61 results on '"Hai-Hui Xue"'
Search Results
2. The Interplay of Transcription and Genome Topology Programs T Cell Development and Differentiation
- Author
-
Xin Zhao, Shaoqi Zhu, Weiqun Peng, and Hai-Hui Xue
- Subjects
CCCTC-Binding Factor ,Genome ,Immunology ,Immunology and Allergy ,Cell Cycle Proteins ,Cell Differentiation ,Chromatin ,Chromosomes - Abstract
T cells are essential for mounting defense against various pathogens and malignantly transformed cells. Thymic development and peripheral T cell differentiation are highly orchestrated biological processes that require precise gene regulation. Higher-order genome organization on multiple scales, in the form of chromatin loops, topologically associating domains and compartments, provides pivotal control of T cell gene expression. CTCF and the cohesin machinery are ubiquitously expressed architectural proteins responsible for establishing chromatin structures. Recent studies indicate that transcription factors, such as T lineage–defining Tcf1 and TCR-induced Batf, may have intrinsic ability and/or engage CTCF to shape chromatin architecture. In this article, we summarize current knowledge on the dynamic changes in genome topology that underlie normal or leukemic T cell development, CD4+ helper T cell differentiation, and CD8+ cytotoxic T cell functions. The knowledge lays a solid foundation for elucidating the causative link of spatial chromatin configuration to transcriptional and functional output in T cells.
- Published
- 2022
- Full Text
- View/download PDF
3. Tcf1–CTCF cooperativity shapes genomic architecture to promote CD8+ T cell homeostasis
- Author
-
Qiang Shan, Shaoqi Zhu, Xia Chen, Jia Liu, Shuang Yuan, Xiang Li, Weiqun Peng, and Hai-Hui Xue
- Subjects
Immunology ,Immunology and Allergy - Published
- 2022
- Full Text
- View/download PDF
4. CTCF mediates CD8+ effector differentiation through dynamic redistribution and genomic reorganization
- Author
-
Jia Liu, Shaoqi Zhu, Wei Hu, Xin Zhao, Qiang Shan, Weiqun Peng, and Hai-Hui Xue
- Subjects
Immunology ,Immunology and Allergy - Abstract
Differentiation of effector CD8+ T cells is instructed by stably and dynamically expressed transcription regulators. Here we show that naive-to-effector differentiation was accompanied by dynamic CTCF redistribution and extensive chromatin architectural changes. Upon CD8+ T cell activation, CTCF acquired de novo binding sites and anchored novel chromatin interactions, and these changes were associated with increased chromatin accessibility and elevated expression of cytotoxic program genes including Tbx21, Ifng, and Klrg1. CTCF was also evicted from its ex-binding sites in naive state, with concomitantly reduced chromatin interactions in effector cells, as observed at memory precursor–associated genes including Il7r, Sell, and Tcf7. Genetic ablation of CTCF indeed diminished cytotoxic gene expression, but paradoxically elevated expression of memory precursor genes. Comparative Hi-C analysis revealed that key memory precursor genes were harbored within insulated neighborhoods demarcated by constitutive CTCF binding, and their induction was likely due to disrupted CTCF-dependent insulation. CTCF thus promotes cytotoxic effector differentiation by integrating local chromatin accessibility control and higher-order genomic reorganization.
- Published
- 2023
- Full Text
- View/download PDF
5. Exploring the stage-specific roles of Tcf-1 in T cell development and malignancy at single-cell resolution
- Author
-
Zhihong Qi, Yaofeng Zhao, Hai-Hui Xue, Yingpeng Yao, Li Bao, Tianyan Zhao, Peng Jiang, Tao Feng, Fang Wang, Shuyang Yu, and G. Yu
- Subjects
0301 basic medicine ,T cell ,BCL11B ,Immunology ,Cell ,T-Cell Transformation ,Biology ,Lymphocyte Activation ,Lymphoma, T-Cell ,medicine.disease_cause ,T-Lymphocytes, Regulatory ,Article ,Mice ,03 medical and health sciences ,0302 clinical medicine ,FYN ,Biomarkers, Tumor ,medicine ,Animals ,Immunology and Allergy ,Hepatocyte Nuclear Factor 1-alpha ,Mice, Knockout ,Gene Expression Profiling ,Cell Differentiation ,Chromatin ,Cell biology ,Mice, Inbred C57BL ,Thymocyte ,Cell Transformation, Neoplastic ,030104 developmental biology ,Infectious Diseases ,medicine.anatomical_structure ,Lymphocyte Specific Protein Tyrosine Kinase p56(lck) ,Single-Cell Analysis ,Carcinogenesis ,030215 immunology - Abstract
Tcf-1 (encoded by Tcf7) not only plays critical roles in promoting T cell development and differentiation but also has been identified as a tumor suppressor involved in preventing T cell malignancy. However, the comprehensive mechanisms of Tcf-1 involved in T cell transformation remain poorly understood. In this study, Tcf7(fl/fl) mice were crossed with Vav-cre, Lck-cre, or Cd4-cre mice to delete Tcf-1 conditionally at the beginning of the HSC, DN2–DN3, or DP stage, respectively. The defective T cell development phenotypes became gradually less severe as the deletion stage became more advanced in distinct mouse models. Interestingly, consistent with Tcf7(−/−) mice, Tcf7(fl/fl)Vav-cre mice developed aggressive T cell lymphoma within 45 weeks, but no tumors were generated in Tcf7(fl/fl)Lck-cre or Tcf7(fl/fl)Cd4-cre mice. Single-cell RNA-seq (ScRNA-seq) indicated that ablation of Tcf-1 at distinct phases can subdivide DN1 cells into three clusters (C1, C2, and C3) and DN2–DN3 cells into three clusters (C4, C5, and C6). Moreover, Tcf-1 deficiency redirects bifurcation among divergent cell fates, and clusters C1 and C4 exhibit high potential for leukemic transformation. Mechanistically, we found that Tcf-1 directly binds and mediates chromatin accessibility for both typical T cell regulators and proto-oncogenes, including Myb, Mycn, Runx1, and Lyl1 in the DN1 phase and Lef1, Id2, Dtx1, Fyn, Bcl11b, and Zfp36l2 in the DN2–DN3 phase. The aberrant expression of these genes due to Tcf-1 deficiency in very early T cells contributes to subsequent tumorigenesis. Thus, we demonstrated that Tcf-1 plays stage-specific roles in regulating early thymocyte development and transformation, providing new insights and evidence for clinical trials on T-ALL leukemia.
- Published
- 2020
- Full Text
- View/download PDF
6. Peripherally induced brain tissue–resident memory CD8+ T cells mediate protection against CNS infection
- Author
-
Isaac J. Jensen, Lecia L. Pewe, Qiang Shan, Hai-Hui Xue, Vladimir P. Badovinac, John T. Harty, and Stina L. Urban
- Subjects
0301 basic medicine ,business.industry ,medicine.medical_treatment ,Immunology ,Central nervous system ,Brain tissue ,Intravital Imaging ,Peripheral ,03 medical and health sciences ,030104 developmental biology ,0302 clinical medicine ,Cytokine ,medicine.anatomical_structure ,medicine ,Immunology and Allergy ,Cytotoxic T cell ,business ,Neuroscience ,Tissue homeostasis ,CD8 ,030215 immunology - Abstract
The central nervous system (CNS) is classically viewed as immune-privileged; however, recent advances highlight interactions between the peripheral immune system and CNS in controlling infections and tissue homeostasis. Tissue-resident memory (TRM) CD8+ T cells in the CNS are generated after brain infections, but it is unknown whether CNS infection is required to generate brain TRM cells. We show that peripheral infections generate antigen-specific CD8+ memory T cells in the brain that adopt a unique TRM signature. Upon depletion of circulating and perivascular memory T cells, this brain signature was enriched and the surveilling properties of brain TRM cells was revealed by intravital imaging. Notably, peripherally induced brain TRM cells showed evidence of rapid activation and enhanced cytokine production and mediated protection after brain infections. These data reveal that peripheral immunizations can generate brain TRM cells and will guide potential use of T cells as therapeutic strategies against CNS infections and neurological diseases.
- Published
- 2020
- Full Text
- View/download PDF
7. Ectopic Tcf1 expression instills a stem-like program in exhausted CD8+ T cells to enhance viral and tumor immunity
- Author
-
Vladimir P. Badovinac, Xia Chen, Chongzhi Zang, Derek B. Danahy, Qiang Shan, Hai-Hui Xue, and Sheng'en Hu
- Subjects
0301 basic medicine ,Receptor expression ,Immunology ,Repressor ,Biology ,Chromatin ,Cell biology ,Transcriptome ,03 medical and health sciences ,030104 developmental biology ,0302 clinical medicine ,Infectious Diseases ,Antigen ,Immunology and Allergy ,Cytotoxic T cell ,Enhancer ,CD8 ,030215 immunology - Abstract
Exhausted CD8+ T (Tex) cells are dysfunctional due to persistent antigen exposure in chronic viral infection and tumor contexts. A stem cell-like Tex (Tex-stem) subset can self-renew and differentiate into terminally exhausted (Tex-term) cells. Here, we show that ectopic Tcf1 expression potently promoted the generation of Tex-stem cells in both a chronic viral infection and preclinical tumor models. Tcf1 overexpression diminished coinhibitory receptor expression and enhanced polycytokine-producing capacity while retaining a heightened responses to checkpoint blockade, leading to enhanced viral and tumor control. Mechanistically, ectopically expressed Tcf1 exploited existing and novel chromatin accessible sites as transcriptional enhancers or repressors and modulated the transcriptome by enforcing pre-existing expression patterns in Tex-stem cells, such as enhanced suppression of Blimp1 and Bim and acquisition of new downstream genes, including Mx1, Tox2, and Runx3. These findings reveal a pronounced impact of ectopic Tcf1 expression on Tex functional restoration and highlight the therapeutic potential of harnessing Tcf1-enforced transcriptional programs.
- Published
- 2020
- Full Text
- View/download PDF
8. Coactivation of NF-κB and Notch signaling is sufficient to induce B-cell transformation and enables B-myeloid conversion
- Author
-
Thomas J. Waldschmidt, Meiling Jin, Hai-Hui Xue, Qianze Dong, Hasem Habelhah, Youzhong Yuan, Wei Wang, Ji Yuan, Chen Zhao, Nicholas Borcherding, Joseph D. Khoury, Iannis Aifantis, Qingchang Li, Brendan F. Boyce, Andrew L. Feldman, Markus Müschen, Lili Wang, Francesco Boccalatte, Shimin Hu, Benjamin W. Darbro, Endi Wang, Adam Bagg, Lin Fu, Yan Xiu, Aaron D. Bossler, Xiaobing Tang, Mariah R. Leidinger, Jose Luis Sardina, Siegfried Janz, and John D. Colgan
- Subjects
Male ,Myeloid ,Immunology ,Notch signaling pathway ,Biology ,Biochemistry ,Mice ,Downregulation and upregulation ,hemic and lymphatic diseases ,medicine ,Animals ,Humans ,Myeloid Cells ,Progenitor cell ,Transcription factor ,B cell ,B-Lymphocytes ,Lymphoid Neoplasia ,Receptors, Notch ,Transdifferentiation ,NF-kappa B ,Myeloid leukemia ,Lymphoma, B-Cell, Marginal Zone ,Cell Biology ,Hematology ,Mice, Inbred C57BL ,Cell Transformation, Neoplastic ,medicine.anatomical_structure ,Cancer research ,Female ,Signal Transduction - Abstract
NF-κB and Notch signaling can be simultaneously activated in a variety of B-cell lymphomas. Patients with B-cell lymphoma occasionally develop clonally related myeloid tumors with poor prognosis. Whether concurrent activation of both pathways is sufficient to induce B-cell transformation and whether the signaling initiates B-myeloid conversion in a pathological context are largely unknown. Here, we provide genetic evidence that concurrent activation of NF-κB and Notch signaling in committed B cells is sufficient to induce B-cell lymphomatous transformation and primes common progenitor cells to convert to myeloid lineage through dedifferentiation, not transdifferentiation. Intriguingly, the converted myeloid cells can further transform, albeit at low frequency, into myeloid leukemia. Mechanistically, coactivation of NF-κB and Notch signaling endows committed B cells with the ability to self renew. Downregulation of BACH2, a lymphoma and myeloid gene suppressor, but not upregulation of CEBPα and/or downregulation of B-cell transcription factors, is an early event in both B-cell transformation and myeloid conversion. Interestingly, a DNA hypomethylating drug not only effectively eliminated the converted myeloid leukemia cells, but also restored the expression of green fluorescent protein, which had been lost in converted myeloid leukemia cells. Collectively, our results suggest that targeting NF-κB and Notch signaling will not only improve lymphoma treatment, but also prevent the lymphoma-to-myeloid tumor conversion. Importantly, DNA hypomethylating drugs might efficiently treat these converted myeloid neoplasms.
- Published
- 2020
- Full Text
- View/download PDF
9. 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
embryonic structures ,Immunology ,Immunology and Allergy - 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
10. Targeting Cbx3/HP1γ Induces LEF-1 and IL-21R to Promote Tumor-Infiltrating CD8 T-Cell Persistence
- Author
-
Phuong T. Le, Ngoc Ha, Ngan K. Tran, Andrew G. Newman, Katharine M. Esselen, John L. Dalrymple, Eva M. Schmelz, Avinash Bhandoola, Hai-Hui Xue, Prim B. Singh, and To-Ha Thai
- Subjects
Chemokine ,Chromosomal Proteins, Non-Histone ,Melanoma, Experimental ,CD8-Positive T-Lymphocytes ,Lymphocyte Activation ,Immunotherapy, Adoptive ,T-Lymphocytes, Regulatory ,Mice ,Neuroblastoma ,LEF-1 ,Tumor Microenvironment ,Cytotoxic T cell ,Immunology and Allergy ,IL-21 receptor ,Original Research ,Mice, Knockout ,Ovarian Neoplasms ,CD8+ T-cell persistence ,biology ,Effector ,Melanoma ,Interleukin-21 Receptor alpha Subunit ,Cell Differentiation ,Tumor Burden ,Gene Expression Regulation, Neoplastic ,ovarian cancer ,Female ,Signal Transduction ,Lymphoid Enhancer-Binding Factor 1 ,Immunology ,Mice, Transgenic ,Cbx3/HP1γ ,Chromatin remodeling ,Lymphocytes, Tumor-Infiltrating ,Cell Line, Tumor ,medicine ,melanoma ,Animals ,Cbx3/HP1 gamma ,RC581-607 ,medicine.disease ,Immune checkpoint ,Coculture Techniques ,Mice, Inbred C57BL ,Chromobox Protein Homolog 5 ,CD8+T-cell persistence ,biology.protein ,Cancer research ,Immunologic diseases. Allergy ,CD8 - Abstract
Immune checkpoint blockade (ICB) relieves CD8+ T-cell exhaustion in most mutated tumors, and TCF-1 is implicated in converting progenitor exhausted cells to functional effector cells. However, identifying mechanisms that can prevent functional senescence and potentiate CD8+ T-cell persistence for ICB non-responsive and resistant tumors remains elusive. We demonstrate that targeting Cbx3/HP1γ in CD8+ T cells augments transcription initiation and chromatin remodeling leading to increased transcriptional activity at Lef1 and Il21r. LEF-1 and IL-21R are necessary for Cbx3/HP1γ-deficient CD8+ effector T cells to persist and control ovarian cancer, melanoma, and neuroblastoma in preclinical models. The enhanced persistence of Cbx3/HP1γ-deficient CD8+ T cells facilitates remodeling of the tumor chemokine/receptor landscape ensuring their optimal invasion at the expense of CD4+ Tregs. Thus, CD8+ T cells heightened effector function consequent to Cbx3/HP1γ deficiency may be distinct from functional reactivation by ICB, implicating Cbx3/HP1γ as a viable cancer T-cell-based therapy target for ICB resistant, non-responsive solid tumors.
