51
|
Krauss PL, Pfeiffenberger M, Damerau A, Buttgereit T, Chen Y, Gaber T, Buttgereit F. Production of IL-6 and Phagocytosis Are the Most Resilient Immune Functions in Metabolically Compromised Human Monocytes. Front Immunol 2021; 12:730672. [PMID: 34737742 PMCID: PMC8562108 DOI: 10.3389/fimmu.2021.730672] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2021] [Accepted: 10/01/2021] [Indexed: 01/23/2023] Open
Abstract
At sites of inflammation, monocytes carry out specific immune functions while facing challenging metabolic restrictions. Here, we investigated the potential of human monocytes to adapt to conditions of gradually inhibited oxidative phosphorylation (OXPHOS) under glucose free conditions. We used myxothiazol, an inhibitor of mitochondrial respiration, to adjust two different levels of decreased mitochondrial ATP production. At these levels, and compared to uninhibited OXPHOS, we assessed phagocytosis, production of reactive oxygen species (ROS) through NADPH oxidase (NOX), expression of surface activation markers CD16, CD80, CD11b, HLA-DR, and production of the inflammatory cytokines IL-1β, IL-6 and TNF-α in human monocytes. We found phagocytosis and the production of IL-6 to be least sensitive to metabolic restrictions while surface expression of CD11b, HLA-DR, production of TNF-α, IL-1β and production of ROS through NOX were most compromised by inhibition of OXPHOS in the absence of glucose. Our data demonstrate a short-term hierarchy of immune functions in human monocytes, which represents novel knowledge potentially leading to the development of new therapeutics in monocyte-mediated inflammatory diseases.
Collapse
Affiliation(s)
- Pierre-Louis Krauss
- Department of Rheumatology and Clinical Immunology, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany.,German Rheumatism Research Centre (DRFZ) Berlin, a Leibniz Institute, Berlin, Germany
| | - Moritz Pfeiffenberger
- Department of Rheumatology and Clinical Immunology, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany.,German Rheumatism Research Centre (DRFZ) Berlin, a Leibniz Institute, Berlin, Germany
| | - Alexandra Damerau
- Department of Rheumatology and Clinical Immunology, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany.,German Rheumatism Research Centre (DRFZ) Berlin, a Leibniz Institute, Berlin, Germany
| | - Thomas Buttgereit
- Department of Rheumatology and Clinical Immunology, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany.,Department of Dermatology, Venerology, and Allergology, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
| | - Yuling Chen
- Department of Rheumatology and Clinical Immunology, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany.,German Rheumatism Research Centre (DRFZ) Berlin, a Leibniz Institute, Berlin, Germany
| | - Timo Gaber
- Department of Rheumatology and Clinical Immunology, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany.,German Rheumatism Research Centre (DRFZ) Berlin, a Leibniz Institute, Berlin, Germany
| | - Frank Buttgereit
- Department of Rheumatology and Clinical Immunology, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany.,German Rheumatism Research Centre (DRFZ) Berlin, a Leibniz Institute, Berlin, Germany
| |
Collapse
|
52
|
Alcántara-Hernández M, Idoyaga J. Mass cytometry profiling of human dendritic cells in blood and tissues. Nat Protoc 2021; 16:4855-4877. [PMID: 34480131 PMCID: PMC10538357 DOI: 10.1038/s41596-021-00599-x] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2020] [Accepted: 06/29/2021] [Indexed: 12/15/2022]
Abstract
The immune system comprises distinct functionally specialized cell populations, which can be characterized in depth by mass cytometry protein profiling. Unfortunately, the low-throughput nature of mass cytometry has made it challenging to analyze minor cell populations. This is the case for dendritic cells, which represent 0.2-2% of all immune cells in tissues and yet perform the critical task of initiating and modulating immune responses. Here, we provide an optimized step-by-step protocol for the characterization of well-known and emerging human dendritic cell populations in blood and tissues using mass cytometry. We provide detailed instructions for the generation of single-cell suspensions, sample enrichment, staining, acquisition and data analysis. We also include a barcoding option that reduces acquisition variability and allows the analysis of low numbers of dendritic cells, i.e., ~20,000. In contrast to other protocols, we emphasize the use of negative selection approaches to enrich for minor populations of immune cells while avoiding their activation. The entire procedure can be completed in 2-3 d and can be conveniently paused at several stages. The procedure described in this robust and reliable protocol allows the analysis of human dendritic cells in health and disease and during vaccination.
Collapse
Affiliation(s)
- Marcela Alcántara-Hernández
- Microbiology & Immunology Department and Immunology Program, Stanford University School of Medicine, Stanford, CA, USA
| | - Juliana Idoyaga
- Microbiology & Immunology Department and Immunology Program, Stanford University School of Medicine, Stanford, CA, USA.
| |
Collapse
|
53
|
Tan RP, Ryder I, Yang N, Lam YT, Santos M, Michael PL, Robinson DA, Ng MK, Wise SG. Macrophage Polarization as a Novel Therapeutic Target for Endovascular Intervention in Peripheral Artery Disease. JACC Basic Transl Sci 2021; 6:693-704. [PMID: 34466756 PMCID: PMC8385566 DOI: 10.1016/j.jacbts.2021.04.008] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/08/2021] [Revised: 04/27/2021] [Accepted: 04/27/2021] [Indexed: 11/19/2022]
Abstract
Peripheral artery disease (PAD) has a significant impact on human health, affecting 200 million people globally. Advanced PAD severely diminishes quality of life, affecting mobility, and in its most severe form leads to limb amputation and death. Treatment of PAD is among the least effective of all endovascular procedures in terms of long-term efficacy. Chronic inflammation is a key driver of PAD; however, stents and coated balloons eluting antiproliferative drugs are most commonly used. As a result, neither stents nor coated balloons produce durable clinical outcomes in the superficial femoral artery, and both have recently been associated with significantly increased mortality. This review summarizes the most common clinical approaches and limitations to treating PAD and highlights the necessity to address the underlying causes of inflammation, identifying macrophages as a novel therapeutic target in the next generation of endovascular PAD intervention.
Collapse
Key Words
- BMS, bare-metal stent
- CAD, coronary artery disease
- DES, drug-eluting stent
- FP, femoropopliteal
- IL, interleukin
- MI, myocardial infarction
- PAD, peripheral artery disease
- PTA, percutaneous transluminal angioplasty
- SFA, superficial femoral artery
- TGF, transforming growth factor
- TNF, tumor necrosis factor
- drug-eluting balloon
- drug-eluting stent
- endovascular intervention
- macrophage polarization
- paclitaxel
- peripheral arterial disease
- vascular healing
- vascular inflammation
Collapse
Affiliation(s)
- Richard P. Tan
- Chronic Diseases, School of Medical Sciences, Faculty of Health and Medicine, University of Sydney, Sydney, New South Wales, Australia
- Charles Perkins Centre, University of Sydney, Sydney, New South Wales, Australia
- Address for correspondence: Dr. Richard P. Tan, Charles Perkins Centre, University of Sydney, Johns Hopkins Drive, Camperdown, Sydney, New South Wales 2006, Australia
| | - Isabelle Ryder
- Charles Perkins Centre, University of Sydney, Sydney, New South Wales, Australia
| | - Nianji Yang
- Chronic Diseases, School of Medical Sciences, Faculty of Health and Medicine, University of Sydney, Sydney, New South Wales, Australia
- Charles Perkins Centre, University of Sydney, Sydney, New South Wales, Australia
| | - Yuen Ting Lam
- Chronic Diseases, School of Medical Sciences, Faculty of Health and Medicine, University of Sydney, Sydney, New South Wales, Australia
- Charles Perkins Centre, University of Sydney, Sydney, New South Wales, Australia
| | - Miguel Santos
- Chronic Diseases, School of Medical Sciences, Faculty of Health and Medicine, University of Sydney, Sydney, New South Wales, Australia
- Charles Perkins Centre, University of Sydney, Sydney, New South Wales, Australia
| | - Praveesuda L. Michael
- Chronic Diseases, School of Medical Sciences, Faculty of Health and Medicine, University of Sydney, Sydney, New South Wales, Australia
- Charles Perkins Centre, University of Sydney, Sydney, New South Wales, Australia
| | - David A. Robinson
- Department of Vascular Surgery, Royal Prince Alfred Hospital, Sydney, New South Wales, Australia
| | - Martin K. Ng
- Department of Medicine, Sydney Medical School, University of Sydney, Sydney, New South Wales, Australia
- Department of Cardiology, Royal Prince Alfred Hospital, Sydney, New South Wales, Australia
| | - Steven G. Wise
- Chronic Diseases, School of Medical Sciences, Faculty of Health and Medicine, University of Sydney, Sydney, New South Wales, Australia
- Charles Perkins Centre, University of Sydney, Sydney, New South Wales, Australia
- University of Sydney Nano Institute, University of Sydney, Sydney, New South Wales, Australia
| |
Collapse
|
54
|
Tong X, Huang T, Zhang M, Chen J, Zhang Z, Li J, Du H, Ling Z, Wu Z, Yang B, Xiao S, Ai H. Four genetic loci affecting swine lung lesions identified by whole-genome sequencing-based association studies. SCIENCE CHINA. LIFE SCIENCES 2021; 64:1571-1574. [PMID: 33521858 DOI: 10.1007/s11427-020-1826-x] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/30/2020] [Accepted: 09/28/2020] [Indexed: 06/12/2023]
Affiliation(s)
- Xinkai Tong
- State Key Laboratory of Pig Genetic Improvement and Production Technology, Jiangxi Agricultural University, Nanchang, 330045, China
| | - Tao Huang
- State Key Laboratory of Pig Genetic Improvement and Production Technology, Jiangxi Agricultural University, Nanchang, 330045, China
| | - Mingpeng Zhang
- State Key Laboratory of Pig Genetic Improvement and Production Technology, Jiangxi Agricultural University, Nanchang, 330045, China
| | - Jiaqi Chen
- State Key Laboratory of Pig Genetic Improvement and Production Technology, Jiangxi Agricultural University, Nanchang, 330045, China
| | - Zhou Zhang
- State Key Laboratory of Pig Genetic Improvement and Production Technology, Jiangxi Agricultural University, Nanchang, 330045, China
| | - Jingquan Li
- State Key Laboratory of Pig Genetic Improvement and Production Technology, Jiangxi Agricultural University, Nanchang, 330045, China
| | - Huipeng Du
- State Key Laboratory of Pig Genetic Improvement and Production Technology, Jiangxi Agricultural University, Nanchang, 330045, China
| | - Ziqi Ling
- State Key Laboratory of Pig Genetic Improvement and Production Technology, Jiangxi Agricultural University, Nanchang, 330045, China
| | - Zhongzi Wu
- State Key Laboratory of Pig Genetic Improvement and Production Technology, Jiangxi Agricultural University, Nanchang, 330045, China
| | - Bin Yang
- State Key Laboratory of Pig Genetic Improvement and Production Technology, Jiangxi Agricultural University, Nanchang, 330045, China
| | - Shijun Xiao
- State Key Laboratory of Pig Genetic Improvement and Production Technology, Jiangxi Agricultural University, Nanchang, 330045, China
| | - Huashui Ai
- State Key Laboratory of Pig Genetic Improvement and Production Technology, Jiangxi Agricultural University, Nanchang, 330045, China.
| |
Collapse
|
55
|
Lepzien R, Nie M, Czarnewski P, Liu S, Yu M, Ravindran A, Kullberg S, Eklund A, Grunewald J, Smed-Sörensen A. Pulmonary and blood dendritic cells from sarcoidosis patients more potently induce IFNγ-producing Th1 cells compared with monocytes. J Leukoc Biol 2021; 111:857-866. [PMID: 34431542 DOI: 10.1002/jlb.5a0321-162r] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Sarcoidosis is a systemic inflammatory disease mainly affecting the lungs. The hallmark of sarcoidosis are granulomas that are surrounded by activated T cells, likely targeting the disease-inducing antigen. IFNγ-producing Th1 and Th17.1 T cells are elevated in sarcoidosis and associate with disease progression. Monocytes and dendritic cells (DCs) are antigen-presenting cells (APCs) and required for T cell activation. Several subsets of monocytes and DCs with different functions were identified in sarcoidosis. However, to what extent different monocyte and DC subsets can support activation and skewing of T cells in sarcoidosis is still unclear. In this study, we performed a transcriptional and functional side-by-side comparison of sorted monocytes and DCs from matched blood and bronchoalveolar lavage (BAL) fluid of sarcoidosis patients. Transcriptomic analysis of all subsets showed upregulation of genes related to T cell activation and antigen presentation in DCs compared with monocytes. Allogeneic T cell proliferation was higher after coculture with monocytes and DCs from blood compared with BAL and DCs induced more T cell proliferation compared with monocytes. After coculture, proliferating T cells showed high expression of the transcription factor Tbet and IFNγ production. We also identified Tbet and RORγt coexpressing T cells that mainly produced IFNγ. Our data show that DCs rather than monocytes from sarcoidosis patients have the ability to activate and polarize T cells towards Th1 and Th17.1 cells. This study provides a useful in vitro tool to better understand the contribution of monocytes and DCs to T cell activation and immunopathology in sarcoidosis.
Collapse
Affiliation(s)
- Rico Lepzien
- Division of Immunology and Allergy, Department of Medicine Solna, Karolinska Institutet, Karolinska University Hospital, Stockholm, Sweden
| | - Mu Nie
- Division of Immunology and Allergy, Department of Medicine Solna, Karolinska Institutet, Karolinska University Hospital, Stockholm, Sweden
| | - Paulo Czarnewski
- Department of Biochemistry and Biophysics, National Bioinformatics Infrastructure Sweden, Science for Life Laboratory, Stockholm University, Solna, Sweden
| | - Sang Liu
- Division of Immunology and Allergy, Department of Medicine Solna, Karolinska Institutet, Karolinska University Hospital, Stockholm, Sweden
| | - Meng Yu
- Division of Immunology and Allergy, Department of Medicine Solna, Karolinska Institutet, Karolinska University Hospital, Stockholm, Sweden
| | - Avinash Ravindran
- Division of Respiratory Medicine, Department of Medicine Solna and Center for Molecular Medicine, Karolinska Institutet, Karolinska University Hospital, Stockholm, Sweden
| | - Susanna Kullberg
- Division of Respiratory Medicine, Department of Medicine Solna and Center for Molecular Medicine, Karolinska Institutet, Karolinska University Hospital, Stockholm, Sweden
- Department of Respiratory Medicine, Theme Inflammation and Infection, Karolinska University Hospital, Stockholm, Sweden
| | - Anders Eklund
- Division of Respiratory Medicine, Department of Medicine Solna and Center for Molecular Medicine, Karolinska Institutet, Karolinska University Hospital, Stockholm, Sweden
- Department of Respiratory Medicine, Theme Inflammation and Infection, Karolinska University Hospital, Stockholm, Sweden
| | - Johan Grunewald
- Division of Respiratory Medicine, Department of Medicine Solna and Center for Molecular Medicine, Karolinska Institutet, Karolinska University Hospital, Stockholm, Sweden
- Department of Respiratory Medicine, Theme Inflammation and Infection, Karolinska University Hospital, Stockholm, Sweden
| | - Anna Smed-Sörensen
- Division of Immunology and Allergy, Department of Medicine Solna, Karolinska Institutet, Karolinska University Hospital, Stockholm, Sweden
| |
Collapse
|
56
|
Brands X, Haak BW, Klarenbeek AM, Butler J, Uhel F, Qin W, Otto NA, Jakobs ME, Faber DR, Lutter R, Wiersinga WJ, van der Poll T, Scicluna BP. An epigenetic and transcriptomic signature of immune tolerance in human monocytes through multi-omics integration. Genome Med 2021; 13:131. [PMID: 34399830 PMCID: PMC8365568 DOI: 10.1186/s13073-021-00948-1] [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: 12/24/2020] [Accepted: 08/05/2021] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND The plasticity of monocytes enables them to exert multiple roles during an immune response, including promoting immune tolerance. How monocytes alter their functions to convey immune tolerance in the context of lower respiratory tract infections in humans is not well understood. Here, we sought to identify epigenetic and transcriptomic features of cytokine production capacity in circulating monocytes during community-acquired pneumonia (CAP). METHODS Circulating CD14+ monocytes were obtained from the blood of CAP patients included in a longitudinal, observational cohort study, on hospitalization (acute stage, n=75), and from the same patients after a 1-month follow-up (recovery stage, n=56). Age and sex-matched non-infectious participants were included as controls (n=41). Ex vivo cytokine production after lipopolysaccharide (LPS) exposure was assessed by multiplex assay. Transcriptomes of circulating monocytes were generated by RNA-sequencing, and DNA methylation levels in the same monocytes were measured by reduced representation bisulfite sequencing. Data were integrated by fitting projection-to-latent-structure models, and signatures derived by partial least squares discrimination. RESULTS Monocytes captured during the acute stage exhibited impaired TNF, IL-1β, IL-6, and IL-10 production after ex vivo stimulation with LPS, relative to controls. IL-6 production was not resolved in recovery monocytes. Multivariate analysis of RNA-sequencing data identified 2938 significantly altered RNA transcripts in acute-stage monocytes (fold expression ≤-1.5 or ≥1.5; adjusted p ≤ 0.01), relative to controls. Comparing DNA methylation levels in circulating monocytes of CAP patients to controls revealed minimal differences, specifically in DNAse hypersensitive sites (HS) of acute-stage monocytes. Data integration identified a cholesterol biosynthesis gene signature and DNAse HS axis of IL-1β and IL-10 production (R2 =0.51). CONCLUSIONS Circulating monocytes obtained from CAP patients during the acute stage exhibited impaired cytokine production capacities, indicative of reprogramming to a state of immune tolerance, which was not fully resolved after 1 month. Our split-sample study showed that 51% of the immune tolerance phenotype can be explained, at least in part, by coordinated shifts in cholesterol biosynthesis gene expression and DNAse HS methylation levels. A multi-scale model identified an epigenetic and transcriptomic signature of immune tolerance in monocytes, with implications for future interventions in immunosuppression. TRIAL REGISTRATION NCT number NCT02928367.
