1
|
Seyedhassantehrani N, Burns CS, Verrinder R, Okafor V, Abbasizadeh N, Spencer JA. Intravital two-photon microscopy of the native mouse thymus. PLoS One 2024; 19:e0307962. [PMID: 39088574 PMCID: PMC11293686 DOI: 10.1371/journal.pone.0307962] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2024] [Accepted: 07/15/2024] [Indexed: 08/03/2024] Open
Abstract
The thymus, a key organ in the adaptive immune system, is sensitive to a variety of insults including cytotoxic preconditioning, which leads to atrophy, compression of the blood vascular system, and alterations in hemodynamics. Although the thymus has innate regenerative capabilities, the production of T cells relies on the trafficking of lymphoid progenitors from the bone marrow through the altered thymic blood vascular system. Our understanding of thymic blood vascular hemodynamics is limited due to technical challenges associated with accessing the native thymus in live mice. To overcome this challenge, we developed an intravital two-photon imaging method to visualize the native thymus in vivo and investigated functional changes to the vascular system following sublethal irradiation. We quantified blood flow velocity and shear rate in cortical blood vessels and identified a subtle but significant increase in vessel leakage and diameter ~24 hrs post-sublethal irradiation. Ex vivo whole organ imaging of optically cleared thymus lobes confirmed a disruption of the thymus vascular structure, resulting in an increase in blood vessel diameter and vessel area, and concurrent thymic atrophy. This novel two-photon intravital imaging method enables a new paradigm for directly investigating the thymic microenvironment in vivo.
Collapse
Affiliation(s)
- Negar Seyedhassantehrani
- Quantitative and Systems Biology Graduate Program, University of California Merced, Merced, California, United States of America
- NSF-CREST Center for Cellular and Biomolecular Machines, University of California Merced, Merced, California, United States of America
| | - Christian S. Burns
- Quantitative and Systems Biology Graduate Program, University of California Merced, Merced, California, United States of America
- NSF-CREST Center for Cellular and Biomolecular Machines, University of California Merced, Merced, California, United States of America
| | - Ruth Verrinder
- NSF-CREST Center for Cellular and Biomolecular Machines, University of California Merced, Merced, California, United States of America
- Department of Bioengineering, University of California Merced, Merced, California, United States of America
| | - Victoria Okafor
- NSF-CREST Center for Cellular and Biomolecular Machines, University of California Merced, Merced, California, United States of America
- Department of Bioengineering, University of California Merced, Merced, California, United States of America
| | - Nastaran Abbasizadeh
- Quantitative and Systems Biology Graduate Program, University of California Merced, Merced, California, United States of America
- NSF-CREST Center for Cellular and Biomolecular Machines, University of California Merced, Merced, California, United States of America
| | - Joel A. Spencer
- Quantitative and Systems Biology Graduate Program, University of California Merced, Merced, California, United States of America
- NSF-CREST Center for Cellular and Biomolecular Machines, University of California Merced, Merced, California, United States of America
- Department of Bioengineering, University of California Merced, Merced, California, United States of America
- Health Science Research Institute, University of California Merced, Merced, California, United States of America
| |
Collapse
|
2
|
Fukuhara T, Ueda Y, Lee SI, Odaka T, Nakajima S, Fujisawa JI, Okuma K, Naganuma M, Okazaki K, Kondo N, Kamioka Y, Matsumoto M, Kinashi T. Thymocyte Development of Humanized Mice Is Promoted by Interactions with Human-Derived Antigen Presenting Cells upon Immunization. Int J Mol Sci 2023; 24:11705. [PMID: 37511462 PMCID: PMC10380196 DOI: 10.3390/ijms241411705] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2023] [Revised: 07/07/2023] [Accepted: 07/15/2023] [Indexed: 07/30/2023] Open
Abstract
Immune responses in humanized mice are generally inefficient without co-transplantation of human thymus or HLA transgenes. Previously, we generated humanized mice via the intra-bone marrow injection of CD133+ cord blood cells into irradiated adult immunodeficient mice (IBMI-huNSG mice), which could mount functional immune responses against HTLV-1, although the underlying mechanisms were still unknown. Here, we investigated thymocyte development in IBMI-huNSG mice, focusing on the roles of human and mouse MHC restriction. IBMI-huNSG mice had normal developmental profiles but aberrant thymic structures. Surprisingly, the thymic medulla-like regions expanded after immunization due to enhanced thymocyte expansion in association with the increase in HLA-DR+ cells, including CD205+ dendritic cells (DCs). The organ culture of thymus from immunized IBMI-huNSG mice with a neutralizing antibody to HLA-DR showed the HLA-DR-dependent expansion of CD4 single positive thymocytes. Mature peripheral T-cells exhibited alloreactive proliferation when co-cultured with human peripheral blood mononuclear cells. Live imaging of the thymus from immunized IBMI-huNSG mice revealed dynamic adhesive contacts of human-derived thymocytes and DCs accompanied by Rap1 activation. These findings demonstrate that an increase in HLA-DR+ cells by immunization promotes HLA-restricted thymocyte expansion in humanized mice, offering a unique opportunity to generate humanized mice with ease.
Collapse
Affiliation(s)
- Takataro Fukuhara
- Division of Gastroenterology and Hepatology, The Third Department of Internal Medicine, Kansai Medical University, Hirakata 573-1010, Osaka, Japan
- Department of Molecular Genetics, Institute of Biomedical Science, Kansai Medical University, Hirakata 573-1010, Osaka, Japan
| | - Yoshihiro Ueda
- Department of Molecular Genetics, Institute of Biomedical Science, Kansai Medical University, Hirakata 573-1010, Osaka, Japan
| | - Sung-Il Lee
- Department of Model Animal, Institute of Biomedical Science, Kansai Medical University, Hirakata 573-1010, Osaka, Japan
| | - Tokifumi Odaka
- Department of Microbiology, Kansai Medical University, Hirakata 573-1010, Osaka, Japan
| | - Shinsuke Nakajima
- Department of Microbiology, Kansai Medical University, Hirakata 573-1010, Osaka, Japan
| | - Jun-Ichi Fujisawa
- Department of Microbiology, Kansai Medical University, Hirakata 573-1010, Osaka, Japan
| | - Kazu Okuma
- Department of Microbiology, Kansai Medical University, Hirakata 573-1010, Osaka, Japan
| | - Makoto Naganuma
- Division of Gastroenterology and Hepatology, The Third Department of Internal Medicine, Kansai Medical University, Hirakata 573-1010, Osaka, Japan
| | - Kazuichi Okazaki
- Division of Gastroenterology and Hepatology, The Third Department of Internal Medicine, Kansai Medical University, Hirakata 573-1010, Osaka, Japan
| | - Naoyuki Kondo
- Department of Molecular Genetics, Institute of Biomedical Science, Kansai Medical University, Hirakata 573-1010, Osaka, Japan
| | - Yuji Kamioka
- Department of Molecular Genetics, Institute of Biomedical Science, Kansai Medical University, Hirakata 573-1010, Osaka, Japan
| | - Mitsuru Matsumoto
- Division of Molecular Immunology, Institute for Enzyme Research, Tokushima University, Kuramoto 770-8503, Tokushima, Japan
| | - Tatsuo Kinashi
- Department of Molecular Genetics, Institute of Biomedical Science, Kansai Medical University, Hirakata 573-1010, Osaka, Japan
| |
Collapse
|
3
|
Orlova E, Loginova O, Shirshev S. Leptin regulates thymic plasmacytoid dendritic cell ability to influence the thymocyte distribution in vitro. Int Immunopharmacol 2023; 117:109912. [PMID: 36857934 DOI: 10.1016/j.intimp.2023.109912] [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: 07/05/2022] [Revised: 02/07/2023] [Accepted: 02/14/2023] [Indexed: 03/03/2023]
Abstract
Leptin, the adipocyte-derived hormone, involved in regulating food intake and body weight, plays an important role in immunity and reproduction. Leptin signals via the specific membrane receptors expressed in most types of immune cells including dendritic cells (DCs) and thymocytes. Leptin enhances thymopoiesis and modulates T-cell-mediated immunity. Thymic plasmacytoid DCs (pDCs) are predominated in the thymus. They play an important role in thymocyte differentiation. We have analyzed whether leptin mediates its effects on human thymocytes by influencing on pDCs. We used leptin at concentration corresponding to its level during II-III trimesters of physiological pregnancy. We cultivated leptin-primed pDCs with autologous thymocytes and estimated the main thymocyte subsets expressing αβ chains of the T-cell receptor (αβTCR), natural regulatory T-cells (tTreg), natural T-helpers producing interleukin-17 (nTh17) and invariant natural killer T-cells (iNKT) in vitro. We have shown that leptin augmented CD86, CD276 expressions and depressed IL-10 productions by pDCs. Leptin-primed pDCs decreased the percentage of CD4+CD8+αβTCR+ thymocytes, increased CD4hiCD8-/loαβTCR+ cells. pDCs cultivated with leptin decreased the number of iNKT precursors, and did not change the number of tTreg and nTh17 precursors. Thus, leptin's important role in regulation of thymic pDC abilities to influence on the thymocyte distribution was indicated in vitro.
