Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 201
Filter
Add more filters

Country/Region as subject
Publication year range
1.
Mol Cell ; 81(7): 1469-1483.e8, 2021 04 01.
Article in English | MEDLINE | ID: mdl-33609448

ABSTRACT

We demonstrate that DNA hypomethylating agent (HMA) treatment can directly modulate the anti-tumor response and effector function of CD8+ T cells. In vivo HMA treatment promotes CD8+ T cell tumor infiltration and suppresses tumor growth via CD8+ T cell-dependent activity. Ex vivo, HMAs enhance primary human CD8+ T cell activation markers, effector cytokine production, and anti-tumor cytolytic activity. Epigenomic and transcriptomic profiling shows that HMAs vastly regulate T cell activation-related transcriptional networks, culminating with over-activation of NFATc1 short isoforms. Mechanistically, demethylation of an intragenic CpG island immediately downstream to the 3' UTR of the short isoform was associated with antisense transcription and alternative polyadenylation of NFATc1 short isoforms. High-dimensional single-cell mass cytometry analyses reveal a selective effect of HMAs on a subset of human CD8+ T cell subpopulations, increasing both the number and abundance of a granzyme Bhigh, perforinhigh effector subpopulation. Overall, our findings support the use of HMAs as a therapeutic strategy to boost anti-tumor immune response.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , CpG Islands/immunology , DNA Methylation/drug effects , Decitabine/pharmacology , Granzymes/immunology , Lymphocyte Activation/drug effects , DNA Methylation/immunology , Humans , NFATC Transcription Factors/immunology , Perforin/immunology
2.
Nat Immunol ; 16(1): 118-28, 2015 Jan.
Article in English | MEDLINE | ID: mdl-25401424

ABSTRACT

The recognition of microbial patterns by Toll-like receptors (TLRs) is critical for activation of the innate immune system. Although TLRs are expressed by human CD4(+) T cells, their function is not well understood. Here we found that engagement of TLR7 in CD4(+) T cells induced intracellular calcium flux with activation of an anergic gene-expression program dependent on the transcription factor NFATc2, as well as unresponsiveness of T cells. As chronic infection with RNA viruses such as human immunodeficiency virus type 1 (HIV-1) induces profound dysfunction of CD4(+) T cells, we investigated the role of TLR7-induced anergy in HIV-1 infection. Silencing of TLR7 markedly decreased the frequency of HIV-1-infected CD4(+) T cells and restored the responsiveness of those HIV-1(+) CD4(+) T cells. Our results elucidate a previously unknown function for microbial pattern-recognition receptors in the downregulation of immune responses.


Subject(s)
CD4-Positive T-Lymphocytes/immunology , HIV Infections/immunology , HIV-1/immunology , Toll-Like Receptor 7/immunology , CD4-Positive T-Lymphocytes/virology , Calcium/immunology , Clonal Anergy/immunology , Down-Regulation , HIV Infections/virology , Humans , Leukocytes, Mononuclear/immunology , Lymphocyte Activation , NFATC Transcription Factors/immunology , RNA, Small Interfering/administration & dosage , RNA, Small Interfering/genetics , Toll-Like Receptor 7/antagonists & inhibitors , Toll-Like Receptor 7/genetics
3.
Immunity ; 47(4): 664-679.e6, 2017 10 17.
Article in English | MEDLINE | ID: mdl-29030115

ABSTRACT

Store-operated Ca2+ entry (SOCE) is the main Ca2+ influx pathway in lymphocytes and is essential for T cell function and adaptive immunity. SOCE is mediated by Ca2+ release-activated Ca2+ (CRAC) channels that are activated by stromal interaction molecule (STIM) 1 and STIM2. SOCE regulates many Ca2+-dependent signaling molecules, including calcineurin, and inhibition of SOCE or calcineurin impairs antigen-dependent T cell proliferation. We here report that SOCE and calcineurin regulate cell cycle entry of quiescent T cells by controlling glycolysis and oxidative phosphorylation. SOCE directs the metabolic reprogramming of naive T cells by regulating the expression of glucose transporters, glycolytic enzymes, and metabolic regulators through the activation of nuclear factor of activated T cells (NFAT) and the PI3K-AKT kinase-mTOR nutrient-sensing pathway. We propose that SOCE controls a critical "metabolic checkpoint" at which T cells assess adequate nutrient supply to support clonal expansion and adaptive immune responses.


