51
|
Iwanowycz S, Ngoi S, Li Y, Hill M, Koivisto C, Parrish M, Guo B, Li Z, Liu B. Type-2 dendritic cells mediate control of cytotoxic T cell-resistant tumors. JCI Insight 2021; 6:e145885. [PMID: 34283809 PMCID: PMC8492342 DOI: 10.1172/jci.insight.145885] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2020] [Accepted: 07/15/2021] [Indexed: 11/17/2022] Open
Abstract
Type 2 DCs (DC2s) comprise the majority of conventional DCs within most tumors; however, little is known about their ability to initiate and sustain antitumor immunity, as most studies have focused on antigen cross-presenting DC1s. Here, we report that DC2 infiltration identified by analysis of multiple human cancer data sets showed a significant correlation with survival across multiple human cancers, with the benefit being seen in tumors resistant to cytotoxic T cell control. Characterization of DC subtype infiltration into an immunotherapy-resistant model of breast cancer revealed that impairment of DC1s through 2 unique models resulted in enhanced DC2 functionality and improved tumor control. BATF3 deficiency depleted intratumoral DC1s, which led to increased DC2 lymph node migration and CD4+ T cell activation. Enhancing DC2 stimulatory potential by genetic deletion of Hsp90b1 (encoding molecular chaperon GP96) led to a similar enhancement of T cell immunity and improved survival in a spontaneous breast cancer model. These data highlight the therapeutic and prognostic potential of DC2s within checkpoint blockade–resistant tumors.
Collapse
Affiliation(s)
- Stephen Iwanowycz
- Department of Microbiology and Immunology, Medical University of South Carolina, Charleston, United States of America
| | - Soo Ngoi
- Department of Internal Medicine, The Ohio State University College of Medicine, Columbus, United States of America
| | - Yingqi Li
- Department of Microbiology and Immunology, Medical University of South Carolina, Charleston, United States of America
| | - Megan Hill
- Department of Internal Medicine, The Ohio State University College of Medicine, Columbus, United States of America
| | - Christopher Koivisto
- Department of Biochemistry and Molecular Biology, Medical University of South Carolina, Charleston, United States of America
| | - Melodie Parrish
- Department of Biochemistry and Molecular Biology, Medical University of South Carolina, Charleston, United States of America
| | - Beichu Guo
- Department of Microbiology and Immunology, Medical University of South Carolina, Charleston, United States of America
| | - Zihai Li
- The Pelotonia Institute for Immuno-Oncology, The Ohio State University College of Medicine, Columbus, United States of America
| | - Bei Liu
- Department of Internal Medicine, The Ohio State University College of Medicine, Columbus, United States of America
| |
Collapse
|
52
|
Tian L, Tomei S, Schreuder J, Weber TS, Amann-Zalcenstein D, Lin DS, Tran J, Audiger C, Chu M, Jarratt A, Willson T, Hilton A, Pang ES, Patton T, Kelly M, Su S, Gouil Q, Diakumis P, Bahlo M, Sargeant T, Kats LM, Hodgkin PD, O'Keeffe M, Ng AP, Ritchie ME, Naik SH. Clonal multi-omics reveals Bcor as a negative regulator of emergency dendritic cell development. Immunity 2021; 54:1338-1351.e9. [PMID: 33862015 DOI: 10.1016/j.immuni.2021.03.012] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2020] [Revised: 02/05/2021] [Accepted: 03/17/2021] [Indexed: 12/13/2022]
Abstract
Despite advances in single-cell multi-omics, a single stem or progenitor cell can only be tested once. We developed clonal multi-omics, in which daughters of a clone act as surrogates of the founder, thereby allowing multiple independent assays per clone. With SIS-seq, clonal siblings in parallel "sister" assays are examined either for gene expression by RNA sequencing (RNA-seq) or for fate in culture. We identified, and then validated using CRISPR, genes that controlled fate bias for different dendritic cell (DC) subtypes. This included Bcor as a suppressor of plasmacytoid DC (pDC) and conventional DC type 2 (cDC2) numbers during Flt3 ligand-mediated emergency DC development. We then developed SIS-skew to examine development of wild-type and Bcor-deficient siblings of the same clone in parallel. We found Bcor restricted clonal expansion, especially for cDC2s, and suppressed clonal fate potential, especially for pDCs. Therefore, SIS-seq and SIS-skew can reveal the molecular and cellular mechanisms governing clonal fate.
Collapse
Affiliation(s)
- Luyi Tian
- Immunology Division, The Walter and Eliza Hall Institute of Medical Research, Parkville, VIC 3052, Australia; Epigenetics and Development Division, The Walter and Eliza Hall Institute of Medical Research, Parkville, VIC 3052, Australia; Department of Medical Biology, University of Melbourne, Parkville, VIC 3052, Australia
| | - Sara Tomei
- Immunology Division, The Walter and Eliza Hall Institute of Medical Research, Parkville, VIC 3052, Australia; Department of Medical Biology, University of Melbourne, Parkville, VIC 3052, Australia
| | - Jaring Schreuder
- Immunology Division, The Walter and Eliza Hall Institute of Medical Research, Parkville, VIC 3052, Australia
| | - Tom S Weber
- Immunology Division, The Walter and Eliza Hall Institute of Medical Research, Parkville, VIC 3052, Australia; Department of Medical Biology, University of Melbourne, Parkville, VIC 3052, Australia
| | - Daniela Amann-Zalcenstein
- Department of Medical Biology, University of Melbourne, Parkville, VIC 3052, Australia; Single Cell Open Research Endeavour (SCORE), The Walter and Eliza Hall Institute of Medical Research, Parkville, VIC 3052, Australia
| | - Dawn S Lin
- Immunology Division, The Walter and Eliza Hall Institute of Medical Research, Parkville, VIC 3052, Australia; Department of Medical Biology, University of Melbourne, Parkville, VIC 3052, Australia
| | - Jessica Tran
- Immunology Division, The Walter and Eliza Hall Institute of Medical Research, Parkville, VIC 3052, Australia
| | - Cindy Audiger
- Immunology Division, The Walter and Eliza Hall Institute of Medical Research, Parkville, VIC 3052, Australia; Department of Medical Biology, University of Melbourne, Parkville, VIC 3052, Australia
| | - Mathew Chu
- Immunology Division, The Walter and Eliza Hall Institute of Medical Research, Parkville, VIC 3052, Australia
| | - Andrew Jarratt
- Department of Medical Biology, University of Melbourne, Parkville, VIC 3052, Australia; Blood Cells and Blood Cancer Division, The Walter and Eliza Hall Institute of Medical Research, Parkville, VIC 3052, Australia
| | - Tracy Willson
- Department of Medical Biology, University of Melbourne, Parkville, VIC 3052, Australia; Blood Cells and Blood Cancer Division, The Walter and Eliza Hall Institute of Medical Research, Parkville, VIC 3052, Australia
| | - Adrienne Hilton
- Department of Medical Biology, University of Melbourne, Parkville, VIC 3052, Australia; Blood Cells and Blood Cancer Division, The Walter and Eliza Hall Institute of Medical Research, Parkville, VIC 3052, Australia
| | - Ee Shan Pang
- Department of Biochemistry and Molecular Biology, Biomedicine Discovery Institute, Monash University, Clayton, VIC 3800, Australia
| | - Timothy Patton
- Department of Biochemistry and Molecular Biology, Biomedicine Discovery Institute, Monash University, Clayton, VIC 3800, Australia
| | - Madison Kelly
- The Peter MacCallum Cancer Centre, Melbourne, VIC 3000, Australia; The Sir Peter MacCallum Department of Oncology, University of Melbourne, Parkville, VIC 3052, Australia
| | - Shian Su
- Epigenetics and Development Division, The Walter and Eliza Hall Institute of Medical Research, Parkville, VIC 3052, Australia; Department of Medical Biology, University of Melbourne, Parkville, VIC 3052, Australia
| | - Quentin Gouil
- Epigenetics and Development Division, The Walter and Eliza Hall Institute of Medical Research, Parkville, VIC 3052, Australia; Department of Medical Biology, University of Melbourne, Parkville, VIC 3052, Australia
| | - Peter Diakumis
- Department of Medical Biology, University of Melbourne, Parkville, VIC 3052, Australia; Population Health and Immunity Division, The Walter and Eliza Hall Institute of Medical Research, Parkville, VIC 3052, Australia
| | - Melanie Bahlo
- Department of Medical Biology, University of Melbourne, Parkville, VIC 3052, Australia; Population Health and Immunity Division, The Walter and Eliza Hall Institute of Medical Research, Parkville, VIC 3052, Australia
| | - Toby Sargeant
- Department of Medical Biology, University of Melbourne, Parkville, VIC 3052, Australia; Blood Cells and Blood Cancer Division, The Walter and Eliza Hall Institute of Medical Research, Parkville, VIC 3052, Australia
| | - Lev M Kats
- The Peter MacCallum Cancer Centre, Melbourne, VIC 3000, Australia; The Sir Peter MacCallum Department of Oncology, University of Melbourne, Parkville, VIC 3052, Australia
| | - Philip D Hodgkin
- Immunology Division, The Walter and Eliza Hall Institute of Medical Research, Parkville, VIC 3052, Australia; Department of Medical Biology, University of Melbourne, Parkville, VIC 3052, Australia
| | - Meredith O'Keeffe
- Department of Biochemistry and Molecular Biology, Biomedicine Discovery Institute, Monash University, Clayton, VIC 3800, Australia
| | - Ashley P Ng
- Department of Medical Biology, University of Melbourne, Parkville, VIC 3052, Australia; Blood Cells and Blood Cancer Division, The Walter and Eliza Hall Institute of Medical Research, Parkville, VIC 3052, Australia
| | - Matthew E Ritchie
- Epigenetics and Development Division, The Walter and Eliza Hall Institute of Medical Research, Parkville, VIC 3052, Australia; Department of Medical Biology, University of Melbourne, Parkville, VIC 3052, Australia
| | - Shalin H Naik
- Immunology Division, The Walter and Eliza Hall Institute of Medical Research, Parkville, VIC 3052, Australia; Department of Medical Biology, University of Melbourne, Parkville, VIC 3052, Australia; Single Cell Open Research Endeavour (SCORE), The Walter and Eliza Hall Institute of Medical Research, Parkville, VIC 3052, Australia.
| |
Collapse
|
53
|
Macrophages and Stem Cells-Two to Tango for Tissue Repair? Biomolecules 2021; 11:biom11050697. [PMID: 34066618 PMCID: PMC8148606 DOI: 10.3390/biom11050697] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2021] [Revised: 04/26/2021] [Accepted: 05/04/2021] [Indexed: 12/25/2022] Open
Abstract
Macrophages (MCs) are present in all tissues, not only supporting homeostasis, but also playing an important role in organogenesis, post-injury regeneration, and diseases. They are a heterogeneous cell population due to their origin, tissue specificity, and polarization in response to aggression factors, depending on environmental cues. Thus, as pro-inflammatory M1 phagocytic MCs, they contribute to tissue damage and even fibrosis, but the anti-inflammatory M2 phenotype participates in repairing processes and wound healing through a molecular interplay with most cells in adult stem cell niches. In this review, we emphasize MC phenotypic heterogeneity in health and disease, highlighting their systemic and systematic contribution to tissue homeostasis and repair. Unraveling the intervention of both resident and migrated MCs on the behavior of stem cells and the regulation of the stem cell niche is crucial for opening new perspectives for novel therapeutic strategies in different diseases.
Collapse
|
54
|
Cueto FJ, Del Fresno C, Brandi P, Combes AJ, Hernández-García E, Sánchez-Paulete AR, Enamorado M, Bromley CP, Gomez MJ, Conde-Garrosa R, Mañes S, Zelenay S, Melero I, Iborra S, Krummel MF, Sancho D. DNGR-1 limits Flt3L-mediated antitumor immunity by restraining tumor-infiltrating type I conventional dendritic cells. J Immunother Cancer 2021; 9:e002054. [PMID: 33980589 PMCID: PMC8118081 DOI: 10.1136/jitc-2020-002054] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/23/2021] [Indexed: 11/08/2022] Open
Abstract
BACKGROUND Conventional type 1 dendritic cells (cDC1s) are central to antitumor immunity and their presence in the tumor microenvironment associates with improved outcomes in patients with cancer. DNGR-1 (CLEC9A) is a dead cell-sensing receptor highly restricted to cDC1s. DNGR-1 has been involved in both cross-presentation of dead cell-associated antigens and processes of disease tolerance, but its role in antitumor immunity has not been clarified yet. METHODS B16 and MC38 tumor cell lines were inoculated subcutaneously into wild-type (WT) and DNGR-1-deficient mice. To overexpress Flt3L systemically, we performed gene therapy through the hydrodynamic injection of an Flt3L-encoding plasmid. To characterize the immune response, we performed flow cytometry and RNA-Seq of tumor-infiltrating cDC1s. RESULTS Here, we found that cross-presentation of tumor antigens in the steady state was DNGR-1-independent. However, on Flt3L systemic overexpression, tumor growth was delayed in DNGR-1-deficient mice compared with WT mice. Of note, this protection was recapitulated by anti-DNGR-1-blocking antibodies in mice following Flt3L gene therapy. This improved antitumor immunity was associated with Batf3-dependent enhanced accumulation of CD8+ T cells and cDC1s within tumors. Mechanistically, the deficiency in DNGR-1 boosted an Flt3L-induced specific inflammatory gene signature in cDC1s, including Ccl5 expression. Indeed, the increased infiltration of cDC1s within tumors and their protective effect rely on CCL5/CCR5 chemoattraction. Moreover, FLT3LG and CCL5 or CCR5 gene expression signatures correlate with an enhanced cDC1 signature and a favorable overall survival in patients with cancer. Notably, cyclophosphamide elevated serum Flt3L levels and, in combination with the absence of DNGR-1, synergized against tumor growth. CONCLUSION DNGR-1 limits the accumulation of tumor-infiltrating cDC1s promoted by Flt3L. Thus, DNGR-1 blockade may improve antitumor immunity in tumor therapy settings associated to high Flt3L expression.
