1
|
Michaels YS, Major MC, Bonham-Carter B, Zhang J, Heydari T, Edgar JM, Siu MM, Greenstreet L, Vilarrasa-Blasi R, Kim S, Castle EL, Forrow A, Ibanez-Rios MI, Zimmerman C, Chung Y, Stach T, Werschler N, Knapp DJHF, Vento-Tormo R, Schiebinger G, Zandstra PW. Tracking the gene expression programs and clonal relationships that underlie mast, myeloid, and T lineage specification from stem cells. Cell Syst 2024; 15:1245-1263.e10. [PMID: 39615483 DOI: 10.1016/j.cels.2024.11.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2023] [Revised: 07/03/2024] [Accepted: 11/01/2024] [Indexed: 12/21/2024]
Abstract
T cells develop from hematopoietic progenitors in the thymus and protect against pathogens and cancer. However, the emergence of human T cell-competent blood progenitors and their subsequent specification to the T lineage have been challenging to capture in real time. Here, we leveraged a pluripotent stem cell differentiation system to understand the transcriptional dynamics and cell fate restriction events that underlie this critical developmental process. Time-resolved single-cell RNA sequencing revealed that downregulation of the multipotent hematopoietic program, upregulation of >90 lineage-associated transcription factors, and cell-cycle exit all occur within a highly coordinated developmental window. Gene-regulatory network inference uncovered a role for YBX1 in T lineage specification. We mapped the differentiation cell fate hierarchy using transcribed lineage barcoding and discovered that mast and myeloid potential bifurcate from each other early in hematopoiesis, upstream of T lineage restriction. Our systems-level analyses provide a quantitative, time-resolved model of human T cell fate specification. A record of this paper's transparent peer review process is included in the supplemental information.
Collapse
Affiliation(s)
- Yale S Michaels
- School of Biomedical Engineering, University of British Columbia, Vancouver, BC V6T 2B9, Canada; Paul Albrechtsen Research Institute CancerCare Manitoba, Winnipeg, MB R3E 0V9, Canada; Department of Biochemistry and Medical Genetics, Rady Faculty of Health Sciences, University of Manitoba, Winnipeg, MB R3E 0W2, Canada
| | - Matthew C Major
- School of Biomedical Engineering, University of British Columbia, Vancouver, BC V6T 2B9, Canada; Paul Albrechtsen Research Institute CancerCare Manitoba, Winnipeg, MB R3E 0V9, Canada
| | - Becca Bonham-Carter
- Department of Mathematics, University of British Columbia, Vancouver, BC V6T 1Z2, Canada
| | - Jingqi Zhang
- Department of Mathematics, University of British Columbia, Vancouver, BC V6T 1Z2, Canada
| | - Tiam Heydari
- School of Biomedical Engineering, University of British Columbia, Vancouver, BC V6T 2B9, Canada
| | - John M Edgar
- School of Biomedical Engineering, University of British Columbia, Vancouver, BC V6T 2B9, Canada
| | - Mona M Siu
- School of Biomedical Engineering, University of British Columbia, Vancouver, BC V6T 2B9, Canada
| | - Laura Greenstreet
- Department of Mathematics, University of British Columbia, Vancouver, BC V6T 1Z2, Canada
| | | | - Seungjoon Kim
- Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton, Cambridge CB10 1SA, UK
| | - Elizabeth L Castle
- School of Biomedical Engineering, University of British Columbia, Vancouver, BC V6T 2B9, Canada
| | - Aden Forrow
- Department of Mathematics and Statistics, University of Maine, Orono, ME 04469-5752, USA
| | - M Iliana Ibanez-Rios
- Institut de recherche en immunologie et en cancérologie and Département de pathologie et biologie cellulaire, Université de Montréal, Montreal, QC H3T 1J4, Canada
| | - Carla Zimmerman
- School of Biomedical Engineering, University of British Columbia, Vancouver, BC V6T 2B9, Canada
| | - Yvonne Chung
- School of Biomedical Engineering, University of British Columbia, Vancouver, BC V6T 2B9, Canada
| | - Tara Stach
- School of Biomedical Engineering, University of British Columbia, Vancouver, BC V6T 2B9, Canada
| | - Nico Werschler
- School of Biomedical Engineering, University of British Columbia, Vancouver, BC V6T 2B9, Canada; Life Sciences Institute, University of British Columbia, Vancouver, BC V6T 1Z3, Canada
| | - David J H F Knapp
- Institut de recherche en immunologie et en cancérologie and Département de pathologie et biologie cellulaire, Université de Montréal, Montreal, QC H3T 1J4, Canada
| | - Roser Vento-Tormo
- Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton, Cambridge CB10 1SA, UK
| | - Geoffrey Schiebinger
- Department of Mathematics, University of British Columbia, Vancouver, BC V6T 1Z2, Canada.
| | - Peter W Zandstra
- School of Biomedical Engineering, University of British Columbia, Vancouver, BC V6T 2B9, Canada; Michael Smith Laboratories, University of British Columbia, Vancouver, BC V6T 1Z4, Canada.
| |
Collapse
|
2
|
A regulatory network of microRNAs confers lineage commitment during early developmental trajectories of B and T lymphocytes. Proc Natl Acad Sci U S A 2021; 118:2104297118. [PMID: 34750254 DOI: 10.1073/pnas.2104297118] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/10/2021] [Indexed: 11/18/2022] Open
Abstract
The commitment of hematopoietic multipotent progenitors (MPPs) toward a particular lineage involves activation of cell type-specific genes and silencing of genes that promote alternate cell fates. Although the gene expression programs of early-B and early-T lymphocyte development are mutually exclusive, we show that these cell types exhibit significantly correlated microRNA (miRNA) profiles. However, their corresponding miRNA targetomes are distinct and predominated by transcripts associated with natural killer, dendritic cell, and myeloid lineages, suggesting that miRNAs function in a cell-autonomous manner. The combinatorial expression of miRNAs miR-186-5p, miR-128-3p, and miR-330-5p in MPPs significantly attenuates their myeloid differentiation potential due to repression of myeloid-associated transcripts. Depletion of these miRNAs caused a pronounced de-repression of myeloid lineage targets in differentiating early-B and early-T cells, resulting in a mixed-lineage gene expression pattern. De novo motif analysis combined with an assay of promoter activities indicates that B as well as T lineage determinants drive the expression of these miRNAs in lymphoid lineages. Collectively, we present a paradigm that miRNAs are conserved between developing B and T lymphocytes, yet they target distinct sets of promiscuously expressed lineage-inappropriate genes to suppress the alternate cell-fate options. Thus, our studies provide a comprehensive compendium of miRNAs with functional implications for B and T lymphocyte development.
Collapse
|
3
|
Goldsmith CD, Donovan T, Vlahovich N, Pyne DB. Unlocking the Role of Exercise on CD4+ T Cell Plasticity. Front Immunol 2021; 12:729366. [PMID: 34759918 PMCID: PMC8573256 DOI: 10.3389/fimmu.2021.729366] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2021] [Accepted: 09/28/2021] [Indexed: 11/26/2022] Open
Abstract
A hallmark of T cell ageing is a loss of effector plasticity. Exercise delays T cell ageing, yet the mechanisms driving the effects of exercise on T cell biology are not well elucidated. T cell plasticity is closely linked with metabolism, and consequently sensitive to metabolic changes induced by exercise. Mitochondrial function is essential for providing the intermediate metabolites necessary to generate and modify epigenetic marks in the nucleus, thus metabolic activity and epigenetic mechanisms are intertwined. In this perspective we propose a role for exercise in CD4+ T cell plasticity, exploring links between exercise, metabolism and epigenetic reprogramming.
Collapse
Affiliation(s)
- Chloé D Goldsmith
- Research Institute for Sport and Exercise, University of Canberra, Canberra, ACT, Australia
| | - Thomasina Donovan
- Research Institute for Sport and Exercise, University of Canberra, Canberra, ACT, Australia.,Australian Centre for Health Services Innovation and Centre for Healthcare Transformation, School of Public Health and Social Work, Faculty of Health, Queensland University of Technology, Brisbane, QLD, Australia
| | - Nicole Vlahovich
- Faculty of Health, University of Canberra, Canberra, ACT, Australia
| | - David B Pyne
- Research Institute for Sport and Exercise, University of Canberra, Canberra, ACT, Australia.,School of Psychology and Life Sciences, Faculty of Science, Engineering and Social Sciences, Canterbury Christ Church University, Canterbury, United Kingdom
| |
Collapse
|
4
|
Anderson MK, Selvaratnam JS. Interaction between γδTCR signaling and the E protein-Id axis in γδ T cell development. Immunol Rev 2020; 298:181-197. [PMID: 33058287 DOI: 10.1111/imr.12924] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2020] [Revised: 08/24/2020] [Accepted: 08/28/2020] [Indexed: 02/06/2023]
Abstract
γδ T cells acquire their functional properties in the thymus, enabling them to exert rapid innate-like responses. To understand how distinct γδ T cell subsets are generated, we have developed a Two-Stage model for γδ T cell development. This model is predicated on the finding that γδTCR signal strength impacts E protein activity through graded upregulation of Id3. Our model proposes that cells enter Stage 1 in response to a γδTCR signaling event in the cortex that activates a γδ T cell-specific gene network. Part of this program includes the upregulation of chemokine receptors that guide them to the medulla. In the medulla, Stage 1 cells receive distinct combinations of γδTCR, cytokine, and/co-stimulatory signals that induce their transit into Stage 2, either toward the γδT1 or the γδT17 lineage. The intersection between γδTCR and cytokine signals can tune Id3 expression, leading to different outcomes even in the presence of strong γδTCR signals. The thymic signaling niches required for γδT17 development are segregated in time and space, providing transient windows of opportunity during ontogeny. Understanding the regulatory context in which E proteins operate at different stages will be key in defining how their activity levels impose functional outcomes.
Collapse
Affiliation(s)
- Michele K Anderson
- Biological Sciences, Sunnybrook Research Institute, Toronto, ON, Canada.,Department of Immunology, University of Toronto, Toronto, ON, Canada
| | - Johanna S Selvaratnam
- Biological Sciences, Sunnybrook Research Institute, Toronto, ON, Canada.,Department of Immunology, University of Toronto, Toronto, ON, Canada
| |
Collapse
|
5
|
Barik S, Cattin-Roy AN, Ukah TK, Miller MM, Teixeiro E, Zaghouani H. Type II Cytokines Fine-Tune Thymic T Cell Selection to Offset Murine Central Nervous System Autoimmunity. THE JOURNAL OF IMMUNOLOGY 2020; 205:2039-2045. [PMID: 32917785 DOI: 10.4049/jimmunol.2000614] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/27/2020] [Accepted: 08/18/2020] [Indexed: 12/26/2022]
Abstract
Early thymic progenitors (ETPs) are bone marrow-derived hematopoietic stem cells that remain multipotent and give rise to a variety of lineage-specific cells. Recently, we discovered a subset of murine ETPs that expresses the IL-4Rα/IL-13Rα1 heteroreceptor (HR) and commits only to the myeloid lineage. This is because IL-4/IL-13 signaling through the HR inhibits their T cell potential and enacts commitment of HR+ETPs to thymic resident CD11c+CD8α+ dendritic cells (DCs). In this study, we discovered that HR+-ETP-derived DCs function as APCs in the thymus and promote deletion of myelin-reactive T cells. Furthermore, this negative T cell selection function of HR+-ETP-derived DCs sustains protection against experimental allergic encephalomyelitis, a mouse model for human multiple sclerosis. These findings, while shedding light on the intricacies underlying ETP lineage commitment, reveal a novel, to our knowledge, function by which IL-4 and IL-13 cytokines condition thymic microenvironment to rheostat T cell selection and fine-tune central tolerance.
Collapse
Affiliation(s)
- Subhasis Barik
- Department of Molecular Microbiology and Immunology, University of Missouri School of Medicine, Columbia, MO 65212
| | - Alexis N Cattin-Roy
- Department of Molecular Microbiology and Immunology, University of Missouri School of Medicine, Columbia, MO 65212
| | - Tobechukwu K Ukah
- Department of Molecular Microbiology and Immunology, University of Missouri School of Medicine, Columbia, MO 65212
| | - Mindy M Miller
- Department of Molecular Microbiology and Immunology, University of Missouri School of Medicine, Columbia, MO 65212
| | - Emma Teixeiro
- Department of Molecular Microbiology and Immunology, University of Missouri School of Medicine, Columbia, MO 65212
| | - Habib Zaghouani
- Department of Molecular Microbiology and Immunology, University of Missouri School of Medicine, Columbia, MO 65212
| |
Collapse
|
6
|
Dash B, Shapiro MJ, Thapa P, Romero Arocha S, Chung JY, Schwab AD, McCue SA, Rajcula MJ, Shapiro VS. The Interaction between NKAP and HDAC3 Is Critical for T Cell Maturation. Immunohorizons 2019; 3:352-367. [PMID: 31387873 DOI: 10.4049/immunohorizons.1900052] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2019] [Accepted: 07/09/2019] [Indexed: 12/14/2022] Open
Abstract
NKAP and HDAC3 are critical for T cell maturation. NKAP and HDAC3 physically associate, and a point mutation in NKAP, NKAP(Y352A), abrogates this interaction. To evaluate the significance of NKAP and HDAC3 association in T cell maturation, transgenic mice were engineered for cre-mediated endogenous NKAP gene deletion coupled to induction of NKAP(Y352A) or a wild type (WT) control transgene, NKAP(WT), in double positive thymocytes or regulatory T cells (Tregs). T cell maturation was normal in mice with endogenous NKAP deletion coupled to NKAP(WT) induction. However, severe defects occurred in T cell and Treg maturation and in iNKT cell development when NKAP(Y352A) was induced, recapitulating NKAP deficiency. Conventional T cells expressing NKAP(Y352A) failed to enter the long-term T cell pool, did not produce cytokines, and remained complement susceptible, whereas Tregs expressing NKAP(Y352A) were eliminated as recent thymic emigrants leading to lethal autoimmunity. Overall, these results demonstrate the significance of NKAP-HDAC3 association in T cells.
