1
|
Yoon J, Sun S, Moon S, Yang H. Repeated Gonadotropin Administration Suppresses T Cell Development in the Mouse Thymus. Dev Reprod 2025; 29:1-11. [PMID: 40256124 PMCID: PMC12004010 DOI: 10.12717/dr.2025.29.1.1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2024] [Revised: 01/14/2025] [Accepted: 02/23/2025] [Indexed: 04/22/2025]
Abstract
Gonadotropins, such as follicle-stimulating hormone (FSH) and human chorionic gonadotropin (hCG), are widely used to induce ovarian hyperovulation during in vitro fertilization and embryo transfer (IVF-ET) for the treatment of infertility. However, the effects of repeated administration of these gonadotropins on immune function, particularly on T cell development in the thymus, remain poorly understood. This study investigated the effects of repeated administration of pregnant mare serum gonadotropin (PMSG) and hCG on thymic T cell development in mice. Histological analysis revealed structural changes in the thymus, including a blurred boundary between the medulla and cortex and reduced vascularization after repeated administration of PMSG and hCG. Quantitative real-time PCR showed increased expression of adipogenesis-related genes [phosphoenolpyruvate carboxykinase (PEPCK), adipocyte fatty acid-binding protein 2 (aP2), peroxisome proliferator-activated receptor gamma (PPARγ)] but no significant changes in thymic epithelial cell-related genes [autoimmune regulator (AIRE), epithelial V-like antigen (EVA), interleukin 7 (IL-7)]. Flow cytometry revealed a decrease in CD4+CD8+ T cells and an increase in CD4-CD8-T cells with altered CD25/CD44 subsets. In addition, CD4+ and CD8+ T cells in the spleen were significantly reduced. These findings suggest that repeated gonadotropin exposure may disrupt thymic T cell development and peripheral T cell populations, potentially impairing immune function. Further research is needed to elucidate the underlying mechanisms and broader immunologic consequences of gonadotropin use in infertility treatment.
Collapse
Affiliation(s)
- Jin Yoon
- Department of Biohealth Convergence, College of Science and Convergence Technology, Seoul Women’s University, Seoul 01797, Korea
| | - Sojung Sun
- Department of Biohealth Convergence, College of Science and Convergence Technology, Seoul Women’s University, Seoul 01797, Korea
| | - Soeun Moon
- Department of Biohealth Convergence, College of Science and Convergence Technology, Seoul Women’s University, Seoul 01797, Korea
| | - Hyunwon Yang
- Department of Biohealth Convergence, College of Science and Convergence Technology, Seoul Women’s University, Seoul 01797, Korea
| |
Collapse
|
2
|
Cao JF, Yang GJ, Zhang YA, Chen J. Contribution of interleukins in the regulation of teleost fish immunity: A review from the perspective of regulating macrophages. FISH & SHELLFISH IMMUNOLOGY 2025; 158:110173. [PMID: 39909123 DOI: 10.1016/j.fsi.2025.110173] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/12/2024] [Revised: 01/20/2025] [Accepted: 02/02/2025] [Indexed: 02/07/2025]
Abstract
Interleukins (ILs) are potent secreted regulators of a wide range of cell types and cellular activities, particularly in the immune system. They are able to participate in intercellular communication in homeostasis and disease, thereby exerting immune functions. Macrophages serve as the innate immune cells of vertebrates and play a pivotal role in defending against and eliminating external pathogens. In mammals, the immune response mounted by macrophages is intricately linked to ILs. Given the fact that teleost fish have evolved an innate immune system that closely resembles those of mammals, particularly in terms of the functionality of macrophages, raises the intriguing possibility that the regulatory function of ILs in macrophage-mediated immunity might be evolutionarily conserved across both mammal and teleost fish lineages. Consequently, from the perspective of interleukin regulation of macrophages, this review outlines the relationship between ILs and macrophages in teleost fish, and elucidates the regulatory role of ILs of immune cell function in teleost fish, thereby contributing to our understanding of the key role of these cytokines in the prevention and control of aquaculture diseases.
Collapse
Affiliation(s)
- Jia-Feng Cao
- State Key Laboratory for Quality and Safety of Agro-products, Ningbo University, Ningbo, 315211, China; Laboratory of Biochemistry and Molecular Biology, School of Marine Sciences, Ningbo University, Ningbo, 315832, China; Key Laboratory of Aquacultural Biotechnology, Ministry of Education, School of Marine Sciences, Ningbo University, Ningbo, Zhejiang, 315832, China
| | - Guan-Jun Yang
- State Key Laboratory for Quality and Safety of Agro-products, Ningbo University, Ningbo, 315211, China; Laboratory of Biochemistry and Molecular Biology, School of Marine Sciences, Ningbo University, Ningbo, 315832, China; Key Laboratory of Aquacultural Biotechnology, Ministry of Education, School of Marine Sciences, Ningbo University, Ningbo, Zhejiang, 315832, China
| | - Yong-An Zhang
- State Key Laboratory of Agricultural Microbiology, College of Fisheries, Huazhong Agricultural University, Wuhan, 430070, China; Hubei Hongshan Laboratory, Huazhong Agricultural University, Wuhan, 430070, China.
| | - Jiong Chen
- State Key Laboratory for Quality and Safety of Agro-products, Ningbo University, Ningbo, 315211, China; Laboratory of Biochemistry and Molecular Biology, School of Marine Sciences, Ningbo University, Ningbo, 315832, China; Key Laboratory of Aquacultural Biotechnology, Ministry of Education, School of Marine Sciences, Ningbo University, Ningbo, Zhejiang, 315832, China.
| |
Collapse
|
3
|
Matsumoto M, Sobral F, Cardoso JS, Oya T, Tsuneyama K, Matsumoto M, Alves NL. The Ins and Outs of Thymic Epithelial Cell Differentiation and Function. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2025; 1471:51-79. [PMID: 40067584 DOI: 10.1007/978-3-031-77921-3_3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/13/2025]
Abstract
The thymus is an essential component of the immune system responsible for producing T cells. It is anatomically divided into two main regions: the outer cortex and the inner medulla. This chapter summarizes our current understanding of thymic stromal cell functions, with a particular focus on the interactions between these cells and T cells. This exploration aims to shed light on the pathogenesis of immune disorders, including autoimmunity.
Collapse
Affiliation(s)
- Minoru Matsumoto
- Department of Molecular Pathology, Tokushima University Graduate School of Biomedical Sciences, Tokushima, Japan
| | - Francisco Sobral
- Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal
- Thymus Development and Function Laboratory, Instituto de Biologia Molecular e Celular, Porto, Portugal
| | - João S Cardoso
- Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal
- Thymus Development and Function Laboratory, Instituto de Biologia Molecular e Celular, Porto, Portugal
| | - Takeshi Oya
- Department of Molecular Pathology, Tokushima University Graduate School of Biomedical Sciences, Tokushima, Japan
| | - Koichi Tsuneyama
- Department of Pathology and Laboratory Medicine, Tokushima University Graduate School of Biomedical Sciences, Tokushima, Japan
| | - Mitsuru Matsumoto
- Division of Molecular Immunology, Institute for Enzyme Research, Tokushima University, Tokushima, Japan.
| | - Nuno L Alves
- Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal.
- Thymus Development and Function Laboratory, Instituto de Biologia Molecular e Celular, Porto, Portugal.
| |
Collapse
|
4
|
Pardini E, Barachini S, Alì G, Infirri GS, Burzi IS, Montali M, Petrini I. Single-cell sequencing has revealed a more complex array of thymic epithelial cells. Immunol Lett 2024; 269:106904. [PMID: 39117004 DOI: 10.1016/j.imlet.2024.106904] [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: 05/13/2024] [Revised: 07/23/2024] [Accepted: 07/29/2024] [Indexed: 08/10/2024]
Abstract
Thymic epithelial cells participate in the maturation and selection of T lymphocytes. This review explores recent insights from single-cell sequencing regarding classifying thymic epithelial cells in both normal and neoplastic thymus. Cortical thymic epithelial cells facilitate thymocyte differentiation and contribute to positive selection. Medullary epithelial cells are distinguished by their expression of AIRE. Cells progress from a pre-AIRE state, containing precursors with cortical and medullary characteristics, termed junctional cells. Mature medullary epithelial cells exhibit promiscuous gene expression and after that downregulate AIRE mRNA. Post-AIRE cells can adopt a Hassall corpuscle-like phenotype or exhibit distinctive differentiation characteristics including tuft cells, ionocytes, neuroendocrine cells, and myoid cells.
Collapse
Affiliation(s)
- Eleonora Pardini
- Department of Translational Research and New Surgical and Medical Technologies, University of Pisa, Pisa, Italy
| | - Serena Barachini
- Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy.
| | - Greta Alì
- Department of Surgical, Medical and Molecular Pathology and Critical Care Medicine, Pisa, Italy
| | - Gisella Sardo Infirri
- Department of Translational Research and New Surgical and Medical Technologies, University of Pisa, Pisa, Italy
| | - Irene Sofia Burzi
- Department of Translational Research and New Surgical and Medical Technologies, University of Pisa, Pisa, Italy
| | - Marina Montali
- Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy
| | - Iacopo Petrini
- Department of Translational Research and New Surgical and Medical Technologies, University of Pisa, Pisa, Italy
| |
Collapse
|
5
|
Yang Y, Jeong J, Su T, Lai S, Zhang P, Garcia-Milian R, Graham M, Liu X, McConnell MJ, Utsumi T, Pereira J, Iwakiri Y. Interleukin-7-based identification of liver lymphatic endothelial cells reveals their unique structural features. JHEP Rep 2024; 6:101069. [PMID: 38966234 PMCID: PMC11222939 DOI: 10.1016/j.jhepr.2024.101069] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/19/2023] [Revised: 03/01/2024] [Accepted: 03/11/2024] [Indexed: 07/06/2024] Open
Abstract
Background & Aims The lymphatic system plays crucial roles in maintaining fluid balance and immune regulation. Studying the liver lymphatics has been considered challenging, as common lymphatic endothelial cell (LyEC) markers are expressed by other liver cells. Additionally, isolation of sufficient numbers of LyECs has been challenging because of their extremely low abundance (<0.01% of entire liver cell population) in a normal liver. Methods Potential LyEC markers was identified using our published single-cell RNA sequencing (scRNA-seq) dataset (GSE147581) in mouse livers. Interleukin-7 (IL7) promoter-driven green fluorescent protein knock-in heterozygous mice were used for the validation of IL7 expression in LyECs in the liver, for the development of liver LyEC isolation protocol, and generating liver ischemia/reperfusion (I/R) injury. Scanning electron microscopy was used for the structural analysis of LyECs. Changes in LyEC phenotypes in livers of mice with I/R were determined by RNA-seq analysis. Results Through scRNA-seq analysis, we have identified IL7 as an exclusive marker for liver LyECs, with no overlap with other liver cell types. Based on IL7 expression in liver LyECs, we have established an LyEC isolation method and observed distinct cell surface structures of LyECs with fenestrae and cellular pores (ranging from 100 to 400 nm in diameter). Furthermore, we identified LyEC genes that undergo alterations during I/R liver injuries. Conclusions This study not only identified IL7 as an exclusively expressed gene in liver LyECs, but also enhanced our understanding of LyEC structures and demonstrated transcriptomic changes in injured livers. Impact and implications Understanding the lymphatic system in the liver is challenging because of the absence of specific markers for liver LyEC. This study has identified IL7 as a reliable marker for LyECs, enabling the development of an effective method for their isolation, elucidating their unique cell surface structure, and identifying LyEC genes that undergo changes during liver damage. The development of IL7 antibodies for detecting it in human liver specimens will further advance our understanding of the liver lymphatic system in the future.
Collapse
Affiliation(s)
- Yilin Yang
- Department of Internal Medicine, Section of Digestive Diseases, Yale University School of Medicine, New Haven, CT, USA
| | - Jain Jeong
- Department of Internal Medicine, Section of Digestive Diseases, Yale University School of Medicine, New Haven, CT, USA
| | - Tingting Su
- Department of Internal Medicine, Section of Digestive Diseases, Yale University School of Medicine, New Haven, CT, USA
- Department of Gastroenterology, First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Sanchuan Lai
- Department of Internal Medicine, Section of Digestive Diseases, Yale University School of Medicine, New Haven, CT, USA
- Department of Gastroenterology, Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Pengpeng Zhang
- Department of Internal Medicine, Section of Digestive Diseases, Yale University School of Medicine, New Haven, CT, USA
- The Transplantation Center of the Third Xiangya Hospital, Central South University, Changsha, China
| | - Rolando Garcia-Milian
- Bioinformatics Support Hub, Cushing/Whitney Medical Library, Yale University School of Medicine, New Haven, CT 06510, USA
| | - Morven Graham
- Center for Cellular and Molecular Imaging, Yale University School of Medicine, New Haven, CT, USA
| | - Xinran Liu
- Center for Cellular and Molecular Imaging, Yale University School of Medicine, New Haven, CT, USA
| | - Matthew J. McConnell
- Department of Internal Medicine, Section of Digestive Diseases, Yale University School of Medicine, New Haven, CT, USA
| | - Teruo Utsumi
- Department of Internal Medicine, Section of Digestive Diseases, Yale University School of Medicine, New Haven, CT, USA
| | - Joao Pereira
- Department of Immunobiology and Yale Stem Cell Center, Yale University School of Medicine, New Haven, CT, USA
| | - Yasuko Iwakiri
- Department of Internal Medicine, Section of Digestive Diseases, Yale University School of Medicine, New Haven, CT, USA
| |
Collapse
|
6
|
Liu T, Xia S. The Proteostasis of Thymic Stromal Cells in Health and Diseases. Protein J 2024; 43:447-463. [PMID: 38622349 DOI: 10.1007/s10930-024-10197-x] [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] [Accepted: 04/05/2024] [Indexed: 04/17/2024]
Abstract
The thymus is the key immune organ for the development of T cells. Different populations of thymic stromal cells interact with T cells, thereby controlling the dynamic development of T cells through their differentiation and function. Proteostasis represents a balance between protein expression, folding, and modification and protein clearance, and its fluctuation usually depends at least partially on related protein regulatory systems for further survival and effects. However, in terms of the substantial requirement for self-antigens and their processing burden, increasing evidence highlights that protein regulation contributes to the physiological effects of thymic stromal cells. Impaired proteostasis may expedite the progression of thymic involution and dysfunction, accompanied by the development of autoimmune diseases or thymoma. Hence, in this review, we summarize the regulation of proteostasis within different types of thymic stromal cells under physiological and pathological conditions to identify potential targets for thymic regeneration and immunotherapy.