Collapse
Affiliation(s)
- Xanthe Brands
- Center for Experimental Molecular Medicine, Amsterdam University Medical Centers, Academic Medical Center, University of Amsterdam, Amsterdam, 1105, AZ, the Netherlands.
| | - Bastiaan W Haak
- Center for Experimental Molecular Medicine, Amsterdam University Medical Centers, Academic Medical Center, University of Amsterdam, Amsterdam, 1105, AZ, the Netherlands
| | - Augustijn M Klarenbeek
- Center for Experimental Molecular Medicine, Amsterdam University Medical Centers, Academic Medical Center, University of Amsterdam, Amsterdam, 1105, AZ, the Netherlands
| | - Joe Butler
- Center for Experimental Molecular Medicine, Amsterdam University Medical Centers, Academic Medical Center, University of Amsterdam, Amsterdam, 1105, AZ, the Netherlands
| | - Fabrice Uhel
- Center for Experimental Molecular Medicine, Amsterdam University Medical Centers, Academic Medical Center, University of Amsterdam, Amsterdam, 1105, AZ, the Netherlands
| | - Wanhai Qin
- Center for Experimental Molecular Medicine, Amsterdam University Medical Centers, Academic Medical Center, University of Amsterdam, Amsterdam, 1105, AZ, the Netherlands
| | - Natasja A Otto
- Center for Experimental Molecular Medicine, Amsterdam University Medical Centers, Academic Medical Center, University of Amsterdam, Amsterdam, 1105, AZ, the Netherlands
| | - Marja E Jakobs
- Laboratory of Genome Analysis, Amsterdam University Medical Centers, Academic Medical Center, University of Amsterdam, Amsterdam, 1105, AZ, the Netherlands
| | | | - René Lutter
- Respiratory Medicine and Experimental Immunology, Amsterdam University Medical Centers, Academic Medical Center, University of Amsterdam, Amsterdam, 1105, AZ, the Netherlands
| | - W Joost Wiersinga
- Center for Experimental Molecular Medicine, Amsterdam University Medical Centers, Academic Medical Center, University of Amsterdam, Amsterdam, 1105, AZ, the Netherlands
- Division of Infectious Diseases, Amsterdam University Medical Centers, Academic Medical Center, University of Amsterdam, Amsterdam, 1105, AZ, the Netherlands
| | - Tom van der Poll
- Center for Experimental Molecular Medicine, Amsterdam University Medical Centers, Academic Medical Center, University of Amsterdam, Amsterdam, 1105, AZ, the Netherlands
- Division of Infectious Diseases, Amsterdam University Medical Centers, Academic Medical Center, University of Amsterdam, Amsterdam, 1105, AZ, the Netherlands
| | - Brendon P Scicluna
- Center for Experimental Molecular Medicine, Amsterdam University Medical Centers, Academic Medical Center, University of Amsterdam, Amsterdam, 1105, AZ, the Netherlands.
- Department of Applied Biomedical Sciences, Faculty of Health Sciences, Mater Dei Hospital, University of Malta, Msida, Malta.
- Centre for Molecular Medicine and Biobanking, University of Malta, Msida, Malta.
| |
Collapse
|
57
|
Zhou X, Yu W, Lyu SC, Macaubas C, Bunning B, He Z, Mellins ED, Nadeau KC. A positive feedback loop reinforces the allergic immune response in human peanut allergy. J Exp Med 2021; 218:e20201793. [PMID: 33944900 PMCID: PMC8103542 DOI: 10.1084/jem.20201793] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2020] [Revised: 12/18/2020] [Accepted: 03/04/2021] [Indexed: 12/13/2022] Open
Abstract
Food allergies are a leading cause of anaphylaxis, and cellular mechanisms involving antigen presentation likely play key roles in their pathogenesis. However, little is known about the response of specific antigen-presenting cell (APC) subsets to food allergens in the setting of food allergies. Here, we show that in peanut-allergic humans, peanut allergen drives the differentiation of CD209+ monocyte-derived dendritic cells (DCs) and CD23+ (FcєRII) myeloid dendritic cells through the action of allergen-specific CD4+ T cells. CD209+ DCs act reciprocally on the same peanut-specific CD4+ T cell population to reinforce Th2 cytokine expression in a positive feedback loop, which may explain the persistence of established food allergy. In support of this novel model, we show clinically that the initiation of oral immunotherapy (OIT) in peanut-allergic patients is associated with a decrease in CD209+ DCs, suggesting that breaking the cycle of positive feedback is associated with therapeutic effect.
Collapse
Affiliation(s)
- Xiaoying Zhou
- Sean N. Parker Center for Allergy & Asthma Research at Stanford University and Division of Pulmonary, Allergy, and Critical Care Medicine, Stanford, CA
| | - Wong Yu
- Sean N. Parker Center for Allergy & Asthma Research at Stanford University and Division of Pulmonary, Allergy, and Critical Care Medicine, Stanford, CA
| | - Shu-Chen Lyu
- Sean N. Parker Center for Allergy & Asthma Research at Stanford University and Division of Pulmonary, Allergy, and Critical Care Medicine, Stanford, CA
| | - Claudia Macaubas
- Department of Pediatrics, Program in Immunology, Stanford University, Stanford, CA
| | - Bryan Bunning
- Sean N. Parker Center for Allergy & Asthma Research at Stanford University and Division of Pulmonary, Allergy, and Critical Care Medicine, Stanford, CA
| | - Ziyuan He
- Sean N. Parker Center for Allergy & Asthma Research at Stanford University and Division of Pulmonary, Allergy, and Critical Care Medicine, Stanford, CA
| | - Elizabeth D. Mellins
- Department of Pediatrics, Program in Immunology, Stanford University, Stanford, CA
| | - Kari C. Nadeau
- Sean N. Parker Center for Allergy & Asthma Research at Stanford University and Division of Pulmonary, Allergy, and Critical Care Medicine, Stanford, CA
| |
Collapse
|
58
|
Nuñez-Reza KJ, Naldi A, Sánchez-Jiménez A, Leon-Apodaca AV, Santana MA, Thomas-Chollier M, Thieffry D, Medina-Rivera A. Logical modelling of in vitro differentiation of human monocytes into dendritic cells unravels novel transcriptional regulatory interactions. Interface Focus 2021; 11:20200061. [PMID: 34123352 PMCID: PMC8193469 DOI: 10.1098/rsfs.2020.0061] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/15/2021] [Indexed: 12/13/2022] Open
Abstract
Dendritic cells (DCs) are the major specialized antigen-presenting cells, thereby connecting innate and adaptive immunity. Because of their role in establishing adaptive immunity, they constitute promising targets for immunotherapy. Monocytes can differentiate into DCs in vitro in the presence of colony-stimulating factor 2 (CSF2) and interleukin 4 (IL4), activating four signalling pathways (MAPK, JAK/STAT, NFKB and PI3K). However, the downstream transcriptional programme responsible for DC differentiation from monocytes (moDCs) remains unknown. By analysing the scientific literature on moDC differentiation, we established a preliminary logical model that helped us identify missing information regarding the activation of genes responsible for this differentiation, including missing targets for key transcription factors (TFs). Using ChIP-seq and RNA-seq data from the Blueprint consortium, we defined active and inactive promoters, together with differentially expressed genes in monocytes, moDCs and macrophages, which correspond to an alternative cell fate. We then used this functional genomic information to predict novel targets for previously identified TFs. By integrating this information, we refined our model and recapitulated the main established facts regarding moDC differentiation. Prospectively, the resulting model should be useful to develop novel immunotherapies targeting moDCs.
Collapse
Affiliation(s)
- Karen J Nuñez-Reza
- Laboratorio Internacional de Investigación sobre el Genoma Humano, Universidad Nacional Autónoma de México, Juriquilla, México
| | - Aurélien Naldi
- Computational Systems Biology team, Institut de Biologie de l'École Normale Supérieure, Inserm, CNRS, Université PSL, Paris, France
| | - Arantza Sánchez-Jiménez
- Laboratorio Internacional de Investigación sobre el Genoma Humano, Universidad Nacional Autónoma de México, Juriquilla, México
| | - Ana V Leon-Apodaca
- Laboratorio Internacional de Investigación sobre el Genoma Humano, Universidad Nacional Autónoma de México, Juriquilla, México
| | - M Angélica Santana
- Centro de Investigación en Dinámica Celular (IICBA), Universidad Autónoma del Estado de Morelos, Cuernavaca, México
| | - Morgane Thomas-Chollier
- Computational Systems Biology team, Institut de Biologie de l'École Normale Supérieure, Inserm, CNRS, Université PSL, Paris, France
| | - Denis Thieffry
- Computational Systems Biology team, Institut de Biologie de l'École Normale Supérieure, Inserm, CNRS, Université PSL, Paris, France
| | - Alejandra Medina-Rivera
- Laboratorio Internacional de Investigación sobre el Genoma Humano, Universidad Nacional Autónoma de México, Juriquilla, México
| |
Collapse
|
59
|
Erra Díaz F, Ochoa V, Merlotti A, Dantas E, Mazzitelli I, Gonzalez Polo V, Sabatté J, Amigorena S, Segura E, Geffner J. Extracellular Acidosis and mTOR Inhibition Drive the Differentiation of Human Monocyte-Derived Dendritic Cells. Cell Rep 2021; 31:107613. [PMID: 32375041 DOI: 10.1016/j.celrep.2020.107613] [Citation(s) in RCA: 40] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2019] [Revised: 01/31/2020] [Accepted: 04/14/2020] [Indexed: 12/13/2022] Open
Abstract
During inflammation, recruited monocytes can differentiate either into macrophages or dendritic cells (DCs); however, little is known about the environmental factors that determine this cell fate decision. Low extracellular pH is a hallmark of a variety of inflammatory processes and solid tumors. Here, we report that low pH dramatically promotes the differentiation of monocytes into DCs (monocyte-derived DCs [mo-DCs]). This process is associated with a reduction in glucose consumption and lactate production, the upregulation of mitochondrial respiratory chain genes, and the inhibition of mTORC1 activity. Interestingly, we also find that both serum starvation and pharmacological inhibition of mTORC1 markedly promote the differentiation of mo-DCs. Our study contributes to better understanding the mechanisms that govern the differentiation of monocytes into DCs and reveals the role of both extracellular pH and mTORC1 as master regulators of monocyte cell fate.
Collapse
Affiliation(s)
- Fernando Erra Díaz
- INBIRS, Universidad de Buenos Aires (UBA)-CONICET, Buenos Aires, Argentina
| | - Valeria Ochoa
- INBIRS, Universidad de Buenos Aires (UBA)-CONICET, Buenos Aires, Argentina
| | | | - Ezequiel Dantas
- INBIRS, Universidad de Buenos Aires (UBA)-CONICET, Buenos Aires, Argentina
| | - Ignacio Mazzitelli
- INBIRS, Universidad de Buenos Aires (UBA)-CONICET, Buenos Aires, Argentina
| | | | - Juan Sabatté
- INBIRS, Universidad de Buenos Aires (UBA)-CONICET, Buenos Aires, Argentina
| | | | - Elodie Segura
- Institut Curie, PSL Research University, INSERM, U932 Paris, France
| | - Jorge Geffner
- INBIRS, Universidad de Buenos Aires (UBA)-CONICET, Buenos Aires, Argentina.
| |
Collapse
|
60
|
Nagel S, Pommerenke C, Meyer C, Drexler HG. NKL Homeobox Gene VENTX Is Part of a Regulatory Network in Human Conventional Dendritic Cells. Int J Mol Sci 2021; 22:ijms22115902. [PMID: 34072771 PMCID: PMC8198381 DOI: 10.3390/ijms22115902] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2021] [Revised: 05/10/2021] [Accepted: 05/27/2021] [Indexed: 01/09/2023] Open
Abstract
Recently, we documented a hematopoietic NKL-code mapping physiological expression patterns of NKL homeobox genes in human myelopoiesis including monocytes and their derived dendritic cells (DCs). Here, we enlarge this map to include normal NKL homeobox gene expressions in progenitor-derived DCs. Analysis of public gene expression profiling and RNA-seq datasets containing plasmacytoid and conventional dendritic cells (pDC and cDC) demonstrated HHEX activity in both entities while cDCs additionally expressed VENTX. The consequent aim of our study was to examine regulation and function of VENTX in DCs. We compared profiling data of VENTX-positive cDC and monocytes with VENTX-negative pDC and common myeloid progenitor entities and revealed several differentially expressed genes encoding transcription factors and pathway components, representing potential VENTX regulators. Screening of RNA-seq data for 100 leukemia/lymphoma cell lines identified prominent VENTX expression in an acute myelomonocytic leukemia cell line, MUTZ-3 containing inv(3)(q21q26) and t(12;22)(p13;q11) and representing a model for DC differentiation studies. Furthermore, extended gene analyses indicated that MUTZ-3 is associated with the subtype cDC2. In addition to analysis of public chromatin immune-precipitation data, subsequent knockdown experiments and modulations of signaling pathways in MUTZ-3 and control cell lines confirmed identified candidate transcription factors CEBPB, ETV6, EVI1, GATA2, IRF2, MN1, SPIB, and SPI1 and the CSF-, NOTCH-, and TNFa-pathways as VENTX regulators. Live-cell imaging analyses of MUTZ-3 cells treated for VENTX knockdown excluded impacts on apoptosis or induced alteration of differentiation-associated cell morphology. In contrast, target gene analysis performed by expression profiling of knockdown-treated MUTZ-3 cells revealed VENTX-mediated activation of several cDC-specific genes including CSFR1, EGR2, and MIR10A and inhibition of pDC-specific genes like RUNX2. Taken together, we added NKL homeobox gene activities for progenitor-derived DCs to the NKL-code, showing that VENTX is expressed in cDCs but not in pDCs and forms part of a cDC-specific gene regulatory network operating in DC differentiation and function.
Collapse
|
61
|
Nazitto R, Amon LM, Mast FD, Aitchison JD, Aderem A, Johnson JS, Diercks AH. ILF3 Is a Negative Transcriptional Regulator of Innate Immune Responses and Myeloid Dendritic Cell Maturation. THE JOURNAL OF IMMUNOLOGY 2021; 206:2949-2965. [PMID: 34031149 DOI: 10.4049/jimmunol.2001235] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/02/2020] [Accepted: 03/31/2021] [Indexed: 12/31/2022]
Abstract
APCs such as myeloid dendritic cells (DCs) are key sentinels of the innate immune system. In response to pathogen recognition and innate immune stimulation, DCs transition from an immature to a mature state that is characterized by widespread changes in host gene expression, which include the upregulation of cytokines, chemokines, and costimulatory factors to protect against infection. Several transcription factors are known to drive these gene expression changes, but the mechanisms that negatively regulate DC maturation are less well understood. In this study, we identify the transcription factor IL enhancer binding factor 3 (ILF3) as a negative regulator of innate immune responses and DC maturation. Depletion of ILF3 in primary human monocyte-derived DCs led to increased expression of maturation markers and potentiated innate responses during stimulation with viral mimetics or classic innate agonists. Conversely, overexpression of short or long ILF3 isoforms (NF90 and NF110) suppressed DC maturation and innate immune responses. Through mutagenesis experiments, we found that a nuclear localization sequence in ILF3, and not its dual dsRNA-binding domains, was required for this function. Mutation of the domain associated with zinc finger motif of ILF3's NF110 isoform blocked its ability to suppress DC maturation. Moreover, RNA-sequencing analysis indicated that ILF3 regulates genes associated with cholesterol homeostasis in addition to genes associated with DC maturation. Together, our data establish ILF3 as a transcriptional regulator that restrains DC maturation and limits innate immune responses through a mechanism that may intersect with lipid metabolism.
Collapse
Affiliation(s)
- Rodolfo Nazitto
- Department of Immunology, University of Washington School of Medicine, Seattle, WA.,Center for Global Infectious Disease Research, Seattle Children's Research Institute, Seattle, WA
| | - Lynn M Amon
- Center for Infectious Disease Research, Seattle, WA; and
| | - Fred D Mast
- Center for Global Infectious Disease Research, Seattle Children's Research Institute, Seattle, WA
| | - John D Aitchison
- Center for Global Infectious Disease Research, Seattle Children's Research Institute, Seattle, WA
| | - Alan Aderem
- Department of Immunology, University of Washington School of Medicine, Seattle, WA.,Center for Global Infectious Disease Research, Seattle Children's Research Institute, Seattle, WA
| | - Jarrod S Johnson
- Center for Infectious Disease Research, Seattle, WA; and.,Department of Biochemistry, University of Utah, Salt Lake City, UT
| | - Alan H Diercks
- Department of Immunology, University of Washington School of Medicine, Seattle, WA;
| |
Collapse
|
62
|
Song K, Cai H, Zheng H, Yang J, Jin L, Xiao H, Zhang J, Zhao Z, Li X, Zhao W, Li X. Multilevel prioritization of gene regulators associated with consensus molecular subtypes of colorectal cancer. Brief Bioinform 2021; 22:6225088. [PMID: 33855351 DOI: 10.1093/bib/bbab077] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2020] [Revised: 02/16/2021] [Accepted: 02/17/2021] [Indexed: 12/31/2022] Open
Abstract
Consensus molecular subtypes (CMSs) are emerging as critical factor for prognosis and treatment of colorectal cancer. Gene regulators, including chromatin regulator, RNA-binding protein and transcriptional factor, are critical modulators of cancer hallmark, yet little is known regarding the underlying functional mechanism in CMSs. Herein, we identified a core set of 235 functional gene regulators (FGRs) by integrating genome, epigenome, transcriptome and interactome of CMSs. FGRs exhibited significant multi-omics alterations and impacts on cell lines growth, as well as significantly enriched cancer driver genes and pathways. Moreover, common FGRs played different roles in the context of CMSs. In accordance with the immune characteristics of CMSs, we found that the anti-tumor immune pathways were mainly activated by FGRs (e.g. STAT1 and CREBBP) in CMS1, while inhibited by FGRs in CMS2-4. FGRs mediated aberrant expression of ligands, which bind to receptor on immune cells, and modulated tumor immune microenvironment of subtypes. Intriguingly, systematic exploration of datasets using genomic and transcriptome co-similarity reveals the coordinated manner in FGRs act in CMSs to orchestrate their pathways and patients' prognosis. Expression signatures of the FGRs revealed an optimized CMS classifier, which demonstrated 88% concordance with the gold-standard classifier, but avoiding the influence of sample composition. Overall, our integrative analysis identified FGRs to regulate core tumorigenic processes/pathways across CMSs.
