151
|
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
|
152
|
Dhariwala MO, Karthikeyan D, Vasquez KS, Farhat S, Weckel A, Taravati K, Leitner EG, Clancy S, Pauli M, Piper ML, Cohen JN, Ashouri JF, Lowe MM, Rosenblum MD, Scharschmidt TC. Developing Human Skin Contains Lymphocytes Demonstrating a Memory Signature. CELL REPORTS MEDICINE 2020; 1:100132. [PMID: 33294857 PMCID: PMC7691438 DOI: 10.1016/j.xcrm.2020.100132] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/19/2020] [Revised: 08/20/2020] [Accepted: 10/13/2020] [Indexed: 12/17/2022]
Abstract
Lymphocytes in barrier tissues play critical roles in host defense and homeostasis. These cells take up residence in tissues during defined developmental windows, when they may demonstrate distinct phenotypes and functions. Here, we utilized mass and flow cytometry to elucidate early features of human skin immunity. Although most conventional αβ T (Tconv) cells in fetal skin have a naive, proliferative phenotype, a subset of CD4+ Tconv and CD8+ cells demonstrate memory-like features and a propensity for interferon (IFN)γ production. Skin regulatory T cells dynamically accumulate over the second trimester in temporal and regional association with hair follicle development. These fetal skin regulatory T cells (Tregs) demonstrate an effector memory phenotype while differing from their adult counterparts in expression of key effector molecules. Thus, we identify features of prenatal skin lymphocytes that may have key implications for understanding antigen and allergen encounters in utero and in infancy. CyTOF reveals a complex lymphocyte landscape in developing human skin Developing skin contains CD45RO+ conventional T cells with propensity to produce IFNγ Regulatory T cells (Tregs) in skin before birth display effector memory properties Skin Tregs increase in conjunction with initial hair follicle morphogenesis
Collapse
Affiliation(s)
- Miqdad O Dhariwala
- Department of Dermatology, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Dhuvarakesh Karthikeyan
- Department of Dermatology, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Kimberly S Vasquez
- Department of Dermatology, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Sepideh Farhat
- Department of Dermatology, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Antonin Weckel
- Department of Dermatology, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Keyon Taravati
- Department of Dermatology, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Elizabeth G Leitner
- Department of Dermatology, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Sean Clancy
- Department of Dermatology, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Mariela Pauli
- Department of Dermatology, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Merisa L Piper
- Division of Plastic and Reconstructive Surgery, Department of Surgery, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Jarish N Cohen
- Department of Dermatology, University of California, San Francisco, San Francisco, CA 94143, USA.,Department of Pathology, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Judith F Ashouri
- Rosalind Russell and Ephraim P. Engleman Rheumatology Research Center, Division of Rheumatology, Department of Medicine, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Margaret M Lowe
- Department of Dermatology, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Michael D Rosenblum
- Department of Dermatology, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Tiffany C Scharschmidt
- Department of Dermatology, University of California, San Francisco, San Francisco, CA 94143, USA
| |
Collapse
|
153
|
Wang T, Shen Y, Luyten S, Yang Y, Jiang X. Tissue-resident memory CD8+ T cells in cancer immunology and immunotherapy. Pharmacol Res 2020; 159:104876. [DOI: 10.1016/j.phrs.2020.104876] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/29/2020] [Revised: 04/26/2020] [Accepted: 04/27/2020] [Indexed: 02/07/2023]
|
154
|
Ruff WE, Greiling TM, Kriegel MA. Host-microbiota interactions in immune-mediated diseases. Nat Rev Microbiol 2020; 18:521-538. [PMID: 32457482 DOI: 10.1038/s41579-020-0367-2] [Citation(s) in RCA: 301] [Impact Index Per Article: 60.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/08/2020] [Indexed: 12/22/2022]
Abstract
Host-microbiota interactions are fundamental for the development of the immune system. Drastic changes in modern environments and lifestyles have led to an imbalance of this evolutionarily ancient process, coinciding with a steep rise in immune-mediated diseases such as autoimmune, allergic and chronic inflammatory disorders. There is an urgent need to better understand these diseases in the context of mucosal and skin microbiota. This Review discusses the mechanisms of how the microbiota contributes to the predisposition, initiation and perpetuation of immune-mediated diseases in the context of a genetically prone host. It is timely owing to the wealth of new studies that recently contributed to this field, ranging from metagenomic studies in humans and mechanistic studies of host-microorganism interactions in gnotobiotic models and in vitro systems, to molecular mechanisms with broader implications across immune-mediated diseases. We focus on the general principles, such as breaches in immune tolerance and barriers, leading to the promotion of immune-mediated diseases by gut, oral and skin microbiota. Lastly, the therapeutic avenues that either target the microbiota, the barrier surfaces or the host immune system to restore tolerance and homeostasis will be explored.
Collapse
Affiliation(s)
- William E Ruff
- Department of Immunobiology, Yale School of Medicine, New Haven, CT, USA
- Department of Neurology, Yale School of Medicine, New Haven, CT, USA
| | - Teri M Greiling
- Department of Dermatology, Oregon Health & Science University, Portland, OR, USA
| | - Martin A Kriegel
- Department of Immunobiology, Yale School of Medicine, New Haven, CT, USA.
| |
Collapse
|
155
|
Lui PP, Cho I, Ali N. Tissue regulatory T cells. Immunology 2020; 161:4-17. [PMID: 32463116 PMCID: PMC7450170 DOI: 10.1111/imm.13208] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2019] [Revised: 05/01/2020] [Accepted: 05/12/2020] [Indexed: 12/21/2022] Open
Abstract
Foxp3+ CD4+ regulatory T cells (Tregs) are an immune cell lineage endowed with immunosuppressive functionality in a wide array of contexts, including both anti-pathogenic and anti-self responses. In the past decades, our understanding of the functional diversity of circulating or lymphoid Tregs has grown exponentially. Only recently, the importance of Tregs residing within non-lymphoid tissues, such as visceral adipose tissue, muscle, skin and intestine, has been recognized. Not only are Tregs critical for influencing the kinetics and strength of immune responses, but the regulation of non-immune or parenchymal cells, also fall within the purview of tissue-resident or infiltrating Tregs. This review focuses on providing a systematic and comprehensive comparison of the molecular maintenance, local adaptation and functional specializations of Treg populations operating within different tissues.
Collapse
Affiliation(s)
- Prudence PokWai Lui
- Centre for Stem Cells and Regenerative MedicineSchool of Basic and Biomedical SciencesKing's College LondonLondonUK
| | - Inchul Cho
- Centre for Stem Cells and Regenerative MedicineSchool of Basic and Biomedical SciencesKing's College LondonLondonUK
| | - Niwa Ali
- Centre for Stem Cells and Regenerative MedicineSchool of Basic and Biomedical SciencesKing's College LondonLondonUK
- The Francis Crick InstituteLondonUK
| |
Collapse
|
156
|
A Complex Acetate-ment: Timing of Exposure Determines Memory T Cell Fate. Cell Metab 2020; 32:325-327. [PMID: 32877687 DOI: 10.1016/j.cmet.2020.08.010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
In this issue of Cell Metabolism, Balmer et al. show that the timing and concentration of acetate exposure is critical to how it is metabolized by and affects the function of CD8 T cells. When abundantly present at the time of reactivation, acetate rewires CD8 T cell metabolism to suppress their reactivation and limit inflammation.
Collapse
|
157
|
Abstract
The skin microbiota is intimately coupled with cutaneous health and disease. Interactions between commensal microbiota and the multiple cell types involved in cutaneous wound healing regulate the immune response and promote barrier restoration. This dialog between host cells and the microbiome is dysregulated in chronic wounds. In this review, we first describe how advances in sequencing approaches and analysis have been used to study the chronic wound microbiota, and how these findings underscored the complexity of microbial communities and their association with clinical outcomes in patients with chronic wound disorders. We also discuss the mechanistic insights gathered from multiple animal models of polymicrobial wound infections. In addition to the well-described role of bacteria residing in polymicrobial biofilms, we also discuss the role of the intracellular bacterial niche in wound healing. We describe how, in contrast to pathogenic species capable of subverting skin immunity, commensals are essential for the regulation of the cutaneous immune system and provide protection from intracellular pathogens through modulation of the antimicrobial molecule, Perforin-2. Despite recent advances, more research is needed to shed light on host-microbiome crosstalk in both healing and nonhealing chronic wounds to appropriately guide therapeutic developments.
Collapse
Affiliation(s)
- Marjana Tomic-Canic
- Dr. Phillip Frost Department of Dermatology and Cutaneous Surgery, Wound Healing and Regenerative Medicine Research Program, University of Miami, Miller School of Medicine, 1600 NW 10th Ave RMSB R-6056, Miami, FL, 33136, USA
| | - Jamie L Burgess
- Dr. Phillip Frost Department of Dermatology and Cutaneous Surgery, Wound Healing and Regenerative Medicine Research Program, University of Miami, Miller School of Medicine, 1600 NW 10th Ave RMSB R-6056, Miami, FL, 33136, USA
| | - Katelyn E O'Neill
- Department of Microbiology and Immunology, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Natasa Strbo
- Department of Microbiology and Immunology, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Irena Pastar
- Dr. Phillip Frost Department of Dermatology and Cutaneous Surgery, Wound Healing and Regenerative Medicine Research Program, University of Miami, Miller School of Medicine, 1600 NW 10th Ave RMSB R-6056, Miami, FL, 33136, USA.
| |
Collapse
|
158
|
Lückel C, Picard FSR, Huber M. Tc17 biology and function: Novel concepts. Eur J Immunol 2020; 50:1257-1267. [DOI: 10.1002/eji.202048627] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2020] [Revised: 07/02/2020] [Accepted: 07/17/2020] [Indexed: 12/13/2022]
Affiliation(s)
- Christina Lückel
- Institute for ImmunologyUniversity Medical Center of the Johannes Gutenberg‐University Mainz Mainz Germany
| | - Felix. S. R. Picard
- Institute for Medical Microbiology and Hospital HygieneUniversity of Marburg Marburg Germany
| | - Magdalena Huber
- Institute for Medical Microbiology and Hospital HygieneUniversity of Marburg Marburg Germany
| |
Collapse
|
159
|
Castro Rocha FA, Duarte-Monteiro AM, Henrique da Mota LM, Matias Dinelly Pinto AC, Fonseca JE. Microbes, helminths, and rheumatic diseases. Best Pract Res Clin Rheumatol 2020; 34:101528. [PMID: 32448639 PMCID: PMC7203059 DOI: 10.1016/j.berh.2020.101528] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
There has been a progressive interest on modifications of the human defense system following insults occurring in the interface between our body and the external environment, as they may provoke or worsen disease states. Studies suggest that billions of germs, which compose the gut microbiota influence one's innate and adaptive immune responses at the intestinal level, but these microorganisms may also impact rheumatic diseases. The microbiota of the skin, respiratory, and urinary tracts may also be relevant in rheumatology. Evidence indicates that changes in the gut microbiome alter the pathogenesis of immune-mediated diseases such as rheumatoid arthritis and ankylosing spondylitis but also of other disorders like atherosclerosis and osteoarthritis. Therapeutic strategies to modify the microbiota, including probiotics and fecal microbiota transplantation, have been received with skepticism, which, in turn, has drawn attention back to previously developed interventions such as antibiotics. Helminths adapted to humans over the evolution process, but their role in disease modulation, particularly immune-mediated diseases, remains to be understood. The present review focuses on data concerning modifications of the immune system induced by interactions with microbes and pluricellular organisms, namely helminths, and their impact on rheumatic diseases. Practical aspects, including specific microbiota-targeted therapies, are also discussed.
