1
|
Das D, Ganesh SM, Mishra A, Netea MG, Thangamani S. The Emerging Fungal Pathogen Candida auris Induces IFNγ to Colonize the Skin. PLoS Pathog 2025; 21:e1013114. [PMID: 40294061 DOI: 10.1371/journal.ppat.1013114] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2025] [Accepted: 04/09/2025] [Indexed: 04/30/2025] Open
Abstract
Candida auris is an emerging multidrug-resistant skin-tropic fungal pathogen that causes serious human infections. However, the factors that regulate C. auris skin infection in vivo are still unclear. In this study, we identified that, unlike Candida albicans, which induces IL-17-secreting protective effector Th17 cells, C. auris predominately induces IFNγ-secreting pathogenic Th1 cells during reinfection. Surprisingly, we found that IFNγ enhances skin infection of C. auris but not C. albicans. Mechanistically, IFNγ enhances skin infection of C. auris by dampening the protective IL-17 responses and increasing dermal damage. Furthermore, we identified that the development of Th1 cells occurs through IL-12, produced by C. auris-induced inflammatory macrophages and monocyte-derived dendritic cells. In addition, our findings reveal that C. auris unique cell wall outer mannan layer regulates the development of Th1 and Th17 cells. Collectively, our findings, for the first time, identified that C. auris induces IFNγ to persist in the skin. These findings help explain why C. auris but not C. albicans preferentially persist in the skin long-term, with the potential to identify novel therapeutic approaches to prevent and treat this emerging fungal pathogen in humans.
Collapse
Affiliation(s)
- Diprasom Das
- Department of Comparative Pathobiology, College of Veterinary Medicine, Purdue University, West Lafayette, Indiana, United States of America
| | - Shrihari M Ganesh
- Department of Comparative Pathobiology, College of Veterinary Medicine, Purdue University, West Lafayette, Indiana, United States of America
| | - Abtar Mishra
- Department of Comparative Pathobiology, College of Veterinary Medicine, Purdue University, West Lafayette, Indiana, United States of America
| | - Mihai G Netea
- Department of Internal Medicine and Radboud Center for Infectious Diseases, Radboud University Medical Center, Nijmegen, the Netherlands
- Department for Immunology and Metabolism, Life and Medical Sciences Institute (LIMES), University of Bonn, Bonn, Germany
| | - Shankar Thangamani
- Department of Comparative Pathobiology, College of Veterinary Medicine, Purdue University, West Lafayette, Indiana, United States of America
- Purdue Institute for Immunology, Inflammation and Infectious Diseases (PI4D), West Lafayette, Indiana, United States of America
| |
Collapse
|
2
|
Schille TB, Sprague JL, Naglik JR, Brunke S, Hube B. Commensalism and pathogenesis of Candida albicans at the mucosal interface. Nat Rev Microbiol 2025:10.1038/s41579-025-01174-x. [PMID: 40247134 DOI: 10.1038/s41579-025-01174-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/18/2025] [Indexed: 04/19/2025]
Abstract
Fungi are important and often underestimated human pathogens. Infections with fungi mostly originate from the environment, from soil or airborne spores. By contrast, Candida albicans, one of the most common and clinically important fungal pathogens, permanently exists in the vast majority of healthy individuals as a member of the human mucosal microbiota. Only under certain circumstances will these commensals cause infections. However, although the pathogenic behaviour and disease manifestation of C. albicans have been at the centre of research for many years, its asymptomatic colonization of mucosal surfaces remains surprisingly understudied. In this Review, we discuss the interplay of the fungus, the host and the microbiome on the dualism of commensal and pathogenic life of C. albicans, and how commensal growth is controlled and permitted. We explore hypotheses that could explain how the mucosal environment shapes C. albicans adaptations to its commensal lifestyle, while still maintaining or even increasing its pathogenic potential.
Collapse
Affiliation(s)
- Tim B Schille
- Department of Microbial Pathogenicity Mechanisms, Leibniz Institute for Natural Product Research and Infection Biology-Hans Knöll Institute (HKI), Jena, Germany
- Cluster of Excellence Balance of the Microverse, Friedrich Schiller University Jena, Jena, Germany
| | - Jakob L Sprague
- Department of Microbial Pathogenicity Mechanisms, Leibniz Institute for Natural Product Research and Infection Biology-Hans Knöll Institute (HKI), Jena, Germany
| | - Julian R Naglik
- Centre for Host-Microbiome Interactions, Faculty of Dentistry, Oral and Craniofacial Sciences, King's College London, London, UK
| | - Sascha Brunke
- Department of Microbial Pathogenicity Mechanisms, Leibniz Institute for Natural Product Research and Infection Biology-Hans Knöll Institute (HKI), Jena, Germany.
| | - Bernhard Hube
- Department of Microbial Pathogenicity Mechanisms, Leibniz Institute for Natural Product Research and Infection Biology-Hans Knöll Institute (HKI), Jena, Germany.
- Cluster of Excellence Balance of the Microverse, Friedrich Schiller University Jena, Jena, Germany.
- Institute of Microbiology, Friedrich Schiller University Jena, Jena, Germany.
| |
Collapse
|
3
|
Chen Y, Gao B, Cai W, Lai J, Lai K, Wang Y. Oral mucosa: anti-inflammatory function, mechanisms, and applications. J Mater Chem B 2025; 13:4059-4072. [PMID: 40062381 DOI: 10.1039/d4tb02845g] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/27/2025]
Abstract
Large soft tissue injuries require several weeks to heal and frequently leave fibrotic scars that can negatively impact tissue function. However, the applicability of traditional skin and mucous membrane transplantation for the treatment of lesions in the ocular surface and urethra is limited owing to the unique locations and functions of these tissues. Oral mucosa has been widely used in the repair of such injuries owing to its reduced propensity for inducing an inflammatory response, angiogenesis, and scarring. Enhancing chronic wound healing while avoiding scar formation requires a broader understanding of the cellular and molecular pathways that drive wound repair in the oral mucosa. This review integrates current knowledge on the mechanisms underlying the resistance of the oral mucosa to inflammation and its application as a graft material, highlighting its challenges and potential advancements. The aim of this review is to offer insights into future therapeutic strategies for wound healing and related conditions.
Collapse
Affiliation(s)
- Yani Chen
- Stomatology Hospital, School of Stomatology, Zhejiang University School of Medicine Zhejiang Provincial Clinical Research Center for Oral Diseases, Key Laboratory of Oral Biomedical Research of Zhejiang Province, Cancer Center of Zhejiang University, Engineering Research Center of Oral Biomaterials and Devices of Zhejiang Province, Hangzhou 310000, P. R. China.
| | - Bicong Gao
- Stomatology Hospital, School of Stomatology, Zhejiang University School of Medicine Zhejiang Provincial Clinical Research Center for Oral Diseases, Key Laboratory of Oral Biomedical Research of Zhejiang Province, Cancer Center of Zhejiang University, Engineering Research Center of Oral Biomaterials and Devices of Zhejiang Province, Hangzhou 310000, P. R. China.
| | - Wenjin Cai
- Stomatology Hospital, School of Stomatology, Zhejiang University School of Medicine Zhejiang Provincial Clinical Research Center for Oral Diseases, Key Laboratory of Oral Biomedical Research of Zhejiang Province, Cancer Center of Zhejiang University, Engineering Research Center of Oral Biomaterials and Devices of Zhejiang Province, Hangzhou 310000, P. R. China.
| | - Junhong Lai
- Stomatology Hospital, School of Stomatology, Zhejiang University School of Medicine Zhejiang Provincial Clinical Research Center for Oral Diseases, Key Laboratory of Oral Biomedical Research of Zhejiang Province, Cancer Center of Zhejiang University, Engineering Research Center of Oral Biomaterials and Devices of Zhejiang Province, Hangzhou 310000, P. R. China.
| | - Kaichen Lai
- Stomatology Hospital, School of Stomatology, Zhejiang University School of Medicine Zhejiang Provincial Clinical Research Center for Oral Diseases, Key Laboratory of Oral Biomedical Research of Zhejiang Province, Cancer Center of Zhejiang University, Engineering Research Center of Oral Biomaterials and Devices of Zhejiang Province, Hangzhou 310000, P. R. China.
| | - Ying Wang
- Stomatology Hospital, School of Stomatology, Zhejiang University School of Medicine Zhejiang Provincial Clinical Research Center for Oral Diseases, Key Laboratory of Oral Biomedical Research of Zhejiang Province, Cancer Center of Zhejiang University, Engineering Research Center of Oral Biomaterials and Devices of Zhejiang Province, Hangzhou 310000, P. R. China.
| |
Collapse
|
4
|
Shang L, Roffel S, Slomka V, D'Agostino EM, Metris A, Buijs MJ, Brandt BW, Deng D, Gibbs S, Krom BP. An in vitro model demonstrating homeostatic interactions between reconstructed human gingiva and a saliva-derived multispecies biofilm. MICROBIOME 2025; 13:58. [PMID: 40022258 PMCID: PMC11869481 DOI: 10.1186/s40168-025-02033-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/12/2024] [Accepted: 01/07/2025] [Indexed: 03/03/2025]
Abstract
BACKGROUND In the oral cavity, host-microbe interactions (HMI) continuously occur and greatly impact oral health. In contrast to the well-studied disease-associated HMI during, for example, periodontitis, HMI that are essential in maintaining oral health have been rarely investigated, especially in a human-relevant context. The aim of this study was to extensively characterize homeostatic HMI between saliva-derived biofilms and a reconstructed human gingiva (RHG). RHG was reconstructed following the structure of native gingiva, composed of a multilayered epithelium formed by keratinocytes and a fibroblast-populated compartment. To mimic the oral environment, RHG were inoculated with pooled human saliva resuspended in different saliva substitute media and incubated for 2 or 4 days. The co-cultured biofilms were retrieved and characterized by viable bacterial counting and compositional profiling (16S rRNA gene sequencing). RHG was investigated for metabolic activity (MTT assay), tissue histology (hematoxylin and eosin staining), epithelial proliferation (Ki67 staining), antimicrobial peptide expression, and cytokine secretion. RESULTS Viable biofilms were detected up to day 4 of co-culturing. Bacterial counts indicated biofilm growth from the inoculation to day 2 and maintained thereafter at a similar level until day 4. All biofilms shared similar composition throughout 4 days, independent of co-culture time and different saliva substitute media used during inoculation. Biofilms were diverse with Streptococcus, Haemophilus, and Neisseria being the dominating genera. While supporting biofilm development, RHG displayed no significant changes in metabolic activity, tissue histology, or epithelial proliferation. However, in the presence of biofilms, the antimicrobial peptides elafin and human β-defensin-2 were upregulated, and the secretion of cytokines IL-6, CXCL1, CXCL8, CCL5, and CCL20 increased. CONCLUSION This model mimicked homeostatic HMI where a healthy gingiva supported a viable, diverse, and stable microbial community, incorporating bacterial genera found on native gingiva. The gingiva model maintained its tissue integrity and exerted protective responses in the presence of biofilms over time. This study adds to the evidence that shows the important role of the host in maintaining homeostatic HMI that are essential for oral health. Video Abstract.
Collapse
Affiliation(s)
- Lin Shang
- Department of Preventive Dentistry, Academic Centre for Dentistry Amsterdam (ACTA), University of Amsterdam and Vrije Universiteit Amsterdam, Amsterdam, 1081 LA, The Netherlands.
| | - Sanne Roffel
- Department of Oral Cell Biology, Academic Centre for Dentistry Amsterdam (ACTA), University of Amsterdam and Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
| | | | | | - Aline Metris
- Unilever Safety and Environmental Assurance Centre (SEAC), Colworth Science Park, Sharnbrook, Bedford, UK
| | - Mark J Buijs
- Department of Preventive Dentistry, Academic Centre for Dentistry Amsterdam (ACTA), University of Amsterdam and Vrije Universiteit Amsterdam, Amsterdam, 1081 LA, The Netherlands
| | - Bernd W Brandt
- Department of Preventive Dentistry, Academic Centre for Dentistry Amsterdam (ACTA), University of Amsterdam and Vrije Universiteit Amsterdam, Amsterdam, 1081 LA, The Netherlands
| | - Dongmei Deng
- Department of Preventive Dentistry, Academic Centre for Dentistry Amsterdam (ACTA), University of Amsterdam and Vrije Universiteit Amsterdam, Amsterdam, 1081 LA, The Netherlands
| | - Susan Gibbs
- Department of Oral Cell Biology, Academic Centre for Dentistry Amsterdam (ACTA), University of Amsterdam and Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
- Department of Molecular Cell Biology and Immunology, Amsterdam University Medical Centre Location Vrije Universiteit Amsterdam, Amsterdam Infection and Immunity Institute, Amsterdam, The Netherlands
| | - Bastiaan P Krom
- Department of Preventive Dentistry, Academic Centre for Dentistry Amsterdam (ACTA), University of Amsterdam and Vrije Universiteit Amsterdam, Amsterdam, 1081 LA, The Netherlands
| |
Collapse
|
5
|
Xie D, Lu G, Mai G, Guo Q, Xu G. Tissue-resident memory T cells in diseases and therapeutic strategies. MedComm (Beijing) 2025; 6:e70053. [PMID: 39802636 PMCID: PMC11725047 DOI: 10.1002/mco2.70053] [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: 08/09/2024] [Revised: 12/05/2024] [Accepted: 12/10/2024] [Indexed: 01/16/2025] Open
Abstract
Tissue-resident memory T (TRM) cells are crucial components of the immune system that provide rapid, localized responses to recurrent pathogens at mucosal and epithelial barriers. Unlike circulating memory T cells, TRM cells are located within peripheral tissues, and they play vital roles in antiviral, antibacterial, and antitumor immunity. Their unique retention and activation mechanisms, including interactions with local epithelial cells and the expression of adhesion molecules, enable their persistence and immediate functionality in diverse tissues. Recent advances have revealed their important roles in chronic inflammation, autoimmunity, and cancer, illuminating both their protective and their pathogenic potential. This review synthesizes current knowledge on TRM cells' molecular signatures, maintenance pathways, and functional dynamics across different tissues. We also explore the interactions of TRM cells with other immune cells, such as B cells, macrophages, and dendritic cells, highlighting the complex network that underpins the efficacy of TRM cells in immune surveillance and response. Understanding the nuanced regulation of TRM cells is essential for developing targeted therapeutic strategies, including vaccines and immunotherapies, to enhance their protective roles while mitigating adverse effects. Insights into TRM cells' biology hold promise for innovative treatments for infectious diseases, cancer, and autoimmune conditions.
