1
|
Tafesse Y, Köhler A, Sanchez Sanchez G, Rodrigues PB, Verce M, Vitsos P, Verdebout I, Rezwani M, Papadopoulou M, Everard A, Flamand V, Vermijlen D. Maternal Administration of Probiotics Augments IL17-Committed γδ T Cells in the Newborn Lung. Eur J Immunol 2025; 55:e202451051. [PMID: 40259457 DOI: 10.1002/eji.202451051] [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: 09/27/2024] [Revised: 03/25/2025] [Accepted: 03/27/2025] [Indexed: 04/23/2025]
Abstract
The early life period is increasingly being recognized as a window of opportunity to shape immunity, where microbiota and related probiotics have an important impact. Innate γδ T cells are the first T cells generated in utero, populating epithelial tissues such as the lung and contributing to tissue protection through, for example, IL17 production. Here, we studied the influence of maternal microbiota and probiotic supplementation during pregnancy on innate γδ T cells in the lung and thymus of newborn mice. Detailed time-kinetic experiments showed that at birth, the murine lung T cell population was specifically dominated by IL17-committed γδ T cells expressing an invariant Vγ6Vδ1 TCR. Single-cell RNA-sequencing showed that the biased IL17-commitment of perinatal lung γδT cells is highly conserved between mice and humans. While maternal microbiota depletion with antibiotics tended to decrease the frequency of the lung Vγ6 T cells of the offspring at birth, the maternal administration of Lacticaseibacillus rhamnosus (L.rhm.), but not of Bifidobacterium animalis subsp. lactis (B.lac.), increased significantly their frequency, resulting in the augmentation of the IL17-commitment of the mouse lung T cell compartment. Altogether, our data indicate that the maternal microbiota contributes to the shaping of IL17-committed γδT cells in the lungs of newborns and that maternal administration of specific probiotic strains can enhance this process.
Collapse
Affiliation(s)
- Yohannes Tafesse
- Department of Pharmacotherapy and Pharmaceutics, Université Libre de Bruxelles (ULB), Brussels, Belgium
- Institute for Medical Immunology (IMI), Université Libre de Bruxelles (ULB), Gosselies, Belgium
- ULB Center for Research in Immunology (U-CRI), Belgium
- WELBIO Department, WEL Research Institute, Wavre, Belgium
| | - Arnaud Köhler
- Institute for Medical Immunology (IMI), Université Libre de Bruxelles (ULB), Gosselies, Belgium
- ULB Center for Research in Immunology (U-CRI), Belgium
| | - Guillem Sanchez Sanchez
- Department of Pharmacotherapy and Pharmaceutics, Université Libre de Bruxelles (ULB), Brussels, Belgium
- Institute for Medical Immunology (IMI), Université Libre de Bruxelles (ULB), Gosselies, Belgium
- ULB Center for Research in Immunology (U-CRI), Belgium
- WELBIO Department, WEL Research Institute, Wavre, Belgium
| | - Patricia Brito Rodrigues
- WELBIO Department, WEL Research Institute, Wavre, Belgium
- Metabolism and Nutrition Research Group, Louvain Drug Research Institute, Université Catholique De Louvain, Brussels, Belgium
| | - Marko Verce
- WELBIO Department, WEL Research Institute, Wavre, Belgium
- Metabolism and Nutrition Research Group, Louvain Drug Research Institute, Université Catholique De Louvain, Brussels, Belgium
| | - Panagiotis Vitsos
- Department of Pharmacotherapy and Pharmaceutics, Université Libre de Bruxelles (ULB), Brussels, Belgium
- Institute for Medical Immunology (IMI), Université Libre de Bruxelles (ULB), Gosselies, Belgium
- ULB Center for Research in Immunology (U-CRI), Belgium
- WELBIO Department, WEL Research Institute, Wavre, Belgium
| | - Isoline Verdebout
- Department of Pharmacotherapy and Pharmaceutics, Université Libre de Bruxelles (ULB), Brussels, Belgium
- Institute for Medical Immunology (IMI), Université Libre de Bruxelles (ULB), Gosselies, Belgium
- ULB Center for Research in Immunology (U-CRI), Belgium
- WELBIO Department, WEL Research Institute, Wavre, Belgium
| | - Moosa Rezwani
- Department of Pharmacotherapy and Pharmaceutics, Université Libre de Bruxelles (ULB), Brussels, Belgium
- Institute for Medical Immunology (IMI), Université Libre de Bruxelles (ULB), Gosselies, Belgium
- ULB Center for Research in Immunology (U-CRI), Belgium
- WELBIO Department, WEL Research Institute, Wavre, Belgium
| | - Maria Papadopoulou
- Department of Pharmacotherapy and Pharmaceutics, Université Libre de Bruxelles (ULB), Brussels, Belgium
- Institute for Medical Immunology (IMI), Université Libre de Bruxelles (ULB), Gosselies, Belgium
- ULB Center for Research in Immunology (U-CRI), Belgium
- WELBIO Department, WEL Research Institute, Wavre, Belgium
| | - Amandine Everard
- WELBIO Department, WEL Research Institute, Wavre, Belgium
- Metabolism and Nutrition Research Group, Louvain Drug Research Institute, Université Catholique De Louvain, Brussels, Belgium
| | - Véronique Flamand
- Institute for Medical Immunology (IMI), Université Libre de Bruxelles (ULB), Gosselies, Belgium
- ULB Center for Research in Immunology (U-CRI), Belgium
| | - David Vermijlen
- Department of Pharmacotherapy and Pharmaceutics, Université Libre de Bruxelles (ULB), Brussels, Belgium
- Institute for Medical Immunology (IMI), Université Libre de Bruxelles (ULB), Gosselies, Belgium
- ULB Center for Research in Immunology (U-CRI), Belgium
- WELBIO Department, WEL Research Institute, Wavre, Belgium
| |
Collapse
|
2
|
Ravi VR, Korkmaz FT, De Ana CL, Lu L, Shao FZ, Odom CV, Barker KA, Ramanujan A, Niszczak EN, Goltry WN, Martin IMC, Ha CT, Quinton LJ, Jones MR, Fine A, Welch JD, Chen F, Belkina AC, Mizgerd JP, Shenoy AT. Lung CD4 + resident memory T cells use airway secretory cells to stimulate and regulate onset of allergic airway neutrophilic disease. Cell Rep 2025; 44:115294. [PMID: 39965565 PMCID: PMC12011213 DOI: 10.1016/j.celrep.2025.115294] [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: 07/04/2024] [Revised: 09/26/2024] [Accepted: 01/20/2025] [Indexed: 02/20/2025] Open
Abstract
Neutrophilic asthma is a vexing disease, but mechanistic and therapeutic advancements will require better models of allergy-induced airway neutrophilia. Here, we find that periodic ovalbumin (OVA) inhalation in sensitized mice elicits rapid allergic airway inflammation and pathophysiology mimicking neutrophilic asthma. OVA-experienced murine lungs harbor diverse clusters of CD4+ resident memory T (TRM) cells, including unconventional RORγtnegative/low T helper 17 (TH17) cells. Acute OVA challenge instigates interleukin (IL)-17A secretion from these TRM cells, driving CXCL5 production from Muc5achigh airway secretory cells, leading to destructive airway neutrophilia. The TRM and epithelial cell signals discovered herein are also observed in adult human asthmatic airways. Epithelial antigen presentation regulates this biology by skewing TRM cells toward TH2 and TH1 fates so that TH1-related interferon (IFN)-γ suppresses IL-17A-driven, CXCL5-mediated airway neutrophilia. Concordantly, in vivo IFN-γ supplementation improves disease outcomes. Thus, using our model of neutrophilic asthma, we identify lung epithelial-CD4+ TRM cell crosstalk as a key rheostat of allergic airway neutrophilia.
Collapse
Affiliation(s)
- Vijay Raaj Ravi
- Department of Microbiology and Immunology, University of Michigan Medical School, Ann Arbor, MI, USA
| | - Filiz T Korkmaz
- Pulmonary Center, Boston University Chobanian & Avedisian School of Medicine, Boston, MA, USA; Department of Medicine, University of Massachusetts Chan Medical School, Worcester, MA, USA
| | - Carolina Lyon De Ana
- Pulmonary Center, Boston University Chobanian & Avedisian School of Medicine, Boston, MA, USA; Department of Virology, Immunology, and Microbiology, Boston University Chobanian & Avedisian School of Medicine, Boston, MA, USA
| | - Lu Lu
- Department of Computational Medicine and Bioinformatics, University of Michigan, Ann Arbor, MI, USA
| | - Feng-Zhi Shao
- Pulmonary Center, Boston University Chobanian & Avedisian School of Medicine, Boston, MA, USA
| | - Christine V Odom
- Pulmonary Center, Boston University Chobanian & Avedisian School of Medicine, Boston, MA, USA; Department of Virology, Immunology, and Microbiology, Boston University Chobanian & Avedisian School of Medicine, Boston, MA, USA
| | - Kimberly A Barker
- Pulmonary Center, Boston University Chobanian & Avedisian School of Medicine, Boston, MA, USA; Department of Virology, Immunology, and Microbiology, Boston University Chobanian & Avedisian School of Medicine, Boston, MA, USA
| | - Aditya Ramanujan
- Pulmonary Center, Boston University Chobanian & Avedisian School of Medicine, Boston, MA, USA
| | - Emma N Niszczak
- Department of Microbiology and Immunology, University of Michigan Medical School, Ann Arbor, MI, USA
| | - Wesley N Goltry
- Pulmonary Center, Boston University Chobanian & Avedisian School of Medicine, Boston, MA, USA
| | - Ian M C Martin
- Pulmonary Center, Boston University Chobanian & Avedisian School of Medicine, Boston, MA, USA
| | - Catherine T Ha
- Pulmonary Center, Boston University Chobanian & Avedisian School of Medicine, Boston, MA, USA
| | - Lee J Quinton
- Pulmonary Center, Boston University Chobanian & Avedisian School of Medicine, Boston, MA, USA; Department of Medicine, University of Massachusetts Chan Medical School, Worcester, MA, USA; Department of Virology, Immunology, and Microbiology, Boston University Chobanian & Avedisian School of Medicine, Boston, MA, USA; Department of Medicine, Boston University Chobanian & Avedisian School of Medicine, Boston, MA, USA; Department of Pathology and Laboratory Medicine, Boston University Chobanian & Avedisian School of Medicine, Boston, MA, USA
| | - Matthew R Jones
- Pulmonary Center, Boston University Chobanian & Avedisian School of Medicine, Boston, MA, USA; Department of Virology, Immunology, and Microbiology, Boston University Chobanian & Avedisian School of Medicine, Boston, MA, USA
| | - Alan Fine
- Pulmonary Center, Boston University Chobanian & Avedisian School of Medicine, Boston, MA, USA
| | - Joshua D Welch
- Department of Computational Medicine and Bioinformatics, University of Michigan, Ann Arbor, MI, USA; Department of Computer Science and Engineering, University of Michigan, Ann Arbor, MI, USA
| | - Felicia Chen
- Pulmonary Center, Boston University Chobanian & Avedisian School of Medicine, Boston, MA, USA
| | - Anna C Belkina
- Pulmonary Center, Boston University Chobanian & Avedisian School of Medicine, Boston, MA, USA; Department of Pathology and Laboratory Medicine, Boston University Chobanian & Avedisian School of Medicine, Boston, MA, USA; Flow Cytometry Core Facility, Boston University Chobanian & Avedisian School of Medicine, Boston, MA, USA
| | - Joseph P Mizgerd
- Pulmonary Center, Boston University Chobanian & Avedisian School of Medicine, Boston, MA, USA; Department of Virology, Immunology, and Microbiology, Boston University Chobanian & Avedisian School of Medicine, Boston, MA, USA; Department of Medicine, Boston University Chobanian & Avedisian School of Medicine, Boston, MA, USA; Department of Biochemistry and Cell Biology, Boston University Chobanian & Avedisian School of Medicine, Boston, MA, USA.
| | - Anukul T Shenoy
- Department of Microbiology and Immunology, University of Michigan Medical School, Ann Arbor, MI, USA; Pulmonary Center, Boston University Chobanian & Avedisian School of Medicine, Boston, MA, USA; Department of Internal Medicine, Division of Pulmonary and Critical Care Medicine, University of Michigan Medical School, Ann Arbor, MI, USA.
| |
Collapse
|
3
|
Cox G, Kobayashi M, Rudd BD, Yoshimoto M. Regulation of HSC development and function by Lin28b. Front Cell Dev Biol 2025; 13:1555877. [PMID: 40143971 PMCID: PMC11936975 DOI: 10.3389/fcell.2025.1555877] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2025] [Accepted: 02/24/2025] [Indexed: 03/28/2025] Open
Abstract
Hematopoietic stem cells (HSCs) provide all kinds of blood cells for life while maintaining self-renewal ability. During development, HSCs are first produced in the mouse embryo around embryonic day (E) 11. At this time, only one or two transplantable HSCs can be detected per embryo. Then, HSCs migrate to the fetal liver, where the number of HSCs rapidly increases, showing enhanced self-renewal ability. After birth, a transition occurs from the rapidly proliferating fetal HSCs to the more slowly dividing adult HSCs, which ends by 3-4 weeks of age. It is known that fetal HSCs express distinct surface markers and transcriptomes and produce a variety of distinct immune cells that are not made by adult HSCs. Accumulating evidence indicates that the ontogeny of the hematopoietic system is driven by a highly conserved and developmentally regulated RNA binding protein known as Lin28b. Lin28b is predominantly expressed in the fetal hematopoietic stem and progenitor cells (HSPCs) and regulates the developmental switch from fetal to adult HSCs. In this review, we will provide an overview of how Lin28b regulates the expansion and differentiation of HSCs in early life. These insights can be taken into consideration when developing ex vivo HSC expansion utilizing such physiological characteristics of HSCs.
Collapse
Affiliation(s)
- Grant Cox
- Department of Neurology, University of Washington, Seattle, WA, United States
| | - Michihiro Kobayashi
- Department of Investigative Medicine, Western Michigan University Homer Stryker M.D. School of Medicine, Kalamazoo, MI, United States
| | - Brian D. Rudd
- Department of Microbiology and Immunology, Cornell University, Ithaca, NY, United States
| | - Momoko Yoshimoto
- Department of Investigative Medicine, Western Michigan University Homer Stryker M.D. School of Medicine, Kalamazoo, MI, United States
| |
Collapse
|
4
|
Inácio D, Amado T, Pamplona A, Sobral D, Cunha C, Santos RF, Oliveira L, Rouquié N, Carmo AM, Lesourne R, Gomes AQ, Silva-Santos B. Signature cytokine-associated transcriptome analysis of effector γδ T cells identifies subset-specific regulators of peripheral activation. Nat Immunol 2025; 26:497-510. [PMID: 39881001 PMCID: PMC11876068 DOI: 10.1038/s41590-024-02073-8] [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: 02/27/2024] [Accepted: 12/20/2024] [Indexed: 01/31/2025]
Abstract
γδ T cells producing either interleukin-17A (γδ17 cells) or interferon-γ (γδIFN cells) are generated in the mouse thymus, but the molecular regulators of their peripheral functions are not fully characterized. Here we established an Il17a-GFP:Ifng-YFP double-reporter mouse strain to analyze at unprecedented depth the transcriptomes of pure γδ17 cell versus γδIFN cell populations from peripheral lymph nodes. Within a very high fraction of differentially expressed genes, we identify a panel of 20 new signature genes in steady-state γδ17 cells versus γδIFN cells, which we further validate in models of experimental autoimmune encephalomyelitis and cerebral malaria, respectively. Among the signature genes, we show that the co-receptor CD6 and the signaling protein Themis promote the activation and proliferation of peripheral γδIFN cells in response to T cell antigen receptor stimulation in vitro and to Plasmodium infection in vivo. This resource can help to understand the distinct activities of effector γδ T cell subsets in pathophysiology.
Collapse
MESH Headings
- Animals
- Mice
- Receptors, Antigen, T-Cell, gamma-delta/metabolism
- Receptors, Antigen, T-Cell, gamma-delta/immunology
- Lymphocyte Activation/immunology
- Lymphocyte Activation/genetics
- Gene Expression Profiling
- Transcriptome
- Encephalomyelitis, Autoimmune, Experimental/immunology
- Interferon-gamma/genetics
- Interferon-gamma/metabolism
- Interferon-gamma/immunology
- T-Lymphocyte Subsets/immunology
- T-Lymphocyte Subsets/metabolism
- Mice, Inbred C57BL
- Interleukin-17/genetics
- Interleukin-17/metabolism
- Interleukin-17/immunology
- Mice, Transgenic
- Malaria, Cerebral/immunology
- Cytokines/metabolism
- Cytokines/genetics
Collapse
Affiliation(s)
- Daniel Inácio
- Gulbenkian Institute for Molecular Medicine, Lisbon, Portugal
- Faculdade de Medicina, Universidade de Lisboa, Lisbon, Portugal
| | - Tiago Amado
- Gulbenkian Institute for Molecular Medicine, Lisbon, Portugal
| | - Ana Pamplona
- Gulbenkian Institute for Molecular Medicine, Lisbon, Portugal
| | - Daniel Sobral
- Instituto Nacional de Saúde Dr. Ricardo Jorge, Lisbon, Portugal
| | - Carolina Cunha
- Gulbenkian Institute for Molecular Medicine, Lisbon, Portugal
| | - Rita F Santos
- Instituto de Investigação e Inovação em Saúde (i3S), Universidade do Porto, Porto, Portugal
- Instituto de Biologia Molecular e Celular (IBMC), Porto, Portugal
- ESS, Politécnico do Porto, Porto, Portugal
| | - Liliana Oliveira
- Instituto de Investigação e Inovação em Saúde (i3S), Universidade do Porto, Porto, Portugal
- Instituto de Biologia Molecular e Celular (IBMC), Porto, Portugal
| | - Nelly Rouquié
- Toulouse Institute for Infectious and Inflammatory Diseases (Infinity), INSERM UMR1291, CNRS UMR5051, University Toulouse III, Toulouse, France
| | - Alexandre M Carmo
- Instituto de Investigação e Inovação em Saúde (i3S), Universidade do Porto, Porto, Portugal
- Instituto de Biologia Molecular e Celular (IBMC), Porto, Portugal
| | - Renaud Lesourne
- Toulouse Institute for Infectious and Inflammatory Diseases (Infinity), INSERM UMR1291, CNRS UMR5051, University Toulouse III, Toulouse, France
| | - Anita Q Gomes
- Gulbenkian Institute for Molecular Medicine, Lisbon, Portugal.
- H&TRC Health and Technology Research Center, Escola Superior de Tecnologia da Saúde, Instituto Politécnico de Lisboa, Lisbon, Portugal.
| | - Bruno Silva-Santos
- Gulbenkian Institute for Molecular Medicine, Lisbon, Portugal.
- Faculdade de Medicina, Universidade de Lisboa, Lisbon, Portugal.
| |
Collapse
|
5
|
Tuor M, Stappers MHT, Desgardin A, Ruchti F, Sparber F, Orr SJ, Gow NAR, LeibundGut-Landmann S. Card9 and MyD88 differentially regulate Th17 immunity to the commensal yeast Malassezia in the murine skin. Mucosal Immunol 2025; 18:205-219. [PMID: 39579986 DOI: 10.1016/j.mucimm.2024.11.004] [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: 06/19/2024] [Revised: 10/28/2024] [Accepted: 11/17/2024] [Indexed: 11/25/2024]
Abstract
The fungal community of the skin microbiome is dominated by a single genus, Malassezia. Besides its symbiotic lifestyle at the host interface, this commensal yeast has also been associated with diverse inflammatory skin diseases in humans and pet animals. Stable colonization is maintained by antifungal type 17 immunity. The mechanisms driving Th17 responses to Malassezia remain, however, unclear. Here, we show that the C-type lectin receptors Mincle, Dectin-1, and Dectin-2 recognize conserved patterns in the cell wall of Malassezia and induce dendritic cell activation in vitro, while only Dectin-2 is required for Th17 activation during experimental skin colonization in vivo. In contrast, Toll-like receptor recognition was redundant in this context. Instead, inflammatory IL-1 family cytokines signaling via MyD88 were also implicated in Th17 activation in a T cell-intrinsic manner. Taken together, we characterized the pathways contributing to protective immunity against the most abundant member of the skin mycobiome. This knowledge contributes to the understanding of barrier immunity and its regulation by commensals and is relevant considering how aberrant immune responses are associated with severe skin pathologies.
