1
|
Singh M, Louie RHY, Samir J, Field MA, Milthorpe C, Adikari T, Mackie J, Roper E, Faulks M, Jackson KJL, Calcino A, Hardy MY, Blombery P, Amos TG, Deveson IW, Wende HV, Floor SN, Read SA, Shek D, Guerin A, Ma CS, Tangye SG, Di Sabatino A, Lenti MV, Pasini A, Ciccocioppo R, Ahlenstiel G, Suan D, Tye-Din JA, Goodnow CC, Luciani F. Expanded T cell clones with lymphoma driver somatic mutations accumulate in refractory celiac disease. Sci Transl Med 2025; 17:eadp6812. [PMID: 40367192 DOI: 10.1126/scitranslmed.adp6812] [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: 04/07/2024] [Accepted: 03/31/2025] [Indexed: 05/16/2025]
Abstract
Intestinal inflammation continues in a subset of patients with celiac disease despite a gluten-free diet. Here, by applying multi-omic single-cell analysis to duodenal biopsies, we found that low-grade malignancies with lymphoma driver mutations in patients with refractory celiac disease type 2 (RCD2) are comprised by surface CD3-negative (sCD3-) lymphocytes stalled at an innate lymphoid cell (ILC)-progenitor T cell stage undergoing extensive TRA, TRB, and TRD TCR recombination. In people with refractory celiac disease type 1 (RCD1), a disease currently lacking explanation, we identified sCD3+ T cells with lymphoma driver mutations in 6 of 10 individuals with RCD1 and in one of the patients with active, recently diagnosed celiac disease. Furthermore, the mutant T cells formed large TCRαβ clones and displayed inflammatory and cytotoxic molecular profiles. Thus, accumulation of lymphoma driver-mutated T cells and sCD3- progenitors may contribute to chronic, nonresponsive celiac disease.
Collapse
Affiliation(s)
- Mandeep Singh
- Garvan Institute of Medical Research, Darlinghurst, NSW 2010, Australia
- School of Clinical Medicine, Faculty of Medicine & Health, UNSW Sydney, Sydney, NSW 2052, Australia
| | - Raymond H Y Louie
- School of Computer Science and Engineering, UNSW Sydney, Sydney, NSW 2052, Australia
- School of Medical Sciences, UNSW Sydney, Sydney, NSW 2052, Australia
| | - Jerome Samir
- School of Medical Sciences, UNSW Sydney, Sydney, NSW 2052, Australia
| | - Matthew A Field
- Garvan Institute of Medical Research, Darlinghurst, NSW 2010, Australia
- Australian Institute of Tropical Health and Medicine and Centre for Tropical Bioinformatics and Molecular Biology, James Cook University, Smithfield, QLD 4878, Australia
| | - Claire Milthorpe
- Garvan Institute of Medical Research, Darlinghurst, NSW 2010, Australia
| | - Thiruni Adikari
- School of Medical Sciences, UNSW Sydney, Sydney, NSW 2052, Australia
| | - Joseph Mackie
- Garvan Institute of Medical Research, Darlinghurst, NSW 2010, Australia
- School of Clinical Medicine, Faculty of Medicine & Health, UNSW Sydney, Sydney, NSW 2052, Australia
| | - Ellise Roper
- Garvan Institute of Medical Research, Darlinghurst, NSW 2010, Australia
| | - Megan Faulks
- Garvan Institute of Medical Research, Darlinghurst, NSW 2010, Australia
| | | | - Andrew Calcino
- Australian Institute of Tropical Health and Medicine and Centre for Tropical Bioinformatics and Molecular Biology, James Cook University, Smithfield, QLD 4878, Australia
| | - Melinda Y Hardy
- Walter and Eliza Hall Institute of Medical Research, Parkville, VIC 3052, Australia
- Department of Medical Biology, University of Melbourne, Parkville, VIC 3010, Australia
| | - Piers Blombery
- Peter MacCallum Cancer Centre and University of Melbourne, Melbourne, VIC 3000, Australia
- University of Melbourne, Melbourne, VIC 3010, Australia
| | - Timothy G Amos
- Garvan Institute of Medical Research, Darlinghurst, NSW 2010, Australia
| | - Ira W Deveson
- Garvan Institute of Medical Research, Darlinghurst, NSW 2010, Australia
- School of Clinical Medicine, Faculty of Medicine & Health, UNSW Sydney, Sydney, NSW 2052, Australia
| | - Helen Vander Wende
- Department of Cell and Tissue Biology, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Stephen N Floor
- Department of Cell and Tissue Biology, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Scott A Read
- Westmead Institute for Medical Research, University of Sydney, Westmead, NSW 2145, Australia
- Blacktown Medical School, Western Sydney University, Blacktown, NSW 2148, Australia
- Blacktown Hospital, Blacktown, NSW 2148, Australia
| | - Dmitri Shek
- Westmead Institute for Medical Research, University of Sydney, Westmead, NSW 2145, Australia
- Blacktown Medical School, Western Sydney University, Blacktown, NSW 2148, Australia
- Blacktown Hospital, Blacktown, NSW 2148, Australia
| | - Antoine Guerin
- Garvan Institute of Medical Research, Darlinghurst, NSW 2010, Australia
- School of Clinical Medicine, Faculty of Medicine & Health, UNSW Sydney, Sydney, NSW 2052, Australia
| | - Cindy S Ma
- Garvan Institute of Medical Research, Darlinghurst, NSW 2010, Australia
- School of Clinical Medicine, Faculty of Medicine & Health, UNSW Sydney, Sydney, NSW 2052, Australia
| | - Stuart G Tangye
- Garvan Institute of Medical Research, Darlinghurst, NSW 2010, Australia
- School of Clinical Medicine, Faculty of Medicine & Health, UNSW Sydney, Sydney, NSW 2052, Australia
| | - Antonio Di Sabatino
- Department of Internal Medicine and Medical Therapeutics, University of Pavia, Pavia 27100, Italy
- First Department of Internal Medicine, Fondazione IRCCS Policlinico San Matteo, Pavia 27100, Italy
| | - Marco V Lenti
- Department of Internal Medicine and Medical Therapeutics, University of Pavia, Pavia 27100, Italy
- First Department of Internal Medicine, Fondazione IRCCS Policlinico San Matteo, Pavia 27100, Italy
| | - Alessandra Pasini
- Department of Internal Medicine and Medical Therapeutics, University of Pavia, Pavia 27100, Italy
- First Department of Internal Medicine, Fondazione IRCCS Policlinico San Matteo, Pavia 27100, Italy
| | - Rachele Ciccocioppo
- Gastroenterology Unit, Department of Medicine, University of Verona and AOUI Verona, Policlinico GB Rossi, Verona 37134, Italy
| | - Golo Ahlenstiel
- Westmead Institute for Medical Research, University of Sydney, Westmead, NSW 2145, Australia
- Blacktown Medical School, Western Sydney University, Blacktown, NSW 2148, Australia
- Blacktown Hospital, Blacktown, NSW 2148, Australia
| | - Dan Suan
- Garvan Institute of Medical Research, Darlinghurst, NSW 2010, Australia
- School of Clinical Medicine, Faculty of Medicine & Health, UNSW Sydney, Sydney, NSW 2052, Australia
| | - Jason A Tye-Din
- Walter and Eliza Hall Institute of Medical Research, Parkville, VIC 3052, Australia
- Gastroenterology Department, Royal Melbourne Hospital, Parkville, VIC 3050, Australia
| | - Christopher C Goodnow
- Garvan Institute of Medical Research, Darlinghurst, NSW 2010, Australia
- Cellular Genomics Futures Institute and School of Biomedical Sciences, UNSW Sydney, Sydney, NSW 2052, Australia
| | - Fabio Luciani
- Garvan Institute of Medical Research, Darlinghurst, NSW 2010, Australia
- School of Medical Sciences, UNSW Sydney, Sydney, NSW 2052, Australia
| |
Collapse
|
2
|
Zhou DY, Bao CF, Zhou G. Intraepithelial lymphocytes in human oral diseases. Front Immunol 2025; 16:1597088. [PMID: 40406112 PMCID: PMC12095017 DOI: 10.3389/fimmu.2025.1597088] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2025] [Accepted: 04/14/2025] [Indexed: 05/26/2025] Open
Abstract
Objective As a distinctive subset of T cells, intraepithelial lymphocytes (IELs) are found in the epithelium of mucosal barrier and serve as the primary defenders of the intestinal mucosal immune system. IELs exhibit phenotypic and functional diversity with high expression of activated marker molecules, tissue-homing integrins, NK cell receptors, cytotoxic T cell-related molecules, and cytokines. Meanwhile, IELs demonstrate differentiation plasticity, antigen recognition diversity, self-reactivity, and rapid "memory" effect, which enable them to play a crucial role in regulating responses, maintaining mucosal barriers, promoting immune tolerance, and providing resistance to infections. In addition, IELs have been explored in autoimmune diseases, inflammatory diseases, and cancers. However, the specific involvement and underlying mechanisms of IELs in oral diseases have not been systematically discussed. Methods A systematic literature review was conducted using the PubMed/MEDLINE databases to identify and analyze relevant literatures on the roles of IELs in oral diseases. Results The literature review revealed the characteristics of IELs and emphasized the potential roles of IELs in the pathogenesis of oral lichen planus, oral cancers, periodontal diseases, graft-versus-host disease, and primary Sjogren's syndrome. Conclusion This review mainly focuses on the involvement of IELs in oral diseases, with a particular emphasis on the main functions and underlying mechanisms by which IELs influence the pathogenesis and progression of these conditions.
Collapse
Affiliation(s)
- Dong-Yang Zhou
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Wuhan, China
| | - Chao-Fan Bao
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Wuhan, China
| | - Gang Zhou
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Wuhan, China
- Department of Oral Medicine, School and Hospital of Stomatology, Wuhan University, Wuhan, China
| |
Collapse
|
3
|
FitzPatrick MEB, Antanaviciute A, Dunstan M, Künnapuu K, Trzupek D, Provine NM, Dooley K, Zhang JY, Irwin SL, Garner LC, Pernes JI, Ferreira RC, Sasson SC, Aschenbrenner D, Agarwal D, Rodrigues A, Howarth L, Brain O, Ruane D, Soilleux E, Teichmann SA, Dendrou CA, Simmons A, Uhlig HH, Todd JA, Klenerman P. Immune-epithelial-stromal networks define the cellular ecosystem of the small intestine in celiac disease. Nat Immunol 2025:10.1038/s41590-025-02146-2. [PMID: 40328997 DOI: 10.1038/s41590-025-02146-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2023] [Accepted: 03/25/2025] [Indexed: 05/08/2025]
Abstract
The immune-epithelial-stromal interactions underpinning intestinal damage in celiac disease (CD) are incompletely understood. To address this, we performed single-cell transcriptomics (RNA sequencing; 86,442 immune, parenchymal and epithelial cells; 35 participants) and spatial transcriptomics (20 participants) on CD intestinal biopsy samples. Here we show that in CD, epithelial populations shifted toward a progenitor state, with interferon-driven transcriptional responses, and perturbation of secretory and enteroendocrine populations. Mucosal T cells showed numeric and functional changes in regulatory and follicular helper-like CD4+ T cells, intraepithelial lymphocytes, CD8+ and γδ T cell subsets, with skewed T cell antigen receptor repertoires. Mucosal changes remained detectable despite treatment, representing a persistent immune-epithelial 'scar'. Spatial transcriptomics defined transcriptional niches beyond those captured in conventional histological scores, including CD-specific lymphoid aggregates containing T cell-B cell interactions. Receptor-ligand spatial analyses integrated with disease susceptibility gene expression defined networks of altered chemokine and morphogen signaling, and provide potential therapeutic targets for CD prevention and treatment.
Collapse
Affiliation(s)
- Michael E B FitzPatrick
- Translational Gastroenterology and Liver Unit, Nuffield Department of Medicine, University of Oxford, John Radcliffe Hospital, Oxford, UK.
| | - Agne Antanaviciute
- MRC Translational Immune Discovery Unit, Weatherall Institute of Molecular Medicine, John Radcliffe Hospital, Oxford, UK
- MRC WIMM Centre for Computational Biology, Weatherall Institute of Molecular Medicine, John Radcliffe Hospital, Oxford, UK
| | - Melanie Dunstan
- Translational Gastroenterology and Liver Unit, Nuffield Department of Medicine, University of Oxford, John Radcliffe Hospital, Oxford, UK
- Centre for Human Genetics, Nuffield Department of Medicine, NIHR Biomedical Research Centres, University of Oxford, Oxford, UK
| | - Karolina Künnapuu
- Translational Gastroenterology and Liver Unit, Nuffield Department of Medicine, University of Oxford, John Radcliffe Hospital, Oxford, UK
| | - Dominik Trzupek
- Centre for Human Genetics, Nuffield Department of Medicine, NIHR Biomedical Research Centres, University of Oxford, Oxford, UK
| | - Nicholas M Provine
- Translational Gastroenterology and Liver Unit, Nuffield Department of Medicine, University of Oxford, John Radcliffe Hospital, Oxford, UK
- Centre for Human Genetics, Nuffield Department of Medicine, NIHR Biomedical Research Centres, University of Oxford, Oxford, UK
- Pandemic Sciences Institute, Nuffield Department of Medicine, University of Oxford, Oxford, UK
| | - Kyla Dooley
- Centre for Human Genetics, Nuffield Department of Medicine, NIHR Biomedical Research Centres, University of Oxford, Oxford, UK
- Pandemic Sciences Institute, Nuffield Department of Medicine, University of Oxford, Oxford, UK
| | - Jia-Yuan Zhang
- MRC WIMM Centre for Computational Biology, Weatherall Institute of Molecular Medicine, John Radcliffe Hospital, Oxford, UK
| | - Sophie L Irwin
- Division of Infectious Diseases, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, USA
| | - Lucy C Garner
- Translational Gastroenterology and Liver Unit, Nuffield Department of Medicine, University of Oxford, John Radcliffe Hospital, Oxford, UK
| | - Jane I Pernes
- Centre for Human Genetics, Nuffield Department of Medicine, NIHR Biomedical Research Centres, University of Oxford, Oxford, UK
| | - Ricardo C Ferreira
- Centre for Human Genetics, Nuffield Department of Medicine, NIHR Biomedical Research Centres, University of Oxford, Oxford, UK
| | - Sarah C Sasson
- Kirby Institute, University of New South Wales, Sydney, New South Wales, Australia
| | | | - Devika Agarwal
- Kennedy Institute of Rheumatology, University of Oxford, Oxford, UK
| | - Astor Rodrigues
- University Children's Hospital, John Radcliffe Hospital, Oxford, UK
| | - Lucy Howarth
- University Children's Hospital, John Radcliffe Hospital, Oxford, UK
| | - Oliver Brain
- Translational Gastroenterology and Liver Unit, Nuffield Department of Medicine, University of Oxford, John Radcliffe Hospital, Oxford, UK
| | - Darren Ruane
- Janssen Research & Development, Immunology Translational Sciences and Medicine, La Jolla, CA, USA
| | | | | | - Calliope A Dendrou
- Centre for Human Genetics, Nuffield Department of Medicine, NIHR Biomedical Research Centres, University of Oxford, Oxford, UK
- Kennedy Institute of Rheumatology, University of Oxford, Oxford, UK
- NIHR Oxford Biomedical Research Centre, University of Oxford, Oxford, UK
| | - Alison Simmons
- Translational Gastroenterology and Liver Unit, Nuffield Department of Medicine, University of Oxford, John Radcliffe Hospital, Oxford, UK
- MRC Translational Immune Discovery Unit, Weatherall Institute of Molecular Medicine, John Radcliffe Hospital, Oxford, UK
| | - Holm H Uhlig
- Translational Gastroenterology and Liver Unit, Nuffield Department of Medicine, University of Oxford, John Radcliffe Hospital, Oxford, UK
- Centre for Human Genetics, Nuffield Department of Medicine, NIHR Biomedical Research Centres, University of Oxford, Oxford, UK
- University Children's Hospital, John Radcliffe Hospital, Oxford, UK
- NIHR Oxford Biomedical Research Centre, University of Oxford, Oxford, UK
| | - John A Todd
- Centre for Human Genetics, Nuffield Department of Medicine, NIHR Biomedical Research Centres, University of Oxford, Oxford, UK
| | - Paul Klenerman
- Translational Gastroenterology and Liver Unit, Nuffield Department of Medicine, University of Oxford, John Radcliffe Hospital, Oxford, UK
- NIHR Oxford Biomedical Research Centre, University of Oxford, Oxford, UK
- Peter Medawar Building for Pathogen Research, Nuffield Department of Medicine, University of Oxford, Oxford, UK
| |
Collapse
|
4
|
Santonicola A, Soldaini C, Ciacci C. New therapies in celiac disease. Curr Opin Gastroenterol 2025; 41:124-131. [PMID: 39862215 PMCID: PMC11970589 DOI: 10.1097/mog.0000000000001080] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/27/2025]
Abstract
PURPOSE OF REVIEW Celiac disease (CeD) is a chronic autoimmune disorder of the small intestine triggered by gluten ingestion in genetically predisposed individuals. The cornerstone of CeD management remains a strict adherence to a lifelong gluten-free diet (GFD), although such a dietary restriction can lead to an altered quality of life and may not be easy to follow for many patients. These challenges highlighted the need for alternative therapies. This review aims to explore the latest advancements in these therapeutic avenues, emphasizing mechanisms of action, clinical efficacy, and safety profiles of drugs currently in advanced stages of clinical testing. RECENT FINDINGS Recent advances in the understanding of CeD pathophysiology have catalyzed the development of new therapeutic approaches, which include strategies to modify gluten processing in the gut, block gluten-triggered immune responses, or restore immune tolerance to gluten. SUMMARY While these therapies are not poised to take the place of GFD, they represent promising treatment alternatives that could enhance the quality of life and minimize long-term consequences in CeD patients. Further research, as well as phase III clinical trials of those already conducted, are needed to establish the feasibility of integrating these novel drugs in the clinical management of CeD.
Collapse
Affiliation(s)
- Antonella Santonicola
- Gastrointestinal Unit, Department of Medicine, Surgery and Dentistry "Scuola Medica Salernitana", University of Salerno, Salerno, Italy
| | | | | |
Collapse
|
5
|
Huysentruyt J, Steels W, Ruiz Pérez M, Verstraeten B, Divert T, Flies K, Lemeire K, Takahashi N, De Bruyn E, Joossens M, Brown AS, Lambrecht BN, Declercq W, Vanden Berghe T, Maelfait J, Vandenabeele P, Tougaard P. RIPK1 ablation in T cells results in spontaneous enteropathy and TNF-driven villus atrophy. EMBO Rep 2025; 26:2654-2682. [PMID: 40307618 PMCID: PMC12117051 DOI: 10.1038/s44319-025-00441-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2023] [Revised: 03/11/2025] [Accepted: 03/18/2025] [Indexed: 05/02/2025] Open
Abstract
RIPK1 is a crucial regulator of cell survival, inflammation and cell death. Human RIPK1 deficiency leads to early-onset intestinal inflammation and peripheral T cell imbalance, though its role in αβT cell-mediated intestinal homeostasis remains unclear. In this study, we demonstrate that mice with RIPK1 ablation in conventional αβT cells (Ripk1ΔCD4) developed a severe small intestinal pathology characterized by small intestinal elongation, crypt hyperplasia, and duodenum-specific villus atrophy. Using mixed bone marrow chimeras reveals a survival disadvantage of αβT cells compared to γδT cells in the small intestine. Broad-spectrum antibiotic treatment ameliorates crypt hyperplasia and prevents intestinal elongation, though villus atrophy persists. Conversely, crossing Ripk1ΔCD4 with TNF receptor 1 Tnfr1-/- knockout mice rescues villus atrophy but not intestinal elongation. Finally, combined ablation of Ripk1∆CD4 and Casp8∆CD4 fully rescues intestinal pathology, revealing that αβT cell apoptosis in Ripk1∆CD4 drives the enteropathy. These findings demonstrate that RIPK1-mediated survival of αβT cells is essential for proximal small intestinal homeostasis. In Ripk1∆CD4 mice, the imbalanced T cell compartment drives microbiome-mediated intestinal elongation and TNF-driven villus atrophy.
