1
|
Gutierrez-Barbosa H, Medina-Moreno S, Perdomo-Celis F, Davis H, Coronel-Ruiz C, Zapata JC, Chua JV. A Comparison of Lymphoid and Myeloid Cells Derived from Human Hematopoietic Stem Cells Xenografted into NOD-Derived Mouse Strains. Microorganisms 2023; 11:1548. [PMID: 37375051 DOI: 10.3390/microorganisms11061548] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2023] [Revised: 06/07/2023] [Accepted: 06/08/2023] [Indexed: 06/29/2023] Open
Abstract
Humanized mice are an invaluable tool for investigating human diseases such as cancer, infectious diseases, and graft-versus-host disease (GvHD). However, it is crucial to understand the strengths and limitations of humanized mice and select the most appropriate model. In this study, we describe the development of the human lymphoid and myeloid lineages using a flow cytometric analysis in four humanized mouse models derived from NOD mice xenotransplanted with CD34+ fetal cord blood from a single donor. Our results showed that all murine strains sustained human immune cells within a proinflammatory environment induced by GvHD. However, the Hu-SGM3 model consistently generated higher numbers of human T cells, monocytes, dendritic cells, mast cells, and megakaryocytes, and a low number of circulating platelets showing an activated profile when compared with the other murine strains. The hu-NOG-EXL model had a similar cell development profile but a higher number of circulating platelets with an inactivated state, and the hu-NSG and hu-NCG developed low frequencies of immune cells compared with the other models. Interestingly, only the hu-SGM3 and hu-EXL models developed mast cells. In conclusion, our findings highlight the importance of selecting the appropriate humanized mouse model for specific research questions, considering the strengths and limitations of each model and the immune cell populations of interest.
Collapse
Affiliation(s)
| | - Sandra Medina-Moreno
- Institute of Human Virology, University of Maryland School of Medicine, Baltimore, MD 21201, USA
| | - Federico Perdomo-Celis
- Instituto de Genética Humana, Facultad de Medicina, Pontificia Universidad Javeriana, Bogotá 110231, Colombia
| | - Harry Davis
- Institute of Human Virology, University of Maryland School of Medicine, Baltimore, MD 21201, USA
| | - Carolina Coronel-Ruiz
- Vice-Chancellor of Research, Virology Group, Universidad El Bosque, Bogotá 110121, Colombia
| | - Juan C Zapata
- Institute of Human Virology, University of Maryland School of Medicine, Baltimore, MD 21201, USA
| | - Joel V Chua
- Institute of Human Virology, University of Maryland School of Medicine, Baltimore, MD 21201, USA
| |
Collapse
|
2
|
Rani R, Nayak M, Nayak B. Exploring the reprogramming potential of B cells and comprehending its clinical and therapeutic perspective. Transpl Immunol 2023; 78:101804. [PMID: 36921730 DOI: 10.1016/j.trim.2023.101804] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2022] [Revised: 02/08/2023] [Accepted: 02/21/2023] [Indexed: 03/14/2023]
Abstract
Initiating from multipotent progenitors, the lineages extrapolated from hematopoietic stem cells are determined by transcription factors specific to each of them. The commitment factors assist in the differentiation of progenitor cells into terminally differentiated cells. B lymphocytes constitute a population of cells that expresses clonally diverse cell surface immunoglobulin (Ig) receptors specific to antigenic epitopes. B cells are a significant facet of the adaptive immune system. The secreted antibodies corresponding to the B cell recognize the antigens via the B cell receptor (BCR). Following antigen recognition, the B cell is activated and thereafter undergoes clonal expansion and proliferation to become memory B cells. The essence of 'cellular reprogramming' has aided in reliably altering the cells to desired tissue type. The potential of reprogramming has been harnessed to decipher and find solutions for various genetically inherited diseases and degenerative disorders. B lymphocytes can be reprogrammed to their initial naive state from where they get differentiated into any lineage or cell type similar to a pluripotent stem cell which can be accomplished by the deletion of master regulators of the B cell lineage. B cells can be reprogrammed into pluripotent stem cells and also can undergo transdifferentiation at the midway of cell differentiation to other cell types. Mandated expression of C/EBP in specialized B cells corresponds to their fast and effective reprogramming into macrophages, reversing the cell fate of these lymphocytes and allowing them to differentiate freshly into other types of cells. The co-expression of C/EBPα and OKSM (Oct4, Sox2, Klf4, c-Myc) amplified the reprogramming efficiency of B lymphocytes. Various human somatic cells including the immune cells are compliant to reprogramming which paves a path for opportunities like autologous tissue grafts, blood transfusion, and cancer immunotherapy. The ability to reprogram B cells offers an unprecedented opportunity for developing a therapeutic approach for several human diseases. Here, we will focus on all the proteins and transcription factors responsible for the developmental commitment of B lymphocytes and how it is harnessed in various applications.
Collapse
Affiliation(s)
- Reetika Rani
- Immunology and Molecular Medicine Laboratory, Department of Life Science, National Institute of Technology, Rourkela, Odisha. 769008, India
| | - Madhusmita Nayak
- Immunology and Molecular Medicine Laboratory, Department of Life Science, National Institute of Technology, Rourkela, Odisha. 769008, India
| | - Bismita Nayak
- Immunology and Molecular Medicine Laboratory, Department of Life Science, National Institute of Technology, Rourkela, Odisha. 769008, India.
| |
Collapse
|
3
|
Fascinating Dendritic Cells—Sentinel Cells of the Immune System a Review. FOLIA VETERINARIA 2021. [DOI: 10.2478/fv-2021-0033] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Abstract
Dendritic cells (DC) are specialized antigen presenting cells which have the unique ability to activate naive T-lymphocytes. Their role in the immune system is much more sophisticated than it seems, as they do not kill the pathogens directly, but provide a long-lasting antigen specific immune response thanks to that sufficiently bridging the innate and the adaptive immunity. In recent years, there has been a growing interest in studies of their role in immune regulation, autoimmune reactions, as well as in immune responses against pathogens and tumours. Processing and presentation capabilities of a highly specific and unique tumour antigen makes them an interesting tool for stimulating effective anti-tumour immunity. In vitro generations of DC represent a preferred model for more detailed studies of DC biology in other fields. The aim of this review was to discuss the main role of dendritic cells in the body as well as their current use as experimental models for further scientific studies.
Collapse
|
4
|
Giza HM, Bozzacco L. Unboxing dendritic cells: Tales of multi-faceted biology and function. Immunology 2021; 164:433-449. [PMID: 34309853 PMCID: PMC8517577 DOI: 10.1111/imm.13394] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2021] [Revised: 07/13/2021] [Accepted: 07/14/2021] [Indexed: 12/14/2022] Open
Abstract
Often referred to as the bridge between innate and adaptive immunity, dendritic cells (DCs) are professional antigen-presenting cells (APCs) that constitute a unique, yet complex cell system. Among other APCs, DCs display the unique property of inducing protective immune responses against invading microbes, or cancer cells, while safeguarding the proper homeostatic equilibrium of the immune system and maintaining self-tolerance. Unsurprisingly, DCs play a role in many diseases such as autoimmunity, allergy, infectious disease and cancer. This makes them attractive but challenging targets for therapeutics. Since their initial discovery, research and understanding of DC biology have flourished. We now recognize the presence of multiple subsets of DCs distributed across tissues. Recent studies of phenotype and gene expression at the single cell level have identified heterogeneity even within the same DC type, supporting the idea that DCs have evolved to greatly expand the flexibility of the immune system to react appropriately to a wide range of threats. This review is meant to serve as a quick and robust guide to understand the basic divisions of DC subsets and their role in the immune system. Between mice and humans, there are some differences in how these subsets are identified and function, and we will point out specific distinctions as necessary. Throughout the text, we are using both fundamental and therapeutic lens to describe overlaps and distinctions and what this could mean for future research and therapies.
Collapse
|
5
|
Hung CH, Wang KY, Liou YH, Wang JP, Huang AYS, Lee TL, Jiang ST, Liao NS, Shyu YC, Shen CKJ. Negative Regulation of the Differentiation of Flk2 - CD34 - LSK Hematopoietic Stem Cells by EKLF/KLF1. Int J Mol Sci 2020; 21:E8448. [PMID: 33182781 PMCID: PMC7697791 DOI: 10.3390/ijms21228448] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2020] [Revised: 10/30/2020] [Accepted: 11/05/2020] [Indexed: 12/13/2022] Open
Abstract
Erythroid Krüppel-like factor (EKLF/KLF1) was identified initially as a critical erythroid-specific transcription factor and was later found to be also expressed in other types of hematopoietic cells, including megakaryocytes and several progenitors. In this study, we have examined the regulatory effects of EKLF on hematopoiesis by comparative analysis of E14.5 fetal livers from wild-type and Eklf gene knockout (KO) mouse embryos. Depletion of EKLF expression greatly changes the populations of different types of hematopoietic cells, including, unexpectedly, the long-term hematopoietic stem cells Flk2- CD34- Lin- Sca1+ c-Kit+ (LSK)-HSC. In an interesting correlation, Eklf is expressed at a relatively high level in multipotent progenitor (MPP). Furthermore, EKLF appears to repress the expression of the colony-stimulating factor 2 receptor β subunit (CSF2RB). As a result, Flk2- CD34- LSK-HSC gains increased differentiation capability upon depletion of EKLF, as demonstrated by the methylcellulose colony formation assay and by serial transplantation experiments in vivo. Together, these data demonstrate the regulation of hematopoiesis in vertebrates by EKLF through its negative regulatory effects on the differentiation of the hematopoietic stem and progenitor cells, including Flk2- CD34- LSK-HSCs.
