1
|
Yao X, Ma K, Zhu Y, Cao S. Innate Lymphoid Cells in Inflammatory Bowel Disease. Cells 2025; 14:825. [PMID: 40498001 PMCID: PMC12155180 DOI: 10.3390/cells14110825] [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: 04/21/2025] [Revised: 05/22/2025] [Accepted: 05/28/2025] [Indexed: 06/19/2025] Open
Abstract
Inflammatory bowel disease (IBD), including Crohn's disease and ulcerative colitis, is a chronic inflammatory disorder of the gastrointestinal tract with rising incidence and an unclear etiology. Innate lymphoid cells (ILCs) have recently emerged as key regulators of mucosal immunity and tissue homeostasis and are increasingly implicated in IBD. Unlike adaptive lymphocytes, ILCs do not require antigen recognition and clonal expansion to respond rapidly to environmental cues and shape immune responses. In a healthy gut, ILCs maintain intestinal homeostasis by guarding the epithelial barrier, protecting against pathogens, and mounting proper responses to external insults. However, their altered differentiation, proliferation, recruitment, activation, and interaction with other host cells, microbiota, and environmental stimuli may contribute to IBD. In this review, we discuss recent advances in understanding murine and human ILCs in the context of intestinal inflammation and IBD. A deeper understanding of ILC-mediated immune mechanisms may offer novel therapeutic strategies for restoring intestinal homeostasis and improving personalized management of IBD.
Collapse
Affiliation(s)
| | | | | | - Siyan Cao
- Division of Gastroenterology, Washington University School of Medicine, St. Louis, MO 63110, USA; (X.Y.); (K.M.); (Y.Z.)
| |
Collapse
|
2
|
Dai WB, Zhang X, Jiang XL, Zhang YZ, Chen LK, Tian WT, Zhou XX, Sun XY, Huang LL, Gu XY, Chen XM, Wu XD, Tian J, Yu WF, Shen L, Su DS. The kynurenine pathway regulated by intestinal innate lymphoid cells mediates postoperative cognitive dysfunction. Mucosal Immunol 2025; 18:53-65. [PMID: 39251184 DOI: 10.1016/j.mucimm.2024.09.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2024] [Revised: 08/28/2024] [Accepted: 09/03/2024] [Indexed: 09/11/2024]
Abstract
Postoperative cognitive dysfunction (POCD) is a prevalent neurological complication that can impair learning and memory for days, months, or even years after anesthesia/surgery. POCD is strongly associated with an altered composition of the gut microbiota (dysbiosis), but the accompanying metabolic changes and their role in gut-brain communication and POCD pathogenesis remain unclear. Here, the present study reports that anesthesia/surgery in aged mice induces elevated intestinal indoleamine 2,3-dioxygenase (IDO) expression and activity, which shifts intestinal tryptophan (TRP) metabolism toward more IDO-catalyzed kynurenine (KYN) and less gut bacteria-catabolized indoleacetic acid (IAA). Both anesthesia/surgery and intraperitoneal KYN administration induce increased KYN levels that correlate with impaired spatial learning and memory, whereas dietary IAA supplementation attenuates the anesthesia/surgery-induced cognitive impairment. Mechanistically, anesthesia/surgery increases interferon-γ (IFN-γ)-producing group 1 innate lymphoid cells (ILC1) in the small intestine lamina propria and elevates intestinal IDO expression and activity, as indicated by the higher ratio of KYN to TRP. The IDO inhibitor 1-MT and antibodies targeting IFN-γ or ILCs mitigate anesthesia/surgery-induced cognitive dysfunction, suggesting that intestinal ILC1 expansion and the ensuing IFN-γ-induced IDO upregulation may be the primary pathway mediating the shift to the KYN pathway in POCD. The ILC1-KYN pathway in the intestine could be a promising therapeutic target for POCD.
Collapse
Affiliation(s)
- Wan-Bing Dai
- Department of Anesthesiology, Renji Hospital, Shanghai Jiaotong University, School of Medicine, Shanghai, China; Key Laboratory of Anesthesiology (Shanghai Jiaotong University), Ministry of Education, China
| | - Xiao Zhang
- Department of Anesthesiology, Renji Hospital, Shanghai Jiaotong University, School of Medicine, Shanghai, China; Key Laboratory of Anesthesiology (Shanghai Jiaotong University), Ministry of Education, China
| | - Xu-Liang Jiang
- Department of Anesthesiology, Renji Hospital, Shanghai Jiaotong University, School of Medicine, Shanghai, China; Key Laboratory of Anesthesiology (Shanghai Jiaotong University), Ministry of Education, China
| | - Yi-Zhe Zhang
- Department of Anesthesiology, Renji Hospital, Shanghai Jiaotong University, School of Medicine, Shanghai, China; Key Laboratory of Anesthesiology (Shanghai Jiaotong University), Ministry of Education, China
| | - Ling-Ke Chen
- Department of Anesthesiology, Renji Hospital, Shanghai Jiaotong University, School of Medicine, Shanghai, China; Key Laboratory of Anesthesiology (Shanghai Jiaotong University), Ministry of Education, China
| | - Wei-Tian Tian
- Department of Anesthesiology, Renji Hospital, Shanghai Jiaotong University, School of Medicine, Shanghai, China; Key Laboratory of Anesthesiology (Shanghai Jiaotong University), Ministry of Education, China
| | - Xiao-Xin Zhou
- Department of Anesthesiology, Renji Hospital, Shanghai Jiaotong University, School of Medicine, Shanghai, China; Key Laboratory of Anesthesiology (Shanghai Jiaotong University), Ministry of Education, China
| | - Xiao-Yu Sun
- Department of Anesthesiology, Renji Hospital, Shanghai Jiaotong University, School of Medicine, Shanghai, China; Key Laboratory of Anesthesiology (Shanghai Jiaotong University), Ministry of Education, China
| | - Li-Li Huang
- Department of Anesthesiology, Renji Hospital, Shanghai Jiaotong University, School of Medicine, Shanghai, China; Key Laboratory of Anesthesiology (Shanghai Jiaotong University), Ministry of Education, China
| | - Xi-Yao Gu
- Department of Anesthesiology, Renji Hospital, Shanghai Jiaotong University, School of Medicine, Shanghai, China; Key Laboratory of Anesthesiology (Shanghai Jiaotong University), Ministry of Education, China
| | - Xue-Mei Chen
- Department of Anesthesiology, Renji Hospital, Shanghai Jiaotong University, School of Medicine, Shanghai, China; Key Laboratory of Anesthesiology (Shanghai Jiaotong University), Ministry of Education, China
| | - Xiao-Dan Wu
- Department of Anesthesiology, Shengli Clinical Medical College of Fujian Medical University, Fujian Provincial Hospital, Fujian, China
| | - Jie Tian
- Department of Anesthesiology, Renji Hospital, Shanghai Jiaotong University, School of Medicine, Shanghai, China; Key Laboratory of Anesthesiology (Shanghai Jiaotong University), Ministry of Education, China
| | - Wei-Feng Yu
- Department of Anesthesiology, Renji Hospital, Shanghai Jiaotong University, School of Medicine, Shanghai, China; Key Laboratory of Anesthesiology (Shanghai Jiaotong University), Ministry of Education, China.
| | - Lei Shen
- Shanghai institute of Immunology, Department of Immunology and Microbiology, and Key Laboratory of Cell Differentiation and Apoptosis of the Chinese Ministry of Education, Shanghai Jiaotong University, School of Medicine, Shanghai, China; Shanghai Key Laboratory of Tumor Microenvironment and Inflammation, Shanghai Jiaotong University, School of Medicine, Shanghai, China.
| | - Dian-San Su
- Department of Anesthesiology, Renji Hospital, Shanghai Jiaotong University, School of Medicine, Shanghai, China; Key Laboratory of Anesthesiology (Shanghai Jiaotong University), Ministry of Education, China; Department of Anesthesiology, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China.
