1
|
Luri-Rey C, Teijeira Á, Wculek SK, de Andrea C, Herrero C, Lopez-Janeiro A, Rodríguez-Ruiz ME, Heras I, Aggelakopoulou M, Berraondo P, Sancho D, Melero I. Cross-priming in cancer immunology and immunotherapy. Nat Rev Cancer 2025; 25:249-273. [PMID: 39881005 DOI: 10.1038/s41568-024-00785-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 12/03/2024] [Indexed: 01/31/2025]
Abstract
Cytotoxic T cell immune responses against cancer crucially depend on the ability of a subtype of professional antigen-presenting cells termed conventional type 1 dendritic cells (cDC1s) to cross-present antigens. Cross-presentation comprises redirection of exogenous antigens taken from other cells to the major histocompatibility complex class I antigen-presenting machinery. In addition, once activated and having sensed viral moieties or T helper cell cooperation via CD40-CD40L interactions, cDC1s provide key co-stimulatory ligands and cytokines to mount and sustain CD8+ T cell immune responses. This regulated process of cognate T cell activation is termed cross-priming. In cancer mouse models, CD8+ T cell cross-priming by cDC1s is crucial for the efficacy of most, if not all, immunotherapy strategies. In patients with cancer, the presence and abundance of cDC1s in the tumour microenvironment is markedly associated with the level of T cell infiltration and responsiveness to immune checkpoint inhibitors. Therapeutic strategies to increase the numbers of cDC1s using FMS-like tyrosine kinase 3 ligand (FLT3L) and/or their activation status show evidence of efficacy in cancer mouse models and are currently being tested in initial clinical trials with promising results so far.
Collapse
Affiliation(s)
- Carlos Luri-Rey
- Program of Immunology and Immunotherapy, Cima Universidad de Navarra, Pamplona, Spain
| | - Álvaro Teijeira
- Program of Immunology and Immunotherapy, Cima Universidad de Navarra, Pamplona, Spain
- Navarra Institute for Health Research (IDISNA), Pamplona, Spain
- Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), Madrid, Spain
| | - Stefanie K Wculek
- Innate Immune Biology Laboratory, Institute for Research in Biomedicine (IRB Barcelona), The Barcelona Institute of Science and Technology (BIST), Barcelona, Spain
| | - Carlos de Andrea
- Department of Pathology, Clínica Universidad de Navarra, Pamplona, Spain
| | - Claudia Herrero
- Program of Immunology and Immunotherapy, Cima Universidad de Navarra, Pamplona, Spain
- Department of Pathology, Clínica Universidad de Navarra, Pamplona, Spain
| | | | | | - Ignacio Heras
- Immunobiology Laboratory, Centro Nacional de Investigaciones Cardiovasculares (CNIC), Madrid, Spain
| | | | - Pedro Berraondo
- Program of Immunology and Immunotherapy, Cima Universidad de Navarra, Pamplona, Spain
- Navarra Institute for Health Research (IDISNA), Pamplona, Spain
- Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), Madrid, Spain
| | - David Sancho
- Immunobiology Laboratory, Centro Nacional de Investigaciones Cardiovasculares (CNIC), Madrid, Spain
| | - Ignacio Melero
- Program of Immunology and Immunotherapy, Cima Universidad de Navarra, Pamplona, Spain.
- Navarra Institute for Health Research (IDISNA), Pamplona, Spain.
- Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), Madrid, Spain.
- Nuffield Department of Medicine, University of Oxford, Oxford, UK.
- Departments of Immunology and Oncology, Clínica Universidad de Navarra, Pamplona, Spain.
| |
Collapse
|
2
|
Ascic E, Pereira CF. Transcription factor-mediated reprogramming to antigen-presenting cells. Curr Opin Genet Dev 2025; 90:102300. [PMID: 39721321 DOI: 10.1016/j.gde.2024.102300] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2024] [Revised: 11/29/2024] [Accepted: 12/05/2024] [Indexed: 12/28/2024]
Abstract
Antigen-presenting cells (APCs) are a heterogenous group of immune cells composed by dendritic cells (DCs) and macrophages (Mϕ), which are critical for orchestrating immunity against cancer or infections. Several strategies have been explored to generate APC subsets, including enrichment from peripheral blood and differentiation from pluripotent or multipotent cells. During development, the generation of APC subsets is instructed by transcription factors (TFs). Direct cell reprogramming, also known as transdifferentiation, offers an approach to harness combinations of TFs to generate APCs from unrelated somatic cells, including cancer cells. In this review, we summarize the transcriptional specification of DC subsets, highlight transcriptional networks for their generation, and discuss future applications of DC reprogramming in cancer immunotherapy.
Collapse
Affiliation(s)
- Ervin Ascic
- Molecular Medicine and Gene Therapy, Lund Stem Cell Centre, Lund University, BMC A12, 221 84 Lund, Sweden; Wallenberg Center for Molecular Medicine at Lund University, BMC A12, 221 84 Lund, Sweden.
| | - Carlos-Filipe Pereira
- Molecular Medicine and Gene Therapy, Lund Stem Cell Centre, Lund University, BMC A12, 221 84 Lund, Sweden; Wallenberg Center for Molecular Medicine at Lund University, BMC A12, 221 84 Lund, Sweden; Asgard Therapeutics AB, Medicon Village, 223 81 Lund, Sweden; CNC - Centre for Neuroscience and Cell Biology, University of Coimbra, Largo Marquês do Pombal, 3004-517 Coimbra, Portugal.
| |
Collapse
|
3
|
Hubing V, Marquis A, Ziemann C, Moriyama H, Moriyama EN, Zhang L. Cytoplasmic Shift of Interferon Regulatory Factors Co-Evolved With Jawed Vertebrate Innate Immunity. J Med Virol 2025; 97:e70247. [PMID: 39977406 PMCID: PMC11841930 DOI: 10.1002/jmv.70247] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2024] [Revised: 11/26/2024] [Accepted: 02/05/2025] [Indexed: 02/22/2025]
Abstract
The emergence of jaws in early vertebrates introduced a novel feeding apparatus and powerful oral defenses, but it also increased the risk of physical injury and pathogen exposure. Interferon regulatory factors (IRFs) play critical roles in orchestrating innate immunity and inflammation in response to invading microbes and tissue damage, with their subcellular localization being essential to some IRFs' function. Our results indicate that IRF members underwent independent expansion and diversification in two distinct vertebrate lineages: jawed and jawless vertebrates. The jawed vertebrate-specific factor, IRF5, has maintained conserved nuclear export sequences throughout evolution, while newly diversified IRF members in jawed vertebrates have acquired cytoplasmic localization. This cytoplasmic shift particularly affected IRFs involved in type I interferon (IFN) signaling (IRF3, IRF5, IRF7, and IRF9), suggesting co-evolution with the development of the type I IFN system in jawed animals. Interestingly, although IRF9 is inherently nuclear, its association with Signal Transducer and Activator of Transcription 2 (STAT2) has led to its cytoplasmic localization. Additionally, IRF6, another jawed vertebrate-specific factor, plays a crucial role in jaw development, reflecting an evolutionary adaptation that aligns structural innovations with immune function. Our findings suggest that the evolution of jaws coincided with the adoption of cytoplasmic localization in IRF members, potentially enhancing rapid immune responses to meet the immunological challenges posed by the predatory lifestyle of early jawed vertebrates.
Collapse
Affiliation(s)
- Vanessa Hubing
- School of Biological SciencesUniversity of NebraskaLincolnNebraskaUSA
| | - Avery Marquis
- School of Biological SciencesUniversity of NebraskaLincolnNebraskaUSA
| | - Chanasei Ziemann
- School of Biological SciencesUniversity of NebraskaLincolnNebraskaUSA
| | - Hideaki Moriyama
- School of Biological SciencesUniversity of NebraskaLincolnNebraskaUSA
| | - Etsuko N. Moriyama
- School of Biological SciencesUniversity of NebraskaLincolnNebraskaUSA
- Center for Plant Science InnovationUniversity of NebraskaLincolnNebraskaUSA
| | - Luwen Zhang
- School of Biological SciencesUniversity of NebraskaLincolnNebraskaUSA
- Nebraska Center for VirologyUniversity of NebraskaLincolnNebraskaUSA
| |
Collapse
|
4
|
Kim S, Liu TT, Ou F, Murphy TL, Murphy KM. Anatomy of a superenhancer. Adv Immunol 2024; 163:51-96. [PMID: 39271259 DOI: 10.1016/bs.ai.2024.08.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/15/2024]
Abstract
Interferon regulatory factor-8 (IRF8) is the lineage determining transcription factor for the type one classical dendritic cell (cDC1) subset, a terminal selector for plasmacytoid dendritic cells and important for the function of monocytes. Studies of Irf8 gene regulation have identified several enhancers controlling its activity during development of progenitors in the bone marrow that precisely regulate expression at distinct developmental stages. Each enhancer responds to distinct transcription factors that are expressed at each stage. IRF8 is first expressed in early progenitors that form the monocyte dendritic cell progenitor (MDP) in response to induction of the transcription factor CCAAT/enhancer-binding protein alpha (C/EBPα) acting at the Irf8 +56 kb enhancer. IRF8 levels increase further as the MDP transits into the common dendritic cell progenitor (CDP) in response to E protein activity at the Irf8 +41 kb enhancer. Upon Nfil3-induction in CDPs leading to specification of the cDC1 progenitor, abrupt induction of BATF3 forms the JUN/BATF3/IRF8 heterotrimer that activates the Irf8 +32 kb enhancer that sustains Irf8 autoactivation throughout the cDC1 lifetime. Deletions of each of these enhancers has revealed their stage dependent activation. Surprisingly, studies of compound heterozygotes for each combination of enhancer deletions revealed that activation of each subsequent enhancer requires the successful activation of the previous enhancer in strictly cis-dependent mechanism. Successful progression of enhancer activation is finely tuned to alter the functional accessibility of subsequent enhancers to factors active in the next stage of development. The molecular basis for these phenomenon is still obscure but could have implications for genomic regulation in a broader developmental context.
Collapse
Affiliation(s)
- Sunkyung Kim
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO, United States.
| | - Tian-Tian Liu
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO, United States
| | - Feiya Ou
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO, United States
| | - Theresa L Murphy
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO, United States
| | - Kenneth M Murphy
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO, United States.
| |
Collapse
|
5
|
Shen X, Li X, Wu T, Guo T, Lv J, He Z, Luo M, Zhu X, Tian Y, Lai W, Dong C, Hu X, Wu L. TRIM33 plays a critical role in regulating dendritic cell differentiation and homeostasis by modulating Irf8 and Bcl2l11 transcription. Cell Mol Immunol 2024; 21:752-769. [PMID: 38822080 PMCID: PMC11214632 DOI: 10.1038/s41423-024-01179-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2024] [Accepted: 04/25/2024] [Indexed: 06/02/2024] Open
Abstract
The development of distinct dendritic cell (DC) subsets, namely, plasmacytoid DCs (pDCs) and conventional DC subsets (cDC1s and cDC2s), is controlled by specific transcription factors. IRF8 is essential for the fate specification of cDC1s. However, how the expression of Irf8 is regulated is not fully understood. In this study, we identified TRIM33 as a critical regulator of DC differentiation and maintenance. TRIM33 deletion in Trim33fl/fl Cre-ERT2 mice significantly impaired DC differentiation from hematopoietic progenitors at different developmental stages. TRIM33 deficiency downregulated the expression of multiple genes associated with DC differentiation in these progenitors. TRIM33 promoted the transcription of Irf8 to facilitate the differentiation of cDC1s by maintaining adequate CDK9 and Ser2 phosphorylated RNA polymerase II (S2 Pol II) levels at Irf8 gene sites. Moreover, TRIM33 prevented the apoptosis of DCs and progenitors by directly suppressing the PU.1-mediated transcription of Bcl2l11, thereby maintaining DC homeostasis. Taken together, our findings identified TRIM33 as a novel and crucial regulator of DC differentiation and maintenance through the modulation of Irf8 and Bcl2l11 expression. The finding that TRIM33 functions as a critical regulator of both DC differentiation and survival provides potential benefits for devising DC-based immune interventions and therapies.
Collapse
Affiliation(s)
- Xiangyi Shen
- Institute for Immunology, School of Basic Medical Sciences, Tsinghua University, 100084, Beijing, China
| | - Xiaoguang Li
- Institute for Immunology, School of Basic Medical Sciences, Tsinghua University, 100084, Beijing, China
- Joint Graduate Program of Peking-Tsinghua-National Institute of Biological Sciences, School of Life Sciences, Tsinghua University, 100084, Beijing, China
| | - Tao Wu
- Institute for Immunology, School of Basic Medical Sciences, Tsinghua University, 100084, Beijing, China
- Tsinghua-Peking Center for Life Sciences, Tsinghua University, 100084, Beijing, China
| | - Tingting Guo
- Institute for Immunology, School of Basic Medical Sciences, Tsinghua University, 100084, Beijing, China
- Tsinghua-Peking Center for Life Sciences, Tsinghua University, 100084, Beijing, China
| | - Jiaoyan Lv
- Institute for Immunology, School of Basic Medical Sciences, Tsinghua University, 100084, Beijing, China
- Tsinghua-Peking Center for Life Sciences, Tsinghua University, 100084, Beijing, China
| | - Zhimin He
- Institute for Immunology, School of Basic Medical Sciences, Tsinghua University, 100084, Beijing, China
- Tsinghua-Peking Center for Life Sciences, Tsinghua University, 100084, Beijing, China
| | - Maocai Luo
- Institute for Immunology, School of Basic Medical Sciences, Tsinghua University, 100084, Beijing, China
| | - Xinyi Zhu
- Institute for Immunology, School of Basic Medical Sciences, Tsinghua University, 100084, Beijing, China
- Tsinghua-Peking Center for Life Sciences, Tsinghua University, 100084, Beijing, China
| | - Yujie Tian
- Institute for Immunology, School of Basic Medical Sciences, Tsinghua University, 100084, Beijing, China
- Joint Graduate Program of Peking-Tsinghua-National Institute of Biological Sciences, School of Life Sciences, Tsinghua University, 100084, Beijing, China
| | - Wenlong Lai
- Institute for Immunology, School of Basic Medical Sciences, Tsinghua University, 100084, Beijing, China
| | - Chen Dong
- Institute for Immunology, School of Basic Medical Sciences, Tsinghua University, 100084, Beijing, China
- Tsinghua-Peking Center for Life Sciences, Tsinghua University, 100084, Beijing, China
- Beijing Key Laboratory for Immunological Research on Chronic Diseases, 100084, Beijing, China
- Westlake University School of Medicine, Hangzhou, 310024, China
| | - Xiaoyu Hu
- Institute for Immunology, School of Basic Medical Sciences, Tsinghua University, 100084, Beijing, China
- Tsinghua-Peking Center for Life Sciences, Tsinghua University, 100084, Beijing, China
- Beijing Key Laboratory for Immunological Research on Chronic Diseases, 100084, Beijing, China
| | - Li Wu
- Institute for Immunology, School of Basic Medical Sciences, Tsinghua University, 100084, Beijing, China.
- Tsinghua-Peking Center for Life Sciences, Tsinghua University, 100084, Beijing, China.
- Beijing Key Laboratory for Immunological Research on Chronic Diseases, 100084, Beijing, China.
| |
Collapse
|
6
|
Li N, Steiger S, Zhong M, Lu M, Lei Y, Tang C, Chen J, Guo Y, Li J, Zhang D, Li J, Zhu E, Zheng Z, Lichtnekert J, Chen Y, Wang X. IRF8 maintains mononuclear phagocyte and neutrophil function in acute kidney injury. Heliyon 2024; 10:e31818. [PMID: 38845872 PMCID: PMC11153194 DOI: 10.1016/j.heliyon.2024.e31818] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2024] [Revised: 05/22/2024] [Accepted: 05/22/2024] [Indexed: 06/09/2024] Open
Abstract
Immune cells are key players in acute tissue injury and inflammation, including acute kidney injury (AKI). Their development, differentiation, activation status, and functions are mediated by a variety of transcription factors, such as interferon regulatory factor 8 (IRF8) and IRF4. We speculated that IRF8 has a pathophysiologic impact on renal immune cells in AKI and found that IRF8 is highly expressed in blood type 1 conventional dendritic cells (cDC1s), monocytes, monocyte-derived dendritic cells (moDCs) and kidney biopsies from patients with AKI. In a mouse model of ischemia‒reperfusion injury (IRI)-induced AKI, Irf8 -/- mice displayed increased tubular cell necrosis and worsened kidney dysfunction associated with the recruitment of a substantial amount of monocytes and neutrophils but defective renal infiltration of cDC1s and moDCs. Mechanistically, global Irf8 deficiency impaired moDC and cDC1 maturation and activation, as well as cDC1 proliferation, antigen uptake, and trafficking to lymphoid organs for T-cell priming in ischemic AKI. Moreover, compared with Irf8 +/+ mice, Irf8 -/- mice exhibited increased neutrophil recruitment and neutrophil extracellular trap (NET) formation following AKI. IRF8 primarily regulates cDC1 and indirectly neutrophil functions, and thereby protects mice from kidney injury and inflammation following IRI. Our results demonstrate that IRF8 plays a predominant immunoregulatory role in cDC1 function and therefore represents a potential therapeutic target in AKI.
