1
|
Colpitts SJ, Jegatheeswaran S, Oakie A, Mashhouri S, Sachewsky N, Murshed H, Mathews JA, Reid KT, Misra PS, Fung VCW, Reichman TW, Nostro MC, Verchere CB, Levings MK, Crome SQ. Cell therapy with human IL-10-producing ILC2s enhances islet function and inhibits allograft rejection. Am J Transplant 2025:S1600-6135(25)00280-1. [PMID: 40412656 DOI: 10.1016/j.ajt.2025.05.023] [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: 11/20/2024] [Revised: 04/30/2025] [Accepted: 05/15/2025] [Indexed: 05/27/2025]
Abstract
Group 2 innate lymphoid cells (ILC2s) that produce IL-10 (IL-10+ILC2s) have demonstrated regulatory and tissue-protective properties in murine studies, but preclinical studies are lacking that explore the potential of human IL-10+ILC2s as a tolerance-promoting cell therapy for transplantation or autoimmunity. Here, we investigated whether human IL-10+ILC2s could enhance islet function and prevent allograft rejection in humanized mouse models of islet transplantation. In vitro, human IL-10+ILC2s did not display cytotoxicity towards allogeneic deceased-donor islets or stem cell-derived islet-like cells, and co-transplantation with IL-10+ILC2s significantly improved glucose control post-transplantation. Allogeneic IL10+ILC2s directly inhibited T cell-mediated cytotoxicity against islet-like cells in vitro, and in an antigen-specific transplant rejection model, prevented T cell-mediated rejection of deceased donor islet grafts. Effects were greater with allogeneic IL-10+ILC2s, as autologous cells did not inhibit T cell IFN-γ production or cytotoxic activity in vitro, and were not sufficient to prevent islet rejection in vivo.Collectively, these studies provide proof-of-principle that human IL-10+ILC2s have therapeutic potential for islet transplantation and type 1 diabetes, and support their use as an allogeneic regulatory cell therapy.
Collapse
Affiliation(s)
- Sarah J Colpitts
- Department of Immunology, Temerty Faculty of Medicine, University of Toronto, Toronto, ON, Canada; Toronto General Hospital Research Institute, Ajmera Transplant Centre, University Health Network, Toronto, ON, Canada
| | - Sinthuja Jegatheeswaran
- Department of Immunology, Temerty Faculty of Medicine, University of Toronto, Toronto, ON, Canada; Toronto General Hospital Research Institute, Ajmera Transplant Centre, University Health Network, Toronto, ON, Canada
| | - Amanda Oakie
- McEwen Stem Cell Institute, University Health Network, Toronto, ON, Canada
| | - Siavash Mashhouri
- Department of Immunology, Temerty Faculty of Medicine, University of Toronto, Toronto, ON, Canada; Toronto General Hospital Research Institute, Ajmera Transplant Centre, University Health Network, Toronto, ON, Canada
| | - Nadia Sachewsky
- Toronto General Hospital Research Institute, Ajmera Transplant Centre, University Health Network, Toronto, ON, Canada
| | - Humaira Murshed
- Toronto General Hospital Research Institute, Ajmera Transplant Centre, University Health Network, Toronto, ON, Canada
| | - Jessica A Mathews
- Toronto General Hospital Research Institute, Ajmera Transplant Centre, University Health Network, Toronto, ON, Canada
| | - Kyle T Reid
- Department of Immunology, Temerty Faculty of Medicine, University of Toronto, Toronto, ON, Canada; Toronto General Hospital Research Institute, Ajmera Transplant Centre, University Health Network, Toronto, ON, Canada
| | - Paraish S Misra
- McEwen Stem Cell Institute, University Health Network, Toronto, ON, Canada; Department of Physiology, University of Toronto, Toronto, ON, Canada
| | - Vivian C W Fung
- Department of Surgery, University of British Columbia, Vancouver, BC, Canada; BC Children's Hospital Research Institute, Vancouver, BC, Canada
| | - Trevor W Reichman
- Toronto General Hospital Research Institute, Ajmera Transplant Centre, University Health Network, Toronto, ON, Canada; Department of Surgery, University of Toronto, Toronto, ON, Canada
| | - M Cristina Nostro
- McEwen Stem Cell Institute, University Health Network, Toronto, ON, Canada; Department of Physiology, University of Toronto, Toronto, ON, Canada
| | - C Bruce Verchere
- Department of Surgery, University of British Columbia, Vancouver, BC, Canada; BC Children's Hospital Research Institute, Vancouver, BC, Canada; Department of Pathology & Laboratory Medicine, University of British Columbia, Vancouver, BC, Canada
| | - Megan K Levings
- Department of Surgery, University of British Columbia, Vancouver, BC, Canada; BC Children's Hospital Research Institute, Vancouver, BC, Canada; School of Biomedical Engineering, University of British Columbia, Vancouver, BC, Canada
| | - Sarah Q Crome
- Department of Immunology, Temerty Faculty of Medicine, University of Toronto, Toronto, ON, Canada; Toronto General Hospital Research Institute, Ajmera Transplant Centre, University Health Network, Toronto, ON, Canada.
| |
Collapse
|
2
|
Roberts LB, Kelly AM, Hepworth MR. There's no place like home: How local tissue microenvironments shape the function of innate lymphoid cells. Mucosal Immunol 2025; 18:279-289. [PMID: 39900201 DOI: 10.1016/j.mucimm.2025.01.012] [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: 11/21/2024] [Revised: 01/16/2025] [Accepted: 01/30/2025] [Indexed: 02/05/2025]
Abstract
Innate lymphoid cells (ILC) have emerged as critical immune effectors with key roles in orchestrating the wider immune response. While ILC are relatively rare cells they are found enriched within discrete microenvironments, predominantly within barrier tissues. An emerging body of evidence implicates complex and multi-layered interactions between cell types, tissue structure and the external environment as key determinants of ILC function within these niches. In this review we will discuss the specific components that constitute ILC-associated microenvironments and consider how they act to determine health and disease. The development of holistic, integrated models of ILC function within complex tissue environments will inform new understanding of the contextual cues and mechanisms that determine the protective versus disease-causing roles of this immune cell family.
Collapse
Affiliation(s)
- Luke B Roberts
- School of Biological Sciences, Faculty of Biology, Medicine and Health, Manchester Academic Health Science Centre, University of Manchester M13 9PL United Kingdom; Lydia Becker Institute of Immunology and Inflammation, University of Manchester, United Kingdom
| | - Alanna M Kelly
- School of Biological Sciences, Faculty of Biology, Medicine and Health, Manchester Academic Health Science Centre, University of Manchester M13 9PL United Kingdom; Lydia Becker Institute of Immunology and Inflammation, University of Manchester, United Kingdom
| | - Matthew R Hepworth
- School of Biological Sciences, Faculty of Biology, Medicine and Health, Manchester Academic Health Science Centre, University of Manchester M13 9PL United Kingdom; Lydia Becker Institute of Immunology and Inflammation, University of Manchester, United Kingdom.
| |
Collapse
|
3
|
Liu J, Gu H, Jia R, Li S, Chen Z, Zheng A, Chang W, Liu G. Effects of Lactobacillus acidophilus on production performance and immunity of broiler chickens and their mechanism. Front Vet Sci 2025; 12:1554502. [PMID: 40196813 PMCID: PMC11974341 DOI: 10.3389/fvets.2025.1554502] [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: 01/02/2025] [Accepted: 03/03/2025] [Indexed: 04/09/2025] Open
Abstract
Introduction Lactobacillus species have attracted more and more attention as a potential antibiotic substitute for human health and animal production due to their remarkable antibacterial effects. However, the underlying mechanism is unclear. This experiment's goal was to investigate the impacts of lactic acid bacteria (LAB) on the growth performance, carcass characteristics, immune function of broiler chickens and their mechanism. Methods One hundred and eighty 1-day-old AA broilers were used and randomly allocated into 3 treatment groups with 6 replicates of 10 chickens per replicate. The 3 treatment groups were control group (CK), L. acidophilus added group (LAB-E, 1.0 × 108 CFU/kg) for the first 7 days; L. acidophilus added group (LAB-A, 1.0 × 108 CFU/kg) for the whole experimental period. Broilers had free access to water and feed. Results The results showed that addition of L. acidophilus for the whole experimental period significantly decreased ADFI, FCR and the abdominal fat percentage of broilers (p < 0.05), tended to increase the levels of IgG in broiler serum (p = 0.093). The LAB-A group had higher HDL-C content and IL-2, IL-4 content, and lower level of LPS in broiler serum compared to the controls (p < 0.05). Discussion In conclusion, L. acidophilus improved feed efficiency and immune function of broilers by controlling nutrient metabolism and inflammation responses of broilers. L. acidophilus can be used as a potential substitute for antibiotics in broiler production.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Wenhuan Chang
- Key Laboratory for Feed Biotechnology of the Ministry of Agriculture and Rural Affairs, Institute of Feed Research, Chinese Academy of Agricultural Sciences, Beijing, China
| | - Guohua Liu
- Key Laboratory for Feed Biotechnology of the Ministry of Agriculture and Rural Affairs, Institute of Feed Research, Chinese Academy of Agricultural Sciences, Beijing, China
| |
Collapse
|
4
|
Roberts LB, Neves JF, Lee DCH, Valpione S, Tachó-Piñot R, Howard JK, Hepworth MR, Lord GM. MicroRNA-142 regulates gut associated lymphoid tissues and group 3 innate lymphoid cells. Mucosal Immunol 2025; 18:39-52. [PMID: 39245145 PMCID: PMC11835792 DOI: 10.1016/j.mucimm.2024.09.001] [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: 09/18/2023] [Revised: 08/26/2024] [Accepted: 09/02/2024] [Indexed: 09/10/2024]
Abstract
The transcriptomic signatures that shape responses of innate lymphoid cells (ILCs) have been well characterised, however post-transcriptional mechanisms which regulate their development and activity remain poorly understood. We demonstrate that ILC groups of the intestinal lamina propria express mature forms of microRNA-142 (miR-142), an evolutionarily conserved microRNA family with several non-redundant regulatory roles within the immune system. Germline Mir142 deletion alters intestinal ILC compositions, resulting in the absence of T-bet+ populations and significant defects in the cellularity and phenotypes of ILC3 subsets including CCR6+ LTi-like ILC3s. These effects were associated with decreased pathology in an innate-immune cell driven model of colitis. Furthermore, Mir142-/- mice demonstrate defective development of gut-associated lymphoid tissues, including a complete absence of mature Peyer's patches. Conditional deletion of Mir142 in ILC3s (RorcΔMir142) supported cell-intrinsic roles for these microRNAs in establishing or maintaining cellularity and functions of LTi-like ILC3s in intestinal associated tissues. RNAseq analysis revealed several target genes and biological pathways potentially regulated by miR-142 microRNAs in these cells. Finally, lack of Mir142 in ILC3 led to elevated IL-17A production. These data broaden our understanding of immune system roles of miR-142 microRNAs, identifying these molecules as critical post-transcriptional regulators of ILC3s and intestinal mucosal immunity.
Collapse
Affiliation(s)
- Luke B Roberts
- Lydia Becker Institute of Immunology and Inflammation, University of Manchester, M13 9PL, United Kingdom.
| | - Joana F Neves
- Centre for Host-Microbiome Interactions, King's College London, Great Maze Pond, London SE1 9RT, United Kingdom
| | - Dave C H Lee
- Lydia Becker Institute of Immunology and Inflammation, University of Manchester, M13 9PL, United Kingdom
| | - Sara Valpione
- The Christie NHS Foundation Trust, 550 Wilmslow Road, M20 4BX Manchester, United Kingdom; Division of Cancer Sciences, The University of Manchester, Oxford Road, M13 9PL Manchester, United Kingdom; Cancer Research UK National Biomarker Centre, Wilmslow Road, M20 4BX Manchester, United Kingdom
| | - Roser Tachó-Piñot
- Lydia Becker Institute of Immunology and Inflammation, University of Manchester, M13 9PL, United Kingdom
| | - Jane K Howard
- School of Cardiovascular and Metabolic Medicine & Sciences, King's College London, Great Maze Pond, London SE1 9RT, United Kingdom
| | - Matthew R Hepworth
- Lydia Becker Institute of Immunology and Inflammation, University of Manchester, M13 9PL, United Kingdom
| | - Graham M Lord
- Lydia Becker Institute of Immunology and Inflammation, University of Manchester, M13 9PL, United Kingdom; Centre for Gene Therapy and Regenerative Medicine, School of Basic and Medical Biosciences, Faculty of Life Sciences and Medicine, King's College London, United Kingdom.
| |
Collapse
|
5
|
He Y, Mohapatra G, Asokan S, Nobs SP, Elinav E. Microbiome modulation of antigen presentation in tolerance and inflammation. Curr Opin Immunol 2024; 91:102471. [PMID: 39277909 DOI: 10.1016/j.coi.2024.102471] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2024] [Revised: 08/24/2024] [Accepted: 08/27/2024] [Indexed: 09/17/2024]
Abstract
The microbiome regulates mammalian immune responses from early life to adulthood. Antigen presentation, orchestrating these responses, integrates commensal and pathogenic signals. However, the temporal and spatial specificity of microbiome impacts on antigen presentation and downstream tolerance versus inflammation remain incompletely understood. Herein, we review the influences of antigen presentation of microbiome-related epitopes on immunity; impacts of microbiome-based modulation of antigen presentation on innate and adaptive immune responses; and their ramifications on homeostasis and immune-related disease, ranging from auto-inflammation to tumorigenesis. We highlight mechanisms driving these influences, such as 'molecular mimicry', in which microbiome auto-antigen presentation aberrantly triggers an immune response driving autoimmunity or influences conferred by microbiome-derived metabolites on antigen-presenting cells in inflammatory bowel disease. We discuss unknowns, controversies, and challenges associated with the study of microbiome regulation of antigen presentation while demonstrating how increasing knowledge may contribute to the development of microbiome-based therapeutics modulating immune responses in a variety of clinical contexts.
Collapse
Affiliation(s)
- Yiming He
- Department of Systems Immunology, Weizmann Institute of Science, Rehovot, Israel
| | - Gayatree Mohapatra
- Department of Systems Immunology, Weizmann Institute of Science, Rehovot, Israel
| | - Sahana Asokan
- Microbiome & Cancer Division, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Samuel Philip Nobs
- Department of Systems Immunology, Weizmann Institute of Science, Rehovot, Israel.
| | - Eran Elinav
- Department of Systems Immunology, Weizmann Institute of Science, Rehovot, Israel; Microbiome & Cancer Division, German Cancer Research Center (DKFZ), Heidelberg, Germany.
| |
Collapse
|
6
|
von Voss L, Arora T, Assis J, Kuentzel KB, Arfelt KN, Nøhr MK, Grevengoed TJ, Arumugam M, Mandrup-Poulsen T, Rosenkilde MM. Sexual Dimorphism in the Immunometabolic Role of Gpr183 in Mice. J Endocr Soc 2024; 8:bvae188. [PMID: 39545055 PMCID: PMC11561910 DOI: 10.1210/jendso/bvae188] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/09/2024] [Indexed: 11/17/2024] Open
Abstract
Context Excessive eating and intake of a Western diet negatively affect the intestinal immune system, resulting in compromised glucose homeostasis and lower gut bacterial diversity. The G protein-coupled receptor GPR183 regulates immune cell migration and intestinal immune response and has been associated with tuberculosis, type 1 diabetes, and inflammatory bowel diseases. Objective We hypothesized that with these implications, GPR183 has an important immunometabolic role and investigated this using a global Gpr183 knockout mouse model. Methods Wild-type (WT) and Gpr183-deficient (Gpr183-/-) mice were fed a high-fat, high-sucrose diet (HFSD) for 15 weeks. We investigated changes in weight, body composition, fecal immunoglobulin A (IgA) levels, fecal microbiome, and glucose tolerance before and after the diet. Macrophage infiltration into visceral fat was determined by flow cytometry, and hepatic gene expression was measured. Results A sexual dimorphism was discovered, whereby female Gpr183-/- mice showed adverse metabolic outcomes compared to WT counterparts with inferior glucose tolerance, lower fecal IgA levels, and increased macrophage infiltration in visceral fat. In contrast, male Gpr183-/- mice had significantly lower fasting blood glucose after diet than male WT mice. Liver gene expression showed reduced inflammation and macrophage markers in Gpr183-/- livers, regardless of sex, while the pancreatic islet area did not differ between the groups. No conclusive differences were found after microbiome sequencing. Conclusion Gpr183 maintains metabolic homeostasis in female but not in male mice independent of diet. If confirmed in humans, future therapy targeting GPR183 should consider this sexual dimorphism.
Collapse
Affiliation(s)
- Liv von Voss
- Molecular and Translational Pharmacology, Department of Biomedical Sciences, University of Copenhagen, DK 2200 Copenhagen, Denmark
| | - Tulika Arora
- Novo Nordisk Foundation Center for Basic Metabolic Research and Medical Sciences, University of Copenhagen, DK 2200 Copenhagen, Denmark
| | - Juliana Assis
- Novo Nordisk Foundation Center for Basic Metabolic Research and Medical Sciences, University of Copenhagen, DK 2200 Copenhagen, Denmark
- National Bioinformatics Infrastructure Sweden, Science for Life Laboratory, Department of Immunotechnology, Lund University, SE 223 63 Lund, Sweden
| | - Katharina B Kuentzel
- Molecular and Translational Pharmacology, Department of Biomedical Sciences, University of Copenhagen, DK 2200 Copenhagen, Denmark
| | - Kristine N Arfelt
- Molecular and Translational Pharmacology, Department of Biomedical Sciences, University of Copenhagen, DK 2200 Copenhagen, Denmark
| | - Mark K Nøhr
- Molecular and Translational Pharmacology, Department of Biomedical Sciences, University of Copenhagen, DK 2200 Copenhagen, Denmark
| | - Trisha J Grevengoed
- Molecular and Translational Pharmacology, Department of Biomedical Sciences, University of Copenhagen, DK 2200 Copenhagen, Denmark
| | - Manimozhiyan Arumugam
- Novo Nordisk Foundation Center for Basic Metabolic Research and Medical Sciences, University of Copenhagen, DK 2200 Copenhagen, Denmark
| | - Thomas Mandrup-Poulsen
- Molecular and Translational Pharmacology, Department of Biomedical Sciences, University of Copenhagen, DK 2200 Copenhagen, Denmark
| | - Mette M Rosenkilde
- Molecular and Translational Pharmacology, Department of Biomedical Sciences, University of Copenhagen, DK 2200 Copenhagen, Denmark
| |
Collapse
|
7
|
Heidari M, Maleki Vareki S, Yaghobi R, Karimi MH. Microbiota activation and regulation of adaptive immunity. Front Immunol 2024; 15:1429436. [PMID: 39445008 PMCID: PMC11496076 DOI: 10.3389/fimmu.2024.1429436] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2024] [Accepted: 08/30/2024] [Indexed: 10/25/2024] Open
Abstract
In the mucosa, T cells and B cells of the immune system are essential for maintaining immune homeostasis by suppressing reactions to harmless antigens and upholding the integrity of intestinal mucosal barrier functions. Host immunity and homeostasis are regulated by metabolites produced by the gut microbiota, which has developed through the long-term coevolution of the host and the gut biome. This is achieved by the immunological system's tolerance for symbiote microbiota, and its ability to generate a proinflammatory response against invasive organisms. The imbalance of the intestinal immune system with commensal organisms is causing a disturbance in the homeostasis of the gut microbiome. The lack of balance results in microbiota dysbiosis, the weakened integrity of the gut barrier, and the development of inflammatory immune reactions toward symbiotic organisms. Researchers may uncover potential therapeutic targets for preventing or regulating inflammatory diseases by understanding the interactions between adaptive immunity and the microbiota. This discussion will explore the connection between adaptive immunity and microbiota.