Collapse
Affiliation(s)
- Ting Liu
- Department of Immunology, School of Medicine, Jiangsu University, 301, Xuefu Road, Zhenjiang, Jiangsu, 212013, China
| | - Sheng Xia
- Department of Immunology, School of Medicine, Jiangsu University, 301, Xuefu Road, Zhenjiang, Jiangsu, 212013, China.
| |
Collapse
|
7
|
Fujimori S, Ohigashi I. The role of thymic epithelium in thymus development and age-related thymic involution. THE JOURNAL OF MEDICAL INVESTIGATION 2024; 71:29-39. [PMID: 38735722 DOI: 10.2152/jmi.71.29] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/14/2024]
Abstract
The establishment of an adaptive immune system is critical for protecting our bodies from neoplastic cancers and invading pathogens such as viruses and bacteria. As a primary lymphoid organ, the thymus generates lymphoid T cells that play a major role in the adaptive immune system. T cell generation in the thymus is controlled by interactions between thymocytes and other thymic cells, primarily thymic epithelial cells. Thus, the normal development and function of thymic epithelial cells are important for the generation of immunocompetent and self-tolerant T cells. On the other hand, the degeneration of the thymic epithelium due to thymic aging causes thymic involution, which is associated with the decline of adaptive immune function. Herein we summarize basic and current knowledge of the development and function of thymic epithelial cells and the mechanism of thymic involution. J. Med. Invest. 71 : 29-39, February, 2024.
Collapse
Affiliation(s)
- Sayumi Fujimori
- Division of Experimental Immunology, Institute of Advanced Medical Sciences, Tokushima University, Tokushima, Japan
| | - Izumi Ohigashi
- Division of Experimental Immunology, Institute of Advanced Medical Sciences, Tokushima University, Tokushima, Japan
| |
Collapse
|
8
|
Lagou MK, Karagiannis GS. Obesity-induced thymic involution and cancer risk. Semin Cancer Biol 2023; 93:3-19. [PMID: 37088128 DOI: 10.1016/j.semcancer.2023.04.008] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2023] [Revised: 04/19/2023] [Accepted: 04/20/2023] [Indexed: 04/25/2023]
Abstract
Declining thymic functions associated either with old age (i.e., age-related thymic involution), or with acute involution as a result of stress, infectious disease, or cytoreductive therapies (e.g., chemotherapy/radiotherapy), have been associated with cancer development. A key mechanism underlying such increased cancer risk is the thymus-dependent debilitation of adaptive immunity, which is responsible for orchestrating immunoediting mechanisms and tumor immune surveillance. In the past few years, a blooming set of evidence has intriguingly linked obesity with cancer development and progression. The majority of such studies has focused on obesity-driven chronic inflammation, steroid/sex hormone and adipokine production, and hyperinsulinemia, as principal factors affecting the tumor microenvironment and driving the development of primary malignancy. However, experimental observations about the negative impact of obesity on T cell development and maturation have existed for more than half a century. Here, we critically discuss the molecular and cellular mechanisms of obesity-driven thymic involution as a previously underrepresented intermediary pathology leading to cancer development and progression. This knowledge could be especially relevant in the context of childhood obesity, because impaired thymic function in young individuals leads to immune system abnormalities, and predisposes to various pediatric cancers. A thorough understanding behind the molecular and cellular circuitries governing obesity-induced thymic involution could therefore help towards the rationalized development of targeted thymic regeneration strategies for obese individuals at high risk of cancer development.
Collapse
Affiliation(s)
- Maria K Lagou
- Department of Microbiology and Immunology, Albert Einstein College of Medicine, Bronx, NY, USA; Tumor Microenvironment of Metastasis Program, Albert Einstein Cancer Center, Bronx, NY, USA
| | - George S Karagiannis
- Department of Microbiology and Immunology, Albert Einstein College of Medicine, Bronx, NY, USA; Tumor Microenvironment of Metastasis Program, Albert Einstein Cancer Center, Bronx, NY, USA; Cancer Dormancy and Tumor Microenvironment Institute, Albert Einstein College of Medicine, Bronx, NY, USA; Gruss-Lipper Biophotonics Center, Albert Einstein College of Medicine, Bronx, NY, USA; Integrated Imaging Program for Cancer Research, Albert Einstein College of Medicine, Bronx, NY, USA.
| |
Collapse
|
9
|
Understanding the Roles of the Hedgehog Signaling Pathway during T-Cell Lymphopoiesis and in T-Cell Acute Lymphoblastic Leukemia (T-ALL). Int J Mol Sci 2023; 24:ijms24032962. [PMID: 36769284 PMCID: PMC9917970 DOI: 10.3390/ijms24032962] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2022] [Revised: 01/27/2023] [Accepted: 01/30/2023] [Indexed: 02/05/2023] Open
Abstract
The Hedgehog (HH) signaling network is one of the main regulators of invertebrate and vertebrate embryonic development. Along with other networks, such as NOTCH and WNT, HH signaling specifies both the early patterning and the polarity events as well as the subsequent organ formation via the temporal and spatial regulation of cell proliferation and differentiation. However, aberrant activation of HH signaling has been identified in a broad range of malignant disorders, where it positively influences proliferation, survival, and therapeutic resistance of neoplastic cells. Inhibitors targeting the HH pathway have been tested in preclinical cancer models. The HH pathway is also overactive in other blood malignancies, including T-cell acute lymphoblastic leukemia (T-ALL). This review is intended to summarize our knowledge of the biological roles and pathophysiology of the HH pathway during normal T-cell lymphopoiesis and in T-ALL. In addition, we will discuss potential therapeutic strategies that might expand the clinical usefulness of drugs targeting the HH pathway in T-ALL.
Collapse
|
10
|
Petrušić M, Stojić-Vukanić Z, Pilipović I, Kosec D, Prijić I, Leposavić G. Thymic changes as a contributing factor in the increased susceptibility of old Albino Oxford rats to EAE development. Exp Gerontol 2023; 171:112009. [PMID: 36334894 DOI: 10.1016/j.exger.2022.112009] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2022] [Revised: 10/26/2022] [Accepted: 10/27/2022] [Indexed: 11/06/2022]
Abstract
The study was aimed to examine putative contribution of thymic involution to ageing-associated increase in susceptibility of Albino Oxford (AO) rats to the development of clinical EAE, and vice versa influence of the disease on the progression of thymic involution. To this end we examined (i) the parameters of thymocyte negative selection efficacy, the thymic generation of CD4+CD25+Foxp3+ T regulatory cells (Tregs) and thymic capacity to instruct/predetermine IL-17-producing T-cell differentiation, and thymopietic efficacy-associated accumulation of "inflammescent" cytotoxic CD28- T cells in the periphery, and (ii) the key underlying mechanisms in young and old non-immunised AO rats and their counterparts immunised for EAE (on the 16th day post-immunisation when the disease in old rats reached the plateau) using flow cytometry analysis and/or RT-qPCR. It was found that thymic involution impairs: (i) the efficacy of negative selection (by affecting thymocyte expression of CD90, negative regulator of selection threshold and the expression of thymic stromal cell integrity factors) and (ii) Treg generation (by diminishing expression of cytokines supporting their differentiation/maturation). Additionally, the results suggest that thymic involution facilitates CD8+ T-cell differentiation into IL-17-producing cells (previously linked to the development of clinical EAE in old AO rats). Furthermore, they confirmed that ageing-related decrease in thymic T-cell output (as indicated by diminished frequency of recent thymic emigrants in peripheral blood) resulted in the accumulation of CD28- T cells in peripheral blood and, upon immunisation, in the target organ. On the other hand, the development of EAE (most likely by increasing circulatory levels of proinflammatory cytokines) contributed to the decline in thymic output of T cells, including Tregs, and thereby to the progression/maintenance of clinical EAE. Thus, in AO rats thymic involution via multi-layered mechanisms may favour the development of clinically manifested autoimmunity, which, in turn, precipitates the thymus atrophy.
Collapse
Affiliation(s)
- Marija Petrušić
- Department of Pathobiology, University of Belgrade, Faculty of Pharmacy, 450 Vojvode Stepe, 11221 Belgrade, Serbia
| | - Zorica Stojić-Vukanić
- Department of Microbiology and Immunology, University of Belgrade, Faculty of Pharmacy, 450 Vojvode Stepe, 11221 Belgrade, Serbia
| | - Ivan Pilipović
- Immunology Research Centre "Branislav Janković", Institute of Virology, Vaccines and Sera "Torlak", 458 Vojvode Stepe, 11221 Belgrade, Serbia
| | - Duško Kosec
- Immunology Research Centre "Branislav Janković", Institute of Virology, Vaccines and Sera "Torlak", 458 Vojvode Stepe, 11221 Belgrade, Serbia
| | - Ivana Prijić
- Immunology Research Centre "Branislav Janković", Institute of Virology, Vaccines and Sera "Torlak", 458 Vojvode Stepe, 11221 Belgrade, Serbia
| | - Gordana Leposavić
- Department of Pathobiology, University of Belgrade, Faculty of Pharmacy, 450 Vojvode Stepe, 11221 Belgrade, Serbia.
| |
Collapse
|
11
|
IL-7: Comprehensive review. Cytokine 2022; 160:156049. [DOI: 10.1016/j.cyto.2022.156049] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2022] [Revised: 09/08/2022] [Accepted: 09/16/2022] [Indexed: 01/08/2023]
|
12
|
Silva CS, Cerqueira MT, Reis RL, Martins A, Neves NM. Laminin-2 immobilized on a 3D fibrous structure impacts cortical thymic epithelial cells behaviour and their interaction with thymocytes. Int J Biol Macromol 2022; 222:3168-3177. [DOI: 10.1016/j.ijbiomac.2022.10.089] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2022] [Revised: 09/15/2022] [Accepted: 10/10/2022] [Indexed: 11/05/2022]
|
13
|
Yoganathan K, Yan A, Rocha J, Trotman-Grant A, Mohtashami M, Wells L, Zúñiga-Pflücker JC, Anderson MK. Regulation of the Signal-Dependent E Protein HEBAlt Through a YYY Motif Is Required for Progression Through T Cell Development. Front Immunol 2022; 13:848577. [PMID: 35990644 PMCID: PMC9385190 DOI: 10.3389/fimmu.2022.848577] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2022] [Accepted: 06/20/2022] [Indexed: 11/17/2022] Open
Abstract
The E protein transcription factors E2A and HEB are critical for many developmental processes, including T cell development. We have shown that the Tcf12 locus gives rise to two distinct HEB proteins, with alternative (HEBAlt) and canonical (HEBCan) N-terminal domains, which are co-expressed during early T cell development. While the functional domains of HEBCan have been well studied, the nature of the HEBAlt-specific (Alt) domain has been obscure. Here we provide compelling evidence that the Alt domain provides a site for the molecular integration of cytokine signaling and E protein activity. Our results indicate that phosphorylation of a unique YYY motif in the Alt domain increases HEBAlt activity by 10-fold, and that this increase is dependent on Janus kinase activity. To enable in vivo studies of HEBAlt in the T cell context, we generated ALT-Tg mice, which can be induced to express a HA-tagged HEBAlt coding cassette in the presence of Cre recombinases. Analysis of ALT-Tg mice on the Vav-iCre background revealed a minor change in the ratio of ISP cells to CD8+ SP cells, and a mild shift in the ratio of T cells to B cells in the spleen, but otherwise the thymus, spleen, and bone marrow lymphocyte subsets were comparable at steady state. However, kinetic analysis of T cell development in OP9-DL4 co-cultures revealed a delay in early T cell development and a partial block at the DN to DP transition when HEBAlt levels or activity were increased. We also observed that HEBCan and HEBAlt displayed significant differences in protein stability that were resolved in the thymocyte context. Finally, a proteomic screen identified STAT1 and Xpo1 as potential members of HEBAlt-containing complexes in thymocytes, consistent with JAK-induced activation of HEBAlt accompanied by translocation to the nucleus. Thus, our results show that the Alt domain confers access to multiple layers of post-translational control to HEBAlt that are not available to HEBCan, and thus may serve as a rheostat to tune E protein activity levels as cells move through different thymic signaling environments during T cell development.