Collapse
Affiliation(s)
- Francisco Airton Castro Rocha
- Departamento de Medicina Clínica, Liga de Reumatologia e Doenças Autoimunes, Faculdade de Medicina, Universidade Federal do Ceará, Fortaleza, CE, Brazil.
| | - Ana Margarida Duarte-Monteiro
- Serviço de Reumatologia e Doenças Ósseas Metabólicas, Hospital de Santa Maria, CHULN and Instituto de Medicina Molecular, Faculdade de Medicina, Universidade de Lisboa, Centro Académico de Medicina de Lisboa, Lisboa, Portugal
| | - Licia Maria Henrique da Mota
- Hospital Universitário de Brasília, Programa de Pós-graduação em Ciências Médicas, Faculdade de Medicina, Universidade de Brasília, Brazil
| | - Ana Carolina Matias Dinelly Pinto
- Departamento de Medicina Clínica, Liga de Reumatologia e Doenças Autoimunes, Faculdade de Medicina, Universidade Federal do Ceará, Fortaleza, CE, Brazil
| | - João Eurico Fonseca
- Serviço de Reumatologia e Doenças Ósseas Metabólicas, Hospital de Santa Maria, CHULN and Instituto de Medicina Molecular, Faculdade de Medicina, Universidade de Lisboa, Centro Académico de Medicina de Lisboa, Lisboa, Portugal
| |
Collapse
|
160
|
|
161
|
Di Domizio J, Belkhodja C, Chenuet P, Fries A, Murray T, Mondéjar PM, Demaria O, Conrad C, Homey B, Werner S, Speiser DE, Ryffel B, Gilliet M. The commensal skin microbiota triggers type I IFN-dependent innate repair responses in injured skin. Nat Immunol 2020; 21:1034-1045. [PMID: 32661363 DOI: 10.1038/s41590-020-0721-6] [Citation(s) in RCA: 118] [Impact Index Per Article: 23.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2019] [Accepted: 05/27/2020] [Indexed: 01/25/2023]
Abstract
Skin wounds heal by coordinated induction of inflammation and tissue repair, but the initiating events are poorly defined. Here we uncover a fundamental role of commensal skin microbiota in this process and show that it is mediated by the recruitment and the activation of type I interferon (IFN)-producing plasmacytoid DC (pDC). Commensal bacteria colonizing skin wounds trigger activation of neutrophils to express the chemokine CXCL10, which recruits pDC and acts as an antimicrobial protein to kill exposed microbiota, leading to the formation of CXCL10-bacterial DNA complexes. These complexes and not complexes with host-derived DNA activate pDC to produce type I IFNs, which accelerate wound closure by triggering skin inflammation and early T cell-independent wound repair responses, mediated by macrophages and fibroblasts that produce major growth factors required for healing. These findings identify a key function of commensal microbiota in driving a central innate wound healing response of the skin.
Collapse
Affiliation(s)
- Jeremy Di Domizio
- Department of Dermatology, CHUV University Hospital, University of Lausanne, Lausanne, Switzerland
| | - Cyrine Belkhodja
- Department of Dermatology, CHUV University Hospital, University of Lausanne, Lausanne, Switzerland
| | - Pauline Chenuet
- Laboratory of Experimental and Molecular Immunology and Neurogenetics, UMR 7355 CNRS-University of Orleans, Orleans, France
| | - Anissa Fries
- Department of Dermatology, CHUV University Hospital, University of Lausanne, Lausanne, Switzerland
| | - Timothy Murray
- Department of Oncology, CHUV University Hospital, University of Lausanne, Lausanne, Switzerland
| | - Paula Marcos Mondéjar
- Department of Oncology, CHUV University Hospital, University of Lausanne, Lausanne, Switzerland
| | - Olivier Demaria
- Department of Dermatology, CHUV University Hospital, University of Lausanne, Lausanne, Switzerland
| | - Curdin Conrad
- Department of Dermatology, CHUV University Hospital, University of Lausanne, Lausanne, Switzerland
| | - Bernhard Homey
- Department of Dermatology, Medical Faculty, Heinrich-Heine-University, Duesseldorf, Germany
| | - Sabine Werner
- ETH Zurich, Institute of Molecular Health Sciences, Zurich, Switzerland
| | - Daniel E Speiser
- Department of Oncology, CHUV University Hospital, University of Lausanne, Lausanne, Switzerland
| | - Bernhard Ryffel
- Laboratory of Experimental and Molecular Immunology and Neurogenetics, UMR 7355 CNRS-University of Orleans, Orleans, France.,Division of Immunology, Department of Pathology, Institute of Infectious Disease and Molecular Medicine, Health Sciences Faculty, University of Cape Town, and South Africa Medical Research Council, Cape Town, South Africa
| | - Michel Gilliet
- Department of Dermatology, CHUV University Hospital, University of Lausanne, Lausanne, Switzerland.
| |
Collapse
|
162
|
Affiliation(s)
- Oliver Harrison
- Center for Fundamental Immunology, Benaroya Research Institute, Seattle, WA, USA.
| |
Collapse
|
163
|
Patra V, Gallais Sérézal I, Wolf P. Potential of Skin Microbiome, Pro- and/or Pre-Biotics to Affect Local Cutaneous Responses to UV Exposure. Nutrients 2020; 12:E1795. [PMID: 32560310 PMCID: PMC7353315 DOI: 10.3390/nu12061795] [Citation(s) in RCA: 38] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2020] [Revised: 06/14/2020] [Accepted: 06/15/2020] [Indexed: 12/12/2022] Open
Abstract
The human skin hosts innumerable microorganisms and maintains homeostasis with the local immune system despite the challenges offered by environmental factors such as ultraviolet radiation (UVR). UVR causes cutaneous alterations such as acute (i.e., sunburn) and chronic inflammation, tanning, photoaging, skin cancer, and immune modulation. Phototherapy on the other hand is widely used to treat inflammatory skin diseases such as psoriasis, atopic dermatitis, polymorphic light eruption and graft-versus-host disease (GvHD), as well as neoplastic skin diseases such as cutaneous T cell lymphoma, among others. Previous work has addressed the use of pro- and pre-biotics to protect against UVR through anti-oxidative, anti-inflammatory, anti-aging, anti-carcinogenic and/or pro-and contra-melanogenic properties. Herein, we discuss and share perspectives of the potential benefits of novel treatment strategies using microbes and pro- and pre-biotics as modulators of the skin response to UVR, and how they could act both for protection against UVR-induced skin damage and as enhancers of the UVR-driven therapeutic effects on the skin.
Collapse
Affiliation(s)
- VijayKumar Patra
- Center for Medical Research, Medical University of Graz, 8010 Graz, Austria;
- Research Unit for Photodermatology, Department of Dermatology, Medical University of Graz, 8010 Graz, Austria
| | - Irène Gallais Sérézal
- Department of Medicine, Unit of Rheumatology, Karolinska Institutet, 171 77 Solna, Sweden;
- Department of Dermatology, Besançon University Hospital, 25000 Besancon, France
| | - Peter Wolf
- Research Unit for Photodermatology, Department of Dermatology, Medical University of Graz, 8010 Graz, Austria
| |
Collapse
|
164
|
The role of the changing human microbiome in the asthma pandemic. J Allergy Clin Immunol 2020; 144:1457-1466. [PMID: 31812180 DOI: 10.1016/j.jaci.2019.10.022] [Citation(s) in RCA: 33] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2019] [Revised: 10/21/2019] [Accepted: 10/23/2019] [Indexed: 02/06/2023]
Abstract
Asthma and allergy incidence continue to increase globally. We have made significant strides in treating disease, but it is becoming more apparent that we need to advance our knowledge into the origins of asthmatic disease. Much recent work has indicated that microbiome composition influences immune regulation and that multiple health care factors have driven a loss in microbiome diversity in modern human populations. Evidence is growing of microbiota-driven influences on immune development, asthma susceptibility, and asthma pathogenesis. The focus of this review is to highlight the strides the field has made in characterizing the constituents of the human gastrointestinal microbiota, such as Helicobacter pylori, other members of the neonatal intestinal microbiota, and microbial peptides and metabolites that influence host immunity and immune response to allergens. As we delve further into this field of research, the goal will be to find actionable and clinical interventions to identify at-risk populations earlier to prevent disease onset. Manipulation of the host microbial community during infancy might be an especially promising approach.
Collapse
|
165
|
Arif S, Gomez-Tourino I, Kamra Y, Pujol-Autonell I, Hanton E, Tree T, Melandri D, Hull C, Wherrett DK, Beam C, Roep BO, Lorenc A, Peakman M. GAD-alum immunotherapy in type 1 diabetes expands bifunctional Th1/Th2 autoreactive CD4 T cells. Diabetologia 2020; 63:1186-1198. [PMID: 32248243 PMCID: PMC7228993 DOI: 10.1007/s00125-020-05130-7] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/22/2020] [Accepted: 02/18/2020] [Indexed: 01/13/2023]
Abstract
AIMS/HYPOTHESIS Antigen-specific therapy aims to modify inflammatory T cell responses in type 1 diabetes and restore immune tolerance. One strategy employs GAD65 conjugated to aluminium hydroxide (GAD-alum) to take advantage of the T helper (Th)2-biasing adjuvant properties of alum and thereby regulate pathological Th1 autoimmunity. We explored the cellular and molecular mechanism of GAD-alum action in the setting of a previously reported randomised placebo-controlled clinical trial conducted by Type 1 Diabetes TrialNet. METHODS In the clinical trial conducted by Type 1 Diabetes TrialNet, participants were immunised with 20 μg GAD-alum (twice or three times) or alum alone and peripheral blood mononuclear cell samples were banked at baseline and post treatment. In the present study, GAD-specific T cell responses were measured in these samples and GAD-specific T cell lines and clones were generated, which were then further characterised. RESULTS At day 91 post immunisation, we detected GAD-specific IL-13+ CD4 T cell responses significantly more frequently in participants immunised with GAD-alum (71% and 94% treated twice or three times, respectively) compared with those immunised with alum alone (38%; p = 0.003 and p = 0.0002, respectively) accompanied by high secreted levels of IL-13, IL-4 and IL-5, confirming a GAD-specific, GAD-alum-induced Th2 response. Of note, GAD-specific, IL-13+ CD4 T cells observed after immunisation co-secreted IFN-γ, displaying a bifunctional Th1/Th2 phenotype. Single-cell transcriptome analysis identified IL13 and IFNG expression in concert with the canonical Th2 and Th1 transcription factor genes GATA3 and TBX21, respectively. T cell receptor β-chain (TCRB) CDR3 regions of GAD-specific bifunctional T cells were identified in circulating naive and central memory CD4 T cell pools of non-immunised participants with new-onset type 1 diabetes and healthy individuals, suggesting the potential for bifunctional responses to be generated de novo by GAD-alum immunisation or via expansion from an existing public repertoire. CONCLUSIONS/INTERPRETATION GAD-alum immunisation activates and propagates GAD-specific CD4 T cells with a distinctive bifunctional phenotype, the functional analysis of which might be important in understanding therapeutic responses.
Collapse
Affiliation(s)
- Sefina Arif
- Peter Gorer Department of Immunobiology, King's College London Faculty of Life Sciences and Medicine, 2nd Floor, Borough Wing, Guy's Hospital, London, SE1 9RT, UK
| | - Iria Gomez-Tourino
- Peter Gorer Department of Immunobiology, King's College London Faculty of Life Sciences and Medicine, 2nd Floor, Borough Wing, Guy's Hospital, London, SE1 9RT, UK
| | - Yogesh Kamra
- Peter Gorer Department of Immunobiology, King's College London Faculty of Life Sciences and Medicine, 2nd Floor, Borough Wing, Guy's Hospital, London, SE1 9RT, UK
| | - Irma Pujol-Autonell
- Peter Gorer Department of Immunobiology, King's College London Faculty of Life Sciences and Medicine, 2nd Floor, Borough Wing, Guy's Hospital, London, SE1 9RT, UK
| | - Emily Hanton
- Peter Gorer Department of Immunobiology, King's College London Faculty of Life Sciences and Medicine, 2nd Floor, Borough Wing, Guy's Hospital, London, SE1 9RT, UK
| | - Timothy Tree
- Peter Gorer Department of Immunobiology, King's College London Faculty of Life Sciences and Medicine, 2nd Floor, Borough Wing, Guy's Hospital, London, SE1 9RT, UK
| | - Daisy Melandri
- Peter Gorer Department of Immunobiology, King's College London Faculty of Life Sciences and Medicine, 2nd Floor, Borough Wing, Guy's Hospital, London, SE1 9RT, UK
| | - Caroline Hull
- Peter Gorer Department of Immunobiology, King's College London Faculty of Life Sciences and Medicine, 2nd Floor, Borough Wing, Guy's Hospital, London, SE1 9RT, UK
| | - Diane K Wherrett
- Division of Endocrinology, Department of Pediatrics, The Hospital for Sick Children, University of Toronto, Toronto, ON, Canada
| | - Craig Beam
- Homer Stryker MD School of Medicine, Western Michigan University, Kalamazoo, MI, USA
| | - Bart O Roep
- Diabetes and Metabolism Research Institute, City of Hope, Duarte, CA, USA
| | - Anna Lorenc
- Peter Gorer Department of Immunobiology, King's College London Faculty of Life Sciences and Medicine, 2nd Floor, Borough Wing, Guy's Hospital, London, SE1 9RT, UK
| | - Mark Peakman
- Peter Gorer Department of Immunobiology, King's College London Faculty of Life Sciences and Medicine, 2nd Floor, Borough Wing, Guy's Hospital, London, SE1 9RT, UK.