Collapse
Affiliation(s)
- Ekaterina Orlova
- Laboratory of Immunoregulation, Perm Federal Research Center, Ural Branch of the Russian Academy of Sciences, 13A Lenina str., 614015 Perm, Russia.
| | - Olga Loginova
- Laboratory of Immunoregulation, Perm Federal Research Center, Ural Branch of the Russian Academy of Sciences, 13A Lenina str., 614015 Perm, Russia.
| | - Sergei Shirshev
- Laboratory of Immunoregulation, Perm Federal Research Center, Ural Branch of the Russian Academy of Sciences, 13A Lenina str., 614015 Perm, Russia.
| |
Collapse
|
4
|
McIntyre LL, Lutes LK, Robey EA. Studying T Cell Development in Neonatal and Adult Thymic Slices. Methods Mol Biol 2023; 2580:233-247. [PMID: 36374461 DOI: 10.1007/978-1-0716-2740-2_14] [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: 06/16/2023]
Abstract
T cell development occurs in the thymus and is coordinated temporally and spatially within the highly complex thymic microenvironment. Therefore, T cell selection and maturation events cannot be fully recapitulated using traditional two-dimensional tissue culture in vitro. The thymic slice system provides a highly versatile system for studying T cell development ex vivo while preserving three-dimensional thymic architecture. Using the thymic slice system, T cell selection and maturation events can be visualized by live imaging and quantified by flow cytometry. Here we describe the process for generating slices from neonatal and adult mice.
Collapse
Affiliation(s)
- Laura L McIntyre
- Department of Molecular and Cell Biology, University of California, Berkeley, CA, USA
| | - Lydia K Lutes
- Department of Molecular and Cell Biology, University of California, Berkeley, CA, USA
| | - Ellen A Robey
- Department of Molecular and Cell Biology, University of California, Berkeley, CA, USA.
| |
Collapse
|
5
|
Hou L, Yuki K. CD11c regulates late-stage T cell development in the thymus. Front Immunol 2022; 13:1040818. [PMID: 36439108 PMCID: PMC9684328 DOI: 10.3389/fimmu.2022.1040818] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2022] [Accepted: 10/26/2022] [Indexed: 10/03/2023] Open
Abstract
CD11c, also named integrin αX, has been deemed solely as a dendritic cell marker for decades while the delineation of its biological function was limited. In the current study, we observed in mice that CD11c deficiency led to a defect in T cell development, demonstrated by the loss of CD4+CD8+ double positive (DP) T cells, CD4+CD8-, and CD4-CD8+ single positive (SP) T cells in the thymus and less mature T cells in the periphery. By using bone marrow chimera, we confirmed that CD11c regulated T cell development in the thymus. We further showed that CD11c deficiency led to an accelerated apoptosis of CD3 positive thymocytes, but not CD4-CD8- double negative (DN) T cells. Overall, this study added one more layer of knowledge on the regulatory mechanism of late-stage T cell development that the presence of CD11c in the thymus is critical for maintaining T cell survival.
Collapse
Affiliation(s)
- Lifei Hou
- Department of Anesthesiology, Critical Care and Pain Medicine, Cardiac Anesthesia Division, Boston Children’s Hospital, Boston, MA, United States
- Departments of Anaesthesia and Immunology, Harvard Medical School, Boston, MA, United States
| | - Koichi Yuki
- Department of Anesthesiology, Critical Care and Pain Medicine, Cardiac Anesthesia Division, Boston Children’s Hospital, Boston, MA, United States
- Departments of Anaesthesia and Immunology, Harvard Medical School, Boston, MA, United States
| |
Collapse
|
6
|
Wang C, Daley SR. How Thymocyte Deletion in the Cortex May Curtail Antigen-Specific T-Regulatory Cell Development in the Medulla. Front Immunol 2022; 13:892498. [PMID: 35693793 PMCID: PMC9176388 DOI: 10.3389/fimmu.2022.892498] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2022] [Accepted: 04/26/2022] [Indexed: 11/13/2022] Open
Abstract
CD4+ T cell responses to self-antigens are pivotal for immunological self-tolerance. Activation of Foxp3– T-conventional (T-conv) cells can precipitate autoimmune disease, whereas activation of Foxp3+ T-regulatory (T-reg) cells is essential to prevent autoimmune disease. This distinction indicates the importance of the thymus in controlling the differentiation of self-reactive CD4+ T cells. Thymocytes and thymic antigen-presenting cells (APC) depend on each other for normal maturation and differentiation. In this Hypothesis and Theory article, we propose this mutual dependence dictates which self-antigens induce T-reg cell development in the thymic medulla. We postulate self-reactive CD4+ CD8– thymocytes deliver signals that stabilize and amplify the presentation of their cognate self-antigen by APC in the thymic medulla, thereby seeding a niche for the development of T-reg cells specific for the same self-antigen. By limiting the number of antigen-specific CD4+ thymocytes in the medulla, thymocyte deletion in the cortex may impede the formation of medullary T-reg niches containing certain self-antigens. Susceptibility to autoimmune disease may arise from cortical deletion creating a “hole” in the self-antigen repertoire recognized by T-reg cells.
Collapse
Affiliation(s)
- Chenglong Wang
- Centre for Immunology and Infection Control, School of Biomedical Sciences, Faculty of Health, Queensland University of Technology, Brisbane, QLD, Australia
| | - Stephen R Daley
- Centre for Immunology and Infection Control, School of Biomedical Sciences, Faculty of Health, Queensland University of Technology, Brisbane, QLD, Australia
| |
Collapse
|
7
|
Balcells F, Martínez Monteros MJ, Gómez AL, Cazorla SI, Perdigón G, Maldonado-Galdeano C. Probiotic Consumption Boosts Thymus in Obesity and Senescence Mouse Models. Nutrients 2022; 14:nu14030616. [PMID: 35276973 PMCID: PMC8838891 DOI: 10.3390/nu14030616] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2021] [Revised: 01/21/2022] [Accepted: 01/23/2022] [Indexed: 02/01/2023] Open
Abstract
The ability of the immune system to respond to different pathogens throughout life requires the constant production and selection of T cells in the thymus. This immune organ is very sensitive to age, infectious processes and nutrition disorders (obesity and malnutrition). Several studies have shown that the incorporation of some probiotic bacteria or probiotic fermented milk in the diet has beneficial effects, not only at the intestinal level but also on distant mucosal tissues, improving the architecture of the thymus in a malnutrition model. The aim of the present study was to determine whether supplementation with the probiotic strain Lactobacillus casei CRL 431 and/or its cell wall could improve body weight, intestinal microbiota and thymus structure and function in both obese and aging mice. We evaluated probiotic administration to BALB/c mice in 2 experimental mouse models: obesity and senescence, including mice of different ages (21, 28, 45, 90 and 180 days). Changes in thymus size and histology were recorded. T-lymphocyte population and cytokine production were also determined. The consumption of probiotics improved the cortical/medullary ratio, the production and regulation of cytokines and the recovery of mature T-lymphocyte populations of the thymus in obese and old mice. Probiotic incorporation into the diet could not only modulate the immune system but also lead to thymus function recovery, thus improving quality of life.
Collapse
Affiliation(s)
- Florencia Balcells
- Laboratorio de Inmunología, Centro de Referencia para Lactobacilos (CERELA-CONICET), San Miguel de Tucumán 4000, Argentina; (F.B.); (M.J.M.M.); (S.I.C.); (G.P.)
| | - María José Martínez Monteros
- Laboratorio de Inmunología, Centro de Referencia para Lactobacilos (CERELA-CONICET), San Miguel de Tucumán 4000, Argentina; (F.B.); (M.J.M.M.); (S.I.C.); (G.P.)
| | - Alba Lorena Gómez
- PatLab Laboratorio de Anatomía Patológica Citopatología e Inmunohistoquímica, San Miguel de Tucumán 4000, Argentina;
| | - Silvia Inés Cazorla
- Laboratorio de Inmunología, Centro de Referencia para Lactobacilos (CERELA-CONICET), San Miguel de Tucumán 4000, Argentina; (F.B.); (M.J.M.M.); (S.I.C.); (G.P.)
- Cátedra de Inmunología, Facultad de Bioquímica, Química y Farmacia, Universidad Nacional de Tucumán, San Miguel de Tucumán 4000, Argentina
| | - Gabriela Perdigón
- Laboratorio de Inmunología, Centro de Referencia para Lactobacilos (CERELA-CONICET), San Miguel de Tucumán 4000, Argentina; (F.B.); (M.J.M.M.); (S.I.C.); (G.P.)
| | - Carolina Maldonado-Galdeano
- Laboratorio de Inmunología, Centro de Referencia para Lactobacilos (CERELA-CONICET), San Miguel de Tucumán 4000, Argentina; (F.B.); (M.J.M.M.); (S.I.C.); (G.P.)
