101
|
Salvermoser J, van Blijswijk J, Papaioannou NE, Rambichler S, Pasztoi M, Pakalniškytė D, Rogers NC, Keppler SJ, Straub T, Reis e Sousa C, Schraml BU. Clec9a-Mediated Ablation of Conventional Dendritic Cells Suggests a Lymphoid Path to Generating Dendritic Cells In Vivo. Front Immunol 2018; 9:699. [PMID: 29713321 PMCID: PMC5911463 DOI: 10.3389/fimmu.2018.00699] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2018] [Accepted: 03/21/2018] [Indexed: 01/01/2023] Open
Abstract
Conventional dendritic cells (cDCs) are versatile activators of immune responses that develop as part of the myeloid lineage downstream of hematopoietic stem cells. We have recently shown that in mice precursors of cDCs, but not of other leukocytes, are marked by expression of DNGR-1/CLEC9A. To genetically deplete DNGR-1-expressing cDC precursors and their progeny, we crossed Clec9a-Cre mice to Rosa-lox-STOP-lox-diphtheria toxin (DTA) mice. These mice develop signs of age-dependent myeloproliferative disease, as has been observed in other DC-deficient mouse models. However, despite efficient depletion of cDC progenitors in these mice, cells with phenotypic characteristics of cDCs populate the spleen. These cells are functionally and transcriptionally similar to cDCs in wild type control mice but show somatic rearrangements of Ig-heavy chain genes, characteristic of lymphoid origin cells. Our studies reveal a previously unappreciated developmental heterogeneity of cDCs and suggest that the lymphoid lineage can generate cells with features of cDCs when myeloid cDC progenitors are impaired.
Collapse
Affiliation(s)
- Johanna Salvermoser
- Walter-Brendel-Centre for Experimental Medicine, University Hospital, LMU Munich, Planegg Martinsried, Germany.,Biomedical Center, LMU Munich, Planegg Martinsried, Germany
| | | | | | - Stephan Rambichler
- Walter-Brendel-Centre for Experimental Medicine, University Hospital, LMU Munich, Planegg Martinsried, Germany.,Biomedical Center, LMU Munich, Planegg Martinsried, Germany
| | - Maria Pasztoi
- Biomedical Center, LMU Munich, Planegg Martinsried, Germany
| | - Dalia Pakalniškytė
- Walter-Brendel-Centre for Experimental Medicine, University Hospital, LMU Munich, Planegg Martinsried, Germany.,Biomedical Center, LMU Munich, Planegg Martinsried, Germany
| | - Neil C Rogers
- Immunobiology Laboratory, The Francis Crick Institute, London, United Kingdom
| | - Selina J Keppler
- Technische Universität München, Klinikum Rechts der Isar, Institut für Klinische Chemie und Pathobiochemie, Munich, Germany
| | - Tobias Straub
- Biomedical Center, LMU Munich, Planegg Martinsried, Germany.,Core Facility Bioinformatics, Biomedical Center (BMC), LMU Munich, Planegg Martinsried, Germany
| | | | - Barbara U Schraml
- Walter-Brendel-Centre for Experimental Medicine, University Hospital, LMU Munich, Planegg Martinsried, Germany.,Biomedical Center, LMU Munich, Planegg Martinsried, Germany
| |
Collapse
|
102
|
Durai V, Bagadia P, Briseño CG, Theisen DJ, Iwata A, Davidson JT, Gargaro M, Fremont DH, Murphy TL, Murphy KM. Altered compensatory cytokine signaling underlies the discrepancy between Flt3-/- and Flt3l-/- mice. J Exp Med 2018; 215:1417-1435. [PMID: 29572360 PMCID: PMC5940266 DOI: 10.1084/jem.20171784] [Citation(s) in RCA: 57] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2017] [Revised: 01/10/2018] [Accepted: 02/22/2018] [Indexed: 12/19/2022] Open
Abstract
The receptor Flt3 and its ligand Flt3L are both critical for dendritic cell (DC) development, but DC deficiency is more severe in Flt3l-/- mice than in Flt3-/- mice. This has led to speculation that Flt3L binds to another receptor that also supports DC development. However, we found that Flt3L administration does not generate DCs in Flt3-/- mice, arguing against a second receptor. Instead, Flt3-/- DC progenitors matured in response to macrophage colony-stimulating factor (M-CSF) or stem cell factor, and deletion of Csf1r in Flt3-/- mice further reduced DC development, indicating that these cytokines could compensate for Flt3. Surprisingly, Flt3-/- DC progenitors displayed enhanced M-CSF signaling, suggesting that loss of Flt3 increased responsiveness to other cytokines. In agreement, deletion of Flt3 in Flt3l-/- mice paradoxically rescued their severe DC deficiency. Thus, multiple cytokines can support DC development, and the discrepancy between Flt3-/- and Flt3l-/- mice results from the increased sensitivity of Flt3-/- progenitors to these cytokines.
Collapse
Affiliation(s)
- Vivek Durai
- Department of Pathology and Immunology, Washington University in St. Louis, School of Medicine, St. Louis, MO
| | - Prachi Bagadia
- Department of Pathology and Immunology, Washington University in St. Louis, School of Medicine, St. Louis, MO
| | - Carlos G Briseño
- Department of Pathology and Immunology, Washington University in St. Louis, School of Medicine, St. Louis, MO
| | - Derek J Theisen
- Department of Pathology and Immunology, Washington University in St. Louis, School of Medicine, St. Louis, MO
| | - Arifumi Iwata
- Department of Pathology and Immunology, Washington University in St. Louis, School of Medicine, St. Louis, MO
| | - Jesse T Davidson
- Department of Pathology and Immunology, Washington University in St. Louis, School of Medicine, St. Louis, MO
| | - Marco Gargaro
- Department of Pathology and Immunology, Washington University in St. Louis, School of Medicine, St. Louis, MO
| | - Daved H Fremont
- Department of Pathology and Immunology, Washington University in St. Louis, School of Medicine, St. Louis, MO
| | - Theresa L Murphy
- Department of Pathology and Immunology, Washington University in St. Louis, School of Medicine, St. Louis, MO
| | - Kenneth M Murphy
- Department of Pathology and Immunology, Washington University in St. Louis, School of Medicine, St. Louis, MO .,Howard Hughes Medical Institute, Washington University in St. Louis, School of Medicine, St. Louis, MO
| |
Collapse
|
103
|
James KD, Jenkinson WE, Anderson G. T-cell egress from the thymus: Should I stay or should I go? J Leukoc Biol 2018; 104:275-284. [PMID: 29485734 PMCID: PMC6174998 DOI: 10.1002/jlb.1mr1217-496r] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2017] [Revised: 01/11/2018] [Accepted: 01/22/2018] [Indexed: 02/02/2023] Open
Abstract
T‐cells bearing the αβTCR play a vital role in defending the host against foreign pathogens and malignant transformation of self. Importantly, T‐cells are required to remain tolerant to the host's own cells and tissues in order to prevent self‐reactive responses that can lead to autoimmune disease. T‐cells achieve the capacity for self/nonself discrimination by undergoing a highly selective and rigorous developmental program during their maturation in the thymus. This organ is unique in its ability to support a program of T‐cell development that ensures the establishment of a functionally diverse αβTCR repertoire within the peripheral T‐cell pool. The thymus achieves this by virtue of specialized stromal microenvironments that contain heterogeneous cell types, whose organization and function underpins their ability to educate, support, and screen different thymocyte subsets through various stages of development. These stages range from the entry of early T‐cell progenitors into the thymus, through to the positive and negative selection of the αβTCR repertoire. The importance of the thymus medulla as a site for T‐cell tolerance and the exit of newly generated T‐cells into the periphery is well established. In this review, we summarize current knowledge on the developmental pathways that take place during αβT‐cell development in the thymus. In addition, we focus on the mechanisms that regulate thymic egress and contribute to the seeding of peripheral tissues with newly selected self‐tolerant αβT‐cells. Review on thymic microenvironments regulation of thymocyte maturation and egress of mature self‐tolerant T cells.
Collapse
Affiliation(s)
- Kieran D James
- Institute of Immunology and Immunotherapy, College of Medical and Dental Sciences, Medical School, University of Birmingham, Birmingham, UK
| | - William E Jenkinson
- Institute of Immunology and Immunotherapy, College of Medical and Dental Sciences, Medical School, University of Birmingham, Birmingham, UK
| | - Graham Anderson
- Institute of Immunology and Immunotherapy, College of Medical and Dental Sciences, Medical School, University of Birmingham, Birmingham, UK
| |
Collapse
|
104
|
Kadel S, Ainsua-Enrich E, Hatipoglu I, Turner S, Singh S, Khan S, Kovats S. A Major Population of Functional KLRG1 - ILC2s in Female Lungs Contributes to a Sex Bias in ILC2 Numbers. Immunohorizons 2018; 2:74-86. [PMID: 29568816 DOI: 10.4049/immunohorizons.1800008] [Citation(s) in RCA: 61] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023] Open
Abstract
Humans show significant sex differences in the incidence and severity of respiratory diseases, including asthma and virus infection. Sex hormones contribute to the female sex bias in type 2 inflammation associated with respiratory diseases, consistent with recent reports that female lungs harbor greater numbers of GATA-3-dependent group 2 innate lymphoid cells (ILC2s). In this study, we determined whether sex hormone levels govern sex differences in the numbers, phenotype, and function of ILC2s in the murine lung and bone marrow (BM). Our data show that lungs of female mice harbor significantly greater ILC2 numbers in homeostasis, in part due to a major subset of ILC2s lacking killer-cell lectin like receptor G1 (KLRG1), a population largely absent in male lungs. The KLRG1- ILC2s were capable of type 2 cytokine production and increased with age after sexual maturity, suggesting that a unique functional subset exists in females. Experiments with gonadectomized mice or mice bearing either global or lymphocyte restricted estrogen receptor α (Esr1) deficiency showed that androgens rather than estrogens regulated numbers of the KLRG1- ILC2 subset and ILC2 functional capacity in the lung and BM, as well as levels of GATA-3 expression in BM ILC2s. Furthermore, the frequency of BM PLZF+ ILC precursors was higher in males and increased by excess androgens, suggesting that androgens act to inhibit the transition of ILC precursors to ILC2s. Taken together, these data show that a functional subset of KLRG1- ILC2s in females contributes to the sex bias in lung ILC2s that is observed after reproductive age.
Collapse
Affiliation(s)
- Sapana Kadel
- Arthritis and Clinical Immunology Program, Oklahoma Medical Research Foundation, Oklahoma City, OK 73104.,Department of Microbiology and Immunology, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104
| | - Erola Ainsua-Enrich
- Arthritis and Clinical Immunology Program, Oklahoma Medical Research Foundation, Oklahoma City, OK 73104
| | - Ibrahim Hatipoglu
- Arthritis and Clinical Immunology Program, Oklahoma Medical Research Foundation, Oklahoma City, OK 73104
| | - Sean Turner
- Arthritis and Clinical Immunology Program, Oklahoma Medical Research Foundation, Oklahoma City, OK 73104
| | - Simar Singh
- Arthritis and Clinical Immunology Program, Oklahoma Medical Research Foundation, Oklahoma City, OK 73104
| | - Sohaib Khan
- Department of Cancer and Cell Biology, College of Medicine, University of Cincinnati, Cincinnati, OH 45267
| | - Susan Kovats
- Arthritis and Clinical Immunology Program, Oklahoma Medical Research Foundation, Oklahoma City, OK 73104.,Department of Microbiology and Immunology, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104
| |
Collapse
|
105
|
Krueger A. Thymus Colonization: Who, How, How Many? Arch Immunol Ther Exp (Warsz) 2017; 66:81-88. [PMID: 29288431 DOI: 10.1007/s00005-017-0503-5] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2017] [Accepted: 11/23/2017] [Indexed: 11/28/2022]
Abstract
De novo generation of T cells depends on continual colonization of the thymus by bone marrow-derived progenitors. Thymus seeding progenitors (TSPs) constitute a heterogeneous population comprising multipotent and lineage-restricted cell types. Entry into the thymic microenvironment is tightly controlled and recent quantitative studies have revealed that the adult murine thymus only contains approximately 160 niches to accommodate TSPs. Of these niches only about 6% are open for seeding on average at steady-state. Here, I review the state of understanding of colonization of the adult murine thymus with a particular focus on past and current controversies in the field. Improving thymus colonization and/or maintaining intact TSP niches during the course of pre-conditioning regimens are likely to be critical for efficient T-cell regeneration after hematopoietic stem cell transplantation.
Collapse
Affiliation(s)
- Andreas Krueger
- Institute for Molecular Medicine, Goethe University Frankfurt am Main, 60590, Frankfurt am Main, Germany.
| |
Collapse
|
106
|
Harly C, Cam M, Kaye J, Bhandoola A. Development and differentiation of early innate lymphoid progenitors. J Exp Med 2017; 215:249-262. [PMID: 29183988 PMCID: PMC5748853 DOI: 10.1084/jem.20170832] [Citation(s) in RCA: 91] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2017] [Revised: 09/19/2017] [Accepted: 10/26/2017] [Indexed: 01/05/2023] Open
Abstract
Early innate lymphoid progenitors (EILPs) have recently been identified in mouse adult bone marrow as a multipotential progenitor population specified toward innate lymphoid cell (ILC) lineages, but their relationship with other described ILC progenitors is still unclear. In this study, we examine the progenitor-successor relationships between EILPs, all-lymphoid progenitors (ALPs), and ILC precursors (ILCps). Functional, bioinformatic, phenotypical, and genetic approaches collectively establish EILPs as an intermediate progenitor between ALPs and ILCps. Our work additionally provides new candidate regulators of ILC development and clearly defines the stage of requirement of transcription factors key for early ILC development.
