104 results on '"Barbara L. Kee"'
Search Results
2. E Protein Transcription Factors as Suppressors of T Lymphocyte Acute Lymphoblastic Leukemia
- Author
-
Geoffrey Parriott and Barbara L. Kee
- Subjects
Leukemia ,E protein ,TAL1 ,LYL1 ,murine ,T lymphocyte ,Immunologic diseases. Allergy ,RC581-607 - Abstract
T Lymphocyte Acute Lymphoblastic Leukemia (ALL) is an aggressive disease arising from transformation of T lymphocytes during their development. The mutation spectrum of T-ALL has revealed critical regulators of the growth and differentiation of normal and leukemic T lymphocytes. Approximately, 60% of T-ALLs show aberrant expression of the hematopoietic stem cell-associated helix-loop-helix transcription factors TAL1 and LYL1. TAL1 and LYL1 function in multiprotein complexes that regulate gene expression in T-ALL but they also antagonize the function of the E protein homodimers that are critical regulators of T cell development. Mice lacking E2A, or ectopically expressing TAL1, LYL1, or other inhibitors of E protein function in T cell progenitors, also succumb to an aggressive T-ALL-like disease highlighting that E proteins promote T cell development and suppress leukemogenesis. In this review, we discuss the role of E2A in T cell development and how alterations in E protein function underlie leukemogenesis. We focus on the role of TAL1 and LYL1 and the genes that are dysregulated in E2a-/- T cell progenitors that contribute to human T-ALL. These studies reveal novel mechanisms of transformation and provide insights into potential therapeutic targets for intervention in this disease.
- Published
- 2022
- Full Text
- View/download PDF
3. Oncogenic and Tumor Suppressor Functions for Lymphoid Enhancer Factor 1 in E2a-/- T Acute Lymphoblastic Leukemia
- Author
-
Tiffany Carr, Stephanie McGregor, Sheila Dias, Mihalis Verykokakis, Michelle M. Le Beau, Hai-Hui Xue, Mikael Sigvardsson, Elizabeth T. Bartom, and Barbara L. Kee
- Subjects
E2a ,Lef1 ,leukemia ,thymus ,lymphocyte ,Immunologic diseases. Allergy ,RC581-607 - Abstract
T lymphocyte acute lymphoblastic leukemia (T-ALL) is a heterogeneous disease affecting T cells at multiple stages of their development and is characterized by frequent genomic alterations. The transcription factor LEF1 is inactivated through mutation in a subset of T-ALL cases but elevated LEF1 expression and activating mutations have also been identified in this disease. Here we show, in a murine model of T-ALL arising due to E2a inactivation, that the developmental timing of Lef1 mutation impacts its ability to function as a cooperative tumor suppressor or oncogene. T cell transformation in the presence of LEF1 allows leukemic cells to become addicted to its presence. In contrast, deletion prior to transformation both accelerates leukemogenesis and results in leukemic cells with altered expression of genes controlling receptor-signaling pathways. Our data demonstrate that the developmental timing of Lef1 mutations impact its apparent oncogenic or tumor suppressive characteristics and demonstrate the utility of mouse models for understanding the cooperation and consequence of mutational order in leukemogenesis.
- Published
- 2022
- Full Text
- View/download PDF
4. Batf Pioneers the Reorganization of Chromatin in Developing Effector T Cells via Ets1-Dependent Recruitment of Ctcf
- Author
-
Duy Pham, Carson E. Moseley, Min Gao, Daniel Savic, Colleen J. Winstead, Mengxi Sun, Barbara L. Kee, Richard M. Myers, Casey T. Weaver, and Robin D. Hatton
- Subjects
Biology (General) ,QH301-705.5 - Abstract
Summary: The basic leucine zipper transcription factor activating transcription factor-like (Batf) contributes to transcriptional programming of multiple effector T cells and is required for T helper 17 (Th17) and T follicular helper (Tfh) cell development. Here, we examine mechanisms by which Batf initiates gene transcription in developing effector CD4 T cells. We find that, in addition to its pioneering function, Batf controls developmentally regulated recruitment of the architectural factor Ctcf to promote chromatin looping that is associated with lineage-specific gene transcription. The chromatin-organizing actions of Batf are largely dependent on Ets1, which appears to be indispensable for the Batf-dependent recruitment of Ctcf. Moreover, most of the Batf-dependent sites to which Ctcf is recruited lie outside of activating protein-1-interferon regulatory factor (Ap-1-Irf) composite elements (AICEs), indicating that direct involvement of Batf-Irf complexes is not required. These results identify a cooperative role for Batf, Ets1, and Ctcf in chromatin reorganization that underpins the transcriptional programming of effector T cells. : Pham et al. uncover mechanisms by which Batf restructures the chromatin landscape during CD4+ effector T cell differentiation. Batf controls Ctcf recruitment to lineage-specifying gene loci in an Ets1-dependent manner to promote chromatin looping and lineage-specific gene transcription, thereby identifying a heretofore unknown cooperativity of these factors in effector T cell development. Keywords: CD4 T cell differentiation, Batf, Ets1, Ctcf, chromatin accessibility, chromatin looping
- Published
- 2019
- Full Text
- View/download PDF
5. Quantitative control of Ets1 dosage by a multi-enhancer hub promotes Th1 cell differentiation and protects from allergic inflammation
- Author
-
Aditi Chandra, Sora Yoon, Michaël F. Michieletto, Naomi Goldman, Emily K. Ferrari, Maryam Abedi, Isabelle Johnson, Maria Fasolino, Kenneth Pham, Leonel Joannas, Barbara L. Kee, Jorge Henao-Mejia, and Golnaz Vahedi
- Subjects
Infectious Diseases ,Immunology ,Immunology and Allergy - Published
- 2023
- Full Text
- View/download PDF
6. Loss of thymocyte competition underlies the tumor suppressive functions of the E2a transcription factor in T lymphocyte acute lymphoblastic leukemia
- Author
-
Geoffrey Parriott, Emma Hegermiller, Rosemary E. Morman, Cameron Frank, Caner Saygin, Wendy Stock, Elizabeth T. Bartom, and Barbara L. Kee
- Subjects
Article - Abstract
T lymphocyte acute lymphoblastic leukemia (T-ALL) is frequently associated with increased expression of the E protein transcription factor inhibitors TAL1 and LYL1. In mouse models, ectopic expression of Tal1 or Lyl1 in T cell progenitors or inactivation of E2a, is sufficient to predispose mice to develop T-ALL. How E2a suppresses thymocyte transformation is currently unknown. Here, we show that early deletion ofE2a, prior to the DN3 stage, was required for robust leukemogenesis and was associated with alterations in thymus cellularity, T cell differentiation, and gene expression in immature CD4+CD8+ thymocytes. Introduction of wild-type thymocytes into mice with early deletion ofE2aprevented leukemogenesis, or delayed disease onset, and impacted the expression of multiple genes associated with transformation and genome instability. Our data indicate that E2a suppresses leukemogenesis by promoting T cell development and enforcing inter-thymocyte competition, a mechanism that is emerging as a safeguard against thymocyte transformation. These studies have implications for understanding how multiple essential regulators of T cell development suppress T-ALL and support the hypothesis that thymus cellularity is a determinant of leukemogenesis.
- Published
- 2023
- Full Text
- View/download PDF
7. A multi-enhancer hub at theEts1locus controls T cell differentiation and allergic inflammation through 3D genome topology
- Author
-
Aditi Chandra, Sora Yoon, Michael F. Michieletto, Naomi Goldman, Emily K. Ferrari, Maria Fasolino, Leonel Joannas, Barbara L. Kee, Jorge Henao-Mejia, and Golnaz Vahedi
- Abstract
Multi-enhancer hubs are spatial clusters of enhancers which have been recently characterized across numerous developmental programs. Yet, the functional relevance of these three-dimensional (3D) structures is poorly understood. Here we show that the multiplicity of enhancers interacting with the transcription factorEts1is essential to control the precise expression level of this gene in response to cellular cues, and the failure to do so can lead to allergic diseases. Focusing on T cells as a model, we identified a highly connected multi-enhancer hub at theEts1locus, comprising a noncoding regulatory element that is a hotspot for sequence variation associated with allergic diseases. We deleted this hotspot and found that the multi-enhancer connectivity is dispensable for T cell development but required for CD4+T helper (Th1) differentiation in response to changes in the cytokine milieu. Mice lacking this hotspot are thus protected from Th1-mediated colitis but demonstrate an overt allergic response to house dust mites, a T cell-mediated response which is dampened by Th1 cells. Mechanistically, the multi-enhancer hub controls the expression level ofEts1that is dispensable for the active enhancer landscape but required for the Th1-specific genome topology through recruitment of CTCF. Together, we establish a paradigm for the functional and mechanistic relevance of multi-enhancer hubs controlling cellular competence to respond specifically to an inductive cue.
- Published
- 2022
- Full Text
- View/download PDF
8. Transforming growth factor-β promotes the postselection thymic development and peripheral function of interferon-γ-producing invariant natural killer T cells
- Author
-
Roxroy C. Morgan, Cameron Frank, Munmun Greger, Mikael Sigvardsson, Elizabeth T. Bartom, and Barbara L. Kee
- Abstract
Interferon-γ producing invariant natural killer T (iNKT1) cells are lipid reactive innate-like lymphocytes that are resident in the thymus and peripheral tissues where they protect against pathogenic infection. The thymic functions of iNKT1 cells are not fully elucidated but subsets of thymic iNKT cells modulate CD8 T cell, dendritic cell, B cell and thymic epithelial cell numbers or function. Here we show that a subset of thymic iNKT1 cells require transforming growth factor (TGF)-β induced signals for their development and for expression of residency associated adhesion receptors. Liver and spleen iNKT1 cells do not share this TGF-β gene signature but nonetheless TGF-β is required for optimal liver iNKT1 cell function. Our findings provide insight into the heterogeneity of mechanisms guiding iNKT1 cell development in different tissues and suggest a close association between a subset of iNKT1 cells and TGF-β producing cells in the thymus.
