the markers of interest in each study. In the validation studies described here, PE channel was used to test the expression of some markers. These features make this panel applicable for immunophenotyping of ILCs in various disease states. © 2020 International Society for Advancement of Cytometry.
Collapse
Affiliation(s)
- Yoichiro Ohne
- Bioscience Asthma, Research and Early Development, Respiratory and Immunology, BioPharmaceuticals R&D, AstraZeneca, Gaithersburg, Maryland, USA
| |
Collapse
|
47
|
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: 2.8] [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
|
48
|
Liu GY, Deng XH, Li X, Cao YJ, Xing YF, Zhou P, Lei AH, Yang Q, Deng K, Zhang H, Zhou J. Expansion of Group 2 Innate Lymphoid Cells in Patients with End-Stage Renal Disease and Their Clinical Significance. THE JOURNAL OF IMMUNOLOGY 2020; 205:36-44. [PMID: 32444391 DOI: 10.4049/jimmunol.1901095] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/10/2019] [Accepted: 04/22/2020] [Indexed: 12/30/2022]
Abstract
Group 2 innate lymphoid cells (ILC2s) play an important role in the control of tissue inflammation and homeostasis. However, the role of ILC2s in patients with end-stage renal disease (ESRD) has never been illustrated. In this study, we investigated ILC2s in ESRD patients and their clinical significance. Results showed that the frequencies and absolute numbers of ILC2s, not group 1 innate lymphoid cells or innate lymphoid cell precursors, were significantly elevated in the peripheral blood of ESRD patients when compared with those from healthy donor controls. Moreover, ILC2s from ESRD patients displayed enhanced type 2 cytokine production and cell proliferation. Plasma from ESRD patients significantly increased ILC2 levels and enhanced their effector function after in vitro treatment. The expression of phosphorylation of STAT5 in ILC2s, as well as the amounts of IL-2 in plasma, were increased in ESRD patients when compared with those from healthy donors. Clinically, ESRD patients with higher ILC2 frequencies displayed lower incidence of infectious complications during a mean of 21 month follow-up study. The proportions of ILC2s were negatively correlated with the prognostic biomarkers of chronic kidney disease, including serum parathyroid hormone, creatinine, and phosphorus, whereas they were positively correlated with serum calcium. These observations indicate that ILC2s may play a protective role in ESRD.
Collapse
Affiliation(s)
- Gao-Yu Liu
- Joint Program in Immunology, Department of Internal Medicine, Affiliated Guangzhou Women and Children's Medical Center, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou 510623, China.,Key Laboratory of Immune Microenvironment and Disease (Ministry of Education), Department of Immunology, School of Basic Medical Sciences, Tianjin Medical University, Tianjin 300070, China.,Institute of Human Virology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou 510080, China
| | - Xiao-Hui Deng
- Joint Program in Immunology, Department of Internal Medicine, Affiliated Guangzhou Women and Children's Medical Center, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou 510623, China.,Key Laboratory of Immune Microenvironment and Disease (Ministry of Education), Department of Immunology, School of Basic Medical Sciences, Tianjin Medical University, Tianjin 300070, China.,Institute of Human Virology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou 510080, China
| | - Xing Li
- Department of Medical Oncology, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou 510630, China
| | - Ying-Jiao Cao
- Joint Program in Immunology, Department of Internal Medicine, Affiliated Guangzhou Women and Children's Medical Center, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou 510623, China.,Key Laboratory of Immune Microenvironment and Disease (Ministry of Education), Department of Immunology, School of Basic Medical Sciences, Tianjin Medical University, Tianjin 300070, China.,Institute of Human Virology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou 510080, China
| | - Yan-Fang Xing
- Department of Nephrology, The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou 510150, China
| | - Pan Zhou
- Institute of Human Virology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou 510080, China
| | - Ai-Hua Lei
- Institute of Human Virology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou 510080, China.,Institute of Pathogenic Biology, Hengyang Medical College, University of South China, Hengyang 421008, China; and
| | - Quan Yang
- Key Laboratory of Immunology, Sino-French Hoffmann Institute, School of Basic Medical Sciences, Guangzhou Medical University, Guangzhou 511436, China
| | - Kai Deng
- Institute of Human Virology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou 510080, China
| | - Hui Zhang
- Institute of Human Virology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou 510080, China
| | - Jie Zhou
- Joint Program in Immunology, Department of Internal Medicine, Affiliated Guangzhou Women and Children's Medical Center, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou 510623, China; .,Key Laboratory of Immune Microenvironment and Disease (Ministry of Education), Department of Immunology, School of Basic Medical Sciences, Tianjin Medical University, Tianjin 300070, China
| |
Collapse
|
49
|
Schulz-Kuhnt A, Greif V, Hildner K, Knipfer L, Döbrönti M, Zirlik S, Fuchs F, Atreya R, Zundler S, López-Posadas R, Neufert C, Ramming A, Kiefer A, Grüneboom A, Strasser E, Wirtz S, Neurath MF, Atreya I. ILC2 Lung-Homing in Cystic Fibrosis Patients: Functional Involvement of CCR6 and Impact on Respiratory Failure. Front Immunol 2020; 11:691. [PMID: 32457736 PMCID: PMC7221160 DOI: 10.3389/fimmu.2020.00691] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2019] [Accepted: 03/26/2020] [Indexed: 01/10/2023] Open
Abstract
Cystic fibrosis patients suffer from a progressive, often fatal lung disease, which is based on a complex interplay between chronic infections, locally accumulating immune cells and pulmonary tissue remodeling. Although group-2 innate lymphoid cells (ILC2s) act as crucial initiators of lung inflammation, our understanding of their involvement in the pathogenesis of cystic fibrosis remains incomplete. Here we report a marked decrease of circulating CCR6+ ILC2s in the blood of cystic fibrosis patients, which significantly correlated with high disease severity and advanced pulmonary failure, strongly implicating increased ILC2 homing from the peripheral blood to the chronically inflamed lung tissue in cystic fibrosis patients. On a functional level, the CCR6 ligand CCL20 was identified as potent promoter of lung-directed ILC2 migration upon inflammatory conditions in vitro and in vivo using a new humanized mouse model with light-sheet fluorescence microscopic visualization of lung-accumulated human ILC2s. In the lung, blood-derived human ILC2s were able to augment local eosinophil and neutrophil accumulation and induced a marked upregulation of pulmonary type-VI collagen expression. Studies in primary human lung fibroblasts additionally revealed ILC2-derived IL-4 and IL-13 as important mediators of this type-VI collagen-inducing effect. Taken together, the here acquired results suggest that pathologically increased CCL20 levels in cystic fibrosis airways induce CCR6-mediated lung homing of circulating human ILC2s. Subsequent ILC2 activation then triggers local production of type-VI collagen and might thereby drive extracellular matrix remodeling potentially influencing pulmonary tissue destruction in cystic fibrosis patients. Thus, modulating the lung homing capacity of circulating ILC2s and their local effector functions opens new therapeutic avenues for cystic fibrosis treatment.