- King's Health Partners Institute of Diabetes, Endocrinology and Obesity, King's College Hospital NHS Foundation Trust, London, UK.
| |
Collapse
|
166
|
Bockerstett KA, Lewis SA, Wolf KJ, Noto CN, Jackson NM, Ford EL, Ahn TH, DiPaolo RJ. Single-cell transcriptional analyses of spasmolytic polypeptide-expressing metaplasia arising from acute drug injury and chronic inflammation in the stomach. Gut 2020; 69:1027-1038. [PMID: 31481545 PMCID: PMC7282188 DOI: 10.1136/gutjnl-2019-318930] [Citation(s) in RCA: 52] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/18/2019] [Revised: 08/21/2019] [Accepted: 08/25/2019] [Indexed: 12/24/2022]
Abstract
OBJECTIVE Spasmolytic polypeptide-expressing metaplasia (SPEM) is a regenerative lesion in the gastric mucosa and is a potential precursor to intestinal metaplasia/gastric adenocarcinoma in a chronic inflammatory setting. The goal of these studies was to define the transcriptional changes associated with SPEM at the individual cell level in response to acute drug injury and chronic inflammatory damage in the gastric mucosa. DESIGN Epithelial cells were isolated from the gastric corpus of healthy stomachs and stomachs with drug-induced and inflammation-induced SPEM lesions. Single cell RNA sequencing (scRNA-seq) was performed on tissue samples from each of these settings. The transcriptomes of individual epithelial cells from healthy, acutely damaged and chronically inflamed stomachs were analysed and compared. RESULTS scRNA-seq revealed a population Mucin 6 (Muc6)+gastric intrinsic factor (Gif)+ cells in healthy tissue, but these cells did not express transcripts associated with SPEM. Furthermore, analyses of SPEM cells from drug injured and chronically inflamed corpus yielded two major findings: (1) SPEM and neck cell hyperplasia/hypertrophy are nearly identical in the expression of SPEM-associated transcripts and (2) SPEM programmes induced by drug-mediated parietal cell ablation and chronic inflammation are nearly identical, although the induction of transcripts involved in immunomodulation was unique to SPEM cells in the chronic inflammatory setting. CONCLUSIONS These data necessitate an expansion of the definition of SPEM to include Tff2+Muc6+ cells that do not express mature chief cell transcripts such as Gif. Our data demonstrate that SPEM arises by a highly conserved cellular programme independent of aetiology and develops immunoregulatory capabilities in a setting of chronic inflammation.
Collapse
Affiliation(s)
- Kevin A Bockerstett
- Department of Molecular Microbiology and Immunology, Saint Louis University School of Medicine, St. Louis, Missouri, USA
| | - Scott A Lewis
- Department of Computer Science, Saint Louis University, Saint Louis, Missouri, USA
| | - Kyle J Wolf
- Department of Molecular Microbiology and Immunology, Saint Louis University School of Medicine, St. Louis, Missouri, USA
| | - Christine N Noto
- Department of Molecular Microbiology and Immunology, Saint Louis University School of Medicine, St. Louis, Missouri, USA
| | - Nicholas M Jackson
- Department of Molecular Microbiology and Immunology, Saint Louis University School of Medicine, St. Louis, Missouri, USA
| | - Eric L Ford
- Department of Molecular Microbiology and Immunology, Saint Louis University School of Medicine, St. Louis, Missouri, USA
| | - Tae-Hyuk Ahn
- Department of Computer Science, Saint Louis University, Saint Louis, Missouri, USA
| | - Richard J DiPaolo
- Department of Molecular Microbiology and Immunology, Saint Louis University School of Medicine, St. Louis, Missouri, USA
| |
Collapse
|
167
|
Berzins SP, Wallace ME, Kannourakis G, Kelly J. A Role for MAIT Cells in Colorectal Cancer. Front Immunol 2020; 11:949. [PMID: 32508830 PMCID: PMC7251153 DOI: 10.3389/fimmu.2020.00949] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2020] [Accepted: 04/22/2020] [Indexed: 12/14/2022] Open
Abstract
MAIT cells are MR1-restricted T cells that are well-known for their anti-microbial properties, but they have recently been associated with different forms of cancer. Several studies have reported activated MAIT cells within the microenvironment of colorectal tumors, but there is conjecture about the nature of their response and whether they are contributing to anti-tumor immunity, or to the progression of the disease. We have reviewed the current state of knowledge about the role of MAIT cells in colorectal cancer, including their likely influence when activated and potential sources of stimulation in the tumor microenvironment. The prospects for MAIT cells being used in clinical settings as biomarkers or as targets of new immunotherapies designed to harness their function are discussed.
Collapse
Affiliation(s)
- Stuart P Berzins
- Fiona Elsey Cancer Research Institute, Ballarat, VIC, Australia.,Federation University Australia, Mount Helen, VIC, Australia.,Department of Microbiology and Immunology, Peter Doherty Institute for Infection and Immunity, University of Melbourne, Melbourne, VIC, Australia
| | - Morgan E Wallace
- Fiona Elsey Cancer Research Institute, Ballarat, VIC, Australia.,Federation University Australia, Mount Helen, VIC, Australia
| | - George Kannourakis
- Fiona Elsey Cancer Research Institute, Ballarat, VIC, Australia.,Federation University Australia, Mount Helen, VIC, Australia
| | - Jason Kelly
- Fiona Elsey Cancer Research Institute, Ballarat, VIC, Australia.,Federation University Australia, Mount Helen, VIC, Australia
| |
Collapse
|
168
|
Type 1 T reg cells promote the generation of CD8 + tissue-resident memory T cells. Nat Immunol 2020; 21:766-776. [PMID: 32424367 DOI: 10.1038/s41590-020-0674-9] [Citation(s) in RCA: 72] [Impact Index Per Article: 14.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2019] [Accepted: 03/30/2020] [Indexed: 12/22/2022]
Abstract
Tissue-resident memory T (TRM) cells, functionally distinct from circulating memory T cells, have a critical role in protective immunity in tissues, are more efficacious when elicited after vaccination and yield more effective antitumor immunity, yet the signals that direct development of TRM cells are incompletely understood. Here we show that type 1 regulatory T (Treg) cells, which express the transcription factor T-bet, promote the generation of CD8+ TRM cells. The absence of T-bet-expressing type 1 Treg cells reduces the presence of TRM cells in multiple tissues and increases pathogen burden upon infectious challenge. Using infection models, we show that type 1 Treg cells are specifically recruited to local inflammatory sites via the chemokine receptor CXCR3. Close proximity with effector CD8+ T cells and Treg cell expression of integrin-β8 endows the bioavailability of transforming growth factor-β in the microenvironment, thereby promoting the generation of CD8+ TRM cells.
Collapse
|
169
|
Amezcua Vesely MC, Pallis P, Bielecki P, Low JS, Zhao J, Harman CCD, Kroehling L, Jackson R, Bailis W, Licona-Limón P, Xu H, Iijima N, Pillai PS, Kaplan DH, Weaver CT, Kluger Y, Kowalczyk MS, Iwasaki A, Pereira JP, Esplugues E, Gagliani N, Flavell RA. Effector T H17 Cells Give Rise to Long-Lived T RM Cells that Are Essential for an Immediate Response against Bacterial Infection. Cell 2020; 178:1176-1188.e15. [PMID: 31442406 DOI: 10.1016/j.cell.2019.07.032] [Citation(s) in RCA: 120] [Impact Index Per Article: 24.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2018] [Revised: 03/19/2019] [Accepted: 07/17/2019] [Indexed: 12/20/2022]
Abstract
Adaptive immunity provides life-long protection by generating central and effector memory T cells and the most recently described tissue resident memory T (TRM) cells. However, the cellular origin of CD4 TRM cells and their contribution to host defense remain elusive. Using IL-17A tracking-fate mouse models, we found that a significant fraction of lung CD4 TRM cells derive from IL-17A-producing effector (TH17) cells following immunization with heat-killed Klebsiella pneumonia (Kp). These exTH17 TRM cells are maintained in the lung by IL-7, produced by lymphatic endothelial cells. During a memory response, neither antibodies, γδ T cells, nor circulatory T cells are sufficient for the rapid host defense required to eliminate Kp. Conversely, using parabiosis and depletion studies, we demonstrated that exTH17 TRM cells play an important role in bacterial clearance. Thus, we delineate the origin and function of airway CD4 TRM cells during bacterial infection, offering novel strategies for targeted vaccine design.
Collapse
Affiliation(s)
- Maria Carolina Amezcua Vesely
- Department of Immunobiology, School of Medicine, Yale University, New Haven, CT, USA; Howard Hughes Medical Institute, Yale University School of Medicine, New Haven, CT, USA
| | - Paris Pallis
- Department of Immunobiology, School of Medicine, Yale University, New Haven, CT, USA
| | - Piotr Bielecki
- Department of Immunobiology, School of Medicine, Yale University, New Haven, CT, USA
| | - Jun Siong Low
- Department of Immunobiology, School of Medicine, Yale University, New Haven, CT, USA
| | - Jun Zhao
- Department of Immunobiology, School of Medicine, Yale University, New Haven, CT, USA; Department of Pathology, Yale School of Medicine, New Haven, CT 06510, USA; Program of Computational Biology and Bioinformatics, Yale University, New Haven, CT 06520, USA
| | - Christian C D Harman
- Department of Immunobiology, School of Medicine, Yale University, New Haven, CT, USA; Howard Hughes Medical Institute, Yale University School of Medicine, New Haven, CT, USA
| | - Lina Kroehling
- Department of Immunobiology, School of Medicine, Yale University, New Haven, CT, USA
| | - Ruaidhrí Jackson
- Department of Immunobiology, School of Medicine, Yale University, New Haven, CT, USA
| | - Will Bailis
- Department of Immunobiology, School of Medicine, Yale University, New Haven, CT, USA
| | - Paula Licona-Limón
- Departamento de Biología Celular y del Desarrollo, Instituto de Fisiología Celular, Universidad Nacional Autónoma de México, Ciudad de México, México
| | - Hao Xu
- Department of Immunobiology, School of Medicine, Yale University, New Haven, CT, USA
| | - Norifumi Iijima
- Department of Immunobiology, School of Medicine, Yale University, New Haven, CT, USA
| | - Padmini S Pillai
- Department of Immunobiology, School of Medicine, Yale University, New Haven, CT, USA; Koch Institute for Integrative Cancer Research and Department of Chemical Engineering, Massachusetts Institute of Technology, Cambridge, MA 02142, USA
| | - Daniel H Kaplan
- Department of Dermatology and Immunology, University of Pittsburgh, Pittsburgh, PA 15261, USA
| | - Casey T Weaver
- Departments of Pathology, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | - Yuval Kluger
- Department of Pathology, Yale School of Medicine, New Haven, CT 06510, USA; Program of Computational Biology and Bioinformatics, Yale University, New Haven, CT 06520, USA; Applied Mathematics Program, Yale University, New Haven, CT 06511, USA
| | | | - Akiko Iwasaki
- Department of Immunobiology, School of Medicine, Yale University, New Haven, CT, USA; Howard Hughes Medical Institute, Yale University School of Medicine, New Haven, CT, USA
| | - Joao P Pereira
- Department of Immunobiology, School of Medicine, Yale University, New Haven, CT, USA
| | - Enric Esplugues
- Department of Immunobiology, School of Medicine, Yale University, New Haven, CT, USA; Laboratory of Molecular and Cellular Immunology, Principe Felipe Research Center (CIPF), 46012 Valencia, Spain
| | - Nicola Gagliani
- Department of Immunobiology, School of Medicine, Yale University, New Haven, CT, USA; I. Medical Department and Department of General, Visceral and Thoracic Surgery, University Medical Center Hamburg-Eppendorf 20246 Hamburg, Germany; Immunology and Allergy Unit, Department of Medicine Solna, Karolinska Institute, 17176 Stockholm, Sweden.
| | - Richard A Flavell
- Department of Immunobiology, School of Medicine, Yale University, New Haven, CT, USA; Howard Hughes Medical Institute, Yale University School of Medicine, New Haven, CT, USA.
| |
Collapse
|
170
|
Künzli M, King CG. Resident Memory T Cells Escape 'Home Quarantine'. Trends Immunol 2020; 41:454-456. [PMID: 32386961 DOI: 10.1016/j.it.2020.04.010] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2020] [Accepted: 04/23/2020] [Indexed: 12/16/2022]
Abstract
T resident memory (Trm) cells provide a first line of defense in non-lymphoid tissues. A new report in Nature Immunology by Fonseca et al. reveals that CD8+ Trm cells can give rise to circulating effector and memory T cells, but remain predisposed to migrate back into their tissue of origin.
