1
|
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
|
2
|
Bessell E, Finlay RE, James LK, Ludewig B, Harris NL, Krebs P, Hepworth MR, Dubey LK. Stromal cell and B cell dialogue potentiates IL-33-enriched lymphoid niches to support eosinophil recruitment and function during type 2 immunity. Cell Rep 2024; 43:114620. [PMID: 39141517 DOI: 10.1016/j.celrep.2024.114620] [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: 11/08/2023] [Revised: 05/27/2024] [Accepted: 07/25/2024] [Indexed: 08/16/2024] Open
Abstract
Eosinophils are involved in host protection against multicellular organisms. However, their recruitment to the mesenteric lymph node (mLN) during type 2 immunity is understudied. Our results demonstrate that eosinophil association with lymphoid stromal niches constructed by fibroblastic reticular cells (FRCs) and lymphatic endothelial cells is diminished in mice selectively lacking interleukin (IL)-4Rα or lymphotoxin-β (LTβ) expression on B cells. Furthermore, eosinophil survival, activation, and enhanced Il1rl1 receptor expression are driven by stromal cell and B cell dialogue. The ligation of lymphotoxin-β receptor (LTβR) on FRCs improves eosinophil survival and significantly augments IL-33 expression and eosinophil homing to the mLN, thus confirming the significance of lymphotoxin signaling for granulocyte recruitment. Eosinophil-deficient ΔdblGATA-1 mice show diminished mLN expansion, reduced interfollicular region (IFR) alarmin expression, and delayed helminth clearance, elucidating their importance in type 2 immunity. These findings provide insight into dialogue between stromal cells and B cells, which govern mLN eosinophilia, and the relevance of these mechanisms during type 2 immunity.
Collapse
Affiliation(s)
- Emily Bessell
- William Harvey Research Institute (WHRI), Barts & The London School of Medicine & Dentistry, Queen Mary University of London (QMUL), London, UK; Institute of Tissue Medicine and Pathology, University of Bern, Bern, Switzerland; Institute of Parasitology, Vetsuisse Faculty, University of Bern, Bern, Switzerland; Graduate School for Cellular and Biomedical Sciences, University of Bern, Bern, Switzerland
| | - Rachel E Finlay
- Division of Immunology, Immunity to Infection and Respiratory Medicine, Faculty of Biology, Medicine and Health, Lydia Becker Institute of Immunology and Inflammation, The University of Manchester, Manchester, UK
| | - Louisa K James
- Centre for Immunobiology, Blizard Institute, Queen Mary University of London, London, UK
| | - Burkhard Ludewig
- Institute of Immunobiology, Kantonsspital St. Gallen, St. Gallen, Switzerland
| | - Nicola L Harris
- Department of Immunology and Pathology, Central Clinical School, Monash University, The Alfred Centre, Melbourne, VIC, Australia
| | - Philippe Krebs
- Institute of Tissue Medicine and Pathology, University of Bern, Bern, Switzerland
| | - Matthew R Hepworth
- Division of Immunology, Immunity to Infection and Respiratory Medicine, Faculty of Biology, Medicine and Health, Lydia Becker Institute of Immunology and Inflammation, The University of Manchester, Manchester, UK
| | - Lalit Kumar Dubey
- William Harvey Research Institute (WHRI), Barts & The London School of Medicine & Dentistry, Queen Mary University of London (QMUL), London, UK; Institute of Tissue Medicine and Pathology, University of Bern, Bern, Switzerland.
| |
Collapse
|
3
|
Tanemoto R, Higashiyama M, Tomioka A, Ito S, Mizoguchi A, Nishii S, Inaba K, Wada A, Sugihara N, Hanawa Y, Horiuchi K, Okada Y, Kurihara C, Akita Y, Narimatsu K, Komoto S, Tomita K, Satoh T, Tsuda H, Hokari R. Chronic skin damage induces small intestinal damage via IL-13-induced apoptosis. Clin Exp Immunol 2024; 217:240-252. [PMID: 38916413 PMCID: PMC11310704 DOI: 10.1093/cei/uxae050] [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: 10/16/2023] [Revised: 05/20/2024] [Accepted: 06/24/2024] [Indexed: 06/26/2024] Open
Abstract
The gut-skin axis has recently been widely recognized, and both the gut and skin have been found to affect each other through a bidirectional connection; however, the precise mechanisms remain to be elucidated. Therefore, we aimed to investigate the effects of chronic skin damage (CSD) on mouse intestines. Following the CSD model, 4% sodium dodecyl sulfate was applied to the back-shaved murine skin six times for 2 weeks after tape stripping. The small and large intestines were analyzed histologically and immunologically, respectively. Intestinal permeability was measured using fluorescein isothiocyanate-conjugated-dextran. The role of interleukin-13 (IL-13) in the ileum was investigated using an anti-IL-13 antibody. Apoptotic intestinal cells were analyzed using TUNEL staining. Villus atrophy was observed in the small intestine in the CSD model, along with increased permeability. Mast cells, but not T cells, eosinophils, or innate lymph cell-2, were increased in the intestinal mucosa. However, no significant changes were observed in the large intestine. mRNA expression of IL-13 was increased only in the ileum of the CSD model. Apoptotic intestinal epithelial cells were significantly increased in the ileum of the CSD model. Administration of an anti-IL-13 antibody ameliorated the intestinal damage caused by CSD, along with decreased apoptotic cells and mast cell infiltration. Skin damage causes morphological changes in the small intestine, accompanied by increased intestinal permeability, possibly through the IL-13-induced apoptosis of mast cells in the epithelium. Surfactant-mediated mechanical skin damage can cause a leaky gut.
Collapse
Affiliation(s)
- Rina Tanemoto
- Department of Gastroenterology, National Defense Medical College, Saitama, Japan
| | - Masaaki Higashiyama
- Department of Gastroenterology, National Defense Medical College, Saitama, Japan
| | - Akira Tomioka
- Department of Gastroenterology, National Defense Medical College, Saitama, Japan
| | - Suguru Ito
- Department of Gastroenterology, National Defense Medical College, Saitama, Japan
| | - Akinori Mizoguchi
- Department of Gastroenterology, National Defense Medical College, Saitama, Japan
| | - Shin Nishii
- Department of Gastroenterology, National Defense Medical College, Saitama, Japan
| | - Kenichi Inaba
- Department of Gastroenterology, National Defense Medical College, Saitama, Japan
| | - Akinori Wada
- Department of Gastroenterology, National Defense Medical College, Saitama, Japan
| | - Nao Sugihara
- Department of Gastroenterology, National Defense Medical College, Saitama, Japan
| | - Yoshinori Hanawa
- Department of Gastroenterology, National Defense Medical College, Saitama, Japan
| | - Kazuki Horiuchi
- Department of Gastroenterology, National Defense Medical College, Saitama, Japan
| | - Yoshikiyo Okada
- Department of Gastroenterology, National Defense Medical College, Saitama, Japan
| | - Chie Kurihara
- Department of Gastroenterology, National Defense Medical College, Saitama, Japan
| | - Yoshihiro Akita
- Department of Gastroenterology, National Defense Medical College, Saitama, Japan
| | - Kazuyuki Narimatsu
- Department of Gastroenterology, National Defense Medical College, Saitama, Japan
| | - Shunsuke Komoto
- Department of Gastroenterology, National Defense Medical College, Saitama, Japan
| | - Kengo Tomita
- Department of Gastroenterology, National Defense Medical College, Saitama, Japan
| | - Takahiro Satoh
- Department of Dermatology, National Defense Medical College, Saitama, Japan
| | - Hitoshi Tsuda
- Department of Basic Pathology, National Defense Medical College, Saitama, Japan
| | - Ryota Hokari
- Department of Gastroenterology, National Defense Medical College, Saitama, Japan
| |
Collapse
|
4
|
Chaudhry MZ, Belz GT. Chameleon impersonation of NK cells and ILC1s. Nat Immunol 2024; 25:1313-1315. [PMID: 38982285 DOI: 10.1038/s41590-024-01886-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/11/2024]
Affiliation(s)
- M Zeeshan Chaudhry
- The University of Queensland Frazer Institute, University of Queensland, Woolloongabba, Queensland, Australia
| | - Gabrielle T Belz
- The University of Queensland Frazer Institute, University of Queensland, Woolloongabba, Queensland, Australia.
| |
Collapse
|
5
|
Mattar P, Reginato A, Lavados C, Das D, Kalyani M, Martinez-Lopez N, Sharma M, Skovbjerg G, Skytte JL, Roostalu U, Subbarayan R, Picarda E, Zang X, Zhang J, Guha C, Schwartz G, Rajbhandari P, Singh R. Insulin and leptin oscillations license food-entrained browning and metabolic flexibility. Cell Rep 2024; 43:114390. [PMID: 38900636 PMCID: PMC11562929 DOI: 10.1016/j.celrep.2024.114390] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2023] [Revised: 04/23/2024] [Accepted: 06/06/2024] [Indexed: 06/22/2024] Open
Abstract
Timed feeding drives adipose browning, although the integrative mechanisms for the same remain unclear. Here, we show that twice-a-night (TAN) feeding generates biphasic oscillations of circulating insulin and leptin, representing their entrainment by timed feeding. Insulin and leptin surges lead to marked cellular, functional, and metabolic remodeling of subcutaneous white adipose tissue (sWAT), resulting in increased energy expenditure. Single-cell RNA-sequencing (scRNA-seq) analyses and flow cytometry demonstrate a role for insulin and leptin surges in innate lymphoid type 2 (ILC2) cell recruitment and sWAT browning, since sWAT depot denervation or loss of leptin or insulin receptor signaling or ILC2 recruitment each dampens TAN feeding-induced sWAT remodeling and energy expenditure. Consistently, recreating insulin and leptin oscillations via once-a-day timed co-injections is sufficient to favorably remodel innervated sWAT. Innervation is necessary for sWAT remodeling, since denervation of sWAT, but not brown adipose tissue (BAT), blocks TAN-induced sWAT remodeling and resolution of inflammation. In sum, reorganization of nutrient-sensitive pathways remodels sWAT and drives the metabolic benefits of timed feeding.
Collapse
Affiliation(s)
- Pamela Mattar
- Department of Medicine, Division of Digestive Diseases, University of Los Angeles, Los Angeles, CA, USA
| | - Andressa Reginato
- Department of Developmental and Molecular Biology, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Christian Lavados
- Department of Developmental and Molecular Biology, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Debajyoti Das
- Department of Medicine, Division of Digestive Diseases, University of Los Angeles, Los Angeles, CA, USA
| | - Manu Kalyani
- Department of Developmental and Molecular Biology, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Nuria Martinez-Lopez
- Department of Medicine, Division of Digestive Diseases, University of Los Angeles, Los Angeles, CA, USA; Comprehensive Liver Research Center at UCLA, University of Los Angeles, Los Angeles, CA, USA
| | - Mridul Sharma
- Department of Medicine, Division of Digestive Diseases, University of Los Angeles, Los Angeles, CA, USA
| | | | | | | | | | - Elodie Picarda
- Department of Microbiology and Immunology, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Xingxing Zang
- Department of Microbiology and Immunology, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Jinghang Zhang
- Department of Microbiology and Immunology, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Chandan Guha
- Radiation Oncology, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Gary Schwartz
- Department of Medicine, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Prashant Rajbhandari
- Department of Medicine, Diabetes, Obesity, and Metabolism Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Rajat Singh
- Department of Medicine, Division of Digestive Diseases, University of Los Angeles, Los Angeles, CA, USA; Department of Developmental and Molecular Biology, Albert Einstein College of Medicine, Bronx, NY, USA; Department of Medicine, Albert Einstein College of Medicine, Bronx, NY, USA; Comprehensive Liver Research Center at UCLA, University of Los Angeles, Los Angeles, CA, USA.
| |
Collapse
|
6
|
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
|
7
|
Hashemi E, McCarthy C, Rao S, Malarkannan S. Transcriptomic diversity of innate lymphoid cells in human lymph nodes compared to BM and spleen. Commun Biol 2024; 7:769. [PMID: 38918571 PMCID: PMC11199704 DOI: 10.1038/s42003-024-06450-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2023] [Accepted: 06/12/2024] [Indexed: 06/27/2024] Open
Abstract
Innate lymphoid cells (ILCs) are largely tissue-resident, mostly described within the mucosal tissues. However, their presence and functions in the human draining lymph nodes (LNs) are unknown. Our study unravels the tissue-specific transcriptional profiles of 47,287 CD127+ ILCs within the human abdominal and thoracic LNs. LNs contain a higher frequency of CD127+ ILCs than in BM or spleen. We define independent stages of ILC development, including EILP and pILC in the BM. These progenitors exist in LNs in addition to naïve ILCs (nILCs) that can differentiate into mature ILCs. We define three ILC1 and four ILC3 sub-clusters in the LNs. ILC1 and ILC3 subsets have clusters with high heat shock protein-encoding genes. We identify previously unrecognized regulons, including the BACH2 family for ILC1 and the ATF family for ILC3. Our study is the comprehensive characterization of ILCs in LNs, providing an in-depth understanding of ILC-mediated immunity in humans.
Collapse
Affiliation(s)
- Elaheh Hashemi
- Blood Research Institute, Versiti, Milwaukee, WI, USA
- Department of Microbiology and Immunology, Medical College of Wisconsin (MCW), Milwaukee, WI, USA
| | | | - Sridhar Rao
- Blood Research Institute, Versiti, Milwaukee, WI, USA
- Division of Hematology, Oncology, and Bone Marrow Transplantation, Department of Pediatrics, MCW, Milwaukee, WI, USA
- Department of Cell Biology, Neurobiology, and Anatomy, MCW, Milwaukee, WI, USA
| | - Subramaniam Malarkannan
- Blood Research Institute, Versiti, Milwaukee, WI, USA.
- Department of Microbiology and Immunology, Medical College of Wisconsin (MCW), Milwaukee, WI, USA.
- Division of Hematology, Oncology, and Bone Marrow Transplantation, Department of Pediatrics, MCW, Milwaukee, WI, USA.
- Division of Hematology and Oncology, Department of Medicine, MCW, Milwaukee, WI, USA.
| |
Collapse
|
8
|
Liang Z, Tang Z, Zhu C, Li F, Chen S, Han X, Zheng R, Hu X, Lin R, Pei Q, Yin C, Wang J, Tang C, Cao N, Zhao J, Wang R, Li X, Luo N, Wen Q, Yu J, Li J, Xia X, Zheng X, Wang X, Huang N, Zhong Z, Mo C, Chen P, Wang Y, Fan J, Guo Y, Zhong H, Liu J, Peng Z, Mao H, Shi GP, Bonventre JV, Chen W, Zhou Y. Intestinal CXCR6 + ILC3s migrate to the kidney and exacerbate renal fibrosis via IL-23 receptor signaling enhanced by PD-1 expression. Immunity 2024; 57:1306-1323.e8. [PMID: 38815582 PMCID: PMC11539045 DOI: 10.1016/j.immuni.2024.05.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2023] [Revised: 02/27/2024] [Accepted: 05/06/2024] [Indexed: 06/01/2024]
Abstract
Group 3 innate lymphoid cells (ILC3s) regulate inflammation and tissue repair at mucosal sites, but whether these functions pertain to other tissues-like the kidneys-remains unclear. Here, we observed that renal fibrosis in humans was associated with increased ILC3s in the kidneys and blood. In mice, we showed that CXCR6+ ILC3s rapidly migrated from the intestinal mucosa and accumulated in the kidney via CXCL16 released from the injured tubules. Within the fibrotic kidney, ILC3s increased the expression of programmed cell death-1 (PD-1) and subsequent IL-17A production to directly activate myofibroblasts and fibrotic niche formation. ILC3 expression of PD-1 inhibited IL-23R endocytosis and consequently amplified the JAK2/STAT3/RORγt/IL-17A pathway that was essential for the pro-fibrogenic effect of ILC3s. Thus, we reveal a hitherto unrecognized migration pathway of ILC3s from the intestine to the kidney and the PD-1-dependent function of ILC3s in promoting renal fibrosis.
