Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 23
Filter
Add more filters










Publication year range
1.
Cancer Discov ; 2024 Jul 29.
Article in English | MEDLINE | ID: mdl-39073085

ABSTRACT

Iron accumulation in tumors contributes to disease progression and chemoresistance. While targeting this process can influence various hallmarks of cancer, the immunomodulatory effects of iron chelation in the tumor microenvironment are unknown. Here, we report that treatment with deferiprone, an FDA-approved iron chelator, unleashes innate immune responses that restrain ovarian cancer. Deferiprone reprogrammed ovarian cancer cells towards an immunostimulatory state characterized by production of type I interferon (IFN) and overexpression of molecules that activate natural killer (NK) cells. Mechanistically, these effects were driven by innate sensing of mitochondrial DNA in the cytosol and concomitant activation of nuclear DNA damage responses triggered upon iron chelation. Deferiprone synergized with chemotherapy and prolonged the survival of mice with ovarian cancer by bolstering type I IFN responses that drove NK cell-dependent control of metastatic disease. Hence, iron chelation may represent an alternative immunotherapeutic strategy for malignancies that are refractory to current T cell-centric modalities.

2.
Medicina (Kaunas) ; 60(6)2024 Jun 20.
Article in English | MEDLINE | ID: mdl-38929626

ABSTRACT

Background and Objectives: As modulators of the tumor microenvironment, macrophages have been extensively studied for their potential in developing anticancer strategies, particularly in regulating macrophage polarization towards an antitumorigenic (M1) phenotype rather than a protumorigenic (M2) one in various experimental models. Here, we evaluated the effect of PD98059, a mitogen-activated protein kinase kinase MAPKK MEK1-linked pathway inhibitor, on the differentiation and polarization of THP-1 monocytes in response to phorbol-12-myristate-13-acetate (PMA) under various culture conditions for tumor microenvironmental application. Materials and Methods: Differentiation and polarization of THP-1 were analyzed by flow cytometry and RT-PCR. Polarized THP-1 subsets with different treatment were compared by motility, phagocytosis, and so on. Results: Clearly, PMA induced THP-1 differentiation occurs in adherent culture conditions more than nonadherent culture conditions by increasing CD11b expression up to 90%, which was not affected by PD98059 when cells were exposed to PMA first (post-PD) but inhibited when PD98059 was treated prior to PMA treatment (pre-PD). CD11bhigh THP-1 cells treated with PMA and PMA-post-PD were categorized into M0 (HLA-DRlow and CD206low), M1 (HLA-DRhigh and CD206low), and M2 (HLA-DRlow and CD206high), resulting in an increased population of M1 macrophages. The transcription levels of markers of macrophage differentiation and polarization confirmed the increased M1 polarization of THP-1 cells with post-PD treatment rather than with PMA-only treatment. The motility and cytotoxicity of THP-1 cells with post-PD treatment were higher than THP-1 cells with PMA, suggesting that post-PD treatment enhanced the anti-tumorigenicity of THP-1 cells. Confocal microscopy and flow cytometry showed the effect of post-PD treatment on phagocytosis by THP-1 cells. Conclusions: We have developed an experimental model of macrophage polarization with THP-1 cells which will be useful for further studies related to the tumor microenvironment.


Subject(s)
Cell Differentiation , Flavonoids , Macrophages , Monocytes , Tetradecanoylphorbol Acetate , Humans , Macrophages/drug effects , Tetradecanoylphorbol Acetate/pharmacology , Flavonoids/pharmacology , Flavonoids/therapeutic use , THP-1 Cells , Cell Differentiation/drug effects , Monocytes/drug effects , Flow Cytometry , Phagocytosis/drug effects
3.
J Clin Invest ; 133(17)2023 09 01.
Article in English | MEDLINE | ID: mdl-37432737

ABSTRACT

Recognition of pathogen-associated molecular patterns can trigger the inositol-requiring enzyme 1 α (IRE1α) arm of the endoplasmic reticulum (ER) stress response in innate immune cells. This process maintains ER homeostasis and also coordinates diverse immunomodulatory programs during bacterial and viral infections. However, the role of innate IRE1α signaling in response to fungal pathogens remains elusive. Here, we report that systemic infection with the human opportunistic fungal pathogen Candida albicans induced proinflammatory IRE1α hyperactivation in myeloid cells that led to fatal kidney immunopathology. Mechanistically, simultaneous activation of the TLR/IL-1R adaptor protein MyD88 and the C-type lectin receptor dectin-1 by C. albicans induced NADPH oxidase-driven generation of ROS, which caused ER stress and IRE1α-dependent overexpression of key inflammatory mediators such as IL-1ß, IL-6, chemokine (C-C motif) ligand 5 (CCL5), prostaglandin E2 (PGE2), and TNF-α. Selective ablation of IRE1α in leukocytes, or treatment with an IRE1α pharmacological inhibitor, mitigated kidney inflammation and prolonged the survival of mice with systemic C. albicans infection. Therefore, controlling IRE1α hyperactivation may be useful for impeding the immunopathogenic progression of disseminated candidiasis.