Collapse
Affiliation(s)
- Kai Song
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin 150086, China
| | - Hao Cai
- Medical Big Data and Bioinformatics Research Center, First Affiliated Hospital of Gannan Medical University, Ganzhou, China
| | - Hailong Zheng
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin 150086, China
| | - Jing Yang
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin 150086, China
| | - Liangliang Jin
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin 150086, China
| | - Huiting Xiao
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin 150086, China
| | - Jiashuai Zhang
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin 150086, China
| | - Zhangxiang Zhao
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin 150086, China
| | | | | | - Xia Li
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin 150086, China
| |
Collapse
|
63
|
Hatscher L, Amon L, Heger L, Dudziak D. Inflammasomes in dendritic cells: Friend or foe? Immunol Lett 2021; 234:16-32. [PMID: 33848562 DOI: 10.1016/j.imlet.2021.04.002] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2021] [Revised: 03/31/2021] [Accepted: 04/03/2021] [Indexed: 12/14/2022]
Abstract
Inflammasomes are cytosolic multiprotein complexes that crucially contribute to host defense against pathogens but are also involved in the pathogenesis of autoinflammatory diseases. Inflammasome formation leads to activation of effector caspases (caspase-1, 4, 5, or 11), the proteolytic maturation of IL-1β and IL-18 as well as cleavage of the pore-forming protein Gasdermin D. Dendritic cells are major regulators of immune responses as they bridge innate and adaptive immunity. We here summarize the current knowledge on inflammasome expression and formation in murine bone marrow-, human monocyte-derived as well as murine and human primary dendritic cells. Further, we discuss both, the beneficial and detrimental, involvement of inflammasome activation in dendritic cells in cancer, infections, and autoimmune diseases. As inflammasome activation is typically accompanied by Gasdermin d-mediated pyroptosis, which is an inflammatory form of programmed cell death, inflammasome formation in dendritic cells seems ill-advised. Therefore, we propose that hyperactivation, which is inflammasome activation without the induction of pyroptosis, may be a general model of inflammasome activation in dendritic cells to enhance Th1, Th17 as well as cytotoxic T cell responses.
Collapse
Affiliation(s)
- Lukas Hatscher
- Laboratory of Dendritic Cell Biology, Department of Dermatology, University Hospital Erlangen, Friedrich-Alexander University of Erlangen-Nürnberg, 91052, Erlangen, Germany
| | - Lukas Amon
- Laboratory of Dendritic Cell Biology, Department of Dermatology, University Hospital Erlangen, Friedrich-Alexander University of Erlangen-Nürnberg, 91052, Erlangen, Germany
| | - Lukas Heger
- Laboratory of Dendritic Cell Biology, Department of Dermatology, University Hospital Erlangen, Friedrich-Alexander University of Erlangen-Nürnberg, 91052, Erlangen, Germany.
| | - Diana Dudziak
- Laboratory of Dendritic Cell Biology, Department of Dermatology, University Hospital Erlangen, Friedrich-Alexander University of Erlangen-Nürnberg, 91052, Erlangen, Germany; Medical Immunology Campus Erlangen, Germany; Deutsches Zentrum Immuntherapie (DZI), Germany; Comprehensive Cancer Center Erlangen-European Metropolitan Area of Nuremberg (CCC ER-EMN), Germany.
| |
Collapse
|
64
|
Lim CX, Lee B, Geiger O, Passegger C, Beitzinger M, Romberger J, Stracke A, Högenauer C, Stift A, Stoiber H, Poidinger M, Zebisch A, Meister G, Williams A, Flavell RA, Henao-Mejia J, Strobl H. miR-181a Modulation of ERK-MAPK Signaling Sustains DC-SIGN Expression and Limits Activation of Monocyte-Derived Dendritic Cells. Cell Rep 2021; 30:3793-3805.e5. [PMID: 32187550 DOI: 10.1016/j.celrep.2020.02.077] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2018] [Revised: 09/29/2019] [Accepted: 02/20/2020] [Indexed: 01/10/2023] Open
Abstract
DC-SIGN+ monocyte-derived dendritic cells (mo-DCs) play important roles in bacterial infections and inflammatory diseases, but the factors regulating their differentiation and proinflammatory status remain poorly defined. Here, we identify a microRNA, miR-181a, and a molecular mechanism that simultaneously regulate the acquisition of DC-SIGN expression and the activation state of DC-SIGN+ mo-DCs. Specifically, we show that miR-181a promotes DC-SIGN expression during terminal mo-DC differentiation and limits its sensitivity and responsiveness to TLR triggering and CD40 ligation. Mechanistically, miR-181a sustains ERK-MAPK signaling in mo-DCs, thereby enabling the maintenance of high levels of DC-SIGN and a high activation threshold. Low miR-181a levels during mo-DC differentiation, induced by inflammatory signals, do not support the high phospho-ERK signal transduction required for DC-SIGNhi mo-DCs and lead to development of proinflammatory DC-SIGNlo/- mo-DCs. Collectively, our study demonstrates that high DC-SIGN expression levels and a high activation threshold in mo-DCs are linked and simultaneously maintained by miR-181a.
Collapse
Affiliation(s)
- Clarice X Lim
- Otto Loewi Research Center, Chair of Immunology and Pathophysiology, Medical University of Graz, 8010 Graz, Austria; DK Inflammation & Immunity Program, Medical University of Vienna, 1090 Vienna, Austria
| | - Bernett Lee
- Singapore Immunology Network (SIgN), Agency for Science, Technology and Research (A(∗)STAR), Biopolis, 138648 Singapore, Singapore
| | - Olivia Geiger
- Division of Hematology, Medical University of Graz, 8010 Graz, Austria
| | - Christina Passegger
- Otto Loewi Research Center, Chair of Immunology and Pathophysiology, Medical University of Graz, 8010 Graz, Austria
| | - Michaela Beitzinger
- Laboratory for RNA Biology, Biochemistry Center Regensburg (BZR), University of Regensburg, 93053 Regensburg, Germany
| | - Johann Romberger
- Otto Loewi Research Center, Chair of Immunology and Pathophysiology, Medical University of Graz, 8010 Graz, Austria
| | - Anika Stracke
- Otto Loewi Research Center, Chair of Immunology and Pathophysiology, Medical University of Graz, 8010 Graz, Austria
| | - Christoph Högenauer
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, Medical University of Graz, 8010 Graz, Austria
| | - Anton Stift
- Department of Surgery, Medical University of Vienna, 1090 Vienna, Austria
| | - Heribert Stoiber
- Division of Virology, Medical University of Innsbruck, 6020 Innsbruck, Austria
| | - Michael Poidinger
- Singapore Immunology Network (SIgN), Agency for Science, Technology and Research (A(∗)STAR), Biopolis, 138648 Singapore, Singapore
| | - Armin Zebisch
- Division of Hematology, Medical University of Graz, 8010 Graz, Austria; Otto Loewi Research Center for Vascular Biology, Immunology and Inflammation, Division of Pharmacology, Medical University of Graz, 8010 Graz, Austria
| | - Gunter Meister
- Laboratory for RNA Biology, Biochemistry Center Regensburg (BZR), University of Regensburg, 93053 Regensburg, Germany
| | - Adam Williams
- The Jackson Laboratory for Genomic Medicine, Department of Genetics and Genomic Sciences, University of Connecticut Health Center, Farmington, CT 06032, USA
| | - Richard A Flavell
- Department of Immunobiology, Yale University School of Medicine, New Haven, CT 06520, USA; Howard Hughes Medical Institute, Yale University, New Haven, CT 06520, USA
| | - Jorge Henao-Mejia
- Institute for Immunology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Herbert Strobl
- Otto Loewi Research Center, Chair of Immunology and Pathophysiology, Medical University of Graz, 8010 Graz, Austria.
| |
Collapse
|
65
|
Johnson JS, De Veaux N, Rives AW, Lahaye X, Lucas SY, Perot BP, Luka M, Garcia-Paredes V, Amon LM, Watters A, Abdessalem G, Aderem A, Manel N, Littman DR, Bonneau R, Ménager MM. A Comprehensive Map of the Monocyte-Derived Dendritic Cell Transcriptional Network Engaged upon Innate Sensing of HIV. Cell Rep 2021; 30:914-931.e9. [PMID: 31968263 PMCID: PMC7039998 DOI: 10.1016/j.celrep.2019.12.054] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2019] [Revised: 06/25/2019] [Accepted: 12/13/2019] [Indexed: 01/12/2023] Open
Abstract
Transcriptional programming of the innate immune response is pivotal for host protection. However, the transcriptional mechanisms that link pathogen sensing with innate activation remain poorly under-stood. During HIV-1 infection, human dendritic cells (DCs) can detect the virus through an innate sensing pathway, leading to antiviral interferon and DC maturation. Here, we develop an iterative experimental and computational approach to map the HIV-1 innate response circuitry in monocyte-derived DCs (MDDCs). By integrating genome-wide chromatin accessibility with expression kinetics, we infer a gene regulatory network that links 542 transcription factors with 21,862 target genes. We observe that an interferon response is required, yet insufficient, to drive MDDC maturation and identify PRDM1 and RARA as essential regulators of the interferon response and MDDC maturation, respectively. Our work provides a resource for interrogation of regulators of HIV replication and innate immunity, highlighting complexity and cooperativity in the regulatory circuit controlling the response to infection. Pathogen sensing leads to host transcriptional reprogramming to protect against infection. However, it is unclear how transcription factor activity is coordinated across the genome. Johnson et al. integrate chromatin accessibility and gene expression data to infer and validate a gene regulatory network that directs the innate immune response to HIV.
Collapse
Affiliation(s)
- Jarrod S Johnson
- Department of Biochemistry, University of Utah, Salt Lake City, UT 84112, USA; Center for Infectious Disease Research, Seattle, WA 98109, USA.
| | - Nicholas De Veaux
- Flatiron Institute, Center for Computational Biology, Simons Foundation, New York, NY 10010, USA
| | - Alexander W Rives
- Flatiron Institute, Center for Computational Biology, Simons Foundation, New York, NY 10010, USA
| | - Xavier Lahaye
- Immunity and Cancer Department, Institut Curie, PSL Research University, INSERM U932, 75005 Paris, France
| | - Sasha Y Lucas
- Center for Infectious Disease Research, Seattle, WA 98109, USA
| | - Brieuc P Perot
- Laboratory of Inflammatory Responses and Transcriptomic Networks in Diseases, Imagine Institute, INSERM UMR 1163, ATIP-Avenir Team, Université de Paris, 24 Boulevard du Montparnasse, 75015 Paris, France
| | - Marine Luka
- Laboratory of Inflammatory Responses and Transcriptomic Networks in Diseases, Imagine Institute, INSERM UMR 1163, ATIP-Avenir Team, Université de Paris, 24 Boulevard du Montparnasse, 75015 Paris, France
| | - Victor Garcia-Paredes
- Laboratory of Inflammatory Responses and Transcriptomic Networks in Diseases, Imagine Institute, INSERM UMR 1163, ATIP-Avenir Team, Université de Paris, 24 Boulevard du Montparnasse, 75015 Paris, France
| | - Lynn M Amon
- Center for Infectious Disease Research, Seattle, WA 98109, USA
| | - Aaron Watters
- Flatiron Institute, Center for Computational Biology, Simons Foundation, New York, NY 10010, USA
| | - Ghaith Abdessalem
- Laboratory of Inflammatory Responses and Transcriptomic Networks in Diseases, Imagine Institute, INSERM UMR 1163, ATIP-Avenir Team, Université de Paris, 24 Boulevard du Montparnasse, 75015 Paris, France
| | - Alan Aderem
- Center for Infectious Disease Research, Seattle, WA 98109, USA; Department of Immunology, University of Washington School of Medicine, Seattle, WA 98109, USA
| | - Nicolas Manel
- Immunity and Cancer Department, Institut Curie, PSL Research University, INSERM U932, 75005 Paris, France
| | - Dan R Littman
- The Kimmel Center for Biology and Medicine of the Skirball Institute, New York University School of Medicine, New York, NY 10016, USA; Howard Hughes Medical Institute, New York University School of Medicine, New York, NY 10016, USA
| | - Richard Bonneau
- Flatiron Institute, Center for Computational Biology, Simons Foundation, New York, NY 10010, USA; Department of Biology, Center for Genomics and Systems Biology, New York University, New York, NY 10003, USA; Center for Data Science, New York University, New York, NY 10011, USA
| | - Mickaël M Ménager
- Laboratory of Inflammatory Responses and Transcriptomic Networks in Diseases, Imagine Institute, INSERM UMR 1163, ATIP-Avenir Team, Université de Paris, 24 Boulevard du Montparnasse, 75015 Paris, France; The Kimmel Center for Biology and Medicine of the Skirball Institute, New York University School of Medicine, New York, NY 10016, USA.
| |
Collapse
|
66
|
Iwabuchi R, Ide K, Terahara K, Wagatsuma R, Iwaki R, Matsunaga H, Tsunetsugu-Yokota Y, Takeyama H, Takahashi Y. Development of an Inflammatory CD14 + Dendritic Cell Subset in Humanized Mice. Front Immunol 2021; 12:643040. [PMID: 33790912 PMCID: PMC8005643 DOI: 10.3389/fimmu.2021.643040] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2020] [Accepted: 02/16/2021] [Indexed: 12/12/2022] Open
Abstract
Humanized mouse models are attractive experimental models for analyzing the development and functions of human dendritic cells (DCs) in vivo. Although various types of DC subsets, including DC type 3 (DC3s), have been identified in humans, it remains unclear whether humanized mice can reproduce heterogeneous DC subsets. CD14, classically known as a monocyte/macrophage marker, is reported as an indicator of DC3s. We previously observed that some CD14+ myeloid cells expressed CD1c, a pan marker for bona fide conventional DC2 (cDC2s), in humanized mouse models in which human FLT3L and GM-CSF genes were transiently expressed using in vivo transfection (IVT). Here, we aimed to elucidate the identity of CD14+CD1c+ DC-like cells in humanized mouse models. We found that CD14+CD1c+ cells were phenotypically different from cDC2s; CD14+CD1c+ cells expressed CD163 but not CD5, whereas cDC2s expressed CD5 but not CD163. Furthermore, CD14+CD1c+ cells primed and polarized naïve CD4+ T cells toward IFN-γ+ Th1 cells more profoundly than cDC2s. Transcriptional analysis revealed that CD14+CD1c+ cells expressed several DC3-specific transcripts, such as CD163, S100A8, and S100A9, and were clearly segregated from cDC2s and monocytes. When lipopolysaccharide was administered to the humanized mice, the frequency of CD14+CD1c+ cells producing IL-6 and TNF-α was elevated, indicating a pro-inflammatory signature. Thus, humanized mice are able to sustain development of functional CD14+CD1c+ DCs, which are equivalent to DC3 subset observed in humans, and they could be useful for analyzing the development and function of DC3s in vivo.
Collapse
Affiliation(s)
- Ryutaro Iwabuchi
- Department of Immunology, National Institute of Infectious Diseases, Tokyo, Japan.,Department of Life Science and Medical Bioscience, Waseda University, Tokyo, Japan.,Computational Bio Big-Data Open Innovation Laboratory, National Institute of Advanced Industrial Science and Technology, Tokyo, Japan
| | - Keigo Ide
- Department of Life Science and Medical Bioscience, Waseda University, Tokyo, Japan.,Computational Bio Big-Data Open Innovation Laboratory, National Institute of Advanced Industrial Science and Technology, Tokyo, Japan
| | - Kazutaka Terahara
- Department of Immunology, National Institute of Infectious Diseases, Tokyo, Japan
| | - Ryota Wagatsuma
- Department of Life Science and Medical Bioscience, Waseda University, Tokyo, Japan
| | - Rieko Iwaki
- Department of Immunology, National Institute of Infectious Diseases, Tokyo, Japan
| | - Hiroko Matsunaga
- Research Organization for Nano and Life Innovation, Waseda University, Tokyo, Japan
| | - Yasuko Tsunetsugu-Yokota
- Department of Immunology, National Institute of Infectious Diseases, Tokyo, Japan.,Department of Medical Technology, School of Human Sciences, Tokyo University of Technology, Tokyo, Japan
| | - Haruko Takeyama
- Department of Life Science and Medical Bioscience, Waseda University, Tokyo, Japan.,Computational Bio Big-Data Open Innovation Laboratory, National Institute of Advanced Industrial Science and Technology, Tokyo, Japan.,Research Organization for Nano and Life Innovation, Waseda University, Tokyo, Japan.,Institute for Advanced Research of Biosystem Dynamics, Waseda Research Institute for Science and Engineering, Waseda University, Tokyo, Japan
| | - Yoshimasa Takahashi
- Department of Immunology, National Institute of Infectious Diseases, Tokyo, Japan
| |
Collapse
|
67
|
Abstract
As the professional antigen-presenting cells of the immune system, dendritic cells (DCs) sense the microenvironment and shape the ensuing adaptive immune response. DCs can induce both immune activation and immune tolerance according to the peripheral cues. Recent work has established that DCs comprise several phenotypically and functionally heterogeneous subsets that differentially regulate T lymphocyte differentiation. This review summarizes both mouse and human DC subset phenotypes, development, diversification, and function. We focus on advances in our understanding of how different DC subsets regulate distinct CD4+ T helper (Th) cell differentiation outcomes, including Th1, Th2, Th17, T follicular helper, and T regulatory cells. We review DC subset intrinsic properties, local tissue microenvironments, and other immune cells that together determine Th cell differentiation during homeostasis and inflammation.
Collapse
Affiliation(s)
- Xiangyun Yin
- Department of Laboratory Medicine and Department of Immunobiology, Yale University School of Medicine, New Haven, Connecticut 06520, USA;
| | - Shuting Chen
- Department of Laboratory Medicine and Department of Immunobiology, Yale University School of Medicine, New Haven, Connecticut 06520, USA;
| | - Stephanie C Eisenbarth
- Department of Laboratory Medicine and Department of Immunobiology, Yale University School of Medicine, New Haven, Connecticut 06520, USA;
| |
Collapse
|
68
|
The epigenetic pioneer EGR2 initiates DNA demethylation in differentiating monocytes at both stable and transient binding sites. Nat Commun 2021; 12:1556. [PMID: 33692344 PMCID: PMC7946903 DOI: 10.1038/s41467-021-21661-y] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2020] [Accepted: 02/04/2021] [Indexed: 12/13/2022] Open
Abstract
The differentiation of human blood monocytes (MO), the post-mitotic precursors of macrophages (MAC) and dendritic cells (moDC), is accompanied by the active turnover of DNA methylation, but the extent, consequences and mechanisms of DNA methylation changes remain unclear. Here, we profile and compare epigenetic landscapes during IL-4/GM-CSF-driven MO differentiation across the genome and detect several thousand regions that are actively demethylated during culture, both with or without accompanying changes in chromatin accessibility or transcription factor (TF) binding. We further identify TF that are globally associated with DNA demethylation processes. While interferon regulatory factor 4 (IRF4) is found to control hallmark dendritic cell functions with less impact on DNA methylation, early growth response 2 (EGR2) proves essential for MO differentiation as well as DNA methylation turnover at its binding sites. We also show that ERG2 interacts with the 5mC hydroxylase TET2, and its consensus binding sequences show a characteristic DNA methylation footprint at demethylated sites with or without detectable protein binding. Our findings reveal an essential role for EGR2 as epigenetic pioneer in human MO and suggest that active DNA demethylation can be initiated by the TET2-recruiting TF both at stable and transient binding sites. DNA methylation turnover is an essential epigenetic process during development. Here, the authors look at the changes in DNA methylation during the differentiation of post-mitotic human monocytes (MO), and find that EGR2 interacts with TET2 and is required for DNA demethylation at its binding sites; revealing EGR2 as an epigenetic pioneer factor in human MO.
