1
|
Ruiz Pérez M, Vandenabeele P, Tougaard P. The thymus road to a T cell: migration, selection, and atrophy. Front Immunol 2024; 15:1443910. [PMID: 39257583 PMCID: PMC11384998 DOI: 10.3389/fimmu.2024.1443910] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2024] [Accepted: 08/08/2024] [Indexed: 09/12/2024] Open
Abstract
The thymus plays a pivotal role in generating a highly-diverse repertoire of T lymphocytes while preventing autoimmunity. Thymus seeding progenitors (TSPs) are a heterogeneous group of multipotent progenitors that migrate to the thymus via CCR7 and CCR9 receptors. While NOTCH guides thymus progenitors toward T cell fate, the absence or disruption of NOTCH signaling renders the thymus microenvironment permissive to other cell fates. Following T cell commitment, developing T cells undergo multiple selection checkpoints by engaging with the extracellular matrix, and interacting with thymic epithelial cells (TECs) and other immune subsets across the different compartments of the thymus. The different selection checkpoints assess the T cell receptor (TCR) performance, with failure resulting in either repurposing (agonist selection), or cell death. Additionally, environmental cues such as inflammation and endocrine signaling induce acute thymus atrophy, contributing to the demise of most developing T cells during thymic selection. We discuss the occurrence of acute thymus atrophy in response to systemic inflammation. The thymus demonstrates high plasticity, shaping inflammation by abrogating T cell development and undergoing profound structural changes, and facilitating regeneration and restoration of T cell development once inflammation is resolved. Despite the challenges, thymic selection ensures a highly diverse T cell repertoire capable of discerning between self and non-self antigens, ultimately egressing to secondary lymphoid organs where they complete their maturation and exert their functions.
Collapse
Affiliation(s)
- Mario Ruiz Pérez
- Molecular Signaling and Cell Death Unit, VIB-UGent, Center for Inflammation Research, Flanders Institute for Biotechnology, Ghent, Belgium
- Department of Biomedical Molecular Biology, Ghent University, Ghent, Belgium
| | - Peter Vandenabeele
- Molecular Signaling and Cell Death Unit, VIB-UGent, Center for Inflammation Research, Flanders Institute for Biotechnology, Ghent, Belgium
- Department of Biomedical Molecular Biology, Ghent University, Ghent, Belgium
| | - Peter Tougaard
- Molecular Signaling and Cell Death Unit, VIB-UGent, Center for Inflammation Research, Flanders Institute for Biotechnology, Ghent, Belgium
- Department of Biomedical Molecular Biology, Ghent University, Ghent, Belgium
- Laboratory of Immunoregulation and Mucosal Immunology, VIB-UGent Center for Inflammation Research, Ghent, Belgium
| |
Collapse
|
2
|
Ruiz Pérez M, Maueröder C, Steels W, Verstraeten B, Lameire S, Xie W, Wyckaert L, Huysentruyt J, Divert T, Roelandt R, Gonçalves A, De Rycke R, Ravichandran K, Lambrecht BN, Taghon T, Leclercq G, Vandenabeele P, Tougaard P. TL1A and IL-18 synergy promotes GM-CSF-dependent thymic granulopoiesis in mice. Cell Mol Immunol 2024; 21:807-825. [PMID: 38839915 PMCID: PMC11291760 DOI: 10.1038/s41423-024-01180-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2023] [Accepted: 04/27/2024] [Indexed: 06/07/2024] Open
Abstract
Acute systemic inflammation critically alters the function of the immune system, often promoting myelopoiesis at the expense of lymphopoiesis. In the thymus, systemic inflammation results in acute thymic atrophy and, consequently, impaired T-lymphopoiesis. The mechanism by which systemic inflammation impacts the thymus beyond suppressing T-cell development is still unclear. Here, we describe how the synergism between TL1A and IL-18 suppresses T-lymphopoiesis to promote thymic myelopoiesis. The protein levels of these two cytokines were elevated in the thymus during viral-induced thymus atrophy infection with murine cytomegalovirus (MCMV) or pneumonia virus of mice (PVM). In vivo administration of TL1A and IL-18 induced acute thymic atrophy, while thymic neutrophils expanded. Fate mapping with Ms4a3-Cre mice demonstrated that thymic neutrophils emerge from thymic granulocyte-monocyte progenitors (GMPs), while Rag1-Cre fate mapping revealed a common developmental path with lymphocytes. These effects could be modeled ex vivo using neonatal thymic organ cultures (NTOCs), where TL1A and IL-18 synergistically enhanced neutrophil production and egress. NOTCH blockade by the LY411575 inhibitor increased the number of neutrophils in the culture, indicating that NOTCH restricted steady-state thymic granulopoiesis. To promote myelopoiesis, TL1A, and IL-18 synergistically increased GM-CSF levels in the NTOC, which was mainly produced by thymic ILC1s. In support, TL1A- and IL-18-induced granulopoiesis was completely prevented in NTOCs derived from Csf2rb-/- mice and by GM-CSFR antibody blockade, revealing that GM-CSF is the essential factor driving thymic granulopoiesis. Taken together, our findings reveal that TL1A and IL-18 synergism induce acute thymus atrophy while promoting extramedullary thymic granulopoiesis in a NOTCH and GM-CSF-controlled manner.
Collapse
Affiliation(s)
- Mario Ruiz Pérez
- Molecular Signaling and Cell Death Unit, VIB-UGent Center for Inflammation Research, Flanders Institute for Biotechnology, Ghent, Belgium
- Department of Biomedical Molecular Biology, Ghent University, Ghent, Belgium
| | - Christian Maueröder
- Department of Biomedical Molecular Biology, Ghent University, Ghent, Belgium
- Cell Clearance in Health and Disease Lab, VIB-UGent Center for Inflammation Research, Flanders Institute for Biotechnology, Ghent, Belgium
| | - Wolf Steels
- Molecular Signaling and Cell Death Unit, VIB-UGent Center for Inflammation Research, Flanders Institute for Biotechnology, Ghent, Belgium
- Department of Biomedical Molecular Biology, Ghent University, Ghent, Belgium
| | - Bruno Verstraeten
- Molecular Signaling and Cell Death Unit, VIB-UGent Center for Inflammation Research, Flanders Institute for Biotechnology, Ghent, Belgium
- Department of Biomedical Molecular Biology, Ghent University, Ghent, Belgium
| | - Sahine Lameire
- Department of Biomedical Molecular Biology, Ghent University, Ghent, Belgium
- Laboratory of Immunoregulation and Mucosal Immunology, VIB-UGent Center for Inflammation Research, Ghent, Belgium
- Department of Internal Medicine and Pediatrics, Ghent University, Ghent, Belgium
| | - Wei Xie
- Molecular Signaling and Cell Death Unit, VIB-UGent Center for Inflammation Research, Flanders Institute for Biotechnology, Ghent, Belgium
- Department of Biomedical Molecular Biology, Ghent University, Ghent, Belgium
| | - Laura Wyckaert
- Molecular Signaling and Cell Death Unit, VIB-UGent Center for Inflammation Research, Flanders Institute for Biotechnology, Ghent, Belgium
- Department of Biomedical Molecular Biology, Ghent University, Ghent, Belgium
| | - Jelle Huysentruyt
- Molecular Signaling and Cell Death Unit, VIB-UGent Center for Inflammation Research, Flanders Institute for Biotechnology, Ghent, Belgium
- Department of Biomedical Molecular Biology, Ghent University, Ghent, Belgium
| | - Tatyana Divert
- Molecular Signaling and Cell Death Unit, VIB-UGent Center for Inflammation Research, Flanders Institute for Biotechnology, Ghent, Belgium
- Department of Biomedical Molecular Biology, Ghent University, Ghent, Belgium
| | - Ria Roelandt
- Molecular Signaling and Cell Death Unit, VIB-UGent Center for Inflammation Research, Flanders Institute for Biotechnology, Ghent, Belgium
- VIB Single Cell Facility, Flanders Institute for Biotechnology, Ghent, Belgium
| | - Amanda Gonçalves
- Department of Biomedical Molecular Biology, Ghent University, Ghent, Belgium
- VIB BioImaging Core, VIB-UGent Center for Inflammation Research, Technologiepark-Zwijnaarde 71, Ghent, 9052, Belgium
| | - Riet De Rycke
- Department of Biomedical Molecular Biology, Ghent University, Ghent, Belgium
- VIB BioImaging Core, VIB-UGent Center for Inflammation Research, Technologiepark-Zwijnaarde 71, Ghent, 9052, Belgium
| | - Kodi Ravichandran
- Department of Biomedical Molecular Biology, Ghent University, Ghent, Belgium
- Cell Clearance in Health and Disease Lab, VIB-UGent Center for Inflammation Research, Flanders Institute for Biotechnology, Ghent, Belgium
- Department of Microbiology, Immunology, and Cancer Biology, University of Virginia, Charlottesville, VA, USA
- Division of Immunobiology, Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO, USA
| | - Bart N Lambrecht
- Laboratory of Immunoregulation and Mucosal Immunology, VIB-UGent Center for Inflammation Research, Ghent, Belgium
- Department of Internal Medicine and Pediatrics, Ghent University, Ghent, Belgium
- Department of Pulmonary Medicine, Erasmus MC, Rotterdam, The Netherlands
| | - Tom Taghon
- Cancer Research Institute Ghent, Ghent, Belgium
- Department of Diagnostic Sciences, Ghent University, Ghent, Belgium
| | - Georges Leclercq
- Cancer Research Institute Ghent, Ghent, Belgium
- Department of Diagnostic Sciences, Ghent University, Ghent, Belgium
| | - Peter Vandenabeele
- Molecular Signaling and Cell Death Unit, VIB-UGent Center for Inflammation Research, Flanders Institute for Biotechnology, Ghent, Belgium.
- Department of Biomedical Molecular Biology, Ghent University, Ghent, Belgium.
| | - Peter Tougaard
- Molecular Signaling and Cell Death Unit, VIB-UGent Center for Inflammation Research, Flanders Institute for Biotechnology, Ghent, Belgium.