Subject(s)
Candidiasis , Protein Serine-Threonine Kinases , Humans , Animals , Mice , Protein Serine-Threonine Kinases/metabolism , Endoribonucleases/metabolism , Endoplasmic Reticulum Stress , Candida albicans , Toll-Like Receptors/metabolism , Adaptor Proteins, Signal Transducing/metabolism
4.
Nat Commun ; 14(1): 120, 2023 01 09.
Article in English | MEDLINE | ID: mdl-36624093

ABSTRACT

IRE1α-XBP1 signaling is emerging as a central orchestrator of malignant progression and immunosuppression in various cancer types. Employing a computational XBP1s detection method applied to TCGA datasets, we demonstrate that expression of the XBP1s mRNA isoform predicts poor survival in non-small cell lung cancer (NSCLC) patients. Ablation of IRE1α in malignant cells delays tumor progression and extends survival in mouse models of NSCLC. This protective effect is accompanied by alterations in intratumoral immune cell subsets eliciting durable adaptive anti-cancer immunity. Mechanistically, cancer cell-intrinsic IRE1α activation sustains mPGES-1 expression, enabling production of the immunosuppressive lipid mediator prostaglandin E2. Accordingly, restoring mPGES-1 expression in IRE1αKO cancer cells rescues normal tumor progression. We have developed an IRE1α gene signature that predicts immune cell infiltration and overall survival in human NSCLC. Our study unveils an immunoregulatory role for cancer cell-intrinsic IRE1α activation and suggests that targeting this pathway may help enhance anti-tumor immunity in NSCLC.


Subject(s)
Carcinoma, Non-Small-Cell Lung , Endoribonucleases , Lung Neoplasms , Protein Serine-Threonine Kinases , Animals , Humans , Mice , Carcinoma, Non-Small-Cell Lung/genetics , Endoribonucleases/genetics , Endoribonucleases/metabolism , Lung Neoplasms/genetics , Protein Serine-Threonine Kinases/genetics , Protein Serine-Threonine Kinases/metabolism , Signal Transduction , X-Box Binding Protein 1/genetics , X-Box Binding Protein 1/metabolism
5.
Res Sq ; 2023 Dec 14.
Article in English | MEDLINE | ID: mdl-38168227

ABSTRACT

Mounting effective immunity against pathogens and tumors relies on the successful metabolic programming of T cells by extracellular fatty acids1-3. During this process, fatty-acid-binding protein 5 (FABP5) imports lipids that fuel mitochondrial respiration and sustain the bioenergetic requirements of protective CD8+ T cells4,5. Importantly, however, the mechanisms governing this crucial immunometabolic axis remain unexplored. Here we report that the cytoskeletal organizer Transgelin 2 (TAGLN2) is necessary for optimal CD8+ T cell fatty acid uptake, mitochondrial respiration, and anti-cancer function. We found that TAGLN2 interacts with FABP5, enabling the surface localization of this lipid importer on activated CD8+ T cells. Analysis of ovarian cancer specimens revealed that endoplasmic reticulum (ER) stress responses elicited by the tumor microenvironment repress TAGLN2 in infiltrating CD8+ T cells, enforcing their dysfunctional state. Restoring TAGLN2 expression in ER-stressed CD8+ T cells bolstered their lipid uptake, mitochondrial respiration, and cytotoxic capacity. Accordingly, chimeric antigen receptor T cells overexpressing TAGLN2 bypassed the detrimental effects of tumor-induced ER stress and demonstrated superior therapeutic efficacy in mice with metastatic ovarian cancer. Our study unveils the role of cytoskeletal TAGLN2 in T cell lipid metabolism and highlights the potential to enhance cellular immunotherapy in solid malignancies by preserving the TAGLN2-FABP5 axis.

6.
J Immunol ; 209(1): 69-76, 2022 07 01.
Article in English | MEDLINE | ID: mdl-35697385

ABSTRACT

Obesity is associated with increased cancer risk and weak responses to vaccination and sepsis treatment. Although dendritic cells (DCs) are fundamental for the initiation and maintenance of competent immune responses against pathogens and tumors, how obesity alters the normal physiology of these myeloid cells remains largely unexplored. In this study, we report that obesity caused by prolonged high-fat diet feeding disrupts the metabolic and functional status of mouse splenic DCs (SpDCs). High-fat diet-induced obesity drastically altered the global transcriptional profile of SpDCs, causing severe changes in the expression of gene programs implicated in lipid metabolism and mitochondrial function. SpDCs isolated from obese mice demonstrated enhanced mitochondrial respiration provoked by increased fatty acid oxidation (FAO), which drove the intracellular accumulation of reactive oxygen species that impaired Ag presentation to T cells. Accordingly, treatment with the FAO inhibitor etomoxir, or antioxidants such as vitamin E or N-acetyl-l-cysteine, restored the Ag-presenting capacity of SpDCs isolated from obese mice. Our findings reveal a major detrimental effect of obesity in DC physiology and suggest that controlling mitochondrial FAO or reactive oxygen species overproduction may help improve DC function in obese individuals.