- Cátedra de Inmunología, Facultad de Bioquímica, Química y Farmacia, Universidad Nacional de Tucumán, San Miguel de Tucumán 4000, Argentina
- Correspondence:
| |
Collapse
|
8
|
Dong M, Chang J, Lebel MÈ, Gervais N, Fournier M, Mallet Gauthier È, Suh WK, Melichar HJ. The ICOS-ICOSL pathway tunes thymic selection. Immunol Cell Biol 2021; 100:205-217. [PMID: 34962663 PMCID: PMC9304562 DOI: 10.1111/imcb.12520] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2021] [Revised: 12/11/2021] [Accepted: 12/27/2021] [Indexed: 11/30/2022]
Abstract
Negative selection of developing T cells plays a significant role in T cell tolerance to self-antigen. This process relies on thymic antigen presenting cells which express both self-antigens as well as co-signaling molecules. Inducible T cell costimulator (ICOS) belongs to the CD28 family of co-signaling molecules and binds to ICOS ligand (ICOSL). The ICOS signaling pathway plays important roles in shaping the immune response to infections, but its role in central tolerance is less well understood. Here we show that ICOSL is expressed by subsets of thymic dendritic cells and medullary thymic epithelial cells as well as thymic B cells. ICOS expression is upregulated as T cells mature in the thymus and correlates with T cell receptor signal strength during thymic selection. We also provide evidence of a role for ICOS signaling in mediating negative selection. Our findings suggest that ICOS may fine-tune T cell receptor signals during thymic selection contributing to the generation of a tolerant T cell population.
Collapse
Affiliation(s)
- Mengqi Dong
- Département de microbiologie, Université de Montréal, infectiologie et immunologie, Montréal, Québec, H3T 1J4, Canada.,Immunology-Oncology Unit, Maisonneuve-Rosemont Hospital Research Center, Montréal, Québec, H1T 2M4, Canada
| | - Jinsam Chang
- Institut de recherches cliniques de Montréal, Montréal, Québec, H2W 1R7, Canada.,Programme de biologie moléculaire, Université de Montréal, Montréal, Québec, H3T 1J4, Canada
| | - Marie-Ève Lebel
- Immunology-Oncology Unit, Maisonneuve-Rosemont Hospital Research Center, Montréal, Québec, H1T 2M4, Canada
| | - Noémie Gervais
- Immunology-Oncology Unit, Maisonneuve-Rosemont Hospital Research Center, Montréal, Québec, H1T 2M4, Canada
| | - Marilaine Fournier
- Immunology-Oncology Unit, Maisonneuve-Rosemont Hospital Research Center, Montréal, Québec, H1T 2M4, Canada
| | - Ève Mallet Gauthier
- Département de microbiologie, Université de Montréal, infectiologie et immunologie, Montréal, Québec, H3T 1J4, Canada.,Immunology-Oncology Unit, Maisonneuve-Rosemont Hospital Research Center, Montréal, Québec, H1T 2M4, Canada
| | - Woong-Kyung Suh
- Département de microbiologie, Université de Montréal, infectiologie et immunologie, Montréal, Québec, H3T 1J4, Canada.,Institut de recherches cliniques de Montréal, Montréal, Québec, H2W 1R7, Canada.,Programme de biologie moléculaire, Université de Montréal, Montréal, Québec, H3T 1J4, Canada
| | - Heather J Melichar
- Immunology-Oncology Unit, Maisonneuve-Rosemont Hospital Research Center, Montréal, Québec, H1T 2M4, Canada.,Département de médecine, Université de Montréal, Montréal, Québec, H3T 1J4, Canada
| |
Collapse
|
9
|
Srinivasan J, Lancaster JN, Singarapu N, Hale LP, Ehrlich LIR, Richie ER. Age-Related Changes in Thymic Central Tolerance. Front Immunol 2021; 12:676236. [PMID: 33968086 PMCID: PMC8100025 DOI: 10.3389/fimmu.2021.676236] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2021] [Accepted: 04/06/2021] [Indexed: 01/03/2023] Open
Abstract
Thymic epithelial cells (TECs) and hematopoietic antigen presenting cells (HAPCs) in the thymus microenvironment provide essential signals to self-reactive thymocytes that induce either negative selection or generation of regulatory T cells (Treg), both of which are required to establish and maintain central tolerance throughout life. HAPCs and TECs are comprised of multiple subsets that play distinct and overlapping roles in central tolerance. Changes that occur in the composition and function of TEC and HAPC subsets across the lifespan have potential consequences for central tolerance. In keeping with this possibility, there are age-associated changes in the cellular composition and function of T cells and Treg. This review summarizes changes in T cell and Treg function during the perinatal to adult transition and in the course of normal aging, and relates these changes to age-associated alterations in thymic HAPC and TEC subsets.
Collapse
Affiliation(s)
- Jayashree Srinivasan
- Department of Molecular Biosciences, Institute of Cellular and Molecular Biology, The University of Texas at Austin, Austin, TX, United States
| | | | - Nandini Singarapu
- Department of Epigenetics and Molecular Carcinogenesis, The University of Texas M.D. Anderson Cancer Center, Smithville, TX, United States
| | - Laura P Hale
- Department of Pathology, Duke University School of Medicine, Durham, NC, United States
| | - Lauren I R Ehrlich
- Department of Molecular Biosciences, Institute of Cellular and Molecular Biology, The University of Texas at Austin, Austin, TX, United States.,Livestrong Cancer Institutes, Dell Medical School, The University of Texas at Austin, Austin, TX, United States
| | - Ellen R Richie
- Department of Epigenetics and Molecular Carcinogenesis, The University of Texas M.D. Anderson Cancer Center, Smithville, TX, United States
| |
Collapse
|
10
|
Kurd NS, Lutes LK, Yoon J, Chan SW, Dzhagalov IL, Hoover AR, Robey EA. A role for phagocytosis in inducing cell death during thymocyte negative selection. eLife 2019; 8:48097. [PMID: 31868579 PMCID: PMC6957271 DOI: 10.7554/elife.48097] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2019] [Accepted: 12/21/2019] [Indexed: 12/11/2022] Open
Abstract
Autoreactive thymocytes are eliminated during negative selection in the thymus, a process important for establishing self-tolerance. Thymic phagocytes serve to remove dead thymocytes, but whether they play additional roles during negative selection remains unclear. Here, using a murine thymic slice model in which thymocytes undergo negative selection in situ, we demonstrate that phagocytosis promotes negative selection, and provide evidence for the escape of autoreactive CD8 T cells to the periphery when phagocytosis in the thymus is impaired. We also show that negative selection is more efficient when the phagocyte also presents the negative selecting peptide. Our findings support a model for negative selection in which the death process initiated following strong TCR signaling is facilitated by phagocytosis. Thus, the phagocytic capability of cells that present self-peptides is a key determinant of thymocyte fate.
Collapse
Affiliation(s)
- Nadia S Kurd
- Division of Immunology and Pathogenesis, Department of Molecular and Cell Biology, University of California, Berkeley, Berkeley, United States
| | - Lydia K Lutes
- Division of Immunology and Pathogenesis, Department of Molecular and Cell Biology, University of California, Berkeley, Berkeley, United States
| | - Jaewon Yoon
- Division of Immunology and Pathogenesis, Department of Molecular and Cell Biology, University of California, Berkeley, Berkeley, United States
| | - Shiao Wei Chan
- Division of Immunology and Pathogenesis, Department of Molecular and Cell Biology, University of California, Berkeley, Berkeley, United States
| | - Ivan L Dzhagalov
- Division of Immunology and Pathogenesis, Department of Molecular and Cell Biology, University of California, Berkeley, Berkeley, United States
| | - Ashley R Hoover
- Division of Immunology and Pathogenesis, Department of Molecular and Cell Biology, University of California, Berkeley, Berkeley, United States
| | - Ellen A Robey
- Division of Immunology and Pathogenesis, Department of Molecular and Cell Biology, University of California, Berkeley, Berkeley, United States
| |
Collapse
|
11
|
Yang J, Shangguan J, Eresen A, Li Y, Wang J, Zhang Z. Dendritic cells in pancreatic cancer immunotherapy: Vaccines and combination immunotherapies. Pathol Res Pract 2019; 215:152691. [PMID: 31676092 DOI: 10.1016/j.prp.2019.152691] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/29/2019] [Revised: 10/04/2019] [Accepted: 10/09/2019] [Indexed: 02/06/2023]
Abstract
Despite significant advances over the past decades of research, pancreatic cancer (PC) continues to have the worst 5-year survival of any malignancy. Dendritic cells (DCs) are the most potent professional antigen-presenting cells and are involved in the induction and regulation of antitumor immune responses. DC-based immunotherapy has been used in clinical trials for PC. Although safety, efficacy, and immune activation were reported in patients with PC, DC vaccines have not yet fulfilled their promise. Additional strategies for combinatorial approaches aimed to augment and sustain the antitumor specific immune response elicited by DC vaccines are currently being investigated. Here, we will discuss DC vaccination immunotherapies that are currently under preclinical and clinical investigation and potential combination approaches for treating and improving the survival of PC patients.