Collapse
Affiliation(s)
- Chun-Hao Hung
- Institute of Molecular Biology, Academia Sinica, Nankang, Taipei 115, Taiwan; (C.-H.H.); (K.-Y.W.); (Y.-H.L.); (J.-P.W.); (A.Y.-S.H.); (T.-L.L.); (N.-S.L.)
| | - Keh-Yang Wang
- Institute of Molecular Biology, Academia Sinica, Nankang, Taipei 115, Taiwan; (C.-H.H.); (K.-Y.W.); (Y.-H.L.); (J.-P.W.); (A.Y.-S.H.); (T.-L.L.); (N.-S.L.)
| | - Yae-Huei Liou
- Institute of Molecular Biology, Academia Sinica, Nankang, Taipei 115, Taiwan; (C.-H.H.); (K.-Y.W.); (Y.-H.L.); (J.-P.W.); (A.Y.-S.H.); (T.-L.L.); (N.-S.L.)
| | - Jing-Ping Wang
- Institute of Molecular Biology, Academia Sinica, Nankang, Taipei 115, Taiwan; (C.-H.H.); (K.-Y.W.); (Y.-H.L.); (J.-P.W.); (A.Y.-S.H.); (T.-L.L.); (N.-S.L.)
| | - Anna Yu-Szu Huang
- Institute of Molecular Biology, Academia Sinica, Nankang, Taipei 115, Taiwan; (C.-H.H.); (K.-Y.W.); (Y.-H.L.); (J.-P.W.); (A.Y.-S.H.); (T.-L.L.); (N.-S.L.)
| | - Tung-Liang Lee
- Institute of Molecular Biology, Academia Sinica, Nankang, Taipei 115, Taiwan; (C.-H.H.); (K.-Y.W.); (Y.-H.L.); (J.-P.W.); (A.Y.-S.H.); (T.-L.L.); (N.-S.L.)
| | - Si-Tse Jiang
- Department of Research and Development, National Laboratory Animal Center, National Applied Research Laboratories, Tainan 74147, Taiwan;
| | - Nah-Shih Liao
- Institute of Molecular Biology, Academia Sinica, Nankang, Taipei 115, Taiwan; (C.-H.H.); (K.-Y.W.); (Y.-H.L.); (J.-P.W.); (A.Y.-S.H.); (T.-L.L.); (N.-S.L.)
| | - Yu-Chiau Shyu
- Institute of Molecular Biology, Academia Sinica, Nankang, Taipei 115, Taiwan; (C.-H.H.); (K.-Y.W.); (Y.-H.L.); (J.-P.W.); (A.Y.-S.H.); (T.-L.L.); (N.-S.L.)
- Department of Nursing, Chang Gung University of Science and Technology, Taoyuan City 333, Taiwan
- Community Medicine Research Center, Chang Gung Memorial Hospital, Keelung Branch, Keelung 204, Taiwan
| | - Che-Kun James Shen
- Institute of Molecular Biology, Academia Sinica, Nankang, Taipei 115, Taiwan; (C.-H.H.); (K.-Y.W.); (Y.-H.L.); (J.-P.W.); (A.Y.-S.H.); (T.-L.L.); (N.-S.L.)
- The PhD Program for Neural Regenerative Medicine, Taipei Medical University, Taipei 115, Taiwan
| |
Collapse
|
6
|
Trzebanski S, Jung S. Plasticity of monocyte development and monocyte fates. Immunol Lett 2020; 227:66-78. [PMID: 32814154 DOI: 10.1016/j.imlet.2020.07.007] [Citation(s) in RCA: 43] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2020] [Revised: 07/30/2020] [Accepted: 07/31/2020] [Indexed: 01/01/2023]
Abstract
Monocytes are circulating myeloid immune precursor cells that are generated in the bone marrow. Mature monocytes are released into the circulation and, in case of need, recruited to peripheral sites of inflammation to differentiate into monocyte-derived effector cells. In absence of overt inflammation, monocytes also extravasate into selected tissues, where they complement tissue-resident macrophage compartments. Adjustment of these homeostatic monocyte infiltrates to local environment is critical to maintain health, as best established for the intestine. Defined gene expression changes that differ between gut segments presumably help strike the fine balance between the crucial function of these monocyte-derived macrophages as tissue rheostats and their detrimental hyperactivation. Environmental factors that dictate local monocyte differentiation remain incompletely understood. Definition of the latter could aid our general understanding of in vivo monocyte functions and their relation to inflammatory disorders. In this review, we summarize recent advances in our understanding of monocyte subsets, their differentiation into tissue macrophages, and selected contributions of monocyte-derived cells to steady-state physiology. Moreover, we will discuss emerging evidence for an intriguing bifurcation of monocyte development in the bone marrow and potential functional implications. Emphasis will be given to points of controversies, but we will largely focus on the healthy organism. For a discussion of monocyte and macrophage contributions to inflammatory conditions, we refer the reader to other dedicated reviews.
Collapse
Affiliation(s)
| | - Steffen Jung
- Department of Immunology, Weizmann Institute of Science, Rehovot 76100, Israel.
| |
Collapse
|
7
|
Coelho-Dos-Reis JGA, Funakoshi R, Huang J, Pereira FV, Iketani S, Tsuji M. Functional Human CD141+ Dendritic Cells in Human Immune System Mice. J Infect Dis 2020; 221:201-213. [PMID: 31647546 DOI: 10.1093/infdis/jiz432] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2018] [Accepted: 08/20/2019] [Indexed: 12/13/2022] Open
Abstract
BACKGROUND For the purpose of studying functional human dendritic cells (DCs) in a humanized mouse model that mimics the human immune system (HIS), a model referred to as HIS mice was established. METHODS Human immune system mice were made by engrafting NOD/SCID/IL2Rgammanull (NSG) mice with human hematopoietic stem cells (HSCs) following the transduction of genes encoding human cytokines and human leukocyte antigen (HLA)-A2.1 by adeno-associated virus serotype 9 (AAV9) vectors. RESULTS Our results indicate that human DC subsets, such as CD141+CD11c+ and CD1c+CD11c+ myeloid DCs, distribute throughout several organs in HIS mice including blood, bone marrow, spleen, and draining lymph nodes. The CD141+CD11c+ and CD1c+CD11c+ human DCs isolated from HIS mice immunized with adenoviruses expressing malaria/human immunodeficiency virus (HIV) epitopes were able to induce the proliferation of malaria/HIV epitopes-specific human CD8+ T cells in vitro. Upregulation of CD1c was also observed in human CD141+ DCs 1 day after immunization with the adenovirus-based vaccines. CONCLUSIONS Establishment of such a humanized mouse model that mounts functional human DCs enables preclinical assessment of the immunogenicity of human vaccines in vivo.
Collapse
Affiliation(s)
- Jordana G A Coelho-Dos-Reis
- Aaron Diamond AIDS Research Center, Affiliate of The Rockefeller University, New York, New York, USA.,Department of Microbiology, Universidade Federal de Minas Gerais, Minas Gerais, Brazil
| | - Ryota Funakoshi
- Aaron Diamond AIDS Research Center, Affiliate of The Rockefeller University, New York, New York, USA
| | - Jing Huang
- Aaron Diamond AIDS Research Center, Affiliate of The Rockefeller University, New York, New York, USA
| | - Felipe Valença Pereira
- Aaron Diamond AIDS Research Center, Affiliate of The Rockefeller University, New York, New York, USA.,Federal University of Sao Paulo, Sao Paulo, Brazil
| | - Sho Iketani
- Aaron Diamond AIDS Research Center, Affiliate of The Rockefeller University, New York, New York, USA.,Department of Microbiology and Immunology, Columbia University Irving Medical Center, New York, New York, USA
| | - Moriya Tsuji
- Aaron Diamond AIDS Research Center, Affiliate of The Rockefeller University, New York, New York, USA
| |
Collapse
|
8
|
Cumano A, Berthault C, Ramond C, Petit M, Golub R, Bandeira A, Pereira P. New Molecular Insights into Immune Cell Development. Annu Rev Immunol 2020; 37:497-519. [PMID: 31026413 DOI: 10.1146/annurev-immunol-042718-041319] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
During development innate lymphoid cells and specialized lymphocyte subsets colonize peripheral tissues, where they contribute to organogenesis and later constitute the first line of protection while maintaining tissue homeostasis. A few of these subsets are produced only during embryonic development and remain in the tissues throughout life. They are generated through a unique developmental program initiated in lympho-myeloid-primed progenitors, which lose myeloid and B cell potential. They either differentiate into innate lymphoid cells or migrate to the thymus to give rise to embryonic T cell receptor-invariant T cells. At later developmental stages, adaptive T lymphocytes are derived from lympho-myeloid progenitors that colonize the thymus, while lymphoid progenitors become specialized in the production of B cells. This sequence of events highlights the requirement for stratification in the establishment of immune functions that determine efficient seeding of peripheral tissues by a limited number of cells.
Collapse
Affiliation(s)
- Ana Cumano
- Unité Lymphopoïèse, Département d'Immunologie, INSERM U1223, Institut Pasteur, 75724 Paris CEDEX 15, France; , , .,Cellule Pasteur, Université Paris Diderot, Sorbonne Paris Cité, 75015 Paris, France
| | - Claire Berthault
- Unité Lymphopoïèse, Département d'Immunologie, INSERM U1223, Institut Pasteur, 75724 Paris CEDEX 15, France; , , .,Cellule Pasteur, Université Paris Diderot, Sorbonne Paris Cité, 75015 Paris, France
| | - Cyrille Ramond
- Unité Lymphopoïèse, Département d'Immunologie, INSERM U1223, Institut Pasteur, 75724 Paris CEDEX 15, France; , ,
| | - Maxime Petit
- Unité Lymphopoïèse, Département d'Immunologie, INSERM U1223, Institut Pasteur, 75724 Paris CEDEX 15, France; , , .,Cellule Pasteur, Université Paris Diderot, Sorbonne Paris Cité, 75015 Paris, France
| | - Rachel Golub
- Unité Lymphopoïèse, Département d'Immunologie, INSERM U1223, Institut Pasteur, 75724 Paris CEDEX 15, France; , , .,Cellule Pasteur, Université Paris Diderot, Sorbonne Paris Cité, 75015 Paris, France
| | - Antonio Bandeira
- Unité Lymphopoïèse, Département d'Immunologie, INSERM U1223, Institut Pasteur, 75724 Paris CEDEX 15, France; , , .,Cellule Pasteur, Université Paris Diderot, Sorbonne Paris Cité, 75015 Paris, France
| | - Pablo Pereira
- Unité Lymphopoïèse, Département d'Immunologie, INSERM U1223, Institut Pasteur, 75724 Paris CEDEX 15, France; , , .,Cellule Pasteur, Université Paris Diderot, Sorbonne Paris Cité, 75015 Paris, France
| |
Collapse
|
9
|
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: 12] [Impact Index Per Article: 2.4] [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
|
10
|
Abstract
During the past few years, there has been a substantial increase in the understanding of innate immunity. Dendritic cells are emerging as key players in the orchestration of this early phase of immune responses, with a role that will translate into the subsequent type of adaptive immune response against infection. Here we provide an overview of dendritic cell differentiation and function, with particular emphasis on those features unique to the immune defense of the peritoneal cavity and in the context of peritoneal dialysis-associated immune responses. The reader is referred to the primary references included in the accompanying list for specific details in this fascinating field.