Collapse
Affiliation(s)
- Na Li
- Department of Nephrology, Center of Kidney and Urology, The Seventh Affiliated Hospital, Sun Yat-Sen University, 518107, Shenzhen, China
- Scientific Research Center, Edmond H. Fischer Translational Medical Research Laboratory, The Seventh Affiliated Hospital, Sun Yat-Sen University, 518107, Shenzhen, China
| | - Stefanie Steiger
- Renal Division, Department of Medicine IV, Ludwig-Maximilians-University Hospital, Ludwig-Maximilian-University Munich, 80336, Munich, Bavaria, Germany
| | - Ming Zhong
- Department of Nephrology, Center of Kidney and Urology, The Seventh Affiliated Hospital, Sun Yat-Sen University, 518107, Shenzhen, China
| | - Meihua Lu
- Department of Geriatrics, People's Hospital of Banan District, 401320, Chongqing, China
| | - Yan Lei
- Department of Nephrology, Center of Kidney and Urology, The Seventh Affiliated Hospital, Sun Yat-Sen University, 518107, Shenzhen, China
| | - Chun Tang
- Department of Nephrology, Center of Kidney and Urology, The Seventh Affiliated Hospital, Sun Yat-Sen University, 518107, Shenzhen, China
| | - Jiasi Chen
- Department of Nephrology, Center of Kidney and Urology, The Seventh Affiliated Hospital, Sun Yat-Sen University, 518107, Shenzhen, China
| | - Yao Guo
- Scientific Research Center, Edmond H. Fischer Translational Medical Research Laboratory, The Seventh Affiliated Hospital, Sun Yat-Sen University, 518107, Shenzhen, China
| | - Jinhong Li
- Department of Nephrology, Center of Kidney and Urology, The Seventh Affiliated Hospital, Sun Yat-Sen University, 518107, Shenzhen, China
| | - Dengyang Zhang
- Scientific Research Center, Edmond H. Fischer Translational Medical Research Laboratory, The Seventh Affiliated Hospital, Sun Yat-Sen University, 518107, Shenzhen, China
| | - Jingyi Li
- Department of Pulmonary & Critical Care Medicine, Shenzhen Hospital of Southern Medical University, 518107, Shenzhen, China
| | - Enyi Zhu
- Department of Nephrology, Center of Kidney and Urology, The Seventh Affiliated Hospital, Sun Yat-Sen University, 518107, Shenzhen, China
| | - Zhihua Zheng
- Department of Nephrology, Center of Kidney and Urology, The Seventh Affiliated Hospital, Sun Yat-Sen University, 518107, Shenzhen, China
| | - Julia Lichtnekert
- Renal Division, Department of Medicine IV, Ludwig-Maximilians-University Hospital, Ludwig-Maximilian-University Munich, 80336, Munich, Bavaria, Germany
| | - Yun Chen
- Scientific Research Center, Edmond H. Fischer Translational Medical Research Laboratory, The Seventh Affiliated Hospital, Sun Yat-Sen University, 518107, Shenzhen, China
| | - Xiaohua Wang
- Department of Nephrology, Center of Kidney and Urology, The Seventh Affiliated Hospital, Sun Yat-Sen University, 518107, Shenzhen, China
| |
Collapse
|
7
|
Bayerl F, Bejarano DA, Bertacchi G, Doffin AC, Gobbini E, Hubert M, Li L, Meiser P, Pedde AM, Posch W, Rupp L, Schlitzer A, Schmitz M, Schraml BU, Uderhardt S, Valladeau-Guilemond J, Wilflingseder D, Zaderer V, Böttcher JP. Guidelines for visualization and analysis of DC in tissues using multiparameter fluorescence microscopy imaging methods. Eur J Immunol 2023; 53:e2249923. [PMID: 36623939 DOI: 10.1002/eji.202249923] [Citation(s) in RCA: 21] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2022] [Revised: 11/07/2022] [Accepted: 11/14/2022] [Indexed: 01/11/2023]
Abstract
This article is part of the Dendritic Cell Guidelines article series, which provides a collection of state-of-the-art protocols for the preparation, phenotype analysis by flow cytometry, generation, fluorescence microscopy, and functional characterization of mouse and human dendritic cells (DC) from lymphoid organs and various non-lymphoid tissues. Here, we provide detailed procedures for a variety of multiparameter fluorescence microscopy imaging methods to explore the spatial organization of DC in tissues and to dissect how DC migrate, communicate, and mediate their multiple functional roles in immunity in a variety of tissue settings. The protocols presented here entail approaches to study DC dynamics and T cell cross-talk by intravital microscopy, large-scale visualization, identification, and quantitative analysis of DC subsets and their functions by multiparameter fluorescence microscopy of fixed tissue sections, and an approach to study DC interactions with tissue cells in a 3D cell culture model. While all protocols were written by experienced scientists who routinely use them in their work, this article was also peer-reviewed by leading experts and approved by all co-authors, making it an essential resource for basic and clinical DC immunologists.
Collapse
Affiliation(s)
- Felix Bayerl
- Institute of Molecular Immunology, School of Medicine, Klinikum Rechts der Isar, Technical University of Munich (TUM), Ismaninger Str. 22, Munich, Germany
| | - David A Bejarano
- Quantitative Systems Biology, Life and Medical Sciences (LIMES) Institute, University of Bonn, Germany
| | - Giulia Bertacchi
- Institute of Hygiene and Medical Microbiology, Medical University of Innsbruck, Innsbruck, Austria
| | - Anne-Claire Doffin
- Cancer Research Center Lyon, UMR INSERM 1052 CNRS 5286, Centre Léon Bérard, 28 rue Laennec, Lyon, France
| | - Elisa Gobbini
- Cancer Research Center Lyon, UMR INSERM 1052 CNRS 5286, Centre Léon Bérard, 28 rue Laennec, Lyon, France
| | - Margaux Hubert
- Cancer Research Center Lyon, UMR INSERM 1052 CNRS 5286, Centre Léon Bérard, 28 rue Laennec, Lyon, France
| | - Lijian Li
- Department of Internal Medicine 3 - Rheumatology and Immunology, Friedrich-Alexander University Erlangen-Nürnberg (FAU) and Universitätsklinikum Erlangen, Erlangen, Germany
- Deutsches Zentrum für Immuntherapie (DZI), Friedrich-Alexander University Erlangen-Nürnberg (FAU) and Universitätsklinikum Erlangen, Erlangen, Germany
- Exploratory Research Unit, Optical Imaging Centre Erlangen (OICE), Friedrich-Alexander University Erlangen-Nürnberg (FAU), Erlangen, Germany
| | - Philippa Meiser
- Institute of Molecular Immunology, School of Medicine, Klinikum Rechts der Isar, Technical University of Munich (TUM), Ismaninger Str. 22, Munich, Germany
| | - Anna-Marie Pedde
- Institute of Molecular Immunology, School of Medicine, Klinikum Rechts der Isar, Technical University of Munich (TUM), Ismaninger Str. 22, Munich, Germany
| | - Wilfried Posch
- Institute of Hygiene and Medical Microbiology, Medical University of Innsbruck, Innsbruck, Austria
| | - Luise Rupp
- Institute of Immunology, Faculty of Medicine Carl Gustav Carus, TU Dresden, Dresden, Germany
| | - Andreas Schlitzer
- Quantitative Systems Biology, Life and Medical Sciences (LIMES) Institute, University of Bonn, Germany
| | - Marc Schmitz
- Institute of Immunology, Faculty of Medicine Carl Gustav Carus, TU Dresden, Dresden, Germany
- National Center for Tumor Diseases (NCT), Partner Site Dresden, Dresden, Germany
- German Cancer Consortium (DKTK), Partner Site Dresden, and German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Barbara U Schraml
- Walter-Brendel-Centre of Experimental Medicine, University Hospital, LMU Munich, Planegg-Martinsried, Germany
- Biomedical Center, Institute for Cardiovascular Physiology and Pathophysiology, Faculty of Medicine, LMU Munich, Planegg-Martinsried, Germany
| | - Stefan Uderhardt
- Department of Internal Medicine 3 - Rheumatology and Immunology, Friedrich-Alexander University Erlangen-Nürnberg (FAU) and Universitätsklinikum Erlangen, Erlangen, Germany
- Deutsches Zentrum für Immuntherapie (DZI), Friedrich-Alexander University Erlangen-Nürnberg (FAU) and Universitätsklinikum Erlangen, Erlangen, Germany
- Exploratory Research Unit, Optical Imaging Centre Erlangen (OICE), Friedrich-Alexander University Erlangen-Nürnberg (FAU), Erlangen, Germany
| | - Jenny Valladeau-Guilemond
- Cancer Research Center Lyon, UMR INSERM 1052 CNRS 5286, Centre Léon Bérard, 28 rue Laennec, Lyon, France
| | - Doris Wilflingseder
- Institute of Hygiene and Medical Microbiology, Medical University of Innsbruck, Innsbruck, Austria
| | - Viktoria Zaderer
- Institute of Hygiene and Medical Microbiology, Medical University of Innsbruck, Innsbruck, Austria
| | - Jan P Böttcher
- Institute of Molecular Immunology, School of Medicine, Klinikum Rechts der Isar, Technical University of Munich (TUM), Ismaninger Str. 22, Munich, Germany
| |
Collapse
|
8
|
Zhang S, Audiger C, Chopin M, Nutt SL. Transcriptional regulation of dendritic cell development and function. Front Immunol 2023; 14:1182553. [PMID: 37520521 PMCID: PMC10382230 DOI: 10.3389/fimmu.2023.1182553] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2023] [Accepted: 06/28/2023] [Indexed: 08/01/2023] Open
Abstract
Dendritic cells (DCs) are sentinel immune cells that form a critical bridge linking the innate and adaptive immune systems. Extensive research addressing the cellular origin and heterogeneity of the DC network has revealed the essential role played by the spatiotemporal activity of key transcription factors. In response to environmental signals DC mature but it is only following the sensing of environmental signals that DC can induce an antigen specific T cell response. Thus, whilst the coordinate action of transcription factors governs DC differentiation, sensing of environmental signals by DC is instrumental in shaping their functional properties. In this review, we provide an overview that focuses on recent advances in understanding the transcriptional networks that regulate the development of the reported DC subsets, shedding light on the function of different DC subsets. Specifically, we discuss the emerging knowledge on the heterogeneity of cDC2s, the ontogeny of pDCs, and the newly described DC subset, DC3. Additionally, we examine critical transcription factors such as IRF8, PU.1, and E2-2 and their regulatory mechanisms and downstream targets. We highlight the complex interplay between these transcription factors, which shape the DC transcriptome and influence their function in response to environmental stimuli. The information presented in this review provides essential insights into the regulation of DC development and function, which might have implications for developing novel therapeutic strategies for immune-related diseases.
Collapse
Affiliation(s)
- Shengbo Zhang
- Immunology Division, Walter and Eliza Hall Institute of Medical Research, Parkville, VIC, Australia
- Department of Medical Biology, University of Melbourne, Parkville, VIC, Australia
| | - Cindy Audiger
- Immunology Division, Walter and Eliza Hall Institute of Medical Research, Parkville, VIC, Australia
- Department of Medical Biology, University of Melbourne, Parkville, VIC, Australia
| | - Michaël Chopin
- Department of Biochemistry and Molecular Biology, Biomedicine Discovery Institute, Monash University, Clayton, VIC, Australia
| | - Stephen L. Nutt
- Immunology Division, Walter and Eliza Hall Institute of Medical Research, Parkville, VIC, Australia
- Department of Medical Biology, University of Melbourne, Parkville, VIC, Australia
| |
Collapse
|
9
|
Johnson KD, Jung MM, Tran VL, Bresnick EH. Interferon regulatory factor-8-dependent innate immune alarm senses GATA2 deficiency to alter hematopoietic differentiation and function. Curr Opin Hematol 2023; 30:117-123. [PMID: 37254854 PMCID: PMC10236032 DOI: 10.1097/moh.0000000000000763] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/01/2023]
Abstract
PURPOSE OF REVIEW Recent discoveries have provided evidence for mechanistic links between the master regulator of hematopoiesis GATA2 and the key component of interferon and innate immunity signaling pathways, interferon-regulatory factor-8 (IRF8). These links have important implications for the control of myeloid differentiation in physiological and pathological states. RECENT FINDINGS GATA2 deficiency resulting from loss of the Gata2 -77 enhancer in progenitors triggers an alarm that instigates the transcriptional induction of innate immune signaling and distorts a myeloid differentiation program. This pathological alteration renders progenitors hyperresponsive to interferon γ, toll-like receptor and interleukin-6 signaling and impaired in granulocyte-macrophage colony-stimulating factor signaling. IRF8 upregulation in -77-/- progenitors promotes monocyte and dendritic cell differentiation while suppressing granulocytic differentiation. As PU.1 promotes transcription of Irf8 and other myeloid and B-lineage genes, GATA2-mediated repression of these genes opposes the PU.1-dependent activating mechanism. SUMMARY As GATA2 deficiency syndrome is an immunodeficiency disorder often involving myelodysplastic syndromes and acute myeloid leukemia, elucidating how GATA2 commissions and decommissions genome activity and developmental regulatory programs will unveil mechanisms that go awry when GATA2 levels and/or activities are disrupted.
Collapse
Affiliation(s)
- Kirby D Johnson
- Wisconsin Blood Cancer Research Institute, Department of Cell and Regenerative Biology, Carbone Cancer Center, University of Wisconsin School of Medicine and Public Health, Madison, Wisconsin, USA
| | | | | | | |
Collapse
|
10
|
In Vitro Human Haematopoietic Stem Cell Expansion and Differentiation. Cells 2023; 12:cells12060896. [PMID: 36980237 PMCID: PMC10046976 DOI: 10.3390/cells12060896] [Citation(s) in RCA: 20] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2023] [Revised: 03/08/2023] [Accepted: 03/09/2023] [Indexed: 03/17/2023] Open
Abstract
The haematopoietic system plays an essential role in our health and survival. It is comprised of a range of mature blood and immune cell types, including oxygen-carrying erythrocytes, platelet-producing megakaryocytes and infection-fighting myeloid and lymphoid cells. Self-renewing multipotent haematopoietic stem cells (HSCs) and a range of intermediate haematopoietic progenitor cell types differentiate into these mature cell types to continuously support haematopoietic system homeostasis throughout life. This process of haematopoiesis is tightly regulated in vivo and primarily takes place in the bone marrow. Over the years, a range of in vitro culture systems have been developed, either to expand haematopoietic stem and progenitor cells or to differentiate them into the various haematopoietic lineages, based on the use of recombinant cytokines, co-culture systems and/or small molecules. These approaches provide important tractable models to study human haematopoiesis in vitro. Additionally, haematopoietic cell culture systems are being developed and clinical tested as a source of cell products for transplantation and transfusion medicine. This review discusses the in vitro culture protocols for human HSC expansion and differentiation, and summarises the key factors involved in these biological processes.
Collapse
|
11
|
Hujoel MLA, Sherman MA, Barton AR, Mukamel RE, Sankaran VG, Terao C, Loh PR. Influences of rare copy-number variation on human complex traits. Cell 2022; 185:4233-4248.e27. [PMID: 36306736 PMCID: PMC9800003 DOI: 10.1016/j.cell.2022.09.028] [Citation(s) in RCA: 30] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2021] [Revised: 07/22/2022] [Accepted: 09/19/2022] [Indexed: 11/06/2022]
Abstract
The human genome contains hundreds of thousands of regions harboring copy-number variants (CNV). However, the phenotypic effects of most such polymorphisms are unknown because only larger CNVs have been ascertainable from SNP-array data generated by large biobanks. We developed a computational approach leveraging haplotype sharing in biobank cohorts to more sensitively detect CNVs. Applied to UK Biobank, this approach accounted for approximately half of all rare gene inactivation events produced by genomic structural variation. This CNV call set enabled a detailed analysis of associations between CNVs and 56 quantitative traits, identifying 269 independent associations (p < 5 × 10-8) likely to be causally driven by CNVs. Putative target genes were identifiable for nearly half of the loci, enabling insights into dosage sensitivity of these genes and uncovering several gene-trait relationships. These results demonstrate the ability of haplotype-informed analysis to provide insights into the genetic basis of human complex traits.
Collapse
Affiliation(s)
- Margaux L A Hujoel
- Division of Genetics, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA; Center for Data Sciences, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA; Program in Medical and Population Genetics, Broad Institute of MIT and Harvard, Cambridge, MA, USA.
| | - Maxwell A Sherman
- Division of Genetics, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA; Center for Data Sciences, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA; Program in Medical and Population Genetics, Broad Institute of MIT and Harvard, Cambridge, MA, USA; Computer Science and Artificial Intelligence Laboratory, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Alison R Barton
- Division of Genetics, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA; Center for Data Sciences, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA; Bioinformatics and Integrative Genomics Program, Harvard Medical School, Boston, MA, USA
| | - Ronen E Mukamel
- Division of Genetics, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA; Center for Data Sciences, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA; Program in Medical and Population Genetics, Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Vijay G Sankaran
- Program in Medical and Population Genetics, Broad Institute of MIT and Harvard, Cambridge, MA, USA; Division of Hematology/Oncology, Boston Children's Hospital, Harvard Medical School, Boston, MA, USA; Department of Pediatric Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, USA
| | - Chikashi Terao
- Laboratory for Statistical and Translational Genetics, RIKEN Center for Integrative Medical Sciences, Yokohama, Japan; Clinical Research Center, Shizuoka General Hospital, Shizuoka, Japan; Department of Applied Genetics, School of Pharmaceutical Sciences, University of Shizuoka, Shizuoka, Japan
| | - Po-Ru Loh
- Division of Genetics, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA; Center for Data Sciences, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA; Program in Medical and Population Genetics, Broad Institute of MIT and Harvard, Cambridge, MA, USA.
| |
Collapse
|
12
|
Moorman HR, Reategui Y, Poschel DB, Liu K. IRF8: Mechanism of Action and Health Implications. Cells 2022; 11:2630. [PMID: 36078039 PMCID: PMC9454819 DOI: 10.3390/cells11172630] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2022] [Revised: 08/17/2022] [Accepted: 08/21/2022] [Indexed: 11/29/2022] Open
Abstract
Interferon regulatory factor 8 (IRF8) is a transcription factor of the IRF protein family. IRF8 was originally identified as an essentialfactor for myeloid cell lineage commitment and differentiation. Deletion of Irf8 leads to massive accumulation of CD11b+Gr1+ immature myeloid cells (IMCs), particularly the CD11b+Ly6Chi/+Ly6G- polymorphonuclear myeloid-derived suppressor cell-like cells (PMN-MDSCs). Under pathological conditions such as cancer, Irf8 is silenced by its promoter DNA hypermethylation, resulting in accumulation of PMN-MDSCs and CD11b+ Ly6G+Ly6Clo monocytic MDSCs (M-MDSCs) in mice. IRF8 is often silenced in MDSCs in human cancer patients. MDSCs are heterogeneous populations of immune suppressive cells that suppress T and NK cell activity to promote tumor immune evasion and produce growth factors to exert direct tumor-promoting activity. Emerging experimental data reveals that IRF8 is also expressed in non-hematopoietic cells. Epithelial cell-expressed IRF8 regulates apoptosis and represses Osteopontin (OPN). Human tumor cells may use the IRF8 promoter DNA methylation as a mechanism to repress IRF8 expression to advance cancer through acquiring apoptosis resistance and OPN up-regulation. Elevated OPN engages CD44 to suppress T cell activation and promote tumor cell stemness to advance cancer. IRF8 thus is a transcription factor that regulates both the immune and non-immune components in human health and diseases.