Collapse
Affiliation(s)
- Daoyuan Xie
- Laboratory of Translational Medicine ResearchDeyang People's Hospital of Chengdu University of Traditional Chinese MedicineDeyangChina
| | - Guanting Lu
- Laboratory of Translational Medicine ResearchDeyang People's Hospital of Chengdu University of Traditional Chinese MedicineDeyangChina
| | - Gang Mai
- Laboratory of Translational Medicine ResearchDeyang People's Hospital of Chengdu University of Traditional Chinese MedicineDeyangChina
| | - Qiuyan Guo
- State Key Laboratory for Quality Ensurance and Sustainable Use of Dao‐di Herbs, Artemisinin Research Center, Institute of Chinese Materia MedicaAcademy of Chinese Medical SciencesBeijingChina
| | - Guofeng Xu
- Inflammation & Allergic Diseases Research UnitThe Affiliated Hospital of Southwest Medical UniversityLuzhouChina
| |
Collapse
|
6
|
Kusumoto Y, Ueda M, Hashimoto M, Takeuchi H, Okada N, Yamamoto J, Nishii A, Fujino A, Kurahashi A, Satoh M, Iwasa Y, Okamura K, Obazaki K, Kumagai R, Sakamoto N, Tanaka Y, Kamiya Y, Hoshida T, Kaisho T, Hemmi H, Katakai T, Honda T, Kikuta J, Kataoka K, Ikebuchi R, Moriya T, Adachi T, Watanabe T, Ishii M, Miyawaki A, Kabashima K, Chtanova T, Tomura M. Sublingual immune cell clusters and dendritic cell distribution in the oral cavity. JCI Insight 2024; 9:e167373. [PMID: 39352752 PMCID: PMC11601585 DOI: 10.1172/jci.insight.167373] [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: 10/04/2024] Open
Abstract
The oral mucosa is the first line of defense against pathogenic bacteria and plays a vital role in maintaining tolerance to food antigens and commensal bacteria. We used CD11c reporter mice to visualize dendritic cells (DCs), a key immune cell population, in the oral cavity. We identified differences in DC density in each oral tissue region. Sublingual immune cell clusters (SLICs) extended from the lamina propria to the epithelium, where DCs and T cells resided in close contact with each other and innate lymphoid cells. Targeted in situ photolabeling revealed that the SLICs comprised mostly CD11c+CD11b+ DCs and were enriched for cDC1s and Langerhans cells. Although the frequency of T cell subsets was similar within and outside the SLICs, tissue-resident memory T cells were significantly enriched within the clusters and cluster size increased in response to inflammation. Collectively, we found that SLICs form a unique microenvironment that facilitates T cell-DC interactions in the steady state and during inflammation. Since the oral mucosa is an important target for needle-free vaccination and sublingual immunotherapy to induce tolerogenic responses, the insight into the localized immunoregulation provided in this study may accelerate the development of these approaches.
Collapse
Affiliation(s)
- Yutaka Kusumoto
- Laboratory of Immunology, Faculty of Pharmacy, Osaka Ohtani University, Tondabayashi, Osaka, Japan
| | - Mizuki Ueda
- Laboratory of Immunology, Faculty of Pharmacy, Osaka Ohtani University, Tondabayashi, Osaka, Japan
| | - Mayuko Hashimoto
- Laboratory of Immunology, Faculty of Pharmacy, Osaka Ohtani University, Tondabayashi, Osaka, Japan
| | - Haruka Takeuchi
- Laboratory of Immunology, Faculty of Pharmacy, Osaka Ohtani University, Tondabayashi, Osaka, Japan
| | - Naoko Okada
- Laboratory of Immunology, Faculty of Pharmacy, Osaka Ohtani University, Tondabayashi, Osaka, Japan
| | - Junya Yamamoto
- Laboratory of Immunology, Faculty of Pharmacy, Osaka Ohtani University, Tondabayashi, Osaka, Japan
| | - Akiko Nishii
- Laboratory of Immunology, Faculty of Pharmacy, Osaka Ohtani University, Tondabayashi, Osaka, Japan
| | - Atsuki Fujino
- Laboratory of Immunology, Faculty of Pharmacy, Osaka Ohtani University, Tondabayashi, Osaka, Japan
| | - Akiho Kurahashi
- Laboratory of Immunology, Faculty of Pharmacy, Osaka Ohtani University, Tondabayashi, Osaka, Japan
| | - Momoka Satoh
- Laboratory of Immunology, Faculty of Pharmacy, Osaka Ohtani University, Tondabayashi, Osaka, Japan
| | - Yuki Iwasa
- Laboratory of Immunology, Faculty of Pharmacy, Osaka Ohtani University, Tondabayashi, Osaka, Japan
| | - Koki Okamura
- Laboratory of Immunology, Faculty of Pharmacy, Osaka Ohtani University, Tondabayashi, Osaka, Japan
| | - Karin Obazaki
- Laboratory of Immunology, Faculty of Pharmacy, Osaka Ohtani University, Tondabayashi, Osaka, Japan
| | - Ryoto Kumagai
- Laboratory of Immunology, Faculty of Pharmacy, Osaka Ohtani University, Tondabayashi, Osaka, Japan
| | - Naruya Sakamoto
- Laboratory of Immunology, Faculty of Pharmacy, Osaka Ohtani University, Tondabayashi, Osaka, Japan
| | - Yuto Tanaka
- Laboratory of Immunology, Faculty of Pharmacy, Osaka Ohtani University, Tondabayashi, Osaka, Japan
| | - Yukika Kamiya
- Laboratory of Immunology, Faculty of Pharmacy, Osaka Ohtani University, Tondabayashi, Osaka, Japan
| | - Tetsushi Hoshida
- Laboratory for Cell Function Dynamics, RIKEN Center for Brain Science, Wako, Saitama, Japan
- Biotechnological Optics Research Team, RIKEN Center for Advanced Photonics, Wako, Saitama, Japan
| | - Tsuneyasu Kaisho
- Department of Immunology, Institute of Advanced Medicine, Wakayama Medical University, Graduate School of Medicine, Wakayama, Wakayama, Japan
| | - Hiroaki Hemmi
- Department of Immunology, Institute of Advanced Medicine, Wakayama Medical University, Graduate School of Medicine, Wakayama, Wakayama, Japan
- Laboratory of Immunology, Faculty of Veterinary Medicine, Okayama, University of Science, Imabari, Ehime, Japan
| | - Tomoya Katakai
- Department of Immunology, Graduate School of Medical and Dental Sciences, Niigata University, Niigata, Niigata, Japan
| | - Tetsuya Honda
- Department of Dermatology, Kyoto University, Graduate School of Medicine, Sakyo-ku, Kyoto, Japan
- Department of Dermatology, Hamamatsu University School of Medicine, Handayama, Hamamatsu, Japan
| | - Junichi Kikuta
- Laboratory of Immunology and Cell Biology, Graduate school of Medicine, Osaka University, Suita, Osaka, Japan
| | - Kosuke Kataoka
- Department of Oral Health Science and Social Welfare, Graduate school of Oral Sciences, Tokushima University, Tokushima, Tokushima, Japan
| | - Ryoyo Ikebuchi
- Laboratory of Immunology, Faculty of Pharmacy, Osaka Ohtani University, Tondabayashi, Osaka, Japan
- Research Fellow of Japan Society for the Promotion of Science, Japan
| | - Taiki Moriya
- Laboratory of Immunology, Faculty of Pharmacy, Osaka Ohtani University, Tondabayashi, Osaka, Japan
| | - Takahiro Adachi
- Department of Precision Health, Medical Research Institute, Tokyo Medical and Dental University, Tokyo, Japan
| | - Takeshi Watanabe
- Laboratory of Immunology, Institute for Life and Medical Sciences, Kyoto University, Sakyo-ku, Kyoto, Japan
| | - Masaru Ishii
- Laboratory of Immunology and Cell Biology, Graduate school of Medicine, Osaka University, Suita, Osaka, Japan
| | - Atsushi Miyawaki
- Laboratory for Cell Function Dynamics, RIKEN Center for Brain Science, Wako, Saitama, Japan
- Biotechnological Optics Research Team, RIKEN Center for Advanced Photonics, Wako, Saitama, Japan
| | - Kenji Kabashima
- Department of Dermatology, Kyoto University, Graduate School of Medicine, Sakyo-ku, Kyoto, Japan
| | - Tatyana Chtanova
- School of Biotechnology and Biomolecular Sciences, Faculty of Science, University of New South Wales Sydney, Kensington, New South Wales, Australia
- Immunology Theme, Garvan Institute of Medical Research, Darlinghurst, New South Wales, Australia
| | - Michio Tomura
- Laboratory of Immunology, Faculty of Pharmacy, Osaka Ohtani University, Tondabayashi, Osaka, Japan
| |
Collapse
|
7
|
You Y, Wu X, Yuan H, He Y, Chen Y, Wang S, Min H, Chen J, Li C. Crystalline silica-induced recruitment and immuno-imbalance of CD4 + tissue resident memory T cells promote silicosis progression. Commun Biol 2024; 7:971. [PMID: 39122899 PMCID: PMC11316055 DOI: 10.1038/s42003-024-06662-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2024] [Accepted: 07/31/2024] [Indexed: 08/12/2024] Open
Abstract
Occupational crystalline silica (CS) particle exposure leads to silicosis. The burden of CS-associated disease remains high, and treatment options are limited due to vague mechanisms. Here we show that pulmonary CD4+ tissue-resident memory T cells (TRM) accumulate in response to CS particles, mediating the pathogenesis of silicosis. The TRM cells are derived from peripheral lymphocyte recruitment and in situ expansion. Specifically, CD69+CD103+ TRM-Tregs depend more on circulating T cell replenishment. CD69 and CD103 can divide the TRM cells into functionally distinct subsets, mirroring the immuno-balance within CD4+ TRM cells. However, targeting CD103+ TRM-Tregs do not mitigate disease phenotype since the TRM subsets exert immunosuppressive but not pro-fibrotic roles. After identifying pathogenic CD69+CD103- subsets, we highlight IL-7 for their maintenance and function, that present a promising avenue for mitigating silicosis. Together, our findings highlight the distinct role of CD4+ TRM cells in mediating CS-induced fibrosis and provide potential therapeutic strategies.
Collapse
Affiliation(s)
- Yichuan You
- Key Laboratory of Environmental Stress and Chronic Disease Control & Prevention (China Medical University), Ministry of Education, No. 77 Puhe Road, Shenyang North New Area, Shenyang, 110122, Liaoning, PR China
- Department of Occupational and Environmental Health, School of Public Health, China Medical University, No. 77 Puhe Road, Shenyang North New Area, Shenyang, 110122, Liaoning, PR China
| | - Xiulin Wu
- Key Laboratory of Environmental Stress and Chronic Disease Control & Prevention (China Medical University), Ministry of Education, No. 77 Puhe Road, Shenyang North New Area, Shenyang, 110122, Liaoning, PR China
- Department of Occupational and Environmental Health, School of Public Health, China Medical University, No. 77 Puhe Road, Shenyang North New Area, Shenyang, 110122, Liaoning, PR China
| | - Haoyang Yuan
- Key Laboratory of Environmental Stress and Chronic Disease Control & Prevention (China Medical University), Ministry of Education, No. 77 Puhe Road, Shenyang North New Area, Shenyang, 110122, Liaoning, PR China
- Department of Occupational and Environmental Health, School of Public Health, China Medical University, No. 77 Puhe Road, Shenyang North New Area, Shenyang, 110122, Liaoning, PR China
| | - Yangyang He
- Key Laboratory of Environmental Stress and Chronic Disease Control & Prevention (China Medical University), Ministry of Education, No. 77 Puhe Road, Shenyang North New Area, Shenyang, 110122, Liaoning, PR China
- Department of Occupational and Environmental Health, School of Public Health, China Medical University, No. 77 Puhe Road, Shenyang North New Area, Shenyang, 110122, Liaoning, PR China
| | - Yinghui Chen
- Key Laboratory of Environmental Stress and Chronic Disease Control & Prevention (China Medical University), Ministry of Education, No. 77 Puhe Road, Shenyang North New Area, Shenyang, 110122, Liaoning, PR China
- Department of Occupational and Environmental Health, School of Public Health, China Medical University, No. 77 Puhe Road, Shenyang North New Area, Shenyang, 110122, Liaoning, PR China
| | - Sisi Wang
- Key Laboratory of Environmental Stress and Chronic Disease Control & Prevention (China Medical University), Ministry of Education, No. 77 Puhe Road, Shenyang North New Area, Shenyang, 110122, Liaoning, PR China
- Department of Occupational and Environmental Health, School of Public Health, China Medical University, No. 77 Puhe Road, Shenyang North New Area, Shenyang, 110122, Liaoning, PR China
| | - Hui Min
- Department of Immunology, College of Basic Medical Sciences, China Medical University, No. 77 Puhe Road, Shenyang North New Area, Shenyang, 110122, Liaoning, PR China
| | - Jie Chen
- Key Laboratory of Environmental Stress and Chronic Disease Control & Prevention (China Medical University), Ministry of Education, No. 77 Puhe Road, Shenyang North New Area, Shenyang, 110122, Liaoning, PR China.
- Department of Occupational and Environmental Health, School of Public Health, China Medical University, No. 77 Puhe Road, Shenyang North New Area, Shenyang, 110122, Liaoning, PR China.
| | - Chao Li
- Key Laboratory of Environmental Stress and Chronic Disease Control & Prevention (China Medical University), Ministry of Education, No. 77 Puhe Road, Shenyang North New Area, Shenyang, 110122, Liaoning, PR China.
- Department of Occupational and Environmental Health, School of Public Health, China Medical University, No. 77 Puhe Road, Shenyang North New Area, Shenyang, 110122, Liaoning, PR China.
| |
Collapse
|
8
|
Shang L, Deng D, Krom BP, Gibbs S. Oral host-microbe interactions investigated in 3D organotypic models. Crit Rev Microbiol 2024; 50:397-416. [PMID: 37166371 DOI: 10.1080/1040841x.2023.2211665] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2023] [Revised: 04/30/2023] [Accepted: 05/02/2023] [Indexed: 05/12/2023]
Abstract
The oral cavity is inhabited by abundant microbes which continuously interact with the host and influence the host's health. Such host-microbe interactions (HMI) are dynamic and complex processes involving e.g. oral tissues, microbial communities and saliva. Due to difficulties in mimicking the in vivo complexity, it is still unclear how exactly HMI influence the transition between healthy status and disease conditions in the oral cavity. As an advanced approach, three-dimensional (3D) organotypic oral tissues (epithelium and mucosa/gingiva) are being increasingly used to study underlying mechanisms. These in vitro models were designed with different complexity depending on the research questions to be answered. In this review, we summarised the existing 3D oral HMI models, comparing designs and readouts, discussing applications as well as future perspectives.
Collapse
Affiliation(s)
- Lin Shang
- Department of Preventive Dentistry, Academic Centre for Dentistry Amsterdam (ACTA), University of Amsterdam and Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
| | - Dongmei Deng
- Department of Preventive Dentistry, Academic Centre for Dentistry Amsterdam (ACTA), University of Amsterdam and Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
| | - Bastiaan P Krom
- Department of Preventive Dentistry, Academic Centre for Dentistry Amsterdam (ACTA), University of Amsterdam and Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
| | - Susan Gibbs
- Department of Oral Cell Biology, Academic Centre for Dentistry Amsterdam (ACTA), University of Amsterdam and Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
- Department of Molecular Cell Biology and Immunology, Amsterdam Infection and Immunity Institute, Amsterdam UMC location Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
| |
Collapse
|
9
|
Muttaleb Asfoor H, Saied Hamied A. Immune response to colonization of Candida albicans in mice treated with Cefoperazone. Cytokine 2024; 179:156611. [PMID: 38640559 DOI: 10.1016/j.cyto.2024.156611] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2024] [Revised: 04/04/2024] [Accepted: 04/08/2024] [Indexed: 04/21/2024]
Abstract
Candida species are a normal human flora in humans' digestive and reproductive systems, oral cavity, skin, and mucosal surfaces. This study aimed to detect the immunological role of Candida infection by using some immunological markers. The results of levels in serum showed high concentrations of IgA (56.20 ± 12 pg/ml,29.55 ± 4.5 pg/ml respectively) and IgG (12.05 ± 3.218 pg/ml, 3.836 ± 1.23 pg/ml respectively) in mice infected with C. albicans and mice treated with Cefoperazone and infected with Candida with significant differences (P value < 0.05). The results showed high serum levels of IL-17(191.5 ± 42.81 pg/ml) and TLR2(7.651 ± 1.5 pg/ml) in group mice infected with C. albicans compared with negative control and group mice treated with Cefoperazone. Also, high levels of IL-17 (91.33 ± 4.816 pg/ml) and TLR2 (2.630 ± 0.5 pg/ml) in group mice treated with Cefoperazone and infected with Candida compared with negative control and group mice treated with Cefoperazone (P value < 0.05). The results of antibodies and immunological markers in the intestine showed high levels of IgA and IgG in mice infected with C.albicans (55.7 ± 4.9 pg/ml, 18.19 ± 0.63 pg/ml respectively).Also,IgA and IgG in mice treated with Cefoperazone and infected with Candida were high level (43.04 ± 2.1 pg/ml, 2.927 ± 0.2 pg/ml respectively) in mice infected with C. albicans with significant differences (P value < 0.05). The results levels of IL-17 and TLR2 were increased in mice infected with C. albicans (191.5 ± 42.81 pg/ml, 7.651 ± 1.5 pg/ml respectively) and mice treated with Cefoperazone and infected with Candida (91.33 ± 4.816 pg/ml,2.630 ± 0.5 pg/ml respectively) with significant differences (P < 0.05). In conclusion, this study demonstrated that cefoperazone treatment and infection by Candida albicans changed the microbiome components in the gut and finally can change host immune responses. It was observed that elevated levels of the antibodies production (IgA and IgG) and immunological markers (IL-17, and TLR2) in serum and the gut.