Collapse
Affiliation(s)
- Kogulan Yoganathan
- Biological Sciences, Sunnybrook Research Institute, Toronto, ON, Canada
- Department of Immunology, University of Toronto, Toronto, ON, Canada
| | - Anqi Yan
- Biological Sciences, Sunnybrook Research Institute, Toronto, ON, Canada
- Department of Immunology, University of Toronto, Toronto, ON, Canada
| | - Juliana Rocha
- Biological Sciences, Sunnybrook Research Institute, Toronto, ON, Canada
| | - Ashton Trotman-Grant
- Biological Sciences, Sunnybrook Research Institute, Toronto, ON, Canada
- Department of Immunology, University of Toronto, Toronto, ON, Canada
| | | | - Lisa Wells
- Biological Sciences, Sunnybrook Research Institute, Toronto, ON, Canada
| | - Juan Carlos Zúñiga-Pflücker
- Biological Sciences, Sunnybrook Research Institute, Toronto, ON, Canada
- Department of Immunology, University of Toronto, Toronto, ON, Canada
| | - Michele K. Anderson
- Biological Sciences, Sunnybrook Research Institute, Toronto, ON, Canada
- Department of Immunology, University of Toronto, Toronto, ON, Canada
- *Correspondence: Michele K. Anderson,
| |
Collapse
|
14
|
Dong X, Liang Z, Zhang J, Zhang Q, Xu Y, Zhang Z, Zhang L, Zhang B, Zhao Y. Trappc1 deficiency impairs thymic epithelial cell development by breaking endoplasmic reticulum homeostasis. Eur J Immunol 2022; 52:1789-1804. [PMID: 35908180 DOI: 10.1002/eji.202249915] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2022] [Revised: 06/28/2022] [Accepted: 07/26/2022] [Indexed: 11/05/2022]
Abstract
Thymic epithelial cells (TECs) are important for T cell development and immune tolerance establishment. Although comprehensive molecular regulation of TEC development has been studied, the role of transport protein particle complexes (Trappcs) in TECs is not clear. Using TEC-specific homozygous or heterozygous Trappc1 deleted mice model, we found that Trappc1 deficiency caused severe thymus atrophy with decreased cell number and blocked maturation of TECs. Mice with a TEC-specific Trappc1 deletion show poor thymic T cell output and have a greater percentage of activated/memory T cells, suffered from spontaneous autoimmune disorders. Our RNA-seq and molecular studies indicated that the decreased endoplasmic reticulum (ER) and Golgi apparatus, enhanced unfolded protein response (UPR) and subsequent Atf4-CHOP-mediated apoptosis, and reactive oxygen species (ROS)-mediated ferroptosis coordinately contributed to the reduction of Trappc1-deleted TECs. Additionally, reduced Aire+ mTECs accompanied by the decreased expression of Irf4, Irf8, and Tbx21 in Trappc1 deficiency mTECs, may further coordinately block the tissue-restricted antigen expression. In this study, we reveal that Trappc1 plays an indispensable role in TEC development and maturation and provide evidence for the importance of inter-organelle traffic and ER homeostasis in TEC development. This article is protected by copyright. All rights reserved.
Collapse
Affiliation(s)
- Xue Dong
- State Key Laboratory of Membrane Biology, Institute of Zoology, Chinese Academy of Sciences.,University of Chinese Academy of Sciences
| | - Zhanfeng Liang
- State Key Laboratory of Membrane Biology, Institute of Zoology, Chinese Academy of Sciences.,University of Chinese Academy of Sciences.,Beijing Institute for Stem Cell and Regeneration
| | - Jiayu Zhang
- State Key Laboratory of Membrane Biology, Institute of Zoology, Chinese Academy of Sciences.,University of Chinese Academy of Sciences
| | - Qian Zhang
- State Key Laboratory of Membrane Biology, Institute of Zoology, Chinese Academy of Sciences.,University of Chinese Academy of Sciences
| | - Yanan Xu
- State Key Laboratory of Membrane Biology, Institute of Zoology, Chinese Academy of Sciences.,University of Chinese Academy of Sciences
| | - Zhaoqi Zhang
- State Key Laboratory of Membrane Biology, Institute of Zoology, Chinese Academy of Sciences.,University of Chinese Academy of Sciences
| | - Lianfeng Zhang
- Key Laboratory of Human Diseases and Comparative Medicine, Ministry of Health, Institute of Laboratory Animal Science, Chinese Academy of Medical Sciences
| | - Baojun Zhang
- Department of Pathogenic Microbiology and Immunology, School of Basic Medical Sciences, Xi'an Jiaotong University
| | - Yong Zhao
- State Key Laboratory of Membrane Biology, Institute of Zoology, Chinese Academy of Sciences.,University of Chinese Academy of Sciences.,Beijing Institute for Stem Cell and Regeneration
| |
Collapse
|
15
|
IL-7 promoted the development of thymic DN3 cells in aged mice via DNA demethylation of Bcl2 and c-Myc genes. Mol Immunol 2022; 147:21-29. [DOI: 10.1016/j.molimm.2022.04.013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2021] [Revised: 03/11/2022] [Accepted: 04/21/2022] [Indexed: 11/18/2022]
|
16
|
Shichkin VP, Antica M. Key Factors for Thymic Function and Development. Front Immunol 2022; 13:926516. [PMID: 35844535 PMCID: PMC9280625 DOI: 10.3389/fimmu.2022.926516] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2022] [Accepted: 05/31/2022] [Indexed: 11/17/2022] Open
Abstract
The thymus is the organ responsible for T cell development and the formation of the adaptive immunity function. Its multicellular environment consists mainly of the different stromal cells and maturing T lymphocytes. Thymus-specific progenitors of epithelial, mesenchymal, and lymphoid cells with stem cell properties represent only minor populations. The thymic stromal structure predominantly determines the function of the thymus. The stromal components, mostly epithelial and mesenchymal cells, form this specialized area. They support the consistent developmental program of functionally distinct conventional T cell subpopulations. These include the MHC restricted single positive CD4+ CD8- and CD4- CD8+ cells, regulatory T lymphocytes (Foxp3+), innate natural killer T cells (iNKT), and γδT cells. Several physiological causes comprising stress and aging and medical treatments such as thymectomy and chemo/radiotherapy can harm the thymus function. The present review summarizes our knowledge of the development and function of the thymus with a focus on thymic epithelial cells as well as other stromal components and the signaling and transcriptional pathways underlying the thymic cell interaction. These critical thymus components are significant for T cell differentiation and restoring the thymic function after damage to reach the therapeutic benefits.
Collapse
|
17
|
Bhalla P, Su DM, van Oers NSC. Thymus Functionality Needs More Than a Few TECs. Front Immunol 2022; 13:864777. [PMID: 35757725 PMCID: PMC9229346 DOI: 10.3389/fimmu.2022.864777] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2022] [Accepted: 05/03/2022] [Indexed: 12/18/2022] Open
Abstract
The thymus, a primary lymphoid organ, produces the T cells of the immune system. Originating from the 3rd pharyngeal pouch during embryogenesis, this organ functions throughout life. Yet, thymopoiesis can be transiently or permanently damaged contingent on the types of systemic stresses encountered. The thymus also undergoes a functional decline during aging, resulting in a progressive reduction in naïve T cell output. This atrophy is evidenced by a deteriorating thymic microenvironment, including, but not limited, epithelial-to-mesenchymal transitions, fibrosis and adipogenesis. An exploration of cellular changes in the thymus at various stages of life, including mouse models of in-born errors of immunity and with single cell RNA sequencing, is revealing an expanding number of distinct cell types influencing thymus functions. The thymus microenvironment, established through interactions between immature and mature thymocytes with thymus epithelial cells (TEC), is well known. Less well appreciated are the contributions of neural crest cell-derived mesenchymal cells, endothelial cells, diverse hematopoietic cell populations, adipocytes, and fibroblasts in the thymic microenvironment. In the current review, we will explore the contributions of the many stromal cell types participating in the formation, expansion, and contraction of the thymus under normal and pathophysiological processes. Such information will better inform approaches for restoring thymus functionality, including thymus organoid technologies, beneficial when an individuals’ own tissue is congenitally, clinically, or accidentally rendered non-functional.
Collapse
Affiliation(s)
- Pratibha Bhalla
- Department of Immunology, The University of Texas Southwestern Medical Center, Dallas, TX, United States
| | - Dong-Ming Su
- Department of Microbiology, Immunology & Genetics, The University of North Texas Health Sciences Center, Fort Worth, TX, United States
| | - Nicolai S C van Oers
- Department of Immunology, The University of Texas Southwestern Medical Center, Dallas, TX, United States.,Department of Microbiology, The University of Texas Southwestern Medical Center, Dallas, TX, United States.,Department of Pediatrics, The University of Texas Southwestern Medical Center, Dallas, TX, United States
| |
Collapse
|
18
|
Structural and Functional Thymic Biomarkers Are Involved in the Pathogenesis of Thymic Epithelial Tumors: An Overview. IMMUNO 2022. [DOI: 10.3390/immuno2020025] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
Abstract
The normal human thymus originates from the third branchial cleft as two paired anlages that descend into the thorax and fuse on the midline of the anterior–superior mediastinum. Alongside the epithelial and lymphoid components, different types of lymphoid accessory cells, stromal mesenchymal and endothelial cells migrate to, or develop in, the thymus. After reaching maximum development during early postnatal life, the human thymus decreases in size and lymphocyte output drops with age. However, thymic immunological functions persist, although they deteriorate progressively. Several major techniques were fundamental to increasing the knowledge of thymic development and function during embryogenesis, postnatal and adult life; these include immunohistochemistry, immunofluorescence, flow cytometry, in vitro colony assays, transplantation in mice models, fetal organ cultures (FTOC), re-aggregated thymic organ cultures (RTOC), and whole-organ thymic scaffolds. The thymic morphological and functional characterization, first performed in the mouse, was then extended to humans. The purpose of this overview is to provide a report on selected structural and functional biomarkers of thymic epithelial cells (TEC) involved in thymus development and lymphoid cell maturation, and on the historical aspects of their characterization, with particular attention being paid to biomarkers also involved in Thymic Epithelial Tumor (TET) pathogenesis. Moreover, a short overview of targeted therapies in TET, based on currently available experimental and clinical data and on potential future advances will be proposed.
Collapse
|
19
|
Abstract
A high diversity of αβ T cell receptors (TCRs), capable of recognizing virtually any pathogen but also self-antigens, is generated during T cell development in the thymus. Nevertheless, a strict developmental program supports the selection of a self-tolerant T cell repertoire capable of responding to foreign antigens. The steps of T cell selection are controlled by cortical and medullary stromal niches, mainly composed of thymic epithelial cells and dendritic cells. The integration of important cues provided by these specialized niches, including (a) the TCR signal strength induced by the recognition of self-peptide-MHC complexes, (b) costimulatory signals, and (c) cytokine signals, critically controls T cell repertoire selection. This review discusses our current understanding of the signals that coordinate positive selection, negative selection, and agonist selection of Foxp3+ regulatory T cells. It also highlights recent advances that have unraveled the functional diversity of thymic antigen-presenting cell subsets implicated in T cell selection.
Collapse
Affiliation(s)
- Magali Irla
- Centre d'Immunologie de Marseille-Luminy (CIML), CNRS, INSERM, Aix-Marseille Université, Marseille, France;
| |
Collapse
|
20
|
Park JY, Won HY, DiPalma DT, Kim HK, Kim TH, Li C, Sato N, Hong C, Abraham N, Gress RE, Park JH. In vivo availability of the cytokine IL-7 constrains the survival and homeostasis of peripheral iNKT cells. Cell Rep 2022; 38:110219. [PMID: 35021100 DOI: 10.1016/j.celrep.2021.110219] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2020] [Revised: 10/06/2021] [Accepted: 12/14/2021] [Indexed: 11/03/2022] Open
Abstract
Understanding the homeostatic mechanism of invariant natural killer T (iNKT) cells is a critical issue in iNKT cell biology. Because interleukin (IL)-15 is required for the thymic generation of iNKT cells, IL-15 has also been considered necessary for the homeostasis of peripheral iNKT cells. Here, we delineated the in vivo cytokine requirement for iNKT cells, and we came to the surprising conclusion that IL-7, not IL-15, is the homeostatic cytokine for iNKT cells. Employing a series of experimental mouse models where the availability of IL-7 or IL-15 was manipulated in peripheral tissues, either by genetic tools or by adult thymectomy and cytokine pump installation, we demonstrate that the abundance of IL-7, and not IL-15, limits the size of the peripheral iNKT cell pool. These results redefine the cytokine requirement for iNKT cells and indicate competition for IL-7 between iNKT and conventional αβ T cells.