Collapse
Affiliation(s)
- Hannah R. Moorman
- Department of Biochemistry and Molecular Biology, Medical College of Georgia, Augusta, GA 30912, USA
| | - Yazmin Reategui
- Department of Biochemistry and Molecular Biology, Medical College of Georgia, Augusta, GA 30912, USA
| | - Dakota B. Poschel
- Department of Biochemistry and Molecular Biology, Medical College of Georgia, Augusta, GA 30912, USA
- Georgia Cancer Center, Augusta, GA 30912, USA
- Charlie Norwood VA Medical Center, Augusta, GA 30904, USA
| | - Kebin Liu
- Department of Biochemistry and Molecular Biology, Medical College of Georgia, Augusta, GA 30912, USA
- Georgia Cancer Center, Augusta, GA 30912, USA
- Charlie Norwood VA Medical Center, Augusta, GA 30904, USA
| |
Collapse
|
13
|
Gargaro M, Scalisi G, Manni G, Briseño CG, Bagadia P, Durai V, Theisen DJ, Kim S, Castelli M, Xu CA, zu Hörste GM, Servillo G, Della Fazia MA, Mencarelli G, Ricciuti D, Padiglioni E, Giacchè N, Colliva C, Pellicciari R, Calvitti M, Zelante T, Fuchs D, Orabona C, Boon L, Bessede A, Colonna M, Puccetti P, Murphy TL, Murphy KM, Fallarino F. Indoleamine 2,3-dioxygenase 1 activation in mature cDC1 promotes tolerogenic education of inflammatory cDC2 via metabolic communication. Immunity 2022; 55:1032-1050.e14. [PMID: 35704993 PMCID: PMC9220322 DOI: 10.1016/j.immuni.2022.05.013] [Citation(s) in RCA: 60] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2021] [Revised: 02/07/2022] [Accepted: 05/17/2022] [Indexed: 12/14/2022]
Abstract
Conventional dendritic cells (cDCs), cDC1 and cDC2, act both to initiate immunity and maintain self-tolerance. The tryptophan metabolic enzyme indoleamine 2,3-dioxygenase 1 (IDO1) is used by cDCs in maintaining tolerance, but its role in different subsets remains unclear. At homeostasis, only mature CCR7+ cDC1 expressed IDO1 that was dependent on IRF8. Lipopolysaccharide treatment induced maturation and IDO1-dependent tolerogenic activity in isolated immature cDC1, but not isolated cDC2. However, both human and mouse cDC2 could induce IDO1 and acquire tolerogenic function when co-cultured with mature cDC1 through the action of cDC1-derived l-kynurenine. Accordingly, cDC1-specific inactivation of IDO1 in vivo exacerbated disease in experimental autoimmune encephalomyelitis. This study identifies a previously unrecognized metabolic communication in which IDO1-expressing cDC1 cells extend their immunoregulatory capacity to the cDC2 subset through their production of tryptophan metabolite l-kynurenine. This metabolic axis represents a potential therapeutic target in treating autoimmune demyelinating diseases.
Collapse
Affiliation(s)
- Marco Gargaro
- Department of Medicine and Surgery, University of Perugia, Perugia, Italy,Department of Pathology and Immunology, Washington University in St. Louis School of Medicine, St. Louis, MO, USA
| | - Giulia Scalisi
- Department of Medicine and Surgery, University of Perugia, Perugia, Italy
| | - Giorgia Manni
- Department of Medicine and Surgery, University of Perugia, Perugia, Italy
| | - Carlos G. Briseño
- Department of Pathology and Immunology, Washington University in St. Louis School of Medicine, St. Louis, MO, USA
| | - Prachi Bagadia
- Department of Pathology and Immunology, Washington University in St. Louis School of Medicine, St. Louis, MO, USA
| | - Vivek Durai
- Department of Pathology and Immunology, Washington University in St. Louis School of Medicine, St. Louis, MO, USA
| | - Derek J. Theisen
- Department of Pathology and Immunology, Washington University in St. Louis School of Medicine, St. Louis, MO, USA
| | - Sunkyung Kim
- Department of Pathology and Immunology, Washington University in St. Louis School of Medicine, St. Louis, MO, USA
| | - Marilena Castelli
- Department of Medicine and Surgery, University of Perugia, Perugia, Italy
| | - Chenling A. Xu
- Department of Electrical Engineering & Computer Science, Center for Computational Biology, University of California, Berkeley, CA, USA
| | - Gerd Meyer zu Hörste
- Department of Neurology with Institute of Translational Neurology, University Hospital Münster, Münster, Germany
| | - Giuseppe Servillo
- Department of Medicine and Surgery, University of Perugia, Perugia, Italy,University research center in functional genomics (c.u.r.ge.f.), University of Perugia, Perugia, Italy
| | | | - Giulia Mencarelli
- Department of Medicine and Surgery, University of Perugia, Perugia, Italy
| | - Doriana Ricciuti
- Department of Medicine and Surgery, University of Perugia, Perugia, Italy
| | | | | | | | | | - Mario Calvitti
- Department of Medicine and Surgery, University of Perugia, Perugia, Italy
| | - Teresa Zelante
- Department of Medicine and Surgery, University of Perugia, Perugia, Italy
| | - Dietmar Fuchs
- Division of Biological Chemistry, Biocenter, Innsbruck Medical University, Innsbruck, Austria
| | - Ciriana Orabona
- Department of Medicine and Surgery, University of Perugia, Perugia, Italy
| | | | | | - Marco Colonna
- Department of Pathology and Immunology, Washington University in St. Louis School of Medicine, St. Louis, MO, USA
| | - Paolo Puccetti
- Department of Medicine and Surgery, University of Perugia, Perugia, Italy,University research center in functional genomics (c.u.r.ge.f.), University of Perugia, Perugia, Italy
| | - Theresa L. Murphy
- Department of Pathology and Immunology, Washington University in St. Louis School of Medicine, St. Louis, MO, USA
| | - Kenneth M. Murphy
- Department of Pathology and Immunology, Washington University in St. Louis School of Medicine, St. Louis, MO, USA,Howard Hughes Medical Institute, Washington University in St. Louis School of Medicine, St. Louis, MO, USA,Corresponding author
| | - Francesca Fallarino
- Department of Medicine and Surgery, University of Perugia, Perugia, Italy; University research center in functional genomics (c.u.r.ge.f.), University of Perugia, Perugia, Italy.
| |
Collapse
|
14
|
Rosa FF, Pires CF, Kurochkin I, Halitzki E, Zahan T, Arh N, Zimmermannová O, Ferreira AG, Li H, Karlsson S, Scheding S, Pereira CF. Single-cell transcriptional profiling informs efficient reprogramming of human somatic cells to cross-presenting dendritic cells. Sci Immunol 2022; 7:eabg5539. [PMID: 35245086 DOI: 10.1126/sciimmunol.abg5539] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023]
Abstract
Type 1 conventional dendritic cells (cDC1s) are rare immune cells critical for the induction of antigen-specific cytotoxic CD8+ T cells, although the genetic program driving human cDC1 specification remains largely unexplored. We previously identified PU.1, IRF8, and BATF3 transcription factors as sufficient to induce cDC1 fate in mouse fibroblasts, but reprogramming of human somatic cells was limited by low efficiency. Here, we investigated single-cell transcriptional dynamics during human cDC1 reprogramming. Human induced cDC1s (hiDC1s) generated from embryonic fibroblasts gradually acquired a global cDC1 transcriptional profile and expressed antigen presentation signatures, whereas other DC subsets were not induced at the single-cell level during the reprogramming process. We extracted gene modules associated with successful reprogramming and identified inflammatory signaling and the cDC1-inducing transcription factor network as key drivers of the process. Combining IFN-γ, IFN-β, and TNF-α with constitutive expression of cDC1-inducing transcription factors led to improvement of reprogramming efficiency by 190-fold. hiDC1s engulfed dead cells, secreted inflammatory cytokines, and performed antigen cross-presentation, key cDC1 functions. This approach allowed efficient hiDC1 generation from adult fibroblasts and mesenchymal stromal cells. Mechanistically, PU.1 showed dominant and independent chromatin targeting at early phases of reprogramming, recruiting IRF8 and BATF3 to shared binding sites. The cooperative binding at open enhancers and promoters led to silencing of fibroblast genes and activation of a cDC1 program. These findings provide mechanistic insights into human cDC1 specification and reprogramming and represent a platform for generating patient-tailored cDC1s, a long-sought DC subset for vaccination strategies in cancer immunotherapy.
Collapse
Affiliation(s)
- Fábio F Rosa
- Molecular Medicine and Gene Therapy, Lund Stem Cell Centre, Lund University, BMC A12, 221 84 Lund, Sweden.,Wallenberg Centre for Molecular Medicine, Lund University, BMC A12, 221 84 Lund, Sweden.,Centre for Neuroscience and Cell Biology, University of Coimbra, Largo Marquês do Pombal 3004-517 Coimbra, Portugal.,Doctoral Programme in Experimental Biology and Biomedicine, University of Coimbra, Coimbra, Portugal
| | - Cristiana F Pires
- Molecular Medicine and Gene Therapy, Lund Stem Cell Centre, Lund University, BMC A12, 221 84 Lund, Sweden.,Wallenberg Centre for Molecular Medicine, Lund University, BMC A12, 221 84 Lund, Sweden.,Centre for Neuroscience and Cell Biology, University of Coimbra, Largo Marquês do Pombal 3004-517 Coimbra, Portugal
| | - Ilia Kurochkin
- Molecular Medicine and Gene Therapy, Lund Stem Cell Centre, Lund University, BMC A12, 221 84 Lund, Sweden.,Wallenberg Centre for Molecular Medicine, Lund University, BMC A12, 221 84 Lund, Sweden.,Skolkovo Institute of Science and Technology, Nobel Street, Building 3, Moscow 143026, Russia
| | - Evelyn Halitzki
- Molecular Medicine and Gene Therapy, Lund Stem Cell Centre, Lund University, BMC A12, 221 84 Lund, Sweden.,Wallenberg Centre for Molecular Medicine, Lund University, BMC A12, 221 84 Lund, Sweden
| | - Tasnim Zahan
- Molecular Medicine and Gene Therapy, Lund Stem Cell Centre, Lund University, BMC A12, 221 84 Lund, Sweden.,Wallenberg Centre for Molecular Medicine, Lund University, BMC A12, 221 84 Lund, Sweden
| | - Nejc Arh
- Molecular Medicine and Gene Therapy, Lund Stem Cell Centre, Lund University, BMC A12, 221 84 Lund, Sweden.,Wallenberg Centre for Molecular Medicine, Lund University, BMC A12, 221 84 Lund, Sweden
| | - Olga Zimmermannová
- Molecular Medicine and Gene Therapy, Lund Stem Cell Centre, Lund University, BMC A12, 221 84 Lund, Sweden.,Wallenberg Centre for Molecular Medicine, Lund University, BMC A12, 221 84 Lund, Sweden
| | - Alexandra G Ferreira
- Molecular Medicine and Gene Therapy, Lund Stem Cell Centre, Lund University, BMC A12, 221 84 Lund, Sweden.,Wallenberg Centre for Molecular Medicine, Lund University, BMC A12, 221 84 Lund, Sweden.,Centre for Neuroscience and Cell Biology, University of Coimbra, Largo Marquês do Pombal 3004-517 Coimbra, Portugal.,Doctoral Programme in Experimental Biology and Biomedicine, University of Coimbra, Coimbra, Portugal
| | - Hongzhe Li
- Division of Molecular Hematology, Department of Laboratory Medicine, Lund University, BMC B12, 221 84 Lund, Sweden
| | - Stefan Karlsson
- Molecular Medicine and Gene Therapy, Lund Stem Cell Centre, Lund University, BMC A12, 221 84 Lund, Sweden
| | - Stefan Scheding
- Division of Molecular Hematology, Department of Laboratory Medicine, Lund University, BMC B12, 221 84 Lund, Sweden.,Department of Hematology, Skåne University Hospital Lund, Skåne, 222 42 Lund, Sweden
| | - Carlos-Filipe Pereira
- Molecular Medicine and Gene Therapy, Lund Stem Cell Centre, Lund University, BMC A12, 221 84 Lund, Sweden.,Wallenberg Centre for Molecular Medicine, Lund University, BMC A12, 221 84 Lund, Sweden.,Centre for Neuroscience and Cell Biology, University of Coimbra, Largo Marquês do Pombal 3004-517 Coimbra, Portugal
| |
Collapse
|
15
|
Dalod M, Scheu S. Dendritic cell functions in vivo: a user's guide to current and next generation mutant mouse models. Eur J Immunol 2022; 52:1712-1749. [PMID: 35099816 DOI: 10.1002/eji.202149513] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2021] [Accepted: 01/14/2022] [Indexed: 11/11/2022]
Abstract
Dendritic cells (DCs) do not just excel in antigen presentation. They orchestrate information transfer from innate to adaptive immunity, by sensing and integrating a variety of danger signals, and translating them to naïve T cells, to mount specifically tailored immune responses. This is accomplished by distinct DC types specialized in different functions and because each DC is functionally plastic, assuming different activation states depending on the input signals received. Mouse models hold the key to untangle this complexity and determine which DC types and activation states contribute to which functions. Here, we aim to provide comprehensive information for selecting the most appropriate mutant mouse strains to address specific research questions on DCs, considering three in vivo experimental approaches: (i) interrogating the roles of DC types through their depletion; (ii) determining the underlying mechanisms by specific genetic manipulations; (iii) deciphering the spatiotemporal dynamics of DC responses. We summarize the advantages, caveats, suggested use and perspectives for a variety of mutant mouse strains, discussing in more detail the most widely used or accurate models. Finally, we discuss innovative strategies to improve targeting specificity, for the next generation mutant mouse models, and briefly address how humanized mouse models can accelerate translation into the clinic. This article is protected by copyright. All rights reserved.
Collapse
Affiliation(s)
- Marc Dalod
- CNRS, Inserm, Aix Marseille Univ, Centre d'Immunologie de Marseille-Luminy (CIML), Turing Center for Living Systems, Marseille, France
| | - Stefanie Scheu
- Institute of Medical Microbiology and Hospital Hygiene, University of Düsseldorf, Düsseldorf, Germany
| |
Collapse
|
16
|
Czeh M, Stäble S, Krämer S, Tepe L, Talyan S, Carrelha J, Meng Y, Heitplatz B, Schwabenland M, Milsom MD, Plass C, Prinz M, Schlesner M, Andrade-Navarro MA, Nerlov C, Jacobsen SEW, Lipka DB, Rosenbauer F. DNMT1 Deficiency Impacts on Plasmacytoid Dendritic Cells in Homeostasis and Autoimmune Disease. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2022; 208:358-370. [PMID: 34903641 PMCID: PMC7612220 DOI: 10.4049/jimmunol.2100624] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/25/2021] [Accepted: 10/28/2021] [Indexed: 01/07/2023]
Abstract
Dendritic cells (DCs) are heterogeneous immune regulators involved in autoimmune diseases. Epigenomic mechanisms orchestrating DC development and DC subset diversification remain insufficiently understood but could be important to modulate DC fate for clinical purposes. By combining whole-genome methylation assessment with the analysis of mice expressing reduced DNA methyltransferase 1 levels, we show that distinct DNA methylation levels and patterns are required for the development of plasmacytoid DC and conventional DC subsets. We provide clonal in vivo evidence for DC lineage establishment at the stem cell level, and we show that a high DNA methylation threshold level is essential for Flt3-dependent survival of DC precursors. Importantly, reducing methylation predominantly depletes plasmacytoid DC and alleviates systemic lupus erythematosus in an autoimmunity mouse model. This study shows how DNA methylation regulates the production of DC subsets and provides a potential rationale for targeting autoimmune disease using hypomethylating agents.
