Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 46
Filter
Add more filters










Publication year range
1.
Allergy ; 71(7): 957-66, 2016 07.
Article in English | MEDLINE | ID: mdl-26850222

ABSTRACT

BACKGROUND: Venom immunotherapy is efficient to desensitize people suffering from insect sting allergies. However, the numerous injections required over several years and important risks of severe side reactions complicate the widespread use of immunotherapy. In the search for novel approaches to blunt the overwhelming pro-allergic Th2 response, we evaluated the therapeutic efficacy of a treatment based on a denatured form of the major allergen, phospholipase A2, associated with microbubbles (PLA2denat -MB) in a mouse model of honeybee venom allergy. METHODS: Antibodies measured by ELISA, T-cell responses assessed by CFSE-based proliferation assays and ELISA, and basophil degranulation were examined after PLA2denat -MB-based therapeutic treatment of sensitized mice. Mice were challenged with a lethal dose of PLA2 to evaluate protection against anaphylaxis. RESULTS: Therapeutic subcutaneous administration of two different PLA2denat -MB formulations, in contrast to PLA2denat alone, reduced allergic symptoms and protected all mice from anaphylaxis-mediated death after allergen challenge. At the functional level, the use of PLA2denat decreased IgE-mediated basophil degranulation as compared to the native form of the allergen. In comparison with PLA2denat alone, both PLA2denat -MB formulations decreased allergen-specific Th2 CD4 T-cell reactivity. At the mechanistic level, PLA2denat -MB containing 20% palmitic acid and PEG induced PLA2-specific IgA and increased Foxp3(+) Treg frequencies and TGF-ß production, whereas the formulation bearing 80% palmitic acid triggered the production of IFN-γ, IgG2a, and IgG3. CONCLUSIONS: In contrast to conventional PLA2 subcutaneous immunotherapy, the therapeutic administration of PLA2-MB treatment to mice that already had established allergy to PLA2 protects all subsequently challenged animals.


Subject(s)
Allergens/immunology , Bee Venoms/immunology , Bees/immunology , Hypersensitivity/immunology , Hypersensitivity/therapy , Microbubbles , Phospholipases A2/administration & dosage , Anaphylaxis/immunology , Anaphylaxis/therapy , Animals , Antibody Specificity/immunology , Basophils/immunology , Basophils/metabolism , Cell Degranulation/immunology , Cytokines/metabolism , Desensitization, Immunologic , Disease Models, Animal , Female , Hypersensitivity/mortality , Immune Tolerance , Immunoglobulin E/immunology , Immunoglobulin G/blood , Immunoglobulin G/immunology , Insect Bites and Stings/immunology , Mice , Rats , T-Lymphocyte Subsets/immunology , T-Lymphocyte Subsets/metabolism
2.
Clin Exp Allergy ; 46(1): 153-62, 2016 Jan.
Article in English | MEDLINE | ID: mdl-25900397

ABSTRACT

BACKGROUND: People suffering from honeybee venom allergy can be treated by venom immunotherapy, which consists in the subcutaneous injection of increasing doses of allergen extracts over a period of 3-5 years. Such a procedure is time-consuming, and the risks of severe side reactions are important. Approaches based on the use of novel adjuvants to blunt pro-allergic Th2-type immune responses represent a sound alternative. OBJECTIVES: In this study, we evaluated in a mouse model of honeybee venom allergy the protection induced by the prophylactic use of the major allergen phospholipase A2 (PLA2) associated with microbubbles (MB). METHODS: Antibody (Ab) and T cell responses, as detected by ELISA and CFSE-based proliferation assays, were first examined after prophylactic immunization of CBA/J mice with PLA2-MB, and second after sensitization with native PLA2. Mice were eventually challenged with a lethal dose of PLA2 to assess protection against anaphylaxis. RESULTS: Prophylactic immunization with PLA2-MB induced PLA2-specific IgG and IgA Ab, triggered the production of IFN-γ and IL-10 and the differentiation of PLA2-specific Foxp3(+) Treg. Immunized/sensitized mice displayed the following: (1) increased titres of potent blocking IgG1, IgG2a and IgG3 Ab, (2) both reduced allergen-specific T cell proliferation and Th2-type cytokine production and (3) elevated frequencies of specific Foxp3(+) Treg and increased production of TGF-ß, as compared to naïve/sensitized animals. Immunomodulation correlated with reduced signs of anaphylaxis after allergen challenge. CONCLUSIONS AND CLINICAL RELEVANCE: Our data demonstrate the ability of PLA2-MB to prophylactically protect mice against subsequent sensitization and death-inducing PLA2 challenge for up to 4 months, revealing so far unravelled immunomodulatory properties of MB. These data, combined with the safe use of MB as contrast agents for in situ imaging in humans, render them an immunotherapeutic agent of great interest for further evaluation.