Collapse
|
69
|
Ackaert C, Smiejkowska N, Xavier C, Sterckx YGJ, Denies S, Stijlemans B, Elkrim Y, Devoogdt N, Caveliers V, Lahoutte T, Muyldermans S, Breckpot K, Keyaerts M. Immunogenicity Risk Profile of Nanobodies. Front Immunol 2021; 12:632687. [PMID: 33767701 PMCID: PMC7985456 DOI: 10.3389/fimmu.2021.632687] [Citation(s) in RCA: 106] [Impact Index Per Article: 35.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2020] [Accepted: 02/15/2021] [Indexed: 12/31/2022] Open
Abstract
Nanobodies (Nbs), the variable domains of camelid heavy chain-only antibodies, are a promising class of therapeutics or in vivo imaging reagents entering the clinic. They possess unique characteristics, including a minimal size, providing fast pharmacokinetics, high-target specificity, and an affinity in the (sub-)nanomolar range in conjunction with an easy selection and production, which allow them to outperform conventional antibodies for imaging and radiotherapeutic purposes. As for all protein theranostics, extended safety assessment and investigation of their possible immunogenicity in particular are required. In this study, we assessed the immunogenicity risk profile of two Nbs that are in phase II clinical trials: a first Nb against Human Epidermal growth factor Receptor 2 (HER2) for PET imaging of breast cancer and a second Nb with specificity to the Macrophage Mannose Receptor (MMR) for PET imaging of tumor-associated macrophages. For the anti-HER2 Nb, we show that only one out of 20 patients had a low amount of pre-existing anti-drug antibodies (ADAs), which only marginally increased 3 months after administering the Nb, and without negative effects of safety and pharmacokinetics. Further in vitro immunogenicity assessment assays showed that both non-humanized Nbs were taken up by human dendritic cells but exhibited no or only a marginal capacity to activate dendritic cells or to induce T cell proliferation. From our data, we conclude that monomeric Nbs present a low immunogenicity risk profile, which is encouraging for their future development toward potential clinical applications.
Collapse
Affiliation(s)
- Chloé Ackaert
- Cellular and Molecular Immunology (CMIM), Vrije Universiteit Brussel (VUB), Brussels, Belgium
| | - Natalia Smiejkowska
- Cellular and Molecular Immunology (CMIM), Vrije Universiteit Brussel (VUB), Brussels, Belgium
| | - Catarina Xavier
- In vivo Cellular and Molecular Imaging Laboratory (ICMI), Vrije Universiteit Brussel, Brussels, Belgium
| | - Yann G J Sterckx
- Cellular and Molecular Immunology (CMIM), Vrije Universiteit Brussel (VUB), Brussels, Belgium.,Laboratory of Medical Biochemistry, University of Antwerp (UA), Wilrijk, Belgium
| | | | - Benoit Stijlemans
- Cellular and Molecular Immunology (CMIM), Vrije Universiteit Brussel (VUB), Brussels, Belgium.,Myeloid Cell Immunology Lab, VIB Center for Inflammation Research, Brussels, Belgium
| | - Yvon Elkrim
- Cellular and Molecular Immunology (CMIM), Vrije Universiteit Brussel (VUB), Brussels, Belgium
| | - Nick Devoogdt
- In vivo Cellular and Molecular Imaging Laboratory (ICMI), Vrije Universiteit Brussel, Brussels, Belgium
| | - Vicky Caveliers
- In vivo Cellular and Molecular Imaging Laboratory (ICMI), Vrije Universiteit Brussel, Brussels, Belgium.,Nuclear Medicine Department, UZ Brussel, Brussels, Belgium
| | - Tony Lahoutte
- In vivo Cellular and Molecular Imaging Laboratory (ICMI), Vrije Universiteit Brussel, Brussels, Belgium.,Nuclear Medicine Department, UZ Brussel, Brussels, Belgium
| | - Serge Muyldermans
- Cellular and Molecular Immunology (CMIM), Vrije Universiteit Brussel (VUB), Brussels, Belgium
| | - Karine Breckpot
- Laboratory for Molecular and Cellular Therapy (LMCT), Vrije Universiteit Brussel, Brussels, Belgium
| | - Marleen Keyaerts
- In vivo Cellular and Molecular Imaging Laboratory (ICMI), Vrije Universiteit Brussel, Brussels, Belgium.,Nuclear Medicine Department, UZ Brussel, Brussels, Belgium
| |
Collapse
|
70
|
Rodriguez E, Boelaars K, Brown K, Eveline Li RJ, Kruijssen L, Bruijns SCM, van Ee T, Schetters STT, Crommentuijn MHW, van der Horst JC, van Grieken NCT, van Vliet SJ, Kazemier G, Giovannetti E, Garcia-Vallejo JJ, van Kooyk Y. Sialic acids in pancreatic cancer cells drive tumour-associated macrophage differentiation via the Siglec receptors Siglec-7 and Siglec-9. Nat Commun 2021; 12:1270. [PMID: 33627655 PMCID: PMC7904912 DOI: 10.1038/s41467-021-21550-4] [Citation(s) in RCA: 137] [Impact Index Per Article: 45.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2020] [Accepted: 01/25/2021] [Indexed: 11/09/2022] Open
Abstract
Changes in glycosylation during tumour progression are a key hallmark of cancer. One of the glycan moieties generally overexpressed in cancer are sialic acids, which can induce immunomodulatory properties via binding to Siglec receptors. We here show that Pancreatic Ductal Adenocarcinoma (PDAC) tumour cells present an increased sialylation that can be recognized by Siglec-7 and Siglec-9 on myeloid cells. We identified the expression of the α2,3 sialyltransferases ST3GAL1 and ST3GAL4 as main contributor to the synthesis of ligands for Siglec-7 and Siglec-9 in tumour cells. Analysing the myeloid composition in PDAC, using single cell and bulk transcriptomics data, we identified monocyte-derived macrophages as contributors to the poor clinical outcome. Tumour-derived sialic acids dictate monocyte to macrophage differentiation via signalling through Siglec-7 and Siglec-9. Moreover, triggering of Siglec-9 in macrophages reduce inflammatory programmes, while increasing PD-L1 and IL-10 expression, illustrating that sialic acids modulate different myeloid cells. This work highlights a critical role for sialylated glycans in controlling immune suppression and provides new potential targets for cancer immunotherapy in PDAC.
Collapse
Affiliation(s)
- Ernesto Rodriguez
- Amsterdam UMC, Vrije Universiteit Amsterdam, Department of Molecular Cell Biology and Immunology, Cancer Center Amsterdam, Amsterdam Infection and Immunity Institute, Amsterdam, Netherlands
| | - Kelly Boelaars
- Amsterdam UMC, Vrije Universiteit Amsterdam, Department of Molecular Cell Biology and Immunology, Cancer Center Amsterdam, Amsterdam Infection and Immunity Institute, Amsterdam, Netherlands
| | - Kari Brown
- Amsterdam UMC, Vrije Universiteit Amsterdam, Department of Molecular Cell Biology and Immunology, Cancer Center Amsterdam, Amsterdam Infection and Immunity Institute, Amsterdam, Netherlands
| | - R J Eveline Li
- Amsterdam UMC, Vrije Universiteit Amsterdam, Department of Molecular Cell Biology and Immunology, Cancer Center Amsterdam, Amsterdam Infection and Immunity Institute, Amsterdam, Netherlands
| | - Laura Kruijssen
- Amsterdam UMC, Vrije Universiteit Amsterdam, Department of Molecular Cell Biology and Immunology, Cancer Center Amsterdam, Amsterdam Infection and Immunity Institute, Amsterdam, Netherlands
| | - Sven C M Bruijns
- Amsterdam UMC, Vrije Universiteit Amsterdam, Department of Molecular Cell Biology and Immunology, Cancer Center Amsterdam, Amsterdam Infection and Immunity Institute, Amsterdam, Netherlands
| | - Thomas van Ee
- Amsterdam UMC, Vrije Universiteit Amsterdam, Department of Molecular Cell Biology and Immunology, Cancer Center Amsterdam, Amsterdam Infection and Immunity Institute, Amsterdam, Netherlands
| | - Sjoerd T T Schetters
- Amsterdam UMC, Vrije Universiteit Amsterdam, Department of Molecular Cell Biology and Immunology, Cancer Center Amsterdam, Amsterdam Infection and Immunity Institute, Amsterdam, Netherlands
| | - Matheus H W Crommentuijn
- Amsterdam UMC, Vrije Universiteit Amsterdam, Department of Molecular Cell Biology and Immunology, Cancer Center Amsterdam, Amsterdam Infection and Immunity Institute, Amsterdam, Netherlands
| | - Joost C van der Horst
- Amsterdam UMC, Vrije Universiteit Amsterdam, Department of Molecular Cell Biology and Immunology, Cancer Center Amsterdam, Amsterdam Infection and Immunity Institute, Amsterdam, Netherlands
| | - Nicole C T van Grieken
- Amsterdam UMC, Vrije Universiteit Amsterdam, Department of Pathology, Amsterdam, Netherlands
| | - Sandra J van Vliet
- Amsterdam UMC, Vrije Universiteit Amsterdam, Department of Molecular Cell Biology and Immunology, Cancer Center Amsterdam, Amsterdam Infection and Immunity Institute, Amsterdam, Netherlands
| | - Geert Kazemier
- Amsterdam UMC, Vrije Universiteit Amsterdam, Department of Surgery, Cancer Center Amsterdam, Amsterdam, Netherlands
| | - Elisa Giovannetti
- Amsterdam UMC, Vrije Universiteit Amsterdam, Department of Medical Oncology, Cancer Center Amsterdam, Amsterdam, Netherlands
- Cancer Pharmacology Lab, AIRC Start-Up Unit, Fondazione Pisana per la Scienza, Pisa, Italy
| | - Juan J Garcia-Vallejo
- Amsterdam UMC, Vrije Universiteit Amsterdam, Department of Molecular Cell Biology and Immunology, Cancer Center Amsterdam, Amsterdam Infection and Immunity Institute, Amsterdam, Netherlands
| | - Yvette van Kooyk
- Amsterdam UMC, Vrije Universiteit Amsterdam, Department of Molecular Cell Biology and Immunology, Cancer Center Amsterdam, Amsterdam Infection and Immunity Institute, Amsterdam, Netherlands.
| |
Collapse
|
71
|
Gingival crevicular fluid infiltrating CD14+ monocytes promote inflammation in periodontitis. Saudi J Biol Sci 2021; 28:3069-3075. [PMID: 34012332 PMCID: PMC8116999 DOI: 10.1016/j.sjbs.2021.02.049] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2021] [Revised: 01/31/2021] [Accepted: 02/14/2021] [Indexed: 12/30/2022] Open
Abstract
Periodontitis is a condition that occurs because of inflammation-mediated tissue degeneration. Many studies have been conducted to identify inflammatory molecules in periodontitis, but the well-defined role of cells from the immune system in the progression of periodontitis as well as in gingival tissue degeneration has not been appropriately established. The objective of the present study was to characterize the monocytes isolated from the gingival crevicular fluid (GCF) in patients with periodontitis. GCF was obtained from periodontitis patients and healthy controls. Cytokine levels of CCL2 were evaluated by ELISA in GCF samples. CD14+ monocytes were separated using magnetic sorting from GCF. RT-qPCR was performed to assess the gene expression. Cytometric bead array analysis was performed to analyze the levels of cytokines and chemokines in the secretome of cells. CD14+ monocytes from GCF secreted higher levels of CCL2 and showed elevated expression of genes responsible for monocyte migration. Additionally, upon lipopolysaccharide stimulation, these monocytes secreted higher levels of inflammatory cytokines and chemokines. This investigation aids in understanding the inflammatory microenvironment of periodontitis by characterizing GCF in terms of infiltrated CD14+ monocytes, cytokines, and molecules secreted by these monocytes, which are specific for cellular differentiation.
Collapse
Key Words
- CCL2, C-C motif chemokine ligand 2
- CCL3, C-C motif chemokine ligand 3
- CCL5, C-C motif chemokine ligand 5
- CCR1, C-C chemokine receptor type 1
- CCR2, C-C chemokine receptor type 2
- CCR5, C-C chemokine receptor type 5
- CD11b (ITGAM), Integrin alpha M
- CD14+ monocytes
- CXCR5/BLR1, C-X-C chemokine receptor type 5
- Gingival crevicular fluid
- IL-1β, Interleukin 1 beta
- IL-6, Interleukin 6
- IL-8, Interleukin 8
- Inflammatory cytokines
- Periodontitis
- STAT1, Signal transducer and activator of transcription 1
- STAT2, Signal transducer and activator of transcription 2
- STAT6, Signal transducer and activator of transcription 6
- TNF-α, Tumor necrosis factor-alpha
Collapse
|
72
|
High-Dimensional Phenotyping of Human Myeloid-Derived Suppressor Cells/Tumor-Associated Macrophages in Tissue by Mass Cytometry. Methods Mol Biol 2021; 2236:57-66. [PMID: 33237540 DOI: 10.1007/978-1-0716-1060-2_6] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Myeloid-derived suppressor cells (MDSCs) and tumor-associated macrophages (TAMs) are heterogeneous cells that share myeloid markers and are not easily distinguishable in human tumors due to their lack of specific markers. These cells are a major player in the tumor microenvironment and are involved in the prognosis and physiopathology of various tumors. Here is presented a scheme to decipher these cells by mass cytometry.
Collapse
|
73
|
Mair F, Liechti T. Comprehensive Phenotyping of Human Dendritic Cells and Monocytes. Cytometry A 2020; 99:231-242. [PMID: 33200508 DOI: 10.1002/cyto.a.24269] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2020] [Revised: 11/02/2020] [Accepted: 11/13/2020] [Indexed: 12/11/2022]
Abstract
Professional antigen-presenting cells (APCs), which include dendritic cells (DCs) and monocytes are essential for inducing and steering adaptive T-cell responses. Recent technological developments in single-cell analysis have significantly advanced our understanding of APC subset heterogeneity. To accurately resolve this functional diversity and to account for tissue-specific adaptation, novel phenotyping markers have been described more recently. While some of these largely overlap with traditionally used markers, more fine-grained phenotyping might be essential during inflammatory settings, where the traditional distinction between monocytes and dendritic cells has become blurred. Within this phenotype report, we provide a concise overview of traditional and recently described markers for the phenotyping of DCs and monocytes in the human system.
Collapse
Affiliation(s)
- Florian Mair
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Research Center, 1100 Fairview Ave N, Seattle, Washington, USA
| | - Thomas Liechti
- ImmunoTechnology Section, Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, 40 Convent Drive, Bethesda, Maryland, USA
| |
Collapse
|
74
|
Carenza C, Franzese S, Calcaterra F, Mavilio D, Della Bella S. Comprehensive Phenotyping of Dendritic Cells in Cancer Patients by Flow Cytometry. Cytometry A 2020; 99:218-230. [PMID: 33098618 DOI: 10.1002/cyto.a.24245] [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: 08/27/2020] [Revised: 10/08/2020] [Accepted: 10/19/2020] [Indexed: 12/12/2022]
Abstract
Dendritic cells (DCs) play a crucial role in the complex interplay between tumor cells and the immune system. During the elimination phase of cancer immunoediting, immunostimulatory DCs are critical for the control of tumor growth. During the escape phase, regulatory DCs sustain tumor tolerance and contribute to the development of the immunosuppressive tumor microenvironment that characterizes this phase. Moreover, increasing evidence indicates that DCs are also critical for the success of cancer immunotherapy. Hence, there is increasing need to fully characterize DC subsets and their activatory/inhibitory profile in cancer patients. In this review, we describe the role played by different DC subsets in the different phases of cancer immunoediting, the function exerted by different activatory and inhibitory molecules expressed on DC surface, and the cytokines produced by distinct DC subsets, in order to provide an overview on the DC features that may be useful to be assessed when dealing with the flow cytometric characterization of DCs in cancer patients. © 2020 International Society for Advancement of Cytometry.
Collapse
Affiliation(s)
- Claudia Carenza
- Department of Medical Biotechnologies and Translational Medicine, University of Milan, Milan, Italy.,Unit of Clinical and Experimental Immunology, Humanitas Clinical and Research Center-IRCCS, Rozzano, Milan, Italy
| | - Sara Franzese
- Department of Medical Biotechnologies and Translational Medicine, University of Milan, Milan, Italy.,Unit of Clinical and Experimental Immunology, Humanitas Clinical and Research Center-IRCCS, Rozzano, Milan, Italy
| | - Francesca Calcaterra
- Department of Medical Biotechnologies and Translational Medicine, University of Milan, Milan, Italy.,Unit of Clinical and Experimental Immunology, Humanitas Clinical and Research Center-IRCCS, Rozzano, Milan, Italy
| | - Domenico Mavilio
- Department of Medical Biotechnologies and Translational Medicine, University of Milan, Milan, Italy.,Unit of Clinical and Experimental Immunology, Humanitas Clinical and Research Center-IRCCS, Rozzano, Milan, Italy
| | - Silvia Della Bella
- Department of Medical Biotechnologies and Translational Medicine, University of Milan, Milan, Italy.,Unit of Clinical and Experimental Immunology, Humanitas Clinical and Research Center-IRCCS, Rozzano, Milan, Italy
| |
Collapse
|
75
|
Vazquez-Madrigal C, Lopez S, Grao-Cruces E, Millan-Linares MC, Rodriguez-Martin NM, Martin ME, Alba G, Santa-Maria C, Bermudez B, Montserrat-de la Paz S. Dietary Fatty Acids in Postprandial Triglyceride-Rich Lipoproteins Modulate Human Monocyte-Derived Dendritic Cell Maturation and Activation. Nutrients 2020; 12:nu12103139. [PMID: 33066622 PMCID: PMC7656296 DOI: 10.3390/nu12103139] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2020] [Revised: 10/10/2020] [Accepted: 10/12/2020] [Indexed: 12/12/2022] Open
Abstract
Dietary fatty acids have been demonstrated to modulate systemic inflammation and induce the postprandial inflammatory response of circulating immune cells. We hypothesized that postprandial triglyceride-rich lipoproteins (TRLs) may have acute effects on immunometabolic homeostasis by modulating dendritic cells (DCs), sentinels of the immunity that link innate and adaptive immune systems. In healthy volunteers, saturated fatty acid (SFA)-enriched meal raised serum levels of granulocyte/macrophage colony-stimulating factor GM-CSF (SFAs > monounsaturated fatty acids (MUFAs) = polyunsaturated fatty acids (PUFAs)) in the postprandial period. Autologous TRL-SFAs upregulated the gene expression of DC maturation (CD123 and CCR7) and DC pro-inflammatory activation (CD80 and CD86) genes while downregulating tolerogenic genes (PD-L1 and PD-L2) in human monocyte-derived DCs (moDCs). These effects were reversed with oleic acid-enriched TRLs. Moreover, postprandial SFAs raised IL-12p70 levels, while TRL-MUFAs and TRL-PUFAs increased IL-10 levels in serum of healthy volunteers and in the medium of TRL-treated moDCs. In conclusion, postprandial TRLs are metabolic entities with DC-related tolerogenic activity, and this function is linked to the type of dietary fat in the meal. This study shows that the intake of meals enriched in MUFAs from olive oil, when compared with meals enriched in SFAs, prevents the postprandial production and priming of circulating pro-inflammatory DCs, and promotes tolerogenic response in healthy subjects. However, functional assays with moDCs generated in the presence of different fatty acids and T cells could increase the knowledge of postprandial TRLs’ effects on DC differentiation and function.