Collapse
Affiliation(s)
- Hussein Muttaleb Asfoor
- Department of Biology, College of Education for pure science Ibn-Al Haitham, University of Baghdad, Baghdad, Iraq
| | - Atyaf Saied Hamied
- Department of Biology, College of Education for pure science Ibn-Al Haitham, University of Baghdad, Baghdad, Iraq.
| |
Collapse
|
10
|
Chong AC, Navarro-Triviño FJ, Su M, Park CO. Fungal Head and Neck Dermatitis: Current Understanding and Management. Clin Rev Allergy Immunol 2024; 66:363-375. [PMID: 39031274 PMCID: PMC11422441 DOI: 10.1007/s12016-024-09000-7] [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] [Accepted: 07/11/2024] [Indexed: 07/22/2024]
Abstract
Head and neck dermatitis (HND) is a form of atopic dermatitis (AD) that affects the seborrheic areas of the body and causes greater quality of life detriments than other types of AD. HND can be challenging to treat since first-line topical therapies may be ineffective or intolerable for long-term use on areas affected by HND while dupilumab may cause dupilumab-associated HND (DAHND). Current evidence implicates fungi, particularly Malassezia spp., in the pathogenesis of HND. Penetration of fungal antigens through the defective AD skin barrier activates the innate and adaptive immune systems to cause cutaneous inflammation via the T helper (Th)17 and/or Th2 axes. Malassezia sensitization may distinguish HND from other forms of AD. Multiple double-blind, placebo-controlled trials have shown antifungals to benefit HND, yet the persistence of symptom relief with sustained use remains unclear. Oral antifungals appear more effective than topical antifungals but may be harmful with long-term use. DAHND may also be fungal-mediated given improvement with antifungals and evidence of an overactive immune response against Malassezia in these patients. Janus kinase inhibitors are effective for HND, including DAHND, but may cause significant side effects when administered systemically. OX40/OX40L inhibitors and tralokinumab may be promising options for HND on the horizon. Demographic and environmental factors influence the host mycobiome and should be considered in future precision-medicine approaches as microbiome composition and diversity are linked to severity of HND.
Collapse
Affiliation(s)
- Albert C Chong
- Department of Internal Medicine, Mayo Clinic Arizona, 13400 E Shea Blvd., Scottsdale, AZ, 85259, USA.
- Keck School of Medicine, University of Southern California, Los Angeles, CA, USA.
| | - Francisco José Navarro-Triviño
- Department of Contact Eczema and Immunoallergic Diseases, Dermatology Service, Hospital Universitario San Cecilio, Granada, Spain
| | - Malcolm Su
- Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Chang Ook Park
- Department of Dermatology & Cutaneous Biology Research Institute, Yonsei University College of Medicine, Seoul, South Korea
| |
Collapse
|
11
|
Wu D, Hailer AA, Wang S, Yuan M, Chan J, El Kurdi A, Rahim M, Kondo A, Han D, Ali H, D'Angio B, Mayer A, Klufas D, Kim E, Shain AH, Choi J, Bhutani T, Simpson G, Grekin RC, Ricardo-Gonzalez R, Purdom E, North JP, Cheng JB, Cho RJ. A single-cell atlas of IL-23 inhibition in cutaneous psoriasis distinguishes clinical response. Sci Immunol 2024; 9:eadi2848. [PMID: 38277466 DOI: 10.1126/sciimmunol.adi2848] [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: 04/17/2023] [Accepted: 11/22/2023] [Indexed: 01/28/2024]
Abstract
Psoriasis vulgaris and other chronic inflammatory diseases improve markedly with therapeutic blockade of interleukin-23 (IL-23) signaling, but the genetic mechanisms underlying clinical responses remain poorly understood. Using single-cell transcriptomics, we profiled immune cells isolated from lesional psoriatic skin before and during IL-23 blockade. In clinically responsive patients, a psoriatic transcriptional signature in skin-resident memory T cells was strongly attenuated. In contrast, poorly responsive patients were distinguished by persistent activation of IL-17-producing T (T17) cells, a mechanism distinct from alternative cytokine signaling or resistance isolated to epidermal keratinocytes. Even in IL-23 blockade-responsive patients, we detected a recurring set of recalcitrant, disease-specific transcriptional abnormalities. This irreversible immunological state may necessitate ongoing IL-23 inhibition. Spatial transcriptomic analyses also suggested that successful IL-23 blockade requires dampening of >90% of IL-17-induced response in lymphocyte-adjacent keratinocytes, an unexpectedly high threshold. Collectively, our data establish a patient-level paradigm for dissecting responses to immunomodulatory treatments.
Collapse
Affiliation(s)
- David Wu
- Department of Dermatology, University of California, San Francisco, San Francisco, CA 94107, USA
| | - Ashley A Hailer
- Department of Dermatology, University of California, San Francisco, San Francisco, CA 94107, USA
- Dermatology Service, San Francisco Veterans Administration Health Care System, San Francisco, CA 94121, USA
| | - Sijia Wang
- Department of Dermatology, University of California, San Francisco, San Francisco, CA 94107, USA
- Dermatology Service, San Francisco Veterans Administration Health Care System, San Francisco, CA 94121, USA
- Department of Dermatology, Second Affiliated Hospital, School of Medicine, Xi'an Jiaotong University, Xi'an 710004, China
| | - Michelle Yuan
- Department of Dermatology, University of California, San Francisco, San Francisco, CA 94107, USA
| | - Jamie Chan
- Dermatopathology Service, University of California, San Francisco, San Francisco, CA 94107, USA
| | - Abdullah El Kurdi
- Department of Biochemistry and Molecular Genetics, American University of Beirut, Beirut, Lebanon
| | - Maha Rahim
- Enable Medicine, Menlo Park, CA 94025, USA
| | | | - David Han
- Enable Medicine, Menlo Park, CA 94025, USA
| | - Hira Ali
- Enable Medicine, Menlo Park, CA 94025, USA
| | | | | | - Daniel Klufas
- Department of Dermatology, University of California, San Francisco, San Francisco, CA 94107, USA
| | - Esther Kim
- Department of Plastic Surgery, University of California, San Francisco, San Francisco, CA 94143, USA
| | - A Hunter Shain
- Department of Dermatology, University of California, San Francisco, San Francisco, CA 94107, USA
| | - Jaehyuk Choi
- Departments of Dermatology and Biochemistry and Molecular Genetics, Feinberg School of Medicine, Chicago, IL 60611, USA
| | - Tina Bhutani
- Department of Dermatology, University of California, San Francisco, San Francisco, CA 94107, USA
| | - Gregory Simpson
- Department of Dermatology, University of California, Fresno, CA 93701,USA
| | - Roy C Grekin
- Department of Dermatology, University of California, San Francisco, San Francisco, CA 94107, USA
| | - Roberto Ricardo-Gonzalez
- Department of Dermatology, University of California, San Francisco, San Francisco, CA 94107, USA
| | - Elizabeth Purdom
- Department of Statistics, University of California, Berkeley, Berkeley, CA 94720, USA
| | - Jeffrey P North
- Dermatopathology Service, University of California, San Francisco, San Francisco, CA 94107, USA
| | - Jeffrey B Cheng
- Department of Dermatology, University of California, San Francisco, San Francisco, CA 94107, USA
- Dermatology Service, San Francisco Veterans Administration Health Care System, San Francisco, CA 94121, USA
| | - Raymond J Cho
- Department of Dermatology, University of California, San Francisco, San Francisco, CA 94107, USA
| |
Collapse
|
12
|
Ruchti F, Tuor M, Mathew L, McCarthy NE, LeibundGut-Landmann S. γδ T cells respond directly and selectively to the skin commensal yeast Malassezia for IL-17-dependent fungal control. PLoS Pathog 2024; 20:e1011668. [PMID: 38215167 PMCID: PMC10810444 DOI: 10.1371/journal.ppat.1011668] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2023] [Revised: 01/25/2024] [Accepted: 12/16/2023] [Indexed: 01/14/2024] Open
Abstract
Stable microbial colonization of the skin depends on tight control by the host immune system. The lipid-dependent yeast Malassezia typically colonizes skin as a harmless commensal and is subject to host type 17 immunosurveillance, but this fungus has also been associated with diverse skin pathologies in both humans and animals. Using a murine model of Malassezia exposure, we show that Vγ4+ dermal γδ T cells expand rapidly and are the major source of IL-17A mediating fungal control in colonized skin. A pool of memory-like Malassezia-responsive Vγ4+ T cells persisted in the skin, were enriched in draining lymph nodes even after fungal clearance, and were protective upon fungal re-exposure up to several weeks later. Induction of γδT17 immunity depended on IL-23 and IL-1 family cytokine signalling, whereas Toll-like and C-type lectin receptors were dispensable. Furthermore, Vγ4+ T cells from Malassezia-exposed hosts were able to respond directly and selectively to Malassezia-derived ligands, independently of antigen-presenting host cells. The fungal moieties detected were shared across diverse species of the Malassezia genus, but not conserved in other Basidiomycota or Ascomycota. These data provide novel mechanistic insight into the induction and maintenance of type 17 immunosurveillance of skin commensal colonization that has significant implications for cutaneous health.
Collapse
Affiliation(s)
- Fiorella Ruchti
- Section of Immunology, Vetsuisse Faculty, University of Zürich, Zürich, Switzerland
- Institute of Experimental Immunology, University of Zürich, Zürich, Switzerland
| | - Meret Tuor
- Section of Immunology, Vetsuisse Faculty, University of Zürich, Zürich, Switzerland
- Institute of Experimental Immunology, University of Zürich, Zürich, Switzerland
| | - Liya Mathew
- Centre for Immunobiology, Bart’s and The London School of Medicine and Dentistry, The Blizard Institute, Queen Mary University of London, London, United Kingdom
| | - Neil E McCarthy
- Centre for Immunobiology, Bart’s and The London School of Medicine and Dentistry, The Blizard Institute, Queen Mary University of London, London, United Kingdom
| | - Salomé LeibundGut-Landmann
- Section of Immunology, Vetsuisse Faculty, University of Zürich, Zürich, Switzerland
- Institute of Experimental Immunology, University of Zürich, Zürich, Switzerland
| |
Collapse
|
13
|
Yu S, Wang K, Cao C, Zhang B, Chen Y, Wu C, Li C, Tang J, Luo W. Tissue-resident memory T cells exhibit phenotypically and functionally heterogeneous in human physiological and pathological nasal mucosa. Clin Immunol 2024; 258:109860. [PMID: 38065369 DOI: 10.1016/j.clim.2023.109860] [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: 08/22/2023] [Revised: 11/27/2023] [Accepted: 11/28/2023] [Indexed: 01/02/2024]
Abstract
Pathogens commonly enter mucosal barrier tissues and tissue-resident memory T cells (TRM) are essential for preventing mucosal lesions. However, the immunological properties of TRM cells in nasal mucosa are poorly known. In comparison with control tissues, decreasing CD103+ TRM cells were observed in Chronic rhinosinusitis with nasal polyps (CRSwNPs) and sinonasal inverted papilloma (SNIP), which presented high capability to produce effector cytokines. In CRSwNPs, we found that CD103+ TRM cells with higher cytokine and Granzyme B coexpressed high PD-1, CD103- TRM cells expressed higher IL-10. Homogenates isolated from CRSwNPs induced CD103 expression on peripheral T cells which could be inhibited by blocking TGF-β. The frequencies of CD103+ TRM cells in CRSwNPs were extremely negatively correlated with neutrophil infiltration. CD103+ TRM cells from Staphylococcus aureus positive CRSwNPs had a stronger response to SEB. Taken together, two phenotypically and functionally distinct subsets of TRM cells exist in nasal tissues and play critical roles in the progress of CRSwNPs and SNIPs.
Collapse
Affiliation(s)
- Sifei Yu
- Institute of translational medicine, The First People's Hospital of Foshan, 81 Lingnan Road, Foshan 528000, PR China
| | - Kai Wang
- Department of Otolaryngology, The First People's Hospital of Foshan, 81 Lingnan Road, Foshan 528000, PR China
| | - Chen Cao
- Department of Otolaryngology, Guangzhou Key Laboratory of Otorhinolaryngology, The First Affiliated Hospital, Sun Yat-sen University, 58 Zhongshan 2nd Road, Guangzhou 510080, PR China
| | - Beiying Zhang
- Institute of translational medicine, The First People's Hospital of Foshan, 81 Lingnan Road, Foshan 528000, PR China
| | - Youmou Chen
- Department of Otolaryngology, The First People's Hospital of Foshan, 81 Lingnan Road, Foshan 528000, PR China; The General Hospital of Western Theater Command, No. 270, Rongdu Avenue, Chengdu 610083, PR China
| | - Changyou Wu
- Clifford Hospital, Jinan University, No.3 Hongfu Road, Guangzhou 511495, PR China
| | - Chunwei Li
- Department of Otolaryngology, Guangzhou Key Laboratory of Otorhinolaryngology, The First Affiliated Hospital, Sun Yat-sen University, 58 Zhongshan 2nd Road, Guangzhou 510080, PR China
| | - Jun Tang
- Department of Otolaryngology, The First People's Hospital of Foshan, 81 Lingnan Road, Foshan 528000, PR China.
| | - Wei Luo
- Institute of translational medicine, The First People's Hospital of Foshan, 81 Lingnan Road, Foshan 528000, PR China.
| |
Collapse
|
14
|
Wang Y, Xue N, Wang Z, Zeng X, Ji N, Chen Q. Targeting Th17 cells: a promising strategy to treat oral mucosal inflammatory diseases. Front Immunol 2023; 14:1236856. [PMID: 37564654 PMCID: PMC10410157 DOI: 10.3389/fimmu.2023.1236856] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2023] [Accepted: 07/06/2023] [Indexed: 08/12/2023] Open
Abstract
With the improved quality of life, oral health is under increased pressure. Numerous common oral mucosal diseases, such as oral lichen planus(OLP) and gingivitis, are related to the destruction of the oral immune barrier. The cytokines secreted by T-helper 17 (Th17) cells are essential for maintaining oral immune homeostasis and play essential roles in immune surveillance. When antigens stimulate the epithelium, Th17 cells expand, differentiate, and generate inflammatory factors to recruit other lymphocytes, such as neutrophils, to clear the infection, which helps to maintain the integrity of the epithelial barrier. In contrast, excessive Th17/IL-17 axis reactions may cause autoimmune damage. Therefore, an in-depth understanding of the role of Th17 cells in oral mucosa may provide prospects for treating oral mucosal diseases. We reviewed the role of Th17 cells in various oral and skin mucosal systemic diseases with oral characteristics, and based on the findings of these reports, we emphasize that Th17 cellular response may be a critical factor in inflammatory diseases of the oral mucosa. In addition, we should pay attention to the role and relationship of "pathogenic Th17" and "non-pathogenic Th17" in oral mucosal diseases. We hope to provide a reference for Th17 cells as a potential therapeutic target for treating oral mucosal inflammatory disorders in the future.