Collapse
Affiliation(s)
- Jelle Huysentruyt
- VIB-UGent Center for Inflammation Research, VIB, Ghent, Belgium
- Department of Biomedical Molecular Biology, Ghent University, Ghent, Belgium
| | - Wolf Steels
- VIB-UGent Center for Inflammation Research, VIB, Ghent, Belgium
- Department of Biomedical Molecular Biology, Ghent University, Ghent, Belgium
| | - Mario Ruiz Pérez
- VIB-UGent Center for Inflammation Research, VIB, Ghent, Belgium
- Department of Biomedical Molecular Biology, Ghent University, Ghent, Belgium
| | - Bruno Verstraeten
- VIB-UGent Center for Inflammation Research, VIB, Ghent, Belgium
- Department of Biomedical Molecular Biology, Ghent University, Ghent, Belgium
| | - Tatyana Divert
- VIB-UGent Center for Inflammation Research, VIB, Ghent, Belgium
- Department of Biomedical Molecular Biology, Ghent University, Ghent, Belgium
| | - Kayleigh Flies
- VIB-UGent Center for Inflammation Research, VIB, Ghent, Belgium
- Department of Biomedical Molecular Biology, Ghent University, Ghent, Belgium
| | - Kelly Lemeire
- VIB-UGent Center for Inflammation Research, VIB, Ghent, Belgium
- Department of Biomedical Molecular Biology, Ghent University, Ghent, Belgium
| | - Nozomi Takahashi
- VIB-UGent Center for Inflammation Research, VIB, Ghent, Belgium
- Department of Biomedical Molecular Biology, Ghent University, Ghent, Belgium
| | - Elke De Bruyn
- Laboratory of Microbiology, Department of Biochemistry and Microbiology, Faculty of Science, Ghent University, Ghent, Belgium
| | - Marie Joossens
- Laboratory of Microbiology, Department of Biochemistry and Microbiology, Faculty of Science, Ghent University, Ghent, Belgium
| | - Andrew S Brown
- VIB-UGent Center for Inflammation Research, VIB, Ghent, Belgium
- Department of Internal Medicine and Pediatrics, Ghent University, Ghent, Belgium
| | - Bart N Lambrecht
- VIB-UGent Center for Inflammation Research, VIB, Ghent, Belgium
- Department of Internal Medicine and Pediatrics, Ghent University, Ghent, Belgium
| | - Wim Declercq
- VIB-UGent Center for Inflammation Research, VIB, Ghent, Belgium
- Department of Biomedical Molecular Biology, Ghent University, Ghent, Belgium
| | - Tom Vanden Berghe
- VIB-UGent Center for Inflammation Research, VIB, Ghent, Belgium
- Department of Biomedical Molecular Biology, Ghent University, Ghent, Belgium
- Department of Biomedical Sciences, University of Antwerp, Antwerp, Belgium
| | - Jonathan Maelfait
- VIB-UGent Center for Inflammation Research, VIB, Ghent, Belgium
- Department of Biomedical Molecular Biology, Ghent University, Ghent, Belgium
| | - Peter Vandenabeele
- VIB-UGent Center for Inflammation Research, VIB, Ghent, Belgium.
- Department of Biomedical Molecular Biology, Ghent University, Ghent, Belgium.
| | - Peter Tougaard
- VIB-UGent Center for Inflammation Research, VIB, Ghent, Belgium
- Department of Biomedical Molecular Biology, Ghent University, Ghent, Belgium
| |
Collapse
|
6
|
Bender MJ, Lucas CL. Decoding Immunobiology Through Genetic Errors of Immunity. Annu Rev Immunol 2025; 43:285-311. [PMID: 39952637 DOI: 10.1146/annurev-immunol-082323-124920] [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] [Indexed: 02/17/2025]
Abstract
Throughout biology, the pursuit of genotype-phenotype relationships has provided foundational knowledge upon which new concepts and hypotheses are built. Genetic perturbation, whether occurring naturally or in experimental settings, is the mainstay of mechanistic dissection in biological systems. The unbiased discovery of causal genetic lesions via forward genetics in patients who have a rare disease elucidates a particularly impactful set of genotype-phenotype relationships. Here, we review the field of genetic errors of immunity, often termed inborn errors of immunity (IEIs), in a framework aimed at highlighting the powerful real-world immunology insights provided collectively and individually by these (approximately) 500 disorders. By conceptualizing essential immune functions in a model of the adaptive arsenal of rapid defenses, we organize IEIs based on immune circuits in which sensors, relays, and executioners cooperate to carry out pathogen clearance functions in an effective yet regulated manner. We review and discuss findings from IEIs that not only reinforce known immunology concepts but also offer surprising phenotypes, prompting an opportunity to refine our understanding of immune system function.
Collapse
Affiliation(s)
- Mackenzie J Bender
- Department of Immunobiology, Yale University, New Haven, Connecticut, USA;
| | - Carrie L Lucas
- Department of Immunobiology, Yale University, New Haven, Connecticut, USA;
| |
Collapse
|
7
|
Porret R, Alcaraz-Serna A, Peter B, Bernier-Latmani J, Cecchin R, Alfageme-Abello O, Ermellino L, Hafezi M, Pace E, du Pré MF, Lana E, Golshayan D, Velin D, Eyquem J, Tang Q, Petrova TV, Coukos G, Irving M, Pot C, Pantaleo G, Sollid LM, Muller YD. T cell receptor precision editing of regulatory T cells for celiac disease. Sci Transl Med 2025; 17:eadr8941. [PMID: 40106579 DOI: 10.1126/scitranslmed.adr8941] [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: 07/28/2024] [Accepted: 02/11/2025] [Indexed: 03/22/2025]
Abstract
Celiac disease, a gluten-sensitive enteropathy, demonstrates a strong human leukocyte antigen (HLA) association, with more than 90% of patients carrying the HLA-DQ2.5 allotype. No therapy is available for the condition except for a lifelong gluten-free diet. To address this gap, we explored the therapeutic potential of regulatory T cells (Tregs). By orthotopic replacement of T cell receptors (TCRs) through homology-directed repair, we generated gluten-reactive HLA-DQ2.5-restricted CD4+ engineered (e) T effector cells (Teffs) and eTregs and performed in vivo experiments in HLA-DQ2.5 transgenic mice. Of five validated TCRs, TCRs specific for two immunodominant and deamidated gluten epitopes (DQ2.5-glia-α1a and DQ2.5-glia-α2) were selected for further evaluation. CD4+ eTeffs exposed to deamidated gluten through oral gavage colocalized with dendritic and B cells in the Peyer's patches and gut-draining lymph nodes and specifically migrated to the intestine. The suppressive function of human eTregs correlated with high TCR functional activity. eTregs specific for one epitope suppressed the proliferation and gut migration of CD4+ eTeffs specific for the same and the other gluten epitope, demonstrating bystander suppression. The suppression requires an antigen-specific activation of eTregs given that polyclonal Tregs failed to suppress CD4+ eTeffs. These findings highlight the potential of gluten-reactive eTregs as a therapeutic for celiac disease.
Collapse
Affiliation(s)
- Raphaël Porret
- Division of Immunology and Allergy, Lausanne University Hospital and University of Lausanne, Lausanne CH-1005, Switzerland
| | - Ana Alcaraz-Serna
- Division of Immunology and Allergy, Lausanne University Hospital and University of Lausanne, Lausanne CH-1005, Switzerland
| | - Benjamin Peter
- Laboratories of Neuroimmunology, Service of Neurology and Neuroscience Research Center, Department of Clinical Neurosciences, Lausanne University Hospital and University of Lausanne, Lausanne CH-1005, Switzerland
| | - Jeremiah Bernier-Latmani
- Department of Oncology, Lausanne University Hospital and University of Lausanne, 1005 Lausanne, Switzerland
| | - Rebecca Cecchin
- Division of Immunology and Allergy, Lausanne University Hospital and University of Lausanne, Lausanne CH-1005, Switzerland
| | - Oscar Alfageme-Abello
- Division of Immunology and Allergy, Lausanne University Hospital and University of Lausanne, Lausanne CH-1005, Switzerland
| | - Laura Ermellino
- Division of Immunology and Allergy, Lausanne University Hospital and University of Lausanne, Lausanne CH-1005, Switzerland
| | - Morteza Hafezi
- Department of Oncology, Lausanne University Hospital and University of Lausanne, 1005 Lausanne, Switzerland
| | - Eleonora Pace
- Division of Immunology and Allergy, Lausanne University Hospital and University of Lausanne, Lausanne CH-1005, Switzerland
| | - M Fleur du Pré
- Norwegian Coeliac Disease Research Centre, Institute of Clinical Medicine, University of Oslo and Department of Immunology, Oslo University Hospital, Oslo NO-0424, Norway
| | - Erica Lana
- Division of Immunology and Allergy, Lausanne University Hospital and University of Lausanne, Lausanne CH-1005, Switzerland
| | - Dela Golshayan
- Transplantation Center, Department of Medicine, Lausanne University Hospital and University of Lausanne, Lausanne CH-1005, Switzerland
| | - Dominique Velin
- Service of Gastroenterology and Hepatology, Lausanne University Hospital and University of Lausanne, Lausanne CH-1005, Switzerland
| | - Justin Eyquem
- Department of Medicine, University of California San Francisco, San Francisco, CA 94143, USA
| | - Qizhi Tang
- Diabetes Center, University of California San Francisco, San Francisco, CA 94143, USA
| | - Tatiana V Petrova
- Department of Oncology, Lausanne University Hospital and University of Lausanne, 1005 Lausanne, Switzerland
| | - George Coukos
- Department of Oncology, Lausanne University Hospital and University of Lausanne, 1005 Lausanne, Switzerland
- Ludwig Institute for Cancer Research Lausanne Branch, 1066 Lausanne, Switzerland
| | - Melita Irving
- Department of Oncology, Lausanne University Hospital and University of Lausanne, 1005 Lausanne, Switzerland
- Ludwig Institute for Cancer Research Lausanne Branch, 1066 Lausanne, Switzerland
| | - Caroline Pot
- Laboratories of Neuroimmunology, Service of Neurology and Neuroscience Research Center, Department of Clinical Neurosciences, Lausanne University Hospital and University of Lausanne, Lausanne CH-1005, Switzerland
| | - Giuseppe Pantaleo
- Division of Immunology and Allergy, Lausanne University Hospital and University of Lausanne, Lausanne CH-1005, Switzerland
| | - Ludvig M Sollid
- Norwegian Coeliac Disease Research Centre, Institute of Clinical Medicine, University of Oslo and Department of Immunology, Oslo University Hospital, Oslo NO-0424, Norway
| | - Yannick D Muller
- Division of Immunology and Allergy, Lausanne University Hospital and University of Lausanne, Lausanne CH-1005, Switzerland
- Centre for Human Immunology Lausanne, Lausanne CH-1005, Switzerland
| |
Collapse
|
8
|
Kowalski MK, Domżał-Magrowska D, Małecka-Wojciesko E. Celiac Disease-Narrative Review on Progress in Celiac Disease. Foods 2025; 14:959. [PMID: 40231983 PMCID: PMC11941517 DOI: 10.3390/foods14060959] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2025] [Revised: 02/28/2025] [Accepted: 03/09/2025] [Indexed: 04/16/2025] Open
Abstract
Celiac disease is defined as a systemic immunological disorder caused by gluten (gliadin and other prolamin) in genetically predisposed individuals, who present with a variety of gluten-dependent symptoms, specific antibodies, the presence of the HLA DQ2 and DQ8 histocompatibility antigen, and enteropathy. Its prevalence, depending on the studied population and methodology, is estimated at 0.75-1.6% of the general population. During the complex immune reaction it induces, most cells involved in inflammatory processes are activated, which leads to the gradual atrophy of intestinal villi and the proliferation of enterocytes within intestinal crypts. The pathogenesis of celiac disease is extremely complicated and is still the subject of research. According to the current diagnostic guidelines, the following criteria should be taken into account: clinical symptoms (intestinal and extraintestinal), the presence of antibodies against tissue transglutaminase in the IgA class, the level of total IgA, and the presence of typical histological changes in duodenal biopsies. Diet-resistant celiac disease is one of the most important clinical challenges, causing serious complications. Currently, the basic method for treating celiac disease is an elimination diet (i.e., the exclusion of products that may contain gluten from the diet), however, new therapeutic strategies are still being sought, mainly based on supplementation with exogenous endopeptidases, modification of the immune response, and the use of zonulin inhibitors and transglutaminase 2 inhibitors. Clinical trials of new drugs are ongoing. The gradually expanding knowledge about the pathogenesis of celiac disease may allow for the development of new therapeutic strategies for both patients with a mild disease course, as well as those that are diet-resistant.
Collapse
Affiliation(s)
| | | | - Ewa Małecka-Wojciesko
- Department of Digestive Tract Diseases, Norbert Barlicki University Hospital, 90-153 Lodz, Poland; (M.K.K.); (D.D.-M.)
| |
Collapse
|
9
|
Galipeau HJ, Verdu EF. Is the microbiome important in celiac disease? J Can Assoc Gastroenterol 2025; 8:S51-S55. [PMID: 39990507 PMCID: PMC11842898 DOI: 10.1093/jcag/gwae047] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/25/2025] Open
Abstract
Celiac disease (CeD) is an autoimmune condition driven by gluten in genetically predisposed individuals. CeD is characterized by small intestinal villous atrophy but presents with a spectrum of gastrointestinal and systemic manifestations. Its only treatment is a strict, life-long adherence to a gluten-free diet, which is difficult to manage and does not always lead to symptomatic or mucosal recovery. About 40% of the population express the CeD-associated risk genes, but only 1%-2% of the worldwide population has CeD. This, along with the rising prevalence of CeD suggests other cofactors in disease pathogenesis. The gut microbiome has been implicated in CeD based on epidemiology studies and clinical associations. Mechanistic studies using relevant in vitro and in vivo preclinical models have begun to elucidate mechanisms through which microbes can influence CeD. Ultimately, a better understanding of these cofactors and their mechanisms will provide rationale intervention strategies and novel therapeutic targets to prevent or treat CeD.
Collapse
Affiliation(s)
- Heather J Galipeau
- Farncombe Family Digestive Health Research Institute, Department of Medicine, McMaster University, Hamilton L8K4K1, Canada
| | - Elena F Verdu
- Farncombe Family Digestive Health Research Institute, Department of Medicine, McMaster University, Hamilton L8K4K1, Canada
| |
Collapse
|
10
|
Lundgren S, Huuhtanen J, Keränen M, Feng X, Patel BA, Ryland GL, Fox LC, Bravo-Perez C, Clemente M, Kerr C, Walldin G, Dufva O, Zaimoku Y, Tuononen T, Myllymäki M, Ebeling F, Jokinen E, Heinonen M, Kasanen T, Klievink J, Lähteenmäki H, Jaatinen T, Kytölä S, Siitonen S, Dulau-Florea A, Braylan R, Heinäniemi M, Nakao S, Hellström-Lindberg E, Maciejewski JP, Blombery P, Young NS, Lähdesmäki H, Mustjoki S. Single-cell analysis of aplastic anemia reveals a convergence of NK and NK-like CD8 + T cells with a disease-associated TCR signature. Sci Transl Med 2025; 17:eadl6758. [PMID: 40009697 DOI: 10.1126/scitranslmed.adl6758] [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: 10/31/2023] [Revised: 08/16/2024] [Accepted: 11/15/2024] [Indexed: 02/28/2025]
Abstract
Immune aplastic anemia (AA) is a life-threatening bone marrow failure disorder driven by an autoimmune T cell attack against hematopoietic stem and progenitor cells (HSPCs). However, the exact autoantigen targets and role of other immune cells in the pathogenesis of AA are unknown. Here, we analyzed a cohort of 218 patients with AA using single-cell RNA and T cell receptor (TCR) αβ sequencing, TCRβ sequencing, flow cytometry, and plasma cytokine profiling. We identified natural killer (NK) cells and CD8+ terminally differentiated effector T (TEMRA) cells expressing NK receptors with AA-associated TCRβ motifs as the most dysregulated immune cell populations in AA bone marrow. Functional coculture experiments using primary HSPCs and immune cells showed that NK cells cannot kill HSPCs alone but may sensitize HSPCs to CD8+ T cell-mediated killing through production of interferons. Furthermore, HSPCs induced activation of T cell clones with CD8+ TEMRA NK-like phenotype in coculture. Our results reveal a convergent phenotype of innate and adaptive immune cells that may drive AA.
