151
|
Hoffmann F, Ender F, Schmudde I, Lewkowich IP, Köhl J, König P, Laumonnier Y. Origin, Localization, and Immunoregulatory Properties of Pulmonary Phagocytes in Allergic Asthma. Front Immunol 2016; 7:107. [PMID: 27047494 PMCID: PMC4803735 DOI: 10.3389/fimmu.2016.00107] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2016] [Accepted: 03/08/2016] [Indexed: 01/21/2023] Open
Abstract
Allergic asthma is a chronic inflammatory disease of the airways that is driven by maladaptive T helper 2 (Th2) and Th17 immune responses against harmless, airborne substances. Pulmonary phagocytes represent the first line of defense in the lung where they constantly sense the local environment for potential threats. They comprise two distinct cell types, i.e., macrophages and dendritic cells (DC) that differ in their origins and functions. Alveolar macrophages quickly take up most of the inhaled allergens, yet do not deliver their cargo to naive T cells sampling in draining lymph nodes. In contrast, pulmonary DCs instruct CD4(+) T cells develop into Th2 and Th17 effectors, initiating the maladaptive immune responses toward harmless environmental substances observed in allergic individuals. Unraveling the mechanisms underlying this mistaken identity of harmless, airborne substances by innate immune cells is one of the great challenges in asthma research. The identification of different pulmonary DC subsets, their role in antigen uptake, migration to the draining lymph nodes, and their potential to instruct distinct T cell responses has set the stage to unravel this mystery. However, at this point, a detailed understanding of the spatiotemporal resolution of DC subset localization, allergen uptake, processing, autocrine and paracrine cellular crosstalk, and the humoral factors that define the activation status of DCs is still lacking. In addition to DCs, at least two distinct macrophage populations have been identified in the lung that are either located in the airway/alveolar lumen or in the interstitium. Recent data suggest that such populations can exert either pro- or anti-inflammatory functions. Similar to the DC subsets, detailed insights into the individual roles of alveolar and interstitial macrophages during the different phases of asthma development are still missing. Here, we will provide an update on the current understanding of the origin, localization, and function of the diverse pulmonary antigen-presenting cell subsets, in particular with regard to the development and regulation of allergic asthma. While most data are from mouse models of experimental asthma, we have also included available human data to judge the translational value of the findings obtained in experimental asthma models.
Collapse
Affiliation(s)
| | - Fanny Ender
- Institute for Systemic Inflammation Research, University of Lübeck, Lübeck, Germany
| | - Inken Schmudde
- Institute for Systemic Inflammation Research, University of Lübeck, Lübeck, Germany
| | - Ian P. Lewkowich
- Division of Immunobiology, Cincinnati Children’s Hospital Medical Center, College of Medicine, University of Cincinnati, Cincinnati, OH, USA
| | - Jörg Köhl
- Institute for Systemic Inflammation Research, University of Lübeck, Lübeck, Germany
- Division of Immunobiology, Cincinnati Children’s Hospital Medical Center, College of Medicine, University of Cincinnati, Cincinnati, OH, USA
- Airway Research Center North (ARCN), German Center for Lung Research (DZL), Giessen, Germany
| | - Peter König
- Institute for Anatomy, University of Lübeck, Lübeck, Germany
- Airway Research Center North (ARCN), German Center for Lung Research (DZL), Giessen, Germany
| | - Yves Laumonnier
- Institute for Systemic Inflammation Research, University of Lübeck, Lübeck, Germany
| |
Collapse
|
152
|
Yu YRA, O’Koren EG, Hotten DF, Kan MJ, Kopin D, Nelson ER, Que L, Gunn MD. A Protocol for the Comprehensive Flow Cytometric Analysis of Immune Cells in Normal and Inflamed Murine Non-Lymphoid Tissues. PLoS One 2016; 11:e0150606. [PMID: 26938654 PMCID: PMC4777539 DOI: 10.1371/journal.pone.0150606] [Citation(s) in RCA: 264] [Impact Index Per Article: 33.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2015] [Accepted: 02/16/2016] [Indexed: 11/18/2022] Open
Abstract
Flow cytometry is used extensively to examine immune cells in non-lymphoid tissues. However, a method of flow cytometric analysis that is both comprehensive and widely applicable has not been described. We developed a protocol for the flow cytometric analysis of non-lymphoid tissues, including methods of tissue preparation, a 10-fluorochrome panel for cell staining, and a standardized gating strategy, that allows the simultaneous identification and quantification of all major immune cell types in a variety of normal and inflamed non-lymphoid tissues. We demonstrate that our basic protocol minimizes cell loss, reliably distinguishes macrophages from dendritic cells (DC), and identifies all major granulocytic and mononuclear phagocytic cell types. This protocol is able to accurately quantify 11 distinct immune cell types, including T cells, B cells, NK cells, neutrophils, eosinophils, inflammatory monocytes, resident monocytes, alveolar macrophages, resident/interstitial macrophages, CD11b- DC, and CD11b+ DC, in normal lung, heart, liver, kidney, intestine, skin, eyes, and mammary gland. We also characterized the expression patterns of several commonly used myeloid and macrophage markers. This basic protocol can be expanded to identify additional cell types such as mast cells, basophils, and plasmacytoid DC, or perform detailed phenotyping of specific cell types. In examining models of primary and metastatic mammary tumors, this protocol allowed the identification of several distinct tumor associated macrophage phenotypes, the appearance of which was highly specific to individual tumor cell lines. This protocol provides a valuable tool to examine immune cell repertoires and follow immune responses in a wide variety of tissues and experimental conditions.
Collapse
Affiliation(s)
- Yen-Rei A. Yu
- Department of Medicine, Division of Pulmonary and Critical Care Medicine, Duke University Medical Center, Durham, North Carolina, United States of America
- * E-mail:
| | - Emily G. O’Koren
- Department of Immunology, Duke University Medical Center, Durham, North Carolina, United States of America
| | - Danielle F. Hotten
- Department of Medicine, Division of Cardiology, Duke University Medical Center, Durham, North Carolina, United States of America
| | - Matthew J. Kan
- Department of Immunology, Duke University Medical Center, Durham, North Carolina, United States of America
| | - David Kopin
- School of Medicine, Duke University Medical Center, Durham, North Carolina, United States of America
| | - Erik R. Nelson
- Department of Molecular & Integrative Physiology, University of Illinois, Champaign, Illinois, United States of America
| | - Loretta Que
- Department of Medicine, Division of Pulmonary and Critical Care Medicine, Duke University Medical Center, Durham, North Carolina, United States of America
| | - Michael D. Gunn
- Department of Medicine, Division of Cardiology, Duke University Medical Center, Durham, North Carolina, United States of America
| |
Collapse
|
153
|
Lymphatic endothelial lineage assemblage during corneal lymphangiogenesis. J Transl Med 2016; 96:270-82. [PMID: 26658452 PMCID: PMC4767586 DOI: 10.1038/labinvest.2015.147] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2015] [Revised: 09/16/2015] [Accepted: 09/28/2015] [Indexed: 02/06/2023] Open
Abstract
Postnatal inflammatory lymphangiogenesis presumably requires precise regulatory processes to properly assemble proliferating lymphatic endothelial cells (LECs). The specific mechanisms that regulate the assembly of LECs during new lymphatic vessel synthesis are unclear. Dynamic endothelial shuffling and rearrangement has been proposed as a mechanism of blood vessel growth. We developed genetic lineage-tracing strategies using an inductive transgenic technology to track the fate of entire tandem dimer tomato-positive (tdT) lymphatic vessels or small, in some cases clonal, populations of LECs. We coupled this platform with a suture-induced mouse model of corneal lymphangiogenesis and used different analytic microscopy techniques including serial live imaging to study the spatial properties of proliferating tdT(+) LEC progenies. LEC precursors and their progeny expanded from the corneal limbal lymphatic vessel and were assembled contiguously to comprise a subunit within a new lymphatic vessel. VE-cadherin blockade induced morphologic abnormalities in newly synthesized lymphatic vessels, but did not disrupt the tdT(+) lymphatic endothelial lineage assembly. Analysis of this static and dynamic data based largely on direct in vivo observations supports a model of lymphatic endothelial lineage assemblage during corneal inflammatory lymphangiogenesis.
Collapse
|
154
|
Muzaki ARBM, Tetlak P, Sheng J, Loh SC, Setiagani YA, Poidinger M, Zolezzi F, Karjalainen K, Ruedl C. Intestinal CD103(+)CD11b(-) dendritic cells restrain colitis via IFN-γ-induced anti-inflammatory response in epithelial cells. Mucosal Immunol 2016; 9:336-51. [PMID: 26174764 PMCID: PMC4801902 DOI: 10.1038/mi.2015.64] [Citation(s) in RCA: 87] [Impact Index Per Article: 10.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2015] [Accepted: 06/15/2015] [Indexed: 02/04/2023]
Abstract
A crosstalk between commensals, gut immune cells, and colonic epithelia is required for a proper function of intestinal mucosal barrier. Here we investigated the importance of two distinct intestinal dendritic cell (DC) subsets in controlling intestinal inflammation. We show that Clec9A-diphtheria toxin receptor (DTR) mice after depletion of CD103(+)CD11b(-) DCs developed severe, low-dose dextran sodium sulfate (DSS)-induced colitis, whereas the lack of CD103(+)CD11b(+) DCs in Clec4a4-DTR mice did not exacerbate intestinal inflammation. The CD103(+)CD11b(-) DC subset has gained a functional specialization that able them to repress inflammation via several epithelial interferon-γ (IFN-γ)-induced proteins. Among others, we identified that epithelial IDO1 and interleukin-18-binding protein (IL-18bp) were strongly modulated by CD103(+)CD11b(-) DCs. Through its preferential property to express IL-12 and IL-15, this particular DC subset can induce lymphocytes in colonic lamina propria and in epithelia to secrete IFN-γ that then can trigger a reversible early anti-inflammatory response in intestinal epithelial cells.
Collapse
Affiliation(s)
- A R B M Muzaki
- School of Biological Sciences, Nanyang Technological University, Singapore, Singapore
| | - P Tetlak
- School of Biological Sciences, Nanyang Technological University, Singapore, Singapore
| | - J Sheng
- School of Biological Sciences, Nanyang Technological University, Singapore, Singapore
| | - S C Loh
- School of Biological Sciences, Nanyang Technological University, Singapore, Singapore
| | - Y A Setiagani
- School of Biological Sciences, Nanyang Technological University, Singapore, Singapore
| | - M Poidinger
- Singapore Immunology Network, Agency for Science, Technology and Research, Singapore, Singapore
| | - F Zolezzi
- Singapore Immunology Network, Agency for Science, Technology and Research, Singapore, Singapore
| | - K Karjalainen
- School of Biological Sciences, Nanyang Technological University, Singapore, Singapore
| | - C Ruedl
- School of Biological Sciences, Nanyang Technological University, Singapore, Singapore,()
| |
Collapse
|
155
|
Satb1 Overexpression Drives Tumor-Promoting Activities in Cancer-Associated Dendritic Cells. Cell Rep 2016; 14:1774-1786. [PMID: 26876172 DOI: 10.1016/j.celrep.2016.01.056] [Citation(s) in RCA: 90] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2015] [Revised: 12/14/2015] [Accepted: 01/14/2016] [Indexed: 02/07/2023] Open
Abstract
Special AT-rich sequence-binding protein 1 (Satb1) governs genome-wide transcriptional programs. Using a conditional knockout mouse, we find that Satb1 is required for normal differentiation of conventional dendritic cells (DCs). Furthermore, Satb1 governs the differentiation of inflammatory DCs by regulating major histocompatibility complex class II (MHC II) expression through Notch1 signaling. Mechanistically, Satb1 binds to the Notch1 promoter, activating Notch expression and driving RBPJ occupancy of the H2-Ab1 promoter, which activates MHC II transcription. However, tumor-driven, unremitting expression of Satb1 in activated Zbtb46(+) inflammatory DCs that infiltrate ovarian tumors results in an immunosuppressive phenotype characterized by increased secretion of tumor-promoting Galectin-1 and IL-6. In vivo silencing of Satb1 in tumor-associated DCs reverses their tumorigenic activity and boosts protective immunity. Therefore, dynamic fluctuations in Satb1 expression govern the generation and immunostimulatory activity of steady-state and inflammatory DCs, but continuous Satb1 overexpression in differentiated DCs converts them into tolerogenic/pro-inflammatory cells that contribute to malignant progression.