Collapse
Affiliation(s)
- Mozhdeh Heidari
- Transplant Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Saman Maleki Vareki
- Department of Oncology, Western University, London, ON, Canada
- Department of Pathology and Laboratory Medicine, Western University, London, ON, Canada
- London Regional Cancer Program, Lawson Health Research Institute, London, ON, Canada
| | - Ramin Yaghobi
- Transplant Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | | |
Collapse
|
8
|
Ilangovan J, Neves JF, Santos AF. Innate lymphoid cells in immunoglobulin E-mediated food allergy. Curr Opin Allergy Clin Immunol 2024; 24:419-425. [PMID: 39132724 PMCID: PMC11356679 DOI: 10.1097/aci.0000000000001018] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/13/2024]
Abstract
PURPOSE OF REVIEW Recognition of the importance of innate lymphoid cells (ILCs) in the immune mechanisms of food allergy has grown in recent years. This review summarizes recent findings of ILCs in immunoglobulin E (IgE)-mediated food allergy. New research on ILCs in the context of the microbiome and other atopic diseases are also considered with respect to how they can inform understanding of the role of ILCs in food allergy. RECENT FINDINGS ILCs can mediate allergic and tolerogenic responses through multiple pathways. A novel subset of interleukin (IL)-10 producing ILC2s are associated with tolerance following immunotherapy to grass pollen, house dust mite allergy and lipid transfer protein allergy. ILC2s can drive food allergen-specific T cell responses in an antigen-specific manner. A memory subset of ILC2s has been identified through studies of other atopic diseases and is associated with effectiveness of response to therapy. SUMMARY The role of ILCs in food allergy and oral tolerance is relatively understudied compared to other diseases. ILCs can modulate immune responses through several mechanisms, and it is likely that these are of importance in the context of food allergy. Better understanding of theses pathways may help to answer fundamental questions regarding the development of food allergy and lead to novel therapeutic targets and treatment.
Collapse
Affiliation(s)
- Janarthanan Ilangovan
- Peter Gorer Department of Immunobiology, School of Immunology and Microbial Sciences, Faculty of Life Sciences and Medicine
- Centre for Host Microbiome Interactions
| | | | - Alexandra F. Santos
- Peter Gorer Department of Immunobiology, School of Immunology and Microbial Sciences, Faculty of Life Sciences and Medicine
- Department of Women and Children's Health (Paediatric Allergy), School of Life Course Sciences, Faculty of Life Sciences and Medicine, King's College London
- Children's Allergy Service, Guy's and St Thomas’ Hospital, London, UK
| |
Collapse
|
9
|
Sasaki T, Ota Y, Takikawa Y, Terrooatea T, Kanaya T, Takahashi M, Taguchi-Atarashi N, Tachibana N, Yabukami H, Surh CD, Minoda A, Kim KS, Ohno H. Food antigens suppress small intestinal tumorigenesis. Front Immunol 2024; 15:1373766. [PMID: 39359724 PMCID: PMC11445177 DOI: 10.3389/fimmu.2024.1373766] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2024] [Accepted: 07/30/2024] [Indexed: 10/04/2024] Open
Abstract
Food components suppressing small intestinal tumorigenesis are not well-defined partly because of the rarity of this tumor type compared to colorectal tumors. Using Apcmin/+ mice, a mouse model for intestinal tumorigenesis, and antigen-free diet, we report here that food antigens serve this function in the small intestine. By depleting Peyer's patches (PPs), immune inductive sites in the small intestine, we found that PPs have a role in the suppression of small intestinal tumors and are important for the induction of small intestinal T cells by food antigens. On the follicle-associated epithelium (FAE) of PPs, microfold (M) cells pass food antigens from lumen to the dendritic cells to induce T cells. Single-cell RNA-seq (scRNA-seq) analysis of immune cells in PPs revealed a significant impact of food antigens on the induction of the PP T cells and the antigen presentation capacity of dendritic cells. These data demonstrate the role of food antigens in the suppression of small intestinal tumorigenesis by PP-mediated immune cell induction.
Collapse
Affiliation(s)
- Takaharu Sasaki
- Laboratory for Intestinal Ecosystem, RIKEN Center for Integrative Medical Sciences, Yokohama, Japan
| | - Yuna Ota
- Laboratory for Intestinal Ecosystem, RIKEN Center for Integrative Medical Sciences, Yokohama, Japan
- Graduate School of Medical Life Science, Yokohama City University, Yokohama, Japan
| | - Yui Takikawa
- Laboratory for Intestinal Ecosystem, RIKEN Center for Integrative Medical Sciences, Yokohama, Japan
- Graduate School of Medical Life Science, Yokohama City University, Yokohama, Japan
| | - Tommy Terrooatea
- Laboratory for Cellular Epigenomics, RIKEN Center for Integrative Medical Sciences, Yokohama, Japan
| | - Takashi Kanaya
- Laboratory for Intestinal Ecosystem, RIKEN Center for Integrative Medical Sciences, Yokohama, Japan
- Graduate School of Medical Life Science, Yokohama City University, Yokohama, Japan
| | - Masumi Takahashi
- Laboratory for Intestinal Ecosystem, RIKEN Center for Integrative Medical Sciences, Yokohama, Japan
| | - Naoko Taguchi-Atarashi
- Laboratory for Intestinal Ecosystem, RIKEN Center for Integrative Medical Sciences, Yokohama, Japan
| | - Naoko Tachibana
- Laboratory for Intestinal Ecosystem, RIKEN Center for Integrative Medical Sciences, Yokohama, Japan
| | - Haruka Yabukami
- Laboratory for Cellular Epigenomics, RIKEN Center for Integrative Medical Sciences, Yokohama, Japan
| | - Charles D. Surh
- Department of Life Sciences, Pohang University of Science and Technology (POSTECH), Pohang, Republic of Korea
| | - Aki Minoda
- Laboratory for Cellular Epigenomics, RIKEN Center for Integrative Medical Sciences, Yokohama, Japan
- Department of Cell Biology, Faculty of Science, Radboud Institute for Molecular Life Sciences, Radboud University, Nijmegen, Netherlands
| | - Kwang Soon Kim
- Department of Life Sciences, Pohang University of Science and Technology (POSTECH), Pohang, Republic of Korea
| | - Hiroshi Ohno
- Laboratory for Intestinal Ecosystem, RIKEN Center for Integrative Medical Sciences, Yokohama, Japan
- Graduate School of Medical Life Science, Yokohama City University, Yokohama, Japan
- Laboratory for Immune Regulation, Graduate School of Medical and Pharmaceutical Sciences, Chiba University, Chiba, Japan
| |
Collapse
|
10
|
Carreto-Binaghi LE, Sztein MB, Booth JS. Role of cellular effectors in the induction and maintenance of IgA responses leading to protective immunity against enteric bacterial pathogens. Front Immunol 2024; 15:1446072. [PMID: 39324143 PMCID: PMC11422102 DOI: 10.3389/fimmu.2024.1446072] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2024] [Accepted: 08/26/2024] [Indexed: 09/27/2024] Open
Abstract
The mucosal immune system is a critical first line of defense to infectious diseases, as many pathogens enter the body through mucosal surfaces, disrupting the balanced interactions between mucosal cells, secretory molecules, and microbiota in this challenging microenvironment. The mucosal immune system comprises of a complex and integrated network that includes the gut-associated lymphoid tissues (GALT). One of its primary responses to microbes is the secretion of IgA, whose role in the mucosa is vital for preventing pathogen colonization, invasion and spread. The mechanisms involved in these key responses include neutralization of pathogens, immune exclusion, immune modulation, and cross-protection. The generation and maintenance of high affinity IgA responses require a delicate balance of multiple components, including B and T cell interactions, innate cells, the cytokine milieu (e.g., IL-21, IL-10, TGF-β), and other factors essential for intestinal homeostasis, including the gut microbiota. In this review, we will discuss the main cellular components (e.g., T cells, innate lymphoid cells, dendritic cells) in the gut microenvironment as mediators of important effector responses and as critical players in supporting B cells in eliciting and maintaining IgA production, particularly in the context of enteric infections and vaccination in humans. Understanding the mechanisms of humoral and cellular components in protection could guide and accelerate the development of more effective mucosal vaccines and therapeutic interventions to efficiently combat mucosal infections.
Collapse
Affiliation(s)
- Laura E. Carreto-Binaghi
- Center for Vaccine Development and Global Health, University of Maryland School of Medicine, Baltimore, MD, United States
- Department of Pediatrics, University of Maryland School of Medicine, Baltimore, MD, United States
- Laboratorio de Inmunobiologia de la Tuberculosis, Instituto Nacional de Enfermedades Respiratorias Ismael Cosio Villegas, Mexico City, Mexico
| | - Marcelo B. Sztein
- Center for Vaccine Development and Global Health, University of Maryland School of Medicine, Baltimore, MD, United States
- Department of Pediatrics, University of Maryland School of Medicine, Baltimore, MD, United States
- Department of Medicine, University of Maryland School of Medicine, Baltimore, MD, United States
- Tumor Immunology and Immunotherapy Program, University of Maryland Marlene and Stewart Greenebaum Comprehensive Cancer Center, Baltimore, MD, United States
| | - Jayaum S. Booth
- Center for Vaccine Development and Global Health, University of Maryland School of Medicine, Baltimore, MD, United States
- Department of Pediatrics, University of Maryland School of Medicine, Baltimore, MD, United States
| |
Collapse
|
11
|
Wang W, Li N, Guo X. The crosstalk between ILC3s and adaptive immunity in diseases. FEBS J 2024; 291:3965-3977. [PMID: 37994218 DOI: 10.1111/febs.17014] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2023] [Revised: 09/26/2023] [Accepted: 11/21/2023] [Indexed: 11/24/2023]
Abstract
RORγt+ group 3 innate lymphoid cells (ILC3s), the innate counterpart of Th17 cells, are enriched in the mucosal area and lymphoid tissues. ILC3s interact with a variety of cells through their effector molecules and play an important role in the host defense against a spectrum of infections. Recent studies suggest that the extensive crosstalk between ILC3s and adaptive immune cells, especially T cells, is essential for maintaining tissue homeostasis. Here we discuss recent advances in the crosstalk between ILC3s and adaptive immune responses in multiple tissues and diseases. Understanding how ILC3s engage with adaptive immune cells will enhance our comprehension of diseases and facilitate the identification of novel therapeutic targets.
Collapse
Affiliation(s)
- Wenyan Wang
- Institute for Immunology, Tsinghua University, Beijing, China
- Department of Basic Medical Sciences, School of Medicine, Tsinghua University, Beijing, China
- Beijing Key Lab for Immunological Research on Chronic Diseases, Tsinghua University, Beijing, China
| | - Na Li
- Institute for Immunology, Tsinghua University, Beijing, China
- Department of Basic Medical Sciences, School of Medicine, Tsinghua University, Beijing, China
- Beijing Key Lab for Immunological Research on Chronic Diseases, Tsinghua University, Beijing, China
| | - Xiaohuan Guo
- Institute for Immunology, Tsinghua University, Beijing, China
- Department of Basic Medical Sciences, School of Medicine, Tsinghua University, Beijing, China
- Beijing Key Lab for Immunological Research on Chronic Diseases, Tsinghua University, Beijing, China
| |
Collapse
|
12
|
Huang C, Zhu W, Li Q, Lei Y, Chen X, Liu S, Chen D, Zhong L, Gao F, Fu S, He D, Li J, Xu H. Antibody Fc-receptor FcεR1γ stabilizes cell surface receptors in group 3 innate lymphoid cells and promotes anti-infection immunity. Nat Commun 2024; 15:5981. [PMID: 39013884 PMCID: PMC11252441 DOI: 10.1038/s41467-024-50266-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2023] [Accepted: 07/03/2024] [Indexed: 07/18/2024] Open
Abstract
Group 3 innate lymphoid cells (ILC3) are crucial for maintaining mucosal homeostasis and regulating inflammatory diseases, but the molecular mechanisms governing their phenotype and function are not fully understood. Here, we show that ILC3s highly express Fcer1g gene, which encodes the antibody Fc-receptor common gamma chain, FcεR1γ. Genetic perturbation of FcεR1γ leads to the absence of critical cell membrane receptors NKp46 and CD16 in ILC3s. Alanine scanning mutagenesis identifies two residues in FcεR1γ that stabilize its binding partners. FcεR1γ expression in ILC3s is essential for effective protective immunity against bacterial and fungal infections. Mechanistically, FcεR1γ influences the transcriptional state and proinflammatory cytokine production of ILC3s, relying on the CD16-FcεR1γ signaling pathway. In summary, our findings highlight the significance of FcεR1γ as an adapter protein that stabilizes cell membrane partners in ILC3s and promotes anti-infection immunity.
Collapse
Affiliation(s)
- Chao Huang
- Key Laboratory of Systems Health Science of Zhejiang Province, School of Life Science, Hangzhou Institute for Advanced Study, University of Chinese Academy of Sciences, Hangzhou, China.
- Laboratory of Systems Immunology, School of Medicine, Westlake University, Hangzhou, Zhejiang, China.
- Key Laboratory of Growth Regulation and Translational Research of Zhejiang Province, School of Life Sciences, Westlake University, Hangzhou, Zhejiang, China.
- Westlake Laboratory of Life Sciences and Biomedicine, Hangzhou, Zhejiang, China.
| | - Wenting Zhu
- Laboratory of Systems Immunology, School of Medicine, Westlake University, Hangzhou, Zhejiang, China
- Key Laboratory of Growth Regulation and Translational Research of Zhejiang Province, School of Life Sciences, Westlake University, Hangzhou, Zhejiang, China
- Westlake Laboratory of Life Sciences and Biomedicine, Hangzhou, Zhejiang, China
- School of Pharmaceutical Science and Technology, Hangzhou Institute for Advanced Study, University of Chinese Academy of Sciences, Hangzhou, China
| | - Qing Li
- Key Laboratory of Multi-Cell Systems, Shanghai Key Laboratory of Molecular Andrology, CAS Center for Excellence in Molecular Cell Science, Shanghai Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai, China
| | - Yuchen Lei
- Key Laboratory of Systems Health Science of Zhejiang Province, School of Life Science, Hangzhou Institute for Advanced Study, University of Chinese Academy of Sciences, Hangzhou, China
| | - Xi Chen
- Laboratory of Systems Immunology, School of Medicine, Westlake University, Hangzhou, Zhejiang, China
- Key Laboratory of Growth Regulation and Translational Research of Zhejiang Province, School of Life Sciences, Westlake University, Hangzhou, Zhejiang, China
- Westlake Laboratory of Life Sciences and Biomedicine, Hangzhou, Zhejiang, China
| | - Shaorui Liu
- Laboratory of Systems Immunology, School of Medicine, Westlake University, Hangzhou, Zhejiang, China
- Key Laboratory of Growth Regulation and Translational Research of Zhejiang Province, School of Life Sciences, Westlake University, Hangzhou, Zhejiang, China
- Westlake Laboratory of Life Sciences and Biomedicine, Hangzhou, Zhejiang, China
| | - Dianyu Chen
- Laboratory of Systems Immunology, School of Medicine, Westlake University, Hangzhou, Zhejiang, China
- Key Laboratory of Growth Regulation and Translational Research of Zhejiang Province, School of Life Sciences, Westlake University, Hangzhou, Zhejiang, China
- Westlake Laboratory of Life Sciences and Biomedicine, Hangzhou, Zhejiang, China
| | - Lijian Zhong
- Key Laboratory of Systems Health Science of Zhejiang Province, School of Life Science, Hangzhou Institute for Advanced Study, University of Chinese Academy of Sciences, Hangzhou, China
| | - Feng Gao
- Laboratory of Systems Immunology, School of Medicine, Westlake University, Hangzhou, Zhejiang, China
- Key Laboratory of Growth Regulation and Translational Research of Zhejiang Province, School of Life Sciences, Westlake University, Hangzhou, Zhejiang, China
- Westlake Laboratory of Life Sciences and Biomedicine, Hangzhou, Zhejiang, China
| | - Shujie Fu
- Laboratory of Systems Immunology, School of Medicine, Westlake University, Hangzhou, Zhejiang, China
- Key Laboratory of Growth Regulation and Translational Research of Zhejiang Province, School of Life Sciences, Westlake University, Hangzhou, Zhejiang, China
| | - Danyang He
- Laboratory of Systems Immunology, School of Medicine, Westlake University, Hangzhou, Zhejiang, China
- Key Laboratory of Growth Regulation and Translational Research of Zhejiang Province, School of Life Sciences, Westlake University, Hangzhou, Zhejiang, China
| | - Jinsong Li
- Key Laboratory of Systems Health Science of Zhejiang Province, School of Life Science, Hangzhou Institute for Advanced Study, University of Chinese Academy of Sciences, Hangzhou, China
- Key Laboratory of Multi-Cell Systems, Shanghai Key Laboratory of Molecular Andrology, CAS Center for Excellence in Molecular Cell Science, Shanghai Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai, China
| | - Heping Xu
- Laboratory of Systems Immunology, School of Medicine, Westlake University, Hangzhou, Zhejiang, China.
- Key Laboratory of Growth Regulation and Translational Research of Zhejiang Province, School of Life Sciences, Westlake University, Hangzhou, Zhejiang, China.
- Westlake Laboratory of Life Sciences and Biomedicine, Hangzhou, Zhejiang, China.
| |
Collapse
|
13
|
Wu S, Yin Y, Du L. The bidirectional relationship of depression and disturbances in B cell homeostasis: Double trouble. Prog Neuropsychopharmacol Biol Psychiatry 2024; 132:110993. [PMID: 38490433 DOI: 10.1016/j.pnpbp.2024.110993] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/09/2023] [Revised: 03/08/2024] [Accepted: 03/12/2024] [Indexed: 03/17/2024]
Abstract
Major depressive disorder (MDD) is a recurrent, persistent, and debilitating neuropsychiatric syndrome with an increasing morbidity and mortality, representing the leading cause of disability worldwide. The dysregulation of immune systems (including innate and adaptive immune systems) has been identified as one of the key contributing factors in the progression of MDD. As the main force of the humoral immunity, B cells have an essential role in the defense against infections, antitumor immunity and autoimmune diseases. Several recent studies have suggested an intriguing connection between disturbances in B cell homeostasis and the pathogenesis of MDD, however, the B-cell-dependent mechanism of MDD remains largely unexplored compared to other immune cells. In this review, we provide an overview of how B cell abnormality regulates the progression of MMD and the potential consequence of the disruption of B cell homeostasis in patients with MDD. Abnormalities of B-cell homeostasis not only promote susceptibility to MDD, but also lead to an increased risk of developing infection, malignancy and autoimmune diseases in patients with MDD. A better understanding of the contribution of B cells underlying MDD would provide opportunities for identification of more targeted treatment approaches and might provide an overall therapeutic benefit to improve the long-term outcomes of patients with MDD.
Collapse
Affiliation(s)
- Shusheng Wu
- Department of Neurology, Affiliated Hospital of Yangzhou University, Jiangsu, China
| | - Yuye Yin
- College of Bioscience and Biotechnology, Yangzhou University, Yangzhou, Jiangsu, China
| | - Longfei Du
- Department of Laboratory Medicine, Affiliated Hospital of Yangzhou University, Yangzhou, Jiangsu, China.
| |
Collapse
|
14
|
Horn V, Sonnenberg GF. Group 3 innate lymphoid cells in intestinal health and disease. Nat Rev Gastroenterol Hepatol 2024; 21:428-443. [PMID: 38467885 PMCID: PMC11144103 DOI: 10.1038/s41575-024-00906-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 02/05/2024] [Indexed: 03/13/2024]
Abstract
The gastrointestinal tract is an immunologically rich organ, containing complex cell networks and dense lymphoid structures that safeguard this large absorptive barrier from pathogens, contribute to tissue physiology and support mucosal healing. Simultaneously, the immune system must remain tolerant to innocuous dietary antigens and trillions of normally beneficial microorganisms colonizing the intestine. Indeed, a dysfunctional immune response in the intestine underlies the pathogenesis of numerous local and systemic diseases, including inflammatory bowel disease, food allergy, chronic enteric infections or cancers. Here, we discuss group 3 innate lymphoid cells (ILC3s), which have emerged as orchestrators of tissue physiology, immunity, inflammation, tolerance and malignancy in the gastrointestinal tract. ILC3s are abundant in the developing and healthy intestine but their numbers or function are altered during chronic disease and cancer. The latest studies provide new insights into the mechanisms by which ILC3s fundamentally shape intestinal homeostasis or disease pathophysiology, and often this functional dichotomy depends on context and complex interactions with other cell types or microorganisms. Finally, we consider how this knowledge could be harnessed to improve current treatments or provoke new opportunities for therapeutic intervention to promote gut health.
Collapse
Affiliation(s)
- Veronika Horn
- Jill Roberts Institute for Research in Inflammatory Bowel Disease, Weill Cornell Medicine, Cornell University, New York, NY, USA
- Joan and Sanford I. Weill Department of Medicine, Division of Gastroenterology & Hepatology, Weill Cornell Medicine, Cornell University, New York, NY, USA
- Department of Microbiology & Immunology, Weill Cornell Medicine, Cornell University, New York, NY, USA
| | - Gregory F Sonnenberg
- Jill Roberts Institute for Research in Inflammatory Bowel Disease, Weill Cornell Medicine, Cornell University, New York, NY, USA.
