Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 9 de 9
Filter
1.
Lab Invest ; 102(4): 352-362, 2022 04.
Article in English | MEDLINE | ID: mdl-34789838

ABSTRACT

Programmed death-ligand 1 (PD-L1) is constitutively expressed by hypoxia-inducible factor 2α (HIF2α). It can be induced by interferon gamma (IFNγ) signaling in clear cell renal cell carcinoma (ccRCC). Clinical trials of metastatic ccRCCs have suggested that a canonical IFNγ signature is a better biomarker for therapeutic response to immune checkpoint inhibitors (ICIs) than PD-L1 expression levels in tumor cells. To understand the therapeutic response to ICIs according to PD-L1 expression levels, we analyzed transcriptional regulation of the PD-L1 promoter by HIF2α and IFNγ-inducible interferon regulatory factor-1 (IRF-1) in ccRCC cells. Here, we present two ccRCC cell models showing differential PD-L1 expression levels in response to IFNγ and hypoxia. Analysis of The Cancer Genome Atlas RNA-sequencing data revealed that PD-L1 expression correlated with JAK2 and STAT1 expression of the canonical IFNγ signature in ccRCC tissues. Upon IFNγ stimulation, PD-L1 was induced by sequential activation of JAK2/STAT1/IRF-1 signaling in both WT- and Mut- VHL ccRCC cells. IFNγ activated the IRF-1α site of the PD-L1 promoter. The IFNγ-mediated increase of PD-L1 expression in Mut-VHL cells was 4.8-fold greater than that in WT-VHL cells. Under normoxia condition, PD-L1 expression in Mut-VHL cells was significantly higher than that in WT-VHL cells due to high basal HIF2α expression. Under hypoxia condition, PD-L1 expression in WT-VHL cells was induced up to 1.8-fold through activation of hypoxia-response elements 2 and 3. In contrast, although PD-L1 in Mut-VHL cells was already highly expressed in the basal state through activation of hypoxia-response elements 2, 3, and 4, it was no longer induced by hypoxia. In conclusion, Mut-VHL ccRCC cells displayed higher PD-L1 expression due to high basal HIF2α expression and a stronger response to IFNγ stimulation than WT-VHL cells. The fact that HIF2α antagonists can potentially reduce PD-L1 expression levels should be considered in ICI combination therapy.


Subject(s)
Carcinoma, Renal Cell , Kidney Neoplasms , B7-H1 Antigen/genetics , B7-H1 Antigen/metabolism , Carcinoma, Renal Cell/pathology , Cell Line, Tumor , Female , Gene Expression Regulation, Neoplastic , Humans , Hypoxia , Interferon-gamma/metabolism , Kidney Neoplasms/metabolism , Male , Von Hippel-Lindau Tumor Suppressor Protein/genetics , Von Hippel-Lindau Tumor Suppressor Protein/metabolism
2.
J Immunol ; 190(11): 5747-56, 2013 Jun 01.
Article in English | MEDLINE | ID: mdl-23636060

ABSTRACT

The chemokine receptor CCR3 is expressed in prominent allergic inflammatory cells, including eosinophils, mast cells, and Th2 cells. We previously identified a functional GATA element within exon 1 of the CCR3 gene that is responsible for GATA-1-mediated CCR3 transcription. Because allergic inflammatory cells exhibit distinct expression patterns of different GATA factors, we investigated whether different GATA factors dictate CCR3 transcription in a cell type-specific manner. GATA-2 was expressed in EoL-1 eosinophilic cells, GATA-1 and GATA-2 were expressed in HMC-1 mast cells, and GATA-3 was preferentially expressed in Jurkat cells. Unlike a wild-type CCR3 reporter, reporters lacking the functional GATA element were not active in any of the three cell types, implying the involvement of different GATA factors in CCR3 transcription. RNA interference assays showed that small interfering RNAs specific for different GATA factors reduced CCR3 reporter activity in a cell type-specific fashion. Consistent with these findings, chromatin immunoprecipitation and EMSA analyses demonstrated cell type-specific binding of GATA factors to the functional GATA site. More importantly, specific inhibition of the CCR3 reporter activity by different GATA small interfering RNAs was well preserved in respective cell types differentiated from cord blood; in particular, GATA-3 was entirely responsible for reporter activity in Th2 cells and replaced the role predominantly played by GATA-1 and GATA-2. These results highlight a mechanistic role of GATA factors in which cell type-specific expression is the primary determinant of transcription of the CCR3 gene in major allergic inflammatory cells.