Collapse
|
156
|
Helft J, Böttcher JP, Chakravarty P, Zelenay S, Huotari J, Schraml BU, Goubau D, Reis e Sousa C. Alive but Confused: Heterogeneity of CD11c(+) MHC Class II(+) Cells in GM-CSF Mouse Bone Marrow Cultures. Immunity 2016; 44:3-4. [PMID: 26789913 DOI: 10.1016/j.immuni.2015.12.014] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Affiliation(s)
- Julie Helft
- Immunobiology Laboratory, The Francis Crick Institute, Lincoln's Inn Fields Laboratory, 44 Lincoln's Inn Fields, London WC2A 3LY, UK
| | - Jan P Böttcher
- Immunobiology Laboratory, The Francis Crick Institute, Lincoln's Inn Fields Laboratory, 44 Lincoln's Inn Fields, London WC2A 3LY, UK
| | - Probir Chakravarty
- Bioinformatics Laboratory, The Francis Crick Institute, Lincoln's Inn Fields Laboratory, 44 Lincoln's Inn Fields, London WC2A 3LY, UK
| | - Santiago Zelenay
- Immunobiology Laboratory, The Francis Crick Institute, Lincoln's Inn Fields Laboratory, 44 Lincoln's Inn Fields, London WC2A 3LY, UK
| | - Jatta Huotari
- Immunobiology Laboratory, The Francis Crick Institute, Lincoln's Inn Fields Laboratory, 44 Lincoln's Inn Fields, London WC2A 3LY, UK
| | - Barbara U Schraml
- Immunobiology Laboratory, The Francis Crick Institute, Lincoln's Inn Fields Laboratory, 44 Lincoln's Inn Fields, London WC2A 3LY, UK
| | - Delphine Goubau
- Immunobiology Laboratory, The Francis Crick Institute, Lincoln's Inn Fields Laboratory, 44 Lincoln's Inn Fields, London WC2A 3LY, UK
| | - Caetano Reis e Sousa
- Immunobiology Laboratory, The Francis Crick Institute, Lincoln's Inn Fields Laboratory, 44 Lincoln's Inn Fields, London WC2A 3LY, UK.
| |
Collapse
|
157
|
Abstract
Dendritic cells are specialized antigen-presenting cells that initiate and orient immune responses. Numerous studies in mice and humans have shown that dendritic cells are heterogeneous and comprise several subsets that can be distinguished by their surface phenotype, ontogeny, and molecular signature. This review gives an overview of mouse and human dendritic cell subsets and their defining features and summarizes the current knowledge of dendritic cell subsets' functional specialization in terms of antigen presentation.
Collapse
Affiliation(s)
- Elodie Segura
- Institut Curie, 26 rue d'Ulm, 75248, Paris Cedex 05, France. .,INSERM U932, 26 rue d'Ulm, 75248, Paris Cedex 05, France.
| |
Collapse
|
158
|
Scott CL, Wright PB, Milling SWF, Mowat AM. Isolation and Identification of Conventional Dendritic Cell Subsets from the Intestine of Mice and Men. Methods Mol Biol 2016; 1423:101-118. [PMID: 27142011 DOI: 10.1007/978-1-4939-3606-9_7] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/05/2023]
Abstract
The identification of conventional dendritic cells (cDCs) in the intestinal mucosa has been hampered by the difficulties associated with isolating cells from the intestine and by the fact that overlapping markers have made it complicated to discriminate them accurately from other intestinal mononuclear phagocytes such as macrophages (MFs). Here we detail the protocols we have developed to isolate live leukocytes from both murine and human small and large intestines and describe reliable strategies which can be used to identify bona fide cDCs in such preparations.
Collapse
Affiliation(s)
- Charlotte L Scott
- Centre for Immunobiology, Institute of Infection, Immunity and Inflammation, College of Veterinary, Medical and Life Sciences, University of Glasgow, Scotland, UK.
- Laboratory of Mucosal Immunology and Immunoregulation, Inflammation Research Centre (IRC), VIB Ghent University, Ghent (Zwijnaarde), Belgium.
- Department of Biomedical Molecular Biology, Ghent University Hospital, Ghent, Belgium.
| | - Pamela B Wright
- Centre for Immunobiology, Institute of Infection, Immunity and Inflammation, College of Veterinary, Medical and Life Sciences, University of Glasgow, Scotland, UK.
| | - Simon W F Milling
- Centre for Immunobiology, Institute of Infection, Immunity and Inflammation, College of Veterinary, Medical and Life Sciences, University of Glasgow, Scotland, UK
| | - Allan McI Mowat
- Centre for Immunobiology, Institute of Infection, Immunity and Inflammation, College of Veterinary, Medical and Life Sciences, University of Glasgow, Scotland, UK
| |
Collapse
|
159
|
Chessa F, Mathow D, Wang S, Hielscher T, Atzberger A, Porubsky S, Gretz N, Burgdorf S, Gröne HJ, Popovic ZV. The renal microenvironment modifies dendritic cell phenotype. Kidney Int 2016; 89:82-94. [DOI: 10.1038/ki.2015.292] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2015] [Revised: 07/20/2015] [Accepted: 08/06/2015] [Indexed: 12/20/2022]
|
160
|
Alexandre YO, Ghilas S, Sanchez C, Le Bon A, Crozat K, Dalod M. XCR1+ dendritic cells promote memory CD8+ T cell recall upon secondary infections with Listeria monocytogenes or certain viruses. J Exp Med 2015; 213:75-92. [PMID: 26694969 PMCID: PMC4710197 DOI: 10.1084/jem.20142350] [Citation(s) in RCA: 80] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2014] [Accepted: 11/20/2015] [Indexed: 12/30/2022] Open
Abstract
Alexandre et al. demonstrate the XCR1+ DCs are instrumental in memory CD8+ T cell responses to Listeria, VSV or vaccinia virus infection, but not CMV. Depending on the infection, robust memory CTL responses require cytokine- and chemokine-dependent cross-talk between XCR1+ DCs and NK cells or other IFN-γ–producing lymphocytes. Naive CD8+ T cell priming during tumor development or many primary infections requires cross-presentation by XCR1+ dendritic cells (DCs). Memory CD8+ T lymphocytes (mCTLs) harbor a lower activation threshold as compared with naive cells. However, whether their recall responses depend on XCR1+ DCs is unknown. By using a new mouse model allowing fluorescent tracking and conditional depletion of XCR1+ DCs, we demonstrate a differential requirement of these cells for mCTL recall during secondary infections by different pathogens. XCR1+ DCs were instrumental to promote this function upon secondary challenges with Listeria monocytogenes, vesicular stomatitis virus, or Vaccinia virus, but dispensable in the case of mouse cytomegalovirus. We deciphered how XCR1+ DCs promote mCTL recall upon secondary infections with Listeria. By visualizing for the first time the in vivo choreography of XCR1+ DCs, NK cells and mCTLs during secondary immune responses, and by neutralizing in vivo candidate molecules, we demonstrate that, very early after infection, mCTLs are activated, and attracted in a CXCR3-dependent manner, by NK cell–boosted, IL-12–, and CXCL9-producing XCR1+ DCs. Hence, depending on the infectious agent, strong recall of mCTLs during secondary challenges can require cytokine- and chemokine-dependent cross-talk with XCR1+ DCs and NK cells.
Collapse
Affiliation(s)
- Yannick O Alexandre
- Centre d'Immunologie de Marseille-Luminy, Aix Marseille Université UM2, Institut National de la Santé et de la Recherche Médicale, U1104, Centre National de la Recherche Scientifique UMR7280, 13288 Marseille, France
| | - Sonia Ghilas
- Centre d'Immunologie de Marseille-Luminy, Aix Marseille Université UM2, Institut National de la Santé et de la Recherche Médicale, U1104, Centre National de la Recherche Scientifique UMR7280, 13288 Marseille, France
| | - Cindy Sanchez
- Centre d'Immunologie de Marseille-Luminy, Aix Marseille Université UM2, Institut National de la Santé et de la Recherche Médicale, U1104, Centre National de la Recherche Scientifique UMR7280, 13288 Marseille, France
| | - Agnès Le Bon
- Institut Cochin, Institut National de la Santé et de la Recherche Médicale, U1016, Centre National de la Recherche Scientifique UMR8104, Université Paris Descartes, Sorbonne Paris Cité, 75014 Paris, France
| | - Karine Crozat
- Centre d'Immunologie de Marseille-Luminy, Aix Marseille Université UM2, Institut National de la Santé et de la Recherche Médicale, U1104, Centre National de la Recherche Scientifique UMR7280, 13288 Marseille, France
| | - Marc Dalod
- Centre d'Immunologie de Marseille-Luminy, Aix Marseille Université UM2, Institut National de la Santé et de la Recherche Médicale, U1104, Centre National de la Recherche Scientifique UMR7280, 13288 Marseille, France
| |
Collapse
|
161
|
Goubau D, van der Veen AG, Chakravarty P, Lin R, Rogers N, Rehwinkel J, Deddouche S, Rosewell I, Hiscott J, Reis e Sousa C. Mouse superkiller-2-like helicase DDX60 is dispensable for type I IFN induction and immunity to multiple viruses. Eur J Immunol 2015; 45:3386-403. [PMID: 26457795 PMCID: PMC4833184 DOI: 10.1002/eji.201545794] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2015] [Revised: 08/12/2015] [Accepted: 09/07/2015] [Indexed: 12/25/2022]
Abstract
IFN-α/β allow cells to fight virus infection by inducing the expression of many genes that encode effectors of antiviral defense. One of these, the Ski2-like DExH-box helicase DDX60, was recently implicated in resistance of human cells to hepatitis C virus, as well as in induction of IFN-α/β by retinoic acid inducible gene 1-like receptors (RLRs) that detect the presence of RNA viruses in a cell-intrinsic manner. Here, we sought to investigate the role of DDX60 in IFN-α/β induction and in resistance to virus infection. Analysis of fibroblasts and myeloid cells from Ddx60-deficient mice revealed no impairment in IFN-α/β production in response to RLR agonists, RNA viruses, or other stimuli. Moreover, overexpression of DDX60 did not potentiate IFN induction and DDX60 did not interact with RLRs or capture RLR agonists from virally infected cells. We also failed to identify any impairment in Ddx60-deficient murine cells or mice in resistance to infection with influenza A virus, encephalomyocarditis virus, Sindbis virus, vaccinia virus, or herpes simplex virus-1. These results put in question the reported role of DDX60 as a broad-acting positive regulator of RLR responses and hint at the possibility that it may function as a restriction factor highly specific for a particular virus or class of viruses.
Collapse
Affiliation(s)
- Delphine Goubau
- Immunobiology LaboratoryThe Francis Crick InstituteLincoln's Inn Fields Laboratory44 Lincoln's Inn FieldsLondonUK
| | - Annemarthe G. van der Veen
- Immunobiology LaboratoryThe Francis Crick InstituteLincoln's Inn Fields Laboratory44 Lincoln's Inn FieldsLondonUK
| | - Probir Chakravarty
- BioinformaticsThe Francis Crick Institute, Lincoln's Inn Fields LaboratoryLondonUK
| | - Rongtuan Lin
- Molecular Oncology GroupLady Davis Institute—Jewish General Hospital, McGill UniversityMontrealQuebecCanada
| | - Neil Rogers
- Immunobiology LaboratoryThe Francis Crick InstituteLincoln's Inn Fields Laboratory44 Lincoln's Inn FieldsLondonUK
| | - Jan Rehwinkel
- Immunobiology LaboratoryThe Francis Crick InstituteLincoln's Inn Fields Laboratory44 Lincoln's Inn FieldsLondonUK
| | - Safia Deddouche
- Immunobiology LaboratoryThe Francis Crick InstituteLincoln's Inn Fields Laboratory44 Lincoln's Inn FieldsLondonUK
| | - Ian Rosewell
- Transgenic ServicesThe Francis Crick Institute, Clare Hall LaboratoryPotters BarHertsUK
| | - John Hiscott
- Vaccine & Gene Therapy Institute of FloridaPort Saint LucieFLUSA
| | - Caetano Reis e Sousa
- Immunobiology LaboratoryThe Francis Crick InstituteLincoln's Inn Fields Laboratory44 Lincoln's Inn FieldsLondonUK
| |
Collapse
|
162
|
Böttcher JP, Zelenay S, Rogers NC, Helft J, Schraml BU, Reis e Sousa C. Oncogenic Transformation of Dendritic Cells and Their Precursors Leads to Rapid Cancer Development in Mice. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2015; 195:5066-76. [PMID: 26459350 PMCID: PMC4635568 DOI: 10.4049/jimmunol.1500889] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/16/2015] [Accepted: 09/15/2015] [Indexed: 12/23/2022]
Abstract
Dendritic cells (DCs) are powerful APCs that can induce Ag-specific adaptive immune responses and are increasingly recognized as important players in innate immunity to both infection and malignancy. Interestingly, although there are multiple described hematological malignancies, DC cancers are rarely observed in humans. Whether this is linked to the immunogenic potential of DCs, which might render them uniquely susceptible to immune control upon neoplastic transformation, has not been fully investigated. To address the issue, we generated a genetically engineered mouse model in which expression of Cre recombinase driven by the C-type lectin domain family 9, member a (Clec9a) locus causes expression of the Kirsten rat sarcoma viral oncogene homolog (Kras)(G12D) oncogenic driver and deletion of the tumor suppressor p53 within developing and differentiated DCs. We show that these Clec9a(Kras-G12D) mice rapidly succumb from disease and display massive accumulation of transformed DCs in multiple organs. In bone marrow chimeras, the development of DC cancer could be induced by a small number of transformed cells and was not prevented by the presence of untransformed DCs. Notably, activation of transformed DCs did not happen spontaneously but could be induced upon stimulation. Although Clec9a(Kras-G12D) mice showed altered thymic T cell development, peripheral T cells were largely unaffected during DC cancer development. Interestingly, transformed DCs were rejected upon adoptive transfer into wild-type but not lymphocyte-deficient mice, indicating that immunological control of DC cancer is in principle possible but does not occur during spontaneous generation in Clec9a(Kras-G12D) mice. Our findings suggest that neoplastic transformation of DCs does not by default induce anti-cancer immunity and can develop unhindered by immunological barriers.