Collapse
Affiliation(s)
- Michelle L. McCully
- The FOCIS Centre for Clinical Immunology and Immunotherapeutics, Robarts Research Institute, and the Departments of Microbiology and Immunology, and Medicine, The University of Western Ontario, London, Ontario, Canada
| | - Joaquín Madrenas
- The FOCIS Centre for Clinical Immunology and Immunotherapeutics, Robarts Research Institute, and the Departments of Microbiology and Immunology, and Medicine, The University of Western Ontario, London, Ontario, Canada
| |
Collapse
|
11
|
Shortman K. Dendritic cell development: A personal historical perspective. Mol Immunol 2020; 119:64-68. [PMID: 31986310 DOI: 10.1016/j.molimm.2019.12.016] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2019] [Revised: 12/02/2019] [Accepted: 12/20/2019] [Indexed: 01/01/2023]
Abstract
Dendritic cells(DCs) were once considered as a single cell type closely related developmentally to macrophages. Now we recognise several subtypes of DCs and have outlined several different pathways that potentially lead to their development. This article outlines some of the research findings that led to these changes in perspective, from the point of view of one of the participants.
Collapse
Affiliation(s)
- Ken Shortman
- The Walter and Eliza Hall Institute, Melbourne, Australia.
| |
Collapse
|
12
|
Guermonprez P, Gerber-Ferder Y, Vaivode K, Bourdely P, Helft J. Origin and development of classical dendritic cells. INTERNATIONAL REVIEW OF CELL AND MOLECULAR BIOLOGY 2019; 349:1-54. [PMID: 31759429 DOI: 10.1016/bs.ircmb.2019.08.002] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Classical dendritic cells (cDCs) are mononuclear phagocytes of hematopoietic origin specialized in the induction and regulation of adaptive immunity. Initially defined by their unique T cell activation potential, it became quickly apparent that cDCs would be difficult to distinguish from other phagocyte lineages, by solely relying on marker-based approaches. Today, cDCs definition increasingly embed their unique ontogenetic features. A growing consensus defines cDCs on multiple criteria including: (1) dependency on the fms-like tyrosine kinase 3 ligand hematopoietic growth factor, (2) development from the common DC bone marrow progenitor, (3) constitutive expression of the transcription factor ZBTB46 and (4) the ability to induce, after adequate stimulation, the activation of naïve T lymphocytes. cDCs are a heterogeneous cell population that contains two main subsets, named type 1 and type 2 cDCs, arising from divergent ontogenetic pathways and populating multiple lymphoid and non-lymphoid tissues. Here, we present recent knowledge on the cellular and molecular pathways controlling the specification and commitment of cDC subsets from murine and human hematopoietic stem cells.
Collapse
Affiliation(s)
- Pierre Guermonprez
- King's College London, Centre for Inflammation Biology and Cancer Immunology, The Peter Gorer Department of Immmunobiology, London, United Kingdom; Université de Paris, CNRS ERL8252, INSERM1149, Centre for Inflammation Research, Paris, France.
| | - Yohan Gerber-Ferder
- Institut Curie, PSL Research University, INSERM U932, SiRIC «Translational Immunotherapy Team», Paris, France; Université de Paris, Immunity and Cancer Department, INSERM U932, Institut Curie, Paris, France
| | - Kristine Vaivode
- King's College London, Centre for Inflammation Biology and Cancer Immunology, The Peter Gorer Department of Immmunobiology, London, United Kingdom
| | - Pierre Bourdely
- King's College London, Centre for Inflammation Biology and Cancer Immunology, The Peter Gorer Department of Immmunobiology, London, United Kingdom
| | - Julie Helft
- Institut Curie, PSL Research University, INSERM U932, SiRIC «Translational Immunotherapy Team», Paris, France; Université de Paris, Immunity and Cancer Department, INSERM U932, Institut Curie, Paris, France.
| |
Collapse
|
13
|
Bagadia P, Huang X, Liu TT, Murphy KM. Shared Transcriptional Control of Innate Lymphoid Cell and Dendritic Cell Development. Annu Rev Cell Dev Biol 2019; 35:381-406. [PMID: 31283378 PMCID: PMC6886469 DOI: 10.1146/annurev-cellbio-100818-125403] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Innate immunity and adaptive immunity consist of highly specialized immune lineages that depend on transcription factors for both function and development. In this review, we dissect the similarities between two innate lineages, innate lymphoid cells (ILCs) and dendritic cells (DCs), and an adaptive immune lineage, T cells. ILCs, DCs, and T cells make up four functional immune modules and interact in concert to produce a specified immune response. These three immune lineages also share transcriptional networks governing the development of each lineage, and we discuss the similarities between ILCs and DCs in this review.
Collapse
Affiliation(s)
- Prachi Bagadia
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, Missouri 63108, USA;
| | - Xiao Huang
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, Missouri 63108, USA;
| | - Tian-Tian Liu
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, Missouri 63108, USA;
| | - Kenneth M Murphy
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, Missouri 63108, USA;
- Howard Hughes Medical Institute, Washington University School of Medicine, St. Louis, Missouri 63108, USA
| |
Collapse
|
14
|
Rogers PB, Schwartz EH. Generation of Large Numbers of Myeloid Progenitors and Dendritic Cell Precursors from Murine Bone Marrow Using a Novel Cell Sorting Strategy. J Vis Exp 2018:57365. [PMID: 30148496 PMCID: PMC6126687 DOI: 10.3791/57365] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023] Open
Abstract
Cultures of monocyte-derived dendritic cells (moDC) generated from mouse bone marrow using Granulocyte-Macrophage Colony Stimulating Factor (GM-CSF) have recently been recognized to be more heterogeneous than previously appreciated. These cultures routinely contain moDC as well monocyte-derived macrophages (moMac), and even some less developed cells such as monocytes. The goal of this protocol is to provide a consistent method for identification and separation of the many cell types present in these cultures as they develop, so that their specific functions may be further investigated. The sorting strategy presented here separates cells first into four populations based on expression of Ly6C and CD115, both of which are expressed transiently by cells as they develop in GM-CSF-driven culture. These four populations include Common myeloid progenitors or CMP (Ly6C-, CD115-), granulocyte/macrophage progenitors or GMP (Ly6C+, CD115-), monocytes (Ly6C+, CD115+), and monocyte-derived macrophages or moMac (Ly6C-, CD115+). CD11c is also added to the sorting strategy to distinguish two populations within the Ly6C-, CD115- population: CMP (CD11c-) and moDC (CD11c+). Finally, two populations may be further distinguished within the Ly6C-, CD115+ population based on the level of MHC class II expression. MoMacs express lower levels of MHC class II, while a monocyte-derived DC precursor (moDP) expresses higher MHC class II. This method allows for the reliable isolation of several developmentally distinct populations in numbers sufficient for a variety of functional and developmental analyses. We highlight one such functional readout, the differential responses of these cell types to stimulation with Pathogen-Associated Molecular Patterns (PAMPs).
Collapse
|
15
|
van Leeuwen-Kerkhoff N, Lundberg K, Westers TM, Kordasti S, Bontkes HJ, Lindstedt M, de Gruijl TD, van de Loosdrecht AA. Human Bone Marrow-Derived Myeloid Dendritic Cells Show an Immature Transcriptional and Functional Profile Compared to Their Peripheral Blood Counterparts and Separate from Slan+ Non-Classical Monocytes. Front Immunol 2018; 9:1619. [PMID: 30061890 PMCID: PMC6055354 DOI: 10.3389/fimmu.2018.01619] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2018] [Accepted: 06/29/2018] [Indexed: 12/11/2022] Open
Abstract
The human bone marrow (BM) gives rise to all distinct blood cell lineages, including CD1c+ (cDC2) and CD141+ (cDC1) myeloid dendritic cells (DC) and monocytes. These cell subsets are also present in peripheral blood (PB) and lymphoid tissues. However, the difference between the BM and PB compartment in terms of differentiation state and immunological role of DC is not yet known. The BM may represent both a site for development as well as a possible effector site and so far, little is known in this light with respect to different DC subsets. Using genome-wide transcriptional profiling we found clear differences between the BM and PB compartment and a location-dependent clustering for cDC2 and cDC1 was demonstrated. DC subsets from BM clustered together and separate from the corresponding subsets from PB, which similarly formed a cluster. In BM, a common proliferating and immature differentiating state was observed for the two DC subsets, whereas DC from the PB showed a more immune-activated mature profile. In contrast, BM-derived slan+ non-classical monocytes were closely related to their PB counterparts and not to DC subsets, implying a homogenous prolife irrespective of anatomical localization. Additional functional tests confirmed these transcriptional findings. DC-like functions were prominently exhibited by PB DC. They surpassed BM DC in maturation capacity, cytokine production, and induction of CD4+ and CD8+ T cell proliferation. This first study on myeloid DC in healthy human BM offers new information on steady state DC biology and could potentially serve as a starting point for further research on these immune cells in healthy conditions as well as in diseases.
Collapse
Affiliation(s)
| | | | - Theresia M Westers
- Cancer Center Amsterdam, Department of Hematology, VU University Medical Center, Amsterdam, Netherlands
| | - Shahram Kordasti
- Department of Haematological Medicine, King's College London, London, United Kingdom
| | - Hetty J Bontkes
- Department of Pathology, VU University Medical Center, Amsterdam, Netherlands
| | - Malin Lindstedt
- Department of Immunotechnology, Lund University, Lund, Sweden
| | - Tanja D de Gruijl
- Cancer Center Amsterdam, Department of Medical Oncology, VU University Medical Center, Amsterdam, Netherlands
| | - Arjan A van de Loosdrecht
- Cancer Center Amsterdam, Department of Hematology, VU University Medical Center, Amsterdam, Netherlands
| |
Collapse
|
16
|
Salem ML, Nassef M, Gomaa S, Essa I. Synergistic combination of murine bone marrow-derived dendritic cells loaded ex vivo with whole tumor lysate and systemic chemotherapy mediates antitumor immune responses in vivo. Biomed Pharmacother 2017; 93:286-295. [PMID: 28648976 DOI: 10.1016/j.biopha.2017.06.046] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2017] [Revised: 06/01/2017] [Accepted: 06/16/2017] [Indexed: 02/07/2023] Open
Abstract
In order to get mature dendritic cells (DC) that is a crucial prerequisite for success in tumor immunotherapy protocols. Herein, we assumed that administration of murine bone marrow (BM)-derived DC (BM-DC), loaded ex vivo with whole Ehrlich ascites carcinoma (EAC) lysate, in the context of systemic chemotherapy cyclophosphamide (CTX) to induce antitumor immune responses, may be a good strategy to improve the presentation of tumor-specific antigens to the immune system. In the first series of experiments, BM cells generated either from BM of naïve mice or from BM of EAC-bearing mice were cultured in the presence of GM-CSF and IL-4 for 6days. At day 7, cells were loaded for 48h with one of the following maturation agents: EAC lysate (1mg/ml), poly-inosinic: polycytidylic acid [poly(I:C)] (25μg/ml) or mRNA encoding human telomerase reverse transcriptase (hTERT-mRNA) (2μg/ml). In the second series of experiments, EAC-bearing mice were intraperitoneally (i.p.) injected with CTX followed by i.p. vaccination with DC, loaded ex vivo with EAC lysate. DC yield and the phenotypic expression of maturity-related surface markers of DC (i.e. CD11b and CD11c) in both series of experiments were investigated. As a result, a significant decrease in the number of DC generated from poly(I:C)-supplemented BM culture from EAC-bearing mice has been detected. Loading of BM cells with poly(I:C), EAC lysate or hTERT-mRNA could induce the expression of CD11b and CD11c. Additionally, vaccination of EAC-bearing mice with DC loaded ex vivo with EAC lysate following CTX treatment, resulted in increases in the percentage of multiple populations of CD11b+CD11c+ in BM, spleen and peripheral blood (PB). To conclude, further researches to clarify the mechanism involved in DC maturation are crucial not only to comprehend DC biology but also to optimize DC immunotherapy protocols.
