1
|
Stankiewicz LN, Salim K, Flaschner EA, Wang YX, Edgar JM, Durland LJ, Lin BZB, Bingham GC, Major MC, Jones RD, Blau HM, Rideout EJ, Levings MK, Zandstra PW, Rossi FMV. Sex-biased human thymic architecture guides T cell development through spatially defined niches. Dev Cell 2025; 60:152-169.e8. [PMID: 39383865 DOI: 10.1016/j.devcel.2024.09.011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2024] [Revised: 06/11/2024] [Accepted: 09/10/2024] [Indexed: 10/11/2024]
Abstract
Within the thymus, regulation of the cellular crosstalk directing T cell development depends on spatial interactions within specialized niches. To create a spatially defined map of tissue niches guiding human postnatal T cell development, we employed the multidimensional imaging platform co-detection by indexing (CODEX) as well as cellular indexing of transcriptomes and epitopes sequencing (CITE-seq) and assay for transposase accessible chromatin sequencing (ATAC-seq). We generated age-matched 4- to 5-month-old human postnatal thymus datasets for male and female donors, identifying significant sex differences in both T cell and thymus biology. We demonstrate a possible role for JAG ligands in directing thymic-like dendritic cell development, identify important functions of a population of extracellular matrix (ECM)- fibroblasts, and characterize the medullary niches surrounding Hassall's corpuscles. Together, these data represent an age-matched spatial multiomic resource to investigate how sex-based differences in thymus regulation and T cell development arise, providing an essential resource to understand the mechanisms underlying immune function and dysfunction in males and females.
Collapse
Affiliation(s)
- Laura N Stankiewicz
- School of Biomedical Engineering, University of British Columbia, 2222 Health Sciences Mall, Vancouver, BC V6T 2B9, Canada
| | - Kevin Salim
- Department of Surgery, University of British Columbia, 2775 Laurel Street, Vancouver, BC V5Z 1M9, Canada; BC Children's Hospital Research Institute, 938 W 28th Ave, Vancouver, BC V5Z 4H4, Canada
| | - Emily A Flaschner
- School of Biomedical Engineering, University of British Columbia, 2222 Health Sciences Mall, Vancouver, BC V6T 2B9, Canada
| | - Yu Xin Wang
- Center for Genetic Disorders and Aging, Sanford Burnham Prebys Medical Discovery Institute, 10901 North Torrey Pines Road, La Jolla, CA 92037, USA
| | - John M Edgar
- School of Biomedical Engineering, University of British Columbia, 2222 Health Sciences Mall, Vancouver, BC V6T 2B9, Canada
| | - Lauren J Durland
- School of Biomedical Engineering, University of British Columbia, 2222 Health Sciences Mall, Vancouver, BC V6T 2B9, Canada
| | - Bruce Z B Lin
- School of Biomedical Engineering, University of British Columbia, 2222 Health Sciences Mall, Vancouver, BC V6T 2B9, Canada
| | - Grace C Bingham
- Department of Biomedical Engineering, University of Virginia, Charlottesville, VA, USA
| | - Matthew C Major
- School of Biomedical Engineering, University of British Columbia, 2222 Health Sciences Mall, Vancouver, BC V6T 2B9, Canada
| | - Ross D Jones
- School of Biomedical Engineering, University of British Columbia, 2222 Health Sciences Mall, Vancouver, BC V6T 2B9, Canada
| | - Helen M Blau
- Baxter Laboratory for Stem Cell Biology, Department of Microbiology and Immunology, Stanford University School of Medicine, 269 Campus Drive, Stanford, CA 94305, USA
| | - Elizabeth J Rideout
- Life Sciences Institute, University of British Columbia, 2350 Health Sciences Mall, Vancouver, BC V6T 1Z3, Canada
| | - Megan K Levings
- School of Biomedical Engineering, University of British Columbia, 2222 Health Sciences Mall, Vancouver, BC V6T 2B9, Canada; Department of Surgery, University of British Columbia, 2775 Laurel Street, Vancouver, BC V5Z 1M9, Canada; BC Children's Hospital Research Institute, 938 W 28th Ave, Vancouver, BC V5Z 4H4, Canada
| | - Peter W Zandstra
- School of Biomedical Engineering, University of British Columbia, 2222 Health Sciences Mall, Vancouver, BC V6T 2B9, Canada; Michael Smith Laboratories, University of British Columbia, 2185 East Mall, Vancouver, BC V6T 1Z4, Canada.
| | - Fabio M V Rossi
- School of Biomedical Engineering, University of British Columbia, 2222 Health Sciences Mall, Vancouver, BC V6T 2B9, Canada; Department of Medical Genetics, University of British Columbia, 2350 Health Sciences Mall, Vancouver, BC V6T 2A1, Canada.
| |
Collapse
|
2
|
Pyaram K, Chang CH. NKT Cells and Other Innate T Cells: The Immune Cells That Do Not Follow the Rules. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2024; 213:3-5. [PMID: 38885470 DOI: 10.4049/jimmunol.2400243] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/01/2024] [Accepted: 05/01/2024] [Indexed: 06/20/2024]
Abstract
This Pillars of Immunology article is a commentary on “A subset of CD4+ thymocytes selected by MHC class I molecules,” a pivotal article by A. Bendelac, N. Killeen, D.R. Littman, and R.H. Schwartz published in Science in 1994, marking the discovery of NKT cells and paving the way for the identification and characterization of other innate T cells. https://doi.org/10.1126/science.7907820.
Collapse
Affiliation(s)
- Kalyani Pyaram
- Department of Cancer Biology, University of Kansas Medical Center, Kansas City, KS
| | | |
Collapse
|
3
|
Locher V, Park S, Bunis DG, Makredes S, Mayer M, Burt TD, Fragiadakis GK, Halkias J. Homeostatic cytokines reciprocally modulate the emergence of prenatal effector PLZF+CD4+ T cells in humans. JCI Insight 2023; 8:e164672. [PMID: 37856221 PMCID: PMC10721317 DOI: 10.1172/jci.insight.164672] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2022] [Accepted: 10/11/2023] [Indexed: 10/21/2023] Open
Abstract
The development of human prenatal adaptive immunity progresses faster than previously appreciated, with the emergence of memory CD4+ T cells alongside regulatory T cells by midgestation. We previously identified a prenatal specific population of promyelocytic leukemia zinc finger-positive (PLZF+) CD4+ T cells with heightened effector potential that were enriched in the developing intestine and accumulated in the cord blood of infants exposed to prenatal inflammation. However, the signals that drive their tissue distribution and effector maturation are unknown. Here, we define the transcriptional and functional heterogeneity of human prenatal PLZF+CD4+ T cells and identify the compartmentalization of T helper-like (Th-like) effector function across the small intestine (SI) and mesenteric lymph nodes (MLNs). IL-7 was more abundant in the SI relative to the MLNs and drove the preferential expansion of naive PLZF+CD4+ T cells via enhanced STAT5 and MEK/ERK signaling. Exposure to IL-7 was sufficient to induce the acquisition of CD45RO expression and rapid effector function in a subset of PLZF+CD4+ T cells, identifying a human analog of memory phenotype CD4+ T cells. Further, IL-7 modulated the differentiation of Th1- and Th17-like PLZF+CD4+ T cells and thus likely contributes to the anatomic compartmentalization of human prenatal CD4+ T cell effector function.
Collapse
Affiliation(s)
- Veronica Locher
- Division of Neonatology, Department of Pediatrics, and
- Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, UCSF, San Francisco, California, USA
- Committee on Immunology, University of Chicago, Chicago, Illinois, USA
| | - Sara Park
- Division of Neonatology, Department of Pediatrics, and
- Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, UCSF, San Francisco, California, USA
| | - Daniel G. Bunis
- Bakar ImmunoX Initiative and
- CoLabs, UCSF, San Francisco, California, USA
| | - Stephanie Makredes
- Division of Neonatology, Department of Pediatrics, and
- Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, UCSF, San Francisco, California, USA
| | - Margareta Mayer
- Division of Neonatology, Department of Pediatrics, and
- Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, UCSF, San Francisco, California, USA
| | - Trevor D. Burt
- Division of Neonatology and the Children’s Health & Discovery Initiative, Department of Pediatrics, Duke University School of Medicine, Durham, North Carolina, USA
| | - Gabriela K. Fragiadakis
- Bakar ImmunoX Initiative and
- CoLabs, UCSF, San Francisco, California, USA
- Division of Rheumatology, Department of Medicine, UCSF, San Francisco, California, USA
| | - Joanna Halkias
- Division of Neonatology, Department of Pediatrics, and
- Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, UCSF, San Francisco, California, USA
- Bakar ImmunoX Initiative and
| |
Collapse
|
4
|
Stankiewicz LN, Salim K, Flaschner EA, Wang YX, Edgar JM, Lin BZB, Bingham GC, Major MC, Jones RD, Blau HM, Rideout EJ, Levings MK, Zandstra PW, Rossi FMV. Sex biased human thymic architecture guides T cell development through spatially defined niches. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.04.13.536804. [PMID: 37090676 PMCID: PMC10120731 DOI: 10.1101/2023.04.13.536804] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/25/2023]
Abstract
Within the thymus, regulation of the cellular cross-talk directing T cell development is dependent on spatial interactions within specialized niches. To create a holistic, spatially defined map of tissue niches guiding postnatal T cell development we employed the multidimensional imaging platform CO-detection by indEXing (CODEX), as well as CITE-seq and ATAC-seq. We generated age-matched 4-5-month-old postnatal thymus datasets for male and female donors, and identify significant sex differences in both T cell and thymus biology. We demonstrate a crucial role for JAG ligands in directing thymic-like dendritic cell development, reveal important functions of a novel population of ECM- fibroblasts, and characterize the medullary niches surrounding Hassall's corpuscles. Together, these data represent a unique age-matched spatial multiomic resource to investigate how sex-based differences in thymus regulation and T cell development arise, and provide an essential resource to understand the mechanisms underlying immune function and dysfunction in males and females.
Collapse
Affiliation(s)
| | - Kevin Salim
- Department of Surgery, University of British Columbia, Canada
- BC Children’s Hospital Research Institute, Canada
| | - Emily A Flaschner
- School of Biomedical Engineering, University of British Columbia, Canada
| | - Yu Xin Wang
- Center for Genetic Disorders and Aging, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, California, USA
| | - John M Edgar
- School of Biomedical Engineering, University of British Columbia, Canada
| | - Bruce ZB Lin
- School of Biomedical Engineering, University of British Columbia, Canada
| | - Grace C Bingham
- Department of Biomedical Engineering, University of Virginia, USA
| | - Matthew C Major
- School of Biomedical Engineering, University of British Columbia, Canada
| | - Ross D Jones
- School of Biomedical Engineering, University of British Columbia, Canada
| | - Helen M Blau
- Baxter Laboratory for Stem Cell Biology, Department of Microbiology and Immunology, Stanford University School of Medicine, Stanford, USA
| | | | - Megan K Levings
- School of Biomedical Engineering, University of British Columbia, Canada
- Department of Surgery, University of British Columbia, Canada
- BC Children’s Hospital Research Institute, Canada
| | - Peter W Zandstra
- School of Biomedical Engineering, University of British Columbia, Canada
- Michael Smith Laboratories, University of British Columbia, Canada
- These authors contributed equally
- Lead contact
| | - Fabio MV Rossi
- School of Biomedical Engineering, University of British Columbia, Canada
- These authors contributed equally
- Lead contact
| |
Collapse
|
5
|
Jardine L, Schim van der Loeff I, Haq IJ, Sproat TDR. Gestational Development of the Human Immune System. Immunol Allergy Clin North Am 2023; 43:1-15. [PMID: 36410996 DOI: 10.1016/j.iac.2022.05.009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
Building an immune system is a monumental task critical to the survival of the fetus and newborn. A functional fetal immune system must complement the maternal immune system in handling in utero infection; abstain from damaging non-self-reactions that would compromise the materno-fetal interface; mobilize in response to infection and equip mucosal tissues for pathogen exposure at birth. There is growing appreciation that immune cells also have noncanonical roles in development and specifically may contribute to tissue morphogenesis. In this review we detail how hematopoietic and lymphoid organs jointly establish cellular constituents of the immune system; how these constituents are organized in 2 mucosal sites-gut and lung-where early life immune function has long-term consequences for health; and how exemplar diseases of prematurity and inborn errors of immunity reveal dominant pathways in prenatal immunity.