Collapse
Affiliation(s)
- Barsha Dash
- Department of Immunology, Mayo Clinic, Rochester, MN 55905
| | | | - Puspa Thapa
- Columbia Center for Translational Immunology, Columbia University Medical Center, New York, NY 10032; and.,Department of Medicine, Columbia University Medical Center, New York, NY 10032
| | | | - Ji-Young Chung
- Department of Immunology, Mayo Clinic, Rochester, MN 55905
| | - Aaron D Schwab
- Department of Immunology, Mayo Clinic, Rochester, MN 55905
| | | | | | | |
Collapse
|
7
|
Gong Z, Shang B, Chu Y, Chen X, Li Q, Liu K, Chen Y, Huang Y, Han Y, Shang Q, Zheng Z, Song L, Li Y, Liu R, Xu C, Zhang X, Liu B, Wang L, Shao C, Wang Y, Shi Y. Fibrotic liver microenvironment promotes Dll4 and SDF-1-dependent T-cell lineage development. Cell Death Dis 2019; 10:440. [PMID: 31165736 PMCID: PMC6549170 DOI: 10.1038/s41419-019-1630-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2019] [Revised: 04/22/2019] [Accepted: 04/29/2019] [Indexed: 11/09/2022]
Abstract
The reconstitution of the T-cell repertoire and quantity is a major challenge in the clinical management of HIV infection/AIDS, cancer, and aging-associated diseases. We previously showed that autologous bone marrow transfusion (BMT) via the hepatic portal vein could effectively restore CD4+ T-cell count in AIDS patients also suffering from decompensated liver cirrhosis. In the current study, we characterized T-cell reconstitution in a mouse model of liver fibrosis induced by CCl4 and found that T-cell reconstitution after BMT via hepatic portal vein was also greatly enhanced. The expression of Dll4 (Delta-like 4), which plays an important role in T-cell progenitor expansion, was elevated in hepatocytes of fibrotic livers when compared to normal livers. This upregulation of Dll4 expression was found to be induced by TNFα in an NFκB-dependent manner. Liver fibroblasts transfected with Dll4 (LF-Dll4) also gained the capacity to promote T-cell lineage development from hematopoietic stem cells (HSCs), resulting in the generation of DN2 (CD4 and CD8 DN 2) and DN3 T-cell progenitors in vitro, which underwent a normal maturation program when adoptively transferred into Rag-2 deficient hosts. We also demonstrated a pivotal role of SDF-1 produced by primary liver fibroblasts (primary LF) in T-lineage differentiation from HSCs. These results suggest that Dll4 and SDF-1 in fibrotic liver microenvironment could promote extrathymic T-cell lineage development. These results expand our knowledge of T-cell development and reconstitution under pathological conditions.
Collapse
Affiliation(s)
- Zheng Gong
- The First Affiliated Hospital of Soochow University, Institutes for Translational Medicine, State Key Laboratory of Radiation Medicine and Protection, Key Laboratory of Stem Cells and Medical Biomaterials of Jiangsu Province, Soochow University Medical College, Suzhou, China
- Key Laboratory of Tissue Microenvironment and Tumor, Institute of Health Sciences, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai, China
| | - Bingxue Shang
- The First Affiliated Hospital of Soochow University, Institutes for Translational Medicine, State Key Laboratory of Radiation Medicine and Protection, Key Laboratory of Stem Cells and Medical Biomaterials of Jiangsu Province, Soochow University Medical College, Suzhou, China
| | - Yunpeng Chu
- The First Affiliated Hospital of Soochow University, Institutes for Translational Medicine, State Key Laboratory of Radiation Medicine and Protection, Key Laboratory of Stem Cells and Medical Biomaterials of Jiangsu Province, Soochow University Medical College, Suzhou, China
| | - Xiaodong Chen
- Key Laboratory of Tissue Microenvironment and Tumor, Institute of Health Sciences, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai, China
| | - Qing Li
- Key Laboratory of Tissue Microenvironment and Tumor, Institute of Health Sciences, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai, China
| | - Keli Liu
- Key Laboratory of Tissue Microenvironment and Tumor, Institute of Health Sciences, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai, China
| | - Yongjing Chen
- The First Affiliated Hospital of Soochow University, Institutes for Translational Medicine, State Key Laboratory of Radiation Medicine and Protection, Key Laboratory of Stem Cells and Medical Biomaterials of Jiangsu Province, Soochow University Medical College, Suzhou, China
| | - Yin Huang
- Key Laboratory of Tissue Microenvironment and Tumor, Institute of Health Sciences, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai, China
| | - Yanyan Han
- Key Laboratory of Tissue Microenvironment and Tumor, Institute of Health Sciences, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai, China
| | - Qianwen Shang
- The First Affiliated Hospital of Soochow University, Institutes for Translational Medicine, State Key Laboratory of Radiation Medicine and Protection, Key Laboratory of Stem Cells and Medical Biomaterials of Jiangsu Province, Soochow University Medical College, Suzhou, China
| | - Zhiyuan Zheng
- The First Affiliated Hospital of Soochow University, Institutes for Translational Medicine, State Key Laboratory of Radiation Medicine and Protection, Key Laboratory of Stem Cells and Medical Biomaterials of Jiangsu Province, Soochow University Medical College, Suzhou, China
| | - Lin Song
- The First Affiliated Hospital of Soochow University, Institutes for Translational Medicine, State Key Laboratory of Radiation Medicine and Protection, Key Laboratory of Stem Cells and Medical Biomaterials of Jiangsu Province, Soochow University Medical College, Suzhou, China
| | - Yanan Li
- The First Affiliated Hospital of Soochow University, Institutes for Translational Medicine, State Key Laboratory of Radiation Medicine and Protection, Key Laboratory of Stem Cells and Medical Biomaterials of Jiangsu Province, Soochow University Medical College, Suzhou, China
| | - Rui Liu
- The First Affiliated Hospital of Soochow University, Institutes for Translational Medicine, State Key Laboratory of Radiation Medicine and Protection, Key Laboratory of Stem Cells and Medical Biomaterials of Jiangsu Province, Soochow University Medical College, Suzhou, China
| | - Chenchang Xu
- The First Affiliated Hospital of Soochow University, Institutes for Translational Medicine, State Key Laboratory of Radiation Medicine and Protection, Key Laboratory of Stem Cells and Medical Biomaterials of Jiangsu Province, Soochow University Medical College, Suzhou, China
| | - Xiaoren Zhang
- Key Laboratory of Tissue Microenvironment and Tumor, Institute of Health Sciences, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai, China
| | - Baochi Liu
- Department of Surgery, Shanghai Public Health Clinical Center Fudan University, Shanghai, China
| | - Luowei Wang
- Department of Gastroenterology, Changhai Hospital, Second Military Medical University, Shanghai, China.
| | - Changshun Shao
- The First Affiliated Hospital of Soochow University, Institutes for Translational Medicine, State Key Laboratory of Radiation Medicine and Protection, Key Laboratory of Stem Cells and Medical Biomaterials of Jiangsu Province, Soochow University Medical College, Suzhou, China.
| | - Ying Wang
- Key Laboratory of Tissue Microenvironment and Tumor, Institute of Health Sciences, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai, China.
| | - Yufang Shi
- The First Affiliated Hospital of Soochow University, Institutes for Translational Medicine, State Key Laboratory of Radiation Medicine and Protection, Key Laboratory of Stem Cells and Medical Biomaterials of Jiangsu Province, Soochow University Medical College, Suzhou, China.
- Key Laboratory of Tissue Microenvironment and Tumor, Institute of Health Sciences, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai, China.
| |
Collapse
|
8
|
Simons L, Cavazzana M, André I. Concise Review: Boosting T-Cell Reconstitution Following Allogeneic Transplantation-Current Concepts and Future Perspectives. Stem Cells Transl Med 2019; 8:650-657. [PMID: 30887712 PMCID: PMC6591542 DOI: 10.1002/sctm.18-0248] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2018] [Accepted: 01/06/2019] [Indexed: 12/14/2022] Open
Abstract
Allogeneic hematopoietic stem cell transplantation (HSCT) is the treatment of choice for a large number of malignant and nonmalignant (inherited) diseases of the hematopoietic system. Nevertheless, non‐HLA identical transplantations are complicated by a severe T‐cell immunodeficiency associated with a high rate of infection, relapse and graft‐versus‐host disease. Initial recovery of T‐cell immunity following HSCT relies on peripheral expansion of memory T cells mostly driven by cytokines. The reconstitution of a diverse, self‐tolerant, and naive T‐cell repertoire, however, may take up to 2 years and crucially relies on the interaction of T‐cell progenitors with the host thymic epithelium, which may be altered by GvHD, age or transplant‐related toxicities. In this review, we summarize current concepts to stimulate reconstitution of a peripheral and polyclonal T‐cell compartment following allogeneic transplantation such as graft manipulation (i.e., T‐cell depletion), transfusion of ex vivo manipulated donor T cells or the exogenous administration of cytokines and growth factors to stimulate host‐thymopoiesis with emphasis on approaches which have led to clinical trials. Particular attention will be given to the development of cellular therapies such as the ex vivo generation of T‐cell precursors to fasten generation of a polyclonal and functional host‐derived T‐cell repertoire. Having been tested so far only in preclinical mouse models, clinical studies are now on the way to validate the efficacy of such T‐cell progenitors in enhancing immune reconstitution following HSCT in various clinical settings. stem cells translational medicine2019;00:1–8
Collapse
Affiliation(s)
- Laura Simons
- Laboratory of Human Lymphohematopoiesis, INSERM UMR 1163, Imagine Institute, Paris, France.,Paris Descartes University-Sorbonne Paris Cité, Imagine Institute, Paris, France.,Department of Biotherapy, Necker Children's Hospital, Assistance Publique-Hôpitaux de Paris, Paris, France
| | - Marina Cavazzana
- Biotherapy Clinical Investigation Center, Groupe Hospitalier Universitaire Ouest, Assistance Publique-Hôpitaux de Paris, INSERM CIC, Paris, France.,Laboratory of Human Lymphohematopoiesis, INSERM UMR 1163, Imagine Institute, Paris, France.,Paris Descartes University-Sorbonne Paris Cité, Imagine Institute, Paris, France.,Department of Biotherapy, Necker Children's Hospital, Assistance Publique-Hôpitaux de Paris, Paris, France
| | - Isabelle André
- Laboratory of Human Lymphohematopoiesis, INSERM UMR 1163, Imagine Institute, Paris, France.,Paris Descartes University-Sorbonne Paris Cité, Imagine Institute, Paris, France
| |
Collapse
|
9
|
Rothenberg EV, Hosokawa H, Ungerbäck J. Mechanisms of Action of Hematopoietic Transcription Factor PU.1 in Initiation of T-Cell Development. Front Immunol 2019; 10:228. [PMID: 30842770 PMCID: PMC6391351 DOI: 10.3389/fimmu.2019.00228] [Citation(s) in RCA: 58] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2018] [Accepted: 01/28/2019] [Indexed: 12/20/2022] Open
Abstract
PU.1 is an ETS-family transcription factor that plays a broad range of roles in hematopoiesis. A direct regulator of myeloid, dendritic-cell, and B cell functional programs, and a well-known antagonist of terminal erythroid cell differentiation, it is also expressed in the earliest stages of T-cell development of each cohort of intrathymic pro-T cells. Its expression in this context appears to give T-cell precursors initial, transient access to myeloid and dendritic cell developmental competence and therefore to represent a source of antagonism or delay of T-cell lineage commitment. However, it has remained uncertain until recently why T-cell development is also intensely dependent upon PU.1. Here, we review recent work that sheds light on the molecular biology of PU.1 action across the genome in pro-T cells and identifies the genes that depend on PU.1 for their correct regulation. This work indicates modes of chromatin engagement, pioneering, and cofactor recruitment (“coregulator theft”) by PU.1 as well as gene network interactions that not only affect specific target genes but also have system-wide regulatory consequences, amplifying the impact of PU.1 beyond its own direct binding targets. The genes directly regulated by PU.1 also suggest a far-reaching transformation of cell biology and signaling potential between the early stages of T-cell development when PU.1 is expressed and when it is silenced. These cell-biological functions can be important to distinguish fetal from adult T-cell development and have the potential to illuminate aspects of thymic function that have so far remained the most mysterious.