Collapse
MESH Headings
- Animals
- Basic-Leucine Zipper Transcription Factors/genetics
- Basic-Leucine Zipper Transcription Factors/metabolism
- CD8-Positive T-Lymphocytes/immunology
- CD8-Positive T-Lymphocytes/metabolism
- Cell Line, Tumor
- Chemokine CCL5/genetics
- Chemokine CCL5/metabolism
- Coculture Techniques
- Colonic Neoplasms/genetics
- Colonic Neoplasms/immunology
- Colonic Neoplasms/metabolism
- Colonic Neoplasms/therapy
- Dendritic Cells/immunology
- Dendritic Cells/metabolism
- Gene Expression Regulation, Neoplastic
- Genetic Therapy
- Lectins, C-Type/genetics
- Lectins, C-Type/metabolism
- Lymphocytes, Tumor-Infiltrating/immunology
- Lymphocytes, Tumor-Infiltrating/metabolism
- Melanoma, Experimental/genetics
- Melanoma, Experimental/immunology
- Melanoma, Experimental/metabolism
- Melanoma, Experimental/therapy
- Membrane Proteins/genetics
- Membrane Proteins/metabolism
- Mice, Inbred C57BL
- Mice, Knockout
- Phenotype
- Receptors, CCR5/genetics
- Receptors, CCR5/metabolism
- Receptors, Immunologic/genetics
- Receptors, Immunologic/metabolism
- Repressor Proteins/genetics
- Repressor Proteins/metabolism
- Signal Transduction
- Skin Neoplasms/genetics
- Skin Neoplasms/immunology
- Skin Neoplasms/metabolism
- Skin Neoplasms/therapy
- Tumor Burden
- Tumor Escape
- Tumor Microenvironment
- Mice
Collapse
Affiliation(s)
- Francisco J Cueto
- Centro Nacional de Investigaciones Cardiovasculares Carlos III (CNIC), Madrid, Spain
| | - Carlos Del Fresno
- Centro Nacional de Investigaciones Cardiovasculares Carlos III (CNIC), Madrid, Spain
- Hospital la Paz Institute for Health Research (IdiPAZ), Madrid, Spain
| | - Paola Brandi
- Centro Nacional de Investigaciones Cardiovasculares Carlos III (CNIC), Madrid, Spain
| | - Alexis J Combes
- Department of Pathology, University of California, San Francisco, California, USA
- ImmunoX Initiative, University of California, San Francisco, California, USA
- UCSF CoLabs, University of California, San Francisco, California, USA
| | - Elena Hernández-García
- Department of Immunology, Ophthalmology and ENT, School of Medicine, Universidad Complutense de Madrid, Madrid, Spain
| | - Alfonso R Sánchez-Paulete
- Division of Immunology and Immunotherapy, Center for Applied Medical Research, University of Navarra, Pamplona, Spain
| | - Michel Enamorado
- Centro Nacional de Investigaciones Cardiovasculares Carlos III (CNIC), Madrid, Spain
- Metaorganism Immunity Section, Laboratory of Host Immunity and Microbiome, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, USA
| | - Christian P Bromley
- Cancer Inflammation and Immunity Group, CRUK Manchester Institute, The University of Manchester, Manchester, UK
| | - Manuel J Gomez
- Centro Nacional de Investigaciones Cardiovasculares Carlos III (CNIC), Madrid, Spain
| | - Ruth Conde-Garrosa
- Centro Nacional de Investigaciones Cardiovasculares Carlos III (CNIC), Madrid, Spain
| | - Santos Mañes
- Department of Immunology and Oncology, Centro Nacional de Biotecnología/CSIC, Darwin, Madrid, Spain
| | - Santiago Zelenay
- Cancer Inflammation and Immunity Group, CRUK Manchester Institute, The University of Manchester, Manchester, UK
| | - Ignacio Melero
- Division of Immunology and Immunotherapy, Center for Applied Medical Research, University of Navarra, Pamplona, Spain
- Instituto de Investigación Sanitaria de Navarra, Pamplona, Spain
- University Clinic, University of Navarra, Pamplona, Spain
- Centro de Investigación Biomédica en Red Cáncer (CIBERONC), Madrid, Spain
| | - Salvador Iborra
- Department of Immunology, Ophthalmology and ENT, School of Medicine, Universidad Complutense de Madrid, Madrid, Spain
| | - Matthew F Krummel
- Department of Pathology, University of California, San Francisco, California, USA
| | - David Sancho
- Centro Nacional de Investigaciones Cardiovasculares Carlos III (CNIC), Madrid, Spain
| |
Collapse
|
55
|
Teo YJ, Ng SL, Mak KW, Setiagani YA, Chen Q, Nair SK, Sheng J, Ruedl C. Renal CD169 ++ resident macrophages are crucial for protection against acute systemic candidiasis. Life Sci Alliance 2021; 4:e202000890. [PMID: 33608410 PMCID: PMC7918719 DOI: 10.26508/lsa.202000890] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2020] [Revised: 01/07/2021] [Accepted: 01/22/2021] [Indexed: 12/17/2022] Open
Abstract
Disseminated candidiasis remains as the most common hospital-acquired bloodstream fungal infection with up to 40% mortality rate despite the advancement of medical and hygienic practices. While it is well established that this infection heavily relies on the innate immune response for host survival, much less is known for the protective role elicited by the tissue-resident macrophage (TRM) subsets in the kidney, the prime organ for Candida persistence. Here, we describe a unique CD169++ TRM subset that controls Candida growth and inflammation during acute systemic candidiasis. Their absence causes severe fungal-mediated renal pathology. CD169++ TRMs, without being actively involved in direct fungal clearance, increase host resistance by promoting IFN-γ release and neutrophil ROS activity.
Collapse
Affiliation(s)
- Yi Juan Teo
- School of Biological Sciences, Nanyang Technological University, Singapore, Singapore
| | - See Liang Ng
- School of Biological Sciences, Nanyang Technological University, Singapore, Singapore
| | - Keng Wai Mak
- School of Biological Sciences, Nanyang Technological University, Singapore, Singapore
| | | | - Qi Chen
- School of Biological Sciences, Nanyang Technological University, Singapore, Singapore
| | - Sajith Kumar Nair
- School of Biological Sciences, Nanyang Technological University, Singapore, Singapore
| | - Jianpeng Sheng
- Department of Hepatobiliary and Pancreatic Surgery, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Christiane Ruedl
- School of Biological Sciences, Nanyang Technological University, Singapore, Singapore
| |
Collapse
|
56
|
Coillard A, Segura E. Antigen presentation by mouse monocyte-derived cells: Re-evaluating the concept of monocyte-derived dendritic cells. Mol Immunol 2021; 135:165-169. [PMID: 33901761 DOI: 10.1016/j.molimm.2021.04.012] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2020] [Revised: 03/22/2021] [Accepted: 04/12/2021] [Indexed: 12/20/2022]
Abstract
Antigen presentation is a key feature of classical dendritic cells (cDCs). Numerous studies have also reported in mouse that, upon inflammation, monocytes enter tissues and differentiate into monocyte-derived DCs (mo-DC) that have the ability to present antigens to T cells. However, a population of inflammatory cDCs sharing phenotypic features with mo-DC has been recently described, challenging the existence of in vivo-generated mo-DC. Here we review studies describing mouse mo-DC in the light of these findings, and evaluate the in vivo evidence for monocyte-derived antigen-presenting cells. We examine the strategies used to demonstrate the monocytic origin of these cells. Finally, we propose that mo-DC play a complementary role to cDCs, by presenting antigens to effector T cells locally in tissues.
Collapse
Affiliation(s)
- Alice Coillard
- Institut Curie, PSL Research University, INSERM, U932, 26 Rue d'Ulm, 75005, Paris, France; Université Paris Descartes, Paris, France
| | - Elodie Segura
- Institut Curie, PSL Research University, INSERM, U932, 26 Rue d'Ulm, 75005, Paris, France.
| |
Collapse
|
57
|
Cueto FJ, Sancho D. The Flt3L/Flt3 Axis in Dendritic Cell Biology and Cancer Immunotherapy. Cancers (Basel) 2021; 13:1525. [PMID: 33810248 PMCID: PMC8037622 DOI: 10.3390/cancers13071525] [Citation(s) in RCA: 58] [Impact Index Per Article: 19.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2021] [Revised: 03/21/2021] [Accepted: 03/23/2021] [Indexed: 12/19/2022] Open
Abstract
Dendritic cells (DCs) prime anti-tumor T cell responses in tumor-draining lymph nodes and can restimulate T effector responses in the tumor site. Thus, in addition to unleashing T cell effector activity, current immunotherapies should be directed to boost DC function. Herein, we review the potential function of Flt3L as a tool for cancer immunotherapy. Flt3L is a growth factor that acts in Flt3-expressing multipotent progenitors and common lymphoid progenitors. Despite the broad expression of Flt3 in the hematopoietic progenitors, the main effect of the Flt3/Flt3L axis, revealed by the characterization of mice deficient in these genes, is the generation of conventional DCs (cDCs) and plasmacytoid DCs (pDCs). However, Flt3 signaling through PI3K and mTOR may also affect the function of mature DCs. We recapitulate the use of Flt3L in preclinical studies either as a single agent or in combination with other cancer therapies. We also analyze the use of Flt3L in clinical trials. The strong correlation between type 1 cDC (cDC1) infiltration of human cancers with overall survival in many cancer types suggests the potential use of Flt3L to boost expansion of this DC subset. However, this may need the combination of Flt3L with other immunomodulatory agents to boost cancer immunotherapy.
Collapse
Affiliation(s)
- Francisco J. Cueto
- Centro Nacional de Investigaciones Cardiovasculares (CNIC), 28029 Madrid, Spain
| | - David Sancho
- Centro Nacional de Investigaciones Cardiovasculares (CNIC), 28029 Madrid, Spain
| |
Collapse
|
58
|
Anders HJ, Wilkens L, Schraml B, Marschner J. One concept does not fit all: the immune system in different forms of acute kidney injury. Nephrol Dial Transplant 2021; 36:29-38. [PMID: 32337558 DOI: 10.1093/ndt/gfaa056] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2019] [Indexed: 02/06/2023] Open
Abstract
Renal and immune systems maintain body homoeostasis during physiological fluctuations and following tissue injury. The immune system plays a central role during acute kidney injury (AKI), adapting evolutional systems programmed for host defence and minimizing unnecessary collateral damage. Indeed, depending upon the disease context, the impact of the immune system upon the manifestations and consequences of AKI can be quite different. Here we provide an overview of the known and unknown involvement of the immune system within the wide range of different forms of AKI, to oppose oversimplification and to endorse deeper insights into the pathogenesis of the different diseases causing kidney injury. This approach may help to overcome some of the current hurdles in translational AKI research and the development of specific treatments for the different diseases, all presenting with an acute increase in serum creatinine or decline in urinary output. One concept does not fit all.
Collapse
Affiliation(s)
- Hans-Joachim Anders
- Department of Medicine IV, Renal Division, University Hospital of the Ludwig Maximilians University, Munich, Germany
| | - Louise Wilkens
- Department of Medicine IV, Renal Division, University Hospital of the Ludwig Maximilians University, Munich, Germany
| | - Barbara Schraml
- Walter-Brendel-Centre of Experimental Medicine, University Hospital, LMU Munich, Planegg-Martinsried, Germany.,Faculty of Medicine, Institute for Cardiovascular Physiology and Pathophysiology, Biomedical Center, LMU Munich, Planegg-Martinsried, Germany
| | - Julian Marschner
- Department of Medicine IV, Renal Division, University Hospital of the Ludwig Maximilians University, Munich, Germany
| |
Collapse
|
59
|
van Uden D, Koudstaal T, van Hulst JAC, Bergen IM, Gootjes C, Morrell NW, van Loo G, von der Thüsen JH, van den Bosch TPP, Ghigna MR, Perros F, Montani D, Kool M, Boomars KA, Hendriks RW. Central Role of Dendritic Cells in Pulmonary Arterial Hypertension in Human and Mice. Int J Mol Sci 2021; 22:ijms22041756. [PMID: 33578743 PMCID: PMC7916474 DOI: 10.3390/ijms22041756] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2021] [Revised: 02/05/2021] [Accepted: 02/06/2021] [Indexed: 12/12/2022] Open
Abstract
The pathogenesis of idiopathic pulmonary arterial hypertension (IPAH) is not fully understood, but evidence is accumulating that immune dysfunction plays a significant role. We previously reported that 31-week-old Tnfaip3DNGR1-KO mice develop pulmonary hypertension (PH) symptoms. These mice harbor a targeted deletion of the TNFα-induced protein-3 (Tnfaip3) gene, encoding the NF-κB regulatory protein A20, specifically in type I conventional dendritic cells (cDC1s). Here, we studied the involvement of dendritic cells (DCs) in PH in more detail. We found various immune cells, including DCs, in the hearts of Tnfaip3DNGR1-KO mice, particularly in the right ventricle (RV). Secondly, in young Tnfaip3DNGR1-KO mice, innate immune activation through airway exposure to toll-like receptor ligands essentially did not result in elevated RV pressures, although we did observe significant RV hypertrophy. Thirdly, PH symptoms in Tnfaip3DNGR1-KO mice were not enhanced by concomitant mutation of bone morphogenetic protein receptor type 2 (Bmpr2), which is the most affected gene in PAH patients. Finally, in human IPAH lung tissue we found co-localization of DCs and CD8+ T cells, representing the main cell type activated by cDC1s. Taken together, these findings support a unique role of cDC1s in PAH pathogenesis, independent of general immune activation or a mutation in the Bmpr2 gene.
Collapse
Affiliation(s)
- Denise van Uden
- Department of Pulmonary Medicine, Erasmus MC, University Medical Center Rotterdam, 3015 GD Rotterdam, The Netherlands; (D.v.U.); (T.K.); (J.A.C.v.H.); (I.M.B.); (C.G.); (M.K.)
| | - Thomas Koudstaal
- Department of Pulmonary Medicine, Erasmus MC, University Medical Center Rotterdam, 3015 GD Rotterdam, The Netherlands; (D.v.U.); (T.K.); (J.A.C.v.H.); (I.M.B.); (C.G.); (M.K.)
| | - Jennifer A. C. van Hulst
- Department of Pulmonary Medicine, Erasmus MC, University Medical Center Rotterdam, 3015 GD Rotterdam, The Netherlands; (D.v.U.); (T.K.); (J.A.C.v.H.); (I.M.B.); (C.G.); (M.K.)
| | - Ingrid M. Bergen
- Department of Pulmonary Medicine, Erasmus MC, University Medical Center Rotterdam, 3015 GD Rotterdam, The Netherlands; (D.v.U.); (T.K.); (J.A.C.v.H.); (I.M.B.); (C.G.); (M.K.)
| | - Chelsea Gootjes
- Department of Pulmonary Medicine, Erasmus MC, University Medical Center Rotterdam, 3015 GD Rotterdam, The Netherlands; (D.v.U.); (T.K.); (J.A.C.v.H.); (I.M.B.); (C.G.); (M.K.)
| | - Nicholas W. Morrell
- Department of Medicine, University of Cambridge & NIHR BioResource for Translational Research & Addenbrooke’s Hospital NHS Foundation Trust & Royal Papworth Hospital NHS Foundation Trust, Cambridge CB2 0QQ, UK;
| | - Geert van Loo
- VIB Center for Inflammation Research, 9052 Ghent, Belgium;
- Department of Biomedical Molecular Biology, Ghent University, 9052 Ghent, Belgium
| | - Jan H. von der Thüsen
- Department of Pathology, Erasmus MC, University Medical Center Rotterdam, 3015 GE Rotterdam, The Netherlands; (J.H.v.d.T.); (T.P.P.v.d.B.)
| | - Thierry P. P. van den Bosch
- Department of Pathology, Erasmus MC, University Medical Center Rotterdam, 3015 GE Rotterdam, The Netherlands; (J.H.v.d.T.); (T.P.P.v.d.B.)
| | - Maria-Rosa Ghigna
- School of Medicine, Université Paris-Saclay, 94270 Le Kremlin-Bicêtre, France; (M.-R.G.); (F.P.); (D.M.)
