1
|
Krayem I, Grusanovic S, Duric I, Pavliuchenko N, Danek P, Borna S, Sekeresova Kralova J, Skopcova T, Pokorna J, Alberich-Jorda M, Brdicka T. WBP1L regulates hematopoietic stem cell function and T cell development. Front Immunol 2024; 15:1421512. [PMID: 39555063 PMCID: PMC11563793 DOI: 10.3389/fimmu.2024.1421512] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2024] [Accepted: 10/14/2024] [Indexed: 11/19/2024] Open
Abstract
WW domain binding protein 1-like (WBP1L), also known as outcome predictor of acute leukemia 1 (OPAL1), is a transmembrane adaptor protein, expression of which was shown to correlate with ETV6-RUNX1 translocation and favorable prognosis in childhood leukemia. It has a broad expression pattern in hematopoietic and non-hematopoietic cells. Our previous work described WBP1L as a regulator of CXCR4 signaling and hematopoiesis. Here, we show that hematopoiesis in the mice with Wbp1l germline deletion is dysregulated, already at the level of hematopoietic stem cells and early progenitors. We further demonstrate that thymi of WBP1L-deficient mice are significantly enlarged and contain increased numbers of thymocytes of all subsets. This can potentially be explained by increased generation of multipotent progenitors 4 (MPP4) in the bone marrow, from which the thymus-seeding progenitors are derived. We also observed increases in multiple cell types in the blood. In addition, we show that WBP1L regulates hematopoietic stem cell functionality and leukocyte progenitor proliferation and gene expression during hematopoietic stem and progenitor cell transplantation, which contribute to more efficient engraftment of WBP1L-deficient cells. WBP1L thus emerges as a regulator of hematopoietic stem and progenitor cell function, which controls leukocyte numbers at the steady state and after bone marrow transplantation.
Collapse
Affiliation(s)
- Imtissal Krayem
- Laboratory of Leukocyte Signalling, Institute of Molecular Genetics of the Czech Academy of Sciences, Prague, Czechia
| | - Srdjan Grusanovic
- Laboratory of Hemato-Oncology, Institute of Molecular Genetics of the Czech Academy of Sciences, Prague, Czechia
| | - Iris Duric
- Laboratory of Leukocyte Signalling, Institute of Molecular Genetics of the Czech Academy of Sciences, Prague, Czechia
- Faculty of Science, Charles University, Prague, Czechia
| | - Nataliia Pavliuchenko
- Laboratory of Leukocyte Signalling, Institute of Molecular Genetics of the Czech Academy of Sciences, Prague, Czechia
| | - Petr Danek
- Laboratory of Hemato-Oncology, Institute of Molecular Genetics of the Czech Academy of Sciences, Prague, Czechia
- Molecular Analysis of Growth Regulation in Animals, Institute of Organic Chemistry and Biochemistry of the Czech Academy of Sciences, Prague, Czechia
| | - Simon Borna
- Laboratory of Leukocyte Signalling, Institute of Molecular Genetics of the Czech Academy of Sciences, Prague, Czechia
| | - Jarmila Sekeresova Kralova
- Laboratory of Leukocyte Signalling, Institute of Molecular Genetics of the Czech Academy of Sciences, Prague, Czechia
| | - Tereza Skopcova
- Laboratory of Leukocyte Signalling, Institute of Molecular Genetics of the Czech Academy of Sciences, Prague, Czechia
| | - Jana Pokorna
- Laboratory of Leukocyte Signalling, Institute of Molecular Genetics of the Czech Academy of Sciences, Prague, Czechia
| | - Meritxell Alberich-Jorda
- Laboratory of Hemato-Oncology, Institute of Molecular Genetics of the Czech Academy of Sciences, Prague, Czechia
- Childhood Leukaemia Investigation Prague, Department of Pediatric Haematology and Oncology, 2Faculty of Medicine, University Hospital Motol, Charles University, Prague, Czechia
| | - Tomas Brdicka
- Laboratory of Leukocyte Signalling, Institute of Molecular Genetics of the Czech Academy of Sciences, Prague, Czechia
| |
Collapse
|
2
|
Shin B, Chang SJ, MacNabb BW, Rothenberg EV. Transcriptional network dynamics in early T cell development. J Exp Med 2024; 221:e20230893. [PMID: 39167073 PMCID: PMC11338287 DOI: 10.1084/jem.20230893] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2024] [Revised: 06/07/2024] [Accepted: 07/10/2024] [Indexed: 08/23/2024] Open
Abstract
The rate at which cells enter the T cell pathway depends not only on the immigration of hematopoietic precursors into the strong Notch signaling environment of the thymus but also on the kinetics with which each individual precursor cell reaches T-lineage commitment once it arrives. Notch triggers a complex, multistep gene regulatory network in the cells in which the steps are stereotyped but the transition speeds between steps are variable. Progenitor-associated transcription factors delay T-lineage differentiation even while Notch-induced transcription factors within the same cells push differentiation forward. Progress depends on regulator cross-repression, on breaching chromatin barriers, and on shifting, competitive collaborations between stage-specific and stably expressed transcription factors, as reviewed here.
Collapse
Affiliation(s)
- Boyoung Shin
- Division of Biology and Biological Engineering California Institute of Technology , Pasadena, CA, USA
| | - Samantha J Chang
- Division of Biology and Biological Engineering California Institute of Technology , Pasadena, CA, USA
| | - Brendan W MacNabb
- Division of Biology and Biological Engineering California Institute of Technology , Pasadena, CA, USA
| | - Ellen V Rothenberg
- Division of Biology and Biological Engineering California Institute of Technology , Pasadena, CA, USA
| |
Collapse
|
3
|
MacNabb BW, Rothenberg EV. Speed and navigation control of thymocyte development by the fetal T-cell gene regulatory network. Immunol Rev 2023; 315:171-196. [PMID: 36722494 PMCID: PMC10771342 DOI: 10.1111/imr.13190] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
Abstract
T-cell differentiation is a tightly regulated developmental program governed by interactions between transcription factors (TFs) and chromatin landscapes and affected by signals received from the thymic stroma. This process is marked by a series of checkpoints: T-lineage commitment, T-cell receptor (TCR)β selection, and positive and negative selection. Dynamically changing combinations of TFs drive differentiation along the T-lineage trajectory, through mechanisms that have been most extensively dissected in adult mouse T-lineage cells. However, fetal T-cell development differs from adult in ways that suggest that these TF mechanisms are not fully deterministic. The first wave of fetal T-cell differentiation occurs during a unique developmental window during thymic morphogenesis, shows more rapid kinetics of differentiation with fewer rounds of cell division, and gives rise to unique populations of innate lymphoid cells (ILCs) and invariant γδT cells that are not generated in the adult thymus. As the characteristic kinetics and progeny biases are cell-intrinsic properties of thymic progenitors, the differences could be based on distinct TF network circuitry within the progenitors themselves. Here, we review recent single-cell transcriptome data that illuminate the TF networks involved in T-cell differentiation in the fetal and adult mouse thymus.
Collapse
Affiliation(s)
- Brendan W MacNabb
- Division of Biology & Biological Engineering, California Institute of Technology, Pasadena, California, USA
| | - Ellen V Rothenberg
- Division of Biology & Biological Engineering, California Institute of Technology, Pasadena, California, USA
| |
Collapse
|
4
|
Shin B, Rothenberg EV. Multi-modular structure of the gene regulatory network for specification and commitment of murine T cells. Front Immunol 2023; 14:1108368. [PMID: 36817475 PMCID: PMC9928580 DOI: 10.3389/fimmu.2023.1108368] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2022] [Accepted: 01/11/2023] [Indexed: 02/04/2023] Open
Abstract
T cells develop from multipotent progenitors by a gradual process dependent on intrathymic Notch signaling and coupled with extensive proliferation. The stages leading them to T-cell lineage commitment are well characterized by single-cell and bulk RNA analyses of sorted populations and by direct measurements of precursor-product relationships. This process depends not only on Notch signaling but also on multiple transcription factors, some associated with stemness and multipotency, some with alternative lineages, and others associated with T-cell fate. These factors interact in opposing or semi-independent T cell gene regulatory network (GRN) subcircuits that are increasingly well defined. A newly comprehensive picture of this network has emerged. Importantly, because key factors in the GRN can bind to markedly different genomic sites at one stage than they do at other stages, the genes they significantly regulate are also stage-specific. Global transcriptome analyses of perturbations have revealed an underlying modular structure to the T-cell commitment GRN, separating decisions to lose "stem-ness" from decisions to block alternative fates. Finally, the updated network sheds light on the intimate relationship between the T-cell program, which depends on the thymus, and the innate lymphoid cell (ILC) program, which does not.