Collapse
Affiliation(s)
- Meret Tuor
- Section of Immunology, Vetsuisse Faculty and Institute of Experimental Immunology, University of Zürich, 8057 Zurich, Switzerland
| | - Mark H T Stappers
- Medical Research Council Centre for Medical Mycology at the University of Exeter, Department of Biosciences, Faculty of Health and Life Sciences, Geoffrey Pope Building, Stocker Road, Exeter EX4 4QD, UK
| | - Alice Desgardin
- Section of Immunology, Vetsuisse Faculty and Institute of Experimental Immunology, University of Zürich, 8057 Zurich, Switzerland
| | - Fiorella Ruchti
- Section of Immunology, Vetsuisse Faculty and Institute of Experimental Immunology, University of Zürich, 8057 Zurich, Switzerland
| | - Florian Sparber
- Section of Immunology, Vetsuisse Faculty and Institute of Experimental Immunology, University of Zürich, 8057 Zurich, Switzerland
| | - Selinda J Orr
- Wellcome-Wolfson Institute for Experimental Medicine, School of Medicine, Dentistry and Biomedical Science, Queen's University Belfast, Belfast BT9 7BL, UK
| | - Neil A R Gow
- Medical Research Council Centre for Medical Mycology at the University of Exeter, Department of Biosciences, Faculty of Health and Life Sciences, Geoffrey Pope Building, Stocker Road, Exeter EX4 4QD, UK
| | - Salomé LeibundGut-Landmann
- Section of Immunology, Vetsuisse Faculty and Institute of Experimental Immunology, University of Zürich, 8057 Zurich, Switzerland; Medical Research Council Centre for Medical Mycology at the University of Exeter, Department of Biosciences, Faculty of Health and Life Sciences, Geoffrey Pope Building, Stocker Road, Exeter EX4 4QD, UK.
| |
Collapse
|
6
|
Garand M, Huang SSY, Dineen B, Glass IA, Eghtesady P. Differential Regulation of Immune-Related Genes in the Developing Heart. Pediatr Cardiol 2025; 46:442-457. [PMID: 38480572 DOI: 10.1007/s00246-024-03441-9] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/21/2023] [Accepted: 02/02/2024] [Indexed: 02/02/2025]
Abstract
In many congenital heart defects, it can be difficult to ascertain primary pathology from secondary consequences from altered flow through the developing heart. The molecular differences between the growing right and left ventricles (RV and LV, respectively) following the completion of septation and the impact of sex on these mechanisms have not been investigated. We analyzed RNA-seq data derived from twelve RV and LVs, one with Hypoplastic Left Heart Syndrome (HLHS), to compare the transcriptomic landscape between the ventricles during development. Differential gene expression analysis revealed a large proportion of genes unique to either the RV or LV as well as sex bias. Our GO enrichment and network analysis strategy highlighted the differential role of immune functions between the RV and LV in the developing heart. Comparatively, RNA-seq analysis of data from C57Bl6/J mice hearts collected at E14 resulted in the enrichment of similar processes related to T cells and leukocyte migration and activation. Differential gene expression analysis of an HLHS case highlighted significant downregulation of chromatin organization pathways and upregulation of genes involved in muscle organ development. This analysis also identified previously unreported upregulation of genes involved in IL-17 production pathways. In conclusion, differences exist between the gene expression profiles of RV versus LV with the expression of immune-related genes being significantly different between these two chambers. The pathogenesis of HLHS may involve alterations in the expression of chromatin and muscle gene organization as well as upregulation of the IL-17 response pathway.
Collapse
Affiliation(s)
- Mathieu Garand
- Division of Pediatric Cardiothoracic Surgery, Department of Surgery, Washington University School of Medicine, St. Louis, MO, USA
| | - Susie S Y Huang
- Division of Pediatric Cardiothoracic Surgery, Department of Surgery, Washington University School of Medicine, St. Louis, MO, USA
| | - Brian Dineen
- Division of Pediatric Cardiothoracic Surgery, Department of Surgery, Washington University School of Medicine, St. Louis, MO, USA
| | - Ian A Glass
- Department of Pediatrics and Medicine, University of Washington, Seattle, WA, USA
| | - Pirooz Eghtesady
- Division of Pediatric Cardiothoracic Surgery, Department of Surgery, Washington University School of Medicine, St. Louis, MO, USA.
| |
Collapse
|
7
|
Chung J, Lee JC, Oh H, Kim Y, Lim S, Lee C, Shim YG, Bang EC, Baek JH. Gut Microbiota Regulates the Homeostasis of Dendritic Epidermal T Cells. Life (Basel) 2024; 14:1695. [PMID: 39768401 PMCID: PMC11677426 DOI: 10.3390/life14121695] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2024] [Revised: 12/12/2024] [Accepted: 12/20/2024] [Indexed: 01/11/2025] Open
Abstract
Dendritic epidermal T cells (DETCs) are a γδ T cell subset residing in the skin epidermis. Although they have been known for decades, the fate of DETCs has largely remained enigmatic. Recent studies have highlighted the relationship between the gut microbiome and γδ T cells in various epithelial and non-epithelial tissues, such as the small intestine, lung, liver, gingiva, and testis. While the skin microbiota has been shown to impact skin γδ T cells, a direct relationship between the gut microbiota and DETCs remains unexplored. In this study, we investigated whether DETCs are regulated by the gut microbiota in the steady-state skin epidermis. We examined the occurrence of DETCs in Balb/c mice, which have a skin epidermis barely populated with DETCs, compared to C57BL/6 mice, under different housing conditions. Our findings reveal that local skin inflammation markedly increases DETC numbers in the ear epidermis of Balb/c mice and that DETCs are activated by environmental factors. Furthermore, an investigation of the gut microbiota under different housing conditions revealed distinct microbial compositions and functional profiles. Taken together, these results suggest a strong connection between DETCs and gut microbiota.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | - Jea-Hyun Baek
- Laboratory of Inflammation Research, School of Life Science, Handong Global University, Pohang 37554, Republic of Korea; (J.C.); (J.-C.L.); (H.O.); (Y.K.); (S.L.); (C.L.); (Y.-G.S.); (E.-C.B.)
| |
Collapse
|
8
|
Mistri SK, Hilton BM, Horrigan KJ, Andretta ES, Savard R, Dienz O, Hampel KJ, Gerrard DL, Rose JT, Sidiropoulos N, Majumdar D, Boyson JE. SLAM/SAP signaling regulates discrete γδ T cell developmental checkpoints and shapes the innate-like γδ TCR repertoire. eLife 2024; 13:RP97229. [PMID: 39656519 PMCID: PMC11630817 DOI: 10.7554/elife.97229] [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] [Indexed: 12/12/2024] Open
Abstract
During thymic development, most γδ T cells acquire innate-like characteristics that are critical for their function in tumor surveillance, infectious disease, and tissue repair. The mechanisms, however, that regulate γδ T cell developmental programming remain unclear. Recently, we demonstrated that the SLAM/SAP signaling pathway regulates the development and function of multiple innate-like γδ T cell subsets. Here, we used a single-cell proteogenomics approach to identify SAP-dependent developmental checkpoints and to define the SAP-dependent γδ TCR repertoire in mice. SAP deficiency resulted in both a significant loss of an immature Gzma+Blk+Etv5+Tox2+ γδT17 precursor population and a significant increase in Cd4+Cd8+Rorc+Ptcra+Rag1+ thymic γδ T cells. SAP-dependent diversion of embryonic day 17 thymic γδ T cell clonotypes into the αβ T cell developmental pathway was associated with a decreased frequency of mature clonotypes in neonatal thymus, and an altered γδ TCR repertoire in the periphery. Finally, we identify TRGV4/TRAV13-4(DV7)-expressing T cells as a novel, SAP-dependent Vγ4 γδT1 subset. Together, the data support a model in which SAP-dependent γδ/αβ T cell lineage commitment regulates γδ T cell developmental programming and shapes the γδ TCR repertoire.
Collapse
MESH Headings
- Animals
- Receptors, Antigen, T-Cell, gamma-delta/metabolism
- Receptors, Antigen, T-Cell, gamma-delta/genetics
- Receptors, Antigen, T-Cell, gamma-delta/immunology
- Mice
- Signal Transduction
- Signaling Lymphocytic Activation Molecule Associated Protein/metabolism
- Signaling Lymphocytic Activation Molecule Associated Protein/genetics
- Immunity, Innate
- Mice, Inbred C57BL
- T-Lymphocyte Subsets/immunology
- T-Lymphocyte Subsets/metabolism
- Thymus Gland/immunology
- Thymus Gland/metabolism
- Cell Differentiation
- Intraepithelial Lymphocytes/immunology
- Intraepithelial Lymphocytes/metabolism
- Signaling Lymphocytic Activation Molecule Family
Collapse
Affiliation(s)
- Somen K Mistri
- Department of Surgery, Larner College of Medicine, University of VermontBurlingtonUnited States
| | - Brianna M Hilton
- Department of Surgery, Larner College of Medicine, University of VermontBurlingtonUnited States
| | - Katherine J Horrigan
- Department of Surgery, Larner College of Medicine, University of VermontBurlingtonUnited States
| | - Emma S Andretta
- Department of Surgery, Larner College of Medicine, University of VermontBurlingtonUnited States
| | - Remi Savard
- Department of Surgery, Larner College of Medicine, University of VermontBurlingtonUnited States
| | - Oliver Dienz
- Department of Surgery, Larner College of Medicine, University of VermontBurlingtonUnited States
| | - Kenneth J Hampel
- Department of Pathology and Laboratory Medicine, Larner College of Medicine, University of Vermont Medical CenterBurlingtonUnited States
| | - Diana L Gerrard
- Department of Pathology and Laboratory Medicine, Larner College of Medicine, University of Vermont Medical CenterBurlingtonUnited States
| | - Joshua T Rose
- Department of Pathology and Laboratory Medicine, Larner College of Medicine, University of Vermont Medical CenterBurlingtonUnited States
| | - Nikoletta Sidiropoulos
- Department of Pathology and Laboratory Medicine, Larner College of Medicine, University of Vermont Medical CenterBurlingtonUnited States
| | - Dev Majumdar
- Department of Surgery, Larner College of Medicine, University of VermontBurlingtonUnited States
| | - Jonathan E Boyson
- Department of Surgery, Larner College of Medicine, University of VermontBurlingtonUnited States
| |
Collapse
|
9
|
Moog MT, Baltes M, Röpke T, Aschenbrenner F, Maus R, Stolper J, Jonigk D, Prinz I, Kolb M, Maus UA. Innate T-cell-derived IL-17A/F protects from bleomycin-induced acute lung injury but not bleomycin or adenoviral TGF-β1-induced lung fibrosis in mice. Eur J Immunol 2024; 54:e2451323. [PMID: 39235361 PMCID: PMC11628887 DOI: 10.1002/eji.202451323] [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: 06/21/2024] [Revised: 08/26/2024] [Accepted: 08/27/2024] [Indexed: 09/06/2024]
Abstract
The pathobiology of IL-17 in lung fibrogenesis is controversial. Here we examined the role of IL-17A/F in bleomycin (BLM) and adenoviral TGF-β1-induced lung fibrosis in mice. In both experimental models, WT and IL17af-/- mice showed increased collagen contents and remodeled lung architecture as assessed by histopathological examination, suggesting that IL-17A/F is dispensable for lung fibrogenesis. However, IL17af-/- mice responded to the BLM challenge with perturbed lung leukocyte subset recruitment. More specifically, bleomycin triggered angiocentric neutrophilic infiltrations of the lung accompanied by increased mortality of IL17af-/- but not WT mice. WT bone marrow transplantation failed to correct this phenotype in BLM-challenged IL17af-/- mice. Conversely, IL17a/f-/- bone marrow transplantation → WT did not perturb lung leukocytic responses upon BLM. At the same time, IL17af-/- mice treated with recombinant IL-17A/F showed an attenuated lung inflammatory response to BLM. Together, the data show that the degree of BLM-driven acute lung injury was critically dependent on the presence of IL-17A/F, while in both models, the fibrotic remodeling process was not.
Collapse
Affiliation(s)
- Marie T. Moog
- Division of Experimental Pneumology, Clinic for PneumologyHannover Medical SchoolHannoverGermany
| | - Melina Baltes
- Division of Experimental Pneumology, Clinic for PneumologyHannover Medical SchoolHannoverGermany
| | - Tina Röpke
- Division of Experimental Pneumology, Clinic for PneumologyHannover Medical SchoolHannoverGermany
| | - Franziska Aschenbrenner
- Division of Experimental Pneumology, Clinic for PneumologyHannover Medical SchoolHannoverGermany
| | - Regina Maus
- Division of Experimental Pneumology, Clinic for PneumologyHannover Medical SchoolHannoverGermany
| | - Jennifer Stolper
- Division of Experimental Pneumology, Clinic for PneumologyHannover Medical SchoolHannoverGermany
| | - Danny Jonigk
- Institute of PathologyRWTH University Medical CenterAachenGermany
- German Center for Lung Research, partner site BREATHHannoverGermany
| | - Immo Prinz
- Institute of Systems ImmunologyUniversity Medical Center Hamburg‐EppendorfGermany
| | - Martin Kolb
- Department of Medicine, Pathology, and Molecular MedicineMcMaster UniversityHamiltonOntarioCanada
| | - Ulrich A. Maus
- Division of Experimental Pneumology, Clinic for PneumologyHannover Medical SchoolHannoverGermany
- German Center for Lung Research, partner site BREATHHannoverGermany
| |
Collapse
|
10
|
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
|
11
|
Wrage M, Holland T, Nüse B, Kaltwasser J, Fröhlich J, Arnold H, Gießler C, Flamann C, Bruns H, Berges J, Daniel C, Hoffmann MH, Anish C, Seeberger PH, Bogdan C, Dettmer K, Rauh M, Mattner J. Cell type-specific modulation of metabolic, immune-regulatory, and anti-microbial pathways by CD101. Mucosal Immunol 2024; 17:892-910. [PMID: 38901763 DOI: 10.1016/j.mucimm.2024.06.004] [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: 09/26/2023] [Revised: 06/05/2024] [Accepted: 06/09/2024] [Indexed: 06/22/2024]
Abstract
T lymphocytes and myeloid cells express the immunoglobulin-like glycoprotein cluster of differentiation (CD)101, notably in the gut. Here, we investigated the cell-specific functions of CD101 during dextran sulfate sodium (DSS)-induced colitis and Salmonella enterica Typhimurium infection. Similar to conventional CD101-/- mice, animals with a regulatory T cell-specific Cd101 deletion developed more severe intestinal pathology than littermate controls in both models. While the accumulation of T helper 1 cytokines in a CD101-deficient environment entertained DSS-induced colitis, it impeded the replication of Salmonella as revealed by studying CD101-/- x interferon-g-/- mice. Moreover, CD101-expressing neutrophils were capable to restrain Salmonella infection in vitro and in vivo. Both cell-intrinsic and -extrinsic mechanisms of CD101 contributed to the control of bacterial growth and spreading. The CD101-dependent containment of Salmonella infection required the expression of Irg-1 and Nox2 and the production of itaconate and reactive oxygen species. The level of intestinal microbial antigens in the sera of inflammatory bowel disease patients correlated inversely with the expression of CD101 on myeloid cells, which is in line with the suppression of CD101 seen in mice following DSS application or Salmonella infection. Thus, depending on the experimental or clinical setting, CD101 helps to limit inflammatory insults or bacterial infections due to cell type-specific modulation of metabolic, immune-regulatory, and anti-microbial pathways.
Collapse
Affiliation(s)
- Marius Wrage
- Mikrobiologisches Institut-Klinische Mikrobiologie, Immunologie und Hygiene, Universitäts-klinikum Erlangen, Friedrich-Alexander-Universität (FAU) Erlangen-Nürnberg, Erlangen, Germany
| | - Tim Holland
- Mikrobiologisches Institut-Klinische Mikrobiologie, Immunologie und Hygiene, Universitäts-klinikum Erlangen, Friedrich-Alexander-Universität (FAU) Erlangen-Nürnberg, Erlangen, Germany
| | - Björn Nüse
- Mikrobiologisches Institut-Klinische Mikrobiologie, Immunologie und Hygiene, Universitäts-klinikum Erlangen, Friedrich-Alexander-Universität (FAU) Erlangen-Nürnberg, Erlangen, Germany
| | - Johanna Kaltwasser
- Mikrobiologisches Institut-Klinische Mikrobiologie, Immunologie und Hygiene, Universitäts-klinikum Erlangen, Friedrich-Alexander-Universität (FAU) Erlangen-Nürnberg, Erlangen, Germany
| | - Jessica Fröhlich
- Mikrobiologisches Institut-Klinische Mikrobiologie, Immunologie und Hygiene, Universitäts-klinikum Erlangen, Friedrich-Alexander-Universität (FAU) Erlangen-Nürnberg, Erlangen, Germany
| | - Harald Arnold
- Mikrobiologisches Institut-Klinische Mikrobiologie, Immunologie und Hygiene, Universitäts-klinikum Erlangen, Friedrich-Alexander-Universität (FAU) Erlangen-Nürnberg, Erlangen, Germany
| | - Claudia Gießler
- Mikrobiologisches Institut-Klinische Mikrobiologie, Immunologie und Hygiene, Universitäts-klinikum Erlangen, Friedrich-Alexander-Universität (FAU) Erlangen-Nürnberg, Erlangen, Germany
| | - Cindy Flamann
- Medizinische Klinik 5, Universitätsklinikum Erlangen, FAU Erlangen-Nürnberg, Erlangen, Germany
| | - Heiko Bruns
- Medizinische Klinik 5, Universitätsklinikum Erlangen, FAU Erlangen-Nürnberg, Erlangen, Germany
| | - Johannes Berges
- Medizinische Klinik 5, Universitätsklinikum Erlangen, FAU Erlangen-Nürnberg, Erlangen, Germany
| | - Christoph Daniel
- Nephropathologische Abteilung, Universitätsklinikum Erlangen, FAU Erlangen-Nürnberg, Erlangen, Germany
| | - Markus H Hoffmann
- Medizinische Klinik 3, Universitätsklinikum Erlangen, FAU Erlangen-Nürnberg, Erlangen, Germany; Klinik für Dermatologie, Allergologie und Venerologie, Universitätsklinikum Schleswig-Holstein, Universität zu Lübeck, Lübeck, Germany
| | - Chakkumkal Anish
- Max Planck Institute of Colloids and Interfaces, Potsdam, Germany; Bacterial Vaccines Discovery and Early Development, Janssen Pharmaceuticals (Johnson & Johnson), CK Leiden, The Netherlands
| | - Peter H Seeberger
- Max Planck Institute of Colloids and Interfaces, Potsdam, Germany; Freie Universität Berlin, Department of Chemistry and Biochemistry, Berlin, Germany
| | - Christian Bogdan
- Mikrobiologisches Institut-Klinische Mikrobiologie, Immunologie und Hygiene, Universitäts-klinikum Erlangen, Friedrich-Alexander-Universität (FAU) Erlangen-Nürnberg, Erlangen, Germany; FAU Profilzentrum Immunmedizin (FAU I-MED), FAU Erlangen-Nürnberg, Erlangen, Germany
| | - Katja Dettmer
- Institut für Funktionelle Genomik, Universität Regensburg, Regensburg, Germany
| | - Manfred Rauh
- Kinder- und Jugendklinik, Universitätsklinikum Erlangen, FAU Erlangen-Nürnberg, Erlangen, Germany
| | - Jochen Mattner
- Mikrobiologisches Institut-Klinische Mikrobiologie, Immunologie und Hygiene, Universitäts-klinikum Erlangen, Friedrich-Alexander-Universität (FAU) Erlangen-Nürnberg, Erlangen, Germany; FAU Profilzentrum Immunmedizin (FAU I-MED), FAU Erlangen-Nürnberg, Erlangen, Germany.
| |
Collapse
|
12
|
Tani-Ichi S, Obwegs D, Yoshikawa A, Watanabe H, Kitano S, Ejima A, Hatano S, Miyachi H, Cui G, Shimba A, Abe S, Hori S, Kondoh G, Sagar, Yoshikai Y, Ikuta K. A RORE-dependent Intronic Enhancer in the IL-7 Receptor-α Locus Controls Glucose Metabolism via Vγ4+ γδT17 Cells. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2024; 213:283-295. [PMID: 39140825 DOI: 10.4049/jimmunol.2300450] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/06/2023] [Accepted: 05/22/2024] [Indexed: 08/15/2024]
Abstract
The IL-7R regulates the homeostasis, activation, and distribution of T cells in peripheral tissues. Although several transcriptional enhancers that regulate IL-7Rα expression in αβ T cells have been identified, enhancers active in γδ T cells remain unknown. In this article, we discovered an evolutionarily conserved noncoding sequence (CNS) in intron 2 of the IL-7Rα-chain (IL-7Rα) locus and named this region CNS9. CNS9 contained a conserved retinoic acid receptor-related orphan receptor (ROR)-responsive element (RORE) and exerted RORγt-dependent enhancer activity in vitro. Mice harboring point mutations in the RORE in CNS9 (CNS9-RORmut) showed reduced IL-7Rα expression in IL-17-producing Vγ4+ γδ T cells. In addition, the cell number and IL-17A production of Vγ4+ γδ T cells were reduced in the adipose tissue of CNS9-RORmut mice. Consistent with the reduction in IL-17A, CNS9-RORmut mice exhibited decreased IL-33 expression in the adipose tissue, resulting in fewer regulatory T cells and glucose intolerance. The CNS9-ROR motif was partially responsible for IL-7Rα expression in RORγt+ regulatory T cells, whereas IL-7Rα expression was unaffected in RORγt-expressing Vγ2+ γδ T cells, Th17 cells, type 3 innate lymphoid cells, and invariant NKT cells. Our results indicate that CNS9 is a RORΕ-dependent, Vγ4+ γδ T cell-specific IL-7Rα enhancer that plays a critical role in adipose tissue homeostasis via regulatory T cells, suggesting that the evolutionarily conserved RORΕ in IL-7Rα intron 2 may influence the incidence of type 2 diabetes.