| |
Collapse
|
3
|
Deng Z, Li L, Meng Z, Zeng G, Cao R, Liu R. Pan-cancer analysis shows that TNFSF4 is a potential prognostic and immunotherapeutic biomarker for multiple cancer types including liver cancer. BMC Cancer 2025; 25:100. [PMID: 39825223 PMCID: PMC11740393 DOI: 10.1186/s12885-025-13479-4] [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: 04/12/2024] [Accepted: 01/08/2025] [Indexed: 01/20/2025] Open
Abstract
BACKGROUND As a member of the tumor necrosis factor (TNF) superfamily, tumor necrosis factor superfamily member 4 (TNFSF4) is expressed on antigen-presenting cells and activated T cells by binding to its receptor TNFRSF4. However, tumorigenicity of TNFSF4 has not been studied in pan-cancer. Therefore, comprehensive bioinformatics analysis of pan-cancer was performed to determine the mechanisms through which TNFSF4 regulates tumorigenesis. METHODS RNA-seq data for 33 cancers were analyzed from UCSC XENA database. Online websites and databases were used to investigate TNFSF4's biological function, epigenetic modifications, genetic alterations, and tumor immunity. Furthermore, cell phenotype experiment and tumor xenotransplantation experiment were performed to determine the biological functions of TNFSF4. RESULTS The pan-cancer analysis showed that TNFSF4 was upregulated in several tumors. Significant relationships between TNFSF4 expression and single-cells data were also observed in numerous cancer types. TNFSF4 expression correlated with the expression of immune checkpoint genes and could influence various drug sensitivity. Vitro and vivo experiments showed that TNFSF4 could promote the development and progression of Hepatocellular Carcinoma (HCC). CONCLUSIONS TNFSF4 was upregulated in multiple cancer types and promoted the development and progression of cancers through several mechanisms including regulation of the tumor-infiltration of immune cells. Our study shows that TNFSF4 is a promising prognostic and immunotherapeutic biomarker in some malignant tumors.
Collapse
Affiliation(s)
- Zhaoda Deng
- Medical School of Chinese PLA, Beijing, China
- Faculty of Hepato-Pancreato-Biliary Surgery, The First Medical Center, Chinese PLA General Hospital, Institute of Hepatobiliary Surgery of Chinese PLA, Beijing, China
- Key Laboratory of Digital Hepatobiliary Surgery, PLA, Beijing, China
| | - Lincheng Li
- Department of Surgery, Second Mobile Corps Hospital of Chinese People's Armed Police Force, Wuxi, China
| | - Zihe Meng
- Inner Mongolia Medical University, Hohhot, China
| | - Guineng Zeng
- Faculty of Hepato-Pancreato-Biliary Surgery, The First Medical Center, Chinese PLA General Hospital, Institute of Hepatobiliary Surgery of Chinese PLA, Beijing, China
- Key Laboratory of Digital Hepatobiliary Surgery, PLA, Beijing, China
- Nankai University School of Medicine, Nankai University, Tianjin, 300300, China
| | - Rui Cao
- Inner Mongolia Medical University, Hohhot, China
| | - Rong Liu
- Medical School of Chinese PLA, Beijing, China.
- Faculty of Hepato-Pancreato-Biliary Surgery, The First Medical Center, Chinese PLA General Hospital, Institute of Hepatobiliary Surgery of Chinese PLA, Beijing, China.
- Key Laboratory of Digital Hepatobiliary Surgery, PLA, Beijing, China.
| |
Collapse
|
4
|
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
|
5
|
Stockis J, Yip T, Moreno-Vicente J, Burton O, Samarakoon Y, Schuijs MJ, Raghunathan S, Garcia C, Luo W, Whiteside SK, Png S, Simpson C, Monk S, Sawle A, Yin K, Barbieri J, Papadopoulos P, Wong H, Rodewald HR, Vyse T, McKenzie ANJ, Cragg MS, Hoare M, Withers DR, Fehling HJ, Roychoudhuri R, Liston A, Halim TYF. Cross-talk between ILC2 and Gata3 high T regs locally constrains adaptive type 2 immunity. Sci Immunol 2024; 9:eadl1903. [PMID: 39028828 DOI: 10.1126/sciimmunol.adl1903] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2023] [Accepted: 06/26/2024] [Indexed: 07/21/2024]
Abstract
Regulatory T cells (Tregs) control adaptive immunity and restrain type 2 inflammation in allergic disease. Interleukin-33 promotes the expansion of tissue-resident Tregs and group 2 innate lymphoid cells (ILC2s); however, how Tregs locally coordinate their function within the inflammatory niche is not understood. Here, we show that ILC2s are critical orchestrators of Treg function. Using spatial, cellular, and molecular profiling of the type 2 inflamed niche, we found that ILC2s and Tregs engage in a direct (OX40L-OX40) and chemotaxis-dependent (CCL1-CCR8) cellular dialogue that enforces the local accumulation of Gata3high Tregs, which are transcriptionally and functionally adapted to the type 2 environment. Genetic interruption of ILC2-Treg communication resulted in uncontrolled type 2 lung inflammation after allergen exposure. Mechanistically, we found that Gata3high Tregs can modulate the local bioavailability of the costimulatory molecule OX40L, which subsequently controlled effector memory T helper 2 cell numbers. Hence, ILC2-Treg interactions represent a critical feedback mechanism to control adaptive type 2 immunity.
Collapse
Affiliation(s)
- Julie Stockis
- CRUK Cambridge Institute, University of Cambridge, Cambridge CB2 0RE, UK
| | - Thomas Yip
- CRUK Cambridge Institute, University of Cambridge, Cambridge CB2 0RE, UK
| | | | - Oliver Burton
- Immunology Programme, Babraham Institute, Cambridge CB22 3AT, UK
- Department of Pathology, University of Cambridge, Cambridge CB2 1QP, UK
| | - Youhani Samarakoon
- CRUK Cambridge Institute, University of Cambridge, Cambridge CB2 0RE, UK
| | - Martijn J Schuijs
- CRUK Cambridge Institute, University of Cambridge, Cambridge CB2 0RE, UK
| | | | - Celine Garcia
- CRUK Cambridge Institute, University of Cambridge, Cambridge CB2 0RE, UK
| | - Weike Luo
- CRUK Cambridge Institute, University of Cambridge, Cambridge CB2 0RE, UK
| | - Sarah K Whiteside
- Department of Pathology, University of Cambridge, Cambridge CB2 1QP, UK
| | - Shaun Png
- CRUK Cambridge Institute, University of Cambridge, Cambridge CB2 0RE, UK
| | - Charlotte Simpson
- CRUK Cambridge Institute, University of Cambridge, Cambridge CB2 0RE, UK
| | - Stela Monk
- CRUK Cambridge Institute, University of Cambridge, Cambridge CB2 0RE, UK
| | - Ashley Sawle
- CRUK Cambridge Institute, University of Cambridge, Cambridge CB2 0RE, UK
| | - Kelvin Yin
- CRUK Cambridge Institute, University of Cambridge, Cambridge CB2 0RE, UK
| | - Johanna Barbieri
- CRUK Cambridge Institute, University of Cambridge, Cambridge CB2 0RE, UK
| | | | - Hannah Wong
- Department of Veterinary Medicine, University of Cambridge, Cambridge CB3 0ES, UK
| | - Hans-Reimer Rodewald
- Division of Cellular Immunology, German Cancer Research Center, Heidelberg 69120, Germany
| | - Timothy Vyse
- Department of Medical and Molecular Genetics, King's College London, London SE1 9RT, UK
| | - Andrew N J McKenzie
- Medical Research Council, Laboratory of Molecular Biology, Cambridge CB2 0QH, UK
| | - Mark S Cragg
- Antibody and Vaccine Group, Centre for Cancer Immunology, School of Cancer Sciences, Faculty of Medicine, University of Southampton, Southampton SO16 6YD, UK
| | - Matthew Hoare
- CRUK Cambridge Institute, University of Cambridge, Cambridge CB2 0RE, UK
- Department of Medicine, University of Cambridge, Cambridge CB2 0QQ, UK
- Early Cancer Institute, Hutchison Research Centre, University of Cambridge, Cambridge CB2 0XZ, UK
| | - David R Withers
- Institute of Immunology and Immunotherapy, University of Birmingham, Birmingham B15 2TT, UK
| | - Hans Jörg Fehling
- Institute of Immunology, University Hospital Ulm, Ulm 89081, Germany
| | | | - Adrian Liston
- Immunology Programme, Babraham Institute, Cambridge CB22 3AT, UK
- Department of Pathology, University of Cambridge, Cambridge CB2 1QP, UK
| | | |
Collapse
|
6
|
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: 11] [Impact Index Per Article: 11.0] [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
|
7
|
Ramanan D, Pratama A, Zhu Y, Venezia O, Sassone-Corsi M, Chowdhary K, Galván-Peña S, Sefik E, Brown C, Gélineau A, Mathis D, Benoist C. Regulatory T cells in the face of the intestinal microbiota. Nat Rev Immunol 2023; 23:749-762. [PMID: 37316560 DOI: 10.1038/s41577-023-00890-w] [Citation(s) in RCA: 49] [Impact Index Per Article: 24.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/10/2023] [Indexed: 06/16/2023]
Abstract
Regulatory T cells (Treg cells) are key players in ensuring a peaceful coexistence with microorganisms and food antigens at intestinal borders. Startling new information has appeared in recent years on their diversity, the importance of the transcription factor FOXP3, how T cell receptors influence their fate and the unexpected and varied cellular partners that influence Treg cell homeostatic setpoints. We also revisit some tenets, maintained by the echo chambers of Reviews, that rest on uncertain foundations or are a subject of debate.
