Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 5 de 5
Filter
Add more filters










Database
Language
Publication year range
1.
Mol Immunol ; 52(3-4): 190-9, 2012 Oct.
Article in English | MEDLINE | ID: mdl-22706073

ABSTRACT

Type-I hypersensitivity reactions play a critical role in the pathogenesis of various allergic diseases. The successful development of the anti-IgE antibody, omalizumab, has validated IgE as an effective therapeutic target for the treatment of various IgE-mediated allergic diseases. Two research groups have reported that mAbs specific for certain parts of CɛmX, a domain of 52 aa residues in human membrane-bound IgE (mIgE), can cause the lysis of mIgE-B cells by apoptosis and antibody-dependent cellular cytotoxicity (ADCC). Herein, we explore virus-like particles formed by hepatitis B virus core antigen (HBcAg) that harbors the entire CɛmX peptide or its fragments as immunogens for inducing anti-CɛmX antibodies. The results showed that mice immunized subcutaneously with these immunogens produced antibodies that bind to recombinant CɛmX-containing human IgE.Fc in ELISA and Western blot analyses, and to genetically engineered human mIgE-expressing Ramos B cell line in fluorescence flow cytometric assays. The IgG antibodies purified from the sera of immunized mice were able to cause the apoptosis of mIgE-expressing Ramos cells through a BCR-dependent caspase pathway. Furthermore, the IgG could mediate ADCC in human mIgE-expressing A20 murine B-cell lymphoma. These studies suggest that HBcAg-CɛmX peptide immunogens warrant further investigation as a therapeutic modality for modulating IgE in patients with IgE-mediated allergic diseases.


Subject(s)
Antibodies, Anti-Idiotypic/immunology , Apoptosis , B-Lymphocytes/immunology , Hepatitis B Core Antigens/immunology , Immunoglobulin E/immunology , Vaccines, Virus-Like Particle/immunology , Animals , Antibodies, Monoclonal/immunology , Antibody-Dependent Cell Cytotoxicity , B-Lymphocytes/metabolism , Cell Line, Tumor , Down-Regulation , Female , Hepatitis B virus/immunology , Humans , Immunoglobulin E/chemistry , Lymphocyte Activation , Mice , Mice, Inbred BALB C , Peptides/chemistry , Peptides/immunology , Receptors, Antigen, B-Cell/immunology
2.
Immunobiology ; 217(7): 676-83, 2012 Jul.
Article in English | MEDLINE | ID: mdl-22226669

ABSTRACT

A new monoclonal antibody (mAb), specific for human IgE, the central mediator of immediate-type hypersensitivity reactions, has been shown to possess a unique set of binding specificities. The mAb, 8D6, binds to a conformational epitope on the CH3 domain of human e immunoglobulin and can compete with omalizumab for binding to IgE. Like omalizumab, it does not bind to IgE bound by the high-affinity IgE.Fc receptor (FcɛRI) on basophils and mast cells. It also does not cause activation and degranulation of IgE-pulsed, human FcɛRI-expressing rat basophilic leukemic cells (RBL SX-38). The mAb can inhibit IgE binding to recombinant α chain of human FcɛRI in ELISA and to human FcɛRI-expressing RBL SX38 cells in fluorescence flow cytometric analysis. However, unlike omalizumab, 8D6 can bind to IgE already bound by the low-affinity IgE.Fc receptors (FcɛRII, or CD23), as revealed in ELISA with recombinant CD23 and in flow cytometric analysis with human B cells. Since earlier investigators have shown that anti-CD23 mAbs can inhibit the synthesis of IgE in lymphocyte culture in vitro and can down-regulate IgE production in treated patients, 8D6 may offer pharmacological mechanisms in addition to those mediated by omalizumab, for controlling IgE in patients with allergic diseases.


