Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 18 de 18
Filter
Add more filters










Publication year range
1.
Immunology ; 172(1): 91-108, 2024 May.
Article in English | MEDLINE | ID: mdl-38303079

ABSTRACT

Natural IgE cytotoxic peptides (nECPs), which are derived from the constant domain of the heavy chain of human IgE producing B cells via endoplasmic reticulum (ER) stress, are decorated onto MHC class 1a molecules (MHCIa) as unique biomarkers for CTL (cytotoxic T lymphocyte)-mediated immune surveillance. Human IgE exhibits only one isotype and lacks polymorphisms; IgE is pivotal in mediating diverse, allergen-specific allergies. Therefore, by disrupting self-IgE tolerance via costimulation, the CTLs induced by nECPs can serve as universal allergy vaccines (UAVs) in humans to dampen IgE production mediated by diverse allergen-specific IgE-secreting B cells and plasma cells expressing surface nECP-MHCIa as targets. The study herein has enabled the identification of nECPs, A32 and SP-1/SP-2 nonameric natural peptides produced through the correspondence principle. Vaccination using nECP induced nECP-specific CTL that profoundly suppressed human IgE production in vitro as well as chimeric human IgE production in human IgE/HLA-A2.01/HLA-B7.02 triple transgenic rodents. Furthermore, nECP-tetramer-specific CTLs were found to be converted into CD4 Tregs that restored IgE competence via the homeostatic principle, mediatepred by SREBP-1c suppressed DCs. Thus, nECPs showed causal efficacy and safety as UAVs for treating categorically type I hypersensitivity IgE-mediated allergies. The applied vaccination concept presented provides the foundation to unify, integrate through a singular class of tetramer-specific TCR clonotypes for regulaing human IgE production. The three signal theory pertains to mechanisms of three cells underlying central tolerance (S), breaking self tolerance (NS) and regaining peripheral tolerance (S) via homeostasis concerning nECP as an efficacious and safe UAV to treat type I IgE-mediated hypersensitivity. The three signal theory impirically extended, may be heuritic for immuno-regulation of adaptive immune repertoire in general.


Subject(s)
Hypersensitivity , Vaccines , Humans , T-Lymphocytes, Cytotoxic , Hypersensitivity/therapy , Immunoglobulin E , Allergens , Peptides , Homeostasis , Epitopes, T-Lymphocyte
2.
bioRxiv ; 2023 Oct 17.
Article in English | MEDLINE | ID: mdl-37904962

ABSTRACT

Natural IgE cytotoxic peptides (nECPs), which are derived from the constant domain of the heavy chain of human IgE producing B cells via endoplasmic reticulum (ER) stress, are decorated onto MHC class 1a molecules (MHCIa) as unique biomarkers for CTL (cytotoxic T lymphocyte)-mediated immune surveillance. Human IgE exhibits only one isotype and lacks polymorphisms; IgE is pivotal in mediating diverse, allergen-specific allergies. Therefore, by disrupting self-IgE tolerance via costimulation, the cytotoxic T lymphocytes (CTLs) induced by nECPs can serve as universal allergy vaccines (UAVs) in humans to dampen IgE production mediated by diverse allergen-specific IgE- secreting B cells and plasma cells expressing surface nECP-MHCIa as targets. The study herein has enabled the identification of nECPs produced through the correspondence principle 1, 2 . Furthermore, nECP-tetramer-specific CTLs were found to be converted into CD4 Tregs that restored IgE competence via the homeostatic principle, mediated by SREBP-1c suppressed DCs. Thus, nECPs showed causal efficacy and safety as UAVs for treating type I hypersensitivity IgE-mediated allergies. The applied vaccination concept presented provides the foundation to unify, integrate through a singular class of tetramer-specific TCR clonotypes. The three signal model is proposed on the mechanisms underlying central tolerance, breaking tolerance and regaining peripheral tolerance via homeostasis concerning nECP as an efficacious and safe UAV to treat type I IgE-mediated hypersensitivity. One Sentence Summary: Human IgE self-peptides are identified as universal allergy vaccines that inhibit IgE synthesis while allowing homeostatic IgE recovery.Graphic abstract textThree cell S/NS/S model of Universal Allergy Vaccines (UAV): Natural IgE peptides (nECPs) presented by enabler DCs break central IgE tolerance (Self), leading to CTLs that inhibit IgE production (Non-self). Generative DCs converted by the metabolic milieu transform the pre-existing nECP-specific CTLs into nECP-specific Tregs leading to homeostatic recovery of IgE competence (S).