Collapse
MESH Headings
- Animals
- Mice
- Introns/genetics
- Receptors, Antigen, T-Cell, gamma-delta/genetics
- Receptors, Antigen, T-Cell, gamma-delta/metabolism
- Enhancer Elements, Genetic/genetics
- Nuclear Receptor Subfamily 1, Group F, Member 3/genetics
- Nuclear Receptor Subfamily 1, Group F, Member 3/metabolism
- Glucose/metabolism
- Receptors, Interleukin-7/genetics
- Receptors, Interleukin-7/metabolism
- Mice, Inbred C57BL
- Th17 Cells/immunology
- Interleukin-17/metabolism
- Interleukin-17/genetics
- Humans
- Adipose Tissue/metabolism
- Adipose Tissue/immunology
Collapse
Affiliation(s)
- Shizue Tani-Ichi
- Laboratory of Immune Regulation, Department of Virus Research, Institute for Life and Medical Sciences, Kyoto University, Kyoto, Japan
- Human Health Sciences, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - David Obwegs
- Department of Medicine II (Gastroenterology, Hepatology, Endocrinology, and Infectious Diseases), Freiburg University Medical Center, Faculty of Medicine, University of Freiburg, Freiburg, Germany
- Faculty of Biology, University of Freiburg, Germany
| | - Alice Yoshikawa
- Human Health Sciences, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Hitomi Watanabe
- Laboratory of Integrative Biological Science, Institute for Life and Medical Sciences, Kyoto University, Kyoto, Japan
| | - Satsuki Kitano
- Reproductive Engineering Team, Institute for Life and Medical Sciences, Kyoto University, Kyoto, Japan
| | - Aki Ejima
- Laboratory of Immune Regulation, Department of Virus Research, Institute for Life and Medical Sciences, Kyoto University, Kyoto, Japan
- Graduate School of Biostudies, Kyoto University, Kyoto, Japan
| | - Shinya Hatano
- Division of Immunology and Genome Biology, Medical Institute of Bioregulation, Kyushu University, Fukuoka, Japan
| | - Hitoshi Miyachi
- Reproductive Engineering Team, Institute for Life and Medical Sciences, Kyoto University, Kyoto, Japan
| | - Guangwei Cui
- Laboratory of Immune Regulation, Department of Virus Research, Institute for Life and Medical Sciences, Kyoto University, Kyoto, Japan
| | - Akihiro Shimba
- Laboratory of Immune Regulation, Department of Virus Research, Institute for Life and Medical Sciences, Kyoto University, Kyoto, Japan
- Human Health Sciences, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Shinya Abe
- Laboratory of Immune Regulation, Department of Virus Research, Institute for Life and Medical Sciences, Kyoto University, Kyoto, Japan
| | - Shohei Hori
- Graduate School of Pharmaceutical Sciences, The University of Tokyo, Tokyo, Japan
| | - Gen Kondoh
- Laboratory of Integrative Biological Science, Institute for Life and Medical Sciences, Kyoto University, Kyoto, Japan
| | - Sagar
- Department of Medicine II (Gastroenterology, Hepatology, Endocrinology, and Infectious Diseases), Freiburg University Medical Center, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Yasunobu Yoshikai
- Division of Host Defense, Medical Institute of Bioregulation, Kyushu University, Fukuoka, Japan
| | - Koichi Ikuta
- Laboratory of Immune Regulation, Department of Virus Research, Institute for Life and Medical Sciences, Kyoto University, Kyoto, Japan
| |
Collapse
|
13
|
Zhang M, Li D, Zhu J, Xia X, Zhang H, Wu J, Wang S, Deng A, Wen Q, Tan J, Hao J, Jiang J, Bao X, Sun G, Lu J, Yang Q, Yang H, Cao G, Yin Z, Wang Q. IL-27 disturbs lipid metabolism and restrains mitochondrial activity to inhibit γδ T17 cell-mediated skin inflammation. Cell Death Dis 2024; 15:491. [PMID: 38982043 PMCID: PMC11233514 DOI: 10.1038/s41419-024-06887-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2024] [Revised: 06/25/2024] [Accepted: 07/02/2024] [Indexed: 07/11/2024]
Abstract
IL-17+ γδ T cells (γδ T17) are kick-starters of inflammation due to their strict immunosurveillance of xenobiotics or cellular damages and rapid response to pro-inflammatory stimulators. IL-27 is a well-recognized pleiotropic immune regulator with potent inhibitory effects on type 17 immune responses. However, its actions on γδ T17 mediated inflammation and the underlying mechanisms are less well understood. Here we find that IL-27 inhibits the production of IL-17 from γδ T cells. Mechanistically, IL-27 promotes lipolysis while inhibits lipogenesis, thus reduces the accumulation of lipids and subsequent membrane phospholipids, which leads to mitochondrial deactivation and ensuing reduction of IL-17. More importantly, Il27ra deficient γδ T cells are more pathogenic in an imiquimod-induced murine psoriasis model, while intracutaneous injection of rmIL-27 ameliorates psoriatic inflammation. In summary, this work uncovered the metabolic basis for the immune regulatory activity of IL-27 in restraining γδ T17 mediated inflammation, which provides novel insights into IL-27/IL-27Ra signaling, γδ T17 biology and the pathogenesis of psoriasis.
Collapse
Affiliation(s)
- Mingyue Zhang
- Guangdong Provincial Key Laboratory of Tumor Interventional Diagnosis and Treatment, Zhuhai Institute of Translational Medicine, Zhuhai People's Hospital (Zhuhai Clinical Medical College of Jinan University), Jinan University, Zhuhai, 519000, China
- State Key Laboratory of Bioactive Molecules and Druggability Assessment, The Biomedical Translational Research Institute, Health Science Center (School of Medicine), Jinan University, Guangzhou, 510632, China
- Key Laboratory of Viral Pathogenesis & Infection Prevention and Control (Jinan University), Ministry of Education, Guangzhou, 510632, China
| | - Dehai Li
- Tianjian Laboratory of Advanced Biomedical Sciences, Institute of Advanced Biomedical Sciences, Zhengzhou University, Zhengzhou, 450001, China
| | - Jing Zhu
- State Key Laboratory of Bioactive Molecules and Druggability Assessment, The Biomedical Translational Research Institute, Health Science Center (School of Medicine), Jinan University, Guangzhou, 510632, China
| | - Xue Xia
- Guangdong Provincial Key Laboratory of Tumor Interventional Diagnosis and Treatment, Zhuhai Institute of Translational Medicine, Zhuhai People's Hospital (Zhuhai Clinical Medical College of Jinan University), Jinan University, Zhuhai, 519000, China
- State Key Laboratory of Bioactive Molecules and Druggability Assessment, The Biomedical Translational Research Institute, Health Science Center (School of Medicine), Jinan University, Guangzhou, 510632, China
- Key Laboratory of Viral Pathogenesis & Infection Prevention and Control (Jinan University), Ministry of Education, Guangzhou, 510632, China
| | - Hua Zhang
- Department of Metabolic and Bariatric Surgery, The First Affiliated Hospital of Jinan University, Guangzhou, 510632, China
| | - Jie Wu
- State Key Laboratory of Bioactive Molecules and Druggability Assessment, The Biomedical Translational Research Institute, Health Science Center (School of Medicine), Jinan University, Guangzhou, 510632, China
| | - Shengli Wang
- Guangdong Provincial Key Laboratory of Tumor Interventional Diagnosis and Treatment, Zhuhai Institute of Translational Medicine, Zhuhai People's Hospital (Zhuhai Clinical Medical College of Jinan University), Jinan University, Zhuhai, 519000, China
- State Key Laboratory of Bioactive Molecules and Druggability Assessment, The Biomedical Translational Research Institute, Health Science Center (School of Medicine), Jinan University, Guangzhou, 510632, China
- Key Laboratory of Viral Pathogenesis & Infection Prevention and Control (Jinan University), Ministry of Education, Guangzhou, 510632, China
| | - Anyi Deng
- Guangdong Provincial Key Laboratory of Tumor Interventional Diagnosis and Treatment, Zhuhai Institute of Translational Medicine, Zhuhai People's Hospital (Zhuhai Clinical Medical College of Jinan University), Jinan University, Zhuhai, 519000, China
- State Key Laboratory of Bioactive Molecules and Druggability Assessment, The Biomedical Translational Research Institute, Health Science Center (School of Medicine), Jinan University, Guangzhou, 510632, China
- Key Laboratory of Viral Pathogenesis & Infection Prevention and Control (Jinan University), Ministry of Education, Guangzhou, 510632, China
| | - Qiong Wen
- Guangdong Provincial Key Laboratory of Tumor Interventional Diagnosis and Treatment, Zhuhai Institute of Translational Medicine, Zhuhai People's Hospital (Zhuhai Clinical Medical College of Jinan University), Jinan University, Zhuhai, 519000, China
- State Key Laboratory of Bioactive Molecules and Druggability Assessment, The Biomedical Translational Research Institute, Health Science Center (School of Medicine), Jinan University, Guangzhou, 510632, China
- Key Laboratory of Viral Pathogenesis & Infection Prevention and Control (Jinan University), Ministry of Education, Guangzhou, 510632, China
| | - Jingyi Tan
- Guangdong Provincial Key Laboratory of Tumor Interventional Diagnosis and Treatment, Zhuhai Institute of Translational Medicine, Zhuhai People's Hospital (Zhuhai Clinical Medical College of Jinan University), Jinan University, Zhuhai, 519000, China
- State Key Laboratory of Bioactive Molecules and Druggability Assessment, The Biomedical Translational Research Institute, Health Science Center (School of Medicine), Jinan University, Guangzhou, 510632, China
- Key Laboratory of Viral Pathogenesis & Infection Prevention and Control (Jinan University), Ministry of Education, Guangzhou, 510632, China
| | - Jianlei Hao
- Guangdong Provincial Key Laboratory of Tumor Interventional Diagnosis and Treatment, Zhuhai Institute of Translational Medicine, Zhuhai People's Hospital (Zhuhai Clinical Medical College of Jinan University), Jinan University, Zhuhai, 519000, China
- State Key Laboratory of Bioactive Molecules and Druggability Assessment, The Biomedical Translational Research Institute, Health Science Center (School of Medicine), Jinan University, Guangzhou, 510632, China
- Key Laboratory of Viral Pathogenesis & Infection Prevention and Control (Jinan University), Ministry of Education, Guangzhou, 510632, China
| | - Jun Jiang
- Guangdong Provincial Key Laboratory of Tumor Interventional Diagnosis and Treatment, Zhuhai Institute of Translational Medicine, Zhuhai People's Hospital (Zhuhai Clinical Medical College of Jinan University), Jinan University, Zhuhai, 519000, China
| | - Xiucong Bao
- School of Biomedical Sciences, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong SAR, China
| | - Guodong Sun
- Guangdong Provincial Key Laboratory of Spine and Spinal Cord Reconstruction, The Fifth Affiliated Hospital (Heyuan Shenhe People's Hospital), Jinan University, Heyuan, 517000, China
| | - Jiajing Lu
- Department of Dermatology, Shanghai Skin Disease Hospital, School of Medicine, Tongji University, Shanghai, 200443, China
- Institute of Psoriasis, School of Medicine, Tongji University, Shanghai, 200443, China
| | - Quanli Yang
- Guangdong Provincial Key Laboratory of Tumor Interventional Diagnosis and Treatment, Zhuhai Institute of Translational Medicine, Zhuhai People's Hospital (Zhuhai Clinical Medical College of Jinan University), Jinan University, Zhuhai, 519000, China
- State Key Laboratory of Bioactive Molecules and Druggability Assessment, The Biomedical Translational Research Institute, Health Science Center (School of Medicine), Jinan University, Guangzhou, 510632, China
| | - Hengwen Yang
- Guangdong Provincial Key Laboratory of Tumor Interventional Diagnosis and Treatment, Zhuhai Institute of Translational Medicine, Zhuhai People's Hospital (Zhuhai Clinical Medical College of Jinan University), Jinan University, Zhuhai, 519000, China.
- State Key Laboratory of Bioactive Molecules and Druggability Assessment, The Biomedical Translational Research Institute, Health Science Center (School of Medicine), Jinan University, Guangzhou, 510632, China.
- Key Laboratory of Viral Pathogenesis & Infection Prevention and Control (Jinan University), Ministry of Education, Guangzhou, 510632, China.
| | - Guangchao Cao
- Guangdong Provincial Key Laboratory of Tumor Interventional Diagnosis and Treatment, Zhuhai Institute of Translational Medicine, Zhuhai People's Hospital (Zhuhai Clinical Medical College of Jinan University), Jinan University, Zhuhai, 519000, China.
- State Key Laboratory of Bioactive Molecules and Druggability Assessment, The Biomedical Translational Research Institute, Health Science Center (School of Medicine), Jinan University, Guangzhou, 510632, China.
- Key Laboratory of Viral Pathogenesis & Infection Prevention and Control (Jinan University), Ministry of Education, Guangzhou, 510632, China.
| | - Zhinan Yin
- Guangdong Provincial Key Laboratory of Tumor Interventional Diagnosis and Treatment, Zhuhai Institute of Translational Medicine, Zhuhai People's Hospital (Zhuhai Clinical Medical College of Jinan University), Jinan University, Zhuhai, 519000, China.
- State Key Laboratory of Bioactive Molecules and Druggability Assessment, The Biomedical Translational Research Institute, Health Science Center (School of Medicine), Jinan University, Guangzhou, 510632, China.
- Key Laboratory of Viral Pathogenesis & Infection Prevention and Control (Jinan University), Ministry of Education, Guangzhou, 510632, China.
| | - Qian Wang
- Guangdong Provincial Key Laboratory of Tumor Interventional Diagnosis and Treatment, Zhuhai Institute of Translational Medicine, Zhuhai People's Hospital (Zhuhai Clinical Medical College of Jinan University), Jinan University, Zhuhai, 519000, China.
- State Key Laboratory of Bioactive Molecules and Druggability Assessment, The Biomedical Translational Research Institute, Health Science Center (School of Medicine), Jinan University, Guangzhou, 510632, China.
- Key Laboratory of Viral Pathogenesis & Infection Prevention and Control (Jinan University), Ministry of Education, Guangzhou, 510632, China.
| |
Collapse
|
14
|
Mistri SK, Hilton BM, Horrigan KJ, Andretta ES, Savard R, Dienz O, Hampel KJ, Gerrard DL, Rose JT, Sidiropoulos N, Majumdar D, Boyson JE. SLAM/SAP signaling regulates discrete γδ T cell developmental checkpoints and shapes the innate-like γδ TCR repertoire. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.01.10.575073. [PMID: 38260519 PMCID: PMC10802474 DOI: 10.1101/2024.01.10.575073] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/24/2024]
Abstract
During thymic development, most γδ T cells acquire innate-like characteristics that are critical for their function in tumor surveillance, infectious disease, and tissue repair. The mechanisms, however, that regulate γδ T cell developmental programming remain unclear. Recently, we demonstrated that the SLAM-SAP signaling pathway regulates the development and function of multiple innate-like γδ T cell subsets. Here, we used a single-cell proteogenomics approach to identify SAP-dependent developmental checkpoints and to define the SAP-dependent γδ TCR repertoire. SAP deficiency resulted in both a significant loss of an immature Gzma + Blk + Etv5 + Tox2 + γδT17 precursor population, and a significant increase in Cd4 + Cd8+ Rorc + Ptcra + Rag1 + thymic γδ T cells. SAP-dependent diversion of embryonic day 17 thymic γδ T cell clonotypes into the αβ T cell developmental pathway was associated with a decreased frequency of mature clonotypes in neonatal thymus, and an altered γδ TCR repertoire in the periphery. Finally, we identify TRGV4/TRAV13-4(DV7)-expressing T cells as a novel, SAP-dependent Vγ4 γδT1 subset. Together, the data suggest that SAP-dependent γδ/αβ T cell lineage commitment regulates γδ T cell developmental programming and shapes the γδ TCR repertoire.
Collapse
Affiliation(s)
- Somen K Mistri
- Department of Surgery, Larner College of Medicine, University of Vermont, Burlington, Vermont 05405, USA
| | - Brianna M. Hilton
- Department of Surgery, Larner College of Medicine, University of Vermont, Burlington, Vermont 05405, USA
| | - Katherine J. Horrigan
- Department of Surgery, Larner College of Medicine, University of Vermont, Burlington, Vermont 05405, USA
| | - Emma S. Andretta
- Department of Surgery, Larner College of Medicine, University of Vermont, Burlington, Vermont 05405, USA
| | - Remi Savard
- Department of Surgery, Larner College of Medicine, University of Vermont, Burlington, Vermont 05405, USA
| | - Oliver Dienz
- Department of Surgery, Larner College of Medicine, University of Vermont, Burlington, Vermont 05405, USA
| | - Kenneth J Hampel
- Department of Pathology and Laboratory Medicine, Larner College of Medicine, University of Vermont Medical Center, Burlington, Vermont 05405, USA
| | - Diana L. Gerrard
- Department of Pathology and Laboratory Medicine, Larner College of Medicine, University of Vermont Medical Center, Burlington, Vermont 05405, USA
| | - Joshua T. Rose
- Department of Pathology and Laboratory Medicine, Larner College of Medicine, University of Vermont Medical Center, Burlington, Vermont 05405, USA
| | - Nikoletta Sidiropoulos
- Department of Pathology and Laboratory Medicine, Larner College of Medicine, University of Vermont Medical Center, Burlington, Vermont 05405, USA
| | - Devdoot Majumdar
- Department of Surgery, Larner College of Medicine, University of Vermont, Burlington, Vermont 05405, USA
| | - Jonathan E. Boyson
- Department of Surgery, Larner College of Medicine, University of Vermont, Burlington, Vermont 05405, USA
| |
Collapse
|
15
|
Ruchti F, Zwicky P, Becher B, Dubrac S, LeibundGut-Landmann S. Epidermal barrier impairment predisposes for excessive growth of the allergy-associated yeast Malassezia on murine skin. Allergy 2024; 79:1531-1547. [PMID: 38385963 DOI: 10.1111/all.16062] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2023] [Revised: 01/15/2024] [Accepted: 01/20/2024] [Indexed: 02/23/2024]
Abstract
BACKGROUND The skin barrier is vital for protection against environmental threats including insults caused by skin-resident microbes. Dysregulation of this barrier is a hallmark of atopic dermatitis (AD) and ichthyosis, with variable consequences for host immune control of colonizing commensals and opportunistic pathogens. While Malassezia is the most abundant commensal fungus of the skin, little is known about the host control of this fungus in inflammatory skin diseases. METHODS In this experimental study, MC903-treated mice were colonized with Malassezia spp. to assess the host-fungal interactions in atopic dermatitis. Additional murine models of AD and ichthyosis, including tape stripping, K5-Nrf2 overexpression and flaky tail mice, were employed to confirm and expand the findings. Skin fungal counts were enumerated. High parameter flow cytometry was used to characterize the antifungal response in the AD-like skin. Structural and functional alterations in the skin barrier were determined by histology and transcriptomics of bulk skin. Finally, differential expression of metabolic genes in Malassezia in atopic and control skin was quantified. RESULTS Malassezia grows excessively in AD-like skin. Fungal overgrowth could, however, not be explained by the altered immune status of the atopic skin. Instead, we found that by upregulating key metabolic genes in the altered cutaneous niche, Malassezia acquired enhanced fitness to efficiently colonise the impaired skin barrier. CONCLUSIONS This study provides evidence that structural and metabolic changes in the dysfunctional epidermal barrier environment provide increased accessibility and an altered lipid profile, to which the lipid-dependent yeast adapts for enhanced nutrient assimilation. Our findings reveal fundamental insights into the implication of the mycobiota in the pathogenesis of common skin barrier disorders.