Collapse
MESH Headings
- Animals
- Cell Transformation, Neoplastic/genetics
- Cell Transformation, Neoplastic/immunology
- Cell Transformation, Neoplastic/pathology
- Dendritic Cells/immunology
- Dendritic Cells/pathology
- Lectins, C-Type/genetics
- Lectins, C-Type/immunology
- Mice
- Mice, Transgenic
- Neoplasms, Experimental/genetics
- Neoplasms, Experimental/immunology
- Neoplasms, Experimental/pathology
- Proto-Oncogene Proteins p21(ras)/genetics
- Proto-Oncogene Proteins p21(ras)/immunology
- Rats
- Receptors, Immunologic/genetics
- Receptors, Immunologic/immunology
- Stem Cells/immunology
- Stem Cells/pathology
Collapse
Affiliation(s)
- Jan P Böttcher
- Immunobiology Laboratory, The Francis Crick Institute, Lincoln's Inn Fields Laboratory, London WC2A 3LY, United Kingdom
| | - Santiago Zelenay
- Immunobiology Laboratory, The Francis Crick Institute, Lincoln's Inn Fields Laboratory, London WC2A 3LY, United Kingdom
| | - Neil C Rogers
- Immunobiology Laboratory, The Francis Crick Institute, Lincoln's Inn Fields Laboratory, London WC2A 3LY, United Kingdom
| | - Julie Helft
- Immunobiology Laboratory, The Francis Crick Institute, Lincoln's Inn Fields Laboratory, London WC2A 3LY, United Kingdom
| | - Barbara U Schraml
- Immunobiology Laboratory, The Francis Crick Institute, Lincoln's Inn Fields Laboratory, London WC2A 3LY, United Kingdom
| | - Caetano Reis e Sousa
- Immunobiology Laboratory, The Francis Crick Institute, Lincoln's Inn Fields Laboratory, London WC2A 3LY, United Kingdom
| |
Collapse
|
163
|
Ludewig P, Gallizioli M, Urra X, Behr S, Brait VH, Gelderblom M, Magnus T, Planas AM. Dendritic cells in brain diseases. Biochim Biophys Acta Mol Basis Dis 2015; 1862:352-67. [PMID: 26569432 DOI: 10.1016/j.bbadis.2015.11.003] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2015] [Revised: 11/05/2015] [Accepted: 11/05/2015] [Indexed: 12/25/2022]
Affiliation(s)
- Peter Ludewig
- Department of Neurology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Mattia Gallizioli
- Department of Brain Ischemia and Neurodegeneration, Institut d'Investigacions Biomèdiques de Barcelona (IIBB), Consejo Superior de Investigaciones Científicas (CSIC), Barcelona, Spain
| | - Xabier Urra
- Functional Unit of Cerebrovascular Diseases, Hospital Clínic, Barcelona, Spain; August Pi i Sunyer Biomedical Research Institute (IDIBAPS), Barcelona, Spain
| | - Sarah Behr
- Department of Neurology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Vanessa H Brait
- August Pi i Sunyer Biomedical Research Institute (IDIBAPS), Barcelona, Spain
| | - Mathias Gelderblom
- Department of Neurology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Tim Magnus
- Department of Neurology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Anna M Planas
- Department of Brain Ischemia and Neurodegeneration, Institut d'Investigacions Biomèdiques de Barcelona (IIBB), Consejo Superior de Investigaciones Científicas (CSIC), Barcelona, Spain; August Pi i Sunyer Biomedical Research Institute (IDIBAPS), Barcelona, Spain.
| |
Collapse
|
164
|
Osorio F, Fuentes C, López MN, Salazar-Onfray F, González FE. Role of Dendritic Cells in the Induction of Lymphocyte Tolerance. Front Immunol 2015; 6:535. [PMID: 26539197 PMCID: PMC4611163 DOI: 10.3389/fimmu.2015.00535] [Citation(s) in RCA: 47] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2015] [Accepted: 10/02/2015] [Indexed: 01/07/2023] Open
Abstract
The ability of dendritic cells (DCs) to trigger tolerance or immunity is dictated by the context in which an antigen is encountered. A large body of evidence indicates that antigen presentation by steady-state DCs induces peripheral tolerance through mechanisms such as the secretion of soluble factors, the clonal deletion of autoreactive T cells, and feedback control of regulatory T cells. Moreover, recent understandings on the function of DC lineages and the advent of murine models of DC depletion have highlighted the contribution of DCs to lymphocyte tolerance. Importantly, these findings are now being applied to human research in the contexts of autoimmune diseases, allergies, and transplant rejection. Indeed, DC-based immunotherapy research has made important progress in the area of human health, particularly in regards to cancer. A better understanding of several DC-related aspects including the features of DC lineages, milieu composition, specific expression of surface molecules, the control of signaling responses, and the identification of competent stimuli able to trigger and sustain a tolerogenic outcome will contribute to the success of DC-based immunotherapy in the area of lymphocyte tolerance. This review will discuss the latest advances in the biology of DC subtypes related to the induction of regulatory T cells, in addition to presenting current ex vivo protocols for tolerogenic DC production. Particular attention will be given to the molecules and signals relevant for achieving an adequate tolerogenic response for the treatment of human pathologies.
Collapse
Affiliation(s)
- Fabiola Osorio
- Millennium Institute on Immunology and Immunotherapy, Faculty of Medicine, University of Chile , Santiago , Chile ; Disciplinary Program of Immunology, Institute of Biomedical Sciences, Faculty of Medicine, University of Chile , Santiago , Chile
| | - Camila Fuentes
- Millennium Institute on Immunology and Immunotherapy, Faculty of Medicine, University of Chile , Santiago , Chile ; Disciplinary Program of Immunology, Institute of Biomedical Sciences, Faculty of Medicine, University of Chile , Santiago , Chile
| | - Mercedes N López
- Millennium Institute on Immunology and Immunotherapy, Faculty of Medicine, University of Chile , Santiago , Chile ; Disciplinary Program of Immunology, Institute of Biomedical Sciences, Faculty of Medicine, University of Chile , Santiago , Chile ; Cell Therapy Laboratory, Blood Bank Service, University of Chile Clinical Hospital , Santiago , Chile
| | - Flavio Salazar-Onfray
- Millennium Institute on Immunology and Immunotherapy, Faculty of Medicine, University of Chile , Santiago , Chile ; Disciplinary Program of Immunology, Institute of Biomedical Sciences, Faculty of Medicine, University of Chile , Santiago , Chile
| | - Fermín E González
- Millennium Institute on Immunology and Immunotherapy, Faculty of Medicine, University of Chile , Santiago , Chile ; Laboratory of Experimental Immunology and Cancer, Faculty of Dentistry, University of Chile , Santiago , Chile
| |
Collapse
|
165
|
Tussiwand R, Gautier EL. Transcriptional Regulation of Mononuclear Phagocyte Development. Front Immunol 2015; 6:533. [PMID: 26539196 PMCID: PMC4609886 DOI: 10.3389/fimmu.2015.00533] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2015] [Accepted: 10/02/2015] [Indexed: 12/23/2022] Open
Abstract
Mononuclear phagocytes (MP) are a quite unique subset of hematopoietic cells, which comprise dendritic cells (DC), monocytes as well as monocyte-derived and tissue-resident macrophages. These cells are extremely diverse with regard to their origin, their phenotype as well as their function. Developmentally, DC and monocytes are constantly replenished from a bone marrow hematopoietic progenitor. The ontogeny of macrophages is more complex and is temporally linked and specified by the organ where they reside, occurring early during embryonic or perinatal life. The functional heterogeneity of MPs is certainly a consequence of the tissue of residence and also reflects the diverse ontogeny of the subsets. In this review, we will highlight the developmental pathways of murine MP, with a particular emphasis on the transcriptional factors that regulate their development and function. Finally, we will discuss and point out open questions in the field.
Collapse
Affiliation(s)
- Roxane Tussiwand
- Department of Biomedicine, University of Basel , Basel , Switzerland
| | - Emmanuel L Gautier
- INSERM UMR_S 1166, Sorbonne Universités, UPMC Univ Paris 06, Pitié-Salpêtrière Hospital , Paris , France
| |
Collapse
|
166
|
Abstract
The mononuclear phagocytes (dendritic cells and macrophages) are closely related immune cells with central roles in anti-infectious defense and maintenance of organ integrity. The canonical function of dendritic cells is the activation of T cells, whereas macrophages remove apoptotic cells and microbes by phagocytosis. In the kidney, these cell types form an intricate system of mononuclear phagocytes that surveys against injury and infection and contributes to organ homeostasis and tissue repair but may also promote progression of CKD. This review summarizes the general functions and classification of dendritic cells and macrophages in the immune system and recapitulates why overlapping definitions and historically separate research have created controversy about their tasks. Their roles in acute kidney disease, CKD, and renal transplantation are described, and therapeutic strategy to modify these cells for therapeutic purposes is discussed.
Collapse
Affiliation(s)
- Christina K Weisheit
- Institute of Experimental Immunology, University Clinic, Rheinische Friedrich-Wilhelms University, Bonn, Germany and Clinic for Anesthesiology and Intensive Care, University Clinic, Rheinische Friedrich-Wilhelms University, Bonn, Germany; and
| | - Daniel R Engel
- Institute of Experimental Immunology, University Clinic, Rheinische Friedrich-Wilhelms University, Bonn, Germany and Institute for Experimental Immunology and Imaging, University Duisburg-Essen and University Hospital Essen, Essen, Germany
| | - Christian Kurts
- Institute of Experimental Immunology, University Clinic, Rheinische Friedrich-Wilhelms University, Bonn, Germany and
| |
Collapse
|
167
|
PI3-Kinase-γ Has a Distinct and Essential Role in Lung-Specific Dendritic Cell Development. Immunity 2015; 43:674-89. [DOI: 10.1016/j.immuni.2015.09.006] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2014] [Revised: 04/21/2015] [Accepted: 07/10/2015] [Indexed: 12/23/2022]
|
168
|
Gottschalk C, Kurts C. The Debate about Dendritic Cells and Macrophages in the Kidney. Front Immunol 2015; 6:435. [PMID: 26388867 PMCID: PMC4556034 DOI: 10.3389/fimmu.2015.00435] [Citation(s) in RCA: 43] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2015] [Accepted: 08/11/2015] [Indexed: 11/13/2022] Open
Abstract
The mononuclear phagocyte system includes macrophages and dendritic cells (DCs), which are usually classified by morphology, phenotypical characteristics, and function. In the last decades, large research communities have gathered substantial knowledge on the roles of these cells in immune homeostasis and anti-infectious defense. However, these communities developed to a degree independent from each other, so that the nomenclature and functions of the numerous DC and macrophage subsets overlap, resulting in the present intense debate about the correct nomenclature. This controversy has also reached the field of experimental nephrology. At present, no mutually accepted way to distinguish renal DC and macrophages is available, so that many important roles in acute and chronic kidney disease have been ascribed to both DCs and macrophages. In this perspective article, we discuss the causes and consequences of the overlapping DC-macrophage classification systems, functional roles of DCs and macrophages, and the transferability of recent findings from other disciplines to the renal mononuclear phagocyte system from the nephrologist's point of view.