3.
Cell Immunol ; 381: 104611, 2022 Sep 27.
Article in English | MEDLINE | ID: mdl-36194940

ABSTRACT

Herein, we show that profound afferent long-term peanut-allergen-specific IgE immunological tolerance for 3 to 9 months induced sustained unresponsiveness (SU) in naïve or peanut-sensitized rodents after peanut allergen immunization. Rodents were vaccinated sublingually with a peanut allergen extract or recombinant peanut allergen in chenodeoxycholate (CDCA), a fanesoid X receptor (FXR, NR1H4) agonist that downregulates SREBP-1c (sterol regulatory element binding protein-1c) and upregulates SHP in bone marrow-derived tolerogenic dendritic cells (DCs). Approximately 90 âˆ¼ 95 % of the total circulating PE-potentiated IgE and Ara h1, Ara h 2, and Ara h 6 peanut allergen-specific IgE responses were suppressed by recombinant peanut allergen-conjugated solid magnetic beads (sensitivity of 0.2 IU/ml). In contrast, peanut allergen-specific IgG production was not affected. Similarly, oleoylethanolamine (OEA), a peroxisome proliferator-activator receptor alpha (PPARα) agonist, and GW9662, a PPARγ antagonist, induced long-term peanut-specific IgE tolerance when administered via the sublingual, oral or i.p. route. Prophylactic Ara h2 DNA immunization with caNRF2 and IL-35 coexpression induced Ara h2 IgE tolerance. In summary, peanut allergen vaccination with select natural molecular ligands of nuclear receptors induced long-term peanut allergen-specific IgE tolerance via the afferent limb, which indicates that vaccination is an immune tolerance-promoting strategy that is effective at the DC level and that differs from Noon's daily desensitization program, which is effective at the mast cell level.

4.
J Leukoc Biol ; 97(6): 1071-80, 2015 Jun.
Article in English | MEDLINE | ID: mdl-25877925

ABSTRACT

IL-12 p40, a common subunit for both IL-12 p70 and IL-23, plays a critical role in the development of Th1 and Th17 cells and autoimmune diseases. Regulation of IL-12 p40 expression is thus considered to be a strategy for developing therapies for Th1- and Th17-mediated autoimmune diseases. The mTOR protein is a subunit mTORC1 and mTORC2. Although mTORC1 has been shown to mediate IL-12 p40 expression in DCs and relevant signaling, the role of mTORC2 in IL-12 p40 expression remains largely unclear. In the present study, we demonstrate that blocking mTORC2 activity using the phytochemical cytopiloyne can specifically inhibit LPS-induced expression of IL-12 p70, IL-23, and IL-12 p40 in human DCs. This regulation by mTORC2 involving Akt activation and the persistent phase of NF-κB activation is further confirmed by siRNA knockdown of Rictor and Sin1 gene expression and the use of alternative inhibition approaches. In terms of IL-12 p40 expression, our findings reveal a new role for the mTORC2 pathway that is antagonistic to that of mTORC1. Our study provides new insight into mTOR regulation of IL-12 p40-mediated Th1 (IFN-γ) and Th17 (IL-17) responses and suggests that the phytochemical cytopiloyne might have useful applications in therapies for Th1 and Th17 cell-mediated inflammatory diseases.


Subject(s)
Dendritic Cells/immunology , Interleukin-12 Subunit p40/immunology , Interleukin-12/immunology , Lipopolysaccharides/pharmacology , Multiprotein Complexes/immunology , Receptors, Interleukin/immunology , TOR Serine-Threonine Kinases/immunology , Adaptor Proteins, Signal Transducing/antagonists & inhibitors , Adaptor Proteins, Signal Transducing/genetics , Adaptor Proteins, Signal Transducing/immunology , Animals , Carrier Proteins/antagonists & inhibitors , Carrier Proteins/immunology , Cell Differentiation , Dendritic Cells/cytology , Dendritic Cells/drug effects , Gene Expression Regulation , Glucosides/pharmacology , Humans , Interleukin-12/genetics , Interleukin-12 Subunit p40/genetics , Mechanistic Target of Rapamycin Complex 2 , Monocytes/cytology , Monocytes/drug effects , Monocytes/immunology , Multiprotein Complexes/antagonists & inhibitors , Multiprotein Complexes/genetics , NF-kappa B/genetics , NF-kappa B/immunology , Polyynes/pharmacology , Primary Cell Culture , Proto-Oncogene Proteins c-akt/genetics , Proto-Oncogene Proteins c-akt/immunology , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism , Rapamycin-Insensitive Companion of mTOR Protein , Receptors, Interleukin/genetics , Signal Transduction , TOR Serine-Threonine Kinases/antagonists & inhibitors , TOR Serine-Threonine Kinases/genetics
5.
Cell Immunol ; 294(1): 44-53, 2015 Mar.
Article in English | MEDLINE | ID: mdl-25684095

ABSTRACT

Galectin-3 (Gal-3), a ß-galactoside-binding lectin, serves as a pattern-recognition receptor (PRR) of dendritic cells (DCs) in regulating proinflammatory cytokine production. Galectin-3 (Gal-3) siRNA downregulates expression of IL-6, IL-1ß and IL-23 p19, while upregulates IL-10 and IL-12 p35 in TLR/NLR stimulated human MoDCs. Furthermore, Gal-3 siRNA-treated MoDCs enhanced IFN-γ production in SEB-stimulated CD45RO CD4 T-cells, but attenuated IL-17A and IL-5 production by CD4 T-cells. Addition of neutralizing antibodies against Gal-3, or recombinant Gal-3 did not differentially modulate IL-23 p19 versus IL-12 p35. The data indicate that intracellular Gal-3 acts as cytokine hub of human DCs in responding to innate immunity signals. Gal-3 downregulation reprograms proinflammatory cytokine production by MoDCs that inhibit Th2/Th17 development.