Subject(s)
Allergens/immunology , Bee Venoms/immunology , Hypersensitivity, Delayed/immunology , Hypersensitivity, Delayed/prevention & control , Microbubbles , Phospholipases A2/administration & dosage , Pre-Exposure Prophylaxis , Th2 Cells/immunology , Animals , Antibodies/immunology , Antibody Specificity/immunology , Antivenins/immunology , Disease Models, Animal , Female , Hypersensitivity, Delayed/metabolism , Immunization , Immunotherapy , Mice , Th1 Cells/immunology , Th1 Cells/metabolism , Th2 Cells/metabolism
3.
Mucosal Immunol ; 7(2): 315-24, 2014 Mar.
Article in English | MEDLINE | ID: mdl-23839063

ABSTRACT

In celiac disease, enhanced permeability to gliadin peptides can result from their apico-basal transport by secretory immunoglobulin A1 (SIgA1) binding to the CD71 receptor ectopically expressed at the gut epithelial surface. Herein, we have established a mouse model in which there is apico-basal transport of the model antigen ovalbumin (OVA) by specific SIgA1 and have analyzed local T-cell activation. Transgenic DO11.10 mice were grafted with a hybridoma-secreting OVA-specific humanized IgA1, which could bind mouse CD71 and which were released in the intestinal lumen as SIgA. CD71 expression was induced at the gut apical surface by treating the mice with tyrphostin A8. Following gavage of the mice with OVA, OVA-specific CD4⁺ T cells isolated from the mesenteric lymph nodes displayed higher expression of the activation marker CD69 and produced more interferon gamma in mice bearing the hybridoma-secreting OVA-specific IgA1, than in ungrafted mice or in mice grafted with an irrelevant hybridoma. These results indicate that the protective role of SIgA1 might be jeopardized in human pathological conditions associated with ectopic expression of CD71 at the gut surface.


Subject(s)
Immunoglobulin A, Secretory/immunology , Immunoglobulin A, Secretory/metabolism , Intestinal Mucosa/immunology , Intestinal Mucosa/metabolism , Ovalbumin/metabolism , Th1 Cells/immunology , Animals , Antigens, CD/metabolism , CD4-Positive T-Lymphocytes/immunology , Celiac Disease/immunology , Celiac Disease/metabolism , Disease Models, Animal , Enterocytes/drug effects , Enterocytes/metabolism , Female , Humans , Lymph Nodes/immunology , Mesentery , Mice , Mice, Transgenic , Protein Binding , Protein Transport , Receptors, Transferrin/metabolism , Tyrphostins/pharmacology , Up-Regulation/drug effects
4.
Mucosal Immunol ; 4(6): 603-11, 2011 Nov.
Article in English | MEDLINE | ID: mdl-21975936

ABSTRACT

Secretory IgA (SIgA) serves as the first line of defense in protecting the intestinal epithelium from enteric toxins and pathogenic microorganisms. Through a process known as immune exclusion, SIgA promotes the clearance of antigens and pathogenic microorganisms from the intestinal lumen by blocking their access to epithelial receptors, entrapping them in mucus, and facilitating their removal by peristaltic and mucociliary activities. In addition, SIgA functions in mucosal immunity and intestinal homeostasis through mechanisms that have only recently been revealed. In just the past several years, SIgA has been identified as having the capacity to directly quench bacterial virulence factors, influence composition of the intestinal microbiota by Fab-dependent and Fab-independent mechanisms, promote retro-transport of antigens across the intestinal epithelium to dendritic cell subsets in gut-associated lymphoid tissue, and, finally, to downregulate proinflammatory responses normally associated with the uptake of highly pathogenic bacteria and potentially allergenic antigens. This review summarizes the intrinsic biological activities now associated with SIgA and their relationships with immunity and intestinal homeostasis.


Subject(s)
Antigen-Antibody Complex/immunology , Immunoglobulin A, Secretory/immunology , Intestinal Mucosa/immunology , Animals , Antigen Presentation , Dendritic Cells/immunology , Homeostasis , Humans , Immunity, Mucosal , Immunomodulation , Metagenome , Mucociliary Clearance
5.
Clin Exp Allergy ; 41(1): 20-8, 2011 Jan.
Article in English | MEDLINE | ID: mdl-21070397

ABSTRACT

Intestinal permeability is a critical feature of the gastrointestinal epithelium as it must allow an efficient passage of nutrients and restrict the entry of larger molecules, such as protein antigen, in order to facilitate appropriate immune responses towards food antigens. The proper regulation of the epithelial barrier relies on multiple, intricate physiological and immunologic mechanisms, in terms of which recent progresses regarding the cellular and molecular components have been unravelled. In genetically predisposed individuals, breakdown of oral tolerance can occur, leading to the inadequate production of allergen-specific IgE and the recruitment of mast cells in the gastrointestinal mucosa. Under such conditions, the intestinal permeability towards allergen is altered via different mechanisms, with IgE-CD23-mediated transport across the mucosa playing an important amplification role. Additionally, during the effector phase of the allergic reaction, when mast cells degranulate, a series of inflammatory mediators, such as proteases and cytokines, are released and further affects intestinal permeability. This leads to an increase in the passage of allergens and hence contributes to perpetuate the inflammatory reaction. In this review, we describe the importance of properly balanced intestinal permeability in oral tolerance induction and address the processes involved in damaging the intestinal barrier in the sensitized epithelium and during allergic reactions. We conclude by speculating on the effect of increased intestinal permeability on the onset of sensitization towards dietary antigens.