Collapse
Affiliation(s)
- Melinda Czeh
- Institute of Molecular Tumor Biology, University of Münster, Münster, Germany
- MRC Molecular Haematology Unit, MRC Weatherall Institute of Molecular Medicine, John Radcliffe Hospital, University of Oxford, Oxford, UK
| | - Sina Stäble
- Section Translational Cancer Epigenomics, Division of Translational Medical Oncology, German Cancer Research Center and National Center for Tumor Diseases Heidelberg, Heidelberg, Germany
| | - Stephen Krämer
- Section Translational Cancer Epigenomics, Division of Translational Medical Oncology, German Cancer Research Center and National Center for Tumor Diseases Heidelberg, Heidelberg, Germany
- Biomedical Informatics, Data Mining and Data Analytics, Faculty of Applied Computer Science and Medical Faculty, University of Augsburg, Germany
- Bioinformatics and Omics Data Analysis, German Cancer Research Center, Heidelberg, Germany
- Faculty of Biosciences, Heidelberg University, Heidelberg, Germany
| | - Lena Tepe
- Institute of Molecular Tumor Biology, University of Münster, Münster, Germany
| | - Sweta Talyan
- Faculty of Biology, Johannes Gutenberg University of Mainz, Mainz, Germany
| | - Joana Carrelha
- MRC Molecular Haematology Unit, MRC Weatherall Institute of Molecular Medicine, John Radcliffe Hospital, University of Oxford, Oxford, UK
| | - Yiran Meng
- MRC Molecular Haematology Unit, MRC Weatherall Institute of Molecular Medicine, John Radcliffe Hospital, University of Oxford, Oxford, UK
| | - Barbara Heitplatz
- Gerhard-Domagk-Institute of Pathology, University Hospital Münster, University of Münster, Münster, Germany
| | - Marius Schwabenland
- Institute of Neuropathology, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Michael D Milsom
- Division of Experimental Hematology, German Cancer Research Center, Heidelberg, Germany
| | - Christoph Plass
- Division of Cancer Epigenomics, German Cancer Research Center, Heidelberg, Germany
| | - Marco Prinz
- Institute of Neuropathology, Faculty of Medicine, University of Freiburg, Freiburg, Germany
- Centre for Biological Signalling Studies and Centre for Integrative Biological Signalling Studies, University of Freiburg, Freiburg, Germany
- Center for Basics in NeuroModulation, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Matthias Schlesner
- Biomedical Informatics, Data Mining and Data Analytics, Faculty of Applied Computer Science and Medical Faculty, University of Augsburg, Germany
- Bioinformatics and Omics Data Analysis, German Cancer Research Center, Heidelberg, Germany
| | | | - Claus Nerlov
- MRC Molecular Haematology Unit, MRC Weatherall Institute of Molecular Medicine, John Radcliffe Hospital, University of Oxford, Oxford, UK
| | - Sten Eirik W Jacobsen
- MRC Molecular Haematology Unit, MRC Weatherall Institute of Molecular Medicine, John Radcliffe Hospital, University of Oxford, Oxford, UK
- Department of Cell and Molecular Biology and Department of Medicine Huddinge, Center for Hematology and Regenerative Medicine, Karolinska Institutet, Stockholm, Sweden; and
- Karolinska University Hospital, Stockholm, Sweden
| | - Daniel B Lipka
- Section Translational Cancer Epigenomics, Division of Translational Medical Oncology, German Cancer Research Center and National Center for Tumor Diseases Heidelberg, Heidelberg, Germany
| | - Frank Rosenbauer
- Institute of Molecular Tumor Biology, University of Münster, Münster, Germany;
| |
Collapse
|
17
|
Murphy TL, Murphy KM. Dendritic cells in cancer immunology. Cell Mol Immunol 2022; 19:3-13. [PMID: 34480145 PMCID: PMC8752832 DOI: 10.1038/s41423-021-00741-5] [Citation(s) in RCA: 144] [Impact Index Per Article: 48.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2021] [Accepted: 06/22/2021] [Indexed: 02/07/2023] Open
Abstract
The clinical success of immune checkpoint therapy (ICT) has produced explosive growth in tumor immunology research because ICT was discovered through basic studies of immune regulation. Much of the current translational efforts are aimed at enhancing ICT by identifying therapeutic targets that synergize with CTLA4 or PD1/PD-L1 blockade and are solidly developed on the basis of currently accepted principles. Expanding these principles through continuous basic research may help broaden translational efforts. With this mindset, we focused this review on three threads of basic research directly relating to mechanisms underlying ICT. Specifically, this review covers three aspects of dendritic cell (DC) biology connected with antitumor immune responses but are not specifically oriented toward therapeutic use. First, we review recent advances in the development of the cDC1 subset of DCs, identifying important features distinguishing these cells from other types of DCs. Second, we review the antigen-processing pathway called cross-presentation, which was discovered in the mid-1970s and remains an enigma. This pathway serves an essential in vivo function unique to cDC1s and may be both a physiologic bottleneck and therapeutic target. Finally, we review the longstanding field of helper cells and the related area of DC licensing, in which CD4 T cells influence the strength or quality of CD8 T cell responses. Each topic is connected with ICT in some manner but is also a fundamental aspect of cell-mediated immunity directed toward intracellular pathogens.
Collapse
Affiliation(s)
- Theresa L. Murphy
- grid.4367.60000 0001 2355 7002Department of Pathology and Immunology, Washington University in St. Louis, School of Medicine, St. Louis, MO 63110 USA
| | - Kenneth M. Murphy
- grid.4367.60000 0001 2355 7002Department of Pathology and Immunology, Washington University in St. Louis, School of Medicine, St. Louis, MO 63110 USA
| |
Collapse
|
18
|
Chauhan KS, Das A, Jaiswal H, Saha I, Kaushik M, Patel VK, Tailor P. IRF8 and BATF3 interaction enhances the cDC1 specific Pfkfb3 gene expression. Cell Immunol 2021; 371:104468. [PMID: 34968772 DOI: 10.1016/j.cellimm.2021.104468] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2021] [Revised: 11/27/2021] [Accepted: 11/28/2021] [Indexed: 11/03/2022]
Abstract
Dendritic cells (DCs) play central role in innate as well as adaptive immune responses regulated by diverse DC subtypes that vary in terms of surface markers, transcriptional profile and functional responses. Generation of DC diversity from progenitor stage is tightly regulated by complex molecular inter-play between transcription factors. We earlier demonstrated that Batf3 and Id2 expression have a synergistic effect on the Irf8 directed classical cDC1 development. In present study, Bi-molecular fluorescence complementation assay suggested that IRF8 interacts with BATF3, and ID2 may aid cDC1 development independently. Genome wide recruitment analysis of IRF8 and BATF3 from different DC subtypes led to identification of the overlapping regions of occupancy by these two transcription factors. Further analysis of overlapping peaks of IRF8 and BATF3 occupancy in promoter region within the cDC1 subtype specific transcriptional pattern identified a metabolically important Pfkfb3 gene. Among various immune cell types; splenic cDC1 subtype displayed enhanced expression of Pfkfb3. Analysis of Irf8-/-, Irf8R294C and Batf3DCKO DC confirmed direct regulation of Pfkfb3 enhanced expression specifically in cDC1 subtype. Further we show that inhibition of PFKFB3 enzymatic activity by a chemical agent PFK15 led to reduction in cDC1 subtype in both in vitro FLDC cultures as well as in vivo mouse spleens. Together, our study identified the direct regulation of cDC1 specific enhanced expression of Pfkfb3 in glycolysis and cDC1 biology.
Collapse
Affiliation(s)
- Kuldeep Singh Chauhan
- Laboratory of Innate Immunity, National Institute of Immunology, New Delhi, India; Department of Molecular Microbiology, Washington University School of Medicine, St. Louis, MO, USA(1)
| | - Annesa Das
- Laboratory of Innate Immunity, National Institute of Immunology, New Delhi, India
| | - Hemant Jaiswal
- Laboratory of Innate Immunity, National Institute of Immunology, New Delhi, India; Laboratory of Molecular Immunology, National Institute of Allergy and, Infectious Diseases, National Institutes of Health, Bethesda, MD, USA(2)
| | - Irene Saha
- Laboratory of Innate Immunity, National Institute of Immunology, New Delhi, India; Division of Immunobiology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA(3)
| | - Monika Kaushik
- Laboratory of Innate Immunity, National Institute of Immunology, New Delhi, India; School of Biotechnology, Jawaharlal Nehru University, New Delhi, India(4)
| | | | - Prafullakumar Tailor
- Laboratory of Innate Immunity, National Institute of Immunology, New Delhi, India; Special Centre for Systems Medicine (SCSM), Jawaharlal Nehru University, New Delhi, India.
| |
Collapse
|
19
|
Tuong ZK, Stewart BJ, Guo SA, Clatworthy MR. Epigenetics and tissue immunity-Translating environmental cues into functional adaptations. Immunol Rev 2021; 305:111-136. [PMID: 34821397 DOI: 10.1111/imr.13036] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2021] [Revised: 10/26/2021] [Accepted: 10/28/2021] [Indexed: 12/21/2022]
Abstract
There is an increasing appreciation that many innate and adaptive immune cell subsets permanently reside within non-lymphoid organs, playing a critical role in tissue homeostasis and defense. The best characterized are macrophages and tissue-resident T lymphocytes that work in concert with organ structural cells to generate appropriate immune responses and are functionally shaped by organ-specific environmental cues. The interaction of tissue epithelial, endothelial and stromal cells is also required to attract, differentiate, polarize and maintain organ immune cells in their tissue niche. All of these processes require dynamic regulation of cellular transcriptional programmes, with epigenetic mechanisms playing a critical role, including DNA methylation and post-translational histone modifications. A failure to appropriately regulate immune cell transcription inevitably results in inadequate or inappropriate immune responses and organ pathology. Here, with a focus on the mammalian kidney, an organ which generates differing regional environmental cues (including hypersalinity and hypoxia) due to its physiological functions, we will review the basic concepts of tissue immunity, discuss the technologies available to profile epigenetic modifications in tissue immune cells, including those that enable single-cell profiling, and consider how these mechanisms influence the development, phenotype, activation and function of different tissue immune cell subsets, as well as the immunological function of structural cells.
Collapse
Affiliation(s)
- Zewen Kelvin Tuong
- Molecular Immunity Unit, Department of Medicine, MRC-Laboratory of Molecular Biology, University of Cambridge, Cambridge, UK.,Cellular Genetics, Wellcome Sanger Institute, Hinxton, UK
| | - Benjamin J Stewart
- Molecular Immunity Unit, Department of Medicine, MRC-Laboratory of Molecular Biology, University of Cambridge, Cambridge, UK.,Cellular Genetics, Wellcome Sanger Institute, Hinxton, UK
| | - Shuang Andrew Guo
- Molecular Immunity Unit, Department of Medicine, MRC-Laboratory of Molecular Biology, University of Cambridge, Cambridge, UK.,Cellular Genetics, Wellcome Sanger Institute, Hinxton, UK
| | - Menna R Clatworthy
- Molecular Immunity Unit, Department of Medicine, MRC-Laboratory of Molecular Biology, University of Cambridge, Cambridge, UK.,Cellular Genetics, Wellcome Sanger Institute, Hinxton, UK.,Cambridge Institute of Therapeutic Immunology and Infectious Diseases, University of Cambridge, Cambridge, UK
| |
Collapse
|
20
|
Zhang S, Chopin M, Nutt SL. Type 1 conventional dendritic cells: ontogeny, function, and emerging roles in cancer immunotherapy. Trends Immunol 2021; 42:1113-1127. [PMID: 34728143 DOI: 10.1016/j.it.2021.10.004] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2021] [Revised: 10/07/2021] [Accepted: 10/08/2021] [Indexed: 12/12/2022]
Abstract
Dendritic cells (DCs) are key immune sentinels that orchestrate protective immune responses against pathogens or cancers. DCs have evolved into multiple phenotypically, anatomically, and functionally distinct cell types. One of these DC types, Type 1 conventional DCs (cDC1s), are uniquely equipped to promote cytotoxic CD8+ T cell differentiation and, therefore, represent a promising target for harnessing antitumor immunity. Indeed, recent studies have highlighted the importance of cDC1s in tumor immunotherapy using immune checkpoint inhibitors. Here, we review the progress in defining the key developmental and functional attributes of cDC1s and the approaches to optimizing the potency of cDC1s for anticancer immunity.
Collapse
Affiliation(s)
- Shengbo Zhang
- The Walter and Eliza Hall Institute of Medical Research, 1G Royal Parade, Parkville, VIC 3052, Australia; Department of Medical Biology, University of Melbourne, Parkville, VIC 3010, Australia
| | - Michaël Chopin
- The Walter and Eliza Hall Institute of Medical Research, 1G Royal Parade, Parkville, VIC 3052, Australia; Department of Medical Biology, University of Melbourne, Parkville, VIC 3010, Australia.
| | - Stephen L Nutt
- The Walter and Eliza Hall Institute of Medical Research, 1G Royal Parade, Parkville, VIC 3052, Australia; Department of Medical Biology, University of Melbourne, Parkville, VIC 3010, Australia.
| |
Collapse
|
21
|
Zhang P, Schlecht A, Wolf J, Boneva S, Laich Y, Koch J, Ludwig F, Boeck M, Thien A, Härdtner C, Kierdorf K, Agostini H, Schlunck G, Prinz M, Hilgendorf I, Wieghofer P, Lange C. The role of interferon regulatory factor 8 for retinal tissue homeostasis and development of choroidal neovascularisation. J Neuroinflammation 2021; 18:215. [PMID: 34544421 PMCID: PMC8454118 DOI: 10.1186/s12974-021-02230-y] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2021] [Accepted: 08/04/2021] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Microglia cells represent the resident innate immune cells of the retina and are important for retinal development and tissue homeostasis. However, dysfunctional microglia can have a negative impact on the structural and functional integrity of the retina under native and pathological conditions. METHODS In this study, we examined interferon-regulatory factor 8 (Irf8)-deficient mice to determine the transcriptional profile, morphology, and temporospatial distribution of microglia lacking Irf8 and to explore the effects on retinal development, tissue homeostasis, and formation of choroidal neovascularisation (CNV). RESULTS Our study shows that Irf8-deficient MG exhibit a considerable loss of microglial signature genes accompanied by a severely altered MG morphology. An in-depth characterisation by fundus photography, fluorescein angiography, optical coherence tomography and electroretinography revealed no major retinal abnormalities during steady state. However, in the laser-induced CNV model, Irf8-deficient microglia showed an increased activity of biological processes critical for inflammation and cell adhesion and a reduced MG cell density near the lesions, which was associated with significantly increased CNV lesion size. CONCLUSIONS Our results suggest that loss of Irf8 in microglia has negligible effects on retinal homeostasis in the steady state. However, under pathological conditions, Irf8 is crucial for the transformation of resident microglia into a reactive phenotype and thus for the suppression of retinal inflammation and CNV formation.
Collapse
Affiliation(s)
- Peipei Zhang
- Medical Faculty, Eye Center, University Hospital, University of Freiburg, Freiburg im Breisgau, Germany
| | - Anja Schlecht
- Medical Faculty, Eye Center, University Hospital, University of Freiburg, Freiburg im Breisgau, Germany.,Institute of Anatomy, Wuerzburg University, Wuerzburg, Germany
| | - Julian Wolf
- Medical Faculty, Eye Center, University Hospital, University of Freiburg, Freiburg im Breisgau, Germany
| | - Stefaniya Boneva
- Medical Faculty, Eye Center, University Hospital, University of Freiburg, Freiburg im Breisgau, Germany
| | - Yannik Laich
- Medical Faculty, Eye Center, University Hospital, University of Freiburg, Freiburg im Breisgau, Germany
| | - Jana Koch
- Medical Faculty, Eye Center, University Hospital, University of Freiburg, Freiburg im Breisgau, Germany
| | - Franziska Ludwig
- Medical Faculty, Eye Center, University Hospital, University of Freiburg, Freiburg im Breisgau, Germany
| | - Myriam Boeck
- Medical Faculty, Eye Center, University Hospital, University of Freiburg, Freiburg im Breisgau, Germany
| | - Adrian Thien
- Medical Faculty, Eye Center, University Hospital, University of Freiburg, Freiburg im Breisgau, Germany
| | - Carmen Härdtner
- Cardiology and Angiology, University Heart Center, University of Freiburg, Freiburg im Breisgau, Germany.,Medical Center and Faculty of Medicine, Institute for Experimental Cardiovascular Medicine, University Heart Center Freiburg Bad Krozingen, University of Freiburg, Freiburg, Germany
| | - Katrin Kierdorf
- Medical Faculty, Institute of Neuropathology, University Hospital, University of Freiburg, Freiburg im Breisgau, Germany.,CIBSS-Centre for Integrative Biological Signalling Studies, University of Freiburg, Freiburg, Germany.,Medical Faculty, Center for Basics in NeuroModulation (NeuroModulBasics), University of Freiburg, Freiburg im Breisgau, Germany
| | - Hansjürgen Agostini
- Medical Faculty, Eye Center, University Hospital, University of Freiburg, Freiburg im Breisgau, Germany
| | - Günther Schlunck
- Medical Faculty, Eye Center, University Hospital, University of Freiburg, Freiburg im Breisgau, Germany
| | - Marco Prinz
- Medical Faculty, Institute of Neuropathology, University Hospital, University of Freiburg, Freiburg im Breisgau, Germany.,Medical Faculty, Center for Basics in NeuroModulation (NeuroModulBasics), University of Freiburg, Freiburg im Breisgau, Germany.,Signalling Research Centres BIOSS and CIBSS, University of Freiburg, Freiburg im Breisgau, Germany
| | - Ingo Hilgendorf
- Cardiology and Angiology, University Heart Center, University of Freiburg, Freiburg im Breisgau, Germany.,Medical Center and Faculty of Medicine, Institute for Experimental Cardiovascular Medicine, University Heart Center Freiburg Bad Krozingen, University of Freiburg, Freiburg, Germany
| | - Peter Wieghofer
- Medical Faculty, Institute of Neuropathology, University Hospital, University of Freiburg, Freiburg im Breisgau, Germany. .,Institute of Anatomy, Leipzig University, Leipzig, Germany.
| | - Clemens Lange
- Medical Faculty, Eye Center, University Hospital, University of Freiburg, Freiburg im Breisgau, Germany.
| |
Collapse
|
22
|
Zhang S, Coughlan HD, Ashayeripanah M, Seizova S, Kueh AJ, Brown DV, Cao W, Jacquelot N, D'Amico A, Lew AM, Zhan Y, Tonkin CJ, Villadangos JA, Smyth GK, Chopin M, Nutt SL. Type 1 conventional dendritic cell fate and function are controlled by DC-SCRIPT. Sci Immunol 2021; 6:6/58/eabf4432. [PMID: 33811060 DOI: 10.1126/sciimmunol.abf4432] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2020] [Accepted: 03/03/2021] [Indexed: 12/13/2022]
Abstract
The functional diversification of dendritic cells (DCs) is a key step in establishing protective immune responses. Despite the importance of DC lineage diversity, its genetic basis is not fully understood. The transcription factor DC-SCRIPT is expressed in conventional DCs (cDCs) and their committed bone marrow progenitors but not in plasmacytoid DCs (pDCs). We show that mice lacking DC-SCRIPT displayed substantially impaired development of IRF8 (interferon regulatory factor 8)-dependent cDC1, whereas cDC2 numbers increased marginally. The residual DC-SCRIPT-deficient cDC1s had impaired capacity to capture and present cell-associated antigens and to secrete IL-12p40, two functional hallmarks of this population. Genome-wide mapping of DC-SCRIPT binding and gene expression analyses revealed a key role for DC-SCRIPT in maintaining cDC1 identity via the direct regulation of cDC1 signature genes, including Irf8 Our study reveals DC-SCRIPT to be a critical component of the gene regulatory program shaping the functional attributes of cDC1s.