- Published
- 2021
- Full Text
- View/download PDF
11. Sepsis leads to lasting changes in phenotype and function of memory CD8 T cells
- Author
-
Mikaela M. Tremblay, Isaac J. Jensen, Thomas S. Griffith, Weiqun Peng, Micaela G. Fosdick, Qiang Shan, Vladimir P. Badovinac, Xiang Li, Jon C. D. Houtman, Patrick W. McGonagill, and Hai-Hui Xue
- Subjects
Male ,Mouse ,Transcription, Genetic ,Cell ,CD8-Positive T-Lymphocytes ,sepsis ,Mice ,Immunology and Inflammation ,Cytotoxic T cell ,homeostatic proliferation ,Biology (General) ,education.field_of_study ,General Neuroscience ,General Medicine ,Middle Aged ,Phenotype ,Vaccination ,medicine.anatomical_structure ,Medicine ,Female ,medicine.symptom ,Research Article ,central memory ,Human ,Adult ,QH301-705.5 ,Science ,Population ,Inflammation ,Biology ,General Biochemistry, Genetics and Molecular Biology ,Sepsis ,Immune system ,medicine ,Animals ,Humans ,education ,Aged ,Cell Proliferation ,General Immunology and Microbiology ,medicine.disease ,Chromatin Assembly and Disassembly ,Listeria monocytogenes ,CD8 T cell ,Case-Control Studies ,Immunology ,Immunologic Memory - Abstract
The global health burden due to sepsis and the associated cytokine storm is substantial. While early intervention has improved survival during the cytokine storm, those that survive can enter a state of chronic immunoparalysis defined by transient lymphopenia and functional deficits of surviving cells. Memory CD8 T cells provide rapid cytolysis and cytokine production following re-encounter with their cognate antigen to promote long-term immunity, and CD8 T cell impairment due to sepsis can pre-dispose individuals to re-infection. While the acute influence of sepsis on memory CD8 T cells has been characterized, if and to what extent pre-existing memory CD8 T cells recover remains unknown. Here, we observed that central memory CD8 T cells (TCM) from septic patients proliferate more than those from healthy individuals. Utilizing LCMV immune mice and a CLP model to induce sepsis, we demonstrated that TCM proliferation is associated with numerical recovery of pathogen-specific memory CD8 T cells following sepsis-induced lymphopenia. This increased proliferation leads to changes in composition of memory CD8 T cell compartment and altered tissue localization. Further, memory CD8 T cells from sepsis survivors have an altered transcriptional profile and chromatin accessibility indicating long-lasting T cell intrinsic changes. The sepsis-induced changes in the composition of the memory CD8 T cell pool and transcriptional landscape culminated in altered T cell function and reduced capacity to control L. monocytogenes infection. Thus, sepsis leads to long-term alterations in memory CD8 T cell phenotype, protective function and localization potentially changing host capacity to respond to re-infection., eLife digest A dirty cut, a nasty burn, a severe COVID infection; there are many ways for someone to develop sepsis. This life-threatening condition emerges when the immune system overreacts to a threat and ends up damaging the body. Even when patients survive, they are often left with a partially impaired immune system that cannot adequately protect against microbes and cancer; this is known as immunoparalysis. Memory CD8 T cells, a type of immune cell that is compromised by sepsis, are a long-lived population of cells that ‘remember’ previous infection or vaccination, and then react faster to prevent the same illness if the person ever encounters the same threat again. Yet it is unclear how exactly sepsis harms the function and representation of memory CD8 T cells, and the immune system in general. Jensen et al. investigated this question, first by showing that sepsis leads to a profound loss of memory CD8 T cells, but that surviving memory CD8 T cells multiply quickly – especially a subpopulation known as central memory CD8 T cells – to re-establish the memory CD8 T cell population. Since the central memory CD8 T cells proliferate better than the other memory T cells this alters the overall composition of the pool of memory CD8 T cells, with central memory cells becoming overrepresented. Further experiments revealed that this biasing toward central memory T cells, due to sepsis, created long-term changes in the distribution of memory CD8 T cells throughout the body. The way the genetic information of these cells was packaged had also been altered, as well as which genes were switched on or off. Overall, these changes reduced the ability of memory CD8 T cells to control infections. Together, these findings help to understand how immunoparalysis can emerge after sepsis, and what could be done to correct it. These findings could also be applied to other conditions – such as COVID-19 – which may cause similar long-term changes to the immune system.
- Published
- 2021
12. Author response: Sepsis leads to lasting changes in phenotype and function of memory CD8 T cells
- Author
-
Thomas S. Griffith, Weiqun Peng, Micaela G. Fosdick, Jon C. D. Houtman, Vladimir P. Badovinac, Xiang Li, Qiang Shan, Isaac J. Jensen, Mikaela M. Tremblay, Patrick W. McGonagill, and Hai-Hui Xue
- Subjects
Sepsis ,business.industry ,Immunology ,Medicine ,Cytotoxic T cell ,business ,medicine.disease ,Phenotype ,Function (biology) - Published
- 2021
- Full Text
- View/download PDF
13. TCF1 and LEF1 Control Treg Competitive Survival and Tfr Development to Prevent Autoimmune Diseases
- Author
-
Bi-Huei Yang, Kristen Jepsen, Ke Wang, Xiaomei Yuan, Li-Fan Lu, Yan Liang, Shuo Wan, Wanqing Xu, Wenxian Fu, Sunglim Cho, Gen-Sheng Feng, Yi Dong, and Hai-Hui Xue
- Subjects
Male ,0301 basic medicine ,Helper-Inducer ,T-Lymphocytes ,Medical Physiology ,Autoimmunity ,Inbred C57BL ,medicine.disease_cause ,T-Lymphocytes, Regulatory ,Mice ,0302 clinical medicine ,Tfr ,homeostasis ,2.1 Biological and endogenous factors ,Hepatocyte Nuclear Factor 1-alpha ,lcsh:QH301-705.5 ,Mice, Knockout ,Competitive fitness ,autoimmunity ,FOXP3 ,Cell Differentiation ,hemic and immune systems ,T-Lymphocytes, Helper-Inducer ,Regulatory ,competitive fitness ,3. Good health ,regulatory T cells ,medicine.anatomical_structure ,Knockout mouse ,embryonic structures ,Female ,Lymphoid Enhancer-Binding Factor 1 ,Knockout ,chemical and pharmacologic phenomena ,Systemic autoimmunity ,Biology ,Autoimmune Disease ,Article ,General Biochemistry, Genetics and Molecular Biology ,Autoimmune Diseases ,03 medical and health sciences ,Genetics ,medicine ,Animals ,LEF1 ,TCF1 ,B cell ,Prevention ,Inflammatory and immune system ,Germinal center ,Germinal Center ,Mice, Inbred C57BL ,030104 developmental biology ,lcsh:Biology (General) ,Immunology ,Biochemistry and Cell Biology ,030217 neurology & neurosurgery ,Homeostasis - Abstract
SUMMARY CD4+ Foxp3+ T regulatory (Treg) cells are key players in preventing lethal autoimmunity. Tregs undertake differentiation processes and acquire diverse functional properties. However, how Treg’s differentiation and functional specification are regulated remains incompletely understood. Here, we report that gradient expression of TCF1 and LEF1 distinguishes Tregs into three distinct subpopulations, particularly highlighting a subset of activated Treg (aTreg) cells. Treg-specific ablation of TCF1 and LEF1 renders the mice susceptible to systemic autoimmunity. TCF1 and LEF1 are dispensable for Treg’s suppressive capacity but essential for maintaining a normal aTreg pool and promoting Treg’s competitive survival. As a consequence, the development of T follicular regulatory (Tfr) cells, which are a subset of aTreg, is abolished in TCF1/LEF1-conditional knockout mice, leading to unrestrained T follicular helper (Tfh) and germinal center B cell responses. Thus, TCF1 and LEF1 act redundantly to control the maintenance and functional specification of Treg subsets to prevent autoimmunity., Graphical Abstract, In Brief Transcriptional regulation of Treg differentiation and function remains incompletely understood. Yang et al. report that two TCF family transcription factors regulate the survival and functional specification of a subset of Treg cells to prevent autoimmunity.
- Published
- 2019
14. TCF-1 limits the formation of Tc17 cells via repression of the MAF–RORγt axis
- Author
-
Matthew A. Firth, Hai-Hui Xue, Laura K. Mackay, Tracy L Putoczki, Dinesh Raghu, Philip D. Hodgkin, Yang Liao, Qiutong Huang, Simone L Park, Brigette C. Duckworth, Ashley E. Franks, Dale I. Godfrey, Lisa A. Mielke, Melissa J. Davis, Michael Chopin, Soroor Hediyeh-Zadeh, Katherine Kedzierska, Vanessa L. Bryant, Gabrielle T. Belz, Wei Shi, Francisca F. Almeida, Jarny Choi, Ella Bridie Clemens, Carolyn A. Bell, and Hui-Fern Koay
- Subjects
0301 basic medicine ,T cell ,Immunology ,CD8-Positive T-Lymphocytes ,Lymphocyte Activation ,Article ,Mice ,03 medical and health sciences ,0302 clinical medicine ,T-Lymphocyte Subsets ,Interferon ,medicine ,Animals ,Humans ,Immunology and Allergy ,Hepatocyte Nuclear Factor 1-alpha ,Transcription factor ,Research Articles ,Mice, Knockout ,Cell growth ,Chemistry ,Interleukin-17 ,Nuclear Receptor Subfamily 1, Group F, Member 3 ,Flow Cytometry ,Lipid Metabolism ,Chromatin ,Cell biology ,Mice, Inbred C57BL ,030104 developmental biology ,medicine.anatomical_structure ,Proto-Oncogene Proteins c-maf ,030220 oncology & carcinogenesis ,T cell differentiation ,Chromatin Immunoprecipitation Sequencing ,Interleukin 17 ,CD8 ,medicine.drug - Abstract
Mielke et al. show that TCF-1 limits IL-17–producing CD8+ T (Tc17) cell development from double-positive thymocytes through the sequential suppression of MAF and RORγt, while cementing conventional CD8+ T cell fate., Interleukin (IL)-17–producing CD8+ T (Tc17) cells have emerged as key players in host-microbiota interactions, infection, and cancer. The factors that drive their development, in contrast to interferon (IFN)-γ–producing effector CD8+ T cells, are not clear. Here we demonstrate that the transcription factor TCF-1 (Tcf7) regulates CD8+ T cell fate decisions in double-positive (DP) thymocytes through the sequential suppression of MAF and RORγt, in parallel with TCF-1–driven modulation of chromatin state. Ablation of TCF-1 resulted in enhanced Tc17 cell development and exposed a gene set signature to drive tissue repair and lipid metabolism, which was distinct from other CD8+ T cell subsets. IL-17–producing CD8+ T cells isolated from healthy humans were also distinct from CD8+IL-17− T cells and enriched in pathways driven by MAF and RORγt. Overall, our study reveals how TCF-1 exerts central control of T cell differentiation in the thymus by normally repressing Tc17 differentiation and promoting an effector fate outcome.
- Published
- 2019
- Full Text
- View/download PDF
15. The transcription factor c-Myb regulates CD8+ T cell stemness and antitumor immunity
- Author
-
Nicholas P. Restifo, Luca Gattinoni, Rahul Roychoudhuri, Timothy P. Bender, Avinash Bhandoola, Philip Brohawn, Veena Kapoor, Wei Zhu, John B. Le Gall, Sanjivan Gautam, Brandon W. Higgs, Devikala Gurusamy, Zhiya Yu, Yun Ji, James D. Hocker, Nga V. Hawk, Neal E. Lacey, Jessica Fioravanti, Jinhui Hu, William G. Telford, and Hai-Hui Xue
- Subjects
0301 basic medicine ,Adoptive cell transfer ,T cell ,Immunology ,Biology ,Article ,Cell biology ,03 medical and health sciences ,Transactivation ,030104 developmental biology ,0302 clinical medicine ,medicine.anatomical_structure ,medicine ,Immunology and Allergy ,Cytotoxic T cell ,MYB ,Stem cell ,Transcription factor ,CD8 ,030215 immunology - Abstract
Stem cells are maintained by transcriptional programs that promote self-renewal and repress differentiation. Here, we found that the transcription factor c-Myb was essential for generating and maintaining stem cells in the CD8+ T cell memory compartment. Following viral infection, CD8+ T cells lacking Myb underwent terminal differentiation and generated fewer stem cell-like central memory cells than did Myb-sufficient T cells. c-Myb acted both as a transcriptional activator of Tcf7 (which encodes the transcription factor Tcf1) to enhance memory development and as a repressor of Zeb2 (which encodes the transcription factor Zeb2) to hinder effector differentiation. Domain-mutagenesis experiments revealed that the transactivation domain of c-Myb was necessary for restraining differentiation, whereas its negative regulatory domain was critical for cell survival. Myb overexpression enhanced CD8+ T cell memory formation, polyfunctionality and recall responses that promoted curative antitumor immunity after adoptive transfer. These findings identify c-Myb as a pivotal regulator of CD8+ T cell stemness and highlight its therapeutic potential.