Collapse
Affiliation(s)
- Joo-Young Park
- Experimental Immunology Branch, Center for Cancer Research, National Cancer Institute, NIH, Building 10, Room 5B17, 10 Center Drive, Bethesda, MD 20892, USA; Department of Oral and Maxillofacial Surgery, Seoul National University School of Dentistry, Seoul National University Dental Hospital, 101 Daehakno, Jongno-gu, Seoul 03080, South Korea.
| | - Hee Yeun Won
- Experimental Immunology Branch, Center for Cancer Research, National Cancer Institute, NIH, Building 10, Room 5B17, 10 Center Drive, Bethesda, MD 20892, USA
| | - Devon T DiPalma
- Experimental Immunology Branch, Center for Cancer Research, National Cancer Institute, NIH, Building 10, Room 5B17, 10 Center Drive, Bethesda, MD 20892, USA
| | - Hye Kyung Kim
- Experimental Transplantation Immunology Branch, Center for Cancer Research, National Cancer Institute, NIH, Bethesda, MD 20892, USA
| | - Tae-Hyoun Kim
- Experimental Immunology Branch, Center for Cancer Research, National Cancer Institute, NIH, Building 10, Room 5B17, 10 Center Drive, Bethesda, MD 20892, USA
| | - Can Li
- Experimental Immunology Branch, Center for Cancer Research, National Cancer Institute, NIH, Building 10, Room 5B17, 10 Center Drive, Bethesda, MD 20892, USA
| | - Noriko Sato
- Molecular Imaging Branch, Center for Cancer Research, National Cancer Institute, NIH, Bethesda, MD 20892, USA
| | - Changwan Hong
- Department of Anatomy, Pusan National University School of Medicine, Yangsan 626-870, South Korea
| | - Ninan Abraham
- Department of Microbiology and Immunology, and Department of Zoology, Life Sciences Institute, University of British Columbia, Vancouver, BC V6T 1Z3, Canada
| | - Ronald E Gress
- Experimental Transplantation Immunology Branch, Center for Cancer Research, National Cancer Institute, NIH, Bethesda, MD 20892, USA
| | - Jung-Hyun Park
- Experimental Immunology Branch, Center for Cancer Research, National Cancer Institute, NIH, Building 10, Room 5B17, 10 Center Drive, Bethesda, MD 20892, USA.
| |
Collapse
|
21
|
ILC Differentiation in the Thymus. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2022; 1365:25-39. [DOI: 10.1007/978-981-16-8387-9_3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
|
22
|
Ikuta K, Hara T, Abe S, Asahi T, Takami D, Cui G. The Roles of IL-7 and IL-15 in Niches for Lymphocyte Progenitors and Immune Cells in Lymphoid Organs. Curr Top Microbiol Immunol 2021; 434:83-101. [PMID: 34850283 DOI: 10.1007/978-3-030-86016-5_4] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
Abstract
Lymphoid organs consist of immune cells and stromal cells. The stromal cells produce various cytokines that support the development, maintenance, and response of the immune cells. IL-7 and IL-15 are the major cytokines produced by stromal cells and are essential for the development and maintenance of lymphocytes and innate lymphoid cells (ILCs). In addition, IL-7 is indispensable for the organogenesis of lymphoid organs. However, because the amount of these two cytokines is relatively low, it has been difficult to directly detect their expression. Recently, several groups succeeded in establishing IL-7 and IL-15 reporter mouse lines. As expected, IL-7 and IL-15 were detected in mesenchymal stromal cells in the bone marrow and lymph nodes and in epithelial cells in the thymus. Furthermore, IL-7 and IL-15 were differentially expressed in lymphatic endothelial cells and blood endothelial cells, respectively. In addition to their expression, many groups have analyzed the local functions of IL-7 and IL-15 by using cell-type-specific knockout mice. From these experiments, CXCL12-expressing mesenchymal stromal cells were identified as the major niche for early B cell precursors. Single-cell RNA sequencing (scRNA-seq) analysis has revealed different subpopulations of stromal cells in the lymphoid organs, including those that express both IL-7 and IL-15. Future research is still needed to elucidate which stromal cells serve as the niche for the early precursors of ILCs and NK cells in the bone marrow.
Collapse
Affiliation(s)
- Koichi Ikuta
- Laboratory of Immune Regulation, Department of Virus Research, Institute for Frontier Life and Medical Sciences, Kyoto University, Kyoto, 606-8507, Japan.
| | - Takahiro Hara
- Laboratory of Immune Regulation, Department of Virus Research, Institute for Frontier Life and Medical Sciences, Kyoto University, Kyoto, 606-8507, Japan
| | - Shinya Abe
- Laboratory of Immune Regulation, Department of Virus Research, Institute for Frontier Life and Medical Sciences, Kyoto University, Kyoto, 606-8507, Japan
| | - Takuma Asahi
- Laboratory of Immune Regulation, Department of Virus Research, Institute for Frontier Life and Medical Sciences, Kyoto University, Kyoto, 606-8507, Japan.,Graduate School of Medicine, Kyoto University, Kyoto, 606-8501, Japan
| | - Daichi Takami
- Laboratory of Immune Regulation, Department of Virus Research, Institute for Frontier Life and Medical Sciences, Kyoto University, Kyoto, 606-8507, Japan.,Graduate School of Pharmaceutical Sciences, Kyoto University, Kyoto, 606-8501, Japan
| | - Guangwei Cui
- Laboratory of Immune Regulation, Department of Virus Research, Institute for Frontier Life and Medical Sciences, Kyoto University, Kyoto, 606-8507, Japan
| |
Collapse
|
23
|
Tao Z, Jiang Y, Xia S. Regulation of thymic T regulatory cell differentiation by TECs in health and disease. Scand J Immunol 2021; 94:e13094. [PMID: 34780092 DOI: 10.1111/sji.13094] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2021] [Revised: 07/26/2021] [Accepted: 07/27/2021] [Indexed: 12/22/2022]
Abstract
The thymus produces self-limiting and self-tolerant T cells through the interaction between thymocytes and thymus epithelial cells (TECs), thereby generating central immune tolerance. The TECs are composed of cortical and medullary thymic epithelial cells, which regulate the positive and negative selection of T cells, respectively. During the process of negative selection, thymocytes with self-reactive ability are deleted or differentiated into regulatory T cells (Tregs). Tregs are a subset of suppressor T cells that are important for maintaining immune homeostasis. The differentiation and development of Tregs depend on the development of TECs and other underlying molecular mechanisms. Tregs regulated by thymic epithelial cells are closely related to human health and are significant in autoimmune diseases, thymoma and pregnancy. In this review, we summarize the current molecular and transcriptional regulatory mechanisms by which TECs affect the development and function of thymic Tregs. We also review the pathophysiological models of thymic epithelial cells regulating thymic Tregs in human diseases and specific physiological conditions.
Collapse
Affiliation(s)
- Zehua Tao
- Department of Immunology, School of Medicine, Jiangsu University, Zhenjiang, China
| | - Yalan Jiang
- Department of Immunology, School of Medicine, Jiangsu University, Zhenjiang, China
| | - Sheng Xia
- Department of Immunology, School of Medicine, Jiangsu University, Zhenjiang, China
| |
Collapse
|
24
|
Abstract
Conventional CD4+ and CD8+ T lymphocytes comprise a mixture of naive and memory cells. Generation and survival of these T-cell subsets is under strict homeostatic control and reflects contact with self-major histocompatibility complex (MHC) and certain cytokines. Naive T cells arise in the thymus via T-cell receptor (TCR)-dependent positive selection to self-peptide/MHC complexes and are then maintained in the periphery through self-MHC interaction plus stimulation via interleukin-7 (IL-7). By contrast, memory T cells are largely MHC-independent for their survival but depend strongly on stimulation via cytokines. Whereas typical memory T cells are generated in response to foreign antigens, some arise spontaneously through contact of naive precursors with self-MHC ligands; we refer to these cells as memory-phenotype (MP) T cells. In this review, we discuss the generation and homeostasis of naive T cells and these two types of memory T cells, focusing on their relative interaction with MHC ligands and cytokines.
Collapse
Affiliation(s)
- Takeshi Kawabe
- Department of Microbiology and Immunology, Tohoku University Graduate School of Medicine, Sendai, Miyagi 980-8575, Japan
| | - Jaeu Yi
- Division of Rheumatology, Department of Internal Medicine, Washington University School of Medicine, St. Louis, Missouri 63110, USA
| | - Jonathan Sprent
- Immunology Division, Garvan Institute of Medical Research, Darlinghurst, New South Wales 2010, Australia
- St. Vincent's Clinical School, University of New South Wales, Sydney, New South Wales 2010, Australia
| |
Collapse
|
25
|
Aghaallaei N, Dick AM, Tsingos E, Inoue D, Hasel E, Thumberger T, Toyoda A, Leptin M, Wittbrodt J, Bajoghli B. αβ/γδ T cell lineage outcome is regulated by intrathymic cell localization and environmental signals. SCIENCE ADVANCES 2021; 7:7/29/eabg3613. [PMID: 34261656 PMCID: PMC8279519 DOI: 10.1126/sciadv.abg3613] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/30/2020] [Accepted: 05/28/2021] [Indexed: 05/07/2023]
Abstract
αβ and γδ T cells are two distinct sublineages that develop in the vertebrate thymus. Thus far, their differentiation from a common progenitor is mostly understood to be regulated by intrinsic mechanisms. However, the proportion of αβ/γδ T cells varies in different vertebrate taxa. How this process is regulated in species that tend to produce a high frequency of γδ T cells is unstudied. Using an in vivo teleost model, the medaka, we report that progenitors first enter a thymic niche where their development into γδ T cells is favored. Translocation from this niche, mediated by chemokine receptor Ccr9b, is a prerequisite for their differentiation into αβ T cells. On the other hand, the thymic niche also generates opposing gradients of the cytokine interleukin-7 and chemokine Ccl25a, and, together, they influence the lineage outcome. We propose a previously unknown mechanism that determines the proportion of αβ/γδ lineages within species.
Collapse
Affiliation(s)
- Narges Aghaallaei
- Department of Hematology, Oncology, Immunology, and Rheumatology, University Hospital of Tübingen, Otfried-Müller-Strasse 10, 72076 Tübingen, Germany
- Centre for Organismal Studies (COS), Heidelberg University, Im Neuenheimer Feld 230, 69120 Heidelberg, Germany
| | - Advaita M Dick
- Department of Hematology, Oncology, Immunology, and Rheumatology, University Hospital of Tübingen, Otfried-Müller-Strasse 10, 72076 Tübingen, Germany
| | - Erika Tsingos
- Centre for Organismal Studies (COS), Heidelberg University, Im Neuenheimer Feld 230, 69120 Heidelberg, Germany
| | - Daigo Inoue
- Centre for Organismal Studies (COS), Heidelberg University, Im Neuenheimer Feld 230, 69120 Heidelberg, Germany
| | - Eva Hasel
- European Molecular Biology Laboratory (EMBL), Meyerhofstrasse 1, 69117 Heidelberg, Germany
| | - Thomas Thumberger
- Centre for Organismal Studies (COS), Heidelberg University, Im Neuenheimer Feld 230, 69120 Heidelberg, Germany
| | - Atsushi Toyoda
- Comparative Genomics Laboratory, Department of Genomics and Evolutionary Biology, National Institute of Genetics, Yata 1111, Mishima, Shizuoka 411-8540, Japan
| | - Maria Leptin
- European Molecular Biology Laboratory (EMBL), Meyerhofstrasse 1, 69117 Heidelberg, Germany
- EMBO, Meyerhofstrasse 1, 69117 Heidelberg, Germany
| | - Joachim Wittbrodt
- Centre for Organismal Studies (COS), Heidelberg University, Im Neuenheimer Feld 230, 69120 Heidelberg, Germany
| | - Baubak Bajoghli
- Department of Hematology, Oncology, Immunology, and Rheumatology, University Hospital of Tübingen, Otfried-Müller-Strasse 10, 72076 Tübingen, Germany.
- European Molecular Biology Laboratory (EMBL), Meyerhofstrasse 1, 69117 Heidelberg, Germany
| |
Collapse
|
26
|
Pinheiro RGR, Alves NL. The Early Postnatal Life: A Dynamic Period in Thymic Epithelial Cell Differentiation. Front Immunol 2021; 12:668528. [PMID: 34220815 PMCID: PMC8250140 DOI: 10.3389/fimmu.2021.668528] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2021] [Accepted: 06/04/2021] [Indexed: 11/20/2022] Open
Abstract
The microenvironments formed by cortical (c) and medullary (m) thymic epithelial cells (TECs) play a non-redundant role in the generation of functionally diverse and self-tolerant T cells. The role of TECs during the first weeks of the murine postnatal life is particularly challenging due to the significant augment in T cell production. Here, we critically review recent studies centered on the timely coordination between the expansion and maturation of TECs during this period and their specialized role in T cell development and selection. We further discuss how aging impacts on the pool of TEC progenitors and maintenance of functionally thymic epithelial microenvironments, and the implications of these chances in the capacity of the thymus to sustain regular thymopoiesis throughout life.