Collapse
Affiliation(s)
- Catherine Gottschalk
- Institute of Experimental Immunology, Rheinische Friedrich-Wilhelms-Universität Bonn , Bonn , Germany
| | - Christian Kurts
- Institute of Experimental Immunology, Rheinische Friedrich-Wilhelms-Universität Bonn , Bonn , Germany
| |
Collapse
|
169
|
Guilliams M, van de Laar L. A Hitchhiker's Guide to Myeloid Cell Subsets: Practical Implementation of a Novel Mononuclear Phagocyte Classification System. Front Immunol 2015; 6:406. [PMID: 26322042 PMCID: PMC4531301 DOI: 10.3389/fimmu.2015.00406] [Citation(s) in RCA: 97] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2015] [Accepted: 07/24/2015] [Indexed: 12/23/2022] Open
Abstract
The classification of mononuclear phagocytes as either dendritic cells or macrophages has been mainly based on morphology, the expression of surface markers, and assumed functional specialization. We have recently proposed a novel classification system of mononuclear phagocytes based on their ontogeny. Here, we discuss the practical application of such a classification system through a number of prototypical examples we have encountered while hitchhiking from one subset to another, across species and between steady-state and inflammatory settings. Finally, we discuss the advantages and drawbacks of such a classification system and propose a number of improvements to move from theoretical concepts to concrete guidelines.
Collapse
Affiliation(s)
- Martin Guilliams
- Laboratory of Immunoregulation, VIB Inflammation Research Center, Ghent University , Ghent , Belgium ; Department of Respiratory Medicine, University Hospital Ghent , Ghent , Belgium
| | - Lianne van de Laar
- Laboratory of Immunoregulation, VIB Inflammation Research Center, Ghent University , Ghent , Belgium ; Department of Respiratory Medicine, University Hospital Ghent , Ghent , Belgium
| |
Collapse
|
170
|
Quintana E, Fernández A, Velasco P, de Andrés B, Liste I, Sancho D, Gaspar ML, Cano E. DNGR-1(+) dendritic cells are located in meningeal membrane and choroid plexus of the noninjured brain. Glia 2015; 63:2231-48. [PMID: 26184558 DOI: 10.1002/glia.22889] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2014] [Revised: 06/17/2015] [Accepted: 06/24/2015] [Indexed: 12/14/2022]
Abstract
The role and different origin of brain myeloid cells in the brain is central to understanding how the central nervous system (CNS) responds to injury. C-type lectin receptor family 9, member A (DNGR-1/CLEC9A) is a marker of specific DC subsets that share functional similarities, such as CD8α(+) DCs in lymphoid tissues and CD103(+) CD11b(low) DCs in peripheral tissues. Here, we analyzed the presence of DNGR-1 in DCs present in the mouse brain (bDCs). Dngr-1/Clec9a mRNA is expressed mainly in the meningeal membranes and choroid plexus (m/Ch), and its expression is enhanced by fms-like tyrosine kinase 3 ligand (Flt3L), a cytokine involved in DC homeostasis. Using Clec9a(egfp/egfp) mice, we show that Flt3L induces accumulation of DNGR-1-EGFP(+) cells in the brain m/Ch. Most of these cells also express major histocompatibility complex class II (MHCII) molecules. We also observed an increase in specific markers of cDC CD8α+ cells such as Batf-3 and Irf-8, but not of costimulatory molecules such as Cd80 and Cd86, indicating an immature phenotype for these bDCs in the noninjured brain. The presence of DNGR-1 in the brain provides a potential marker for the study of this specific brain cell subset. Knowledge and targeting of brain antigen presenting cells (APCs) has implications for the fight against brain diseases such as neuroinflammation-based neurodegenerative diseases, microbe-induced encephalitis, and brain tumors such as gliomas.
Collapse
Affiliation(s)
- Elena Quintana
- Neuroinflammation Unit. Unidad Funcional De Investigación De Enfermedades Crónicas, Instituto De Salud Carlos III, Majadahonda, Madrid, Spain
| | - Andrés Fernández
- Neuroinflammation Unit. Unidad Funcional De Investigación De Enfermedades Crónicas, Instituto De Salud Carlos III, Majadahonda, Madrid, Spain
| | - Patricia Velasco
- Neuroinflammation Unit. Unidad Funcional De Investigación De Enfermedades Crónicas, Instituto De Salud Carlos III, Majadahonda, Madrid, Spain
| | - Belén de Andrés
- Department of Immunology, Centro Nacional De Microbiología, Instituto De Salud Carlos III, Madrid, Spain
| | - Isabel Liste
- Instituto De Salud Carlos III, Neural Regeneration Unit. Unidad Funcional De Investigación De Enfermedades Crónicas, Majadahonda, Madrid, Spain
| | - David Sancho
- Centro Nacional De Investigaciones Cardiovasculares (CNIC), Madrid, Spain
| | - María Luisa Gaspar
- Department of Immunology, Centro Nacional De Microbiología, Instituto De Salud Carlos III, Madrid, Spain
| | - Eva Cano
- Neuroinflammation Unit. Unidad Funcional De Investigación De Enfermedades Crónicas, Instituto De Salud Carlos III, Majadahonda, Madrid, Spain
| |
Collapse
|
171
|
Gutiérrez-Martínez E, Planès R, Anselmi G, Reynolds M, Menezes S, Adiko AC, Saveanu L, Guermonprez P. Cross-Presentation of Cell-Associated Antigens by MHC Class I in Dendritic Cell Subsets. Front Immunol 2015; 6:363. [PMID: 26236315 PMCID: PMC4505393 DOI: 10.3389/fimmu.2015.00363] [Citation(s) in RCA: 117] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2015] [Accepted: 07/05/2015] [Indexed: 12/12/2022] Open
Abstract
Dendritic cells (DCs) have the unique ability to pick up dead cells carrying antigens in tissue and migrate to the lymph nodes where they can cross-present cell-associated antigens by MHC class I to CD8+ T cells. There is strong in vivo evidence that the mouse XCR1+ DCs subset acts as a key player in this process. The intracellular processes underlying cross-presentation remain controversial and several pathways have been proposed. Indeed, a wide number of studies have addressed the cellular process of cross-presentation in vitro using a variety of sources of antigen and antigen-presenting cells. Here, we review the in vivo and in vitro evidence supporting the current mechanistic models and disscuss their physiological relevance to the cross-presentation of cell-associated antigens by DCs subsets.
Collapse
Affiliation(s)
- Enric Gutiérrez-Martínez
- Laboratory of Phagocyte Immunobiology, Peter Gorer Department of Immunobiology, CMCBI, King's College London , London , UK
| | - Remi Planès
- Laboratory of Phagocyte Immunobiology, Peter Gorer Department of Immunobiology, CMCBI, King's College London , London , UK
| | - Giorgio Anselmi
- Laboratory of Phagocyte Immunobiology, Peter Gorer Department of Immunobiology, CMCBI, King's College London , London , UK
| | - Matthew Reynolds
- Laboratory of Phagocyte Immunobiology, Peter Gorer Department of Immunobiology, CMCBI, King's College London , London , UK
| | - Shinelle Menezes
- Laboratory of Phagocyte Immunobiology, Peter Gorer Department of Immunobiology, CMCBI, King's College London , London , UK
| | - Aimé Cézaire Adiko
- Laboratory of Phagocyte Immunobiology, Peter Gorer Department of Immunobiology, Centre for Molecular & Cellular Biology of Inflammation (CMCBI), King's College London , Paris , France ; Sorbonne Paris Cité, Université Paris Diderot , Paris , France
| | - Loredana Saveanu
- Laboratory of Phagocyte Immunobiology, Peter Gorer Department of Immunobiology, Centre for Molecular & Cellular Biology of Inflammation (CMCBI), King's College London , Paris , France ; Sorbonne Paris Cité, Université Paris Diderot , Paris , France
| | - Pierre Guermonprez
- Laboratory of Phagocyte Immunobiology, Peter Gorer Department of Immunobiology, CMCBI, King's College London , London , UK
| |
Collapse
|
172
|
Chousterman BG, Boissonnas A, Poupel L, Baudesson de Chanville C, Adam J, Tabibzadeh N, Licata F, Lukaszewicz AC, Lombès A, Deterre P, Payen D, Combadière C. Ly6Chigh Monocytes Protect against Kidney Damage during Sepsis via a CX3CR1-Dependent Adhesion Mechanism. J Am Soc Nephrol 2015; 27:792-803. [PMID: 26160897 DOI: 10.1681/asn.2015010009] [Citation(s) in RCA: 52] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2015] [Accepted: 05/20/2015] [Indexed: 12/24/2022] Open
Abstract
Monocytes have a crucial role in both proinflammatory and anti-inflammatory phenomena occurring during sepsis. Monocyte recruitment and activation are orchestrated by the chemokine receptors CX3CR1 and CCR2 and their cognate ligands. However, little is known about the roles of these cells and chemokines during the acute phase of inflammation in sepsis. Using intravital microscopy in a murine model of polymicrobial sepsis, we showed that inflammatory Ly6C(high) monocytes infiltrated kidneys, exhibited altered motility, and adhered strongly to the renal vascular wall in a chemokine receptor CX3CR1-dependent manner. Adoptive transfer of Cx3cr1-proficient monocyte-enriched bone marrow cells into septic Cx3cr1-depleted mice prevented kidney damage and promoted mouse survival. Modulation of CX3CR1 activation in septic mice controlled monocyte adhesion, regulated proinflammatory and anti-inflammatory cytokine expression, and was associated with the extent of kidney lesions such that the number of lesions decreased when CX3CR1 activity increased. Consistent with these results, the pro-adhesive I249 CX3CR1 allele in humans was associated with a lower incidence of AKI in patients with sepsis. These data show that inflammatory monocytes have a protective effect during sepsis via a CX3CR1-dependent adhesion mechanism. This receptor might be a new therapeutic target for kidney injury during sepsis.