Collapse
Affiliation(s)
| | - Alvin Pratama
- Department of Immunology, Harvard Medical School, Boston, MA, USA
| | - Yangyang Zhu
- Department of Immunology, Harvard Medical School, Boston, MA, USA
| | - Olivia Venezia
- Department of Immunology, Harvard Medical School, Boston, MA, USA
| | | | | | | | - Esen Sefik
- Department of Immunology, Yale University, New Haven, CT, USA
| | - Chrysothemis Brown
- Immuno-Oncology, Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Department of Paediatrics, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Immunology and Microbial Pathogenesis Program, Weill Cornell Medicine Graduate School of Medical Sciences, New York, NY, USA
| | | | - Diane Mathis
- Department of Immunology, Harvard Medical School, Boston, MA, USA
| | | |
Collapse
|
8
|
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
|
9
|
Ryu S, Lim M, Kim J, Kim HY. Versatile roles of innate lymphoid cells at the mucosal barrier: from homeostasis to pathological inflammation. Exp Mol Med 2023; 55:1845-1857. [PMID: 37696896 PMCID: PMC10545731 DOI: 10.1038/s12276-023-01022-z] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2023] [Revised: 03/31/2023] [Accepted: 04/04/2023] [Indexed: 09/13/2023] Open
Abstract
Innate lymphoid cells (ILCs) are innate lymphocytes that do not express antigen-specific receptors and largely reside and self-renew in mucosal tissues. ILCs can be categorized into three groups (ILC1-3) based on the transcription factors that direct their functions and the cytokines they produce. Their signature transcription factors and cytokines closely mirror those of their Th1, Th2, and Th17 cell counterparts. Accumulating studies show that ILCs are involved in not only the pathogenesis of mucosal tissue diseases, especially respiratory diseases, and colitis, but also the resolution of such diseases. Here, we discuss recent advances regarding our understanding of the biology of ILCs in mucosal tissue health and disease. In addition, we describe the current research on the immune checkpoints by which other cells regulate ILC activities: for example, checkpoint molecules are potential new targets for therapies that aim to control ILCs in mucosal diseases. In addition, we review approved and clinically- trialed drugs and drugs in clinical trials that can target ILCs and therefore have therapeutic potential in ILC-mediated diseases. Finally, since ILCs also play important roles in mucosal tissue homeostasis, we explore the hitherto sparse research on cell therapy with regulatory ILCs. This review highlights various therapeutic approaches that could be used to treat ILC-mediated mucosal diseases and areas of research that could benefit from further investigation.
Collapse
Affiliation(s)
- Seungwon Ryu
- Department of Microbiology, Gachon University College of Medicine, Incheon, 21999, South Korea
| | - MinYeong Lim
- Laboratory of Mucosal Immunology, Department of Biomedical Sciences, Seoul National University College of Medicine, Seoul, 03080, South Korea
- Institute of Allergy and Clinical Immunology, Seoul National University Medical Research Center, Seoul, South Korea
- CIRNO, Sungkyunkwan University, Suwon, South Korea
| | - Jinwoo Kim
- Laboratory of Mucosal Immunology, Department of Biomedical Sciences, Seoul National University College of Medicine, Seoul, 03080, South Korea
- Institute of Allergy and Clinical Immunology, Seoul National University Medical Research Center, Seoul, South Korea
- CIRNO, Sungkyunkwan University, Suwon, South Korea
| | - Hye Young Kim
- Laboratory of Mucosal Immunology, Department of Biomedical Sciences, Seoul National University College of Medicine, Seoul, 03080, South Korea.
- Institute of Allergy and Clinical Immunology, Seoul National University Medical Research Center, Seoul, South Korea.
- CIRNO, Sungkyunkwan University, Suwon, South Korea.
| |
Collapse
|
10
|
Xie J, Gu A, He H, Zhao Q, Yu Y, Chen J, Cheng Z, Zhou P, Zhou Q, Jin M. Autoimmune thyroid disease disrupts immune homeostasis in the endometrium of unexplained infertility women-a single-cell RNA transcriptome study during the implantation window. Front Endocrinol (Lausanne) 2023; 14:1185147. [PMID: 37501789 PMCID: PMC10368980 DOI: 10.3389/fendo.2023.1185147] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/13/2023] [Accepted: 05/23/2023] [Indexed: 07/29/2023] Open
Abstract
Objective Autoimmune thyroid disease (AITD) is known to be associated with unexplained infertility in women. Although the presence of antithyroid antibodies have been speculated to be a marker of an immune imbalance that might lead to implantation failure, its underlying mechanism influencing the endometrial receptivity remains to be elucidated. In this study, we used single-cell RNA sequencing (scRNA-seq) to dissect immune microenvironment in endometrium of AITD patients during window of implantation (WOI). Methods We collected CD45+ immune cell populations of endometrium samples of unexplained infertile women with AITD (n=3), as well as samples of AITD- controls (n=3). The cells were then processed with 10X Genomics Chromium for further analysis. Results We characterized 28 distinct immune cell subtypes totally, and uncovered differences in the composition and gene expression patterns between AITD patients and controls. The proportions of T CD4+, cNK, ILC3, T CD8+ GZMK+, T CD8+ Cytotoxic and ILC3 CD3E - cells were increased, and CD366+ uNK1 was decreased in AITD+ patients. And the abnormal expression of GNLY and chemokines was observed in AITD patients. In addition, uNK and T CD8+ Cytotoxic cells showed lower cytotoxicity but activation of immune response. Genes enriched in cell adhesion of ILC3 and Tregs were downregulated, while the number of ILC3 and Tregs were increased. Conclusion Immune imbalance exists in endometrium during WOI, which may impact embryo implantation.