Collapse
Affiliation(s)
- Zhou Liang
- Department of Nephrology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou 510080, China; NHC Key Laboratory of Clinical Nephrology (Sun Yat-sen University) and Guangdong Provincial Key Laboratory of Nephrology, Guangzhou 510080, China
| | - Ziwen Tang
- Department of Nephrology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou 510080, China; NHC Key Laboratory of Clinical Nephrology (Sun Yat-sen University) and Guangdong Provincial Key Laboratory of Nephrology, Guangzhou 510080, China
| | - Changjian Zhu
- Department of Nephrology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou 510080, China; NHC Key Laboratory of Clinical Nephrology (Sun Yat-sen University) and Guangdong Provincial Key Laboratory of Nephrology, Guangzhou 510080, China
| | - Feng Li
- Department of Nephrology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou 510080, China; NHC Key Laboratory of Clinical Nephrology (Sun Yat-sen University) and Guangdong Provincial Key Laboratory of Nephrology, Guangzhou 510080, China
| | - Shuaijiabin Chen
- State Key Laboratory of Membrane Biology, Beijing Advanced Innovation Center for Structural Biology, Beijing Frontier Research Center for Biological Structure, School of Life Sciences, Tsinghua University, Beijing 100084, China
| | - Xu Han
- Department of Nephrology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou 510080, China; NHC Key Laboratory of Clinical Nephrology (Sun Yat-sen University) and Guangdong Provincial Key Laboratory of Nephrology, Guangzhou 510080, China
| | - Ruilin Zheng
- Department of Nephrology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou 510080, China; NHC Key Laboratory of Clinical Nephrology (Sun Yat-sen University) and Guangdong Provincial Key Laboratory of Nephrology, Guangzhou 510080, China
| | - Xinrong Hu
- Department of Nephrology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou 510080, China; NHC Key Laboratory of Clinical Nephrology (Sun Yat-sen University) and Guangdong Provincial Key Laboratory of Nephrology, Guangzhou 510080, China
| | - Ruoni Lin
- Department of Nephrology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou 510080, China; NHC Key Laboratory of Clinical Nephrology (Sun Yat-sen University) and Guangdong Provincial Key Laboratory of Nephrology, Guangzhou 510080, China
| | - Qiaoqiao Pei
- Department of Nephrology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou 510080, China; NHC Key Laboratory of Clinical Nephrology (Sun Yat-sen University) and Guangdong Provincial Key Laboratory of Nephrology, Guangzhou 510080, China
| | - Changjun Yin
- Precision Medicine Research Center, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou 510080, China
| | - Ji Wang
- Precision Medicine Research Center, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou 510080, China
| | - Ce Tang
- Precision Medicine Research Center, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou 510080, China
| | - Nan Cao
- Key Laboratory for Stem Cells and Tissue Engineering (Sun Yat-Sen University), Ministry of Education, Guangzhou 510080, China
| | - Jincun Zhao
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou 510182, China
| | - Rong Wang
- Department of Nephrology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou 510080, China; NHC Key Laboratory of Clinical Nephrology (Sun Yat-sen University) and Guangdong Provincial Key Laboratory of Nephrology, Guangzhou 510080, China
| | - Xiaoyan Li
- Department of Nephrology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou 510080, China; NHC Key Laboratory of Clinical Nephrology (Sun Yat-sen University) and Guangdong Provincial Key Laboratory of Nephrology, Guangzhou 510080, China
| | - Ning Luo
- Department of Nephrology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou 510080, China; NHC Key Laboratory of Clinical Nephrology (Sun Yat-sen University) and Guangdong Provincial Key Laboratory of Nephrology, Guangzhou 510080, China
| | - Qiong Wen
- Department of Nephrology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou 510080, China; NHC Key Laboratory of Clinical Nephrology (Sun Yat-sen University) and Guangdong Provincial Key Laboratory of Nephrology, Guangzhou 510080, China
| | - Jianwen Yu
- Department of Nephrology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou 510080, China; NHC Key Laboratory of Clinical Nephrology (Sun Yat-sen University) and Guangdong Provincial Key Laboratory of Nephrology, Guangzhou 510080, China
| | - Jianbo Li
- Department of Nephrology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou 510080, China; NHC Key Laboratory of Clinical Nephrology (Sun Yat-sen University) and Guangdong Provincial Key Laboratory of Nephrology, Guangzhou 510080, China
| | - Xi Xia
- Department of Nephrology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou 510080, China; NHC Key Laboratory of Clinical Nephrology (Sun Yat-sen University) and Guangdong Provincial Key Laboratory of Nephrology, Guangzhou 510080, China
| | - Xunhua Zheng
- Department of Nephrology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou 510080, China; NHC Key Laboratory of Clinical Nephrology (Sun Yat-sen University) and Guangdong Provincial Key Laboratory of Nephrology, Guangzhou 510080, China
| | - Xin Wang
- Department of Nephrology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou 510080, China; NHC Key Laboratory of Clinical Nephrology (Sun Yat-sen University) and Guangdong Provincial Key Laboratory of Nephrology, Guangzhou 510080, China
| | - Naya Huang
- Department of Nephrology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou 510080, China; NHC Key Laboratory of Clinical Nephrology (Sun Yat-sen University) and Guangdong Provincial Key Laboratory of Nephrology, Guangzhou 510080, China
| | - Zhong Zhong
- Department of Nephrology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou 510080, China; NHC Key Laboratory of Clinical Nephrology (Sun Yat-sen University) and Guangdong Provincial Key Laboratory of Nephrology, Guangzhou 510080, China
| | - Chengqiang Mo
- Department of Urology, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou 510080, China
| | - Peisong Chen
- Department of Laboratory Medicine, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou 510080, China
| | - Yating Wang
- Department of Nephrology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou 510080, China; NHC Key Laboratory of Clinical Nephrology (Sun Yat-sen University) and Guangdong Provincial Key Laboratory of Nephrology, Guangzhou 510080, China
| | - Jinjin Fan
- Department of Nephrology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou 510080, China; NHC Key Laboratory of Clinical Nephrology (Sun Yat-sen University) and Guangdong Provincial Key Laboratory of Nephrology, Guangzhou 510080, China
| | - Yun Guo
- Department of Nephrology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou 510080, China; NHC Key Laboratory of Clinical Nephrology (Sun Yat-sen University) and Guangdong Provincial Key Laboratory of Nephrology, Guangzhou 510080, China
| | - Haojie Zhong
- Department of Nephrology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou 510080, China; NHC Key Laboratory of Clinical Nephrology (Sun Yat-sen University) and Guangdong Provincial Key Laboratory of Nephrology, Guangzhou 510080, China
| | - Jiaqi Liu
- Department of Nephrology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou 510080, China; NHC Key Laboratory of Clinical Nephrology (Sun Yat-sen University) and Guangdong Provincial Key Laboratory of Nephrology, Guangzhou 510080, China
| | - Zhenwei Peng
- Department of Radiation Oncology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou 510080, China
| | - Haiping Mao
- Department of Nephrology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou 510080, China; NHC Key Laboratory of Clinical Nephrology (Sun Yat-sen University) and Guangdong Provincial Key Laboratory of Nephrology, Guangzhou 510080, China
| | - Guo-Ping Shi
- Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Joseph V Bonventre
- Department of Nephrology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA.
| | - Wei Chen
- Department of Nephrology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou 510080, China; NHC Key Laboratory of Clinical Nephrology (Sun Yat-sen University) and Guangdong Provincial Key Laboratory of Nephrology, Guangzhou 510080, China.
| | - Yi Zhou
- Department of Nephrology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou 510080, China; NHC Key Laboratory of Clinical Nephrology (Sun Yat-sen University) and Guangdong Provincial Key Laboratory of Nephrology, Guangzhou 510080, China.
| |
Collapse
|
9
|
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
|
10
|
Cruz de Casas P, Knöpper K, Dey Sarkar R, Kastenmüller W. Same yet different - how lymph node heterogeneity affects immune responses. Nat Rev Immunol 2024; 24:358-374. [PMID: 38097778 DOI: 10.1038/s41577-023-00965-8] [Citation(s) in RCA: 10] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/30/2023] [Indexed: 05/04/2024]
Abstract
Lymph nodes are secondary lymphoid organs in which immune responses of the adaptive immune system are initiated and regulated. Distributed throughout the body and embedded in the lymphatic system, local lymph nodes are continuously informed about the state of the organs owing to a constant drainage of lymph. The tissue-derived lymph carries products of cell metabolism, proteins, carbohydrates, lipids, pathogens and circulating immune cells. Notably, there is a growing body of evidence that individual lymph nodes differ from each other in their capacity to generate immune responses. Here, we review the structure and function of the lymphatic system and then focus on the factors that lead to functional heterogeneity among different lymph nodes. We will discuss how lymph node heterogeneity impacts on cellular and humoral immune responses and the implications for vaccination, tumour development and tumour control by immunotherapy.
Collapse
Affiliation(s)
- Paulina Cruz de Casas
- Max Planck Research Group, Würzburg Institute of Systems Immunology, Julius-Maximilians-Universität Würzburg, Würzburg, Germany
| | - Konrad Knöpper
- Howard Hughes Medical Institute, University of California, San Francisco, San Francisco, CA, USA
- Department of Microbiology and Immunology, University of California, San Francisco, San Francisco, CA, USA
| | - Rupak Dey Sarkar
- Max Planck Research Group, Würzburg Institute of Systems Immunology, Julius-Maximilians-Universität Würzburg, Würzburg, Germany
| | - Wolfgang Kastenmüller
- Max Planck Research Group, Würzburg Institute of Systems Immunology, Julius-Maximilians-Universität Würzburg, Würzburg, Germany.
| |
Collapse
|
11
|
Saadh MJ, Ahmed HM, Alani ZK, Al Zuhairi RAH, Almarhoon ZM, Ahmad H, Ubaid M, Alwan NH. The Role of Gut-derived Short-Chain Fatty Acids in Multiple Sclerosis. Neuromolecular Med 2024; 26:14. [PMID: 38630350 DOI: 10.1007/s12017-024-08783-4] [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: 02/04/2024] [Accepted: 03/08/2024] [Indexed: 04/19/2024]
Abstract
Multiple sclerosis (MS) is a chronic condition affecting the central nervous system (CNS), where the interplay of genetic and environmental factors influences its pathophysiology, triggering immune responses and instigating inflammation. Contemporary research has been notably dedicated to investigating the contributions of gut microbiota and their metabolites in modulating inflammatory reactions within the CNS. Recent recognition of the gut microbiome and dietary patterns as environmental elements impacting MS development emphasizes the potential influence of small, ubiquitous molecules from microbiota, such as short-chain fatty acids (SCFAs). These molecules may serve as vital molecular signals or metabolic substances regulating host cellular metabolism in the intricate interplay between microbiota and the host. A current emphasis lies on optimizing the health-promoting attributes of colonic bacteria to mitigate urinary tract issues through dietary management. This review aims to spotlight recent investigations on the impact of SCFAs on immune cells pivotal in MS, the involvement of gut microbiota and SCFAs in MS development, and the considerable influence of probiotics on gastrointestinal disruptions in MS. Comprehending the gut-CNS connection holds promise for the development of innovative therapeutic approaches, particularly probiotic-based supplements, for managing MS.
Collapse
Affiliation(s)
- Mohamed J Saadh
- Faculty of Pharmacy, Middle East University, Amman, 11831, Jordan
| | - Hani Moslem Ahmed
- Department of Dental Industry Techniques, Al-Noor University College, Nineveh, Iraq
| | - Zaid Khalid Alani
- College of Health and Medical Technical, Al-Bayan University, Baghdad, Iraq
| | | | - Zainab M Almarhoon
- Department of Chemistry, College of Science, King Saud University, 11451, Riyadh, Saudi Arabia
| | - Hijaz Ahmad
- Section of Mathematics, International Telematic University Uninettuno, Corso Vittorio Emanuele II, 39, 00186, Rome, Italy.
- Center for Applied Mathematics and Bioinformatics, Gulf University for Science and Technology, Mubarak Al-Abdullah, Kuwait.
- Department of Computer Science and Mathematics, Lebanese American University, Beirut, Lebanon.
| | - Mohammed Ubaid
- Medical Technical College, Al-Farahidi University, Baghdad, Iraq
| | | |
Collapse
|
12
|
Chen H, Sun L, Feng L, Han X, Zhang Y, Zhai W, Zhang Z, Mulholland M, Zhang W, Yin Y. Intermittent fasting promotes type 3 innate lymphoid cells secreting IL-22 contributing to the beigeing of white adipose tissue. eLife 2024; 12:RP91060. [PMID: 38536726 PMCID: PMC10972562 DOI: 10.7554/elife.91060] [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] [Indexed: 04/04/2024] Open
Abstract
Mechanism underlying the metabolic benefit of intermittent fasting remains largely unknown. Here, we reported that intermittent fasting promoted interleukin-22 (IL-22) production by type 3 innate lymphoid cells (ILC3s) and subsequent beigeing of subcutaneous white adipose tissue. Adoptive transfer of intestinal ILC3s increased beigeing of white adipose tissue in diet-induced-obese mice. Exogenous IL-22 significantly increased the beigeing of subcutaneous white adipose tissue. Deficiency of IL-22 receptor (IL-22R) attenuated the beigeing induced by intermittent fasting. Single-cell sequencing of sorted intestinal immune cells revealed that intermittent fasting increased aryl hydrocarbon receptor signaling in ILC3s. Analysis of cell-cell ligand receptor interactions indicated that intermittent fasting may stimulate the interaction of ILC3s with dendritic cells and macrophages. These results establish the role of intestinal ILC3s in beigeing of white adipose tissue, suggesting that ILC3/IL-22/IL-22R axis contributes to the metabolic benefit of intermittent fasting.
