Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 104
Filter
1.
Antibodies (Basel) ; 12(4)2023 Nov 16.
Article in English | MEDLINE | ID: mdl-37987253

ABSTRACT

We have previously produced a toolkit of antibodies, comprising recombinant human antibodies of all but one of the human isotypes, directed against the polcalcin family antigen Phl p 7. In this work, we complete the toolkit of human antibody isotypes with the IgD version of the anti-Phl p 7 monoclonal antibody. We also raised a set of nanobodies against the IgD anti-Phl p 7 antibody and identify and characterize one paratope-specific nanobody. This nanobody also binds to the IgE isotype of this antibody, which shares the same idiotype, and orthosterically inhibits the interaction with Phl p 7. The 2.1 Å resolution X-ray crystal structure of the nanobody in complex with the IgD Fab is described.

2.
Nat Struct Mol Biol ; 30(7): 866-869, 2023 Jul.
Article in English | MEDLINE | ID: mdl-37433905
3.
Mol Immunol ; 159: 28-37, 2023 07.
Article in English | MEDLINE | ID: mdl-37267832

ABSTRACT

Antibodies of the IgD isotype remain the least well characterized of the mammalian immunoglobulin isotypes. Here we report three-dimensional structures for the Fab region of IgD, based on four different crystal structures, at resolutions of 1.45-2.75 Å. These IgD Fab crystals provide the first high-resolution views of the unique Cδ1 domain. Structural comparisons identify regions of conformational diversity within the Cδ1 domain, as well as among the homologous domains of Cα1, Cγ1 and Cµ1. The IgD Fab structure also possesses a unique conformation of the upper hinge region, which may contribute to the overall disposition of the very long linker sequence between the Fab and Fc regions found in human IgD. Structural similarities observed between IgD and IgG, and differences with IgA and IgM, are consistent with predicted evolutionary relationships for the mammalian antibody isotypes.


Subject(s)
Immunoglobulin Fab Fragments , Immunoglobulin Isotypes , Animals , Humans , Mammals
4.
Plants (Basel) ; 12(7)2023 Apr 04.
Article in English | MEDLINE | ID: mdl-37050178

ABSTRACT

Impatiens glandulifera or Himalayan balsam is one of the most invasive weeds across Europe and can seriously reduce native plant diversity. It often forms continuous monocultures along river banks, but the mechanisms of this arrested succession are largely unknown. Here, we investigated the effect of arbuscular mycorrhizal (AM) fungi on balsam competitive ability with two native plant species, Plantago lanceolata and Holcus lanatus. We also studied how competition with Impatiens affects colonisation by foliar endophytes and mycorrhizas of two other co-occurring native species, Urtica dioica and Cirsium arvense. Mycorrhizal colonisation reduced balsam growth when the plants were grown singly, but appeared to have little effect when balsam experienced intra- or interspecific competition. Competition with balsam together with the addition of mycorrhizas had no effect on P. lanceolata biomass, suggesting that the fungi were beneficial to the latter, enabling it to compete effectively with balsam. However, this was not so with H. lanatus. Meanwhile, competition with Impatiens reduced endophyte numbers and mycorrhizal colonisation in U. dioica and C. arvense, leading to enhanced susceptibility of these plants to insect attack. Himalayan balsam is known to degrade soil fungal populations and can also reduce foliar beneficial fungi in neighbouring plants. This allows the plant to compete effectively with itself and other native species, thereby leading to the continuous monocultures.

5.
Int J Mol Sci ; 22(13)2021 Jun 29.
Article in English | MEDLINE | ID: mdl-34209905

ABSTRACT

Both non-immune "natural" and antigen-induced "immune" IgM are important for protection against pathogens and for regulation of immune responses to self-antigens. Since the bona fide IgM Fc receptor (FcµR) was identified in humans by a functional cloning strategy in 2009, the roles of FcµR in these IgM effector functions have begun to be explored. In this short essay, we describe the differences between human and mouse FcµRs in terms of their identification processes, cellular distributions and ligand binding activities with emphasis on our recent findings from the mutational analysis of human FcµR. We have identified at least three sites of human FcµR, i.e., Asn66 in the CDR2, Lys79 to Arg83 in the DE loop and Asn109 in the CDR3, responsible for its constitutive IgM-ligand binding. Results of computational structural modeling analysis are consistent with these mutational data and a model of the ligand binding, Ig-like domain of human FcµR is proposed. Serendipitously, substitution of Glu41 and Met42 in the CDR1 of human FcµR with mouse equivalents Gln and Leu, either single or more prominently in combination, enhances both the receptor expression and IgM binding. These findings would help in the future development of preventive and therapeutic interventions targeting FcµR.