Collapse
Affiliation(s)
- Boyoung Shin
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA, United States
| | - Ellen V. Rothenberg
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA, United States
| |
Collapse
|
5
|
Lisovska N. Multilevel mechanism of immune checkpoint inhibitor action in solid tumors: History, present issues and future development (Review). Oncol Lett 2022; 23:190. [PMID: 35527781 PMCID: PMC9073577 DOI: 10.3892/ol.2022.13310] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2022] [Accepted: 03/31/2022] [Indexed: 11/12/2022] Open
Abstract
Immunotherapy with checkpoint inhibitors (antibodies that target and block immune checkpoints in the tumor microenvironment) is included in the standard of care for patients with different types of malignancy, such as melanoma, renal cell and urothelial carcinoma, lung cancer etc. The introduction of this new immunotherapy has altered the view on potential targets for treatment of solid tumors from tumor cells themselves to their immune microenvironment; this has led to a reconsideration of the mechanisms of tumor-associated immunity. However, only a subset of patients benefit from immunotherapy and patient response is often unpredictable, even with known initial levels of prognostic markers; the biomarkers for favorable response are still being investigated. Mechanisms of immune checkpoint inhibitors efficiency, as well as the origins of treatment failure, require further investigation. From a clinical standpoint, discrepancies between the theoretical explanation of inhibitors of immune checkpoint actions at the cellular level and their deployment at a tissue/organ level impede the effective clinical implementation of novel immune therapy. The present review assessed existing experimental and clinical data on functional activity of inhibitors of immune checkpoints to provide a more comprehensive picture of their mechanisms of action on a cellular and higher levels of biological organization.
Collapse
Affiliation(s)
- Natalya Lisovska
- Chemotherapy Department, Center of Oncology, ‘Cyber Clinic of Spizhenko’, Kapitanovka, Kyiv 08112, Ukraine
| |
Collapse
|
6
|
The sialyltransferase ST3Gal-IV guides murine T-cell progenitors to the thymus. Blood Adv 2021; 4:1930-1941. [PMID: 32380539 DOI: 10.1182/bloodadvances.2019001046] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2019] [Accepted: 04/12/2020] [Indexed: 12/15/2022] Open
Abstract
T lymphocytes are important players in beneficial and detrimental immune responses. In contrast to other lymphocyte populations that develop in the bone marrow, T-cell precursors need to migrate to the thymus for further development. The interaction of P-selectin and P-selectin glycoprotein ligand-1 (PSGL-1) is crucial for thymic entry of T-cell precursors during settings of T-cell lineage reconstitution. PSGL-1 has to be sialylated to function as a ligand for P-selectin, and the sialyltransferase ST3Gal-IV might play a critical role in this process. We therefore investigated the role of ST3Gal-IV for T-cell development using competitive mixed bone marrow chimeric mice. We found that ST3Gal-IV is dispensable for homing and engraftment of hematopoietic precursors in the bone marrow. However, ST3Gal-IV deficiency affects seeding of the thymus by early T-cell progenitors, leading to impaired restoration of the peripheral T-cell compartment. This defect could be restored by ectopic retroviral expression of ST3Gal-IV in hematopoietic stem cells derived from ST3Gal-IV-deficient donor mice. Our findings show that ST3Gal-IV plays a critical and nonredundant role for efficient T-cell lineage reconstitution after bone marrow transplantation.
Collapse
|
7
|
Modeling the Dynamics of T-Cell Development in the Thymus. ENTROPY 2021; 23:e23040437. [PMID: 33918050 PMCID: PMC8069328 DOI: 10.3390/e23040437] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/15/2021] [Revised: 04/01/2021] [Accepted: 04/05/2021] [Indexed: 12/24/2022]
Abstract
The thymus hosts the development of a specific type of adaptive immune cells called T cells. T cells orchestrate the adaptive immune response through recognition of antigen by the highly variable T-cell receptor (TCR). T-cell development is a tightly coordinated process comprising lineage commitment, somatic recombination of Tcr gene loci and selection for functional, but non-self-reactive TCRs, all interspersed with massive proliferation and cell death. Thus, the thymus produces a pool of T cells throughout life capable of responding to virtually any exogenous attack while preserving the body through self-tolerance. The thymus has been of considerable interest to both immunologists and theoretical biologists due to its multi-scale quantitative properties, bridging molecular binding, population dynamics and polyclonal repertoire specificity. Here, we review experimental strategies aimed at revealing quantitative and dynamic properties of T-cell development and how they have been implemented in mathematical modeling strategies that were reported to help understand the flexible dynamics of the highly dividing and dying thymic cell populations. Furthermore, we summarize the current challenges to estimating in vivo cellular dynamics and to reaching a next-generation multi-scale picture of T-cell development.
Collapse
|
8
|
Liu C, Lan Y, Liu B, Zhang H, Hu H. T Cell Development: Old Tales Retold By Single-Cell RNA Sequencing. Trends Immunol 2021; 42:165-175. [PMID: 33446417 DOI: 10.1016/j.it.2020.12.004] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2020] [Revised: 12/07/2020] [Accepted: 12/07/2020] [Indexed: 02/07/2023]
Abstract
Mammalian T cell development initiates from the migration of hematopoietic progenitors to the thymus, which undergo cell proliferation, T-lineage specification and commitment, as well as positive and negative selection. These processes are precisely controlled at multiple levels and have been intensively studied using gene-modified animal models and in vitro coculture systems. However, several long-standing questions, including the characterization of the rare but crucial progenitors/precursors and the molecular mechanisms underlying their fate decision, have been dampened because of cell scarcity and lack of appropriate techniques. Single-cell RNA sequencing (scRNA-seq) makes it possible to investigate and resolve some of these questions, leading to new remarkable progress in identifying and characterizing early thymic progenitors and delineating the refined developmental trajectories of conventional and unconventional T cells.
Collapse
Affiliation(s)
- Chen Liu
- State Key Laboratory of Proteomics, Academy of Military Medical Sciences, Academy of Military Sciences, Beijing 100071, China
| | - Yu Lan
- Key Laboratory for Regenerative Medicine of Ministry of Education, Institute of Hematology, School of Medicine, Jinan University, Guangzhou 510632, China
| | - Bing Liu
- State Key Laboratory of Proteomics, Academy of Military Medical Sciences, Academy of Military Sciences, Beijing 100071, China; Key Laboratory for Regenerative Medicine of Ministry of Education, Institute of Hematology, School of Medicine, Jinan University, Guangzhou 510632, China; State Key Laboratory of Experimental Hematology, Fifth Medical Center of Chinese PLA General Hospital, Beijing 100071, China
| | - Huiyuan Zhang
- Department of Rheumatology and Immunology, National Clinical Research Center for Geriatrics, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Collaborative Innovation Center for Biotherapy, Chengdu 610041, China.
| | - Hongbo Hu
- Department of Rheumatology and Immunology, National Clinical Research Center for Geriatrics, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Collaborative Innovation Center for Biotherapy, Chengdu 610041, China.
| |
Collapse
|
9
|
Zhou W, Yui MA, Williams BA, Yun J, Wold BJ, Cai L, Rothenberg EV. Single-Cell Analysis Reveals Regulatory Gene Expression Dynamics Leading to Lineage Commitment in Early T Cell Development. Cell Syst 2019; 9:321-337.e9. [PMID: 31629685 PMCID: PMC6932747 DOI: 10.1016/j.cels.2019.09.008] [Citation(s) in RCA: 67] [Impact Index Per Article: 11.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2019] [Revised: 08/10/2019] [Accepted: 09/18/2019] [Indexed: 02/04/2023]
Abstract
Intrathymic T cell development converts multipotent precursors to committed pro-T cells, silencing progenitor genes while inducing T cell genes, but the underlying steps have remained obscure. Single-cell profiling was used to define the order of regulatory changes, employing single-cell RNA sequencing (scRNA-seq) for full-transcriptome analysis, plus sequential multiplexed single-molecule fluorescent in situ hybridization (seqFISH) to quantitate functionally important transcripts in intrathymic precursors. Single-cell cloning verified high T cell precursor frequency among the immunophenotypically defined "early T cell precursor" (ETP) population; a discrete committed granulocyte precursor subset was also distinguished. We established regulatory phenotypes of sequential ETP subsets, confirmed initial co-expression of progenitor with T cell specification genes, defined stage-specific relationships between cell cycle and differentiation, and generated a pseudotime model from ETP to T lineage commitment, supported by RNA velocity and transcription factor perturbations. This model was validated by developmental kinetics of ETP subsets at population and clonal levels. The results imply that multilineage priming is integral to T cell specification.