Subject(s)
Calcium Channels/immunology , Calcium Signaling/immunology , Calcium/immunology , T-Lymphocytes/immunology , Animals , Calcineurin/immunology , Calcineurin/metabolism , Calcium/metabolism , Calcium Channels/metabolism , Cell Division/immunology , Cells, Cultured , Female , Glycolysis/immunology , HEK293 Cells , Humans , Immunoblotting , Male , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Microscopy, Confocal , NFATC Transcription Factors/genetics , NFATC Transcription Factors/immunology , NFATC Transcription Factors/metabolism , Phosphatidylinositol 3-Kinases/immunology , Phosphatidylinositol 3-Kinases/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Signal Transduction/immunology , Stromal Interaction Molecule 1/genetics , Stromal Interaction Molecule 1/immunology , Stromal Interaction Molecule 1/metabolism , Stromal Interaction Molecule 2/genetics , Stromal Interaction Molecule 2/immunology , Stromal Interaction Molecule 2/metabolism , T-Lymphocytes/metabolism
4.
Immunity ; 47(6): 1129-1141.e5, 2017 12 19.
Article in English | MEDLINE | ID: mdl-29246443

ABSTRACT

During chronic stimulation, CD8+ T cells acquire an exhausted phenotype characterized by expression of inhibitory receptors, down-modulation of effector function, and metabolic impairments. T cell exhaustion protects from excessive immunopathology but limits clearance of virus-infected or tumor cells. We transcriptionally profiled antigen-specific T cells from mice infected with lymphocytic choriomeningitis virus strains that cause acute or chronic disease. T cell exhaustion during chronic infection was driven by high amounts of T cell receptor (TCR)-induced transcription factors IRF4, BATF, and NFATc1. These regulators promoted expression of inhibitory receptors, including PD-1, and mediated impaired cellular metabolism. Furthermore, they repressed the expression of TCF1, a transcription factor required for memory T cell differentiation. Reducing IRF4 expression restored the functional and metabolic properties of antigen-specific T cells and promoted memory-like T cell development. These findings indicate that IRF4 functions as a central node in a TCR-responsive transcriptional circuit that establishes and sustains T cell exhaustion during chronic infection.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Immunologic Memory , Interferon Regulatory Factors/immunology , Lymphocytic Choriomeningitis/immunology , Lymphocytic choriomeningitis virus/immunology , Receptors, Antigen, T-Cell/immunology , Animals , Basic-Leucine Zipper Transcription Factors/genetics , Basic-Leucine Zipper Transcription Factors/immunology , CD8-Positive T-Lymphocytes/virology , Cell Differentiation , Gene Expression Regulation , HEK293 Cells , Hepatocyte Nuclear Factor 1-alpha/genetics , Hepatocyte Nuclear Factor 1-alpha/immunology , Humans , Interferon Regulatory Factors/deficiency , Interferon Regulatory Factors/genetics , Lymphocyte Activation , Lymphocyte Depletion , Lymphocytic Choriomeningitis/genetics , Lymphocytic Choriomeningitis/virology , Lymphocytic choriomeningitis virus/growth & development , Mice , Mice, Knockout , NFATC Transcription Factors/genetics , NFATC Transcription Factors/immunology , Programmed Cell Death 1 Receptor/genetics , Programmed Cell Death 1 Receptor/immunology , Receptors, Antigen, T-Cell/genetics , Signal Transduction
5.
Immunity ; 38(2): 225-36, 2013 Feb 21.
Article in English | MEDLINE | ID: mdl-23415911

ABSTRACT

It is widely appreciated that T cells increase glycolytic flux during activation, but the role of mitochondrial flux is unclear. Here, we have shown that mitochondrial metabolism in the absence of glucose metabolism is sufficient to support interleukin-2 (IL-2) induction. Furthermore, we used mice with reduced mitochondrial reactive oxygen species (mROS) production in T cells (T-Uqcrfs(-/-) mice) to show that mitochondria are required for T cell activation to produce mROS for activation of nuclear factor of activated T cells (NFAT) and subsequent IL-2 induction. These mice could not induce antigen-specific expansion of T cells in vivo, but Uqcrfs1(-/-) T cells retained the ability to proliferate in vivo under lymphopenic conditions. This suggests that Uqcrfs1(-/-) T cells were not lacking bioenergetically but rather lacked specific ROS-dependent signaling events needed for antigen-specific expansion. Thus, mitochondrial metabolism is a critical component of T cell activation through the production of complex III ROS.