- Published
- 2022
- Full Text
- View/download PDF
9. Combinatorial ETS1-Dependent Control of Oncogenic NOTCH1 Enhancers in T-cell Leukemia
- Author
-
Yiran Liu, Cher Sha, Alberto Ambesi-Impiombato, Michael C. Ostrowski, Erin Kim, Theresa M. Keeley, Jahnavi K. Nalamolu, Qing Wang, Mark Y. Chiang, Adolfo A. Ferrando, Ran Yan, Rohan Kodgule, Russell J.H. Ryan, Arvind Rao, Nicholas Kunnath, Linda C. Samuelson, Barbara L. Kee, Giusy Della Gatta, Anna C. McCarter, Rork Kuick, Ashley Melnick, and Mengxi Sun
- Subjects
Leukemia, T-Cell ,Carcinogenesis ,Effector ,Lymphoblastic Leukemia ,T-cell leukemia ,General Medicine ,Biology ,Precursor T-Cell Lymphoblastic Leukemia-Lymphoma ,Article ,Chromatin ,Proto-Oncogene Protein c-ets-1 ,Mice ,ETS1 ,Antineoplastic Combined Chemotherapy Protocols ,Cancer research ,Animals ,Receptor, Notch1 ,Enhancer ,Gene ,Transcription factor ,Signal Transduction - Abstract
Notch activation is highly prevalent among cancers, in particular T-cell acute lymphoblastic leukemia (T-ALL). However, the use of pan-Notch inhibitors to treat cancers has been hampered by adverse effects, particularly intestinal toxicities. To circumvent this barrier in T-ALL, we aimed to inhibit ETS1, a developmentally important T-cell transcription factor previously shown to cobind Notch response elements. Using complementary genetic approaches in mouse models, we show that ablation of Ets1 leads to strong Notch-mediated suppressive effects on T-cell development and leukemogenesis but milder intestinal effects than pan-Notch inhibitors. Mechanistically, genome-wide chromatin profiling studies demonstrate that Ets1 inactivation impairs recruitment of multiple Notch-associated factors and Notch-dependent activation of transcriptional elements controlling major Notch-driven oncogenic effector pathways. These results uncover previously unrecognized hierarchical heterogeneity of Notch-controlled genes and point to Ets1-mediated enucleation of Notch–Rbpj transcriptional complexes as a target for developing specific anti-Notch therapies in T-ALL that circumvent the barriers of pan-Notch inhibition. Significance: Notch signaling controls developmentally important and tissue-specific activities, raising barriers for developing anti-Notch therapies. Pivoting away from pan-Notch inhibitors, we show antileukemic but less toxic effects of targeting ETS1, a T-cell NOTCH1 cofactor. These results demonstrate the feasibility of context-dependent suppression of NOTCH1 programs for the treatment of T-ALL. This article is highlighted in the In This Issue feature, p. 127
- Published
- 2020
- Full Text
- View/download PDF
10. Ezh2 Represses Transcription of Innate Lymphoid Genes in B Lymphocyte Progenitors and Maintains the B-2 Cell Fate
- Author
-
Mikael Sigvardsson, Elizabeth T. Bartom, Jennifer A. Jacobsen, and Barbara L. Kee
- Subjects
Transcription, Genetic ,Lymphocyte ,Immunology ,Gene Expression ,Cell fate determination ,Methylation ,Article ,Histones ,Mice ,Gene expression ,medicine ,Animals ,Immunology and Allergy ,Cell Lineage ,Enhancer of Zeste Homolog 2 Protein ,Psychological repression ,Gene ,Transcription factor ,B-Lymphocytes ,biology ,Precursor Cells, B-Lymphoid ,EZH2 ,RNA-Binding Proteins ,Cell Differentiation ,Immunity, Innate ,Cell biology ,Mice, Inbred C57BL ,medicine.anatomical_structure ,Histone ,biology.protein ,Transcription Factors - Abstract
Lymphocyte lineage specification and commitment requires the activation of lineage-specific genes and repression of alternative lineage genes, respectively. The mechanisms governing alternative lineage gene repression and commitment in lymphocytes are largely unknown. In this study, we demonstrate that Ezh2, which represses gene expression through methylation of histone 3 lysine 27, was essential for repression of numerous genes, including genes encoding innate lymphocyte transcription factors, specifically in murine B lymphocyte progenitors, but these cells maintained their B lymphocyte identity. However, adult Ezh2-deficient B lymphocytes expressed Lin28b, which encodes an RNA-binding protein associated with fetal hematopoietic gene expression programs, and these cells acquired a fetal B-1 lymphocyte phenotype in vitro and in vivo. Therefore, Ezh2 coordinates the repression of multiple gene programs in B lymphocytes and maintains the adult B-2 cell fate.
- Published
- 2020
- Full Text
- View/download PDF
11. Genomic and Transcriptional Mechanisms Governing Innate-like T Lymphocyte Development
- Author
-
Roxroy C. Morgan and Barbara L. Kee
- Subjects
Immunology ,Immunology and Allergy ,Cytokines ,Natural Killer T-Cells ,Cell Differentiation ,Genomics ,Immunity, Innate - Abstract
Innate-like lymphocytes are a subset of lymphoid cells that function as a first line of defense against microbial infection. These cells are activated by proinflammatory cytokines or broadly expressed receptors and are able to rapidly perform their effector functions owing to a uniquely primed chromatin state that is acquired as a part of their developmental program. These cells function in many organs to protect against disease, but they release cytokines and cytotoxic mediators that can also lead to severe tissue pathologies. Therefore, harnessing the capabilities of these cells for therapeutic interventions will require a deep understanding of how these cells develop and regulate their effector functions. In this review we discuss recent advances in the identification of the transcription factors and the genomic regions that guide the development and function of invariant NKT cells and we highlight related mechanisms in other innate-like lymphocytes.
- Published
- 2022
12. Molecular Switches of the Immune System: The E Protein/Id Axis in Hematopoietic Development and Function
- Author
-
Michele Kay Anderson, Juan Carlos Zuniga-Pflucker, Mikael Sigvardsson, and Barbara L. Kee
- Published
- 2022
- Full Text
- View/download PDF
13. Oncogenic and Tumor Suppressor Functions for Lymphoid Enhancer Factor 1 in
- Author
-
Tiffany, Carr, Stephanie, McGregor, Sheila, Dias, Mihalis, Verykokakis, Michelle M, Le Beau, Hai-Hui, Xue, Mikael, Sigvardsson, Elizabeth T, Bartom, and Barbara L, Kee
- Subjects
Mice ,Lymphoid Enhancer-Binding Factor 1 ,Animals ,Oncogenes ,Precursor Cell Lymphoblastic Leukemia-Lymphoma ,Precursor T-Cell Lymphoblastic Leukemia-Lymphoma ,TCF Transcription Factors ,Transcription Factors - Abstract
T lymphocyte acute lymphoblastic leukemia (T-ALL) is a heterogeneous disease affecting T cells at multiple stages of their development and is characterized by frequent genomic alterations. The transcription factor LEF1 is inactivated through mutation in a subset of T-ALL cases but elevated LEF1 expression and activating mutations have also been identified in this disease. Here we show, in a murine model of T-ALL arising due to
- Published
- 2021
14. Oncogenic and Tumor Suppressor Functions for Lymphoid Enhancer Factor 1 in a Murine Model of T Acute Lymphoblastic Leukemia
- Author
-
Hai-Hui Xue, Sheila Dias, Barbara L. Kee, Michelle M. LeBeau, Elizabeth T. Bartom, Stephanie M. McGregor, Mihalis Verykokakis, and Tiffany Carr
- Subjects
Mutation ,Oncogene ,T-Cell Transformation ,T lymphocyte ,Biology ,medicine.disease_cause ,law.invention ,T Acute Lymphoblastic Leukemia ,law ,embryonic structures ,Cancer research ,medicine ,Suppressor ,Enhancer ,Transcription factor - Abstract
T lymphocyte acute lymphoblastic leukemia (T-ALL) is a heterogeneous disease affecting T cells at multiple stages of their development and is characterized by frequent genomic alterations. The transcription factor LEF1 is inactivated through mutation in a subset of T-ALL cases but elevated LEF1 expression and activating mutations have also been identified in this disease. Here we show, in a murine model of T-ALL, that the developmental timing of Lef1 mutation impacts its ability to function as a cooperative tumor suppressor or oncogene. T cell transformation in the presence of LEF1 allows leukemic cells to become addicted to its presence. In contrast, deletion prior to transformation both accelerates leukemogenesis and results in leukemic cells with altered expression of genes controlling receptor signaling pathways. Our data demonstrate that the developmental timing of Lef1 mutations impact its apparent oncogenic or tumor suppressive characteristics and demonstrate the utility of mouse models for understanding the cooperation and consequence of mutational order in leukemogenesis.
- Published
- 2021
- Full Text
- View/download PDF
15. Cryptic activation of an Irf8 enhancer governs cDC1 fate specification
- Author
-
Kenneth M. Murphy, Gary E. Grajales-Reyes, Rodney D. Newberry, Carlos G. Briseño, Prachi Bagadia, Howard Y. Chang, Theresa L. Murphy, Vivek Durai, Swapneel J. Patel, Xiao Huang, Hiromi Tagoh, Jeffrey M. Granja, Miriam Wöhner, Jesse T. Davidson, Barbara L. Kee, Ansuman T. Satpathy, Tian Tian Liu, Renee Wu, Meinrad Busslinger, Arifumi Iwata, and Devesha H. Kulkarni
- Subjects
0301 basic medicine ,Cellular differentiation ,Immunology ,Biology ,Article ,Monocytes ,03 medical and health sciences ,Mice ,0302 clinical medicine ,Tumor Cells, Cultured ,Immunology and Allergy ,Animals ,Cell Lineage ,Progenitor cell ,Enhancer ,Transcription factor ,Regulation of gene expression ,Mice, Knockout ,Macrophages ,Stem Cells ,Cell Differentiation ,Dendritic cell ,Dendritic Cells ,Cell biology ,Chromatin ,Mice, Inbred C57BL ,030104 developmental biology ,Enhancer Elements, Genetic ,Gene Expression Regulation ,Interferon Regulatory Factors ,IRF8 ,CRISPR-Cas Systems ,030215 immunology - Abstract
Induction of the transcription factor Irf8 in the common dendritic cell progenitor (CDP) is required for classical type 1 dendritic cell (cDC1) fate specification, but the mechanisms controlling this induction are unclear. In the present study Irf8 enhancers were identified via chromatin profiling of dendritic cells and CRISPR/Cas9 genome editing was used to assess their roles in Irf8 regulation. An enhancer 32 kilobases (kb) downstream of the Irf8 transcriptional start site (+32-kb Irf8) that was active in mature cDC1s was required for the development of this lineage, but not for its specification. Instead, a +41-kb Irf8 enhancer, previously thought to be active only in plasmacytoid dendritic cells, was found to also be transiently accessible in cDC1 progenitors, and deleting this enhancer prevented the induction of Irf8 in CDPs and abolished cDC1 specification. Thus, cryptic activation of the +41-kb Irf8 enhancer in dendritic cell progenitors is responsible for cDC1 fate specification. The transcription factor IRF8 is essential for classical type 1 dendritic cell (cDC1) development. Murphy and colleagues investigate in detail the molecular control of cDC1 fate specification by systematically unpicking the IRF8 enhancer regions.