- Joan and Sanford I. Weill Department of Medicine, Division of Gastroenterology & Hepatology, Weill Cornell Medicine, Cornell University, New York, NY, USA.
- Department of Microbiology & Immunology, Weill Cornell Medicine, Cornell University, New York, NY, USA.
| |
Collapse
|
15
|
Tyler CJ, Hoti I, Griffiths DD, Cuff SM, Andrews R, Keisker M, Ahmed R, Hansen HP, Lindsay JO, Stagg AJ, Moser B, McCarthy NE, Eberl M. IL-21 conditions antigen-presenting human γδ T-cells to promote IL-10 expression in naïve and memory CD4 + T-cells. DISCOVERY IMMUNOLOGY 2024; 3:kyae008. [PMID: 38903247 PMCID: PMC11187773 DOI: 10.1093/discim/kyae008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 03/02/2024] [Revised: 03/30/2024] [Accepted: 05/09/2024] [Indexed: 06/22/2024]
Abstract
Direct interaction between T-cells exerts a major influence on tissue immunity and inflammation across multiple body sites including the human gut, which is highly enriched in 'unconventional' lymphocytes such as γδ T-cells. We previously reported that microbial activation of human Vγ9/Vδ2+ γδ T-cells in the presence of the mucosal damage-associated cytokine IL-15 confers the ability to promote epithelial barrier defence, specifically via induction of IL-22 expression in conventional CD4+ T-cells. In the current report, we assessed whether other cytokines enriched in the gut milieu also functionally influence microbe-responsive Vγ9/Vδ2 T-cells. When cultured in the presence of IL-21, Vγ9/Vδ2 T-cells acquired the ability to induce expression of the immunoregulatory cytokine IL-10 in both naïve and memory CD4+ T-cells, at levels surpassing those induced by monocytes or monocyte-derived DCs. These findings identify an unexpected influence of IL-21 on Vγ9/Vδ2 T-cell modulation of CD4+ T-cell responses. Further analyses suggested a possible role for CD30L and/or CD40L reverse signalling in mediating IL-10 induction by IL-21 conditioned Vγ9/Vδ2 T-cells. Our findings indicate that the local microenvironment exerts a profound influence on Vγ9/Vδ2 T-cell responses to microbial challenge, leading to induction of distinct functional profiles among CD4+ T-cells that may influence inflammatory events at mucosal surfaces. Targeting these novel pathways may offer therapeutic benefit in disorders such as inflammatory bowel disease.
Collapse
Affiliation(s)
- Christopher J Tyler
- Division of Infection and Immunity, School of Medicine, Cardiff University, Cardiff, UK
| | - Inva Hoti
- Centre for Immunobiology, The Blizard Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, UK
| | - Daniel D Griffiths
- Division of Infection and Immunity, School of Medicine, Cardiff University, Cardiff, UK
| | - Simone M Cuff
- Division of Infection and Immunity, School of Medicine, Cardiff University, Cardiff, UK
| | - Robert Andrews
- Systems Immunity Research Institute, Cardiff University, Cardiff, UK
| | - Maximilian Keisker
- Division of Infection and Immunity, School of Medicine, Cardiff University, Cardiff, UK
| | - Raya Ahmed
- Division of Infection and Immunity, School of Medicine, Cardiff University, Cardiff, UK
| | - Hinrich P Hansen
- Department of Internal Medicine I, University of Cologne, Cologne, Germany
| | - James O Lindsay
- Centre for Immunobiology, The Blizard Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, UK
- Department of Gastroenterology, The Royal London Hospital, Barts Health NHS Trust, London, UK
| | - Andrew J Stagg
- Centre for Immunobiology, The Blizard Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, UK
| | - Bernhard Moser
- Division of Infection and Immunity, School of Medicine, Cardiff University, Cardiff, UK
- Systems Immunity Research Institute, Cardiff University, Cardiff, UK
| | - Neil E McCarthy
- Centre for Immunobiology, The Blizard Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, UK
| | - Matthias Eberl
- Division of Infection and Immunity, School of Medicine, Cardiff University, Cardiff, UK
- Systems Immunity Research Institute, Cardiff University, Cardiff, UK
| |
Collapse
|
16
|
Das S, Chauhan KS, Ahmed M, Akter S, Lu L, Colonna M, Khader SA. Lung type 3 innate lymphoid cells respond early following Mycobacterium tuberculosis infection. mBio 2024; 15:e0329923. [PMID: 38407132 PMCID: PMC11005430 DOI: 10.1128/mbio.03299-23] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2023] [Accepted: 01/17/2024] [Indexed: 02/27/2024] Open
Abstract
Tuberculosis is the leading cause of death due to an infectious disease worldwide. Innate lymphoid type 3 cells (ILC3s) mediate early protection during Mycobacterium tuberculosis (Mtb) infection. However, the early signaling mechanisms that govern ILC3 activation or recruitment within the lung during Mtb infection are unclear. scRNA-seq analysis of Mtb-infected mouse lung innate lymphoid cells (ILCs) has revealed the presence of different clusters of ILC populations, suggesting heterogeneity. Using mouse models, we show that during Mtb infection, interleukin-1 receptor (IL-1R) signaling on epithelial cells drives ILC3 expansion and regulates ILC3 accumulation in the lung. Furthermore, our data revealed that C-X-C motif chemokine receptor 5 (CXCR5) signaling plays a crucial role in ILC3 recruitment from periphery during Mtb infection. Our study thus establishes the early responses that drive ILC3 accumulation during Mtb infection and points to ILC3s as a potential vaccine target. IMPORTANCE Tuberculosis is a leading cause of death due to a single infectious agent accounting for 1.6 million deaths each year. In our study, we determined the role of type 3 innate lymphoid cells in early immune events necessary for achieving protection during Mtb infection. Our study reveals distinct clusters of ILC2, ILC3, and ILC3/ILC1-like cells in Mtb infection. Moreover, our study reveal that IL-1R signaling on lung type 2 epithelial cells plays a key role in lung ILC3 accumulation during Mtb infection. CXCR5 on ILC3s is involved in ILC3 homing from periphery during Mtb infection. Thus, our study provides novel insights into the early immune mechanisms governed by innate lymphoid cells that can be targeted for potential vaccine-induced protection.
Collapse
Affiliation(s)
- Shibali Das
- Department of Molecular Microbiology, Washington University in St. Louis, St. Louis, Missouri, USA
| | - Kuldeep Singh Chauhan
- Department of Molecular Microbiology, Washington University in St. Louis, St. Louis, Missouri, USA
- Department of Microbiology, University of Chicago, Chicago, Illinois, USA
| | - Mushtaq Ahmed
- Department of Molecular Microbiology, Washington University in St. Louis, St. Louis, Missouri, USA
- Department of Microbiology, University of Chicago, Chicago, Illinois, USA
| | - Sadia Akter
- Department of Molecular Microbiology, Washington University in St. Louis, St. Louis, Missouri, USA
- Department of Microbiology, University of Chicago, Chicago, Illinois, USA
| | - Lan Lu
- Department of Molecular Microbiology, Washington University in St. Louis, St. Louis, Missouri, USA
| | - Marco Colonna
- Department of Pathology and Immunology, Division of Immunobiology, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Shabaana A. Khader
- Department of Molecular Microbiology, Washington University in St. Louis, St. Louis, Missouri, USA
- Department of Microbiology, University of Chicago, Chicago, Illinois, USA
| |
Collapse
|
17
|
Sepúlveda-Alfaro J, Catalán EA, Vallejos OP, Ramos-Tapia I, Madrid-Muñoz C, Mendoza-León MJ, Suazo ID, Rivera-Asin E, Silva PH, Alvarez-Mardones O, Castillo-Godoy DP, Riedel CA, Schinnerling K, Ugalde JA, Soto JA, Bueno SM, Kalergis AM, Melo-Gonzalez F. Human metapneumovirus respiratory infection affects both innate and adaptive intestinal immunity. Front Immunol 2024; 15:1330209. [PMID: 38404579 PMCID: PMC10884822 DOI: 10.3389/fimmu.2024.1330209] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2023] [Accepted: 01/15/2024] [Indexed: 02/27/2024] Open
Abstract
Introduction Respiratory infections are one of the leading causes of morbidity and mortality worldwide, mainly in children, immunocompromised people, and the elderly. Several respiratory viruses can induce intestinal inflammation and alterations in intestinal microbiota composition. Human metapneumovirus (HMPV) is one of the major respiratory viruses contributing to infant mortality in children under 5 years of age worldwide, and the effect of this infection at the gut level has not been studied. Methods Here, we evaluated the distal effects of HMPV infection on intestinal microbiota and inflammation in a murine model, analyzing several post-infection times (days 1, 3, and 5). Six to eight-week-old C57BL/6 mice were infected intranasally with HMPV, and mice inoculated with a non-infectious supernatant (Mock) were used as a control group. Results We did not detect HMPV viral load in the intestine, but we observed significant changes in the transcription of IFN-γ in the colon, analyzed by qPCR, at day 1 post-infection as compared to the control group. Furthermore, we analyzed the frequencies of different innate and adaptive immune cells in the colonic lamina propria, using flow cytometry. The frequency of monocyte populations was altered in the colon of HMPV -infected mice at days 1 and 3, with no significant difference from control mice at day 5 post-infection. Moreover, colonic CD8+ T cells and memory precursor effector CD8+ T cells were significantly increased in HMPV-infected mice at day 5, suggesting that HMPV may also alter intestinal adaptive immunity. Additionally, we did not find alterations in antimicrobial peptide expression, the frequency of colonic IgA+ plasma cells, and levels of fecal IgA. Some minor alterations in the fecal microbiota composition of HMPV -infected mice were detected using 16s rRNA sequencing. However, no significant differences were found in β-diversity and relative abundance at the genus level. Discussion To our knowledge, this is the first report describing the alterations in intestinal immunity following respiratory infection with HMPV infection. These effects do not seem to be mediated by direct viral infection in the intestinal tract. Our results indicate that HMPV can affect colonic innate and adaptive immunity but does not significantly alter the microbiota composition, and further research is required to understand the mechanisms inducing these distal effects in the intestine.
Collapse
Affiliation(s)
- Javiera Sepúlveda-Alfaro
- Millennium Institute on Immunology and Immunotherapy, Departamento de Genética Molecular y Microbiología, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Eduardo A. Catalán
- Millennium Institute on Immunology and Immunotherapy, Departamento de Genética Molecular y Microbiología, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile
- Millennium Institute on Immunology and Immunotherapy, Departamento de Ciencias Biológicas, Facultad de Ciencias de la Vida, Universidad Andrés Bello, Santiago, Chile
| | - Omar P. Vallejos
- Millennium Institute on Immunology and Immunotherapy, Departamento de Genética Molecular y Microbiología, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Ignacio Ramos-Tapia
- Center for Bioinformatics and Integrative Biology, Facultad de Ciencias de la Vida, Universidad Andrés Bello, Santiago, Chile
| | | | - María J. Mendoza-León
- Millennium Institute on Immunology and Immunotherapy, Departamento de Ciencias Biológicas, Facultad de Ciencias de la Vida, Universidad Andrés Bello, Santiago, Chile
| | - Isidora D. Suazo
- Millennium Institute on Immunology and Immunotherapy, Departamento de Genética Molecular y Microbiología, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Elizabeth Rivera-Asin
- Millennium Institute on Immunology and Immunotherapy, Departamento de Ciencias Biológicas, Facultad de Ciencias de la Vida, Universidad Andrés Bello, Santiago, Chile
| | - Pedro H. Silva
- Millennium Institute on Immunology and Immunotherapy, Departamento de Genética Molecular y Microbiología, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Oscar Alvarez-Mardones
- Millennium Institute on Immunology and Immunotherapy, Departamento de Ciencias Biológicas, Facultad de Ciencias de la Vida, Universidad Andrés Bello, Santiago, Chile
| | | | - Claudia A. Riedel
- Millennium Institute on Immunology and Immunotherapy, Departamento de Ciencias Biológicas, Facultad de Ciencias de la Vida, Universidad Andrés Bello, Santiago, Chile
| | | | - Juan A. Ugalde
- Center for Bioinformatics and Integrative Biology, Facultad de Ciencias de la Vida, Universidad Andrés Bello, Santiago, Chile
| | - Jorge A. Soto
- Millennium Institute on Immunology and Immunotherapy, Departamento de Ciencias Biológicas, Facultad de Ciencias de la Vida, Universidad Andrés Bello, Santiago, Chile
| | - Susan M. Bueno
- Millennium Institute on Immunology and Immunotherapy, Departamento de Genética Molecular y Microbiología, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Alexis M. Kalergis
- Millennium Institute on Immunology and Immunotherapy, Departamento de Genética Molecular y Microbiología, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile
- Departamento de Endocrinología, Facultad de Medicina, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Felipe Melo-Gonzalez
- Millennium Institute on Immunology and Immunotherapy, Departamento de Ciencias Biológicas, Facultad de Ciencias de la Vida, Universidad Andrés Bello, Santiago, Chile
| |
Collapse
|
18
|
Kurihara S, Suzuki K, Yokota M, Ito T, Hayashi Y, Kikuchi R, Kageyama T, Meguro K, Tanaka S, Iwata A, Goto Y, Suto A, Nakajima H. Eosinophils Contribute to Oral Tolerance via Induction of RORγt-Positive Antigen-Presenting Cells and RORγt-Positive Regulatory T Cells. Biomolecules 2024; 14:89. [PMID: 38254689 PMCID: PMC10813120 DOI: 10.3390/biom14010089] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2023] [Revised: 01/05/2024] [Accepted: 01/09/2024] [Indexed: 01/24/2024] Open
Abstract
Oral tolerance has been defined as the specific suppression of immune responses to an antigen by prior oral administration of the antigen. It has been thought to serve to suppress food allergy. Previous studies have shown that dendritic cells (DCs) and regulatory T cells (Tregs) are involved in the induction of oral tolerance. However, the detailed mechanisms of Treg induction in oral tolerance remain largely unknown. Eosinophils have been recognized as effector cells in allergic diseases, but in recent years, the diverse functions of tissue-resident eosinophils have been reported. Eosinophils in the intestine have been reported to induce Tregs by releasing TGF-β, but the role of eosinophils in oral tolerance is still controversial. In this study, we analyzed the roles of eosinophils in oral tolerance using eosinophil-deficient ΔdblGATA mice (mice lacking a high-affinity GATA-binding site in the GATA1 promoter). ΔdblGATA mice showed impaired antigen-induced oral tolerance compared to wild-type mice. The induction of RORγt+ Tregs in mesenteric lymph nodes (MLNs) by oral tolerance induction was impaired in ΔdblGATA mice compared to wild-type mice. An increase in RORγt+ antigen-presenting cells (APCs), which are involved in RORγt+ Treg differentiation, in the intestine and MLNs was not seen in ΔdblGATA mice. Notably, the expansion of group 3 innate lymphoid cells (ILC3s), a subset of RORγt+ APCs, by oral tolerance induction was seen in wild-type mice but not ΔdblGATA mice. These results suggest that eosinophils are crucial in the induction of oral tolerance, possibly via the induction of RORγt+ APCs and RORγt+ Tregs.
Collapse
Affiliation(s)
- Shunjiro Kurihara
- Department of Allergy and Clinical Immunology, Graduate School of Medicine, Chiba University, 1-8-1 Inohana, Chiba 260-8670, Japan; (S.K.); (T.I.); (R.K.); (T.K.); (K.M.); (S.T.); (A.I.); (A.S.)
| | - Kotaro Suzuki
- Department of Allergy and Clinical Immunology, Graduate School of Medicine, Chiba University, 1-8-1 Inohana, Chiba 260-8670, Japan; (S.K.); (T.I.); (R.K.); (T.K.); (K.M.); (S.T.); (A.I.); (A.S.)
| | - Masaya Yokota
- Department of Allergy and Clinical Immunology, Graduate School of Medicine, Chiba University, 1-8-1 Inohana, Chiba 260-8670, Japan; (S.K.); (T.I.); (R.K.); (T.K.); (K.M.); (S.T.); (A.I.); (A.S.)
| | - Takashi Ito
- Department of Allergy and Clinical Immunology, Graduate School of Medicine, Chiba University, 1-8-1 Inohana, Chiba 260-8670, Japan; (S.K.); (T.I.); (R.K.); (T.K.); (K.M.); (S.T.); (A.I.); (A.S.)
- Synergy Institute for Futuristic Mucosal Vaccine Research and Development (cSIMVa), Chiba University, Chiba 260-8670, Japan;
| | - Yuki Hayashi
- Department of Allergy and Clinical Immunology, Graduate School of Medicine, Chiba University, 1-8-1 Inohana, Chiba 260-8670, Japan; (S.K.); (T.I.); (R.K.); (T.K.); (K.M.); (S.T.); (A.I.); (A.S.)
| | - Ryo Kikuchi
- Department of Allergy and Clinical Immunology, Graduate School of Medicine, Chiba University, 1-8-1 Inohana, Chiba 260-8670, Japan; (S.K.); (T.I.); (R.K.); (T.K.); (K.M.); (S.T.); (A.I.); (A.S.)
| | - Takahiro Kageyama
- Department of Allergy and Clinical Immunology, Graduate School of Medicine, Chiba University, 1-8-1 Inohana, Chiba 260-8670, Japan; (S.K.); (T.I.); (R.K.); (T.K.); (K.M.); (S.T.); (A.I.); (A.S.)
| | - Kazuyuki Meguro
- Department of Allergy and Clinical Immunology, Graduate School of Medicine, Chiba University, 1-8-1 Inohana, Chiba 260-8670, Japan; (S.K.); (T.I.); (R.K.); (T.K.); (K.M.); (S.T.); (A.I.); (A.S.)
| | - Shigeru Tanaka
- Department of Allergy and Clinical Immunology, Graduate School of Medicine, Chiba University, 1-8-1 Inohana, Chiba 260-8670, Japan; (S.K.); (T.I.); (R.K.); (T.K.); (K.M.); (S.T.); (A.I.); (A.S.)
| | - Arifumi Iwata
- Department of Allergy and Clinical Immunology, Graduate School of Medicine, Chiba University, 1-8-1 Inohana, Chiba 260-8670, Japan; (S.K.); (T.I.); (R.K.); (T.K.); (K.M.); (S.T.); (A.I.); (A.S.)
| | - Yoshiyuki Goto
- Synergy Institute for Futuristic Mucosal Vaccine Research and Development (cSIMVa), Chiba University, Chiba 260-8670, Japan;
- Division of Molecular Immunology, Medical Mycology Research Center, Chiba University, Chiba 260-8670, Japan
- Division of Pandemic and Post-Disaster Infectious Diseases, Research Institute of Disaster Medicine, Chiba University, Chiba 260-8670, Japan
- Division of Infectious Disease Vaccine R&D, Research Institute of Disaster Medicine, Chiba University, Chiba 260-8670, Japan
| | - Akira Suto
- Department of Allergy and Clinical Immunology, Graduate School of Medicine, Chiba University, 1-8-1 Inohana, Chiba 260-8670, Japan; (S.K.); (T.I.); (R.K.); (T.K.); (K.M.); (S.T.); (A.I.); (A.S.)
| | - Hiroshi Nakajima
- Department of Allergy and Clinical Immunology, Graduate School of Medicine, Chiba University, 1-8-1 Inohana, Chiba 260-8670, Japan; (S.K.); (T.I.); (R.K.); (T.K.); (K.M.); (S.T.); (A.I.); (A.S.)
