Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 481
Filter
Add more filters

Publication year range
1.
Nat Immunol ; 17(2): 169-78, 2016 Feb.
Article in English | MEDLINE | ID: mdl-26595886

ABSTRACT

The transcription factor GATA-3 is indispensable for the development of all innate lymphoid cells (ILCs) that express the interleukin 7 receptor α-chain (IL-7Rα). However, the function of low GATA-3 expression in committed group 3 ILCs (ILC3 cells) has not been identified. We found that GATA-3 regulated the homeostasis of ILC3 cells by controlling IL-7Rα expression. In addition, GATA-3 served a critical function in the development of the NKp46(+) ILC3 subset by regulating the balance between the transcription factors T-bet and RORγt. Among NKp46(+) ILC3 cells, although GATA-3 positively regulated genes specific to the NKp46(+) ILC3 subset, it negatively regulated genes specific to lymphoid tissue-inducer (LTi) or LTi-like ILC3 cells. Furthermore, GATA-3 was required for IL-22 production in both ILC3 subsets. Thus, despite its low expression, GATA-3 was critical for the homeostasis, development and function of ILC3 subsets.


Subject(s)
Cell Differentiation , GATA3 Transcription Factor/metabolism , Lymphocyte Subsets/cytology , Lymphocyte Subsets/metabolism , Animals , Antigens, Ly/genetics , Antigens, Ly/metabolism , Cell Differentiation/genetics , Cell Differentiation/immunology , Cell Lineage/genetics , Cell Lineage/immunology , Cluster Analysis , GATA3 Transcription Factor/deficiency , GATA3 Transcription Factor/genetics , Gene Expression Profiling , Gene Expression Regulation , Homeostasis , Immunity, Innate/genetics , Immunophenotyping , Interleukins/biosynthesis , Lymphocyte Subsets/immunology , Mice , Mice, Knockout , Mice, Transgenic , Natural Cytotoxicity Triggering Receptor 1/genetics , Natural Cytotoxicity Triggering Receptor 1/metabolism , Nuclear Receptor Subfamily 1, Group F, Member 3/genetics , Nuclear Receptor Subfamily 1, Group F, Member 3/metabolism , Phenotype , Protein Binding , Receptors, Interleukin-7/genetics , Receptors, Interleukin-7/metabolism , T-Box Domain Proteins/metabolism , Interleukin-22
2.
Immunity ; 47(1): 171-182.e4, 2017 07 18.
Article in English | MEDLINE | ID: mdl-28723549

ABSTRACT

Interleukin-7 (IL-7) availability determines the size and proliferative state of the resting T cell pool. However, the mechanisms that regulate steady-state IL-7 amounts are unclear. Using experimental lymphopenic mouse models and IL-7-induced homeostatic proliferation to measure IL-7 availability in vivo, we found that radioresistant cells were the source of IL-7 for both CD4+ and CD8+ T cells. Hematopoietic lineage cells, although irrelevant as a source of IL-7, were primarily responsible for limiting IL-7 availability via their expression of IL-7R. Unexpectedly, innate lymphoid cells were found to have a potent influence on IL-7 amounts in the primary and secondary lymphoid tissues. These results demonstrate that IL-7 homeostasis is achieved through consumption by multiple subsets of innate and adaptive immune cells.


Subject(s)
Hematopoietic Stem Cells/physiology , Interleukin-7/metabolism , Lymphocytes/immunology , Lymphopenia/immunology , T-Lymphocytes/physiology , Adaptive Immunity , Animals , Cell Proliferation , Cells, Cultured , Disease Models, Animal , Homeostasis , Humans , Immunity, Innate , Interleukin-7/genetics , Interleukin-7/immunology , Mice , Mice, Inbred C57BL , Mice, Knockout , Radiation Tolerance , Receptors, Interleukin-7/genetics , Receptors, Interleukin-7/metabolism
3.
EMBO Rep ; 25(8): 3601-3626, 2024 Aug.
Article in English | MEDLINE | ID: mdl-38956225

ABSTRACT

Signals emanating from the T-cell receptor (TCR), co-stimulatory receptors, and cytokine receptors each influence CD8 T-cell fate. Understanding how these signals respond to homeostatic and microenvironmental cues can reveal new ways to therapeutically direct T-cell function. Through forward genetic screening in mice, we discover that loss-of-function mutations in LDL receptor-related protein 10 (Lrp10) cause naive and central memory CD8 T cells to accumulate in peripheral lymphoid organs. Lrp10 encodes a conserved cell surface protein of unknown immunological function. T-cell activation induces Lrp10 expression, which post-translationally suppresses IL7 receptor (IL7R) levels. Accordingly, Lrp10 deletion enhances T-cell homeostatic expansion through IL7R signaling. Lrp10-deficient mice are also intrinsically resistant to syngeneic tumors. This phenotype depends on dense tumor infiltration of CD8 T cells, which display increased memory cell characteristics, reduced terminal exhaustion, and augmented responses to immune checkpoint inhibition. Here, we present Lrp10 as a new negative regulator of CD8 T-cell homeostasis and a host factor that controls tumor resistance with implications for immunotherapy.


