1
|
Kabil A, Nayyar N, Xu C, Brassard J, Hill LA, Shin SB, Chopra S, Lo B, Li Y, Bal M, Theret M, Rossi FM, Underhill TM, Hughes MR, McNagny KM. Functional targeting of ILC2s and ILC3s reveals selective roles in intestinal fibrosis and homeostasis. J Exp Med 2025; 222:e20241671. [PMID: 40434419 PMCID: PMC12118370 DOI: 10.1084/jem.20241671] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2024] [Revised: 03/06/2025] [Accepted: 04/28/2025] [Indexed: 05/29/2025] Open
Abstract
Innate lymphoid cells (ILCs) are long-lived, tissue-resident cell analogs to T helper subsets that lack antigen-specific receptors. Understanding the roles of specific ILCs in chronic inflammation and fibrosis has been limited by inadequate tools for selective targeting. Here, we used Il17rb-CreERT2-eGFP and Rorc-Cre strains to selectively delete RORα in ILC2s and ILC3/Th17 cells, respectively. RORα deletion in ILC2s caused significant loss of gastrointestinal ILC2s, increased ILC3 abundance, elevated Th17-type responses, and heightened susceptibility to Crohn's disease-like fibrosis. Conversely, RORα deletion in ILC3/Th17 cells reduced IL-17 production, protecting against fibrosis. Using isolithocholic acid (isoLCA), a microbial secondary bile acid and RORγt inverse agonist, we confirmed the role of ILC3s/Th17 cells in fibrosis. In RORγt reporter and Th17-deficient Rag1-/- mice, isoLCA reduced IL-17 production by ILC3s and attenuated intestinal fibrosis by dampening RORγt-dependent ILC3/Th17 responses. These findings reveal a novel interplay between ILC2s and ILC3s in gut homeostasis and demonstrate the therapeutic potential of targeting RORγt in ILC3s as a strategy for preventing fibrosis.
Collapse
MESH Headings
- Animals
- Homeostasis/immunology
- Fibrosis
- Mice
- Immunity, Innate
- Lymphocytes/immunology
- Th17 Cells/immunology
- Th17 Cells/metabolism
- Intestines/pathology
- Intestines/immunology
- Nuclear Receptor Subfamily 1, Group F, Member 3/metabolism
- Nuclear Receptor Subfamily 1, Group F, Member 3/genetics
- Nuclear Receptor Subfamily 1, Group F, Member 1/metabolism
- Nuclear Receptor Subfamily 1, Group F, Member 1/genetics
- Mice, Knockout
- Interleukin-17/metabolism
- Mice, Inbred C57BL
- Crohn Disease/immunology
- Crohn Disease/pathology
Collapse
Affiliation(s)
- Ahmed Kabil
- School of Biomedical Engineering, University of British Columbia, Vancouver, Canada
| | - Natalia Nayyar
- School of Biomedical Engineering, University of British Columbia, Vancouver, Canada
| | - Chengxi Xu
- School of Biomedical Engineering, University of British Columbia, Vancouver, Canada
| | - Julyanne Brassard
- School of Biomedical Engineering, University of British Columbia, Vancouver, Canada
| | - Lesley A. Hill
- Department of Cellular and Physiological Sciences, University of British Columbia, Vancouver, Canada
| | - Samuel B. Shin
- School of Biomedical Engineering, University of British Columbia, Vancouver, Canada
| | - Sameeksha Chopra
- School of Biomedical Engineering, University of British Columbia, Vancouver, Canada
| | - Bernard Lo
- Department of Pathology and Rogel Cancer Center, University of Michigan, Ann Arbor, MI, USA
| | - Yicong Li
- School of Biomedical Engineering, University of British Columbia, Vancouver, Canada
| | - Mya Bal
- School of Biomedical Engineering, University of British Columbia, Vancouver, Canada
| | - Marine Theret
- School of Biomedical Engineering, University of British Columbia, Vancouver, Canada
| | - Fabio M.V. Rossi
- School of Biomedical Engineering, University of British Columbia, Vancouver, Canada
- The Department of Medical Genetics, University of British Columbia, Vancouver, Canada
| | - T. Michael Underhill
- School of Biomedical Engineering, University of British Columbia, Vancouver, Canada
- Department of Cellular and Physiological Sciences, University of British Columbia, Vancouver, Canada
| | - Michael R. Hughes
- School of Biomedical Engineering, University of British Columbia, Vancouver, Canada
| | - Kelly M. McNagny
- School of Biomedical Engineering, University of British Columbia, Vancouver, Canada
- The Department of Medical Genetics, University of British Columbia, Vancouver, Canada
- The Center for Heart Lung Innovation, St Paul’s Hospital, Vancouver, Canada
| |
Collapse
|
2
|
Churchill MJ, Pandeya A, Bauer R, Christopher T, Krug S, Honodel R, Smita S, Warner L, Mooney BM, Gibson AR, Mitchell PS, Tait Wojno ED, Rauch I. Enteric tuft cell inflammasome activation drives NKp46+ILC3 IL22 via PGD2 and inhibits Salmonella. J Exp Med 2025; 222:e20230803. [PMID: 40079814 PMCID: PMC11905811 DOI: 10.1084/jem.20230803] [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: 05/11/2023] [Revised: 12/12/2024] [Accepted: 02/10/2025] [Indexed: 03/15/2025] Open
Abstract
To distinguish pathogens from commensals, the intestinal epithelium employs cytosolic innate immune sensors. Activation of the NAIP-NLRC4 inflammasome initiates extrusion of infected intestinal epithelial cells (IEC) upon cytosolic bacterial sensing. We previously reported that activation of the inflammasome in tuft cells, which are primarily known for their role in parasitic infections, leads to the release of prostaglandin D2 (PGD2). We observe that NAIP-NLRC4 inflammasome activation in tuft cells leads to an antibacterial response with increased IL-22 and antimicrobial protein levels within the small intestine, which is dependent on PGD2 signaling. A NKp46+ subset of ILC3 expresses the PGD2 receptor CRTH2 and is the source of the increased IL-22. Inflammasome activation in tuft cells also leads to better control of Salmonella Typhimurium in the distal small intestine. However, tuft cells in the cecum and colon are dispensable for antibacterial immunity. These data support that intestinal tuft cells can also induce antibacterial responses, possibly in a tissue-specific manner.
Collapse
Affiliation(s)
- Madeline J. Churchill
- Department of Molecular Microbiology and Immunology, Oregon Health and Science University, Portland, OR, USA
| | - Ankit Pandeya
- Department of Molecular Microbiology and Immunology, Oregon Health and Science University, Portland, OR, USA
| | - Renate Bauer
- Department of Biosciences and Medical Biology, Paris Lodron University of Salzburg, Salzburg, Austria
| | - Tighe Christopher
- Department of Immunology, University of Washington, Seattle, WA, USA
| | - Stefanie Krug
- Department of Microbiology, University of Washington, Seattle, WA, USA
| | - Roslyn Honodel
- Department of Molecular Microbiology and Immunology, Oregon Health and Science University, Portland, OR, USA
| | - Shuchi Smita
- Department of Immunology, University of Washington, Seattle, WA, USA
| | - Lindsey Warner
- Department of Immunology, University of Washington, Seattle, WA, USA
| | - Bridget M. Mooney
- Department of Immunology, University of Washington, Seattle, WA, USA
| | - Alexis R. Gibson
- Department of Molecular Microbiology and Immunology, Oregon Health and Science University, Portland, OR, USA
| | - Patrick S. Mitchell
- Department of Microbiology, University of Washington, Seattle, WA, USA
- Howard Hughes Medical Institute, University of Washington, Seattle, WA, USA
| | | | - Isabella Rauch
- Department of Molecular Microbiology and Immunology, Oregon Health and Science University, Portland, OR, USA
| |
Collapse
|
3
|
Gao X, Yang C, Feng Z, Liu P, Liu Z. The signature of the small intestinal epithelial and immune cells in health and diseases. Chin Med J (Engl) 2025:00029330-990000000-01558. [PMID: 40394804 DOI: 10.1097/cm9.0000000000003615] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2024] [Indexed: 05/22/2025] Open
Abstract
ABSTRACT The small intestine is essential for digestion, nutrient absorption, immune regulation, and microbial balance. Its epithelial lining, containing specialized cells like Paneth and tuft cells, is crucial for maintaining intestinal homeostasis. Paneth cells produce antimicrobial peptides and growth factors that support microbial regulation and intestinal stem cells, while tuft cells act as chemosensors, detecting environmental changes and modulating immune responses. Along with immune cells such as intraepithelial lymphocytes, innate lymphoid cells, T cells, and macrophages, they form a strong defense system that protects the epithelial barrier. Disruptions in this balance contribute to chronic inflammation, microbial dysbiosis, and compromised barrier function-key features of inflammatory bowel disease, celiac disease, and metabolic syndromes. Furthermore, dysfunctions in the small intestine and immune cells are linked to systemic diseases like obesity, diabetes, and autoimmune disorders. Recent research highlights promising therapeutic strategies, including modulation of epithelial and immune cell functions, probiotics, and gene editing to restore gut health and address systemic effects. This review emphasizes the pivotal roles of small intestinal epithelia and immune cells in maintaining intestinal homeostasis, their involvement in disease development, and emerging treatments for intestinal and systemic disorders.
Collapse
Affiliation(s)
- Xiang Gao
- Center for Inflammatory Bowel Disease Research, Department of Gastroenterology, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai 200072, China
| | - Cuiping Yang
- Department of Gastroenterology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 201801, China
| | - Zhongsheng Feng
- Center for Inflammatory Bowel Disease Research, Department of Gastroenterology, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai 200072, China
| | - Ping Liu
- Department of Gastroenterology, Wuhu First People's Hospital, Wuhu, Anhui 241000, China
| | - Zhanju Liu
- Center for Inflammatory Bowel Disease Research, Department of Gastroenterology, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai 200072, China
| |
Collapse
|
4
|
Xu H, Wang Y, Wang W, Fu YX, Qiu J, Shi Y, Yuan L, Dong C, Hu X, Chen YG, Guo X. ILC3s promote intestinal tuft cell hyperplasia and anthelmintic immunity through RANK signaling. Sci Immunol 2025; 10:eadn1491. [PMID: 40378237 DOI: 10.1126/sciimmunol.adn1491] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2023] [Revised: 08/01/2024] [Accepted: 04/24/2025] [Indexed: 05/18/2025]
Abstract
Helminth infections, particularly in developing countries, remain a notable health burden worldwide. Group 3 innate lymphoid cells (ILC3s) are enriched in the intestine and play a critical role in immunity against extracellular bacteria and fungi. However, whether ILC3s are involved in intestinal helminth infection is still unclear. Here, we report that helminth infection reprograms ILC3s, which, in turn, promote anthelmintic immunity. ILC3-derived RANKL [receptor activator of NF-κB (nuclear factor κB) ligand] synergizes with interleukin-13 (IL-13) to facilitate intestinal tuft cell expansion after helminth infection, which further activates the tuft cell-group 2 innate lymphoid cell (ILC2) circuit to control helminth infection. Deletion of RANKL in ILC3s or deletion of RANK or its downstream adaptor RelB in intestinal epithelial cells substantially diminishes tuft cell hyperplasia and dampens anthelmintic immunity. Thus, ILC3s play an indispensable role in protecting against helminth infection through the regulation of intestinal tuft cell hyperplasia and type 2 immunity.
Collapse
Affiliation(s)
- Hongkai Xu
- Institute for Immunology, Tsinghua University, Beijing 100084, China
- School of Basic Medical Sciences, Tsinghua Medicine, Tsinghua University, Beijing 100084, China
- Beijing Key Laboratory of Immunological Research of Allergy (LIRA), Tsinghua University, Beijing 100084, China
| | - Yibo Wang
- Institute for Immunology, Tsinghua University, Beijing 100084, China
- School of Basic Medical Sciences, Tsinghua Medicine, Tsinghua University, Beijing 100084, China
- Beijing Key Laboratory of Immunological Research of Allergy (LIRA), Tsinghua University, Beijing 100084, China
- State Key Laboratory of Molecular Oncology, School of Basic Medical Sciences, Tsinghua University, Beijing 100084, China
| | - Wenyan Wang
- School of Basic Medical Sciences, Tsinghua Medicine, Tsinghua University, Beijing 100084, China
- State Key Laboratory of Molecular Oncology, School of Basic Medical Sciences, Tsinghua University, Beijing 100084, China
| | - Yang-Xin Fu
- School of Basic Medical Sciences, Tsinghua Medicine, Tsinghua University, Beijing 100084, China
- State Key Laboratory of Molecular Oncology, School of Basic Medical Sciences, Tsinghua University, Beijing 100084, China
| | - Ju Qiu
- CAS Key Laboratory of Tissue Microenvironment and Tumor, Shanghai Institute of Nutrition and Health, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai 200031, China
| | - Yan Shi
- Institute for Immunology, Tsinghua University, Beijing 100084, China
- School of Basic Medical Sciences, Tsinghua Medicine, Tsinghua University, Beijing 100084, China
- Beijing Key Laboratory of Immunological Research of Allergy (LIRA), Tsinghua University, Beijing 100084, China
| | - Lei Yuan
- Institute for Immunology, Tsinghua University, Beijing 100084, China
| | - Chen Dong
- Westlake University, Hangzhou 310030, China
| | - Xiaoyu Hu
- Institute for Immunology, Tsinghua University, Beijing 100084, China
- School of Basic Medical Sciences, Tsinghua Medicine, Tsinghua University, Beijing 100084, China
- Beijing Key Laboratory of Immunological Research of Allergy (LIRA), Tsinghua University, Beijing 100084, China
| | - Ye-Guang Chen
- State Key Laboratory of Membrane Biology, Tsinghua-Peking Center for Life Sciences, School of Life Sciences, Tsinghua University, Beijing, 100084, China
| | - Xiaohuan Guo
- Institute for Immunology, Tsinghua University, Beijing 100084, China
- School of Basic Medical Sciences, Tsinghua Medicine, Tsinghua University, Beijing 100084, China
- Beijing Key Laboratory of Immunological Research of Allergy (LIRA), Tsinghua University, Beijing 100084, China
- State Key Laboratory of Molecular Oncology, School of Basic Medical Sciences, Tsinghua University, Beijing 100084, China
- SXMU-Tsinghua Collaborative Innovation Center for Frontier Medicine, Shanxi Medical University, Taiyuan, Shanxi 030001, China
| |
Collapse
|
5
|
Rodrigues PF, Wu S, Trsan T, Panda SK, Fachi JL, Liu Y, Du S, de Oliveira S, Antonova AU, Khantakova D, Sudan R, Desai P, Diamond MS, Gilfillan S, Anderson SK, Cella M, Colonna M. Rorγt-positive dendritic cells are required for the induction of peripheral regulatory T cells in response to oral antigens. Cell 2025; 188:2720-2737.e22. [PMID: 40185101 DOI: 10.1016/j.cell.2025.03.020] [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: 08/26/2024] [Revised: 01/26/2025] [Accepted: 03/10/2025] [Indexed: 04/07/2025]
Abstract
The intestinal immune system maintains tolerance to harmless food proteins and gut microbiota through peripherally derived RORγt+ Tregs (pTregs), which prevent food intolerance and inflammatory bowel disease. Recent studies suggested that RORγt+ antigen-presenting cells (APCs), which encompass rare dendritic cell (DC) subsets and type 3 innate lymphoid cells (ILC3s), are key to pTreg induction. Here, we developed a mouse with reduced RORγt+ APCs by deleting a specific cis-regulatory element of Rorc encoding RORγt. Single-cell RNA sequencing and flow cytometry analyses confirmed the depletion of a RORγt+ DC subset and ILC3s. These mice showed a secondary reduction in pTregs, impaired tolerance to oral antigens, and an increase in T helper (Th)2 cells. Conversely, ILC3-deficient mice showed no pTregs or Th2 cell abnormalities. Lineage tracing revealed that RORγt+ DCs share a lymphoid origin with ILC3s, consistent with their similar phenotypic traits. These findings highlight the role of lymphoid RORγt+ DCs in maintaining intestinal immune balance and preventing conditions like food allergies.
Collapse
Affiliation(s)
- Patrick Fernandes Rodrigues
- Department of Pathology and Immunology, Washington University School of Medicine in Saint Louis, Saint Louis, MO, USA
| | - Shitong Wu
- Department of Pathology and Immunology, Washington University School of Medicine in Saint Louis, Saint Louis, MO, USA
| | - Tihana Trsan
- Department of Pathology and Immunology, Washington University School of Medicine in Saint Louis, Saint Louis, MO, USA
| | - Santosh K Panda
- Department of Pathology and Immunology, Washington University School of Medicine in Saint Louis, Saint Louis, MO, USA
| | - José Luís Fachi
- Department of Pathology and Immunology, Washington University School of Medicine in Saint Louis, Saint Louis, MO, USA
| | - Yizhou Liu
- Department of Pathology and Immunology, Washington University School of Medicine in Saint Louis, Saint Louis, MO, USA
| | - Siling Du
- Department of Pathology and Immunology, Washington University School of Medicine in Saint Louis, Saint Louis, MO, USA
| | - Sarah de Oliveira
- Department of Genetics and Evolution, Microbiology and Immunology, Institute of Biology, University of Campinas, Campinas, Brazil
| | - Alina Ulezko Antonova
- Department of Pathology and Immunology, Washington University School of Medicine in Saint Louis, Saint Louis, MO, USA
| | - Darya Khantakova
- Department of Pathology and Immunology, Washington University School of Medicine in Saint Louis, Saint Louis, MO, USA
| | - Raki Sudan
- Department of Pathology and Immunology, Washington University School of Medicine in Saint Louis, Saint Louis, MO, USA
| | - Pritesh Desai
- Department of Medicine, Washington University School of Medicine in Saint Louis, Saint Louis, MO, USA
| | - Michael S Diamond
- Department of Pathology and Immunology, Washington University School of Medicine in Saint Louis, Saint Louis, MO, USA; Department of Medicine, Washington University School of Medicine in Saint Louis, Saint Louis, MO, USA
| | - Susan Gilfillan
- Department of Pathology and Immunology, Washington University School of Medicine in Saint Louis, Saint Louis, MO, USA
| | - Stephen K Anderson
- Basic Science Program, Frederick National Laboratory for Cancer Research, National Cancer Institute, Frederick, MD 21702, USA
| | - Marina Cella
- Department of Pathology and Immunology, Washington University School of Medicine in Saint Louis, Saint Louis, MO, USA
| | - Marco Colonna
- Department of Pathology and Immunology, Washington University School of Medicine in Saint Louis, Saint Louis, MO, USA.
| |
Collapse
|
6
|
Kao YS, Lauterbach M, Lopez Krol A, Distler U, Godoy GJ, Klein M, Argüello RJ, Boukhallouk F, Vallejo Fuente S, Braband KL, Nurbekova A, Romero M, Mamareli P, Silva L, Damasceno LEA, Rampoldi F, Berod L, Lynch L, Hiller K, Sparwasser T. Metabolic reprogramming of interleukin-17-producing γδ T cells promotes ACC1-mediated de novo lipogenesis under psoriatic conditions. Nat Metab 2025; 7:966-984. [PMID: 40360755 PMCID: PMC12116387 DOI: 10.1038/s42255-025-01276-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/11/2024] [Accepted: 03/13/2025] [Indexed: 05/15/2025]
Abstract
Metabolic reprogramming determines γδ T cell fate during thymic development; however, the metabolic requirements of interleukin (IL)-17A-producing γδ T cells (γδT17 cells) under psoriatic conditions are unclear. Combining high-throughput techniques, including RNA sequencing, SCENITH, proteomics and stable isotope tracing, we demonstrated that psoriatic inflammation caused γδT17 cells to switch toward aerobic glycolysis. Under psoriatic conditions, γδT17 cells upregulated ATP-citrate synthase to convert citrate to acetyl-CoA, linking carbohydrate metabolism and fatty acid synthesis (FAS). Accordingly, we used a pharmacological inhibitor, Soraphen A, which blocks acetyl-CoA carboxylase (ACC), to impair FAS in γδT17 cells, reducing their intracellular lipid stores and ability to produce IL-17A under psoriatic conditions in vitro. We pinpointed the pathogenic role of ACC1 in γδT17 cells in vivo by genetic ablation, ameliorating inflammation in a psoriatic mouse model. Furthermore, ACC inhibition limited human IL-17A-producing γδT17 cells. Targeting ACC1 to attenuate pathogenic γδT17 cell function has important implications for psoriasis management.
Collapse
Affiliation(s)
- Yu-San Kao
- Institute of Medical Microbiology and Hygiene, University Medical Center of the Johannes Gutenberg University Mainz, Mainz, Germany.