Subject(s)
Apoptosis Regulatory Proteins/genetics , Apoptosis Regulatory Proteins/metabolism , Membrane Proteins/genetics , Membrane Proteins/metabolism , Animals , Apoptosis Regulatory Proteins/chemistry , Binding Sites , Cloning, Molecular , Humans , Immunoglobulin M/metabolism , Ligands , Membrane Proteins/chemistry , Mice , Models, Molecular , Mutation , Protein Binding , Protein Conformation
6.
FEBS Open Bio ; 11(7): 1827-1840, 2021 07.
Article in English | MEDLINE | ID: mdl-34075727

ABSTRACT

Immunoglobulin E (IgE) is a central regulatory and triggering molecule of allergic immune responses. IgE's interaction with CD23 modulates both IgE production and functional activities.CD23 is a noncanonical immunoglobulin receptor, unrelated to receptors of other antibody isotypes. Human CD23 is a calcium-dependent (C-type) lectin-like domain that has apparently lost its carbohydrate-binding capability. The calcium-binding site classically required for carbohydrate binding in C-type lectins is absent in human CD23 but is present in the murine molecule. To determine whether the absence of this calcium-binding site affects the structure and function of human CD23, CD23 mutant proteins with increasingly "murine-like" sequences were generated. Restoration of the calcium-binding site was confirmed by NMR spectroscopy, and structures of mutant human CD23 proteins were determined by X-ray crystallography, although no electron density for calcium was observed. This study offers insights into the evolutionary differences between murine and human CD23 and some of the functional differences between CD23 in different species.


Subject(s)
Calcium , Receptors, IgE , Animals , Binding Sites , Calcium/metabolism , Crystallography, X-Ray , Humans , Immunoglobulin E/metabolism , Lectins, C-Type , Mice , Receptors, IgE/chemistry , Receptors, IgE/metabolism
7.
Allergy ; 75(9): 2309-2318, 2020 09.
Article in English | MEDLINE | ID: mdl-32248566

ABSTRACT

BACKGROUND: Understanding the discrepancy between IgE sensitization and allergic reactions to peanut could facilitate diagnosis and lead to novel means of treating peanut allergy. OBJECTIVE: To identify differences in IgE and IgG4 binding to peanut peptides between peanut-allergic (PA) and peanut-sensitized but tolerant (PS) children. METHODS: PA (n = 56), PS (n = 42) and nonsensitized nonallergic (NA, n = 10) patients were studied. Synthetic overlapping 15-mer peptides of peanut allergens (Ara h 1-11) were spotted onto microarray slides, and patients' samples were tested for IgE and IgG4 binding using immunofluorescence. IgE and IgG4 levels to selected peptides were quantified using ImmunoCAP. Diagnostic model comparisons were performed using likelihood-ratio tests between each specified nominal logistic regression models. RESULTS: Seven peptides on Ara h 1, Ara h 2, and Ara h 3 were bound more by IgE of PA compared to PS patients on the microarray. IgE binding to one peptide on Ara h 5 and IgG4 binding to one Ara h 9 peptide were greater in PS than in PA patients. Using ImmunoCAP, IgE to the Ara h 2 peptides enhanced the diagnostic accuracy of Ara h 2-specific IgE. Ratios of IgG4/IgE to 4 out of the 7 peptides were higher in PS than in PA subjects. CONCLUSIONS: Ara h 2 peptide-specific IgE added diagnostic value to Ara h 2-specific IgE. Ability of peptide-specific IgG4 to surmount their IgE counterpart seems to be important in established peanut tolerance.