Collapse
Affiliation(s)
- Shengbo Zhang
- Walter and Eliza Hall Institute of Medical Research, 1G Royal Parade, Parkville, VIC 3052, Australia.,Department of Medical Biology, University of Melbourne, Parkville, VIC 3010, Australia
| | - Hannah D Coughlan
- Walter and Eliza Hall Institute of Medical Research, 1G Royal Parade, Parkville, VIC 3052, Australia.,Department of Medical Biology, University of Melbourne, Parkville, VIC 3010, Australia
| | - Mitra Ashayeripanah
- Department of Microbiology and Immunology, University of Melbourne at Peter Doherty Institute of Infection and Immunity, Melbourne, VIC 3010, Australia
| | - Simona Seizova
- Walter and Eliza Hall Institute of Medical Research, 1G Royal Parade, Parkville, VIC 3052, Australia.,Department of Medical Biology, University of Melbourne, Parkville, VIC 3010, Australia
| | - Andrew J Kueh
- Walter and Eliza Hall Institute of Medical Research, 1G Royal Parade, Parkville, VIC 3052, Australia.,Department of Medical Biology, University of Melbourne, Parkville, VIC 3010, Australia
| | - Daniel V Brown
- Walter and Eliza Hall Institute of Medical Research, 1G Royal Parade, Parkville, VIC 3052, Australia.,Department of Medical Biology, University of Melbourne, Parkville, VIC 3010, Australia
| | - Wang Cao
- Walter and Eliza Hall Institute of Medical Research, 1G Royal Parade, Parkville, VIC 3052, Australia.,Department of Medical Biology, University of Melbourne, Parkville, VIC 3010, Australia
| | - Nicolas Jacquelot
- Walter and Eliza Hall Institute of Medical Research, 1G Royal Parade, Parkville, VIC 3052, Australia.,Department of Medical Biology, University of Melbourne, Parkville, VIC 3010, Australia
| | - Angela D'Amico
- Walter and Eliza Hall Institute of Medical Research, 1G Royal Parade, Parkville, VIC 3052, Australia.,Department of Medical Biology, University of Melbourne, Parkville, VIC 3010, Australia
| | - Andrew M Lew
- Walter and Eliza Hall Institute of Medical Research, 1G Royal Parade, Parkville, VIC 3052, Australia.,Department of Medical Biology, University of Melbourne, Parkville, VIC 3010, Australia
| | - Yifan Zhan
- Walter and Eliza Hall Institute of Medical Research, 1G Royal Parade, Parkville, VIC 3052, Australia.,Department of Medical Biology, University of Melbourne, Parkville, VIC 3010, Australia.,Drug Discovery, Shanghai Huaota Biopharmaceutical Co. Ltd., Shanghai, China
| | - Christopher J Tonkin
- Walter and Eliza Hall Institute of Medical Research, 1G Royal Parade, Parkville, VIC 3052, Australia.,Department of Medical Biology, University of Melbourne, Parkville, VIC 3010, Australia
| | - Jose A Villadangos
- Department of Microbiology and Immunology, University of Melbourne at Peter Doherty Institute of Infection and Immunity, Melbourne, VIC 3010, Australia.,Department of Biochemistry and Molecular Biology, Bio21 Molecular Science and Biotechnology Institute, University of Melbourne, Parkville, VIC, Australia
| | - Gordon K Smyth
- Walter and Eliza Hall Institute of Medical Research, 1G Royal Parade, Parkville, VIC 3052, Australia.,School of Mathematics and Statistics, University of Melbourne, Parkville, VIC 3010, Australia
| | - Michaël Chopin
- Walter and Eliza Hall Institute of Medical Research, 1G Royal Parade, Parkville, VIC 3052, Australia. .,Department of Medical Biology, University of Melbourne, Parkville, VIC 3010, Australia
| | - Stephen L Nutt
- Walter and Eliza Hall Institute of Medical Research, 1G Royal Parade, Parkville, VIC 3052, Australia. .,Department of Medical Biology, University of Melbourne, Parkville, VIC 3010, Australia
| |
Collapse
|
23
|
Johnson KD, Conn DJ, Shishkova E, Katsumura KR, Liu P, Shen S, Ranheim EA, Kraus SG, Wang W, Calvo KR, Hsu AP, Holland SM, Coon JJ, Keles S, Bresnick EH. Constructing and deconstructing GATA2-regulated cell fate programs to establish developmental trajectories. J Exp Med 2021; 217:151996. [PMID: 32736380 PMCID: PMC7596813 DOI: 10.1084/jem.20191526] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2019] [Revised: 02/08/2020] [Accepted: 06/18/2020] [Indexed: 12/13/2022] Open
Abstract
Stem and progenitor cell fate transitions constitute key decision points in organismal development that enable access to a developmental path or actively preclude others. Using the hematopoietic system, we analyzed the relative importance of cell fate–promoting mechanisms versus negating fate-suppressing mechanisms to engineer progenitor cells with multilineage differentiation potential. Deletion of the murine Gata2−77 enhancer, with a human equivalent that causes leukemia, downregulates the transcription factor GATA2 and blocks progenitor differentiation into erythrocytes, megakaryocytes, basophils, and granulocytes, but not macrophages. Using multiomics and single-cell analyses, we demonstrated that the enhancer orchestrates a balance between pro- and anti-fate circuitry in single cells. By increasing GATA2 expression, the enhancer instigates a fate-promoting mechanism while abrogating an innate immunity–linked, fate-suppressing mechanism. During embryogenesis, the suppressing mechanism dominated in enhancer mutant progenitors, thus yielding progenitors with a predominant monocytic differentiation potential. Coordinating fate-promoting and -suppressing circuits therefore averts deconstruction of a multifate system into a monopotent system and maintains critical progenitor heterogeneity and functionality.
Collapse
Affiliation(s)
- Kirby D Johnson
- University of Wisconsin-Madison Blood Research Program, Department of Cell and Regenerative Biology, Wisconsin Institutes for Medical Research, University of Wisconsin School of Medicine and Public Health, Madison, WI
| | - Daniel J Conn
- Department of Biostatistics and Medical Informatics, University of Wisconsin School of Medicine and Public Health, Madison, WI
| | - Evgenia Shishkova
- Department of Biomolecular Chemistry, University of Wisconsin School of Medicine and Public Health, Madison, WI
| | - Koichi R Katsumura
- University of Wisconsin-Madison Blood Research Program, Department of Cell and Regenerative Biology, Wisconsin Institutes for Medical Research, University of Wisconsin School of Medicine and Public Health, Madison, WI
| | - Peng Liu
- University of Wisconsin Carbone Cancer Center, Department of Biostatistics and Medical Informatics, University of Wisconsin School of Medicine and Public Health, Madison, WI
| | - Siqi Shen
- Department of Statistics, University of Wisconsin, Madison, WI
| | - Erik A Ranheim
- Department of Pathology and Laboratory Medicine, University of Wisconsin School of Medicine and Public Health, Madison, WI
| | - Sean G Kraus
- University of Wisconsin-Madison Blood Research Program, Department of Cell and Regenerative Biology, Wisconsin Institutes for Medical Research, University of Wisconsin School of Medicine and Public Health, Madison, WI
| | - Weixin Wang
- Department of Laboratory Medicine, Clinical Center, National Institutes of Health, Bethesda, MD
| | - Katherine R Calvo
- Department of Laboratory Medicine, Clinical Center, National Institutes of Health, Bethesda, MD
| | - Amy P Hsu
- Laboratory of Clinical Immunology and Microbiology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD
| | - Steven M Holland
- Laboratory of Clinical Immunology and Microbiology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD
| | - Joshua J Coon
- Department of Biomolecular Chemistry, University of Wisconsin School of Medicine and Public Health, Madison, WI
| | - Sunduz Keles
- Department of Biostatistics and Medical Informatics, University of Wisconsin School of Medicine and Public Health, Madison, WI
| | - Emery H Bresnick
- University of Wisconsin-Madison Blood Research Program, Department of Cell and Regenerative Biology, Wisconsin Institutes for Medical Research, University of Wisconsin School of Medicine and Public Health, Madison, WI
| |
Collapse
|
24
|
Murakami K, Sasaki H, Nishiyama A, Kurotaki D, Kawase W, Ban T, Nakabayashi J, Kanzaki S, Sekita Y, Nakajima H, Ozato K, Kimura T, Tamura T. A RUNX-CBFβ-driven enhancer directs the Irf8 dose-dependent lineage choice between DCs and monocytes. Nat Immunol 2021; 22:301-311. [PMID: 33603226 DOI: 10.1038/s41590-021-00871-y] [Citation(s) in RCA: 37] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2020] [Accepted: 01/11/2021] [Indexed: 01/30/2023]
Abstract
The transcription factor IRF8 is essential for the development of monocytes and dendritic cells (DCs), whereas it inhibits neutrophilic differentiation. It is unclear how Irf8 expression is regulated and how this single transcription factor supports the generation of both monocytes and DCs. Here, we identified a RUNX-CBFβ-driven enhancer 56 kb downstream of the Irf8 transcription start site. Deletion of this enhancer in vivo significantly decreased Irf8 expression throughout the myeloid lineage from the progenitor stages, thus resulting in loss of common DC progenitors and overproduction of Ly6C+ monocytes. We demonstrated that high, low or null expression of IRF8 in hematopoietic progenitor cells promotes differentiation toward type 1 conventional DCs, Ly6C+ monocytes or neutrophils, respectively, via epigenetic regulation of distinct sets of enhancers in cooperation with other transcription factors. Our results illustrate the mechanism through which IRF8 controls the lineage choice in a dose-dependent manner within the myeloid cell system.
Collapse
Affiliation(s)
- Koichi Murakami
- Department of Immunology, Yokohama City University Graduate School of Medicine, Kanagawa, Japan
- Advanced Medical Research Center, Yokohama City University, Kanagawa, Japan
| | - Haruka Sasaki
- Department of Immunology, Yokohama City University Graduate School of Medicine, Kanagawa, Japan
| | - Akira Nishiyama
- Department of Immunology, Yokohama City University Graduate School of Medicine, Kanagawa, Japan.
| | - Daisuke Kurotaki
- Department of Immunology, Yokohama City University Graduate School of Medicine, Kanagawa, Japan
| | - Wataru Kawase
- Department of Immunology, Yokohama City University Graduate School of Medicine, Kanagawa, Japan
| | - Tatsuma Ban
- Department of Immunology, Yokohama City University Graduate School of Medicine, Kanagawa, Japan
| | - Jun Nakabayashi
- Advanced Medical Research Center, Yokohama City University, Kanagawa, Japan
| | - Satoko Kanzaki
- Laboratory of Stem Cell Biology, Department of Biosciences, Kitasato University School of Science, Kanagawa, Japan
| | - Yoichi Sekita
- Laboratory of Stem Cell Biology, Department of Biosciences, Kitasato University School of Science, Kanagawa, Japan
| | - Hideaki Nakajima
- Department of Stem Cell and Immune Regulation, Yokohama City University Graduate School of Medicine, Kanagawa, Japan
| | - Keiko Ozato
- Program in Genomics of Differentiation, Eunice Kennedy Shriver National Institute of Child Health and Human Development, NIH, Bethesda, MD, USA
| | - Tohru Kimura
- Laboratory of Stem Cell Biology, Department of Biosciences, Kitasato University School of Science, Kanagawa, Japan
| | - Tomohiko Tamura
- Department of Immunology, Yokohama City University Graduate School of Medicine, Kanagawa, Japan.
- Advanced Medical Research Center, Yokohama City University, Kanagawa, Japan.
| |
Collapse
|
25
|
Abstract
The innate immune response is a rapid response to pathogens or danger signals. It is precisely activated not only to efficiently eliminate pathogens but also to avoid excessive inflammation and tissue damage. cis-Regulatory element-associated chromatin architecture shaped by epigenetic factors, which we define as the epiregulome, endows innate immune cells with specialized phenotypes and unique functions by establishing cell-specific gene expression patterns, and it also contributes to resolution of the inflammatory response. In this review, we focus on two aspects: (a) how niche signals during lineage commitment or following infection and pathogenic stress program epiregulomes by regulating gene expression levels, enzymatic activities, or gene-specific targeting of chromatin modifiers and (b) how the programed epiregulomes in turn mediate regulation of gene-specific expression, which contributes to controlling the development of innate cells, or the response to infection and inflammation, in a timely manner. We also discuss the effects of innate immunometabolic rewiring on epiregulomes and speculate on several future challenges to be encountered during the exploration of the master regulators of epiregulomes in innate immunity and inflammation.
Collapse
Affiliation(s)
- Qian Zhang
- Department of Immunology, Center for Immunotherapy, Institute of Basic Medical Sciences, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing 100005, China; , .,National Key Laboratory of Medical Immunology, Institute of Immunology, Navy Military Medical University, Shanghai 200433, China
| | - Xuetao Cao
- Department of Immunology, Center for Immunotherapy, Institute of Basic Medical Sciences, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing 100005, China; , .,National Key Laboratory of Medical Immunology, Institute of Immunology, Navy Military Medical University, Shanghai 200433, China.,Laboratory of Immunity and Inflammation, College of Life Sciences, Nankai University, Tianjin 300071, China
| |
Collapse
|
26
|
Anderson DA, Dutertre CA, Ginhoux F, Murphy KM. Genetic models of human and mouse dendritic cell development and function. Nat Rev Immunol 2021; 21:101-115. [PMID: 32908299 PMCID: PMC10955724 DOI: 10.1038/s41577-020-00413-x] [Citation(s) in RCA: 191] [Impact Index Per Article: 47.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/28/2020] [Indexed: 12/13/2022]
Abstract
Dendritic cells (DCs) develop in the bone marrow from haematopoietic progenitors that have numerous shared characteristics between mice and humans. Human counterparts of mouse DC progenitors have been identified by their shared transcriptional signatures and developmental potential. New findings continue to revise models of DC ontogeny but it is well accepted that DCs can be divided into two main functional groups. Classical DCs include type 1 and type 2 subsets, which can detect different pathogens, produce specific cytokines and present antigens to polarize mainly naive CD8+ or CD4+ T cells, respectively. By contrast, the function of plasmacytoid DCs is largely innate and restricted to the detection of viral infections and the production of type I interferon. Here, we discuss genetic models of mouse DC development and function that have aided in correlating ontogeny with function, as well as how these findings can be translated to human DCs and their progenitors.
Collapse
Affiliation(s)
- David A Anderson
- Department of Pathology and Immunology, School of Medicine, Washington University in St. Louis, St. Louis, MO, USA
| | | | - Florent Ginhoux
- Singapore Immunology Network, Agency for Science, Technology and Research, Singapore, Singapore
- Shanghai Institute of Immunology, Department of Immunology and Microbiology, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Translational Immunology Institute, SingHealth Duke-NUS Academic Medical Centre, Singapore, Singapore
| | - Kenneth M Murphy
- Department of Pathology and Immunology, School of Medicine, Washington University in St. Louis, St. Louis, MO, USA.
- Howard Hughes Medical Institute, School of Medicine, Washington University in St. Louis, St. Louis, MO, USA.
| |
Collapse
|
27
|
Robinson A, Han CZ, Glass CK, Pollard JW. Monocyte Regulation in Homeostasis and Malignancy. Trends Immunol 2021; 42:104-119. [PMID: 33446416 PMCID: PMC7877795 DOI: 10.1016/j.it.2020.12.001] [Citation(s) in RCA: 90] [Impact Index Per Article: 22.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2020] [Revised: 11/27/2020] [Accepted: 12/05/2020] [Indexed: 12/14/2022]
Abstract
Monocytes are progenitors to macrophages and a subclass of dendritic cells (monocyte-derived dendritic cells, MoDCs), but they also act as circulating sensors that respond to environmental changes and disease. Technological advances have defined the production of classical monocytes in the bone marrow through the identification of lineage-determining transcription factors (LDTFs) and have proposed alternative routes of differentiation. Monocytes released into the circulation can be recruited to tissues by specific chemoattractants where they respond to sequential niche-specific signals that determine their differentiation into terminal effector cells. New aspects of monocyte biology in the circulation are being revealed, exemplified by the influence of cancer on the systemic alteration of monocyte subset abundance and transcriptional profiles. These changes can act to enhance the metastatic spread of primary cancers and may offer therapeutic opportunities.