Subject(s)
Diet, High-Fat , Fatty Acids , Animals , Dendritic Cells , Diet, High-Fat/adverse effects , Fatty Acids/metabolism , Homeostasis , Lipid Metabolism , Mice , Mice, Obese , Mitochondria/metabolism , Obesity/metabolism , Oxidation-Reduction , Reactive Oxygen Species/metabolism
7.
Cancer Discov ; 12(8): 1904-1921, 2022 08 05.
Article in English | MEDLINE | ID: mdl-35552618

ABSTRACT

Lysophosphatidic acid (LPA) is a bioactive lipid enriched in the tumor microenvironment of immunosuppressive malignancies such as ovarian cancer. Although LPA enhances the tumorigenic attributes of cancer cells, the immunomodulatory activity of this phospholipid messenger remains largely unexplored. Here, we report that LPA operates as a negative regulator of type I interferon (IFN) responses in ovarian cancer. Ablation of the LPA-generating enzyme autotaxin (ATX) in ovarian cancer cells reprogrammed the tumor immune microenvironment, extended host survival, and improved the effects of therapies that elicit protective responses driven by type I IFN. Mechanistically, LPA sensing by dendritic cells triggered PGE2 biosynthesis that suppressed type I IFN signaling via autocrine EP4 engagement. Moreover, we identified an LPA-controlled, immune-derived gene signature associated with poor responses to combined PARP inhibition and PD-1 blockade in patients with ovarian cancer. Controlling LPA production or sensing in tumors may therefore be useful to improve cancer immunotherapies that rely on robust induction of type I IFN. SIGNIFICANCE: This study uncovers that ATX-LPA is a central immunosuppressive pathway in the ovarian tumor microenvironment. Ablating this axis sensitizes ovarian cancer hosts to various immunotherapies by unleashing protective type I IFN responses. Understanding the immunoregulatory programs induced by LPA could lead to new biomarkers predicting resistance to immunotherapy in patients with cancer. See related commentary by Conejo-Garcia and Curiel, p. 1841. This article is highlighted in the In This Issue feature, p. 1825.


Subject(s)
Interferon Type I , Lysophospholipids , Ovarian Neoplasms , Female , Humans , Lysophospholipids/genetics , Lysophospholipids/metabolism , Ovarian Neoplasms/drug therapy , Ovarian Neoplasms/genetics , Ovarian Neoplasms/pathology , Receptors, Lysophosphatidic Acid/genetics , Receptors, Lysophosphatidic Acid/metabolism , Tumor Microenvironment
8.
Science ; 365(6450)2019 07 19.
Article in English | MEDLINE | ID: mdl-31320508

ABSTRACT

Inositol-requiring enzyme 1[α] (IRE1[α])-X-box binding protein spliced (XBP1) signaling maintains endoplasmic reticulum (ER) homeostasis while controlling immunometabolic processes. Yet, the physiological consequences of IRE1α-XBP1 activation in leukocytes remain unexplored. We found that induction of prostaglandin-endoperoxide synthase 2 (Ptgs2/Cox-2) and prostaglandin E synthase (Ptges/mPGES-1) was compromised in IRE1α-deficient myeloid cells undergoing ER stress or stimulated through pattern recognition receptors. Inducible biosynthesis of prostaglandins, including the pro-algesic mediator prostaglandin E2 (PGE2), was decreased in myeloid cells that lack IRE1α or XBP1 but not other ER stress sensors. Functional XBP1 transactivated the human PTGS2 and PTGES genes to enable optimal PGE2 production. Mice that lack IRE1α-XBP1 in leukocytes, or that were treated with IRE1α inhibitors, demonstrated reduced pain behaviors in PGE2-dependent models of pain. Thus, IRE1α-XBP1 is a mediator of prostaglandin biosynthesis and a potential target to control pain.


Subject(s)
Dinoprostone/biosynthesis , Endoribonucleases/metabolism , Leukocytes/metabolism , Pain, Postoperative/metabolism , Protein Serine-Threonine Kinases/metabolism , Visceral Pain/metabolism , X-Box Binding Protein 1/metabolism , Animals , Cells, Cultured , Cyclooxygenase 2/genetics , Endoribonucleases/genetics , Humans , Mice , Mice, Inbred C57BL , Myeloid Cells/metabolism , Pain, Postoperative/genetics , Promoter Regions, Genetic , Prostaglandin-E Synthases/genetics , Protein Serine-Threonine Kinases/genetics , Signal Transduction , Unfolded Protein Response , Visceral Pain/genetics , X-Box Binding Protein 1/genetics
9.
Nature ; 562(7727): 423-428, 2018 10.
Article in English | MEDLINE | ID: mdl-30305738