Collapse
Affiliation(s)
- Jilai Xie
- Second Affiliated Hospital, School of Medicine, Zhejiang University, Department of Reproductive Medicine, Hangzhou, China
| | - Aiyuan Gu
- Ministry of Education (MOE) Laboratory of Biosystems Homeostasis & Protection and Zhejiang Provincial Key Laboratory for Cancer Molecular Cell Biology, Life Sciences Institute, Zhejiang University, Hangzhou, China
| | - Huangyi He
- Ministry of Education (MOE) Laboratory of Biosystems Homeostasis & Protection and Zhejiang Provincial Key Laboratory for Cancer Molecular Cell Biology, Life Sciences Institute, Zhejiang University, Hangzhou, China
| | - Qiaohang Zhao
- Second Affiliated Hospital, School of Medicine, Zhejiang University, Department of Reproductive Medicine, Hangzhou, China
| | - Ya Yu
- Second Affiliated Hospital, School of Medicine, Zhejiang University, Department of Reproductive Medicine, Hangzhou, China
| | - Jian Chen
- Second Affiliated Hospital, School of Medicine, Zhejiang University, Department of Reproductive Medicine, Hangzhou, China
| | - Zhangliang Cheng
- Ministry of Education (MOE) Laboratory of Biosystems Homeostasis & Protection and Zhejiang Provincial Key Laboratory for Cancer Molecular Cell Biology, Life Sciences Institute, Zhejiang University, Hangzhou, China
| | - Ping Zhou
- Second Affiliated Hospital, School of Medicine, Zhejiang University, Department of Reproductive Medicine, Hangzhou, China
| | - Qi Zhou
- Second Affiliated Hospital, School of Medicine, Zhejiang University, Department of Reproductive Medicine, Hangzhou, China
- Ministry of Education (MOE) Laboratory of Biosystems Homeostasis & Protection and Zhejiang Provincial Key Laboratory for Cancer Molecular Cell Biology, Life Sciences Institute, Zhejiang University, Hangzhou, China
- Department of Neuroscience and Developmental Biology, University of Vienna, Vienna, Austria
| | - Min Jin
- Second Affiliated Hospital, School of Medicine, Zhejiang University, Department of Reproductive Medicine, Hangzhou, China
| |
Collapse
|
11
|
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
|
12
|
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
|
13
|
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
|
14
|
Hoffman K, Brownell Z, Doyle WJ, Ochoa-Repáraz J. The immunomodulatory roles of the gut microbiome in autoimmune diseases of the central nervous system: Multiple sclerosis as a model. J Autoimmun 2023; 137:102957. [PMID: 36435700 PMCID: PMC10203067 DOI: 10.1016/j.jaut.2022.102957] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2022] [Accepted: 11/14/2022] [Indexed: 11/24/2022]
Abstract
The gut-associated lymphoid tissue is a primary activation site for immune responses to infection and immunomodulation. Experimental evidence using animal disease models suggests that specific gut microbes significantly regulate inflammation and immunoregulatory pathways. Furthermore, recent clinical findings indicate that gut microbes' composition, collectively named gut microbiota, is altered under disease state. This review focuses on the functional mechanisms by which gut microbes promote immunomodulatory responses that could be relevant in balancing inflammation associated with autoimmunity in the central nervous system. We also propose therapeutic interventions that target the composition of the gut microbiota as immunomodulatory mechanisms to control neuroinflammation.
Collapse
Affiliation(s)
- Kristina Hoffman
- Department of Biological Sciences, Boise State University, Boise, ID, 83725, USA
| | - Zackariah Brownell
- Department of Biological Sciences, Arizona State University, Tempe, AZ, 85281, USA
| | - William J Doyle
- Department of Biological Sciences, Boise State University, Boise, ID, 83725, USA
| | - Javier Ochoa-Repáraz
- Department of Biological Sciences, Boise State University, Boise, ID, 83725, USA.
| |
Collapse
|
15
|
Iriki H, Takahashi H, Amagai M. Diverse Role of OX40 on T Cells as a Therapeutic Target for Skin Diseases. J Invest Dermatol 2023; 143:545-553. [PMID: 36842860 DOI: 10.1016/j.jid.2022.11.009] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2022] [Revised: 11/22/2022] [Accepted: 11/24/2022] [Indexed: 02/26/2023]
Abstract
OX40 is an important costimulatory molecule for T-cell expansion and survival. Because OX40 is expressed on most T-cell subsets, it is an attractive therapeutic target for a variety of T-cell‒mediated diseases. Clinical trials are already underway for some skin inflammatory diseases. In this review, we present various observations that improve our understanding of how OX40-targeted therapy can be applied for skin inflammatory diseases, such as atopic dermatitis and psoriasis, T helper (Th)2- and Th17-mediated diseases, respectively. The important OX40/OX40L-mediated interaction between T cells and other immune cells is also discussed in terms of skin autoimmune diseases, such as alopecia areata and pemphigus. Regulatory T cells (Tregs) highly express OX40, and the skin harbors a large Treg population; thus, understanding how OX40-targeted treatment acts on Tregs is vital for the development of therapeutic strategies for various skin diseases.
Collapse
Affiliation(s)
- Hisato Iriki
- Department of Dermatology, Keio University School of Medicine, Tokyo, Japan
| | - Hayato Takahashi
- Department of Dermatology, Keio University School of Medicine, Tokyo, Japan
| | - Masayuki Amagai
- Department of Dermatology, Keio University School of Medicine, Tokyo, Japan; Laboratory for Skin Homeostasis, RIKEN Center for Integrative Medical Sciences, Yokohama, Japan.
| |
Collapse
|
16
|
Lv X, Zhu S, Wu J, Chen J. Group 3 innate lymphoid cells: intestinal patrolling guardians bullied by T cells. Cell Mol Immunol 2023; 20:115-116. [PMID: 36385184 PMCID: PMC9886995 DOI: 10.1038/s41423-022-00948-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2022] [Accepted: 10/28/2022] [Indexed: 11/17/2022] Open
Affiliation(s)
- Xinping Lv
- Cancer Center, First Hospital of Jilin University, Changchun, 130021, Jilin, China
- Laboratory for Tumor Immunology, First Hospital of Jilin University, Changchun, 130061, Jilin, China
| | - Shan Zhu
- Cancer Center, First Hospital of Jilin University, Changchun, 130021, Jilin, China
- Laboratory for Tumor Immunology, First Hospital of Jilin University, Changchun, 130061, Jilin, China
| | - Jing Wu
- Cancer Center, First Hospital of Jilin University, Changchun, 130021, Jilin, China
- Laboratory for Tumor Immunology, First Hospital of Jilin University, Changchun, 130061, Jilin, China
| | - Jingtao Chen
- Cancer Center, First Hospital of Jilin University, Changchun, 130021, Jilin, China.
- Laboratory for Tumor Immunology, First Hospital of Jilin University, Changchun, 130061, Jilin, China.
| |
Collapse
|
17
|
Pan ZY, Liu HQ, Zhuang YP, Tan HB, Yang XY, Zhong HJ, He XX. Reduced type 3 innate lymphoid cells related to worsening kidney function in renal dysfunction. Exp Biol Med (Maywood) 2023; 248:242-252. [PMID: 36670544 PMCID: PMC10107398 DOI: 10.1177/15353702221147561] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2022] [Accepted: 11/22/2022] [Indexed: 01/22/2023] Open
Abstract
Intestinal mucosa barrier injury and immunity imbalance contribute to chronic kidney disease (CKD) progression. Type 3 innate lymphoid cells (ILC3s) are essential for normal intestinal homeostasis. Nevertheless, the relationship between ILC3s and CKD remains largely unknown. The aim of this study was to investigate the relationship linking ILC3s to clinical indicators among patients with renal dysfunction. The levels of circulating ILC3s and dendritic cells, as well as their subsets, in patients with renal dysfunction and healthy controls were determined through flow cytometry. The levels of human plasma granulocyte-macrophage colony-stimulating factor (GM-CSF) were measured using enzyme-linked immunosorbent assay. Renal function was evaluated by measuring the estimated glomerular filtration rate (eGFR), as well as the levels of serum creatinine, blood urea nitrogen (BUN), and uric acid. The results revealed that the proportion of peripheral ILC3s was significantly decreased in patients with renal dysfunction. This reduction was positively associated with the levels of eGFR, and inversely associated with the levels of BUN and uric acid. Similarly, the percentage of circulating C-C motif chemokine receptor 6-positive (CCR6 +) ILC3s was also obviously reduced, and demonstrated positive and negative associations with the levels of eGFR and BUN, respectively. Furthermore, the levels of CCR6 + ILC3s correlated positively with those of GM-CSF, as well as type 1 conventional dendritic cells (cDC1s), which also decreased in parallel with kidney function. Thus, the reduction of ILC3s, particularly CCR6 + ILC3s, was related to worsening kidney function in patients with renal dysfunction. This effect may delay renal function impairment by regulating cDC1s via the secretion of GM-CSF, indicating that CCR6 + ILC3s may serve as efficient biomarkers for evaluating kidney function.