Subject(s)
CD8-Positive T-Lymphocytes , Homeostasis , Receptors, Interleukin-7 , Animals , CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/metabolism , Mice , Receptors, Interleukin-7/metabolism , Receptors, Interleukin-7/genetics , LDL-Receptor Related Proteins/metabolism , LDL-Receptor Related Proteins/genetics , Signal Transduction , Lymphocyte Activation/immunology , Mice, Knockout , Mice, Inbred C57BL , Immunologic Memory , Neoplasms/immunology , Neoplasms/genetics , Humans
4.
J Immunol ; 213(3): 283-295, 2024 Aug 01.
Article in English | MEDLINE | ID: mdl-39140825

ABSTRACT

The IL-7R regulates the homeostasis, activation, and distribution of T cells in peripheral tissues. Although several transcriptional enhancers that regulate IL-7Rα expression in αß T cells have been identified, enhancers active in γδ T cells remain unknown. In this article, we discovered an evolutionarily conserved noncoding sequence (CNS) in intron 2 of the IL-7Rα-chain (IL-7Rα) locus and named this region CNS9. CNS9 contained a conserved retinoic acid receptor-related orphan receptor (ROR)-responsive element (RORE) and exerted RORγt-dependent enhancer activity in vitro. Mice harboring point mutations in the RORE in CNS9 (CNS9-RORmut) showed reduced IL-7Rα expression in IL-17-producing Vγ4+ γδ T cells. In addition, the cell number and IL-17A production of Vγ4+ γδ T cells were reduced in the adipose tissue of CNS9-RORmut mice. Consistent with the reduction in IL-17A, CNS9-RORmut mice exhibited decreased IL-33 expression in the adipose tissue, resulting in fewer regulatory T cells and glucose intolerance. The CNS9-ROR motif was partially responsible for IL-7Rα expression in RORγt+ regulatory T cells, whereas IL-7Rα expression was unaffected in RORγt-expressing Vγ2+ γδ T cells, Th17 cells, type 3 innate lymphoid cells, and invariant NKT cells. Our results indicate that CNS9 is a RORΕ-dependent, Vγ4+ γδ T cell-specific IL-7Rα enhancer that plays a critical role in adipose tissue homeostasis via regulatory T cells, suggesting that the evolutionarily conserved RORΕ in IL-7Rα intron 2 may influence the incidence of type 2 diabetes.


Subject(s)
Enhancer Elements, Genetic , Introns , Nuclear Receptor Subfamily 1, Group F, Member 3 , Receptors, Antigen, T-Cell, gamma-delta , Animals , Mice , Introns/genetics , Receptors, Antigen, T-Cell, gamma-delta/genetics , Receptors, Antigen, T-Cell, gamma-delta/metabolism , Enhancer Elements, Genetic/genetics , Nuclear Receptor Subfamily 1, Group F, Member 3/genetics , Nuclear Receptor Subfamily 1, Group F, Member 3/metabolism , Glucose/metabolism , Receptors, Interleukin-7/genetics , Receptors, Interleukin-7/metabolism , Mice, Inbred C57BL , Th17 Cells/immunology , Interleukin-17/metabolism , Interleukin-17/genetics , Humans , Adipose Tissue/metabolism , Adipose Tissue/immunology
5.
Development ; 149(8)2022 04 15.
Article in English | MEDLINE | ID: mdl-35072209

ABSTRACT

Tissue-resident lymphoid cells (TLCs) span the spectrum of innate-to-adaptive immune function. Unlike traditional, circulating lymphocytes that are continuously generated from hematopoietic stem cells (HSCs), many TLCs are of fetal origin and poorly generated from adult HSCs. Here, we sought to further understand murine TLC development and the roles of Flk2 and IL7Rα, two cytokine receptors with known function in traditional lymphopoiesis. Using Flk2- and Il7r-Cre lineage tracing, we found that peritoneal B1a cells, splenic marginal zone B (MZB) cells, lung ILC2s and regulatory T cells (Tregs) were highly labeled. Despite high labeling, loss of Flk2 minimally affected the generation of these cells. In contrast, loss of IL7Rα, or combined deletion of Flk2 and IL7Rα, dramatically reduced the number of B1a cells, MZBs, ILC2s and Tregs, both in situ and upon transplantation, indicating an intrinsic and essential role for IL7Rα. Surprisingly, reciprocal transplants of wild-type HSCs showed that an IL7Rα-/- environment selectively impaired reconstitution of TLCs when compared with TLC numbers in situ. Taken together, our data defined Flk2- and IL7Rα-positive TLC differentiation paths, and revealed functional roles of Flk2 and IL7Rα in TLC establishment.