Collapse
Affiliation(s)
- Benjamin G Chousterman
- Sorbonne Universités, Université Pierre et Marie Curie (UPMC), University of Paris 06, Paris, France; Institut National de la Santé et de la Recherche Médicale (INSERM), U1135, Paris, France; Centre National de la Recherche Scientifique (CNRS), Paris, France; Département d'Anesthésie-Réanimation-Service d'Aide Médicale Urgente (SMUR), Hôpital Lariboisière, Assistance Publique-Hôpitaux de Paris, Paris, France
| | - Alexandre Boissonnas
- Sorbonne Universités, Université Pierre et Marie Curie (UPMC), University of Paris 06, Paris, France; Institut National de la Santé et de la Recherche Médicale (INSERM), U1135, Paris, France; Centre National de la Recherche Scientifique (CNRS), Paris, France;
| | - Lucie Poupel
- Sorbonne Universités, Université Pierre et Marie Curie (UPMC), University of Paris 06, Paris, France; Institut National de la Santé et de la Recherche Médicale (INSERM), U1135, Paris, France; Centre National de la Recherche Scientifique (CNRS), Paris, France
| | - Camille Baudesson de Chanville
- Sorbonne Universités, Université Pierre et Marie Curie (UPMC), University of Paris 06, Paris, France; Institut National de la Santé et de la Recherche Médicale (INSERM), U1135, Paris, France; Centre National de la Recherche Scientifique (CNRS), Paris, France
| | - Julien Adam
- Institut Gustave-Roussy, Université Paris-Sud Villejuif, France
| | - Nahid Tabibzadeh
- Sorbonne Universités, Université Pierre et Marie Curie (UPMC), University of Paris 06, Paris, France; Service des Explorations Fonctionnelles and Institut National de la Santé et de la Recherche Médicale, Assistance Publique-Hôpitaux de Paris, Hôpital Tenon, Paris, France; and
| | - Fabrice Licata
- Sorbonne Universités, Université Pierre et Marie Curie (UPMC), University of Paris 06, Paris, France; Institut National de la Santé et de la Recherche Médicale (INSERM), U1135, Paris, France; Centre National de la Recherche Scientifique (CNRS), Paris, France
| | - Anne-Claire Lukaszewicz
- Département d'Anesthésie-Réanimation-Service d'Aide Médicale Urgente (SMUR), Hôpital Lariboisière, Assistance Publique-Hôpitaux de Paris, Paris, France; INSERM, U1160, Paris, France
| | - Amélie Lombès
- Sorbonne Universités, Université Pierre et Marie Curie (UPMC), University of Paris 06, Paris, France; Institut National de la Santé et de la Recherche Médicale (INSERM), U1135, Paris, France; Centre National de la Recherche Scientifique (CNRS), Paris, France
| | - Philippe Deterre
- Sorbonne Universités, Université Pierre et Marie Curie (UPMC), University of Paris 06, Paris, France; Institut National de la Santé et de la Recherche Médicale (INSERM), U1135, Paris, France; Centre National de la Recherche Scientifique (CNRS), Paris, France
| | - Didier Payen
- Département d'Anesthésie-Réanimation-Service d'Aide Médicale Urgente (SMUR), Hôpital Lariboisière, Assistance Publique-Hôpitaux de Paris, Paris, France; INSERM, U1160, Paris, France
| | - Christophe Combadière
- Sorbonne Universités, Université Pierre et Marie Curie (UPMC), University of Paris 06, Paris, France; Institut National de la Santé et de la Recherche Médicale (INSERM), U1135, Paris, France; Centre National de la Recherche Scientifique (CNRS), Paris, France;
| |
Collapse
|
173
|
Heung LJ, Jhingran A, Hohl TM. Deploying FLAREs to Visualize Functional Outcomes of Host-Pathogen Encounters. PLoS Pathog 2015; 11:e1004912. [PMID: 26158781 PMCID: PMC4497593 DOI: 10.1371/journal.ppat.1004912] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Affiliation(s)
- Lena J. Heung
- Infectious Diseases Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, New York, United States of America
| | - Anupam Jhingran
- Infectious Diseases Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, New York, United States of America
| | - Tobias M. Hohl
- Infectious Diseases Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, New York, United States of America
- Immunology Program, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, New York, United States of America
- * E-mail:
| |
Collapse
|
174
|
Reynolds G, Haniffa M. Human and Mouse Mononuclear Phagocyte Networks: A Tale of Two Species? Front Immunol 2015; 6:330. [PMID: 26124761 PMCID: PMC4479794 DOI: 10.3389/fimmu.2015.00330] [Citation(s) in RCA: 93] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2015] [Accepted: 06/12/2015] [Indexed: 12/22/2022] Open
Abstract
Dendritic cells (DCs), monocytes, and macrophages are a heterogeneous population of mononuclear phagocytes that are involved in antigen processing and presentation to initiate and regulate immune responses to pathogens, vaccines, tumor, and tolerance to self. In addition to their afferent sentinel function, DCs and macrophages are also critical as effectors and coordinators of inflammation and homeostasis in peripheral tissues. Harnessing DCs and macrophages for therapeutic purposes has major implications for infectious disease, vaccination, transplantation, tolerance induction, inflammation, and cancer immunotherapy. There has been a paradigm shift in our understanding of the developmental origin and function of the cellular constituents of the mononuclear phagocyte system. Significant progress has been made in tandem in both human and mouse mononuclear phagocyte biology. This progress has been accelerated by comparative biology analysis between mouse and human, which has proved to be an exceptionally fruitful strategy to harmonize findings across species. Such analyses have provided unexpected insights and facilitated productive reciprocal and iterative processes to inform our understanding of human and mouse mononuclear phagocytes. In this review, we discuss the strategies, power, and utility of comparative biology approaches to integrate recent advances in human and mouse mononuclear phagocyte biology and its potential to drive forward clinical translation of this knowledge. We also present a functional framework on the parallel organization of human and mouse mononuclear phagocyte networks.
Collapse
Affiliation(s)
- Gary Reynolds
- Human Dendritic Cell Laboratory, Institute of Cellular Medicine, Newcastle University , Newcastle upon Tyne , UK ; Musculoskeletal Research Group, Institute of Cellular Medicine, Newcastle University , Newcastle upon Tyne , UK
| | - Muzlifah Haniffa
- Human Dendritic Cell Laboratory, Institute of Cellular Medicine, Newcastle University , Newcastle upon Tyne , UK
| |
Collapse
|
175
|
Scott CL, Henri S, Guilliams M. Mononuclear phagocytes of the intestine, the skin, and the lung. Immunol Rev 2015; 262:9-24. [PMID: 25319324 DOI: 10.1111/imr.12220] [Citation(s) in RCA: 75] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Tissues that are in direct contact with the outside world face particular immunological challenges. The intestine, the skin, and the lung possess important mononuclear phagocyte populations to deal with these challenges, but the cellular origin of these phagocytes is strikingly different from one subset to another, with some cells derived from embryonic precursors and some from bone marrow-derived circulating monocytes. Here, we review the current knowledge regarding the developmental pathways that control the differentiation of mononuclear phagocytes in these barrier tissues. We have also attempted to build a theoretical model that could explain the distinct cellular origin of mononuclear phagocytes in these tissues.
Collapse
Affiliation(s)
- Charlotte L Scott
- Laboratory of Immunoregulation, VIB Inflammation Research Center, Ghent, Belgium; Department of Respiratory Medicine, Ghent University, Ghent, Belgium; Centre for Immunobiology, Institute of Infection, Immunity and Inflammation, University of Glasgow, Glasgow, UK
| | | | | |
Collapse
|
176
|
Helft J, Böttcher J, Chakravarty P, Zelenay S, Huotari J, Schraml BU, Goubau D, Reis e Sousa C. GM-CSF Mouse Bone Marrow Cultures Comprise a Heterogeneous Population of CD11c(+)MHCII(+) Macrophages and Dendritic Cells. Immunity 2015; 42:1197-211. [PMID: 26084029 DOI: 10.1016/j.immuni.2015.05.018] [Citation(s) in RCA: 605] [Impact Index Per Article: 67.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2014] [Revised: 03/04/2015] [Accepted: 03/31/2015] [Indexed: 12/23/2022]
Abstract
Dendritic cells (DCs) are key players in the immune system. Much of their biology has been elucidated via culture systems in which hematopoietic precursors differentiate into DCs under the aegis of cytokines. A widely used protocol involves the culture of murine bone marrow (BM) cells with granulocyte-macrophage colony-stimulating factor (GM-CSF) to generate BM-derived DCs (BMDCs). BMDCs express CD11c and MHC class II (MHCII) molecules and share with DCs isolated from tissues the ability to present exogenous antigens to T cells and to respond to microbial stimuli by undergoing maturation. We demonstrate that CD11c(+)MHCII(+) BMDCs are in fact a heterogeneous group of cells that comprises conventional DCs and monocyte-derived macrophages. DCs and macrophages in GM-CSF cultures both undergo maturation upon stimulation with lipopolysaccharide but respond differentially to the stimulus and remain separable entities. These results have important implications for the interpretation of a vast array of data obtained with DC culture systems.
Collapse
Affiliation(s)
- Julie Helft
- Immunobiology Laboratory, The Francis Crick Institute, Lincoln's Inn Fields Laboratory, 44 Lincoln's Inn Fields, London WC2A 3LY, UK
| | - Jan Böttcher
- Immunobiology Laboratory, The Francis Crick Institute, Lincoln's Inn Fields Laboratory, 44 Lincoln's Inn Fields, London WC2A 3LY, UK
| | - Probir Chakravarty
- Bioinformatics Laboratory, The Francis Crick Institute, Lincoln's Inn Fields Laboratory, 44 Lincoln's Inn Fields, London WC2A 3LY, UK
| | - Santiago Zelenay
- Immunobiology Laboratory, The Francis Crick Institute, Lincoln's Inn Fields Laboratory, 44 Lincoln's Inn Fields, London WC2A 3LY, UK
| | - Jatta Huotari
- Immunobiology Laboratory, The Francis Crick Institute, Lincoln's Inn Fields Laboratory, 44 Lincoln's Inn Fields, London WC2A 3LY, UK
| | - Barbara U Schraml
- Immunobiology Laboratory, The Francis Crick Institute, Lincoln's Inn Fields Laboratory, 44 Lincoln's Inn Fields, London WC2A 3LY, UK
| | - Delphine Goubau
- Immunobiology Laboratory, The Francis Crick Institute, Lincoln's Inn Fields Laboratory, 44 Lincoln's Inn Fields, London WC2A 3LY, UK
| | - Caetano Reis e Sousa
- Immunobiology Laboratory, The Francis Crick Institute, Lincoln's Inn Fields Laboratory, 44 Lincoln's Inn Fields, London WC2A 3LY, UK.
| |
Collapse
|
177
|
Vu Manh TP, Bertho N, Hosmalin A, Schwartz-Cornil I, Dalod M. Investigating Evolutionary Conservation of Dendritic Cell Subset Identity and Functions. Front Immunol 2015; 6:260. [PMID: 26082777 PMCID: PMC4451681 DOI: 10.3389/fimmu.2015.00260] [Citation(s) in RCA: 73] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2015] [Accepted: 05/11/2015] [Indexed: 12/14/2022] Open
Abstract
Dendritic cells (DCs) were initially defined as mononuclear phagocytes with a dendritic morphology and an exquisite efficiency for naïve T-cell activation. DC encompass several subsets initially identified by their expression of specific cell surface molecules and later shown to excel in distinct functions and to develop under the instruction of different transcription factors or cytokines. Very few cell surface molecules are expressed in a specific manner on any immune cell type. Hence, to identify cell types, the sole use of a small number of cell surface markers in classical flow cytometry can be deceiving. Moreover, the markers currently used to define mononuclear phagocyte subsets vary depending on the tissue and animal species studied and even between laboratories. This has led to confusion in the definition of DC subset identity and in their attribution of specific functions. There is a strong need to identify a rigorous and consensus way to define mononuclear phagocyte subsets, with precise guidelines potentially applicable throughout tissues and species. We will discuss the advantages, drawbacks, and complementarities of different methodologies: cell surface phenotyping, ontogeny, functional characterization, and molecular profiling. We will advocate that gene expression profiling is a very rigorous, largely unbiased and accessible method to define the identity of mononuclear phagocyte subsets, which strengthens and refines surface phenotyping. It is uniquely powerful to yield new, experimentally testable, hypotheses on the ontogeny or functions of mononuclear phagocyte subsets, their molecular regulation, and their evolutionary conservation. We propose defining cell populations based on a combination of cell surface phenotyping, expression analysis of hallmark genes, and robust functional assays, in order to reach a consensus and integrate faster the huge but scattered knowledge accumulated by different laboratories on different cell types, organs, and species.
Collapse
Affiliation(s)
- Thien-Phong Vu Manh
- UM2, Centre d'Immunologie de Marseille-Luminy (CIML), Aix-Marseille University , Marseille , France ; U1104, Institut National de la Santé et de la Recherche Médicale (INSERM) , Marseille , France ; UMR7280, Centre National de la Recherche Scientifique (CNRS) , Marseille , France
| | - Nicolas Bertho
- Virologie et Immunologie Moléculaires UR892, Institut National de la Recherche Agronomique , Jouy-en-Josas , France
| | - Anne Hosmalin
- INSERM U1016, Institut Cochin , Paris , France ; CNRS UMR8104 , Paris , France ; Université Paris Descartes , Paris , France ; Assistance Publique-Hôpitaux de Paris (AP-HP), Hôpital Cochin , Paris , France
| | - Isabelle Schwartz-Cornil
- Virologie et Immunologie Moléculaires UR892, Institut National de la Recherche Agronomique , Jouy-en-Josas , France
| | - Marc Dalod
- UM2, Centre d'Immunologie de Marseille-Luminy (CIML), Aix-Marseille University , Marseille , France ; U1104, Institut National de la Santé et de la Recherche Médicale (INSERM) , Marseille , France ; UMR7280, Centre National de la Recherche Scientifique (CNRS) , Marseille , France
| |
Collapse
|
178
|
Rutkowski MR, Svoronos N, Perales-Puchalt A, Conejo-Garcia JR. The Tumor Macroenvironment: Cancer-Promoting Networks Beyond Tumor Beds. Adv Cancer Res 2015. [PMID: 26216635 DOI: 10.1016/bs.acr.2015.04.011] [Citation(s) in RCA: 45] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
During tumor progression, alterations within the systemic tumor environment, or macroenvironment, result in the promotion of tumor growth, tumor invasion to distal organs, and eventual metastatic disease. Distally produced hormones, commensal microbiota residing within mucosal surfaces, myeloid cells and even the bone marrow impact the systemic immune system, tumor growth, and metastatic spread. Understanding the reciprocal interactions between the cells and soluble factors within the macroenvironment and the primary tumor will enable the design of specific therapies that have the potential to prevent dissemination and metastatic spread. This chapter will summarize recent findings detailing how the primary tumor and systemic tumor macroenvironment coordinate malignant progression.