Collapse
Affiliation(s)
- Wen Zhou
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA 91125, USA
| | - Mary A Yui
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA 91125, USA
| | - Brian A Williams
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA 91125, USA
| | - Jina Yun
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA 91125, USA
| | - Barbara J Wold
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA 91125, USA
| | - Long Cai
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA 91125, USA
| | - Ellen V Rothenberg
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA 91125, USA.
| |
Collapse
|
10
|
Klein F, Mitrovic M, Roux J, Engdahl C, von Muenchow L, Alberti-Servera L, Fehling HJ, Pelczar P, Rolink A, Tsapogas P. The transcription factor Duxbl mediates elimination of pre-T cells that fail β-selection. J Exp Med 2019; 216:638-655. [PMID: 30765463 PMCID: PMC6400535 DOI: 10.1084/jem.20181444] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2018] [Revised: 12/13/2018] [Accepted: 01/22/2019] [Indexed: 12/13/2022] Open
Abstract
During β-selection, T cells without productive TCRβ rearrangements are eliminated. Klein et al. show that the transcription factor Duxbl regulates this process by inducing apoptosis through activation of the Oas/RNaseL pathway. Successful TCRβ rearrangement rescues cells by pre-TCR–mediated Duxbl suppression. T cell development is critically dependent on successful rearrangement of antigen-receptor chains. At the β-selection checkpoint, only cells with a functional rearrangement continue in development. However, how nonselected T cells proceed in their dead-end fate is not clear. We identified low CD27 expression to mark pre-T cells that have failed to rearrange their β-chain. Expression profiling and single-cell transcriptome clustering identified a developmental trajectory through β-selection and revealed specific expression of the transcription factor Duxbl at a stage of high recombination activity before β-selection. Conditional transgenic expression of Duxbl resulted in a developmental block at the DN3-to-DN4 transition due to reduced proliferation and enhanced apoptosis, whereas RNA silencing of Duxbl led to a decrease in apoptosis. Transcriptome analysis linked Duxbl to elevated expression of the apoptosis-inducing Oas/RNaseL pathway. RNaseL deficiency or sustained Bcl2 expression led to a partial rescue of cells in Duxbl transgenic mice. These findings identify Duxbl as a regulator of β-selection by inducing apoptosis in cells with a nonfunctional rearrangement.
Collapse
Affiliation(s)
- Fabian Klein
- Developmental and Molecular Immunology, Department of Biomedicine, University of Basel, Basel, Switzerland
| | - Mladen Mitrovic
- Immune Regulation, Department of Biomedicine, University of Basel, Basel, Switzerland
| | - Julien Roux
- Bioinformatics Core Facility, Department of Biomedicine, University of Basel, Basel, Switzerland.,Swiss Institute of Bioinformatics, Lausanne, Switzerland
| | - Corinne Engdahl
- Developmental and Molecular Immunology, Department of Biomedicine, University of Basel, Basel, Switzerland
| | - Lilly von Muenchow
- Developmental and Molecular Immunology, Department of Biomedicine, University of Basel, Basel, Switzerland
| | - Llucia Alberti-Servera
- Developmental and Molecular Immunology, Department of Biomedicine, University of Basel, Basel, Switzerland
| | | | - Pawel Pelczar
- Center for Transgenic Models, University of Basel, Basel, Switzerland
| | - Antonius Rolink
- Developmental and Molecular Immunology, Department of Biomedicine, University of Basel, Basel, Switzerland
| | - Panagiotis Tsapogas
- Developmental and Molecular Immunology, Department of Biomedicine, University of Basel, Basel, Switzerland
| |
Collapse
|
11
|
Zhang X, Huo L, Song L, Hu Z, Wang X, Han Y, Wang Y, Xu P, Zhang J, Hua ZC. Dominant Negative FADD/MORT1 Inhibits the Development of Intestinal Intraepithelial Lymphocytes With a Marked Defect on CD8αα+TCRγδ+ T Cells. Front Immunol 2018; 9:2038. [PMID: 30250469 PMCID: PMC6139313 DOI: 10.3389/fimmu.2018.02038] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2018] [Accepted: 08/17/2018] [Indexed: 11/13/2022] Open
Abstract
Intestinal intraepithelial lymphocytes (IELs) play a critical role in mucosal immune system, which differ from thymus-derived cells and develop locally in gut. Although the development of IELs has been studied in some detail, the molecular cues controlling their local development remain unclear. Here, we demonstrate that FADD, a classic adaptor protein required for death-receptor-induced apoptosis, is a critical regulator of the intestinal IEL development. The mice with a dominant negative mutant of FADD (FADD-DN) display an abnormal development of intestinal IELs with a marked reduction in the numbers of CD8αα+TCRγδ+ T cells. As a precursor for CD8αα+ development, lamina propria lymphocytes in lin-negative expression (lin- LPLs) were analyzed and the massive accumulation of IL-7R-lin- LPLs was observed in FADD-DN mice. As IL-7R is one of Notch1-target genes, we further observed that the level of Notch1 expression was lower in Lin- LPLs from FADD-DN mice compared with normal mice. The downregulation of Notch1 expression induced by FADD-DN overexpression was also confirmed in Jurkat T cells. Considering that IL-7 and its receptor IL7-R play a differentiation inducing role in the development of intestinal IELs, the influence of FADD via its DD domain on Notch1 expression might be a possible molecular signal involved in the early IELs development. In addition, loss of γδ T-IELs in FADD-DN mice aggravates DSS-induced colitis, suggesting that FADD is a relevant contribution to the field of mucosal immunology and intestinal homeostasis.
Collapse
Affiliation(s)
- Xuerui Zhang
- The State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Nanjing University, Nanjing, China.,Changzhou High-Tech Research Institute of Nanjing University and Jiangsu Target Pharma Laboratories Inc., Changzhou, China
| | - Lina Huo
- The State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Nanjing University, Nanjing, China
| | - Lulu Song
- The State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Nanjing University, Nanjing, China
| | - Zhaoqing Hu
- The State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Nanjing University, Nanjing, China
| | - Xinran Wang
- The State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Nanjing University, Nanjing, China
| | - Yuheng Han
- The State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Nanjing University, Nanjing, China
| | - Ying Wang
- The State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Nanjing University, Nanjing, China.,Changzhou High-Tech Research Institute of Nanjing University and Jiangsu Target Pharma Laboratories Inc., Changzhou, China
| | - Peipei Xu
- Department of Hematology, Drum Tower Hospital, School of Medicine, Nanjing University, Nanjing, China
| | - Jing Zhang
- The State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Nanjing University, Nanjing, China.,Changzhou High-Tech Research Institute of Nanjing University and Jiangsu Target Pharma Laboratories Inc., Changzhou, China
| | - Zi-Chun Hua
- The State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Nanjing University, Nanjing, China.,Changzhou High-Tech Research Institute of Nanjing University and Jiangsu Target Pharma Laboratories Inc., Changzhou, China.,Shenzhen Research Institute of Nanjing University, Shenzhen, China
| |
Collapse
|
12
|
Krueger A. Thymus Colonization: Who, How, How Many? Arch Immunol Ther Exp (Warsz) 2017; 66:81-88. [PMID: 29288431 DOI: 10.1007/s00005-017-0503-5] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2017] [Accepted: 11/23/2017] [Indexed: 11/28/2022]
Abstract
De novo generation of T cells depends on continual colonization of the thymus by bone marrow-derived progenitors. Thymus seeding progenitors (TSPs) constitute a heterogeneous population comprising multipotent and lineage-restricted cell types. Entry into the thymic microenvironment is tightly controlled and recent quantitative studies have revealed that the adult murine thymus only contains approximately 160 niches to accommodate TSPs. Of these niches only about 6% are open for seeding on average at steady-state. Here, I review the state of understanding of colonization of the adult murine thymus with a particular focus on past and current controversies in the field. Improving thymus colonization and/or maintaining intact TSP niches during the course of pre-conditioning regimens are likely to be critical for efficient T-cell regeneration after hematopoietic stem cell transplantation.
Collapse
Affiliation(s)
- Andreas Krueger
- Institute for Molecular Medicine, Goethe University Frankfurt am Main, 60590, Frankfurt am Main, Germany.
| |
Collapse
|
13
|
Mainardi C, Ebinger M, Enkel S, Feuchtinger T, Teltschik HM, Eyrich M, Schumm M, Rabsteyn A, Schlegel P, Seitz C, Schwarze CP, Müller I, Greil J, Bader P, Schlegel PG, Martin D, Holzer U, Döring M, Handgretinger R, Lang P. CD34 + selected stem cell boosts can improve poor graft function after paediatric allogeneic stem cell transplantation. Br J Haematol 2017; 180:90-99. [PMID: 29205259 DOI: 10.1111/bjh.15012] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2017] [Accepted: 09/19/2017] [Indexed: 12/13/2022]
Abstract
Poor graft function (PGF) is a severe complication of haematopoietic stem cell transplantation (HSCT) and administration of donor stem cell boosts (SCBs) represents a therapeutic option. We report 50 paediatric patients with PGF who received 61 boosts with CD34+ selected peripheral blood stem cells (PBSC) after transplantation from matched unrelated (n = 25) or mismatched related (n = 25) donors. Within 8 weeks, a significant increase in median neutrophil counts (0·6 vs. 1·516 × 109 /l, P < 0·05) and a decrease in red blood cell and platelet transfusion requirement (median frequencies 1 and 7 vs. 0, P < 0·0001 and <0·001), were observed, and 78·8% of patients resolved one or two of their cytopenias. 36·5% had a complete haematological response. Median lymphocyte counts for CD3+ , CD3+ CD4+ , CD19+ and CD56+ increased 8·3-, 14·2-, 22.- and 1·6-fold. The rate of de novo acute graft-versus-host disease (GvHD) grade I-III was only 6% and resolved completely. No GvHD grade IV or chronic GvHD occurred. Patients who responded to SCB displayed a trend toward better overall survival (OS) (P = 0·07). Thus, administration of CD34+ selected SCBs from alternative donors is safe and effective. Further studies are warranted to clarify the impact on immune reconstitution and survival.