Subject(s)
Hematopoietic Stem Cells/immunology , Lymphopoiesis/genetics , Receptors, Interleukin-7/genetics , fms-Like Tyrosine Kinase 3/genetics , Adaptive Immunity/genetics , Animals , B-Lymphocytes/immunology , Cell Differentiation/genetics , Cell Differentiation/immunology , Cell Lineage/genetics , Cell Lineage/immunology , Gene Expression Regulation, Developmental/genetics , Hematopoietic Stem Cells/cytology , Immunity, Innate/genetics , Lymphocytes/cytology , Lymphocytes/immunology , Lymphoid Tissue/cytology , Lymphoid Tissue/immunology , Lymphopoiesis/immunology , Mice , Organ Specificity/genetics , T-Lymphocytes, Regulatory/immunology
6.
Nat Immunol ; 14(2): 127-35, 2013 Feb.
Article in English | MEDLINE | ID: mdl-23263556

ABSTRACT

Interleukin 7 (IL-7) has a critical role in the development of early CD4(-)CD8(-) double-negative (DN) thymocytes. Although the transcription factor STAT5 is an important component of IL-7 signaling, differences in the phenotypes of mice deficient in STAT5, IL-7, IL-7 receptor alpha (IL-7rα) or the kinase Jak3 suggest the existence of STAT5-independent IL-7 signaling. Here we found that IL-7-Jak3 signals activated the transcription factor NFATc1 in DN thymocytes by phosphorylating Tyr371 in the regulatory region of NFATc1. This NFAT-activation pathway was critical for the survival and development of DN thymocytes, as deficiency in NFATc1 blocked thymocyte development at the DN1 stage, leading to T cell lymphopenia. In addition, our results demonstrated a cooperative function for NFATc1 and STAT5 in guiding thymocyte development in response to IL-7 signals.


Subject(s)
Interleukin-7/genetics , NFATC Transcription Factors/genetics , STAT5 Transcription Factor/genetics , Signal Transduction/immunology , Thymocytes/cytology , Animals , CD4 Antigens/genetics , CD4 Antigens/immunology , CD8 Antigens/genetics , CD8 Antigens/immunology , Cell Differentiation , Gene Expression Regulation , Interleukin-7/immunology , Janus Kinase 3/genetics , Janus Kinase 3/immunology , Lymphopenia/immunology , Lymphopenia/pathology , Mice , Mice, Transgenic , NFATC Transcription Factors/deficiency , Phosphorylation , Promoter Regions, Genetic , Receptors, Interleukin-7/genetics , Receptors, Interleukin-7/immunology , STAT5 Transcription Factor/immunology , Thymocytes/immunology , Transcription, Genetic
7.
Blood ; 142(13): 1113-1130, 2023 09 28.
Article in English | MEDLINE | ID: mdl-37369082

ABSTRACT

Although absence of interleukin-7 (IL-7) signaling completely abrogates T and B lymphopoiesis in mice, patients with severe combined immunodeficiency caused by mutations in the IL-7 receptor α chain (IL-7Rα) still generate peripheral blood B cells. Consequently, human B lymphopoiesis has been thought to be independent of IL-7 signaling. Using flow cytometric analysis and single-cell RNA sequencing of bone marrow samples from healthy controls and patients who are IL-7Rα deficient, in combination with in vitro modeling of human B-cell differentiation, we demonstrate that IL-7R signaling plays a crucial role in human B lymphopoiesis. IL-7 drives proliferation and expansion of early B-cell progenitors but not of pre-BII large cells and has a limited role in the prevention of cell death. Furthermore, IL-7 guides cell fate decisions by enhancing the expression of BACH2, EBF1, and PAX5, which jointly orchestrate the specification and commitment of early B-cell progenitors. In line with this observation, early B-cell progenitors of patients with IL-7Rα deficiency still expressed myeloid-specific genes. Collectively, our results unveil a previously unknown role for IL-7 signaling in promoting the B-lymphoid fate and expanding early human B-cell progenitors while defining important differences between mice and humans. Our results have implications for hematopoietic stem cell transplantation strategies in patients with T- B+ severe combined immunodeficiency and provide insights into the role of IL-7R signaling in leukemogenesis.


Subject(s)
Interleukin-7 , Severe Combined Immunodeficiency , Humans , Animals , Mice , Interleukin-7/metabolism , Receptors, Interleukin-7/genetics , Cell Differentiation , Hematopoiesis
8.
Blood ; 141(14): 1708-1717, 2023 04 06.
Article in English | MEDLINE | ID: mdl-36599086

ABSTRACT

The downstream signaling of the interleukin-7 (IL-7) receptor (IL-7R) plays important physiological and pathological roles, including the differentiation of lymphoid cells and proliferation of acute lymphoblastic leukemia cells. Gain-of-function mutations in the IL-7Rα chain, the specific component of the receptor for IL-7, result in constitutive, IL-7-independent signaling and trigger acute lymphoblastic leukemia. Here, we show that the loss of the phosphoinositide 5-phosphatase INPP5K is associated with increased levels of the INPP5K substrate phosphatidylinositol 4,5-bisphosphate (PtdIns[4,5]P2) and causes an altered dynamic structure of the IL-7 receptor. We discovered that the IL-7Rα chain contains a very conserved positively charged polybasic amino acid sequence in its cytoplasmic juxtamembrane region; this region establish stronger ionic interactions with negatively charged PtdIns(4,5)P2 in the absence of INPP5K, freezing the IL-7Rα chain structure. This dynamic structural alteration causes defects in IL-7R signaling, culminating in decreased expressions of EBF1 and PAX5 transcription factors, in microdomain formation, cytoskeletal reorganization, and bone marrow B-cell differentiation. Similar alterations after the reduced INPP5K expression also affected mutated, constitutively activated IL-7Rα chains that trigger leukemia development, leading to reduced cell proliferation. Altogether, our results indicate that the lipid 5-phosphatase INPP5K hydrolyzes PtdIns(4,5)P2, allowing the requisite conformational changes of the IL-7Rα chain for optimal signaling.