- INSERM UMR_S 999, Pulmonary Hypertension: Pathology and Novel Therapies, Hôpital Marie Lannelongue, 92350 Le Plessis Robinson, France
- Division of Pathology, Marie Lannelongue Hospital, 92350 Le Plessis Robinson, France
| | - Frédéric Perros
- School of Medicine, Université Paris-Saclay, 94270 Le Kremlin-Bicêtre, France; (M.-R.G.); (F.P.); (D.M.)
- INSERM UMR_S 999, Pulmonary Hypertension: Pathology and Novel Therapies, Hôpital Marie Lannelongue, 92350 Le Plessis Robinson, France
| | - David Montani
- School of Medicine, Université Paris-Saclay, 94270 Le Kremlin-Bicêtre, France; (M.-R.G.); (F.P.); (D.M.)
- INSERM UMR_S 999, Pulmonary Hypertension: Pathology and Novel Therapies, Hôpital Marie Lannelongue, 92350 Le Plessis Robinson, France
- Department of Respiratory and Intensive Care Medicine, Pulmonary Hypertension National Referral Center, Assistance Publique—Hôpitaux de Paris (AP-HP), Hôpital Bicêtre, 94270 Le Kremlin-Bicêtre, France
| | - Mirjam Kool
- Department of Pulmonary Medicine, Erasmus MC, University Medical Center Rotterdam, 3015 GD Rotterdam, The Netherlands; (D.v.U.); (T.K.); (J.A.C.v.H.); (I.M.B.); (C.G.); (M.K.)
| | - Karin A. Boomars
- Department of Pulmonary Medicine, Erasmus MC, University Medical Center Rotterdam, 3015 GD Rotterdam, The Netherlands; (D.v.U.); (T.K.); (J.A.C.v.H.); (I.M.B.); (C.G.); (M.K.)
- Correspondence: (K.A.B.); (R.W.H.); Tel.: +316-50031911 (K.A.B.); +31-10-7043700 (R.W.H.)
| | - Rudi W. Hendriks
- Department of Pulmonary Medicine, Erasmus MC, University Medical Center Rotterdam, 3015 GD Rotterdam, The Netherlands; (D.v.U.); (T.K.); (J.A.C.v.H.); (I.M.B.); (C.G.); (M.K.)
- Correspondence: (K.A.B.); (R.W.H.); Tel.: +316-50031911 (K.A.B.); +31-10-7043700 (R.W.H.)
| |
Collapse
|
60
|
Canton J, Blees H, Henry CM, Buck MD, Schulz O, Rogers NC, Childs E, Zelenay S, Rhys H, Domart MC, Collinson L, Alloatti A, Ellison CJ, Amigorena S, Papayannopoulos V, Thomas DC, Randow F, Reis e Sousa C. The receptor DNGR-1 signals for phagosomal rupture to promote cross-presentation of dead-cell-associated antigens. Nat Immunol 2021; 22:140-153. [PMID: 33349708 PMCID: PMC7116638 DOI: 10.1038/s41590-020-00824-x] [Citation(s) in RCA: 97] [Impact Index Per Article: 32.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2020] [Accepted: 10/19/2020] [Indexed: 12/16/2022]
Abstract
Type 1 conventional dendritic (cDC1) cells are necessary for cross-presentation of many viral and tumor antigens to CD8+ T cells. cDC1 cells can be identified in mice and humans by high expression of DNGR-1 (also known as CLEC9A), a receptor that binds dead-cell debris and facilitates XP of corpse-associated antigens. Here, we show that DNGR-1 is a dedicated XP receptor that signals upon ligand engagement to promote phagosomal rupture. This allows escape of phagosomal contents into the cytosol, where they access the endogenous major histocompatibility complex class I antigen processing pathway. The activity of DNGR-1 maps to its signaling domain, which activates SYK and NADPH oxidase to cause phagosomal damage even when spliced into a heterologous receptor and expressed in heterologous cells. Our data reveal the existence of innate immune receptors that couple ligand binding to endocytic vesicle damage to permit MHC class I antigen presentation of exogenous antigens and to regulate adaptive immunity.
Collapse
Affiliation(s)
- Johnathan Canton
- Immunobiology Laboratory, The Francis Crick Institute, London, UK
| | - Hanna Blees
- Immunobiology Laboratory, The Francis Crick Institute, London, UK
| | - Conor M Henry
- Immunobiology Laboratory, The Francis Crick Institute, London, UK
| | - Michael D Buck
- Immunobiology Laboratory, The Francis Crick Institute, London, UK
| | - Oliver Schulz
- Immunobiology Laboratory, The Francis Crick Institute, London, UK
| | - Neil C Rogers
- Immunobiology Laboratory, The Francis Crick Institute, London, UK
| | - Eleanor Childs
- Immunobiology Laboratory, The Francis Crick Institute, London, UK
| | - Santiago Zelenay
- Cancer Inflammation and Immunity Group, CRUK Manchester Institute, The University of Manchester, Alderley Park, UK
| | - Hefin Rhys
- Flow Cytometry STP, The Francis Crick Institute, London, UK
| | | | - Lucy Collinson
- Electron Microscopy STP, The Francis Crick Institute, London, UK
| | - Andres Alloatti
- Centre de Recherche, INSERM U932, Institut Curie, Paris, France
| | - Cara J Ellison
- Division of Protein and Nucleic Acid Chemistry, MRC Laboratory of Molecular Biology, Cambridge, UK
- Department of Medicine, University of Cambridge, Addenbrooke's Hospital, Cambridge, UK
| | | | | | - David C Thomas
- Immunity and Inflammation, 9NC, Imperial College, London, UK
| | - Felix Randow
- Division of Protein and Nucleic Acid Chemistry, MRC Laboratory of Molecular Biology, Cambridge, UK
- Department of Medicine, University of Cambridge, Addenbrooke's Hospital, Cambridge, UK
| | | |
Collapse
|
61
|
Anderson DA, Dutertre CA, Ginhoux F, Murphy KM. Genetic models of human and mouse dendritic cell development and function. Nat Rev Immunol 2021; 21:101-115. [PMID: 32908299 PMCID: PMC10955724 DOI: 10.1038/s41577-020-00413-x] [Citation(s) in RCA: 163] [Impact Index Per Article: 54.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/28/2020] [Indexed: 12/13/2022]
Abstract
Dendritic cells (DCs) develop in the bone marrow from haematopoietic progenitors that have numerous shared characteristics between mice and humans. Human counterparts of mouse DC progenitors have been identified by their shared transcriptional signatures and developmental potential. New findings continue to revise models of DC ontogeny but it is well accepted that DCs can be divided into two main functional groups. Classical DCs include type 1 and type 2 subsets, which can detect different pathogens, produce specific cytokines and present antigens to polarize mainly naive CD8+ or CD4+ T cells, respectively. By contrast, the function of plasmacytoid DCs is largely innate and restricted to the detection of viral infections and the production of type I interferon. Here, we discuss genetic models of mouse DC development and function that have aided in correlating ontogeny with function, as well as how these findings can be translated to human DCs and their progenitors.
Collapse
Affiliation(s)
- David A Anderson
- Department of Pathology and Immunology, School of Medicine, Washington University in St. Louis, St. Louis, MO, USA
| | | | - Florent Ginhoux
- Singapore Immunology Network, Agency for Science, Technology and Research, Singapore, Singapore
- Shanghai Institute of Immunology, Department of Immunology and Microbiology, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Translational Immunology Institute, SingHealth Duke-NUS Academic Medical Centre, Singapore, Singapore
| | - Kenneth M Murphy
- Department of Pathology and Immunology, School of Medicine, Washington University in St. Louis, St. Louis, MO, USA.
- Howard Hughes Medical Institute, School of Medicine, Washington University in St. Louis, St. Louis, MO, USA.
| |
Collapse
|
62
|
Cabeza-Cabrerizo M, Cardoso A, Minutti CM, Pereira da Costa M, Reis E Sousa C. Dendritic Cells Revisited. Annu Rev Immunol 2021; 39:131-166. [PMID: 33481643 DOI: 10.1146/annurev-immunol-061020-053707] [Citation(s) in RCA: 357] [Impact Index Per Article: 119.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Dendritic cells (DCs) possess the ability to integrate information about their environment and communicate it to other leukocytes, shaping adaptive and innate immunity. Over the years, a variety of cell types have been called DCs on the basis of phenotypic and functional attributes. Here, we refocus attention on conventional DCs (cDCs), a discrete cell lineage by ontogenetic and gene expression criteria that best corresponds to the cells originally described in the 1970s. We summarize current knowledge of mouse and human cDC subsets and describe their hematopoietic development and their phenotypic and functional attributes. We hope that our effort to review the basic features of cDC biology and distinguish cDCs from related cell types brings to the fore the remarkable properties of this cell type while shedding some light on the seemingly inordinate complexity of the DC field.
Collapse
Affiliation(s)
- Mar Cabeza-Cabrerizo
- Immunobiology Laboratory, The Francis Crick Institute, London NW1 1AT, United Kingdom;
| | - Ana Cardoso
- Immunobiology Laboratory, The Francis Crick Institute, London NW1 1AT, United Kingdom;
| | - Carlos M Minutti
- Immunobiology Laboratory, The Francis Crick Institute, London NW1 1AT, United Kingdom;
| | | | - Caetano Reis E Sousa
- Immunobiology Laboratory, The Francis Crick Institute, London NW1 1AT, United Kingdom;
| |
Collapse
|
63
|
Environmental signals rather than layered ontogeny imprint the function of type 2 conventional dendritic cells in young and adult mice. Nat Commun 2021; 12:464. [PMID: 33469015 PMCID: PMC7815729 DOI: 10.1038/s41467-020-20659-2] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2020] [Accepted: 12/13/2020] [Indexed: 01/29/2023] Open
Abstract
Conventional dendritic cells (cDC) are key activators of naive T cells, and can be targeted in adults to induce adaptive immunity, but in early life are considered under-developed or functionally immature. Here we show that, in early life, when the immune system develops, cDC2 exhibit a dual hematopoietic origin and, like other myeloid and lymphoid cells, develop in waves. Developmentally distinct cDC2 in early life, despite being distinguishable by fate mapping, are transcriptionally and functionally similar. cDC2 in early and adult life, however, are exposed to distinct cytokine environments that shape their transcriptional profile and alter their ability to sense pathogens, secrete cytokines and polarize T cells. We further show that cDC2 in early life, despite being distinct from cDC2 in adult life, are functionally competent and can induce T cell responses. Our results thus highlight the potential of harnessing cDC2 for boosting immunity in early life.
Collapse
|
64
|
Wu X, Brombacher F, Chroneos ZC, Norbury CC, Gowda DC. IL-4Rα signaling by CD8α + dendritic cells contributes to cerebral malaria by enhancing inflammatory, Th1, and cytotoxic CD8 + T cell responses. J Biol Chem 2021; 296:100615. [PMID: 33798555 PMCID: PMC8100064 DOI: 10.1016/j.jbc.2021.100615] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2020] [Revised: 03/12/2021] [Accepted: 03/29/2021] [Indexed: 11/20/2022] Open
Abstract
Persistent high levels of proinflammatory and Th1 responses contribute to cerebral malaria (CM). Suppression of inflammatory responses and promotion of Th2 responses prevent pathogenesis. IL-4 commonly promotes Th2 responses and inhibits inflammatory and Th1 responses. Therefore, IL-4 is widely considered as a beneficial cytokine via its Th2-promoting role that is predicted to provide protection against severe malaria by inhibiting inflammatory responses. However, IL-4 may also induce inflammatory responses, as the result of IL-4 action depends on the timing and levels of its production and the tissue environment in which it is produced. Recently, we showed that dendritic cells (DCs) produce IL-4 early during malaria infection in response to a parasite protein and that this IL-4 response may contribute to severe malaria. However, the mechanism by which IL-4 produced by DCs contributing to lethal malaria is unknown. Using Plasmodium berghei ANKA-infected C57BL/6 mice, a CM model, we show here that mice lacking IL-4Rα only in CD8α+ DCs are protected against CM pathogenesis and survive, whereas WT mice develop CM and die. Compared with WT mice, mice lacking IL-4Rα in CD11c+ or CD8α+ DCs showed reduced inflammatory responses leading to decreased Th1 and cytotoxic CD8+ T cell responses, lower infiltration of CD8+ T cells to the brain, and negligible brain pathology. The novel results presented here reveal a paradoxical role of IL-4Rα signaling in CM pathogenesis that promotes CD8α+ DC-mediated inflammatory responses that generate damaging Th1 and cytotoxic CD8+ T cell responses.
