51
|
Devi KSP, Anandasabapathy N. The origin of DCs and capacity for immunologic tolerance in central and peripheral tissues. Semin Immunopathol 2016; 39:137-152. [PMID: 27888331 DOI: 10.1007/s00281-016-0602-0] [Citation(s) in RCA: 55] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2016] [Accepted: 10/28/2016] [Indexed: 12/20/2022]
Abstract
Dendritic cells (DCs) are specialized immune sentinels that play key role in maintaining immune homeostasis by efficiently regulating the delicate balance between protective immunity and tolerance to self. Although DCs respond to maturation signals present in the surrounding milieu, multiple layers of suppression also co-exist that reduce the infringement of tolerance against self-antigens. These tolerance inducing properties of DCs are governed by their origin and a range of other factors including distribution, cytokines, growth factors, and transcriptional programing, that collectively impart suppressive functions to these cells. DCs directing tolerance secrete anti-inflammatory cytokines and induce naïve T cells or B cells to differentiate into regulatory T cells (Tregs) or B cells. In this review, we provide a detailed outlook on the molecular mechanisms that induce functional specialization to govern central or peripheral tolerance. The tolerance-inducing nature of DCs can be exploited to overcome autoimmunity and rejection in graft transplantation.
Collapse
Affiliation(s)
- K Sanjana P Devi
- Department of Dermatology/Harvard Skin Disease Research Center, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Niroshana Anandasabapathy
- Department of Dermatology/Harvard Skin Disease Research Center, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
52
|
Single-cell RNA-seq identifies a PD-1hi ILC progenitor and defines its development pathway. Nature 2016; 539:102-106. [DOI: 10.1038/nature20105] [Citation(s) in RCA: 205] [Impact Index Per Article: 25.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2016] [Accepted: 09/22/2016] [Indexed: 02/06/2023]
|
53
|
Li HS, Liu C, Xiao Y, Chu F, Liang X, Peng W, Hu J, Neelapu SS, Sun SC, Hwu P, Watowich SS. Bypassing STAT3-mediated inhibition of the transcriptional regulator ID2 improves the antitumor efficacy of dendritic cells. Sci Signal 2016; 9:ra94. [PMID: 27678219 PMCID: PMC5061503 DOI: 10.1126/scisignal.aaf3957] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Despite the potent ability of dendritic cells (DCs) to stimulate lymphocyte responses and host immunity, granulocyte-macrophage colony-stimulating factor-derived DCs (GM-DCs) used as antitumor vaccines have demonstrated relatively modest success in cancer immunotherapy. We found that injecting GM-DCs into melanoma tumors in mice, or culturing GM-DCs with melanoma-secreted cytokines or melanoma-conditioned medium, rapidly suppressed DC-intrinsic expression of the gene encoding inhibitor of differentiation 2 (ID2), a transcriptional regulator. Melanoma-associated cytokines repressed Id2 transcription in murine DCs through the activation of signal transducer and activator of transcription 3 (STAT3). Enforced expression of ID2 in GM-DCs (ID2-GM-DCs) suppressed their production of the proinflammatory cytokine tumor necrosis factor-α (TNF-α). Vaccination with ID2-GM-DCs slowed the progression of melanoma tumors and enhanced animal survival, which was associated with an increased abundance of tumor-infiltrating interferon-γ-positive CD4(+) effector and CD8(+) cytotoxic T cells and a decreased number of tumor-infiltrating regulatory CD4(+) T cells. The efficacy of the ID2-GM-DC vaccine was improved by combinatorial treatment with a blocking antibody to programmed cell death protein-1 (PD-1), a current immunotherapy that overcomes suppressive immune checkpoint signaling. Collectively, our data reveal a previously unrecognized STAT3-mediated immunosuppressive mechanism in DCs and indicate that DC-intrinsic ID2 promotes tumor immunity by modulating tumor-associated CD4(+) T cell responses. Thus, inhibiting STAT3 or overexpressing ID2 selectively in DCs may improve the efficiency of DC vaccines in cancer therapy.
Collapse
Affiliation(s)
- Haiyan S Li
- Department of Immunology, University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Chengwen Liu
- Department of Melanoma Medical Oncology, University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Yichuan Xiao
- Department of Immunology, University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Fuliang Chu
- Department of Lymphoma and Myeloma, University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Xiaoxuan Liang
- Department of Immunology, University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Weiyi Peng
- Department of Melanoma Medical Oncology, University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Jianhua Hu
- Department of Biostatistics, University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Sattva S Neelapu
- Department of Lymphoma and Myeloma, University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Shao-Cong Sun
- Department of Immunology, University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA. University of Texas Graduate School of Biomedical Sciences, Houston, TX 77030, USA
| | - Patrick Hwu
- Department of Melanoma Medical Oncology, University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA. University of Texas Graduate School of Biomedical Sciences, Houston, TX 77030, USA
| | - Stephanie S Watowich
- Department of Immunology, University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA. University of Texas Graduate School of Biomedical Sciences, Houston, TX 77030, USA.
| |
Collapse
|
54
|
CXCR5+ follicular cytotoxic T cells control viral infection in B cell follicles. Nat Immunol 2016; 17:1187-96. [DOI: 10.1038/ni.3543] [Citation(s) in RCA: 315] [Impact Index Per Article: 39.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2016] [Accepted: 07/29/2016] [Indexed: 12/12/2022]
|
55
|
Ghisi M, Kats L, Masson F, Li J, Kratina T, Vidacs E, Gilan O, Doyle MA, Newbold A, Bolden JE, Fairfax KA, de Graaf CA, Firth M, Zuber J, Dickins RA, Corcoran LM, Dawson MA, Belz GT, Johnstone RW. Id2 and E Proteins Orchestrate the Initiation and Maintenance of MLL-Rearranged Acute Myeloid Leukemia. Cancer Cell 2016; 30:59-74. [PMID: 27374225 DOI: 10.1016/j.ccell.2016.05.019] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/11/2015] [Revised: 02/29/2016] [Accepted: 05/31/2016] [Indexed: 12/19/2022]
Abstract
E proteins and their antagonists, the Id proteins, are transcriptional regulators important for normal hematopoiesis. We found that Id2 acts as a key regulator of leukemia stem cell (LSC) potential in MLL-rearranged acute myeloid leukemia (AML). Low endogenous Id2 expression is associated with LSC enrichment while Id2 overexpression impairs MLL-AF9-leukemia initiation and growth. Importantly, MLL-AF9 itself controls the E-protein pathway by suppressing Id2 while directly activating E2-2 expression, and E2-2 depletion phenocopies Id2 overexpression in MLL-AF9-AML cells. Remarkably, Id2 tumor-suppressive function is conserved in t(8;21) AML. Low expression of Id2 and its associated gene signature are associated with poor prognosis in MLL-rearranged and t(8;21) AML patients, identifying the Id2/E-protein axis as a promising new therapeutic target in AML.
Collapse
MESH Headings
- Animals
- Cell Proliferation
- Chromosomes, Human, Pair 21/genetics
- Chromosomes, Human, Pair 8/genetics
- Gene Expression Regulation, Leukemic
- Humans
- Inhibitor of Differentiation Protein 2/genetics
- Inhibitor of Differentiation Protein 2/metabolism
- Leukemia, Myeloid, Acute/genetics
- Leukemia, Myeloid, Acute/metabolism
- Leukemia, Myeloid, Acute/pathology
- Mice
- Myeloid-Lymphoid Leukemia Protein/genetics
- Myeloid-Lymphoid Leukemia Protein/metabolism
- Neoplasms, Experimental
- Oncogene Proteins, Fusion/genetics
- Oncogene Proteins, Fusion/metabolism
- Prognosis
- Stem Cells/cytology
- Stem Cells/metabolism
- Survival Analysis
- Transcription Factor 7-Like 2 Protein/genetics
- Transcription Factor 7-Like 2 Protein/metabolism
- Translocation, Genetic
Collapse
Affiliation(s)
- Margherita Ghisi
- Cancer Therapeutics Program, Peter MacCallum Cancer Centre, St. Andrews Place, East Melbourne, 3002 VIC, Australia; Australian Centre for Blood Diseases, Monash University, Melbourne, 3004 VIC, Australia.
| | - Lev Kats
- Cancer Therapeutics Program, Peter MacCallum Cancer Centre, St. Andrews Place, East Melbourne, 3002 VIC, Australia; The Sir Peter MacCallum Department of Oncology, University of Melbourne, Parkville, 3010 VIC, Australia
| | - Frederick Masson
- Cancer Inflammation Laboratory, Olivia Newton-John Cancer Research Institute and La Trobe University School of Cancer Medicine, Heidelberg, 3084 VIC, Australia; Molecular Immunology Division, The Walter and Eliza Hall Institute of Medical Research, Parkville, 3052 VIC, Australia; Department of Medical Biology, The University of Melbourne, Parkville, 3010 VIC, Australia
| | - Jason Li
- The Sir Peter MacCallum Department of Oncology, University of Melbourne, Parkville, 3010 VIC, Australia; Bioinformatics Core, Peter MacCallum Cancer Centre, St. Andrews Place, East Melbourne, 3002 VIC, Australia
| | - Tobias Kratina
- Molecular Immunology Division, The Walter and Eliza Hall Institute of Medical Research, Parkville, 3052 VIC, Australia; Department of Medical Biology, The University of Melbourne, Parkville, 3010 VIC, Australia
| | - Eva Vidacs
- Cancer Therapeutics Program, Peter MacCallum Cancer Centre, St. Andrews Place, East Melbourne, 3002 VIC, Australia
| | - Omer Gilan
- Cancer Therapeutics Program, Peter MacCallum Cancer Centre, St. Andrews Place, East Melbourne, 3002 VIC, Australia; The Sir Peter MacCallum Department of Oncology, University of Melbourne, Parkville, 3010 VIC, Australia
| | - Maria A Doyle
- Research Computing Facility, Peter MacCallum Cancer Centre, St. Andrews Place, East Melbourne, 3002 VIC, Australia
| | - Andrea Newbold
- Cancer Therapeutics Program, Peter MacCallum Cancer Centre, St. Andrews Place, East Melbourne, 3002 VIC, Australia; The Sir Peter MacCallum Department of Oncology, University of Melbourne, Parkville, 3010 VIC, Australia
| | - Jessica E Bolden
- Department of Medical Biology, The University of Melbourne, Parkville, 3010 VIC, Australia; Molecular Medicine Division, The Walter and Eliza Hall Institute of Medical Research, 1G Royal Parade, Parkville, 3052 VIC, Australia
| | - Kirsten A Fairfax
- Department of Medical Biology, The University of Melbourne, Parkville, 3010 VIC, Australia; Molecular Medicine Division, The Walter and Eliza Hall Institute of Medical Research, 1G Royal Parade, Parkville, 3052 VIC, Australia
| | - Carolyn A de Graaf
- Department of Medical Biology, The University of Melbourne, Parkville, 3010 VIC, Australia; Molecular Medicine Division, The Walter and Eliza Hall Institute of Medical Research, 1G Royal Parade, Parkville, 3052 VIC, Australia
| | - Matthew Firth
- Molecular Immunology Division, The Walter and Eliza Hall Institute of Medical Research, Parkville, 3052 VIC, Australia; Department of Medical Biology, The University of Melbourne, Parkville, 3010 VIC, Australia
| | - Johannes Zuber
- Research Institute of Molecular Pathology (IMP), Vienna Biocenter (VBC), Dr. Bohr-Gasse 7, 1030 Vienna, Austria
| | - Ross A Dickins
- Australian Centre for Blood Diseases, Monash University, Melbourne, 3004 VIC, Australia
| | - Lynn M Corcoran
- Molecular Immunology Division, The Walter and Eliza Hall Institute of Medical Research, Parkville, 3052 VIC, Australia; Department of Medical Biology, The University of Melbourne, Parkville, 3010 VIC, Australia
| | - Mark A Dawson
- Cancer Therapeutics Program, Peter MacCallum Cancer Centre, St. Andrews Place, East Melbourne, 3002 VIC, Australia; The Sir Peter MacCallum Department of Oncology, University of Melbourne, Parkville, 3010 VIC, Australia
| | - Gabrielle T Belz
- Molecular Immunology Division, The Walter and Eliza Hall Institute of Medical Research, Parkville, 3052 VIC, Australia; Department of Medical Biology, The University of Melbourne, Parkville, 3010 VIC, Australia
| | - Ricky W Johnstone
- Cancer Therapeutics Program, Peter MacCallum Cancer Centre, St. Andrews Place, East Melbourne, 3002 VIC, Australia; The Sir Peter MacCallum Department of Oncology, University of Melbourne, Parkville, 3010 VIC, Australia.
| |
Collapse
|
56
|
Gloury R, Zotos D, Zuidscherwoude M, Masson F, Liao Y, Hasbold J, Corcoran LM, Hodgkin PD, Belz GT, Shi W, Nutt SL, Tarlinton DM, Kallies A. Dynamic changes in Id3 and E-protein activity orchestrate germinal center and plasma cell development. J Exp Med 2016; 213:1095-111. [PMID: 27217539 PMCID: PMC4886367 DOI: 10.1084/jem.20152003] [Citation(s) in RCA: 43] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2015] [Accepted: 04/15/2016] [Indexed: 12/27/2022] Open
Abstract
Gloury et al. reveal an essential role for the Id3–E-protein axis in the transcriptional regulation of humoral immunity. The generation of high-affinity antibodies requires germinal center (GC) development and differentiation of long-lived plasma cells in a multilayered process that is tightly controlled by the activity of multiple transcription factors. Here, we reveal a new layer of complexity by demonstrating that dynamic changes in Id3 and E-protein activity govern both GC and plasma cell differentiation. We show that down-regulation of Id3 in B cells is essential for releasing E2A and E2-2, which in a redundant manner are required for antigen-induced B cell differentiation. We demonstrate that this pathway controls the expression of multiple key factors, including Blimp1, Xbp1, and CXCR4, and is therefore critical for establishing the transcriptional network that controls GC B cell and plasma cell differentiation.