- Department of Biomedical Molecular Biology, Ghent University, Ghent, Belgium.
| |
Collapse
|
3
|
Sado T, Cart JB, Lee CL. Mechanisms Underlying the Development of Murine T-Cell Lymphoblastic Lymphoma/Leukemia Induced by Total-Body Irradiation. Cancers (Basel) 2024; 16:2224. [PMID: 38927929 PMCID: PMC11201593 DOI: 10.3390/cancers16122224] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2024] [Revised: 06/11/2024] [Accepted: 06/12/2024] [Indexed: 06/28/2024] Open
Abstract
Exposure to ionizing radiation is associated with an increased risk of hematologic malignancies in myeloid and lymphoid lineages in humans and experimental mice. Given that substantial evidence links radiation exposure with the risk of hematologic malignancies, it is imperative to deeply understand the mechanisms underlying cellular and molecular changes during the latency period between radiation exposure and the emergence of fully transformed malignant cells. One experimental model widely used in the field of radiation and cancer biology to study hematologic malignancies induced by radiation exposure is mouse models of radiation-induced thymic lymphoma. Murine radiation-induced thymic lymphoma is primarily driven by aberrant activation of Notch signaling, which occurs frequently in human precursor T-cell lymphoblastic lymphoma (T-LBL) and T-cell lymphoblastic leukemia (T-ALL). Here, we summarize the literature elucidating cell-autonomous and non-cell-autonomous mechanisms underlying cancer initiation, progression, and malignant transformation in the thymus following total-body irradiation (TBI) in mice.
Collapse
Affiliation(s)
- Toshihiko Sado
- National Institute of Radiological Sciences, Chiba 263-0024, Japan
| | - John B. Cart
- Department of Radiation Oncology, Duke University School of Medicine, Durham, NC 27710, USA
- Department of Pathology, Duke University School of Medicine, Durham, NC 27710, USA
| | - Chang-Lung Lee
- Department of Radiation Oncology, Duke University School of Medicine, Durham, NC 27710, USA
- Department of Pathology, Duke University School of Medicine, Durham, NC 27710, USA
| |
Collapse
|
4
|
Zhang Y, Wu T, He Z, Lai W, Shen X, Lv J, Wang Y, Wu L. Regulation of pDC fate determination by histone deacetylase 3. eLife 2023; 12:e80477. [PMID: 38011375 PMCID: PMC10732571 DOI: 10.7554/elife.80477] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2022] [Accepted: 11/22/2023] [Indexed: 11/29/2023] Open
Abstract
Dendritic cells (DCs), the key antigen-presenting cells, are primary regulators of immune responses. Transcriptional regulation of DC development had been one of the major research interests in DC biology; however, the epigenetic regulatory mechanisms during DC development remains unclear. Here, we report that Histone deacetylase 3 (Hdac3), an important epigenetic regulator, is highly expressed in pDCs, and its deficiency profoundly impaired the development of pDCs. Significant disturbance of homeostasis of hematopoietic progenitors was also observed in HDAC3-deficient mice, manifested by altered cell numbers of these progenitors and defective differentiation potentials for pDCs. Using the in vitro Flt3L supplemented DC culture system, we further demonstrated that HDAC3 was required for the differentiation of pDCs from progenitors at all developmental stages. Mechanistically, HDAC3 deficiency resulted in enhanced expression of cDC1-associated genes, owing to markedly elevated H3K27 acetylation (H3K27ac) at these gene sites in BM pDCs. In contrast, the expression of pDC-associated genes was significantly downregulated, leading to defective pDC differentiation.
Collapse
Affiliation(s)
- Yijun Zhang
- Institute for Immunology, Tsinghua-Peking Center for Life Sciences, School of Medicine, Tsinghua UniversityBeijingChina
- Beijing Key Laboratory for Immunological Research on Chronic DiseasesBeijingChina
| | - Tao Wu
- Institute for Immunology, Tsinghua-Peking Center for Life Sciences, School of Medicine, Tsinghua UniversityBeijingChina
- Beijing Key Laboratory for Immunological Research on Chronic DiseasesBeijingChina
| | - Zhimin He
- Institute for Immunology, Tsinghua-Peking Center for Life Sciences, School of Medicine, Tsinghua UniversityBeijingChina
- Beijing Key Laboratory for Immunological Research on Chronic DiseasesBeijingChina
| | - Wenlong Lai
- Institute for Immunology, Tsinghua-Peking Center for Life Sciences, School of Medicine, Tsinghua UniversityBeijingChina
- Beijing Key Laboratory for Immunological Research on Chronic DiseasesBeijingChina
| | - Xiangyi Shen
- Institute for Immunology, Tsinghua-Peking Center for Life Sciences, School of Medicine, Tsinghua UniversityBeijingChina
- Beijing Key Laboratory for Immunological Research on Chronic DiseasesBeijingChina
| | - Jiaoyan Lv
- Institute for Immunology, Tsinghua-Peking Center for Life Sciences, School of Medicine, Tsinghua UniversityBeijingChina
- Beijing Key Laboratory for Immunological Research on Chronic DiseasesBeijingChina
| | - Yuanhao Wang
- Institute for Immunology, Tsinghua-Peking Center for Life Sciences, School of Medicine, Tsinghua UniversityBeijingChina
- Beijing Key Laboratory for Immunological Research on Chronic DiseasesBeijingChina
| | - Li Wu
- Institute for Immunology, Tsinghua-Peking Center for Life Sciences, School of Medicine, Tsinghua UniversityBeijingChina
- Beijing Key Laboratory for Immunological Research on Chronic DiseasesBeijingChina
| |
Collapse
|
5
|
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
|
6
|
Clonal Analysis of Human Dendritic Cell Progenitors Using a Stromal Cell Culture. Methods Mol Biol 2023; 2618:155-170. [PMID: 36905516 DOI: 10.1007/978-1-0716-2938-3_12] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/12/2023]
Abstract
Dendritic cells (DCs) are a heterogenous population of professional antigen-presenting cells that play an "educator" role in immunity. Multiple DC subsets collaboratively initiate and orchestrate innate and adaptive immune responses. Recent advances in our ability to investigate cellular transcription, signaling, and function at the single-cell level have opened opportunities to examine heterogeneous populations at unprecedented resolutions. Culturing of mouse DC subsets from single bone marrow hematopoietic progenitor cells, that is, clonal analysis, has enabled identification of multiple progenitors with distinct potentials and furthered understanding of mouse DC development. However, studies of human DC development have been hampered by the lack of a corresponding system to generate multiple human DC subsets. Here, we describe a protocol to functionally profile the differentiation potentials of single human hematopoietic stem and progenitor cells (HSPCs) to multiple DC subsets, myeloid and lymphoid cells that will facilitate investigation of human DC lineage specification and reveal its molecular bases.
Collapse
|
7
|
Anderson DA, Ou F, Kim S, Murphy TL, Murphy KM. Transition from cMyc to L-Myc during dendritic cell development coordinated by rising levels of IRF8. J Exp Med 2022; 219:e20211483. [PMID: 34958351 PMCID: PMC8713298 DOI: 10.1084/jem.20211483] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2021] [Revised: 10/25/2021] [Accepted: 12/02/2021] [Indexed: 01/01/2023] Open
Abstract
During dendritic cell (DC) development, Myc expression in progenitors is replaced by Mycl in mature DCs, but when and how this transition occurs is unknown. We evaluated DC development using reporters for MYC, MYCL, and cell cycle proteins Geminin and CDT1 in wild-type and various mutant mice. For classical type 1 dendritic cells (cDC1s) and plasmacytoid DCs (pDCs), the transition occurred upon their initial specification from common dendritic cell progenitors (CDPs) or common lymphoid progenitors (CLPs), respectively. This transition required high levels of IRF8 and interaction with PU.1, suggesting the use of EICEs within Mycl enhancers. In pDCs, maximal MYCL induction also required the +41kb Irf8 enhancer that controls pDC IRF8 expression. IRF8 also contributed to repression of MYC. While MYC is expressed only in rapidly dividing DC progenitors, MYCL is most highly expressed in DCs that have exited the cell cycle. Thus, IRF8 levels coordinate the Myc-Mycl transition during DC development.
Collapse
Affiliation(s)
| | | | | | | | - Kenneth M. Murphy
- Department of Pathology and Immunology, Washington University in St. Louis, School of Medicine, St. Louis, MO
| |
Collapse
|
8
|
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
|
9
|
A regulatory network of microRNAs confers lineage commitment during early developmental trajectories of B and T lymphocytes. Proc Natl Acad Sci U S A 2021; 118:2104297118. [PMID: 34750254 DOI: 10.1073/pnas.2104297118] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/10/2021] [Indexed: 11/18/2022] Open
Abstract
The commitment of hematopoietic multipotent progenitors (MPPs) toward a particular lineage involves activation of cell type-specific genes and silencing of genes that promote alternate cell fates. Although the gene expression programs of early-B and early-T lymphocyte development are mutually exclusive, we show that these cell types exhibit significantly correlated microRNA (miRNA) profiles. However, their corresponding miRNA targetomes are distinct and predominated by transcripts associated with natural killer, dendritic cell, and myeloid lineages, suggesting that miRNAs function in a cell-autonomous manner. The combinatorial expression of miRNAs miR-186-5p, miR-128-3p, and miR-330-5p in MPPs significantly attenuates their myeloid differentiation potential due to repression of myeloid-associated transcripts. Depletion of these miRNAs caused a pronounced de-repression of myeloid lineage targets in differentiating early-B and early-T cells, resulting in a mixed-lineage gene expression pattern. De novo motif analysis combined with an assay of promoter activities indicates that B as well as T lineage determinants drive the expression of these miRNAs in lymphoid lineages. Collectively, we present a paradigm that miRNAs are conserved between developing B and T lymphocytes, yet they target distinct sets of promiscuously expressed lineage-inappropriate genes to suppress the alternate cell-fate options. Thus, our studies provide a comprehensive compendium of miRNAs with functional implications for B and T lymphocyte development.