Collapse
Affiliation(s)
- Marco Künzli
- Department of Biomedicine, University of Basel, University Hospital of Basel, CH-4031 Basel, Switzerland
| | - Carolyn G King
- Department of Biomedicine, University of Basel, University Hospital of Basel, CH-4031 Basel, Switzerland.
| |
Collapse
|
171
|
Ronchese F, Hilligan KL, Mayer JU. Dendritic cells and the skin environment. Curr Opin Immunol 2020; 64:56-62. [PMID: 32387901 DOI: 10.1016/j.coi.2020.03.006] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2019] [Revised: 03/01/2020] [Accepted: 03/10/2020] [Indexed: 12/11/2022]
Abstract
The skin is inhabited by several immune cell populations that serve as a first line of defence against pathogen invasion. Amongst these populations are dendritic cells, which play an essential sentinel function by taking up antigen or infectious agents and transporting them to the lymph node for T cell recognition and the priming of immune responses. In this review, we briefly summarise recent advances showing how skin dendritic cells are connected to a network of epithelial and stromal cells, which provide structural support, growth factors, spatial cues, contact with the external environment and the skin microbiome, and favour interactions with other immune cells. We propose that this network creates a unique skin environment that may condition dendritic cell phenotype and function.
Collapse
Affiliation(s)
- Franca Ronchese
- Malaghan Institute of Medical Research, Wellington 6012, New Zealand.
| | - Kerry L Hilligan
- Malaghan Institute of Medical Research, Wellington 6012, New Zealand; Immunobiology Section, Laboratory of Parasitic Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda 20892, Maryland, USA
| | - Johannes U Mayer
- Malaghan Institute of Medical Research, Wellington 6012, New Zealand
| |
Collapse
|
172
|
Boothby IC, Cohen JN, Rosenblum MD. Regulatory T cells in skin injury: At the crossroads of tolerance and tissue repair. Sci Immunol 2020; 5:eaaz9631. [PMID: 32358172 PMCID: PMC7274208 DOI: 10.1126/sciimmunol.aaz9631] [Citation(s) in RCA: 97] [Impact Index Per Article: 19.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2020] [Accepted: 04/08/2020] [Indexed: 12/21/2022]
Abstract
Skin injury is a highly inflammatory process that is carefully regulated to mitigate tissue damage and allow for proper barrier repair. Regulatory T cells (Tregs) are crucial coordinators of the immune response to injury in several organs. Here, we review the emerging role of Tregs in facilitating skin repair after injury. We focus on recently discovered interactions between lymphocytes and nonhematopoietic cells during wound healing and discuss how these interactions are regulated both by "classical" suppressive mechanisms of Tregs and by "nonclassical" reparative Treg functions.
Collapse
Affiliation(s)
- Ian C Boothby
- Department of Dermatology, University of California, San Francisco, 513 Parnassus Avenue, San Francisco, CA 94143, USA
- Medical Scientist Training Program, University of California, San Francisco, 513 Parnassus Avenue, San Francisco, CA 94143, USA
| | - Jarish N Cohen
- Department of Pathology, University of California, San Francisco, 513 Parnassus Avenue, San Francisco, CA 94143, USA
| | - Michael D Rosenblum
- Department of Dermatology, University of California, San Francisco, 513 Parnassus Avenue, San Francisco, CA 94143, USA.
| |
Collapse
|
173
|
Ordovas-Montanes J, Beyaz S, Rakoff-Nahoum S, Shalek AK. Distribution and storage of inflammatory memory in barrier tissues. Nat Rev Immunol 2020; 20:308-320. [PMID: 32015472 PMCID: PMC7547402 DOI: 10.1038/s41577-019-0263-z] [Citation(s) in RCA: 54] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/06/2019] [Indexed: 12/17/2022]
Abstract
Memories of previous immune events enable barrier tissues to rapidly recall distinct environmental exposures. To effectively inform future responses, these past experiences can be stored in cell types that are long-term residents or essential constituents of tissues. There is an emerging understanding that, in addition to antigen-specific immune cells, diverse haematopoietic, stromal, parenchymal and neuronal cell types can store inflammatory memory. Here, we explore the impact of previous immune activity on various cell lineages with the goal of presenting a unified view of inflammatory memory to environmental exposures (such as allergens, antigens, noxious agents and microorganisms) at barrier tissues. We propose that inflammatory memory is distributed across diverse cell types and stored through shifts in cell states, and we provide a framework to guide future experiments. This distribution and storage may promote adaptation or maladaptation in homeostatic, maintenance and disease settings - especially if the distribution of memory favours cellular cooperation during storage or recall.
Collapse
Affiliation(s)
- Jose Ordovas-Montanes
- Division of Gastroenterology, Boston Children's Hospital, Boston, MA, USA.
- Institute for Medical Engineering and Science (IMES), MIT, Cambridge, MA, USA.
- Department of Chemistry, MIT, Cambridge, MA, USA.
- Koch Institute for Integrative Cancer Research, MIT, Cambridge, MA, USA.
- Broad Institute of MIT and Harvard, Cambridge, MA, USA.
- Ragon Institute of MGH, MIT and Harvard, Cambridge, MA, USA.
- Harvard Stem Cell Institute, Cambridge, MA, USA.
| | - Semir Beyaz
- Cold Spring Harbor Laboratory, Cold Spring Harbor, NY, USA
| | - Seth Rakoff-Nahoum
- Division of Gastroenterology, Boston Children's Hospital, Boston, MA, USA
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Division of Infectious Diseases, Boston Children's Hospital, Boston, MA, USA
| | - Alex K Shalek
- Institute for Medical Engineering and Science (IMES), MIT, Cambridge, MA, USA
- Department of Chemistry, MIT, Cambridge, MA, USA
- Koch Institute for Integrative Cancer Research, MIT, Cambridge, MA, USA
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Ragon Institute of MGH, MIT and Harvard, Cambridge, MA, USA
- Harvard Stem Cell Institute, Cambridge, MA, USA
- Harvard-MIT Division of Health Sciences & Technology, Cambridge, MA, USA
| |
Collapse
|
174
|
Sepulveda M, Pirozzolo I, Alegre ML. Impact of the microbiota on solid organ transplant rejection. Curr Opin Organ Transplant 2020; 24:679-686. [PMID: 31577594 DOI: 10.1097/mot.0000000000000702] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
PURPOSE OF REVIEW The microbiota in mammalian hosts can affect maturation and function of the immune system and has been associated with health and disease. We will review new findings on how this dynamic environmental factor impacts alloimmunity and therapy in transplant hosts. RECENT FINDINGS The microbiota changes after transplantation and immunosuppressive therapy. New data indicate that different microbial community structures have distinct impact on graft outcome, from promoting, to inhibiting or being neutral to transplant survival. In addition, we will address reciprocal interactions between the microbiota and immunosuppressive drugs, as well as the suitability of the microbiota as a predictive biomarker and its utility as adjunct therapy in transplantation. SUMMARY Advances in microbiome sequencing and wider availability of gnotobiotic facilities are enabling mechanistic investigations into the commensal communities and pathways that modulate allograft outcome, responsiveness to immunosuppression and side effects of drugs. A better understanding of the functions of the microbiota may help mitigate drug toxicity, predict drug dosage and dampen alloimmunity in transplant patients.
Collapse
|
175
|
Abusleme L, O'Gorman H, Dutzan N, Greenwell-Wild T, Moutsopoulos NM. Establishment and Stability of the Murine Oral Microbiome. J Dent Res 2020; 99:721-729. [PMID: 32345105 DOI: 10.1177/0022034520915485] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023] Open
Abstract
Commensal microbiomes exert critical functions at barrier sites. In particular, establishment of the commensal microbiome after birth dictates immune functionality and tissue homeostasis at mucosal surfaces. To investigate the establishment and stability of the oral mucosal microbiome in mice, we evaluated oral microbiome communities shortly after birth, through adulthood, and up to 1 y of life in a controlled manner, using sequential oral samples from the same mice over time. We further evaluated transmissibility of oral microbiomes from parents and during cohousing experiments and evaluated susceptibility to oral inflammatory disease in mice harboring distinct microbiomes. Our work reveals basic principles in the establishment and stability of a health-associated oral microbiome after birth and provides insights that may be important for host-microbiome experimentation in animal models.
Collapse
Affiliation(s)
- L Abusleme
- Laboratory of Oral Microbiology, Faculty of Dentistry, University of Chile, Santiago, Chile.,Laboratory for Craniofacial Translational Research, Faculty of Dentistry, University of Chile, Santiago, Chile
| | - H O'Gorman
- National Institute of Dental and Craniofacial Research, National Institutes of Health, Bethesda, MD, USA
| | - N Dutzan
- Laboratory of Oral Microbiology, Faculty of Dentistry, University of Chile, Santiago, Chile.,Laboratory for Craniofacial Translational Research, Faculty of Dentistry, University of Chile, Santiago, Chile
| | - T Greenwell-Wild
- National Institute of Dental and Craniofacial Research, National Institutes of Health, Bethesda, MD, USA
| | - N M Moutsopoulos
- National Institute of Dental and Craniofacial Research, National Institutes of Health, Bethesda, MD, USA
| |
Collapse
|
176
|
Alves de Lima K, Rustenhoven J, Kipnis J. Meningeal Immunity and Its Function in Maintenance of the Central Nervous System in Health and Disease. Annu Rev Immunol 2020; 38:597-620. [DOI: 10.1146/annurev-immunol-102319-103410] [Citation(s) in RCA: 109] [Impact Index Per Article: 21.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Neuroimmunology, albeit a relatively established discipline, has recently sparked numerous exciting findings on microglia, the resident macrophages of the central nervous system (CNS). This review addresses meningeal immunity, a less-studied aspect of neuroimmune interactions. The meninges, a triple layer of membranes—the pia mater, arachnoid mater, and dura mater—surround the CNS, encompassing the cerebrospinal fluid produced by the choroid plexus epithelium. Unlike the adjacent brain parenchyma, the meninges contain a wide repertoire of immune cells. These constitute meningeal immunity, which is primarily concerned with immune surveillance of the CNS, and—according to recent evidence—also participates in postinjury CNS recovery, chronic neurodegenerative conditions, and even higher brain function. Meningeal immunity has recently come under the spotlight owing to the characterization of meningeal lymphatic vessels draining the CNS. Here, we review the current state of our understanding of meningeal immunity and its effects on healthy and diseased brains.
Collapse
Affiliation(s)
- Kalil Alves de Lima
- Center for Brain Immunology and Glia (BIG) and Department of Neuroscience, School of Medicine, University of Virginia, Charlottesville, Virginia 22908, USA;,
| | - Justin Rustenhoven
- Center for Brain Immunology and Glia (BIG) and Department of Neuroscience, School of Medicine, University of Virginia, Charlottesville, Virginia 22908, USA;,
| | - Jonathan Kipnis
- Center for Brain Immunology and Glia (BIG) and Department of Neuroscience, School of Medicine, University of Virginia, Charlottesville, Virginia 22908, USA;,
| |
Collapse
|
177
|
Frizzell H, Fonseca R, Christo SN, Evrard M, Cruz-Gomez S, Zanluqui NG, von Scheidt B, Freestone D, Park SL, McWilliam HEG, Villadangos JA, Carbone FR, Mackay LK. Organ-specific isoform selection of fatty acid–binding proteins in tissue-resident lymphocytes. Sci Immunol 2020; 5:5/46/eaay9283. [DOI: 10.1126/sciimmunol.aay9283] [Citation(s) in RCA: 53] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2019] [Accepted: 02/06/2020] [Indexed: 12/16/2022]
Abstract
Tissue-resident memory T (TRM) cells exist throughout the body, where they are poised to mediate local immune responses. Although studies have defined a common mechanism of residency independent of location, there is likely to be a level of specialization that adapts TRM cells to their given tissue of lodgment. It has been shown that TRM cells in the skin rely on the uptake of exogenous fatty acids for their survival and up-regulate fatty acid–binding protein 4 (FABP4) and FABP5 as part of their transcriptional program. However, FABPs exist as a larger family of isoforms, with different members selected in a tissue-specific fashion that is optimized for local fatty acid availability. Here, we show that although TRM cells in a range of tissue widely express FABPs, they are not restricted to FABP4 and FABP5. Instead, TRM cells show varying patterns of isoform usage that are determined by tissue-derived factors. These patterns are malleable because TRM cells relocated to different organs modify their FABP expression in line with their new location. As a consequence, these results argue for tissue-specific overlays to the TRM cell residency program, including FABP expression that is tailored to the particular tissue of TRM cell lodgment.