Collapse
Affiliation(s)
- Christelle Harly
- Laboratory of Genome Integrity, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD
| | - Maggie Cam
- CCR Collaborative Bioinformatics Resource, Office of Science and Technology Resources, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD
| | - Jonathan Kaye
- Research Division of Immunology, Department of Biomedical Sciences, Samuel Oschin Comprehensive Cancer Institute, Cedars-Sinai Medical Center, Los Angeles, CA.,Research Division of Immunology, Department of Medicine, Samuel Oschin Comprehensive Cancer Institute, Cedars-Sinai Medical Center, Los Angeles, CA
| | - Avinash Bhandoola
- Laboratory of Genome Integrity, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD
| |
Collapse
|
107
|
Saluzzo S, Gorki AD, Rana BMJ, Martins R, Scanlon S, Starkl P, Lakovits K, Hladik A, Korosec A, Sharif O, Warszawska JM, Jolin H, Mesteri I, McKenzie ANJ, Knapp S. First-Breath-Induced Type 2 Pathways Shape the Lung Immune Environment. Cell Rep 2017; 18:1893-1905. [PMID: 28228256 PMCID: PMC5329122 DOI: 10.1016/j.celrep.2017.01.071] [Citation(s) in RCA: 196] [Impact Index Per Article: 24.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2016] [Revised: 12/27/2016] [Accepted: 01/26/2017] [Indexed: 01/07/2023] Open
Abstract
From birth onward, the lungs are exposed to the external environment and therefore harbor a complex immunological milieu to protect this organ from damage and infection. We investigated the homeostatic role of the epithelium-derived alarmin interleukin-33 (IL-33) in newborn mice and discovered the immediate upregulation of IL-33 from the first day of life, closely followed by a wave of IL-13-producing type 2 innate lymphoid cells (ILC2s), which coincided with the appearance of alveolar macrophages (AMs) and their early polarization to an IL-13-dependent anti-inflammatory M2 phenotype. ILC2s contributed to lung quiescence in homeostasis by polarizing tissue resident AMs and induced an M2 phenotype in transplanted macrophage progenitors. ILC2s continued to maintain the M2 AM phenotype during adult life at the cost of a delayed response to Streptococcus pneumoniae infection in mice. These data highlight the homeostatic role of ILC2s in setting the activation threshold in the lung and underline their implications in anti-bacterial defenses. The first breath triggers IL-33 induction by AEC2 in lungs of newborn mice IL-33 promotes the perinatal expansion and activation of ST2-expressing ILC2s ILC2-derived IL-13 polarizes newborn’s AMs into an M2 phenotype This homeostatic type 2 pathway delays antibacterial effector responses
Collapse
Affiliation(s)
- Simona Saluzzo
- CeMM Research Center for Molecular Medicine of the Austrian Academy of Sciences, Vienna 1090, Austria; Department of Medicine I, Laboratory of Infection Biology, Medical University of Vienna, Vienna 1090, Austria; MRC Laboratory of Molecular Biology, Francis Crick Avenue, Cambridge CB2 0QH, UK
| | - Anna-Dorothea Gorki
- CeMM Research Center for Molecular Medicine of the Austrian Academy of Sciences, Vienna 1090, Austria; Department of Medicine I, Laboratory of Infection Biology, Medical University of Vienna, Vienna 1090, Austria
| | - Batika M J Rana
- MRC Laboratory of Molecular Biology, Francis Crick Avenue, Cambridge CB2 0QH, UK
| | - Rui Martins
- CeMM Research Center for Molecular Medicine of the Austrian Academy of Sciences, Vienna 1090, Austria; Department of Medicine I, Laboratory of Infection Biology, Medical University of Vienna, Vienna 1090, Austria
| | - Seth Scanlon
- MRC Laboratory of Molecular Biology, Francis Crick Avenue, Cambridge CB2 0QH, UK
| | - Philipp Starkl
- CeMM Research Center for Molecular Medicine of the Austrian Academy of Sciences, Vienna 1090, Austria; Department of Medicine I, Laboratory of Infection Biology, Medical University of Vienna, Vienna 1090, Austria
| | - Karin Lakovits
- CeMM Research Center for Molecular Medicine of the Austrian Academy of Sciences, Vienna 1090, Austria; Department of Medicine I, Laboratory of Infection Biology, Medical University of Vienna, Vienna 1090, Austria
| | - Anastasiya Hladik
- CeMM Research Center for Molecular Medicine of the Austrian Academy of Sciences, Vienna 1090, Austria; Department of Medicine I, Laboratory of Infection Biology, Medical University of Vienna, Vienna 1090, Austria
| | - Ana Korosec
- CeMM Research Center for Molecular Medicine of the Austrian Academy of Sciences, Vienna 1090, Austria; Department of Medicine I, Laboratory of Infection Biology, Medical University of Vienna, Vienna 1090, Austria
| | - Omar Sharif
- CeMM Research Center for Molecular Medicine of the Austrian Academy of Sciences, Vienna 1090, Austria; Department of Medicine I, Laboratory of Infection Biology, Medical University of Vienna, Vienna 1090, Austria
| | - Joanna M Warszawska
- CeMM Research Center for Molecular Medicine of the Austrian Academy of Sciences, Vienna 1090, Austria; Department of Medicine I, Laboratory of Infection Biology, Medical University of Vienna, Vienna 1090, Austria
| | - Helen Jolin
- MRC Laboratory of Molecular Biology, Francis Crick Avenue, Cambridge CB2 0QH, UK
| | - Ildiko Mesteri
- Institute of Pathology Überlingen, Überlingen 88662, Germany
| | - Andrew N J McKenzie
- MRC Laboratory of Molecular Biology, Francis Crick Avenue, Cambridge CB2 0QH, UK.
| | - Sylvia Knapp
- CeMM Research Center for Molecular Medicine of the Austrian Academy of Sciences, Vienna 1090, Austria; Department of Medicine I, Laboratory of Infection Biology, Medical University of Vienna, Vienna 1090, Austria.
| |
Collapse
|
108
|
Abstract
Purpose of review Hematopoietic stem cell (HSC) transplantation has yielded tremendous information on experimental properties of HSCs. Yet, it remains unclear whether transplantation reflects the physiology of hematopoiesis. A limitation is the difficulty in accessing HSC functions without isolation, in-vitro manipulation and readout for potential. New genetic fate mapping and clonal marking techniques now shed light on hematopoiesis under physiological conditions. Recent findings Transposon-based genetic marks were introduced across the entire hematopoietic system to follow the clonal dynamics of these tags over time. A polyclonal source downstream from stem cells was found responsible for the production of at least granulocytes. In independent experiments, HSCs were genetically marked in adult mice, and the kinetics of label emergence throughout the system was followed over time. These experiments uncovered that during physiological steady-state hematopoiesis large numbers of HSCs yield differentiated progeny. Individual HSCs were active only rarely, indicating their very slow periodicity of differentiation rather than quiescence. Summary Noninvasive genetic experiments in mice have identified a major role of stem and progenitor cells downstream from HSCs as drivers of adult hematopoiesis, and revealed that post-transplantation hematopoiesis differs quantitatively from normal steady-state hematopoiesis.
Collapse
|
109
|
Mao AP, Ishizuka IE, Kasal DN, Mandal M, Bendelac A. A shared Runx1-bound Zbtb16 enhancer directs innate and innate-like lymphoid lineage development. Nat Commun 2017; 8:863. [PMID: 29038474 PMCID: PMC5643357 DOI: 10.1038/s41467-017-00882-0] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2017] [Accepted: 08/02/2017] [Indexed: 02/07/2023] Open
Abstract
Zbtb16-encoded PLZF is a signature transcription factor (TF) that directs the acquisition of T-helper effector programs during the development of multiple innate lymphocyte lineages, including natural killer T cell, innate lymphoid cell, mucosal-associated invariant T cell and γδ lineages. PLZF is also essential in osteoblast and spermatogonial development. How Zbtb16 itself is regulated in different lineages is incompletely understood. Here, by systematic CRISPR/Cas9-assisted deletions of chromatin accessible regions within the Zbtb16 locus in mouse, we identify a critical enhancer controlling PLZF expression exclusively in innate lymphoid lineages. Multiple sites within this enhancer express canonical motifs for the TF Runx1, which is essential for the development of these lineages. Notably, some regulatory sites control the kinetic rather than the overall level of PLZF expression. Thus, our comprehensive, unbiased analysis of regulatory elements in vivo reveals critical mechanisms of Zbtb16 regulation shared between innate and innate-like lymphoid lineages. Zbtb16-encoded transcription factor PLZF directs the differentiation of multiple innate and innate-like cell lineages, but how Zbtb16 itself is regulated remains unclear. Here the authors show, using CRISPR gene editing, ATAC-seq and ChIP-seq, that specific Runx1-bound enhancer elements critically modulate lineage-dependent expressions of PLZF.
Collapse
Affiliation(s)
- Ai-Ping Mao
- Committee on Immunology, University of Chicago, Chicago, IL, 60637, USA.,Department of Pathology, University of Chicago, Chicago, IL, 60637, USA
| | - Isabel E Ishizuka
- Committee on Immunology, University of Chicago, Chicago, IL, 60637, USA.,Department of Pathology, University of Chicago, Chicago, IL, 60637, USA
| | - Darshan N Kasal
- Committee on Immunology, University of Chicago, Chicago, IL, 60637, USA.,Department of Pathology, University of Chicago, Chicago, IL, 60637, USA
| | - Malay Mandal
- Committee on Immunology, University of Chicago, Chicago, IL, 60637, USA.,Department of Medicine, Section of Rheumatology and Gwen Knapp Center for Lupus and Immunology Research, University of Chicago, Chicago, IL, 60637, USA
| | - Albert Bendelac
- Committee on Immunology, University of Chicago, Chicago, IL, 60637, USA. .,Department of Pathology, University of Chicago, Chicago, IL, 60637, USA.
| |
Collapse
|
110
|
Alberti-Servera L, von Muenchow L, Tsapogas P, Capoferri G, Eschbach K, Beisel C, Ceredig R, Ivanek R, Rolink A. Single-cell RNA sequencing reveals developmental heterogeneity among early lymphoid progenitors. EMBO J 2017; 36:3619-3633. [PMID: 29030486 DOI: 10.15252/embj.201797105] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2017] [Revised: 08/11/2017] [Accepted: 09/13/2017] [Indexed: 12/21/2022] Open
Abstract
Single-cell RNA sequencing is a powerful technology for assessing heterogeneity within defined cell populations. Here, we describe the heterogeneity of a B220+CD117intCD19-NK1.1- uncommitted hematopoietic progenitor having combined lymphoid and myeloid potential. Phenotypic and functional assays revealed four subpopulations within the progenitor with distinct lineage developmental potentials. Among them, the Ly6D+SiglecH-CD11c- fraction was lymphoid-restricted exhibiting strong B-cell potential, whereas the Ly6D-SiglecH-CD11c- fraction showed mixed lympho-myeloid potential. Single-cell RNA sequencing of these subsets revealed that the latter population comprised a mixture of cells with distinct lymphoid and myeloid transcriptional signatures and identified a subgroup as the potential precursor of Ly6D+SiglecH-CD11c- Subsequent functional assays confirmed that B220+CD117intCD19-NK1.1- single cells are, with rare exceptions, not bipotent for lymphoid and myeloid lineages. A B-cell priming gradient was observed within the Ly6D+SiglecH-CD11c- subset and we propose a herein newly identified subgroup as the direct precursor of the first B-cell committed stage. Therefore, the apparent multipotency of B220+CD117intCD19-NK1.1- progenitors results from underlying heterogeneity at the single-cell level and highlights the validity of single-cell transcriptomics for resolving cellular heterogeneity and developmental relationships among hematopoietic progenitors.
Collapse
Affiliation(s)
- Llucia Alberti-Servera
- Developmental and Molecular Immunology, Department of Biomedicine, University of Basel, Basel, Switzerland
| | - Lilly von Muenchow
- Developmental and Molecular Immunology, Department of Biomedicine, University of Basel, Basel, Switzerland
| | - Panagiotis Tsapogas
- Developmental and Molecular Immunology, Department of Biomedicine, University of Basel, Basel, Switzerland
| | - Giuseppina Capoferri
- Developmental and Molecular Immunology, Department of Biomedicine, University of Basel, Basel, Switzerland
| | - Katja Eschbach
- Genomics Facility, Department of Biosystems Science and Engineering, ETH Zurich, Basel, Switzerland
| | - Christian Beisel
- Genomics Facility, Department of Biosystems Science and Engineering, ETH Zurich, Basel, Switzerland
| | - Rhodri Ceredig
- Discipline of Physiology, College of Medicine & Nursing Health Science National University of Ireland, Galway, Ireland
| | - Robert Ivanek
- Department of Biomedicine, University of Basel and University Hospital Basel, Basel, Switzerland.,Swiss Institute of Bioinformatics, Basel, Switzerland
| | - Antonius Rolink
- Developmental and Molecular Immunology, Department of Biomedicine, University of Basel, Basel, Switzerland
| |
Collapse
|
111
|
Loughran SJ, Comoglio F, Hamey FK, Giustacchini A, Errami Y, Earp E, Göttgens B, Jacobsen SEW, Mead AJ, Hendrich B, Green AR. Mbd3/NuRD controls lymphoid cell fate and inhibits tumorigenesis by repressing a B cell transcriptional program. J Exp Med 2017; 214:3085-3104. [PMID: 28899870 PMCID: PMC5626393 DOI: 10.1084/jem.20161827] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2016] [Revised: 07/04/2017] [Accepted: 07/25/2017] [Indexed: 02/02/2023] Open
Abstract
Differentiation of lineage-committed cells from multipotent progenitors requires the establishment of accessible chromatin at lineage-specific transcriptional enhancers and promoters, which is mediated by pioneer transcription factors that recruit activating chromatin remodeling complexes. Here we show that the Mbd3/nucleosome remodeling and deacetylation (NuRD) chromatin remodeling complex opposes this transcriptional pioneering during B cell programming of multipotent lymphoid progenitors by restricting chromatin accessibility at B cell enhancers and promoters. Mbd3/NuRD-deficient lymphoid progenitors therefore prematurely activate a B cell transcriptional program and are biased toward overproduction of pro-B cells at the expense of T cell progenitors. The striking reduction in early thymic T cell progenitors results in compensatory hyperproliferation of immature thymocytes and development of T cell lymphoma. Our results reveal that Mbd3/NuRD can regulate multilineage differentiation by constraining the activation of dormant lineage-specific enhancers and promoters. In this way, Mbd3/NuRD protects the multipotency of lymphoid progenitors, preventing B cell-programming transcription factors from prematurely enacting lineage commitment. Mbd3/NuRD therefore controls the fate of lymphoid progenitors, ensuring appropriate production of lineage-committed progeny and suppressing tumor formation.