Collapse
|
10
|
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: 12] [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
|
11
|
Balhara J, Koussih L, Mohammed A, Shan L, Lamkhioued B, Gounni AS. PTX3 Deficiency Promotes Enhanced Accumulation and Function of CD11c +CD11b + DCs in a Murine Model of Allergic Inflammation. Front Immunol 2021; 12:641311. [PMID: 34305885 PMCID: PMC8299994 DOI: 10.3389/fimmu.2021.641311] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2020] [Accepted: 06/21/2021] [Indexed: 11/17/2022] Open
Abstract
PTX3 is a unique member of the long pentraxins family and plays an indispensable role in regulating the immune system. We previously showed that PTX3 deletion aggravates allergic inflammation via a Th17 -dominant phenotype and enhanced CD4 T cell survival using a murine model of ovalbumin (OVA) induced allergic inflammation. In this study, we identified that upon OVA exposure, increased infiltration of CD11c+CD11b+ dendritic cells (DCs) was observed in the lungs of PTX3-/- mice compared to wild type littermate. Further analysis showed that a short-term OVA exposure led to an increased number of bone marrow common myeloid progenitors (CMP) population concomitantly with increased Ly6Chigh CCR2high monocytes and CD11c+CD11b+ DCs in the lungs. Also, pulmonary CD11c+CD11b+ DCs from OVA-exposed PTX3-/- mice exhibited enhanced expression of maturation markers, chemokines receptors CCR2, and increased OVA uptake and processing compared to wild type controls. Taken together, our data suggest that PTX3 deficiency heightened lung CD11c+CD11b+DC numbers and function, hence exacerbating airway inflammatory response.
Collapse
Affiliation(s)
- Jyoti Balhara
- Department of Immunology, Max-Rady College of Medicine, Rady Faculty of Health Sciences, University of Manitoba, Winnipeg, MB, Canada
| | - Latifa Koussih
- Department of Immunology, Max-Rady College of Medicine, Rady Faculty of Health Sciences, University of Manitoba, Winnipeg, MB, Canada.,Department des Sciences Experimentales, Université de Saint-Boniface, Winnipeg, MB, Canada
| | - Ashfaque Mohammed
- Department of Immunology, Max-Rady College of Medicine, Rady Faculty of Health Sciences, University of Manitoba, Winnipeg, MB, Canada
| | - Lianyu Shan
- Department of Immunology, Max-Rady College of Medicine, Rady Faculty of Health Sciences, University of Manitoba, Winnipeg, MB, Canada
| | - Bouchaib Lamkhioued
- Laboratoire d'Immunologie et de Biotechnologies, EA7509-IRMAIC, Pôle-Santé, Université de Reims Champagne-Ardenne, Reims, France
| | - Abdelilah S Gounni
- Department of Immunology, Max-Rady College of Medicine, Rady Faculty of Health Sciences, University of Manitoba, Winnipeg, MB, Canada
| |
Collapse
|
12
|
IDH1 mutation contributes to myeloid dysplasia in mice by disturbing heme biosynthesis and erythropoiesis. Blood 2021; 137:945-958. [PMID: 33254233 DOI: 10.1182/blood.2020007075] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2020] [Accepted: 11/18/2020] [Indexed: 12/22/2022] Open
Abstract
Isocitrate dehydrogenase (IDH) mutations are common genetic alterations in myeloid disorders, including acute myeloid leukemia (AML) and myelodysplastic syndrome (MDS). Epigenetic changes, including abnormal histone and DNA methylation, have been implicated in the pathogenic build-up of hematopoietic progenitors, but it is still unclear whether and how IDH mutations themselves affect hematopoiesis. Here, we show that IDH1-mutant mice develop myeloid dysplasia in that these animals exhibit anemia, ineffective erythropoiesis, and increased immature progenitors and erythroblasts. In erythroid cells of these mice, D-2-hydroxyglutarate, an aberrant metabolite produced by the mutant IDH1 enzyme, inhibits oxoglutarate dehydrogenase activity and diminishes succinyl-coenzyme A (CoA) production. This succinyl-CoA deficiency attenuates heme biosynthesis in IDH1-mutant hematopoietic cells, thus blocking erythroid differentiation at the late erythroblast stage and the erythroid commitment of hematopoietic stem cells, while the exogenous succinyl-CoA or 5-ALA rescues erythropoiesis in IDH1-mutant erythroid cells. Heme deficiency also impairs heme oxygenase-1 expression, which reduces levels of important heme catabolites such as biliverdin and bilirubin. These deficits result in accumulation of excessive reactive oxygen species that induce the cell death of IDH1-mutant erythroid cells. Our results clearly show the essential role of IDH1 in normal erythropoiesis and describe how its mutation leads to myeloid disorders. These data thus have important implications for the devising of new treatments for IDH-mutant tumors.
Collapse
|
13
|
Anderson DA, Dutertre CA, Ginhoux F, Murphy KM. Genetic models of human and mouse dendritic cell development and function. Nat Rev Immunol 2021; 21:101-115. [PMID: 32908299 PMCID: PMC10955724 DOI: 10.1038/s41577-020-00413-x] [Citation(s) in RCA: 163] [Impact Index Per Article: 54.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/28/2020] [Indexed: 12/13/2022]
Abstract
Dendritic cells (DCs) develop in the bone marrow from haematopoietic progenitors that have numerous shared characteristics between mice and humans. Human counterparts of mouse DC progenitors have been identified by their shared transcriptional signatures and developmental potential. New findings continue to revise models of DC ontogeny but it is well accepted that DCs can be divided into two main functional groups. Classical DCs include type 1 and type 2 subsets, which can detect different pathogens, produce specific cytokines and present antigens to polarize mainly naive CD8+ or CD4+ T cells, respectively. By contrast, the function of plasmacytoid DCs is largely innate and restricted to the detection of viral infections and the production of type I interferon. Here, we discuss genetic models of mouse DC development and function that have aided in correlating ontogeny with function, as well as how these findings can be translated to human DCs and their progenitors.
Collapse
Affiliation(s)
- David A Anderson
- Department of Pathology and Immunology, School of Medicine, Washington University in St. Louis, St. Louis, MO, USA
| | | | - Florent Ginhoux
- Singapore Immunology Network, Agency for Science, Technology and Research, Singapore, Singapore
- Shanghai Institute of Immunology, Department of Immunology and Microbiology, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Translational Immunology Institute, SingHealth Duke-NUS Academic Medical Centre, Singapore, Singapore
| | - Kenneth M Murphy
- Department of Pathology and Immunology, School of Medicine, Washington University in St. Louis, St. Louis, MO, USA.
- Howard Hughes Medical Institute, School of Medicine, Washington University in St. Louis, St. Louis, MO, USA.
| |
Collapse
|
14
|
Cabeza-Cabrerizo M, Cardoso A, Minutti CM, Pereira da Costa M, Reis E Sousa C. Dendritic Cells Revisited. Annu Rev Immunol 2021; 39:131-166. [PMID: 33481643 DOI: 10.1146/annurev-immunol-061020-053707] [Citation(s) in RCA: 357] [Impact Index Per Article: 119.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Dendritic cells (DCs) possess the ability to integrate information about their environment and communicate it to other leukocytes, shaping adaptive and innate immunity. Over the years, a variety of cell types have been called DCs on the basis of phenotypic and functional attributes. Here, we refocus attention on conventional DCs (cDCs), a discrete cell lineage by ontogenetic and gene expression criteria that best corresponds to the cells originally described in the 1970s. We summarize current knowledge of mouse and human cDC subsets and describe their hematopoietic development and their phenotypic and functional attributes. We hope that our effort to review the basic features of cDC biology and distinguish cDCs from related cell types brings to the fore the remarkable properties of this cell type while shedding some light on the seemingly inordinate complexity of the DC field.
Collapse
Affiliation(s)
- Mar Cabeza-Cabrerizo
- Immunobiology Laboratory, The Francis Crick Institute, London NW1 1AT, United Kingdom;
| | - Ana Cardoso
- Immunobiology Laboratory, The Francis Crick Institute, London NW1 1AT, United Kingdom;
| | - Carlos M Minutti
- Immunobiology Laboratory, The Francis Crick Institute, London NW1 1AT, United Kingdom;
| | | | - Caetano Reis E Sousa
- Immunobiology Laboratory, The Francis Crick Institute, London NW1 1AT, United Kingdom;
| |
Collapse
|
15
|
Environmental signals rather than layered ontogeny imprint the function of type 2 conventional dendritic cells in young and adult mice. Nat Commun 2021; 12:464. [PMID: 33469015 PMCID: PMC7815729 DOI: 10.1038/s41467-020-20659-2] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2020] [Accepted: 12/13/2020] [Indexed: 01/29/2023] Open
Abstract
Conventional dendritic cells (cDC) are key activators of naive T cells, and can be targeted in adults to induce adaptive immunity, but in early life are considered under-developed or functionally immature. Here we show that, in early life, when the immune system develops, cDC2 exhibit a dual hematopoietic origin and, like other myeloid and lymphoid cells, develop in waves. Developmentally distinct cDC2 in early life, despite being distinguishable by fate mapping, are transcriptionally and functionally similar. cDC2 in early and adult life, however, are exposed to distinct cytokine environments that shape their transcriptional profile and alter their ability to sense pathogens, secrete cytokines and polarize T cells. We further show that cDC2 in early life, despite being distinct from cDC2 in adult life, are functionally competent and can induce T cell responses. Our results thus highlight the potential of harnessing cDC2 for boosting immunity in early life.
Collapse
|
16
|
Schmid H, Ribeiro EM, Secker KA, Duerr-Stoerzer S, Keppeler H, Dong R, Munz T, Schulze-Osthoff K, Hailfinger S, Schneidawind C, Schneidawind D. Human invariant natural killer T cells promote tolerance by preferential apoptosis induction of conventional dendritic cells. Haematologica 2021; 107:427-436. [PMID: 33440919 PMCID: PMC8804566 DOI: 10.3324/haematol.2020.267583] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2020] [Indexed: 11/20/2022] Open
Abstract
Graft-versus-host disease (GvHD) is a major cause of morbidity and mortality after allogeneic hematopoietic cell transplantation. We recently showed in murine studies and in vitro human models that adoptively transferred invariant natural killer T (iNKT) cells protect from GvHD and promote graft-versus-leukemia effects. The cellular mechanisms underlying GvHD prevention by iNKT cells in humans, however, remain unknown. In order to study relevant cellular interactions, dendritic cells (DC) were either generated from monocytes or isolated directly from blood of healthy donors or GvHD patients and co-cultured in a mixed lymphocyte reaction (MLR) with T cells obtained from healthy donors or transplantation bags. Addition of culture-expanded iNKT cells to the MLR-induced DC apoptosis in a cell contact-dependent manner, thereby preventing T-cell activation and proliferation. Annexin V/propidium iodide staining and image stream assays showed that CD4+CD8–, CD4–CD8+ and double negative iNKT cells are similarly able to induce DC apoptosis. Further MLR assays revealed that conventional DC (cDC) but not plasmacytoid DC (pDC) could induce alloreactive T-cell activation and proliferation. Interestingly, cDC were also more susceptible to apoptosis induced by iNKT cells, which correlates with their higher CD1d expression, leading to a bias in favor of pDC. Remarkably, these results could also be observed in GvHD patients. We propose a new mechanism how ex vivo expanded human iNKT cells prevent alloreactivity of T cells. iNKT cells modulate T-cell responses by selective apoptosis of DC subsets, resulting in suppression of T-cell activation and proliferation while enabling beneficial immune responses through pDC.