Collapse
Affiliation(s)
- H. Frizzell
- Department of Microbiology and Immunology, University of Melbourne, Peter Doherty Institute for Infection and Immunity, Melbourne, VIC, Australia
| | - R. Fonseca
- Department of Microbiology and Immunology, University of Melbourne, Peter Doherty Institute for Infection and Immunity, Melbourne, VIC, Australia
| | - S. N. Christo
- Department of Microbiology and Immunology, University of Melbourne, Peter Doherty Institute for Infection and Immunity, Melbourne, VIC, Australia
| | - M. Evrard
- Department of Microbiology and Immunology, University of Melbourne, Peter Doherty Institute for Infection and Immunity, Melbourne, VIC, Australia
| | - S. Cruz-Gomez
- Department of Microbiology and Immunology, University of Melbourne, Peter Doherty Institute for Infection and Immunity, Melbourne, VIC, Australia
| | - N. G. Zanluqui
- Department of Microbiology and Immunology, University of Melbourne, Peter Doherty Institute for Infection and Immunity, Melbourne, VIC, Australia
- Department of Immunology, Institute of Biomedical Sciences, University of São Paulo, São Paulo, SP, Brazil
| | - B. von Scheidt
- Department of Microbiology and Immunology, University of Melbourne, Peter Doherty Institute for Infection and Immunity, Melbourne, VIC, Australia
| | - D. Freestone
- Department of Microbiology and Immunology, University of Melbourne, Peter Doherty Institute for Infection and Immunity, Melbourne, VIC, Australia
| | - S. L. Park
- Department of Microbiology and Immunology, University of Melbourne, Peter Doherty Institute for Infection and Immunity, Melbourne, VIC, Australia
| | - H. E. G. McWilliam
- Department of Microbiology and Immunology, University of Melbourne, Peter Doherty Institute for Infection and Immunity, Melbourne, VIC, Australia
- Department of Biochemistry and Molecular Biology, University of Melbourne, Bio21 Molecular Science and Biotechnology Institute, Melbourne, VIC, Australia
| | - J. A. Villadangos
- Department of Microbiology and Immunology, University of Melbourne, Peter Doherty Institute for Infection and Immunity, Melbourne, VIC, Australia
- Department of Biochemistry and Molecular Biology, University of Melbourne, Bio21 Molecular Science and Biotechnology Institute, Melbourne, VIC, Australia
| | - F. R. Carbone
- Department of Microbiology and Immunology, University of Melbourne, Peter Doherty Institute for Infection and Immunity, Melbourne, VIC, Australia
| | - L. K. Mackay
- Department of Microbiology and Immunology, University of Melbourne, Peter Doherty Institute for Infection and Immunity, Melbourne, VIC, Australia
| |
Collapse
|
178
|
Zemmour D, Kiner E, Benoist C. CD4 + teff cell heterogeneity: the perspective from single-cell transcriptomics. Curr Opin Immunol 2020; 63:61-67. [PMID: 32259715 PMCID: PMC7198319 DOI: 10.1016/j.coi.2020.02.004] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2020] [Accepted: 02/18/2020] [Indexed: 12/15/2022]
Abstract
Single-cell transcriptomics (scRNAseq) holds the promise to generate definitive atlases of cell types. We review scRNAseq studies of conventional CD4+ αβ T cells performed in a variety of challenged contexts (infection, tumor, allergy) that aimed to parse the complexity and representativity of previously defined CD4+ T cell types, lineages, and cosmologies. With a few years' experience, the field has realized the difficulties and pitfalls of scRNAseq. With the very high-dimensionality of scRNAseq data, subset definitions based on low-dimensionality marker combinations tend to fade or blur: cell types prove more complex than expected; transcripts of key defining transcripts (cytokines, chemokines) are distributed as broad and partially overlapping continua; boundaries with innate lymphocytes are blurred. Tissue location and activation, either cytokine-driven or TCR-driven, determine Teff heterogeneity in sometimes unexpected ways. Emerging techniques for lineage and trajectory tracing, and RNA-protein connections, will further help define the space of differentiated CD4+ T cell heterogeneity.
Collapse
Affiliation(s)
- David Zemmour
- Department of Immunology, Harvard Medical School, and Evergrande Center for Immunologic Diseases, Harvard Medical School and Brigham and Women's Hospital, Boston, MA 02115, USA
| | - Evgeny Kiner
- Department of Immunology, Harvard Medical School, and Evergrande Center for Immunologic Diseases, Harvard Medical School and Brigham and Women's Hospital, Boston, MA 02115, USA
| | - Christophe Benoist
- Department of Immunology, Harvard Medical School, and Evergrande Center for Immunologic Diseases, Harvard Medical School and Brigham and Women's Hospital, Boston, MA 02115, USA.
| |
Collapse
|
179
|
Bukhari S, Mertz AF, Naik S. Eavesdropping on the conversation between immune cells and the skin epithelium. Int Immunol 2020; 31:415-422. [PMID: 30721971 DOI: 10.1093/intimm/dxy088] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2018] [Accepted: 01/04/2019] [Indexed: 11/12/2022] Open
Abstract
The skin epithelium covers our body and serves as a vital interface with the external environment. Here, we review the context-specific interactions between immune cells and the epithelium that underlie barrier fitness and function. We highlight the mechanisms by which these two systems engage each other and how immune-epithelial interactions are tuned by microbial and inflammatory stimuli. Epithelial homeostasis relies on a delicate balance of immune surveillance and tolerance, breakdown of which results in disease. In addition to their canonical immune functions, resident and recruited immune cells also supply the epithelium with instructive signals to promote repair. Decoding the dialogue between immunity and the epithelium therefore has great potential for boosting barrier function or mitigating inflammatory epithelial diseases.
Collapse
Affiliation(s)
- Shoiab Bukhari
- Department of Pathology, Department of Medicine and Ronald O. Perelman Department of Dermatology, NYU School of Medicine, New York, NY, USA
| | - Aaron F Mertz
- Laboratory of Mammalian Cell Biology and Development, The Rockefeller University, New York, NY, USA
| | - Shruti Naik
- Department of Pathology, Department of Medicine and Ronald O. Perelman Department of Dermatology, NYU School of Medicine, New York, NY, USA
| |
Collapse
|
180
|
Mansfield K, Naik S. Unraveling Immune-Epithelial Interactions in Skin Homeostasis and Injury. THE YALE JOURNAL OF BIOLOGY AND MEDICINE 2020; 93:133-143. [PMID: 32226343 PMCID: PMC7087067] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
The skin serves as a front line of defense against harmful environmental elements and thus is vital for organismal survival. This barrier is comprised of a water-tight epithelial structure reinforced by an arsenal of immune cells. The epithelial and immune components of the skin are interdependent and actively dialogue to maintain health and combat infectious, injurious, and noxious stimuli. Here, we discuss the molecular mediators of this crosstalk that establish tissue homeostasis and their dynamic adaptations to various stress conditions. In particular, we focus on immune-epithelial interactions in homeostatic tissue regeneration, during natural cycling of the hair follicle, and following skin injury. We also highlight the epithelial derived factors that orchestrate immunity. A comprehensive and mechanistic understanding of dynamic interactions between cutaneous immune cells and the epithelium can be leveraged to develop novel therapies to treat of range of skin diseases and boost skin health.
Collapse
Affiliation(s)
| | - Shruti Naik
- To whom all correspondence should be addressed: Shruti Naik, Department of Pathology, Department of Medicine, and Ronald O. Perelman Department of Dermatology, New York University School of Medicine, New York, NY;
| |
Collapse
|
181
|
Abstract
The terminal differentiation of the epidermis is a complex physiological process. During the past few decades, medical genetics has shown that defects in the stratum corneum (SC) permeability barrier cause a myriad of pathological conditions, ranging from common dry skin to lethal ichthyoses. Contrarily, molecular phylogenetics has revealed that amniotes have acquired a specialized form of cytoprotection cornification that provides mechanical resilience to the SC. This superior biochemical property, along with desiccation tolerance, is attributable to the proper formation of the macromolecular protein-lipid complex termed cornified cell envelopes (CE). Cornification largely depends on the peculiar biochemical and biophysical properties of loricrin, which is a major CE component. Despite its quantitative significance, loricrin knockout (LKO) mice have revealed it to be dispensable for the SC permeability barrier. Nevertheless, LKO mice have brought us valuable lessons. It is also becoming evident that absent loricrin affects skin homeostasis more profoundly in many more aspects than previously expected. Through an extensive review of aggregate evidence, we discuss herein the functional significance of the thiol-rich protein loricrin from a biochemical, genetic, pathological, metabolic, or immunological aspect with some theoretical and speculative perspectives.
Collapse
Affiliation(s)
- Yosuke Ishitsuka
- Department of Dermatology, Faculty of Medicine, University of Tsukuba 1-1-1 Tennodai, Tsukuba, Ibaraki 305-8575, Japan
| | - Dennis R. Roop
- Department of Dermatology and Charles C. Gates Center for Regenerative Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA;
| |
Collapse
|
182
|
Hadian Y, Fregoso D, Nguyen C, Bagood MD, Dahle SE, Gareau MG, Isseroff RR. Microbiome-skin-brain axis: A novel paradigm for cutaneous wounds. Wound Repair Regen 2020; 28:282-292. [PMID: 32034844 DOI: 10.1111/wrr.12800] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2019] [Revised: 01/21/2020] [Accepted: 01/24/2020] [Indexed: 12/12/2022]
Abstract
Chronic wounds cause a significant burden on society financially, medically, and psychologically. Unfortunately, patients with nonhealing wounds often suffer from comorbidities that further compound their disability. Given the high rate of depressive symptoms experienced by patients with chronic wounds, further studies are needed to investigate the potentially linked pathophysiological changes in wounds and depression in order to improve patient care. The English literature on wound healing, inflammatory and microbial changes in chronic wounds and depression, and antiinflammatory and probiotic therapy was reviewed on PubMed. Chronic wound conditions and depression were demonstrated to share common pathologic features of dysregulated inflammation and altered microbiome, indicating a possible relationship. Furthermore, alternative treatment strategies such as immune-targeted and probiotic therapy showed promising potential by addressing both pathophysiological pathways. However, many existing studies are limited to a small study population, a cross-sectional design that does not establish temporality, or a wide range of confounding variables in the context of a highly complex and multifactorial disease process. Therefore, additional preclinical studies in suitable wound models, as well as larger clinical cohort studies and trials are necessary to elucidate the relationship between wound microbiome, healing, and depression, and ultimately guide the most effective therapeutic and management plan for chronic wound patients.