Collapse
Affiliation(s)
- Amy Robinson
- Medical Research Council (MRC) Centre for Reproductive Health, Queen's Medical Research Institute, University of Edinburgh, Edinburgh EH16 4TJ, UK
| | - Claudia Z Han
- Department of Cellular and Molecular Medicine, School of Medicine, University of California San Diego, La Jolla, CA, 92037, USA
| | - Christopher K Glass
- Department of Cellular and Molecular Medicine, School of Medicine, University of California San Diego, La Jolla, CA, 92037, USA
| | - Jeffrey W Pollard
- Medical Research Council (MRC) Centre for Reproductive Health, Queen's Medical Research Institute, University of Edinburgh, Edinburgh EH16 4TJ, UK.
| |
Collapse
|
28
|
Chauvistré H, Seré K. Epigenetic aspects of DC development and differentiation. Mol Immunol 2020; 128:116-124. [PMID: 33126080 DOI: 10.1016/j.molimm.2020.10.011] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2019] [Revised: 09/09/2020] [Accepted: 10/14/2020] [Indexed: 02/06/2023]
Abstract
In this review we introduce the basic principles of epigenetic gene regulation and discuss them in the context of dendritic cell (DC) development and differentiation. Epigenetic mechanisms control the accessibility of chromatin for DNA binding proteins and thus they control gene expression. These mechanisms comprise chemical modifications of DNA and histones, chromatin remodeling and chromatin conformation. The variety of epigenetic mechanisms allow high-end fine tuning and flexibility of gene expression, a prerequisite in the process of DC lineage development.
Collapse
Affiliation(s)
- Heike Chauvistré
- Department of Dermatology, University Hospital Essen, West German Cancer Center, University Duisburg-Essen and the German Cancer Consortium (DKTK), Essen, Germany
| | - Kristin Seré
- Institute of Biomedical Engineering, Department of Cell Biology, RWTH Aachen University Medical School, Helmholtz Institute for Biomedical Engineering, RWTH Aachen University, Aachen, Germany.
| |
Collapse
|
29
|
Kurotaki D, Yoshida H, Tamura T. Epigenetic and transcriptional regulation of osteoclast differentiation. Bone 2020; 138:115471. [PMID: 32526404 DOI: 10.1016/j.bone.2020.115471] [Citation(s) in RCA: 61] [Impact Index Per Article: 12.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/31/2020] [Revised: 06/04/2020] [Accepted: 06/04/2020] [Indexed: 01/28/2023]
Abstract
Osteoclasts are derived from mononuclear phagocyte lineage cells and are indispensable for bone resorption. Recent findings suggest that fetal yolk sac macrophage progenitors give rise to neonatal osteoclasts, while hematopoietic stem cell-derived cells, such as monocytes, contribute to maintaining osteoclast syncytia in vivo. Osteoclast differentiation is dependent on macrophage colony-stimulating factor (M-CSF) and receptor activator of nuclear factor-κB ligand (RANKL) signaling that mediates global epigenetic and transcriptional changes. PU.1 is a transcription factor that establishes cell type-specific enhancer landscapes in osteoclast precursors and mature osteoclasts by collaborating with interferon regulatory factor-8 (IRF8) and nuclear factor of activated T-cells (NFATc1), respectively. Irf8 and Nfatc1 genes are tightly controlled by epigenetic mechanisms such as DNA methylation and histone modifications during osteoclastogenesis. Thus, key transcription factors orchestrate osteoclast-specific transcription regulatory networks through epigenetic modifications. In this review, we discuss recent advances in our understanding of the molecular mechanisms involved in osteoclast development.
Collapse
Affiliation(s)
- Daisuke Kurotaki
- Department of Immunology, Yokohama City University Graduate School of Medicine, Yokohama 236-0004, Japan.
| | - Haruka Yoshida
- Department of Immunology, Yokohama City University Graduate School of Medicine, Yokohama 236-0004, Japan
| | - Tomohiko Tamura
- Department of Immunology, Yokohama City University Graduate School of Medicine, Yokohama 236-0004, Japan
| |
Collapse
|
30
|
Kramer ED, Abrams SI. Granulocytic Myeloid-Derived Suppressor Cells as Negative Regulators of Anticancer Immunity. Front Immunol 2020; 11:1963. [PMID: 32983128 PMCID: PMC7481329 DOI: 10.3389/fimmu.2020.01963] [Citation(s) in RCA: 38] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2020] [Accepted: 07/21/2020] [Indexed: 12/24/2022] Open
Abstract
The immune system plays a critical role in cancer progression and response to therapy. However, the immune system can be compromised during the neoplastic process. Notably, the myeloid lineage, which gives rise to granulocytic cells, including neutrophils, is a well-recognized target of tumor-mediated immune suppression. Ordinarily, granulocytic cells are integral for host defense, but in neoplasia the normal process of granulocyte differentiation (i.e., granulopoiesis) can be impaired leading instead to the formation of granulocytic (or PMN)-myeloid-derived suppressor cells (MDSCs). Such cells comprise various stages of myeloid differentiation and are defined functionally by their highly pro-tumorigenic and immune suppressive activities. Thus, considerable interest has been devoted to impeding the negative contributions of PMN-MDSCs to the antitumor response. Understanding their biology has the potential to unveil novel therapeutic opportunities to hamper PMN-MDSC production in the bone marrow, their mobilization, or their effector functions within the tumor microenvironment and, therefore, bolster anticancer therapies that require a competent myeloid compartment. In this review, we will highlight mechanisms by which the neoplastic process skews granulopoiesis to produce PMN-MDSCs, summarize mechanisms by which they execute their pro-tumorigenic activities and, lastly, underscore strategies to obstruct their role as negative regulators of antitumor immunity.
Collapse
Affiliation(s)
- Elliot D Kramer
- Department of Immunology, Roswell Park Comprehensive Cancer Center, Buffalo, NY, United States
| | - Scott I Abrams
- Department of Immunology, Roswell Park Comprehensive Cancer Center, Buffalo, NY, United States
| |
Collapse
|
31
|
De N, Nandi M, Banik S, Roy S. Bi-stability of the master gene regulatory network of the common dendritic precursor cell: Implications for cell differentiation. IUBMB Life 2020; 72:2225-2232. [PMID: 32790022 DOI: 10.1002/iub.2355] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2020] [Accepted: 07/10/2020] [Indexed: 11/08/2022]
Abstract
In cell lineage commitment decisions, a gene regulatory network (GRN) consisting of a limited number of transcription factors forms the regulatory pivot. Myeloid lineage dendritic cells or DCs are specialized cells having the antigen-presenting ability and are of immense importance in immune surveillance. In this report, we analyze the GRN that governs the lineage commitment of Common DC Progenitor (CDP) cells to conventional dendritic cells (cDC) and plasmacytoid dendritic cells (pDC). We have analyzed the quantitative behavior of the master regulatory circuit of CDP that governs the lineage commitment. Simulations showed that the GRN displays a bi-stable behavior within a range of parameter values. Several transcription factors, PU.1, IRF8, Flt3, and Stat3, whose concentrations vary significantly in the two steady states, appear to be the key players. We hypothesize that the two stable steady states are precursors of cDC and pDC, and the variation of concentration of these key transcription factors in the two states may be responsible for early events in lineage commitment.
Collapse
Affiliation(s)
- Nayan De
- Department of Biophysics, Bose Institute, Kolkata, India
| | - Mintu Nandi
- Department of Chemistry, University of Calcutta, Kolkata, India
| | - Suman Banik
- Department of Chemistry, Bose Institute, Kolkata, India
| | - Siddhartha Roy
- Department of Biophysics, Bose Institute, Kolkata, India
| |
Collapse
|
32
|
Fourier N, Zolty M, Azriel A, Tedesco D, Levi BZ. MafK Mediates Chromatin Remodeling to Silence IRF8 Expression in Non-immune Cells in a Cell Type-SpecificManner. J Mol Biol 2020; 432:4544-4560. [PMID: 32534063 DOI: 10.1016/j.jmb.2020.06.005] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2020] [Revised: 06/02/2020] [Accepted: 06/05/2020] [Indexed: 11/17/2022]
Abstract
The regulation of gene expression is a result of a complex interplay between chromatin remodeling, transcription factors, and signaling molecules. Cell differentiation is accompanied by chromatin remodeling of specific loci to permanently silence genes that are not essential for the differentiated cell activity. The molecular cues that recruit the chromatin remodeling machinery are not well characterized. IRF8 is an immune-cell specific transcription factor and its expression is augmented by interferon-γ. Therefore, it serves as a model gene to elucidate the molecular mechanisms governing its silencing in non-immune cells. Ahigh-throughput shRNA library screen in IRF8 expression-restrictive cells enabled the identification of MafK as modulator of IRF8 silencing, affecting chromatin architecture. ChIP-Seq analysis revealed three MafK binding regions (-25 kb, -20 kb, and IRF8 6th intron) within the IRF8 locus. These MafK binding sites are sufficient to repress a reporter gene when cloned in genome-integrated lentiviral reporter constructs in only expression-restrictive cells. Conversely, plasmid-based constructs do not demonstrate such repressive effect. These results highlight the role of these MafK binding sites in mediating repressed chromatin assembly. Finally, a more thorough genomic analysis was performed, using CRISPR-Cas9 to delete MafK-int6 binding region in IRF8 expression-restrictive cells. Deleted clones exhibited an accessible chromatin conformation within the IRF8 locus that was accompanied by a significant increase in basal expression of IRF8 that was further induced by interferon-γ. Taken together, we identified and characterized several MafK binding elements within the IRF8 locus that mediate repressive chromatin conformation resulting in the silencing of IRF8 expression in a celltype-specific manner.
Collapse
Affiliation(s)
- Nitsan Fourier
- Department of Biotechnology and Food Engineering, Technion-Israel Institute of Technology, Haifa, Israel
| | - Maya Zolty
- Department of Biotechnology and Food Engineering, Technion-Israel Institute of Technology, Haifa, Israel
| | - Aviva Azriel
- Department of Biotechnology and Food Engineering, Technion-Israel Institute of Technology, Haifa, Israel
| | | | - Ben-Zion Levi
- Department of Biotechnology and Food Engineering, Technion-Israel Institute of Technology, Haifa, Israel.
| |
Collapse
|
33
|
Nutt SL, Chopin M. Transcriptional Networks Driving Dendritic Cell Differentiation and Function. Immunity 2020; 52:942-956. [DOI: 10.1016/j.immuni.2020.05.005] [Citation(s) in RCA: 110] [Impact Index Per Article: 22.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2020] [Revised: 04/23/2020] [Accepted: 05/15/2020] [Indexed: 12/13/2022]
|
34
|
Wang H, Morse HC, Bolland S. Transcriptional Control of Mature B Cell Fates. Trends Immunol 2020; 41:601-613. [PMID: 32446878 DOI: 10.1016/j.it.2020.04.011] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2019] [Revised: 04/28/2020] [Accepted: 04/30/2020] [Indexed: 02/05/2023]
Abstract
The mature naïve B cell repertoire consists of three well-defined populations: B1, B2 (follicular B, FOB), and marginal zone B (MZB) cells. FOB cells are the dominant mature B cell population in the secondary lymphoid organs and blood of both humans and mice. The driving forces behind mature B lineage selection have been linked to B cell receptor (BCR) signaling strength and environmental cues, but how these fate-determination factors are transcriptionally regulated remains poorly understood. We summarize emerging data on the role of transcription factors (TFs) - particularly the ETS and IRF families - in regulating MZB and FOB lineage selection. Indeed, genomic analyses have identified four major groups of target genes that are crucial for FOB differentiation, revealing previously unrecognized pathways that ultimately determine biological responses specific to this lineage.
Collapse
Affiliation(s)
- Hongsheng Wang
- Laboratory of Immunogenetics, National Institute of Allergy and Infectious Diseases (NIAID), National Institutes of Health (NIH), Rockville, MD, USA.
| | - Herbert C Morse
- Laboratory of Immunogenetics, National Institute of Allergy and Infectious Diseases (NIAID), National Institutes of Health (NIH), Rockville, MD, USA
| | - Silvia Bolland
- Laboratory of Immunogenetics, National Institute of Allergy and Infectious Diseases (NIAID), National Institutes of Health (NIH), Rockville, MD, USA.
| |
Collapse
|
35
|
Fischer-Riepe L, Daber N, Schulte-Schrepping J, Véras De Carvalho BC, Russo A, Pohlen M, Fischer J, Chasan AI, Wolf M, Ulas T, Glander S, Schulz C, Skryabin B, Wollbrink Dipl-Ing A, Steingraeber N, Stremmel C, Koehle M, Gärtner F, Vettorazzi S, Holzinger D, Gross J, Rosenbauer F, Stoll M, Niemann S, Tuckermann J, Schultze JL, Roth J, Barczyk-Kahlert K. CD163 expression defines specific, IRF8-dependent, immune-modulatory macrophages in the bone marrow. J Allergy Clin Immunol 2020; 146:1137-1151. [PMID: 32199911 DOI: 10.1016/j.jaci.2020.02.034] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2019] [Revised: 01/13/2020] [Accepted: 02/11/2020] [Indexed: 12/29/2022]
Abstract
BACKGROUND Scavenger receptor CD163 is exclusively expressed on monocytes/macrophages and is widely used as a marker for alternatively activated macrophages. However, the role of CD163 is not yet clear. OBJECTIVES We sought to examine the function of CD163 in steady-state as well as in sterile and infectious inflammation. METHODS Expression of CD163 was analyzed under normal and inflammatory conditions in mice. Functional relevance of CD163 was investigated in models of inflammation in wild-type and CD163-/- mice. RESULTS We describe a subpopulation of bone marrow-resident macrophages (BMRMs) characterized by a high expression of CD163 and functionally distinct from classical bone marrow-derived macrophages. Development of CD163+ BMRMs is strictly dependent on IFN regulatory factor-8. CD163+ BMRMs show a specific transcriptome and cytokine secretion pattern demonstrating a specific immunomodulatory profile of these cells. Accordingly, CD163-/- mice show a stronger inflammation in allergic contact dermatitis, indicating a regulatory role of CD163. However, CD163-/- mice are highly susceptible to S aureus infections, demonstrating the relevance of CD163 for antimicrobial defense as well. CONCLUSIONS Our data indicate that anti-inflammatory and immunosuppressive mechanisms are not necessarily associated with a decreased antimicrobial activity. In contrast, our data define a novel macrophage population that controls overwhelming inflammation on one hand but is also necessary for an effective control of infections on the other hand.
Collapse
Affiliation(s)
| | - Niklas Daber
- Institute of Immunology, University of Muenster, Muenster, Germany
| | - Jonas Schulte-Schrepping
- Genomics and Immunoregulation, Life and Medical Sciences Institute, University of Bonn, Bonn, Germany
| | | | - Antonella Russo
- Institute of Immunology, University of Muenster, Muenster, Germany
| | - Michele Pohlen
- Institute of Immunology, University of Muenster, Muenster, Germany; Department of Medicine A, Hematology and Oncology, University Hospital of Muenster, Muenster, Germany
| | - Josephine Fischer
- Institute of Molecular Tumor Biology, University of Muenster, Muenster, Germany
| | | | - Marc Wolf
- Institute of Immunology, University of Muenster, Muenster, Germany
| | - Thomas Ulas
- Genomics and Immunoregulation, Life and Medical Sciences Institute, University of Bonn, Bonn, Germany
| | - Shirin Glander
- Institute of Human Genetics, Genetic Epidemiology, University of Muenster, Muenster, Germany
| | - Christian Schulz
- Medizinische Klinik und Poliklinik I, Klinikum der Universität München, Ludwig-Maximilians-Universität, Munich, Germany; DZHK (German Centre for Cardiovascular Research), partner site Munich Heart Alliance, Munich, Germany
| | - Boris Skryabin
- Department of Medicine, Transgenic Animal and Genetic Engineering Models, University of Muenster, Muenster, Germany
| | | | - Nadine Steingraeber
- Institute for Biomagnetism and Biosignalanalysis, University of Muenster, Muenster, Germany
| | - Christopher Stremmel
- Medizinische Klinik und Poliklinik I, Klinikum der Universität München, Ludwig-Maximilians-Universität, Munich, Germany
| | - Megan Koehle
- Medizinische Klinik und Poliklinik I, Klinikum der Universität München, Ludwig-Maximilians-Universität, Munich, Germany
| | - Florian Gärtner
- Medizinische Klinik und Poliklinik I, Klinikum der Universität München, Ludwig-Maximilians-Universität, Munich, Germany
| | - Sabine Vettorazzi
- Institute of Comparative Molecular Endocrinology (CME), University of Ulm, Ulm, Germany
| | - Dirk Holzinger
- Institute of Immunology, University of Muenster, Muenster, Germany
| | - Joachim Gross
- Institute for Biomagnetism and Biosignalanalysis, University of Muenster, Muenster, Germany
| | - Frank Rosenbauer
- Institute of Molecular Tumor Biology, University of Muenster, Muenster, Germany
| | - Monika Stoll
- Institute of Human Genetics, Genetic Epidemiology, University of Muenster, Muenster, Germany
| | - Silke Niemann
- Institute of Medical Microbiology, University Hospital Muenster, Muenster, Germany
| | - Jan Tuckermann
- Institute of Comparative Molecular Endocrinology (CME), University of Ulm, Ulm, Germany
| | - Joachim L Schultze
- Genomics and Immunoregulation, Life and Medical Sciences Institute, University of Bonn, Bonn, Germany; Platform for Single Cell Genomics and Epigenomics (PRECISE) at the DZNE and the University of Bonn, Bonn, Germany
| | - Johannes Roth
- Institute of Immunology, University of Muenster, Muenster, Germany; Interdisciplinary Centre for Clinical Research, University of Muenster, Muenster, Germany
| | | |
Collapse
|
36
|
Transcriptional regulation of DC fate specification. Mol Immunol 2020; 121:38-46. [PMID: 32151907 PMCID: PMC7187805 DOI: 10.1016/j.molimm.2020.02.021] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2019] [Revised: 02/26/2020] [Accepted: 02/28/2020] [Indexed: 12/12/2022]
Abstract
Dendritic cells function in the immune system to instruct adaptive immune cells to respond accordingly to different threats. While conventional dendritic cells can be subdivided into two main subtypes, termed cDC1s and cDC2s, it is clear that further heterogeneity exists within these subtypes, particularly for cDC2s. Understanding the signals involved in specifying each of these lineages and subtypes thereof is crucial to (i) enable us to determine their specific functions and (ii) put us in a position to be able to target these cells to promote or prevent a specific function in any given disease setting. Although we still have much to learn regarding the specification of these cells, here we review the most recent advances in our understanding of this and highlight some of the next questions for the future.