Collapse
Affiliation(s)
- Hannes Schmid
- Department of Medicine II, University Hospital Tuebingen, Eberhard Karls University, Tuebingen
| | - Emmanuelle M Ribeiro
- Department of Medicine II, University Hospital Tuebingen, Eberhard Karls University, Tuebingen
| | - Kathy-Ann Secker
- Department of Medicine II, University Hospital Tuebingen, Eberhard Karls University, Tuebingen
| | - Silke Duerr-Stoerzer
- Department of Medicine II, University Hospital Tuebingen, Eberhard Karls University, Tuebingen
| | - Hildegard Keppeler
- Department of Medicine II, University Hospital Tuebingen, Eberhard Karls University, Tuebingen
| | - Ruoyun Dong
- Department of Medicine II, University Hospital Tuebingen, Eberhard Karls University, Tuebingen
| | - Timo Munz
- Department of Medicine II, University Hospital Tuebingen, Eberhard Karls University, Tuebingen
| | | | - Stephan Hailfinger
- Interfaculty Institute of Biochemistry, Eberhard Karls University, Tuebingen
| | - Corina Schneidawind
- Department of Medicine II, University Hospital Tuebingen, Eberhard Karls University, Tuebingen
| | - Dominik Schneidawind
- Department of Medicine II, University Hospital Tuebingen, Eberhard Karls University, Tuebingen.
| |
Collapse
|
17
|
Audiger C, Lesage S. FLT3 Ligand Is Dispensable for the Final Stage of Type 1 Conventional Dendritic Cell Differentiation. THE JOURNAL OF IMMUNOLOGY 2020; 205:2117-2127. [PMID: 32887750 DOI: 10.4049/jimmunol.2000742] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/22/2020] [Accepted: 08/10/2020] [Indexed: 12/29/2022]
Abstract
Conventional dendritic cells (cDCs) are comprised of two major subsets, type 1 cDC (cDC1) and type 2 cDC (cDC2). As each cDC subset differentially influences the nature of immune responses, we sought factors that would allow the manipulation of their relative abundance. Notably, cDC1 are less abundant than cDC2 in both lymphoid and nonlymphoid organs. We demonstrate that this bias is already apparent in bone marrow precommitted precursors. However, comparison of five common inbred strains revealed a disparity in precursor-product relationship, in which mice with fewer precursors to cDC1 had more cDC1. This disparity associated with contrasting variations in CD135 (FLT3) expression on cDC subsets. Hence, we characterized the response to FLT3 ligand during cDC1 and cDC2 lineage differentiation and find that although FLT3 ligand is required throughout cDC2 differentiation, it is surprisingly dispensable during late-stage cDC1 differentiation. Overall, we find that tight regulation of FLT3 ligand levels throughout cDC differentiation dictates the cDC1 to cDC2 ratio in lymphoid organs.
Collapse
Affiliation(s)
- Cindy Audiger
- Department of Immunology-Oncology, Maisonneuve-Rosemont Hospital, Montreal, Quebec H1T 2M4, Canada; and Département de Microbiologie, Infectiologie et Immunologie, Université de Montréal, Montreal, Quebec H3C 3J7, Canada
| | - Sylvie Lesage
- Department of Immunology-Oncology, Maisonneuve-Rosemont Hospital, Montreal, Quebec H1T 2M4, Canada; and Département de Microbiologie, Infectiologie et Immunologie, Université de Montréal, Montreal, Quebec H3C 3J7, Canada
| |
Collapse
|
18
|
Naik SH. Dendritic cell development at a clonal level within a revised 'continuous' model of haematopoiesis. Mol Immunol 2020; 124:190-197. [PMID: 32593782 DOI: 10.1016/j.molimm.2020.06.012] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2020] [Revised: 04/15/2020] [Accepted: 06/11/2020] [Indexed: 12/17/2022]
Abstract
Understanding development of the dendritic cell (DC) subtypes continues to evolve. The origin and relationship of conventional DC type 1 (cDC1), cDC type 2 (cDC2) and plasmacytoid DCs (pDCs) to each other, and in relation to classic myeloid and lymphoid cells, has had a long and controversial history and is still not fully resolved. This review summarises the technological developments and findings that have been achieved at a clonal level, and how that has enhanced our knowledge of the process. It summarises the single cell lineage tracing technologies that have emerged, their application in in vitro and in vivo studies, in both mouse and human settings, and places the findings in a wider context of understanding haematopoiesis at a single cell or clonal level. In particular, it addresses the fate heterogeneity observed in many phenotypically defined progenitor subsets and how these findings have led to a departure from the classic ball-and-stick models of haematopoiesis to the emerging continuous model. Prior contradictions in DC development may be reconciled if they are framed within this revised model, where commitment to a lineage or cell type does not occur in an all-or-nothing process in defined progenitors but rather can occur at many stages of haematopoiesis in a dynamic process.
Collapse
Affiliation(s)
- Shalin H Naik
- Immunology Division, The Walter & Eliza Hall Institute of Medical Research, Parkville, Australia; The Department of Medical Biology, The University of Melbourne, Parkville, Australia.
| |
Collapse
|
19
|
Bigley V, Collin M. Insights from Patients with Dendritic Cell Immunodeficiency. Mol Immunol 2020; 122:116-123. [PMID: 32344243 DOI: 10.1016/j.molimm.2020.04.019] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2020] [Revised: 03/27/2020] [Accepted: 04/16/2020] [Indexed: 12/15/2022]
Abstract
Dendritic Cells (DCs), derived from haematopoietic stem cells, are critical to the dynamic and balanced functioning of the intact immune system and are of great interest as vehicles of immunotherapy. Genetically modified mouse models have proved powerful tools to map DC development and function in vivo but human studies have previously relied heavily on in vitro systems. Human dendritic cell immunodeficiency, resulting from single gene mutations, offers new opportunities to dissect the role of human DCs in vivo, determine the genetic requirements for their development and map their haematopoietic differentiation pathways. This review will summarise the clinical phenotypes of mutations in GATA2, IRF8 and IKZF1 genes which result in global or subset specific dendritic cell deficiencies, discuss the functional consequences of these cytopenias and how these syndromes have informed our knowledge of DC differentiation and human haematopoiesis.
Collapse
Affiliation(s)
- Venetia Bigley
- Translational and Clinical Research Institute, Newcastle University, Newcastle upon Tyne, UK; NIHR Newcastle Biomedical Research Centre at Newcastle upon Tyne Hospitals NHS Foundation Trust, Newcastle upon Tyne, UK.
| | - Matthew Collin
- Translational and Clinical Research Institute, Newcastle University, Newcastle upon Tyne, UK; NIHR Newcastle Biomedical Research Centre at Newcastle upon Tyne Hospitals NHS Foundation Trust, Newcastle upon Tyne, UK
| |
Collapse
|
20
|
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
|
21
|
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: 1.5] [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
|
22
|
Leylek R, Idoyaga J. The versatile plasmacytoid dendritic cell: Function, heterogeneity, and plasticity. INTERNATIONAL REVIEW OF CELL AND MOLECULAR BIOLOGY 2019; 349:177-211. [PMID: 31759431 DOI: 10.1016/bs.ircmb.2019.10.002] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Since their identification as the natural interferon-producing cell two decades ago, plasmacytoid dendritic cells (pDCs) have been attributed diverse functions in the immune response. Their most well characterized function is innate, i.e., their rapid and robust production of type-I interferon (IFN-I) in response to viruses. However, pDCs have also been implicated in antigen presentation, activation of adaptive immune responses and immunoregulation. The mechanisms by which pDCs enact these diverse functions are poorly understood. One central debate is whether these functions are carried out by different pDC subpopulations or by plasticity in the pDC compartment. This chapter summarizes the latest reports regarding pDC function, heterogeneity, cell conversion and environmentally influenced plasticity, as well as the role of pDCs in infection, autoimmunity and cancer.
Collapse
Affiliation(s)
- Rebecca Leylek
- Department of Microbiology and Immunology, and Immunology Program, Stanford University School of Medicine, Stanford, CA, United States
| | - Juliana Idoyaga
- Department of Microbiology and Immunology, and Immunology Program, Stanford University School of Medicine, Stanford, CA, United States.
| |
Collapse
|
23
|
Satpathy AT, Granja JM, Yost KE, Qi Y, Meschi F, McDermott GP, Olsen BN, Mumbach MR, Pierce SE, Corces MR, Shah P, Bell JC, Jhutty D, Nemec CM, Wang J, Wang L, Yin Y, Giresi PG, Chang ALS, Zheng GXY, Greenleaf WJ, Chang HY. Massively parallel single-cell chromatin landscapes of human immune cell development and intratumoral T cell exhaustion. Nat Biotechnol 2019; 37:925-936. [PMID: 31375813 PMCID: PMC7299161 DOI: 10.1038/s41587-019-0206-z] [Citation(s) in RCA: 523] [Impact Index Per Article: 104.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2019] [Accepted: 07/01/2019] [Indexed: 02/08/2023]
Abstract
Understanding complex tissues requires single-cell deconstruction of gene regulation with precision and scale. Here, we assess the performance of a massively parallel droplet-based method for mapping transposase-accessible chromatin in single cells using sequencing (scATAC-seq). We apply scATAC-seq to obtain chromatin profiles of more than 200,000 single cells in human blood and basal cell carcinoma. In blood, application of scATAC-seq enables marker-free identification of cell type-specific cis- and trans-regulatory elements, mapping of disease-associated enhancer activity and reconstruction of trajectories of cellular differentiation. In basal cell carcinoma, application of scATAC-seq reveals regulatory networks in malignant, stromal and immune cells in the tumor microenvironment. Analysis of scATAC-seq profiles from serial tumor biopsies before and after programmed cell death protein 1 blockade identifies chromatin regulators of therapy-responsive T cell subsets and reveals a shared regulatory program that governs intratumoral CD8+ T cell exhaustion and CD4+ T follicular helper cell development. We anticipate that scATAC-seq will enable the unbiased discovery of gene regulatory factors across diverse biological systems.