Subject(s)
Mitochondria/metabolism , NFATC Transcription Factors/genetics , T-Lymphocytes/metabolism , Tumor Necrosis Factor Receptor Superfamily, Member 7/genetics , Animals , Cell Proliferation , Electron Transport Complex III/metabolism , Female , Gene Expression , Homeodomain Proteins/genetics , Homeodomain Proteins/immunology , Interleukin-2/biosynthesis , Interleukin-2/immunology , Iron-Sulfur Proteins/deficiency , Iron-Sulfur Proteins/genetics , Lymphocyte Activation , Lymphopenia/immunology , Lymphopenia/metabolism , Mice , Mice, Knockout , Mitochondria/genetics , Mitochondria/immunology , NFATC Transcription Factors/immunology , Reactive Oxygen Species/metabolism , Signal Transduction , T-Lymphocytes/immunology , Tumor Necrosis Factor Receptor Superfamily, Member 7/immunology
6.
Immunity ; 38(2): 237-49, 2013 Feb 21.
Article in English | MEDLINE | ID: mdl-23313588

ABSTRACT

Interactions with antigen-presenting cells (APCs) interrupt T cell migration through tissues and trigger signaling pathways that converge on the activation of transcriptional regulators, including nuclear factor of activated T cells (NFAT), which control T cell function and differentiation. Both stable and unstable modes of cognate T cell-APC interactions have been observed in vivo, but the functional significance of unstable, serial contacts has remained unclear. Here we used multiphoton intravital microscopy in lymph nodes and tumors to show that while NFAT nuclear import was fast (t(1/2 max)∼1 min), nuclear export was slow (t(1/2)∼20 min) in T cells. During delayed export, nuclear NFAT constituted a short-term imprint of transient TCR signals and remained transcriptionally active for the T cell tolerance gene Egr2, but not for the effector gene Ifng, which required continuous TCR triggering for expression. This provides a potential mechanistic basis for the observation that a predominance of unstable APC interactions correlates with the induction of T cell tolerance.


Subject(s)
Antigen-Presenting Cells/metabolism , Immune Tolerance , Immunologic Memory , Lymph Nodes/metabolism , NFATC Transcription Factors/genetics , T-Lymphocytes/metabolism , Animals , Antigen-Presenting Cells/immunology , Antigen-Presenting Cells/pathology , Cell Communication , Cell Differentiation , Cell Movement , Cell Nucleus/metabolism , Cytosol/metabolism , Early Growth Response Protein 2/genetics , Early Growth Response Protein 2/immunology , Gene Expression Regulation , Humans , Interferon-gamma/genetics , Interferon-gamma/immunology , Lymph Nodes/immunology , Lymph Nodes/pathology , Mice , Microscopy, Fluorescence, Multiphoton , NFATC Transcription Factors/immunology , Protein Transport , Receptors, Antigen, T-Cell , Signal Transduction , T-Lymphocytes/immunology , Tumor Cells, Cultured
7.
J Immunol ; 205(12): 3311-3318, 2020 12 15.
Article in English | MEDLINE | ID: mdl-33188077

ABSTRACT

IL-13 plays a critical role in mediating many biological processes responsible for allergic inflammation. Mast cells express Il13 mRNA and produce IL-13 protein in response to antigenic stimulation. Enhancers are essential in promoting gene transcription and are thought to activate transcription by delivering essential accessory cofactors to the promoter to potentiate gene transcription. However, enhancers mediating Il13 have not been identified. Furthermore, which Il13 enhancers detect signals triggered by antigenic stimulation have not yet been defined. In this study, we identified potential mouse Il13 enhancers using histone modification monomethylation at lysine residue 4 on histone 3 (H3K4me1) chromatin immunoprecipitation sequencing and acetylation at lysine residue 27 on histone 3 (H3K27ac) chromatin immunoprecipitation sequencing. We used Omni-assay for transposase-accessible chromatin sequencing to determine which accessible regions within the potential Il13 enhancers that responded to IgE receptor crosslinking. We also demonstrated that the transcription factor cluster consisting of the NFATC2, STAT5, GATA2, AP1, and RUNX1 binding sites at the proximal Il13 enhancer and the transcription factor cluster consisting of the EGR2 binding site at the distal Il13 E+6.5 enhancer are critical in sensing the signals triggered by antigenic stimulation. Those enhancers, which are responsive to antigenic stimulation and are constitutively active, cooperate to generate greater transcriptional outputs. Our study reveals a novel mechanism underlying how antigenic stimulation induces robust Il13 mRNA expression in mouse mast cells.


Subject(s)
Antigens/immunology , Core Binding Factor Alpha 2 Subunit/immunology , Early Growth Response Protein 2/immunology , GATA2 Transcription Factor/immunology , Interleukin-13/immunology , Mast Cells/immunology , NFATC Transcription Factors/immunology , Response Elements/immunology , STAT5 Transcription Factor/immunology , Transcription Factor AP-1/immunology , Transcription, Genetic/immunology , Animals , Cell Line , Mast Cells/cytology , Mice
8.
Nat Immunol ; 10(3): 306-13, 2009 Mar.
Article in English | MEDLINE | ID: mdl-19169262

ABSTRACT

The influence of signals transmitted by the phosphatase calcineurin and the transcription factor NFAT on the development and function of natural killer T (NKT) cells is unclear. In this report, we demonstrate that the transcription factor early growth response 2 (Egr2), a target gene of NFAT, was specifically required for the ontogeny of NKT cells but not that of conventional CD4(+) or CD8(+) T cells. NKT cells developed normally in the absence of Egr1 or Egr3, which suggests that Egr2 is a specific regulator of NKT cell differentiation. We found that Egr2 was important in the selection, survival and maturation of NKT cells. Our findings emphasize the importance of the calcineurin-NFAT-Egr2 pathway in the development of the NKT lymphocyte lineage.