Collapse
Affiliation(s)
- Laura Jardine
- Biosciences Institute, Newcastle University, Faculty of Medical Sciences, Newcastle Upon Tyne NE2 4HH, United Kingdom; Haematology Department, Freeman Hospital, Newcastle Hospitals NHS Foundation Trust, Newcastle Upon Tyne, United Kingdom.
| | - Ina Schim van der Loeff
- Translational and Clinical Research Institute, Newcastle University, Faculty of Medical Sciences, Newcastle Upon Tyne NE2 4HH, United Kingdom
| | - Iram J Haq
- Translational and Clinical Research Institute, Newcastle University, Faculty of Medical Sciences, Newcastle Upon Tyne NE2 4HH, United Kingdom; Department of Paediatric Respiratory Medicine, Great North Children's Hospital, Newcastle Hospitals NHS Foundation Trust, Newcastle Upon Tyne, United Kingdom
| | - Thomas D R Sproat
- Neonatal Unit, Royal Victoria Infirmary, Newcastle Hospitals NHS Foundation Trust, Richardson Road, Newcastle Upon Tyne NE1 4LP, United Kingdom
| |
Collapse
|
6
|
LeBlanc G, Kreissl F, Melamed J, Sobel AL, Constantinides MG. The role of unconventional T cells in maintaining tissue homeostasis. Semin Immunol 2022; 61-64:101656. [PMID: 36306662 PMCID: PMC9828956 DOI: 10.1016/j.smim.2022.101656] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/03/2022] [Revised: 09/01/2022] [Accepted: 09/21/2022] [Indexed: 01/12/2023]
Affiliation(s)
- Gabrielle LeBlanc
- Department of Immunology & Microbiology, Scripps Research, La Jolla, CA 92037, USA,These authors contributed equally
| | - Felix Kreissl
- Department of Immunology & Microbiology, Scripps Research, La Jolla, CA 92037, USA,These authors contributed equally
| | - Jonathan Melamed
- Department of Immunology & Microbiology, Scripps Research, La Jolla, CA 92037, USA,These authors contributed equally
| | - Adam L. Sobel
- Department of Immunology & Microbiology, Scripps Research, La Jolla, CA 92037, USA,These authors contributed equally
| | | |
Collapse
|
7
|
Suo C, Dann E, Goh I, Jardine L, Kleshchevnikov V, Park JE, Botting RA, Stephenson E, Engelbert J, Tuong ZK, Polanski K, Yayon N, Xu C, Suchanek O, Elmentaite R, Domínguez Conde C, He P, Pritchard S, Miah M, Moldovan C, Steemers AS, Mazin P, Prete M, Horsfall D, Marioni JC, Clatworthy MR, Haniffa M, Teichmann SA. Mapping the developing human immune system across organs. Science 2022; 376:eabo0510. [PMID: 35549310 PMCID: PMC7612819 DOI: 10.1126/science.abo0510] [Citation(s) in RCA: 152] [Impact Index Per Article: 50.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Single-cell genomics studies have decoded the immune-cell composition of several human prenatal organs but were limited in understanding the developing immune system as a distributed network across tissues. We profiled nine prenatal tissues combining single-cell RNA sequencing, antigen-receptor sequencing, and spatial transcriptomics to reconstruct the developing human immune system. This revealed the late acquisition of immune effector functions by myeloid and lymphoid cell subsets and the maturation of monocytes and T cells prior to peripheral tissue seeding. Moreover, we uncovered system-wide blood and immune cell development beyond primary hematopoietic organs, characterized human prenatal B1 cells, and shed light on the origin of unconventional T cells. Our atlas provides both valuable data resources and biological insights that will facilitate cell engineering, regenerative medicine, and disease understanding.
Collapse
Affiliation(s)
- Chenqu Suo
- Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton, Cambridge, UK.,Department of Paediatrics, Cambridge University Hospitals, Hills Road, Cambridge, UK
| | - Emma Dann
- Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton, Cambridge, UK
| | - Issac Goh
- Biosciences Institute, Newcastle University, Newcastle upon Tyne, UK
| | - Laura Jardine
- Biosciences Institute, Newcastle University, Newcastle upon Tyne, UK.,Haematology Department, Freeman Hospital, Newcastle upon Tyne Hospitals NHS Foundation Trust, Newcastle upon Tyne, UK
| | | | - Jong-Eun Park
- Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton, Cambridge, UK.,Graduate School of Medical Science and Engineering, Korea Advanced Institute of Science and Technology (KAIST), Daejeon, Korea
| | - Rachel A Botting
- Biosciences Institute, Newcastle University, Newcastle upon Tyne, UK
| | - Emily Stephenson
- Biosciences Institute, Newcastle University, Newcastle upon Tyne, UK
| | - Justin Engelbert
- Biosciences Institute, Newcastle University, Newcastle upon Tyne, UK
| | - Zewen Kelvin Tuong
- Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton, Cambridge, UK.,Molecular Immunity Unit, University of Cambridge Department of Medicine, Cambridge, UK
| | - Krzysztof Polanski
- Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton, Cambridge, UK
| | - Nadav Yayon
- Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton, Cambridge, UK.,European Molecular Biology Laboratory European Bioinformatics Institute, Hinxton, Cambridge, UK
| | - Chuan Xu
- Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton, Cambridge, UK
| | - Ondrej Suchanek
- Molecular Immunity Unit, University of Cambridge Department of Medicine, Cambridge, UK
| | - Rasa Elmentaite
- Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton, Cambridge, UK
| | | | - Peng He
- Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton, Cambridge, UK.,European Molecular Biology Laboratory European Bioinformatics Institute, Hinxton, Cambridge, UK
| | - Sophie Pritchard
- Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton, Cambridge, UK
| | - Mohi Miah
- Biosciences Institute, Newcastle University, Newcastle upon Tyne, UK
| | - Corina Moldovan
- Department of Cellular Pathology, Newcastle upon Tyne Hospitals NHS Foundation Trust, Newcastle upon Tyne, UK
| | | | - Pavel Mazin
- Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton, Cambridge, UK
| | - Martin Prete
- Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton, Cambridge, UK
| | - Dave Horsfall
- Biosciences Institute, Newcastle University, Newcastle upon Tyne, UK
| | - John C Marioni
- Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton, Cambridge, UK.,European Molecular Biology Laboratory European Bioinformatics Institute, Hinxton, Cambridge, UK.,Cancer Research UK Cambridge Institute, Li Ka Shing Centre, University of Cambridge, Cambridge, UK
| | - Menna R Clatworthy
- Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton, Cambridge, UK.,Molecular Immunity Unit, University of Cambridge Department of Medicine, Cambridge, UK
| | - Muzlifah Haniffa
- Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton, Cambridge, UK.,Biosciences Institute, Newcastle University, Newcastle upon Tyne, UK.,Department of Dermatology and NIHR Newcastle Biomedical Research Centre, Newcastle upon Tyne Hospitals NHS Foundation Trust, Newcastle upon Tyne, UK
| | - Sarah A Teichmann
- Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton, Cambridge, UK.,Theory of Condensed Matter, Cavendish Laboratory, Department of Physics, University of Cambridge, Cambridge, UK
| |
Collapse
|
8
|
Homeostatic serum IgE is secreted by plasma cells in the thymus and enhances mast cell survival. Nat Commun 2022; 13:1418. [PMID: 35301301 PMCID: PMC8930980 DOI: 10.1038/s41467-022-29032-x] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2021] [Accepted: 02/23/2022] [Indexed: 12/29/2022] Open
Abstract
Increased serum levels of immunoglobulin E (IgE) is a risk factor for various diseases, including allergy and anaphylaxis. However, the source and ontogeny of B cells producing IgE under steady state conditions are not well defined. Here, we show plasma cells that develop in the thymus and potently secrete IgE and other immunoglobulins, including IgM, IgA, and IgG. The development of these IgE-secreting plasma cells are induced by IL-4 produced by invariant Natural Killer T cells, independent of CD1d-mediated interaction. Single-cell transcriptomics suggest the developmental landscape of thymic B cells, and the thymus supports development of transitional, mature, and memory B cells in addition to plasma cells. Furthermore, thymic plasma cells produce polyclonal antibodies without somatic hypermutation, indicating they develop via the extra-follicular pathway. Physiologically, thymic-derived IgEs increase the number of mast cells in the gut and skin, which correlates with the severity of anaphylaxis. Collectively, we define the ontogeny of thymic plasma cells and show that steady state thymus-derived IgEs regulate mast cell homeostasis, opening up new avenues for studying the genetic causes of allergic disorders. Elevated levels of IgE is associated with a range of allergic pathology but the source of such IgE producing B cells during the steady state is poorly understood. Here, Kwon et al. show that homeostatic IgE is secreted by plasma cells in the thymus and link this to mast cell survival.
Collapse
|
9
|
Krovi SH, Loh L, Spengler A, Brunetti T, Gapin L. Current insights in mouse iNKT and MAIT cell development using single cell transcriptomics data. Semin Immunol 2022; 60:101658. [PMID: 36182863 DOI: 10.1016/j.smim.2022.101658] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/02/2022] [Revised: 08/17/2022] [Accepted: 09/21/2022] [Indexed: 01/15/2023]
Abstract
Innate T (Tinn) cells are a collection of T cells with important regulatory functions that have a crucial role in immunity towards tumors, bacteria, viruses, and in cell-mediated autoimmunity. In mice, the two main αβ Tinn cell subsets include the invariant NKT (iNKT) cells that recognize glycolipid antigens presented by non-polymorphic CD1d molecules and the mucosal associated invariant T (MAIT) cells that recognize vitamin B metabolites presented by the non-polymorphic MR1 molecules. Due to their ability to promptly secrete large quantities of cytokines either after T cell antigen receptor (TCR) activation or upon exposure to tissue- and antigen-presenting cell-derived cytokines, Tinn cells are thought to act as a bridge between the innate and adaptive immune systems and have the ability to shape the overall immune response. Their swift response reflects the early acquisition of helper effector programs during their development in the thymus, independently of pathogen exposure and prior to taking up residence in peripheral tissues. Several studies recently profiled, in an unbiased manner, the transcriptomes of mouse thymic iNKT and MAIT cells at the single cell level. Based on these data, we re-examine in this review how Tinn cells develop in the mouse thymus and undergo effector differentiation.
Collapse
Affiliation(s)
| | - Liyen Loh
- University of Colorado Anschutz Medical Campus, Aurora, USA
| | | | - Tonya Brunetti
- University of Colorado Anschutz Medical Campus, Aurora, USA
| | - Laurent Gapin
- University of Colorado Anschutz Medical Campus, Aurora, USA.
| |
Collapse
|
10
|
Feyaerts D, Urbschat C, Gaudillière B, Stelzer IA. Establishment of tissue-resident immune populations in the fetus. Semin Immunopathol 2022; 44:747-766. [PMID: 35508672 PMCID: PMC9067556 DOI: 10.1007/s00281-022-00931-x] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2022] [Accepted: 03/17/2022] [Indexed: 12/15/2022]
Abstract
The immune system establishes during the prenatal period from distinct waves of stem and progenitor cells and continuously adapts to the needs and challenges of early postnatal and adult life. Fetal immune development not only lays the foundation for postnatal immunity but establishes functional populations of tissue-resident immune cells that are instrumental for fetal immune responses amidst organ growth and maturation. This review aims to discuss current knowledge about the development and function of tissue-resident immune populations during fetal life, focusing on the brain, lung, and gastrointestinal tract as sites with distinct developmental trajectories. While recent progress using system-level approaches has shed light on the fetal immune landscape, further work is required to describe precise roles of prenatal immune populations and their migration and adaptation to respective organ environments. Defining points of prenatal susceptibility to environmental challenges will support the search for potential therapeutic targets to positively impact postnatal health.