Collapse
Affiliation(s)
- Yasmin Hadian
- Department of Dermatology, School of Medicine, University of California, Davis, California.,Dermatology Section, VA Northern California Health Care System, Mather, California
| | - Daniel Fregoso
- Department of Dermatology, School of Medicine, University of California, Davis, California
| | - Chuong Nguyen
- Department of Dermatology, School of Medicine, University of California, Davis, California
| | - Michelle D Bagood
- Department of Dermatology, School of Medicine, University of California, Davis, California
| | - Sara E Dahle
- Department of Dermatology, School of Medicine, University of California, Davis, California.,Podiatry Section, VA Northern California Health Care System, Mather, California
| | - Melanie G Gareau
- Department of Anatomy, Physiology and Cell Biology, School of Veterinary Medicine, University of California Davis, Davis, California
| | - Roslyn Rivkah Isseroff
- Department of Dermatology, School of Medicine, University of California, Davis, California.,Dermatology Section, VA Northern California Health Care System, Mather, California
| |
Collapse
|
183
|
Liang C, Yang KY, Chan VW, Li X, Fung TH, Wu Y, Tian XY, Huang Y, Qin L, Lau JY, Lui KO. CD8 + T-cell plasticity regulates vascular regeneration in type-2 diabetes. Theranostics 2020; 10:4217-4232. [PMID: 32226549 PMCID: PMC7086373 DOI: 10.7150/thno.40663] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2019] [Accepted: 01/02/2020] [Indexed: 12/13/2022] Open
Abstract
In this study, we observe that the ischemic tissues of type-2 diabetic (T2D) patients and mice have significantly more CD8+ T-cells than that of their normoglycemic counterparts, respectively. However, the role of CD8+ T-cells in the pathogenesis of diabetic vascular complication has been less studied. Methods: We employed loss-of-function studies in mouse models using the non-lytic anti-CD8 antibody that blocks tissue infiltration of CD8+ T-cells into the injured tissue. We also performed genome-wide, single-cell RNA-sequencing of CD8+ T-cells to uncover their role in the pathogenesis of diabetic vascular diseases. Results: The vascular density is negatively correlated with the number of CD8+ T-cells in the ischemic tissues of patients and mice after injury. CD8+ T-cells or their supernatant can directly impair human and murine angiogenesis. Compared to normoglycemic mice that can regenerate their blood vessels after injury, T2D mice fail in this regeneration. Treatment with the CD8 checkpoint blocking antibody increases the proliferation and function of endothelial cells in both Leprdb/db mice and diet-induced diabetic Cdh5-Cre;Rosa-YFP lineage-tracing mice after ischemic injury. Furthermore, single-cell transcriptomic profiling reveals that CD8+ T-cells of T2D mice showed a de novo cell fate change from the angiogenic, tissue-resident memory cells towards the effector and effector memory cells after injury. Functional revascularization by CD8 checkpoint blockade is mediated through unleashing such a poised lineage commitment of CD8+ T-cells from T2D mice. Conclusion: Our results reveal that CD8+ T-cell plasticity regulates vascular regeneration; and give clinically relevant insights into the potential development of immunotherapy targeting vascular diseases associated with obesity and diabetes.
Collapse
|
184
|
Salerno F, Turner M, Wolkers MC. Dynamic Post-Transcriptional Events Governing CD8+ T Cell Homeostasis and Effector Function. Trends Immunol 2020; 41:240-254. [DOI: 10.1016/j.it.2020.01.001] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2019] [Revised: 01/08/2020] [Accepted: 01/08/2020] [Indexed: 12/31/2022]
|
185
|
Pierce SK, Schwartzberg PL, Shah NN, Taylor N. Women in immunology: 2020 and beyond. Nat Immunol 2020; 21:254-258. [PMID: 32094649 PMCID: PMC11549733 DOI: 10.1038/s41590-020-0618-4] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2020] [Accepted: 01/24/2020] [Indexed: 12/27/2022]
Abstract
Women have been at the forefront of tremendous achievements in immunology in the past decade. However, disparities still exist, limiting upward potential and further advancements. As four NIH intramural women scientists who care deeply about scientific progress and the progress of women in our field, we review ongoing challenges and discuss potential approaches to help advance the promotion of women in the sciences.
Collapse
Affiliation(s)
- Susan K Pierce
- Laboratory of Immunogenetics, National Institute of Allergy and Infectious Diseases, NIH, Bethesda, MD, USA.
| | - Pamela L Schwartzberg
- Laboratory of Immune System Biology, National Institute of Allergy and Infectious Diseases, NIH, Bethesda, MD, USA.
| | - Nirali N Shah
- Pediatric Oncology Branch, National Cancer Institute, NIH, Bethesda, MD, USA.
| | - Naomi Taylor
- Pediatric Oncology Branch, National Cancer Institute, NIH, Bethesda, MD, USA.
- IGMM, Université de Montpellier, CNRS, Montpellier, France.
| |
Collapse
|
186
|
Platts-Mills TAE, Commins SP, Biedermann T, van Hage M, Levin M, Beck LA, Diuk-Wasser M, Jappe U, Apostolovic D, Minnicozzi M, Plaut M, Wilson JM. On the cause and consequences of IgE to galactose-α-1,3-galactose: A report from the National Institute of Allergy and Infectious Diseases Workshop on Understanding IgE-Mediated Mammalian Meat Allergy. J Allergy Clin Immunol 2020; 145:1061-1071. [PMID: 32057766 DOI: 10.1016/j.jaci.2020.01.047] [Citation(s) in RCA: 82] [Impact Index Per Article: 16.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2019] [Revised: 01/29/2020] [Accepted: 01/31/2020] [Indexed: 12/16/2022]
Abstract
The mammalian meat allergy known as the "α-Gal syndrome" relates to IgE specific for galactose-α-1,3-galactose (α-Gal), an oligosaccharide that is present in cells and tissues of nonprimate mammals. The recognition of delayed reactions to food derived from mammals in patients with IgE to α-Gal and also the association with tick bites have been increasing worldwide. In 2018, the National Institute of Allergy and Infectious Diseases, Division of Allergy, Immunology and Transplantation, sponsored a workshop on this emerging tick-related disease. International experts from the fields of tick biology, allergy, immunology, infectious disease, and dermatology discussed the current state of our understanding of this emerging medical condition. The participants provided suggestions for specific research priorities and for the development of resources to advance our knowledge of the mechanisms, diagnosis, management, and prevention of this allergic disease. This publication is a summary of the workshop and the panel's recommendations are presented herein.
Collapse
Affiliation(s)
| | - Scott P Commins
- Departments of Medicine & Pediatrics, University of North Carolina, Chapel Hill, NC
| | - Tilo Biedermann
- Department of Dermatology and Allergy, Technical University of Munich and Unit Clinical Allergology (EKA), Helmholtz Zentrum München, Munich, Germany
| | - Marianne van Hage
- Division of Immunology and Allergy, Department of Medicine Solna, Karolinska Institutet and University Hospital, Stockholm, Sweden
| | - Michael Levin
- Division of Paediatric Allergy, University of Cape Town, Cape Town, South Africa
| | - Lisa A Beck
- Department of Dermatology, University of Rochester Medical Center, Rochester, NY
| | - Maria Diuk-Wasser
- Department of Ecology, Evolution, and Environmental Biology, Columbia University, New York, NY
| | - Uta Jappe
- Division of Clinical and Molecular Allergology, Research Center Borstel, Airway Research, Center North (ARCN), Member of the German Center for Lung Research, Borstel, Germany; Interdisciplinary Allergy Outpatient Clinic, Department of Pneumology, University of Lübeck, Lübeck, Germany
| | - Danijela Apostolovic
- Division of Immunology and Allergy, Department of Medicine Solna, Karolinska Institutet and University Hospital, Stockholm, Sweden
| | - Michael Minnicozzi
- Division of Allergy, Immunology and Transplantation, Allergy, Asthma and Airway Biology Branch, National Institute of Allergy and Infectious Diseases, NIH, Rockville, Md
| | - Marshall Plaut
- Division of Allergy, Immunology and Transplantation, Allergy, Asthma and Airway Biology Branch, National Institute of Allergy and Infectious Diseases, NIH, Rockville, Md
| | - Jeffrey M Wilson
- Division of Allergy & Clinical Immunology, University of Virginia, Charlottesville, Va
| |
Collapse
|
187
|
Clay SL, Bravo-Blas A, Wall DM, MacLeod MKL, Milling SWF. Regulatory T cells control the dynamic and site-specific polarization of total CD4 T cells following Salmonella infection. Mucosal Immunol 2020; 13:946-957. [PMID: 32457450 PMCID: PMC7567643 DOI: 10.1038/s41385-020-0299-1] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2019] [Revised: 04/10/2020] [Accepted: 04/21/2020] [Indexed: 02/06/2023]
Abstract
FoxP3+ regulatory T cells (Tregs) control inflammation and maintain mucosal homeostasis, but their functions during infection are poorly understood. Th1, Th2, and Th17 cells can be identified by master transcription factors (TFs) T-bet, GATA3, and RORγT; Tregs also express these TFs. While T-bet+ Tregs can selectively suppress Th1 cells, it is unclear whether distinct Treg populations can alter Th bias. To address this, we used Salmonella enterica serotype Typhimurium to induce nonlethal colitis. Following infection, we observed an early colonic Th17 response within total CD4 T cells, followed by a Th1 bias. The early Th17 response, which contains both Salmonella-specific and non-Salmonella-specific cells, parallels an increase in T-bet+ Tregs. Later, Th1 cells and RORγT+ Tregs dominate. This reciprocal dynamic may indicate that Tregs selectively suppress Th cells, shaping the immune response. Treg depletion 1-2 days post-infection shifted the early Th17 response to a Th1 bias; however, Treg depletion 6-7 days post-infection abrogated the Th1 bias. Thus, Tregs are necessary for the early Th17 response, and for a maximal Th1 response later. These data show that Tregs shape the overall tissue CD4 T cell response and highlight the potential for subpopulations of Tregs to be used in targeted therapeutic approaches.
Collapse
Affiliation(s)
- Slater L. Clay
- grid.8756.c0000 0001 2193 314XInstitute of Infection, Immunity and Inflammation, College of Medical, Veterinary and Life Sciences, University of Glasgow, 120 University Place, Glasgow, UK ,grid.38142.3c000000041936754XDepartment of Immunology and Infectious Diseases, Harvard T.H. Chan School of Public Health, 665 Huntington Avenue, Boston, MA 02115 USA
| | - Alberto Bravo-Blas
- grid.8756.c0000 0001 2193 314XInstitute of Infection, Immunity and Inflammation, College of Medical, Veterinary and Life Sciences, University of Glasgow, 120 University Place, Glasgow, UK ,grid.23636.320000 0000 8821 5196Institute of Cancer Sciences, University of Glasgow and Cancer Research UK Beatson Institute, Glasgow, G61 1BD UK
| | - Daniel M. Wall
- grid.8756.c0000 0001 2193 314XInstitute of Infection, Immunity and Inflammation, College of Medical, Veterinary and Life Sciences, University of Glasgow, 120 University Place, Glasgow, UK
| | - Megan K. L. MacLeod
- grid.8756.c0000 0001 2193 314XInstitute of Infection, Immunity and Inflammation, College of Medical, Veterinary and Life Sciences, University of Glasgow, 120 University Place, Glasgow, UK
| | - Simon W. F. Milling
- grid.8756.c0000 0001 2193 314XInstitute of Infection, Immunity and Inflammation, College of Medical, Veterinary and Life Sciences, University of Glasgow, 120 University Place, Glasgow, UK
| |
Collapse
|
188
|
Larsen SB, Cowley CJ, Fuchs E. Epithelial cells: liaisons of immunity. Curr Opin Immunol 2019; 62:45-53. [PMID: 31874430 DOI: 10.1016/j.coi.2019.11.004] [Citation(s) in RCA: 65] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2019] [Accepted: 11/13/2019] [Indexed: 01/12/2023]
Abstract
The surface and lining tissues of our body are exposed to the external environment, and as such these epithelial tissues must form structural barriers able to defend against microbes, environmental toxins, and mechanical stress. Their cells are equipped to detect a diverse array of surface perturbations, and then launch signaling relays to the immune system. The aim of these liaisons is to coordinate the requisite immune cell response needed to preserve and/or restore barrier integrity and defend the host. It has been recently appreciated that epithelial cells learn from these experiences. Following inflammatory exposure, long-lived stem cells within the tissue retain an epigenetic memory that endows them with heightened responsiveness to subsequent encounters with stress. Here, we review the recent literature on how epithelial cells sense signals from microbes, allergens, and injury at the tissue surface, and transmit this information to immune cells, while embedding a memory of the experience within their chromatin.
Collapse
Affiliation(s)
- Samantha B Larsen
- Robin Chemers Neustein Laboratory of Mammalian Cell Biology and Development, Howard Hughes Medical Institute, The Rockefeller University, New York, NY 10065, USA; New York University School of Medicine, Neuroscience Institute, New York, NY 10016, USA
| | - Christopher J Cowley
- Robin Chemers Neustein Laboratory of Mammalian Cell Biology and Development, Howard Hughes Medical Institute, The Rockefeller University, New York, NY 10065, USA
| | - Elaine Fuchs
- Robin Chemers Neustein Laboratory of Mammalian Cell Biology and Development, Howard Hughes Medical Institute, The Rockefeller University, New York, NY 10065, USA.
| |
Collapse
|
189
|
Shi H, Chi H. Metabolic Control of Treg Cell Stability, Plasticity, and Tissue-Specific Heterogeneity. Front Immunol 2019; 10:2716. [PMID: 31921097 PMCID: PMC6917616 DOI: 10.3389/fimmu.2019.02716] [Citation(s) in RCA: 131] [Impact Index Per Article: 21.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2019] [Accepted: 11/05/2019] [Indexed: 12/21/2022] Open
Abstract
Regulatory T (Treg) cells are crucial for peripheral immune tolerance and prevention of autoimmunity and tissue damage. Treg cells are inherently defined by the expression of the transcription factor Foxp3, which enforces lineage development and immune suppressive function of these cells. Under various conditions as observed in autoimmunity, cancer and non-lymphoid tissues, a proportion of Treg cells respond to specific environmental signals and display altered stability, plasticity and tissue-specific heterogeneity, which further shape their context-dependent suppressive functions. Recent studies have revealed that metabolic programs play pivotal roles in controlling these processes in Treg cells, thereby considerably expanding our understanding of Treg cell biology. Here we summarize these recent advances that highlight how cell-extrinsic factors, such as nutrients, vitamins and metabolites, and cell-intrinsic metabolic programs, orchestrate Treg cell stability, plasticity, and tissue-specific heterogeneity. Understanding metabolic regulation of Treg cells should provide new insight into immune homeostasis and disease, with important therapeutic implications for autoimmunity, cancer, and other immune-mediated disorders.