Subject(s)
Cytokines/biosynthesis , Dendritic Cells/immunology , Galectin 3/biosynthesis , Inflammation/immunology , Receptors, Pattern Recognition/genetics , Cell Differentiation/immunology , Cell Line , Dendritic Cells/cytology , Down-Regulation , Galectin 3/genetics , HT29 Cells , Humans , Interleukin-10/biosynthesis , Interleukin-12 Subunit p35/biosynthesis , Interleukin-1beta/biosynthesis , Interleukin-23 Subunit p19/biosynthesis , Interleukin-6/biosynthesis , RNA Interference , RNA, Small Interfering , Receptors, Pattern Recognition/biosynthesis , Th17 Cells/cytology , Th17 Cells/immunology , Th2 Cells/cytology , Th2 Cells/immunology
6.
Biochem J ; 460(1): 69-78, 2014 May 15.
Article in English | MEDLINE | ID: mdl-24593306

ABSTRACT

Fucosylation regulates various pathological events in cells. We reported that different levels of CRT (calreticulin) affect the cell adhesion and metastasis of bladder cancer. However, the precise mechanism of tumour metastasis regulated by CRT remains unclear. Using a DNA array, we identified FUT1 (fucosyltransferase 1) as a gene regulated by CRT expression levels. CRT regulated cell adhesion through α1,2-linked fucosylation of ß1 integrin and this modification was catalysed by FUT1. To clarify the roles for FUT1 in bladder cancer, we transfected the human FUT1 gene into CRT-RNAi stable cell lines. FUT1 overexpression in CRT-RNAi cells resulted in increased levels of ß1 integrin fucosylation and rescued cell adhesion to type-I collagen. Treatment with UEA-1 (Ulex europaeus agglutinin-1), a lectin that recognizes FUT1-modified glycosylation structures, did not affect cell adhesion. In contrast, a FUT1-specific fucosidase diminished the activation of ß1 integrin. These results indicated that α1,2-fucosylation of ß1 integrin was not involved in integrin-collagen interaction, but promoted ß1 integrin activation. Moreover, we demonstrated that CRT regulated FUT1 mRNA degradation at the 3'-UTR. In conclusion, the results of the present study suggest that CRT stabilized FUT1 mRNA, thereby leading to an increase in fucosylation of ß1 integrin. Furthermore, increased fucosylation levels activate ß1 integrin, rather than directly modifying the integrin-binding sites.


Subject(s)
Calreticulin/biosynthesis , Fucosyltransferases/physiology , Integrin beta1/metabolism , Urinary Bladder Neoplasms/metabolism , Cell Adhesion/genetics , Cell Line, Tumor , Fucosyltransferases/genetics , Humans , Integrin beta1/genetics , Protein Stability , RNA Stability/genetics , Urinary Bladder Neoplasms/genetics , Urinary Bladder Neoplasms/pathology , Galactoside 2-alpha-L-fucosyltransferase
7.
Cancer Immunol Immunother ; 61(11): 1989-2002, 2012 Nov.
Article in English | MEDLINE | ID: mdl-22527248

ABSTRACT

Immunogenic cell death is characterized by damage-associated molecular patterns, which can enhance the maturation and antigen uptake of dendritic cells. Shikonin, an anti-inflammatory and antitumor phytochemical, was exploited here as an adjuvant for dendritic cell-based cancer vaccines via induction of immunogenic cell death. Shikonin can effectively activate both receptor- and mitochondria-mediated apoptosis and increase the expression of all five tested damage-associated molecular patterns in the resultant tumor cell lysates. The combination treatment with damage-associated molecular patterns and LPS activates dendritic cells to a high maturation status and enhances the priming of Th1/Th17 effector cells. Shikonin-tumor cell lysate-loaded mature dendritic cells exhibit a high level of CD86 and MHC class II and activate Th1 cells. The shikonin-tumor cell lysate-loaded dendritic cell vaccines result in a strong induction of cytotoxic activity of splenocytes against target tumor cells, a retardation in tumor growth, and an increase in the survival of test mice. The much enhanced immunogenicity and efficacy of the current cancer vaccine formulation, that is, the use of shikonin-treated tumor cells as cell lysates for the pulse of dendritic cells in culture, may suggest a new ex vivo approach for developing individualized, dendritic cells-based anticancer vaccines.


Subject(s)
Apoptosis/drug effects , Cancer Vaccines/therapeutic use , Dendritic Cells/transplantation , Melanoma, Experimental/therapy , Naphthoquinones/administration & dosage , Skin Neoplasms/therapy , Animals , Apoptosis/immunology , B7-2 Antigen/immunology , Cells, Cultured , Dendritic Cells/immunology , Female , Genes, MHC Class II/immunology , Mice , Mice, Inbred C57BL , Spleen/immunology , Th1 Cells/drug effects , Th1 Cells/immunology , Th17 Cells/drug effects , Th17 Cells/immunology
8.
Stem Cells ; 29(11): 1763-73, 2011 Nov.
Article in English | MEDLINE | ID: mdl-21915944