- Published
- 2019
16. Cutting Edge: Lymphomyeloid-Primed Progenitor Cell Fates Are Controlled by the Transcription Factor Tal1
- Author
-
Sheila Dias, Michael K. Okoreeh, Barbara L. Kee, Mikael Sigvardsson, Elisabeth T. Bartom, Renée F. de Pooter, and Munmun Chowdhury
- Subjects
Myeloid ,Immunology ,hemic and immune systems ,Biology ,Cell biology ,T Acute Lymphoblastic Leukemia ,03 medical and health sciences ,Haematopoiesis ,0302 clinical medicine ,medicine.anatomical_structure ,hemic and lymphatic diseases ,medicine ,Immunology and Allergy ,Stem cell ,Progenitor cell ,Transcription factor ,030215 immunology ,Megakaryopoiesis ,TAL1 - Abstract
Lymphoid specification is the process by which hematopoietic stem cells (HSCs) and their progeny become restricted to differentiation through the lymphoid lineages. The basic helix-loop-helix transcription factors E2A and Lyl1 form a complex that promotes lymphoid specification. In this study, we demonstrate that Tal1, a Lyl1-related basic helix-loop-helix transcription factor that promotes T acute lymphoblastic leukemia and is required for HSC specification, erythropoiesis, and megakaryopoiesis, is a negative regulator of murine lymphoid specification. We demonstrate that Tal1 limits the expression of multiple E2A target genes in HSCs and controls the balance of myeloid versus T lymphocyte differentiation potential in lymphomyeloid-primed progenitors. Our data provide insight into the mechanisms controlling lymphocyte specification and may reveal a basis for the unique functions of Tal1 and Lyl1 in T acute lymphoblastic leukemia.
- Published
- 2019
- Full Text
- View/download PDF
17. The transcriptional repressor ID2 supports natural killer cell maturation by controlling TCF1 amplitude
- Author
-
Mengxi Sun, Mark Maienschein-Cline, Rosemary E. Morman, Zhong Yin Li, Mikael Sigvardsson, Elizabeth T. Bartom, Emma Hegermiller, and Barbara L. Kee
- Subjects
Transcription, Genetic ,Cellular differentiation ,Innate Immunity and Inflammation ,Immunology ,Cell ,Gene Expression ,chemical and pharmacologic phenomena ,Biology ,Article ,Natural killer cell ,Interferon-gamma ,Mice ,Cell surface receptor ,medicine ,Animals ,Immunology and Allergy ,Hepatocyte Nuclear Factor 1-alpha ,Melanoma ,Transcription factor ,Inhibitor of Differentiation Protein 2 ,Cell growth ,Effector ,Cell Differentiation ,Hematopoiesis ,Cell biology ,Oncolytic virus ,Killer Cells, Natural ,Mice, Inbred C57BL ,medicine.anatomical_structure ,Immunologi ,Cytokines ,Transcription Factors - Abstract
ID2 is an essential transcription factor for natural killer cell development. Through epistasis analysis, Li et al. show that ID2 controls the amplitude of TCF1 and allows for its temporal downregulation, thereby supporting natural killer cell maturation., Gaining a mechanistic understanding of the expansion and maturation program of natural killer (NK) cells will provide opportunities for harnessing their inflammation-inducing and oncolytic capacity for therapeutic purposes. Here, we demonstrated that ID2, a transcriptional regulatory protein constitutively expressed in NK cells, supports NK cell effector maturation by controlling the amplitude and temporal dynamics of the transcription factor TCF1. TCF1 promotes immature NK cell expansion and restrains differentiation. The increased TCF1 expression in ID2-deficient NK cells arrests their maturation and alters cell surface receptor expression. Moreover, TCF1 limits NK cell functions, such as cytokine-induced IFN-γ production and the ability to clear metastatic melanoma in ID2-deficient NK cells. Our data demonstrate that ID2 sets a threshold for TCF1 during NK cell development, thus controlling the balance of immature and terminally differentiated cells that support future NK cell responses., Graphical Abstract
- Published
- 2021
- Full Text
- View/download PDF
18. It's a Phase That EBF1 Is Going Through
- Author
-
Barbara L. Kee
- Subjects
0301 basic medicine ,B-Lymphocytes ,Prions ,Immunology ,biochemical phenomena, metabolism, and nutrition ,Biology ,humanities ,Chromatin ,Domain (software engineering) ,Cell biology ,03 medical and health sciences ,030104 developmental biology ,0302 clinical medicine ,Infectious Diseases ,Gene Expression Regulation ,030220 oncology & carcinogenesis ,Phase (matter) ,Immunology and Allergy ,Nucleosome ,Transcription factor ,Transcription Factors - Abstract
EBF1 is a pioneer transcription factor involved in B lymphocyte specification. In this issue of Immunity, Wang et al. localize EBF1's pioneering activity to a prion-like domain that mediates recruitment of the nucleosome remodeler Brg1 and FUS-assisted liquid-liquid phase separation.
- Published
- 2020
19. Transcriptional regulation of natural killer cell development and maturation
- Author
-
Barbara L, Kee, Rosmary E, Morman, and Mengxi, Sun
- Subjects
Killer Cells, Natural ,Mice ,Gene Expression Regulation ,Animals ,Cytokines ,Humans ,Cell Differentiation ,Immunity, Innate ,Transcription Factors - Abstract
Natural killer cells are lymphocytes that respond rapidly to intracellular pathogens or cancer/stressed cells by producing pro-inflammatory cytokines or chemokines and by killing target cells through direct cytolysis. NK cells are distinct from B and T lymphocytes in that they become activated through a series of broadly expressed germ line encoded activating and inhibitory receptors or through the actions of inflammatory cytokines. They are the founding member of the innate lymphoid cell family, which mirror the functions of T lymphocytes, with NK cells being the innate counterpart to CD8 T lymphocytes. Despite the functional relationship between NK cells and CD8 T cells, the mechanisms controlling their specification, differentiation and maturation are distinct, with NK cells emerging from multipotent lymphoid progenitors in the bone marrow under the control of a unique transcriptional program. Over the past few years, substantial progress has been made in understanding the developmental pathways and the factors involved in generating mature and functional NK cells. NK cells have immense therapeutic potential and understanding how to acquire large numbers of functional cells and how to endow them with potent activity to control hematopoietic and non-hematopoietic malignancies and autoimmunity is a major clinical goal. In this review, we examine basic aspects of conventional NK cell development in mice and humans and discuss multiple transcription factors that are known to guide the development of these cells.
- Published
- 2020
20. Transcriptional and epigenetic regulation of innate-like T lymphocyte development
- Author
-
Barbara L. Kee and Mihalis Verykokakis
- Subjects
0301 basic medicine ,Transcription, Genetic ,Lymphocyte ,T cell ,Immunology ,Population ,Biology ,Article ,Epigenesis, Genetic ,03 medical and health sciences ,0302 clinical medicine ,T-Lymphocyte Subsets ,medicine ,Animals ,Humans ,Immunology and Allergy ,Epigenetics ,Lymphopoiesis ,education ,Regulation of gene expression ,education.field_of_study ,T-cell receptor ,Gene Expression Regulation, Developmental ,Cell Differentiation ,T lymphocyte ,Immunity, Innate ,Cell biology ,030104 developmental biology ,medicine.anatomical_structure ,Natural Killer T-Cells ,030215 immunology - Abstract
Invariant Natural Killer T (iNKT) cells are a heterogeneous innate T cell population that recognizes lipid antigens. Despite the monospecific nature of their T cell receptor, iNKT cells differentiate into stable sublineages during thymic development, prior to foreign antigen encounter. How iNKT cell subsets acquire and maintain their functional programs is a central question in innate lymphocyte biology. Global transcriptional and epigenetic profiling of iNKT subsets has provided insights into the internal wiring of these subsets that defines their identity. Comparison of the iNKT transcriptional programs with those of other adaptive and innate lymphocyte lineages revealed common core regulatory circuits that may dictate effector functions. In this review, we summarize recent advances on the molecular mechanisms involved in iNKT cell development.
- Published
- 2018
- Full Text
- View/download PDF
21. Murine thymic NK cells are distinct from ILC1s and have unique transcription factor requirements
- Author
-
April Bell, Barbara L. Kee, Loris Zamai, Mengxi Sun, Erin C. Zook, Renée F. de Pooter, and Sara Gabrielli
- Subjects
0301 basic medicine ,CD11b Antigen ,Intracellular parasite ,Cellular differentiation ,Immunology ,Cell ,Innate lymphoid cell ,Biology ,Cell biology ,03 medical and health sciences ,030104 developmental biology ,0302 clinical medicine ,medicine.anatomical_structure ,Integrin alpha M ,ETS1 ,medicine ,biology.protein ,Immunology and Allergy ,Transcription factor ,030215 immunology - Abstract
Group 1 innate lymphoid cells include natural killer (NK) cells and ILC1s, which mediate the response to intracellular pathogens. Thymic NK (tNK) cells were described with hybrid features of immature NK cells and ILC1 but whether these cells are related to NK cells or ILC1 has not been fully investigated. We report that murine tNK cells expressed the NK-cell associated transcription factor EOMES and developed independent of the essential ILC1 factor TBET, confirming their placement within the NK lineage. Moreover, tNK cells resemble NK cells rather than ILC1 in their requirements for the E protein transcription factor inhibitor ID2. We provide further insight into the mechanisms governing tNK-cell development by showing that the transcription factor ETS1 prevented tNK cell acquisition of the conventional NK-cell maturation markers CD11b and KLRG1. Our data reveal few ILC1 in the thymus and clarify the identity and developmental requirements of tNK cells.
- Published
- 2017
- Full Text
- View/download PDF
22. Applying the TOR(C)QUE in iNKT cells: A new twist in an old tale
- Author
-
Barbara L. Kee and Mihalis Verykokakis
- Subjects
0301 basic medicine ,Immunology ,mTORC1 ,Biology ,mTORC2 ,Article ,Mice ,03 medical and health sciences ,T-Lymphocyte Subsets ,Animals ,Immunology and Allergy ,Cytotoxic T cell ,Transcription factor ,Protein kinase B ,PI3K/AKT/mTOR pathway ,Mice, Knockout ,Effector ,TOR Serine-Threonine Kinases ,digestive, oral, and skin physiology ,Cell Differentiation ,Natural killer T cell ,Cell biology ,030104 developmental biology ,Natural Killer T-Cells ,Interleukin-4 - Abstract
The mammalian Target of Rapamycin (mTOR) protein controls the machinery necessary for T-cell activation, differentiation, and memory formation, as a component of mTOR complex 1 (mTORC1) and mTORC2, which function both downstream and upstream of AKT. Invariant natural killer T (iNKT) cells are a unique T-cell subset that exist in a primed state, capable of rapid activation, and produce large quantities of cytokines. iNKT-cell effector differentiation is dependent on the mTORC1 complex; however, the requirements for mTORC2 in iNKT cells have been controversial. In this issue, Sklarz et al. [Eur. J. Immunol. 2017. 47: 516-526] provide a careful analysis of the requirements for the mTORC2 component Rictor in iNKT cells, providing a new twist in this unfolding tale. The authors demonstrate that Rictor is required for iNKT-cell proliferation and survival during the key stage of intrathymic expansion and that Rictor supports the development of NKT17 cells, an effector subset which depends on the transcription factor RORγt and produces interleukin (IL)-17, in both the thymus and the lung. IL-4-producing NKT2 cells develop in the absence of Rictor but the cytotoxic potential of iNKT cells is Rictor-dependent.