Subject(s)
Food Hypersensitivity/immunology , Food Hypersensitivity/metabolism , Intestinal Mucosa/metabolism , Animals , Humans , Intestines/immunology , Permeability
6.
Clin Exp Allergy ; 40(1): 153-62, 2010 Jan.
Article in English | MEDLINE | ID: mdl-19689461

ABSTRACT

BACKGROUND: Food allergy has reached an epidemic level in westernized countries and although central mechanisms have been described, the variability associated with genetic diversity underscores the still unresolved complexity of these disorders. OBJECTIVE: To develop models of food allergy and oral tolerance, both strictly induced by the intestinal route, and to compare antigen-specific responses. METHODS: BALB/c mice were mucosally sensitized to ovalbumin (OVA) in the presence of the mucosal adjuvant cholera toxin, or tolerized by intra-gastric administrations of OVA alone. Antibody titres and cytokines were determined by ELISA, and allergic status was determined through several physiologic parameters including decline in temperature, diarrhoea, mast cell degranulation and intestinal permeability. RESULTS: OVA-specific antibodies (IgE, IgGs and IgA in serum and feces) were produced in sensitized mice exclusively. Upon intra-gastric challenge with OVA, sensitized mice developed anaphylactic reactions associated with a decline of temperature, diarrhoea, degranulation of mast cells, which were only moderately recruited in the small intestine, and increased intestinal permeability. Cytokines produced by immune cells from sensitized mice included T-helper type 2 cytokines (IL-5, IL-13), but also IL-10, IFN-gamma and IL-17. In contrast, all markers of allergy were totally absent in tolerized animals, and yet the latter were protected from subsequent sensitization, demonstrating that oral tolerance took place efficiently. CONCLUSION: This work allows for the first time an appropriate comparison between sensitized and tolerized BALB/c mice towards OVA. It highlights important differences from other models of allergy, and thus questions some of the generally accepted notions of allergic reactions, such as the protective role of IFN-gamma, the importance of antigen-specific secretory IgA and the role of mucosal mast cells in intestinal anaphylaxis. In addition, it suggests that IL-17 might be an effector cytokine in food allergy. Finally, it demonstrates that intestinal permeability towards the allergen is increased during challenge.


Subject(s)
Allergens/immunology , Antibodies/immunology , Cytokines/immunology , Food Hypersensitivity/immunology , Immune Tolerance , Intestinal Mucosa/immunology , Mast Cells/immunology , Administration, Oral , Animals , Antibodies/blood , Antibody Specificity , Cell Degranulation , Cells, Cultured , Disease Models, Animal , Female , Immunoglobulin A, Secretory/immunology , Interleukin-17/biosynthesis , Interleukin-17/immunology , Intestinal Mucosa/pathology , Lymphocytes/immunology , Lymphocytes/metabolism , Mice , Mice, Inbred BALB C , Ovalbumin/immunology
7.
Clin Exp Allergy ; 39(4): 527-36, 2009 Apr.
Article in English | MEDLINE | ID: mdl-19178541

ABSTRACT

BACKGROUND: Food allergy is a common allergic disorder--especially in early childhood. The avoidance of the allergenic food is the only available method to prevent further reactions in sensitized patients. A better understanding of the immunologic mechanisms involved in this reaction would help to develop therapeutic approaches applicable to the prevention of food allergy. OBJECTIVE: To establish a multi-cell in vitro model of sensitized intestinal epithelium that mimics the intestinal epithelial barrier to study the capacity of probiotic microorganisms to modulate permeability, translocation and immunoreactivity of ovalbumin (OVA) used as a model antigen. METHODS: Polarized Caco-2 cell monolayers were conditioned by basolateral basophils and used to examine apical to basolateral transport of OVA by ELISA. Activation of basophils with translocated OVA was measured by beta-hexosaminidase release assay. This experimental setting was used to assess how microorganisms added apically affected these parameters. Basolateral secretion of cytokine/chemokines by polarized Caco-2 cell monolayers was analysed by ELISA. RESULTS: Basophils loaded with OVA-specific IgE responded to OVA in a dose-dependent manner. OVA transported across polarized Caco-2 cell monolayers was found to trigger basolateral basophil activation. Microorganisms including lactobacilli and Escherichia coli increased transepithelial electrical resistance while promoting OVA passage capable to trigger basophil activation. Non-inflammatory levels of IL-8 and thymic stromal lymphopoietin were produced basolaterally by Caco-2 cells exposed to microorganisms. CONCLUSION: The complex model designed in here is adequate to learn about the consequence of the interaction between microorganisms and epithelial cells vis-a-vis the barrier function and antigen translocation, two parameters essential to mucosal homeostasis. It can further serve as a direct tool to search for microorganisms with anti-allergic and anti-inflammatory properties.