Collapse
Affiliation(s)
- Carlos Vazquez-Madrigal
- Department of Medical Biochemistry, Molecular Biology and Immunology, School of Medicine, Universidad de Sevilla, 41009 Seville, Spain; (C.V.-M.); (S.L.); (E.G.-C.); (G.A.)
| | - Soledad Lopez
- Department of Medical Biochemistry, Molecular Biology and Immunology, School of Medicine, Universidad de Sevilla, 41009 Seville, Spain; (C.V.-M.); (S.L.); (E.G.-C.); (G.A.)
| | - Elena Grao-Cruces
- Department of Medical Biochemistry, Molecular Biology and Immunology, School of Medicine, Universidad de Sevilla, 41009 Seville, Spain; (C.V.-M.); (S.L.); (E.G.-C.); (G.A.)
| | - Maria C. Millan-Linares
- Department of Food & Health, Instituto de la Grasa, CSIC, 41013 Seville, Spain; (M.C.M.-L.); (N.M.R.-M.)
| | - Noelia M. Rodriguez-Martin
- Department of Food & Health, Instituto de la Grasa, CSIC, 41013 Seville, Spain; (M.C.M.-L.); (N.M.R.-M.)
| | - Maria E. Martin
- Department of Cell Biology, Faculty of Biology, Universidad de Sevilla, 41012 Seville, Spain; (M.E.M.); (B.B.)
| | - Gonzalo Alba
- Department of Medical Biochemistry, Molecular Biology and Immunology, School of Medicine, Universidad de Sevilla, 41009 Seville, Spain; (C.V.-M.); (S.L.); (E.G.-C.); (G.A.)
| | - Consuelo Santa-Maria
- Department of Biochemistry and Molecular Biology, School of Pharmacy, Universidad de Sevilla, 41012 Seville, Spain;
| | - Beatriz Bermudez
- Department of Cell Biology, Faculty of Biology, Universidad de Sevilla, 41012 Seville, Spain; (M.E.M.); (B.B.)
| | - Sergio Montserrat-de la Paz
- Department of Medical Biochemistry, Molecular Biology and Immunology, School of Medicine, Universidad de Sevilla, 41009 Seville, Spain; (C.V.-M.); (S.L.); (E.G.-C.); (G.A.)
- Correspondence: ; Tel.: +34-954-559-850
| |
Collapse
|
76
|
Chen Y, Wang J, Li X, Hu N, Voelcker NH, Xie X, Elnathan R. Emerging Roles of 1D Vertical Nanostructures in Orchestrating Immune Cell Functions. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2020; 32:e2001668. [PMID: 32844502 PMCID: PMC7461044 DOI: 10.1002/adma.202001668] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/09/2020] [Revised: 05/16/2020] [Indexed: 05/07/2023]
Abstract
Engineered nano-bio cellular interfaces driven by 1D vertical nanostructures (1D-VNS) are set to prompt radical progress in modulating cellular processes at the nanoscale. Here, tuneable cell-VNS interfacial interactions are probed and assessed, highlighting the use of 1D-VNS in immunomodulation, and intracellular delivery into immune cells-both crucial in fundamental and translational biomedical research. With programmable topography and adaptable surface functionalization, 1D-VNS provide unique biophysical and biochemical cues to orchestrate innate and adaptive immunity, both ex vivo and in vivo. The intimate nanoscale cell-VNS interface leads to membrane penetration and cellular deformation, facilitating efficient intracellular delivery of diverse bioactive cargoes into hard-to-transfect immune cells. The unsettled interfacial mechanisms reported to be involved in VNS-mediated intracellular delivery are discussed. By identifying up-to-date progress and fundamental challenges of current 1D-VNS technology in immune-cell manipulation, it is hoped that this report gives timely insights for further advances in developing 1D-VNS as a safe, universal, and highly scalable platform for cell engineering and enrichment in advanced cancer immunotherapy such as chimeric antigen receptor-T therapy.
Collapse
Affiliation(s)
- Yaping Chen
- Monash Institute of Pharmaceutical SciencesMonash University381 Royal ParadeParkvilleVIC3052Australia
- Melbourne Centre for NanofabricationVictorian Node of the Australian National Fabrication Facility151 Wellington RoadClayton3168Australia
| | - Ji Wang
- The First Affiliated Hospital of Sun Yat‐sen UniversitySun Yat‐sen UniversityGuangzhou510006China
| | - Xiangling Li
- State Key Laboratory of Optoelectronic Materials and TechnologiesSchool of Electronics and Information TechnologySun Yat‐sen UniversityGuangzhou510006China
| | - Ning Hu
- State Key Laboratory of Optoelectronic Materials and TechnologiesSchool of Electronics and Information TechnologySun Yat‐sen UniversityGuangzhou510006China
| | - Nicolas H. Voelcker
- Monash Institute of Pharmaceutical SciencesMonash University381 Royal ParadeParkvilleVIC3052Australia
- Melbourne Centre for NanofabricationVictorian Node of the Australian National Fabrication Facility151 Wellington RoadClayton3168Australia
- Department of Materials Science and EngineeringMonash University22 Alliance LaneClaytonVIC3168Australia
- Commonwealth Scientific and Industrial Research Organisation (CSIRO)ClaytonVIC3168Australia
- INM‐Leibniz Institute for New MaterialsCampus D2 2Saarbrücken66123Germany
| | - Xi Xie
- The First Affiliated Hospital of Sun Yat‐sen UniversitySun Yat‐sen UniversityGuangzhou510006China
- State Key Laboratory of Optoelectronic Materials and TechnologiesSchool of Electronics and Information TechnologySun Yat‐sen UniversityGuangzhou510006China
| | - Roey Elnathan
- Monash Institute of Pharmaceutical SciencesMonash University381 Royal ParadeParkvilleVIC3052Australia
- Melbourne Centre for NanofabricationVictorian Node of the Australian National Fabrication Facility151 Wellington RoadClayton3168Australia
- Department of Materials Science and EngineeringMonash University22 Alliance LaneClaytonVIC3168Australia
| |
Collapse
|
77
|
Carvalheiro T, Garcia S, Pascoal Ramos MI, Giovannone B, Radstake TRDJ, Marut W, Meyaard L. Leukocyte Associated Immunoglobulin Like Receptor 1 Regulation and Function on Monocytes and Dendritic Cells During Inflammation. Front Immunol 2020; 11:1793. [PMID: 32973751 PMCID: PMC7466540 DOI: 10.3389/fimmu.2020.01793] [Citation(s) in RCA: 36] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2020] [Accepted: 07/06/2020] [Indexed: 12/30/2022] Open
Abstract
Inhibitory receptors are crucial immune regulators and are essential to prevent exacerbated responses, thus contributing to immune homeostasis. Leukocyte associated immunoglobulin like receptor 1 (LAIR-1) is an immune inhibitory receptor which has collagen and collagen domain containing proteins as ligands. LAIR-1 is broadly expressed on immune cells and has a large availability of ligands in both circulation and tissues, implicating a need for tight regulation of this interaction. In the current study, we sought to examine the regulation and function of LAIR-1 on monocyte, dendritic cell (DC) and macrophage subtypes, using different in vitro models. We found that LAIR-1 is highly expressed on intermediate monocytes as well as on plasmacytoid DCs. LAIR-1 is also expressed on skin immune cells, mainly on tissue CD14+ cells, macrophages and CD1c+ DCs. In vitro, monocyte and type-2 conventional DC stimulation leads to LAIR-1 upregulation, which may reflect the importance of LAIR-1 as negative regulator under inflammatory conditions. Indeed, we demonstrate that LAIR-1 ligation on monocytes inhibits toll like receptor (TLR)4 and Interferon (IFN)-α- induced signals. Furthermore, LAIR-1 is downregulated on GM-CSF and IFN-γ monocyte-derived macrophages and monocyte-derived DCs. In addition, LAIR-1 triggering during monocyte derived-DC differentiation results in significant phenotypic changes, as well as a different response to TLR4 and IFN-α stimulation. This indicates a role for LAIR-1 in skewing DC function, which impacts the cytokine expression profile of these cells. In conclusion, we demonstrate that LAIR-1 is consistently upregulated on monocytes and DC during the inflammatory phase of the immune response and tends to restore its expression during the resolution phase. Under inflammatory conditions, LAIR-1 has an inhibitory function, pointing toward to a potential intervention opportunity targeting LAIR-1 in inflammatory conditions.
Collapse
Affiliation(s)
- Tiago Carvalheiro
- Center for Translational Immunology, University Medical Center Utrecht, University of Utrecht, Utrecht, Netherlands.,Department of Rheumatology & Clinical Immunology, University Medical Center Utrecht, University of Utrecht, Utrecht, Netherlands
| | - Samuel Garcia
- Center for Translational Immunology, University Medical Center Utrecht, University of Utrecht, Utrecht, Netherlands.,Department of Rheumatology & Clinical Immunology, University Medical Center Utrecht, University of Utrecht, Utrecht, Netherlands.,Rheumatology & Immuno-Mediated Diseases Research Group (IRIDIS), Galicia Sur Health Research Institute (IIS Galicia Sur), SERGAS-UVIGO, Vigo, Spain.,Rheumatology Department, University Hospital Complex of Vigo, Vigo, Spain
| | - M Inês Pascoal Ramos
- Center for Translational Immunology, University Medical Center Utrecht, University of Utrecht, Utrecht, Netherlands.,Oncode Institute, Utrecht, Netherlands
| | - Barbara Giovannone
- Center for Translational Immunology, University Medical Center Utrecht, University of Utrecht, Utrecht, Netherlands.,Department of Dermatology, University Medical Center Utrecht, Utrecht University, Utrecht, Netherlands
| | - Timothy R D J Radstake
- Center for Translational Immunology, University Medical Center Utrecht, University of Utrecht, Utrecht, Netherlands.,Department of Rheumatology & Clinical Immunology, University Medical Center Utrecht, University of Utrecht, Utrecht, Netherlands
| | - Wioleta Marut
- Center for Translational Immunology, University Medical Center Utrecht, University of Utrecht, Utrecht, Netherlands.,Department of Rheumatology & Clinical Immunology, University Medical Center Utrecht, University of Utrecht, Utrecht, Netherlands
| | - Linde Meyaard
- Center for Translational Immunology, University Medical Center Utrecht, University of Utrecht, Utrecht, Netherlands.,Oncode Institute, Utrecht, Netherlands
| |
Collapse
|
78
|
Bourdely P, Anselmi G, Vaivode K, Ramos RN, Missolo-Koussou Y, Hidalgo S, Tosselo J, Nuñez N, Richer W, Vincent-Salomon A, Saxena A, Wood K, Lladser A, Piaggio E, Helft J, Guermonprez P. Transcriptional and Functional Analysis of CD1c + Human Dendritic Cells Identifies a CD163 + Subset Priming CD8 +CD103 + T Cells. Immunity 2020; 53:335-352.e8. [PMID: 32610077 PMCID: PMC7445430 DOI: 10.1016/j.immuni.2020.06.002] [Citation(s) in RCA: 205] [Impact Index Per Article: 51.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2019] [Revised: 04/15/2020] [Accepted: 05/29/2020] [Indexed: 02/04/2023]
Abstract
Dendritic cells (DCs) are antigen-presenting cells controlling T cell activation. In humans, the diversity, ontogeny, and functional capabilities of DC subsets are not fully understood. Here, we identified circulating CD88-CD1c+CD163+ DCs (called DC3s) as immediate precursors of inflammatory CD88-CD14+CD1c+CD163+FcεRI+ DCs. DC3s develop via a specific pathway activated by GM-CSF, independent of cDC-restricted (CDP) and monocyte-restricted (cMoP) progenitors. Like classical DCs but unlike monocytes, DC3s drove activation of naive T cells. In vitro, DC3s displayed a distinctive ability to prime CD8+ T cells expressing a tissue homing signature and the epithelial homing alpha-E integrin (CD103) through transforming growth factor β (TGF-β) signaling. In vivo, DC3s infiltrated luminal breast cancer primary tumors, and DC3 infiltration correlated positively with CD8+CD103+CD69+ tissue-resident memory T cells. Together, these findings define DC3s as a lineage of inflammatory DCs endowed with a strong potential to regulate tumor immunity.
Collapse
Affiliation(s)
- Pierre Bourdely
- Centre for Inflammation Biology and Cancer Immunology, The Peter Gorer Department of Immunobiology, School of Immunology & Microbial Sciences, King's College London, London, UK; Cancer Research UK King's Health Partner Cancer Centre, King's College London, London, UK
| | - Giorgio Anselmi
- Centre for Inflammation Biology and Cancer Immunology, The Peter Gorer Department of Immunobiology, School of Immunology & Microbial Sciences, King's College London, London, UK; Cancer Research UK King's Health Partner Cancer Centre, King's College London, London, UK
| | - Kristine Vaivode
- Centre for Inflammation Biology and Cancer Immunology, The Peter Gorer Department of Immunobiology, School of Immunology & Microbial Sciences, King's College London, London, UK; Cancer Research UK King's Health Partner Cancer Centre, King's College London, London, UK
| | - Rodrigo Nalio Ramos
- PSL Research University, Institut Curie Research Center, Translational Immunotherapy Team, INSERM U932, Paris, France
| | - Yoann Missolo-Koussou
- PSL Research University, Institut Curie Research Center, Translational Immunotherapy Team, INSERM U932, Paris, France
| | - Sofia Hidalgo
- PSL Research University, Institut Curie Research Center, Translational Immunotherapy Team, INSERM U932, Paris, France; Laboratory of Immuno-oncology, Fundación Ciencia & Vida, Santiago, Chile
| | - Jimena Tosselo
- PSL Research University, Institut Curie Research Center, Translational Immunotherapy Team, INSERM U932, Paris, France
| | - Nicolas Nuñez
- PSL Research University, Institut Curie Research Center, Translational Immunotherapy Team, INSERM U932, Paris, France
| | - Wilfrid Richer
- PSL Research University, Institut Curie Research Center, Translational Immunotherapy Team, INSERM U932, Paris, France
| | - Anne Vincent-Salomon
- PSL Research University, Institut Curie, Department of Biopathology, Paris, France
| | - Alka Saxena
- National Institute of Health Research Biomedical Research Centre at Guy's and St Thomas' Hospital and King's College London, London, UK
| | - Kristie Wood
- National Institute of Health Research Biomedical Research Centre at Guy's and St Thomas' Hospital and King's College London, London, UK
| | - Alvaro Lladser
- Laboratory of Immuno-oncology, Fundación Ciencia & Vida, Santiago, Chile; Facultad de Medicina y Ciencia, Universidad San Sebastián, Santiago, Chile
| | - Eliane Piaggio
- PSL Research University, Institut Curie Research Center, Translational Immunotherapy Team, INSERM U932, Paris, France
| | - Julie Helft
- PSL Research University, Institut Curie Research Center, Translational Immunotherapy Team, INSERM U932, Paris, France
| | - Pierre Guermonprez
- Centre for Inflammation Biology and Cancer Immunology, The Peter Gorer Department of Immunobiology, School of Immunology & Microbial Sciences, King's College London, London, UK; Cancer Research UK King's Health Partner Cancer Centre, King's College London, London, UK; Université de Paris, Centre for Inflammation Research, CNRS ERL8252, INSERM1149 Paris, France.
| |
Collapse
|
79
|
Cirovic B, de Bree LCJ, Groh L, Blok BA, Chan J, van der Velden WJFM, Bremmers MEJ, van Crevel R, Händler K, Picelli S, Schulte-Schrepping J, Klee K, Oosting M, Koeken VACM, van Ingen J, Li Y, Benn CS, Schultze JL, Joosten LAB, Curtis N, Netea MG, Schlitzer A. BCG Vaccination in Humans Elicits Trained Immunity via the Hematopoietic Progenitor Compartment. Cell Host Microbe 2020; 28:322-334.e5. [PMID: 32544459 PMCID: PMC7295478 DOI: 10.1016/j.chom.2020.05.014] [Citation(s) in RCA: 272] [Impact Index Per Article: 68.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2020] [Revised: 04/16/2020] [Accepted: 05/12/2020] [Indexed: 01/13/2023]
Abstract
Induction of trained immunity by Bacille-Calmette-Guérin (BCG) vaccination mediates beneficial heterologous effects, but the mechanisms underlying its persistence and magnitude remain elusive. In this study, we show that BCG vaccination in healthy human volunteers induces a persistent transcriptional program connected to myeloid cell development and function within the hematopoietic stem and progenitor cell (HSPC) compartment in the bone marrow. We identify hepatic nuclear factor (HNF) family members 1a and b as crucial regulators of this transcriptional shift. These findings are corroborated by higher granulocyte numbers in BCG-vaccinated infants, HNF1 SNP variants that correlate with trained immunity, and elevated serum concentrations of the HNF1 target alpha-1 antitrypsin. Additionally, transcriptomic HSPC remodeling was epigenetically conveyed to peripheral CD14+ monocytes, displaying an activated transcriptional signature three months after BCG vaccination. Taken together, transcriptomic, epigenomic, and functional reprogramming of HSPCs and peripheral monocytes is a hallmark of BCG-induced trained immunity in humans.
