1
|
Arbogast F, Sal-Carro R, Boufenghour W, Frenger Q, Bouis D, Filippi De La Palavesa L, Fauny JD, Griso O, Puccio H, Fima R, Huby T, Gautier EL, Molitor A, Carapito R, Bahram S, Romani N, Clausen BE, Voisin B, Mueller CG, Gros F, Flacher V. Epidermal maintenance of Langerhans cells relies on autophagy-regulated lipid metabolism. J Cell Biol 2025; 224:e202403178. [PMID: 39535446 PMCID: PMC11561468 DOI: 10.1083/jcb.202403178] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2024] [Revised: 09/12/2024] [Accepted: 10/23/2024] [Indexed: 11/16/2024] Open
Abstract
Macroautophagy (often-named autophagy), a catabolic process involving autophagy-related (Atg) genes, prevents the accumulation of harmful cytoplasmic components and mobilizes energy reserves in long-lived and self-renewing cells. Autophagy deficiency affects antigen presentation in conventional dendritic cells (DCs) without impacting their survival. However, previous studies did not address epidermal Langerhans cells (LCs). Here, we demonstrate that deletion of either Atg5 or Atg7 in LCs leads to their gradual depletion. ATG5-deficient LCs showed metabolic dysregulation and accumulated neutral lipids. Despite increased mitochondrial respiratory capacity, they were unable to process lipids, eventually leading them to ferroptosis. Finally, metabolically impaired LCs upregulated proinflammatory transcripts and showed decreased expression of neuronal interaction receptors. Altogether, autophagy represents a critical regulator of lipid storage and metabolism in LCs, allowing their maintenance in the epidermis.
Collapse
Affiliation(s)
- Florent Arbogast
- Laboratory CNRS I2CT/UPR3572 Immunology, Immunopathology and Therapeutic Chemistry, Strasbourg Drug Discovery and Development Institute (IMS), Institut de Biologie Moléculaire et Cellulaire, Strasbourg, France
- Université de Strasbourg, Strasbourg, France
| | - Raquel Sal-Carro
- Laboratory CNRS I2CT/UPR3572 Immunology, Immunopathology and Therapeutic Chemistry, Strasbourg Drug Discovery and Development Institute (IMS), Institut de Biologie Moléculaire et Cellulaire, Strasbourg, France
| | - Wacym Boufenghour
- Laboratory CNRS I2CT/UPR3572 Immunology, Immunopathology and Therapeutic Chemistry, Strasbourg Drug Discovery and Development Institute (IMS), Institut de Biologie Moléculaire et Cellulaire, Strasbourg, France
| | | | - Delphine Bouis
- Laboratory CNRS I2CT/UPR3572 Immunology, Immunopathology and Therapeutic Chemistry, Strasbourg Drug Discovery and Development Institute (IMS), Institut de Biologie Moléculaire et Cellulaire, Strasbourg, France
| | - Louise Filippi De La Palavesa
- Laboratory CNRS I2CT/UPR3572 Immunology, Immunopathology and Therapeutic Chemistry, Strasbourg Drug Discovery and Development Institute (IMS), Institut de Biologie Moléculaire et Cellulaire, Strasbourg, France
| | - Jean-Daniel Fauny
- Laboratory CNRS I2CT/UPR3572 Immunology, Immunopathology and Therapeutic Chemistry, Strasbourg Drug Discovery and Development Institute (IMS), Institut de Biologie Moléculaire et Cellulaire, Strasbourg, France
| | - Olivier Griso
- Institut de Génétique et de Biologie Moléculaire et Cellulaire, INSERM U1258/CNRS UMR7104, Illkirch, France
| | - Hélène Puccio
- Institut de Génétique et de Biologie Moléculaire et Cellulaire, INSERM U1258/CNRS UMR7104, Illkirch, France
| | - Rebecca Fima
- Sorbonne Université, INSERM UMR_S 1166 ICAN, Paris, France
| | - Thierry Huby
- Sorbonne Université, INSERM UMR_S 1166 ICAN, Paris, France
| | | | - Anne Molitor
- Laboratoire d’Immunorhumatologie Moléculaire, Plateforme GENOMAX, INSERM UMR_S 1109, Faculté de Médecine, Fédération Hospitalo-Universitaire OMICARE, ITI TRANSPLANTEX NG, Université de Strasbourg, Strasbourg, France
- Strasbourg Federation of Translational Medicine (FMTS), Strasbourg University, Strasbourg, France
| | - Raphaël Carapito
- Laboratoire d’Immunorhumatologie Moléculaire, Plateforme GENOMAX, INSERM UMR_S 1109, Faculté de Médecine, Fédération Hospitalo-Universitaire OMICARE, ITI TRANSPLANTEX NG, Université de Strasbourg, Strasbourg, France
- Strasbourg Federation of Translational Medicine (FMTS), Strasbourg University, Strasbourg, France
- Service d’Immunologie Biologique, Plateau Technique de Biologie, Pôle de Biologie, Nouvel Hôpital Civil, Hôpitaux Universitaires de Strasbourg, Strasbourg, France
| | - Seiamak Bahram
- Laboratoire d’Immunorhumatologie Moléculaire, Plateforme GENOMAX, INSERM UMR_S 1109, Faculté de Médecine, Fédération Hospitalo-Universitaire OMICARE, ITI TRANSPLANTEX NG, Université de Strasbourg, Strasbourg, France
- Strasbourg Federation of Translational Medicine (FMTS), Strasbourg University, Strasbourg, France
- Service d’Immunologie Biologique, Plateau Technique de Biologie, Pôle de Biologie, Nouvel Hôpital Civil, Hôpitaux Universitaires de Strasbourg, Strasbourg, France
| | - Nikolaus Romani
- Department of Dermatology, Venereology and Allergology, Medical University of Innsbruck, Innsbruck, Austria
| | - Björn E. Clausen
- Institute for Molecular Medicine and Paul Klein Center for Immunotherapy (PKZI), University Medical Center of the Johannes Gutenberg-University Mainz, Mainz, Germany
| | - Benjamin Voisin
- Laboratory CNRS I2CT/UPR3572 Immunology, Immunopathology and Therapeutic Chemistry, Strasbourg Drug Discovery and Development Institute (IMS), Institut de Biologie Moléculaire et Cellulaire, Strasbourg, France
| | - Christopher G. Mueller
- Laboratory CNRS I2CT/UPR3572 Immunology, Immunopathology and Therapeutic Chemistry, Strasbourg Drug Discovery and Development Institute (IMS), Institut de Biologie Moléculaire et Cellulaire, Strasbourg, France
| | - Frédéric Gros
- Laboratory CNRS I2CT/UPR3572 Immunology, Immunopathology and Therapeutic Chemistry, Strasbourg Drug Discovery and Development Institute (IMS), Institut de Biologie Moléculaire et Cellulaire, Strasbourg, France
- Université de Strasbourg, Strasbourg, France
| | - Vincent Flacher
- Laboratory CNRS I2CT/UPR3572 Immunology, Immunopathology and Therapeutic Chemistry, Strasbourg Drug Discovery and Development Institute (IMS), Institut de Biologie Moléculaire et Cellulaire, Strasbourg, France
| |
Collapse
|
2
|
Raquer-McKay HM, Maqueda-Alfaro RA, Saravanan S, Arroyo Hornero R, Clausen BE, Gottfried-Blackmore A, Idoyaga J. Monocytes give rise to Langerhans cells that preferentially migrate to lymph nodes at steady state. Proc Natl Acad Sci U S A 2024; 121:e2404927121. [PMID: 39541348 PMCID: PMC11588065 DOI: 10.1073/pnas.2404927121] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2024] [Accepted: 09/21/2024] [Indexed: 11/16/2024] Open
Abstract
Current evidence suggests that ontogeny may account for the functional heterogeneity of some tissue macrophages, but not others. Here, we asked whether developmental origin drives different functions of skin Langerhans cells (LCs), an embryo-derived mononuclear phagocyte with features of both tissue macrophages and dendritic cells. Using time-course analyses, bone marrow chimeras, and fate tracing models, we found that the complete elimination of embryo-derived LCs at steady state results in their repopulation from circulating monocytes. However, monocyte-derived LCs inefficiently replenished the epidermal niche. Instead, these cells preferentially migrated to skin-draining lymph nodes. Mechanistically, we show that the enhanced migratory capability of monocyte-derived LCs is associated with higher expression of CD207/Langerin, a C-type lectin involved in the capture of skin microbes. Our data demonstrate that ontogeny plays a role in the migratory behavior of epidermal LCs.
Collapse
Affiliation(s)
- Hayley M. Raquer-McKay
- Microbiology and Immunology Department, Stanford University School of Medicine, Stanford, CA94305
- Immunology Program, Stanford University School of Medicine, Stanford, CA94304
| | - Raul A. Maqueda-Alfaro
- Pharmacology Department, School of Medicine, University of California San Diego, La Jolla, CA92093
| | - Sanjana Saravanan
- Microbiology and Immunology Department, Stanford University School of Medicine, Stanford, CA94305
- Immunology Program, Stanford University School of Medicine, Stanford, CA94304
| | - Rebeca Arroyo Hornero
- Microbiology and Immunology Department, Stanford University School of Medicine, Stanford, CA94305
- Immunology Program, Stanford University School of Medicine, Stanford, CA94304
| | - Björn E. Clausen
- Institute for Molecular Medicine, Paul Klein Center for Immune Intervention, University Medical Center of the Johannes Gutenberg-University Mainz, Mainz55131, Germany
- Research Center for Immunotherapy (Forschungs-Zentrum für Immuntherapie), University Medical Center of the Johannes Gutenberg-University Mainz, Mainz55131, Germany
| | - Andres Gottfried-Blackmore
- Pharmacology Department, School of Medicine, University of California San Diego, La Jolla, CA92093
- Department of Medicine, Division of Gastroenterology, University of California San Diego, La Jolla, CA92093
- Veterans Affairs San Diego Healthcare System, Gastroenterology Section, La Jolla, CA92161
| | - Juliana Idoyaga
- Microbiology and Immunology Department, Stanford University School of Medicine, Stanford, CA94305
- Immunology Program, Stanford University School of Medicine, Stanford, CA94304
- Pharmacology Department, School of Medicine, University of California San Diego, La Jolla, CA92093
- Molecular Biology Department, School of Biological Sciences, University of California San Diego, La Jolla, CA92093
| |
Collapse
|
3
|
Bayerl F, Bejarano DA, Bertacchi G, Doffin AC, Gobbini E, Hubert M, Li L, Meiser P, Pedde AM, Posch W, Rupp L, Schlitzer A, Schmitz M, Schraml BU, Uderhardt S, Valladeau-Guilemond J, Wilflingseder D, Zaderer V, Böttcher JP. Guidelines for visualization and analysis of DC in tissues using multiparameter fluorescence microscopy imaging methods. Eur J Immunol 2023; 53:e2249923. [PMID: 36623939 DOI: 10.1002/eji.202249923] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2022] [Revised: 11/07/2022] [Accepted: 11/14/2022] [Indexed: 01/11/2023]
Abstract
This article is part of the Dendritic Cell Guidelines article series, which provides a collection of state-of-the-art protocols for the preparation, phenotype analysis by flow cytometry, generation, fluorescence microscopy, and functional characterization of mouse and human dendritic cells (DC) from lymphoid organs and various non-lymphoid tissues. Here, we provide detailed procedures for a variety of multiparameter fluorescence microscopy imaging methods to explore the spatial organization of DC in tissues and to dissect how DC migrate, communicate, and mediate their multiple functional roles in immunity in a variety of tissue settings. The protocols presented here entail approaches to study DC dynamics and T cell cross-talk by intravital microscopy, large-scale visualization, identification, and quantitative analysis of DC subsets and their functions by multiparameter fluorescence microscopy of fixed tissue sections, and an approach to study DC interactions with tissue cells in a 3D cell culture model. While all protocols were written by experienced scientists who routinely use them in their work, this article was also peer-reviewed by leading experts and approved by all co-authors, making it an essential resource for basic and clinical DC immunologists.
Collapse
Affiliation(s)
- Felix Bayerl
- Institute of Molecular Immunology, School of Medicine, Klinikum Rechts der Isar, Technical University of Munich (TUM), Ismaninger Str. 22, Munich, Germany
| | - David A Bejarano
- Quantitative Systems Biology, Life and Medical Sciences (LIMES) Institute, University of Bonn, Germany
| | - Giulia Bertacchi
- Institute of Hygiene and Medical Microbiology, Medical University of Innsbruck, Innsbruck, Austria
| | - Anne-Claire Doffin
- Cancer Research Center Lyon, UMR INSERM 1052 CNRS 5286, Centre Léon Bérard, 28 rue Laennec, Lyon, France
| | - Elisa Gobbini
- Cancer Research Center Lyon, UMR INSERM 1052 CNRS 5286, Centre Léon Bérard, 28 rue Laennec, Lyon, France
| | - Margaux Hubert
- Cancer Research Center Lyon, UMR INSERM 1052 CNRS 5286, Centre Léon Bérard, 28 rue Laennec, Lyon, France
| | - Lijian Li
- Department of Internal Medicine 3 - Rheumatology and Immunology, Friedrich-Alexander University Erlangen-Nürnberg (FAU) and Universitätsklinikum Erlangen, Erlangen, Germany
- Deutsches Zentrum für Immuntherapie (DZI), Friedrich-Alexander University Erlangen-Nürnberg (FAU) and Universitätsklinikum Erlangen, Erlangen, Germany
- Exploratory Research Unit, Optical Imaging Centre Erlangen (OICE), Friedrich-Alexander University Erlangen-Nürnberg (FAU), Erlangen, Germany
| | - Philippa Meiser
- Institute of Molecular Immunology, School of Medicine, Klinikum Rechts der Isar, Technical University of Munich (TUM), Ismaninger Str. 22, Munich, Germany
| | - Anna-Marie Pedde
- Institute of Molecular Immunology, School of Medicine, Klinikum Rechts der Isar, Technical University of Munich (TUM), Ismaninger Str. 22, Munich, Germany
| | - Wilfried Posch
- Institute of Hygiene and Medical Microbiology, Medical University of Innsbruck, Innsbruck, Austria
| | - Luise Rupp
- Institute of Immunology, Faculty of Medicine Carl Gustav Carus, TU Dresden, Dresden, Germany
| | - Andreas Schlitzer
- Quantitative Systems Biology, Life and Medical Sciences (LIMES) Institute, University of Bonn, Germany
| | - Marc Schmitz
- Institute of Immunology, Faculty of Medicine Carl Gustav Carus, TU Dresden, Dresden, Germany
- National Center for Tumor Diseases (NCT), Partner Site Dresden, Dresden, Germany
- German Cancer Consortium (DKTK), Partner Site Dresden, and German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Barbara U Schraml
- Walter-Brendel-Centre of Experimental Medicine, University Hospital, LMU Munich, Planegg-Martinsried, Germany
- Biomedical Center, Institute for Cardiovascular Physiology and Pathophysiology, Faculty of Medicine, LMU Munich, Planegg-Martinsried, Germany
| | - Stefan Uderhardt
- Department of Internal Medicine 3 - Rheumatology and Immunology, Friedrich-Alexander University Erlangen-Nürnberg (FAU) and Universitätsklinikum Erlangen, Erlangen, Germany
- Deutsches Zentrum für Immuntherapie (DZI), Friedrich-Alexander University Erlangen-Nürnberg (FAU) and Universitätsklinikum Erlangen, Erlangen, Germany
- Exploratory Research Unit, Optical Imaging Centre Erlangen (OICE), Friedrich-Alexander University Erlangen-Nürnberg (FAU), Erlangen, Germany
| | - Jenny Valladeau-Guilemond
- Cancer Research Center Lyon, UMR INSERM 1052 CNRS 5286, Centre Léon Bérard, 28 rue Laennec, Lyon, France
| | - Doris Wilflingseder
- Institute of Hygiene and Medical Microbiology, Medical University of Innsbruck, Innsbruck, Austria
| | - Viktoria Zaderer
- Institute of Hygiene and Medical Microbiology, Medical University of Innsbruck, Innsbruck, Austria
| | - Jan P Böttcher
- Institute of Molecular Immunology, School of Medicine, Klinikum Rechts der Isar, Technical University of Munich (TUM), Ismaninger Str. 22, Munich, Germany
| |
Collapse
|
4
|
The Roles of Skin Langerhans Cells in Immune Tolerance and Cancer Immunity. Vaccines (Basel) 2022; 10:vaccines10091380. [PMID: 36146458 PMCID: PMC9503294 DOI: 10.3390/vaccines10091380] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2022] [Revised: 08/14/2022] [Accepted: 08/19/2022] [Indexed: 12/19/2022] Open
Abstract
Langerhans cells (LC) are a unique population of tissue-resident macrophages with dendritic cell (DC) functionality that form a network of cells across the epidermis of the skin. Their location at the skin barrier suggests an important role for LC as immune sentinels at the skin surface. The classification of LC as DC over the past few decades has driven the scientific community to extensively study how LC function as DC-like cells that prime T cell immunity. However, LC are a unique type of tissue-resident macrophages, and recent evidence also supports an immunoregulatory role of LC at steady state and during specific inflammatory conditions, highlighting the impact of cutaneous environment in shaping LC functionality. In this mini review, we discuss the recent literature on the immune tolerance function of LC in homeostasis and disease conditions, including malignant transformation and progression; as well as LC functional plasticity for adaption to microenvironmental cues and the potential connection between LC population heterogeneity and functional diversity. Future investigation into the molecular mechanisms that LC use to integrate different microenvironment cues and adapt immunological responses for controlling LC functional plasticity is needed for future breakthroughs in tumor immunology, vaccine development, and treatments for inflammatory skin diseases.