Subject(s)
Antigens, Plant , Peanut Hypersensitivity , 2S Albumins, Plant , Allergens , Arachis , Child , Epitopes , Humans , Immunoglobulin E , Peanut Hypersensitivity/diagnosis , Plant Proteins
8.
Acta Crystallogr F Struct Biol Commun ; 76(Pt 3): 116-129, 2020 Mar 01.
Article in English | MEDLINE | ID: mdl-32133997

ABSTRACT

Immunoglobulin E (IgE) plays a central role in the allergic response, in which cross-linking of allergen by FcεRI-bound IgE triggers mast cell and basophil degranulation and the release of inflammatory mediators. The high-affinity interaction between IgE and FcεRI is a long-standing target for therapeutic intervention in allergic disease. Omalizumab is a clinically approved anti-IgE monoclonal antibody that binds to free IgE, also with high affinity, preventing its interaction with FcεRI. All attempts to crystallize the pre-formed complex between the omalizumab Fab and the Fc region of IgE (IgE-Fc), to understand the structural basis for its mechanism of action, surprisingly failed. Instead, the Fab alone selectively crystallized in different crystal forms, but their structures revealed intermolecular Fab/Fab interactions that were clearly strong enough to disrupt the Fab/IgE-Fc complexes. Some of these interactions were common to other Fab crystal structures. Mutations were therefore designed to disrupt two recurring packing interactions observed in the omalizumab Fab crystal structures without interfering with the ability of the omalizumab Fab to recognize IgE-Fc; this led to the successful crystallization and subsequent structure determination of the Fab/IgE-Fc complex. The mutagenesis strategy adopted to achieve this result is applicable to other intractable Fab/antigen complexes or systems in which Fabs are used as crystallization chaperones.


Subject(s)
Antibodies, Anti-Idiotypic/metabolism , Crystallization/methods , Immunoglobulin E/metabolism , Immunoglobulin Fab Fragments/metabolism , Immunoglobulin Fc Fragments/metabolism , Omalizumab/metabolism , Antibodies, Anti-Idiotypic/chemistry , Crystallography, X-Ray/methods , Humans , Immunoglobulin E/chemistry , Immunoglobulin Fab Fragments/chemistry , Immunoglobulin Fc Fragments/chemistry , Omalizumab/pharmacology , Protein Structure, Secondary , Protein Structure, Tertiary
9.
Front Immunol ; 11: 618327, 2020.
Article in English | MEDLINE | ID: mdl-33584711

ABSTRACT

Both non-immune "natural" and antigen-induced "immune" IgM are important for protection against infections and for regulation of immune responses to self-antigens. The roles of its Fc receptor (FcµR) in these IgM effector functions have begun to be explored. In the present study, by taking advantage of the difference in IgM-ligand binding of FcµRs of human (constitutive binding) and mouse (transient binding), we replaced non-conserved amino acid residues of human FcµR with mouse equivalents before establishment of cell lines stably expressing mutant or wild-type (WT) receptors. The resultant eight-different mutant FcµR-bearing cells were compared with WT receptor-bearing cells for cell-surface expression and IgM-binding by flow cytometric assessments using receptor-specific mAbs and IgM paraproteins as ligands. Three sites Asn66, Lys79-Arg83, and Asn109, which are likely in the CDR2, DE loop and CDR3 of the human FcµR Ig-like domain, respectively, were responsible for constitutive IgM binding. Intriguingly, substitution of Glu41 and Met42 in the presumed CDR1 with the corresponding mouse residues Gln and Leu, either single or more prominently in combination, enhanced both the receptor expression and IgM binding. A four-aa stretch of Lys24-Gly27 in the predicted A ß-strand of human FcµR appeared to be essential for maintenance of its proper receptor conformation on plasma membranes because of reduction of both receptor expression and IgM-binding potential when these were mutated. Results from a computational structural modeling analysis were consistent with these mutational data and identified a possible mode of binding of FcµR with IgM involving the loops including Asn66, Arg83 and Asn109 of FcµR interacting principally with the Cµ4 domain including Gln510 and to a lesser extent Cµ3 domain including Glu398, of human IgM. To our knowledge, this is the first experimental report describing the identification of amino acid residues of human FcµR critical for binding to IgM Fc.