Subject(s)
Cell Differentiation , Early Growth Response Protein 2/metabolism , NFATC Transcription Factors/metabolism , Natural Killer T-Cells/immunology , Animals , CD4-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/immunology , Calcineurin/metabolism , Cells, Cultured , Early Growth Response Protein 2/genetics , Early Growth Response Protein 2/immunology , Humans , Mice , Mice, Inbred C57BL , Mice, Knockout , NFATC Transcription Factors/genetics , NFATC Transcription Factors/immunology
9.
J Immunol ; 202(9): 2616-2627, 2019 05 01.
Article in English | MEDLINE | ID: mdl-30910863

ABSTRACT

Appropriate T cell responses are controlled by strict balance between activatory and inhibitory pathways downstream of TCR. Although mice or humans with impaired TCR signaling develop autoimmunity, the precise molecular mechanisms linking reduced TCR signaling to autoimmunity are not fully understood. Engagement of TCR activates Ca2+ signaling mainly through store-operated Ca2+ entry activated by stromal interaction molecule (Stim) 1 and Stim2. Despite defective T cell activation, mice deficient in both Stim1 and Stim2 in T cells (conditional double knockout [cDKO]) developed lymphoproliferative disorders and skin inflammation with a concomitant increase in serum IgG1 and IgE levels. In cDKO mice, follicular helper T (Tfh) cells were dramatically increased in number, and they produced IL-4 spontaneously. These inflammatory symptoms were abolished by the deletion of IL-4 in cDKO mice. Tfh development and inflammatory symptoms in cDKO mice were abrogated by further deletion of NFAT2 in T cells. These findings suggest that Tfh cells spontaneously developed in the absence of Ca2+ signaling and caused unregulated type 2 responses.


Subject(s)
Immune System Diseases/immunology , Stromal Interaction Molecule 1/deficiency , Stromal Interaction Molecule 2/deficiency , T-Lymphocytes, Helper-Inducer/immunology , Animals , Calcium Signaling/genetics , Calcium Signaling/immunology , Immune System Diseases/genetics , Immune System Diseases/pathology , Immunoglobulin E/genetics , Immunoglobulin E/immunology , Immunoglobulin G/genetics , Immunoglobulin G/immunology , Interleukin-4/genetics , Interleukin-4/immunology , Lymphoproliferative Disorders/genetics , Lymphoproliferative Disorders/immunology , Lymphoproliferative Disorders/pathology , Mice , Mice, Knockout , NFATC Transcription Factors/genetics , NFATC Transcription Factors/immunology , Skin Diseases/genetics , Skin Diseases/immunology , Skin Diseases/pathology , T-Lymphocytes, Helper-Inducer/pathology
10.
Int J Mol Sci ; 22(5)2021 Mar 08.
Article in English | MEDLINE | ID: mdl-33800389

ABSTRACT

Nuclear factor of activated T cells (NFAT), which is the pharmacological target of immunosuppressants cyclosporine and tacrolimus, has been shown to play an important role not only in T cells (immune system), from which their name is derived, but also in many biological events. Therefore, functional and/or structural abnormalities of NFAT are linked to the pathogenesis of diseases in various organs. The NFAT protein family consists of five isoforms, and each isoform performs diverse functions and has unique expression patterns in the target tissues. This diversity has made it difficult to obtain ideal pharmacological output for immunosuppressants that inhibit the activity of almost all NFAT family members, causing serious and wide-ranging side effects. Moreover, it remains unclear whether isoform-selective NFAT regulation can be achieved by targeting the structural differences among NFAT isoforms and whether this strategy can lead to the development of better drugs than the existing ones. This review summarizes the role of the NFAT family members in biological events, including the development of various diseases, as well as the usefulness of and problems associated with NFAT-targeting therapies, including those dependent on current immunosuppressants. Finally, we propose a novel therapeutic strategy based on the molecular mechanisms that enable selective regulation of specific NFAT isoforms.