Collapse
Affiliation(s)
- Renee Gloury
- The Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria 3052, Australia Department of Medical Biology, University of Melbourne, Parkville, Victoria 3010, Australia
| | - Dimitra Zotos
- The Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria 3052, Australia Department of Medical Biology, University of Melbourne, Parkville, Victoria 3010, Australia
| | - Malou Zuidscherwoude
- The Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria 3052, Australia Department of Medical Biology, University of Melbourne, Parkville, Victoria 3010, Australia
| | - Frederick Masson
- The Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria 3052, Australia Department of Medical Biology, University of Melbourne, Parkville, Victoria 3010, Australia
| | - Yang Liao
- The Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria 3052, Australia Department of Medical Biology, University of Melbourne, Parkville, Victoria 3010, Australia
| | - Jhaguaral Hasbold
- The Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria 3052, Australia Department of Medical Biology, University of Melbourne, Parkville, Victoria 3010, Australia
| | - Lynn M Corcoran
- The Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria 3052, Australia Department of Medical Biology, University of Melbourne, Parkville, Victoria 3010, Australia
| | - Phil D Hodgkin
- The Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria 3052, Australia Department of Medical Biology, University of Melbourne, Parkville, Victoria 3010, Australia
| | - Gabrielle T Belz
- The Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria 3052, Australia Department of Medical Biology, University of Melbourne, Parkville, Victoria 3010, Australia
| | - Wei Shi
- The Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria 3052, Australia Department of Computing and Information Systems, University of Melbourne, Parkville, Victoria 3010, Australia
| | - Stephen L Nutt
- The Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria 3052, Australia Department of Medical Biology, University of Melbourne, Parkville, Victoria 3010, Australia
| | - David M Tarlinton
- The Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria 3052, Australia Department of Medical Biology, University of Melbourne, Parkville, Victoria 3010, Australia
| | - Axel Kallies
- The Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria 3052, Australia Department of Medical Biology, University of Melbourne, Parkville, Victoria 3010, Australia
| |
Collapse
|
57
|
Scott CL, Soen B, Martens L, Skrypek N, Saelens W, Taminau J, Blancke G, Van Isterdael G, Huylebroeck D, Haigh J, Saeys Y, Guilliams M, Lambrecht BN, Berx G. The transcription factor Zeb2 regulates development of conventional and plasmacytoid DCs by repressing Id2. J Exp Med 2016; 213:897-911. [PMID: 27185854 PMCID: PMC4886362 DOI: 10.1084/jem.20151715] [Citation(s) in RCA: 105] [Impact Index Per Article: 13.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2015] [Accepted: 04/15/2016] [Indexed: 12/21/2022] Open
Abstract
Lambrecht et al. show that the transcription factor Zeb2 regulates commitment toward both the pDC and cDC2 lineages by repressing Id2. Plasmacytoid dendritic cells (DCs [pDCs]) develop from pre-pDCs, whereas two lineages of conventional DCs (cDCs; cDC1s and cDC2s) develop from lineage-committed pre-cDCs. Several transcription factors (TFs) have been implicated in regulating the development of pDCs (E2-2 and Id2) and cDC1s (Irf8, Id2, and Batf3); however, those required for the early commitment of pre-cDCs toward the cDC2 lineage are unknown. Here, we identify the TF zinc finger E box–binding homeobox 2 (Zeb2) to play a crucial role in regulating DC development. Zeb2 was expressed from the pre-pDC and pre-cDC stage onward and highly expressed in mature pDCs and cDC2s. Mice conditionally lacking Zeb2 in CD11c+ cells had a cell-intrinsic reduction in pDCs and cDC2s, coupled with an increase in cDC1s. Conversely, mice in which CD11c+ cells overexpressed Zeb2 displayed a reduction in cDC1s. This was accompanied by altered expression of Id2, which was up-regulated in cDC2s and pDCs from conditional knockout mice. Zeb2 chromatin immunoprecipitation analysis revealed Id2 to be a direct target of Zeb2. Thus, we conclude that Zeb2 regulates commitment to both the cDC2 and pDC lineages through repression of Id2.
Collapse
Affiliation(s)
- Charlotte L Scott
- Laboratory of Immunoregulation and Mucosal Immunology, Inflammation Research Center, Vlaams Instituut voor Biotechnologie, 9052 Ghent, Belgium Department of Respiratory Medicine, Ghent University, 9000 Ghent, Belgium Department of Biomedical Molecular Biology, Ghent University, 9000 Ghent, Belgium
| | - Bieke Soen
- Unit of Molecular and Cellular Oncology, Inflammation Research Center, Vlaams Instituut voor Biotechnologie, 9052 Ghent, Belgium Department of Biomedical Molecular Biology, Ghent University, 9000 Ghent, Belgium
| | - Liesbet Martens
- Data Mining and Modeling for Biomedicine, Inflammation Research Center, Vlaams Instituut voor Biotechnologie, 9052 Ghent, Belgium Department of Respiratory Medicine, Ghent University, 9000 Ghent, Belgium
| | - Nicolas Skrypek
- Unit of Molecular and Cellular Oncology, Inflammation Research Center, Vlaams Instituut voor Biotechnologie, 9052 Ghent, Belgium Department of Biomedical Molecular Biology, Ghent University, 9000 Ghent, Belgium
| | - Wouter Saelens
- Data Mining and Modeling for Biomedicine, Inflammation Research Center, Vlaams Instituut voor Biotechnologie, 9052 Ghent, Belgium Department of Respiratory Medicine, Ghent University, 9000 Ghent, Belgium
| | - Joachim Taminau
- Unit of Molecular and Cellular Oncology, Inflammation Research Center, Vlaams Instituut voor Biotechnologie, 9052 Ghent, Belgium Department of Biomedical Molecular Biology, Ghent University, 9000 Ghent, Belgium
| | - Gillian Blancke
- Unit of Molecular and Cellular Oncology, Inflammation Research Center, Vlaams Instituut voor Biotechnologie, 9052 Ghent, Belgium Department of Biomedical Molecular Biology, Ghent University, 9000 Ghent, Belgium
| | - Gert Van Isterdael
- Laboratory of Immunoregulation and Mucosal Immunology, Inflammation Research Center, Vlaams Instituut voor Biotechnologie, 9052 Ghent, Belgium Department of Respiratory Medicine, Ghent University, 9000 Ghent, Belgium
| | - Danny Huylebroeck
- Laboratory of Molecular Biology (Celgen), Department of Development and Regeneration, Katholieke Universiteit Leuven, 3000 Leuven, Belgium Department of Cell Biology, Erasmus University Medical Center, 3015 GE Rotterdam, Netherlands
| | - Jody Haigh
- Mammalian Functional Genetics Laboratory, Division of Blood Cancers, Australian Centre for Blood Diseases, Monash University, Melbourne, Victoria 3800, Australia
| | - Yvan Saeys
- Data Mining and Modeling for Biomedicine, Inflammation Research Center, Vlaams Instituut voor Biotechnologie, 9052 Ghent, Belgium Department of Respiratory Medicine, Ghent University, 9000 Ghent, Belgium
| | - Martin Guilliams
- Laboratory of Immunoregulation and Mucosal Immunology, Inflammation Research Center, Vlaams Instituut voor Biotechnologie, 9052 Ghent, Belgium Department of Biomedical Molecular Biology, Ghent University, 9000 Ghent, Belgium
| | - Bart N Lambrecht
- Laboratory of Immunoregulation and Mucosal Immunology, Inflammation Research Center, Vlaams Instituut voor Biotechnologie, 9052 Ghent, Belgium Department of Respiratory Medicine, Ghent University, 9000 Ghent, Belgium Department of Pulmonary Medicine, Erasmus University Medical Center, 3015 GE Rotterdam, Netherlands
| | - Geert Berx
- Unit of Molecular and Cellular Oncology, Inflammation Research Center, Vlaams Instituut voor Biotechnologie, 9052 Ghent, Belgium Department of Biomedical Molecular Biology, Ghent University, 9000 Ghent, Belgium
| |
Collapse
|
58
|
Carpentier S, Vu Manh TP, Chelbi R, Henri S, Malissen B, Haniffa M, Ginhoux F, Dalod M. Comparative genomics analysis of mononuclear phagocyte subsets confirms homology between lymphoid tissue-resident and dermal XCR1(+) DCs in mouse and human and distinguishes them from Langerhans cells. J Immunol Methods 2016; 432:35-49. [PMID: 26966045 PMCID: PMC4859332 DOI: 10.1016/j.jim.2016.02.023] [Citation(s) in RCA: 42] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2015] [Revised: 02/01/2016] [Accepted: 02/25/2016] [Indexed: 12/24/2022]
Abstract
Dendritic cells (DC) are mononuclear phagocytes which exhibit a branching (dendritic) morphology and excel at naïve T cell activation. DC encompass several subsets initially identified by their expression of cell surface molecules and later shown to possess distinct functions. DC subset differentiation is orchestrated by transcription factors, growth factors and cytokines. Identifying DC subsets is challenging as very few cell surface molecules are uniquely expressed on any one of these cell populations. There is no standard consensus to identify mononuclear phagocyte subsets; varying antigens are employed depending on the tissue and animal species studied and between laboratories. This has led to confusion in how to accurately define and classify DCs across tissues and between species. Here we report a comparative genomics strategy that enables universal definition of DC and other mononuclear phagocyte subsets across species. We performed a meta-analysis of several public datasets of human and mouse mononuclear phagocyte subsets isolated from blood, spleen, skin or cutaneous lymph nodes, including by using a novel and user friendly software, BubbleGUM, which generates and integrates gene signatures for high throughput gene set enrichment analysis. This analysis demonstrates the equivalence between human and mouse skin XCR1(+) DCs, and between mouse and human Langerhans cells.
Collapse
Affiliation(s)
- Sabrina Carpentier
- Mi-mAbs (C/O Centre d'Immunologie de Marseille-Luminy), F-13009 Marseille, France
| | - Thien-Phong Vu Manh
- Centre d'Immunologie de Marseille-Luminy, Aix Marseille Université UM2, Inserm, U1104, CNRS UMR7280, F-13288 Marseille Cedex 09, France
| | - Rabie Chelbi
- Centre d'Immunologie de Marseille-Luminy, Aix Marseille Université UM2, Inserm, U1104, CNRS UMR7280, F-13288 Marseille Cedex 09, France
| | - Sandrine Henri
- Centre d'Immunologie de Marseille-Luminy, Aix Marseille Université UM2, Inserm, U1104, CNRS UMR7280, F-13288 Marseille Cedex 09, France
| | - Bernard Malissen
- Centre d'Immunologie de Marseille-Luminy, Aix Marseille Université UM2, Inserm, U1104, CNRS UMR7280, F-13288 Marseille Cedex 09, France
| | - Muzlifah Haniffa
- Human Dendritic Cell Laboratory, Institute of Cellular Medicine, Newcastle University, Newcastle upon Tyne, UK.
| | - Florent Ginhoux
- Singapore Immunology Network (SIgN), Agency for Science, Technology and Research (A*STAR), Singapore.
| | - Marc Dalod
- Centre d'Immunologie de Marseille-Luminy, Aix Marseille Université UM2, Inserm, U1104, CNRS UMR7280, F-13288 Marseille Cedex 09, France.
| |
Collapse
|
59
|
Cohen SB, Smith NL, McDougal C, Pepper M, Shah S, Yap GS, Acha-Orbea H, Jiang A, Clausen BE, Rudd BD, Denkers EY. Beta-catenin signaling drives differentiation and proinflammatory function of IRF8-dependent dendritic cells. THE JOURNAL OF IMMUNOLOGY 2016; 194:210-22. [PMID: 25416805 DOI: 10.4049/jimmunol.1402453] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Abstract
Beta-catenin signaling has recently been tied to the emergence of tolerogenic dendritic cells (DCs). In this article, we demonstrate a novel role for beta-catenin in directing DC subset development through IFN regulatory factor 8 (IRF8) activation. We found that splenic DC precursors express beta-catenin, and DCs from mice with CD11c-specific constitutive beta-catenin activation upregulated IRF8 through targeting of the Irf8 promoter, leading to in vivo expansion of IRF8-dependent CD8a+, plasmacytoid, and CD103+ CD11b2 DCs. beta-catenin–stabilized CD8a+ DCs secreted elevated IL-12 upon in vitro microbial stimulation, and pharmacological beta-catenin inhibition blocked this response in wild-type cells. Upon infections with Toxoplasma gondii and vaccinia virus, mice with stabilized DC beta-catenin displayed abnormally high Th1 and CD8+ T lymphocyte responses, respectively. Collectively, these results reveal a novel and unexpected function for beta-catenin in programming DC differentiation toward subsets that orchestrate proinflammatory immunity to infection.