Collapse
Affiliation(s)
- Mohamed L Salem
- Zoology Department, Faculty of Science, Tanta University, El-Giesh St., Gharbia province, Tanta 31527, Egypt; Center of Excellence in Cancer Research, Tanta University, El-Giesh St., Gharbia province, Tanta 31527, Egypt
| | - Mohamed Nassef
- Zoology Department, Faculty of Science, Tanta University, El-Giesh St., Gharbia province, Tanta 31527, Egypt.
| | - Soha Gomaa
- Zoology Department, Faculty of Science, Tanta University, El-Giesh St., Gharbia province, Tanta 31527, Egypt
| | - Ibrahim Essa
- Zoology Department, Faculty of Science, Tanta University, El-Giesh St., Gharbia province, Tanta 31527, Egypt
| |
Collapse
|
17
|
Abstract
The hematopoietic stem cell (HSC) is a multipotent stem cell that resides in the bone marrow and has the ability to form all of the cells of the blood and immune system. Since its first purification in 1988, additional studies have refined the phenotype and functionality of HSCs and characterized all of their downstream progeny. The hematopoietic lineage is divided into two main branches: the myeloid and lymphoid arms. The myeloid arm is characterized by the common myeloid progenitor and all of its resulting cell types. The stages of hematopoiesis have been defined in both mice and humans. During embryological development, the earliest hematopoiesis takes place in yolk sac blood islands and then migrates to the fetal liver and hematopoietic organs. Some adult myeloid populations develop directly from yolk sac progenitors without apparent bone marrow intermediates, such as tissue-resident macrophages. Hematopoiesis also changes over time, with a bias of the dominating HSCs toward myeloid development as animals age. Defects in myelopoiesis contribute to many hematologic disorders, and some of these can be overcome with therapies that target the aberrant stage of development. Furthermore, insights into myeloid development have informed us of mechanisms of programmed cell removal. The CD47/SIRPα axis, a myeloid-specific immune checkpoint, limits macrophage removal of HSCs but can be exploited by hematologic and solid malignancies. Therapeutics targeting CD47 represent a new strategy for treating cancer. Overall, an understanding of hematopoiesis and myeloid cell development has implications for regenerative medicine, hematopoietic cell transplantation, malignancy, and many other diseases.
Collapse
|
18
|
Huang A, Yang Y, Chen S, Xia F, Sun D, Fang D, Xiong S, Jin L, Zhang J. MiR-34a promotes DCs development and inhibits their function on T cell activation by targeting WNT1. Oncotarget 2017; 8:17191-17201. [PMID: 28199987 PMCID: PMC5370032 DOI: 10.18632/oncotarget.15228] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2016] [Accepted: 01/16/2017] [Indexed: 01/22/2023] Open
Abstract
MicroRNAs serve important functions in numerous biological processes. Whether microRNAs also act on dendritic cell (DC) differentiation and function remains unclear. In this study, both conventional DCs (cDCs) and plasmacytoid DCs (pDCs) were increased in miR-34a overexpressing bone marrow chimeric and transgenic (TG) mice. Further experiments showed that miR-34a promoted preDC differentiated into cDCs and pDCs without affecting the proliferation and apoptosis of DCs. Luciferase report assay and Western blot experiments demonstrated that WNT1 is the direct target of miR-34a in DCs. Interestingly, miR-34a overexpressing cDCs also produced a large amount of IL-17a and suppressed T cell activation because of the inhibition of TCF1 expression, thus increasing RORγT expression. Taken together, miR-34a promotes preDC to differentiate into cDCs and pDCs, as well as inhibits the function of cDCs on the activation of CD4+ T cells by producing IL-17a.
Collapse
Affiliation(s)
- Anfei Huang
- Institutes of Biology and Medical Sciences, Soochow University, Suzhou, Jiangsu Province 215123, People's Republic of China
| | - Yi Yang
- Institutes of Biology and Medical Sciences, Soochow University, Suzhou, Jiangsu Province 215123, People's Republic of China
| | - Si Chen
- Institutes of Biology and Medical Sciences, Soochow University, Suzhou, Jiangsu Province 215123, People's Republic of China
| | - Fei Xia
- Institutes of Biology and Medical Sciences, Soochow University, Suzhou, Jiangsu Province 215123, People's Republic of China
| | - Di Sun
- Institutes of Biology and Medical Sciences, Soochow University, Suzhou, Jiangsu Province 215123, People's Republic of China
| | - Deyu Fang
- Department of Pathology, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA
| | - Sidong Xiong
- Institutes of Biology and Medical Sciences, Soochow University, Suzhou, Jiangsu Province 215123, People's Republic of China
| | - Liping Jin
- Clinical and Translational Research Center, Shanghai First Maternity and Infant Hospital, Tongji University School of Medicine, Shanghai 200040, China
| | - Jinping Zhang
- Institutes of Biology and Medical Sciences, Soochow University, Suzhou, Jiangsu Province 215123, People's Republic of China
| |
Collapse
|
19
|
Rogers PB, Driessnack MG, Hiltbold Schwartz E. Analysis of the developmental stages, kinetics, and phenotypes exhibited by myeloid cells driven by GM-CSF in vitro. PLoS One 2017; 12:e0181985. [PMID: 28750033 PMCID: PMC5531556 DOI: 10.1371/journal.pone.0181985] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2017] [Accepted: 07/10/2017] [Indexed: 12/24/2022] Open
Abstract
The developmental progression of conventional DC has been quite well defined, yet the developmental pathway of monocyte-derived, GM-CSF-driven DC is less well understood. We addressed this issue by establishing an isolation strategy that identifies five distinct GM-CSF derived cell types. Expression of Ly6C and CD115 (Csf-1R) was used to identify and isolate four populations. One of the populations could be further separated based on CD11c expression, distinguishing five populations. We further defined these cells based on expression of transcription factors and markers of early and later stages of myeloid development. These discreet developmental stages corresponded well with previously defined populations: Common Myeloid Progenitors (CMP), Granulocyte/Macrophage Progenitors (GMP), Monocytes, as well as Monocyte-derived macrophages (moMac) and Monocyte-derived DC (moDC). Finally, within the moMac population we also identified moDC precursor activity (moDP) that could be distinguished from moMac and moDC based on their level of MHC class II expression and developmental plasticity.
Collapse
Affiliation(s)
- Peter B. Rogers
- Department of Biological Sciences, Auburn University, Auburn AL, United States of America
| | - Michelle G. Driessnack
- Department of Biological Sciences, Auburn University, Auburn AL, United States of America
| | | |
Collapse
|
20
|
Helft J, Anjos-Afonso F, van der Veen AG, Chakravarty P, Bonnet D, Reis e Sousa C. Dendritic Cell Lineage Potential in Human Early Hematopoietic Progenitors. Cell Rep 2017; 20:529-537. [PMID: 28723558 PMCID: PMC5529209 DOI: 10.1016/j.celrep.2017.06.075] [Citation(s) in RCA: 53] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2017] [Revised: 05/22/2017] [Accepted: 06/23/2017] [Indexed: 01/16/2023] Open
Abstract
Conventional dendritic cells (cDCs) are thought to descend from a DC precursor downstream of the common myeloid progenitor (CMP). However, a mouse lymphoid-primed multipotent progenitor has been shown to generate cDCs following a DC-specific developmental pathway independent of monocyte and granulocyte poiesis. Similarly, here we show that, in humans, a large fraction of multipotent lymphoid early progenitors (MLPs) gives rise to cDCs, in particular the subset known as cDC1, identified by co-expression of DNGR-1 (CLEC9A) and CD141 (BDCA-3). Single-cell analysis indicates that over one-third of MLPs have the potential to efficiently generate cDCs. cDC1s generated from CMPs or MLPs do not exhibit differences in transcriptome or phenotype. These results demonstrate an early imprinting of the cDC lineage in human hematopoiesis and highlight the plasticity of developmental pathways giving rise to human DCs.
Collapse
Affiliation(s)
- Julie Helft
- Immunobiology Laboratory, The Francis Crick Institute, 1 Midland Road, London NW1 1AT, UK.
| | - Fernando Anjos-Afonso
- Haematopoietic Stem Cell Laboratory, The Francis Crick Institute, 1 Midland Road, London NW1 1AT, UK; Cardiff University, European Cancer Stem Cell Institute, Cardiff, CF10 3XQ, UK
| | | | - Probir Chakravarty
- Bioinformatics, The Francis Crick Institute, 1 Midland Road, London NW1 1AT, UK
| | - Dominique Bonnet
- Haematopoietic Stem Cell Laboratory, The Francis Crick Institute, 1 Midland Road, London NW1 1AT, UK
| | - Caetano Reis e Sousa
- Immunobiology Laboratory, The Francis Crick Institute, 1 Midland Road, London NW1 1AT, UK.
| |
Collapse
|
21
|
Hsia HC, Stopford CM, Zhang Z, Damania B, Baldwin AS. Signal transducer and activator of transcription 3 ( Stat3) regulates host defense and protects mice against herpes simplex virus-1 (HSV-1) infection. J Leukoc Biol 2016; 101:1053-1064. [PMID: 27965384 DOI: 10.1189/jlb.4a1016-199rr] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2016] [Revised: 10/31/2016] [Accepted: 11/17/2016] [Indexed: 12/31/2022] Open
Abstract
Signal transducer and activator of transcription 3 (STAT3) mediates cellular responses to multiple cytokines, governs gene expression, and regulates the development and activation of immune cells. STAT3 also modulates reactivation of latent herpes simplex virus-1 (HSV-1) in ganglia. However, it is unclear how STAT3 regulates the innate immune response during the early phase of HSV-1 lytic infection. Many cell types critical for the innate immunity are derived from the myeloid lineage. Therefore, in this study, we used myeloid-specific Stat3 knockout mice to investigate the role of STAT3 in the innate immune response against HSV-1. Our results demonstrate that Stat3 knockout bone marrow-derived macrophages (BMMs) expressed decreased levels of interferon-α (IFN-α) and interferon-stimulated genes (ISGs) upon HSV-1 infection. In vivo, knockout mice were more susceptible to HSV-1, as marked by higher viral loads and more significant weight loss. Splenic expression of IFN-α and ISGs was reduced in the absence of STAT3, indicating that STAT3 is required for optimal type I interferon response to HSV-1. Expression of TNF-α and IL-12, cytokines that have been shown to limit HSV-1 replication and pathogenesis, was also significantly lower in knockout mice. Interestingly, Stat3 knockout mice failed to expand the CD8+ conventional DC (cDC) population upon HSV-1 infection, and this was accompanied by impaired NK and CD8 T cell activation. Collectively, our data demonstrate that myeloid-specific Stat3 deletion causes defects in multiple aspects of the immune system and that STAT3 has a protective role at the early stage of systemic HSV-1 infection.