- Published
- 2019
16. Publisher Correction: Tcf1 preprograms the mobilization of glycolysis in central memory CD8+ T cells during recall responses
- Author
-
Qiang Shan, Shengen Shawn Hu, Shaoqi Zhu, Xia Chen, Vladimir P. Badovinac, Weiqun Peng, Chongzhi Zang, and Hai-Hui Xue
- Subjects
Immunology ,Immunology and Allergy - Published
- 2022
- Full Text
- View/download PDF
17. Tle corepressors are differentially partitioned to instruct CD8+ T cell lineage choice and identity
- Author
-
Weiqun Peng, David A. Sweetser, Shaojun Xing, Shuyang Yu, Xudong Zhao, Wooseok Seo, Fengyin Li, Jianfeng Wang, Peng Shao, Ichiro Taniuchi, Xiang Li, Justin C. Wheat, Selvi Ramasamy, Chengyu Liu, and Hai-Hui Xue
- Subjects
CD4-Positive T-Lymphocytes ,0301 basic medicine ,animal structures ,Co-Repressor Proteins ,Lineage (genetic) ,T cell ,Immunology ,CD8-Positive T-Lymphocytes ,Biology ,behavioral disciplines and activities ,Article ,03 medical and health sciences ,medicine ,Animals ,Immunology and Allergy ,Cytotoxic T cell ,Cell Lineage ,Gene ,Transcription factor ,Research Articles ,FOXP3 ,nervous system diseases ,Cell biology ,Mice, Inbred C57BL ,Repressor Proteins ,Core Binding Factor Alpha 3 Subunit ,030104 developmental biology ,medicine.anatomical_structure ,nervous system ,psychological phenomena and processes ,Gene Deletion ,CD8 ,Transcription Factors - Abstract
Xing et al demonstrate the requirements for Tle transcriptional corepressors in CD8+ T cell development. Tle proteins are differentially partitioned to the Runx and Tcf/Lef complexes to promote CD8+ lineage choice and establish CD8+ T cell identity, respectively., Tle/Groucho proteins are transcriptional corepressors interacting with Tcf/Lef and Runx transcription factors, but their physiological roles in T cell development remain unknown. Conditional targeting of Tle1, Tle3 and Tle4 revealed gene dose–dependent requirements for Tle proteins in CD8+ lineage cells. Upon ablating all three Tle proteins, generation of CD8+ T cells was greatly diminished, largely owing to redirection of MHC-I–selected thymocytes to CD4+ lineage; the remaining CD8-positive T cells showed aberrant up-regulation of CD4+ lineage-associated genes including Cd4, Thpok, St8sia6, and Foxp3. Mechanistically, Tle3 bound to Runx-occupied Thpok silencer, in post-selection double-positive thymocytes to prevent excessive ThPOK induction and in mature CD8+ T cells to silence Thpok expression. Tle3 also bound to Tcf1-occupied sites in a few CD4+ lineage-associated genes, including Cd4 silencer and St8sia6 introns, to repress their expression in mature CD8+ T cells. These findings indicate that Tle corepressors are differentially partitioned to Runx and Tcf/Lef complexes to instruct CD8+ lineage choice and cooperatively establish CD8+ T cell identity, respectively., Graphical Abstract
- Published
- 2018
- Full Text
- View/download PDF
18. Differential Requirements for Tcf1 Long Isoforms in CD8+ and CD4+ T Cell Responses to Acute Viral Infection
- Author
-
Weiqun Peng, Vladimir P. Badovinac, Jodi A. Gullicksrud, Zhouhao Zeng, Hai-Hui Xue, Shaojun Xing, John T. Harty, and Fengyin Li
- Subjects
0301 basic medicine ,Cellular differentiation ,T cell ,Immunology ,Biology ,Cell biology ,TCIRG1 ,03 medical and health sciences ,Interleukin 21 ,030104 developmental biology ,medicine.anatomical_structure ,medicine ,Immunology and Allergy ,Cytotoxic T cell ,IL-2 receptor ,Antigen-presenting cell ,CD8 - Abstract
In response to acute viral infection, activated naive T cells give rise to effector T cells that clear the pathogen and memory T cells that persist long-term and provide heightened protection. T cell factor 1 (Tcf1) is essential for several of these differentiation processes. Tcf1 is expressed in multiple isoforms, with all isoforms sharing the same HDAC and DNA-binding domains and the long isoforms containing a unique N-terminal β-catenin–interacting domain. In this study, we specifically ablated Tcf1 long isoforms in mice, while retaining expression of Tcf1 short isoforms. During CD8+ T cell responses, Tcf1 long isoforms were dispensable for generating cytotoxic CD8+ effector T cells and maintaining memory CD8+ T cell pool size, but they contributed to optimal maturation of central memory CD8+ T cells and their optimal secondary expansion in a recall response. In contrast, Tcf1 long isoforms were required for differentiation of T follicular helper (TFH) cells, but not TH1 effectors, elicited by viral infection. Although Tcf1 short isoforms adequately supported Bcl6 and ICOS expression in TFH cells, Tcf1 long isoforms remained important for suppressing the expression of Blimp1 and TH1-associated genes and for positively regulating Id3 to restrain germinal center TFH cell differentiation. Furthermore, formation of memory TH1 and memory TFH cells strongly depended on Tcf1 long isoforms. These data reveal that Tcf1 long and short isoforms have distinct, yet complementary, functions and may represent an evolutionarily conserved means to ensure proper programming of CD8+ and CD4+ T cell responses to viral infection.
- Published
- 2017
- Full Text
- View/download PDF
19. The transcription factor Runx3 guards cytotoxic CD8+ effector T cells against deviation towards follicular helper T cell lineage
- Author
-
Natalija Van Braeckel-Budimir, Steven M. Varga, Fengyin Li, Jodi A. Gullicksrud, Ichiro Taniuchi, Vladimir P. Badovinac, Matthew D. Martin, Qiang Shan, Weiqun Peng, Stacey M. Hartwig, Hai-Hui Xue, Yao Su, Samarchith P. Kurup, John T. Harty, Zhouhao Zeng, and Shaojun Xing
- Subjects
0301 basic medicine ,T cell ,Immunology ,Innate lymphoid cell ,Transcription factor complex ,Biology ,BCL6 ,digestive system diseases ,03 medical and health sciences ,Interleukin 21 ,030104 developmental biology ,medicine.anatomical_structure ,Cancer research ,medicine ,Immunology and Allergy ,Cytotoxic T cell ,IL-2 receptor ,CD8 - Abstract
Activated CD8+ T cells differentiate into cytotoxic effector (TEFF) cells that eliminate target cells. How TEFF cell identity is established and maintained is not fully understood. We found that Runx3 deficiency limited clonal expansion and impaired upregulation of cytotoxic molecules in TEFF cells. Runx3-deficient CD8+ TEFF cells aberrantly upregulated genes characteristic of follicular helper T (TFH) cell lineage, including Bcl6, Tcf7 and Cxcr5. Mechanistically, the Runx3-CBFβ transcription factor complex deployed H3K27me3 to Bcl6 and Tcf7 genes to suppress the TFH program. Ablating Tcf7 in Runx3-deficient CD8+ TEFF cells prevented the upregulation of TFH genes and ameliorated their defective induction of cytotoxic genes. As such, Runx3-mediated Tcf7 repression coordinately enforced acquisition of cytotoxic functions and protected the cytotoxic lineage integrity by preventing TFH-lineage deviation.
- Published
- 2017
- Full Text
- View/download PDF
20. Control of Lymphocyte Fate, Infection, and Tumor Immunity by TCF-1
- Author
-
Lisa A. Mielke, Dinesh Raghu, and Hai-Hui Xue
- Subjects
0301 basic medicine ,Lineage (genetic) ,Lymphocyte ,T cell ,Immunology ,Biology ,CD8-Positive T-Lymphocytes ,03 medical and health sciences ,Mice ,0302 clinical medicine ,Neoplasms ,medicine ,T Cell Transcription Factor 1 ,Immunology and Allergy ,Animals ,Humans ,Transcription factor ,Disease Resistance ,Innate lymphoid cell ,Immunity ,Cell Differentiation ,Immune checkpoint ,Cell biology ,030104 developmental biology ,medicine.anatomical_structure ,Gene Expression Regulation ,Virus Diseases ,Histone deacetylase ,CD8 ,030215 immunology - Abstract
T cell factor-1 (TCF-1), encoded by Tcf7, is a transcription factor and histone deacetylase (HDAC) essential for commitment to both the T cell and the innate lymphoid cell (ILC) lineages in mammals. In this review, we discuss the multifunctional role of TCF-1 in establishing these lineages and the requirement for TCF-1 throughout lineage differentiation and maintenance of lineage stability. We highlight recent reports showing promise for TCF-1 as a novel biomarker to identify recently characterized subsets of exhausted CD8+ T cells that may help to predict patient responses to immune checkpoint blockade (ICB).
- Published
- 2019
21. Bystander responses impact accurate detection of murine and human antigen-specific CD8 T cells
- Author
-
Hai-Hui Xue, Isaac J. Jensen, Andrew S. Ishizuka, Qiang Shan, Mitchell N. Lefebvre, Vladimir P. Badovinac, John T. Harty, Matthew D. Martin, and Robert A. Seder
- Subjects
0301 basic medicine ,T cell ,Biology ,CD8-Positive T-Lymphocytes ,Vaccines, Attenuated ,03 medical and health sciences ,Interferon-gamma ,Mice ,0302 clinical medicine ,Immune system ,Antigen ,Malaria Vaccines ,Bystander effect ,medicine ,Cytotoxic T cell ,Animals ,Humans ,Antigens ,Brefeldin A ,Malaria vaccine ,General Medicine ,Bystander Effect ,Acquired immune system ,Malaria ,Mice, Inbred C57BL ,030104 developmental biology ,medicine.anatomical_structure ,Phenotype ,030220 oncology & carcinogenesis ,Immunology ,Leukocytes, Mononuclear ,Cytokines ,Female ,Immunization ,Immunologic Memory ,CD8 ,Spleen ,Signal Transduction ,Research Article - Abstract
Induction of memory CD8(+) T cells is important for controlling infections such as malaria and HIV/AIDS and for cancer immunotherapy. Accurate assessment of antigen-specific (Ag-specific) CD8(+) T cells is critical for vaccine optimization and for defining correlates of protection. However, conditions for determining Ag-specific CD8(+) T cell responses ex vivo using intracellular cytokine staining (ICS) may be variable, especially in humans with complex antigens. Here, we used an attenuated whole parasite malaria vaccine model in humans and various experimental infections in mice to show that the duration of antigenic stimulation and timing of brefeldin A (BFA) addition influence the magnitude of Ag-specific and bystander T cell responses. Indeed, after immunization with an attenuated whole sporozoite malaria vaccine in humans, significantly higher numbers of IFN-γ–producing memory CD8(+) T cells comprising Ag-specific and bystander responses were detected when the duration of Ag stimulation prior to addition of BFA was increased. Mechanistic analyses of virus-specific CD8(+) T cells in mice revealed that the increase in IFN-γ–producing CD8(+) T cells was due to bystander activation of Ag-experienced memory CD8(+) T cells, and correlated with the proportion of Ag-experienced CD8(+) T cells in the stimulated populations. Incubation with anti-cytokine antibodies (e.g., IL-12) improved accuracy in detecting bona fide memory CD8(+) T cell responses, suggesting this as the mechanism for the bystander activation. These data have important implications for accurate assessment of immune responses generated by vaccines intended to elicit protective memory CD8(+) T cells.
- Published
- 2019
22. Cutting Edge: Tcf1 Instructs T Follicular Helper Cell Differentiation by Repressing Blimp1 in Response to Acute Viral Infection
- Author
-
Fengyin Li, Hai-Hui Xue, Peng Shao, Chengyu Liu, Xia Chen, and Jinyong Wang
- Subjects
Cellular differentiation ,Immunology ,Cell ,Biology ,Lymphocyte Activation ,Article ,03 medical and health sciences ,Mice ,0302 clinical medicine ,Downregulation and upregulation ,medicine ,Immunology and Allergy ,Animals ,Hepatocyte Nuclear Factor 1-alpha ,Gene ,Psychological repression ,T follicular helper cell differentiation ,Germinal center ,Cell Differentiation ,T-Lymphocytes, Helper-Inducer ,BCL6 ,Germinal Center ,Mice, Mutant Strains ,Cell biology ,Mice, Inbred C57BL ,medicine.anatomical_structure ,Gene Expression Regulation ,Virus Diseases ,Positive Regulatory Domain I-Binding Factor 1 ,030215 immunology - Abstract
Differentiation of T follicular helper (TFH) cells is regulated by a complex transcriptional network, with mutually antagonistic Bcl6–Blimp1 as a core regulatory axis. It is well established that Tcf1 acts upstream of Bcl6 for its optimal induction to program TFH cell differentiation. In this study, we show that whereas genetic ablation of Tcf1 in mice greatly diminished TFH cells in response to viral infection, compound deletion of Blimp1 with Tcf1 restored TFH cell frequency, numbers, and generation of germinal center B cells. Aberrant upregulation of T-bet and Id2 in Tcf1-deficient TFH cells was also largely rectified by ablating Blimp1. Tcf1 chromatin immunoprecipitation sequencing in TFH cells identified two strong Tcf1 binding sites in the Blimp1 gene at a 24-kb upstream and an intron-3 element. Deletion of the intron-3 element, but not the 24-kb upstream element, compromised production of TFH cells. Our data demonstrate that Tcf1-mediated Blimp1 repression is functionally critical for safeguarding TFH cell differentiation.