Collapse
Affiliation(s)
- Fiorella Ruchti
- Section of Immunology, Vetsuisse Faculty, University of Zürich, Zürich, Switzerland
- Institute of Experimental Immunology, University of Zürich, Zürich, Switzerland
| | - Pascale Zwicky
- Institute of Experimental Immunology, University of Zürich, Zürich, Switzerland
| | - Burkhard Becher
- Institute of Experimental Immunology, University of Zürich, Zürich, Switzerland
| | - Sandrine Dubrac
- Department of Dermatology, Venereology and Allergology, Medical University of Innsbruck, Innsbruck, Austria
| | - Salomé LeibundGut-Landmann
- Section of Immunology, Vetsuisse Faculty, University of Zürich, Zürich, Switzerland
- Institute of Experimental Immunology, University of Zürich, Zürich, Switzerland
| |
Collapse
|
16
|
Guo J, Chowdhury RR, Mallajosyula V, Xie J, Dubey M, Liu Y, Li J, Wei YL, Palanski BA, Wang C, Qiu L, Ohanyan M, Kask O, Sola E, Kamalyan L, Lewis DB, Scriba TJ, Davis MM, Dodd D, Zeng X, Chien YH. γδ T cell antigen receptor polyspecificity enables T cell responses to a broad range of immune challenges. Proc Natl Acad Sci U S A 2024; 121:e2315592121. [PMID: 38227652 PMCID: PMC10823224 DOI: 10.1073/pnas.2315592121] [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: 09/15/2023] [Accepted: 11/22/2023] [Indexed: 01/18/2024] Open
Abstract
γδ T cells are essential for immune defense and modulating physiological processes. While they have the potential to recognize large numbers of antigens through somatic gene rearrangement, the antigens which trigger most γδ T cell response remain unidentified, and the role of antigen recognition in γδ T cell function is contentious. Here, we show that some γδ T cell receptors (TCRs) exhibit polyspecificity, recognizing multiple ligands of diverse molecular nature. These ligands include haptens, metabolites, neurotransmitters, posttranslational modifications, as well as peptides and proteins of microbial and host origin. Polyspecific γδ T cells are enriched among activated cells in naive mice and the responding population in infection. They express diverse TCR sequences, have different functional potentials, and include the innate-like γδ T cells, such as the major IL-17 responders in various pathological/physiological conditions. We demonstrate that encountering their antigenic microbiome metabolite maintains their homeostasis and functional response, indicating that their ability to recognize multiple ligands is essential for their function. Human γδ T cells with similar polyspecificity also respond to various immune challenges. This study demonstrates that polyspecificity is a prevalent feature of γδ T cell antigen recognition, which enables rapid and robust T cell responses to a wide range of challenges, highlighting a unique function of γδ T cells.
Collapse
Affiliation(s)
- Jing Guo
- Department of Microbiology and Immunology, Stanford University School of Medicine, Stanford, CA94305
- Program in Immunology, Stanford University School of Medicine, Stanford, CA94305
| | - Roshni Roy Chowdhury
- Department of Microbiology and Immunology, Stanford University School of Medicine, Stanford, CA94305
- Program in Immunology, Stanford University School of Medicine, Stanford, CA94305
| | - Vamsee Mallajosyula
- Institute for Immunity, Transplantation and Infection, Stanford University School of Medicine, Stanford, CA94305
| | - Jianming Xie
- Department of Microbiology and Immunology, Stanford University School of Medicine, Stanford, CA94305
- Program in Immunology, Stanford University School of Medicine, Stanford, CA94305
| | - Megha Dubey
- Department of Microbiology and Immunology, Stanford University School of Medicine, Stanford, CA94305
- Program in Immunology, Stanford University School of Medicine, Stanford, CA94305
| | - Yuanyuan Liu
- Department of Pathology, Stanford University School of Medicine, Stanford, CA94305
| | - Jing Li
- Institute for Immunity, Transplantation and Infection, Stanford University School of Medicine, Stanford, CA94305
| | - Yu-ling Wei
- Department of Microbiology and Immunology, Stanford University School of Medicine, Stanford, CA94305
- Program in Immunology, Stanford University School of Medicine, Stanford, CA94305
| | | | - Conghua Wang
- Department of Microbiology and Immunology, Stanford University School of Medicine, Stanford, CA94305
- Program in Immunology, Stanford University School of Medicine, Stanford, CA94305
| | - Lingfeng Qiu
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou310003, China
- National Medical Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou310003, China
| | - Mané Ohanyan
- Department of Microbiology and Immunology, Stanford University School of Medicine, Stanford, CA94305
- Program in Immunology, Stanford University School of Medicine, Stanford, CA94305
| | - Oliver Kask
- Department of Microbiology and Immunology, Stanford University School of Medicine, Stanford, CA94305
- Program in Immunology, Stanford University School of Medicine, Stanford, CA94305
| | - Elsa Sola
- Institute for Immunity, Transplantation and Infection, Stanford University School of Medicine, Stanford, CA94305
| | - Lilit Kamalyan
- Institute for Immunity, Transplantation and Infection, Stanford University School of Medicine, Stanford, CA94305
| | - David B. Lewis
- Program in Immunology, Stanford University School of Medicine, Stanford, CA94305
- Department of Pediatrics, Stanford University School of Medicine, Stanford, CA94305
| | - Thomas J. Scriba
- South African Tuberculosis Vaccine Initiative, Institute of Infectious Disease and Molecular Medicine and Division of Immunology, Department of Pathology, University of Cape Town, Cape Town7700, South Africa
| | - Mark M. Davis
- Department of Microbiology and Immunology, Stanford University School of Medicine, Stanford, CA94305
- Institute for Immunity, Transplantation and Infection, Stanford University School of Medicine, Stanford, CA94305
- HHMI, Stanford University School of Medicine, Stanford, CA94305
| | - Dylan Dodd
- Department of Microbiology and Immunology, Stanford University School of Medicine, Stanford, CA94305
- Department of Pathology, Stanford University School of Medicine, Stanford, CA94305
| | - Xun Zeng
- Department of Microbiology and Immunology, Stanford University School of Medicine, Stanford, CA94305
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou310003, China
- National Medical Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou310003, China
- Research Units of Infectious disease and Microecology, Chinese Academy of Medical Sciences, Beijing100730, China
| | - Yueh-hsiu Chien
- Department of Microbiology and Immunology, Stanford University School of Medicine, Stanford, CA94305
- Program in Immunology, Stanford University School of Medicine, Stanford, CA94305
| |
Collapse
|
17
|
Kwon DI, Park S, Jeong YL, Kim YM, Min J, Lee C, Choi JA, Choi YH, Kong HJ, Choi Y, Baek S, Lee KJ, Kang YW, Jeong C, You G, Oh Y, Im SK, Song M, Kim JK, Chang J, Choi D, Lee SW. Fc-fused IL-7 provides broad antiviral effects against respiratory virus infections through IL-17A-producing pulmonary innate-like T cells. Cell Rep Med 2024; 5:101362. [PMID: 38232693 PMCID: PMC10829794 DOI: 10.1016/j.xcrm.2023.101362] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2023] [Revised: 10/15/2023] [Accepted: 12/12/2023] [Indexed: 01/19/2024]
Abstract
Repeated pandemics caused by the influenza virus and severe acute respiratory syndrome coronavirus (SARS-CoV) have resulted in serious problems in global public health, emphasizing the need for broad-spectrum antiviral therapeutics against respiratory virus infections. Here, we show the protective effects of long-acting recombinant human interleukin-7 fused with hybrid Fc (rhIL-7-hyFc) against major respiratory viruses, including influenza virus, SARS-CoV-2, and respiratory syncytial virus. Administration of rhIL-7-hyFc in a therapeutic or prophylactic regimen induces substantial antiviral effects. During an influenza A virus (IAV) infection, rhIL-7-hyFc treatment increases pulmonary T cells composed of blood-derived interferon γ (IFNγ)+ conventional T cells and locally expanded IL-17A+ innate-like T cells. Single-cell RNA transcriptomics reveals that rhIL-7-hyFc upregulates antiviral genes in pulmonary T cells and induces clonal expansion of type 17 innate-like T cells. rhIL-7-hyFc-mediated disease prevention is dependent on IL-17A in both IAV- and SARS-CoV-2-infected mice. Collectively, we suggest that rhIL-7-hyFc can be used as a broadly active therapeutic for future respiratory virus pandemic.
Collapse
Affiliation(s)
- Dong-Il Kwon
- Department of Life Sciences, Pohang University of Science and Technology, Pohang 37666, Republic of Korea
| | - Subin Park
- Department of Life Sciences, Pohang University of Science and Technology, Pohang 37666, Republic of Korea
| | - Yujin L Jeong
- Department of Life Sciences, Pohang University of Science and Technology, Pohang 37666, Republic of Korea
| | - Young-Min Kim
- Department of Life Sciences, Pohang University of Science and Technology, Pohang 37666, Republic of Korea
| | - Jeongyong Min
- Department of Life Sciences, Pohang University of Science and Technology, Pohang 37666, Republic of Korea
| | - Changhyung Lee
- Department of Life Sciences, Pohang University of Science and Technology, Pohang 37666, Republic of Korea
| | - Jung-Ah Choi
- Science Unit, International Vaccine Institute, Seoul 08826, Republic of Korea
| | - Yoon Ha Choi
- Department of Life Sciences, Pohang University of Science and Technology, Pohang 37666, Republic of Korea
| | - Hyun-Jung Kong
- Graduate School of Pharmaceutical Sciences, Ewha Womans University, Seoul 03760, Republic of Korea
| | - Youngwon Choi
- Graduate School of Pharmaceutical Sciences, Ewha Womans University, Seoul 03760, Republic of Korea
| | - Seungtae Baek
- Research Institute of NeoImmuneTech Co., Ltd., Pohang 37666, Republic of Korea
| | - Kun-Joo Lee
- Department of Life Sciences, Pohang University of Science and Technology, Pohang 37666, Republic of Korea
| | - Yeon-Woo Kang
- Department of Life Sciences, Pohang University of Science and Technology, Pohang 37666, Republic of Korea
| | - Chaerim Jeong
- Department of Life Sciences, Pohang University of Science and Technology, Pohang 37666, Republic of Korea
| | - Gihoon You
- Department of Life Sciences, Pohang University of Science and Technology, Pohang 37666, Republic of Korea
| | - Youngsik Oh
- Department of Life Sciences, Pohang University of Science and Technology, Pohang 37666, Republic of Korea
| | - Sun-Kyoung Im
- Research Institute of NeoImmuneTech Co., Ltd., Pohang 37666, Republic of Korea
| | - Manki Song
- Science Unit, International Vaccine Institute, Seoul 08826, Republic of Korea
| | - Jong Kyoung Kim
- Department of Life Sciences, Pohang University of Science and Technology, Pohang 37666, Republic of Korea
| | - Jun Chang
- Graduate School of Pharmaceutical Sciences, Ewha Womans University, Seoul 03760, Republic of Korea
| | - Donghoon Choi
- Research Institute of NeoImmuneTech Co., Ltd., Pohang 37666, Republic of Korea.
| | - Seung-Woo Lee
- Department of Life Sciences, Pohang University of Science and Technology, Pohang 37666, Republic of Korea.
| |
Collapse
|
18
|
Nah J, Lee Y, Seong RH. PRDM16 regulates γδT17 cell differentiation via controlling type 17 program and lipid-dependent cell fitness. Front Immunol 2024; 14:1332386. [PMID: 38239368 PMCID: PMC10794300 DOI: 10.3389/fimmu.2023.1332386] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2023] [Accepted: 12/13/2023] [Indexed: 01/22/2024] Open
Abstract
γδT17 cells are a subset of γδT cells producing IL-17, which is crucial for protection against bacterial and fungal infections. It has recently been shown that γδT17 cells have enriched lipid storage and lipid metabolism. However, the regulation of γδT17 cell function and differentiation with respect to lipids remains unknown. Here, we report that PRDM16 is a critical regulator of γδT17 cell differentiation, controlling type 17 immunity gene expression program and lipid-dependent cell fitness. We demonstrated that γδT17 cells have higher lipid-dependent cell fitness, which is negatively correlated with the expression of Prdm16. Loss of Prdm16 enhances the function and differentiation of γδT17 cells, and increases their fitness in lipid-rich environments. Specifically, loss of Prdm16 exacerbates development of psoriasis in the skin, a lipid-rich organ, and Prdm16 controls lipid-mediated differentiation of Vγ4+ γδT17 cells, which are the major source of IL-17 during the onset of psoriasis. Our study highlights the potential impact of PRDM16 on lipid-dependent fitness and protective immune function of γδT cells and also on the immunotherapy of psoriasis and inflammatory diseases.
Collapse
Affiliation(s)
| | | | - Rho H. Seong
- School of Biological Sciences, Institute of Molecular Biology and Genetics, Seoul National University, Seoul, Republic of Korea
| |
Collapse
|
19
|
Ruchti F, Tuor M, Mathew L, McCarthy NE, LeibundGut-Landmann S. γδ T cells respond directly and selectively to the skin commensal yeast Malassezia for IL-17-dependent fungal control. PLoS Pathog 2024; 20:e1011668. [PMID: 38215167 PMCID: PMC10810444 DOI: 10.1371/journal.ppat.1011668] [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: 09/07/2023] [Revised: 01/25/2024] [Accepted: 12/16/2023] [Indexed: 01/14/2024] Open
Abstract
Stable microbial colonization of the skin depends on tight control by the host immune system. The lipid-dependent yeast Malassezia typically colonizes skin as a harmless commensal and is subject to host type 17 immunosurveillance, but this fungus has also been associated with diverse skin pathologies in both humans and animals. Using a murine model of Malassezia exposure, we show that Vγ4+ dermal γδ T cells expand rapidly and are the major source of IL-17A mediating fungal control in colonized skin. A pool of memory-like Malassezia-responsive Vγ4+ T cells persisted in the skin, were enriched in draining lymph nodes even after fungal clearance, and were protective upon fungal re-exposure up to several weeks later. Induction of γδT17 immunity depended on IL-23 and IL-1 family cytokine signalling, whereas Toll-like and C-type lectin receptors were dispensable. Furthermore, Vγ4+ T cells from Malassezia-exposed hosts were able to respond directly and selectively to Malassezia-derived ligands, independently of antigen-presenting host cells. The fungal moieties detected were shared across diverse species of the Malassezia genus, but not conserved in other Basidiomycota or Ascomycota. These data provide novel mechanistic insight into the induction and maintenance of type 17 immunosurveillance of skin commensal colonization that has significant implications for cutaneous health.
Collapse
Affiliation(s)
- Fiorella Ruchti
- Section of Immunology, Vetsuisse Faculty, University of Zürich, Zürich, Switzerland
- Institute of Experimental Immunology, University of Zürich, Zürich, Switzerland
| | - Meret Tuor
- Section of Immunology, Vetsuisse Faculty, University of Zürich, Zürich, Switzerland
- Institute of Experimental Immunology, University of Zürich, Zürich, Switzerland
| | - Liya Mathew
- Centre for Immunobiology, Bart’s and The London School of Medicine and Dentistry, The Blizard Institute, Queen Mary University of London, London, United Kingdom
| | - Neil E McCarthy
- Centre for Immunobiology, Bart’s and The London School of Medicine and Dentistry, The Blizard Institute, Queen Mary University of London, London, United Kingdom
| | - Salomé LeibundGut-Landmann
- Section of Immunology, Vetsuisse Faculty, University of Zürich, Zürich, Switzerland
- Institute of Experimental Immunology, University of Zürich, Zürich, Switzerland
| |
Collapse
|
20
|
Sinton MC, Chandrasegaran PRG, Capewell P, Cooper A, Girard A, Ogunsola J, Perona-Wright G, M Ngoyi D, Kuispond N, Bucheton B, Camara M, Kajimura S, Bénézech C, Mabbott NA, MacLeod A, Quintana JF. IL-17 signalling is critical for controlling subcutaneous adipose tissue dynamics and parasite burden during chronic murine Trypanosoma brucei infection. Nat Commun 2023; 14:7070. [PMID: 37923768 PMCID: PMC10624677 DOI: 10.1038/s41467-023-42918-8] [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: 05/26/2023] [Accepted: 10/25/2023] [Indexed: 11/06/2023] Open
Abstract
In the skin, Trypanosoma brucei colonises the subcutaneous white adipose tissue, and is proposed to be competent for forward transmission. The interaction between parasites, adipose tissue, and the local immune system is likely to drive the adipose tissue wasting and weight loss observed in cattle and humans infected with T. brucei. However, mechanistically, events leading to subcutaneous white adipose tissue wasting are not fully understood. Here, using several complementary approaches, including mass cytometry by time of flight, bulk and single cell transcriptomics, and in vivo genetic models, we show that T. brucei infection drives local expansion of several IL-17A-producing cells in the murine WAT, including TH17 and Vγ6+ cells. We also show that global IL-17 deficiency, or deletion of the adipocyte IL-17 receptor protect from infection-induced WAT wasting and weight loss. Unexpectedly, we find that abrogation of adipocyte IL-17 signalling results in a significant accumulation of Dpp4+ Pi16+ interstitial preadipocytes and increased extravascular parasites in the WAT, highlighting a critical role for IL-17 signalling in controlling preadipocyte fate, subcutaneous WAT dynamics, and local parasite burden. Taken together, our study highlights the central role of adipocyte IL-17 signalling in controlling WAT responses to infection, suggesting that adipocytes are critical coordinators of tissue dynamics and immune responses to T. brucei infection.
Collapse
Affiliation(s)
- Matthew C Sinton
- Wellcome Centre for Integrative Parasitology, University of Glasgow, Glasgow, UK.
- School of Biodiversity, One Health and Veterinary Medicine, University of Glasgow, Glasgow, UK.
- Division of Cardiovascular Science, University of Manchester, Manchester, UK.
| | - Praveena R G Chandrasegaran
- Wellcome Centre for Integrative Parasitology, University of Glasgow, Glasgow, UK
- School of Biodiversity, One Health and Veterinary Medicine, University of Glasgow, Glasgow, UK
| | - Paul Capewell
- Wellcome Centre for Integrative Parasitology, University of Glasgow, Glasgow, UK
- School of Biodiversity, One Health and Veterinary Medicine, University of Glasgow, Glasgow, UK
| | - Anneli Cooper
- Wellcome Centre for Integrative Parasitology, University of Glasgow, Glasgow, UK
- School of Biodiversity, One Health and Veterinary Medicine, University of Glasgow, Glasgow, UK
| | - Alex Girard
- Wellcome Centre for Integrative Parasitology, University of Glasgow, Glasgow, UK
- School of Biodiversity, One Health and Veterinary Medicine, University of Glasgow, Glasgow, UK
| | - John Ogunsola
- Wellcome Centre for Integrative Parasitology, University of Glasgow, Glasgow, UK
- School of Biodiversity, One Health and Veterinary Medicine, University of Glasgow, Glasgow, UK
| | - Georgia Perona-Wright
- Wellcome Centre for Integrative Parasitology, University of Glasgow, Glasgow, UK
- School of Infection and Immunity, University of Glasgow, Glasgow, UK
| | - Dieudonné M Ngoyi
- Department of Parasitology, National Institute of Biomedical Research, Kinshasa, Democratic Republic of Congo
- Member of TrypanoGEN, Kinshasa, Democratic Republic of Congo
| | - Nono Kuispond
- Department of Parasitology, National Institute of Biomedical Research, Kinshasa, Democratic Republic of Congo
- Member of TrypanoGEN, Kinshasa, Democratic Republic of Congo
| | - Bruno Bucheton
- Member of TrypanoGEN, Kinshasa, Democratic Republic of Congo
- Institut de Recherche pour le Développement, Unité Mixte de Recherche IRD-CIRAD 177, Campus International de Baillarguet, Montpellier, France
| | - Mamadou Camara
- Member of TrypanoGEN, Kinshasa, Democratic Republic of Congo
- Programme National de Lutte contre la Trypanosomiase Humaine Africaine, Ministère de la Santé, Conakry, Guinea
| | - Shingo Kajimura
- Division of Endocrinology, Diabetes and Metabolism, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, MA, USA
- Howard Hughes Medical Institute, Chevy Chase, MD, USA
| | - Cécile Bénézech
- Centre for Cardiovascular Science, University of Edinburgh, Edinburgh, EH16 4TJ, Scotland, UK
| | - Neil A Mabbott
- The Roslin Institute and Royal (Dick) School of Veterinary Studies, University of Edinburgh, Edinburgh, UK
| | - Annette MacLeod
- Wellcome Centre for Integrative Parasitology, University of Glasgow, Glasgow, UK
- School of Biodiversity, One Health and Veterinary Medicine, University of Glasgow, Glasgow, UK
- School of Infection and Immunity, University of Glasgow, Glasgow, UK
- Member of TrypanoGEN, Kinshasa, Democratic Republic of Congo
| | - Juan F Quintana
- Wellcome Centre for Integrative Parasitology, University of Glasgow, Glasgow, UK.
- School of Biodiversity, One Health and Veterinary Medicine, University of Glasgow, Glasgow, UK.
- Lydia Becker Institute of Immunology and Inflammation, University of Manchester, Manchester, UK.