Subject(s)
Basophils/immunology , Cell Membrane Permeability/immunology , Intestinal Mucosa/immunology , Intestinal Mucosa/microbiology , Models, Immunological , Animals , Basophil Degranulation Test , Caco-2 Cells , Cell Line, Tumor , Coculture Techniques , Cytokines/immunology , Escherichia coli/immunology , Female , Humans , Lactobacillus/immunology , Mice , Mice, Inbred BALB C , Ovalbumin/immunology , Rats , beta-N-Acetylhexosaminidases/analysis , beta-N-Acetylhexosaminidases/immunology
8.
Clin Exp Allergy ; 36(6): 803-14, 2006 Jun.
Article in English | MEDLINE | ID: mdl-16776682

ABSTRACT

BACKGROUND: Food allergy in developed countries represents a growing concern as reflected by epidemiological studies, indicating that up to 4% of the overall population is affected. Reduction of symptoms takes place following eviction or processing of some allergens. However, it cannot be predicted which structural changes will be associated with significant effects on the allergenicity. OBJECTIVE: To determine how various treatments of bovine beta-lactoglobulin (BLG) used as a model antigen alters its immunoreactivity and transepithelial transport, and whether this correlates with reduced allergenicity using an in vitro basophil activation assay. METHODS: BLG was subjected to reduction/alkylation, trypsin digestion or exposed to Lactococcus lactis. The remaining immunoreactivity toward IgG raised against native BLG was assessed by ELISA. Transepithelial transport of BLG and derivatives was examined using polarized Caco-2 cell monolayers mimicking the intestinal epithelium. Selective passage of tryptic peptides was determined using colchicine and cytochalasin D. Basophil activation was measured following stimulation with BLG and derivatives. RESULTS: Reduction/alkylation, trypsin digestion or incubation with L. lactis was associated with decreased BLG recognition by IgG antibodies raised against the native protein. All treatments also resulted in a more efficient transepithelial transport of BLG. BLG crossed the Caco-2 monolayer through passage across the cell, whereas tryptic peptides followed both the para- and transcellular routes. With the exception of denaturation by reduction/alkylation, cross-linking of IgE antibodies by BLG derivatives led to lower basophil degranulation. CONCLUSION: In vitro dissection of antigenicity and allergenicity may be a valid and convenient alternative to evaluate the effects of biotechnological processing on dietary proteins. In addition, it can help to define the molecular and cellular mechanisms that will provide improved means of diagnosis and possibly therapy of food-allergic disorders.


Subject(s)
Allergens/administration & dosage , Intestinal Mucosa/metabolism , Lactococcus lactis , Lactoglobulins/metabolism , Milk Hypersensitivity/immunology , Allergens/immunology , Animals , Basophils/immunology , Caco-2 Cells , Cattle , Cell Line, Tumor , Enzyme-Linked Immunosorbent Assay/methods , Humans , Hydrolysis , Immunoglobulin G/immunology , Intestinal Mucosa/microbiology , Lactoglobulins/analysis , Lactoglobulins/immunology , Microscopy, Confocal , Microscopy, Fluorescence , Protein Transport , Rats
10.
J Immunol ; 166(10): 6250-6, 2001 May 15.
Article in English | MEDLINE | ID: mdl-11342648

ABSTRACT

Both IgG and IgA Abs have been implicated in host defense against bacterial infections, although their relative contributions remain unclear. We generated a unique panel of human chimeric Abs of all human IgG and IgA subclasses with identical V genes against porin A, a major subcapsular protein Ag of Neisseria meningitidis and a vaccine candidate. Chimeric Abs were produced in baby hamster kidney cells, and IgA-producing clones were cotransfected with human J chain and/or human secretory component. Although IgG (isotypes IgG1-3) mediated efficient complement-dependent lysis, IgA was unable to. However, IgA proved equally active to IgG in stimulating polymorphonuclear leukocyte respiratory burst. Remarkably, although porin-specific monomeric, dimeric, and polymeric IgA triggered efficient phagocytosis, secretory IgA did not. These studies reveal unique and nonoverlapping roles for IgG and IgA Abs in defense against meningococcal infections.


Subject(s)
Immunity, Innate , Immunoglobulin A/physiology , Immunoglobulin G/physiology , Neisseria meningitidis/immunology , Animals , Bacteriolysis/genetics , Bacteriolysis/immunology , Binding Sites, Antibody/genetics , Blood Bactericidal Activity/genetics , Blood Bactericidal Activity/immunology , Cell Line , Complement Activation/immunology , Cricetinae , Dimerization , Humans , Immunity, Innate/genetics , Immunoglobulin A/biosynthesis , Immunoglobulin A/genetics , Immunoglobulin A/metabolism , Immunoglobulin A, Secretory/metabolism , Immunoglobulin G/biosynthesis , Immunoglobulin G/genetics , Immunoglobulin G/metabolism , Neutrophils/immunology , Neutrophils/metabolism , Phagocytosis/genetics , Phagocytosis/immunology , Porins/immunology , Receptors, Fc/metabolism , Recombinant Fusion Proteins/biosynthesis , Recombinant Fusion Proteins/metabolism , Respiratory Burst/immunology
11.
J Immunol ; 166(5): 3612-21, 2001 Mar 01.
Article in English | MEDLINE | ID: mdl-11207323

ABSTRACT

Phospholipase A(2) (PLA(2)) is one of the major honey bee venom allergens for humans. To assess the long-term prevention of allergic reactions by DNA vaccination, a PLA(2)-CBA/J mouse model was employed using empty or PLA(2) sequence-carrying DNA plasmids. Early skin application of either DNA construct before (prophylactic approach) or after (therapeutic approach) sensitization with PLA(2)/alum led to reduced PLA(2)-specific IgE and IgG1 titers at 7 mo, with concomitant rise in IgG2a and IgG3. Splenocytes recovered at 5-6 mo after the last DNA administration exhibited a sustained IFN-gamma and IL-10 secretion and reduced IL-4 production. Recall challenge with PLA(2) boosted IFN-gamma and IL-10 secretion, suggesting the reactivation of quiescent memory Th1 lymphocytes. Mice from the prophylactic groups were fully protected against anaphylaxis, whereas 65% of the animals recovered in the therapeutic groups. Th1-polarized immune responses were also active in mice vaccinated with an empty plasmid 32 wk before sensitization with another Ag (OVA). This is the first demonstration that the Ag-coding sequence in DNA vaccine is not necessary to promote immune modulation in naive and sensitized animals for a prolonged period, and has relevance for the understanding of the innate and induced mechanisms underlying gene immunotherapy in long-term treatment of allergy.