Collapse
Affiliation(s)
- Melanie R Rutkowski
- Tumor Microenvironment and Metastasis Program, The Wistar Institute, Philadelphia, Pennsylvania, USA
| | - Nikolaos Svoronos
- Tumor Microenvironment and Metastasis Program, The Wistar Institute, Philadelphia, Pennsylvania, USA
| | - Alfredo Perales-Puchalt
- Tumor Microenvironment and Metastasis Program, The Wistar Institute, Philadelphia, Pennsylvania, USA
| | - Jose R Conejo-Garcia
- Tumor Microenvironment and Metastasis Program, The Wistar Institute, Philadelphia, Pennsylvania, USA.
| |
Collapse
|
179
|
Abstract
Dendritic cells (DCs) are a heterogeneous group of mononuclear phagocytes with versatile roles in immunity. They are classified predominantly based on phenotypic and functional properties, namely their stellate morphology, expression of the integrin CD11c, and major histocompatibility class II molecules, as well as their superior capacity to migrate to secondary lymphoid organs and stimulate naïve T cells. However, these attributes are not exclusive to DCs and often change within inflammatory or infectious environments. This led to debates over cell identification and questioned even the mere existence of DCs as distinct leukocyte lineage. Here, we review experimental approaches taken to fate map DCs and discuss how these have shaped our understanding of DC ontogeny and lineage affiliation. Considering the ontogenetic properties of DCs will help to overcome the inherent shortcomings of purely phenotypic- and function-based approaches to cell definition and will yield a more robust way of DC classification.
Collapse
Affiliation(s)
- Mateusz Pawel Poltorak
- Institute for Medical Microbiology, Immunology and Hygiene, Technische Universität München , Munich , Germany
| | - Barbara Ursula Schraml
- Institute for Medical Microbiology, Immunology and Hygiene, Technische Universität München , Munich , Germany
| |
Collapse
|
180
|
Toward defining a ‘lineage’ – The case for dendritic cells. Semin Cell Dev Biol 2015; 41:3-8. [DOI: 10.1016/j.semcdb.2015.02.004] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2014] [Accepted: 02/10/2015] [Indexed: 12/23/2022]
|
181
|
Schlitzer A, McGovern N, Ginhoux F. Dendritic cells and monocyte-derived cells: Two complementary and integrated functional systems. Semin Cell Dev Biol 2015; 41:9-22. [DOI: 10.1016/j.semcdb.2015.03.011] [Citation(s) in RCA: 158] [Impact Index Per Article: 17.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2014] [Revised: 03/27/2015] [Accepted: 03/31/2015] [Indexed: 12/23/2022]
|
182
|
Eckert C, Klein N, Kornek M, Lukacs-Kornek V. The complex myeloid network of the liver with diverse functional capacity at steady state and in inflammation. Front Immunol 2015; 6:179. [PMID: 25941527 PMCID: PMC4403526 DOI: 10.3389/fimmu.2015.00179] [Citation(s) in RCA: 54] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2015] [Accepted: 03/30/2015] [Indexed: 12/23/2022] Open
Abstract
In recent years, it has been an explosion of information regarding the role of various myeloid cells in liver pathology. Macrophages and dendritic cell (DC) play crucial roles in multiple chronic liver diseases such as fibrosis and non-alcoholic fatty liver disease (NAFLD). The complexity of myeloid cell populations and the missing exclusive marker combination make the interpretation of the data often extremely difficult. The current review aims to summarize the multiple roles of macrophages and DCs in chronic liver diseases, especially pointing out how these cells influence liver immune and parenchymal cells thereby altering liver function and pathology. Moreover, the review outlines the currently known marker combinations for the identification of these cell populations for the study of their role in liver immunology.
Collapse
Affiliation(s)
- Christoph Eckert
- Department of Medicine II, Saarland University Medical Center , Homburg , Germany
| | - Niklas Klein
- Department of Medicine II, Saarland University Medical Center , Homburg , Germany
| | - Miroslaw Kornek
- Department of Medicine II, Saarland University Medical Center , Homburg , Germany
| | | |
Collapse
|
183
|
Batal I, De Serres SA, Safa K, Bijol V, Ueno T, Onozato ML, Iafrate AJ, Herter JM, Lichtman AH, Mayadas TN, Guleria I, Rennke HG, Najafian N, Chandraker A. Dendritic Cells in Kidney Transplant Biopsy Samples Are Associated with T Cell Infiltration and Poor Allograft Survival. J Am Soc Nephrol 2015; 26:3102-13. [PMID: 25855773 DOI: 10.1681/asn.2014080804] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2014] [Accepted: 02/03/2015] [Indexed: 12/16/2022] Open
Abstract
Progress in long-term renal allograft survival continues to lag behind the progress in short-term transplant outcomes. Dendritic cells are the most efficient antigen-presenting cells, but surprisingly little attention has been paid to their presence in transplanted kidneys. We used dendritic cell-specific intercellular adhesion molecule-3-grabbing nonintegrin as a marker of dendritic cells in 105 allograft biopsy samples from 105 kidney transplant recipients. High dendritic cell density was associated with poor allograft survival independent of clinical variables. Moreover, high dendritic cell density correlated with greater T cell proliferation and poor outcomes in patients with high total inflammation scores, including inflammation in areas of tubular atrophy. We then explored the association between dendritic cells and histologic variables associated with poor prognosis. Multivariate analysis revealed an independent association between the densities of dendritic cells and T cells. In biopsy samples with high dendritic cell density, electron microscopy showed direct physical contact between infiltrating lymphocytes and cells that have the ultrastructural morphologic characteristics of dendritic cells. The origin of graft dendritic cells was sought in nine sex-mismatched recipients using XY fluorescence in situ hybridization. Whereas donor dendritic cells predominated initially, the majority of dendritic cells in late allograft biopsy samples were of recipient origin. Our data highlight the prognostic value of dendritic cell density in allograft biopsy samples, suggest a new role for these cells in shaping graft inflammation, and provide a rationale for targeting dendritic cell recruitment to promote long-term allograft survival.
Collapse
Affiliation(s)
- Ibrahim Batal
- Department of Pathology, Brigham and Women's Hospital and Harvard Medical School, Boston, Massachusetts;
| | - Sacha A De Serres
- Renal Division, Schuster Family Transplantation Research Center, Brigham and Women's Hospital, Boston Children's Hospital, and Harvard Medical School, Boston, Massachusetts; and
| | - Kassem Safa
- Renal Division, Schuster Family Transplantation Research Center, Brigham and Women's Hospital, Boston Children's Hospital, and Harvard Medical School, Boston, Massachusetts; and
| | - Vanesa Bijol
- Department of Pathology, Brigham and Women's Hospital and Harvard Medical School, Boston, Massachusetts
| | - Takuya Ueno
- Renal Division, Schuster Family Transplantation Research Center, Brigham and Women's Hospital, Boston Children's Hospital, and Harvard Medical School, Boston, Massachusetts; and
| | - Maristela L Onozato
- Department of Pathology, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts
| | - A John Iafrate
- Department of Pathology, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts
| | - Jan M Herter
- Department of Pathology, Brigham and Women's Hospital and Harvard Medical School, Boston, Massachusetts
| | - Andrew H Lichtman
- Department of Pathology, Brigham and Women's Hospital and Harvard Medical School, Boston, Massachusetts
| | - Tanya N Mayadas
- Department of Pathology, Brigham and Women's Hospital and Harvard Medical School, Boston, Massachusetts
| | - Indira Guleria
- Renal Division, Schuster Family Transplantation Research Center, Brigham and Women's Hospital, Boston Children's Hospital, and Harvard Medical School, Boston, Massachusetts; and
| | - Helmut G Rennke
- Department of Pathology, Brigham and Women's Hospital and Harvard Medical School, Boston, Massachusetts
| | - Nader Najafian
- Renal Division, Schuster Family Transplantation Research Center, Brigham and Women's Hospital, Boston Children's Hospital, and Harvard Medical School, Boston, Massachusetts; and
| | - Anil Chandraker
- Renal Division, Schuster Family Transplantation Research Center, Brigham and Women's Hospital, Boston Children's Hospital, and Harvard Medical School, Boston, Massachusetts; and
| |
Collapse
|
184
|
Koscsó B, Gowda K, Schell TD, Bogunovic M. Purification of dendritic cell and macrophage subsets from the normal mouse small intestine. J Immunol Methods 2015; 421:1-13. [PMID: 25796561 DOI: 10.1016/j.jim.2015.02.013] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2014] [Revised: 02/27/2015] [Accepted: 02/28/2015] [Indexed: 12/13/2022]
Abstract
Mononuclear phagocytes are essential for protecting against pathogens breaching the intestinal mucosa and maintaining the integrity of the gastrointestinal tract. The mononuclear phagocyte family of the healthy intestine is represented by a small population of hematopoietic cells including dendritic cells and macrophages. Distinct mononuclear phagocyte subsets strategically accumulate within and below the mucosal epithelium and are distributed in the submucosa and muscularis externa. Shaped by its unique microenvironment, each mononuclear phagocyte subset is developmentally and functionally unique and phenotypically distinct. Here we summarize our recent advances on identifying and purifying various intestinal mononuclear phagocyte subsets by flow cytometry in the context of their developmental properties and location within the intestinal tissue.
Collapse
Affiliation(s)
- Balázs Koscsó
- Department of Microbiology and Immunology, Penn State University College of Medicine and Milton S. Hershey Medical Center, Hershey, PA 17033, USA
| | - Kavitha Gowda
- Department of Microbiology and Immunology, Penn State University College of Medicine and Milton S. Hershey Medical Center, Hershey, PA 17033, USA
| | - Todd D Schell
- Department of Microbiology and Immunology, Penn State University College of Medicine and Milton S. Hershey Medical Center, Hershey, PA 17033, USA
| | - Milena Bogunovic
- Department of Microbiology and Immunology, Penn State University College of Medicine and Milton S. Hershey Medical Center, Hershey, PA 17033, USA.
| |
Collapse
|
185
|
Nakano H, Moran TP, Nakano K, Gerrish KE, Bortner CD, Cook DN. Complement receptor C5aR1/CD88 and dipeptidyl peptidase-4/CD26 define distinct hematopoietic lineages of dendritic cells. THE JOURNAL OF IMMUNOLOGY 2015; 194:3808-19. [PMID: 25769922 DOI: 10.4049/jimmunol.1402195] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/27/2014] [Accepted: 02/13/2015] [Indexed: 12/17/2022]
Abstract
Differential display of the integrins CD103 and CD11b are widely used to distinguish two major dendritic cell (DC) subsets in nonlymphoid tissues. CD103(+) DCs arise from FLT3-dependent DC precursors (preDCs), whereas CD11b(hi) DCs can arise either from preDCs or FLT3-independent monocytes. Functional characterization of these two lineages of CD11b(hi) DCs has been hindered by the lack of a widely applicable method to distinguish between them. We performed gene expression analysis of fractionated lung DCs from C57BL/6 mice and found that monocyte-derived DCs (moDCs), including CD11b(hi)Ly-6C(lo) tissue-resident and CD11b(hi)Ly-6C(hi) inflammatory moDCs, express the complement 5a receptor 1/CD88, whereas preDC-derived conventional DCs (cDCs), including CD103(+) and CD11b(hi) cDCs, express dipeptidyl peptidase-4/CD26. Flow cytometric analysis of multiple organs, including the kidney, liver, lung, lymph nodes, small intestine, and spleen, confirmed that reciprocal display of CD88 and CD26 can reliably distinguish FLT3-independent moDCs from FLT3-dependent cDCs in C57BL/6 mice. Similar results were obtained when DCs from BALB/c mice were analyzed. Using this novel approach to study DCs in mediastinal lymph nodes, we observed that most blood-derived lymph node-resident DCs, as well as tissue-derived migratory DCs, are cDCs. Furthermore, cDCs, but not moDCs, stimulated naive T cell proliferation. We anticipate that the use of Abs against CD88 and CD26 to distinguish moDCs and cDCs in multiple organs and mouse strains will facilitate studies aimed at assigning specific functions to distinct DC lineages in immune responses.