Collapse
Affiliation(s)
- Hong Chen
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Center for Reproductive Medicine, Third Hospital, Peking UniversityBeijingChina
- State Key Laboratory of Female Fertility Promote, Center for Reproductive Medicine, Department of Obstetrics and Gynecology, Peking University Third HospitalBeijingChina
| | - Lijun Sun
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Center for Reproductive Medicine, Third Hospital, Peking UniversityBeijingChina
| | - Lu Feng
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Center for Reproductive Medicine, Third Hospital, Peking UniversityBeijingChina
| | - Xue Han
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Center for Reproductive Medicine, Third Hospital, Peking UniversityBeijingChina
| | - Yunhua Zhang
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Center for Reproductive Medicine, Third Hospital, Peking UniversityBeijingChina
| | - Wenbo Zhai
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Center for Reproductive Medicine, Third Hospital, Peking UniversityBeijingChina
| | - Zehe Zhang
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Center for Reproductive Medicine, Third Hospital, Peking UniversityBeijingChina
| | - Michael Mulholland
- Department of Surgery, University of Michigan Medical CenterAnn ArborUnited States
| | - Weizhen Zhang
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Center for Reproductive Medicine, Third Hospital, Peking UniversityBeijingChina
- Department of Surgery, University of Michigan Medical CenterAnn ArborUnited States
| | - Yue Yin
- Department of Pharmacology, School of Basic Medical Sciences, and Key Laboratory of Molecular Cardiovascular Science, Ministry of Education, Peking UniversityBeijingChina
| |
Collapse
|
13
|
Uddin MJ, Thompson B, Leslie JL, Fishman C, Sol-church K, Kumar P, Petri WA. Investigating the impact of antibiotic-induced dysbiosis on protection from Clostridium difficile colitis by mouse colonic innate lymphoid cells. mBio 2024; 15:e0333823. [PMID: 38376154 PMCID: PMC11209775 DOI: 10.1128/mbio.03338-23] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2023] [Accepted: 01/18/2024] [Indexed: 02/21/2024] Open
Abstract
Innate lymphoid cells (ILCs) play a critical role in maintaining intestinal health in homeostatic and diseased conditions. During Clostridium difficile infection (CDI), IL-33 activates ILC2 to protect from colonic damage and mortality. The function of IL-33 and ILC is tightly regulated by the intestinal microbiota. We set out to determine the impact of antibiotic-induced disruption of the microbiome on ILC function. Our goal was to understand antibiotic-induced changes in ILC function on susceptibility to C. difficile colitis in a mouse model. We utilized high-throughput single-cell RNAseq to investigate the phenotypic features of colonic ILC at baseline, after antibiotic administration with or without IL-33 treatment. We identified a heterogeneous landscape of colonic ILCs with gene signatures of inflammatory, anti-inflammatory, migratory, progenitor, plastic, and antigen-presenting ILCs. Antibiotic treatment decreased ILC2 while coordinately increasing ILC1 and ILC3 phenotypes. Notably, Ifng+, Ccl5+, and Il23r+ ILC increased after antibiotics. IL-33 treatment counteracted the antibiotic effect by downregulating ILC1 and ILC3 and activating ILC2. In addition, IL-33 treatment markedly induced the expression of type 2 genes, including Areg and Il5. Finally, we identified amphiregulin, produced by ILC2, as protective during C. difficile infection. Together, our data expand our understanding of how antibiotics induce susceptibility to C. difficile colitis through their impact on ILC subsets and function.IMPORTANCEClostridium difficile infection (CDI) accounts for around 500,000 symptomatic cases and over 20,000 deaths annually in the United States alone. A major risk factor of CDI is antibiotic-induced dysbiosis of the gut. Microbiota-regulated IL-33 and innate lymphoid cells (ILCs) are important in determining the outcomes of C. difficile infection. Understanding how antibiotic and IL-33 treatment alter the phenotype of colon ILCs is important to identify potential therapeutics. Here, we performed single-cell RNAseq of mouse colon ILCs collected at baseline, after antibiotic treatment, and after IL-33 treatment. We identified heterogeneous subpopulations of all three ILC subtypes in the mouse colon. Our analysis revealed several potential pathways of antibiotic-mediated increased susceptibility to intestinal infection. Our discovery that Areg is abundantly expressed by ILCs, and the protection of mice from CDI by amphiregulin treatment, suggests that the amphiregulin-epidermal growth factor receptor pathway is a potential therapeutic target for treating intestinal colitis.
Collapse
Affiliation(s)
- Md Jashim Uddin
- Division of Infectious Diseases and International Health, Department of Medicine, University of Virginia School of Medicine, Charlottesville, Virginia, USA
| | - Brandon Thompson
- Division of Infectious Diseases and International Health, Department of Medicine, University of Virginia School of Medicine, Charlottesville, Virginia, USA
| | - Jhansi L. Leslie
- Division of Infectious Diseases and International Health, Department of Medicine, University of Virginia School of Medicine, Charlottesville, Virginia, USA
- Arcus Biosciences, Hayward, California, USA
| | - Casey Fishman
- Division of Infectious Diseases and International Health, Department of Medicine, University of Virginia School of Medicine, Charlottesville, Virginia, USA
| | - Katia Sol-church
- Genome Analysis and Technology Core, University of Virginia School of Medicine, Charlottesville, Virginia, USA
| | - Pankaj Kumar
- Department of Biochemistry and Molecular Genetics, University of Virginia School of Medicine, Charlottesville, Virginia, USA
| | - William A. Petri
- Division of Infectious Diseases and International Health, Department of Medicine, University of Virginia School of Medicine, Charlottesville, Virginia, USA
| |
Collapse
|
14
|
Fol M, Karpik W, Zablotni A, Kulesza J, Kulesza E, Godkowicz M, Druszczynska M. Innate Lymphoid Cells and Their Role in the Immune Response to Infections. Cells 2024; 13:335. [PMID: 38391948 PMCID: PMC10886880 DOI: 10.3390/cells13040335] [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: 01/05/2024] [Revised: 02/07/2024] [Accepted: 02/10/2024] [Indexed: 02/24/2024] Open
Abstract
Over the past decade, a group of lymphocyte-like cells called innate lymphoid cells (ILCs) has gained considerable attention due to their crucial role in regulating immunity and tissue homeostasis. ILCs, lacking antigen-specific receptors, are a group of functionally differentiated effector cells that act as tissue-resident sentinels against infections. Numerous studies have elucidated the characteristics of ILC subgroups, but the mechanisms controlling protective or pathological responses to pathogens still need to be better understood. This review summarizes the functions of ILCs in the immunology of infections caused by different intracellular and extracellular pathogens and discusses their possible therapeutic potential.
Collapse
Affiliation(s)
- Marek Fol
- Department of Immunology and Infectious Biology, Institute of Microbiology, Biotechnology and Immunology, Faculty of Biology and Environmental Protection, University of Lodz, 90-237 Lodz, Poland; (M.F.); (W.K.); (M.G.)
| | - Wojciech Karpik
- Department of Immunology and Infectious Biology, Institute of Microbiology, Biotechnology and Immunology, Faculty of Biology and Environmental Protection, University of Lodz, 90-237 Lodz, Poland; (M.F.); (W.K.); (M.G.)
| | - Agnieszka Zablotni
- Department of Bacterial Biology, Faculty of Biology and Environmental Protection, University of Lodz, 90-237 Lodz, Poland;
| | - Jakub Kulesza
- Department of Internal Diseases and Clinical Pharmacology, Medical University of Lodz, 91-347 Lodz, Poland;
| | - Ewelina Kulesza
- Department of Rheumatology and Internal Diseases, Medical University of Lodz, 90-549 Lodz, Poland;
| | - Magdalena Godkowicz
- Department of Immunology and Infectious Biology, Institute of Microbiology, Biotechnology and Immunology, Faculty of Biology and Environmental Protection, University of Lodz, 90-237 Lodz, Poland; (M.F.); (W.K.); (M.G.)
- Lodz Institutes of the Polish Academy of Sciences, The Bio-Med-Chem Doctoral School, University of Lodz, 90-237 Lodz, Poland
| | - Magdalena Druszczynska
- Department of Immunology and Infectious Biology, Institute of Microbiology, Biotechnology and Immunology, Faculty of Biology and Environmental Protection, University of Lodz, 90-237 Lodz, Poland; (M.F.); (W.K.); (M.G.)
| |
Collapse
|
15
|
Galván-Peña S, Zhu Y, Hanna BS, Mathis D, Benoist C. A dynamic atlas of immunocyte migration from the gut. Sci Immunol 2024; 9:eadi0672. [PMID: 38181094 PMCID: PMC10964343 DOI: 10.1126/sciimmunol.adi0672] [Citation(s) in RCA: 11] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2023] [Accepted: 12/06/2023] [Indexed: 01/07/2024]
Abstract
Dysbiosis in the gut microbiota affects several systemic diseases, possibly by driving the migration of perturbed intestinal immunocytes to extraintestinal tissues. Combining Kaede photoconvertible mice and single-cell genomics, we generated a detailed map of migratory trajectories from the colon, at baseline, and in several models of intestinal and extraintestinal inflammation. All lineages emigrated from the colon in an S1P-dependent manner. B lymphocytes represented the largest contingent, with the unexpected circulation of nonexperienced follicular B cells, which carried a gut-imprinted transcriptomic signature. T cell emigration included distinct groups of RORγ+ and IEL-like CD160+ subsets. Gut inflammation curtailed emigration, except for dendritic cells disseminating to lymph nodes. Colon-emigrating cells distributed differentially to distinct sites of extraintestinal models of inflammation (psoriasis-like skin, arthritic synovium, and tumors). Thus, specific cellular trails originating in the gut and influenced by microbiota may shape peripheral immunity in varied ways.
Collapse
Affiliation(s)
| | - Yangyang Zhu
- Department of Immunology, Harvard Medical School, Boston, MA, USA
| | - Bola S. Hanna
- Department of Immunology, Harvard Medical School, Boston, MA, USA
| | - Diane Mathis
- Department of Immunology, Harvard Medical School, Boston, MA, USA
| | | |
Collapse
|
16
|
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: 18] [Impact Index Per Article: 18.0] [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
|
17
|
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
|
18
|
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: 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: 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
|
19
|
McKendrick JG, Jones GR, Elder SS, Watson E, T'Jonck W, Mercer E, Magalhaes MS, Rocchi C, Hegarty LM, Johnson AL, Schneider C, Becher B, Pridans C, Mabbott N, Liu Z, Ginhoux F, Bajenoff M, Gentek R, Bain CC, Emmerson E. CSF1R-dependent macrophages in the salivary gland are essential for epithelial regeneration after radiation-induced injury. Sci Immunol 2023; 8:eadd4374. [PMID: 37922341 DOI: 10.1126/sciimmunol.add4374] [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: 06/11/2022] [Accepted: 10/03/2023] [Indexed: 11/05/2023]
Abstract
The salivary glands often become damaged in individuals receiving radiotherapy for head and neck cancer, resulting in chronic dry mouth. This leads to detrimental effects on their health and quality of life, for which there is no regenerative therapy. Macrophages are the predominant immune cell in the salivary glands and are attractive therapeutic targets due to their unrivaled capacity to drive tissue repair. Yet, the nature and role of macrophages in salivary gland homeostasis and how they may contribute to tissue repair after injury are not well understood. Here, we show that at least two phenotypically and transcriptionally distinct CX3CR1+ macrophage populations are present in the adult salivary gland, which occupy anatomically distinct niches. CD11c+CD206-CD163- macrophages typically associate with gland epithelium, whereas CD11c-CD206+CD163+ macrophages associate with blood vessels and nerves. Using a suite of complementary fate mapping systems, we show that there are highly dynamic changes in the ontogeny and composition of salivary gland macrophages with age. Using an in vivo model of radiation-induced salivary gland injury combined with genetic or antibody-mediated depletion of macrophages, we demonstrate an essential role for macrophages in clearance of cells with DNA damage. Furthermore, we show that epithelial-associated macrophages are indispensable for effective tissue repair and gland function after radiation-induced injury, with their depletion resulting in reduced saliva production. Our data, therefore, provide a strong case for exploring the therapeutic potential of manipulating macrophages to promote tissue repair and thus minimize salivary gland dysfunction after radiotherapy.
Collapse
Affiliation(s)
- John G McKendrick
- Centre for Regenerative Medicine, Institute for Regeneration and Repair, University of Edinburgh, 4-5 Little France Drive, Edinburgh, EH16 4UU, UK
- Centre for Inflammation Research, Institute for Regeneration and Repair, University of Edinburgh, 4-5 Little France Drive, Edinburgh, EH16 4UU, UK
| | - Gareth-Rhys Jones
- Centre for Inflammation Research, Institute for Regeneration and Repair, University of Edinburgh, 4-5 Little France Drive, Edinburgh, EH16 4UU, UK
| | - Sonia S Elder
- Centre for Regenerative Medicine, Institute for Regeneration and Repair, University of Edinburgh, 4-5 Little France Drive, Edinburgh, EH16 4UU, UK
| | - Erin Watson
- Centre for Regenerative Medicine, Institute for Regeneration and Repair, University of Edinburgh, 4-5 Little France Drive, Edinburgh, EH16 4UU, UK
| | - Wouter T'Jonck
- Centre for Inflammation Research, Institute for Regeneration and Repair, University of Edinburgh, 4-5 Little France Drive, Edinburgh, EH16 4UU, UK
| | - Ella Mercer
- Centre for Regenerative Medicine, Institute for Regeneration and Repair, University of Edinburgh, 4-5 Little France Drive, Edinburgh, EH16 4UU, UK
| | - Marlene S Magalhaes
- Centre for Inflammation Research, Institute for Regeneration and Repair, University of Edinburgh, 4-5 Little France Drive, Edinburgh, EH16 4UU, UK
- Centre for Reproductive Health, Institute for Regeneration and Repair, University of Edinburgh, 4-5 Little France Drive, Edinburgh, EH16 4UU, UK
| | - Cecilia Rocchi
- Centre for Regenerative Medicine, Institute for Regeneration and Repair, University of Edinburgh, 4-5 Little France Drive, Edinburgh, EH16 4UU, UK
| | - Lizi M Hegarty
- Centre for Inflammation Research, Institute for Regeneration and Repair, University of Edinburgh, 4-5 Little France Drive, Edinburgh, EH16 4UU, UK
| | - Amanda L Johnson
- Centre for Regenerative Medicine, Institute for Regeneration and Repair, University of Edinburgh, 4-5 Little France Drive, Edinburgh, EH16 4UU, UK
| | | | - Burkhard Becher
- Institute of Experimental Immunology, University of Zurich, Zurich, Switzerland
| | - Clare Pridans
- Centre for Inflammation Research, Institute for Regeneration and Repair, University of Edinburgh, 4-5 Little France Drive, Edinburgh, EH16 4UU, UK
| | - Neil Mabbott
- Roslin Institute & Royal (Dick) School of Veterinary Studies, University of Edinburgh, Easter Bush Campus, Midlothian EH25 9RG, UK
| | - Zhaoyuan Liu
- Shanghai Institute of Immunology, Department of Immunology and Microbiology, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Florent Ginhoux
- Shanghai Institute of Immunology, Department of Immunology and Microbiology, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
- Singapore Immunology Network, Agency for Science, Technology and Research, Singapore 138648, Singapore
- Translational Immunology Institute, SingHealth Duke-NUS Academic Medical Centre, Singapore, Singapore
| | - Marc Bajenoff
- Centre d'Immunologie de Marseille-Luminy, Aix Marseille Université UM2, INSERM, U1104, CNRS UMR7280, Marseille 13288, France
| | - Rebecca Gentek
- Centre for Inflammation Research, Institute for Regeneration and Repair, University of Edinburgh, 4-5 Little France Drive, Edinburgh, EH16 4UU, UK
- Centre for Reproductive Health, Institute for Regeneration and Repair, University of Edinburgh, 4-5 Little France Drive, Edinburgh, EH16 4UU, UK
| | - Calum C Bain
- Centre for Inflammation Research, Institute for Regeneration and Repair, University of Edinburgh, 4-5 Little France Drive, Edinburgh, EH16 4UU, UK
| | - Elaine Emmerson
- Centre for Regenerative Medicine, Institute for Regeneration and Repair, University of Edinburgh, 4-5 Little France Drive, Edinburgh, EH16 4UU, UK
| |
Collapse
|
20
|
Sonar SA, Watanabe M, Nikolich JŽ. Disorganization of secondary lymphoid organs and dyscoordination of chemokine secretion as key contributors to immune aging. Semin Immunol 2023; 70:101835. [PMID: 37651849 PMCID: PMC10840697 DOI: 10.1016/j.smim.2023.101835] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/26/2023] [Revised: 08/18/2023] [Accepted: 08/18/2023] [Indexed: 09/02/2023]
Abstract
Aging is characterized by progressive loss of organ and tissue function, and the immune system is no exception to that inevitable principle. Of all the age-related changes in the body, reduction of the size of, and naïve T (Tn) cell output from, the thymus occurs earliest, being prominent already before or by the time of puberty. Therefore, to preserve immunity against new infections, over much of their lives, vertebrates dominantly rely on peripheral maintenance of the Tn cell pool in the secondary lymphoid organs (SLO). However, SLO structure and function subsequently also deteriorate with aging. Several recent studies have made a convincing case that this deterioration is of major importance to the erosion of protective immunity in the last third of life. Specifically, the SLO were found to accumulate multiple degenerative changes with aging. Importantly, the results from adoptive transfer and parabiosis studies teach us that the old microenvironment is the limiting factor for protective immunity in old mice. In this review, we discuss the extent, mechanisms, and potential role of stromal cell aging in the age-related alteration of T cell homeostatic maintenance and immune function decline. We use that discussion to frame the potential strategies to correct the SLO stromal aging defects - in the context of other immune rejuvenation approaches, - to improve functional immune responses and protective immunity in older adults.