Collapse
Affiliation(s)
- Sara B Cohen
- Department of Microbiology and Immunology, Cornell University College of Veterinary Medicine, Ithaca, NY 14867
| | - Norah L Smith
- Department of Microbiology and Immunology, Cornell University College of Veterinary Medicine, Ithaca, NY 14867
| | - Courtney McDougal
- Department of Microbiology and Immunology, Cornell University College of Veterinary Medicine, Ithaca, NY 14867
| | - Marion Pepper
- Department of Immunology, University of Washington School of Medicine, Seattle, WA 98101
| | - Suhagi Shah
- Center for Immunity and Inflammation, New Jersey Medical School, Rutgers, The State University of New Jersey, Newark, NJ 07101
| | - George S Yap
- Center for Immunity and Inflammation, New Jersey Medical School, Rutgers, The State University of New Jersey, Newark, NJ 07101
| | - Hans Acha-Orbea
- Department of Biochemistry, University of Lausanne, CH-1066 Epalinges, Switzerland
| | - Aimin Jiang
- Department of Immunology, Roswell Park Cancer Institute, Buffalo, NY 14263
| | - Bjorn E Clausen
- Institute for Molecular Medicine, University Medical Center of the Johannes Gutenberg-University Mainz, 55131 Mainz, Germany
| | - Brian D Rudd
- Department of Microbiology and Immunology, Cornell University College of Veterinary Medicine, Ithaca, NY 14867
| | - Eric Y Denkers
- Department of Microbiology and Immunology, Cornell University College of Veterinary Medicine, Ithaca, NY 14867
| |
Collapse
|
60
|
Molecular Mechanisms of Induction of Tolerant and Tolerogenic Intestinal Dendritic Cells in Mice. J Immunol Res 2016; 2016:1958650. [PMID: 26981546 PMCID: PMC4766351 DOI: 10.1155/2016/1958650] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2015] [Revised: 01/06/2016] [Accepted: 01/17/2016] [Indexed: 12/31/2022] Open
Abstract
How does the host manage to tolerate its own intestinal microbiota? A simple question leading to complicated answers. In order to maintain balanced immune responses in the intestine, the host immune system must tolerate commensal bacteria in the gut while it has to simultaneously keep the ability to fight pathogens and to clear infections. If this tender equilibrium is disturbed, severe chronic inflammatory reactions can result. Tolerogenic intestinal dendritic cells fulfil a crucial role in balancing immune responses and therefore creating homeostatic conditions and preventing from uncontrolled inflammation. Although several dendritic cell subsets have already been characterized to play a pivotal role in this process, less is known about definite molecular mechanisms of how intestinal dendritic cells are converted into tolerogenic ones. Here we review how gut commensal bacteria interact with intestinal dendritic cells and why this bacteria-host cell interaction is crucial for induction of dendritic cell tolerance in the intestine. Hereby, different commensal bacteria can have distinct effects on the phenotype of intestinal dendritic cells and these effects are mainly mediated by impacting toll-like receptor signalling in dendritic cells.
Collapse
|
61
|
Delconte RB, Shi W, Sathe P, Ushiki T, Seillet C, Minnich M, Kolesnik TB, Rankin LC, Mielke LA, Zhang JG, Busslinger M, Smyth MJ, Hutchinson DS, Nutt SL, Nicholson SE, Alexander WS, Corcoran LM, Vivier E, Belz GT, Carotta S, Huntington ND. The Helix-Loop-Helix Protein ID2 Governs NK Cell Fate by Tuning Their Sensitivity to Interleukin-15. Immunity 2016; 44:103-115. [PMID: 26795246 DOI: 10.1016/j.immuni.2015.12.007] [Citation(s) in RCA: 94] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2015] [Revised: 07/28/2015] [Accepted: 09/30/2015] [Indexed: 12/21/2022]
Abstract
The inhibitor of DNA binding 2 (Id2) is essential for natural killer (NK) cell development with its canonical role being to antagonize E-protein function and alternate lineage fate. Here we have identified a key role for Id2 in regulating interleukin-15 (IL-15) receptor signaling and homeostasis of NK cells by repressing multiple E-protein target genes including Socs3. Id2 deletion in mature NK cells was incompatible with their homeostasis due to impaired IL-15 receptor signaling and metabolic function and this could be rescued by strong IL-15 receptor stimulation or genetic ablation of Socs3. During NK cell maturation, we observed an inverse correlation between E-protein target genes and Id2. These results shift the current paradigm on the role of ID2, indicating that it is required not only to antagonize E-proteins during NK cell commitment, but constantly required to titrate E-protein activity to regulate NK cell fitness and responsiveness to IL-15.
Collapse
Affiliation(s)
- Rebecca B Delconte
- The Walter and Eliza Hall Institute of Medical Research, 1G Royal Parade, Parkville, VIC 3052, Australia; Department of Medical Biology, University of Melbourne, VIC 3010 Australia
| | - Wei Shi
- The Walter and Eliza Hall Institute of Medical Research, 1G Royal Parade, Parkville, VIC 3052, Australia; Department of Medical Biology, University of Melbourne, VIC 3010 Australia; Department of Computing and Information Systems, The University of Melbourne, VIC 3010, Australia
| | - Priyanka Sathe
- The Walter and Eliza Hall Institute of Medical Research, 1G Royal Parade, Parkville, VIC 3052, Australia; Department of Medical Biology, University of Melbourne, VIC 3010 Australia
| | - Takashi Ushiki
- The Walter and Eliza Hall Institute of Medical Research, 1G Royal Parade, Parkville, VIC 3052, Australia; Department of Medical Biology, University of Melbourne, VIC 3010 Australia
| | - Cyril Seillet
- The Walter and Eliza Hall Institute of Medical Research, 1G Royal Parade, Parkville, VIC 3052, Australia; Department of Medical Biology, University of Melbourne, VIC 3010 Australia
| | - Martina Minnich
- Research Institute of Molecular Pathology, Dr. Bohr-Gasse 7, 1030 Vienna, Austria
| | - Tatiana B Kolesnik
- The Walter and Eliza Hall Institute of Medical Research, 1G Royal Parade, Parkville, VIC 3052, Australia; Department of Medical Biology, University of Melbourne, VIC 3010 Australia
| | - Lucille C Rankin
- The Walter and Eliza Hall Institute of Medical Research, 1G Royal Parade, Parkville, VIC 3052, Australia; Department of Medical Biology, University of Melbourne, VIC 3010 Australia
| | - Lisa A Mielke
- The Walter and Eliza Hall Institute of Medical Research, 1G Royal Parade, Parkville, VIC 3052, Australia; Department of Medical Biology, University of Melbourne, VIC 3010 Australia
| | - Jian-Guo Zhang
- The Walter and Eliza Hall Institute of Medical Research, 1G Royal Parade, Parkville, VIC 3052, Australia; Department of Medical Biology, University of Melbourne, VIC 3010 Australia
| | - Meinrad Busslinger
- Research Institute of Molecular Pathology, Dr. Bohr-Gasse 7, 1030 Vienna, Austria
| | - Mark J Smyth
- QIMR Berghofer Medical Research Institute, Herston, QLD 4006, Australia; School of Medicine, University of Queensland, Herston, QLD 4006, Australia
| | - Dana S Hutchinson
- Drug Discovery Biology, Monash Institute of Pharmacological Science, Parkville, VIC 3052, Australia
| | - Stephen L Nutt
- The Walter and Eliza Hall Institute of Medical Research, 1G Royal Parade, Parkville, VIC 3052, Australia; Department of Medical Biology, University of Melbourne, VIC 3010 Australia
| | - Sandra E Nicholson
- The Walter and Eliza Hall Institute of Medical Research, 1G Royal Parade, Parkville, VIC 3052, Australia; Department of Medical Biology, University of Melbourne, VIC 3010 Australia
| | - Warren S Alexander
- The Walter and Eliza Hall Institute of Medical Research, 1G Royal Parade, Parkville, VIC 3052, Australia; Department of Medical Biology, University of Melbourne, VIC 3010 Australia
| | - Lynn M Corcoran
- The Walter and Eliza Hall Institute of Medical Research, 1G Royal Parade, Parkville, VIC 3052, Australia; Department of Medical Biology, University of Melbourne, VIC 3010 Australia
| | - Eric Vivier
- Centre d'Immunologie de Marseille-Luminy, Aix Marseille Université UM2, Inserm, U1104, CNRS UMR7280, 13288 Marseille, France; Immunologie, Hôpital de la Comception, Assistance Publique des Hôpitaux de Marseille, 13385 Marseille, France
| | - Gabrielle T Belz
- The Walter and Eliza Hall Institute of Medical Research, 1G Royal Parade, Parkville, VIC 3052, Australia.
| | - Sebastian Carotta
- The Walter and Eliza Hall Institute of Medical Research, 1G Royal Parade, Parkville, VIC 3052, Australia
| | - Nicholas D Huntington
- The Walter and Eliza Hall Institute of Medical Research, 1G Royal Parade, Parkville, VIC 3052, Australia.
| |
Collapse
|
62
|
Park JH, Choi AJ, Kim SJ, Jeong SY. 3,3'-Diindolylmethane Inhibits Flt3L/GM-CSF-induced-bone Marrow-derived CD103(+) Dendritic Cell Differentiation Regulating Phosphorylation of STAT3 and STAT5. Immune Netw 2015; 15:278-90. [PMID: 26770182 PMCID: PMC4700404 DOI: 10.4110/in.2015.15.6.278] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2015] [Revised: 11/05/2015] [Accepted: 11/07/2015] [Indexed: 01/05/2023] Open
Abstract
The intestinal immune system maintains oral tolerance to harmless antigens or nutrients. One mechanism of oral tolerance is mediated by regulatory T cell (Treg)s, of which differentiation is regulated by a subset of dendritic cell (DC)s, primarily CD103+ DCs. The aryl hydrocarbon receptor (AhR), a ligand-activated transcription factor, plays an important role in regulating immunity. The intestines are exposed to various AhR ligands, including endogenous metabolites and phytochemicals. It was previously reported that AhR activation induced tolerogenic DCs in mice or in cultures of bone marrow-derived DCs. However, given the variety of tolerogenic DCs, which type of tolerogenic DCs is regulated by AhR remains unknown. In this study, we found that AhR ligand 3,3'-diindolylmethane (DIM) inhibited the development of CD103+ DCs from mouse bone marrow cells stimulated with Flt3L and GM-CSF. DIM interfered with phosphorylation of STAT3 and STAT5 inhibiting the expression of genes, including Id2, E2-2, IDO-1, and Aldh1a2, which are associated with DC differentiation and functions. Finally, DIM suppressed the ability of CD103+ DCs to induce Foxp3+ Tregs.
Collapse
Affiliation(s)
- Joo-Hung Park
- Department of Biology, Changwon National University, Changwon 51140, Korea
| | - Ah-Jeong Choi
- Department of Biology, Changwon National University, Changwon 51140, Korea
| | - Soo-Ji Kim
- Department of Biology, Changwon National University, Changwon 51140, Korea
| | - So-Yeon Jeong
- Department of Biology, Changwon National University, Changwon 51140, Korea
| |
Collapse
|
63
|
Murphy TL, Grajales-Reyes GE, Wu X, Tussiwand R, Briseño CG, Iwata A, Kretzer NM, Durai V, Murphy KM. Transcriptional Control of Dendritic Cell Development. Annu Rev Immunol 2015; 34:93-119. [PMID: 26735697 DOI: 10.1146/annurev-immunol-032713-120204] [Citation(s) in RCA: 316] [Impact Index Per Article: 35.1] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
The dendritic cells (DCs) of the immune system function in innate and adaptive responses by directing activity of various effector cells rather than serving as effectors themselves. DCs and closely related myeloid lineages share expression of many surface receptors, presenting a challenge in distinguishing their unique in vivo functions. Recent work has taken advantage of unique transcriptional programs to identify and manipulate murine DCs in vivo. This work has assigned several nonredundant in vivo functions to distinct DC lineages, consisting of plasmacytoid DCs and several subsets of classical DCs that promote different immune effector modules in response to pathogens. In parallel, a correspondence between human and murine DC subsets has emerged, underlying structural similarities for the DC lineages between these species. Recent work has begun to unravel the transcriptional circuitry that controls the development and diversification of DCs from common progenitors in the bone marrow.