Collapse
Affiliation(s)
- Sofie Lundgren
- Hematology Research Unit Helsinki, Department of Hematology, University of Helsinki and Helsinki University Hospital Comprehensive Cancer Center, Helsinki 00290, Finland
- Translational Immunology Research Program, University of Helsinki, Helsinki 00290, Finland
- ICAN Digital Precision Cancer Medicine Flagship, University of Helsinki and Helsinki University Hospital Comprehensive Cancer Center, Helsinki 00290, Finland
| | - Jani Huuhtanen
- Hematology Research Unit Helsinki, Department of Hematology, University of Helsinki and Helsinki University Hospital Comprehensive Cancer Center, Helsinki 00290, Finland
- Translational Immunology Research Program, University of Helsinki, Helsinki 00290, Finland
- ICAN Digital Precision Cancer Medicine Flagship, University of Helsinki and Helsinki University Hospital Comprehensive Cancer Center, Helsinki 00290, Finland
- Department of Computer Science, Aalto University School of Science, Espoo 02150, Finland
| | - Mikko Keränen
- Hematology Research Unit Helsinki, Department of Hematology, University of Helsinki and Helsinki University Hospital Comprehensive Cancer Center, Helsinki 00290, Finland
- Translational Immunology Research Program, University of Helsinki, Helsinki 00290, Finland
- ICAN Digital Precision Cancer Medicine Flagship, University of Helsinki and Helsinki University Hospital Comprehensive Cancer Center, Helsinki 00290, Finland
- Department of Hematology, Helsinki University Hospital Comprehensive Cancer Center, Helsinki 00290, Finland
| | - Xingmin Feng
- National Heart Lung and Blood Institute (NHLBI), National Institutes of Health (NIH), Bethesda, MD 20892, USA
| | - Bhavisha A Patel
- National Heart Lung and Blood Institute (NHLBI), National Institutes of Health (NIH), Bethesda, MD 20892, USA
| | - Georgina L Ryland
- Sir Peter MacCallum Department of Oncology, University of Melbourne, Melbourne, VIC 3052, Australia
| | - Lucy C Fox
- Sir Peter MacCallum Department of Oncology, University of Melbourne, Melbourne, VIC 3052, Australia
| | - Carlos Bravo-Perez
- Department of Translational Hematology and Oncology Research, Taussig Cancer Institute, Cleveland Clinic, Cleveland, OH 44106, USA
- Department of Hematology and Medical Oncology, Hospital Universitario Morales Meseguer, University of Murcia, IMIB-Pascual Parrilla, CIBERER-Instituto de Salud Carlos III, Murcia 30008, Spain
| | - Michael Clemente
- Department of Translational Hematology and Oncology Research, Taussig Cancer Institute, Cleveland Clinic, Cleveland, OH 44106, USA
| | - Cassandra Kerr
- Department of Translational Hematology and Oncology Research, Taussig Cancer Institute, Cleveland Clinic, Cleveland, OH 44106, USA
| | - Gunilla Walldin
- Center for Hematology and Regenerative Medicine, Department of Medicine, Karolinska Institutet, Karolinska University Hospital Huddinge, Huddinge 14157, Sweden
| | - Olli Dufva
- Hematology Research Unit Helsinki, Department of Hematology, University of Helsinki and Helsinki University Hospital Comprehensive Cancer Center, Helsinki 00290, Finland
- Translational Immunology Research Program, University of Helsinki, Helsinki 00290, Finland
- ICAN Digital Precision Cancer Medicine Flagship, University of Helsinki and Helsinki University Hospital Comprehensive Cancer Center, Helsinki 00290, Finland
| | - Yoshitaka Zaimoku
- Department of Hematology, Faculty of Medicine, Institute of Medical Pharmaceutical and Health Sciences, Kanazawa University, Ishikawa 920-1192, Japan
| | - Tiina Tuononen
- School of Medicine, University of Eastern Finland, Kuopio 70211, Finland
| | - Mikko Myllymäki
- Hematology Research Unit Helsinki, Department of Hematology, University of Helsinki and Helsinki University Hospital Comprehensive Cancer Center, Helsinki 00290, Finland
- Translational Immunology Research Program, University of Helsinki, Helsinki 00290, Finland
- ICAN Digital Precision Cancer Medicine Flagship, University of Helsinki and Helsinki University Hospital Comprehensive Cancer Center, Helsinki 00290, Finland
| | - Freja Ebeling
- Department of Hematology, Helsinki University Hospital Comprehensive Cancer Center, Helsinki 00290, Finland
| | - Emmi Jokinen
- Hematology Research Unit Helsinki, Department of Hematology, University of Helsinki and Helsinki University Hospital Comprehensive Cancer Center, Helsinki 00290, Finland
- Translational Immunology Research Program, University of Helsinki, Helsinki 00290, Finland
- ICAN Digital Precision Cancer Medicine Flagship, University of Helsinki and Helsinki University Hospital Comprehensive Cancer Center, Helsinki 00290, Finland
- Department of Computer Science, Aalto University School of Science, Espoo 02150, Finland
| | - Markus Heinonen
- Department of Computer Science, Aalto University School of Science, Espoo 02150, Finland
- Helsinki Institute for Information Technology HIIT, Espoo 02150, Finland
| | - Tiina Kasanen
- Hematology Research Unit Helsinki, Department of Hematology, University of Helsinki and Helsinki University Hospital Comprehensive Cancer Center, Helsinki 00290, Finland
- Translational Immunology Research Program, University of Helsinki, Helsinki 00290, Finland
- ICAN Digital Precision Cancer Medicine Flagship, University of Helsinki and Helsinki University Hospital Comprehensive Cancer Center, Helsinki 00290, Finland
| | - Jay Klievink
- Hematology Research Unit Helsinki, Department of Hematology, University of Helsinki and Helsinki University Hospital Comprehensive Cancer Center, Helsinki 00290, Finland
- Translational Immunology Research Program, University of Helsinki, Helsinki 00290, Finland
- ICAN Digital Precision Cancer Medicine Flagship, University of Helsinki and Helsinki University Hospital Comprehensive Cancer Center, Helsinki 00290, Finland
| | - Hanna Lähteenmäki
- Hematology Research Unit Helsinki, Department of Hematology, University of Helsinki and Helsinki University Hospital Comprehensive Cancer Center, Helsinki 00290, Finland
- Translational Immunology Research Program, University of Helsinki, Helsinki 00290, Finland
- ICAN Digital Precision Cancer Medicine Flagship, University of Helsinki and Helsinki University Hospital Comprehensive Cancer Center, Helsinki 00290, Finland
| | - Taina Jaatinen
- Histocompatibility Testing Laboratory, Finnish Red Cross Blood Service, Vantaa 01730, Finland
| | - Sari Kytölä
- Department of Hematology, Helsinki University Hospital Comprehensive Cancer Center, Helsinki 00290, Finland
| | - Sanna Siitonen
- Department of Clinical Chemistry, HUS Diagnostic Centre, Helsinki University Hospital and University of Helsinki, Helsinki 00290, Finland
| | - Alina Dulau-Florea
- Hematology Laboratory, Department of Laboratory Medicine/Clinical Center, National Institutes of Health (NIH), Bethesda, MD 20892, USA
| | - Raul Braylan
- Hematology Laboratory, Department of Laboratory Medicine/Clinical Center, National Institutes of Health (NIH), Bethesda, MD 20892, USA
| | - Merja Heinäniemi
- School of Medicine, University of Eastern Finland, Kuopio 70211, Finland
| | - Shinji Nakao
- Department of Hematology, Faculty of Medicine, Institute of Medical Pharmaceutical and Health Sciences, Kanazawa University, Ishikawa 920-1192, Japan
| | - Eva Hellström-Lindberg
- Center for Hematology and Regenerative Medicine, Department of Medicine, Karolinska Institutet, Karolinska University Hospital Huddinge, Huddinge 14157, Sweden
| | - Jaroslaw P Maciejewski
- Department of Translational Hematology and Oncology Research, Taussig Cancer Institute, Cleveland Clinic, Cleveland, OH 44106, USA
| | - Piers Blombery
- Sir Peter MacCallum Department of Oncology, University of Melbourne, Melbourne, VIC 3052, Australia
| | - Neal S Young
- National Heart Lung and Blood Institute (NHLBI), National Institutes of Health (NIH), Bethesda, MD 20892, USA
| | - Harri Lähdesmäki
- Department of Computer Science, Aalto University School of Science, Espoo 02150, Finland
| | - Satu Mustjoki
- Hematology Research Unit Helsinki, Department of Hematology, University of Helsinki and Helsinki University Hospital Comprehensive Cancer Center, Helsinki 00290, Finland
- Translational Immunology Research Program, University of Helsinki, Helsinki 00290, Finland
- ICAN Digital Precision Cancer Medicine Flagship, University of Helsinki and Helsinki University Hospital Comprehensive Cancer Center, Helsinki 00290, Finland
- Department of Clinical Chemistry and Hematology, University of Helsinki, Helsinki 00290, Finland
| |
Collapse
|
11
|
Rotondi Aufiero V, Iacomino G, De Chiara G, Picariello E, Iaquinto G, Troncone R, Mazzarella G. Neutralizing IL-15 Inhibits Tissue-Damaging Immune Response in Ex Vivo Cultured Untreated Celiac Intestinal Mucosa. Cells 2025; 14:234. [PMID: 39937025 PMCID: PMC11818035 DOI: 10.3390/cells14030234] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2024] [Revised: 01/27/2025] [Accepted: 02/01/2025] [Indexed: 02/13/2025] Open
Abstract
In celiac disease (CeD), interleukin 15 (IL-15) affects the epithelial barrier by acting on intraepithelial lymphocytes, promoting interferon γ (IFN-γ) production and inducing strong cytotoxic activity as well as eliciting apoptotic death of enterocytes by the Fas/Fas ligand system. This study investigates the effects of a monoclonal antibody neutralizing the effects of IL-15 (aIL-15) on tissue-damaging immune response in untreated CeD patients by using an organ culture system. Jejunal biopsies from 10 untreated CeD patients were cultured ex vivo with or without aIL-15. Epithelial expressions of CD95/Fas, HLA-E and perforin were analyzed by immunohistochemistry. Apoptosis was detected in the epithelium by using the terminal deoxynucleotidyl transferase-mediated dUTP nick-end labeling (TUNEL) assay. Additionally, the surface epithelium compartment of ex vivo cultured biopsy samples was isolated by laser capture microdissection (LCM). RNA from each LCM sample was extracted and the relative expression of IFN-γ was evaluated by quantitative reverse transcriptase-PCR (qRT-PCR). Biopsies cultured with the aIL-15 antibody showed a reduction in Fas, HLA-E and perforin epithelial expression, as well as a decrease in epithelial TUNEL+ cells compared to biopsies cultured without the aIL-15 antibody. Moreover, downregulation of epithelial IFN-γ expression was recorded in biopsies incubated with aIL-15, compared to those cultured without aIL-15. Our findings suggest that neutralizing the effects of IL-15 in ex vivo cultured untreated CeD intestinal mucosa could block apoptosis by downregulating Fas and HLA-E expression and the release of cytotoxic proteins, such as perforin. Furthermore, it can dampen the hyperactive immune response by reducing IFN-γ expression. More generally, our study provides new evidence for the effects of anti-IL-15 neutralizing monoclonal antibodies in preventing or repairing epithelial damage and further supports the concept that IL-15 is a meaningful therapeutic target in CeD, or inflammatory diseases associated with the upregulation of IL-15.
Collapse
Affiliation(s)
- Vera Rotondi Aufiero
- Institute of Food Science, National Research Council (ISA-CNR), 83100 Avellino, Italy; (V.R.A.)
- European Laboratory for the Investigation of Food Induced Diseases (ELFID), Via Pansini 5, 80131 Napoli, Italy
| | - Giuseppe Iacomino
- Institute of Food Science, National Research Council (ISA-CNR), 83100 Avellino, Italy; (V.R.A.)
| | | | - Errico Picariello
- Institute of Food Science, National Research Council (ISA-CNR), 83100 Avellino, Italy; (V.R.A.)
| | | | - Riccardo Troncone
- European Laboratory for the Investigation of Food Induced Diseases (ELFID), Via Pansini 5, 80131 Napoli, Italy
- Department of Medical Translational Sciences, University Federico II, 80131 Naples, Italy
| | - Giuseppe Mazzarella
- Institute of Food Science, National Research Council (ISA-CNR), 83100 Avellino, Italy; (V.R.A.)
- European Laboratory for the Investigation of Food Induced Diseases (ELFID), Via Pansini 5, 80131 Napoli, Italy
| |
Collapse
|
12
|
Hada A, Xiao Z. Ligands for Intestinal Intraepithelial T Lymphocytes in Health and Disease. Pathogens 2025; 14:109. [PMID: 40005486 PMCID: PMC11858322 DOI: 10.3390/pathogens14020109] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2024] [Revised: 01/17/2025] [Accepted: 01/22/2025] [Indexed: 02/27/2025] Open
Abstract
The intestinal tract is constantly exposed to a diverse mixture of luminal antigens, such as those derived from commensals, dietary substances, and potential pathogens. It also serves as a primary route of entry for pathogens. At the forefront of this intestinal defense is a single layer of epithelial cells that forms a critical barrier between the gastrointestinal (GI) lumen and the underlying host tissue. The intestinal intraepithelial T lymphocytes (T-IELs), one of the most abundant lymphocyte populations in the body, play a crucial role in actively surveilling and maintaining the integrity of this barrier by tolerating non-harmful factors such as commensal microbiota and dietary components, promoting epithelial turnover and renewal while also defending against pathogens. This immune balance is maintained through interactions between ligands in the GI microenvironment and receptors on T-IELs. This review provides a detailed examination of the ligands present in the intestinal epithelia and the corresponding receptors expressed on T-IELs, including T cell receptors (TCRs) and non-TCRs, as well as how these ligand-receptor interactions influence T-IEL functions under both steady-state and pathological conditions. By understanding these engagements, we aim to shed light on the mechanisms that govern T-IEL activities within the GI microenvironment. This knowledge may help in developing strategies to target GI ligands and modulate T-IEL receptor expression, offering precise approaches for treating intestinal disorders.
Collapse
Affiliation(s)
| | - Zhengguo Xiao
- Department of Animal and Avian Sciences, University of Maryland, College Park, MD 20742, USA;
| |
Collapse
|
13
|
Aitella E, Cozzolino D, Ginaldi L, Romano C. Celiac Disease: A Transitional Point of View. Nutrients 2025; 17:234. [PMID: 39861364 PMCID: PMC11767334 DOI: 10.3390/nu17020234] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2024] [Revised: 01/03/2025] [Accepted: 01/04/2025] [Indexed: 01/27/2025] Open
Abstract
Celiac disease (CeD) is a chronic, lifelong, multifactorial, polygenic, and autoimmune disorder, characteristically triggered by exposure to the exogenous factor "gluten" in genetically predisposed individuals, with resulting duodenal inflammation and enteropathy, as well as heterogeneous multisystemic and extraintestinal manifestations. The immunopathogenesis of CeD is complex, favored by a peculiar human leukocyte antigen (HLA) genetic predisposition, leading to gluten presentation by antigen-presenting cells to CD4+ T helper (Th) cells, T cell-B cell interactions, and production of specific antibodies, resulting in the immune-mediated killing of enterocytes and, macroscopically, in duodenal inflammation. Here, the most relevant correlations between cellular and molecular aspects and clinical manifestations of this complex disease are reviewed, with final considerations on nutritional aspects for disease management.
Collapse
Affiliation(s)
- Ernesto Aitella
- Department of Clinical Medicine, Public Health, Life and Environmental Sciences, University of L’Aquila, 67100 L’Aquila, Italy; (E.A.); (L.G.)
- Allergy and Clinical Immunology Unit, “G. Mazzini” Hospital, ASL Teramo, 64100 Teramo, Italy
| | - Domenico Cozzolino
- Division of Internal Medicine, Department of Precision Medicine, “Luigi Vanvitelli” University of Campania, 80131 Naples, Italy;
| | - Lia Ginaldi
- Department of Clinical Medicine, Public Health, Life and Environmental Sciences, University of L’Aquila, 67100 L’Aquila, Italy; (E.A.); (L.G.)
- Allergy and Clinical Immunology Unit, “G. Mazzini” Hospital, ASL Teramo, 64100 Teramo, Italy
| | - Ciro Romano
- Clinical Immunology Outpatient Clinic, Division of Internal Medicine, Department of Advanced Medical and Surgical Sciences, “Luigi Vanvitelli” University of Campania, 80131 Naples, Italy
| |
Collapse
|
14
|
Fiz-López A, De Prado Á, Arribas-Rodríguez E, García-Alonso FJ, Izquierdo S, Martín-Muñoz Á, Garrote JA, Arranz E, Barrio J, Fernández-Salazar L, Bernardo D. Biological variability of human intraepithelial lymphocytes throughout the human gastrointestinal tract in health and coeliac disease. Eur J Clin Invest 2024; 54:e14304. [PMID: 39210517 DOI: 10.1111/eci.14304] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/04/2024] [Accepted: 08/08/2024] [Indexed: 09/04/2024]
Abstract
BACKGROUND Intraepithelial lymphocytes are the first line of defence of the human intestinal immune system. Besides, their composition is altered on patients with coeliac disease (CD), so they are considered as biomarkers with utility on their diagnose and/or monitoring. Our aim is to address their variability through the human gastrointestinal tract in health and characterized them in further depth in the coeliac duodenum. METHODS Intraepithelial lymphocytes were isolated from human gastric, duodenal, ileal and colonic biopsies, then stained with specific antibodies and acquired by flow cytometry. RESULTS Our results confirmed that the profile of Intraepithelial lymphocytes change through the length of the human gastrointestinal tract. Besides and given the central role that Interleukin-15 (IL-15) elicits on CD pathogenesis; we also assessed the expression of its receptor revealing that there was virtually no functional IL-15 receptor on duodenal Intraepithelial lymphocytes. Nevertheless and contrary to our expectations, the active IL-15 receptor was not increased either on Intraepithelial lymphocytes from CD patients. CONCLUSIONS IL-15 might require additional stimulus to activate intraepithelial lymphocytes. These findings may provide novel tools to aid on a CD diagnosis and/or monitoring, at the time that provide the bases to perform functional studies in order of getting a deeper insight in the specific function that Intraepithelial lymphocytes elicit on CD pathogenesis.
Collapse
Affiliation(s)
- Aida Fiz-López
- Mucosal Immunology Lab, Unidad de Excelencia Instituto Biomedicina y Genética Molecular (IBGM), Universidad de Valladolid-CSIC, Valladolid, Spain
| | - Ángel De Prado
- Mucosal Immunology Lab, Unidad de Excelencia Instituto Biomedicina y Genética Molecular (IBGM), Universidad de Valladolid-CSIC, Valladolid, Spain
- Servicio de Gastroenterología, Hospital Universitario Río Hortega, Valladolid, Spain
| | - Elisa Arribas-Rodríguez
- Mucosal Immunology Lab, Unidad de Excelencia Instituto Biomedicina y Genética Molecular (IBGM), Universidad de Valladolid-CSIC, Valladolid, Spain
| | | | - Sandra Izquierdo
- Servicio de Gastroenterología, Hospital Clínico Universitario, Valladolid, Spain
| | - Álvaro Martín-Muñoz
- Cytometry Facility, Unidad de Excelencia Instituto Biomedicina y Genética Molecular (IBGM, Universidad de Valladolid-CSIC), Valladolid, Spain
| | - José A Garrote
- Mucosal Immunology Lab, Unidad de Excelencia Instituto Biomedicina y Genética Molecular (IBGM), Universidad de Valladolid-CSIC, Valladolid, Spain
| | - Eduardo Arranz
- Mucosal Immunology Lab, Unidad de Excelencia Instituto Biomedicina y Genética Molecular (IBGM), Universidad de Valladolid-CSIC, Valladolid, Spain
| | - Jesús Barrio
- Servicio de Gastroenterología, Hospital Universitario Río Hortega, Valladolid, Spain
| | - Luis Fernández-Salazar
- Servicio de Gastroenterología, Hospital Clínico Universitario, Valladolid, Spain
- Departamento de Medicina, Dermatología y Toxicología, Universidad de Valladolid, Valladolid, Spain
| | - David Bernardo
- Mucosal Immunology Lab, Unidad de Excelencia Instituto Biomedicina y Genética Molecular (IBGM), Universidad de Valladolid-CSIC, Valladolid, Spain
- Centro de Investigaciones Biomédicas en Red de Enfermedades Infecciosas (CIBERINFEC), Madrid, Spain
| |
Collapse
|
15
|
Yosri M, Dokhan M, Aboagye E, Al Moussawy M, Abdelsamed HA. Mechanisms governing bystander activation of T cells. Front Immunol 2024; 15:1465889. [PMID: 39669576 PMCID: PMC11635090 DOI: 10.3389/fimmu.2024.1465889] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2024] [Accepted: 10/31/2024] [Indexed: 12/14/2024] Open
Abstract
The immune system is endowed with the capacity to distinguish between self and non-self, so-called immune tolerance or "consciousness of the immune system." This type of awareness is designed to achieve host protection by eliminating cells expressing a wide range of non-self antigens including microbial-derived peptides. Such a successful immune response is associated with the secretion of a whole spectrum of soluble mediators, e.g., cytokines and chemokines, which not only contribute to the clearance of infected host cells but also activate T cells that are not specific to the original cognate antigen. This kind of non-specific T-cell activation is called "bystander activation." Although it is well-established that this phenomenon is cytokine-dependent, there is evidence in the literature showing the involvement of peptide/MHC recognition depending on the type of T-cell subset (naive vs. memory). Here, we will summarize our current understanding of the mechanism(s) of bystander T-cell activation as well as its biological significance in a wide range of diseases including microbial infections, cancer, auto- and alloimmunity, and chronic inflammatory diseases such as atherosclerosis.
Collapse
Affiliation(s)
- Mohammed Yosri
- The Regional Center for Mycology and Biotechnology, Al-Azhar University, Cairo, Egypt
| | - Mohamed Dokhan
- Immunology Center of Georgia (IMMCG), Medical College of Georgia (MCG), Augusta University, Augusta, GA, United States
| | - Elizabeth Aboagye
- Immunology Center of Georgia (IMMCG), Medical College of Georgia (MCG), Augusta University, Augusta, GA, United States
| | - Mouhamad Al Moussawy
- Starzl Transplantation Institute, School of Medicine, University of Pittsburgh, Pittsburgh, PA, United States
| | - Hossam A. Abdelsamed
- Immunology Center of Georgia (IMMCG), Medical College of Georgia (MCG), Augusta University, Augusta, GA, United States
- Department of Physiology, Augusta University, Augusta, GA, United States
| |
Collapse
|
16
|
Drosu N, Bjornevik K, Cortese M, Levy M, Sollid LM. Coeliac disease as a model for understanding multiple sclerosis. Nat Rev Neurol 2024; 20:685-690. [PMID: 39379493 DOI: 10.1038/s41582-024-01025-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/11/2024] [Indexed: 10/10/2024]
Abstract
The genetic architecture of multiple sclerosis (MS) is similar to that of coeliac disease, with human leukocyte antigen (HLA) being the greatest genetic determinant in both diseases. Furthermore, similar to the involvement of gluten in coeliac disease, Epstein-Barr virus (EBV) infection is now widely considered to be an important environmental factor in MS. The molecular basis for the HLA association in coeliac disease is well defined, and B cells have a clear role in antigen presentation to gluten-specific CD4+ T cells. By contrast, the mechanisms underlying the HLA association of MS are unknown but accumulating evidence indicates a similar role of B cells acting as antigen-presenting cells. The growing parallels suggest that much could be learned about the mechanisms of MS by using coeliac disease as a model. In this Perspective article, we discuss the insights that could be gained from these parallels and consider the possibility of antiviral treatment against EBV as a therapy for MS that is analogous to the gluten-free diet in coeliac disease.
Collapse
Affiliation(s)
- Natalia Drosu
- Department of Neurology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - Kjetil Bjornevik
- Department of Nutrition, Harvard T.H. Chan School of Public Health, Boston, MA, USA
- Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, MA, USA
| | - Marianna Cortese
- Department of Nutrition, Harvard T.H. Chan School of Public Health, Boston, MA, USA
| | - Michael Levy
- Department of Neurology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - Ludvig M Sollid
- Institute of Clinical Medicine, University of Oslo, Oslo, Norway.
- Department of Immunology, Oslo University Hospital-Rikshospitalet, Oslo, Norway.
| |
Collapse
|
17
|
Drosu N, Anderson M, Bilodeau PA, Nishiyama S, Mikami T, Bobrowski-Khoury N, Cabot J, Housman D, Levy M. CD4 T cells restricted to DRB1*15:01 recognize two Epstein-Barr virus glycoproteins capable of intracellular antigen presentation. Proc Natl Acad Sci U S A 2024; 121:e2416097121. [PMID: 39432795 PMCID: PMC11536159 DOI: 10.1073/pnas.2416097121] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2024] [Accepted: 09/06/2024] [Indexed: 10/23/2024] Open
Abstract
Both genetic and environmental factors contribute to multiple sclerosis (MS) risk. Infection with the Epstein-Barr virus (EBV) is the strongest environmental risk factor, and HLA-DR15 is the strongest genetic risk factor for MS. We employed computational methods and in vitro assays for CD4 T cell activation to investigate the DR15-restricted response to EBV. Using a machine learning-based HLA ligand predictor, the EBV glycoprotein B (gB) was predicted to be enriched in epitopes restricted to presentation by DRB1*15:01. In DR15-positive individuals, two epitopes comprised the major CD4 T cell response to gB. Surprisingly, the expression of recombinant gB in a DR15-homozygous B cell line or primary autologous B cells elicited a CD4 T cell response, indicating that intracellular gB was loaded onto HLA class II molecules. By deleting the signal sequence of gB, we determined that this pathway for direct activation of CD4 T cells was dependent on trafficking to the endoplasmic reticulum (ER) within the B cell. We screened seven recombinant EBV antigens from the ER compartment for immune responses in DR15-negative vs. DR15-homozygous individuals. In addition to gB, gH was a key CD4 T cell target in individuals homozygous for DR15. Compared to non-DR15 controls, DR15-homozygotes had significantly higher T cell responses to both gB and gH but not to EBV latent or lytic antigens overall. Responses to gB and gH were slightly elevated in DR15 homozygotes with MS. Our results link MS environmental and genetic risk factors by demonstrating that HLA-DR15 dictates CD4 T cell immunity to EBV antigens.