Collapse
Affiliation(s)
- Sandip Ashok Sonar
- Department of Immunobiology, University of Arizona College of Medicine-Tucson, Tucson, AZ, USA; The University of Arizona Center on Aging, University of Arizona College of Medicine-Tucson, Tucson, AZ, USA
| | - Makiko Watanabe
- Department of Immunobiology, University of Arizona College of Medicine-Tucson, Tucson, AZ, USA; The University of Arizona Center on Aging, University of Arizona College of Medicine-Tucson, Tucson, AZ, USA
| | - Janko Ž Nikolich
- Department of Immunobiology, University of Arizona College of Medicine-Tucson, Tucson, AZ, USA; The University of Arizona Center on Aging, University of Arizona College of Medicine-Tucson, Tucson, AZ, USA; the Aegis Consortium for Pandemic-free Future, University of Arizona Health Sciences, USA; BIO5 Institute, University of Arizona, Tucson, AZ, USA.
| |
Collapse
|
21
|
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
|
22
|
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: 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: 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
|
23
|
Guo Y, Liu Y, Rui B, Lei Z, Ning X, Liu Y, Li M. Crosstalk between the gut microbiota and innate lymphoid cells in intestinal mucosal immunity. Front Immunol 2023; 14:1171680. [PMID: 37304260 PMCID: PMC10249960 DOI: 10.3389/fimmu.2023.1171680] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2023] [Accepted: 05/02/2023] [Indexed: 06/13/2023] Open
Abstract
The human gastrointestinal mucosa is colonized by thousands of microorganisms, which participate in a variety of physiological functions. Intestinal dysbiosis is closely associated with the pathogenesis of several human diseases. Innate lymphoid cells (ILCs), which include NK cells, ILC1s, ILC2s, ILC3s and LTi cells, are a type of innate immune cells. They are enriched in the mucosal tissues of the body, and have recently received extensive attention. The gut microbiota and its metabolites play important roles in various intestinal mucosal diseases, such as inflammatory bowel disease (IBD), allergic disease, and cancer. Therefore, studies on ILCs and their interaction with the gut microbiota have great clinical significance owing to their potential for identifying pharmacotherapy targets for multiple related diseases. This review expounds on the progress in research on ILCs differentiation and development, the biological functions of the intestinal microbiota, and its interaction with ILCs in disease conditions in order to provide novel ideas for disease treatment in the future.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Ming Li
- *Correspondence: Yinhui Liu, ; Ming Li,
| |
Collapse
|
24
|
Fukaya T, Uto T, Mitoma S, Takagi H, Nishikawa Y, Tominaga M, Choijookhuu N, Hishikawa Y, Sato K. Gut dysbiosis promotes the breakdown of oral tolerance mediated through dysfunction of mucosal dendritic cells. Cell Rep 2023; 42:112431. [PMID: 37099426 DOI: 10.1016/j.celrep.2023.112431] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2021] [Revised: 02/21/2023] [Accepted: 04/10/2023] [Indexed: 04/27/2023] Open
Abstract
While dysbiosis in the gut is implicated in the impaired induction of oral tolerance generated in mesenteric lymph nodes (MesLNs), how dysbiosis affects this process remains unclear. Here, we describe that antibiotic-driven gut dysbiosis causes the dysfunction of CD11c+CD103+ conventional dendritic cells (cDCs) in MesLNs, preventing the establishment of oral tolerance. Deficiency of CD11c+CD103+ cDCs abrogates the generation of regulatory T cells in MesLNs to establish oral tolerance. Antibiotic treatment triggers the intestinal dysbiosis linked to the impaired generation of colony-stimulating factor 2 (Csf2)-producing group 3 innate lymphoid cells (ILC3s) for regulating the tolerogenesis of CD11c+CD103+ cDCs and the reduced expression of tumor necrosis factor (TNF)-like ligand 1A (TL1A) on CD11c+CD103+ cDCs for generating Csf2-producing ILC3s. Thus, antibiotic-driven intestinal dysbiosis leads to the breakdown of crosstalk between CD11c+CD103+ cDCs and ILC3s for maintaining the tolerogenesis of CD11c+CD103+ cDCs in MesLNs, responsible for the failed establishment of oral tolerance.
Collapse
Affiliation(s)
- Tomohiro Fukaya
- Division of Immunology, Department of Infectious Diseases, Faculty of Medicine, University of Miyazaki, 5200 Kihara, Kiyotake, Miyazaki 889-1692, Japan; Japan Agency for Medical Research and Development (AMED), 1-7-1 Otemachi, Chiyoda-ku, Tokyo 100-0004, Japan
| | - Tomofumi Uto
- Division of Immunology, Department of Infectious Diseases, Faculty of Medicine, University of Miyazaki, 5200 Kihara, Kiyotake, Miyazaki 889-1692, Japan; Japan Agency for Medical Research and Development (AMED), 1-7-1 Otemachi, Chiyoda-ku, Tokyo 100-0004, Japan
| | - Shuya Mitoma
- Division of Immunology, Department of Infectious Diseases, Faculty of Medicine, University of Miyazaki, 5200 Kihara, Kiyotake, Miyazaki 889-1692, Japan; Japan Agency for Medical Research and Development (AMED), 1-7-1 Otemachi, Chiyoda-ku, Tokyo 100-0004, Japan
| | - Hideaki Takagi
- Division of Immunology, Department of Infectious Diseases, Faculty of Medicine, University of Miyazaki, 5200 Kihara, Kiyotake, Miyazaki 889-1692, Japan; Japan Agency for Medical Research and Development (AMED), 1-7-1 Otemachi, Chiyoda-ku, Tokyo 100-0004, Japan
| | - Yotaro Nishikawa
- Division of Immunology, Department of Infectious Diseases, Faculty of Medicine, University of Miyazaki, 5200 Kihara, Kiyotake, Miyazaki 889-1692, Japan; Department of Dermatology, Faculty of Medicine, University of Miyazaki, 5200 Kihara, Kiyotake, Miyazaki 889-1692, Japan
| | - Moe Tominaga
- Division of Immunology, Department of Infectious Diseases, Faculty of Medicine, University of Miyazaki, 5200 Kihara, Kiyotake, Miyazaki 889-1692, Japan; Department of Oral and Maxillofacial Surgery, Faculty of Medicine, University of Miyazaki, 5200 Kihara, Kiyotake, Miyazaki 889-1692, Japan
| | - Narantsog Choijookhuu
- Division of Histochemistry and Cell Biology, Department of Anatomy, Faculty of Medicine, University of Miyazaki, Miyazaki 889-1692, Japan
| | - Yoshitaka Hishikawa
- Division of Histochemistry and Cell Biology, Department of Anatomy, Faculty of Medicine, University of Miyazaki, Miyazaki 889-1692, Japan
| | - Katsuaki Sato
- Division of Immunology, Department of Infectious Diseases, Faculty of Medicine, University of Miyazaki, 5200 Kihara, Kiyotake, Miyazaki 889-1692, Japan; Japan Agency for Medical Research and Development (AMED), 1-7-1 Otemachi, Chiyoda-ku, Tokyo 100-0004, Japan; Frontier Science Research Center, University of Miyazaki, 5200 Kihara, Kiyotake, Miyazaki 889-1692, Japan.
| |
Collapse
|
25
|
Molostvov G, Gachechiladze M, Shaaban AM, Hayward S, Dean I, Dias IHK, Badr N, Danial I, Mohammed F, Novitskaya V, Paniushkina L, Speirs V, Hanby A, Nazarenko I, Withers DR, van Laere S, Long HM, Berditchevski F. Tspan6 stimulates the chemoattractive potential of breast cancer cells for B cells in an EV- and LXR-dependent manner. Cell Rep 2023; 42:112207. [PMID: 36867531 DOI: 10.1016/j.celrep.2023.112207] [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: 06/06/2022] [Revised: 01/11/2023] [Accepted: 02/15/2023] [Indexed: 03/04/2023] Open
Abstract
The immune microenvironment in breast cancer (BCa) is controlled by a complex network of communication between various cell types. Here, we find that recruitment of B lymphocytes to BCa tissues is controlled via mechanisms associated with cancer cell-derived extracellular vesicles (CCD-EVs). Gene expression profiling identifies the Liver X receptor (LXR)-dependent transcriptional network as a key pathway that controls both CCD-EVs-induced migration of B cells and accumulation of B cells in BCa tissues. The increased accumulation oxysterol ligands for LXR (i.e., 25-hydroxycholesterol and 27-hydroxycholesterol) in CCD-EVs is regulated by the tetraspanin 6 (Tspan6). Tspan6 stimulates the chemoattractive potential of BCa cells for B cells in an EV- and LXR-dependent manner. These results demonstrate that tetraspanins control intercellular trafficking of oxysterols via CCD-EVs. Furthermore, tetraspanin-dependent changes in the oxysterol composition of CCD-EVs and the LXR signaling axis play a key role in specific changes in the tumor immune microenvironment.
Collapse
Affiliation(s)
- Guerman Molostvov
- Institute of Cancer and Genomic Sciences, The University of Birmingham, Edgbaston, Birmingham B15 2TT, UK
| | - Mariam Gachechiladze
- Institute of Cancer and Genomic Sciences, The University of Birmingham, Edgbaston, Birmingham B15 2TT, UK; Department of Clinical and Molecular Pathology, Palacky Univerzity, 7779 00 Olomouc, Czech Republic
| | - Abeer M Shaaban
- Institute of Cancer and Genomic Sciences, The University of Birmingham, Edgbaston, Birmingham B15 2TT, UK
| | - Steven Hayward
- Institute of Cancer and Genomic Sciences, The University of Birmingham, Edgbaston, Birmingham B15 2TT, UK
| | - Isaac Dean
- Institute of Immunology and Immunotherapy, The University of Birmingham, Edgbaston, Birmingham B15 2TT, UK
| | - Irundika H K Dias
- Aston Medical Research Institute, Aston Medical School, Aston University, Birmingham B4 7ET, UK
| | - Nahla Badr
- Institute of Cancer and Genomic Sciences, The University of Birmingham, Edgbaston, Birmingham B15 2TT, UK; Department of Pathology, Faculty of Medicine, Menoufia University, Shebin El-Kom, Egypt
| | - Irini Danial
- Institute of Cancer and Genomic Sciences, The University of Birmingham, Edgbaston, Birmingham B15 2TT, UK
| | - Fiyaz Mohammed
- Institute of Immunology and Immunotherapy, The University of Birmingham, Edgbaston, Birmingham B15 2TT, UK
| | - Vera Novitskaya
- Institute of Cancer and Genomic Sciences, The University of Birmingham, Edgbaston, Birmingham B15 2TT, UK
| | - Liliia Paniushkina
- Faculty of Medicine, Institute for Infection Prevention and Hospital Epidemiology, Medical Center - University of Freiburg, 79106 Freiburg, Germany
| | - Valerie Speirs
- Leeds Institute of Medical Research, University of Leeds, St James's University Hospital, Leeds LS9 7TF, UK; Institute of Medical Sciences, University of Aberdeen, Aberdeen AB25 2ZD, UK
| | - Andrew Hanby
- Leeds Institute of Medical Research, University of Leeds, St James's University Hospital, Leeds LS9 7TF, UK
| | - Irina Nazarenko
- Faculty of Medicine, Institute for Infection Prevention and Hospital Epidemiology, Medical Center - University of Freiburg, 79106 Freiburg, Germany; German Cancer Consortium (DKTK), Partner Site Freiburg and German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany
| | - David R Withers
- Institute of Immunology and Immunotherapy, The University of Birmingham, Edgbaston, Birmingham B15 2TT, UK
| | - Steven van Laere
- Translational Cancer Research Unit Center for Oncological Research, University Antwerp, Antwerp 2610, Belgium
| | - Heather M Long
- Institute of Immunology and Immunotherapy, The University of Birmingham, Edgbaston, Birmingham B15 2TT, UK.
| | - Fedor Berditchevski
- Institute of Cancer and Genomic Sciences, The University of Birmingham, Edgbaston, Birmingham B15 2TT, UK.
| |
Collapse
|
26
|
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
|
27
|
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
|
28
|
Mincham KT, Snelgrove RJ. OMIP-086: Full spectrum flow cytometry for high-dimensional immunophenotyping of mouse innate lymphoid cells. Cytometry A 2023; 103:110-116. [PMID: 36331092 PMCID: PMC10953369 DOI: 10.1002/cyto.a.24702] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2022] [Revised: 10/17/2022] [Accepted: 10/24/2022] [Indexed: 11/06/2022]
Abstract
This 25-parameter, 22-color full spectrum flow cytometry panel was designed and optimized for the comprehensive enumeration and functional characterization of innate lymphoid cell (ILC) subsets in mouse tissues. The panel presented here allows the discrimination of ILC progenitors (ILCP), ILC1, ILC2, NCR+ ILC3, NCR- ILC3, CCR6+ lymphoid tissue-inducer (LTi)-like ILC3 and mature natural killer (NK) cell populations. Further characterization of ILC and NK cell functional profiles in response to stimulation is provided by the inclusion of subset-specific cytokine markers, and proliferation markers. Development and optimization of this panel was performed on freshly isolated cells from adult BALB/c lungs and small intestine lamina propria, and ex vivo stimulation with phorbol 12-myrisate 13-acetate, ionomycin, and pro-ILC activating cytokines.
Collapse
Affiliation(s)
- Kyle T. Mincham
- National Heart and Lung InstituteImperial College LondonLondonUK
| | | |
Collapse
|
29
|
Ware MB, Wolfarth AA, Goon JB, Ezeanya UI, Dhar S, Ferrando-Martinez S, Lee BH. The Role of Interleukin-7 in the Formation of Tertiary Lymphoid Structures and Their Prognostic Value in Gastrointestinal Cancers. JOURNAL OF IMMUNOTHERAPY AND PRECISION ONCOLOGY 2022; 5:105-117. [PMID: 36483588 PMCID: PMC9714415 DOI: 10.36401/jipo-22-10] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/19/2022] [Revised: 08/19/2022] [Accepted: 08/25/2022] [Indexed: 06/17/2023]
Abstract
Immunotherapies for the treatment of solid tumors continue to develop in preclinical and clinical research settings. Unfortunately, for many patients the tumor fails to respond or becomes resistant to therapies such as checkpoint inhibitors (CPIs) targeting programmed cell death protein-1 (PD-1), programmed death-ligand 1 (PD-L1), and cytotoxic T lymphocyte antigen-4 (CTLA-4). In many cancers, failed response to CPIs can be attributed to poor T cell infiltration, dominant immunosuppression, and exhausted immune responses. In gastrointestinal (GI) cancers T cell infiltration can be dismal, with several reports finding that CD8+ T cells compose less than 2% of all cells within the tumor. Organized aggregates of lymphocytes, antigen-presenting cells, and vessels, together termed tertiary lymphoid structures (TLSs), are hypothesized to be a major source of T cells within solid tumors. The intratumoral formation of these organized immune centers appears to rely on intricate cytokine and chemokine signaling to heterogeneous cell populations such as B and T cells, innate lymphoid cells, fibroblasts, and dendritic cells. In GI cancers, the presence and density of TLSs provide prognostic value for predicting outcome and survival. Further, TLS presence and density associates with favorable responses to CPIs in many cancers. This review highlights the prognostic value of TLSs in GI cancers, the role of the homeostatic cytokine interleukin-7 (IL-7) in TLS formation, and the induction of TLSs in solid tumors by novel therapeutics.