Collapse
|
5
|
Dalod M, Scheu S. Dendritic cell functions in vivo: a user's guide to current and next generation mutant mouse models. Eur J Immunol 2022; 52:1712-1749. [PMID: 35099816 DOI: 10.1002/eji.202149513] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2021] [Accepted: 01/14/2022] [Indexed: 11/11/2022]
Abstract
Dendritic cells (DCs) do not just excel in antigen presentation. They orchestrate information transfer from innate to adaptive immunity, by sensing and integrating a variety of danger signals, and translating them to naïve T cells, to mount specifically tailored immune responses. This is accomplished by distinct DC types specialized in different functions and because each DC is functionally plastic, assuming different activation states depending on the input signals received. Mouse models hold the key to untangle this complexity and determine which DC types and activation states contribute to which functions. Here, we aim to provide comprehensive information for selecting the most appropriate mutant mouse strains to address specific research questions on DCs, considering three in vivo experimental approaches: (i) interrogating the roles of DC types through their depletion; (ii) determining the underlying mechanisms by specific genetic manipulations; (iii) deciphering the spatiotemporal dynamics of DC responses. We summarize the advantages, caveats, suggested use and perspectives for a variety of mutant mouse strains, discussing in more detail the most widely used or accurate models. Finally, we discuss innovative strategies to improve targeting specificity, for the next generation mutant mouse models, and briefly address how humanized mouse models can accelerate translation into the clinic. This article is protected by copyright. All rights reserved.
Collapse
Affiliation(s)
- Marc Dalod
- CNRS, Inserm, Aix Marseille Univ, Centre d'Immunologie de Marseille-Luminy (CIML), Turing Center for Living Systems, Marseille, France
| | - Stefanie Scheu
- Institute of Medical Microbiology and Hospital Hygiene, University of Düsseldorf, Düsseldorf, Germany
| |
Collapse
|
6
|
Murray MP, Crosby CM, Marcovecchio P, Hartmann N, Chandra S, Zhao M, Khurana A, Zahner SP, Clausen BE, Coleman FT, Mizgerd JP, Mikulski Z, Kronenberg M. Stimulation of a subset of natural killer T cells by CD103 + DC is required for GM-CSF and protection from pneumococcal infection. Cell Rep 2022; 38:110209. [PMID: 35021099 DOI: 10.1016/j.celrep.2021.110209] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2021] [Revised: 11/17/2021] [Accepted: 12/13/2021] [Indexed: 11/18/2022] Open
Abstract
Innate-like T cells, including invariant natural killer T cells, mucosal-associated invariant T cells, and γδ T cells, are present in various barrier tissues, including the lung, where they carry out protective responses during infections. Here, we investigate their roles during pulmonary pneumococcal infection. Following infection, innate-like T cells rapidly increase in lung tissue, in part through recruitment, but T cell antigen receptor activation and cytokine production occur mostly in interleukin-17-producing NKT17 and γδ T cells. NKT17 cells are preferentially located within lung tissue prior to infection, as are CD103+ dendritic cells, which are important both for antigen presentation to NKT17 cells and γδ T cell activation. Whereas interleukin-17-producing γδ T cells are numerous, granulocyte-macrophage colony-stimulating factor is exclusive to NKT17 cells and is required for optimal protection. These studies demonstrate how particular cellular interactions and responses of functional subsets of innate-like T cells contribute to protection from pathogenic lung infection.
Collapse
Affiliation(s)
- Mallory Paynich Murray
- Division of Developmental Immunology, La Jolla Institute for Immunology, La Jolla, CA 92037, USA
| | - Catherine M Crosby
- Division of Developmental Immunology, La Jolla Institute for Immunology, La Jolla, CA 92037, USA
| | - Paola Marcovecchio
- Microscopy and Histology Core Facility, La Jolla Institute for Immunology, La Jolla, CA 92037, USA
| | - Nadine Hartmann
- Division of Developmental Immunology, La Jolla Institute for Immunology, La Jolla, CA 92037, USA
| | - Shilpi Chandra
- Division of Developmental Immunology, La Jolla Institute for Immunology, La Jolla, CA 92037, USA
| | - Meng Zhao
- Division of Developmental Immunology, La Jolla Institute for Immunology, La Jolla, CA 92037, USA
| | - Archana Khurana
- Division of Developmental Immunology, La Jolla Institute for Immunology, La Jolla, CA 92037, USA
| | - Sonja P Zahner
- Division of Developmental Immunology, La Jolla Institute for Immunology, La Jolla, CA 92037, USA
| | - Björn E Clausen
- Institute for Molecular Medicine, University Medical Center of the Johannes Gutenberg-University Mainz, Mainz 55131, Germany
| | - Fadie T Coleman
- Pulmonary Center, Boston University School of Medicine, Boston, MA 02118, USA
| | - Joseph P Mizgerd
- Pulmonary Center, Boston University School of Medicine, Boston, MA 02118, USA
| | - Zbigniew Mikulski
- Microscopy and Histology Core Facility, La Jolla Institute for Immunology, La Jolla, CA 92037, USA
| | - Mitchell Kronenberg
- Division of Developmental Immunology, La Jolla Institute for Immunology, La Jolla, CA 92037, USA; Division of Biological Sciences, University of California, San Diego, La Jolla, CA 92037, USA.
| |
Collapse
|
7
|
Kawakubo A, Miyagi M, Yokozeki Y, Nakawaki M, Takano S, Satoh M, Itakura M, Inoue G, Takaso M, Uchida K. Origin of M2 Mϕ and its macrophage polarization by TGF-β in a mice intervertebral injury model. Int J Immunopathol Pharmacol 2022; 36:3946320221103792. [PMID: 35592891 PMCID: PMC9174651 DOI: 10.1177/03946320221103792] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
Introduction Studies have identified the presence of M1 and M2 macrophages (Mϕ) in injured intervertebral discs (IVDs). However, the origin and polarization-regulatory factor of M2 Mϕ are not fully understood. TGF-β is a regulatory factor for M2 polarization in several tissues. Here, we investigated the source of M2 Mϕ and the role of TGF-β on M2 polarization using a mice disc-puncture injury model. Methods To investigate the origin of M2 macrophages, 30 GFP chimeric mice were created by bone marrow transplantation. IVDs were obtained from both groups on pre-puncture (control) and post-puncture days 1, 3, 7, and 14 and CD86 (M1 marker)- and CD206 (M2 marker)-positive cells evaluated by flow cytometry (n = 5 at each time point). To investigate the role of TGF-β on M2 polarization, TGF-β inhibitor (SB431542) was also injected on post-puncture days (PPD) 5 and 6 and CD206 expression was evaluated on day 7 by flow cytometry (n = 5) and real time PCR (n = 10). Results The proportion of CD86+ Mϕ within the GFP+ population was significantly increased at PPD 1, 3, 7, and 14 compared to control. CD206-positive cells in GFP-populations were significantly increased on PPD 7 and 14. In addition, the percentage of CD206-positive cells was significantly higher in GFP-populations than in GFP+ populations. TGF-β inhibitor reduced CD206-positive cells and Cd206 expression at 7 days after puncture. Conclusion Our findings suggest that M2 Mϕ following IVD injury may originate from resident Mϕ. TGF-β is a key factor for M2 polarization of macrophages following IVD injury.
Collapse
Affiliation(s)
- Ayumu Kawakubo
- Department of Orthopedic Surgery, 38088Kitasato University School of Medicine, Sagamihara, Japan
| | - Masayuki Miyagi
- Department of Orthopedic Surgery, 38088Kitasato University School of Medicine, Sagamihara, Japan
| | - Yuji Yokozeki
- Department of Orthopedic Surgery, 38088Kitasato University School of Medicine, Sagamihara, Japan
| | - Mitsufumi Nakawaki
- Department of Orthopedic Surgery, 38088Kitasato University School of Medicine, Sagamihara, Japan
| | - Shotaro Takano
- Department of Orthopedic Surgery, 38088Kitasato University School of Medicine, Sagamihara, Japan
| | - Masashi Satoh
- Department of Immunology, 38088Kitasato University School of Medicine, Sagamihara, Japan
| | - Makoto Itakura
- Department of Biochemistry, 38088Kitasato University School of Medicine, Sagamihara, Japan
| | - Gen Inoue
- Department of Orthopedic Surgery, 38088Kitasato University School of Medicine, Sagamihara, Japan
| | - Masashi Takaso
- Department of Orthopedic Surgery, 38088Kitasato University School of Medicine, Sagamihara, Japan
| | - Kentaro Uchida
- Department of Orthopedic Surgery, 38088Kitasato University School of Medicine, Sagamihara, Japan
| |
Collapse
|
8
|
Lopez Kostka S, Kautz-Neu K, Yogev N, Lukas D, Holzmann B, Waisman A, Clausen BE, von Stebut E. Exclusive Expression of MyD88 on Dendritic Cells Is Lopez Kostka Sufficient to Induce Protection against Experimental Leishmaniasis. J Invest Dermatol 2021; 142:1230-1233. [PMID: 34570998 DOI: 10.1016/j.jid.2021.07.184] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2021] [Revised: 06/28/2021] [Accepted: 07/31/2021] [Indexed: 10/20/2022]
Affiliation(s)
- Susanna Lopez Kostka
- Department of Dermatology and Venereology, University Medical Center, Johannes Gutenberg University Mainz, Mainz, Germany
| | - Kordula Kautz-Neu
- Department of Dermatology and Venereology, University Medical Center, Johannes Gutenberg University Mainz, Mainz, Germany
| | - Nir Yogev
- Department of Dermatology and Venereology, Faculty of Medicine, University of Cologne, Cologne, Germany
| | - Dominika Lukas
- Department of Dermatology and Venereology, Faculty of Medicine, University of Cologne, Cologne, Germany
| | - Bernhard Holzmann
- Department of Surgery, Faculty of Medicine, Technische Universität München, Munich, Germany
| | - Ari Waisman
- Institute for Molecular Medicine Mainz, University Medical Center, Johannes Gutenberg University Mainz, Mainz, Germany
| | - Björn E Clausen
- Institute for Molecular Medicine Mainz, University Medical Center, Johannes Gutenberg University Mainz, Mainz, Germany
| | - Esther von Stebut
- Department of Dermatology and Venereology, Faculty of Medicine, University of Cologne, Cologne, Germany.
| |
Collapse
|
9
|
Yokozeki Y, Kawakubo A, Miyagi M, Kuroda A, Sekiguchi H, Inoue G, Takaso M, Uchida K. Reduced TGF- β Expression and CD206-Positive Resident Macrophages in the Intervertebral Discs of Aged Mice. BIOMED RESEARCH INTERNATIONAL 2021; 2021:7988320. [PMID: 34337052 PMCID: PMC8289593 DOI: 10.1155/2021/7988320] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/05/2021] [Accepted: 07/03/2021] [Indexed: 11/18/2022]
Abstract
Age is a key factor in intervertebral disc (IVD) degeneration; however, the changes that occur in IVDs with age are not fully understood. Tissue-resident macrophages are critical for tissue homeostasis and are regulated by transforming growth factor- (TGF-) β. We examined changes in the proportion of resident macrophages in young versus aged mice and the role of TGF-β in regulating resident macrophages in IVDs. IVDs were harvested from 4-month (young) and 18-month-old (aged) C57BL/6J mice. The proportion of macrophages in IVDs was determined using flow cytometry (n = 5 for each time point) and the expression of Cd11b, Cd206, and Tgfb genes, which encode CD11b, CD206, and TGF-β protein, respectively, using real-time PCR. To study the role of TGF-β in the polarization of resident macrophages, resident macrophages isolated from IVDs from young and aged mice were treated with recombinant TGF-β with and without a TGF-β inhibitor (SB431542). Additionally, SB431542 was intraperitoneally injected into young and aged mice, and Cd206 expression was examined using real-time PCR (n = 10 for each time point). The proportion of CD11b+ and CD11b+ CD206+ cells was significantly reduced in aged versus young mice, as was Cd11b, Cd206, and Tgfb expression. TGF-β/IL10 stimulation significantly increased the expression of Cd206, an M2 macrophage marker, in disc macrophages from both young and aged mice. Meanwhile, administration of a TGF-β inhibitor significantly reduced Cd206 expression compared to vehicle control in both groups. Conclusion. Resident macrophages decrease with age in IVDs, which may be associated with the concomitant decrease in TGF-β. Our findings provide new insight into the mechanisms of age-related IVD pathology.