Collapse
Affiliation(s)
- Ansuman T Satpathy
- Center for Personal Dynamic Regulomes, Stanford University School of Medicine, Stanford, CA, USA
- Department of Pathology, Stanford University School of Medicine, Stanford, CA, USA
| | - Jeffrey M Granja
- Center for Personal Dynamic Regulomes, Stanford University School of Medicine, Stanford, CA, USA
- Department of Genetics, Stanford University School of Medicine, Stanford, CA, USA
- Biophysics Program, Stanford University School of Medicine, Stanford, CA, USA
| | - Kathryn E Yost
- Center for Personal Dynamic Regulomes, Stanford University School of Medicine, Stanford, CA, USA
- Cancer Biology Program, Stanford University School of Medicine, Stanford, CA, USA
- Department of Dermatology, Stanford University School of Medicine, Redwood City, CA, USA
| | - Yanyan Qi
- Center for Personal Dynamic Regulomes, Stanford University School of Medicine, Stanford, CA, USA
- Department of Dermatology, Stanford University School of Medicine, Redwood City, CA, USA
| | | | | | | | - Maxwell R Mumbach
- Center for Personal Dynamic Regulomes, Stanford University School of Medicine, Stanford, CA, USA
- Department of Genetics, Stanford University School of Medicine, Stanford, CA, USA
| | - Sarah E Pierce
- Department of Genetics, Stanford University School of Medicine, Stanford, CA, USA
- Cancer Biology Program, Stanford University School of Medicine, Stanford, CA, USA
| | - M Ryan Corces
- Center for Personal Dynamic Regulomes, Stanford University School of Medicine, Stanford, CA, USA
- Department of Dermatology, Stanford University School of Medicine, Redwood City, CA, USA
| | | | | | | | | | - Jean Wang
- 10x Genomics, Inc., Pleasanton, CA, USA
| | - Li Wang
- 10x Genomics, Inc., Pleasanton, CA, USA
| | | | | | - Anne Lynn S Chang
- Department of Dermatology, Stanford University School of Medicine, Redwood City, CA, USA
| | | | - William J Greenleaf
- Center for Personal Dynamic Regulomes, Stanford University School of Medicine, Stanford, CA, USA.
- Department of Genetics, Stanford University School of Medicine, Stanford, CA, USA.
- Department of Applied Physics, Stanford University, Stanford, CA, USA.
- Chan Zuckerberg Biohub, San Francisco, CA, USA.
| | - Howard Y Chang
- Center for Personal Dynamic Regulomes, Stanford University School of Medicine, Stanford, CA, USA.
- Department of Genetics, Stanford University School of Medicine, Stanford, CA, USA.
- Department of Dermatology, Stanford University School of Medicine, Redwood City, CA, USA.
- Howard Hughes Medical Institute, Stanford University School of Medicine, Stanford, CA, USA.
| |
Collapse
|
24
|
McPherson SW, Heuss ND, Lehmann U, Roehrich H, Abedin M, Gregerson DS. The retinal environment induces microglia-like properties in recruited myeloid cells. J Neuroinflammation 2019; 16:151. [PMID: 31325968 PMCID: PMC6642741 DOI: 10.1186/s12974-019-1546-9] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2019] [Accepted: 07/11/2019] [Indexed: 12/31/2022] Open
Abstract
Background Microglia are essential to the development of the CNS and its homeostasis. Our prior findings suggested a niche model to describe the behaviors of retinal microglia. Here, we ask whether new myeloid cells recruited to the retina are constrained to resemble endogenous microglia morphologically and functionally. Methods Use of CD11cDTR/GFP transgenic mouse allowed identification of two niches of retinal microglia distinguished by being GFPlo or GFPhi. We also used transgenic mice in which CX3CR1+ cells expressed YFP and were depletable following tamoxifen-induced expression of diphtheria toxin subunit A. We employed several ablation and injury stimulation protocols to examine the origin and fate of myeloid cells repopulating the retina. Analysis of retinal myeloid cells was done by microscopy, flow cytometry, and qRT-PCR. Results We found that the origin of new GFPhi and GFPlo myeloid cells in the retina of CD11cDTR/GFP mice, whether recruited or local, depended on the ablation and stimulation protocols. Regardless of origin, new GFPlo and GFPhi retinal myeloid cells were CD45medCD11b+Ly6G−Ly6CloIba1+F4/80+, similar to endogenous microglia. Following tamoxifen-induced diphtheria toxin ablation, myeloid cell repopulation differed in the retina compared to the brain and optic nerve. Stimulation of replacement GFPhi cells was substantially attenuated in repopulating retinas after tamoxifen-induced diphtheria toxin ablation compared to control or radiation-ablated mice. In radiation bone marrow chimeric mice, replacement GFPhi myeloid cells from the circulation were slow to repopulate the retina unless stimulated by an optic nerve crush injury. However, once stimulated, recruited GFPhi cells were found to concentrate on injured retinal ganglion cells and were morphologically similar to GFPhi cells in non-ablated control CD11cDTR/GFP mice. Conclusions The results support the idea that GFPhi cells in the CD11cDTR/GFP mouse, whether recruited or from resident microglia, mark a unique niche of activated retinal myeloid cells. We conclude that the retinal environment has a potent influence on the function, morphology, and proliferative capacity of new myeloid cells regardless of their origin, compelling them to be equivalent to the endogenous microglia. Electronic supplementary material The online version of this article (10.1186/s12974-019-1546-9) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Scott W McPherson
- Department of Ophthalmology and Visual Neurosciences, University of Minnesota, 2001 6th Street SE, LRB Room 314, Minneapolis, MN, 55455, USA.
| | - Neal D Heuss
- Department of Ophthalmology and Visual Neurosciences, University of Minnesota, 2001 6th Street SE, LRB Room 314, Minneapolis, MN, 55455, USA
| | - Ute Lehmann
- Department of Ophthalmology and Visual Neurosciences, University of Minnesota, 2001 6th Street SE, LRB Room 314, Minneapolis, MN, 55455, USA
| | - Heidi Roehrich
- Department of Ophthalmology and Visual Neurosciences, University of Minnesota, 2001 6th Street SE, LRB Room 314, Minneapolis, MN, 55455, USA
| | - Md Abedin
- Department of Ophthalmology and Visual Neurosciences, University of Minnesota, 2001 6th Street SE, LRB Room 314, Minneapolis, MN, 55455, USA
| | - Dale S Gregerson
- Department of Ophthalmology and Visual Neurosciences, University of Minnesota, 2001 6th Street SE, LRB Room 314, Minneapolis, MN, 55455, USA
| |
Collapse
|
25
|
Musumeci A, Lutz K, Winheim E, Krug AB. What Makes a pDC: Recent Advances in Understanding Plasmacytoid DC Development and Heterogeneity. Front Immunol 2019; 10:1222. [PMID: 31191558 PMCID: PMC6548821 DOI: 10.3389/fimmu.2019.01222] [Citation(s) in RCA: 89] [Impact Index Per Article: 17.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2018] [Accepted: 05/13/2019] [Indexed: 12/16/2022] Open
Abstract
Dendritic cells (DCs) are professional antigen presenting cells (APCs) that originate in the bone marrow and are continuously replenished from hematopoietic progenitor cells. Conventional DCs (cDCs) and plasmacytoid DCs (pDCs) are distinguished by morphology and function, and can be easily discriminated by surface marker expression, both in mouse and man. Classification of DCs based on their ontology takes into account their origin as well as their requirements for transcription factor (TF) expression. cDCs and pDCs of myeloid origin differentiate from a common DC progenitor (CDP) through committed pre-DC stages. pDCs have also been shown to originate from a lymphoid progenitor derived IL-7R+ FLT3+ precursor population containing cells with pDC or B cell potential. Technological advancements in recent years have allowed unprecedented resolution in the analysis of cell states, down to the single cell level, providing valuable information on the commitment, and dynamics of differentiation of all DC subsets. However, the heterogeneity and functional diversification of pDCs still raises the question whether different ontogenies generate restricted pDC subsets, or fully differentiated pDCs retain plasticity in response to challenges. The emergence of novel techniques for the integration of high-resolution data in individual cells promises interesting discoveries regarding DC development and plasticity in the near future.
Collapse
Affiliation(s)
- Andrea Musumeci
- Institute for Immunology, Biomedical Center, Ludwig-Maximilian-University, Munich, Germany
| | - Konstantin Lutz
- Institute for Immunology, Biomedical Center, Ludwig-Maximilian-University, Munich, Germany
| | - Elena Winheim
- Institute for Immunology, Biomedical Center, Ludwig-Maximilian-University, Munich, Germany
| | - Anne Barbara Krug
- Institute for Immunology, Biomedical Center, Ludwig-Maximilian-University, Munich, Germany
| |
Collapse
|
26
|
Bouderlique T, Peña-Pérez L, Kharazi S, Hils M, Li X, Krstic A, De Paepe A, Schachtrup C, Gustafsson C, Holmberg D, Schachtrup K, Månsson R. The Concerted Action of E2-2 and HEB Is Critical for Early Lymphoid Specification. Front Immunol 2019; 10:455. [PMID: 30936870 PMCID: PMC6433000 DOI: 10.3389/fimmu.2019.00455] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2018] [Accepted: 02/20/2019] [Indexed: 12/21/2022] Open
Abstract
The apparition of adaptive immunity in Gnathostomata correlates with the expansion of the E-protein family to encompass E2-2, HEB, and E2A. Within the family, E2-2 and HEB are more closely evolutionarily related but their concerted action in hematopoiesis remains to be explored. Here we show that the combined disruption of E2-2 and HEB results in failure to express the early lymphoid program in Common lymphoid precursors (CLPs) and a near complete block in B-cell development. In the thymus, Early T-cell progenitors (ETPs) were reduced and T-cell development perturbed, resulting in reduced CD4 T- and increased γδ T-cell numbers. In contrast, hematopoietic stem cells (HSCs), erythro-myeloid progenitors, and innate immune cells were unaffected showing that E2-2 and HEB are dispensable for the ancestral hematopoietic lineages. Taken together, this E-protein dependence suggests that the appearance of the full Gnathostomata E-protein repertoire was critical to reinforce the gene regulatory circuits that drove the emergence and expansion of the lineages constituting humoral immunity.