Collapse
Affiliation(s)
- Hung-Ching Hsia
- Department of Cell Biology and Physiology, University of North Carolina, Chapel Hill, North Carolina, USA.,Lineberger Comprehensive Cancer Center, University of North Carolina, Chapel Hill, North Carolina, USA; and
| | - Charles M Stopford
- Lineberger Comprehensive Cancer Center, University of North Carolina, Chapel Hill, North Carolina, USA; and
| | - Zhigang Zhang
- Lineberger Comprehensive Cancer Center, University of North Carolina, Chapel Hill, North Carolina, USA; and
| | - Blossom Damania
- Lineberger Comprehensive Cancer Center, University of North Carolina, Chapel Hill, North Carolina, USA; and.,Department of Microbiology and Immunology, University of North Carolina, Chapel Hill, North Carolina, USA
| | - Albert S Baldwin
- Department of Cell Biology and Physiology, University of North Carolina, Chapel Hill, North Carolina, USA; .,Lineberger Comprehensive Cancer Center, University of North Carolina, Chapel Hill, North Carolina, USA; and
| |
Collapse
|
22
|
Devi KSP, Anandasabapathy N. The origin of DCs and capacity for immunologic tolerance in central and peripheral tissues. Semin Immunopathol 2016; 39:137-152. [PMID: 27888331 DOI: 10.1007/s00281-016-0602-0] [Citation(s) in RCA: 55] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2016] [Accepted: 10/28/2016] [Indexed: 12/20/2022]
Abstract
Dendritic cells (DCs) are specialized immune sentinels that play key role in maintaining immune homeostasis by efficiently regulating the delicate balance between protective immunity and tolerance to self. Although DCs respond to maturation signals present in the surrounding milieu, multiple layers of suppression also co-exist that reduce the infringement of tolerance against self-antigens. These tolerance inducing properties of DCs are governed by their origin and a range of other factors including distribution, cytokines, growth factors, and transcriptional programing, that collectively impart suppressive functions to these cells. DCs directing tolerance secrete anti-inflammatory cytokines and induce naïve T cells or B cells to differentiate into regulatory T cells (Tregs) or B cells. In this review, we provide a detailed outlook on the molecular mechanisms that induce functional specialization to govern central or peripheral tolerance. The tolerance-inducing nature of DCs can be exploited to overcome autoimmunity and rejection in graft transplantation.
Collapse
Affiliation(s)
- K Sanjana P Devi
- Department of Dermatology/Harvard Skin Disease Research Center, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Niroshana Anandasabapathy
- Department of Dermatology/Harvard Skin Disease Research Center, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
23
|
Perié L, Duffy KR. Retracing thein vivohaematopoietic tree using single-cell methods. FEBS Lett 2016; 590:4068-4083. [DOI: 10.1002/1873-3468.12299] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2016] [Revised: 07/08/2016] [Accepted: 07/09/2016] [Indexed: 12/14/2022]
Affiliation(s)
- Leïla Perié
- Institut Curie; PSL Research University; CNRS UMR168; Paris France
- Sorbonne Universités; UPMC Univ Paris 06; France
| | - Ken R. Duffy
- Hamilton Institute; Maynooth University; Co Kildare Ireland
| |
Collapse
|
24
|
Puhr S, Lee J, Zvezdova E, Zhou YJ, Liu K. Dendritic cell development-History, advances, and open questions. Semin Immunol 2016; 27:388-96. [PMID: 27040276 DOI: 10.1016/j.smim.2016.03.012] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/10/2015] [Accepted: 03/16/2016] [Indexed: 01/22/2023]
Abstract
Dendritic cells (DCs) are uniquely potent in orchestrating T cell immune response, thus they are indispensable immune sentinels. They originate from progenitors in the bone marrow through hematopoiesis, a highly regulated developmental process involving multiple cellular and molecular events. This review highlights studies of DC development-from the discovery of DCs as glass-adherent antigen presenting cells to the debate and resolution of their origin and lineage map. In particular, we summarize the roles of lineage-specific cytokines, the placement of distinct hematopoietic progenitors within the DC lineage and transcriptional programs governing DC development, which together have allowed us to diagram the current view of DC hematopoiesis. Important open questions and debates on the DC development and relevant models are also discussed.
Collapse
Affiliation(s)
- Sarah Puhr
- Columbia University Medical Center, Department of Microbiology and Immunology, New York, NY 10032, USA.
| | - Jaeyop Lee
- Columbia University Medical Center, Department of Microbiology and Immunology, New York, NY 10032, USA.
| | - Ekaterina Zvezdova
- Columbia University Medical Center, Department of Microbiology and Immunology, New York, NY 10032, USA.
| | - Yu J Zhou
- Columbia University Medical Center, Department of Microbiology and Immunology, New York, NY 10032, USA
| | - Kang Liu
- Columbia University Medical Center, Department of Microbiology and Immunology, New York, NY 10032, USA.
| |
Collapse
|
25
|
Duriancik DM, Gardner EM. Energy restriction impairs dendritic cell development in C57BL/6J mice. Mech Ageing Dev 2016; 154:9-19. [PMID: 26876761 DOI: 10.1016/j.mad.2016.02.002] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2015] [Revised: 01/05/2016] [Accepted: 02/05/2016] [Indexed: 12/13/2022]
Abstract
Dendritic cells (DC) are antigen-presenting cells known for stimulating naïve T lymphocytes. The sequential stages of DC development from common myeloid progenitors have been elucidated in murine bone marrow. Energy-restriction (ER) is a pro-longevity dietary intervention with mixed immunological outcomes. The objective of this study was to examine the development of DC in adult C57Bl6J mice fed a 40% ER diet. We observed increased myeloid progenitors, but decreased common DC progenitors, precursor conventional DC and plasmacytoid DC. Furthermore, we observed increased macrophages and cells expressing CD169 in the bone marrow of ER mice. There was no significant difference in DC subsets from unfractionated ER and ad libitum-fed murine bone marrow samples cultured in GM-CSF-supplemented media or Flt3L-supplemented media. Examining rates of proliferation with 6h BrdU incorporation and Ki-67 staining showed these DC progenitor populations have different proliferation rates in ER compared with AL mice. We show here, for the first time, ER results in altered myelopoiesis resulting in reduced DC development but enhanced monocyte/macrophage development in steady-state C57Bl6J mice. In conclusion, these data may partially explain prior observations of impaired early innate immune responses to primary infection such as influenza in ER mice.
Collapse
Affiliation(s)
- David M Duriancik
- Department of Food Science & Human Nutrition, Michigan State University, East Lansing, MI 48824-1224, United States
| | - Elizabeth M Gardner
- Department of Food Science & Human Nutrition, Michigan State University, East Lansing, MI 48824-1224, United States.
| |
Collapse
|
26
|
Murphy TL, Grajales-Reyes GE, Wu X, Tussiwand R, Briseño CG, Iwata A, Kretzer NM, Durai V, Murphy KM. Transcriptional Control of Dendritic Cell Development. Annu Rev Immunol 2015; 34:93-119. [PMID: 26735697 DOI: 10.1146/annurev-immunol-032713-120204] [Citation(s) in RCA: 323] [Impact Index Per Article: 32.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
The dendritic cells (DCs) of the immune system function in innate and adaptive responses by directing activity of various effector cells rather than serving as effectors themselves. DCs and closely related myeloid lineages share expression of many surface receptors, presenting a challenge in distinguishing their unique in vivo functions. Recent work has taken advantage of unique transcriptional programs to identify and manipulate murine DCs in vivo. This work has assigned several nonredundant in vivo functions to distinct DC lineages, consisting of plasmacytoid DCs and several subsets of classical DCs that promote different immune effector modules in response to pathogens. In parallel, a correspondence between human and murine DC subsets has emerged, underlying structural similarities for the DC lineages between these species. Recent work has begun to unravel the transcriptional circuitry that controls the development and diversification of DCs from common progenitors in the bone marrow.
Collapse
Affiliation(s)
- Theresa L Murphy
- Department of Pathology and Immunology, Washington University School of Medicine in St. Louis, Missouri 63110;
| | - Gary E Grajales-Reyes
- Department of Pathology and Immunology, Washington University School of Medicine in St. Louis, Missouri 63110;
| | - Xiaodi Wu
- Department of Pathology and Immunology, Washington University School of Medicine in St. Louis, Missouri 63110;
| | - Roxane Tussiwand
- Department of Biomedicine, University of Basel, 4058 Basel, Switzerland
| | - Carlos G Briseño
- Department of Pathology and Immunology, Washington University School of Medicine in St. Louis, Missouri 63110;
| | - Arifumi Iwata
- Department of Pathology and Immunology, Washington University School of Medicine in St. Louis, Missouri 63110;
| | - Nicole M Kretzer
- Department of Pathology and Immunology, Washington University School of Medicine in St. Louis, Missouri 63110;
| | - Vivek Durai
- Department of Pathology and Immunology, Washington University School of Medicine in St. Louis, Missouri 63110;
| | - Kenneth M Murphy
- Department of Pathology and Immunology, Washington University School of Medicine in St. Louis, Missouri 63110; .,Howard Hughes Medical Institute, Washington University School of Medicine in St. Louis, Missouri 63110
| |
Collapse
|
27
|
Kubicka-Sierszen A, Grzegorczyk JŁ. The influence of infectious factors on dendritic cell apoptosis. Arch Med Sci 2015; 11:1044-51. [PMID: 26528349 PMCID: PMC4624750 DOI: 10.5114/aoms.2015.54860] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/08/2013] [Revised: 06/04/2013] [Accepted: 10/02/2013] [Indexed: 12/23/2022] Open
Abstract
Pathogens can have a negative influence on dendritic cells (DCs), causing their apoptosis, which prevents active presentation of foreign antigens. It results in a state of immunosuppression which makes the body susceptible to secondary infections. Infected immature DCs have lower expression of co-stimulatory and adhesion molecules, reduced ability to secrete cytokines and an inhibited maturation process and are incapable of effective antigen presentation and activation of T-lymphocytes. In some cases, the ability of DCs to undergo rapid apoptosis is important for the body defense, which is probably because of DCs' ability to cross-present and cooperate with other cells. Apoptotic bodies released from the infected DCs are phagocytosed by other DCs, which then stimulate the effector cells and present antigens more efficiently than infected cells. The aim of this article is to review how the DCs respond to viral and bacterial factors and which biochemical mechanisms are responsible for their apoptosis.