Collapse
Affiliation(s)
- Theresa L Murphy
- Department of Pathology and Immunology, Washington University School of Medicine in St. Louis, Missouri 63110;
| | - Gary E Grajales-Reyes
- Department of Pathology and Immunology, Washington University School of Medicine in St. Louis, Missouri 63110;
| | - Xiaodi Wu
- Department of Pathology and Immunology, Washington University School of Medicine in St. Louis, Missouri 63110;
| | - Roxane Tussiwand
- Department of Biomedicine, University of Basel, 4058 Basel, Switzerland
| | - Carlos G Briseño
- Department of Pathology and Immunology, Washington University School of Medicine in St. Louis, Missouri 63110;
| | - Arifumi Iwata
- Department of Pathology and Immunology, Washington University School of Medicine in St. Louis, Missouri 63110;
| | - Nicole M Kretzer
- Department of Pathology and Immunology, Washington University School of Medicine in St. Louis, Missouri 63110;
| | - Vivek Durai
- Department of Pathology and Immunology, Washington University School of Medicine in St. Louis, Missouri 63110;
| | - Kenneth M Murphy
- Department of Pathology and Immunology, Washington University School of Medicine in St. Louis, Missouri 63110; .,Howard Hughes Medical Institute, Washington University School of Medicine in St. Louis, Missouri 63110
| |
Collapse
|
64
|
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
|
65
|
Lin Q, Chauvistré H, Costa IG, Gusmao EG, Mitzka S, Hänzelmann S, Baying B, Klisch T, Moriggl R, Hennuy B, Smeets H, Hoffmann K, Benes V, Seré K, Zenke M. Epigenetic program and transcription factor circuitry of dendritic cell development. Nucleic Acids Res 2015; 43:9680-93. [PMID: 26476451 PMCID: PMC4787753 DOI: 10.1093/nar/gkv1056] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2015] [Accepted: 10/03/2015] [Indexed: 12/18/2022] Open
Abstract
Dendritic cells (DC) are professional antigen presenting cells that develop from hematopoietic stem cells through successive steps of lineage commitment and differentiation. Multipotent progenitors (MPP) are committed to DC restricted common DC progenitors (CDP), which differentiate into specific DC subsets, classical DC (cDC) and plasmacytoid DC (pDC). To determine epigenetic states and regulatory circuitries during DC differentiation, we measured consecutive changes of genome-wide gene expression, histone modification and transcription factor occupancy during the sequel MPP-CDP-cDC/pDC. Specific histone marks in CDP reveal a DC-primed epigenetic signature, which is maintained and reinforced during DC differentiation. Epigenetic marks and transcription factor PU.1 occupancy increasingly coincide upon DC differentiation. By integrating PU.1 occupancy and gene expression we devised a transcription factor regulatory circuitry for DC commitment and subset specification. The circuitry provides the transcription factor hierarchy that drives the sequel MPP-CDP-cDC/pDC, including Irf4, Irf8, Tcf4, Spib and Stat factors. The circuitry also includes feedback loops inferred for individual or multiple factors, which stabilize distinct stages of DC development and DC subsets. In summary, here we describe the basic regulatory circuitry of transcription factors that drives DC development.
Collapse
Affiliation(s)
- Qiong Lin
- Institute for Biomedical Engineering, Department of Cell Biology, RWTH Aachen University Medical School, 52074 Aachen, Germany Helmholtz Institute for Biomedical Engineering, RWTH Aachen University, 52074 Aachen, Germany
| | - Heike Chauvistré
- Institute for Biomedical Engineering, Department of Cell Biology, RWTH Aachen University Medical School, 52074 Aachen, Germany Helmholtz Institute for Biomedical Engineering, RWTH Aachen University, 52074 Aachen, Germany Department of Dermatology, University Hospital Essen, 45147 Essen, Germany
| | - Ivan G Costa
- IZKF Computational Biology Research Group, RWTH Aachen University Medical School, 52074 Aachen, Germany Aachen Institute for Advanced Study in Computational Engineering Science (AICES), RWTH Aachen University, 52062 Aachen, Germany
| | - Eduardo G Gusmao
- IZKF Computational Biology Research Group, RWTH Aachen University Medical School, 52074 Aachen, Germany
| | - Saskia Mitzka
- Institute for Biomedical Engineering, Department of Cell Biology, RWTH Aachen University Medical School, 52074 Aachen, Germany Helmholtz Institute for Biomedical Engineering, RWTH Aachen University, 52074 Aachen, Germany
| | - Sonja Hänzelmann
- IZKF Computational Biology Research Group, RWTH Aachen University Medical School, 52074 Aachen, Germany
| | - Bianka Baying
- Genomics Core Facilities GeneCore, European Molecular Biology Laboratory (EMBL), 69117 Heidelberg, Germany
| | - Theresa Klisch
- Institute for Biomedical Engineering, Department of Cell Biology, RWTH Aachen University Medical School, 52074 Aachen, Germany Helmholtz Institute for Biomedical Engineering, RWTH Aachen University, 52074 Aachen, Germany
| | - Richard Moriggl
- Ludwig Boltzmann Institute for Cancer Research, University of Veterinary Medicine, Medical University Vienna, 1090 Vienna, Austria
| | - Benoit Hennuy
- GIGA-Genomics, University of Liège, 4000 Liège, Belgium
| | - Hubert Smeets
- Department of Genetics and Cell Biology, Maastricht University Medical Center, 6200 MD Maastricht, The Netherlands Research Schools CARIM and GROW, Maastricht University Medical Center, 6200 MD Maastricht, The Netherlands
| | - Kurt Hoffmann
- Institute of Molecular Biotechnology, RWTH Aachen University, 52074 Aachen, Germany
| | - Vladimir Benes
- Genomics Core Facilities GeneCore, European Molecular Biology Laboratory (EMBL), 69117 Heidelberg, Germany
| | - Kristin Seré
- Institute for Biomedical Engineering, Department of Cell Biology, RWTH Aachen University Medical School, 52074 Aachen, Germany Helmholtz Institute for Biomedical Engineering, RWTH Aachen University, 52074 Aachen, Germany
| | - Martin Zenke
- Institute for Biomedical Engineering, Department of Cell Biology, RWTH Aachen University Medical School, 52074 Aachen, Germany Helmholtz Institute for Biomedical Engineering, RWTH Aachen University, 52074 Aachen, Germany
| |
Collapse
|
66
|
Seillet C, Belz GT. Differentiation and diversity of subsets in group 1 innate lymphoid cells. Int Immunol 2015; 28:3-11. [PMID: 26346810 DOI: 10.1093/intimm/dxv051] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2015] [Accepted: 08/24/2015] [Indexed: 02/04/2023] Open
Abstract
NK cells were first identified in 1975 and represent the prototypical group 1 innate lymphoid cell (ILC). More recently, the discovery of new members of the ILC family has highlighted the complexity of this innate lymphoid lineage. Importantly, it has been recognized that different subsets exist within the group 1 ILC, which have potential roles in mediating immune protection and immunosurveillance, and in regulating tissue homeostasis and inflammation. Here, we review the developmental relationships between the different group 1 ILC, which have been identified to date and discuss how heterogeneity within this expanding family may have arisen.
Collapse
Affiliation(s)
- Cyril Seillet
- Walter and Eliza Hall Institute of Medical Research, 1G Royal Parade, Parkville, Melbourne, Victoria 3052, Australia Department of Medical Biology, University of Melbourne, Melbourne, Victoria 3010, Australia
| | - Gabrielle T Belz
- Walter and Eliza Hall Institute of Medical Research, 1G Royal Parade, Parkville, Melbourne, Victoria 3052, Australia Department of Medical Biology, University of Melbourne, Melbourne, Victoria 3010, Australia
| |
Collapse
|
67
|
Cohen SB, Denkers EY. Impact of Toxoplasma gondii on Dendritic Cell Subset Function in the Intestinal Mucosa. THE JOURNAL OF IMMUNOLOGY 2015; 195:2754-62. [PMID: 26283477 DOI: 10.4049/jimmunol.1501137] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/18/2015] [Accepted: 07/20/2015] [Indexed: 01/10/2023]
Abstract
The function of mucosal dendritic cell (DC) subsets in immunity and inflammation is not well understood. In this study, we define four DC subsets present within the lamina propria and mesenteric lymph node compartments based on expression of CD103 and CD11b. Using IL-12p40 YFP (Yet40) reporter mice, we show that CD103(+)CD11b(-) mucosal DCs are primary in vivo sources of IL-12p40; we also identified CD103(-)CD11b(-) mucosal DCs as a novel population producing this cytokine. Infection was preferentially found in CD11b(+) DCs that were negative for CD103. Lamina propria DCs containing parasites were negative for IL-12p40. Instead, production of the cytokine was strictly a property of noninfected cells. We also show that vitamin A metabolism, as measured by ALDH activity, was preferentially found in CD103(+)CD11b(+) DC and was strongly downregulated in all mucosal DC subsets during infection. Finally, overall apoptosis of lamina propria DC subsets was increased during infection. Combined, these results highlight the ability of intestinal Toxoplasma infection to alter mucosal DC activity at both the whole population level and at the level of individual subsets.
Collapse
Affiliation(s)
- Sara B Cohen
- Department of Microbiology and Immunology, College of Veterinary Medicine, Cornell University, Ithaca, NY 14853
| | - Eric Y Denkers
- Department of Microbiology and Immunology, College of Veterinary Medicine, Cornell University, Ithaca, NY 14853
| |
Collapse
|
68
|
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
|
69
|
Granja AG, Leal E, Pignatelli J, Castro R, Abós B, Kato G, Fischer U, Tafalla C. Identification of Teleost Skin CD8α+ Dendritic-like Cells, Representing a Potential Common Ancestor for Mammalian Cross-Presenting Dendritic Cells. THE JOURNAL OF IMMUNOLOGY 2015; 195:1825-37. [PMID: 26179908 DOI: 10.4049/jimmunol.1500322] [Citation(s) in RCA: 64] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/10/2015] [Accepted: 06/17/2015] [Indexed: 01/09/2023]
Abstract
Although fish constitute the most ancient animal group in which an acquired immune system is present, the presence of dendritic cells (DCs) in teleosts has been addressed only briefly, and the identification of a specific DC subset in teleosts remained elusive because of the lack of specific Abs. In mice, DCs expressing CD8α(+) in lymphoid tissues have the capacity to cross-present extracellular Ags to T cells through MHC I, similarly to tissue-derived CD103(+) DCs and the human CD141(+) DC population. In the current study, we identified a large and highly complex subpopulation of leukocytes coexpressing MHC class II and CD8α. This CD8α(+) MHC II(+) DC-like subpopulation constituted ∼1.2% of the total leukocyte population in the skin, showing phenotypical and functional characteristics of semimature DCs that seem to locally regulate mucosal immunity and tolerance in a species lacking lymph nodes. Furthermore, we identified trout homologs for CD141 and CD103 and demonstrated that, in trout, this skin CD8(+) DC-like subpopulation expresses both markers. To our knowledge, these results provide the first evidence of a specific DC-like subtype in nonimmune tissue in teleosts and support the hypothesis of a common origin for all mammalian cross-presenting DCs.
Collapse
Affiliation(s)
- Aitor G Granja
- Centro de Investigación en Sanidad Animal, Valdeolmos, 28130 Madrid, Spain; and
| | - Esther Leal
- Centro de Investigación en Sanidad Animal, Valdeolmos, 28130 Madrid, Spain; and
| | - Jaime Pignatelli
- Centro de Investigación en Sanidad Animal, Valdeolmos, 28130 Madrid, Spain; and
| | - Rosario Castro
- Centro de Investigación en Sanidad Animal, Valdeolmos, 28130 Madrid, Spain; and
| | - Beatriz Abós
- Centro de Investigación en Sanidad Animal, Valdeolmos, 28130 Madrid, Spain; and
| | - Goshi Kato
- Friedrich-Loeffler-Institute, Federal Research Institute for Animal Health, 17493 Greifswald-Insel Riems, Germany
| | - Uwe Fischer
- Friedrich-Loeffler-Institute, Federal Research Institute for Animal Health, 17493 Greifswald-Insel Riems, Germany
| | - Carolina Tafalla
- Centro de Investigación en Sanidad Animal, Valdeolmos, 28130 Madrid, Spain; and
| |
Collapse
|
70
|
Walker JA, Oliphant CJ, Englezakis A, Yu Y, Clare S, Rodewald HR, Belz G, Liu P, Fallon PG, McKenzie ANJ. Bcl11b is essential for group 2 innate lymphoid cell development. ACTA ACUST UNITED AC 2015; 212:875-82. [PMID: 25964370 PMCID: PMC4451131 DOI: 10.1084/jem.20142224] [Citation(s) in RCA: 122] [Impact Index Per Article: 13.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2014] [Accepted: 04/14/2015] [Indexed: 11/23/2022]
Abstract
Walker et al. demonstrate that the transcription factor Bcl11b is expressed in mouse lymphoid ILC2 precursors in the bone marrow and is required for their development. Mice deficient in Bcl11b exhibit a lack of ILC2 development and an expansion of RORγt+ ILC3s and are unable to clear Nippostrongylus brasiliensis worm infection, but can clear Citrobacter rodentium. Group 2 innate lymphoid cells (ILC2s) are often found associated with mucosal surfaces where they contribute to protective immunity, inappropriate allergic responses, and tissue repair. Although we know they develop from a common lymphoid progenitor in the bone marrow (BM), the specific lineage path and transcriptional regulators that are involved are only starting to emerge. After ILC2 gene expression analysis we investigated the role of Bcl11b, a factor previously linked to T cell commitment, in ILC2 development. Using combined Bcl11b-tom and Id2-gfp reporter mice, we show that Bcl11b is expressed in ILC2 precursors in the BM and maintained in mature ILC2s. In vivo deletion of Bcl11b, by conditional tamoxifen-induced depletion or by Bcl11b−/− fetal liver chimera reconstitution, demonstrates that ILC2s are wholly dependent on Bcl11b for their development. Notably, in the absence of Bcl11b there is a concomitant expansion of the RORγt+ ILC3 population, suggesting that Bcl11b may negatively regulate this lineage. Using Nippostrongylus brasiliensis infection, we reveal that the absence of Bcl11b leads to impaired worm expulsion, caused by a deficit in ILC2s, whereas Citrobacter rodentium infection is cleared efficiently. These data clearly establish Bcl11b as a new factor in the differentiation of ILC2s.