- Division of Immunology, Immunity to Infection and Health, Manchester Academic Health Science Centre, University of Manchester, Manchester, UK.
| |
Collapse
|
21
|
Yang T, Barros-Martins J, Wang Z, Wencker M, Zhang J, Smout J, Gambhir P, Janssen A, Schimrock A, Georgiev H, León-Lara X, Weiss S, Huehn J, Prinz I, Krueger A, Foerster R, Walzer T, Ravens S. RORγt + c-Maf + Vγ4 + γδ T cells are generated in the adult thymus but do not reach the periphery. Cell Rep 2023; 42:113230. [PMID: 37815917 DOI: 10.1016/j.celrep.2023.113230] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2023] [Revised: 08/30/2023] [Accepted: 09/21/2023] [Indexed: 10/12/2023] Open
Abstract
T cell receptor (TCR) Vγ4-expressing γδ T cells comprise interferon γ (IFNγ)- and interleukin-17 (IL-17)-producing effector subsets, with a preference for IL-17 effector fate decisions during early ontogeny. The existence of adult-thymus-derived IL-17+ T cells (γδ17) remains controversial. Here, we use a mouse model in which T cells are generated exclusively in the adult thymus and employ single-cell chromatin state analysis to study their development. We identify adult-thymus-derived Vγ4 T cells that have all the molecular programs to become IL-17 producers. However, they have reduced IL-17 production capabilities and rarely reach the periphery. Moreover, this study provides high-resolution profiles of Vγ4 T cells in the adult thymus and lymph nodes and identifies Zeb1 as a potential γδ17 cell regulator. Together, this study provides valuable insights into the developmental traits of Vγ4 T cells during adulthood and supports the idea of age-specific signals required for thymic export and/or peripheral maturation of γδ17 cells.
Collapse
Affiliation(s)
- Tao Yang
- Institute of Immunology, Hannover Medical School, 30625 Hannover, Germany
| | | | - Ziqing Wang
- Institute of Immunology, Hannover Medical School, 30625 Hannover, Germany
| | - Melanie Wencker
- Centre International de Recherche en Infectiologie, INSERM U1111, École Normale Supérieure de Lyon, Claude Bernard University Lyon 1, CNRS, UMR 5308, 69365 Lyon, France
| | - Jiang Zhang
- Centre International de Recherche en Infectiologie, INSERM U1111, École Normale Supérieure de Lyon, Claude Bernard University Lyon 1, CNRS, UMR 5308, 69365 Lyon, France
| | - Justine Smout
- Experimental Immunology, Helmholtz Centre for Infection Research, 39124 Braunschweig, Germany
| | - Prerna Gambhir
- Molecular Immunology, Justus-Liebig-University, 35392 Gießen, Germany
| | - Anika Janssen
- Institute of Immunology, Hannover Medical School, 30625 Hannover, Germany
| | - Anja Schimrock
- Institute of Immunology, Hannover Medical School, 30625 Hannover, Germany
| | - Hristo Georgiev
- Institute of Immunology, Hannover Medical School, 30625 Hannover, Germany
| | - Ximena León-Lara
- Institute of Immunology, Hannover Medical School, 30625 Hannover, Germany
| | - Siegfried Weiss
- Institute of Immunology, Hannover Medical School, 30625 Hannover, Germany; Cluster of Excellence RESIST (EXC 2155), Hannover Medical School, 30625 Hannover, Germany
| | - Jochen Huehn
- Experimental Immunology, Helmholtz Centre for Infection Research, 39124 Braunschweig, Germany; Cluster of Excellence RESIST (EXC 2155), Hannover Medical School, 30625 Hannover, Germany
| | - Immo Prinz
- Institute of Systems Immunology, University Hamburg-Eppendorf, 20246 Hamburg, Germany; Cluster of Excellence RESIST (EXC 2155), Hannover Medical School, 30625 Hannover, Germany
| | - Andreas Krueger
- Molecular Immunology, Justus-Liebig-University, 35392 Gießen, Germany
| | - Reinhold Foerster
- Institute of Immunology, Hannover Medical School, 30625 Hannover, Germany; Cluster of Excellence RESIST (EXC 2155), Hannover Medical School, 30625 Hannover, Germany
| | - Thierry Walzer
- Centre International de Recherche en Infectiologie, INSERM U1111, École Normale Supérieure de Lyon, Claude Bernard University Lyon 1, CNRS, UMR 5308, 69365 Lyon, France
| | - Sarina Ravens
- Institute of Immunology, Hannover Medical School, 30625 Hannover, Germany; Cluster of Excellence RESIST (EXC 2155), Hannover Medical School, 30625 Hannover, Germany.
| |
Collapse
|
22
|
Cui TX, Brady AE, Zhang YJ, Anderson C, Popova AP. IL-17a-producing γδT cells and NKG2D signaling mediate bacterial endotoxin-induced neonatal lung injury: implications for bronchopulmonary dysplasia. Front Immunol 2023; 14:1156842. [PMID: 37744375 PMCID: PMC10514485 DOI: 10.3389/fimmu.2023.1156842] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2023] [Accepted: 08/24/2023] [Indexed: 09/26/2023] Open
Abstract
Bronchopulmonary dysplasia (BPD) is a chronic lung disease in preterm birth survivors characterized by inflammation, impaired alveolarization and dysmorphic vasculature. Activated IL-17A+ lymphocytes are key drivers of inflammation in preterm infants. We have shown that in immature mice chronic airway exposure to lipopolysaccharide (LPS) induces pulmonary inflammation, increased IL-17a expression, and hypoalveolarization, a BPD-like phenotype. The source of IL-17a and contribution to lung pathology is unknown. The natural-killer group 2, member D (NKG2D) receptor mediates activation and IL-17a production in γδ T cells by binding to stress molecules. LPS induces NKG2D ligand expression, including Rae-1 and MULT1. We hypothesized that IL-17a+ γδ T cells and NKG2D signaling mediate neonatal LPS-induced lung injury. Immature C57BL/6J (wild type), Nkg2d-/- or Tcrd-/- (lacking γδ T cells) mice were inoculated with 3ug/10ul of LPS from E. coli O26:B6 or 10ul of PBS intranasally on day of life 3, 5, 7, and 10. Selected mice were treated with neutralizing antibodies against IL-17a, or NKG2D intraperitoneally. Lung immune cells were assessed by flow cytometry and gene expression was analyzed by qPCR. Alveolar growth was assessed by lung morphometry. We established that anti-IL-17a antibody treatment attenuated LPS-induced hypoalveolarization. We found that LPS induced the fraction of IL-17a+NKG2D+ γδ T cells, a major source of IL-17a in the neonatal lung. LPS also induced lung mRNA expression of NKG2D, Rae-1, MULT1, and the DNA damage regulator p53. Anti-NKG2D treatment attenuated the effect of LPS on γδ T cell IL-17a expression, immune cell infiltration and hypoalveolarization. LPS-induced hypoalveolarization was also attenuated in Nkg2d-/- and Tcrd-/- mice. In tracheal aspirates of preterm infants IL-17A and its upstream regulator IL-23 were higher in infants who later developed BPD. Also, human ligands of NKG2D, MICA and MICB were present in the aspirates and MICA correlated with median FiO2. Our novel findings demonstrate a central role for activated IL-17a+ γδ T cells and NKG2D signaling in neonatal LPS-induced lung injury. Future studies will determine the role of NKG2D ligands and effectors, other NKG2D+ cells in early-life endotoxin-induced lung injury and inflammation with a long-term goal to understand how inflammation contributes to BPD pathogenesis.
Collapse
Affiliation(s)
| | | | | | | | - Antonia P. Popova
- Department of Pediatrics, University of Michigan Medical School, Ann Arbor, MI, United States
| |
Collapse
|
23
|
Quintana JF, Sinton MC, Chandrasegaran P, Lestari AN, Heslop R, Cheaib B, Ogunsola J, Ngoyi DM, Kuispond Swar NR, Cooper A, Mabbott NA, Coffelt SB, MacLeod A. γδ T cells control murine skin inflammation and subcutaneous adipose wasting during chronic Trypanosoma brucei infection. Nat Commun 2023; 14:5279. [PMID: 37644007 PMCID: PMC10465518 DOI: 10.1038/s41467-023-40962-y] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2023] [Accepted: 08/17/2023] [Indexed: 08/31/2023] Open
Abstract
African trypanosomes colonise the skin to ensure parasite transmission. However, how the skin responds to trypanosome infection remains unresolved. Here, we investigate the local immune response of the skin in a murine model of infection using spatial and single cell transcriptomics. We detect expansion of dermal IL-17A-producing Vγ6+ cells during infection, which occurs in the subcutaneous adipose tissue. In silico cell-cell communication analysis suggests that subcutaneous interstitial preadipocytes trigger T cell activation via Cd40 and Tnfsf18 signalling, amongst others. In vivo, we observe that female mice deficient for IL-17A-producing Vγ6+ cells show extensive inflammation and limit subcutaneous adipose tissue wasting, independently of parasite burden. Based on these observations, we propose that subcutaneous adipocytes and Vγ6+ cells act in concert to limit skin inflammation and adipose tissue wasting. These studies provide new insights into the role of γδ T cell and subcutaneous adipocytes as homeostatic regulators of skin immunity during chronic infection.
Collapse
Affiliation(s)
- Juan F Quintana
- Wellcome Centre for Integrative Parasitology (WCIP), University of Glasgow, Glasgow, UK.
- School of Biodiversity, One Health, Veterinary Medicine (SBOHVM), College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow, UK.
- Division of Immunology, Immunity to Infection and Respiratory Medicine, Lydia Becker Institute of Immunology and Inflammation. University of Manchester, Manchester, UK.
| | - Matthew C Sinton
- Wellcome Centre for Integrative Parasitology (WCIP), University of Glasgow, Glasgow, UK
- School of Biodiversity, One Health, Veterinary Medicine (SBOHVM), College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow, UK
- Division of Cardiovascular Sciences, University of Manchester, Manchester, UK
| | - Praveena Chandrasegaran
- Wellcome Centre for Integrative Parasitology (WCIP), University of Glasgow, Glasgow, UK
- School of Biodiversity, One Health, Veterinary Medicine (SBOHVM), College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow, UK
| | - Agatha Nabilla Lestari
- Wellcome Centre for Integrative Parasitology (WCIP), University of Glasgow, Glasgow, UK
- School of Biodiversity, One Health, Veterinary Medicine (SBOHVM), College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow, UK
| | - Rhiannon Heslop
- Wellcome Centre for Integrative Parasitology (WCIP), University of Glasgow, Glasgow, UK
- School of Biodiversity, One Health, Veterinary Medicine (SBOHVM), College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow, UK
| | - Bachar Cheaib
- Wellcome Centre for Integrative Parasitology (WCIP), University of Glasgow, Glasgow, UK
- School of Biodiversity, One Health, Veterinary Medicine (SBOHVM), College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow, UK
- Translational Lung Research Center Heidelberg (TLRC), Center for Infectious Diseases, Heidelberg University Hospital, 69120, Heidelberg, Germany
| | - John Ogunsola
- Wellcome Centre for Integrative Parasitology (WCIP), University of Glasgow, Glasgow, UK
- School of Biodiversity, One Health, Veterinary Medicine (SBOHVM), College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow, UK
| | - Dieudonne Mumba Ngoyi
- Department of Parasitology, National Institute of Biomedical Research, Kinshasa, Democratic Republic of the Congo
| | - Nono-Raymond Kuispond Swar
- Wellcome Centre for Integrative Parasitology (WCIP), University of Glasgow, Glasgow, UK
- Department of Parasitology, National Institute of Biomedical Research, Kinshasa, Democratic Republic of the Congo
| | - Anneli Cooper
- Wellcome Centre for Integrative Parasitology (WCIP), University of Glasgow, Glasgow, UK
- School of Biodiversity, One Health, Veterinary Medicine (SBOHVM), College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow, UK
| | - Neil A Mabbott
- The Roslin Institute and Royal (Dick) School of Veterinary Studies, University of Edinburgh, Edinburgh, UK
| | - Seth B Coffelt
- School of Cancer Sciences, University of Glasgow, Glasgow, UK
- Cancer Research UK Beatson Institute, Glasgow, UK
| | - Annette MacLeod
- Wellcome Centre for Integrative Parasitology (WCIP), University of Glasgow, Glasgow, UK.
- School of Biodiversity, One Health, Veterinary Medicine (SBOHVM), College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow, UK.
| |
Collapse
|
24
|
Huang HI, Xue Y, Jewell ML, Tan CY, Theriot B, Aggarwal N, Dockterman J, Lin YD, Schroeder EA, Wang D, Xiong N, Coers J, Shinohara ML, Surana NK, Hammer GE. A binary module for microbiota-mediated regulation of γδ17 cells, hallmarked by microbiota-driven expression of programmed cell death protein 1. Cell Rep 2023; 42:112951. [PMID: 37556321 PMCID: PMC10588736 DOI: 10.1016/j.celrep.2023.112951] [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: 04/11/2022] [Revised: 05/12/2023] [Accepted: 07/24/2023] [Indexed: 08/11/2023] Open
Abstract
Little is known about how microbiota regulate innate-like γδ T cells or how these restrict their effector functions within mucosal barriers, where microbiota provide chronic stimulation. Here, we show that microbiota-mediated regulation of γδ17 cells is binary, where microbiota instruct in situ interleukin-17 (IL-17) production and concomitant expression of the inhibitory receptor programmed cell death protein 1 (PD-1). Microbiota-driven expression of PD-1 and IL-17 and preferential adoption of a PD-1high phenotype are conserved for γδ17 cells across multiple mucosal barriers. Importantly, microbiota-driven PD-1 inhibits in situ IL-17 production by mucosa-resident γδ17 effectors, linking microbiota to their simultaneous activation and suppression. We further show the dynamic nature of this microbiota-driven module and define an inflammation-associated activation state for γδ17 cells marked by augmented PD-1, IL-17, and lipid uptake, thus linking the microbiota to dynamic subset-specific activation and metabolic remodeling to support γδ17 effector functions in a microbiota-dense tissue environment.
Collapse
Affiliation(s)
- Hsin-I Huang
- Department of Pathology, Division of Microbiology and Immunology, University of Utah, Salt Lake City, UT 84112, USA
| | - Yue Xue
- Department of Immunology, Duke University Medical Center, Durham, NC 27710, USA
| | - Mark L Jewell
- Department of Pathology, Division of Microbiology and Immunology, University of Utah, Salt Lake City, UT 84112, USA
| | - Chin Yee Tan
- Department of Molecular Genetics and Microbiology, Duke University Medical Center, Durham, NC 27710, USA; Department of Pediatrics, Duke University Medical Center, Durham, NC 27710, USA
| | - Barbara Theriot
- Department of Pediatrics, Duke University Medical Center, Durham, NC 27710, USA
| | - Nupur Aggarwal
- Department of Immunology, Duke University Medical Center, Durham, NC 27710, USA
| | - Jacob Dockterman
- Department of Immunology, Duke University Medical Center, Durham, NC 27710, USA
| | - Yang-Ding Lin
- Department of Microbiology, Immunology and Molecular Genetics, University of Texas Health Science Center San Antonio, San Antonio, TX 78229, USA
| | - Erin A Schroeder
- Department of Molecular Genetics and Microbiology, Duke University Medical Center, Durham, NC 27710, USA
| | - Donghai Wang
- Department of Medicine, Division of Rheumatology and Immunology, Duke University Medical Center, Durham, NC 27710, USA
| | - Na Xiong
- Department of Microbiology, Immunology and Molecular Genetics, University of Texas Health Science Center San Antonio, San Antonio, TX 78229, USA; Department of Medicine, Division of Dermatology and Cutaneous Surgery, University of Texas Health Science Center San Antonio, San Antonio, TX 78229, USA
| | - Jörn Coers
- Department of Immunology, Duke University Medical Center, Durham, NC 27710, USA; Department of Molecular Genetics and Microbiology, Duke University Medical Center, Durham, NC 27710, USA
| | - Mari L Shinohara
- Department of Immunology, Duke University Medical Center, Durham, NC 27710, USA; Department of Molecular Genetics and Microbiology, Duke University Medical Center, Durham, NC 27710, USA
| | - Neeraj K Surana
- Department of Immunology, Duke University Medical Center, Durham, NC 27710, USA; Department of Molecular Genetics and Microbiology, Duke University Medical Center, Durham, NC 27710, USA; Department of Pediatrics, Duke University Medical Center, Durham, NC 27710, USA
| | - Gianna Elena Hammer
- Department of Pathology, Division of Microbiology and Immunology, University of Utah, Salt Lake City, UT 84112, USA; Department of Immunology, Duke University Medical Center, Durham, NC 27710, USA.
| |
Collapse
|
25
|
Ishikawa J, Suto A, Abe K, Hayashi Y, Suga K, Tanaka S, Kageyama T, Iwata A, Suzuki K, Suzuki K, Nakajima H. IL-21 is required for the maintenance and pathogenesis of murine Vγ4 + IL-17-producing γδT cells. Front Immunol 2023; 14:1211620. [PMID: 37662923 PMCID: PMC10473412 DOI: 10.3389/fimmu.2023.1211620] [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: 04/25/2023] [Accepted: 08/04/2023] [Indexed: 09/05/2023] Open
Abstract
Murine IL-17-producing γδT (γδT17) cells are divided into two subsets: natural γδT17 (nγδT17) cells, whose development is restricted to the fetal thymus, and inducible γδT17 cells, which require antigen exposure for their IL-17 production and are presumed to develop from Rorc + Il17a - CCR9 + immature γδT17 cells in the adult thymus and whose T cell receptor (TCR) is biased toward Vγ4. Although IL-23 is known to be involved in developing γδT17 cells, the roles of other cytokines, such as IL-21, which is involved in developing Th17 cells like IL-23, in the development, maintenance, and pathophysiology of γδT17 cells remain unknown. Here, we show that IL-21 is dispensable for the fetal thymic development of nγδT17 cells but is required for the peripheral maintenance of Vγ4+nγδT17 cells. Upon stimulation with γδTCR, IL-1 plus IL-21 induces the proliferation of Vγ4+nγδT17 cells via STAT3 as effectively as IL-1 plus IL-23. Using bone marrow chimeric mice, we demonstrated that immature γδT17 cells are produced de novo in the adult mice from donor adult bone marrow cells and that IL-21 is dispensable for their development. Instead, IL-21 is required to expand newly induced Vγ4+γδT17 cells in the periphery upon immunization. Finally, using adoptive transfer experiments of γδT17 cells, we found that IL-21 receptors on γδT17 cells are involved in maintaining Vγ4+γδT17 cells, subsequent infiltration of Th17 cells into the spinal cord, and exacerbation of experimental autoimmune encephalomyelitis. Collectively, IL-21 plays a vital role in the maintenance and pathogenesis of Vγ4+γδT17 cells.
Collapse
Affiliation(s)
- Junichi Ishikawa
- Department of Allergy and Clinical Immunology, Graduate School of Medicine, Chiba University, Chiba, Japan
| | - Akira Suto
- Department of Allergy and Clinical Immunology, Graduate School of Medicine, Chiba University, Chiba, Japan
- Institute for Advanced Academic Research, Chiba University, Chiba, Japan
| | - Kazuya Abe
- Department of Allergy and Clinical Immunology, Graduate School of Medicine, Chiba University, Chiba, Japan
| | - Yuki Hayashi
- Department of Allergy and Clinical Immunology, Graduate School of Medicine, Chiba University, Chiba, Japan
| | - Kensuke Suga
- Department of Allergy and Clinical Immunology, Graduate School of Medicine, Chiba University, Chiba, Japan
| | - Shigeru Tanaka
- Department of Allergy and Clinical Immunology, Graduate School of Medicine, Chiba University, Chiba, Japan
| | - Takahiro Kageyama
- Department of Allergy and Clinical Immunology, Graduate School of Medicine, Chiba University, Chiba, Japan
- Institute for Advanced Academic Research, Chiba University, Chiba, Japan
| | - Arifumi Iwata
- Department of Allergy and Clinical Immunology, Graduate School of Medicine, Chiba University, Chiba, Japan
| | - Kazumasa Suzuki
- Department of Allergy and Clinical Immunology, Graduate School of Medicine, Chiba University, Chiba, Japan
| | - Kotaro Suzuki
- Department of Allergy and Clinical Immunology, Graduate School of Medicine, Chiba University, Chiba, Japan
| | - Hiroshi Nakajima
- Department of Allergy and Clinical Immunology, Graduate School of Medicine, Chiba University, Chiba, Japan
- Chiba University Synergy Institute for Futuristic Mucosal Vaccine Research and Development (cSIMVa), Chiba, Japan
| |
Collapse
|
26
|
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
|
27
|
Boodhoo N, Matsuyama-Kato A, Raj S, Fazel F, St-Denis M, Sharif S. Effect of Pre-Treatment with a Recombinant Chicken Interleukin-17A on Vaccine Induced Immunity against a Very Virulent Marek's Disease Virus. Viruses 2023; 15:1633. [PMID: 37631976 PMCID: PMC10459749 DOI: 10.3390/v15081633] [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: 06/22/2023] [Revised: 07/23/2023] [Accepted: 07/24/2023] [Indexed: 08/27/2023] Open
Abstract
The host response to pathogenic microbes can lead to expression of interleukin (IL)-17, which has antimicrobial and anti-viral activity. However, relatively little is known about the basic biological role of chicken IL-17A against avian viruses, particularly against Marek's disease virus (MDV). We demonstrate that, following MDV infection, upregulation of IL-17A mRNA and an increase in the frequency of IL-17A+ T cells in the spleen occur compared to control chickens. To elaborate on the role of chIL-17A in MD, the full-length chIL-17A coding sequence was cloned into a pCDNA3.1-V5/HIS TOPO plasmid. The effect of treatment with pcDNA:chIL-17A plasmid in combination with a vaccine (HVT) and very virulent(vv)MDV challenge or vvMDV infection was assessed. In combination with HVT vaccination, chickens that were inoculated with the pcDNA:chIL-17A plasmid had reduced tumor incidence compared to chickens that received the empty vector control or that were vaccinated only (66.6% in the HVT + empty vector group and 73.33% in HVT group versus 53.3% in the HVT + pcDNA:chIL-17A). Further analysis demonstrated that the chickens that received the HVT vaccine and/or plasmid expressing IL-17A had lower MDV-Meq transcripts in the spleen. In conclusion, chIL-17A can influence the immunity conferred by HVT vaccination against MDV infection in chickens.