Collapse
Affiliation(s)
- Agata Kubicka-Sierszen
- Department of Microbiology and Laboratory Medical Immunology, Faculty of Medicine, Medical University of Lodz, Lodz, Poland
| | - Janina Ł Grzegorczyk
- Department of Microbiology and Laboratory Medical Immunology, Faculty of Medicine, Medical University of Lodz, Lodz, Poland
| |
Collapse
|
28
|
Abstract
Dendritic cells (DCs) are a heterogeneous group of mononuclear phagocytes with versatile roles in immunity. They are classified predominantly based on phenotypic and functional properties, namely their stellate morphology, expression of the integrin CD11c, and major histocompatibility class II molecules, as well as their superior capacity to migrate to secondary lymphoid organs and stimulate naïve T cells. However, these attributes are not exclusive to DCs and often change within inflammatory or infectious environments. This led to debates over cell identification and questioned even the mere existence of DCs as distinct leukocyte lineage. Here, we review experimental approaches taken to fate map DCs and discuss how these have shaped our understanding of DC ontogeny and lineage affiliation. Considering the ontogenetic properties of DCs will help to overcome the inherent shortcomings of purely phenotypic- and function-based approaches to cell definition and will yield a more robust way of DC classification.
Collapse
Affiliation(s)
- Mateusz Pawel Poltorak
- Institute for Medical Microbiology, Immunology and Hygiene, Technische Universität München , Munich , Germany
| | - Barbara Ursula Schraml
- Institute for Medical Microbiology, Immunology and Hygiene, Technische Universität München , Munich , Germany
| |
Collapse
|
29
|
Toward defining a ‘lineage’ – The case for dendritic cells. Semin Cell Dev Biol 2015; 41:3-8. [DOI: 10.1016/j.semcdb.2015.02.004] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2014] [Accepted: 02/10/2015] [Indexed: 12/23/2022]
|
30
|
Kim SJ, Diamond B. Modulation of tolerogenic dendritic cells and autoimmunity. Semin Cell Dev Biol 2015; 41:49-58. [PMID: 24747368 PMCID: PMC9973561 DOI: 10.1016/j.semcdb.2014.04.020] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2013] [Revised: 04/07/2014] [Accepted: 04/09/2014] [Indexed: 12/23/2022]
Abstract
A key function of dendritic cells (DCs) is to induce either immune tolerance or immune activation. Many new DC subsets are being recognized, and it is now clear that each DC subset has a specialized function. For example, different DC subsets may express different cell surface molecules and respond differently to activation by secretion of a unique cytokine profile. Apart from intrinsic differences among DC subsets, various immune modulators in the microenvironment may influence DC function; inappropriate DC function is closely related to the development of immune disorders. The most exciting recent advance in DC biology is appreciation of human DC subsets. In this review, we discuss functionally different mouse and human DC subsets both in lymphoid organs and non-lymphoid organs, the molecules that regulate DC function, and the emerging understanding of the contribution of DCs to autoimmune diseases.
Collapse
Affiliation(s)
| | - Betty Diamond
- The Center for Autoimmune and Musculoskeletal Diseases, The Feinstein Institute for Medical Research, United States.
| |
Collapse
|
31
|
Tan EMS, Chudakova DA, Davis PF, Brasch HD, Itinteang T, Tan ST. Characterisation of subpopulations of myeloid cells in infantile haemangioma. J Clin Pathol 2015; 68:571-4. [DOI: 10.1136/jclinpath-2014-202846] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2014] [Accepted: 03/12/2015] [Indexed: 11/03/2022]
|
32
|
Abstract
In the 40 years since their discovery, dendritic cells (DCs) have been recognized as central players in immune regulation. DCs sense microbial stimuli through pathogen-recognition receptors (PRRs) and decode, integrate, and present information derived from such stimuli to T cells, thus stimulating immune responses. DCs can also regulate the quality of immune responses. Several functionally specialized subsets of DCs exist, but DCs also display functional plasticity in response to diverse stimuli. In addition to sensing pathogens via PRRs, emerging evidence suggests that DCs can also sense stress signals, such as amino acid starvation, through ancient stress and nutrient sensing pathways, to stimulate adaptive immunity. Here, I discuss these exciting advances in the context of a historic perspective on the discovery of DCs and their role in immune regulation. I conclude with a discussion of emerging areas in DC biology in the systems immunology era and suggest that the impact of DCs on immunity can be usefully contextualized in a hierarchy-of-organization model in which DCs, their receptors and signaling networks, cell-cell interactions, tissue microenvironment, and the host macroenvironment represent different levels of the hierarchy. Immunity or tolerance can then be represented as a complex function of each of these hierarchies.
Collapse
Affiliation(s)
- Bali Pulendran
- Emory Vaccine Center and Yerkes National Primate Research Center, Emory University, Atlanta, Georgia 30329;
| |
Collapse
|
33
|
Wu X, Xu F. Dendritic cells during Staphylococcus aureus infection: subsets and roles. J Transl Med 2014; 12:358. [PMID: 25519813 PMCID: PMC4279898 DOI: 10.1186/s12967-014-0358-z] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2014] [Accepted: 12/10/2014] [Indexed: 12/22/2022] Open
Abstract
Dendritic cells (DCs) are professional antigen-presenting cells (APCs) that play a crucial role in both innate and adaptive immune responses. DCs orient the immune responses by modulating the balance between protective immunity to pathogens and tolerance to self-antigens. Staphylococcus aureus (S. aureus) is a common member of human skin microbiota and can cause severe infections with significant morbidity and mortality. Protective immunity to pathogens by DCs is required for clearance of S. aureus. DCs sense the presence of the staphylococcal components using pattern recognition receptors (PRRs) and then orchestrate immune systems to resolve infections. This review summarizes the possible roles of DCs, in particular their Toll-like receptors (TLRs) involved in S. aureus infection and strategies by which the pathogen affects activation and function of DCs.
Collapse
Affiliation(s)
- Xuejie Wu
- Department of Infectious Diseases, Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310009, China.
| | - Feng Xu
- Department of Infectious Diseases, Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310009, China.
| |
Collapse
|
34
|
Strauss V, Kolle SN, Honarvar N, Dammann M, Groeters S, Faulhammer F, Landsiedel R, van Ravenzwaay B. Immunophenotyping does not improve predictivity of the local lymph node assay in mice. J Appl Toxicol 2014; 35:434-45. [DOI: 10.1002/jat.3042] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2014] [Revised: 06/05/2014] [Accepted: 06/05/2014] [Indexed: 11/06/2022]
Affiliation(s)
- Volker Strauss
- BASF SE; Experimental Toxicology and Ecology; Z470 67056 Ludwigshafen Germany
| | - Susanne N. Kolle
- BASF SE; Experimental Toxicology and Ecology; Z470 67056 Ludwigshafen Germany
| | - Naveed Honarvar
- BASF SE; Experimental Toxicology and Ecology; Z470 67056 Ludwigshafen Germany
| | - Martina Dammann
- BASF SE; Experimental Toxicology and Ecology; Z470 67056 Ludwigshafen Germany
| | - Sibylle Groeters
- BASF SE; Experimental Toxicology and Ecology; Z470 67056 Ludwigshafen Germany
| | - Frank Faulhammer
- BASF SE; Product Safety, Regulatory Toxicology; Ludwigshafen Germany
| | - Robert Landsiedel
- BASF SE; Experimental Toxicology and Ecology; Z470 67056 Ludwigshafen Germany
| | | |
Collapse
|
35
|
Wang H, Yan M, Sun J, Jain S, Yoshimi R, Abolfath SM, Ozato K, Coleman WG, Ng AP, Metcalf D, DiRago L, Nutt SL, Morse HC. A reporter mouse reveals lineage-specific and heterogeneous expression of IRF8 during lymphoid and myeloid cell differentiation. THE JOURNAL OF IMMUNOLOGY 2014; 193:1766-77. [PMID: 25024380 DOI: 10.4049/jimmunol.1301939] [Citation(s) in RCA: 62] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
The IFN regulatory factor family member 8 (IRF8) regulates differentiation of lymphoid and myeloid lineage cells by promoting or suppressing lineage-specific genes. How IRF8 promotes hematopoietic progenitors to commit to one lineage while preventing the development of alternative lineages is not known. In this study, we report an IRF8-EGFP fusion protein reporter mouse that revealed previously unrecognized patterns of IRF8 expression. Differentiation of hematopoietic stem cells into oligopotent progenitors is associated with progressive increases in IRF8-EGFP expression. However, significant induction of IRF8-EGFP is found in granulocyte-myeloid progenitors and the common lymphoid progenitors but not the megakaryocytic-erythroid progenitors. Surprisingly, IRF8-EGFP identifies three subsets of the seemingly homogeneous granulocyte-myeloid progenitors with an intermediate level of expression of EGFP defining bipotent progenitors that differentiation into either EGFP(hi) monocytic progenitors or EGFP(lo) granulocytic progenitors. Also surprisingly, IRF8-EGFP revealed a highly heterogeneous pre-pro-B population with a fluorescence intensity ranging from background to 4 orders above background. Interestingly, IRF8-EGFP readily distinguishes true B cell committed (EGFP(int)) from those that are noncommitted. Moreover, dendritic cell progenitors expressed extremely high levels of IRF8-EGFP. Taken together, the IRF8-EGFP reporter revealed previously unrecognized subsets with distinct developmental potentials in phenotypically well-defined oligopotent progenitors, providing new insights into the dynamic heterogeneity of developing hematopoietic progenitors.