Collapse
Affiliation(s)
- Ruben G R Pinheiro
- i3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal.,IBMC - Instituto de Biologia Molecular e Celular, Universidade do Porto, Porto, Portugal.,Doctoral Program in Cell and Molecular Biology, Instituto de Ciências Biomédicas, Universidade do Porto, Porto, Portugal
| | - Nuno L Alves
- i3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal.,IBMC - Instituto de Biologia Molecular e Celular, Universidade do Porto, Porto, Portugal
| |
Collapse
|
27
|
Han J, Zúñiga-Pflücker JC. A 2020 View of Thymus Stromal Cells in T Cell Development. THE JOURNAL OF IMMUNOLOGY 2021; 206:249-256. [PMID: 33397738 DOI: 10.4049/jimmunol.2000889] [Citation(s) in RCA: 40] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/28/2020] [Accepted: 10/29/2020] [Indexed: 12/12/2022]
Abstract
The thymus is an intricate primary lymphoid organ, wherein bone marrow-derived lymphoid progenitor cells are induced to develop into functionally competent T cells that express a diverse TCR repertoire, which is selected to allow for the recognition of foreign Ags while avoiding self-reactivity or autoimmunity. Thymus stromal cells, which can include all non-T lineage cells, such as thymic epithelial cells, endothelial cells, mesenchymal/fibroblast cells, dendritic cells, and B cells, provide signals that are essential for thymocyte development as well as for the homeostasis of the thymic stroma itself. In this brief review, we focus on the key roles played by thymic stromal cells during early stages of T cell development, such as promoting the homing of thymic-seeding progenitors, inducing T lineage differentiation, and supporting thymocyte survival and proliferation. We also discuss recent advances on the transcriptional regulation that govern thymic epithelial cell function as well as the cellular and molecular changes that are associated with thymic involution and regeneration.
Collapse
Affiliation(s)
- Jianxun Han
- Sunnybrook Research Institute, Toronto, Ontario M4N 3M5, Canada; and
| | - Juan Carlos Zúñiga-Pflücker
- Sunnybrook Research Institute, Toronto, Ontario M4N 3M5, Canada; and.,Department of Immunology, University of Toronto, Toronto, Ontario M5S 1A8, Canada
| |
Collapse
|
28
|
Podlesnaja M, Pilmane M, Sumeraga G. Cytokines, Proliferation Markers, Antimicrobial Factors and Neuropeptide-Containing Innervation in Human Nasal Mucosa after Rhinoseptoplasty Procedure. Med Sci (Basel) 2021; 9:medsci9020025. [PMID: 33919374 PMCID: PMC8167725 DOI: 10.3390/medsci9020025] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2021] [Revised: 04/16/2021] [Accepted: 04/19/2021] [Indexed: 12/19/2022] Open
Abstract
The nasal cavity lined by nasal mucosa, is a significant part of respiratory system of human. However, there are no studies aimed to detect a molecular phenotype of healthy and normal functioning nasal mucosa, obtained after rhinoseptoplasty procedure, to understand its physiology and growth and inflammation processes. Thus, our aim is to identify human healthy nasal mucosa cytokines, neuropeptide-containing innervation and cell proliferation markers to form a control group for further tissue investigation of human nasal polyposis as the next step of our research. The study included surgery materials from 17 healthy humans. Biotin-streptavidin immunohistochemistry was performed for detection of tissue PGP9.5, Ki-67, β-Defensin 2, IL-1, IL-4, IL-6, IL-7, IL-8, IL-10, IL-12. Results were evaluated semi-quantitatively and by Friedman ANOVA and Spearman rang correlation tests. All factors were more widely expressed by superficial epithelium than by glandular one. Abundance of ILs-8, -10 and -12 positive cells was detected in comparison with moderate to numerous distributions of IL-1, IL-6 and β-Defensin 2. Moderate number of PGP 9.5-containing nerve fibers and only few to moderate Ki-67 positive cells were found in healthy nasal mucosa. We revealed statistically significant difference between Ki-67 and ILs-4, -6, -7, -8, -10, -12 both in healthy nasal mucosa superficial and glandular epithelium. From nasal epithelia, commonly the surface one displays more cytokines and β-Defensin 2 in comparison to the glandular one. Numerous to abundant expression of ILs-4, -6, -7, -8, -10, -12 and β-Defensin 2 in nasal superficial and glandular epithelia proves probably these factors' role into the common immune response of tissue and stimulation of immune cell differentiation.
Collapse
Affiliation(s)
- Marija Podlesnaja
- Institute of Anatomy and Anthropology, Riga Stradins University, LV-1010 Riga, Latvia;
- Correspondence:
| | - Mara Pilmane
- Institute of Anatomy and Anthropology, Riga Stradins University, LV-1010 Riga, Latvia;
| | - Gunta Sumeraga
- Department of Otorhinolaryngology, Pauls Stradiņš Clinical University Hospital, LV-1002 Riga, Latvia;
| |
Collapse
|
29
|
Bednarz-Misa I, Bromke MA, Krzystek-Korpacka M. Interleukin (IL)-7 Signaling in the Tumor Microenvironment. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2021; 1290:9-49. [PMID: 33559853 DOI: 10.1007/978-3-030-55617-4_2] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Interleukin (IL)-7 plays an important immunoregulatory role in different types of cells. Therefore, it attracts researcher's attention, but despite the fact, many aspects of its modulatory action, as well as other functionalities, are still poorly understood. The review summarizes current knowledge on the interleukin-7 and its signaling cascade in context of cancer development. Moreover, it provides a cancer-type focused description of the involvement of IL-7 in solid tumors, as well as hematological malignancies.The interleukin has been discovered as a growth factor crucial for the early lymphocyte development and supporting the growth of malignant cells in certain leukemias and lymphomas. Therefore, its targeting has been explored as a treatment modality in hematological malignancies, while the unique ability to expand lymphocyte populations selectively and without hyperinflammation has been used in experimental immunotherapies in patients with lymphopenia. Ever since the early research demonstrated a reduced growth of solid tumors in the presence of IL-7, the interleukin application in boosting up the anticancer immunity has been investigated. However, a growing body of evidence indicative of IL-7 upregulation in carcinomas, facilitating tumor growth and metastasis and aiding drug-resistance, is accumulating. It therefore becomes increasingly apparent that the response to the IL-7 stimulus strongly depends on cell type, their developmental stage, and microenvironmental context. The interleukin exerts its regulatory action mainly through phosphorylation events in JAK/STAT and PI3K/Akt pathways, while the significance of MAPK pathway seems to be limited to solid tumors. Given the unwavering interest in IL-7 application in immunotherapy, a better understanding of interleukin role, source in tumor microenvironment, and signaling pathways, as well as the identification of cells that are likely to respond should be a research priority.
Collapse
Affiliation(s)
- Iwona Bednarz-Misa
- Department of Medical Biochemistry, Wroclaw Medical University, Wroclaw, Poland
| | - Mariusz A Bromke
- Department of Medical Biochemistry, Wroclaw Medical University, Wroclaw, Poland
| | | |
Collapse
|
30
|
Irla M. RANK Signaling in the Differentiation and Regeneration of Thymic Epithelial Cells. Front Immunol 2021; 11:623265. [PMID: 33552088 PMCID: PMC7862717 DOI: 10.3389/fimmu.2020.623265] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2020] [Accepted: 12/07/2020] [Indexed: 11/24/2022] Open
Abstract
Thymic epithelial cells (TECs) provide essential clues for the proliferation, survival, migration, and differentiation of thymocytes. Recent advances in mouse and human have revealed that TECs constitute a highly heterogeneous cell population with distinct functional properties. Importantly, TECs are sensitive to thymic damages engendered by myeloablative conditioning regimen used for bone marrow transplantation. These detrimental effects on TECs delay de novo T-cell production, which can increase the risk of morbidity and mortality in many patients. Alike that TECs guide the development of thymocytes, reciprocally thymocytes control the differentiation and organization of TECs. These bidirectional interactions are referred to as thymic crosstalk. The tumor necrosis factor receptor superfamily (TNFRSF) member, receptor activator of nuclear factor kappa-B (RANK) and its cognate ligand RANKL have emerged as key players of the crosstalk between TECs and thymocytes. RANKL, mainly provided by positively selected CD4+ thymocytes and a subset of group 3 innate lymphoid cells, controls mTEC proliferation/differentiation and TEC regeneration. In this review, I discuss recent advances that have unraveled the high heterogeneity of TECs and the implication of the RANK-RANKL signaling axis in TEC differentiation and regeneration. Targeting this cell-signaling pathway opens novel therapeutic perspectives to recover TEC function and T-cell production.
Collapse
Affiliation(s)
- Magali Irla
- Aix Marseille Univ, CNRS, INSERM, CIML, Centre d'Immunologie de Marseille-Luminy, Marseille, France
| |
Collapse
|
31
|
Trujillo-Ocampo A, Cho HW, Clowers M, Pareek S, Ruiz-Vazquez W, Lee SE, Im JS. IL-7 During Antigenic Stimulation Using Allogeneic Dendritic Cells Promotes Expansion of CD45RA -CD62L +CD4 + Invariant NKT Cells With Th-2 Biased Cytokine Production Profile. Front Immunol 2020; 11:567406. [PMID: 33329531 PMCID: PMC7728799 DOI: 10.3389/fimmu.2020.567406] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2020] [Accepted: 10/29/2020] [Indexed: 11/18/2022] Open
Abstract
Invariant natural killer T (iNKT) cells are innate-like T lymphocytes cells that recognize glycolipid antigens associated with CD1d, non-classical antigen presenting proteins. They can drive either pro-inflammatory (Th-1) or anti-inflammatory (Th-2) immune microenvironment through the production of both Th-1 and Th-2 type cytokines upon activation, thus play a vital role in cancer, infection, and autoimmune diseases. Adoptive cell therapy using ex vivo expanded iNKT cells is a promising approach to enhance anti-tumor immunity or immunosuppression. However, overcoming phenotypic and functional heterogeneity and promoting in vivo persistency of iNKT cells remains to be a challenge. Here, we compared various methods for ex vivo expansion of human iNKT cells and assessed the quality of expansion, phenotype, and cytokine production profile of expanded iNKT cells. While a direct stimulation of iNKT cells in peripheral blood mononuclear cells with agonist glycolipid led to the expansion of iNKT cells in varying degrees, stimulation of enriched iNKT cells by irradiated autologous peripheral blood mononuclear cells or allogeneic dendritic cells resulted in consistent expansion of highly pure iNKT cells. Interestingly, the mode of antigenic stimulation influenced the dominant subtype of expanded iNKT cells. Further, we evaluated whether additional IL-7 or IL-15 during antigenic stimulation with allogeneic dendritic cells can improve the phenotypic heterogeneity and modify cytokine production profile of iNKT cells expanded from 18 consecutive donors. The presence of IL-7 or IL-15 during antigenic stimulation did not affect the fold of expansion or purity of expanded iNKT cells. However, IL-7, but not IL-15, led to a better expansion of CD4+ iNKT cells, enhanced Th-2 type cytokine production of CD4+ iNKT cells, and maintained the expansion of central memory (CD45RA-CD62L+) CD4+ iNKT cells. Our results suggest the addition of IL-7 during antigenic stimulation with allogeneic dendritic cells can promote the expansion of CD62L+Th-2+CD4+ human iNKT cells that can be used as novel immunotherapeutic to control excessive inflammation to treat various autoimmune diseases.
Collapse
Affiliation(s)
- Abel Trujillo-Ocampo
- Department of Stem Cell Transplantation and Cellular Therapy, University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Hyun-Woo Cho
- Department of Stem Cell Transplantation and Cellular Therapy, University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Michael Clowers
- Department of Stem Cell Transplantation and Cellular Therapy, University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Sumedha Pareek
- Department of Stem Cell Transplantation and Cellular Therapy, University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Wilfredo Ruiz-Vazquez
- Department of Stem Cell Transplantation and Cellular Therapy, University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Sung-Eun Lee
- Department of Stem Cell Transplantation and Cellular Therapy, University of Texas MD Anderson Cancer Center, Houston, TX, United States.,Department of Hematology, Seoul St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Seoul, South Korea
| | - Jin S Im
- Department of Stem Cell Transplantation and Cellular Therapy, University of Texas MD Anderson Cancer Center, Houston, TX, United States.,Department of Hematology, Seoul St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Seoul, South Korea
| |
Collapse
|
32
|
Miao R, Lim VY, Kothapalli N, Ma Y, Fossati J, Zehentmeier S, Sun R, Pereira JP. Hematopoietic Stem Cell Niches and Signals Controlling Immune Cell Development and Maintenance of Immunological Memory. Front Immunol 2020; 11:600127. [PMID: 33324418 PMCID: PMC7726109 DOI: 10.3389/fimmu.2020.600127] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2020] [Accepted: 10/29/2020] [Indexed: 12/11/2022] Open
Abstract
Studies over the last couple of decades have shown that hematopoietic stem cells (HSCs) are critically dependent on cytokines such as Stem Cell Factor and other signals provided by bone marrow niches comprising of mesenchymal stem and progenitor cells (MSPCs) and endothelial cells (ECs). Because of their critical roles in HSC maintenance the niches formed by MSPCs and ECs are commonly referred to as HSC niches. For the most part, the signals required for HSC maintenance act in a short-range manner, which imposes the necessity for directional and positional cues in order for HSCs to localize and be retained properly in stem cell niches. The chemokine CXCL12 and its Gαi protein coupled receptor CXCR4, besides promoting HSC quiescence directly, also play instrumental roles in enabling HSCs to access bone marrow stem cell niches. Recent studies have revealed, however, that HSC niches also provide a constellation of hematopoietic cytokines that are critical for the production of most, if not all, blood cell types. Some hematopoietic cytokines, namely IL-7 and IL-15 produced by HSC niches, are not only required for lymphopoiesis but are also essential for memory T cell maintenance. Consequently, hematopoietic progenitors and differentiated immune cells, such as memory T cell subsets, also depend on the CXCL12/CXCR4 axis for migration into bone marrow and interactions with MSPCs and ECs. Similarly, subsets of antibody-secreting plasma cells also reside in close association with CXCL12-producing MSPCs in the bone marrow and require the CXCR4/CXCL12 axis for survival and long-term maintenance. Collectively, these studies demonstrate a broad range of key physiological roles, spanning blood cell production and maintenance of immunological memory, that are orchestrated by stem cell niches through a common and simple mechanism: CXCL12/CXCR4-mediated cell recruitment followed by receipt of a maintenance and/or instructive signal. A fundamental flaw of this type of cellular organization is revealed by myeloid and lymphoid leukemias, which target stem cell niches and induce profound transcriptomic changes that result in reduced hematopoietic activity and altered mesenchymal cell differentiation.