Subject(s)
GATA Transcription Factors/metabolism , Hypersensitivity/genetics , Receptors, CCR3/genetics , Transcription, Genetic , Binding Sites , Cell Differentiation/genetics , Cell Line , Cells, Cultured , Eosinophils/cytology , Eosinophils/metabolism , Fetal Blood/cytology , GATA Transcription Factors/genetics , Gene Expression Regulation , Gene Order , Gene Silencing , Humans , Mast Cells/cytology , Mast Cells/metabolism , Organ Specificity/genetics , Protein Binding , RNA Interference , Th2 Cells/cytology , Th2 Cells/metabolism
3.
J Oral Pathol Med ; 39(7): 533-9, 2010 Aug 01.
Article in English | MEDLINE | ID: mdl-20141577

ABSTRACT

OBJECTIVE: Membranous (M) cell of the human palatine tonsil is an antigen entry site for mucosal infection, but its location is obscure in histological sections. Recently, a microarray analysis has demonstrated that clusterin, annexin A5, CD44, MMP14, and beta-tubulin are candidate genes of M cell marker in mice. Among these genes, we here describe class II beta-tubulin as a new marker for human tonsillar M cells and follicular dendritic cells (FDCs), and present its usefulness for diagnosis of angioimmunoblastic T-cell lymphomas (AILTs). MATERIALS AND METHODS: Immunohistochemistry and Western blotting for class II beta-tubulin were performed using 81 cases of lymphoid, gastrointestinal and thyroid tissues, and an FDC cell line, respectively. Double immunostaining with clusterin and class II beta-tubulin were carried out. RESULTS: Class II beta-tubulin localized the M cells and FDCs in the palatine tonsils (10/10, 100%) and adenoids (10/10, 100%). It was colocalized with clusterin in the palatine tonsils. However, class II beta-tubulin staining did not identify intestinal M cells in the intestines. Immunoblot analysis revealed that class II beta-tubulin expression was upregulated in HK cells, a normal FDC cell line. Class II beta-tubulin immunostaining highlighted hyperplastic FDC meshworks in all AILTs (14/14, 100%). CONCLUSION: Class II beta-tubulin is a specific histochemical marker for human tonsillar M cells and FDCs. Thus, class II beta-tubulin immunostaining may be useful to identify tonsillar M cells and to diagnose FDC proliferative lesions such as AILT.


Subject(s)
Biomarkers, Tumor/analysis , Dendritic Cells, Follicular/pathology , Palatine Tonsil/pathology , Tubulin/analysis , Adenoids/pathology , Appendix/pathology , Blotting, Western , Castleman Disease/diagnosis , Castleman Disease/pathology , Cell Line , Cell Line, Tumor , Choristoma/pathology , Clusterin/analysis , Gastric Mucosa/pathology , Gastritis/microbiology , Gastritis/pathology , Hashimoto Disease/pathology , Helicobacter Infections/pathology , Helicobacter pylori , Humans , Hyperplasia , Ileum/pathology , Immunohistochemistry , Intestinal Mucosa/pathology , Lymph Nodes/pathology , Lymphoid Tissue/pathology , Lymphoma, T-Cell/diagnosis , Lymphoma, T-Cell/pathology , Thyroid Gland/pathology , Up-Regulation
4.
Pharmacol Rep ; 69(6): 1140-1144, 2017 Dec.
Article in English | MEDLINE | ID: mdl-29128792