ABSTRACT

Lysophosphatidic acid (LPA), an extracellular lipid mediator, exerts multiple bioactivities through activating G protein-coupled receptors. LPA receptor 3 (LPA(3)) is a member of the endothelial differentiation gene family, which regulates differentiation and development of the circulation system. However, the relationship among the LPA receptors (LPARs) and erythropoiesis is still not clear. In this study, we found that erythroblasts expressed both LPA(1) and LPA(3), and erythropoietic defects were observed in zLPA(3) antisense morpholino oligonucleotide-injected zebrafish embryos. In human model, our results showed that LPA enhanced the erythropoiesis in the cord blood-derived human hematopoietic stem cells (hHSCs) with erythropoietin (EPO) addition in the plasma-free culture. When hHSCs were treated with Ki16425, an antagonist of LPA(1) and LPA(3), erythropoietic process of hHSCs was also blocked, as detected by mRNA and protein expressions of CD71 and GlyA. In the knockdown study, we further demonstrated that specific knockdown of LPA(3), not LPA(1), blocked the erythropoiesis. The translocation of ß-catenin into the nucleus, a downstream response of LPAR activation, was blocked by Ki16425 treatment. In addition, upregulation of erythropoiesis by LPA was also blocked by quercetin, an inhibitor of the ß-catenin/T-cell factor pathway. Furthermore, the enhancement of LPA on erythropoiesis was diminished by blocking c-Jun-activated kinase/signal transducer and activator of transcription and phosphatidylinositol 3-kinase/AKT activation, the downstream signaling pathways of EPO receptor, suggested that LPA might play a synergistic role with EPO to regulate erythropoietic process. In conclusion, we first reported that LPA participates in EPO-dependent erythropoiesis through activating LPA(3).


Subject(s)
Erythropoiesis/drug effects , Lysophospholipids/pharmacology , Receptors, Lysophosphatidic Acid/agonists , Receptors, Lysophosphatidic Acid/metabolism , AC133 Antigen , Animals , Antigens, CD/metabolism , Cells, Cultured , Embryo, Nonmammalian , Flow Cytometry , Glycoproteins/metabolism , Hematopoietic Stem Cells/cytology , Hematopoietic Stem Cells/drug effects , Hematopoietic Stem Cells/metabolism , Humans , Isoxazoles/pharmacology , Peptides/metabolism , Propionates/pharmacology , Receptors, Lysophosphatidic Acid/antagonists & inhibitors , Zebrafish
9.
J Biomed Sci ; 18: 44, 2011 Jun 20.
Article in English | MEDLINE | ID: mdl-21689407

ABSTRACT

BACKGROUND: Damage-associated molecular patterns (DAMPs) are associated with immunogenic cell death and have the ability to enhance maturation and antigen presentation of dendritic cells (DCs). Specific microtubule-depolymerizing agents (MDAs) such as colchicine have been shown to confer anti-cancer activity and also trigger activation of DCs. METHODS: In this study, we evaluated the ability of three MDAs (colchicine and two 2-phenyl-4-quinolone analogues) to induce immunogenic cell death in test tumor cells, activate DCs, and augment T-cell proliferation activity. These MDAs were further evaluated for use as an adjuvant in a tumor cell lysate-pulsed DC vaccine. RESULTS: The three test phytochemicals considerably increased the expression of DAMPs including HSP70, HSP90 and HMGB1, but had no effect on expression of calreticulin (CRT). DC vaccines pulsed with MDA-treated tumor cell lysates had a significant effect on tumor growth, showed cytotoxic T-lymphocyte activity against tumors, and increased the survival rate of test mice. In vivo antibody depletion experiments suggested that CD8+ and NK cells, but not CD4+ cells, were the main effector cells responsible for the observed anti-tumor activity. In addition, culture of DCs with GM-CSF and IL-4 during the pulsing and stimulation period significantly increased the production of IL-12 and decreased production of IL-10. MDAs also induced phenotypic maturation of DCs and augmented CD4+ and CD8+ T-cell proliferation when co-cultured with DCs. CONCLUSIONS: Specific MDAs including the clinical drug, colchicine, can induce immunogenic cell death in tumor cells, and DCs pulsed with MDA-treated tumor cell lysates (TCLs) can generate potent anti-tumor immunity in mice. This approach may warrant future clinical evaluation as a cancer vaccine.


Subject(s)
Cancer Vaccines/immunology , Colchicine/therapeutic use , Dendritic Cells/immunology , Melanoma, Experimental/immunology , Melanoma, Experimental/therapy , Quinolones/therapeutic use , Adjuvants, Immunologic , Animals , Cancer Vaccines/therapeutic use , Cell Death , Cell Line, Tumor , Colchicine/pharmacology , Dendritic Cells/cytology , Lymphocyte Activation , Male , Mice , Mice, Inbred C57BL , Quinolones/pharmacology
10.
Cytokine ; 55(1): 79-89, 2011 Jul.
Article in English | MEDLINE | ID: mdl-21486701

ABSTRACT

In this study, we compared the effects of zymosan and LPS on human monocyte-derived dendritic cells. The specific effects of zymosan on the expression of several key cytokines, including granulocyte-macrophage colony-stimulating factor (GM-CSF) and interleukins (IL-1α, IL-1ß and IL-12 p70) were quite distinct from the effects of LPS. Unlike activation with LPS, DCs activated by zymosan expressed little or no IL-12 p70 due to lack of expression of the p35 subunit. However, treatment with zymosan resulted in a substantial increase in Th1 and Th17 cell-polarizing capacity of DCs. Furthermore, the GM-CSF secreted by zymosan-activated DCs enhanced IL-23 production, resulting in activation of a Th17 response. GM-CSF and IL-27, rather than IL-12 p70, were both major direct contributors to the activation of a Th1 response. This signaling mechanism is distinct and yet complementary to LPS-mediated T-cell activation. We suggest that this novel zymosan-induced GM-CSF-mediated signaling network may play a key role in regulating specific immune cell type activities.