- Published
- 2017
- Full Text
- View/download PDF
23. Transcriptional regulation of natural killer cell development and maturation
- Author
-
Mengxi Sun, Rosmary E Morman, and Barbara L. Kee
- Subjects
Chemokine ,NFIL3 ,Innate lymphoid cell ,Biology ,Cell biology ,Natural killer cell ,03 medical and health sciences ,Haematopoiesis ,0302 clinical medicine ,medicine.anatomical_structure ,medicine ,biology.protein ,Cytotoxic T cell ,Bone marrow ,CD8 ,030215 immunology - Abstract
Natural killer cells are lymphocytes that respond rapidly to intracellular pathogens or cancer/stressed cells by producing pro-inflammatory cytokines or chemokines and by killing target cells through direct cytolysis. NK cells are distinct from B and T lymphocytes in that they become activated through a series of broadly expressed germ line encoded activating and inhibitory receptors or through the actions of inflammatory cytokines. They are the founding member of the innate lymphoid cell family, which mirror the functions of T lymphocytes, with NK cells being the innate counterpart to CD8 T lymphocytes. Despite the functional relationship between NK cells and CD8 T cells, the mechanisms controlling their specification, differentiation and maturation are distinct, with NK cells emerging from multipotent lymphoid progenitors in the bone marrow under the control of a unique transcriptional program. Over the past few years, substantial progress has been made in understanding the developmental pathways and the factors involved in generating mature and functional NK cells. NK cells have immense therapeutic potential and understanding how to acquire large numbers of functional cells and how to endow them with potent activity to control hematopoietic and non-hematopoietic malignancies and autoimmunity is a major clinical goal. In this review, we examine basic aspects of conventional NK cell development in mice and humans and discuss multiple transcription factors that are known to guide the development of these cells.
- Published
- 2020
- Full Text
- View/download PDF
24. The transcription factor BCL-6 controls early development of innate-like T cells
- Author
-
Barbara L. Kee, Marianthi Gioulbasani, Alexander L. Dent, Mihalis Verykokakis, Sofia Grammenoudi, Panagiotis Moulos, Pantelis Hatzis, Mikael Sigvardsson, and Alexandros Galaras
- Subjects
0301 basic medicine ,Naive T cell ,Cellular differentiation ,T cell ,Immunology ,Population ,Biology ,Lymphocyte Activation ,Mucosal-Associated Invariant T Cells ,Article ,03 medical and health sciences ,Mice ,0302 clinical medicine ,medicine ,Immunology and Allergy ,Animals ,Promyelocytic Leukemia Zinc Finger Protein ,education ,Clonal Selection, Antigen-Mediated ,Transcription factor ,Cells, Cultured ,Mice, Knockout ,education.field_of_study ,Gene Expression Regulation, Developmental ,Cell Differentiation ,T lymphocyte ,BCL6 ,Chromatin ,Immunity, Innate ,Cell biology ,Mice, Inbred C57BL ,030104 developmental biology ,medicine.anatomical_structure ,Proto-Oncogene Proteins c-bcl-6 ,Natural Killer T-Cells ,030215 immunology - Abstract
Innate T cells, including invariant natural killer T (iNKT) and mucosal-associated innate T (MAIT) cells, are a heterogeneous T lymphocyte population with effector properties pre-programmed during their thymic differentiation. How this program is initiated is currently unclear. Here, we show that the transcription factor BCL-6 was transiently expressed in iNKT cells upon exit from positive selection and was required for their proper development beyond stage 0. Notably, development of MAIT cells was also impaired in the absence of Bcl6. BCL-6–deficient iNKT cells had reduced expression of genes that were associated with the innate T cell lineage, including Zbtb16, which encodes PLZF, and PLZF-targeted genes. BCL-6 contributed to a chromatin accessibility landscape that was permissive for the expression of development-related genes and inhibitory for genes associated with naïve T cell programs. Our results revealed novel functions for BCL-6 and illuminated how this transcription factor controls early iNKT cell development.
- Published
- 2019
25. Lnc'ing Id2 to ILC1
- Author
-
Mengxi Sun and Barbara L. Kee
- Subjects
0301 basic medicine ,Immunology ,Cell ,Innate lymphoid cell ,RNA ,Biology ,Regulatory Sequences, Nucleic Acid ,Molecular biology ,Immunity, Innate ,Article ,body regions ,03 medical and health sciences ,030104 developmental biology ,Infectious Diseases ,medicine.anatomical_structure ,Immunity ,Regulatory sequence ,medicine ,Transcriptional Repressor ,Nucleic acid ,Immunology and Allergy ,Humans ,RNA, Long Noncoding ,Lymphocytes ,skin and connective tissue diseases - Abstract
Commitment to the innate lymphoid cells (ILC) lineage is determined by Id2, a transcriptional regulator that antagonizes T and B cell-specific gene expression programs. Yet how Id2 expression is regulated in each ILC subset remains poorly understood. We identified a cis-regulatory element demarcated by a long non-coding RNA (lncRNA) that controls the function and lineage identity of group 1 ILCs, while being dispensable for early ILC development and homeostasis of ILC2s and ILC3s. The locus encoding this lncRNA, which we termed Rroid, directly interacted with the promoter of its neighboring gene, Id2, in group 1 ILCs. Moreover, the Rroid locus, but not the lncRNA itself, controlled the identity and function of ILC1s by promoting chromatin accessibility and deposition of STAT5 at the promoter of Id2 in response to interleukin (IL)-15. Thus, non-coding elements responsive to extracellular cues unique to each ILC subset represent a key regulatory layer for controlling the identity and function of ILCs.
- Published
- 2017
26. Transcription factor ID2 prevents E proteins from enforcing a naïve T lymphocyte gene program during NK cell development
- Author
-
Aimee M. Beaulieu, Erin C. Zook, Mikael Sigvardsson, Joseph C. Sun, Zhong Yin Li, Mark Maienschein-Cline, Yiying Xu, Barbara L. Kee, Renée F. de Pooter, Mihalis Verykokakis, and Anna Lasorella
- Subjects
0301 basic medicine ,medicine.medical_treatment ,T-Lymphocytes ,Immunology ,Biology ,Article ,03 medical and health sciences ,Chemokine receptor ,0302 clinical medicine ,medicine ,Basic Helix-Loop-Helix Transcription Factors ,Cytotoxic T cell ,Animals ,Transcription factor ,Inhibitor of Differentiation Protein 2 ,Mice, Knockout ,Naive T-Lymphocyte ,Innate lymphoid cell ,Cell Differentiation ,General Medicine ,Chromatin ,Cell biology ,Killer Cells, Natural ,Mice, Inbred C57BL ,030104 developmental biology ,Cytokine ,Gene Expression Regulation ,Inhibitor of Differentiation Proteins ,CD8 ,030215 immunology - Abstract
All innate lymphoid cells (ILC) require the small helix-loop-helix transcription factor ID2 but the functions of ID2 are not well understood in these cells. Here we show that mature natural killer (NK) cells, the prototypic ILC, developed in mice lacking ID2 but remained as precursor CD27(+)CD11b(−) cells that failed to differentiate into CD27(−)CD11b(+) cytotoxic effectors. We show that ID2 limited chromatin accessibility at E protein binding sites near naïve T lymphocyte-associated genes including multiple chemokine receptors, cytokine receptors, and signaling molecules and altered the NK cell response to inflammatory cytokines. In the absence of ID2, CD27(+)CD11b(−) NK cells expressed ID3, a helix-loop-helix protein associated with naïve T cells, and they transitioned from a CD8 memory-precursor-like to a naïve-like chromatin accessibility state. We demonstrate that ID3 was required for the development of ID2-deficient NK cells indicating that completely unfettered E protein function is incompatible with NK cell development. These data solidify the roles of ID2 and ID3 as mediators of effector and naïve gene programs, respectively, and revealed a critical role for ID2 in promoting a chromatin state and transcriptional program in CD27(+)CD11b(−) NK cells that supports cytotoxic effector differentiation and cytokine responses.
- Published
- 2017
27. EZH2 regulates the developmental timing of effectors of the pre-antigen receptor checkpoints
- Author
-
Barbara L. Kee, Jennifer Woodard, Malay Mandal, Jennifer A. Jacobsen, Elizabeth T. Bartom, Mikael Sigvardsson, and Marcus R. Clark
- Subjects
0301 basic medicine ,T cell ,Lymphocyte ,T-Lymphocytes ,Immunology ,macromolecular substances ,Biology ,Adaptive Immunity ,Article ,03 medical and health sciences ,Mice ,0302 clinical medicine ,medicine ,Immunology and Allergy ,Animals ,Enhancer of Zeste Homolog 2 Protein ,Lymphopoiesis ,Cyclin-Dependent Kinase Inhibitor p16 ,Regulation of gene expression ,B-Lymphocytes ,Effector ,EZH2 ,Acquired immune system ,Genes, p53 ,Killer Cells, Natural ,Receptors, Antigen ,030104 developmental biology ,medicine.anatomical_structure ,Gene Expression Regulation ,Histone methyltransferase ,Cancer research ,030215 immunology - Abstract
The histone methyltransferase EZH2 is required for B and T cell development; however, the molecular mechanisms underlying this requirement remain elusive. In a murine model of lymphoid-specific EZH2 deficiency we found that EZH2 was required for proper development of adaptive, but not innate, lymphoid cells. In adaptive lymphoid cells EZH2 prevented the premature expression of Cdkn2a and the consequent stabilization of p53, an effector of the pre–Ag receptor checkpoints. Deletion of Cdkn2a in EZH2-deficient lymphocytes prevented p53 stabilization, extended lymphocyte survival, and restored differentiation resulting in the generation of mature B and T lymphocytes. Our results uncover a crucial role for EZH2 in adaptive lymphocytes to control the developmental timing of effectors of the pre–Ag receptor checkpoints.