Collapse
Affiliation(s)
- Zhao-Yu Pan
- Department of Gastroenterology, The
First Affiliated Hospital of Guangdong Pharmaceutical University, Guangzhou 510080,
China
| | - Hong-Qian Liu
- Department of Gastroenterology, The
First Affiliated Hospital of Guangdong Pharmaceutical University, Guangzhou 510080,
China
| | - Yu-Pei Zhuang
- Department of Gastroenterology, The
First Affiliated Hospital of Guangdong Pharmaceutical University, Guangzhou 510080,
China
- The First Clinical Medical College,
Nanjing University of Chinese Medicine, Nanjing 210023, China
| | - Hai-Bo Tan
- Shenzhen Traditional Chinese Medicine
Hospital Affiliated to Nanjing University of Chinese Medicine, Shenzhen 518033,
China
| | - Xiao-Ya Yang
- Department of Physiology, Guangzhou
Health Science College, Guangzhou 510450, China
| | - Hao-Jie Zhong
- Department of Gastroenterology, The
First Affiliated Hospital of Guangdong Pharmaceutical University, Guangzhou 510080,
China
- School of Biology and Biological
Engineering, South China University of Technology, Guangzhou 510080, China
| | - Xing-Xiang He
- Department of Gastroenterology, The
First Affiliated Hospital of Guangdong Pharmaceutical University, Guangzhou 510080,
China
- Xing-Xiang He.
| |
Collapse
|
18
|
Song P, Cao K, Mao Y, Ai S, Sun F, Hu Q, Liu S, Wang M, Lu X, Guan W, Shen X. Tissue specific imprinting on innate lymphoid cells during homeostasis and disease process revealed by integrative inference of single-cell transcriptomics. Front Immunol 2023; 14:1127413. [PMID: 36960063 PMCID: PMC10028295 DOI: 10.3389/fimmu.2023.1127413] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2022] [Accepted: 02/20/2023] [Indexed: 03/09/2023] Open
Abstract
Introduction Innate lymphoid cells (ILCs) are key components of the immune system, yet the similarity and distinction of the properties across tissues under homeostasis, inflammation and tumor process remain elusive. Methods Here we performed integrative inference of ILCs to reveal their transcriptional profiles and heterogeneity from single-cell genomics. We collected a large number of ILCs from human six different tissues which can represent unique immune niches (circulation, lymphoid tissue, normal and inflamed mucosa, tumor microenvironment), to systematically address the transcriptional imprinting. Results ILCs are profoundly imprinted by their organ of residence, and tissue-specific distinctions are apparent under pathological conditions. In the hepatocellular carcinoma microenvironment, we identified intermediate c-kit+ ILC2 population, and lin-CD127- NK-like cells that expressed markers of cytotoxicity including CCL5 and IFNG. Additionally, CD127+CD94+ ILC1s were preferentially enriched in inflamed ileum from patients with Crohn's disease. Discussion These analyses depicted a comprehensive characterization of ILC anatomical distribution and subset heterogeneity, and provided a base line for future temporal or spatial studies focused on tissue-specific ILC-mediated immunity.
Collapse
Affiliation(s)
- Peng Song
- Department of Gastrointestinal Surgery, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, China
- Department of Gastrointestinal Surgery, Nanjing Drum Tower Hospital, Drum Tower Clinical Medical College of Nanjing Medical University, Nanjing, China
| | - Ke Cao
- Department of Critical Care Medicine, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, China
| | - Yonghuan Mao
- Department of Gastrointestinal Surgery, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, China
- Department of Gastrointestinal Surgery, Nanjing Drum Tower Hospital, Drum Tower Clinical Medical College of Nanjing Medical University, Nanjing, China
| | - Shichao Ai
- Department of Gastrointestinal Surgery, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, China
| | - Feng Sun
- Department of Gastrointestinal Surgery, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, China
| | - Qiongyuan Hu
- Department of Gastrointestinal Surgery, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, China
| | - Song Liu
- Department of Gastrointestinal Surgery, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, China
| | - Meng Wang
- Department of Gastrointestinal Surgery, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, China
| | - Xiaofeng Lu
- Department of Gastrointestinal Surgery, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, China
- *Correspondence: Xiaofei Shen, ; Wenxian Guan, ; Xiaofeng Lu,
| | - Wenxian Guan
- Department of Gastrointestinal Surgery, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, China
- *Correspondence: Xiaofei Shen, ; Wenxian Guan, ; Xiaofeng Lu,
| | - Xiaofei Shen
- Department of Gastrointestinal Surgery, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, China
- Department of Gastrointestinal Surgery, Nanjing Drum Tower Hospital, Drum Tower Clinical Medical College of Nanjing Medical University, Nanjing, China
- *Correspondence: Xiaofei Shen, ; Wenxian Guan, ; Xiaofeng Lu,
| |
Collapse
|
19
|
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
|
20
|
Curio S, Belz GT. ZBTB46 in ILC3: shared transcriptional infrastructure defines gut-protective capabilities. Trends Immunol 2022; 43:690-692. [PMID: 35953346 DOI: 10.1016/j.it.2022.07.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2022] [Accepted: 07/29/2022] [Indexed: 11/16/2022]
Abstract
Group 3 innate lymphoid cells (ILC3s) are distributed along the gastrointestinal tract at the interface between the immune system and the gut lumen, which carries a significant microbial burden. In a new study, Zhou et al. investigated the expression of transcription factor ZBTB46, normally thought to be restricted to classical dendritic cells (cDCs), and discovered that ZBTB46 expression by ILC3s in the mouse colon forms an essential part of the gastrointestinal armory to calibrate inflammatory responses.
Collapse
Affiliation(s)
- Sophie Curio
- The University of Queensland Diamantina Institute, Faculty of Medicine, The University of Queensland, Woolloongabba, QLD 4102, Australia
| | - Gabrielle T Belz
- The University of Queensland Diamantina Institute, Faculty of Medicine, The University of Queensland, Woolloongabba, QLD 4102, Australia.
| |
Collapse
|
21
|
Queiroz-Glauss CP, Vieira MS, Gonçalves-Pereira MH, Almeida SS, Freire RH, Gomes MA, Alvarez-Leite JI, Santiago HC. Helminth infection modulates number and function of adipose tissue Tregs in high fat diet-induced obesity. PLoS Negl Trop Dis 2022; 16:e0010105. [PMID: 35499991 PMCID: PMC9098094 DOI: 10.1371/journal.pntd.0010105] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2021] [Revised: 05/12/2022] [Accepted: 04/01/2022] [Indexed: 11/23/2022] Open
Abstract
Background Epidemiological and experimental studies have shown a protective effect of helminth infections in weight gain and against the development of metabolic dysfunctions in the host. However, the mechanisms Treg cells exert in the helminth-obesity interface has been poorly investigated. The present study aimed to verify the influence of Heligmosomoides polygyrus infection in early stages of high fat diet-induced obesity. Principal findings The presence of infection was able to prevent exacerbated weight gain in mice fed with high fat diet when compared to non-infected controls. In addition, infected animals displayed improved insulin sensitivity and decreased fat accumulation in the liver. Obesity-associated inflammation was reduced in the presence of infection, demonstrated by lower levels of leptin and resistin, lower infiltration of Th1 and Th17 cells in adipose tissue, higher expression of IL10 and adiponectin, increased infiltration of Th2 and eosinophils in adipose tissue of infected animals. Of note, the parasite infection was associated with increased Treg frequency in adipose tissue which showed higher expression of cell surface markers of function and activation, like LAP and CD134. The infection could also increase adipose Treg suppressor function in animals on high fat diet. Conclusion These data suggest that H. polygyrus modulates adipose tissue Treg cells with implication for weight gain and metabolic syndrome. Helminth infections are known to modulate the immune system being responsible for protecting the host from developing allergic and autoimmune disorders (Hygiene Hypothesis). We hypothesized that the same immunomodulatory effect could have an impact on immunometabolic diseases, such as obesity and its linked diseases such as type 2 diabetes. Weight disorders have reached epidemic levels, nearly tripling since 1975 and being responsible for almost 5 million premature deaths each year, but have been spared in areas of high helminth prevalence. To test our hypothesis C57BL/6 male mice were fed control or high fat diet, for five weeks, in the presence or not of infection with the worm Heligmosomoides polygyrus. Weight gain, development of metabolic disorders, inflammation and cellular migration to the adipose tissue were evaluated. In accordance with our hypothesis, we found that the presence of infection prevented the exacerbated weight gain and also improved metabolic parameters in animals fed a high fat diet. This was associated with the infection’s ability to modulate parameters of a cell responsible for regulatory functions: Tregs. In the light of these findings, helminth infection could be protective against weight gain and metabolic disturbances.