Collapse
Affiliation(s)
| | | | | | | | | | - Shayan Sharif
- Department of Pathobiology, Ontario Veterinary College, University of Guelph, Guelph, ON N1G 2W1, Canada; (N.B.); (A.M.-K.); (S.R.); (F.F.); (M.S.-D.)
| |
Collapse
|
28
|
Abou-El-Hassan H, Rezende RM, Izzy S, Gabriely G, Yahya T, Tatematsu BK, Habashy KJ, Lopes JR, de Oliveira GLV, Maghzi AH, Yin Z, Cox LM, Krishnan R, Butovsky O, Weiner HL. Vγ1 and Vγ4 gamma-delta T cells play opposing roles in the immunopathology of traumatic brain injury in males. Nat Commun 2023; 14:4286. [PMID: 37463881 DOI: 10.1038/s41467-023-39857-9] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2022] [Accepted: 06/30/2023] [Indexed: 07/20/2023] Open
Abstract
Traumatic brain injury (TBI) is a leading cause of morbidity and mortality. The innate and adaptive immune responses play an important role in the pathogenesis of TBI. Gamma-delta (γδ) T cells have been shown to affect brain immunopathology in multiple different conditions, however, their role in acute and chronic TBI is largely unknown. Here, we show that γδ T cells affect the pathophysiology of TBI as early as one day and up to one year following injury in a mouse model. TCRδ-/- mice are characterized by reduced inflammation in acute TBI and improved neurocognitive functions in chronic TBI. We find that the Vγ1 and Vγ4 γδ T cell subsets play opposing roles in TBI. Vγ4 γδ T cells infiltrate the brain and secrete IFN-γ and IL-17 that activate microglia and induce neuroinflammation. Vγ1 γδ T cells, however, secrete TGF-β that maintains microglial homeostasis and dampens TBI upon infiltrating the brain. These findings provide new insights on the role of different γδ T cell subsets after brain injury and lay down the principles for the development of targeted γδ T-cell-based therapy for TBI.
Collapse
Affiliation(s)
- Hadi Abou-El-Hassan
- Ann Romney Center for Neurologic Diseases, Brigham & Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Rafael M Rezende
- Ann Romney Center for Neurologic Diseases, Brigham & Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Saef Izzy
- Ann Romney Center for Neurologic Diseases, Brigham & Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Galina Gabriely
- Ann Romney Center for Neurologic Diseases, Brigham & Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Taha Yahya
- Ann Romney Center for Neurologic Diseases, Brigham & Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Bruna K Tatematsu
- Ann Romney Center for Neurologic Diseases, Brigham & Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Karl J Habashy
- Ann Romney Center for Neurologic Diseases, Brigham & Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Juliana R Lopes
- Ann Romney Center for Neurologic Diseases, Brigham & Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Gislane L V de Oliveira
- Ann Romney Center for Neurologic Diseases, Brigham & Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Amir-Hadi Maghzi
- Ann Romney Center for Neurologic Diseases, Brigham & Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Zhuoran Yin
- Ann Romney Center for Neurologic Diseases, Brigham & Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Laura M Cox
- Ann Romney Center for Neurologic Diseases, Brigham & Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Rajesh Krishnan
- Ann Romney Center for Neurologic Diseases, Brigham & Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Oleg Butovsky
- Ann Romney Center for Neurologic Diseases, Brigham & Women's Hospital, Harvard Medical School, Boston, MA, USA
- Evergrande Center for Immunologic Diseases, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Howard L Weiner
- Ann Romney Center for Neurologic Diseases, Brigham & Women's Hospital, Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
29
|
Moos S, Regen T, Wanke F, Tian Y, Arendholz LT, Hauptmann J, Heinen AP, Bleul L, Bier K, El Malki K, Reinhardt C, Prinz I, Diefenbach A, Wolz C, Schittek B, Waisman A, Kurschus FC. IL-17 Signaling in Keratinocytes Orchestrates the Defense against Staphylococcus aureus Skin Infection. J Invest Dermatol 2023; 143:1257-1267.e10. [PMID: 36736996 DOI: 10.1016/j.jid.2023.01.016] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2022] [Revised: 12/05/2022] [Accepted: 01/05/2023] [Indexed: 02/04/2023]
Abstract
Keratinocytes (KCs) form the outer epithelial barrier of the body, protecting against invading pathogens. Mice lacking the IL-17RA or both IL-17A and IL-17F develop spontaneous Staphylococcusaureus skin infections. We found a marked expansion of T17 cells, comprised of RORγt-expressing γδ T cells and T helper 17 cells in the skin-draining lymph nodes of these mice. Contradictory to previous suggestions, this expansion was not a result of a direct negative feedback loop because we found no expansion of T17 cells in mice lacking IL-17 signaling specifically in T cells. Instead, we found that the T17 expansion depended on the microbiota and was observed only when KCs were deficient for IL-17RA signaling. Indeed, mice that lack IL-17RA only in KCs showed an increased susceptibility to experimental epicutaneous infection with S. aureus together with an accumulation of IL-17A-producing γδ T cells. We conclude that deficiency of IL-17RA on KCs leads to microbiota dysbiosis in the skin, which triggers the expansion of IL-17A-producing T cells. Our data show that KCs are the primary target cells of IL-17A and IL-17F, coordinating the defense against microbial invaders in the skin.
Collapse
Affiliation(s)
- Sonja Moos
- Department of Dermatology, Heidelberg University Hospital, Heidelberg, Germany; Institute for Molecular Medicine, Paul Klein Center for Immune Intervention, University Medical Center of the Johannes Gutenberg-University Mainz, Mainz, Germany
| | - Tommy Regen
- Institute for Molecular Medicine, Paul Klein Center for Immune Intervention, University Medical Center of the Johannes Gutenberg-University Mainz, Mainz, Germany
| | - Florian Wanke
- Institute for Molecular Medicine, Paul Klein Center for Immune Intervention, University Medical Center of the Johannes Gutenberg-University Mainz, Mainz, Germany; Neuroscience and Rare Diseases (NRD), Discovery and Translational Area, Roche Pharma Research & Early Development (pRED), Roche Innovation Center, Basel, Switzerland
| | - Yizhu Tian
- Department of Dermatology, Heidelberg University Hospital, Heidelberg, Germany
| | - Lucas T Arendholz
- Department of Dermatology, Heidelberg University Hospital, Heidelberg, Germany
| | - Judith Hauptmann
- Institute for Molecular Medicine, Paul Klein Center for Immune Intervention, University Medical Center of the Johannes Gutenberg-University Mainz, Mainz, Germany
| | - André P Heinen
- Institute for Molecular Medicine, Paul Klein Center for Immune Intervention, University Medical Center of the Johannes Gutenberg-University Mainz, Mainz, Germany
| | - Lisa Bleul
- Interfakultäres Institute for Microbiology, Infectious Diseases, Eberhard Karls University, Tübingen, Germany; Cluster of Excellence EXC 2124 "Controlling Microbes to Fight Infections", Eberhard Karls University, Tübingen, Germany
| | - Katharina Bier
- Division of Dermatooncology, Department of Dermatology, Eberhard Karls University, Tübingen, Germany
| | - Khalifa El Malki
- Institute for Molecular Medicine, Paul Klein Center for Immune Intervention, University Medical Center of the Johannes Gutenberg-University Mainz, Mainz, Germany; Department of Pediatric Hematology/Oncology, Center for Pediatric and Adolescent Medicine, University Medical Center of the Johannes Gutenberg-University, Mainz, Germany
| | - Christoph Reinhardt
- Center for Thrombosis and Hemostasis, University Medical Center of the Johannes Gutenberg-University Mainz, Mainz, Germany; German Center for Cardiovascular Research (DZHK), Partner Site RhineMain, 55131 Mainz, Germany
| | - Immo Prinz
- Institute of Immunology, Hannover Medical School, Hannover, Germany; Institute of Systems Immunology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Andreas Diefenbach
- Institute for Medical Microbiology and Hygiene, University Medical Center of the Johannes Gutenberg-University Mainz, Mainz, Germany; Institute of Microbiology, Infectious Diseases and Immunology, Charite University Medical Center Berlin, Berlin, Germany
| | - Christiane Wolz
- Interfakultäres Institute for Microbiology, Infectious Diseases, Eberhard Karls University, Tübingen, Germany; Cluster of Excellence EXC 2124 "Controlling Microbes to Fight Infections", Eberhard Karls University, Tübingen, Germany
| | - Birgit Schittek
- Division of Dermatooncology, Department of Dermatology, Eberhard Karls University, Tübingen, Germany
| | - Ari Waisman
- Institute for Molecular Medicine, Paul Klein Center for Immune Intervention, University Medical Center of the Johannes Gutenberg-University Mainz, Mainz, Germany; Focus Program Translational Neurosciences, University Medical Center of the Johannes Gutenberg-University Mainz, Mainz, Germany; Research Center for Immunotherapy, University Medical Center of the Johannes Gutenberg-University Mainz, Mainz, Germany
| | - Florian C Kurschus
- Department of Dermatology, Heidelberg University Hospital, Heidelberg, Germany.
| |
Collapse
|
30
|
Teichman S, Wang H, Lee CR, Mohtashami M, Foerster E, Han J, Trotman-Grant AC, Winer S, Tsui H, Philpott DJ, Zúñiga-Pflücker JC. Recent Thymic Emigrants Require RBPJ-Dependent Notch Signaling to Transition into Functionally Mature Naive T Cells. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2023; 211:81-90. [PMID: 37154711 PMCID: PMC10330261 DOI: 10.4049/jimmunol.2300140] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/24/2023] [Accepted: 04/18/2023] [Indexed: 05/10/2023]
Abstract
Recent thymic emigrant (RTE) cells are nascent T cells that continue their post-thymic maturation in the periphery and dominate T cell immune responses in early life and in adults having undergone lymphodepletion regimens. However, the events that govern their maturation and their functionality as they transition to mature naive T cells have not been clearly defined. Using RBPJind mice, we were able to identify different stages of RTE maturation and interrogate their immune function using a T cell transfer model of colitis. As CD45RBlo RTE cells mature, they transition through a CD45RBint immature naive T (INT) cell population that is more immunocompetent but shows a bias toward IL-17 production at the expense of IFN-γ. Additionally, the levels of IFN-γ and IL-17 produced in INT cells are highly dependent on whether Notch signals are received during INT cell maturation or during their effector function. IL-17 production by INT cells showed a total requirement for Notch signaling. Loss of Notch signaling at any stage of INT cells resulted in an impaired colitogenic effect of INT cells. RNA sequencing of INT cells that had matured in the absence of Notch signals showed a reduced inflammatory profile compared with Notch-responsive INT cells. Overall, we have elucidated a previously unknown INT cell stage, revealed its intrinsic bias toward IL-17 production, and demonstrated a role for Notch signaling in INT cell peripheral maturation and effector function in the context of a T cell transfer model of colitis.
Collapse
Affiliation(s)
- Sintia Teichman
- Biological Sciences, Sunnybrook Research Institute, Toronto, ON, Canada
- Department of Immunology, University of Toronto, Toronto, ON, Canada
| | - Helen Wang
- Biological Sciences, Sunnybrook Research Institute, Toronto, ON, Canada
- Department of Immunology, University of Toronto, Toronto, ON, Canada
| | - Christina R. Lee
- Biological Sciences, Sunnybrook Research Institute, Toronto, ON, Canada
| | | | | | - Jianxun Han
- Biological Sciences, Sunnybrook Research Institute, Toronto, ON, Canada
| | - Ashton C. Trotman-Grant
- Biological Sciences, Sunnybrook Research Institute, Toronto, ON, Canada
- Department of Immunology, University of Toronto, Toronto, ON, Canada
| | - Shawn Winer
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, ON, Canada
| | - Hubert Tsui
- Biological Sciences, Sunnybrook Research Institute, Toronto, ON, Canada
- Department of Immunology, University of Toronto, Toronto, ON, Canada
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, ON, Canada
- Division of Hematological Pathology, Department of Laboratory Medicine and Molecular Diagnostics, Precision Diagnostics and Therapeutics Program, Sunnybrook Health Sciences Centre, Toronto, ON, Canada
| | - Dana J. Philpott
- Department of Immunology, University of Toronto, Toronto, ON, Canada
| | - Juan Carlos Zúñiga-Pflücker
- Biological Sciences, Sunnybrook Research Institute, Toronto, ON, Canada
- Department of Immunology, University of Toronto, Toronto, ON, Canada
| |
Collapse
|
31
|
Faragher JL, Auger JL, Osinski V, Meier LA, Engelson BJ, Firulyova MM, Gonzalez-Torres MI, Brombacher F, Zaitsev K, Marath A, Binstadt BA. Autoimmune Valvular Carditis Requires Endothelial Cell TNFR1 Expression. Arterioscler Thromb Vasc Biol 2023; 43:943-957. [PMID: 37021574 PMCID: PMC10213135 DOI: 10.1161/atvbaha.122.319025] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2023] [Accepted: 03/21/2023] [Indexed: 04/07/2023]
Abstract
BACKGROUND Inflammation is a key driver of cardiovascular pathology, and many systemic autoimmune/rheumatic diseases are accompanied by increased cardiac risk. In the K/B.g7 mouse model of coexisting systemic autoantibody-mediated arthritis and valvular carditis, valve inflammation depends on macrophage production of TNF (tumor necrosis factor) and IL-6 (interleukin-6). Here, we sought to determine if other canonical inflammatory pathways participate and to determine whether TNF signaling through TNFR1 (tumor necrosis factor receptor 1) on endothelial cells is required for valvular carditis. METHODS We first asked if type 1, 2, or 3 inflammatory cytokine systems (typified by IFNγ, IL-4, and IL-17, respectively) were critical for valvular carditis in K/B.g7 mice, using a combination of in vivo monoclonal antibody blockade and targeted genetic ablation studies. To define the key cellular targets of TNF, we conditionally deleted its main proinflammatory receptor, TNFR1, in endothelial cells. We analyzed how the absence of endothelial cell TNFR1 affected valve inflammation, lymphangiogenesis, and the expression of proinflammatory genes and molecules. RESULTS We found that typical type 1, 2, and 3 inflammatory cytokine systems were not required for valvular carditis, apart from a known initial requirement of IL-4 for autoantibody production. Despite expression of TNFR1 on a wide variety of cell types in the cardiac valve, deleting TNFR1 specifically on endothelial cells protected K/B.g7 mice from valvular carditis. This protection was accompanied by reduced expression of VCAM-1 (vascular cell adhesion molecule), fewer valve-infiltrating macrophages, reduced pathogenic lymphangiogenesis, and diminished proinflammatory gene expression. CONCLUSIONS TNF and IL-6 are the main cytokines driving valvular carditis in K/B.g7 mice. The interaction of TNF with TNFR1 specifically on endothelial cells promotes cardiovascular pathology in the setting of systemic autoimmune/rheumatic disease, suggesting that therapeutic targeting of the TNF:TNFR1 interaction could be beneficial in this clinical context.
Collapse
Affiliation(s)
- Jessica L. Faragher
- Center for Immunology, University of Minnesota, Minneapolis, MN
- Department of Pediatrics, University of Minnesota, Minneapolis, MN
- University of Minnesota, Minneapolis, MN
| | - Jennifer L Auger
- Center for Immunology, University of Minnesota, Minneapolis, MN
- Department of Pediatrics, University of Minnesota, Minneapolis, MN
- University of Minnesota, Minneapolis, MN
| | - Victoria Osinski
- Center for Immunology, University of Minnesota, Minneapolis, MN
- Department of Pediatrics, University of Minnesota, Minneapolis, MN
- University of Minnesota, Minneapolis, MN
| | - Lee A Meier
- Center for Immunology, University of Minnesota, Minneapolis, MN
- Department of Pediatrics, University of Minnesota, Minneapolis, MN
- University of Minnesota, Minneapolis, MN
- Department of Surgery, University of Colorado, Aurora, CO
| | - Brianna J Engelson
- Center for Immunology, University of Minnesota, Minneapolis, MN
- University of Minnesota, Minneapolis, MN
| | - Maria M. Firulyova
- Almazov National Medical Research Centre, Saint-Petersburg, Russia
- Computer Technologies Laboratory, ITMO University, Saint Petersburg, Russia
| | | | - Frank Brombacher
- International Centre for Genetic Engineering and Biotechnology, Cape Town, South Africa
| | - Konstantin Zaitsev
- Computer Technologies Laboratory, ITMO University, Saint Petersburg, Russia
| | | | - Bryce A Binstadt
- Center for Immunology, University of Minnesota, Minneapolis, MN
- Department of Pediatrics, University of Minnesota, Minneapolis, MN
- University of Minnesota, Minneapolis, MN
| |
Collapse
|
32
|
James KD, White AJ, Jenkinson WE, Anderson G. The medulla controls effector primed γδT-cell development in the adult mouse thymus. Eur J Immunol 2023; 53:e2350388. [PMID: 36929102 PMCID: PMC10947249 DOI: 10.1002/eji.202350388] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2023] [Revised: 02/21/2023] [Accepted: 03/14/2023] [Indexed: 03/18/2023]
Abstract
γδT cells are produced in the thymus throughout life and provide immunity at epithelial-rich sites. Unlike conventional αβT cells, γδT-cell development involves intrathymic acquisition of effector function, with priming for either IL17 or IFN-γ production occurring during embryonic or adult life, respectively. How the thymus controls effector-primed γδT-cell generation in adulthood is poorly understood. Here, we distinguished de novo γδT cells from those undergoing thymus recirculation and/or retention using Rag2GFP mice alongside markers of maturation/effector priming including CD24, CD25, CD73, and IFN-γ, the latter by crossing with IFN-γYFP GREAT mice. We categorize newly developing γδT-cells into an ordered sequence where CD25+ CD73- IFN-γYFP- precursors are followed sequentially by CD25- CD73+ IFN-γYFP- intermediates and CD25- CD73+ IFN-γYFP+ effectors. To determine intrathymic requirements controlling this sequence, we examined γδT-cell development in Relb-/- thymus grafts that lack medullary microenvironments. Interestingly, medulla deficiency did not alter CD25+ γδT-cell precursor generation, but significantly impaired development of effector primed stages. This impact on γδT-cell priming was mirrored in plt/plt mice lacking the medullary chemoattractants CCL19 and CCL21, and also Ccl21a-/- but not Ccl19-/- mice. Collectively, we identify the medulla as an important site for effector priming during adult γδT-cell development and demonstrate a specific role for the medullary epithelial product CCL21 in this process.
Collapse
Affiliation(s)
- Kieran D. James
- Institute of Immunology and ImmunotherapyUniversity of BirminghamBirminghamUK
| | - Andrea J. White
- Institute of Immunology and ImmunotherapyUniversity of BirminghamBirminghamUK
| | | | - Graham Anderson
- Institute of Immunology and ImmunotherapyUniversity of BirminghamBirminghamUK
| |
Collapse
|
33
|
Wang Y, Qin H, Cai Y, Chen X, Li H, Montoya-Durango DE, Ding C, Hu X, Chariker JH, Sarojini H, Chien S, Rouchka EC, Zhang HG, Zheng J, Qiu F, Yan J. Natural γδT17 cell development and functional acquisition is governed by the mTORC2- c-Maf-controlled mitochondrial fission pathway. iScience 2023; 26:106630. [PMID: 37192973 PMCID: PMC10182300 DOI: 10.1016/j.isci.2023.106630] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2022] [Revised: 02/17/2023] [Accepted: 04/04/2023] [Indexed: 05/18/2023] Open
Abstract
Natural IL-17-producing γδ T cells (γδT17 cells) are unconventional innate-like T cells that undergo functional programming in the fetal thymus. However, the intrinsic metabolic mechanisms of γδT17 cell development remain undefined. Here, we demonstrate that mTORC2, not mTORC1, selectively controls the functional fate commitment of γδT17 cells through regulating transcription factor c-Maf expression. scRNA-seq data suggest that fetal and adult γδT17 cells predominately utilize mitochondrial metabolism. mTORC2 deficiency results in impaired Drp1-mediated mitochondrial fission and mitochondrial dysfunction characterized by mitochondrial membrane potential (ΔΨm) loss, reduced oxidative phosphorylation (OXPHOS), and subsequent ATP depletion. Treatment with the Drp1 inhibitor Mdivi-1 alleviates imiquimod-induced skin inflammation. Reconstitution of intracellular ATP levels by ATP-encapsulated liposome completely rescues γδT17 defect caused by mTORC2 deficiency, revealing the fundamental role of metabolite ATP in γδT17 development. These results provide an in-depth insight into the intrinsic link between the mitochondrial OXPHOS pathway and γδT17 thymic programming and functional acquisition.