Collapse
|
30
|
Kedmi R, Najar TA, Mesa KR, Grayson A, Kroehling L, Hao Y, Hao S, Pokrovskii M, Xu M, Talbot J, Wang J, Germino J, Lareau CA, Satpathy AT, Anderson MS, Laufer TM, Aifantis I, Bartleson JM, Allen PM, Paidassi H, Gardner JM, Stoeckius M, Littman DR. A RORγt + cell instructs gut microbiota-specific T reg cell differentiation. Nature 2022; 610:737-743. [PMID: 36071167 PMCID: PMC9908423 DOI: 10.1038/s41586-022-05089-y] [Citation(s) in RCA: 120] [Impact Index Per Article: 40.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2021] [Accepted: 07/08/2022] [Indexed: 01/19/2023]
Abstract
The mutualistic relationship of gut-resident microbiota and the host immune system promotes homeostasis that ensures maintenance of the microbial community and of a largely non-aggressive immune cell compartment1,2. The consequences of disturbing this balance include proximal inflammatory conditions, such as Crohn's disease, and systemic illnesses. This equilibrium is achieved in part through the induction of both effector and suppressor arms of the adaptive immune system. Helicobacter species induce T regulatory (Treg) and T follicular helper (TFH) cells under homeostatic conditions, but induce inflammatory T helper 17 (TH17) cells when induced Treg (iTreg) cells are compromised3,4. How Helicobacter and other gut bacteria direct T cells to adopt distinct functions remains poorly understood. Here we investigated the cells and molecular components required for iTreg cell differentiation. We found that antigen presentation by cells expressing RORγt, rather than by classical dendritic cells, was required and sufficient for induction of Treg cells. These RORγt+ cells-probably type 3 innate lymphoid cells and/or Janus cells5-require the antigen-presentation machinery, the chemokine receptor CCR7 and the TGFβ activator αv integrin. In the absence of any of these factors, there was expansion of pathogenic TH17 cells instead of iTreg cells, induced by CCR7-independent antigen-presenting cells. Thus, intestinal commensal microbes and their products target multiple antigen-presenting cells with pre-determined features suited to directing appropriate T cell differentiation programmes, rather than a common antigen-presenting cell that they endow with appropriate functions.
Collapse
Affiliation(s)
- Ranit Kedmi
- Molecular Pathogenesis Program, The Kimmel Center for Biology and Medicine of the Skirball Institute, New York University School of Medicine, New York, NY, USA
| | - Tariq A Najar
- Molecular Pathogenesis Program, The Kimmel Center for Biology and Medicine of the Skirball Institute, New York University School of Medicine, New York, NY, USA
| | - Kailin R Mesa
- Molecular Pathogenesis Program, The Kimmel Center for Biology and Medicine of the Skirball Institute, New York University School of Medicine, New York, NY, USA
| | - Allyssa Grayson
- Molecular Pathogenesis Program, The Kimmel Center for Biology and Medicine of the Skirball Institute, New York University School of Medicine, New York, NY, USA.,Howard Hughes Medical Institute, New York, NY, USA
| | - Lina Kroehling
- Molecular Pathogenesis Program, The Kimmel Center for Biology and Medicine of the Skirball Institute, New York University School of Medicine, New York, NY, USA
| | - Yuhan Hao
- Center for Genomics and Systems Biology, New York University, New York, NY, USA.,New York Genome Center, New York, NY, USA
| | - Stephanie Hao
- Technology Innovation Lab, New York Genome Center, New York, NY, USA
| | - Maria Pokrovskii
- Molecular Pathogenesis Program, The Kimmel Center for Biology and Medicine of the Skirball Institute, New York University School of Medicine, New York, NY, USA.,Calico Life Sciences, LLC, South San Francisco, CA, USA
| | - Mo Xu
- Molecular Pathogenesis Program, The Kimmel Center for Biology and Medicine of the Skirball Institute, New York University School of Medicine, New York, NY, USA.,National Institute for Biological Sciences, Beijing, China
| | - Jhimmy Talbot
- Molecular Pathogenesis Program, The Kimmel Center for Biology and Medicine of the Skirball Institute, New York University School of Medicine, New York, NY, USA.,Fred Hutchinson Cancer Center, Seattle, WA, USA
| | - Jiaxi Wang
- Diabetes Center, University of California, San Francisco, San Francisco, CA, USA
| | - Joe Germino
- Diabetes Center, University of California, San Francisco, San Francisco, CA, USA
| | - Caleb A Lareau
- Department of Pathology, Stanford University, Stanford, CA, USA.,Parker Institute for Cancer Immunotherapy, Stanford University, Stanford, CA, USA.,Gladstone-UCSF Institute of Genomic Immunology, San Francisco, CA, USA
| | - Ansuman T Satpathy
- Department of Pathology, Stanford University, Stanford, CA, USA.,Parker Institute for Cancer Immunotherapy, Stanford University, Stanford, CA, USA.,Gladstone-UCSF Institute of Genomic Immunology, San Francisco, CA, USA
| | - Mark S Anderson
- Diabetes Center, University of California, San Francisco, San Francisco, CA, USA
| | - Terri M Laufer
- Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA.,Department of Medicine, C. Michael Crescenz Veterans Administration Medical Center, Philadelphia, PA, USA
| | - Iannis Aifantis
- Department of Pathology, New York University School of Medicine, New York, NY, USA
| | - Juliet M Bartleson
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO, USA.,Federation Bio, South San Francisco, CA, USA
| | - Paul M Allen
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO, USA
| | - Helena Paidassi
- CIRI, Centre International de Recherche en Infectiologie, Université de Lyon, INSERM U1111, Université Claude Bernard Lyon 1, CNRS UMR5308, ENS de Lyon, Lyon, France
| | - James M Gardner
- Diabetes Center, University of California, San Francisco, San Francisco, CA, USA.,Department of Surgery, University of California, San Francisco, San Francisco, CA, USA
| | - Marlon Stoeckius
- Technology Innovation Lab, New York Genome Center, New York, NY, USA.,10X Genomics, Stockholm, Sweden
| | - Dan R Littman
- Molecular Pathogenesis Program, The Kimmel Center for Biology and Medicine of the Skirball Institute, New York University School of Medicine, New York, NY, USA. .,Howard Hughes Medical Institute, New York, NY, USA.
| |
Collapse
|
31
|
Lyu M, Suzuki H, Kang L, Gaspal F, Zhou W, Goc J, Zhou L, Zhou J, Zhang W, Shen Z, Fox JG, Sockolow RE, Laufer TM, Fan Y, Eberl G, Withers DR, Sonnenberg GF. ILC3s select microbiota-specific regulatory T cells to establish tolerance in the gut. Nature 2022; 610:744-751. [PMID: 36071169 PMCID: PMC9613541 DOI: 10.1038/s41586-022-05141-x] [Citation(s) in RCA: 141] [Impact Index Per Article: 47.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2021] [Accepted: 07/25/2022] [Indexed: 02/07/2023]
Abstract
Microbial colonization of the mammalian intestine elicits inflammatory or tolerogenic T cell responses, but the mechanisms controlling these distinct outcomes remain poorly understood, and accumulating evidence indicates that aberrant immunity to intestinal microbiota is causally associated with infectious, inflammatory and malignant diseases1-8. Here we define a critical pathway controlling the fate of inflammatory versus tolerogenic T cells that respond to the microbiota and express the transcription factor RORγt. We profiled all RORγt+ immune cells at single-cell resolution from the intestine-draining lymph nodes of mice and reveal a dominant presence of T regulatory (Treg) cells and lymphoid tissue inducer-like group 3 innate lymphoid cells (ILC3s), which co-localize at interfollicular regions. These ILC3s are distinct from extrathymic AIRE-expressing cells, abundantly express major histocompatibility complex class II, and are necessary and sufficient to promote microbiota-specific RORγt+ Treg cells and prevent their expansion as inflammatory T helper 17 cells. This occurs through ILC3-mediated antigen presentation, αV integrin and competition for interleukin-2. Finally, single-cell analyses suggest that interactions between ILC3s and RORγt+ Treg cells are impaired in inflammatory bowel disease. Our results define a paradigm whereby ILC3s select for antigen-specific RORγt+ Treg cells, and against T helper 17 cells, to establish immune tolerance to the microbiota and intestinal health.
Collapse
Affiliation(s)
- Mengze Lyu
- Joan and Sanford I. Weill Department of Medicine, Division of Gastroenterology and Hepatology, Weill Cornell Medicine, Cornell University, New York, NY, USA
- Department of Microbiology and Immunology, Weill Cornell Medicine, Cornell University, New York, NY, USA
- Jill Roberts Institute for Research in Inflammatory Bowel Disease, Weill Cornell Medicine, Cornell University, New York, NY, USA
| | - Hiroaki Suzuki
- Joan and Sanford I. Weill Department of Medicine, Division of Gastroenterology and Hepatology, Weill Cornell Medicine, Cornell University, New York, NY, USA
- Department of Microbiology and Immunology, Weill Cornell Medicine, Cornell University, New York, NY, USA
- Jill Roberts Institute for Research in Inflammatory Bowel Disease, Weill Cornell Medicine, Cornell University, New York, NY, USA
- EA Pharma, Kanagawa, Japan
| | - Lan Kang
- Joan and Sanford I. Weill Department of Medicine, Division of Gastroenterology and Hepatology, Weill Cornell Medicine, Cornell University, New York, NY, USA
- Department of Microbiology and Immunology, Weill Cornell Medicine, Cornell University, New York, NY, USA
- Jill Roberts Institute for Research in Inflammatory Bowel Disease, Weill Cornell Medicine, Cornell University, New York, NY, USA
| | - Fabrina Gaspal
- Institute of Immunology and Immunotherapy, College of Medical and Dental Sciences, University of Birmingham, Birmingham, UK
| | - Wenqing Zhou
- Joan and Sanford I. Weill Department of Medicine, Division of Gastroenterology and Hepatology, Weill Cornell Medicine, Cornell University, New York, NY, USA
- Department of Microbiology and Immunology, Weill Cornell Medicine, Cornell University, New York, NY, USA
- Jill Roberts Institute for Research in Inflammatory Bowel Disease, Weill Cornell Medicine, Cornell University, New York, NY, USA
| | - Jeremy Goc
- Joan and Sanford I. Weill Department of Medicine, Division of Gastroenterology and Hepatology, Weill Cornell Medicine, Cornell University, New York, NY, USA
- Department of Microbiology and Immunology, Weill Cornell Medicine, Cornell University, New York, NY, USA
- Jill Roberts Institute for Research in Inflammatory Bowel Disease, Weill Cornell Medicine, Cornell University, New York, NY, USA
| | - Lei Zhou
- Joan and Sanford I. Weill Department of Medicine, Division of Gastroenterology and Hepatology, Weill Cornell Medicine, Cornell University, New York, NY, USA
- Department of Microbiology and Immunology, Weill Cornell Medicine, Cornell University, New York, NY, USA
- Jill Roberts Institute for Research in Inflammatory Bowel Disease, Weill Cornell Medicine, Cornell University, New York, NY, USA
| | - Jordan Zhou
- Joan and Sanford I. Weill Department of Medicine, Division of Gastroenterology and Hepatology, Weill Cornell Medicine, Cornell University, New York, NY, USA
- Department of Microbiology and Immunology, Weill Cornell Medicine, Cornell University, New York, NY, USA
- Jill Roberts Institute for Research in Inflammatory Bowel Disease, Weill Cornell Medicine, Cornell University, New York, NY, USA
| | - Wen Zhang
- Joan and Sanford I. Weill Department of Medicine, Division of Gastroenterology and Hepatology, Weill Cornell Medicine, Cornell University, New York, NY, USA
- Department of Microbiology and Immunology, Weill Cornell Medicine, Cornell University, New York, NY, USA
- Jill Roberts Institute for Research in Inflammatory Bowel Disease, Weill Cornell Medicine, Cornell University, New York, NY, USA
| | - Zeli Shen
- Division of Comparative Medicine, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - James G Fox
- Division of Comparative Medicine, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Robbyn E Sockolow
- Department of Pediatrics, Division of Gastroenterology, Hepatology, and Nutrition, Weill Cornell Medicine, Cornell University, New York, NY, USA
| | - Terri M Laufer
- Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Philadelphia Veterans Affairs Medical Center, Philadelphia, PA, USA
| | - Yong Fan
- Institute of Cellular Therapeutics, Allegheny Health Network, Pittsburgh, PA, USA
| | - Gerard Eberl
- Microenvironment and Immunity Unit, Institut Pasteur, Paris, France
| | - David R Withers
- Institute of Immunology and Immunotherapy, College of Medical and Dental Sciences, University of Birmingham, Birmingham, UK
| | - Gregory F Sonnenberg
- Joan and Sanford I. Weill Department of Medicine, Division of Gastroenterology and Hepatology, Weill Cornell Medicine, Cornell University, New York, NY, USA.
- Department of Microbiology and Immunology, Weill Cornell Medicine, Cornell University, New York, NY, USA.
- Jill Roberts Institute for Research in Inflammatory Bowel Disease, Weill Cornell Medicine, Cornell University, New York, NY, USA.
| |
Collapse
|
32
|
Inflammation triggers ILC3 patrolling of the intestinal barrier. Nat Immunol 2022; 23:1317-1323. [PMID: 35999393 PMCID: PMC9477741 DOI: 10.1038/s41590-022-01284-1] [Citation(s) in RCA: 32] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2021] [Accepted: 07/07/2022] [Indexed: 11/09/2022]
Abstract
An orchestrated cellular network, including adaptive lymphocytes and group 3 innate lymphoid cells (ILC3s), maintains intestinal barrier integrity and homeostasis. T cells can monitor environmental insults through constitutive circulation, scanning tissues and forming immunological contacts, a process named immunosurveillance. In contrast, the dynamics of intestinal ILC3s are unknown. Using intravital imaging, we observed that villus ILC3s were largely immotile at steady state but acquired migratory ‘patrolling’ attributes and enhanced cytokine expression in response to inflammation. We showed that T cells, the chemokine CCL25 and bacterial ligands regulated intestinal ILC3 behavior and that loss of patrolling behavior by interleukin-22 (IL-22)-producing ILC3s altered the intestinal barrier through increased epithelial cell death. Collectively, we identified notable differences between the behavior of ILC3s and T cells, with a prominent adaptation of intestinal ILC3s toward mucosal immunosurveillance after inflammation. Serafini and colleagues show that intestinal villus ILC3s, which are largely immotile at steady state, develop a patrolling behavior in response to inflammation.