Collapse
Affiliation(s)
- Stephen J Loughran
- Department of Haematology, Cambridge Institute for Medical Research, University of Cambridge, Cambridge, England, UK
- Wellcome Trust/MRC Stem Cell Institute, University of Cambridge, Cambridge, England, UK
| | - Federico Comoglio
- Department of Haematology, Cambridge Institute for Medical Research, University of Cambridge, Cambridge, England, UK
- Wellcome Trust/MRC Stem Cell Institute, University of Cambridge, Cambridge, England, UK
| | - Fiona K Hamey
- Department of Haematology, Cambridge Institute for Medical Research, University of Cambridge, Cambridge, England, UK
- Wellcome Trust/MRC Stem Cell Institute, University of Cambridge, Cambridge, England, UK
| | - Alice Giustacchini
- MRC Weatherall Institute of Molecular Medicine, University of Oxford, Oxford, England, UK
| | - Youssef Errami
- Department of Haematology, Cambridge Institute for Medical Research, University of Cambridge, Cambridge, England, UK
- Wellcome Trust/MRC Stem Cell Institute, University of Cambridge, Cambridge, England, UK
| | - Eleanor Earp
- Department of Haematology, Cambridge Institute for Medical Research, University of Cambridge, Cambridge, England, UK
- Wellcome Trust/MRC Stem Cell Institute, University of Cambridge, Cambridge, England, UK
| | - Berthold Göttgens
- Department of Haematology, Cambridge Institute for Medical Research, University of Cambridge, Cambridge, England, UK
- Wellcome Trust/MRC Stem Cell Institute, University of Cambridge, Cambridge, England, UK
| | - Sten Eirik W Jacobsen
- MRC Weatherall Institute of Molecular Medicine, University of Oxford, Oxford, England, UK
- Wallenberg Institute for Regenerative Medicine, Department of Cell and Molecular Biology and Department of Medicine Huddinge, Karolinska Institutet and Center for Hematology and Regenerative Medicine, Karolinska University Hospital Huddinge, Stockholm, Sweden
| | - Adam J Mead
- MRC Weatherall Institute of Molecular Medicine, University of Oxford, Oxford, England, UK
| | - Brian Hendrich
- Wellcome Trust/MRC Stem Cell Institute, University of Cambridge, Cambridge, England, UK
- Department of Biochemistry, University of Cambridge, Cambridge, England, UK
| | - Anthony R Green
- Department of Haematology, Cambridge Institute for Medical Research, University of Cambridge, Cambridge, England, UK
- Wellcome Trust/MRC Stem Cell Institute, University of Cambridge, Cambridge, England, UK
| |
Collapse
|
112
|
Moore AJ, In TS, Trotman-Grant A, Yoganathan K, Montpellier B, Guidos CJ, Zúñiga-Pflücker JC, Anderson MK. A key role for IL-7R in the generation of microenvironments required for thymic dendritic cells. Immunol Cell Biol 2017; 95:933-942. [PMID: 28890536 PMCID: PMC5698111 DOI: 10.1038/icb.2017.74] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2016] [Revised: 08/10/2017] [Accepted: 08/24/2017] [Indexed: 11/21/2022]
Abstract
Interleukin-7 receptor (IL-7R) signaling is critical for multiple stages of T-cell development, but a role in the establishment of the mature thymic architecture needed for T-cell development and thymocyte selection has not been established. Crosstalk signals between developing thymocytes and thymic epithelial cell (TEC) precursors are critical for their differentiation into cortical TECs (cTECs) and medullary TECs (mTECs). In addition, mTEC-derived factors have been implicated in the recruitment of thymic dendritic cells (DCs) and intrathymic DC development. We therefore examined corticomedullary structure and DC populations in the thymus of Il7r−/− mice. Analysis of TEC phenotype and spatial organization revealed a striking shift in the mTEC to cTEC ratio, accompanied by disorganized corticomedullary structure. Several of the thymic subsets known to have DC potential were nearly absent, accompanied by reductions in DC cell numbers. We also examined chemokine expression in the Il7r−/− thymus, and found a significant decrease in mTEC-derived CCR7 ligand expression, and high levels of cTEC-derived chemokines, including CCL25 and CXCL12. Although splenic DCs were similarly affected, bone marrow (BM) precursors capable of giving rise to DCs were unperturbed. Finally, BM chimeras showed that there was no intrinsic need for IL-7R signaling in the development or recruitment of thymic DCs, but that the provision of wild-type progenitors enhanced reconstitution of thymic DCs from Il7r−/− progenitors. Our results are therefore supportive of a model in which Il7r-dependent cells are required to set up the microenvironments that allow accumulation of thymic DCs.
Collapse
Affiliation(s)
- Amanda J Moore
- Biological Sciences, Sunnybrook Research Institute, Toronto, Ontario, Canada.,Department of Immunology, University of Toronto, Toronto, Ontario, Canada
| | - Tracy Sh In
- Biological Sciences, Sunnybrook Research Institute, Toronto, Ontario, Canada.,Department of Immunology, University of Toronto, Toronto, Ontario, Canada
| | - Ashton Trotman-Grant
- Biological Sciences, Sunnybrook Research Institute, Toronto, Ontario, Canada.,Department of Immunology, University of Toronto, Toronto, Ontario, Canada
| | - Kogulan Yoganathan
- Biological Sciences, Sunnybrook Research Institute, Toronto, Ontario, Canada.,Department of Immunology, University of Toronto, Toronto, Ontario, Canada
| | - Bertrand Montpellier
- Department of Immunology, University of Toronto, Toronto, Ontario, Canada.,Program in Developmental and Stem Cell Biology, Hospital for Sick Children Research Institute, Toronto, Ontario, Canada
| | - Cynthia J Guidos
- Department of Immunology, University of Toronto, Toronto, Ontario, Canada.,Program in Developmental and Stem Cell Biology, Hospital for Sick Children Research Institute, Toronto, Ontario, Canada
| | - Juan Carlos Zúñiga-Pflücker
- Biological Sciences, Sunnybrook Research Institute, Toronto, Ontario, Canada.,Department of Immunology, University of Toronto, Toronto, Ontario, Canada
| | - Michele K Anderson
- Biological Sciences, Sunnybrook Research Institute, Toronto, Ontario, Canada.,Department of Immunology, University of Toronto, Toronto, Ontario, Canada
| |
Collapse
|
113
|
Barik S, Miller MM, Cattin-Roy AN, Ukah TK, Chen W, Zaghouani H. IL-4/IL-13 Signaling Inhibits the Potential of Early Thymic Progenitors To Commit to the T Cell Lineage. THE JOURNAL OF IMMUNOLOGY 2017; 199:2767-2776. [PMID: 28893952 DOI: 10.4049/jimmunol.1700498] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/05/2017] [Accepted: 08/15/2017] [Indexed: 11/19/2022]
Abstract
Early thymic progenitors (ETPs) are endowed with diverse potencies and can give rise to myeloid and lymphoid lineage progenitors. How the thymic environment guides ETP commitment and maturation toward a specific lineage remains obscure. We have previously shown that ETPs expressing the heteroreceptor (HR) comprising IL-4Rα and IL-13Rα1 give rise to myeloid cells but not T cells. In this article, we show that signaling through the HR inhibits ETP maturation to the T cell lineage but enacts commitment toward the myeloid cells. Indeed, HR+ ETPs, but not HR- ETPs, exhibit activated STAT6 transcription factor, which parallels with downregulation of Notch1, a critical factor for T cell development. Meanwhile, the myeloid-specific transcription factor C/EBPα, usually under the control of Notch1, is upregulated. Furthermore, in vivo inhibition of STAT6 phosphorylation restores Notch1 expression in HR+ ETPs, which regain T lineage potential. In addition, upon stimulation with IL-4 or IL-13, HR- ETPs expressing virally transduced HR also exhibit STAT6 phosphorylation and downregulation of Notch1, leading to inhibition of lymphoid, but not myeloid, lineage potential. These observations indicate that environmental cytokines play a role in conditioning ETP lineage choice, which would impact T cell development.
Collapse
Affiliation(s)
- Subhasis Barik
- Department of Molecular Microbiology and Immunology, University of Missouri School of Medicine, Columbia, MO 65212
| | - Mindy M Miller
- Department of Molecular Microbiology and Immunology, University of Missouri School of Medicine, Columbia, MO 65212
| | - Alexis N Cattin-Roy
- Department of Molecular Microbiology and Immunology, University of Missouri School of Medicine, Columbia, MO 65212
| | - Tobechukwu K Ukah
- Department of Molecular Microbiology and Immunology, University of Missouri School of Medicine, Columbia, MO 65212
| | - Weirong Chen
- Department of Molecular Microbiology and Immunology, University of Missouri School of Medicine, Columbia, MO 65212
| | - Habib Zaghouani
- Department of Molecular Microbiology and Immunology, University of Missouri School of Medicine, Columbia, MO 65212; .,Department of Child Health, University of Missouri School of Medicine, Columbia, MO 65212; and.,Department of Neurology, University of Missouri School of Medicine, Columbia, MO 65212
| |
Collapse
|
114
|
The E-Id Protein Axis Specifies Adaptive Lymphoid Cell Identity and Suppresses Thymic Innate Lymphoid Cell Development. Immunity 2017; 46:818-834.e4. [PMID: 28514688 DOI: 10.1016/j.immuni.2017.04.022] [Citation(s) in RCA: 72] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2016] [Revised: 02/26/2017] [Accepted: 04/27/2017] [Indexed: 01/24/2023]
Abstract
Innate and adaptive lymphoid development is orchestrated by the activities of E proteins and their antagonist Id proteins, but how these factors regulate early T cell progenitor (ETP) and innate lymphoid cell (ILC) development remains unclear. Using multiple genetic strategies, we demonstrated that E proteins E2A and HEB acted in synergy in the thymus to establish T cell identity and to suppress the aberrant development of ILCs, including ILC2s and lymphoid-tissue-inducer-like cells. E2A and HEB orchestrated T cell fate and suppressed the ILC transcription signature by activating the expression of genes associated with Notch receptors, T cell receptor (TCR) assembly, and TCR-mediated signaling. E2A and HEB acted in ETPs to establish and maintain a T-cell-lineage-specific enhancer repertoire, including regulatory elements associated with the Notch1, Rag1, and Rag2 loci. On the basis of these and previous observations, we propose that the E-Id protein axis specifies innate and adaptive lymphoid cell fate.
Collapse
|
115
|
Berthault C, Ramond C, Burlen-Defranoux O, Soubigou G, Chea S, Golub R, Pereira P, Vieira P, Cumano A. Asynchronous lineage priming determines commitment to T cell and B cell lineages in fetal liver. Nat Immunol 2017; 18:1139-1149. [PMID: 28825702 DOI: 10.1038/ni.3820] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2017] [Accepted: 07/31/2017] [Indexed: 12/15/2022]
Abstract
The molecular events that initiate lymphoid-lineage specification remain unidentified because the stages of differentiation during which lineage commitment occurs are difficult to characterize. We isolated fetal liver progenitor cells undergoing restriction of their differentiation potential toward the T cell-innate lymphoid cell lineage or the B cell lineage. Transcripts that defined the molecular signatures of these two subsets were sequentially upregulated in lympho-myeloid precursor cells and in common lymphoid progenitor cells, respectively, and this preceded lineage restriction; this indicates that T cell-versus-B cell commitment is not a binary fate 'decision'. The T cell-bias and B cell-bias transcriptional programs were frequently co-expressed in common lymphoid progenitor cells and were segregated in subsets biased toward T cell differentiation or B cell differentiation, after interleukin 7 (IL-7) signaling that controlled the number of progenitor cells engaging in T cell differentiation versus B cell differentiation.
Collapse
Affiliation(s)
- Claire Berthault
- Unit for Lymphopoiesis, Pasteur Institute, Paris, France. Immunology department.,INSERM U1223, Paris, France.,Université Paris Diderot, Sorbonne Paris Cité, Cellule Pasteur, Paris, France
| | - Cyrille Ramond
- Unit for Lymphopoiesis, Pasteur Institute, Paris, France. Immunology department.,INSERM U1223, Paris, France.,Université Pierre et Marie Curie, Paris, France
| | - Odile Burlen-Defranoux
- Unit for Lymphopoiesis, Pasteur Institute, Paris, France. Immunology department.,INSERM U1223, Paris, France.,Université Paris Diderot, Sorbonne Paris Cité, Cellule Pasteur, Paris, France
| | - Guillaume Soubigou
- Département Génomes et Génétique, Plate-forme Transcriptome et Epigénome, Institut Pasteur, Paris, France
| | - Sylvestre Chea
- Unit for Lymphopoiesis, Pasteur Institute, Paris, France. Immunology department.,INSERM U1223, Paris, France.,Université Paris Diderot, Sorbonne Paris Cité, Cellule Pasteur, Paris, France
| | - Rachel Golub
- Unit for Lymphopoiesis, Pasteur Institute, Paris, France. Immunology department.,INSERM U1223, Paris, France.,Université Paris Diderot, Sorbonne Paris Cité, Cellule Pasteur, Paris, France
| | - Pablo Pereira
- Unit for Lymphopoiesis, Pasteur Institute, Paris, France. Immunology department.,INSERM U1223, Paris, France.,Université Paris Diderot, Sorbonne Paris Cité, Cellule Pasteur, Paris, France
| | - Paulo Vieira
- Unit for Lymphopoiesis, Pasteur Institute, Paris, France. Immunology department.,INSERM U1223, Paris, France.,Université Paris Diderot, Sorbonne Paris Cité, Cellule Pasteur, Paris, France
| | - Ana Cumano
- Unit for Lymphopoiesis, Pasteur Institute, Paris, France. Immunology department.,INSERM U1223, Paris, France.,Université Paris Diderot, Sorbonne Paris Cité, Cellule Pasteur, Paris, France
| |
Collapse
|
116
|
Interleukin-7 in the transition of bone marrow progenitors to the thymus. Immunol Cell Biol 2017; 95:916-924. [PMID: 28811625 DOI: 10.1038/icb.2017.68] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2016] [Revised: 08/01/2017] [Accepted: 08/01/2017] [Indexed: 02/03/2023]
Abstract
Interleukin-7 (IL-7) is essential for the development of T cells in humans and mice where deficiencies in IL-7 signaling result in severe immunodeficiency. T cells require IL-7 at multiple points during development; however, it is unclear when IL-7 is first necessary. We observed that mice with impaired IL-7 signaling had a large reduction in the number of early thymic progenitors (ETPs) while mice that overexpress IL-7 had greatly increased numbers of ETPs. These results indicated that the development of ETPs is sensitive to IL-7. Bone marrow progenitors of ETP are present in normal numbers in mice with impaired IL-7 signaling (IL-7Rα449F) and were efficiently recruited to the thymus. Furthermore, ETPs and their progenitors from IL-7Rα449F mice did not undergo increased apoptosis and proliferate normally compared to WT cells. Mixed bone marrow chimeras demonstrated that IL-7 signaling has a cell-intrinsic role in ETP development but was not required for development of bone marrow progenitors. We have shown a novel role for IL-7 signaling in the development of ETPs that is distinct from classic mechanisms of IL-7 regulating survival and proliferation.
Collapse
|
117
|
Cortez VS, Ulland TK, Cervantes-Barragan L, Bando JK, Robinette ML, Wang Q, White AJ, Gilfillan S, Cella M, Colonna M. SMAD4 impedes the conversion of NK cells into ILC1-like cells by curtailing non-canonical TGF-β signaling. Nat Immunol 2017; 18:995-1003. [PMID: 28759002 DOI: 10.1038/ni.3809] [Citation(s) in RCA: 265] [Impact Index Per Article: 33.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2017] [Accepted: 06/28/2017] [Indexed: 12/12/2022]
Abstract
Among the features that distinguish type 1 innate lymphoid cells (ILC1s) from natural killer (NK) cells is a gene signature indicative of 'imprinting' by cytokines of the TGF-β family. We studied mice in which ILC1s and NK cells lacked SMAD4, a signal transducer that facilitates the canonical signaling pathway common to all cytokines of the TGF-β family. While SMAD4 deficiency did not affect ILC1 differentiation, NK cells unexpectedly acquired an ILC1-like gene signature and were unable to control tumor metastasis or viral infection. Mechanistically, SMAD4 restrained non-canonical TGF-β signaling mediated by the cytokine receptor TGFβR1 in NK cells. NK cells from a SMAD4-deficient person affected by polyposis were also hyper-responsive to TGF-β. These results identify SMAD4 as a previously unknown regulator that restricts non-canonical TGF-β signaling in NK cells.