- Published
- 2019
23. The transcription factor TCF-1 enforces commitment to the innate lymphoid cell lineage
- Author
-
Binbin Lai, Keji Zhao, Avinash Bhandoola, Tobias Raabe, Gang Ren, Christelle Harly, Hai-Hui Xue, Qi Yang, Devin Kenney, Margaret C. Cam, Bernardo, Elizabeth, Immunobiology of Human αβ and γδ T Cells and Immunotherapeutic Applications (CRCINA-ÉQUIPE 1), Centre de Recherche en Cancérologie et Immunologie Nantes-Angers (CRCINA), Université d'Angers (UA)-Université de Nantes (UN)-Institut National de la Santé et de la Recherche Médicale (INSERM)-Centre National de la Recherche Scientifique (CNRS)-Centre hospitalier universitaire de Nantes (CHU Nantes)-Université d'Angers (UA)-Université de Nantes (UN)-Institut National de la Santé et de la Recherche Médicale (INSERM)-Centre National de la Recherche Scientifique (CNRS)-Centre hospitalier universitaire de Nantes (CHU Nantes), Laboratory of Genome Integrity [Bethesda, MD, USA] (Center for Cancer Research), National Institutes of Health [Bethesda] (NIH)-National Cancer Institute [Bethesda] (NCI-NIH), National Institutes of Health [Bethesda] (NIH), Systems Biology Center [Bethesda, MD, USA] (NIH/NHLBI ), National Institutes of Health [Bethesda] (NIH)-National Heart, Lung, and Blood Institute [Bethesda] (NHLBI), Division of Translational Medicine and Human Genetics [Philadelphia, PL, USA] (Department of Medicine), University of Pennsylvania-Perelman School of Medicine, University of Pennsylvania, Department of Immunology and Microbial Disease [Albany, NY, USA], Albany Medical College, Office of Science and Technology Resources [Bethesda, MD, USA] (CCR/NIH), Department of Microbiology, Interdisciplinary Immunology Graduate Program [Iowa City, IA, USA], Carver College of Medicine, University of Iowa, This research was supported by the Intramural Research Program of the NIH, National Cancer Institute and Center for Cancer Research, and by grants from the NIH (AI121080 and AI139874 to H.-H.X.), Veteran Affairs BLR&D Merit Review Program (BX002903A to H.H.X.) and Foundation pour la Recherche Médicale (DEQ20170839118 to C.H.)., Institut National de la Santé et de la Recherche Médicale (INSERM)-Université de Nantes - UFR de Médecine et des Techniques Médicales (UFR MEDECINE), Université de Nantes (UN)-Université de Nantes (UN)-Centre hospitalier universitaire de Nantes (CHU Nantes)-Centre National de la Recherche Scientifique (CNRS)-Université d'Angers (UA)-Institut National de la Santé et de la Recherche Médicale (INSERM)-Université de Nantes - UFR de Médecine et des Techniques Médicales (UFR MEDECINE), Université de Nantes (UN)-Université de Nantes (UN)-Centre hospitalier universitaire de Nantes (CHU Nantes)-Centre National de la Recherche Scientifique (CNRS)-Université d'Angers (UA), University of Pennsylvania [Philadelphia]-Perelman School of Medicine, and University of Pennsylvania [Philadelphia]
- Subjects
0301 basic medicine ,Transcription, Genetic ,T-Lymphocytes ,T cell ,Immunology ,innate lymphoid cells ,lineage commitment ,[SDV.CAN]Life Sciences [q-bio]/Cancer ,Biology ,DC ,Article ,Mice ,03 medical and health sciences ,0302 clinical medicine ,[SDV.CAN] Life Sciences [q-bio]/Cancer ,Transcription (biology) ,medicine ,Animals ,Immunology and Allergy ,Cell Lineage ,Hepatocyte Nuclear Factor 1-alpha ,Progenitor cell ,skin and connective tissue diseases ,Transcription factor ,Cells, Cultured ,Tissue homeostasis ,TCF-1 ,Innate lymphoid cell ,Cell Differentiation ,Dendritic Cells ,Dendritic cell ,Lymphoid Progenitor Cells ,Cell biology ,Chromatin ,body regions ,Mice, Inbred C57BL ,030104 developmental biology ,medicine.anatomical_structure ,ILC ,Gene Expression Regulation ,030215 immunology - Abstract
International audience; Innate lymphoid cells (ILCs) play important functions in immunity and tissue homeostasis, but their development is poorly understood. Through the use of single-cell approaches, we examined the transcriptional and functional heterogeneity of ILC progenitors, and studied the precursor-product relationships that link the subsets identified. This analysis identified two successive stages of ILC development within T cell factor 1-positive (TCF-1+) early innate lymphoid progenitors (EILPs), which we named 'specified EILPs' and 'committed EILPs'. Specified EILPs generated dendritic cells, whereas this potential was greatly decreased in committed EILPs. TCF-1 was dispensable for the generation of specified EILPs, but required for the generation of committed EILPs. TCF-1 used a pre-existing regulatory landscape established in upstream lymphoid precursors to bind chromatin in EILPs. Our results provide insight into the mechanisms by which TCF-1 promotes developmental progression of ILC precursors, while constraining their dendritic cell lineage potential and enforcing commitment to ILC fate.
- Published
- 2019
- Full Text
- View/download PDF
24. CD4+ T cell effector commitment coupled to self-renewal by asymmetric cell divisions
- Author
-
Kyra D. Zens, William C. Adams, Nyanza J. Rothman, Wen-Hsuan W. Lin, Yen-Hua Chen, Radomir Kratchmarov, Bonnie Yen, Simone A. Nish, Avinash Bhandoola, Steven L. Reiner, Hai-Hui Xue, and Donna L. Farber
- Subjects
CD4-Positive T-Lymphocytes ,0301 basic medicine ,Cell division ,T cell ,Cellular differentiation ,Immunology ,Cell ,Cell fate determination ,Biology ,Mice ,Phosphatidylinositol 3-Kinases ,03 medical and health sciences ,0302 clinical medicine ,Antigen ,medicine ,Animals ,Immunology and Allergy ,Hepatocyte Nuclear Factor 1-alpha ,Cells, Cultured ,Research Articles ,Effector ,TOR Serine-Threonine Kinases ,Asymmetric Cell Division ,Brief Definitive Report ,Cell biology ,Mice, Inbred C57BL ,030104 developmental biology ,medicine.anatomical_structure ,Signal transduction ,Cell Division ,030215 immunology - Abstract
Nish et al. report that production of a fully committed Th1 effector cell occurs during an asymmetric cell division wherein the other daughter cell remains memory cell–like. Unequal transmission of metabolic signaling may be the driver of this regenerative behavior., Upon infection, an activated CD4+ T cell produces terminally differentiated effector cells and renews itself for continued defense. In this study, we show that differentiation and self-renewal arise as opposing outcomes of sibling CD4+ T cells. After influenza challenge, antigen-specific cells underwent several divisions in draining lymph nodes (LN; DLNs) while maintaining expression of TCF1. After four or five divisions, some cells silenced, whereas some cells maintained TCF1 expression. TCF1-silenced cells were T helper 1–like effectors and concentrated in the lungs. Cells from earliest divisions were memory-like and concentrated in nondraining LN. TCF1-expressing cells from later divisions in the DLN could self-renew, clonally yielding a TCF1-silenced daughter cell as well as a sibling cell maintaining TCF1 expression. Some TCF1-expressing cells in DLNs acquired an alternative, follicular helper-like fate. Modeled differentiation experiments in vitro suggested that unequal PI3K/mechanistic target of rapamycin signaling drives intraclonal cell fate heterogeneity. Asymmetric division enables self-renewal to be coupled to production of differentiated CD4+ effector T cells during clonal selection.
- Published
- 2016
- Full Text
- View/download PDF
25. CD8 + T Cells Utilize Highly Dynamic Enhancer Repertoires and Regulatory Circuitry in Response to Infections
- Author
-
Peng Gao, Bing He, Jodi A. Gullicksrud, Kai Tan, John T. Harty, Vladimir P. Badovinac, Hai-Hui Xue, Changya Chen, Qiang Shan, Shaojun Xing, Li Teng, Matthew D. Martin, and Shuyang Yu
- Subjects
0301 basic medicine ,Genetics ,T cell ,Immunology ,Enhancer RNAs ,Biology ,Cell biology ,03 medical and health sciences ,030104 developmental biology ,0302 clinical medicine ,Infectious Diseases ,medicine.anatomical_structure ,030220 oncology & carcinogenesis ,medicine ,Immunology and Allergy ,Cytotoxic T cell ,Epigenetics ,Enhancer ,Transcription factor ,Chromatin immunoprecipitation ,Epigenomics - Abstract
Differentiation of effector and memory CD8+ T cells is accompanied by extensive changes in the transcriptome and histone modifications at gene promoters; however, the enhancer repertoire and associated gene regulatory networks are poorly defined. Using histone mark chromatin immunoprecipitation coupled with deep sequencing, we mapped the enhancer and super-enhancer landscapes in antigen-specific naive, differentiated effector, and central memory CD8+ T cells during LCMV infection. Epigenomics-based annotation revealed a highly dynamic repertoire of enhancers, which were inherited, de novo activated, decommissioned and re-activated during CD8+ T cell responses. We employed a computational algorithm to pair enhancers with target gene promoters. On average, each enhancer targeted three promoters and each promoter was regulated by two enhancers. By identifying enriched transcription factor motifs in enhancers, we defined transcriptional regulatory circuitry at each CD8+ T cell response stage. These multi-dimensional datasets provide a blueprint for delineating molecular mechanisms underlying functional differentiation of CD8+ T cells.
- Published
- 2016
- Full Text
- View/download PDF
26. Defining CD8+ T cells that provide the proliferative burst after PD-1 therapy
- Author
-
Hai-Hui Xue, Haydn T. Kissick, Se Jin Im, Arlene H. Sharpe, Gordon J. Freeman, Helder I. Nakaya, Rafi Ahmed, Qiang Shan, J. Scott Hale, Michael Y. Gerner, Tahseen H. Nasti, Ronald N. Germain, Matheus Carvalho Bürger, Judong Lee, Masao Hashimoto, and Junghwa Lee
- Subjects
0301 basic medicine ,T cell ,Cellular differentiation ,Programmed Cell Death 1 Receptor ,CD8-Positive T-Lymphocytes ,Lymphocytic Choriomeningitis ,Biology ,Inducible T-Cell Co-Stimulator Protein ,Mice ,03 medical and health sciences ,Interleukin 21 ,0302 clinical medicine ,CD28 Antigens ,T-Lymphocyte Subsets ,medicine ,Animals ,Lymphocytic choriomeningitis virus ,Cytotoxic T cell ,Hepatocyte Nuclear Factor 1-alpha ,Cell Self Renewal ,Cell Proliferation ,Multidisciplinary ,CD28 ,Cell Differentiation ,T-Lymphocytes, Helper-Inducer ,Hematopoietic Stem Cells ,CÉLULAS ,Haematopoiesis ,030104 developmental biology ,medicine.anatomical_structure ,030220 oncology & carcinogenesis ,Immunology ,Cancer research ,Female ,Immunotherapy ,Stem cell ,CD8 - Abstract
Chronic viral infections are characterized by a state of CD8+ T-cell dysfunction that is associated with expression of the programmed cell death 1 (PD-1) inhibitory receptor. A better understanding of the mechanisms that regulate CD8+ T-cell responses during chronic infection is required to improve immunotherapies that restore function in exhausted CD8+ T cells. Here we identify a population of virus-specific CD8+ T cells that proliferate after blockade of the PD-1 inhibitory pathway in mice chronically infected with lymphocytic choriomeningitis virus (LCMV). These LCMV-specific CD8+ T cells expressed the PD-1 inhibitory receptor, but also expressed several costimulatory molecules such as ICOS and CD28. This CD8+ T-cell subset was characterized by a unique gene signature that was related to that of CD4+ T follicular helper (TFH) cells, CD8+ T cell memory precursors and haematopoietic stem cell progenitors, but that was distinct from that of CD4+ TH1 cells and CD8+ terminal effectors. This CD8+ T-cell population was found only in lymphoid tissues and resided predominantly in the T-cell zones along with naive CD8+ T cells. These PD-1+CD8+ T cells resembled stem cells during chronic LCMV infection, undergoing self-renewal and also differentiating into the terminally exhausted CD8+ T cells that were present in both lymphoid and non-lymphoid tissues. The proliferative burst after PD-1 blockade came almost exclusively from this CD8+ T-cell subset. Notably, the transcription factor TCF1 had a cell-intrinsic and essential role in the generation of this CD8+ T-cell subset. These findings provide a better understanding of T-cell exhaustion and have implications in the optimization of PD-1-directed immunotherapy in chronic infections and cancer.