Subject(s)
Dendritic Cells/drug effects , Dendritic Cells/immunology , Granulocyte-Macrophage Colony-Stimulating Factor/metabolism , Lymphocyte Activation/drug effects , Th1 Cells/immunology , Th17 Cells/immunology , Zymosan/pharmacology , Biomarkers/metabolism , Cell Differentiation/drug effects , Cell Shape/drug effects , Cytokines/genetics , Cytokines/metabolism , Dendritic Cells/cytology , Dendritic Cells/metabolism , Gene Expression Regulation/drug effects , Humans , Phenotype , RNA, Messenger/genetics , RNA, Messenger/metabolism , Reproducibility of Results , Th1 Cells/cytology , Th1 Cells/drug effects , Th17 Cells/cytology , Th17 Cells/drug effects
11.
BMC Immunol ; 11: 34, 2010 Jun 30.
Article in English | MEDLINE | ID: mdl-20591187

ABSTRACT

BACKGROUND: At present, it is highly controversial whether pure mast cells can serve as antigen presenting cells, and it is not known whether the capacity of antigen presenting function is temporally restricted to a particular subset of differentiated mast cells. Evidence is presented for a novel surface FcepsilonRIhi , MHC II +, and c-kit + pure mast cell subset, temporally restricted as antigen-presenting cells in the immune axis of T-cell activation. RESULTS: Bone marrow-derived mast cells (BMMC) cultured in the presence of IL-3 for three weeks are pure mast cells based on surface expression of lineage-specific marker, c-kit and FcepsilonRI. Herein we present the first demonstration that approximately 98.7% c-kit + and FcepsilonRI expressing BMMC, further depleted of any contaminated professional antigen-presenting cells, are still fully capable of presenting antigens, i.e., OVA protein, OVA peptide, and IgE-TNP-OVA, to OVA peptide-specific T-cell hybridomas. Notably, IgE-dependent antigen presentation is more efficient compared to that resulting from direct antigen uptake. Importantly, we present the novel finding that only surface FcepsilonRIhi mast cells, also expressing surface MHC II exhibited antigen-presenting function. In contrast, surface FcepsilonRIlo mast cells without expressing surface MHC II were not capable of antigen presentation. Interestingly, the antigen-presenting function of BMMC was irrevocably lost during the third and fourth week in IL-3 or SCF containing cultures. CONCLUSIONS: This is the first observation to attribute a spatiotemporally restricted antigen-presenting function to a subset of three-week old pure BMMC expressing both high levels of surface FcepsilonRI and surface MHC II. We propose that mast cells play an important role in immune deviating and/or sustaining the activation of infiltrating CD4 T-cells, and modulating T-cell mediated allergic inflammation via its flexibility to present antigens and antigen-IgE complexes.


Subject(s)
Antigen Presentation/immunology , Bone Marrow Cells/cytology , Bone Marrow Cells/immunology , Histocompatibility Antigens Class II/immunology , Mast Cells/immunology , Receptors, IgE/immunology , Animals , Antigen-Presenting Cells/immunology , Antigens/immunology , Cells, Cultured , Female , Intracellular Space/metabolism , Mast Cells/cytology , Mice , Time Factors
12.
Cell Immunol ; 254(1): 28-38, 2008.
Article in English | MEDLINE | ID: mdl-18674755

ABSTRACT

Effect of IgE peptide-specific CTL on IgE antibody production was studied in mouse models. CTL elicited in B6.A2Kb tg mice against a human IgE peptide nonamer, pWV, lysed human IgE-secreting U266 myeloma cells and inhibit IgE production by these cells. U266 transfected with mouse A2Kb transgene (U266-A2Kb) were optimally lysed by these CTL, because the alpha3 domain of A2Kb interacts well with the CD8 co-receptors. The CTL generated were more effective in inhibiting IgE production by U266-A2Kb cells than lysing these cells. IgE production by and progression of U266 myeloma were suppressed in B6.A2Kb tg mice rendered tolerant to these cells and vaccinated with pWV along with CpG. We also studied the CTL response elicited in wild-type mice by a mouse nonameric IgE peptide, PI-1, along with CpG. This treatment caused a transient suppression of the IgE response in mice previously sensitized to an antigen. In mice treated with this regimen repeatedly, the IgE response was fully recovered 20 days after each treatment. Notably, while IgE peptide/CpG-treated mice remained unresponsive to antigen challenge in vivo, antigen-specific IgE production can be elicited by antigen in cultured splenocytes from these mice. Moreover, IgE peptide/CpG also inhibited an on-going IgE response, including IgE production by bone marrow cells. Taken together, these observations indicate that a CTL-based IgE peptide vaccine targeting IgE-secreting B/plasma cells may be safely employed as a therapeutic approach for suppressing IgE production.