ABSTRACT

Tumours evade immune control by creating hostile microenvironments that perturb T cell metabolism and effector function1-4. However, it remains unclear how intra-tumoral T cells integrate and interpret metabolic stress signals. Here we report that ovarian cancer-an aggressive malignancy that is refractory to standard treatments and current immunotherapies5-8-induces endoplasmic reticulum stress and activates the IRE1α-XBP1 arm of the unfolded protein response9,10 in T cells to control their mitochondrial respiration and anti-tumour function. In T cells isolated from specimens collected from patients with ovarian cancer, upregulation of XBP1 was associated with decreased infiltration of T cells into tumours and with reduced IFNG mRNA expression. Malignant ascites fluid obtained from patients with ovarian cancer inhibited glucose uptake and caused N-linked protein glycosylation defects in T cells, which triggered IRE1α-XBP1 activation that suppressed mitochondrial activity and IFNγ production. Mechanistically, induction of XBP1 regulated the abundance of glutamine carriers and thus limited the influx of glutamine that is necessary to sustain mitochondrial respiration in T cells under glucose-deprived conditions. Restoring N-linked protein glycosylation, abrogating IRE1α-XBP1 activation or enforcing expression of glutamine transporters enhanced mitochondrial respiration in human T cells exposed to ovarian cancer ascites. XBP1-deficient T cells in the metastatic ovarian cancer milieu exhibited global transcriptional reprogramming and improved effector capacity. Accordingly, mice that bear ovarian cancer and lack XBP1 selectively in T cells demonstrate superior anti-tumour immunity, delayed malignant progression and increased overall survival. Controlling endoplasmic reticulum stress or targeting IRE1α-XBP1 signalling may help to restore the metabolic fitness and anti-tumour capacity of T cells in cancer hosts.


Subject(s)
Endoribonucleases/metabolism , Mitochondria/metabolism , Ovarian Neoplasms/immunology , Protein Serine-Threonine Kinases/metabolism , T-Lymphocytes/cytology , T-Lymphocytes/immunology , X-Box Binding Protein 1/metabolism , Amino Acid Transport Systems, Basic , Animals , Ascites/metabolism , Cell Respiration , Disease Progression , Endoplasmic Reticulum Stress , Female , Gene Expression Regulation, Neoplastic , Glucose/metabolism , Glutamine/metabolism , Glycosylation , Humans , Interferon-gamma/biosynthesis , Interferon-gamma/genetics , Mice , Neoplasm Metastasis , Neoplasm Transplantation , Ovarian Neoplasms/pathology , Signal Transduction , Survival Rate , T-Lymphocytes/metabolism , Tumor Escape/immunology , Unfolded Protein Response , X-Box Binding Protein 1/biosynthesis , X-Box Binding Protein 1/deficiency
10.
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
11.
Cancer Immunol Immunother ; 66(8): 969-977, 2017 Aug.
Article in English | MEDLINE | ID: mdl-28214928

ABSTRACT

Immune-based therapies that induce remarkable and durable responses against melanoma and lung cancer have unfortunately demonstrated limited success in ovarian cancer patients. This is likely due to the exceptional immunoregulatory nature of ovarian tumors, which employ numerous strategies to effectively suppress anti-tumor immunity. Here, we summarize a decade of research indicating that ovarian cancers possess an exquisite capacity to subvert the activity of host dendritic cells (DCs) as a key mechanism to impede the development and maintenance of protective T cell-based immune responses. Identifying, understanding, and disabling the precise mechanisms promoting DC dysfunction in ovarian cancer are, therefore, fundamental requirements for devising the next generation of successful immunotherapies against this devastating malignancy.


Subject(s)
Cancer Vaccines/immunology , Dendritic Cells/immunology , Immunotherapy , Myeloid-Derived Suppressor Cells/immunology , Ovarian Neoplasms/immunology , Animals , Female , Humans , Immunity , Immunomodulation , Ovarian Neoplasms/therapy , Tumor Escape , Tumor Microenvironment
12.
Sci Rep ; 6: 19453, 2016 Jan 18.
Article in English | MEDLINE | ID: mdl-26777750

ABSTRACT

Allergic contact hypersensitivity (CHS) is an inflammatory skin disease mediated by allergen specific T cells. In this study, we investigated the role of transcription factor NFAT1 in the pathogenesis of contact hypersensitivity. NFAT1 knock out (KO) mice spontaneously developed CHS-like skin inflammation in old age. Healthy young NFAT1 KO mice displayed enhanced susceptibility to hapten-induced CHS. Both CD4(+) and CD8(+) T cells from NFAT1 KO mice displayed hyper-activated properties and produced significantly enhanced levels of inflammatory T helper 1(Th1)/Th17 type cytokines. NFAT1 KO T cells were more resistant to activation induced cell death (AICD), and regulatory T cells derived from these mice showed a partial defect in their suppressor activity. NFAT1 KO T cells displayed a reduced expression of apoptosis associated BCL-2/BH3 family members. Ectopic expression of NFAT1 restored the AICD defect in NFAT1 KO T cells and increased AICD in normal T cells. Recipient Rag2(-/-) mice transferred with NFAT1 KO T cells showed more severe CHS sensitivity due to a defect in activation induced hapten-reactive T cell apoptosis. Collectively, our results suggest the NFAT1 plays a pivotal role as a genetic switch in CD4(+)/CD8(+) T cell tolerance by regulating AICD process in the T cell mediated skin inflammation.