Collapse
Affiliation(s)
- Victor S Cortez
- Department of Pathology and Immunology, Washington University School of Medicine, St Louis, Missouri, USA
| | - Tyler K Ulland
- Department of Pathology and Immunology, Washington University School of Medicine, St Louis, Missouri, USA
| | - Luisa Cervantes-Barragan
- Department of Pathology and Immunology, Washington University School of Medicine, St Louis, Missouri, USA
| | - Jennifer K Bando
- Department of Pathology and Immunology, Washington University School of Medicine, St Louis, Missouri, USA
| | - Michelle L Robinette
- Department of Pathology and Immunology, Washington University School of Medicine, St Louis, Missouri, USA
| | - Qianli Wang
- Department of Pathology and Immunology, Washington University School of Medicine, St Louis, Missouri, USA
| | - Andrew J White
- Division of Pediatric Rheumatology and Department of Pediatrics, Washington University School of Medicine, St Louis, Missouri, USA
| | - Susan Gilfillan
- Department of Pathology and Immunology, Washington University School of Medicine, St Louis, Missouri, USA
| | - Marina Cella
- Department of Pathology and Immunology, Washington University School of Medicine, St Louis, Missouri, USA
| | - Marco Colonna
- Department of Pathology and Immunology, Washington University School of Medicine, St Louis, Missouri, USA
| |
Collapse
|
118
|
Schwartz C, Khan AR, Floudas A, Saunders SP, Hams E, Rodewald HR, McKenzie ANJ, Fallon PG. ILC2s regulate adaptive Th2 cell functions via PD-L1 checkpoint control. J Exp Med 2017; 214:2507-2521. [PMID: 28747424 PMCID: PMC5584124 DOI: 10.1084/jem.20170051] [Citation(s) in RCA: 102] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2017] [Revised: 06/02/2017] [Accepted: 07/05/2017] [Indexed: 12/21/2022] Open
Abstract
Group 2 innate lymphoid cells (ILC2s) are important effector cells driving the initiation of type 2 immune responses leading to adaptive T helper 2 (Th2) immunity. Here we show that ILC2s dynamically express the checkpoint inhibitor molecule PD-L1 during type 2 pulmonary responses. Surprisingly, PD-L1:PD-1 interaction between ILC2s and CD4+ T cells did not inhibit the T cell response, but PD-L1-expressing ILC2s stimulated increased expression of GATA3 and production of IL-13 by Th2 cells both in vitro and in vivo. Conditional deletion of PD-L1 on ILC2s impaired early Th2 polarization and cytokine production, leading to delayed worm expulsion during infection with the gastrointestinal helminth Nippostrongylus brasiliensis Our results identify a novel PD-L1-controlled mechanism for type 2 polarization, with ILC2s mediating an innate checkpoint to control adaptive T helper responses, which has important implications for the treatment of type 2 inflammation.
Collapse
Affiliation(s)
- Christian Schwartz
- Trinity Biomedical Sciences Institute, School of Medicine, Trinity College Dublin, Dublin, Ireland.,Trinity Translational Medicine Institute, Trinity College Dublin, Dublin, Ireland
| | - Adnan R Khan
- Trinity Biomedical Sciences Institute, School of Medicine, Trinity College Dublin, Dublin, Ireland.,Trinity Translational Medicine Institute, Trinity College Dublin, Dublin, Ireland
| | - Achilleas Floudas
- Trinity Biomedical Sciences Institute, School of Medicine, Trinity College Dublin, Dublin, Ireland.,Trinity Translational Medicine Institute, Trinity College Dublin, Dublin, Ireland
| | - Sean P Saunders
- Trinity Biomedical Sciences Institute, School of Medicine, Trinity College Dublin, Dublin, Ireland.,Trinity Translational Medicine Institute, Trinity College Dublin, Dublin, Ireland
| | - Emily Hams
- Trinity Biomedical Sciences Institute, School of Medicine, Trinity College Dublin, Dublin, Ireland.,Trinity Translational Medicine Institute, Trinity College Dublin, Dublin, Ireland
| | - Hans-Reimer Rodewald
- Division of Cellular Immunology, German Cancer Research Center, Heidelberg, Germany
| | | | - Padraic G Fallon
- Trinity Biomedical Sciences Institute, School of Medicine, Trinity College Dublin, Dublin, Ireland .,Trinity Translational Medicine Institute, Trinity College Dublin, Dublin, Ireland.,National Children's Research Centre, Our Lady's Children's Hospital, Crumlin, Dublin, Ireland
| |
Collapse
|
119
|
Helft J, Anjos-Afonso F, van der Veen AG, Chakravarty P, Bonnet D, Reis e Sousa C. Dendritic Cell Lineage Potential in Human Early Hematopoietic Progenitors. Cell Rep 2017; 20:529-537. [PMID: 28723558 PMCID: PMC5529209 DOI: 10.1016/j.celrep.2017.06.075] [Citation(s) in RCA: 53] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2017] [Revised: 05/22/2017] [Accepted: 06/23/2017] [Indexed: 01/16/2023] Open
Abstract
Conventional dendritic cells (cDCs) are thought to descend from a DC precursor downstream of the common myeloid progenitor (CMP). However, a mouse lymphoid-primed multipotent progenitor has been shown to generate cDCs following a DC-specific developmental pathway independent of monocyte and granulocyte poiesis. Similarly, here we show that, in humans, a large fraction of multipotent lymphoid early progenitors (MLPs) gives rise to cDCs, in particular the subset known as cDC1, identified by co-expression of DNGR-1 (CLEC9A) and CD141 (BDCA-3). Single-cell analysis indicates that over one-third of MLPs have the potential to efficiently generate cDCs. cDC1s generated from CMPs or MLPs do not exhibit differences in transcriptome or phenotype. These results demonstrate an early imprinting of the cDC lineage in human hematopoiesis and highlight the plasticity of developmental pathways giving rise to human DCs.
Collapse
Affiliation(s)
- Julie Helft
- Immunobiology Laboratory, The Francis Crick Institute, 1 Midland Road, London NW1 1AT, UK.
| | - Fernando Anjos-Afonso
- Haematopoietic Stem Cell Laboratory, The Francis Crick Institute, 1 Midland Road, London NW1 1AT, UK; Cardiff University, European Cancer Stem Cell Institute, Cardiff, CF10 3XQ, UK
| | | | - Probir Chakravarty
- Bioinformatics, The Francis Crick Institute, 1 Midland Road, London NW1 1AT, UK
| | - Dominique Bonnet
- Haematopoietic Stem Cell Laboratory, The Francis Crick Institute, 1 Midland Road, London NW1 1AT, UK
| | - Caetano Reis e Sousa
- Immunobiology Laboratory, The Francis Crick Institute, 1 Midland Road, London NW1 1AT, UK.
| |
Collapse
|
120
|
Newland SA, Mohanta S, Clément M, Taleb S, Walker JA, Nus M, Sage AP, Yin C, Hu D, Kitt LL, Finigan AJ, Rodewald HR, Binder CJ, McKenzie ANJ, Habenicht AJ, Mallat Z. Type-2 innate lymphoid cells control the development of atherosclerosis in mice. Nat Commun 2017; 8:15781. [PMID: 28589929 PMCID: PMC5467269 DOI: 10.1038/ncomms15781] [Citation(s) in RCA: 93] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2016] [Accepted: 04/27/2017] [Indexed: 01/06/2023] Open
Abstract
Type-2 innate lymphoid cells (ILC2) are a prominent source of type II cytokines and are found constitutively at mucosal surfaces and in visceral adipose tissue. Despite their role in limiting obesity, how ILC2s respond to high fat feeding is poorly understood, and their direct influence on the development of atherosclerosis has not been explored. Here, we show that ILC2 are present in para-aortic adipose tissue and lymph nodes and display an inflammatory-like phenotype atypical of adipose resident ILC2. High fat feeding alters both the number of ILC2 and their type II cytokine production. Selective genetic ablation of ILC2 in Ldlr−/− mice accelerates the development of atherosclerosis, which is prevented by reconstitution with wild type but not Il5−/− or Il13−/− ILC2. We conclude that ILC2 represent a major innate cell source of IL-5 and IL-13 required for mounting atheroprotective immunity, which can be altered by high fat diet. Type-2 innate lymphoid cells (ILC2) affect adipose tissue metabolism and function. Here the authors show that the ILC2 are present in para-aortic adipose tissue and represent a major source of IL-5 and IL-13 required for mounting atheroprotective immunity, which can be altered by high fat diet.
Collapse
Affiliation(s)
- Stephen A Newland
- Department of Medicine, Division of Cardiovascular Medicine, University of Cambridge, Cambridge CB2 0SZ, UK
| | - Sarajo Mohanta
- Institute for Cardiovascular Prevention, Ludwig-Maximilians-University (LMU), 80336 Munich, Germany
| | - Marc Clément
- Department of Medicine, Division of Cardiovascular Medicine, University of Cambridge, Cambridge CB2 0SZ, UK
| | - Soraya Taleb
- Institut National de la Santé et de la Recherche Médicale, U970 Paris, France
| | - Jennifer A Walker
- Division of Protein and Nucleic Acid Chemistry, MRC Laboratory of Molecular Biology, Cambridge CB2 0QH, UK
| | - Meritxell Nus
- Department of Medicine, Division of Cardiovascular Medicine, University of Cambridge, Cambridge CB2 0SZ, UK
| | - Andrew P Sage
- Department of Medicine, Division of Cardiovascular Medicine, University of Cambridge, Cambridge CB2 0SZ, UK
| | - Changjun Yin
- Institute for Cardiovascular Prevention, Ludwig-Maximilians-University (LMU), 80336 Munich, Germany
| | - Desheng Hu
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, Xiamen University, Xiamen, Fujian 361102, China
| | - Lauren L Kitt
- Department of Medicine, Division of Cardiovascular Medicine, University of Cambridge, Cambridge CB2 0SZ, UK
| | - Alison J Finigan
- Department of Medicine, Division of Cardiovascular Medicine, University of Cambridge, Cambridge CB2 0SZ, UK
| | - Hans-Reimer Rodewald
- Division of Cellular Immunology, German Cancer Research Center, 69120 Heidelberg, Germany
| | - Christoph J Binder
- Department of Laboratory Medicine, Medical University of Vienna and Center for Molecular Medicine (CeMM) of the Austrian Academy of Sciences, 1090 Vienna, Austria
| | - Andrew N J McKenzie
- Division of Protein and Nucleic Acid Chemistry, MRC Laboratory of Molecular Biology, Cambridge CB2 0QH, UK
| | - Andreas J Habenicht
- Institute for Cardiovascular Prevention, Ludwig-Maximilians-University (LMU), 80336 Munich, Germany
| | - Ziad Mallat
- Department of Medicine, Division of Cardiovascular Medicine, University of Cambridge, Cambridge CB2 0SZ, UK.,Institut National de la Santé et de la Recherche Médicale, U970 Paris, France
| |
Collapse
|
121
|
Bando JK, Colonna M. Innate lymphoid cell function in the context of adaptive immunity. Nat Immunol 2017; 17:783-9. [PMID: 27328008 DOI: 10.1038/ni.3484] [Citation(s) in RCA: 91] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2016] [Accepted: 05/04/2016] [Indexed: 12/12/2022]
Abstract
Innate lymphoid cells (ILCs) are a family of innate immune cells that have diverse functions during homeostasis and disease. Subsets of ILCs have phenotypes that mirror those of polarized helper T cell subsets in their expression of core transcription factors and effector cytokines. Given the similarities between these two classes of lymphocytes, it is important to understand which functions of ILCs are specialized and which are redundant with those of T cells. Here we discuss genetic mouse models that have been used to delineate the contributions of ILCs versus those of T cells and review the current understanding of the specialized in vivo functions of ILCs.
Collapse
Affiliation(s)
- Jennifer K Bando
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Marco Colonna
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, Missouri, USA
| |
Collapse
|
122
|
Development of innate lymphoid cells. Nat Immunol 2017; 17:775-82. [PMID: 27328007 DOI: 10.1038/ni.3481] [Citation(s) in RCA: 164] [Impact Index Per Article: 20.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2016] [Accepted: 05/04/2016] [Indexed: 12/13/2022]
Abstract
Innate lymphoid cells (ILCs) are a family of immune effector cells that have important roles in host defense, metabolic homeostasis and tissue repair but can also contribute to inflammatory diseases such as asthma and colitis. These cells can be categorized into three groups on the basis of the transcription factors that direct their function and the cytokines they produce, which parallel the effector functions of T lymphocytes. The hierarchy of cell-fate-restriction events that occur as common lymphoid progenitors become committed to each of the ILC lineages further underscores the relationship between these innate immune cells and T lymphocytes. In this Review we discuss the developmental program of ILCs and transcription factors that guide ILC lineage specification and commitment.
Collapse
|
123
|
Jacobsen JA, Woodard J, Mandal M, Clark MR, Bartom ET, Sigvardsson M, Kee BL. EZH2 Regulates the Developmental Timing of Effectors of the Pre-Antigen Receptor Checkpoints. THE JOURNAL OF IMMUNOLOGY 2017; 198:4682-4691. [PMID: 28490575 DOI: 10.4049/jimmunol.1700319] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/01/2017] [Accepted: 04/17/2017] [Indexed: 12/11/2022]
Abstract
The histone methyltransferase EZH2 is required for B and T cell development; however, the molecular mechanisms underlying this requirement remain elusive. In a murine model of lymphoid-specific EZH2 deficiency we found that EZH2 was required for proper development of adaptive, but not innate, lymphoid cells. In adaptive lymphoid cells EZH2 prevented the premature expression of Cdkn2a and the consequent stabilization of p53, an effector of the pre-Ag receptor checkpoints. Deletion of Cdkn2a in EZH2-deficient lymphocytes prevented p53 stabilization, extended lymphocyte survival, and restored differentiation resulting in the generation of mature B and T lymphocytes. Our results uncover a crucial role for EZH2 in adaptive lymphocytes to control the developmental timing of effectors of the pre-Ag receptor checkpoints.