Collapse
Affiliation(s)
- Thibault Bouderlique
- Department of Laboratory Medicine, Center for Hematology and Regenerative Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Lucia Peña-Pérez
- Department of Laboratory Medicine, Center for Hematology and Regenerative Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Shabnam Kharazi
- Department of Laboratory Medicine, Center for Hematology and Regenerative Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Miriam Hils
- Faculty of Medicine & Faculty of Biology, Center for Chronic Immunodeficiency, Medical Center, University of Freiburg, Freiburg, Germany
| | - Xiaoze Li
- Department of Laboratory Medicine, Center for Hematology and Regenerative Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Aleksandra Krstic
- Department of Laboratory Medicine, Center for Hematology and Regenerative Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Ayla De Paepe
- Department of Laboratory Medicine, Center for Hematology and Regenerative Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Christian Schachtrup
- Faculty of Medicine, Institute of Anatomy and Cell Biology, University of Freiburg, Freiburg, Germany
| | - Charlotte Gustafsson
- Department of Laboratory Medicine, Center for Hematology and Regenerative Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Dan Holmberg
- Lund University Diabetes Center, Lund University, Malmö, Sweden
| | - Kristina Schachtrup
- Faculty of Medicine & Faculty of Biology, Center for Chronic Immunodeficiency, Medical Center, University of Freiburg, Freiburg, Germany
| | - Robert Månsson
- Department of Laboratory Medicine, Center for Hematology and Regenerative Medicine, Karolinska Institutet, Stockholm, Sweden.,Hematology Center, Karolinska University Hospital, Stockholm, Sweden
| |
Collapse
|
27
|
He Z, Zhu X, Shi Z, Wu T, Wu L. Metabolic Regulation of Dendritic Cell Differentiation. Front Immunol 2019; 10:410. [PMID: 30930893 PMCID: PMC6424910 DOI: 10.3389/fimmu.2019.00410] [Citation(s) in RCA: 40] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2018] [Accepted: 02/15/2019] [Indexed: 12/11/2022] Open
Abstract
Dendritic cells (DCs) are important antigen-presenting cells (APCs) that play essential roles in bridging innate and adaptive immune responses. Differentiation stages of DC subsets from bone marrow progenitor cells have been well-defined during the past decades. Features that distinguish DC progenitor cells from each differentiation stages, related signaling pathways and transcription factors that are crucial for DC lineage commitment have been well-elucidated in numerous studies. Recently, growing evidence are showing that cellular metabolism, as one of the most fundamental process of cells, has essential role in the modulation of immune system. There have been multiple reports and reviews that focus on the metabolic modulations on DC functions, however little attention had been paid to the metabolic regulation of DC development and differentiation. In recent years, increasing evidence suggests that metabolic regulations also exert significant impact on DC differentiation, as well as on the homeostasis of tissue resident DCs. The focus of this review is to summarize the findings from recent studies on the metabolic regulation of DC differentiation and to discuss the impacts of the three major aspects of metabolism on the processes of DC development and differentiation, namely the changes in metabolic pathways, the molecular signaling pathways that modulate cell metabolism, and the effects of metabolites and nutrients. The aim of this review is to draw attentions to this important and exciting research field where the effects of metabolic process and their regulation in DC differentiation need to be further explored.
Collapse
Affiliation(s)
- Zhimin He
- School of Medicine, Institute for Immunology, Tsinghua University, Beijing, China.,Tsinghua-Peking Center for Life Science, Beijing, China
| | - Xinyi Zhu
- School of Medicine, Institute for Immunology, Tsinghua University, Beijing, China
| | - Zhen Shi
- School of Medicine, Institute for Immunology, Tsinghua University, Beijing, China
| | - Tao Wu
- School of Medicine, Institute for Immunology, Tsinghua University, Beijing, China
| | - Li Wu
- School of Medicine, Institute for Immunology, Tsinghua University, Beijing, China.,Tsinghua-Peking Center for Life Science, Beijing, China
| |
Collapse
|
28
|
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: 1.7] [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
|
29
|
Salvermoser J, van Blijswijk J, Papaioannou NE, Rambichler S, Pasztoi M, Pakalniškytė D, Rogers NC, Keppler SJ, Straub T, Reis e Sousa C, Schraml BU. Clec9a-Mediated Ablation of Conventional Dendritic Cells Suggests a Lymphoid Path to Generating Dendritic Cells In Vivo. Front Immunol 2018; 9:699. [PMID: 29713321 PMCID: PMC5911463 DOI: 10.3389/fimmu.2018.00699] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2018] [Accepted: 03/21/2018] [Indexed: 01/01/2023] Open
Abstract
Conventional dendritic cells (cDCs) are versatile activators of immune responses that develop as part of the myeloid lineage downstream of hematopoietic stem cells. We have recently shown that in mice precursors of cDCs, but not of other leukocytes, are marked by expression of DNGR-1/CLEC9A. To genetically deplete DNGR-1-expressing cDC precursors and their progeny, we crossed Clec9a-Cre mice to Rosa-lox-STOP-lox-diphtheria toxin (DTA) mice. These mice develop signs of age-dependent myeloproliferative disease, as has been observed in other DC-deficient mouse models. However, despite efficient depletion of cDC progenitors in these mice, cells with phenotypic characteristics of cDCs populate the spleen. These cells are functionally and transcriptionally similar to cDCs in wild type control mice but show somatic rearrangements of Ig-heavy chain genes, characteristic of lymphoid origin cells. Our studies reveal a previously unappreciated developmental heterogeneity of cDCs and suggest that the lymphoid lineage can generate cells with features of cDCs when myeloid cDC progenitors are impaired.
Collapse
Affiliation(s)
- Johanna Salvermoser
- Walter-Brendel-Centre for Experimental Medicine, University Hospital, LMU Munich, Planegg Martinsried, Germany.,Biomedical Center, LMU Munich, Planegg Martinsried, Germany
| | | | | | - Stephan Rambichler
- Walter-Brendel-Centre for Experimental Medicine, University Hospital, LMU Munich, Planegg Martinsried, Germany.,Biomedical Center, LMU Munich, Planegg Martinsried, Germany
| | - Maria Pasztoi
- Biomedical Center, LMU Munich, Planegg Martinsried, Germany
| | - Dalia Pakalniškytė
- Walter-Brendel-Centre for Experimental Medicine, University Hospital, LMU Munich, Planegg Martinsried, Germany.,Biomedical Center, LMU Munich, Planegg Martinsried, Germany
| | - Neil C Rogers
- Immunobiology Laboratory, The Francis Crick Institute, London, United Kingdom
| | - Selina J Keppler
- Technische Universität München, Klinikum Rechts der Isar, Institut für Klinische Chemie und Pathobiochemie, Munich, Germany
| | - Tobias Straub
- Biomedical Center, LMU Munich, Planegg Martinsried, Germany.,Core Facility Bioinformatics, Biomedical Center (BMC), LMU Munich, Planegg Martinsried, Germany
| | | | - Barbara U Schraml
- Walter-Brendel-Centre for Experimental Medicine, University Hospital, LMU Munich, Planegg Martinsried, Germany.,Biomedical Center, LMU Munich, Planegg Martinsried, Germany
| |
Collapse
|
30
|
Lutz MB, Strobl H, Schuler G, Romani N. GM-CSF Monocyte-Derived Cells and Langerhans Cells As Part of the Dendritic Cell Family. Front Immunol 2017; 8:1388. [PMID: 29109731 PMCID: PMC5660299 DOI: 10.3389/fimmu.2017.01388] [Citation(s) in RCA: 45] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2017] [Accepted: 10/09/2017] [Indexed: 12/21/2022] Open
Abstract
Dendritic cells (DCs) and macrophages (Mph) share many characteristics as components of the innate immune system. The criteria to classify the multitude of subsets within the mononuclear phagocyte system are currently phenotype, ontogeny, transcription patterns, epigenetic adaptations, and function. More recently, ontogenetic, transcriptional, and proteomic research approaches uncovered major developmental differences between Flt3L-dependent conventional DCs as compared with Mphs and monocyte-derived DCs (MoDCs), the latter mainly generated in vitro from murine bone marrow-derived DCs (BM-DCs) or human CD14+ peripheral blood monocytes. Conversely, in vitro GM-CSF-dependent monocyte-derived Mphs largely resemble MoDCs whereas tissue-resident Mphs show a common embryonic origin from yolk sac and fetal liver with Langerhans cells (LCs). The novel ontogenetic findings opened discussions on the terminology of DCs versus Mphs. Here, we bring forward arguments to facilitate definitions of BM-DCs, MoDCs, and LCs. We propose a group model of terminology for all DC subsets that attempts to encompass both ontogeny and function.
Collapse
Affiliation(s)
- Manfred B Lutz
- Institute for Virology and Immunobiology, University of Würzburg, Würzburg, Germany
| | - Herbert Strobl
- Institute of Pathophysiology and Immunology, Medical University of Graz, Graz, Austria
| | - Gerold Schuler
- Department of Dermatology, University Hospital Erlangen, Erlangen, Germany
| | - Nikolaus Romani
- Department of Dermatology, Venereology and Allergology, Medical University of Innsbruck, Innsbruck, Austria
| |
Collapse
|
31
|
Li S, Wu J, Zhu S, Liu YJ, Chen J. Disease-Associated Plasmacytoid Dendritic Cells. Front Immunol 2017; 8:1268. [PMID: 29085361 PMCID: PMC5649186 DOI: 10.3389/fimmu.2017.01268] [Citation(s) in RCA: 63] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2016] [Accepted: 09/22/2017] [Indexed: 12/20/2022] Open
Abstract
Plasmacytoid dendritic cells (pDCs), also called natural interferon (IFN)-producing cells, represent a specialized cell type within the innate immune system. pDCs are specialized in sensing viral RNA and DNA by toll-like receptor-7 and -9 and have the ability to rapidly produce massive amounts of type 1 IFNs upon viral encounter. After producing type 1 IFNs, pDCs differentiate into professional antigen-presenting cells, which are capable of stimulating T cells of the adaptive immune system. Chronic activation of human pDCs by self-DNA or mitochondrial DNA contributes to the pathogenesis of systemic lupus erythematosis and IFN-related autoimmune diseases. Under steady-state conditions, pDCs play an important role in immune tolerance. In many types of human cancers, recruitment of pDCs to the tumor microenvironment contributes to the induction of immune tolerance. Here, we provide a systemic review of recent progress in studies on the role of pDCs in human diseases, including cancers and autoimmune/inflammatory diseases.