Collapse
Affiliation(s)
- Ellen V Rothenberg
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA, United States
| | - Hiroyuki Hosokawa
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA, United States
| | - Jonas Ungerbäck
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA, United States
| |
Collapse
|
10
|
Ha VL, Luong A, Li F, Casero D, Malvar J, Kim YM, Bhatia R, Crooks GM, Parekh C. The T-ALL related gene BCL11B regulates the initial stages of human T-cell differentiation. Leukemia 2017; 31:2503-2514. [PMID: 28232744 PMCID: PMC5599326 DOI: 10.1038/leu.2017.70] [Citation(s) in RCA: 44] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2016] [Revised: 01/16/2017] [Accepted: 02/15/2017] [Indexed: 02/07/2023]
Abstract
The initial stages of T-cell differentiation are characterized by a progressive commitment to the T-cell lineage, a process that involves the loss of alternative (myelo-erythroid, NK, B) lineage potentials. Aberrant differentiation during these stages can result in T-cell acute lymphoblastic leukemia (T-ALL). However, the mechanisms regulating the initial stages of human T-cell differentiation are obscure. Through loss of function studies, we showed BCL11B, a transcription factor recurrently mutated T-ALL, is essential for T-lineage commitment, particularly the repression of NK and myeloid potentials, and the induction of T-lineage genes, during the initial stages of human T-cell differentiation. In gain of function studies, BCL11B inhibited growth of and induced a T-lineage transcriptional program in T-ALL cells. We found previously unknown differentiation stage-specific DNA binding of BCL11B at multiple T-lineage genes; target genes showed BCL11B-dependent expression, suggesting a transcriptional activator role for BCL11B at these genes. Transcriptional analyses revealed differences in the regulatory actions of BCL11B between human and murine thymopoiesis. Our studies show BCL11B is a key regulator of the initial stages of human T-cell differentiation and delineate the BCL11B transcriptional program, enabling the dissection of the underpinnings of normal T-cell differentiation and providing a resource for understanding dysregulations in T-ALL.
Collapse
Affiliation(s)
- VL Ha
- Children’s Center for Cancer and Blood Disease, Children’s Hospital Los Angeles, Los Angeles, California, USA
| | - A Luong
- Children’s Center for Cancer and Blood Disease, Children’s Hospital Los Angeles, Los Angeles, California, USA
| | - F Li
- MiNGS Core Laboratory, Children’s Hospital Los Angeles, Los Angeles, California, USA
| | - D Casero
- Department of Pathology & Laboratory Medicine, David Geffen School of Medicine University of California, Los Angeles, Los Angeles, California, USA
| | - J Malvar
- Children’s Center for Cancer and Blood Disease, Children’s Hospital Los Angeles, Los Angeles, California, USA
| | - YM Kim
- Children’s Center for Cancer and Blood Disease, Children’s Hospital Los Angeles, Los Angeles, California, USA
- Department of Pediatrics, Keck School of Medicine, University of Southern California, Los Angeles, California, USA
| | - R Bhatia
- Division of Hematology and Oncology, University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - GM Crooks
- Department of Pathology & Laboratory Medicine, David Geffen School of Medicine University of California, Los Angeles, Los Angeles, California, USA
- Eli and Edythe Broad Center for Regenerative Medicine and Stem Cell Research, University of California, Los Angeles, Los Angeles, California, USA
- Department of Pediatrics, David Geffen School of Medicine at University of California Los Angeles, Los Angeles, California, USA
- Jonsson Comprehensive Cancer Center, University of California Los Angeles, Los Angeles, California, USA
| | - C Parekh
- Children’s Center for Cancer and Blood Disease, Children’s Hospital Los Angeles, Los Angeles, California, USA
- Department of Pediatrics, Keck School of Medicine, University of Southern California, Los Angeles, California, USA
| |
Collapse
|
11
|
GATA3 Abundance Is a Critical Determinant of T Cell Receptor β Allelic Exclusion. Mol Cell Biol 2017; 37:MCB.00052-17. [PMID: 28320875 DOI: 10.1128/mcb.00052-17] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2017] [Accepted: 03/14/2017] [Indexed: 12/30/2022] Open
Abstract
Allelic exclusion describes the essential immunological process by which feedback repression of sequential DNA rearrangements ensures that only one autosome expresses a functional T or B cell receptor. In wild-type mammals, approximately 60% of cells have recombined the DNA of one T cell receptor β (TCRβ) V-to-DJ-joined allele in a functional configuration, while the second allele has recombined only the DJ sequences; the other 40% of cells have recombined the V to the DJ segments on both alleles, with only one of the two alleles predicting a functional TCRβ protein. Here we report that the transgenic overexpression of GATA3 leads predominantly to biallelic TCRβ gene (Tcrb) recombination. We also found that wild-type immature thymocytes can be separated into distinct populations based on intracellular GATA3 expression and that GATA3LO cells had almost exclusively recombined only one Tcrb locus (that predicted a functional receptor sequence), while GATA3HI cells had uniformly recombined both Tcrb alleles (one predicting a functional and the other predicting a nonfunctional rearrangement). These data show that GATA3 abundance regulates the recombination propensity at the Tcrb locus and provide new mechanistic insight into the historic immunological conundrum for how Tcrb allelic exclusion is mediated.
Collapse
|
12
|
Ettersperger J, Montcuquet N, Malamut G, Guegan N, Lopez-Lastra S, Gayraud S, Reimann C, Vidal E, Cagnard N, Villarese P, Andre-Schmutz I, Gomes Domingues R, Godinho-Silva C, Veiga-Fernandes H, Lhermitte L, Asnafi V, Macintyre E, Cellier C, Beldjord K, Di Santo JP, Cerf-Bensussan N, Meresse B. Interleukin-15-Dependent T-Cell-like Innate Intraepithelial Lymphocytes Develop in the Intestine and Transform into Lymphomas in Celiac Disease. Immunity 2016; 45:610-625. [PMID: 27612641 DOI: 10.1016/j.immuni.2016.07.018] [Citation(s) in RCA: 109] [Impact Index Per Article: 12.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2015] [Revised: 05/12/2016] [Accepted: 06/02/2016] [Indexed: 01/19/2023]
Abstract
The nature of gut intraepithelial lymphocytes (IELs) lacking antigen receptors remains controversial. Herein we showed that, in humans and in mice, innate intestinal IELs expressing intracellular CD3 (iCD3(+)) differentiate along an Id2 transcription factor (TF)-independent pathway in response to TF NOTCH1, interleukin-15 (IL-15), and Granzyme B signals. In NOTCH1-activated human hematopoietic precursors, IL-15 induced Granzyme B, which cleaved NOTCH1 into a peptide lacking transcriptional activity. As a result, NOTCH1 target genes indispensable for T cell differentiation were silenced and precursors were reprogrammed into innate cells with T cell marks including intracellular CD3 and T cell rearrangements. In the intraepithelial lymphoma complicating celiac disease, iCD3(+) innate IELs acquired gain-of-function mutations in Janus kinase 1 or Signal transducer and activator of transcription 3, which enhanced their response to IL-15. Overall we characterized gut T cell-like innate IELs, deciphered their pathway of differentiation and showed their malignant transformation in celiac disease.
Collapse
Affiliation(s)
- Julien Ettersperger
- INSERM UMR1163, Laboratory of Intestinal Immunity, Institut Imagine, 75015 Paris, France; Université Paris Descartes-Sorbonne Paris Cité and Institut Imagine, 75015 Paris, France
| | - Nicolas Montcuquet
- INSERM UMR1163, Laboratory of Intestinal Immunity, Institut Imagine, 75015 Paris, France; Université Paris Descartes-Sorbonne Paris Cité and Institut Imagine, 75015 Paris, France
| | - Georgia Malamut
- INSERM UMR1163, Laboratory of Intestinal Immunity, Institut Imagine, 75015 Paris, France; Université Paris Descartes-Sorbonne Paris Cité and Institut Imagine, 75015 Paris, France; AP-HP, Department of Gastroenterology, Hôpital Européen Georges Pompidou, 75015 Paris, France
| | - Nicolas Guegan
- INSERM UMR1163, Laboratory of Intestinal Immunity, Institut Imagine, 75015 Paris, France; Université Paris Descartes-Sorbonne Paris Cité and Institut Imagine, 75015 Paris, France
| | - Silvia Lopez-Lastra
- Innate Immunity Unit, Institut Pasteur, 75015 Paris, France; INSERM U 668, 75015 Paris, France
| | - Ségolène Gayraud
- INSERM UMR1163, Laboratory of Intestinal Immunity, Institut Imagine, 75015 Paris, France; Université Paris Descartes-Sorbonne Paris Cité and Institut Imagine, 75015 Paris, France
| | - Christian Reimann
- Université Paris Descartes-Sorbonne Paris Cité and Institut Imagine, 75015 Paris, France; INSERM UMR1163, Laboratory of Human Lymphohematopoiesis, 75015 Paris, France
| | - Elodie Vidal
- Université Paris Descartes-Sorbonne Paris Cité, Institut Necker-Enfants-Malades, INSERM UMR1151 and, Biological Hematology, AP-HP Necker-Enfants-Malades, 75015 Paris, France
| | | | - Patrick Villarese
- Université Paris Descartes-Sorbonne Paris Cité, Institut Necker-Enfants-Malades, INSERM UMR1151 and, Biological Hematology, AP-HP Necker-Enfants-Malades, 75015 Paris, France
| | - Isabelle Andre-Schmutz
- Université Paris Descartes-Sorbonne Paris Cité and Institut Imagine, 75015 Paris, France; INSERM UMR1163, Laboratory of Human Lymphohematopoiesis, 75015 Paris, France
| | - Rita Gomes Domingues
- Instituto de Medicina Molecular, Faculdade de Medicina de Lisboa, 1649-028 Lisboa, Portugal
| | - Cristina Godinho-Silva
- Instituto de Medicina Molecular, Faculdade de Medicina de Lisboa, 1649-028 Lisboa, Portugal
| | | | - Ludovic Lhermitte
- Université Paris Descartes-Sorbonne Paris Cité, Institut Necker-Enfants-Malades, INSERM UMR1151 and, Biological Hematology, AP-HP Necker-Enfants-Malades, 75015 Paris, France
| | - Vahid Asnafi
- Université Paris Descartes-Sorbonne Paris Cité, Institut Necker-Enfants-Malades, INSERM UMR1151 and, Biological Hematology, AP-HP Necker-Enfants-Malades, 75015 Paris, France
| | - Elizabeth Macintyre
- Université Paris Descartes-Sorbonne Paris Cité, Institut Necker-Enfants-Malades, INSERM UMR1151 and, Biological Hematology, AP-HP Necker-Enfants-Malades, 75015 Paris, France
| | - Christophe Cellier
- INSERM UMR1163, Laboratory of Intestinal Immunity, Institut Imagine, 75015 Paris, France; Université Paris Descartes-Sorbonne Paris Cité and Institut Imagine, 75015 Paris, France; AP-HP, Department of Gastroenterology, Hôpital Européen Georges Pompidou, 75015 Paris, France
| | - Kheira Beldjord
- Université Paris Descartes-Sorbonne Paris Cité, Institut Necker-Enfants-Malades, INSERM UMR1151 and, Biological Hematology, AP-HP Necker-Enfants-Malades, 75015 Paris, France; Institut Universitaire d'Hématologie, Hôpital Saint-Louis, 75010 Paris, France
| | - James P Di Santo
- Innate Immunity Unit, Institut Pasteur, 75015 Paris, France; INSERM U 668, 75015 Paris, France
| | - Nadine Cerf-Bensussan
- INSERM UMR1163, Laboratory of Intestinal Immunity, Institut Imagine, 75015 Paris, France; Université Paris Descartes-Sorbonne Paris Cité and Institut Imagine, 75015 Paris, France.
| | - Bertrand Meresse
- INSERM UMR1163, Laboratory of Intestinal Immunity, Institut Imagine, 75015 Paris, France; Université Paris Descartes-Sorbonne Paris Cité and Institut Imagine, 75015 Paris, France.
| |
Collapse
|
13
|
TNF-alpha and Notch signaling regulates the expression of HOXB4 and GATA3 during early T lymphopoiesis. In Vitro Cell Dev Biol Anim 2016; 52:920-934. [PMID: 27251160 DOI: 10.1007/s11626-016-0055-8] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2016] [Accepted: 05/04/2016] [Indexed: 10/21/2022]
Abstract
During the early thymus colonization, Notch signaling activation on hematopoietic progenitor cells (HPCs) drives proliferation and T cell commitment. Although these processes are driven by transcription factors such as HOXB4 and GATA3, there is no evidence that Notch directly regulates their transcription. To evaluate the role of NOTCH and TNF signaling in this process, human CD34+ HPCs were cocultured with OP9-DL1 cells, in the presence or absence of TNF. The use of a Notch signaling inhibitor and a protein synthesis inhibitor allowed us to distinguish primary effects, mediated by direct signaling downstream Notch and TNF, from secondary effects, mediated by de novo synthesized proteins. A low and physiologically relevant concentration of TNF promoted T lymphopoiesis in OP9-DL1 cocultures. TNF positively modulated the expression of both transcripts in a Notch-dependent manner; however, GATA3 induction was mediated by a direct mechanism, while HOXB4 induction was indirect. Induction of both transcripts was repressed by a GSK3β inhibitor, indicating that activation of canonical Wnt signaling inhibits rather than induces their expression. Our study provides novel evidences of the mechanisms integrating Notch and TNF-alpha signaling in the transcriptional induction of GATA3 and HOXB4. This mechanism has direct implications in the control of self-renewal, proliferation, commitment, and T cell differentiation.