Subject(s)
Antigens/physiology , Bee Venoms/immunology , Desensitization, Immunologic/methods , Immunosuppressive Agents/administration & dosage , Phospholipases A/immunology , Vaccines, DNA/immunology , Adjuvants, Immunologic/administration & dosage , Adjuvants, Immunologic/genetics , Adjuvants, Immunologic/therapeutic use , Anaphylaxis/immunology , Anaphylaxis/prevention & control , Animals , Antibody Specificity/genetics , Antigens/administration & dosage , Bee Venoms/administration & dosage , CHO Cells , Cells, Cultured , Cricetinae , Cytokines/biosynthesis , Cytokines/metabolism , Female , Genetic Vectors/administration & dosage , Genetic Vectors/immunology , Genetic Vectors/therapeutic use , Immunoglobulin E/biosynthesis , Immunoglobulin G/biosynthesis , Lymphocyte Activation/genetics , Mice , Mice, Inbred CBA , Ovalbumin/administration & dosage , Ovalbumin/immunology , Peptide Fragments/administration & dosage , Peptide Fragments/genetics , Peptide Fragments/immunology , Phospholipases A/administration & dosage , Phospholipases A/genetics , T-Lymphocyte Subsets/immunology , T-Lymphocyte Subsets/metabolism , Th1 Cells/immunology , Th1 Cells/metabolism , Transfection , Up-Regulation/genetics , Up-Regulation/immunology , Vaccines, DNA/administration & dosage , Vaccines, DNA/therapeutic use
12.
Tumour Biol ; 22(1): 36-44, 2001.
Article in English | MEDLINE | ID: mdl-11054025

ABSTRACT

Single chain Fv antibody fragments (scFv) binding to purified Helicobacter pylori urease were selected from a nonimmune human antibody repertoire displayed on filamentous phage. After three rounds of screening on solid phase urease, 44 clones were found to bind the enzyme and four distinct scFv were identified by sequencing their heavy and light chain variable region genes (V(H) and V(L)). Two of the selected human scFv (scFv B4 and scFv D9) inhibited the activity of H. pylori urease with inhibitory constants (K(i)) of 7 and 2 microM, respectively. Their affinity (K(d)) for H. pylori urease as determined by surface plasmon resonance ranged from 17 to 42 nM. Both scFv were able to bind to urease present on the surface of living H. pylori organisms as demonstrated by flow cytometry analysis. The binding sites of scFv B4 and D9 were mapped by the use of two random hexapeptide libraries (X6 and CX6C) displayed on filamentous bacteriophage. The selected peptide sequences were shown to inhibit scFv binding to H. pylori urease and thus could be used in a vaccination strategy as epitopes mimicking (mimotopes) the region of urease recognized by these human scFv antibody fragments.


Subject(s)
Antibodies, Bacterial/immunology , Bacterial Proteins/immunology , Helicobacter pylori/immunology , Immunoglobulin Fragments/immunology , Urease/immunology , Amino Acid Sequence , Antibodies, Bacterial/isolation & purification , Antibodies, Bacterial/metabolism , Antibodies, Bacterial/pharmacology , Antibody Affinity , Antibody Specificity , Antigen-Antibody Reactions , Bacterial Proteins/antagonists & inhibitors , Bacterial Proteins/metabolism , Bacterial Vaccines , Drug Design , Enzyme Inhibitors/immunology , Enzyme Inhibitors/isolation & purification , Enzyme-Linked Immunosorbent Assay , Epitopes/immunology , Helicobacter pylori/enzymology , Humans , Immunoglobulin Fragments/isolation & purification , Immunoglobulin Fragments/metabolism , Immunoglobulin Fragments/pharmacology , Molecular Mimicry , Molecular Sequence Data , Peptide Library , Sequence Alignment , Sequence Homology, Amino Acid , Surface Plasmon Resonance , Urease/antagonists & inhibitors , Urease/metabolism
13.
J Immunol ; 165(5): 2544-55, 2000 Sep 01.
Article in English | MEDLINE | ID: mdl-10946281