Collapse
Affiliation(s)
| | | | | | | | - Ning Ji
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, Research Unit of Oral Carcinogenesis and Management, Chinese Academy of Medical Sciences, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, China
| | | |
Collapse
|
15
|
Abstract
Specialized subpopulations of CD4+ T cells survey major histocompatibility complex class II-peptide complexes to control phagosomal infections, help B cells, regulate tissue homeostasis and repair or perform immune regulation. Memory CD4+ T cells are positioned throughout the body and not only protect the tissues from reinfection and cancer, but also participate in allergy, autoimmunity, graft rejection and chronic inflammation. Here we provide updates on our understanding of the longevity, functional heterogeneity, differentiation, plasticity, migration and human immunodeficiency virus reservoirs as well as key technological advances that are facilitating the characterization of memory CD4+ T cell biology.
Collapse
Affiliation(s)
- Marco Künzli
- Center for Immunology, Department of Microbiology and Immunology, University of Minnesota, Minneapolis, MN, USA
| | - David Masopust
- Center for Immunology, Department of Microbiology and Immunology, University of Minnesota, Minneapolis, MN, USA.
| |
Collapse
|
16
|
Ruchti F, LeibundGut-Landmann S. New insights into immunity to skin fungi shape our understanding of health and disease. Parasite Immunol 2023; 45:e12948. [PMID: 36047038 PMCID: PMC10078452 DOI: 10.1111/pim.12948] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2022] [Revised: 08/18/2022] [Accepted: 08/25/2022] [Indexed: 01/31/2023]
Abstract
Fungi represent an integral part of the skin microbiota. Their complex interaction network with the host shapes protective immunity during homeostasis. If host defences are breached, skin-resident fungi including Malassezia and Candida, and environmental fungi such as dermatophytes can cause cutaneous infections. In addition, fungi are associated with diverse non-infectious skin disorders. Despite their multiple roles in health and disease, fungi remain elusive and understudied, and the mechanisms underlying the emergence of pathological conditions linked to fungi are largely unclear. The identification of IL-17 as an important antifungal effector mechanism represents a milestone for understanding homeostatic antifungal immunity. At the same time, host-adverse, disease-promoting roles of IL-17 have been delineated, as in psoriasis. Fungal dysbiosis represents another feature of many pathological skin conditions with an unknown causal link of intra- and interkingdom interactions to disease pathogenesis. The emergence of new fungal pathogens such as Candida auris highlights the need for more research into fungal immunology to understand how antifungal responses shape health and diseases. Recent technological advances for genetically manipulating fungi to target immunomodulatory fungal determinants, multi-omics approaches for studying immune cells in the human skin, and novel experimental models open up a promising future for skin fungal immunity.
Collapse
Affiliation(s)
- Fiorella Ruchti
- Section of Immunology, Vetsuisse Faculty, University of Zürich, Zürich, Switzerland.,Institute of Experimental Immunology, University of Zürich, Zürich, Switzerland
| | - Salomé LeibundGut-Landmann
- Section of Immunology, Vetsuisse Faculty, University of Zürich, Zürich, Switzerland.,Institute of Experimental Immunology, University of Zürich, Zürich, Switzerland
| |
Collapse
|
17
|
Singh AK, Majumder S, Wang X, Song R, Sun W. Lung Resident Memory T Cells Activated by Oral Vaccination Afford Comprehensive Protection against Pneumonic Yersinia pestis Infection. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2023; 210:259-270. [PMID: 36480265 PMCID: PMC9851976 DOI: 10.4049/jimmunol.2200487] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/07/2022] [Accepted: 11/17/2022] [Indexed: 01/03/2023]
Abstract
A growing body of evidence has shown that resident memory T (TRM) cells formed in tissue after mucosal infection or vaccination are crucial for counteracting reinfection by pathogens. However, whether lung TRM cells activated by oral immunization with Yptb1(pYA5199) play a protective role against pneumonic plague remains unclear. In this study, we demonstrated that lung CD4+ and CD8+ TRM cells significantly accumulated in the lungs of orally Yptb1(pYA5199)-vaccinated mice and dramatically expanded with elevated IL-17A, IFN-γ, and/or TNF-α production after pulmonary Yersinia pestis infection and afforded significant protection. Short-term or long-term treatment of immunized mice with FTY720 did not affect lung TRM cell formation and expansion or protection against pneumonic plague. Moreover, the intratracheal transfer of both lung CD4+ and CD8+ TRM cells conferred comprehensive protection against pneumonic plague in naive recipient mice. Lung TRM cell-mediated protection was dramatically abolished by the neutralization of both IFN-γ and IL-17A. Our findings reveal that lung TRM cells can be activated via oral Yptb1(pYA5199) vaccination, and that IL-17A and IFN-γ production play an essential role in adaptive immunity against pulmonary Y. pestis infection. This study highlights an important new target for developing an effective pneumonic plague vaccine.
Collapse
Affiliation(s)
- Amit K. Singh
- Department of Immunology and Microbial Disease, Albany Medical College, Albany, NY, 12208, USA
| | - Saugata Majumder
- Department of Immunology and Microbial Disease, Albany Medical College, Albany, NY, 12208, USA
| | - Xiuran Wang
- Department of Immunology and Microbial Disease, Albany Medical College, Albany, NY, 12208, USA
| | - Renjie Song
- Immunology Core at Wadsworth Center, New York State Department of Health, Albany, NY, 12208, USA
| | - Wei Sun
- Department of Immunology and Microbial Disease, Albany Medical College, Albany, NY, 12208, USA
| |
Collapse
|
18
|
Jacobsen ID. The Role of Host and Fungal Factors in the Commensal-to-Pathogen Transition of Candida albicans. CURRENT CLINICAL MICROBIOLOGY REPORTS 2023; 10:55-65. [PMID: 37151578 PMCID: PMC10154278 DOI: 10.1007/s40588-023-00190-w] [Citation(s) in RCA: 23] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/16/2023] [Indexed: 05/09/2023]
Abstract
Abstract Purpose of Review The fungus Candida albicans has evolved to live in close association with warm-blooded hosts and is found frequently on mucosal surfaces of healthy humans. As an opportunistic pathogen, C. albicans can also cause mucosal and disseminated infections (candidiasis). This review describes the features that differentiate the fungus in the commensal versus pathogenic state and the main factors underlying C. albicans commensal-to-pathogen transition. Recent Findings Adhesion, invasion, and tissue damage are critical steps in the infection process. Especially invasion and damage require transcriptional and morphological changes that differentiate C. albicans in the pathogenic from the commensal state. While the commensal-to-pathogen transition has some conserved causes and features in the oral cavity, the female urogenital tract, and the gut, site-specific differences have been identified in recent years. Summary This review highlights how specific factors in the different mucosal niches affect development of candidiasis. Recent evidence suggests that colonization of the gut is not only a risk factor for systemic candidiasis but might also provide beneficial effects to the host.
Collapse
Affiliation(s)
- Ilse D. Jacobsen
- Research Group Microbial Immunology, Leibniz Institute for Natural Product Research and Infection Biology, Jena, Germany
- Institute of Microbiology, Friedrich Schiller University, Jena, Germany
| |
Collapse
|
19
|
Das D, HogenEsch H, Thangamani S. Intestinal colonization with Candida auris and mucosal immune response in mice treated with cefoperazone oral antibiotic. Front Immunol 2023; 14:1123200. [PMID: 37114044 PMCID: PMC10126271 DOI: 10.3389/fimmu.2023.1123200] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2022] [Accepted: 03/29/2023] [Indexed: 04/29/2023] Open
Abstract
Candida auris, an emerging multi-drug resistant fungal pathogen, causes invasive infections in humans. The factors regulating the colonization of C. auris in host niches are not well understood. In this study, we examined the effect of antibiotic-induced gut dysbiosis on C. auris intestinal colonization, dissemination, microbiome composition and the mucosal immune response. Our results indicate that mice treated with cefoperazone alone had a significant increase in C. auris intestinal colonization compared to untreated control groups. A significant increase in the dissemination of C. auris from the intestine to internal organs was observed in antibiotic-treated immunosuppressed mice. Intestinal colonization of C. auris alters the microbiome composition of antibiotic-treated mice. Relative abundance of firmicutes members mainly Clostridiales and Paenibacillus were considerably increased in the cefoperazone-treated mice infected with C. auris compared to cefoperazone-treated uninfected mice. Next, we examined the mucosal immune response of C. auris infected mice and compared the results with Candida albicans infection. The number of CD11b+ CX3CR1+ macrophages was significantly decreased in the intestine of C. auris infected mice when compared to C. albicans infection. On the other hand, both C. auris and C. albicans infected mice had a comparable increase of the number of Th17 and Th22 cells in the intestine. A significant increase in Candida-specific IgA was observed in the serum of C. auris but not in the C. albicans infected mice. Taken together, treatment with broad-spectrum antibiotic increased the colonization and dissemination of C. auris from the intestine. Furthermore, findings from this study for the first time revealed the microbiome composition, innate and adaptive cellular immune response to intestinal infection with C. auris.
Collapse
Affiliation(s)
- Diprasom Das
- Department of Comparative Pathobiology, College of Veterinary Medicine, Purdue University, West Lafayette, IN, United States
| | - Harm HogenEsch
- Department of Comparative Pathobiology, College of Veterinary Medicine, Purdue University, West Lafayette, IN, United States
- Purdue Institute for Immunology, Inflammation and Infectious Diseases (PI4D), West Lafayette, IN, United States
| | - Shankar Thangamani
- Department of Comparative Pathobiology, College of Veterinary Medicine, Purdue University, West Lafayette, IN, United States
- Purdue Institute for Immunology, Inflammation and Infectious Diseases (PI4D), West Lafayette, IN, United States
- *Correspondence: Shankar Thangamani,
| |
Collapse
|
20
|
Lopes JP, Lionakis MS. Pathogenesis and virulence of Candida albicans. Virulence 2022; 13:89-121. [PMID: 34964702 PMCID: PMC9728475 DOI: 10.1080/21505594.2021.2019950] [Citation(s) in RCA: 191] [Impact Index Per Article: 63.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2021] [Revised: 12/08/2021] [Accepted: 12/14/2021] [Indexed: 12/18/2022] Open
Abstract
Candida albicans is a commensal yeast fungus of the human oral, gastrointestinal, and genital mucosal surfaces, and skin. Antibiotic-induced dysbiosis, iatrogenic immunosuppression, and/or medical interventions that impair the integrity of the mucocutaneous barrier and/or perturb protective host defense mechanisms enable C. albicans to become an opportunistic pathogen and cause debilitating mucocutaneous disease and/or life-threatening systemic infections. In this review, we synthesize our current knowledge of the tissue-specific determinants of C. albicans pathogenicity and host immune defense mechanisms.
Collapse
Affiliation(s)
- José Pedro Lopes
- From the Fungal Pathogenesis Section, Laboratory of Clinical Immunology and Microbiology (LCIM), National Institute of Allergy and Infectious Diseases (NIAID), Bethesda, MD, USA
| | - Michail S. Lionakis
- From the Fungal Pathogenesis Section, Laboratory of Clinical Immunology and Microbiology (LCIM), National Institute of Allergy and Infectious Diseases (NIAID), Bethesda, MD, USA
| |
Collapse
|
21
|
Liu Y, Qv W, Ma Y, Zhang Y, Ding C, Chu M, Chen F. The interplay between oral microbes and immune responses. Front Microbiol 2022. [DOI: 10.3389/fmicb.2022.1009018] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Oral microbes play a critical role in maintaining oral homeostasis. Microbial dysbiosis promotes disease pathogenesis through several mechanisms. Recent studies have revealed that microbial imbalance and sustained inflammation are involved in disease progression. The adverse interaction triggered by a host immune response to microorganisms can lead to oral and systemic diseases. Here, we reviewed how oral microbes communicate with hosts during the development of local and distant inflammation. Elucidation of these processes may reveal future directions in this field and the potential targets of novel biological therapies for oral and systemic diseases.
Collapse
|
22
|
Vitte J, Michel M, Malinovschi A, Caminati M, Odebode A, Annesi-Maesano I, Caimmi DP, Cassagne C, Demoly P, Heffler E, Menu E, Nwaru BI, Sereme Y, Ranque S, Raulf M, Feleszko W, Janson C, Galán C. Fungal exposome, human health, and unmet needs: A 2022 update with special focus on allergy. Allergy 2022; 77:3199-3216. [PMID: 35976185 DOI: 10.1111/all.15483] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2022] [Revised: 08/01/2022] [Accepted: 08/13/2022] [Indexed: 01/28/2023]
Abstract
Humans inhale, ingest, and touch thousands of fungi each day. The ubiquity and diversity of the fungal kingdom, reflected by its complex taxonomy, are in sharp contrast with our scarce knowledge about its distribution, pathogenic effects, and effective interventions at the environmental and individual levels. Here, we present an overview of salient features of fungi as permanent players of the human exposome and key determinants of human health, through the lens of fungal allergy and other fungal hypersensitivity reactions. Improved understanding of the fungal exposome sheds new light on the epidemiology of fungal-related hypersensitivity diseases, their immunological substratum, the currently available methods, and biomarkers for environmental and medical fungi. Unmet needs are described and potential approaches are highlighted as perspectives.