- Published
- 2017
28. Dose-Dependent Requirements for ETS1 in Invariant Natural Killer T (iNKT) Cell Development
- Author
-
Roxroy C Morgan and Barbara L Kee
- Subjects
Immunology ,Immunology and Allergy - Abstract
iNKT cells are innate-like T cells that use an invariant T cell receptor to recognize glycolipids presented by the non-classical major histocompatibility protein CD1d. Upon recognition, iNKTs rapidly produce cytokines that stimulate innate cells to produce cytokines and prime adaptive cells for activation upon antigenic stimulation. Studies in Ets1 germline-deficient mice showed that the transcription factor ETS1 is required for the development of CD4+ NK1.1+ NKT cells. Development of CD1d tetramers and the recent elucidation of multiple effector iNKT cell subsets has improved our understanding of NKT cell development and function, and revealed that CD4+ NK1.1+ iNKT cells are a minor subset of total NKTs. Using conditional deletion for Ets1 in thymocytes, we show that Ets1 is a dose dependent regulator of all iNKT cells and appears to be particularly important for development of interferon-g-producing iNKT1 cells. Deletion of Ets1 in developing iNKT1 cells using Tbx21Cre demonstrated the critical role of ETS1 in this iNKT cell effector subset. Our findings set the stage for future studies focused on the molecular mechanism by which ETS1 controls iNKT cell development and function.
- Published
- 2019
- Full Text
- View/download PDF
29. The transcription factor TCF1 is a critical target of ID2 that limits natural killer cell maturation
- Author
-
Barbara L Kee, Rosemary Morman, Mengxi Sun, and Zhong-Yin Li
- Subjects
Immunology ,Immunology and Allergy - Abstract
ID2 is an inhibitor of the E protein transcription factors that is expressed in all innate lymphoid cells. In natural killer (NK) cells, ID2 prevents immature cells from acquiring a naïve T cell – like gene program that restricts NK cell maturation and function. Here we show that Tcf7 (encoding the transcription factor TCF1) is the critical E protein target gene in immature NK cells that maintains NK cell progenitors. TCF1 binds to a subset of key “naïve T cell” genes that are normally expressed at low, but detectable, levels in immature NK cells and whose expression is deregulated in the absence of ID2. Importantly, combined deficiency of TCF1 and ID2 restores expression of these naïve T cell genes to near wild-type levels. Moreover, in the absence of TCF1, ID2-deficient NK cells mature and acquire functional capacity but fail to expand to wild-type numbers. Our data indicate that ID2 is essential for NK cell maturation because it controls TCF1, whose heightened expression arrests NK cell maturation and activates a naïve – like gene program.
- Published
- 2019
- Full Text
- View/download PDF
30. Essential Functions for ID Proteins at Multiple Checkpoints in Invariant NKT Cell Development
- Author
-
Mikael Sigvardsson, Veena Krishnamoorthy, Mihalis Verykokakis, Antonio Iavarone, Barbara L. Kee, and Anna Lasorella
- Subjects
Cell growth ,Immunology ,Innate lymphoid cell ,Cell ,Gene rearrangement ,Biology ,Natural killer T cell ,Cell biology ,medicine.anatomical_structure ,medicine ,Immunology and Allergy ,Lymphopoiesis ,Transcription factor ,Interleukin 4 - Abstract
Invariant NKT (iNKT) cells display characteristics of both adaptive and innate lymphoid cells (ILCs). Like other ILCs, iNKT cells constitutively express ID proteins, which antagonize the E protein transcription factors that are essential for adaptive lymphocyte development. However, unlike ILCs, ID2 is not essential for thymic iNKT cell development. In this study, we demonstrated that ID2 and ID3 redundantly promoted iNKT cell lineage specification involving the induction of the signature transcription factor PLZF and that ID3 was critical for development of TBET-dependent NKT1 cells. In contrast, both ID2 and ID3 limited iNKT cell numbers by enforcing the postselection checkpoint in conventional thymocytes. Therefore, iNKT cells show both adaptive and innate-like requirements for ID proteins at distinct checkpoints during iNKT cell development.
- Published
- 2013
- Full Text
- View/download PDF
31. Murine thymic NK cells are distinct from ILC1s and have unique transcription factor requirements
- Author
-
Sara, Gabrielli, Mengxi, Sun, April, Bell, Erin C, Zook, Renee F, de Pooter, Loris, Zamai, and Barbara L, Kee
- Subjects
CD11b Antigen ,Thymocytes ,Cell Differentiation ,Thymus Gland ,Immunity, Innate ,Killer Cells, Natural ,Proto-Oncogene Protein c-ets-1 ,Mice ,Animals ,Cell Lineage ,Lectins, C-Type ,Lymphocytes ,Receptors, Immunologic ,T-Box Domain Proteins ,Inhibitor of Differentiation Protein 2 ,Transcription Factors - Abstract
Group 1 innate lymphoid cells include natural killer (NK) cells and ILC1s, which mediate the response to intracellular pathogens. Thymic NK (tNK) cells were described with hybrid features of immature NK cells and ILC1 but whether these cells are related to NK cells or ILC1 has not been fully investigated. We report that murine tNK cells expressed the NK-cell associated transcription factor EOMES and developed independent of the essential ILC1 factor TBET, confirming their placement within the NK lineage. Moreover, tNK cells resemble NK cells rather than ILC1 in their requirements for the E protein transcription factor inhibitor ID2. We provide further insight into the mechanisms governing tNK-cell development by showing that the transcription factor ETS1 prevented tNK cell acquisition of the conventional NK-cell maturation markers CD11b and KLRG1. Our data reveal few ILC1 in the thymus and clarify the identity and developmental requirements of tNK cells.
- Published
- 2016
32. Development of innate lymphoid cells
- Author
-
Erin C. Zook and Barbara L. Kee
- Subjects
0301 basic medicine ,Cellular differentiation ,T-Lymphocytes ,Immunology ,Biology ,03 medical and health sciences ,0302 clinical medicine ,Immunity ,medicine ,Immunology and Allergy ,Animals ,Humans ,Lymphoid progenitors ,Cell Lineage ,Lymphocytes ,Colitis ,skin and connective tissue diseases ,Transcription factor ,Innate immune system ,Innate lymphoid cell ,Cell Differentiation ,Lymphoid Progenitor Cells ,medicine.disease ,Immunity, Innate ,body regions ,030104 developmental biology ,Cytokines ,Function (biology) ,030215 immunology ,Transcription Factors - Abstract
Innate lymphoid cells (ILCs) are a family of immune effector cells that have important roles in host defense, metabolic homeostasis and tissue repair but can also contribute to inflammatory diseases such as asthma and colitis. These cells can be categorized into three groups on the basis of the transcription factors that direct their function and the cytokines they produce, which parallel the effector functions of T lymphocytes. The hierarchy of cell-fate-restriction events that occur as common lymphoid progenitors become committed to each of the ILC lineages further underscores the relationship between these innate immune cells and T lymphocytes. In this Review we discuss the developmental program of ILCs and transcription factors that guide ILC lineage specification and commitment.
- Published
- 2016
33. Epigenetic repression of the Igk locus by STAT5-mediated Ezh2 recruitment
- Author
-
Aaron R. Dinner, Marcus R. Clark, Sarah E. Powers, Mark Maienschein-Cline, Elizabeth T. Bartom, Barbara L. Kee, Malay Mandal, and Keith M. Hamel
- Subjects
Immunology ,Immunoglobulins ,macromolecular substances ,Biology ,Binding, Competitive ,Methylation ,Article ,Epigenesis, Genetic ,Histones ,Mice ,03 medical and health sciences ,Histone H3 ,0302 clinical medicine ,Chlorocebus aethiops ,Histone methylation ,Histone H2A ,Basic Helix-Loop-Helix Transcription Factors ,STAT5 Transcription Factor ,Animals ,Cluster Analysis ,Immunology and Allergy ,Enhancer of Zeste Homolog 2 Protein ,Enhancer ,Transcription factor ,030304 developmental biology ,Mice, Knockout ,Mice, Inbred BALB C ,0303 health sciences ,Binding Sites ,Receptors, Interleukin-7 ,Base Sequence ,Gene Expression Profiling ,EZH2 ,Polycomb Repressive Complex 2 ,Histone-Lysine N-Methyltransferase ,Molecular biology ,Mice, Inbred C57BL ,Gene Expression Regulation ,Histone methyltransferase ,Epigenetic Repression ,COS Cells ,Protein Binding ,Signal Transduction ,030215 immunology - Abstract
During B lymphopoiesis, recombination of the locus encoding the immunoglobulin κ-chain complex (Igk) requires expression of the precursor to the B cell antigen receptor (pre-BCR) and escape from signaling via the interleukin 7 receptor (IL-7R). By activating the transcription factor STAT5, IL-7R signaling maintains proliferation and represses Igk germline transcription by unknown mechanisms. We demonstrate that a STAT5 tetramer bound the Igk intronic enhancer (E(κi)), which led to recruitment of the histone methyltransferase Ezh2. Ezh2 marked trimethylation of histone H3 at Lys27 (H3K27me3) throughout the κ-chain joining region (J(κ)) to the κ-chain constant region (C(κ)). In the absence of Ezh2, IL-7 failed to repress Igk germline transcription. H3K27me3 modifications were lost after termination of IL-7R-STAT5 signaling, and the transcription factor E2A bound E(κi), which resulted in acquisition of H3K4me1 and acetylated histone H4 (H4Ac). Genome-wide analyses showed a STAT5 tetrameric binding motif associated with transcriptional repression. Our data demonstrate how IL-7R signaling represses Igk germline transcription and provide a general model for STAT5-mediated epigenetic transcriptional repression.
- Published
- 2011
34. Early B-cell factor regulates the expression of Hemokinin-1 in the olfactory epithelium and differentiating B lymphocytes
- Author
-
Alexandra Berger, Anne H. Tran, Christopher J. Paige, Gillian E. Wu, and Barbara L. Kee
- Subjects
Nervous system ,Molecular Sequence Data ,Immunology ,Regulator ,Gene Expression ,Cell Separation ,Biology ,Transfection ,Hemokinin-1 ,Mice ,Immune system ,Olfactory Mucosa ,Tachykinins ,medicine ,Animals ,Immunology and Allergy ,Protein Precursors ,Promoter Regions, Genetic ,Gene ,Transcription factor ,B-Lymphocytes ,Base Sequence ,Reverse Transcriptase Polymerase Chain Reaction ,Cell Differentiation ,Blotting, Northern ,Flow Cytometry ,Cell biology ,Mice, Inbred C57BL ,medicine.anatomical_structure ,Bridge (graph theory) ,Gene Expression Regulation ,Neurology ,Mutagenesis, Site-Directed ,Trans-Activators ,Female ,Neurology (clinical) ,Olfactory epithelium - Abstract
Hemokinin-1, encoded by the TAC4 gene, is a tachykinin most closely related to substance P. Previous studies have shown that TAC4 distinguishes itself from other tachykinins by its predominantly non-neuronal expression profile, particularly in cells of the immune system. Here we report for the first time that the highest levels of TAC4 expression are found in the olfactory epithelium. Furthermore, we identify olfactory neuron-specific transcription factor (Olf-1), also known as early B-cell factor (EBF), as a novel regulator of TAC4 expression. EBF present in the olfactory epithelium and in B cells binds to two sites in the TAC4 promoter and modulates expression in developing B cells. Our findings suggest a role for TAC4 in cell differentiation, and represent a regulatory bridge between the nervous system and the immune system.