Collapse
Affiliation(s)
- Yuji Yokozeki
- Department of Orthopedic Surgery, Kitasato University School of Medicine, 1-15-1 Minami-ku Kitasato, Sagamihara City, Kanagawa 252-0374, Japan
| | - Ayumu Kawakubo
- Department of Orthopedic Surgery, Kitasato University School of Medicine, 1-15-1 Minami-ku Kitasato, Sagamihara City, Kanagawa 252-0374, Japan
| | - Masayuki Miyagi
- Department of Orthopedic Surgery, Kitasato University School of Medicine, 1-15-1 Minami-ku Kitasato, Sagamihara City, Kanagawa 252-0374, Japan
| | - Akiyoshi Kuroda
- Department of Orthopedic Surgery, Kitasato University School of Medicine, 1-15-1 Minami-ku Kitasato, Sagamihara City, Kanagawa 252-0374, Japan
| | - Hiroyuki Sekiguchi
- Shonan University of Medical Sciences Research Institute, Nishikubo 500, Chigasaki City, Kanagawa 253-0083, Japan
| | - Gen Inoue
- Department of Orthopedic Surgery, Kitasato University School of Medicine, 1-15-1 Minami-ku Kitasato, Sagamihara City, Kanagawa 252-0374, Japan
| | - Masashi Takaso
- Department of Orthopedic Surgery, Kitasato University School of Medicine, 1-15-1 Minami-ku Kitasato, Sagamihara City, Kanagawa 252-0374, Japan
| | - Kentaro Uchida
- Department of Orthopedic Surgery, Kitasato University School of Medicine, 1-15-1 Minami-ku Kitasato, Sagamihara City, Kanagawa 252-0374, Japan
- Shonan University of Medical Sciences Research Institute, Nishikubo 500, Chigasaki City, Kanagawa 253-0083, Japan
| |
Collapse
|
10
|
Lelios I, Stifter SA, Cecconi V, Petrova E, Lutz M, Cansever D, Utz SG, Becher B, van den Broek M, Greter M. Monocytes promote UV-induced epidermal carcinogenesis. Eur J Immunol 2021; 51:1799-1808. [PMID: 33759186 PMCID: PMC8359952 DOI: 10.1002/eji.202048841] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2020] [Revised: 02/03/2021] [Accepted: 03/16/2021] [Indexed: 11/10/2022]
Abstract
Mononuclear phagocytes consisting of monocytes, macrophages, and DCs play a complex role in tumor development by either promoting or restricting tumor growth. Cutaneous squamous cell carcinoma (cSCC) is the second most common nonmelanoma skin cancer arising from transformed epidermal keratinocytes. While present at high numbers, the role of tumor-infiltrating and resident myeloid cells in the formation of cSCC is largely unknown. Using transgenic mice and depleting antibodies to eliminate specific myeloid cell types in the skin, we investigated the involvement of mononuclear phagocytes in the development of UV-induced cSCC in K14-HPV8-E6 transgenic mice. Although resident Langerhans cells were enriched in the tumor, their contribution to tumor formation was negligible. Equally, dermal macrophages were dispensable for the development of cSCC. In contrast, mice lacking circulating monocytes were completely resistant to UV-induced cSCC, indicating that monocytes promote tumor development. Collectively, these results demonstrate a critical role for classical monocytes in the initiation of skin cancer.
Collapse
Affiliation(s)
- Iva Lelios
- Institute of Experimental Immunology, University of Zurich, Zurich, Switzerland
| | - Sebastian A Stifter
- Institute of Experimental Immunology, University of Zurich, Zurich, Switzerland
| | - Virginia Cecconi
- Institute of Experimental Immunology, University of Zurich, Zurich, Switzerland.,Comprehensive Cancer Center Zurich, Zurich, Switzerland
| | - Ekaterina Petrova
- Institute of Experimental Immunology, University of Zurich, Zurich, Switzerland
| | - Mirjam Lutz
- Institute of Experimental Immunology, University of Zurich, Zurich, Switzerland
| | - Dilay Cansever
- Institute of Experimental Immunology, University of Zurich, Zurich, Switzerland
| | - Sebastian G Utz
- Institute of Experimental Immunology, University of Zurich, Zurich, Switzerland
| | - Burkhard Becher
- Institute of Experimental Immunology, University of Zurich, Zurich, Switzerland.,Comprehensive Cancer Center Zurich, Zurich, Switzerland
| | - Maries van den Broek
- Institute of Experimental Immunology, University of Zurich, Zurich, Switzerland.,Comprehensive Cancer Center Zurich, Zurich, Switzerland
| | - Melanie Greter
- Institute of Experimental Immunology, University of Zurich, Zurich, Switzerland
| |
Collapse
|
11
|
Anderson DA, Dutertre CA, Ginhoux F, Murphy KM. Genetic models of human and mouse dendritic cell development and function. Nat Rev Immunol 2021; 21:101-115. [PMID: 32908299 PMCID: PMC10955724 DOI: 10.1038/s41577-020-00413-x] [Citation(s) in RCA: 176] [Impact Index Per Article: 44.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/28/2020] [Indexed: 12/13/2022]
Abstract
Dendritic cells (DCs) develop in the bone marrow from haematopoietic progenitors that have numerous shared characteristics between mice and humans. Human counterparts of mouse DC progenitors have been identified by their shared transcriptional signatures and developmental potential. New findings continue to revise models of DC ontogeny but it is well accepted that DCs can be divided into two main functional groups. Classical DCs include type 1 and type 2 subsets, which can detect different pathogens, produce specific cytokines and present antigens to polarize mainly naive CD8+ or CD4+ T cells, respectively. By contrast, the function of plasmacytoid DCs is largely innate and restricted to the detection of viral infections and the production of type I interferon. Here, we discuss genetic models of mouse DC development and function that have aided in correlating ontogeny with function, as well as how these findings can be translated to human DCs and their progenitors.
Collapse
Affiliation(s)
- David A Anderson
- Department of Pathology and Immunology, School of Medicine, Washington University in St. Louis, St. Louis, MO, USA
| | | | - Florent Ginhoux
- Singapore Immunology Network, Agency for Science, Technology and Research, Singapore, Singapore
- Shanghai Institute of Immunology, Department of Immunology and Microbiology, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Translational Immunology Institute, SingHealth Duke-NUS Academic Medical Centre, Singapore, Singapore
| | - Kenneth M Murphy
- Department of Pathology and Immunology, School of Medicine, Washington University in St. Louis, St. Louis, MO, USA.
- Howard Hughes Medical Institute, School of Medicine, Washington University in St. Louis, St. Louis, MO, USA.
| |
Collapse
|
12
|
Vroman H, Uden D, Bergen IM, Hulst JAC, Lukkes M, Loo G, Clausen BE, Boon L, Lambrecht BN, Hammad H, Hendriks RW, Kool M. Tnfaip3 expression in pulmonary conventional type 1 Langerin-expressing dendritic cells regulates T helper 2-mediated airway inflammation in mice. Allergy 2020; 75:2587-2598. [PMID: 32329078 PMCID: PMC7687104 DOI: 10.1111/all.14334] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2019] [Revised: 03/10/2020] [Accepted: 03/23/2020] [Indexed: 12/14/2022]
Abstract
Background Conventional type 1 dendritic cells (cDC1s) control anti‐viral and anti‐tumor immunity by inducing antigen‐specific cytotoxic CD8+ T‐cell responses. Controversy exists whether cDC1s also control CD4+ T helper 2 (Th2) cell responses, since suppressive and activating roles have been reported. DC activation status, controlled by the transcription factor NF‐κB, might determine the precise outcome of Th‐cell differentiation upon encounter with cDC1s. To investigate the role of activated cDC1s in Th2‐driven immune responses, pulmonary cDC1s were activated by targeted deletion of A20/Tnfaip3, a negative regulator of NF‐κB signaling. Methods To target pulmonary cDC1s, Cd207 (Langerin)‐mediated excision of A20/Tnfaip3 was used, generating Tnfaip3fl/flxCd207+/cre (Tnfaip3Lg‐KO) mice. Mice were exposed to house dust mite (HDM) to provoke Th2‐mediated immune responses. Results Mice harboring Tnfaip3‐deficient cDC1s did not develop Th2‐driven eosinophilic airway inflammation upon HDM exposure, but rather showed elevated numbers of IFNγ‐expressing CD8+ T cells. In addition, Tnfaip3Lg‐KO mice harbored increased numbers of IL‐12–expressing cDC1s and elevated PD‐L1 expression in all pulmonary DC subsets. Blocking either IL‐12 or IFNγ in Tnfaip3Lg‐KO mice restored Th2 responses, whereas administration of recombinant IFNγ during HDM sensitization in C57Bl/6 mice blocked Th2 development. Conclusions These findings indicate that the activation status of cDC1s, shown by their specific expression of co‐inhibitory molecules and cytokines, critically contributes to the development of Th2 cell–mediated disorders, most likely by influencing IFNγ production in CD8+ T cells.
Collapse
Affiliation(s)
- Heleen Vroman
- Department of Pulmonary Medicine Erasmus MC Rotterdam The Netherlands
- VIB Center for Inflammation Research Ghent Belgium
| | - Denise Uden
- Department of Pulmonary Medicine Erasmus MC Rotterdam The Netherlands
| | - Ingrid M. Bergen
- Department of Pulmonary Medicine Erasmus MC Rotterdam The Netherlands
| | | | - Melanie Lukkes
- Department of Pulmonary Medicine Erasmus MC Rotterdam The Netherlands
| | - Geert Loo
- VIB Center for Inflammation Research Ghent Belgium
- Department of Biomedical Molecular Biology Ghent University Ghent Belgium
| | - Björn E. Clausen
- Institute for Molecular Medicine University Medical Center of the Johannes Gutenberg‐University Mainz Mainz Germany
| | | | - Bart N. Lambrecht
- Department of Pulmonary Medicine Erasmus MC Rotterdam The Netherlands
- VIB Center for Inflammation Research Ghent Belgium
- Department of Respiratory Medicine Ghent University Ghent Belgium
| | - Hamida Hammad
- VIB Center for Inflammation Research Ghent Belgium
- Department of Internal Medicine Ghent University Ghent Belgium
| | - Rudi W. Hendriks
- Department of Pulmonary Medicine Erasmus MC Rotterdam The Netherlands
| | - Mirjam Kool
- Department of Pulmonary Medicine Erasmus MC Rotterdam The Netherlands
| |
Collapse
|
13
|
Horev Y, Salameh R, Nassar M, Capucha T, Saba Y, Barel O, Zubeidat K, Matanes D, Leibovich A, Heyman O, Eli-Berchoer L, Hanhan S, Betser-Cohen G, Shapiro H, Elinav E, Bercovier H, Wilensky A, Hovav AH. Niche rather than origin dysregulates mucosal Langerhans cells development in aged mice. Mucosal Immunol 2020; 13:767-776. [PMID: 32457449 DOI: 10.1038/s41385-020-0301-y] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2020] [Revised: 04/02/2020] [Accepted: 04/19/2020] [Indexed: 02/04/2023]
Abstract
Unlike epidermal Langerhans cells (LCs) that originate from embryonic precursors and are self-renewed locally, mucosal LCs arise and are replaced by circulating bone marrow (BM) precursors throughout life. While the unique lifecycle of epidermal LCs is associated with an age-dependent decrease in their numbers, whether and how aging has an impact on mucosal LCs remains unclear. Focusing on gingival LCs we found that mucosal LCs are reduced with age but exhibit altered morphology with that observed in aged epidermal LCs. The reduction of gingival but not epidermal LCs in aged mice was microbiota-dependent; nevertheless, the impact of the microbiota on gingival LCs was indirect. We next compared the ability of young and aged BM precursors to differentiate to mucosal LCs. Mixed BM chimeras, as well as differentiation cultures, demonstrated that aged BM has intact if not superior capacity to differentiate into LCs than young BM. This was in line with the higher percentages of mucosal LC precursors, pre-DCs, and monocytes, detected in aged BM. These findings suggest that while aging is associated with reduced LC numbers, the niche rather than the origin controls this process in mucosal barriers.
Collapse
Affiliation(s)
- Yael Horev
- The Institute of Dental Sciences, Faculty of Dental Medicine, Hebrew University, Jerusalem, Israel.,Department of Periodontology, Faculty of Dental Medicine, Hebrew University-Hadassah Medical Center, Jerusalem, Israel
| | - Rana Salameh
- The Institute of Dental Sciences, Faculty of Dental Medicine, Hebrew University, Jerusalem, Israel
| | - Maria Nassar
- The Institute of Dental Sciences, Faculty of Dental Medicine, Hebrew University, Jerusalem, Israel
| | - Tal Capucha
- The Institute of Dental Sciences, Faculty of Dental Medicine, Hebrew University, Jerusalem, Israel
| | - Yasmin Saba
- The Institute of Dental Sciences, Faculty of Dental Medicine, Hebrew University, Jerusalem, Israel
| | - Or Barel
- The Institute of Dental Sciences, Faculty of Dental Medicine, Hebrew University, Jerusalem, Israel
| | - Khaled Zubeidat
- The Institute of Dental Sciences, Faculty of Dental Medicine, Hebrew University, Jerusalem, Israel
| | - Daniela Matanes
- The Institute of Dental Sciences, Faculty of Dental Medicine, Hebrew University, Jerusalem, Israel
| | - Amit Leibovich
- The Institute of Dental Sciences, Faculty of Dental Medicine, Hebrew University, Jerusalem, Israel
| | - Oded Heyman
- Department of Periodontology, Faculty of Dental Medicine, Hebrew University-Hadassah Medical Center, Jerusalem, Israel
| | - Luba Eli-Berchoer
- The Institute of Dental Sciences, Faculty of Dental Medicine, Hebrew University, Jerusalem, Israel
| | - Salem Hanhan
- The Institute of Dental Sciences, Faculty of Dental Medicine, Hebrew University, Jerusalem, Israel
| | - Gili Betser-Cohen
- The Lautenberg Center for Immunology and Cancer Research, Faculty of Medicine, Hebrew University, Jerusalem, Israel
| | - Hagit Shapiro
- Department of Immunology, Weizmann Institute of Science, Rehovot, Israel
| | - Eran Elinav
- Department of Immunology, Weizmann Institute of Science, Rehovot, Israel
| | - Herve Bercovier
- Department of Microbiology and Molecular Genetics, Faculty of Medicine, Hebrew University, Jerusalem, Israel
| | - Asaf Wilensky
- Department of Periodontology, Faculty of Dental Medicine, Hebrew University-Hadassah Medical Center, Jerusalem, Israel.
| | - Avi-Hai Hovav
- The Institute of Dental Sciences, Faculty of Dental Medicine, Hebrew University, Jerusalem, Israel.
| |
Collapse
|
14
|
Tran S, Baba I, Poupel L, Dussaud S, Moreau M, Gélineau A, Marcelin G, Magréau-Davy E, Ouhachi M, Lesnik P, Boissonnas A, Le Goff W, Clausen BE, Yvan-Charvet L, Sennlaub F, Huby T, Gautier EL. Impaired Kupffer Cell Self-Renewal Alters the Liver Response to Lipid Overload during Non-alcoholic Steatohepatitis. Immunity 2020; 53:627-640.e5. [PMID: 32562600 DOI: 10.1016/j.immuni.2020.06.003] [Citation(s) in RCA: 218] [Impact Index Per Article: 43.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2019] [Revised: 04/14/2020] [Accepted: 05/29/2020] [Indexed: 02/06/2023]
Abstract
Kupffer cells (KCs) are liver-resident macrophages that self-renew by proliferation in the adult independently from monocytes. However, how they are maintained during non-alcoholic steatohepatitis (NASH) remains ill defined. We found that a fraction of KCs derived from Ly-6C+ monocytes during NASH, underlying impaired KC self-renewal. Monocyte-derived KCs (MoKCs) gradually seeded the KC pool as disease progressed in a response to embryo-derived KC (EmKC) death. Those MoKCs were partly immature and exhibited a pro-inflammatory status compared to EmKCs. Yet, they engrafted the KC pool for the long term as they remained following disease regression while acquiring mature EmKC markers. While KCs as a whole favored hepatic triglyceride storage during NASH, EmKCs promoted it more efficiently than MoKCs, and the latter exacerbated liver damage, highlighting functional differences among KCs with different origins. Overall, our data reveal that KC homeostasis is impaired during NASH, altering the liver response to lipids, as well as KC ontogeny.