Subject(s)
Antibodies, Anti-Idiotypic/immunology , Antibodies, Monoclonal/immunology , Hypersensitivity, Immediate/immunology , Immunoglobulin E/immunology , Receptors, IgE/immunology , Animals , Antibodies, Anti-Idiotypic/metabolism , Antibodies, Anti-Idiotypic/pharmacology , Antibodies, Monoclonal/metabolism , Antibodies, Monoclonal/pharmacology , Antibodies, Monoclonal, Humanized/immunology , Antibodies, Monoclonal, Humanized/metabolism , Antibodies, Monoclonal, Humanized/pharmacology , Antibody Specificity , B-Lymphocytes/drug effects , B-Lymphocytes/immunology , B-Lymphocytes/metabolism , Basophils/drug effects , Basophils/immunology , Basophils/metabolism , Binding, Competitive , Enzyme-Linked Immunosorbent Assay , Epitopes , Humans , Hypersensitivity, Immediate/drug therapy , Hypersensitivity, Immediate/metabolism , Immunoglobulin E/metabolism , Mast Cells/drug effects , Mast Cells/immunology , Mast Cells/metabolism , Mice , Mice, Inbred BALB C , Omalizumab , Protein Binding , Protein Structure, Tertiary , Rats , Receptors, IgE/metabolism
3.
Immunogenetics ; 64(4): 279-87, 2012 Apr.
Article in English | MEDLINE | ID: mdl-22068888

ABSTRACT

Membrane-bound IgE (mIgE) on B lymphocytes is essential for IgE production. Earlier studies showed that the ε chain of mIgE (mε) on human B cells has a "long" isoform, with an extra "CεmX" domain of 52 amino acid (aa) residues between the CH4 domain and the membrane-anchor segment, as compared to the conventional "short" isoform. Because CεmX provides an antigenic site for targeting IgE-expressing B cells to down-regulate IgE production in patients with allergy, analysis of CεmX in various animals is of great interest. Hence, we analyzed the ε Ig gene, in particular, its membrane exon regions encoding the membrane anchor peptide segment and CεmX domain, of 26 species of the order Primates and 12 species of seven non-Primate orders using data obtained experimentally or retrieved from GenBank. Our analyses reveal the unexpected finding that the genes of three extant tarsier species do not contain the membrane exons for mIgE. Another striking finding is that early evolved Strepsirhini primates such as lemurs and lorises do not have gene segments for the long isoform, whereas New World monkeys such as marmosets and squirrel monkeys allow the transcription of only the long isoform. In Old World monkeys and apes, including humans, the ε gene allows the transcription of both isoforms. This work thus reveals the dramatic differences in the gene segment encoding the mε C terminal region among the four major primate lineages: the Strepsirhini primates, the tarsiers, New World monkeys, and Old World monkeys and apes/humans.


Subject(s)
Evolution, Molecular , Genetic Variation , Immunoglobulin E/genetics , Primates/genetics , Amino Acid Sequence , Animals , Base Sequence , Genomic Library , Humans , Molecular Sequence Data , Phylogeny , Primates/classification , Sequence Analysis, DNA , Sequence Homology, Amino Acid , Sequence Homology, Nucleic Acid , Species Specificity
4.
J Immunol ; 184(4): 1748-56, 2010 Feb 15.
Article in English | MEDLINE | ID: mdl-20083663

ABSTRACT

Membrane-bound IgE (mIgE) is part of the IgE-BCR and is essential for generating isotype-specific IgE responses. On mIgE(+) B cells, the membrane-bound epsilon-chain (mepsilon) exists predominantly in the long isoform, mepsilon(L), containing an extra 52 aa CepsilonmX domain between CH4 and the C-terminal membrane-anchoring segment; the short isoform of mepsilon, mepsilon(S), exists in minor proportions. CepsilonmX thus provides an attractive site for immunologic targeting of mIgE(+) B cells. In this study, we show that nine newly prepared CepsilonmX-specific mAbs, as well as the previously reported a20, bound to mIgE.Fc(L)-expressing CHO cells, while only 4B12 and 26H2 bound to mIgE.Fc(L)-expressing B cell line Ramos cells. The mAb 4B12 bound to the N-terminal part, 26H2 the middle part, and all others the C-terminal part of CepsilonmX. Expression of Igalpha and Igbeta on the mIgE.Fc(L)-CHO cells reduces the binding of a20 to CepsilonmX as compared with that of 4B12 and 26H2. The chimeric mAbs c4B12 and c26H2, when cross-linked by secondary antibodies, lysed mIgE.Fc(L)-Ramos cells by apoptosis through a BCR-dependent caspase pathway. Using PBMCs as the source of effector cells, c4B12 and c26H2 demonstrated Ab-dependent cellular cytotoxicity toward mIgE.Fc(L)-Ramos cells in a dose-dependent fashion. In cultures of PBMCs from atopic dermatitis patients, c4B12 and c26H2 inhibited the synthesis of IgE driven by anti-CD40 and IL-4. These results suggest that 4B12 and 26H2 and an immunogen using the peptide segments recognized by these mAbs are potentially useful for targeting mIgE(+) B cells to control IgE production.