Collapse
|
14
|
Li Y, Wang H, Lu H, Hua S. Regulation of Memory T Cells by Interleukin-23. Int Arch Allergy Immunol 2016; 169:157-62. [PMID: 27100864 DOI: 10.1159/000445834] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022] Open
Abstract
Interleukin-23 (IL-23), a member of the IL-12 family of cytokines, is a heterodimeric cytokine. It is composed of subunits p40 (shared with IL-12) and p19 (an IL-12 p35-related subunit) and is secreted by several types of immune cells, such as natural killer cells and dendritic cells. The IL-23 receptor is composed of the subunit IL-12Rβ1 and the IL-23-specific subunit IL-23R. The binding of IL-23 to its specific cell surface receptor regulates a number of functions, including proliferation and differentiation of cells and secretion of cell factors. Memory T cells are a subset of T cells that secrete numerous important cell factors, and they function in the immune response to infection and diseases like cancer, autoimmune disease and bronchial asthma. IL-23R is expressed on the surface of memory T cells, which suggests that it can specifically regulate memory T cell function. IL-23 has been widely used as a clinical indicator in immune-related diseases and shows potential for use in disease treatment. Here we review the current progress in the study of the role of IL-23 in the regulation of memory T cells.
Collapse
Affiliation(s)
- Yanchun Li
- Division 1, Pediatric Respiratory Department, The First Hospital of Jilin University, Changchun, China
| | | | | | | |
Collapse
|
15
|
Malleshaiah M, Padi M, Rué P, Quackenbush J, Martinez-Arias A, Gunawardena J. Nac1 Coordinates a Sub-network of Pluripotency Factors to Regulate Embryonic Stem Cell Differentiation. Cell Rep 2016; 14:1181-1194. [PMID: 26832399 DOI: 10.1016/j.celrep.2015.12.101] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2015] [Revised: 10/19/2015] [Accepted: 12/23/2015] [Indexed: 12/15/2022] Open
Abstract
Pluripotent cells give rise to distinct cell types during development and are regulated by often self-reinforcing molecular networks. How such networks allow cells to differentiate is less well understood. Here, we use integrative methods to show that external signals induce reorganization of the mouse embryonic stem cell pluripotency network and that a sub-network of four factors, Nac1, Oct4, Tcf3, and Sox2, regulates their differentiation into the alternative mesendodermal and neuroectodermal fates. In the mesendodermal fate, Nac1 and Oct4 were constrained within quantitative windows, whereas Sox2 and Tcf3 were repressed. In contrast, in the neuroectodermal fate, Sox2 and Tcf3 were constrained while Nac1 and Oct4 were repressed. In addition, we show that Nac1 coordinates differentiation by activating Oct4 and inhibiting both Sox2 and Tcf3. Reorganization of progenitor cell networks around shared factors might be a common differentiation strategy and our integrative approach provides a general methodology for delineating such networks.
Collapse
Affiliation(s)
- Mohan Malleshaiah
- Department of Systems Biology, Harvard Medical School, 200 Longwood Avenue, Boston, MA 02115, USA.
| | - Megha Padi
- Biostatistics and Computational Biology, Dana-Farber Cancer Institute, 450 Brookline Avenue, Boston, MA 02215, USA
| | - Pau Rué
- Department of Genetics, University of Cambridge, Downing Street, Cambridge CB2 3EH, UK
| | - John Quackenbush
- Biostatistics and Computational Biology, Dana-Farber Cancer Institute, 450 Brookline Avenue, Boston, MA 02215, USA
| | | | - Jeremy Gunawardena
- Department of Systems Biology, Harvard Medical School, 200 Longwood Avenue, Boston, MA 02115, USA.
| |
Collapse
|
16
|
Kitajima K, Kawaguchi M, Miyashita K, Nakajima M, Kanokoda M, Hara T. Efficient production of T cells from mouse pluripotent stem cells by controlled expression of Lhx2. Genes Cells 2015; 20:720-38. [PMID: 26153538 DOI: 10.1111/gtc.12266] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2014] [Accepted: 06/03/2015] [Indexed: 01/01/2023]
Abstract
LIM-homeobox transcription factor Lhx2 induces ex vivo amplification of adult hematopoietic stem cells (HSCs) in mice. We previously showed that engraftable HSC-like cells are generated from mouse embryonic stem cells (ESCs) and induced pluripotent stem cells by enforced expression of Lhx2. However, when these HSC-like cells were transplanted into irradiated congenic mice, donor-derived T cells were barely detectable, whereas other lineages of hematopoietic cells were continuously produced. Here we investigated T-cell differentiation potential of the Lhx2-induced HSC-like cells using ESCs carrying doxycycline (dox)-inducible Lhx2 expression cassette. Dox-mediated over-expression of Lhx2 conferred a self-renewing activity to ESC-derived c-Kit(+) CD41(+) embryonic hematopoietic progenitor cells (HPCs), thereby converting them to HSC-like cells. When these HSC-like cells were transplanted into irradiated immunodeficient mice and they were supplied with a dox-containing water, CD4/8 double negative T cells were detected in their thymi. Once the Lhx2 expression was terminated, differentiation of CD4/8 double positive and single positive T cells was initiated in the thymi of transplanted mice and mature T cells were released in the peripheral blood. These results showed that engraftable HSC-like cells with full hematopoietic potential can be obtained from ESCs by the conditional expression of Lhx2.
Collapse
Affiliation(s)
- Kenji Kitajima
- Stem Cell Project, Tokyo Metropolitan Institute of Medical Science, 2-1-6 Kamikitazawa, Setagaya-ku, Tokyo 156-8506, Japan
| | - Manami Kawaguchi
- Stem Cell Project, Tokyo Metropolitan Institute of Medical Science, 2-1-6 Kamikitazawa, Setagaya-ku, Tokyo 156-8506, Japan.,Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, 1-5-45 Yushima, Bunkyo-ku, Tokyo 113-8510, Japan
| | - Kazuya Miyashita
- Stem Cell Project, Tokyo Metropolitan Institute of Medical Science, 2-1-6 Kamikitazawa, Setagaya-ku, Tokyo 156-8506, Japan.,Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, 1-5-45 Yushima, Bunkyo-ku, Tokyo 113-8510, Japan
| | - Marino Nakajima
- Stem Cell Project, Tokyo Metropolitan Institute of Medical Science, 2-1-6 Kamikitazawa, Setagaya-ku, Tokyo 156-8506, Japan.,Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, 1-5-45 Yushima, Bunkyo-ku, Tokyo 113-8510, Japan
| | - Mai Kanokoda
- Stem Cell Project, Tokyo Metropolitan Institute of Medical Science, 2-1-6 Kamikitazawa, Setagaya-ku, Tokyo 156-8506, Japan.,Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, 1-5-45 Yushima, Bunkyo-ku, Tokyo 113-8510, Japan
| | - Takahiko Hara
- Stem Cell Project, Tokyo Metropolitan Institute of Medical Science, 2-1-6 Kamikitazawa, Setagaya-ku, Tokyo 156-8506, Japan.,Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, 1-5-45 Yushima, Bunkyo-ku, Tokyo 113-8510, Japan
| |
Collapse
|
17
|
Yu VWC, Saez B, Cook C, Lotinun S, Pardo-Saganta A, Wang YH, Lymperi S, Ferraro F, Raaijmakers MHGP, Wu JY, Zhou L, Rajagopal J, Kronenberg HM, Baron R, Scadden DT. Specific bone cells produce DLL4 to generate thymus-seeding progenitors from bone marrow. ACTA ACUST UNITED AC 2015; 212:759-74. [PMID: 25918341 PMCID: PMC4419348 DOI: 10.1084/jem.20141843] [Citation(s) in RCA: 111] [Impact Index Per Article: 11.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2014] [Accepted: 03/26/2015] [Indexed: 12/12/2022]
Abstract
Osteocalcin (Ocn)-expressing bone marrow cells produce the Notch ligand DLL4, and this is required for lymphoid progenitor cells to seed the thymus. Production of the cells that ultimately populate the thymus to generate α/β T cells has been controversial, and their molecular drivers remain undefined. Here, we report that specific deletion of bone-producing osteocalcin (Ocn)-expressing cells in vivo markedly reduces T-competent progenitors and thymus-homing receptor expression among bone marrow hematopoietic cells. Decreased intrathymic T cell precursors and decreased generation of mature T cells occurred despite normal thymic function. The Notch ligand DLL4 is abundantly expressed on bone marrow Ocn+ cells, and selective depletion of DLL4 from these cells recapitulated the thymopoietic abnormality. These data indicate that specific mesenchymal cells in bone marrow provide key molecular drivers enforcing thymus-seeding progenitor generation and thereby directly link skeletal biology to the production of T cell–based adaptive immunity.
Collapse
Affiliation(s)
- Vionnie W C Yu
- Center for Regenerative Medicine, Massachusetts General Hospital, Boston, MA 02215 Harvard Stem Cell Institute, Cambridge, MA 02215 Department of Stem Cell and Regenerative Biology, Harvard University, Cambridge, MA 02215
| | - Borja Saez
- Center for Regenerative Medicine, Massachusetts General Hospital, Boston, MA 02215 Harvard Stem Cell Institute, Cambridge, MA 02215 Department of Stem Cell and Regenerative Biology, Harvard University, Cambridge, MA 02215
| | - Colleen Cook
- Center for Regenerative Medicine, Massachusetts General Hospital, Boston, MA 02215 Harvard Stem Cell Institute, Cambridge, MA 02215 Department of Stem Cell and Regenerative Biology, Harvard University, Cambridge, MA 02215
| | - Sutada Lotinun
- Department of Oral Medicine, Infection and Immunity, Harvard School of Dental Medicine, Boston, MA 02215 Department of Physiology and STAR on Craniofacial and Skeletal Disorders, Chulalongkorn University, Bangkok 10330, Thailand
| | - Ana Pardo-Saganta
- Center for Regenerative Medicine, Massachusetts General Hospital, Boston, MA 02215 Harvard Stem Cell Institute, Cambridge, MA 02215 Departments of Internal Medicine and Pediatrics, Pulmonary and Critical Care Unit, Massachusetts General Hospital, Boston, MA 02215
| | - Ying-Hua Wang
- Center for Regenerative Medicine, Massachusetts General Hospital, Boston, MA 02215 Harvard Stem Cell Institute, Cambridge, MA 02215 Department of Stem Cell and Regenerative Biology, Harvard University, Cambridge, MA 02215
| | - Stefania Lymperi
- Center for Regenerative Medicine, Massachusetts General Hospital, Boston, MA 02215 Harvard Stem Cell Institute, Cambridge, MA 02215 Department of Stem Cell and Regenerative Biology, Harvard University, Cambridge, MA 02215
| | - Francesca Ferraro
- Center for Regenerative Medicine, Massachusetts General Hospital, Boston, MA 02215 Harvard Stem Cell Institute, Cambridge, MA 02215 Department of Stem Cell and Regenerative Biology, Harvard University, Cambridge, MA 02215
| | - Marc H G P Raaijmakers
- Department of Hematology and Erasmus Stem Cell Institute, Erasmus University Medical Center Cancer Institute, 3015 CE Rotterdam, Netherlands
| | - Joy Y Wu
- Endocrine Unit, Department of Medicine, Massachusetts General Hospital and Harvard Medical School, Boston, MA 02215
| | - Lan Zhou
- Department of Pathology, Case Western Reserve University, Cleveland, OH 44106
| | - Jayaraj Rajagopal
- Center for Regenerative Medicine, Massachusetts General Hospital, Boston, MA 02215 Harvard Stem Cell Institute, Cambridge, MA 02215 Departments of Internal Medicine and Pediatrics, Pulmonary and Critical Care Unit, Massachusetts General Hospital, Boston, MA 02215
| | - Henry M Kronenberg
- Endocrine Unit, Department of Medicine, Massachusetts General Hospital and Harvard Medical School, Boston, MA 02215
| | - Roland Baron
- Department of Oral Medicine, Infection and Immunity, Harvard School of Dental Medicine, Boston, MA 02215 Endocrine Unit, Department of Medicine, Massachusetts General Hospital and Harvard Medical School, Boston, MA 02215
| | - David T Scadden
- Center for Regenerative Medicine, Massachusetts General Hospital, Boston, MA 02215 Harvard Stem Cell Institute, Cambridge, MA 02215 Department of Stem Cell and Regenerative Biology, Harvard University, Cambridge, MA 02215
| |
Collapse
|
18
|
Champhekar A, Damle SS, Freedman G, Carotta S, Nutt SL, Rothenberg EV. Regulation of early T-lineage gene expression and developmental progression by the progenitor cell transcription factor PU.1. Genes Dev 2015; 29:832-48. [PMID: 25846797 PMCID: PMC4403259 DOI: 10.1101/gad.259879.115] [Citation(s) in RCA: 54] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2015] [Accepted: 03/17/2015] [Indexed: 12/28/2022]
Abstract
In the thymus, high PU.1 expression persists through multiple cell divisions in early stages but then falls sharply during T-cell lineage commitment. Here, Champhekar et al. show that PU.1 is needed for full proliferation, restricting access to some non-T fates, and controlling the timing of T-cell developmental progression. Genome-wide transcriptome analysis identifies novel targets of PU.1-positive and PU.1-negative regulation affecting progenitor cell signaling and cell biology and indicating distinct regulatory effects on different subsets of progenitor cell transcription factors. The ETS family transcription factor PU.1 is essential for the development of several blood lineages, including T cells, but its function in intrathymic T-cell precursors has been poorly defined. In the thymus, high PU.1 expression persists through multiple cell divisions in early stages but then falls sharply during T-cell lineage commitment. PU.1 silencing is critical for T-cell commitment, but it has remained unknown how PU.1 activities could contribute positively to T-cell development. Here we employed conditional knockout and modified antagonist PU.1 constructs to perturb PU.1 function stage-specifically in early T cells. We show that PU.1 is needed for full proliferation, restricting access to some non-T fates, and controlling the timing of T-cell developmental progression such that removal or antagonism of endogenous PU.1 allows precocious access to T-cell differentiation. Dominant-negative effects reveal that this repression by PU.1 is mediated indirectly. Genome-wide transcriptome analysis identifies novel targets of PU.1 positive and negative regulation affecting progenitor cell signaling and cell biology and indicating distinct regulatory effects on different subsets of progenitor cell transcription factors. Thus, in addition to supporting early T-cell proliferation, PU.1 regulates the timing of activation of the core T-lineage developmental program.