Collapse
Affiliation(s)
- Hao Shi
- Department of Immunology, St. Jude Children's Research Hospital, Memphis, TN, United States
| | - Hongbo Chi
- Department of Immunology, St. Jude Children's Research Hospital, Memphis, TN, United States
| |
Collapse
|
190
|
McGinty JW, von Moltke J. A three course menu for ILC and bystander T cell activation. Curr Opin Immunol 2019; 62:15-21. [PMID: 31830683 DOI: 10.1016/j.coi.2019.11.005] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2019] [Accepted: 11/11/2019] [Indexed: 02/06/2023]
Abstract
The varied list of agonists that activate innate lymphoid cells (ILCs) continues to grow, but whether and how these signals interact is not well defined, especially in vivo. ILC subsets share master transcription factors, chromatin landscapes, and effector cytokines with their corresponding T helper (Th) cell subsets. Here we discuss how studies of these two cell types can inform each other. Specifically, we outline a framework in which ILC agonists are grouped by the transcription factors they activate. Optimal ILC activation requires at least three items from a 'menu' of non-redundant signals that collectively replicate the STAT and TCR signaling that drives effector Th cell function. This conceptual model may also apply to TCR-independent 'bystander' activation of Th cells.
Collapse
Affiliation(s)
- John W McGinty
- Department of Immunology, University of Washington School of Medicine, Seattle, WA, 98109, USA
| | - Jakob von Moltke
- Department of Immunology, University of Washington School of Medicine, Seattle, WA, 98109, USA.
| |
Collapse
|
191
|
Quoi de neuf en recherche ? Ann Dermatol Venereol 2019; 146:12S19-12S23. [DOI: 10.1016/s0151-9638(20)30102-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
|
192
|
Barrat FJ, Crow MK, Ivashkiv LB. Interferon target-gene expression and epigenomic signatures in health and disease. Nat Immunol 2019; 20:1574-1583. [PMID: 31745335 PMCID: PMC7024546 DOI: 10.1038/s41590-019-0466-2] [Citation(s) in RCA: 349] [Impact Index Per Article: 58.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2019] [Accepted: 07/10/2019] [Indexed: 02/06/2023]
Abstract
Multiple type I interferons and interferon-γ (IFN-γ) are expressed under physiological conditions and are increased by stress and infections, and in autoinflammatory and autoimmune diseases. Interferons activate the Jak-STAT signaling pathway and induce overlapping patterns of expression, called 'interferon signatures', of canonical interferon-stimulated genes (ISGs) encoding molecules important for antiviral responses, antigen presentation, autoimmunity and inflammation. It has now become clear that interferons also induce an 'interferon epigenomic signature' by activating latent enhancers and 'bookmarking' chromatin, thus reprogramming cell responses to environmental cues. The interferon epigenomic signature affects ISGs and other gene sets, including canonical targets of the transcription factor NF-κB that encode inflammatory molecules, and is involved in the priming of immune cells, tolerance and the training of innate immune memory. Here we review the mechanisms through which interferon signatures and interferon epigenomic signatures are generated, as well as the expression and functional consequences of these signatures in homeostasis and autoimmune diseases, including systemic lupus erythematosus, rheumatoid arthritis and systemic sclerosis.
Collapse
Affiliation(s)
- Franck J Barrat
- Research Institute and David Z. Rosensweig Genomics Research Center, Hospital for Special Surgery, New York, NY, USA
- Department of Microbiology and Immunology, Weill Cornell Medicine, New York, NY, USA
- Immunology and Microbial Pathogenesis Program, Weill Cornell Medicine, New York, NY, USA
| | - Mary K Crow
- Research Institute and David Z. Rosensweig Genomics Research Center, Hospital for Special Surgery, New York, NY, USA
- Immunology and Microbial Pathogenesis Program, Weill Cornell Medicine, New York, NY, USA
- Department of Medicine, Weill Cornell Medicine, New York, NY, USA
| | - Lionel B Ivashkiv
- Research Institute and David Z. Rosensweig Genomics Research Center, Hospital for Special Surgery, New York, NY, USA.
- Immunology and Microbial Pathogenesis Program, Weill Cornell Medicine, New York, NY, USA.
- Department of Medicine, Weill Cornell Medicine, New York, NY, USA.
| |
Collapse
|
193
|
Li XV, Leonardi I, Iliev ID. Gut Mycobiota in Immunity and Inflammatory Disease. Immunity 2019; 50:1365-1379. [PMID: 31216461 DOI: 10.1016/j.immuni.2019.05.023] [Citation(s) in RCA: 165] [Impact Index Per Article: 27.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2019] [Revised: 05/01/2019] [Accepted: 05/29/2019] [Indexed: 02/08/2023]
Abstract
The mammalian intestine is colonized by a wealth of microorganisms-including bacteria, viruses, protozoa, and fungi-that are all integrated into a functional trans-kingdom community. Characterization of the composition of the fungal community-the mycobiota-has advanced further than the much-needed mechanistic studies. Recent findings have revealed roles for the gut mycobiota in the regulation of host immunity and in the development and progression of human diseases of inflammatory origin. We review these findings here while placing them in the context of the current understanding of the pathways and cellular networks that induce local and systemic immune responses to fungi in the gastrointestinal tract. We discuss gaps in knowledge and argue for the importance of considering bacteria-fungal interactions as we aim to define the roles of mycobiota in immune homeostasis and immune-associated pathologies.
Collapse
Affiliation(s)
- Xin V Li
- Gastroenterology and Hepatology Division, Joan and Sanford I. Weill Department of Medicine, Weill Cornell Medicine, Cornell University, New York, NY 10021, USA; Jill Roberts Institute for Research in Inflammatory Bowel Disease, Weill Cornell Medicine, Cornell University, New York, NY 10021, USA
| | - Irina Leonardi
- Gastroenterology and Hepatology Division, Joan and Sanford I. Weill Department of Medicine, Weill Cornell Medicine, Cornell University, New York, NY 10021, USA; Jill Roberts Institute for Research in Inflammatory Bowel Disease, Weill Cornell Medicine, Cornell University, New York, NY 10021, USA
| | - Iliyan D Iliev
- Gastroenterology and Hepatology Division, Joan and Sanford I. Weill Department of Medicine, Weill Cornell Medicine, Cornell University, New York, NY 10021, USA; Jill Roberts Institute for Research in Inflammatory Bowel Disease, Weill Cornell Medicine, Cornell University, New York, NY 10021, USA; Department of Microbiology and Immunology, Weill Cornell Medicine, Cornell University, New York, NY 10065, USA; Immunology and Microbial Pathogenesis Program, Weill Cornell Graduate School of Medical Sciences, Weill Cornell Medicine, Cornell University, New York, NY 10065, USA.
| |
Collapse
|
194
|
Keratinocyte-intrinsic MHCII expression controls microbiota-induced Th1 cell responses. Proc Natl Acad Sci U S A 2019; 116:23643-23652. [PMID: 31672911 DOI: 10.1073/pnas.1912432116] [Citation(s) in RCA: 48] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
The cross-talk between the microbiota and the immune system plays a fundamental role in the control of host physiology. However, the tissue-specific factors controlling this dialogue remain poorly understood. Here we demonstrate that T cell responses to commensal colonization are associated with the development of organized cellular clusters within the skin epithelium. These organized lymphocyte clusters are surrounded by keratinocytes expressing a discrete program associated with antigen presentation and antimicrobial defense. Notably, IL-22-mediated keratinocyte-intrinsic MHC class II expression was required for the selective accumulation of commensal-induced IFN-γ, but not IL-17A-producing CD4+ T cells within the skin. Taking these data together, this work uncovers an unexpected role for MHC class II expression by keratinocytes in the control of homeostatic type 1 responses to the microbiota. Our findings have important implications for the understanding of the tissue-specific rules governing the dialogue between a host and its microbiota.
Collapse
|
195
|
Owen DL, Sjaastad LE, Farrar MA. Regulatory T Cell Development in the Thymus. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2019; 203:2031-2041. [PMID: 31591259 PMCID: PMC6910132 DOI: 10.4049/jimmunol.1900662] [Citation(s) in RCA: 65] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/13/2019] [Accepted: 08/23/2019] [Indexed: 12/19/2022]
Abstract
Development of a comprehensive regulatory T (Treg) cell compartment in the thymus is required to maintain immune homeostasis and prevent autoimmunity. In this study, we review cellular and molecular determinants of Treg cell development in the thymus. We focus on the evidence for a self-antigen-focused Treg cell repertoire as well as the APCs responsible for presenting self-antigens to developing thymocytes. We also cover the contribution of different cytokines to thymic Treg development and the cellular populations that produce these cytokines. Finally, we update the originally proposed "two-step" model of thymic Treg differentiation by incorporating new evidence demonstrating that Treg cells develop from two Treg progenitor populations and discuss the functional importance of Treg cells generated via either progenitor pathway.
Collapse
Affiliation(s)
- David L Owen
- Center for Immunology, University of Minnesota, Minneapolis, MN 55455; Masonic Cancer Center, University of Minnesota, Minneapolis, MN 55455; and Department of Laboratory Medicine and Pathology, University of Minnesota, Minneapolis, MN 55455
| | - Louisa E Sjaastad
- Center for Immunology, University of Minnesota, Minneapolis, MN 55455; Masonic Cancer Center, University of Minnesota, Minneapolis, MN 55455; and Department of Laboratory Medicine and Pathology, University of Minnesota, Minneapolis, MN 55455
| | - Michael A Farrar
- Center for Immunology, University of Minnesota, Minneapolis, MN 55455; Masonic Cancer Center, University of Minnesota, Minneapolis, MN 55455; and Department of Laboratory Medicine and Pathology, University of Minnesota, Minneapolis, MN 55455
| |
Collapse
|
196
|
Zhang Z, Tang H, Chen P, Xie H, Tao Y. Demystifying the manipulation of host immunity, metabolism, and extraintestinal tumors by the gut microbiome. Signal Transduct Target Ther 2019; 4:41. [PMID: 31637019 PMCID: PMC6799818 DOI: 10.1038/s41392-019-0074-5] [Citation(s) in RCA: 153] [Impact Index Per Article: 25.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2019] [Revised: 08/27/2019] [Accepted: 08/27/2019] [Indexed: 02/06/2023] Open
Abstract
The trillions of microorganisms in the gut microbiome have attracted much attention recently owing to their sophisticated and widespread impacts on numerous aspects of host pathophysiology. Remarkable progress in large-scale sequencing and mass spectrometry has increased our understanding of the influence of the microbiome and/or its metabolites on the onset and progression of extraintestinal cancers and the efficacy of cancer immunotherapy. Given the plasticity in microbial composition and function, microbial-based therapeutic interventions, including dietary modulation, prebiotics, and probiotics, as well as fecal microbial transplantation, potentially permit the development of novel strategies for cancer therapy to improve clinical outcomes. Herein, we summarize the latest evidence on the involvement of the gut microbiome in host immunity and metabolism, the effects of the microbiome on extraintestinal cancers and the immune response, and strategies to modulate the gut microbiome, and we discuss ongoing studies and future areas of research that deserve focused research efforts.