Subject(s)
Cyclosporine/therapeutic use , Immunosuppressive Agents/therapeutic use , NFATC Transcription Factors/antagonists & inhibitors , NFATC Transcription Factors/immunology , T-Lymphocytes/immunology , Tacrolimus/therapeutic use , Animals , Humans , Protein Isoforms
11.
Br J Haematol ; 191(5): 906-919, 2020 12.
Article in English | MEDLINE | ID: mdl-32729137

ABSTRACT

Severe acquired aplastic anaemia (AA) is a serious disease characterised by autoreactive T cells attacking haematopoietic stem cells, leading to marrow hypoplasia and pancytopenia. Immunosuppressive therapy combined with antithymocyte globulin and ciclosporin can rescue most patients with AA. However, the relapse after ciclosporin withdrawal and the severe side effects of long-term ciclosporin administration remain unresolved. As such, new strategies should be developed to supplement current therapeutics and treat AA. In this study, the possibility of all-trans-retinoic acid (ATRA) as an alternative AA treatment was tested by using an immune-mediated mouse model of AA. Results revealed that ATRA inhibited T-cell proliferation, activation and effector function. It also restrained the Fas/Fasl pathway, shifted Th1 towards Th2 cell development, rebalanced T-cell subsets at a relatively high level and corrected the Th1/Th2 ratio by targeting NFAT1 signalling. In addition, ATRA inhibited Th17 cell differentiation and promoted regulatory T-cell development. Therefore, ATRA was an effective agent to improve AA treatment outcomes.


Subject(s)
Anemia, Aplastic/immunology , Cell Differentiation/drug effects , NFATC Transcription Factors/immunology , Signal Transduction/immunology , Th1 Cells/immunology , Th2 Cells/immunology , Tretinoin/pharmacology , Anemia, Aplastic/pathology , Animals , Cell Differentiation/immunology , Female , Male , Mice , Mice, Inbred BALB C , Signal Transduction/drug effects , Th1 Cells/pathology , Th17 Cells/immunology , Th17 Cells/pathology , Th2 Cells/pathology
12.
Immunity ; 34(4): 479-91, 2011 Apr 22.
Article in English | MEDLINE | ID: mdl-21458306

ABSTRACT

The transcription factor FOXP3 is essential for the suppressive function of regulatory T cells that are required for maintaining self-tolerance. We have solved the crystal structure of the FOXP3 forkhead domain as a ternary complex with the DNA-binding domain of the transcription factor NFAT1 and a DNA oligonucleotide from the interleukin-2 promoter. A striking feature of this structure is that FOXP3 forms a domain-swapped dimer that bridges two molecules of DNA. Structure-guided or autoimmune disease (IPEX)-associated mutations in the domain-swap interface diminished dimer formation by the FOXP3 forkhead domain without compromising FOXP3 DNA binding. These mutations also eliminated T cell-suppressive activity conferred by FOXP3, both in vitro and in a murine model of autoimmune diabetes in vivo. We conclude that FOXP3-mediated suppressor function requires dimerization through the forkhead domain and that mutations in the dimer interface can lead to the systemic autoimmunity observed in IPEX patients.


Subject(s)
Forkhead Transcription Factors/chemistry , Forkhead Transcription Factors/immunology , T-Lymphocytes, Regulatory/chemistry , T-Lymphocytes, Regulatory/immunology , Amino Acid Sequence , Animals , Humans , Mice , Models, Molecular , Molecular Sequence Data , Mutation , NFATC Transcription Factors/chemistry , NFATC Transcription Factors/immunology , Protein Multimerization , Protein Structure, Quaternary , Protein Structure, Tertiary , Sequence Alignment
13.
Immunity ; 34(5): 703-14, 2011 May 27.
Article in English | MEDLINE | ID: mdl-21530328

ABSTRACT

A chief Ca(2+) entry pathway in immune cells is store-operated Ca(2+) (SOC) influx, which is triggered by depletion of Ca(2+) from the endoplasmic reticulum (ER). However, its physiological role in B cells remains elusive. Here, we show that ER calcium sensors STIM1- and STIM2-induced SOC influx is critical for B cell regulatory function. B cell-specific deletion of STIM1 and STIM2 in mice caused a profound defect in B cell receptor (BCR)-induced SOC influx and proliferation. However, B cell development and antibody responses were unaffected. Remarkably, B cells lacking both STIM proteins failed to produce the anti-inflammatory cytokine IL-10 because of defective activation of nuclear factor of activated T cells (NFAT) after BCR stimulation. This resulted in exacerbation of experimental autoimmune encephalomyelitis, a mouse model of multiple sclerosis. Our data establish STIM-dependent SOC influx as a key signal for B cell regulatory function required to limit autoimmunity.