- Published
- 2016
- Full Text
- View/download PDF
27. Tcf1 and Lef1 are required for the immunosuppressive function of regulatory T cells
- Author
-
Hai-Hui Xue, William J. Paradee, Xin Zhao, David K. Meyerholz, Zhouhao Zeng, Xiang Li, Weiqun Peng, Kexin Gai, Shaojun Xing, Peng Shao, Xia Chen, and Xudong Zhao
- Subjects
Male ,Lymphoid Enhancer-Binding Factor 1 ,T cell ,Immunology ,chemical and pharmacologic phenomena ,Mice, Transgenic ,Biology ,CD8-Positive T-Lymphocytes ,medicine.disease_cause ,T-Lymphocytes, Regulatory ,Article ,Immune tolerance ,Autoimmunity ,03 medical and health sciences ,Mice ,0302 clinical medicine ,immune system diseases ,hemic and lymphatic diseases ,PRDM1 ,medicine ,Immune Tolerance ,Immunology and Allergy ,Cytotoxic T cell ,Animals ,Hepatocyte Nuclear Factor 1-alpha ,Research Articles ,030304 developmental biology ,0303 health sciences ,Effector ,Dextran Sulfate ,FOXP3 ,hemic and immune systems ,Cell Differentiation ,Colitis ,3. Good health ,Cell biology ,Mice, Inbred C57BL ,Disease Models, Animal ,medicine.anatomical_structure ,embryonic structures ,Female ,Transcriptome ,CD8 ,030215 immunology - Abstract
Tcf1 and Lef1 are underexpressed in T reg cells compared with conventional CD4+ T cells. Xing et al. demonstrate that genetic ablation of both factors impairs immunosuppressive function of T reg cells and leads to spontaneous multi-organ autoimmunity., Tcf1 and Lef1 have versatile functions in regulating T cell development and differentiation, but intrinsic requirements for these factors in regulatory T (T reg) cells remain to be unequivocally defined. Specific ablation of Tcf1 and Lef1 in T reg cells resulted in spontaneous multi-organ autoimmunity that became more evident with age. Tcf1/Lef1-deficient T regs showed reduced protection against experimentally induced colitis, indicative of diminished immuno-suppressive capacity. Transcriptomic analysis revealed that Tcf1 and Lef1 were responsible for positive regulation of a subset of T reg–overrepresented signature genes such as Ikzf4 and Izumo1r. Unexpectedly, Tcf1 and Lef1 were necessary for restraining expression of cytotoxic CD8+ effector T cell–associated genes in T reg cells, including Prdm1 and Ifng. Tcf1 ChIP-seq revealed substantial overlap between Tcf1 and Foxp3 binding peaks in the T reg cell genome, with Tcf1-Foxp3 cooccupancy observed at key T reg signature and cytotoxic effector genes. Our data collectively indicate that Tcf1 and Lef1 are critical for sustaining T reg suppressive functions and preventing loss of self-tolerance., Graphical Abstract
- Published
- 2018
28. Polymicrobial sepsis influences NK-cell-mediated immunity by diminishing NK-cell-intrinsic receptor-mediated effector responses to viral ligands or infections
- Author
-
Christina S. Winborn, Thomas S. Griffith, Mikaela M. Tremblay, Christopher M. Snyder, Sandeep K. Tripathy, Qiang Shan, Micaela G. Fosdick, Hai-Hui Xue, Peng Shao, Jon C. D. Houtman, Isaac J. Jensen, and Vladimir P. Badovinac
- Subjects
0301 basic medicine ,Muromegalovirus ,Physiology ,Gene Expression ,Apoptosis ,Pathology and Laboratory Medicine ,Mice ,0302 clinical medicine ,Cell Signaling ,Immune Physiology ,Medicine and Health Sciences ,Receptor ,lcsh:QH301-705.5 ,Cells, Cultured ,Mice, Knockout ,Immunity, Cellular ,Innate Immune System ,Cell Death ,Effector ,Experimental Design ,Signal transducing adaptor protein ,3. Good health ,Killer Cells, Natural ,Research Design ,Cell Processes ,Cytomegalovirus Infections ,Cytokines ,NK Cell Lectin-Like Receptor Subfamily A ,Research Article ,Signal Transduction ,lcsh:Immunologic diseases. Allergy ,Secondary infection ,Immunology ,Surgical and Invasive Medical Procedures ,Biology ,Research and Analysis Methods ,Microbiology ,Sepsis ,03 medical and health sciences ,Signs and Symptoms ,Immunity ,Diagnostic Medicine ,Virology ,medicine ,Genetics ,Animals ,Calcium Signaling ,Molecular Biology ,Perforin ,Biology and Life Sciences ,Cell Biology ,Molecular Development ,medicine.disease ,Mice, Inbred C57BL ,030104 developmental biology ,lcsh:Biology (General) ,Immune System ,Parasitology ,Cytokine storm ,lcsh:RC581-607 ,Spleen ,030215 immunology ,Developmental Biology - Abstract
The sepsis-induced cytokine storm leads to severe lymphopenia and reduced effector capacity of remaining/surviving cells. This results in a prolonged state of immunoparalysis, that contributes to enhanced morbidity/mortality of sepsis survivors upon secondary infection. The impact of sepsis on several lymphoid subsets has been characterized, yet its impact on NK-cells remains underappreciated–despite their critical role in controlling infection(s). Here, we observed numerical loss of NK-cells in multiple tissues after cecal-ligation-and-puncture (CLP)-induced sepsis. To elucidate the sepsis-induced lesions in surviving NK-cells, transcriptional profiles were evaluated and indicated changes consistent with impaired effector functionality. A corresponding deficit in NK-cell capacity to produce effector molecules following secondary infection and/or cytokine stimulation (IL-12,IL-18) further suggested a sepsis-induced NK-cell intrinsic impairment. To specifically probe NK-cell receptor-mediated function, the activating Ly49H receptor, that recognizes the murine cytomegalovirus (MCMV) m157 protein, served as a model receptor. Although relative expression of Ly49H receptor did not change, the number of Ly49H+ NK-cells in CLP hosts was reduced leading to impaired in vivo cytotoxicity and the capacity of NK-cells (on per-cell basis) to perform Ly49H-mediated degranulation, killing, and effector molecule production in vitro was also severely reduced. Mechanistically, Ly49H adaptor protein (DAP12) activation and clustering, assessed by TIRF microscopy, was compromised. This was further associated with diminished AKT phosphorylation and capacity to flux calcium following receptor stimulation. Importantly, DAP12 overexpression in NK-cells restored Ly49H/D receptors-mediated effector functions in CLP hosts. Finally, as a consequence of sepsis-dependent numerical and functional lesions in Ly49H+ NK-cells, host capacity to control MCMV infection was significantly impaired. Importantly, IL-2 complex (IL-2c) therapy after CLP improved numbers but not a function of NK-cells leading to enhanced immunity to MCMV challenge. Thus, the sepsis-induced immunoparalysis state includes numerical and NK-cell-intrinsic functional impairments, an instructive notion for future studies aimed in restoring NK-cell immunity in sepsis survivors., Author summary Sepsis is an exaggerated host response to infection that can initially lead to significant morbidity/mortality and a long-lasting state of immunoparalysis in sepsis survivors. Sepsis-induced immunoparalysis functionally impairs numerous lymphocyte populations, including NK-cells. However, the scope and underlying mechanisms of NK-cell impairment and the consequences for NK-cell-mediated pathogen control remain underappreciated. NK-cells contribute to early host control of pathogens through a balance of activating and inhibitory receptors, and alterations in the number and capacity of NK-cells to exert receptor-mediated immunity can lead to dramatic impairment in host control of infection. The present study defines sepsis-induced numerical and cell-intrinsic functional impairments in NK-cell response to cytokine stimulation and receptor signaling that contribute to impaired host capacity to mount NK-cell-mediated effector responses and provide protection to bacterial and/or viral pathogens. Impairments in receptor signaling were due to reduced expression of adaptor protein DAP12. Importantly, the diminished ability of NK-cells from CLP hosts to provide anti-viral (MCMV) immunity is partially restored by IL-2 complex (IL-2c) therapy, which increased the number, but not function, of protective Ly49H+ NK-cells. Thus, these findings define sepsis-induced changes of the NK-cell compartment and provide insight into potential therapeutic interventions aimed at resolving sepsis-induced immunoparalysis in sepsis survivors.
- Published
- 2018
29. Time and Antigen-Stimulation History Influence Memory CD8 T Cell Bystander Responses
- Author
-
Matthew D. Martin, Qiang Shan, Hai-Hui Xue, and Vladimir P. Badovinac
- Subjects
0301 basic medicine ,lcsh:Immunologic diseases. Allergy ,medicine.medical_treatment ,Immunology ,Inflammation ,CD8 T cells ,Biology ,Proinflammatory cytokine ,bystander responses ,antigen-exposure history ,memory ,03 medical and health sciences ,0302 clinical medicine ,Antigen ,Bystander effect ,medicine ,Immunology and Allergy ,Cytotoxic T cell ,IL-2 receptor ,Original Research ,time-dependent functions ,cytokines ,Cytolysis ,030104 developmental biology ,Cytokine ,medicine.symptom ,lcsh:RC581-607 ,030215 immunology - Abstract
Memory CD8 T cells can be activated and induced to produce cytokines and increase stores of cytolytic proteins not only in response to cognate antigen (Ag), but also in response to inflammatory cytokines (bystander responses). Importantly, bystander memory CD8 T cell functions have been shown to be dependent upon memory CD8 T cell fitness, since exhausted CD8 T cells have diminished capacity to respond to inflammatory cues. While it is known that memory CD8 T cell functional abilities, including ability to produce cytokines in response to cognate Ag, change with time after initial Ag encounter and upon multiple Ag-stimulations (e.g. primary vs. tertiary CD8 T cell responses), it is unknown if bystander memory CD8 T cell responses are influenced by time or by Ag-exposure history. Here, we examined time and Ag-stimulation history-dependent alterations in virus-specific memory CD8 T cell bystander functions in response to inflammatory cytokines and unrelated bacterial infection. We found that expression of cytokine receptors and ability to produce IFN- following heterologous infection or incubation with inflammatory cytokines decreases with time following initial Ag encounter and increases with additional Ag encounters, suggesting that the ability to sense inflammation and respond with bystander cytokine production is dependent on age and Ag-stimulation history of memory CD8 T cells. These data shed further light on the regulation of memory CD8 T cell effector functions and have important implications for the development of vaccines designed to elicit protective memory CD8 T cells.
- Published
- 2017
- Full Text
- View/download PDF
30. TCF-1 and LEF-1 act upstream of Th-POK to promote the CD4+ T cell fate and interact with Runx3 to silence Cd4 in CD8+ T cells
- Author
-
Hiroshi Kawamoto, Bo Zhou, Shuyang Yu, Kai Tan, Bing He, Hai-Hui Xue, Jun Zhu, Wenjing Yang, Farrah C. Steinke, and Xinyuan Zhou
- Subjects
CD4-Positive T-Lymphocytes ,Male ,animal structures ,Lymphoid Enhancer-Binding Factor 1 ,T cell ,Immunology ,CD8-Positive T-Lymphocytes ,Biology ,Article ,Mice ,03 medical and health sciences ,Interleukin 21 ,0302 clinical medicine ,T Cell Transcription Factor 1 ,medicine ,Animals ,Immunology and Allergy ,Cytotoxic T cell ,Cell Lineage ,Hepatocyte Nuclear Factor 1-alpha ,IL-2 receptor ,Antigen-presenting cell ,STAT4 ,030304 developmental biology ,0303 health sciences ,ZAP70 ,fungi ,Natural killer T cell ,Molecular biology ,Core Binding Factor Alpha 3 Subunit ,medicine.anatomical_structure ,CD4 Antigens ,embryonic structures ,Female ,Transcription Factors ,030215 immunology - Abstract
The transcription factors TCF-1 and LEF-1 are essential for early T cell development, but their roles beyond the CD4(+)CD8(+) double-positive (DP) stage are unknown. By specific ablation of these factors in DP thymocytes, we demonstrated that deficiency in TCF-1 and LEF-1 diminished the output of CD4(+) T cells and redirected CD4(+) T cells to a CD8(+) T cell fate. The role of TCF-1 and LEF-1 in the CD4-versus-CD8 lineage 'choice' was mediated in part by direct positive regulation of the transcription factor Th-POK. Furthermore, loss of TCF-1 and LEF-1 unexpectedly caused derepression of CD4 expression in T cells committed to the CD8(+) lineage without affecting the expression of Runx transcription factors. Instead, TCF-1 physically interacted with Runx3 to cooperatively silence Cd4. Thus, TCF-1 and LEF-1 adopted distinct genetic 'wiring' to promote the CD4(+) T cell fate and establish CD8(+) T cell identity.
- Published
- 2014
- Full Text
- View/download PDF
31. Cutting Edge: β-Catenin-Interacting Tcf1 Isoforms Are Essential for Thymocyte Survival but Dispensable for Thymic Maturation Transitions
- Author
-
Yu-Bin Du, Jodi A. Gullicksrud, Shaojun Xing, Chengyu Liu, Qiang Shan, Zhe Xu, Thomas B. Bair, and Hai-Hui Xue
- Subjects
0301 basic medicine ,Gene isoform ,Cell Survival ,T cell ,T-Lymphocytes ,Immunology ,Apoptosis ,Thymus Gland ,Biology ,Article ,Transcriptome ,03 medical and health sciences ,Mice ,Coactivator ,medicine ,T Cell Transcription Factor 1 ,Immunology and Allergy ,Animals ,Protein Isoforms ,beta Catenin ,Regulation of gene expression ,Mice, Knockout ,Thymocytes ,Cell Differentiation ,Cell biology ,Mice, Inbred C57BL ,Thymocyte ,030104 developmental biology ,medicine.anatomical_structure ,Gene Expression Regulation ,Catenin ,Signal transduction ,Protein Binding ,Signal Transduction - Abstract
T cell factor 1 (Tcf1) is essential for T cell development; however, it remains controversial whether β-catenin, a known coactivator of Tcf1, has a role. Tcf1 is expressed in multiple isoforms in T lineage cells, with the long isoforms interacting with β-catenin through an N-terminal domain. In this study, we specifically ablated Tcf1 long isoforms in mice (p45−/−mice) to abrogate β-catenin interaction. Although thymic cellularity was diminished in p45−/− mice, transition of thymocytes through the maturation stages was unaffected, with no overt signs of developmental blocks. p45−/− thymocytes showed increased apoptosis and alterations in transcriptome, but these changes were substantially more modest than in thymocytes lacking all Tcf1 isoforms. These data indicate that Tcf1–β-catenin interaction is necessary for promoting thymocyte survival to maintain thymic output. Rather than being dominant-negative regulators, Tcf1 short isoforms are adequate in supporting developing thymocytes to traverse through maturation steps and in regulating the expression of most Tcf1 target genes.