Subject(s)
HLA-A2 Antigen/immunology , Immunoglobulin E/immunology , Immunoglobulin Fragments/immunology , Peptide Fragments/immunology , T-Lymphocytes, Cytotoxic/immunology , Vaccines, Subunit/immunology , Animals , Antibody Formation , Enzyme-Linked Immunosorbent Assay , Flow Cytometry , Humans , Mice , Mice, Transgenic , Transfection
13.
Biochem Biophys Res Commun ; 374(3): 409-14, 2008 Sep 26.
Article in English | MEDLINE | ID: mdl-18619414

ABSTRACT

GFP-Ckappa fusion protein was previously shown selectable on ribosome display platform with solid phase antibodies against GFP determinant [Y.-M. Yang, T.J. Barankiewicz, M. He, M. Taussig, S.-S. Chen, Selection of antigenic markers on a GFP-Ckappa fusion scaffold with high sensitivity by eukaryotic ribosome display, Biochem. Biophys. Res. Commun. 359 (2007) 251-257]. Herein, we show that members of aptameric peptide library constructed within the site 6 and site 8/9 loops of GFP of the ribosome display construct are selectable upon binding to the solid phase IgE antigen. An input of 1.0 microg of the dual site aptameric GFP library exhibiting a diversity of 7.5x10(11) was transcribed, translated and incubated with solid phase IgE. RT-PCR products were amplified from mRNA of the aptamer-ribosome-mRNA (ARM) complex captured on the solid phase IgE. Clones of aptameric GFP were prepared from RT-PCR product of ARM complex following repetitive selection. Recombinant aptameric GFP proteins from the selected clones bind IgE coated on the 96-well plate, and the binding was abrogated by incubation with soluble human IgE but not human IgG. Selected aptameric GFP proteins also exhibit binding to three different sources of human IgE (IgE PS, BED, and JW8) but not irrelevant proteins. These observations indicate that appropriately selected aptameric GFP on a solid phase ligand by ribosome display may serve as an affinity reagent for blocking reactivity of a biological ligand.


Subject(s)
Aptamers, Peptide/isolation & purification , Biosensing Techniques , Green Fluorescent Proteins/isolation & purification , Immunoglobulin E/metabolism , Recombinant Fusion Proteins/isolation & purification , Animals , Aptamers, Peptide/metabolism , Green Fluorescent Proteins/metabolism , Humans , Mice , Multiple Myeloma/immunology , Peptide Library , Recombinant Fusion Proteins/metabolism , Ribosomes/metabolism
14.
J Immunol Methods ; 333(1-2): 10-23, 2008 Apr 20.
Article in English | MEDLINE | ID: mdl-18281056

ABSTRACT

Green fluorescent protein (GFP) exhibits a rigid central beta-barrel, formed by eleven beta-strands with floppy loops spanning between the stands. Herein, we evaluate whether the rigid beta-barrel may serve as a scaffold that can constrain the loops of a foreign protein, and thus its antigenicity. The spanning loops, site 6 of GFP, were engineered with RE cloning sites for inserting oligonucleotides corresponding to FcepsilonRI-binding sequence of human IgE. In a high-throughput format, shortened oligonucleotides encoding eight amino acid residues of the receptor-binding regions were inserted into site 6 of GFP by PCR, followed by enabling sequences for in vitro transcription and translation at the 5' end. Antigenized C2-3 linker (C2-3L) was shown by immuno-blots with polyclonal anti-IgE under native gel electrophoresis and transfer. Recombinant antigenized GFP was expressed and purified to homogeneity by metal affinity column, followed by Sephacryl S-200 high resolution gel filtration. Hyperimmune sera from mice immunized with C2-3L antigenized GFP contain anti-IgE reactive with JW8 murine/human chimeric IgE. Further, elevated serum anti-C2-3L and affinity pure antibodies effectively inhibits binding of JW8 IgE to recombinant FcepsilonRIalpha, and desensitizes JW8 to rat RBL-2H3 transfected with human FcepsilonRIalpha. This observation raised the possibility that active IgE vaccine may be employed in raising active protective anti-IgE in allergic patients as an alternative to passive immunization with MAb-E25 anti-IgE. Taken together, GFP appears suitable protein scaffold for spanning/constraining the C2-3L of human IgE as active vaccine; and this technique may be generally employed for eliciting antibodies to specific B-cell epitopes of other proteins.


Subject(s)
Green Fluorescent Proteins/immunology , Hypersensitivity/immunology , Immunization/methods , Immunoglobulin E/immunology , Animals , Base Sequence , Enzyme-Linked Immunosorbent Assay , Epitopes, B-Lymphocyte/immunology , Green Fluorescent Proteins/chemistry , Green Fluorescent Proteins/genetics , Hypersensitivity/prevention & control , Immunoglobulin E/chemistry , Immunoglobulin E/genetics , Immunoglobulin epsilon-Chains/immunology , Mast Cells , Mice , Mice, Inbred BALB C , Molecular Conformation , Mutagenesis, Insertional , Protein Conformation , Recombinant Proteins/chemistry , Recombinant Proteins/immunology , Transfection
15.
Biochem Biophys Res Commun ; 359(2): 251-7, 2007 Jul 27.
Article in English | MEDLINE | ID: mdl-17537405