Collapse
Affiliation(s)
- Branko Cirovic
- Quantitative Systems Biology, Life & Medical Sciences Institute, University of Bonn, 53115 Bonn, Germany
| | - L Charlotte J de Bree
- Department of Internal Medicine, Radboud Institute of Molecular Life Sciences (RIMLS) and Radboud Center for Infectious Diseases (RCI), Radboud University Nijmegen Medical Centre, 6526 GA Nijmegen, the Netherlands; Research Center for Vitamins and Vaccines, Bandim Health Project, Statens Serum Institut, Copenhagen, Denmark; Odense Patient Data Explorative Network, University of Southern Denmark/Odense University Hospital, Odense, Denmark
| | - Laszlo Groh
- Department of Internal Medicine, Radboud Institute of Molecular Life Sciences (RIMLS) and Radboud Center for Infectious Diseases (RCI), Radboud University Nijmegen Medical Centre, 6526 GA Nijmegen, the Netherlands
| | - Bas A Blok
- Department of Internal Medicine, Radboud Institute of Molecular Life Sciences (RIMLS) and Radboud Center for Infectious Diseases (RCI), Radboud University Nijmegen Medical Centre, 6526 GA Nijmegen, the Netherlands; Research Center for Vitamins and Vaccines, Bandim Health Project, Statens Serum Institut, Copenhagen, Denmark; Odense Patient Data Explorative Network, University of Southern Denmark/Odense University Hospital, Odense, Denmark
| | - Joyce Chan
- Department of Paediatrics, The University of Melbourne & Murdoch Children's Research Institute, The Royal Children's Hospital Melbourne, Parkville, Australia
| | | | - M E J Bremmers
- Department of Haematology, Radboud University Medical Center, Nijmegen, the Netherlands
| | - Reinout van Crevel
- Department of Internal Medicine, Radboud Institute of Molecular Life Sciences (RIMLS) and Radboud Center for Infectious Diseases (RCI), Radboud University Nijmegen Medical Centre, 6526 GA Nijmegen, the Netherlands
| | - Kristian Händler
- Single Cell Genomics and Epigenomics Unit at the German Center for Neurodegenerative Diseases and the University of Bonn, 53175 Bonn, Germany
| | - Simone Picelli
- Single Cell Genomics and Epigenomics Unit at the German Center for Neurodegenerative Diseases and the University of Bonn, 53175 Bonn, Germany
| | - Jonas Schulte-Schrepping
- Genomics and Immunoregulation, Life & Medical Sciences Institute, University of Bonn, 53115 Bonn, Germany
| | - Kathrin Klee
- Genomics and Immunoregulation, Life & Medical Sciences Institute, University of Bonn, 53115 Bonn, Germany
| | - Marije Oosting
- Department of Internal Medicine, Radboud Institute of Molecular Life Sciences (RIMLS) and Radboud Center for Infectious Diseases (RCI), Radboud University Nijmegen Medical Centre, 6526 GA Nijmegen, the Netherlands
| | - Valerie A C M Koeken
- Department of Internal Medicine, Radboud Institute of Molecular Life Sciences (RIMLS) and Radboud Center for Infectious Diseases (RCI), Radboud University Nijmegen Medical Centre, 6526 GA Nijmegen, the Netherlands
| | - Jakko van Ingen
- Department of Medical Microbiology, Radboud University Medical Center, Nijmegen, the Netherlands
| | - Yang Li
- Department of Internal Medicine, Radboud Institute of Molecular Life Sciences (RIMLS) and Radboud Center for Infectious Diseases (RCI), Radboud University Nijmegen Medical Centre, 6526 GA Nijmegen, the Netherlands; Centre for Individualised Infection Medicine (CiiM) & TWINCORE, joint ventures between the Helmholtz-Centre for Infection Research (HZI) and the Hannover Medical School (MHH), 30625 Hannover, Germany
| | - Christine S Benn
- Research Center for Vitamins and Vaccines, Bandim Health Project, Statens Serum Institut, Copenhagen, Denmark; Odense Patient Data Explorative Network, University of Southern Denmark/Odense University Hospital, Odense, Denmark
| | - Joachim L Schultze
- Single Cell Genomics and Epigenomics Unit at the German Center for Neurodegenerative Diseases and the University of Bonn, 53175 Bonn, Germany; Genomics and Immunoregulation, Life & Medical Sciences Institute, University of Bonn, 53115 Bonn, Germany
| | - Leo A B Joosten
- Department of Internal Medicine, Radboud Institute of Molecular Life Sciences (RIMLS) and Radboud Center for Infectious Diseases (RCI), Radboud University Nijmegen Medical Centre, 6526 GA Nijmegen, the Netherlands
| | - Nigel Curtis
- Department of Paediatrics, The University of Melbourne & Murdoch Children's Research Institute, The Royal Children's Hospital Melbourne, Parkville, Australia
| | - Mihai G Netea
- Department of Internal Medicine, Radboud Institute of Molecular Life Sciences (RIMLS) and Radboud Center for Infectious Diseases (RCI), Radboud University Nijmegen Medical Centre, 6526 GA Nijmegen, the Netherlands; Immunology and Metabolism, Life & Medical Sciences Institute, University of Bonn, 53115 Bonn, Germany.
| | - Andreas Schlitzer
- Quantitative Systems Biology, Life & Medical Sciences Institute, University of Bonn, 53115 Bonn, Germany; Single Cell Genomics and Epigenomics Unit at the German Center for Neurodegenerative Diseases and the University of Bonn, 53175 Bonn, Germany.
| |
Collapse
|
80
|
Schetters STT, Kruijssen LJW, Crommentuijn MHW, Kalay H, den Haan JMM, van Kooyk Y. Immunological dynamics after subcutaneous immunization with a squalene-based oil-in-water adjuvant. FASEB J 2020; 34:12406-12418. [PMID: 33411367 PMCID: PMC7496326 DOI: 10.1096/fj.202000848r] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2020] [Revised: 06/24/2020] [Accepted: 07/06/2020] [Indexed: 12/25/2022]
Abstract
The clinically successful adjuvant MF59 is used in seasonal influenza vaccines, which is proposed to enhance immunity by creating an immune-competent microenvironment in the muscle that allows recruitment of immune cells that drive adaptive immune responses. Here, we examined whether the clinically successful adjuvants MF59/AddaVax could be used for subcutaneous use and how antigen delivery can be synergized with cellular dynamics at the vaccination site. Subcutaneous injection of AddaVax leads to thickening of the skin, characterized by a neutrophil-monocyte recruitment sequence. Skin-infiltrating CCR2+Ly6Chigh monocytes showed differentiation to CD11b+Ly6C+MHCII+CD11c+CD64+ monocyte-derived DCs over time in the hypodermal layers of the skin, expressing high levels of CD209a/mDC-SIGN. Surprisingly, skin thickening was accompanied with increased white adipose tissue highly enriched with monocytes. Analysis of the skin-draining lymph nodes revealed early increases in neutrophils and moDCs at 12 hours after injection and later increases in migratory cDC2s. Subcutaneous vaccination with AddaVax enhanced antigen-specific CD8+ and CD4+ T cell responses, while moDC targeting using antigen-coupled CD209a antibody additionally boosted humoral responses. Hence, oil-in-water emulsions provide an attractive immune modulatory adjuvants aimed at increasing cellular responses, as well as antibody responses when combined with moDC targeting.
Collapse
Affiliation(s)
- Sjoerd T T Schetters
- Department of Molecular Cell Biology and Immunology, Amsterdam University Medical Center, Amsterdam, the Netherlands.,Cancer Center Amsterdam, Amsterdam Infection and Immunity Institute, VU University, Amsterdam, the Netherlands
| | - Laura J W Kruijssen
- Department of Molecular Cell Biology and Immunology, Amsterdam University Medical Center, Amsterdam, the Netherlands.,Cancer Center Amsterdam, Amsterdam Infection and Immunity Institute, VU University, Amsterdam, the Netherlands
| | - Matheus H W Crommentuijn
- Department of Molecular Cell Biology and Immunology, Amsterdam University Medical Center, Amsterdam, the Netherlands.,Cancer Center Amsterdam, Amsterdam Infection and Immunity Institute, VU University, Amsterdam, the Netherlands
| | - Hakan Kalay
- Department of Molecular Cell Biology and Immunology, Amsterdam University Medical Center, Amsterdam, the Netherlands.,Cancer Center Amsterdam, Amsterdam Infection and Immunity Institute, VU University, Amsterdam, the Netherlands
| | - Joke M M den Haan
- Department of Molecular Cell Biology and Immunology, Amsterdam University Medical Center, Amsterdam, the Netherlands.,Cancer Center Amsterdam, Amsterdam Infection and Immunity Institute, VU University, Amsterdam, the Netherlands
| | - Yvette van Kooyk
- Department of Molecular Cell Biology and Immunology, Amsterdam University Medical Center, Amsterdam, the Netherlands.,Cancer Center Amsterdam, Amsterdam Infection and Immunity Institute, VU University, Amsterdam, the Netherlands
| |
Collapse
|
81
|
Bsat M, Chapuy L, Rubio M, Sarfati M. A two-step human culture system replicates intestinal monocyte maturation cascade: Conversion of tissue-like inflammatory monocytes into macrophages. Eur J Immunol 2020; 50:1676-1690. [PMID: 32557554 DOI: 10.1002/eji.202048555] [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: 01/17/2020] [Revised: 05/05/2020] [Accepted: 06/17/2020] [Indexed: 12/14/2022]
Abstract
Monocyte maturation program into macrophages (MΦ) is well defined in murine gut under homeostatic or inflammatory conditions. Obviously, in vivo tracking of monocytes in inflamed tissues remains difficult in humans. Furthermore, in vitro models fall short in generating the surrogates of transient extravasated tissue inflammatory monocytes. Here, we aimed to unravel environmental cues that replicated the human monocyte "waterfall" process in vitro by first, generating tissue-like inflammatory monocytes, which were then shifted toward MΦ. Purified CD14+ CD16- monocytes, cultured with granulocyte-macrophage colony-stimulating factor (GM-CSF), IFN-γ and IL23, differentiated into CD14+ CD163- cells that displayed a monocyte-like morphology. In vitro generated inflammatory CD14+ CD163- (inflammatory monocyte-like cells) cells promoted IL-1β-dependent memory Th17 and Th17/Th1 responses, like the CD14+ CD163- mo-like cells that accumulate in inflamed colon of Crohn's disease patients. Next, in vitro generated inflammatory monocyte-like cells converted to functional CD163+ MΦ following exposure to TGF-β and IL10. Gene set enrichment analysis further revealed a shared molecular signature between converted CD163+ MΦ and MΦ detected in various inflamed nonlymphoid and lymphoid diseased tissues. Our findings propose a two-step in vitro culture that recapitulates human monocyte maturation cascade in inflamed tissue. Manipulation of this process might open therapeutic avenues for chronic inflammatory disorders.
Collapse
Affiliation(s)
- Marwa Bsat
- Immunoregulation Laboratory, Centre de Recherche du Centre Hospitalier de l'Université de Montréal (CRCHUM), Montreal, Quebec, Canada
| | - Laurence Chapuy
- Immunoregulation Laboratory, Centre de Recherche du Centre Hospitalier de l'Université de Montréal (CRCHUM), Montreal, Quebec, Canada
| | - Manuel Rubio
- Immunoregulation Laboratory, Centre de Recherche du Centre Hospitalier de l'Université de Montréal (CRCHUM), Montreal, Quebec, Canada
| | - Marika Sarfati
- Immunoregulation Laboratory, Centre de Recherche du Centre Hospitalier de l'Université de Montréal (CRCHUM), Montreal, Quebec, Canada
| |
Collapse
|
82
|
Branchett WJ, O'Garra A, Lloyd CM. Transcriptomic analysis reveals diverse gene expression changes in airway macrophages during experimental allergic airway disease. Wellcome Open Res 2020; 5:101. [PMID: 32587903 PMCID: PMC7309452 DOI: 10.12688/wellcomeopenres.15875.2] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/17/2020] [Indexed: 12/26/2022] Open
Abstract
Background: Airway macrophages (AMs) are the most abundant leukocytes in the healthy airway lumen and have a highly specialised but plastic phenotype that is governed by signals in the local microenvironment. AMs are thought to maintain immunological homeostasis in the steady state, but have also been implicated in the pathogenesis of allergic airway disease (AAD). In this study, we aimed to better understand these potentially contrasting AM functions using transcriptomic analysis. Methods: Bulk RNA sequencing was performed on AMs (CD11c + Siglec F + CD64 + CD45 + SSC hi) flow cytometry sorted from C57BL/6 mice during experimental AAD driven by repeated house dust mite inhalation (AMs HDM), compared to control AMs from non-allergic mice. Differentially expressed genes were further analysed by hierarchical clustering and biological pathway analysis. Results: AMs HDM showed increased expression of genes associated with antigen presentation, inflammatory cell recruitment and tissue repair, including several chemokine and matrix metalloproteinase genes. This was accompanied by increased expression of mitochondrial electron transport chain subunit genes and the retinoic acid biosynthetic enzyme gene Raldh2. Conversely, AMs HDM displayed decreased expression of a number of cell cycle genes, genes related to cytoskeletal functions and a subset of genes implicated in antimicrobial innate immunity, such as Tlr5, Il18 and Tnf. Differential gene expression in AMs HDM was consistent with upstream effects of the cytokines IL-4 and IFN-γ, both of which were present at increased concentrations in lung tissue after HDM treatment. Conclusions: These data highlight diverse gene expression changes in the total AM population in a clinically relevant mouse model of AAD, collectively suggestive of contributions to inflammation and tissue repair/remodelling, but with decreases in certain steady state cellular and immunological functions.
Collapse
Affiliation(s)
- William J. Branchett
- National Heart and Lung Institute, Imperial College London, London, SW7 2AZ, UK
- Asthma UK Centre in Allergic Mechanisms of Asthma, Imperial College London, London, W2 1NY, UK
| | - Anne O'Garra
- National Heart and Lung Institute, Imperial College London, London, SW7 2AZ, UK
- Laboratory of Immunoregulation and Infection, The Francis Crick Institute, London, NW1 1AT, UK
| | - Clare M. Lloyd
- National Heart and Lung Institute, Imperial College London, London, SW7 2AZ, UK
- Asthma UK Centre in Allergic Mechanisms of Asthma, Imperial College London, London, W2 1NY, UK
| |
Collapse
|
83
|
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: 44] [Impact Index Per Article: 11.0] [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]
|
84
|
Branchett WJ, O'Garra A, Lloyd CM. Transcriptomic analysis reveals diverse gene expression changes in airway macrophages during experimental allergic airway disease. Wellcome Open Res 2020; 5:101. [PMID: 32587903 PMCID: PMC7309452 DOI: 10.12688/wellcomeopenres.15875.1] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/07/2020] [Indexed: 02/12/2024] Open
Abstract
Background: Airway macrophages (AMs) are the most abundant leukocytes in the healthy airway lumen and have a highly specialised but plastic phenotype that is governed by signals in the local microenvironment. AMs are thought to maintain immunological homeostasis in the steady state, but have also been implicated in the pathogenesis of allergic airway disease (AAD). In this study, we aimed to better understand these potentially contrasting AM functions using transcriptomic analysis. Methods: Bulk RNA sequencing was performed on AMs flow cytometry sorted from C57BL/6 mice during experimental AAD driven by repeated house dust mite inhalation (AMs HDM), compared to control AMs from non-allergic mice. Differentially expressed genes were further analysed by hierarchical clustering and biological pathway analysis. Results: AMs HDM showed increased expression of genes associated with antigen presentation, inflammatory cell recruitment and tissue repair, including several chemokine and matrix metalloproteinase genes. This was accompanied by increased expression of mitochondrial electron transport chain subunit genes and the retinoic acid biosynthetic enzyme gene Raldh2. Conversely, AMs HDM displayed decreased expression of a number of cell cycle genes, genes related to cytoskeletal functions and a subset of genes implicated in antimicrobial innate immunity, such as Tlr5, Il18 and Tnf. Differential gene expression in AMs HDM was consistent with upstream effects of the cytokines IL-4 and IFN-γ, both of which were present at increased concentrations in lung tissue after HDM treatment. Conclusions: These data highlight diverse gene expression changes in the total AM population in a clinically relevant mouse model of AAD, collectively suggestive of contributions to inflammation and tissue repair/remodelling, but with decreases in certain steady state cellular and immunological functions.
Collapse
Affiliation(s)
- William J. Branchett
- National Heart and Lung Institute, Imperial College London, London, SW7 2AZ, UK
- Asthma UK Centre in Allergic Mechanisms of Asthma, Imperial College London, London, W2 1NY, UK
| | - Anne O'Garra
- National Heart and Lung Institute, Imperial College London, London, SW7 2AZ, UK
- Laboratory of Immunoregulation and Infection, The Francis Crick Institute, London, NW1 1AT, UK
| | - Clare M. Lloyd
- National Heart and Lung Institute, Imperial College London, London, SW7 2AZ, UK
- Asthma UK Centre in Allergic Mechanisms of Asthma, Imperial College London, London, W2 1NY, UK
| |
Collapse
|
85
|
Yu L, Fernandez S, Brock G. Power analysis for RNA-Seq differential expression studies using generalized linear mixed effects models. BMC Bioinformatics 2020; 21:198. [PMID: 32429934 PMCID: PMC7236949 DOI: 10.1186/s12859-020-3541-7] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2019] [Accepted: 05/07/2020] [Indexed: 11/10/2022] Open
Abstract
Background Power analysis becomes an inevitable step in experimental design of current biomedical research. Complex designs allowing diverse correlation structures are commonly used in RNA-Seq experiments. However, the field currently lacks statistical methods to calculate sample size and estimate power for RNA-Seq differential expression studies using such designs. To fill the gap, simulation based methods have a great advantage by providing numerical solutions, since theoretical distributions of test statistics are typically unavailable for such designs. Results In this paper, we propose a novel simulation based procedure for power estimation of differential expression with the employment of generalized linear mixed effects models for correlated expression data. We also propose a new procedure for power estimation of differential expression with the use of a bivariate negative binomial distribution for paired designs. We compare the performance of both the likelihood ratio test and Wald test under a variety of simulation scenarios with the proposed procedures. The simulated distribution was used to estimate the null distribution of test statistics in order to achieve the desired false positive control and was compared to the asymptotic Chi-square distribution. In addition, we applied the procedure for paired designs to the TCGA breast cancer data set. Conclusions In summary, we provide a framework for power estimation of RNA-Seq differential expression under complex experimental designs. Simulation results demonstrate that both the proposed procedures properly control the false positive rate at the nominal level.