- Published
- 2016
32. Critical roles of mTOR Complex 1 and 2 for T follicular helper cell differentiation and germinal center responses
- Author
-
Yun Pan, Xiao-Ping Zhong, Jimin Gao, Xingguang Lin, Hai-Hui Xue, Pengcheng Chen, Jinli Wang, Jialong Yang, and Hongxiang Huang
- Subjects
0301 basic medicine ,Mouse ,QH301-705.5 ,Cellular differentiation ,T cell ,Science ,Immunology ,Mechanistic Target of Rapamycin Complex 2 ,Mechanistic Target of Rapamycin Complex 1 ,Biology ,mTORC2 ,General Biochemistry, Genetics and Molecular Biology ,Mice ,03 medical and health sciences ,0302 clinical medicine ,germinal center B cell ,medicine ,Animals ,T Follicular Helper Cell ,Biology (General) ,Protein kinase B ,PI3K/AKT/mTOR pathway ,B cell ,T follicular helper cell differentiation ,lymphocyte subsets ,General Immunology and Microbiology ,General Neuroscience ,Germinal center ,Cell Differentiation ,T-Lymphocytes, Helper-Inducer ,General Medicine ,Germinal Center ,3. Good health ,Cell biology ,030104 developmental biology ,medicine.anatomical_structure ,mTOR ,Medicine ,signal transduction ,Research Article ,030215 immunology - Abstract
T follicular helper (Tfh) cells play critical roles for germinal center responses and effective humoral immunity. We report here that mTOR in CD4 T cells is essential for Tfh differentiation. In Mtorf/f-Cd4Cre mice, both constitutive and inducible Tfh differentiation is severely impaired, leading to defective germinal center B cell formation and antibody production. Moreover, both mTORC1 and mTORC2 contribute to Tfh and GC B cell development but may do so via distinct mechanisms. mTORC1 mainly promotes CD4 T cell proliferation to reach the cell divisions necessary for Tfh differentiation, while Rictor/mTORC2 regulates Tfh differentiation by promoting Akt activation and TCF1 expression without grossly influencing T cell proliferation. Together, our results reveal crucial but distinct roles for mTORC1 and mTORC2 in CD4 T cells during Tfh differentiation and germinal center responses. DOI: http://dx.doi.org/10.7554/eLife.17936.001
- Published
- 2016
33. The TCF-1 and LEF-1 Transcription Factors Have Cooperative and Opposing Roles in T Cell Development and Malignancy
- Author
-
Xuefang Jing, Xinyuan Zhou, Dong Mei Zhao, Shann Ching Chen, Chengyu Liu, C. Michael Knudson, Shuyang Yu, Oksana Zagorodna, Yoshifumi Yokota, Farrah C. Steinke, Charles G. Mullighan, David K. Meyerholz, and Hai-Hui Xue
- Subjects
animal structures ,Lymphoid Enhancer-Binding Factor 1 ,CD8 Antigens ,T-Lymphocytes ,T cell ,Immunology ,Biology ,Bioinformatics ,Malignancy ,Lymphoma, T-Cell ,Precursor T-Cell Lymphoblastic Leukemia-Lymphoma ,Article ,Malignant transformation ,Mice ,Downregulation and upregulation ,T Cell Transcription Factor 1 ,medicine ,Animals ,Humans ,Immunology and Allergy ,Hepatocyte Nuclear Factor 1-alpha ,Transcription factor ,Inhibitor of Differentiation Protein 2 ,Progenitor ,Thymocytes ,Receptors, Notch ,medicine.disease ,Up-Regulation ,Mice, Inbred C57BL ,Thymocyte ,Cell Transformation, Neoplastic ,Infectious Diseases ,medicine.anatomical_structure ,CD4 Antigens ,embryonic structures ,Cancer research ,CD8 ,Transcription Factors ,Lymphoid enhancer-binding factor 1 - Abstract
Summary The TCF-1 and LEF-1 transcription factors are known to play critical roles in normal thymocyte development. Unexpectedly, we found that TCF-1-deficient ( Tcf7 −/− ) mice developed aggressive T cell malignancy, resembling human T cell acute lymphoblastic leukemia (T-ALL). LEF-1 was aberrantly upregulated in premalignant Tcf7 −/− early thymocytes and lymphoma cells. We further demonstrated that TCF-1 directly repressed LEF-1 expression in early thymocytes and that conditional inactivation of Lef1 greatly delayed or prevented T cell malignancy in Tcf7 −/− mice. In human T-ALLs, an early thymic progenitor (ETP) subtype was associated with diminished TCF7 expression, and two of the ETP-ALL cases harbored TCF7 gene deletions. We also showed that TCF-1 and LEF-1 were dispensable for T cell lineage commitment but instead were required for early thymocytes to mature beyond the CD4 − CD8 − stage. TCF-1 thus has dual roles, i.e., acting cooperatively with LEF-1 to promote thymocyte maturation while restraining LEF-1 expression to prevent malignant transformation of developing thymocytes.
- Published
- 2012
- Full Text
- View/download PDF
34. MLL4 keeps Foxp3 in the loop
- Author
-
Hai-Hui Xue and Dong-Mei Zhao
- Subjects
0301 basic medicine ,biology ,Immunology ,Lymphocyte differentiation ,FOXP3 ,Locus (genetics) ,complex mixtures ,Molecular biology ,Chromatin ,03 medical and health sciences ,030104 developmental biology ,Histone ,biology.protein ,bacteria ,Immunology and Allergy ,Cytotoxic T cell ,IL-2 receptor ,Transcription factor - Abstract
The histone lysine methyltransferase MLL4 primes the locus encoding the transcription factor Foxp3 for transcriptional activation in thymus-derived and inducible regulatory T cells.
- Published
- 2017
- Full Text
- View/download PDF
35. Repetitive Antigen Stimulation Induces Stepwise Transcriptome Diversification but Preserves a Core Signature of Memory CD8+ T Cell Differentiation
- Author
-
Vladimir P. Badovinac, Thomas B. Bair, Deepa Rai, Thomas Wirth, Hai-Hui Xue, Jaime L. Sabel, and John T. Harty
- Subjects
Ovalbumin ,Cellular differentiation ,T cell ,Immunology ,Immunization, Secondary ,Mice, Transgenic ,CD8-Positive T-Lymphocytes ,Biology ,Article ,Immunophenotyping ,Transcriptome ,Mice ,03 medical and health sciences ,0302 clinical medicine ,Antigen ,medicine ,Animals ,Immunology and Allergy ,Cytotoxic T cell ,Listeriosis ,Transgenes ,Antigens ,MOLIMMUNO ,Cells, Cultured ,030304 developmental biology ,0303 health sciences ,SYSBIO ,Gene Expression Profiling ,Cell Differentiation ,Microarray Analysis ,Molecular biology ,Lymphocyte Subsets ,Mice, Inbred C57BL ,Gene expression profiling ,Infectious Diseases ,medicine.anatomical_structure ,CELLIMMUNO ,T cell differentiation ,Immunologic Memory ,CD8 ,030215 immunology - Abstract
Repetitive antigen-stimulation by prime-boost vaccination or pathogen re-encounter increases memory CD8+ T cell numbers, however the impact on memory CD8+ T cell differentiation is unknown. Here we showed that repetitive antigen-stimulations induced accumulation of memory CD8+ T cells with uniform effector memory characteristics. However, genome-wide microarray analyses revealed that each additional antigen-challenge resulted in the differential regulation of several hundred new genes in the ensuing memory CD8+ T cell populations and therefore in stepwise diversification of CD8+ T cell transcriptomes. Thus, primary and repeatedly stimulated (secondary, tertiary, quaternary) memory CD8+ T cells differ substantially in their molecular signature while sharing expression of a small group of genes and biological pathways, which may constitute a core signature of memory differentiation. These results provide new insight into the complex regulation of memory CD8+ T cell differentiation and identify a spectrum of potential new molecular targets to dissect the function of memory cells generated by repeated antigen-stimulation.
- Published
- 2010
- Full Text
- View/download PDF
36. Constitutive Expression of IL-7 Receptor α Does Not Support Increased Expansion or Prevent Contraction of Antigen-Specific CD4 or CD8 T Cells following Listeria monocytogenes Infection
- Author
-
Jodie S. Haring, Julie Bollenbacher-Reilley, Hai-Hui Xue, Warren J. Leonard, Xuefang Jing, and John T. Harty
- Subjects
CD4-Positive T-Lymphocytes ,Cell Survival ,Secondary infection ,T cell ,Immunology ,Epitopes, T-Lymphocyte ,Mice, Transgenic ,CD8-Positive T-Lymphocytes ,Biology ,Lymphocyte Activation ,Epitope ,Mice ,medicine ,Animals ,Immunology and Allergy ,Cytotoxic T cell ,Listeriosis ,Lymphocyte Count ,Interleukin-7 receptor ,Cell Proliferation ,Receptors, Interleukin-7 ,Cell Death ,Cell growth ,Effector ,Listeria monocytogenes ,Growth Inhibitors ,Cell biology ,Mice, Inbred C57BL ,medicine.anatomical_structure ,Immunologic Memory ,Memory T cell - Abstract
Expression of IL-7Rα (CD127) has been suggested as a major determinant in the survival of memory T cell precursors. We investigated whether constitutive expression of IL-7Rα on T cells increased expansion and/or decreased contraction of endogenous Ag-specific CD4 and CD8 T cells following infection with Listeria monocytogenes. The results indicate that constitutive expression of IL-7Rα alone was not enough to impart an expansion or survival advantage to CD8 T cells responding to infection, and did not increase memory CD8 T cell numbers over those observed in wild-type controls. Constitutive expression of IL-7Rα did allow for slightly prolonged expansion of Ag-specific CD4 T cells; however, it did not alter the contraction phase or protect against the waning of memory T cell numbers at later times after infection. Memory CD4 and CD8 T cells generated in IL-7Rα transgenic mice expanded similarly to wild-type T cells after secondary infection, and immunized IL-7Rα transgenic mice were fully protected against lethal bacterial challenge demonstrating that constitutive expression of IL-7Rα does not impair, or markedly improve memory/secondary effector T cell function. These results indicate that expression of IL-7Rα alone does not support increased survival of effector Ag-specific CD4 or CD8 T cells into the memory phase following bacterial infection.
- Published
- 2008
- Full Text
- View/download PDF
37. β-Catenin is required for the differentiation of iNKT2 and iNKT17 cells that augment IL-25-dependent lung inflammation
- Author
-
Archna Sharma, Farrah C. Steinke, Cheong Hee Chang, Yeung Hyen Kim, Dil Afroz Sultana, Hai-Hui Xue, Nicola M. Heller, Rosa Berga-Bolaños, Kalyani Pyaram, Jessie Fang, and Jyoti Misra Sen
- Subjects
iNKT2 ,iNKT17, IL-17RB ,Cell type ,Cellular differentiation ,Immunology ,Cell ,Mice, Transgenic ,Inflammation ,Biology ,medicine ,Animals ,beta Catenin ,iNKT cells ,Effector ,Interleukin-17 ,Cell Differentiation ,Pneumonia ,Natural killer T cell ,Cell biology ,Mice, Inbred C57BL ,medicine.anatomical_structure ,Beta-catenin ,Catenin ,Natural Killer T-Cells ,Interleukin 17 ,Bronchial Hyperreactivity ,medicine.symptom ,Research Article - Abstract
Background Invariant Natural Killer T (iNKT) cells have been implicated in lung inflammation in humans and also shown to be a key cell type in inducing allergic lung inflammation in mouse models. iNKT cells differentiate and acquire functional characteristics during development in the thymus. However, the correlation between development of iNKT cells in the thymus and role in lung inflammation remains unknown. In addition, transcriptional control of differentiation of iNKT cells into iNKT cell effector subsets in the thymus during development is also unclear. In this report we show that β-catenin dependent mechanisms direct differentiation of iNKT2 and iNKT17 subsets but not iNKT1 cells. Methods To study the role for β-catenin in lung inflammation we utilize mice with conditional deletion and enforced expression of β-catenin in a well-established mouse model for IL-25-dependen lung inflammation. Results Specifically, we demonstrate that conditional deletion of β-catenin permitted development of mature iNKT1 cells while impeding maturation of iNKT2 and 17 cells. A role for β-catenin expression in promoting iNKT2 and iNKT17 subsets was confirmed when we noted that enforced transgenic expression of β-catenin in iNKT cell precursors enhanced the frequency and number of iNKT2 and iNKT17 cells at the cost of iNKT1 cells. This effect of expression of β-catenin in iNKT cell precursors was cell autonomous. Furthermore, iNKT2 cells acquired greater capability to produce type-2 cytokines when β-catenin expression was enhanced. Discussion This report shows that β-catenin deficiency resulted in a profound decrease in iNKT2 and iNKT17 subsets of iNKT cells whereas iNKT1 cells developed normally. By contrast, enforced expression of β-catenin promoted the development of iNKT2 and iNKT17 cells. It was important to note that the majority of iNKT cells in the thymus of C57BL/6 mice were iNKT1 cells and enforced expression of β-catenin altered the pattern to iNKT2 and iNKT17 cells suggesting that β-catenin may be a major factor in the distinct pathways that critically direct differentiation of iNKT effector subsets. Conclusions Thus, we demonstrate that β-catenin expression in iNKT cell precursors promotes differentiation toward iNKT2 and iNKT17 effector subsets and supports enhanced capacity to produce type 2 and 17 cytokines which in turn augment lung inflammation in mice.
- Published
- 2015
- Full Text
- View/download PDF
38. The Timing of Stimulation and IL-2 Signaling Regulate Secondary CD8 T Cell Responses
- Author
-
John T. Harty, Vladimir P. Badovinac, Shaniya H. Khan, Hai-Hui Xue, Gabriel R. Starbeck-Miller, and Matthew D. Martin
- Subjects
Interleukin 2 ,lcsh:Immunologic diseases. Allergy ,Time Factors ,Cellular differentiation ,Immunoblotting ,Immunology ,CD8-Positive T-Lymphocytes ,Lymphocyte Activation ,Polymerase Chain Reaction ,Microbiology ,Mice ,Immune system ,Virology ,Genetics ,medicine ,Animals ,Cytotoxic T cell ,IL-2 receptor ,Molecular Biology ,lcsh:QH301-705.5 ,CD40 ,biology ,Lymphokine ,CD28 ,Cell Differentiation ,Flow Cytometry ,Adoptive Transfer ,Cell biology ,Mice, Inbred C57BL ,lcsh:Biology (General) ,biology.protein ,Interleukin-2 ,Parasitology ,lcsh:RC581-607 ,Immunologic Memory ,Signal Transduction ,Research Article ,medicine.drug - Abstract
Memory CD8 T cells provide protection to immune hosts by eliminating pathogen-infected cells during re-infection. While parameters influencing the generation of primary (1°) CD8 T cells are well established, the factors controlling the development of secondary (2°) CD8 T cell responses remain largely unknown. Here, we address the mechanisms involved in the generation and development of 2° memory (M) CD8 T cells. We observed that the time at which 1° M CD8 T cells enter into immune response impacts their fate and differentiation into 2° M CD8 T cells. Late-entry of 1° M CD8 T cells into an immune response (relative to the onset of infection) not only facilitated the expression of transcription factors associated with memory formation in 2° effector CD8 T cells, but also influenced the ability of 2° M CD8 T cells to localize within the lymph nodes, produce IL-2, and undergo Ag-driven proliferation. The timing of stimulation of 1° M CD8 T cells also impacted the duration of expression of the high-affinity IL-2 receptor (CD25) on 2° effector CD8 T cells and their sensitivity to IL-2 signaling. Importantly, by blocking or enhancing IL-2 signaling in developing 2° CD8 T cells, we provide direct evidence for the role of IL-2 in controlling the differentiation of Ag-driven 2° CD8 T cell responses. Thus, our data suggest that the process of 1° M to 2° M CD8 T cell differentiation is not fixed and can be manipulated, a notion with relevance for the design of future prime-boost vaccination approaches., Author Summary Since memory CD8 T cells afford hosts increased protection, extensive research has been devoted to understanding the parameters that affect the generation of these cells. Humans are typically infected with pathogens more than once, thus leading to re-stimulation of existing primary memory CD8 T cell populations. The factors influencing the development of CD8 T cells responding to repetitive antigen stimulations remain unknown. We demonstrate that the time at which primary memory CD8 T cells encounter antigen and are re-stimulated during infection influences the outcome of a secondary pathogen-specific CD8 T cell response. We show that at the time of antigen re-encounter, interleukin-2 cytokine signals received by developing secondary CD8 T cells impact the rate of acquiring secondary memory CD8 T cell characteristics. These data indicate that secondary memory CD8 T cell generation is a process that can be manipulated, which may have implications in the development of consecutive prime-boost immunization strategies.