Collapse
Affiliation(s)
- Xianzhu Wu
- Department of Biochemistry and Molecular Biology, The Pennsylvania State University College of Medicine, Hershey, Pennsylvania, USA
| | - Frank Brombacher
- Component & Division of Immunology, Faculty of Health Science, International Centre for Genetic Engineering and Biotechnology (ICGEB), University of Cape Town, Cape Town, South Africa
| | - Zissis C Chroneos
- Department of Pediatrics, The Pennsylvania State University College of Medicine, Hershey, Pennsylvania, USA
| | - Christopher C Norbury
- Department of Microbiology and Immunology, The Pennsylvania State University College of Medicine, Hershey, Pennsylvania, USA
| | - D Channe Gowda
- Department of Biochemistry and Molecular Biology, The Pennsylvania State University College of Medicine, Hershey, Pennsylvania, USA.
| |
Collapse
|
65
|
Koudstaal T, van Hulst JAC, Das T, Neys SFH, Merkus D, Bergen IM, de Raaf MA, Bogaard HJ, Boon L, van Loo G, Aerts JGJV, Boomars KA, Kool M, Hendriks RW. DNGR1-Cre-mediated Deletion of Tnfaip3/A20 in Conventional Dendritic Cells Induces Pulmonary Hypertension in Mice. Am J Respir Cell Mol Biol 2020; 63:665-680. [PMID: 32755457 DOI: 10.1165/rcmb.2019-0443oc] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Chronic perivascular inflammation is a prominent feature in the lungs of idiopathic pulmonary arterial hypertension. Although the proportions of conventional dendritic cells (cDCs) and plasmacytoid DCs are increased in idiopathic pulmonary arterial hypertension lungs, it remains unknown whether activated cDCs play a pathogenic role. The Tnfaip3 gene encodes the ubiquitin-binding protein A20, which is a negative regulator of NF-κB, critically involved in DC activation. Targeting of Tnfaip3/A20 in cDCs was achieved by Clec9a (DNGR1)-Cre-mediated excision of the Tnfaip3 gene in Tnfaip3DNGR1-KO mice. Mice were evaluated for signs of pulmonary hypertension (PH) using right heart catheterization, echocardiography, and measurement of the Fulton index. Inflammation was assessed by immunohistochemistry and flow cytometry. Pulmonary cDCs and monocyte-derived DCs from 31-week-old Tnfaip3DNGR1-KO mice showed modulated expression of cell surface activation markers compared with Tnfaip3DNGR1-WT mice. Tnfaip3DNGR1-KO mice developed elevated right ventricular systolic pressure and right ventricular hypertrophy. The lungs of these mice displayed increased vascular remodeling and perivascular and peribronchial immune cell infiltration resembling tertiary lymphoid organs. Proportions of activated T cells and expression of IL-1β, IL-6, and IL-10 were enhanced in the lungs of Tnfaip3DNGR1-KO mice. Autoreactive IgA and IgG1 was detected in BAL and autoreactive IgA recognizing pulmonary endothelial antigens was present in the serum of Tnfaip3DNGR1-KO mice. All signs of PH were ameliorated in Tnfaip3DNGR1-KO mice by anti-IL-6 antibody treatment. These results indicate that activation of the NF-κB pathway in DCs, through deletion of A20/Tnfaip3, leads to experimental PH with accompanied pulmonary inflammation in an IL-6-dependent fashion.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Michiel A de Raaf
- Department of Neonatology, Erasmus Medical Centre, Rotterdam, the Netherlands.,VU Medical Centre, Amsterdam, the Netherlands
| | | | - Louis Boon
- Polpharma Biologics, Utrecht, the Netherlands
| | - Geert van Loo
- VIB Center for Inflammation Research, Ghent, Belgium; and.,Department of Biomedical Molecular Biology, Ghent University, Ghent, Belgium
| | | | | | | | | |
Collapse
|
66
|
Venosa A. Senescence in Pulmonary Fibrosis: Between Aging and Exposure. Front Med (Lausanne) 2020; 7:606462. [PMID: 33282895 PMCID: PMC7689159 DOI: 10.3389/fmed.2020.606462] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2020] [Accepted: 10/23/2020] [Indexed: 12/15/2022] Open
Abstract
To date, chronic pulmonary pathologies represent the third leading cause of death in the elderly population. Evidence-based projections suggest that >65 (years old) individuals will account for approximately a quarter of the world population before the turn of the century. Genomic instability, telomere attrition, epigenetic alterations, loss of proteostasis, deregulated nutrient sensing, mitochondrial dysfunction, cellular senescence, stem cell exhaustion, and altered intercellular communication, are described as the nine “hallmarks” that govern cellular fitness. Any deviation from the normal pattern initiates a complex cascade of events culminating to a disease state. This blueprint, originally employed to describe aberrant changes in cancer cells, can be also used to describe aging and fibrosis. Pulmonary fibrosis (PF) is the result of a progressive decline in injury resolution processes stemming from endogenous (physiological decline or somatic mutations) or exogenous stress. Environmental, dietary or occupational exposure accelerates the pathogenesis of a senescent phenotype based on (1) window of exposure; (2) dose, duration, recurrence; and (3) cells type being targeted. As the lung ages, the threshold to generate an irreversibly senescent phenotype is lowered. However, we do not have sufficient knowledge to make accurate predictions. In this review, we provide an assessment of the literature that interrogates lung epithelial, mesenchymal, and immune senescence at the intersection of aging, environmental exposure and pulmonary fibrosis.
Collapse
Affiliation(s)
- Alessandro Venosa
- Department of Pharmacology and Toxicology, University of Utah College of Pharmacy, Salt Lake City, UT, United States
| |
Collapse
|
67
|
Abstract
After both sterile and infectious insults, damage is inflicted on tissues leading to accidental or programmed cell death. In addition, events of programmed cell death also take place under homeostatic conditions, such as in embryo development or in the turnover of hematopoietic cells. Mammalian tissues are seeded with myeloid immune cells, which harbor a plethora of receptors that allow the detection of cell death, modulating immune responses. The myeloid C-type lectin receptors (CLRs) are one of the most prominent families of receptors involved in tailoring immunity after sensing dead cells. In this chapter, we will cover a diversity of signals arising from different forms of cell death and how they are recognized by myeloid CLRs. We will also explore how myeloid cells develop their sentinel function, exploring how some of these CLRs identify cell death and the type of responses triggered thereof. In particular, we will focus on DNGR-1 (CLEC9A), Mincle (CLEC4E), CLL-1 (CLEC12A), LOX-1 (OLR1), CD301 (CLEC10A) and DEC-205 (LY75) as paradigmatic death-sensing CLRs expressed by myeloid cells. The molecular processes triggered after cell death recognition by myeloid CLRs contribute to the regulation of immune responses in pathologies associated with tissue damage, such as infection, autoimmunity and cancer. A better understanding of these processes may help to improve the current approaches for therapeutic intervention.
Collapse
|
68
|
Gallizioli M, Miró-Mur F, Otxoa-de-Amezaga A, Cugota R, Salas-Perdomo A, Justicia C, Brait VH, Ruiz-Jaén F, Arbaizar-Rovirosa M, Pedragosa J, Bonfill-Teixidor E, Gelderblom M, Magnus T, Cano E, Del Fresno C, Sancho D, Planas AM. Dendritic Cells and Microglia Have Non-redundant Functions in the Inflamed Brain with Protective Effects of Type 1 cDCs. Cell Rep 2020; 33:108291. [PMID: 33086061 PMCID: PMC7578563 DOI: 10.1016/j.celrep.2020.108291] [Citation(s) in RCA: 41] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2020] [Revised: 09/02/2020] [Accepted: 09/29/2020] [Indexed: 01/14/2023] Open
Abstract
Brain CD11c+ cells share features with microglia and dendritic cells (DCs). Sterile inflammation increases brain CD11c+ cells, but their phenotype, origin, and functions remain largely unknown. We report that, after cerebral ischemia, microglia attract DCs to the inflamed brain, and astroglia produce Flt3 ligand, supporting development and expansion of CD11c+ cells. CD11c+ cells in the inflamed brain are a complex population derived from proliferating microglia and infiltrating DCs, including a major subset of OX40L+ conventional cDC2, and also cDC1, plasmacytoid, and monocyte-derived DCs. Despite sharing certain morphological features and markers, CD11c+ microglia and DCs display differential expression of pattern recognition receptors and chemokine receptors. DCs excel CD11c- and CD11c+ microglia in the capacity to present antigen through MHCI and MHCII. Of note, cDC1s protect from brain injury after ischemia. We thus reveal aspects of the dynamics and functions of brain DCs in the regulation of inflammation and immunity.
Collapse
Affiliation(s)
- Mattia Gallizioli
- Department of Brain Ischemia and Neurodegeneration, Institut d'Investigacions Biomèdiques de Barcelona (IIBB), Consejo Superior de Investigaciones Científicas (CSIC), Barcelona 08036, Spain; Area of Neurosciences, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona 08036, Spain
| | - Francesc Miró-Mur
- Area of Neurosciences, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona 08036, Spain; Fundació Clínic, Barcelona 08036, Spain
| | - Amaia Otxoa-de-Amezaga
- Department of Brain Ischemia and Neurodegeneration, Institut d'Investigacions Biomèdiques de Barcelona (IIBB), Consejo Superior de Investigaciones Científicas (CSIC), Barcelona 08036, Spain; Area of Neurosciences, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona 08036, Spain
| | - Roger Cugota
- Department of Brain Ischemia and Neurodegeneration, Institut d'Investigacions Biomèdiques de Barcelona (IIBB), Consejo Superior de Investigaciones Científicas (CSIC), Barcelona 08036, Spain
| | - Angélica Salas-Perdomo
- Department of Brain Ischemia and Neurodegeneration, Institut d'Investigacions Biomèdiques de Barcelona (IIBB), Consejo Superior de Investigaciones Científicas (CSIC), Barcelona 08036, Spain; Fundació Clínic, Barcelona 08036, Spain
| | - Carles Justicia
- Department of Brain Ischemia and Neurodegeneration, Institut d'Investigacions Biomèdiques de Barcelona (IIBB), Consejo Superior de Investigaciones Científicas (CSIC), Barcelona 08036, Spain; Area of Neurosciences, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona 08036, Spain
| | - Vanessa H Brait
- Area of Neurosciences, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona 08036, Spain
| | - Francisca Ruiz-Jaén
- Department of Brain Ischemia and Neurodegeneration, Institut d'Investigacions Biomèdiques de Barcelona (IIBB), Consejo Superior de Investigaciones Científicas (CSIC), Barcelona 08036, Spain; Area of Neurosciences, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona 08036, Spain
| | - Maria Arbaizar-Rovirosa
- Department of Brain Ischemia and Neurodegeneration, Institut d'Investigacions Biomèdiques de Barcelona (IIBB), Consejo Superior de Investigaciones Científicas (CSIC), Barcelona 08036, Spain; Area of Neurosciences, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona 08036, Spain
| | - Jordi Pedragosa
- Department of Brain Ischemia and Neurodegeneration, Institut d'Investigacions Biomèdiques de Barcelona (IIBB), Consejo Superior de Investigaciones Científicas (CSIC), Barcelona 08036, Spain; Area of Neurosciences, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona 08036, Spain
| | - Ester Bonfill-Teixidor
- Department of Brain Ischemia and Neurodegeneration, Institut d'Investigacions Biomèdiques de Barcelona (IIBB), Consejo Superior de Investigaciones Científicas (CSIC), Barcelona 08036, Spain
| | - Mathias Gelderblom
- Department of Neurology, University Medical Center Hamburg-Eppendorf, Hamburg 20251, Germany
| | - Tim Magnus
- Department of Neurology, University Medical Center Hamburg-Eppendorf, Hamburg 20251, Germany
| | - Eva Cano
- Neuroinflammation Unit, Unidad Funcional de Investigación de Enfermedades Crónicas, Instituto de Salud Carlos III, Majadahonda, Madrid 28222, Spain
| | - Carlos Del Fresno
- Centro Nacional de Investigaciones Cardiovasculares (CNIC), Madrid 28029, Spain
| | - David Sancho
- Centro Nacional de Investigaciones Cardiovasculares (CNIC), Madrid 28029, Spain
| | - Anna M Planas
- Department of Brain Ischemia and Neurodegeneration, Institut d'Investigacions Biomèdiques de Barcelona (IIBB), Consejo Superior de Investigaciones Científicas (CSIC), Barcelona 08036, Spain; Area of Neurosciences, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona 08036, Spain.
| |
Collapse
|
69
|
Sun T, Nguyen A, Gommerman JL. Dendritic Cell Subsets in Intestinal Immunity and Inflammation. THE JOURNAL OF IMMUNOLOGY 2020; 204:1075-1083. [PMID: 32071090 DOI: 10.4049/jimmunol.1900710] [Citation(s) in RCA: 49] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/24/2019] [Accepted: 10/11/2019] [Indexed: 12/21/2022]
Abstract
The mammalian intestine is a complex environment that is constantly exposed to Ags derived from food, microbiota, and metabolites. Intestinal dendritic cells (DC) have the responsibility of establishing oral tolerance against these Ags while initiating immune responses against mucosal pathogens. We now know that DC are a heterogeneous population of innate immune cells composed of classical and monocyte-derived DC, Langerhans cells, and plasmacytoid DC. In the intestine, DC are found in organized lymphoid tissues, such as the mesenteric lymph nodes and Peyer's patches, as well as in the lamina propria. In this Brief Review, we review recent work that describes a division of labor between and collaboration among gut DC subsets in the context of intestinal homeostasis and inflammation. Understanding relationships between DC subtypes and their biological functions will rationalize oral vaccine design and will provide insights into treatments that quiet pathological intestinal inflammation.
Collapse
Affiliation(s)
- Tian Sun
- Department of Immunology, Faculty of Medicine, University of Toronto, Toronto, Ontario M5S1A8, Canada
| | - Albert Nguyen
- Department of Immunology, Faculty of Medicine, University of Toronto, Toronto, Ontario M5S1A8, Canada
| | - Jennifer L Gommerman
- Department of Immunology, Faculty of Medicine, University of Toronto, Toronto, Ontario M5S1A8, Canada
| |
Collapse
|
70
|
de Winde CM, Munday C, Acton SE. Molecular mechanisms of dendritic cell migration in immunity and cancer. Med Microbiol Immunol 2020; 209:515-529. [PMID: 32451606 PMCID: PMC7395046 DOI: 10.1007/s00430-020-00680-4] [Citation(s) in RCA: 42] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2020] [Accepted: 05/07/2020] [Indexed: 12/18/2022]
Abstract
Dendritic cells (DCs) are a heterogeneous population of antigen-presenting cells that act to bridge innate and adaptive immunity. DCs are critical in mounting effective immune responses to tissue damage, pathogens and cancer. Immature DCs continuously sample tissues and engulf antigens via endocytic pathways such as phagocytosis or macropinocytosis, which result in DC activation. Activated DCs undergo a maturation process by downregulating endocytosis and upregulating surface proteins controlling migration to lymphoid tissues where DC-mediated antigen presentation initiates adaptive immune responses. To traffic to lymphoid tissues, DCs must adapt their motility mechanisms to migrate within a wide variety of tissue types and cross barriers to enter lymphatics. All steps of DC migration involve cell-cell or cell-substrate interactions. This review discusses DC migration mechanisms in immunity and cancer with a focus on the role of cytoskeletal processes and cell surface proteins, including integrins, lectins and tetraspanins. Understanding the adapting molecular mechanisms controlling DC migration in immunity provides the basis for therapeutic interventions to dampen immune activation in autoimmunity, or to improve anti-tumour immune responses.
Collapse
Affiliation(s)
- Charlotte M de Winde
- Stromal Immunology Group, MRC Laboratory for Molecular Cell Biology, University College London, Gower Street, London, WC1E 6BT, UK.
| | - Clare Munday
- Stromal Immunology Group, MRC Laboratory for Molecular Cell Biology, University College London, Gower Street, London, WC1E 6BT, UK
| | - Sophie E Acton
- Stromal Immunology Group, MRC Laboratory for Molecular Cell Biology, University College London, Gower Street, London, WC1E 6BT, UK
| |
Collapse
|
71
|
Ramachandran P, Matchett KP, Dobie R, Wilson-Kanamori JR, Henderson NC. Single-cell technologies in hepatology: new insights into liver biology and disease pathogenesis. Nat Rev Gastroenterol Hepatol 2020; 17:457-472. [PMID: 32483353 DOI: 10.1038/s41575-020-0304-x] [Citation(s) in RCA: 131] [Impact Index Per Article: 32.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 04/08/2020] [Indexed: 12/19/2022]
Abstract
Liver disease is a major global health-care problem, affecting an estimated 844 million people worldwide. Despite this substantial burden, therapeutic options for liver disease remain limited, in part owing to a paucity of detailed analyses defining the cellular and molecular mechanisms that drive these conditions in humans. Single-cell transcriptomic technologies are transforming our understanding of cellular diversity and function in health and disease. In this Review, we discuss how these technologies have been applied in hepatology, advancing our understanding of cellular heterogeneity and providing novel insights into fundamental liver biology such as the metabolic zonation of hepatocytes, endothelial cells and hepatic stellate cells, and the cellular mechanisms underpinning liver regeneration. Application of these methodologies is also uncovering critical pathophysiological changes driving disease states such as hepatic fibrosis, where distinct populations of macrophages, endothelial cells and mesenchymal cells reside within a spatially distinct fibrotic niche and interact to promote scar formation. In addition, single-cell approaches are starting to dissect key cellular and molecular functions in liver cancer. In the near future, new techniques such as spatial transcriptomics and multiomic approaches will further deepen our understanding of disease pathogenesis, enabling the identification of novel therapeutic targets for patients across the spectrum of liver diseases.