- Ludwig Cancer Research Institute, Princeton Branch, Princeton University, Princeton, NJ, USA.
| | - Mario Lauterbach
- Department of Bioinformatics and Biochemistry, Braunschweig Integrated Centre of Systems Biology, Technical University of Braunschweig, Braunschweig, Germany
| | - Aleksandra Lopez Krol
- Institute of Medical Microbiology and Hygiene, University Medical Center of the Johannes Gutenberg University Mainz, Mainz, Germany
| | - Ute Distler
- Research Center for Immunotherapy (FZI), University Medical Center of the Johannes Gutenberg University Mainz, Mainz, Germany
- Institute for Immunology, University Medical Center of the Johannes Gutenberg-University Mainz, Mainz, Germany
| | - Gloria Janet Godoy
- Institute of Medical Microbiology and Hygiene, University Medical Center of the Johannes Gutenberg University Mainz, Mainz, Germany
- Institute of Molecular Medicine, University Medical Center of the Johannes Gutenberg University Mainz, Mainz, Germany
| | - Matthias Klein
- Research Center for Immunotherapy (FZI), University Medical Center of the Johannes Gutenberg University Mainz, Mainz, Germany
- Institute for Immunology, University Medical Center of the Johannes Gutenberg-University Mainz, Mainz, Germany
| | - Rafael Jose Argüello
- Aix Marseille University, CNRS, INSERM, CIML, Centre d'Immunologie de Marseille-Luminy, Marseille, France
| | - Fatima Boukhallouk
- Institute of Medical Microbiology and Hygiene, University Medical Center of the Johannes Gutenberg University Mainz, Mainz, Germany
| | - Sara Vallejo Fuente
- Institute of Medical Microbiology and Hygiene, University Medical Center of the Johannes Gutenberg University Mainz, Mainz, Germany
| | - Kathrin Luise Braband
- Institute of Medical Microbiology and Hygiene, University Medical Center of the Johannes Gutenberg University Mainz, Mainz, Germany
- Institute for Immunology, University Medical Center of the Johannes Gutenberg University Mainz, Mainz, Germany
| | - Assel Nurbekova
- Institute for Immunology, University Medical Center of the Johannes Gutenberg-University Mainz, Mainz, Germany
| | - Monica Romero
- Institute of Medical Microbiology and Hygiene, University Medical Center of the Johannes Gutenberg University Mainz, Mainz, Germany
| | - Panagiota Mamareli
- Institute of Medical Microbiology and Hygiene, University Medical Center of the Johannes Gutenberg University Mainz, Mainz, Germany
| | - Luana Silva
- Institute of Medical Microbiology and Hygiene, University Medical Center of the Johannes Gutenberg University Mainz, Mainz, Germany
| | - Luis Eduardo Alves Damasceno
- Institute of Medical Microbiology and Hygiene, University Medical Center of the Johannes Gutenberg University Mainz, Mainz, Germany
- Center for Research in Inflammatory Diseases (CRID), Department of Pharmacology, Ribeirao Preto Medical School, University of Sao Paulo, São Paulo, Brazil
| | - Francesca Rampoldi
- Institute of Medical Microbiology and Hygiene, University Medical Center of the Johannes Gutenberg University Mainz, Mainz, Germany
- DKFZ German Cancer Research Center, Heidelberg, Germany
| | - Luciana Berod
- Research Center for Immunotherapy (FZI), University Medical Center of the Johannes Gutenberg University Mainz, Mainz, Germany
- Institute of Molecular Medicine, University Medical Center of the Johannes Gutenberg University Mainz, Mainz, Germany
| | - Lydia Lynch
- Molecular Biology, Princeton University, Princeton, NJ, USA
- Ludwig Cancer Research Institute, Princeton Branch, Princeton University, Princeton, NJ, USA
| | - Karsten Hiller
- Department of Bioinformatics and Biochemistry, Braunschweig Integrated Centre of Systems Biology, Technical University of Braunschweig, Braunschweig, Germany
| | - Tim Sparwasser
- Institute of Medical Microbiology and Hygiene, University Medical Center of the Johannes Gutenberg University Mainz, Mainz, Germany.
- Research Center for Immunotherapy (FZI), University Medical Center of the Johannes Gutenberg University Mainz, Mainz, Germany.
| |
Collapse
|
7
|
Fonseca-Pereira D, Bae S, Clay SL, Michaud M, MacDonald MH, Glickman JN, Garrett WS. The metabolic sensor LKB1 regulates ILC3 homeostasis and mitochondrial function. Cell Rep 2025; 44:115456. [PMID: 40120107 DOI: 10.1016/j.celrep.2025.115456] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2024] [Revised: 02/06/2025] [Accepted: 03/03/2025] [Indexed: 03/25/2025] Open
Abstract
Group 3 innate lymphoid cells (ILC3s) are tissue-resident cells that sense environmental cues, control infections, and promote tissue homeostasis at mucosal surfaces. The metabolic sensor liver kinase B1 (LKB1) integrates intracellular stress, metabolism, and mitochondrial function to promote the development and effector functions of a variety of immune cells; however, the role of LKB1 in ILC3 function was unknown. Here, we show that LKB1 is crucial for adult ILC3 homeostasis, cytokine production, and mitochondrial function. ILC3-specific LKB1 deletion resulted in a reduced number of ILC3s and interleukin-22 (IL-22) production. LKB1-deficient ILC3s had decreased survival, mitochondrial dysfunction, cytoplasmic lipid accumulation, and altered bioenergetics. Using LKB1 downstream kinase modulators, we found that LKB1 regulation of ILC3 survival and IL-22 production requires signaling through microtubule affinity-regulating kinases (MARKs). Mechanistically, LKB1 deficiency resulted in increased reactive oxygen species (ROS) production and NFAT2 and PD-1 expression. Our work reveals that metabolic regulation of enteric ILC3 function by an LKB1-dependent signaling network is crucial for intestinal immunity and tissue homeostasis.
Collapse
Affiliation(s)
- Diogo Fonseca-Pereira
- Department of Immunology and Infectious Diseases, Harvard T.H. Chan School of Public Health, Boston, MA, USA; Harvard T.H. Chan Microbiome in Public Health Center, Boston, MA, USA
| | - Sena Bae
- Department of Immunology and Infectious Diseases, Harvard T.H. Chan School of Public Health, Boston, MA, USA; Harvard T.H. Chan Microbiome in Public Health Center, Boston, MA, USA
| | - Slater L Clay
- Department of Immunology and Infectious Diseases, Harvard T.H. Chan School of Public Health, Boston, MA, USA; Harvard T.H. Chan Microbiome in Public Health Center, Boston, MA, USA
| | - Monia Michaud
- Department of Immunology and Infectious Diseases, Harvard T.H. Chan School of Public Health, Boston, MA, USA; Harvard T.H. Chan Microbiome in Public Health Center, Boston, MA, USA
| | - Meghan H MacDonald
- Department of Immunology and Infectious Diseases, Harvard T.H. Chan School of Public Health, Boston, MA, USA; Harvard T.H. Chan Microbiome in Public Health Center, Boston, MA, USA
| | - Jonathan N Glickman
- Gastrointestinal Pathology, Massachusetts General Hospital, Boston, MA, USA; Department of Pathology, Harvard Medical School, Boston, MA, USA
| | - Wendy S Garrett
- Department of Immunology and Infectious Diseases, Harvard T.H. Chan School of Public Health, Boston, MA, USA; Harvard T.H. Chan Microbiome in Public Health Center, Boston, MA, USA; Department of Molecular Metabolism, Harvard T.H. Chan School of Public Health, Boston, MA, USA; Broad Institute of MIT and Harvard, Cambridge, MA, USA; Division of Medical Sciences, Harvard Medical School, Boston, MA, USA; Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA.
| |
Collapse
|
8
|
Shirley S, Ichise H, Di Natale V, Jin J, Wu C, Zou R, Zhang W, Fang Y, Zhang Y, Chen M, Peng S, Basu U, Que J, Huang Y. A vasculature-resident innate lymphoid cell population in mouse lungs. Nat Commun 2025; 16:3718. [PMID: 40253407 PMCID: PMC12009297 DOI: 10.1038/s41467-025-58982-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2024] [Accepted: 04/08/2025] [Indexed: 04/21/2025] Open
Abstract
Tissue-resident immune cells such as innate lymphoid cells (ILC) are known to reside in the parenchymal compartments of tissues and modulate local immune protection. Here we use intravascular cell labeling, parabiosis and multiplex 3D imaging to identify a population of group 3 ILCs in mice that are present within the intravascular space of lung blood vessels (vILC3). vILC3s are distributed broadly in alveolar capillary beds from which inhaled pathogens enter the lung parenchyma. By contrast, conventional ILC3s in tissue parenchyma are enriched in lymphoid clusters in proximity to large veins. In a mouse model of pneumonia, Pseudomonas aeruginosa infection results in rapid vILC3 expansion and production of chemokines including CCL4. Blocking CCL4 in vivo attenuates neutrophil recruitment to the lung at the early stage of infection, resulting in prolonged inflammation and delayed bacterial clearance. Our findings thus define the intravascular space as a site of ILC residence in mice, and reveal a unique immune cell population that interfaces with tissue alarmins and the circulating immune system for timely host defense.
Collapse
Affiliation(s)
- Simon Shirley
- Department of Microbiology & Immunology, Columbia University Medical Center, New York, NY, USA
| | - Hiroshi Ichise
- Lymphocyte Biology Section, Laboratory of Immune Systems Biology, National Institute of Allergy and Infectious Disease, National Institutes of Health, Bethesda, 20892, MD, USA
| | - Vincenzo Di Natale
- Department of Microbiology & Immunology, Columbia University Medical Center, New York, NY, USA
| | - Jiacheng Jin
- Department of Microbiology & Immunology, Columbia University Medical Center, New York, NY, USA
| | - Christine Wu
- Department of Microbiology & Immunology, Columbia University Medical Center, New York, NY, USA
| | - Raymond Zou
- Department of Microbiology & Immunology, Columbia University Medical Center, New York, NY, USA
| | - Wanwei Zhang
- Department of Microbiology & Immunology, Columbia University Medical Center, New York, NY, USA
| | - Yinshan Fang
- Department of Medicine, Columbia Center for Human Development, Columbia University Medical Center, New York, NY, USA
| | - Yingyu Zhang
- Department of Microbiology & Immunology, Columbia University Medical Center, New York, NY, USA
| | - Miao Chen
- Department of Microbiology & Immunology, Columbia University Medical Center, New York, NY, USA
| | - Sophia Peng
- Department of Microbiology & Immunology, Columbia University Medical Center, New York, NY, USA
| | - Uttiya Basu
- Department of Microbiology & Immunology, Columbia University Medical Center, New York, NY, USA
| | - Jianwen Que
- Department of Medicine, Columbia Center for Human Development, Columbia University Medical Center, New York, NY, USA.
| | - Yuefeng Huang
- Department of Microbiology & Immunology, Columbia University Medical Center, New York, NY, USA.
| |
Collapse
|
9
|
Fu L, Upadhyay R, Pokrovskii M, Chen FM, Romero-Meza G, Griesemer A, Littman DR. PRDM16-dependent antigen-presenting cells induce tolerance to gut antigens. Nature 2025:10.1038/s41586-025-08982-4. [PMID: 40228524 DOI: 10.1038/s41586-025-08982-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2024] [Accepted: 04/04/2025] [Indexed: 04/16/2025]
Abstract
The gastrointestinal tract is continuously exposed to foreign antigens in food and commensal microorganisms with potential to induce adaptive immune responses. Peripherally induced T regulatory (pTreg) cells are essential for mitigating inflammatory responses to these agents1-4. Although RORγt+ antigen-presenting cells (APCs) have been shown to programme gut microbiota-specific pTreg cells5-7, their definition remains incomplete, and the APC responsible for food tolerance has remained unknown. Here we identify an APC subset that is required for differentiation of both food- and microbiota-specific pTreg cells and for establishment of oral tolerance. Development and function of these APCs require expression of the transcription factors PRDM16 and RORγt, as well as a unique Rorc(t) cis-regulatory element. Gene expression, chromatin accessibility, and surface marker analysis establish the pTreg-inducing APCs as myeloid in origin, distinct from type 3 innate lymphoid cells, and sharing epigenetic profiles with classical dendritic cells, and designate them PRDM16+RORγt+ tolerizing dendritic cells (tolDCs). Upon genetic perturbation of tolDCs, we observe a substantial increase in food antigen-specific T helper 2 cells in lieu of pTreg cells, leading to compromised tolerance in mouse models of asthma and food allergy. Single-cell analyses of freshly resected mesenteric lymph nodes from a human organ donor, as well as multiple specimens of human intestine and tonsil, reveal candidate tolDCs with co-expression of PRDM16 and RORC and an extensive transcriptome shared with tolDCs from mice, highlighting an evolutionarily conserved role across species. Our findings suggest that a better understanding of how tolDCs develop and how they regulate T cell responses to food and microbial antigens could offer new insights into developing therapeutic strategies for autoimmune and allergic diseases as well as organ transplant tolerance.
Collapse
Affiliation(s)
- Liuhui Fu
- Department of Cell Biology, New York University School of Medicine, New York, NY, USA
| | - Rabi Upadhyay
- Department of Cell Biology, New York University School of Medicine, New York, NY, USA
- Perlmutter Cancer Center, NYU Langone Health, New York, NY, USA
| | - Maria Pokrovskii
- Department of Cell Biology, New York University School of Medicine, New York, NY, USA
- Calico Life Sciences, South San Francisco, CA, USA
| | - Francis M Chen
- Department of Cell Biology, New York University School of Medicine, New York, NY, USA
| | - Gabriela Romero-Meza
- Department of Cell Biology, New York University School of Medicine, New York, NY, USA
- Howard Hughes Medical Institute, New York, NY, USA
| | - Adam Griesemer
- NYU Langone Transplant Institute, NYU Langone Health, New York, NY, USA
| | - Dan R Littman
- Department of Cell Biology, New York University School of Medicine, New York, NY, USA.
- Perlmutter Cancer Center, NYU Langone Health, New York, NY, USA.
- Howard Hughes Medical Institute, New York, NY, USA.
| |
Collapse
|
10
|
Narasimhan H, Richter ML, Shakiba R, Papaioannou NE, Stehle C, Ravi Rengarajan K, Ulmert I, Kendirli A, de la Rosa C, Kuo PY, Altman A, Münch P, Mahboubi S, Küntzel V, Sayed A, Stange EL, Pes J, Ulezko Antonova A, Pereira CF, Klein L, Dudziak D, Colonna M, Torow N, Hornef MW, Clausen BE, Kerschensteiner M, Lahl K, Romagnani C, Colomé-Tatché M, Schraml BU. RORγt-expressing dendritic cells are functionally versatile and evolutionarily conserved antigen-presenting cells. Proc Natl Acad Sci U S A 2025; 122:e2417308122. [PMID: 39993193 PMCID: PMC11892598 DOI: 10.1073/pnas.2417308122] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2024] [Accepted: 01/13/2025] [Indexed: 02/26/2025] Open
Abstract
Conventional dendritic cells (cDCs) are potent antigen-presenting cells (APCs) that integrate signals from their environment allowing them to direct situation-adapted immunity. Thereby they harbor great potential for being targeted in vaccination, autoimmunity, and cancer. Here, we use fate mapping, functional analyses, and comparative cross-species transcriptomics to show that RORγt+ DCs are a conserved, functionally versatile, and transcriptionally distinct type of DCs. RORγt+ DCs entail various populations described in different contexts including Janus cells/RORγt-expressing extrathymic Aire-expressing cells (eTACs), subtypes of Thetis cells, RORγt+-DC (R-DC) like cells, cDC2C and ACY3+ DCs. We show that in response to inflammatory triggers, RORγt+ DCs can migrate to lymph nodes and in the spleen can activate naïve CD4+ T cells. These findings expand the functional repertoire of RORγt+ DCs beyond the known role of eTACs and Thetis cells in inducing T cell tolerance to self-antigens and intestinal microbes in mice. We further show that RORγt+ DCs with proinflammatory features accumulate in autoimmune neuroinflammation in mice and men. Thus, our work establishes RORγt+ DCs as immune sentinel cells that exhibit a broad functional spectrum ranging from inducing peripheral T cell tolerance to T cell activation depending on signals they integrate from their environment.
Collapse
Affiliation(s)
- Hamsa Narasimhan
- Institute for Immunology, Biomedical Center Munich, Faculty of Medicine, Ludwig-Maximilians-Universität in Munich, Planegg-Martinsried82152, Germany
- Biomedical Center, Institute of Cardiovascular Physiology and Pathophysiology, Faculty of Medicine, Ludwig-Maximilians-Universität in Munich, Planegg-Martinsried82152, Germany
| | - Maria L. Richter
- Biomedical Center, Physiological Chemistry, Faculty of Medicine, Ludwig-Maximilians-Universität in Munich, Planegg-Martinsried82152, Germany
| | - Ramin Shakiba
- Institute for Immunology, Biomedical Center Munich, Faculty of Medicine, Ludwig-Maximilians-Universität in Munich, Planegg-Martinsried82152, Germany
- Biomedical Center, Institute of Cardiovascular Physiology and Pathophysiology, Faculty of Medicine, Ludwig-Maximilians-Universität in Munich, Planegg-Martinsried82152, Germany
| | - Nikos E. Papaioannou
- Biomedical Center, Institute of Cardiovascular Physiology and Pathophysiology, Faculty of Medicine, Ludwig-Maximilians-Universität in Munich, Planegg-Martinsried82152, Germany
| | - Christina Stehle
- Institute for Medical Immunology, Charité Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt Universität zu Berlin, Berlin10117, Germany
| | - Kaushikk Ravi Rengarajan
- Institute for Immunology, Biomedical Center Munich, Faculty of Medicine, Ludwig-Maximilians-Universität in Munich, Planegg-Martinsried82152, Germany
- Biomedical Center, Institute of Cardiovascular Physiology and Pathophysiology, Faculty of Medicine, Ludwig-Maximilians-Universität in Munich, Planegg-Martinsried82152, Germany
| | - Isabel Ulmert
- Section for Experimental and Translational Immunology, Institute for Health Technology, Technical University of Denmark, Kongens Lyngby2800, Denmark
| | - Arek Kendirli
- Institute of Clinical Neuroimmunology, University Hospital, Ludwig-Maximilians-Universität in Munich, Planegg-Martinsried82152, Germany
- Biomedical Center, Medical Faculty, Ludwig-Maximilians-Universität in Munich, Planegg-Martinsried82152, Germany
- Munich Cluster of Systems Neurology (SyNergy), Munich81377, Germany
| | - Clara de la Rosa
- Institute of Clinical Neuroimmunology, University Hospital, Ludwig-Maximilians-Universität in Munich, Planegg-Martinsried82152, Germany
- Biomedical Center, Medical Faculty, Ludwig-Maximilians-Universität in Munich, Planegg-Martinsried82152, Germany
- Graduate School of Systemic Neurosciences, LMU, Planegg-Martinsried82152, Germany
| | - Pin-Yu Kuo
- Institute for Immunology, Biomedical Center Munich, Faculty of Medicine, Ludwig-Maximilians-Universität in Munich, Planegg-Martinsried82152, Germany
- Biomedical Center, Institute of Cardiovascular Physiology and Pathophysiology, Faculty of Medicine, Ludwig-Maximilians-Universität in Munich, Planegg-Martinsried82152, Germany
| | - Abigail Altman
- Molecular Medicine and Gene Therapy, Lund Stem Cell Centre, Lund University, Lund221 84, Sweden
- Wallenberg Centre for Molecular Medicine at Lund University, Lund221 84, Sweden
| | - Philipp Münch
- Institute for Molecular Medicine and Research Center for Immunotherapy (Forschungszentrum für Immuntherapie), University Medical Center Johannes Gutenberg-University Mainz, Mainz55131, Germany
| | - Saba Mahboubi
- Institute for Molecular Medicine and Research Center for Immunotherapy (Forschungszentrum für Immuntherapie), University Medical Center Johannes Gutenberg-University Mainz, Mainz55131, Germany
| | - Vanessa Küntzel
- Biomedical Center, Institute of Cardiovascular Physiology and Pathophysiology, Faculty of Medicine, Ludwig-Maximilians-Universität in Munich, Planegg-Martinsried82152, Germany
| | - Amina Sayed
- Institute for Immunology, Biomedical Center Munich, Faculty of Medicine, Ludwig-Maximilians-Universität in Munich, Planegg-Martinsried82152, Germany
| | - Eva-Lena Stange
- Institute of Medical Microbiology, Rheinisch-Westfälische Technische Hochschule Aachen University Hospital, Aachen52074, Germany
| | - Jonas Pes
- Institute of Medical Microbiology, Rheinisch-Westfälische Technische Hochschule Aachen University Hospital, Aachen52074, Germany
| | - Alina Ulezko Antonova
- Department of Pathology and Immunology, Washington University School of Medicine, Saint Louis, MO63110
| | - Carlos-Filipe Pereira
- Molecular Medicine and Gene Therapy, Lund Stem Cell Centre, Lund University, Lund221 84, Sweden
- Wallenberg Centre for Molecular Medicine at Lund University, Lund221 84, Sweden
| | - Ludger Klein
- Institute for Immunology, Biomedical Center Munich, Faculty of Medicine, Ludwig-Maximilians-Universität in Munich, Planegg-Martinsried82152, Germany
| | - Diana Dudziak
- Institute of Immunology, Jena University Hospital of the Friedrich-Schiller-University, Jena07747, Germany
| | - Marco Colonna
- Department of Pathology and Immunology, Washington University School of Medicine, Saint Louis, MO63110
| | - Natalia Torow
- Institute of Medical Microbiology, Rheinisch-Westfälische Technische Hochschule Aachen University Hospital, Aachen52074, Germany
| | - Mathias W. Hornef
- Institute of Medical Microbiology, Rheinisch-Westfälische Technische Hochschule Aachen University Hospital, Aachen52074, Germany
| | - Björn E. Clausen
- Institute for Molecular Medicine and Research Center for Immunotherapy (Forschungszentrum für Immuntherapie), University Medical Center Johannes Gutenberg-University Mainz, Mainz55131, Germany
| | - Martin Kerschensteiner
- Institute of Clinical Neuroimmunology, University Hospital, Ludwig-Maximilians-Universität in Munich, Planegg-Martinsried82152, Germany
- Biomedical Center, Medical Faculty, Ludwig-Maximilians-Universität in Munich, Planegg-Martinsried82152, Germany
- Munich Cluster of Systems Neurology (SyNergy), Munich81377, Germany
| | - Katharina Lahl
- Section for Experimental and Translational Immunology, Institute for Health Technology, Technical University of Denmark, Kongens Lyngby2800, Denmark
- Immunology Section, Lund University, Lund221 84, Sweden
- Calvin, Phoebe and Joan Snyder Institute for Chronic Diseases, University of Calgary, Calgary, ABT2N 1N4, Canada
- Department of Microbiology, Immunology, and Infectious Diseases, Cumming School of Medicine, University of Calgary, Calgary, ABT2N 1N4, Canada
| | - Chiara Romagnani
- Institute for Medical Immunology, Charité Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt Universität zu Berlin, Berlin10117, Germany
| | - Maria Colomé-Tatché
- Biomedical Center, Physiological Chemistry, Faculty of Medicine, Ludwig-Maximilians-Universität in Munich, Planegg-Martinsried82152, Germany
| | - Barbara U. Schraml
- Institute for Immunology, Biomedical Center Munich, Faculty of Medicine, Ludwig-Maximilians-Universität in Munich, Planegg-Martinsried82152, Germany
- Biomedical Center, Institute of Cardiovascular Physiology and Pathophysiology, Faculty of Medicine, Ludwig-Maximilians-Universität in Munich, Planegg-Martinsried82152, Germany
| |
Collapse
|
11
|
Wang W, Li N, Xu H, Wei S, Li Y, Ou J, Hao J, Zhang J, Dong L, Qiu Y, Hu X, Fu YX, Guo X. ILC3s regulate the gut microbiota via host intestinal galactosylation to limit pathogen infection in mice. Nat Microbiol 2025; 10:654-666. [PMID: 39962279 DOI: 10.1038/s41564-025-01933-9] [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: 02/28/2024] [Accepted: 01/13/2025] [Indexed: 02/27/2025]
Abstract
Host immunity and commensal bacteria synergistically maintain intestinal homeostasis and mediate colonization resistance against pathogens. However, the molecular and cellular mechanisms remain unclear. Here, with a mouse infection model of Citrobacter rodentium, a natural mouse intestinal pathogen that mimics human enteropathogenic Escherichia coli and enterohaemorrhagic Escherichia coli, we find that group 3 innate lymphoid cells (ILC3s) can protect the host from infection by regulating gut microbiota. Mechanistically, ILC3s can control gut dysbiosis through IL-22-dependent regulation of intestinal galactosylation in mice. ILC3 deficiency led to an increase in intestinal galactosylation and the expansion of commensal Akkermansia muciniphila in colonic mucus. The increased A. muciniphila and A. muciniphila-derived metabolic product succinate further promoted the expression of pathogen virulence factors tir and ler, resulting in increased susceptibility to C. rodentium infection. Together, our data reveal a mechanism for ILC3s in protecting against pathogen infection through the regulation of intestinal glycosylation and gut microbiota metabolism.