Collapse
Affiliation(s)
- Hongsheng Wang
- Virology and Cellular Immunology Section, Laboratory of Immunogenetics, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, MD 20852;
| | - Ming Yan
- Laboratory of Biochemistry and Genetics, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, MD 20892
| | - Jiafang Sun
- Virology and Cellular Immunology Section, Laboratory of Immunogenetics, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, MD 20852
| | - Shweta Jain
- Virology and Cellular Immunology Section, Laboratory of Immunogenetics, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, MD 20852
| | - Ryusuke Yoshimi
- Department of Internal Medicine and Clinical Immunology, Yokohama City University Graduate School of Medicine, Yokohama 236-0004, Japan
| | - Sanaz Momben Abolfath
- Virology and Cellular Immunology Section, Laboratory of Immunogenetics, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, MD 20852
| | - Keiko Ozato
- Program in Genomics of Differentiation, National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD 20892
| | - William G Coleman
- Laboratory of Biochemistry and Genetics, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, MD 20892
| | - Ashley P Ng
- Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria 3052, Australia; and Department of Medical Biology, University of Melbourne, Parkville, Victoria 3010, Australia
| | - Donald Metcalf
- Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria 3052, Australia; and Department of Medical Biology, University of Melbourne, Parkville, Victoria 3010, Australia
| | - Ladina DiRago
- Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria 3052, Australia; and
| | - Stephen L Nutt
- Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria 3052, Australia; and Department of Medical Biology, University of Melbourne, Parkville, Victoria 3010, Australia
| | - Herbert C Morse
- Virology and Cellular Immunology Section, Laboratory of Immunogenetics, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, MD 20852;
| |
Collapse
|
36
|
Briseño CG, Murphy TL, Murphy KM. Complementary diversification of dendritic cells and innate lymphoid cells. Curr Opin Immunol 2014; 29:69-78. [PMID: 24874447 PMCID: PMC5161034 DOI: 10.1016/j.coi.2014.04.006] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2014] [Revised: 03/28/2014] [Accepted: 04/29/2014] [Indexed: 01/25/2023]
Abstract
Dendritic cells (DCs) are professional antigen presenting cells conventionally thought to mediate cellular adaptive immune responses. Recent studies have led to the recognition of a non-redundant role for DCs in orchestrating innate immune responses, and in particular, for DC subset-specific interactions with innate lymphoid cells (ILCs). Recently recognized as important effectors of early immune responses, ILCs develop into subsets which mirror the transcriptional and cytokine profile of their T cell subset counterparts. DC diversification into functional subsets provides for modules of pathogen sensing and cytokine production that direct pathogen-appropriate ILC and T cell responses. This review focuses on the recent advances in the understanding of DC development, and their function in orchestrating the innate immune modules.
Collapse
Affiliation(s)
- Carlos G Briseño
- Department of Pathology and Immunology, Washington University in St. Louis, School of Medicine, 660 S. Euclid Avenue, St. Louis, MO 63110, USA
| | - Theresa L Murphy
- Department of Pathology and Immunology, Washington University in St. Louis, School of Medicine, 660 S. Euclid Avenue, St. Louis, MO 63110, USA
| | - Kenneth M Murphy
- Department of Pathology and Immunology, Washington University in St. Louis, School of Medicine, 660 S. Euclid Avenue, St. Louis, MO 63110, USA; Howard Hughes Medical Institute, Washington University in St. Louis, School of Medicine, 660 S. Euclid Avenue, St. Louis, MO 63110, USA.
| |
Collapse
|
37
|
Kyoizumi S, Kubo Y, Kajimura J, Yoshida K, Hayashi T, Nakachi K, Young LF, Moore MA, van den Brink MRM, Kusunoki Y. Linkage between dendritic and T cell commitments in human circulating hematopoietic progenitors. THE JOURNAL OF IMMUNOLOGY 2014; 192:5749-60. [PMID: 24835400 DOI: 10.4049/jimmunol.1303260] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
The relationships between commitments of dendritic cells (DCs) and T cells in human hematopoietic stem cells are not well understood. In this study, we enumerate and characterize conventional DC and plasmacytoid DC precursors in association with T cell and thymus-derived types of NK cell precursors among CD34(+) hematopoietic progenitor cells (HPCs) circulating in human peripheral blood. By limiting-dilution analyses using coculture with stroma cells expressing Notch1 ligand, the precursor frequencies (PFs) of DCs in HPCs were found to significantly correlate with T cell PFs, but not with NK cell PFs, among healthy donors. Clonal analyses showed that the majority of T/NK dual- and T single-lineage precursors-but only a minority of NK single-lineage precursors-were associated with the generation of DC progenies. All clones producing both DC and T cell progenies were found with monocyte and/or granulocyte progenies, suggesting DC differentiation via myeloid DC pathways. Analyses of peripheral blood HPC subpopulations revealed that the lineage split between DC and T/NK cell progenitor occurs at the stage prior to bifurcation into T and NK cell lineages. The findings suggest a strong linkage between DC and T cell commitments, which may be imprinted in circulating lymphoid-primed multipotent progenitors or in more upstream HPCs.
Collapse
Affiliation(s)
- Seishi Kyoizumi
- Department of Radiobiology/Molecular Epidemiology, Radiation Effects Research Foundation, Hiroshima 732-0815, Japan;
| | - Yoshiko Kubo
- Department of Radiobiology/Molecular Epidemiology, Radiation Effects Research Foundation, Hiroshima 732-0815, Japan
| | - Junko Kajimura
- Department of Radiobiology/Molecular Epidemiology, Radiation Effects Research Foundation, Hiroshima 732-0815, Japan
| | - Kengo Yoshida
- Department of Radiobiology/Molecular Epidemiology, Radiation Effects Research Foundation, Hiroshima 732-0815, Japan
| | - Tomonori Hayashi
- Department of Radiobiology/Molecular Epidemiology, Radiation Effects Research Foundation, Hiroshima 732-0815, Japan
| | - Kei Nakachi
- Department of Radiobiology/Molecular Epidemiology, Radiation Effects Research Foundation, Hiroshima 732-0815, Japan
| | - Lauren F Young
- Department of Medicine and Immunology, Memorial Sloan-Kettering Cancer Center, New York, NY 10065; and
| | - Malcolm A Moore
- Cell Biology Program, Memorial Sloan-Kettering Cancer Center, New York, NY 10065
| | - Marcel R M van den Brink
- Department of Medicine and Immunology, Memorial Sloan-Kettering Cancer Center, New York, NY 10065; and
| | - Yoichiro Kusunoki
- Department of Radiobiology/Molecular Epidemiology, Radiation Effects Research Foundation, Hiroshima 732-0815, Japan
| |
Collapse
|
38
|
Paul F, Amit I. Plasticity in the transcriptional and epigenetic circuits regulating dendritic cell lineage specification and function. Curr Opin Immunol 2014; 30:1-8. [PMID: 24820527 DOI: 10.1016/j.coi.2014.04.004] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2014] [Accepted: 04/09/2014] [Indexed: 12/23/2022]
Abstract
Dendritic cells (DC) are critical and functionally versatile innate immune sentinels. Here, we coarsely partition the adult DC lineage into three developmental subtypes and argue that pioneer transcription factors and chromatin remodeling are responsible for specification and plasticity between the DC subsets. Subsequently, intricate signaling-dependent transcription factor networks generate different functional states in response to pathogen stimuli within a specified DC subtype. To expand our understanding of lineage heterogeneity and functional activation states, we discuss the use of single cell genomics approaches in the context of a newly emerging systems immunology era, complementing the dichotomous definition of immune cells based solely on their surface marker expression. Rapid developments in single cell genomics are beginning to provide us with robust tools to potentially revise the working models of DC specification and the common hematopoietic tree.
Collapse
Affiliation(s)
- Franziska Paul
- Department of Immunology, Weizmann Institute of Science, Rehovot 76100, Israel
| | - Ido Amit
- Department of Immunology, Weizmann Institute of Science, Rehovot 76100, Israel.
| |
Collapse
|
39
|
Determining Lineage Pathways from Cellular Barcoding Experiments. Cell Rep 2014; 6:617-24. [DOI: 10.1016/j.celrep.2014.01.016] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2013] [Revised: 12/09/2013] [Accepted: 01/14/2014] [Indexed: 11/21/2022] Open
|
40
|
Merad M, Sathe P, Helft J, Miller J, Mortha A. The dendritic cell lineage: ontogeny and function of dendritic cells and their subsets in the steady state and the inflamed setting. Annu Rev Immunol 2013; 31:563-604. [PMID: 23516985 DOI: 10.1146/annurev-immunol-020711-074950] [Citation(s) in RCA: 1752] [Impact Index Per Article: 146.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Dendritic cells (DCs) form a remarkable cellular network that shapes adaptive immune responses according to peripheral cues. After four decades of research, we now know that DCs arise from a hematopoietic lineage distinct from other leukocytes, establishing the DC system as a unique hematopoietic branch. Recent work has also established that tissue DCs consist of developmentally and functionally distinct subsets that differentially regulate T lymphocyte function. This review discusses major advances in our understanding of the regulation of DC lineage commitment, differentiation, diversification, and function in situ.
Collapse
Affiliation(s)
- Miriam Merad
- Department of Oncological Sciences, Mount Sinai Medical School, New York, NY 10029, USA.
| | | | | | | | | |
Collapse
|
41
|
Saito Y, Boddupalli CS, Borsotti C, Manz MG. Dendritic cell homeostasis is maintained by nonhematopoietic and T-cell-produced Flt3-ligand in steady state and during immune responses. Eur J Immunol 2013; 43:1651-8. [PMID: 23519969 DOI: 10.1002/eji.201243163] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2012] [Revised: 02/01/2013] [Accepted: 03/18/2013] [Indexed: 12/19/2022]
Abstract
Lymphoid-tissue dendritic cells (DCs) are short-lived and need to be continuously replenished from bone marrow-derived DC progenitor cells. Fms-related tyrosine kinase 3 is expressed during cellular development from hematopoietic progenitors to lymphoid-tissue DCs. Fms-related tyrosine kinase 3 ligand (Flt3L) is an essential, nonredundant cytokine for DC progenitor to lymphoid tissue DC differentiation and maintenance. However, which cells contribute to Flt3L production and how Flt3L cytokine levels are regulated in steady state and during immune reactions remains to be determined. Here we demonstrate that besides nonhematopoietic cells, WT T cells produce Flt3L and contribute to the generation of both classical DCs (cDCs) and plasmacytoid DCs in Flt3L(-/-) mice. Upon stimulation in vitro, CD4(+) T cells produce more Flt3L than CD8(+) T cells. Moreover, in vivo stimulation of naïve OT-II CD4(+) T cells with OVA leads to increase of pre-cDCs and cDCs in draining lymph nodes of Flt3L(-/-) mice in a partially Flt3L-dependent manner. Thus, Flt3L-mediated lymphoid tissue DC homeostasis is regulated by steady-state T cells as well as by proliferative T cells, fostering local development of lymphoid organ resident DCs.