ABSTRACT

BACKGROUND: Recently, a peptide vaccine (B4T) was developed that prevents high fat diet (HFD)-induced obesity and liver steatosis in wild type mice and appears to target an epitope present in ApoB100 but not ApoB48. Here, we ask whether B4T remains effective in ApoE knockout (ApoE-ko) mice, which exhibit a greatly increased ApoB48/ApoB100 ratio and develop atherosclerosis under HFD. METHODS: HFD-fed male ApoE-ko mice were injected with B4T or vehicle 3 times between 5 and 15 weeks of age. Until 45 weeks of age, they were regularly weighed and antibody titers determined. In the end, adiposity and organ histologies were examined. RESULTS: We find that in the ApoE-ko mice, B4T prevents HFD-induced body weight increases (p<0.01) to a comparable degree as previously shown in wild type mice. Also, liver steatosis was prevented as previously shown in wild type mice. By contrast, atherosclerotic plaque formation was not prevented in any of the vaccinated mice studied, in line with the observation that antibody production paralleled the weight reduction but largely preceded atherogenesis. CONCLUSION: The findings demonstrate effectiveness of B4T despite the increased ApoB48/B100 ratio, but argue against an effect on de novo plaque formation. At least under the current vaccination schedule, the obesity- and atherosclerosis-related roles of ApoB appear to be dissociable.


Subject(s)
Apolipoprotein B-100/immunology , Fatty Liver/prevention & control , Obesity/prevention & control , Peptides/administration & dosage , Vaccines/administration & dosage , Animals , Apolipoproteins E/genetics , Atherosclerosis/prevention & control , Diet, High-Fat/adverse effects , Disease Models, Animal , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Peptides/immunology , Peptides/pharmacology , Plaque, Atherosclerotic/prevention & control , Vaccines/immunology , Vaccines/pharmacology
5.
Sci Rep ; 7(1): 17810, 2017 12 19.
Article in English | MEDLINE | ID: mdl-29259270

ABSTRACT

Programmed death-ligand 1 (PD-L1) acts as an immune checkpoint inhibitor in various cancers. PD-L1 is known to be more frequently expressed in EBV (+) gastric cancer (GC). However, the mechanisms underlying the regulation of PD-L1 expression in EBV (+) GC remain unclear. We investigated the basal and inducible PD-L1 expressions in GC cells. PD-L1 expression was upregulated upon treatment with IFNγ in both EBV (-) and EBV (+) GC cells. Upon stimulation with the same concentration of IFNγ for 24 h, EBV (+) SNU-719 cells showed dramatically higher PD-L1 expression levels by activating JAK2/STAT1/IRF-1 signaling than those of EBV (-) AGS cells. PD-L1 promoter assays, chromatin immunoprecipitation, and electrophoretic mobility shift assays revealed that IFNγ-inducible PD-L1 overexpression is primarily mediated by the putative IRF-1α site of the PD-L1 promoter in EBV (+) SNU-719 cells. Moreover, EBNA1 knockdown reduced both constitutive and IFNγ-inducible PD-L1 promoter activity by decreasing the transcript and protein levels of JAK2 and subsequently STAT1/IRF-1/PD-L1 signaling. EBNA1 is suggested to be moderately enhance both constitutive and IFNγ-inducible PD-L1 expression in EBV (+) GC cells. Thus, the signaling proteins and EBNA1 that regulate PD-L1 expression are potential therapeutic targets in EBV (+) GC.


Subject(s)
Interferon-gamma/genetics , Proteins/genetics , Signal Transduction/genetics , Stomach Neoplasms/genetics , B7-H1 Antigen/genetics , Cell Line, Tumor , Epstein-Barr Virus Infections/genetics , Epstein-Barr Virus Infections/virology , Gene Expression Regulation, Neoplastic/genetics , Herpesvirus 4, Human/pathogenicity , Humans , Interferon Regulatory Factor-1/genetics , Janus Kinase 2/genetics , STAT1 Transcription Factor/genetics , Stomach Neoplasms/virology
6.
Immune Netw ; 16(3): 176-82, 2016 Jun.
Article in English | MEDLINE | ID: mdl-27340386