Collapse
Affiliation(s)
- Wenyan Wang
- Institute for Immunology, Tsinghua University, Beijing, China.
- School of Basic Medical Sciences, Tsinghua University, Beijing, China.
- State Key Laboratory of Molecular Oncology, School of Basic Medical Sciences, Tsinghua University, Beijing, China.
| | - Na Li
- Institute for Immunology, Tsinghua University, Beijing, China
- School of Basic Medical Sciences, Tsinghua University, Beijing, China
- State Key Laboratory of Molecular Oncology, School of Basic Medical Sciences, Tsinghua University, Beijing, China
| | - Hongkai Xu
- Institute for Immunology, Tsinghua University, Beijing, China
- School of Basic Medical Sciences, Tsinghua University, Beijing, China
| | - Siting Wei
- Institute for Immunology, Tsinghua University, Beijing, China
- School of Basic Medical Sciences, Tsinghua University, Beijing, China
- State Key Laboratory of Molecular Oncology, School of Basic Medical Sciences, Tsinghua University, Beijing, China
| | - Yiping Li
- Institute for Immunology, Tsinghua University, Beijing, China
- School of Basic Medical Sciences, Tsinghua University, Beijing, China
| | - Jiayao Ou
- Institute for Immunology, Tsinghua University, Beijing, China
- School of Basic Medical Sciences, Tsinghua University, Beijing, China
| | - Jiacheng Hao
- Institute for Immunology, Tsinghua University, Beijing, China
- School of Basic Medical Sciences, Tsinghua University, Beijing, China
- State Key Laboratory of Molecular Oncology, School of Basic Medical Sciences, Tsinghua University, Beijing, China
| | - Jing Zhang
- Institute for Immunology, Tsinghua University, Beijing, China
- School of Basic Medical Sciences, Tsinghua University, Beijing, China
| | | | - Ying Qiu
- School of Basic Medical Sciences, Tsinghua University, Beijing, China
- State Key Laboratory of Molecular Oncology, School of Basic Medical Sciences, Tsinghua University, Beijing, China
| | - Xiaoyu Hu
- Institute for Immunology, Tsinghua University, Beijing, China
- School of Basic Medical Sciences, Tsinghua University, Beijing, China
- Tsinghua-Peking Center for Life Sciences, Beijing, China
| | - Yang-Xin Fu
- School of Basic Medical Sciences, Tsinghua University, Beijing, China
- State Key Laboratory of Molecular Oncology, School of Basic Medical Sciences, Tsinghua University, Beijing, China
| | - Xiaohuan Guo
- Institute for Immunology, Tsinghua University, Beijing, China.
- School of Basic Medical Sciences, Tsinghua University, Beijing, China.
- State Key Laboratory of Molecular Oncology, School of Basic Medical Sciences, Tsinghua University, Beijing, China.
- SXMU-Tsinghua Collaborative Innovation Center for Frontier Medicine, Shanxi Medical University, Taiyuan, China.
| |
Collapse
|
12
|
Li J, Jacobse J, Pilat JM, Kaur H, Gu W, Kang SW, Rusznak M, Huang HI, Barrera J, Oloo PA, Roland JT, Hawkins CV, Pahnke AP, Khalil M, Washington MK, Wilson KT, Williams CS, Peebles RS, Konnikova L, Choksi YA, Hammer GE, Lau KS, Goettel JA. Interleukin-10 production by innate lymphoid cells restricts intestinal inflammation in mice. Mucosal Immunol 2025:S1933-0219(25)00023-6. [PMID: 39988202 DOI: 10.1016/j.mucimm.2025.02.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2024] [Revised: 02/13/2025] [Accepted: 02/18/2025] [Indexed: 02/25/2025]
Abstract
Interleukin-10 (IL-10) is an immunomodulatory cytokine critical for intestinal immune homeostasis. IL-10 is produced by various immune cells but IL-10 receptor signaling in intestinal CX3CR1+ mononuclear phagocytes is necessary to prevent spontaneous colitis in mice. Here, we utilized fluorescent protein reporters and cell-specific targeting and found that Rorc-expressing innate lymphoid cells (ILCs) produce IL-10 in response to anti-CD40-mediated intestinal inflammation. Deletion of Il10 specifically in Rorc-expressing ILCs led to phenotypic changes in intestinal macrophages and exacerbated both innate and adaptive immune-mediated models of experimental colitis. The population of IL-10+ producing ILCs shared markers with both ILC2 and ILC3 with nearly all ILC3s being of the NCR+ subtype. Interestingly, Ccl26 was enriched in IL-10+ ILCs and was markedly reduced in IL-10-deficient ILC3s. Since CCL26 is a ligand for CX3CR1, we employed RNA in situ hybridization and observed increased numbers of ILCs in close proximity to Cx3cr1-expressing cells under inflammatory conditions. Finally, we generated transgenic RorctdTomato reporter mice that faithfully marked RORγt+ cells that could rescue disease pathology and aberrant macrophage phenotype following adoptive transfer into mice with selective Il10 deficiency in ILC3s. These results demonstrate that IL-10 production by a population of ILCs functions to promote immune homeostasis in the intestine possibly via direct effects on intestinal macrophages.
Collapse
Affiliation(s)
- Jing Li
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Justin Jacobse
- Department of Medicine, Division of Gastroenterology, Hepatology and Nutrition, Vanderbilt University Medical Center, Nashville, TN, USA; Willem-Alexander Children's Hospital, Department of Pediatrics, Leiden University Medical Center, Leiden, the Netherlands; Veterans Affairs Tennessee Valley Healthcare System, Nashville, TN USA 37212
| | - Jennifer M Pilat
- Department of Medicine, Division of Gastroenterology, Hepatology and Nutrition, Vanderbilt University Medical Center, Nashville, TN, USA; Program in Cancer Biology, Vanderbilt University School of Medicine, Nashville, TN, USA
| | - Harsimran Kaur
- Program in Chemical and Physical Biology, Vanderbilt University School of Medicine, Nashville, TN, USA
| | - Weihong Gu
- Department of Pediatrics, Yale Medical School, New Haven, CT 06520, USA
| | - Seung Woo Kang
- Department of Cell and Developmental Biology, Vanderbilt University School of Medicine, Nashville, TN, USA
| | - Mark Rusznak
- Department of Internal Medicine Vanderbilt University Medical Center, Nashville, TN, USA
| | - Hsin-I Huang
- Department of Pathology, Division of Microbiology and Immunology, University of Utah, Salt Lake City, UT 84112, USA
| | - Julio Barrera
- Department of Pathology, Division of Microbiology and Immunology, University of Utah, Salt Lake City, UT 84112, USA
| | - Pauline A Oloo
- Department of Pathology, Division of Microbiology and Immunology, University of Utah, Salt Lake City, UT 84112, USA
| | - Joseph T Roland
- Department of Cell and Developmental Biology, Vanderbilt University School of Medicine, Nashville, TN, USA; Department of Internal Medicine Vanderbilt University Medical Center, Nashville, TN, USA
| | - Caroline V Hawkins
- Department of Medicine, Division of Gastroenterology, Hepatology and Nutrition, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Andrew P Pahnke
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Marian Khalil
- Department of Medicine, Division of Gastroenterology, Hepatology and Nutrition, Vanderbilt University Medical Center, Nashville, TN, USA
| | - M Kay Washington
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, Nashville, TN, USA; Center for Mucosal Inflammation and Cancer, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Keith T Wilson
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, Nashville, TN, USA; Department of Medicine, Division of Gastroenterology, Hepatology and Nutrition, Vanderbilt University Medical Center, Nashville, TN, USA; Veterans Affairs Tennessee Valley Healthcare System, Nashville, TN USA 37212; Program in Cancer Biology, Vanderbilt University School of Medicine, Nashville, TN, USA; Center for Mucosal Inflammation and Cancer, Vanderbilt University Medical Center, Nashville, TN, USA; Vanderbilt Institute for Infection, Immunology and Inflammation, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Christopher S Williams
- Department of Medicine, Division of Gastroenterology, Hepatology and Nutrition, Vanderbilt University Medical Center, Nashville, TN, USA; Veterans Affairs Tennessee Valley Healthcare System, Nashville, TN USA 37212; Program in Cancer Biology, Vanderbilt University School of Medicine, Nashville, TN, USA; Center for Mucosal Inflammation and Cancer, Vanderbilt University Medical Center, Nashville, TN, USA
| | - R Stokes Peebles
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, Nashville, TN, USA; Veterans Affairs Tennessee Valley Healthcare System, Nashville, TN USA 37212; Department of Internal Medicine Vanderbilt University Medical Center, Nashville, TN, USA
| | - Liza Konnikova
- Department of Pediatrics, Yale Medical School, New Haven, CT 06520, USA; Department of Immunobiology, Yale School of Medicine, New Haven, CT 06520, USA; Center for Systems and Engineering Immunology, Yale School of Medicine, New Haven, CT 06520, USA; Human and Translational Immunology Program, Yale School of Medicine, New Haven, CT 06520, USA; Program in Translational Biomedicine, Yale School of Medicine, New Haven, CT 06520, USA; Department of Obstetrics Gynecology and Reproductive Sciences, Yale School of Medicine, New Haven, CT 06520, USA
| | - Yash A Choksi
- Department of Medicine, Division of Gastroenterology, Hepatology and Nutrition, Vanderbilt University Medical Center, Nashville, TN, USA; Veterans Affairs Tennessee Valley Healthcare System, Nashville, TN USA 37212; Program in Cancer Biology, Vanderbilt University School of Medicine, Nashville, TN, USA; Center for Mucosal Inflammation and Cancer, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Gianna Elena Hammer
- Department of Pathology, Division of Microbiology and Immunology, University of Utah, Salt Lake City, UT 84112, USA
| | - Ken S Lau
- Program in Chemical and Physical Biology, Vanderbilt University School of Medicine, Nashville, TN, USA; Department of Cell and Developmental Biology, Vanderbilt University School of Medicine, Nashville, TN, USA; Department of Pathology, Division of Microbiology and Immunology, University of Utah, Salt Lake City, UT 84112, USA; Department of Surgery, Vanderbilt University Medical Center, Nashville, TN, USA; Epithelial Biology Center, Vanderbilt University Medical Center, Nashville, TN, USA; Center for Computational Systems Biology, Vanderbilt University, Nashville, TN, USA
| | - Jeremy A Goettel
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, Nashville, TN, USA; Department of Medicine, Division of Gastroenterology, Hepatology and Nutrition, Vanderbilt University Medical Center, Nashville, TN, USA; Program in Cancer Biology, Vanderbilt University School of Medicine, Nashville, TN, USA; Center for Mucosal Inflammation and Cancer, Vanderbilt University Medical Center, Nashville, TN, USA; Vanderbilt Institute for Infection, Immunology and Inflammation, Vanderbilt University Medical Center, Nashville, TN, USA.
| |
Collapse
|
13
|
Zeng Q, Guo H, Tang N, Renavikar PS, Karandikar NJ, Lovett-Racke AE, Racke MK, Yan C, Tang R, Sinha S, Ghosh K, Ryal JP, Ouyang S, Chen M, Amari F, Vincenzo C, Pope RM, Li Y, Yang H, Langdon WY, Zhang J. K27-linked RORγt ubiquitination by Nedd4 potentiates Th17-mediated autoimmunity. J Biomed Sci 2025; 32:26. [PMID: 39972304 PMCID: PMC11841259 DOI: 10.1186/s12929-025-01120-2] [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: 05/28/2024] [Accepted: 01/17/2025] [Indexed: 02/21/2025] Open
Abstract
BACKGROUND The HECT E3 ubiquitin ligase Nedd4 has been shown to positively regulate T cell responses, but its role in T helper (Th) cell differentiation and autoimmunity is unknown. Th17 cells are believed to play a pivotal role in the development and pathogenesis of autoimmune diseases. Nevertheless, the regulation of RORγt activation during Th17 cell differentiation by TCR signaling is yet to be elucidated. These uncharted aspects inspire us to explore the potential role of Nedd4 in Th17-mediated autoimmunity. METHODS We evaluated the impact of Nedd4 deficiency on mouse T cell development and differentiation using flow cytometry and siRNA transfection, and subsequently validated these findings in T cells from patients with multiple sclerosis (MS). Furthermore, we investigated the influence of Nedd4 deficiency on Th17-mediated autoimmunity through experimental autoimmune encephalomyelitis (EAE), a mouse model of MS. Subsequently, we elucidated the molecular mechanism underlying the interaction between Nedd4 and RORgt through immunoprecipitation, mass spectrometry analysis, and lentiviral transduction. Additionally, we identified Nedd4 as an E3 ubiquitin ligase for RORγt. Moreover, we characterized the tyrosine residue sites and polyubiquitination patterns involved in RORγt ubiquitination. RESULTS In this study, we report that loss of Nedd4 in T cells specifically impairs pathogenic and non-pathogenic Th17 responses, and Th17-mediated EAE development. At the molecular level, Nedd4 binds to the PPLY motif within the ligand binding domain of RORγt, and targets RORγt at K112 for K27-linked polyubiquitination, thus augmenting its activity. CONCLUSION Nedd4 is a crucial E3 ubiquitin ligase for RORγt in the regulating Th17 cell development and offers potential therapeutic benefits for treating Th17-mediated autoimmune diseases.
Collapse
MESH Headings
- Nedd4 Ubiquitin Protein Ligases/genetics
- Nedd4 Ubiquitin Protein Ligases/metabolism
- Nedd4 Ubiquitin Protein Ligases/immunology
- Animals
- Th17 Cells/immunology
- Mice
- Ubiquitination
- Humans
- Autoimmunity/genetics
- Nuclear Receptor Subfamily 1, Group F, Member 3/genetics
- Nuclear Receptor Subfamily 1, Group F, Member 3/immunology
- Nuclear Receptor Subfamily 1, Group F, Member 3/metabolism
- Encephalomyelitis, Autoimmune, Experimental/immunology
- Encephalomyelitis, Autoimmune, Experimental/genetics
- Multiple Sclerosis/immunology
- Multiple Sclerosis/genetics
- Mice, Inbred C57BL
- Cell Differentiation
Collapse
Affiliation(s)
- Qiuming Zeng
- Department of Neurology, Xiangya Hospital, Central South University, Changsha, Hunan, 410008, People's Republic of China.
- Department of Pathology, The University of Iowa Roy J. and Lucille A. Carver College of Medicine, Iowa City, IA, 52242, USA.
- Department of Microbial Infection and Immunity, The Ohio State University, Columbus, OH, 43210, USA.
- Clinical Research Center for Neuroimmune and Neuromuscular Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, 410008, People's Republic of China.
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, 410008, People's Republic of China.
| | - Hui Guo
- Department of Pathology, The University of Iowa Roy J. and Lucille A. Carver College of Medicine, Iowa City, IA, 52242, USA.
- Department of Microbial Infection and Immunity, The Ohio State University, Columbus, OH, 43210, USA.
| | - Na Tang
- Department of Pathology, The University of Iowa Roy J. and Lucille A. Carver College of Medicine, Iowa City, IA, 52242, USA
- Department of Microbial Infection and Immunity, The Ohio State University, Columbus, OH, 43210, USA
| | - Pranav S Renavikar
- Department of Pathology, The University of Iowa Roy J. and Lucille A. Carver College of Medicine, Iowa City, IA, 52242, USA
| | - Nitin J Karandikar
- Department of Pathology, The University of Iowa Roy J. and Lucille A. Carver College of Medicine, Iowa City, IA, 52242, USA
| | - Amy E Lovett-Racke
- Department of Microbial Infection and Immunity, The Ohio State University, Columbus, OH, 43210, USA
| | - Michael K Racke
- Department of Neurology, The Ohio State University, Columbus, OH, 43210, USA
| | - Chengkai Yan
- Department of Pathology, The University of Iowa Roy J. and Lucille A. Carver College of Medicine, Iowa City, IA, 52242, USA
- Department of Microbial Infection and Immunity, The Ohio State University, Columbus, OH, 43210, USA
| | - Rong Tang
- Department of Microbial Infection and Immunity, The Ohio State University, Columbus, OH, 43210, USA
| | - Sushmita Sinha
- Department of Pathology, The University of Iowa Roy J. and Lucille A. Carver College of Medicine, Iowa City, IA, 52242, USA
| | - Krishnendu Ghosh
- Department of Pathology, The University of Iowa Roy J. and Lucille A. Carver College of Medicine, Iowa City, IA, 52242, USA
| | - Jeremy P Ryal
- Department of Pathology, The University of Iowa Roy J. and Lucille A. Carver College of Medicine, Iowa City, IA, 52242, USA
| | - Song Ouyang
- Department of Microbial Infection and Immunity, The Ohio State University, Columbus, OH, 43210, USA
| | - Min Chen
- Genetically Engineered Mouse Modeling Core, The Ohio State University, Columbus, OH, 43210, USA
| | - Foued Amari
- Genetically Engineered Mouse Modeling Core, The Ohio State University, Columbus, OH, 43210, USA
| | - Coppola Vincenzo
- Genetically Engineered Mouse Modeling Core, The Ohio State University, Columbus, OH, 43210, USA
- Department of Cancer Biology and Genetics, The Ohio State University, Columbus, OH, 43210, USA
| | - R Marshall Pope
- Proteomics Facility, The University of Iowa Roy J. and Lucille A. Carver College of Medicine, Iowa City, IA, USA
| | - Yalan Li
- Genetically Engineered Mouse Modeling Core, The Ohio State University, Columbus, OH, 43210, USA
| | - Huan Yang
- Department of Pathology, The University of Iowa Roy J. and Lucille A. Carver College of Medicine, Iowa City, IA, 52242, USA
- Clinical Research Center for Neuroimmune and Neuromuscular Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, 410008, People's Republic of China
| | - Wallace Y Langdon
- School of Biomedical Sciences, The University of Western Australia, Perth, Australia
| | - Jian Zhang
- Department of Pathology, The University of Iowa Roy J. and Lucille A. Carver College of Medicine, Iowa City, IA, 52242, USA.