Collapse
|
33
|
Kabil A, Shin SB, Hughes MR, McNagny KM. “Just one word, plastic!”: Controversies and caveats in innate lymphoid cell plasticity. Front Immunol 2022; 13:946905. [PMID: 36052086 PMCID: PMC9427196 DOI: 10.3389/fimmu.2022.946905] [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: 05/18/2022] [Accepted: 07/26/2022] [Indexed: 11/13/2022] Open
Abstract
Innate lymphoid cells (ILCs) are frontline immune effectors involved in the early stages of host defense and maintenance of tissue homeostasis, particularly at mucosal surfaces such as the intestine, lung, and skin. Canonical ILCs are described as tissue-resident cells that populate peripheral tissues early in life and respond appropriately based on environmental exposure and their anatomical niche and tissue microenvironment. Intriguingly, there are accumulating reports of ILC “plasticity” that note the existence of non-canonical ILCs that exhibit distinct patterns of master transcription factor expression and cytokine production profiles in response to tissue inflammation. Yet this concept of ILC-plasticity is controversial due to several confounding caveats that include, among others, the independent large-scale recruitment of new ILC subsets from distal sites and the local, in situ, differentiation of uncommitted resident precursors. Nevertheless, the ability of ILCs to acquire unique characteristics and adapt to local environmental cues is an attractive paradigm because it would enable the rapid adaptation of innate responses to a wider array of pathogens even in the absence of pre-existing ‘prototypical’ ILC responder subsets. Despite the impressive recent progress in understanding ILC biology, the true contribution of ILC plasticity to tissue homeostasis and disease and how it is regulated remains obscure. Here, we detail current methodologies used to study ILC plasticity in mice and review the mechanisms that drive and regulate functional ILC plasticity in response to polarizing signals in their microenvironment and different cytokine milieus. Finally, we discuss the physiological relevance of ILC plasticity and its implications for potential therapeutics and treatments.
Collapse
Affiliation(s)
- Ahmed Kabil
- School of Biomedical Engineering, University of British Columbia, Vancouver, BC, Canada
| | - Samuel B. Shin
- School of Biomedical Engineering, University of British Columbia, Vancouver, BC, Canada
| | - Michael R. Hughes
- School of Biomedical Engineering, University of British Columbia, Vancouver, BC, Canada
| | - Kelly M. McNagny
- School of Biomedical Engineering, University of British Columbia, Vancouver, BC, Canada
- Department of Medical Genetics, University of British Columbia, Vancouver, BC, Canada
- Centre for Heart and Lung Innovation (HLI), St Paul’s Hospital, University of British Columbia, Vancouver, BC, Canada
- *Correspondence: Kelly M. McNagny,
| |
Collapse
|
34
|
Richard AC. Divide and Conquer: Phenotypic and Temporal Heterogeneity Within CD8 + T Cell Responses. Front Immunol 2022; 13:949423. [PMID: 35911755 PMCID: PMC9334874 DOI: 10.3389/fimmu.2022.949423] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2022] [Accepted: 06/22/2022] [Indexed: 11/23/2022] Open
Abstract
The advent of technologies that can characterize the phenotypes, functions and fates of individual cells has revealed extensive and often unexpected levels of diversity between cells that are nominally of the same subset. CD8+ T cells, also known as cytotoxic T lymphocytes (CTLs), are no exception. Investigations of individual CD8+ T cells both in vitro and in vivo have highlighted the heterogeneity of cellular responses at the levels of activation, differentiation and function. This review takes a broad perspective on the topic of heterogeneity, outlining different forms of variation that arise during a CD8+ T cell response. Specific attention is paid to the impact of T cell receptor (TCR) stimulation strength on heterogeneity. In particular, this review endeavors to highlight connections between variation at different cellular stages, presenting known mechanisms and key open questions about how variation between cells can arise and propagate.
Collapse
|
35
|
Whyte CE, Singh K, Burton OT, Aloulou M, Kouser L, Veiga RV, Dashwood A, Okkenhaug H, Benadda S, Moudra A, Bricard O, Lienart S, Bielefeld P, Roca CP, Naranjo-Galindo FJ, Lombard-Vadnais F, Junius S, Bending D, Ono M, Hochepied T, Halim TY, Schlenner S, Lesage S, Dooley J, Liston A. Context-dependent effects of IL-2 rewire immunity into distinct cellular circuits. J Exp Med 2022; 219:e20212391. [PMID: 35699942 PMCID: PMC9202720 DOI: 10.1084/jem.20212391] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2021] [Revised: 04/06/2022] [Accepted: 05/16/2022] [Indexed: 12/17/2022] Open
Abstract
Interleukin 2 (IL-2) is a key homeostatic cytokine, with therapeutic applications in both immunogenic and tolerogenic immune modulation. Clinical use has been hampered by pleiotropic functionality and widespread receptor expression, with unexpected adverse events. Here, we developed a novel mouse strain to divert IL-2 production, allowing identification of contextual outcomes. Network analysis identified priority access for Tregs and a competitive fitness cost of IL-2 production among both Tregs and conventional CD4 T cells. CD8 T and NK cells, by contrast, exhibited a preference for autocrine IL-2 production. IL-2 sourced from dendritic cells amplified Tregs, whereas IL-2 produced by B cells induced two context-dependent circuits: dramatic expansion of CD8+ Tregs and ILC2 cells, the latter driving a downstream, IL-5-mediated, eosinophilic circuit. The source-specific effects demonstrate the contextual influence of IL-2 function and potentially explain adverse effects observed during clinical trials. Targeted IL-2 production therefore has the potential to amplify or quench particular circuits in the IL-2 network, based on clinical desirability.
Collapse
Affiliation(s)
- Carly E. Whyte
- Immunology Programme, The Babraham Institute, Cambridge, UK
| | - Kailash Singh
- Immunology Programme, The Babraham Institute, Cambridge, UK
| | - Oliver T. Burton
- Immunology Programme, The Babraham Institute, Cambridge, UK
- VIB Center for Brain and Disease Research, Vlaams Instituut voor Biotechnologie, Leuven, Belgium
- Department of Microbiology, Immunology and Transplantation, KU Leuven—University of Leuven, Leuven, Belgium
| | - Meryem Aloulou
- Immunology Programme, The Babraham Institute, Cambridge, UK
- Toulouse Institute for Infectious and Inflammatory Diseases (Infinity), Centre national de la recherche scientifique U5051, Institut national de la santé et de la recherche médicale U1291, University of Toulouse III, Toulouse, France
| | - Lubna Kouser
- Immunology Programme, The Babraham Institute, Cambridge, UK
| | | | - Amy Dashwood
- Immunology Programme, The Babraham Institute, Cambridge, UK
| | | | - Samira Benadda
- Immunology Programme, The Babraham Institute, Cambridge, UK
- Centre de Recherche Sur L’inflammation, Centre national de la recherche scientifique ERL8252, Institut national de la santé et de la recherche médicale U1149, Université de Paris, Paris, France
| | - Alena Moudra
- Immunology Programme, The Babraham Institute, Cambridge, UK
| | - Orian Bricard
- Immunology Programme, The Babraham Institute, Cambridge, UK
| | | | | | - Carlos P. Roca
- Immunology Programme, The Babraham Institute, Cambridge, UK
| | | | - Félix Lombard-Vadnais
- Department of Microbiology and Immunology, McGill University, Montréal, Quebec, Canada
- Department of Immunology-Oncology, Maisonneuve-Rosemont Hospital, Montréal, Quebec, Canada
| | - Steffie Junius
- VIB Center for Brain and Disease Research, Vlaams Instituut voor Biotechnologie, Leuven, Belgium
- Department of Microbiology, Immunology and Transplantation, KU Leuven—University of Leuven, Leuven, Belgium
| | - David Bending
- Institute of Immunology and Immunotherapy, College of Medical and Dental Sciences, University of Birmingham, Birmingham, UK
| | - Masahiro Ono
- Department of Life Sciences, Imperial College London, London, UK
| | - Tino Hochepied
- Department of Biomedical Molecular Biology, Ghent University, Ghent, Belgium
- VIB Center for Inflammation Research, Vlaams Instituut voor Biotechnologie, Ghent, Belgium
| | | | - Susan Schlenner
- Department of Microbiology, Immunology and Transplantation, KU Leuven—University of Leuven, Leuven, Belgium
| | - Sylvie Lesage
- Centre de Recherche Sur L’inflammation, Centre national de la recherche scientifique ERL8252, Institut national de la santé et de la recherche médicale U1149, Université de Paris, Paris, France
- Département de Microbiologie, Infectiologie et Immunologie, Université de Montréal, Montréal, Quebec, Canada
| | - James Dooley
- Immunology Programme, The Babraham Institute, Cambridge, UK
- VIB Center for Brain and Disease Research, Vlaams Instituut voor Biotechnologie, Leuven, Belgium
- Department of Microbiology, Immunology and Transplantation, KU Leuven—University of Leuven, Leuven, Belgium
| | - Adrian Liston
- Immunology Programme, The Babraham Institute, Cambridge, UK
- VIB Center for Brain and Disease Research, Vlaams Instituut voor Biotechnologie, Leuven, Belgium
- Department of Microbiology, Immunology and Transplantation, KU Leuven—University of Leuven, Leuven, Belgium
| |
Collapse
|
36
|
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
|
37
|
Rethacker L, Boy M, Bisio V, Roussin F, Denizeau J, Vincent-Salomon A, Borcoman E, Sedlik C, Piaggio E, Toubert A, Dulphy N, Caignard A. Innate lymphoid cells: NK and cytotoxic ILC3 subsets infiltrate metastatic breast cancer lymph nodes. Oncoimmunology 2022; 11:2057396. [PMID: 35371620 PMCID: PMC8973349 DOI: 10.1080/2162402x.2022.2057396] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022] Open
Abstract
Innate lymphoid cells (ILCs) – which include cytotoxic Natural Killer (NK) cells and helper-type ILC – are important regulators of tissue immune homeostasis, with possible roles in tumor surveillance. We analyzed ILC and their functionality in human lymph nodes (LN). In LN, NK cells and ILC3 were the prominent subpopulations. Among the ILC3s, we identified a CD56+/ILC3 subset with a phenotype close to ILC3 but also expressing cytotoxicity genes shared with NK. In tumor-draining LNs (TD-LNs) and tumor samples from breast cancer (BC) patients, NK cells were prominent, and proportions of ILC3 subsets were low. In tumors and TD-LN, NK cells display reduced levels of NCR (Natural cytotoxicity receptors), despite high transcript levels and included a small subset CD127− CD56− NK cells with reduced function. Activated by cytokines CD56+/ILC3 cells from donor and patients LN acquired cytotoxic capacity and produced IFNg. In TD-LN, all cytokine activated ILC populations produced TNFα in response to BC cell line. Analyses of cytotoxic and helper ILC indicate a switch toward NK cells in TD-LN. The local tumor microenvironment inhibited NK cell functions through downregulation of NCR, but cytokine stimulation restored their functionality.
Collapse
Affiliation(s)
- Louise Rethacker
- INSERM U1160, Institut de Recherche Saint-Louis, Hôpital Saint Louis, Paris, France
| | - Maxime Boy
- INSERM U1160, Institut de Recherche Saint-Louis, Hôpital Saint Louis, Paris, France
| | - Valeria Bisio
- INSERM U1160, Institut de Recherche Saint-Louis, Hôpital Saint Louis, Paris, France
| | - France Roussin
- Service d’Anesthésie-Réanimation, AP-HP, Hôpital Saint-Louis, Paris, France
| | - Jordan Denizeau
- INSERM U932, Département de Recherche Translationelle, Institut Curie, Université de Recherche Paris Sciences & Lettres (PSL), Institut National de la Santé et de la Recherche Médicale (INSERM), Paris, France
| | - Anne Vincent-Salomon
- Diagnostic and Theranostic Medicine Division, Institut Curie, PSL Research University, Paris, France
| | - Edith Borcoman
- Department of Medical Oncology, Institut Curie, Paris, France
- Université Paris Diderot, Université de Paris, Paris, France
| | - Christine Sedlik
- INSERM U932, Département de Recherche Translationelle, Institut Curie, Université de Recherche Paris Sciences & Lettres (PSL), Institut National de la Santé et de la Recherche Médicale (INSERM), Paris, France
| | - Eliane Piaggio
- INSERM U932, Département de Recherche Translationelle, Institut Curie, Université de Recherche Paris Sciences & Lettres (PSL), Institut National de la Santé et de la Recherche Médicale (INSERM), Paris, France
| | - Antoine Toubert
- INSERM U1160, Institut de Recherche Saint-Louis, Hôpital Saint Louis, Paris, France
- Université Paris Diderot, Université de Paris, Paris, France
- Assistance Publique–Hôpitaux de Paris (AP–HP), Hôpital Saint-Louis, Laboratoire d’Immunologie et Histocompatibilité, Paris, France
| | - Nicolas Dulphy
- INSERM U1160, Institut de Recherche Saint-Louis, Hôpital Saint Louis, Paris, France
- Université Paris Diderot, Université de Paris, Paris, France
- Assistance Publique–Hôpitaux de Paris (AP–HP), Hôpital Saint-Louis, Laboratoire d’Immunologie et Histocompatibilité, Paris, France
| | - Anne Caignard
- INSERM U1160, Institut de Recherche Saint-Louis, Hôpital Saint Louis, Paris, France
| |
Collapse
|
38
|
Alawam AS, Cosway EJ, James KD, Lucas B, Bacon A, Parnell SM, White AJ, Jenkinson WE, Anderson G. Failures in thymus medulla regeneration during immune recovery cause tolerance loss and prime recipients for auto-GVHD. J Exp Med 2022; 219:212911. [PMID: 34910105 PMCID: PMC8679781 DOI: 10.1084/jem.20211239] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2021] [Revised: 10/20/2021] [Accepted: 11/17/2021] [Indexed: 12/11/2022] Open
Abstract
Bone marrow transplantation (BMT) is a widely used therapy for blood cancers and primary immunodeficiency. Following transplant, the thymus plays a key role in immune reconstitution by generating a naive αβT cell pool from transplant-derived progenitors. While donor-derived thymopoiesis during the early post-transplant period is well studied, the ability of the thymus to synchronize T cell development with essential tolerance mechanisms is poorly understood. Using a syngeneic mouse transplant model, we analyzed T cell recovery alongside the regeneration and function of intrathymic microenvironments. We report a specific and prolonged failure in the post-transplant recovery of medullary thymic epithelial cells (mTECs). This manifests as loss of medulla-dependent tolerance mechanisms, including failures in Foxp3+ regulatory T cell development and formation of the intrathymic dendritic cell pool. In addition, defective negative selection enables escape of self-reactive conventional αβT cells that promote autoimmunity. Collectively, we show that post-transplant T cell recovery involves an uncoupling of thymopoiesis from thymic tolerance, which results in autoimmune reconstitution caused by failures in thymic medulla regeneration.
Collapse
Affiliation(s)
- Abdullah S Alawam
- Institute for Immunology and Immunotherapy, College of Medical and Dental Sciences, Medical School, University of Birmingham, Birmingham, UK
| | - Emilie J Cosway
- Institute for Immunology and Immunotherapy, College of Medical and Dental Sciences, Medical School, University of Birmingham, Birmingham, UK
| | - Kieran D James
- Institute for Immunology and Immunotherapy, College of Medical and Dental Sciences, Medical School, University of Birmingham, Birmingham, UK
| | - Beth Lucas
- Institute for Immunology and Immunotherapy, College of Medical and Dental Sciences, Medical School, University of Birmingham, Birmingham, UK
| | - Andrea Bacon
- Institute for Immunology and Immunotherapy, College of Medical and Dental Sciences, Medical School, University of Birmingham, Birmingham, UK
| | - Sonia M Parnell
- Institute for Immunology and Immunotherapy, College of Medical and Dental Sciences, Medical School, University of Birmingham, Birmingham, UK
| | - Andrea J White
- Institute for Immunology and Immunotherapy, College of Medical and Dental Sciences, Medical School, University of Birmingham, Birmingham, UK
| | - William E Jenkinson
- Institute for Immunology and Immunotherapy, College of Medical and Dental Sciences, Medical School, University of Birmingham, Birmingham, UK
| | - Graham Anderson
- Institute for Immunology and Immunotherapy, College of Medical and Dental Sciences, Medical School, University of Birmingham, Birmingham, UK
| |
Collapse
|
39
|
Das A, Harly C, Ding Y, Bhandoola A. ILC Differentiation from Progenitors in the Bone Marrow. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2022; 1365:7-24. [DOI: 10.1007/978-981-16-8387-9_2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/01/2022]
|
40
|
Abstract
Inflammatory bowel disease (IBD) is a chronic and nonspecific intestinal inflammatory condition with high relapse rate. Its pathogenesis has been linked to dysbacteriosis, genetic and environmental factors. In recent years, a new type of lymphocytes, termed innate lymphoid cells, has been described and classified into three subtypes of innate lymphoid cells-group 1, group 2 and group 3. An imbalance among these subsets' interaction with gut microbiome, and other immune cells affects intestinal mucosal homeostasis. Understanding the role of innate lymphoid cells may provide ideas for developing novel and targeted approaches for treatment of IBD.