Subject(s)
Interleukin-7 , Precursor Cell Lymphoblastic Leukemia-Lymphoma , Humans , Interleukin-7/genetics , Interleukin-7/metabolism , Phosphatidylinositol 4,5-Diphosphate , Receptors, Interleukin-7/genetics , Receptors, Interleukin-7/metabolism , Signal Transduction/genetics
9.
Blood ; 142(2): 158-171, 2023 07 13.
Article in English | MEDLINE | ID: mdl-37023368

ABSTRACT

T-cell acute lymphoblastic leukemia (T-ALL) is an aggressive hematological malignancy with a dismal prognosis related to refractory/relapsing diseases, raising the need for new targeted therapies. Activating mutations of interleukin-7-receptor pathway genes (IL-7Rp) play a proven leukemia-supportive role in T-ALL. JAK inhibitors, such as ruxolitinib, have recently demonstrated preclinical efficacy. However, prediction markers for sensitivity to JAK inhibitors are still lacking. Herein, we show that IL-7R (CD127) expression is more frequent (∼70%) than IL-7Rp mutations in T-ALL (∼30%). We compared the so-called nonexpressers (no IL-7R expression/IL-7Rp mutation), expressers (IL7R expression without IL-7Rp mutation), and mutants (IL-7Rp mutations). Integrative multiomics analysis outlined IL-7R deregulation in virtually all T-ALL subtypes, at the epigenetic level in nonexpressers, genetic level in mutants, and posttranscriptional level in expressers. Ex vivo data using primary-derived xenografts support that IL-7Rp is functional whenever the IL-7R is expressed, regardless of the IL-7Rp mutational status. Consequently, ruxolitinib impaired T-ALL survival in both expressers and mutants. Interestingly, we show that expressers displayed ectopic IL-7R expression and IL-7Rp addiction conferring a deeper sensitivity to ruxolitinib. Conversely, mutants were more sensitive to venetoclax than expressers. Overall, the combination of ruxolitinib and venetoclax resulted in synergistic effects in both groups. We illustrate the clinical relevance of this association by reporting the achievement of complete remission in 2 patients with refractory/relapsed T-ALL. This provides proof of concept for translation of this strategy into clinics as a bridge-to-transplantation therapy. IL7R expression can be used as a biomarker for sensitivity to JAK inhibition, thereby expanding the fraction of patients with T-ALL eligible for ruxolitinib up to nearly ∼70% of T-ALL cases.


Subject(s)
Janus Kinase Inhibitors , Precursor T-Cell Lymphoblastic Leukemia-Lymphoma , Humans , Receptors, Interleukin-7/genetics , Receptors, Interleukin-7/metabolism , Precursor T-Cell Lymphoblastic Leukemia-Lymphoma/drug therapy , Precursor T-Cell Lymphoblastic Leukemia-Lymphoma/genetics , Precursor T-Cell Lymphoblastic Leukemia-Lymphoma/metabolism , Janus Kinase Inhibitors/therapeutic use , T-Lymphocytes/pathology
10.
Immunology ; 173(1): 93-105, 2024 Sep.
Article in English | MEDLINE | ID: mdl-38778445

ABSTRACT

Cytokines of the common-γ receptor chain (γc) family are crucial for T-cell differentiation and dysregulation of γc cytokine pathways is involved in the pathogenesis of autoimmune diseases. There is increasing evidence that the availability of the γc receptor (CD132) for the associated receptor chains has implications for T-cell functions. Here we studied the influence of differential γc expression on the expression of the IL-2Rα (CD25), the IL-7Rα (CD127) and the differentiation of activated naïve T cells. We fine-tuned the regulation of γc expression in human primary naïve T cells by lentiviral transduction using small hairpin (sh)RNAs and γc cDNA. Differential γc levels were then analysed for effects on T-cell phenotype and function after activation. Differential γc expression markedly affected IL-2Rα and IL-7Rα expression on activated naïve T cells. High γc expression (γc-high) induced significantly higher expression of IL-2Rα and re-expression of IL-7Rα after activation. Inhibition of γc caused lower IL-2Rα/IL-7Rα expression and impaired proliferation of activated naïve T cells. In contrast, γc-high T cells secreted significantly higher concentrations of effector cytokines (i.e., IFN-γ, IL-6) and showed higher cytokine-receptor induced STAT5 phosphorylation during initial stages as well as persistently higher pSTAT1 and pSTAT3 levels after activation. Finally, accelerated transition towards a CD45RO expressing effector/memory phenotype was seen especially for CD4+ γc-high naïve T cells. These results suggested that high expression of γc promotes expression of IL-2Rα and IL-7Rα on activated naïve T cells with significant effects on differentiation and effector cytokine expression.