Collapse
Affiliation(s)
- Jia Yang
- Department of Radiology, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
| | - Junjie Shangguan
- Department of Radiology, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
| | - Aydin Eresen
- Department of Radiology, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
| | - Yu Li
- Department of Radiology, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA; Department of General Surgery, The Affiliated Hospital of Qingdao University, Qingdao, Shandong, China
| | - Jian Wang
- Department of Radiology, Southwest Hospital, Chongqing, China.
| | - Zhuoli Zhang
- Department of Radiology, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA; Robert H. Lurie Comprehensive Cancer Center, Northwestern University, Chicago, IL, USA.
| |
Collapse
|
12
|
De Niz M, Meehan GR, Tavares J. Intravital microscopy: Imaging host-parasite interactions in lymphoid organs. Cell Microbiol 2019; 21:e13117. [PMID: 31512335 DOI: 10.1111/cmi.13117] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2019] [Revised: 08/25/2019] [Accepted: 09/01/2019] [Indexed: 12/11/2022]
Abstract
Intravital microscopy allows imaging of biological phenomena within living animals, including host-parasite interactions. This has advanced our understanding of both, the function of lymphoid organs during parasitic infections, and the effect of parasites on such organs to allow their survival. In parasitic research, recent developments in this technique have been crucial for the direct study of host-parasite interactions within organs at depths, speeds and resolution previously difficult to achieve. Lymphoid organs have gained more attention as we start to understand their function during parasitic infections and the effect of parasites on them. In this review, we summarise technical and biological findings achieved by intravital microscopy with respect to the interaction of various parasites with host lymphoid organs, namely the bone marrow, thymus, lymph nodes, spleen and the mucosa-associated lymphoid tissue, and present a view into possible future applications.
Collapse
Affiliation(s)
- Mariana De Niz
- Institute of Cell Biology, Heussler Lab, University of Bern, Bern, Switzerland
| | - Gavin R Meehan
- Wellcome Centre for Integrative Parasitology, University of Glasgow, Glasgow, UK
| | - Joana Tavares
- i3S-Instituto de Investigação e Inovação em Saúde, University of Porto, Porto, Portugal.,IBMC-Instituto de Biologia Molecular e Celular, University of Porto, Porto, Portugal
| |
Collapse
|
13
|
Yap JY, Wirasinha RC, Chan A, Howard DR, Goodnow CC, Daley SR. Indirect presentation in the thymus limits naive and regulatory T-cell differentiation by promoting deletion of self-reactive thymocytes. Immunology 2018; 154:522-532. [PMID: 29411880 DOI: 10.1111/imm.12904] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2017] [Revised: 01/23/2018] [Accepted: 01/26/2018] [Indexed: 12/18/2022] Open
Abstract
Acquisition of T-cell central tolerance involves distinct pathways of self-antigen presentation to thymocytes. One pathway termed indirect presentation requires a self-antigen transfer step from thymic epithelial cells (TECs) to bone marrow-derived cells before the self-antigen is presented to thymocytes. The role of indirect presentation in central tolerance is context-dependent, potentially due to variation in self-antigen expression, processing and presentation in the thymus. Here, we report experiments in mice in which TECs expressed a membrane-bound transgenic self-antigen, hen egg lysozyme (HEL), from either the insulin (insHEL) or thyroglobulin (thyroHEL) promoter. Intrathymic HEL expression was less abundant and more confined to the medulla in insHEL mice compared with thyroHEL mice. When indirect presentation was impaired by generating mice lacking MHC class II expression in bone marrow-derived antigen-presenting cells, insHEL-mediated thymocyte deletion was abolished, whereas thyroHEL-mediated deletion occurred at a later stage of thymocyte development and Foxp3+ regulatory T-cell differentiation increased. Indirect presentation increased the strength of T-cell receptor signalling that both self-antigens induced in thymocytes, as assessed by Helios expression. Hence, indirect presentation limits the differentiation of naive and regulatory T cells by promoting deletion of self-reactive thymocytes.
Collapse
Affiliation(s)
- Jin Yan Yap
- Department of Immunology and Infectious Disease, The John Curtin School of Medical Research, The Australian National University, Canberra, ACT, Australia
| | - Rushika C Wirasinha
- Infection and Immunity Programme, Monash Biomedicine Discovery Institute and Department of Biochemistry and Molecular Biology, Monash University, Melbourne, VIC, Australia
| | - Anna Chan
- Infection and Immunity Programme, Monash Biomedicine Discovery Institute and Department of Biochemistry and Molecular Biology, Monash University, Melbourne, VIC, Australia
| | - Debbie R Howard
- Department of Immunology and Infectious Disease, The John Curtin School of Medical Research, The Australian National University, Canberra, ACT, Australia
| | - Christopher C Goodnow
- Immunology Division, Garvan Institute of Medical Research, Sydney, NSW, Australia.,St Vincent's Clinical School, University of New South Wales, Sydney, NSW, Australia
| | - Stephen R Daley
- Department of Immunology and Infectious Disease, The John Curtin School of Medical Research, The Australian National University, Canberra, ACT, Australia.,Infection and Immunity Programme, Monash Biomedicine Discovery Institute and Department of Biochemistry and Molecular Biology, Monash University, Melbourne, VIC, Australia
| |
Collapse
|
14
|
Lancaster JN, Li Y, Ehrlich LIR. Chemokine-Mediated Choreography of Thymocyte Development and Selection. Trends Immunol 2017; 39:86-98. [PMID: 29162323 DOI: 10.1016/j.it.2017.10.007] [Citation(s) in RCA: 36] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2017] [Revised: 10/19/2017] [Accepted: 10/27/2017] [Indexed: 12/30/2022]
Abstract
As they differentiate, thymocytes encounter spatially restricted cues critical for differentiation and selection of a functional, self-tolerant T cell repertoire. Sequential migration of developing T cells through distinct thymic microenvironments is enforced by the ordered expression of chemokine receptors. Herein, we provide an updated perspective on T cell differentiation through the lens of recent advances that illuminate the dynamics of chemokine-driven thymocyte migration, localization, and interactions with stromal cells. We consider these findings in the context of earlier groundwork exploring the contribution of chemokines to T cell development, recent advances regarding the specificity of chemokine signaling, and novel techniques for evaluating the T cell repertoire. We suggest future research should amalgamate visualization of localized cellular interactions with downstream molecular signals.
Collapse
Affiliation(s)
- Jessica N Lancaster
- Department of Molecular Biosciences, The University of Texas at Austin, Austin, TX 78712, USA
| | - Yu Li
- Department of Molecular Biosciences, The University of Texas at Austin, Austin, TX 78712, USA
| | - Lauren I R Ehrlich
- Department of Molecular Biosciences, The University of Texas at Austin, Austin, TX 78712, USA.
| |
Collapse
|
15
|
Lodygin D, Flügel A. Intravital real-time analysis of T-cell activation in health and disease. Cell Calcium 2017; 64:118-129. [DOI: 10.1016/j.ceca.2016.12.007] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2016] [Accepted: 12/23/2016] [Indexed: 01/27/2023]
|
16
|
Daley SR, Teh C, Hu DY, Strasser A, Gray DH. Cell death and thymic tolerance. Immunol Rev 2017; 277:9-20. [DOI: 10.1111/imr.12532] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Affiliation(s)
- Stephen R. Daley
- Infection and Immunity Program; Biomedicine Discovery Institute and Department of Biochemistry and Molecular Biology; Monash University; Melbourne VIC Australia
| | - Charis Teh
- The Walter and Eliza Hall Institute of Medical Research; Melbourne VIC Australia
- Department of Medical Biology; The University of Melbourne; Parkville VIC Australia
| | | | - Andreas Strasser
- The Walter and Eliza Hall Institute of Medical Research; Melbourne VIC Australia
- Department of Medical Biology; The University of Melbourne; Parkville VIC Australia
| | - Daniel H.D. Gray
- The Walter and Eliza Hall Institute of Medical Research; Melbourne VIC Australia
- Department of Medical Biology; The University of Melbourne; Parkville VIC Australia
| |
Collapse
|
17
|
Abstract
The ability of T cells to respond to a wide array of foreign antigens while avoiding reactivity to self is largely determined by cellular selection of developing T cells in the thymus. While a great deal is known about the cell types and molecules involved in T-cell selection in the thymus, our understanding of the spatial and temporal aspects of this process remain relatively poorly understood. Thymocytes are highly motile within the thymus and travel between specialized microenvironments at different phases of their development while interacting with distinct sets of self-peptides and peptide presenting cells. A knowledge of when, where, and how thymocytes encounter self-peptide MHC ligands at different stages of thymic development is key to understanding T-cell selection. In the past several years, our laboratory has investigated this topic using two-photon time-lapse microscopy to directly visualize thymocyte migration and signaling events, together with a living thymic slice preparation to provide a synchronized experimental model of T-cell selection in situ. Here, we discuss recent advances in our understanding of the temporal and spatial aspects of T-cell selection, highlighting our own work, and placing them in the context of work from other groups.