Collapse
Affiliation(s)
- Chiara Mainardi
- Department of Paediatric Oncology, Children's University Hospital, University of Padova, Padova, Italy.,Department of Paediatric Haematology/Oncology, Children's University Hospital, Tübingen, Germany
| | - Martin Ebinger
- Department of Paediatric Haematology/Oncology, Children's University Hospital, Tübingen, Germany
| | - Sigrid Enkel
- Transfusion Medicine Department, Tübingen University Hospital, University of Tübingen, Tübingen, Germany
| | - Tobias Feuchtinger
- Dr. von Hauner Children's Hospital, Ludwig Maximilians University, Munich, Germany
| | - Heiko-Manuel Teltschik
- Department of Paediatric Haematology/Oncology, Children's University Hospital, Tübingen, Germany
| | - Matthias Eyrich
- Department of Paediatric Oncology, University Children's Hospital, University of Würzburg, Würzburg, Germany
| | - Michael Schumm
- Department of Paediatric Haematology/Oncology, Children's University Hospital, Tübingen, Germany
| | - Armin Rabsteyn
- Department of Paediatric Haematology/Oncology, Children's University Hospital, Tübingen, Germany
| | - Patrick Schlegel
- Department of Paediatric Haematology/Oncology, Children's University Hospital, Tübingen, Germany
| | - Christian Seitz
- Department of Paediatric Haematology/Oncology, Children's University Hospital, Tübingen, Germany
| | - Carl-Phillip Schwarze
- Department of Paediatric Haematology/Oncology, Children's University Hospital, Tübingen, Germany
| | - Ingo Müller
- Department of Paediatric Haematology and Oncology, University Hospital Eppendorf, Hamburg, Germany
| | - Johann Greil
- Department of Paediatric Oncology, Haematology and Immunology, University of Heidelberg, Heidelberg, Germany
| | - Peter Bader
- Clinic for Paediatric and Adolescent Medicine, University Hospital Frankfurt, Frankfurt/Main, Germany
| | - Paul-Gerhardt Schlegel
- Department of Paediatric Oncology, University Children's Hospital, University of Würzburg, Würzburg, Germany
| | - David Martin
- Department of Paediatric Haematology/Oncology, Children's University Hospital, Tübingen, Germany.,Filderklinik, Filderstadt-Bonlanden, Germany
| | - Ursula Holzer
- Department of Paediatric Haematology/Oncology, Children's University Hospital, Tübingen, Germany
| | - Michaela Döring
- Department of Paediatric Haematology/Oncology, Children's University Hospital, Tübingen, Germany
| | - Rupert Handgretinger
- Department of Paediatric Haematology/Oncology, Children's University Hospital, Tübingen, Germany
| | - Peter Lang
- Department of Paediatric Haematology/Oncology, Children's University Hospital, Tübingen, Germany
| |
Collapse
|
14
|
Interleukin-7 in the transition of bone marrow progenitors to the thymus. Immunol Cell Biol 2017; 95:916-924. [PMID: 28811625 DOI: 10.1038/icb.2017.68] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2016] [Revised: 08/01/2017] [Accepted: 08/01/2017] [Indexed: 02/03/2023]
Abstract
Interleukin-7 (IL-7) is essential for the development of T cells in humans and mice where deficiencies in IL-7 signaling result in severe immunodeficiency. T cells require IL-7 at multiple points during development; however, it is unclear when IL-7 is first necessary. We observed that mice with impaired IL-7 signaling had a large reduction in the number of early thymic progenitors (ETPs) while mice that overexpress IL-7 had greatly increased numbers of ETPs. These results indicated that the development of ETPs is sensitive to IL-7. Bone marrow progenitors of ETP are present in normal numbers in mice with impaired IL-7 signaling (IL-7Rα449F) and were efficiently recruited to the thymus. Furthermore, ETPs and their progenitors from IL-7Rα449F mice did not undergo increased apoptosis and proliferate normally compared to WT cells. Mixed bone marrow chimeras demonstrated that IL-7 signaling has a cell-intrinsic role in ETP development but was not required for development of bone marrow progenitors. We have shown a novel role for IL-7 signaling in the development of ETPs that is distinct from classic mechanisms of IL-7 regulating survival and proliferation.
Collapse
|
15
|
Krueger A, Ziętara N, Łyszkiewicz M. T Cell Development by the Numbers. Trends Immunol 2016; 38:128-139. [PMID: 27842955 DOI: 10.1016/j.it.2016.10.007] [Citation(s) in RCA: 55] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2016] [Revised: 10/21/2016] [Accepted: 10/25/2016] [Indexed: 01/01/2023]
Abstract
T cells are continually generated in the thymus in a highly dynamic process comprising discrete steps of lineage commitment, T cell receptor (TCR) gene rearrangement, and selection. These steps are linked to distinct rates of proliferation, survival, and cell death, but a quantitative picture of T cell development is only beginning to emerge. Here we summarize recent technical advances, including genetic fate mapping, barcoding, and molecular timers, that have allowed the implementation of computational models to quantify developmental dynamics in the thymus. Coupling new techniques with mathematical models has recently resulted in the emergence of new paradigms in early hematopoiesis and might similarly open new perspectives on T cell development.
Collapse
Affiliation(s)
- Andreas Krueger
- Institute of Molecular Medicine, Goethe University Frankfurt am Main, 60590 Frankfurt am Main, Germany.
| | - Natalia Ziętara
- Dr von Hauner Children's Hospital, Ludwig Maximilian University, 80337 Munich, Germany
| | - Marcin Łyszkiewicz
- Dr von Hauner Children's Hospital, Ludwig Maximilian University, 80337 Munich, Germany
| |
Collapse
|
16
|
Golec DP, Henao Caviedes LM, Baldwin TA. RasGRP1 and RasGRP3 Are Required for Efficient Generation of Early Thymic Progenitors. THE JOURNAL OF IMMUNOLOGY 2016; 197:1743-53. [PMID: 27465532 DOI: 10.4049/jimmunol.1502107] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/29/2015] [Accepted: 06/28/2016] [Indexed: 11/19/2022]
Abstract
T cell development is dependent on the migration of progenitor cells from the bone marrow to the thymus. Upon reaching the thymus, progenitors undergo a complex developmental program that requires inputs from various highly conserved signaling pathways including the Notch and Wnt pathways. To date, Ras signaling has not been implicated in the very earliest stages of T cell differentiation, but members of a family of Ras activators called RasGRPs have been shown to be involved at multiple stages of T cell development. We examined early T cell development in mice lacking RasGRP1, RasGRP3, and RasGRPs 1 and 3. We report that RasGRP1- and RasGRP3-deficient thymi show significantly reduced numbers of early thymic progenitors (ETPs) relative to wild type thymi. Furthermore, RasGRP1/3 double-deficient thymi show significant reductions in ETP numbers compared with either RasGRP1 or RasGRP3 single-deficient thymi, suggesting that both RasGRP1 and RasGRP3 regulate the generation of ETPs. In addition, competitive bone marrow chimera experiments reveal that RasGRP1/3 double-deficient progenitors intrinsically generate ETPs less efficiently than wild type progenitors. Finally, RasGRP1/3-deficient progenitors show impaired migration toward the CCR9 ligand, CCL25, suggesting that RasGRP1 and RasGRP3 may regulate progenitor entry into the thymus through a CCR9-dependent mechanism. These data demonstrate that, in addition to Notch and Wnt, the highly conserved Ras pathway is critical for the earliest stages of T cell development and further highlight the importance of Ras signaling during thymocyte maturation.
Collapse
Affiliation(s)
- Dominic P Golec
- Department of Medical Microbiology and Immunology, University of Alberta, Edmonton, Alberta T6G 2S2, Canada
| | - Laura M Henao Caviedes
- Department of Medical Microbiology and Immunology, University of Alberta, Edmonton, Alberta T6G 2S2, Canada
| | - Troy A Baldwin
- Department of Medical Microbiology and Immunology, University of Alberta, Edmonton, Alberta T6G 2S2, Canada
| |
Collapse
|
17
|
Rothenberg EV, Ungerbäck J, Champhekar A. Forging T-Lymphocyte Identity: Intersecting Networks of Transcriptional Control. Adv Immunol 2015; 129:109-74. [PMID: 26791859 DOI: 10.1016/bs.ai.2015.09.002] [Citation(s) in RCA: 53] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
T-lymphocyte development branches off from other lymphoid developmental programs through its requirement for sustained environmental signals through the Notch pathway. In the thymus, Notch signaling induces a succession of T-lineage regulatory factors that collectively create the T-cell identity through distinct steps. This process involves both the staged activation of T-cell identity genes and the staged repression of progenitor-cell-inherited regulatory genes once their roles in self-renewal and population expansion are no longer needed. With the recent characterization of innate lymphoid cells (ILCs) that share transcriptional regulation programs extensively with T-cell subsets, T-cell identity can increasingly be seen as defined in modular terms, as the processes selecting and actuating effector function are potentially detachable from the processes generating and selecting clonally unique T-cell receptor structures. The developmental pathways of different classes of T cells and ILCs are distinguished by the numbers of prerequisites of gene rearrangement, selection, and antigen contact before the cells gain access to nearly common regulatory mechanisms for choosing effector function. Here, the major classes of transcription factors that interact with Notch signals during T-lineage specification are discussed in terms of their roles in these programs, the evidence for their spectra of target genes at different stages, and their cross-regulatory and cooperative actions with each other. Specific topics include Notch modulation of PU.1 and GATA-3, PU.1-Notch competition, the relationship between PU.1 and GATA-3, and the roles of E proteins, Bcl11b, and GATA-3 in guiding acquisition of T-cell identity while avoiding redirection to an ILC fate.