- Synergy Institute for Futuristic Mucosal Vaccine Research and Development (cSIMVa), Chiba University, Chiba 260-8670, Japan;
| |
Collapse
|
19
|
Rivera JC, Opazo MC, Hernández-Armengol R, Álvarez O, Mendoza-León MJ, Caamaño E, Gatica S, Bohmwald K, Bueno SM, González PA, Neunlist M, Boudin H, Kalergis AM, Riedel CA. Transient gestational hypothyroxinemia accelerates and enhances ulcerative colitis-like disorder in the male offspring. Front Endocrinol (Lausanne) 2024; 14:1269121. [PMID: 38239991 PMCID: PMC10794346 DOI: 10.3389/fendo.2023.1269121] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/29/2023] [Accepted: 11/06/2023] [Indexed: 01/22/2024] Open
Abstract
Introduction Gestational hypothyroxinemia (HTX) is a condition that occurs frequently at the beginning of pregnancy, and it correlates with cognitive impairment, autism, and attentional deficit in the offspring. Evidence in animal models suggests that gestational HTX can increase the susceptibility of the offspring to develop strong inflammation in immune-mediated inflammatory diseases. Ulcerative colitis (UC) is a frequent inflammatory bowel disease with unknown causes. Therefore, the intensity of ulcerative colitis-like disorder (UCLD) and the cellular and molecular factors involved in proinflammatory or anti-inflammatory responses were analyzed in the offspring gestated in HTX (HTX-offspring) and compared with the offspring gestated in euthyroidism (Control-offspring). Methods Gestational HTX was induced by the administration of 2-mercapto-1-methylimidazole in drinking water to pregnant mice during E10-E14. The HTX-offspring were induced with UCLD by the acute administration of dextran sodium sulfate (DSS). The score of UCLD symptomatology was registered every day, and colon histopathology, immune cells, and molecular factors involved in the inflammatory or anti-inflammatory response were analyzed on day 6 of DSS treatment. Results The HTX-offspring displayed earlier UCLD pathological symptoms compared with the Control-offspring. After 6 days of DSS treatment, the HTX-offspring almost doubled the score of the Control-offspring. The histopathological analyses of the colon samples showed signs of inflammation at the distal and medial colon for both the HTX-offspring and Control-offspring. However, significantly more inflammatory features were detected in the proximal colon of the HTX-offspring induced with UCLD compared with the Control-offspring induced with UCLD. Significantly reduced mRNA contents encoding for protective molecules like glutamate-cysteine ligase catalytic subunit (GCLC) and mucin-2 (MUC-2) were found in the colon of the HTX-offspring as compared with the Control-offspring. Higher percentages of Th17 lymphocytes were detected in the colon tissues of the HTX-offspring induced or not with UCLD as compared with the Control-offspring. Discussion Gestational HTX accelerates the onset and increases the intensity of UCLD in the offspring. The low expression of MUC-2 and GCLC together with high levels of Th17 Lymphocytes in the colon tissue suggests that the HTX-offspring has molecular and cellular features that favor inflammation and tissue damage. These results are important evidence to be aware of the impact of gestational HTX as a risk factor for UCLD development in offspring.
Collapse
Affiliation(s)
- Juan Carlos Rivera
- Laboratorio de Endocrino-inmunología, Departamento de Ciencias Biológicas, Facultad de Ciencias de la Vida, Universidad Andrés Bello, Santiago, Chile
- Millennium Institute on Immunology and Immunotherapy, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Ma. Cecilia Opazo
- Millennium Institute on Immunology and Immunotherapy, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile
- Facultad de Medicina Veterinaria y Agronomía, Instituto de Ciencias Naturales, Universidad de las Américas, Santiago, Chile
| | - Rosario Hernández-Armengol
- Laboratorio de Endocrino-inmunología, Departamento de Ciencias Biológicas, Facultad de Ciencias de la Vida, Universidad Andrés Bello, Santiago, Chile
- Millennium Institute on Immunology and Immunotherapy, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Oscar Álvarez
- Laboratorio de Endocrino-inmunología, Departamento de Ciencias Biológicas, Facultad de Ciencias de la Vida, Universidad Andrés Bello, Santiago, Chile
- Millennium Institute on Immunology and Immunotherapy, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - María José Mendoza-León
- Laboratorio de Endocrino-inmunología, Departamento de Ciencias Biológicas, Facultad de Ciencias de la Vida, Universidad Andrés Bello, Santiago, Chile
- Millennium Institute on Immunology and Immunotherapy, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Esteban Caamaño
- Laboratorio de Endocrino-inmunología, Departamento de Ciencias Biológicas, Facultad de Ciencias de la Vida, Universidad Andrés Bello, Santiago, Chile
- Millennium Institute on Immunology and Immunotherapy, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Sebastian Gatica
- Laboratorio de Endocrino-inmunología, Departamento de Ciencias Biológicas, Facultad de Ciencias de la Vida, Universidad Andrés Bello, Santiago, Chile
- Millennium Institute on Immunology and Immunotherapy, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Karen Bohmwald
- Millennium Institute on Immunology and Immunotherapy, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile
- Instituto de Ciencias Biomédicas, Facultad de Ciencias de la Salud, Universidad Autónoma de Chile, Santiago, Chile
| | - Susan M. Bueno
- Millennium Institute on Immunology and Immunotherapy, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile
- Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Pablo A. González
- Millennium Institute on Immunology and Immunotherapy, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile
- Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Michel Neunlist
- Université de Nantes, Inserm, TENS, The Enteric Nervous System in Gut and Brain Disorders, IMAD, Nantes, France
| | - Helene Boudin
- Université de Nantes, Inserm, TENS, The Enteric Nervous System in Gut and Brain Disorders, IMAD, Nantes, France
| | - Alexis M. Kalergis
- Millennium Institute on Immunology and Immunotherapy, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile
- Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile
- Departamento de Endocrinología, Facultad de Medicina, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Claudia A. Riedel
- Laboratorio de Endocrino-inmunología, Departamento de Ciencias Biológicas, Facultad de Ciencias de la Vida, Universidad Andrés Bello, Santiago, Chile
- Millennium Institute on Immunology and Immunotherapy, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile
| |
Collapse
|
20
|
Calabrò A, Drommi F, De Pasquale C, Navarra G, Carrega P, Bonaccorsi I, Ferlazzo G, Campana S. Microbiota and plasticity of antigen-presenting ILC3s: impact on antitumor immune response. Gut Microbes 2024; 16:2390135. [PMID: 39161185 PMCID: PMC11340763 DOI: 10.1080/19490976.2024.2390135] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 08/21/2024] Open
Abstract
Growing evidence highlights the pivotal role of RORγt-innate lymphoid cells (ILCs) in the establishment of antitumor immune response and in enhancing tumor sensitivity to immunotherapy. Noteworthy, type 3 ILCs (ILC3s) have been recently acknowledged as an important class of antigen-presenting cells (APCs) in the context of host-microorganism interactions shaping the adaptive immune response in the intestinal mucosa. Although a broad range of mouse models has led to significant progress in untangling the role of ILC3s as APCs, the outcome of major histocompatibility complex (MHC)-dependent ILC-T cell crosstalk in colorectal cancer (CRC) remains underexplored in human. Moreover, expression of MHCII is confined to ILC3 subset, endowed with lymphoid tissue-inducing properties, that adopts tissue-specific fates and functions. Intestinal microbiota could dictate the plasticity of antigen-presenting ILC3s and we here summarize our current understanding of the functions of these cells in both mouse and human CRC discussing the role of microbiota as a key modulator of their tumor-suppressive activity.
Collapse
Affiliation(s)
- Alessia Calabrò
- Laboratory of Immunology and Biotherapy, Department of Human Pathology “G.Barresi”, University of Messina, Messina, Italy
| | - Fabiana Drommi
- Laboratory of Immunology and Biotherapy, Department of Human Pathology “G.Barresi”, University of Messina, Messina, Italy
| | - Claudia De Pasquale
- Laboratory of Immunology and Biotherapy, Department of Human Pathology “G.Barresi”, University of Messina, Messina, Italy
| | - Giuseppe Navarra
- Oncologic Surgery, Department of Human Pathology of Adult and Evolutive Age, University of Messina, Messina, Italy
| | - Paolo Carrega
- Laboratory of Immunology and Biotherapy, Department of Human Pathology “G.Barresi”, University of Messina, Messina, Italy
| | - Irene Bonaccorsi
- Laboratory of Immunology and Biotherapy, Department of Human Pathology “G.Barresi”, University of Messina, Messina, Italy
- Clinical Pathology Unit, University Hospital Policlinico “G. Martino”, Messina, Italy
| | - Guido Ferlazzo
- Department of Experimental Medicine (DIMES), University of Genoa, Genova, Italy
- Unit of Experimental Pathology and Immunology, IRCCS Ospedale Policlinico San Martino, Genova, Italy
| | - Stefania Campana
- Laboratory of Immunology and Biotherapy, Department of Human Pathology “G.Barresi”, University of Messina, Messina, Italy
| |
Collapse
|
21
|
Abramson J, Dobeš J, Lyu M, Sonnenberg GF. The emerging family of RORγt + antigen-presenting cells. Nat Rev Immunol 2024; 24:64-77. [PMID: 37479834 PMCID: PMC10844842 DOI: 10.1038/s41577-023-00906-5] [Citation(s) in RCA: 22] [Impact Index Per Article: 22.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/12/2023] [Indexed: 07/23/2023]
Abstract
Antigen-presenting cells (APCs) are master regulators of the immune response by directly interacting with T cells to orchestrate distinct functional outcomes. Several types of professional APC exist, including conventional dendritic cells, B cells and macrophages, and numerous other cell types have non-classical roles in antigen presentation, such as thymic epithelial cells, endothelial cells and granulocytes. Accumulating evidence indicates the presence of a new family of APCs marked by the lineage-specifying transcription factor retinoic acid receptor-related orphan receptor-γt (RORγt) and demonstrates that these APCs have key roles in shaping immunity, inflammation and tolerance, particularly in the context of host-microorganism interactions. These RORγt+ APCs include subsets of group 3 innate lymphoid cells, extrathymic autoimmune regulator-expressing cells and, potentially, other emerging populations. Here, we summarize the major findings that led to the discovery of these RORγt+ APCs and their associated functions. We discuss discordance in recent reports and identify gaps in our knowledge in this burgeoning field, which has tremendous potential to advance our understanding of fundamental immune concepts.
Collapse
Affiliation(s)
- Jakub Abramson
- Department of Immunology and Regenerative Biology, Weizmann Institute of Science, Rehovot, Israel.
| | - Jan Dobeš
- Department of Cell Biology, Faculty of Science, Charles University, Prague, Czech Republic
| | - Mengze Lyu
- Jill Roberts Institute for Research in Inflammatory Bowel Disease, Weill Cornell Medicine, Cornell University, New York, NY, USA
- Joan and Sanford I. Weill Department of Medicine, Division of Gastroenterology & Hepatology, Weill Cornell Medicine, Cornell University, New York, NY, USA
- Department of Microbiology & Immunology, Weill Cornell Medicine, Cornell University, New York, NY, USA
| | - Gregory F Sonnenberg
- Jill Roberts Institute for Research in Inflammatory Bowel Disease, Weill Cornell Medicine, Cornell University, New York, NY, USA.
- Joan and Sanford I. Weill Department of Medicine, Division of Gastroenterology & Hepatology, Weill Cornell Medicine, Cornell University, New York, NY, USA.
- Department of Microbiology & Immunology, Weill Cornell Medicine, Cornell University, New York, NY, USA.
| |
Collapse
|
22
|
Mori A, Ohno H, Satoh-Takayama N. Disease pathogenesis and barrier functions regulated by group 3 innate lymphoid cells. Semin Immunopathol 2024; 45:509-519. [PMID: 38305897 DOI: 10.1007/s00281-024-01000-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2023] [Accepted: 01/16/2024] [Indexed: 02/03/2024]
Abstract
The mucosal surface is in constant contact with foreign antigens and is regulated by unique mechanisms that are different from immune responses in the peripheral organs. For the last several decades, only adaptive immune cells such as helper T (Th) cells, Th1, Th2, or Th17 were targeted to study a wide variety of immune responses in the mucosal tissues. However, since their discovery, innate lymphoid cells (ILCs) have been attracting attention as a unique subset of immune cells that provide border defense with various functions and tissue specificity. ILCs are classified into different groups based on cell differentiation and functions. Group 3 innate lymphoid cells (ILC3s) are particularly in close proximity to mucosal surfaces and therefore have the opportunity to be exposed to a variety of bacteria including pathogenic bacteria. In recent years, studies have also provided much evidence that ILC3s contribute to disease pathogenesis as well as the defense of mucosal surfaces by rapidly responding to pathogens and coordinating other immune cells. As the counterpart of helper T cells, ILC3s together with other ILC subsets establish the immune balance between adaptive and innate immunity in protecting us from invasion or encounter with non-self-antigens for maintaining a complex homeostasis. In this review, we summarize recent advances in our understanding of ILCs, with a particular focus on the function of ILC3s in their involvement in bacterial infection and disease pathogenesis.
Collapse
Affiliation(s)
- Ayana Mori
- Immunobiology Laboratory, School of Science, Yokohama City University, 1-7-22, Suehiro, Tsurumi, Yokohama, 230-0045, Japan
- Laboratory for Intestinal Ecosystem, RIKEN Center for Integrative Medical Sciences, 1-7-22, Suehiro, Tsurumi, Yokohama City, Kanagawa, 230-0045, Japan
| | - Hiroshi Ohno
- Laboratory for Intestinal Ecosystem, RIKEN Center for Integrative Medical Sciences, 1-7-22, Suehiro, Tsurumi, Yokohama City, Kanagawa, 230-0045, Japan
- Immunobiology Laboratory, Graduate School of Medical Life Science, Yokohama City University, 1-7-22, Suehiro, Tsurumi, Yokohama, 230-0045, Japan
- Laboratory for Immune Regulation, Graduate School of Medicine, Chiba University, Chiba, 260-8670, Japan
| | - Naoko Satoh-Takayama
- Laboratory for Intestinal Ecosystem, RIKEN Center for Integrative Medical Sciences, 1-7-22, Suehiro, Tsurumi, Yokohama City, Kanagawa, 230-0045, Japan.
- Immunobiology Laboratory, Graduate School of Medical Life Science, Yokohama City University, 1-7-22, Suehiro, Tsurumi, Yokohama, 230-0045, Japan.
| |
Collapse
|
23
|
Roland MM, Peacock TE, Hall N, Mohammed AD, Ball R, Jolly A, Alexeev S, Dopkins N, Nagarkatti M, Nagarkatti P, Kubinak JL. B-cell-specific MhcII regulates microbiota composition in a primarily IgA-independent manner. Front Immunol 2023; 14:1253674. [PMID: 38187389 PMCID: PMC10766766 DOI: 10.3389/fimmu.2023.1253674] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2023] [Accepted: 11/30/2023] [Indexed: 01/09/2024] Open
Abstract
Background The expression of major histocompatibility complex class II (MhcII) molecules on B cells is required for the development of germinal centers (GCs) in lymphoid follicles; the primary sites for the generation of T-cell-dependent (TD) antibody responses. Peyer's patches (PPs) are secondary lymphoid tissues (SLOs) in the small intestine (SI) that give rise to high-affinity, TD antibodies (mainly immunoglobulin A (IgA)) generated against the microbiota. While several studies have demonstrated that MhcII antigen presentation by other immune cells coordinate TD IgA responses and regulate microbiota composition, whether or not B-cell-specific MhcII influences gut microbial ecology is unknown. Methods Here, we developed a novel Rag1 -/- adoptive co-transfer model to answer this question. In this model, Rag1 -/- mice were reconstituted with naïve CD4+ T cells and either MhcII-sufficient or MhcII-deficient naïve B cells. Subsequent to this, resulting shifts in microbiota composition was characterized via 16S rRNA gene sequencing of SI-resident and fecal bacterial communities. Results Results from our experiments indicate that SLO development and reconstitution of an anti-commensal TD IgA response can be induced in Rag1 -/- mice receiving T cells and MhcII-sufficient B cells, but not in mice receiving T cells and MhcII-deficient B cells. Results from our 16S experiments confirmed that adaptive immunity is a relevant host factor shaping microbial ecology in the gut, and that its impact was most pronounced on SI-resident bacterial communities. Conclusion Our data also clearly establishes that MhcII-mediated cognate interactions between B cells and T cells regulates this effect by maintaining species richness in the gut, which is a phenotype commonly associated with good health. Finally, contrary to expectations, our experimental results indicate that IgA was not responsible for driving any of the effects on the microbiota ascribed to the loss of B cell-specific MhcII. Collectively, results from our experiments support that MhcII-mediated antigen presentation by B cells regulates microbiota composition and promotes species richness through an IgA-independent mechanism.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | | | - Jason L. Kubinak
- Department of Pathology, Microbiology, and Immunology, University of South Carolina School of Medicine, Columbia, SC, United States
| |
Collapse
|
24
|
Li F, Liang Z, Zhong H, Hu X, Tang Z, Zhu C, Shen J, Han X, Lin R, Zheng R, Tang R, Peng H, Zheng X, Mo C, Chen P, Wang X, Wen Q, Li J, Xia X, Ye H, Qiu Y, Yu J, Fu D, Liu J, Wang R, Xie H, Guo Y, Li X, Fan J, Liu Q, Mao H, Chen W, Zhou Y. Group 3 Innate Lymphoid Cells Exacerbate Lupus Nephritis by Promoting B Cell Activation in Kidney Ectopic Lymphoid Structures. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2023; 10:e2302804. [PMID: 37915129 PMCID: PMC10724443 DOI: 10.1002/advs.202302804] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/03/2023] [Revised: 08/28/2023] [Indexed: 11/03/2023]
Abstract
Group 3 innate lymphoid cells (ILC3s) represent a new population in immune regulation, yet their role in lupus nephritis (LN) remains elusive. In the present work, systemic increases in ILC3s, particularly in the kidney, are observed to correlate strongly with disease severity in both human and murine LN. Using MRL/lpr lupus mice and a nephrotoxic serum-induced LN model, this study demonstrates that ILC3s accumulated in the kidney migrate predominantly from the intestine. Furthermore, intestinal ILC3s accelerate LN progression, manifested by exacerbated autoimmunity and kidney injuries. In LN kidneys, ILC3s are located adjacent to B cells within ectopic lymphoid structures (ELS), directly activating B cell differentiation into plasma cells and antibody production in a Delta-like1 (DLL1)/Notch-dependent manner. Blocking DLL1 attenuates ILC3s' effects and protects against LN. Altogether, these findings reveal a novel pathogenic role of ILC3s in B cell activation, renal ELS formation and autoimmune injuries during LN, shedding light on the therapeutic value of targeting ILC3s for LN.