Collapse
Affiliation(s)
- Jennifer A Walker
- Medical Research Council (MRC) Laboratory of Molecular Biology, Cambridge CB2 0QH, England, UK
| | - Christopher J Oliphant
- Medical Research Council (MRC) Laboratory of Molecular Biology, Cambridge CB2 0QH, England, UK
| | - Alexandros Englezakis
- Medical Research Council (MRC) Laboratory of Molecular Biology, Cambridge CB2 0QH, England, UK
| | - Yong Yu
- Wellcome Trust Sanger Institute, Hinxton, Cambridge CB10 1HH, England, UK
| | - Simon Clare
- Wellcome Trust Sanger Institute, Hinxton, Cambridge CB10 1HH, England, UK
| | - Hans-Reimer Rodewald
- Division of Cellular Immunology, German Cancer Research Center, 69120 Heidelberg, Germany
| | - Gabrielle Belz
- The Walter and Eliza Hall Institute of Medical Research, Parkville, Melbourne, Victoria 3052, Australia Department of Medical Biology, University of Melbourne, Melbourne, Victoria 3010, Australia
| | - Pentao Liu
- Wellcome Trust Sanger Institute, Hinxton, Cambridge CB10 1HH, England, UK
| | - Padraic G Fallon
- Trinity Biomedical Sciences Institute and Institute of Molecular Medicine, Trinity College Dublin, Dublin 2, Ireland Trinity Biomedical Sciences Institute and Institute of Molecular Medicine, Trinity College Dublin, Dublin 2, Ireland National Children's Research Centre, Our Lady's Children's Hospital, Crumlin, Dublin 12, Ireland
| | - Andrew N J McKenzie
- Medical Research Council (MRC) Laboratory of Molecular Biology, Cambridge CB2 0QH, England, UK
| |
Collapse
|
71
|
Regulation of myelopoiesis by the transcription factor IRF8. Int J Hematol 2015; 101:342-51. [DOI: 10.1007/s12185-015-1761-9] [Citation(s) in RCA: 63] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2015] [Revised: 02/23/2015] [Accepted: 02/24/2015] [Indexed: 10/23/2022]
|
72
|
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
|
73
|
Lim AWY, McKenzie ANJ. Deciphering the transcriptional switches of innate lymphoid cell programming: the right factors at the right time. Genes Immun 2015; 16:177-86. [PMID: 25611557 PMCID: PMC4409422 DOI: 10.1038/gene.2014.83] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2014] [Revised: 12/17/2014] [Accepted: 12/19/2014] [Indexed: 12/17/2022]
Abstract
Innate lymphoid cells (ILCs) are increasingly recognised as an innate immune counterpart of adaptive TH cells. In addition to their similar effector cytokine production, there is a strong parallel between the transcription factors that control the differentiation of TH1, TH2 and TH17 cells and ILC Groups 1, 2 and 3, respectively. Here, we review the transcriptional circuit that specifies the development of a common ILC progenitor and its subsequent programming into distinct ILC groups. Notch, GATA-3, Nfil3 and Id2 are identified as early factors that suppress B and T cell potentials and are turned on in favour of ILC commitment. Natural killer cells, which are the cytotoxic ILCs, develop along a pathway distinct from the rest of the helper-like ILCs that are derived from a common progenitor to all helper-like innate lymphoid cells (CHILPs). PLZF− CHILPs give rise to lymphoid tissue inducer cells while PLZF+ CHILPs have multi-lineage potential and could give rise to ILCs 1, 2 and 3. Such lineage specificity is dictated by the controlled expression of T-bet, RORα, RORγt and AHR. In addition to the type of transcription factors, the developmental stages at which these factors are expressed are crucial in specifying the fate of the ILCs.
Collapse
Affiliation(s)
- A W Y Lim
- Division of Protein and Nucleic Acid Chemistry, MRC Laboratory of Molecular Biology, Cambridge, UK
| | - A N J McKenzie
- Division of Protein and Nucleic Acid Chemistry, MRC Laboratory of Molecular Biology, Cambridge, UK
| |
Collapse
|
74
|
Abstract
In this issue of Blood, Mayer et al describe a new method of generating high numbers of Batf3- and Irf8-dependent CD103+ conventional dendritic cells (cDCs), providing new opportunities to study this subset of antigen-presenting cells specialized in crosspresentation.
Collapse
|
75
|
|
76
|
Mott KR, Maazi H, Allen SJ, Zandian M, Matundan H, Ghiasi YN, Sharifi BG, Underhill D, Akbari O, Ghiasi H. Batf3 deficiency is not critical for the generation of CD8α⁺ dendritic cells. Immunobiology 2014; 220:518-24. [PMID: 25468565 DOI: 10.1016/j.imbio.2014.10.019] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2014] [Revised: 09/17/2014] [Accepted: 10/21/2014] [Indexed: 12/14/2022]
Abstract
Recently, we have reported that CD8α(+) DCs, rather than CD8(+) T cells, are involved in the establishment and maintenance of HSV-1 latency in the trigeminal ganglia (TG) of ocularly infected mice. In the current study, we investigated whether similar results can be obtained using Batf3(-/-) mice that previously were reported to lack CD8α(+) DCs. However, our results demonstrate that Batf3(-/-) mice, without any known infection, express CD8α(+) DCs. Consequently, due to the presence of CD8α(+) DCs, no differences were detected in the level of HSV-1 latency between Batf3(-/-) mice compared with wild type control mice.
Collapse
Affiliation(s)
- Kevin R Mott
- Center for Neurobiology and Vaccine Development, Ophthalmology Research, Department of Surgery, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Hadi Maazi
- Department of Molecular Microbiology and Immunology, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - Sariah J Allen
- Center for Neurobiology and Vaccine Development, Ophthalmology Research, Department of Surgery, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Mandana Zandian
- Center for Neurobiology and Vaccine Development, Ophthalmology Research, Department of Surgery, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Harry Matundan
- Center for Neurobiology and Vaccine Development, Ophthalmology Research, Department of Surgery, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Yasamin N Ghiasi
- Department of Molecular Microbiology and Immunology, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - Behrooz G Sharifi
- Division of Cardiology, Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - David Underhill
- Inflammatory Bowel & Immunobiology Research Institute, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Omid Akbari
- Department of Molecular Microbiology and Immunology, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - Homayon Ghiasi
- Center for Neurobiology and Vaccine Development, Ophthalmology Research, Department of Surgery, Cedars-Sinai Medical Center, Los Angeles, CA, USA.
| |
Collapse
|
77
|
Holmes ML, Huntington ND, Thong RPL, Brady J, Hayakawa Y, Andoniou CE, Fleming P, Shi W, Smyth GK, Degli-Esposti MA, Belz GT, Kallies A, Carotta S, Smyth MJ, Nutt SL. Peripheral natural killer cell maturation depends on the transcription factor Aiolos. EMBO J 2014; 33:2721-34. [PMID: 25319415 DOI: 10.15252/embj.201487900] [Citation(s) in RCA: 62] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022] Open
Abstract
Natural killer (NK) cells are an innate lymphoid cell lineage characterized by their capacity to provide rapid effector functions, including cytokine production and cytotoxicity. Here, we identify the Ikaros family member, Aiolos, as a regulator of NK-cell maturation. Aiolos expression is initiated at the point of lineage commitment and maintained throughout NK-cell ontogeny. Analysis of cell surface markers representative of distinct stages of peripheral NK-cell maturation revealed that Aiolos was required for the maturation in the spleen of CD11b(high)CD27(-) NK cells. The differentiation block was intrinsic to the NK-cell lineage and resembled that found in mice lacking either T-bet or Blimp1; however, genetic analysis revealed that Aiolos acted independently of all other known regulators of NK-cell differentiation. NK cells lacking Aiolos were strongly hyper-reactive to a variety of NK-cell-mediated tumor models, yet impaired in controlling viral infection, suggesting a regulatory function for CD27(-) NK cells in balancing these two arms of the immune response. These data place Aiolos in the emerging gene regulatory network controlling NK-cell maturation and function.
Collapse
Affiliation(s)
- Melissa L Holmes
- The Walter and Eliza Hall Institute of Medical Research, Parkville, Vic., Australia
| | - Nicholas D Huntington
- The Walter and Eliza Hall Institute of Medical Research, Parkville, Vic., Australia Department of Medical Biology, The University of Melbourne, Parkville, Vic., Australia
| | - Rebecca P L Thong
- The Walter and Eliza Hall Institute of Medical Research, Parkville, Vic., Australia
| | - Jason Brady
- Cancer Immunology Program, The Peter MacCallum Cancer Centre, East Melbourne, Vic., Australia
| | - Yoshihiro Hayakawa
- Division of Pathogenic Biochemistry, Institute of Natural Medicine, University of Toyama, Toyama, Japan
| | - Christopher E Andoniou
- Immunology and Virology Program, Centre for Ophthalmology and Visual Science, The University of Western Australia, Nedlands, WA, Australia Centre for Experimental Immunology, Lions Eye Institute, Nedlands, WA, Australia
| | - Peter Fleming
- Immunology and Virology Program, Centre for Ophthalmology and Visual Science, The University of Western Australia, Nedlands, WA, Australia Centre for Experimental Immunology, Lions Eye Institute, Nedlands, WA, Australia
| | - Wei Shi
- The Walter and Eliza Hall Institute of Medical Research, Parkville, Vic., Australia Department of Computing and Information Systems, University of Melbourne, Parkville, Vic., Australia
| | - Gordon K Smyth
- The Walter and Eliza Hall Institute of Medical Research, Parkville, Vic., Australia The Department of Mathematics and Statistics, University of Melbourne, Parkville, Vic., Australia
| | - Mariapia A Degli-Esposti
- Immunology and Virology Program, Centre for Ophthalmology and Visual Science, The University of Western Australia, Nedlands, WA, Australia Centre for Experimental Immunology, Lions Eye Institute, Nedlands, WA, Australia
| | - Gabrielle T Belz
- The Walter and Eliza Hall Institute of Medical Research, Parkville, Vic., Australia Department of Medical Biology, The University of Melbourne, Parkville, Vic., Australia
| | - Axel Kallies
- The Walter and Eliza Hall Institute of Medical Research, Parkville, Vic., Australia Department of Medical Biology, The University of Melbourne, Parkville, Vic., Australia
| | - Sebastian Carotta
- The Walter and Eliza Hall Institute of Medical Research, Parkville, Vic., Australia Department of Medical Biology, The University of Melbourne, Parkville, Vic., Australia
| | - Mark J Smyth
- QIMR Berghofer Medical Research Institute, Herston, Qld, Australia School of Medicine, University of Queensland, Herston, Qld, Australia
| | - Stephen L Nutt
- The Walter and Eliza Hall Institute of Medical Research, Parkville, Vic., Australia Department of Medical Biology, The University of Melbourne, Parkville, Vic., Australia
| |
Collapse
|
78
|
Petersen TR, Knight DA, Tang CW, Osmond TL, Hermans IF. Batf3-independent langerin- CX3CR1- CD8α+ splenic DCs represent a precursor for classical cross-presenting CD8α+ DCs. J Leukoc Biol 2014; 96:1001-10. [PMID: 25170118 DOI: 10.1189/jlb.1a0314-130r] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
This study tests the hypothesis that CD8α(+) DCs in the spleen of mice contain an immature precursor for functionally mature, "classical" cross-presenting CD8α(+) DCs. The lymphoid tissues contain a network of phenotypically distinct DCs with unique roles in surveillance and immunity. Splenic CD8α(+) DCs have been shown to exhibit a heightened capacity for phagocytosis of cellular material, secretion of IL-12, and cross-priming of CD8(+) T cells. However, this population can be subdivided further on the basis of expression of both langerin/CD207 and CX(3)CR1. We therefore evaluated the functional capacities of these different subsets. The CX(3)CR1(+) CD8α(+) DC subset does not express langerin and does not exhibit the classical features above. The CX(3)CR1(-) CD8α(+) DC can be divided into langerin-positive and negative populations, both of which express DEC205, Clec9A, and high basal levels of CD86. However, the langerin(+) CX(3)CR1(-) CD8α(+) subset has a superior capacity for acquiring cellular material and producing IL-12 and is more susceptible to activation-induced cell death. Significantly, following purification and adoptive transfer into new hosts, the langerin(-) CX(3)CR1(-) CD8α(+) subset survives longer, up-regulates expression of langerin, and becomes more susceptible to activation-induced cell death. Last, in contrast to langerin(+) CX(3)CR1(-) CD8α(+), the langerin(-) CX(3)CR1(-) CD8α(+) are still present in Batf3(-/-) mice. We conclude that the classical attributes of CD8α(+) DC are confined primarily to the langerin(+) CX(3)CR1(-) CD8α(+) DC population and that the langerin(-) CX(3)CR1(-) subset represents a Batf3-independent precursor to this mature population.