Subject(s)
B-Lymphocytes/immunology , Calcium/metabolism , Interleukin-10/immunology , Membrane Glycoproteins/immunology , Animals , B-Lymphocytes/cytology , Brain Diseases/immunology , Brain Diseases/pathology , Calcium Channels , Cell Proliferation , Cell Survival , Encephalitis , Hashimoto Disease/immunology , Hashimoto Disease/pathology , Interleukin-10/biosynthesis , Membrane Glycoproteins/deficiency , Mice , NFATC Transcription Factors/immunology , Receptors, Antigen, B-Cell/immunology , Stromal Interaction Molecule 1 , Stromal Interaction Molecule 2
14.
Am J Physiol Renal Physiol ; 316(6): F1133-F1140, 2019 06 01.
Article in English | MEDLINE | ID: mdl-30785353

ABSTRACT

Chemotherapy-induced hemorrhagic cystitis is characterized by bladder pain and voiding dysfunction caused by hemorrhage and inflammation. Novel therapeutic options to treat hemorrhagic cystitis are needed. We previously reported that systemic administration of the Schistosomiasis hematobium-derived protein H-IPSEH06 (IL-4-inducing principle from Schistosoma mansoni eggs) is superior to three doses of MESNA in alleviating hemorrhagic cystitis (Mbanefo EC, Le L, Pennington LF, Odegaard JI, Jardetzky TS, Alouffi A, Falcone FH, Hsieh MH. FASEB J 32: 4408-4419, 2018). Based on prior reports by others on S. mansoni IPSE (M-IPSE) and additional work by our group, we reasoned that H-IPSE mediates its effects on hemorrhagic cystitis by binding IgE on basophils and inducing IL-4 expression, promoting urothelial proliferation, and translocating to the nucleus to modulate expression of genes implicated in relieving bladder dysfunction. We speculated that local bladder injection of the S. hematobium IPSE ortholog IPSEH03, hereafter called H-IPSEH03, might be more efficacious in preventing hemorrhagic cystitis compared with systemic administration of IPSEH06. We report that H-IPSEH03, like M-IPSE and H-IPSEH06, activates IgE-bearing basophils in a nuclear factor of activated T-cells reporter assay, indicating activation of the cytokine pathway. Furthermore, H-IPSEH03 attenuates ifosfamide-induced increases in bladder wet weight in an IL-4-dependent fashion. H-IPSEH03 relieves hemorrhagic cystitis-associated allodynia and modulates voiding patterns in mice. Finally, H-IPSEH03 drives increased urothelial cell proliferation, suggesting that IPSE induces bladder repair mechanisms. Taken together, H-IPSEH03 may be a potential novel therapeutic to treat hemorrhagic cystitis by basophil activation, attenuation of allodynia, and promotion of urothelial cell proliferation.


Subject(s)
Cell Proliferation/drug effects , Cystitis/prevention & control , Egg Proteins/administration & dosage , Helminth Proteins/administration & dosage , Hemorrhage/prevention & control , Immunologic Factors/administration & dosage , Urinary Bladder/drug effects , Urothelium/drug effects , Administration, Intravesical , Animals , Basophils/drug effects , Basophils/immunology , Basophils/metabolism , Cell Line , Cystitis/chemically induced , Cystitis/immunology , Cystitis/metabolism , Disease Models, Animal , Female , Hemorrhage/chemically induced , Hemorrhage/immunology , Hemorrhage/metabolism , Humans , Ifosfamide , Immunoglobulin E/immunology , Immunoglobulin E/metabolism , Injections, Intravenous , Interleukin-4/immunology , Interleukin-4/metabolism , Mice, Inbred C57BL , NFATC Transcription Factors/immunology , NFATC Transcription Factors/metabolism , Signal Transduction , Urinary Bladder/immunology , Urinary Bladder/metabolism , Urinary Bladder/pathology , Urodynamics/drug effects , Urothelium/immunology , Urothelium/metabolism , Urothelium/pathology
15.
J Immunol ; 198(1): 196-204, 2017 01 01.
Article in English | MEDLINE | ID: mdl-27895176

ABSTRACT

Despite a long history and extensive usage of insoluble aluminum salts (alum) as vaccine adjuvants, the molecular mechanisms underpinning Ag-specific immunity upon vaccination remain unclear. Dendritic cells (DCs) are crucial initiators of immune responses, but little is known about the molecular pathways used by DCs to sense alum and, in turn, activate T and B cells. In this article, we show that alum adjuvanticity requires IL-2 specifically released by DCs, even when T cell secretion of IL-2 is intact. We demonstrate that alum, as well as other sterile particulates, such as uric acid crystals, induces DCs to produce IL-2 following initiation of actin-mediated phagocytosis that leads to Src and Syk kinase activation, Ca2+ mobilization, and calcineurin-dependent activation of NFAT, the master transcription factor regulating IL-2 expression. Using chimeric mice, we show that DC-derived IL-2 is required for maximal Ag-specific proliferation of CD4+ T cells and optimal humoral responses following alum-adjuvanted immunization. These data identify DC-derived IL-2 as a key mediator of alum adjuvanticity in vivo and the Src-Syk pathway as a potential leverage point in the rational design of novel adjuvants.