ABSTRACT

T560, a mouse B lymphoma that originated in gut-associated lymphoid tissue, expresses receptors that bind dimeric IgA and IgM in a mutually inhibitory manner but have little affinity for monomeric IgA. Evidence presented in this paper indicates that the receptor is poly-Ig receptor (pIgR) known in humans and domestic cattle to bind both IgA and IgM. The evidence includes the demonstration that binding of IgM is J chain dependent, and that pIg-precipitated receptor has an appropriate Mr of 116-120 kDa and can be detected on immunoblots with specific rabbit anti-mouse pIgR. Overlapping RT-PCR performed using template mRNA from T560 cells and oligonucleotide primer pairs designed from the published sequence of mouse liver pIgR indicate that T560 cells express mRNA virtually identical with that of the epithelial cell pIgR throughout its external, transmembrane, and intracytoplasmic coding regions. Studies using mutant IgAs suggest that the Calpha2 domain of dimeric IgA is not involved in high-affinity binding to the T560 pIgR. Inasmuch as this mouse B cell pIgR binds IgM better than IgA, it is similar to human pIgR and differs from rat, mouse, and rabbit epithelial cell pIgRs that bind IgA but not IgM. Possible explanations for this difference are discussed. All clones of T560 contain some cells that spontaneously secrete both IgG2a and IgA, but all of the IgA recoverable from the medium and from cell lysates is monomeric; it cannot be converted to secretory IgA by T560 cells.


Subject(s)
Immunoglobulin A/metabolism , Immunoglobulin M/metabolism , Lymphoma, B-Cell/immunology , Receptors, Fc/biosynthesis , Receptors, Polymeric Immunoglobulin/biosynthesis , Amino Acid Sequence , Animals , Base Sequence , Binding Sites, Antibody , Binding, Competitive/immunology , Carbohydrate Metabolism , Carbohydrates/immunology , Chemical Precipitation , Cross Reactions , Enzyme Activation/immunology , Epitopes, B-Lymphocyte/metabolism , Epitopes, T-Lymphocyte/metabolism , Humans , Immunoblotting , Immunoglobulin A, Secretory/metabolism , Immunoglobulin J-Chains/physiology , Lymphoma, B-Cell/metabolism , Mice , Mice, Inbred BALB C , Molecular Sequence Data , Molecular Weight , Phosphatidylinositol Diacylglycerol-Lyase , Protein Kinase C/metabolism , RNA, Messenger/biosynthesis , Rats , Receptors, Fc/isolation & purification , Receptors, Polymeric Immunoglobulin/genetics , Receptors, Polymeric Immunoglobulin/isolation & purification , Tumor Cells, Cultured , Type C Phospholipases/metabolism
14.
J Immunol ; 164(4): 1952-60, 2000 Feb 15.
Article in English | MEDLINE | ID: mdl-10657645

ABSTRACT

The two exotoxins A and B produced by Clostridium difficile are responsible for antibiotic-associated enterocolitis in human and animals. When added apically to human colonic carcinoma-derived T84 cell monolayers, toxin A, but not toxin B, abolished the transepithelial electrical resistance and altered the morphological integrity. Apical addition of suboptimal concentration of toxin A made the cell monolayer sensitive to toxin B. Both toxins induced drastic and rapid epithelial alterations when applied basolaterally with a complete disorganization of tight junctions and vacuolization of the cells. Toxin A-specific IgG2a from hybridoma PCG-4 added apically with toxin A alone or in combination with toxin B abolished the toxin-induced epithelial alterations for up to 8 h. The Ab neutralized basolateral toxin A for 4 h, but not the mixture of the two toxins. Using an identical Ab:Ag ratio, we found that recombinant polymeric IgA (IgAd/p) with the same Fv fragments extended protection against toxin A for at least 24 h in both compartments. In contrast, the recombinant monomeric IgA counterpart behaved as the PCG-4 IgG2a Ab. The direct comparison between different Ig isotype and molecular forms, but of unique specificity, demonstrates that IgAd/p Ab is more efficient in neutralizing toxin A than monomeric IgG and IgA. We conclude that immune protection against C. difficile toxins requires toxin A-specific secretory Abs in the intestinal lumen and IgAd/p specific for both toxins in the lamina propria.


Subject(s)
Bacterial Proteins , Bacterial Toxins/toxicity , Clostridioides difficile/immunology , Enterotoxins/toxicity , Epithelial Cells/immunology , Immunoglobulin A/physiology , Immunoglobulin G/physiology , Immunoglobulin Variable Region/physiology , Antibody Specificity , Bacterial Toxins/antagonists & inhibitors , Bacterial Toxins/immunology , Cell Line , Cell Membrane/immunology , Cell Membrane/metabolism , Enterotoxins/antagonists & inhibitors , Enterotoxins/immunology , Epithelial Cells/pathology , Humans , Immunoglobulin G/pharmacology , Tight Junctions/immunology , Tight Junctions/pathology , Time Factors
15.
Biol Chem ; 380(11): 1251-62, 1999 Nov.
Article in English | MEDLINE | ID: mdl-10614817

ABSTRACT

Immune responses taking place in mucosal tissues are typified by secretory immunoglobulin A (S-IgA) molecules, which are assembled from proteins expressed in two cell lineages. The heavy and light chains as well as the J chain are produced in plasma cells, whereas the secretory component (SC) is associated to the immunoglobulin complex during transcytosis across the epithelial layer. S-IgA antibodies represent the predominant immunoglobulin class in external secretions, and the best defined entity providing specific immune protection for mucosal surfaces by blocking attachment of bacteria and viruses. S-IgA constitutes greater than 80% of all antibodies produced in mucosa-associated lymphoid tissues in humans. The existence of a common mucosal immune system permits immunization on one mucosal surface to induce secretion of antigen-specific S-IgA at distant sites. In addition, S-IgA antibodies not only function in external secretions, but also exert their antimicrobial properties within the epithelial cell during transport across the epithelium. Passive mucosal delivery of monoclonal IgA molecules neutralizes pathogens responsible for gastrointestinal and respiratory infections. Mucosal and systemic immunity can be achieved by orally administered recombinant S-IgA molecules carrying a protective bacterial epitope within the SC polypeptide primary sequence.