Collapse
Affiliation(s)
- Ellen V Rothenberg
- Division of Biology & Biological Engineering, California Institute of Technology, Pasadena, California, USA.
| | - Jonas Ungerbäck
- Division of Biology & Biological Engineering, California Institute of Technology, Pasadena, California, USA; Department of Clinical and Experimental Medicine, Experimental Hematopoiesis Unit, Faculty of Health Sciences, Linköping University, Linköping, Sweden
| | - Ameya Champhekar
- Department of Microbiology, Immunology, and Molecular Genetics, University of California Los Angeles, Los Angeles, California, USA
| |
Collapse
|
18
|
Regelin M, Blume J, Pommerencke J, Vakilzadeh R, Witzlau K, Łyszkiewicz M, Ziętara N, Saran N, Schambach A, Krueger A. Responsiveness of Developing T Cells to IL-7 Signals Is Sustained by miR-17∼92. THE JOURNAL OF IMMUNOLOGY 2015; 195:4832-40. [PMID: 26475928 DOI: 10.4049/jimmunol.1402248] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/02/2014] [Accepted: 09/04/2015] [Indexed: 12/13/2022]
Abstract
miRNAs regulate a large variety of developmental processes including development of the immune system. T cell development is tightly controlled through the interplay of transcriptional programs and cytokine-mediated signals. However, the role of individual miRNAs in this process remains largely elusive. In this study, we demonstrated that hematopoietic cell-specific loss of miR-17∼92, a cluster of six miRNAs implicated in B and T lineage leukemogenesis, resulted in profound defects in T cell development both at the level of prethymic T cell progenitors as well as intrathymically. We identified reduced surface expression of IL-7R and concomitant limited responsiveness to IL-7 signals as a common mechanism resulting in reduced cell survival of common lymphoid progenitors and thymocytes at the double-negative to double-positive transition. In conclusion, we identified miR-17∼92 as a critical modulator of multiple stages of T cell development.
Collapse
Affiliation(s)
- Malte Regelin
- Institute of Immunology, Hannover Medical School, 30625 Hannover, Germany
| | - Jonas Blume
- Institute of Immunology, Hannover Medical School, 30625 Hannover, Germany
| | - Jens Pommerencke
- Institute of Immunology, Hannover Medical School, 30625 Hannover, Germany
| | - Ramin Vakilzadeh
- Institute of Immunology, Hannover Medical School, 30625 Hannover, Germany
| | - Katrin Witzlau
- Institute of Immunology, Hannover Medical School, 30625 Hannover, Germany
| | - Marcin Łyszkiewicz
- Institute of Immunology, Hannover Medical School, 30625 Hannover, Germany
| | - Natalia Ziętara
- Institute of Immunology, Hannover Medical School, 30625 Hannover, Germany
| | - Namita Saran
- Institute of Immunology, Hannover Medical School, 30625 Hannover, Germany
| | - Axel Schambach
- Institute of Experimental Hematology, Hannover Medical School, 30625 Hannover, Germany; and Division of Hematology and Oncology, Boston Children's Hospital, Harvard Medical School, Boston, MA 02115
| | - Andreas Krueger
- Institute of Immunology, Hannover Medical School, 30625 Hannover, Germany;
| |
Collapse
|
19
|
Ziętara N, Łyszkiewicz M, Puchałka J, Witzlau K, Reinhardt A, Förster R, Pabst O, Prinz I, Krueger A. Multicongenic fate mapping quantification of dynamics of thymus colonization. ACTA ACUST UNITED AC 2015; 212:1589-601. [PMID: 26347471 PMCID: PMC4577840 DOI: 10.1084/jem.20142143] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2014] [Accepted: 08/10/2015] [Indexed: 12/30/2022]
Abstract
Ziętara et al demonstrate with multicongenic fate mapping that thymus seeding is directly restricted to the duration of niche occupancy rather than long-range effects. Postnatal T cell development depends on continuous colonization of the thymus by BM-derived T lineage progenitors. Both quantitative parameters and the mechanisms of thymus seeding remain poorly understood. Here, we determined the number of dedicated thymus-seeding progenitor niches (TSPNs) capable of supporting productive T cell development, turnover rates of niche occupancy, and feedback mechanisms. To this end, we established multicongenic fate mapping combined with mathematical modeling to quantitate individual events of thymus colonization. We applied this method to study thymus colonization in CCR7−/−CCR9−/− (DKO) mice, whose TSPNs are largely unoccupied. We showed that ∼160–200 TSPNs are present in the adult thymus and, on average, 10 of these TSPNs were open for recolonization at steady state. Preconditioning of wild-type mice revealed a similar number of TSPNs, indicating that preconditioning can generate space efficiently for transplanted T cell progenitors. To identify potential cellular feedback loops restricting thymus colonization, we performed serial transfer experiments. These experiments indicated that thymus seeding was directly restricted by the duration of niche occupancy rather than long-range effects, thus challenging current paradigms of thymus colonization.
Collapse
Affiliation(s)
- Natalia Ziętara
- Institute of Immunology, Hannover Medical School, D-30625 Hannover, Germany
| | - Marcin Łyszkiewicz
- Institute of Immunology, Hannover Medical School, D-30625 Hannover, Germany
| | - Jacek Puchałka
- Dr. von Haunersches Kinderspital, University Children's Hospital, Ludwig Maximilian University, D-80337 Munich, Germany
| | - Katrin Witzlau
- Institute of Immunology, Hannover Medical School, D-30625 Hannover, Germany
| | - Annika Reinhardt
- Institute of Immunology, Hannover Medical School, D-30625 Hannover, Germany
| | - Reinhold Förster
- Institute of Immunology, Hannover Medical School, D-30625 Hannover, Germany
| | - Oliver Pabst
- Institute of Immunology, Hannover Medical School, D-30625 Hannover, Germany Institute of Molecular Medicine, RWTH Aachen University, D-52074 Aachen, Germany
| | - Immo Prinz
- Institute of Immunology, Hannover Medical School, D-30625 Hannover, Germany
| | - Andreas Krueger
- Institute of Immunology, Hannover Medical School, D-30625 Hannover, Germany
| |
Collapse
|
20
|
Bose TO, Colpitts SL, Pham QM, Puddington L, Lefrançois L. CD11a is essential for normal development of hematopoietic intermediates. THE JOURNAL OF IMMUNOLOGY 2014; 193:2863-72. [PMID: 25108025 DOI: 10.4049/jimmunol.1301820] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
The process of lymphopoiesis begins in the bone marrow (BM) and requires multiple cellular intermediates. For T cell production, lymphoid progenitors exit the BM and home to the thymus where maturation and selection ensue. These processes are dependent on a number of factors, including chemokines and adhesion molecules. Although the β2 integrin CD11a plays an important role in the migration of lymphocytes to lymph nodes, the role of CD11a in T cell development is largely undefined. Our studies now show that, in CD11a(-/-) mice, thymic cellularity was decreased and early T cell development was partially impaired. Remarkably, CD11a was critical for generation of common lymphoid progenitors (CLPs) and lymphoid-primed multipotent progenitors. However, in intact CD11a(-/-) mice, peripheral B and T cell subsets were only modestly altered, suggesting that compensatory mechanisms were operating. In contrast, competitive BM-reconstitution assays revealed an essential role for CD11a in the generation of thymocytes and mature T and B cells. This defect was linked to the requirement for CD11a in the development of CLPs. Furthermore, our results identified CLPs, and not lymphoid-primed multipotent progenitors, as the requisite CD11a-dependent precursor for lymphocyte development. Thus, these findings established a key role for CD11a in lymphopoiesis.