Collapse
Affiliation(s)
- Ameya Champhekar
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, California 91125, USA
| | - Sagar S Damle
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, California 91125, USA
| | - George Freedman
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, California 91125, USA
| | - Sebastian Carotta
- Division of Molecular Immunology, Walter and Eliza Hall Institute of Medical Research, Melbourne, Victoria 3052, Australia; Department of Medical Biology, The University of Melbourne, Parkville, Victoria 3010, Australia
| | - Stephen L Nutt
- Division of Molecular Immunology, Walter and Eliza Hall Institute of Medical Research, Melbourne, Victoria 3052, Australia; Department of Medical Biology, The University of Melbourne, Parkville, Victoria 3010, Australia
| | - Ellen V Rothenberg
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, California 91125, USA;
| |
Collapse
|
19
|
Steinke FC, Xue HH. From inception to output, Tcf1 and Lef1 safeguard development of T cells and innate immune cells. Immunol Res 2015; 59:45-55. [PMID: 24847765 DOI: 10.1007/s12026-014-8545-9] [Citation(s) in RCA: 54] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Transcription factors have recurring roles during T cell development and activation. Tcf1 and Lef1 are known to be essential for early stages of thymocyte maturation. Recent research has revealed several novel aspects of their functionality. Tcf1 is induced at the very earliest step of specifying hematopoietic progenitors to the T cell lineage as a key target gene downstream of Notch activation. In addition to promoting maturation of T-lineage-committed thymocytes, Tcf1 functions as a tumor suppressor in developing thymocytes, and this is mediated, paradoxically, by restraining Lef1 expression. After positive selection, Tcf1 and Lef1 act together to direct CD4(+)CD8(+) double positive thymocytes to a CD4(+) T cell fate. Although not required for CD8(+) T cell differentiation, Tcf1 and Lef1 cooperate with Runx factors to achieve stable silencing of the Cd4 gene in CD8(+) T cells. Tcf1 is also found to have versatile roles in innate immune cells, which partly mirror its functions in mature T helper cells. Discrepancy in requirements of Tcf1/Lef1 and β-catenin in T cells has been a long-standing enigma. We will review other protein factors interacting with Tcf1 and Lef1 and discuss their regulatory roles independent of β-catenin.
Collapse
Affiliation(s)
- Farrah C Steinke
- Department of Microbiology, Carver College of Medicine, University of Iowa, Iowa City, IA, 52242, USA
| | | |
Collapse
|
20
|
Abstract
The lymphocyte family has expanded significantly in recent years to include not only the adaptive lymphocytes (T cells, B cells) and NK cells, but also several additional innate lymphoid cell (ILC) types. ILCs lack clonally distributed antigen receptors characteristic of adaptive lymphocytes and instead respond exclusively to signaling via germline-encoded receptors. ILCs resemble T cells more closely than any other leukocyte lineage at the transcriptome level and express many elements of the core T cell transcriptional program, including Notch, Gata3, Tcf7, and Bcl11b. We present our current understanding of the shared and distinct transcriptional regulatory mechanisms involved in the development of adaptive T lymphocytes and closely related ILCs. We discuss the possibility that a core set of transcriptional regulators common to ILCs and T cells establish enhancers that enable implementation of closely aligned effector pathways. Studies of the transcriptional regulation of lymphopoiesis will support the development of novel therapeutic approaches to correct early lymphoid developmental defects and aberrant lymphocyte function.
Collapse
Affiliation(s)
- Maria Elena De Obaldia
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania 19104
| | | |
Collapse
|
21
|
Abstract
The hematopoietically expressed homeobox gene, Hhex, is a transcription factor that is important for development of definitive hematopoietic stem cells (HSCs) and B cells, and that causes T-cell leukemia when overexpressed. Here, we have used an Hhex inducible knockout mouse model to study the role of Hhex in adult hematopoiesis. We found that loss of Hhex was tolerated in HSCs and myeloid lineages, but resulted in a progressive loss of B lymphocytes in the circulation. This was accompanied by a complete loss of B-cell progenitors in the bone marrow and of transitional B-cell subsets in the spleen. In addition, transplantation and in vitro culture experiments demonstrated an almost complete failure of Hhex-null HSCs to contribute to lymphoid lineages beyond the common lymphoid precursor stage, including T cells, B cells, NK cells, and dendritic cells. Gene expression analysis of Hhex-deleted progenitors demonstrated deregulated expression of a number of cell cycle regulators. Overexpression of one of these, cyclin D1, could rescue the B-cell developmental potential of Hhex-null lymphoid precursors. Thus, Hhex is a key regulator of early lymphoid development, functioning, at least in part, via regulation of the cell cycle.
Collapse
|
22
|
Smeets MFMA, Wiest DL, Izon DJ. Fli-1 regulates the DN2 to DN3 thymocyte transition and promotes γδ T-cell commitment by enhancing TCR signal strength. Eur J Immunol 2014; 44:2617-24. [PMID: 24935715 PMCID: PMC5242326 DOI: 10.1002/eji.201444442] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2014] [Revised: 05/22/2014] [Accepted: 06/10/2014] [Indexed: 01/05/2023]
Abstract
Friend leukemia integration 1 (Fli-1) is a member of the Ets transcription factor family and is expressed during T-cell development; however, the role Fli-1 plays in early T-cell differentiation has not been elucidated. In this report, we demonstrate that in mouse, Fli-1 overexpression retards the CD4(-) CD8(-) double-negative (DN) to CD4(+) CD8(+) double-positive (DP) transition by deregulating normal DN thymocyte development. Specifically, Fli-1 expression moderates the DN2 and DN3 developmental transitions. We further show that Fli-1 overexpression partially mimics strong TCR signals in developing DN thymocytes and thereby enhances γδ T-cell development. Conversely, Fli-1 knockdown by small hairpin RNA reverses the lineage bias from γδ T cells and directs DN cells to the αβ lineage by attenuating TCR signaling. Therefore, Fli-1 plays a critical role in both the DN2 to DN3 transition and αβ/γδ lineage commitment.
Collapse
MESH Headings
- Animals
- Cells, Cultured
- Mice
- Proto-Oncogene Protein c-fli-1/genetics
- Proto-Oncogene Protein c-fli-1/immunology
- Receptors, Antigen, T-Cell, alpha-beta/genetics
- Receptors, Antigen, T-Cell, alpha-beta/immunology
- Receptors, Antigen, T-Cell, gamma-delta/genetics
- Receptors, Antigen, T-Cell, gamma-delta/immunology
- Signal Transduction/genetics
- Signal Transduction/immunology
- T-Lymphocytes/cytology
- T-Lymphocytes/immunology
- Thymocytes/cytology
- Thymocytes/immunology
Collapse
Affiliation(s)
- Monique F M A Smeets
- Haematology and Leukaemia Unit, St. Vincent's Institute, Fitzroy, Victoria, Australia
| | | | | |
Collapse
|
23
|
Yui MA, Rothenberg EV. Developmental gene networks: a triathlon on the course to T cell identity. Nat Rev Immunol 2014; 14:529-45. [PMID: 25060579 PMCID: PMC4153685 DOI: 10.1038/nri3702] [Citation(s) in RCA: 238] [Impact Index Per Article: 21.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Cells acquire their ultimate identities by activating combinations of transcription factors that initiate and sustain expression of the appropriate cell type-specific genes. T cell development depends on the progression of progenitor cells through three major phases, each of which is associated with distinct transcription factor ensembles that control the recruitment of these cells to the thymus, their proliferation, lineage commitment and responsiveness to T cell receptor signals, all before the allocation of cells to particular effector programmes. All three phases are essential for proper T cell development, as are the mechanisms that determine the boundaries between each phase. Cells that fail to shut off one set of regulators before the next gene network phase is activated are predisposed to leukaemic transformation.
Collapse
Affiliation(s)
- Mary A Yui
- Division of Biology 156-29, California Institute of Technology, Pasadena, California 91125, USA
| | - Ellen V Rothenberg
- Division of Biology 156-29, California Institute of Technology, Pasadena, California 91125, USA
| |
Collapse
|
24
|
Treanor LM, Zhou S, Janke L, Churchman ML, Ma Z, Lu T, Chen SC, Mullighan CG, Sorrentino BP. Interleukin-7 receptor mutants initiate early T cell precursor leukemia in murine thymocyte progenitors with multipotent potential. ACTA ACUST UNITED AC 2014; 211:701-13. [PMID: 24687960 PMCID: PMC3978278 DOI: 10.1084/jem.20122727] [Citation(s) in RCA: 78] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022]
Abstract
Early T cell precursor acute lymphoblastic leukemia (ETP-ALL) exhibits lymphoid, myeloid, and stem cell features and is associated with a poor prognosis. Whole genome sequencing of human ETP-ALL cases has identified recurrent mutations in signaling, histone modification, and hematopoietic development genes but it remains to be determined which of these abnormalities are sufficient to initiate leukemia. We show that activating mutations in the interleukin-7 receptor identified in human pediatric ETP-ALL cases are sufficient to generate ETP-ALL in mice transplanted with primitive transduced thymocytes from p19(Arf-/-) mice. The cellular mechanism by which these mutant receptors induce ETP-ALL is the block of thymocyte differentiation at the double negative 2 stage at which myeloid lineage and T lymphocyte developmental potential coexist. Analyses of samples from pediatric ETP-ALL cases and our murine ETP-ALL model show uniformly high levels of LMO2 expression, very low to undetectable levels of BCL11B expression, and a relative lack of activating NOTCH1 mutations. We report that pharmacological blockade of Jak-Stat signaling with ruxolitinib has significant antileukemic activity in this ETP-ALL model. This new murine model recapitulates several important cellular and molecular features of ETP-ALL and should be useful to further define novel therapeutic approaches for this aggressive leukemia.