Collapse
Affiliation(s)
- Lianbo Yu
- Center for Biostatistics, Department of Biomedical Informatics, The Ohio State University, 1800 Cannon Dr., Columbus, 43210, OH, USA.
| | - Soledad Fernandez
- Center for Biostatistics, Department of Biomedical Informatics, The Ohio State University, 1800 Cannon Dr., Columbus, 43210, OH, USA
| | - Guy Brock
- Center for Biostatistics, Department of Biomedical Informatics, The Ohio State University, 1800 Cannon Dr., Columbus, 43210, OH, USA
| |
Collapse
|
86
|
Luo XL, Dalod M. The quest for faithful in vitro models of human dendritic cells types. Mol Immunol 2020; 123:40-59. [PMID: 32413788 DOI: 10.1016/j.molimm.2020.04.018] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2019] [Revised: 04/06/2020] [Accepted: 04/16/2020] [Indexed: 12/13/2022]
Abstract
Dendritic cells (DCs) are mononuclear phagocytes that are specialized in the induction and functional polarization of effector lymphocytes, thus orchestrating immune defenses against infections and cancer. The population of DC encompasses distinct cell types that vary in their efficacy for complementary functions and are thus likely involved in defending the body against different threats. Plasmacytoid DCs specialize in the production of high levels of the antiviral cytokines type I interferons. Type 1 conventional DCs (cDC1s) excel in the activation of cytotoxic CD8+ T cells (CTLs) which are critical for defense against cancer and infections by intracellular pathogens. Type 2 conventional DCs (cDC2s) prime helper CD4+ T cells for the production of type 2 cytokines underpinning immune defenses against worms or of IL-17 promoting control of infections by extracellular bacteria or fungi. Hence, clinically manipulating the development and functions of DC types could have a major impact for improving treatments against many diseases. However, the rarity and fragility of human DC types is impeding advancement towards this goal. To overcome this roadblock, major efforts are ongoing to generate in vitro large numbers of distinct human DC types. We review here the current state of this research field, emphasizing recent breakthrough and proposing future priorities. We also pinpoint the necessity to develop a consensus nomenclature and rigorous methodologies to ensure proper identification and characterization of human DC types. Finally, we elaborate on how faithful in vitro models of human DC types can accelerate our understanding of the biology of these cells and the engineering of next generation vaccines or immunotherapies against viral infections or cancer.
Collapse
Affiliation(s)
- Xin-Long Luo
- Aix Marseille Univ, CNRS, INSERM, CIML, Centre d'Immunologie de Marseille-Luminy, Turing Center for Living Systems, Marseille, France
| | - Marc Dalod
- Aix Marseille Univ, CNRS, INSERM, CIML, Centre d'Immunologie de Marseille-Luminy, Turing Center for Living Systems, Marseille, France.
| |
Collapse
|
87
|
Villar J, Segura E. Recent advances towards deciphering human dendritic cell development. Mol Immunol 2020; 122:109-115. [PMID: 32339957 DOI: 10.1016/j.molimm.2020.04.004] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2019] [Revised: 03/10/2020] [Accepted: 04/09/2020] [Indexed: 10/24/2022]
Abstract
Dendritic cell (DC) populations are the orchestrators of immune responses and arise from hematopoietic progenitors. Studies to unravel DC ontogeny have been conducted mainly in mice due to historical and practical reasons. However, understanding DC development in humans is a prerequisite for manipulating this process for therapeutic design. Here, we review the advantages and limitations of methods used to study human DC development in vitro and in vivo. In particular, we examine the in vitro culture systems that support the differentiation of all or some DC subpopulations. We also review recent discoveries regarding human DC precursors and factors that regulate their differentiation.
Collapse
Affiliation(s)
- Javiera Villar
- Institut Curie, PSL Research University, INSERM, U932, 26 rue d'Ulm, 75005 Paris, France
| | - Elodie Segura
- Institut Curie, PSL Research University, INSERM, U932, 26 rue d'Ulm, 75005 Paris, France.
| |
Collapse
|
88
|
Chapuy L, Sarfati M. Single-Cell Protein and RNA Expression Analysis of Mononuclear Phagocytes in Intestinal Mucosa and Mesenteric Lymph Nodes of Ulcerative Colitis and Crohn's Disease Patients. Cells 2020; 9:E813. [PMID: 32230977 PMCID: PMC7226791 DOI: 10.3390/cells9040813] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2020] [Revised: 03/19/2020] [Accepted: 03/24/2020] [Indexed: 12/21/2022] Open
Abstract
Inflammatory bowel diseases (IBDs), which include Crohn's disease (CD) and ulcerative colitis (UC), are driven by an abnormal immune response to commensal microbiota in genetically susceptible hosts. In addition to epithelial and stromal cells, innate and adaptive immune systems are both involved in IBD immunopathogenesis. Given the advances driven by single-cell technologies, we here reviewed the immune landscape and function of mononuclear phagocytes in inflamed non-lymphoid and lymphoid tissues of CD and UC patients. Immune cell profiling of IBD tissues using scRNA sequencing combined with multi-color cytometry analysis identifies unique clusters of monocyte-like cells, macrophages, and dendritic cells. These clusters reflect either distinct cell lineages (nature), or distinct or intermediate cell types with identical ontogeny, adapting their phenotype and function to the surrounding milieu (nurture and tissue imprinting). These advanced technologies will provide an unprecedented view of immune cell networks in health and disease, and thus may offer a personalized medicine approach to patients with IBD.
Collapse
Affiliation(s)
| | - Marika Sarfati
- Immunoregulation Laboratory, CRCHUM, Montreal, QC H2X 0A9, Canada;
| |
Collapse
|
89
|
Chapuy L, Bsat M, Rubio M, Harvey F, Motta V, Schwenter F, Wassef R, Richard C, Deslandres C, Nguyen BN, Soucy G, Hacohen N, Fritz J, Villani AC, Mehta H, Sarfati M. Transcriptomic Analysis and High-dimensional Phenotypic Mapping of Mononuclear Phagocytes in Mesenteric Lymph Nodes Reveal Differences Between Ulcerative Colitis and Crohn's Disease. J Crohns Colitis 2020; 14:393-405. [PMID: 31541232 PMCID: PMC7068244 DOI: 10.1093/ecco-jcc/jjz156] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
BACKGROUND AND AIMS Crohn's disease [CD] and ulcerative colitis [UC] are distinct forms of inflammatory bowel disease. Heterogeneity of HLA-DR+SIRPα + mononuclear phagocytes [MNPs], including macrophages [MΦ], monocyte-derived [Mono] cells, and dendritic cells [DCs], was reported in gut tissue but not yet investigated in mesenteric lymph nodes [MLNs] of IBD patients. We here compared the phenotype, function, and molecular profile of HLA-DR+SIRPα + MNPs in CD and UC MLNs. METHODS Cell distribution, morphology, immune function, and transcriptomic [bulk RNAseq] and high-dimensional protein expression profiles [CyTOF] of HLA-DR+SIRPα + MNPs were examined in MLNs of UC [n = 14], CD [n = 35], and non-IBD [n = 12] patients. RESULTS Elevated frequencies of CD14+CD64+CD163+ [Mono/MΦ-like] MNPs displaying monocyte/MΦ morphology and phagocytic function were a distinct feature of UC MLNs. In CD, the proportion of CD14-CD64-CD163- [DC-like] cells was augmented relative to Mono/MΦ-like cells; DC-like cells drove naïve T cell proliferation, Th1 polarisation, and Th17 TCM plasticity. Gene expression profile corroborated the nature of DC-like cells, best represented by BTLA, SERPINF, IGJ and, of Mono/MΦ-like cells, defined by CD163, MARCO, MAFB, CD300E, S100A9 expression. CyTOF analysis showed that CD123+ plasmacytoid cells predominated over conventional DCs in DC-like cells. Four CD163+ clusters were revealed in Mono/MΦ-like cells, two of which were enriched in MARCO-CD68dimHLA-DRdim monocyte-like cells and MARCOhiCD68hiHLA-DRhi Mɸ, whose proportion increased in UC relative to CD. CONCLUSIONS Defining the landscape of MNPs in MLNs provided evidence for expansion of CD163+ Mono/MΦ-like cells in UC only, highlighting a distinction between UC and CD, and thus the potential contribution of monocyte-like cells in driving colitis.
Collapse
Affiliation(s)
- Laurence Chapuy
- Immunoregulation Laboratory, Centre de Recherche du Centre Hospitalier de l’Université de Montréal [CRCHUM], Montréal, QC, Canada
| | - Marwa Bsat
- Immunoregulation Laboratory, Centre de Recherche du Centre Hospitalier de l’Université de Montréal [CRCHUM], Montréal, QC, Canada
| | - Manuel Rubio
- Immunoregulation Laboratory, Centre de Recherche du Centre Hospitalier de l’Université de Montréal [CRCHUM], Montréal, QC, Canada
| | - François Harvey
- Department of Biomedical Informatics, Centre de Recherche du Centre Hospitalier de l’Université de Montréal [CRCHUM], Montréal, QC, Canada
| | - Vinicius Motta
- McGill Goodman Research Center, McGill University, Montréal, QC, Canada
| | - Frank Schwenter
- Digestive Surgery Department, Centre Hospitalier de l’Université de Montréal [CHUM], Montréal, QC, Canada
| | - Ramses Wassef
- Digestive Surgery Department, Centre Hospitalier de l’Université de Montréal [CHUM], Montréal, QC, Canada
| | - Carole Richard
- Digestive Surgery Department, Centre Hospitalier de l’Université de Montréal [CHUM], Montréal, QC, Canada
| | - Colette Deslandres
- Division of Gastroenterology, Hepatology and Nutrition, Department of Pediatrics, CHU Sainte-Justine, Université de Montreal, QC, Canada
| | - Bich N Nguyen
- Pathology Department, Centre Hospitalier de l’Université de Montréal [CHUM], Montréal, QC, Canada
| | - Geneviève Soucy
- Pathology Department, Centre Hospitalier de l’Université de Montréal [CHUM], Montréal, QC, Canada
| | - Nir Hacohen
- Broad Institute of MIT and Harvard, Cambridge, MA USA
| | - Jorge Fritz
- Department of Microbiology and Immunology, McGill University, Montréal, Qc, Canada
| | - Alexandra-Chloé Villani
- Broad Institute of MIT and Harvard, Cambridge, MA USA
- Center for Immunology and Inflammatory Diseases, Massachusetts General Hospital, Boston, MA, USA
| | - Heena Mehta
- Immunoregulation Laboratory, Centre de Recherche du Centre Hospitalier de l’Université de Montréal [CRCHUM], Montréal, QC, Canada
| | - Marika Sarfati
- Immunoregulation Laboratory, Centre de Recherche du Centre Hospitalier de l’Université de Montréal [CRCHUM], Montréal, QC, Canada
| |
Collapse
|
90
|
Hinneburg H, Pedersen JL, Bokil NJ, Pralow A, Schirmeister F, Kawahara R, Rapp E, Saunders BM, Thaysen-Andersen M. High-resolution longitudinal N- and O-glycoprofiling of human monocyte-to-macrophage transition. Glycobiology 2020; 30:679-694. [DOI: 10.1093/glycob/cwaa020] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2019] [Revised: 02/18/2020] [Accepted: 03/02/2020] [Indexed: 12/12/2022] Open
Abstract
Abstract
Protein glycosylation impacts the development and function of innate immune cells. The glycophenotypes and the glycan remodelling associated with the maturation of macrophages from monocytic precursor populations remain incompletely described. Herein, label-free porous graphitised carbon–liquid chromatography–tandem mass spectrometry (PGC-LC-MS/MS) was employed to profile with high resolution the N- and O-glycome associated with human monocyte-to-macrophage transition. Primary blood-derived CD14+ monocytes were differentiated ex vivo in the absence of strong anti- and proinflammatory stimuli using a conventional 7-day granulocyte-macrophage colony-stimulating factor differentiation protocol with longitudinal sampling. Morphology and protein expression monitored by light microscopy and proteomics validated the maturation process. Glycomics demonstrated that monocytes and macrophages display similar N-glycome profiles, comprising predominantly paucimannosidic (Man1-3GlcNAc2Fuc0–1, 22.1–30.8%), oligomannosidic (Man5-9GlcNAc2, 29.8–35.7%) and α2,3/6-sialylated complex-type N-glycans with variable core fucosylation (27.6–39.1%). Glycopeptide analysis validated conjugation of these glycans to human proteins, while quantitative proteomics monitored the glycoenzyme expression levels during macrophage differentiation. Significant interperson glycome variations were observed suggesting a considerable physiology-dependent or heritable heterogeneity of CD14+ monocytes. Only few N-glycome changes correlated with the monocyte-to-macrophage transition across donors including decreased core fucosylation and reduced expression of mannose-terminating (paucimannosidic-/oligomannosidic-type) N-glycans in macrophages, while lectin flow cytometry indicated that more dramatic cell surface glycan remodelling occurs during maturation. The less heterogeneous core 1-rich O-glycome showed a minor decrease in core 2-type O-glycosylation but otherwise remained unchanged with macrophage maturation. This high-resolution glycome map underpinning normal monocyte-to-macrophage transition, the most detailed to date, aids our understanding of the molecular makeup pertaining to two vital innate immune cell types and forms an important reference for future glycoimmunological studies.
Collapse
Affiliation(s)
- Hannes Hinneburg
- Department of Molecular Sciences, Macquarie University, NSW 2109 Sydney, Australia
- Biomolecular Discovery Research Centre, Macquarie University, NSW 2109 Sydney, Australia
| | - Jessica L Pedersen
- School of Life Sciences, Faculty of Science, University of Technology, NSW 2007 Sydney, Australia
| | - Nilesh J Bokil
- School of Life Sciences, Faculty of Science, University of Technology, NSW 2007 Sydney, Australia
| | - Alexander Pralow
- Max Planck Institute for Dynamics of Complex Technical Systems (Bioprocess Engineering), 39106 Magdeburg, Germany
| | | | - Rebeca Kawahara
- Department of Molecular Sciences, Macquarie University, NSW 2109 Sydney, Australia
- Biomolecular Discovery Research Centre, Macquarie University, NSW 2109 Sydney, Australia
| | - Erdmann Rapp
- Max Planck Institute for Dynamics of Complex Technical Systems (Bioprocess Engineering), 39106 Magdeburg, Germany
- GlyXera GmbH, 39120 Magdeburg, Germany
| | - Bernadette M Saunders
- School of Life Sciences, Faculty of Science, University of Technology, NSW 2007 Sydney, Australia
| | - Morten Thaysen-Andersen
- Department of Molecular Sciences, Macquarie University, NSW 2109 Sydney, Australia
- Biomolecular Discovery Research Centre, Macquarie University, NSW 2109 Sydney, Australia
| |
Collapse
|
91
|
Lira-Junior R, Holmström SB, Clark R, Zwicker S, Majster M, Johannsen G, Axtelius B, Åkerman S, Svensson M, Klinge B, Boström EA. S100A12 Expression Is Modulated During Monocyte Differentiation and Reflects Periodontitis Severity. Front Immunol 2020; 11:86. [PMID: 32082330 PMCID: PMC7005221 DOI: 10.3389/fimmu.2020.00086] [Citation(s) in RCA: 37] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2019] [Accepted: 01/13/2020] [Indexed: 12/23/2022] Open
Abstract
S100A12 is a calcium-binding protein of the S100 subfamily of myeloid-related proteins that acts as an alarmin to induce a pro-inflammatory innate immune response. It has been linked to several chronic inflammatory diseases, however its role in the common oral immunopathology periodontitis is largely unknown. Previous in vitro monoculture experiments indicate that S100A12 production decreases during monocyte differentiation stages, while the regulation within tissue is poorly defined. This study evaluated S100A12 expression in monocyte subsets, during monocyte-to-macrophage differentiation and following polarization, both in monoculture and in a tissue context, utilizing a three-dimensional co-culture oral tissue model. Further, we explored the involvement of S100A12 in periodontitis by analyzing its expression in peripheral circulation and gingival tissue, as well as in saliva. We found that S100A12 expression was higher in classical than in non-classical monocytes. S100A12 expression and protein secretion declined significantly during monocyte-to-macrophage differentiation, while polarization of monocyte-derived macrophages had no effect on either. Peripheral monocytes from periodontitis patients had higher S100A12 expression than monocytes from controls, a difference particularly observed in the intermediate and non-classical monocyte subsets. Further, monocytes from periodontitis patients displayed an increased secretion of S100A12 compared with monocytes from controls. In oral tissue cultures, monocyte differentiation resulted in increased S100A12 secretion over time, which further increased after inflammatory stimuli. Likewise, S100A12 expression was higher in gingival tissue from periodontitis patients where monocyte-derived cells exhibited higher expression of S100A12 in comparison to non-periodontitis tissue. In line with our findings, patients with severe periodontitis had significantly higher levels of S100A12 in saliva compared to non-periodontitis patients, and the levels correlated to clinical periodontal parameters. Taken together, S100A12 is predominantly secreted by monocytes rather than by monocyte-derived cells. Moreover, S100A12 is increased in inflamed tissue cultures, potentially as a result of enhanced production by monocyte-derived cells. This study implicates the involvement of S100A12 in periodontitis pathogenesis, as evidenced by increased S100A12 expression in inflamed gingival tissue, which may be due to altered circulatory monocytes in periodontitis.