Subject(s)
Cell Differentiation , Lymphocyte Activation , Humans , Cell Differentiation/immunology , Interleukin Receptor Common gamma Subunit/genetics , Interleukin Receptor Common gamma Subunit/metabolism , Receptors, Interleukin-7/metabolism , Receptors, Interleukin-7/genetics , Cells, Cultured , Interleukin-2 Receptor alpha Subunit/metabolism , T-Lymphocytes/immunology , T-Lymphocytes/metabolism , Signal Transduction , Phosphorylation , STAT5 Transcription Factor/metabolism , Gene Expression Regulation
11.
Cancer Immunol Immunother ; 73(6): 98, 2024 Apr 15.
Article in English | MEDLINE | ID: mdl-38619641

ABSTRACT

CAR-T-cell therapy has shown promise in treating hematological malignancies but faces challenges in treating solid tumors due to impaired T-cell function in the tumor microenvironment. To provide optimal T-cell activation, we developed a B7 homolog 3 protein (B7H3)-targeting CAR construct consisting of three activation signals: CD3ζ (signal 1), 41BB (signal 2), and the interleukin 7 receptor alpha (IL7Rα) cytoplasmic domain (signal 3). We generated B7H3 CAR-T cells with different lengths of the IL7Rα cytoplasmic domain, including the full length (IL7R-L), intermediate length (IL7R-M), and short length (IL7R-S) domains, and evaluated their functionality in vitro and in vivo. All the B7H3-IL7Rα CAR-T cells exhibited a less differentiated phenotype and effectively eliminated B7H3-positive glioblastoma in vitro. Superiority was found in B7H3 CAR-T cells contained the short length of the IL7Rα cytoplasmic domain. Integration of the IL7R-S cytoplasmic domain maintained pSTAT5 activation and increased T-cell proliferation while reducing activation-induced cell death. Moreover, RNA-sequencing analysis of B7H3-IL7R-S CAR-T cells after coculture with a glioblastoma cell line revealed downregulation of proapoptotic genes and upregulation of genes associated with T-cell proliferation compared with those in 2nd generation B7H3 CAR-T cells. In animal models, compared with conventional CAR-T cells, B7H3-IL7R-S CAR-T cells suppressed tumor growth and prolonged overall survival. Our study demonstrated the therapeutic potential of IL7Rα-incorporating CAR-T cells for glioblastoma treatment, suggesting a promising strategy for augmenting the effectiveness of CAR-T cell therapy.


Subject(s)
Glioblastoma , Receptors, Chimeric Antigen , Animals , Glioblastoma/therapy , Receptors, Chimeric Antigen/genetics , Receptors, Interleukin-7/genetics , Signal Transduction , T-Lymphocytes , Tumor Microenvironment , Humans
12.
RNA ; 28(8): 1058-1073, 2022 08.
Article in English | MEDLINE | ID: mdl-35613883

ABSTRACT

The interleukin 7 receptor (IL7R) is strongly associated with increased risk to develop multiple sclerosis (MS), an autoimmune disease of the central nervous system, and this association is likely driven by up-regulation of the soluble isoform of IL7R (sIL7R). Expression of sIL7R is determined by exclusion of the alternative exon 6 from IL7R transcripts, and our previous work revealed that the MS risk allele of the SNP rs6897932 within this exon enhances the expression of sIL7R by promoting exclusion of exon 6. sIL7R potentiates the activity of IL7, leading to enhanced expansion of T cells and increased disability in the experimental autoimmune encephalomyelitis (EAE) murine model of MS. This role in modulating T cell-driven immunity positions sIL7R as an attractive therapeutic target whose expression could be reduced for treatment of MS or increased for treatment of cancers. In this study, we identified novel antisense oligonucleotides (ASOs) that effectively control the inclusion (anti-sIL7R ASOs) or exclusion (pro-sIL7R ASOs) of this exon in a dose-dependent fashion. These ASOs provided excellent control of exon 6 splicing and sIL7R secretion in human primary CD4+ T cells. Supporting their potential for therapeutic targeting, we showed that lead anti-sIL7R ASOs correct the enhanced exon 6 exclusion imposed by the MS risk allele of rs6897932, whereas lead pro-sIL7R ASOs phenocopy it. The data presented here form the foundation for future preclinical studies that will test the therapeutic potential of these ASOs in MS and immuno-oncology.


Subject(s)
CD4-Positive T-Lymphocytes , Multiple Sclerosis , Receptors, Interleukin-7 , Animals , Exons , Humans , Mice , Multiple Sclerosis/genetics , Oligonucleotides, Antisense/genetics , Oligonucleotides, Antisense/pharmacology , RNA Splicing , Receptors, Interleukin-7/genetics , T-Lymphocytes
13.
J Immunol ; 209(10): 1942-1949, 2022 11 15.
Article in English | MEDLINE | ID: mdl-36426945

ABSTRACT

IL-7 and IL-7R are essential for T lymphocyte differentiation by driving proliferation and survival of specific developmental stages. Although early T lineage progenitors (ETPs), the most immature thymocyte population known, have a history of IL-7R expression, it is unclear whether IL-7R is required at this stage. In this study, we show that mice lacking IL-7 or IL-7R have a marked loss of ETPs that results mostly from a cell-autonomous defect in proliferation and survival, although no changes were detected in Bcl2 protein levels. Furthermore, a fraction of ETPs responded to IL-7 stimulation ex vivo by phosphorylating Stat5, and IL-7R was enriched in the most immature Flt3+Ccr9+ ETPs. Consistently, IL-7 promoted the expansion of Flt3+ but not Flt3- ETPs on OP9-DLL4 cocultures, without affecting differentiation at either stage. Taken together, our data show that IL-7/IL-7R is necessary following thymus seeding by promoting proliferation and survival of the most immature thymocytes.