Subject(s)
Amino Acids/chemistry , Binding Sites, Antibody , Models, Molecular , Receptors, Fc/chemistry , Animals , Complementarity Determining Regions/chemistry , Computer Simulation , Humans , Mice
10.
Biochim Biophys Acta Proteins Proteom ; 1868(1): 140266, 2020 01.
Article in English | MEDLINE | ID: mdl-31449905

ABSTRACT

FcµR is a high-affinity receptor for the Fc portion of human IgM. It participates in B cell activation, cell survival and proliferation, but the full range of its functions remains to be elucidated. The receptor has an extracellular immunoglobulin (Ig)-like domain homologous to those in Fcα/µR and pIgR, but unlike these two other IgM receptors which also bind IgA, FcµR exhibits a binding specificity for only IgM-Fc. Previous studies have suggested that the IgM/FcµR interaction mainly involves the Cµ4 domains with possible contributions from either Cµ3 or Cµ2. To define the binding site more precisely, we generated three recombinant IgM-Fc proteins with specific mutations in the Cµ3 and Cµ4 domains, as well as a construct lacking the Cµ2 domains, and analyzed their interaction with the extracellular Ig-like domain of FcµR using surface plasmon resonance analysis. There is a binding site for FcµR in each IgM heavy chain. Neither the absence of the Cµ2 domains nor the quadruple mutant D340S/Q341G/D342S/T343S (in Cµ3 adjacent to Cµ2) affected FcµR binding, whereas double mutant K361D/D416R (in Cµ3 at the Cµ4 interface) substantially decreased binding, and a single mutation Q510R (in Cµ4) completely abolished FcµR binding. We conclude that glutamine at position 510 in Cµ4 is critical for IgM binding to FcµR. This will facilitate discrimination between the distinct effects of FcµR interactions with soluble IgM and with the IgM BCR.


Subject(s)
Binding Sites , Immunoglobulin Fc Fragments/chemistry , Immunoglobulin M/chemistry , Receptors, Fc/chemistry , Humans
11.
Antibodies (Basel) ; 8(1)2019 Feb 22.
Article in English | MEDLINE | ID: mdl-31544825

ABSTRACT

Immunoglobulin E (IgE) antibodies are well known for their role in mediating allergic reactions, and their powerful effector functions activated through binding to Fc receptors FcεRI and FcεRII/CD23. Structural studies of IgE-Fc alone, and when bound to these receptors, surprisingly revealed not only an acutely bent Fc conformation, but also subtle allosteric communication between the two distant receptor-binding sites. The ability of IgE-Fc to undergo more extreme conformational changes emerged from structures of complexes with anti-IgE antibodies, including omalizumab, in clinical use for allergic disease; flexibility is clearly critical for IgE function, but may also be exploited by allosteric interference to inhibit IgE activity for therapeutic benefit. In contrast, the power of IgE may be harnessed to target cancer. Efforts to improve the effector functions of therapeutic antibodies for cancer have almost exclusively focussed on IgG1 and IgG4 subclasses, but IgE offers an extremely high affinity for FcεRI receptors on immune effector cells known to infiltrate solid tumours. Furthermore, while tumour-resident inhibitory Fc receptors can modulate the effector functions of IgG antibodies, no inhibitory IgE Fc receptors are known to exist. The development of tumour antigen-specific IgE antibodies may therefore provide an improved immune functional profile and enhanced anti-cancer efficacy. We describe proof-of-concept studies of IgE immunotherapies against solid tumours, including a range of in vitro and in vivo evaluations of efficacy and mechanisms of action, as well as ex vivo and in vivo safety studies. The first anti-cancer IgE antibody, MOv18, the clinical translation of which we discuss herein, has now reached clinical testing, offering great potential to direct this novel therapeutic modality against many other tumour-specific antigens. This review highlights how our understanding of IgE structure and function underpins these exciting clinical developments.