Collapse
Affiliation(s)
- Yunke Wang
- Division of Immunotherapy, The Hiram C. Polk, Jr., MD Department of Surgery, Immuno-Oncology Program, Brown Cancer Center, University of Louisville, Louisville, KY, USA
- Key Laboratory of Tumor Microenvironment and Immune Therapy of Zhejiang Province, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
- Department of Medical Oncology, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Hui Qin
- Division of Immunotherapy, The Hiram C. Polk, Jr., MD Department of Surgery, Immuno-Oncology Program, Brown Cancer Center, University of Louisville, Louisville, KY, USA
- Department of Dermatology, Shanghai Jiaotong University Ruijin Hospital, Shanghai, China
| | - Yihua Cai
- Division of Immunotherapy, The Hiram C. Polk, Jr., MD Department of Surgery, Immuno-Oncology Program, Brown Cancer Center, University of Louisville, Louisville, KY, USA
| | - Xu Chen
- Division of Immunotherapy, The Hiram C. Polk, Jr., MD Department of Surgery, Immuno-Oncology Program, Brown Cancer Center, University of Louisville, Louisville, KY, USA
- Department of Clinical Immunology, Shanghai Ninth People’s Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Hong Li
- Functional Immunomics Core, Brown Cancer Center, University of Louisville, Louisville, KY, USA
| | - Diego Elias Montoya-Durango
- Division of Immunotherapy, The Hiram C. Polk, Jr., MD Department of Surgery, Immuno-Oncology Program, Brown Cancer Center, University of Louisville, Louisville, KY, USA
| | - Chuanlin Ding
- Division of Immunotherapy, The Hiram C. Polk, Jr., MD Department of Surgery, Immuno-Oncology Program, Brown Cancer Center, University of Louisville, Louisville, KY, USA
| | - Xiaoling Hu
- Division of Immunotherapy, The Hiram C. Polk, Jr., MD Department of Surgery, Immuno-Oncology Program, Brown Cancer Center, University of Louisville, Louisville, KY, USA
| | - Julia H. Chariker
- Department of Anatomical Sciences and Neurobiology, University of Louisville, Louisville, KY, USA
- Kentucky Biomedical Research Infrastructure Network Bioinformatics Core University of Louisville, Louisville, KY, USA
| | - Harshini Sarojini
- Division of Immunotherapy, The Hiram C. Polk, Jr., MD Department of Surgery, Immuno-Oncology Program, Brown Cancer Center, University of Louisville, Louisville, KY, USA
| | - Sufan Chien
- Division of Immunotherapy, The Hiram C. Polk, Jr., MD Department of Surgery, Immuno-Oncology Program, Brown Cancer Center, University of Louisville, Louisville, KY, USA
| | - Eric C. Rouchka
- Kentucky Biomedical Research Infrastructure Network Bioinformatics Core University of Louisville, Louisville, KY, USA
- Department of Biochemistry and Molecular Genetics, University of Louisville, Louisville, KY, USA
| | - Huang-Ge Zhang
- Department of Microbiology and Immunology, Brown Cancer Center, University of Louisville, Louisville, KY, USA
| | - Jie Zheng
- Department of Dermatology, Shanghai Jiaotong University Ruijin Hospital, Shanghai, China
| | - Fuming Qiu
- Key Laboratory of Tumor Microenvironment and Immune Therapy of Zhejiang Province, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
- Department of Medical Oncology, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Jun Yan
- Division of Immunotherapy, The Hiram C. Polk, Jr., MD Department of Surgery, Immuno-Oncology Program, Brown Cancer Center, University of Louisville, Louisville, KY, USA
| |
Collapse
|
34
|
MacNabb BW, Rothenberg EV. Speed and navigation control of thymocyte development by the fetal T-cell gene regulatory network. Immunol Rev 2023; 315:171-196. [PMID: 36722494 PMCID: PMC10771342 DOI: 10.1111/imr.13190] [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] [Indexed: 02/02/2023]
Abstract
T-cell differentiation is a tightly regulated developmental program governed by interactions between transcription factors (TFs) and chromatin landscapes and affected by signals received from the thymic stroma. This process is marked by a series of checkpoints: T-lineage commitment, T-cell receptor (TCR)β selection, and positive and negative selection. Dynamically changing combinations of TFs drive differentiation along the T-lineage trajectory, through mechanisms that have been most extensively dissected in adult mouse T-lineage cells. However, fetal T-cell development differs from adult in ways that suggest that these TF mechanisms are not fully deterministic. The first wave of fetal T-cell differentiation occurs during a unique developmental window during thymic morphogenesis, shows more rapid kinetics of differentiation with fewer rounds of cell division, and gives rise to unique populations of innate lymphoid cells (ILCs) and invariant γδT cells that are not generated in the adult thymus. As the characteristic kinetics and progeny biases are cell-intrinsic properties of thymic progenitors, the differences could be based on distinct TF network circuitry within the progenitors themselves. Here, we review recent single-cell transcriptome data that illuminate the TF networks involved in T-cell differentiation in the fetal and adult mouse thymus.
Collapse
Affiliation(s)
- Brendan W MacNabb
- Division of Biology & Biological Engineering, California Institute of Technology, Pasadena, California, USA
| | - Ellen V Rothenberg
- Division of Biology & Biological Engineering, California Institute of Technology, Pasadena, California, USA
| |
Collapse
|
35
|
Wilson A, Bogie B, Chaaban H, Burge K. The Nonbacterial Microbiome: Fungal and Viral Contributions to the Preterm Infant Gut in Health and Disease. Microorganisms 2023; 11:909. [PMID: 37110332 PMCID: PMC10144239 DOI: 10.3390/microorganisms11040909] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2023] [Revised: 03/27/2023] [Accepted: 03/30/2023] [Indexed: 04/29/2023] Open
Abstract
The intestinal microbiome is frequently implicated in necrotizing enterocolitis (NEC) pathogenesis. While no particular organism has been associated with NEC development, a general reduction in bacterial diversity and increase in pathobiont abundance has been noted preceding disease onset. However, nearly all evaluations of the preterm infant microbiome focus exclusively on the bacterial constituents, completely ignoring any fungi, protozoa, archaea, and viruses present. The abundance, diversity, and function of these nonbacterial microbes within the preterm intestinal ecosystem are largely unknown. Here, we review findings on the role of fungi and viruses, including bacteriophages, in preterm intestinal development and neonatal intestinal inflammation, with potential roles in NEC pathogenesis yet to be determined. In addition, we highlight the importance of host and environmental influences, interkingdom interactions, and the role of human milk in shaping fungal and viral abundance, diversity, and function within the preterm intestinal ecosystem.
Collapse
Affiliation(s)
| | | | - Hala Chaaban
- Division of Neonatal-Perinatal Medicine, Department of Pediatrics, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA
| | - Kathryn Burge
- Division of Neonatal-Perinatal Medicine, Department of Pediatrics, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA
| |
Collapse
|
36
|
Hahn AM, Vogg L, Brey S, Schneider A, Schäfer S, Palmisano R, Pavlova A, Sandrock I, Tan L, Fichtner AS, Prinz I, Ravens S, Winkler TH. A monoclonal Trd chain supports the development of the complete set of functional γδ T cell lineages. Cell Rep 2023; 42:112253. [PMID: 36920908 DOI: 10.1016/j.celrep.2023.112253] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2022] [Revised: 11/14/2022] [Accepted: 02/28/2023] [Indexed: 03/16/2023] Open
Abstract
The clonal selection theory describes key features of adaptive immune responses of B and T cells. For αβ T cells and B cells, antigen recognition and selection principles are known at a detailed molecular level. The precise role of the antigen receptor in γδ T cells remains less well understood. To better understand the role of the γδ T cell receptor (TCR), we generate an orthotopic TCRδ transgenic mouse model. We demonstrate a multi-layered functionality of γδ TCRs and diverse roles of CDR3δ-mediated selection during γδ T cell development. Whereas epithelial populations using Vγ5 or Vγ7 chains are almost unaffected in their biology in the presence of the transgenic TCRδ chain, pairing with Vγ1 positively selects γδ T cell subpopulations with distinct programs in several organs, thereby distorting the repertoire. In conclusion, our data support dictation of developmental tropism together with adaptive-like recognition principles in a single antigen receptor.
Collapse
Affiliation(s)
- Anne M Hahn
- Division of Genetics, Department Biology, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), 91054 Erlangen, Germany
| | - Lisa Vogg
- Division of Genetics, Department Biology, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), 91054 Erlangen, Germany
| | - Stefanie Brey
- Division of Genetics, Department Biology, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), 91054 Erlangen, Germany
| | - Andrea Schneider
- Division of Genetics, Department Biology, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), 91054 Erlangen, Germany
| | - Simon Schäfer
- Division of Genetics, Department Biology, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), 91054 Erlangen, Germany
| | - Ralph Palmisano
- Optical Imaging Centre Erlangen (OICE), Competence Unit, FAU, 91058 Erlangen, Germany
| | - Anna Pavlova
- Division of Genetics, Department Biology, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), 91054 Erlangen, Germany
| | | | - Likai Tan
- Medizinische Hochschule Hannover, Hannover, Germany
| | | | - Immo Prinz
- Medizinische Hochschule Hannover, Hannover, Germany; Institute for Systems Immunology, Universitätsklinikum Hamburg-Eppendorf, Hamburg, Germany
| | | | - Thomas H Winkler
- Division of Genetics, Department Biology, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), 91054 Erlangen, Germany.
| |
Collapse
|
37
|
Dou Y, Shan S, Zhang J. UcTCRdb: An unconventional T cell receptor sequence database with online analysis functions. Front Immunol 2023; 14:1158295. [PMID: 36993970 PMCID: PMC10040587 DOI: 10.3389/fimmu.2023.1158295] [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: 02/03/2023] [Accepted: 03/02/2023] [Indexed: 03/14/2023] Open
Abstract
Unlike conventional major histocompatibility complex (MHC) class I and II molecules reactive T cells, the unconventional T cell subpopulations recognize various non-polymorphic antigen-presenting molecules and are typically characterized by simplified patterns of T cell receptors (TCRs), rapid effector responses and ‘public’ antigen specificities. Dissecting the recognition patterns of the non-MHC antigens by unconventional TCRs can help us further our understanding of the unconventional T cell immunity. The small size and irregularities of the released unconventional TCR sequences are far from high-quality to support systemic analysis of unconventional TCR repertoire. Here we present UcTCRdb, a database that contains 669,900 unconventional TCRs collected from 34 corresponding studies in humans, mice, and cattle. In UcTCRdb, users can interactively browse TCR features of different unconventional T cell subsets in different species, search and download sequences under different conditions. Additionally, basic and advanced online TCR analysis tools have been integrated into the database, which will facilitate the study of unconventional TCR patterns for users with different backgrounds. UcTCRdb is freely available at http://uctcrdb.cn/.
Collapse
|
38
|
Stromal regulation of the intestinal barrier. Mucosal Immunol 2023; 16:221-231. [PMID: 36708806 DOI: 10.1016/j.mucimm.2023.01.006] [Citation(s) in RCA: 19] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2022] [Revised: 12/27/2022] [Accepted: 01/12/2023] [Indexed: 01/26/2023]
Abstract
The intestinal barrier is a complex structure that allows the absorption of nutrients while ensuring protection against intestinal pathogens and balanced immunity. The development and maintenance of a functional intestinal barrier is a multifactorial process that is only partially understood. Here we review novel findings on the emerging role of mesenchymal cells in this process using insights gained from lineage tracing approaches, Cre-based gene deletion, and single-cell transcriptomics. The current evidence points toward a key organizer role for distinct mesenchymal lineages in intestinal development and homeostasis, regulating both epithelial and immune components of the intestinal barrier. We further discuss recent findings on functional mesenchymal heterogeneity and implications for intestinal regeneration and inflammatory intestinal pathologies.
Collapse
|
39
|
Muro R, Narita T, Nitta T, Takayanagi H. Spleen tyrosine kinase mediates the γδTCR signaling required for γδT cell commitment and γδT17 differentiation. Front Immunol 2023; 13:1045881. [PMID: 36713401 PMCID: PMC9878111 DOI: 10.3389/fimmu.2022.1045881] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2022] [Accepted: 12/23/2022] [Indexed: 01/13/2023] Open
Abstract
The γδT cells that produce IL-17 (γδT17 cells) play a key role in various pathophysiologic processes in host defense and homeostasis. The development of γδT cells in the thymus requires γδT cell receptor (γδTCR) signaling mediated by the spleen tyrosine kinase (Syk) family proteins, Syk and Zap70. Here, we show a critical role of Syk in the early phase of γδT cell development using mice deficient for Syk specifically in lymphoid lineage cells (Syk-conditional knockout (cKO) mice). The development of γδT cells in the Syk-cKO mice was arrested at the precursor stage where the expression of Rag genes and αβT-lineage-associated genes were retained, indicating that Syk is required for γδT-cell lineage commitment. Loss of Syk in γδT cells weakened TCR signal-induced phosphorylation of Erk and Akt, which is mandatory for the thymic development of γδT17 cells. Syk-cKO mice exhibited a loss of γδT17 cells in the thymus as well as throughout the body, and thereby are protected from γδT17-dependent psoriasis-like skin inflammation. Collectively, our results indicate that Syk is a key player in the lineage commitment of γδT cells and the priming of γδT17 cell differentiation.
Collapse
Affiliation(s)
- Ryunosuke Muro
- Department of Immunology, Graduate School of Medicine and Faculty of Medicine, The University of Tokyo, Tokyo, Japan
| | - Tomoya Narita
- Department of Pharmacotherapeutics, Research Institute of Pharmaceutical Sciences and Faculty of Pharmacy, Musashino University, Tokyo, Japan
| | - Takeshi Nitta
- Department of Immunology, Graduate School of Medicine and Faculty of Medicine, The University of Tokyo, Tokyo, Japan,*Correspondence: Takeshi Nitta,
| | - Hiroshi Takayanagi
- Department of Immunology, Graduate School of Medicine and Faculty of Medicine, The University of Tokyo, Tokyo, Japan
| |
Collapse
|
40
|
Contreras AV, Wiest DL. Development of γδ T Cells: Soldiers on the Front Lines of Immune Battles. Methods Mol Biol 2023; 2580:71-88. [PMID: 36374451 DOI: 10.1007/978-1-0716-2740-2_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] [Indexed: 06/14/2023]
Abstract
While the functions of αβ T cells in host resistance to pathogen infection are understood in far more detail than those of γδ lineage T cells, γδ T cells perform critical, essential functions during immune responses that cannot be compensated for by αβ T cells. Accordingly, it is critical to understand how the development of γδ T cells is controlled so that their generation and function might be manipulated in future for therapeutic benefit. This introductory chapter will focus primarily on the basic processes that underlie γδ T cell development in the thymus, as well as the current understanding of how they are controlled.
Collapse
Affiliation(s)
- Alejandra V Contreras
- Blood Cell Development and Function Program, Fox Chase Cancer Center, Philadelphia, PA, USA
| | - David L Wiest
- Blood Cell Development and Function Program, Fox Chase Cancer Center, Philadelphia, PA, USA.
| |
Collapse
|
41
|
Ling S, You Z, Li Y, Zhang J, Zhao S, He Y, Chen X. The role of γδ T17 cells in cardiovascular disease. J Leukoc Biol 2022; 112:1649-1661. [PMID: 36073777 DOI: 10.1002/jlb.3mr0822-761rr] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2022] [Revised: 06/23/2022] [Accepted: 08/03/2022] [Indexed: 01/04/2023] Open
Abstract
Due to the ability of γδ T cells to bridge adaptive and innate immunity, γδ T cells can respond to a variety of molecular cues and acquire the ability to induce a variety of cytokines such as IL-17 family, IFN-γ, IL-4, and IL-10. IL-17+ γδ T cells (γδ T17 cells) populations have recently received considerable interest as they are the major early source of IL-17A in many immune response models. However, the exact mechanism of γδ T17 cells is still poorly understood, especially in the context of cardiovascular disease (CVD). CVD is the leading cause of death in the world, and it tends to be younger. Here, we offer a review of the cardiovascular inflammatory and immune functions of γδ T17 cells in order to understand their role in CVD, which may be the key to developing new clinical applications.
Collapse
Affiliation(s)
- Shaoxue Ling
- College of Pharmaceutical Engineering of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, 10 Poyanghu Road, Tianjin, 301617, China
| | - Zonghao You
- College of Pharmaceutical Engineering of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, 10 Poyanghu Road, Tianjin, 301617, China
| | - Yang Li
- School of Chinese Materia Medica, Tianjin University of Traditional Chinese Medicine, 10 Poyanghu Road, Tianjin, 301617, China
| | - Jian Zhang
- School of Chinese Materia Medica, Tianjin University of Traditional Chinese Medicine, 10 Poyanghu Road, Tianjin, 301617, China
| | - Shuwu Zhao
- School of Intergrative Medicine, Tianjin University of Traditional Chinese Medicine, 10 Poyanghu Road, Tianjin, 301617, China
| | - Yongzhi He
- College of Pharmaceutical Engineering of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, 10 Poyanghu Road, Tianjin, 301617, China
| | - Xi Chen
- College of Pharmaceutical Engineering of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, 10 Poyanghu Road, Tianjin, 301617, China
| |
Collapse
|
42
|
Zelenka T, Klonizakis A, Tsoukatou D, Papamatheakis DA, Franzenburg S, Tzerpos P, Tzonevrakis IR, Papadogkonas G, Kapsetaki M, Nikolaou C, Plewczynski D, Spilianakis C. The 3D enhancer network of the developing T cell genome is shaped by SATB1. Nat Commun 2022; 13:6954. [PMID: 36376298 PMCID: PMC9663569 DOI: 10.1038/s41467-022-34345-y] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2021] [Accepted: 10/24/2022] [Indexed: 11/16/2022] Open
Abstract
Mechanisms of tissue-specific gene expression regulation via 3D genome organization are poorly understood. Here we uncover the regulatory chromatin network of developing T cells and identify SATB1, a tissue-specific genome organizer, enriched at the anchors of promoter-enhancer loops. We have generated a T-cell specific Satb1 conditional knockout mouse which allows us to infer the molecular mechanisms responsible for the deregulation of its immune system. H3K27ac HiChIP and Hi-C experiments indicate that SATB1-dependent promoter-enhancer loops regulate expression of master regulator genes (such as Bcl6), the T cell receptor locus and adhesion molecule genes, collectively being critical for cell lineage specification and immune system homeostasis. SATB1-dependent regulatory chromatin loops represent a more refined layer of genome organization built upon a high-order scaffold provided by CTCF and other factors. Overall, our findings unravel the function of a tissue-specific factor that controls transcription programs, via spatial chromatin arrangements complementary to the chromatin structure imposed by ubiquitously expressed genome organizers.
Collapse
Affiliation(s)
- Tomas Zelenka
- Department of Biology, University of Crete, Heraklion, Crete, Greece
- Institute of Molecular Biology and Biotechnology-Foundation for Research and Technology Hellas, Heraklion, Crete, Greece
- Department of Immunology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL, USA
| | | | - Despina Tsoukatou
- Institute of Molecular Biology and Biotechnology-Foundation for Research and Technology Hellas, Heraklion, Crete, Greece
| | - Dionysios-Alexandros Papamatheakis
- Department of Biology, University of Crete, Heraklion, Crete, Greece
- Institute of Molecular Biology and Biotechnology-Foundation for Research and Technology Hellas, Heraklion, Crete, Greece
| | | | - Petros Tzerpos
- Department of Biology, University of Crete, Heraklion, Crete, Greece
- Department of Biochemistry and Molecular Biology, Faculty of Medicine, University of Debrecen, Debrecen, HU-4032, Hungary
| | | | - George Papadogkonas
- Department of Biology, University of Crete, Heraklion, Crete, Greece
- Institute of Molecular Biology and Biotechnology-Foundation for Research and Technology Hellas, Heraklion, Crete, Greece
| | - Manouela Kapsetaki
- Institute of Molecular Biology and Biotechnology-Foundation for Research and Technology Hellas, Heraklion, Crete, Greece
| | - Christoforos Nikolaou
- Department of Biology, University of Crete, Heraklion, Crete, Greece
- Institute of Molecular Biology and Biotechnology-Foundation for Research and Technology Hellas, Heraklion, Crete, Greece
- Institute for Bioinnovation, Biomedical Sciences Research Centre "Alexander Fleming", 16672, Vari, Greece
| | - Dariusz Plewczynski
- Laboratory of Bioinformatics and Computational Genomics, Faculty of Mathematics and Information Science, Warsaw University of Technology, Warsaw, Poland
- Laboratory of Functional and Structural Genomics, Centre of New Technologies, University of Warsaw, Warsaw, Poland
| | - Charalampos Spilianakis
- Department of Biology, University of Crete, Heraklion, Crete, Greece.