- Department of Microbial Infection and Immunity, The Ohio State University, Columbus, OH, 43210, USA.
| |
Collapse
|
14
|
Roberts LB, Neves JF, Lee DCH, Valpione S, Tachó-Piñot R, Howard JK, Hepworth MR, Lord GM. MicroRNA-142 regulates gut associated lymphoid tissues and group 3 innate lymphoid cells. Mucosal Immunol 2025; 18:39-52. [PMID: 39245145 PMCID: PMC11835792 DOI: 10.1016/j.mucimm.2024.09.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2023] [Revised: 08/26/2024] [Accepted: 09/02/2024] [Indexed: 09/10/2024]
Abstract
The transcriptomic signatures that shape responses of innate lymphoid cells (ILCs) have been well characterised, however post-transcriptional mechanisms which regulate their development and activity remain poorly understood. We demonstrate that ILC groups of the intestinal lamina propria express mature forms of microRNA-142 (miR-142), an evolutionarily conserved microRNA family with several non-redundant regulatory roles within the immune system. Germline Mir142 deletion alters intestinal ILC compositions, resulting in the absence of T-bet+ populations and significant defects in the cellularity and phenotypes of ILC3 subsets including CCR6+ LTi-like ILC3s. These effects were associated with decreased pathology in an innate-immune cell driven model of colitis. Furthermore, Mir142-/- mice demonstrate defective development of gut-associated lymphoid tissues, including a complete absence of mature Peyer's patches. Conditional deletion of Mir142 in ILC3s (RorcΔMir142) supported cell-intrinsic roles for these microRNAs in establishing or maintaining cellularity and functions of LTi-like ILC3s in intestinal associated tissues. RNAseq analysis revealed several target genes and biological pathways potentially regulated by miR-142 microRNAs in these cells. Finally, lack of Mir142 in ILC3 led to elevated IL-17A production. These data broaden our understanding of immune system roles of miR-142 microRNAs, identifying these molecules as critical post-transcriptional regulators of ILC3s and intestinal mucosal immunity.
Collapse
Affiliation(s)
- Luke B Roberts
- Lydia Becker Institute of Immunology and Inflammation, University of Manchester, M13 9PL, United Kingdom.
| | - Joana F Neves
- Centre for Host-Microbiome Interactions, King's College London, Great Maze Pond, London SE1 9RT, United Kingdom
| | - Dave C H Lee
- Lydia Becker Institute of Immunology and Inflammation, University of Manchester, M13 9PL, United Kingdom
| | - Sara Valpione
- The Christie NHS Foundation Trust, 550 Wilmslow Road, M20 4BX Manchester, United Kingdom; Division of Cancer Sciences, The University of Manchester, Oxford Road, M13 9PL Manchester, United Kingdom; Cancer Research UK National Biomarker Centre, Wilmslow Road, M20 4BX Manchester, United Kingdom
| | - Roser Tachó-Piñot
- Lydia Becker Institute of Immunology and Inflammation, University of Manchester, M13 9PL, United Kingdom
| | - Jane K Howard
- School of Cardiovascular and Metabolic Medicine & Sciences, King's College London, Great Maze Pond, London SE1 9RT, United Kingdom
| | - Matthew R Hepworth
- Lydia Becker Institute of Immunology and Inflammation, University of Manchester, M13 9PL, United Kingdom
| | - Graham M Lord
- Lydia Becker Institute of Immunology and Inflammation, University of Manchester, M13 9PL, United Kingdom; Centre for Gene Therapy and Regenerative Medicine, School of Basic and Medical Biosciences, Faculty of Life Sciences and Medicine, King's College London, United Kingdom.
| |
Collapse
|
15
|
Wu J, Wang K, Qi X, Zhou S, Zhao S, Lu M, Nie Q, Li M, Han M, Luo X, Yun C, Wang P, Li R, Zhong C, Yu X, Yin WB, Jiang C, Qiao J, Pang Y. The intestinal fungus Aspergillus tubingensis promotes polycystic ovary syndrome through a secondary metabolite. Cell Host Microbe 2025; 33:119-136.e11. [PMID: 39788092 DOI: 10.1016/j.chom.2024.12.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2024] [Revised: 09/25/2024] [Accepted: 12/06/2024] [Indexed: 01/12/2025]
Abstract
Polycystic ovary syndrome (PCOS) affects 6%-10% of women of reproductive age and is known to be associated with disruptions in the gut bacteria. However, the role of the gut mycobiota in PCOS pathology remains unclear. Using culture-dependent and internal transcribed spacer 2 (ITS2)-sequencing methods, we discovered an enrichment of the gut-colonizable fungus Aspergillus tubingensis in 226 individuals, with or without PCOS, from 3 different geographical areas within China. Colonization of mice with A. tubingensis led to a PCOS-like phenotype due to inhibition of Aryl hydrocarbon receptor (AhR) signaling and reduced interleukin (IL)-22 secretion in intestinal group 3 innate lymphoid cells (ILC3s). By developing a strain-diversity-based-activity metabolite screening workflow, we identified secondary metabolite AT-C1 as an endogenous AhR antagonist and a key mediator of PCOS. Our findings demonstrate that an intestinal fungus and its secondary metabolite play a critical role in PCOS pathogenesis, offering a therapeutic strategy for improving the management of the disease.
Collapse
Affiliation(s)
- Jiayu Wu
- State Key Laboratory of Female Fertility Promotion, Center for Reproductive Medicine, Department of Obstetrics and Gynecology, Peking University Third Hospital, Beijing, China; Department of Immunology, School of Basic Medical Sciences, State Key Laboratory of Vascular Homeostasis and Remodeling, Peking University, Beijing 100191, China; NHC Key Laboratory of Medical Immunology, Peking University, Beijing, China
| | - Kai Wang
- State Key Laboratory of Female Fertility Promotion, Center for Reproductive Medicine, Department of Obstetrics and Gynecology, Peking University Third Hospital, Beijing, China; Department of Immunology, School of Basic Medical Sciences, State Key Laboratory of Vascular Homeostasis and Remodeling, Peking University, Beijing 100191, China; NHC Key Laboratory of Medical Immunology, Peking University, Beijing, China
| | - Xinyu Qi
- State Key Laboratory of Female Fertility Promotion, Center for Reproductive Medicine, Department of Obstetrics and Gynecology, Peking University Third Hospital, Beijing, China; Institute of Advanced Clinical Medicine, Peking University, National Clinical Research Center for Obstetrics and Gynecology (Peking University Third Hospital), Beijing, China; Beijing Key Laboratory of Reproductive Endocrinology and Assisted Reproductive Technology, Beijing, China
| | - Shuang Zhou
- State Key Laboratory of Female Fertility Promotion, Center for Reproductive Medicine, Department of Obstetrics and Gynecology, Peking University Third Hospital, Beijing, China; Department of Immunology, School of Basic Medical Sciences, State Key Laboratory of Vascular Homeostasis and Remodeling, Peking University, Beijing 100191, China; NHC Key Laboratory of Medical Immunology, Peking University, Beijing, China
| | - Shuyun Zhao
- Reproductive Medicine Center, Department of Obstetrics and Gynecology, The Affiliated Hospital of Guizhou Medical University, Guiyang, Guizhou, China
| | - Meisong Lu
- The First Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang, China
| | - Qixing Nie
- State Key Laboratory of Female Fertility Promotion, Center for Reproductive Medicine, Department of Obstetrics and Gynecology, Peking University Third Hospital, Beijing, China
| | - Meng Li
- Department of Physiology and Pathophysiology, Center for Obesity and Metabolic Disease Research, School of Basic Medical Sciences, State Key Laboratory of Vascular Homeostasis and Remodeling, Peking University, Beijing 100191, China
| | - Mengwei Han
- Institute of Systems Biomedicine, School of Basic Medical Sciences, Peking University Health Science Center, Beijing Key Laboratory of Tumor Systems Biology, Beijing, China
| | - Xi Luo
- Department of Physiology and Pathophysiology, Center for Obesity and Metabolic Disease Research, School of Basic Medical Sciences, State Key Laboratory of Vascular Homeostasis and Remodeling, Peking University, Beijing 100191, China
| | - Chuyu Yun
- State Key Laboratory of Female Fertility Promotion, Center for Reproductive Medicine, Department of Obstetrics and Gynecology, Peking University Third Hospital, Beijing, China; Institute of Advanced Clinical Medicine, Peking University, National Clinical Research Center for Obstetrics and Gynecology (Peking University Third Hospital), Beijing, China; Beijing Key Laboratory of Reproductive Endocrinology and Assisted Reproductive Technology, Beijing, China
| | - Pengcheng Wang
- Center of Basic Medical Research, Institute of Medical Innovation and Research, Peking University Third Hospital, Beijing, China
| | - Rong Li
- State Key Laboratory of Female Fertility Promotion, Center for Reproductive Medicine, Department of Obstetrics and Gynecology, Peking University Third Hospital, Beijing, China; Institute of Advanced Clinical Medicine, Peking University, National Clinical Research Center for Obstetrics and Gynecology (Peking University Third Hospital), Beijing, China; Beijing Key Laboratory of Reproductive Endocrinology and Assisted Reproductive Technology, Beijing, China
| | - Chao Zhong
- Department of Immunology, School of Basic Medical Sciences, State Key Laboratory of Vascular Homeostasis and Remodeling, Peking University, Beijing 100191, China
| | - Xiaofei Yu
- State Key Laboratory of Genetic Engineering, School of Life Sciences, Fudan University, Shanghai 200438, China
| | - Wen-Bing Yin
- State Key Laboratory of Mycology, Institute of Microbiology, Chinese Academy of Sciences, Beijing 100101, China
| | - Changtao Jiang
- State Key Laboratory of Female Fertility Promotion, Center for Reproductive Medicine, Department of Obstetrics and Gynecology, Peking University Third Hospital, Beijing, China; Department of Immunology, School of Basic Medical Sciences, State Key Laboratory of Vascular Homeostasis and Remodeling, Peking University, Beijing 100191, China
| | - Jie Qiao
- State Key Laboratory of Female Fertility Promotion, Center for Reproductive Medicine, Department of Obstetrics and Gynecology, Peking University Third Hospital, Beijing, China; Institute of Advanced Clinical Medicine, Peking University, National Clinical Research Center for Obstetrics and Gynecology (Peking University Third Hospital), Beijing, China; Beijing Key Laboratory of Reproductive Endocrinology and Assisted Reproductive Technology, Beijing, China.
| | - Yanli Pang
- State Key Laboratory of Female Fertility Promotion, Center for Reproductive Medicine, Department of Obstetrics and Gynecology, Peking University Third Hospital, Beijing, China; Institute of Advanced Clinical Medicine, Peking University, National Clinical Research Center for Obstetrics and Gynecology (Peking University Third Hospital), Beijing, China; Beijing Key Laboratory of Reproductive Endocrinology and Assisted Reproductive Technology, Beijing, China.
| |
Collapse
|
16
|
Huang J, Zhang J, Song P, Huang J, Yang Z, Han J, Wu L, Guo X. p38α-eIF6-Nsun2 axis promotes ILC3's rapid response to protect host from intestinal inflammation. J Exp Med 2025; 222:e20240624. [PMID: 39589554 PMCID: PMC11602552 DOI: 10.1084/jem.20240624] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2024] [Revised: 09/01/2024] [Accepted: 10/08/2024] [Indexed: 11/27/2024] Open
Abstract
Group 3 innate lymphoid cells (ILC3s) are important for maintaining gut homeostasis. Upon stimulation, ILC3s can rapidly produce cytokines to protect against infections and colitis. However, the regulation of ILC3 quick response is still unclear. Here, we find that eIF6 aggregates with Nsun2 and cytokine mRNA in ILC3s at steady state, which inhibits the methyltransferase activity of Nsun2 and the nuclear export of cytokine mRNA, resulting in the nuclear reservation of cytokine mRNA. Upon stimulation, phosphorylated p38α phosphorylates eIF6, which in turn releases Nsun2 activity, and promotes the nuclear export of cytokine mRNA and rapid cytokine production. Genetic disruption of p38α, Nsun2, or eIF6 in ILC3s influences the mRNA nuclear export and protein expression of the protective cytokines, thus leading to increased susceptibility to colitis. Together, our data identify a crucial role of the p38α-eIF6-Nsun2 axis in regulating rapid ILC3 immune response at the posttranscriptional level, which is critical for gut homeostasis maintenance and protection against gut inflammation.
Collapse
Affiliation(s)
- Jida Huang
- Institute for Immunology, Tsinghua University, Beijing, China
- School of Basic Medical Sciences, Tsinghua University, Beijing, China
- Beijing Key Lab for Immunological Research on Chronic Diseases, Tsinghua University, Beijing, China
- State Key Laboratory of Molecular Oncology, School of Basic Medical Sciences, Tsinghua University, Beijing, China
| | - Jing Zhang
- Institute for Immunology, Tsinghua University, Beijing, China
- School of Basic Medical Sciences, Tsinghua University, Beijing, China
- Beijing Key Lab for Immunological Research on Chronic Diseases, Tsinghua University, Beijing, China
- State Key Laboratory of Molecular Oncology, School of Basic Medical Sciences, Tsinghua University, Beijing, China
| | - Panwei Song
- Institute for Immunology, Tsinghua University, Beijing, China
- School of Basic Medical Sciences, Tsinghua University, Beijing, China
- Beijing Key Lab for Immunological Research on Chronic Diseases, Tsinghua University, Beijing, China
- State Key Laboratory of Molecular Oncology, School of Basic Medical Sciences, Tsinghua University, Beijing, China
| | - Jiaoyan Huang
- Institute for Immunology, Tsinghua University, Beijing, China
- School of Basic Medical Sciences, Tsinghua University, Beijing, China
- Beijing Key Lab for Immunological Research on Chronic Diseases, Tsinghua University, Beijing, China
| | - Zi Yang
- Protein Preparation and Identification Facilities at Technology Center for Protein Science, School of Life Sciences, Tsinghua University, Beijing, China
| | - Jiahuai Han
- State Key Laboratory of Cellular Stress Biology, School of Life Sciences, Innovation Center for Cell Signaling Network, Xiamen University, Xiamen, China
| | - Li Wu
- Institute for Immunology, Tsinghua University, Beijing, China
- School of Basic Medical Sciences, 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
- School of Basic Medical Sciences, Tsinghua University, Beijing, China
- Beijing Key Lab for Immunological Research on Chronic Diseases, Tsinghua University, Beijing, China
- State Key Laboratory of Molecular Oncology, School of Basic Medical Sciences, Tsinghua University, Beijing, China
- SXMU-Tsinghua Collaborative Innovation Center for Frontier Medicine, Shanxi Medical University, Taiyuan, China
| |
Collapse
|
17
|
Onji M, Sigl V, Lendl T, Novatchkova M, Ullate-Agote A, Andersson-Rolf A, Kozieradzki I, Koglgruber R, Pai TP, Lichtscheidl D, Nayak K, Zilbauer M, Carranza García NA, Sievers LK, Falk-Paulsen M, Cronin SJF, Hagelkruys A, Sawa S, Osborne LC, Rosenstiel P, Pasparakis M, Ruland J, Takayanagi H, Clevers H, Koo BK, Penninger JM. RANK drives structured intestinal epithelial expansion during pregnancy. Nature 2025; 637:156-166. [PMID: 39633049 PMCID: PMC11666467 DOI: 10.1038/s41586-024-08284-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2023] [Accepted: 10/24/2024] [Indexed: 12/07/2024]
Abstract
During reproduction, multiple species such as insects and all mammals undergo extensive physiological and morphological adaptions to ensure health and survival of the mother and optimal development of the offspring. Here we report that the intestinal epithelium undergoes expansion during pregnancy and lactation in mammals. This enlargement of the intestinal surface area results in a novel geometry of expanded villi. Receptor activator of nuclear factor-κΒ (RANK, encoded by TNFRSF11A) and its ligand RANKL were identified as a molecular pathway involved in this villous expansion of the small intestine in vivo in mice and in intestinal mouse and human organoids. Mechanistically, RANK-RANKL protects gut epithelial cells from cell death and controls the intestinal stem cell niche through BMP receptor signalling, resulting in the elongation of villi and a prominent increase in the intestinal surface. As a transgenerational consequence, babies born to female mice that lack Rank in the intestinal epithelium show reduced weight and develop glucose intolerance after metabolic stress. Whereas gut epithelial remodelling in pregnancy/lactation is reversible, constitutive expression of an active form of RANK is sufficient to drive intestinal expansion followed by loss of villi and stem cells, and prevents the formation of Apcmin-driven small intestinal stem cell tumours. These data identify RANK-RANKL as a pathway that drives intestinal epithelial expansion in pregnancy/lactation, one of the most elusive and fundamental tissue remodelling events in mammalian life history and evolution.
Collapse
Affiliation(s)
- Masahiro Onji
- Institute of Molecular Biotechnology of the Austrian Academy of Sciences (IMBA), Vienna BioCenter (VBC), Vienna, Austria.
- Department of Laboratory Medicine, Medical University of Vienna, Vienna, Austria.
| | - Verena Sigl
- Institute of Molecular Biotechnology of the Austrian Academy of Sciences (IMBA), Vienna BioCenter (VBC), Vienna, Austria
| | - Thomas Lendl
- Institute of Molecular Pathology (IMP), Vienna BioCenter (VBC), Vienna, Austria
| | - Maria Novatchkova
- Institute of Molecular Pathology (IMP), Vienna BioCenter (VBC), Vienna, Austria
| | - Asier Ullate-Agote
- Biomedical Engineering Program, Center for Applied Medical Research (CIMA), Universidad de Navarra, Instituto de Investigación Sanitaria de Navarra (IdiSNA), Pamplona, Spain
| | - Amanda Andersson-Rolf
- Oncode Institute, Hubrecht Institute, Royal Netherlands Academy of Arts and Sciences (KNAW) and University Medical Center, Utrecht, The Netherlands
| | - Ivona Kozieradzki
- Department of Medical Genetics, Life Sciences Institute, University of British Columbia, Vancouver, British Columbia, Canada
| | - Rubina Koglgruber
- Institute of Molecular Biotechnology of the Austrian Academy of Sciences (IMBA), Vienna BioCenter (VBC), Vienna, Austria
- Department of Laboratory Medicine, Medical University of Vienna, Vienna, Austria
| | - Tsung-Pin Pai
- Institute of Molecular Biotechnology of the Austrian Academy of Sciences (IMBA), Vienna BioCenter (VBC), Vienna, Austria
| | - Dominic Lichtscheidl
- Institute of Molecular Biotechnology of the Austrian Academy of Sciences (IMBA), Vienna BioCenter (VBC), Vienna, Austria
| | - Komal Nayak
- Wellcome-MRC Cambridge Stem Cell Institute, University of Cambridge, Cambridge, UK
- Department of Paediatrics, University of Cambridge, Cambridge, UK
| | - Matthias Zilbauer
- Wellcome-MRC Cambridge Stem Cell Institute, University of Cambridge, Cambridge, UK
- Department of Paediatrics, University of Cambridge, Cambridge, UK
- Department of Paediatric Gastroenterology, Hepatology and Nutrition, Cambridge University Hospitals (CUH), Addenbrooke's, Cambridge, UK
| | - Natalia A Carranza García
- Department of Microbiology and Immunology, Life Sciences Institute, University of British Columbia, Vancouver, British Columbia, Canada
| | - Laura Katharina Sievers
- Institute of Clinical Molecular Biology, Kiel University and University Hospital Schleswig-Holstein, Kiel, Germany
| | - Maren Falk-Paulsen
- Institute of Clinical Molecular Biology, Kiel University and University Hospital Schleswig-Holstein, Kiel, Germany
| | - Shane J F Cronin
- Institute of Molecular Biotechnology of the Austrian Academy of Sciences (IMBA), Vienna BioCenter (VBC), Vienna, Austria
- Department of Laboratory Medicine, Medical University of Vienna, Vienna, Austria
| | - Astrid Hagelkruys
- Institute of Molecular Biotechnology of the Austrian Academy of Sciences (IMBA), Vienna BioCenter (VBC), Vienna, Austria
- Department of Laboratory Medicine, Medical University of Vienna, Vienna, Austria
| | - Shinichiro Sawa
- Division of Mucosal Immunology, Research Center for Systems Immunology, Medical Institute of Bioregulation, Kyushu University, Fukuoka, Japan
| | - Lisa C Osborne
- Department of Microbiology and Immunology, Life Sciences Institute, University of British Columbia, Vancouver, British Columbia, Canada
| | - Philip Rosenstiel
- Institute of Clinical Molecular Biology, Kiel University and University Hospital Schleswig-Holstein, Kiel, Germany
| | - Manolis Pasparakis
- Institute for Genetics and Cologne Excellence Cluster on Cellular Stress Responses in Aging-Associated Diseases (CECAD), University of Cologne, Cologne, Germany
| | - Jürgen Ruland
- Institute of Clinical Chemistry and Pathobiochemistry, School of Medicine and Health, TUM University Hospital, Munich, Germany
- Center for Translational Cancer Research (TranslaTUM), Munich, Germany
| | - Hiroshi Takayanagi
- Department of Immunology, Graduate School of Medicine and Faculty of Medicine, University of Tokyo, Tokyo, Japan
| | - Hans Clevers
- Oncode Institute, Hubrecht Institute, Royal Netherlands Academy of Arts and Sciences (KNAW) and University Medical Center, Utrecht, The Netherlands
- The Princess Maxima Center for Pediatric Oncology, Utrecht, The Netherlands
- Institute of Human Biology (IHB), Roche Pharma Research and Early Development, Roche innovation Centre, Basel, Switzerland
| | - Bon-Kyoung Koo
- Center for Genome Engineering, Institute for Basic Science, Daejeon, Republic of Korea
| | - Josef M Penninger
- Institute of Molecular Biotechnology of the Austrian Academy of Sciences (IMBA), Vienna BioCenter (VBC), Vienna, Austria.