Collapse
Affiliation(s)
- Dorien Feyaerts
- grid.168010.e0000000419368956Department of Anesthesiology, Perioperative and Pain Medicine, Stanford University School of Medicine, Palo Alto, CA USA
| | - Christopher Urbschat
- grid.13648.380000 0001 2180 3484Division of Experimental Feto-Maternal Medicine, Department of Obstetrics and Fetal Medicine, University Medical Center Hamburg, Hamburg, Germany
| | - Brice Gaudillière
- grid.168010.e0000000419368956Department of Anesthesiology, Perioperative and Pain Medicine, Stanford University School of Medicine, Palo Alto, CA USA ,grid.168010.e0000000419368956Department of Pediatrics, Stanford University School of Medicine, Palo Alto, CA USA
| | - Ina A. Stelzer
- grid.168010.e0000000419368956Department of Anesthesiology, Perioperative and Pain Medicine, Stanford University School of Medicine, Palo Alto, CA USA
| |
Collapse
|
11
|
Dong M, Mallet Gauthier È, Fournier M, Melichar HJ. Developing the right tools for the job: Lin28 regulation of early life T-cell development and function. FEBS J 2021; 289:4416-4429. [PMID: 34077615 DOI: 10.1111/febs.16045] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2020] [Revised: 04/29/2021] [Accepted: 06/01/2021] [Indexed: 12/14/2022]
Abstract
T cells comprise a functionally heterogeneous cell population that has important roles in the immune system. While T cells are broadly considered to be a component of the antigen-specific adaptive immune response, certain T-cell subsets display innate-like effector characteristics whereas others perform immunosuppressive functions. These functionally diverse T-cell populations preferentially arise at different stages of ontogeny and are tailored to the immunological priorities of the organism over time. Many differences in early life versus adult T-cell phenotypes can be attributed to the cell-intrinsic properties of the distinct progenitors that seed the thymus throughout development. It is becoming clear that Lin28, an evolutionarily conserved, heterochronic RNA-binding protein that is differentially expressed among early life and adult hematopoietic progenitor cells, plays a substantial role in influencing early T-cell development and function. Here, we discuss the mechanisms by which Lin28 shapes the T-cell landscape to protect the developing fetus and newborn. Manipulation of the Lin28 gene regulatory network is being considered as one means of improving hematopoietic stem cell transplant outcomes; as such, understanding the impact of Lin28 on T-cell function is of clinical relevance.
Collapse
Affiliation(s)
- Mengqi Dong
- Immunology-Oncology Unit, Maisonneuve-Rosemont Hospital Research Center, Montréal, QC, Canada.,Département de microbiologie, infectiologie et immunologie, Université de Montréal, Montréal, QC, Canada
| | - Ève Mallet Gauthier
- Immunology-Oncology Unit, Maisonneuve-Rosemont Hospital Research Center, Montréal, QC, Canada.,Département de microbiologie, infectiologie et immunologie, Université de Montréal, Montréal, QC, Canada
| | - Marilaine Fournier
- Immunology-Oncology Unit, Maisonneuve-Rosemont Hospital Research Center, Montréal, QC, Canada
| | - Heather J Melichar
- Immunology-Oncology Unit, Maisonneuve-Rosemont Hospital Research Center, Montréal, QC, Canada.,Département de médecine, Université de Montréal, Montréal, QC, Canada
| |
Collapse
|
12
|
Classical MHC expression by DP thymocytes impairs the selection of non-classical MHC restricted innate-like T cells. Nat Commun 2021; 12:2308. [PMID: 33863906 PMCID: PMC8052364 DOI: 10.1038/s41467-021-22589-z] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2020] [Accepted: 03/10/2021] [Indexed: 02/02/2023] Open
Abstract
Conventional T cells are selected by peptide-MHC expressed by cortical epithelial cells in the thymus, and not by cortical thymocytes themselves that do not express MHC I or MHC II. Instead, cortical thymocytes express non-peptide presenting MHC molecules like CD1d and MR1, and promote the selection of PLZF+ iNKT and MAIT cells, respectively. Here, we report an inducible class-I transactivator mouse that enables the expression of peptide presenting MHC I molecules in different cell types. We show that MHC I expression in DP thymocytes leads to expansion of peptide specific PLZF+ innate-like (PIL) T cells. Akin to iNKT cells, PIL T cells differentiate into three functional effector subsets in the thymus, and are dependent on SAP signaling. We demonstrate that PIL and NKT cells compete for a narrow niche, suggesting that the absence of peptide-MHC on DP thymocytes facilitates selection of non-peptide specific lymphocytes.
Collapse
|
13
|
Bhalla P, Wysocki CA, van Oers NSC. Molecular Insights Into the Causes of Human Thymic Hypoplasia With Animal Models. Front Immunol 2020; 11:830. [PMID: 32431714 PMCID: PMC7214791 DOI: 10.3389/fimmu.2020.00830] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2020] [Accepted: 04/14/2020] [Indexed: 12/30/2022] Open
Abstract
22q11.2 deletion syndrome (DiGeorge), CHARGE syndrome, Nude/SCID and otofaciocervical syndrome type 2 (OTFCS2) are distinct clinical conditions in humans that can result in hypoplasia and occasionally, aplasia of the thymus. Thymic hypoplasia/aplasia is first suggested by absence or significantly reduced numbers of recent thymic emigrants, revealed in standard-of-care newborn screens for T cell receptor excision circles (TRECs). Subsequent clinical assessments will often indicate whether genetic mutations are causal to the low T cell output from the thymus. However, the molecular mechanisms leading to the thymic hypoplasia/aplasia in diverse human syndromes are not fully understood, partly because the problems of the thymus originate during embryogenesis. Rodent and Zebrafish models of these clinical syndromes have been used to better define the underlying basis of the clinical presentations. Results from these animal models are uncovering contributions of different cell types in the specification, differentiation, and expansion of the thymus. Cell populations such as epithelial cells, mesenchymal cells, endothelial cells, and thymocytes are variably affected depending on the human syndrome responsible for the thymic hypoplasia. In the current review, findings from the diverse animal models will be described in relation to the clinical phenotypes. Importantly, these results are suggesting new strategies for regenerating thymic tissue in patients with distinct congenital disorders.
Collapse
Affiliation(s)
- Pratibha Bhalla
- Department of Immunology, The University of Texas Southwestern Medical Center, Dallas, TX, United States
| | - Christian A. Wysocki
- Department of Pediatrics, The University of Texas Southwestern Medical Center, Dallas, TX, United States
| | - Nicolai S. C. van Oers
- Department of Immunology, 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
- Department of Microbiology, The University of Texas Southwestern Medical Center, Dallas, TX, United States
| |
Collapse
|
14
|
Variants of innate CD8 + T cells are associated with Grip2 and Klf15 genes. Cell Mol Immunol 2020; 17:1007-1009. [PMID: 31919398 PMCID: PMC7608311 DOI: 10.1038/s41423-019-0357-3] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2019] [Accepted: 12/17/2019] [Indexed: 11/13/2022] Open
|
15
|
Halkias J, Rackaityte E, Hillman SL, Aran D, Mendoza VF, Marshall LR, MacKenzie TC, Burt TD. CD161 contributes to prenatal immune suppression of IFNγ-producing PLZF+ T cells. J Clin Invest 2019; 129:3562-3577. [PMID: 31145102 DOI: 10.1172/jci125957] [Citation(s) in RCA: 45] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
Abstract
BACKGROUND While the human fetal immune system defaults to a program of tolerance, there is concurrent need for protective immunity to meet the antigenic challenges encountered after birth. Activation of T cells in utero is associated with the fetal inflammatory response with broad implications for the health of the fetus and of the pregnancy. However, the characteristics of the fetal effector T cells that contribute to this process are largely unknown. METHODS We analyzed primary human fetal lymphoid and mucosal tissues and performed phenotypic, functional, and transcriptional analysis to identify T cells with pro-inflammatory potential. The frequency and function of fetal-specific effector T cells was assessed in the cord blood of infants with localized and systemic inflammatory pathologies and compared to healthy term controls. RESULTS We identified a transcriptionally distinct population of CD4+ T cells characterized by expression of the transcription factor Promyelocytic Leukemia Zinc Finger (PLZF). PLZF+ CD4+ T cells were specifically enriched in the fetal intestine, possessed an effector memory phenotype, and rapidly produced pro-inflammatory cytokines. Engagement of the C-type lectin CD161 on these cells inhibited TCR-dependent production of IFNγ in a fetal-specific manner. IFNγ-producing PLZF+ CD4+ T cells were enriched in the cord blood of infants with gastroschisis, a natural model of chronic inflammation originating from the intestine, as well as in preterm birth, suggesting these cells contribute to fetal systemic immune activation. CONCLUSION Our work reveals a fetal-specific program of protective immunity whose dysregulation is associated with fetal and neonatal inflammatory pathologies.
Collapse
Affiliation(s)
| | - Elze Rackaityte
- Biomedical Sciences Program, UCSF, San Francisco, California, USA
| | - Sara L Hillman
- Maternal and Fetal Medicine Department, Institute for Women's Health, University College London, London, United Kingdom
| | - Dvir Aran
- Institute for Computational Health Sciences, UCSF, San Francisco, California, USA
| | - Ventura F Mendoza
- Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, UCSF, San Francisco, California, USA
| | - Lucy R Marshall
- Division of Infection Immunity and Inflammation, University College London Great Ormond Street Institute of Child Health, London, United Kingdom
| | - Tippi C MacKenzie
- Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, UCSF, San Francisco, California, USA.,Department of Surgery, UCSF, San Francisco, California, USA
| | - Trevor D Burt
- Division of Neonatology, Department of Pediatrics, and.,Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, UCSF, San Francisco, California, USA
| |
Collapse
|
16
|
Dashtsoodol N, Bortoluzzi S, Schmidt-Supprian M. T Cell Receptor Expression Timing and Signal Strength in the Functional Differentiation of Invariant Natural Killer T Cells. Front Immunol 2019; 10:841. [PMID: 31080448 PMCID: PMC6497757 DOI: 10.3389/fimmu.2019.00841] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2019] [Accepted: 04/01/2019] [Indexed: 12/19/2022] Open
Abstract
The CD1d-restricted Vα14 invariant NKT (iNKT) cell lineage in mice (Vα24 in humans) represents an evolutionary conserved innate-like immune cell type that recognizes glycolipid antigens. Because of their unique ability to promptly secrete copious amounts of both pro-inflammatory and anti-inflammatory cytokines, typically produced by different T helper cell types, iNKT cells are implicated in the regulation of various pathologic conditions such as infection, allergy, autoimmune disease, maintenance of transplantation tolerance, and cancer. This striking multifaceted role in immune regulation is correlated with the presence of multiple functionally distinct iNKT cell subsets that can be distinguished based on the expression of characteristic surface markers and transcription factors. However, to date it, remains largely unresolved how this puzzling diversity of iNKT cell functional subsets emerges and what factors dictate the type of effector cell differentiation during the thymic differentiation considering the mono-specific nature of their T cell receptor (TCR) and their selecting molecule CD1d. Here, we summarize recent findings focusing on the role of TCR-mediated signaling and discuss possible mechanisms that may influence the sub-lineage choice of iNKT cells.
Collapse
Affiliation(s)
- Nyambayar Dashtsoodol
- Department of Hematology and Medical Oncology, Klinikum rechts der Isar and TranslaTUM Cancer Center, Technische Universität München, München, Germany.,Department of Microbiology and Immunology, School of Biomedicine, Mongolian National University of Medical Sciences, Ulaanbaatar, Mongolia
| | - Sabrina Bortoluzzi
- Department of Hematology and Medical Oncology, Klinikum rechts der Isar and TranslaTUM Cancer Center, Technische Universität München, München, Germany
| | - Marc Schmidt-Supprian
- Department of Hematology and Medical Oncology, Klinikum rechts der Isar and TranslaTUM Cancer Center, Technische Universität München, München, Germany
| |
Collapse
|
17
|
White AJ, Lucas B, Jenkinson WE, Anderson G. Invariant NKT Cells and Control of the Thymus Medulla. THE JOURNAL OF IMMUNOLOGY 2019; 200:3333-3339. [PMID: 29735644 DOI: 10.4049/jimmunol.1800120] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/26/2018] [Accepted: 02/20/2018] [Indexed: 12/29/2022]
Abstract
Most αβ T cells that form in the thymus are generated during mainstream conventional thymocyte development and involve the generation and selection of a diverse αβ TCR repertoire that recognizes self-peptide/MHC complexes. Additionally, the thymus also supports the production of T cell subsets that express αβ TCRs but display unique developmental and functional features distinct from conventional αβ T cells. These include multiple lineages of CD1d-restricted invariant NKT (iNKT) cells that express an invariant αβ TCR, branch off from mainstream thymocytes at the CD4+CD8+ stage, and are potent producers of polarizing cytokines. Importantly, and despite their differences, iNKT cells and conventional αβ T cells share common requirements for thymic epithelial microenvironments during their development. Moreover, emerging evidence suggests that constitutive cytokine production by iNKT cells influences both conventional thymocyte development and the intrathymic formation of additional innate CD8+ αβ T cells with memory-like properties. In this article, we review evidence for an intrathymic innate lymphocyte network in which iNKT cells play key roles in multiple aspects of thymus function.