Subject(s)
Dermatitis, Contact/immunology , Dermatitis, Contact/metabolism , NFATC Transcription Factors/metabolism , T-Lymphocyte Subsets/immunology , T-Lymphocyte Subsets/metabolism , Animals , Apoptosis/genetics , Cell Death/genetics , Cell Death/immunology , Dermatitis, Allergic Contact/genetics , Dermatitis, Allergic Contact/immunology , Dermatitis, Allergic Contact/metabolism , Disease Models, Animal , Gene Expression Regulation , Immune Tolerance , Immunomodulation , Lymphocyte Activation/genetics , Lymphocyte Activation/immunology , Mice , Mice, Knockout , NFATC Transcription Factors/genetics , T-Lymphocytes, Regulatory/immunology , T-Lymphocytes, Regulatory/metabolism
13.
J Immunol ; 194(4): 1963-74, 2015 Feb 15.
Article in English | MEDLINE | ID: mdl-25595785

ABSTRACT

IL-31 is a key mediator of itching in atopic dermatitis (AD) and is preferentially produced by activated CD4(+) T cells and Th2 cells. Although pathophysiological functions of IL-31 have been suggested in diverse immune disorders, the molecular events underlying IL-31 gene regulation are still unclear. In this study we identified the transcription start site and functional promoter involved in IL-31 gene regulation in mouse CD4(+) T cells. TCR stimulation-dependent IL-31 expression was found to be closely linked with in vivo binding of NFAT1 and JunB to the IL-31 promoter. Although NFAT1 alone enhanced IL-31 promoter activity, it was further enhanced in the presence of JunB. Conversely, knockdown of either NFAT1 or JunB resulted in reduced IL-31 expression. NFAT1-deficient CD4(+) T cells showed a significant defect in IL-31 expression compared with wild-type CD4(+) T cells. In agreement with these findings, mice subjected to atopic conditions showed much higher levels of IL-31, which were closely correlated with a significant increase in the number of infiltrated NFAT1(+)CD4(+) T cells into the AD ears. Amelioration of AD progression by cyclosporin A treatment was well correlated with downregulation of IL-31 expressions in CD4(+) T cells and total ear residual cells. In summary, our results suggest a functional cooperation between NFAT1 and JunB in mediating IL-31 gene expression in CD4(+) T cells and indicate that interference with this interaction or their activity has the potential of reducing IL-31-mediated AD symptoms.


Subject(s)
CD4-Positive T-Lymphocytes/immunology , Dermatitis, Atopic/immunology , Gene Expression Regulation/immunology , Interleukins/biosynthesis , NFATC Transcription Factors/immunology , Transcription Factors/immunology , Animals , Chromatin Immunoprecipitation , Dermatitis, Atopic/genetics , Dermatitis, Atopic/metabolism , Disease Models, Animal , Female , Immunohistochemistry , Interleukins/genetics , Interleukins/immunology , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mutagenesis, Site-Directed , NFATC Transcription Factors/genetics , RNA, Small Interfering , Real-Time Polymerase Chain Reaction , Transcription Factors/genetics , Transcriptome , Transfection
14.
PLoS Biol ; 12(6): e1001881, 2014 Jun.
Article in English | MEDLINE | ID: mdl-24914685

ABSTRACT

Rheumatoid arthritis (RA) is a systemic autoimmune disorder that manifests as chronic inflammation and joint tissue destruction. However, the etiology and pathogenesis of RA have not been fully elucidated. Here, we explored the role of the hypoxia-inducible factors (HIFs), HIF-1α (encoded by HIF1A) and HIF-2α (encoded by EPAS1). HIF-2α was markedly up-regulated in the intimal lining of RA synovium, whereas HIF-1α was detected in a few cells in the sublining and deep layer of RA synovium. Overexpression of HIF-2α in joint tissues caused an RA-like phenotype, whereas HIF-1α did not affect joint architecture. Moreover, a HIF-2α deficiency in mice blunted the development of experimental RA. HIF-2α was expressed mainly in fibroblast-like synoviocytes (FLS) of RA synovium and regulated their proliferation, expression of RANKL (receptor activator of nuclear factor-κB ligand) and various catabolic factors, and osteoclastogenic potential. Moreover, HIF-2α-dependent up-regulation of interleukin (IL)-6 in FLS stimulated differentiation of TH17 cells-crucial effectors of RA pathogenesis. Additionally, in the absence of IL-6 (Il6-/- mice), overexpression of HIF-2α in joint tissues did not cause an RA phenotype. Thus, our results collectively suggest that HIF-2α plays a pivotal role in the pathogenesis of RA by regulating FLS functions, independent of HIF-1α.