Collapse
Affiliation(s)
- Louise M Treanor
- Department of Hematology and 2 Department of Pathology, Division of Experimental Hematology, St. Jude Children's Research Hospital, Memphis, TN 38105
| | | | | | | | | | | | | | | | | |
Collapse
|
25
|
Illegitimate V(D)J recombination-mediated deletions in Notch1 and Bcl11b are not sufficient for extensive clonal expansion and show minimal age or sex bias in frequency or junctional processing. Mutat Res 2014; 761:34-48. [PMID: 24530429 DOI: 10.1016/j.mrfmmm.2014.01.007] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2013] [Revised: 12/23/2013] [Accepted: 01/28/2014] [Indexed: 01/22/2023]
Abstract
Illegitimate V(D)J recombination at oncogenes and tumor suppressor genes is implicated in formation of several T cell malignancies. Notch1 and Bcl11b, genes involved in developing T cell specification, selection, proliferation, and survival, were previously shown to contain hotspots for deletional illegitimate V(D)J recombination associated with radiation-induced thymic lymphoma. Interestingly, these deletions were also observed in wild-type animals. In this study, we conducted frequency, clonality, and junctional processing analyses of Notch1 and Bcl11b deletions during mouse development and compared results to published analyses of authentic V(D)J rearrangements at the T cell receptor beta (TCRβ) locus and illegitimate V(D)J deletions observed at the human, nonimmune HPRT1 locus not involved in T cell malignancies. We detect deletions in Notch1 and Bcl11b in thymic and splenic T cell populations, consistent with cells bearing deletions in the circulating lymphocyte pool. Deletions in thymus can occur in utero, increase in frequency between fetal and postnatal stages, are detected at all ages examined between fetal and 7 months, exhibit only limited clonality (contrasting with previous results in radiation-sensitive mouse strains), and consistent with previous reports are more frequent in Bcl11b, partially explained by relatively high Recombination Signal Information Content (RIC) scores. Deletion junctions in Bcl11b exhibit greater germline nucleotide loss, while in Notch1 palindromic (P) nucleotides are more abundant, although average P nucleotide length is similar for both genes and consistent with results at the TCRβ locus. Non-templated (N) nucleotide insertions appear to increase between fetal and postnatal stages for Notch1, consistent with normal terminal deoxynucleotidyl transferase (TdT) activity; however, neonatal Bcl11b junctions contain elevated levels of N insertions. Finally, contrasting with results at the HPRT1 locus, we find no obvious age or gender bias in junctional processing, and inverted repeats at recessed coding ends (Pr nucleotides) correspond mostly to single-base additions consistent with normal TdT activity.
Collapse
|
26
|
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
|
27
|
Abstract
A systems approach to blood diseases can help make essential contributions to our ability to diagnose, treat, and perhaps even prevent common diseases in humans. Using blood as a window, one can study health and disease through this unique tool box with reactive biological fluids that mirrors the prevailing hemodynamics of the vessel walls and the various blood cell types. Many blood diseases, rare and common, can and have been exploited using systems biology approaches with successful results and therefore ideal models for systems medicine. More importantly, hematopoiesis offers one of the best studied systems with insight into stem cell biology, cellular interaction, development; linage programming and reprogramming that are influenced every day by the most mature and understood regulatory networks.
Collapse
|
28
|
Saki N, Abroun S, Soleimani M, Mortazavi Y, Kaviani S, Arefian E. The roles of miR-146a in the differentiation of Jurkat T-lymphoblasts. Hematology 2013; 19:141-7. [DOI: 10.1179/1607845413y.0000000105] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/31/2022] Open
Affiliation(s)
- Najmaldin Saki
- Department of Hematology and Blood BankingFaculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran
| | - Saeid Abroun
- Department of Hematology and Blood BankingFaculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran
| | - Masoud Soleimani
- Department of Hematology and Blood BankingFaculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran
| | - Yousef Mortazavi
- Department of Pathology Faculty of Medicine, Zanjan University of Medical Sciences, Zanjan, Iran
| | - Saeid Kaviani
- Department of Hematology and Blood BankingFaculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran
| | - Ehsan Arefian
- Department of Stem Cell Biology DepartmentStem Cell Technology Research Center, Tehran, Iran
| |
Collapse
|
29
|
Lotem J, Levanon D, Negreanu V, Leshkowitz D, Friedlander G, Groner Y. Runx3-mediated transcriptional program in cytotoxic lymphocytes. PLoS One 2013; 8:e80467. [PMID: 24236182 PMCID: PMC3827420 DOI: 10.1371/journal.pone.0080467] [Citation(s) in RCA: 49] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2013] [Accepted: 10/02/2013] [Indexed: 12/03/2022] Open
Abstract
The transcription factor Runx3 is highly expressed in CD8+ T and NK cytotoxic lymphocytes and is required for their effective activation and proliferation but molecular insights into the transcription program regulated by Runx3 in these cells are still missing. Using Runx3-ChIP-seq and transcriptome analysis of wild type vs. Runx3-/- primary cells we have now identified Runx3-regulated genes in the two cell types at both resting and IL-2-activated states. Runx3-bound genomic regions in both cell types were distantly located relative to gene transcription start sites and were enriched for RUNX and ETS motifs. Bound genomic regions significantly overlapped T-bet and p300-bound enhancer regions in Runx3-expressing Th1 helper cells. Compared to resting cells, IL-2-activated CD8+ T and NK cells contain three times more Runx3-regulated genes that are common to both cell types. Functional annotation of shared CD8+ T and NK Runx3-regulated genes revealed enrichment for immune-associated terms including lymphocyte activation, proliferation, cytotoxicity, migration and cytokine production, highlighting the role of Runx3 in CD8+ T and NK activated cells.
Collapse
MESH Headings
- Animals
- Core Binding Factor Alpha 3 Subunit/genetics
- Enhancer Elements, Genetic
- Gene Expression Profiling
- Gene Expression Regulation/drug effects
- Histones/metabolism
- Interleukin-2/metabolism
- Interleukin-2/pharmacology
- Killer Cells, Natural/drug effects
- Killer Cells, Natural/immunology
- Killer Cells, Natural/metabolism
- Lymphocyte Activation/genetics
- Lymphocyte Activation/immunology
- Mice
- Mice, Knockout
- Nucleotide Motifs
- Position-Specific Scoring Matrices
- Protein Binding
- Resting Phase, Cell Cycle/genetics
- T-Lymphocytes, Cytotoxic/drug effects
- T-Lymphocytes, Cytotoxic/immunology
- T-Lymphocytes, Cytotoxic/metabolism
- Transcription Factor AP-1/metabolism
- Transcription Initiation Site
- Transcription, Genetic
Collapse
Affiliation(s)
- Joseph Lotem
- Department of Molecular Genetics, Weizmann Institute of Science, Rehovot, Israel
| | - Ditsa Levanon
- Department of Molecular Genetics, Weizmann Institute of Science, Rehovot, Israel
| | - Varda Negreanu
- Department of Molecular Genetics, Weizmann Institute of Science, Rehovot, Israel
| | - Dena Leshkowitz
- Israel National Center for Personalized Medicine Bioinformatics Unit, Weizmann Institute of Science, Rehovot, Israel
| | - Gilgi Friedlander
- Israel National Center for Personalized Medicine Bioinformatics Unit, Weizmann Institute of Science, Rehovot, Israel
| | - Yoram Groner
- Department of Molecular Genetics, Weizmann Institute of Science, Rehovot, Israel
- * E-mail:
| |
Collapse
|
30
|
Russ BE, Prier JE, Rao S, Turner SJ. T cell immunity as a tool for studying epigenetic regulation of cellular differentiation. Front Genet 2013; 4:218. [PMID: 24273551 PMCID: PMC3824109 DOI: 10.3389/fgene.2013.00218] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2013] [Accepted: 10/08/2013] [Indexed: 12/21/2022] Open
Abstract
Cellular differentiation is regulated by the strict spatial and temporal control of gene expression. This is achieved, in part, by regulating changes in histone post-translational modifications (PTMs) and DNA methylation that in turn, impact transcriptional activity. Further, histone PTMs and DNA methylation are often propagated faithfully at cell division (termed epigenetic propagation), and thus contribute to maintaining cellular identity in the absence of signals driving differentiation. Cardinal features of adaptive T cell immunity include the ability to differentiate in response to infection, resulting in acquisition of immune functions required for pathogen clearance; and the ability to maintain this functional capacity in the long-term, allowing more rapid and effective pathogen elimination following re-infection. These characteristics underpin vaccination strategies by effectively establishing a long-lived T cell population that contributes to an immunologically protective state (termed immunological memory). As we discuss in this review, epigenetic mechanisms provide attractive and powerful explanations for key aspects of T cell-mediated immunity – most obviously and notably, immunological memory, because of the capacity of epigenetic circuits to perpetuate cellular identities in the absence of the initial signals that drive differentiation. Indeed, T cell responses to infection are an ideal model system for studying how epigenetic factors shape cellular differentiation and development generally. This review will examine how epigenetic mechanisms regulate T cell function and differentiation, and how these model systems are providing general insights into the epigenetic regulation of gene transcription during cellular differentiation.
Collapse
Affiliation(s)
- Brendan E Russ
- Department of Microbiology and Immunology, The University of Melbourne Parkville, VIC, Australia
| | | | | | | |
Collapse
|
31
|
Abstract
The evolutionary emergence of vertebrates was accompanied by major morphological and functional innovations, including the development of an adaptive immune system. Vertebrate adaptive immunity is based on the clonal expression of somatically diversifying antigen receptors on lymphocytes. This is a common feature of both the jawless and jawed vertebrates , although these two groups of extant vertebrates employ structurally different types of antigen receptors and principal mechanisms for their somatic diversification . These observations suggest that the common vertebrate ancestor must have already possessed a complex immune system, including B- and T-like lymphocyte lineages and primary lymphoid organs, such as the thymus, but possibly lacked the facilities for somatic diversification of antigen receptors. Interestingly, memory formation, previously considered to be a defining feature of adaptive immunity, also occurs in the context of innate immune responses and can even be observed in unicellular organisms, attesting to the convergent evolutionary history of distinct aspects of adaptive immunity.
Collapse
Affiliation(s)
- Thomas Boehm
- Max Planck Institute of Immunobiology and Epigenetics, 79108 Freiburg, Germany; ,
| | | |
Collapse
|
32
|
Rothenberg EV, Champhekar A, Damle S, Del Real MM, Kueh HY, Li L, Yui MA. Transcriptional establishment of cell-type identity: dynamics and causal mechanisms of T-cell lineage commitment. COLD SPRING HARBOR SYMPOSIA ON QUANTITATIVE BIOLOGY 2013; 78:31-41. [PMID: 24135716 DOI: 10.1101/sqb.2013.78.020271] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
Abstract
Precursor cell entry into the T-cell developmental pathway can be divided into two phases by the closure of T-lineage commitment. As cells decide against the last alternative options to the T-cell fate, they turn on the transcription factor Bcl11b and silence expression of a group of multipotent progenitor regulatory factors that include hematopoietic transcription factor PU.1. Functional perturbation tests show that Bcl11b is needed for commitment while PU.1 actively participates in keeping open access to alternative fates, until it is silenced; however, PU.1 and Bcl11b both contribute positively to T-cell development. Our recent work reviewed here sheds light on the transcriptional regulatory network that determines the timing and irreversibility of Bcl11b activation, the ways that Notch signaling from the thymic microenvironment restricts the action of PU.1 to prevent it from diverting cells to non-T fates, and the target genes that PU.1 still regulates under the influence of Notch signaling to contribute to T-cell generation. We argue that T-cell development depends on the sequential operation of two interlaced, but mutually antagonistic, gene regulatory networks, one initially supporting expansion before commitment and the other imposing a "terminal" differentiation process on committed cells.
Collapse
Affiliation(s)
- Ellen V Rothenberg
- Division of Biology 156-29, California Institute of Technology, Pasadena, California 91125
| | - Ameya Champhekar
- Division of Biology 156-29, California Institute of Technology, Pasadena, California 91125
| | - Sagar Damle
- Division of Biology 156-29, California Institute of Technology, Pasadena, California 91125
| | | | - Hao Yuan Kueh
- Division of Biology 156-29, California Institute of Technology, Pasadena, California 91125
| | - Long Li
- Division of Biology 156-29, California Institute of Technology, Pasadena, California 91125
| | - Mary A Yui
- Division of Biology 156-29, California Institute of Technology, Pasadena, California 91125
| |
Collapse
|
33
|
Phelan JD, Saba I, Zeng H, Kosan C, Messer MS, Olsson HA, Fraszczak J, Hildeman DA, Aronow BJ, Möröy T, Grimes HL. Growth factor independent-1 maintains Notch1-dependent transcriptional programming of lymphoid precursors. PLoS Genet 2013; 9:e1003713. [PMID: 24068942 PMCID: PMC3772063 DOI: 10.1371/journal.pgen.1003713] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2012] [Accepted: 06/25/2013] [Indexed: 11/19/2022] Open
Abstract
Growth factor independent 1 (Gfi1) is a transcriptional repressor originally identified as a gene activated in T-cell leukemias induced by Moloney-murine-leukemia virus infection. Notch1 is a transmembrane receptor that is frequently mutated in human T-cell acute lymphoblastic leukemia (T-ALL). Gfi1 is an important factor in the initiation and maintenance of lymphoid leukemias and its deficiency significantly impedes Notch dependent initiation of T-ALL in animal models. Here, we show that immature hematopoietic cells require Gfi1 to competently integrate Notch-activated signaling. Notch1 activation coupled with Gfi1 deficiency early in T-lineage specification leads to a dramatic loss of T-cells, whereas activation in later stages leaves development unaffected. In Gfi1 deficient multipotent precursors, Notch activation induces lethality and is cell autonomous. Further, without Gfi1, multipotent progenitors do not maintain Notch1-activated global expression profiles typical for T-lineage precursors. In agreement with this, we find that both lymphoid-primed multipotent progenitors (LMPP) and early T lineage progenitors (ETP) do not properly form or function in Gfi1−/− mice. These defects correlate with an inability of Gfi1−/− progenitors to activate lymphoid genes, including IL7R, Rag1, Flt3 and Notch1. Our data indicate that Gfi1 is required for hematopoietic precursors to withstand Notch1 activation and to maintain Notch1 dependent transcriptional programming to determine early T-lymphoid lineage identity. Understanding the mechanisms that protect lymphoid cells from transformation is a critical first step in developing therapies against blood cancers. Recently, we demonstrated that the Growth factor independent-1 transcriptional repressor protein is required for cancer development driven by activation of Notch1 signaling. Here, we investigated the mechanisms by which Gfi1 protects lymphoid transformation. Using complex genetic mouse models to delete Gfi1 and activate Notch1, we demonstrate that Gfi1 is required to maintain both the homeostatic levels of Notch1 target genes in normal lymphoid precursors in the bone marrow, as well as to maintain the supraphysiologic levels of Notch1 signaling present in pre-malignant lymphoid progenitors. Consequently, without Gfi1 the pool of premalignant cells available for transformation is depleted. Our data provide additional insight into the multiple mechanisms by which developmental networks may have evolved to protect lymphoid cells from transformation.