Collapse
Affiliation(s)
- Andrea J White
- Institute of Immunology and Immunotherapy, College of Medical and Dental Sciences, Medical School, University of Birmingham, Birmingham B15 2TT, United Kingdom
| | - Beth Lucas
- Institute of Immunology and Immunotherapy, College of Medical and Dental Sciences, Medical School, University of Birmingham, Birmingham B15 2TT, United Kingdom
| | - William E Jenkinson
- Institute of Immunology and Immunotherapy, College of Medical and Dental Sciences, Medical School, University of Birmingham, Birmingham B15 2TT, United Kingdom
| | - Graham Anderson
- Institute of Immunology and Immunotherapy, College of Medical and Dental Sciences, Medical School, University of Birmingham, Birmingham B15 2TT, United Kingdom
| |
Collapse
|
18
|
Kim YH, Zhu L, Pyaram K, Lopez C, Ohye RG, Garcia JV, Green GE, Chang CH. PLZF-expressing CD4 T cells show the characteristics of terminally differentiated effector memory CD4 T cells in humans. Eur J Immunol 2018; 48:1255-1257. [PMID: 29572809 PMCID: PMC6748641 DOI: 10.1002/eji.201747426] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2017] [Revised: 02/12/2018] [Accepted: 03/14/2018] [Indexed: 01/31/2023]
Abstract
We show the presence of lymphoid tissue-resident PLZF+ CD45RA+ RO+ CD4 T cells in humans. They express HLA-DR, granzyme B, and perforin and are low on CCR7 like terminally differentiated effector memory (Temra) cells and are likely generated from effector T cells (Te) or from central (Tcm) or effector (Tem) memory T (Tcm) cells during immune responses. Tn, Naïve T cells.
Collapse
Affiliation(s)
- Yeung-Hyen Kim
- Department of Microbiology and Immunology, University of Michigan Medical School, Ann Arbor, MI, USA
- Department of New Business Development, Medytox R&D center, Medytox, Suwon, Korea
| | - Lingqiao Zhu
- Department of Microbiology and Immunology, University of Michigan Medical School, Ann Arbor, MI, USA
- Shanghai Mabgeek Biotech, Shanghai, P. R. China
| | - Kalyani Pyaram
- Department of Microbiology and Immunology, University of Michigan Medical School, Ann Arbor, MI, USA
| | - Cesar Lopez
- Division of Infectious, Disease, University of North Carolina at Chapel Hill, School of Medicine, Chapel Hill, NC, USA
| | - Richard G. Ohye
- Department of Cardiac Surgery, University of Michigan Medical School, Ann Arbor, MI, USA
| | - J Victor Garcia
- Division of Infectious, Disease, University of North Carolina at Chapel Hill, School of Medicine, Chapel Hill, NC, USA
| | - Glenn E. Green
- Department of Otolaryngology-Head and Neck Surgery, University of Michigan Medical School, Ann Arbor, MI, USA
| | - Cheong-Hee Chang
- Department of Microbiology and Immunology, University of Michigan Medical School, Ann Arbor, MI, USA
| |
Collapse
|
19
|
Foreign antigen-independent memory-phenotype CD4 + T cells: a new player in innate immunity? Nat Rev Immunol 2018; 18:1. [PMID: 29480288 DOI: 10.1038/nri.2018.13] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
|
20
|
Kwon DI, Lee YJ. Lineage Differentiation Program of Invariant Natural Killer T Cells. Immune Netw 2017; 17:365-377. [PMID: 29302250 PMCID: PMC5746607 DOI: 10.4110/in.2017.17.6.365] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2017] [Revised: 10/27/2017] [Accepted: 11/02/2017] [Indexed: 02/07/2023] Open
Abstract
Invariant natural killer T (iNKT) cells are innate T cells restricted by CD1d molecules. They are positively selected in the thymic cortex and migrate to the medullary area, in which they differentiate into 3 different lineages. Promyelocytic leukemia zinc finger (PLZF) modulates this process, and PLZFhigh, PLZFintermediate, and PLZFlow iNKT cells are designated as NKT2, NKT17, and NKT1 cells, respectively. Analogous to conventional helper CD4 T cells, each subset expresses distinct combinations of transcription factors and produces different cytokines. In lymphoid organs, iNKT subsets have unique localizations, which determine their cytokine responses upon antigenic challenge. The lineage differentiation programs of iNKT cells are differentially regulated in various mice strains in a cell-intrinsic manner, and BALB/c mice contain a high frequency of NKT2 cells. In the thymic medulla, steady state IL-4 from NKT2 cells directly conditions CD8 T cells to become memory-like cells expressing Eomesodermin, which function as premade memory effectors. The genetic signature of iNKT cells is more similar to that of γδ T cells and innate lymphoid cells (ILCs) than of conventional helper T cells, suggesting that ILCs and innate T cells share common developmental programs.
Collapse
Affiliation(s)
- Dong-Il Kwon
- Academy of Immunology and Microbiology, Institute for Basic Science, Pohang 37673, Korea.,Division of Integrative Biosciences and Biotechnology, Pohang University of Science and Technology, Pohang 37673, Korea
| | - You Jeong Lee
- Academy of Immunology and Microbiology, Institute for Basic Science, Pohang 37673, Korea.,Division of Integrative Biosciences and Biotechnology, Pohang University of Science and Technology, Pohang 37673, Korea
| |
Collapse
|
21
|
Breed ER, Lee ST, Hogquist KA. Directing T cell fate: How thymic antigen presenting cells coordinate thymocyte selection. Semin Cell Dev Biol 2017; 84:2-10. [PMID: 28800929 DOI: 10.1016/j.semcdb.2017.07.045] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2017] [Revised: 06/30/2017] [Accepted: 07/30/2017] [Indexed: 01/02/2023]
Abstract
The development of a self-tolerant and effective T cell receptor repertoire is dependent on interactions coordinated by various antigen presenting cells (APC) within the thymus. T cell receptor-self-peptide-MHC interactions are essential for determining T cell fate, however different cytokine and co-stimulatory signals provided by the diverse APCs within the thymus are also critical. Here, we outline the different localization and functional capabilities of thymic APCs. We also discuss how these distinct APCs work collectively to facilitate the establishment of a diverse T cell receptor repertoire that is tolerant to an array of different self-antigens.
Collapse
Affiliation(s)
- Elise R Breed
- The Center for Immunology, Department of Laboratory Medicine & Pathology, University of Minnesota, Minneapolis, MN, USA
| | - S Thera Lee
- The Center for Immunology, Department of Laboratory Medicine & Pathology, University of Minnesota, Minneapolis, MN, USA
| | - Kristin A Hogquist
- The Center for Immunology, Department of Laboratory Medicine & Pathology, University of Minnesota, Minneapolis, MN, USA.
| |
Collapse
|
22
|
White JT, Cross EW, Kedl RM. Antigen-inexperienced memory CD8 + T cells: where they come from and why we need them. Nat Rev Immunol 2017; 17:391-400. [PMID: 28480897 DOI: 10.1038/nri.2017.34] [Citation(s) in RCA: 116] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Memory-phenotype CD8+ T cells exist in substantial numbers within hosts that have not been exposed to either foreign antigen or overt lymphopenia. These antigen-inexperienced memory-phenotype T cells can be divided into two major subsets: 'innate memory' T cells and 'virtual memory' T cells. Although these two subsets are nearly indistinguishable by surface markers alone, notable developmental and functional differences exist between the two subsets, which suggests that they represent distinct populations. In this Opinion article, we review the available literature on each subset, highlighting the key differences between these populations. Furthermore, we suggest a unifying model for the categorization of antigen-inexperienced memory-phenotype CD8+ T cells.
Collapse
Affiliation(s)
- Jason T White
- Department of Microbiology and Immunology, The Peter Doherty Institute, University of Melbourne, 792 Elizabeth Street, Melbourne, Victoria 3000, Australia
| | - Eric W Cross
- Department of Immunology and Microbiology, University of Colorado Denver at Anschutz Medical Campus, School of Medicine, Mail Stop 8333, Room P18-8115, 12800 East 19th Avenue, Aurora, Colorado 80045-2537, USA
| | - Ross M Kedl
- Department of Immunology and Microbiology, University of Colorado Denver at Anschutz Medical Campus, School of Medicine, Mail Stop 8333, Room P18-8115, 12800 East 19th Avenue, Aurora, Colorado 80045-2537, USA
| |
Collapse
|
23
|
Kang BH, Park HJ, Park HJ, Lee JII, Park SH, Jung KC. PLZF(+) Innate T Cells Support the TGF-β-Dependent Generation of Activated/Memory-Like Regulatory T Cells. Mol Cells 2016; 39:468-76. [PMID: 27101876 PMCID: PMC4916398 DOI: 10.14348/molcells.2016.0004] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2016] [Revised: 03/28/2016] [Accepted: 03/31/2016] [Indexed: 11/27/2022] Open
Abstract
PLZF-expressing invariant natural killer T cells and CD4 T cells are unique subsets of innate T cells. Both are selected via thymocyte-thymocyte interaction, and they contribute to the generation of activated/memory-like CD4 and CD8 T cells in the thymus via the production of IL-4. Here, we investigated whether PLZF(+) innate T cells also affect the development and function of Foxp3(+) regulatory CD4 T cells. Flow cytometry analysis of the thymus and spleen from both CIITA transgenic C57BL/6 and wild-type BALB/c mice, which have abundant PLZF(+) CD4 T cells and invariant natural killer T cells, respectively, revealed that Foxp3(+) T cells in these mice exhibited a CD103(+) activated/memory-like phenotype. The frequency of CD103(+) regulatory T cells was considerably decreased in PLZF(+) cell-deficient CIITA(Tg)Plzf(lu/lu) and BALB/c.CD1d(-/-) mice as well as in an IL-4-deficient background, such as in CIITA(Tg)IL-4(-/-) and BALB/c.lL-4(-/-) mice, indicating that the acquisition of an activated/memory-like phenotype was dependent on PLZF(+) innate T cells and IL-4. Using fetal thymic organ culture, we further demonstrated that IL-4 in concert with TGF-β enhanced the acquisition of the activated/memory-like phenotype of regulatory T cells. In functional aspects, the activated/memory-like phenotype of Treg cells was directly related to their suppressive function; regulatory T cells of CIITA(Tg)PIV(-/-) mice more efficiently suppressed ovalbumin-induced allergic airway inflammation compared with their counterparts from wild-type mice. All of these findings suggest that PLZF(+) innate T cells also augmented the generation of activated/memory-like regulation via IL-4 production.