Collapse
Affiliation(s)
- Sophie Tran
- Institut National de la Santé et de la Recherche Médicale (Inserm, UMR_S 1166), Sorbonne Université, Hôpital de la Pitié-Salpêtrière, Paris, France
| | - Ines Baba
- Institut National de la Santé et de la Recherche Médicale (Inserm, UMR_S 1166), Sorbonne Université, Hôpital de la Pitié-Salpêtrière, Paris, France
| | - Lucie Poupel
- Institut National de la Santé et de la Recherche Médicale (Inserm, UMR_S 1166), Sorbonne Université, Hôpital de la Pitié-Salpêtrière, Paris, France
| | - Sébastien Dussaud
- Institut National de la Santé et de la Recherche Médicale (Inserm, UMR_S 1166), Sorbonne Université, Hôpital de la Pitié-Salpêtrière, Paris, France
| | - Martine Moreau
- Institut National de la Santé et de la Recherche Médicale (Inserm, UMR_S 1166), Sorbonne Université, Hôpital de la Pitié-Salpêtrière, Paris, France
| | - Adélaïde Gélineau
- Institut National de la Santé et de la Recherche Médicale (Inserm, UMR_S 1166), Sorbonne Université, Hôpital de la Pitié-Salpêtrière, Paris, France
| | - Geneviève Marcelin
- Institut National de la Santé et de la Recherche Médicale (Inserm, UMR_S 1269), Sorbonne Université, Hôpital de la Pitié-Salpêtrière, Paris, France
| | - Elissa Magréau-Davy
- Institut National de la Santé et de la Recherche Médicale (Inserm, UMR_S 1166), Sorbonne Université, Hôpital de la Pitié-Salpêtrière, Paris, France
| | - Melissa Ouhachi
- Institut National de la Santé et de la Recherche Médicale (Inserm, UMR_S 1166), Sorbonne Université, Hôpital de la Pitié-Salpêtrière, Paris, France
| | - Philippe Lesnik
- Institut National de la Santé et de la Recherche Médicale (Inserm, UMR_S 1166), Sorbonne Université, Hôpital de la Pitié-Salpêtrière, Paris, France
| | - Alexandre Boissonnas
- Institut National de la Santé et de la Recherche Médicale (Inserm, UMR1135), Sorbonne Université, Centre National de la Recherche Scientifique (CNRS, ERL8255), Centre d'Immunologie et des Maladies Infectieuses CIMI, Paris, France
| | - Wilfried Le Goff
- Institut National de la Santé et de la Recherche Médicale (Inserm, UMR_S 1166), Sorbonne Université, Hôpital de la Pitié-Salpêtrière, Paris, France
| | - Björn E Clausen
- Institute for Molecular Medicine, University Medical Center of the Johannes Gutenberg-University Mainz, Mainz, Germany
| | - Laurent Yvan-Charvet
- Institut National de la Santé et de la Recherche Médicale (Inserm, U1065), Université Côte d'Azur, Centre Méditerranéen de Médecine Moléculaire (C3M), Atip-Avenir, Fédération Hospitalo-Universitaire (FHU) Oncoage, Nice, France
| | - Florian Sennlaub
- Institut de la Vision, Sorbonne Université, Institut National de la Santé et de la Recherche Médicale (Inserm), Centre National de la Recherche Scientifique (CNRS), Paris, France
| | - Thierry Huby
- Institut National de la Santé et de la Recherche Médicale (Inserm, UMR_S 1166), Sorbonne Université, Hôpital de la Pitié-Salpêtrière, Paris, France
| | - Emmanuel L Gautier
- Institut National de la Santé et de la Recherche Médicale (Inserm, UMR_S 1166), Sorbonne Université, Hôpital de la Pitié-Salpêtrière, Paris, France.
| |
Collapse
|
15
|
Selective AhR knockout in langerin-expressing cells abates Langerhans cells and polarizes Th2/Tr1 in epicutaneous protein sensitization. Proc Natl Acad Sci U S A 2020; 117:12980-12990. [PMID: 32461368 DOI: 10.1073/pnas.1917479117] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
The aryl hydrocarbon receptor (AhR) represents an environmental sensor regulating immune responses. In the skin, AhR is expressed in several cell types, including keratinocytes, epidermal Langerhans cells (LC), and dermal dendritic cells (DC). The mechanisms how AhR activates or inhibits cutaneous immune responses remain controversial, owing to differences in the cell-specific functions of AhR and the different activating ligands. Therefore, we sought to investigate the role of AhR in LC and langerin+ and negative DC in the skin. To this aim, we generated Langerin-specific and CD11c-specific knockout (-/-) mice lacking AhR, respectively, in LC and Langerin+ dermal DC and in all CD11c+ cells. These were then tested in an epicutaneous protein (ovalbumin, Ova) sensitization model. Immunofluorescence microscopy and flow cytometry revealed that Langerin-AhR-/- but not CD11c-AhR-/- mice harbored a decreased number of LC with fewer and stunted dendrites in the epidermis as well as a decreased number of LC in skin-draining lymph nodes (LN). Moreover, in the absence of AhR, we detected an enhanced T helper type-2 (Th2) [increased interleukin 5 (IL-5) and interleukin 13 (IL-13)] and T regulatory type-1 (Tr1) (IL-10) response when LN cells were challenged with Ova in vitro, though the number of regulatory T cells (Treg) in the LN remained comparable. Langerin-AhR-/- mice also exhibited increased blood levels of Ova-specific immunoglobulin E (IgE). In conclusion, deletion of AhR in langerin-expressing cells diminishes the number and activation of LC, while enhancing Th2 and Tr1 responses upon epicutaneous protein sensitization.
Collapse
|
16
|
Huang L, Li GH, Yu Q, Xu Y, Cvetkovski S, Wang X, Parajuli N, Udo-Inyang I, Kaplan D, Zhou L, Yao Z, Mi QS. Smad2/4 Signaling Pathway Is Critical for Epidermal Langerhans Cell Repopulation Under Inflammatory Condition but Not Required for Their Homeostasis at Steady State. Front Immunol 2020; 11:912. [PMID: 32457763 PMCID: PMC7221176 DOI: 10.3389/fimmu.2020.00912] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2019] [Accepted: 04/20/2020] [Indexed: 01/17/2023] Open
Abstract
Epidermal Langerhans cells (LCs) are skin-resident dendritic cells that are essential for the induction of skin immunity and tolerance. Transforming growth factor-β 1 (TGFβ1) is a crucial factor for LC maintenance and function. However, the underlying TGFβ1 signaling pathways remain unclear. Our previous research has shown that the TGFβ1/Smad3 signaling pathway does not impact LC homeostasis and maturation. In this study, we generated mice with conditional deletions of either individual Smad2, Smad4, or both Smad2 and Smad4 in the LC lineage or myeloid lineage, to further explore the impact of TGFβ1/Smad signaling pathways on LCs. We found that interruption of Smad2 or Smad4 individually or simultaneously in the LC lineage did not significantly impact the maintenance, maturation, antigen uptake, and migration of LCs in vivo or in vitro during steady state. However, the interruption of both Smad2 and Smad4 pathways in the myeloid lineage led to a dramatic inhibition of bone marrow-derived LCs in the inflammatory state. Overall, our data suggest that canonical TGFβ1/Smad2/4 signaling pathways are dispensable for epidermal LC homeostasis and maturation at steady state, but are critical for the long-term LC repopulation directly originating from the bone marrow in the inflammatory state.
Collapse
Affiliation(s)
- Linting Huang
- Department of Dermatology, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.,Center for Cutaneous Biology and Immunology, Department of Dermatology, Henry Ford Health System, Detroit, MI, United States.,Immunology Research Program, Henry Ford Cancer Institute, Henry Ford Health System, Detroit, MI, United States.,Institute of Dermatology, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Gui-Hua Li
- Center for Cutaneous Biology and Immunology, Department of Dermatology, Henry Ford Health System, Detroit, MI, United States.,Immunology Research Program, Henry Ford Cancer Institute, Henry Ford Health System, Detroit, MI, United States
| | - Qian Yu
- Center for Cutaneous Biology and Immunology, Department of Dermatology, Henry Ford Health System, Detroit, MI, United States.,Immunology Research Program, Henry Ford Cancer Institute, Henry Ford Health System, Detroit, MI, United States
| | - Yingping Xu
- Center for Cutaneous Biology and Immunology, Department of Dermatology, Henry Ford Health System, Detroit, MI, United States.,Immunology Research Program, Henry Ford Cancer Institute, Henry Ford Health System, Detroit, MI, United States
| | - Steven Cvetkovski
- Center for Cutaneous Biology and Immunology, Department of Dermatology, Henry Ford Health System, Detroit, MI, United States.,Immunology Research Program, Henry Ford Cancer Institute, Henry Ford Health System, Detroit, MI, United States
| | - Xuan Wang
- Center for Cutaneous Biology and Immunology, Department of Dermatology, Henry Ford Health System, Detroit, MI, United States.,Immunology Research Program, Henry Ford Cancer Institute, Henry Ford Health System, Detroit, MI, United States
| | - Nirmal Parajuli
- Center for Cutaneous Biology and Immunology, Department of Dermatology, Henry Ford Health System, Detroit, MI, United States.,Immunology Research Program, Henry Ford Cancer Institute, Henry Ford Health System, Detroit, MI, United States
| | - Imo Udo-Inyang
- Center for Cutaneous Biology and Immunology, Department of Dermatology, Henry Ford Health System, Detroit, MI, United States.,Immunology Research Program, Henry Ford Cancer Institute, Henry Ford Health System, Detroit, MI, United States
| | - Daniel Kaplan
- Department of Dermatology, University of Pittsburgh, Pittsburgh, PA, United States.,Department of Immunology, University of Pittsburgh, Pittsburgh, PA, United States
| | - Li Zhou
- Center for Cutaneous Biology and Immunology, Department of Dermatology, Henry Ford Health System, Detroit, MI, United States.,Immunology Research Program, Henry Ford Cancer Institute, Henry Ford Health System, Detroit, MI, United States.,Department of Internal Medicine, Henry Ford Health System, Detroit, MI, United States
| | - Zhirong Yao
- Department of Dermatology, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.,Institute of Dermatology, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Qing-Sheng Mi
- Center for Cutaneous Biology and Immunology, Department of Dermatology, Henry Ford Health System, Detroit, MI, United States.,Immunology Research Program, Henry Ford Cancer Institute, Henry Ford Health System, Detroit, MI, United States.,Department of Internal Medicine, Henry Ford Health System, Detroit, MI, United States
| |
Collapse
|
17
|
Maier B, Leader AM, Chen ST, Tung N, Chang C, LeBerichel J, Chudnovskiy A, Maskey S, Walker L, Finnigan JP, Kirkling ME, Reizis B, Ghosh S, D'Amore NR, Bhardwaj N, Rothlin CV, Wolf A, Flores R, Marron T, Rahman AH, Kenigsberg E, Brown BD, Merad M. A conserved dendritic-cell regulatory program limits antitumour immunity. Nature 2020; 580:257-262. [PMID: 32269339 DOI: 10.1038/s41586-020-2134-y] [Citation(s) in RCA: 513] [Impact Index Per Article: 102.6] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2019] [Accepted: 01/17/2020] [Indexed: 11/09/2022]
Abstract
Checkpoint blockade therapies have improved cancer treatment, but such immunotherapy regimens fail in a large subset of patients. Conventional type 1 dendritic cells (DC1s) control the response to checkpoint blockade in preclinical models and are associated with better overall survival in patients with cancer, reflecting the specialized ability of these cells to prime the responses of CD8+ T cells1-3. Paradoxically, however, DC1s can be found in tumours that resist checkpoint blockade, suggesting that the functions of these cells may be altered in some lesions. Here, using single-cell RNA sequencing in human and mouse non-small-cell lung cancers, we identify a cluster of dendritic cells (DCs) that we name 'mature DCs enriched in immunoregulatory molecules' (mregDCs), owing to their coexpression of immunoregulatory genes (Cd274, Pdcd1lg2 and Cd200) and maturation genes (Cd40, Ccr7 and Il12b). We find that the mregDC program is expressed by canonical DC1s and DC2s upon uptake of tumour antigens. We further find that upregulation of the programmed death ligand 1 protein-a key checkpoint molecule-in mregDCs is induced by the receptor tyrosine kinase AXL, while upregulation of interleukin (IL)-12 depends strictly on interferon-γ and is controlled negatively by IL-4 signalling. Blocking IL-4 enhances IL-12 production by tumour-antigen-bearing mregDC1s, expands the pool of tumour-infiltrating effector T cells and reduces tumour burden. We have therefore uncovered a regulatory module associated with tumour-antigen uptake that reduces DC1 functionality in human and mouse cancers.