Collapse
Affiliation(s)
- Camila P. Queiroz-Glauss
- Departamento de Bioquímica e Imunologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Mariana S. Vieira
- Departamento de Bioquímica e Imunologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Marcela Helena Gonçalves-Pereira
- Departamento de Bioquímica e Imunologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Stephanie S. Almeida
- Departamento de Bioquímica e Imunologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Rachel H. Freire
- Departamento de Bioquímica e Imunologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Maria A. Gomes
- Departamento de Parasitologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Jacqueline I. Alvarez-Leite
- Departamento de Bioquímica e Imunologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Helton C. Santiago
- Departamento de Bioquímica e Imunologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
- * E-mail:
| |
Collapse
|
22
|
Schroeder JH, Howard JK, Lord GM. Transcription factor-driven regulation of ILC1 and ILC3. Trends Immunol 2022; 43:564-579. [PMID: 35618586 PMCID: PMC10166716 DOI: 10.1016/j.it.2022.04.009] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2022] [Revised: 04/26/2022] [Accepted: 04/28/2022] [Indexed: 10/18/2022]
Abstract
Mammalian innate lymphoid cells (ILCs) have functional relevance under both homeostatic and disease settings, such as inflammatory bowel disease (IBD), particularly in the context of maintaining the integrity of mucosal surfaces. Early reports highlighted group 1 and 3 ILC regulatory transcription factors (TFs), T-box expressed in T cells (T-bet; Tbx21) and RAR-related orphan nuclear receptor γt (RORγt; Rorc), as key regulators of ILC biology. Since then, other canonical TFs have been shown to have a role in the development and function of ILC subsets. In this review, we focus on recent insights into the balance between mature ILC1 and ILC3 based on these TFs and how they interact with other key cell-intrinsic molecular pathways. We outline how this TF interplay might be explored to identify novel candidate therapeutic avenues for human diseases.
Collapse
|
23
|
Chung DC, Jacquelot N, Ghaedi M, Warner K, Ohashi PS. Innate Lymphoid Cells: Role in Immune Regulation and Cancer. Cancers (Basel) 2022; 14:2071. [PMID: 35565201 PMCID: PMC9102917 DOI: 10.3390/cancers14092071] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2022] [Revised: 04/13/2022] [Accepted: 04/19/2022] [Indexed: 02/04/2023] Open
Abstract
Immune regulation is composed of a complex network of cellular and molecular pathways that regulate the immune system and prevent tissue damage. It is increasingly clear that innate lymphoid cells (ILCs) are also armed with immunosuppressive capacities similar to well-known immune regulatory cells (i.e., regulatory T cells). In cancer, immunoregulatory ILCs have been shown to inhibit anti-tumour immune response through various mechanisms including: (a) direct suppression of anti-tumour T cells or NK cells, (b) inhibiting T-cell priming, and (c) promoting other immunoregulatory cells. To provide a framework of understanding the role of immunosuppressive ILCs in the context of cancer, we first outline a brief history and challenges related to defining immunosuppressive ILCs. Furthermore, we focus on the mechanisms of ILCs in suppressing anti-tumour immunity and consequentially promoting tumour progression.
Collapse
Affiliation(s)
- Douglas C. Chung
- Department of Immunology, University of Toronto, Toronto, ON M5S 1A8, Canada
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON M5G 2M9, Canada; (N.J.); (M.G.); (K.W.)
| | - Nicolas Jacquelot
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON M5G 2M9, Canada; (N.J.); (M.G.); (K.W.)
| | - Maryam Ghaedi
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON M5G 2M9, Canada; (N.J.); (M.G.); (K.W.)
| | - Kathrin Warner
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON M5G 2M9, Canada; (N.J.); (M.G.); (K.W.)
| | - Pamela S. Ohashi
- Department of Immunology, University of Toronto, Toronto, ON M5S 1A8, Canada
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON M5G 2M9, Canada; (N.J.); (M.G.); (K.W.)
| |
Collapse
|
24
|
Wang S, van de Pavert SA. Innate Lymphoid Cells in the Central Nervous System. Front Immunol 2022; 13:837250. [PMID: 35185929 PMCID: PMC8852840 DOI: 10.3389/fimmu.2022.837250] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2021] [Accepted: 01/17/2022] [Indexed: 12/19/2022] Open
Abstract
Immune cells are present within the central nervous system and play important roles in neurological inflammation and disease. As relatively new described immune cell population, Innate Lymphoid Cells are now increasingly recognized within the central nervous system and associated diseases. Innate Lymphoid Cells are generally regarded as tissue resident and early responders, while conversely within the central nervous system at steady-state their presence is limited. This review describes the current understandings on Innate Lymphoid Cells in the central nervous system at steady-state and its borders plus their involvement in major neurological diseases like ischemic stroke, Alzheimer's disease and Multiple Sclerosis.
Collapse
Affiliation(s)
| | - Serge A. van de Pavert
- Aix-Marseille Université, Centre National de la Recherche Scientifique (CNRS), Institut National de la Santé et de la Recherche Médicale (INSERM), Centre d’Immunologie de Marseille-Luminy (CIML), Marseille, France
| |
Collapse
|
25
|
Jacquelot N, Ghaedi M, Warner K, Chung DC, Crome SQ, Ohashi PS. Immune Checkpoints and Innate Lymphoid Cells-New Avenues for Cancer Immunotherapy. Cancers (Basel) 2021; 13:5967. [PMID: 34885076 PMCID: PMC8657134 DOI: 10.3390/cancers13235967] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2021] [Revised: 11/23/2021] [Accepted: 11/24/2021] [Indexed: 12/21/2022] Open
Abstract
Immune checkpoints (IC) are broadly characterized as inhibitory pathways that tightly regulate the activation of the immune system. These molecular "brakes" are centrally involved in the maintenance of immune self-tolerance and represent a key mechanism in avoiding autoimmunity and tissue destruction. Antibody-based therapies target these inhibitory molecules on T cells to improve their cytotoxic function, with unprecedented clinical efficacies for a number of malignancies. Many of these ICs are also expressed on innate lymphoid cells (ILC), drawing interest from the field to understand their function, impact for anti-tumor immunity and potential for immunotherapy. In this review, we highlight ILC specificities at different tissue sites and their migration potential upon inflammatory challenge. We further summarize the current understanding of IC molecules on ILC and discuss potential strategies for ILC modulation as part of a greater anti-cancer armamentarium.
Collapse
Affiliation(s)
- Nicolas Jacquelot
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON M5G 2M9, Canada; (M.G.); (K.W.); (D.C.C.)
| | - Maryam Ghaedi
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON M5G 2M9, Canada; (M.G.); (K.W.); (D.C.C.)
| | - Kathrin Warner
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON M5G 2M9, Canada; (M.G.); (K.W.); (D.C.C.)
| | - Douglas C. Chung
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON M5G 2M9, Canada; (M.G.); (K.W.); (D.C.C.)