- Published
- 2011
- Full Text
- View/download PDF
35. Alternative Promoter Usage at the Notch1 Locus Supports Ligand-Independent Signaling in T Cell Development and Leukemogenesis
- Author
-
Juan Miguel Redondo, Marc Vooijs, Freddy Radtke, Jiangwen Zhang, Feifei Liu, Audrey F. Jackson, Taku Naito, Pablo Gómez-del Arco, Barbara L. Kee, Mariko Kashiwagi, Katia Georgopoulos, MUMC+: MA Radiotherapie OC (9), Radiotherapie, and RS: GROW - School for Oncology and Reproduction
- Subjects
Epigenomics ,T cell ,T-Lymphocytes ,Immunology ,Notch signaling pathway ,Biology ,Lymphocyte Activation ,Precursor T-Cell Lymphoblastic Leukemia-Lymphoma ,Article ,03 medical and health sciences ,Ikaros Transcription Factor ,Mice ,0302 clinical medicine ,hemic and lymphatic diseases ,medicine ,Animals ,Immunology and Allergy ,Receptor, Notch1 ,Promoter Regions, Genetic ,Transcription factor ,030304 developmental biology ,Regulation of gene expression ,0303 health sciences ,Gene Expression Regulation, Leukemic ,Promoter ,Chromatin ,medicine.anatomical_structure ,Infectious Diseases ,Genetic Loci ,030220 oncology & carcinogenesis ,embryonic structures ,Cancer research ,cardiovascular system ,sense organs ,Signal transduction ,biological phenomena, cell phenomena, and immunity ,Signal Transduction - Abstract
Loss of Ikaros has been correlated with Notch activation in T cell acute lymphoblastic leukemia (T-ALL), however, the mechanism remains unknown. We identified promoters in Notch1 that drive expression of Notch1 proteins active in the absence of ligand. Ikaros bound to both canonical and alternative Notch1 promoters and its loss increased permissive chromatin, facilitating recruitment of transcription regulators. At early stages of leukemogenesis, increased basal expression from the canonical and 5’-alternative promoters initiated a feed-back loop, progressively augmenting Notch1 signaling. Ikaros also repressed intragenic promoters that are cryptic in wild-type, poised in pre-leukemic, and active in leukemic cells and which also produced ligand-independent Notch1 proteins. Only ligand-independent Notch1 isoforms were required for Ikaros-mediated leukemogenesis. Notch1 alternative-promoter usage was observed at stages of T cell development dependent on Notch signaling and during T-ALL progression. These studies identify a network of epigenetic and transcriptional regulators that control conventional and unconventional Notch signaling during normal development and leukemogenesis.
- Published
- 2010
36. E proteins and the regulation of early lymphocyte development
- Author
-
Renée F. de Pooter and Barbara L. Kee
- Subjects
Genetics ,Regulation of gene expression ,Myeloid ,Cellular differentiation ,Immunology ,Context (language use) ,Biology ,Cell biology ,Haematopoiesis ,medicine.anatomical_structure ,Multipotent Stem Cell ,hemic and lymphatic diseases ,medicine ,Immunology and Allergy ,Lymphoid Progenitor Cells ,Lymphopoiesis - Abstract
Lymphopoiesis generates mature B, T, and NK lymphocytes from hematopoietic stem cells via a series of increasingly restricted developmental intermediates. The transcriptional networks that regulate these fate choices are composed of both common and lineage-specific components, which combine to create a cellular context that informs the developmental response to external signals. E proteins are an important factor during lymphopoiesis, and E2A in particular is required for normal T- and B-cell development. Although the other E proteins, HEB and E2-2, are expressed during lymphopoiesis and can compensate for some of E2A's activity, E2A proteins have non-redundant functions during early T-cell development and at multiple checkpoints throughout B lymphopoiesis. More recently, a role for E2A has been demonstrated in the generation of lymphoid-primed multipotent progenitors and shown to favor their specification toward lymphoid over myeloid lineages. This review summarizes both our current understanding of the wide-ranging functions of E proteins during the development of adaptive lymphocytes and the novel functions of E2A in orchestrating a lymphoid-biased cellular context in early multipotent progenitors.
- Published
- 2010
- Full Text
- View/download PDF
37. Pioneer transcription factor BATF controls chromatin accessibility and CTCF-mediated chromatin architecture in CD4+ T cells
- Author
-
Duy Pham, Carson E. Moseley, Daniel Savic, Richard Myers, Barbara L. Kee, Robin D. Hatton, and Casey T. Weaver
- Subjects
Immunology ,Immunology and Allergy - Abstract
Pioneer transcription factors are a unique class of proteins that access heterochromatin, facilitate chromatin accessibility, and recruit other transcription factors to regulate gene transcription. The basic leucine zipper transcription factor ATF-like (BATF) is a critical factor for the development of T helper cell lineages. While BATF has been proposed as a pioneer factor in both T and B cells, its molecular functions are incompletely understood. In this study, we have identified a novel aspect of the pioneer function of BATF in controlling the chromatin landscape in developing effector T cells. We find that BATF is indispensable for the recruitment of chromatin-organizing complexes containing the DNA binding factor CTCF and other co-factors such as Oct1 and Ets1 to a subset of enhancer and chromatin-looping nodes during effector T cell lineage specification. Accordingly, BATF has a unique function in modulating genomic spatial organization in a CTCF-dependent manner. This study establishes BATF as a bona fide pioneer factor and provides novel insight into its role as a spatial genome organizer that modulates chromatin accessibility and initiates chromatin looping for the coordinate and differential gene expression in CD4+ T cells.
- Published
- 2018
- Full Text
- View/download PDF
38. Transcriptional regulation of natural killer cell development
- Author
-
Barbara L. Kee and Kevin Ramirez
- Subjects
Lymphokine-activated killer cell ,medicine.medical_treatment ,Immunology ,Biology ,Natural killer T cell ,Cell biology ,Interleukin 21 ,Cytokine ,Interleukin 15 ,medicine ,Interleukin 12 ,Immunology and Allergy ,Lymphopoiesis ,Transcription factor - Abstract
Natural killer (NK) cells are a subset of lymphocytes that kill virus-infected or cancerous cells and influence adaptive immune responses via production of inflammatory cytokines. Unlike B and T lymphocytes, no transcription factors have been identified that are essential for the emergence of NK cell progenitors from their multipotent precursors. We argue that this dearth of essential factors is because of the expression of redundant transcription factors that function at the earliest stages of development. However, multiple essential transcription factors have been identified at later stages of development. Recent studies have revealed novel subsets of NK cells with differing potential for target cell lysis and cytokine production. How these subsets arise from the conventional pathway of NK cell development and identification of the transcriptional networks that control their development are major challenges for future studies.
- Published
- 2010
- Full Text
- View/download PDF
39. Ras orchestrates exit from the cell cycle and light-chain recombination during early B cell development
- Author
-
Malay Mandal, Katia Georgopoulos, Harinder Singh, Sarah E. Powers, Marcus R. Clark, Kyoko Ochiai, and Barbara L. Kee
- Subjects
education.field_of_study ,Cellular differentiation ,Immunology ,Population ,Biology ,Cell cycle ,medicine.anatomical_structure ,Cancer research ,medicine ,Immunology and Allergy ,Signal transduction ,education ,Enhancer ,Interleukin-7 receptor ,Transcription factor ,B cell - Abstract
How signals through the pre–B cell antigen receptor (pre-BCR) and IL-7 receptor (IL-7R) coordinate population expansion of pre-B cells with subsequent recombination of the immunoglobulin κ-chain locus is unclear. Clark and colleagues show that pre-BCR signaling via the Ras-MEK-Erk pathway poises pre–B cells to undergo differentiation after escaping IL-7R signaling. Signals through the pre–B cell antigen receptor (pre-BCR) and interleukin 7 receptor (IL-7R) coordinate pre–B cell population expansion with subsequent recombination of the locus encoding immunoglobulin κ-chain (Igk). Although many 'downstream' effectors of each receptor are known, how they integrate to mediate development has remained unclear. Here we report that pre-BCR-mediated activation of the Ras-MEK-Erk signaling pathway silenced transcription of Ccnd3 (encoding cyclin D3) and coordinated exit from the cell cycle with induction of the transcription factor E2A and the initiation of Igk recombination. IL-7R-mediated activation of the transcription factor STAT5 opposed this pathway by promoting Ccnd3 expression and concomitantly inhibiting Igk transcription by binding to the Igk intronic enhancer and preventing E2A recruitment. Our data show how pre-BCR signaling poises pre–B cells to undergo differentiation after escape from IL-7R signaling.
- Published
- 2009
- Full Text
- View/download PDF
40. E and ID proteins branch out
- Author
-
Barbara L. Kee
- Subjects
History ,T-Lymphocytes ,Cell lineage ,Biology ,Education ,chemistry.chemical_compound ,Basic Helix-Loop-Helix Transcription Factors ,Animals ,Humans ,Cell Lineage ,Lymphopoiesis ,B-Lymphocytes ,Extramural ,Transcriptional Networks ,Dendritic Cells ,Computer Science Applications ,Cell biology ,DNA-Binding Proteins ,Killer Cells, Natural ,Haematopoiesis ,chemistry ,Inhibitor of Differentiation Proteins ,Stem cell ,DNA ,Function (biology) - Abstract
E and inhibitor of DNA binding (ID) proteins are transcriptional regulators that function in many developmental processes in vertebrates and invertebrates. One subset of E proteins, the E2A proteins, have a central role in the transcriptional regulatory networks that promote commitment to and differentiation of the B- and T-cell lineages, and their function in these lineages is modulated by ID proteins. In this Review, I discuss recent studies that reveal a more extensive role for E and ID proteins in the transcriptional networks that drive the differentiation of many lymphoid lineages, as well as new functions for these proteins in haematopoietic stem cells and their multipotent, but lymphoid-primed, progeny.