Collapse
Affiliation(s)
- Tina O Bose
- Department of Immunology, Center for Integrated Immunology and Vaccine Research, University of Connecticut Health Center, Farmington, CT 06030
| | - Sara L Colpitts
- Department of Immunology, Center for Integrated Immunology and Vaccine Research, University of Connecticut Health Center, Farmington, CT 06030
| | - Quynh-Mai Pham
- Department of Immunology, Center for Integrated Immunology and Vaccine Research, University of Connecticut Health Center, Farmington, CT 06030
| | - Lynn Puddington
- Department of Immunology, Center for Integrated Immunology and Vaccine Research, University of Connecticut Health Center, Farmington, CT 06030
| | - Leo Lefrançois
- Department of Immunology, Center for Integrated Immunology and Vaccine Research, University of Connecticut Health Center, Farmington, CT 06030
| |
Collapse
|
21
|
Zhang SL, Wang X, Manna S, Zlotoff DA, Bryson JL, Blazar BR, Bhandoola A. Chemokine treatment rescues profound T-lineage progenitor homing defect after bone marrow transplant conditioning in mice. Blood 2014; 124:296-304. [PMID: 24876562 PMCID: PMC4093685 DOI: 10.1182/blood-2014-01-552794] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2014] [Accepted: 05/22/2014] [Indexed: 12/13/2022] Open
Abstract
Development of T cells in the thymus requires continuous importation of T-lineage progenitors from the bone marrow via the circulation. Following bone marrow transplant, recovery of a normal peripheral T-cell pool depends on production of naïve T cells in the thymus; however, delivery of progenitors to the thymus limits T-lineage reconstitution. Here, we examine homing of intravenously delivered progenitors to the thymus following irradiation and bone marrow reconstitution. Surprisingly, following host conditioning by irradiation, we find that homing of lymphoid-primed multipotent progenitors and common lymphoid progenitors to the thymus decreases more than 10-fold relative to unirradiated mice. The reduction in thymic homing in irradiated mice is accompanied by a significant reduction in CCL25, an important chemokine ligand for thymic homing. We show that pretreatment of bone marrow progenitors with CCL25 and CCL21 corrects the defect in thymic homing after irradiation and promotes thymic reconstitution. These data suggest new therapeutic approaches to promote T-cell regeneration.
Collapse
Affiliation(s)
- Shirley L Zhang
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA; and
| | - Xinxin Wang
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA; and
| | - Sugata Manna
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA; and
| | - Daniel A Zlotoff
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA; and
| | - Jerrod L Bryson
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA; and
| | - Bruce R Blazar
- Department of Pediatrics, Division of Blood and Marrow Transplantation, University of Minnesota, Minneapolis, MN
| | - Avinash Bhandoola
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA; and
| |
Collapse
|
22
|
Image-guided intrathymic injection of multipotent stem cells supports lifelong T-cell immunity and facilitates targeted immunotherapy. Blood 2014; 123:2797-805. [PMID: 24652996 DOI: 10.1182/blood-2013-10-535401] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
T-cell deficiency related to disease, medical treatment, or aging represents a major clinical challenge and is associated with significant morbidity and mortality in cancer and bone marrow transplantation recipients. This study describes several innovative and clinically relevant strategies to manipulate thymic function based on an interventional radiology technique for intrathymic injection of cells or drugs. We show that intrathymic injection of multipotent hematopoietic stem/progenitor cells into irradiated syngeneic or allogeneic young or aged recipients resulted in efficient and long-lasting generation of functional donor T cells. Persistence of intrathymic donor cells was associated with intrathymic presence of cells resembling long-term hematopoietic stem cells, suggesting a self-renewal capacity of the intrathymically injected cells. Furthermore, our approach enabled the induction of long-term antigen-specific T-cell-mediated antitumor immunity following intrathymic injection of progenitor cells harboring a transgenic T-cell receptor gene. The intrathymic injection of interleukin-7 prior to irradiation conferred radioprotection. In addition, thymopoiesis of aged mice improved with a single intrathymic administration of low-dose keratinocyte growth factor, an effect that was sustained even in the setting of radiation-induced injury. Taken together, we established a preclinical framework for the development of novel clinical protocols to establish lifelong antigen-specific T-cell immunity.
Collapse
|
23
|
Abstract
T and B cells share a common somatic gene rearrangement mechanism for assembling the genes that code for their antigen receptors; they also have developmental pathways with many parallels. Shared usage of basic helix-loop-helix E proteins as transcriptional drivers underlies these common features. However, the transcription factor networks in which these E proteins are embedded are different both in membership and in architecture for T and B cell gene regulatory programs. These differences permit lineage commitment decisions to be made in different hierarchical orders. Furthermore, in contrast to B cell gene networks, the T cell gene network architecture for effector differentiation is sufficiently modular so that E protein inputs can be removed. Complete T cell-like effector differentiation can proceed without T cell receptor rearrangement or selection when E proteins are neutralized, yielding natural killer and other innate lymphoid cells.
Collapse
Affiliation(s)
- Ellen V Rothenberg
- Division of Biology, California Institute of Technology, Pasadena, California 91125;
| |
Collapse
|
24
|
Kyoizumi S, Kubo Y, Kajimura J, Yoshida K, Imai K, Hayashi T, Nakachi K, Young LF, Moore MA, van den Brink MRM, Kusunoki Y. Age-associated changes in the differentiation potentials of human circulating hematopoietic progenitors to T- or NK-lineage cells. THE JOURNAL OF IMMUNOLOGY 2013; 190:6164-72. [PMID: 23670190 DOI: 10.4049/jimmunol.1203189] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
Age-associated changes of T and NK cell (T/NK) potential of human hematopoietic stem cells are unknown. In this study, we enumerate and characterize T/NK precursors among CD34(+)Lin(-) cell populations circulating in normal human adult peripheral blood (PB) by a limiting-dilution assay using coculture with OP9-DL1 stroma cells expressing Notch 1 ligand, Delta-like 1. The frequency of T cell precursors in CD34(+)Lin(-) cells was found to decrease with donor age, whereas the ratio of NK to T cell precursor frequency (NK/T ratio) increased with age, suggesting that lymphoid differentiation potential of PB progenitors shifts from T to NK cell lineage with aging. Clonal analyses of CD34(+)Lin(-) cells showed that differences in the NK/T ratio were attributable to different distributions of single- and dual-lineage T/NK precursor clones. Because nearly all of the clones retained monocyte and/or granulocyte differentiation potentials in coculture with OP9-DL1 cells, T/NK precursors in PB are considered to be contained in the pool of T/NK/myeloid multipotent progenitors. The age-associated increase in NK over T cell commitment might occur in precursor cells with T/NK/myeloid potential.
Collapse
Affiliation(s)
- Seishi Kyoizumi
- Department of Radiobiology/Molecular Epidemiology, Radiation Effects Research Foundation, Hiroshima 732-0815, Japan.
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
25
|
Luche H, Nageswara Rao T, Kumar S, Tasdogan A, Beckel F, Blum C, Martins VC, Rodewald HR, Fehling HJ. In vivo fate mapping identifies pre-TCRα expression as an intra- and extrathymic, but not prethymic, marker of T lymphopoiesis. ACTA ACUST UNITED AC 2013; 210:699-714. [PMID: 23509324 PMCID: PMC3620354 DOI: 10.1084/jem.20122609] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2023]
Abstract
A novel pre-TCRα (pTα) reporter mouse reveals that expression of pTα is confined to the T lineage and does not occur on prethymic progenitors. Expression of the pre–T cell receptor α (pTα) gene has been exploited in previous studies as a molecular marker to identify tiny cell populations in bone marrow (BM) and blood that were suggested to contain physiologically relevant thymus settling progenitors (TSPs). But to what extent these cells genuinely contribute to thymopoiesis has remained obscure. We have generated a novel pTαiCre knockin mouse line and performed lineage-tracing experiments to precisely quantitate the contribution of pTα-expressing progenitors to distinct differentiation pathways and to the genealogy of mature hematopoietic cells under physiological in vivo conditions. Using these mice in combination with fluorescent reporter strains, we observe highly consistent labeling patterns that identify pTα expression as a faithful molecular marker of T lineage commitment. Specifically, the fate of pTα-expressing progenitors was found to include all αβ and most γδ T cells but, in contrast to previous assumptions, to exclude B, NK, and thymic dendritic cells. Although we could detect small numbers of T cell progenitors with a history of pTα expression in BM and blood, our data clearly exclude these populations as physiologically important precursors of thymopoiesis and indicate that they instead belong to a pathway of T cell maturation previously defined as extrathymic.
Collapse
Affiliation(s)
- Hervé Luche
- Institute of Immunology, University Clinics Ulm, D-89081 Ulm, Germany
| | | | | | | | | | | | | | | | | |
Collapse
|
26
|
Mendes-da-Cruz DA, Stimamiglio MA, Muñoz JJ, Alfaro D, Terra-Granado E, Garcia-Ceca J, Alonso-Colmenar LM, Savino W, Zapata AG. Developing T-cell migration: role of semaphorins and ephrins. FASEB J 2012; 26:4390-9. [PMID: 22815386 DOI: 10.1096/fj.11-202952] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
Cell migration is a crucial event for normal T-cell development, and various ligand/receptor pairs have been implicated. Most of them, including chemokines and extracellular matrix proteins, have attractant properties on thymocytes. We discuss herein two further groups of ligand/receptor pairs, semaphorins/neuropilins and ephs/ephrins, which are constitutively expressed by thymocytes and thymic microenvironmental cells. Evidence shows that the corresponding interactions are relevant for developing T-cell migration, including the entry of bone marrow progenitor cells, migration of CD4/CD8-defined thymocyte subpopulations triggered by chemokines and/or extracellular matrix proteins, and thymocyte export. Conceptually, the data summarized here show that thymocyte migration results from a complex network of molecular interactions, which generate not only attraction, but also repulsion of migrating T-cell precursors.