Collapse
Affiliation(s)
- Hideki Nakano
- Immunity, Inflammation and Disease Laboratory, Division of Intramural Research, National Institute of Environmental Health Sciences, National Institutes of Health, Research Triangle Park, NC 27709;
| | - Timothy P Moran
- Immunity, Inflammation and Disease Laboratory, Division of Intramural Research, National Institute of Environmental Health Sciences, National Institutes of Health, Research Triangle Park, NC 27709; Division of Allergy and Immunology, Department of Pediatrics, Duke University Medical Center, Durham, NC 27705
| | - Keiko Nakano
- Immunity, Inflammation and Disease Laboratory, Division of Intramural Research, National Institute of Environmental Health Sciences, National Institutes of Health, Research Triangle Park, NC 27709
| | - Kevin E Gerrish
- Molecular Genetics Core Laboratory, Division of Intramural Research, National Institute of Environmental Health Sciences, National Institutes of Health, Research Triangle Park, NC 27709; and
| | - Carl D Bortner
- Signal Transduction Laboratory, Division of Intramural Research, National Institute of Environmental Health Sciences, National Institutes of Health, Research Triangle Park, NC 27709
| | - Donald N Cook
- Immunity, Inflammation and Disease Laboratory, Division of Intramural Research, National Institute of Environmental Health Sciences, National Institutes of Health, Research Triangle Park, NC 27709
| |
Collapse
|
186
|
Kopf M, Schneider C, Nobs SP. The development and function of lung-resident macrophages and dendritic cells. Nat Immunol 2015; 16:36-44. [PMID: 25521683 DOI: 10.1038/ni.3052] [Citation(s) in RCA: 388] [Impact Index Per Article: 43.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2014] [Accepted: 11/10/2014] [Indexed: 12/12/2022]
Abstract
Gas exchange is the vital function of the lungs. It occurs in the alveoli, where oxygen and carbon dioxide diffuse across the alveolar epithelium and the capillary endothelium surrounding the alveoli, separated only by a fused basement membrane 0.2-0.5 μm in thickness. This tenuous barrier is exposed to dangerous or innocuous particles, toxins, allergens and infectious agents inhaled with the air or carried in the blood. The lung immune system has evolved to ward off pathogens and restrain inflammation-mediated damage to maintain gas exchange. Lung-resident macrophages and dendritic cells are located in close proximity to the epithelial surface of the respiratory system and the capillaries to sample and examine the air-borne and blood-borne material. In communication with alveolar epithelial cells, they set the threshold and the quality of the immune response.
Collapse
Affiliation(s)
- Manfred Kopf
- Institute of Molecular Health Sciences, Department of Biology, Swiss Federal Institute of Technology Zurich, Zurich, Switzerland
| | - Christoph Schneider
- Institute of Molecular Health Sciences, Department of Biology, Swiss Federal Institute of Technology Zurich, Zurich, Switzerland
| | - Samuel P Nobs
- Institute of Molecular Health Sciences, Department of Biology, Swiss Federal Institute of Technology Zurich, Zurich, Switzerland
| |
Collapse
|
187
|
Li J, Ahmet F, Sullivan LC, Brooks AG, Kent SJ, De Rose R, Salazar AM, Reis e Sousa C, Shortman K, Lahoud MH, Heath WR, Caminschi I. Antibodies targeting Clec9A promote strong humoral immunity without adjuvant in mice and non-human primates. Eur J Immunol 2015; 45:854-64. [PMID: 25487143 DOI: 10.1002/eji.201445127] [Citation(s) in RCA: 68] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2014] [Revised: 11/04/2014] [Accepted: 12/03/2014] [Indexed: 11/07/2022]
Abstract
Targeting antigens to dendritic cell (DC) surface receptors using antibodies has been successfully used to generate strong immune responses and is currently in clinical trials for cancer immunotherapy. Whilst cancer immunotherapy focuses on the induction of CD8(+) T-cell responses, many successful vaccines to pathogens or their toxins utilize humoral immunity as the primary effector mechanism. Universally, these approaches have used adjuvants or pathogen material that augment humoral responses. However, adjuvants are associated with safety issues. One approach, successfully used in the mouse, to generate strong humoral responses in the absence of adjuvant is to target antigen to Clec9A, also known as DNGR-1, a receptor on CD8α(+) DCs. Here, we address two issues relating to clinical application. First, we address the issue of variable adjuvant-dependence for different antibodies targeting mouse Clec9A. We show that multiple sites on Clec9A can be successfully targeted, but that strong in vivo binding and provision of suitable helper T cell determinants was essential for efficacy. Second, we show that induction of humoral immunity to CLEC9A-targeted antigens is extremely effective in nonhuman primates, in an adjuvant-free setting. Our findings support extending this vaccination approach to humans and offer important insights into targeting design.
Collapse
Affiliation(s)
- Jessica Li
- Centre for Biomedical Research, Burnet Institute, Melbourne, Victoria, Australia; Department of Microbiology and Immunology, Peter Doherty Institute for Infection and Immunity, The University of Melbourne, Victoria, Australia
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
188
|
Alpaerts K, Buckinx R, Adriaensen D, Van Nassauw L, Timmermans JP. Identification and Putative Roles of Distinct Subtypes of Intestinal Dendritic Cells in Neuroimmune Communication: What can be Learned from Other Organ Systems? Anat Rec (Hoboken) 2015; 298:903-16. [DOI: 10.1002/ar.23106] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2014] [Revised: 09/13/2014] [Accepted: 11/08/2014] [Indexed: 12/13/2022]
Affiliation(s)
- Katrien Alpaerts
- Laboratory of Cell biology and Histology; Department of Veterinary Sciences; University of Antwerp; Antwerp Belgium
| | - Roeland Buckinx
- Laboratory of Cell biology and Histology; Department of Veterinary Sciences; University of Antwerp; Antwerp Belgium
| | - Dirk Adriaensen
- Laboratory of Cell biology and Histology; Department of Veterinary Sciences; University of Antwerp; Antwerp Belgium
| | - Luc Van Nassauw
- Laboratory of Human Anatomy and Embryology; Faculty of Medicine and Health Sciences; University of Antwerp; Antwerp Belgium
| | - Jean-Pierre Timmermans
- Laboratory of Cell biology and Histology; Department of Veterinary Sciences; University of Antwerp; Antwerp Belgium
| |
Collapse
|
189
|
Defining dendritic cells. Curr Opin Immunol 2015; 32:13-20. [DOI: 10.1016/j.coi.2014.11.001] [Citation(s) in RCA: 126] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2014] [Revised: 11/04/2014] [Accepted: 11/17/2014] [Indexed: 12/23/2022]
|
190
|
Bekiaris V, Persson EK, Agace WW. Intestinal dendritic cells in the regulation of mucosal immunity. Immunol Rev 2015; 260:86-101. [PMID: 24942684 DOI: 10.1111/imr.12194] [Citation(s) in RCA: 108] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
The intestine presents a huge surface area to the outside environment, a property that is of critical importance for its key functions in nutrient digestion, absorption, and waste disposal. As such, the intestine is constantly exposed to dietary and microbial-derived foreign antigens, to which immune cells within the mucosa must suitably respond to maintain intestinal integrity, while also providing the ability to mount effective immune responses to potential pathogens. Dendritic cells (DCs) are sentinel immune cells that play a central role in the initiation and differentiation of adaptive immune responses. In the intestinal mucosa, DCs are located diffusely throughout the intestinal lamina propria, within gut-associated lymphoid tissues, including Peyer's patches and smaller lymphoid aggregates, as well as in intestinal-draining lymph nodes, including mesenteric lymph nodes. The recognition that dietary nutrients and microbial communities in the intestine influence both mucosal and systemic immune cell development and function as well as immune-mediated disease has led to an explosion of literature in mucosal immunology in recent years and a growing interest in the functionality of intestinal DCs. In the current review, we discuss recent findings from our group and others that have provided important insights regarding murine and human intestinal lamina propria DCs and highlighted marked developmental and functional heterogeneity within this compartment. A thorough understanding of the role these subsets play in the regulation of intestinal immune homeostasis and inflammation will help to define novel strategies for the treatment of intestinal pathologies and contribute to improved rational design of mucosal vaccines.
Collapse
|
191
|
Abstract
The intestine contains the largest pool of macrophages in the body which are essential for maintaining mucosal homeostasis in the face of the microbiota and the constant need for epithelial renewal but are also important components of protective immunity and are involved in the pathology of inflammatory bowel disease (IBD). However, defining the biological roles of intestinal macrophages has been impeded by problems in defining the phenotype and origins of different populations of myeloid cells in the mucosa. Here, we discuss how multiple parameters can be used in combination to discriminate between functionally distinct myeloid cells and discuss the roles of macrophages during homeostasis and how these may change when inflammation ensues. We also discuss the evidence that intestinal macrophages do not fit the current paradigm that tissue-resident macrophages are derived from embryonic precursors that self-renew in situ, but require constant replenishment by blood monocytes. We describe our recent work demonstrating that classical monocytes constantly enter the intestinal mucosa and how the environment dictates their subsequent fate. We believe that understanding the factors that drive intestinal macrophage development in the steady state and how these may change in response to pathogens or inflammation could provide important insights into the treatment of IBD.
Collapse
Affiliation(s)
- Calum C Bain
- Centre for Immunobiology, Institute of Infection, Immunity and Inflammation, University of Glasgow, Glasgow, UK
| | | |
Collapse
|
192
|
Abstract
The intestine contains the largest pool of macrophages in the body which are essential for maintaining mucosal homeostasis in the face of the microbiota and the constant need for epithelial renewal but are also important components of protective immunity and are involved in the pathology of inflammatory bowel disease (IBD). However, defining the biological roles of intestinal macrophages has been impeded by problems in defining the phenotype and origins of different populations of myeloid cells in the mucosa. Here, we discuss how multiple parameters can be used in combination to discriminate between functionally distinct myeloid cells and discuss the roles of macrophages during homeostasis and how these may change when inflammation ensues. We also discuss the evidence that intestinal macrophages do not fit the current paradigm that tissue-resident macrophages are derived from embryonic precursors that self-renew in situ, but require constant replenishment by blood monocytes. We describe our recent work demonstrating that classical monocytes constantly enter the intestinal mucosa and how the environment dictates their subsequent fate. We believe that understanding the factors that drive intestinal macrophage development in the steady state and how these may change in response to pathogens or inflammation could provide important insights into the treatment of IBD.
Collapse
Affiliation(s)
- Calum C Bain
- Centre for Immunobiology, Institute of Infection, Immunity and Inflammation, University of Glasgow, Glasgow, UK
| | | |
Collapse
|
193
|
Engel DR, Krause TA, Snelgrove SL, Thiebes S, Hickey MJ, Boor P, Kitching AR, Kurts C. CX3CR1 reduces kidney fibrosis by inhibiting local proliferation of profibrotic macrophages. THE JOURNAL OF IMMUNOLOGY 2015; 194:1628-38. [PMID: 25595779 DOI: 10.4049/jimmunol.1402149] [Citation(s) in RCA: 58] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
A dense network of macrophages and dendritic cells (DC) expressing the chemokine receptor CX3CR1 populates most tissues. We recently reported that CX3CR1 regulates the abundance of CD11c(+) DC in the kidney and thereby promotes renal inflammation in glomerulonephritis. Given that chronic inflammation usually causes fibrosis, we hypothesized that CX3CR1 deficiency should attenuate renal fibrosis. However, when we tested this hypothesis using the DC-independent murine fibrosis model of unilateral ureteral obstruction, kidney fibrosis was unexpectedly more severe, despite less intrarenal inflammation. Two-photon imaging and flow cytometry revealed in kidneys of CX3CR1-deficient mice more motile Ly6C/Gr-1(+) macrophages. Flow cytometry verified that renal macrophages were more abundant in the absence of CX3CR1 and produced more of the key profibrotic mediator, TGF-β. Macrophages accumulated because of higher intrarenal proliferation, despite reduced monocyte recruitment and higher signs of apoptosis within the kidney. These findings support the theory that tissue macrophage numbers are regulated through local proliferation and identify CX3CR1 as a regulator of such proliferation. Thus, CX3CR1 inhibition should be avoided in DC-independent inflammatory diseases because it may promote fibrosis.