Collapse
Affiliation(s)
- Byung Hyun Kang
- Postgraduate Course of Translational Medicine, Medical Research Center, Seoul National University College of Medicine, Seoul 03080,
Korea
| | - Hyo Jin Park
- Department of Pathology, Medical Research Center, Seoul National University College of Medicine, Seoul 03080,
Korea
- Department of Pathology, Seoul National University Bundang Hospital, Sungnam 13620,
Korea
| | - Hi Jung Park
- Postgraduate Course of Translational Medicine, Medical Research Center, Seoul National University College of Medicine, Seoul 03080,
Korea
| | - Jae-II Lee
- Postgraduate Course of Translational Medicine, Medical Research Center, Seoul National University College of Medicine, Seoul 03080,
Korea
- Transplantation Research Institute, Medical Research Center, Seoul National University College of Medicine, Seoul 03080,
Korea
| | - Seong Hoe Park
- Postgraduate Course of Translational Medicine, Medical Research Center, Seoul National University College of Medicine, Seoul 03080,
Korea
- Transplantation Research Institute, Medical Research Center, Seoul National University College of Medicine, Seoul 03080,
Korea
| | - Kyeong Cheon Jung
- Postgraduate Course of Translational Medicine, Medical Research Center, Seoul National University College of Medicine, Seoul 03080,
Korea
- Department of Pathology, Medical Research Center, Seoul National University College of Medicine, Seoul 03080,
Korea
- Transplantation Research Institute, Medical Research Center, Seoul National University College of Medicine, Seoul 03080,
Korea
- Department of Pathology, Seoul National University Hospital, Seoul 03080,
Korea
| |
Collapse
|
24
|
Affiliation(s)
- Seong Hoe Park
- Transplantation Research Institute, Medical Research Center, Seoul National University College of Medicine, Seoul, Korea
| |
Collapse
|
25
|
Development of a diverse human T-cell repertoire despite stringent restriction of hematopoietic clonality in the thymus. Proc Natl Acad Sci U S A 2015; 112:E6020-7. [PMID: 26483497 DOI: 10.1073/pnas.1519118112] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023] Open
Abstract
The fate and numbers of hematopoietic stem cells (HSC) and their progeny that seed the thymus constitute a fundamental question with important clinical implications. HSC transplantation is often complicated by limited T-cell reconstitution, especially when HSC from umbilical cord blood are used. Attempts to improve immune reconstitution have until now been unsuccessful, underscoring the need for better insight into thymic reconstitution. Here we made use of the NOD-SCID-IL-2Rγ(-/-) xenograft model and lentiviral cellular barcoding of human HSCs to study T-cell development in the thymus at a clonal level. Barcoded HSCs showed robust (>80% human chimerism) and reproducible myeloid and lymphoid engraftment, with T cells arising 12 wk after transplantation. A very limited number of HSC clones (<10) repopulated the xenografted thymus, with further restriction of the number of clones during subsequent development. Nevertheless, T-cell receptor rearrangements were polyclonal and showed a diverse repertoire, demonstrating that a multitude of T-lymphocyte clones can develop from a single HSC clone. Our data imply that intrathymic clonal fitness is important during T-cell development. As a consequence, immune incompetence after HSC transplantation is not related to the transplantation of limited numbers of HSC but to intrathymic events.
Collapse
|
26
|
Lee A, Park SP, Park CH, Kang BH, Park SH, Ha SJ, Jung KC. IL-4 Induced Innate CD8+ T Cells Control Persistent Viral Infection. PLoS Pathog 2015; 11:e1005193. [PMID: 26452143 PMCID: PMC4599894 DOI: 10.1371/journal.ppat.1005193] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2015] [Accepted: 09/06/2015] [Indexed: 12/20/2022] Open
Abstract
Memory-like CD8+ T cells expressing eomesodermin are a subset of innate T cells initially identified in a number of genetically modified mice, and also exist in wild mice and human. The acquisition of memory phenotype and function by these T cells is dependent on IL–4 produced by PLZF+ innate T cells; however, their physiologic function is still not known. Here we found that these IL-4-induced innate CD8+ T cells are critical for accelerating the control of chronic virus infection. In CIITA-transgenic mice, which have a substantial population of IL-4-induced innate CD8+ T cells, this population facilitated rapid control of viremia and induction of functional anti-viral T-cell responses during infection with chronic form of lymphocytic choriomeningitis virus. Characteristically, anti-viral innate CD8+ T cells accumulated sufficiently during early phase of infection. They produced a robust amount of IFN-γ and TNF-α with enhanced expression of a degranulation marker. Furthermore, this finding was confirmed in wild-type mice. Taken together, the results from our study show that innate CD8+ T cells works as an early defense mechanism against chronic viral infection. Over the course of viral infection there may be a limited time period during which the host system can eliminate the virus. When viruses are not eliminated within this period of time, virus can establish persistent infection. Here, we show that IL-4-induced innate CD8+ T cells are able to effectively control chronic virus infection. Innate T cells are heterogeneous population of T cells that acquire effector/memory phenotype as a result of their maturation process in thymus, unlike conventional T cells that differentiate into memory cells after antigen encounter in periphery. Previous data suggest that innate T cells might serve as a first-line of defense against certain bacterial pathogens. IL-4-induced innate CD8+ T cells are a unique subset of innate T cells that were recently identified in both mouse and human. We found that IL-4-induced innate CD8+ T cells immediately accumulated after viral infection and produced a robust amount of effector cytokines. Thereby, IL-4-induced innate CD8+ T cells provide an effective barrier to the establishment of persistent infection via effective virus control during the early phase of viral infection. Collectively our data show that IL-4-induced innate CD8+ T cells works as an early defense mechanism against chronic viral infection.
Collapse
Affiliation(s)
- Ara Lee
- Department of Biochemistry, College of Life Science & Biotechnology, Yonsei University, Seoul, Korea
| | - Seung Pyo Park
- Transplantation Research Institute, Medical Research Center, Seoul National University College of Medicine, Seoul, Korea
| | - Chan Hee Park
- Department of Biochemistry, College of Life Science & Biotechnology, Yonsei University, Seoul, Korea
| | - Byung Hyun Kang
- Graduate School of Translational Medicine, Seoul National University College of Medicine, Seoul, Korea
| | - Seong Hoe Park
- Transplantation Research Institute, Medical Research Center, Seoul National University College of Medicine, Seoul, Korea
- Graduate School of Translational Medicine, Seoul National University College of Medicine, Seoul, Korea
| | - Sang-Jun Ha
- Department of Biochemistry, College of Life Science & Biotechnology, Yonsei University, Seoul, Korea
- * E-mail: (SJH); (KCJ)
| | - Kyeong Cheon Jung
- Transplantation Research Institute, Medical Research Center, Seoul National University College of Medicine, Seoul, Korea
- Graduate School of Translational Medicine, Seoul National University College of Medicine, Seoul, Korea
- Department of Pathology, Seoul National University College of Medicine, Seoul, Korea
- * E-mail: (SJH); (KCJ)
| |
Collapse
|
27
|
Zhao HB, Zhang XY, Feng GQ, Guo MM, Chang P, Qi C, Zhong XP, Zhou QC, Wang JL. Expression of plzfa in embryo and adult of medaka Oryzias latipes. JOURNAL OF FISH BIOLOGY 2015; 87:231-240. [PMID: 26077174 DOI: 10.1111/jfb.12713] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/14/2014] [Accepted: 04/22/2015] [Indexed: 06/04/2023]
Abstract
In this study, a homologous gene named plzfa was identified and characterized in medaka Oryzias latipes. Oryzias latipes plzfa was detected in all the tissues including brain, gill, muscle, liver, intestine, kidney, spleen, testis and ovary using reverse transcriptase (RT)-PCR. plzfa was detected in the oocytes of the ovary and in the spermatogonia and somitic cells of the testis by in situ hybridization. plzfa had a maternal origin with continuous and dynamic expression during embryonic development. plzfa was observed in the brain, neural rod and sensor organs including the eyes, ears and nose during embryogenesis. plzfa was also detected in the neural crest, somite, pectoral fin, intestine and skin. These results indicate that plzfa is a pleiotropic gene that may play major roles in various tissues.
Collapse
Affiliation(s)
- H B Zhao
- Hubei Key Laboratory of Genetic Regulation and Integrative Biology, School of Life Sciences, Central China Normal University, Wuhan, 430079, China
| | - X Y Zhang
- Hubei Key Laboratory of Genetic Regulation and Integrative Biology, School of Life Sciences, Central China Normal University, Wuhan, 430079, China
| | - G Q Feng
- Hubei Key Laboratory of Genetic Regulation and Integrative Biology, School of Life Sciences, Central China Normal University, Wuhan, 430079, China
| | - M M Guo
- Hubei Key Laboratory of Genetic Regulation and Integrative Biology, School of Life Sciences, Central China Normal University, Wuhan, 430079, China
| | - P Chang
- Hubei Key Laboratory of Genetic Regulation and Integrative Biology, School of Life Sciences, Central China Normal University, Wuhan, 430079, China
| | - C Qi
- Hubei Key Laboratory of Genetic Regulation and Integrative Biology, School of Life Sciences, Central China Normal University, Wuhan, 430079, China
| | - X P Zhong
- Hubei Key Laboratory of Genetic Regulation and Integrative Biology, School of Life Sciences, Central China Normal University, Wuhan, 430079, China
| | - Q C Zhou
- Hubei Key Laboratory of Genetic Regulation and Integrative Biology, School of Life Sciences, Central China Normal University, Wuhan, 430079, China
| | - J L Wang
- Hubei Key Laboratory of Genetic Regulation and Integrative Biology, School of Life Sciences, Central China Normal University, Wuhan, 430079, China
| |
Collapse
|
28
|
Van Kaer L. Innate and virtual memory T cells in man. Eur J Immunol 2015; 45:1916-20. [PMID: 26013879 DOI: 10.1002/eji.201545761] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2015] [Revised: 05/13/2015] [Accepted: 05/21/2015] [Indexed: 12/11/2022]
Abstract
A hallmark of the antigen-specific B and T lymphocytes of the adaptive immune system is their capacity to "remember" pathogens long after they are first encountered, a property that forms the basis for effective vaccine development. However, studies in mice have provided strong evidence that some naive T cells can develop characteristics of memory T cells in the absence of foreign antigen encounters. Such innate memory T cells may develop in response to lymphopenia or the presence of high levels of the cytokine IL-4, and have also been identified in unmanipulated animals, a phenomenal referred to as "virtual memory." While the presence of innate memory T cells in mice is now widely accepted, their presence in humans has not yet been fully validated. In this issue of the European Journal of Immunology, Jacomet et al. [Eur. J. Immunol. 2015. 45:1926-1933] provide the best evidence to date for innate memory T cells in humans. These findings may contribute significantly to our understanding of human immunity to microbial pathogens and tumors.
Collapse
Affiliation(s)
- Luc Van Kaer
- Department of Pathology, Microbiology and Immunology, Vanderbilt University School of Medicine, Nashville, TN, USA
| |
Collapse
|
29
|
Kang BH, Park HJ, Yum HI, Park SP, Park JK, Kang EH, Lee JI, Lee EB, Park CG, Jung KC, Park SH. Thymic low affinity/avidity interaction selects natural Th1 cells. THE JOURNAL OF IMMUNOLOGY 2015; 194:5861-71. [PMID: 25972479 DOI: 10.4049/jimmunol.1401628] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/26/2014] [Accepted: 04/15/2015] [Indexed: 12/13/2022]
Abstract
Identification of intrathymic eomesodermin(+) (Eomes(+)) CD4 T cells creates a novel idea that there is more than one way for the generation of innate CD4 T cells. Promyelocytic leukemia zinc finger protein(+) T cells and natural Th17 cells are known to be generated by sensing a high and persistent TCR strength, whereas this is not the case for Eomes(+) CD4 T cells. These cells go through low-level signal during the entire maturation pathway, which subsequently leads to induction of high susceptibility to cytokine IL-4. This event seems to be a major determinant for the generation of this type of cell. These T cells are functionally equivalent to Th1 cells that are present in the periphery, and this event takes place both in transgenic and in wild-type mice. There is additional evidence that this type of Eomes(+) innate CD4 T cell is also present in human cord blood.