Collapse
Affiliation(s)
- Joana Vitte
- IDESP, University of Montpellier and INSERM, Montpellier, France.,MEPHI, IHU Méditerranée Infection, IRD, APHM, Aix-Marseille Univ, Marseille, France
| | - Moïse Michel
- IDESP, University of Montpellier and INSERM, Montpellier, France.,MEPHI, IHU Méditerranée Infection, IRD, APHM, Aix-Marseille Univ, Marseille, France.,Immunology Laboratory, University Hospital Nîmes, Nîmes, France
| | - Andrei Malinovschi
- Department of Medical Sciences Clinical Physiology, Uppsala University, Uppsala, Sweden
| | - Marco Caminati
- Asthma, Allergy and Clinical Immunology Section, Department of Medicine, University of Verona, Verona, Italy
| | - Adeyinka Odebode
- Department of Basic Science, Kampala International University, Kampala, Uganda
| | | | - Davide Paolo Caimmi
- IDESP, University of Montpellier and INSERM, Montpellier, France.,Departement of Pneumology, University Hospital of Montpellier, Montpellier, France
| | - Carole Cassagne
- VITROME, IHU Méditerranée Infection, IRD, APHM, Aix-Marseille Univ, Marseille, France
| | - Pascal Demoly
- IDESP, University of Montpellier and INSERM, Montpellier, France.,Departement of Pneumology, University Hospital of Montpellier, Montpellier, France
| | - Enrico Heffler
- Personalized Medicine, Asthma and Allergy Humanitas Clinical and Research Center IRCCS Rozzano, Rozzano, Italy.,Department of Biomedical Sciences, Humanitas University, Pieve Emanuele, Italy
| | - Estelle Menu
- VITROME, IHU Méditerranée Infection, IRD, APHM, Aix-Marseille Univ, Marseille, France
| | - Bright I Nwaru
- Krefting Research Centre, Institute of Medicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden.,Wallenberg Centre for Molecular and Translational Medicine, University of Gothenburg, Gothenburg, Sweden
| | - Youssouf Sereme
- MEPHI, IHU Méditerranée Infection, IRD, APHM, Aix-Marseille Univ, Marseille, France.,Department of Immunology, Infectiology and Hematology, Institut Necker-Enfants Malades (INEM), INSERM U1151, CNRS UMR 8253, Université Paris Descartes, Paris, France
| | - Stéphane Ranque
- VITROME, IHU Méditerranée Infection, IRD, APHM, Aix-Marseille Univ, Marseille, France
| | - Monika Raulf
- Department of Allergology and Immunology, Institute for Prevention and Occupational Medicine of the German Social Accident Insurance, Institute of the Ruhr-Universität Bochum (IPA), Bochum, Germany
| | - Wojciech Feleszko
- Department of Pediatric Pulmonology and Allergy, Medical University of Warsaw, Warsaw, Poland
| | - Christer Janson
- Department of Medical Sciences Respiratory, Allergy and Sleep Research, Uppsala University, Uppsala, Sweden
| | - Carmen Galán
- International Campus of Excellence on Agrifood (ceiA3), University of Cordoba, Córdoba, Spain.,Andalusian Inter-University Institute for Earth System Research (IISTA), University of Cordoba, Córdoba, Spain
| | | |
Collapse
|
23
|
Michaudel C, Danne C, Agus A, Magniez A, Aucouturier A, Spatz M, Lefevre A, Kirchgesner J, Rolhion N, Wang Y, Lavelle A, Galbert C, Da Costa G, Poirier M, Lapière A, Planchais J, Nádvorník P, Illes P, Oeuvray C, Creusot L, Michel ML, Benech N, Bourrier A, Nion-Larmurier I, Landman C, Richard ML, Emond P, Seksik P, Beaugerie L, Arguello RR, Moulin D, Mani S, Dvorák Z, Bermúdez-Humarán LG, Langella P, Sokol H. Rewiring the altered tryptophan metabolism as a novel therapeutic strategy in inflammatory bowel diseases. Gut 2022:gutjnl-2022-327337. [PMID: 36270778 DOI: 10.1136/gutjnl-2022-327337] [Citation(s) in RCA: 100] [Impact Index Per Article: 33.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/11/2022] [Accepted: 10/06/2022] [Indexed: 11/04/2022]
Abstract
OBJECTIVE The extent to which tryptophan (Trp) metabolism alterations explain or influence the outcome of inflammatory bowel diseases (IBDs) is still unclear. However, several Trp metabolism end-products are essential to intestinal homeostasis. Here, we investigated the role of metabolites from the kynurenine pathway. DESIGN Targeted quantitative metabolomics was performed in two large human IBD cohorts (1069 patients with IBD). Dextran sodium sulphate-induced colitis experiments in mice were used to evaluate effects of identified metabolites. In vitro, ex vivo and in vivo experiments were used to decipher mechanisms involved. Effects on energy metabolism were evaluated by different methods including Single Cell mEtabolism by profiling Translation inHibition. RESULTS In mice and humans, intestinal inflammation severity negatively correlates with the amount of xanthurenic (XANA) and kynurenic (KYNA) acids. Supplementation with XANA or KYNA decreases colitis severity through effects on intestinal epithelial cells and T cells, involving Aryl hydrocarbon Receptor (AhR) activation and the rewiring of cellular energy metabolism. Furthermore, direct modulation of the endogenous tryptophan metabolism, using the recombinant enzyme aminoadipate aminotransferase (AADAT), responsible for the generation of XANA and KYNA, was protective in rodent colitis models. CONCLUSION Our study identified a new mechanism linking Trp metabolism to intestinal inflammation and IBD. Bringing back XANA and KYNA has protective effects involving AhR and the rewiring of the energy metabolism in intestinal epithelial cells and CD4+ T cells. This study paves the way for new therapeutic strategies aiming at pharmacologically correcting its alterations in IBD by manipulating the endogenous metabolic pathway with AADAT.
Collapse
Affiliation(s)
- Chloé Michaudel
- Université Paris-Saclay, INRAe, AgroParisTech, Micalis institute, Jouy-en-Josas, France.,Paris Center for Microbiome Medicine (PaCeMM) FHU, Paris, France
| | - Camille Danne
- Université Paris-Saclay, INRAe, AgroParisTech, Micalis institute, Jouy-en-Josas, France.,Paris Center for Microbiome Medicine (PaCeMM) FHU, Paris, France.,Sorbonne Université, INSERM UMRS-938, Centre de Recherche Saint-Antoine, CRSA, AP-HP, Paris, France
| | - Allison Agus
- Université Paris-Saclay, INRAe, AgroParisTech, Micalis institute, Jouy-en-Josas, France.,Paris Center for Microbiome Medicine (PaCeMM) FHU, Paris, France
| | - Aurélie Magniez
- Université Paris-Saclay, INRAe, AgroParisTech, Micalis institute, Jouy-en-Josas, France.,Paris Center for Microbiome Medicine (PaCeMM) FHU, Paris, France
| | - Anne Aucouturier
- Université Paris-Saclay, INRAe, AgroParisTech, Micalis institute, Jouy-en-Josas, France.,Paris Center for Microbiome Medicine (PaCeMM) FHU, Paris, France
| | - Madeleine Spatz
- Université Paris-Saclay, INRAe, AgroParisTech, Micalis institute, Jouy-en-Josas, France.,Paris Center for Microbiome Medicine (PaCeMM) FHU, Paris, France
| | - Antoine Lefevre
- UMR 1253, iBrain, University of Tours, Inserm, 37044 Tours, France
| | - Julien Kirchgesner
- Paris Center for Microbiome Medicine (PaCeMM) FHU, Paris, France.,Gastroenterology department, Saint Antoine Hospital, APHP, Paris, France
| | - Nathalie Rolhion
- Paris Center for Microbiome Medicine (PaCeMM) FHU, Paris, France.,Sorbonne Université, INSERM UMRS-938, Centre de Recherche Saint-Antoine, CRSA, AP-HP, Paris, France
| | - Yazhou Wang
- Université Paris-Saclay, INRAe, AgroParisTech, Micalis institute, Jouy-en-Josas, France.,Paris Center for Microbiome Medicine (PaCeMM) FHU, Paris, France
| | - Aonghus Lavelle
- Paris Center for Microbiome Medicine (PaCeMM) FHU, Paris, France.,Sorbonne Université, INSERM UMRS-938, Centre de Recherche Saint-Antoine, CRSA, AP-HP, Paris, France
| | - Chloé Galbert
- Paris Center for Microbiome Medicine (PaCeMM) FHU, Paris, France.,Sorbonne Université, INSERM UMRS-938, Centre de Recherche Saint-Antoine, CRSA, AP-HP, Paris, France
| | - Gregory Da Costa
- Université Paris-Saclay, INRAe, AgroParisTech, Micalis institute, Jouy-en-Josas, France.,Paris Center for Microbiome Medicine (PaCeMM) FHU, Paris, France
| | - Maxime Poirier
- Université Paris-Saclay, INRAe, AgroParisTech, Micalis institute, Jouy-en-Josas, France.,Paris Center for Microbiome Medicine (PaCeMM) FHU, Paris, France
| | - Alexia Lapière
- Université Paris-Saclay, INRAe, AgroParisTech, Micalis institute, Jouy-en-Josas, France.,Paris Center for Microbiome Medicine (PaCeMM) FHU, Paris, France
| | - Julien Planchais
- Université Paris-Saclay, INRAe, AgroParisTech, Micalis institute, Jouy-en-Josas, France.,Paris Center for Microbiome Medicine (PaCeMM) FHU, Paris, France
| | - Petr Nádvorník
- Department of Cell Biology and Genetics, Faculty of Science, Palacky University, Olomouc, Czech Republic
| | - Peter Illes
- Department of Cell Biology and Genetics, Faculty of Science, Palacky University, Olomouc, Czech Republic
| | - Cyriane Oeuvray
- Paris Center for Microbiome Medicine (PaCeMM) FHU, Paris, France.,Sorbonne Université, INSERM UMRS-938, Centre de Recherche Saint-Antoine, CRSA, AP-HP, Paris, France
| | - Laura Creusot
- Paris Center for Microbiome Medicine (PaCeMM) FHU, Paris, France.,Sorbonne Université, INSERM UMRS-938, Centre de Recherche Saint-Antoine, CRSA, AP-HP, Paris, France
| | - Marie-Laure Michel
- Université Paris-Saclay, INRAe, AgroParisTech, Micalis institute, Jouy-en-Josas, France.,Paris Center for Microbiome Medicine (PaCeMM) FHU, Paris, France
| | - Nicolas Benech
- Paris Center for Microbiome Medicine (PaCeMM) FHU, Paris, France.,Sorbonne Université, INSERM UMRS-938, Centre de Recherche Saint-Antoine, CRSA, AP-HP, Paris, France.,Gastroenterology department, Saint Antoine Hospital, APHP, Paris, France
| | - Anne Bourrier
- Paris Center for Microbiome Medicine (PaCeMM) FHU, Paris, France.,Gastroenterology department, Saint Antoine Hospital, APHP, Paris, France
| | - Isabelle Nion-Larmurier
- Paris Center for Microbiome Medicine (PaCeMM) FHU, Paris, France.,Gastroenterology department, Saint Antoine Hospital, APHP, Paris, France
| | - Cecilia Landman
- Paris Center for Microbiome Medicine (PaCeMM) FHU, Paris, France.,Gastroenterology department, Saint Antoine Hospital, APHP, Paris, France
| | - Mathias L Richard
- Université Paris-Saclay, INRAe, AgroParisTech, Micalis institute, Jouy-en-Josas, France.,Paris Center for Microbiome Medicine (PaCeMM) FHU, Paris, France
| | - Patrick Emond
- UMR 1253, iBrain, University of Tours, Inserm, 37044 Tours, France.,CHRU Tours, Medical Biology Center, Tours, France
| | - Philippe Seksik
- Paris Center for Microbiome Medicine (PaCeMM) FHU, Paris, France.,Sorbonne Université, INSERM UMRS-938, Centre de Recherche Saint-Antoine, CRSA, AP-HP, Paris, France.,Gastroenterology department, Saint Antoine Hospital, APHP, Paris, France
| | - Laurent Beaugerie
- Paris Center for Microbiome Medicine (PaCeMM) FHU, Paris, France.,Gastroenterology department, Saint Antoine Hospital, APHP, Paris, France
| | - Rafael Rose Arguello
- Aix Marseille Univ, CNRS, INSERM, CIML, Centre d'Immunologie de Marseille-Luminy, Marseille, France
| | - David Moulin
- CNRS, IMoPA, Université de Lorraine, Vandoeuvre-lès-Nancy, France
| | - Sridhar Mani
- Molecular Pharmacology, Genetics and Medicine, Albert Einstein College of Medicine, Bronx, New York, USA
| | - Zdenek Dvorák
- Department of Cell Biology and Genetics, Faculty of Science, Palacky University, Olomouc, Czech Republic
| | - Luis G Bermúdez-Humarán
- Université Paris-Saclay, INRAe, AgroParisTech, Micalis institute, Jouy-en-Josas, France.,Paris Center for Microbiome Medicine (PaCeMM) FHU, Paris, France
| | - Philippe Langella
- Université Paris-Saclay, INRAe, AgroParisTech, Micalis institute, Jouy-en-Josas, France.,Paris Center for Microbiome Medicine (PaCeMM) FHU, Paris, France
| | - Harry Sokol
- Université Paris-Saclay, INRAe, AgroParisTech, Micalis institute, Jouy-en-Josas, France .,Paris Center for Microbiome Medicine (PaCeMM) FHU, Paris, France.,Sorbonne Université, INSERM UMRS-938, Centre de Recherche Saint-Antoine, CRSA, AP-HP, Paris, France.,Gastroenterology department, Saint Antoine Hospital, APHP, Paris, France
| |
Collapse
|
24
|
Bystrom J, Taher TE, Henson SM, Gould DJ, Mageed RA. Metabolic requirements of Th17 cells and of B cells: Regulation and defects in health and in inflammatory diseases. Front Immunol 2022; 13:990794. [PMCID: PMC9614365 DOI: 10.3389/fimmu.2022.990794] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2022] [Accepted: 09/06/2022] [Indexed: 11/13/2022] Open
Abstract
The immune system protects from infections and cancer through complex cellular networks. For this purpose, immune cells require well-developed mechanisms of energy generation. However, the immune system itself can also cause diseases when defective regulation results in the emergence of autoreactive lymphocytes. Recent studies provide insights into how differential patterns of immune cell responses are associated with selective metabolic pathways. This review will examine the changing metabolic requirements of Th17 cells and of B cells at different stages of their development and activation. Both cells provide protection but can also mediate diseases through the production of autoantibodies and the production of proinflammatory mediators. In health, B cells produce antibodies and cytokines and present antigens to T cells to mount specific immunity. Th17 cells, on the other hand, provide protection against extra cellular pathogens at mucosal surfaces but can also drive chronic inflammation. The latter cells can also promote the differentiation of B cells to plasma cells to produce more autoantibodies. Metabolism-regulated checkpoints at different stages of their development ensure the that self-reactive B cells clones and needless production of interleukin (IL-)17 are limited. The metabolic regulation of the two cell types has some similarities, e.g. the utility of hypoxia induced factor (HIF)1α during low oxygen tension, to prevent autoimmunity and regulate inflammation. There are also clear differences, as Th17 cells only are vulnerable to the lack of certain amino acids. B cells, unlike Th17 cells, are also dependent of mechanistic target of rapamycin 2 (mTORC2) to function. Significant knowledge has recently been gained, particularly on Th17 cells, on how metabolism regulates these cells through influencing their epigenome. Metabolic dysregulation of Th17 cells and B cells can lead to chronic inflammation. Disease associated alterations in the genome can, in addition, cause dysregulation to metabolism and, thereby, result in epigenetic alterations in these cells. Recent studies highlight how pathology can result from the cooperation between the two cell types but only few have so far addressed the key metabolic alterations in such settings. Knowledge of the impact of metabolic dysfunction on chronic inflammation and pathology can reveal novel therapeutic targets to treat such diseases.
Collapse
Affiliation(s)
- Jonas Bystrom
- Centre for Translational Medicine and Therapeutics, William Harvey Research Institute, Queen Mary University of London, London, United Kingdom
- *Correspondence: Jonas Bystrom, ; Taher E. Taher,
| | - Taher E. Taher
- Institute of Immunology and Immunotherapy, College of Medical and Dental Sciences, University of Birmingham, Birmingham, United Kingdom
- *Correspondence: Jonas Bystrom, ; Taher E. Taher,
| | - Sian M. Henson
- Centre for Translational Medicine and Therapeutics, William Harvey Research Institute, Queen Mary University of London, London, United Kingdom
| | - David J. Gould
- Centre for Biochemical Pharmacology, William Harvey Research Institute, Queen Mary University of London, London, United Kingdom
| | - Rizgar A. Mageed
- Centre for Translational Medicine and Therapeutics, William Harvey Research Institute, Queen Mary University of London, London, United Kingdom
| |
Collapse
|
25
|
Progranulin aggravates lethal Candida albicans sepsis by regulating inflammatory response and antifungal immunity. PLoS Pathog 2022; 18:e1010873. [PMID: 36121866 PMCID: PMC9521894 DOI: 10.1371/journal.ppat.1010873] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2022] [Revised: 09/29/2022] [Accepted: 09/09/2022] [Indexed: 11/19/2022] Open
Abstract
Candida albicans is the most frequent pathogen of fungal sepsis associated with substantial mortality in critically ill patients and those who are immunocompromised. Identification of novel immune-based therapeutic targets from a better understanding of its molecular pathogenesis is required. Here, we reported that the production of progranulin (PGRN) levels was significantly increased in mice after invasive C.albicans infection. Mice that lacked PGRN exhibited attenuated kidney injury and increased survival upon a lethal systemic infection with C. albicans. In mice, PGRN deficiency protected against systemic candidiasis by decreasing aberrant inflammatory reactions that led to renal immune cell apoptosis and kidney injury, and by enhancing antifungal capacity of macrophages and neutrophils that limited fungal burden in the kidneys. PGRN in hematopoietic cell compartment was important for this effect. Moreover, anti-PGRN antibody treatment limited renal inflammation and fungal burden and prolonged survival after invasive C. albicans infection. In vitro, PGRN loss increased phagocytosis, phagosome formation, reactive oxygen species production, neutrophil extracellular traps release, and killing activity in macrophages or neutrophils. Mechanistic studies demonstrated that PGRN loss up-regulated Dectin-2 expression, and enhanced spleen tyrosine kinase phosphorylation and extracellular signal-regulated kinase activation in macrophages and neutrophils. In summary, we identified PGRN as a critical factor that contributes to the immunopathology of invasive C.albicans infection, suggesting that targeting PGRN might serve as a novel treatment for fungal infection.