ABSTRACT

CCR3 is a chemokine receptor that mediates the accumulation of allergic inflammatory cells, including eosinophils and Th2 cells, at inflamed sites. The regulatory sequence of the CCR3 gene, contains two Runt-related transcription factor (RUNX) 1 sites and two PU.1 sites, in addition to a functional GATA site for transactivation of the CCR3 gene. In the present study, we examined the effects of the cis-acting elements of RUNX1 and PU.1 on transcription of the gene in EoL-1 eosinophilic cells and Jurkat T cells, both of which expressed functional surface CCR3 and these two transcription factors. Introduction of RUNX1 siRNA or PU.1 siRNA resulted in a modest decrease in CCR3 reporter activity in both cell types, compared with transfection of GATA-1 siRNA. Cotransfection of the two siRNAs led to inhibition in an additive manner. EMSA analysis showed that RUNX1, in particular, bound to its binding motifs. Mutagenesis analysis revealed that all point mutants lacking RUNX1- and PU.1-binding sites exhibited reduced reporter activities. These results suggest that RUNX1 and PU.1 participate in transcriptional regulation of the CCR3 gene.

7.
J Leukoc Biol ; 100(4): 711-723, 2016 10.
Article in English | MEDLINE | ID: mdl-27154355

ABSTRACT

Oligodendrocyte transcription factor 2, a basic helix-loop-helix transcription factor that binds to E-box motifs, is known to have a key role in determining lineage specification of oligodendrocytes and motor neurons. In the present study, we report that oligodendrocyte transcription factor 2 is expressed in human eosinophils and involved in transcriptional activation of the gene encoding sialic acid binding immunoglobulin-like lectin 8 (Siglec-8), a late eosinophil-differentiation marker known to exert eosinophil apoptosis. When cord blood CD34+ hematopoietic stem cells differentiated toward eosinophils during a 24-d culture period, oligodendrocyte transcription factor 2 protein was expressed in cord blood eosinophils on d 24, a time when cord blood eosinophils are considered fully differentiated, whereas it was not detectable on d 18 or at earlier time points. Oligodendrocyte transcription factor 2 protein was also abundantly expressed in human peripheral-blood eosinophils but not in neutrophils, monocytes, lymphocytes, or cord blood mast cells. RNA sequencing analysis showed that numerous genes, especially those encoding eosinophil surface molecules, were highly up-regulated along with OLIG2 Among the genes examined, SIGLEC-8 messenger RNA and protein were markedly down-regulated in parallel with OLIG2 by an oligodendrocyte transcription factor 2 small interfering RNA or a short hairpin RNA, as evidenced by real-time polymerase chain reaction, fluorescence-activated cell sorting, and Western blot analyses. In reporter gene and chromatin immunoprecipitation experiments, an E-box in the first intron was found to stimulate SIGLEC-8 gene transcription and to bind oligodendrocyte transcription factor 2. Hence, at least one important aspect of eosinophil differentiation is regulated by oligodendrocyte transcription factor 2, a transcription factor that has not previously been reported, to our knowledge, in normal granulocytes.


Subject(s)
Antigens, CD/biosynthesis , Antigens, Differentiation, B-Lymphocyte/biosynthesis , Basic Helix-Loop-Helix Transcription Factors/physiology , Eosinophils/cytology , Gene Expression Regulation, Developmental , Lectins/biosynthesis , Myelopoiesis/genetics , Nerve Tissue Proteins/physiology , Antigens, CD/genetics , Antigens, Differentiation, B-Lymphocyte/genetics , Basic Helix-Loop-Helix Transcription Factors/biosynthesis , Basic Helix-Loop-Helix Transcription Factors/genetics , Cells, Cultured , E-Box Elements/genetics , Eosinophils/metabolism , Fetal Blood/cytology , Genes, Reporter , Hematopoietic Stem Cells/cytology , Humans , Hypersensitivity, Immediate/blood , Introns/genetics , Lectins/genetics , Mast Cells/metabolism , Membrane Proteins/biosynthesis , Membrane Proteins/genetics , Nerve Tissue Proteins/biosynthesis , Nerve Tissue Proteins/genetics , Neutrophils/metabolism , Oligodendrocyte Transcription Factor 2 , RNA Interference , RNA, Messenger/biosynthesis , RNA, Small Interfering/genetics , Sequence Analysis, RNA
8.
Exp Mol Med ; 48(1): e202, 2016 01 08.
Article in English | MEDLINE | ID: mdl-27534531