Collapse
Affiliation(s)
- Natalia Drosu
- Department of Neurology, Division of Neuroimmunology & Neuroinfectious Disease, Massachusetts General Hospital and Harvard Medical School, Boston, MA02114
- Department of Biology, Massachusetts Institute of Technology, Cambridge, MA02139
| | - Monique Anderson
- Department of Neurology, Division of Neuroimmunology & Neuroinfectious Disease, Massachusetts General Hospital and Harvard Medical School, Boston, MA02114
| | - Philippe A. Bilodeau
- Department of Neurology, Division of Neuroimmunology & Neuroinfectious Disease, Massachusetts General Hospital and Harvard Medical School, Boston, MA02114
| | - Shuhei Nishiyama
- Department of Neurology, Division of Neuroimmunology & Neuroinfectious Disease, Massachusetts General Hospital and Harvard Medical School, Boston, MA02114
| | - Takahisa Mikami
- Department of Neurology, Division of Neuroimmunology & Neuroinfectious Disease, Massachusetts General Hospital and Harvard Medical School, Boston, MA02114
| | - Natasha Bobrowski-Khoury
- Department of Neurology, Division of Neuroimmunology & Neuroinfectious Disease, Massachusetts General Hospital and Harvard Medical School, Boston, MA02114
| | - Jackson Cabot
- Department of Neurology, Division of Neuroimmunology & Neuroinfectious Disease, Massachusetts General Hospital and Harvard Medical School, Boston, MA02114
| | - David Housman
- Department of Biology, Massachusetts Institute of Technology, Cambridge, MA02139
| | - Michael Levy
- Department of Neurology, Division of Neuroimmunology & Neuroinfectious Disease, Massachusetts General Hospital and Harvard Medical School, Boston, MA02114
| |
Collapse
|
18
|
Dong B, Obermajer N, Tsuji T, Matsuzaki J, Bonura CM, Sander C, Withers H, Long MD, Chavel C, Olejniczak SH, Minderman H, Kirkwood JM, Edwards RP, Storkus WJ, Romero P, Kalinski P. NK Receptor Signaling Lowers TCR Activation Threshold, Enhancing Selective Recognition of Cancer Cells by TAA-Specific CTLs. Cancer Immunol Res 2024; 12:1421-1437. [PMID: 38949179 PMCID: PMC11706306 DOI: 10.1158/2326-6066.cir-24-0061] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2024] [Revised: 05/10/2024] [Accepted: 06/26/2024] [Indexed: 07/02/2024]
Abstract
Cytotoxic CD8+ T lymphocyte (CTL) recognition of non-mutated tumor-associated antigens (TAA), present on cancer cells and also in healthy tissues, is an important element of cancer immunity, but the mechanism of its selectivity for cancer cells and opportunities for its enhancement remain elusive. In this study, we found that CTL expression of the NK receptors (NKR) DNAM1 and NKG2D was associated with the effector status of CD8+ tumor-infiltrating lymphocytes and long-term survival of patients with melanoma. Using MART1 and NY-ESO-1 as model TAAs, we demonstrated that DNAM1 and NKG2D regulate T-cell receptor (TCR) functional avidity and set the threshold for TCR activation of human TAA-specific CTLs. Superior co-stimulatory effects of DNAM1 over CD28 involved enhanced TCR signaling, CTL killer function, and polyfunctionality. Double transduction of human CTLs with TAA-specific TCR and NKRs resulted in strongly enhanced antigen sensitivity, without a reduction in antigen specificity and selectivity of killer function. In addition, the elevation of NKR ligand expression on cancer cells due to chemotherapy also increased CTL recognition of cancer cells expressing low levels of TAAs. Our data help explain the ability of self-antigens to mediate tumor rejection in the absence of autoimmunity and support the development of dual-targeting adoptive T-cell therapies that use NKRs to enhance the potency and selectivity of recognition of TAA-expressing cancer cells.
Collapse
MESH Headings
- Humans
- Receptors, Antigen, T-Cell/metabolism
- Receptors, Antigen, T-Cell/immunology
- Signal Transduction
- T-Lymphocytes, Cytotoxic/immunology
- T-Lymphocytes, Cytotoxic/metabolism
- Antigens, Neoplasm/immunology
- Antigens, Neoplasm/metabolism
- Melanoma/immunology
- Melanoma/metabolism
- NK Cell Lectin-Like Receptor Subfamily K/metabolism
- Lymphocyte Activation/immunology
- Lymphocytes, Tumor-Infiltrating/immunology
- Lymphocytes, Tumor-Infiltrating/metabolism
- Antigens, Differentiation, T-Lymphocyte/metabolism
- Antigens, Differentiation, T-Lymphocyte/immunology
- Cell Line, Tumor
- MART-1 Antigen/immunology
- MART-1 Antigen/metabolism
- Cytotoxicity, Immunologic
- T Lineage-Specific Activation Antigen 1
Collapse
Affiliation(s)
- Bowen Dong
- Department of Immunology, Roswell Park Comprehensive Cancer Center; Buffalo, NY, United States
- Department of Medicine, Roswell Park Comprehensive Cancer Center; Buffalo, NY, United States
| | - Nataša Obermajer
- Department of Surgery, University of Pittsburgh School of Medicine; Pittsburgh, PA, United States
| | - Takemasa Tsuji
- Department of Immunology, Roswell Park Comprehensive Cancer Center; Buffalo, NY, United States
| | - Junko Matsuzaki
- Department of Immunology, Roswell Park Comprehensive Cancer Center; Buffalo, NY, United States
| | - Cindy M. Bonura
- Department of Immunology, Roswell Park Comprehensive Cancer Center; Buffalo, NY, United States
| | - Cindy Sander
- Department of Medicine, University of Pittsburgh School of Medicine; Pittsburgh, PA, United States
| | - Henry Withers
- Department of Biostatistics and Bioinformatics, Roswell Park Comprehensive Cancer Center; Buffalo, NY, United States
| | - Mark D. Long
- Department of Biostatistics and Bioinformatics, Roswell Park Comprehensive Cancer Center; Buffalo, NY, United States
| | - Colin Chavel
- Department of Immunology, Roswell Park Comprehensive Cancer Center; Buffalo, NY, United States
| | - Scott H. Olejniczak
- Department of Immunology, Roswell Park Comprehensive Cancer Center; Buffalo, NY, United States
| | - Hans Minderman
- Department of Flow and Immune Analysis Shared Resource, Roswell Park Comprehensive Cancer Center; Buffalo, NY, United States
| | - John M. Kirkwood
- Department of Medicine, University of Pittsburgh School of Medicine; Pittsburgh, PA, United States
| | - Robert P. Edwards
- Department of OB-GYN, University of Pittsburgh School of Medicine; Pittsburgh, PA, United States
| | - Walter J. Storkus
- Department of Dermatology, University of Pittsburgh School of Medicine; Pittsburgh, PA, United States
- Department of Immunology, University of Pittsburgh School of Medicine; Pittsburgh, PA, United States
| | - Pedro Romero
- University of Lausanne and Ludwig Institute for Cancer Research; Lausanne, Switzerland
| | - Pawel Kalinski
- Department of Immunology, Roswell Park Comprehensive Cancer Center; Buffalo, NY, United States
- Department of Medicine, Roswell Park Comprehensive Cancer Center; Buffalo, NY, United States
- Department of Surgery, University of Pittsburgh School of Medicine; Pittsburgh, PA, United States
- Department of Immunology, University of Pittsburgh School of Medicine; Pittsburgh, PA, United States
| |
Collapse
|
19
|
Kivelä L, Lindfors K, Lundin KEA, Størdal K. Review article: Faecal biomarkers for assessing small intestinal damage in coeliac disease and environmental enteropathy. Aliment Pharmacol Ther 2024; 60:988-1004. [PMID: 39233618 DOI: 10.1111/apt.18234] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/03/2024] [Revised: 06/19/2024] [Accepted: 08/20/2024] [Indexed: 09/06/2024]
Abstract
BACKGROUND In coeliac disease and environmental enteropathy, dietary gluten and enteric infections cause reversible inflammation and morphological changes to the small intestinal mucosa that can be detected in biopsy samples obtained by endoscopy. However, there is a clear need for non-invasive biomarkers. Constant shedding of mucosal material into the bowel lumen and faeces, together with easy availability of stool, makes it an interesting sample matrix. AIMS To conduct a systematic literature search and summarize the existing evidence for host mucosa-derived faecal biomarkers in evaluating small intestinal damage. METHODS We searched for studies on PubMed (MEDLINE) until 1 March 2024. RESULTS We identified 494 studies and included 35 original case-control and cohort studies. These assessed host mucosal transcripts and 14 other markers aiming specifically to reflect inflammation and cell-mediated, innate and gluten-induced immune responses. In coeliac disease, faecal calprotectin and anti-gliadin, tissue transglutaminase, endomysium and deamidated gliadin peptide antibodies were the most studied but with inconsistent results. Single studies reported positive findings about microRNA transcripts, β-defensin-2, lipocalin-2, zonulin-related proteins and angiotensin-converting enzyme. In environmental enteropathy, a non-significant association was reported between calprotectin and urine lactulose/mannitol ratio; there were conflicting results for neopterin, myeloperoxidase and host transcripts. Single studies reported a positive association for lactoferrin, and a negative association for regenerating islet-derived protein 1. Studies comparing faecal markers against small intestinal biopsy findings were not identified in environmental enteropathy. CONCLUSIONS Further studies are needed to determine reliable faecal markers as a proxy for small intestinal mucosal damage.
Collapse
Affiliation(s)
- Laura Kivelä
- Department of Pediatric Research, Faculty of Medicine, University of Oslo, Oslo, Norway
- Celiac Disease Research Center, Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland
- Children's Hospital, Helsinki University Hospital, Helsinki, Finland
- Department of Pediatrics, Tampere University Hospital, Wellbeing Services County of Pirkanmaa, Tampere, Finland
| | - Katri Lindfors
- Celiac Disease Research Center, Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland
| | - Knut E A Lundin
- Department of Gastroenterology, Oslo University Hospital Rikshospitalet, Oslo, Norway
- Norwegian Coeliac Disease Research Centre, Institute of Clinical Medicine, Faculty of Medicine, University of Oslo, Oslo, Norway
| | - Ketil Størdal
- Department of Pediatric Research, Faculty of Medicine, University of Oslo, Oslo, Norway
- Department of Pediatric and Adolescent Medicine, Oslo University Hospital, Oslo, Norway
| |
Collapse
|
20
|
Verdelho Machado M. Refractory Celiac Disease: What the Gastroenterologist Should Know. Int J Mol Sci 2024; 25:10383. [PMID: 39408713 PMCID: PMC11477276 DOI: 10.3390/ijms251910383] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2024] [Revised: 09/20/2024] [Accepted: 09/25/2024] [Indexed: 10/20/2024] Open
Abstract
Fewer than 1% of patients with celiac disease (CD) will develop refractory CD (RCD). As such, most gastroenterologists might never need to manage patients with RCD. However, all gastroenterologists must be familiarized with the basic concepts of RCD and non-responsive CD (NRCD), since it can present as a severe disease with high mortality, not only due to intestinal failure, but also due to progression to enteropathy-associated T cell lymphoma (EATL) and a higher susceptibility to life-threatening infections. The diagnostic workup and differential diagnosis with other causes of gastrointestinal symptoms and villous atrophy, as well as the differentiation between type I and II RCD, are complex, and may require specialized laboratories and reference hospitals. Immunosuppression is efficient in the milder RCDI; however, the treatment of RCDII falls short, with current options probably only providing transient clinical improvement and delaying EATL development. This review summarizes the current diagnostic and therapeutic approach for patients with RCD that all doctors that manage patients with CD should know.
Collapse
Affiliation(s)
- Mariana Verdelho Machado
- Gastroenterology Department, Hospital de Vila Franca de Xira, 2600-009 Lisbon, Portugal; ; Tel.: +351-912620306
- Gastroenterology Department, Faculdade de Medicina, Lisbon University, 1649-028 Lisboa, Portugal
| |
Collapse
|
21
|
Ge HJ, Chen XL. Advances in understanding and managing celiac disease: Pathophysiology and treatment strategies. World J Gastroenterol 2024; 30:3932-3941. [PMID: 39351055 PMCID: PMC11438662 DOI: 10.3748/wjg.v30.i35.3932] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/06/2024] [Revised: 08/12/2024] [Accepted: 08/26/2024] [Indexed: 09/13/2024] Open
Abstract
In this editorial, we comment on an article published in the recent issue of the World Journal of Gastroenterology. Celiac disease (CeD) is a disease occurring in genetically susceptible individuals, which is mainly characterized by gluten intolerance in the small intestine and clinical symptoms such as abdominal pain, diarrhea, and malnutrition. Therefore, patients often need a lifelong gluten-free diet, which greatly affects the quality of life and expenses of patients. The gold standard for diagnosis is intestinal mucosal biopsy, combined with serological and genetic tests. At present, the lack of safe, effective, and satisfactory drugs for CeD is mainly due to the complexity of its pathogenesis, and it is difficult to find a perfect target to solve the multi-level needs of patients. In this editorial, we mainly review the pathological mechanism of CeD and describe the current experimental and improved drugs for various pathological aspects.
Collapse
Affiliation(s)
- Hao-Jie Ge
- Department of Burns, The First Affiliated Hospital of Anhui Medical University, Hefei 230022, Anhui Province, China
| | - Xu-Lin Chen
- Department of Burns, The First Affiliated Hospital of Anhui Medical University, Hefei 230022, Anhui Province, China
| |
Collapse
|
22
|
Levescot A, Cerf-Bensussan N. Loss of tolerance to dietary proteins: From mouse models to human model diseases. Immunol Rev 2024; 326:173-190. [PMID: 39295093 DOI: 10.1111/imr.13395] [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: 09/21/2024]
Abstract
The critical importance of the immunoregulatory mechanisms, which prevent adverse responses to dietary proteins is demonstrated by the consequences of their failure in two common but distinct human pathological conditions, food allergy and celiac disease. The mechanisms of tolerance to dietary proteins have been extensively studied in mouse models but the extent to which the results in mice can be extrapolated to humans remains unclear. Here, after summarizing the mechanisms known to control oral tolerance in mouse models, we discuss how the monogenic immune disorders associated with food allergy on the one hand, and celiac disease, on the other hand, represent model diseases to gain insight into the key immunoregulatory pathways that control immune responses to food antigens in humans. The spectrum of monogenic disorders, in which the dysfunction of a single gene, is strongly associated with TH2-mediated food allergy suggests an important overlap between the mechanisms that regulate TH2 and IgE responses to food antigens in humans and mice. In contrast, celiac disease provides a unique example of the link between autoimmunity and loss of tolerance to a food antigen.
Collapse
Affiliation(s)
- Anais Levescot
- Laboratory of Intestinal Immunity, INSERM UMR 1163 and Imagine Institute, Université Paris Cité, Paris, France
| | - Nadine Cerf-Bensussan
- Laboratory of Intestinal Immunity, INSERM UMR 1163 and Imagine Institute, Université Paris Cité, Paris, France
| |
Collapse
|
23
|
Srinivas N, Peiffer L, Horny K, Lei KC, Buus TB, Kubat L, Luo M, Yin M, Spassova I, Sucker A, Farahpour F, Kehrmann J, Ugurel S, Livingstone E, Gambichler T, Ødum N, Becker JC. Single-cell RNA and T-cell receptor sequencing unveil mycosis fungoides heterogeneity and a possible gene signature. Front Oncol 2024; 14:1408614. [PMID: 39169943 PMCID: PMC11337020 DOI: 10.3389/fonc.2024.1408614] [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: 03/28/2024] [Accepted: 07/19/2024] [Indexed: 08/23/2024] Open
Abstract
Background Mycosis fungoides (MF) is the most common subtype of cutaneous T-cell lymphoma (CTCL). Comprehensive analysis of MF cells in situ and ex vivo is complicated by the fact that is challenging to distinguish malignant from reactive T cells with certainty. Methods To overcome this limitation, we performed combined single-cell RNA (scRNAseq) and T-cell receptor TCR sequencing (scTCRseq) of skin lesions of cutaneous MF lesions from 12 patients. A sufficient quantity of living T cells was obtained from 9 patients, but 2 had to be excluded due to unclear diagnoses (coexisting CLL or revision to a fixed toxic drug eruption). Results From the remaining patients we established single-cell mRNA expression profiles and the corresponding TCR repertoire of 18,630 T cells. TCR clonality unequivocally identified 13,592 malignant T cells. Reactive T cells of all patients clustered together, while malignant cells of each patient formed a unique cluster expressing genes typical of naive/memory, such as CD27, CCR7 and IL7R, or cytotoxic T cells, e.g., GZMA, NKG7 and GNLY. Genes encoding classic CTCL markers were not detected in all clusters, consistent with the fact that mRNA expression does not correlate linearly with protein expression. Nevertheless, we successfully pinpointed distinctive gene signatures differentiating reactive malignant from malignant T cells: keratins (KRT81, KRT86), galectins (LGALS1, LGALS3) and S100 genes (S100A4, S100A6) being overexpressed in malignant cells. Conclusions Combined scRNAseq and scTCRseq not only allows unambiguous identification of MF cells, but also revealed marked heterogeneity between and within patients with unexpected functional phenotypes. While the correlation between mRNA and protein abundance was limited with respect to established MF markers, we were able to identify a single-cell gene expression signature that distinguishes malignant from reactive T cells.
Collapse
Affiliation(s)
- Nalini Srinivas
- Translational Skin Cancer Research, German Cancer Consortium (DKTK), University Medicine Essen, Essen, Germany
- German Cancer Research Center (DKFZ), Heidelberg, Germany
- Department of Dermatology, University Hospital Essen, and German Cancer Consortium (DKTK) partner site Essen/Düsseldorf, Essen, Germany
| | - Lukas Peiffer
- Translational Skin Cancer Research, German Cancer Consortium (DKTK), University Medicine Essen, Essen, Germany
- German Cancer Research Center (DKFZ), Heidelberg, Germany
- LEO Foundation Skin Immunology Research Center, Department of Immunology and Microbiology, University of Copenhagen, Copenhagen, Denmark
| | - Kai Horny
- Translational Skin Cancer Research, German Cancer Consortium (DKTK), University Medicine Essen, Essen, Germany
- German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Kuan Cheok Lei
- Translational Skin Cancer Research, German Cancer Consortium (DKTK), University Medicine Essen, Essen, Germany
- German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Terkild B. Buus
- LEO Foundation Skin Immunology Research Center, Department of Immunology and Microbiology, University of Copenhagen, Copenhagen, Denmark
| | - Linda Kubat
- Translational Skin Cancer Research, German Cancer Consortium (DKTK), University Medicine Essen, Essen, Germany
- German Cancer Research Center (DKFZ), Heidelberg, Germany
- Department of Dermatology, University Hospital Essen, and German Cancer Consortium (DKTK) partner site Essen/Düsseldorf, Essen, Germany
| | - Meng Luo
- Translational Skin Cancer Research, German Cancer Consortium (DKTK), University Medicine Essen, Essen, Germany
- German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Menghong Yin
- Translational Skin Cancer Research, German Cancer Consortium (DKTK), University Medicine Essen, Essen, Germany
- German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Ivelina Spassova
- Translational Skin Cancer Research, German Cancer Consortium (DKTK), University Medicine Essen, Essen, Germany
- German Cancer Research Center (DKFZ), Heidelberg, Germany
- Department of Dermatology, University Hospital Essen, and German Cancer Consortium (DKTK) partner site Essen/Düsseldorf, Essen, Germany
| | - Antje Sucker
- Department of Dermatology, University Hospital Essen, and German Cancer Consortium (DKTK) partner site Essen/Düsseldorf, Essen, Germany
| | - Farnoush Farahpour
- Bioinformatics and Computational Biophysics, University Duisburg-Essen, and Group of Molecular Cell Biology, Institute for Cell Biology (Cancer Research), University Hospital Essen, Essen, Germany
| | - Jan Kehrmann
- Institute of Medical Microbiology, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
| | - Selma Ugurel
- Department of Dermatology, University Hospital Essen, and German Cancer Consortium (DKTK) partner site Essen/Düsseldorf, Essen, Germany
| | - Elisabeth Livingstone
- Department of Dermatology, University Hospital Essen, and German Cancer Consortium (DKTK) partner site Essen/Düsseldorf, Essen, Germany
| | - Thilo Gambichler
- Department of Dermatology, Ruhr-University Bochum, Bochum, Germany
- Department of Dermatology, Dortmund Hospital, University Witten/Herdecke, Dortmund, Germany
| | - Niels Ødum
- LEO Foundation Skin Immunology Research Center, Department of Immunology and Microbiology, University of Copenhagen, Copenhagen, Denmark
| | - Jürgen C. Becker
- Translational Skin Cancer Research, German Cancer Consortium (DKTK), University Medicine Essen, Essen, Germany
- German Cancer Research Center (DKFZ), Heidelberg, Germany
- Department of Dermatology, University Hospital Essen, and German Cancer Consortium (DKTK) partner site Essen/Düsseldorf, Essen, Germany
| |
Collapse
|
24
|
Santos AJM, van Unen V, Lin Z, Chirieleison SM, Ha N, Batish A, Chan JE, Cedano J, Zhang ET, Mu Q, Guh-Siesel A, Tomaske M, Colburg D, Varma S, Choi SS, Christophersen A, Baghdasaryan A, Yost KE, Karlsson K, Ha A, Li J, Dai H, Sellers ZM, Chang HY, Dunn JCY, Zhang BM, Mellins ED, Sollid LM, Fernandez-Becker NQ, Davis MM, Kuo CJ. A human autoimmune organoid model reveals IL-7 function in coeliac disease. Nature 2024; 632:401-410. [PMID: 39048815 PMCID: PMC11747932 DOI: 10.1038/s41586-024-07716-2] [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: 04/04/2022] [Accepted: 06/14/2024] [Indexed: 07/27/2024]
Abstract
In vitro models of autoimmunity are constrained by an inability to culture affected epithelium alongside the complex tissue-resident immune microenvironment. Coeliac disease (CeD) is an autoimmune disease in which dietary gluten-derived peptides bind to the major histocompatibility complex (MHC) class II human leukocyte antigen molecules (HLA)-DQ2 or HLA-DQ8 to initiate immune-mediated duodenal mucosal injury1-4. Here, we generated air-liquid interface (ALI) duodenal organoids from intact fragments of endoscopic biopsies that preserve epithelium alongside native mesenchyme and tissue-resident immune cells as a unit without requiring reconstitution. The immune diversity of ALI organoids spanned T cells, B and plasma cells, natural killer (NK) cells and myeloid cells, with extensive T-cell and B-cell receptor repertoires. HLA-DQ2.5-restricted gluten peptides selectively instigated epithelial destruction in HLA-DQ2.5-expressing organoids derived from CeD patients, and this was antagonized by blocking MHC-II or NKG2C/D. Gluten epitopes stimulated a CeD organoid immune network response in lymphoid and myeloid subsets alongside anti-transglutaminase 2 (TG2) autoantibody production. Functional studies in CeD organoids revealed that interleukin-7 (IL-7) is a gluten-inducible pathogenic modulator that regulates CD8+ T-cell NKG2C/D expression and is necessary and sufficient for epithelial destruction. Furthermore, endogenous IL-7 was markedly upregulated in patient biopsies from active CeD compared with remission disease from gluten-free diets, predominantly in lamina propria mesenchyme. By preserving the epithelium alongside diverse immune populations, this human in vitro CeD model recapitulates gluten-dependent pathology, enables mechanistic investigation and establishes a proof of principle for the organoid modelling of autoimmunity.