Collapse
Affiliation(s)
- Barbara Maier
- The Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA.,The Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA.,Department of Oncological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Andrew M Leader
- The Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA.,The Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA.,Department of Oncological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Steven T Chen
- The Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA.,The Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA.,Department of Oncological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Navpreet Tung
- The Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA.,The Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Christie Chang
- The Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA.,Human Immune Monitoring Center, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Jessica LeBerichel
- The Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA.,The Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA.,Department of Oncological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Aleksey Chudnovskiy
- The Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA.,The Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA.,Department of Oncological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA.,Laboratory of Lymphocyte Dynamics, The Rockefeller University, New York, NY, USA
| | - Shrisha Maskey
- The Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA.,The Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA.,Department of Oncological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Laura Walker
- The Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA.,Human Immune Monitoring Center, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - John P Finnigan
- The Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA.,The Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Margaret E Kirkling
- Department of Pathology and Department of Medicine, New York University School of Medicine, New York, NY, USA.,Graduate Program in Genetics and Development, Columbia University Medical Center, New York, NY, USA
| | - Boris Reizis
- Department of Pathology and Department of Medicine, New York University School of Medicine, New York, NY, USA
| | - Sourav Ghosh
- Department of Neurology & Department of Pharmacology, Yale University School of Medicine, New Haven, CT, USA
| | | | - Nina Bhardwaj
- The Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA.,The Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA.,Department of Hematology/Oncology, Icahn School of Medicine at Mount Sinai, New York, NY, USA.,Parker Institute for Cancer Immunotherapy, San Francisco, CA, USA
| | - Carla V Rothlin
- Department of Immunobiology & Department of Pharmacology, Yale University School of Medicine, New Haven, CT, USA
| | - Andrea Wolf
- Department of Thoracic Surgery, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Raja Flores
- Department of Thoracic Surgery, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Thomas Marron
- The Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA.,The Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA.,Department of Hematology/Oncology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Adeeb H Rahman
- The Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA.,Human Immune Monitoring Center, Icahn School of Medicine at Mount Sinai, New York, NY, USA.,Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Ephraim Kenigsberg
- The Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA.,Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA.,Icahn Institute for Genomics and Multiscale Biology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Brian D Brown
- The Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA.,The Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA.,Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Miriam Merad
- The Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA. .,The Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA. .,Department of Oncological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA. .,Human Immune Monitoring Center, Icahn School of Medicine at Mount Sinai, New York, NY, USA.
| |
Collapse
|
18
|
Borek I, Köffel R, Feichtinger J, Spies M, Glitzner-Zeis E, Hochgerner M, Sconocchia T, Krump C, Tam-Amersdorfer C, Passegger C, Benezeder T, Tittes J, Redl A, Painsi C, Thallinger GG, Wolf P, Stary G, Sibilia M, Strobl H. BMP7 aberrantly induced in the psoriatic epidermis instructs inflammation-associated Langerhans cells. J Allergy Clin Immunol 2019; 145:1194-1207.e11. [PMID: 31870764 DOI: 10.1016/j.jaci.2019.12.011] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2019] [Revised: 12/08/2019] [Accepted: 12/13/2019] [Indexed: 12/31/2022]
Abstract
BACKGROUND Epidermal hyperplasia represents a morphologic hallmark of psoriatic skin lesions. Langerhans cells (LCs) in the psoriatic epidermis engage with keratinocytes (KCs) in tight physical interactions; moreover, they induce T-cell-mediated immune responses critical to psoriasis. OBJECTIVE This study sought to improve the understanding of epidermal factors in psoriasis pathogenesis. METHODS BMP7-LCs versus TGF-β1-LCs were phenotypically characterized and their functional properties were analyzed using flow cytometry, cell kinetic studies, co-culture with CD4 T cells, and cytokine measurements. Furthermore, immunohistology of healthy and psoriatic skin was performed. Additionally, in vivo experiments with Junf/fJunBf/fK5cre-ERT mice were carried out to assess the role of bone morphogenetic protein (BMP) signaling in psoriatic skin inflammation. RESULTS This study identified a KC-derived signal (ie, BMP signaling) to promote epidermal changes in psoriasis. Whereas BMP7 is strictly confined to the basal KC layer in the healthy skin, it is expressed at high levels throughout the lesional psoriatic epidermis. BMP7 instructs precursor cells to differentiate into LCs that phenotypically resemble psoriatic LCs. These BMP7-LCs exhibit proliferative activity and increased sensitivity to bacterial stimulation. Moreover, aberrant high BMP signaling in the lesional epidermis is mediated by a KC intrinsic mechanism, as suggested from murine data and clinical outcome after topical antipsoriatic treatment in human patients. CONCLUSIONS These data indicate that available TGF-β family members within the lesional psoriatic epidermis preferentially signal through the canonical BMP signaling cascade to instruct inflammatory-type LCs and to promote psoriatic epidermal changes. Targeting BMP signaling might allow to therapeutically interfere with cutaneous psoriatic manifestations.
Collapse
Affiliation(s)
- Izabela Borek
- Otto Loewi Research Center, Chair of Immunology and Pathophysiology, Medical University of Graz, Graz, Austria
| | - René Köffel
- Institute of Anatomy, University of Bern, Bern, Switzerland
| | - Julia Feichtinger
- Division of Cell Biology, Histology and Embryology, Gottfried Schatz Research Center for Cell Signaling, Metabolism and Aging, Medical University of Graz, Graz, Austria
| | - Melanie Spies
- Otto Loewi Research Center, Chair of Immunology and Pathophysiology, Medical University of Graz, Graz, Austria
| | - Elisabeth Glitzner-Zeis
- Institute of Cancer Research, Department of Internal Medicine I, Medical University of Vienna, Vienna, Austria
| | - Mathias Hochgerner
- Institute of Cancer Research, Department of Internal Medicine I, Medical University of Vienna, Vienna, Austria
| | - Tommaso Sconocchia
- Otto Loewi Research Center, Chair of Immunology and Pathophysiology, Medical University of Graz, Graz, Austria
| | - Corinna Krump
- Otto Loewi Research Center, Chair of Immunology and Pathophysiology, Medical University of Graz, Graz, Austria
| | - Carmen Tam-Amersdorfer
- Otto Loewi Research Center, Chair of Immunology and Pathophysiology, Medical University of Graz, Graz, Austria
| | - Christina Passegger
- Otto Loewi Research Center, Chair of Immunology and Pathophysiology, Medical University of Graz, Graz, Austria
| | - Theresa Benezeder
- Department of Dermatology, Medical University of Graz, Graz, Austria
| | - Julia Tittes
- Division of Immunology, Allergy and Infectious Diseases, Department of Dermatology, Medical University of Vienna, Vienna, Austria
| | - Anna Redl
- Division of Immunology, Allergy and Infectious Diseases, Department of Dermatology, Medical University of Vienna, Vienna, Austria
| | - Clemens Painsi
- Department of Dermatology, State Hospital Klagenfurt, Klagenfurt, Austria
| | - Gerhard G Thallinger
- Institute of Computational Biotechnology, Graz University of Technology, Graz, Austria; Omics Center Graz, BioTechMed Graz, Graz, Austria
| | - Peter Wolf
- Department of Dermatology, Medical University of Graz, Graz, Austria
| | - Georg Stary
- Division of Immunology, Allergy and Infectious Diseases, Department of Dermatology, Medical University of Vienna, Vienna, Austria
| | - Maria Sibilia
- Institute of Cancer Research, Department of Internal Medicine I, Medical University of Vienna, Vienna, Austria
| | - Herbert Strobl
- Otto Loewi Research Center, Chair of Immunology and Pathophysiology, Medical University of Graz, Graz, Austria.
| |
Collapse
|
19
|
Sil P, Suwanpradid J, Muse G, Gruzdev A, Liu L, Corcoran DL, Willson CJ, Janardhan K, Grimm S, Myers P, Degraff LM, MacLeod AS, Martinez J. Noncanonical autophagy in dermal dendritic cells mediates immunosuppressive effects of UV exposure. J Allergy Clin Immunol 2019; 145:1389-1405. [PMID: 31837371 DOI: 10.1016/j.jaci.2019.11.041] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2019] [Revised: 10/10/2019] [Accepted: 11/05/2019] [Indexed: 12/14/2022]
Abstract
BACKGROUND Control of the inflammatory response is critical to maintaining homeostasis, and failure to do so contributes to the burden of chronic inflammation associated with several disease states. The mechanisms that underlie immunosuppression, however, remain largely unknown. Although defects in autophagy machinery have been associated with inflammatory pathologic conditions, we now appreciate that autophagic components participate in noncanonical pathways distinct from classical autophagy. We have previously demonstrated that LC3-associated phagocytosis (LAP), a noncanonical autophagic process dependent on Rubicon (rubicon autophagy regulator [RUBCN]), contributes to immunosuppression. OBJECTIVE We used Rubcn-/- mice to examine the role of the LAP pathway in mediating the UV-induced immunotolerant program in a model of contact hypersensitivity (CHS). METHODS Flow cytometry and transcriptional analysis were used to measure immune cell infiltration and activation in the skin of Rubcn+/+ and Rubcn-/- mice during the CHS response. RESULTS Here, we demonstrate that LAP is required for UV-induced immunosuppression and that UV exposure induces a broadly anti-inflammatory transcriptional program dependent on Rubicon. Rubcn-/- mice are resistant to UV-induced immunosuppression and instead display exaggerated inflammation in a model of CHS. Specifically, RUBCN deficiency in CD301b+ dermal dendritic cells results in their increased antigen presentation capacity and subsequent hyperactivation of the CD8+ T-cell response. CONCLUSIONS LAP functions to limit the immune response and is critical in maintaining the balance between homeostasis and inflammation.
Collapse
Affiliation(s)
- Payel Sil
- Immunity, Inflammation, and Disease Laboratory, National Institute of Environmental Health Sciences, National Institutes of Health, Research Triangle Park, NC
| | | | - Ginger Muse
- Immunity, Inflammation, and Disease Laboratory, National Institute of Environmental Health Sciences, National Institutes of Health, Research Triangle Park, NC
| | - Artiom Gruzdev
- Knockout Mouse Core Laboratory, Reproductive and Developmental Biology Laboratory, National Institute of Environmental Health Sciences, National Institutes of Health, Research Triangle Park, NC
| | - Liwen Liu
- Molecular Genomics Core Laboratory, Signal Transduction Laboratory, National Institute of Environmental Health Sciences, National Institutes of Health, Research Triangle Park, NC
| | - David L Corcoran
- Duke Center for Genomic and Computational Biology, Duke University Medical Center, Durham, NC
| | | | | | - Sara Grimm
- Division of Intramural Research, Research Triangle Park, NC
| | - Page Myers
- Comparative Medicine Branch, National Institute of Environmental Health Sciences, National Institutes of Health, Research Triangle Park, NC
| | - Laura Miller Degraff
- Immunity, Inflammation, and Disease Laboratory, National Institute of Environmental Health Sciences, National Institutes of Health, Research Triangle Park, NC
| | - Amanda S MacLeod
- Department of Dermatology, Duke University, Durham, NC; Department of Immunology, Duke University, Durham, NC; Department of Molecular Genetics and Microbiology, Duke University, Durham, NC
| | - Jennifer Martinez
- Immunity, Inflammation, and Disease Laboratory, National Institute of Environmental Health Sciences, National Institutes of Health, Research Triangle Park, NC.
| |
Collapse
|
20
|
Solmaz G, Puttur F, Francozo M, Lindenberg M, Guderian M, Swallow M, Duhan V, Khairnar V, Kalinke U, Ludewig B, Clausen BE, Wagner H, Lang KS, Sparwasser TD. TLR7 Controls VSV Replication in CD169 + SCS Macrophages and Associated Viral Neuroinvasion. Front Immunol 2019; 10:466. [PMID: 30930901 PMCID: PMC6428728 DOI: 10.3389/fimmu.2019.00466] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2018] [Accepted: 02/21/2019] [Indexed: 01/21/2023] Open
Abstract
Vesicular stomatitis virus (VSV) is an insect-transmitted rhabdovirus that is neurovirulent in mice. Upon peripheral VSV infection, CD169+ subcapsular sinus (SCS) macrophages capture VSV in the lymph, support viral replication, and prevent CNS neuroinvasion. To date, the precise mechanisms controlling VSV infection in SCS macrophages remain incompletely understood. Here, we show that Toll-like receptor-7 (TLR7), the main sensing receptor for VSV, is central in controlling lymph-borne VSV infection. Following VSV skin infection, TLR7−/− mice display significantly less VSV titers in the draining lymph nodes (dLN) and viral replication is attenuated in SCS macrophages. In contrast to effects of TLR7 in impeding VSV replication in the dLN, TLR7−/− mice present elevated viral load in the brain and spinal cord highlighting their susceptibility to VSV neuroinvasion. By generating novel TLR7 floxed mice, we interrogate the impact of cell-specific TLR7 function in anti-viral immunity after VSV skin infection. Our data suggests that TLR7 signaling in SCS macrophages supports VSV replication in these cells, increasing LN infection and may account for the delayed onset of VSV-induced neurovirulence observed in TLR7−/− mice. Overall, we identify TLR7 as a novel and essential host factor that critically controls anti-viral immunity to VSV. Furthermore, the novel mouse model generated in our study will be of valuable importance to shed light on cell-intrinsic TLR7 biology in future studies.
Collapse
Affiliation(s)
- Gülhas Solmaz
- Institute of Infection Immunology, TWINCORE, Centre for Experimental and Clinical Infection Research, A Joint Venture Between the Medical School Hannover and the Helmholtz Centre for Infection Research (HZI), Hannover, Germany
| | - Franz Puttur
- Institute of Infection Immunology, TWINCORE, Centre for Experimental and Clinical Infection Research, A Joint Venture Between the Medical School Hannover and the Helmholtz Centre for Infection Research (HZI), Hannover, Germany
| | - Marcela Francozo
- Institute of Infection Immunology, TWINCORE, Centre for Experimental and Clinical Infection Research, A Joint Venture Between the Medical School Hannover and the Helmholtz Centre for Infection Research (HZI), Hannover, Germany
| | - Marc Lindenberg
- Institute of Infection Immunology, TWINCORE, Centre for Experimental and Clinical Infection Research, A Joint Venture Between the Medical School Hannover and the Helmholtz Centre for Infection Research (HZI), Hannover, Germany
| | - Melanie Guderian
- Institute of Infection Immunology, TWINCORE, Centre for Experimental and Clinical Infection Research, A Joint Venture Between the Medical School Hannover and the Helmholtz Centre for Infection Research (HZI), Hannover, Germany
| | - Maxine Swallow
- Institute of Infection Immunology, TWINCORE, Centre for Experimental and Clinical Infection Research, A Joint Venture Between the Medical School Hannover and the Helmholtz Centre for Infection Research (HZI), Hannover, Germany
| | - Vikas Duhan
- Institute of Immunology of the University Hospital in Essen, Medical Faculty, University of Duisburg-Essen, Essen, Germany
| | - Vishal Khairnar
- Institute of Immunology of the University Hospital in Essen, Medical Faculty, University of Duisburg-Essen, Essen, Germany
| | - Ulrich Kalinke
- Institute of Experimental Infection Research, TWINCORE, Centre for Experimental and Clinical Infection Research, A Joint Venture Between the Hannover Medical School and the Helmholtz Centre for Infection Research (HZI), Hannover, Germany
| | - Burkhard Ludewig
- Institute of Immunobiology, Kantonsspital St. Gallen, St. Gallen, Switzerland
| | - Björn E Clausen
- Institute for Molecular Medicine, University Medical Center of the Johannes Gutenberg-University Mainz, Mainz, Germany
| | - Hermann Wagner
- Institute for Medical Microbiology, Immunology and Hygiene, Technical University Munich, Munich, Germany
| | - Karl S Lang
- Institute of Immunology of the University Hospital in Essen, Medical Faculty, University of Duisburg-Essen, Essen, Germany
| | - Tim D Sparwasser
- Institute of Infection Immunology, TWINCORE, Centre for Experimental and Clinical Infection Research, A Joint Venture Between the Medical School Hannover and the Helmholtz Centre for Infection Research (HZI), Hannover, Germany.,Department of Medical Microbiology and Hygiene, University Medical Center of the Johannes Gutenberg University Mainz, Mainz, Germany
| |
Collapse
|
21
|
Honda T, Egawa G, Kabashima K. Antigen presentation and adaptive immune responses in skin. Int Immunol 2019; 31:423-429. [DOI: 10.1093/intimm/dxz005] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2018] [Accepted: 01/15/2019] [Indexed: 01/06/2023] Open
Abstract
Abstract
For the induction of adequate cutaneous immune responses, the antigen presentation and recognition that occur in both the skin and skin-draining lymph nodes are essential. In each process of cutaneous immune responses, several distinct subsets of immune cells, including dendritic cells and T cells, are involved, and they elicit their respective functions in a harmonious manner. For example, in the elicitation phase of cutaneous acquired immunity, immune cells form a specific lymphoid structure named inducible skin-associated lymphoid tissue (iSALT) to facilitate efficient antigen presentation in situ. In this short review, we will overview the mechanisms of how antigens are presented and how cutaneous adaptive immune responses are conducted in the skin, especially focusing on contact hypersensitivity, a prototypic adaptive immune response in the skin.