- Department of Laboratory Medicine, Medical University of Vienna, Vienna, Austria.
- Department of Medical Genetics, Life Sciences Institute, University of British Columbia, Vancouver, British Columbia, Canada.
- Helmholtz Centre for Infection Research, Braunschweig, Germany.
| |
Collapse
|
18
|
Parker ME, Mehta NU, Liao TC, Tomaszewski WH, Snyder SA, Busch J, Ciofani M. Restriction of innate Tγδ17 cell plasticity by an AP-1 regulatory axis. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.10.15.618522. [PMID: 39463970 PMCID: PMC11507935 DOI: 10.1101/2024.10.15.618522] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 10/29/2024]
Abstract
IL-17-producing γδ T (Tγδ17) cells are innate-like mediators of intestinal barrier immunity. While Th17 cell and ILC3 plasticity have been extensively studied, the mechanisms governing Tγδ17 cell effector flexibility remain undefined. Here, we combined type 3 fate-mapping with single cell ATAC/RNA-seq multiome profiling to define the cellular features and regulatory networks underlying Tγδ17 cell plasticity. During homeostasis, Tγδ17 cell effector identity was stable across tissues, including for intestinal T-bet+ Tγδ17 cells that restrained IFNγ production. However, S. typhimurium infection induced intestinal Vγ6+ Tγδ17 cell conversion into type 1 effectors, with loss of IL-17A production and partial RORγt downregulation. Multiome analysis revealed a trajectory along Vγ6+ Tγδ17 effector conversion, with TIM-3 marking ex-Tγδ17 cells with enhanced type 1 functionality. Lastly, we characterized and validated a critical AP-1 regulatory axis centered around JunB and Fosl2 that controls Vγ6+ Tγδ17 cell plasticity by stabilizing type 3 identity and restricting type 1 effector conversion.
Collapse
Affiliation(s)
- Morgan E Parker
- Department of Integrative Immunobiology, Duke University Medical Center, Durham, NC, USA
- Center for Advanced Genomic Technologies, Duke University, Durham, NC, USA
| | - Naren U Mehta
- Department of Integrative Immunobiology, Duke University Medical Center, Durham, NC, USA
- Center for Advanced Genomic Technologies, Duke University, Durham, NC, USA
| | - Tzu-Chieh Liao
- Department of Integrative Immunobiology, Duke University Medical Center, Durham, NC, USA
- Center for Advanced Genomic Technologies, Duke University, Durham, NC, USA
| | - William H Tomaszewski
- Department of Integrative Immunobiology, Duke University Medical Center, Durham, NC, USA
| | - Stephanie A Snyder
- Department of Integrative Immunobiology, Duke University Medical Center, Durham, NC, USA
- Center for Advanced Genomic Technologies, Duke University, Durham, NC, USA
| | - Julia Busch
- Department of Integrative Immunobiology, Duke University Medical Center, Durham, NC, USA
- Center for Advanced Genomic Technologies, Duke University, Durham, NC, USA
| | - Maria Ciofani
- Department of Integrative Immunobiology, Duke University Medical Center, Durham, NC, USA
- Center for Advanced Genomic Technologies, Duke University, Durham, NC, USA
- Department of Molecular Genetics and Microbiology, Duke University Medical Center, Durham, NC, USA
| |
Collapse
|
19
|
Biniaris-Georgallis SI, Aschman T, Stergioula K, Schreiber F, Jafari V, Taranko A, Karmalkar T, Kasapi A, Lenac Rovis T, Jelencic V, Bejarano DA, Fabry L, Papacharalampous M, Mattiola I, Molgora M, Hou J, Hublitz KW, Heinrich F, Guerra GM, Durek P, Patone G, Lindberg EL, Maatz H, Hölsken O, Krönke G, Mortha A, Voll RE, Clarke AJ, Hauser AE, Colonna M, Thurley K, Schlitzer A, Schneider C, Stamatiades EG, Mashreghi MF, Jonjic S, Hübner N, Diefenbach A, Kanda M, Triantafyllopoulou A. Amplification of autoimmune organ damage by NKp46-activated ILC1s. Nature 2024; 634:952-960. [PMID: 39137897 DOI: 10.1038/s41586-024-07907-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2022] [Accepted: 08/05/2024] [Indexed: 08/15/2024]
Abstract
In systemic lupus erythematosus, loss of immune tolerance, autoantibody production and immune complex deposition are required but not sufficient for organ damage1. How inflammatory signals are initiated and amplified in the setting of autoimmunity remains elusive. Here we set out to dissect layers and hierarchies of autoimmune kidney inflammation to identify tissue-specific cellular hubs that amplify autoinflammatory responses. Using high-resolution single-cell profiling of kidney immune and parenchymal cells, in combination with antibody blockade and genetic deficiency, we show that tissue-resident NKp46+ innate lymphoid cells (ILCs) are crucial signal amplifiers of disease-associated macrophage expansion and epithelial cell injury in lupus nephritis, downstream of autoantibody production. NKp46 signalling in a distinct subset of group 1 ILCs (ILC1s) instructed an unconventional immune-regulatory transcriptional program, which included the expression of the myeloid cell growth factor CSF2. CSF2 production by NKp46+ ILCs promoted the population expansion of monocyte-derived macrophages. Blockade of the NKp46 receptor (using the antibody clone mNCR1.15; ref. 2) or genetic deficiency of NKp46 abrogated epithelial cell injury. The same cellular and molecular patterns were operative in human lupus nephritis. Our data provide support for the idea that NKp46+ ILC1s promote parenchymal cell injury by granting monocyte-derived macrophages access to epithelial cell niches. NKp46 activation in ILC1s therefore constitutes a previously unrecognized, crucial tissue rheostat that amplifies organ damage in autoimmune hosts, with broad implications for inflammatory pathologies and therapies.
Collapse
Affiliation(s)
- Stylianos-Iason Biniaris-Georgallis
- Department of Rheumatology and Clinical Immunology, Charité-Universitätsmedizin Berlin Campus Mitte, Berlin, Germany
- German Rheumatology Research Center (DRFZ), A Leibniz Institute, Berlin, Germany
- Institute of Microbiology, Infectious Diseases and Immunology (I-MIDI), Charité-Universitätsmedizin Berlin Campus Benjamin Franklin, Berlin, Germany
- Department of Biology, Chemistry and Pharmacy, Free University of Berlin, Berlin, Germany
- Department of Nephrology and Medical Intensive Care, Charité-Universitätsmedizin Berlin Campus Mitte, Berlin, Germany
| | - Tom Aschman
- Department of Rheumatology and Clinical Immunology, Charité-Universitätsmedizin Berlin Campus Mitte, Berlin, Germany
- German Rheumatology Research Center (DRFZ), A Leibniz Institute, Berlin, Germany
- Institute of Microbiology, Infectious Diseases and Immunology (I-MIDI), Charité-Universitätsmedizin Berlin Campus Benjamin Franklin, Berlin, Germany
- Department of Rheumatology and Clinical Immunology, Medical Center -University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
- Department of Neuropathology, Charité-Universitätsmedizin Berlin Campus Mitte, Berlin, Germany
| | - Katerina Stergioula
- Department of Rheumatology and Clinical Immunology, Charité-Universitätsmedizin Berlin Campus Mitte, Berlin, Germany
- German Rheumatology Research Center (DRFZ), A Leibniz Institute, Berlin, Germany
- Institute of Microbiology, Infectious Diseases and Immunology (I-MIDI), Charité-Universitätsmedizin Berlin Campus Benjamin Franklin, Berlin, Germany
- Department of Biology, Chemistry and Pharmacy, Free University of Berlin, Berlin, Germany
| | - Frauke Schreiber
- Department of Rheumatology and Clinical Immunology, Charité-Universitätsmedizin Berlin Campus Mitte, Berlin, Germany
- German Rheumatology Research Center (DRFZ), A Leibniz Institute, Berlin, Germany
- Institute of Microbiology, Infectious Diseases and Immunology (I-MIDI), Charité-Universitätsmedizin Berlin Campus Benjamin Franklin, Berlin, Germany
| | - Vajiheh Jafari
- Department of Rheumatology and Clinical Immunology, Charité-Universitätsmedizin Berlin Campus Mitte, Berlin, Germany
- German Rheumatology Research Center (DRFZ), A Leibniz Institute, Berlin, Germany
- Institute of Microbiology, Infectious Diseases and Immunology (I-MIDI), Charité-Universitätsmedizin Berlin Campus Benjamin Franklin, Berlin, Germany
- Department of Biology, Chemistry and Pharmacy, Free University of Berlin, Berlin, Germany
| | - Anna Taranko
- Department of Rheumatology and Clinical Immunology, Charité-Universitätsmedizin Berlin Campus Mitte, Berlin, Germany
- German Rheumatology Research Center (DRFZ), A Leibniz Institute, Berlin, Germany
- Institute of Microbiology, Infectious Diseases and Immunology (I-MIDI), Charité-Universitätsmedizin Berlin Campus Benjamin Franklin, Berlin, Germany
- Department of Biology, Chemistry and Pharmacy, Free University of Berlin, Berlin, Germany
| | - Tejal Karmalkar
- Department of Rheumatology and Clinical Immunology, Charité-Universitätsmedizin Berlin Campus Mitte, Berlin, Germany
- German Rheumatology Research Center (DRFZ), A Leibniz Institute, Berlin, Germany
- Institute of Microbiology, Infectious Diseases and Immunology (I-MIDI), Charité-Universitätsmedizin Berlin Campus Benjamin Franklin, Berlin, Germany
- Department of Biology, Chemistry and Pharmacy, Free University of Berlin, Berlin, Germany
| | - Ana Kasapi
- Department of Rheumatology and Clinical Immunology, Charité-Universitätsmedizin Berlin Campus Mitte, Berlin, Germany
- German Rheumatology Research Center (DRFZ), A Leibniz Institute, Berlin, Germany
- Institute of Microbiology, Infectious Diseases and Immunology (I-MIDI), Charité-Universitätsmedizin Berlin Campus Benjamin Franklin, Berlin, Germany
| | - Tihana Lenac Rovis
- Center for Proteomics, Faculty of Medicine, University of Rijeka, Rijeka, Croatia
| | - Vedrana Jelencic
- Center for Proteomics, Faculty of Medicine, University of Rijeka, Rijeka, Croatia
| | - David A Bejarano
- Quantitative Systems Biology, Life and Medical Sciences (LIMES) Institute, University of Bonn, Bonn, Germany
| | - Lea Fabry
- Department of Rheumatology and Clinical Immunology, Charité-Universitätsmedizin Berlin Campus Mitte, Berlin, Germany
- German Rheumatology Research Center (DRFZ), A Leibniz Institute, Berlin, Germany
- Institute of Microbiology, Infectious Diseases and Immunology (I-MIDI), Charité-Universitätsmedizin Berlin Campus Benjamin Franklin, Berlin, Germany
| | - Michail Papacharalampous
- Department of Rheumatology and Clinical Immunology, Medical Center -University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Irene Mattiola
- German Rheumatology Research Center (DRFZ), A Leibniz Institute, Berlin, Germany
- Institute of Microbiology, Infectious Diseases and Immunology (I-MIDI), Charité-Universitätsmedizin Berlin Campus Benjamin Franklin, Berlin, Germany
| | - Martina Molgora
- Department of Pathology and Immunology, Washington University School of Medicine, St Louis, MO, USA
- Department of Immunology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL, USA
| | - Jinchao Hou
- Department of Pathology and Immunology, Washington University School of Medicine, St Louis, MO, USA
- Department of Anesthesiology, Children's Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child Health, Hangzhou, China
| | - Karolin W Hublitz
- Institute of Microbiology, Infectious Diseases and Immunology (I-MIDI), Charité-Universitätsmedizin Berlin Campus Benjamin Franklin, Berlin, Germany
| | - Frederik Heinrich
- German Rheumatology Research Center (DRFZ), A Leibniz Institute, Berlin, Germany
| | | | - Pawel Durek
- German Rheumatology Research Center (DRFZ), A Leibniz Institute, Berlin, Germany
| | - Giannino Patone
- Cardiovascular and Metabolic Sciences, Max Delbrück Center for Molecular Medicine in the Helmholtz Association (MDC), Berlin, Germany
| | - Eric L Lindberg
- Cardiovascular and Metabolic Sciences, Max Delbrück Center for Molecular Medicine in the Helmholtz Association (MDC), Berlin, Germany
| | - Henrike Maatz
- Cardiovascular and Metabolic Sciences, Max Delbrück Center for Molecular Medicine in the Helmholtz Association (MDC), Berlin, Germany
- DZHK, German Centre for Cardiovascular Research, Partner Site Berlin, Berlin, Germany
| | - Oliver Hölsken
- German Rheumatology Research Center (DRFZ), A Leibniz Institute, Berlin, Germany
- Institute of Microbiology, Infectious Diseases and Immunology (I-MIDI), Charité-Universitätsmedizin Berlin Campus Benjamin Franklin, Berlin, Germany
- Department of Anesthesiology and Intensive Care Medicine, Charité-Universitätsmedizin Berlin Campus Benjamin Franklin, Berlin, Germany
- Berlin Institute of Health at Charité Universitätsmedizin, Berlin, BIH Academy, Junior Clinician Scientist Program, Berlin, Germany
| | - Gerhard Krönke
- Department of Rheumatology and Clinical Immunology, Charité-Universitätsmedizin Berlin Campus Mitte, Berlin, Germany
- German Rheumatology Research Center (DRFZ), A Leibniz Institute, Berlin, Germany
| | - Arthur Mortha
- Department of Immunology, University of Toronto, Toronto, Ontario, Canada
| | - Reinhard E Voll
- Department of Rheumatology and Clinical Immunology, Medical Center -University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | | | - Anja E Hauser
- Department of Rheumatology and Clinical Immunology, Charité-Universitätsmedizin Berlin Campus Mitte, Berlin, Germany
- German Rheumatology Research Center (DRFZ), A Leibniz Institute, Berlin, Germany
| | - Marco Colonna
- Department of Pathology and Immunology, Washington University School of Medicine, St Louis, MO, USA
| | - Kevin Thurley
- German Rheumatology Research Center (DRFZ), A Leibniz Institute, Berlin, Germany
- Biomathematics Division, Institute of Experimental Oncology, University Hospital Bonn, Bonn, Germany
| | - Andreas Schlitzer
- Quantitative Systems Biology, Life and Medical Sciences (LIMES) Institute, University of Bonn, Bonn, Germany
| | | | - Efstathios G Stamatiades
- Institute of Microbiology, Infectious Diseases and Immunology (I-MIDI), Charité-Universitätsmedizin Berlin Campus Benjamin Franklin, Berlin, Germany
| | - Mir-Farzin Mashreghi
- German Rheumatology Research Center (DRFZ), A Leibniz Institute, Berlin, Germany
- German Center for Child and Adolescent Health (DZKJ), Partner Site Berlin, Berlin, Germany
| | - Stipan Jonjic
- Center for Proteomics, Faculty of Medicine, University of Rijeka, Rijeka, Croatia
| | - Norbert Hübner
- Cardiovascular and Metabolic Sciences, Max Delbrück Center for Molecular Medicine in the Helmholtz Association (MDC), Berlin, Germany
- DZHK, German Centre for Cardiovascular Research, Partner Site Berlin, Berlin, Germany
- Charité-Universitätsmedizin, Berlin, Germany
| | - Andreas Diefenbach
- German Rheumatology Research Center (DRFZ), A Leibniz Institute, Berlin, Germany.
- Institute of Microbiology, Infectious Diseases and Immunology (I-MIDI), Charité-Universitätsmedizin Berlin Campus Benjamin Franklin, Berlin, Germany.
| | - Masatoshi Kanda
- Cardiovascular and Metabolic Sciences, Max Delbrück Center for Molecular Medicine in the Helmholtz Association (MDC), Berlin, Germany.
- Department of Rheumatology and Clinical Immunology, Sapporo Medical University School of Medicine, Sapporo, Hokkaido, Japan.
| | - Antigoni Triantafyllopoulou
- Department of Rheumatology and Clinical Immunology, Charité-Universitätsmedizin Berlin Campus Mitte, Berlin, Germany.
- German Rheumatology Research Center (DRFZ), A Leibniz Institute, Berlin, Germany.
- Institute of Microbiology, Infectious Diseases and Immunology (I-MIDI), Charité-Universitätsmedizin Berlin Campus Benjamin Franklin, Berlin, Germany.
| |
Collapse
|
20
|
Karmoker JR, Bounds SE, Cai J. Aryl hydrocarbon receptor (AhR)-mediated immune responses to degeneration of the retinal pigment epithelium. Biochim Biophys Acta Mol Basis Dis 2024; 1870:167351. [PMID: 39004382 PMCID: PMC11330344 DOI: 10.1016/j.bbadis.2024.167351] [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: 03/07/2024] [Revised: 07/05/2024] [Accepted: 07/08/2024] [Indexed: 07/16/2024]
Abstract
Injuries to the retinal pigment epithelium (RPE) trigger immune responses, orchestrating interactions within the innate and adaptive immune systems in the outer retina and choroid. We previously reported that interleukin 17 (IL-17) is a pivotal signaling molecule originating from choroidal γδ T cells, exerting protective effects by mediating functional connections between the RPE and subretinal microglia. In this current study, we generated mice with aryl hydrocarbon receptor (AhR) knockout specifically in IL-17-producing cells. These animals had deficiency in IL-17 production from γδ T cells, and exhibited increased sensitivity to both acute and chronic insults targeting the RPE. These findings imply that IL-17 plays a crucial role as a signaling cytokine in preserving the homeostasis of the outer retina and choroid.
Collapse
Affiliation(s)
- James Regun Karmoker
- Department of Biochemistry & Physiology, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA.
| | - Sarah E Bounds
- Department of Biochemistry & Physiology, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA.
| | - Jiyang Cai
- Department of Biochemistry & Physiology, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA; Dean McGee Eye Institute, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA.
| |
Collapse
|
21
|
Xing Q, Chang D, Xie S, Zhao X, Zhang H, Wang X, Bai X, Dong C. BCL6 is required for the thymic development of TCRαβ +CD8αα + intraepithelial lymphocyte lineage. Sci Immunol 2024; 9:eadk4348. [PMID: 38335269 DOI: 10.1126/sciimmunol.adk4348] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2023] [Accepted: 12/13/2023] [Indexed: 02/12/2024]
Abstract
TCRαβ+CD8αα+ intraepithelial lymphocytes (CD8αα+ αβ IELs) are a specialized subset of T cells in the gut epithelium that develop from thymic agonist selected IEL precursors (IELps). The molecular mechanisms underlying the selection and differentiation of this T cell type in the thymus are largely unknown. Here, we found that Bcl6 deficiency in αβ T cells resulted in the near absence of CD8αα+ αβ IELs. BCL6 was expressed by approximately 50% of CD8αα+ αβ IELs and by the majority of thymic PD1+ IELps after agonist selection. Bcl6 deficiency blocked early IELp generation in the thymus, and its expression in IELps was induced by thymic TCR signaling in an ERK-dependent manner. As a result of Bcl6 deficiency, the precursors of IELps among CD4+CD8+ double-positive thymocytes exhibited increased apoptosis during agonist selection and impaired IELp differentiation and maturation. Together, our results elucidate BCL6 as a crucial transcription factor during the thymic development of CD8αα+ αβ IELs.