Subject(s)
Adjuvants, Immunologic/pharmacology , Alum Compounds/pharmacology , Dendritic Cells/immunology , Signal Transduction/immunology , Animals , CD4-Positive T-Lymphocytes/immunology , Enzyme-Linked Immunosorbent Assay , Flow Cytometry , Interleukin-2/immunology , Lymphocyte Activation/drug effects , Mice , Mice, Inbred C57BL , Models, Animal , NFATC Transcription Factors/immunology , Signal Transduction/drug effects , Syk Kinase/immunology
16.
J Immunol ; 199(9): 3051-3062, 2017 11 01.
Article in English | MEDLINE | ID: mdl-28972088

ABSTRACT

The transcription factor NFAT1 plays a pivotal role in the homeostasis of T lymphocytes. However, its functional importance in non-CD4+ T cells, especially in systemic immune disorders, is largely unknown. In this study, we report that NFAT1 regulates dendritic cell (DC) tolerance and suppresses systemic autoimmunity using the experimental autoimmune myasthenia gravis (EAMG) as a model. Myasthenia gravis and EAMG are T cell-dependent, Ab-mediated autoimmune disorders in which the acetylcholine receptor is the major autoantigen. NFAT1-knockout mice showed higher susceptibility to EAMG development with enhanced Th1/Th17 cell responses. NFAT1 deficiency led to a phenotypic alteration of DCs that show hyperactivation of NF-κB-mediated signaling pathways and enhanced binding of NF-κB (p50) to the promoters of IL-6 and IL-12. As a result, NFAT1-knockout DCs produced much higher levels of proinflammatory cytokines such as IL-1ß, IL-6, IL-12, and TNF-α, which preferentially induce Th1/Th17 cell differentiation. Our data suggest that NFAT1 may limit the hyperactivation of the NF-κB-mediated proinflammatory response in DCs and suppress autoimmunity by serving as a key regulator of DC tolerance.


Subject(s)
Dendritic Cells/immunology , Lymphocyte Activation , Myasthenia Gravis, Autoimmune, Experimental/immunology , NFATC Transcription Factors/immunology , Signal Transduction/immunology , Animals , Cytokines/genetics , Cytokines/immunology , Dendritic Cells/pathology , Immune Tolerance/genetics , Mice , Mice, Transgenic , Myasthenia Gravis, Autoimmune, Experimental/genetics , Myasthenia Gravis, Autoimmune, Experimental/pathology , NF-kappa B/genetics , NF-kappa B/immunology , NFATC Transcription Factors/genetics , Signal Transduction/genetics , Th1 Cells/immunology , Th1 Cells/pathology , Th17 Cells/immunology , Th17 Cells/pathology
17.
Biochem Biophys Res Commun ; 495(2): 1864-1870, 2018 01 08.
Article in English | MEDLINE | ID: mdl-29225169

ABSTRACT

Orai1-dependent Ca2+ entry plays an essential role in inflammatory response through regulating T cell and macrophage activation and neutrophil infiltration. However, whether Orai1 Ca2+ entry contributes to endothelial activation, one of the early steps of vascular inflammation, remains elusive. In the present study, we observed that knockdown of Orai1 reduced, whereas overexpression of Orai1 potentiated, TNFα-induced expression of adhesion molecules such as ICAM-1 and VCAM-1 in HUVECs, and subsequently blocked adhesion of monocyte to HUVECs. In vivo, Orai1 downregulation attenuated TNFα-induced ICAM-1 and VCAM-1 expression in mouse aorta and the levels of pro-inflammatory cytokines in the serum. In addition, Orai1 knockdown also dramatically decreased the expression of pro-inflammatory cytokines and neutrophil infiltration in the lung after TNFα treatment, and thus protected lung tissue injury. Notably, among all isoforms of nuclear factor of activated T cells (NFATs), TNFα only triggered NFATc4 nuclear accumulation in HUVECs. Knockdown of Orai1 or inhibition of calcineurin prevented TNFα-induced NFATc4 nuclear translocation and reduced ICAM-1 and VCAM-1 expression in HUVECs. Overexpression of NFATc4 further enhanced ICAM-1 and VCAM-1 expression induced by TNFα. Our study demonstrates that Orai1-Ca2+-calcineurin-NFATc4 signaling is an essential inflammatory pathway required for TNFα-induced endothelial cell activation and vascular inflammation. Therefore, Orai1 may be a potential therapeutic target for treatment of inflammatory diseases.