Collapse
MESH Headings
- Humans
- Autoantibodies/immunology
- Autoimmunity
- B-Lymphocytes/immunology
- B-Lymphocytes/metabolism
- Biopsy
- Celiac Disease/immunology
- Celiac Disease/pathology
- Celiac Disease/metabolism
- Duodenum/immunology
- Duodenum/pathology
- Duodenum/metabolism
- Epitopes/immunology
- Glutens/immunology
- Glutens/metabolism
- GTP-Binding Proteins/metabolism
- GTP-Binding Proteins/immunology
- HLA-DQ Antigens/immunology
- HLA-DQ Antigens/metabolism
- Interleukin-7/metabolism
- Intestinal Mucosa/immunology
- Intestinal Mucosa/metabolism
- Intestinal Mucosa/pathology
- Killer Cells, Natural/immunology
- Models, Biological
- Myeloid Cells/immunology
- Organoids/immunology
- Organoids/metabolism
- Organoids/pathology
- Protein Glutamine gamma Glutamyltransferase 2/immunology
- Receptors, Antigen, B-Cell/immunology
- Receptors, Antigen, B-Cell/metabolism
- Receptors, Antigen, T-Cell/immunology
- Receptors, Antigen, T-Cell/metabolism
- T-Lymphocytes/immunology
- T-Lymphocytes/metabolism
Collapse
Affiliation(s)
- António J M Santos
- Division of Hematology, Department of Medicine, Stanford University School of Medicine, Stanford, CA, USA
| | - Vincent van Unen
- Division of Hematology, Department of Medicine, Stanford University School of Medicine, Stanford, CA, USA
- Department of Microbiology and Immunology, Stanford University School of Medicine, Stanford, CA, USA
- Institute for Immunity, Transplantation and Infection, Stanford University School of Medicine, Stanford, CA, USA
| | - Zhongqi Lin
- Division of Hematology, Department of Medicine, Stanford University School of Medicine, Stanford, CA, USA
| | - Steven M Chirieleison
- Division of Hematology, Department of Medicine, Stanford University School of Medicine, Stanford, CA, USA
- Department of Pathology, Stanford University School of Medicine, Stanford, CA, USA
| | - Nhi Ha
- Division of Hematology, Department of Medicine, Stanford University School of Medicine, Stanford, CA, USA
| | - Arpit Batish
- Division of Hematology, Department of Medicine, Stanford University School of Medicine, Stanford, CA, USA
| | - Joshua E Chan
- Division of Hematology, Department of Medicine, Stanford University School of Medicine, Stanford, CA, USA
| | - Jose Cedano
- Division of Hematology, Department of Medicine, Stanford University School of Medicine, Stanford, CA, USA
| | - Elisa T Zhang
- Division of Hematology, Department of Medicine, Stanford University School of Medicine, Stanford, CA, USA
| | - Qinghui Mu
- Division of Hematology, Department of Medicine, Stanford University School of Medicine, Stanford, CA, USA
| | - Alexander Guh-Siesel
- Division of Hematology, Department of Medicine, Stanford University School of Medicine, Stanford, CA, USA
| | - Madeline Tomaske
- Division of Hematology, Department of Medicine, Stanford University School of Medicine, Stanford, CA, USA
| | - Deana Colburg
- Department of Pathology, Stanford University School of Medicine, Stanford, CA, USA
| | - Sushama Varma
- Department of Pathology, Stanford University School of Medicine, Stanford, CA, USA
| | - Shannon S Choi
- Division of Hematology, Department of Medicine, Stanford University School of Medicine, Stanford, CA, USA
| | - Asbjørn Christophersen
- K. G. Jebsen Coeliac Disease Research Centre, Institute of Clinical Medicine, University of Oslo, Oslo, Norway
- Department of Immunology, Oslo University Hospital, Oslo, Norway
- Department of Rheumatology, Dermatology and Infectious Diseases, Oslo University Hospital, Oslo, Norway
| | - Ani Baghdasaryan
- Department of Chemistry, Stanford University School of Medicine, Stanford, CA, USA
| | - Kathryn E Yost
- Center for Personal Dynamic Regulomes, Stanford University School of Medicine, Stanford, CA, USA
- Department of Dermatology, Stanford University School of Medicine, Stanford, CA, USA
| | - Kasper Karlsson
- Division of Hematology, Department of Medicine, Stanford University School of Medicine, Stanford, CA, USA
- Division of Oncology, Department of Medicine, Stanford University School of Medicine, Stanford, CA, USA
- Department of Genetics, Stanford University School of Medicine, Stanford, CA, USA
| | - Andrew Ha
- Division of Hematology, Department of Medicine, Stanford University School of Medicine, Stanford, CA, USA
| | - Jing Li
- Institute for Immunity, Transplantation and Infection, Stanford University School of Medicine, Stanford, CA, USA
| | - Hongjie Dai
- Department of Chemistry, Stanford University School of Medicine, Stanford, CA, USA
| | - Zachary M Sellers
- Department of Pediatrics, Stanford University School of Medicine, Stanford, CA, USA
| | - Howard Y Chang
- Center for Personal Dynamic Regulomes, Stanford University School of Medicine, Stanford, CA, USA
- Department of Dermatology, Stanford University School of Medicine, Stanford, CA, USA
- Department of Genetics, Stanford University School of Medicine, Stanford, CA, USA
- Howard Hughes Medical Institute, Stanford University School of Medicine, Stanford, CA, USA
| | - James C Y Dunn
- Department of Pediatric Surgery, Stanford University School of Medicine, Stanford, CA, USA
| | - Bing M Zhang
- Department of Pathology, Stanford University School of Medicine, Stanford, CA, USA
| | - Elizabeth D Mellins
- Department of Pediatrics, Stanford University School of Medicine, Stanford, CA, USA
| | - Ludvig M Sollid
- K. G. Jebsen Coeliac Disease Research Centre, Institute of Clinical Medicine, University of Oslo, Oslo, Norway
- Department of Immunology, Oslo University Hospital, Oslo, Norway
| | - Nielsen Q Fernandez-Becker
- Division of Gastroenterology and Hepatology, Department of Medicine, Stanford University School of Medicine, Stanford, CA, USA
| | - Mark M Davis
- Department of Microbiology and Immunology, Stanford University School of Medicine, Stanford, CA, USA
- Institute for Immunity, Transplantation and Infection, Stanford University School of Medicine, Stanford, CA, USA
- Howard Hughes Medical Institute, Stanford University School of Medicine, Stanford, CA, USA
| | - Calvin J Kuo
- Division of Hematology, Department of Medicine, Stanford University School of Medicine, Stanford, CA, USA.
| |
Collapse
|
25
|
Al-Mansori A, Al-Sbiei A, Bashir GH, Qureshi MM, Tariq S, Altahrawi A, al-Ramadi BK, Fernandez-Cabezudo MJ. Effect of acetylcholinesterase inhibition on immune cells in the murine intestinal mucosa. Heliyon 2024; 10:e33849. [PMID: 39071679 PMCID: PMC11283160 DOI: 10.1016/j.heliyon.2024.e33849] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2024] [Revised: 06/27/2024] [Accepted: 06/27/2024] [Indexed: 07/30/2024] Open
Abstract
The gastrointestinal tract (GI) is the largest immune organ whose function is controlled by a complex network of neurons from the enteric nervous system (ENS) as well as the sympathetic and parasympathetic system. Evolving evidence indicates that cross-communication between gut-innervating neurons and immune cells regulates many essential physiological functions including protection against mucosal infections. We previously demonstrated that following paraoxon treatment, 70 % of the mice were able to survive an oral infection with S. typhimurium, a virulent strain of Salmonella enterica serovar Typhimurium. The present study aims to investigate the effect that rivastigmine, a reversible AChE inhibitor used for the treatment of neurodegenerative diseases, has on the murine immune defenses of the intestinal mucosa. Our findings show that, similar to what is observed with paraoxon, administration of rivastigmine promoted the release of secretory granules from goblet and Paneth cells, resulting in increased mucin layer. Surprisingly, however, and unlike paraoxon, rivastigmine treatment did not affect overall mortality of infected mice. In order to investigate the mechanistic basis for the differential effects observed between paraoxon and rivastigmine, we used multi-color flowcytometric analysis to characterize the immune cell landscape in the intraepithelial (IE) and lamina propria (LP) compartments of intestinal mucosa. Our data indicate that treatment with paraoxon, but not rivastigmine, led to an increase of resident CD3+CD8+ T lymphocytes in the ileal mucosa (epithelium and lamina propria) and CD11b- CD11c+ dendritic cells in the LP. Our findings indicate the requirement for persistent cholinergic pathway engagement to effect a change in the cellular landscape of the mucosal tissue that is necessary for protection against lethal bacterial infections. Moreover, optimal protection requires a collaboration between innate and adaptive mucosal immune responses in the intestine.
Collapse
Affiliation(s)
- Alreem Al-Mansori
- Department of Biochemistry and Molecular Biology, College of Medicine and Health Sciences, United Arab University, Al-Ain, United Arab Emirates
| | - Ashraf Al-Sbiei
- Department of Biochemistry and Molecular Biology, College of Medicine and Health Sciences, United Arab University, Al-Ain, United Arab Emirates
| | - Ghada H. Bashir
- Department of Medical Microbiology and Immunology, College of Medicine and Health Sciences, United Arab University, Al-Ain, United Arab Emirates
| | - Mohammed M. Qureshi
- Department of Biochemistry and Molecular Biology, College of Medicine and Health Sciences, United Arab University, Al-Ain, United Arab Emirates
| | - Saeed Tariq
- Department of Anatomy, College of Medicine and Health Sciences, United Arab University, Al-Ain, United Arab Emirates
| | - Abeer Altahrawi
- Department of Pathology, College of Medicine and Health Sciences, United Arab University, Al-Ain, United Arab Emirates
| | - Basel K. al-Ramadi
- Department of Medical Microbiology and Immunology, College of Medicine and Health Sciences, United Arab University, Al-Ain, United Arab Emirates
- Zayed Center for Health Sciences, United Arab Emirates University, Al Ain, United Arab Emirates
| | - Maria J. Fernandez-Cabezudo
- Department of Biochemistry and Molecular Biology, College of Medicine and Health Sciences, United Arab University, Al-Ain, United Arab Emirates
- Zayed Center for Health Sciences, United Arab Emirates University, Al Ain, United Arab Emirates
| |
Collapse
|
26
|
González-Castro AM, Fernández-Bañares F, Zabana Y, Farago-Pérez G, Ortega-Barrionuevo J, Expósito E, Guagnozzi D. Microscopic Colitis and Celiac Disease: Sharing More than a Diagnostic Overlap. Nutrients 2024; 16:2233. [PMID: 39064676 PMCID: PMC11279699 DOI: 10.3390/nu16142233] [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: 06/02/2024] [Revised: 07/01/2024] [Accepted: 07/04/2024] [Indexed: 07/28/2024] Open
Abstract
Microscopic colitis (MC) is an emergent group of chronic inflammatory diseases of the colon, and celiac disease (CD) is a chronic gluten-induced immune-mediated enteropathy affecting the small bowel. We performed a narrative review to provide an overview regarding the relationship between both disorders, analyzing the most recent studies published at the epidemiological, clinical and pathophysiological levels. In fact, MC and CD are concomitantly prevalent in approximately 6% of the cases, mainly in the subset of refractory patients. Thus, physicians should screen refractory patients with CD against MC and vice versa. Both disorders share more than a simple epidemiological association, being multifactorial diseases involving innate and adaptive immune responses to known or unknown luminal factors based on a rather common genetic ground. Moreover, autoimmunity is a shared characteristic between the patients with MC and those with CD, with autoimmunity in the latter being quite well-established. Furthermore, CD and MC share some common clinical symptoms and risk factors and overlap with other gastrointestinal diseases, but some differences exist between both disorders. More studies are therefore needed to better understand the complex mechanisms involving the common pathogenetic ground contributing to the CD and MC epidemiological association.
Collapse
Affiliation(s)
- Ana María González-Castro
- Translational Mucosal Immunology Laboratory, Vall d’Hebron Institut de Recerca, 08035 Barcelona, Spain; (A.M.G.-C.); (E.E.)
- Neuro-Immuno-Gastroenterology Laboratory, Vall d’Hebron Institut de Recerca, 08035 Barcelona, Spain
| | - Fernando Fernández-Bañares
- Gastroenterology Department, University Hospital Mútua Terrassa, 08221 Terrassa, Spain (Y.Z.)
- Centro de Investigación Biomédica en Red en Enfermedades Hepáticas y Digestivas (CIBERehd, Instituto Carlos III), 28029 Madrid, Spain
| | - Yamile Zabana
- Gastroenterology Department, University Hospital Mútua Terrassa, 08221 Terrassa, Spain (Y.Z.)
- Centro de Investigación Biomédica en Red en Enfermedades Hepáticas y Digestivas (CIBERehd, Instituto Carlos III), 28029 Madrid, Spain
| | - Georgina Farago-Pérez
- Translational Mucosal Immunology Laboratory, Vall d’Hebron Institut de Recerca, 08035 Barcelona, Spain; (A.M.G.-C.); (E.E.)
| | - Jonathan Ortega-Barrionuevo
- Translational Mucosal Immunology Laboratory, Vall d’Hebron Institut de Recerca, 08035 Barcelona, Spain; (A.M.G.-C.); (E.E.)
| | - Elba Expósito
- Translational Mucosal Immunology Laboratory, Vall d’Hebron Institut de Recerca, 08035 Barcelona, Spain; (A.M.G.-C.); (E.E.)
- Neuro-Immuno-Gastroenterology Laboratory, Vall d’Hebron Institut de Recerca, 08035 Barcelona, Spain
| | - Danila Guagnozzi
- Translational Mucosal Immunology Laboratory, Vall d’Hebron Institut de Recerca, 08035 Barcelona, Spain; (A.M.G.-C.); (E.E.)
- Neuro-Immuno-Gastroenterology Laboratory, Vall d’Hebron Institut de Recerca, 08035 Barcelona, Spain
- Centro de Investigación Biomédica en Red en Enfermedades Hepáticas y Digestivas (CIBERehd, Instituto Carlos III), 28029 Madrid, Spain
- Gastroenterology Department, University Hospital Vall d’Hebron, 08035 Barcelona, Spain
| |
Collapse
|
27
|
Iijima N. The emerging role of effector functions exerted by tissue-resident memory T cells. OXFORD OPEN IMMUNOLOGY 2024; 5:iqae006. [PMID: 39193473 PMCID: PMC11213632 DOI: 10.1093/oxfimm/iqae006] [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: 11/23/2023] [Revised: 04/14/2024] [Accepted: 06/04/2024] [Indexed: 08/29/2024] Open
Abstract
The magnitude of the effector functions of memory T cells determines the consequences of the protection against invading pathogens and tumor development or the pathogenesis of autoimmune and allergic diseases. Tissue-resident memory T cells (TRM cells) are unique T-cell populations that persist in tissues for long periods awaiting re-encounter with their cognate antigen. Although TRM cell reactivation primarily requires the presentation of cognate antigens, recent evidence has shown that, in addition to the conventional concept, TRM cells can be reactivated without the presentation of cognate antigens. Non-cognate TRM cell activation is triggered by cross-reactive antigens or by several combinations of cytokines, including interleukin (IL)-2, IL-7, IL-12, IL-15 and IL-18. The activation mode of TRM cells reinforces their cytotoxic activity and promotes the secretion of effector cytokines (such as interferon-gamma and tumor necrosis factor-alpha). This review highlights the key features of TRM cell maintenance and reactivation and discusses the importance of effector functions that TRM cells exert upon being presented with cognate and/or non-cognate antigens, as well as cytokines secreted by TRM and non-TRM cells within the tissue microenvironment.
Collapse
Affiliation(s)
- Norifumi Iijima
- Center for Drug Design Research, National Institutes of Biomedical Innovation, Health and Nutrition (NIBN), Ibaraki, Osaka, Japan
| |
Collapse
|
28
|
Maurice NJ, Dalzell TS, Jarjour NN, DePauw TA, Jameson SC. Steady-state, therapeutic, and helminth-induced IL-4 compromise protective CD8 T cell bystander activation. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.06.10.598293. [PMID: 38915668 PMCID: PMC11195063 DOI: 10.1101/2024.06.10.598293] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/26/2024]
Abstract
Memory CD8 T cells (Tmem) can be activated into innate-like killers by cytokines like IL-12, IL-15, and/or IL-18; but mechanisms regulating this phenomenon (termed bystander activation) are not fully resolved. We found strain-intrinsic deficiencies in bystander activation using specific pathogen-free mice, whereby basal IL-4 signals antagonize IL-18 sensing. We show that therapeutic and helminth-induced IL-4 impairs protective bystander-mediated responses against pathogens. However, this IL-4/IL-18 axis does not completely abolish bystander activation but rather tunes the expression of direct versus indirect mediators of cytotoxicity (granzymes and interferon-γ, respectively). We show that antigen-experience overrides strain-specific deficiencies in bystander activation, leading to uniform IL-18 receptor expression and enhanced capacity for bystander activation/cytotoxicity. Our data highlight that bystander activation is not a binary process but tuned/deregulated by other cytokines that are elevated by contemporaneous infections. Further, our findings underscore the importance of antigen-experienced Tmem to dissect the contributions of bystander Tmem in health and disease.