- Published
- 2009
- Full Text
- View/download PDF
41. Gut microbiota regulates K/BxN autoimmune arthritis through Tfh but not Th17 cells
- Author
-
Alexander L. Dent, Haochu Huang, Zhong Zheng, Barbara L. Kee, and Katharine E. Block
- Subjects
0301 basic medicine ,T cell ,Segmented filamentous bacteria ,Cellular differentiation ,Immunology ,Arthritis ,Gut flora ,digestive system ,Article ,Arthritis, Rheumatoid ,03 medical and health sciences ,0302 clinical medicine ,Immune system ,Mice, Inbred NOD ,medicine ,Immunology and Allergy ,Animals ,Mucous Membrane ,biology ,Cell Differentiation ,T-Lymphocytes, Helper-Inducer ,biology.organism_classification ,BCL6 ,medicine.disease ,Germinal Center ,Phenotype ,Arthritis, Experimental ,Gastrointestinal Microbiome ,DNA-Binding Proteins ,Mice, Inbred C57BL ,Disease Models, Animal ,030104 developmental biology ,medicine.anatomical_structure ,Proto-Oncogene Proteins c-bcl-6 ,Th17 Cells ,030215 immunology - Abstract
The bacterial community that colonizes mucosal surfaces helps shape the development and function of the immune system. The K/BxN autoimmune arthritis model is dependent on the microbiota, and particularly on segmented filamentous bacteria, for the autoimmune phenotype. The mechanisms of how the gut microbiota affects arthritis development are not well understood. In this study, we investigate the contribution of two T cell subsets, Th17 and follicular helper T (Tfh), to arthritis and how microbiota modulates their differentiation. Using genetic approaches, we demonstrate that IL-17 is dispensable for arthritis. Antibiotic treatment inhibits disease in IL-17–deficient animals, suggesting that the gut microbiota regulates arthritis independent of Th17 cells. In contrast, conditional deletion of Bcl6 in T cells blocks Tfh cell differentiation and arthritis development. Furthermore, Tfh cell differentiation is defective in antibiotic-treated mice. Taken together, we conclude that gut microbiota regulates arthritis through Tfh but not Th17 cells. These findings have implications in our understanding of how environmental factors contribute to the development of autoimmune diseases.
- Published
- 2016
42. Development of Natural Killer Cells and ILC1
- Author
-
Barbara L. Kee
- Subjects
Interleukin 21 ,Lymphokine-activated killer cell ,medicine.anatomical_structure ,Interleukin 15 ,Immunology ,Innate lymphoid cell ,medicine ,Interleukin 12 ,Lymphopoiesis ,Biology ,Natural killer T cell ,Natural killer cell ,Cell biology - Abstract
Group 1 innate lymphoid cells (ILCs) include natural killer (NK) cells and ILC1. NK cells function in response to intracellular pathogens such as viruses and they directly kill infected cells as well as produce cytokines that activate innate and adaptive components of the immune response. While the critical functions of ILC1 are still under investigation, these cells share many features with NK cells including expression of a common core of transcription factors and a requirement for the T-box binding factors for their development. In this article I will discuss the essential features of NK cell and ILC1 development, and I will discuss the transcription factors that are known to be critical for their development from their common ancestor, the common lymphoid progenitor.
- Published
- 2016
- Full Text
- View/download PDF
43. The ETS1 transcription factor is required for the development and cytokine-induced expansion of ILC2
- Author
-
Erin C. Zook, Kevin Ramirez, Eric C. Svensson, Grant van der Voort, Xiaohuan Guo, Barbara L. Kee, Mikael Sigvardsson, and Yang Xin Fu
- Subjects
0301 basic medicine ,medicine.medical_treatment ,Cell- och molekylärbiologi ,Immunology ,Inflammation ,Biology ,Proto-Oncogene Protein c-ets-1 ,Mice ,03 medical and health sciences ,0302 clinical medicine ,ETS1 ,medicine ,Animals ,Immunology and Allergy ,Lymphocytes ,Progenitor cell ,Transcription factor ,Research Articles ,Inhibitor of Differentiation Protein 2 ,Mice, Knockout ,Innate lymphoid cell ,Brief Definitive Report ,Immunity, Innate ,Cell biology ,Interleukin 33 ,030104 developmental biology ,Cytokine ,Interleukin 13 ,Cytokines ,medicine.symptom ,Cell and Molecular Biology ,030215 immunology - Abstract
Zook et al. use a novel mouse model to demonstrate a requirement for the transcription factor ETS1 in the development and function of group 2 innate lymphoid cells., Group 2 innate lymphoid cells (ILC2s) are a subset of ILCs that play a protective role in the response to helminth infection, but they also contribute to allergic lung inflammation. Here, we report that the deletion of the ETS1 transcription factor in lymphoid cells resulted in a loss of ILC2s in the bone marrow and lymph nodes and that ETS1 promotes the fitness of the common progenitor of all ILCs. ETS1-deficient ILC2 progenitors failed to up-regulate messenger RNA for the E protein transcription factor inhibitor ID2, a critical factor for ILCs, and these cells were unable to expand in cytokine-driven in vitro cultures. In vivo, ETS1 was required for the IL-33–induced accumulation of lung ILC2s and for the production of the T helper type 2 cytokines IL-5 and IL-13. IL-25 also failed to elicit an expansion of inflammatory ILC2s when these cells lacked ETS1. Our data reveal ETS1 as a critical regulator of ILC2 expansion and cytokine production and implicate ETS1 in the regulation of Id2 at the inception of ILC2 development.
- Published
- 2016
44. Extrinsic and intrinsic regulation of early natural killer cell development
- Author
-
Kevin Ramirez, Markus D. Boos, and Barbara L. Kee
- Subjects
Immunology ,Bone Marrow Cells ,Thymus Gland ,Biology ,Natural killer cell ,Interleukin 21 ,Bone Marrow ,medicine ,Animals ,Humans ,Cell Lineage ,Receptors, Cytokine ,Innate immune system ,Lymphokine-activated killer cell ,Helix-Loop-Helix Motifs ,Innate lymphoid cell ,Natural killer T cell ,Cell biology ,Killer Cells, Natural ,medicine.anatomical_structure ,Myeloid-derived Suppressor Cell ,Interleukin 12 ,Cytokines ,Lymph Nodes ,Transcription Factors - Abstract
Natural killer (NK) cells are lymphocytes that play a critical role in both adaptive and innate immune responses. These cells develop from multipotent progenitors in the embryonic thymus and neonatal or adult bone marrow and recent evidence suggests that a subset of these cells may develop in the thymus. Thymus- and bone marrow-derived NK cells have unique phenotypes and functional abilities supporting the hypothesis that the microenvironment dictates the outcome of NK cell development. A detailed understanding of the mechanisms controlling this developmental program will be required to determine how alterations in NK cell development lead to disease and to determine how to harness this developmental program for therapeutic purposes. In this review, we discuss some of the known extrinsic stromal-cell derived factors and cell intrinsic transcription factors that function in guiding NK cell development.
- Published
- 2007
- Full Text
- View/download PDF
45. Interferon-producing killer dendritic cells (IKDCs) arise via a unique differentiation pathway from primitive c-kitHiCD62L+ lymphoid progenitors
- Author
-
Rosana Pelayo, Karla P. Garrett, Paul W. Kincade, Xiao Hong Sun, Xinrong Chen, S. Scott Perry, Barbara L. Kee, and Robert S. Welner
- Subjects
CpG Oligodeoxynucleotide ,Cellular differentiation ,Immunology ,Bone Marrow Cells ,Biology ,Biochemistry ,Interferon-gamma ,Mice ,Interferon ,medicine ,Animals ,Interferon gamma ,Lymphocytes ,L-Selectin ,Progenitor cell ,Immunobiology ,Mice, Inbred BALB C ,Stem Cells ,TLR9 ,Cell Differentiation ,hemic and immune systems ,Dendritic Cells ,Cell Biology ,Hematology ,Cell biology ,Killer Cells, Natural ,Mice, Inbred C57BL ,Transplantation ,Proto-Oncogene Proteins c-kit ,Toll-Like Receptor 9 ,Interferons ,Stem cell ,medicine.drug - Abstract
Interferon-producing killer dendritic cells (IKDCs) have only recently been described and they share some properties with plasmacytoid dendritic cells (pDCs). We now show that they can arise from some of the same progenitors. However, IKDCs expressed little or no RAG-1, Spi-B, or TLR9, but responded to the TLR9 agonist CpG ODN by production of IFNγ. The RAG-1−pDC2 subset was more similar to IKDCs than RAG-1+ pDC1s with respect to IFNγ production. The Id-2 transcriptional inhibitor was essential for production of IKDCs and natural killer (NK) cells, but not pDCs. IKDCs developed from lymphoid progenitors in culture but, unlike pDCs, were not affected by Notch receptor ligation. While IKDCs could be made from estrogen-sensitive progenitors, they may have a slow turnover because their numbers did not rapidly decline in hormone-treated mice. Four categories of progenitors were compared for IKDC-producing ability in transplantation assays. Of these, Lin−Sca-1+c-KitHiThy1.1−L-selectin+ lymphoid progenitors (LSPs) were the best source. While NK cells resemble IKDCs in several respects, they develop from different progenitors. These observations suggest that IKDCs may arise from a unique differentiation pathway, and one that diverges early from those responsible for NK cells, pDCs, and T and B cells.
- Published
- 2007
- Full Text
- View/download PDF
46. Growth factor independent 1B (Gfi1b) is an E2A target gene that modulates Gata3 in T-cell lymphomas
- Author
-
Barbara L. Kee and Wei Xu
- Subjects
Cell Survival ,medicine.medical_treatment ,T cell ,Immunology ,chemical and pharmacologic phenomena ,GATA3 Transcription Factor ,Thymus Gland ,Biology ,Lymphoma, T-Cell ,Biochemistry ,Proto-Oncogene Proteins ,hemic and lymphatic diseases ,Basic Helix-Loop-Helix Transcription Factors ,Tumor Cells, Cultured ,medicine ,Humans ,RNA, Messenger ,Lymphopoiesis ,Progenitor cell ,Transcription factor ,Cell Proliferation ,Regulation of gene expression ,Gene Expression Profiling ,Growth factor ,GATA3 ,hemic and immune systems ,Cell Biology ,Hematology ,Gene Expression Regulation, Neoplastic ,Repressor Proteins ,medicine.anatomical_structure ,Cancer research ,Ectopic expression ,tissues - Abstract
The E2A transcription factors are required for normal T lymphopoiesis and to prevent T-lymphocyte progenitor transformation. Ectopic expression of E2A proteins in E2A-deficient lymphomas results in growth arrest and apoptosis, indicating that these cells remain responsive to the targets of E2A. Here we identify the transcriptional repressor growth factor independent 1B (Gfi1b) as a target of E2A that promotes growth arrest and apoptosis in lymphomas. Gfi1b expression in primary T-lymphocyte progenitors is dependent on E2A and excess Gfi1b prevents the outgrowth of T lymphocyte progenitors in vitro. Gfi1b represses expression of Gata3, a transcription factor whose appropriate regulation is required for survival of lymphomas and T-lymphocyte progenitors. We also show that ectopic expression of Gata3 in lymphomas promotes expression of Gfi1b, indicating that these proteins may function in an autoregulatory loop that maintains appropriate levels of Gata3. Therefore, we propose that E2A proteins prevent lymphoma cell expansion, at least in part through regulation of Gfi1b and modulation of Gata3 expression.