Collapse
Affiliation(s)
| | - Jennifer Woodard
- Committee on Immunology, The University of Chicago, Chicago, IL 60637
| | - Malay Mandal
- Division of Rheumatology, Department of Medicine, The University of Chicago, Chicago, IL 60637
| | - Marcus R Clark
- Committee on Immunology, The University of Chicago, Chicago, IL 60637.,Division of Rheumatology, Department of Medicine, The University of Chicago, Chicago, IL 60637
| | | | - Mikael Sigvardsson
- Department of Molecular Hematology, Lund University, 22184 Lund, Sweden; and
| | - Barbara L Kee
- Committee on Immunology, The University of Chicago, Chicago, IL 60637; .,Department of Pathology, The University of Chicago, Chicago, IL 60637
| |
Collapse
|
124
|
Chen YCE, Mapp S, Blumenthal A, Burgess ML, Mazzieri R, Mattarollo SR, Mollee P, Gill D, Saunders NA. The duality of macrophage function in chronic lymphocytic leukaemia. Biochim Biophys Acta Rev Cancer 2017; 1868:176-182. [PMID: 28347751 DOI: 10.1016/j.bbcan.2017.03.006] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2017] [Revised: 03/09/2017] [Accepted: 03/21/2017] [Indexed: 12/23/2022]
Abstract
Chronic lymphocytic leukaemia (CLL) is the most common adult leukaemia and, in some patients, is accompanied by resistance to both chemotherapeutics and immunotherapeutics. In this review we will discuss the role of tumour associated macrophages (TAMs) in promoting CLL cell survival and resistance to immunotherapeutics. In addition, we will discuss mechanisms by which TAMs suppress T-cell mediated antitumour responses. Thus, targeting macrophages could be used to i) reduce the leukaemic burden via the induction of T-cell-mediated antitumour responses, ii) to reduce pro-survival signalling and enhance response to conventional chemotherapeutics or iii) enhance the response to therapeutic antibodies in current clinical use.
Collapse
Affiliation(s)
- Y C E Chen
- University of Queensland Diamantina Institute, University of Queensland, Translational Research Institute, Princess Alexandra Hospital, Brisbane, Qld, Australia
| | - S Mapp
- Department of Haematology, Division of Cancer Services, Princess Alexandra Hospital, Brisbane, Qld, Australia
| | - A Blumenthal
- University of Queensland Diamantina Institute, University of Queensland, Translational Research Institute, Princess Alexandra Hospital, Brisbane, Qld, Australia
| | - M L Burgess
- University of Queensland Diamantina Institute, University of Queensland, Translational Research Institute, Princess Alexandra Hospital, Brisbane, Qld, Australia; Department of Haematology, Division of Cancer Services, Princess Alexandra Hospital, Brisbane, Qld, Australia
| | - R Mazzieri
- University of Queensland Diamantina Institute, University of Queensland, Translational Research Institute, Princess Alexandra Hospital, Brisbane, Qld, Australia
| | - S R Mattarollo
- University of Queensland Diamantina Institute, University of Queensland, Translational Research Institute, Princess Alexandra Hospital, Brisbane, Qld, Australia
| | - P Mollee
- Department of Haematology, Division of Cancer Services, Princess Alexandra Hospital, Brisbane, Qld, Australia
| | - D Gill
- University of Queensland Diamantina Institute, University of Queensland, Translational Research Institute, Princess Alexandra Hospital, Brisbane, Qld, Australia; Department of Haematology, Division of Cancer Services, Princess Alexandra Hospital, Brisbane, Qld, Australia
| | - N A Saunders
- University of Queensland Diamantina Institute, University of Queensland, Translational Research Institute, Princess Alexandra Hospital, Brisbane, Qld, Australia.
| |
Collapse
|
125
|
Rothenberg EV, Kueh HY, Yui MA, Zhang JA. Hematopoiesis and T-cell specification as a model developmental system. Immunol Rev 2016; 271:72-97. [PMID: 27088908 DOI: 10.1111/imr.12417] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
The pathway to generate T cells from hematopoietic stem cells guides progenitors through a succession of fate choices while balancing differentiation progression against proliferation, stage to stage. Many elements of the regulatory system that controls this process are known, but the requirement for multiple, functionally distinct transcription factors needs clarification in terms of gene network architecture. Here, we compare the features of the T-cell specification system with the rule sets underlying two other influential types of gene network models: first, the combinatorial, hierarchical regulatory systems that generate the orderly, synchronized increases in complexity in most invertebrate embryos; second, the dueling 'master regulator' systems that are commonly used to explain bistability in microbial systems and in many fate choices in terminal differentiation. The T-cell specification process shares certain features with each of these prevalent models but differs from both of them in central respects. The T-cell system is highly combinatorial but also highly dose-sensitive in its use of crucial regulatory factors. The roles of these factors are not always T-lineage-specific, but they balance and modulate each other's activities long before any mutually exclusive silencing occurs. T-cell specification may provide a new hybrid model for gene networks in vertebrate developmental systems.
Collapse
Affiliation(s)
- Ellen V Rothenberg
- Division of Biology & Biological Engineering, California Institute of Technology, Pasadena, CA, USA
| | - Hao Yuan Kueh
- Division of Biology & Biological Engineering, California Institute of Technology, Pasadena, CA, USA
| | - Mary A Yui
- Division of Biology & Biological Engineering, California Institute of Technology, Pasadena, CA, USA
| | - Jingli A Zhang
- Division of Biology & Biological Engineering, California Institute of Technology, Pasadena, CA, USA
| |
Collapse
|
126
|
Krueger A, Ziętara N, Łyszkiewicz M. T Cell Development by the Numbers. Trends Immunol 2016; 38:128-139. [PMID: 27842955 DOI: 10.1016/j.it.2016.10.007] [Citation(s) in RCA: 55] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2016] [Revised: 10/21/2016] [Accepted: 10/25/2016] [Indexed: 01/01/2023]
Abstract
T cells are continually generated in the thymus in a highly dynamic process comprising discrete steps of lineage commitment, T cell receptor (TCR) gene rearrangement, and selection. These steps are linked to distinct rates of proliferation, survival, and cell death, but a quantitative picture of T cell development is only beginning to emerge. Here we summarize recent technical advances, including genetic fate mapping, barcoding, and molecular timers, that have allowed the implementation of computational models to quantify developmental dynamics in the thymus. Coupling new techniques with mathematical models has recently resulted in the emergence of new paradigms in early hematopoiesis and might similarly open new perspectives on T cell development.
Collapse
Affiliation(s)
- Andreas Krueger
- Institute of Molecular Medicine, Goethe University Frankfurt am Main, 60590 Frankfurt am Main, Germany.
| | - Natalia Ziętara
- Dr von Hauner Children's Hospital, Ludwig Maximilian University, 80337 Munich, Germany
| | - Marcin Łyszkiewicz
- Dr von Hauner Children's Hospital, Ludwig Maximilian University, 80337 Munich, Germany
| |
Collapse
|
127
|
Lucas B, James KD, Cosway EJ, Parnell SM, Tumanov AV, Ware CF, Jenkinson WE, Anderson G. Lymphotoxin β Receptor Controls T Cell Progenitor Entry to the Thymus. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2016; 197:2665-72. [PMID: 27549174 PMCID: PMC5026032 DOI: 10.4049/jimmunol.1601189] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/08/2016] [Accepted: 07/29/2016] [Indexed: 11/19/2022]
Abstract
The recruitment of lymphoid progenitors to the thymus is essential to sustain T cell production throughout life. Importantly, it also limits T lineage regeneration following bone marrow transplantation, and so contributes to the secondary immunodeficiency that is caused by delayed immune reconstitution. Despite this significance, the mechanisms that control thymus colonization are poorly understood. In this study, we show that in both the steady-state and after bone marrow transplant, lymphotoxin β receptor (LTβR) controls entry of T cell progenitors to the thymus. We show that this requirement maps to thymic stroma, further underlining the key importance of this TNFR superfamily member in regulation of thymic microenvironments. Importantly, analysis of the requirement for LTβR in relationship to known regulators of thymus seeding suggests that it acts independently of its regulation of thymus-homing chemokines. Rather, we show that LTβR differentially regulates intrathymic expression of adhesion molecules known to play a role in T cell progenitor entry to the thymus. Finally, Ab-mediated in vivo LTβR stimulation following bone marrow transplant enhances initial thymus recovery and boosts donor-derived T cell numbers, which correlates with increased adhesion molecule expression by thymic stroma. Collectively, we reveal a novel link between LTβR and thymic stromal cells in thymus colonization, and highlight its potential as an immunotherapeutic target to boost T cell reconstitution after transplantation.
Collapse
Affiliation(s)
- Beth Lucas
- Medical Research Council Centre for Immune Regulation, Institute for Immunology and Immunotherapy, University of Birmingham, Birmingham B15 2TT, United Kingdom
| | - Kieran D James
- Medical Research Council Centre for Immune Regulation, Institute for Immunology and Immunotherapy, University of Birmingham, Birmingham B15 2TT, United Kingdom
| | - Emilie J Cosway
- Medical Research Council Centre for Immune Regulation, Institute for Immunology and Immunotherapy, University of Birmingham, Birmingham B15 2TT, United Kingdom
| | - Sonia M Parnell
- Medical Research Council Centre for Immune Regulation, Institute for Immunology and Immunotherapy, University of Birmingham, Birmingham B15 2TT, United Kingdom
| | | | - Carl F Ware
- Infectious and Inflammatory Diseases Research Center, Sanford Burnham Medical Research Institute, La Jolla, CA 92037
| | - William E Jenkinson
- Medical Research Council Centre for Immune Regulation, Institute for Immunology and Immunotherapy, University of Birmingham, Birmingham B15 2TT, United Kingdom
| | - Graham Anderson
- Medical Research Council Centre for Immune Regulation, Institute for Immunology and Immunotherapy, University of Birmingham, Birmingham B15 2TT, United Kingdom;
| |
Collapse
|
128
|
Riemke P, Czeh M, Fischer J, Walter C, Ghani S, Zepper M, Agelopoulos K, Lettermann S, Gebhardt ML, Mah N, Weilemann A, Grau M, Gröning V, Haferlach T, Lenze D, Delwel R, Prinz M, Andrade-Navarro MA, Lenz G, Dugas M, Müller-Tidow C, Rosenbauer F. Myeloid leukemia with transdifferentiation plasticity developing from T-cell progenitors. EMBO J 2016; 35:2399-2416. [PMID: 27572462 DOI: 10.15252/embj.201693927] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2016] [Revised: 07/25/2016] [Accepted: 07/27/2016] [Indexed: 11/09/2022] Open
Abstract
Unfavorable patient survival coincides with lineage plasticity observed in human acute leukemias. These cases are assumed to arise from hematopoietic stem cells, which have stable multipotent differentiation potential. However, here we report that plasticity in leukemia can result from instable lineage identity states inherited from differentiating progenitor cells. Using mice with enhanced c-Myc expression, we show, at the single-cell level, that T-lymphoid progenitors retain broad malignant lineage potential with a high capacity to differentiate into myeloid leukemia. These T-cell-derived myeloid blasts retain expression of a defined set of T-cell transcription factors, creating a lymphoid epigenetic memory that confers growth and propagates myeloid/T-lymphoid plasticity. Based on these characteristics, we identified a correlating human leukemia cohort and revealed targeting of Jak2/Stat3 signaling as a therapeutic possibility. Collectively, our study suggests the thymus as a source for myeloid leukemia and proposes leukemic plasticity as a driving mechanism. Moreover, our results reveal a pathway-directed therapy option against thymus-derived myeloid leukemogenesis and propose a model in which dynamic progenitor differentiation states shape unique neoplastic identities and therapy responses.
Collapse
Affiliation(s)
- Pia Riemke
- Institute of Molecular Tumor Biology, University of Münster, Münster, Germany
| | - Melinda Czeh
- Institute of Molecular Tumor Biology, University of Münster, Münster, Germany
| | - Josephine Fischer
- Institute of Molecular Tumor Biology, University of Münster, Münster, Germany
| | - Carolin Walter
- Institute of Medical Informatics, University of Münster, Münster, Germany
| | - Saeed Ghani
- Department of Hematology, Oncology, and Tumor Immunology, Robert-Rössle-Clinic, Berlin, Germany
| | - Matthias Zepper
- Institute of Molecular Tumor Biology, University of Münster, Münster, Germany
| | - Konstantin Agelopoulos
- Department of Dermatology, Competence Center Chronic Pruritus University of Münster, Münster, Germany
| | - Stephanie Lettermann
- Molecular Hematology and Oncology, Medical Clinics A, University of Münster, Münster, Germany
| | - Marie L Gebhardt
- Department of Computational Biology and Data Mining, Max-Delbrück-Center for Molecular Medicine, Berlin, Germany
| | - Nancy Mah
- Berlin-Brandenburger Center for Regenerative Therapies, Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Andre Weilemann
- Translational Oncology, Medical Clinics A, University of Münster, Münster, Germany.,Cluster of Excellence EXC 1003, Cells in Motion, Münster, Germany
| | - Michael Grau
- Translational Oncology, Medical Clinics A, University of Münster, Münster, Germany.,Cluster of Excellence EXC 1003, Cells in Motion, Münster, Germany
| | - Verena Gröning
- Institute of Molecular Tumor Biology, University of Münster, Münster, Germany
| | | | - Dido Lenze
- Institute of Pathology, Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Ruud Delwel
- Department of Hematology, Erasmus University Medical Center, Rotterdam, the Netherlands
| | - Marco Prinz
- Institute of Neuropathology, University of Freiburg, Freiburg, Germany.,BIOSS Centre for Biological Signalling Studies, University of Freiburg, Freiburg, Germany
| | - Miguel A Andrade-Navarro
- Department of Medical Informatics and Biomathematics, Institute of Molecular Biology Johannes Gutenberg University of Mainz, Mainz, Germany
| | - Georg Lenz
- Translational Oncology, Medical Clinics A, University of Münster, Münster, Germany.,Cluster of Excellence EXC 1003, Cells in Motion, Münster, Germany
| | - Martin Dugas
- Institute of Medical Informatics, University of Münster, Münster, Germany
| | - Carsten Müller-Tidow
- Department of Internal Medicine, Hematology and Oncology, University of Halle-Wittenberg, Halle, Germany
| | - Frank Rosenbauer
- Institute of Molecular Tumor Biology, University of Münster, Münster, Germany
| |
Collapse
|
129
|
Abstract
Genetics tools, and especially the ability to enforce, by transgenesis, or disrupt, by homologous recombination, gene expression in a cell-specific manner, have revolutionized the study of immunology and propelled the laboratory mouse as the main model to study immune responses. Perhaps more than any other aspect of immunology, the study of T cell development has benefited from these technologies. This brief chapter summarizes genetic tools specific to T cell development studies, focusing on mouse strains with lineage- and stage-specific expression of the Cre recombinase, or expressing unique antigen receptor specificities. It ends with a broader discussion of strategies to enforce ectopic lineage and stage-specific gene expression.