Collapse
|
26
|
Taylor TC, Li Y, Li DD, Majumder S, McGeachy MJ, Biswas PS, Gingras S, Gaffen SL. Arid5a Mediates an IL-17-Dependent Pathway That Drives Autoimmunity but Not Antifungal Host Defense. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2022; 209:1138-1145. [PMID: 35940634 PMCID: PMC9492638 DOI: 10.4049/jimmunol.2200132] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/15/2022] [Accepted: 07/11/2022] [Indexed: 01/04/2023]
Abstract
IL-17 contributes to the pathogenesis of certain autoimmune diseases, but conversely is essential for host defense against fungi. Ab-based biologic drugs that neutralize IL-17 are effective in autoimmunity but can be accompanied by adverse side effects. Candida albicans is a commensal fungus that is the primary causative agent of oropharyngeal and disseminated candidiasis. Defects in IL-17 signaling cause susceptibility to candidiasis in mice and humans. A key facet of IL-17 receptor signaling involves RNA-binding proteins, which orchestrate the fate of target mRNA transcripts. In tissue culture models we showed that the RNA-binding protein AT-rich interaction domain 5A (Arid5a) promotes the stability and/or translation of multiple IL-17-dependent mRNAs. Moreover, during oropharyngeal candidiasis, Arid5a is elevated within the oral mucosa in an IL-17-dependent manner. However, the contribution of Arid5a to IL-17-driven events in vivo is poorly defined. In this study, we used CRISPR-Cas9 to generate mice lacking Arid5a. Arid5a -/- mice were fully resistant to experimental autoimmune encephalomyelitis, an autoimmune setting in which IL-17 signaling drives pathology. Surprisingly, Arid5a -/- mice were resistant to oropharyngeal candidiasis and systemic candidiasis, similar to immunocompetent wild-type mice and contrasting with mice defective in IL-17 signaling. Therefore, Arid5a-dependent signals mediate pathology in autoimmunity and yet are not required for immunity to candidiasis, indicating that selective targeting of IL-17 signaling pathway components may be a viable strategy for development of therapeutics that spare IL-17-driven host defense.
Collapse
Affiliation(s)
- Tiffany C Taylor
- Division of Rheumatology & Clinical Immunology, University of Pittsburgh, Pittsburgh, PA; and
| | - Yang Li
- Division of Rheumatology & Clinical Immunology, University of Pittsburgh, Pittsburgh, PA; and
| | - De-Dong Li
- Division of Rheumatology & Clinical Immunology, University of Pittsburgh, Pittsburgh, PA; and
| | - Saikat Majumder
- Division of Rheumatology & Clinical Immunology, University of Pittsburgh, Pittsburgh, PA; and
| | - Mandy J McGeachy
- Division of Rheumatology & Clinical Immunology, University of Pittsburgh, Pittsburgh, PA; and
| | - Partha S Biswas
- Division of Rheumatology & Clinical Immunology, University of Pittsburgh, Pittsburgh, PA; and
| | | | - Sarah L Gaffen
- Division of Rheumatology & Clinical Immunology, University of Pittsburgh, Pittsburgh, PA; and
| |
Collapse
|
27
|
Sharma J, Mudalagiriyappa S, Nanjappa SG. T cell responses to control fungal infection in an immunological memory lens. Front Immunol 2022; 13:905867. [PMID: 36177012 PMCID: PMC9513067 DOI: 10.3389/fimmu.2022.905867] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2022] [Accepted: 08/22/2022] [Indexed: 11/24/2022] Open
Abstract
In recent years, fungal vaccine research emanated significant findings in the field of antifungal T-cell immunity. The generation of effector T cells is essential to combat many mucosal and systemic fungal infections. The development of antifungal memory T cells is integral for controlling or preventing fungal infections, and understanding the factors, regulators, and modifiers that dictate the generation of such T cells is necessary. Despite the deficiency in the clear understanding of antifungal memory T-cell longevity and attributes, in this review, we will compile some of the existing literature on antifungal T-cell immunity in the context of memory T-cell development against fungal infections.
Collapse
Affiliation(s)
| | | | - Som Gowda Nanjappa
- Department of Pathobiology, College of Veterinary Medicine, University of Illinois at Urbana-Champaign, Urbana, IL, United States
| |
Collapse
|
28
|
Le Bars P, Kouadio AA, Bandiaky ON, Le Guéhennec L, de La Cochetière MF. Host's Immunity and Candida Species Associated with Denture Stomatitis: A Narrative Review. Microorganisms 2022; 10:microorganisms10071437. [PMID: 35889156 PMCID: PMC9323190 DOI: 10.3390/microorganisms10071437] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2022] [Revised: 07/12/2022] [Accepted: 07/13/2022] [Indexed: 02/04/2023] Open
Abstract
Denture-related Candida stomatitis, which has been described clinically in the literature, is either localized or generalized inflammation of the oral mucosa in connection with a removable prosthesis. During this inflammatory process, the mycobacterial biofilm and the host’s immune response play an essential role. Among microorganisms of this mixed biofilm, the Candida species proliferates easily and changes from a commensal to an opportunistic pathogen. In this situation, the relationship between the Candida spp. and the host is influenced by the presence of the denture and conditioned both by the immune response and the oral microbiota. Specifically, this fungus is able to hijack the innate immune system of its host to cause infection. Additionally, older edentulous wearers of dentures may experience an imbalanced and decreased oral microbiome diversity. Under these conditions, the immune deficiency of these aging patients often promotes the spread of commensals and pathogens. The present narrative review aimed to analyze the innate and adaptive immune responses of patients with denture stomatitis and more particularly the involvement of Candida albicans sp. associated with this pathology.
Collapse
Affiliation(s)
- Pierre Le Bars
- Department of Prosthetic Dentistry, Faculty of Dentistry, Nantes University, 1 Place Alexis Ricordeau, 44042 Nantes, France; (A.A.K.); (O.N.B.); (L.L.G.)
- Correspondence: authors:
| | - Alain Ayepa Kouadio
- Department of Prosthetic Dentistry, Faculty of Dentistry, Nantes University, 1 Place Alexis Ricordeau, 44042 Nantes, France; (A.A.K.); (O.N.B.); (L.L.G.)
- Department of Prosthetic Dentistry, Faculty of Dentistry, CHU, Abidjan P.O. Box 612, Côte d’Ivoire
| | - Octave Nadile Bandiaky
- Department of Prosthetic Dentistry, Faculty of Dentistry, Nantes University, 1 Place Alexis Ricordeau, 44042 Nantes, France; (A.A.K.); (O.N.B.); (L.L.G.)
| | - Laurent Le Guéhennec
- Department of Prosthetic Dentistry, Faculty of Dentistry, Nantes University, 1 Place Alexis Ricordeau, 44042 Nantes, France; (A.A.K.); (O.N.B.); (L.L.G.)
| | - Marie-France de La Cochetière
- EA 3826 Thérapeutiques Cliniques Et expérimentales des Infections, Faculté de Médecine, CHU Hôtel-Dieu, Université de Nantes, 1, rue G. Veil, 44000 Nantes, France;
| |
Collapse
|
29
|
Datta A, Hernandez-Franco JF, Park S, Olson MR, HogenEsch H, Thangamani S. Bile Acid Regulates Mononuclear Phagocytes and T Helper 17 Cells to Control Candida albicans in the Intestine. J Fungi (Basel) 2022; 8:jof8060610. [PMID: 35736093 PMCID: PMC9224641 DOI: 10.3390/jof8060610] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2022] [Revised: 06/02/2022] [Accepted: 06/04/2022] [Indexed: 02/06/2023] Open
Abstract
Invasive Candida albicans (CA) infections often arise from the intestine and cause life-threatening infections in immunocompromised individuals. The role of gut commensal microbiota, metabolites, and host factors in the regulation of CA colonization in the intestine is poorly understood. Previous findings from our lab indicate that taurocholic acid (TCA), a major bile acid present in the intestine, promotes CA colonization and dissemination. Here, we report that oral administration of TCA to CA-infected mice significantly decreased the number of mononuclear phagocytes and CD4+ IL17A+ T helper 17 cells that play a critical role in controlling CA in the intestine. Collectively, our results indicate that TCA modulates mucosal innate and adaptive immune responses to promote CA colonization in the intestine.
Collapse
Affiliation(s)
- Abhishek Datta
- Department of Comparative Pathobiology, College of Veterinary Medicine, Purdue University, West Lafayette, IN 47906, USA; (A.D.); (J.F.H.-F.); (H.H.)
| | - Juan F. Hernandez-Franco
- Department of Comparative Pathobiology, College of Veterinary Medicine, Purdue University, West Lafayette, IN 47906, USA; (A.D.); (J.F.H.-F.); (H.H.)
| | - Sungtae Park
- Department of Biological Sciences, Purdue University, West Lafayette, IN 47906, USA; (S.P.); (M.R.O.)
| | - Matthew R. Olson
- Department of Biological Sciences, Purdue University, West Lafayette, IN 47906, USA; (S.P.); (M.R.O.)
| | - Harm HogenEsch
- Department of Comparative Pathobiology, College of Veterinary Medicine, Purdue University, West Lafayette, IN 47906, USA; (A.D.); (J.F.H.-F.); (H.H.)
- Purdue Institute for Immunology, Inflammation and Infectious Diseases (PI4D), West Lafayette, IN 47906, USA
| | - Shankar Thangamani
- Department of Comparative Pathobiology, College of Veterinary Medicine, Purdue University, West Lafayette, IN 47906, USA; (A.D.); (J.F.H.-F.); (H.H.)
- Purdue Institute for Immunology, Inflammation and Infectious Diseases (PI4D), West Lafayette, IN 47906, USA
- Correspondence: ; Tel.: +1-765-494-0763
| |
Collapse
|
30
|
Bruno M, Davidson L, Koenen HJPM, van den Reek JMPA, van Cranenbroek B, de Jong EMGJ, van de Veerdonk FL, Kullberg BJ, Netea MG. Immunological effects of anti-IL-17/12/23 therapy in patients with psoriasis complicated by Candida infections. J Invest Dermatol 2022; 142:2929-2939.e8. [PMID: 35662644 DOI: 10.1016/j.jid.2022.05.1083] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2022] [Revised: 05/08/2022] [Accepted: 05/24/2022] [Indexed: 12/12/2022]
Abstract
Biologics that block the T-helper-17 pathway are very effective in the treatment of psoriasis and other inflammatory diseases. However, interleukin-17 is also crucial for antifungal host defense, and clinical trial data suggest an increase in the incidence of Candida infections during IL-17 inhibitor (IL-17i) therapy. We investigated the innate and adaptive immune responses of psoriasis patients with a history of skin and/or mucosal candidiasis during IL-17i or IL-12/23i therapy, comparing those responses to healthy controls. Psoriasis patients with IL-17i showed significantly lower CD4+Th1-like (CCR6-CXCR3+CCR4-) and Th1Th17-like (CD4+CCR6+CXCR3+CCR4-) cell percentages. Patient cells stimulated with Candida albicans produced significantly lower IL-6 in the IL-12/23i group and IL-1β production in the IL-17i group, while the release of TNF-α and ROS was similar between patients and controls. IFN-γ and IL-10 production in response to several stimuli after 7 days was particularly decreased in patients receiving IL-17i therapy. Finally, after stimulation with the polarizing cytokines IL-1β and IL-23, the Th17 cytokine response was significantly lower in the IL-17i patient group. These innate and adaptive immune response defects can diminish antifungal host immune response and thereby increase susceptibility to candidiasis in patients treated with IL-17i or IL-12/23i.
Collapse
Affiliation(s)
- Mariolina Bruno
- Radboud University Medical Center Center for Infectious Diseases (RCI), Department of Internal Medicine, Radboud University Medical Center, Nijmegen, the Netherlands; Department of Internal Medicine, Radboud University Medical Center, Nijmegen, the Netherlands.
| | - Linda Davidson
- Radboud University Medical Center Center for Infectious Diseases (RCI), Department of Internal Medicine, Radboud University Medical Center, Nijmegen, the Netherlands
| | - Hans J P M Koenen
- Laboratory Medical Immunology, Radboud University Medical Center, the Netherlands
| | | | - Bram van Cranenbroek
- Laboratory Medical Immunology, Radboud University Medical Center, the Netherlands
| | - Elke M G J de Jong
- Department of Dermatology, Radboud University Medical Center, Nijmegen, the Netherlands
| | - Frank L van de Veerdonk
- Radboud University Medical Center Center for Infectious Diseases (RCI), Department of Internal Medicine, Radboud University Medical Center, Nijmegen, the Netherlands; Department of Internal Medicine, Radboud University Medical Center, Nijmegen, the Netherlands; Radboud Center for Infectious Disease (RCI), Radboud University Medical Center, Nijmegen, the Netherlands
| | - Bart-Jan Kullberg
- Radboud University Medical Center Center for Infectious Diseases (RCI), Department of Internal Medicine, Radboud University Medical Center, Nijmegen, the Netherlands
| | - Mihai G Netea
- Department of Internal Medicine, Radboud University Medical Center, Nijmegen, the Netherlands; Radboud Center for Infectious Disease (RCI), Radboud University Medical Center, Nijmegen, the Netherlands; Department of Immunology and Metabolism, Life and Medical Sciences Institute (LIMES), University of Bonn, Bonn, Germany
| |
Collapse
|
31
|
Gao C, Li X, Zhao X, Yang P, Wang X, Chen X, Chen N, Chen F. Standardized studies of the oral microbiome: From technology-driven to hypothesis-driven. IMETA 2022; 1:e19. [PMID: 38868569 PMCID: PMC10989927 DOI: 10.1002/imt2.19] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/30/2021] [Revised: 03/05/2022] [Accepted: 03/06/2022] [Indexed: 06/14/2024]
Abstract
The microbiome is in a symbiotic relationship with the host. Among the microbial consortia in the human body, that in the oral cavity is complex. Instead of repeatedly confirming biomarkers of oral and systemic diseases, recent studies have focused on a unified clinical diagnostic standard in microbiology that reduces the heterogeneity caused by individual discrepancies. Research has also been conducted on other topics of greater clinical importance, including bacterial pathogenesis, and the effects of drugs and treatments. In this review, we divide existing research into technology-driven and hypothesis-driven, according to whether there is a clear research hypothesis. This classification allows the demonstration of shifts in the direction of oral microbiology research. Based on the shifts, we suggested that establishing clear hypotheses may be the solution to major research challenges.
Collapse
Affiliation(s)
- Chuqi Gao
- Central LaboratoryPeking University Hospital of StomatologyBeijingChina
| | - Xuantao Li
- Central LaboratoryPeking University Hospital of StomatologyBeijingChina
| | - Xiaole Zhao
- Central LaboratoryPeking University Hospital of StomatologyBeijingChina
| | - Peiyue Yang
- Central LaboratoryPeking University Hospital of StomatologyBeijingChina
| | - Xiao Wang
- Central LaboratoryPeking University Hospital of StomatologyBeijingChina
| | - Xiaoli Chen
- Central LaboratoryPeking University Hospital of StomatologyBeijingChina
| | - Ning Chen
- Department of GastroenterologyPeking University People's HospitalBeijingChina
| | - Feng Chen
- Central LaboratoryPeking University Hospital of StomatologyBeijingChina
| |
Collapse
|
32
|
Swidergall M, LeibundGut-Landmann S. Immunosurveillance of Candida albicans commensalism by the adaptive immune system. Mucosal Immunol 2022; 15:829-836. [PMID: 35778599 PMCID: PMC9385492 DOI: 10.1038/s41385-022-00536-5] [Citation(s) in RCA: 28] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2022] [Revised: 05/27/2022] [Accepted: 05/31/2022] [Indexed: 02/04/2023]
Abstract
The fungal microbiota (mycobiota) is an integral part of the microbial community colonizing the body surfaces and is involved in many key aspects of human physiology, while an imbalance of the fungal communities, termed fungal dysbiosis, has been described in pathologies ranging from infections to inflammatory bowel disease. Commensal organisms, such as the fungus Candida albicans, induce antigen-specific immune responses that maintain immune homeostasis. Adaptive immune mechanisms are vital in this process, while deficiencies in adaptive immunity are linked to fungal infections. We start to understand the mechanisms by which a shift in mycobiota composition, in particular in C. albicans abundance, is linked to immunopathological conditions. This review discusses the mechanisms that ensure continuous immunosurveillance of C. albicans during mucosal colonization, how these protective adaptive immune responses can also promote immunopathology, and highlight therapeutic advances against C. albicans-associated disease.