Collapse
Affiliation(s)
- Byung Hyun Kang
- Graduate School of Immunology, Seoul National University College of Medicine, Seoul 110-799, Korea
| | - Hyo Jin Park
- Department of Pathology, Seoul National University College of Medicine, Seoul 110-799, Korea; Department of Pathology, Seoul National University Bundang Hospital, SungNam 463-707, Korea
| | - Hye In Yum
- Graduate School of Immunology, Seoul National University College of Medicine, Seoul 110-799, Korea
| | - Seung Pyo Park
- Transplantation Research Institute, Medical Research Center, Seoul National University College of Medicine, Seoul 110-799, Korea
| | - Jin Kyun Park
- Department of Internal Medicine, Seoul National University College of Medicine, Seoul 110-799, Korea; Division of Rheumatology, Department of Internal Medicine, Seoul National University Hospital, Seoul 110-744, Korea
| | - Eun Ha Kang
- Department of Internal Medicine, Seoul National University College of Medicine, Seoul 110-799, Korea; Division of Rheumatology, Department of Internal Medicine, Seoul National University Bundang Hospital, SungNam 463-707, Korea
| | - Jae-Il Lee
- Graduate School of Immunology, Seoul National University College of Medicine, Seoul 110-799, Korea; Transplantation Research Institute, Medical Research Center, Seoul National University College of Medicine, Seoul 110-799, Korea
| | - Eun Bong Lee
- Department of Internal Medicine, Seoul National University College of Medicine, Seoul 110-799, Korea; Division of Rheumatology, Department of Internal Medicine, Seoul National University Hospital, Seoul 110-744, Korea
| | - Chung-Gyu Park
- Transplantation Research Institute, Medical Research Center, Seoul National University College of Medicine, Seoul 110-799, Korea; Department of Microbiology and Immunology, Seoul National University College of Medicine, Seoul 110-799, Korea; Translational Xenotransplantation Research Center, Seoul National University College of Medicine, Seoul 110-799, Korea; and
| | - Kyeong Cheon Jung
- Graduate School of Immunology, Seoul National University College of Medicine, Seoul 110-799, Korea; Department of Pathology, Seoul National University College of Medicine, Seoul 110-799, Korea; Transplantation Research Institute, Medical Research Center, Seoul National University College of Medicine, Seoul 110-799, Korea; Department of Pathology, Seoul National University Hospital, Seoul 110-744, Korea
| | - Seong Hoe Park
- Graduate School of Immunology, Seoul National University College of Medicine, Seoul 110-799, Korea; Transplantation Research Institute, Medical Research Center, Seoul National University College of Medicine, Seoul 110-799, Korea;
| |
Collapse
|
30
|
Abstract
The immune system can be divided into innate and adaptive components that differ in their rate and mode of cellular activation, with innate immune cells being the first responders to invading pathogens. Recent advances in the identification and characterization of innate lymphoid cells have revealed reiterative developmental programs that result in cells with effector fates that parallel those of adaptive lymphoid cells and are tailored to effectively eliminate a broad spectrum of pathogenic challenges. However, activation of these cells can also be associated with pathologies such as autoimmune disease. One major distinction between innate and adaptive immune system cells is the constitutive expression of ID proteins in the former and inducible expression in the latter. ID proteins function as antagonists of the E protein transcription factors that play critical roles in lymphoid specification as well as B- and T-lymphocyte development. In this review, we examine the transcriptional mechanisms controlling the development of innate lymphocytes, including natural killer cells and the recently identified innate lymphoid cells (ILC1, ILC2, and ILC3), and innate-like lymphocytes, including natural killer T cells, with an emphasis on the known requirements for the ID proteins.
Collapse
Affiliation(s)
- Mihalis Verykokakis
- Committee on Immunology and Department of Pathology, The University of Chicago, Chicago, IL, USA
| | | | | |
Collapse
|
31
|
Miyazaki M, Miyazaki K, Chen S, Chandra V, Wagatsuma K, Agata Y, Rodewald HR, Saito R, Chang AN, Varki N, Kawamoto H, Murre C. The E-Id protein axis modulates the activities of the PI3K-AKT-mTORC1-Hif1a and c-myc/p19Arf pathways to suppress innate variant TFH cell development, thymocyte expansion, and lymphomagenesis. Genes Dev 2015; 29:409-25. [PMID: 25691468 PMCID: PMC4335296 DOI: 10.1101/gad.255331.114] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
Miyazaki et al. show that Id2 and Id3 suppress the development and expansion of innate variant TFH cells by acting upstream of the Hif1a/Foxo/AKT/mTORC1 pathway as well as the c-myc/p19Arf module. Mice depleted for Id2 and Id3 expression developed colitis and αβ T-cell lymphomas, and the transcription signatures of Id2- and Id3-depleted lymphomas revealed similarities to genetic deficiencies associated with Burkitt lymphoma. It is now well established that the E and Id protein axis regulates multiple steps in lymphocyte development. However, it remains unknown how E and Id proteins mechanistically enforce and maintain the naïve T-cell fate. Here we show that Id2 and Id3 suppressed the development and expansion of innate variant follicular helper T (TFH) cells. Innate variant TFH cells required major histocompatibility complex (MHC) class I-like signaling and were associated with germinal center B cells. We found that Id2 and Id3 induced Foxo1 and Foxp1 expression to antagonize the activation of a TFH transcription signature. We show that Id2 and Id3 acted upstream of the Hif1a/Foxo/AKT/mTORC1 pathway as well as the c-myc/p19Arf module to control cellular expansion. We found that mice depleted for Id2 and Id3 expression developed colitis and αβ T-cell lymphomas. Lymphomas depleted for Id2 and Id3 expression displayed elevated levels of c-myc, whereas p19Arf abundance declined. Transcription signatures of Id2- and Id3-depleted lymphomas revealed similarities to genetic deficiencies associated with Burkitt lymphoma. We propose that, in response to antigen receptor and/or cytokine signaling, the E–Id protein axis modulates the activities of the PI3K–AKT–mTORC1–Hif1a and c-myc/p19Arf pathways to control cellular expansion and homeostatic proliferation.
Collapse
Affiliation(s)
- Masaki Miyazaki
- Department of Molecular Biology, University of California at San Diego, La Jolla, California 92093, USA
| | - Kazuko Miyazaki
- Department of Molecular Biology, University of California at San Diego, La Jolla, California 92093, USA
| | - Shuwen Chen
- Department of Molecular Biology, University of California at San Diego, La Jolla, California 92093, USA
| | - Vivek Chandra
- Department of Molecular Biology, University of California at San Diego, La Jolla, California 92093, USA
| | - Keisuke Wagatsuma
- Department of Biochemistry and Molecular Biology, Shiga University of Medical School, Shiga 520-2192, Japan
| | - Yasutoshi Agata
- Department of Biochemistry and Molecular Biology, Shiga University of Medical School, Shiga 520-2192, Japan
| | - Hans-Reimer Rodewald
- Division of Cellular Immunology, German Cancer Research Center, D-69120 Heidelberg, Germany
| | - Rintaro Saito
- Department of Medicine, University of California at San Diego, La Jolla, California 92093, USA
| | - Aaron N Chang
- Center for Computational Biology, Institute for Genomic Medicine, University of California at San Diego, La Jolla, California 92093, USA
| | - Nissi Varki
- Department of Pathology, University of California at San Diego, La Jolla, California 92093, USA
| | - Hiroshi Kawamoto
- Department of Immunology, Institute for Frontier Medical Sciences, Kyoto University, Kyoto 606-8507, Japan
| | - Cornelis Murre
- Department of Molecular Biology, University of California at San Diego, La Jolla, California 92093, USA;
| |
Collapse
|
32
|
Singh SP, Zhang HH, Tsang H, Gardina PJ, Myers TG, Nagarajan V, Lee CH, Farber JM. PLZF regulates CCR6 and is critical for the acquisition and maintenance of the Th17 phenotype in human cells. THE JOURNAL OF IMMUNOLOGY 2015; 194:4350-61. [PMID: 25833398 DOI: 10.4049/jimmunol.1401093] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Subscribe] [Scholar Register] [Received: 04/29/2014] [Accepted: 03/01/2015] [Indexed: 12/31/2022]
Abstract
Th17 cells, which express the chemokine receptor CCR6, are implicated in many immune-mediated disorders, such as psoriasis and multiple sclerosis. We found that expression levels of CCR6 on human effector/memory CD4(+) T cells reflect a continuum of Th17 differentiation. By evaluating the transcriptome in cells with increasing CCR6, we detected progressive upregulation of ZBTB16, which encodes the broad complex, tramtrack, bric-à-brac-zinc finger transcription factor promyelocytic leukemia zinc finger protein (PLZF). Using chromatin immunoprecipitation for modified histones, p300, and PLZF, we identified enhancer-like sites at -9/-10 and -13/-14 kb from the upstream transcription start site of CCR6 that bind PLZF in CCR6(+) cells. For Th cells from adult blood, both in the CCR6(+) memory population and in naive cells activated ex vivo, knockdown of ZBTB16 downregulated CCR6 and other Th17-associated genes. ZBTB16 and RORC (which encodes the "master regulator" RORγt) cross-regulate each other, and PLZF binds at the RORC promoter in CCR6(+) cells. In naive Th cells from cord blood, ZBTB16 expression was confined to CD161(+) cells, which are Th17 cell precursors. ZBTB16 was not expressed in mouse Th17 cells, and Th17 cells could be made from luxoid mice, which harbor an inactivating mutation in Zbtb16. These studies demonstrate a role for PLZF as an activator of transcription important both for Th17 differentiation and the maintenance of the Th17 phenotype in human cells, expand the role of PLZF as a critical regulator in the human adaptive immune system, and identify a novel, essential element in a regulatory network that is of significant therapeutic interest.
Collapse
Affiliation(s)
- Satya P Singh
- Laboratory of Molecular Immunology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892
| | - Hongwei H Zhang
- Laboratory of Molecular Immunology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892
| | - Hsinyi Tsang
- Laboratory of Molecular Immunology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892
| | - Paul J Gardina
- Research Technologies Branch, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892; and
| | - Timothy G Myers
- Research Technologies Branch, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892; and
| | - Vijayaraj Nagarajan
- Bioinformatics and Computational Biosciences Branch, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892
| | - Chang Hoon Lee
- Laboratory of Molecular Immunology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892
| | - Joshua M Farber
- Laboratory of Molecular Immunology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892;
| |
Collapse
|
33
|
Analyses of the TCR repertoire of MHC class II-restricted innate CD4⁺ T cells. Exp Mol Med 2015; 47:e154. [PMID: 25813222 PMCID: PMC4351420 DOI: 10.1038/emm.2015.7] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2014] [Revised: 11/05/2014] [Accepted: 12/04/2014] [Indexed: 11/08/2022] Open
Abstract
Analysis of the T-cell receptor (TCR) repertoire of innate CD4(+) T cells selected by major histocompatibility complex (MHC) class II-dependent thymocyte-thymocyte (T-T) interaction (T-T CD4(+) T cells) is essential for predicting the characteristics of the antigens that bind to these T cells and for distinguishing T-T CD4(+) T cells from other types of innate T cells. Using the TCR(mini) Tg mouse model, we show that the repertoire of TCRα chains in T-T CD4(+) T cells was extremely diverse, in contrast to the repertoires previously described for other types of innate T cells. The TCRα chain sequences significantly overlapped between T-T CD4(+) T cells and conventional CD4(+) T cells in the thymus and spleen. However, the diversity of the TCRα repertoire of T-T CD4(+) T cells seemed to be restricted compared with that of conventional CD4(+) T cells. Interestingly, the frequency of the parental OT-II TCRα chains was significantly reduced in the process of T-T interaction. This diverse and shifted repertoire in T-T CD4(+) T cells has biological relevance in terms of defense against diverse pathogens and a possible regulatory role during peripheral T-T interaction.
Collapse
|
34
|
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: 4.1] [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
|
35
|
Abstract
Memory T cells are usually considered to be a feature of a successful immune response against a foreign antigen, and such cells can mediate potent immunity. However, in mice, alternative pathways have been described, through which naïve T cells can acquire the characteristics and functions of memory T cells without encountering specific foreign antigen or the typical signals required for conventional T cell differentiation. Such cells reflect a response to the internal rather the external environment, and hence such cells are called innate memory T cells. In this review, we describe how innate memory subsets were identified, the signals that induce their generation and their functional properties and potential role in the normal immune response. The existence of innate memory T cells in mice raises questions about whether parallel populations exist in humans, and we discuss the evidence for such populations during human T cell development and differentiation.