Collapse
Affiliation(s)
- Yasuyuki Saito
- Division of Hematology, University Hospital Zurich, Zurich, Switzerland
| | | | | | | |
Collapse
|
42
|
Bone marrow dendritic cells are reduced in patients with high-risk myelodysplastic syndromes. Leuk Res 2013; 37:266-73. [DOI: 10.1016/j.leukres.2012.10.010] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2012] [Revised: 10/14/2012] [Accepted: 10/15/2012] [Indexed: 12/19/2022]
|
43
|
Moore AJ, Anderson MK. Dendritic cell development: a choose-your-own-adventure story. Adv Hematol 2013; 2013:949513. [PMID: 23476654 PMCID: PMC3588201 DOI: 10.1155/2013/949513] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2012] [Accepted: 12/27/2012] [Indexed: 12/13/2022] Open
Abstract
Dendritic cells (DCs) are essential components of the immune system and contribute to immune responses by activating or tolerizing T cells. DCs comprise a heterogeneous mixture of subsets that are located throughout the body and possess distinct and specialized functions. Although numerous defined precursors from the bone marrow and spleen have been identified, emerging data in the field suggests many alternative routes of DC differentiation from precursors with multilineage potential. Here, we discuss how the combinatorial expression of transcription factors can promote one DC lineage over another as well as the integration of cytokine signaling in this process.
Collapse
Affiliation(s)
- Amanda J. Moore
- Division of Biological Sciences, Sunnybrook Research Institute, 2075 Bayview Avenue, Toronto, ON, Canada M4N 3M5
- Department of Immunology, University of Toronto, Toronto, ON, Canada M5S 1A8
| | - Michele K. Anderson
- Division of Biological Sciences, Sunnybrook Research Institute, 2075 Bayview Avenue, Toronto, ON, Canada M4N 3M5
- Department of Immunology, University of Toronto, Toronto, ON, Canada M5S 1A8
| |
Collapse
|
44
|
Gwin K, Dolence JJ, Shapiro MB, Medina KL. Differential requirement for Hoxa9 in the development and differentiation of B, NK, and DC-lineage cells from Flt3+ multipotential progenitors. BMC Immunol 2013; 14:5. [PMID: 23363389 PMCID: PMC3565875 DOI: 10.1186/1471-2172-14-5] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2012] [Accepted: 01/25/2013] [Indexed: 12/13/2022] Open
Abstract
Hoxa9 is a homeodomain transcription factor important for the generation of Flt3+hiIL-7R- lymphoid biased-multipotential progenitors, Flt3+IL-7R+ common lymphoid progenitors (CLPs), and B cell precursors (BCP) in bone marrow (BM). In addition to B-cell, Flt3+IL-7R+ CLPs possess NK and DC developmental potentials, although DCs arise from Flt3+IL-7R- myeloid progenitors as well. In this study, we investigated the requirement for Hoxa9, from Flt3+ or Flt3- progenitor subsets, in the development of NK and DC lineage cells in BM. Flt3+IL-7R+Ly6D- CLPs and their Flt3+IL-7R+Ly6D+ B lineage-restricted progeny (BLP) were significantly reduced in hoxa9−/− mice. Interestingly, the reduction in Flt3+IL-7R+ CLPs in hoxa9−/− mice had no impact on the generation of NK precursor (NKP) subsets, the differentiation of NKP into mature NK cells, or NK homeostasis. Similarly, percentages and numbers of common dendritic progenitors (CDP), as well as their plasmacytoid or conventional dendritic cell progeny in hoxa9−/− mice were comparable to wildtype. These findings reveal distinct requirements for Hoxa9 or Hoxa9/Flt3 molecular circuits in regulation of B versus NK and DC development in BM.
Collapse
Affiliation(s)
- Kimberly Gwin
- Department of Immunology, College of Medicine, Mayo Clinic, 200 First Street SW, Rochester, MN 55905, USA
| | | | | | | |
Collapse
|
45
|
Kim SJ, Gregersen PK, Diamond B. Regulation of dendritic cell activation by microRNA let-7c and BLIMP1. J Clin Invest 2013; 123:823-33. [PMID: 23298838 DOI: 10.1172/jci64712] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2012] [Accepted: 11/08/2012] [Indexed: 01/02/2023] Open
Abstract
Mice with a DC-specific deletion of the transcriptional repressor B lymphocyte-induced maturation protein-1 (Blimp1) exhibit a lupus-like phenotype, secondary to enhanced DC production of IL-6. Here we explored further phenotypic changes in Blimp1-deficient DCs, the molecular mechanism underlying these changes, and their relevance to human disease. Blimp1-deficient DCs exhibited elevated expression of MHC II, and exposure to TLR agonists increased secretion of proinflammatory cytokines. This phenotype reflects enhanced expression of the microRNA let-7c, which is regulated by BLIMP1. Let-7c reciprocally inhibited Blimp1 and also blocked LPS-induced suppressor of cytokine signaling-1 (SOCS1) expression, contributing to the proinflammatory phenotype of Blimp1-deficient DCs. DCs from Blimp1 SLE-risk allele carriers exhibited analogous phenotypic changes, including decreased BLIMP1 expression, increased let-7c expression, and increased expression of proinflammatory cytokines. These results suggest that let-7c regulates DC phenotype and confirm the importance of BLIMP1 in maintaining tolerogenic DCs in both mice and humans.
Collapse
Affiliation(s)
- Sun Jung Kim
- Center for Autoimmune and Musculoskeletal Diseases, The Feinstein Institute for Medical Research (FIMR), Manhasset, New York, USA
| | | | | |
Collapse
|
46
|
Rahman AH, Aloman C. Dendritic cells and liver fibrosis. Biochim Biophys Acta Mol Basis Dis 2013; 1832:998-1004. [PMID: 23313573 DOI: 10.1016/j.bbadis.2013.01.005] [Citation(s) in RCA: 41] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2012] [Revised: 12/31/2012] [Accepted: 01/02/2013] [Indexed: 12/23/2022]
Abstract
Dendritic cells are a relative rare population of specialized antigen presenting cells that are distributed through most lymphoid and non-lymphoid tissues and play a critical role in linking the innate and adaptive arms of the immune system. The liver contains a heterogeneous population of dendritic cells that may contribute to liver inflammation and fibrosis through a number of mechanisms. This review summarizes current knowledge on the development and characterization of liver dendritic cells and their potential impact on liver fibrosis. This article is part of a Special Issue entitled: Fibrosis: Translation of basic research to human disease.
Collapse
Affiliation(s)
- Adeeb H Rahman
- Division of Liver Diseases, Mount Sinai School of Medicine, New York, NY, USA
| | | |
Collapse
|
47
|
Shortman K, Sathe P, Vremec D, Naik S, O’Keeffe M. Plasmacytoid Dendritic Cell Development. Adv Immunol 2013; 120:105-26. [DOI: 10.1016/b978-0-12-417028-5.00004-1] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
|
48
|
Abstract
Dendritic cells (DCs) drive both adaptive and innate immunity. Recent findings support the notion that distinct subsets of classical DCs favor alternative modules of immunity, acting on innate lymphoid-like cells (ILCs) and T cells similarly to promote either ILC1/Th1/CTL- or ILC3/Th17-type responses. Coordination between DC subsets and their favored immune module might imply that the genetic programs for DC diversification preceded the emergence of recombination-activating gene-dependent adaptive immunity and operate initially in coordinating ILC repertoires for appropriate responses against pathogens. Consequently, understanding the molecular basis of DC developmental and diversification is important for an underlying appreciation of immune regulation. Currently, the basis for DC development into the recognized subsets/lineages is only partially understood, based on the requirements for several transcription factors including PU.1, Bcl11a, Irf8, E2-2, Id2, Irf4, Irf8, Batf3, and other BATF family members. This chapter will briefly review recent transcriptional aspects of DC development and function and then highlight some currently unresolved questions.
Collapse
Affiliation(s)
- Kenneth M Murphy
- School of Medicine, Department of Pathology and Immunology, Washington University, St. Louis, Missouri, USA; School of Medicine, Howard Hughes Medical Institute, Washington University, St. Louis, Missouri, USA.
| |
Collapse
|
49
|
Satpathy AT, Wu X, Albring JC, Murphy KM. Re(de)fining the dendritic cell lineage. Nat Immunol 2012; 13:1145-54. [PMID: 23160217 DOI: 10.1038/ni.2467] [Citation(s) in RCA: 333] [Impact Index Per Article: 25.6] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2012] [Accepted: 09/27/2012] [Indexed: 12/12/2022]
Abstract
Dendritic cells (DCs) are essential mediators of innate and adaptive immune responses. Study of these critical cells has been complicated by their similarity to other hematopoietic lineages, particularly monocytes and macrophages. Progress has been made in three critical areas of DC biology: the characterization of lineage-restricted progenitors in the bone marrow, the identification of cytokines and transcription factors required during differentiation, and the development of genetic tools for the visualization and depletion of DCs in vivo. Collectively, these advances have clarified the nature of the DC lineage and have provided novel insights into their function during health and disease.
Collapse
Affiliation(s)
- Ansuman T Satpathy
- Department of Pathology and Immunology, Washington University in St. Louis, School of Medicine, St. Louis, Missouri, USA
| | | | | | | |
Collapse
|
50
|
Abstract
The mouse thymus supports T-cell development, but also contains non-T-cell lineages such as dendritic cells, macrophages, and granulocytes that are necessary for T-cell repertoire selection and apoptotic thymocyte clearance. Early thymic progenitors (ETPs) are not committed to the T-cell lineage, as demonstrated by both in vitro and in vivo assays. Whether ETPs realize non-T-cell lineage potentials in vivo is not well understood and indeed is controversial. In the present study, we investigated whether ETPs are the major precursors of any non-T-lineage cells in the thymus. We analyzed the development of these populations under experimental circumstances in which ETPs are nearly absent due to either abrogated thymic settling or inhibition of early thymic development by genetic ablation of IL-7 receptorα or Hes1. Results obtained using multiple in vivo approaches indicate that the majority of thymic granulocytes derive from ETPs. These data indicate that myelolymphoid progenitors settle the thymus and thus clarify the pathways by which stem cells give rise to downstream blood cell lineages.
Collapse
|