ABSTRACT

Ribosome display is a cell-free system permitting gene selection through the physical association of genetic material (mRNA) and its phenotypic (protein) product. While often used to select single-chain antibodies from large libraries by panning against immobilized antigens, we have adapted ribosome display for use in the 'reverse' format in order to select high affinity antigenic determinants against solid-phase antibody. To create an antigenic scaffold, DNA encoding green fluorescent protein (GFP) was fused to a light chain constant domain (Ckappa) with stop codon deleted, and with 5' signals (T7 promoter, Kozak) enabling coupled transcription/translation in a eukaryotic cell-free system. Epitopes on either GFP (5') or Ckappa (3') were selected by anti-GFP or anti-Ckappa antibodies, respectively, coupled to magnetic beads. After selection, mRNA was amplified directly from protein-ribosome-mRNA (PRM) complexes by in situ PCR followed by internal amplification and reassembly PCR. As little as 10fg of the 1kb DNA construct, i.e. approximately 7500 molecules, could be recovered following a single round of interaction with solid-phase anti-GFP antibody. This platform is highly specific and sensitive for the antigen-antibody interaction and may permit selection and reshaping of high affinity antigenic variants of scaffold proteins.


Subject(s)
Antigens/chemistry , Genetic Markers/genetics , Green Fluorescent Proteins/chemistry , Ribosomes/metabolism , Animals , Biochemistry/methods , Cell-Free System , Green Fluorescent Proteins/metabolism , Models, Genetic , Peptide Library , Protein Biosynthesis , Protein Interaction Mapping , RNA, Messenger/metabolism , Rabbits , Recombinant Fusion Proteins/chemistry , Ribosomal Proteins/chemistry , Ribosomes/chemistry
16.
Cell Immunol ; 233(1): 11-22, 2005 Jan.
Article in English | MEDLINE | ID: mdl-15913585

ABSTRACT

Immunoglobulin E (IgE) plays a central role in IgE-mediated immediate type hypersensitivity. Since production of IgE depends on Th2, efforts to block IgE production and control allergic reactions include tolerization of Th2 or deviating development of Th2. We hypothesized that cytotoxic T lymphocytes targeting natural IgE peptides/MHC I complexes can eliminate IgE-producing cells and inhibit centrally IgE production. CTL to self-IgE peptides were elicited in mice immunized with nonameric p109-117, p113-121, and p103-141 (CHepsilon2 domain), which encompass both peptides with an OVA helper peptide (OVAp restricted for H-2d/b) in liposomes and presented by dendritic cells (DC). CTL from BALB/c lysed IgE peptide-pulsed P815 target as well as IgE-producing 26.82 hybridomas (H-2d). Natural tolerance to self-IgE peptides was tested in IgE sufficient (IgE +/+) as well as IgE-deficient (IgE -/-) 129/SvEv mice (H-2b). Comparable magnitude of CTL responses was observed in both strains immunized with p109-117 or p103-141 concomitantly with CD4 T-cell costimulation. CTL from 129/SvEv lysed not only IgE peptide-pulsed EL-4 but also IgE-producing B4 hybridomas (H-2b). This observation strongly suggests a correspondence of epitope of immunogenic peptide to that of physiologically processed IgE peptides presented on IgE-producing cells. Moreover, CTL were generated in 129/SvEv, immunized with the recombinant antigenized antibody in liposomes encompassing p107-123, p109-117, and p113-121 expressed in CDR3 of VH62/human gamma1. Polyclonal IgE production was inhibited by coincubation with MHC I-restricted CTL in vitro. Furthermore, antigen-specific IgE responses were inhibited in mice, immunized with p109-117 and p103-141 while IgG responses were not suppressed. Since IgE peptide sequences of CHepsilon2 are ubiquitous to all murine IgE heavy chain, peptides made as such can serve as a universal IgE vaccine to prevent allergy for a myriad of allergens in rodents. This observation suggests that similar human IgE peptides should be identified and employed to downregulate human IgE production.


Subject(s)
Down-Regulation/immunology , Immunoglobulin E/immunology , Peptide Fragments/immunology , T-Lymphocytes, Cytotoxic/immunology , Animals , Antibodies/genetics , Antibodies/immunology , B-Lymphocytes/immunology , B-Lymphocytes/metabolism , CD40 Ligand/immunology , CD8-Positive T-Lymphocytes/immunology , Cell Line, Tumor , Cell Transplantation , Complementarity Determining Regions/genetics , Cytotoxicity Tests, Immunologic , Dendritic Cells/immunology , Dendritic Cells/transplantation , Female , H-2 Antigens/immunology , H-2 Antigens/metabolism , Hemocyanins/immunology , Histocompatibility Antigens Class I/immunology , Hybridomas/immunology , Hypersensitivity/immunology , Hypersensitivity/therapy , Immunization , Immunoglobulin E/genetics , Immunotherapy, Active/methods , Mice , Mice, Inbred BALB C , Mice, Knockout , Ovalbumin/immunology , Peptide Fragments/genetics , Peptide Fragments/metabolism , Protein Binding/immunology , Recombinant Proteins/immunology , Self Tolerance/immunology
17.
Immunol Invest ; 33(3): 295-307, 2004 Aug.
Article in English | MEDLINE | ID: mdl-15495789