Collapse
|
41
|
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
|
42
|
Teng F, Tachó-Piñot R, Sung B, Farber DL, Worgall S, Hammad H, Lambrecht BN, Hepworth MR, Sonnenberg GF. ILC3s control airway inflammation by limiting T cell responses to allergens and microbes. Cell Rep 2021; 37:110051. [PMID: 34818549 PMCID: PMC8635287 DOI: 10.1016/j.celrep.2021.110051] [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: 06/02/2021] [Revised: 09/28/2021] [Accepted: 11/02/2021] [Indexed: 02/06/2023] Open
Abstract
Group 3 innate lymphoid cells (ILC3s) critically regulate host-microbe
interactions in the gastrointestinal tract, but their role in the airway remains
poorly understood. Here, we demonstrate that lymphoid-tissue-inducer (LTi)-like
ILC3s are enriched in the lung-draining lymph nodes of healthy mice and humans.
These ILC3s abundantly express major histocompatibility complex class II (MHC
class II) and functionally restrict the expansion of allergen-specific
CD4+ T cells upon experimental airway challenge. In a mouse model
of house-dust-mite-induced allergic airway inflammation, MHC class
II+ ILC3s limit T helper type 2 (Th2) cell responses,
eosinophilia, and airway hyperresponsiveness. Furthermore, MHC class
II+ ILC3s limit a concomitant Th17 cell response and airway
neutrophilia. This exacerbated Th17 cell response requires exposure of the lung
to microbial stimuli, which can be found associated with house dust mites. These
findings demonstrate a critical role for antigen-presenting ILC3s in
orchestrating immune tolerance in the airway by restricting pro-inflammatory T
cell responses to both allergens and microbes. In this study, Teng et al. demonstrate that an innate immune cell type,
ILC3, is enriched in the lung draining lymph node of healthy humans and mice and
functions to limit airway inflammation through antigen presentation and control
of T cell responses directed against allergens or microbes.
Collapse
Affiliation(s)
- Fei Teng
- Joan and Sanford I. Weill Department of Medicine, Division of Gastroenterology, Weill Cornell Medicine, Cornell University, New York, NY, USA; Department of Microbiology and Immunology, Weill Cornell Medicine, Cornell University, New York, NY, USA; Jill Roberts Institute for Research in Inflammatory Bowel Disease, Weill Cornell Medicine, Cornell University, New York, NY, USA
| | - Roser Tachó-Piñot
- Lydia Becker Institute of Immunology and Inflammation, University of Manchester, Manchester, UK; Division of Infection, Immunity and Respiratory Medicine, School of Biological Sciences, Manchester Academic Health Science Centre, University of Manchester, Manchester, UK
| | - Biin Sung
- Department of Pediatrics, Weill Cornell Medicine, New York, New York, USA
| | - Donna L Farber
- Columbia Center for Translational Immunology and Departments of Surgery and Microbiology and Immunology, Columbia University Medical Center, New York, New York, USA
| | - Stefan Worgall
- Department of Pediatrics, Weill Cornell Medicine, New York, New York, USA; Department of Genetic Medicine, Weill Cornell Medicine, New York, New York, USA; Drukier Institute for Children's Health, Weill Cornell Medicine, New York, New York, USA
| | - Hamida Hammad
- Laboratory of Immunoregulation and Mucosal Immunology, VIB-UGent Center for Inflammation Research, Ghent, Belgium; Department of Internal Medicine and Pediatrics, Ghent University, Ghent, Belgium
| | - Bart N Lambrecht
- Laboratory of Immunoregulation and Mucosal Immunology, VIB-UGent Center for Inflammation Research, Ghent, Belgium; Department of Internal Medicine and Pediatrics, Ghent University, Ghent, Belgium; Department of Pulmonary Medicine, Erasmus University Medical Center Rotterdam, Rotterdam, the Netherlands
| | - Matthew R Hepworth
- Lydia Becker Institute of Immunology and Inflammation, University of Manchester, Manchester, UK; Division of Infection, Immunity and Respiratory Medicine, School of Biological Sciences, Manchester Academic Health Science Centre, University of Manchester, Manchester, UK.
| | - Gregory F Sonnenberg
- Joan and Sanford I. Weill Department of Medicine, Division of Gastroenterology, Weill Cornell Medicine, Cornell University, New York, NY, USA; Department of Microbiology and Immunology, Weill Cornell Medicine, Cornell University, New York, NY, USA; Jill Roberts Institute for Research in Inflammatory Bowel Disease, Weill Cornell Medicine, Cornell University, New York, NY, USA.
| |
Collapse
|
43
|
Yu HB, Yang H, Allaire JM, Ma C, Graef FA, Mortha A, Liang Q, Bosman ES, Reid GS, Waschek JA, Osborne LC, Sokol H, Vallance BA, Jacobson K. Vasoactive intestinal peptide promotes host defense against enteric pathogens by modulating the recruitment of group 3 innate lymphoid cells. Proc Natl Acad Sci U S A 2021; 118:e2106634118. [PMID: 34625492 PMCID: PMC8521691 DOI: 10.1073/pnas.2106634118] [Citation(s) in RCA: 40] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/30/2021] [Indexed: 01/10/2023] Open
Abstract
Group 3 innate lymphoid cells (ILC3s) control the formation of intestinal lymphoid tissues and play key roles in intestinal defense. They express neuropeptide vasoactive intestinal peptide (VIP) receptor 2 (VPAC2), through which VIP modulates their function, but whether VIP exerts other effects on ILC3 remains unclear. We show that VIP promotes ILC3 recruitment to the intestine through VPAC1 independent of the microbiota or adaptive immunity. VIP is also required for postnatal formation of lymphoid tissues as well as the maintenance of local populations of retinoic acid (RA)-producing dendritic cells, with RA up-regulating gut-homing receptor CCR9 expression by ILC3s. Correspondingly, mice deficient in VIP or VPAC1 suffer a paucity of intestinal ILC3s along with impaired production of the cytokine IL-22, rendering them highly susceptible to the enteric pathogen Citrobacter rodentium This heightened susceptibility to C. rodentium infection was ameliorated by RA supplementation, adoptive transfer of ILC3s, or by recombinant IL-22. Thus, VIP regulates the recruitment of intestinal ILC3s and formation of postnatal intestinal lymphoid tissues, offering protection against enteric pathogens.
Collapse
Affiliation(s)
- Hong Bing Yu
- Division of Gastroenterology, Hepatology and Nutrition, Department of Pediatrics, The University of British Columbia, Vancouver, BC, V5Z 4H4, Canada;
| | - Hyungjun Yang
- Division of Gastroenterology, Hepatology and Nutrition, Department of Pediatrics, The University of British Columbia, Vancouver, BC, V5Z 4H4, Canada
| | - Joannie M Allaire
- Division of Gastroenterology, Hepatology and Nutrition, Department of Pediatrics, The University of British Columbia, Vancouver, BC, V5Z 4H4, Canada
| | - Caixia Ma
- Division of Gastroenterology, Hepatology and Nutrition, Department of Pediatrics, The University of British Columbia, Vancouver, BC, V5Z 4H4, Canada
| | - Franziska A Graef
- Division of Gastroenterology, Hepatology and Nutrition, Department of Pediatrics, The University of British Columbia, Vancouver, BC, V5Z 4H4, Canada
| | - Arthur Mortha
- Department of Immunology, University of Toronto, Toronto, ON, M5S 1A8, Canada
| | - Qiaochu Liang
- Division of Gastroenterology, Hepatology and Nutrition, Department of Pediatrics, The University of British Columbia, Vancouver, BC, V5Z 4H4, Canada
| | - Else S Bosman
- Division of Gastroenterology, Hepatology and Nutrition, Department of Pediatrics, The University of British Columbia, Vancouver, BC, V5Z 4H4, Canada
| | - Gregor S Reid
- Division of Oncology, Department of Pediatrics, The University of British Columbia, Vancouver, BC, V5Z 4H4, Canada
| | - James A Waschek
- The Semel Institute and Department of Psychiatry, The David Geffen School of Medicine, University of California, Los Angeles, CA 90095
| | - Lisa C Osborne
- Department of Microbiology and Immunology, Life Sciences Institute, University of British Columbia, Vancouver, BC, V6T 1Z3, Canada
| | - Harry Sokol
- Gastroenterology Department, INSERM, Centre de Recherche Saint Antoine, Sorbonne Université, Paris, F-75012, France
- Institut national de la recherche agronomique, Micalis Institute and AgroParisTech, Jouy en Josas, F-78350, France
- Paris Center for Microbiome Medicine, Fédérations Hospitalo-universitaires, Paris, F-75012, France
| | - Bruce A Vallance
- Division of Gastroenterology, Hepatology and Nutrition, Department of Pediatrics, The University of British Columbia, Vancouver, BC, V5Z 4H4, Canada;
| | - Kevan Jacobson
- Division of Gastroenterology, Hepatology and Nutrition, Department of Pediatrics, The University of British Columbia, Vancouver, BC, V5Z 4H4, Canada;
| |
Collapse
|
44
|
Stehle C, Rückert T, Fiancette R, Gajdasik DW, Willis C, Ulbricht C, Durek P, Mashreghi MF, Finke D, Hauser AE, Withers DR, Chang HD, Zimmermann J, Romagnani C. T-bet and RORα control lymph node formation by regulating embryonic innate lymphoid cell differentiation. Nat Immunol 2021; 22:1231-1244. [PMID: 34556887 PMCID: PMC7614953 DOI: 10.1038/s41590-021-01029-6] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2020] [Accepted: 08/12/2021] [Indexed: 11/09/2022]
Abstract
The generation of lymphoid tissues during embryogenesis relies on group 3 innate lymphoid cells (ILC3) displaying lymphoid tissue inducer (LTi) activity and expressing the master transcription factor RORγt. Accordingly, RORγt-deficient mice lack ILC3 and lymphoid structures, including lymph nodes (LN). Whereas T-bet affects differentiation and functions of ILC3 postnatally, the role of T-bet in regulating fetal ILC3 and LN formation remains completely unknown. Using multiple mouse models and single-cell analyses of fetal ILCs and ILC progenitors (ILCP), here we identify a key role for T-bet during embryogenesis and show that its deficiency rescues LN formation in RORγt-deficient mice. Mechanistically, T-bet deletion skews the differentiation fate of fetal ILCs and promotes the accumulation of PLZFhi ILCP expressing central LTi molecules in a RORα-dependent fashion. Our data unveil an unexpected role for T-bet and RORα during embryonic ILC function and highlight that RORγt is crucial in counteracting the suppressive effects of T-bet.
Collapse
Affiliation(s)
- Christina Stehle
- Innate Immunity, German Rheumatism Research Centre-a Leibniz Institute, Berlin, Germany
| | - Timo Rückert
- Innate Immunity, German Rheumatism Research Centre-a Leibniz Institute, Berlin, Germany
| | - Rémi Fiancette
- Institute of Immunology and Immunotherapy, College of Medical and Dental Sciences, University of Birmingham, Birmingham, UK
| | - Dominika W Gajdasik
- Institute of Immunology and Immunotherapy, College of Medical and Dental Sciences, University of Birmingham, Birmingham, UK
| | - Claire Willis
- Institute of Immunology and Immunotherapy, College of Medical and Dental Sciences, University of Birmingham, Birmingham, UK
| | - Carolin Ulbricht
- Immune Dynamics, German Rheumatism Research Centre-a Leibniz Institute, Berlin, Germany
- Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Department of Rheumatology and Clinical Immunology, Berlin, Germany
| | - Pawel Durek
- Cell Biology, German Rheumatism Research Centre-a Leibniz Institute, Berlin, Germany
| | - Mir-Farzin Mashreghi
- Therapeutic Gene Regulation, German Rheumatism Research Centre-a Leibniz Institute, Berlin, Germany
- Berlin Institute of Health (BIH) at Charité-Universitätsmedizin Berlin, BIH Center for Regenerative Therapies (BCRT), Berlin, Germany
| | - Daniela Finke
- Department of Biomedicine and University Children's Hospital of Basel, University of Basel, Basel, Switzerland
| | - Anja Erika Hauser
- Immune Dynamics, German Rheumatism Research Centre-a Leibniz Institute, Berlin, Germany
- Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Department of Rheumatology and Clinical Immunology, Berlin, Germany
| | - David R Withers
- Institute of Immunology and Immunotherapy, College of Medical and Dental Sciences, University of Birmingham, Birmingham, UK
| | - Hyun-Dong Chang
- Schwiete Laboratory for Microbiota and Inflammation, German Rheumatism Research Centre-a Leibniz Institute, Berlin, Germany
- Department of Cytometry, Institute of Biotechnology, Technische Universität Berlin, Berlin, Germany
| | - Jakob Zimmermann
- Maurice Müller Laboratories, Universitätsklinik für Viszerale Chirurgie und Medizin Inselspital, University of Bern, Bern, Switzerland
| | - Chiara Romagnani
- Innate Immunity, German Rheumatism Research Centre-a Leibniz Institute, Berlin, Germany.
- Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Department of Gastroenterology, Infectious Diseases, Rheumatology, Berlin, Germany.
- Leibniz-Science Campus Chronic Inflammation, Berlin, Germany.
| |
Collapse
|
45
|
Goc J, Lv M, Bessman NJ, Flamar AL, Sahota S, Suzuki H, Teng F, Putzel GG, Eberl G, Withers DR, Arthur JC, Shah MA, Sonnenberg GF. Dysregulation of ILC3s unleashes progression and immunotherapy resistance in colon cancer. Cell 2021; 184:5015-5030.e16. [PMID: 34407392 PMCID: PMC8454863 DOI: 10.1016/j.cell.2021.07.029] [Citation(s) in RCA: 124] [Impact Index Per Article: 31.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2019] [Revised: 03/08/2021] [Accepted: 07/21/2021] [Indexed: 02/07/2023]
Abstract
Group 3 innate lymphoid cells (ILC3s) regulate immunity and inflammation, yet their role in cancer remains elusive. Here, we identify that colorectal cancer (CRC) manifests with altered ILC3s that are characterized by reduced frequencies, increased plasticity, and an imbalance with T cells. We evaluated the consequences of these changes in mice and determined that a dialog between ILC3s and T cells via major histocompatibility complex class II (MHCII) is necessary to support colonization with microbiota that subsequently induce type-1 immunity in the intestine and tumor microenvironment. As a result, mice lacking ILC3-specific MHCII develop invasive CRC and resistance to anti-PD-1 immunotherapy. Finally, humans with dysregulated intestinal ILC3s harbor microbiota that fail to induce type-1 immunity and immunotherapy responsiveness when transferred to mice. Collectively, these data define a protective role for ILC3s in cancer and indicate that their inherent disruption in CRC drives dysfunctional adaptive immunity, tumor progression, and immunotherapy resistance.