Collapse
Affiliation(s)
- Nadia Kurd
- Division of Immunology and Pathogenesis, Department of Molecular and Cell Biology, University of California, Berkeley, Berkeley, CA, 94720, USA
| | - Ellen A Robey
- Division of Immunology and Pathogenesis, Department of Molecular and Cell Biology, University of California, Berkeley, Berkeley, CA, 94720, USA
| |
Collapse
|
18
|
Hu Z, Lancaster JN, Ehrlich LIR. The Contribution of Chemokines and Migration to the Induction of Central Tolerance in the Thymus. Front Immunol 2015; 6:398. [PMID: 26300884 PMCID: PMC4528182 DOI: 10.3389/fimmu.2015.00398] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2015] [Accepted: 07/20/2015] [Indexed: 02/01/2023] Open
Abstract
As T cells develop, they migrate throughout the thymus where they undergo essential bi-directional signaling with stromal cells in distinct thymic microenvironments. Immature thymocyte progenitors are located in the thymic cortex. Following T cell receptor expression and positive selection, thymocytes undergo a dramatic transition: they become rapidly motile and relocate to the thymic medulla. Antigen-presenting cells (APCs) within the cortex and medulla display peptides derived from a wide array of self-proteins, which promote thymocyte self-tolerance. If a thymocyte is auto-reactive against such antigens, it undergoes either negative selection, via apoptosis, or differentiation into the regulatory T cell lineage. This induction of central tolerance is critical for prevention of autoimmunity. Chemokines and adhesion molecules play an essential role in tolerance induction, as they promote migration of developing thymocytes through the different thymic microenvironments and enhance interactions with APCs displaying self-antigens. Herein, we review the contribution of chemokines and other regulators of thymocyte localization and motility to T cell development, with a focus on their contribution to the induction of central tolerance.
Collapse
Affiliation(s)
- Zicheng Hu
- Ehrlich Laboratory, Department of Molecular Biosciences, Institute for Cellular and Molecular Biology, The University of Texas at Austin , Austin, TX , USA
| | - Jessica Naomi Lancaster
- Ehrlich Laboratory, Department of Molecular Biosciences, Institute for Cellular and Molecular Biology, The University of Texas at Austin , Austin, TX , USA
| | - Lauren I R Ehrlich
- Ehrlich Laboratory, Department of Molecular Biosciences, Institute for Cellular and Molecular Biology, The University of Texas at Austin , Austin, TX , USA
| |
Collapse
|
19
|
Conserved and divergent aspects of human T-cell development and migration in humanized mice. Immunol Cell Biol 2015; 93:716-26. [PMID: 25744551 PMCID: PMC4575952 DOI: 10.1038/icb.2015.38] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2014] [Revised: 02/09/2015] [Accepted: 02/09/2015] [Indexed: 02/07/2023]
Abstract
Humanized mice represent an important model to study the development and function of the human immune system. While it is known that mouse thymic stromal cells can support human T-cell development, the extent of interspecies cross-talk and the degree to which these systems recapitulate normal human T-cell development remain unclear. To address these questions, we compared conventional and non-conventional T-cell development in a neonatal chimera humanized mouse model with that seen in human fetal and neonatal thymus samples, and also examined the impact of a human HLA-A2 transgene expressed by the mouse stroma. Given that dynamic migration and cell–cell interactions are essential for T-cell differentiation, we also studied the intrathymic migration pattern of human thymocytes developing in a murine thymic environment. We found that both conventional T-cell development and intra-thymic migration patterns in humanized mice closely resemble human thymopoiesis. Additionally, we show that developing human thymocytes engage in short, serial interactions with other human hematopoietic-derived cells. However, non-conventional T-cell differentiation in humanized mice differed from both fetal and neonatal human thymopoiesis, including a marked deficiency of Foxp3+ T-cell development. These data suggest that although the murine thymic microenvironment can support a number of aspects of human T-cell development, important differences remain, and additional human-specific factors may be required.
Collapse
|
20
|
Halkias J, Melichar HJ, Taylor KT, Robey EA. Tracking migration during human T cell development. Cell Mol Life Sci 2014; 71:3101-17. [PMID: 24682469 PMCID: PMC11113765 DOI: 10.1007/s00018-014-1607-2] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2013] [Revised: 02/21/2014] [Accepted: 03/11/2014] [Indexed: 01/06/2023]
Abstract
Specialized microenvironments within the thymus are comprised of unique cell types with distinct roles in directing the development of a diverse, functional, and self-tolerant T cell repertoire. As they differentiate, thymocytes transit through a number of developmental intermediates that are associated with unique localization and migration patterns. For example, during one particular developmental transition, immature thymocytes more than double in speed as they become mature T cells that are among the fastest cells in the body. This transition is associated with dramatic changes in the expression of chemokine receptors and their antagonists, cell adhesion molecules, and cytoskeletal components to direct the maturing thymocyte population from the cortex to medulla. Here we discuss the dynamic changes in behavior that occur throughout thymocyte development, and provide an overview of the cell-intrinsic and extrinsic mechanisms that regulate human thymocyte migration.
Collapse
Affiliation(s)
- Joanna Halkias
- Division of Immunology and Pathogenesis, Department of Molecular and Cell Biology, University of California, Berkeley, 142 Life Sciences Addition, #3200, Berkeley, CA, 94720-3200, USA,
| | | | | | | |
Collapse
|
21
|
Distinct phases in the positive selection of CD8+ T cells distinguished by intrathymic migration and T-cell receptor signaling patterns. Proc Natl Acad Sci U S A 2014; 111:E2550-8. [PMID: 24927565 DOI: 10.1073/pnas.1408482111] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023] Open
Abstract
Positive selection of CD8 T cells in the thymus is thought to be a multistep process lasting 3-4 d; however, the discrete steps involved are poorly understood. Here, we examine phenotypic changes, calcium signaling, and intrathymic migration in a synchronized cohort of MHC class I-specific thymocytes undergoing positive selection in situ. Transient elevations in intracellular calcium concentration ([Ca(2+)]i) and migratory pauses occurred throughout the first 24 h of positive selection, becoming progressively briefer and accompanied by a gradual shift in basal [Ca(2+)]i over time. Changes in chemokine-receptor expression and relocalization from the cortex to medulla occurred between 12 and 24 h after the initial encounter with positive-selecting ligands, a time frame at which the majority of thymocytes retain CD4 and CD8 expression and still require T-cell receptor (TCR) signaling to efficiently complete positive selection. Our results identify distinct phases in the positive selection of MHC class I-specific thymocytes that are distinguished by their TCR-signaling pattern and intrathymic location and provide a framework for understanding the multistep process of positive selection in the thymus.
Collapse
|
22
|
Melichar HJ, Ross JO, Herzmark P, Hogquist KA, Robey EA. Distinct temporal patterns of T cell receptor signaling during positive versus negative selection in situ. Sci Signal 2013; 6:ra92. [PMID: 24129702 DOI: 10.1126/scisignal.2004400] [Citation(s) in RCA: 73] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
The recognition by the T cell receptor (TCR) of self-peptides presented by the major histocompatibility complex (MHC) on antigen-presenting cells, such as dendritic cells and thymic epithelial cells, controls T cell fate in the thymus, with weak TCR signals inducing survival (positive selection) and stronger signals inducing death (negative selection). In vitro studies indicate that peptide ligands that induce positive selection stimulate a low, but sustained, pattern of TCR signaling; however, the temporal pattern of TCR signaling in MHC class I-restricted thymocytes (thymocytes that are presented with peptides by MHC class I) in the thymus, under conditions that support positive selection, is unknown. We addressed this question by examining intracellular Ca(2+) dynamics and migratory changes in thymocytes undergoing positive and negative selection in thymic slices. Brief, serial signaling events that were separated by migratory periods and low cytosolic Ca(2+) concentrations correlated with the positive selection of MHC class I-restricted thymocytes, whereas sustained Ca(2+) signaling and the arrest of thymocytes were associated with negative selection. Low-avidity peptides and the presentation of peptides by cortical thymic epithelial cells, rather than dendritic cells, failed to induce strong migratory arrest of thymocytes, which led to transient TCR signaling. Thus, we provide a comparison of positive and negative selection signals in situ and suggest that the absence of strong stop signals distinguishes between positive and negative selection.