12.
J Immunol ; 203(7): 1693-1700, 2019 10 01.
Article in English | MEDLINE | ID: mdl-31462504

ABSTRACT

An allergic reaction is rapidly generated when allergens bind and cross-link IgE bound to its receptor FcεRI on effector cells, resulting in cell degranulation and release of proinflammatory mediators. The extent of effector cell activation is linked to allergen affinity, oligomeric state, valency, and spacing of IgE-binding epitopes on the allergen. Whereas most of these observations come from studies using synthetic allergens, in this study we have used Timothy grass pollen allergen Phl p 7 and birch pollen allergen Bet v 4 to study these effects. Despite the high homology of these polcalcin family allergens, Phl p 7 and Bet v 4 display different binding characteristics toward two human patient-derived polcalcin-specific IgE Abs. We have used native polcalcin dimers and engineered multimeric allergens to test the effects of affinity and oligomeric state on IgE binding and effector cell activation. Our results indicate that polcalcin multimers are required to stimulate high levels of effector cell degranulation when using the humanized RBL-SX38 cell model and that multivalency can overcome the need for high-affinity interactions.


Subject(s)
Allergens/immunology , Antibody Affinity , Antigens, Plant/immunology , Calcium-Binding Proteins/immunology , Cell Degranulation , Immunoglobulin E/immunology , Models, Immunological , Plant Proteins/immunology , Allergens/genetics , Antigens, Plant/genetics , Calcium-Binding Proteins/genetics , Epitopes/genetics , Epitopes/immunology , HEK293 Cells , Humans , Plant Proteins/genetics , Protein Multimerization/genetics , Protein Multimerization/immunology
13.
IEEE Trans Neural Netw Learn Syst ; 30(6): 1920-1926, 2019 Jun.
Article in English | MEDLINE | ID: mdl-30387748

ABSTRACT

Probabilistic spin logic is a recently proposed computing paradigm based on unstable stochastic units called probabilistic bits ( p -bits) that can be correlated to form probabilistic circuits (p-circuits). These p-circuits can be used to solve the problems of optimization, inference, and implement precise Boolean functions in an "inverted" mode, where a given Boolean circuit can operate in reverse to find the input combinations that are consistent with a given output. In this brief, we present a scalable field-programmable gate array implementation of such invertible p-circuits. We implement a "weighted" p -bit that combines stochastic units with localized memory structures. We also present a generalized tile of weighted p -bits to which a large class of problems beyond invertible Boolean logic can be mapped and how invertibility can be applied to interesting problems such as the NP-complete subset sum problem by solving a small instance of this problem in hardware.

14.
Angew Chem Int Ed Engl ; 57(52): 17194-17199, 2018 12 21.
Article in English | MEDLINE | ID: mdl-30408305

ABSTRACT

Immunoglobulins are biomolecules involved in defence against foreign substances. Flexibility is key to their functional properties in relation to antigen binding and receptor interactions. We have developed an integrative strategy combining ion mobility mass spectrometry (IM-MS) with molecular modelling to study the conformational dynamics of human IgG antibodies. Predictive models of all four human IgG subclasses were assembled and their dynamics sampled in the transition from extended to collapsed state during IM-MS. Our data imply that this collapse of IgG antibodies is related to their intrinsic structural features, including Fab arm flexibility, collapse towards the Fc region, and the length of their hinge regions. The workflow presented here provides an accurate structural representation in good agreement with the observed collision cross section for these flexible IgG molecules. These results have implications for studying other nonglobular flexible proteins.


Subject(s)
Immunoglobulin G/chemistry , Gases/chemistry , Mass Spectrometry , Models, Molecular , Protein Conformation
15.
FEBS J ; 285(24): 4575-4589, 2018 12.
Article in English | MEDLINE | ID: mdl-30387270