Collapse
Affiliation(s)
- Prakash Ramachandran
- Centre for Inflammation Research, The Queen's Medical Research Institute, University of Edinburgh, Edinburgh, UK
| | - Kylie P Matchett
- Centre for Inflammation Research, The Queen's Medical Research Institute, University of Edinburgh, Edinburgh, UK
| | - Ross Dobie
- Centre for Inflammation Research, The Queen's Medical Research Institute, University of Edinburgh, Edinburgh, UK
| | - John R Wilson-Kanamori
- Centre for Inflammation Research, The Queen's Medical Research Institute, University of Edinburgh, Edinburgh, UK
| | - Neil C Henderson
- Centre for Inflammation Research, The Queen's Medical Research Institute, University of Edinburgh, Edinburgh, UK. .,MRC Human Genetics Unit, Institute of Genetics and Molecular Medicine, University of Edinburgh, Edinburgh, UK.
| |
Collapse
|
72
|
Balam S, Kesselring R, Eggenhofer E, Blaimer S, Evert K, Evert M, Schlitt HJ, Geissler EK, van Blijswijk J, Lee S, Reis e Sousa C, Brunner SM, Fichtner-Feigl S. Cross-presentation of dead-cell-associated antigens by DNGR-1 + dendritic cells contributes to chronic allograft rejection in mice. Eur J Immunol 2020; 50:2041-2054. [PMID: 32640051 DOI: 10.1002/eji.201948501] [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: 12/23/2019] [Revised: 05/12/2020] [Accepted: 07/02/2020] [Indexed: 01/06/2023]
Abstract
The purpose of this study was to elucidate whether DC NK lectin group receptor-1 (DNGR-1)-dependent cross-presentation of dead-cell-associated antigens occurs after transplantation and contributes to CD8+ T cell responses, chronic allograft rejection (CAR), and fibrosis. BALB/c or C57BL/6 hearts were heterotopically transplanted into WT, Clec9a-/- , or Batf3-/- recipient C57BL/6 mice. Allografts were analyzed for cell infiltration, CD8+ T cell activation, fibrogenesis, and CAR using immunohistochemistry, Western blot, qRT2 -PCR, and flow cytometry. Allografts displayed infiltration by recipient DNGR-1+ DCs, signs of CAR, and fibrosis. Allografts in Clec9a-/- recipients showed reduced CAR (p < 0.0001), fibrosis (P = 0.0137), CD8+ cell infiltration (P < 0.0001), and effector cytokine levels compared to WT recipients. Batf3-deficiency greatly reduced DNGR-1+ DC-infiltration, CAR (P < 0.0001), and fibrosis (P = 0.0382). CD8 cells infiltrating allografts of cytochrome C treated recipients, showed reduced production of CD8 effector cytokines (P < 0.05). Further, alloreactive CD8+ T cell response in indirect pathway IFN-γ ELISPOT was reduced in Clec9a-/- recipient mice (P = 0.0283). Blockade of DNGR-1 by antibody, similar to genetic elimination of the receptor, reduced CAR (P = 0.0003), fibrosis (P = 0.0273), infiltration of CD8+ cells (p = 0.0006), and effector cytokine levels. DNGR-1-dependent alloantigen cross-presentation by DNGR-1+ DCs induces alloreactive CD8+ cells that induce CAR and fibrosis. Antibody against DNGR-1 can block this process and prevent CAR and fibrosis.
Collapse
Affiliation(s)
- Saidou Balam
- Department of Surgery, University Medical Center Regensburg, Regensburg, Germany
| | - Rebecca Kesselring
- Department of Surgery, University Medical Center Regensburg, Regensburg, Germany
| | - Elke Eggenhofer
- Department of Surgery, University Medical Center Regensburg, Regensburg, Germany
| | - Stephanie Blaimer
- Department of Surgery, University Medical Center Regensburg, Regensburg, Germany
| | - Katja Evert
- Department of Pathology, University Medical Center Regensburg, Regensburg, Germany
| | - Matthias Evert
- Department of Pathology, University Medical Center Regensburg, Regensburg, Germany
| | - Hans J Schlitt
- Department of Surgery, University Medical Center Regensburg, Regensburg, Germany
| | - Edward K Geissler
- Department of Surgery, University Medical Center Regensburg, Regensburg, Germany
| | | | - Sonia Lee
- Immunobiology Laboratory, The Francis Crick Institute, London, UK
| | | | - Stefan M Brunner
- Department of Surgery, University Medical Center Regensburg, Regensburg, Germany
| | - Stefan Fichtner-Feigl
- Department of Surgery, University Medical Center Regensburg, Regensburg, Germany.,Department of General and Visceral Surgery, University Medical Center Freiburg, Freiburg, Germany
| |
Collapse
|
73
|
Naik SH. Dendritic cell development at a clonal level within a revised 'continuous' model of haematopoiesis. Mol Immunol 2020; 124:190-197. [PMID: 32593782 DOI: 10.1016/j.molimm.2020.06.012] [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: 02/04/2020] [Revised: 04/15/2020] [Accepted: 06/11/2020] [Indexed: 12/17/2022]
Abstract
Understanding development of the dendritic cell (DC) subtypes continues to evolve. The origin and relationship of conventional DC type 1 (cDC1), cDC type 2 (cDC2) and plasmacytoid DCs (pDCs) to each other, and in relation to classic myeloid and lymphoid cells, has had a long and controversial history and is still not fully resolved. This review summarises the technological developments and findings that have been achieved at a clonal level, and how that has enhanced our knowledge of the process. It summarises the single cell lineage tracing technologies that have emerged, their application in in vitro and in vivo studies, in both mouse and human settings, and places the findings in a wider context of understanding haematopoiesis at a single cell or clonal level. In particular, it addresses the fate heterogeneity observed in many phenotypically defined progenitor subsets and how these findings have led to a departure from the classic ball-and-stick models of haematopoiesis to the emerging continuous model. Prior contradictions in DC development may be reconciled if they are framed within this revised model, where commitment to a lineage or cell type does not occur in an all-or-nothing process in defined progenitors but rather can occur at many stages of haematopoiesis in a dynamic process.
Collapse
Affiliation(s)
- Shalin H Naik
- Immunology Division, The Walter & Eliza Hall Institute of Medical Research, Parkville, Australia; The Department of Medical Biology, The University of Melbourne, Parkville, Australia.
| |
Collapse
|
74
|
PD-L1 expression by dendritic cells is a key regulator of T-cell immunity in cancer. ACTA ACUST UNITED AC 2020; 1:681-691. [DOI: 10.1038/s43018-020-0075-x] [Citation(s) in RCA: 137] [Impact Index Per Article: 34.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2020] [Accepted: 05/13/2020] [Indexed: 12/12/2022]
|
75
|
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]
|
76
|
Ferrer IR, West HC, Henderson S, Ushakov DS, Santos E Sousa P, Strid J, Chakraverty R, Yates AJ, Bennett CL. A wave of monocytes is recruited to replenish the long-term Langerhans cell network after immune injury. Sci Immunol 2020; 4:4/38/eaax8704. [PMID: 31444235 DOI: 10.1126/sciimmunol.aax8704] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2019] [Accepted: 07/15/2019] [Indexed: 12/21/2022]
Abstract
A dense population of embryo-derived Langerhans cells (eLCs) is maintained within the sealed epidermis without contribution from circulating cells. When this network is perturbed by transient exposure to ultraviolet light, short-term LCs are temporarily reconstituted from an initial wave of monocytes but thought to be superseded by more permanent repopulation with undefined LC precursors. However, the extent to which this process is relevant to immunopathological processes that damage LC population integrity is not known. Using a model of allogeneic hematopoietic stem cell transplantation, where alloreactive T cells directly target eLCs, we have asked whether and how the original LC network is ultimately restored. We find that donor monocytes, but not dendritic cells, are the precursors of long-term LCs in this context. Destruction of eLCs leads to recruitment of a wave of monocytes that engraft in the epidermis and undergo a sequential pathway of differentiation via transcriptionally distinct EpCAM+ precursors. Monocyte-derived LCs acquire the capacity of self-renewal, and proliferation in the epidermis matched that of steady-state eLCs. However, we identified a bottleneck in the differentiation and survival of epidermal monocytes, which, together with the slow rate of renewal of mature LCs, limits repair of the network. Furthermore, replenishment of the LC network leads to constitutive entry of cells into the epidermal compartment. Thus, immune injury triggers functional adaptation of mechanisms used to maintain tissue-resident macrophages at other sites, but this process is highly inefficient in the skin.
Collapse
Affiliation(s)
- Ivana R Ferrer
- Institute for Immunity and Transplantation, Division of Infection and Immunity, University College London, London NW3 2PF, UK and Cancer Institute Department of Haematology, Division of Cancer Studies, University College London, London WC1E 6DD, UK
| | - Heather C West
- Institute for Immunity and Transplantation, Division of Infection and Immunity, University College London, London NW3 2PF, UK and Cancer Institute Department of Haematology, Division of Cancer Studies, University College London, London WC1E 6DD, UK
| | - Stephen Henderson
- Bill Lyons Informatics Centre, Cancer Institute, University College London, London WC1E 6DD, UK
| | - Dmitry S Ushakov
- Peter Gorer Department of Immunobiology, School of Immunology and Microbial Sciences, King's College London, New Hunt's House, Newcomen Street, London SE1 1UL, UK
| | - Pedro Santos E Sousa
- Institute for Immunity and Transplantation, Division of Infection and Immunity, University College London, London NW3 2PF, UK and Cancer Institute Department of Haematology, Division of Cancer Studies, University College London, London WC1E 6DD, UK
| | - Jessica Strid
- Division of Immunology and Inflammation, Imperial College London, Hammersmith Campus, London W12 0NN, UK
| | - Ronjon Chakraverty
- Institute for Immunity and Transplantation, Division of Infection and Immunity, University College London, London NW3 2PF, UK and Cancer Institute Department of Haematology, Division of Cancer Studies, University College London, London WC1E 6DD, UK
| | - Andrew J Yates
- Department of Pathology and Cell Biology, Columbia University Medical Center, New York, NY 10032, USA
| | - Clare L Bennett
- Institute for Immunity and Transplantation, Division of Infection and Immunity, University College London, London NW3 2PF, UK and Cancer Institute Department of Haematology, Division of Cancer Studies, University College London, London WC1E 6DD, UK.
| |
Collapse
|
77
|
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
|
78
|
Inflammatory Type 2 cDCs Acquire Features of cDC1s and Macrophages to Orchestrate Immunity to Respiratory Virus Infection. Immunity 2020; 52:1039-1056.e9. [PMID: 32392463 PMCID: PMC7207120 DOI: 10.1016/j.immuni.2020.04.005] [Citation(s) in RCA: 238] [Impact Index Per Article: 59.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2019] [Revised: 03/05/2020] [Accepted: 04/14/2020] [Indexed: 02/07/2023]
Abstract
The phenotypic and functional dichotomy between IRF8+ type 1 and IRF4+ type 2 conventional dendritic cells (cDC1s and cDC2s, respectively) is well accepted; it is unknown how robust this dichotomy is under inflammatory conditions, when additionally monocyte-derived cells (MCs) become competent antigen-presenting cells (APCs). Using single-cell technologies in models of respiratory viral infection, we found that lung cDC2s acquired expression of the Fc receptor CD64 shared with MCs and of IRF8 shared with cDC1s. These inflammatory cDC2s (inf-cDC2s) were superior in inducing CD4+ T helper (Th) cell polarization while simultaneously presenting antigen to CD8+ T cells. When carefully separated from inf-cDC2s, MCs lacked APC function. Inf-cDC2s matured in response to cell-intrinsic Toll-like receptor and type 1 interferon receptor signaling, upregulated an IRF8-dependent maturation module, and acquired antigens via convalescent serum and Fc receptors. Because hybrid inf-cDC2s are easily confused with monocyte-derived cells, their existence could explain why APC functions have been attributed to MCs. Type I interferon drives differentiation of inf-cDC2s that closely resemble MCs Inf-cDC2s prime CD4+ and CD8+ T cells, whereas MCs lack APC function Inf-cDC2s internalize antibody-complexed antigen via Fc receptors IRF8 controls maturation gene module in inf-cDC2s
Collapse
|
79
|
Kidney dendritic cells: fundamental biology and functional roles in health and disease. Nat Rev Nephrol 2020; 16:391-407. [PMID: 32372062 DOI: 10.1038/s41581-020-0272-y] [Citation(s) in RCA: 60] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/18/2020] [Indexed: 02/06/2023]
Abstract
Dendritic cells (DCs) are chief inducers of adaptive immunity and regulate local inflammatory responses across the body. Together with macrophages, the other main type of mononuclear phagocyte, DCs constitute the most abundant component of the intrarenal immune system. This network of functionally specialized immune cells constantly surveys its microenvironment for signs of injury or infection, which trigger the initiation of an immune response. In the healthy kidney, DCs coordinate effective immune responses, for example, by recruiting neutrophils for bacterial clearance in pyelonephritis. The pro-inflammatory actions of DCs can, however, also contribute to tissue damage in various types of acute kidney injury and chronic glomerulonephritis, as DCs recruit and activate effector T cells, which release toxic mediators and maintain tubulointerstitial immune infiltrates. These actions are counterbalanced by DC subsets that promote the activation and maintenance of regulatory T cells to support resolution of the immune response and allow kidney repair. Several studies have investigated the multiple roles for DCs in kidney homeostasis and disease, but it has become clear that current tools and subset markers are not sufficient to accurately distinguish DCs from macrophages. Multidimensional transcriptomic analysis studies promise to improve mononuclear phagocyte classification and provide a clearer view of DC ontogeny and subsets.