ABSTRACT

Epidemiological data showed that total IgE and IL-4 levels in cigarette smokers were elevated, comparable to those in the asthmatics. The etiological agent(s) elevating IgE production are not clear. We evaluate whether tobacco polyphenols potentiate IgE production in a rodent model. Mice were fed with rutin or CGA in drinking water during antigen sensitization, followed by antigenic challenge i.p. in alum. CGA and rutin were also delivered in a bolus intraperitoneally or intranasally along with antigens during immunization. Antigen-specific IgE and IgG responses were measured. Enhancement of total IgE responses via i.p. and drinking routes can be achieved at concentrations as low as 0.1% CGA. Furthermore, IgG1 responses but not IgG2a and IgG2b were augmented, indicating a Th2 type of response by CGA. Moreover, both antigen-specific and serum IgE production can be achieved when CGA and antigenic challenges were delivered intranasally in the absence of alum. In contrast, nicotine does not enhance antigen-specific IgE production, and only marginally affects serum IgE levels. The more polarized Th2 development in CGA-treated mice may account for enhancement of both antigen-specific and total IgE responses. High levels of IL-4 but not IFN-gamma or IL-12, were observed in antigen-challenged mesenteric lymph nodes (MLN) cultures from CGA-treated mice. In contrast, significant levels of IL-4, IL-12, and IFN-gamma were observed in antigen-challenged cultures from nicotine-treated mice. This study shows that tobacco polyphenols, CGA and rutin potentiated IgE production in vivo. Polyphenolic antioxidants enhance Th2 development. We propose that IgE production and T cell dichotomy may be critically influenced by the redox microenvironment. Enhanced Th2 development and IgE production henceforth may counteract more severe Th1-mediated tissue damage triggered by environmental oxidative stress.


Subject(s)
Antioxidants/pharmacology , Flavonoids/immunology , Flavonoids/pharmacology , Immunoglobulin E/blood , Immunoglobulin E/drug effects , Phenols/immunology , Phenols/pharmacology , Animals , Antioxidants/administration & dosage , Chlorogenic Acid/administration & dosage , Chlorogenic Acid/immunology , Chlorogenic Acid/pharmacology , Female , Flavonoids/administration & dosage , Mice , Mice, Inbred BALB C , Phenols/administration & dosage , Polyphenols , Rutin/administration & dosage , Rutin/immunology , Rutin/pharmacology , Th2 Cells/immunology , Tobacco, Smokeless/chemistry
18.
Int Immunopharmacol ; 3(13-14): 1841-52, 2003 Dec.
Article in English | MEDLINE | ID: mdl-14636833

ABSTRACT

Antigen-presenting cells (APC) provide two essential signals, e.g., antigenic peptides as well as costimulatory molecules for T-cell activation. Small molecules of smoke tobacco extracts (SM-STE) inhibited antigen presentation of A20 to OVAp-specific T-cell hybridomas. Pretreatment of A20 but not T hybridomas abrogates the APC function. Viability of APC and levels of MHCII, CD40 and B7 of APC were not affected by this treatment. The active principle, inhibiting APC was reproduced with pure tobacco polyphenols, quercetin and its glycoside, rutin. Antioxidant activity of rutin is relevant since rutin downregulated levels of reactive oxygen species (ROS) in phorbol ester-stimulated A20; moreover, another antioxidant, N-acetyl cysteine (NAC) also inhibited antigen presentation, albeit at a higher concentration. Other types of APC, such as bone marrow-derived mast cells (BMMC), MHCII-transfected fibroblast, and splenocytes are affected by tobacco polyphenols. We propose that polyphenols may affect redox-sensitive signal transduction pathway since APC function of PD 98059, MEK inhibitor-pretreated A20 were similarly abrogated. Taken together, we propose that maintaining appropriate intracellular redox of APC is crucial for its antigen-presenting function.


Subject(s)
Antigen Presentation/drug effects , Antigen-Presenting Cells/drug effects , Antioxidants/pharmacology , Flavonoids/pharmacology , Phenols/pharmacology , Acetylcysteine/pharmacology , Animals , Antigen Presentation/immunology , Antigen-Presenting Cells/immunology , Antigen-Presenting Cells/metabolism , Antigens, CD/drug effects , Antigens, CD/immunology , B-Lymphocytes/drug effects , B-Lymphocytes/immunology , B-Lymphocytes/metabolism , B7-2 Antigen , Bone Marrow Cells/drug effects , CD40 Antigens/drug effects , CD40 Antigens/immunology , Chlorogenic Acid/chemistry , Chlorogenic Acid/isolation & purification , Chlorogenic Acid/pharmacology , Cysteine Endopeptidases , Down-Regulation , Fibroblasts/cytology , Histocompatibility Antigens Class II/immunology , Hybridomas , Interleukin-2/antagonists & inhibitors , Interleukin-2/biosynthesis , Intracellular Signaling Peptides and Proteins , Lymphocyte Activation/immunology , Mast Cells/drug effects , Mast Cells/metabolism , Membrane Glycoproteins/drug effects , Membrane Glycoproteins/immunology , Mice , Mice, Inbred BALB C , Nuclear Proteins , Plant Extracts/chemistry , Plant Leaves/chemistry , Polyphenols , Protein Biosynthesis , Proteins/antagonists & inhibitors , Quercetin/chemistry , Quercetin/isolation & purification , Quercetin/pharmacology , Reactive Oxygen Species/antagonists & inhibitors , Rutin/chemistry , Rutin/isolation & purification , Rutin/pharmacology , Signal Transduction , Spleen/cytology , Spleen/drug effects , T-Lymphocytes/cytology , T-Lymphocytes/immunology
SELECTION OF CITATIONS
SEARCH DETAIL
...