Collapse
Affiliation(s)
- Tetsuya Honda
- Department of Dermatology, Graduate School of Medicine, Kyoto University, Sakyo, Kyoto, Japan
| | - Gyohei Egawa
- Department of Dermatology, Graduate School of Medicine, Kyoto University, Sakyo, Kyoto, Japan
| | - Kenji Kabashima
- Department of Dermatology, Graduate School of Medicine, Kyoto University, Sakyo, Kyoto, Japan
- Singapore Immunology Network (SIgN) and Skin Research Institute of Singapore (SRIS), Agency for Science, Technology and Research (A*STAR), Biopolis, Immunos, Singapore
| |
Collapse
|
22
|
RARα supports the development of Langerhans cells and langerin-expressing conventional dendritic cells. Nat Commun 2018; 9:3896. [PMID: 30254197 PMCID: PMC6156335 DOI: 10.1038/s41467-018-06341-8] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2017] [Accepted: 08/29/2018] [Indexed: 01/11/2023] Open
Abstract
Langerhans cells (LC) are the prototype langerin-expressing dendritic cells (DC) that reside specifically in the epidermis, but langerin-expressing conventional DCs also reside in the dermis and other tissues, yet the factors that regulate their development are unclear. Because retinoic acid receptor alpha (RARα) is highly expressed by LCs, we investigate the functions of RARα and retinoic acid (RA) in regulating the langerin-expressing DCs. Here we show that the development of LCs from embryonic and bone marrow-derived progenitors and langerin+ conventional DCs is profoundly regulated by the RARα-RA axis. During LC differentiation, RARα is required for the expression of a LC-promoting transcription factor Runx3, but suppresses that of LC-inhibiting C/EBPβ. RARα promotes the development of LCs and langerin+ conventional DCs only in hypo-RA conditions, a function effectively suppressed at systemic RA levels. Our findings identify positive and negative regulatory mechanisms to tightly regulate the development of the specialized DC populations. Langerhans cells (LC) and langerin-expressing conventional dendritic cells are made from distinct progenitors and enriched in the distinct microenvironments of the skin. Here the authors show that these immune cells are regulated by retinoic acid receptor alpha (RARα) via simultaneous induction of LC-promoting Runx3 and repression of LC-inhibiting C/EBPβ.
Collapse
|
23
|
Otsuka M, Egawa G, Kabashima K. Uncovering the Mysteries of Langerhans Cells, Inflammatory Dendritic Epidermal Cells, and Monocyte-Derived Langerhans Cell-Like Cells in the Epidermis. Front Immunol 2018; 9:1768. [PMID: 30105033 PMCID: PMC6077183 DOI: 10.3389/fimmu.2018.01768] [Citation(s) in RCA: 44] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2018] [Accepted: 07/17/2018] [Indexed: 11/25/2022] Open
Abstract
The identity of Langerhans cells (LCs) has been called into question of late due to the increasing evidence that LCs originate from macrophage lineage instead of dendritic cell (DC) lineage as previously thought. For many years, LCs have been assumed to be DCs due to its migratory capabilities. However, recent studies have demonstrated that LCs are from macrophage lineage of the adult fetal liver (FL) progenitor. Bona fide LCs are now considered tissue-resident macrophages as they originate from the FL as shown by fate mapping models. In recent years, studies have shown that there are three types of antigen-presenting cells present in the epidermis, such as LCs, monocyte-derived LC-like cells, and inflammatory dendritic epidermal cells (IDECs). Of these, LC-like cells have been characterized in both human and mouse studies, while IDECs have only been described in human studies. This has shed a new light on the area of epidermal macrophages, suggesting that there might be a misidentification and misclassification of LCs. IDECs and LC-like cells have been shown to be present in both steady state and inflammatory state, but they are present in more significant amounts under inflammatory conditions such as atopic dermatitis, ultra violet injury, and psoriasis. In this review, we discuss what is already known and discuss the possible roles of LCs, LC-like cells, and IDECs during inflammation. Most intriguingly, we discuss the possibility of LCs having a dual identity as both a macrophage and a DC. This is shown as LCs are the only tissue-resident macrophage to have shown migratory property-like DCs.
Collapse
Affiliation(s)
- Masayuki Otsuka
- Department of Dermatology, Graduate School of Medicine, Kyoto, Japan
| | - Gyohei Egawa
- Department of Dermatology, Graduate School of Medicine, Kyoto, Japan
| | - Kenji Kabashima
- Department of Dermatology, Graduate School of Medicine, Kyoto, Japan.,Singapore Immunology Network (SIgN), Singapore, Singapore.,Institute of Medical Biology (IMB), Agency for Science, Technology and Research (ASTAR), Biopolis, Singapore, Singapore
| |
Collapse
|
24
|
Deckers J, Hammad H, Hoste E. Langerhans Cells: Sensing the Environment in Health and Disease. Front Immunol 2018; 9:93. [PMID: 29449841 PMCID: PMC5799717 DOI: 10.3389/fimmu.2018.00093] [Citation(s) in RCA: 118] [Impact Index Per Article: 16.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2017] [Accepted: 01/12/2018] [Indexed: 12/17/2022] Open
Abstract
In the last few decades, our understanding of Langerhans cells (LCs) has drastically changed based on novel findings regarding the developmental origin and biological functions of these epidermis-specific resident immune cells. It has become clear that LCs not only exert pivotal roles in immune surveillance and homeostasis but also impact on pathology by either inducing tolerance or mediating inflammation. Their unique capabilities to self-renew within the epidermis, while also being able to migrate to lymph nodes in order to present antigen, place LCs in a key position to sample the local environment and decide on the appropriate cutaneous immune response. Exciting new data distinguishing LCs from Langerin+ dermal dendritic cells (DCs) on a functional and ontogenic level reveal crucial roles for LCs in trauma and various skin pathologies, which will be thoroughly discussed here. However, despite rapid progress in the field, the exact role of LCs during immune responses has not been completely elucidated. This review focuses on what mouse models that have been developed in order to enable the study of murine LCs and other Langerin-expressing DCs have taught us about LC development and function.
Collapse
Affiliation(s)
- Julie Deckers
- VIB Center for Inflammation Research, Ghent, Belgium.,Department of Internal Medicine, Ghent University, Ghent, Belgium
| | - Hamida Hammad
- VIB Center for Inflammation Research, Ghent, Belgium.,Department of Internal Medicine, Ghent University, Ghent, Belgium
| | - Esther Hoste
- VIB Center for Inflammation Research, Ghent, Belgium.,Department of Biomedical Molecular Biology, Ghent University, Ghent, Belgium
| |
Collapse
|
25
|
Abstract
The Cre/loxP system is a widely applied technology for site-specific genetic manipulation in mice. This system allows for deletion of the genes of interest in specific cells, tissues, and whole organism to generate a diversity of conditional knockout mouse strains. Additionally, the Cre/loxP system is useful for development of cell- and tissue-specific reporter mice for lineage tracing, and cell-specific conditional depletion models in mice. Recently, the Cre/loxP technique was extensively adopted to characterize the monocyte/macrophage biology in mouse models. Compared to other relatively homogenous immune cell types such as neutrophils, mast cells, and basophils, monocytes/macrophages represent a highly heterogeneous population which lack specific markers or transcriptional factors. Though great efforts have been made toward establishing macrophage-specific Cre driver mice in the past decade, all of the current available strains are not perfect with regard to their depletion efficiency and targeting specificity for endogenous macrophages. Here we overview the commonly used Cre driver mouse strains targeting macrophages and discuss their major applications and limitations.
Collapse
|
26
|
Clayton K, Vallejo AF, Davies J, Sirvent S, Polak ME. Langerhans Cells-Programmed by the Epidermis. Front Immunol 2017; 8:1676. [PMID: 29238347 PMCID: PMC5712534 DOI: 10.3389/fimmu.2017.01676] [Citation(s) in RCA: 91] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2017] [Accepted: 11/15/2017] [Indexed: 12/24/2022] Open
Abstract
Langerhans cells (LCs) reside in the epidermis as a dense network of immune system sentinels. These cells determine the appropriate adaptive immune response (inflammation or tolerance) by interpreting the microenvironmental context in which they encounter foreign substances. In a normal physiological, "non-dangerous" situation, LCs coordinate a continuous state of immune tolerance, preventing unnecessary and harmful immune activation. Conversely, when they sense a danger signal, for example during infection or when the physical integrity of skin has been compromised as a result of a trauma, they instruct T lymphocytes of the adaptive immune system to mount efficient effector responses. Recent advances investigating the molecular mechanisms underpinning the cross talk between LCs and the epidermal microenvironment reveal its importance for programming LC biology. This review summarizes the novel findings describing LC origin and function through the analysis of the transcriptomic programs and gene regulatory networks (GRNs). Review and meta-analysis of publicly available datasets clearly delineates LCs as distinct from both conventional dendritic cells (DCs) and macrophages, suggesting a primary role for the epidermal microenvironment in programming LC biology. This concept is further supported by the analysis of the effect of epidermal pro-inflammatory signals, regulating key GRNs in human and murine LCs. Applying whole transcriptome analyses and in silico analysis has advanced our understanding of how LCs receive, integrate, and process signals from the steady-state and diseased epidermis. Interestingly, in homeostasis and under immunological stress, the molecular network in LCs remains relatively stable, reflecting a key evolutionary need related to tissue localization. Importantly, to fulfill their key role in orchestrating antiviral adaptive immune responses, LC share specific transcriptomic modules with other DC types able to cross-present antigens to cytotoxic CD8+ T cells, pointing to a possible evolutionary convergence mechanism. With the development of more advanced technologies allowing delineation of the molecular networks at the level of chromatin organization, histone modifications, protein translation, and phosphorylation, future "omics" investigations will bring in-depth understanding of the complex molecular mechanisms underpinning human LC biology.
Collapse
Affiliation(s)
- Kalum Clayton
- Systems Immmunology Group, Clinical and Experimental Sciences, Sir Henry Wellcome Laboratories, Faculty of Medicine, University of Southampton, Southampton, United Kingdom
| | - Andres F Vallejo
- Systems Immmunology Group, Clinical and Experimental Sciences, Sir Henry Wellcome Laboratories, Faculty of Medicine, University of Southampton, Southampton, United Kingdom
| | - James Davies
- Systems Immmunology Group, Clinical and Experimental Sciences, Sir Henry Wellcome Laboratories, Faculty of Medicine, University of Southampton, Southampton, United Kingdom
| | - Sofia Sirvent
- Systems Immmunology Group, Clinical and Experimental Sciences, Sir Henry Wellcome Laboratories, Faculty of Medicine, University of Southampton, Southampton, United Kingdom
| | - Marta E Polak
- Systems Immmunology Group, Clinical and Experimental Sciences, Sir Henry Wellcome Laboratories, Faculty of Medicine, University of Southampton, Southampton, United Kingdom
| |
Collapse
|
27
|
The Cytokine TGF-β Promotes the Development and Homeostasis of Alveolar Macrophages. Immunity 2017; 47:903-912.e4. [PMID: 29126797 DOI: 10.1016/j.immuni.2017.10.007] [Citation(s) in RCA: 233] [Impact Index Per Article: 29.1] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2017] [Revised: 07/25/2017] [Accepted: 10/11/2017] [Indexed: 12/23/2022]
Abstract
Alveolar macrophages (AMs) derive from fetal liver monocytes, which colonize the lung during embryonic development and give rise to fully mature AMs perinatally. AM differentiation requires granulocyte macrophage colony-stimulating factor (GM-CSF), but whether additional factors are involved in AM regulation is not known. Here we report that AMs, in contrast to most other tissue macrophages, were also dependent on transforming growth factor-β receptor (TGF-βR) signaling. Conditional deletion of TGF-βR in mice at different time points halted the development and differentiation of AMs. In adult mice, TGF-β was also critical for AM homeostasis. The source of TGF-β was AMs themselves, indicative of an autocrine loop that promotes AM self-maintenance. Mechanistically, TGF-βR signaling resulted in upregulation of PPAR-γ, a signature transcription factor essential for the development of AMs. These findings reveal an additional layer of complexity regarding the guidance cues, which govern the genesis, maturation, and survival of AMs.
Collapse
|
28
|
TGFβR signalling controls CD103 +CD11b + dendritic cell development in the intestine. Nat Commun 2017; 8:620. [PMID: 28931816 PMCID: PMC5607002 DOI: 10.1038/s41467-017-00658-6] [Citation(s) in RCA: 71] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2016] [Accepted: 07/19/2017] [Indexed: 01/01/2023] Open
Abstract
CD103+CD11b+ dendritic cells (DCs) are unique to the intestine, but the factors governing their differentiation are unclear. Here we show that transforming growth factor receptor 1 (TGFβR1) has an indispensable, cell intrinsic role in the development of these cells. Deletion of Tgfbr1 results in markedly fewer intestinal CD103+CD11b+ DCs and a reciprocal increase in the CD103−CD11b+ dendritic cell subset. Transcriptional profiling identifies markers that define the CD103+CD11b+ DC lineage, including CD101, TREM1 and Siglec-F, and shows that the absence of CD103+CD11b+ DCs in CD11c-Cre.Tgfbr1fl/fl mice reflects defective differentiation from CD103−CD11b+ intermediaries, rather than an isolated loss of CD103 expression. The defect in CD103+CD11b+ DCs is accompanied by reduced generation of antigen-specific, inducible FoxP3+ regulatory T cells in vitro and in vivo, and by reduced numbers of endogenous Th17 cells in the intestinal mucosa. Thus, TGFβR1-mediated signalling may explain the tissue-specific development of these unique DCs. Developmental cues for the different dendritic cell (DC) subsets in the intestine are yet to be defined. Here the authors show that TGFβR1 signalling is needed for development of CD103+CD11b+ intestinal DCs from CD103−CD11b+ cells and that they contribute to the generation of Th17 and regulatory T cells
Collapse
|
29
|
Abstract
Dendritic cells (DCs) play critical roles in activating innate immune cells and initiating adaptive immune responses. The functions of DCs were originally obscured by their overlap with other mononuclear phagocytes, but new mouse models have allowed for the selective ablation of subsets of DCs and have helped to identify their non-redundant roles in the immune system. These tools have elucidated the functions of DCs in host defense against pathogens, autoimmunity, and cancer. This review will describe the mouse models generated to interrogate the role of DCs and will discuss how their use has progressively clarified our understanding of the unique functions of DC subsets.