Collapse
Affiliation(s)
- Stephen C Jameson
- Center for Immunology, University of Minnesota Medical School, Minneapolis, Minnesota, USA.
| | - You Jeong Lee
- Center for Immunology, University of Minnesota Medical School, Minneapolis, Minnesota, USA
| | - Kristin A Hogquist
- Center for Immunology, University of Minnesota Medical School, Minneapolis, Minnesota, USA.
| |
Collapse
|
36
|
Prince AL, Kraus Z, Carty SA, Ng C, Yin CC, Jordan MS, Schwartzberg PL, Berg LJ. Development of innate CD4+ and CD8+ T cells in Itk-deficient mice is regulated by distinct pathways. THE JOURNAL OF IMMUNOLOGY 2014; 193:688-99. [PMID: 24943215 DOI: 10.4049/jimmunol.1302059] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
T cell development in the thymus produces multiple lineages of cells, including innate T cells such as γδ TCR(+) cells, invariant NKT cells, mucosal-associated invariant T cells, and H2-M3-specific cells. Although innate cells are generally a minor subset of thymocytes, in several strains of mice harboring mutations in T cell signaling proteins or transcriptional regulators, conventional CD8(+) T cells develop as innate cells with characteristics of memory T cells. Thus, in Itk-deficient mice, mature CD4(-)CD8(+) (CD8 single-positive [SP]) thymocytes express high levels of the transcription factor eomesodermin (Eomes) and are dependent on IL-4 being produced in the thymic environment by a poorly characterized subset of CD4(+) thymocytes expressing the transcriptional regulator promyelocytic leukemia zinc finger. In this study, we show that a sizeable proportion of mature CD4(+)CD8(-) (CD4SP) thymocytes in itk(-/-) mice also develop as innate Eomes-expressing T cells. These cells are dependent on MHC class II and IL-4 signaling for their development, indicating that they are conventional CD4(+) T cells that have been converted to an innate phenotype. Surprisingly, neither CD4SP nor CD8SP innate Eomes(+) thymocytes in itk(-/-) or SLP-76(Y145F) mice are dependent on γδ T cells for their development. Instead, we find that the predominant population of Eomes(+) innate itk(-/-) CD4SP thymocytes is largely absent in mice lacking CD1d-specific invariant NKT cells, with no effect on innate itk(-/-) CD8SP thymocytes. In contrast, both subsets of innate Eomes(+)itk(-/-) T cells require the presence of a novel promyelocytic leukemia zinc finger-expressing, SLAM family receptor adapter protein-dependent thymocyte population that is essential for the conversion of conventional CD4(+) and CD8(+) T cells into innate T cells with a memory phenotype.
Collapse
Affiliation(s)
- Amanda L Prince
- Department of Pathology, University of Massachusetts Medical School, Worcester, MA 01655
| | - Zachary Kraus
- National Human Genome Research Institute, National Institutes of Health, Bethesda, MD 20892; and
| | - Shannon A Carty
- Department of Pathology and Laboratory Medicine, University of Pennsylvania, Philadelphia, PA 19104
| | - Caleb Ng
- Department of Pathology and Laboratory Medicine, University of Pennsylvania, Philadelphia, PA 19104
| | - Catherine C Yin
- Department of Pathology, University of Massachusetts Medical School, Worcester, MA 01655
| | - Martha S Jordan
- Department of Pathology and Laboratory Medicine, University of Pennsylvania, Philadelphia, PA 19104
| | - Pamela L Schwartzberg
- National Human Genome Research Institute, National Institutes of Health, Bethesda, MD 20892; and
| | - Leslie J Berg
- Department of Pathology, University of Massachusetts Medical School, Worcester, MA 01655;
| |
Collapse
|
37
|
Prince AL, Watkin LB, Yin CC, Selin LK, Kang J, Schwartzberg PL, Berg LJ. Innate PLZF+CD4+ αβ T cells develop and expand in the absence of Itk. THE JOURNAL OF IMMUNOLOGY 2014; 193:673-87. [PMID: 24928994 DOI: 10.4049/jimmunol.1302058] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
T cell development in the thymus produces multiple lineages of cells, including innate T cells. Studies in mice harboring alterations in TCR signaling proteins or transcriptional regulators have revealed an expanded population of CD4(+) innate T cells in the thymus that produce IL-4 and express the transcription factor promyelocytic leukemia zinc finger (PLZF). In these mice, IL-4 produced by the CD4(+)PLZF(+) T cell population leads to the conversion of conventional CD8(+) thymocytes into innate CD8(+) T cells resembling memory T cells expressing eomesodermin. The expression of PLZF, the signature invariant NKT cell transcription factor, in these innate CD4(+) T cells suggests that they might be a subset of αβ or γδ TCR(+) NKT cells or mucosal-associated invariant T (MAIT) cells. To address these possibilities, we characterized the CD4(+)PLZF(+) innate T cells in itk(-/-) mice. We show that itk(-/-) innate PLZF(+)CD4(+) T cells are not CD1d-dependent NKT cells, MR1-dependent MAIT cells, or γδ T cells. Furthermore, although the itk(-/-) innate PLZF(+)CD4(+) T cells express αβ TCRs, neither β2-microglobulin-dependent MHC class I nor any MHC class II molecules are required for their development. In contrast to invariant NKT cells and MAIT cells, this population has a highly diverse TCRα-chain repertoire. Analysis of peripheral tissues indicates that itk(-/-) innate PLZF(+)CD4(+) T cells preferentially home to spleen and mesenteric lymph nodes owing to increased expression of gut-homing receptors, and that their expansion is regulated by commensal gut flora. These data support the conclusion that itk(-/-) innate PLZF(+)CD4(+) T cells are a novel subset of innate T cells.
Collapse
Affiliation(s)
- Amanda L Prince
- Department of Pathology, University of Massachusetts Medical School, Worcester, MA 01655; and
| | - Levi B Watkin
- Department of Pathology, University of Massachusetts Medical School, Worcester, MA 01655; and
| | - Catherine C Yin
- Department of Pathology, University of Massachusetts Medical School, Worcester, MA 01655; and
| | - Liisa K Selin
- Department of Pathology, University of Massachusetts Medical School, Worcester, MA 01655; and
| | - Joonsoo Kang
- Department of Pathology, University of Massachusetts Medical School, Worcester, MA 01655; and
| | - Pamela L Schwartzberg
- National Human Genome Research Institute, National Institutes of Health, Bethesda, MD 20814
| | - Leslie J Berg
- Department of Pathology, University of Massachusetts Medical School, Worcester, MA 01655; and
| |
Collapse
|
38
|
Hashiguchi T, Oyamada A, Sakuraba K, Shimoda K, Nakayama KI, Iwamoto Y, Yoshikai Y, Yamada H. Tyk2-Dependent Bystander Activation of Conventional and Nonconventional Th1 Cell Subsets Contributes to Innate Host Defense againstListeria monocytogenesInfection. THE JOURNAL OF IMMUNOLOGY 2014; 192:4739-47. [DOI: 10.4049/jimmunol.1303067] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
|
39
|
TL1A induces TCR independent IL-6 and TNF-α production and growth of PLZF⁺ leukocytes. PLoS One 2014; 9:e85793. [PMID: 24416448 PMCID: PMC3885722 DOI: 10.1371/journal.pone.0085793] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2013] [Accepted: 12/08/2013] [Indexed: 12/24/2022] Open
Abstract
An elevated level of the cytokine TL1A is known to be associated with several autoimmune diseases, e.g. rheumatoid arthritis and inflammatory bowel disease. However, the mode of action of TL1A remains elusive. In this study, we investigated the role of TL1A in a pro-inflammatory setting, using human leukocytes purified from healthy donors. We show that TL1A, together with IL-12, IL-15 and IL-18, directly induces the production of IL-6 and TNF-α from leukocytes. Interestingly, TL1A-induced IL-6 was not produced by CD14+ monocytes. We further show that the produced IL-6 is fully functional, as measured by its ability to signal through the IL-6 receptor, and that the induction of IL-6 is independent of TCR stimulation. Furthermore, the transcription factor PLZF was induced in stimulated cells. These results offer a substantial explanation for the role of TL1A, since TNF-α and IL-6 are directly responsible for much of the inflammatory state in many autoimmune diseases. Our study suggests that TL1A is a possible target for the treatment of autoimmune diseases.
Collapse
|
40
|
The role of BTB-zinc finger transcription factors during T cell development and in the regulation of T cell-mediated immunity. Curr Top Microbiol Immunol 2014; 381:21-49. [PMID: 24850219 DOI: 10.1007/82_2014_374] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
The proper regulation of the development and function of peripheral helper and cytotoxic T cell lineages is essential for T cell-mediated adaptive immunity. Progress made during the last 10-15 years led to the identification of several transcription factors and transcription factor networks that control the development and function of T cell subsets. Among the transcription factors identified are also several members of the so-called BTB/POZ domain containing zinc finger (ZF) transcription factor family (BTB-ZF), and important roles of BTB-ZF factors have been described. In this review, we will provide an up-to-date overview about the role of BTB-ZF factors during T cell development and in peripheral T cells.
Collapse
|
41
|
Zhu L, Qiao Y, Choi ES, Das J, Sant'angelo DB, Chang CH. A transgenic TCR directs the development of IL-4+ and PLZF+ innate CD4 T cells. THE JOURNAL OF IMMUNOLOGY 2013; 191:737-44. [PMID: 23776174 DOI: 10.4049/jimmunol.1300862] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
MHC class II-expressing thymocytes can efficiently mediate positive selection of CD4 T cells in mice. Thymocyte-selected CD4 (T-CD4) T cells have an innate-like phenotype similar to invariant NKT cells. To investigate the development and function of T-CD4 T cells in-depth, we cloned TCR genes from T-CD4 T cells and generated transgenic mice. Remarkably, positive selection of T-CD4 TCR transgenic (T3) thymocytes occurred more efficiently when MHC class II was expressed by thymocytes than by thymic epithelial cells. Similar to polyclonal T-CD4 T cells and also invariant NKT cells, T3 CD4 T cell development is controlled by signaling lymphocyte activation molecule/signaling lymphocyte activation molecule-associated protein signaling, and the cells expressed both IL-4 and promyelocytic leukemia zinc finger (PLZF). Surprisingly, the selected T3 CD4 T cells were heterogeneous in that only half expressed IL-4 and only half expressed PLZF. IL-4- and PLZF-expressing cells were first found at the double-positive cell stage. Thus, the expression of IL-4 and PLZF seems to be determined by an unidentified event that occurs postselection and is not solely dependent on TCR specificity or the selection process, per se. Taken together, our data show for the first time, to our knowledge, that the TCR specificity regulates but does not determine the development of innate CD4 T cells by thymocytes.
Collapse
Affiliation(s)
- Lingqiao Zhu
- Department of Microbiology and Immunology, University of Michigan Medical School, Ann Arbor, MI 48109, USA
| | | | | | | | | | | |
Collapse
|
42
|
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: 3.9] [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
|
43
|
Dutta M, Kraus ZJ, Gomez-Rodriguez J, Hwang SH, Cannons JL, Cheng J, Lee SY, Wiest DL, Wakeland EK, Schwartzberg PL. A role for Ly108 in the induction of promyelocytic zinc finger transcription factor in developing thymocytes. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2013; 190:2121-8. [PMID: 23355739 PMCID: PMC3578000 DOI: 10.4049/jimmunol.1202145] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Abstract
The promyelocytic zinc finger transcription factor (PLZF) is required for the development of activated phenotypes in NKT and other innate T lymphocytes. Although strong TCR stimulation has been implicated in the induction of PLZF, the factors regulating PLZF expression are incompletely understood. We show in this study that costimulation of preselection double-positive thymocytes through the signaling lymphocyte activation molecule family receptor Ly108 markedly enhanced PLZF expression compared with that induced by TCR stimulation alone. Costimulation with Ly108 increased expression of early growth response protein (Egr)-2 and binding of Egr-2 to the promoter of Zbtb16, which encodes PLZF, and resulted in PLZF levels similar to those seen in NKT cells. In contrast, costimulation with anti-CD28 failed to enhance Egr-2 binding and Zbtb16 expression. Moreover, mice lacking Ly108 showed decreased numbers of PLZF-expressing CD4(+) T cells. Together, these results support a potential role for Ly108 in the induction of PLZF.