Collapse
Affiliation(s)
- Feng Li
- Department of NephrologyThe First Affiliated Hospital, Sun Yat‐sen UniversityGuangzhou510080China
- NHC Key Laboratory of Clinical Nephrology (Sun Yat‐Sen University) and Guangdong Provincial Key Laboratory of NephrologyGuangzhou510080China
| | - Zhou Liang
- Department of NephrologyThe First Affiliated Hospital, Sun Yat‐sen UniversityGuangzhou510080China
- NHC Key Laboratory of Clinical Nephrology (Sun Yat‐Sen University) and Guangdong Provincial Key Laboratory of NephrologyGuangzhou510080China
| | - Haojie Zhong
- NHC Key Laboratory of Clinical Nephrology (Sun Yat‐Sen University) and Guangdong Provincial Key Laboratory of NephrologyGuangzhou510080China
- Department of Hepatobiliary and Pancreatic SurgeryThe First Affiliated Hospital, Shenzhen UniversityShenzhen518000China
| | - Xinrong Hu
- Department of NephrologyThe First Affiliated Hospital, Sun Yat‐sen UniversityGuangzhou510080China
- NHC Key Laboratory of Clinical Nephrology (Sun Yat‐Sen University) and Guangdong Provincial Key Laboratory of NephrologyGuangzhou510080China
| | - Ziwen Tang
- Department of NephrologyThe First Affiliated Hospital, Sun Yat‐sen UniversityGuangzhou510080China
- NHC Key Laboratory of Clinical Nephrology (Sun Yat‐Sen University) and Guangdong Provincial Key Laboratory of NephrologyGuangzhou510080China
| | - Changjian Zhu
- Department of NephrologyThe First Affiliated Hospital, Sun Yat‐sen UniversityGuangzhou510080China
- NHC Key Laboratory of Clinical Nephrology (Sun Yat‐Sen University) and Guangdong Provincial Key Laboratory of NephrologyGuangzhou510080China
| | - Jiani Shen
- Department of NephrologyThe First Affiliated Hospital, Sun Yat‐sen UniversityGuangzhou510080China
- NHC Key Laboratory of Clinical Nephrology (Sun Yat‐Sen University) and Guangdong Provincial Key Laboratory of NephrologyGuangzhou510080China
| | - Xu Han
- Department of NephrologyThe First Affiliated Hospital, Sun Yat‐sen UniversityGuangzhou510080China
- NHC Key Laboratory of Clinical Nephrology (Sun Yat‐Sen University) and Guangdong Provincial Key Laboratory of NephrologyGuangzhou510080China
| | - Ruoni Lin
- Department of NephrologyThe First Affiliated Hospital, Sun Yat‐sen UniversityGuangzhou510080China
- NHC Key Laboratory of Clinical Nephrology (Sun Yat‐Sen University) and Guangdong Provincial Key Laboratory of NephrologyGuangzhou510080China
| | - Ruilin Zheng
- Department of NephrologyThe First Affiliated Hospital, Sun Yat‐sen UniversityGuangzhou510080China
- NHC Key Laboratory of Clinical Nephrology (Sun Yat‐Sen University) and Guangdong Provincial Key Laboratory of NephrologyGuangzhou510080China
| | - Ruihan Tang
- Department of NephrologyThe First Affiliated Hospital, Sun Yat‐sen UniversityGuangzhou510080China
- NHC Key Laboratory of Clinical Nephrology (Sun Yat‐Sen University) and Guangdong Provincial Key Laboratory of NephrologyGuangzhou510080China
| | - Huajing Peng
- Department of NephrologyThe First Affiliated Hospital, Sun Yat‐sen UniversityGuangzhou510080China
- NHC Key Laboratory of Clinical Nephrology (Sun Yat‐Sen University) and Guangdong Provincial Key Laboratory of NephrologyGuangzhou510080China
| | - Xunhua Zheng
- Department of NephrologyThe First Affiliated Hospital, Sun Yat‐sen UniversityGuangzhou510080China
- NHC Key Laboratory of Clinical Nephrology (Sun Yat‐Sen University) and Guangdong Provincial Key Laboratory of NephrologyGuangzhou510080China
| | - Chengqiang Mo
- Department of UrologyThe First Affiliated Hospital, Sun Yat‐sen UniversityGuangzhou510080China
| | - Peisong Chen
- Department of Laboratory MedicineThe First Affiliated Hospital, Sun Yat‐sen UniversityGuangzhou510080China
| | - Xin Wang
- Department of NephrologyThe First Affiliated Hospital, Sun Yat‐sen UniversityGuangzhou510080China
- NHC Key Laboratory of Clinical Nephrology (Sun Yat‐Sen University) and Guangdong Provincial Key Laboratory of NephrologyGuangzhou510080China
| | - Qiong Wen
- Department of NephrologyThe First Affiliated Hospital, Sun Yat‐sen UniversityGuangzhou510080China
- NHC Key Laboratory of Clinical Nephrology (Sun Yat‐Sen University) and Guangdong Provincial Key Laboratory of NephrologyGuangzhou510080China
| | - Jianbo Li
- Department of NephrologyThe First Affiliated Hospital, Sun Yat‐sen UniversityGuangzhou510080China
- NHC Key Laboratory of Clinical Nephrology (Sun Yat‐Sen University) and Guangdong Provincial Key Laboratory of NephrologyGuangzhou510080China
| | - Xi Xia
- Department of NephrologyThe First Affiliated Hospital, Sun Yat‐sen UniversityGuangzhou510080China
- NHC Key Laboratory of Clinical Nephrology (Sun Yat‐Sen University) and Guangdong Provincial Key Laboratory of NephrologyGuangzhou510080China
| | - Hongjian Ye
- Department of NephrologyThe First Affiliated Hospital, Sun Yat‐sen UniversityGuangzhou510080China
- NHC Key Laboratory of Clinical Nephrology (Sun Yat‐Sen University) and Guangdong Provincial Key Laboratory of NephrologyGuangzhou510080China
| | - Yagui Qiu
- Department of NephrologyThe First Affiliated Hospital, Sun Yat‐sen UniversityGuangzhou510080China
- NHC Key Laboratory of Clinical Nephrology (Sun Yat‐Sen University) and Guangdong Provincial Key Laboratory of NephrologyGuangzhou510080China
| | - Jianwen Yu
- Department of NephrologyThe First Affiliated Hospital, Sun Yat‐sen UniversityGuangzhou510080China
- NHC Key Laboratory of Clinical Nephrology (Sun Yat‐Sen University) and Guangdong Provincial Key Laboratory of NephrologyGuangzhou510080China
| | - Dongying Fu
- Department of NephrologyThe First Affiliated Hospital, Sun Yat‐sen UniversityGuangzhou510080China
- NHC Key Laboratory of Clinical Nephrology (Sun Yat‐Sen University) and Guangdong Provincial Key Laboratory of NephrologyGuangzhou510080China
| | - Jiaqi Liu
- Department of NephrologyThe First Affiliated Hospital, Sun Yat‐sen UniversityGuangzhou510080China
- NHC Key Laboratory of Clinical Nephrology (Sun Yat‐Sen University) and Guangdong Provincial Key Laboratory of NephrologyGuangzhou510080China
| | - Rong Wang
- Department of NephrologyThe First Affiliated Hospital, Sun Yat‐sen UniversityGuangzhou510080China
- NHC Key Laboratory of Clinical Nephrology (Sun Yat‐Sen University) and Guangdong Provincial Key Laboratory of NephrologyGuangzhou510080China
| | - Huixin Xie
- Department of NephrologyThe First Affiliated Hospital, Sun Yat‐sen UniversityGuangzhou510080China
- NHC Key Laboratory of Clinical Nephrology (Sun Yat‐Sen University) and Guangdong Provincial Key Laboratory of NephrologyGuangzhou510080China
| | - Yun Guo
- Department of NephrologyThe First Affiliated Hospital, Sun Yat‐sen UniversityGuangzhou510080China
- NHC Key Laboratory of Clinical Nephrology (Sun Yat‐Sen University) and Guangdong Provincial Key Laboratory of NephrologyGuangzhou510080China
| | - Xiaoyan Li
- Department of NephrologyThe First Affiliated Hospital, Sun Yat‐sen UniversityGuangzhou510080China
- NHC Key Laboratory of Clinical Nephrology (Sun Yat‐Sen University) and Guangdong Provincial Key Laboratory of NephrologyGuangzhou510080China
| | - Jinjin Fan
- Department of NephrologyThe First Affiliated Hospital, Sun Yat‐sen UniversityGuangzhou510080China
- NHC Key Laboratory of Clinical Nephrology (Sun Yat‐Sen University) and Guangdong Provincial Key Laboratory of NephrologyGuangzhou510080China
| | - Qinghua Liu
- Department of NephrologyThe First Affiliated Hospital, Sun Yat‐sen UniversityGuangzhou510080China
- NHC Key Laboratory of Clinical Nephrology (Sun Yat‐Sen University) and Guangdong Provincial Key Laboratory of NephrologyGuangzhou510080China
| | - Haiping Mao
- Department of NephrologyThe First Affiliated Hospital, Sun Yat‐sen UniversityGuangzhou510080China
- NHC Key Laboratory of Clinical Nephrology (Sun Yat‐Sen University) and Guangdong Provincial Key Laboratory of NephrologyGuangzhou510080China
| | - Wei Chen
- Department of NephrologyThe First Affiliated Hospital, Sun Yat‐sen UniversityGuangzhou510080China
- NHC Key Laboratory of Clinical Nephrology (Sun Yat‐Sen University) and Guangdong Provincial Key Laboratory of NephrologyGuangzhou510080China
| | - Yi Zhou
- Department of NephrologyThe First Affiliated Hospital, Sun Yat‐sen UniversityGuangzhou510080China
- NHC Key Laboratory of Clinical Nephrology (Sun Yat‐Sen University) and Guangdong Provincial Key Laboratory of NephrologyGuangzhou510080China
| |
Collapse
|
25
|
Tachó-Piñot R, Stamper CT, King JI, Matei-Rascu V, Richardson E, Li Z, Roberts LB, Bassett JW, Melo-Gonzalez F, Fiancette R, Lin IH, Dent A, Harada Y, Finlay C, Mjösberg J, Withers DR, Hepworth MR. Bcl6 is a subset-defining transcription factor of lymphoid tissue inducer-like ILC3. Cell Rep 2023; 42:113425. [PMID: 37950867 PMCID: PMC7615641 DOI: 10.1016/j.celrep.2023.113425] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2023] [Revised: 09/18/2023] [Accepted: 10/26/2023] [Indexed: 11/13/2023] Open
Abstract
Innate lymphoid cells (ILCs) are tissue-resident effector cells with roles in tissue homeostasis, protective immunity, and inflammatory disease. Group 3 ILCs (ILC3s) are classically defined by the master transcription factor RORγt. However, ILC3 can be further subdivided into subsets that share type 3 effector modules that exhibit significant ontological, transcriptional, phenotypic, and functional heterogeneity. Notably lymphoid tissue inducer (LTi)-like ILC3s mediate effector functions not typically associated with other RORγt-expressing lymphocytes, suggesting that additional transcription factors contribute to dictate ILC3 subset phenotypes. Here, we identify Bcl6 as a subset-defining transcription factor of LTi-like ILC3s in mice and humans. Deletion of Bcl6 results in dysregulation of the LTi-like ILC3 transcriptional program and markedly enhances expression of interleukin-17A (IL-17A) and IL-17F in LTi-like ILC3s in a manner in part dependent upon the commensal microbiota-and associated with worsened inflammation in a model of colitis. Together, these findings redefine our understanding of ILC3 subset biology.
Collapse
Affiliation(s)
- Roser Tachó-Piñot
- Lydia Becker Institute of Immunology and Inflammation, University of Manchester, Manchester M13 9PL, UK; Division of Immunology, Immunity to Infection and Respiratory Medicine, School of Biological Sciences, Faculty of Biology, Medicine and Health, Manchester Academic Health Science Centre, University of Manchester, Manchester M13 9PL, UK
| | - Christopher T Stamper
- Center for Infectious Medicine, Department of Medicine Huddinge, Karolinska Institutet, Stockholm, Sweden; Medical Unit for Lung and Allergy Diseases, Karolinska University Hospital, Stockholm, Sweden
| | - James I King
- Lydia Becker Institute of Immunology and Inflammation, University of Manchester, Manchester M13 9PL, UK; Division of Immunology, Immunity to Infection and Respiratory Medicine, School of Biological Sciences, Faculty of Biology, Medicine and Health, Manchester Academic Health Science Centre, University of Manchester, Manchester M13 9PL, UK
| | - Veronika Matei-Rascu
- Institute of Immunology and Immunotherapy, College of Medical and Dental Sciences, University of Birmingham, Birmingham B15 2TT, UK
| | - Erin Richardson
- Institute of Immunology and Immunotherapy, College of Medical and Dental Sciences, University of Birmingham, Birmingham B15 2TT, UK
| | - Zhi Li
- Institute of Immunology and Immunotherapy, College of Medical and Dental Sciences, University of Birmingham, Birmingham B15 2TT, UK
| | - Luke B Roberts
- Lydia Becker Institute of Immunology and Inflammation, University of Manchester, Manchester M13 9PL, UK; Division of Immunology, Immunity to Infection and Respiratory Medicine, School of Biological Sciences, Faculty of Biology, Medicine and Health, Manchester Academic Health Science Centre, University of Manchester, Manchester M13 9PL, UK
| | - John W Bassett
- Center for Infectious Medicine, Department of Medicine Huddinge, Karolinska Institutet, Stockholm, Sweden; Medical Unit for Lung and Allergy Diseases, Karolinska University Hospital, Stockholm, Sweden
| | - Felipe Melo-Gonzalez
- Lydia Becker Institute of Immunology and Inflammation, University of Manchester, Manchester M13 9PL, UK; Division of Immunology, Immunity to Infection and Respiratory Medicine, School of Biological Sciences, Faculty of Biology, Medicine and Health, Manchester Academic Health Science Centre, University of Manchester, Manchester M13 9PL, UK
| | - Rémi Fiancette
- Institute of Immunology and Immunotherapy, College of Medical and Dental Sciences, University of Birmingham, Birmingham B15 2TT, UK
| | - I-Hsuan Lin
- Bioinformatics Core Facility, University of Manchester, Manchester M13 9PL, UK
| | - Alexander Dent
- Department of Microbiology and Immunology, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Yohsuke Harada
- Laboratory of Pharmaceutical Immunology, Faculty of Pharmaceutical Sciences, Tokyo University of Science, 2641 Yamazaki, Noda, Chiba 278-8510, Japan
| | - Conor Finlay
- Lydia Becker Institute of Immunology and Inflammation, University of Manchester, Manchester M13 9PL, UK; Division of Immunology, Immunity to Infection and Respiratory Medicine, School of Biological Sciences, Faculty of Biology, Medicine and Health, Manchester Academic Health Science Centre, University of Manchester, Manchester M13 9PL, UK; School of Medicine, Trinity Translational Medicine Institute, Trinity College Dublin, Dublin, Ireland
| | - Jenny Mjösberg
- Center for Infectious Medicine, Department of Medicine Huddinge, Karolinska Institutet, Stockholm, Sweden; Medical Unit for Lung and Allergy Diseases, Karolinska University Hospital, Stockholm, Sweden
| | - David R Withers
- Institute of Immunology and Immunotherapy, College of Medical and Dental Sciences, University of Birmingham, Birmingham B15 2TT, UK
| | - Matthew R Hepworth
- Lydia Becker Institute of Immunology and Inflammation, University of Manchester, Manchester M13 9PL, UK; Division of Immunology, Immunity to Infection and Respiratory Medicine, School of Biological Sciences, Faculty of Biology, Medicine and Health, Manchester Academic Health Science Centre, University of Manchester, Manchester M13 9PL, UK.
| |
Collapse
|
26
|
Koprivica I, Stanisavljević S, Mićanović D, Jevtić B, Stojanović I, Miljković Đ. ILC3: a case of conflicted identity. Front Immunol 2023; 14:1271699. [PMID: 37915588 PMCID: PMC10616800 DOI: 10.3389/fimmu.2023.1271699] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2023] [Accepted: 10/02/2023] [Indexed: 11/03/2023] Open
Abstract
Innate lymphoid cells type 3 (ILC3s) are the first line sentinels at the mucous tissues, where they contribute to the homeostatic immune response in a major way. Also, they have been increasingly appreciated as important modulators of chronic inflammatory and autoimmune responses, both locally and systemically. The proper identification of ILC3 is of utmost importance for meaningful studies on their role in immunity. Flow cytometry is the method of choice for the detection and characterization of ILC3. However, the analysis of ILC3-related papers shows inconsistency in ILC3 phenotypic definition, as different inclusion and exclusion markers are used for their identification. Here, we present these discrepancies in the phenotypic characterization of human and mouse ILC3s. We discuss the pros and cons of using various markers for ILC3 identification. Furthermore, we consider the possibilities for the efficient isolation and propagation of ILC3 from different organs and tissues for in-vitro and in-vivo studies. This paper calls upon uniformity in ILC3 definition, isolation, and propagation for the increased possibility of confluent interpretation of ILC3's role in immunity.
Collapse
Affiliation(s)
| | | | | | | | | | - Đorđe Miljković
- Department of Immunology, Institute for Biological Research “Siniša Stanković” - National Institute of Republic of Serbia, University of Belgrade, Belgrade, Serbia
| |
Collapse
|
27
|
King JI, Melo-Gonzalez F, Malengier-Devlies B, Tachó-Piñot R, Magalhaes MS, Hodge SH, Romero Ros X, Gentek R, Hepworth MR. Bcl-2 supports survival and metabolic fitness of quiescent tissue-resident ILC3. Mucosal Immunol 2023; 16:658-670. [PMID: 37453568 PMCID: PMC10564625 DOI: 10.1016/j.mucimm.2023.07.001] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2023] [Revised: 07/07/2023] [Accepted: 07/10/2023] [Indexed: 07/18/2023]
Abstract
Group 3 innate lymphoid cells (ILC3) are potent effector cells with critical roles in enforcing immunity, barrier integrity and tissue homeostasis along the gastrointestinal tract. ILC3 are considered primarily tissue-resident cells, seeding the gastrointestinal tract during embryonic stages and early life. However, the mechanisms through which mature ILC3 are maintained within adult tissues are poorly understood. Here, we report that lymphoid tissue-inducer-like (LTi-like) ILC3 exhibit minimal turnover in the healthy adult intestinal tract, persist for extended periods of time, and display a quiescent phenotype. Strikingly, during enteric bacterial infection LTi-like ILC3 also exhibit negligible hematopoietic replenishment and remain non-proliferative, despite robustly producing cytokines. Survival of LTi-like ILC3 was found to be dependent upon the balance between the metabolic activity required to drive effector function and anti-apoptotic programs. Notably, the pro-survival protein B-cell lymphoma-2 (Bcl-2) was required for the survival of LTi-like ILC3 ex vivo but was rendered partially dispensable if mitochondrial respiration was inhibited. Together we demonstrate LTi-like ILC3 are a tissue-resident, quiescent population that persist independently of hematopoietic replenishment to survive within the intestinal microenvironment.
Collapse
Affiliation(s)
- James I King
- Lydia Becker Institute of Immunology and Inflammation, University of Manchester, Manchester, United Kingdom; Division of Immunology, Immunity to Infection and Respiratory Medicine, School of Biological Sciences, Faculty of Biology, Medicine and Health, Manchester Academic Health Science Centre, University of Manchester, Manchester, United Kingdom
| | - Felipe Melo-Gonzalez
- Lydia Becker Institute of Immunology and Inflammation, University of Manchester, Manchester, United Kingdom; Division of Immunology, Immunity to Infection and Respiratory Medicine, School of Biological Sciences, Faculty of Biology, Medicine and Health, Manchester Academic Health Science Centre, University of Manchester, Manchester, United Kingdom
| | - Bert Malengier-Devlies
- Institute for Regeneration and Repair, Centre for Inflammation Research & Centre for Reproductive Health, University of Edinburgh, Edinburgh, United Kingdom
| | - Roser Tachó-Piñot
- Lydia Becker Institute of Immunology and Inflammation, University of Manchester, Manchester, United Kingdom; Division of Immunology, Immunity to Infection and Respiratory Medicine, School of Biological Sciences, Faculty of Biology, Medicine and Health, Manchester Academic Health Science Centre, University of Manchester, Manchester, United Kingdom
| | - Marlene S Magalhaes
- Institute for Regeneration and Repair, Centre for Inflammation Research & Centre for Reproductive Health, University of Edinburgh, Edinburgh, United Kingdom
| | - Suzanne H Hodge
- Lydia Becker Institute of Immunology and Inflammation, University of Manchester, Manchester, United Kingdom; Division of Immunology, Immunity to Infection and Respiratory Medicine, School of Biological Sciences, Faculty of Biology, Medicine and Health, Manchester Academic Health Science Centre, University of Manchester, Manchester, United Kingdom
| | - Xavier Romero Ros
- Bioscience Asthma, Research and Early Development, Respiratory & Immunology, BioPharmaceuticals R&D, AstraZeneca, Cambridge, United Kingdom
| | - Rebecca Gentek
- Institute for Regeneration and Repair, Centre for Inflammation Research & Centre for Reproductive Health, University of Edinburgh, Edinburgh, United Kingdom
| | - Matthew R Hepworth
- Lydia Becker Institute of Immunology and Inflammation, University of Manchester, Manchester, United Kingdom; Division of Immunology, Immunity to Infection and Respiratory Medicine, School of Biological Sciences, Faculty of Biology, Medicine and Health, Manchester Academic Health Science Centre, University of Manchester, Manchester, United Kingdom.
| |
Collapse
|
28
|
Liang B, Xing D. Unveiling the mystery of ILC3s: Their functions and interactions in mucosal immunity. Int Immunopharmacol 2023; 123:110772. [PMID: 37552906 DOI: 10.1016/j.intimp.2023.110772] [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: 03/26/2023] [Revised: 08/02/2023] [Accepted: 08/03/2023] [Indexed: 08/10/2023]
Abstract
Innate lymphoid cells (ILCs) are a recently discovered subset of immune cells that play a crucial role in preserving tissue health and combating infections. Among these, ILC3s are particularly vital in regulating mucosal immunity across multiple organs such as the gut, lungs, and skin. The purpose of this article is to present a comprehensive and detailed overview of current knowledge on ILC3s, with a specific emphasis on their intricate interactions with various components of the intestinal microenvironment. Recent research on the complex, bidirectional communication pathways between ILC3s and intestinal epithelial cells, stromal cells, immune cells, microbiota, their metabolites, and diet are highlighted. Furthermore, this review comprehensively examines the diverse functions of ILC3s, which include lymphoid tissue development, tissue repair, infection, inflammation, and metabolic diseases, as well as the effector molecules that facilitate these functions. Overall, this review provides valuable insights into the biological and functional aspects of ILC3s and underscores their potential for developing innovative therapies for immune-mediated disorders, while also acknowledging the remaining knowledge gaps and challenges that need to be addressed.