- Department of Immunology, University of Toronto, Toronto, ON M5S 1A8, Canada;
| | - Sarah Q. Crome
- Department of Immunology, University of Toronto, Toronto, ON M5S 1A8, Canada;
- Ajmera Transplant Centre, Toronto General Hospital Research Institute, University Health Network, Toronto, ON M5G 2C4, Canada
| | - Pamela S. Ohashi
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON M5G 2M9, Canada; (M.G.); (K.W.); (D.C.C.)
- Department of Immunology, University of Toronto, Toronto, ON M5S 1A8, Canada;
| |
Collapse
|
26
|
Miljković Đ, Jevtić B, Stojanović I, Dimitrijević M. ILC3, a Central Innate Immune Component of the Gut-Brain Axis in Multiple Sclerosis. Front Immunol 2021; 12:657622. [PMID: 33912185 PMCID: PMC8071931 DOI: 10.3389/fimmu.2021.657622] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2021] [Accepted: 03/15/2021] [Indexed: 12/12/2022] Open
Abstract
Gut immune cells have been increasingly appreciated as important players in the central nervous system (CNS) autoimmunity in animal models of multiple sclerosis (MS). Among the gut immune cells, innate lymphoid cell type 3 (ILC3) is of special interest in MS research, as they represent the innate cell counterpart of the major pathogenic cell population in MS, i.e. T helper (Th)17 cells. Importantly, these cells have been shown to stimulate regulatory T cells (Treg) and to counteract pathogenic Th17 cells in animal models of autoimmune diseases. Besides, they are also well known for their ability to stabilize the intestinal barrier and to shape the immune response to the gut microbiota. Thus, proper maintenance of the intestinal barrier and the establishment of the regulatory milieu in the gut performed by ILC3 may prevent activation of CNS antigen-specific Th17 cells by the molecular mimicry. Recent findings on the role of ILC3 in the gut-CNS axis and their relevance for MS pathogenesis will be discussed in this paper. Possibilities of ILC3 functional modulation for the benefit of MS patients will be addressed, as well.
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
| | - Bojan Jevtić
- Department of Immunology, Institute for Biological Research "Siniša Stanković" - National Institute of Republic of Serbia, University of Belgrade, Belgrade, Serbia
| | - Ivana Stojanović
- Department of Immunology, Institute for Biological Research "Siniša Stanković" - National Institute of Republic of Serbia, University of Belgrade, Belgrade, Serbia
| | - Mirjana Dimitrijević
- Department of Immunology, Institute for Biological Research "Siniša Stanković" - National Institute of Republic of Serbia, University of Belgrade, Belgrade, Serbia
| |
Collapse
|
27
|
Xian Y, Lv X, Xie M, Xiao F, Kong C, Ren Y. Physiological function and regulatory signal of intestinal type 3 innate lymphoid cell(s). Life Sci 2020; 262:118504. [PMID: 32991877 DOI: 10.1016/j.lfs.2020.118504] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2020] [Revised: 09/19/2020] [Accepted: 09/23/2020] [Indexed: 02/07/2023]
Abstract
Of the three groups of innate lymphoid cells, the type 3 innate lymphoid cell(s) (ILC3) include the subgroup of enteric ILC3 that participates in many physiological functions of the organism, such as promoting the repair of damaged mucosa, maintaining the homeostasis of gut symbiotic microorganisms, and presenting specific antigens. ILC3 also includes splenic and decidual ILC3. Like other physiological processes in the organism, enteric ILC3 functions are precisely regulated at the endogenous and exogenous levels. However, there has been no review on the physiological functions and regulatory signals of intestinal ILC3. In this paper, based on the current research on the physiological functions of enteric ILC3 in animals and the human, we summarize the signals that regulate cytokine secretion, antigen presentation and the quantity of ILC3 under normal intestinal conditions. We discuss for the first time the classification of the promoting mechanism of secretagogues of ILC3 into direct and indirect types. We also propose that ILC3 can promote intestinal homeostasis, and intestinal homeostasis can ensure the physiological phenotype of ILC3. If homeostasis is disturbed, ILC3 may participate in intestinal pathological changes. Therefore, regulating ILC3 and maintaining intestinal homeostasis are critical to the body.
Collapse
Affiliation(s)
- Yin Xian
- Department of General Surgery, and Institute of Hepato-Biliary-Pancreas and Intestinal Disease, Affiliated Hospital of North Sichuan Medical College, Nanchong 637000, PR China
| | - Xiaodong Lv
- Department of General Surgery, and Institute of Hepato-Biliary-Pancreas and Intestinal Disease, Affiliated Hospital of North Sichuan Medical College, Nanchong 637000, PR China
| | - Minjia Xie
- School of Clinical Medicine, North Sichuan Medical College, Nanchong 637000, PR China
| | - Fuyang Xiao
- School of Clinical Medicine, North Sichuan Medical College, Nanchong 637000, PR China
| | - Chenyang Kong
- School of Clinical Medicine, North Sichuan Medical College, Nanchong 637000, PR China
| | - Yixing Ren
- Department of General Surgery, and Institute of Hepato-Biliary-Pancreas and Intestinal Disease, Affiliated Hospital of North Sichuan Medical College, Nanchong 637000, PR China.
| |
Collapse
|
28
|
Song D, Lai L, Ran Z. Metabolic Regulation of Group 3 Innate Lymphoid Cells and Their Role in Inflammatory Bowel Disease. Front Immunol 2020; 11:580467. [PMID: 33193381 PMCID: PMC7649203 DOI: 10.3389/fimmu.2020.580467] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2020] [Accepted: 09/30/2020] [Indexed: 12/16/2022] Open
Abstract
Inflammatory bowel disease (IBD) is characterized by chronic and relapsing inflammatory disorder of the intestine. IBD is associated with complex pathogenesis, and considerable data suggest that innate lymphoid cells contribute to the development and progression of the condition. Group 3 innate lymphoid cells (ILC3s) not only play a protective role in maintaining intestinal homeostasis and gut barrier function, but also a pathogenic role in intestinal inflammation. ILC3s can sense environmental and host-derived signals and combine these cues to modulate cell expansion, migration and function, and transmit information to the broader immune system. Herein, we review current knowledge of how ILC3s can be regulated by dietary nutrients, microbiota and their metabolites, as well as other metabolites. In addition, we describe the phenotypic and functional alterations of ILC3s in IBD and discuss the therapeutic potential of ILC3s in the treatment of IBD.
Collapse
Affiliation(s)
| | | | - Zhihua Ran
- Division of Gastroenterology and Hepatology, Key Laboratory of Gastroenterology and Hepatology, Ministry of Health, Inflammatory Bowel Disease Research Center, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai Institute of Digestive Disease, Shanghai, China
| |
Collapse
|
29
|
Th1 responses in vivo require cell-specific provision of OX40L dictated by environmental cues. Nat Commun 2020; 11:3421. [PMID: 32647184 PMCID: PMC7347572 DOI: 10.1038/s41467-020-17293-3] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2019] [Accepted: 06/23/2020] [Indexed: 12/24/2022] Open
Abstract
The OX40-OX40L pathway provides crucial co-stimulatory signals for CD4 T cell responses, however the precise cellular interactions critical for OX40L provision in vivo and when these occur, remains unclear. Here, we demonstrate that provision of OX40L by dendritic cells (DCs), but not T cells, B cells nor group 3 innate lymphoid cells (ILC3s), is critical specifically for the effector Th1 response to an acute systemic infection with Listeria monocytogenes (Lm). OX40L expression by DCs is regulated by cross-talk with NK cells, with IFNγ signalling to the DC to enhance OX40L in a mechanism conserved in both mouse and human DCs. Strikingly, DC expression of OX40L is redundant in a chronic intestinal Th1 response and expression by ILC3s is necessary. Collectively these data reveal tissue specific compartmentalisation of the cellular provision of OX40L and define a mechanism controlling DC expression of OX40L in vivo. The OX40-OX40L axis is a crucial component of the costimulatory requirement of CD4 T cell responses. Here, the authors show context and cell type specific expression of OX40L for driving Th1 cell generation during acute and chronic models of infection.