Collapse
Affiliation(s)
- Heather J Melichar
- 1Division of Immunology and Pathogenesis, Department of Molecular and Cell Biology, University of California, Berkeley, Berkeley, CA 94720, USA
| | | | | | | | | |
Collapse
|
23
|
Pranzatelli MR, Tate ED, McGee NR, Ransohoff RM. CCR7 signaling in pediatric opsoclonus–myoclonus: Upregulated serum CCL21 expression is steroid-responsive. Cytokine 2013; 64:331-6. [DOI: 10.1016/j.cyto.2013.05.020] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2013] [Revised: 05/02/2013] [Accepted: 05/17/2013] [Indexed: 11/25/2022]
|
24
|
Halkias J, Melichar HJ, Taylor KT, Ross JO, Yen B, Cooper SB, Winoto A, Robey EA. Opposing chemokine gradients control human thymocyte migration in situ. J Clin Invest 2013; 123:2131-42. [PMID: 23585474 DOI: 10.1172/jci67175] [Citation(s) in RCA: 47] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2012] [Accepted: 02/15/2013] [Indexed: 12/23/2022] Open
Abstract
The ordered migration of thymocytes from the cortex to the medulla is critical for the appropriate selection of the mature T cell repertoire. Most studies of thymocyte migration rely on mouse models, but we know relatively little about how human thymocytes find their appropriate anatomical niches within the thymus. Moreover, the signals that retain CD4+CD8+ double-positive (DP) thymocytes in the cortex and prevent them from entering the medulla prior to positive selection have not been identified in mice or humans. Here, we examined the intrathymic migration of human thymocytes in both mouse and human thymic stroma and found that human thymocyte subsets localized appropriately to the cortex on mouse thymic stroma and that MHC-dependent interactions between human thymocytes and mouse stroma could maintain the activation and motility of DP cells. We also showed that CXCR4 was required to retain human DP thymocytes in the cortex, whereas CCR7 promoted migration of mature human thymocytes to the medulla. Thus, 2 opposing chemokine gradients control the migration of thymocytes from the cortex to the medulla. These findings point to significant interspecies conservation in thymocyte-stroma interactions and provide the first evidence that chemokines not only attract mature thymocytes to the medulla, but also play an active role in retaining DP thymocytes in the cortex prior to positive selection.
Collapse
Affiliation(s)
- Joanna Halkias
- Division of Immunology and Pathogenesis, Department of Molecular and Cell Biology, UC Berkeley, Berkeley, California 94720-3200, USA
| | | | | | | | | | | | | | | |
Collapse
|
25
|
Abstract
Ten years ago, in 2002, the introduction of dynamic in vivo imaging to immunologists set a new standard for studying immune responses. In particular, two-photon imaging has provided tremendous insights into immune cell dynamics in various contexts, including infection, cancer, transplantation and autoimmunity. Whereas initial studies were restricted to the migration of and interactions between immune cells, recent advances are bringing intravital imaging to a new level in which cell dynamics and function can be investigated simultaneously. These exciting developments further broaden the applications of immunoimaging and provide unprecedented opportunities to probe and decode immune cell communication in situ.
Collapse
|
26
|
Guerri L, Peguillet I, Geraldo Y, Nabti S, Premel V, Lantz O. Analysis of APC types involved in CD4 tolerance and regulatory T cell generation using reaggregated thymic organ cultures. THE JOURNAL OF IMMUNOLOGY 2013; 190:2102-10. [PMID: 23365074 DOI: 10.4049/jimmunol.1202883] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Tolerance to self-Ags is generated in the thymus. Both epithelial and hematopoietic thymic stromal cells play an active and essential role in this process. However, the role of each of the various stromal cell types remains unresolved. To our knowledge, we describe the first comparative analysis of several types of thymic hematopoietic stromal cells (THSCs) for their ability to induce CD4 tolerance to self, in parallel with the thymic epithelium. The THSCs--two types of conventional dendritic cells (cDCs), plasmacytoid dendritic cells, macrophages (MΦs), B lymphocytes, and eosinophils--were first characterized and quantified in adult mouse thymus. They were then examined in reaggregated thymic organ cultures containing mixtures of monoclonal and polyclonal thymocytes. This thymocyte mixture allows for the analysis of Ag-specific events while avoiding the extreme skewing frequently seen in purely monoclonal systems. Our data indicate that thymic epithelium alone is capable of promoting self-tolerance by eliminating autoreactive CD4 single-positive thymocytes and by supporting regulatory T cell (Treg) development. We also show that both non-Treg CD4 single-positive thymocytes and Tregs are efficiently deleted by the two populations of cDCs present in the thymus, as well as to a lesser extent by MΦs. Plasmacytoid dendritic cells, B lymphocytes, and eosinophils were not able to do so. Finally, cDCs were also the most efficient THSCs at supporting Treg development in the thymus, suggesting that although they may share some characteristics required for negative selection with MΦs, they do not share those required for the support of Treg development, making cDCs a unique cell subset in the thymus.
Collapse
Affiliation(s)
- Lucia Guerri
- INSERM U932 and Centre d'Investigation Clinique, CIC-BT507, Institut Curie, Paris 75005, France.
| | | | | | | | | | | |
Collapse
|
27
|
Abstract
Dendritic cells (DCs) are specialized sentinels responsible for coordinating adaptive immunity. This function is dependent upon coupled sensitivity to environmental signs of inflammation and infection to cellular maturation-the programmed alteration of DC phenotype and function to enhance immune cell activation. Although DCs are thus well equipped to respond to pathogens, maturation triggers are not unique to infection. Given that immune cells are exquisitely sensitive to the biological functions of DCs, we now appreciate that multiple layers of suppression are required to restrict the environmental sensitivity, cellular maturation, and even life span of DCs to prevent aberrant immune activation during the steady state. At the same time, steady-state DCs are not quiescent but rather perform key functions that support homeostasis of numerous cell types. Here we review these functions and molecular mechanisms of suppression that control steady-state DC maturation. Corruption of these steady-state operatives has diverse immunological consequences and pinpoints DCs as potent drivers of autoimmune and inflammatory disease.
Collapse
Affiliation(s)
- Gianna Elena Hammer
- Department of Medicine, University of California, San Francisco, California 94143
| | - Averil Ma
- Department of Medicine, University of California, San Francisco, California 94143
| |
Collapse
|
28
|
Abstract
The development of CD4(+) helper and CD8(+) cytotoxic T-cells expressing the αβ form of the T-cell receptor (αβTCR) takes place in the thymus, a primary lymphoid organ containing distinct cortical and medullary microenvironments. While the cortex represents a site of early T-cell precursor development, and the positive selection of CD4(+)8(+) thymocytes, the thymic medulla plays a key role in tolerance induction, ensuring that thymic emigrants are purged of autoreactive αβTCR specificities. In recent years, advances have been made in understanding the development and function of thymic medullary epithelial cells, most notably the subset defined by expression of the Autoimmune Regulator (Aire) gene. Here, we summarize current knowledge of the developmental mechanisms regulating thymus medulla development, and examine the role of the thymus medulla in recessive (negative selection) and dominant (T-regulatory cell) tolerance.
Collapse
|
29
|
Dzhagalov I, Phee H. How to find your way through the thymus: a practical guide for aspiring T cells. Cell Mol Life Sci 2011; 69:663-82. [PMID: 21842411 DOI: 10.1007/s00018-011-0791-6] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2011] [Revised: 07/25/2011] [Accepted: 07/25/2011] [Indexed: 01/16/2023]
Abstract
Thymocytes must complete an elaborate developmental program in the thymus to ultimately generate T cells that express functional but neither harmful nor useless TCRs. Each developmental step coincides with dynamic relocation of the thymocytes between anatomically discrete thymic microenvironments, suggesting that thymocytes' migration is tightly regulated by their developmental status. Chemokines produced by thymic stromal cells and chemokine receptors on the thymocytes play an indispensable role in guiding developing thymocytes into the different microenvironments. In addition to long-range migration, chemokines increase the thymocytes' motility, enhancing their interaction with stromal cells. During the past several years, much progress has been made to determine the various signals that guide thymocytes on their journey within the thymus. In this review, we summarize the progress in identifying chemokines and other chemoattractant signals that direct intrathymic migration. Furthermore, we discuss the recent advances of two-photon microscopy in determining dynamic motility and interaction behavior of thymocytes within distinct compartments to provide a better understanding of the relationship between thymocyte motility and development.