Collapse
Affiliation(s)
- Shuang Li
- Institute of Translational Medicine, The First Hospital, Jilin University, Changchun, China
| | - Jing Wu
- Institute of Translational Medicine, The First Hospital, Jilin University, Changchun, China
| | - Shan Zhu
- Institute of Translational Medicine, The First Hospital, Jilin University, Changchun, China
| | - Yong-Jun Liu
- Institute of Translational Medicine, The First Hospital, Jilin University, Changchun, China.,Sanofi Research and Development, Cambridge, MA, United States
| | - Jingtao Chen
- Institute of Translational Medicine, The First Hospital, Jilin University, Changchun, China
| |
Collapse
|
32
|
Sichien D, Lambrecht BN, Guilliams M, Scott CL. Development of conventional dendritic cells: from common bone marrow progenitors to multiple subsets in peripheral tissues. Mucosal Immunol 2017; 10:831-844. [PMID: 28198365 DOI: 10.1038/mi.2017.8] [Citation(s) in RCA: 132] [Impact Index Per Article: 18.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2016] [Accepted: 01/14/2017] [Indexed: 02/04/2023]
Abstract
Our understanding of conventional dendritic cell (cDC) development and the functional specializations of distinct subsets in the peripheral tissues has increased greatly in recent years. Here, we review cDC development from the distinct progenitors in the bone marrow through to the distinct cDC subsets found in barrier tissues, providing an overview of the different subsets described in each location. In addition, we detail the transcription factors and local signals that have been proposed to control this developmental process. Importantly, despite these significant advances, numerous questions remain to be answered regarding cDC development. For example, it remains unclear whether the different subsets described, such as the CD103+CD11b+ and CD103-CD11b+ cDCs in the intestines, truly represent different populations or rather distinct developmental or activation stages. Furthermore, whether distinct progenitors exist for these cDC subsets remains to be determined. Thus in the last part of this review we discuss what we believe will be the main questions facing the field for the coming years.
Collapse
Affiliation(s)
- D Sichien
- Laboratory of Immunoregulation and Mucosal Immunology, VIB-UGent Center for Inflammation Research, Ghent, Belgium
- Department of Biomedical Molecular Biology, Ghent University, Ghent, Belgium
| | - B N Lambrecht
- Laboratory of Immunoregulation and Mucosal Immunology, VIB-UGent Center for Inflammation Research, Ghent, Belgium
- Department of Respiratory Medicine, Ghent University, Ghent, Belgium
- Department of Pulmonary Medicine, ErasmusMC, Rotterdam, The Netherlands
| | - M Guilliams
- Laboratory of Immunoregulation and Mucosal Immunology, VIB-UGent Center for Inflammation Research, Ghent, Belgium
- Department of Biomedical Molecular Biology, Ghent University, Ghent, Belgium
| | - C L Scott
- Laboratory of Immunoregulation and Mucosal Immunology, VIB-UGent Center for Inflammation Research, Ghent, Belgium
- Department of Biomedical Molecular Biology, Ghent University, Ghent, Belgium
| |
Collapse
|
33
|
Lineage specification of human dendritic cells is marked by IRF8 expression in hematopoietic stem cells and multipotent progenitors. Nat Immunol 2017. [PMID: 28650480 PMCID: PMC5743223 DOI: 10.1038/ni.3789] [Citation(s) in RCA: 92] [Impact Index Per Article: 13.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
The origin and specification of human dendritic cells (DCs) have not been investigated at clonal level. Using clonal assays combined with statistical computation to quantify the yield of granulocytes, monocytes, lymphocytes and three subsets of DCs from single human CD34+ progenitor cells, we show DC lineage specification occurs in parallel with myeloid and lymphoid lineages in HSCs, starting as a lineage bias defined by specific transcriptional programs correlated with the relative IRF8/PU.1 ratios, which is transmitted to most progeny and reinforced by FLT3L-driven IRF8 upregulation over division. We propose a model in which DC lineage specification is driven by parallel and inheritable transcriptional programs in HSCs, and reinforced over cell division by recursive interaction between transcriptional programs and extrinsic signals.
Collapse
|
34
|
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: 1.1] [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
|
35
|
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: 6.9] [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
|
36
|
Bullinger L. T-lymphoid progenitors - we know what they are, but know not what they may be. EMBO J 2016; 35:2383-2385. [PMID: 27670761 DOI: 10.15252/embj.201695613] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Affiliation(s)
- Lars Bullinger
- Department of Internal Medicine III, University Hospital of Ulm, Ulm, Germany
| |
Collapse
|
37
|
Investigating B Cell Development, Natural and Primary Antibody Responses in Ly-6A/Sca-1 Deficient Mice. PLoS One 2016; 11:e0157271. [PMID: 27322740 PMCID: PMC4913937 DOI: 10.1371/journal.pone.0157271] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2015] [Accepted: 05/26/2016] [Indexed: 11/24/2022] Open
Abstract
Ly-6A/Stem cell antigen-1 (Ly-6A/Sca-1) is a glycosylphosphatidylinositol-anchored protein expressed on many cell types including hematopoietic stem cells (HSCs) and early lymphoid-specific progenitors. Ly-6A/Sca-1 is expressed on CD4+ T cells and plays a role in regulating cellular responses to foreign antigens. The role of Ly-6A/Sca-1 in primary antibody responses has not been defined. To investigate whether Ly-6A/Sca-1 functions in humoral immunity, we first injected Ly-6A/Sca-1-deficient and wild-type control mice with chicken ovalbumin (c-Ova) protein mixed with an adjuvant. We then assessed the ability of the mice to generate a primary antibody response against cOva. We further examined the development of B cells and circulating antibody isotypes in non-immunized Ly-6A/Sca-1deficient mice to determine if Ly6A/Sca-1 functions in development irrespective of antigen-specific immune activation. Ly-6A/Sca-1/Sca-1-deficient mice did not show any significant changes in the number of B lymphocytes in the bone marrow and peripheral lymphoid tissues. Interestingly, Ly-6A/Sca-1/Sca-1-/- mice have significantly elevated serum levels of IgA with λ light chains compared to wild type controls. B cell clusters with high reactivity to anti-IgA λ monoclonal antibody were detected in the lamina propria of the gut, though this was not observed in the bone marrow and peripheral lymphoid tissues. Despite these differences, the Ly-6A/Sca-1deficient mice generated a similar primary antibody response when compared to the wild-type mice. In summary, we conclude that the primary antibody response to cOva antigen is similar in Ly-6A/Sca-1deficient and sufficient mice. In addition, we report significantly higher expression of the immunoglobulin λ light chain by B cells in lamina propria of Ly-6A/Sca-1deficient mice when compared to the wild-type control.
Collapse
|
38
|
GATA2 regulates dendritic cell differentiation. Blood 2016; 128:508-18. [PMID: 27259979 DOI: 10.1182/blood-2016-02-698118] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2016] [Accepted: 05/18/2016] [Indexed: 12/29/2022] Open
Abstract
Dendritic cells (DCs) are critical immune response regulators; however, the mechanism of DC differentiation is not fully understood. Heterozygous germ line GATA2 mutations induce GATA2-deficiency syndrome, characterized by monocytopenia, a predisposition to myelodysplasia/acute myeloid leukemia, and a profoundly reduced DC population, which is associated with increased susceptibility to viral infections, impaired phagocytosis, and decreased cytokine production. To define the role of GATA2 in DC differentiation and function, we studied Gata2 conditional knockout and haploinsufficient mice. Gata2 conditional deficiency significantly reduced the DC count, whereas Gata2 haploinsufficiency did not affect this population. GATA2 was required for the in vitro generation of DCs from Lin(-)Sca-1(+)Kit(+) cells, common myeloid-restricted progenitors, and common dendritic cell precursors, but not common lymphoid-restricted progenitors or granulocyte-macrophage progenitors, suggesting that GATA2 functions in the myeloid pathway of DC differentiation. Moreover, expression profiling demonstrated reduced expression of myeloid-related genes, including mafb, and increased expression of T-lymphocyte-related genes, including Gata3 and Tcf7, in Gata2-deficient DC progenitors. In addition, GATA2 was found to bind an enhancer element 190-kb downstream region of Gata3, and a reporter assay exhibited significantly reduced luciferase activity after adding this enhancer region to the Gata3 promoter, which was recovered by GATA sequence deletion within Gata3 +190. These results suggest that GATA2 plays an important role in cell-fate specification toward the myeloid vs T-lymphocyte lineage by regulating lineage-specific transcription factors in DC progenitors, thereby contributing to DC differentiation.
Collapse
|
39
|
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: 25] [Impact Index Per Article: 3.1] [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
|
40
|
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: 0.9] [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
|
41
|
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: 316] [Impact Index Per Article: 35.1] [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
|
42
|
Jang SH, Lee S, Chung HY. Characterization of Leukemia-Inducing Genes Using a Proto-Oncogene/Homeobox Gene Retroviral Human cDNA Library in a Mouse In Vivo Model. PLoS One 2015; 10:e0143240. [PMID: 26606454 PMCID: PMC4659616 DOI: 10.1371/journal.pone.0143240] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2015] [Accepted: 11/01/2015] [Indexed: 11/18/2022] Open
Abstract
The purpose of this research is to develop a method to screen a large number of potential driver mutations of acute myeloid leukemia (AML) using a retroviral cDNA library and murine bone marrow transduction-transplantation system. As a proof-of-concept, murine bone marrow (BM) cells were transduced with a retroviral cDNA library encoding well-characterized oncogenes and homeobox genes, and the virus-transduced cells were transplanted into lethally irradiated mice. The proto-oncogenes responsible for leukemia initiation were identified by PCR amplification of cDNA inserts from genomic DNA isolated from leukemic cells. In an initial screen of ten leukemic mice, the MYC proto-oncogene was detected in all the leukemic mice. Of ten leukemic mice, 3 (30%) had MYC as the only transgene, and seven mice (70%) had additional proto-oncogene inserts. We repeated the same experiment after removing MYC-related genes from the library to characterize additional leukemia-inducing gene combinations. Our second screen using the MYC-deleted proto-oncogene library confirmed MEIS1and the HOX family as cooperating oncogenes in leukemia pathogenesis. The model system we introduced in this study will be valuable in functionally screening novel combinations of genes for leukemogenic potential in vivo, and the system will help in the discovery of new targets for leukemia therapy.