Subject(s)
Interleukin-7 , Receptors, Interleukin-7 , T-Lymphocytes , Animals , Mice , Cell Differentiation , Receptors, Interleukin-7/genetics , Thymocytes , Thymus Gland , T-Lymphocytes/immunology , Cell Lineage
14.
J Immunol ; 208(5): 1155-1169, 2022 03 01.
Article in English | MEDLINE | ID: mdl-35110421

ABSTRACT

CD8+ T cells are critical for the immune response to pathogens and tumors, and CD8+ T cell memory protects against repeat infections. In this study, we identify the activating transcription factor 7 interacting protein (ATF7ip) as a critical regulator of CD8+ T cell immune responses. Mice with a T cell-specific deletion of ATF7ip have a CD8+ T cell-intrinsic enhancement of Il7r expression and Il2 expression leading to enhanced effector and memory responses. Chromatin immunoprecipitation sequencing studies identified ATF7ip as a repressor of Il7r and Il2 gene expression through the deposition of the repressive histone mark H3K9me3 at the Il7r gene and Il2-Il21 intergenic region. Interestingly, ATF7ip targeted transposable elements for H3K9me3 deposition at both the IL7r locus and the Il2-Il21 intergenic region, indicating that ATF7ip silencing of transposable elements is important for regulating CD8+ T cell function. These results demonstrate a new epigenetic pathway by which IL-7R and IL-2 production are constrained in CD8+ T cells, and this may open up new avenues for modulating their production.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Immunologic Memory/immunology , Interleukin-2/biosynthesis , Receptors, Interleukin-7/biosynthesis , Repressor Proteins/metabolism , Animals , CD4-Positive T-Lymphocytes/immunology , Cells, Cultured , Chromatin Immunoprecipitation , DNA Transposable Elements/genetics , Gene Deletion , Gene Silencing , Histones/genetics , Humans , Interleukin-2/metabolism , Listeria monocytogenes/immunology , Listeriosis/immunology , Listeriosis/microbiology , Mice , Mice, Inbred C57BL , Mice, Knockout , Receptors, Interleukin-7/genetics , Receptors, Interleukin-7/metabolism , Repressor Proteins/genetics
15.
J Immunol ; 208(1): 155-168, 2022 01 01.
Article in English | MEDLINE | ID: mdl-34872976

ABSTRACT

CD8+ memory T (TM) cells play a critical role in immune defense against infection. Two common γ-chain family cytokines, IL-2 and IL-7, although triggering the same mTORC1-S6K pathway, distinctly induce effector T (TE) cells and TM cells, respectively, but the underlying mechanism(s) remains elusive. In this study, we generated IL-7R-/and AMPKα1-knockout (KO)/OTI mice. By using genetic and pharmaceutical tools, we demonstrate that IL-7 deficiency represses expression of FOXO1, TCF1, p-AMPKα1 (T172), and p-ULK1 (S555) and abolishes T cell memory differentiation in IL-7R KO T cells after Listeria monocytogenesis rLmOVA infection. IL-2- and IL-7-stimulated strong and weak S6K (IL-2/S6Kstrong and IL-7/S6Kweak) signals control short-lived IL-7R-CD62L-KLRG1+ TE and long-term IL-7R+CD62L+KLRG1- TM cell formations, respectively. To assess underlying molecular pathway(s), we performed flow cytometry, Western blotting, confocal microscopy, and Seahorse assay analyses by using the IL-7/S6Kweak-stimulated TM (IL-7/TM) and the control IL-2/S6Kstrong-stimulated TE (IL-2/TE) cells. We determine that the IL-7/S6Kweak signal activates transcriptional FOXO1, TCF1, and Id3 and metabolic p-AMPKα1, p-ULK1, and ATG7 molecules in IL-7/TM cells. IL-7/TM cells upregulate IL-7R and CD62L, promote mitochondria biogenesis and fatty acid oxidation metabolism, and show long-term cell survival and functional recall responses. Interestingly, AMPKα1 deficiency abolishes the AMPKα1 but maintains the FOXO1 pathway and induces a metabolic switch from fatty acid oxidation to glycolysis in AMPKα1 KO IL-7/TM cells, leading to loss of cell survival and recall responses. Taken together, our data demonstrate that IL-7-stimulated weak strength of mTORC1-S6K signaling controls T cell memory via activation of transcriptional FOXO1-TCF1-Id3 and metabolic AMPKα1-ULK1-ATG7 pathways. This (to our knowledge) novel finding provides a new mechanism for a distinct IL-2/IL-7 stimulation model in T cell memory and greatly impacts vaccine development.