Collapse
|
37
|
Pires CF, Rosa FF, Kurochkin I, Pereira CF. Understanding and Modulating Immunity With Cell Reprogramming. Front Immunol 2019; 10:2809. [PMID: 31921109 PMCID: PMC6917620 DOI: 10.3389/fimmu.2019.02809] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2019] [Accepted: 11/15/2019] [Indexed: 12/30/2022] Open
Abstract
Cell reprogramming concepts have been classically developed in the fields of developmental and stem cell biology and are currently being explored for regenerative medicine, given its potential to generate desired cell types for replacement therapy. Cell fate can be experimentally reversed or modified by enforced expression of lineage specific transcription factors leading to pluripotency or attainment of another somatic cell type identity. The possibility to reprogram fibroblasts into induced dendritic cells (DC) competent for antigen presentation creates a paradigm shift for understanding and modulating the immune system with direct cell reprogramming. PU.1, IRF8, and BATF3 were identified as sufficient and necessary to impose DC fate in unrelated cell types, taking advantage of Clec9a, a C-type lectin receptor with restricted expression in conventional DC type 1. The identification of such minimal gene regulatory networks helps to elucidate the molecular mechanisms governing development and lineage heterogeneity along the hematopoietic hierarchy. Furthermore, the generation of patient-tailored reprogrammed immune cells provides new and exciting tools for the expanding field of cancer immunotherapy. Here, we summarize cell reprogramming concepts and experimental approaches, review current knowledge at the intersection of cell reprogramming with hematopoiesis, and propose how cell fate engineering can be merged to immunology, opening new opportunities to understand the immune system in health and disease.
Collapse
Affiliation(s)
- Cristiana F. Pires
- Cell Reprogramming in Hematopoiesis and Immunity Laboratory, Lund Stem Cell Center, Molecular Medicine and Gene Therapy, Lund University, Lund, Sweden
- Wallenberg Center for Molecular Medicine, Lund University, Lund, Sweden
- Center for Neuroscience and Cell Biology, University of Coimbra, Coimbra, Portugal
| | - Fábio F. Rosa
- Cell Reprogramming in Hematopoiesis and Immunity Laboratory, Lund Stem Cell Center, Molecular Medicine and Gene Therapy, Lund University, Lund, Sweden
- Wallenberg Center for Molecular Medicine, Lund University, Lund, Sweden
- Center for Neuroscience and Cell Biology, University of Coimbra, Coimbra, Portugal
| | - Ilia Kurochkin
- Center for Neurobiology and Brain Restoration, Skolkovo Institute of Science and Technology, Moscow, Russia
| | - Carlos-Filipe Pereira
- Cell Reprogramming in Hematopoiesis and Immunity Laboratory, Lund Stem Cell Center, Molecular Medicine and Gene Therapy, Lund University, Lund, Sweden
- Wallenberg Center for Molecular Medicine, Lund University, Lund, Sweden
- Center for Neuroscience and Cell Biology, University of Coimbra, Coimbra, Portugal
| |
Collapse
|
38
|
Amon L, Lehmann CHK, Baranska A, Schoen J, Heger L, Dudziak D. Transcriptional control of dendritic cell development and functions. INTERNATIONAL REVIEW OF CELL AND MOLECULAR BIOLOGY 2019; 349:55-151. [PMID: 31759434 DOI: 10.1016/bs.ircmb.2019.10.001] [Citation(s) in RCA: 55] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Dendritic cells (DCs) are major regulators of adaptive immunity, as they are not only capable to induce efficient immune responses, but are also crucial to maintain peripheral tolerance and thereby inhibit autoimmune reactions. DCs bridge the innate and the adaptive immune system by presenting peptides of self and foreign antigens as peptide MHC complexes to T cells. These properties render DCs as interesting target cells for immunomodulatory therapies in cancer, but also autoimmune diseases. Several subsets of DCs with special properties and functions have been described. Recent achievements in understanding transcriptional programs on single cell level, together with the generation of new murine models targeting specific DC subsets, advanced our current understanding of DC development and function. Thus, DCs arise from precursor cells in the bone marrow with distinct progenitor cell populations splitting the monocyte populations and macrophage populations from the DC lineage, which upon lineage commitment can be separated into conventional cDC1, cDC2, and plasmacytoid DCs (pDCs). The DC populations harbor intrinsic programs enabling them to react for specific pathogens in dependency on the DC subset, and thereby orchestrate T cell immune responses. Similarities, but also varieties, between human and murine DC subpopulations are challenging, and will require further investigation of human specimens under consideration of the influence of the tissue micromilieu and DC subset localization in the future.
Collapse
Affiliation(s)
- Lukas Amon
- Laboratory of Dendritic Cell Biology, Department of Dermatology, University Hospital Erlangen, Friedrich-Alexander University of Erlangen-Nürnberg, Erlangen, Germany
| | - Christian H K Lehmann
- Laboratory of Dendritic Cell Biology, Department of Dermatology, University Hospital Erlangen, Friedrich-Alexander University of Erlangen-Nürnberg, Erlangen, Germany
| | - Anna Baranska
- Laboratory of Dendritic Cell Biology, Department of Dermatology, University Hospital Erlangen, Friedrich-Alexander University of Erlangen-Nürnberg, Erlangen, Germany
| | - Janina Schoen
- Laboratory of Dendritic Cell Biology, Department of Dermatology, University Hospital Erlangen, Friedrich-Alexander University of Erlangen-Nürnberg, Erlangen, Germany
| | - Lukas Heger
- Laboratory of Dendritic Cell Biology, Department of Dermatology, University Hospital Erlangen, Friedrich-Alexander University of Erlangen-Nürnberg, Erlangen, Germany
| | - Diana Dudziak
- Laboratory of Dendritic Cell Biology, Department of Dermatology, University Hospital Erlangen, Friedrich-Alexander University of Erlangen-Nürnberg, Erlangen, Germany.
| |
Collapse
|
39
|
Guermonprez P, Gerber-Ferder Y, Vaivode K, Bourdely P, Helft J. Origin and development of classical dendritic cells. INTERNATIONAL REVIEW OF CELL AND MOLECULAR BIOLOGY 2019; 349:1-54. [PMID: 31759429 DOI: 10.1016/bs.ircmb.2019.08.002] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Classical dendritic cells (cDCs) are mononuclear phagocytes of hematopoietic origin specialized in the induction and regulation of adaptive immunity. Initially defined by their unique T cell activation potential, it became quickly apparent that cDCs would be difficult to distinguish from other phagocyte lineages, by solely relying on marker-based approaches. Today, cDCs definition increasingly embed their unique ontogenetic features. A growing consensus defines cDCs on multiple criteria including: (1) dependency on the fms-like tyrosine kinase 3 ligand hematopoietic growth factor, (2) development from the common DC bone marrow progenitor, (3) constitutive expression of the transcription factor ZBTB46 and (4) the ability to induce, after adequate stimulation, the activation of naïve T lymphocytes. cDCs are a heterogeneous cell population that contains two main subsets, named type 1 and type 2 cDCs, arising from divergent ontogenetic pathways and populating multiple lymphoid and non-lymphoid tissues. Here, we present recent knowledge on the cellular and molecular pathways controlling the specification and commitment of cDC subsets from murine and human hematopoietic stem cells.
Collapse
Affiliation(s)
- Pierre Guermonprez
- King's College London, Centre for Inflammation Biology and Cancer Immunology, The Peter Gorer Department of Immmunobiology, London, United Kingdom; Université de Paris, CNRS ERL8252, INSERM1149, Centre for Inflammation Research, Paris, France.
| | - Yohan Gerber-Ferder
- Institut Curie, PSL Research University, INSERM U932, SiRIC «Translational Immunotherapy Team», Paris, France; Université de Paris, Immunity and Cancer Department, INSERM U932, Institut Curie, Paris, France
| | - Kristine Vaivode
- King's College London, Centre for Inflammation Biology and Cancer Immunology, The Peter Gorer Department of Immmunobiology, London, United Kingdom
| | - Pierre Bourdely
- King's College London, Centre for Inflammation Biology and Cancer Immunology, The Peter Gorer Department of Immmunobiology, London, United Kingdom
| | - Julie Helft
- Institut Curie, PSL Research University, INSERM U932, SiRIC «Translational Immunotherapy Team», Paris, France; Université de Paris, Immunity and Cancer Department, INSERM U932, Institut Curie, Paris, France.
| |
Collapse
|
40
|
Bagadia P, Huang X, Liu TT, Murphy KM. Shared Transcriptional Control of Innate Lymphoid Cell and Dendritic Cell Development. Annu Rev Cell Dev Biol 2019; 35:381-406. [PMID: 31283378 PMCID: PMC6886469 DOI: 10.1146/annurev-cellbio-100818-125403] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Innate immunity and adaptive immunity consist of highly specialized immune lineages that depend on transcription factors for both function and development. In this review, we dissect the similarities between two innate lineages, innate lymphoid cells (ILCs) and dendritic cells (DCs), and an adaptive immune lineage, T cells. ILCs, DCs, and T cells make up four functional immune modules and interact in concert to produce a specified immune response. These three immune lineages also share transcriptional networks governing the development of each lineage, and we discuss the similarities between ILCs and DCs in this review.
Collapse
Affiliation(s)
- Prachi Bagadia
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, Missouri 63108, USA;
| | - Xiao Huang
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, Missouri 63108, USA;
| | - Tian-Tian Liu
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, Missouri 63108, USA;
| | - Kenneth M Murphy
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, Missouri 63108, USA;
- Howard Hughes Medical Institute, Washington University School of Medicine, St. Louis, Missouri 63108, USA
| |
Collapse
|
41
|
Durai V, Bagadia P, Granja JM, Satpathy AT, Kulkarni DH, Davidson JT, Wu R, Patel SJ, Iwata A, Liu TT, Huang X, Briseño CG, Grajales-Reyes GE, Wöhner M, Tagoh H, Kee BL, Newberry RD, Busslinger M, Chang HY, Murphy TL, Murphy KM. Cryptic activation of an Irf8 enhancer governs cDC1 fate specification. Nat Immunol 2019; 20:1161-1173. [PMID: 31406378 PMCID: PMC6707878 DOI: 10.1038/s41590-019-0450-x] [Citation(s) in RCA: 122] [Impact Index Per Article: 20.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2018] [Accepted: 06/17/2019] [Indexed: 01/25/2023]
Abstract
Induction of the transcription factor Irf8 in the common dendritic cell progenitor (CDP) is required for classical type 1 dendritic cell (cDC1) fate specification, but the mechanisms controlling this induction are unclear. In the present study Irf8 enhancers were identified via chromatin profiling of dendritic cells and CRISPR/Cas9 genome editing was used to assess their roles in Irf8 regulation. An enhancer 32 kilobases (kb) downstream of the Irf8 transcriptional start site (+32-kb Irf8) that was active in mature cDC1s was required for the development of this lineage, but not for its specification. Instead, a +41-kb Irf8 enhancer, previously thought to be active only in plasmacytoid dendritic cells, was found to also be transiently accessible in cDC1 progenitors, and deleting this enhancer prevented the induction of Irf8 in CDPs and abolished cDC1 specification. Thus, cryptic activation of the +41-kb Irf8 enhancer in dendritic cell progenitors is responsible for cDC1 fate specification.
Collapse
Affiliation(s)
- Vivek Durai
- Department of Pathology and Immunology, Washington University in St Louis, School of Medicine, St Louis, MO, USA
| | - Prachi Bagadia
- Department of Pathology and Immunology, Washington University in St Louis, School of Medicine, St Louis, MO, USA
| | - Jeffrey M Granja
- Center for Personal Dynamic Regulomes, Stanford University School of Medicine, Stanford, CA, USA
- Deparment of Genetics, Stanford University School of Medicine, Stanford, CA, USA
- Biophysics Program, Stanford University School of Medicine, Stanford, CA, USA
| | - Ansuman T Satpathy
- Center for Personal Dynamic Regulomes, Stanford University School of Medicine, Stanford, CA, USA
- Department of Pathology, Stanford University School of Medicine, Stanford, CA, USA
| | - Devesha H Kulkarni
- Division of Gastroenterology, John T. Milliken Department of Medicine, Washington University in St Louis, School of Medicine, St Louis, MO, USA
| | - Jesse T Davidson
- Department of Pathology and Immunology, Washington University in St Louis, School of Medicine, St Louis, MO, USA
| | - Renee Wu
- Department of Pathology and Immunology, Washington University in St Louis, School of Medicine, St Louis, MO, USA
| | - Swapneel J Patel
- Division of Rheumatology, John T. Milliken Department of Medicine, Washington University in St Louis, School of Medicine, St Louis, MO, USA
| | - Arifumi Iwata
- Department of Pathology and Immunology, Washington University in St Louis, School of Medicine, St Louis, MO, USA
| | - Tian-Tian Liu
- Department of Pathology and Immunology, Washington University in St Louis, School of Medicine, St Louis, MO, USA
- Howard Hughes Medical Institute, Washington University in St Louis, School of Medicine, St Louis, MO, USA
| | - Xiao Huang
- Department of Pathology and Immunology, Washington University in St Louis, School of Medicine, St Louis, MO, USA
| | - Carlos G Briseño
- Department of Pathology and Immunology, Washington University in St Louis, School of Medicine, St Louis, MO, USA
| | - Gary E Grajales-Reyes
- Department of Pathology and Immunology, Washington University in St Louis, School of Medicine, St Louis, MO, USA
| | - Miriam Wöhner
- Research Institute of Molecular Pathology, Campus-Vienna-Biocenter 1, Vienna, Austria
| | - Hiromi Tagoh
- Research Institute of Molecular Pathology, Campus-Vienna-Biocenter 1, Vienna, Austria
| | - Barbara L Kee
- Department of Pathology and Committee on Immunology, University of Chicago, Chicago, IL, USA
| | - Rodney D Newberry
- Division of Gastroenterology, John T. Milliken Department of Medicine, Washington University in St Louis, School of Medicine, St Louis, MO, USA
| | - Meinrad Busslinger
- Research Institute of Molecular Pathology, Campus-Vienna-Biocenter 1, Vienna, Austria
| | - Howard Y Chang
- Center for Personal Dynamic Regulomes, Stanford University School of Medicine, Stanford, CA, USA
- Howard Hughes Medical Institute, Stanford University School of Medicine, Stanford, CA, USA
| | - Theresa L Murphy
- Department of Pathology and Immunology, Washington University in St Louis, School of Medicine, St Louis, MO, USA
| | - Kenneth M Murphy
- Department of Pathology and Immunology, Washington University in St Louis, School of Medicine, St Louis, MO, USA.
- Howard Hughes Medical Institute, Washington University in St Louis, School of Medicine, St Louis, MO, USA.
| |
Collapse
|
42
|
|
43
|
Fontela MG, Notario L, Alari-Pahissa E, Lorente E, Lauzurica P. The Conserved Non-Coding Sequence 2 (CNS2) Enhances CD69 Transcription through Cooperation between the Transcription Factors Oct1 and RUNX1. Genes (Basel) 2019; 10:genes10090651. [PMID: 31466317 PMCID: PMC6770821 DOI: 10.3390/genes10090651] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2019] [Revised: 07/29/2019] [Accepted: 08/23/2019] [Indexed: 02/02/2023] Open
Abstract
The immune regulatory receptor CD69 is expressed upon activation in all types of leukocytes and is strongly regulated at the transcriptional level. We previously described that, in addition to the CD69 promoter, there are four conserved noncoding regions (CNS1-4) upstream of the CD69 promoter. Furthermore, we proposed that CNS2 is the main enhancer of CD69 transcription. In the present study, we mapped the transcription factor (TF) binding sites (TFBS) from ChIP-seq databases within CNS2. Through luciferase reporter assays, we defined a ~60 bp sequence that acts as the minimum enhancer core of mouse CNS2, which includes the Oct1 TFBS. This enhancer core establishes cooperative interactions with the 3′ and 5′ flanking regions, which contain RUNX1 BS. In agreement with the luciferase reporter data, the inhibition of RUNX1 and Oct1 TF expression by siRNA suggests that they synergistically enhance endogenous CD69 gene transcription. In summary, we describe an enhancer core containing RUNX1 and Oct1 BS that is important for the activity of the most potent CD69 gene transcription enhancer.
Collapse
Affiliation(s)
- Miguel G. Fontela
- Microbiology National Center, Instituto de Salud Carlos III, Majadahonda, 28220 Madrid, Spain
| | - Laura Notario
- Microbiology National Center, Instituto de Salud Carlos III, Majadahonda, 28220 Madrid, Spain
| | - Elisenda Alari-Pahissa
- Department of Experimental and Health Science, University Pompeu Fabra, 08003 Barcelona, Spain
| | - Elena Lorente
- Microbiology National Center, Instituto de Salud Carlos III, Majadahonda, 28220 Madrid, Spain
| | - Pilar Lauzurica
- Microbiology National Center, Instituto de Salud Carlos III, Majadahonda, 28220 Madrid, Spain
- Correspondence: ; Tel.: +34-918222720
| |
Collapse
|
44
|
Wardowska A, Komorniczak M, Bułło-Piontecka B, Dȩbska-Ślizień MA, Pikuła M. Transcriptomic and Epigenetic Alterations in Dendritic Cells Correspond With Chronic Kidney Disease in Lupus Nephritis. Front Immunol 2019; 10:2026. [PMID: 31507612 PMCID: PMC6718474 DOI: 10.3389/fimmu.2019.02026] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2019] [Accepted: 08/12/2019] [Indexed: 12/11/2022] Open
Abstract
Systemic lupus erythematosus (SLE) is a serious autoimmune disease with variety of organ manifestations. The most dreadful one, affecting the majority of SLE patients, is kidney manifestation-lupus nephritis (LN). Dendritic cells (DC) are believed to be one of the culprits of immune dysregulation in LN. Flow cytometry analysis was applied to identify the frequency and activity of peripheral blood DCs subpopulations: myeloid and plasmacytoid, in LN patients. Magnetically isolated mDCs and pDCs were subjected to molecular analysis of genes expression, evaluation of global DNA methylation and histone H3 methylation. We observed distinctive features of DCs associated with the stages of nephritis in LN patients. Lower numbers of pDCs were observed in patients with severe LN, while increased co-stimulatory potential of mDCs was connected with the early, mild stage of this disease. IRF1 transcript upregulation was specific for mDCs from total LN patients, while exceptional amount of IRF1 mRNA was detected in mDCs from severe LN patients. DCs DNA hypermethylation seemed characteristic for severe LN, whereas a decrease in H3K4me3 and H3K27me3 marks was significant for the early stages of LN. These findings present dendritic cell alterations that may reflect renal involvement in SLE, laying foundations for new strategy of diagnosis and monitoring of LN patients, omitting invasive kidney biopsies.