Collapse
Affiliation(s)
- Mala Dutta
- National Human Genome Research Institute, NIH, Bethesda, MD 20892
- George Washington University Institute of Biomedical Sciences, Washington, DC 20052
| | - Zachary J. Kraus
- National Human Genome Research Institute, NIH, Bethesda, MD 20892
| | | | - Sun-hee Hwang
- Department of Immunology, University of Texas Southwestern Medical Center, Dallas, TX
| | | | - Jun Cheng
- National Human Genome Research Institute, NIH, Bethesda, MD 20892
| | - Sang-Yun Lee
- Immune Cell Development and Host Defense Program, Fox Chase Cancer Center, Philadelphia, PA 19111
| | - David L. Wiest
- Immune Cell Development and Host Defense Program, Fox Chase Cancer Center, Philadelphia, PA 19111
| | - Edward K. Wakeland
- Department of Immunology, University of Texas Southwestern Medical Center, Dallas, TX
| | | |
Collapse
|
44
|
Systemic Human T Cell Developmental Processes in Humanized Mice Cotransplanted With Human Fetal Thymus/Liver Tissue and Hematopoietic Stem Cells. Transplantation 2012; 94:1095-102. [DOI: 10.1097/tp.0b013e318270f392] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
|
45
|
Development of promyelocytic leukemia zinc finger-expressing innate CD4 T cells requires stronger T-cell receptor signals than conventional CD4 T cells. Proc Natl Acad Sci U S A 2012; 109:16264-9. [PMID: 22988097 DOI: 10.1073/pnas.1207528109] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
MHC class II-expressing thymocytes and thymic epithelial cells can mediate CD4 T-cell selection resulting in functionally distinct thymocyte-selected CD4 (T-CD4) and epithelial-selected CD4 (E-CD4) T cells, respectively. However, little is known about how T-cell receptor (TCR) signaling influences the development of these two CD4 T-cell subsets. To study TCR signaling for T-CD4 T-cell development, we used a GFP reporter system of Nur77 in which GFP intensity directly correlates with TCR signaling strength. T-CD4 T cells expressed higher levels of GFP than E-CD4 T cells, suggesting that T-CD4 T cells received stronger TCR signaling than E-CD4 T cells during selection. Elimination of Ras GTPase-activating protein enhanced E-CD4 but decreased T-CD4 T-cell selection efficiency, suggesting a shift to negative selection. Conversely, the absence of IL-2-inducible T-cell kinase that causes poor E-CD4 T-cell selection due to insufficient TCR signaling improved T-CD4 T-cell generation, consistent with rescue from negative selection. Strong TCR signaling during T-CD4 T-cell development correlates with the expression of the transcription factor promyelocytic leukemia zinc finger protein. However, although modulation of the signaling strength affected the efficiency of T-CD4 T-cell development during positive and negative selection, the signaling strength is not as important for the effector function of T-CD4 T cells. These findings indicate that innate T-CD4 T cells, together with invariant natural killer T cells and γδ T cells, receive strong TCR signals during their development and that signaling requirements for the development and the effector functions are distinct.
Collapse
|
46
|
Jeon YK, Go H, Nam SJ, Keam B, Kim TM, Jung KC, Kang HJ, Lee DS, Huh JR, Park SH. Expression of the promyelocytic leukemia zinc-finger in T-lymphoblastic lymphoma and leukemia has strong implications for their cellular origin and greater association with initial bone marrow involvement. Mod Pathol 2012; 25:1236-45. [PMID: 22555178 DOI: 10.1038/modpathol.2012.82] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023]
Abstract
The promyelocytic leukemia zinc-finger (PLZF) is essential for the development of innate T cells (as represented by natural killer T cells) for acquisition of their unique innate immune properties. We evaluated the PLZF protein expression in a variety of immature and mature lymphoid malignancies. PLZF was preferentially expressed in T-lymphoblastic lymphoma/acute lymphoblastic leukemia (T-LBL/ALL) in 50% of the 54 cases. Among 51 cases of peripheral T-cell lymphoma not otherwise specified, only one (2%) expressed PLZF. One mycosis fungoides case expressed PLZF in lymph node involved by tumor. Otherwise, PLZF was not detected in any other type of lymphoma. In T-LBL/ALL, PLZF expression was more common in CD4/CD8 double-negative (67%) or CD8 single-positive subtypes (73%) than in CD4/CD8 double-positive (13%) and CD4 single-positive subtypes (0%) (P=0.001). Importantly, PLZF and CD1a expression were mutually exclusive in T-LBL/ALL (P=0.001). This was also the case for T-cell receptor βF1 expression (P=0.000). Most (96%) of the PLZF-positive T-LBL/ALL cases showed initial bone marrow involvement compared with 39% of PLZF-negative cases (P=0.000). Based on these findings, we suggest that T-LBL/ALLs that express PLZF arise from early immature double-negative thymocytes when the T-cell receptor β chain has not yet expressed or innate T-cell precursors, and strongly imply bone marrow involvement.
Collapse
Affiliation(s)
- Yoon K Jeon
- Department of Pathology, Seoul National University Hospital, Seoul, South Korea.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
47
|
Leishmania major infection in humanized mice induces systemic infection and provokes a nonprotective human immune response. PLoS Negl Trop Dis 2012; 6:e1741. [PMID: 22848771 PMCID: PMC3404120 DOI: 10.1371/journal.pntd.0001741] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2012] [Accepted: 06/07/2012] [Indexed: 12/03/2022] Open
Abstract
Background Leishmania (L.) species are the causative agent of leishmaniasis. Due to the lack of efficient vaccine candidates, drug therapies are the only option to deal with cutaneous leishmaniasis. Unfortunately, chemotherapeutic interventions show high toxicity in addition to an increased risk of dissemination of drug-resistant parasites. An appropriate laboratory animal based model is still missing which allows testing of new drug strategies in the context of human immune cells in vivo. Methodology/Principal Findings Humanized mice were infected subcutaneously with stationary phase promastigote L. major into the footpad. The human immune response against the pathogen and the parasite host interactions were analyzed. In addition we proved the versatility of this new model to conduct drug research studies by the inclusion of orally given Miltefosine. We show that inflammatory human macrophages get infected with Leishmania parasites at the site of infection. Furthermore, a Leishmania-specific human-derived T cell response is initiated. However, the human immune system is not able to prevent systemic infection. Thus, we treated the mice with Miltefosine to reduce the parasitic load. Notably, this chemotherapy resulted in a reduction of the parasite load in distinct organs. Comparable to some Miltefosine treated patients, humanized mice developed severe side effects, which are not detectable in the classical murine model of experimental leishmaniasis. Conclusions/Significance This study describes for the first time L. major infection in humanized mice, characterizes the disease development, the induction of human adaptive and innate immune response including cytokine production and the efficiency of Miltefosine treatment in these animals. In summary, humanized mice might be beneficial for future preclinical chemotherapeutic studies in systemic (visceral) leishmaniasis allowing the investigation of human immune response, side effects of the drug due to cytokine production of activated humane immune cells and the efficiency of the treatment to eliminate also not replicating (“hiding”) parasites. As many as 12 million people suffer from Leishmania (L.) infection worldwide with about one to two million newly infected people every year. Due to the lack of vaccine strategies, the only option is chemotherapeutic intervention which can cause serious side effects. Therefore, new prevention or treatment strategies are urgently needed in addition to an appropriate animal model for testing. We infected humanized mice in the footpad with stationary phase promastigote L. major and analyzed the human innate and adaptive immune response by flow cytometry, histology, and quantitative PCR. Infected macrophages were detectable at the site of infection and in lymphoid organs. Additionally, we were able to measure Leishmania-specific T cell priming in humanized mice. However, these human immune defense mechanisms were not sufficient to prevent systemic spreading and lethality. By the inclusion of Miltefosine, we tested this novel model for its versatility in conducting drug studies. The oral treatment was able to reduce parasitic load but also revealed side effects which are described in humans but not in mice. Therefore, we propose humanized mice as a novel model, which offers the opportunity to study new therapy strategies in chronic leishmaniasis in the context of a human immune system.
Collapse
|
48
|
Yuan J, Nguyen CK, Liu X, Kanellopoulou C, Muljo SA. Lin28b reprograms adult bone marrow hematopoietic progenitors to mediate fetal-like lymphopoiesis. Science 2012; 335:1195-200. [PMID: 22345399 DOI: 10.1126/science.1216557] [Citation(s) in RCA: 265] [Impact Index Per Article: 20.4] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/02/2022]
Abstract
The immune system develops in waves during ontogeny; it is initially populated by cells generated from fetal hematopoietic stem cells (HSCs) and later by cells derived from adult HSCs. Remarkably, the genetic programs that control these two distinct stem cell fates remain poorly understood. We report that Lin28b is specifically expressed in mouse and human fetal liver and thymus, but not in adult bone marrow or thymus. We demonstrate that ectopic expression of Lin28 reprograms hematopoietic stem/progenitor cells (HSPCs) from adult bone marrow, which endows them with the ability to mediate multilineage reconstitution that resembles fetal lymphopoiesis, including increased development of B-1a, marginal zone B, gamma/delta (γδ) T cells, and natural killer T (NKT) cells.
Collapse
Affiliation(s)
- Joan Yuan
- Laboratory of Immunology, National Institute of Allergy and Infectious Diseases (NIAID), National Institutes of Health, Bethesda, MD 20892, USA
| | | | | | | | | |
Collapse
|
49
|
Qiao Y, Gray BM, Sofi MH, Bauler LD, Eaton KA, O'Riordan MXD, Chang CH. Innate-like CD4 T cells selected by thymocytes suppress adaptive immune responses against bacterial infections. ACTA ACUST UNITED AC 2011; 2:25-39. [PMID: 23264931 DOI: 10.4236/oji.2012.21004] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
We have reported a new innate-like CD4 T cell population that expresses cell surface makers of effector/memory cells and produce Th1 and Th2 cytokines immediately upon activation. Unlike conventional CD4 T cells that are selected by thymic epithelial cells, these CD4 T cells, named T-CD4 T cells, are selected by MHC class II expressing thymocytes. Previously, we showed that the presence of T-CD4 T cells protected mice from airway inflammation suggesting an immune regulatory role of T-CD4 T cells. To further understand the function of T-CD4 T cells, we investigated immune responses mediated by T-CD4 T cells during bacterial infection because the generation of antigen specific CD4 T cells contributes to clearance of infection and for the development of immune memory. The current study shows a suppressive effect of T-CD4 T cells on both CD8 and CD4 T cell-mediated immune responses during Listeria and Helicobacter infections. In the mouse model of Listeria monocytogenes infection, T-CD4 T cells resulted in decreasedfrequency of Listeria-specific CD8 T cells and the killing activity of them. Furthermore, mice with T-CD4 T cells developed poor immune memory, demonstrated by reduced expansion of antigen-specific T cells and high bacterial burden upon re-infection. Similarly, the presence of T-CD4 T cells suppressed the generation of antigen-specific CD4 T cells in Helicobacter pylori infected mice. Thus, our studies reveal a novel function of T-CD4 T cells in suppressing anti-bacterial immunity.
Collapse
Affiliation(s)
- Yu Qiao
- Department of Microbiology and Immunology, University of Michigan, Ann Arbor, USA
| | | | | | | | | | | | | |
Collapse
|
50
|
Abstract
Human mucosal associated invariant T (MAIT) CD8(+) and Tc17 cells are important tissue-homing cell populations, characterized by high expression of CD161 ((++)) and type-17 differentiation, but their origins and relationships remain poorly defined. By transcriptional and functional analyses, we demonstrate that a pool of polyclonal, precommitted type-17 CD161(++)CD8αβ(+) T cells exist in cord blood, from which a prominent MAIT cell (TCR Vα7.2(+)) population emerges post-natally. During this expansion, CD8αα T cells appear exclusively within a CD161(++)CD8(+)/MAIT subset, sharing cytokine production, chemokine-receptor expression, TCR-usage, and transcriptional profiles with their CD161(++)CD8αβ(+) counterparts. Our data demonstrate the origin and differentiation pathway of MAIT-cells from a naive type-17 precommitted CD161(++)CD8(+) T-cell pool and the distinct phenotype and function of CD8αα cells in man.
Collapse
|