Subject(s)
Arthritis, Experimental/etiology , Arthritis, Rheumatoid/etiology , Basic Helix-Loop-Helix Transcription Factors/metabolism , Animals , Arthritis, Experimental/metabolism , Arthritis, Rheumatoid/metabolism , Basic Helix-Loop-Helix Transcription Factors/immunology , Cell Differentiation , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Interleukin-6/metabolism , Male , Mice, Inbred C57BL , Mice, Inbred DBA , Phenotype , Synovial Membrane/metabolism , Th17 Cells/cytology , Up-Regulation
15.
J Biol Chem ; 287(19): 15445-57, 2012 May 04.
Article in English | MEDLINE | ID: mdl-22427656

ABSTRACT

IL-9 regulates diverse inflammatory immune responses. Although the functional importance of IL-9 has been investigated in various pathophysiological conditions, molecular mechanisms by which TCR stimulation induced IL-9 gene expression are still unclear. In this study, we investigated the functional importance of the NFAT1 and NF-κB (p65) in IL-9 gene transcription in CD4(+) T cells. In vivo binding of NFAT1 and NF-κB (p65) to the IL-9 promoter was observed. NFAT1 binding induced a transcriptionally active chromatin configuration at the IL-9 promoter locus, whereas NF-κB (p65) binding transactivated the IL-9 promoter. Mouse deficient in NFAT1 shows a significant down-regulation of IL-9 expression that resulted from an inaccessible chromatin configuration at the IL-9 promoter. In parallel, knockdown of NF-κB (p65) also resulted in reduced IL-9 expression. In this process, NFAT1 plays a pivotal role as a core protein that creates an accessible platform for the assembly of transcription activators. The presence of NFAT1 correlates with recruitment of NF-κB (p65), p300, and active histone markers on the IL-9 promoter, resulting in a transcriptionally competent promoter. NFAT1 deficiency significantly reduced the recruitment of the above activation complex to the IL-9 promoter. In summary, our data suggest that functional cooperation of NFAT1 and NF-κB synergistically enhances IL-9 transcription in CD4(+) T cells.


Subject(s)
Chromatin/metabolism , Interleukin-9/genetics , NFATC Transcription Factors/metabolism , Transcription Factor RelA/metabolism , Transcriptional Activation , Animals , Base Sequence , Binding Sites/genetics , CD4-Positive T-Lymphocytes/cytology , CD4-Positive T-Lymphocytes/metabolism , Cells, Cultured , Chromatin/genetics , Chromatin Immunoprecipitation , HEK293 Cells , Humans , Immunoblotting , Mice , Mice, Inbred C57BL , Mice, Knockout , Molecular Sequence Data , Mutation , NFATC Transcription Factors/genetics , Promoter Regions, Genetic/genetics , Protein Binding , RNA Interference , Sequence Homology, Nucleic Acid , Transcription Factor RelA/genetics
16.
J Immunol ; 188(5): 2244-53, 2012 Mar 01.
Article in English | MEDLINE | ID: mdl-22266280

ABSTRACT

IL-10 is a multifunctional cytokine that plays a crucial role in immunity and tolerance. IL-10 is produced by diverse immune cell types, including B cells and subsets of T cells. Although Th1 produce IL-10, their expression levels are much lower than Th2 cells under conventional stimulation conditions. The potential role of E26 transformation-specific 1 (Ets-1) transcription factor as a negative regulator for Il10 gene expression in CD4(+) T cells has been implicated previously. In this study, we investigated the underlying mechanism of Ets-1-mediated Il10 gene repression in Th1 cells. Compared with wild type Th1 cells, Ets-1 knockout Th1 cells expressed a significantly higher level of IL-10, which is comparable with that of wild type Th2 cells. Upregulation of IL-10 expression in Ets-1 knockout Th1 cells was accompanied by enhanced chromatin accessibility and increased recruitment of histone H3 acetylation at the Il10 regulatory regions. Reciprocally, Ets-1 deficiency significantly decreased histone deacetylase 1 (HDAC1) enrichment at the Il10 regulatory regions. Treatment with trichostatin A, an inhibitor of HDAC family, significantly increased Il10 gene expression by increasing histone H3 acetylation recruitment. We further demonstrated a physical interaction between Ets-1 and HDAC1. Coexpression of Ets-1 with HDAC1 synergistically repressed IL-10 transcription activity. In summary, our data suggest that an interaction of Ets-1 with HDAC1 represses the Il10 gene expression in Th1 cells.