Collapse
Affiliation(s)
- Ronaldo Lira-Junior
- Division of Oral Diseases, Department of Dental Medicine, Karolinska Institutet, Huddinge, Sweden
| | - Sofia Björnfot Holmström
- Division of Oral Diseases, Department of Dental Medicine, Karolinska Institutet, Huddinge, Sweden
| | - Reuben Clark
- Division of Oral Diseases, Department of Dental Medicine, Karolinska Institutet, Huddinge, Sweden
| | - Stephanie Zwicker
- Division of Oral Diseases, Department of Dental Medicine, Karolinska Institutet, Huddinge, Sweden
| | - Mirjam Majster
- Division of Oral Diseases, Department of Dental Medicine, Karolinska Institutet, Huddinge, Sweden
| | - Gunnar Johannsen
- Division of Oral Diseases, Department of Dental Medicine, Karolinska Institutet, Huddinge, Sweden
| | - Björn Axtelius
- Department of Oral Diagnostics, Faculty of Odontology, Malmö University, Malmö, Sweden
| | - Sigvard Åkerman
- Department of Orofacial Pain and Jaw Function, Faculty of Odontology, Malmö University, Malmö, Sweden
| | - Mattias Svensson
- Department of Medicine, Center for Infectious Medicine, Karolinska Institutet, Huddinge, Sweden
| | - Björn Klinge
- Division of Oral Diseases, Department of Dental Medicine, Karolinska Institutet, Huddinge, Sweden.,Department of Periodontology, Faculty of Odontology, Malmö University, Malmö, Sweden
| | - Elisabeth A Boström
- Division of Oral Diseases, Department of Dental Medicine, Karolinska Institutet, Huddinge, Sweden
| |
Collapse
|
92
|
Factor VIII Fc Fusion Protein but not FVIII Drives Human Monocyte-Derived Dendritic Cell Activation via FcγRIIa. Hemasphere 2020; 4:e330. [PMID: 32072146 PMCID: PMC7000470 DOI: 10.1097/hs9.0000000000000330] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2019] [Accepted: 11/26/2019] [Indexed: 12/12/2022] Open
Abstract
This study compares the effect of recombinant Factor VIII Fc fusion protein (rFVIII-Fc) with recombinant FVIII (rFVIII) on monocyte-derived dendritic cells (moDC's). Cells treated with rFVIII-Fc showed morphological changes typical for cell activation, had a significant up-regulation of cell activation markers and produced higher levels of pro-inflammatory cytokines. Even after stimulation with Lipopolysaccharides, the addition of rFVIII-Fc led to increased expression of activation markers, indicating that rFVIII-Fc is capable of amplifying the maturation signal. On the contrary, cultivation of moDC's with rFVIII did not alter cell morphology or increase surface activation marker expression and pro-inflammatory cytokine production. The binding of the Fc domain to the activating Fcγ receptor IIa (FcγRIIa) can cause cell activation. Therefore, the effect of rFVIII-Fc on FcγRIIa was analyzed in detail. Cultivation of moDC's with rFVIII-Fc led to increased phosphorylation of FcγRIIa, which was not detected for rFVIII. Blocking FcγRIIa prior to the cultivation with rFVIII-Fc significantly reduced the activating effect of rFVIII-Fc, indicating that rFVIII-Fc-induced moDC activation was caused by FcγRIIa. Moreover, rFVIII-Fc bound to FCGR2A-transfected human embryonic kidney 293 cells. Taken together, our data present a new mechanism of moDC activation by rFVIII-Fc via FcγRIIa.
Collapse
|
93
|
Brombacher EC, Everts B. Shaping of Dendritic Cell Function by the Metabolic Micro-Environment. Front Endocrinol (Lausanne) 2020; 11:555. [PMID: 33013685 PMCID: PMC7493661 DOI: 10.3389/fendo.2020.00555] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/07/2020] [Accepted: 07/07/2020] [Indexed: 12/13/2022] Open
Abstract
Nutrients are required for growth and survival of all cells, but are also crucially involved in cell fate determination of many cell types, including immune cells. There is a growing appreciation that the metabolic micro-environment also plays a major role in shaping the functional properties of dendritic cells (DCs). Under pathological conditions nutrient availability can range from a very restricted supply, such as seen in a tumor micro-environment, to an overabundance of nutrients found in for example obese adipose tissue. In this review we will discuss what is currently known about the metabolic requirements for DC differentiation and immunogenicity and compare that to how function and fate of DCs under pathological conditions can be affected by alterations in environmental levels of carbohydrates, lipids and amino acids as well as by other metabolic cues, including availability of oxygen, redox homeostasis and lactate levels. Many of these insights have been generated using in vitro model systems, which have revealed highly diverse effects of different metabolic cues on DC function. However, they also stress the importance of shifting toward more physiologically relevant experimental settings to be able to fully delineate the role of the metabolic surroundings in its full complexity in shaping the functional properties of DCs in health and disease.
Collapse
|
94
|
Monaghan KL, Zheng W, Hu G, Wan ECK. Monocytes and Monocyte-Derived Antigen-Presenting Cells Have Distinct Gene Signatures in Experimental Model of Multiple Sclerosis. Front Immunol 2019; 10:2779. [PMID: 31849962 PMCID: PMC6889845 DOI: 10.3389/fimmu.2019.02779] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2019] [Accepted: 11/13/2019] [Indexed: 12/11/2022] Open
Abstract
Multiple sclerosis (MS) is a chronic inflammatory disease mediated by a complex interaction between the autoreactive lymphocytes and the effector myeloid cells within the central nervous system (CNS). In a murine model of MS, experimental autoimmune encephalomyelitis (EAE), Ly6Chi monocytes migrate into the CNS and further differentiate into antigen-presenting cells (APCs) during disease progression. Currently, there is no information about gene signatures that can distinguish between monocytes and the monocyte-derived APCs. We developed a surface marker-based strategy to distinguish between these two cell types during the stage of EAE when the clinical symptoms were most severe, and performed transcriptome analysis to compare their gene expression. We report here that the inflammatory CNS environment substantially alters gene expression of monocytes, compared to the monocyte differentiation process within CNS. Monocytes in the CNS express genes that encode proinflammatory cytokines and chemokines, and their expression is mostly maintained when the cells differentiate. Moreover, monocyte-derived APCs express surface markers associated with both dendritic cells and macrophages, and have a significant up-regulation of genes that are critical for antigen presentation. Furthermore, we found that Ccl17, Ccl22, and Ccr7 are expressed in monocyte-derived APCs but not the Ly6Chi monocytes. These findings may shed light on identifying molecular signals that control monocyte differentiation and functions during EAE.
Collapse
Affiliation(s)
- Kelly L. Monaghan
- Department of Microbiology, Immunology, and Cell Biology, West Virginia University, Morgantown, WV, United States
| | - Wen Zheng
- Department of Microbiology, Immunology, and Cell Biology, West Virginia University, Morgantown, WV, United States
| | - Gangqing Hu
- Department of Microbiology, Immunology, and Cell Biology, West Virginia University, Morgantown, WV, United States
- Bioinformatics Core, West Virginia University, Morgantown, WV, United States
| | - Edwin C. K. Wan
- Department of Microbiology, Immunology, and Cell Biology, West Virginia University, Morgantown, WV, United States
- Department of Neuroscience, West Virginia University, Morgantown, WV, United States
- Rockefeller Neuroscience Institute, West Virginia University, Morgantown, WV, United States
| |
Collapse
|
95
|
Carstensen LS, Lie-Andersen O, Obers A, Crowther MD, Svane IM, Hansen M. Long-Term Exposure to Inflammation Induces Differential Cytokine Patterns and Apoptosis in Dendritic Cells. Front Immunol 2019; 10:2702. [PMID: 31824496 PMCID: PMC6882286 DOI: 10.3389/fimmu.2019.02702] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2019] [Accepted: 11/04/2019] [Indexed: 12/21/2022] Open
Abstract
The activation of dendritic cells (DCs) has profound implications and governs the control of adaptive immunity. However, long-term activation might drive exhaustion of immune cells and negatively affect functionality. Here, long-term vs. short-term exposure to bacterial lipopolysaccharide and interferon (IFN)γ was evaluated on human monocyte-derived DCs. Long-term activated DC1s began to undergo apoptosis concomitant with a profound TAM-receptor and efferocytosis-dependent induction of interleukin (IL)-10. Whereas, levels of IL-12p70 and IL-10 were positively correlated upon short-term activation, an inverse association occured upon long-term activation and, while short-term activated CD1a+ DCs were main producers of IL-12p70, CD1a− DCs were the main fraction that underwent apoptosis and released IL-10 upon long-term activation. Moreover, pre-apoptotic long-term activated DCs were no longer able to activate alloreactive IFNγ-responsive T cells present in peripheral blood mononuclear cells from healthy volunteers. The IFNγ response was mediated by IL-12p70, as a strong reduction in IFNγ was observed following blockade with an IL-12p70 neutralizing antibody. Finally, multiplex analysis of DC supernatants revealed a particular pattern of proteins associated with apoptosis, cancer and chronic inflammation partly overlapping with gold standard DCs well-known for their inability to secrete IL-12p70. In conclusion, long-term activated DC1s significantly changed their profile toward a non-functional, tumor-promoting and anti-inflammatory phenotype.
Collapse
Affiliation(s)
- Laura Stentoft Carstensen
- National Center for Cancer Immune Therapy (CCIT-DK), Department of Oncology, Copenhagen University Hospital Herlev, Herlev, Denmark
| | - Olivia Lie-Andersen
- National Center for Cancer Immune Therapy (CCIT-DK), Department of Oncology, Copenhagen University Hospital Herlev, Herlev, Denmark.,Department of Bioengineering, Technical University of Denmark, Lyngby, Denmark.,Immunitrack ApS, Copenhagen, Denmark
| | - Andreas Obers
- National Center for Cancer Immune Therapy (CCIT-DK), Department of Oncology, Copenhagen University Hospital Herlev, Herlev, Denmark
| | - Michael Douglas Crowther
- National Center for Cancer Immune Therapy (CCIT-DK), Department of Oncology, Copenhagen University Hospital Herlev, Herlev, Denmark
| | - Inge Marie Svane
- National Center for Cancer Immune Therapy (CCIT-DK), Department of Oncology, Copenhagen University Hospital Herlev, Herlev, Denmark
| | - Morten Hansen
- National Center for Cancer Immune Therapy (CCIT-DK), Department of Oncology, Copenhagen University Hospital Herlev, Herlev, Denmark
| |
Collapse
|
96
|
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: 3.4] [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
|
97
|
Polyacrylic acid-coated iron oxide nanoparticles could be a useful tool for tracking inflammatory monocytes. Future Sci OA 2019; 5:FSO423. [PMID: 31827892 PMCID: PMC6900970 DOI: 10.2144/fsoa-2019-0066] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
Aim: To establish the effect of poly(acrylic acid)-coated iron oxide nanoparticles (PAC-IONs) and later exposure to a magnetic field on the differentiation of mononuclear phagocytes into macrophages. Methods: By flow cytometry, cell death was evaluated with DIOC6 and PI, Poly (ADP-ribose) Polymerases (PARP) fragmentation, H2AX phosphorylation and TUNEL assay. Cytokines by Cytokine bead array and the intracellular amount of iron by atomic absorption spectrometry. Results: PAC-IONs did not induce apoptosis, modify the cell membrane integrity or alter the mitochondrial membrane potential. They did not affect the cell morphology, the pattern of cytokine accumulation or the activating role of differentiation of mononuclear phagocytes into macrophages on the proliferation of autologous T cells. Conclusion: This evidence indicates that the PAC-IONs are safe and biocompatible. Moreover, the selectivity of the PAC-IONs for mononuclear phagocytes, as well as their increased uptake by non-classical monocytes, warrant future research with a view to their use as a contrast agent, a useful tool for in vivo tracking of tissue-infiltrating mononuclear phagocytes. In the search for materials that allow the study of inflammatory processes when biopsies are not feasible, magnetic nanoparticles have become an alternative tool for use in MRI. This article examined whether supermagnetic iron nanoparticles can affect the basic function of phagocytic cells, with a view to their use in clinical imaging applications.
Collapse
|
98
|
Günther P, Schultze JL. Mind the Map: Technology Shapes the Myeloid Cell Space. Front Immunol 2019; 10:2287. [PMID: 31636632 PMCID: PMC6787770 DOI: 10.3389/fimmu.2019.02287] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2019] [Accepted: 09/10/2019] [Indexed: 12/14/2022] Open
Abstract
The myeloid cell system shows very high plasticity, which is crucial to quickly adapt to changes during an immune response. From the beginning, this high plasticity has made cell type classification within the myeloid cell system difficult. Not surprising, naming schemes have been frequently changed. Recent advancements in multidimensional technologies, including mass cytometry and single-cell RNA sequencing, are challenging our current understanding of cell types, cell subsets, and functional states of cells. Despite the power of these technologies to create new reference maps for the myeloid cell system, it is essential to put these new results into context with previous knowledge that was established over decades. Here we report on earlier attempts of cell type classification in the myeloid cell system, discuss current approaches and their pros and cons, and propose future strategies for cell type classification within the myeloid cell system that can be easily extended to other cell types.
Collapse
Affiliation(s)
- Patrick Günther
- Genomics and Immunoregulation, Life and Medical Sciences Institute (LIMES), University of Bonn, Bonn, Germany.,Platform for Single Cell Genomics and Epigenomics, German Center for Neurodegenerative Diseases and University of Bonn, Bonn, Germany
| | - Joachim L Schultze
- Genomics and Immunoregulation, Life and Medical Sciences Institute (LIMES), University of Bonn, Bonn, Germany.,Platform for Single Cell Genomics and Epigenomics, German Center for Neurodegenerative Diseases and University of Bonn, Bonn, Germany
| |
Collapse
|
99
|
Rodriguez RM, Suarez-Alvarez B, Lavín JL, Ascensión AM, Gonzalez M, Lozano JJ, Raneros AB, Bulnes PD, Vidal-Castiñeira JR, Huidobro C, Martin-Martin C, Sanz AB, Ruiz-Ortega M, Puig-Kröger A, Corbí AL, Araúzo-Bravo MJ, Aransay AM, Lopez-Larrea C. Signal Integration and Transcriptional Regulation of the Inflammatory Response Mediated by the GM-/M-CSF Signaling Axis in Human Monocytes. Cell Rep 2019; 29:860-872.e5. [DOI: 10.1016/j.celrep.2019.09.035] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2019] [Revised: 06/27/2019] [Accepted: 09/11/2019] [Indexed: 12/15/2022] Open
|
100
|
Martin JC, Chang C, Boschetti G, Ungaro R, Giri M, Grout JA, Gettler K, Chuang LS, Nayar S, Greenstein AJ, Dubinsky M, Walker L, Leader A, Fine JS, Whitehurst CE, Mbow ML, Kugathasan S, Denson LA, Hyams JS, Friedman JR, Desai PT, Ko HM, Laface I, Akturk G, Schadt EE, Salmon H, Gnjatic S, Rahman AH, Merad M, Cho JH, Kenigsberg E. Single-Cell Analysis of Crohn's Disease Lesions Identifies a Pathogenic Cellular Module Associated with Resistance to Anti-TNF Therapy. Cell 2019; 178:1493-1508.e20. [PMID: 31474370 PMCID: PMC7060942 DOI: 10.1016/j.cell.2019.08.008] [Citation(s) in RCA: 492] [Impact Index Per Article: 98.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2018] [Revised: 05/06/2019] [Accepted: 08/05/2019] [Indexed: 10/26/2022]
Abstract
Clinical benefits of cytokine blockade in ileal Crohn's disease (iCD) are limited to a subset of patients. Here, we applied single-cell technologies to iCD lesions to address whether cellular heterogeneity contributes to treatment resistance. We found that a subset of patients expressed a unique cellular module in inflamed tissues that consisted of IgG plasma cells, inflammatory mononuclear phagocytes, activated T cells, and stromal cells, which we named the GIMATS module. Analysis of ligand-receptor interaction pairs identified a distinct network connectivity that likely drives the GIMATS module. Strikingly, the GIMATS module was also present in a subset of patients in four independent iCD cohorts (n = 441), and its presence at diagnosis correlated with failure to achieve durable corticosteroid-free remission upon anti-TNF therapy. These results emphasize the limitations of current diagnostic assays and the potential for single-cell mapping tools to identify novel biomarkers of treatment response and tailored therapeutic opportunities.
Collapse
Affiliation(s)
- Jerome C Martin
- Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; Department of Oncological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Christie Chang
- Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; Department of Oncological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Gilles Boschetti
- Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; Department of Oncological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Ryan Ungaro
- The Dr. Henry D. Janowitz Division of Gastroenterology, Icahn School of Medicine at Mount Sinai, New York City, NY 10029, USA
| | - Mamta Giri
- Charles Bronfman Institute for Personalized Medicine, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - John A Grout
- Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; Department of Oncological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Kyle Gettler
- Charles Bronfman Institute for Personalized Medicine, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Ling-Shiang Chuang
- Charles Bronfman Institute for Personalized Medicine, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Shikha Nayar
- Charles Bronfman Institute for Personalized Medicine, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Alexander J Greenstein
- Department of Colorectal Surgery, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Marla Dubinsky
- Department of Pediatrics, Susan and Leonard Feinstein IBD Clinical Center, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Laura Walker
- Human Immune Monitoring Center, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Andrew Leader
- Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; Department of Oncological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Jay S Fine
- Boehringer Ingelheim Pharmaceuticals, Immunology and Respiratory Diseases Research, Ridgefield, CT 06877, USA
| | - Charles E Whitehurst
- Boehringer Ingelheim Pharmaceuticals, Immunology and Respiratory Diseases Research, Ridgefield, CT 06877, USA
| | - M Lamine Mbow
- Boehringer Ingelheim Pharmaceuticals, Immunology and Respiratory Diseases Research, Ridgefield, CT 06877, USA
| | - Subra Kugathasan
- Division of Pediatric Gastroenterology, Emory University School of Medicine, Atlanta, GA, USA
| | - Lee A Denson
- Division of Pediatric Gastroenterology, Hepatology, and Nutrition, Cincinnati Children's Hospital Medical Center and the University of Cincinnati College of Medicine, Cincinnati, OH, USA
| | - Jeffrey S Hyams
- Division of Digestive Diseases, Hepatology, and Nutrition, Connecticut Children's Medical Center, Hartford, CT, USA
| | | | | | - Huaibin M Ko
- The Dr. Henry D. Janowitz Division of Gastroenterology, Icahn School of Medicine at Mount Sinai, New York City, NY 10029, USA; Department of Pathology, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Ilaria Laface
- Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; Human Immune Monitoring Center, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Guray Akturk
- Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; Human Immune Monitoring Center, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Eric E Schadt
- Department of Genetics and Genomics Sciences, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; Icahn Institute for Genomics and Multiscale Biology, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Helene Salmon
- Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; Department of Oncological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Sacha Gnjatic
- Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; Human Immune Monitoring Center, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; Department of Medicine, Division of Hematology Oncology, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Adeeb H Rahman
- Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; Human Immune Monitoring Center, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; Department of Genetics and Genomics Sciences, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Miriam Merad
- Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; Department of Oncological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; Human Immune Monitoring Center, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA.
| | - Judy H Cho
- Charles Bronfman Institute for Personalized Medicine, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA.
| | - Ephraim Kenigsberg
- Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; Department of Genetics and Genomics Sciences, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; Icahn Institute for Genomics and Multiscale Biology, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA.
| |
Collapse
|