Collapse
Affiliation(s)
- Runfeng Miao
- Department of Immunobiology and Yale Stem Cell Center, Yale University School of Medicine, New Haven, CT, United States
| | - Vivian Y Lim
- Department of Immunobiology and Yale Stem Cell Center, Yale University School of Medicine, New Haven, CT, United States
| | - Neeharika Kothapalli
- Department of Immunobiology and Yale Stem Cell Center, Yale University School of Medicine, New Haven, CT, United States
| | - Yifan Ma
- Department of Immunobiology and Yale Stem Cell Center, Yale University School of Medicine, New Haven, CT, United States
| | - Julia Fossati
- Department of Immunobiology and Yale Stem Cell Center, Yale University School of Medicine, New Haven, CT, United States
| | - Sandra Zehentmeier
- Department of Immunobiology and Yale Stem Cell Center, Yale University School of Medicine, New Haven, CT, United States
| | - Ruifeng Sun
- Department of Immunobiology and Yale Stem Cell Center, Yale University School of Medicine, New Haven, CT, United States
| | - João P Pereira
- Department of Immunobiology and Yale Stem Cell Center, Yale University School of Medicine, New Haven, CT, United States
| |
Collapse
|
33
|
García-Ceca J, Montero-Herradón S, Zapata AG. Intrathymic Selection and Defects in the Thymic Epithelial Cell Development. Cells 2020; 9:cells9102226. [PMID: 33023072 PMCID: PMC7601110 DOI: 10.3390/cells9102226] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2020] [Revised: 09/29/2020] [Accepted: 09/30/2020] [Indexed: 02/07/2023] Open
Abstract
Intimate interactions between thymic epithelial cells (TECs) and thymocytes (T) have been repeatedly reported as essential for performing intrathymic T-cell education. Nevertheless, it has been described that animals exhibiting defects in these interactions were capable of a proper positive and negative T-cell selection. In the current review, we first examined distinct types of TECs and their possible role in the immune surveillance. However, EphB-deficient thymi that exhibit profound thymic epithelial (TE) alterations do not exhibit important immunological defects. Eph and their ligands, the ephrins, are implicated in cell attachment/detachment and govern, therefore, TEC–T interactions. On this basis, we hypothesized that a few normal TE areas could be enough for a proper phenotypical and functional maturation of T lymphocytes. Then, we evaluated in vivo how many TECs would be necessary for supporting a normal T-cell differentiation, concluding that a significantly low number of TEC are still capable of supporting normal T lymphocyte maturation, whereas with fewer numbers, T-cell maturation is not possible.
Collapse
Affiliation(s)
- Javier García-Ceca
- Department of Cell Biology, Faculty of Biology, Complutense University of Madrid, 28040 Madrid, Spain; (J.G.-C.); (S.M.-H.)
- Health Research Institute, Hospital 12 de Octubre (imas12), 28041 Madrid, Spain
| | - Sara Montero-Herradón
- Department of Cell Biology, Faculty of Biology, Complutense University of Madrid, 28040 Madrid, Spain; (J.G.-C.); (S.M.-H.)
- Health Research Institute, Hospital 12 de Octubre (imas12), 28041 Madrid, Spain
| | - Agustín G. Zapata
- Department of Cell Biology, Faculty of Biology, Complutense University of Madrid, 28040 Madrid, Spain; (J.G.-C.); (S.M.-H.)
- Health Research Institute, Hospital 12 de Octubre (imas12), 28041 Madrid, Spain
- Correspondence: ; Tel.: +34-91-394-4979
| |
Collapse
|
34
|
Dumont-Lagacé M, Daouda T, Depoërs L, Zumer J, Benslimane Y, Brochu S, Harrington L, Lemieux S, Perreault C. Qualitative Changes in Cortical Thymic Epithelial Cells Drive Postpartum Thymic Regeneration. Front Immunol 2020; 10:3118. [PMID: 32010151 PMCID: PMC6974522 DOI: 10.3389/fimmu.2019.03118] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2019] [Accepted: 12/20/2019] [Indexed: 12/05/2022] Open
Abstract
During gestation, sex hormones cause a significant thymic involution which enhances fertility. This thymic involution is rapidly corrected following parturition. As thymic epithelial cells (TECs) are responsible for the regulation of thymopoiesis, we analyzed the sequential phenotypic and transcriptomic changes in TECs during the postpartum period in order to identify mechanisms triggering postpartum thymic regeneration. In particular, we performed flow cytometry analyses and deep RNA-sequencing on purified TEC subsets at several time points before and after parturition. We report that pregnancy-induced involution is not caused by loss of TECs since their number does not change during or after pregnancy. However, during pregnancy, we observed a significant depletion of all thymocyte subsets downstream of the double-negative 1 (DN1) differentiation stage. Variations in thymocyte numbers correlated with conspicuous changes in the transcriptome of cortical TECs (cTECs). The transcriptomic changes affected predominantly cTEC expression of Foxn1, its targets and several genes that are essential for thymopoiesis. By contrast, medullary TECs (mTECs) showed very little transcriptomic changes in the early postpartum regenerative phase, but seemed to respond to the expansion of single-positive (SP) thymocytes in the late phase of regeneration. Together, these results show that postpartum thymic regeneration is orchestrated by variations in expression of a well-defined subset of cTEC genes, that occur very early after parturition.
Collapse
Affiliation(s)
- Maude Dumont-Lagacé
- Immunobiology Research Unit, Department of Medicine, Institute for Research in Immunology and Cancer, Université de Montréal, Montréal, QC, Canada
| | - Tariq Daouda
- Immunobiology Research Unit, Department of Medicine, Institute for Research in Immunology and Cancer, Université de Montréal, Montréal, QC, Canada.,Functional and Structural Bioinformatics Research Unit, Department of Biochemistry and Molecular Medicine, Institute for Research in Immunology and Cancer, Université de Montréal, Montréal, QC, Canada
| | - Lucyle Depoërs
- Immunobiology Research Unit, Department of Medicine, Institute for Research in Immunology and Cancer, Université de Montréal, Montréal, QC, Canada
| | - Jérémie Zumer
- Functional and Structural Bioinformatics Research Unit, Department of Biochemistry and Molecular Medicine, Institute for Research in Immunology and Cancer, Université de Montréal, Montréal, QC, Canada
| | - Yahya Benslimane
- Telomere Length Homeostasis and Genomic Instability Research Unit, Department of Medicine, Institute for Research in Immunology and Cancer, Université de Montréal, Montréal, QC, Canada
| | - Sylvie Brochu
- Immunobiology Research Unit, Department of Medicine, Institute for Research in Immunology and Cancer, Université de Montréal, Montréal, QC, Canada
| | - Lea Harrington
- Telomere Length Homeostasis and Genomic Instability Research Unit, Department of Medicine, Institute for Research in Immunology and Cancer, Université de Montréal, Montréal, QC, Canada
| | - Sébastien Lemieux
- Functional and Structural Bioinformatics Research Unit, Department of Biochemistry and Molecular Medicine, Institute for Research in Immunology and Cancer, Université de Montréal, Montréal, QC, Canada
| | - Claude Perreault
- Immunobiology Research Unit, Department of Medicine, Institute for Research in Immunology and Cancer, Université de Montréal, Montréal, QC, Canada
| |
Collapse
|
35
|
Abstract
The generation of a functional T cell repertoire in the thymus is mainly orchestrated by thymic epithelial cells (TECs), which provide developing T cells with cues for their navigation, proliferation, differentiation and survival. The TEC compartment has been segregated historically into two major populations of medullary TECs and cortical TECs, which differ in their anatomical localization, molecular characteristics and functional roles. However, recent studies have shown that TECs are highly heterogeneous and comprise multiple subpopulations with distinct molecular and functional characteristics, including tuft cell-like or corneocyte-like phenotypes. Here, we review the most recent advances in our understanding of TEC heterogeneity from a molecular, functional and developmental perspective. In particular, we highlight the key insights that were recently provided by single-cell genomic technologies and in vivo fate mapping and discuss them in the context of previously published data.
Collapse
|
36
|
Shi L, Chen X, Zang A, Li T, Hu Y, Ma S, Lü M, Yin H, Wang H, Zhang X, Zhang B, Leng Q, Yang J, Xiao H. TSC1/mTOR-controlled metabolic-epigenetic cross talk underpins DC control of CD8+ T-cell homeostasis. PLoS Biol 2019; 17:e3000420. [PMID: 31433805 PMCID: PMC6719877 DOI: 10.1371/journal.pbio.3000420] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2019] [Revised: 09/03/2019] [Accepted: 08/01/2019] [Indexed: 02/07/2023] Open
Abstract
Dendritic cells (DCs) play pivotal roles in T-cell homeostasis and activation, and metabolic programing has been recently linked to DC development and function. However, the metabolic underpinnings corresponding to distinct DC functions remain largely unresolved. Here, we demonstrate a special metabolic–epigenetic coupling mechanism orchestrated by tuberous sclerosis complex subunit 1 (TSC1)-mechanistic target of rapamycin (mTOR) for homeostatic DC function. Specific ablation of Tsc1 in the DC compartment (Tsc1DC-KO) largely preserved DC development but led to pronounced reduction in naïve and memory–phenotype cluster of differentiation (CD)8+ T cells, a defect fully rescued by concomitant ablation of mTor or regulatory associated protein of MTOR, complex 1 (Rptor) in DCs. Moreover, Tsc1DC-KO mice were unable to launch efficient antigen-specific CD8+ T effector responses required for containing Listeria monocytogenes and B16 melanomas. Mechanistically, our data suggest that the steady-state DCs tend to tune down de novo fatty acid synthesis and divert acetyl-coenzyme A (acetyl-CoA) for histone acetylation, a process critically controlled by TSC1-mTOR. Correspondingly, TSC1 deficiency elevated acetyl-CoA carboxylase 1 (ACC1) expression and fatty acid synthesis, leading to impaired epigenetic imprinting on selective genes such as major histocompatibility complex (MHC)-I and interleukin (IL)-7. Remarkably, tempering ACC1 activity was able to divert cytosolic acetyl-CoA for histone acetylation and restore the gene expression program compromised by TSC1 deficiency. Taken together, our results uncover a crucial role for TSC1-mTOR in metabolic programing of the homeostatic DCs for T-cell homeostasis and implicate metabolic-coupled epigenetic imprinting as a paradigm for DC specification. Dendritic cells (DCs) play pivotal roles in T cell homeostasis and activation, but the basis of the metabolic programming of distinct DC functions remains unclear. This study identifies a novel metabolic-epigenetic node enabling DC control of CD8 T cell homeostasis, involving mTOR-ACC1 as a rheostat that balances fatty-acid synthesis and histone acetylation.