Collapse
|
80
|
The MYCL and MXD1 transcription factors regulate the fitness of murine dendritic cells. Proc Natl Acad Sci U S A 2020; 117:4885-4893. [PMID: 32071205 PMCID: PMC7060746 DOI: 10.1073/pnas.1915060117] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
We previously found that MYCL is required by a Batf3-dependent classical dendritic cell subset (cDC1) for optimal CD8 T cell priming, but the underlying mechanism has remained unclear. The MAX-binding proteins encompass a family of transcription factors with overlapping DNA-binding specificities, conferred by a C-terminal basic helix-loop-helix domain, which mediates heterodimerization. Thus, regulation of transcription by these factors is dependent on divergent N-terminal domains. The MYC family, including MYCL, has actions that are reciprocal to the MXD family, which is mediated through the recruitment of higher-order activator and repressor complexes, respectively. As potent proto-oncogenes, models of MYC family function have been largely derived from their activity at supraphysiological levels in tumor cell lines. MYC and MYCN have been studied extensively, but empirical analysis of MYCL function had been limited due to highly restricted, lineage-specific expression in vivo. Here we observed that Mycl is expressed in immature cDC1s but repressed on maturation, concomitant with Mxd1 induction in mature cDC1s. We hypothesized that MYCL and MXD1 regulate a shared, but reciprocal, transcriptional program during cDC1 maturation. In agreement, immature cDC1s in Mycl -/- -deficient mice exhibited reduced expression of genes that regulate core biosynthetic processes. Mature cDC1s from Mxd1 -/- mice exhibited impaired ability to inhibit the transcriptional signature otherwise supported by MYCL. The present study reveals LMYC and MXD1 as regulators of a transcriptional program that is modulated during the maturation of Batf3-dependent cDC1s.
Collapse
|
81
|
Drouin M, Saenz J, Chiffoleau E. C-Type Lectin-Like Receptors: Head or Tail in Cell Death Immunity. Front Immunol 2020; 11:251. [PMID: 32133013 PMCID: PMC7040094 DOI: 10.3389/fimmu.2020.00251] [Citation(s) in RCA: 68] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2019] [Accepted: 01/30/2020] [Indexed: 12/12/2022] Open
Abstract
C-type lectin-like receptors (CLRs) represent a family of transmembrane pattern recognition receptors, expressed primarily by myeloid cells. They recognize not only pathogen moieties for host defense, but also modified self-antigens such as damage-associated molecular patterns released from dead cells. Upon ligation, CLR signaling leads to the production of inflammatory mediators to shape amplitude, duration and outcome of the immune response. Thus, following excessive injury, dysregulation of these receptors leads to the development of inflammatory diseases. Herein, we will focus on four CLRs of the "Dectin family," shown to decode the immunogenicity of cell death. CLEC9A on dendritic cells links F-actin exposed by dying cells to favor cross-presentation of dead-cell associated antigens to CD8+ T cells. Nevertheless, CLEC9A exerts also feedback mechanisms to temper neutrophil recruitment and prevent additional tissue damage. MINCLE expressed by macrophages binds nuclear SAP130 released by necrotic cells to potentiate pro-inflammatory responses. However, the consequent inflammation can exacerbate pathogenesis of inflammatory diseases. Moreover, in a tumor microenvironment, MINCLE induces macrophage-induced immune suppression and cancer progression. Similarly, triggering of LOX-1 by oxidized LDL, amplifies pro-inflammatory response but promotes tumor immune escape and metastasis. Finally, CLEC12A that recognizes monosodium urate crystals formed during cell death, inhibits activating signals to prevent detrimental inflammation. Interestingly, CLEC12A also sustains type-I IFN response to finely tune immune responses in case of viral-induced collateral damage. Therefore, CLRs acting in concert as sensors of injury, could be used in a targeted way to treat numerous diseases such as allergies, obesity, tumors, and autoimmunity.
Collapse
Affiliation(s)
- Marion Drouin
- Université de Nantes, Inserm, Centre de Recherche en Transplantation et Immunologie, UMR 1064, ITUN, Nantes, France.,OSE Immunotherapeutics, Nantes, France
| | - Javier Saenz
- Université de Nantes, Inserm, Centre de Recherche en Transplantation et Immunologie, UMR 1064, ITUN, Nantes, France
| | - Elise Chiffoleau
- Université de Nantes, Inserm, Centre de Recherche en Transplantation et Immunologie, UMR 1064, ITUN, Nantes, France
| |
Collapse
|
82
|
Cueto FJ, Del Fresno C, Sancho D. DNGR-1, a Dendritic Cell-Specific Sensor of Tissue Damage That Dually Modulates Immunity and Inflammation. Front Immunol 2020; 10:3146. [PMID: 32117205 PMCID: PMC7018937 DOI: 10.3389/fimmu.2019.03146] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2019] [Accepted: 12/27/2019] [Indexed: 11/13/2022] Open
Abstract
DNGR-1 (encoded by CLEC9A) is a C-type lectin receptor (CLR) with an expression profile that is mainly restricted to type 1 conventional dendritic cells (cDC1s) both in mice and humans. This delimited expression pattern makes it appropriate for defining a cDC1 signature and for therapeutic targeting of this population, promoting immunity in mouse models. Functionally, DNGR-1 binds F-actin, which is confined within the intracellular space in healthy cells, but is exposed when plasma membrane integrity is compromised, as happens in necrosis. Upon F-actin binding, DNGR-1 signals through SYK and mediates cross-presentation of dead cell-associated antigens. Cross-presentation to CD8+ T cells promoted by DNGR-1 during viral infections is key for cross-priming tissue-resident memory precursors in the lymph node. However, in contrast to other closely related CLRs such as Dectin-1, DNGR-1 does not activate NFκB. Instead, recent findings show that DNGR-1 can activate SHP-1 to limit inflammation triggered by heterologous receptors, which results in reduced production of inflammatory chemokines and neutrophil recruitment into damaged tissues in both sterile and infectious processes. Hence, DNGR-1 reduces immunopathology associated with tissue damage, promoting disease tolerance to safeguard tissue integrity. How DNGR-1 signals are conditioned by the microenvironment and the detailed molecular mechanisms underlying DNGR-1 function have not been elucidated. Here, we review the expression pattern and structural features of DNGR-1, and the biological relevance of the detection of tissue damage through this CLR.
Collapse
Affiliation(s)
- Francisco J Cueto
- Laboratory of Immunobiology, Centro Nacional de Investigaciones Cardiovasculares, Madrid, Spain
| | - Carlos Del Fresno
- Laboratory of Immunobiology, Centro Nacional de Investigaciones Cardiovasculares, Madrid, Spain
| | - David Sancho
- Laboratory of Immunobiology, Centro Nacional de Investigaciones Cardiovasculares, Madrid, Spain
| |
Collapse
|
83
|
Böhner AMC, Viehmann SF, Kurts C. Unraveling the Complexity of the Renal Mononuclear Phagocyte System by Genetic Cell Lineage Tracing. J Am Soc Nephrol 2020; 31:233-235. [PMID: 31949050 DOI: 10.1681/asn.2019121295] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Affiliation(s)
| | - Susanne F Viehmann
- Institute of Experimental Immunology, University Hospital Bonn, Bonn, Germany
| | - Christian Kurts
- Institute of Experimental Immunology, University Hospital Bonn, Bonn, Germany
| |
Collapse
|
84
|
Laban KG, Rijken R, Hiddingh S, Mertens JS, van der Veen RLP, Eenhorst CAE, Pandit A, Radstake TRDJ, de Boer JH, Kalmann R, Kuiper JJW. cDC2 and plasmacytoid dendritic cells diminish from tissues of patients with non-Hodgkin orbital lymphoma and idiopathic orbital inflammation. Eur J Immunol 2020; 50:548-557. [PMID: 31841217 PMCID: PMC7187234 DOI: 10.1002/eji.201948370] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2019] [Revised: 10/21/2019] [Accepted: 12/10/2019] [Indexed: 12/21/2022]
Abstract
Non-Hodgkin orbital lymphoma (NHOL) and idiopathic orbital inflammation (IOI) are common orbital conditions with largely unknown pathophysiology. To investigate the immune cell composition of these diseases, we performed standardized 29 parameter flow cytometry phenotyping in peripheral blood mononuclear cells of 18 NHOL patients, 21 IOI patients, and 41 unaffected controls. Automatic gating by FlowSOM revealed decreased abundance of meta-clusters containing dendritic cells in patients, which we confirmed by manual gating. A decreased percentage of (HLA-DR+ CD303+ CD123+ ) plasmacytoid dendritic cells (pDC) in the circulation of IOI patients and decreased (HLA-DR+ CD11c+ CD1c+ ) conventional dendritic cells (cDC) type-2 for IOI patients were replicated in an independent cohort of patients and controls. Meta-analysis of both cohorts demonstrated that pDCs are also decreased in blood of NHOL patients and highlighted that the decrease in blood cDC type-2 was specific for IOI patients compared to NHOL or controls. Deconvolution-based estimation of immune cells in transcriptomic data of 48 orbital biopsies revealed a decrease in the abundance of pDC and cDC populations within the orbital microenvironment of IOI patients. Collectively, these data suggest a previously underappreciated role for dendritic cells in orbital disorders.
Collapse
Affiliation(s)
- Kamil G Laban
- Ophthalmo-Immunology Unit, University Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands.,Department of Ophthalmology, University Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands.,Laboratory of Translational Immunology, University Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands
| | - Rianne Rijken
- Laboratory of Translational Immunology, University Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands.,Department of Rheumatology & Clinical Immunology, University Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands
| | - Sanne Hiddingh
- Ophthalmo-Immunology Unit, University Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands.,Department of Ophthalmology, University Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands.,Laboratory of Translational Immunology, University Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands
| | - Jorre S Mertens
- Laboratory of Translational Immunology, University Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands.,Department of Rheumatology & Clinical Immunology, University Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands.,Department of Dermatology, Radboud University Medical Centre, Radboud University, Nijmegen, The Netherlands
| | - Rob L P van der Veen
- Department of Ophthalmology, University Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands
| | - Christine A E Eenhorst
- Department of Ophthalmology, University Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands
| | - Aridaman Pandit
- Laboratory of Translational Immunology, University Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands.,Department of Rheumatology & Clinical Immunology, University Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands
| | - Timothy R D J Radstake
- Ophthalmo-Immunology Unit, University Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands.,Laboratory of Translational Immunology, University Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands.,Department of Rheumatology & Clinical Immunology, University Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands
| | - Joke H de Boer
- Ophthalmo-Immunology Unit, University Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands.,Department of Ophthalmology, University Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands
| | - Rachel Kalmann
- Ophthalmo-Immunology Unit, University Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands.,Department of Ophthalmology, University Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands
| | - Jonas J W Kuiper
- Ophthalmo-Immunology Unit, University Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands.,Department of Ophthalmology, University Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands.,Laboratory of Translational Immunology, University Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands
| |
Collapse
|
85
|
Salei N, Rambichler S, Salvermoser J, Papaioannou NE, Schuchert R, Pakalniškytė D, Li N, Marschner JA, Lichtnekert J, Stremmel C, Cernilogar FM, Salvermoser M, Walzog B, Straub T, Schotta G, Anders HJ, Schulz C, Schraml BU. The Kidney Contains Ontogenetically Distinct Dendritic Cell and Macrophage Subtypes throughout Development That Differ in Their Inflammatory Properties. J Am Soc Nephrol 2020; 31:257-278. [PMID: 31932472 DOI: 10.1681/asn.2019040419] [Citation(s) in RCA: 60] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2019] [Accepted: 10/20/2019] [Indexed: 12/16/2022] Open
Abstract
BACKGROUND Mononuclear phagocytes (MPs), including macrophages, monocytes, and dendritic cells (DCs), are phagocytic cells with important roles in immunity. The developmental origin of kidney DCs has been highly debated because of the large phenotypic overlap between macrophages and DCs in this tissue. METHODS We used fate mapping, RNA sequencing, flow cytometry, confocal microscopy, and histo-cytometry to assess the origin and phenotypic and functional properties of renal DCs in healthy kidney and of DCs after cisplatin and ischemia reperfusion-induced kidney injury. RESULTS Adult kidney contains at least four subsets of MPs with prominent Clec9a-expression history indicating a DC origin. We demonstrate that these populations are phenotypically, functionally, and transcriptionally distinct from each other. We also show these kidney MPs exhibit unique age-dependent developmental heterogeneity. Kidneys from newborn mice contain a prominent population of embryonic-derived MHCIInegF4/80hiCD11blow macrophages that express T cell Ig and mucin domain containing 4 (TIM-4) and MER receptor tyrosine kinase (MERTK). These macrophages are replaced within a few weeks after birth by phenotypically similar cells that express MHCII but lack TIM-4 and MERTK. MHCII+F4/80hi cells exhibit prominent Clec9a-expression history in adulthood but not early life, indicating additional age-dependent developmental heterogeneity. In AKI, MHCIInegF4/80hi cells reappear in adult kidneys as a result of MHCII downregulation by resident MHCII+F4/80hi cells, possibly in response to prostaglandin E2 (PGE2). RNA sequencing further suggests MHCII+F4/80hi cells help coordinate the recruitment of inflammatory cells during renal injury. CONCLUSIONS Distinct developmental programs contribute to renal DC and macrophage populations throughout life, which could have important implications for therapies targeting these cells.
Collapse
Affiliation(s)
- Natallia Salei
- Walter Brendel Centre of Experimental Medicine, University Hospital Munich.,Institute for Cardiovascular Physiology and Pathophysiology
| | - Stephan Rambichler
- Walter Brendel Centre of Experimental Medicine, University Hospital Munich.,Institute for Cardiovascular Physiology and Pathophysiology
| | - Johanna Salvermoser
- Walter Brendel Centre of Experimental Medicine, University Hospital Munich.,Institute for Cardiovascular Physiology and Pathophysiology
| | - Nikos E Papaioannou
- Walter Brendel Centre of Experimental Medicine, University Hospital Munich.,Institute for Cardiovascular Physiology and Pathophysiology
| | - Ronja Schuchert
- Medical Clinic and Polyclinic I and.,DZHK (Deutsches Zentrum für Herz-Kreislaufforschung [German Center for Cardiovascular Research]), Partner Site Munich Heart Alliance, Munich, Germany; and
| | - Dalia Pakalniškytė
- Walter Brendel Centre of Experimental Medicine, University Hospital Munich.,Institute for Cardiovascular Physiology and Pathophysiology
| | - Na Li
- Division of Nephrology, The Seventh Affiliated Hospital, Sun Yat-sen University, Shen Zhen, China.,Division of Nephrology, Medical Clinic and Polyclinic IV, University Hospital Munich, Ludwig Maximilian University of Munich, Munich, Germany
| | - Julian A Marschner
- Division of Nephrology, Medical Clinic and Polyclinic IV, University Hospital Munich, Ludwig Maximilian University of Munich, Munich, Germany
| | - Julia Lichtnekert
- Division of Nephrology, Medical Clinic and Polyclinic IV, University Hospital Munich, Ludwig Maximilian University of Munich, Munich, Germany
| | - Christopher Stremmel
- Medical Clinic and Polyclinic I and.,DZHK (Deutsches Zentrum für Herz-Kreislaufforschung [German Center for Cardiovascular Research]), Partner Site Munich Heart Alliance, Munich, Germany; and
| | | | - Melanie Salvermoser
- Walter Brendel Centre of Experimental Medicine, University Hospital Munich.,Institute for Cardiovascular Physiology and Pathophysiology
| | - Barbara Walzog
- Walter Brendel Centre of Experimental Medicine, University Hospital Munich.,Institute for Cardiovascular Physiology and Pathophysiology
| | | | - Gunnar Schotta
- Division of Molecular Biology.,Center for Integrated Protein Science Munich, Biomedical Center, Faculty of Medicine, Ludwig Maximilian University of Munich, Martinsried, Germany
| | - Hans-Joachim Anders
- Division of Nephrology, Medical Clinic and Polyclinic IV, University Hospital Munich, Ludwig Maximilian University of Munich, Munich, Germany
| | - Christian Schulz
- Medical Clinic and Polyclinic I and.,DZHK (Deutsches Zentrum für Herz-Kreislaufforschung [German Center for Cardiovascular Research]), Partner Site Munich Heart Alliance, Munich, Germany; and
| | - Barbara U Schraml
- Walter Brendel Centre of Experimental Medicine, University Hospital Munich, .,Institute for Cardiovascular Physiology and Pathophysiology
| |
Collapse
|
86
|
Abstract
C-type lectin receptors (CLRs) are a family of transmembrane proteins having at least one C-type lectin-like domain (CTLD) on the cell surface and either a short intracellular signaling tail or a transmembrane domain that facilitates interaction with a second protein, often the Fc receptor common gamma chain (FcRγ), that mediates signaling. Many CLRs directly recognize microbial cell walls and influence innate immunity by activating inflammatory and antimicrobial responses in phagocytes. In this review, we examine the contributions of certain CLRs to activation and regulation of phagocytosis in cells such as macrophages, dendritic cells and neutrophils.