Subject(s)
Aortitis/immunology , Calcineurin/immunology , Calcium/immunology , Cell Adhesion Molecules/immunology , Endothelium, Vascular/immunology , NFATC Transcription Factors/immunology , ORAI1 Protein/immunology , Animals , Aortitis/pathology , Cells, Cultured , Down-Regulation/immunology , Humans , Inflammation Mediators/immunology , Metabolic Networks and Pathways/immunology , Mice , Mice, Inbred C57BL
19.
Immunity ; 31(4): 587-97, 2009 Oct 16.
Article in English | MEDLINE | ID: mdl-19818650

ABSTRACT

T helper 17 (Th17) cells play major roles in autoimmunity and bacterial infections, yet how T cell receptor (TCR) signaling affects Th17 cell differentiation is relatively unknown. We demonstrate that CD4(+) T cells lacking Itk, a tyrosine kinase required for full TCR-induced phospholipase C-gamma (PLC-gamma1) activation, exhibit decreased interleukin-17A (IL-17A) expression in vitro and in vivo, despite relatively normal expression of retinoic acid receptor-related orphan receptor-gammaT (ROR-gammaT) and IL-17F. IL-17A expression was rescued by pharmacologically induced Ca(2+) influx or constitutively activated nuclear factor of activated T cells (NFAT). Conversely, decreased TCR stimulation or calcineurin inhibition preferentially reduced IL-17A expression. We further found that the promoter of Il17a but not Il17f has a conserved NFAT binding site that bound NFATc1 in wild-type but not Itk-deficient cells, even though both exhibited open chromatin conformations. Finally, Itk(-/-) mice also showed differential regulation of IL-17A and IL-17F in vivo. Our results suggest that Itk specifically couples TCR signaling to Il17a expression and the differential regulation of Th17 cell cytokines through NFATc1.


Subject(s)
CD4-Positive T-Lymphocytes/immunology , Cytokines/immunology , Interleukin-17/biosynthesis , Protein Serine-Threonine Kinases/metabolism , Protein-Tyrosine Kinases/metabolism , Animals , CD4-Positive T-Lymphocytes/metabolism , Calcium/immunology , Calcium/metabolism , Cytokines/metabolism , Lung/immunology , Lung/pathology , Mice , Mice, Inbred C57BL , Mice, Knockout , NFATC Transcription Factors/immunology , NFATC Transcription Factors/metabolism , Nuclear Receptor Subfamily 1, Group F, Member 3 , Phospholipase C gamma/immunology , Phospholipase C gamma/metabolism , Promoter Regions, Genetic/immunology , Protein Serine-Threonine Kinases/genetics , Protein Serine-Threonine Kinases/immunology , Protein-Tyrosine Kinases/genetics , Protein-Tyrosine Kinases/immunology , Receptors, Antigen, T-Cell/immunology , Receptors, Antigen, T-Cell/metabolism , Receptors, Retinoic Acid/immunology , Receptors, Retinoic Acid/metabolism , Receptors, Thyroid Hormone/immunology , Receptors, Thyroid Hormone/metabolism , Signal Transduction/immunology
20.
J Immunol ; 196(5): 2015-9, 2016 Mar 01.
Article in English | MEDLINE | ID: mdl-26851216

ABSTRACT

Follicular CD4(+) Th (Tfh) cells provide B cell help in germinal center reactions that support class switching, somatic hypermutation, and the generation of high-affinity Abs. In this article, we show that deficiency in NFAT1 and NFAT2 in CD4(+) T cells leads to impaired germinal center reactions upon viral infection because of reduced Tfh cell differentiation and defective expression of proteins involved in T/B interactions and B cell help, including ICOS, PD-1, and SLAM family receptors. Genome-wide chromatin immunoprecipitation data suggest that NFAT proteins likely directly participate in regulation of genes important for Tfh cell differentiation and function. NFAT proteins are important TCR and Ca(2+)-dependent regulators of T cell biology, and in this article we demonstrate a major positive role of NFAT family members in Tfh differentiation.


Subject(s)
Cell Differentiation/immunology , Gene Expression Regulation/immunology , NFATC Transcription Factors/immunology , T-Lymphocytes, Helper-Inducer/immunology , Virus Diseases/immunology , Animals , Chromatin Immunoprecipitation , Germinal Center/immunology , Lymphocyte Activation/immunology , Mice , Mice, Transgenic , T-Lymphocytes, Helper-Inducer/cytology
SELECTION OF CITATIONS
SEARCH DETAIL