Collapse
Affiliation(s)
- Thomas Ciucci
- Laboratory of Immune Cell Biology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | | | | |
Collapse
|
130
|
Höfer T, Busch K, Klapproth K, Rodewald HR. Fate Mapping and Quantitation of Hematopoiesis In Vivo. Annu Rev Immunol 2016; 34:449-78. [DOI: 10.1146/annurev-immunol-032414-112019] [Citation(s) in RCA: 51] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Affiliation(s)
- Thomas Höfer
- Division of Theoretical Systems Biology, German Cancer Research Center (DKFZ), D-69120 Heidelberg, Germany;
| | - Katrin Busch
- Division of Cellular Immunology, German Cancer Research Center (DKFZ), D-69120 Heidelberg, Germany;
| | - Kay Klapproth
- Division of Cellular Immunology, German Cancer Research Center (DKFZ), D-69120 Heidelberg, Germany;
| | - Hans-Reimer Rodewald
- Division of Cellular Immunology, German Cancer Research Center (DKFZ), D-69120 Heidelberg, Germany;
| |
Collapse
|
131
|
Chea S, Perchet T, Petit M, Verrier T, Guy-Grand D, Banchi EG, Vosshenrich CAJ, Di Santo JP, Cumano A, Golub R. Notch signaling in group 3 innate lymphoid cells modulates their plasticity. Sci Signal 2016; 9:ra45. [DOI: 10.1126/scisignal.aaf2223] [Citation(s) in RCA: 43] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
|
132
|
Zook EC, Ramirez K, Guo X, van der Voort G, Sigvardsson M, Svensson EC, Fu YX, Kee BL. The ETS1 transcription factor is required for the development and cytokine-induced expansion of ILC2. J Exp Med 2016; 213:687-96. [PMID: 27069114 PMCID: PMC4854726 DOI: 10.1084/jem.20150851] [Citation(s) in RCA: 63] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2015] [Accepted: 02/29/2016] [Indexed: 12/02/2022] Open
Abstract
Zook et al. use a novel mouse model to demonstrate a requirement for the transcription factor ETS1 in the development and function of group 2 innate lymphoid cells. Group 2 innate lymphoid cells (ILC2s) are a subset of ILCs that play a protective role in the response to helminth infection, but they also contribute to allergic lung inflammation. Here, we report that the deletion of the ETS1 transcription factor in lymphoid cells resulted in a loss of ILC2s in the bone marrow and lymph nodes and that ETS1 promotes the fitness of the common progenitor of all ILCs. ETS1-deficient ILC2 progenitors failed to up-regulate messenger RNA for the E protein transcription factor inhibitor ID2, a critical factor for ILCs, and these cells were unable to expand in cytokine-driven in vitro cultures. In vivo, ETS1 was required for the IL-33–induced accumulation of lung ILC2s and for the production of the T helper type 2 cytokines IL-5 and IL-13. IL-25 also failed to elicit an expansion of inflammatory ILC2s when these cells lacked ETS1. Our data reveal ETS1 as a critical regulator of ILC2 expansion and cytokine production and implicate ETS1 in the regulation of Id2 at the inception of ILC2 development.
Collapse
Affiliation(s)
- Erin C Zook
- Committee on Immunology, The University of Chicago, Chicago, IL 60637
| | - Kevin Ramirez
- Committee on Immunology, The University of Chicago, Chicago, IL 60637
| | - Xiaohuan Guo
- Committee on Immunology, The University of Chicago, Chicago, IL 60637
| | | | - Mikael Sigvardsson
- Experimental Hematopoiesis Unit, Department of Clinical and Experimental Medicine, Faculty of Medicine for Health Sciences, Linköping University, 58183 Linköping, Sweden
| | - Eric C Svensson
- Department of Medicine, Section of Cardiology, The University of Chicago, Chicago, IL 60637
| | - Yang-Xin Fu
- Committee on Immunology, The University of Chicago, Chicago, IL 60637 Department of Pathology, The University of Chicago, Chicago, IL 60637
| | - Barbara L Kee
- Committee on Immunology, The University of Chicago, Chicago, IL 60637 Department of Pathology, The University of Chicago, Chicago, IL 60637
| |
Collapse
|
133
|
Arginase 1 is an innate lymphoid-cell-intrinsic metabolic checkpoint controlling type 2 inflammation. Nat Immunol 2016; 17:656-65. [PMID: 27043409 PMCID: PMC4873382 DOI: 10.1038/ni.3421] [Citation(s) in RCA: 205] [Impact Index Per Article: 22.8] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2015] [Accepted: 02/24/2016] [Indexed: 12/13/2022]
Abstract
Group 2 innate lymphoid cells (ILC2s) regulate tissue inflammation and repair following activation by cell-extrinsic factors including host-derived cytokines. However, the cell-intrinsic metabolic pathways that control ILC2 function are undefined. Here we demonstrate that expression of the enzyme Arginase 1 (Arg1) is a conserved trait of murine and human ILC2s during acute or chronic lung inflammation. Deletion of murine ILC-intrinsic Arg1 abrogated type 2 lung inflammation by restraining ILC2 proliferation and dampening cytokine production. Mechanistically, inhibition of Arg1 enzymatic activity disrupted multiple components of ILC2 metabolic programming by altering arginine catabolism, impairing polyamine biosynthesis and reducing aerobic glycolysis. These data identify Arg1 as a key regulator of ILC2 bioenergetics, controlling proliferative capacity and pro-inflammatory functions that promote type 2 inflammation.
Collapse
|
134
|
Humblet-Baron S, Franckaert D, Dooley J, Bornschein S, Cauwe B, Schönefeldt S, Bossuyt X, Matthys P, Baron F, Wouters C, Liston A. IL-2 consumption by highly activated CD8 T cells induces regulatory T-cell dysfunction in patients with hemophagocytic lymphohistiocytosis. J Allergy Clin Immunol 2016; 138:200-209.e8. [PMID: 26947179 DOI: 10.1016/j.jaci.2015.12.1314] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2015] [Revised: 11/25/2015] [Accepted: 12/14/2015] [Indexed: 12/15/2022]
Abstract
BACKGROUND Hemophagocytic lymphohistiocytosis (HLH) is a severe inflammatory condition driven by excessive CD8(+) T-cell activation. HLH occurs as both acquired and familial hemophagocytic lymphohistiocytosis (FHL) forms. In both conditions, a sterile or infectious trigger is required for disease initiation, which then becomes self-sustaining and life-threatening. Recent studies have attributed the key distal event to excessive IFN-γ production; however, the proximal events driving immune dysregulation have remained undefined. OBJECTIVE We sought to investigate the role of regulatory T (Treg) cells in the pathophysiology of experimental FHL. METHODS Because mutation in perforin is a common cause of FHL, we used an experimental FHL mouse model in which disease in perforin-deficient mice is triggered by lymphocytic choriomeningitis virus (LCMV). We assessed Treg and CD8(+) T-cell homeostasis and activation during the changing systemic conditions in the mice. In addition, human blood samples were collected and analyzed during the HLH episode. RESULTS We found no primary Treg cell defects in perforin-deficient mice. However, Treg cell numbers collapsed after LCMV inoculation. The collapse of Treg cell numbers in LCMV-triggered perforin-deficient, but not wild-type, mice was accompanied by the combination of lower IL-2 secretion by conventional CD4(+) T cells, increased IL-2 consumption by activated CD8(+) T cells, and secretion of competitive soluble CD25. Moreover low Treg cell numbers were observed in untreated patients experiencing HLH flares. CONCLUSION These results demonstrate that excessive CD8(+) T-cell activation rewires the IL-2 homeostatic network away from Treg cell maintenance and toward feed-forward inflammation. These results also provide a potential mechanistic pathway for the progression of infectious inflammation to persistent inflammation in patients with HLH.
Collapse
Affiliation(s)
- Stéphanie Humblet-Baron
- Translational Immunology Laboratory, VIB, Leuven, Belgium; Department of Microbiology and Immunology, KUL-University of Leuven, Leuven, Belgium
| | - Dean Franckaert
- Translational Immunology Laboratory, VIB, Leuven, Belgium; Department of Microbiology and Immunology, KUL-University of Leuven, Leuven, Belgium
| | - James Dooley
- Translational Immunology Laboratory, VIB, Leuven, Belgium; Department of Microbiology and Immunology, KUL-University of Leuven, Leuven, Belgium
| | - Simon Bornschein
- Translational Immunology Laboratory, VIB, Leuven, Belgium; Department of Microbiology and Immunology, KUL-University of Leuven, Leuven, Belgium
| | - Bénédicte Cauwe
- Translational Immunology Laboratory, VIB, Leuven, Belgium; Department of Microbiology and Immunology, KUL-University of Leuven, Leuven, Belgium
| | - Susann Schönefeldt
- Translational Immunology Laboratory, VIB, Leuven, Belgium; Department of Microbiology and Immunology, KUL-University of Leuven, Leuven, Belgium
| | - Xavier Bossuyt
- Department of Microbiology and Immunology, KUL-University of Leuven, Leuven, Belgium; University Hospitals Leuven, Leuven, Belgium
| | | | - Frédéric Baron
- GIGA I(3) and the Department of Hematology, University of Liège, Liege, Belgium
| | - Carine Wouters
- University Hospitals Leuven, Leuven, Belgium; Pediatric Immunology, KUL-University of Leuven, Leuven, Belgium
| | - Adrian Liston
- Translational Immunology Laboratory, VIB, Leuven, Belgium; Department of Microbiology and Immunology, KUL-University of Leuven, Leuven, Belgium.
| |
Collapse
|
135
|
Hanafusa T, Kato K, Azukizawa H, Miyazaki JI, Takeda J, Katayama I. B-1 B cell progenitors transiently and partially express keratin 5 during differentiation in bone marrow. J Dermatol Sci 2016; 81:173-81. [DOI: 10.1016/j.jdermsci.2015.11.010] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2015] [Revised: 10/14/2015] [Accepted: 11/17/2015] [Indexed: 01/16/2023]
|
136
|
Jensen CT, Strid T, Sigvardsson M. Exploring the multifaceted nature of the common lymphoid progenitor compartment. Curr Opin Immunol 2016; 39:121-6. [PMID: 26871596 DOI: 10.1016/j.coi.2016.01.009] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2015] [Revised: 01/13/2016] [Accepted: 01/18/2016] [Indexed: 01/03/2023]
Abstract
While the common lymphoid progenitor compartment was originally thought to be a rather homogenous cell population, it has become increasingly clear that this compartment is highly heterogeneous both with regard to phenotypic and functional features. The exploration of this cellular complexity has generated novel molecular insights into regulatory events in lymphoid lineage restriction and provided support for the idea that multiple lineage restriction events occur at this developmental stage. Furthermore, the identification of multiple lineage-restricted progenitors with mixed lineage potential challenges a strictly hierarchical model for lymphoid development. Instead we propose a model based on competence windows during which cell fates are established through the action of lineage determining factors.
Collapse
Affiliation(s)
| | - Tobias Strid
- Department of Clinical and Experimental Medicine, Linköping University, Sweden
| | - Mikael Sigvardsson
- Division of Molecular Hematology, Lund University, Sweden; Department of Clinical and Experimental Medicine, Linköping University, Sweden.
| |
Collapse
|
137
|
Chea S, Schmutz S, Berthault C, Perchet T, Petit M, Burlen-Defranoux O, Goldrath A, Rodewald HR, Cumano A, Golub R. Single-Cell Gene Expression Analyses Reveal Heterogeneous Responsiveness of Fetal Innate Lymphoid Progenitors to Notch Signaling. Cell Rep 2016; 14:1500-1516. [PMID: 26832410 DOI: 10.1016/j.celrep.2016.01.015] [Citation(s) in RCA: 63] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2015] [Revised: 12/01/2015] [Accepted: 01/02/2016] [Indexed: 11/27/2022] Open
Abstract
T and innate lymphoid cells (ILCs) share some aspects of their developmental programs. However, although Notch signaling is strictly required for T cell development, it is dispensable for fetal ILC development. Constitutive activation of Notch signaling, at the common lymphoid progenitor stage, drives T cell development and abrogates ILC development by preventing Id2 expression. By combining single-cell transcriptomics and clonal culture strategies, we characterize two heterogeneous α4β7-expressing lymphoid progenitor compartments. αLP1 (Flt3(+)) still retains T cell potential and comprises the global ILC progenitor, while αLP2 (Flt3(-)) consists of ILC precursors that are primed toward the different ILC lineages. Only a subset of αLP2 precursors is sensitive to Notch signaling required for their proliferation. Our study identifies, in a refined manner, the diversity of transitional stages of ILC development, their transcriptional signatures, and their differential dependence on Notch signaling.
Collapse
|
138
|
Staal FJT, Wiekmeijer AS, Brugman MH, Pike-Overzet K. The functional relationship between hematopoietic stem cells and developing T lymphocytes. Ann N Y Acad Sci 2016; 1370:36-44. [PMID: 26773328 DOI: 10.1111/nyas.12995] [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] [Indexed: 12/18/2022]
Abstract
In contrast to all other blood and immune cells, T lymphocytes do not develop in the bone marrow (BM), but in the specialized microenvironment provided by the thymus. Similar to the other lineages, however, all T cells arise from multipotent hematopoietic stem cells (HSCs) that reside in the BM. Not all HSCs give rise to T cells; but how many and what kind of developmental checkpoints are located along this intricate differentiation path is the subject of intense research. Traditionally, this process has been studied almost exclusively using mouse cells, but recent advances in immunodeficient mouse models, high-speed cell sorting, lentiviral transduction protocols, and deep sequencing techniques have allowed these questions to be addressed using human cells. Here we review the process of thymic seeding by BM-derived cells and T cell commitment in humans, discussing recent insights into the clonal composition of the thymus and the definition of developmental checkpoints, on the basis of insights from human severe combined immunodeficiency patients.
Collapse
Affiliation(s)
- Frank J T Staal
- Department of Immunohematology and Blood Transfusion, Leiden University Medical Center, Leiden, the Netherlands
| | - Anna-Sophia Wiekmeijer
- Department of Immunohematology and Blood Transfusion, Leiden University Medical Center, Leiden, the Netherlands
| | - Martijn H Brugman
- Department of Immunohematology and Blood Transfusion, Leiden University Medical Center, Leiden, the Netherlands
| | - Karin Pike-Overzet
- Department of Immunohematology and Blood Transfusion, Leiden University Medical Center, Leiden, the Netherlands
| |
Collapse
|
139
|
Halim TYF, Hwang YY, Scanlon ST, Zaghouani H, Garbi N, Fallon PG, McKenzie ANJ. Group 2 innate lymphoid cells license dendritic cells to potentiate memory TH2 cell responses. Nat Immunol 2016; 17:57-64. [PMID: 26523868 PMCID: PMC4685755 DOI: 10.1038/ni.3294] [Citation(s) in RCA: 250] [Impact Index Per Article: 27.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2015] [Accepted: 09/14/2015] [Indexed: 12/13/2022]
Abstract
Rapid activation of memory CD4(+) T helper 2 (TH2) cells during allergic inflammation requires their recruitment into the affected tissue. Here we demonstrate that group 2 innate lymphoid (ILC2) cells have a crucial role in memory TH2 cell responses, with targeted depletion of ILC2 cells profoundly impairing TH2 cell localization to the lungs and skin of sensitized mice after allergen re-challenge. ILC2-derived interleukin 13 (IL-13) is critical for eliciting production of the TH2 cell-attracting chemokine CCL17 by IRF4(+)CD11b(+)CD103(-) dendritic cells (DCs). Consequently, the sentinel function of DCs is contingent on ILC2 cells for the generation of an efficient memory TH2 cell response. These results elucidate a key innate mechanism in the regulation of the immune memory response to allergens.