Collapse
Affiliation(s)
- Bing Liang
- Cancer Institute, The Affiliated Hospital of Qingdao University, Qingdao, China; Qingdao Cancer Institute, Qingdao University, Qingdao, China.
| | - Dongming Xing
- Cancer Institute, The Affiliated Hospital of Qingdao University, Qingdao, China; Qingdao Cancer Institute, Qingdao University, Qingdao, China; School of Life Sciences, Tsinghua University, Beijing, China
| |
Collapse
|
29
|
Sah P, Zenewicz LA. Modulation of innate lymphoid cells by enteric bacterial pathogens. Front Immunol 2023; 14:1219072. [PMID: 37483638 PMCID: PMC10358831 DOI: 10.3389/fimmu.2023.1219072] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2023] [Accepted: 06/22/2023] [Indexed: 07/25/2023] Open
Abstract
Innate lymphoid cells (ILCs) are key regulators of tissue homeostasis, inflammation, and immunity to infections. ILCs rapidly respond to environmental cues such as cytokines, microbiota and invading pathogens which regulate their function and phenotype. Even though ILCs are rare cells, they are enriched at barrier surfaces such as the gastrointestinal (GI) tract, and they are often critical to the host's immune response to eliminate pathogens. On the other side of host-pathogen interactions, pathogenic bacteria also have the means to modulate these immune responses. Manipulation or evasion of the immune cells is often to the pathogen's benefit and/or to the detriment of competing microbiota. In some instances, specific bacterial virulence factors or toxins have been implicated in how the pathogen modulates immunity. In this review, we discuss the recent progress made towards understanding the role of non-cytotoxic ILCs during enteric bacterial infections, how these pathogens can modulate the immune response, and the implications these have on developing new therapies to combat infection.
Collapse
Affiliation(s)
| | - Lauren A. Zenewicz
- Department of Microbiology and Immunology, College of Medicine, The University of Oklahoma Health Sciences Center, Oklahoma City, OK, United States
| |
Collapse
|
30
|
Lv X, Zhu S, Wu J, Shi J, Wei Q, Li T, Yang N, Liu C, Qi L, Zang G, Cheng H, Yang Z, Jin C, Wang Y, Cui J, Ueno H, Liu YJ, Chen J. Reciprocal costimulatory molecules control the activation of mucosal type 3 innate lymphoid cells during engagement with B cells. Cell Mol Immunol 2023; 20:808-819. [PMID: 37225838 PMCID: PMC10310834 DOI: 10.1038/s41423-023-01041-w] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2022] [Accepted: 05/09/2023] [Indexed: 05/26/2023] Open
Abstract
Innate lymphoid cells (ILCs) are the counterpart of T helper cells in the innate immune system and share multiple phenotypes with T helper cells. Inducible T-cell costimulator (ICOS) is recognized on T cells and participates in T-cell activation and T and B-cell engagement in lymphoid tissues. However, the role of ICOS in ILC3s and ILC3-involved interactions with the immune microenvironment remains unclear. Here, we found that ICOS expression on human ILC3s was correlated with the activated state of ILC3s. ICOS costimulation enhanced the survival, proliferation, and capacity of ILC3s to produce cytokines (IL-22, IL-17A, IFN-γ, TNF, and GM-CSF). Via synergistic effects of ICOS and CD40 signaling, B cells promoted ILC3 functions, and ILC3-induced T-cell-independent B-cell IgA and IgM secretion primarily required CD40 signaling. Hence, ICOS is essential for the nonredundant role of ILC3s and their interaction with adjacent B cells.
Collapse
Affiliation(s)
- Xinping Lv
- Cancer Center, First Hospital of Jilin University, Changchun, Jilin, 130021, China
- Laboratory for Tumor Immunology, First Hospital of Jilin University, Changchun, Jilin, 130061, China
| | - Shan Zhu
- Cancer Center, First Hospital of Jilin University, Changchun, Jilin, 130021, China
- Laboratory for Tumor Immunology, First Hospital of Jilin University, Changchun, Jilin, 130061, China
| | - Jing Wu
- Cancer Center, First Hospital of Jilin University, Changchun, Jilin, 130021, China
- Laboratory for Tumor Immunology, First Hospital of Jilin University, Changchun, Jilin, 130061, China
| | - Jinfeng Shi
- Department of Otolaryngology Head and Neck Surgery, First Hospital of Jilin University, Changchun, Jilin, 130021, China
| | - Qiuyu Wei
- Laboratory for Tumor Immunology, First Hospital of Jilin University, Changchun, Jilin, 130061, China
| | - Tete Li
- Laboratory for Tumor Immunology, First Hospital of Jilin University, Changchun, Jilin, 130061, China
- Department of Translational Medicine, Changchun GeneScience Pharmaceuticals Co., Ltd., Changchun, Jilin, 130012, China
| | - Ning Yang
- Laboratory for Tumor Immunology, First Hospital of Jilin University, Changchun, Jilin, 130061, China
| | - Chunyan Liu
- Laboratory for Tumor Immunology, First Hospital of Jilin University, Changchun, Jilin, 130061, China
- Department of Gynecology, First Hospital of Jilin University, Changchun, Jilin, 130021, China
| | - Lingli Qi
- Laboratory for Tumor Immunology, First Hospital of Jilin University, Changchun, Jilin, 130061, China
- Department of Pediatric Gastroenterology, First Hospital of Jilin University, Changchun, Jilin, 130021, China
| | - Guoxia Zang
- Laboratory for Tumor Immunology, First Hospital of Jilin University, Changchun, Jilin, 130061, China
| | - Hang Cheng
- Laboratory for Tumor Immunology, First Hospital of Jilin University, Changchun, Jilin, 130061, China
- Department of Pediatrics, First Hospital of Jilin University, Changchun, Jilin, 130021, China
| | - Zhiguang Yang
- Department of Thoracic Surgery, First Hospital of Jilin University, Changchun, Jilin, 130021, China
| | - Chengyan Jin
- Department of Thoracic Surgery, Second Hospital of Jilin University, Changchun, Jilin, 130041, China
| | - Yusheng Wang
- Department of Otolaryngology Head and Neck Surgery, First Hospital of Jilin University, Changchun, Jilin, 130021, China
| | - Jiuwei Cui
- Cancer Center, First Hospital of Jilin University, Changchun, Jilin, 130021, China
| | - Hideki Ueno
- Department of Immunology, Graduate School of Medicine, Kyoto University, Kyoto, Japan
- ASHBi Institute for the Advanced Study of Human Biology, Kyoto University, Kyoto, Japan
| | - Yong-Jun Liu
- Laboratory for Tumor Immunology, First Hospital of Jilin University, Changchun, Jilin, 130061, China.
| | - Jingtao Chen
- Cancer Center, First Hospital of Jilin University, Changchun, Jilin, 130021, China.
- Laboratory for Tumor Immunology, First Hospital of Jilin University, Changchun, Jilin, 130061, China.
| |
Collapse
|
31
|
Li JH, Hepworth MR, O'Sullivan TE. Regulation of systemic metabolism by tissue-resident immune cell circuits. Immunity 2023; 56:1168-1186. [PMID: 37315533 PMCID: PMC10321269 DOI: 10.1016/j.immuni.2023.05.001] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2023] [Revised: 04/11/2023] [Accepted: 05/02/2023] [Indexed: 06/16/2023]
Abstract
Recent studies have demonstrated that tissue homeostasis and metabolic function are dependent on distinct tissue-resident immune cells that form functional cell circuits with structural cells. Within these cell circuits, immune cells integrate cues from dietary contents and commensal microbes in addition to endocrine and neuronal signals present in the tissue microenvironment to regulate structural cell metabolism. These tissue-resident immune circuits can become dysregulated during inflammation and dietary overnutrition, contributing to metabolic diseases. Here, we review the evidence describing key cellular networks within and between the liver, gastrointestinal tract, and adipose tissue that control systemic metabolism and how these cell circuits become dysregulated during certain metabolic diseases. We also identify open questions in the field that have the potential to enhance our understanding of metabolic health and disease.
Collapse
Affiliation(s)
- Joey H Li
- Department of Microbiology, Immunology, and Molecular Genetics, David Geffen School of Medicine at UCLA, Los Angeles, CA 900953, USA; Medical Scientist Training Program, David Geffen School of Medicine at UCLA, Los Angeles, CA, USA
| | - Matthew R Hepworth
- Division of Immunology, Immunity to Infection and Respiratory Medicine, Faculty of Biology, Medicine and Health, Manchester Collaborative Centre for Inflammation Research, Lydia Becker Institute of Immunology and Inflammation, Manchester Academic Health Science Centre, The University of Manchester, Manchester, UK
| | - Timothy E O'Sullivan
- Department of Microbiology, Immunology, and Molecular Genetics, David Geffen School of Medicine at UCLA, Los Angeles, CA 900953, USA.
| |
Collapse
|
32
|
Drommi F, Calabrò A, Vento G, Pezzino G, Cavaliere R, Omero F, Muscolino P, Granata B, D'Anna F, Silvestris N, De Pasquale C, Ferlazzo G, Campana S. Crosstalk between ILC3s and Microbiota: Implications for Colon Cancer Development and Treatment with Immune Check Point Inhibitors. Cancers (Basel) 2023; 15:cancers15112893. [PMID: 37296855 DOI: 10.3390/cancers15112893] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2023] [Revised: 05/17/2023] [Accepted: 05/22/2023] [Indexed: 06/12/2023] Open
Abstract
Type 3 innate lymphoid cells (ILC3s) are primarily tissue-resident cells strategically localized at the intestinal barrier that exhibit the fast-acting responsiveness of classic innate immune cells. Populations of these lymphocytes depend on the transcription factor RAR-related orphan receptor and play a key role in maintaining intestinal homeostasis, keeping host-microbial mutualism in check. Current evidence has indicated a bidirectional relationship between microbiota and ILC3s. While ILC3 function and maintenance in the gut are influenced by commensal microbiota, ILC3s themselves can control immune responses to intestinal microbiota by providing host defense against extracellular bacteria, helping to maintain a diverse microbiota and inducing immune tolerance for commensal bacteria. Thus, ILC3s have been linked to host-microbiota interactions and the loss of their normal activity promotes dysbiosis, chronic inflammation and colon cancer. Furthermore, recent evidence has suggested that a healthy dialog between ILC3s and gut microbes is necessary to support antitumor immunity and response to immune checkpoint inhibitor (ICI) therapy. In this review, we summarize the functional interactions occurring between microbiota and ILC3s in homeostasis, providing an overview of the molecular mechanisms orchestrating these interactions. We focus on how alterations in this interplay promote gut inflammation, colorectal cancer and resistance to therapies with immune check point inhibitors.
Collapse
Affiliation(s)
- Fabiana Drommi
- Laboratory of Immunology and Biotherapy, Department Human Pathology "G.Barresi", University of Messina, 98122 Messina, Italy
| | - Alessia Calabrò
- Laboratory of Immunology and Biotherapy, Department Human Pathology "G.Barresi", University of Messina, 98122 Messina, Italy
| | - Grazia Vento
- Department of Experimental Medicine (DIMES), University of Genoa, 16132 Genova, Italy
| | - Gaetana Pezzino
- Unit of Experimental Pathology and Immunology, IRCCS Ospedale Policlinico San Martino, 16132 Genova, Italy
| | - Riccardo Cavaliere
- Laboratory of Immunology and Biotherapy, Department Human Pathology "G.Barresi", University of Messina, 98122 Messina, Italy
| | - Fausto Omero
- Medical Oncology Unit, Department of Human Pathology "G.Barresi", University of Messina, 98125 Messina, Italy
| | - Paola Muscolino
- Medical Oncology Unit, Department of Human Pathology "G.Barresi", University of Messina, 98125 Messina, Italy
| | - Barbara Granata
- Medical Oncology Unit, Department of Human Pathology "G.Barresi", University of Messina, 98125 Messina, Italy
| | - Federica D'Anna
- Medical Oncology Unit, Department of Human Pathology "G.Barresi", University of Messina, 98125 Messina, Italy
| | - Nicola Silvestris
- Medical Oncology Unit, Department of Human Pathology "G.Barresi", University of Messina, 98125 Messina, Italy
| | - Claudia De Pasquale
- Laboratory of Immunology and Biotherapy, Department Human Pathology "G.Barresi", University of Messina, 98122 Messina, Italy
| | - Guido Ferlazzo
- Department of Experimental Medicine (DIMES), University of Genoa, 16132 Genova, Italy
- Unit of Experimental Pathology and Immunology, IRCCS Ospedale Policlinico San Martino, 16132 Genova, Italy
| | - Stefania Campana
- Laboratory of Immunology and Biotherapy, Department Human Pathology "G.Barresi", University of Messina, 98122 Messina, Italy
| |
Collapse
|
33
|
Du Y, Gao Y, Hu M, Hou J, Yang L, Wang X, Du W, Liu J, Xu Q. Colonization and development of the gut microbiome in calves. J Anim Sci Biotechnol 2023; 14:46. [PMID: 37031166 PMCID: PMC10082981 DOI: 10.1186/s40104-023-00856-x] [Citation(s) in RCA: 45] [Impact Index Per Article: 22.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2022] [Accepted: 02/16/2023] [Indexed: 04/10/2023] Open
Abstract
Colonization and development of the gut microbiome are crucial for the growth and health of calves. In this review, we summarized the colonization, beneficial nutrition, immune function of gut microbiota, function of the gut barrier, and the evolution of core microbiota in the gut of calves of different ages. Homeostasis of gut microbiome is beneficial for nutritional and immune system development of calves. Disruption of the gut microbiome leads to digestive diseases in calves, such as diarrhea and intestinal inflammation. Microbiota already exists in the gut of calf fetuses, and the colonization of microbiota continues to change dynamically under the influence of various factors, which include probiotics, diet, age, and genotype. Colonization depends on the interaction between the gut microbiota and the immune system of calves. The abundance and diversity of these commensal microbiota stabilize and play a critical role in the health of calves.
Collapse
Affiliation(s)
- Yufeng Du
- College of Animal Sciences and Technology, Huazhong Agricultural University, Wuhan, 430070, China
| | - Ya Gao
- College of Animal Sciences and Technology, Huazhong Agricultural University, Wuhan, 430070, China
| | - Mingyang Hu
- College of Animal Sciences and Technology, Huazhong Agricultural University, Wuhan, 430070, China
| | - Jinxiu Hou
- College of Animal Sciences and Technology, Huazhong Agricultural University, Wuhan, 430070, China
| | - Linhai Yang
- College of Animal Sciences and Technology, Huazhong Agricultural University, Wuhan, 430070, China
| | - Xianghuang Wang
- College of Animal Sciences and Technology, Huazhong Agricultural University, Wuhan, 430070, China
| | - Wenjuan Du
- College of Animal Sciences and Technology, Huazhong Agricultural University, Wuhan, 430070, China
| | - Jianxin Liu
- MoE Key Laboratory of Molecular Animal Nutrition, College of Animal Sciences, Zhejiang University, Hangzhou, 310058, China
| | - Qingbiao Xu
- College of Animal Sciences and Technology, Huazhong Agricultural University, Wuhan, 430070, China.
- MoE Key Laboratory of Molecular Animal Nutrition, College of Animal Sciences, Zhejiang University, Hangzhou, 310058, China.
| |
Collapse
|
34
|
Zhang Y, Feng X, Chen J, Liu J, Wu J, Tan H, Mi Z, Rong P. Controversial role of ILC3s in intestinal diseases: A novelty perspective on immunotherapy. Front Immunol 2023; 14:1134636. [PMID: 37063879 PMCID: PMC10090672 DOI: 10.3389/fimmu.2023.1134636] [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: 12/30/2022] [Accepted: 03/13/2023] [Indexed: 03/31/2023] Open
Abstract
ILC3s have been identified as crucial immune regulators that play a role in maintaining host homeostasis and modulating the antitumor response. Emerging evidence supports the idea that LTi cells play an important role in initiating lymphoid tissue development, while other ILC3s can promote host defense and orchestrate adaptive immunity, mainly through the secretion of specific cytokines and crosstalk with other immune cells or tissues. Additionally, dysregulation of ILC3-mediated overexpression of cytokines, changes in subset abundance, and conversion toward other ILC subsets are closely linked with the occurrence of tumors and inflammatory diseases. Regulation of ILC3 cytokines, ILC conversion and LTi-induced TLSs may be a novel strategy for treating tumors and intestinal or extraintestinal inflammatory diseases. Herein, we discuss the development of ILCs, the biology of ILC3s, ILC plasticity, the correlation of ILC3s and adaptive immunity, crosstalk with the intestinal microenvironment, controversial roles of ILC3s in intestinal diseases and potential applications for treatment.
Collapse
Affiliation(s)
- Yunshu Zhang
- Department of Radiology, The Third Xiangya Hospital, Central South University, Changsha, Hunan, China
- Xiangya School of Medicine, Central South University, Changsha, China
| | - Xuefei Feng
- Department of Government & Public Administration, The Chinese University of Hong Kong, Hong Kong, Hong Kong SAR, China
| | - Juan Chen
- Department of Radiology, The Third Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Jiahao Liu
- Department of Radiology, The Third Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Jianmin Wu
- Department of Radiology, The Third Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Hongpei Tan
- Department of Radiology, The Third Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Ze Mi
- Department of Radiology, The Third Xiangya Hospital, Central South University, Changsha, Hunan, China
- *Correspondence: Ze Mi, ; Pengfei Rong,
| | - Pengfei Rong
- Department of Radiology, The Third Xiangya Hospital, Central South University, Changsha, Hunan, China
- Key Laboratory of Biological Nanotechnology of National Health Commission, Xiangya Hospital, Central South University, Changsha, Hunan, China
- *Correspondence: Ze Mi, ; Pengfei Rong,
| |
Collapse
|
35
|
Mattiola I, Diefenbach A. Regulation of innate immune system function by the microbiome: Consequences for tumor immunity and cancer immunotherapy. Semin Immunol 2023; 66:101724. [PMID: 36758379 DOI: 10.1016/j.smim.2023.101724] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/20/2022] [Revised: 01/03/2023] [Accepted: 01/17/2023] [Indexed: 02/10/2023]
Abstract
Innate effector cells are immune cells endowed with host protective features and cytotoxic functions. By sensing the tissue environment, innate cells have an important role in regulating the transition from homeostasis to inflammation and the establishment of pathological states, including the onset and development of cancer. The tumor microenvironment induces molecular and functional modifications in innate cells, dampening their capability to initiate and sustain anti-tumor immune responses. Emerging studies clearly showed a contribution of the microbiota in modulating the functions of innate cells in cancer. Commensal microorganisms can not only directly interact with innate cells in the tumor microenvironment but can also exert immunomodulatory features from non-tumor sites through the release of microbial products. The microbiota can mediate the priming of innate cells at mucosal tissues and determine the strength of immune responses mediated by such cells when they migrate to non-mucosal tissues, having an impact on cancer. Finally, several evidences reported a strong contribution of the microbiota in promoting innate immune responses during anti-cancer therapies leading to enhanced therapeutic efficacy. In this review, we considered the current knowledge on the role of the microbiota in shaping host innate immune responses in cancer.
Collapse
Affiliation(s)
- Irene Mattiola
- Laboratory of Innate Immunity, Institute of Microbiology, Infectious Diseases and Immunology (I-MIDI), Campus Benjamin Franklin, Charité - Universitätsmedizin Berlin, Germany; Mucosal and Developmental Immunology, Deutsches Rheuma-Forschungszentrum (DRFZ), an Institute of the Leibniz Association, Berlin, Germany.
| | - Andreas Diefenbach
- Laboratory of Innate Immunity, Institute of Microbiology, Infectious Diseases and Immunology (I-MIDI), Campus Benjamin Franklin, Charité - Universitätsmedizin Berlin, Germany; Mucosal and Developmental Immunology, Deutsches Rheuma-Forschungszentrum (DRFZ), an Institute of the Leibniz Association, Berlin, Germany; Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany.
| |
Collapse
|
36
|
van de Pavert SA. Layered origins of lymphoid tissue inducer cells. Immunol Rev 2023; 315:71-78. [PMID: 36705244 DOI: 10.1111/imr.13189] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023]
Abstract
The Innate Lymphoid Cell (ILC) family is a relatively recently described immune cell family involved in innate immune responses and tissue homeostasis. Lymphoid Tissue Inducer (LTi) cells are part of the type 3 (ILC3) family. The ILC3 family is the main ILC population within the embryo, in which the LTi cells are critically associated with embryonic lymph node formation. Recent studies have shown more insights in ILC origin and residency from local embryonic and tissue resident precursors. Embryonic LTi cells originating from a different hemogenic endothelial source were shown to be replaced by HSC derived progenitors in adult. This review will discuss the layered origin of the ILC3 family with an emphasis on the LTi cell lineage.