Collapse
Affiliation(s)
- James D. Phelan
- Division of Cellular and Molecular Immunology; Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio, United States of America
| | - Ingrid Saba
- Institut de recherches cliniques de Montréal IRCM, Montréal, Québec, Canada
- Département de microbiologie, infectiologie et immunologie, Université de Montréal, Montréal, Québec, Canada
| | - Hui Zeng
- Institute of Infectious Diseases, Beijing Ditan Hospital, Capital Medical University, Beijing, China
| | - Christian Kosan
- Institut de recherches cliniques de Montréal IRCM, Montréal, Québec, Canada
| | - Malynda S. Messer
- Division of Cellular and Molecular Immunology; Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio, United States of America
| | - H. Andre Olsson
- Division of Cellular and Molecular Immunology; Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio, United States of America
| | - Jennifer Fraszczak
- Institut de recherches cliniques de Montréal IRCM, Montréal, Québec, Canada
| | - David A. Hildeman
- Division of Cellular and Molecular Immunology; Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio, United States of America
| | - Bruce J. Aronow
- Division of Biomedical Informatics, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio, United States of America
| | - Tarik Möröy
- Institut de recherches cliniques de Montréal IRCM, Montréal, Québec, Canada
- Département de microbiologie, infectiologie et immunologie, Université de Montréal, Montréal, Québec, Canada
- * E-mail: (TM); (HLG)
| | - H. Leighton Grimes
- Division of Cellular and Molecular Immunology; Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio, United States of America
- Division of Experimental Hematology; Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio, United States of America
- * E-mail: (TM); (HLG)
| |
Collapse
|
34
|
Essential, dose-dependent role for the transcription factor Gata3 in the development of IL-5+ and IL-13+ type 2 innate lymphoid cells. Proc Natl Acad Sci U S A 2013; 110:10240-5. [PMID: 23733962 DOI: 10.1073/pnas.1217158110] [Citation(s) in RCA: 182] [Impact Index Per Article: 15.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Group 2 innate lymphoid cells (ILC2s; also called nuocytes, innate helper cells, or natural helper cells) provide protective immunity during helminth infection and play an important role in influenza-induced and allergic airway hyperreactivity. Whereas the transcription factor GATA binding protein 3 (Gata3) is important for the production of IL-5 and -13 by ILC2s in response to IL-33 or -25 stimulation, it is not known whether Gata3 is required for ILC2 development from hematopoietic stem cells. Here, we show that chimeric mice generated with Gata3-deficient fetal liver hematopoietic stem cells fail to develop systemically dispersed ILC2s. In these chimeric mice, in vivo administration of IL-33 or -25 fails to expand ILC2 numbers or to induce characteristic ILC2-dependent IL-5 or -13 production. Moreover, cell-intrinsic Gata3 expression is required for ILC2 development in vitro and in vivo. Using mutant and transgenic mice in which Gata3 gene copy number is altered, we show that ILC2 generation from common lymphoid progenitors, as well as ILC2 homeostasis and cytokine production, is regulated by Gata3 expression levels in a dose-dependent fashion. Collectively, these results identify Gata3 as a critical early regulator of ILC2 development, thereby extending the paradigm of Gata3-dependent control of type 2 immunity to include both innate and adaptive lymphocytes.
Collapse
|
35
|
Fallah P, Arefian E, Naderi M, Aghaee-Bakhtiari SH, Atashi A, Ahmadi K, Shafiee A, Soleimani M. miR-146a and miR-150 promote the differentiation of CD133+ cells into T-lymphoid lineage. Mol Biol Rep 2013; 40:4713-9. [PMID: 23673476 DOI: 10.1007/s11033-013-2567-6] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2012] [Accepted: 04/29/2013] [Indexed: 12/12/2022]
Abstract
MicroRNAs control the genes involved in hematopoietic stem cell (HSCs) survival, proliferation and differentiation. The over-expression of miR-146 and miR-150 has been reported during differentiation of HSCs into T-lymphoid lineage. Therefore, in this study we evaluated the effect of their over-expression on CD133+ cells differentiation to T cells. miR-146a and miR-150 were separately and jointly transduced to human cord blood derived CD133+ cells (>97% purity). We used qRT-PCR to assess the expression of CD2, CD3ε, CD4, CD8, CD25, T cell receptor alpha (TCR-α) and Ikaros genes in differentiated cells 4 and 8 days after transduction of the miRNAs. Following the over-expression of miR-146a, significant up-regulation of CD2, CD4, CD25 and Ikaros genes were observed (P<0.01). On the other hand, over-expression of miR-150 caused an increase in the expression of Ikaros, CD4, CD25 and TCR-α. To evaluate the combinatorial effect of miR-146a and miR-150, transduction of both miRNAs was concurrently performed which led to increase in the expression of Ikaros, CD4 and CD3 genes. In conclusion, it seems that the effect of miR-150 and miR-146a on the promotion of T cell differentiation is time-dependant. Moreover, miRNAs could be used either as substitutes or complements of the conventional differentiation protocols for higher efficiency.
Collapse
Affiliation(s)
- Parviz Fallah
- Department of Molecular Biology and Genetic Engineering, Stem Cell Technology Research Center, 1997775555, Tehran, Iran
| | | | | | | | | | | | | | | |
Collapse
|
36
|
Abstract
In this issue of Blood, García-Ojeda et al demonstrate a key mechanism that drives T and B lymphocytes to divergent fates from early in their development.
Collapse
|
37
|
Mingueneau M, Kreslavsky T, Gray D, Heng T, Cruse R, Ericson J, Bendall S, Spitzer MH, Nolan GP, Kobayashi K, von Boehmer H, Mathis D, Benoist C, Best AJ, Knell J, Goldrath A, Joic V, Koller D, Shay T, Regev A, Cohen N, Brennan P, Brenner M, Kim F, Nageswara Rao T, Wagers A, Heng T, Ericson J, Rothamel K, Ortiz-Lopez A, Mathis D, Benoist C, Bezman NA, Sun JC, Min-Oo G, Kim CC, Lanier LL, Miller J, Brown B, Merad M, Gautier EL, Jakubzick C, Randolph GJ, Monach P, Blair DA, Dustin ML, Shinton SA, Hardy RR, Laidlaw D, Collins J, Gazit R, Rossi DJ, Malhotra N, Sylvia K, Kang J, Kreslavsky T, Fletcher A, Elpek K, Bellemare-Pelletier A, Malhotra D, Turley S. The transcriptional landscape of αβ T cell differentiation. Nat Immunol 2013; 14:619-32. [PMID: 23644507 PMCID: PMC3660436 DOI: 10.1038/ni.2590] [Citation(s) in RCA: 226] [Impact Index Per Article: 18.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2012] [Accepted: 03/19/2013] [Indexed: 12/11/2022]
Abstract
αβT cell differentiation from thymic precursors is a complex process, explored here with the breadth of ImmGen expression datasets, analyzing how differentiation of thymic precursors gives rise to transcriptomes. After surprisingly gradual changes though early T commitment, transit through the CD4+CD8+ stage involves a shutdown or rare breadth, and correlating tightly with MYC. MHC-driven selection promotes a large-scale transcriptional reactivation. We identify distinct signatures that mark cells destined for positive selection versus apoptotic deletion. Differential expression of surprisingly few genes accompany CD4 or CD8 commitment, a similarity that carries through to peripheral T cells and their activation, revealed by mass cytometry phosphoproteomics. The novel transcripts identified as candidate mediators of key transitions help define the “known unknown” of thymocyte differentiation.
Collapse
Affiliation(s)
- Michael Mingueneau
- Division of Immunology, Department of Microbiology and Immunobiology, Harvard Medical School, Boston, Massachusetts, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
38
|
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
|
39
|
Yui MA, Feng N, Zhang JA, Liaw CY, Rothenberg EV, Longmate JA. Loss of T cell progenitor checkpoint control underlies leukemia initiation in Rag1-deficient nonobese diabetic mice. THE JOURNAL OF IMMUNOLOGY 2013; 190:3276-88. [PMID: 23440410 DOI: 10.4049/jimmunol.1202970] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
NOD mice exhibit major defects in the earliest stages of T cell development in the thymus. Genome-wide genetic and transcriptome analyses were used to investigate the origins and consequences of an early T cell developmental checkpoint breakthrough in Rag1-deficient NOD mice. Quantitative trait locus analysis mapped the presence of checkpoint breakthrough cells to several known NOD diabetes susceptibility regions, particularly insulin-dependent diabetes susceptibility genes (Idd)9/11 on chromosome 4, suggesting common genetic origins for T cell defects affecting this trait and autoimmunity. Genome-wide RNA deep-sequencing of NOD and B6 Rag1-deficient thymocytes revealed the effects of genetic background prior to breakthrough, as well as the cellular consequences of the breakthrough. Transcriptome comparison between the two strains showed enrichment in differentially expressed signal transduction genes, prominently tyrosine kinase and actin-binding genes, in accord with their divergent sensitivities to activating signals. Emerging NOD breakthrough cells aberrantly expressed both stem cell-associated proto-oncogenes, such as Lmo2, Hhex, Lyl1, and Kit, which are normally repressed at the commitment checkpoint, and post-β-selection checkpoint genes, including Cd2 and Cd5. Coexpression of genes characteristic of multipotent progenitors and more mature T cells persists in the expanding population of thymocytes and in the thymic leukemias that emerge with age in these mice. These results show that Rag1-deficient NOD thymocytes have T cell defects that can collapse regulatory boundaries at two early T cell checkpoints, which may predispose them to both leukemia and autoimmunity.
Collapse
Affiliation(s)
- Mary A Yui
- Division of Biology, California Institute of Technology, Pasadena, CA 91125, USA.
| | | | | | | | | | | |
Collapse
|
40
|
Mangel M, Bonsall MB. Stem cell biology is population biology: differentiation of hematopoietic multipotent progenitors to common lymphoid and myeloid progenitors. Theor Biol Med Model 2013; 10:5. [PMID: 23327512 PMCID: PMC3765094 DOI: 10.1186/1742-4682-10-5] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2012] [Accepted: 12/22/2012] [Indexed: 01/28/2023] Open
Abstract
The hematopoietic stem cell (HSC) system is a demand control system, with the demand coming from the organism, since the products of the common myeloid and lymphoid progenitor (CMP, CLP respectively) cells are essential for activity and defense against disease. We show how ideas from population biology (combining population dynamics and evolutionary considerations) can illuminate the feedback control of the HSC system by the fully differentiated products, which has recently been verified experimentally. We develop models for the penultimate differentiation of HSC Multipotent Progenitors (MPPs) into CLP and CMP and introduce two concepts from population biology into stem cell biology. The first concept is the Multipotent Progenitor Commitment Response (MPCR) which is the probability that a multipotent progenitor cell follows a CLP route rather than a CMP route. The second concept is the link between the MPCR and a measure of Darwinian fitness associated with organismal performance and the levels of differentiated lymphoid and myeloid cells. We show that many MPCRs are consistent with homeostasis, but that they will lead to different dynamics of cells and signals following a wound or injury and thus have different consequences for Darwinian fitness. We show how coupling considerations of life history to dynamics of the HSC system and its products allows one to compute the selective pressures on cellular processes. We discuss ways that this framework can be used and extended.