Subject(s)
AMP-Activated Protein Kinases/metabolism , Autophagy-Related Protein 7/metabolism , Autophagy-Related Protein-1 Homolog/metabolism , CD8-Positive T-Lymphocytes/immunology , Forkhead Box Protein O1/metabolism , Hepatocyte Nuclear Factor 1-alpha/metabolism , Inhibitor of Differentiation Proteins/metabolism , Interleukin-7/metabolism , Listeria monocytogenes/physiology , Listeriosis/immunology , Mechanistic Target of Rapamycin Complex 1/metabolism , Memory T Cells/immunology , Ribosomal Protein S6 Kinases, 90-kDa/metabolism , AMP-Activated Protein Kinases/genetics , Animals , Cell Differentiation , Cell Survival , Cells, Cultured , Cytotoxicity, Immunologic , Fatty Acids/metabolism , Forkhead Box Protein O1/genetics , Gene Expression Regulation , Glycolysis , Hepatocyte Nuclear Factor 1-alpha/genetics , Immunologic Memory , Inhibitor of Differentiation Proteins/genetics , Interleukin-7/genetics , Mice , Mice, Inbred C57BL , Mice, Knockout , Receptors, Interleukin-7/genetics , Signal Transduction , Vaccine Development
16.
Int J Mol Sci ; 25(7)2024 Apr 04.
Article in English | MEDLINE | ID: mdl-38612837

ABSTRACT

Hashimoto's thyroiditis (HT) and Graves' disease (GD) are common autoimmune endocrine disorders in children. Studies indicate that apart from environmental factors, genetic background significantly contributes to the development of these diseases. This study aimed to assess the prevalence of selected single-nucleotide polymorphisms (SNPs) of Il7R, CD226, CAPSL, and CLEC16A genes in children with autoimmune thyroid diseases. We analyzed SNPs at the locus rs3194051, rs6897932 of IL7R, rs763361 of CD226, rs1010601 of CAPSL, and rs725613 of CLEC16A gene in 56 HT patients, 124 GD patients, and 156 healthy children. We observed significant differences in alleles IL7R (rs6897932) between HT males and the control group (C > T, p = 0.028) and between all GD patients and healthy children (C > T, p = 0.035) as well as GD females and controls (C > T, p = 0.018). Moreover, the C/T genotype was less frequent in GD patients at rs6897932 locus and in HT males at rs1010601 locus. The presence of the T allele in the IL7R (rs6897932) locus appears to have a protective effect against HT in males and GD in all children. Similarly, the presence of the T allele in the CAPSL locus (rs1010601) seems to reduce the risk of HT development in all patients.


Subject(s)
Autoimmune Diseases , Graves Disease , Hashimoto Disease , Child , Female , Male , Humans , Adolescent , Prevalence , Alleles , Hashimoto Disease/genetics , Polymorphism, Single Nucleotide , Graves Disease/genetics , Receptors, Interleukin-7/genetics , Monosaccharide Transport Proteins , Lectins, C-Type/genetics
17.
Nat Immunol ; 12(6): 544-50, 2011 Jun.
Article in English | MEDLINE | ID: mdl-21532575

ABSTRACT

The molecular mechanisms that underlie T cell quiescence are poorly understood. Here we report that mature naive CD8(+) T cells lacking the transcription factor Foxp1 gained effector phenotype and function and proliferated directly in response to interleukin 7 (IL-7) in vitro. Foxp1 repressed expression of the IL-7 receptor α-chain (IL-7Rα) by antagonizing Foxo1 and negatively regulated signaling by the kinases MEK and Erk. Acute deletion of Foxp1 induced naive T cells to gain an effector phenotype and proliferate in lympho-replete mice. Foxp1-deficient naive CD8(+) T cells proliferated even in lymphopenic mice deficient in major histocompatibility complex class I. Our results demonstrate that Foxp1 exerts essential cell-intrinsic regulation of naive T cell quiescence, providing direct evidence that lymphocyte quiescence is achieved through actively maintained mechanisms that include transcriptional regulation.


Subject(s)
Cell Proliferation , Forkhead Transcription Factors/immunology , Repressor Proteins/immunology , T-Lymphocytes/immunology , Animals , Butadienes/pharmacology , CD8-Positive T-Lymphocytes/drug effects , CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/metabolism , Cells, Cultured , Enzyme Inhibitors/pharmacology , Extracellular Signal-Regulated MAP Kinases/antagonists & inhibitors , Extracellular Signal-Regulated MAP Kinases/immunology , Extracellular Signal-Regulated MAP Kinases/metabolism , Female , Flow Cytometry , Forkhead Box Protein O1 , Forkhead Transcription Factors/genetics , Forkhead Transcription Factors/metabolism , Gene Expression Regulation/drug effects , Imidazoles/pharmacology , Immunoblotting , Interleukin-7/pharmacology , MAP Kinase Kinase Kinases/antagonists & inhibitors , MAP Kinase Kinase Kinases/immunology , MAP Kinase Kinase Kinases/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Nitriles/pharmacology , Protein Binding , Pyridines/pharmacology , Receptors, Interleukin-7/genetics , Receptors, Interleukin-7/immunology , Receptors, Interleukin-7/metabolism , Repressor Proteins/genetics , Repressor Proteins/metabolism , Reverse Transcriptase Polymerase Chain Reaction , T-Lymphocytes/drug effects , T-Lymphocytes/metabolism
19.
Blood ; 138(12): 1040-1052, 2021 09 23.
Article in English | MEDLINE | ID: mdl-33970999