Collapse
Affiliation(s)
- Ivan Dzhagalov
- LSA, Room 479, Department of Molecular and Cell Biology, Division of Immunology and Pathogenesis, University of California Berkeley, Berkeley, CA 94720-3200, USA.
| | | |
Collapse
|
30
|
Padfield D, Rittscher J, Roysam B. Coupled minimum-cost flow cell tracking for high-throughput quantitative analysis. Med Image Anal 2011; 15:650-68. [DOI: 10.1016/j.media.2010.07.006] [Citation(s) in RCA: 112] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2009] [Revised: 07/16/2010] [Accepted: 07/21/2010] [Indexed: 11/26/2022]
|
31
|
Sanos SL, Nowak J, Fallet M, Bajenoff M. Stromal Cell Networks Regulate Thymocyte Migration and Dendritic Cell Behavior in the Thymus. THE JOURNAL OF IMMUNOLOGY 2011; 186:2835-41. [DOI: 10.4049/jimmunol.1003563] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
|
32
|
Nitta T, Ohigashi I, Nakagawa Y, Takahama Y. Cytokine crosstalk for thymic medulla formation. Curr Opin Immunol 2010; 23:190-7. [PMID: 21194915 DOI: 10.1016/j.coi.2010.12.002] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2010] [Revised: 11/24/2010] [Accepted: 12/03/2010] [Indexed: 01/12/2023]
Abstract
The medullary microenvironment of the thymus plays a crucial role in the establishment of self-tolerance through the deletion of self-reactive thymocytes and the generation of regulatory T cells. Crosstalk or bidirectional signal exchanges between developing thymocytes and medullary thymic epithelial cells (mTECs) contribute to the formation of the thymic medulla. Recent studies have identified the molecules that mediate thymic crosstalk. Tumor necrosis factor superfamily cytokines, including RANKL, CD40L, and lymphotoxin, produced by positively selected thymocytes and lymphoid tissue inducer cells promote the proliferation and differentiation of mTECs. In return, CCR7 ligand chemokines produced by mTECs facilitate the migration of positively selected thymocytes to the medulla. The cytokine crosstalk between developing thymocytes and mTECs nurtures the formation of the thymic medulla and thereby regulates the establishment of self-tolerance.
Collapse
Affiliation(s)
- Takeshi Nitta
- Division of Experimental Immunology, Institute for Genome Research, University of Tokushima, Tokushima 770-8503, Japan
| | | | | | | |
Collapse
|
33
|
Bunting MD, Comerford I, McColl SR. Finding their niche: chemokines directing cell migration in the thymus. Immunol Cell Biol 2010; 89:185-96. [PMID: 21135866 DOI: 10.1038/icb.2010.142] [Citation(s) in RCA: 49] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
T lymphocytes are generated throughout life, arising from bone marrow-derived progenitors that complete an essential developmental process in the thymus. Thymic T cell education leads to the generation of a self-restricted and largely self-tolerant peripheral T-cell pool and is facilitated by interactions with thymic stromal cells residing in distinct supportive niches. The signals governing thymocyte precursor migration into the thymus, directing thymocyte navigation through thymic microenvironments and mature T-cell egress into circulation were, until recently, largely unknown, but presumed to be mediated to a large extent by chemokine signalling. Recent studies have now uncovered various specific functions for members of the chemokine superfamily in the thymus. These studies have not only revealed distinct but also in some cases overlapping roles for several chemokine family members in various thymocyte migration events and have also shown that homing and positioning of other cells in the thymus, such as dendritic cells and natural killer T cells is also chemokine-dependent. Here, we discuss current understanding of the role of chemokines in the thymus and highlight key future avenues for investigation in this field.
Collapse
Affiliation(s)
- Mark D Bunting
- Chemokine Biology Laboratory, Discipline of Microbiology and Immunology, The School of Molecular and Biomedical Science, The University of Adelaide, South Australia, Australia
| | | | | |
Collapse
|
34
|
Quantifying subcellular distribution of fluorescent fusion proteins in cells migrating within tissues. Immunol Cell Biol 2010; 89:549-57. [PMID: 20956985 DOI: 10.1038/icb.2010.122] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
The movement of proteins within cells can provide dynamic indications of cell signaling and cell polarity, but methods are needed to track and quantify subcellular protein movement within tissue environments. Here we present a semiautomated approach to quantify subcellular protein location for hundreds of migrating cells within intact living tissue using retrovirally expressed fluorescent fusion proteins and time-lapse two-photon microscopy of intact thymic lobes. We have validated the method using GFP-PKCζ, a marker for cell polarity, and LAT-GFP, a marker for T-cell receptor signaling, and have related the asymmetric distribution of these proteins to the direction and speed of cell migration. These approaches could be readily adapted to other fluorescent fusion proteins, tissues and biological questions.
Collapse
|
35
|
Regulation of thymocyte positive selection and motility by GIT2. Nat Immunol 2010; 11:503-11. [PMID: 20431621 DOI: 10.1038/ni.1868] [Citation(s) in RCA: 40] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2010] [Accepted: 03/22/2010] [Indexed: 11/08/2022]
Abstract
Thymocytes are highly motile cells that migrate under the influence of chemokines in distinct thymic compartments as they mature. The motility of thymocytes is tightly regulated; however, the molecular mechanisms that control thymocyte motility are not well understood. Here we report that G protein-coupled receptor kinase-interactor 2 (GIT2) was required for efficient positive selection. Notably, Git2(-/-) double-positive thymocytes showed greater activation of the small GTPase Rac, actin polymerization and migration toward the chemokines CXCL12 (SDF-1) and CCL25 in vitro. By two-photon laser-scanning microscopy, we found that the scanning activity of Git2(-/-) thymocytes was compromised in the thymic cortex, which suggests GIT2 has a key role in regulating the chemokine-mediated motility of double-positive thymocytes.
Collapse
|
36
|
Abstract
Natural regulatory T cells (nTregs) are defined by their inherent ability to establish and maintain peripheral self-tolerance. In recent years, the development of nTregs has come under close examination with the advent of Forkhead Box P3 protein (FOXP3)-green fluorescent protein reporter mice that pinpointed the initiation of FOXP3 expression within the thymus. The mechanism and pathway of nTreg development has only recently been studied in detail and to a large degree remains unclear. In this review, we will discuss our current understanding of nTreg lineage choice and development from a cellular and intracellular standpoint.
Collapse
Affiliation(s)
- Matthew L Bettini
- Department of Immunology, St. Jude Children's Research Hospital, Memphis, Tennessee 38105-3678, USA
| | | |
Collapse
|
37
|
Morley SC, Wang C, Lo WL, Lio CWJ, Zinselmeyer BH, Miller MJ, Brown EJ, Allen PM. The actin-bundling protein L-plastin dissociates CCR7 proximal signaling from CCR7-induced motility. THE JOURNAL OF IMMUNOLOGY 2010; 184:3628-38. [PMID: 20194718 DOI: 10.4049/jimmunol.0903851] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/23/2023]
Abstract
Chemokines promote lymphocyte motility by triggering F-actin rearrangements and inducing cellular polarization. Chemokines can also enhance cell-cell adhesion and costimulate T cells. In this study, we establish a requirement for the actin-bundling protein L-plastin (LPL) in CCR7- and sphingosine-1-phosphate-mediated T cell chemotaxis using LPL(-/-) mice. Disrupted motility of mature LPL(-/-) thymocytes manifested in vivo as diminished thymic egress. Two-photon microscopy of LPL(-/-) lymphocytes revealed reduced velocity and motility in lymph nodes. Defective migration resulted from defective cellular polarization following CCR7 ligation, as CCR7 did not polarize to the leading edge in chemokine-stimulated LPL(-/-) T cells. However, CCR7 signaling to F-actin polymerization and CCR7-mediated costimulation was intact in LPL(-/-) lymphocytes. The differential requirement for LPL in CCR7-induced cellular adhesion and CCR7-induced motility allowed assessment of the contribution of CCR7-mediated motility to positive selection of thymocytes and lineage commitment. Results suggest that normal motility is not required for CCR7 to function in positive selection and lineage commitment. We thus identify LPL as a molecule critical for CCR7-mediated motility but dispensable for early CCR7 signaling. The requirement for actin bundling by LPL for polarization reveals a novel mechanism of regulating actin dynamics during T cell motility.
Collapse
Affiliation(s)
- Sharon Celeste Morley
- Department of Pathology and Immunology, Washington University School of Medicine, St Louis, MO 63110, USA
| | | | | | | | | | | | | | | |
Collapse
|
38
|
Ehrlich LIR, Oh DY, Weissman IL, Lewis RS. Differential contribution of chemotaxis and substrate restriction to segregation of immature and mature thymocytes. Immunity 2009; 31:986-98. [PMID: 19962328 DOI: 10.1016/j.immuni.2009.09.020] [Citation(s) in RCA: 83] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2009] [Revised: 08/30/2009] [Accepted: 09/21/2009] [Indexed: 12/24/2022]
Abstract
T cell development requires sequential localization of thymocyte subsets to distinct thymic microenvironments. To address mechanisms governing this segregation, we used two-photon microscopy to visualize migration of purified thymocyte subsets in defined microenvironments within thymic slices. Double-negative (CD4(-)8(-)) and double-positive (CD4(+)8(+)) thymocytes were confined to cortex where they moved slowly without directional bias. DP cells accumulated and migrated more rapidly in a specialized inner-cortical microenvironment, but were unable to migrate on medullary substrates. In contrast, CD4 single positive (SP) thymocytes migrated directionally toward the medulla, where they accumulated and moved very rapidly. Our results revealed a requisite two-step process governing CD4 SP cell medullary localization: the chemokine receptor CCR7 mediated chemotaxis of CD4 SP cells towards medulla, whereas a distinct pertussis-toxin sensitive pathway was required for medullary entry. These findings suggest that developmentally regulated responses to both chemotactic signals and specific migratory substrates guide thymocytes to specific locations in the thymus.