Collapse
Affiliation(s)
- Qi Xing
- Shanghai Immune Therapy Institute, New Cornerstone Science Laboratory, Shanghai Jiao Tong University School of Medicine-affiliated Renji Hospital, Shanghai 200127, China
- Institute for Immunology and School of Medicine, Tsinghua University, Beijing 100084, China
| | - Dehui Chang
- Institute for Immunology and School of Medicine, Tsinghua University, Beijing 100084, China
| | - Shiyuan Xie
- Institute for Advanced Interdisciplinary Studies and Peking University-Tsinghua University-National Institute of Biological Sciences Joint Graduate Program, Peking University, Beijing 100084, China
| | - Xiaohong Zhao
- Institute for Immunology and School of Medicine, Tsinghua University, Beijing 100084, China
| | - Hao Zhang
- Institute for Immunology and School of Medicine, Tsinghua University, Beijing 100084, China
| | - Xiaohu Wang
- Institute for Immunology and School of Medicine, Tsinghua University, Beijing 100084, China
| | - Xue Bai
- Institute for Immunology and School of Medicine, Tsinghua University, Beijing 100084, China
| | - Chen Dong
- Shanghai Immune Therapy Institute, New Cornerstone Science Laboratory, Shanghai Jiao Tong University School of Medicine-affiliated Renji Hospital, Shanghai 200127, China
- Research Unit of Immune Regulation and Immune Diseases of Chinese Academy of Medical Sciences, Shanghai Jiao Tong University School of Medicine-Affiliated Renji Hospital, Shanghai 200127, China
- Westlake University School of Medicine-affiliated Hangzhou First Hospital, Hangzhou 310024, China
| |
Collapse
|
22
|
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
|
23
|
Xing X, Ni X, Wang J, Shi J. Necroptosis in recurrent implantation failure: A bioinformatics analysis of key genes and therapeutic targets. Medicine (Baltimore) 2024; 103:e38907. [PMID: 39058876 PMCID: PMC11272259 DOI: 10.1097/md.0000000000038907] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/30/2023] [Accepted: 06/20/2024] [Indexed: 07/28/2024] Open
Abstract
Recurrent implantation failure (RIF), a major issue in assisted reproductive technology (ART), may be influenced by necroptosis, a form of cell death linked to several diseases. This study was aimed at investigating the involvement of necroptosis in RIF. Using RNA-sequencing data from the Gene Expression Omnibus database, we identified differentially expressed necroptosis-related genes (DENRGs) in RIF patients compared with those in controls. Functional enrichment, protein-protein interaction (PPI) networks, and transcription factor (TF) regulatory networks were analyzed to identify key genes. Immune cell infiltration was analyzed using the single-sample gene set enrichment analysis (ssGSEA) algorithm. Finally, potential therapeutic drugs targeting key genes were explored using a Drug Gene Interaction Database. In total, 20 DENRGs associated with RIF were identified, with a focus on 6 key genes (MLKL, FASLG, XIAP, CASP1, BIRC3, and TLR3) implicated in necroptosis and immune processes. These genes were used to develop a predictive model for RIF, which was validated in 2 datasets. The model and TF network analysis underscored the importance of TLR3. Immune infiltration analysis showed reduced levels of 16 immune cells in RIF patients, highlighting immune system alterations. Several drugs targeting CASP1, such as nivocasan and emricasan, were identified as potential treatments. The study sheds light on the role of necroptosis in RIF, identifying key genes and immune alterations that could serve as biomarkers and therapeutic targets. These findings pave the way for future experimental research and clinical applications targeting necroptosis in RIF treatment.
Collapse
Affiliation(s)
- Xiuye Xing
- Department of Reproductive Medicine, Haidian District Maternal and Child Health Care Hospital, Beijing, China
| | - Xiaoxiao Ni
- Department of Obstetrics, Haidian District Maternal and Child Health Care Hospital, Beijing, China
| | - Jiaojiao Wang
- Department of Obstetrics, Haidian District Maternal and Child Health Care Hospital, Beijing, China
| | - Junmei Shi
- Department of Obstetrics, Haidian District Maternal and Child Health Care Hospital, Beijing, China
| |
Collapse
|
24
|
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
|
25
|
Tan CY, Jiang D, Theriot BS, Rao MV, Surana NK. A commensal-derived sugar protects against metabolic disease. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.06.12.598703. [PMID: 38915674 PMCID: PMC11195190 DOI: 10.1101/2024.06.12.598703] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/26/2024]
Abstract
Obesity is a worsening global epidemic that is regulated by the microbiota through unknown bacterial factors. We discovered a human-derived commensal bacterium, Clostridium immunis , that protects against metabolic disease by secreting a phosphocholine-modified exopolysaccharide. Genetic interruption of the phosphocholine biosynthesis locus ( licABC ) results in a functionally inactive exopolysaccharide, which demonstrates the critical requirement for this phosphocholine moiety. This C. immunis exopolysaccharide acts via group 3 innate lymphoid cells and modulating IL-22 levels, which results in a reduction in serum triglycerides, body weight, and visceral adiposity. Importantly, phosphocholine biosynthesis genes are less abundant in humans with obesity or hypertriglyceridemia, findings that suggest the role of bacterial phosphocholine is conserved across mice and humans. These results define a bacterial molecule-and its key structural motif-that regulates host metabolism. More broadly, they highlight how small molecules, such as phosphocholine, may help fine-tune microbiome- immune-metabolism interactions.
Collapse
|
26
|
Yu H, Tang J, Dong L, Tang M, Arif A, Zhang T, Zhang G, Xie K, Zhao Z, Dai G. Transcriptome analysis reveals that gga-miR-2954 inhibits the inflammatory response against Eimeria tenella infection. Int J Biol Macromol 2024; 269:131807. [PMID: 38670189 DOI: 10.1016/j.ijbiomac.2024.131807] [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: 12/05/2023] [Revised: 01/29/2024] [Accepted: 03/15/2024] [Indexed: 04/28/2024]
Abstract
Coccidiosis is an important parasitic protozoan disease in poultry farming, causing huge economic losses in the global poultry industry every year. MicroRNAs (miRNAs) are a class of RNA macromolecules that play important roles in the immune response to pathogens. However, the expression profiles and functions of miRNAs during Eimeria tenella (E. tenella) infection in chickens remain mostly uncharacterized. In this study, high-throughput sequencing of cecal tissues of control (JC), resistant (JR), and susceptible (JS) chickens led to the identification of 35 differentially expressed miRNAs among the three groups. Functional enrichment analysis showed that the differentially expressed miRNAs were mainly associated with the TGF-beta, NF-kB, and Jak-STAT signaling pathways. Notably, gga-miR-2954 was found to be significantly upregulated after coccidial infection. Functional analysis showed that gga-miR-2954 inhibited the production of the inflammatory cytokines IL-6, IL-1β, TNF-α, and IL-8 in sporozoite-stimulated DF-1 cells. Mechanistically, we found that gga-miR-2954 targeted the RORC gene and that RORC promoted the inflammatory response in sporozoite-stimulated DF-1 cells. In conclusion, our study was the first to identify differentially expressed miRNAs in chicken cecal tissue during E. tenella infection and found that gga-miR-2954 regulates the host immune response to coccidial infection in chickens by targeting the RORC gene.
Collapse
Affiliation(s)
- Hailiang Yu
- College of Animal Science and Technology, Yangzhou University, Yangzhou, China
| | - Jianqiang Tang
- College of Animal Science and Technology, Yangzhou University, Yangzhou, China
| | - Liyue Dong
- College of Animal Science and Technology, Yangzhou University, Yangzhou, China
| | - Meihui Tang
- College of Animal Science and Technology, Yangzhou University, Yangzhou, China
| | - AreeJ Arif
- College of Animal Science and Technology, Yangzhou University, Yangzhou, China
| | - Tao Zhang
- College of Animal Science and Technology, Yangzhou University, Yangzhou, China
| | - Genxi Zhang
- College of Animal Science and Technology, Yangzhou University, Yangzhou, China
| | - Kaizhou Xie
- College of Animal Science and Technology, Yangzhou University, Yangzhou, China
| | - Zhenhua Zhao
- Poultry Institute, Chinese Academy of Agricultural Sciences, Yangzhou, China
| | - Guojun Dai
- College of Animal Science and Technology, Yangzhou University, Yangzhou, China.
| |
Collapse
|
27
|
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
|
28
|
Abstract
The intestinal epithelium, which segregates the highly stimulatory lumen from the underlying tissue, harbors one of the largest lymphocyte populations in the body, intestinal intraepithelial lymphocytes (IELs). IELs must balance tolerance, resistance, and tissue protection to maintain epithelial homeostasis and barrier integrity. This review discusses the ontogeny, environmental imprinting, T cell receptor (TCR) repertoire, and function of intestinal IELs. Despite distinct developmental pathways, IEL subsets share core traits including an epithelium-adapted profile, innate-like properties, cytotoxic potential, and limited TCR diversity. IELs also receive important developmental and functional cues through interactions with epithelial cells, microbiota, and dietary components. The restricted TCR diversity of IELs suggests that a limited set of intestinal antigens drives IEL responses, with potential functional consequences. Finally, IELs play a key role in promoting homeostatic immunity and epithelial barrier integrity but can become pathogenic upon dysregulation. Therefore, IELs represent intriguing but underexamined therapeutic targets for inflammatory diseases and cancer.
Collapse
Affiliation(s)
- Ainsley Lockhart
- Laboratory of Mucosal Immunology, The Rockefeller University, New York, NY, USA; ,
- Current affiliation: Immunology Program, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Daniel Mucida
- Laboratory of Mucosal Immunology, The Rockefeller University, New York, NY, USA; ,
- Howard Hughes Medical Institute, The Rockefeller University, New York, NY, USA
| | - Angelina M Bilate
- Laboratory of Mucosal Immunology, The Rockefeller University, New York, NY, USA; ,
| |
Collapse
|
29
|
Cao S, Fachi JL, Ma K, Ulezko Antonova A, Wang Q, Cai Z, Kaufman RJ, Ciorba MA, Deepak P, Colonna M. The IRE1α/XBP1 pathway sustains cytokine responses of group 3 innate lymphoid cells in inflammatory bowel disease. J Clin Invest 2024; 134:e174198. [PMID: 38722686 PMCID: PMC11214543 DOI: 10.1172/jci174198] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2023] [Accepted: 05/02/2024] [Indexed: 05/14/2024] Open
Abstract
Group 3 innate lymphoid cells (ILC3s) are key players in intestinal homeostasis. ER stress is linked to inflammatory bowel disease (IBD). Here, we used cell culture, mouse models, and human specimens to determine whether ER stress in ILC3s affects IBD pathophysiology. We show that mouse intestinal ILC3s exhibited a 24-hour rhythmic expression pattern of the master ER stress response regulator inositol-requiring kinase 1α/X-box-binding protein 1 (IRE1α/XBP1). Proinflammatory cytokine IL-23 selectively stimulated IRE1α/XBP1 in mouse ILC3s through mitochondrial ROS (mtROS). IRE1α/XBP1 was activated in ILC3s from mice exposed to experimental colitis and in inflamed human IBD specimens. Mice with Ire1α deletion in ILC3s (Ire1αΔRorc) showed reduced expression of the ER stress response and cytokine genes including Il22 in ILC3s and were highly vulnerable to infections and colitis. Administration of IL-22 counteracted their colitis susceptibility. In human ILC3s, IRE1 inhibitors suppressed cytokine production, which was upregulated by an IRE1 activator. Moreover, the frequencies of intestinal XBP1s+ ILC3s in patients with Crohn's disease before administration of ustekinumab, an anti-IL-12/IL-23 antibody, positively correlated with the response to treatment. We demonstrate that a noncanonical mtROS-IRE1α/XBP1 pathway augmented cytokine production by ILC3s and identify XBP1s+ ILC3s as a potential biomarker for predicting the response to anti-IL-23 therapies in IBD.
Collapse
Affiliation(s)
- Siyan Cao
- Division of Gastroenterology, Department of Medicine and
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Jose L. Fachi
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Kaiming Ma
- Division of Gastroenterology, Department of Medicine and
| | - Alina Ulezko Antonova
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Qianli Wang
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Zhangying Cai
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Randal J. Kaufman
- Degenerative Diseases Program, Center for Genetic Disorders and Aging Research, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, California, USA
| | | | | | - Marco Colonna
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, Missouri, USA
| |
Collapse
|
30
|
Hao J, Liu C, Gu Z, Yang X, Lan X, Guo X. Dysregulation of Wnt/β-catenin signaling contributes to intestinal inflammation through regulation of group 3 innate lymphoid cells. Nat Commun 2024; 15:2820. [PMID: 38561332 PMCID: PMC10985070 DOI: 10.1038/s41467-024-45616-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2023] [Accepted: 01/30/2024] [Indexed: 04/04/2024] Open
Abstract
RORγt+ group 3 innate lymphoid cells (ILC3s) are essential for intestinal homeostasis. Dysregulation of ILC3s has been found in the gut of patients with inflammatory bowel disease and colorectal cancer, yet the specific mechanisms still require more investigation. Here we observe increased β-catenin in intestinal ILC3s from inflammatory bowel disease and colon cancer patients compared with healthy donors. In contrast to promoting RORγt expression in T cells, activation of Wnt/β-catenin signaling in ILC3s suppresses RORγt expression, inhibits its proliferation and function, and leads to a deficiency of ILC3s and subsequent intestinal inflammation in mice. Activated β-catenin and its interacting transcription factor, TCF-1, cannot directly suppress RORγt expression, but rather alters global chromatin accessibility and inhibits JunB expression, which is essential for RORγt expression in ILC3s. Together, our findings suggest that dysregulated Wnt/β-catenin signaling impairs intestinal ILC3s through TCF-1/JunB/RORγt regulation, further disrupting intestinal homeostasis, and promoting inflammation and cancer.
Collapse
Affiliation(s)
- Jiacheng Hao
- Institute for Immunology, Tsinghua University, 100084, Beijing, China
- Department of Basic Medical Sciences, School of Medicine, Tsinghua University, 100084, Beijing, China
- School of Life Sciences, Tsinghua University, 100084, Beijing, China
- Beijing Key Lab for Immunological Research on Chronic Diseases, Tsinghua University, 100084, Beijing, China
- Tsinghua-Peking Center for Life Sciences, Tsinghua University, Beijing, China
| | - Chang Liu
- Department of Basic Medical Sciences, School of Medicine, Tsinghua University, 100084, Beijing, China
| | - Zhijie Gu
- Institute for Immunology, Tsinghua University, 100084, Beijing, China
- Department of Basic Medical Sciences, School of Medicine, Tsinghua University, 100084, Beijing, China
- School of Life Sciences, Tsinghua University, 100084, Beijing, China
- Beijing Key Lab for Immunological Research on Chronic Diseases, Tsinghua University, 100084, Beijing, China
- Tsinghua-Peking Center for Life Sciences, Tsinghua University, Beijing, China
| | - Xuanming Yang
- Sheng Yushou Center of Cell Biology and Immunology, School of Life Sciences and Biotechnology, Shanghai Jiao Tong University, 200240, Shanghai, China
- Joint International Research Laboratory of Metabolic and Developmental Sciences, Shanghai Jiao Tong University, 200240, Shanghai, China
- Key Laboratory of Systems Biomedicine (Ministry of Education), Shanghai Center for Systems Biomedicine, Shanghai Jiao Tong University, 200240, Shanghai, China
| | - Xun Lan
- Department of Basic Medical Sciences, School of Medicine, Tsinghua University, 100084, Beijing, China
- Tsinghua-Peking Center for Life Sciences, Tsinghua University, Beijing, China
| | - Xiaohuan Guo
- Institute for Immunology, Tsinghua University, 100084, Beijing, China.
- Department of Basic Medical Sciences, School of Medicine, Tsinghua University, 100084, Beijing, China.
- Beijing Key Lab for Immunological Research on Chronic Diseases, Tsinghua University, 100084, Beijing, China.
| |
Collapse
|
31
|
Naik AK, Dauphars DJ, Corbett E, Simpson L, Schatz DG, Krangel MS. RORγt up-regulates RAG gene expression in DP thymocytes to expand the Tcra repertoire. Sci Immunol 2024; 9:eadh5318. [PMID: 38489350 PMCID: PMC11005092 DOI: 10.1126/sciimmunol.adh5318] [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: 03/09/2023] [Accepted: 02/21/2024] [Indexed: 03/17/2024]
Abstract
Recombination activating gene (RAG) expression increases as thymocytes transition from the CD4-CD8- double-negative (DN) to the CD4+CD8+ double-positive (DP) stage, but the physiological importance and mechanism of transcriptional up-regulation are unknown. Here, we show that a DP-specific component of the recombination activating genes antisilencer (DPASE) provokes elevated RAG expression in DP thymocytes. Mouse DP thymocytes lacking the DPASE display RAG expression equivalent to that in DN thymocytes, but this supports only a partial Tcra repertoire due to inefficient secondary Vα-Jα rearrangement. These data indicate that RAG up-regulation is required for a replete Tcra repertoire and that RAG expression is fine-tuned during lymphocyte development to meet the requirements of distinct antigen receptor loci. We further show that transcription factor RORγt directs RAG up-regulation in DP thymocytes by binding to the DPASE and that RORγt influences the Tcra repertoire by binding to the Tcra enhancer. These data, together with prior work showing RORγt to control Tcra rearrangement by regulating DP thymocyte proliferation and survival, reveal RORγt to orchestrate multiple pathways that support formation of the Tcra repertoire.
Collapse
Affiliation(s)
- Abani Kanta Naik
- Department of Integrative Immunobiology, Duke University School of Medicine, Durham, NC, USA
| | - Danielle J Dauphars
- Department of Integrative Immunobiology, Duke University School of Medicine, Durham, NC, USA
| | - Elizabeth Corbett
- Department of Immunobiology and Department of Molecular Biophysics and Biochemistry, Yale University School of Medicine, New Haven, CT, USA
| | - Lunden Simpson
- Department of Integrative Immunobiology, Duke University School of Medicine, Durham, NC, USA
| | - David G Schatz
- Department of Immunobiology and Department of Molecular Biophysics and Biochemistry, Yale University School of Medicine, New Haven, CT, USA
| | - Michael S Krangel
- Department of Integrative Immunobiology, Duke University School of Medicine, Durham, NC, USA
| |
Collapse
|
32
|
Ohara D, Takeuchi Y, Watanabe H, Lee Y, Mukoyama H, Ohteki T, Kondoh G, Hirota K. Notch2 with retinoic acid license IL-23 expression by intestinal EpCAM+ DCIR2+ cDC2s in mice. J Exp Med 2024; 221:e20230923. [PMID: 38180443 PMCID: PMC10770806 DOI: 10.1084/jem.20230923] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2023] [Revised: 11/06/2023] [Accepted: 12/07/2023] [Indexed: 01/06/2024] Open
Abstract
Despite the importance of IL-23 in mucosal host defense and disease pathogenesis, the mechanisms regulating the development of IL-23-producing mononuclear phagocytes remain poorly understood. Here, we employed an Il23aVenus reporter strain to investigate the developmental identity and functional regulation of IL-23-producing cells. We showed that flagellin stimulation or Citrobacter rodentium infection led to robust induction of IL-23-producing EpCAM+ DCIR2+ CD103- cDC2s, termed cDCIL23, which was confined to gut-associated lymphoid tissues, including the mesenteric lymph nodes, cryptopatches, and isolated lymphoid follicles. Furthermore, we demonstrated that Notch2 signaling was crucial for the development of EpCAM+ DCIR2+ cDC2s, and the combination of Notch2 signaling with retinoic acid signaling controlled their terminal differentiation into cDCIL23, supporting a two-step model for the development of gut cDCIL23. Our findings provide fundamental insights into the developmental pathways and cellular dynamics of IL-23-producing cDC2s at steady state and during pathogen infection.