ABSTRACT

Tight regulation of IL-7Rα expression is essential for normal T-cell development. IL-7Rα gain-of-function mutations are known drivers of T-cell acute lymphoblastic leukemia (T-ALL). Although a subset of patients with T-ALL display high IL7R messenger RNA levels and cases with IL7R gains have been reported, the impact of IL-7Rα overexpression, rather than mutational activation, during leukemogenesis remains unclear. In this study, overexpressed IL-7Rα in tetracycline-inducible Il7r transgenic and Rosa26 IL7R knockin mice drove potential thymocyte self-renewal, and thymus hyperplasia related to increased proliferation of T-cell precursors, which subsequently infiltrated lymph nodes, spleen, and bone marrow, ultimately leading to fatal leukemia. The tumors mimicked key features of human T-ALL, including heterogeneity in immunophenotype and genetic subtype between cases, frequent hyperactivation of the PI3K/Akt pathway paralleled by downregulation of p27Kip1 and upregulation of Bcl-2, and gene expression signatures evidencing activation of JAK/STAT, PI3K/Akt/mTOR and Notch signaling. Notably, we also found that established tumors may no longer require high levels of IL-7R expression upon secondary transplantation and progressed in the absence of IL-7, but remain sensitive to inhibitors of IL-7R-mediated signaling ruxolitinib (Jak1), AZD1208 (Pim), dactolisib (PI3K/mTOR), palbociclib (Cdk4/6), and venetoclax (Bcl-2). The relevance of these findings for human disease are highlighted by the fact that samples from patients with T-ALL with high wild-type IL7R expression display a transcriptional signature resembling that of IL-7-stimulated pro-T cells and, critically, of IL7R-mutant cases of T-ALL. Overall, our study demonstrates that high expression of IL-7Rα can promote T-cell tumorigenesis, even in the absence of IL-7Rα mutational activation.


Subject(s)
Carcinogenesis , Gene Expression Regulation, Leukemic , Mutation , Neoplasm Proteins , Neoplasms, Experimental , Precursor T-Cell Lymphoblastic Leukemia-Lymphoma , Receptors, Interleukin-7 , Animals , Carcinogenesis/genetics , Carcinogenesis/metabolism , Humans , Mice , Mice, Transgenic , Neoplasm Proteins/biosynthesis , Neoplasm Proteins/genetics , Neoplasms, Experimental/genetics , Neoplasms, Experimental/metabolism , Precursor T-Cell Lymphoblastic Leukemia-Lymphoma/genetics , Precursor T-Cell Lymphoblastic Leukemia-Lymphoma/metabolism , Receptors, Interleukin-7/biosynthesis , Receptors, Interleukin-7/genetics , Signal Transduction , Thymocytes/metabolism
20.
Immunity ; 40(3): 378-88, 2014 Mar 20.
Article in English | MEDLINE | ID: mdl-24631153

ABSTRACT

Innate lymphoid cells (ILCs) are critical in innate immune responses to pathogens and lymphoid organ development. Similar to CD4(+) T helper (Th) cell subsets, ILC subsets positive for interleukin-7 receptor α (IL-7Rα) produce distinct sets of effector cytokines. However, the molecular control of IL-7Rα(+) ILC development and maintenance is unclear. Here, we report that GATA3 was indispensable for the development of all IL-7Rα(+) ILC subsets and T cells but was not required for the development of classical natural killer cells. Conditionally Gata3-deficient mice had no lymph nodes and were susceptible to Citrobactor rodentium infection. After the ILCs had fully developed, GATA3 remained important for the maintenance and functions of ILC2s. Genome-wide gene expression analyses indicated that GATA3 regulated a similar set of cytokines and receptors in Th2 cells and ILC2s, but not in ILC3s. Thus, GATA3 plays parallel roles in regulating the development and functions of CD4(+) T cells and IL-7Rα(+) ILCs.


Subject(s)
GATA3 Transcription Factor/metabolism , Gene Expression Regulation , Immunity, Innate/genetics , Lymphocyte Subsets/metabolism , Receptors, Interleukin-7/genetics , Animals , Citrobacter rodentium/immunology , Enterobacteriaceae Infections/genetics , Enterobacteriaceae Infections/immunology , GATA3 Transcription Factor/genetics , Genetic Predisposition to Disease , Lymphocyte Subsets/immunology , Mice , Mice, Knockout , Models, Immunological , Proto-Oncogene Proteins c-vav/genetics , Proto-Oncogene Proteins c-vav/metabolism , Receptors, Interleukin-7/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL