Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 380
Filter
1.
J Leukoc Biol ; 108(3): 859-866, 2020 09.
Article in English | MEDLINE | ID: mdl-32480423

ABSTRACT

Mast cell activation through the high-affinity IgE receptor (FcεRI) plays a central role in allergic reactions. FcεRI-mediated activation triggers multiple signaling pathways leading to degranulation and synthesis of different inflammatory mediators. IgE-mediated mast cell activation can be modulated by different molecules, including several drugs. Herein, we investigated the immunomodulatory activity of the histone deacetylase inhibitor valproic acid (VPA) on IgE-mediated mast cell activation. To this end, bone marrow-derived mast cells (BMMC) were sensitized with IgE and treated with VPA followed by FcεRI cross-linking. The results indicated that VPA reduced mast cell IgE-dependent degranulation and cytokine release. VPA also induced a significant reduction in the cell surface expression of FcεRI and CD117, but not other mast cell surface molecules. Interestingly, VPA treatment inhibited the phosphorylation of PLCγ2, a key signaling molecule involved in IgE-mediated degranulation and cytokine secretion. However, VPA did not affect the phosphorylation of other key components of the FcεRI signaling pathway, such as Syk, Akt, ERK1/2, or p38. Altogether, our data demonstrate that VPA affects PLCγ2 phosphorylation, which in turn decreases IgE-mediated mast cell activation. These results suggest that VPA might be a key modulator of allergic reactions and might be a promising therapeutic candidate.


Subject(s)
Histone Deacetylase Inhibitors/pharmacology , Immunoglobulin E/immunology , Mast Cells/drug effects , Phospholipase C gamma/antagonists & inhibitors , Receptors, IgE/drug effects , Valproic Acid/pharmacology , Animals , Cell Degranulation/drug effects , Down-Regulation/drug effects , Interleukin-13/metabolism , Interleukin-6/metabolism , Mast Cells/cytology , Mice , Phospholipase C gamma/physiology , Receptors, IgE/biosynthesis , Receptors, IgE/genetics , Tumor Necrosis Factor-alpha/metabolism
2.
Hum Pathol ; 89: 71-80, 2019 07.
Article in English | MEDLINE | ID: mdl-31054894

ABSTRACT

Mantle cell lymphoma (MCL) is usually CD23 negative, a feature helpful in distinguishing MCL from chronic lymphocytic leukemia/small lymphocytic lymphoma. However, a subset of MCL cases can be CD23+. Limited data are available regarding the clinicopathological features and prognosis of patients with CD23+ MCL. In this study, we reviewed 798 cases of MCL and identified 103 (13%) that were CD23+ by flow cytometry, all of which were positive for cyclin D1 and/or associated with CCND1/IGH. In all cases of CD23+ MCL, CD23 expression was dim partial or dim, unlike moderate to bright CD23 expression observed in chronic lymphocytic leukemia/small lymphocytic lymphoma. The clinicopathological features and outcome of patients with CD23+ MCL were compared with 240 patients with typical MCL negative for CD23. Patients with CD23+ MCL more often had an elevated leukocyte count (33% versus 18%, P = .009), bone marrow involvement (89% versus 78%, P = .02), stage 4 disease (87% versus 77%, P = .03), and a leukemic presentation (42% versus 11%, P = .0001). CD23+ MCL was also more often positive for CD200 (17% versus. 4.6%, P = .0005) and less commonly positive for SOX11 (55% versus. 74%, P = .027). All other clinicopathological features were similar. With similar treatment regimens and observation times, patients with CD23+ MCL had a significant better overall survival (P = .02) and progression-free survival (P = .029). In conclusion, CD23 expression was observed in 13% of MCL cases and is associated with a better prognosis in patients with MCL. CD23 is associated with leukocytosis, a leukemic presentation, bone marrow involvement, CD200 expression, and a lower frequency of SOX11 positivity.


Subject(s)
Lymphoma, Mantle-Cell/pathology , Receptors, IgE/biosynthesis , Adult , Aged , Aged, 80 and over , Antigens, CD/biosynthesis , Biomarkers, Tumor/analysis , Female , Humans , Immunophenotyping , Lymphoma, Mantle-Cell/metabolism , Lymphoma, Mantle-Cell/mortality , Male , Middle Aged , Prognosis , Progression-Free Survival , Retrospective Studies
3.
Stem Cells ; 37(3): 430-440, 2019 03.
Article in English | MEDLINE | ID: mdl-30537419

ABSTRACT

Previously, we reported that although the HSPC frequency in bone marrow cells (BMC) was comparable between ß2-/- and ß2+/+ mice, transplantation of ß2-/- BMC into lethally irradiated CD45.1 recipient resulted in more myeloid cell production than ß2+/+ BMC. The objective of this study is to address if integrin ß2 deficiency skews granulocyte/macrophage progenitor (GMP) proliferation. FACS analysis demonstrated that GMP frequency and cell number were higher and megakaryocyte/erythrocyte progenitor frequency and cell number were lower in ß2-/- mice than ß2+/+ mice. However, the common myeloid progenitors (CMP) frequency and cell number were similar between the two groups. The increased GMP number was due to GMP proliferation as evidenced by the percentage of BrdU-incorporating GMP. Whole genome transcriptome analysis identified increased FcεRIα expression in ß2-/- CMP compared to ß2+/+ CMP. FcεRIα expression on ß2-/- GMP was detected increased in ß2-/- mice by qRT-PCR and FACS. Although transplantation of FcεRIαhi GMP or FcεRIαlo GMP into lethally irradiated CD45.1 recipient resulted in comparable myeloid cell production, transplantation of ß2 deficient FcεRIαhi GMP generated more myeloid cells than ß2+/+ FcεRIαhi GMP. GATA2 expression was increased in ß2-/- GMP. Using a luciferase reporter assay, we demonstrated that mutation of the GATA2 binding site in the FcεRIα promoter region diminished FcεRIα transcription. In vitro, the addition of IgE, the ligand of FcεRIα, promoted GMP expansion, which was abrogated by inhibition of JNK phosphorylation. Integrin ß2 deficiency promoted GMP proliferation and myeloid cell production, which was mediated via FcεRIα/IgE-induced JNK phosphorylation in GMP. Stem Cells 2019;37:430-440.


Subject(s)
CD18 Antigens/metabolism , Cell Proliferation , Granulocyte-Macrophage Progenitor Cells/metabolism , Animals , CD18 Antigens/genetics , GATA2 Transcription Factor/genetics , GATA2 Transcription Factor/metabolism , Gene Expression Regulation , MAP Kinase Kinase 4 , Mice , Mice, Knockout , Receptors, IgE/biosynthesis , Receptors, IgE/genetics , Transcription, Genetic
5.
J Cell Biochem ; 119(5): 4142-4149, 2018 05.
Article in English | MEDLINE | ID: mdl-29243845

ABSTRACT

Despite large number of investigations, the etiology of chronic rhinosinusitis (CRS) remains unclear. Several factors are likely involved in its onset. The genetic susceptibility of IgE-responsiveness likely caused by polymorphism(s) in high affinity receptor for IgE (FcɛR1α) gene can help in understanding the pathophysiology of CRS with nasal polyposis (CRSwNP). A population-based case-control association analysis was conducted to assess the risk of CRSwNP conferred by single nucleotide polymorphisms (SNPs) in FcɛR1α gene in a North Indian cohort. Two promoter and three exonic regions of FcɛR1α gene were amplified and sequenced to investigate five SNPs: rs2427827, rs2251746, rs2298804, rs2298805, and rs2269718. BLAST analysis and subsequent multiple alignments, with known sequences available in the NCBI database, were performed. Total serum IgE and FcɛR1α antibody levels were estimated. Patient IgE level of 461.22 ± 436.43 in comparison to 83.62 ± 58.043 IU/mL in controls (P < 0.0001), and FcɛR1α antibody level of 292.38 ± 115.27 in comparison to 160.56 ± 105.9 in controls (P < 0.0001), depicts their highly significant associations with CRSwNP disease. However, no SNP showed evidence of association with CRSwNP; although relatively higher Odds ratios were observed with rs2427827, rs2251746, and rs2298804. Patient stratification revealed a significant association (P < 0.05) of rs2427827 SNP with high IgE level CRSwNP patients. Nonetheless, we found no SNP associated with low serum IgE level patients. SNP (rs2427827) in the FcɛR1α gene region and high IgE levels may confer susceptibility to CRSwNP in north Indian population. However, further studies including larger sample size, gene-gene, and gene-environment interactions are required for its elucidation.


Subject(s)
Immunoglobulin E/blood , Nasal Polyps , Polymorphism, Single Nucleotide , Receptors, IgE , Rhinitis , Sinusitis , Adult , Chronic Disease , Female , Humans , Male , Nasal Polyps/blood , Nasal Polyps/genetics , Nasal Polyps/pathology , Receptors, IgE/biosynthesis , Receptors, IgE/genetics , Rhinitis/blood , Rhinitis/genetics , Rhinitis/pathology , Sinusitis/blood , Sinusitis/genetics , Sinusitis/pathology
6.
J Immunol ; 198(1): 229-238, 2017 01 01.
Article in English | MEDLINE | ID: mdl-27852746

ABSTRACT

Basophils represent <1% of circulating leukocytes. They play a crucial role during allergy and helminth-induced Th2 responses. However, recent data also suggest a contribution to the pathogenesis of autoimmune diseases. Basophils from patients with systemic lupus erythematosus show an activated phenotype, correlating to disease activity. Furthermore, murine basophils or their mediators enhance memory responses and plasma cell (PC) survival, suggesting that they directly modulate the function of B cells. This is highly relevant with respect to human allergy and autoimmunity because a possible modulation of B cell differentiation by basophils could point to new therapeutic targets. Therefore, the interaction between human B cells and basophils and the mechanism underlying this interaction were investigated in detail. Using two different methods to induce PC differentiation, we found that human basophils enhance B cell proliferation, class switching, differentiation into PC, maturation of PC, and production of Igs, especially IgG. Basophil supernatants enhanced the expression of the B cell markers CD23 and CD40, which are important for B cell differentiation into IgG-producing PC. This was mainly IL-4 dependent. IL-3 amplified the number of PC in vitro, and acted synergistically with basophils in enhancing Ab production. Thus, human basophils modulate B cell differentiation into Ab-producing PC. Their contribution as modulators and effectors during allergy and autoimmunity should be considered when designing new therapeutic options.


Subject(s)
Basophils/immunology , Cell Differentiation/immunology , Lymphocyte Activation/immunology , Plasma Cells/cytology , CD40 Antigens/biosynthesis , Cells, Cultured , Enzyme-Linked Immunosorbent Assay , Enzyme-Linked Immunospot Assay , Flow Cytometry , Humans , Receptors, IgE/biosynthesis , Reverse Transcriptase Polymerase Chain Reaction
7.
Int J Mol Med ; 37(2): 501-8, 2016 Feb.
Article in English | MEDLINE | ID: mdl-26707911

ABSTRACT

Arctium lappa fruit has been used in traditional medicine, and it is known to exert beneficial effects, such as antioxidant, anti-inflammatory and anticancer effects. However, the effects of the Arctium lappa fruit on the allergic response remain unknown. In this study, we evaluated the anti-allergic effects of Arctium lappa fruit extract (AFE) and its fermented form (F-AFE) using immunoglobulin E (IgE)-activated RBL­2H3 cells. To investigate the anti-allergic effects of AFE or F-AFE, we examined the release of ß-hexosaminidase, a key biomarker of degranulation during an allergic reaction, and the production of pro-inflammatory mediators, such as tumor necrosis factor-α (TNF-α) and prostaglandin E2 (PGE2) in the cells treated with or without the above-mentioned extracts. AFE weakly inhibited the release of ß-hexosaminidase, whereas F-AFE significantly suppressed the release of ß-hexosaminidase in a dose-dependent manner. Consistently, F-AFE suppressed the production of TNF-α and PGE2 in a dose-dependent manner. F-AFE exerted an inhibitory effect on the production of ß-hexosaminidase, TNF-α and PGE2 with an IC50 value of 30.73, 46.96 and 36.27 µg/ml, respectively. Furthermore, F-AFE inhibited the phosphorylation of Lyn, Fyn and Syk, which are involved in the FcεRI signaling pathway, that of phosphoinositide phospholipase C (PLC)γ1/2 and protein kinase C (PKC)δ, which are associated with the degranulation process, as well as that of extracellular signal-regulated kinase 1/2 (ERK1/2), c-Jun N-terminal kinase 1/2 (JNK), p38 and Akt, which are associated with cytokine expression. In the late phase, F-AFE partially suppressed the phosphorylation of cytosolic phospholipase A2 (cPLA2), but not the expression of cyclooxygenase (COX)-2. To compare and identify the major components of the two extracts, we used high-performance liquid chromatography. The levels of arctigenin, one of the major compounds, were elevated 6-fold in F-AFE compared with AFE, whereas the levels of arctiin, an arctigenin glycoside, were decreased in F-AFE by approximately 57.40%. These results suggest that arctigenin plays an important role in the anti-allergic effects of F-AFE. Taken together, F-AFE containing anti-allergic phytochemicals, including arctigenin, inhibited the activation of the FcεRI receptor induced by the antigen­IgE complex. Such effects may provide further information for the development of a phytomedicine for allergic diseases.


Subject(s)
Hypersensitivity/drug therapy , Immunoglobulin E/biosynthesis , Plant Extracts/administration & dosage , Receptors, IgE/biosynthesis , Arctium/chemistry , Fermentation , Fruit/chemistry , Gene Expression Regulation/drug effects , Humans , Hypersensitivity/immunology , Hypersensitivity/pathology , Immunoglobulin E/immunology , Intracellular Signaling Peptides and Proteins/biosynthesis , Plant Extracts/chemistry , Protein-Tyrosine Kinases/biosynthesis , Proto-Oncogene Proteins c-fyn/biosynthesis , Receptors, IgE/immunology , Signal Transduction/drug effects , Syk Kinase , Tumor Necrosis Factor-alpha , src-Family Kinases/biosynthesis
8.
J Immunol ; 195(7): 3427-35, 2015 Oct 01.
Article in English | MEDLINE | ID: mdl-26297757

ABSTRACT

FcεRI, which is composed of α, ß, and γ subunits, plays an important role in IgE-mediated allergic responses. TGF-ß1 has been reported to suppress FcεRI and stem cell factor receptor c-Kit expression on mast cell surfaces and to suppress mast cell activation induced by cross-linking of FcεRI. However, the molecular mechanism by which these expressions and activation are suppressed by TGF-ß1 remains unclear. In this study, we found that the expression of Ets homologous factor (Ehf), a member of the Ets family transcriptional factors, is upregulated by TGF-ß/Smad signaling in mouse bone marrow-derived mast cells (BMMCs). Forced expression of Ehf in BMMCs repressed the transcription of genes encoding FcεRIα, FcεRIß, and c-Kit, resulting in a reduction in cell surface FcεRI and c-Kit expression. Additionally, forced expression of Ehf suppressed FcεRI-mediated degranulation and cytokine production. Ehf inhibited the promoter activity of genes encoding FcεRIα, FcεRIß, and c-Kit by binding to these gene promoters. Furthermore, the mRNA levels of Gata1, Gata2, and Stat5b were lower in BMMCs stably expressing Ehf compared with control cells. Because GATA-1 and GATA-2 are positive regulators of FcεRI and c-Kit expression, decreased expression of GATAs may be also involved in the reduction of FcεRI and c-Kit expression. Decreased expression of Stat5 may contribute to the suppression of cytokine production by BMMCs. In part, mast cell response to TGF-ß1 was mimicked by forced expression of Ehf, suggesting that TGF-ß1 suppresses FcεRI and c-Kit expression and suppresses FcεRI-mediated activation through upregulation of Ehf.


Subject(s)
Proto-Oncogene Proteins c-kit/biosynthesis , Receptors, IgE/immunology , Transcription Factors/immunology , Transforming Growth Factor beta1/metabolism , Animals , Bone Marrow Cells , Cell Degranulation/immunology , Cells, Cultured , Cytokines/biosynthesis , GATA1 Transcription Factor/biosynthesis , GATA1 Transcription Factor/genetics , GATA2 Transcription Factor/biosynthesis , GATA2 Transcription Factor/genetics , Immunoglobulin E/immunology , Mast Cells/immunology , Mice , Mice, Inbred BALB C , Promoter Regions, Genetic/genetics , Proto-Oncogene Proteins c-kit/genetics , RNA Interference , RNA, Messenger/metabolism , RNA, Small Interfering , Receptors, IgE/biosynthesis , STAT5 Transcription Factor/biosynthesis , STAT5 Transcription Factor/genetics , Signal Transduction/immunology , Smad Proteins/metabolism , Transcription Factors/biosynthesis , Transcription, Genetic/genetics , Transcriptional Activation
9.
J Immunol Res ; 2015: 351829, 2015.
Article in English | MEDLINE | ID: mdl-25973435

ABSTRACT

Mast cells (MCs) are versatile effector and regulatory cells in various physiologic, immunologic, and pathologic processes. In addition to the well-characterized IgE/FcεRI-mediated degranulation, a variety of biological substances can induce MCs activation and release of their granule content. Sex steroids, mainly estradiol and progesterone, have been demonstrated to elicit MCs activation. Most published studies have been conducted on MCs lines or freshly isolated peritoneal and bone marrow-derived MC without addressing gender impact on MC response. Our goal was to investigate if the effect of estradiol, progesterone, testosterone, and dihydrotestosterone (DHT) on MCs may differ depending on whether female or male rats are used as MCs donors. Our results demonstrated that effect of sex steroids on MCs histamine release is dose- and gender-dependent and can be direct, synergistic, or inhibitory depending on whether hormones are used alone or to pretreat MCs followed by substance P-stimulation or upon IgE-mediated stimulation. In contrast, sex steroids did not have effect on the MC expression of the IgE high affinity receptor, FcεRI, no matter female or male rats were used. In conclusion, MCs degranulation is modulated by sex hormones in a gender-selective fashion, with MC from females being more susceptible than MC from males to the effects of sex steroids.


Subject(s)
Gonadal Steroid Hormones/pharmacology , Histamine Release/drug effects , Mast Cells/immunology , Peritoneum/cytology , Receptors, IgE/biosynthesis , Animals , Cell Degranulation/immunology , Cells, Cultured , Dihydrotestosterone/pharmacology , Estradiol/pharmacology , Female , Immunoglobulin E/immunology , Male , Mast Cells/cytology , Progesterone/pharmacology , Rats , Rats, Sprague-Dawley , Sex Factors , Substance P/metabolism , Testosterone/pharmacology
10.
J Immunol ; 194(1): 364-78, 2015 Jan 01.
Article in English | MEDLINE | ID: mdl-25452561

ABSTRACT

Helminth parasites provoke multicellular immune responses in their hosts that can suppress concomitant disease. The gut lumen-dwelling tapeworm Hymenolepis diminuta, unlike other parasites assessed as helminth therapy, causes no host tissue damage while potently suppressing murine colitis. With the goal of harnessing the immunomodulatory capacity of infection with H. diminuta, we assessed the putative generation of anti-colitic regulatory B cells following H. diminuta infection. Splenic CD19(+) B cells isolated from mice infected 7 [HdBc(7(d))] and 14 d (but not 3 d) previously with H. diminuta and transferred to naive mice significantly reduced the severity of dinitrobenzene sulfonic acid (DNBS)-, oxazolone-, and dextran-sodium sulfate-induced colitis. Mechanistic studies with the DNBS model, revealed the anti-colitic HdBc(7(d)) was within the follicular B cell population and its phenotype was not dependent on IL-4 or IL-10. The HdBc(7(d)) were not characterized by increased expression of CD1d, CD5, CD23, or IL-10 production, but did spontaneously, and upon LPS plus anti-CD40 stimulation, produce more TGF-ß than CD19(+) B cells from controls. DNBS-induced colitis in RAG1(-/-) mice was inhibited by administration of HdBc(7(d)), indicating a lack of a requirement for T and B cells in the recipient; however, depletion of macrophages in recipient mice abrogated the anti-colitic effect of HdBc(7(d)). Thus, in response to H. diminuta, a putatively unique splenic CD19(+) B cell with a functional immunoregulatory program is generated that promotes the suppression of colitis dominated by TH1, TH2, or TH1-plus-TH2 events, and may do so via the synthesis of TGF-ß and the generation of, or cooperation with, a regulatory macrophage.


Subject(s)
B-Lymphocytes/immunology , Colitis/immunology , Hymenolepiasis/immunology , Hymenolepis diminuta/immunology , Macrophages/immunology , Animals , Antigens, CD19/biosynthesis , Antigens, CD1d/biosynthesis , Benzenesulfonates , CD40 Antigens/immunology , CD5 Antigens/biosynthesis , Colitis/chemically induced , Colitis/therapy , Dextran Sulfate , Homeodomain Proteins/genetics , Hymenolepiasis/parasitology , Immunomodulation/immunology , Immunotherapy , Interleukin-10/biosynthesis , Interleukin-10/immunology , Interleukin-4/immunology , Lipopolysaccharides , Male , Mast Cells/immunology , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Knockout , Oxazolone , Receptors, IgE/biosynthesis , Th1 Cells/immunology , Th2 Cells/immunology , Transforming Growth Factor beta/biosynthesis
11.
Immunology ; 143(2): 230-40, 2014 Oct.
Article in English | MEDLINE | ID: mdl-24750112

ABSTRACT

The IgE Fcε3 domain is an active immunotherapeutic target for asthma and other allergic diseases. However, previous methods for preparing IgE fusion protein vaccines are complex. Antigen 43 (Ag43) is a surface protein found in Escherichia coli that contains α and ß subunits (the α subunit contains multiple T epitopes). Here we constructed a novel Ag43 surface display system (Ag43 system) to express Ag43 chimeric proteins to disrupt immune tolerance against IgE. The Ag43 system was constructed from the E. coli strain Tan109, in which the Ag43 gene was deleted and a recombinant plasmid (pETAg43) expressing a partial Ag43 gene was introduced. The Fcε3 domain of the IgE gene was then subcloned into plasmid pETAg43, resulting in a recombinant plasmid pETAg43/Fcε3, which was used to transform Tan109 for Ag43/Fcε3 surface expression. Thereafter, Ag43/Fcε3 was investigated as an asthma vaccine in a mouse model. Ag43/Fcε3 was expressed on and could be separated from the bacterial surface by heating to 60° while retaining activity. Ag43/Fcε3, as a protein vaccine, produced neutralizing autoantibodies to murine IgE, induced significant anti-asthma effects, and regulated IgE and T helper cytokines in a murine asthma model. Data show that Ag43/Fcε3 chimeric protein is a potential model vaccine for asthma treatment, and that the Ag43 system may be an effective tool for novel vaccine preparation to break immune tolerance to other self-molecules.


Subject(s)
Adhesins, Escherichia coli/immunology , Asthma/prevention & control , Bronchial Hyperreactivity/prevention & control , Receptors, IgE/immunology , Vaccines, Synthetic/immunology , Adhesins, Escherichia coli/biosynthesis , Adhesins, Escherichia coli/genetics , Adoptive Transfer , Animals , Antibodies, Neutralizing/blood , Asthma/blood , Asthma/immunology , Asthma/physiopathology , Autoantibodies/blood , Bronchial Hyperreactivity/blood , Bronchial Hyperreactivity/immunology , Bronchial Hyperreactivity/physiopathology , Bronchoconstriction , Cells, Cultured , Cloning, Molecular , Cytokines/metabolism , Disease Models, Animal , Histamine/metabolism , Immune Tolerance , Immunoglobulin E/immunology , Male , Mice , Mice, Inbred BALB C , Molecular Sequence Data , Ovalbumin/immunology , Receptors, IgE/biosynthesis , Receptors, IgE/genetics , Recombinant Fusion Proteins/immunology , T-Lymphocytes, Helper-Inducer/immunology , Time Factors , Vaccines, Synthetic/biosynthesis , Vaccines, Synthetic/genetics
13.
Protein Expr Purif ; 96: 1-7, 2014 Apr.
Article in English | MEDLINE | ID: mdl-24468271

ABSTRACT

Secretory human interleukin 4 (hIL4) is an N-glycosylated pleiotropic cytokine. It is unknown if these N-linked glycans are required and essential for hIL4 protein stability, expression, secretion, and activity in vivo, and hIL4 expressed from Pichia pastoris yeast has not been tested to date. In this study, we successfully expressed human hIL4 in P. pastoris, the methylotrophic yeast, with a yield of 15.0mg/L. Using the site-directed mutagenesis technique, we made two mutant hIL4 cDNA clones (N38A and N105L) and subsequently expressed them in P. pastoris to analyze the relevant function of each N-glycosylation site on hIL4. Our results demonstrate that the glycosylation only occurs at position Asn38, but not Asn105. The glycosylated form of hIL4 unexpectedly has lower biological activity and lower stability when compared to its non-glycosylated form. The implications of this are discussed.


Subject(s)
Interleukin-4/genetics , Pichia/genetics , Pichia/metabolism , Recombinant Proteins/genetics , Cloning, Molecular , Gene Expression , Glycoproteins/genetics , Glycoproteins/metabolism , Glycosylation , Humans , Interleukin-4/biosynthesis , Interleukin-4/metabolism , Mutagenesis, Site-Directed , Receptors, IgE/biosynthesis
14.
J Korean Med Sci ; 29(1): 43-7, 2014 Jan.
Article in English | MEDLINE | ID: mdl-24431904

ABSTRACT

Increased FcεR1α expression with upregulated CD203c expression on peripheral basophils is seen in patients with chronic urticaria (CU). However, there has been no published report on the association between CD203c expression level and clinical disease activity in CU patients. To investigate whether the increase of basophil activation is associated with the disease activity of CU, we measured basophil CD203c expression using a tricolor flow cytometric method in 82 CU patients and 21 normal controls. The relationship between the percentage of CD203c-expressing basophils and clinical parameters was analyzed. The mean basophil CD203c expression was significantly higher in CU patients than in healthy controls (57.5% vs 11.6%, P < 0.001). The basophil CD203c expression in severe CU patients was significantly higher than in non-severe CU (66.5% ± 23.3% vs 54.0% ± 23.3%, P = 0.033). Multiple logistic regression analysis indicated that both ≥ 72% basophil CD203c expression and urticaria activity score (UAS)≥ 13 were significant predictors of severe CU (P = 0.005 and P = 0.032, respectively). These findings suggest that the quantification of basophil activation with CD203c at baseline may be used as a potential predictor of severe CU requiring another treatment option beyond antihistamines.


Subject(s)
Basophils/immunology , Phosphoric Diester Hydrolases/immunology , Pyrophosphatases/immunology , Urticaria/immunology , Adult , Autoantibodies/blood , Female , Flow Cytometry , Humans , Immunoglobulin E/blood , Immunoglobulin E/immunology , Male , Phosphoric Diester Hydrolases/biosynthesis , Pyrophosphatases/biosynthesis , Receptors, IgE/biosynthesis
15.
Clin Exp Allergy ; 44(2): 238-49, 2014 Feb.
Article in English | MEDLINE | ID: mdl-24118172

ABSTRACT

BACKGROUND: We recently reported that the interaction between Lyn and FcεRIß is indispensable for FcεRI-mediated human mast cell (MC) activation and that FcεRIß functions as an amplifier of FcεRI-mediated activation signal. Some of FcεRIß in cytoplasm appeared not to be co-localized with FcεRIα. The function of FcεRIß in the cytoplasm remains unknown. METHODS: The localization of FcεRIß and FcεRIα in giant papillae specimens from patients with allergic keratoconjunctivitis and of FcεRIß, FcεRIα, and Lyn in cultured human MCs was examined using confocal microscopy. FcεRIß was overexpressed using an adenovirus vector system. Mediators were measured by enzyme immunoassays or enzyme-linked immunosorbent assays. RESULTS: In the subepithelial region, FcεRIß was mainly localized in the cell membrane of MCs. In the perivascular region, FcεRIß expression was scattered throughout the cytoplasm and in the cell membrane of MCs. Overexpression of FcεRIß in MCs mainly increased its cytoplasmic expression and slightly up-regulated cell surface FcεRI expression. However, overexpression of FcεRIß in MCs resulted in down-regulation of the tyrosine phosphorylation levels of FcεRIß and Syk and down-regulation of the Ca(2+) influx soon after FcεRI aggregation and then resulted in down-regulation of degranulation, PGD2 synthesis, and production of a set of cytokines. This negative regulatory effect may be due to inhibition of the redistribution of Lyn to small patches within the plasma membrane. CONCLUSION: Cytoplasmic FcεRIß, which is not co-localized with FcεRIα, may function as a negative regulator, as it can capture important signalling molecules such as Lyn.


Subject(s)
Calcium Signaling , Down-Regulation , Hypersensitivity/metabolism , Keratoconjunctivitis/metabolism , Mast Cells/metabolism , Receptors, IgE/biosynthesis , Adult , Cell Line , Cytoplasm , Female , Humans , Hypersensitivity/immunology , Hypersensitivity/pathology , Intracellular Signaling Peptides and Proteins/immunology , Intracellular Signaling Peptides and Proteins/metabolism , Keratoconjunctivitis/immunology , Keratoconjunctivitis/pathology , Male , Mast Cells/immunology , Mast Cells/pathology , Protein-Tyrosine Kinases/immunology , Protein-Tyrosine Kinases/metabolism , Receptors, IgE/immunology , Syk Kinase , src-Family Kinases/immunology , src-Family Kinases/metabolism
16.
Mol Immunol ; 57(2): 129-37, 2014 Feb.
Article in English | MEDLINE | ID: mdl-24084099

ABSTRACT

Dysregulation of T-cell survival and apoptosis is the common cause of autoimmune diseases such as multiple sclerosis (MS). However, the factors inducing imbalance of T-cell survival and apoptosis in MS remains unclear. Here, we show that the resistance to apoptosis was associated with high levels of B-cell activating factor (BAFF). Blockade of BAFF with TACI (transmembrane activator and calcium modulator and cyclophilin ligand interactor)-IgG significantly reduced T-cell survival in myelin oligodendroglia glycoprotein (MOG)-induced chronic experimental allergic encephalitis (EAE). Furthermore, BAFF induced anti-apoptotic molecule Bcl2 expression in T cells by up-regulating osteopontin (OPN) secretion from B cells. BAFF mainly induced OPN expression in splenic CD21(-)CD23(+) B cells via a NF-kB dependent signaling pathway. In addition, we found that BAFF and OPN levels were increased in MS patients similar to the results obtained from our mice research. The study suggests that BAFF regulates T-cell survival by inducing OPN secretion in B cells in autoimmune diseases.


Subject(s)
B-Cell Activating Factor/metabolism , B-Lymphocytes/metabolism , Encephalomyelitis, Autoimmune, Experimental/immunology , Multiple Sclerosis/immunology , Osteopontin/metabolism , Animals , Apoptosis , B-Cell Activating Factor/biosynthesis , B-Lymphocytes/immunology , Cell Survival/immunology , Cells, Cultured , Humans , Immunoglobulin G , Mice , Mice, Inbred C57BL , Myelin-Oligodendrocyte Glycoprotein/immunology , NF-kappa B/metabolism , Osteopontin/biosynthesis , Peptide Fragments/immunology , Receptors, Complement 3d/biosynthesis , Receptors, IgE/biosynthesis , Signal Transduction/immunology , T-Lymphocytes/immunology , Transmembrane Activator and CAML Interactor Protein/immunology , Transmembrane Activator and CAML Interactor Protein/metabolism
17.
Article in English | WPRIM (Western Pacific) | ID: wpr-200228

ABSTRACT

Increased FcepsilonR1alpha expression with upregulated CD203c expression on peripheral basophils is seen in patients with chronic urticaria (CU). However, there has been no published report on the association between CD203c expression level and clinical disease activity in CU patients. To investigate whether the increase of basophil activation is associated with the disease activity of CU, we measured basophil CD203c expression using a tricolor flow cytometric method in 82 CU patients and 21 normal controls. The relationship between the percentage of CD203c-expressing basophils and clinical parameters was analyzed. The mean basophil CD203c expression was significantly higher in CU patients than in healthy controls (57.5% vs 11.6%, P or = 72% basophil CD203c expression and urticaria activity score (UAS)> or = 13 were significant predictors of severe CU (P = 0.005 and P = 0.032, respectively). These findings suggest that the quantification of basophil activation with CD203c at baseline may be used as a potential predictor of severe CU requiring another treatment option beyond antihistamines.


Subject(s)
Adult , Female , Humans , Male , Autoantibodies/blood , Basophils/immunology , Flow Cytometry , Immunoglobulin E/blood , Phosphoric Diester Hydrolases/biosynthesis , Pyrophosphatases/biosynthesis , Receptors, IgE/biosynthesis , Urticaria/immunology
18.
J Immunol ; 191(9): 4505-13, 2013 Nov 01.
Article in English | MEDLINE | ID: mdl-24068671

ABSTRACT

We previously demonstrated that TGF-ß1 suppresses IgE-mediated signaling in human and mouse mast cells in vitro, an effect that correlated with decreased expression of the high-affinity IgE receptor, FcεRI. The in vivo effects of TGF-ß1 and the means by which it suppresses mast cells have been less clear. This study shows that TGF-ß1 suppresses FcεRI and c-Kit expression in vivo. By examining changes in cytokine production concurrent with FcεRI expression, we found that TGF-ß1 suppresses TNF production independent of FcεRI levels. Rather, IgE-mediated signaling was altered. TGF-ß1 significantly reduced expression of Fyn and Stat5, proteins critical for cytokine induction. These changes may partly explain the effects of TGF-ß1, because Stat5B overexpression blocked TGF-mediated suppression of IgE-induced cytokine production. We also found that Stat5B is required for mast cell migration toward stem cell factor, and that TGF-ß1 reduced this migration. We found evidence that genetic background may alter TGF responses. TGF-ß1 greatly reduced mast cell numbers in Th1-prone C57BL/6, but not Th2-prone 129/Sv mice. Furthermore, TGF-ß1 did not suppress IgE-induced cytokine release and did increase c-Kit-mediated migration in 129/Sv mast cells. These data correlated with high basal Fyn and Stat5 expression in 129/Sv cells, which was not reduced by TGF-ß1 treatment. Finally, primary human mast cell populations also showed variable sensitivity to TGF-ß1-mediated changes in Stat5 and IgE-mediated IL-6 secretion. We propose that TGF-ß1 regulates mast cell homeostasis, and that this feedback suppression may be dependent on genetic context, predisposing some individuals to atopic disease.


Subject(s)
Immunoglobulin E/immunology , Mast Cells/metabolism , Receptors, IgE/immunology , STAT5 Transcription Factor/metabolism , Transforming Growth Factor beta1/metabolism , Animals , Cell Movement/immunology , Cells, Cultured , Cytokines/metabolism , Humans , Immunoglobulin E/metabolism , Mast Cells/immunology , Mice , Mice, Inbred C57BL , Proto-Oncogene Proteins c-fyn/metabolism , Proto-Oncogene Proteins c-kit/metabolism , RNA Interference , RNA, Small Interfering , Receptors, IgE/biosynthesis , Receptors, IgE/metabolism , STAT5 Transcription Factor/genetics , STAT5 Transcription Factor/immunology , Signal Transduction/immunology , Transforming Growth Factor beta1/immunology , Tumor Necrosis Factors/biosynthesis
19.
Cell Immunol ; 283(1-2): 38-44, 2013.
Article in English | MEDLINE | ID: mdl-23850674

ABSTRACT

There is increasing evidence that histamine regulates the immune system via histamine H4 receptors, therefore we sought to investigate the functions of the H4 receptor on mast cells. Mast cells were differentiated from murine bone marrow stem cells, and the expression of mast cell surface markers FcεRI and CD117 were measured using flow cytometry. Real-time qRT-PCR was used to determine the expression of mH4R; as a measure of antigen-dependent degranulation, ß-hexosaminidase release assay was carried out using IgE sensitized mast cells. We determined that the expression kinetics of FcεRI and mH4R can be described with a function that has one maximum value in the time range of the culture's differentiation. Antigen-dependent degranulation of murine bone marrow-derived mast cells could be inhibited by a selective H4 antagonist/inverse agonist only when it was present during the IgE sensitization phase of degranulation. In addition, flow cytometric analysis revealed that the H4 antagonist/inverse agonist also inhibited IgE induced FcεRI upregulation. The inhibition percentage of H4 antagonist on IgE induced FcεRI upregulation was determined to be dependent upon the maturity of the mast cell cultures, and this time-dependency was consistent with the expression kinetics of both mH4R and FcεRI. These results imply that H4R has regulatory roles in FcεRI expression and FcεRI mediated functions in mast cells. In conclusion the present study shows that H4 receptors potentially play a role in IgE induced FcεRI upregulation and in the sensitization phase but not the effector phase of mast cell degranulation.


Subject(s)
Cell Degranulation/immunology , Mast Cells/metabolism , Receptors, Histamine/metabolism , Receptors, IgE/biosynthesis , Animals , Bone Marrow Cells/immunology , Bone Marrow Cells/metabolism , Flow Cytometry , Immunoglobulin E/immunology , Male , Mast Cells/immunology , Mice , Mice, Inbred BALB C , Real-Time Polymerase Chain Reaction , Receptors, Histamine/immunology , Receptors, IgE/immunology , Up-Regulation
20.
J Mol Histol ; 44(3): 327-38, 2013 Jun.
Article in English | MEDLINE | ID: mdl-23377922

ABSTRACT

Although CD23-dependent transcytosis of IgE and IgE-derived immune complexes across respiratory epithelial cells is likely to play a pivotal role in the initiation and development of airway allergic inflammation, there is currently a lack of physiological support for this phenomena to suggest that the targeting of CD23 could be used as a means of therapeutic intervention. The present study was designed to detect the CD23 expression in the nasal mucosa of allergic rhinitis (AR) murine model by immunohistochemistry and western blotting, and to investigate whether intranasal anti-CD23 treatment could inhibit allergen-induced upper airway inflammation in the AR model. This is the first report to show that CD23 was constitutively expressed in murine nasal epithelial cells, and its expression was significantly up-regulated in the AR murine model. In vivo, the up-regulation of CD23 expression was correlated with increased serum IL-4 levels. Following intranasal anti-CD23 treatment, nasal symptoms were alleviated and histopathologic examination showed a significant decrease in eosinophilic infiltration. Meanwhile, ELISA analysis showed levels of serum leukotriene C4 (LTC4), eosinophil cation protein (ECP), ovalbumin (OVA)-specific IgE and IL-4 also significantly decreased, as were LTC4 and OVA-specific IgE in the nasal lavage fluid. Furthermore, Western blotting analysis showed that ECP expression in the nasal mucosa was down-regulated. Finally, flow cytometric analysis revealed anti-CD23 treatment inhibited Th2 cell responses. These results indicate that intranasal anti-CD23 treatment can reduce allergic responses in a murine model of allergic rhinitis.


Subject(s)
Hypersensitivity/drug therapy , Hypersensitivity/immunology , Receptors, IgE/antagonists & inhibitors , Receptors, IgE/immunology , Rhinitis, Allergic, Perennial/drug therapy , Rhinitis, Allergic, Perennial/immunology , Administration, Intranasal , Allergens/immunology , Animals , Budesonide/pharmacology , Disease Models, Animal , Down-Regulation/immunology , Eosinophil Cationic Protein/blood , Eosinophil Cationic Protein/immunology , Eosinophils/immunology , Eosinophils/pathology , Epithelial Cells/immunology , Epithelial Cells/pathology , Female , Hypersensitivity/blood , Hypersensitivity/pathology , Immunoglobulin E/blood , Immunoglobulin E/immunology , Inflammation/blood , Inflammation/immunology , Inflammation/pathology , Interleukin-4/blood , Interleukin-4/immunology , Leukotriene C4/blood , Leukotriene C4/immunology , Mice , Mice, Inbred BALB C , Nasal Mucosa/immunology , Nasal Mucosa/pathology , Ovalbumin/immunology , Ovalbumin/pharmacology , Random Allocation , Receptors, IgE/biosynthesis , Rhinitis, Allergic , Rhinitis, Allergic, Perennial/blood , Rhinitis, Allergic, Perennial/pathology , Th2 Cells/immunology , Th2 Cells/pathology , Up-Regulation/immunology
SELECTION OF CITATIONS
SEARCH DETAIL
...