Collapse
Affiliation(s)
- Troels R Petersen
- Malaghan Institute of Medical Research, Wellington, New Zealand; and
| | - Deborah A Knight
- Malaghan Institute of Medical Research, Wellington, New Zealand; and
| | - Ching-Wen Tang
- Malaghan Institute of Medical Research, Wellington, New Zealand; and
| | - Taryn L Osmond
- Malaghan Institute of Medical Research, Wellington, New Zealand; and School of Biological Sciences, Victoria University of Wellington, New Zealand
| | - Ian F Hermans
- Malaghan Institute of Medical Research, Wellington, New Zealand; and School of Biological Sciences, Victoria University of Wellington, New Zealand
| |
Collapse
|
79
|
Selective and efficient generation of functional Batf3-dependent CD103+ dendritic cells from mouse bone marrow. Blood 2014; 124:3081-91. [PMID: 25100743 DOI: 10.1182/blood-2013-12-545772] [Citation(s) in RCA: 147] [Impact Index Per Article: 14.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
Multiple subsets of FMS-like tyrosine kinase 3 ligand (FLT3L)-dependent dendritic cells (DCs) control T-cell tolerance and immunity. In mice, Batf3-dependent CD103(+) DCs efficiently enter lymph nodes and cross-present antigens, rendering this conserved DC subset a promising target for tolerance induction or vaccination. However, only limited numbers of CD103(+) DCs can be isolated with current methods. Established bone marrow culture protocols efficiently generate monocyte-derived DCs or produce a mixture of FLT3L-dependent DC subsets. We show that CD103(+) DC development requires prolonged culture time and continuous action of both FLT3L and granulocyte macrophage colony-stimulating factor (GM-CSF), explained by a dual effect of GM-CSF on DC precursors and differentiating CD103(+) DCs. Accordingly, we established a novel method to generate large numbers of CD103(+) DCs (iCD103-DCs) with limited presence of other DC subsets. iCD103-DCs develop in a Batf3- and Irf8-dependent fashion, express a CD8α/CD103 DC gene signature, cross-present cell-associated antigens, and respond to TLR3 stimulation. Thus, iCD103-DCs reflect key features of tissue CD103(+) DCs. Importantly, iCD103-DCs express high levels of CCR7 upon maturation and migrate to lymph nodes more efficiently than classical monocyte-derived DCs. Finally, iCD103-DCs induce T cell-mediated protective immunity in vivo. Our study provides insights into CD103(+) DC development and function.
Collapse
|
80
|
Dendritic cells, monocytes and macrophages: a unified nomenclature based on ontogeny. Nat Rev Immunol 2014; 14:571-8. [PMID: 25033907 DOI: 10.1038/nri3712] [Citation(s) in RCA: 1311] [Impact Index Per Article: 131.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
The mononuclear phagocyte system (MPS) has historically been categorized into monocytes, dendritic cells and macrophages on the basis of functional and phenotypical characteristics. However, considering that these characteristics are often overlapping, the distinction between and classification of these cell types has been challenging. In this Opinion article, we propose a unified nomenclature for the MPS. We suggest that these cells can be classified primarily by their ontogeny and secondarily by their location, function and phenotype. We believe that this system permits a more robust classification during both steady-state and inflammatory conditions, with the benefit of spanning different tissues and across species.
Collapse
|
81
|
Ghosh HS, Ceribelli M, Matos I, Lazarovici A, Bussemaker HJ, Lasorella A, Hiebert SW, Liu K, Staudt LM, Reizis B. ETO family protein Mtg16 regulates the balance of dendritic cell subsets by repressing Id2. ACTA ACUST UNITED AC 2014; 211:1623-35. [PMID: 24980046 PMCID: PMC4113936 DOI: 10.1084/jem.20132121] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Dendritic cells (DCs) comprise two major subsets, the interferon (IFN)-producing plasmacytoid DCs (pDCs) and antigen-presenting classical DCs (cDCs). The development of pDCs is promoted by E protein transcription factor E2-2, whereas E protein antagonist Id2 is specifically absent from pDCs. Conversely, Id2 is prominently expressed in cDCs and promotes CD8(+) cDC development. The mechanisms that control the balance between E and Id proteins during DC subset specification remain unknown. We found that the loss of Mtg16, a transcriptional cofactor of the ETO protein family, profoundly impaired pDC development and pDC-dependent IFN response. The residual Mtg16-deficient pDCs showed aberrant phenotype, including the expression of myeloid marker CD11b. Conversely, the development of cDC progenitors (pre-DCs) and of CD8(+) cDCs was enhanced. Genome-wide expression and DNA-binding analysis identified Id2 as a direct target of Mtg16. Mtg16-deficient cDC progenitors and pDCs showed aberrant induction of Id2, and the deletion of Id2 facilitated the impaired development of Mtg16-deficient pDCs. Thus, Mtg16 promotes pDC differentiation and restricts cDC development in part by repressing Id2, revealing a cell-intrinsic mechanism that controls subset balance during DC development.
Collapse
Affiliation(s)
- Hiyaa S Ghosh
- Department of Microbiology and Immunology, Center for Computational Biology and Bioinformatics, Institute for Cancer Genetics, Department of Pathology, and Department of Pediatrics, Columbia University Medical Center and Department of Biological Sciences and Department of Electrical Engineering, Columbia University, New York, NY 10032
| | - Michele Ceribelli
- Lymphoid Malignancies Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892
| | - Ines Matos
- Department of Microbiology and Immunology, Center for Computational Biology and Bioinformatics, Institute for Cancer Genetics, Department of Pathology, and Department of Pediatrics, Columbia University Medical Center and Department of Biological Sciences and Department of Electrical Engineering, Columbia University, New York, NY 10032
| | - Allan Lazarovici
- Department of Microbiology and Immunology, Center for Computational Biology and Bioinformatics, Institute for Cancer Genetics, Department of Pathology, and Department of Pediatrics, Columbia University Medical Center and Department of Biological Sciences and Department of Electrical Engineering, Columbia University, New York, NY 10032Department of Microbiology and Immunology, Center for Computational Biology and Bioinformatics, Institute for Cancer Genetics, Department of Pathology, and Department of Pediatrics, Columbia University Medical Center and Department of Biological Sciences and Department of Electrical Engineering, Columbia University, New York, NY 10032
| | - Harmen J Bussemaker
- Department of Microbiology and Immunology, Center for Computational Biology and Bioinformatics, Institute for Cancer Genetics, Department of Pathology, and Department of Pediatrics, Columbia University Medical Center and Department of Biological Sciences and Department of Electrical Engineering, Columbia University, New York, NY 10032Department of Microbiology and Immunology, Center for Computational Biology and Bioinformatics, Institute for Cancer Genetics, Department of Pathology, and Department of Pediatrics, Columbia University Medical Center and Department of Biological Sciences and Department of Electrical Engineering, Columbia University, New York, NY 10032
| | - Anna Lasorella
- Department of Microbiology and Immunology, Center for Computational Biology and Bioinformatics, Institute for Cancer Genetics, Department of Pathology, and Department of Pediatrics, Columbia University Medical Center and Department of Biological Sciences and Department of Electrical Engineering, Columbia University, New York, NY 10032Department of Microbiology and Immunology, Center for Computational Biology and Bioinformatics, Institute for Cancer Genetics, Department of Pathology, and Department of Pediatrics, Columbia University Medical Center and Department of Biological Sciences and Department of Electrical Engineering, Columbia University, New York, NY 10032Department of Microbiology and Immunology, Center for Computational Biology and Bioinformatics, Institute for Cancer Genetics, Department of Pathology, and Department of Pediatrics, Columbia University Medical Center and Department of Biological Sciences and Department of Electrical Engineering, Columbia University, New York, NY 10032
| | - Scott W Hiebert
- Department of Biochemistry, Vanderbilt University School of Medicine, Nashville, TN 37232
| | - Kang Liu
- Department of Microbiology and Immunology, Center for Computational Biology and Bioinformatics, Institute for Cancer Genetics, Department of Pathology, and Department of Pediatrics, Columbia University Medical Center and Department of Biological Sciences and Department of Electrical Engineering, Columbia University, New York, NY 10032
| | - Louis M Staudt
- Lymphoid Malignancies Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892
| | - Boris Reizis
- Department of Microbiology and Immunology, Center for Computational Biology and Bioinformatics, Institute for Cancer Genetics, Department of Pathology, and Department of Pediatrics, Columbia University Medical Center and Department of Biological Sciences and Department of Electrical Engineering, Columbia University, New York, NY 10032
| |
Collapse
|
82
|
The closely related CD103+ dendritic cells (DCs) and lymphoid-resident CD8+ DCs differ in their inflammatory functions. PLoS One 2014; 9:e91126. [PMID: 24637385 PMCID: PMC3956455 DOI: 10.1371/journal.pone.0091126] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2013] [Accepted: 02/07/2014] [Indexed: 01/01/2023] Open
Abstract
Migratory CD103+ and lymphoid-resident CD8+ dendritic cells (DCs) share many attributes, such as dependence on the same transcription factors, cross-presenting ability and expression of certain surface molecules, such that it has been proposed they belong to a common sub-lineage. The functional diversity of the two DC types is nevertheless incompletely understood. Here we reveal that upon skin infection with herpes simplex virus, migratory CD103+ DCs from draining lymph nodes were more potent at inducing Th17 cytokine production by CD4+ T cells than CD8+ DCs. This superior capacity to drive Th17 responses was also evident in CD103+ DCs from uninfected mice. Their differential potency to induce Th17 differentiation was reflected by higher production of IL-1β and IL-6 by CD103+ DCs compared with CD8+ DCs upon stimulation. The two types of DCs from isolated lymph nodes also differ in expression of certain pattern recognition receptors. Furthermore, elevated levels of GM-CSF, typical of those found in inflammation, substantially increased the pool size of CD103+ DCs in lymph nodes and skin. We argue that varied levels of GM-CSF may explain the contrasting reports regarding the positive role of GM-CSF in regulating development of CD103+ DCs. Together, we find that these two developmentally closely-related DC subsets display functional differences and that GM-CSF has differential effect on the two types of DCs.
Collapse
|
83
|
Toubai T, Mathewson N, Reddy P. The role of dendritic cells in graft-versus-tumor effect. Front Immunol 2014; 5:66. [PMID: 24600454 PMCID: PMC3930914 DOI: 10.3389/fimmu.2014.00066] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2013] [Accepted: 02/05/2014] [Indexed: 12/23/2022] Open
Abstract
Dendritic cells (DCs) are the most potent antigen presenting cells. DCs play a pivotal role in determining the character and magnitude of immune responses to tumors. Host and donor hematopoietic-derived DCs play a critical role in the development of graft-versus-host disease (GVHD) following allogeneic hematopoietic cell transplantation. GVHD is tightly linked with the graft-versus-tumor (GVT) effect. Although both host and donor DCs are important regulators of GVHD, the role of DCs in GVT is poorly understood. GVT is caused by donor T cells that attack recipient tumor cells. The donor T cells recognize alloantigens, and tumor specific antigens (TSAs) are mediating GVHD. The process of presentation of these antigens, especially TSAs remains unknown. Recent data suggested that DC may be essential role for inducing GVT. The mechanisms that DCs possess may include direct presentation, cross-presentation, cross-dressing. The role they play in GVT will be reviewed.