Subject(s)
Down-Regulation/immunology , Gene Expression Regulation/immunology , Histone Deacetylase 1/physiology , Interleukin-10/antagonists & inhibitors , Interleukin-10/biosynthesis , Proto-Oncogene Protein c-ets-1/physiology , Th1 Cells/immunology , Th1 Cells/metabolism , Animals , Cell Differentiation/genetics , Cell Differentiation/immunology , Cells, Cultured , Down-Regulation/genetics , HEK293 Cells , Histone Deacetylase 1/antagonists & inhibitors , Histone Deacetylase 1/metabolism , Humans , Interleukin-10/genetics , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Knockout , Proto-Oncogene Protein c-ets-1/deficiency , Proto-Oncogene Protein c-ets-1/metabolism , Th1 Cells/cytology , Up-Regulation/genetics , Up-Regulation/immunology
17.
PLoS One ; 7(12): e52119, 2012.
Article in English | MEDLINE | ID: mdl-23284891

ABSTRACT

Probiotics are live bacteria that confer health benefits to the host physiology. Although protective role of probiotics have been reported in diverse diseases, no information is available whether probiotics can modulate neuromuscular immune disorders. We have recently demonstrated that IRT5 probiotics, a mixture of 5 probiotics, could suppress diverse experimental disorders in mice model. In this study we further investigated whether IRT5 probiotics could modulate the progression of experimental autoimmune myasthenia gravis (EAMG). Myasthenia gravis (MG) is a T cell dependent antibody mediated autoimmune disorder in which acetylcholine receptor (AChR) at the neuromuscular junction is the major auto-antigen. Oral administration of IRT5 probiotics significantly reduced clinical symptoms of EAMG such as weight loss, body trembling and grip strength. Prophylactic effect of IRT5 probiotics on EMAG is mediated by down-regulation of effector function of AChR-reactive T cells and B cells. Administration of IRT5 probiotics decreased AChR-reactive lymphocyte proliferation, anti-AChR reactive IgG levels and inflammatory cytokine levels such as IFN-γ, TNF-α, IL-6 and IL-17. Down-regulation of inflammatory mediators in AChR-reactive lymphocytes by IRT5 probiotics is mediated by the generation of regulatory dendritic cells (rDCs) that express increased levels of IL-10, TGF-ß, arginase 1 and aldh1a2. Furthermore, DCs isolated from IRT5 probiotics-fed group effectively converted CD4(+) T cells into CD4(+)Foxp3(+) regulatory T cells compared with control DCs. Our data suggest that IRT5 probiotics could be applicable to modulate antibody mediated autoimmune diseases including myasthenia gravis.


Subject(s)
Myasthenia Gravis, Autoimmune, Experimental/prevention & control , Probiotics/therapeutic use , Administration, Oral , Animals , Antibodies/immunology , Antibodies/metabolism , Autoantibodies/immunology , Autoantibodies/metabolism , Cytokines/immunology , Cytokines/metabolism , Dendritic Cells/immunology , Disease Progression , Female , Immune Tolerance , Inflammation Mediators/immunology , Inflammation Mediators/metabolism , Lymph Nodes/immunology , Lymphocyte Activation/immunology , Lymphocytes/immunology , Lymphocytes/metabolism , Mesentery/immunology , Myasthenia Gravis, Autoimmune, Experimental/immunology , Myasthenia Gravis, Autoimmune, Experimental/metabolism , Probiotics/administration & dosage , Rats , Receptors, Cholinergic/immunology , Receptors, Cholinergic/metabolism
18.
PLoS One ; 6(7): e22042, 2011.
Article in English | MEDLINE | ID: mdl-21799768

ABSTRACT

Nuclear factor of activated T cells (NFAT) is a family of transcription factors composed of five proteins. Among them, NFAT1 is a predominant NFAT protein in CD4(+) T cells. NFAT1 positively regulates transcription of a large number of inducible cytokine genes including IL-2, IL-4, IL-5 and other cytokines. However, disruption of NFAT1 results in an unexpected increase of IL-4. In this study, we have investigated the role of NFAT1 in regulation of IL-4 gene expression in T helper 2 cells (Th2) from an epigenetic viewpoint. NFAT1 deficient Th2 cells showed a sustained IL-4 expression while wild type (WT) cells reduced its expression. We tested whether epigenetic maintenance and changes in the chromatin architecture of IL-4 promoter locus play a role in differential IL-4 transcription between in WT and NFAT1 deficient Th2 cells. Compared with WT, NFAT1 deficient CD4(+) Th2 cells exhibited enhanced chromatin accessibility with permissive histone modification and DNA demethylation in the IL-4 promoter region. Transcription factors bound to IL-4 promoter region in the absence of NFAT1 were identified by Micro-LC/LC-MS/MS analysis. Among the candidates, preferential recruitment of JUNB to the IL-4 promoter was confirmed by chromatin immunoprecipitation analysis. Overexpression of JUNB together with SATB1 synergistically upregulated IL-4 promoter activity, while knockdown JUNB significantly reduced IL-4 expression. Our results suggest that the prolonged IL-4 expression in NFAT1 deficient Th2 cells is mediated by preferential binding of JUNB/SATB1 to the IL-4 promoter with permissive chromatin architecture.