Collapse
Affiliation(s)
- Su Hwa Jang
- Department of Biomedical Science, Graduate School of Biomedical Science and Engineering, Hanyang University, Seoul, Republic of Korea
| | - Sohyun Lee
- Department of Biomedical Science, Graduate School of Biomedical Science and Engineering, Hanyang University, Seoul, Republic of Korea
| | - Hee Yong Chung
- Department of Biomedical Science, Graduate School of Biomedical Science and Engineering, Hanyang University, Seoul, Republic of Korea
- * E-mail:
| |
Collapse
|
43
|
Chang S, Pai LM, Lee CK. In Vitro Generation of Murine Plasmacytoid Dendritic Cells from Common Lymphoid Progenitors using the AC-6 Feeder System. J Vis Exp 2015. [PMID: 26650046 DOI: 10.3791/53211] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2023] Open
Abstract
Plasmacytoid dendritic cells (pDCs) are powerful type I interferon (IFN-I) producing cells that are activated in response to infection or during inflammatory responses. Unfortunately, study of pDC function is hindered by their low frequency in lymphoid organs, and existing methods for in vitro DC generation predominantly favor the production of cDCs over pDCs. Here we present a unique approach to efficiently generate pDCs from common lymphoid progenitors (CLPs) in vitro. Specifically, the protocol described details how to purify CLPs from bone marrow and generate pDCs by coculturing with γ-irradiated AC-6 feeder cells in the presence of Flt3 ligand. A unique characteristic of this culture system is that the CLPs migrate underneath the AC-6 cells and become cobblestone area-forming cells, a critical step for expanding pDCs. Morphologically distinct DCs, namely pDCs and cDCs, were generated after approximately 2 weeks with a composition of 70-90% pDCs under optimal conditions. Typically, the number of pDCs generated by this method is roughly 100-fold of the number of CLPs seeded. Therefore, this is a novel system with which to robustly generate the large numbers of pDCs required to facilitate further studies into the development and function of these cells.
Collapse
Affiliation(s)
- Shiun Chang
- Graduate Institute of Immunology, National Taiwan University College of Medicine
| | - Li-Mei Pai
- Department of Biochemistry, Chang Gung University College of Medicine; Molecular Medicine Research Center, Chang Gung University College of Medicine
| | - Chien-Kuo Lee
- Graduate Institute of Immunology, National Taiwan University College of Medicine;
| |
Collapse
|
44
|
Podestà MA, Cucchiari D, Ponticelli C. The diverging roles of dendritic cells in kidney allotransplantation. Transplant Rev (Orlando) 2015; 29:114-20. [DOI: 10.1016/j.trre.2015.04.001] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2014] [Revised: 03/09/2015] [Accepted: 04/08/2015] [Indexed: 12/23/2022]
|
45
|
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: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2014] [Accepted: 02/10/2015] [Indexed: 12/23/2022]
|
46
|
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: 3.0] [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
|
47
|
Fan X, Liu Z, Jin H, Yan J, Liang HP. Alterations of dendritic cells in sepsis: featured role in immunoparalysis. BIOMED RESEARCH INTERNATIONAL 2015; 2015:903720. [PMID: 25821827 PMCID: PMC4363672 DOI: 10.1155/2015/903720] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/15/2014] [Revised: 05/25/2014] [Accepted: 07/28/2014] [Indexed: 12/12/2022]
Abstract
Sepsis, the leading cause of mortality in intensive care unit, is characterized by hyperinflammatory response in the early stage and followed by a period of immunosuppression. This immune disorder is believed to be the potent factor that is tightly associated with high mortality in sepsis. Dendritic cells (DCs) serve as professional antigen-presenting cells that play a vital role in immune response by activating T lymphocytes. During the progression of sepsis, DCs have been reported to take part in the aberrant immune response and be necessary for survival. Therefore, a better understanding of the DCs pathology will be undoubtedly beneficial for resolving the problems occurring in sepsis. This review discusses effects of sepsis on DCs number and function, including surface molecules expression, cytokines secretion, and T cell activation, and the underlying mechanism as well as some potential therapeutic strategies.
Collapse
Affiliation(s)
- Xia Fan
- State Key Laboratory of Trauma, Burns and Combined Injury, Research Institute of Surgery, Daping Hospital, The Third Military Medical University, Chongqing 400042, China
| | - Zheng Liu
- State Key Laboratory of Trauma, Burns and Combined Injury, Research Institute of Surgery, Daping Hospital, The Third Military Medical University, Chongqing 400042, China
| | - He Jin
- State Key Laboratory of Trauma, Burns and Combined Injury, Research Institute of Surgery, Daping Hospital, The Third Military Medical University, Chongqing 400042, China
| | - Jun Yan
- State Key Laboratory of Trauma, Burns and Combined Injury, Research Institute of Surgery, Daping Hospital, The Third Military Medical University, Chongqing 400042, China
| | - Hua-ping Liang
- State Key Laboratory of Trauma, Burns and Combined Injury, Research Institute of Surgery, Daping Hospital, The Third Military Medical University, Chongqing 400042, China
| |
Collapse
|
48
|
Abstract
The lymphocyte family has expanded significantly in recent years to include not only the adaptive lymphocytes (T cells, B cells) and NK cells, but also several additional innate lymphoid cell (ILC) types. ILCs lack clonally distributed antigen receptors characteristic of adaptive lymphocytes and instead respond exclusively to signaling via germline-encoded receptors. ILCs resemble T cells more closely than any other leukocyte lineage at the transcriptome level and express many elements of the core T cell transcriptional program, including Notch, Gata3, Tcf7, and Bcl11b. We present our current understanding of the shared and distinct transcriptional regulatory mechanisms involved in the development of adaptive T lymphocytes and closely related ILCs. We discuss the possibility that a core set of transcriptional regulators common to ILCs and T cells establish enhancers that enable implementation of closely aligned effector pathways. Studies of the transcriptional regulation of lymphopoiesis will support the development of novel therapeutic approaches to correct early lymphoid developmental defects and aberrant lymphocyte function.
Collapse
Affiliation(s)
- Maria Elena De Obaldia
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania 19104
| | | |
Collapse
|
49
|
Becker AM, Walcheck B, Bhattacharya D. ADAM17 limits the expression of CSF1R on murine hematopoietic progenitors. Exp Hematol 2015; 43:44-52.e1-3. [PMID: 25308957 PMCID: PMC4268392 DOI: 10.1016/j.exphem.2014.10.001] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2014] [Revised: 08/05/2014] [Accepted: 10/01/2014] [Indexed: 01/07/2023]
Abstract
All-lymphoid progenitors (ALPs) yield few myeloid cells in vivo, but readily generate such cells in vitro. The basis for this difference remains unknown. We hypothesized that ALPs limit responsiveness to in vivo concentrations of myeloid-promoting cytokines by reducing expression of the corresponding receptors, potentially through posttranscriptional mechanisms. Consistent with such a mechanism, ALPs express higher levels of CSF1R transcripts than their upstream precursors, yet show limited cell-surface protein expression of colony-stimulating factor 1 receptor (CSF1R). All-lymphoid progenitors and other hematopoietic progenitors deficient in A disintegrin and metalloproteinase domain 17 (ADAM17), display elevated cell surface CSF1R expression. ADAM17(-/-) ALPs, however, fail to yield myeloid cells upon transplantation into irradiated recipients. Moreover, ADAM17(-/-) ALPs yield fewer macrophages in vitro than control ALPs at high concentrations of macrophage colony stimulating factor. Mice with hematopoietic-specific deletion of ADAM17 have normal numbers of myeloid and lymphoid progenitors and mature cells in vivo. These data demonstrate that ADAM17 limits CSF1R protein expression on hematopoietic progenitors, but that compensatory mechanisms prevent elevated CSF1R levels from altering lymphoid progenitor potential.
Collapse
Affiliation(s)
- Amy M Becker
- Department of Pathology and Immunology, Washington University School of Medicine, Saint Louis, MO, United States
| | - Bruce Walcheck
- Department of Veterinary and Biomedical Sciences, University of Minnesota, St. Paul, MN, USA
| | - Deepta Bhattacharya
- Department of Pathology and Immunology, Washington University School of Medicine, Saint Louis, MO, United States.
| |
Collapse
|
50
|
Bieber K, Autenrieth SE. Insights how monocytes and dendritic cells contribute and regulate immune defense against microbial pathogens. Immunobiology 2014; 220:215-26. [PMID: 25468558 DOI: 10.1016/j.imbio.2014.10.025] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2014] [Revised: 10/17/2014] [Accepted: 10/23/2014] [Indexed: 12/19/2022]
Abstract
The immune system protects from infections primarily by detecting and eliminating invading pathogens. Beside neutrophils, monocytes and dendritic cells (DCs) have been recently identified as important sentinels and effectors in combating microbial pathogens. In the steady state mononuclear phagocytes like monocytes and DCs patrol the blood and the tissues. Mammalian monocytes contribute to antimicrobial defense by supplying tissues with macrophage and DC precursors. DCs recognize pathogens and are essential in presenting antigens to initiate antigen-specific adaptive immune responses, thereby bridging the innate and adaptive immune systems. Both, monocytes and DCs play distinct roles in the shaping of immune response. In this review we will focus on the contributions of monocytes and lymphoid organ DCs to immune defense against microbial pathogens in the mouse and their dynamic regulation from steady state to infection.
Collapse
Affiliation(s)
- Kristin Bieber
- Department of Internal Medicine II, University of Tübingen, Germany
| | | |
Collapse
|