Collapse
Affiliation(s)
- Lei Shi
- School of Life Sciences, Lanzhou University, Lanzhou, Gansu, China
- CAS Key Laboratory of Molecular Virology and Immunology, Institut Pasteur of Shanghai; CAS Center for Excellence in Molecular Cell Science; University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Xia Chen
- School of Life Sciences, Lanzhou University, Lanzhou, Gansu, China
- CAS Key Laboratory of Molecular Virology and Immunology, Institut Pasteur of Shanghai; CAS Center for Excellence in Molecular Cell Science; University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Aiping Zang
- CAS Key Laboratory of Molecular Virology and Immunology, Institut Pasteur of Shanghai; CAS Center for Excellence in Molecular Cell Science; University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Tiantian Li
- CAS Key Laboratory of Molecular Virology and Immunology, Institut Pasteur of Shanghai; CAS Center for Excellence in Molecular Cell Science; University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Yanxiang Hu
- Department of Immunology, Medical College of Qingdao University, Qingdao, Shandong, China
| | - Shixin Ma
- CAS Key Laboratory of Molecular Virology and Immunology, Institut Pasteur of Shanghai; CAS Center for Excellence in Molecular Cell Science; University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Mengdie Lü
- Affiliated Cancer Hospital and Institute of Guangzhou Medical University, State Key Laboratory of Respiratory Diseases, Guangzhou, Guangdong, China
| | - Huiyong Yin
- CAS Key Laboratory of Nutrition, Metabolism and Food Safety, Shanghai Institute of Nutrition and Health, Chinese Academy of Sciences, Shanghai, China
| | - Haikun Wang
- CAS Key Laboratory of Molecular Virology and Immunology, Institut Pasteur of Shanghai; CAS Center for Excellence in Molecular Cell Science; University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Xiaoming Zhang
- CAS Key Laboratory of Molecular Virology and Immunology, Institut Pasteur of Shanghai; CAS Center for Excellence in Molecular Cell Science; University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Bei Zhang
- Department of Immunology, Medical College of Qingdao University, Qingdao, Shandong, China
| | - Qibin Leng
- Affiliated Cancer Hospital and Institute of Guangzhou Medical University, State Key Laboratory of Respiratory Diseases, Guangzhou, Guangdong, China
- * E-mail: (HX); (JY); (QL)
| | - Jinbo Yang
- School of Life Sciences, Lanzhou University, Lanzhou, Gansu, China
- * E-mail: (HX); (JY); (QL)
| | - Hui Xiao
- CAS Key Laboratory of Molecular Virology and Immunology, Institut Pasteur of Shanghai; CAS Center for Excellence in Molecular Cell Science; University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, China
- * E-mail: (HX); (JY); (QL)
| |
Collapse
|
37
|
Buffière A, Uzan B, Aucagne R, Hermetet F, Mas M, Nassurdine S, Aznague A, Carmignac V, Tournier B, Bouchot O, Ballerini P, Barata JT, Bastie JN, Delva L, Pflumio F, Quéré R. T-cell acute lymphoblastic leukemia displays autocrine production of Interleukin-7. Oncogene 2019; 38:7357-7365. [PMID: 31417180 DOI: 10.1038/s41388-019-0921-4] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2019] [Revised: 06/17/2019] [Accepted: 07/22/2019] [Indexed: 11/09/2022]
Abstract
T-cell acute lymphoblastic leukemia (T-ALL) is an aggressive hematological malignancy characterized by an accumulation of immature T cells. Although patient outcomes have improved, novel targeted therapies are needed to reduce the intensity of chemotherapy and improve the prognosis of high-risk patients. Interleukin-7 (IL-7) modulates the survival and proliferation of normal and malignant T cells. Targeting the IL-7 signaling pathway is thus a potentially effective therapeutic strategy. To achieve such aim, it is essential to first understand how the IL-7 signaling pathway is activated. Although IL-7 production has been observed from multiple stromal tissues, T-ALL autocrine IL-7 secretion has not yet been described. Interestingly, using T-ALL cell lines, primary and patient-derived xenotransplanted (PDX) T-ALL cells, we demonstrate that T-ALL cells produce IL-7 whereas normal T cells do not. Finally, using knock down of IL7 gene in T-ALL cells, we describe to what extent IL-7 autocrine secretion is involved in the T-ALL cells propagation in bone marrow and how it affects the number of leukemia-initiating cells in PDX mice. Together, these results demonstrate how the autocrine production of the IL-7 cytokine mediated by T-ALL cells can be involved in the oncogenic development of T-ALL and offer novel insights into T-ALL spreading.
Collapse
Affiliation(s)
- Anne Buffière
- UMR1231, Inserm/Université Bourgogne Franche-Comté, Dijon, France.,LipSTIC Labex, Dijon, France
| | - Benjamin Uzan
- UMR967, Inserm/CEA/Université Paris 7/Université Paris 11, Fontenay-aux-Roses, France.,LSHL, IRCM/CEA, Fontenay-aux-Roses, France
| | - Romain Aucagne
- UMR1231, Inserm/Université Bourgogne Franche-Comté, Dijon, France.,LipSTIC Labex, Dijon, France
| | - François Hermetet
- UMR1231, Inserm/Université Bourgogne Franche-Comté, Dijon, France.,LipSTIC Labex, Dijon, France
| | - Manon Mas
- UMR1231, Inserm/Université Bourgogne Franche-Comté, Dijon, France
| | | | - Aziza Aznague
- UMR1231, Inserm/Université Bourgogne Franche-Comté, Dijon, France.,LipSTIC Labex, Dijon, France
| | | | - Benjamin Tournier
- Hôpital Universitaire François Mitterrand, Service de Génétique des Cancers, Dijon, France
| | - Olivier Bouchot
- Hôpital Universitaire François Mitterrand, Chirurgie Cardiovasculaire, Dijon, France
| | - Paola Ballerini
- Assistance Publique-Hôpitaux de Paris, Laboratoire d'Hématologie, Hôpital Trousseau, Paris, France
| | - João T Barata
- Instituto de Medicina Molecular, Faculdade de Medicina, Universidade de Lisboa, Lisboa, Portugal
| | - Jean-Noël Bastie
- UMR1231, Inserm/Université Bourgogne Franche-Comté, Dijon, France.,LipSTIC Labex, Dijon, France.,Hôpital Universitaire François Mitterrand, Service d'Hématologie Clinique, Dijon, France.,Hôpital Universitaire François Mitterrand, CRB Ferdinand Cabanne, BB-0033-00044, Dijon, France
| | - Laurent Delva
- UMR1231, Inserm/Université Bourgogne Franche-Comté, Dijon, France.,LipSTIC Labex, Dijon, France
| | - Françoise Pflumio
- UMR967, Inserm/CEA/Université Paris 7/Université Paris 11, Fontenay-aux-Roses, France.,LSHL, IRCM/CEA, Fontenay-aux-Roses, France
| | - Ronan Quéré
- UMR1231, Inserm/Université Bourgogne Franche-Comté, Dijon, France. .,LipSTIC Labex, Dijon, France.
| |
Collapse
|
38
|
Luan R, Liang Z, Zhang Q, Sun L, Zhao Y. Molecular regulatory networks of thymic epithelial cell differentiation. Differentiation 2019; 107:42-49. [PMID: 31238242 DOI: 10.1016/j.diff.2019.06.002] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2018] [Revised: 06/04/2019] [Accepted: 06/12/2019] [Indexed: 01/15/2023]
Abstract
Functional mature T cells are generated in the thymus. Thymic epithelial cells (TECs) provide the essential microenvironment for T cell development and maturation. According to their function and localization, TECs are roughly divided into cortical TECs (cTECs) and medullary TECs (mTECs), which are responsible for positive and negative selection, respectively. This review summarizes the current understanding of TEC biology, the identification of fetal and adult bipotent TEC progenitors, and the signaling pathways that control the development and maturation of TECs. The understanding of the ontogeny, differentiation, maturation and function of cTECs lags behind that of mTECs. Better understanding TEC biology will provide clues about TEC development and the applications of thymus engineering.
Collapse
Affiliation(s)
- Rong Luan
- State Key Laboratory of Membrane Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China
| | - Zhanfeng Liang
- State Key Laboratory of Membrane Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China
| | - Qian Zhang
- State Key Laboratory of Membrane Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China; University of Chinese Academy of Sciences, Beijing, China
| | - Liguang Sun
- Institute of Translational Medicine, The First Hospital, Jilin University, Changchun, Jilin, China.
| | - Yong Zhao
- State Key Laboratory of Membrane Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China; University of Chinese Academy of Sciences, Beijing, China; Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing, China.
| |
Collapse
|
39
|
Zhou L, Che Z, Zhang X, Zhou P, Li X, Xu X, Shi Q, Li D, Ju X. Influence of neonatal gender on cord blood CD34 + cell amplification and gene expression. Exp Ther Med 2019; 18:105-118. [PMID: 31258643 PMCID: PMC6566080 DOI: 10.3892/etm.2019.7549] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2018] [Accepted: 03/12/2019] [Indexed: 12/18/2022] Open
Abstract
The present study attempted to evaluate whether neonatal gender affects the hematopoietic potential of cord blood (CB) transplants and, if so, to determine the underlying molecular mechanisms. CD34+ cells from CB were isolated and divided into male and female groups. CD34+CD38− cell populations were then compared using fluorescence-assisted cell sorting (FACS) and a colony formation assay was performed. Next, a Genechip microarray analysis was used to identify differentially expressed genes (DEGs). Finally, the Genechip results were validated by FACS analysis. It was revealed that the male group had higher amplification efficiency. Gene ontology analysis indicated differences in the biological function of the DEGs between the two groups. Kyoto Encyclopedia of Genes and Genomes analysis suggested that the hematopoietic cell lineage signaling pathway was upregulated in the male group along with high expression levels of genes including interleukin (IL) 6 signal transducer (glycoprotein 130), IL-7 and IL-7 receptor. It was speculated that this may be partially due to numerous upregulated DEGs being involved in chromosomal segregation and hematopoietic cell lineage signaling pathways in CD34+ cells from the male group.
Collapse
Affiliation(s)
- Liping Zhou
- Department of Pediatrics, The Sixth People's Hospital of Ji'nan, Jinan, Shandong 250200, P.R. China.,Cryomedicine Laboratory, Qilu Hospital of Shandong University, Jinan, Shandong 250012, P.R. China
| | - Zhe Che
- Health and Family Planning Inspection Agency of Zhangqiu District, Jinan, Shandong 250200, P.R. China
| | - Xiaowei Zhang
- Department of Pediatrics, The Sixth People's Hospital of Ji'nan, Jinan, Shandong 250200, P.R. China
| | - Panpan Zhou
- Cryomedicine Laboratory, Qilu Hospital of Shandong University, Jinan, Shandong 250012, P.R. China
| | - Xue Li
- Cryomedicine Laboratory, Qilu Hospital of Shandong University, Jinan, Shandong 250012, P.R. China
| | - Xuejing Xu
- Cryomedicine Laboratory, Qilu Hospital of Shandong University, Jinan, Shandong 250012, P.R. China
| | - Qing Shi
- Cryomedicine Laboratory, Qilu Hospital of Shandong University, Jinan, Shandong 250012, P.R. China
| | - Dong Li
- Cryomedicine Laboratory, Qilu Hospital of Shandong University, Jinan, Shandong 250012, P.R. China.,Stem Cell and Regenerative Medicine Research Center, Shandong University, Jinan, Shandong 250012, P.R. China
| | - Xiuli Ju
- Cryomedicine Laboratory, Qilu Hospital of Shandong University, Jinan, Shandong 250012, P.R. China.,Stem Cell and Regenerative Medicine Research Center, Shandong University, Jinan, Shandong 250012, P.R. China
| |
Collapse
|
40
|
Balzano M, De Grandis M, Vu Manh TP, Chasson L, Bardin F, Farina A, Sergé A, Bidaut G, Charbord P, Hérault L, Bailly AL, Cartier-Michaud A, Boned A, Dalod M, Duprez E, Genever P, Coles M, Bajenoff M, Xerri L, Aurrand-Lions M, Schiff C, Mancini SJ. Nidogen-1 Contributes to the Interaction Network Involved in Pro-B Cell Retention in the Peri-sinusoidal Hematopoietic Stem Cell Niche. Cell Rep 2019; 26:3257-3271.e8. [DOI: 10.1016/j.celrep.2019.02.065] [Citation(s) in RCA: 37] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2018] [Revised: 12/24/2018] [Accepted: 02/15/2019] [Indexed: 12/11/2022] Open
|
41
|
Tabata R, Tabata C, Yamamoto N, Shibuya S, Yasumizu R, Kojima M. Progressive peripheral CD8+ T lymphocytosis complicated by pure red cell aplasia following immunosuppressive therapy for thymoma-associated myasthenia gravis. Int Immunopharmacol 2018; 63:14-18. [DOI: 10.1016/j.intimp.2018.07.030] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2018] [Revised: 07/12/2018] [Accepted: 07/25/2018] [Indexed: 10/28/2022]
|
42
|
Murine Bone Marrow Niches from Hematopoietic Stem Cells to B Cells. Int J Mol Sci 2018; 19:ijms19082353. [PMID: 30103411 PMCID: PMC6121419 DOI: 10.3390/ijms19082353] [Citation(s) in RCA: 31] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2018] [Revised: 08/08/2018] [Accepted: 08/08/2018] [Indexed: 01/03/2023] Open
Abstract
After birth, the development of hematopoietic cells occurs in the bone marrow. Hematopoietic differentiation is finely tuned by cell-intrinsic mechanisms and lineage-specific transcription factors. However, it is now clear that the bone marrow microenvironment plays an essential role in the maintenance of hematopoietic stem cells (HSC) and their differentiation into more mature lineages. Mesenchymal and endothelial cells contribute to a protective microenvironment called hematopoietic niches that secrete specific factors and establish a direct contact with developing hematopoietic cells. A number of recent studies have addressed in mouse models the specific molecular events that are involved in the cellular crosstalk between hematopoietic subsets and their niches. This has led to the concept that hematopoietic differentiation and commitment towards a given hematopoietic pathway is a dynamic process controlled at least partially by the bone marrow microenvironment. In this review, we discuss the evolving view of murine hematopoietic–stromal cell crosstalk that is involved in HSC maintenance and commitment towards B cell differentiation.
Collapse
|
43
|
Jouan Y, Patin EC, Hassane M, Si-Tahar M, Baranek T, Paget C. Thymic Program Directing the Functional Development of γδT17 Cells. Front Immunol 2018; 9:981. [PMID: 29867959 PMCID: PMC5951931 DOI: 10.3389/fimmu.2018.00981] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2018] [Accepted: 04/20/2018] [Indexed: 12/27/2022] Open
Abstract
γδT cells comprise a unique T cell sublineage endowed with a wide functional repertoire, which allow them to play important—sometimes opposite—roles in many immune responses associated with infection, cancer, and inflammatory processes. This is largely dependent on the existence of pre-programmed discrete functional subsets that differentiate within the thymus at specific temporal windows of life. Since they represent a major early source of interleukin-17A in many models of immune responses, the γδT17 cell population has recently gained considerable interest. Thus, a better dissection of the developmental program of this effector γδT subset appears critical in understanding their associated immune functions. Several recent reports have provided new exciting insights into the developmental mechanisms that control γδT cell lineage commitment and differentiation. Here, we review the importance of thymic cues and intrinsic factors that shape the developmental program of γδT17 cells. We also discuss the potential future areas of research in γδT17 cell development especially in regards to the recently provided data from deep RNA sequencing technology. Pursuing our understanding into this complex mechanism will undoubtedly provide important clues into the biology of this particular T cell sublineage.