- Published
- 2015
39. Cell-autonomous requirement for TCF1 and LEF1 in the development of Natural Killer T cells
- Author
-
Hai-Hui Xue, Rosa Berga-Bolaños, Wandi S. Zhu, Jyoti Misra Sen, and Farrah C. Steinke
- Subjects
Lymphoid Enhancer-Binding Factor 1 ,Cellular differentiation ,Immunology ,CD1 ,chemical and pharmacologic phenomena ,Biology ,Article ,Mice ,Cell autonomous ,Animals ,Hepatocyte Nuclear Factor 1-alpha ,Molecular Biology ,Transcription factor ,Mice, Knockout ,Thymocytes ,hemic and immune systems ,Cell Differentiation ,Natural killer T cell ,Cell biology ,Mice, Inbred C57BL ,Thymocyte ,embryonic structures ,Natural Killer T-Cells ,CD8 ,Lymphoid enhancer-binding factor 1 - Abstract
Natural killer T (NKT) cells develop from common CD4(+) CD8(+) thymocyte precursors. Transcriptional programs that regulate the development of NKT cells in the thymus development remain to be fully delineated. Here, we demonstrate a cell-intrinsic requirement for transcription factors TCF1 and LEF1 for the development of all subsets of NKT cells. Conditional deletion of TCF1 alone results in a substantial reduction in NKT cells. The remaining NKT cells are eliminated when TCF1 and LEF1 are both deleted. These data reveal an essential role for TCF1 and LEF1 in development of NKT cells.
- Published
- 2015
40. LEF-1 and TCF-1 orchestrate T(FH) differentiation by regulating differentiation circuits upstream of the transcriptional repressor Bcl6
- Author
-
Jodi A. Gullicksrud, Zhouhao Zeng, Shaojun Xing, Paul E. Love, Weiqun Peng, Shane Crotty, Youn Soo Choi, Qiang Shan, Fengyin Li, and Hai-Hui Xue
- Subjects
B cell lymphoma 6 (Bcl6) ,medicine.medical_specialty ,Blimp1 ,Lymphoid Enhancer-Binding Factor 1 ,Cellular differentiation ,T cell factor (TCF)-1 ,Immunology ,IL-6 signal transducer (gp130) ,Biology ,Article ,Mice ,Internal medicine ,medicine ,Cytokine Receptor gp130 ,Immunology and Allergy ,Animals ,Hepatocyte Nuclear Factor 1-alpha ,inducible costimulator (ICOS) ,Transcription factor ,Regulation of gene expression ,B-Lymphocytes ,T follicular helper cell differentiation ,Gene Expression Profiling ,Lymphocyte differentiation ,Germinal center ,Cell Differentiation ,T-Lymphocytes, Helper-Inducer ,BCL6 ,Germinal Center ,Receptors, Interleukin-6 ,Cell biology ,DNA-Binding Proteins ,Endocrinology ,Lymphoid enhancer factor (LEF)-1 ,Gene Expression Regulation ,IL-6 receptor alpha (IL-6Rα) ,embryonic structures ,Proto-Oncogene Proteins c-bcl-6 ,Prdm1 ,Lymphoid enhancer-binding factor 1 - Abstract
Follicular helper T cells (T(FH) cells) are specialized effector CD4(+) T cells that help B cells develop germinal centers (GCs) and memory. However, the transcription factors that regulate the differentiation of T(FH) cells remain incompletely understood. Here we report that selective loss of Lef1 or Tcf7 (which encode the transcription factor LEF-1 or TCF-1, respectively) resulted in T(FH) cell defects, while deletion of both Lef1 and Tcf7 severely impaired the differentiation of T(FH) cells and the formation of GCs. Forced expression of LEF-1 enhanced T(FH) differentiation. LEF-1 and TCF-1 coordinated such differentiation by two general mechanisms. First, they established the responsiveness of naive CD4(+) T cells to T(FH) cell signals. Second, they promoted early T(FH) differentiation via the multipronged approach of sustaining expression of the cytokine receptors IL-6Rα and gp130, enhancing expression of the costimulatory receptor ICOS and promoting expression of the transcriptional repressor Bcl6.
- Published
- 2015
41. Maturation stage–specific regulation of megakaryopoiesis by pointed-domain Ets proteins
- Author
-
Warren J. Leonard, Mortimer Poncz, Gerd A. Blobel, Liyan Pang, Dennis K. Watson, Hai-Hui Xue, Gabor Szalai, Xun Wang, and Yuhuan Wang
- Subjects
Proto-Oncogene Protein c-fli-1 ,Transcription, Genetic ,Immunology ,Mice, Transgenic ,Biology ,Biochemistry ,Proto-Oncogene Protein c-ets-1 ,Mice ,Megakaryocyte ,Chlorocebus aethiops ,Gene expression ,medicine ,Animals ,Gene ,Transcription factor ,Megakaryopoiesis ,Genetics ,Binding Sites ,ETS transcription factor family ,fungi ,Cell Biology ,Hematology ,Hematopoiesis ,Cell biology ,Mice, Inbred C57BL ,medicine.anatomical_structure ,Liver ,Regulatory sequence ,COS Cells ,NIH 3T3 Cells ,Megakaryocytes - Abstract
Numerous megakaryocyte-specific genes contain signature Ets-binding sites in their regulatory regions. Fli-1 (friend leukemia integration 1), an Ets transcription factor, is required for the normal maturation of megakaryocytes and controls the expression of multiple megakaryocyte-specific genes. However, in Fli-1–/– mice, early megakaryopoiesis persists, and the expression of the early megakaryocyte-specific genes, αIIb and cMpl, is maintained, consistent with functional compensation by a related Ets factor(s). Here we identify the Ets protein GABPα (GA-binding protein α) as a regulator of early megakaryocyte-specific genes. Notably, GABPα preferentially occupies Ets elements of early megakaryocyte-specific genes in vitro and in vivo, whereas Fli-1 binds both early and late megakaryocyte-specific genes. Moreover, the ratio of GABPα/Fli-1 expression declines throughout megakaryocyte maturation. Consistent with this expression pattern, primary fetal liver–derived megakaryocytes from Fli-1–deficient murine embryos exhibit reduced expression of genes associated with late stages of maturation (glycoprotein [GP] Ibα, GPIX, and platelet factor 4 [PF4]), whereas GABPα-deficient megakaryocytes were mostly impaired in the expression of early megakaryocyte-specific genes (αIIb and cMpl). Finally, mechanistic experiments revealed that GABPα, like Fli-1, can impart transcriptional synergy between the hematopoietic transcription factor GATA-1 and its cofactor FOG-1 (friend of GATA-1). In concert, these data reveal disparate, but overlapping, functions of Ets transcription factors at distinct stages of megakaryocyte maturation.
- Published
- 2006
- Full Text
- View/download PDF
42. GA binding protein regulates interleukin 7 receptor α-chain gene expression in T cells
- Author
-
J. Philip McCoy, Warren J. Leonard, Radhika Tripuraneni, Valentina Rovella, Yu-Bin Du, Chengyu Liu, Hai-Hui Xue, Ann Williams, and Julie Bollenbacher
- Subjects
T-Lymphocytes ,T cell ,Amino Acid Motifs ,Molecular Sequence Data ,Immunology ,Settore MED/09 ,Biology ,Settore MED/04 ,GA-Binding Protein ,Mice ,Receptors ,medicine ,Animals ,Deoxyribonuclease I ,Humans ,Immunology and Allergy ,Amino Acid Sequence ,Interleukin-7 receptor ,Transcription factor ,B cell ,Interleukin 3 ,Fetus ,Receptors, Interleukin-7 ,Base Sequence ,Interleukin-7 ,Embryo ,Settore MED/16 ,GA-Binding Protein Transcription Factor ,Molecular biology ,DNA-Binding Proteins ,medicine.anatomical_structure ,Gene Expression Regulation ,Transcription Factors - Abstract
The interleukin 7 receptor alpha-chain (IL-7Ralpha) is essential for T cell development in both humans and mice and for B cell development in mice. Whereas the transcription factor PU.1 regulates IL-7Ralpha expression in mouse pro-B cells via a GGAA motif, we demonstrate here that GA binding protein (GABP) bound to this site and was essential in the regulation of IL-7Ralpha expression in T cells, where PU.1 is not expressed. Moreover, IL-7Ralpha expression was diminished substantially in thymocytes but was normal on B220(+) fetal liver cells from mouse embryos with diminished expression of GABPalpha. Thus, GABP is essential for the regulation of IL-7Ralpha expression in T cells, and the differential regulation of IL-7Ralpha in distinct lymphoid lineages is achieved at least in part by differential recruitment of factors to the same GGAA motif.
- Published
- 2004
- Full Text
- View/download PDF
43. Serine phosphorylation of Stat5 proteins in lymphocytes stimulated with IL-2
- Author
-
Warren J. Leonard, Hai-Hui Xue, Donald W. Fink, Christoph W. Turck, Xiaolong Zhang, and Jun Qin
- Subjects
animal structures ,Immunology ,Enzyme Activators ,Mass Spectrometry ,Phosphorylation cascade ,Serine ,Mice ,chemistry.chemical_compound ,STAT5 Transcription Factor ,Animals ,Humans ,Immunology and Allergy ,Protein phosphorylation ,Lymphocytes ,Phosphorylation ,STAT5 ,Serine/threonine-specific protein kinase ,biology ,Tumor Suppressor Proteins ,food and beverages ,Tyrosine phosphorylation ,General Medicine ,Milk Proteins ,Precipitin Tests ,Molecular biology ,DNA-Binding Proteins ,chemistry ,Mutagenesis, Site-Directed ,Trans-Activators ,biology.protein ,Interleukin-2 ,Signal transduction ,Phosphorus Radioisotopes - Abstract
Tyrosine phosphorylation regulates cytokine-induced dimerization of STAT proteins. Serine phosphorylation has also been found to occur in a number of STAT proteins, including Stat1, Sat3, Stat4, Stat5a, Stat5b and Stat6, and was shown to be important for maximal transcriptional activation mediated by Stat1, Stat3 and Stat4, but not for Stat5a or Stat5b. As these latter proteins were studied in transiently transfected COS-7 cells stimulated with prolactin, we sought to further investigate the significance of their serine phosphorylation in a more physiologically based system in response to IL-2. Both Stat5a and Stat5b were rapidly phosphorylated on serine in response to IL-2 and the phosphorylation site in Stat5a was mapped to Ser780, which is not conserved in Stat5b. In vitro studies with reporter constructs, and experiments in which wild-type and mutant Stat5a retroviruses were used to transduce Stat5a-deficient splenocytes revealed that the serine mutant constructs were not diminished in their ability to mediate IL-2 signaling and if anything exhibited augmented proliferative capability. Thus, in contrast to the apparent importance of serine phosphorylation for transcriptional activation by Stat1, Stat3 and Stat4 in response to IFN, IL-6 and IL-12 respectively, serine phosphorylation of Stat5a does not enhance Stat5a-mediated signaling in response to IL-2.
- Published
- 2002
- Full Text
- View/download PDF
44. Polymicrobial sepsis impairs bystander recruitment of effector cells to infected skin despite optimal sensing and alarming function of skin resident memory CD8 T cells
- Author
-
Isaac J. Jensen, John T. Harty, Vladimir P. Badovinac, Stacey M. Hartwig, Hai-Hui Xue, Qiang Shan, Derek B. Danahy, Scott M. Anthony, and Thomas S. Griffith
- Subjects
0301 basic medicine ,Otology ,Ear Infections ,CD8-Positive T-Lymphocytes ,Pathology and Laboratory Medicine ,Memory T cells ,0302 clinical medicine ,Cellular types ,Cytotoxic T cell ,Biology (General) ,Skin ,Effector ,Immune cells ,3. Good health ,Infectious Diseases ,Cytokines ,White blood cells ,Anatomy ,Research Article ,Skin Infections ,Cell biology ,Blood cells ,QH301-705.5 ,Secondary infection ,Immunology ,Ear infection ,T cells ,Vaccinia virus ,Cytotoxic T cells ,Surgical and Invasive Medical Procedures ,Dermatology ,Biology ,Microbiology ,Sepsis ,Interferon-gamma ,03 medical and health sciences ,Signs and Symptoms ,Immune system ,Antigen ,Diagnostic Medicine ,Virology ,Genetics ,medicine ,Animals ,Antigens ,Molecular Biology ,Medicine and health sciences ,Biology and life sciences ,RC581-607 ,medicine.disease ,030104 developmental biology ,Animal cells ,Otorhinolaryngology ,Ears ,Parasitology ,Immunologic diseases. Allergy ,Cytokine storm ,Immunologic Memory ,Head ,030215 immunology - Abstract
Sepsis is a systemic infection that enhances host vulnerability to secondary infections normally controlled by T cells. Using CLP sepsis model, we observed that sepsis induces apoptosis of circulating memory CD8 T-cells (TCIRCM) and diminishes their effector functions, leading to impaired CD8 T-cell mediated protection to systemic pathogen re-infection. In the context of localized re-infections, tissue resident memory CD8 T-cells (TRM) provide robust protection in a variety of infectious models. TRM rapidly ‘sense’ infection in non-lymphoid tissues and ‘alarm’ the host by enhancing immune cell recruitment to the site of the infection to accelerate pathogen clearance. Here, we show that compared to pathogen-specific TCIRCM, sepsis does not invoke significant numerical decline of Vaccinia virus induced skin-TRM keeping their effector functions (e.g., Ag-dependent IFN-γ production) intact. IFN-γ-mediated recruitment of immune cells to the site of localized infection was, however, reduced in CLP hosts despite TRM maintaining their ‘sensing and alarming’ functions. The capacity of memory CD8 T-cells in the septic environment to respond to inflammatory cues and arrive to the site of secondary infection/antigen exposure remained normal suggesting T-cell-extrinsic factors contributed to the observed lesion. Mechanistically, we showed that IFN-γ produced rapidly during sepsis-induced cytokine storm leads to reduced IFN-γR1 expression on vascular endothelium. As a consequence, decreased expression of adhesion molecules and/or chemokines (VCAM1 and CXCL9) on skin endothelial cells in response to TRM-derived IFN-γ was observed, leading to sub-optimal bystander-recruitment of effector cells and increased susceptibility to pathogen re-encounter. Importantly, as visualized by intravital 2-photon microscopy, exogenous administration of CXCL9/10 was sufficient to correct sepsis-induced impairments in recruitment of effector cells at the localized site of TRM antigen recognition. Thus, sepsis has the capacity to alter skin TRM anamnestic responses without directly impacting TRM number and/or function, an observation that helps to further define the immunoparalysis phase in sepsis survivors., Author summary Infectious pathogens are generally relegated within barrier tissues; however, when infections enter the bloodstream the host enters a septic state that (when severe enough) can lead to widespread tissue damage and death. After resolution of acute stage of sepsis, patients often display enhanced susceptibility to secondary infections resulting from quantitative and qualitative alterations in the immune response. Previously, we and others have shown that sepsis dramatically reduces the number and function of memory CD8 T cells within the host, contributing to the state of immunoparalysis early after sepsis induction. The present study directly examines the impact of sepsis on tissue resident memory CD8 T cells (TRM) that are restricted to barrier tissues and provide protection to localized infections. In contrast to circulating memory T cells found within lymphoid tissues, the number and function of TRM within non-lymphoid peripheral tissue (such as the skin) was maintained during sepsis suggesting a protective niche for memory CD8 T cells within barrier tissues of septic hosts. However, the capacity of TRM to provide protection upon re-infection was severely diminished in septic hosts, which was attributed to a sepsis-induced lesion in TRM-extrinsic factors. Thus, this report supports the notion that sepsis has the capacity to influence host response to pathogen re-infection either by directly influencing memory CD8 T cell populations or by preventing other cell types to properly recognize localized pathogen-induced alarming signals delivered by resident memory CD8 T cells.