Collapse
Affiliation(s)
- Tomomi Toubai
- Blood and Marrow Transplantation Program, Division of Hematology and Oncology, Department of Internal Medicine, University of Michigan Comprehensive Cancer Center , Ann Arbor, MI , USA
| | - Nathan Mathewson
- Blood and Marrow Transplantation Program, Division of Hematology and Oncology, Department of Internal Medicine, University of Michigan Comprehensive Cancer Center , Ann Arbor, MI , USA
| | - Pavan Reddy
- Blood and Marrow Transplantation Program, Division of Hematology and Oncology, Department of Internal Medicine, University of Michigan Comprehensive Cancer Center , Ann Arbor, MI , USA
| |
Collapse
|
84
|
Seillet C, Huntington ND, Gangatirkar P, Axelsson E, Minnich M, Brady HJM, Busslinger M, Smyth MJ, Belz GT, Carotta S. Differential requirement for Nfil3 during NK cell development. THE JOURNAL OF IMMUNOLOGY 2014; 192:2667-76. [PMID: 24532575 DOI: 10.4049/jimmunol.1302605] [Citation(s) in RCA: 100] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
NK cells can be grouped into distinct subsets that are localized to different organs and exhibit a different capacity to secrete cytokines and mediate cytotoxicity. Despite these hallmarks that reflect tissue-specific specialization in NK cells, little is known about the factors that control the development of these distinct subsets. The basic leucine zipper transcription factor Nfil3 (E4bp4) is essential for bone marrow-derived NK cell development, but it is not clear whether Nfil3 is equally important for all NK cell subsets or how it induces NK lineage commitment. In this article, we show that Nfil3 is required for the formation of Eomes-expressing NK cells, including conventional medullary and thymic NK cells, whereas TRAIL(+) Eomes(-) NK cells develop independently of Nfil3. Loss of Nfil3 during the development of bone marrow-derived NK cells resulted in reduced expression of Eomes and, conversely, restoration of Eomes expression in Nfil3(-/-) progenitors rescued NK cell development and maturation. Collectively, these findings demonstrate that Nfil3 drives the formation of mature NK cells by inducing Eomes expression and reveal the differential requirements of NK cell subsets for Nfil3.
Collapse
Affiliation(s)
- Cyril Seillet
- Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria 3052, Australia
| | | | | | | | | | | | | | | | | | | |
Collapse
|
85
|
Abstract
The family of inhibitor of differentiation (Id) proteins is a group of evolutionarily conserved molecules, which play important regulatory roles in organisms ranging from Drosophila to humans. Id proteins are small polypeptides harboring a helix-loop-helix (HLH) motif, which are best known to mediate dimerization with other basic HLH proteins, primarily E proteins. Because Id proteins do not possess the basic amino acids adjacent to the HLH motif necessary for DNA binding, Id proteins inhibit the function of E protein homodimers, as well as heterodimers between E proteins and tissue-specific bHLH proteins. However, Id proteins have also been shown to have E protein-independent functions. The Id genes are broadly but differentially expressed in a variety of cell types. Transcription of the Id genes is controlled by transcription factors such as C/EBPβ and Egr as well as by signaling pathways triggered by different stimuli, which include bone morphogenic proteins, cytokines, and ligands of T cell receptors. In general, Id proteins are capable of inhibiting the differentiation of progenitors of different cell types, promoting cell-cycle progression, delaying cellular senescence, and facilitating cell migration. These properties of Id proteins enable them to play significant roles in stem cell maintenance, vasculogenesis, tumorigenesis and metastasis, the development of the immune system, and energy metabolism. In this review, we intend to highlight the current understanding of the function of Id proteins and discuss gaps in our knowledge about the mechanisms whereby Id proteins exert their diverse effects in multiple cellular processes.
Collapse
Affiliation(s)
- Flora Ling
- Immunobiology Cancer Research Program, Oklahoma Medical Research Foundation, Oklahoma City, Oklahoma, USA
| | - Bin Kang
- Immunobiology Cancer Research Program, Oklahoma Medical Research Foundation, Oklahoma City, Oklahoma, USA
| | - Xiao-Hong Sun
- Immunobiology Cancer Research Program, Oklahoma Medical Research Foundation, Oklahoma City, Oklahoma, USA.
| |
Collapse
|
86
|
Abstract
Dendritic cells (DCs) are professional antigen presenting cells involved critically not only in provoking innate immune responses but also in establishing adaptive immune responses. Dendritic cells are heterogenous and divided into several subsets, including plasmactyoid DCs (pDCs) and several types of conventional DCs (cDCs), which show subset-specific functions. Plasmactyoid DCs are featured by their ability to produce large amounts of type I interferons (IFNs) in response to nucleic acid sensors, TLR7 and TLR9 and involved in anti-viral immunity and pathogenesis of certain autoimmune disorders such as psoriasis. Conventional DCs include the DC subsets with high crosspresentation activity, which contributes to anti-viral and anti-tumor immunity. These subsets are generated from hematopoietic stem cells (HSCs) via several intermediate progenitors and the development is regulated by the transcriptional mechanisms in which subset-specific transcription factors play major roles. We have recently found that an Ets family transcription factor, SPI-B, which is abundantly expressed in pDCs among DC subsets, plays critical roles in functions and late stage development of pDCs. SPI-B functions in cooperation with other transcription factors, especially, interferon regulatory factor (IRF) family members. Here we review the transcription factor-based molecular mechanisms for generation and functions of DCs, mainly by focusing on the roles of SPI-B and its relatives.
Collapse
|
87
|
Chopin M, Seillet C, Chevrier S, Wu L, Wang H, Morse HC, Belz GT, Nutt SL. Langerhans cells are generated by two distinct PU.1-dependent transcriptional networks. ACTA ACUST UNITED AC 2013; 210:2967-80. [PMID: 24249112 PMCID: PMC3865480 DOI: 10.1084/jem.20130930] [Citation(s) in RCA: 87] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
Langerhans cell homeostasis and differentiation depends on PU.1, the latter via regulation of TGF-β–dependent binding of PU.1 to the regulatory elements of RUNX3. Langerhans cells (LCs) are the unique dendritic cells found in the epidermis. While a great deal of attention has focused on defining the developmental origins of LCs, reports addressing the transcriptional network ruling their differentiation remain sparse. We addressed the function of a group of key DC transcription factors—PU.1, ID2, IRF4, and IRF8—in the establishment of the LC network. We show that although steady-state LC homeostasis depends on PU.1 and ID2, the latter is dispensable for bone marrow–derived LCs. PU.1 controls LC differentiation by regulating the expression of the critical TGF-β responsive transcription factor RUNX3. PU.1 directly binds to the Runx3 regulatory elements in a TGF-β–dependent manner, whereas ectopic expression of RUNX3 rescued LC differentiation in the absence of PU.1 and promoted LC differentiation from PU.1-sufficient progenitors. These findings highlight the dual molecular network underlying LC differentiation, and show the central role of PU.1 in these processes.
Collapse
Affiliation(s)
- Michaël Chopin
- The Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria 3052, Australia
| | | | | | | | | | | | | | | |
Collapse
|
88
|
Jaiswal H, Kaushik M, Sougrat R, Gupta M, Dey A, Verma R, Ozato K, Tailor P. Batf3 and Id2 have a synergistic effect on Irf8-directed classical CD8α+ dendritic cell development. THE JOURNAL OF IMMUNOLOGY 2013; 191:5993-6001. [PMID: 24227775 DOI: 10.4049/jimmunol.1203541] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Dendritic cells (DCs) are heterogeneous cell populations represented by different subtypes, each varying in terms of gene expression patterns and specific functions. Recent studies identified transcription factors essential for the development of different DC subtypes, yet molecular mechanisms for the developmental program and functions remain poorly understood. In this study, we developed and characterized a mouse DC progenitor-like cell line, designated DC9, from Irf8(-/-) bone marrow cells as a model for DC development and function. Expression of Irf8 in DC9 cells led to plasmacytoid DCs and CD8α(+) DC-like cells, with a concomitant increase in plasmacytoid DC- and CD8α(+) DC-specific gene transcripts and induction of type I IFNs and IL12p40 following TLR ligand stimulation. Irf8 expression in DC9 cells led to an increase in Id2 and Batf3 transcript levels, transcription factors shown to be important for the development of CD8α(+) DCs. We show that, without Irf8, expression of Id2 and Batf3 was not sufficient for directing classical CD8α(+) DC development. When coexpressed with Irf8, Batf3 and Id2 had a synergistic effect on classical CD8α(+) DC development. We demonstrate that Irf8 is upstream of Batf3 and Id2 in the classical CD8α(+) DC developmental program and define the hierarchical relationship of transcription factors important for classical CD8α(+) DC development.
Collapse
Affiliation(s)
- Hemant Jaiswal
- Laboratory of Innate Immunity, National Institute of Immunology, New Delhi 110067, India
| | | | | | | | | | | | | | | |
Collapse
|
89
|
Mielke LA, Groom JR, Rankin LC, Seillet C, Masson F, Putoczki T, Belz GT. TCF-1 controls ILC2 and NKp46+RORγt+ innate lymphocyte differentiation and protection in intestinal inflammation. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2013; 191:4383-91. [PMID: 24038093 DOI: 10.4049/jimmunol.1301228] [Citation(s) in RCA: 110] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
Innate lymphocyte populations play a central role in conferring protective immunity at the mucosal frontier. In this study, we demonstrate that T cell factor 1 (TCF-1; encoded by Tcf7), a transcription factor also important for NK and T cell differentiation, is expressed by multiple innate lymphoid cell (ILC) subsets, including GATA3(+) nuocytes (ILC2) and NKp46(+) ILCs (ILC3), which confer protection against lung and intestinal inflammation. TCF-1 was intrinsically required for the differentiation of both ILC2 and NKp46(+) ILC3. Loss of TCF-1 expression impaired the capacity of these ILC subsets to produce IL-5, IL-13, and IL-22 and resulted in crippled responses to intestinal infection with Citrobacter rodentium. Furthermore, a reduction in T-bet expression required for Notch-2-dependent development of NKp46(+) ILC3 showed a dose-dependent reduction in TCF-1 expression. Collectively, our findings demonstrate an essential requirement for TCF-1 in ILC2 differentiation and reveal a link among Tcf7, Notch, and Tbx21 in NKp46(+) ILC3 development.
Collapse
Affiliation(s)
- Lisa A Mielke
- Division of Molecular Immunology, Walter and Eliza Hall Institute of Medical Research, Melbourne, Victoria 3052, Australia
| | | | | | | | | | | | | |
Collapse
|
90
|
Persson EK, Scott CL, Mowat AM, Agace WW. Dendritic cell subsets in the intestinal lamina propria: ontogeny and function. Eur J Immunol 2013; 43:3098-107. [PMID: 23966272 PMCID: PMC3933733 DOI: 10.1002/eji.201343740] [Citation(s) in RCA: 111] [Impact Index Per Article: 10.1] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2013] [Revised: 07/02/2013] [Accepted: 07/09/2013] [Indexed: 12/21/2022]
Abstract
The intestinal mucosa is exposed to large amounts of foreign antigen (Ag) derived from commensal bacteria, dietary Ags, and intestinal pathogens. Dendritic cells (DCs) are believed to be involved in the induction of tolerance to harmless Ags and in mounting protective immune responses to pathogens and, as such, to play key roles in regulating intestinal immune homeostasis. The characterization of classical DCs (cDCs) in the intestinal lamina propria has been under intense investigation in recent years but the use of markers (including CD11c, CD11b, MHC class II), which are also expressed by intestinal MΦs, has led to some controversy regarding their definition. Here we review recent studies that help to distinguish cDCs subsets from monocyte-derived cells in the intestinal mucosa. We address the phenotype and ontogeny of these cDC subsets and highlight recent findings indicating that these subsets play distinct roles in the regulation of mucosal immune responses in vivo.
Collapse
Affiliation(s)
- Emma K Persson
- Immunology Section, Department of Experimental Medical Science, Lund University, Lund, Sweden
| | | | | | | |
Collapse
|
91
|
Masson F, Minnich M, Olshansky M, Bilic I, Mount AM, Kallies A, Speed TP, Busslinger M, Nutt SL, Belz GT. Id2-mediated inhibition of E2A represses memory CD8+ T cell differentiation. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2013; 190:4585-94. [PMID: 23536629 PMCID: PMC3631715 DOI: 10.4049/jimmunol.1300099] [Citation(s) in RCA: 73] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/11/2013] [Accepted: 02/20/2013] [Indexed: 01/13/2023]
Abstract
The transcription factor inhibitor of DNA binding (Id)2 modulates T cell fate decisions, but the molecular mechanism underpinning this regulation is unclear. In this study we show that loss of Id2 cripples effector differentiation and instead programs CD8(+) T cells to adopt a memory fate with increased Eomesodermin and Tcf7 expression. We demonstrate that Id2 restrains CD8(+) T cell memory differentiation by inhibiting E2A-mediated direct activation of Tcf7 and that Id2 expression level mirrors T cell memory recall capacity. As a result of the defective effector differentiation, Id2-deficient CD8(+) T cells fail to induce sufficient Tbx21 expression to generate short-lived effector CD8(+) T cells. Our findings reveal that the Id2/E2A axis orchestrates T cell differentiation through the induction or repression of downstream transcription factors essential for effector and memory T cell differentiation.