Collapse
Affiliation(s)
- Nicholas J Maurice
- Center for Immunology, University of Minnesota Medical School, Minneapolis, MN
- Department of Laboratory Medicine and Pathology, University of Minnesota Medical School, Minneapolis, MN
| | - Talia S Dalzell
- Center for Immunology, University of Minnesota Medical School, Minneapolis, MN
- Department of Laboratory Medicine and Pathology, University of Minnesota Medical School, Minneapolis, MN
| | - Nicholas N Jarjour
- Center for Immunology, University of Minnesota Medical School, Minneapolis, MN
- Department of Laboratory Medicine and Pathology, University of Minnesota Medical School, Minneapolis, MN
| | - Taylor A DePauw
- Center for Immunology, University of Minnesota Medical School, Minneapolis, MN
- Department of Laboratory Medicine and Pathology, University of Minnesota Medical School, Minneapolis, MN
| | - Stephen C Jameson
- Center for Immunology, University of Minnesota Medical School, Minneapolis, MN
- Department of Laboratory Medicine and Pathology, University of Minnesota Medical School, Minneapolis, MN
| |
Collapse
|
29
|
Esgalhado AJ, Reste-Ferreira D, Weinhold S, Uhrberg M, Cardoso EM, Arosa FA. In vitro IL-15-activated human naïve CD8+ T cells down-modulate the CD8β chain and become CD8αα T cells. Front Immunol 2024; 15:1252439. [PMID: 38903513 PMCID: PMC11188365 DOI: 10.3389/fimmu.2024.1252439] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2023] [Accepted: 05/21/2024] [Indexed: 06/22/2024] Open
Abstract
Antigen-driven human effector-memory CD8+ T cells expressing low levels of the CD8β chain have been previously described. However, little is known on a possible antigen-independent trigger. We have examined the impact that IL-15 has on the expression of CD8β on purified human naïve CD8+ T cells after CFSE labeling and culture with IL-15. As expected, IL-15 induced naïve CD8+ T cells to proliferate and differentiate. Remarkably, the process was associated with a cell-cycle dependent down-modulation of CD8β from the cell surface, leading to the generation of CD8αβlow and CD8αβ- (i.e., CD8αα) T cells. In contrast, expression of the CD8α chain remained steady or even increased. Neither IL-2 nor IL-7 reproduced the effect of IL-15. Determination of mRNA levels for CD8α and CD8β isoforms by qPCR revealed that IL-15 promoted a significant decrease in mRNA levels of the CD8β M-4 isoform, while levels of the M-1/M-2 isoforms and of CD8α increased. Noteworthy, CD8+ T cell blasts obtained after culture of CD8+ T cells with IL-15 showed a cell-cycle dependent increase in the level of the tyrosine kinase Lck, when compared to CD8+ T cells at day 0. This study has shown for the first time that IL-15 generates CD8αα+αβlow and CD8αα+αβ- T cells containing high levels of Lck, suggesting that they may be endowed with unique functional features.
Collapse
Affiliation(s)
- André J. Esgalhado
- Health Sciences Research Centre, University of Beira Interior (CICS-UBI), Covilhã, Portugal
| | - Débora Reste-Ferreira
- Health Sciences Research Centre, University of Beira Interior (CICS-UBI), Covilhã, Portugal
| | - Sandra Weinhold
- Institute for Transplantation Diagnostics and Cell Therapeutics, Medical Faculty, University Hospital Düsseldorf, Heinrich-Heine-University, Düsseldorf, Germany
| | - Markus Uhrberg
- Institute for Transplantation Diagnostics and Cell Therapeutics, Medical Faculty, University Hospital Düsseldorf, Heinrich-Heine-University, Düsseldorf, Germany
| | - Elsa M. Cardoso
- Health Sciences Research Centre, University of Beira Interior (CICS-UBI), Covilhã, Portugal
- School of Health Sciences, Polytechnic of Guarda (ESS-IPG), Guarda, Portugal
| | - Fernando A. Arosa
- Health Sciences Research Centre, University of Beira Interior (CICS-UBI), Covilhã, Portugal
- Faculty of Health Sciences, University of Beira Interior (FCS-UBI), Covilhã, Portugal
| |
Collapse
|
30
|
Galipeau HJ, Hinterleitner R, Leonard MM, Caminero A. Non-Host Factors Influencing Onset and Severity of Celiac Disease. Gastroenterology 2024; 167:34-50. [PMID: 38286392 PMCID: PMC11653303 DOI: 10.1053/j.gastro.2024.01.030] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/10/2023] [Revised: 01/19/2024] [Accepted: 01/22/2024] [Indexed: 01/31/2024]
Abstract
Celiac disease (CeD) is a chronic autoimmune condition driven by gluten ingestion in genetically predisposed individuals, resulting in inflammatory lesions in the proximal small intestine. Although the presence of specific HLA-linked haplotypes and gluten consumption are necessary for disease development, they alone do not account for the variable onset of CeD in susceptible individuals. This review explores the multifaceted role of non-host factors in CeD development, including dietary and microbial influences. We discuss clinical associations and observations highlighting the impact of these factors on disease onset and severity. Furthermore, we discuss studies in CeD-relevant animal models that offer mechanistic insights into how diet, the microbiome, and enteric infections modulate CeD pathogenesis. Finally, we address the clinical implications and therapeutic potential of understanding these cofactors offering a promising avenue for preventive and therapeutic interventions in CeD management.
Collapse
Affiliation(s)
- Heather J Galipeau
- Farncombe Family Digestive Health Research Institute, Department of Medicine, McMaster University, Hamilton, Ontario, Canada.
| | - Reinhard Hinterleitner
- Department of Immunology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
| | - Maureen M Leonard
- Division of Pediatric Gastroenterology and Nutrition, Department of Pediatrics, MassGeneral Hospital for Children, Harvard Medical School, Boston, Massachusetts; Center for Celiac Research and Treatment, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts
| | - Alberto Caminero
- Farncombe Family Digestive Health Research Institute, Department of Medicine, McMaster University, Hamilton, Ontario, Canada
| |
Collapse
|
31
|
Abstract
The intestinal epithelium, which segregates the highly stimulatory lumen from the underlying tissue, harbors one of the largest lymphocyte populations in the body, intestinal intraepithelial lymphocytes (IELs). IELs must balance tolerance, resistance, and tissue protection to maintain epithelial homeostasis and barrier integrity. This review discusses the ontogeny, environmental imprinting, T cell receptor (TCR) repertoire, and function of intestinal IELs. Despite distinct developmental pathways, IEL subsets share core traits including an epithelium-adapted profile, innate-like properties, cytotoxic potential, and limited TCR diversity. IELs also receive important developmental and functional cues through interactions with epithelial cells, microbiota, and dietary components. The restricted TCR diversity of IELs suggests that a limited set of intestinal antigens drives IEL responses, with potential functional consequences. Finally, IELs play a key role in promoting homeostatic immunity and epithelial barrier integrity but can become pathogenic upon dysregulation. Therefore, IELs represent intriguing but underexamined therapeutic targets for inflammatory diseases and cancer.
Collapse
Affiliation(s)
- Ainsley Lockhart
- Laboratory of Mucosal Immunology, The Rockefeller University, New York, NY, USA; ,
- Current affiliation: Immunology Program, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Daniel Mucida
- Laboratory of Mucosal Immunology, The Rockefeller University, New York, NY, USA; ,
- Howard Hughes Medical Institute, The Rockefeller University, New York, NY, USA
| | - Angelina M Bilate
- Laboratory of Mucosal Immunology, The Rockefeller University, New York, NY, USA; ,
| |
Collapse
|
32
|
Abadie V, Han AS, Jabri B, Sollid LM. New Insights on Genes, Gluten, and Immunopathogenesis of Celiac Disease. Gastroenterology 2024; 167:4-22. [PMID: 38670280 PMCID: PMC11283582 DOI: 10.1053/j.gastro.2024.03.042] [Citation(s) in RCA: 15] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/12/2023] [Revised: 03/10/2024] [Accepted: 03/11/2024] [Indexed: 04/28/2024]
Abstract
Celiac disease (CeD) is a gluten-induced enteropathy that develops in genetically susceptible individuals upon consumption of cereal gluten proteins. It is a unique and complex immune disorder to study as the driving antigen is known and the tissue targeted by the immune reaction can be interrogated. This review integrates findings gained from genetic, biochemical, and immunologic studies, which together have revealed mechanisms of gluten peptide modification and HLA binding, thereby enabling a maladapted anti-gluten immune response. Observations in human samples combined with experimental mouse models have revealed that the gluten-induced immune response involves CD4+ T cells, cytotoxic CD8+ T cells, and B cells; their cross-talks are critical for the tissue-damaging response. The emergence of high-throughput technologies is increasing our understanding of the phenotype, location, and presumably function of the gluten-specific cells, which are all required to identify novel therapeutic targets and strategies for CeD.
Collapse
Affiliation(s)
- Valérie Abadie
- Department of Medicine, University of Chicago, Chicago, Illinois; Section of Gastroenterology, Nutrition and Hepatology, University of Chicago, Chicago, Illinois; Committee on Immunology, University of Chicago, Chicago, Illinois.
| | - Arnold S Han
- Columbia Center for Translational Immunology, Columbia University, New York, New York; Department of Microbiology and Immunology, Columbia University, New York, New York; Department of Medicine, Digestive and Liver Diseases, Columbia University, New York, New York
| | - Bana Jabri
- Department of Medicine, University of Chicago, Chicago, Illinois; Section of Gastroenterology, Nutrition and Hepatology, University of Chicago, Chicago, Illinois; Committee on Immunology, University of Chicago, Chicago, Illinois; Department of Pathology, University of Chicago, Chicago, Illinois; Department of Pediatrics, University of Chicago, Chicago, Illinois
| | - Ludvig M Sollid
- Norwegian Coeliac Disease Research Centre, Institute of Clinical Medicine, University of Oslo, Oslo, Norway; Department of Immunology, Oslo University Hospital-Rikshospitalet, Oslo, Norway
| |
Collapse
|
33
|
Sollid LM. Tolerance-inducing therapies in coeliac disease - mechanisms, progress and future directions. Nat Rev Gastroenterol Hepatol 2024; 21:335-347. [PMID: 38336920 DOI: 10.1038/s41575-024-00895-3] [Citation(s) in RCA: 9] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 01/08/2024] [Indexed: 02/12/2024]
Abstract
Coeliac disease is an autoinflammatory condition caused by immune reactions to cereal gluten proteins. Currently, the only available treatment for the condition is a lifelong avoidance of gluten proteins in the diet. There is an unmet need for alternative therapies. Coeliac disease has a strong association with certain HLA-DQ allotypes (DQ2.5, DQ2.2 and DQ8), and these disease-associated HLA-DQ molecules present deamidated gluten peptides to gluten-specific CD4+ T cells. The gluten-specific CD4+ T cells are the drivers of the immune reactions leading to coeliac disease. Once established, the clonotypes of gluten-specific CD4+ T cells persist for decades, explaining why patients must adhere to a gluten-free diet for life. Given the key pathogenic role of gluten-specific CD4+ T cells, tolerance-inducing therapies that target these T cells are attractive for treatment of the disorder. Lessons learned from coeliac disease might provide clues for treatment of other HLA-associated diseases for which the disease-driving antigens are unknown. Thus, intensive efforts have been and are currently implemented to bring an effective tolerance-inducing therapy for coeliac disease. This Review discusses mechanisms of the various approaches taken, summarizing the progress made, and highlights future directions in this field.
Collapse
Affiliation(s)
- Ludvig M Sollid
- Norwegian Coeliac Disease Research Centre, Institute of Clinical Medicine, University of Oslo, Oslo, Norway.
- Department of Immunology, Oslo University Hospital, Oslo, Norway.
| |
Collapse
|
34
|
Rupert PB, Buerger M, Girard EJ, Frutoso M, Parrilla D, Ng K, Gooley T, Groh V, Strong RK. Preclinical characterization of Pan-NKG2D ligand-binding NKG2D receptor decoys. Heliyon 2024; 10:e28583. [PMID: 38586421 PMCID: PMC10998067 DOI: 10.1016/j.heliyon.2024.e28583] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2023] [Revised: 03/18/2024] [Accepted: 03/20/2024] [Indexed: 04/09/2024] Open
Abstract
NKG2D and its ligands are critical regulators of protective immune responses controlling infections and cancer, defining a crucial immune signaling axis. Current therapeutic efforts targeting this axis almost exclusively aim at enhancing NKG2D-mediated effector functions. However, this axis can drive disease processes when dysregulated, in particular, driving stem-like cancer cell reprogramming and tumorigenesis through receptor/ligand self-stimulation on tumor cells. Despite complexities with its structure and biology, we developed multiple novel engineered proteins that functionally serve as axis-blocking NKG2D "decoys" and report biochemical, structural, in vitro, and in vivo evaluation of their functionality.
Collapse
Affiliation(s)
- Peter B Rupert
- Division of Basic Science, Fred Hutchinson Cancer Center, Seattle, WA, United States
| | - Matthew Buerger
- Division of Basic Science, Fred Hutchinson Cancer Center, Seattle, WA, United States
| | - Emily J Girard
- Clinical Research Division, Fred Hutchinson Cancer Center, Seattle, WA, United States
| | - Marie Frutoso
- Division of Basic Science, Fred Hutchinson Cancer Center, Seattle, WA, United States
| | - Don Parrilla
- Division of Basic Science, Fred Hutchinson Cancer Center, Seattle, WA, United States
| | - Kevin Ng
- Division of Basic Science, Fred Hutchinson Cancer Center, Seattle, WA, United States
| | - Theodore Gooley
- Division of Basic Science, Fred Hutchinson Cancer Center, Seattle, WA, United States
| | - Veronika Groh
- Division of Basic Science, Fred Hutchinson Cancer Center, Seattle, WA, United States
| | - Roland K Strong
- Division of Basic Science, Fred Hutchinson Cancer Center, Seattle, WA, United States
| |
Collapse
|
35
|
Ahmadi N, Zareinejad M, Ameri M, Mahmoudi Maymand E, Nooreddin Faraji S, Ghaderi A, Ramezani A. Enhancing cancer immunotherapy with Anti-NKG2D/IL-15(N72D)/Sushi fusion protein: Targeting cytotoxic immune cells and boosting IL-15 efficacy. Cytokine 2024; 176:156505. [PMID: 38301357 DOI: 10.1016/j.cyto.2024.156505] [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: 10/18/2023] [Revised: 01/07/2024] [Accepted: 01/12/2024] [Indexed: 02/03/2024]
Abstract
BACKGROUND There are a number of distinct challenges and complexities associated with administering IL-15 for cancer immunotherapy that must be taken into consideration. OBJECTIVE The purpose of this study was to design a fusion protein for targeting cytotoxic immune cells and enhance IL-15 efficiency. METHODS A fusokine that contains IL-15(N72D), a Sushi domain, and anti-NKG2D scFv was designed. The fusion protein was in-silico modeled using the Swiss model server, followed by docking and molecular dynamics simulations. The in-vitro purified fusokine was evaluated using dot blot and Western blot. Then, flow cytometry was employed to evaluate biological properties such as proliferation, cytotoxicity, and degranulation. RESULTS Fusokine and IL-15(N72D)/Sushi, which had molecular weights of about 52 kDa and 26 kDa, respectively, were expressed in CHO-K1 cells. The fusokine binds 69.6 % of the CHO-NKG2D+ cells that express 83.1 % NKG2D. Both the fusokine and the IL-15(N72D)/Sushi significantly stimulate the proliferation of lymphocytes. After 14 days of growth, the vitality of untreated cells decreased to about 17.5 %, but 82.2 % and 56.6 % of cells were still alive when fusokine and IL-15(N72D)/Sushi were present. Furthermore, administration of fusokine was associated with the highest rates of target tumor cell cytotoxicity. Additionally, although it was not statistically significant, fusokine increased the expression of CD107a and granzyme B by 1.25 times and 2.4 times, respectively. CONCLUSION The fusokine possesses the capability to stimulate the survival and multiplication of lymphocytes, as well as their ability to eliminate tumors. These characteristics have led to its consideration as a potential treatment for immunotherapy.
Collapse
Affiliation(s)
- Nahid Ahmadi
- Department of Medical Biotechnology, School of Advanced Medical Sciences and Technologies, Shiraz University of Medical Sciences, Shiraz, Iran; Shiraz Institute for Cancer Research, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Mohammadrasul Zareinejad
- Shiraz Institute for Cancer Research, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Mehrdad Ameri
- Department of Medical Biotechnology, School of Advanced Medical Sciences and Technologies, Shiraz University of Medical Sciences, Shiraz, Iran; Shiraz Institute for Cancer Research, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Elham Mahmoudi Maymand
- Shiraz Institute for Cancer Research, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Seyed Nooreddin Faraji
- Department of Pathology, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Abbas Ghaderi
- Shiraz Institute for Cancer Research, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Amin Ramezani
- Department of Medical Biotechnology, School of Advanced Medical Sciences and Technologies, Shiraz University of Medical Sciences, Shiraz, Iran; Shiraz Institute for Cancer Research, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran.
| |
Collapse
|
36
|
Passerini L, Amodio G, Bassi V, Vitale S, Mottola I, Di Stefano M, Fanti L, Sgaramella P, Ziparo C, Furio S, Auricchio R, Barera G, Di Nardo G, Troncone R, Gianfrani C, Gregori S. IL-10-producing regulatory cells impact on celiac disease evolution. Clin Immunol 2024; 260:109923. [PMID: 38316201 PMCID: PMC10905269 DOI: 10.1016/j.clim.2024.109923] [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: 10/13/2023] [Revised: 01/19/2024] [Accepted: 02/01/2024] [Indexed: 02/07/2024]
Abstract
Celiac Disease (CD) is a T-cell mediated disorder caused by immune response to gluten, although the mechanisms underlying CD progression are still elusive. We analyzed immune cell composition, plasma cytokines, and gliadin-specific T-cell responses in patients with positive serology and normal intestinal mucosa (potential-CD) or villous atrophy (acute-CD), and after gluten-free diet (GFD). We found: an inflammatory signature and the presence of circulating gliadin-specific IFN-γ+ T cells in CD patients regardless of mucosal damage; an increased frequency of IL-10-secreting dendritic cells (DC-10) in the gut and of circulating gliadin-specific IL-10-secreting T cells in potential-CD; IL-10 inhibition increased IFN-γ secretion by gliadin-specific intestinal T cells from acute- and potential-CD. On GFD, inflammatory cytokines normalized, while IL-10-producing T cells accumulated in the gut. We show that IL-10-producing cells are fundamental in controlling pathological T-cell responses to gluten: DC-10 protect the intestinal mucosa from damage and represent a marker of potential-CD.
Collapse
Affiliation(s)
- Laura Passerini
- Mechanisms of Peripheral Tolerance Unit, San Raffaele Telethon Institute for Gene Therapy (SR-Tiget), IRCCS San Raffaele Scientific Institute, Via Olgettina 60, Milan 20132, Italy
| | - Giada Amodio
- Mechanisms of Peripheral Tolerance Unit, San Raffaele Telethon Institute for Gene Therapy (SR-Tiget), IRCCS San Raffaele Scientific Institute, Via Olgettina 60, Milan 20132, Italy
| | - Virginia Bassi
- Mechanisms of Peripheral Tolerance Unit, San Raffaele Telethon Institute for Gene Therapy (SR-Tiget), IRCCS San Raffaele Scientific Institute, Via Olgettina 60, Milan 20132, Italy
| | - Serena Vitale
- Institute of Biochemistry and Cell Biology, CNR, Via P. Castellino 111, Naples 80131, Italy
| | - Ilaria Mottola
- Institute of Biochemistry and Cell Biology, CNR, Via P. Castellino 111, Naples 80131, Italy
| | - Marina Di Stefano
- Department of Paediatrics, IRCCS San Raffaele Scientific Institute, Via Olgettina 60, Milan 20132, Italy
| | - Lorella Fanti
- Gastroenterology and Gastrointestinal Endoscopy Unit, IRCCS San Raffaele Scientific Institute, Via Olgettina 60, Milan 20132, Italy
| | - Paola Sgaramella
- Department of Paediatrics, IRCCS San Raffaele Scientific Institute, Via Olgettina 60, Milan 20132, Italy
| | - Chiara Ziparo
- NESMOS Department, School of Medicine and Psychology, Sapienza University of Rome, Sant' Andrea University Hospital, Via di Grottarossa 1035, Rome 00189, Italy
| | - Silvia Furio
- NESMOS Department, School of Medicine and Psychology, Sapienza University of Rome, Sant' Andrea University Hospital, Via di Grottarossa 1035, Rome 00189, Italy
| | - Renata Auricchio
- European Laboratory for the Investigation of Food Induced Diseases (ELFID), Department of Translational Medical Science, Section of Pediatrics, Via Pansini 5, University Federico II, Naples 80131, Italy
| | - Graziano Barera
- Department of Paediatrics, IRCCS San Raffaele Scientific Institute, Via Olgettina 60, Milan 20132, Italy
| | - Giovanni Di Nardo
- NESMOS Department, School of Medicine and Psychology, Sapienza University of Rome, Sant' Andrea University Hospital, Via di Grottarossa 1035, Rome 00189, Italy
| | - Riccardo Troncone
- European Laboratory for the Investigation of Food Induced Diseases (ELFID), Department of Translational Medical Science, Section of Pediatrics, Via Pansini 5, University Federico II, Naples 80131, Italy
| | - Carmen Gianfrani
- Institute of Biochemistry and Cell Biology, CNR, Via P. Castellino 111, Naples 80131, Italy
| | - Silvia Gregori
- Mechanisms of Peripheral Tolerance Unit, San Raffaele Telethon Institute for Gene Therapy (SR-Tiget), IRCCS San Raffaele Scientific Institute, Via Olgettina 60, Milan 20132, Italy.
| |
Collapse
|
37
|
Roque A, Pereira SG. Bacteria: Potential Make-or-Break Determinants of Celiac Disease. Int J Mol Sci 2024; 25:2090. [PMID: 38396767 PMCID: PMC10889687 DOI: 10.3390/ijms25042090] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2023] [Revised: 12/25/2023] [Accepted: 12/28/2023] [Indexed: 02/25/2024] Open
Abstract
Celiac disease is an autoimmune disease triggered by dietary gluten in genetically susceptible individuals that primarily affects the small intestinal mucosa. The sole treatment is a gluten-free diet that places a social and economic burden on patients and fails, in some, to lead to symptomatic or mucosal healing. Thus, an alternative treatment has long been sought after. Clinical studies on celiac disease have shown an association between the presence of certain microbes and disease outcomes. However, the mechanisms that underlie the effects of microbes in celiac disease remain unclear. Recent studies have employed disease models that have provided insights into disease mechanisms possibly mediated by bacteria in celiac disease. Here, we have reviewed the bacteria and related mechanisms identified so far that might protect from or incite the development of celiac disease. Evidence indicates bacteria play a role in celiac disease and it is worth continuing to explore this, particularly since few studies, to the best of our knowledge, have focused on establishing a mechanistic link between bacteria and celiac disease. Uncovering host-microbe interactions and their influence on host responses to gluten may enable the discovery of pathogenic targets and development of new therapeutic or preventive approaches.