Collapse
Affiliation(s)
- Lauren I Richie Ehrlich
- Institute for Stem Cell Biology and Regenerative Medicine, Stanford University, Stanford, CA 94305, USA
| | | | | | | |
Collapse
|
39
|
Li A, Qin L, Zhu D, Zhu R, Sun J, Wang S. Signalling pathways involved in the activation of dendritic cells by layered double hydroxide nanoparticles. Biomaterials 2009; 31:748-56. [PMID: 19853910 DOI: 10.1016/j.biomaterials.2009.09.095] [Citation(s) in RCA: 48] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2009] [Accepted: 09/29/2009] [Indexed: 12/18/2022]
Abstract
Layered double hydroxide (LDH) nanoparticles are attractive as potential drug vectors for the targeting not only of tissues, but also of intracellular organelles, and particularly the acidic endolysosomes created after cell endocytosis. The purpose of this study was to investigate the ability of LDH nanoparticles designed as vectors to activate dendritic cells (DCs), as measured by various cellular functions. The study also explored the possible signaling pathway through which the LDH nanoparticles exerted their effects on the cellular functions of DCs. First, LDH nanoparticles with different ratios of Mg(OH)(2) to Al(OH)(3) (1:1, 2:1 and 3:1, called R1, R2 and R3 respectively) were optimized and had a hydrodynamic diameter of 57 nm with a zeta potential of +35 mV. Then, the efficient endocytosis of the optimized LDH nanoparticles by bone marrow-derived dendritic cells (MDDCs) was monitored by fluorescence-activated cell sorting. The effect of R1, R2 and R3 on the expression of the pro- and anti-inflammatory cytokines (TNF-alpha, IL-6, and IL-12) and the co-stimulatory molecules (CD40, CD80, CD86, and MHC class II) in MDDCs was examined. The exposure of R1 caused a dose-dependent increase in the expression of TNF-alpha, IL-12, CD86 and CD40, while R2 and R3 did not up-regulate these cytokines and co-stimulatory molecules. Migration assays showed that R1 could increase the migration capacity of DCs to CCL21 and up-regulate the expression of CCR7. Furthermore, we found that R1 significantly increased the NF-kappaB expression in the nucleus (in a dose-dependent manner) and promoted the degradation of total IkappaBalpha levels, indicating that the NF-kappaB signaling pathway might involve in an R1-induced DC activation. Our results suggested that LDH nanoparticles, in the future, may function as a useful vector for ex vivo engineering to promote vaccine delivery in immune cells.
Collapse
Affiliation(s)
- Ang Li
- Shanghai key laboratory of cell signaling and diseases, School of Life Science and Technology, Tongji University, Shanghai 200092, PR China
| | | | | | | | | | | |
Collapse
|
40
|
Baba T, Nakamoto Y, Mukaida N. Crucial contribution of thymic Sirp alpha+ conventional dendritic cells to central tolerance against blood-borne antigens in a CCR2-dependent manner. THE JOURNAL OF IMMUNOLOGY 2009; 183:3053-63. [PMID: 19675159 DOI: 10.4049/jimmunol.0900438] [Citation(s) in RCA: 103] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
Thymic dendritic cells (DCs) as well as thymic epithelial cells are presumed to be major sentinels in central tolerance by inducing the apoptosis of autoreactive T progenitor cells. The thymic DC population is composed of heterogeneous subsets including CD11c(+)B220(+) plasmacytoid DCs, CD11c(+)B220(-)CD8alpha(+) signal regulatory protein alpha (Sirpalpha)(-) and CD11c(+)B220(-)CD8alpha(-)Sirpalpha(+) conventional DCs (cDCs). However, the distinctive role of each DC subset remains undefined. We show herein that Sirpalpha(+) cDCs, a minor subpopulation, was disseminated in the thymic cortical area with some of them uniquely localized inside perivascular regions and nearby small vessels in the thymus. The Sirpalpha(+) but not Sirpalpha(-) cDC subset can selectively capture blood-circulating Ags. Moreover, in CCR2-deficient mice, the thymic Sirpalpha(+) cDC subset, but not other thymic cell components, was moderately decreased especially in the perivascular regions. Concomitantly, these mice exhibited a modest impairment in intrathymic negative selection against blood-borne Ags, with the reduced capacity to uptake blood-borne Ags. Given their intrathymic cortical localization, CD11c(+)B220(-)CD8alpha(-)Sirpalpha(+) cDCs can have a unique role in the development of central tolerance against circulating peripheral Ags, at least partially in a CCR2-dependent manner.
Collapse
Affiliation(s)
- Tomohisa Baba
- Division of Molecular Bioregulation, Cancer Research Institute, Kanazawa University, Kanazawa, Ishikawa, Japan
| | | | | |
Collapse
|
41
|
The impact of negative selection on thymocyte migration in the medulla. Nat Immunol 2009; 10:823-30. [PMID: 19543275 PMCID: PMC2793676 DOI: 10.1038/ni.1761] [Citation(s) in RCA: 116] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2009] [Accepted: 06/01/2009] [Indexed: 01/14/2023]
Abstract
Developing thymocytes are screened for self-reactivity before exiting the thymus, but how thymocytes scan the medulla for self-antigens is unclear. Using two-photon microscopy, we observed that medullary thymocytes migrated rapidly and made frequent, transient contacts with dendritic cells. In the presence of a negative selecting ligand, thymocytes slowed, became confined to areas of approximately 30 microns in diameter, and had increased contact with dendritic cells surrounding confinement zones. One third of polyclonal medullary thymocytes also exhibited confined, slower migration, and may correspond to auto-reactive thymocytes. Our data suggest that many auto-reactive thymocytes do not undergo immediate arrest and death upon encounter with a negative selecting ligand, but rather adopt an altered migration program while remaining within the medullary microenvironment.
Collapse
|
42
|
Chen Y, Ladi E, Herzmark P, Robey E, Roysam B. Automated 5-D analysis of cell migration and interaction in the thymic cortex from time-lapse sequences of 3-D multi-channel multi-photon images. J Immunol Methods 2008; 340:65-80. [PMID: 18992251 DOI: 10.1016/j.jim.2008.09.024] [Citation(s) in RCA: 41] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2008] [Revised: 09/24/2008] [Accepted: 09/30/2008] [Indexed: 11/16/2022]
Abstract
This paper presents automated methods to quantify dynamic phenomena such as cell-cell interactions and cell migration patterns from time-lapse series of multi-channel three-dimensional image stacks of living specimens. Various 5-dimensional (x, y, z, t, lambda) images containing dendritic cells (DC), and T-cells or thymocytes in the developing mouse thymic cortex and lymph node were acquired by two-photon laser scanning microscopy (TPLSM). The cells were delineated automatically using a mean-shift clustering algorithm. This enables morphological measurements to be computed. A robust multiple-hypothesis tracking algorithm was used to track thymocytes (the DC were stationary). The tracking data enable dynamic measurements to be computed, including migratory patterns of thymocytes, and duration of thymocyte-DC contacts. Software was developed for efficient inspection, corrective editing, and validation of the automated analysis results. Our software-generated results agreed with manually generated measurements to within 8%.
Collapse
Affiliation(s)
- Ying Chen
- Department of Electrical, Computer, and System Engineering, Rensselaer Polytechnic Institute, Troy, NY 12180, USA
| | | | | | | | | |
Collapse
|
43
|
McCaughtry TM, Hogquist KA. Central tolerance: what have we learned from mice? Semin Immunopathol 2008; 30:399-409. [PMID: 19015857 DOI: 10.1007/s00281-008-0137-0] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2008] [Accepted: 10/01/2008] [Indexed: 10/21/2022]
Abstract
Producing a healthy immune system capable of defending against pathogens, while avoiding autoimmunity, is dependent on thymic selection. Positive selection yields functional T cells that have the potential to recognize both self and foreign antigens. Therefore, negative selection exists to manage potentially self-reactive cells. Negative selection results from the induction of anergy, receptor editing, clonal diversion (agonist selection), and/or clonal deletion (apoptosis) in self-reactive clones. Clonal deletion has been inherently difficult to study because the cells of interest are undergoing apoptosis and being eliminated quickly. Furthermore, analysis of clonal deletion in humans has proved even more difficult due to availability of samples and lack of reagents. Mouse models have thus been instrumental in achieving our current understanding of central tolerance, and the evolution of elegant model systems has led to an explosion of new data to be assimilated. This review will focus on recent advances in the field of clonal deletion with respect to three aspects: the development of physiological model systems, signaling pathways that lead to apoptosis, and antigen presenting cell types involved in the induction of clonal deletion.
Collapse
Affiliation(s)
- Tom M McCaughtry
- Center for Immunology, Laboratory Medicine & Pathology, University of Minnesota, Mayo Mail Code 334, 420 Delaware Street SE, Minneapolis, MN, 55454, USA
| | | |
Collapse
|