Collapse
|
27
|
Abstract
Despite its complexity, blood is probably the best understood developmental system, largely due to seminal experimentation in the mouse. Clinically, hematopoietic stem cell (HSC) transplantation represents the most widely deployed regenerative therapy, but human HSCs have only been characterized relatively recently. The discovery that immune-deficient mice could be engrafted with human cells provided a powerful approach for studying HSCs. We highlight 2 decades of studies focusing on isolation and molecular regulation of human HSCs, therapeutic applications, and early lineage commitment steps, and compare mouse and humanized models to identify both conserved and species-specific mechanisms that will aid future preclinical research.
Collapse
Affiliation(s)
- Sergei Doulatov
- Division of Stem Cell and Developmental Biology, Campbell Family Institute for Cancer Research/Ontario Cancer Institute, Toronto, ON M5G 1L7, Canada
| | | | | | | |
Collapse
|
28
|
Extra-thymic physiological T lineage progenitor activity is exclusively confined to cells expressing either CD127, CD90, or high levels of CD117. PLoS One 2012; 7:e30864. [PMID: 22355330 PMCID: PMC3280270 DOI: 10.1371/journal.pone.0030864] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2011] [Accepted: 12/28/2011] [Indexed: 11/19/2022] Open
Abstract
T cell development depends on continuous recruitment of progenitors from bone marrow (BM) to the thymus via peripheral blood. However, both phenotype and functional characteristics of physiological T cell precursors remain ill-defined. Here, we characterized a putative CD135+CD27+ T cell progenitor population, which lacked expression of CD127, CD90, and high levels of CD117 and was therefore termed triple negative precursor (TNP). TNPs were present in both BM and blood and displayed robust T lineage potential, but virtually no myeloid or B lineage potential, in vitro. However, TNPs did not efficiently generate T lineage progeny after intravenous or intrathymic transfer, suggesting that a physiological thymic microenvironment does not optimally support T cell differentiation from TNPs. Thus, we propose that physiological T cell precursors are confined to populations expressing either CD127, CD90, or high levels of CD117 in addition to CD135 and CD27 and that TNPs may have other physiological functions.
Collapse
|
29
|
|
30
|
Rothenberg EV. T cell lineage commitment: identity and renunciation. THE JOURNAL OF IMMUNOLOGY 2011; 186:6649-55. [PMID: 21646301 DOI: 10.4049/jimmunol.1003703] [Citation(s) in RCA: 59] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Precursors undertaking T cell development shed their access to other pathways in a sequential process that begins before entry into the thymus and continues through many cell cycles afterward. This process involves three levels of regulatory change, in which the cells' intrinsic transcriptional regulatory factors, expression of signaling receptors (e.g., Notch1), and expression of distinct homing receptors separately contribute to confirmation of T cell identity. Each alternative potential has a different underlying molecular basis that is neutralized and then permanently silenced through different mechanisms in early T cell precursors. This regulatory mosaic has notable implications for the hierarchy of relationships linking T lymphocytes to other hematopoietic fates.
Collapse
Affiliation(s)
- Ellen V Rothenberg
- Division of Biology, California Institute of Technology, Pasadena, CA 91125, USA.
| |
Collapse
|
31
|
Thompson PK, Zúñiga-Pflücker JC. On becoming a T cell, a convergence of factors kick it up a Notch along the way. Semin Immunol 2011; 23:350-9. [PMID: 21981947 DOI: 10.1016/j.smim.2011.08.007] [Citation(s) in RCA: 45] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/18/2010] [Accepted: 08/19/2011] [Indexed: 12/18/2022]
Abstract
The thymus is seeded by bone marrow-derived progenitors, which undergo a series of differentiation and proliferation events in order to generate functional T lymphocytes. The Notch signaling pathway, together with multiple transcription factors, act in concert to commit progenitors to a T-lineage fate, extinguishing non-T cell potential, inducing thymocyte differentiation and supporting proliferation and survival along the way to becoming a mature T cell. This review focuses on recent evidence regarding the complex interplay between the Notch pathway and other key transcription factors at specific lineage-decision points during the program of T cell development.
Collapse
Affiliation(s)
- Patrycja K Thompson
- Department of Immunology, University of Toronto, Sunnybrook Research Institute, Toronto, Ontario M4N 3M5, Canada
| | | |
Collapse
|
32
|
Abstract
Negative selection in the thymus prevents the generation of self-reactive T cells through the deletion of thymocytes with high affinity for self-antigens. Within the thymus, self-antigens are presented by thymic epithelial cells and DCs. Both cell types can mediate negative selection, although the relative contribution of each cell type remains elusive. Similar to DCs of other lymphoid organs, thymic DCs come in different flavors. Over the past years, various lines of evidence have emerged that either favor a common origin for some thymic DCs and thymocytes or, conversely, indicate the existence of separate intrathymic T lineage and DC precursors. In this issue of the European Journal of Immunology, a study reports the identification of an intrathymic DC precursor that is likely to be unrelated to the earliest physiological T-cell progenitors. Thus this intrathymic DC precursor may constitute a "missing link" between extrathymic DC precursor-types, which are able to generate DCs in secondary lymphoid organs and intrathymic DCs, and supports the notion that intrathymic DCs and thymocytes arise from different precursors.
Collapse
Affiliation(s)
- Andreas Krueger
- Institute for Immunology, Hannover Medical School, Hannover, Germany.
| |
Collapse
|
33
|
Gui J, Morales AJ, Maxey SE, Bessette KA, Ratcliffe NR, Kelly JA, Craig RW. MCL1 increases primitive thymocyte viability in female mice and promotes thymic expansion into adulthood. Int Immunol 2011; 23:647-59. [PMID: 21937457 DOI: 10.1093/intimm/dxr073] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Increasing the pool of cells at early T-cell developmental stages enhances thymopoiesis and is especially beneficial when T-cell production is compromised by radiation or aging. Within the immature double-negative (DN; CD4(-)CD8(-)) thymocyte subpopulation, the DN1 subset contains the most primitive cells including the rare early T-cell progenitors (ETPs). In the present study, a human MCL1 transgene, under the control of its endogenous promoter, resulted in enlargement of an undistorted thymus in C57/BL6 mice. Enlargement occurred in females but not males, being seen at 1 month of age and maintained during progression into adulthood as the thymus underwent involution. The small DN1 subset was expanded disproportionally (ETPs increasing from ∼0.016 to 0.03% of thymocytes), while more mature thymocytes were increased proportionally (1.5-fold) along with the stroma. DN1 cells from transgenic females exhibited increased viability with maintained proliferation, and their survival in primary culture was extended. Exposure of transgenic females to γ-irradiation also revealed an expanded pool of radioresistant DN1 cells exhibiting increased viability. While the viability of DN1 cells from transgenic males was equivalent to that of their non-transgenic counterparts directly after harvest, it was enhanced in culture-suggesting that the effect of the transgene was suppressed in the in vivo environment of the male. Viability was increased in ETPs from transgenic females, but unchanged in more mature thymocytes, indicating that primitive cells were affected selectively. The MCL1 transgene thus increases the viability and pool size of primitive ETP/DN1 cells, promoting thymopoiesis and radioresistance in peripubescent females and into adulthood.
Collapse
Affiliation(s)
- Jingang Gui
- Department of Pharmacology and Toxicology, Dartmouth Medical School, Hanover, NH 03755, USA
| | | | | | | | | | | | | |
Collapse
|
34
|
Wils EJ, Rombouts EJC, van Mourik I, Spits H, Legrand N, Braakman E, Cornelissen JJ. Stem Cell Factor Consistently Improves Thymopoiesis after Experimental Transplantation of Murine or Human Hematopoietic Stem Cells in Immunodeficient Mice. THE JOURNAL OF IMMUNOLOGY 2011; 187:2974-81. [DOI: 10.4049/jimmunol.1004209] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
|
35
|
Love PE, Bhandoola A. Signal integration and crosstalk during thymocyte migration and emigration. Nat Rev Immunol 2011; 11:469-77. [PMID: 21701522 PMCID: PMC3710714 DOI: 10.1038/nri2989] [Citation(s) in RCA: 153] [Impact Index Per Article: 10.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
The thymus produces self-tolerant functionally competent T cells. This process involves the import of multipotent haematopoietic progenitors that are then signalled to adopt the T cell fate. Expression of T cell-specific genes, including those encoding the T cell receptor (TCR), is followed by positive and negative selection and the eventual export of mature T cells. Significant progress has been made in elucidating the signals that direct progenitor cell trafficking to, within and out of the thymus. These advances are the subject of this Review, with a particular focus on the role of reciprocal cooperative and regulatory interactions between TCR- and chemokine receptor-mediated signalling.