Collapse
Affiliation(s)
- Serge A van de Pavert
- Aix-Marseille Univ, CNRS, INSERM, Centre d'Immunologie de Marseille-Luminy (CIML), Marseille, France
| |
Collapse
|
37
|
Bickel J, Bermudez LE. Induction of killing of Mycobacterium avium subsp. hominissuis in macrophages by cytokine stimulated innate-like lymphoid cells is negatively affected by the pathogen. INTERNATIONAL MICROBIOLOGY : THE OFFICIAL JOURNAL OF THE SPANISH SOCIETY FOR MICROBIOLOGY 2023:10.1007/s10123-023-00326-4. [PMID: 36662342 PMCID: PMC10397143 DOI: 10.1007/s10123-023-00326-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/01/2022] [Revised: 01/03/2023] [Accepted: 01/10/2023] [Indexed: 01/21/2023]
Abstract
Mycobacterium avium subsp. hominissuis (MAH) is a common environmental bacterium that causes infection in immunocompromised patients such as those with HIV/AIDS, or patients with chronic lung disease such as cystic fibrosis. There are many strains of MAH with varying levels of virulence. Infection with MAH strains 100 and 104 has been associated with different immune responses in mice and outcome of the disease. While MAH 100 infection tends to be cleared from mice, MAH 104 is virulent and grows in host tissue. What is currently unknown are the mechanisms related to this difference in host defense and virulence. Our hypothesis is that differences in circulating innate lymphocytes response are associated with increased protection from infection. Innate lymphoid cells (ILC) are lymphoid cells with an important role in regulation of innate immune systems. ILCs can be categorized into three subpopulations ILC1, ILC2, and ILC3 based on their cytokine production and regulatory transcription factors. Investigation was carried out on how macrophage anti-MAH response change depending on activation by primary mouse lymphocytes activated with IL-12, IL-33, and IL-23, triggering differentiation into ILC-like subpopulations. Our results do not affirm the role of any one ILC subpopulation in macrophage anti-M. avium ability. Our findings instead support the conclusion that MAH infection of macrophages suppresses the stimulatory function of ILCs.
Collapse
Affiliation(s)
- Jay Bickel
- Department of Biomedical Science, College of Veterinary Medicine, Oregon State University, Corvallis, OR, USA
| | - Luiz E Bermudez
- Department of Biomedical Science, College of Veterinary Medicine, Oregon State University, Corvallis, OR, USA.
- Department of Microbiology, College of Science, Oregon State University, Corvallis, OR, USA.
| |
Collapse
|
38
|
Li M, Wang Z, Jiang W, Lu Y, Zhang J. The role of group 3 innate lymphoid cell in intestinal disease. Front Immunol 2023; 14:1171826. [PMID: 37122757 PMCID: PMC10140532 DOI: 10.3389/fimmu.2023.1171826] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2023] [Accepted: 04/03/2023] [Indexed: 05/02/2023] Open
Abstract
Group 3 innate lymphoid cells (ILC3s), a novel subpopulation of lymphocytes enriched in the intestinal mucosa, are currently considered as key sentinels in maintaining intestinal immune homeostasis. ILC3s can secrete a series of cytokines such as IL-22 to eliminate intestinal luminal antigens, promote epithelial tissue repair and mucosal barrier integrity, and regulate intestinal immunity by integrating multiple signals from the environment and the host. However, ILC3 dysfunction may be associated with the development and progression of various diseases in the gut. Therefore, in this review, we will discuss the role of ILC3 in intestinal diseases such as enteric infectious diseases, intestinal inflammation, and tumors, with a focus on recent research advances and discoveries to explore potential therapeutic targets.
Collapse
|
39
|
Akuzum B, Lee JY. Context-Dependent Regulation of Type17 Immunity by Microbiota at the Intestinal Barrier. Immune Netw 2022; 22:e46. [PMID: 36627936 PMCID: PMC9807962 DOI: 10.4110/in.2022.22.e46] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2022] [Revised: 07/26/2022] [Accepted: 08/01/2022] [Indexed: 12/30/2022] Open
Abstract
T-helper-17 (Th17) cells and related IL-17-producing (type17) lymphocytes are abundant at the epithelial barrier. In response to bacterial and fungal infection, the signature cytokines IL-17A/F and IL-22 mediate the antimicrobial immune response and contribute to wound healing of injured tissues. Despite their protective function, type17 lymphocytes are also responsible for various chronic inflammatory disorders, including inflammatory bowel disease (IBD) and colitis associated cancer (CAC). A deeper understanding of type17 regulatory mechanisms could ultimately lead to the discovery of therapeutic strategies for the treatment of chronic inflammatory disorders and the prevention of cancer. In this review, we discuss the current understanding of the development and function of type17 immune cells at the intestinal barrier, focusing on the impact of microbiota-immune interactions on intestinal barrier homeostasis and disease etiology.
Collapse
Affiliation(s)
- Begum Akuzum
- Department of Microbiology and Immunology, Yonsei University College of Medicine, Seoul 03722, Korea
| | - June-Yong Lee
- Department of Microbiology and Immunology, Yonsei University College of Medicine, Seoul 03722, Korea
- Institute for Immunology and Immunological Diseases, Yonsei University College of Medicine, Seoul 03722, Korea
- Brain Korea 21 PLUS Project for Medical Sciences, Yonsei University College of Medicine, Seoul 03722, Korea
| |
Collapse
|
40
|
Wu X, Khatun A, Kasmani MY, Chen Y, Zheng S, Atkinson S, Nguyen C, Burns R, Taparowsky EJ, Salzman NH, Hand TW, Cui W. Group 3 innate lymphoid cells require BATF to regulate gut homeostasis in mice. J Exp Med 2022; 219:e20211861. [PMID: 36048018 PMCID: PMC9440727 DOI: 10.1084/jem.20211861] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2021] [Revised: 04/18/2022] [Accepted: 08/03/2022] [Indexed: 12/22/2022] Open
Abstract
Group 3 innate lymphoid cells (ILC3s) are crucial for the maintenance of host-microbiota homeostasis in gastrointestinal mucosal tissues. The mechanisms that maintain lineage identity of intestinal ILC3s and ILC3-mediated orchestration of microbiota and mucosal T cell immunity are elusive. Here, we identified BATF as a gatekeeper of ILC3 homeostasis in the gut. Depletion of BATF in ILC3s resulted in excessive interferon-γ production, dysbiosis, aberrant T cell immune responses, and spontaneous inflammatory bowel disease (IBD), which was considerably ameliorated by the removal of adaptive immunity, interferon-γ blockade, or antibiotic treatment. Mechanistically, BATF directly binds to the cis-regulatory elements of type 1 effector genes, restrains their chromatin accessibility, and inhibits their expression. Conversely, BATF promotes chromatin accessibility of genes involved in MHCII antigen processing and presentation pathways, which in turn directly promotes the transition of precursor ILC3s to MHCII+ ILC3s. Collectively, our findings reveal that BATF is a key transcription factor for maintaining ILC3 stability and coordinating ILC3-mediated control of intestinal homeostasis.
Collapse
Affiliation(s)
- Xiaopeng Wu
- Blood Research Institute, Versiti Wisconsin, Milwaukee, WI
| | - Achia Khatun
- Blood Research Institute, Versiti Wisconsin, Milwaukee, WI
- Department of Microbiology and Immunology, Medical College of Wisconsin, Milwaukee, WI
| | - Moujtaba Y. Kasmani
- Blood Research Institute, Versiti Wisconsin, Milwaukee, WI
- Department of Microbiology and Immunology, Medical College of Wisconsin, Milwaukee, WI
| | - Yao Chen
- Blood Research Institute, Versiti Wisconsin, Milwaukee, WI
- Department of Microbiology and Immunology, Medical College of Wisconsin, Milwaukee, WI
| | - Shikan Zheng
- Blood Research Institute, Versiti Wisconsin, Milwaukee, WI
| | - Samantha Atkinson
- Department of Pediatrics, Division of Gastroenterology and Center for Microbiome Research, Medical College of Wisconsin, Milwaukee, WI
| | - Christine Nguyen
- Blood Research Institute, Versiti Wisconsin, Milwaukee, WI
- Department of Microbiology and Immunology, Medical College of Wisconsin, Milwaukee, WI
| | - Robert Burns
- Blood Research Institute, Versiti Wisconsin, Milwaukee, WI
| | - Elizabeth J. Taparowsky
- Department of Biological Sciences and Purdue University Center for Cancer Research, Purdue University, West Lafayette, IN
| | - Nita H. Salzman
- Department of Pediatrics, Division of Gastroenterology and Center for Microbiome Research, Medical College of Wisconsin, Milwaukee, WI
| | - Timothy W. Hand
- R.K. Mellon Institute for Pediatric Research, Pediatrics Department, Infectious Disease Section, UPMC Children’s Hospital of Pittsburgh, University of Pittsburgh, Pittsburgh, PA
| | - Weiguo Cui
- Blood Research Institute, Versiti Wisconsin, Milwaukee, WI
- Department of Microbiology and Immunology, Medical College of Wisconsin, Milwaukee, WI
| |
Collapse
|
41
|
Bruchard M, Spits H. The role of ILC subsets in cancer. Semin Immunol 2022; 61-64:101654. [PMID: 36306660 DOI: 10.1016/j.smim.2022.101654] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/07/2022] [Revised: 09/17/2022] [Accepted: 09/19/2022] [Indexed: 12/15/2022]
Abstract
The family of innate lymphoid cells (ILCs) are composed of five canonical subsets, NK cells, ILC1, ILC2, ILC3 and Lymphoid tissue inducer cells. ILCs have important functions in early stages of immune response towards infectious agents. ILCs are highly plastic enabling rapid modification of their functions dependent on the type of microbe and tissue environment to optimally counter these microbes. Data that still accumulate in a rapid pace indicate that these cells are also involved in immunity against tumor cells. Paradoxically ILC subsets have been shown to have tumor suppressing and tumor promoting activities. In this brief review we provide a snapshot of our current knowledge of characteristics and functions of tumor infiltrating ILC subsets and speculate on how these cells can be harnessed to mediate anti-tumor immunity.
Collapse
Affiliation(s)
- Mélanie Bruchard
- INSERM U1231, Equipe Labellisée Ligue Contre le Cancer, Dijon, France, Cancer Biology Transfer Platform, Georges-Francois Leclerc Cancer Center-UNICANCER, Dijon, France, University of Burgundy and Franche Comté, Dijon, France.
| | - Hergen Spits
- Amsterdam UMC location University of Amsterdam, department of Experimental Immunology, Cancer Center Amsterdam, Amsterdam Institute for Infection & Immunity, Meibergdreef 9, Amsterdam, Netherlands.
| |
Collapse
|
42
|
Wen Y, Zhang W, Yang R, Jiang L, Zhang X, Wang B, Hua Y, Ji P, Yuan Z, Wei Y, Yao W. Regulation of Yujin Powder alcoholic extracts on ILC3s-TD IgA-colonic mucosal flora axis of DSS-induced ulcerative colitis. Front Microbiol 2022; 13:1039884. [PMID: 36338041 PMCID: PMC9633017 DOI: 10.3389/fmicb.2022.1039884] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2022] [Accepted: 10/07/2022] [Indexed: 11/29/2022] Open
Abstract
The intestinal flora maintained by the immune system plays an important role in healthy colon. However, the role of ILC3s-TD IgA-colonic mucosal flora axis in ulcerative colitis (UC) and whether it could become an innovative pathway for the treatment of UC is unknown. Yujin Powder is a classic prescription for treatment of dampness-heat type intestine disease in traditional Chinese medicine and has therapeutic effects on UC. Hence, the present study aimed to investigate the regulatory mechanism of Yujin Powder alcoholic extracts (YJP-A) on UC via ILC3s-TD IgA-colonic mucosal flora axis. The UC mouse model was induced by drinking 3.5% dextran sodium sulfate (DSS), meanwhile, YJP-A was given orally for prevention. During the experiment, the clinical symptoms of mice were recorded. Then the intestinal injury and inflammatory response of mice about UC were detected after the experiment. In addition, the relevant indicators of ILC3s-TD IgA-colonic mucosal flora axis were detected. The results showed that YJP-A had good therapy effects on DSS-induced mice UC: improved the symptoms, increased body weight and the length of colon, decreased the disease activity index score, ameliorated the intestinal injury, and reduced the inflammation etc. Also, YJP-A significantly increased the ILC3s proportion and the expression level of MHC II; significantly decreased the proportion of Tfh cells and B cells and the expression levels of Bcl6, IL-4, Aicda in mesenteric lymph nodes of colon in UC mice and IgA in colon. In addition, by 16S rDNA sequencing, YJP-A could restore TD IgA targets colonic mucus flora in UC mice by decreasing the relative abundance of Mucispirillum, Lachnospiraceae and increasing the relative abundance of Allprevotella, Alistipes, and Ruminococcaceae etc. In conclusion, our results demonstrated that the ILC3s-TD IgA-colonic mucosal flora axis was disordered in UC mice. YJP-A could significantly promote the proliferation of ILC3s to inhibit Tfh responses and B cells class switching through MHC II, further to limit TD IgA responses toward colonic mucosal flora. Our findings suggested that this axis may be a novel and promising strategy to prevent UC.
Collapse
Affiliation(s)
- Yanqiao Wen
- Institute of Traditional Chinese Veterinary Medicine, College of Veterinary Medicine, Gansu Agricultural University, Lanzhou, China
| | - Wangdong Zhang
- Laboratory of Veterinary Pathology, College of Veterinary Medicine, Gansu Agricultural University, Lanzhou, China
| | - Rong Yang
- Institute of Traditional Chinese Veterinary Medicine, College of Veterinary Medicine, Gansu Agricultural University, Lanzhou, China
| | - Lidong Jiang
- Institute of Traditional Chinese Veterinary Medicine, College of Veterinary Medicine, Gansu Agricultural University, Lanzhou, China
| | - Xiaosong Zhang
- Institute of Traditional Chinese Veterinary Medicine, College of Veterinary Medicine, Gansu Agricultural University, Lanzhou, China
| | - Baoshan Wang
- Laboratory of Veterinary Pathology, College of Veterinary Medicine, Gansu Agricultural University, Lanzhou, China
| | - Yongli Hua
- Institute of Traditional Chinese Veterinary Medicine, College of Veterinary Medicine, Gansu Agricultural University, Lanzhou, China
| | - Peng Ji
- Institute of Traditional Chinese Veterinary Medicine, College of Veterinary Medicine, Gansu Agricultural University, Lanzhou, China
| | - Ziwen Yuan
- Institute of Traditional Chinese Veterinary Medicine, College of Veterinary Medicine, Gansu Agricultural University, Lanzhou, China
| | - Yanming Wei
- Institute of Traditional Chinese Veterinary Medicine, College of Veterinary Medicine, Gansu Agricultural University, Lanzhou, China
- *Correspondence: Yanming Wei; Wanling Yao,
| | - Wanling Yao
- Institute of Traditional Chinese Veterinary Medicine, College of Veterinary Medicine, Gansu Agricultural University, Lanzhou, China
- *Correspondence: Yanming Wei; Wanling Yao,
| |
Collapse
|
43
|
Liu A, Wang X, Liang X, Wang W, Li C, Qian J, Zhang X. Human umbilical cord mesenchymal stem cells regulate immunoglobulin a secretion and remodel the diversification of intestinal microbiota to improve colitis. Front Cell Infect Microbiol 2022; 12:960208. [PMID: 36118029 PMCID: PMC9478446 DOI: 10.3389/fcimb.2022.960208] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2022] [Accepted: 08/10/2022] [Indexed: 11/20/2022] Open
Abstract
BACKGROUND Mesenchymal stem cell (MSC) therapy has emerged as a promising novel therapeutic strategy for managing inflammatory bowel disease (IBD) mainly via dampening inflammation, regulating immune disorders, and promoting mucosal tissue repair. However, in the process, the associated changes in the gut microbiota and the underlying mechanism are not yet clear. METHODS In the present study, dextran sulfate sodium (DSS) was used to induce colitis in mice. Mice with colitis were treated with intraperitoneal infusions of MSCs from human umbilical cord mesenchymal stem cells (HUMSCs) and evaluated for severity of inflammation including weight reduction, diarrhea, bloody stools, histopathology, and mortality. The proportion of regulatory T cells (Tregs) and immunoglobulin A-positive (IgA+) plasmacytes in gut-associated lymphoid tissue were determined. The intestinal and fecal levels of IgA were tested, and the proportion of IgA-coated bacteria was also determined. Fecal microbiome was analyzed using 16S rRNA gene sequencing analyses. RESULTS Treatment with HUMSCs ameliorated the clinical abnormalities and histopathologic severity of acute colitis in mice. Furthermore, the proportion of Tregs in both Peyer's patches and lamina propria of the small intestine was significantly increased. Meanwhile, the proportion of IgA+ plasmacytes was also substantially higher in the MSCs group than that of the DSS group, resulting in elevated intestinal and fecal levels of IgA. The proportion of IgA-coated bacteria was also upregulated in the MSCs group. In addition, the microbiome alterations in mice with colitis were partially restored to resemble those of healthy mice following treatment with HUMSCs. CONCLUSIONS Therapeutically administered HUMSCs ameliorate DSS-induced colitis partially via regulating the Tregs-IgA response, promoting the secretion of IgA, and facilitating further the restoration of intestinal microbiota, which provides a potential therapeutic mechanism for HUMSCs in the treatment of IBD.
Collapse
Affiliation(s)
- Airu Liu
- Hebei Key Laboratory of Gastroenterology, Hebei Clinical Research Center for Digestive Diseases, Department of Gastroenterology, Hebei Institute of Gastroenterology, The Second Hospital of Hebei Medical University, Shijiazhuang, China
| | - Xing Wang
- Hebei Key Laboratory of Gastroenterology, Hebei Clinical Research Center for Digestive Diseases, Department of Gastroenterology, Hebei Institute of Gastroenterology, The Second Hospital of Hebei Medical University, Shijiazhuang, China
| | - Xiaonan Liang
- Hebei Key Laboratory of Gastroenterology, Hebei Clinical Research Center for Digestive Diseases, Department of Gastroenterology, Hebei Institute of Gastroenterology, The Second Hospital of Hebei Medical University, Shijiazhuang, China
| | - Wenxin Wang
- Hebei Key Laboratory of Gastroenterology, Hebei Clinical Research Center for Digestive Diseases, Department of Gastroenterology, Hebei Institute of Gastroenterology, The Second Hospital of Hebei Medical University, Shijiazhuang, China
| | - Chenyang Li
- Hebei Key Laboratory of Gastroenterology, Hebei Clinical Research Center for Digestive Diseases, Department of Gastroenterology, Hebei Institute of Gastroenterology, The Second Hospital of Hebei Medical University, Shijiazhuang, China
| | - Jiaming Qian
- Hebei Key Laboratory of Gastroenterology, Hebei Clinical Research Center for Digestive Diseases, Department of Gastroenterology, Hebei Institute of Gastroenterology, The Second Hospital of Hebei Medical University, Shijiazhuang, China
- Department of Gastroenterology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Xiaolan Zhang
- Hebei Key Laboratory of Gastroenterology, Hebei Clinical Research Center for Digestive Diseases, Department of Gastroenterology, Hebei Institute of Gastroenterology, The Second Hospital of Hebei Medical University, Shijiazhuang, China
| |
Collapse
|
44
|
Penny HA, Domingues RG, Krauss MZ, Melo-Gonzalez F, Lawson MA, Dickson S, Parkinson J, Hurry M, Purse C, Jegham E, Godinho-Silva C, Rendas M, Veiga-Fernandes H, Bechtold DA, Grencis RK, Toellner KM, Waisman A, Swann JR, Gibbs JE, Hepworth MR. Rhythmicity of intestinal IgA responses confers oscillatory commensal microbiota mutualism. Sci Immunol 2022; 7:eabk2541. [PMID: 36054336 PMCID: PMC7613662 DOI: 10.1126/sciimmunol.abk2541] [Citation(s) in RCA: 34] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Interactions between the mammalian host and commensal microbiota are enforced through a range of immune responses that confer metabolic benefits and promote tissue health and homeostasis. Immunoglobulin A (IgA) responses directly determine the composition of commensal species that colonize the intestinal tract but require substantial metabolic resources to fuel antibody production by tissue-resident plasma cells. Here, we demonstrate that IgA responses are subject to diurnal regulation over the course of a circadian day. Specifically, the magnitude of IgA secretion, as well as the transcriptome of intestinal IgA+ plasma cells, was found to exhibit rhythmicity. Oscillatory IgA responses were found to be entrained by time of feeding and were also found to be in part coordinated by the plasma cell-intrinsic circadian clock via deletion of the master clock gene Arntl. Moreover, reciprocal interactions between the host and microbiota dictated oscillatory dynamics among the commensal microbial community and its associated transcriptional and metabolic activity in an IgA-dependent manner. Together, our findings suggest that circadian networks comprising intestinal IgA, diet, and the microbiota converge to align circadian biology in the intestinal tract and to ensure host-microbial mutualism.