Collapse
Affiliation(s)
- Marc Mangel
- Department of Applied Mathematics and Statistics, University of California, Santa Cruz, CA, USA.
| | | |
Collapse
|
41
|
GATA-3 promotes T-cell specification by repressing B-cell potential in pro-T cells in mice. Blood 2013; 121:1749-59. [PMID: 23287858 DOI: 10.1182/blood-2012-06-440065] [Citation(s) in RCA: 76] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Transcription factors orchestrate T-lineage differentiation in the thymus. One critical checkpoint involves Notch1 signaling that instructs T-cell commitment at the expense of the B-lineage program. While GATA-3 is required for T-cell specification, its mechanism of action is poorly understood. We show that GATA-3 works in concert with Notch1 to commit thymic progenitors to the T-cell lineage via 2 distinct pathways. First, GATA-3 orchestrates a transcriptional “repertoire” that is required for thymocyte maturation up to and beyond the pro-T-cell stage. Second, GATA-3 critically suppresses a latent B-cell potential in pro–T cells. As such, GATA-3 is essential to sealing in Notch-induced T-cell fate in early thymocyte precursors by promoting T-cell identity through the repression of alternative developmental options.
Collapse
|
42
|
Yu Y, Wang J, Khaled W, Burke S, Li P, Chen X, Yang W, Jenkins NA, Copeland NG, Zhang S, Liu P. Bcl11a is essential for lymphoid development and negatively regulates p53. ACTA ACUST UNITED AC 2012; 209:2467-83. [PMID: 23230003 PMCID: PMC3526365 DOI: 10.1084/jem.20121846] [Citation(s) in RCA: 152] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Bcl11a regulates development of lymphoid cells in adult mice in part by inhibiting expression of p53. Transcription factors play important roles in lymphopoiesis. We have previously demonstrated that Bcl11a is essential for normal lymphocyte development in the mouse embryo. We report here that, in the adult mouse, Bcl11a is expressed in most hematopoietic cells and is highly enriched in B cells, early T cell progenitors, common lymphoid progenitors (CLPs), and hematopoietic stem cells (HSCs). In the adult mouse, Bcl11a deletion causes apoptosis in early B cells and CLPs and completely abolishes the lymphoid development potential of HSCs to B, T, and NK cells. Myeloid development, in contrast, is not obviously affected by the loss of Bcl11a. Bcl11a regulates expression of Bcl2, Bcl2-xL, and Mdm2, which inhibits p53 activities. Overexpression of Bcl2 and Mdm2, or p53 deficiency, rescues both lethality and proliferative defects in Bcl11a-deficient early B cells and enables the mutant CLPs to differentiate to lymphocytes. Bcl11a is therefore essential for lymphopoiesis and negatively regulates p53 activities. Deletion of Bcl11a may represent a new approach for generating a mouse model that completely lacks an adaptive immune system.
Collapse
Affiliation(s)
- Yong Yu
- Key Laboratory of Agricultural Animal Genetics, Breeding, and Reproduction of Ministry of Education, Huazhong Agricultural University, Wuhan 430070, China
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
43
|
The transcription factor Lyl-1 regulates lymphoid specification and the maintenance of early T lineage progenitors. Nat Immunol 2012; 13:761-9. [PMID: 22772404 DOI: 10.1038/ni.2365] [Citation(s) in RCA: 41] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2011] [Accepted: 06/04/2012] [Indexed: 01/19/2023]
Abstract
Thymopoiesis depends on the recruitment and expansion of bone marrow-derived progenitor populations; tight regulation of these processes is required for maintenance of the homeostasis of the T lineage. Lyl-1, a transcription factor that regulates hematopoietic progenitors, is expressed in thymocyte progenitors until T cell commitment. Here we demonstrate a requirement for Lyl-1 in lymphoid specification and the maintenance of early T lineage progenitors (ETPs). Lyl-1 deficiency resulted in profound defects in the generation of lymphoid-primed multipotent progenitors (LMPPs), common lymphoid progenitors (CLPs) and ETPs. Lyl-1-deficient ETPs and thymocyte progenitors at the CD4(-)CD8(-) double-negative 2 (DN2) stage showed more apoptosis, blocked differentiation and impaired population expansion. We identified Gfi1 as a critical transcriptional target of Lyl-1-mediated lymphopoiesis of T cells. Thus, Lyl-1 is a pivotal component of a transcriptional program that controls the lymphoid specification and maintenance of ETPs.
Collapse
|
44
|
Belyaev NN, Biró J, Athanasakis D, Fernandez-Reyes D, Potocnik AJ. Global transcriptional analysis of primitive thymocytes reveals accelerated dynamics of T cell specification in fetal stages. Immunogenetics 2012; 64:591-604. [PMID: 22581009 PMCID: PMC3395349 DOI: 10.1007/s00251-012-0620-6] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2011] [Accepted: 04/24/2012] [Indexed: 11/24/2022]
Abstract
T cell development constitutes a multistage process allowing the dissection of events resulting in cellular commitment and functional specification in a specialized microenvironment. This process is guided by the appropriate expression of regulatory genetic factors like transcriptional activators or repressors which are, in part, dependent on instructive signals of the microenvironment. To date, it remains unclear whether exactly the same genetic mechanism acts in adult compared to fetal T cell development. In order to directly compare T cell commitment during adult and fetal differentiation, we isolated subsequent stages of intrathymic subpopulations starting with early canonical T cell progenitors up to irreversibly committed T cell precursors. The genome-wide analysis revealed several distinct gene clusters with a specific pattern of gene regulation for each subset. The largest cluster contained genes upregulated after transition through the most primitive pool into the next transitory population with a consistently elevated expression of elements associated with ongoing T cell fate specification, like Gata3 and Tcf7, in fetal progenitors. Furthermore, adult and fetal T cell progenitors occupied distinct "transcriptional territories" revealing a precise land map of the progression to final T cell commitment operating in different developmental windows. The presence and/or elevated expression of elements associated with an ongoing establishment of a T cell signature in the most primitive fetal subset is highly suggestive for an extrathymic initiation of T cell specification and underlines the fundamental differences in fetal versus adult lymphopoiesis.
Collapse
Affiliation(s)
- Nikolai N Belyaev
- Division of Molecular Immunology, MRC National Institute for Medical Research, London, UK
| | | | | | | | | |
Collapse
|
45
|
Fibroblast growth factor-7 partially reverses murine thymocyte progenitor aging by repression of Ink4a. Blood 2012; 119:5715-21. [PMID: 22555975 DOI: 10.1182/blood-2011-12-400002] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
Involution of the thymus results in reduced production of naive T cells, and this in turn is thought to contribute to impaired immunity in the elderly. Early T-cell progenitors (ETPs), the most immature intrathymic T-cell precursors, harvested from the involuted thymus exhibit a diminished proliferative potential and increased rate of apoptosis and as a result their number is significantly reduced. In the present study, we show that these age-induced alterations result in part from increased expression of the Ink4a tumor-suppressor gene in ETPs. We also show that repression of Ink4a in aged ETPs results in their partial rejuvenation and that this can be accomplished by in vivo fibroblast growth factor 7 administration. These results define a genetic basis for thymocyte progenitor aging and demonstrate that the senescence-associated gene Ink4a can be pharmacologically repressed in ETPs to partially reverse the effects of aging.
Collapse
|
46
|
Berent-Maoz B, Montecino-Rodriguez E, Dorshkind K. Genetic regulation of thymocyte progenitor aging. Semin Immunol 2012; 24:303-8. [PMID: 22559986 DOI: 10.1016/j.smim.2012.04.006] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/17/2012] [Revised: 03/27/2012] [Accepted: 04/09/2012] [Indexed: 02/06/2023]
Abstract
The number of T cell progenitors is significantly reduced in the involuted thymus, and the growth and developmental potential of the few cells that are present is severely attenuated. This review provides an overview of how aging affects T cell precursors before and following entry into the thymus and discusses the age-related genetic changes that may occur in them. Finally, interventions that rejuvenate thymopoiesis in the elderly by targeting T cell progenitors are discussed.
Collapse
Affiliation(s)
- Beata Berent-Maoz
- Department of Pathology and Laboratory Medicine, David Geffen School of Medicine at UCLA, Los Angeles, CA 90095, United States
| | | | | |
Collapse
|
47
|
Intrathymic programming of effector fates in three molecularly distinct γδ T cell subtypes. Nat Immunol 2012; 13:511-8. [PMID: 22473038 PMCID: PMC3427768 DOI: 10.1038/ni.2247] [Citation(s) in RCA: 155] [Impact Index Per Article: 11.9] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2011] [Accepted: 01/17/2012] [Indexed: 12/13/2022]
Abstract
Innate γδ T cells function in the early phase of immune responses. Although innate γδ T cells have often been studied as one homogenous population, they can be functionally classified into effector subsets on the basis of the production of signature cytokines, analogous to adaptive helper T cell subsets. However, unlike the function of adaptive T cells, γδ effector T cell function correlates with genomically encoded T cell antigen receptor (TCR) chains, which suggests that clonal TCR selection is not the main determinant of the differentiation of γδ effector cells. A high-resolution transcriptome analysis of all emergent γδ thymocyte subsets segregated on the basis of use of the TCR γ-chain or δ-chain indicated the existence of three separate subtypes of γδ effector cells in the thymus. The immature γδ subsets were distinguished by unique transcription-factor modules that program effector function.
Collapse
|
48
|
Rothenberg EV. Transcriptional drivers of the T-cell lineage program. Curr Opin Immunol 2012; 24:132-8. [PMID: 22264928 DOI: 10.1016/j.coi.2011.12.012] [Citation(s) in RCA: 55] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2011] [Accepted: 12/31/2011] [Indexed: 11/28/2022]
Abstract
The T-cell development program is specifically triggered by Notch-Delta signaling, but most transcription factors needed to establish T-cell lineage identity also have crossover roles in other hematopoietic lineages. This factor sharing complicates full definition of the core gene regulatory circuits required for T-cell specification. But new advances illuminate the roles of three of the most T-cell specific transcription factors. Commitment to the T-cell lineage is now shown to depend on Bcl11b, while initiation of the T-cell differentiation program begins earlier with the induction of TCF-1 (Tcf7 gene product) and GATA-3. Several reports now reveal how TCF-1 and GATA-3 are mobilized in early T cells and the pathways for their T-lineage specific effects.
Collapse
Affiliation(s)
- Ellen V Rothenberg
- Division of Biology 156-29, California Institute of Technology, Pasadena, CA 91125, USA.
| |
Collapse
|
49
|
Kominami R. Role of the transcription factor Bcl11b in development and lymphomagenesis. PROCEEDINGS OF THE JAPAN ACADEMY. SERIES B, PHYSICAL AND BIOLOGICAL SCIENCES 2012; 88:72-87. [PMID: 22450536 PMCID: PMC3365246 DOI: 10.2183/pjab.88.72] [Citation(s) in RCA: 52] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 10/07/2011] [Accepted: 01/11/2012] [Indexed: 05/31/2023]
Abstract
Bcl11b is a lineage-specific transcription factor expressed in various cell types and its expression is important for development of T cells, neurons and others. On the other hand, Bcl11b is a haploinsufficient tumor suppressor and loss of a Bcl11b allele provides susceptibility to mouse thymic lymphoma and human T-cell acute lymphoblastic leukemia. Although there are many transcription factors affecting both cell differentiation and cancer development, Bcl11b has several unique properties. This review describes phenotypes given by loss of Bcl11b and roles of Bcl11b in cell proliferation, differentiation and apoptosis, taking tissue development and lymphomagenesis into consideration.
Collapse
Affiliation(s)
- Ryo Kominami
- Department of Molecular Genetics, Niigata University Graduate School of Medical and Dental Sciences, Niigata, Japan.
| |
Collapse
|
50
|
Expression of BMPRIA on human thymic NK cell precursors: role of BMP signaling in intrathymic NK cell development. Blood 2011; 119:1861-71. [PMID: 22210872 DOI: 10.1182/blood-2011-07-370650] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
The bone morphogenetic protein (BMP) signaling pathway regulates survival, proliferation, and differentiation of several cell types in multiple tissues, including the thymus. Previous reports have shown that BMP signaling negatively regulates T-cell development. Here, we study the subpopulation of early human intrathymic progenitors expressing the type IA BMP receptor (BMPRIA) and provide evidence that CD34(+)CD1a(-)BMPRIA(+) precursor cells mostly express surface cell markers and transcription factors typically associated with NK cell lineage. These CD34(+) cells mostly differentiate into functional CD56(+) natural killer (NK) cells when they are cocultured with thymic stromal cells in chimeric human-mouse fetal thymic organ cultures and also in the presence of SCF and IL-15. Moreover, autocrine BMP signaling can promote the differentiation of thymic NK cells by regulating the expression of key transcription factors required for NK cell lineage (eg, Id3 and Nfil3) as well as one of the components of IL-15 receptor, CD122. Subsequently, the resulting population of IL-15-responsive NK cell precursors can be expanded by IL-15, whose action is mediated by BMP signaling during the last steps of thymic NK cell differentiation. Our results strongly suggest that BMPRIA expression identifies human thymic NK cell precursors and that BMP signaling is relevant for NK cell differentiation in the human thymus.
Collapse
|