Collapse
Affiliation(s)
- Marc Swidergall
- Division of Infectious Diseases, Harbor-UCLA Medical Center, Torrance, CA, USA
- The Lundquist Institute for Biomedical Innovation at Harbor-UCLA Medical Center, Torrance, CA, USA
- David Geffen School of Medicine at UCLA, Los Angeles, CA, USA
| | - Salomé LeibundGut-Landmann
- Section of Immunology, Vetsuisse Faculty, University of Zürich, Zürich, Switzerland.
- Institute of Experimental Immunology, University of Zürich, Zürich, Switzerland.
| |
Collapse
|
33
|
Oja AE, van Lier RAW, Hombrink P. Two sides of the same coin: Protective versus pathogenic CD4 + resident memory T cells. Sci Immunol 2022; 7:eabf9393. [PMID: 35394815 DOI: 10.1126/sciimmunol.abf9393] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2022]
Abstract
The ability of the adaptive immune system to form memory is key to providing protection against secondary infections. Resident memory T cells (TRM) are specialized T cell populations that reside within tissue sites where they await reencounter with their cognate antigen. TRM are distinct from circulating memory cells, including central and effector memory T cells, both functionally and transcriptionally. Since the discovery of TRM, most research has focused on CD8+ TRM, despite that CD4+ TRM are also abundant in most tissues. In the past few years, more evidence has emerged that CD4+ TRM can contribute both protective and pathogenic roles in disease. A complexity inherent to the CD4+ TRM field is the ability of CD4+ T cells to polarize into a multitude of distinct subsets and recognize not only viruses and intracellular bacteria but also extracellular bacteria, fungi, and parasites. In this review, we outline the key features of CD4+ TRM in health and disease, including their contributions to protection against SARS-CoV-2 and potential contributions to immunopathology associated with COVID-19.
Collapse
Affiliation(s)
- Anna E Oja
- Department of Hematopoiesis, Sanquin Research and Landsteiner Laboratory, Amsterdam UMC, University of Amsterdam, Amsterdam, Netherlands
| | - René A W van Lier
- Department of Hematopoiesis, Sanquin Research and Landsteiner Laboratory, Amsterdam UMC, University of Amsterdam, Amsterdam, Netherlands
| | - Pleun Hombrink
- Department of Hematopoiesis, Sanquin Research and Landsteiner Laboratory, Amsterdam UMC, University of Amsterdam, Amsterdam, Netherlands
| |
Collapse
|
34
|
Lemberg C, Martinez de San Vicente K, Fróis-Martins R, Altmeier S, Tran VDT, Mertens S, Amorim-Vaz S, Rai LS, d’Enfert C, Pagni M, Sanglard D, LeibundGut-Landmann S. Candida albicans commensalism in the oral mucosa is favoured by limited virulence and metabolic adaptation. PLoS Pathog 2022; 18:e1010012. [PMID: 35404986 PMCID: PMC9041809 DOI: 10.1371/journal.ppat.1010012] [Citation(s) in RCA: 31] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2021] [Revised: 04/26/2022] [Accepted: 03/17/2022] [Indexed: 12/19/2022] Open
Abstract
As part of the human microbiota, the fungus Candida albicans colonizes the oral cavity and other mucosal surfaces of the human body. Commensalism is tightly controlled by complex interactions of the fungus and the host to preclude fungal elimination but also fungal overgrowth and invasion, which can result in disease. As such, defects in antifungal T cell immunity render individuals susceptible to oral thrush due to interrupted immunosurveillance of the oral mucosa. The factors that promote commensalism and ensure persistence of C. albicans in a fully immunocompetent host remain less clear. Using an experimental model of C. albicans oral colonization in mice we explored fungal determinants of commensalism in the oral cavity. Transcript profiling of the oral isolate 101 in the murine tongue tissue revealed a characteristic metabolic profile tailored to the nutrient poor conditions in the stratum corneum of the epithelium where the fungus resides. Metabolic adaptation of isolate 101 was also reflected in enhanced nutrient acquisition when grown on oral mucosa substrates. Persistent colonization of the oral mucosa by C. albicans also correlated inversely with the capacity of the fungus to induce epithelial cell damage and to elicit an inflammatory response. Here we show that these immune evasive properties of isolate 101 are explained by a strong attenuation of a number of virulence genes, including those linked to filamentation. De-repression of the hyphal program by deletion or conditional repression of NRG1 abolished the commensal behaviour of isolate 101, thereby establishing a central role of this factor in the commensal lifestyle of C. albicans in the oral niche of the host. The oral microbiota represents an important part of the human microbiota and includes several hundreds to several thousands of bacterial and fungal species. One of the most prominent fungus colonizing the oral cavity is the yeast Candida albicans. While the presence of C. albicans usually remains unnoticed, the fungus can under certain circumstances cause lesions on the lining of the mouth referred to as oral thrush or contribute to other common oral diseases such as caries. Maintaining C. albicans commensalism in the oral mucosa is therefore of utmost importance for oral health and overall wellbeing. While overt fungal growth and disease is limited by immunosurveillance mechanisms during homeostasis, C. albicans strives to survive and evades elimination from the host. Here, we show that while commensalism in the oral cavity is characterized by a restricted fungal virulence and hyphal program, enforcing filamentation in a commensal isolate is sufficient for driving pathogenicity and fungus-induced inflammation in the oral mucosa thwarting persistent colonization. Our results further support a critical role for specialized nutrient acquisition allowing the fungus to thrive in the nutrient poor environment of the squamous epithelium. Together, this work revealed key determinants of C. albicans commensalism in the oral niche.
Collapse
Affiliation(s)
- Christina Lemberg
- Section of Immunology, Vetsuisse Faculty, University of Zürich, Zürich, Switzerland
- Institute of Experimental Immunology, University of Zürich, Zürich, Switzerland
| | - Kontxi Martinez de San Vicente
- Section of Immunology, Vetsuisse Faculty, University of Zürich, Zürich, Switzerland
- Institute of Experimental Immunology, University of Zürich, Zürich, Switzerland
| | - Ricardo Fróis-Martins
- Section of Immunology, Vetsuisse Faculty, University of Zürich, Zürich, Switzerland
- Institute of Experimental Immunology, University of Zürich, Zürich, Switzerland
| | - Simon Altmeier
- Section of Immunology, Vetsuisse Faculty, University of Zürich, Zürich, Switzerland
- Institute of Experimental Immunology, University of Zürich, Zürich, Switzerland
| | - Van Du T. Tran
- Vital-IT Group, SIB Swiss Institute of Bioinformatics, Lausanne, Switzerland
| | - Sarah Mertens
- Section of Immunology, Vetsuisse Faculty, University of Zürich, Zürich, Switzerland
- Institute of Experimental Immunology, University of Zürich, Zürich, Switzerland
| | - Sara Amorim-Vaz
- Institute of Microbiology, University of Lausanne and University Hospital Center, Lausanne, Switzerland
| | - Laxmi Shanker Rai
- Institut Pasteur, Université de Paris, INRAE, USC2019, Unité Biologie et Pathogénicité Fongiques, Paris, France
| | - Christophe d’Enfert
- Institut Pasteur, Université de Paris, INRAE, USC2019, Unité Biologie et Pathogénicité Fongiques, Paris, France
| | - Marco Pagni
- Vital-IT Group, SIB Swiss Institute of Bioinformatics, Lausanne, Switzerland
| | - Dominique Sanglard
- Institute of Microbiology, University of Lausanne and University Hospital Center, Lausanne, Switzerland
| | - Salomé LeibundGut-Landmann
- Section of Immunology, Vetsuisse Faculty, University of Zürich, Zürich, Switzerland
- Institute of Experimental Immunology, University of Zürich, Zürich, Switzerland
- * E-mail:
| |
Collapse
|
35
|
Li S, Su B, He QS, Wu H, Zhang T. Alterations in the oral microbiome in HIV infection: causes, effects and potential interventions. Chin Med J (Engl) 2021; 134:2788-2798. [PMID: 34670249 PMCID: PMC8667981 DOI: 10.1097/cm9.0000000000001825] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2021] [Indexed: 12/02/2022] Open
Abstract
ABSTRACT A massive depletion of CD4+ T lymphocytes has been described in early and acute human immunodeficiency virus (HIV) infection, leading to an imbalance between the human microbiome and immune responses. In recent years, a growing interest in the alterations in gut microbiota in HIV infection has led to many studies; however, only few studies have been conducted to explore the importance of oral microbiome in HIV-infected individuals. Evidence has indicated the dysbiosis of oral microbiota in people living with HIV (PLWH). Potential mechanisms might be related to the immunodeficiency in the oral cavity of HIV-infected individuals, including changes in secretory components such as reduced levels of enzymes and proteins in saliva and altered cellular components involved in the reduction and dysfunction of innate and adaptive immune cells. As a result, disrupted oral immunity in HIV-infected individuals leads to an imbalance between the oral microbiome and local immune responses, which may contribute to the development of HIV-related diseases and HIV-associated non-acquired immunodeficiency syndrome comorbidities. Although the introduction of antiretroviral therapy (ART) has led to a significant decrease in occurrence of the opportunistic oral infections in HIV-infected individuals, the dysbiosis in oral microbiome persists. Furthermore, several studies with the aim to investigate the ability of probiotics to regulate the dysbiosis of oral microbiota in HIV-infected individuals are ongoing. However, the effects of ART and probiotics on oral microbiome in HIV-infected individuals remain unclear. In this article, we review the composition of the oral microbiome in healthy and HIV-infected individuals and the possible effect of oral microbiome on HIV-associated oral diseases. We also discuss how ART and probiotics influence the oral microbiome in HIV infection. We believe that a deeper understanding of composition and function of the oral microbiome is critical for the development of effective preventive and therapeutic strategies for HIV infection.
Collapse
Affiliation(s)
- Shuang Li
- Beijing Key Laboratory for HIV/AIDS Research, Clinical and Research Center for Infectious Diseases, Beijing Youan Hospital, Capital Medical University, Beijing 100069, China
| | - Bin Su
- Beijing Key Laboratory for HIV/AIDS Research, Clinical and Research Center for Infectious Diseases, Beijing Youan Hospital, Capital Medical University, Beijing 100069, China
| | - Qiu-Shui He
- Institute of Biomedicine, Research Center for Infections and Immunity, University of Turku, Turku 20520, Finland
| | - Hao Wu
- Beijing Key Laboratory for HIV/AIDS Research, Clinical and Research Center for Infectious Diseases, Beijing Youan Hospital, Capital Medical University, Beijing 100069, China
| | - Tong Zhang
- Beijing Key Laboratory for HIV/AIDS Research, Clinical and Research Center for Infectious Diseases, Beijing Youan Hospital, Capital Medical University, Beijing 100069, China
| |
Collapse
|
36
|
Li S, Zhu J, Su B, Wei H, Chen F, Liu H, Wei J, Yang X, Zhang Q, Xia W, Wu H, He Q, Zhang T. Alteration in Oral Microbiome Among Men Who Have Sex With Men With Acute and Chronic HIV Infection on Antiretroviral Therapy. Front Cell Infect Microbiol 2021; 11:695515. [PMID: 34336719 PMCID: PMC8317457 DOI: 10.3389/fcimb.2021.695515] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2021] [Accepted: 06/24/2021] [Indexed: 12/12/2022] Open
Abstract
Despite the antiretroviral therapy (ART), human immunodeficiency virus (HIV)-related oral disease remains a common problem for people living with HIV (PLWH). Evidence suggests that impairment of immune function in HIV infection might lead to the conversion of commensal bacteria to microorganisms with increased pathogenicity. However, limited information is available about alteration in oral microbiome in PLWH on ART. We performed a longitudinal comparative study on men who have sex with men (MSM) with acute HIV infection (n=15), MSM with chronic HIV infection (n=15), and HIV-uninfected MSM controls (n=15). Throat swabs were collected when these subjects were recruited (W0) and 12 weeks after ART treatment (W12) from the patients. Genomic DNAs were extracted and 16S rRNA gene sequencing was performed. Microbiome diversity was significantly decreased in patients with acute and chronic HIV infections compared with those in controls at the sampling time of W0 and the significant difference remained at W12. An increased abundance of unidentified Prevotellaceae was found in patients with acute and chronic HIV infections. Moreover, increased abundances of Prevotella in subjects with acute HIV infection and Streptococcus in subjects with chronic HIV infection were observed. In contrast, greater abundance in Lactobacillus, Rothia, Lautropia, and Bacteroides was found in controls. After effective ART, Bradyrhizobium was enriched in both acute and chronic HIV infections, whereas in controls, Lactobacillus, Rothia, Clostridia, Actinobacteria, and Ruminococcaceae were enriched. In addition, we found that lower CD4+ T-cell counts (<200 cells/mm3) were associated with lower relative abundances of Haemophilus, Actinomyces, unidentified Ruminococcaceae, and Rothia. This study has shown alteration in oral microbiome resulting from HIV infection and ART. The results obtained warrant further studies in a large number of subjects with different ethnics. It might contribute to improved oral health in HIV-infected individuals.
Collapse
Affiliation(s)
- Shuang Li
- Beijing Key Laboratory for HIV/AIDS Research, Center for Infectious Diseases, Beijing Youan Hospital, Capital Medical University, Beijing, China
| | - Junping Zhu
- Department of Medical Microbiology, Capital Medical University, Beijing, China
| | - Bin Su
- Beijing Key Laboratory for HIV/AIDS Research, Center for Infectious Diseases, Beijing Youan Hospital, Capital Medical University, Beijing, China
| | - Huanhuan Wei
- Department of Medical Microbiology, Capital Medical University, Beijing, China
| | - Fei Chen
- Department of Medical Microbiology, Capital Medical University, Beijing, China
| | - Hongshan Liu
- Department of Medical Microbiology, Capital Medical University, Beijing, China
| | - Jiaqi Wei
- Beijing Key Laboratory for HIV/AIDS Research, Center for Infectious Diseases, Beijing Youan Hospital, Capital Medical University, Beijing, China
| | - Xiaodong Yang
- Beijing Key Laboratory for HIV/AIDS Research, Center for Infectious Diseases, Beijing Youan Hospital, Capital Medical University, Beijing, China
| | - Qiuyue Zhang
- Beijing Key Laboratory for HIV/AIDS Research, Center for Infectious Diseases, Beijing Youan Hospital, Capital Medical University, Beijing, China
| | - Wei Xia
- Beijing Key Laboratory for HIV/AIDS Research, Center for Infectious Diseases, Beijing Youan Hospital, Capital Medical University, Beijing, China
| | - Hao Wu
- Beijing Key Laboratory for HIV/AIDS Research, Center for Infectious Diseases, Beijing Youan Hospital, Capital Medical University, Beijing, China
| | - Qiushui He
- Department of Medical Microbiology, Capital Medical University, Beijing, China.,Institute of Biomedicine, Research Center for Infections and Immunity, University of Turku, Turku, Finland
| | - Tong Zhang
- Beijing Key Laboratory for HIV/AIDS Research, Center for Infectious Diseases, Beijing Youan Hospital, Capital Medical University, Beijing, China
| |
Collapse
|
37
|
Zubeidat K, Hovav AH. Shaped by the epithelium - postnatal immune mechanisms of oral homeostasis. Trends Immunol 2021; 42:622-634. [PMID: 34083119 DOI: 10.1016/j.it.2021.05.006] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2021] [Revised: 05/05/2021] [Accepted: 05/06/2021] [Indexed: 02/07/2023]
Abstract
The first encounter of mucosal barriers with the microbiota initiates host-microbiota feedback loops instructing the tailored development of both the immune system and microbiota at each mucosal site. Once established, balanced immunological interactions enable symbiotic relationships with the microbiota in adult life. This process has been extensively investigated in the mammalian monolayer epithelium-covered intestine and lung mucosae; however, the postnatal mechanisms engaged by the oral mucosa to establish homeostasis are currently being discovered. Here, we discuss the early life dialogue between the oral mucosa and the microbiota, with particular emphasis on the central role the multilayer epithelium plays to protect the oral mucosa. These intricate and unique postnatal immunological processes shape oral homeostasis, which can potentially affect buccal and systemic health in adult life.