Collapse
Affiliation(s)
- Hugo A. Penny
- Lydia Becker Institute of Immunology and Inflammation, University of Manchester, M13 9PL, Manchester, United Kingdom
- School of Biological Sciences, Faculty of Biology, Medicine and Health, Manchester Academic Health Science Centre, University of Manchester, M13 9PL, Manchester, United Kingdom
| | - Rita G. Domingues
- Lydia Becker Institute of Immunology and Inflammation, University of Manchester, M13 9PL, Manchester, United Kingdom
- School of Biological Sciences, Faculty of Biology, Medicine and Health, Manchester Academic Health Science Centre, University of Manchester, M13 9PL, Manchester, United Kingdom
| | - Maria Z. Krauss
- Lydia Becker Institute of Immunology and Inflammation, University of Manchester, M13 9PL, Manchester, United Kingdom
- School of Biological Sciences, Faculty of Biology, Medicine and Health, Manchester Academic Health Science Centre, University of Manchester, M13 9PL, Manchester, United Kingdom
| | - Felipe Melo-Gonzalez
- Lydia Becker Institute of Immunology and Inflammation, University of Manchester, M13 9PL, Manchester, United Kingdom
- School of Biological Sciences, Faculty of Biology, Medicine and Health, Manchester Academic Health Science Centre, University of Manchester, M13 9PL, Manchester, United Kingdom
| | - Melissa A.E. Lawson
- Lydia Becker Institute of Immunology and Inflammation, University of Manchester, M13 9PL, Manchester, United Kingdom
- School of Biological Sciences, Faculty of Biology, Medicine and Health, Manchester Academic Health Science Centre, University of Manchester, M13 9PL, Manchester, United Kingdom
| | - Suzanna Dickson
- Lydia Becker Institute of Immunology and Inflammation, University of Manchester, M13 9PL, Manchester, United Kingdom
- School of Biological Sciences, Faculty of Biology, Medicine and Health, Manchester Academic Health Science Centre, University of Manchester, M13 9PL, Manchester, United Kingdom
- Centre for Biological Timing, Faculty of Biology, Medicine and Health, University of Manchester, M13 9PL, Manchester, United Kingdom
| | - James Parkinson
- Lydia Becker Institute of Immunology and Inflammation, University of Manchester, M13 9PL, Manchester, United Kingdom
- School of Biological Sciences, Faculty of Biology, Medicine and Health, Manchester Academic Health Science Centre, University of Manchester, M13 9PL, Manchester, United Kingdom
| | - Madeleine Hurry
- Lydia Becker Institute of Immunology and Inflammation, University of Manchester, M13 9PL, Manchester, United Kingdom
- School of Biological Sciences, Faculty of Biology, Medicine and Health, Manchester Academic Health Science Centre, University of Manchester, M13 9PL, Manchester, United Kingdom
| | - Catherine Purse
- Lydia Becker Institute of Immunology and Inflammation, University of Manchester, M13 9PL, Manchester, United Kingdom
- School of Biological Sciences, Faculty of Biology, Medicine and Health, Manchester Academic Health Science Centre, University of Manchester, M13 9PL, Manchester, United Kingdom
| | - Emna Jegham
- Lydia Becker Institute of Immunology and Inflammation, University of Manchester, M13 9PL, Manchester, United Kingdom
- School of Biological Sciences, Faculty of Biology, Medicine and Health, Manchester Academic Health Science Centre, University of Manchester, M13 9PL, Manchester, United Kingdom
| | | | - Miguel Rendas
- Champalimaud Research, Champalimaud Centre for the Unknown, Lisbon, 1400-038, Portugal
| | | | - David A. Bechtold
- Centre for Biological Timing, Faculty of Biology, Medicine and Health, University of Manchester, M13 9PL, Manchester, United Kingdom
| | - Richard K. Grencis
- Lydia Becker Institute of Immunology and Inflammation, University of Manchester, M13 9PL, Manchester, United Kingdom
- School of Biological Sciences, Faculty of Biology, Medicine and Health, Manchester Academic Health Science Centre, University of Manchester, M13 9PL, Manchester, United Kingdom
- Wellcome Centre for Cell Matrix Research, University of Manchester, M13 9PL, Manchester, United Kingdom
| | - Kai-Michael Toellner
- Institute of Immunology and Immunotherapy, College of Medical & Dental Sciences, Medical School, University of Birmingham, Birmingham, B15 2TT, UK
| | - Ari Waisman
- Institute for Molecular Medicine, University Medical Center of the Johannes Gutenberg-University Mainz, Mainz, Germany
| | - Jonathan R. Swann
- School of Human Development and Health, Faculty of Medicine, University of Southampton, SO16 6YD, Southampton, United Kingdom
| | - Julie E. Gibbs
- Lydia Becker Institute of Immunology and Inflammation, University of Manchester, M13 9PL, Manchester, United Kingdom
- School of Biological Sciences, Faculty of Biology, Medicine and Health, Manchester Academic Health Science Centre, University of Manchester, M13 9PL, Manchester, United Kingdom
- Centre for Biological Timing, Faculty of Biology, Medicine and Health, University of Manchester, M13 9PL, Manchester, United Kingdom
| | - Matthew R. Hepworth
- Lydia Becker Institute of Immunology and Inflammation, University of Manchester, M13 9PL, Manchester, United Kingdom
- School of Biological Sciences, Faculty of Biology, Medicine and Health, Manchester Academic Health Science Centre, University of Manchester, M13 9PL, Manchester, United Kingdom
| |
Collapse
|
45
|
Cerutti A, Filipska M, Fa XM, Tachó-Piñot R. Impact of the mucosal milieu on antibody responses to allergens. J Allergy Clin Immunol 2022; 150:503-512. [PMID: 36075636 DOI: 10.1016/j.jaci.2022.07.007] [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: 06/02/2022] [Revised: 07/19/2022] [Accepted: 07/20/2022] [Indexed: 11/30/2022]
Abstract
Respiratory and digestive mucosal surfaces are continually exposed to common environmental antigens, which include potential allergens. Although innocuous in healthy individuals, allergens cause allergy in predisposed subjects and do so by triggering a pathologic TH2 cell response that induces IgE class switching and somatic hypermutation in allergen-specific B cells. The ensuing affinity maturation and plasma cell differentiation lead to the abnormal release of high-affinity IgE that binds to powerful FcεRI receptors on basophils and mast cells. When cross-linked by allergen, FcεRI-bound IgE instigates the release of prestored and de novo-induced proinflammatory mediators. Aside from causing type I hypersensitivity reactions underlying allergy, IgE affords protection against nematodes or venoms from insects and snakes, which raises questions as to the fundamental differences between protective and pathogenic IgE responses. In this review, we discuss the impact of the mucosal environment, including the epithelial and mucus barriers, on the induction of protective IgE responses against environmental antigens. We further discuss how perturbations of these barriers may contribute to the induction of pathogenic IgE production.
Collapse
Affiliation(s)
- Andrea Cerutti
- Catalan Institute for Research and Advanced Studies, Barcelona, Spain; Institut Hospital del Mar d'Investigacions Mèdiques, Barcelona Biomedical Research Park, Barcelona, Spain; Division of Clinical Immunology, Department of Medicine, Mount Sinai School of Medicine, New York.
| | - Martyna Filipska
- Institut Hospital del Mar d'Investigacions Mèdiques, Barcelona Biomedical Research Park, Barcelona, Spain
| | - Xavi Marcos Fa
- Institut Hospital del Mar d'Investigacions Mèdiques, Barcelona Biomedical Research Park, Barcelona, Spain
| | - Roser Tachó-Piñot
- Lydia Becher Institute of Immunology and Inflammation, University of Manchester, Manchester, United Kingdom
| |
Collapse
|
46
|
Einenkel R, Ehrhardt J, Zygmunt M, Muzzio DO. Oxygen regulates ILC3 antigen presentation potential and pregnancy-related hormone actions. Reprod Biol Endocrinol 2022; 20:109. [PMID: 35906658 PMCID: PMC9336067 DOI: 10.1186/s12958-022-00979-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/17/2022] [Accepted: 07/19/2022] [Indexed: 11/24/2022] Open
Abstract
Early pregnancy is marked by placentation and embryogenesis, which take place under physiological low oxygen concentrations. This oxygen condition is crucial for many aspects of placentation, trophoblast function, vascularization and immune function. Recently, a new family of innate lymphoid cells has been found to be expressed at the fetomaternal interface. Among these, type 3 innate lymphoid cells (ILC3) are important antigen presenting cells in the context of MHC-II. The expression of MHC-II on ILC3s during pregnancy is reduced. We tested the hypothesis that low oxygen concentrations reduce the potential of ILC3s to present antigens promoting fetal tolerance.Using an in vitro approach, NCR+ ILC3s generated from cord blood stem cell precursors were incubated under different O2 concentrations in the presence or absence of the pregnancy-related hormones hCG and TGF-β1. The expression of MHC-II, accessory molecules and an activation marker were assessed by flow cytometry. We observed that 1% O2 reduced the expression of the MHC-II molecule HLA-DR as compared to 21% O2 and modulated the relative effects of hCG and TGF-β1.Our data indicate that low oxygen concentrations reduce the antigen presentation potential of NCR+ ILC3s and suggest that it may promote fetal tolerance during the first trimester of pregnancy.
Collapse
Affiliation(s)
- Rebekka Einenkel
- Department of Obstetrics and Gynecology, University Medicine Greifswald, Greifswald, Germany
- Present address: Gynecologic Endocrinology and Reproductive Medicine, University Hospital Bonn, Bonn, Germany
| | - Jens Ehrhardt
- Department of Obstetrics and Gynecology, University Medicine Greifswald, Greifswald, Germany
| | - Marek Zygmunt
- Department of Obstetrics and Gynecology, University Medicine Greifswald, Greifswald, Germany
| | - Damián Oscar Muzzio
- Department of Obstetrics and Gynecology, University Medicine Greifswald, Greifswald, Germany.
| |
Collapse
|
47
|
Guo K, Huang J, Zhou Z. Host gene effects on gut microbiota in type 1 diabetes. Biochem Soc Trans 2022; 50:1133-1142. [PMID: 35521897 PMCID: PMC9246325 DOI: 10.1042/bst20220004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2022] [Revised: 04/07/2022] [Accepted: 04/12/2022] [Indexed: 12/03/2022]
Abstract
Type 1 diabetes (T1D) is an organ-specific autoimmune disease characterized by progressive pancreatic β-cell loss. Both a predisposing genetic background, that may encompass mutations in several genes, as well as exposure to environmental factors can affect the progression of autoimmune responses to multiple pancreatic islet autoantigens. Many genetic variants that increase the risk of T1D are found in immunity genes involved in sensing and responding to microorganisms. Although increasing evidence indicates that the gut microbiome composition may promote or prevent T1D development, little is known about the link between gut microbiota and T1D susceptibility genes in patients with T1D. Recent studies in the inbred non-obese diabetic (NOD) mouse, a widely used model of T1D, have suggested that many genetic loci can influence gut microbiome composition to modulate islet autoimmunity. This review summarizes evidence that examines the effect of host genes on gut microbiota diversity and function during T1D development. Knowledge of the host gene-gut microbiota interactions at play during T1D progression may help us identify new diagnostic and prognostic tools and help also design effective strategies for disease treatment.
Collapse
Affiliation(s)
- Keyu Guo
- National Clinical Research Center for Metabolic Diseases, Key Laboratory of Diabetes Immunology (Central South University), Ministry of Education, and Department of Metabolism and Endocrinology, The Second Xiangya Hospital of Central South University, Changsha, China
| | - Juan Huang
- National Clinical Research Center for Metabolic Diseases, Key Laboratory of Diabetes Immunology (Central South University), Ministry of Education, and Department of Metabolism and Endocrinology, The Second Xiangya Hospital of Central South University, Changsha, China
- Section of Endocrinology, Department of Internal Medicine, School of Medicine, Yale University, New Haven, CT, U.S.A
| | - Zhiguang Zhou
- National Clinical Research Center for Metabolic Diseases, Key Laboratory of Diabetes Immunology (Central South University), Ministry of Education, and Department of Metabolism and Endocrinology, The Second Xiangya Hospital of Central South University, Changsha, China
| |
Collapse
|
48
|
Extrathymic expression of Aire controls the induction of effective T H17 cell-mediated immune response to Candida albicans. Nat Immunol 2022; 23:1098-1108. [PMID: 35761088 DOI: 10.1038/s41590-022-01247-6] [Citation(s) in RCA: 38] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2019] [Accepted: 05/18/2022] [Indexed: 01/06/2023]
Abstract
Patients with loss of function in the gene encoding the master regulator of central tolerance AIRE suffer from a devastating disorder called autoimmune polyendocrine syndrome type 1 (APS-1), characterized by a spectrum of autoimmune diseases and severe mucocutaneous candidiasis. Although the key mechanisms underlying the development of autoimmunity in patients with APS-1 are well established, the underlying cause of the increased susceptibility to Candida albicans infection remains less understood. Here, we show that Aire+MHCII+ type 3 innate lymphoid cells (ILC3s) could sense, internalize and present C. albicans and had a critical role in the induction of Candida-specific T helper 17 (TH17) cell clones. Extrathymic Rorc-Cre-mediated deletion of Aire resulted in impaired generation of Candida-specific TH17 cells and subsequent overgrowth of C. albicans in the mucosal tissues. Collectively, our observations identify a previously unrecognized regulatory mechanism for effective defense responses against fungal infections.
Collapse
|
49
|
Fu W, Wang W, Zhang J, Zhao Y, Chen K, Wang Y, Zhang J, Xiong Y, Guo X, Ding S. Dynamic change of circulating innate and adaptive lymphocytes subtypes during a cascade of gastric lesions. J Leukoc Biol 2022; 112:931-938. [PMID: 35657091 DOI: 10.1002/jlb.5ma0422-505r] [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: 11/30/2021] [Revised: 04/28/2022] [Indexed: 11/07/2022] Open
Abstract
According to the Correa model, the intestinal-type gastric cancer (GC) is preceded by premalignant lesions, including chronic gastritis, intestinal metaplasia and dysplasia. However, the dynamic change of innate and adaptive immune response during this process has not been studied comprehensively. In this study, we performed a comprehensive and trajectory analysis of circulating innate lymphoid cells (ILCs) and adaptive Th lymphocytes subtypes in patients spanning a cascade of gastric lesions. Increased circulating ILC2s frequency was found in the gastritis, premalignant stage and GC group, whereas further decreased ILC2s were detected in the GC group compared with the premalignant group. Moreover, ILC3s level was higher in both gastritis, premalignant lesion and GC stage, compared with healthy controls. Furthermore, up-regulated T follicular helper (Tfh) cell proportions were detected in the gastritis and premalignant process. In conclusion, by analyzing the circulating ILCs and Th cells frequency and the key cytokine production or immunoglobulin level, we demonstrated the potential involvement of ILC3 and Tfh in the gastric diseases. These findings will help to understand the immunologic mechanisms in both GC and the premalignant process and contribute to serve potential therapeutic targets to prevent the GC development.
Collapse
Affiliation(s)
- Weiwei Fu
- Department of Gastroenterology, Peking University Third Hospital, Beijing, China.,Beijing Key Laboratory for Helicobacter Pylori Infection and Upper Gastrointestinal Diseases, Beijing, China
| | - Wenyan Wang
- Department of Basic Medical Sciences, School of Medicine, Tsinghua University, Beijing, China, Beijing, China.,Institute for Immunology, Tsinghua University, Beijing, China
| | - Jing Zhang
- Department of Gastroenterology, Peking University Third Hospital, Beijing, China.,Beijing Key Laboratory for Helicobacter Pylori Infection and Upper Gastrointestinal Diseases, Beijing, China
| | - Yang Zhao
- Department of Laboratory Medicine, Peking University Third Hospital, Beijing, China
| | - Keyan Chen
- Department of Gastroenterology, Peking University Third Hospital, Beijing, China.,Beijing Key Laboratory for Helicobacter Pylori Infection and Upper Gastrointestinal Diseases, Beijing, China
| | - Ye Wang
- Department of Gastroenterology, Peking University Third Hospital, Beijing, China.,Beijing Key Laboratory for Helicobacter Pylori Infection and Upper Gastrointestinal Diseases, Beijing, China
| | - Jing Zhang
- Department of Gastroenterology, Peking University Third Hospital, Beijing, China.,Beijing Key Laboratory for Helicobacter Pylori Infection and Upper Gastrointestinal Diseases, Beijing, China
| | - Ying Xiong
- Department of Gastroenterology, Peking University Third Hospital, Beijing, China.,Beijing Key Laboratory for Helicobacter Pylori Infection and Upper Gastrointestinal Diseases, Beijing, China
| | - Xiaohuan Guo
- Department of Basic Medical Sciences, School of Medicine, Tsinghua University, Beijing, China, Beijing, China.,Institute for Immunology, Tsinghua University, Beijing, China
| | - Shigang Ding
- Department of Gastroenterology, Peking University Third Hospital, Beijing, China.,Beijing Key Laboratory for Helicobacter Pylori Infection and Upper Gastrointestinal Diseases, Beijing, China
| |
Collapse
|
50
|
Wang R, Zhang J, Li D, Liu G, Fu Y, Li Q, Zhang L, Qian L, Hao L, Wang Y, Harris DCH, Wang D, Cao Q. Imbalance of circulating innate lymphoid cell subpopulations in patients with chronic kidney disease. Clin Immunol 2022; 239:109029. [PMID: 35525476 DOI: 10.1016/j.clim.2022.109029] [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: 07/22/2021] [Revised: 04/27/2022] [Accepted: 04/29/2022] [Indexed: 11/19/2022]
Abstract
Innate lymphoid cells (ILCs) are a newly identified heterogeneous family of innate immune cells. We conducted this study to investigate the frequency of circulating ILC subsets in various chronic kidney diseases (CKD). In DN, the proportion of total ILCs and certain ILC subgroups increased significantly. Positive correlations between proportion of total ILCs, ILC1s and body mass index, glycated hemoglobin were observed in DN. In LN, a significantly increased proportion of ILC1s was found in parallel with a reduced proportion of ILC2s. The proportions of total ILCs and ILC1s were correlated with WBC count and the level of C3. In all enrolled patients, the proportion of total ILCs and ILC1s was significantly correlated with the levels of ACR and GFR. In the present study, the proportion of circulating ILC subsets increased significantly in various types of CKD and correlated with clinico-pathological features, which suggests a possible role for ILCs in CKD.
Collapse
Affiliation(s)
- Ruifeng Wang
- Department of Nephrology, The Second Affiliated Hospital of Anhui Medical University, Hefei, China; Centre for Transplant and Renal Research, Westmead Institute for Medical Research, The University of Sydney, Sydney, NSW, Australia; Department of Nephrology, The Second Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Jingjing Zhang
- Department of Nephrology, The Second Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Dandan Li
- Department of Nephrology, The Second Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Guiling Liu
- Department of Nephrology, The Second Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Yuqin Fu
- Department of Nephrology, The Second Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Qing Li
- The Central Laboratory of Medical Research Center, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
| | - Lei Zhang
- Department of Rheumatology, The Second Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Long Qian
- Department of Rheumatology, The Second Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Li Hao
- Department of Nephrology, The Second Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Yiping Wang
- Centre for Transplant and Renal Research, Westmead Institute for Medical Research, The University of Sydney, Sydney, NSW, Australia
| | - David C H Harris
- Centre for Transplant and Renal Research, Westmead Institute for Medical Research, The University of Sydney, Sydney, NSW, Australia
| | - Deguang Wang
- Department of Nephrology, The Second Affiliated Hospital of Anhui Medical University, Hefei, China.
| | - Qi Cao
- Centre for Transplant and Renal Research, Westmead Institute for Medical Research, The University of Sydney, Sydney, NSW, Australia.
| |
Collapse
|