Collapse
|
30
|
Ebihara T. Dichotomous Regulation of Acquired Immunity by Innate Lymphoid Cells. Cells 2020; 9:cells9051193. [PMID: 32403291 PMCID: PMC7290502 DOI: 10.3390/cells9051193] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2020] [Revised: 05/04/2020] [Accepted: 05/08/2020] [Indexed: 12/12/2022] Open
Abstract
The concept of innate lymphoid cells (ILCs) includes both conventional natural killer (NK) cells and helper ILCs, which resemble CD8+ killer T cells and CD4+ helper T cells in acquired immunity, respectively. Conventional NK cells are migratory cytotoxic cells that find tumor cells or cells infected with microbes. Helper ILCs are localized at peripheral tissue and are responsible for innate helper-cytokine production. Helper ILCs are classified into three subpopulations: TH1-like ILC1s, TH2-like ILC2s, and TH17/TH22-like ILC3s. Because of the functional similarities between ILCs and T cells, ILCs can serve as an innate component that augments each corresponding type of acquired immunity. However, the physiological functions of ILCs are more plastic and complicated than expected and are affected by environmental cues and types of inflammation. Here, we review recent advances in understanding the interaction between ILCs and acquired immunity, including T- and B-cell responses at various conditions. Immune suppressive activities by ILCs in particular are discussed in comparison to their immune stimulatory effects to gain precise knowledge of ILC biology and the physiological relevance of ILCs in human diseases.
Collapse
Affiliation(s)
- Takashi Ebihara
- Department of Medical Biology, Akita University Graduate School of Medicine Affiliation, 1-1-1 Hondo, Akita 010-8543, Japan
| |
Collapse
|
31
|
Zhang Z, Tang H, Chen P, Xie H, Tao Y. Demystifying the manipulation of host immunity, metabolism, and extraintestinal tumors by the gut microbiome. Signal Transduct Target Ther 2019; 4:41. [PMID: 31637019 PMCID: PMC6799818 DOI: 10.1038/s41392-019-0074-5] [Citation(s) in RCA: 156] [Impact Index Per Article: 26.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2019] [Revised: 08/27/2019] [Accepted: 08/27/2019] [Indexed: 02/06/2023] Open
Abstract
The trillions of microorganisms in the gut microbiome have attracted much attention recently owing to their sophisticated and widespread impacts on numerous aspects of host pathophysiology. Remarkable progress in large-scale sequencing and mass spectrometry has increased our understanding of the influence of the microbiome and/or its metabolites on the onset and progression of extraintestinal cancers and the efficacy of cancer immunotherapy. Given the plasticity in microbial composition and function, microbial-based therapeutic interventions, including dietary modulation, prebiotics, and probiotics, as well as fecal microbial transplantation, potentially permit the development of novel strategies for cancer therapy to improve clinical outcomes. Herein, we summarize the latest evidence on the involvement of the gut microbiome in host immunity and metabolism, the effects of the microbiome on extraintestinal cancers and the immune response, and strategies to modulate the gut microbiome, and we discuss ongoing studies and future areas of research that deserve focused research efforts.
Collapse
Affiliation(s)
- Ziying Zhang
- Key Laboratory of Carcinogenesis and Cancer Invasion, Ministry of Education, Department of Pathology, Xiangya Hospital, Central South University, 410078 Hunan, China
- NHC Key Laboratory of Carcinogenesis (Central South University), Cancer Research Institute and School of Basic Medicine, Central South University, 410078 Changsha, Hunan China
- Hunan Key Laboratory of Tumor Models and Individualized Medicine, Department of Thoracic Surgery, Second Xiangya Hospital, Central South University, 410011 Changsha, China
- Department of Oncology, Third Xiangya Hospital, Central South University, 410013 Changsha, China
| | - Haosheng Tang
- Key Laboratory of Carcinogenesis and Cancer Invasion, Ministry of Education, Department of Pathology, Xiangya Hospital, Central South University, 410078 Hunan, China
- NHC Key Laboratory of Carcinogenesis (Central South University), Cancer Research Institute and School of Basic Medicine, Central South University, 410078 Changsha, Hunan China
- Hunan Key Laboratory of Tumor Models and Individualized Medicine, Department of Thoracic Surgery, Second Xiangya Hospital, Central South University, 410011 Changsha, China
| | - Peng Chen
- Department of Urology, Xiangya Hospital, Central South University, 410008 Changsha, China
| | - Hui Xie
- Department of Thoracic and Cardiovascular Surgery, Second Xiangya Hospital of Central South University, 410011 Changsha, China
| | - Yongguang Tao
- Key Laboratory of Carcinogenesis and Cancer Invasion, Ministry of Education, Department of Pathology, Xiangya Hospital, Central South University, 410078 Hunan, China
- NHC Key Laboratory of Carcinogenesis (Central South University), Cancer Research Institute and School of Basic Medicine, Central South University, 410078 Changsha, Hunan China
- Hunan Key Laboratory of Tumor Models and Individualized Medicine, Department of Thoracic Surgery, Second Xiangya Hospital, Central South University, 410011 Changsha, China
| |
Collapse
|
32
|
Abstract
Despite continuous exposure to trillions of microbes, the intestinal immune system protects the mucosa by balancing barrier protection, tolerance, and immunity. As both sentinel and effector, the mucosal innate immune system plays a central role in coordinating these responses. By integrating signals from the intestinal microbiota, mononuclear phagocytes (MNPs) serve as a critical link in regulating effector functions of group 3 innate lymphoid cells (ILC3s). Our recent work identified the role for MNP production of the IBD-linked protein TNF-like ligand 1A (TL1A) in modulating microbial regulation of ILC3 barrier immunity. These findings highlight a broader role for ILC3s in local control of T cell immunity and their potential role in the pathogenesis and treatment of inflammatory disease.
Collapse
Affiliation(s)
- Jim G. Castellanos
- Jill Roberts Institute for Research in IBD, Division of Gastroenterology and Hepatology, Department of Medicine, Weill Cornell Medicine, New York, USA
| | - Randy S. Longman
- Jill Roberts Institute for Research in IBD, Division of Gastroenterology and Hepatology, Department of Medicine, Weill Cornell Medicine, New York, USA,CONTACT Randy S. Longman Jill Roberts Institute for Research in IBD, Division of Gastroenterology and Hepatology, Department of Medicine, Weill Cornell Medicine, New York, USA
| |
Collapse
|
33
|
Functional interactions between innate lymphoid cells and adaptive immunity. Nat Rev Immunol 2019; 19:599-613. [PMID: 31350531 PMCID: PMC6982279 DOI: 10.1038/s41577-019-0194-8] [Citation(s) in RCA: 184] [Impact Index Per Article: 30.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/27/2019] [Indexed: 12/19/2022]
Abstract
Innate lymphoid cells (ILCs) are enriched at barrier surfaces of the mammalian body where they rapidly respond to host, microbial or environmental stimuli to promote immunity or tissue homeostasis. Furthermore, ILCs are dysregulated in multiple human diseases. Over the past decade, substantial advances have been made in identifying the heterogeneity and functional diversity of ILCs, which have revealed striking similarities to T cell subsets. However, emerging evidence indicates that ILCs also have a complex role in directly influencing the adaptive immune response in the context of development, homeostasis, infection or inflammation. In turn, adaptive immunity reciprocally regulates ILCs, which indicates that these interactions are a crucial determinant of immune responses within tissues. Here, we summarize our current understanding of functional interactions between ILCs and the adaptive immune system, discuss limitations and future areas of investigation, and consider the potential for these interactions to be therapeutically harnessed to benefit human health.
Collapse
|