- Institute of Molecular Biology and Biotechnology-Foundation for Research and Technology Hellas, Heraklion, Crete, Greece.
| |
Collapse
|
43
|
Boehme L, Roels J, Taghon T. Development of γδ T cells in the thymus - A human perspective. Semin Immunol 2022; 61-64:101662. [PMID: 36374779 DOI: 10.1016/j.smim.2022.101662] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/26/2022] [Accepted: 10/05/2022] [Indexed: 12/14/2022]
Abstract
γδ T cells are increasingly emerging as crucial immune regulators that can take on innate and adaptive roles in the defence against pathogens. Although they arise within the thymus from the same hematopoietic precursors as conventional αβ T cells, the development of γδ T cells is less well understood. In this review, we focus on summarising the current state of knowledge about the cellular and molecular processes involved in the generation of γδ T cells in human.
Collapse
Affiliation(s)
- Lena Boehme
- Department of Diagnostic Sciences, Ghent University, Ghent, Belgium
| | - Juliette Roels
- Department of Diagnostic Sciences, Ghent University, Ghent, Belgium
| | - Tom Taghon
- Department of Diagnostic Sciences, Ghent University, Ghent, Belgium; Cancer Research Institute Ghent (CRIG), Ghent, Belgium.
| |
Collapse
|
44
|
Darrigues J, Almeida V, Conti E, Ribot JC. The multisensory regulation of unconventional T cell homeostasis. Semin Immunol 2022; 61-64:101657. [PMID: 36370671 DOI: 10.1016/j.smim.2022.101657] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/14/2022] [Revised: 08/29/2022] [Accepted: 09/21/2022] [Indexed: 12/14/2022]
Abstract
Unconventional T cells typically group γδ T cells, invariant Natural Killer T cells (NKT) and Mucosal Associated Invariant T (MAIT) cells. With their pre-activated status and biased tropism for non-lymphoid organs, they provide a rapid (innate-like) and efficient first line of defense against pathogens at strategical barrier sites, while they can also trigger chronic inflammation, and unexpectedly contribute to steady state physiology. Thus, a tight control of their homeostasis is critical to maintain tissue integrity. In this review, we discuss the recent advances of our understanding of the factors, from neuroimmune to inflammatory regulators, shaping the size and functional properties of unconventional T cell subsets in non-lymphoid organs. We present a general overview of the mechanisms common to these populations, while also acknowledging specific aspects of their diversity. We mainly focus on their maintenance at steady state and upon inflammation, highlighting some key unresolved issues and raising upcoming technical, fundamental and translational challenges.
Collapse
Affiliation(s)
- Julie Darrigues
- Instituto de Medicina Molecular João Lobo Antunes, Faculdade de Medicina, Universidade de Lisboa, Av. Professor Egas Moniz, 1649-028 Lisboa, Portugal.
| | - Vicente Almeida
- Instituto de Medicina Molecular João Lobo Antunes, Faculdade de Medicina, Universidade de Lisboa, Av. Professor Egas Moniz, 1649-028 Lisboa, Portugal
| | - Eller Conti
- Instituto de Medicina Molecular João Lobo Antunes, Faculdade de Medicina, Universidade de Lisboa, Av. Professor Egas Moniz, 1649-028 Lisboa, Portugal
| | - Julie C Ribot
- Instituto de Medicina Molecular João Lobo Antunes, Faculdade de Medicina, Universidade de Lisboa, Av. Professor Egas Moniz, 1649-028 Lisboa, Portugal.
| |
Collapse
|
45
|
Identification of distinct functional thymic programming of fetal and pediatric human γδ thymocytes via single-cell analysis. Nat Commun 2022; 13:5842. [PMID: 36195611 PMCID: PMC9532436 DOI: 10.1038/s41467-022-33488-2] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2021] [Accepted: 09/21/2022] [Indexed: 12/12/2022] Open
Abstract
Developmental thymic waves of innate-like and adaptive-like γδ T cells have been described, but the current understanding of γδ T cell development is mainly limited to mouse models. Here, we combine single cell (sc) RNA gene expression and sc γδ T cell receptor (TCR) sequencing on fetal and pediatric γδ thymocytes in order to understand the ontogeny of human γδ T cells. Mature fetal γδ thymocytes (both the Vγ9Vδ2 and nonVγ9Vδ2 subsets) are committed to either a type 1, a type 3 or a type 2-like effector fate displaying a wave-like pattern depending on gestation age, and are enriched for public CDR3 features upon maturation. Strikingly, these effector modules express different CDR3 sequences and follow distinct developmental trajectories. In contrast, the pediatric thymus generates only a small effector subset that is highly biased towards Vγ9Vδ2 TCR usage and shows a mixed type 1/type 3 effector profile. Thus, our combined dataset of gene expression and detailed TCR information at the single-cell level identifies distinct functional thymic programming of γδ T cell immunity in human. Knowledge about the ontogeny of T cells in the thymus relies heavily on mouse studies because of difficulty to obtain human material. Here the authors perform a single cell analysis of thymocytes from human fetal and paediatric thymic samples to characterise the development of human γδ T cells in the thymus.
Collapse
|
46
|
Deng L, Harms A, Ravens S, Prinz I, Tan L. Systematic pattern analyses of Vδ2+ TCRs reveal that shared “public” Vδ2+ γδ T cell clones are a consequence of rearrangement bias and a higher expansion status. Front Immunol 2022; 13:960920. [DOI: 10.3389/fimmu.2022.960920] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2022] [Accepted: 09/06/2022] [Indexed: 11/13/2022] Open
Abstract
BackgroundVγ9Vδ2+ T cells are a major innate T cell subset in human peripheral blood. Their Vδ2+ VDJ-rearrangements are short and simple in the fetal thymus and gradually increase in diversity and CDR3 length along with development. So-called “public” versions of Vδ2+ TCRs are shared among individuals of all ages. However, it is unclear whether such frequently occurring “public” Vγ9Vδ2+ T cell clones are derived from the fetal thymus and whether they are fitter to proliferate and persist than infrequent “private” clones.MethodsShared “public” Vδ2+ TCRs were identified from Vδ2+ TCR-repertoires collected from 89 individuals, including newborns (cord blood), infants, and adults (peripheral blood). Distance matrices of Vδ2+ CDR3 were generated by TCRdist3 and then embedded into a UMAP for visualizing the heterogeneity of Vδ2+ TCRs.ResultsVδ2+ CDR3 distance matrix embedded by UMAP revealed that the heterogeneity of Vδ2+ TCRs is primarily determined by the J-usage and CDR3aa length, while age or publicity-specific motifs were not found. The most prevalent public Vδ2+ TCRs showed germline-like rearrangement with low N-insertions. Age-related features were also identified. Public Vδ2+TRDJ1 TCRs from cord blood showed higher N-insertions and longer CDR3 lengths. Synonymous codons resulting from VDJ rearrangement also contribute to the generation of public Vδ2+ TCRs. Each public TCR was always produced by multiple different transcripts, even with different D gene usage, and the publicity of Vδ2+ TCRs was positively associated with expansion status.ConclusionTo conclude, the heterogeneity of Vδ2+ TCRs is mainly determined by TRDJ-usage and the length of CDR3aa sequences. Public Vδ2+ TCRs result from germline-like rearrangement and synonymous codons, associated with a higher expansion status.
Collapse
|
47
|
Ritter K, Behrends J, Rückerl D, Hölscher A, Volz J, Prinz I, Hölscher C. High-Dose Mycobacterium tuberculosis H37rv Infection in IL-17A- and IL-17A/F-Deficient Mice. Cells 2022; 11:cells11182875. [PMID: 36139450 PMCID: PMC9496946 DOI: 10.3390/cells11182875] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2022] [Revised: 09/02/2022] [Accepted: 09/13/2022] [Indexed: 11/16/2022] Open
Abstract
During experimental tuberculosis (TB), interleukin (IL)-17A appears to be involved in the formation of lung granulomas, possibly through the attraction of neutrophils to the sites of infection. However, the protective impact of cytokine appears to depend on the degree of its induction. Hence, robust production of IL-17A in mice infected with the hypervirulent isolate Mycobacterium tuberculosis (Mtb) HN878 mediates protection, while the cytokine is dispensable for protective immune responses against low-dose infection with the less virulent strain H37rv. Here, we show that after experimental infection with high doses of Mtb H37rv, IL-17A-deficient (−/−) mice exhibited high susceptibility to the infection, which was mediated by the strong accumulation of neutrophils in the infected lung tissue. Accordingly, we observed nearly unrestricted bacterial replication within the neutrophils, indicating that they may serve as a survival niche for Mtb. By use of IL-17A/IL-17F-double-deficient mice, we demonstrated that the susceptibility in the absence of IL-17A is mediated by a compensatory expression of IL-17F, which, however, appeared not to be dependent on neutrophils. Together, our results illustrate the compensatory potential of the Th17-secreted cytokines IL-17A and IL-17F in the context of experimental TB and once again emphasize the detrimental effect of excessive neutrophil infiltration in response to Mtb.
Collapse
Affiliation(s)
- Kristina Ritter
- Infection Immunology, Research Center Borstel, D-23845 Borstel, Germany
| | - Jochen Behrends
- Fluorescence Cytometry Core Unit, Research Center Borstel, D-23845 Borstel, Germany
| | - Dominik Rückerl
- Lydia Becker Institute of Immunology and Inflammation, School of Biological Sciences, University of Manchester, Manchester M13 9PL, UK
| | | | - Johanna Volz
- Infection Immunology, Research Center Borstel, D-23845 Borstel, Germany
| | - Immo Prinz
- Center for Molecular Neurobiology Hamburg, Eppendorf University Medical Center, D-20246 Hamburg, Germany
| | - Christoph Hölscher
- Infection Immunology, Research Center Borstel, D-23845 Borstel, Germany
- Thematic Translational Unit Tuberculosis, German Center for Infection Research (DZIF), D-38124 Braunschweig, Germany
- Correspondence:
| |
Collapse
|
48
|
Triiodothyronine-stimulated dendritic cell vaccination boosts antitumor immunity against murine colon cancer. Int Immunopharmacol 2022; 110:109016. [DOI: 10.1016/j.intimp.2022.109016] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2022] [Revised: 06/20/2022] [Accepted: 06/28/2022] [Indexed: 11/22/2022]
|
49
|
Ataide MA, Knöpper K, Cruz de Casas P, Ugur M, Eickhoff S, Zou M, Shaikh H, Trivedi A, Grafen A, Yang T, Prinz I, Ohlsen K, Gomez de Agüero M, Beilhack A, Huehn J, Gaya M, Saliba AE, Gasteiger G, Kastenmüller W. Lymphatic migration of unconventional T cells promotes site-specific immunity in distinct lymph nodes. Immunity 2022; 55:1813-1828.e9. [PMID: 36002023 DOI: 10.1016/j.immuni.2022.07.019] [Citation(s) in RCA: 35] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2022] [Revised: 06/06/2022] [Accepted: 07/27/2022] [Indexed: 12/31/2022]
Abstract
Lymphatic transport of molecules and migration of myeloid cells to lymph nodes (LNs) continuously inform lymphocytes on changes in drained tissues. Here, using LN transplantation, single-cell RNA-seq, spectral flow cytometry, and a transgenic mouse model for photolabeling, we showed that tissue-derived unconventional T cells (UTCs) migrate via the lymphatic route to locally draining LNs. As each tissue harbored a distinct spectrum of UTCs with locally adapted differentiation states and distinct T cell receptor repertoires, every draining LN was thus populated by a distinctive tissue-determined mix of these lymphocytes. By making use of single UTC lineage-deficient mouse models, we found that UTCs functionally cooperated in interconnected units and generated and shaped characteristic innate and adaptive immune responses that differed between LNs that drained distinct tissues. Lymphatic migration of UTCs is, therefore, a key determinant of site-specific immunity initiated in distinct LNs with potential implications for vaccination strategies and immunotherapeutic approaches.
Collapse
Affiliation(s)
- Marco A Ataide
- Würzburg Institute of Systems Immunology, Max Planck Research Group at the Julius-Maximilians-Universität Würzburg, 97078 Würzburg, Germany.
| | - Konrad Knöpper
- Würzburg Institute of Systems Immunology, Max Planck Research Group at the Julius-Maximilians-Universität Würzburg, 97078 Würzburg, Germany
| | - Paulina Cruz de Casas
- Würzburg Institute of Systems Immunology, Max Planck Research Group at the Julius-Maximilians-Universität Würzburg, 97078 Würzburg, Germany
| | - Milas Ugur
- Würzburg Institute of Systems Immunology, Max Planck Research Group at the Julius-Maximilians-Universität Würzburg, 97078 Würzburg, Germany
| | - Sarah Eickhoff
- Würzburg Institute of Systems Immunology, Max Planck Research Group at the Julius-Maximilians-Universität Würzburg, 97078 Würzburg, Germany
| | - Mangge Zou
- Experimental Immunology, Helmholtz Centre for Infection Research, 38124 Braunschweig, Germany
| | - Haroon Shaikh
- Department of Medicine II and Pediatrics, Würzburg University Hospital, ZEMM, 97078 Würzburg, Germany
| | - Apurwa Trivedi
- Centre d'Immunologie de Marseille-Luminy (CIML), Department of Immunology, 13288 Marseille, France
| | - Anika Grafen
- Würzburg Institute of Systems Immunology, Max Planck Research Group at the Julius-Maximilians-Universität Würzburg, 97078 Würzburg, Germany
| | - Tao Yang
- Institute of Immunology, Hannover Medical School, 30625 Hannover, Germany
| | - Immo Prinz
- Institute of Systems Immunology, University Medical Center Hamburg-Eppendorf, 20251 Hamburg, Germany
| | - Knut Ohlsen
- Institute for Molecular Infection Biology (IMIB), 97078 Würzburg, Germany
| | - Mercedes Gomez de Agüero
- Würzburg Institute of Systems Immunology, Max Planck Research Group at the Julius-Maximilians-Universität Würzburg, 97078 Würzburg, Germany
| | - Andreas Beilhack
- Department of Medicine II and Pediatrics, Würzburg University Hospital, ZEMM, 97078 Würzburg, Germany
| | - Jochen Huehn
- Experimental Immunology, Helmholtz Centre for Infection Research, 38124 Braunschweig, Germany; Cluster of Excellence RESIST (EXC 2155), Hannover Medical School, 30625 Hannover, Germany
| | - Mauro Gaya
- Centre d'Immunologie de Marseille-Luminy (CIML), Department of Immunology, 13288 Marseille, France
| | - Antoine-Emmanuel Saliba
- Helmholtz Institute for RNA-Based Infection Research (HIRI), Helmholtz-Center for Infection Research (HZI), 97078 Würzburg, Germany
| | - Georg Gasteiger
- Würzburg Institute of Systems Immunology, Max Planck Research Group at the Julius-Maximilians-Universität Würzburg, 97078 Würzburg, Germany
| | - Wolfgang Kastenmüller
- Würzburg Institute of Systems Immunology, Max Planck Research Group at the Julius-Maximilians-Universität Würzburg, 97078 Würzburg, Germany.
| |
Collapse
|
50
|
Ding C, Xu H, Yu Z, Roulis M, Qu R, Zhou J, Oh J, Crawford J, Gao Y, Jackson R, Sefik E, Li S, Wei Z, Skadow M, Yin Z, Ouyang X, Wang L, Zou Q, Su B, Hu W, Flavell RA, Li HB. RNA m 6A demethylase ALKBH5 regulates the development of γδ T cells. Proc Natl Acad Sci U S A 2022; 119:e2203318119. [PMID: 35939687 PMCID: PMC9388086 DOI: 10.1073/pnas.2203318119] [Citation(s) in RCA: 31] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
γδ T cells are an abundant T cell population at the mucosa and are important in providing immune surveillance as well as maintaining tissue homeostasis. However, despite γδ T cells' origin in the thymus, detailed mechanisms regulating γδ T cell development remain poorly understood. N6-methyladenosine (m6A) represents one of the most common posttranscriptional modifications of messenger RNA (mRNA) in mammalian cells, but whether it plays a role in γδ T cell biology is still unclear. Here, we show that depletion of the m6A demethylase ALKBH5 in lymphocytes specifically induces an expansion of γδ T cells, which confers enhanced protection against gastrointestinal Salmonella typhimurium infection. Mechanistically, loss of ALKBH5 favors the development of γδ T cell precursors by increasing the abundance of m6A RNA modification in thymocytes, which further reduces the expression of several target genes including Notch signaling components Jagged1 and Notch2. As a result, impairment of Jagged1/Notch2 signaling contributes to enhanced proliferation and differentiation of γδ T cell precursors, leading to an expanded mature γδ T cell repertoire. Taken together, our results indicate a checkpoint role of ALKBH5 and m6A modification in the regulation of γδ T cell early development.
Collapse
Affiliation(s)
- Chenbo Ding
- aDepartment of Geriatrics, Center for Immune-Related Diseases, Shanghai Institute of Immunology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
- bShanghai Jiao Tong University School of Medicine–Yale University Institute for Immune Metabolism, Shanghai 200025, China
| | - Hao Xu
- cDepartment of Immunobiology, Yale University School of Medicine, New Haven, CT 06520
| | - Zhibin Yu
- aDepartment of Geriatrics, Center for Immune-Related Diseases, Shanghai Institute of Immunology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
- bShanghai Jiao Tong University School of Medicine–Yale University Institute for Immune Metabolism, Shanghai 200025, China
| | - Manolis Roulis
- cDepartment of Immunobiology, Yale University School of Medicine, New Haven, CT 06520
| | - Rihao Qu
- cDepartment of Immunobiology, Yale University School of Medicine, New Haven, CT 06520
- dProgram of Computational Biology and Bioinformatics, Yale University, New Haven, CT 06520
- eDepartment of Pathology, Yale University School of Medicine, New Haven, CT 06510
| | - Jing Zhou
- aDepartment of Geriatrics, Center for Immune-Related Diseases, Shanghai Institute of Immunology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
- bShanghai Jiao Tong University School of Medicine–Yale University Institute for Immune Metabolism, Shanghai 200025, China
| | - Joonseok Oh
- fDepartment of Chemistry, Yale University, New Haven, CT 06520
- gChemical Biology Institute, Yale University, West Haven, CT 06516
| | - Jason Crawford
- fDepartment of Chemistry, Yale University, New Haven, CT 06520
- gChemical Biology Institute, Yale University, West Haven, CT 06516
- hDepartment of Microbial Pathogenesis, Yale University School of Medicine, New Haven, CT 06520
| | - Yimeng Gao
- iSection of Hematology, Yale Cancer Center and Department of Internal Medicine, Yale University School of Medicine, New Haven, CT 06520
- jYale Stem Cell Center, Yale University School of Medicine, New Haven, CT 06520
- kYale RNA Center, Yale University School of Medicine, New Haven, CT 06520
| | - Ruaidhrí Jackson
- cDepartment of Immunobiology, Yale University School of Medicine, New Haven, CT 06520
| | - Esen Sefik
- cDepartment of Immunobiology, Yale University School of Medicine, New Haven, CT 06520
| | - Simiao Li
- cDepartment of Immunobiology, Yale University School of Medicine, New Haven, CT 06520
| | - Zheng Wei
- cDepartment of Immunobiology, Yale University School of Medicine, New Haven, CT 06520
| | - Mathias Skadow
- cDepartment of Immunobiology, Yale University School of Medicine, New Haven, CT 06520
| | - Zhinan Yin
- lZhuhai Precision Medical Center, Zhuhai People’s Hospital, Jinan University, Zhuhai 519000, Guangdong, China
- mBiomedical Translational Research Institute, Faculty of Medical Science, Jinan University, Guangzhou 510632, Guangdong, China
| | - Xinshou Ouyang
- nSection of Digestive Diseases, Department of Internal Medicine, Yale University School of Medicine, New Haven, CT 06520
| | - Lei Wang
- aDepartment of Geriatrics, Center for Immune-Related Diseases, Shanghai Institute of Immunology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Qiang Zou
- aDepartment of Geriatrics, Center for Immune-Related Diseases, Shanghai Institute of Immunology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Bing Su
- aDepartment of Geriatrics, Center for Immune-Related Diseases, Shanghai Institute of Immunology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
- bShanghai Jiao Tong University School of Medicine–Yale University Institute for Immune Metabolism, Shanghai 200025, China
| | - Weiguo Hu
- aDepartment of Geriatrics, Center for Immune-Related Diseases, Shanghai Institute of Immunology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
- 2To whom correspondence may be addressed. , , or
| | - Richard A. Flavell
- cDepartment of Immunobiology, Yale University School of Medicine, New Haven, CT 06520
- oHHMI, Yale University School of Medicine, New Haven, CT 06520
- 2To whom correspondence may be addressed. , , or
| | - Hua-Bing Li
- aDepartment of Geriatrics, Center for Immune-Related Diseases, Shanghai Institute of Immunology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
- bShanghai Jiao Tong University School of Medicine–Yale University Institute for Immune Metabolism, Shanghai 200025, China
- 2To whom correspondence may be addressed. , , or
| |
Collapse
|