Subject(s)
Immunity, Mucosal , Immunization , Immunoglobulin A, Secretory/genetics , Immunoglobulin A, Secretory/immunology , Animals , Antigens/metabolism , Humans , Immunity, Mucosal/drug effects , Immunoglobulin A, Secretory/chemistry , Inflammation/immunology , Recombinant Proteins/genetics , Recombinant Proteins/immunology , Recombinant Proteins/pharmacology , Vaccines/immunology
16.
J Biol Chem ; 274(44): 31445-55, 1999 Oct 29.
Article in English | MEDLINE | ID: mdl-10531346

ABSTRACT

Recombinant secretory immunoglobulin A containing a bacterial epitope in domain I of the secretory component (SC) moiety can serve as a mucosal delivery vehicle triggering both mucosal and systemic responses (Corthésy, B., Kaufmann, M., Phalipon, A., Peitsch, M., Neutra, M. R., and Kraehenbuhl, J.-P. (1996) J. Biol. Chem. 271, 33670-33677). To load recombinant secretory IgA with multiple B and T epitopes and extend its biological functions, we selected, based on molecular modeling, five surface-exposed sites in domains II and III of murine SC. Loops predicted to be exposed at the surface of SC domains were replaced with the DYKDDDDK octapeptide (FLAG). Another two mutants were obtained with the FLAG inserted in between domains II and III or at the carboxyl terminus of SC. As shown by mass spectrometry, internal substitution of the FLAG into four of the mutants induced the formation of disulfide-linked homodimers. Three of the dimers and two of the monomers from SC mutants could be affinity-purified using an antibody to the FLAG, mapping them as candidates for insertion. FLAG-induced dimerization also occurred with the polymeric immunoglobulin receptor (pIgR) and might reflect the so-far nondemonstrated capacity of the receptor to oligomerize. By co-expressing in COS-7 cells and epithelial Caco-2 cells two pIgR constructs tagged at the carboxyl terminus with hexahistidine or FLAG, we provide the strongest evidence reported to date that the pIgR dimerizes noncovalently in the plasma membrane in the absence of polymeric IgA ligand. The implication of this finding is discussed in terms of IgA transport and specific antibody response at mucosal surfaces.


Subject(s)
Immunoconjugates/metabolism , Immunoglobulin A, Secretory/metabolism , Receptors, Polymeric Immunoglobulin/metabolism , Animals , COS Cells , Caco-2 Cells , Cell Membrane , Dimerization , Enzyme-Linked Immunosorbent Assay , Epitopes , Humans , Immunoconjugates/chemistry , Immunoglobulin A, Secretory/chemistry , Ligands , Mice , Models, Molecular , Oligopeptides , Peptides , Precipitin Tests , Protein Engineering , Receptors, Polymeric Immunoglobulin/chemistry , Spectrometry, Mass, Matrix-Assisted Laser Desorption-Ionization
17.
J Biol Chem ; 274(44): 31456-62, 1999 Oct 29.
Article in English | MEDLINE | ID: mdl-10531347

ABSTRACT

We have identified sites for epitope insertion in the murine secretory component (SC) by replacing individual surface-exposed loops in domains I, II, and III with the FLAG sequence (Crottet, P., Peitsch, M. C., Servis, C., and Corthésy, B. (1999) J. Biol. Chem. 274, 31445-31455). We had previously shown that epitope-carrying SC reassociated with dimeric IgA (IgA(d)) can serve as a mucosal delivery vehicle. When analyzing the capacity of SC mutants to associate with IgA(d), we found that all domain II and III mutants bound specifically with immobilized IgA(d), and their affinity for IgA(d) was comparable to that of the wild type protein (IC(50) approximately 1 nM). We conclude that domains II and III in SC are permissive to local mutation and represent convenient sites to antigenize the SC molecule. No mutant bound to monomeric IgA. SC mutants exposing the FLAG at their surface maintained this property once bound to IgA(d), thereby defining regions not required for high affinity binding to IgA(d). Association of IgA(d) with SC mutants carrying a buried FLAG did not expose de novo the epitope, consistent with limited, local changes in the SC structure upon binding. Only wild type and two mutant SCs bound covalently to IgA(d), thus implicating domains II and III in the correct positioning of the reactive cysteine in SC. This establishes that the integrity of murine SC domains II and III is not essential to preserve specific IgA(d) binding but is necessary for covalency to take place. Finally, SC mutants existing in the monomeric and dimeric forms exhibited the same IgA(d) binding capacity as monomeric wild type SC known to bind with a 1:1 stoichiometry.