Collapse
Affiliation(s)
- Vivek Durai
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO 63110, USA.
| | - Kenneth M Murphy
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO 63110, USA; Howard Hughes Medical Institute, Washington University School of Medicine, St. Louis, MO 63110, USA.
| |
Collapse
|
30
|
Vroman H, Hendriks RW, Kool M. Dendritic Cell Subsets in Asthma: Impaired Tolerance or Exaggerated Inflammation? Front Immunol 2017; 8:941. [PMID: 28848549 PMCID: PMC5552666 DOI: 10.3389/fimmu.2017.00941] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2017] [Accepted: 07/24/2017] [Indexed: 12/14/2022] Open
Abstract
Asthma is a prevalent chronic heterogeneous inflammatory disease of the airways, leading to reversible airway obstruction, in which various inflammatory responses can be observed. Mild to moderate asthma patients often present with a Th2-mediated eosinophilic inflammation whereas in severe asthma patients, a Th17-associated neutrophilic or combined Th2 and Th17-mediated eosinophilic/neutrophilic inflammation is observed. The differentiation of these effector Th2 and Th17-cells is induced by allergen-exposed dendritic cells (DCs) that migrate toward the lung draining lymph node. The DC lineage comprises conventional DCs (cDCs) and plasmacytoid DCs (pDCs), of which the cDC lineage consists of type 1 cDCs (cDC1s) and cDC2s. During inflammation, also monocytes can differentiate into so-called monocyte-derived DCs (moDCs). These DC subsets differ both in ontogeny, localization, and in their functional properties. New identification tools and the availability of transgenic mice targeting specific DC subsets enable the investigation of how these different DC subsets contribute to or suppress asthma pathogenesis. In this review, we will discuss mechanisms used by different DC subsets to elicit or hamper the pathogenesis of both Th2-mediated eosinophilic asthma and more severe Th17-mediated neutrophilic inflammation.
Collapse
Affiliation(s)
- Heleen Vroman
- Department of Pulmonary Medicine, Erasmus MC, Rotterdam, Netherlands
| | - Rudi W Hendriks
- Department of Pulmonary Medicine, Erasmus MC, Rotterdam, Netherlands
| | - Mirjam Kool
- Department of Pulmonary Medicine, Erasmus MC, Rotterdam, Netherlands
| |
Collapse
|
31
|
T Cell Zone Resident Macrophages Silently Dispose of Apoptotic Cells in the Lymph Node. Immunity 2017; 47:349-362.e5. [DOI: 10.1016/j.immuni.2017.07.019] [Citation(s) in RCA: 70] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2017] [Revised: 05/19/2017] [Accepted: 07/25/2017] [Indexed: 02/07/2023]
|
32
|
Abstract
Skin is the largest organ of the body with a complex network of multitude of cell types that perform plastic and dynamic cellular communication to maintain several vital processes such as inflammation, immune response including induction of tolerance and disease prevention, wound healing, and angiogenesis. Of paramount importance are immunological functions of the skin that protect from harmful exposure coming from external and internal environments. Awareness of skin immunity can provide a better comprehension of inflammation, autoimmunity, cancer, graft-versus-host disease, vaccination, and immunotherapy approaches. This paper will update on what we currently know about immune sentinels contributing to skin immunity.
Collapse
Affiliation(s)
- Agata Matejuk
- Faculty of Health Science, Wroclaw Medical University, Wrocław, Poland. .,Faculty of Science and Technology, Karkonosze College, Jelenia Góra, Poland.
| |
Collapse
|
33
|
Sanjabi S, Oh SA, Li MO. Regulation of the Immune Response by TGF-β: From Conception to Autoimmunity and Infection. Cold Spring Harb Perspect Biol 2017; 9:cshperspect.a022236. [PMID: 28108486 DOI: 10.1101/cshperspect.a022236] [Citation(s) in RCA: 401] [Impact Index Per Article: 50.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Transforming growth factor β (TGF-β) is a pleiotropic cytokine involved in both suppressive and inflammatory immune responses. After 30 years of intense study, we have only begun to elucidate how TGF-β alters immunity under various conditions. Under steady-state conditions, TGF-β regulates thymic T-cell selection and maintains homeostasis of the naïve T-cell pool. TGF-β inhibits cytotoxic T lymphocyte (CTL), Th1-, and Th2-cell differentiation while promoting peripheral (p)Treg-, Th17-, Th9-, and Tfh-cell generation, and T-cell tissue residence in response to immune challenges. Similarly, TGF-β controls the proliferation, survival, activation, and differentiation of B cells, as well as the development and functions of innate cells, including natural killer (NK) cells, macrophages, dendritic cells, and granulocytes. Collectively, TGF-β plays a pivotal role in maintaining peripheral tolerance against self- and innocuous antigens, such as food, commensal bacteria, and fetal alloantigens, and in controlling immune responses to pathogens.
Collapse
Affiliation(s)
- Shomyseh Sanjabi
- Institute of Virology and Immunology, Gladstone Institutes, San Francisco, California 94158.,Department of Microbiology and Immunology, University of California, San Francisco, California 94143
| | - Soyoung A Oh
- Immunology Program, Memorial Sloan Kettering Cancer Center, New York, New York 10065
| | - Ming O Li
- Immunology Program, Memorial Sloan Kettering Cancer Center, New York, New York 10065
| |
Collapse
|
34
|
Nirschl CJ, Anandasabapathy N. Duality at the gate: Skin dendritic cells as mediators of vaccine immunity and tolerance. Hum Vaccin Immunother 2016; 12:104-16. [PMID: 26836327 DOI: 10.1080/21645515.2015.1066050] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
Abstract
Since Edward Jenner's discovery that intentional exposure to cowpox could provide lifelong protection from smallpox, vaccinations have been a major focus of medical research. However, while the protective benefits of many vaccines have been successfully translated into the clinic, the cellular and molecular mechanisms that differentiate effective vaccines from sub-optimal ones are not well understood. Dendritic cells (DCs) are the gatekeepers of the immune system, and are ultimately responsible for the generation of adaptive immunity and lifelong protective memory through interactions with T cells. In addition to lymph node and spleen resident DCs, a number of tissue resident DC populations have been identified at barrier tissues, such as the skin, which migrate to the local lymph node (migDC). These populations have unique characteristics, and play a key role in the function of cutaneous vaccinations by shuttling antigen from the vaccination site to the draining lymph node, rapidly capturing freely draining antigens in the lymph node, and providing key stimuli to T cells. However, while migDCs are responsible for the generation of immunity following exposure to certain pathogens and vaccines, recent work has identified a tolerogenic role for migDCs in the steady state as well as during protein immunization. Here, we examine the roles and functions of skin DC populations in the generation of protective immunity, as well as their role as regulators of the immune system.
Collapse
Affiliation(s)
- Christopher J Nirschl
- a Department of Dermatology ; Harvard Skin Disease Research Center; Brigham and Women's Hospital ; Boston , MA USA
| | - Niroshana Anandasabapathy
- a Department of Dermatology ; Harvard Skin Disease Research Center; Brigham and Women's Hospital ; Boston , MA USA
| |
Collapse
|
35
|
Haas K, Weighardt H, Deenen R, Köhrer K, Clausen B, Zahner S, Boukamp P, Bloch W, Krutmann J, Esser C. Aryl Hydrocarbon Receptor in Keratinocytes Is Essential for Murine Skin Barrier Integrity. J Invest Dermatol 2016; 136:2260-2269. [DOI: 10.1016/j.jid.2016.06.627] [Citation(s) in RCA: 63] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2016] [Revised: 06/21/2016] [Accepted: 06/28/2016] [Indexed: 12/28/2022]
|
36
|
Sichien D, Scott C, Martens L, Vanderkerken M, Van Gassen S, Plantinga M, Joeris T, De Prijck S, Vanhoutte L, Vanheerswynghels M, Van Isterdael G, Toussaint W, Madeira F, Vergote K, Agace W, Clausen B, Hammad H, Dalod M, Saeys Y, Lambrecht B, Guilliams M. IRF8 Transcription Factor Controls Survival and Function of Terminally Differentiated Conventional and Plasmacytoid Dendritic Cells, Respectively. Immunity 2016; 45:626-640. [DOI: 10.1016/j.immuni.2016.08.013] [Citation(s) in RCA: 154] [Impact Index Per Article: 17.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2015] [Revised: 06/13/2016] [Accepted: 06/28/2016] [Indexed: 12/13/2022]
|
37
|
Abstract
PURPOSE OF REVIEW This article summarizes recent research on the ontogeny of Langerhans cells and regulation of their homeostasis in quiescent and inflamed conditions. RECENT FINDINGS Langerhans cells originate prenatally and may endure throughout life, independently of bone marrow-derived precursors. Fate-mapping experiments have recently resolved the relative contribution of primitive yolk sac and fetal liver hematopoiesis to the initial formation of Langerhans cells. In postnatal life, local self-renewal restores Langerhans cell numbers following chronic or low-grade inflammatory insults. However, severe inflammation recruits de-novo bone marrow-derived precursors in two waves; a transient population of classical monocytes followed by uncharacterized myeloid precursors that form a stable self-renewing Langerhans cell network as inflammation subsides. Human CD1c⁺ dendritic cells have Langerhans cell potential in vitro, raising the possibility that dendritic cell progenitors provide the second wave. Langerhans cell development depends upon transforming growth factor beta receptor signaling with distinct pathways active during differentiation and homeostasis. Langerhans cell survival is mediated by multiple pathways including mechanistic target of rapamycin and extracellular signal-regulated kinase signaling, mechanisms that become highly relevant in Langerhans cell neoplasia. SUMMARY The study of Langerhans cells continues to provide novel and unexpected insights into the origin and regulation of myeloid cell populations. The melding of macrophage and dendritic cell biology, shaped by a unique habitat, is a special feature of Langerhans cells.
Collapse
Affiliation(s)
- Matthew Collin
- Human Dendritic Cell Laboratory, Institute of Cellular Medicine, Newcastle University, Framlington Place, Newcastle upon Tyne, UK
| | | |
Collapse
|
38
|
Clausen BE, Stoitzner P. Functional Specialization of Skin Dendritic Cell Subsets in Regulating T Cell Responses. Front Immunol 2015; 6:534. [PMID: 26557117 PMCID: PMC4617171 DOI: 10.3389/fimmu.2015.00534] [Citation(s) in RCA: 120] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2015] [Accepted: 10/02/2015] [Indexed: 12/18/2022] Open
Abstract
Dendritic cells (DC) are a heterogeneous family of professional antigen-presenting cells classically recognized as most potent inducers of adaptive immune responses. In this respect, Langerhans cells have long been considered to be prototypic immunogenic DC in the skin. More recently this view has considerably changed. The generation of in vivo cell ablation and lineage tracing models revealed the complexity of the skin DC network and, in particular, established the existence of a number of phenotypically distinct Langerin+ and negative DC populations in the dermis. Moreover, by now we appreciate that DC also exert important regulatory functions and are required for the maintenance of tolerance toward harmless foreign and self-antigens. This review summarizes our current understanding of the skin-resident DC system in the mouse and discusses emerging concepts on the functional specialization of the different skin DC subsets in regulating T cell responses. Special consideration is given to antigen cross-presentation as well as immune reactions toward contact sensitizers, cutaneous pathogens, and tumors. These studies form the basis for the manipulation of the human counterparts of the murine DC subsets to promote immunity or tolerance for the treatment of human disease.
Collapse
Affiliation(s)
- Björn E Clausen
- Institute for Molecular Medicine, University Medical Center of the Johannes Gutenberg-University Mainz , Mainz , Germany
| | - Patrizia Stoitzner
- Department of Dermatology and Venereology, Division of Experimental Dermatology, Medical University of Innsbruck , Innsbruck , Austria
| |
Collapse
|
39
|
Ono S, Kabashima K. Novel insights into the role of immune cells in skin and inducible skin-associated lymphoid tissue (iSALT). ALLERGO JOURNAL 2015. [DOI: 10.1007/s15007-015-0911-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/23/2022]
|
40
|
Novel insights into the role of immune cells in skin and inducible skin-associated lymphoid tissue (iSALT). ACTA ACUST UNITED AC 2015; 24:170-179. [PMID: 27069837 PMCID: PMC4792357 DOI: 10.1007/s40629-015-0065-1] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2015] [Accepted: 05/11/2015] [Indexed: 12/22/2022]
Abstract
The skin is equipped with serial barriers that provide rapid and efficient protection against external intruders. Beneath the epidermal physical barriers of the stratum corneum and the tight junctions, the integrated immune systems in both the epidermis and the dermis act in a coordinated manner to protect the host. This “immunological” barrier is composed of various cells, including skin-resident cells, such as keratinocytes, dendritic cells, tissue-resident macrophages, resident memory T cells, mast cells, and innate lymphoid cells. Additionally, infiltrating memory T cells, monocytes, neutrophils, basophils, and eosinophils are recruited in support of the host immunity. In addition to discussing the role of each of these cellular populations, we describe the concept of skin associated lymphoid tissue (SALT), which reminds us that the skin is an important component of the lymphatic system. We further describe the newly discovered phenomenon of multiple cell gathering under skin inflammation, which can be referred to as inducible SALT (iSALT). iSALT contributes to our understanding of SALT by highlighting the importance of direct cell-cell interaction in skin immunity.
Collapse
|
41
|
Croxford A, Lanzinger M, Hartmann F, Schreiner B, Mair F, Pelczar P, Clausen B, Jung S, Greter M, Becher B. The Cytokine GM-CSF Drives the Inflammatory Signature of CCR2+ Monocytes and Licenses Autoimmunity. Immunity 2015; 43:502-14. [DOI: 10.1016/j.immuni.2015.08.010] [Citation(s) in RCA: 314] [Impact Index Per Article: 31.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2015] [Revised: 05/04/2015] [Accepted: 06/01/2015] [Indexed: 11/28/2022]
|
42
|
The TGF-β superfamily in dendritic cell biology. Cytokine Growth Factor Rev 2015; 26:647-57. [PMID: 26115564 DOI: 10.1016/j.cytogfr.2015.06.002] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2015] [Accepted: 06/15/2015] [Indexed: 12/18/2022]
Abstract
The TGF-β superfamily consists of a large group of pleiotropic cytokines that are involved in the regulation of many developmental, physiological and pathological processes. Dendritic cells are antigen-presenting cells that play a key role in innate and adaptive immune responses. Dendritic cells have a complex relationship with the TGF-β cytokine superfamily being both source and targets for many of these cytokines. Some TGF-β family members are expressed by dendritic cells and modulate immune responses, for instance through the induction of T cell polarization. Others play a crucial role in the development and function of the different dendritic cell subsets. This review summarizes the current knowledge on the role of TGF-β family cytokines in dendritic cell biology, focusing on TGF-β as well as on other, less characterized, members of these important immune mediators.