Collapse
Affiliation(s)
- Daiya Ohara
- Laboratory of Integrative Biological Science, Institute for Life and Medical Sciences, Kyoto University, Kyoto, Japan
| | - Yusuke Takeuchi
- Laboratory of Integrative Biological Science, Institute for Life and Medical Sciences, Kyoto University, Kyoto, Japan
| | - Hitomi Watanabe
- Laboratory of Integrative Biological Science, Institute for Life and Medical Sciences, Kyoto University, Kyoto, Japan
| | - Yoonha Lee
- Laboratory of Integrative Biological Science, Institute for Life and Medical Sciences, Kyoto University, Kyoto, Japan
| | - Hiroki Mukoyama
- Laboratory of Integrative Biological Science, Institute for Life and Medical Sciences, Kyoto University, Kyoto, Japan
| | - Toshiaki Ohteki
- Department of Biodefense Research, Medical Research Institute, Tokyo Medical and Dental University, Tokyo, Japan
| | - Gen Kondoh
- Laboratory of Integrative Biological Science, Institute for Life and Medical Sciences, Kyoto University, Kyoto, Japan
| | - Keiji Hirota
- Laboratory of Integrative Biological Science, Institute for Life and Medical Sciences, Kyoto University, Kyoto, Japan
| |
Collapse
|
33
|
Akama Y, Murao A, Aziz M, Wang P. Extracellular CIRP induces CD4CD8αα intraepithelial lymphocyte cytotoxicity in sepsis. Mol Med 2024; 30:17. [PMID: 38302880 PMCID: PMC10835974 DOI: 10.1186/s10020-024-00790-2] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2023] [Accepted: 01/23/2024] [Indexed: 02/03/2024] Open
Abstract
BACKGROUND In sepsis, intestinal barrier dysfunction is often caused by the uncontrolled death of intestinal epithelial cells (IECs). CD4CD8αα intraepithelial lymphocytes (IELs), a subtype of CD4+ T cells residing within the intestinal epithelium, exert cytotoxicity by producing granzyme B (GrB) and perforin (Prf). Extracellular cold-inducible RNA-binding protein (eCIRP) is a recently identified alarmin which stimulates TLR4 on immune cells to induce proinflammatory responses. Here, we hypothesized that eCIRP enhances CD4CD8αα IEL cytotoxicity and induces IEC death in sepsis. METHODS We subjected wild-type (WT) and CIRP-/- mice to sepsis by cecal ligation and puncture (CLP) and collected the small intestines to isolate IELs. The expression of GrB and Prf in CD4CD8αα IELs was assessed by flow cytometry. IELs isolated from WT and TLR4-/- mice were challenged with recombinant mouse CIRP (eCIRP) and assessed the expression of GrB and Prf in CD4CD8αα by flow cytometry. Organoid-derived IECs were co-cultured with eCIRP-treated CD4CD8αα cells in the presence/absence of GrB and Prf inhibitors and assessed IEC death by flow cytometry. RESULTS We found a significant increase in the expression of GrB and Prf in CD4CD8αα IELs of septic mice compared to sham mice. We found that GrB and Prf levels in CD4CD8αα IELs were increased in the small intestines of WT septic mice, while CD4CD8αα IELs of CIRP-/- mice did not show an increase in those cytotoxic granules after sepsis. We found that eCIRP upregulated GrB and Prf in CD4CD8αα IELs isolated from WT mice but not from TLR4-/- mice. Furthermore, we also revealed that eCIRP-treated CD4CD8αα cells induced organoid-derived IEC death, which was mitigated by GrB and Prf inhibitors. Finally, histological analysis of septic mice revealed that CIRP-/- mice were protected from tissue injury and cell death in the small intestines compared to WT mice. CONCLUSION In sepsis, the cytotoxicity initiated by the eCIRP/TLR4 axis in CD4CD8αα IELs is associated with intestinal epithelial cell (IEC) death, which could lead to gut injury.
Collapse
Affiliation(s)
- Yuichi Akama
- Center for Immunology and Inflammation, The Feinstein Institutes for Medical Research, 350 Community Dr, 11030, Manhasset, NY, USA
| | - Atsushi Murao
- Center for Immunology and Inflammation, The Feinstein Institutes for Medical Research, 350 Community Dr, 11030, Manhasset, NY, USA
| | - Monowar Aziz
- Center for Immunology and Inflammation, The Feinstein Institutes for Medical Research, 350 Community Dr, 11030, Manhasset, NY, USA.
- Departments of Surgery and Molecular Medicine, Zucker School of Medicine at Hofstra/Northwell, Manhasset, NY, USA.
| | - Ping Wang
- Center for Immunology and Inflammation, The Feinstein Institutes for Medical Research, 350 Community Dr, 11030, Manhasset, NY, USA.
- Departments of Surgery and Molecular Medicine, Zucker School of Medicine at Hofstra/Northwell, Manhasset, NY, USA.
| |
Collapse
|
34
|
Abramson J, Dobeš J, Lyu M, Sonnenberg GF. The emerging family of RORγt + antigen-presenting cells. Nat Rev Immunol 2024; 24:64-77. [PMID: 37479834 PMCID: PMC10844842 DOI: 10.1038/s41577-023-00906-5] [Citation(s) in RCA: 22] [Impact Index Per Article: 22.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/12/2023] [Indexed: 07/23/2023]
Abstract
Antigen-presenting cells (APCs) are master regulators of the immune response by directly interacting with T cells to orchestrate distinct functional outcomes. Several types of professional APC exist, including conventional dendritic cells, B cells and macrophages, and numerous other cell types have non-classical roles in antigen presentation, such as thymic epithelial cells, endothelial cells and granulocytes. Accumulating evidence indicates the presence of a new family of APCs marked by the lineage-specifying transcription factor retinoic acid receptor-related orphan receptor-γt (RORγt) and demonstrates that these APCs have key roles in shaping immunity, inflammation and tolerance, particularly in the context of host-microorganism interactions. These RORγt+ APCs include subsets of group 3 innate lymphoid cells, extrathymic autoimmune regulator-expressing cells and, potentially, other emerging populations. Here, we summarize the major findings that led to the discovery of these RORγt+ APCs and their associated functions. We discuss discordance in recent reports and identify gaps in our knowledge in this burgeoning field, which has tremendous potential to advance our understanding of fundamental immune concepts.
Collapse
Affiliation(s)
- Jakub Abramson
- Department of Immunology and Regenerative Biology, Weizmann Institute of Science, Rehovot, Israel.
| | - Jan Dobeš
- Department of Cell Biology, Faculty of Science, Charles University, Prague, Czech Republic
| | - Mengze Lyu
- Jill Roberts Institute for Research in Inflammatory Bowel Disease, Weill Cornell Medicine, Cornell University, New York, NY, USA
- Joan and Sanford I. Weill Department of Medicine, Division of Gastroenterology & Hepatology, Weill Cornell Medicine, Cornell University, New York, NY, USA
- Department of Microbiology & Immunology, Weill Cornell Medicine, Cornell University, New York, NY, USA
| | - Gregory F Sonnenberg
- Jill Roberts Institute for Research in Inflammatory Bowel Disease, Weill Cornell Medicine, Cornell University, New York, NY, USA.
- Joan and Sanford I. Weill Department of Medicine, Division of Gastroenterology & Hepatology, Weill Cornell Medicine, Cornell University, New York, NY, USA.
- Department of Microbiology & Immunology, Weill Cornell Medicine, Cornell University, New York, NY, USA.
| |
Collapse
|
35
|
Pastwińska J, Karwaciak I, Karaś K, Bachorz RA, Ratajewski M. RORγT agonists as immune modulators in anticancer therapy. Biochim Biophys Acta Rev Cancer 2023; 1878:189021. [PMID: 37951483 DOI: 10.1016/j.bbcan.2023.189021] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2023] [Revised: 10/26/2023] [Accepted: 11/04/2023] [Indexed: 11/14/2023]
Abstract
RORγT is a transcription factor that directs the development of Th17 lymphocytes and other IL-17-expressing cells (e.g., Tc17 and ILC3 cells). These cells are involved in the body's defense against pathogenic bacteria and fungi, but they also participate in maintaining the proinflammatory environment in some autoimmune diseases and play a role in the immune system's response to cancer. Similar to other members of the nuclear receptor superfamily, the activity of RORγT is regulated by low-molecular-weight ligands. Therefore, extensive efforts have been dedicated to identifying inverse agonists that diminish the activity of this receptor and subsequently inhibit the development of autoimmune diseases. Unfortunately, in the pursuit of an ideal inverse agonist, the development of agonists has been overlooked. It is important to remember that these types of compounds, by stimulating lymphocytes expressing RORγT (Th17 and Tc17), can enhance the immune system's response to tumors. In this review, we present recent advancements in the biology of RORγT agonists and their potential application in anticancer therapy.
Collapse
Affiliation(s)
- Joanna Pastwińska
- Laboratory of Epigenetics, Institute of Medical Biology, Polish Academy of Sciences, Lodowa 106, 93-232 Lodz, Poland
| | - Iwona Karwaciak
- Laboratory of Epigenetics, Institute of Medical Biology, Polish Academy of Sciences, Lodowa 106, 93-232 Lodz, Poland
| | - Kaja Karaś
- Laboratory of Epigenetics, Institute of Medical Biology, Polish Academy of Sciences, Lodowa 106, 93-232 Lodz, Poland
| | - Rafał A Bachorz
- Laboratory of Molecular Modeling, Institute of Medical Biology, Polish Academy of Sciences, Lodowa 106, 93-232 Lodz, Poland
| | - Marcin Ratajewski
- Laboratory of Epigenetics, Institute of Medical Biology, Polish Academy of Sciences, Lodowa 106, 93-232 Lodz, Poland.
| |
Collapse
|
36
|
Kemter AM, Patry RT, Arnold J, Hesser LA, Campbell E, Ionescu E, Mimee M, Wang S, Nagler CR. Commensal bacteria signal through TLR5 and AhR to improve barrier integrity and prevent allergic responses to food. Cell Rep 2023; 42:113153. [PMID: 37742185 PMCID: PMC10697505 DOI: 10.1016/j.celrep.2023.113153] [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: 11/30/2022] [Revised: 08/07/2023] [Accepted: 09/01/2023] [Indexed: 09/26/2023] Open
Abstract
The increasing prevalence of food allergies has been linked to reduced commensal microbial diversity. In this article, we describe two features of allergy-protective Clostridia that contribute to their beneficial effects. Some Clostridial taxa bear flagella (a ligand for TLR5) and produce indole (a ligand for the aryl hydrocarbon receptor [AhR]). Lysates and flagella from a Clostridia consortium induced interleukin-22 (IL-22) secretion from ileal explants. IL-22 production is abrogated in explants from mice in which TLR5 or MyD88 signaling is deficient either globally or conditionally in CD11c+ antigen-presenting cells. AhR signaling in RORγt+ cells is necessary for the induction of IL-22. Mice deficient in AhR in RORγt+ cells exhibit increased intestinal permeability and are more susceptible to an anaphylactic response to food. Our findings implicate TLR5 and AhR signaling in a molecular mechanism by which commensal Clostridia protect against allergic responses to food.
Collapse
Affiliation(s)
- Andrea M Kemter
- Department of Pathology, The University of Chicago, Chicago, IL 60637, USA
| | - Robert T Patry
- Department of Pathology, The University of Chicago, Chicago, IL 60637, USA
| | - Jack Arnold
- Pritzker School of Molecular Engineering, The University of Chicago, Chicago, IL 60637, USA
| | - Lauren A Hesser
- Pritzker School of Molecular Engineering, The University of Chicago, Chicago, IL 60637, USA
| | - Evelyn Campbell
- Committee on Microbiology, The University of Chicago, Chicago, IL 60637, USA
| | - Edward Ionescu
- Pritzker School of Molecular Engineering, The University of Chicago, Chicago, IL 60637, USA
| | - Mark Mimee
- Department of Microbiology, The University of Chicago, Chicago, IL 60637, USA; Pritzker School of Molecular Engineering, The University of Chicago, Chicago, IL 60637, USA; Committee on Microbiology, The University of Chicago, Chicago, IL 60637, USA
| | - Shan Wang
- Department of Pathology, The University of Chicago, Chicago, IL 60637, USA
| | - Cathryn R Nagler
- Department of Pathology, The University of Chicago, Chicago, IL 60637, USA; Pritzker School of Molecular Engineering, The University of Chicago, Chicago, IL 60637, USA; Committee on Immunology, The University of Chicago, Chicago, IL 60637, USA.
| |
Collapse
|
37
|
Rizk J, Mörbe UM, Agerholm R, Baglioni MV, Catafal Tardos E, Fares da Silva MGF, Ulmert I, Kadekar D, Viñals MT, Bekiaris V. The cIAP ubiquitin ligases sustain type 3 γδ T cells and ILC during aging to promote barrier immunity. J Exp Med 2023; 220:e20221534. [PMID: 37440178 PMCID: PMC10345214 DOI: 10.1084/jem.20221534] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2022] [Revised: 04/10/2023] [Accepted: 06/22/2023] [Indexed: 07/14/2023] Open
Abstract
Early-life cues shape the immune system during adulthood. However, early-life signaling pathways and their temporal functions are not well understood. Herein, we demonstrate that the cellular inhibitor of apoptosis proteins 1 and 2 (cIAP1/2), which are E3 ubiquitin ligases, sustain interleukin (IL)-17-producing γ δ T cells (γδT17) and group 3 innate lymphoid cells (ILC3) during late neonatal and prepubescent life. We show that cell-intrinsic deficiency of cIAP1/2 at 3-4 wk of life leads to downregulation of the transcription factors cMAF and RORγt and failure to enter the cell cycle, followed by progressive loss of γδT17 cells and ILC3 during aging. Mice deficient in cIAP1/2 have severely reduced γδT17 cells and ILC3, present with suboptimal γδT17 responses in the skin, lack intestinal isolated lymphoid follicles, and cannot control intestinal bacterial infection. Mechanistically, these effects appear to be dependent on overt activation of the non-canonical NF-κB pathway. Our data identify cIAP1/2 as early-life molecular switches that establish effective type 3 immunity during aging.
Collapse
Affiliation(s)
- John Rizk
- Department of Health Technology, Technical University of Denmark, Kgs Lyngby, Denmark
- Department of Immunology and Microbiology, LEO Foundation Skin Immunology Research Center, University of Copenhagen, Copenhagen, Denmark
| | - Urs M. Mörbe
- Department of Health Technology, Technical University of Denmark, Kgs Lyngby, Denmark
| | - Rasmus Agerholm
- Department of Health Technology, Technical University of Denmark, Kgs Lyngby, Denmark
| | | | - Elisa Catafal Tardos
- Department of Health Technology, Technical University of Denmark, Kgs Lyngby, Denmark
| | | | - Isabel Ulmert
- Department of Health Technology, Technical University of Denmark, Kgs Lyngby, Denmark
| | - Darshana Kadekar
- Department of Health Technology, Technical University of Denmark, Kgs Lyngby, Denmark
| | | | - Vasileios Bekiaris
- Department of Health Technology, Technical University of Denmark, Kgs Lyngby, Denmark
| |
Collapse
|
38
|
Shen X, Gao X, Luo Y, Xu Q, Fan Y, Hong S, Huang Z, Liu X, Wang Q, Chen Z, Wang D, Lu L, Wu C, Liang H, Wang L. Cxxc finger protein 1 maintains homeostasis and function of intestinal group 3 innate lymphoid cells with aging. NATURE AGING 2023; 3:965-981. [PMID: 37429951 DOI: 10.1038/s43587-023-00453-7] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/25/2022] [Accepted: 06/09/2023] [Indexed: 07/12/2023]
Abstract
Aging is accompanied by homeostatic and functional dysregulation of multiple immune cell subsets. Group 3 innate lymphoid cells (ILC3s) constitute a heterogeneous cell population that plays pivotal roles in intestinal immunity. In this study, we found that ILC3s in aged mice exhibited dysregulated homeostasis and function, leading to bacterial and fungal infection susceptibility. Moreover, our data revealed that the enrichment of the H3K4me3 modification in effector genes of aged gut CCR6+ ILC3s was specifically decreased compared to young mice counterparts. Disruption of Cxxc finger protein 1 (Cxxc1) activity, a key subunit of H3K4 methyltransferase, in ILC3s led to similar aging-related phenotypes. An integrated analysis revealed Kruppel-like factor 4 (Klf4) as a potential Cxxc1 target. Klf4 overexpression partially restored the differentiation and functional defects seen in both aged and Cxxc1-deficient intestinal CCR6+ ILC3s. Therefore, these data suggest that targeting intestinal ILC3s may provide strategies to protect against age-related infections.
Collapse
Affiliation(s)
- Xin Shen
- Institute of Immunology and Bone Marrow Transplantation Center, First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
- Co-Facility Center, Zhejiang University School of Medicine, Hangzhou, China
- Zhejiang University School of Medicine, Hangzhou, China
- Liangzhu Laboratory, Zhejiang University Medical Center, Hangzhou, China
| | - Xianzhi Gao
- Institute of Immunology and Bone Marrow Transplantation Center, First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
- Zhejiang University School of Medicine, Hangzhou, China
- Liangzhu Laboratory, Zhejiang University Medical Center, Hangzhou, China
- Zhejiang University-University of Edinburgh Institute, Zhejiang University School of Medicine, Hangzhou, China
- Edinburgh Medical School: Biomedical Sciences, College of Medicine and Veterinary Medicine, University of Edinburgh, Edinburgh, UK
| | - Yikai Luo
- Department of Bioinformatics and Computational Biology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
- Program of Quantitative and Computational Biosciences, Baylor College of Medicine, Houston, TX, USA
| | - Qianying Xu
- Zhejiang University School of Medicine, Hangzhou, China
| | - Ying Fan
- Laboratory Animal Center, Zhejiang University, Hangzhou, China
| | - Shenghui Hong
- Laboratory Animal Center, Zhejiang University, Hangzhou, China
| | | | - Xiaoqian Liu
- Institute of Immunology and Bone Marrow Transplantation Center, First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
- Zhejiang University School of Medicine, Hangzhou, China
| | - Qianqian Wang
- Laboratory Animal Center, Zhejiang University, Hangzhou, China
| | - Zuojia Chen
- Experimental Immunology Branch, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Di Wang
- Zhejiang University School of Medicine, Hangzhou, China
| | - Linrong Lu
- Zhejiang University School of Medicine, Hangzhou, China
| | - Chuan Wu
- Experimental Immunology Branch, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA.
| | - Han Liang
- Department of Bioinformatics and Computational Biology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA.
- Program of Quantitative and Computational Biosciences, Baylor College of Medicine, Houston, TX, USA.
- Department of Systems Biology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA.
| | - Lie Wang
- Institute of Immunology and Bone Marrow Transplantation Center, First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China.
- Zhejiang University School of Medicine, Hangzhou, China.
- Liangzhu Laboratory, Zhejiang University Medical Center, Hangzhou, China.
- Zhejiang University-University of Edinburgh Institute, Zhejiang University School of Medicine, Hangzhou, China.
- Laboratory Animal Center, Zhejiang University, Hangzhou, China.
- Future Health Laboratory, Innovation Center of Yangtze River Delta, Zhejiang University, Jiaxing, China.
| |
Collapse
|
39
|
Gao KM, Chiang K, Korkmaz FT, Janardhan HP, Trivedi CM, Quinton LJ, Gingras S, Fitzgerald KA, Marshak-Rothstein A. Expression of a STING Gain-of-function Mutation in Endothelial Cells Initiates Lymphocytic Infiltration of the Lungs. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.07.27.550897. [PMID: 37547024 PMCID: PMC10402179 DOI: 10.1101/2023.07.27.550897] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/08/2023]
Abstract
Patients afflicted with STING gain-of-function mutations frequently present with debilitating interstitial lung disease ( ILD ) that is recapitulated in mice expressing the STING V154M mutation ( VM ). Prior radiation chimera studies revealed an unexpected and critical role for non-hematopoietic cells in the initiation of ILD. To identify STING-expressing non-hematopoietic cell types relevant to ILD, we generated a conditional knock-in ( CKI ) model in which expression of the VM allele was directed to hematopoietic cells, fibroblasts, epithelial cells, or endothelial cells. Only endothelial cell-targeted expression of the mutant allele resulted in the recruitment of immune cells to the lung and the formation of bronchus-associated lymphoid tissue, as seen in the parental VM strain. These findings reveal the importance of endothelial cells as instigators of STING-driven lung disease and suggest that therapeutic targeting of STING inhibitors to endothelial cells could potentially mitigate inflammation in the lungs of SAVI patients or patients afflicted with other ILD-related disorders. Summary Patients with STING gain-of-function (GOF) mutations develop life-threatening lung autoinflammation. In this study, Gao et al. utilize a mouse model of conditional STING GOF to demonstrate a role for endothelial STING GOF in initiating immune cell recruitment into lung tissues of SAVI mice.
Collapse
|
40
|
Horodincu L, Solcan C. Influence of Different Light Spectra on Melatonin Synthesis by the Pineal Gland and Influence on the Immune System in Chickens. Animals (Basel) 2023; 13:2095. [PMID: 37443893 DOI: 10.3390/ani13132095] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2023] [Revised: 06/12/2023] [Accepted: 06/21/2023] [Indexed: 07/15/2023] Open
Abstract
It is well known that the pineal gland in birds influences behavioural and physiological functions, including those of the immune system. The purpose of this research is to examine the endocrine-immune correlations between melatonin and immune system activity. Through a description of the immune-pineal axis, we formulated the objective to determine and describe: the development of the pineal gland; how light influences secretory activity; and how melatonin influences the activity of primary and secondary lymphoid organs. The pineal gland has the ability to turn light information into an endocrine signal suitable for the immune system via the membrane receptors Mel1a, Mel1b, and Mel1c, as well as the nuclear receptors RORα, RORβ, and RORγ. We can state the following findings: green monochromatic light (560 nm) increased serum melatonin levels and promoted a stronger humoral and cellular immune response by proliferating B and T lymphocytes; the combination of green and blue monochromatic light (560-480 nm) ameliorated the inflammatory response and protected lymphoid organs from oxidative stress; and red monochromatic light (660 nm) maintained the inflammatory response and promoted the growth of pathogenic bacteria. Melatonin can be considered a potent antioxidant and immunomodulator and is a critical element in the coordination between external light stimulation and the body's internal response.