Collapse
Affiliation(s)
- Anna Wardowska
- Laboratory of Tissue Engineering and Regenerative Medicine, Department of Embryology, Medical University of Gdansk, Gdansk, Poland.,Department of Clinical Immunology and Transplantology, Medical University of Gdansk, Gdansk, Poland
| | - Michał Komorniczak
- Department of Nephrology, Transplantology, and Internal Diseases, Medical University of Gdansk, Gdansk, Poland
| | - Barbara Bułło-Piontecka
- Department of Nephrology, Transplantology, and Internal Diseases, Medical University of Gdansk, Gdansk, Poland
| | - M Alicja Dȩbska-Ślizień
- Department of Nephrology, Transplantology, and Internal Diseases, Medical University of Gdansk, Gdansk, Poland
| | - Michał Pikuła
- Laboratory of Tissue Engineering and Regenerative Medicine, Department of Embryology, Medical University of Gdansk, Gdansk, Poland.,Department of Clinical Immunology and Transplantology, Medical University of Gdansk, Gdansk, Poland
| |
Collapse
|
45
|
Epigenetic control of early dendritic cell lineage specification by the transcription factor IRF8 in mice. Blood 2019; 133:1803-1813. [PMID: 30796024 DOI: 10.1182/blood-2018-06-857789] [Citation(s) in RCA: 43] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2018] [Accepted: 02/13/2019] [Indexed: 01/04/2023] Open
Abstract
Dendritic cells (DCs), which are vital for immune responses, are derived from bone marrow hematopoietic stem cells via common DC progenitors (CDPs). DC lineage fate decisions occurring at stages much earlier than CDPs have recently been recognized, yet the mechanism remains elusive. By single-cell RNA-sequencing, in vivo cell transfer experiments, and an assay for transposase-accessible chromatin sequencing using wild-type, IRF8-GFP chimera knock-in or IRF8-knockout mice, we demonstrate that IRF8 regulates chromatin at the lymphoid-primed multipotent progenitor (LMPP) stage to induce early commitment toward DCs. A low but significant expression of IRF8, a transcription factor essential for DC and monocyte development, was initiated in a subpopulation within LMPPs. These IRF8+ LMPPs were derived from IRF8- LMPPs and predominantly produced DCs, especially classical DC1s, potentially via known progenitors, such as monocyte-DC progenitors, CDPs, and preclassical DCs. IRF8+ LMPPs did not generate significant numbers of monocytes, neutrophils, or lymphocytes. Although IRF8- and IRF8+ LMPPs displayed very similar global gene expression patterns, the chromatin of enhancers near DC lineage genes was more accessible in IRF8+ LMPPs than in IRF8- LMPPs, an epigenetic change dependent on IRF8. The majority of the genes epigenetically primed by IRF8 were still transcriptionally inactive at the LMPP stage, but were highly expressed in the downstream DC lineage populations such as CDPs. Therefore, early expression of the key transcription factor IRF8 changes chromatin states in otherwise multipotent progenitors, biasing their fate decision toward DCs.
Collapse
|
46
|
Theisen DJ, Ferris ST, Briseño CG, Kretzer N, Iwata A, Murphy KM, Murphy TL. Batf3-Dependent Genes Control Tumor Rejection Induced by Dendritic Cells Independently of Cross-Presentation. Cancer Immunol Res 2019; 7:29-39. [PMID: 30482745 DOI: 10.1158/2326-6066.cir-18-0138] [Citation(s) in RCA: 41] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2018] [Revised: 09/12/2018] [Accepted: 11/21/2018] [Indexed: 11/16/2022]
Abstract
The BATF3-dependent cDC1 lineage of conventional dendritic cells (cDC) is required for rejection of immunogenic sarcomas and for rejection of progressive sarcomas during checkpoint blockade therapy. One unique function of the cDC1 lineage is the efficient cross-presentation of tumor-derived neoantigens to CD8+ T cells, but it is not clear that this is the only unique function of cDC1 required for tumor rejection. We previously showed that BATF3 functions during cDC1 lineage commitment to maintain IRF8 expression in the specified cDC1 progenitor. However, since cDC1 progenitors do not develop into mature cDC1s in Batf3 -/- mice, it is still unclear whether BATF3 has additional functions in mature cDC1 cells. A transgenic Irf8-Venus reporter allele increases IRF8 protein concentration sufficiently to allow autonomous cDC1 development in spleens of Batf3 -/- mice. These restored Batf3 -/- cDC1s are transcriptionally similar to control wild-type cDC1s but have reduced expression of a restricted set of cDC1-specific genes. Restored Batf3 -/- cDC1s are able to cross-present cell-associated antigens both in vitro and in vivo However, Batf3 -/- cDC1 exhibit altered characteristics in vivo and are unable to mediate tumor rejection. These results show that BATF3, in addition to regulating Irf8 expression to stabilize cDC1 lineage commitment, also controls expression of a small set of genes required for cDC1-mediated tumor rejection. These BATF3-regulated genes may be useful targets in immunotherapies aimed at promoting tumor rejection.
Collapse
Affiliation(s)
- Derek J Theisen
- Department of Pathology and Immunology, Washington University in St. Louis, School of Medicine, St. Louis, Missouri
| | - Stephen T Ferris
- Department of Pathology and Immunology, Washington University in St. Louis, School of Medicine, St. Louis, Missouri
| | - Carlos G Briseño
- Department of Pathology and Immunology, Washington University in St. Louis, School of Medicine, St. Louis, Missouri
| | - Nicole Kretzer
- Department of Pathology and Immunology, Washington University in St. Louis, School of Medicine, St. Louis, Missouri
| | - Arifumi Iwata
- Department of Pathology and Immunology, Washington University in St. Louis, School of Medicine, St. Louis, Missouri
| | - Kenneth M Murphy
- Department of Pathology and Immunology, Washington University in St. Louis, School of Medicine, St. Louis, Missouri
- Howard Hughes Medical Institute, Washington University in St. Louis, School of Medicine, St. Louis, Missouri
| | - Theresa L Murphy
- Department of Pathology and Immunology, Washington University in St. Louis, School of Medicine, St. Louis, Missouri.
| |
Collapse
|
47
|
Rosa FF, Pires CF, Kurochkin I, Ferreira AG, Gomes AM, Palma LG, Shaiv K, Solanas L, Azenha C, Papatsenko D, Schulz O, e Sousa CR, Pereira CF. Direct reprogramming of fibroblasts into antigen-presenting dendritic cells. Sci Immunol 2018; 3:3/30/eaau4292. [DOI: 10.1126/sciimmunol.aau4292] [Citation(s) in RCA: 45] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2018] [Accepted: 10/03/2018] [Indexed: 12/31/2022]
Abstract
Ectopic expression of transcription factors has been used to reprogram differentiated somatic cells toward pluripotency or to directly reprogram them to other somatic cell lineages. This concept has been explored in the context of regenerative medicine. Here, we set out to generate dendritic cells (DCs) capable of presenting antigens from mouse and human fibroblasts. By screening combinations of 18 transcription factors that are expressed in DCs, we have identified PU.1, IRF8, and BATF3 transcription factors as being sufficient to reprogram both mouse and human fibroblasts to induced DCs (iDCs). iDCs acquire a conventional DC type 1–like transcriptional program, with features of interferon-induced maturation. iDCs secrete inflammatory cytokines and have the ability to engulf, process, and present antigens to T cells. Furthermore, we demonstrate that murine iDCs generated here were able to cross-present antigens to CD8+ T cells. Our reprogramming system should facilitate better understanding of DC specification programs and serve as a platform for the development of patient-specific DCs for immunotherapy.
Collapse
|
48
|
Weston BR, Li L, Tyson JJ. Mathematical Analysis of Cytokine-Induced Differentiation of Granulocyte-Monocyte Progenitor Cells. Front Immunol 2018; 9:2048. [PMID: 30279691 PMCID: PMC6153365 DOI: 10.3389/fimmu.2018.02048] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2018] [Accepted: 08/20/2018] [Indexed: 01/01/2023] Open
Abstract
Granulocyte-monocyte progenitor (GMP) cells play a vital role in the immune system by maturing into a variety of white blood cells, including neutrophils and macrophages, depending on exposure to cytokines such as various types of colony stimulating factors (CSF). Granulocyte-CSF (G-CSF) induces granulopoiesis and macrophage-CSF (M-CSF) induces monopoiesis, while granulocyte/macrophage-CSF (GM-CSF) favors monocytic and granulocytic differentiation at low and high concentrations, respectively. Although these differentiation pathways are well documented, the mechanisms behind the diverse behavioral responses of GMP cells to CSFs are not well understood. In this paper, we propose a mechanism of interacting CSF-receptors and transcription factors that control GMP differentiation, convert the mechanism into a set of differential equations, and explore the properties of this mathematical model using dynamical systems theory. Our model reproduces numerous experimental observations of GMP cell differentiation in response to varying dosages of G-CSF, M-CSF, and GM-CSF. In particular, we are able to reproduce the concentration-dependent behavior of GM-CSF induced differentiation, and propose a mechanism driving this behavior. In addition, we explore the differentiation of a fourth phenotype, monocytic myeloid-derived suppressor cells (M-MDSC), showing how they might fit into the classical pathways of GMP differentiation and how progenitor cells can be primed for M-MDSC differentiation. Finally, we use the model to make novel predictions that can be explored by future experimental studies.
Collapse
Affiliation(s)
- Bronson R Weston
- Program in Genetics, Bioinformatics, and Computational Biology, Virginia Polytechnic Institute and State University, Blacksburg, VA, United States
| | - Liwu Li
- Department of Biological Sciences, Virginia Polytechnic Institute and State University, Blacksburg, VA, United States
| | - John J Tyson
- Department of Biological Sciences, Virginia Polytechnic Institute and State University, Blacksburg, VA, United States
| |
Collapse
|
49
|
Carey HA, Hildreth BE, Geisler JA, Nickel MC, Cabrera J, Ghosh S, Jiang Y, Yan J, Lee J, Makam S, Young NA, Valiente GR, Jarjour WN, Huang K, Rosol TJ, Toribio RE, Charles JF, Ostrowski MC, Sharma SM. Enhancer variants reveal a conserved transcription factor network governed by PU.1 during osteoclast differentiation. Bone Res 2018; 6:8. [PMID: 29619268 PMCID: PMC5874256 DOI: 10.1038/s41413-018-0011-1] [Citation(s) in RCA: 31] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2017] [Revised: 01/12/2018] [Accepted: 02/16/2018] [Indexed: 12/20/2022] Open
Abstract
Genome-wide association studies (GWASs) have been instrumental in understanding complex phenotypic traits. However, they have rarely been used to understand lineage-specific pathways and functions that contribute to the trait. In this study, by integrating lineage-specific enhancers from mesenchymal and myeloid compartments with bone mineral density loci, we were able to segregate osteoblast- and osteoclast (OC)-specific functions. Specifically, in OCs, a PU.1-dependent transcription factor (TF) network was revealed. Deletion of PU.1 in OCs in mice resulted in severe osteopetrosis. Functional genomic analysis indicated PU.1 and MITF orchestrated a TF network essential for OC differentiation. Several of these TFs were regulated by cooperative binding of PU.1 with BRD4 to form superenhancers. Further, PU.1 is essential for conformational changes in the superenhancer region of Nfatc1. In summary, our study demonstrates that combining GWASs with genome-wide binding studies and model organisms could decipher lineage-specific pathways contributing to complex disease states. Genetic variation in non-coding regions of DNA could raise osteoporosis risk by affecting osteoclast differentiation. Osteoporosis occurs when the normal process of bone remodeling by osteoblasts and osteoclasts falls out of balance. Genome-wide association studies (GWAS) have identified numerous single nucleotide polymorphisms (SNPs) associated with osteoporosis, but how these affect specific cell types was unclear. Sudarshana Sharma and Michael Ostrowski at the Medical University of South Carolina and colleagues wondered if variations in non-coding ‘enhancer’ regions of DNA, might shed light on the molecular underpinnings of osteoporosis. So, they overlaid SNPs associated with reduced bone mineral density onto enhancers in mesenchymal and myeloid cells—the precursors of osteoblasts and osteoclasts—identifying a transcription factor network in myeloid cells that drives the differentiation of osteoclasts. When this was disrupted in mice, severe defects in osteoclast differentiation and function resulted.
Collapse
Affiliation(s)
- Heather A Carey
- 1Department of Cancer Biology and Genetics and Comprehensive Cancer Center, The Ohio State University Wexner Medical Center, Columbus, OH 43210 USA
| | - Blake E Hildreth
- 1Department of Cancer Biology and Genetics and Comprehensive Cancer Center, The Ohio State University Wexner Medical Center, Columbus, OH 43210 USA.,2College of Veterinary Medicine, The Ohio State University, Columbus, OH 43210 USA.,3Department of Biochemistry and Molecular Biology and Hollings Cancer Center, Medical University of South Carolina, Charleston, SC 29425 USA
| | - Jennifer A Geisler
- 1Department of Cancer Biology and Genetics and Comprehensive Cancer Center, The Ohio State University Wexner Medical Center, Columbus, OH 43210 USA.,2College of Veterinary Medicine, The Ohio State University, Columbus, OH 43210 USA
| | - Mara C Nickel
- 1Department of Cancer Biology and Genetics and Comprehensive Cancer Center, The Ohio State University Wexner Medical Center, Columbus, OH 43210 USA
| | - Jennifer Cabrera
- 1Department of Cancer Biology and Genetics and Comprehensive Cancer Center, The Ohio State University Wexner Medical Center, Columbus, OH 43210 USA
| | - Sankha Ghosh
- 1Department of Cancer Biology and Genetics and Comprehensive Cancer Center, The Ohio State University Wexner Medical Center, Columbus, OH 43210 USA
| | - Yue Jiang
- 1Department of Cancer Biology and Genetics and Comprehensive Cancer Center, The Ohio State University Wexner Medical Center, Columbus, OH 43210 USA
| | - Jing Yan
- 4Division of Rheumatology, Immunology and Allergy, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA USA
| | - James Lee
- 1Department of Cancer Biology and Genetics and Comprehensive Cancer Center, The Ohio State University Wexner Medical Center, Columbus, OH 43210 USA
| | - Sandeep Makam
- 1Department of Cancer Biology and Genetics and Comprehensive Cancer Center, The Ohio State University Wexner Medical Center, Columbus, OH 43210 USA
| | - Nicholas A Young
- 5Division of Rheumatology and Immunology, Department of Internal Medicine, The Ohio State University Wexner Medical Center, Columbus, OH 43210 USA
| | - Giancarlo R Valiente
- 5Division of Rheumatology and Immunology, Department of Internal Medicine, The Ohio State University Wexner Medical Center, Columbus, OH 43210 USA
| | - Wael N Jarjour
- 5Division of Rheumatology and Immunology, Department of Internal Medicine, The Ohio State University Wexner Medical Center, Columbus, OH 43210 USA
| | - Kun Huang
- 6Department of Biomedical Informatics, The Ohio State University Wexner Medical Center, Columbus, OH 43210 USA
| | - Thomas J Rosol
- 2College of Veterinary Medicine, The Ohio State University, Columbus, OH 43210 USA
| | - Ramiro E Toribio
- 2College of Veterinary Medicine, The Ohio State University, Columbus, OH 43210 USA
| | - Julia F Charles
- 4Division of Rheumatology, Immunology and Allergy, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA USA
| | - Michael C Ostrowski
- 1Department of Cancer Biology and Genetics and Comprehensive Cancer Center, The Ohio State University Wexner Medical Center, Columbus, OH 43210 USA.,3Department of Biochemistry and Molecular Biology and Hollings Cancer Center, Medical University of South Carolina, Charleston, SC 29425 USA
| | - Sudarshana M Sharma
- 1Department of Cancer Biology and Genetics and Comprehensive Cancer Center, The Ohio State University Wexner Medical Center, Columbus, OH 43210 USA.,3Department of Biochemistry and Molecular Biology and Hollings Cancer Center, Medical University of South Carolina, Charleston, SC 29425 USA
| |
Collapse
|
50
|
Dress RJ, Wong AYW, Ginhoux F. Homeostatic control of dendritic cell numbers and differentiation. Immunol Cell Biol 2018; 96:463-476. [DOI: 10.1111/imcb.12028] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2018] [Revised: 02/18/2018] [Accepted: 02/19/2018] [Indexed: 12/20/2022]
Affiliation(s)
- Regine J Dress
- Singapore Immunology Network (SIgN); Agency for Science; Technology, and Research (A*STAR); Singapore 138648 Singapore
| | - Alicia YW Wong
- Singapore Immunology Network (SIgN); Agency for Science; Technology, and Research (A*STAR); Singapore 138648 Singapore
| | - Florent Ginhoux
- Singapore Immunology Network (SIgN); Agency for Science; Technology, and Research (A*STAR); Singapore 138648 Singapore
| |
Collapse
|