ABSTRACT

Aspirin hypersensitivity is a hallmark of aspirin-exacerbated respiratory disease (AERD), a clinical syndrome characterized by the severe inflammation of the respiratory tract after ingestion of cyclooxygenase-1 inhibitors. We investigated the capacity of aspirin to induce interleukin-4 (IL-4) production in inflammatory cells relevant to AERD pathogenesis and examined the associated biochemical and molecular pathways. We also compared IL-4 production in peripheral blood mononuclear cells (PBMCs) from patients with AERD vs aspirin-tolerant asthma (ATA) upon exposure to aspirin. Aspirin induced IL-4 expression and activated the IL-4 promoter in a report assay. The capacity of aspirin to induce IL-4 expression correlated with its activity to activate mitogen-activated protein kinases, to form DNA-protein complexes on P elements in the IL-4 promoter and to synthesize nuclear factor of activated T cells, critical transcription factors for IL-4 transcription. Of clinical importance, aspirin upregulated IL-4 production twice as much in PBMCs from patients with AERD compared with PBMCs from patients with ATA. Our results suggest that IL-4 is an inflammatory component mediating intolerance reactions to aspirin, and thus is crucial for AERD pathogenesis.


Subject(s)
Aspirin/adverse effects , Cyclooxygenase Inhibitors/adverse effects , Inflammation/chemically induced , Interleukin-4/immunology , Respiration Disorders/chemically induced , Cell Line , Humans , Inflammation/genetics , Inflammation/immunology , Interleukin-4/genetics , RNA, Messenger/genetics , Respiration Disorders/genetics , Respiration Disorders/immunology , Up-Regulation/drug effects
9.
Cancer Lett ; 269(1): 37-45, 2008 Sep 28.
Article in English | MEDLINE | ID: mdl-18502033

ABSTRACT

Tissue inhibitor of metalloproteinase-1 (TIMP1) is a survival factor of germinal center (GC) B cells, and its over-expression is correlated with aggressive B cell lymphomas and classical Hodgkin lymphomas. We previously demonstrated that TIMP1 down-regulates B-cell receptor and BCL6, and activates interleukins-6,-10 (ILs)/signal transducer and activator of transcription-3 (STAT3) signaling in GC B cells. The activation of ILs/STAT3 signaling can amplify CD44 function, and vice versa, and induce protein-tyrosine phosphatase SHP1 activity by a negative feedback mechanism. Here, we show that TIMP1 up-regulates cell surface CD44 (standard and variants 3 and 7-10) and induces the activity and nuclear localization of SHP1 in an Epstein Barr virus (EBV)-negative Burkitt lymphoma cell line, the neoplastic counterpart of GC centroblasts. These results suggest that TIMP1 functions as a differentiating and survival factor of GC B cells by modulating CD44 and SHP1 in the late centrocyte/post-GC stage, regardless of EBV infection.


Subject(s)
Cell Differentiation , Cell Nucleus/metabolism , Enterocytes/cytology , Germinal Center/cytology , Hyaluronan Receptors/biosynthesis , Protein Tyrosine Phosphatase, Non-Receptor Type 6/metabolism , Tissue Inhibitor of Metalloproteinase-1/physiology , Active Transport, Cell Nucleus , Cells, Cultured , Humans , Hyaluronan Receptors/analysis , Interferon Regulatory Factors/analysis , Protein Tyrosine Phosphatase, Non-Receptor Type 6/analysis , Tissue Inhibitor of Metalloproteinase-1/analysis
SELECTION OF CITATIONS
SEARCH DETAIL