Collapse
Affiliation(s)
- Loredana Horodincu
- Preclinics Department, Faculty of Veterinary Medicine, "Ion Ionescu de la Brad" Iasi University of Life Sciences, Mihail Sadoveanu Alley, 700489 Iasi, Romania
| | - Carmen Solcan
- Preclinics Department, Faculty of Veterinary Medicine, "Ion Ionescu de la Brad" Iasi University of Life Sciences, Mihail Sadoveanu Alley, 700489 Iasi, Romania
| |
Collapse
|
41
|
Korchagina AA, Shein SA, Koroleva E, Tumanov AV. Transcriptional control of ILC identity. Front Immunol 2023; 14:1146077. [PMID: 36969171 PMCID: PMC10033543 DOI: 10.3389/fimmu.2023.1146077] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2023] [Accepted: 02/21/2023] [Indexed: 03/12/2023] Open
Abstract
Innate lymphoid cells (ILCs) are heterogeneous innate immune cells which participate in host defense, mucosal repair and immunopathology by producing effector cytokines similarly to their adaptive immune cell counterparts. The development of ILC1, 2, and 3 subsets is controlled by core transcription factors: T-bet, GATA3, and RORγt, respectively. ILCs can undergo plasticity and transdifferentiate to other ILC subsets in response to invading pathogens and changes in local tissue environment. Accumulating evidence suggests that the plasticity and the maintenance of ILC identity is controlled by a balance between these and additional transcription factors such as STATs, Batf, Ikaros, Runx3, c-Maf, Bcl11b, and Zbtb46, activated in response to lineage-guiding cytokines. However, how interplay between these transcription factors leads to ILC plasticity and the maintenance of ILC identity remains hypothetical. In this review, we discuss recent advances in understanding transcriptional regulation of ILCs in homeostatic and inflammatory conditions.
Collapse
|
42
|
Teghanemt A, Misel-Wuchter K, Heath J, Thurman A, Pulipati P, Dixit G, Geesala R, Meyerholz DK, Maretzky T, Pezzulo A, Issuree PD. DNA demethylation fine-tunes IL-2 production during thymic regulatory T cell differentiation. EMBO Rep 2023; 24:e55543. [PMID: 36880575 PMCID: PMC10157375 DOI: 10.15252/embr.202255543] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2022] [Revised: 02/10/2023] [Accepted: 02/16/2023] [Indexed: 03/08/2023] Open
Abstract
Regulatory T (T reg) cells developing in the thymus are essential to maintain tolerance and prevent fatal autoimmunity in mice and humans. Expression of the T reg lineage-defining transcription factor FoxP3 is critically dependent upon T cell receptor (TCR) and interleukin-2 (IL-2) signaling. Here, we report that ten-eleven translocation (Tet) enzymes, which are DNA demethylases, are required early during double-positive (DP) thymic T cell differentiation and prior to the upregulation of FoxP3 in CD4 single-positive (SP) thymocytes, to promote Treg differentiation. We show that Tet3 selectively controls the development of CD25- FoxP3lo CD4SP Treg cell precursors in the thymus and is critical for TCR-dependent IL-2 production, which drive chromatin remodeling at the FoxP3 locus as well as other Treg-effector gene loci in an autocrine/paracrine manner. Together, our results demonstrate a novel role for DNA demethylation in regulating the TCR response and promoting Treg cell differentiation. These findings highlight a novel epigenetic pathway to promote the generation of endogenous Treg cells for mitigation of autoimmune responses.
Collapse
Affiliation(s)
- Athmane Teghanemt
- Department of Internal Medicine, Roy J. and Lucille A. Carver College of Medicine, University of Iowa, Iowa City, IA, USA.,Inflammation Program, Roy J. and Lucille A. Carver College of Medicine, University of Iowa, Iowa City, IA, USA
| | - Kara Misel-Wuchter
- Inflammation Program, Roy J. and Lucille A. Carver College of Medicine, University of Iowa, Iowa City, IA, USA.,Molecular Medicine Graduate Program, Iowa City, IA, USA
| | - Jace Heath
- Inflammation Program, Roy J. and Lucille A. Carver College of Medicine, University of Iowa, Iowa City, IA, USA.,Immunology Graduate Program, Iowa City, IA, USA
| | - Andrew Thurman
- Department of Internal Medicine, Roy J. and Lucille A. Carver College of Medicine, University of Iowa, Iowa City, IA, USA
| | - Priyanjali Pulipati
- Department of Internal Medicine, Roy J. and Lucille A. Carver College of Medicine, University of Iowa, Iowa City, IA, USA.,Inflammation Program, Roy J. and Lucille A. Carver College of Medicine, University of Iowa, Iowa City, IA, USA
| | - Garima Dixit
- Department of Internal Medicine, Roy J. and Lucille A. Carver College of Medicine, University of Iowa, Iowa City, IA, USA.,Inflammation Program, Roy J. and Lucille A. Carver College of Medicine, University of Iowa, Iowa City, IA, USA
| | - Ramasatya Geesala
- Department of Internal Medicine, Roy J. and Lucille A. Carver College of Medicine, University of Iowa, Iowa City, IA, USA.,Inflammation Program, Roy J. and Lucille A. Carver College of Medicine, University of Iowa, Iowa City, IA, USA
| | | | - Thorsten Maretzky
- Department of Internal Medicine, Roy J. and Lucille A. Carver College of Medicine, University of Iowa, Iowa City, IA, USA.,Inflammation Program, Roy J. and Lucille A. Carver College of Medicine, University of Iowa, Iowa City, IA, USA
| | - Alejandro Pezzulo
- Department of Internal Medicine, Roy J. and Lucille A. Carver College of Medicine, University of Iowa, Iowa City, IA, USA
| | - Priya D Issuree
- Department of Internal Medicine, Roy J. and Lucille A. Carver College of Medicine, University of Iowa, Iowa City, IA, USA.,Inflammation Program, Roy J. and Lucille A. Carver College of Medicine, University of Iowa, Iowa City, IA, USA.,Molecular Medicine Graduate Program, Iowa City, IA, USA.,Immunology Graduate Program, Iowa City, IA, USA
| |
Collapse
|
43
|
Glaser KM, Tarrant TK, Lämmermann T. Combinatorial depletions of G-protein coupled receptor kinases in immune cells identify pleiotropic and cell type-specific functions. Front Immunol 2022; 13:1039803. [DOI: 10.3389/fimmu.2022.1039803] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2022] [Accepted: 11/01/2022] [Indexed: 11/16/2022] Open
Abstract
G-protein coupled receptor kinases (GRKs) participate in the regulation of chemokine receptors by mediating receptor desensitization. They can be recruited to agonist-activated G-protein coupled receptors (GPCRs) and phosphorylate their intracellular parts, which eventually blocks signal propagation and often induces receptor internalization. However, there is growing evidence that GRKs can also control cellular functions beyond GPCR regulation. Immune cells commonly express two to four members of the GRK family (GRK2, GRK3, GRK5, GRK6) simultaneously, but we have very limited knowledge about their interplay in primary immune cells. In particular, we are missing comprehensive studies comparing the role of this GRK interplay for (a) multiple GPCRs within one leukocyte type, and (b) one specific GPCR between several immune cell subsets. To address this issue, we generated mouse models of single, combinatorial and complete GRK knockouts in four primary immune cell types (neutrophils, T cells, B cells and dendritic cells) and systematically addressed the functional consequences on GPCR-controlled cell migration and tissue localization. Our study shows that combinatorial depletions of GRKs have pleiotropic and cell-type specific effects in leukocytes, many of which could not be predicted. Neutrophils lacking all four GRK family members show increased chemotactic migration responses to a wide range of GPCR ligands, whereas combinatorial GRK depletions in other immune cell types lead to pro- and anti-migratory responses. Combined depletion of GRK2 and GRK6 in T cells and B cells shows distinct functional outcomes for (a) one GPCR type in different cell types, and (b) different GPCRs in one cell type. These GPCR-type and cell-type specific effects reflect in altered lymphocyte chemotaxis in vitro and localization in vivo. Lastly, we provide evidence that complete GRK deficiency impairs dendritic cell homeostasis, which unexpectedly results from defective dendritic cell differentiation and maturation in vitro and in vivo. Together, our findings demonstrate the complexity of GRK functions in immune cells, which go beyond GPCR desensitization in specific leukocyte types. Furthermore, they highlight the need for studying GRK functions in primary immune cells to address their specific roles in each leukocyte subset.
Collapse
|
44
|
Hall JA, Pokrovskii M, Kroehling L, Kim BR, Kim SY, Wu L, Lee JY, Littman DR. Transcription factor RORα enforces stability of the Th17 cell effector program by binding to a Rorc cis-regulatory element. Immunity 2022; 55:2027-2043.e9. [PMID: 36243007 DOI: 10.1016/j.immuni.2022.09.013] [Citation(s) in RCA: 40] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2022] [Revised: 08/05/2022] [Accepted: 09/20/2022] [Indexed: 11/05/2022]
Abstract
T helper 17 (Th17) cells regulate mucosal barrier defenses but also promote multiple autoinflammatory diseases. Although many molecular determinants of Th17 cell differentiation have been elucidated, the transcriptional programs that sustain Th17 cells in vivo remain obscure. The transcription factor RORγt is critical for Th17 cell differentiation; however, it is not clear whether the closely related RORα, which is co-expressed in Th17 cells, has a distinct role. Here, we demonstrated that although dispensable for Th17 cell differentiation, RORα was necessary for optimal Th17 responses in peripheral tissues. The absence of RORα in T cells led to reductions in both RORγt expression and effector function among Th17 cells. Cooperative binding of RORα and RORγt to a previously unidentified Rorc cis-regulatory element was essential for Th17 lineage maintenance in vivo. These data point to a non-redundant role of RORα in Th17 lineage maintenance via reinforcement of the RORγt transcriptional program.
Collapse
Affiliation(s)
- Jason A Hall
- The Kimmel Center for Biology and Medicine of the Skirball Institute, New York University School of Medicine, New York, NY 10016, USA
| | - Maria Pokrovskii
- The Kimmel Center for Biology and Medicine of the Skirball Institute, New York University School of Medicine, New York, NY 10016, USA
| | - Lina Kroehling
- The Kimmel Center for Biology and Medicine of the Skirball Institute, New York University School of Medicine, New York, NY 10016, USA
| | - Bo-Ram Kim
- Department of Microbiology and Immunology, Yonsei University College of Medicine, Seoul 03722, Republic of Korea
| | - Seung Yong Kim
- Department of Microbiology and Immunology, Yonsei University College of Medicine, Seoul 03722, Republic of Korea; Brain Korea 21 PLUS Project for Medical Sciences, Yonsei University College of Medicine, Seoul 03722, Republic of Korea
| | - Lin Wu
- The Kimmel Center for Biology and Medicine of the Skirball Institute, New York University School of Medicine, New York, NY 10016, USA
| | - June-Yong Lee
- The Kimmel Center for Biology and Medicine of the Skirball Institute, New York University School of Medicine, New York, NY 10016, USA; Department of Microbiology and Immunology, Yonsei University College of Medicine, Seoul 03722, Republic of Korea; Brain Korea 21 PLUS Project for Medical Sciences, Yonsei University College of Medicine, Seoul 03722, Republic of Korea; Institute for Immunology and Immunological Diseases, Yonsei University College of Medicine, Seoul 03722, Republic of Korea.
| | - Dan R Littman
- The Kimmel Center for Biology and Medicine of the Skirball Institute, New York University School of Medicine, New York, NY 10016, USA; Howard Hughes Medical Institute, New York, NY 10016, USA.
| |
Collapse
|
45
|
Peng V, Cao S, Trsan T, Bando JK, Avila-Pacheco J, Cleveland JL, Clish C, Xavier RJ, Colonna M. Ornithine decarboxylase supports ILC3 responses in infectious and autoimmune colitis through positive regulation of IL-22 transcription. Proc Natl Acad Sci U S A 2022; 119:e2214900119. [PMID: 36279426 PMCID: PMC9659397 DOI: 10.1073/pnas.2214900119] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2022] [Accepted: 09/27/2022] [Indexed: 01/14/2023] Open
Abstract
Group 3 innate lymphoid cells (ILC3s) are RORγT+ lymphocytes that are predominately enriched in mucosal tissues and produce IL-22 and IL-17A. They are the innate counterparts of Th17 cells. While Th17 lymphocytes utilize unique metabolic pathways in their differentiation program, it is unknown whether ILC3s make similar metabolic adaptations. We employed single-cell RNA sequencing and metabolomic profiling of intestinal ILC subsets to identify an enrichment of polyamine biosynthesis in ILC3s, converging on the rate-limiting enzyme ornithine decarboxylase (ODC1). In vitro and in vivo studies demonstrated that exogenous supplementation with the polyamine putrescine or its biosynthetic substrate, ornithine, enhanced ILC3 production of IL-22. Conditional deletion of ODC1 in ILC3s impaired mouse antibacterial defense against Citrobacter rodentium infection, which was associated with a decrease in anti-microbial peptide production by the intestinal epithelium. Furthermore, in a model of anti-CD40 colitis, deficiency of ODC1 in ILC3s markedly reduced the production of IL-22 and severity of inflammatory colitis. We conclude that ILC3-intrinsic polyamine biosynthesis facilitates efficient defense against enteric pathogens as well as exacerbates autoimmune colitis, thus representing an attractive target to modulate ILC3 function in intestinal disease.
Collapse
Affiliation(s)
- Vincent Peng
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO 63110
| | - Siyan Cao
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO 63110
- Division of Gastroenterology, Washington University School of Medicine, St. Louis, MO 63110
| | - Tihana Trsan
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO 63110
| | - Jennifer K. Bando
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO 63110
| | | | - John L. Cleveland
- Department of Tumor Biology, H. Lee Moffitt Cancer Center & Research Institute, Tampa, FL 33612
| | | | - Ramnik J. Xavier
- Broad Institute, Cambridge, MA 02412
- Center for Computational and Integrative Biology and Department of Molecular Biology, Massachusetts General Hospital and Harvard Medical School, Cambridge Street, Boston, MA 02114
| | - Marco Colonna
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO 63110
| |
Collapse
|
46
|
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: 141] [Impact Index Per Article: 47.0] [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
|
47
|
Zhou W, Zhou L, Zhou J, Chu C, Zhang C, Sockolow RE, Eberl G, Sonnenberg GF. ZBTB46 defines and regulates ILC3s that protect the intestine. Nature 2022; 609:159-165. [PMID: 35831503 PMCID: PMC9528687 DOI: 10.1038/s41586-022-04934-4] [Citation(s) in RCA: 30] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2021] [Accepted: 06/06/2022] [Indexed: 12/28/2022]
Abstract
RORγt is a lineage-specifying transcription factor that is expressed by immune cells that are enriched in the gastrointestinal tract and promote immunity, inflammation and tissue homeostasis1-15. However, fundamental questions remain with regard to the cellular heterogeneity among these cell types, the mechanisms that control protective versus inflammatory properties and their functional redundancy. Here we define all RORγt+ immune cells in the intestine at single-cell resolution and identify a subset of group 3 innate lymphoid cells (ILC3s) that expresses ZBTB46, a transcription factor specifying conventional dendritic cells16-20. ZBTB46 is robustly expressed by CCR6+ lymphoid-tissue-inducer-like ILC3s that are developmentally and phenotypically distinct from conventional dendritic cells, and its expression is imprinted by RORγt, fine-tuned by microbiota-derived signals and increased by pro-inflammatory cytokines. ZBTB46 restrains the inflammatory properties of ILC3s, including the OX40L-dependent expansion of T helper 17 cells and the exacerbated intestinal inflammation that occurs after enteric infection. Finally, ZBTB46+ ILC3s are a major source of IL-22, and selective depletion of this population renders mice susceptible to enteric infection and associated intestinal inflammation. These results show that ZBTB46 is a transcription factor that is shared between conventional dendritic cells and ILC3s, and identify a cell-intrinsic function for ZBTB46 in restraining the pro-inflammatory properties of ILC3s and a non-redundant role for ZBTB46+ ILC3s in orchestrating intestinal health.
Collapse
Affiliation(s)
- Wenqing Zhou
- Joan and Sanford I. Weill Department of Medicine, Division of Gastroenterology & 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 & 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 & 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
| | - Coco Chu
- Jill Roberts Institute for Research in Inflammatory Bowel Disease, Weill Cornell Medicine, Cornell University, New York, NY, USA
| | - Chao Zhang
- Department of Medicine, Division of Computational Biomedicine, Boston University, Boston, MA, USA
| | - Robbyn E Sockolow
- Department of Pediatrics, Division of Gastroenterology and Nutrition, Weill Cornell Medicine, Cornell University, New York, NY, USA
| | - Gerard Eberl
- Microenvironment and Immunity Unit, Institut Pasteur, Paris, France
| | - Gregory F Sonnenberg
- Joan and Sanford I. Weill Department of Medicine, Division of Gastroenterology & 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
|
48
|
Extrathymic expression of Aire controls the induction of effective T H17 cell-mediated immune response to Candida albicans. Nat Immunol 2022; 23:1098-1108. [PMID: 35761088 DOI: 10.1038/s41590-022-01247-6] [Citation(s) in RCA: 38] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2019] [Accepted: 05/18/2022] [Indexed: 01/06/2023]
Abstract
Patients with loss of function in the gene encoding the master regulator of central tolerance AIRE suffer from a devastating disorder called autoimmune polyendocrine syndrome type 1 (APS-1), characterized by a spectrum of autoimmune diseases and severe mucocutaneous candidiasis. Although the key mechanisms underlying the development of autoimmunity in patients with APS-1 are well established, the underlying cause of the increased susceptibility to Candida albicans infection remains less understood. Here, we show that Aire+MHCII+ type 3 innate lymphoid cells (ILC3s) could sense, internalize and present C. albicans and had a critical role in the induction of Candida-specific T helper 17 (TH17) cell clones. Extrathymic Rorc-Cre-mediated deletion of Aire resulted in impaired generation of Candida-specific TH17 cells and subsequent overgrowth of C. albicans in the mucosal tissues. Collectively, our observations identify a previously unrecognized regulatory mechanism for effective defense responses against fungal infections.
Collapse
|
49
|
Programme of self-reactive innate-like T cell-mediated cancer immunity. Nature 2022; 605:139-145. [PMID: 35444279 DOI: 10.1038/s41586-022-04632-1] [Citation(s) in RCA: 60] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2019] [Accepted: 03/10/2022] [Indexed: 12/12/2022]
Abstract
Cellular transformation induces phenotypically diverse populations of tumour-infiltrating T cells1-5, and immune checkpoint blockade therapies preferentially target T cells that recognize cancer cell neoantigens6,7. Yet, how other classes of tumour-infiltrating T cells contribute to cancer immunosurveillance remains elusive. Here, in a survey of T cells in mouse and human malignancies, we identified a population of αβ T cell receptor (TCR)-positive FCER1G-expressing innate-like T cells with high cytotoxic potential8 (ILTCKs). These cells were broadly reactive to unmutated self-antigens, arose from distinct thymic progenitors following early encounter with cognate antigens, and were continuously replenished by thymic progenitors during tumour progression. Notably, expansion and effector differentiation of intratumoural ILTCKs depended on interleukin-15 (IL-15) expression in cancer cells, and inducible activation of IL-15 signalling in adoptively transferred ILTCK progenitors suppressed tumour growth. Thus, the antigen receptor self-reactivity, unique ontogeny, and distinct cancer cell-sensing mechanism distinguish ILTCKs from conventional cytotoxic T cells, and define a new class of tumour-elicited immune response.
Collapse
|
50
|
Bolivar-Wagers S, Larson JH, Jin S, Blazar BR. Cytolytic CD4 + and CD8 + Regulatory T-Cells and Implications for Developing Immunotherapies to Combat Graft-Versus-Host Disease. Front Immunol 2022; 13:864748. [PMID: 35493508 PMCID: PMC9040077 DOI: 10.3389/fimmu.2022.864748] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2022] [Accepted: 03/16/2022] [Indexed: 02/03/2023] Open
Abstract
Regulatory T-cells (Treg) are critical for the maintenance of immune homeostasis and tolerance induction. While the immunosuppressive mechanisms of Treg have been extensively investigated for decades, the mechanisms responsible for Treg cytotoxicity and their therapeutic potential in regulating immune responses have been incompletely explored and exploited. Conventional cytotoxic T effector cells (Teffs) are known to be important for adaptive immune responses, particularly in the settings of viral infections and cancer. CD4+ and CD8+ Treg subsets may also share similar cytotoxic properties with conventional Teffs. Cytotoxic effector Treg (cyTreg) are a heterogeneous population in the periphery that retain the capacity to suppress T-cell proliferation and activation, induce cellular apoptosis, and migrate to tissues to ensure immune homeostasis. The latter can occur through several cytolytic mechanisms, including the Granzyme/Perforin and Fas/FasL signaling pathways. This review focuses on the current knowledge and recent advances in our understanding of cyTreg and their potential application in the treatment of human disease, particularly Graft-versus-Host Disease (GVHD).
Collapse
Affiliation(s)
| | | | | | - Bruce R. Blazar
- Department of Pediatrics, Division of Blood & Marrow Transplant & Cellular Therapy, University of Minnesota, Minneapolis, MN, United States
| |
Collapse
|