Collapse
Affiliation(s)
- Frederick Masson
- Division of Molecular Immunology, Walter and Eliza Hall Institute of Medical Research, Melbourne, Victoria 3052, Australia
| | - Martina Minnich
- Research Institute of Molecular Pathology, Vienna Biocenter, 1030 Vienna, Austria
| | - Moshe Olshansky
- Division of Bioinformatics, Walter and Eliza Hall Institute of Medical Research, Melbourne, Victoria 3052, Australia
| | - Ivan Bilic
- Research Institute of Molecular Pathology, Vienna Biocenter, 1030 Vienna, Austria
| | - Adele M. Mount
- Research Institute of Molecular Pathology, Vienna Biocenter, 1030 Vienna, Austria
| | - Axel Kallies
- Division of Molecular Immunology, Walter and Eliza Hall Institute of Medical Research, Melbourne, Victoria 3052, Australia
- Department of Medical Biology, University of Melbourne, Melbourne, Victoria 3010, Australia; and
| | - Terence P. Speed
- Division of Bioinformatics, Walter and Eliza Hall Institute of Medical Research, Melbourne, Victoria 3052, Australia
- Department of Mathematics and Statistics, University of Melbourne, Melbourne, Victoria 3010, Australia
| | - Meinrad Busslinger
- Research Institute of Molecular Pathology, Vienna Biocenter, 1030 Vienna, Austria
| | - Stephen L. Nutt
- Division of Molecular Immunology, Walter and Eliza Hall Institute of Medical Research, Melbourne, Victoria 3052, Australia
- Department of Medical Biology, University of Melbourne, Melbourne, Victoria 3010, Australia; and
| | - Gabrielle T. Belz
- Division of Molecular Immunology, Walter and Eliza Hall Institute of Medical Research, Melbourne, Victoria 3052, Australia
- Department of Medical Biology, University of Melbourne, Melbourne, Victoria 3010, Australia; and
| |
Collapse
|
92
|
Hancock DG, Guy TV, Shklovskaya E, Fazekas de St Groth B. Experimental models to investigate the function of dendritic cell subsets: challenges and implications. Clin Exp Immunol 2013; 171:147-54. [PMID: 23286941 DOI: 10.1111/cei.12027] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/25/2012] [Indexed: 11/29/2022] Open
Abstract
The dendritic cell (DC) lineage is remarkably heterogeneous. It has been postulated that specialized DC subsets have evolved in order to select and support the multitude of possible T cell differentiation pathways. However, defining the function of individual DC subsets has proven remarkably difficult, and DC subset control of key T cell fates such as tolerance, T helper cell commitment and regulatory T cell induction is still not well understood. While the difficulty in assigning unique functions to particular DC subsets may be due to sharing of functions, it may also reflect a lack of appropriate physiological in-vivo models for studying DC function. In this paper we review the limitations associated with many of the current DC models and highlight some of the underlying difficulties involved in studying the function of murine DC subsets.
Collapse
Affiliation(s)
- D G Hancock
- Centenary Institute of Cancer Medicine and Cell Biology and the Discipline of Dermatology, University of Sydney, Sydney, NSW, Australia
| | | | | | | |
Collapse
|
93
|
Rankin L, Groom J, Mielke LA, Seillet C, Belz GT. Diversity, function, and transcriptional regulation of gut innate lymphocytes. Front Immunol 2013; 4:22. [PMID: 23508190 PMCID: PMC3600536 DOI: 10.3389/fimmu.2013.00022] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2012] [Accepted: 01/16/2013] [Indexed: 12/19/2022] Open
Abstract
The innate immune system plays a critical early role in host defense against viruses, bacteria, and tumor cells. Until recently, natural killer (NK) cells and lymphoid tissue inducer (LTi) cells were the primary members of the innate lymphocyte family: NK cells form the front-line interface between the external environment and the adaptive immune system, while LTi cells are essential for secondary lymphoid tissue formation. More recently, it has become apparent that the composition of this family is much more diverse than previously appreciated and newly recognized populations play distinct and essential functions in tissue protection. Despite the importance of these cells, the developmental relationships between different innate lymphocyte populations remain unclear. Here we review recent advances in our understanding of the development of different innate immune cell subsets, the transcriptional programs that might be involved in driving fate decisions during development, and their relationship to NK cells.
Collapse
Affiliation(s)
- Lucille Rankin
- Division of Molecular Immunology, The Walter and Eliza Hall Institute of Medical ResearchMelbourne, VIC, Australia
- Department of Medical Biology, University of MelbourneMelbourne, VIC, Australia
| | - Joanna Groom
- Division of Molecular Immunology, The Walter and Eliza Hall Institute of Medical ResearchMelbourne, VIC, Australia
- Department of Medical Biology, University of MelbourneMelbourne, VIC, Australia
| | - Lisa A. Mielke
- Division of Molecular Immunology, The Walter and Eliza Hall Institute of Medical ResearchMelbourne, VIC, Australia
- Department of Medical Biology, University of MelbourneMelbourne, VIC, Australia
| | - Cyril Seillet
- Division of Molecular Immunology, The Walter and Eliza Hall Institute of Medical ResearchMelbourne, VIC, Australia
- Department of Medical Biology, University of MelbourneMelbourne, VIC, Australia
| | - Gabrielle T. Belz
- Division of Molecular Immunology, The Walter and Eliza Hall Institute of Medical ResearchMelbourne, VIC, Australia
- Department of Medical Biology, University of MelbourneMelbourne, VIC, Australia
| |
Collapse
|
94
|
Rankin LC, Groom JR, Chopin M, Herold MJ, Walker JA, Mielke LA, McKenzie ANJ, Carotta S, Nutt SL, Belz GT. The transcription factor T-bet is essential for the development of NKp46+ innate lymphocytes via the Notch pathway. Nat Immunol 2013; 14:389-95. [PMID: 23455676 DOI: 10.1038/ni.2545] [Citation(s) in RCA: 238] [Impact Index Per Article: 21.6] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2012] [Accepted: 01/10/2013] [Indexed: 12/12/2022]
Abstract
NKp46+ innate lymphoid cells (ILCs) serve important roles in regulating the intestinal microbiota and defense against pathogens. Whether NKp46+ ILCs arise directly from lymphoid tissue-inducer (LTi) cells or represent a separate lineage remains controversial. We report here that the transcription factor T-bet (encoded by Tbx21) was essential for the development of NKp46+ ILCs but not of LTi cells or nuocytes. Deficiency in interleukin 22 (IL-22)-producing NKp46+ ILCs resulted in greater susceptibility of Tbx21-/- mice to intestinal infection. Haploinsufficient T-bet expression resulted in lower expression of the signaling molecule Notch, and Notch signaling was necessary for the transition of LTi cells into NKp46+ ILCs. Furthermore, NKp46+ ILCs differentiated solely from the CD4- LTi population, not the CD4+ LTi population. Our results pinpoint the regulation of Notch signaling by T-bet as a distinct molecular pathway that guides the development of NKp46+ ILCs.
Collapse
Affiliation(s)
- Lucille C Rankin
- The Walter and Eliza Hall Institute of Medical Research, Melbourne, Australia
| | | | | | | | | | | | | | | | | | | |
Collapse
|
95
|
Abstract
Pulmonary dendritic cells (DCs) constantly sample the tissue and traffic inhaled antigens to the lung-draining lymph node where they normally orchestrate an appropriate immune response. The dynamic ability of these professional antigen-presenting cells to promote tolerance or immunity has been intensively studied by several groups, including ours. Distinct DC subsets in both lymphoid and non-lymphoid tissues have been described based on their surface molecule expression and location. Current efforts to unravel DC development and function are providing insight into the various roles each subset offers the immune system. Elucidating DC functions, particularly in the lung, may then allow use of the inherent ability of these cells for enhanced vaccine strategies and therapeutics for pulmonary infections and diseases.
Collapse
Affiliation(s)
- A. Nicole Desch
- Integrated Department of Immunology, University of Colorado School of Medicine, Denver, CO, USA
| | - Peter M. Henson
- Department of Pediatrics, National Jewish Health, 1400 Jackson Street, Denver, CO 80206, USA
- Integrated Department of Immunology, University of Colorado School of Medicine, Denver, CO, USA
| | - Claudia V. Jakubzick
- Department of Pediatrics, National Jewish Health, 1400 Jackson Street, Denver, CO 80206, USA,
- Integrated Department of Immunology, University of Colorado School of Medicine, Denver, CO, USA
| |
Collapse
|
96
|
Abstract
Key Points
Transcription factors Batf3, Id2, and Nfil3 are not essential for induced CD8α+ DC generation. Induced CD8α+ DCs can cross-present cellular antigens.
Collapse
|
97
|
Moore AJ, Anderson MK. Dendritic cell development: a choose-your-own-adventure story. Adv Hematol 2013; 2013:949513. [PMID: 23476654 PMCID: PMC3588201 DOI: 10.1155/2013/949513] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2012] [Accepted: 12/27/2012] [Indexed: 12/13/2022] Open
Abstract
Dendritic cells (DCs) are essential components of the immune system and contribute to immune responses by activating or tolerizing T cells. DCs comprise a heterogeneous mixture of subsets that are located throughout the body and possess distinct and specialized functions. Although numerous defined precursors from the bone marrow and spleen have been identified, emerging data in the field suggests many alternative routes of DC differentiation from precursors with multilineage potential. Here, we discuss how the combinatorial expression of transcription factors can promote one DC lineage over another as well as the integration of cytokine signaling in this process.
Collapse
Affiliation(s)
- Amanda J. Moore
- Division of Biological Sciences, Sunnybrook Research Institute, 2075 Bayview Avenue, Toronto, ON, Canada M4N 3M5
- Department of Immunology, University of Toronto, Toronto, ON, Canada M5S 1A8
| | - Michele K. Anderson
- Division of Biological Sciences, Sunnybrook Research Institute, 2075 Bayview Avenue, Toronto, ON, Canada M4N 3M5
- Department of Immunology, University of Toronto, Toronto, ON, Canada M5S 1A8
| |
Collapse
|
98
|
Seré K, Baek JH, Ober-Blöbaum J, Müller-Newen G, Tacke F, Yokota Y, Zenke M, Hieronymus T. Two distinct types of Langerhans cells populate the skin during steady state and inflammation. Immunity 2013; 37:905-16. [PMID: 23159228 DOI: 10.1016/j.immuni.2012.07.019] [Citation(s) in RCA: 154] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2011] [Accepted: 07/30/2012] [Indexed: 11/20/2022]
Abstract
Langerhans cells (LCs), the dendritic cells (DCs) in skin epidermis, possess an exceptional life cycle and developmental origin. Here we identified two types of LCs, short-term and long-term LCs, which transiently or stably reconstitute the LC compartment, respectively. Short-term LCs developed from Gr-1(hi) monocytes under inflammatory conditions and occurred independently of the transcription factor Id2. Long-term LCs arose from bone marrow in steady state and were critically dependent on Id2. Surface marker and gene expression analysis positioned short-term LCs close to Gr-1(hi) monocytes, which is indicative of their monocytic origin. We also show that LC reconstitution after UV light exposure occurs in two waves: an initial fast and transient wave of Gr-1(hi) monocyte-derived short-term LCs is followed by a second wave of steady-state precursor-derived long-term LCs. Our data demonstrate the presence of two types of LCs that develop through different pathways in inflammation and steady state.
Collapse
Affiliation(s)
- Kristin Seré
- Institute for Biomedical Engineering, Department of Cell Biology, Medical Faculty, RWTH Aachen University, 52074 Aachen, Germany
| | | | | | | | | | | | | | | |
Collapse
|
99
|
Huntington ND, Nutt SL, Carotta S. Regulation of murine natural killer cell commitment. Front Immunol 2013; 4:14. [PMID: 23386852 PMCID: PMC3558707 DOI: 10.3389/fimmu.2013.00014] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2012] [Accepted: 01/08/2013] [Indexed: 02/02/2023] Open
Abstract
Natural killer (NK) cells can derive from the same precursors as B and T cells, however, to achieve lineage specificity, several transcription factors need to be activated or annulled. While a few important transcription factors have been identified for NK genesis the mechanisms of how this is achieved is far from resolved. Adding to the complexity of this, NK cells are found and potentially develop in diverse locations in vivo and it remains to be addressed if a common NK cell precursor seeds diverse niches and how transcription factors may differentially regulate NK cell commitment in distinct microenvironments. Here we will summarize some recent findings in NK cell commitment and discuss how a NK cell transcriptional network might be organized, while addressing some misconceptions and anomalies along the way.
Collapse
Affiliation(s)
- Nicholas D Huntington
- Molecular Immunology Division, Walter and Eliza Hall Institute of Medical Research Parkville, VIC, Australia ; Department of Medical Biology, The University of Melbourne Parkville, VIC, Australia
| | | | | |
Collapse
|
100
|
Abstract
Innate lymphoid cells (ILCs) are newly identified members of the lymphoid lineage that have emerging roles in mediating immune responses and in regulating tissue homeostasis and inflammation. Here, we review the developmental relationships between the various ILC lineages that have been identified to date and summarize their functions in protective immunity to infection and their pathological roles in allergic and autoimmune diseases.
Collapse
|