- Published
- 2017
- Full Text
- View/download PDF
45. Prostaglandin E1 and Its Analog Misoprostol Inhibit Human CML Stem Cell Self-Renewal via EP4 Receptor Activation and Repression of AP-1
- Author
-
Bing He, Chen Zhao, Fengyin Li, Monica L. Guzman, Rupali R. Bhave, Xiaoke Ma, Steven R. Lentz, Hai-Hui Xue, Shuyang Yu, and Kai Tan
- Subjects
0301 basic medicine ,Agonist ,Transcription, Genetic ,medicine.drug_class ,CD34 ,EP4 Receptor ,Biology ,Article ,Mice ,03 medical and health sciences ,chemistry.chemical_compound ,Cell Line, Tumor ,Leukemia, Myelogenous, Chronic, BCR-ABL Positive ,hemic and lymphatic diseases ,Genetics ,medicine ,Animals ,Humans ,Alprostadil ,Cell Self Renewal ,Progenitor cell ,Prostaglandin E1 ,neoplasms ,beta Catenin ,Drug Synergism ,Cell Biology ,Hematopoietic Stem Cells ,medicine.disease ,Xenograft Model Antitumor Assays ,Transcription Factor AP-1 ,030104 developmental biology ,chemistry ,Immunology ,Imatinib Mesylate ,Neoplastic Stem Cells ,Cancer research ,Molecular Medicine ,lipids (amino acids, peptides, and proteins) ,Stem cell ,Proto-Oncogene Proteins c-fos ,Receptors, Prostaglandin E, EP4 Subtype ,Misoprostol ,FOSB ,Chronic myelogenous leukemia - Abstract
Summary Effective treatment of chronic myelogenous leukemia (CML) largely depends on the eradication of CML leukemic stem cells (LSCs). We recently showed that CML LSCs depend on Tcf1 and Lef1 factors for self-renewal. Using a connectivity map, we identified prostaglandin E1 (PGE1) as a small molecule that partly elicited the gene expression changes in LSCs caused by Tcf1/Lef1 deficiency. Although it has little impact on normal hematopoiesis, we found that PGE1 treatment impaired the persistence and activity of LSCs in a pre-clinical murine CML model and a xenograft model of transplanted CML patient CD34 + stem/progenitor cells. Mechanistically, PGE1 acted on the EP4 receptor and repressed Fosb and Fos AP-1 factors in a β-catenin-independent manner. Misoprostol, an FDA-approved EP4 agonist, conferred similar protection against CML. These findings suggest that activation of this PGE1-EP4 pathway specifically targets CML LSCs and that the combination of PGE1/misoprostol with conventional tyrosine-kinase inhibitors could provide effective therapy for CML.
- Published
- 2017
- Full Text
- View/download PDF
46. From inception to output, Tcf1 and Lef1 safeguard development of T cells and innate immune cells
- Author
-
Farrah C. Steinke and Hai-Hui Xue
- Subjects
CD4-Positive T-Lymphocytes ,Lymphoid Enhancer-Binding Factor 1 ,T cell ,Immunology ,Portraits as Topic ,Biology ,CD8-Positive T-Lymphocytes ,medicine ,Cytotoxic T cell ,Animals ,Humans ,IL-2 receptor ,Hepatocyte Nuclear Factor 1-alpha ,Antigen-presenting cell ,beta Catenin ,Thymocytes ,ZAP70 ,Innate lymphoid cell ,Core Binding Factor alpha Subunits ,Cell Differentiation ,Immunity, Innate ,Cell biology ,Thymocyte ,medicine.anatomical_structure ,embryonic structures ,CD4 Antigens ,CD8 - 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.
- Published
- 2014
47. IL-12 and type I interferon prolong the division of activated CD8 T cells by maintaining high-affinity IL-2 signaling in vivo
- Author
-
John T. Harty, Gabriel R. Starbeck-Miller, and Hai-Hui Xue
- Subjects
Interleukin 2 ,Cell Survival ,medicine.medical_treatment ,T cell ,Immunology ,Immunoblotting ,Biology ,CD8-Positive T-Lymphocytes ,Lymphocyte Activation ,Article ,TCIRG1 ,Interleukin 21 ,Mice ,medicine ,Immunology and Allergy ,Cytotoxic T cell ,Animals ,IL-2 receptor ,Cell Proliferation ,DNA Primers ,Reverse Transcriptase Polymerase Chain Reaction ,ZAP70 ,Forkhead Box Protein M1 ,Interleukin-2 Receptor alpha Subunit ,Forkhead Transcription Factors ,Microarray Analysis ,Adoptive Transfer ,Interleukin-12 ,3. Good health ,Cell biology ,Mice, Inbred C57BL ,Cytokine ,medicine.anatomical_structure ,Bromodeoxyuridine ,Interferon Type I ,Interleukin-2 ,medicine.drug ,Signal Transduction - Abstract
The signal 3 cytokines interleukin-12 and type I interferon sustain CD8 T cell division by prolonging expression of CD25 in vivo., TCR ligation and co-stimulation induce cellular division; however, optimal accumulation of effector CD8 T cells requires direct inflammatory signaling by signal 3 cytokines, such as IL-12 or type I IFNs. Although in vitro studies suggest that IL-12/type I IFN may enhance T cell survival or early proliferation, the mechanisms underlying optimal accumulation of CD8 T cells in vivo are unknown. In particular, it is unclear if disparate signal 3 cytokines optimize effector CD8 T cell accumulation by the same mechanism and how these inflammatory cytokines, which are transiently produced early after infection, affect T cell accumulation many days later at the peak of the immune response. Here, we show that transient exposure of CD8 T cells to IL-12 or type I IFN does not promote survival or confer an early proliferative advantage in vivo, but rather sustains surface expression of CD25, the high-affinity IL-2 receptor. This prolongs division of CD8 T cells in response to basal IL-2, through activation of the PI3K pathway and expression of FoxM1, a positive regulator of cell cycle progression genes. Thus, signal 3 cytokines use a common pathway to optimize effector CD8 T cell accumulation through a temporally orchestrated sequence of cytokine signals that sustain division rather than survival.
- Published
- 2013
48. Cutting edge: generation of memory precursors and functional memory CD8+ T cells depends on T cell factor-1 and lymphoid enhancer-binding factor-1
- Author
-
Hai-Hui Xue and Xinyuan Zhou
- Subjects
animal structures ,Lymphoid Enhancer-Binding Factor 1 ,T cell ,Immunology ,Mice, Transgenic ,Biology ,CD8-Positive T-Lymphocytes ,Article ,TCIRG1 ,Interleukin 21 ,Mice ,medicine ,T Cell Transcription Factor 1 ,Immunology and Allergy ,Cytotoxic T cell ,Animals ,Humans ,IL-2 receptor ,Hepatocyte Nuclear Factor 1-alpha ,Mice, Knockout ,Precursor Cells, T-Lymphoid ,Cell Differentiation ,Listeria monocytogenes ,Cell biology ,Thymocyte ,medicine.anatomical_structure ,embryonic structures ,Immunologic Memory ,CD8 ,Lymphoid enhancer-binding factor 1 - Abstract
T cell factor (TCF)-1 and lymphoid enhancer-binding factor (LEF)-1 transcription factors have redundant roles in promoting thymocyte maturation. TCF-1 has been recently shown to critically regulate memory CD8+ T cell differentiation and persistence. The complete spectra of regulatory roles for TCF-1 and LEF-1 in CD8+ T cell responses are yet unknown. We conditionally targeted LEF-1, and by combination with germline deletion of TCF-1, we found that loss of both factors completely abrogated the generation of KLR G1loIL-7Rα+ memory precursors in effector CD8+ T cell populations in response to Listeria monocytogenes infection. Whereas CD8+ effectors deficient for TCF-1 and LEF-1 retained the capacity to express IFN-γ, granzyme B, and perforin, they were defective in TNF-α production. In the memory phase, the Ag-specific CD8+ T cells lacking TCF-1 and LEF-1 exhibited an effector phenotype and were severely impaired in secondary expansion upon rechallenge. Thus, TCF-1 and LEF-1 cooperatively regulate generation of memory precursors and protective memory CD8+ T cells.
- Published
- 2012
49. GABP controls a critical transcription regulatory module that is essential for maintenance and differentiation of hematopoietic stem/progenitor cells
- Author
-
Keji Zhao, Xuefang Jing, Raja Jothi, Dong-Mei Zhao, Kairong Cui, Shuyang Yu, and Hai-Hui Xue
- Subjects
Mice, 129 Strain ,Hematopoiesis and Stem Cells ,Cellular differentiation ,Immunology ,Kruppel-Like Transcription Factors ,Mice, Transgenic ,Biology ,Biochemistry ,Mice ,Animals ,Humans ,Epigenetics ,Progenitor cell ,Transcription factor ,Genetics ,Mice, Knockout ,Binding Sites ,Proto-Oncogene Proteins c-ets ,Gene Expression Profiling ,Forkhead Box Protein O3 ,PTEN Phosphohydrolase ,Gene Expression Regulation, Developmental ,Cell Differentiation ,Forkhead Transcription Factors ,Cell Biology ,Hematology ,Hematopoietic Stem Cells ,GA-Binding Protein Transcription Factor ,Chromatin ,Cell biology ,Hematopoiesis ,Mice, Inbred C57BL ,Repressor Proteins ,Haematopoiesis ,Proto-Oncogene Proteins c-bcl-2 ,Stem cell ,Genome-Wide Association Study - Abstract
Maintaining a steady pool of self-renewing hematopoietic stem cells (HSCs) is critical for sustained production of multiple blood lineages. Many transcription factors and molecules involved in chromatin and epigenetic modifications have been found to be critical for HSC self-renewal and differentiation; however, their interplay is less understood. The transcription factor GA binding protein (GABP), consisting of DNA-binding subunit GABPα and transactivating subunit GABPβ, is essential for lymphopoiesis as shown in our previous studies. Here we demonstrate cell-intrinsic, absolute dependence on GABPα for maintenance and differentiation of hematopoietic stem/progenitor cells. Through genome-wide mapping of GABPα binding and transcriptomic analysis of GABPα-deficient HSCs, we identified Zfx and Etv6 transcription factors and prosurvival Bcl-2 family members including Bcl-2, Bcl-XL, and Mcl-1 as direct GABP target genes, underlying its pivotal role in HSC survival. GABP also directly regulates Foxo3 and Pten and hence sustains HSC quiescence. Furthermore, GABP activates transcription of DNA methyltransferases and histone acetylases including p300, contributing to regulation of HSC self-renewal and differentiation. These systematic analyses revealed a GABP-controlled gene regulatory module that programs multiple aspects of HSC biology. Our studies thus constitute a critical first step in decoding how transcription factors are orchestrated to regulate maintenance and multipotency of HSCs.
- Published
- 2010
50. Differentiation and persistence of memory CD8(+) T cells depend on T cell factor 1
- Author
-
Dong-Mei Zhao, John T. Harty, Shuyang Yu, Hai-Hui Xue, Xinyuan Zhou, and Vladimir P. Badovinac
- Subjects
Transcription, Genetic ,T cell ,Cellular differentiation ,Immunology ,Biology ,CD8-Positive T-Lymphocytes ,Regulatory Sequences, Nucleic Acid ,Mice ,medicine ,T Cell Transcription Factor 1 ,Immunology and Allergy ,Cytotoxic T cell ,Animals ,Hepatocyte Nuclear Factor 1-alpha ,MOLIMMUNO ,Transcription factor ,beta Catenin ,Interleukin-15 ,Mice, Knockout ,Wnt signaling pathway ,Cell Differentiation ,Molecular biology ,Listeria monocytogenes ,Mice, Inbred C57BL ,Wnt Proteins ,Infectious Diseases ,medicine.anatomical_structure ,Phenotype ,Gene Expression Regulation ,Interleukin 15 ,CELLIMMUNO ,Signal transduction ,T-Box Domain Proteins ,Immunologic Memory ,CD8 ,Protein Binding ,Signal Transduction - Abstract
T cell factor 1 (TCF-1) is a transcription factor known to act downstream of the canonical Wnt pathway and is essential for normal T cell development. However, its physiological roles in mature CD8(+) T cell responses are unknown. Here we showed that TCF-1 deficiency limited proliferation of CD8(+) effector T cells and impaired their differentiation toward a central memory phenotype. Moreover, TCF-1-deficient memory CD8(+) T cells were progressively lost over time, exhibiting reduced expression of the antiapoptotic molecule Bcl-2 and interleukin-2 receptor beta chain and diminished IL-15-driven proliferation. TCF-1 was directly associated with the Eomes allele and the Wnt-TCF-1 pathway was necessary and sufficient for optimal Eomes expression in naive and memory CD8(+) T cells. Importantly, forced expression of Eomes partly protected TCF-1-deficient memory CD8(+) T cells from time-dependent attrition. Our studies thus identify TCF-1 as a critical player in a transcriptional program that regulates memory CD8 differentiation and longevity.
- Published
- 2010
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.