Collapse
Affiliation(s)
- Jeremy Goc
- 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 and Hepatology, Weill Cornell Medicine, Cornell University, New York, NY, USA; Department of Microbiology and Immunology, Weill Cornell Medicine, Cornell University, New York, NY, USA
| | - Mengze Lv
- 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 and Hepatology, Weill Cornell Medicine, Cornell University, New York, NY, USA; Department of Microbiology and Immunology, Weill Cornell Medicine, Cornell University, New York, NY, USA
| | - Nicholas J Bessman
- 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 and Hepatology, Weill Cornell Medicine, Cornell University, New York, NY, USA; Department of Microbiology and Immunology, Weill Cornell Medicine, Cornell University, New York, NY, USA
| | - Anne-Laure Flamar
- 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 and Hepatology, Weill Cornell Medicine, Cornell University, New York, NY, USA; Department of Microbiology and Immunology, Weill Cornell Medicine, Cornell University, New York, NY, USA
| | - Sheena Sahota
- 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 and Hepatology, Weill Cornell Medicine, Cornell University, New York, NY, USA; Department of Microbiology and Immunology, Weill Cornell Medicine, Cornell University, New York, NY, USA
| | - Hiroaki Suzuki
- 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 and Hepatology, Weill Cornell Medicine, Cornell University, New York, NY, USA; Department of Microbiology and Immunology, Weill Cornell Medicine, Cornell University, New York, NY, USA
| | - Fei Teng
- 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 and Hepatology, Weill Cornell Medicine, Cornell University, New York, NY, USA; Department of Microbiology and Immunology, Weill Cornell Medicine, Cornell University, New York, NY, USA
| | - Gregory G Putzel
- Jill Roberts Institute for Research in Inflammatory Bowel Disease, Weill Cornell Medicine, Cornell University, New York, NY, USA
| | - Gerard Eberl
- Microenvironment and Immunity Unit, Institut Pasteur, Paris, France
| | - David R Withers
- College of Medical and Dental Sciences, Institute of Immunology and Immunotherapy, University of Birmingham, Birmingham, UK
| | - Janelle C Arthur
- Center for Gastrointestinal Biology and Disease, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA; Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA; Department of Microbiology and Immunology, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Manish A Shah
- Division of Hematology and Medical Oncology, Department of Medicine, Weill Cornell Medicine, Cornell University, New York, NY, USA; Gastrointestinal Oncology Program, Center for Advanced Digestive Care, Sandra and Edward Meyer Cancer Center, New York-Presbyterian Hospital, 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 and Hepatology, Weill Cornell Medicine, Cornell University, New York, NY, USA; Department of Microbiology and Immunology, Weill Cornell Medicine, Cornell University, New York, NY, USA.
| |
Collapse
|
46
|
Lok LSC, Walker JA, Jolin HE, Scanlon ST, Ishii M, Fallon PG, McKenzie ANJ, Clatworthy MR. Group 2 Innate Lymphoid Cells Exhibit Tissue-Specific Dynamic Behaviour During Type 2 Immune Responses. Front Immunol 2021; 12:711907. [PMID: 34484215 PMCID: PMC8415880 DOI: 10.3389/fimmu.2021.711907] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2021] [Accepted: 07/29/2021] [Indexed: 12/25/2022] Open
Abstract
Group 2 innate lymphoid cells (ILC2s) are early effectors of mucosal type 2 immunity, producing cytokines such as interleukin (IL)-13 to mediate responses to helminth infection and allergen-induced inflammation. ILC2s are also present in lymph nodes (LNs) and can express molecules required for antigen presentation, but to date there are limited data on their dynamic behaviour. We used a CD2/IL-13 dual fluorescent reporter mouse for in vivo imaging of ILC2s and Th2 T cells in real time following a type 2 priming helminth infection or egg injection. After helminth challenge, we found that ILC2s were the main source of IL-13 in lymphoid organs (Peyer’s patches and peripheral LNs), and were located in T cell areas. Intravital imaging demonstrated an increase in IL-13+ ILC2 size and movement following helminth infection, but reduced duration of interactions with T cells compared with those in homeostasis. In contrast, in the intestinal mucosa, we observed an increase in ILC2-T cell interactions post-infection, including some of prolonged duration, as well as increased IL-13+ ILC2 movement. These data suggest that ILC2 activation enhances cell motility, with the potential to increase the area of distribution of cytokines to optimise the early generation of type 2 responses. The prolonged ILC2 interactions with T cells within the intestinal mucosa are consistent with the conclusion that contact-based T cell activation may occur within inflamed tissues rather than lymphoid organs. Our findings have important implications for our understanding of the in vivo biology of ILC2s and the way in which these cells facilitate adaptive immune responses.
Collapse
Affiliation(s)
- Laurence S C Lok
- Molecular Immunity Unit, Department of Medicine, MRC Laboratory of Molecular Biology, University of Cambridge, Cambridge, United Kingdom.,Cambridge Institute for Therapeutic Immunology and Infectious Diseases, University of Cambridge, Cambridge, United Kingdom.,Department of Immunology and Cell Biology, Graduate School of Medicine, Osaka University, Osaka, Japan.,Immunology Frontier Research Center, Osaka University, Osaka, Japan
| | - Jennifer A Walker
- Medical Research Council Laboratory of Molecular Biology, Cambridge, United Kingdom
| | - Helen E Jolin
- Medical Research Council Laboratory of Molecular Biology, Cambridge, United Kingdom
| | - Seth T Scanlon
- Medical Research Council Laboratory of Molecular Biology, Cambridge, United Kingdom
| | - Masaru Ishii
- Department of Immunology and Cell Biology, Graduate School of Medicine, Osaka University, Osaka, Japan.,Immunology Frontier Research Center, Osaka University, Osaka, Japan
| | | | - Andrew N J McKenzie
- Medical Research Council Laboratory of Molecular Biology, Cambridge, United Kingdom
| | - Menna R Clatworthy
- Molecular Immunity Unit, Department of Medicine, MRC Laboratory of Molecular Biology, University of Cambridge, Cambridge, United Kingdom.,Cambridge Institute for Therapeutic Immunology and Infectious Diseases, University of Cambridge, Cambridge, United Kingdom.,Cellular Genetics, Wellcome Sanger Institute, Hinxton, United Kingdom
| |
Collapse
|
47
|
Huang C, Li F, Wang J, Tian Z. Innate-like Lymphocytes and Innate Lymphoid Cells in Asthma. Clin Rev Allergy Immunol 2021; 59:359-370. [PMID: 31776937 DOI: 10.1007/s12016-019-08773-6] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023]
Abstract
Asthma is a chronic pulmonary disease, highly associated with immune disorders. The typical symptoms of asthma include airway hyperresponsiveness (AHR), airway remodeling, mucus overproduction, and airflow limitation. The etiology of asthma is multifactorial and affected by genetic and environmental factors. Increasing trends toward dysbiosis, smoking, stress, air pollution, and a western lifestyle may account for the increasing incidence of asthma. Based on the presence or absence of eosinophilic inflammation, asthma is mainly divided into T helper 2 (Th2) and non-Th2 asthma. Th2 asthma is mediated by allergen-specific Th2 cells, and eosinophils activated by Th2 cells via the secretion of interleukin (IL)-4, IL-5, and IL-13. Different from Th2 asthma, non-Th2 asthma shows little eosinophilic inflammation, resists to corticosteroid treatment, and occurs mainly in severe asthmatic patients. Previous studies of asthma primarily focused on the function of Th2 cells, but, with the discovery of non-Th2 asthma and the involvement of innate lymphoid cells (ILCs) in the pathogenesis of asthma, tissue-resident innate immune cells in the lung have become the focus of attention in asthma research. Currently, innate-like lymphocytes (ILLs) and ILCs as important components of the innate immune system in mucosal tissues are reportedly involved in the pathogenesis of or protection against both Th2 and non-Th2 asthma. These findings of the functions of different subsets of ILLs and ILCs may provide clues for the treatment of asthma.
Collapse
Affiliation(s)
- Chao Huang
- Institute of Immunology, University of Science and Technology of China, Hefei, 230027, Anhui, China
| | - Fengqi Li
- Institute of Molecular Health Sciences, ETH Zürich, 8093, Zürich, Switzerland
| | - Jian Wang
- Neuroimmunology and MS Research Section (NIMS), Neurology Clinic, University of Zürich, University Hospital Zürich, 8091, Zürich, Switzerland.
| | - Zhigang Tian
- Institute of Immunology, University of Science and Technology of China, Hefei, 230027, Anhui, China.
| |
Collapse
|
48
|
Saez A, Gomez-Bris R, Herrero-Fernandez B, Mingorance C, Rius C, Gonzalez-Granado JM. Innate Lymphoid Cells in Intestinal Homeostasis and Inflammatory Bowel Disease. Int J Mol Sci 2021; 22:ijms22147618. [PMID: 34299236 PMCID: PMC8307624 DOI: 10.3390/ijms22147618] [Citation(s) in RCA: 107] [Impact Index Per Article: 26.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2021] [Revised: 07/13/2021] [Accepted: 07/14/2021] [Indexed: 02/07/2023] Open
Abstract
Inflammatory bowel disease (IBD) is a heterogeneous state of chronic intestinal inflammation of unknown cause encompassing Crohn’s disease (CD) and ulcerative colitis (UC). IBD has been linked to genetic and environmental factors, microbiota dysbiosis, exacerbated innate and adaptive immunity and epithelial intestinal barrier dysfunction. IBD is classically associated with gut accumulation of proinflammatory Th1 and Th17 cells accompanied by insufficient Treg numbers and Tr1 immune suppression. Inflammatory T cells guide innate cells to perpetuate a constant hypersensitivity to microbial antigens, tissue injury and chronic intestinal inflammation. Recent studies of intestinal mucosal homeostasis and IBD suggest involvement of innate lymphoid cells (ILCs). These lymphoid-origin cells are innate counterparts of T cells but lack the antigen receptors expressed on B and T cells. ILCs play important roles in the first line of antimicrobial defense and contribute to organ development, tissue protection and regeneration, and mucosal homeostasis by maintaining the balance between antipathogen immunity and commensal tolerance. Intestinal homeostasis requires strict regulation of the quantity and activity of local ILC subpopulations. Recent studies demonstrated that changes to ILCs during IBD contribute to disease development. A better understanding of ILC behavior in gastrointestinal homeostasis and inflammation will provide valuable insights into new approaches to IBD treatment. This review summarizes recent research into ILCs in intestinal homeostasis and the latest advances in the understanding of the role of ILCs in IBD, with particular emphasis on the interaction between microbiota and ILC populations and functions.
Collapse
Affiliation(s)
- Angela Saez
- LamImSys Lab, Instituto de Investigación Hospital 12 de Octubre (imas12), 28041 Madrid, Spain; (A.S.); (R.G.-B.); (B.H.-F.); (C.M.)
- Facultad de Ciencias Experimentales, Universidad Francisco de Vitoria (UFV), 28223 Madrid, Spain
| | - Raquel Gomez-Bris
- LamImSys Lab, Instituto de Investigación Hospital 12 de Octubre (imas12), 28041 Madrid, Spain; (A.S.); (R.G.-B.); (B.H.-F.); (C.M.)
- Departamento de Fisiología, Facultad de Medicina, Universidad Autónoma de Madrid (UAM), 28029 Madrid, Spain
| | - Beatriz Herrero-Fernandez
- LamImSys Lab, Instituto de Investigación Hospital 12 de Octubre (imas12), 28041 Madrid, Spain; (A.S.); (R.G.-B.); (B.H.-F.); (C.M.)
- Departamento de Fisiología, Facultad de Medicina, Universidad Autónoma de Madrid (UAM), 28029 Madrid, Spain
| | - Claudia Mingorance
- LamImSys Lab, Instituto de Investigación Hospital 12 de Octubre (imas12), 28041 Madrid, Spain; (A.S.); (R.G.-B.); (B.H.-F.); (C.M.)
| | - Cristina Rius
- Faculty of Biomedical and Health Sciences, Universidad Europea de Madrid (UEM), Villaviciosa de Odón, 28670 Madrid, Spain;
- Centro Nacional de Investigaciones Cardiovasculares (CNIC), 28029 Madrid, Spain
- CIBER de Enfermedades Cardiovasculares, 28029 Madrid, Spain
| | - Jose M. Gonzalez-Granado
- LamImSys Lab, Instituto de Investigación Hospital 12 de Octubre (imas12), 28041 Madrid, Spain; (A.S.); (R.G.-B.); (B.H.-F.); (C.M.)
- Departamento de Fisiología, Facultad de Medicina, Universidad Autónoma de Madrid (UAM), 28029 Madrid, Spain
- Centro Nacional de Investigaciones Cardiovasculares (CNIC), 28029 Madrid, Spain
- CIBER de Enfermedades Cardiovasculares, 28029 Madrid, Spain
- Correspondence: ; Tel.: +34-913908766
| |
Collapse
|
49
|
Sécca C, Bando JK, Fachi JL, Gilfillan S, Peng V, Di Luccia B, Cella M, McDonald KG, Newberry RD, Colonna M. Spatial distribution of LTi-like cells in intestinal mucosa regulates type 3 innate immunity. Proc Natl Acad Sci U S A 2021; 118:e2101668118. [PMID: 34083442 PMCID: PMC8201890 DOI: 10.1073/pnas.2101668118] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
Abstract
Lymphoid tissue inducer (LTi)-like cells are tissue resident innate lymphocytes that rapidly secrete cytokines that promote gut epithelial integrity and protect against extracellular bacterial infections.Here, we report that the retention of LTi-like cells in conventional solitary intestinal lymphoid tissue (SILT) is essential for controlling LTi-like cell function and is maintained by expression of the chemokine receptor CXCR5. Deletion of Cxcr5 functionally unleashed LTi-like cells in a cell intrinsic manner, leading to uncontrolled IL-17 and IL-22 production. The elevated production of IL-22 in Cxcr5-deficient mice improved gut barrier integrity and protected mice during infection with the opportunistic pathogen Clostridium difficile Interestingly, Cxcr5-/- mice developed LTi-like cell aggregates that were displaced from their typical niche at the intestinal crypt, and LTi-like cell hyperresponsiveness was associated with the local formation of this unconventional SILT. Thus, LTi-like cell positioning within mucosa controls their activity via niche-specific signals that temper cytokine production during homeostasis.
Collapse
Affiliation(s)
- Cristiane Sécca
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO 63110
| | - Jennifer K Bando
- Department of Microbiology and Immunology, Stanford University School of Medicine, Stanford, CA 94305
| | - José L Fachi
- Laboratory of Immunoinflammation, Institute of Biology, University of Campinas, Campinas 13083-862, Brazil
- Department of Genetics, Evolution, Microbiology, and Immunology, Institute of Biology, University of Campinas, Campinas 13083-862, Brazil
| | - Susan Gilfillan
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO 63110
| | - Vincent Peng
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO 63110
| | - Blanda Di Luccia
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO 63110
| | - Marina Cella
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO 63110
| | - Keely G McDonald
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO 63110
| | - Rodney D Newberry
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO 63110
| | - Marco Colonna
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO 63110;
| |
Collapse
|
50
|
Dynamic regulation of innate lymphoid cells in the mucosal immune system. Cell Mol Immunol 2021; 18:1387-1394. [PMID: 33980994 PMCID: PMC8167116 DOI: 10.1038/s41423-021-00689-6] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2021] [Accepted: 04/18/2021] [Indexed: 02/03/2023] Open
Abstract
The mucosal immune system is considered a local immune system, a term that implies regional restriction. Mucosal tissues are continually exposed to a wide range of antigens. The regulation of mucosal immune cells is tightly associated with the progression of mucosal diseases. Innate lymphoid cells (ILCs) are abundant in mucosal barriers and serve as first-line defenses against pathogens. The subtype changes and translocation of ILCs are accompanied by the pathologic processes of mucosal diseases. Here, we review the plasticity and circulation of ILCs in the mucosal immune system under physiological and pathological conditions. We also discuss the signaling pathways involved in dynamic ILC changes and the related targets in mucosal diseases.
Collapse
|