Subject(s)
Immunoglobulin A, Secretory/metabolism , Receptors, Polymeric Immunoglobulin/metabolism , Animals , Binding Sites , Dimerization , Epitopes , Immunoglobulin A/metabolism , Mice , Oligopeptides , Peptides , Protein Binding , Protein Structure, Tertiary , Receptors, Polymeric Immunoglobulin/genetics
18.
Eur J Biochem ; 262(3): 774-80, 1999 Jun.
Article in English | MEDLINE | ID: mdl-10411639

ABSTRACT

Urease is an important virulence factor for Helicobacter pylori and is critical for bacterial colonization of the human gastric mucosa. Specific inhibition of urease activity has been proposed as a possible strategy to fight this bacteria which infects billions of individual throughout the world and can lead to severe pathological conditions in a limited number of cases. We have selected peptides which specifically bind and inhibit H. pylori urease from libraries of random peptides displayed on filamentous phage in the context of pIII coat protein. Screening of a highly diverse 25-mer combinatorial library and two newly constructed random 6-mer peptide libraries on solid phase H. pylori urease holoenzyme allowed the identification of two peptides, 24-mer TFLPQPRCSALLRYLSEDGVIVPS and 6-mer YDFYWW that can bind and inhibit the activity of urease purified from H. pylori. These two peptides were chemically synthesized and their inhibition constants (Ki) were found to be 47 microM for the 24-mer and 30 microM for the 6-mer peptide. Both peptides specifically inhibited the activity of H. pylori urease but not that of Bacillus pasteurii.


Subject(s)
Bacterial Proteins/antagonists & inhibitors , Bacteriophage M13/genetics , Helicobacter pylori/enzymology , Peptide Library , Urease/antagonists & inhibitors , Amino Acid Sequence , Bacterial Proteins/isolation & purification , Base Sequence , Enzyme Inhibitors/pharmacology , Helicobacter pylori/chemistry , Holoenzymes/metabolism , Kinetics , Molecular Sequence Data , Peptides/genetics , Peptides/pharmacology , Protein Binding , Substrate Specificity , Urease/isolation & purification
19.
Biochem J ; 341 ( Pt 2): 299-306, 1999 Jul 15.
Article in English | MEDLINE | ID: mdl-10393086

ABSTRACT

Reconstitution of secretory IgA (S-IgA) by the association in vitro of secretory component (SC) and polymeric IgA (pIgA) obtained from hybridomas is a valuable tool in the study of the structure-function relationship in this particular class of antibody. Although dimeric IgA (dIgA) can be obtained and purified from hybridoma clones, SC remains tedious to isolate in sufficient amounts from colostral milk. Several murine models for the study of mucosal immunity are available, which could potentially benefit from the use of cognate IgA antibodies in various molecular forms, including dIgA and S-IgA. We report here on the establishment of two expression systems allowing the production of milligram amounts of pure recombinant murine SC (rmSC) with preserved murine pIgA-binding capability. The first system relies on the use of recombinant vaccinia virus to prompt infected HeLa cells to express the murine SC protein, whereas the second system is based on a stably transfected cell clone exhibiting murine glycosylation. The second source of rmSC will permit the study of the role of its sugar moieties in pathogen-host interactions, and the evaluation of its function in passive protection without risking adverse immune responses. The extensive biochemical characterization conducted in this study demonstrates that rmSC is a dependable and convenient alternative to the natural product, and indicates that the J chain is dispensable in the recognition of pIgA and SC in vitro, whereas it is required for proper pIgA-polymeric Ig receptor interaction in vivo.


Subject(s)
Immunoglobulin A , Secretory Component , Animals , Chromatography, Affinity , Cloning, Molecular , Immunity, Mucosal , Immunoglobulin A/genetics , Immunoglobulin A/immunology , Mice , Plasmids , Recombinant Proteins/genetics , Recombinant Proteins/immunology , Recombinant Proteins/isolation & purification , Secretory Component/genetics , Secretory Component/immunology , Secretory Component/isolation & purification
20.
Proc Natl Acad Sci U S A ; 96(6): 3029-34, 1999 Mar 16.
Article in English | MEDLINE | ID: mdl-10077631

ABSTRACT

The hallmark of a mucosal immune response is the production of antigen-specific secretory IgA (S-IgA) antibodies in external secretions. S-IgA consists of ten polypeptides produced in two different cell lineages. The heavy and light chains in plasma cells assemble into IgA, which on association with J chain become polymerized, whereas secretory component (SC) is added during transport across the epithelium. Recombinant chimeric mouse-human monomeric, dimeric, and S-IgA antibodies have been produced in a single CHO cell sequentially transfected with expression vectors carrying three independent selective markers for chimeric heavy and light chains, human J chain, and human SC, respectively. Biochemical characterization of the various molecular forms indicates that the assembly of the various polypeptides resulted in species of the expected size and covalence. All chimeric IgA antibodies retained the antigen-binding capacity of the parent mouse IgA antibody. The resistance of S-IgA to protease-rich intestinal washes was enhanced when compared with dimeric IgA lacking associated SC. Up to 20 micrograms of recombinant S-IgA per 1 x 10(6) cells were recovered in 24 h with the best producing clones. We conclude that CHO cells programmed de novo with four different genetic elements can assemble functional chimeric S-IgA.


Subject(s)
CHO Cells , Immunoglobulin A/immunology , Animals , Antigen-Antibody Reactions , Cell Lineage , Cricetinae , Humans , Immunoglobulin A/biosynthesis , Immunoglobulin A/genetics , Mice , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/immunology
SELECTION OF CITATIONS
SEARCH DETAIL
...