Collapse
Affiliation(s)
- Ziying Zhang
- Key Laboratory of Carcinogenesis and Cancer Invasion, Ministry of Education, Department of Pathology, Xiangya Hospital, Central South University, 410078 Hunan, China
- NHC Key Laboratory of Carcinogenesis (Central South University), Cancer Research Institute and School of Basic Medicine, Central South University, 410078 Changsha, Hunan China
- Hunan Key Laboratory of Tumor Models and Individualized Medicine, Department of Thoracic Surgery, Second Xiangya Hospital, Central South University, 410011 Changsha, China
- Department of Oncology, Third Xiangya Hospital, Central South University, 410013 Changsha, China
| | - Haosheng Tang
- Key Laboratory of Carcinogenesis and Cancer Invasion, Ministry of Education, Department of Pathology, Xiangya Hospital, Central South University, 410078 Hunan, China
- NHC Key Laboratory of Carcinogenesis (Central South University), Cancer Research Institute and School of Basic Medicine, Central South University, 410078 Changsha, Hunan China
- Hunan Key Laboratory of Tumor Models and Individualized Medicine, Department of Thoracic Surgery, Second Xiangya Hospital, Central South University, 410011 Changsha, China
| | - Peng Chen
- Department of Urology, Xiangya Hospital, Central South University, 410008 Changsha, China
| | - Hui Xie
- Department of Thoracic and Cardiovascular Surgery, Second Xiangya Hospital of Central South University, 410011 Changsha, China
| | - Yongguang Tao
- Key Laboratory of Carcinogenesis and Cancer Invasion, Ministry of Education, Department of Pathology, Xiangya Hospital, Central South University, 410078 Hunan, China
- NHC Key Laboratory of Carcinogenesis (Central South University), Cancer Research Institute and School of Basic Medicine, Central South University, 410078 Changsha, Hunan China
- Hunan Key Laboratory of Tumor Models and Individualized Medicine, Department of Thoracic Surgery, Second Xiangya Hospital, Central South University, 410011 Changsha, China
| |
Collapse
|
197
|
Tax G, Lia A, Santino A, Roversi P. Modulation of ERQC and ERAD: A Broad-Spectrum Spanner in the Works of Cancer Cells? JOURNAL OF ONCOLOGY 2019; 2019:8384913. [PMID: 31662755 PMCID: PMC6791201 DOI: 10.1155/2019/8384913] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/20/2019] [Accepted: 08/27/2019] [Indexed: 12/21/2022]
Abstract
Endoplasmic reticulum glycoprotein folding quality control (ERQC) and ER-associated degradation (ERAD) preside over cellular glycoprotein secretion and maintain steady glycoproteostasis. When cells turn malignant, cancer cell plasticity is affected and supported either by point mutations, preferential isoform selection, altered expression levels, or shifts to conformational equilibria of a secreted glycoprotein. Such changes are crucial in mediating altered extracellular signalling, metabolic behavior, and adhesion properties of cancer cells. It is therefore conceivable that interference with ERQC and/or ERAD can be used to selectively damage cancers. Indeed, inhibitors of the late stages of ERAD are already in the clinic against cancers such as multiple myeloma. Here, we review recent advances in our understanding of the complex relationship between glycoproteostasis and cancer biology and discuss the potential of ERQC and ERAD modulators for the selective targeting of cancer cell plasticity.
Collapse
Affiliation(s)
- Gábor Tax
- Leicester Institute of Structural and Chemical Biology, Department of Molecular and Cell Biology, University of Leicester, Henry Wellcome Building, Lancaster Road, Leicester LE1 7RH, UK
| | - Andrea Lia
- Leicester Institute of Structural and Chemical Biology, Department of Molecular and Cell Biology, University of Leicester, Henry Wellcome Building, Lancaster Road, Leicester LE1 7RH, UK
- Institute of Sciences of Food Production, C.N.R. Unit of Lecce, via Monteroni, I-73100 Lecce, Italy
| | - Angelo Santino
- Institute of Sciences of Food Production, C.N.R. Unit of Lecce, via Monteroni, I-73100 Lecce, Italy
| | - Pietro Roversi
- Leicester Institute of Structural and Chemical Biology, Department of Molecular and Cell Biology, University of Leicester, Henry Wellcome Building, Lancaster Road, Leicester LE1 7RH, UK
| |
Collapse
|
198
|
Leng T, Akther HD, Hackstein CP, Powell K, King T, Friedrich M, Christoforidou Z, McCuaig S, Neyazi M, Arancibia-Cárcamo CV, Hagel J, Powrie F, Peres RS, Millar V, Ebner D, Lamichhane R, Ussher J, Hinks TSC, Marchi E, Willberg C, Klenerman P. TCR and Inflammatory Signals Tune Human MAIT Cells to Exert Specific Tissue Repair and Effector Functions. Cell Rep 2019; 28:3077-3091.e5. [PMID: 31533032 PMCID: PMC6899450 DOI: 10.1016/j.celrep.2019.08.050] [Citation(s) in RCA: 177] [Impact Index Per Article: 29.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2018] [Revised: 04/17/2019] [Accepted: 08/15/2019] [Indexed: 01/10/2023] Open
Abstract
MAIT cells are an unconventional T cell population that can be activated through both TCR-dependent and TCR-independent mechanisms. Here, we examined the impact of combinations of TCR-dependent and TCR-independent signals in human CD8+ MAIT cells. TCR-independent activation of these MAIT cells from blood and gut was maximized by extending the panel of cytokines to include TNF-superfamily member TL1A. RNA-seq experiments revealed that TCR-dependent and TCR-independent signals drive MAIT cells to exert overlapping and specific effector functions, affecting both host defense and tissue homeostasis. Although TCR triggering alone is insufficient to drive sustained activation, TCR-triggered MAIT cells showed specific enrichment of tissue-repair functions at the gene and protein levels and in in vitro assays. Altogether, these data indicate the blend of TCR-dependent and TCR-independent signaling to CD8+ MAIT cells may play a role in controlling the balance between healthy and pathological processes of tissue inflammation and repair.
Collapse
Affiliation(s)
- Tianqi Leng
- Peter Medawar Building for Pathogen Research, South Parks Road, Oxford OX1 3SY, UK
| | - Hossain Delowar Akther
- Peter Medawar Building for Pathogen Research, South Parks Road, Oxford OX1 3SY, UK; Translational Gastroenterology Unit, Nuffield Department of Medicine, University of Oxford, Oxford OX3 9DU, UK
| | - Carl-Philipp Hackstein
- Peter Medawar Building for Pathogen Research, South Parks Road, Oxford OX1 3SY, UK; Translational Gastroenterology Unit, Nuffield Department of Medicine, University of Oxford, Oxford OX3 9DU, UK
| | - Kate Powell
- Peter Medawar Building for Pathogen Research, South Parks Road, Oxford OX1 3SY, UK; Department of Microbiology and Immunology, University of Otago, Otago, New Zealand
| | - Thomas King
- Peter Medawar Building for Pathogen Research, South Parks Road, Oxford OX1 3SY, UK
| | - Matthias Friedrich
- The Kennedy Institute of Rheumatology, Roosevelt Dr., Oxford OX3 7FY, UK
| | - Zoe Christoforidou
- Translational Gastroenterology Unit, Nuffield Department of Medicine, University of Oxford, Oxford OX3 9DU, UK
| | - Sarah McCuaig
- The Kennedy Institute of Rheumatology, Roosevelt Dr., Oxford OX3 7FY, UK
| | - Mastura Neyazi
- The Kennedy Institute of Rheumatology, Roosevelt Dr., Oxford OX3 7FY, UK
| | | | - Joachim Hagel
- Peter Medawar Building for Pathogen Research, South Parks Road, Oxford OX1 3SY, UK
| | - Fiona Powrie
- The Kennedy Institute of Rheumatology, Roosevelt Dr., Oxford OX3 7FY, UK
| | | | - Val Millar
- Target Discovery Institute, Roosevelt Dr., Oxford OX3 7FZ, UK
| | - Daniel Ebner
- Target Discovery Institute, Roosevelt Dr., Oxford OX3 7FZ, UK
| | - Rajesh Lamichhane
- Department of Microbiology and Immunology, University of Otago, Otago, New Zealand
| | - James Ussher
- Department of Microbiology and Immunology, University of Otago, Otago, New Zealand
| | - Timothy S C Hinks
- NIHR Biomedical Research Centre, John Radcliffe Hospital, Oxford OX3 9DU, UK; Respiratory Medicine Unit, Nuffield Department of Medicine Experimental Medicine, University of Oxford, Oxford OX3 9DU, UK; Department of Microbiology and Immunology, Peter Doherty Institute for Infection and Immunity, University of Melbourne, Melbourne, VIC 3000, Australia
| | - Emanuele Marchi
- Peter Medawar Building for Pathogen Research, South Parks Road, Oxford OX1 3SY, UK
| | - Chris Willberg
- Peter Medawar Building for Pathogen Research, South Parks Road, Oxford OX1 3SY, UK
| | - Paul Klenerman
- Peter Medawar Building for Pathogen Research, South Parks Road, Oxford OX1 3SY, UK; Translational Gastroenterology Unit, Nuffield Department of Medicine, University of Oxford, Oxford OX3 9DU, UK; NIHR Biomedical Research Centre, John Radcliffe Hospital, Oxford OX3 9DU, UK.
| |
Collapse
|
199
|
Leonard WJ, Lin JX, O'Shea JJ. The γ c Family of Cytokines: Basic Biology to Therapeutic Ramifications. Immunity 2019; 50:832-850. [PMID: 30995502 DOI: 10.1016/j.immuni.2019.03.028] [Citation(s) in RCA: 260] [Impact Index Per Article: 43.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2019] [Revised: 03/22/2019] [Accepted: 03/26/2019] [Indexed: 12/15/2022]
Abstract
The common cytokine receptor γ chain, γc, is a component of the receptors for interleukin-2 (IL-2), IL-4, IL-7, IL-9, IL-15, and IL-21. Mutation of the gene encoding γc results in X-linked severe combined immunodeficiency in humans, and γc family cytokines collectively regulate development, proliferation, survival, and differentiation of immune cells. Here, we review the basic biology of these cytokines, highlighting mechanisms of signaling and gene regulation that have provided insights for immunodeficiency, autoimmunity, allergic diseases, and cancer. Moreover, we discuss how studies of this family stimulated the development of JAK3 inhibitors and present an overview of current strategies targeting these pathways in the clinic, including novel antibodies, antagonists, and partial agonists. The diverse roles of these cytokines on a range of immune cells have important therapeutic implications.
Collapse
Affiliation(s)
- Warren J Leonard
- Laboratory of Molecular Immunology and the Immunology Center, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD 20892-1674, USA.
| | - Jian-Xin Lin
- Laboratory of Molecular Immunology and the Immunology Center, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD 20892-1674, USA.
| | - John J O'Shea
- Molecular Immunology and Inflammation Branch, National Institute of Arthritis, Metabolic, and Skin Diseases, National Institutes of Health, Bethesda, MD 20892-1674, USA.
| |
Collapse
|
200
|
Deckers J, Lambrecht BN, Hammad H. How a farming environment protects from atopy. Curr Opin Immunol 2019; 60:163-169. [PMID: 31499321 DOI: 10.1016/j.coi.2019.08.001] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2019] [Accepted: 08/02/2019] [Indexed: 12/15/2022]
Abstract
It is now well established that the exposure to certain environments such as farms has the potential to protect from the development of allergies later in life. This protection is achieved when repeated exposure to the farming environment occurs early in life, but persists when children spend sufficient amount of time in contact with livestock and hay, and drink unpasteurized milk. The capacity of farm dust to protect from allergy development lies, amongst others, in the microbe composition in the farm. These protective microbes release various metabolites and cell wall components that change farmers' home dust composition, when compared to urbanized home dust. Additionally, they can colonize various barrier sites (skin, lung, intestine) in farmers' children, leading to persistent changes in the way their immune system and their barrier cells respond to environmental allergens.
Collapse
Affiliation(s)
- Julie Deckers
- Laboratory of Immunoregulation and Mucosal Immunology, VIB Center for Inflammation Research, Technologiepark-Zwijnaarde 71, B-9052 Ghent (Zwijnaarde), Belgium; Department of Internal Medicine and Pediatrics, Ghent University, Ghent, Belgium
| | - Bart N Lambrecht
- Laboratory of Immunoregulation and Mucosal Immunology, VIB Center for Inflammation Research, Technologiepark-Zwijnaarde 71, B-9052 Ghent (Zwijnaarde), Belgium; Department of Internal Medicine and Pediatrics, Ghent University, Ghent, Belgium; Department of Respiratory Medicine, Erasmus Medical Center, Rotterdam, The Netherlands.
| | - Hamida Hammad
- Laboratory of Immunoregulation and Mucosal Immunology, VIB Center for Inflammation Research, Technologiepark-Zwijnaarde 71, B-9052 Ghent (Zwijnaarde), Belgium; Department of Internal Medicine and Pediatrics, Ghent University, Ghent, Belgium
| |
Collapse
|