Collapse
Affiliation(s)
- Paul E Love
- Eunice Kennedy Schriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, Maryland, USA.
| | | |
Collapse
|
36
|
Delivery of progenitors to the thymus limits T-lineage reconstitution after bone marrow transplantation. Blood 2011; 118:1962-70. [PMID: 21659540 DOI: 10.1182/blood-2010-12-324954] [Citation(s) in RCA: 53] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
T-cell production depends on the recruitment of hematopoietic progenitors into the thymus. T cells are among the last of the hematopoietic lineages to recover after bone marrow transplantation (BMT), but the reasons for this delay are not well understood. Under normal physiologic conditions, thymic settling is selective and either CCR7 or CCR9 is required for progenitor access into the thymus. The mechanisms of early thymic reconstitution after BMT, however, are unknown. Here we report that thymic settling is briefly CCR7/CCR9-independent after BMT but continues to rely on the selectin ligand PSGL-1. The CCR7/CCR9 independence is transient, and by 3 weeks after BMT these receptors are again strictly required. Despite the normalization of thymic settling signals, the rare bone marrow progenitors that can efficiently repopulate the thymus are poorly reconstituted for at least 4 weeks after BMT. Consistent with reduced progenitor input to the thymus, intrathymic progenitor niches remain unsaturated for at least 10 weeks after BMT. Finally, we show that thymic recovery is limited by the number of progenitors entering the thymus after BMT. Hence, T-lineage reconstitution after BMT is limited by progenitor supply to the thymus.
Collapse
|
37
|
Abstract
Although most hematopoietic lineages develop in the bone marrow (BM), T cells uniquely complete their development in the specialized environment of the thymus. Hematopoietic stem cells with long-term self-renewal capacity are not present in the thymus. As a result, continuous T cell development requires that BM-derived progenitors be imported into the thymus throughout adult life. The process of thymic homing begins with the mobilization of progenitors out of the BM, continues with their circulation in the bloodstream, and concludes with their settling in the thymus. This review will discuss each of these steps as they occur in the unirradiated and postirradiation scenarios, focusing on the molecular mechanisms of regulation. Improved knowledge about these early steps in T cell generation may accelerate the development of new therapeutic options in patients with impaired T cell number or function.
Collapse
Affiliation(s)
- Daniel A Zlotoff
- Department of Pathology and Laboratory Medicine, University of Pennsylvania School of Medicine, Philadelphia, Pennsylvania, USA
| | | |
Collapse
|
38
|
Ichii M, Shimazu T, Welner RS, Garrett KP, Zhang Q, Esplin BL, Kincade PW. Functional diversity of stem and progenitor cells with B-lymphopoietic potential. Immunol Rev 2010; 237:10-21. [PMID: 20727026 DOI: 10.1111/j.1600-065x.2010.00933.x] [Citation(s) in RCA: 40] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Technical advances have made it possible to separate hematopoietic tissues such as the bone marrow into ever smaller populations, complicating our understanding of immune system replenishment. Patterns of surface marker expression and transcription profiles as well as results obtained with reporter mice suggest that lymphopoietic cells are not closely synchronized, and there is considerable cell to cell variation. Loss of differentiation options is gradual, and ultimate fate can be established at different stages of lineage progression. For example, individual hematopoietic stem cells can be biased such that some are very poor sources of lymphocytes as contrasted to ones with balanced outputs. Still other hematopoietic stem cells are effective at generating B and T cells but are defective with respect to expansion and difficult to distinguish from early lymphoid progenitors. That diversity carries forward to later events, and similar appearing cells in the immune system can arise from alternate differentiation pathways. In fact, new categories of lymphoid progenitors are still being discovered. Heterogeneity provides adaptability as hematopoiesis can be dramatically altered during infections, influencing numbers and types of cells that are produced.
Collapse
Affiliation(s)
- Michiko Ichii
- Immunobiology and Cancer Program, Oklahoma Medical Research Foundation, Oklahoma City, OK 73104, USA
| | | | | | | | | | | | | |
Collapse
|
39
|
Braunstein M, Rajkumar P, Claus CL, Vaccarelli G, Moore AJ, Wang D, Anderson MK. HEBAlt enhances the T-cell potential of fetal myeloid-biased precursors. Int Immunol 2010; 22:963-72. [PMID: 21115673 DOI: 10.1093/intimm/dxq450] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023] Open
Abstract
Hematopoiesis is controlled by the interplay between transcription factors and environmental signals. One of the primary determinants of the T-lineage choice is Delta-like (DL)-Notch signaling, which promotes T-cell development and inhibits B-cell development. We have found that the transcription factor HEBAlt is up-regulated in early hematopoietic precursors in response to DL-Notch signaling and that it can promote early T-cell development. Here, we identified a population of lineage-negative Sca-1⁻c-kit(+) (LK) cells in the mouse fetal liver that rapidly gave rise to myeloid cells and B cells but exhibited very little T-cell potential. However, forced expression of HEBAlt in these precursors restored their ability to develop into T cells. We also showed that Ikaros and Notch1 are up-regulated in response to HEBAlt over-expression and that activated Notch1 enhances the ability of LK cells to enter the T-cell lineage. Furthermore, the myeloid transcription factor C/EBPα is down-regulated in response to HEBAlt. We therefore propose that HEBAlt plays a role in the network that enforces the T-lineage fate and limits myeloid fate during hematopoiesis.
Collapse
Affiliation(s)
- Marsela Braunstein
- Division of Molecular and Cellular Biology, Sunnybrook Research Institute, Toronto, Ontario M4N 3M5, Canada
| | | | | | | | | | | | | |
Collapse
|
40
|
Vicente R, Swainson L, Marty-Grès S, De Barros SC, Kinet S, Zimmermann VS, Taylor N. Molecular and cellular basis of T cell lineage commitment. Semin Immunol 2010; 22:270-5. [PMID: 20630771 DOI: 10.1016/j.smim.2010.04.016] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/15/2010] [Accepted: 04/23/2010] [Indexed: 12/16/2022]
Abstract
The thymus forms as an alymphoid thymic primordium with T cell differentiation requiring the seeding of this anlage. This review will focus on the characteristics of the hematopoietic progenitors which colonize the thymus and their subsequent commitment/differentiation, both in mice and men. Within the thymus, the interplay between Notch1 and IL-7 signals is crucial for the orchestration of T cell development, but the precise requirements for these factors in murine and human thympoeisis are not synonymous. Recent advances in our understanding of the mechanisms regulating precursor entry and their maintenance in the thymus will also be presented.
Collapse
Affiliation(s)
- Rita Vicente
- Institut de Génétique Moléculaire de Montpellier, CNRS UMR 5535/IFR 122, 34293 Montpellier Cedex 5, France
| | | | | | | | | | | | | |
Collapse
|
41
|
Revised map of the human progenitor hierarchy shows the origin of macrophages and dendritic cells in early lymphoid development. Nat Immunol 2010; 11:585-93. [PMID: 20543838 DOI: 10.1038/ni.1889] [Citation(s) in RCA: 352] [Impact Index Per Article: 23.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2010] [Accepted: 05/18/2010] [Indexed: 12/11/2022]
Abstract
The classical model of hematopoiesis posits the segregation of lymphoid and myeloid lineages as the earliest fate decision. The validity of this model in the mouse has been questioned; however, little is known about the lineage potential of human progenitors. Here we provide a comprehensive analysis of the human hematopoietic hierarchy by clonally mapping the developmental potential of seven progenitor classes from neonatal cord blood and adult bone marrow. Human multilymphoid progenitors, identified as a distinct population of Thy-1(neg-lo)CD45RA(+) cells in the CD34(+)CD38(-) stem cell compartment, gave rise to all lymphoid cell types, as well as monocytes, macrophages and dendritic cells, which indicated that these myeloid lineages arise in early lymphoid lineage specification. Thus, as in the mouse, human hematopoiesis does not follow a rigid model of myeloid-lymphoid segregation.
Collapse
|
42
|
Sultana DA, Bell JJ, Zlotoff DA, De Obaldia ME, Bhandoola A. Eliciting the T cell fate with Notch. Semin Immunol 2010; 22:254-60. [PMID: 20627765 DOI: 10.1016/j.smim.2010.04.011] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/15/2010] [Accepted: 04/23/2010] [Indexed: 10/19/2022]
Abstract
Multipotent progenitors arrive at the thymus via the blood. Constraining the non-T cell fates of these progenitors while promoting the T cell fate is a major task of the thymus. Notch appears to be the initial trigger for a developmental program that eventually results in T cell lineage commitment. Several downstream targets of Notch are known, but the specific roles of each are poorly understood. A greater understanding of how Notch and other thymic signals direct progenitors to a T cell fate could be useful for translational work. For example, such work could eventually allow for the generation of fully competent T cells in vitro that could supplement the waning T cell numbers and function in the elderly and boost T cell-mediated immunity in patients with immunodeficiency and after stem cell transplantation.
Collapse
Affiliation(s)
- Dil Afroz Sultana
- Department of Pathology and Laboratory Medicine, University of Pennsylvania, School of Medicine, Philadelphia, PA 19104, USA
| | | | | | | | | |
Collapse
|
43
|
Feeding the fire: the role of defective bone marrow function in exacerbating thymic involution. Trends Immunol 2010; 31:191-8. [DOI: 10.1016/j.it.2010.02.002] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2009] [Revised: 02/02/2010] [Accepted: 02/25/2010] [Indexed: 12/28/2022]
|