Collapse
Affiliation(s)
- Anja Schulz-Kuhnt
- Department of Medicine 1, University Hospital of Erlangen, Erlangen, Germany
| | - Vicky Greif
- Department of Medicine 1, University Hospital of Erlangen, Erlangen, Germany
| | - Kai Hildner
- Department of Medicine 1, University Hospital of Erlangen, Erlangen, Germany
| | - Lisa Knipfer
- Department of Medicine 1, University Hospital of Erlangen, Erlangen, Germany
| | - Michael Döbrönti
- Department of Medicine 1, University Hospital of Erlangen, Erlangen, Germany
| | - Sabine Zirlik
- Department of Medicine 1, University Hospital of Erlangen, Erlangen, Germany
| | - Florian Fuchs
- Department of Medicine 1, University Hospital of Erlangen, Erlangen, Germany
| | - Raja Atreya
- Department of Medicine 1, University Hospital of Erlangen, Erlangen, Germany
| | - Sebastian Zundler
- Department of Medicine 1, University Hospital of Erlangen, Erlangen, Germany
| | - Rocío López-Posadas
- Department of Medicine 1, University Hospital of Erlangen, Erlangen, Germany
| | - Clemens Neufert
- Department of Medicine 1, University Hospital of Erlangen, Erlangen, Germany
| | - Andreas Ramming
- Department of Medicine 3, University Hospital of Erlangen, Erlangen, Germany
| | - Alexander Kiefer
- Department of Pediatrics and Adolescent Medicine, University Hospital of Erlangen, Erlangen, Germany
| | - Anika Grüneboom
- Department of Medicine 3, University Hospital of Erlangen, Erlangen, Germany
| | - Erwin Strasser
- Department of Transfusion Medicine and Haemostaseology, University Hospital of Erlangen, Erlangen, Germany
| | - Stefan Wirtz
- Department of Medicine 1, University Hospital of Erlangen, Erlangen, Germany
| | - Markus F Neurath
- Department of Medicine 1, University Hospital of Erlangen, Erlangen, Germany
| | - Imke Atreya
- Department of Medicine 1, University Hospital of Erlangen, Erlangen, Germany
| |
Collapse
|
50
|
Cytokines regulate the antigen-presenting characteristics of human circulating and tissue-resident intestinal ILCs. Nat Commun 2020; 11:2049. [PMID: 32341343 PMCID: PMC7184749 DOI: 10.1038/s41467-020-15695-x] [Citation(s) in RCA: 39] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2019] [Accepted: 03/23/2020] [Indexed: 12/13/2022] Open
Abstract
ILCs and T helper cells have been shown to exert bi-directional regulation in mice. However, how crosstalk between ILCs and CD4+ T cells influences immune function in humans is unknown. Here we show that human intestinal ILCs co-localize with T cells in healthy and colorectal cancer tissue and display elevated HLA-DR expression in tumor and tumor-adjacent areas. Although mostly lacking co-stimulatory molecules ex vivo, intestinal and peripheral blood (PB) ILCs acquire antigen-presenting characteristics triggered by inflammasome-associated cytokines IL-1β and IL-18. IL-1β drives the expression of HLA-DR and co-stimulatory molecules on PB ILCs in an NF-κB-dependent manner, priming them as efficient inducers of cytomegalovirus-specific memory CD4+ T-cell responses. This effect is strongly inhibited by the anti-inflammatory cytokine TGF-β. Our results suggest that circulating and tissue-resident ILCs have the intrinsic capacity to respond to the immediate cytokine milieu and regulate local CD4+ T-cell responses, with potential implications for anti-tumor immunity and inflammation. Murine ILCs can modulate T cell responses in MHCII-dependent manner. Here the authors show that human ILCs process and present antigens and induce T-cell responses upon exposure to IL-1-family cytokines; along with the article by Lehmann et al, this work elucidates how cytokines set context specificity of ILC-T cell crosstalk by regulating ILC antigen presentation.
Collapse
|