Collapse
|
87
|
Pires CF, Rosa FF, Kurochkin I, Pereira CF. Understanding and Modulating Immunity With Cell Reprogramming. Front Immunol 2019; 10:2809. [PMID: 31921109 PMCID: PMC6917620 DOI: 10.3389/fimmu.2019.02809] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2019] [Accepted: 11/15/2019] [Indexed: 12/30/2022] Open
Abstract
Cell reprogramming concepts have been classically developed in the fields of developmental and stem cell biology and are currently being explored for regenerative medicine, given its potential to generate desired cell types for replacement therapy. Cell fate can be experimentally reversed or modified by enforced expression of lineage specific transcription factors leading to pluripotency or attainment of another somatic cell type identity. The possibility to reprogram fibroblasts into induced dendritic cells (DC) competent for antigen presentation creates a paradigm shift for understanding and modulating the immune system with direct cell reprogramming. PU.1, IRF8, and BATF3 were identified as sufficient and necessary to impose DC fate in unrelated cell types, taking advantage of Clec9a, a C-type lectin receptor with restricted expression in conventional DC type 1. The identification of such minimal gene regulatory networks helps to elucidate the molecular mechanisms governing development and lineage heterogeneity along the hematopoietic hierarchy. Furthermore, the generation of patient-tailored reprogrammed immune cells provides new and exciting tools for the expanding field of cancer immunotherapy. Here, we summarize cell reprogramming concepts and experimental approaches, review current knowledge at the intersection of cell reprogramming with hematopoiesis, and propose how cell fate engineering can be merged to immunology, opening new opportunities to understand the immune system in health and disease.
Collapse
Affiliation(s)
- Cristiana F. Pires
- Cell Reprogramming in Hematopoiesis and Immunity Laboratory, Lund Stem Cell Center, Molecular Medicine and Gene Therapy, Lund University, Lund, Sweden
- Wallenberg Center for Molecular Medicine, Lund University, Lund, Sweden
- Center for Neuroscience and Cell Biology, University of Coimbra, Coimbra, Portugal
| | - Fábio F. Rosa
- Cell Reprogramming in Hematopoiesis and Immunity Laboratory, Lund Stem Cell Center, Molecular Medicine and Gene Therapy, Lund University, Lund, Sweden
- Wallenberg Center for Molecular Medicine, Lund University, Lund, Sweden
- Center for Neuroscience and Cell Biology, University of Coimbra, Coimbra, Portugal
| | - Ilia Kurochkin
- Center for Neurobiology and Brain Restoration, Skolkovo Institute of Science and Technology, Moscow, Russia
| | - Carlos-Filipe Pereira
- Cell Reprogramming in Hematopoiesis and Immunity Laboratory, Lund Stem Cell Center, Molecular Medicine and Gene Therapy, Lund University, Lund, Sweden
- Wallenberg Center for Molecular Medicine, Lund University, Lund, Sweden
- Center for Neuroscience and Cell Biology, University of Coimbra, Coimbra, Portugal
| |
Collapse
|
88
|
Netravali IA, Cariappa A, Yates K, Haining WN, Bertocchi A, Allard-Chamard H, Rosenberg I, Pillai S. 9-O-acetyl sialic acid levels identify committed progenitors of plasmacytoid dendritic cells. Glycobiology 2019; 29:861-875. [PMID: 31411667 DOI: 10.1093/glycob/cwz062] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2019] [Revised: 07/24/2019] [Accepted: 08/01/2019] [Indexed: 11/12/2022] Open
Abstract
The origins of plasmacytoid dendritic cells (pDCs) have long been controversial and progenitors exclusively committed to this lineage have not been described. We show here that the fate of hematopoietic progenitors is determined in part by their surface levels of 9-O-acetyl sialic acid. Pro-pDCs were identified as lineage negative 9-O-acetyl sialic acid low progenitors that lack myeloid and lymphoid potential but differentiate into pre-pDCs. The latter cells are also lineage negative, 9-O-acetyl sialic acid low cells but are exclusively committed to the pDC lineage. Levels of 9-O-acetyl sialic acid provide a distinct way to define progenitors and thus facilitate the study of hematopoietic differentiation.
Collapse
Affiliation(s)
- Ilka A Netravali
- Ragon Institute of MGH, MIT and Harvard, Cambridge MA 02139 and The MGH Cancer Center, Massachusetts General Hospital, Harvard Medical School, Charlestown, MA 02129, USA
| | - Annaiah Cariappa
- Ragon Institute of MGH, MIT and Harvard, Cambridge MA 02139 and The MGH Cancer Center, Massachusetts General Hospital, Harvard Medical School, Charlestown, MA 02129, USA
| | - Kathleen Yates
- Dana-Farber Cancer Institute, Pediatric Oncology, Harvard Medical School, Boston, MA 02115, USA
| | - W Nicholas Haining
- Dana-Farber Cancer Institute, Pediatric Oncology, Harvard Medical School, Boston, MA 02115, USA
| | - Alice Bertocchi
- Ragon Institute of MGH, MIT and Harvard, Cambridge MA 02139 and The MGH Cancer Center, Massachusetts General Hospital, Harvard Medical School, Charlestown, MA 02129, USA
| | - Hugues Allard-Chamard
- Ragon Institute of MGH, MIT and Harvard, Cambridge MA 02139 and The MGH Cancer Center, Massachusetts General Hospital, Harvard Medical School, Charlestown, MA 02129, USA.,Division of Rheumatology, Faculté de Médecine et des Sciences de la Santé de l', Université de Sherbrooke et Centre de Recherche Clinique Étienne-Le Bel, Sherbrooke, Québec, Canada, J1K 2R1
| | - Ian Rosenberg
- Ragon Institute of MGH, MIT and Harvard, Cambridge MA 02139 and The MGH Cancer Center, Massachusetts General Hospital, Harvard Medical School, Charlestown, MA 02129, USA
| | - Shiv Pillai
- Ragon Institute of MGH, MIT and Harvard, Cambridge MA 02139 and The MGH Cancer Center, Massachusetts General Hospital, Harvard Medical School, Charlestown, MA 02129, USA
| |
Collapse
|
89
|
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
|
90
|
Clement M, Raffort J, Lareyre F, Tsiantoulas D, Newland S, Lu Y, Masters L, Harrison J, Saveljeva S, Ma MKL, Ozsvar-Kozma M, Lam BYH, Yeo GSH, Binder CJ, Kaser A, Mallat Z. Impaired Autophagy in CD11b + Dendritic Cells Expands CD4 + Regulatory T Cells and Limits Atherosclerosis in Mice. Circ Res 2019; 125:1019-1034. [PMID: 31610723 PMCID: PMC6844650 DOI: 10.1161/circresaha.119.315248] [Citation(s) in RCA: 33] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Supplemental Digital Content is available in the text. Atherosclerosis is a chronic inflammatory disease. Recent studies have shown that dysfunctional autophagy in endothelial cells, smooth muscle cells, and macrophages, plays a detrimental role during atherogenesis, leading to the suggestion that autophagy-stimulating approaches may provide benefit.
Collapse
Affiliation(s)
- Marc Clement
- From the Division of Cardiovascular Medicine, University of Cambridge, Cambridge, United Kingdom (M.C., J.R., F.L., D.T., S.N., Y.L., L.M., J.H., Z.M.)
| | - Juliette Raffort
- From the Division of Cardiovascular Medicine, University of Cambridge, Cambridge, United Kingdom (M.C., J.R., F.L., D.T., S.N., Y.L., L.M., J.H., Z.M.).,Université CÔte d'Azur, Institut National de la Santé et de la Recherche Médicale, Centre Mediterranéen de Recherche Moléculaire, University Hospital of Nice, France (J.R., F.L.)
| | - Fabien Lareyre
- From the Division of Cardiovascular Medicine, University of Cambridge, Cambridge, United Kingdom (M.C., J.R., F.L., D.T., S.N., Y.L., L.M., J.H., Z.M.).,Université CÔte d'Azur, Institut National de la Santé et de la Recherche Médicale, Centre Mediterranéen de Recherche Moléculaire, University Hospital of Nice, France (J.R., F.L.)
| | - Dimitrios Tsiantoulas
- From the Division of Cardiovascular Medicine, University of Cambridge, Cambridge, United Kingdom (M.C., J.R., F.L., D.T., S.N., Y.L., L.M., J.H., Z.M.)
| | - Stephen Newland
- From the Division of Cardiovascular Medicine, University of Cambridge, Cambridge, United Kingdom (M.C., J.R., F.L., D.T., S.N., Y.L., L.M., J.H., Z.M.)
| | - Yuning Lu
- From the Division of Cardiovascular Medicine, University of Cambridge, Cambridge, United Kingdom (M.C., J.R., F.L., D.T., S.N., Y.L., L.M., J.H., Z.M.)
| | - Leanne Masters
- From the Division of Cardiovascular Medicine, University of Cambridge, Cambridge, United Kingdom (M.C., J.R., F.L., D.T., S.N., Y.L., L.M., J.H., Z.M.)
| | - James Harrison
- From the Division of Cardiovascular Medicine, University of Cambridge, Cambridge, United Kingdom (M.C., J.R., F.L., D.T., S.N., Y.L., L.M., J.H., Z.M.)
| | - Svetlana Saveljeva
- Department of Gastroenterology and Hepatology, University of Cambridge, United Kingdom (S.S., A.K.)
| | - Marcella K L Ma
- MRC Metabolic Diseases Unit, University of Cambridge Metabolic Research Laboratories, Wellcome Trust-MRC Institute of Metabolic Science, Genomics and Transcriptomics Core, Addenbrooke's Hospital, Cambridge, United Kingdom (M.K.L.M., B.Y.H.L., G.S.H.Y.)
| | - Maria Ozsvar-Kozma
- Department of Laboratory Medicine, Medical University of Vienna and Center for Molecular Medicine (CeMM) of the Austrian Academy of Sciences Vienna, Austria (M.O.-K., C.J.B)
| | - Brian Y H Lam
- MRC Metabolic Diseases Unit, University of Cambridge Metabolic Research Laboratories, Wellcome Trust-MRC Institute of Metabolic Science, Genomics and Transcriptomics Core, Addenbrooke's Hospital, Cambridge, United Kingdom (M.K.L.M., B.Y.H.L., G.S.H.Y.)
| | - Giles S H Yeo
- MRC Metabolic Diseases Unit, University of Cambridge Metabolic Research Laboratories, Wellcome Trust-MRC Institute of Metabolic Science, Genomics and Transcriptomics Core, Addenbrooke's Hospital, Cambridge, United Kingdom (M.K.L.M., B.Y.H.L., G.S.H.Y.)
| | - Christoph J Binder
- Department of Laboratory Medicine, Medical University of Vienna and Center for Molecular Medicine (CeMM) of the Austrian Academy of Sciences Vienna, Austria (M.O.-K., C.J.B)
| | - Arthur Kaser
- Department of Gastroenterology and Hepatology, University of Cambridge, United Kingdom (S.S., A.K.)
| | - Ziad Mallat
- From the Division of Cardiovascular Medicine, University of Cambridge, Cambridge, United Kingdom (M.C., J.R., F.L., D.T., S.N., Y.L., L.M., J.H., Z.M.).,Institut National de la Santé et de la Recherche Médicale, Paris Cardiovascular Research Center, France (Z.M.)
| |
Collapse
|
91
|
Yona S, Mildner A. Good things come in threes. Sci Immunol 2019; 3:3/30/eaav5545. [PMID: 30530728 DOI: 10.1126/sciimmunol.aav5545] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2018] [Accepted: 11/02/2018] [Indexed: 01/20/2023]
Abstract
Ectopic expression of PU.1, IRF8, and BATF3 reprograms mouse and human fibroblasts into dendritic cells. See related Research Article by Rosa et al.
Collapse
Affiliation(s)
- Simon Yona
- Division of Medicine, University College London, London, England, UK
| | | |
Collapse
|
92
|
The impact of endoplasmic reticulum stress responses in dendritic cell immunobiology. INTERNATIONAL REVIEW OF CELL AND MOLECULAR BIOLOGY 2019; 349:153-176. [PMID: 31759430 DOI: 10.1016/bs.ircmb.2019.08.004] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Dendritic cells (DCs) are critical for bridging innate and adaptive immunity. They do so by presenting antigens to T cells, and by expressing diverse molecules that further promote T cell activation, differentiation and memory formation. During this process, intracellular and extracellular factors can perturb the protein-folding capacity of endoplasmic reticulum (ER) and induce a cellular state of "ER stress," which is controlled and resolved by the unfolded protein response (UPR). Interestingly, various studies have shown that DCs can activate UPR-related pathways even in the absence of global ER stress, and that this process can modulate their normal activity. In other settings, such as cancer, adverse microenvironmental conditions have been demonstrated to evoke severe ER stress and persistent activation of the UPR in tumor-infiltrating DCs. This process disrupts their metabolism and local antigen-presenting capacity, hence impeding the initiation and maintenance of anti-cancer immunity. Here, we review recent findings on how canonical and non-canonical UPR activation impacts DC immunobiology at the steady-state, upon activation via pattern recognition receptors, and under diverse pathological conditions. We also discuss the potential therapeutic implications that targeting the UPR in DCs may have in the context of cancer and in other pathologies such as graft-versus-host disease.