Subject(s)
B-Lymphocyte Subsets/immunology , B-Lymphocyte Subsets/metabolism , Epitopes, B-Lymphocyte/genetics , Epitopes, B-Lymphocyte/metabolism , Gene Targeting , Immunoglobulin E/biosynthesis , Immunoglobulin epsilon-Chains/biosynthesis , Receptors, Antigen, B-Cell/metabolism , Amino Acid Sequence , Animals , Antibody-Dependent Cell Cytotoxicity/genetics , Apoptosis/genetics , Apoptosis/immunology , Binding Sites, Antibody/genetics , CHO Cells , Cell Line , Cell Line, Tumor , Clone Cells , Cricetinae , Cricetulus , Humans , Immunoglobulin E/genetics , Immunoglobulin E/metabolism , Immunoglobulin epsilon-Chains/metabolism , Mice , Mice, Inbred BALB C , Molecular Sequence Data , Protein Isoforms/biosynthesis , Protein Isoforms/genetics , Protein Isoforms/metabolism
5.
Adv Immunol ; 93: 63-119, 2007.
Article in English | MEDLINE | ID: mdl-17383539

ABSTRACT

The pharmacological purposes of the anti-IgE therapy are to neutralize IgE and to inhibit its production to attenuate type I hypersensitivity reactions. The therapy is based on humanized IgG1 antibodies that bind to free IgE and to membrane-bound IgE on B cells, but not to IgE bound by the high-affinity IgE.Fc receptors on basophils and mast cells or by the low-affinity IgE.Fc receptors on B cells. After nearly 20 years since inception, therapeutic anti-IgE antibodies (anti-IgE) have been studied in about 30 Phase II and III clinical trials in many allergy indications, and a lead antibody, omalizumab, has been approved for treating patients (12 years and older) with moderate-to-severe allergic asthma. Anti-IgE has confirmed the roles of IgE in the pathogenesis of asthma and helped define the concept "allergic asthma" in clinical practice. It has been shown to be safe and efficacious in treating pediatric allergic asthma and treating allergic rhinitis and is being investigated for treating peanut allergy, atopic dermatitis, latex allergy, and others. It has potential for use to combine with specific and rush immunotherapy for increased safety and efficacy. Anti-IgE thus appears to provide a prophylactic and therapeutic option for moderate to severe cases of many allergic diseases and conditions in which IgE plays a significant role. This chapter reviews the evolution of the anti-IgE concept and the clinical studies of anti-IgE on various disease indications, and presents a comprehensive analysis on the multiple intricate immunoregulatory pharmacological effects of anti-IgE. Finally, it reviews other approaches that target IgE or IgE-expressing B cells.


Subject(s)
Anti-Allergic Agents/therapeutic use , Antibodies, Anti-Idiotypic/therapeutic use , Hypersensitivity/drug therapy , Hypersensitivity/immunology , Immunoglobulin E/immunology , Immunotherapy/methods , Animals , Antibodies, Anti-Idiotypic/immunology , Antibodies, Monoclonal/therapeutic use , Antibodies, Monoclonal, Humanized , Clinical Trials as Topic , Desensitization, Immunologic , Humans , Omalizumab
SELECTION OF CITATIONS
SEARCH DETAIL
...