Collapse
Affiliation(s)
- Timotheus YF Halim
- Medical Research Council, Laboratory of Molecular Biology, Cambridge, Cambridgeshire, CB2 0QH, United Kingdom
| | - You Yi Hwang
- Medical Research Council, Laboratory of Molecular Biology, Cambridge, Cambridgeshire, CB2 0QH, United Kingdom
| | - Seth T Scanlon
- Medical Research Council, Laboratory of Molecular Biology, Cambridge, Cambridgeshire, CB2 0QH, United Kingdom
| | - Habib Zaghouani
- Department of Molecular Microbiology and Immunology, University of Missouri School of Medicine, Columbia, Missouri, United States of America
| | - Natalio Garbi
- Department of Molecular Immunology, Institutes of Molecular Medicine and Experimental Immunology, University of Bonn, Bonn D-53105, Germany
| | - Padraic G Fallon
- Trinity Biomedical Sciences Institute, Trinity College Dublin, Ireland
- National Children’s Research Centre, Our Lady’s Children’s Hospital, Dublin, Ireland
- Institute of Molecular Medicine, Trinity College Dublin, Dublin 2, Ireland
| | - Andrew NJ McKenzie
- Medical Research Council, Laboratory of Molecular Biology, Cambridge, Cambridgeshire, CB2 0QH, United Kingdom
| |
Collapse
|
140
|
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: 325] [Impact Index Per Article: 32.5] [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
|
141
|
Hams E, Bermingham R, Wurlod FA, Hogan AE, O'Shea D, Preston RJ, Rodewald HR, McKenzie ANJ, Fallon PG. The helminth T2 RNase ω1 promotes metabolic homeostasis in an IL-33- and group 2 innate lymphoid cell-dependent mechanism. FASEB J 2015; 30:824-35. [PMID: 26490658 PMCID: PMC4973506 DOI: 10.1096/fj.15-277822] [Citation(s) in RCA: 67] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2015] [Accepted: 10/13/2015] [Indexed: 12/17/2022]
Abstract
Induction of a type 2 cellular response in the white adipose tissue leads to weight loss and improves glucose homeostasis in obese animals. Injection of obese mice with recombinant helminth-derived Schistosoma mansoni egg-derived ω1 (ω1), a potent inducer of type 2 activation, improves metabolic status involving a mechanism reliant upon release of the type 2 initiator cytokine IL-33. IL-33 initiates the accumulation of group 2 innate lymphoid cells (ILC2s), eosinophils, and alternatively activated macrophages in the adipose tissue. IL-33 release from cells in the adipose tissue is mediated by the RNase activity of ω1; however, the ability of ω1 to improve metabolic status is reliant upon effective binding of ω1 to CD206. We demonstrate a novel mechanism for RNase-mediated release of IL-33 inducing ILC2-dependent improvements in the metabolic status of obese animals.
Collapse
Affiliation(s)
- Emily Hams
- *Trinity Biomedical Sciences Institute, School of Medicine, Trinity College Dublin, Dublin, Ireland; National Children's Research Centre, Our Lady's Children's Hospital, Dublin, Ireland; Obesity Immunology, Education and Research Centre, St. Vincent's University Hospital, University College Dublin, Dublin, Ireland; Department of Endocrinology, St. Vincent's University Hospital, Elm Park, Dublin, Ireland; Division of Cellular Immunology, German Cancer Research Center, Heidelberg, Germany; and Medical Research Council Laboratory of Molecular Biology, Cambridge, United Kingdom
| | - Rachel Bermingham
- *Trinity Biomedical Sciences Institute, School of Medicine, Trinity College Dublin, Dublin, Ireland; National Children's Research Centre, Our Lady's Children's Hospital, Dublin, Ireland; Obesity Immunology, Education and Research Centre, St. Vincent's University Hospital, University College Dublin, Dublin, Ireland; Department of Endocrinology, St. Vincent's University Hospital, Elm Park, Dublin, Ireland; Division of Cellular Immunology, German Cancer Research Center, Heidelberg, Germany; and Medical Research Council Laboratory of Molecular Biology, Cambridge, United Kingdom
| | - Felicity A Wurlod
- *Trinity Biomedical Sciences Institute, School of Medicine, Trinity College Dublin, Dublin, Ireland; National Children's Research Centre, Our Lady's Children's Hospital, Dublin, Ireland; Obesity Immunology, Education and Research Centre, St. Vincent's University Hospital, University College Dublin, Dublin, Ireland; Department of Endocrinology, St. Vincent's University Hospital, Elm Park, Dublin, Ireland; Division of Cellular Immunology, German Cancer Research Center, Heidelberg, Germany; and Medical Research Council Laboratory of Molecular Biology, Cambridge, United Kingdom
| | - Andrew E Hogan
- *Trinity Biomedical Sciences Institute, School of Medicine, Trinity College Dublin, Dublin, Ireland; National Children's Research Centre, Our Lady's Children's Hospital, Dublin, Ireland; Obesity Immunology, Education and Research Centre, St. Vincent's University Hospital, University College Dublin, Dublin, Ireland; Department of Endocrinology, St. Vincent's University Hospital, Elm Park, Dublin, Ireland; Division of Cellular Immunology, German Cancer Research Center, Heidelberg, Germany; and Medical Research Council Laboratory of Molecular Biology, Cambridge, United Kingdom
| | - Donal O'Shea
- *Trinity Biomedical Sciences Institute, School of Medicine, Trinity College Dublin, Dublin, Ireland; National Children's Research Centre, Our Lady's Children's Hospital, Dublin, Ireland; Obesity Immunology, Education and Research Centre, St. Vincent's University Hospital, University College Dublin, Dublin, Ireland; Department of Endocrinology, St. Vincent's University Hospital, Elm Park, Dublin, Ireland; Division of Cellular Immunology, German Cancer Research Center, Heidelberg, Germany; and Medical Research Council Laboratory of Molecular Biology, Cambridge, United Kingdom
| | - Roger J Preston
- *Trinity Biomedical Sciences Institute, School of Medicine, Trinity College Dublin, Dublin, Ireland; National Children's Research Centre, Our Lady's Children's Hospital, Dublin, Ireland; Obesity Immunology, Education and Research Centre, St. Vincent's University Hospital, University College Dublin, Dublin, Ireland; Department of Endocrinology, St. Vincent's University Hospital, Elm Park, Dublin, Ireland; Division of Cellular Immunology, German Cancer Research Center, Heidelberg, Germany; and Medical Research Council Laboratory of Molecular Biology, Cambridge, United Kingdom
| | - Hans-Reimer Rodewald
- *Trinity Biomedical Sciences Institute, School of Medicine, Trinity College Dublin, Dublin, Ireland; National Children's Research Centre, Our Lady's Children's Hospital, Dublin, Ireland; Obesity Immunology, Education and Research Centre, St. Vincent's University Hospital, University College Dublin, Dublin, Ireland; Department of Endocrinology, St. Vincent's University Hospital, Elm Park, Dublin, Ireland; Division of Cellular Immunology, German Cancer Research Center, Heidelberg, Germany; and Medical Research Council Laboratory of Molecular Biology, Cambridge, United Kingdom
| | - Andrew N J McKenzie
- *Trinity Biomedical Sciences Institute, School of Medicine, Trinity College Dublin, Dublin, Ireland; National Children's Research Centre, Our Lady's Children's Hospital, Dublin, Ireland; Obesity Immunology, Education and Research Centre, St. Vincent's University Hospital, University College Dublin, Dublin, Ireland; Department of Endocrinology, St. Vincent's University Hospital, Elm Park, Dublin, Ireland; Division of Cellular Immunology, German Cancer Research Center, Heidelberg, Germany; and Medical Research Council Laboratory of Molecular Biology, Cambridge, United Kingdom
| | - Padraic G Fallon
- *Trinity Biomedical Sciences Institute, School of Medicine, Trinity College Dublin, Dublin, Ireland; National Children's Research Centre, Our Lady's Children's Hospital, Dublin, Ireland; Obesity Immunology, Education and Research Centre, St. Vincent's University Hospital, University College Dublin, Dublin, Ireland; Department of Endocrinology, St. Vincent's University Hospital, Elm Park, Dublin, Ireland; Division of Cellular Immunology, German Cancer Research Center, Heidelberg, Germany; and Medical Research Council Laboratory of Molecular Biology, Cambridge, United Kingdom
| |
Collapse
|
142
|
Kubicka-Sierszen A, Grzegorczyk JŁ. The influence of infectious factors on dendritic cell apoptosis. Arch Med Sci 2015; 11:1044-51. [PMID: 26528349 PMCID: PMC4624750 DOI: 10.5114/aoms.2015.54860] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/08/2013] [Revised: 06/04/2013] [Accepted: 10/02/2013] [Indexed: 12/23/2022] Open
Abstract
Pathogens can have a negative influence on dendritic cells (DCs), causing their apoptosis, which prevents active presentation of foreign antigens. It results in a state of immunosuppression which makes the body susceptible to secondary infections. Infected immature DCs have lower expression of co-stimulatory and adhesion molecules, reduced ability to secrete cytokines and an inhibited maturation process and are incapable of effective antigen presentation and activation of T-lymphocytes. In some cases, the ability of DCs to undergo rapid apoptosis is important for the body defense, which is probably because of DCs' ability to cross-present and cooperate with other cells. Apoptotic bodies released from the infected DCs are phagocytosed by other DCs, which then stimulate the effector cells and present antigens more efficiently than infected cells. The aim of this article is to review how the DCs respond to viral and bacterial factors and which biochemical mechanisms are responsible for their apoptosis.
Collapse
Affiliation(s)
- Agata Kubicka-Sierszen
- Department of Microbiology and Laboratory Medical Immunology, Faculty of Medicine, Medical University of Lodz, Lodz, Poland
| | - Janina Ł Grzegorczyk
- Department of Microbiology and Laboratory Medical Immunology, Faculty of Medicine, Medical University of Lodz, Lodz, Poland
| |
Collapse
|
143
|
Yang Q, Li F, Harly C, Xing S, Ye L, Xia X, Wang H, Wang X, Yu S, Zhou X, Cam M, Xue HH, Bhandoola A. TCF-1 upregulation identifies early innate lymphoid progenitors in the bone marrow. Nat Immunol 2015; 16:1044-50. [PMID: 26280998 PMCID: PMC4575643 DOI: 10.1038/ni.3248] [Citation(s) in RCA: 219] [Impact Index Per Article: 21.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2015] [Accepted: 07/15/2015] [Indexed: 02/07/2023]
Abstract
The cellular and molecular events that drive the early development of innate lymphoid cells (ILCs) remain poorly understood. We show that the transcription factor TCF-1 is required for the efficient generation of all known adult ILC subsets and their precursors. Using novel reporter mice, we identified a new subset of early ILC progenitors (EILPs) expressing high amounts of TCF-1. EILPs lacked efficient T and B lymphocyte potential but efficiently gave rise to NK cells and all known adult helper ILC lineages, indicating that they are the earliest ILC-committed progenitors identified so far. Our results suggest that upregulation of TCF-1 expression denotes the earliest stage of ILC fate specification. The discovery of EILPs provides a basis for deciphering additional signals that specify ILC fate.
Collapse
Affiliation(s)
- Qi Yang
- T-Cell Biology and Development Unit, Laboratory of Genome Integrity, Center for Cancer Research, National Cancer Institute, Bethesda, Maryland, USA
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Fengyin Li
- Department of Microbiology, Carver College of Medicine, University of Iowa, Iowa City, Iowa, USA
| | - Christelle Harly
- T-Cell Biology and Development Unit, Laboratory of Genome Integrity, Center for Cancer Research, National Cancer Institute, Bethesda, Maryland, USA
| | - Shaojun Xing
- Department of Microbiology, Carver College of Medicine, University of Iowa, Iowa City, Iowa, USA
| | - Longyun Ye
- T-Cell Biology and Development Unit, Laboratory of Genome Integrity, Center for Cancer Research, National Cancer Institute, Bethesda, Maryland, USA
| | - Xuefeng Xia
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Haikun Wang
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Xinxin Wang
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Shuyang Yu
- Department of Microbiology, Carver College of Medicine, University of Iowa, Iowa City, Iowa, USA
| | - Xinyuan Zhou
- Department of Microbiology, Carver College of Medicine, University of Iowa, Iowa City, Iowa, USA
| | - Maggie Cam
- CCR Collaborative Bioinformatics Resource, National Cancer Institute, Bethesda, Maryland, USA
| | - Hai-Hui Xue
- Department of Microbiology, Carver College of Medicine, University of Iowa, Iowa City, Iowa, USA
| | - Avinash Bhandoola
- T-Cell Biology and Development Unit, Laboratory of Genome Integrity, Center for Cancer Research, National Cancer Institute, Bethesda, Maryland, USA
| |
Collapse
|
144
|
Ziętara N, Łyszkiewicz M, Puchałka J, Witzlau K, Reinhardt A, Förster R, Pabst O, Prinz I, Krueger A. Multicongenic fate mapping quantification of dynamics of thymus colonization. ACTA ACUST UNITED AC 2015; 212:1589-601. [PMID: 26347471 PMCID: PMC4577840 DOI: 10.1084/jem.20142143] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2014] [Accepted: 08/10/2015] [Indexed: 12/30/2022]
Abstract
Ziętara et al demonstrate with multicongenic fate mapping that thymus seeding is directly restricted to the duration of niche occupancy rather than long-range effects. Postnatal T cell development depends on continuous colonization of the thymus by BM-derived T lineage progenitors. Both quantitative parameters and the mechanisms of thymus seeding remain poorly understood. Here, we determined the number of dedicated thymus-seeding progenitor niches (TSPNs) capable of supporting productive T cell development, turnover rates of niche occupancy, and feedback mechanisms. To this end, we established multicongenic fate mapping combined with mathematical modeling to quantitate individual events of thymus colonization. We applied this method to study thymus colonization in CCR7−/−CCR9−/− (DKO) mice, whose TSPNs are largely unoccupied. We showed that ∼160–200 TSPNs are present in the adult thymus and, on average, 10 of these TSPNs were open for recolonization at steady state. Preconditioning of wild-type mice revealed a similar number of TSPNs, indicating that preconditioning can generate space efficiently for transplanted T cell progenitors. To identify potential cellular feedback loops restricting thymus colonization, we performed serial transfer experiments. These experiments indicated that thymus seeding was directly restricted by the duration of niche occupancy rather than long-range effects, thus challenging current paradigms of thymus colonization.