Collapse
Affiliation(s)
| | - Sónia Gonçalves Pereira
- Center for Innovative Care and Health Technology (ciTechCare), School of Health Sciences, Polytechnic of Leiria, 2410-541 Leiria, Portugal;
| |
Collapse
|
38
|
Balint E, Feng E, Giles EC, Ritchie TM, Qian AS, Vahedi F, Montemarano A, Portillo AL, Monteiro JK, Trigatti BL, Ashkar AA. Bystander activated CD8 + T cells mediate neuropathology during viral infection via antigen-independent cytotoxicity. Nat Commun 2024; 15:896. [PMID: 38316762 PMCID: PMC10844499 DOI: 10.1038/s41467-023-44667-0] [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: 02/09/2022] [Accepted: 12/21/2023] [Indexed: 02/07/2024] Open
Abstract
Although many viral infections are linked to the development of neurological disorders, the mechanism governing virus-induced neuropathology remains poorly understood, particularly when the virus is not directly neuropathic. Using a mouse model of Zika virus (ZIKV) infection, we found that the severity of neurological disease did not correlate with brain ZIKV titers, but rather with infiltration of bystander activated NKG2D+CD8+ T cells. Antibody depletion of CD8 or blockade of NKG2D prevented ZIKV-associated paralysis, suggesting that CD8+ T cells induce neurological disease independent of TCR signaling. Furthermore, spleen and brain CD8+ T cells exhibited antigen-independent cytotoxicity that correlated with NKG2D expression. Finally, viral infection and inflammation in the brain was necessary but not sufficient to induce neurological damage. We demonstrate that CD8+ T cells mediate virus-induced neuropathology via antigen-independent, NKG2D-mediated cytotoxicity, which may serve as a therapeutic target for treatment of virus-induced neurological disease.
Collapse
Affiliation(s)
- Elizabeth Balint
- McMaster Immunology Research Centre, Department of Medicine, McMaster University, Hamilton, ON, Canada
| | - Emily Feng
- McMaster Immunology Research Centre, Department of Medicine, McMaster University, Hamilton, ON, Canada
| | - Elizabeth C Giles
- McMaster Immunology Research Centre, Department of Medicine, McMaster University, Hamilton, ON, Canada
| | - Tyrah M Ritchie
- McMaster Immunology Research Centre, Department of Medicine, McMaster University, Hamilton, ON, Canada
| | - Alexander S Qian
- Thrombosis and Atherosclerosis Research Institute, Department of Biochemistry and Biomedical Sciences, McMaster University, Hamilton Health Sciences, Hamilton, ON, Canada
| | - Fatemeh Vahedi
- McMaster Immunology Research Centre, Department of Medicine, McMaster University, Hamilton, ON, Canada
| | - Amelia Montemarano
- McMaster Immunology Research Centre, Department of Medicine, McMaster University, Hamilton, ON, Canada
| | - Ana L Portillo
- McMaster Immunology Research Centre, Department of Medicine, McMaster University, Hamilton, ON, Canada
| | - Jonathan K Monteiro
- McMaster Immunology Research Centre, Department of Medicine, McMaster University, Hamilton, ON, Canada
| | - Bernardo L Trigatti
- Thrombosis and Atherosclerosis Research Institute, Department of Biochemistry and Biomedical Sciences, McMaster University, Hamilton Health Sciences, Hamilton, ON, Canada
| | - Ali A Ashkar
- McMaster Immunology Research Centre, Department of Medicine, McMaster University, Hamilton, ON, Canada.
| |
Collapse
|
39
|
Lee H, Park SH, Shin EC. IL-15 in T-Cell Responses and Immunopathogenesis. Immune Netw 2024; 24:e11. [PMID: 38455459 PMCID: PMC10917573 DOI: 10.4110/in.2024.24.e11] [Citation(s) in RCA: 13] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2024] [Revised: 02/01/2024] [Accepted: 02/01/2024] [Indexed: 03/09/2024] Open
Abstract
IL-15 belongs to the common gamma chain cytokine family and has pleiotropic immunological functions. IL-15 is a homeostatic cytokine essential for the development and maintenance of NK cells and memory CD8+ T cells. In addition, IL-15 plays a critical role in the activation, effector functions, tissue residency, and senescence of CD8+ T cells. IL-15 also activates virtual memory T cells, mucosal-associated invariant T cells and γδ T cells. Recently, IL-15 has been highlighted as a major trigger of TCR-independent activation of T cells. This mechanism is involved in T cell-mediated immunopathogenesis in diverse diseases, including viral infections and chronic inflammatory diseases. Deeper understanding of IL-15-mediated T-cell responses and their underlying mechanisms could optimize therapeutic strategies to ameliorate host injury by T cell-mediated immunopathogenesis. This review highlights recent advancements in comprehending the role of IL-15 in relation to T cell responses and immunopathogenesis under various host conditions.
Collapse
Affiliation(s)
- Hoyoung Lee
- The Center for Viral Immunology, Korea Virus Research Institute, Institute for Basic Science (IBS), Daejeon 34126, Korea
| | - Su-Hyung Park
- Graduate School of Medical Science and Engineering, Korea Advanced Institute of Science and Technology (KAIST), Daejeon 34141, Korea
| | - Eui-Cheol Shin
- The Center for Viral Immunology, Korea Virus Research Institute, Institute for Basic Science (IBS), Daejeon 34126, Korea
- Graduate School of Medical Science and Engineering, Korea Advanced Institute of Science and Technology (KAIST), Daejeon 34141, Korea
| |
Collapse
|
40
|
Lee LW, Shafiani S, Crossley B, Emerson RO, Williamson D, Bunin A, Vargas J, Han AS, Kaplan IM, Green PHR, Kirsch I, Bhagat G. Characterisation of T cell receptor repertoires in coeliac disease. J Clin Pathol 2024; 77:116-124. [PMID: 36522177 PMCID: PMC10850686 DOI: 10.1136/jcp-2022-208541] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2022] [Accepted: 11/23/2022] [Indexed: 12/16/2022]
Abstract
AIMS Characterise T-cell receptor gene (TR) repertoires of small intestinal T cells of patients with newly diagnosed (active) coeliac disease (ACD), refractory CD type I (RCD I) and patients with CD on a gluten-free diet (GFD). METHODS Next-generation sequencing of complementarity-determining region 3 (CDR3) of rearranged T cell receptor β (TRB) and γ (TRG) genes was performed using DNA extracted from intraepithelial cell (IEC) and lamina propria cell (LPC) fractions and a small subset of peripheral blood mononuclear cell (PBMC) samples obtained from CD and non-CD (control) patients. Several parameters were assessed, including relative abundance and enrichment. RESULTS TRB and TRG repertoires of CD IEC and LPC samples demonstrated lower clonality but higher frequency of rearranged TRs compared with controls. No CD-related differences were detected in the limited number of PBMC samples. Previously published LP gliadin-specific TRB sequences were more frequently detected in LPC samples from patients with CD compared with non-CD controls. TRG repertoires of IECs from both ACD and GFD patients demonstrated increased abundance of certain CDR3 amino acid (AA) motifs compared with controls, which were encoded by multiple nucleotide variants, including one motif that was enriched in duodenal IECs versus the PBMCs of CD patients. CONCLUSIONS Small intestinal TRB and TRG repertoires of patients with CD are more diverse than individuals without CD, likely due to mucosal recruitment and accumulation of T cells because of protracted inflammation. Enrichment of the unique TRG CDR3 AA sequence in the mucosa of patients with CD may suggest disease-associated changes in the TCRγδ IE lymphocyte (IEL) landscape.
Collapse
Affiliation(s)
- Lik Wee Lee
- Computational Biology and Translational Medicine, Adaptive Biotechnologies Corp, Seattle, Washington, USA
| | - Shahin Shafiani
- Computational Biology and Translational Medicine, Adaptive Biotechnologies Corp, Seattle, Washington, USA
| | - Beryl Crossley
- Computational Biology and Translational Medicine, Adaptive Biotechnologies Corp, Seattle, Washington, USA
| | - Ryan O Emerson
- Computational Biology and Translational Medicine, Adaptive Biotechnologies Corp, Seattle, Washington, USA
| | - David Williamson
- Computational Biology and Translational Medicine, Adaptive Biotechnologies Corp, Seattle, Washington, USA
| | - Anna Bunin
- Department of Medicine, Celiac Disease Center, Columbia University Irving Medical Center, New York, New York, USA
| | - Justin Vargas
- Department of Medicine, Celiac Disease Center, Columbia University Irving Medical Center, New York, New York, USA
| | - Arnold S Han
- Department of Medicine, Celiac Disease Center, Columbia University Irving Medical Center, New York, New York, USA
| | - Ian M Kaplan
- Computational Biology and Translational Medicine, Adaptive Biotechnologies Corp, Seattle, Washington, USA
| | - Peter H R Green
- Department of Medicine, Celiac Disease Center, Columbia University Irving Medical Center, New York, New York, USA
| | - Ilan Kirsch
- Computational Biology and Translational Medicine, Adaptive Biotechnologies Corp, Seattle, Washington, USA
| | - Govind Bhagat
- Department of Pathology and Cell Biology and Department of Medicine, Celiac Disease Center, Columbia University Irving Medical Center, New York, New York, USA
| |
Collapse
|
41
|
Wei L, Xiang Z, Zou Y. The Role of NKG2D and Its Ligands in Autoimmune Diseases: New Targets for Immunotherapy. Int J Mol Sci 2023; 24:17545. [PMID: 38139373 PMCID: PMC10744089 DOI: 10.3390/ijms242417545] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2023] [Revised: 12/08/2023] [Accepted: 12/13/2023] [Indexed: 12/24/2023] Open
Abstract
Natural killer (NK) cells and CD8+ T cells can clear infected and transformed cells and generate tolerance to themselves, which also prevents autoimmune diseases. Natural killer group 2 member D (NKG2D) is an important activating immune receptor that is expressed on NK cells, CD8+ T cells, γδ T cells, and a very small percentage of CD4+ T cells. In contrast, the NKG2D ligand (NKG2D-L) is generally not expressed on normal cells but is overexpressed under stress. Thus, the inappropriate expression of NKG2D-L leads to the activation of self-reactive effector cells, which can trigger or exacerbate autoimmunity. In this review, we discuss the role of NKG2D and NKG2D-L in systemic lupus erythematosus (SLE), rheumatoid arthritis (RA), multiple sclerosis (MS), type I diabetes (T1DM), inflammatory bowel disease (IBD), and celiac disease (CeD). The data suggest that NKG2D and NKG2D-L play a pathogenic role in some autoimmune diseases. Therefore, the development of strategies to block the interaction of NKG2D and NKG2D-L may have therapeutic effects in some autoimmune diseases.
Collapse
Affiliation(s)
| | | | - Yizhou Zou
- Department of Immunology, School of Basic Medical, Central South University, Changsha 410083, China; (L.W.); (Z.X.)
| |
Collapse
|
42
|
Ruera CN, Perez F, Iribarren ML, Guzman L, Menendez L, Garbi L, Chirdo FG. Coexistence of apoptosis, pyroptosis, and necroptosis pathways in celiac disease. Clin Exp Immunol 2023; 214:328-340. [PMID: 37455655 PMCID: PMC10719221 DOI: 10.1093/cei/uxad082] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2023] [Revised: 06/12/2023] [Accepted: 07/14/2023] [Indexed: 07/18/2023] Open
Abstract
Usually, the massive elimination of cells under steady-state conditions occurs by apoptosis, which is also acknowledged to explain the loss of enterocytes in the small intestine of celiac disease (CD) patients. However, little is known about the role of proinflammatory cell death pathways in CD. Here, we have used confocal microscopy, western blot, and RT-qPCR analysis to assess the presence of regulated cell death pathways in the duodenum of CD patients. We found an increased number of dead (TUNEL+) cells in the lamina propria of small intestine of CD patients, most of them are plasma cells (CD138+). Many dying cells expressed FAS and were in close contact with CD3+ T cells. Caspase-8 and caspase-3 expression was increased in CD, confirming the activation of apoptosis. In parallel, caspase-1, IL-1β, and GSDMD were increased in CD samples indicating the presence of inflammasome-dependent pyroptosis. Necroptosis was also present, as shown by the increase of RIPK3 and phosphorylate MLKL. Analysis of published databases confirmed that CD has an increased expression of regulated cell death -related genes. Together, these results reveal that CD is characterized by cell death of different kinds. In particular, the presence of proinflammatory cell death pathways may contribute to mucosal damage.
Collapse
Affiliation(s)
- Carolina N Ruera
- Instituto de Estudios Inmunológicos y Fisiopatológicos (IIFP), (UNLP-CONICET-CIC) Facultad de Ciencias Exactas, Universidad Nacional de La Plata, La Plata, Argentina
| | - Federico Perez
- Instituto de Estudios Inmunológicos y Fisiopatológicos (IIFP), (UNLP-CONICET-CIC) Facultad de Ciencias Exactas, Universidad Nacional de La Plata, La Plata, Argentina
| | - María Luz Iribarren
- Instituto de Estudios Inmunológicos y Fisiopatológicos (IIFP), (UNLP-CONICET-CIC) Facultad de Ciencias Exactas, Universidad Nacional de La Plata, La Plata, Argentina
| | - Luciana Guzman
- Servicio de Gastroenterología Hospital de Niños “Sor María Ludovica”, La Plata, Argentina
| | - Lorena Menendez
- Servicio de Gastroenterología Hospital de Niños “Sor María Ludovica”, La Plata, Argentina
| | - Laura Garbi
- Servicio de Gastroenterología, HospitalSan Martin, La Plata, Argentina
| | - Fernando G Chirdo
- Instituto de Estudios Inmunológicos y Fisiopatológicos (IIFP), (UNLP-CONICET-CIC) Facultad de Ciencias Exactas, Universidad Nacional de La Plata, La Plata, Argentina
| |
Collapse
|
43
|
Besser HA, Khosla C. Celiac disease: mechanisms and emerging therapeutics. Trends Pharmacol Sci 2023; 44:949-962. [PMID: 37839914 PMCID: PMC10843302 DOI: 10.1016/j.tips.2023.09.006] [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: 09/01/2023] [Revised: 09/14/2023] [Accepted: 09/20/2023] [Indexed: 10/17/2023]
Abstract
Celiac disease (CeD) is a widespread, gluten-induced, autoimmune disorder that lacks any medicinal therapy. Towards the goal of developing non-dietary treatments for CeD, research has focused on elucidating its molecular and cellular etiology. A model of pathogenesis has emerged centered on interactions between three molecular families: specific class II MHC proteins on antigen-presenting cells (APCs), deamidated gluten-derived peptides, and T cell receptors (TCRs) on inflammatory CD4+ T cells. Growing evidence suggests that this pathogenic axis can be pharmacologically targeted to protect patients from some of the adverse effects of dietary gluten. Further studies have revealed the existence of additional host and environmental contributors to disease initiation and tissue damage. This review summarizes our current understanding of CeD pathogenesis and how it is being harnessed for therapeutic design and development.
Collapse
Affiliation(s)
- Harrison A Besser
- Department of Chemistry, Stanford University, Stanford, CA 94305, USA; Stanford Medical Scientist Training Program, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Chaitan Khosla
- Department of Chemistry, Stanford University, Stanford, CA 94305, USA; Department of Chemical Engineering, Stanford University, Stanford, CA 94305, USA; Sarafan ChEM-H (Chemistry, Engineering and Medicine for Human Health), Stanford University, Stanford, CA 94305, USA.
| |
Collapse
|
44
|
Wang Q, Lu Q, Jia S, Zhao M. Gut immune microenvironment and autoimmunity. Int Immunopharmacol 2023; 124:110842. [PMID: 37643491 DOI: 10.1016/j.intimp.2023.110842] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2023] [Revised: 08/14/2023] [Accepted: 08/19/2023] [Indexed: 08/31/2023]
Abstract
A variety of immune cells or tissues are present in the gut to form the gut immune microenvironment by interacting with gut microbiota, and to maintain the gut immune homeostasis. Accumulating evidence indicated that gut microbiota dysbiosis might break the homeostasis of the gut immune microenvironment, which was associated with many health problems including autoimmune diseases. Moreover, disturbance of the gut immune microenvironment can also induce extra-intestinal autoimmune disorders through the migration of intestinal pro-inflammatory effector cells from the intestine to peripheral inflamed sites. This review discussed the composition of the gut immune microenvironment and its association with autoimmunity. These findings are expected to provide new insights into the pathogenesis of various autoimmune disorders, as well as novel strategies for the prevention and treatment against related diseases.
Collapse
Affiliation(s)
- Qiaolin Wang
- Hospital for Skin Diseases, Institute of Dermatology, Chinese Academy of Medical Sciences and Peking Union Medical College, Nanjing 210042, China; Key Laboratory of Basic and Translational Research on Immune-Mediated Skin Diseases, Chinese Academy of Medical Sciences, Nanjing 210042, China
| | - Qianjin Lu
- Hospital for Skin Diseases, Institute of Dermatology, Chinese Academy of Medical Sciences and Peking Union Medical College, Nanjing 210042, China; Key Laboratory of Basic and Translational Research on Immune-Mediated Skin Diseases, Chinese Academy of Medical Sciences, Nanjing 210042, China
| | - Sujie Jia
- Department of Pharmacy, Chinese Academy of Medical Sciences and Peking Union Medical College, Nanjing 210042, China.
| | - Ming Zhao
- Hospital for Skin Diseases, Institute of Dermatology, Chinese Academy of Medical Sciences and Peking Union Medical College, Nanjing 210042, China; Key Laboratory of Basic and Translational Research on Immune-Mediated Skin Diseases, Chinese Academy of Medical Sciences, Nanjing 210042, China.
| |
Collapse
|
45
|
Seitz V, Gennermann K, Elezkurtaj S, Groth D, Schaper S, Dröge A, Lachmann N, Berg E, Lenze D, Kühl AA, Husemann C, Kleo K, Horst D, Lennerz V, Hennig S, Hummel M, Schumann M. Specific T-cell receptor beta-rearrangements of gluten-triggered CD8 + T-cells are enriched in celiac disease patients' duodenal mucosa. Clin Immunol 2023; 256:109795. [PMID: 37769786 DOI: 10.1016/j.clim.2023.109795] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2023] [Revised: 09/12/2023] [Accepted: 09/25/2023] [Indexed: 10/02/2023]
Abstract
Celiac disease (CeD) is an autoimmune disorder affecting the small intestine with gluten as disease trigger. Infections including Influenza A, increase the CeD risk. While gluten-specific CD4+ T-cells, recognizing HLA-DQ2/DQ8 presented gluten-peptides, initiate and sustain the celiac immune response, CD8+ α/β intraepithelial T-cells elicit mucosal damage. Here, we subjected TCRs from a cohort of 56 CeD patients and 22 controls to an analysis employing 749 published CeD-related TCRβ-rearrangements derived from gluten-specific CD4+ T-cells and gluten-triggered peripheral blood CD8+ T-cells. We show, that in addition to TCRs from gluten-specific CD4+ T-cells, TCRs of gluten-triggered CD8+ T-cells are significantly enriched in CeD duodenal tissue samples. TCRβ-rearrangements of gluten-triggered CD8+ T-cells were even more expanded in patients than TCRs from gluten-specific CD4+ T-cells (p < 0.0002) and highest in refractory CeD. Sequence alignments with TCR-antigen databases suggest that a subgroup of these most likely indirectly gluten-triggered TCRs recognize microbial, viral, and autoantigens.