Subject(s)
Chromatin/metabolism , Gene Expression Regulation , Interleukin-4/genetics , NFATC Transcription Factors/deficiency , Proto-Oncogene Proteins c-jun/metabolism , Th2 Cells/metabolism , Amino Acid Sequence , Animals , Cell Adhesion Molecules, Neuronal/chemistry , Cell Adhesion Molecules, Neuronal/metabolism , Chromatin/genetics , CpG Islands , DNA Methylation , Female , Humans , Jurkat Cells , Mice , Molecular Sequence Data , Promoter Regions, Genetic/genetics , Proto-Oncogene Proteins c-jun/chemistry , Th2 Cells/cytology , Transcriptional Activation
19.
Biochem Biophys Res Commun ; 407(1): 141-7, 2011 Apr 01.
Article in English | MEDLINE | ID: mdl-21371435

ABSTRACT

The function and differentiation of induced regulatory T (iTreg) cells are tightly regulated by various signaling cascade. In this study, we have investigated the role of TCR signaling to induce Foxp3 gene expression in cooperation with TGF-ß/IL-2 stimulation. Activation of CD4(+) T cells by TCR signaling or TGF-ß/IL-2 alone failed to enhance Foxp3 expression. Only when TCR stimulation is coupled together with TGF-ß/IL-2, CD4(+) T cells expressed high levels of Foxp3 by maintaining open chromatin structure around its promoter region. Under this condition, stimulation-dependent recruitment of JunB together with c-Rel enhanced Foxp3 expression. Over expression of JunB and c-Rel significantly enhanced Foxp3 promoter activity while treatment of JunB siRNA or inhibition of TCR signaling by MAPK inhibitors significantly reduced Foxp3 expression. Collectively our results suggest that TCR signaling together with TGF-ß/IL-2 stimulation cooperatively enhance Foxp3 gene expression by maintaining accessible chromatin structure and by actively recruiting key transcription factors JunB and c-Rel.


Subject(s)
Forkhead Transcription Factors/genetics , Gene Expression Regulation , Lymphocyte Activation/genetics , Proto-Oncogene Proteins c-jun/metabolism , Proto-Oncogene Proteins c-rel/metabolism , T-Lymphocytes, Regulatory/immunology , Animals , Cell Differentiation/genetics , Chromatin/metabolism , Chromatin/ultrastructure , Epigenomics , Interleukin-2/pharmacology , Mice , Mice, Inbred C57BL , Promoter Regions, Genetic , Receptors, Antigen, T-Cell/metabolism , T-Lymphocytes, Regulatory/drug effects , Transforming Growth Factor beta/pharmacology
20.
Life Sci ; 88(7-8): 358-66, 2011 Feb 14.
Article in English | MEDLINE | ID: mdl-21167838

ABSTRACT

AIMS: We previously reported that Lactobacillus casei (L. casei) has beneficial effects in experimental rheumatoid arthritis (RA) by suppressing inflammatory immune responses. The major purpose of this study was to evaluate therapeutic effects of L. casei on pathological responses in experimental rodent model of osteoarthritis (OA). MAIN METHODS: Experimental OA was induced by intra-articular injection of monosodium iodoacetate (MIA) in Wistar rats. L. casei alone or together with type II collagen (CII) and glucosamine (Gln) was orally administered into OA rats. The pathophysiological aspects of OA were investigated by analyzing mechanical hyperalgesia and histology of articular tissues. Expression of inflammatory molecules was analyzed in CD4(+) T cells, synovial fibroblasts, and chondrocytes by quantitative real-time PCR. KEY FINDINGS: Oral administration of L. casei together with CII and Gln more effectively reduced pain, cartilage destruction, and lymphocyte infiltration than the treatment of Gln or L. casei alone. This co-administration also decreased expression of various pro-inflammatory cytokines (interleukin-1ß (IL-1ß), IL-2, IL-6, IL-12, IL-17, tumor necrosis factor-α (TNF-α), and interferon-γ (IFN-γ)) and matrix metalloproteinases (MMP1, MMP3, and MMP13), while up-regulating anti-inflammatory cytokines (IL-4 and IL-10). These results are concomitant with reduced translocation of NF-κB into the nucleus and increased expression of the tissue inhibitor of MMP1 (TIMP1) and CII in chondrocytes. SIGNIFICANCE: Our study provides evidence that L. casei could act as a potent nutraceutical modulator for OA treatment by reducing pain, inflammatory responses, and articular cartilage degradation.


Subject(s)
Collagen Type II/agonists , Glucosamine/agonists , Inflammation/therapy , Lacticaseibacillus casei/physiology , Osteoarthritis/therapy , Probiotics/therapeutic use , Animals , CD4-Positive T-Lymphocytes/chemistry , CD4-Positive T-Lymphocytes/drug effects , CD4-Positive T-Lymphocytes/physiology , Chondrocytes/chemistry , Chondrocytes/drug effects , Chondrocytes/physiology , Collagen Type II/physiology , Cytokines/analysis , Female , Glucosamine/physiology , Inflammation/immunology , NF-kappa B/analysis , Osteoarthritis/microbiology , Pain Measurement , Rats , Rats, Wistar , Reverse Transcriptase Polymerase Chain Reaction , Synovial Fluid/cytology , Synovial Fluid/drug effects , Synovial Fluid/physiology
SELECTION OF CITATIONS
SEARCH DETAIL