Collapse
|
93
|
Balan S, Saxena M, Bhardwaj N. Dendritic cell subsets and locations. INTERNATIONAL REVIEW OF CELL AND MOLECULAR BIOLOGY 2019; 348:1-68. [PMID: 31810551 DOI: 10.1016/bs.ircmb.2019.07.004] [Citation(s) in RCA: 188] [Impact Index Per Article: 37.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Dendritic cells (DCs) are a unique class of immune cells that act as a bridge between innate and adaptive immunity. The discovery of DCs by Cohen and Steinman in 1973 laid the foundation for DC biology, and the advances in the field identified different versions of DCs with unique properties and functions. DCs originate from hematopoietic stem cells, and their differentiation is modulated by Flt3L. They are professional antigen-presenting cells that patrol the environmental interphase, sites of infection, or infiltrate pathological tissues looking for antigens that can be used to activate effector cells. DCs are critical for the initiation of the cellular and humoral immune response and protection from infectious diseases or tumors. DCs can take up antigens using specialized surface receptors such as endocytosis receptors, phagocytosis receptors, and C type lectin receptors. Moreover, DCs are equipped with an array of extracellular and intracellular pattern recognition receptors for sensing different danger signals. Upon sensing the danger signals, DCs get activated, upregulate costimulatory molecules, produce various cytokines and chemokines, take up antigen and process it and migrate to lymph nodes where they present antigens to both CD8 and CD4 T cells. DCs are classified into different subsets based on an integrated approach considering their surface phenotype, expression of unique and conserved molecules, ontogeny, and functions. They can be broadly classified as conventional DCs consisting of two subsets (DC1 and DC2), plasmacytoid DCs, inflammatory DCs, and Langerhans cells.
Collapse
Affiliation(s)
- Sreekumar Balan
- The Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, United States.
| | - Mansi Saxena
- The Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, United States
| | - Nina Bhardwaj
- The Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, United States; Parker Institute for Cancer Immunotherapy, San Francisco, CA, United States
| |
Collapse
|
94
|
Abstract
The C-type lectins are a superfamily of proteins that recognize a broad repertoire of ligands and that regulate a diverse range of physiological functions. Most research attention has focused on the ability of C-type lectins to function in innate and adaptive antimicrobial immune responses, but these proteins are increasingly being recognized to have a major role in autoimmune diseases and to contribute to many other aspects of multicellular existence. Defects in these molecules lead to developmental and physiological abnormalities, as well as altered susceptibility to infectious and non-infectious diseases. In this Review, we present an overview of the roles of C-type lectins in immunity and homeostasis, with an emphasis on the most exciting recent discoveries.
Collapse
|
95
|
Das T, Bergen IM, Koudstaal T, van Hulst JA, van Loo G, Boonstra A, Vanwolleghem T, Leung PS, Gershwin ME, Hendriks RW, Kool M. DNGR1-mediated deletion of A20/Tnfaip3 in dendritic cells alters T and B-cell homeostasis and promotes autoimmune liver pathology. J Autoimmun 2019; 102:167-178. [DOI: 10.1016/j.jaut.2019.05.007] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2019] [Revised: 04/27/2019] [Accepted: 05/05/2019] [Indexed: 02/06/2023]
|
96
|
The orphan nuclear receptor NR4A3 controls the differentiation of monocyte-derived dendritic cells following microbial stimulation. Proc Natl Acad Sci U S A 2019; 116:15150-15159. [PMID: 31285338 DOI: 10.1073/pnas.1821296116] [Citation(s) in RCA: 38] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023] Open
Abstract
In response to microbial stimulation, monocytes can differentiate into macrophages or monocyte-derived dendritic cells (MoDCs) but the molecular requirements guiding these possible fates are poorly understood. In addition, the physiological importance of MoDCs in the host cellular and immune responses to microbes remains elusive. Here, we demonstrate that the nuclear orphan receptor NR4A3 is required for the proper differentiation of MoDCs but not for other types of DCs. Indeed, the generation of DC-SIGN+ MoDCs in response to LPS was severely impaired in Nr4a3 -/- mice, which resulted in the inability to mount optimal CD8+ T cell responses to gram-negative bacteria. Transcriptomic analyses revealed that NR4A3 is required to skew monocyte differentiation toward MoDCs, at the expense of macrophages, and allows the acquisition of migratory characteristics required for MoDC function. Altogether, our data identify that the NR4A3 transcription factor is required to guide the fate of monocytes toward MoDCs.
Collapse
|
97
|
Lin DS, Kan A, Gao J, Crampin EJ, Hodgkin PD, Naik SH. DiSNE Movie Visualization and Assessment of Clonal Kinetics Reveal Multiple Trajectories of Dendritic Cell Development. Cell Rep 2019. [PMID: 29514085 DOI: 10.1016/j.celrep.2018.02.046] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
A thorough understanding of cellular development is incumbent on assessing the complexities of fate and kinetics of individual clones within a population. Here, we develop a system for robust periodical assessment of lineage outputs of thousands of transient clones and establishment of bona fide cellular trajectories. We appraise the development of dendritic cells (DCs) in fms-like tyrosine kinase 3 ligand culture from barcode-labeled hematopoietic stem and progenitor cells (HSPCs) by serially measuring barcode signatures and visualize these multidimensional data using developmental interpolated t-distributed stochastic neighborhood embedding (DiSNE) time-lapse movies. We identify multiple cellular trajectories of DC development that are characterized by distinct fate bias and expansion kinetics and determine that these are intrinsically programmed. We demonstrate that conventional DC and plasmacytoid DC trajectories are largely separated already at the HSPC stage. This framework allows systematic evaluation of clonal dynamics and can be applied to other steady-state or perturbed developmental systems.
Collapse
Affiliation(s)
- Dawn S Lin
- Molecular Medicine Division, Walter and Eliza Hall Institute, Parkville, VIC 3052, Australia; Immunology Division, Walter and Eliza Hall Institute, Parkville, VIC 3052, Australia; Faculty of Medicine, Dentistry & Health Sciences, University of Melbourne, Parkville, VIC 3010, Australia
| | - Andrey Kan
- Immunology Division, Walter and Eliza Hall Institute, Parkville, VIC 3052, Australia; Faculty of Medicine, Dentistry & Health Sciences, University of Melbourne, Parkville, VIC 3010, Australia
| | - Jerry Gao
- Molecular Medicine Division, Walter and Eliza Hall Institute, Parkville, VIC 3052, Australia
| | - Edmund J Crampin
- Systems Biology Laboratory, University of Melbourne, Parkville, VIC 3010, Australia; Centre for Systems Genomics, University of Melbourne, Parkville, VIC 3010, Australia; ARC Centre of Excellence in Convergent Bio-Nano Science and Technology, Melbourne School of Engineering, University of Melbourne, Parkville, VIC 3010, Australia
| | - Philip D Hodgkin
- Immunology Division, Walter and Eliza Hall Institute, Parkville, VIC 3052, Australia; Faculty of Medicine, Dentistry & Health Sciences, University of Melbourne, Parkville, VIC 3010, Australia
| | - Shalin H Naik
- Molecular Medicine Division, Walter and Eliza Hall Institute, Parkville, VIC 3052, Australia; Immunology Division, Walter and Eliza Hall Institute, Parkville, VIC 3052, Australia; Faculty of Medicine, Dentistry & Health Sciences, University of Melbourne, Parkville, VIC 3010, Australia.
| |
Collapse
|
98
|
Musumeci A, Lutz K, Winheim E, Krug AB. What Makes a pDC: Recent Advances in Understanding Plasmacytoid DC Development and Heterogeneity. Front Immunol 2019; 10:1222. [PMID: 31191558 PMCID: PMC6548821 DOI: 10.3389/fimmu.2019.01222] [Citation(s) in RCA: 89] [Impact Index Per Article: 17.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2018] [Accepted: 05/13/2019] [Indexed: 12/16/2022] Open
Abstract
Dendritic cells (DCs) are professional antigen presenting cells (APCs) that originate in the bone marrow and are continuously replenished from hematopoietic progenitor cells. Conventional DCs (cDCs) and plasmacytoid DCs (pDCs) are distinguished by morphology and function, and can be easily discriminated by surface marker expression, both in mouse and man. Classification of DCs based on their ontology takes into account their origin as well as their requirements for transcription factor (TF) expression. cDCs and pDCs of myeloid origin differentiate from a common DC progenitor (CDP) through committed pre-DC stages. pDCs have also been shown to originate from a lymphoid progenitor derived IL-7R+ FLT3+ precursor population containing cells with pDC or B cell potential. Technological advancements in recent years have allowed unprecedented resolution in the analysis of cell states, down to the single cell level, providing valuable information on the commitment, and dynamics of differentiation of all DC subsets. However, the heterogeneity and functional diversification of pDCs still raises the question whether different ontogenies generate restricted pDC subsets, or fully differentiated pDCs retain plasticity in response to challenges. The emergence of novel techniques for the integration of high-resolution data in individual cells promises interesting discoveries regarding DC development and plasticity in the near future.
Collapse
Affiliation(s)
- Andrea Musumeci
- Institute for Immunology, Biomedical Center, Ludwig-Maximilian-University, Munich, Germany
| | - Konstantin Lutz
- Institute for Immunology, Biomedical Center, Ludwig-Maximilian-University, Munich, Germany
| | - Elena Winheim
- Institute for Immunology, Biomedical Center, Ludwig-Maximilian-University, Munich, Germany
| | - Anne Barbara Krug
- Institute for Immunology, Biomedical Center, Ludwig-Maximilian-University, Munich, Germany
| |
Collapse
|
99
|
Tintelnot J, Ufer F, Engler JB, Winkler H, Lücke K, Mittrücker HW, Friese MA. Arc/Arg3.1 defines dendritic cells and Langerhans cells with superior migratory ability independent of phenotype and ontogeny in mice. Eur J Immunol 2019; 49:724-736. [PMID: 30786014 DOI: 10.1002/eji.201847797] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2018] [Revised: 12/14/2018] [Accepted: 02/18/2019] [Indexed: 01/22/2023]
Abstract
The key function of migratory dendritic cells (migDCs) is to take up antigens in peripheral tissues and migrate to draining lymph nodes (dLN) to initiate immune responses. Recently, we discovered that in the mouse immune system activity-regulated cytoskeleton associated protein/activity-regulated gene 3.1 (Arc/Arg3.1) is exclusively expressed by migDCs and is a central driver of fast inflammatory migration. However, the frequency of Arc/Arg3.1-expressing cells in different migDC subsets and Langerhans cells (LCs), their phylogenetic origin, transcription factor dependency, and functional role remain unclear. Here, we found that Arc/Arg3.1+ migDCs derived from common DC precursors and radio-resistant LCs. We detected Arc/Arg3.1+ migDCs in varying frequencies within each migDC subset and LCs. Consistently, they showed superiority in inflammatory migration. Arc/Arg3.1 expression was independent of the transcription factors Irf4 or Batf3 in vivo. In intradermal Staphylococcus aureus infection that relies on inflammatory antigen transport, Arc/Arg3.1 deletion reduced T-cell responses. By contrast, Arc/Arg3.1 deficiency did not hamper the immune response to systemic Listeria monocytogenes infection, which does not require antigen transport. Thus, Arc/Arg3.1 expression is independent of ontogeny and phenotype and although it is restricted to a small fraction within each migDC subset and LCs, Arc/Arg3.1+ migDCs are important to facilitate infectious migration.
Collapse
Affiliation(s)
- Joseph Tintelnot
- Institut für Neuroimmunologie und Multiple Sklerose, Universitätsklinikum Hamburg-Eppendorf, Hamburg, Germany
| | - Friederike Ufer
- Institut für Neuroimmunologie und Multiple Sklerose, Universitätsklinikum Hamburg-Eppendorf, Hamburg, Germany
| | - Jan Broder Engler
- Institut für Neuroimmunologie und Multiple Sklerose, Universitätsklinikum Hamburg-Eppendorf, Hamburg, Germany
| | - Hana Winkler
- Institut für Neuroimmunologie und Multiple Sklerose, Universitätsklinikum Hamburg-Eppendorf, Hamburg, Germany
| | - Karsten Lücke
- Institut für Immunologie, Universitätsklinikum Hamburg-Eppendorf, Hamburg, Germany
| | | | - Manuel A Friese
- Institut für Neuroimmunologie und Multiple Sklerose, Universitätsklinikum Hamburg-Eppendorf, Hamburg, Germany
| |
Collapse
|
100
|
Arnold IC, Zhang X, Urban S, Artola-Borán M, Manz MG, Ottemann KM, Müller A. NLRP3 Controls the Development of Gastrointestinal CD11b + Dendritic Cells in the Steady State and during Chronic Bacterial Infection. Cell Rep 2019; 21:3860-3872. [PMID: 29281833 DOI: 10.1016/j.celrep.2017.12.015] [Citation(s) in RCA: 46] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2017] [Revised: 10/24/2017] [Accepted: 12/01/2017] [Indexed: 12/14/2022] Open
Abstract
The gastric lamina propria is largely uncharted immunological territory. Here we describe the evolution and composition of the gastric, small intestinal, and colonic lamina propria mononuclear phagocyte system during the steady state and infection with the gastric pathogen Helicobacter pylori. We show that monocytes, CX3CR1hi macrophages, and CD11b+ dendritic cells are recruited to the infected stomach in a CCR2-dependent manner. All three populations, but not BATF3-dependent CD103+ DCs, sample red fluorescent protein (RFP)+Helicobacter pylori (H. pylori). Mice reconstituted with human hematopoietic stem cells recapitulate several features of the myeloid cell-H. pylori interaction. The differentiation in and/or recruitment to gastrointestinal, lung, and lymphoid tissues of CD11b+ DCs requires NLRP3, but not apoptosis-associated speck-like protein containing a carboxy-terminal CARD (ASC) or caspase-1, during steady-state and chronic infection. NLRP3-/- mice fail to generate Treg responses to H. pylori and control the infection more effectively than wild-type mice. The results demonstrate a non-canonical inflammasome-independent function of NLRP3 in DC development and immune regulation.
Collapse
Affiliation(s)
- Isabelle C Arnold
- Institute of Molecular Cancer Research , University of Zürich, 8057 Zürich, Switzerland.
| | - Xiaozhou Zhang
- Institute of Molecular Cancer Research , University of Zürich, 8057 Zürich, Switzerland
| | - Sabine Urban
- Institute of Molecular Cancer Research , University of Zürich, 8057 Zürich, Switzerland
| | - Mariela Artola-Borán
- Institute of Molecular Cancer Research , University of Zürich, 8057 Zürich, Switzerland
| | - Markus G Manz
- Department of Hematology, University of Zürich, 8057 Zürich, Switzerland
| | - Karen M Ottemann
- Department of Microbiology and Environmental Toxicology, University of California, Santa Cruz, Santa Cruz, CA, USA
| | - Anne Müller
- Institute of Molecular Cancer Research , University of Zürich, 8057 Zürich, Switzerland.
| |
Collapse
|