Collapse
Affiliation(s)
- Daniel R Engel
- Institute of Experimental Immunology, Rheinische Friedrich-Wilhelms University, 53105 Bonn, Germany; Institute for Experimental Immunology and Imaging, University Duisburg-Essen and University Hospital Essen, 45147 Essen, Germany
| | - Torsten A Krause
- Institute of Experimental Immunology, Rheinische Friedrich-Wilhelms University, 53105 Bonn, Germany
| | - Sarah L Snelgrove
- Centre for Inflammatory Diseases, Department of Medicine, Monash University, Clayton, Victoria 3168, Australia
| | - Stephanie Thiebes
- Institute of Experimental Immunology, Rheinische Friedrich-Wilhelms University, 53105 Bonn, Germany
| | - Michael J Hickey
- Centre for Inflammatory Diseases, Department of Medicine, Monash University, Clayton, Victoria 3168, Australia
| | - Peter Boor
- Institute of Pathology, Rheinisch-Westfälische Technische Hochschule, 52074 Aachen, Germany; and Department of Nephrology, Rheinisch-Westfälische Technische Hochschule, 52074 Aachen, Germany
| | - A Richard Kitching
- Centre for Inflammatory Diseases, Department of Medicine, Monash University, Clayton, Victoria 3168, Australia
| | - Christian Kurts
- Institute of Experimental Immunology, Rheinische Friedrich-Wilhelms University, 53105 Bonn, Germany;
| |
Collapse
|
194
|
van Blijswijk J, Schraml BU, Rogers NC, Whitney PG, Zelenay S, Acton SE, Reis e Sousa C. Altered lymph node composition in diphtheria toxin receptor-based mouse models to ablate dendritic cells. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2015; 194:307-15. [PMID: 25411201 PMCID: PMC4272857 DOI: 10.4049/jimmunol.1401999] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Dendritic cells (DCs) are key regulators of innate and adaptive immunity. Our understanding of immune function has benefited greatly from mouse models allowing for selective ablation of DCs. Many such models rely on transgenic diphtheria toxin receptor (DTR) expression driven by DC-restricted promoters. This renders DCs sensitive to DT but is otherwise thought to have no effect on immune physiology. In this study, we report that, unexpectedly, mice in which DTR is expressed on conventional DCs display marked lymph node (LN) hypocellularity and reduced frequency of DCs in the same organs but not in spleen or nonlymphoid tissues. Intriguingly, in mixed bone marrow chimeras the phenotype conferred by DTR-expressing DCs is dominant over control bone marrow-derived cells, leading to small LNs and an overall paucity of DCs independently of the genetic ability to express DTR. The finding of alterations in LN composition and size independently of DT challenge suggests that caution must be exercised when interpreting results of experiments obtained with mouse models to inducibly deplete DCs. It further indicates that DTR, a member of the epidermal growth factor family, is biologically active in mice. Its use in cell ablation experiments needs to be considered in light of this activity.
Collapse
Affiliation(s)
- Janneke van Blijswijk
- Immunobiology Laboratory, Cancer Research UK, London Research Institute, London WC2A 3LY, United Kingdom
| | - Barbara U Schraml
- Immunobiology Laboratory, Cancer Research UK, London Research Institute, London WC2A 3LY, United Kingdom
| | - Neil C Rogers
- Immunobiology Laboratory, Cancer Research UK, London Research Institute, London WC2A 3LY, United Kingdom
| | - Paul G Whitney
- Immunobiology Laboratory, Cancer Research UK, London Research Institute, London WC2A 3LY, United Kingdom
| | - Santiago Zelenay
- Immunobiology Laboratory, Cancer Research UK, London Research Institute, London WC2A 3LY, United Kingdom
| | - Sophie E Acton
- Immunobiology Laboratory, Cancer Research UK, London Research Institute, London WC2A 3LY, United Kingdom
| | - Caetano Reis e Sousa
- Immunobiology Laboratory, Cancer Research UK, London Research Institute, London WC2A 3LY, United Kingdom
| |
Collapse
|
195
|
|
196
|
Dambuza IM, Brown GD. C-type lectins in immunity: recent developments. Curr Opin Immunol 2014; 32:21-7. [PMID: 25553393 PMCID: PMC4589735 DOI: 10.1016/j.coi.2014.12.002] [Citation(s) in RCA: 333] [Impact Index Per Article: 33.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2014] [Revised: 12/03/2014] [Accepted: 12/10/2014] [Indexed: 12/18/2022]
Abstract
CLRs play an essential role in immunity to fungi and mycobacteria. CLRs are involved in the regulation of homeostasis, autoimmunity and allergy. CLRs recognise and trigger cellular responses to dead and cancerous cells.
C-type lectin receptors (CLRs) comprise a large superfamily of proteins, which recognise a diverse range of ligands, and are defined by the presence of at least one C-type lectin-like domain (CTLD). Of particular interest are the single extracellular CTLD-containing receptors of the ‘Dectin-1’ and ‘Dectin-2’ clusters, which associate with signalling adaptors or possess integral intracellular signalling domains. These CLRs have traditionally been associated with the recognition of fungi, but recent discoveries have revealed diverse and unexpected functions. In this review, we describe their newly identified roles in anti-microbial host defence, homeostasis, autoimmunity, allergy and their functions in the recognition and response to dead and cancerous cells.
Collapse
Affiliation(s)
- Ivy M Dambuza
- Aberdeen Fungal Group, Division of Applied Medicine, Immunity, Infection and Inflammation Programme, Institute of Medical Sciences, University of Aberdeen, Aberdeen AB25 2ZD, UK
| | - Gordon D Brown
- Aberdeen Fungal Group, Division of Applied Medicine, Immunity, Infection and Inflammation Programme, Institute of Medical Sciences, University of Aberdeen, Aberdeen AB25 2ZD, UK.
| |
Collapse
|
197
|
Fossum E, Grødeland G, Terhorst D, Tveita AA, Vikse E, Mjaaland S, Henri S, Malissen B, Bogen B. Vaccine molecules targeting Xcr1 on cross-presenting DCs induce protective CD8+T-cell responses against influenza virus. Eur J Immunol 2014; 45:624-35. [DOI: 10.1002/eji.201445080] [Citation(s) in RCA: 79] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2014] [Revised: 10/20/2014] [Accepted: 11/17/2014] [Indexed: 12/24/2022]
Affiliation(s)
- Even Fossum
- K.G. Jebsen Center for Influenza Vaccine Research; Institute of Immunology; University of Oslo and Oslo University Hospital; Oslo Norway
| | - Gunnveig Grødeland
- K.G. Jebsen Center for Influenza Vaccine Research; Institute of Immunology; University of Oslo and Oslo University Hospital; Oslo Norway
| | - Dorothea Terhorst
- Department of Dermatology; Charité University Medicine Berlin; Berlin Germany
- Centre d'Immunologie de Marseille-Luminy (CIML); Aix-Marseille Université; Marseille France
- INSERM U1104; Marseille France
- CNRS UMR7280; Marseille France
| | - Anders A. Tveita
- Center for Immune Regulation; Institute of Immunology; University of Oslo and Oslo University Hospital; Oslo Norway
| | - Elisabeth Vikse
- K.G. Jebsen Center for Influenza Vaccine Research; Institute of Immunology; University of Oslo and Oslo University Hospital; Oslo Norway
| | - Siri Mjaaland
- K.G. Jebsen Center for Influenza Vaccine Research; Institute of Immunology; University of Oslo and Oslo University Hospital; Oslo Norway
- Division for Infectious Disease Control; Department of Bacteriology and Infection Immunology; Norwegian Institute of Public Health; Oslo Norway
| | - Sandrine Henri
- Centre d'Immunologie de Marseille-Luminy (CIML); Aix-Marseille Université; Marseille France
| | - Bernard Malissen
- Centre d'Immunologie de Marseille-Luminy (CIML); Aix-Marseille Université; Marseille France
| | - Bjarne Bogen
- K.G. Jebsen Center for Influenza Vaccine Research; Institute of Immunology; University of Oslo and Oslo University Hospital; Oslo Norway
- Center for Immune Regulation; Institute of Immunology; University of Oslo and Oslo University Hospital; Oslo Norway
| |
Collapse
|
198
|
Bloy N, Pol J, Aranda F, Eggermont A, Cremer I, Fridman WH, Fučíková J, Galon J, Tartour E, Spisek R, Dhodapkar MV, Zitvogel L, Kroemer G, Galluzzi L. Trial watch: Dendritic cell-based anticancer therapy. Oncoimmunology 2014; 3:e963424. [PMID: 25941593 DOI: 10.4161/21624011.2014.963424] [Citation(s) in RCA: 61] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2014] [Accepted: 09/05/2014] [Indexed: 02/06/2023] Open
Abstract
The use of patient-derived dendritic cells (DCs) as a means to elicit therapeutically relevant immune responses in cancer patients has been extensively investigated throughout the past decade. In this context, DCs are generally expanded, exposed to autologous tumor cell lysates or loaded with specific tumor-associated antigens (TAAs), and then reintroduced into patients, often in combination with one or more immunostimulatory agents. As an alternative, TAAs are targeted to DCs in vivo by means of monoclonal antibodies, carbohydrate moieties or viral vectors specific for DC receptors. All these approaches have been shown to (re)activate tumor-specific immune responses in mice, often mediating robust therapeutic effects. In 2010, the first DC-based preparation (sipuleucel-T, also known as Provenge®) has been approved by the US Food and Drug Administration (FDA) for use in humans. Reflecting the central position occupied by DCs in the regulation of immunological tolerance and adaptive immunity, the interest in harnessing them for the development of novel immunotherapeutic anticancer regimens remains high. Here, we summarize recent advances in the preclinical and clinical development of DC-based anticancer therapeutics.
Collapse
Key Words
- DC, dendritic cell
- DC-based vaccination
- FDA, Food and Drug Administration
- IFN, interferon
- MRC1, mannose receptor, C type 1
- MUC1, mucin 1
- TAA, tumor-associated antigen
- TLR, Toll-like receptor
- Toll-like receptor agonists
- Treg, regulatory T cell
- WT1, Wilms tumor 1
- antigen cross-presentation
- autophagy
- iDC, immature DC
- immunogenic cell death
- mDC, mature DC
- pDC, plasmacytoid DC
- regulatory T cells
Collapse
Affiliation(s)
- Norma Bloy
- Gustave Roussy Cancer Campus ; Villejuif, France ; INSERM , U1138; Paris France ; Equipe 11 labellisée par la Ligue Nationale contre le Cancer; Centre de Recherche des Cordeliers ; Paris France ; Université Paris-Sud/Paris XI ; Orsay, France
| | - Jonathan Pol
- Gustave Roussy Cancer Campus ; Villejuif, France ; INSERM , U1138; Paris France ; Equipe 11 labellisée par la Ligue Nationale contre le Cancer; Centre de Recherche des Cordeliers ; Paris France
| | - Fernando Aranda
- Gustave Roussy Cancer Campus ; Villejuif, France ; INSERM , U1138; Paris France ; Equipe 11 labellisée par la Ligue Nationale contre le Cancer; Centre de Recherche des Cordeliers ; Paris France
| | | | - Isabelle Cremer
- INSERM , U1138; Paris France ; Equipe 13; Centre de Recherche des Cordeliers ; Paris France ; Université Pierre et Marie Curie/Paris VI ; Paris France
| | - Wolf Hervé Fridman
- INSERM , U1138; Paris France ; Equipe 13; Centre de Recherche des Cordeliers ; Paris France ; Université Pierre et Marie Curie/Paris VI ; Paris France
| | - Jitka Fučíková
- Department of Immunology; 2nd Medical School Charles University and University Hospital Motol ; Prague, Czech Republic ; Sotio a.s. ; Prague, Czech Republic
| | - Jérôme Galon
- INSERM , U1138; Paris France ; Université Pierre et Marie Curie/Paris VI ; Paris France ; Laboratory of Integrative Cancer Immunology; Centre de Recherche des Cordeliers ; Paris France ; Université Paris Descartes/Paris V; Sorbonne Paris Cité ; Paris France
| | - Eric Tartour
- Université Paris Descartes/Paris V; Sorbonne Paris Cité ; Paris France ; INSERM , U970; Paris France ; Pôle de Biologie; Hôpital Européen Georges Pompidou, AP-HP ; Paris France
| | - Radek Spisek
- Department of Immunology; 2nd Medical School Charles University and University Hospital Motol ; Prague, Czech Republic ; Sotio a.s. ; Prague, Czech Republic
| | - Madhav V Dhodapkar
- Department of Medicine; Immunobiology and Yale Cancer Center; Yale University ; New Haven, CT USA
| | - Laurence Zitvogel
- Gustave Roussy Cancer Campus ; Villejuif, France ; INSERM, U1015, CICBT507 ; Villejuif, France
| | - Guido Kroemer
- INSERM , U1138; Paris France ; Equipe 11 labellisée par la Ligue Nationale contre le Cancer; Centre de Recherche des Cordeliers ; Paris France ; Université Paris Descartes/Paris V; Sorbonne Paris Cité ; Paris France ; Pôle de Biologie; Hôpital Européen Georges Pompidou, AP-HP ; Paris France ; Metabolomics and Cell Biology Platforms; Gustave Roussy Cancer Campus ; Villejuif, France
| | - Lorenzo Galluzzi
- Gustave Roussy Cancer Campus ; Villejuif, France ; INSERM , U1138; Paris France ; Equipe 11 labellisée par la Ligue Nationale contre le Cancer; Centre de Recherche des Cordeliers ; Paris France ; Université Paris Descartes/Paris V; Sorbonne Paris Cité ; Paris France
| |
Collapse
|
199
|
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: 2.6] [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
|
200
|
den Haan JM, Arens R, van Zelm MC. The activation of the adaptive immune system: Cross-talk between antigen-presenting cells, T cells and B cells. Immunol Lett 2014; 162:103-12. [DOI: 10.1016/j.imlet.2014.10.011] [Citation(s) in RCA: 81] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
|