Collapse
Affiliation(s)
- Khaled Zubeidat
- The Institute of Biomedical and Oral Research, Faculty of Dental Medicine, Hebrew University, Jerusalem, Israel
| | - Avi-Hai Hovav
- The Institute of Biomedical and Oral Research, Faculty of Dental Medicine, Hebrew University, Jerusalem, Israel.
| |
Collapse
|
38
|
LeibundGut-Landmann S. Tissue-Resident Memory T Cells in Antifungal Immunity. Front Immunol 2021; 12:693055. [PMID: 34113356 PMCID: PMC8185520 DOI: 10.3389/fimmu.2021.693055] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2021] [Accepted: 05/10/2021] [Indexed: 12/26/2022] Open
Abstract
Fungi are an integral part of the mammalian microbiota colonizing most if not all mucosal surfaces and the skin. Maintaining stable colonization on these surfaces is critical for preventing fungal dysbiosis and infection, which in some cases can lead to life threatening consequences. The epithelial barriers are protected by T cells and additional controlling immune mechanisms. Noncirculating memory T cells that reside stably in barrier tissues play an important role for host protection from commensals and recurrent pathogens due to their fast response and local activity, which provides them a strategic advantage. So far, only a few specific examples of tissue resident memory T cells (TRMs) that act against fungi have been reported. This review provides an overview of the characteristics and functional attributes of TRMs that have been established based on human and mouse studies with various microbes. It highlights what is currently known about fungi specific TRMs mediating immunosurveillance, how they have been targeted in preclinical vaccination approaches and how they can promote immunopathology, if not controlled. A better appreciation of the host protective and damaging roles of TRMs might accelerate the development of novel tissue specific preventive strategies against fungal infections and fungi-driven immunopathologies.
Collapse
Affiliation(s)
- Salomé LeibundGut-Landmann
- Section of Immunology, Vetsuisse Faculty, University of Zürich, Zürich, Switzerland
- Institute of Experimental Immunology, University of Zürich, Zürich, Switzerland
| |
Collapse
|
39
|
Kume M, Kiyohara E, Matsumura Y, Koguchi-Yoshioka H, Tanemura A, Hanaoka Y, Taminato M, Tashima H, Tomita K, Kubo T, Watanabe R, Fujimoto M. Ganglioside GD3 May Suppress the Functional Activities of Benign Skin T Cells in Cutaneous T-Cell Lymphoma. Front Immunol 2021; 12:651048. [PMID: 33859643 PMCID: PMC8042233 DOI: 10.3389/fimmu.2021.651048] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2021] [Accepted: 03/15/2021] [Indexed: 01/11/2023] Open
Abstract
In cutaneous T-cell lymphoma (CTCL), which arises from skin-tropic memory T cells, malignant T cells and benign T cells are confined in the same skin lesions. It is thus difficult to evaluate the phenotypic characteristics and functional activities of benign T cells in CTCL. Disialoganglioside with three glycosyl groups (GD3) is increasingly expressed on the surface of solid malignant tumor cells and takes part in tumor progression and suppression of tumor immunity. However, the role of GD3 in CTCL is not well-understood. In this study, the malignant and benign T cells in CTCL skin lesions were distinguished by flow cytometry and their phenotypic characteristics were compared with those of T cells from control skin specimens. In CTCL skin lesions, the benign T cells included limited resident memory T cells (TRM), which are sessile in skin and known to exert strong antitumor function. The benign T cells showed diminished Th17 property, and the expression of GD3 was high in the malignant T cells. The expression of GD3 in the malignant T cells inversely correlated with IL-17A production from the benign CD4 T cells. GD3 from the malignant T cells was implied to be involved in suppressing the Th17 activity of the benign T cells independent of the regulation of TRM differentiation in CTCL. Revealing the role of GD3 in inhibiting the production of IL-17A in CTCL would aid the understanding of the suppressive mechanism of the antitumor activity by malignant tumor cells.
Collapse
Affiliation(s)
- Miki Kume
- Department of Dermatology, Course of Integrated Medicine, Graduate School of Medicine, Osaka University, Osaka, Japan
| | - Eiji Kiyohara
- Department of Dermatology, Course of Integrated Medicine, Graduate School of Medicine, Osaka University, Osaka, Japan
| | - Yutaka Matsumura
- Department of Dermatology, Course of Integrated Medicine, Graduate School of Medicine, Osaka University, Osaka, Japan
| | - Hanako Koguchi-Yoshioka
- Department of Dermatology, Course of Integrated Medicine, Graduate School of Medicine, Osaka University, Osaka, Japan.,Department of Integrative Medicine for Allergic and Immunological Diseases, Course of Integrated Medicine, Graduate School of Medicine, Osaka University, Osaka, Japan
| | - Atsushi Tanemura
- Department of Dermatology, Course of Integrated Medicine, Graduate School of Medicine, Osaka University, Osaka, Japan
| | - Yuma Hanaoka
- Department of Dermatology, Course of Integrated Medicine, Graduate School of Medicine, Osaka University, Osaka, Japan
| | - Mifue Taminato
- Department of Plastic Surgery, Course of Organ Regulation Medicine, Graduate School of Medicine, Osaka University, Osaka, Japan
| | - Hiroki Tashima
- Department of Plastic Surgery, Course of Organ Regulation Medicine, Graduate School of Medicine, Osaka University, Osaka, Japan
| | - Koichi Tomita
- Department of Plastic Surgery, Course of Organ Regulation Medicine, Graduate School of Medicine, Osaka University, Osaka, Japan
| | - Tateki Kubo
- Department of Plastic Surgery, Course of Organ Regulation Medicine, Graduate School of Medicine, Osaka University, Osaka, Japan
| | - Rei Watanabe
- Department of Dermatology, Course of Integrated Medicine, Graduate School of Medicine, Osaka University, Osaka, Japan.,Department of Integrative Medicine for Allergic and Immunological Diseases, Course of Integrated Medicine, Graduate School of Medicine, Osaka University, Osaka, Japan
| | - Manabu Fujimoto
- Department of Dermatology, Course of Integrated Medicine, Graduate School of Medicine, Osaka University, Osaka, Japan
| |
Collapse
|
40
|
Estrada Brull A, Rost F, Oderbolz J, Kirchner FR, Leibundgut-Landmann S, Oxenius A, Joller N. CD85k Contributes to Regulatory T Cell Function in Chronic Viral Infections. Int J Mol Sci 2020; 22:E31. [PMID: 33375121 PMCID: PMC7792974 DOI: 10.3390/ijms22010031] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2020] [Revised: 12/16/2020] [Accepted: 12/17/2020] [Indexed: 12/20/2022] Open
Abstract
Regulatory T cells (Tregs) prevent excessive immune responses and limit immune pathology upon infections. To fulfill this role in different immune environments elicited by different types of pathogens, Tregs undergo functional specialization into distinct subsets. During acute type 1 immune responses, type 1 Tregs are induced and recruited to the site of ongoing Th1 responses to efficiently control Th1 responses. However, whether a similar specialization process also takes place following chronic infections is still unknown. In this study, we investigated Treg specialization in persistent viral infections using lymphocytic choriomeningitis virus (LCMV) and murine cytomegalovirus (MCMV) infection as models for chronic and latent infections, respectively. We identify CD85k as a Th1-specific co-inhibitory receptor with sustained expression in persistent viral infections and show that recombinant CD85k inhibits LCMV-specific effector T cells. Furthermore, expression of the CD85k ligand ALCAM is induced on LCMV-specific and exhausted T cells during chronic LCMV infection. Finally, we demonstrate that type 1 Tregs arising during chronic LCMV infection suppress Th1 effector cells in an ALCAM-dependent manner. These results extend the current knowledge of Treg specialization from acute to persistent viral infections and reveal an important functional role of CD85k in Treg-mediated suppression of type 1 immunity.
Collapse
MESH Headings
- Animals
- Antigens, CD/immunology
- Antigens, CD/metabolism
- Cell Adhesion Molecules, Neuronal/immunology
- Cell Adhesion Molecules, Neuronal/metabolism
- Cell Line
- Cells, Cultured
- Herpesviridae Infections/immunology
- Herpesviridae Infections/metabolism
- Herpesviridae Infections/virology
- Lymphocytic Choriomeningitis/immunology
- Lymphocytic Choriomeningitis/metabolism
- Lymphocytic Choriomeningitis/virology
- Lymphocytic choriomeningitis virus/immunology
- Lymphocytic choriomeningitis virus/physiology
- Membrane Glycoproteins/immunology
- Membrane Glycoproteins/metabolism
- Mice, Inbred C57BL
- Muromegalovirus/immunology
- Muromegalovirus/physiology
- Receptors, Immunologic/immunology
- Receptors, Immunologic/metabolism
- T-Lymphocytes, Regulatory/immunology
- T-Lymphocytes, Regulatory/metabolism
- T-Lymphocytes, Regulatory/virology
- Th1 Cells/immunology
- Th1 Cells/metabolism
Collapse
Affiliation(s)
- Anna Estrada Brull
- Institute of Experimental Immunology, University of Zurich, 8057 Zurich, Switzerland; (A.E.B.); (F.R.); (F.R.K.); (S.L.-L.)
| | - Felix Rost
- Institute of Experimental Immunology, University of Zurich, 8057 Zurich, Switzerland; (A.E.B.); (F.R.); (F.R.K.); (S.L.-L.)
| | - Josua Oderbolz
- ETH Zurich, Institute of Microbiology, 8093 Zurich, Switzerland; (J.O.); (A.O.)
| | - Florian R. Kirchner
- Institute of Experimental Immunology, University of Zurich, 8057 Zurich, Switzerland; (A.E.B.); (F.R.); (F.R.K.); (S.L.-L.)
- Section of Immunology, Vetsuisse Faculty, University of Zurich, 8057 Zurich, Switzerland
| | - Salomé Leibundgut-Landmann
- Institute of Experimental Immunology, University of Zurich, 8057 Zurich, Switzerland; (A.E.B.); (F.R.); (F.R.K.); (S.L.-L.)
- Section of Immunology, Vetsuisse Faculty, University of Zurich, 8057 Zurich, Switzerland
| | - Annette Oxenius
- ETH Zurich, Institute of Microbiology, 8093 Zurich, Switzerland; (J.O.); (A.O.)
| | - Nicole Joller
- Institute of Experimental Immunology, University of Zurich, 8057 Zurich, Switzerland; (A.E.B.); (F.R.); (F.R.K.); (S.L.-L.)
| |
Collapse
|
41
|
Scheffold A, Bacher P, LeibundGut-Landmann S. T cell immunity to commensal fungi. Curr Opin Microbiol 2020; 58:116-123. [PMID: 33120172 DOI: 10.1016/j.mib.2020.09.008] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2020] [Revised: 09/12/2020] [Accepted: 09/15/2020] [Indexed: 02/07/2023]
Abstract
Fungi are an important part of the microbiota in healthy barrier tissues. Fungal dysbiosis in turn is associated with local and distal inflammatory diseases. Recent advances have shed light on the antigen-specific IL-17-dependent mechanisms that regulate fungal commensalism and prevent fungal overgrowth during homeostasis. Progress in our understanding of species-specific differences in fungus-host interactions provides new hypotheses of why Candida albicans-targeting T cells exceed those directed against other fungal species in the human T cell repertoire. Importantly, C. albicans-specific Th17 cells can also contribute to immune pathology in distant organs such as the lung via cross-reaction with heterologous antigens.
Collapse
Affiliation(s)
- Alexander Scheffold
- Institute of Immunology, Christian-Albrechts Universität zu Kiel and Universitätsklinik Schleswig-Holstein, Kiel, Germany
| | - Petra Bacher
- Institute of Immunology, Christian-Albrechts Universität zu Kiel and Universitätsklinik Schleswig-Holstein, Kiel, Germany; Institute of Clinical Molecular Biology, Christian-Albrechts Universität zu Kiel, Kiel, Germany
| | - Salomé LeibundGut-Landmann
- Section of Immunology, Vetsuisse Faculty, University of Zürich, Switzerland; Institute of Experimental Immunology, University of Zürich, Switzerland.
| |
Collapse
|
42
|
Millet N, Solis NV, Swidergall M. Mucosal IgA Prevents Commensal Candida albicans Dysbiosis in the Oral Cavity. Front Immunol 2020; 11:555363. [PMID: 33193324 PMCID: PMC7642201 DOI: 10.3389/fimmu.2020.555363] [Citation(s) in RCA: 38] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2020] [Accepted: 09/03/2020] [Indexed: 12/15/2022] Open
Abstract
The fungus Candida albicans colonizes the oral mucosal surface of 30–70% of healthy individuals. Due to local or systemic immunosuppression, this commensal fungus is able to proliferate resulting in oral disease, called oropharyngeal candidiasis (OPC). However, in healthy individuals C. albicans causes no harm. Unlike humans mice do not host C. albicans in their mycobiome. Thus, oral fungal challenge generates an acute immune response in a naive host. Therefore, we utilized C. albicans clinical isolates which are able to persist in the oral cavity without causing disease to analyze adaptive responses to oral fungal commensalism. We performed RNA sequencing to determine the transcriptional host response landscape during C. albicans colonization. Pathway analysis revealed an upregulation of adaptive host responses due to C. albicans oral persistence, including the upregulation of the immune network for IgA production. Fungal colonization increased cross-specific IgA levels in the saliva and the tongue, and IgA+ cells migrated to foci of fungal colonization. Binding of IgA prevented fungal epithelial adhesion and invasion resulting in a dampened proinflammatory epithelial response. Besides CD19+ CD138− B cells, plasmablasts, and plasma cells were enriched in the tongue of mice colonized with C. albicans suggesting a potential role of B lymphocytes during oral fungal colonization. B cell deficiency increased the oral fungal load without causing severe OPC. Thus, in the oral cavity B lymphocytes contribute to control commensal C. albicans carriage by secreting IgA at foci of colonization thereby preventing fungal dysbiosis.
Collapse
Affiliation(s)
- Nicolas Millet
- Division of Infectious Diseases, Harbor-UCLA Medical Center, Torrance, CA, United States.,Institute for Infection and Immunity, The Lundquist Institute at Harbor-UCLA Medical Center, Torrance, CA, United States
| | - Norma V Solis
- Division of Infectious Diseases, Harbor-UCLA Medical Center, Torrance, CA, United States.,Institute for Infection and Immunity, The Lundquist Institute at Harbor-UCLA Medical Center, Torrance, CA, United States
| | - Marc Swidergall
- Division of Infectious Diseases, Harbor-UCLA Medical Center, Torrance, CA, United States.,Institute for Infection and Immunity, The Lundquist Institute at Harbor-UCLA Medical Center, Torrance, CA, United States.,David Geffen School of Medicine at UCLA, Los Angeles, CA, United States
| |
Collapse
|