Collapse
Affiliation(s)
- Natalia Ziętara
- Institute of Immunology, Hannover Medical School, D-30625 Hannover, Germany
| | - Marcin Łyszkiewicz
- Institute of Immunology, Hannover Medical School, D-30625 Hannover, Germany
| | - Jacek Puchałka
- Dr. von Haunersches Kinderspital, University Children's Hospital, Ludwig Maximilian University, D-80337 Munich, Germany
| | - Katrin Witzlau
- Institute of Immunology, Hannover Medical School, D-30625 Hannover, Germany
| | - Annika Reinhardt
- Institute of Immunology, Hannover Medical School, D-30625 Hannover, Germany
| | - Reinhold Förster
- Institute of Immunology, Hannover Medical School, D-30625 Hannover, Germany
| | - Oliver Pabst
- Institute of Immunology, Hannover Medical School, D-30625 Hannover, Germany Institute of Molecular Medicine, RWTH Aachen University, D-52074 Aachen, Germany
| | - Immo Prinz
- Institute of Immunology, Hannover Medical School, D-30625 Hannover, Germany
| | - Andreas Krueger
- Institute of Immunology, Hannover Medical School, D-30625 Hannover, Germany
| |
Collapse
|
145
|
Hu Z, Lancaster JN, Ehrlich LIR. The Contribution of Chemokines and Migration to the Induction of Central Tolerance in the Thymus. Front Immunol 2015; 6:398. [PMID: 26300884 PMCID: PMC4528182 DOI: 10.3389/fimmu.2015.00398] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2015] [Accepted: 07/20/2015] [Indexed: 02/01/2023] Open
Abstract
As T cells develop, they migrate throughout the thymus where they undergo essential bi-directional signaling with stromal cells in distinct thymic microenvironments. Immature thymocyte progenitors are located in the thymic cortex. Following T cell receptor expression and positive selection, thymocytes undergo a dramatic transition: they become rapidly motile and relocate to the thymic medulla. Antigen-presenting cells (APCs) within the cortex and medulla display peptides derived from a wide array of self-proteins, which promote thymocyte self-tolerance. If a thymocyte is auto-reactive against such antigens, it undergoes either negative selection, via apoptosis, or differentiation into the regulatory T cell lineage. This induction of central tolerance is critical for prevention of autoimmunity. Chemokines and adhesion molecules play an essential role in tolerance induction, as they promote migration of developing thymocytes through the different thymic microenvironments and enhance interactions with APCs displaying self-antigens. Herein, we review the contribution of chemokines and other regulators of thymocyte localization and motility to T cell development, with a focus on their contribution to the induction of central tolerance.
Collapse
Affiliation(s)
- Zicheng Hu
- Ehrlich Laboratory, Department of Molecular Biosciences, Institute for Cellular and Molecular Biology, The University of Texas at Austin , Austin, TX , USA
| | - Jessica Naomi Lancaster
- Ehrlich Laboratory, Department of Molecular Biosciences, Institute for Cellular and Molecular Biology, The University of Texas at Austin , Austin, TX , USA
| | - Lauren I R Ehrlich
- Ehrlich Laboratory, Department of Molecular Biosciences, Institute for Cellular and Molecular Biology, The University of Texas at Austin , Austin, TX , USA
| |
Collapse
|
146
|
Abstract
The thymus is an essential organ for the generation of the adaptive immune system. By now, the cellular selection events taking place in ongoing life before sexual maturity have been worked out even at the molecular level, and thus thymic lymphocyte development represents one of the best-studied systems in mammalian development. Because thymic lymphocyte development involves ample proliferation and generation of new cells, it is not astonishing that the thymus also represents an organ where malignancy can develop. In this Masters of Immunology primer, the development of lymphocytes and the role of intracellular Notch 1 and cyclins in lymphocytic malignancy are reviewed, offering new therapeutic possibilities.
Collapse
Affiliation(s)
- Harald von Boehmer
- Author's Affiliations: Harvard Medical School; Dana-Farber Cancer Institute, Boston, Massachusetts; University of Florida, Gainesville, Florida; and University of Munich, Munich, Germany
| |
Collapse
|
147
|
Becker AM, Callahan DJ, Richner JM, Choi J, DiPersio JF, Diamond MS, Bhattacharya D. GPR18 Controls Reconstitution of Mouse Small Intestine Intraepithelial Lymphocytes following Bone Marrow Transplantation. PLoS One 2015. [PMID: 26197390 PMCID: PMC4510063 DOI: 10.1371/journal.pone.0133854] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022] Open
Abstract
Specific G protein coupled receptors (GPRs) regulate the proper positioning, function, and development of immune lineage subsets. Here, we demonstrate that GPR18 regulates the reconstitution of intraepithelial lymphocytes (IELs) of the small intestine following bone marrow transplantation. Through analysis of transcriptional microarray data, we find that GPR18 is highly expressed in IELs, lymphoid progenitors, and mature follicular B cells. To establish the physiological role of this largely uncharacterized GPR, we generated Gpr18-/- mice. Despite high levels of GPR18 expression in specific hematopoietic progenitors, Gpr18-/- mice have no defects in lymphopoiesis or myelopoiesis. Moreover, antibody responses following immunization with hapten-protein conjugates or infection with West Nile virus are normal in Gpr18-/- mice. Steady-state numbers of IELs are also normal in Gpr18-/- mice. However, competitive bone marrow reconstitution experiments demonstrate that GPR18 is cell-intrinsically required for the optimal restoration of small intestine TCRγδ+ and TCRαβ+ CD8αα+ IELs. In contrast, GPR18 is dispensable for the reconstitution of large intestine IELs. Moreover, Gpr18-/- bone marrow reconstitutes small intestine IELs similarly to controls in athymic recipients. Gpr18-/- chimeras show no changes in susceptibility to intestinal insults such as Citrobacter rodentium infections or graft versus host disease. These data reveal highly specific requirements for GPR18 in the development and reconstitution of thymus-derived intestinal IEL subsets in the steady-state and after bone marrow transplantation.
Collapse
MESH Headings
- Animals
- B-Lymphocytes/cytology
- Bone Marrow Cells/cytology
- Bone Marrow Transplantation
- Citrobacter
- Female
- Graft vs Host Disease
- Hematopoietic Stem Cells/cytology
- Intestinal Mucosa/metabolism
- Intestine, Small/metabolism
- Lymphocytes/cytology
- Male
- Mice
- Mice, Inbred C57BL
- Mice, Transgenic
- Myelopoiesis
- Oligonucleotide Array Sequence Analysis
- Receptors, Antigen, T-Cell, alpha-beta/metabolism
- Receptors, Antigen, T-Cell, gamma-delta/metabolism
- Receptors, G-Protein-Coupled/metabolism
- Thymus Gland/metabolism
- Transplantation, Homologous
- West Nile virus
Collapse
Affiliation(s)
- Amy M. Becker
- Department of Pathology and Immunology, Washington University School of Medicine, Saint Louis, Missouri, United States of America
| | - Derrick J. Callahan
- Department of Pathology and Immunology, Washington University School of Medicine, Saint Louis, Missouri, United States of America
| | - Justin M. Richner
- Department of Medicine, Washington University School of Medicine, Saint Louis, Missouri, United States of America
- Department of Molecular Microbiology, Washington University School of Medicine, Saint Louis, Missouri, United States of America
| | - Jaebok Choi
- Department of Medicine, Washington University School of Medicine, Saint Louis, Missouri, United States of America
- Division of Oncology, Washington University School of Medicine, Saint Louis, Missouri, United States of America
| | - John F. DiPersio
- Department of Medicine, Washington University School of Medicine, Saint Louis, Missouri, United States of America
- Division of Oncology, Washington University School of Medicine, Saint Louis, Missouri, United States of America
| | - Michael S. Diamond
- Department of Pathology and Immunology, Washington University School of Medicine, Saint Louis, Missouri, United States of America
- Department of Medicine, Washington University School of Medicine, Saint Louis, Missouri, United States of America
- Department of Molecular Microbiology, Washington University School of Medicine, Saint Louis, Missouri, United States of America
| | - Deepta Bhattacharya
- Department of Pathology and Immunology, Washington University School of Medicine, Saint Louis, Missouri, United States of America
- * E-mail:
| |
Collapse
|
148
|
Gutiérrez-Martínez E, Planès R, Anselmi G, Reynolds M, Menezes S, Adiko AC, Saveanu L, Guermonprez P. Cross-Presentation of Cell-Associated Antigens by MHC Class I in Dendritic Cell Subsets. Front Immunol 2015; 6:363. [PMID: 26236315 PMCID: PMC4505393 DOI: 10.3389/fimmu.2015.00363] [Citation(s) in RCA: 121] [Impact Index Per Article: 12.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2015] [Accepted: 07/05/2015] [Indexed: 12/12/2022] Open
Abstract
Dendritic cells (DCs) have the unique ability to pick up dead cells carrying antigens in tissue and migrate to the lymph nodes where they can cross-present cell-associated antigens by MHC class I to CD8+ T cells. There is strong in vivo evidence that the mouse XCR1+ DCs subset acts as a key player in this process. The intracellular processes underlying cross-presentation remain controversial and several pathways have been proposed. Indeed, a wide number of studies have addressed the cellular process of cross-presentation in vitro using a variety of sources of antigen and antigen-presenting cells. Here, we review the in vivo and in vitro evidence supporting the current mechanistic models and disscuss their physiological relevance to the cross-presentation of cell-associated antigens by DCs subsets.
Collapse
Affiliation(s)
- Enric Gutiérrez-Martínez
- Laboratory of Phagocyte Immunobiology, Peter Gorer Department of Immunobiology, CMCBI, King's College London , London , UK
| | - Remi Planès
- Laboratory of Phagocyte Immunobiology, Peter Gorer Department of Immunobiology, CMCBI, King's College London , London , UK
| | - Giorgio Anselmi
- Laboratory of Phagocyte Immunobiology, Peter Gorer Department of Immunobiology, CMCBI, King's College London , London , UK
| | - Matthew Reynolds
- Laboratory of Phagocyte Immunobiology, Peter Gorer Department of Immunobiology, CMCBI, King's College London , London , UK
| | - Shinelle Menezes
- Laboratory of Phagocyte Immunobiology, Peter Gorer Department of Immunobiology, CMCBI, King's College London , London , UK
| | - Aimé Cézaire Adiko
- Laboratory of Phagocyte Immunobiology, Peter Gorer Department of Immunobiology, Centre for Molecular & Cellular Biology of Inflammation (CMCBI), King's College London , Paris , France ; Sorbonne Paris Cité, Université Paris Diderot , Paris , France
| | - Loredana Saveanu
- Laboratory of Phagocyte Immunobiology, Peter Gorer Department of Immunobiology, Centre for Molecular & Cellular Biology of Inflammation (CMCBI), King's College London , Paris , France ; Sorbonne Paris Cité, Université Paris Diderot , Paris , France
| | - Pierre Guermonprez
- Laboratory of Phagocyte Immunobiology, Peter Gorer Department of Immunobiology, CMCBI, King's College London , London , UK
| |
Collapse
|
149
|
Oh J, Shin JS. The Role of Dendritic Cells in Central Tolerance. Immune Netw 2015; 15:111-20. [PMID: 26140042 PMCID: PMC4486773 DOI: 10.4110/in.2015.15.3.111] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2015] [Revised: 05/26/2015] [Accepted: 06/02/2015] [Indexed: 12/16/2022] Open
Abstract
Dendritic cells (DCs) play a significant role in establishing self-tolerance through their ability to present self-antigens to developing T cells in the thymus. DCs are predominantly localized in the medullary region of thymus and present a broad range of self-antigens, which include tissue-restricted antigens expressed and transferred from medullary thymic epithelial cells, circulating antigens directly captured by thymic DCs through coticomedullary junction blood vessels, and peripheral tissue antigens captured and transported by peripheral tissue DCs homing to the thymus. When antigen-presenting DCs make a high affinity interaction with antigen-specific thymocytes, this interaction drives the interacting thymocytes to death, a process often referred to as negative selection, which fundamentally blocks the self-reactive thymocytes from differentiating into mature T cells. Alternatively, the interacting thymocytes differentiate into the regulatory T (Treg) cells, a distinct T cell subset with potent immune suppressive activities. The specific mechanisms by which thymic DCs differentiate Treg cells have been proposed by several laboratories. Here, we review the literatures that elucidate the contribution of thymic DCs to negative selection and Treg cell differentiation, and discusses its potential mechanisms and future directions.
Collapse
Affiliation(s)
- Jaehak Oh
- Department of Microbiology and Immunology, Sandler Asthma Basic Research Center, University of California San Francisco, San Francisco, CA 94143, USA
| | - Jeoung-Sook Shin
- Department of Microbiology and Immunology, Sandler Asthma Basic Research Center, University of California San Francisco, San Francisco, CA 94143, USA
| |
Collapse
|
150
|
Sreenivasan J, Schlenner S, Franckaert D, Dooley J, Liston A. The thymoprotective function of leptin is indirectly mediated via suppression of obesity. Immunology 2015; 146:122-9. [PMID: 26059465 DOI: 10.1111/imm.12488] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2015] [Revised: 05/19/2015] [Accepted: 06/01/2015] [Indexed: 12/17/2022] Open
Abstract
Leptin is an adipokine that regulates metabolism and plays an important role as a neuroendocrine hormone. Leptin mediates these functions via the leptin receptor, and deficiency in either leptin or its receptor leads to obesity in humans and mice. Leptin has far reaching effects on the immune system, as observed in obese mice, which display decreased thymic function and increased inflammatory responses. With expression of the leptin receptor on T cells and supporting thymic epithelium, aberrant signalling through the leptin receptor has been thought to be the direct cause of thymic involution in obese mice. Here, we demonstrate that the absence of leptin receptor on either thymic epithelial cells or T cells does not lead to the loss of thymic function, demonstrating that the thymoprotective effect of leptin is mediated by obesity suppression rather than direct signalling to the cellular components of the thymus.
Collapse
Affiliation(s)
- Jayasree Sreenivasan
- VIB, Leuven, Belgium.,Department of Microbiology and Immunology, University of Leuven, Leuven, Belgium
| | - Susan Schlenner
- VIB, Leuven, Belgium.,Department of Microbiology and Immunology, University of Leuven, Leuven, Belgium
| | - Dean Franckaert
- VIB, Leuven, Belgium.,Department of Microbiology and Immunology, University of Leuven, Leuven, Belgium
| | - James Dooley
- VIB, Leuven, Belgium.,Department of Microbiology and Immunology, University of Leuven, Leuven, Belgium
| | - Adrian Liston
- VIB, Leuven, Belgium.,Department of Microbiology and Immunology, University of Leuven, Leuven, Belgium
| |
Collapse
|