- Published
- 2007
- Full Text
- View/download PDF
47. NFIL3 Orchestrates the Emergence of Common Helper Innate Lymphoid Cell Precursors
- Author
-
Lara Moreira-Santos, Yasutaka Motomura, Franck Bihl, Manuela Ferreira, Hélder Ribeiro, Veronique M. Braud, Henrique Veiga-Fernandes, Mark Coles, Silvia Lopez-Lastra, Christian A. J. Vosshenrich, Hugh J.M. Brady, Barbara L. Kee, Masato Kubo, James P. Di Santo, Wei Xu, Rita G. Domingues, Diogo Fonseca-Pereira, Immunité Innée - Innate Immunity, Institut Pasteur [Paris] (IP)-Institut National de la Santé et de la Recherche Médicale (INSERM), Instituto de Medicina Molecular (iMM), Faculdade de Medicina [Lisboa], Universidade de Lisboa = University of Lisbon (ULISBOA)-Universidade de Lisboa = University of Lisbon (ULISBOA), RIKEN - Institute of Physical and Chemical Research [Japon] (RIKEN), Institut de pharmacologie moléculaire et cellulaire (IPMC), Université Nice Sophia Antipolis (1965 - 2019) (UNS), COMUE Université Côte d'Azur (2015-2019) (COMUE UCA)-COMUE Université Côte d'Azur (2015-2019) (COMUE UCA)-Centre National de la Recherche Scientifique (CNRS)-Université Côte d'Azur (UCA), University of Chicago, Imperial College London, University of York [York, UK], R.G.D., D.F.-P., andM.F.weresupported by Fundaçaoo para a Ciencia e Tecnologia, Portugal, H.V.-F. by Fundaçao para aCiencia e Tecnologia, Portugal, EMBO (1648) and ERC (207057), and J.P.D.by grants from the Institut Pasteur, Inserm, LNCC (Equipe Labellisée LigueContre le Cancer) and the Agence National pour la Recherche (Program‘‘Blanc’’ Gut_ILC). J.P.D. is a founder and stakeholder in the biotechnologycompany AXENIS (Paris, France), ANR-10-BLAN-1305,Gut_ILC,Les cellules lymphoïdes innées dans l'homéostasie et la défense immune intestinale(2010), European Project: 207057,EC:FP7:ERC,ERC-2007-StG,RETIMMUNEFUNCTION(2008), Repositório da Universidade de Lisboa, Institut Pasteur [Paris]-Institut National de la Santé et de la Recherche Médicale (INSERM), Universidade de Lisboa (ULISBOA)-Universidade de Lisboa (ULISBOA), Centre National de la Recherche Scientifique (CNRS)-Université Nice Sophia Antipolis (... - 2019) (UNS), COMUE Université Côte d'Azur (2015-2019) (COMUE UCA)-COMUE Université Côte d'Azur (2015-2019) (COMUE UCA)-Université Côte d'Azur (UCA), Di Santo, James, BLANC - Les cellules lymphoïdes innées dans l'homéostasie et la défense immune intestinale - - Gut_ILC2010 - ANR-10-BLAN-1305 - BLANC - VALID, and Role of the proto-oncogene Ret during lymphocyte development and function - RETIMMUNEFUNCTION - - EC:FP7:ERC2008-11-01 - 2013-10-31 - 207057 - VALID
- Subjects
[SDV.IMM] Life Sciences [q-bio]/Immunology ,Regulator ,Biology ,General Biochemistry, Genetics and Molecular Biology ,03 medical and health sciences ,0302 clinical medicine ,Animals ,Cell Lineage ,Lymphocytes ,Lymphopoiesis ,Progenitor cell ,10. No inequality ,skin and connective tissue diseases ,lcsh:QH301-705.5 ,030304 developmental biology ,Progenitor ,0303 health sciences ,Effector ,Innate lymphoid cell ,NFIL3 ,Gene targeting ,Cell Differentiation ,Lymphoid Progenitor Cells ,Immunity, Innate ,Killer Cells, Natural ,Mice, Inbred C57BL ,body regions ,Basic-Leucine Zipper Transcription Factors ,lcsh:Biology (General) ,Cancer research ,[SDV.IMM]Life Sciences [q-bio]/Immunology ,030215 immunology - Abstract
© 2015 The Authors. Published by Elsevier Inc., Innate lymphoid cells (ILCs) are a family of effectors that originate from a common innate lymphoid cell progenitor. However, the transcriptional program that sets the identity of the ILC lineage remains elusive. Here, we show that NFIL3 is a critical regulator of the common helper-like innate lymphoid cell progenitor (CHILP). Cell-intrinsic Nfil3 ablation led to variably impaired development of fetal and adult ILC subsets. Conditional gene targeting demonstrated that NFIL3 exerted its function prior to ILC subset commitment. Accordingly, NFIL3 ablation resulted in loss of ID2(+) CHILP and PLZF(+) ILC progenitors. Nfil3 expression in lymphoid progenitors was under the control of the mesenchyme-derived hematopoietin IL-7, and NFIL3 exerted its function via direct Id2 regulation in the CHILP. Moreover, ectopic Id2 expression in Nfil3-null precursors rescued defective ILC lineage development in vivo. Our data establish NFIL3 as a key regulator of common helper-like ILC progenitors as they emerge during early lymphopoiesis., R.G.D., D.F.-P., and M.F. were supported by Fundação para a Ciência e Tecnologia, Portugal; H.V.-F. by Fundação para a Ciência e Tecnologia, Portugal, EMBO (1648) and ERC (207057); and J.P.D. by grants from the Institut Pasteur, Inserm, LNCC (Equipe Labellisée Ligue Contre le Cancer) and the Agence National pour la Recherche (Program “Blanc” Gut_ILC).
- Published
- 2015
- Full Text
- View/download PDF
48. Targeting the NF-κB signaling pathway in Notch1-induced T-cell leukemia
- Author
-
Malay Mandal, Iannis Aifantis, Teresa Palomero, Adolfo A. Ferrando, Fanyong Meng, Lucio Miele, Barbara L. Kee, Benjamin J. Thompson, Tomas Vilimas, Christina Spaulding, Joaquina Mascarenhas, Silvia Buonamici, Maria-Luisa Alegre, and Sami Macaroun
- Subjects
Leukemia, T-Cell ,Cell Survival ,CD8 Antigens ,Green Fluorescent Proteins ,T-cell leukemia ,IκB kinase ,General Biochemistry, Genetics and Molecular Biology ,Cell Line ,Bortezomib ,Mice ,Cell Line, Tumor ,Chlorocebus aethiops ,Animals ,Humans ,Medicine ,Neoplastic transformation ,Receptor, Notch1 ,Progenitor cell ,Mice, Knockout ,Leukemia, Experimental ,Microscopy, Confocal ,business.industry ,Gene Expression Profiling ,NF-kappa B ,General Medicine ,medicine.disease ,Boronic Acids ,Survival Analysis ,DNA-Binding Proteins ,Mice, Inbred C57BL ,Haematopoiesis ,Leukemia ,Pyrazines ,CD4 Antigens ,COS Cells ,Mutation ,Immunology ,Cancer research ,Stem cell ,Signal transduction ,business ,Interleukin Receptor Common gamma Subunit ,Signal Transduction - Abstract
T-cell acute lymphoblastic leukemia (T-ALL), unlike other ALL types, is only infrequently associated with chromosomal aberrations, but it was recently shown that most individuals with T-ALL carry activating mutations in the NOTCH1 gene. However, the signaling pathways and target genes responsible for Notch1-induced neoplastic transformation remain undefined. We report here that constitutively active Notch1 activates the NF-kappaB pathway transcriptionally and via the IkappaB kinase (IKK) complex, thereby causing increased expression of several well characterized target genes of NF-kappaB in bone marrow hematopoietic stem cells and progenitors. Our observations demonstrate that the NF-kappaB pathway is highly active in established human T-ALL and that inhibition of the pathway can efficiently restrict tumor growth both in vitro and in vivo. These findings identify NF-kappaB as one of the major mediators of Notch1-induced transformation and suggest that the NF-kappaB pathway is a potential target of future therapies of T-ALL.
- Published
- 2006
- Full Text
- View/download PDF
49. A Gradient of Template Dependence Defines Distinct Biological Roles for Family X Polymerases in Nonhomologous End Joining
- Author
-
Jody M. Havener, Stephanie A. Nick McElhinny, Dale A. Ramsden, Katarzyna Bebenek, Thomas A. Kunkel, Miguel Garcia-Diaz, Raquel Juárez, Barbara L. Kee, and Luis Blanco
- Subjects
Models, Molecular ,DNA Repair ,viruses ,Molecular Sequence Data ,Gene Expression ,Context (language use) ,DNA-Directed DNA Polymerase ,Transfection ,Cell Line ,Substrate Specificity ,03 medical and health sciences ,chemistry.chemical_compound ,Immunoglobulin kappa-Chains ,Mice ,0302 clinical medicine ,DNA Nucleotidylexotransferase ,Animals ,Humans ,Amino Acid Sequence ,Molecular Biology ,Polymerase ,DNA Polymerase beta ,030304 developmental biology ,Genetics ,Gene Rearrangement ,Recombination, Genetic ,0303 health sciences ,B-Lymphocytes ,biology ,Sequence Homology, Amino Acid ,fungi ,DNA ,Templates, Genetic ,Cell Biology ,DNA polymerase lambda ,Recombinant Proteins ,Cell biology ,Non-homologous end joining ,enzymes and coenzymes (carbohydrates) ,chemistry ,030220 oncology & carcinogenesis ,Coding strand ,embryonic structures ,biology.protein ,Immunoglobulin Joining Region ,Primer (molecular biology) ,Recombination - Abstract
Three Pol X family members have been linked to nonhomologous end joining (NHEJ) in mammals. Template-independent TdT promotes diversity during NHEJ-dependent repair of V(D)J recombination intermediates, but the roles of the template-dependent polymerases mu and lambda in NHEJ remain unclear. We show here that pol mu and pol lambda are similarly recruited by NHEJ factors to fill gaps when ends have partially complementary overhangs, suggesting equivalent roles promoting accuracy in NHEJ. However, only pol mu promotes accuracy during immunoglobulin kappa recombination. This distinctive in vivo role correlates with the TdT-like ability of pol mu, but not pol lambda, to act when primer termini lack complementary bases in the template strand. However, unlike TdT, synthesis by pol mu in this context is primarily instructed by a template from another DNA molecule. This apparent gradient of template dependence is largely attributable to a small structural element that is present but different in all three polymerases.
- Published
- 2005
- Full Text
- View/download PDF
50. Lymphoid development: it's not 'all Greek to us' any more
- Author
-
Barbara L. Kee and Barbara A. Osborne
- Subjects
Regulation of gene expression ,Genetics ,Lineage (genetic) ,Immunology ,Immunology and Allergy ,Identity (social science) ,Biology ,Lineage specification - Abstract
Lymphocyte lineage specification involves multiple regulatory factors that act in reciprocal fashion to ensure lineage committment and identity. New insights on how these factors interact were presented at the second Aegean Workshop on Gene Regulation in Lymphocyte Development.
- Published
- 2005
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.