Collapse
Affiliation(s)
- Youenn Jouan
- INSERM, Centre d'Etude des Pathologies Respiratoires (CEPR), UMR 1100, Tours, France.,Université de Tours, Tours, France.,Service de Médecine Intensive Réanimation, Centre Hospitalier Régional Universitaire de Tours, Tours, France
| | - Emmanuel C Patin
- Division of Radiotherapy and Imaging, Targeted Therapy Team, The Institute of Cancer Research, London, United Kingdom
| | - Maya Hassane
- Department of Biochemistry and Molecular Genetics, Faculty of Medicine, American University of Beirut, Beirut, Lebanon
| | - Mustapha Si-Tahar
- INSERM, Centre d'Etude des Pathologies Respiratoires (CEPR), UMR 1100, Tours, France.,Université de Tours, Tours, France
| | - Thomas Baranek
- INSERM, Centre d'Etude des Pathologies Respiratoires (CEPR), UMR 1100, Tours, France.,Université de Tours, Tours, France
| | - Christophe Paget
- INSERM, Centre d'Etude des Pathologies Respiratoires (CEPR), UMR 1100, Tours, France.,Université de Tours, Tours, France
| |
Collapse
|
44
|
Hosokawa H, Rothenberg EV. Cytokines, Transcription Factors, and the Initiation of T-Cell Development. Cold Spring Harb Perspect Biol 2018; 10:cshperspect.a028621. [PMID: 28716889 DOI: 10.1101/cshperspect.a028621] [Citation(s) in RCA: 99] [Impact Index Per Article: 14.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Multipotent blood progenitor cells migrate into the thymus and initiate the T-cell differentiation program. T-cell progenitor cells gradually acquire T-cell characteristics while shedding their multipotentiality for alternative fates. This process is supported by extracellular signaling molecules, including Notch ligands and cytokines, provided by the thymic microenvironment. T-cell development is associated with dynamic change of gene regulatory networks of transcription factors, which interact with these environmental signals. Together with Notch or pre-T-cell-receptor (TCR) signaling, cytokines always control proliferation, survival, and differentiation of early T cells, but little is known regarding their cross talk with transcription factors. However, recent results suggest ways that cytokines expressed in distinct intrathymic niches can specifically modulate key transcription factors. This review discusses how stage-specific roles of cytokines and transcription factors can jointly guide development of early T cells.
Collapse
Affiliation(s)
- Hiroyuki Hosokawa
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, California 91125
| | - Ellen V Rothenberg
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, California 91125
| |
Collapse
|
45
|
Raeber ME, Zurbuchen Y, Impellizzieri D, Boyman O. The role of cytokines in T-cell memory in health and disease. Immunol Rev 2018; 283:176-193. [DOI: 10.1111/imr.12644] [Citation(s) in RCA: 99] [Impact Index Per Article: 14.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Affiliation(s)
- Miro E. Raeber
- Department of Immunology; University Hospital Zurich; Zurich Switzerland
| | - Yves Zurbuchen
- Department of Immunology; University Hospital Zurich; Zurich Switzerland
| | | | - Onur Boyman
- Department of Immunology; University Hospital Zurich; Zurich Switzerland
- Faculty of Medicine; University of Zurich; Zurich Switzerland
| |
Collapse
|
46
|
Oyler-Yaniv A, Krichevsky O. Imaging Cytokine Concentration Fields Using PlaneView Imaging Devices. Bio Protoc 2018; 8:e2788. [PMID: 34286011 DOI: 10.21769/bioprotoc.2788] [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: 12/17/2017] [Revised: 03/15/2018] [Accepted: 03/22/2018] [Indexed: 11/02/2022] Open
Abstract
We describe here a method to visualize concentration fields of cytokines around cytokine-secreting cells. The main challenge is that physiological cytokine concentrations can be very low, in the pico-molar range. Since it is currently impossible to measure such concentrations directly, we rely on cell's response to the cytokines-the phosphorylation of a transcription factor-that can be visualized through antibody staining. Our devices aim at mimicking conditions in dense tissues, such as lymph nodes. A small number of secreting cells is deposited on a polylysine-coated glass and covered by multiple layers of cytokine-consuming. The cells are left to communicate for 1 h, after which the top layers are removed and the bottom layer of cells is antibody labeled for the response to cytokines. Then a cross-section of cytokine fields can be visualized by standard fluorescence microscopy. This manuscript summarized our method to quantify the extent of cytokine-mediated cell-to-cell communications in dense collection of cells in vitro.
Collapse
Affiliation(s)
- Alon Oyler-Yaniv
- Physics Department, Ben-Gurion University of the Negev, Beer-Sheva, Israel
| | - Oleg Krichevsky
- Physics Department, Ben-Gurion University of the Negev, Beer-Sheva, Israel.,Ilse Kats Center for Nanoscience, Ben-Gurion University of the Negev, Beer-Sheva, Israel
| |
Collapse
|
47
|
Xiao S, Zhang W, Manley NR. Thymic B cell development is controlled by the B potential of progenitors via both hematopoietic-intrinsic and thymic microenvironment-intrinsic regulatory mechanisms. PLoS One 2018; 13:e0193189. [PMID: 29462202 PMCID: PMC5819817 DOI: 10.1371/journal.pone.0193189] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2017] [Accepted: 02/06/2018] [Indexed: 12/23/2022] Open
Abstract
BACKGROUND Hematopoietic stem cells (HSCs) derived from birth through adult possess differing differentiation potential for T or B cell fate in the thymus; neonatal bone marrow (BM) cells also have a higher potential for B cell production in BM compared to adult HSCs. We hypothesized that this hematopoietic-intrinsic B potential might also regulate B cell development in the thymus during ontogeny. METHODS Foxn1lacZ mutant mice are a model in which down regulation of a thymic epithelial cell (TEC) specific transcription factor beginning one week postnatal causes a dramatic reduction of thymocytes production. In this study, we found that while T cells were decreased, the frequency of thymic B cells was greatly increased in these mutants in the perinatal period. We used this model to characterize the mechanisms in the thymus controlling B cell development. RESULTS Foxn1lacZ mutants, T cell committed intrathymic progenitors (DN1a,b) were progressively reduced beginning one week after birth, while thymic B cells peaked at 3-4 weeks with pre-B-II progenitor phenotype, and originated in the thymus. Heterochronic chimeras showed that the capacity for thymic B cell production was due to a combination of higher B potential of neonatal HSCs, combined with a thymic microenvironment deficiency including reduction of DL4 and increase of IL-7 that promoted B cell fate. CONCLUSION Our findings indicate that the capacity and time course for thymic B-cell production are primarily controlled by the hematopoietic-intrinsic potential for B cells themselves during ontogeny, but that signals from TECs microenvironment also influence the frequency and differentiation potential of B cell development in the thymus.
Collapse
Affiliation(s)
- Shiyun Xiao
- Department of Genetics, Paul D. Coverdell Center, University of Georgia, Athens, Georgia, United States of America
| | - Wen Zhang
- Department of Genetics, Paul D. Coverdell Center, University of Georgia, Athens, Georgia, United States of America
| | - Nancy R. Manley
- Department of Genetics, Paul D. Coverdell Center, University of Georgia, Athens, Georgia, United States of America
| |
Collapse
|
48
|
Rodrigues PM, Ribeiro AR, Serafini N, Meireles C, Di Santo JP, Alves NL. Intrathymic Deletion of IL-7 Reveals a Contribution of the Bone Marrow to Thymic Rebound Induced by Androgen Blockade. THE JOURNAL OF IMMUNOLOGY 2018; 200:1389-1398. [DOI: 10.4049/jimmunol.1701112] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/30/2023]
Abstract
Abstract
Despite the well-documented effect of castration in thymic regeneration, the singular contribution of the bone marrow (BM) versus the thymus to this process remains unclear. The chief role of IL-7 in pre- and intrathymic stages of T lymphopoiesis led us to investigate the impact of disrupting this cytokine during thymic rebound induced by androgen blockade. We found that castration promoted thymopoiesis in young and aged wild-type mice. In contrast, only young germline IL-7–deficient (Il7−/−) mice consistently augmented thymopoiesis after castration. The increase in T cell production was accompanied by the expansion of the sparse medullary thymic epithelial cell and the peripheral T cell compartment in young Il7−/− mice. In contrast to young Il7−/− and wild-type mice, the poor thymic response of aged Il7−/− mice after castration was associated with a defect in the expansion of BM hematopoietic progenitors. These findings suggest that BM-derived T cell precursors contribute to thymic rebound driven by androgen blockade. To assess the role of IL-7 within the thymus, we generated mice with conditional deletion of IL-7 (Il7 conditional knockout [cKO]) in thymic epithelial cells. As expected, Il7cKO mice presented a profound defect in T cell development while maintaining an intact BM hematopoietic compartment across life. Unlike Il7−/− mice, castration promoted the expansion of BM precursors and enhanced thymic activity in Il7cKO mice independently of age. Our findings suggest that the mobilization of BM precursors acts as a prime catalyst of castration-driven thymopoiesis.
Collapse
Affiliation(s)
- Pedro M. Rodrigues
- *Instituto de Investigação e Inovação em Saúde, Universidade do Porto, 4200-135 Porto, Portugal
- †Thymus Development and Function Laboratory, Institute for Molecular and Cellular Biology, 4200-135 Porto, Portugal
- ‡Doctoral Program in Biomedical Sciences, Abel Salazar Biomedical Sciences Institute, University of Porto, 4050-313 Porto, Portugal
| | - Ana R. Ribeiro
- *Instituto de Investigação e Inovação em Saúde, Universidade do Porto, 4200-135 Porto, Portugal
- †Thymus Development and Function Laboratory, Institute for Molecular and Cellular Biology, 4200-135 Porto, Portugal
| | - Nicolas Serafini
- §Innate Immunity Unit, Pasteur Institute, 75724 Paris, France; and
- ¶INSERM U1223, 75015 Paris, France
| | - Catarina Meireles
- *Instituto de Investigação e Inovação em Saúde, Universidade do Porto, 4200-135 Porto, Portugal
- †Thymus Development and Function Laboratory, Institute for Molecular and Cellular Biology, 4200-135 Porto, Portugal
| | - James P. Di Santo
- §Innate Immunity Unit, Pasteur Institute, 75724 Paris, France; and
- ¶INSERM U1223, 75015 Paris, France
| | - Nuno L. Alves
- *Instituto de Investigação e Inovação em Saúde, Universidade do Porto, 4200-135 Porto, Portugal
- †Thymus Development and Function Laboratory, Institute for Molecular and Cellular Biology, 4200-135 Porto, Portugal
| |
Collapse
|
49
|
Peng Y. Forced expression of IL-7R promotes CD8 T cell cytotoxicity to self antigen. PLoS One 2017; 12:e0188112. [PMID: 29272267 PMCID: PMC5741212 DOI: 10.1371/journal.pone.0188112] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2016] [Accepted: 11/01/2017] [Indexed: 01/22/2023] Open
Abstract
Cross-presentation of apoptotic cell associated antigens by immature dendritic cells prevents the activation of self reactive CD8 T cells. Tolerized self reactive CD8 T cells down-regulate IL-7R expression on their surface. Whether over-expression of IL-7R can reverse their fate and function has not been examined. In this paper, we showed forced expression of IL-7R in OT-I T cells by a transgene enhanced CD8 T cell mediated diabetes in the RIP-mOVA model. Although IL-7R Tg (transgenic) did not completely reverse the deletion of OT-I T cells, it provided a significant survival advantage over w.t OT-I T cells. Furthermore, IL7R Tg OT-I T cells isolated from diabetic pancreata displayed increased production of IFN-γ, higher expression of T-bet, and increased externalization of CD107a. We also found that immature DCs containing apoptotic cells expressed high levels of PDL-1 on their surface. Although IL-7R Tg did not change PD1 expression on activated OT-I cells in vivo, the transgene enabled a significantly lower number of OT-I T cells to induce diabetes in the absence of PDL-1. Our results demonstrated that forced expression of IL-7R not only improved the functionality of tolerized CD8 T cells, it also acted in synergy with PDL-1 deficiency to further promote CD8 T cell cytotoxicity to self antigens.
Collapse
Affiliation(s)
- YuFeng Peng
- Division of Rheumatology, Department of Medicine, University of Washington, Seattle, Washington, United States of America
- * E-mail:
| |
Collapse
|
50
|
Interleukin-7 Availability Is Maintained by a Hematopoietic Cytokine Sink Comprising Innate Lymphoid Cells and T Cells. Immunity 2017; 47:171-182.e4. [DOI: 10.1016/j.immuni.2017.07.005] [Citation(s) in RCA: 81] [Impact Index Per Article: 10.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2017] [Revised: 05/05/2017] [Accepted: 07/05/2017] [Indexed: 01/09/2023]
|