Collapse
|
43
|
Abstract
In the 40 years since their discovery, dendritic cells (DCs) have been recognized as central players in immune regulation. DCs sense microbial stimuli through pathogen-recognition receptors (PRRs) and decode, integrate, and present information derived from such stimuli to T cells, thus stimulating immune responses. DCs can also regulate the quality of immune responses. Several functionally specialized subsets of DCs exist, but DCs also display functional plasticity in response to diverse stimuli. In addition to sensing pathogens via PRRs, emerging evidence suggests that DCs can also sense stress signals, such as amino acid starvation, through ancient stress and nutrient sensing pathways, to stimulate adaptive immunity. Here, I discuss these exciting advances in the context of a historic perspective on the discovery of DCs and their role in immune regulation. I conclude with a discussion of emerging areas in DC biology in the systems immunology era and suggest that the impact of DCs on immunity can be usefully contextualized in a hierarchy-of-organization model in which DCs, their receptors and signaling networks, cell-cell interactions, tissue microenvironment, and the host macroenvironment represent different levels of the hierarchy. Immunity or tolerance can then be represented as a complex function of each of these hierarchies.
Collapse
Affiliation(s)
- Bali Pulendran
- Emory Vaccine Center and Yerkes National Primate Research Center, Emory University, Atlanta, Georgia 30329;
| |
Collapse
|
44
|
Sparber F, Tripp CH, Komenda K, Scheffler JM, Clausen BE, Huber LA, Romani N, Stoitzner P. The late endosomal adaptor molecule p14 (LAMTOR2) regulates TGFβ1-mediated homeostasis of Langerhans cells. J Invest Dermatol 2015; 135:119-129. [PMID: 25078666 PMCID: PMC4285575 DOI: 10.1038/jid.2014.324] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2014] [Revised: 06/18/2014] [Accepted: 06/30/2014] [Indexed: 11/09/2022]
Abstract
Langerhans cells (LCs), a sub-population of dendritic cells (DCs) in the skin, participate in the regulation of immunity and peripheral tolerance. The adaptor molecule p14 is part of the late endosomal/lysosomal adaptor and mitogen-activated protein kinase and mammalian target of rapamycin (mTOR) activator/regulator (LAMTOR) complex, which mediates the activation of lysosome-associated extracellular signaling-regulated kinase (ERK) and the mTOR cascade. In previous work, we demonstrated that CD11c-specific deficiency of p14 disrupts LC homeostasis by affecting the LAMTOR-mediated ERK and mTOR signaling. In this study, we extended our analysis on p14 deficiency specifically in LCs. Langerin-specific ablation of p14 caused a complete loss of LCs, accompanied by an increased maturational phenotype of LCs. The absence of LCs in p14-deficient mice reduced contact hypersensitivity (CHS) responses to the contact sensitizer trinitrochlorobenzene. Analysis using bone marrow-derived DCs (BMDCs) revealed that p14 deficiency in DCs/LCs interfered with the LC-relevant transforming growth factor β1 (TGFβ1) pathway, by lowering TGFβ receptor II expression on BMDCs and LCs, as well as surface binding of TGFβ1 on BMDCs. We conclude that p14 deficiency affects TGFβ1 sensitivity of LCs, which is mandatory for their homeostasis and subsequently for their immunological function during CHS.
Collapse
Affiliation(s)
- Florian Sparber
- Department of Dermatology and Venereology, Innsbruck Medical University, Innsbruck, Austria
| | - Christoph H Tripp
- Department of Dermatology and Venereology, Innsbruck Medical University, Innsbruck, Austria
| | - Kerstin Komenda
- Department of Dermatology and Venereology, Innsbruck Medical University, Innsbruck, Austria
| | | | - Björn E Clausen
- Institute for Molecular Medicine, University Medical Center of the Johannes Gutenberg-University Mainz, Mainz, Germany
| | - Lukas A Huber
- Division of Cell Biology, Biocenter Innsbruck, Innsbruck, Austria
| | - Nikolaus Romani
- Department of Dermatology and Venereology, Innsbruck Medical University, Innsbruck, Austria
| | - Patrizia Stoitzner
- Department of Dermatology and Venereology, Innsbruck Medical University, Innsbruck, Austria.
| |
Collapse
|
45
|
Arnold-Schrauf C, Berod L, Sparwasser T. Dendritic cell specific targeting of MyD88 signalling pathways in vivo. Eur J Immunol 2014; 45:32-9. [DOI: 10.1002/eji.201444747] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2014] [Revised: 11/09/2014] [Accepted: 11/11/2014] [Indexed: 12/19/2022]
Affiliation(s)
- Catharina Arnold-Schrauf
- Institute for Infection Immunology; TWINCORE, Center for Experimental and Clinical Infection Research, a Joint Venture between the Medical School Hannover (MHH) and the Helmholtz Center for Infection Research (HZI); Hannover Germany
| | - Luciana Berod
- Institute for Infection Immunology; TWINCORE, Center for Experimental and Clinical Infection Research, a Joint Venture between the Medical School Hannover (MHH) and the Helmholtz Center for Infection Research (HZI); Hannover Germany
| | - Tim Sparwasser
- Institute for Infection Immunology; TWINCORE, Center for Experimental and Clinical Infection Research, a Joint Venture between the Medical School Hannover (MHH) and the Helmholtz Center for Infection Research (HZI); Hannover Germany
| |
Collapse
|
46
|
Comparative analysis of the efficiency and specificity of myeloid-Cre deleting strains using ROSA-EYFP reporter mice. J Immunol Methods 2014; 408:89-100. [PMID: 24857755 DOI: 10.1016/j.jim.2014.05.009] [Citation(s) in RCA: 368] [Impact Index Per Article: 33.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2014] [Revised: 05/06/2014] [Accepted: 05/07/2014] [Indexed: 12/22/2022]
Abstract
Since the first example of conditional gene targeting in mice in 1994, the use of Cre recombinase and loxP flanked sequences has become an invaluable technique to generate tissue and temporal specific gene knockouts. The number of mouse strains expressing floxed-gene sequences, and tissue-specific or temporal-specific Cre-recombinase that have been reported in the literature has grown exponentially. However, increased use of this technology has highlighted several problems that can impact the interpretation of any phenotype observed in these mouse models. In particular, accurate knowledge of the specificity of Cre expression in each strain is critical in order to make conclusions about the role of specific cell types in the phenotypes observed. Cre-mediated deletion specificity and efficiency have been described in many different ways in the literature, making direct comparisons between these Cre strains impossible. Here we report crossing thirteen different myeloid-Cre mouse strains to ROSA-EYFP reporter mice and assaying YFP expression in a variety of naïve unstimulated hematopoietic cells, in parallel. By focusing on myeloid subsets, we directly compare the relative efficiency and specificity of myeloid deletion in these strains under steady-state conditions.
Collapse
|
47
|
Berres ML, Lim KPH, Peters T, Price J, Takizawa H, Salmon H, Idoyaga J, Ruzo A, Lupo PJ, Hicks MJ, Shih A, Simko SJ, Abhyankar H, Chakraborty R, Leboeuf M, Beltrão M, Lira SA, Heym KM, Bigley V, Collin M, Manz MG, McClain K, Merad M, Allen CE. BRAF-V600E expression in precursor versus differentiated dendritic cells defines clinically distinct LCH risk groups. ACTA ACUST UNITED AC 2014; 211:669-83. [PMID: 24638167 PMCID: PMC3978272 DOI: 10.1084/jem.20130977] [Citation(s) in RCA: 286] [Impact Index Per Article: 26.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023]
Abstract
Langerhans cell histiocytosis (LCH) is a clonal disorder with elusive etiology, characterized by the accumulation of CD207(+) dendritic cells (DCs) in inflammatory lesions. Recurrent BRAF-V600E mutations have been reported in LCH. In this study, lesions from 100 patients were genotyped, and 64% carried the BRAF-V600E mutation within infiltrating CD207(+) DCs. BRAF-V600E expression in tissue DCs did not define specific clinical risk groups but was associated with increased risk of recurrence. Strikingly, we found that patients with active, high-risk LCH also carried BRAF-V600E in circulating CD11c(+) and CD14(+) fractions and in bone marrow (BM) CD34(+) hematopoietic cell progenitors, whereas the mutation was restricted to lesional CD207(+) DC in low-risk LCH patients. Importantly, BRAF-V600E expression in DCs was sufficient to drive LCH-like disease in mice. Consistent with our findings in humans, expression of BRAF-V600E in BM DC progenitors recapitulated many features of the human high-risk LCH, whereas BRAF-V600E expression in differentiated DCs more closely resembled low-risk LCH. We therefore propose classification of LCH as a myeloid neoplasia and hypothesize that high-risk LCH arises from somatic mutation of a hematopoietic progenitor, whereas low-risk disease arises from somatic mutation of tissue-restricted precursor DCs.
Collapse
Affiliation(s)
- Marie-Luise Berres
- Department of Oncological Sciences, 2 Tisch Cancer Institute, and 3 Immunology Institute, Mount Sinai School of Medicine, New York, NY 10029
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
48
|
Ravindran A, Mohammed J, Gunderson AJ, Cui X, Glick AB. Tumor-promoting role of TGFβ1 signaling in ultraviolet B-induced skin carcinogenesis is associated with cutaneous inflammation and lymph node migration of dermal dendritic cells. Carcinogenesis 2013; 35:959-66. [PMID: 24363069 DOI: 10.1093/carcin/bgt486] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
Transforming growth factor beta 1 (TGFβ1) is a pleiotropic cytokine in the skin that can function both as a tumor promoter and suppressor in chemically induced skin carcinogenesis, but the function in ultraviolet B (UVB) carcinogenesis is not well understood. Treatment of SKH1 hairless mice with the activin-like kinase 5 (ALK5) inhibitor SB431542 to block UVB-induced activation of cutaneous TGFβ1 signaling suppressed skin tumor formation but did not alter tumor size or tumor cell proliferation. Tumors that arose in SB-treated mice after 30 weeks had significantly reduced percentage of IFNγ(+) tumor-infiltrating lymphocytes compared with control mice. SB431542 blocked acute and chronic UVB-induced skin inflammation and T-cell activation in the skin-draining lymph node (SDLN) and skin but did not alter UVB-induced epidermal proliferation. We tested the effect of SB431542 on migration of skin dendritic cell (DC) populations because DCs are critical mediators of T-cell activation and cutaneous inflammation. SB431542 blocked (i) UVB-induced Smad2 phosphorylation in dermal DC (dDC) and (ii) SDLN and ear explant migration of CD103(+) CD207(+) and CD207(-) skin DC subsets but did not affect basal or UV-induced migration of Langerhans cells. Mice expressing a dominant-negative TGFβ type II receptor in CD11c(+) cells had reduced basal and UVB-induced SDLN migration of CD103(+) CD207(+) and CD207(-) DC subsets and a reduced percentage of CD86(high) dDC following UVB irradiation. Together, these suggest that TGFβ1 signaling has a tumor-promoting role in UVB-induced skin carcinogenesis and this is mediated in part through its role in UVB-induced migration of dDC and cutaneous inflammation.
Collapse
Affiliation(s)
- Anand Ravindran
- Department of Veterinary and Biomedical Sciences, Center for Molecular Toxicology and Carcinogenesis, The Pennsylvania State University, University Park, PA, USA
| | | | | | | | | |
Collapse
|
49
|
Yasmin N, Bauer T, Modak M, Wagner K, Schuster C, Köffel R, Seyerl M, Stöckl J, Elbe-Bürger A, Graf D, Strobl H. Identification of bone morphogenetic protein 7 (BMP7) as an instructive factor for human epidermal Langerhans cell differentiation. ACTA ACUST UNITED AC 2013; 210:2597-610. [PMID: 24190429 PMCID: PMC3832935 DOI: 10.1084/jem.20130275] [Citation(s) in RCA: 78] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/03/2022]
Abstract
Bone morphogenetic protein 7 (BMP7) promotes the differentiation of Langerhans cells in the epidermis during prenatal development. Human Langerhans cell (LC) precursors populate the epidermis early during prenatal development and thereafter undergo massive proliferation. The prototypic antiproliferative cytokine TGF-β1 is required for LC differentiation from human CD34+ hematopoietic progenitor cells and blood monocytes in vitro. Similarly, TGF-β1 deficiency results in LC loss in vivo. However, immunohistology studies revealed that human LC niches in early prenatal epidermis and adult basal (germinal) keratinocyte layers lack detectable TGF-β1. Here we demonstrated that these LC niches express high levels of bone morphogenetic protein 7 (BMP7) and that Bmp7-deficient mice exhibit substantially diminished LC numbers, with the remaining cells appearing less dendritic. BMP7 induces LC differentiation and proliferation by activating the BMP type-I receptor ALK3 in the absence of canonical TGF-β1–ALK5 signaling. Conversely, TGF-β1–induced in vitro LC differentiation is mediated via ALK3; however, co-induction of ALK5 diminished TGF-β1–driven LC generation. Therefore, selective ALK3 signaling by BMP7 promotes high LC yields. Within epidermis, BMP7 shows an inverse expression pattern relative to TGF-β1, the latter induced in suprabasal layers and up-regulated in outer layers. We observed that TGF-β1 inhibits microbial activation of BMP7-generated LCs. Therefore, TGF-β1 in suprabasal/outer epidermal layers might inhibit LC activation, resulting in LC network maintenance.
Collapse
Affiliation(s)
- Nighat Yasmin
- Institute of Pathophysiology and Immunology, Center for Molecular Medicine and 2 Center for Medical Research, Medical University Graz, A-8036 Graz, Austria
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
50
|
Langerin+ dermal DC, but not Langerhans cells, are required for effective CD8-mediated immune responses after skin scarification with vaccinia virus. J Invest Dermatol 2013; 134:686-694. [PMID: 24126845 PMCID: PMC3945525 DOI: 10.1038/jid.2013.418] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2013] [Revised: 09/11/2013] [Accepted: 09/13/2013] [Indexed: 02/08/2023]
Abstract
Skin scarification (s.s.) with Vaccinia virus (VACV) is essential for generation of an optimal protective T cell memory immune response. Dendritic Cells (DC), which are professional antigen presenting cells, are required for naïve T cell priming and activation. At least three subsets of skin resident DC have been identified: Langerhans Cells (LC), Dermal Langerin+ DC (Lang+dDC) and Dermal Langerin− DC (Lang−dDC). Using Langerin-diphtheria toxin receptor mice and established mouse model of VACV delivered by s.s., we demonstrated that Lang+dDC, but not LC, are absolutely required for the induction of a rapid and robust antigen-specific CD8+ T cell response after s.s. with VACV. The depletion of Lang+dDC led to a significant delay in the priming and proliferation of antigen-specific CD8+ T cells. Moreover CD8+ T cells generated after VACV s.s. in the absence of Lang+dDC lacked effector cytotoxic functions both in vitro and in vivo. While s.s.-immunized WT and LC depleted mice controlled the progression of OVA257–264 expressing T cell lymphoma EG7 (injected intradermally), the depletion of Lang+dDC led to rapid lymphoma progression and mortality. These data indicate that of all skin DC subsets, Lang+dDC the most critical for the generation of robust CD8+ T cell immunity after s.s. with VACV.
Collapse
|