Collapse
Affiliation(s)
- V Seitz
- Institute of Pathology, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany; HS Diagnomics GmbH, Berlin, Germany
| | | | - S Elezkurtaj
- Institute of Pathology, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
| | - D Groth
- Bioinformatics, Institute of Biochemistry and Biology, University of Potsdam, Potsdam, Germany
| | | | - A Dröge
- HS Diagnomics GmbH, Berlin, Germany
| | - N Lachmann
- Centre for Tumor Medicine, Histocompatibility & Immunogenetics Laboratory, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
| | - E Berg
- Institute of Pathology, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
| | - D Lenze
- Institute of Pathology, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
| | - A A Kühl
- iPATH.Berlin - Core Unit of the Charité Universitätsmedizin Berlin, corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
| | - C Husemann
- Institute of Pathology, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany; German Cancer Consortium (DKTK), Partner Site Berlin, Berlin, Germany; German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - K Kleo
- Institute of Pathology, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
| | - D Horst
- Institute of Pathology, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
| | | | - S Hennig
- HS Diagnomics GmbH, Berlin, Germany
| | - M Hummel
- Institute of Pathology, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany; German Cancer Consortium (DKTK), Partner Site Berlin, Berlin, Germany; German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - M Schumann
- Medizinische Klinik m. S. Gastroenterologie, Infektiologie und Rheumatologie, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany.
| |
Collapse
|
46
|
Lutz S, Klausz K, Albici AM, Ebinger L, Sellmer L, Teipel H, Frenzel A, Langner A, Winterberg D, Krohn S, Hust M, Schirrmann T, Dübel S, Scherließ R, Humpe A, Gramatzki M, Kellner C, Peipp M. Novel NKG2D-directed bispecific antibodies enhance antibody-mediated killing of malignant B cells by NK cells and T cells. Front Immunol 2023; 14:1227572. [PMID: 37965326 PMCID: PMC10641740 DOI: 10.3389/fimmu.2023.1227572] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2023] [Accepted: 10/05/2023] [Indexed: 11/16/2023] Open
Abstract
The activating receptor natural killer group 2, member D (NKG2D) represents an attractive target for immunotherapy as it exerts a crucial role in cancer immunosurveillance by regulating the activity of cytotoxic lymphocytes. In this study, a panel of novel NKG2D-specific single-chain fragments variable (scFv) were isolated from naïve human antibody gene libraries and fused to the fragment antigen binding (Fab) of rituximab to obtain [CD20×NKG2D] bibodies with the aim to recruit cytotoxic lymphocytes to lymphoma cells. All bispecific antibodies bound both antigens simultaneously. Two bibody constructs, [CD20×NKG2D#3] and [CD20×NKG2D#32], efficiently activated natural killer (NK) cells in co-cultures with CD20+ lymphoma cells. Both bibodies triggered NK cell-mediated lysis of lymphoma cells and especially enhanced antibody-dependent cell-mediated cytotoxicity (ADCC) by CD38 or CD19 specific monoclonal antibodies suggesting a synergistic effect between NKG2D and FcγRIIIA signaling pathways in NK cell activation. The [CD20×NKG2D] bibodies were not effective in redirecting CD8+ T cells as single agents, but enhanced cytotoxicity when combined with a bispecific [CD19×CD3] T cell engager, indicating that NKG2D signaling also supports CD3-mediated T cell activation. In conclusion, engagement of NKG2D with bispecific antibodies is attractive to directly activate cytotoxic lymphocytes or to support their activation by monoclonal antibodies or bispecific T cell engagers. As a perspective, co-targeting of two tumor antigens may allow fine-tuning of antibody cancer therapies. Our proposed combinatorial approach is potentially applicable for many existing immunotherapies but further testing in different preclinical models is necessary to explore the full potential.
Collapse
Affiliation(s)
- Sebastian Lutz
- Department of Transfusion Medicine, Cell Therapeutics and Hemostaseology, University Hospital, Ludwig Maximilians University (LMU) Munich, Munich, Germany
- Division of Antibody-Based Immunotherapy, Department of Medicine II, Kiel University, Kiel, Germany
| | - Katja Klausz
- Division of Antibody-Based Immunotherapy, Department of Medicine II, Kiel University, Kiel, Germany
| | - Anca-Maria Albici
- Division of Antibody-Based Immunotherapy, Department of Medicine II, Kiel University, Kiel, Germany
| | - Lea Ebinger
- Division of Antibody-Based Immunotherapy, Department of Medicine II, Kiel University, Kiel, Germany
| | - Lea Sellmer
- Division of Antibody-Based Immunotherapy, Department of Medicine II, Kiel University, Kiel, Germany
| | - Hannah Teipel
- Division of Antibody-Based Immunotherapy, Department of Medicine II, Kiel University, Kiel, Germany
| | | | - Anna Langner
- Division of Antibody-Based Immunotherapy, Department of Medicine II, Kiel University, Kiel, Germany
| | - Dorothee Winterberg
- Division of Antibody-Based Immunotherapy, Department of Medicine II, Kiel University, Kiel, Germany
| | - Steffen Krohn
- Division of Antibody-Based Immunotherapy, Department of Medicine II, Kiel University, Kiel, Germany
| | - Michael Hust
- YUMAB GmbH, Braunschweig, Germany
- Technische Universität Braunschweig, Institut für Biochemie, Biotechnologie und Bioinformatik, Abteilung Biotechnologie, Braunschweig, Germany
| | | | - Stefan Dübel
- Technische Universität Braunschweig, Institut für Biochemie, Biotechnologie und Bioinformatik, Abteilung Biotechnologie, Braunschweig, Germany
| | - Regina Scherließ
- Department of Pharmaceutics and Biopharmaceutics, Kiel University, Kiel, Germany
| | - Andreas Humpe
- Department of Transfusion Medicine, Cell Therapeutics and Hemostaseology, University Hospital, Ludwig Maximilians University (LMU) Munich, Munich, Germany
| | - Martin Gramatzki
- Division of Antibody-Based Immunotherapy, Department of Medicine II, Kiel University, Kiel, Germany
| | - Christian Kellner
- Department of Transfusion Medicine, Cell Therapeutics and Hemostaseology, University Hospital, Ludwig Maximilians University (LMU) Munich, Munich, Germany
| | - Matthias Peipp
- Division of Antibody-Based Immunotherapy, Department of Medicine II, Kiel University, Kiel, Germany
| |
Collapse
|
47
|
Westmeier J, Brochtrup A, Paniskaki K, Karakoese Z, Werner T, Sutter K, Dolff S, Limmer A, Mittermüller D, Liu J, Zheng X, Koval T, Kaidashev I, Berger MM, Herbstreit F, Brenner T, Witzke O, Trilling M, Lu M, Yang D, Babel N, Westhoff T, Dittmer U, Zelinskyy G. Macrophage migration inhibitory factor receptor CD74 expression is associated with expansion and differentiation of effector T cells in COVID-19 patients. Front Immunol 2023; 14:1236374. [PMID: 37946732 PMCID: PMC10631787 DOI: 10.3389/fimmu.2023.1236374] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2023] [Accepted: 10/02/2023] [Indexed: 11/12/2023] Open
Abstract
Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) caused millions of COVID-19 cases and deaths worldwide. Severity of pulmonary pathologies and poor prognosis were reported to be associated with the activation non-virus-specific bystander T cells. In addition, high concentrations of the macrophage migration inhibitory factor (MIF) were found in serum of COVID-19 patients. We hypothesized that these two pathogenic factors might be related and analyzed the expression of receptors for MIF on T cells in COVID-19. T cells from PBMCs of hospitalized patients with mild and severe COVID-19 were characterized. A significantly higher proportion of CD4+ and CD8+ T cells from COVID-19 patients expressed CD74 on the cell surface compared to healthy controls. To induce intracellular signaling upon MIF binding, CD74 forms complexes with CD44, CXCR2, or CXCR4. The vast majority of CD74+ T cells expressed CD44, whereas expression of CXCR2 and CXCR4 was low in controls but increased upon SARS-CoV-2 infection. Hence, T cells in COVID-19 patients express receptors that render them responsive to MIF. A detailed analysis of CD74+ T cell populations revealed that most of them had a central memory phenotype early in infection, while cells with an effector and effector memory phenotype arose later during infection. Furthermore, CD74+ T cells produced more cytotoxic molecules and proliferation markers. Our data provide new insights into the MIF receptor and co-receptor repertoire of bystander T cells in COVID-19 and uncovers a novel and potentially druggable aspect of the immunological footprint of SARS-CoV-2.
Collapse
Affiliation(s)
- Jaana Westmeier
- Institute for Virology, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
| | - Annika Brochtrup
- Institute for Virology, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
| | - Krystallenia Paniskaki
- Department of Infectious Diseases, West German Centre of Infectious Diseases, University Hospital Essen, University Duisburg-Essen, Essen, Germany
- Center for Translational Medicine, Medical Department I, Marien Hospital Herne, University Hospital of the Ruhr-University Bochum, Herne, Germany
| | - Zehra Karakoese
- Institute for Virology, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
| | - Tanja Werner
- Institute for Virology, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
| | - Kathrin Sutter
- Institute for Virology, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
- Joint International Laboratory of Infection and Immunity, Huazhong University of Science and Technology (HUST), Wuhan, China
| | - Sebastian Dolff
- Department of Infectious Diseases, West German Centre of Infectious Diseases, University Hospital Essen, University Duisburg-Essen, Essen, Germany
| | - Andreas Limmer
- Department of Anesthesiology, University Hospital Essen, University Duisburg-Essen, Essen, Germany
- Department of Pediatric Heart Surgery, Friedrich-Alexander- Universität Erlangen-Nürnberg, Erlangen, Germany
| | - Daniela Mittermüller
- Institute for Virology, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
| | - Jia Liu
- Joint International Laboratory of Infection and Immunity, Huazhong University of Science and Technology (HUST), Wuhan, China
- Department of Infectious Diseases, Union Hospital of Tonji Medical College, Huazhong University of Science and Technology (HUST), Wuhan, China
| | - Xin Zheng
- Joint International Laboratory of Infection and Immunity, Huazhong University of Science and Technology (HUST), Wuhan, China
- Department of Infectious Diseases, Union Hospital of Tonji Medical College, Huazhong University of Science and Technology (HUST), Wuhan, China
| | - Tetiana Koval
- Department of Infectious Diseases with Epidemiology, Poltava State Medical University, Poltava, Ukraine
| | - Igor Kaidashev
- Department of Internal Medicine №3 with Phthisiology, Poltava State Medical University, Poltava, Ukraine
| | - Marc Moritz Berger
- Department of Anesthesiology, University Hospital Essen, University Duisburg-Essen, Essen, Germany
| | - Frank Herbstreit
- Department of Anesthesiology, University Hospital Essen, University Duisburg-Essen, Essen, Germany
| | - Thorsten Brenner
- Department of Anesthesiology, University Hospital Essen, University Duisburg-Essen, Essen, Germany
| | - Oliver Witzke
- Department of Infectious Diseases, West German Centre of Infectious Diseases, University Hospital Essen, University Duisburg-Essen, Essen, Germany
| | - Mirko Trilling
- Institute for Virology, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
- Joint International Laboratory of Infection and Immunity, Huazhong University of Science and Technology (HUST), Wuhan, China
| | - Mengji Lu
- Institute for Virology, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
- Joint International Laboratory of Infection and Immunity, Huazhong University of Science and Technology (HUST), Wuhan, China
| | - Dongliang Yang
- Joint International Laboratory of Infection and Immunity, Huazhong University of Science and Technology (HUST), Wuhan, China
- Department of Infectious Diseases, Union Hospital of Tonji Medical College, Huazhong University of Science and Technology (HUST), Wuhan, China
| | - Nina Babel
- Center for Translational Medicine, Medical Department I, Marien Hospital Herne, University Hospital of the Ruhr-University Bochum, Herne, Germany
- Charité – Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin-Brandenburg Center for Regenerative Therapies, Berlin, Germany
| | - Timm Westhoff
- Medical Department I, Marien Hospital Herne, University Hospital of the Ruhr University of Bochum, Herne, Germany
| | - Ulf Dittmer
- Institute for Virology, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
- Joint International Laboratory of Infection and Immunity, Huazhong University of Science and Technology (HUST), Wuhan, China
| | - Gennadiy Zelinskyy
- Institute for Virology, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
- Joint International Laboratory of Infection and Immunity, Huazhong University of Science and Technology (HUST), Wuhan, China
| |
Collapse
|
48
|
Belei O, Jugănaru I, Basaca DG, Munteanu AI, Mărginean O. The Role of Intestinal Microbiota in Celiac Disease and Further Therapeutic Perspectives. Life (Basel) 2023; 13:2039. [PMID: 37895421 PMCID: PMC10608277 DOI: 10.3390/life13102039] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2023] [Revised: 10/01/2023] [Accepted: 10/04/2023] [Indexed: 10/29/2023] Open
Abstract
Celiac disease (CD) is an immune-mediated enteropathy caused by exposure to gluten and related prolamins in genetically susceptible individuals. It is a complex genetic disorder with multiple contributing genes. Linkage studies have identified several genomic regions that probably contain CD susceptibility genes. The most important genetic factors are HLA-DQ2 and DQ8. Several known environmental triggers promote the onset of CD at any age after gluten introduction in individuals with a genetic background, such as viral infections and intestinal dysbiosis. Recent publications have described the interference of the intestinal microbiome in gluten metabolism, modulation of local immune reactions, and in maintaining normal gut permeability. These results have promoted further lines of research on the benefit of probiotic administration to prevent disease onset or alleviate clinical symptoms along with a gluten-free diet (GFD). The relationship between gut microbiome changes and the onset of CD is incompletely understood, still being the subject of current research. This narrative review analyzes the interplay between environmental factors, intestinal microbiome alterations, and the course of CD. Furthermore, this review sets out to discuss if modulation of intestinal microflora with pre- and probiotics along with a GFD could represent a reliable therapeutic target for celiac patients.
Collapse
Affiliation(s)
- Oana Belei
- First Pediatric Clinic, Disturbances of Growth and Development on Children Research Center, “Victor Babeș” University of Medicine and Pharmacy, 300041 Timișoara, Romania; (O.B.); (D.-G.B.); (A.I.M.); (O.M.)
- First Pediatric Clinic, “Victor Babeș” University of Medicine and Pharmacy, 300041 Timișoara, Romania
| | - Iulius Jugănaru
- First Pediatric Clinic, Disturbances of Growth and Development on Children Research Center, “Victor Babeș” University of Medicine and Pharmacy, 300041 Timișoara, Romania; (O.B.); (D.-G.B.); (A.I.M.); (O.M.)
- First Pediatric Clinic, “Victor Babeș” University of Medicine and Pharmacy, 300041 Timișoara, Romania
| | - Diana-Georgiana Basaca
- First Pediatric Clinic, Disturbances of Growth and Development on Children Research Center, “Victor Babeș” University of Medicine and Pharmacy, 300041 Timișoara, Romania; (O.B.); (D.-G.B.); (A.I.M.); (O.M.)
- First Pediatric Clinic, “Victor Babeș” University of Medicine and Pharmacy, 300041 Timișoara, Romania
| | - Andrei Ioan Munteanu
- First Pediatric Clinic, Disturbances of Growth and Development on Children Research Center, “Victor Babeș” University of Medicine and Pharmacy, 300041 Timișoara, Romania; (O.B.); (D.-G.B.); (A.I.M.); (O.M.)
- First Pediatric Clinic, “Victor Babeș” University of Medicine and Pharmacy, 300041 Timișoara, Romania
| | - Otilia Mărginean
- First Pediatric Clinic, Disturbances of Growth and Development on Children Research Center, “Victor Babeș” University of Medicine and Pharmacy, 300041 Timișoara, Romania; (O.B.); (D.-G.B.); (A.I.M.); (O.M.)
- First Pediatric Clinic, “Victor Babeș” University of Medicine and Pharmacy, 300041 Timișoara, Romania
| |
Collapse
|
49
|
Brown H, Komnick MR, Brigleb PH, Dermody TS, Esterházy D. Lymph node sharing between pancreas, gut, and liver leads to immune crosstalk and regulation of pancreatic autoimmunity. Immunity 2023; 56:2070-2085.e11. [PMID: 37557168 PMCID: PMC11040372 DOI: 10.1016/j.immuni.2023.07.008] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2022] [Revised: 05/03/2023] [Accepted: 07/12/2023] [Indexed: 08/11/2023]
Abstract
Lymph nodes (LNs) are critical sites for shaping tissue-specific adaptive immunity. However, the impact of LN sharing between multiple organs on such tailoring is less understood. Here, we describe the drainage hierarchy of the pancreas, liver, and the upper small intestine (duodenum) into three murine LNs. Migratory dendritic cells (migDCs), key in instructing adaptive immune outcome, exhibited stronger pro-inflammatory signatures when originating from the pancreas or liver than from the duodenum. Qualitatively different migDC mixing in each shared LN influenced pancreatic β-cell-reactive T cells to acquire gut-homing and tolerogenic phenotypes proportional to duodenal co-drainage. However, duodenal viral infections rendered non-intestinal migDCs and β-cell-reactive T cells more pro-inflammatory in all shared LNs, resulting in elevated pancreatic islet lymphocyte infiltration. Our study uncovers immune crosstalk through LN co-drainage as a powerful force regulating pancreatic autoimmunity.
Collapse
Affiliation(s)
- Hailey Brown
- Department of Pathology, University of Chicago, Chicago, IL, USA
| | - Macy R Komnick
- Department of Pathology, University of Chicago, Chicago, IL, USA
| | - Pamela H Brigleb
- Department of Microbiology and Molecular Genetics, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA; Institute of Infection, Inflammation, and Immunity, UPMC Children's Hospital of Pittsburgh, Pittsburgh, PA, USA
| | - Terence S Dermody
- Department of Microbiology and Molecular Genetics, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA; Institute of Infection, Inflammation, and Immunity, UPMC Children's Hospital of Pittsburgh, Pittsburgh, PA, USA; Department of Pediatrics, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Daria Esterházy
- Department of Pathology, University of Chicago, Chicago, IL, USA.
| |
Collapse
|
50
|
Farzam-Kia N, Moratalla AC, Lemaître F, Levert A, Da Cal S, Margarido C, Carpentier Solorio Y, Arbour N. GM-CSF distinctly impacts human monocytes and macrophages via ERK1/2-dependent pathways. Immunol Lett 2023; 261:47-55. [PMID: 37516253 DOI: 10.1016/j.imlet.2023.07.009] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2023] [Revised: 07/20/2023] [Accepted: 07/24/2023] [Indexed: 07/31/2023]
Abstract
Human monocytes and macrophages are two major myeloid cell subsets with similar and distinct functions in tissue homeostasis and immune responses. GM-CSF plays a fundamental role in myeloid cell differentiation and activation. Hence, we compared the effects of GM-CSF on the expression of several immune mediators by human monocytes and monocyte-derived macrophages obtained from healthy donors. We report that GM-CSF similarly elevated the expression of CD80 and ICAM-1 and reduced HLA-DR levels on both myeloid cell subsets. However, GM-CSF increased the percentage of macrophages expressing surface IL-15 but reduced the proportion of monocytes carrying surface IL-15. Moreover, GM-CSF significantly increased the secretion of IL-4, IL-6, TNF, CXCL10, and IL-27 by macrophages while reducing the secretion of IL-4 and CXCL10 by monocytes. We show that GM-CSF triggered ERK1/2, STAT3, STAT5, and SAPK/JNK pathways in both myeloid subsets. Using a pharmacological inhibitor (U0126) preventing ERK phosphorylation, we demonstrated that this pathway was involved in both the GM-CSF-induced increase and decrease of the percentage of IL-15+ macrophages and monocytes, respectively. Moreover, ERK1/2 contributed to GM-CSF-triggered secretion of IL-4, IL-6, TNF, IL-27 and CXCL10 by macrophages. However, the ERK1/2 pathway exhibited different roles in monocytes and macrophages for the GM-CSF-mediated impact on surface makers (CD80, HLA-DR, and ICAM-1). Our data demonstrate that GM-CSF stimulation induces differential responses by human monocytes and monocyte-derived macrophages and that some but not all of these effects are ERK-dependent.
Collapse
Affiliation(s)
- Negar Farzam-Kia
- Department of Neurosciences, Faculty of Medicine, Université de Montréal, QC, Canada; Centre de Recherche du Centre Hospitalier de l'Université de Montréal (CRCHUM), Montreal, QC, Canada
| | - Ana Carmena Moratalla
- Department of Neurosciences, Faculty of Medicine, Université de Montréal, QC, Canada; Centre de Recherche du Centre Hospitalier de l'Université de Montréal (CRCHUM), Montreal, QC, Canada
| | - Florent Lemaître
- Department of Neurosciences, Faculty of Medicine, Université de Montréal, QC, Canada; Centre de Recherche du Centre Hospitalier de l'Université de Montréal (CRCHUM), Montreal, QC, Canada
| | - Annie Levert
- Centre de Recherche du Centre Hospitalier de l'Université de Montréal (CRCHUM), Montreal, QC, Canada
| | - Sandra Da Cal
- Centre de Recherche du Centre Hospitalier de l'Université de Montréal (CRCHUM), Montreal, QC, Canada
| | - Clara Margarido
- Centre de Recherche du Centre Hospitalier de l'Université de Montréal (CRCHUM), Montreal, QC, Canada
| | - Yves Carpentier Solorio
- Department of Neurosciences, Faculty of Medicine, Université de Montréal, QC, Canada; Centre de Recherche du Centre Hospitalier de l'Université de Montréal (CRCHUM), Montreal, QC, Canada
| | - Nathalie Arbour
- Department of Neurosciences, Faculty of Medicine, Université de Montréal, QC, Canada; Centre de Recherche du Centre Hospitalier de l'Université de Montréal (CRCHUM), Montreal, QC, Canada.
| |
Collapse
|