ABSTRACT

The identification of new strategies to fight bacterial infections in view of the spread of multiple resistance to antibiotics has become mandatory. It has been demonstrated that several bacteria develop poly-γ-glutamic acid (γ-PGA) capsules as a protection from external insults and/or host defence systems. Among the pathogens that shield themselves in these capsules are Bacillus anthracis, Francisella tularensis and several Staphylococcus strains. These are important pathogens with a profound influence on human health. The recently characterised γ-PGA hydrolases, which can dismantle the γ-PGA-capsules, are an attractive new direction that can offer real hope for the development of alternatives to antibiotics, particularly in cases of multidrug resistant bacteria. We have characterised in detail the cleaving mechanism and stereospecificity of the enzyme PghL (previously named YndL) from Bacillus subtilis encoded by a gene of phagic origin and dramatically efficient in degrading the long polymeric chains of γ-PGA. We used X-ray crystallography to solve the three-dimensional structures of the enzyme in its zinc-free, zinc-bound and complexed forms. The protein crystallised with a γ-PGA hexapeptide substrate and thus reveals details of the interaction which could explain the stereospecificity observed and give hints on the catalytic mechanism of this class of hydrolytic enzymes.


Subject(s)
Bacillus subtilis/enzymology , Bacterial Proteins/chemistry , Bacterial Proteins/metabolism , Hydrolases/chemistry , Hydrolases/metabolism , Polyglutamic Acid/analogs & derivatives , Amino Acid Sequence , Crystallography, X-Ray , Humans , Models, Molecular , Polyglutamic Acid/metabolism , Protein Conformation , Sequence Homology
16.
Proc Natl Acad Sci U S A ; 115(37): E8707-E8716, 2018 09 11.
Article in English | MEDLINE | ID: mdl-30150373

ABSTRACT

Antibodies classically bind antigens via their complementarity-determining regions, but an alternative mode of interaction involving V-domain framework regions has been observed for some B cell "superantigens." We report the crystal structure of an antibody employing both modes of interaction simultaneously and binding two antigen molecules. This human antibody from an allergic individual binds to the grass pollen allergen Phl p 7. Not only are two allergen molecules bound to each antibody fragment (Fab) but also each allergen molecule is bound by two Fabs: One epitope is recognized classically, the other in a superantigen-like manner. A single allergen molecule thus cross-links two identical Fabs, contrary to the one-antibody-one-epitope dogma, which dictates that a dimeric allergen at least is required for this to occur. Allergens trigger immediate hypersensitivity reactions by cross-linking receptor-bound IgE molecules on effector cells. We found that monomeric Phl p 7 induced degranulation of basophils sensitized solely with this monoclonal antibody expressed as an IgE, demonstrating that the dual specificity has functional consequences. The monomeric state of Phl p 7 and two structurally related allergens was confirmed by size-exclusion chromatography and multiangle laser light scattering, and the results were supported by degranulation studies with the related allergens, a second patient-derived allergen-specific antibody lacking the nonclassical binding site, and mutagenesis of the nonclassically recognized allergen epitope. The antibody dual reactivity and cross-linking mechanism not only have implications for understanding allergenicity and allergen potency but, importantly, also have broader relevance to antigen recognition by membrane Ig and cross-linking of the B cell receptor.


Subject(s)
Antibodies, Monoclonal/immunology , Antigens, Plant/immunology , Calcium-Binding Proteins/immunology , Epitopes/immunology , Superantigens/immunology , Antibodies, Monoclonal/chemistry , Antibodies, Monoclonal/metabolism , Antibody Specificity/immunology , Antigens, Plant/chemistry , Antigens, Plant/metabolism , Basophils/immunology , Basophils/physiology , Calcium-Binding Proteins/chemistry , Calcium-Binding Proteins/metabolism , Cell Degranulation/immunology , Cross Reactions/immunology , Crystallography, X-Ray , Epitopes/chemistry , Epitopes/metabolism , Humans , Immunoglobulin E/chemistry , Immunoglobulin E/immunology , Immunoglobulin E/metabolism , Models, Molecular , Protein Binding , Protein Conformation , Superantigens/chemistry , Superantigens/metabolism
17.
Sci Rep ; 8(1): 11548, 2018 08 01.
Article in English | MEDLINE | ID: mdl-30069035

ABSTRACT

Immunoglobulin E (IgE) antibodies play a central role in the allergic response: interaction with FcεRI on mast cells and basophils leads to immediate hypersensitivity reactions upon allergen challenge, while interaction with CD23/FcεRII, expressed on a variety of cells, regulates IgE synthesis among other activities. The receptor-binding IgE-Fc region has recently been found to display remarkable flexibility, from acutely bent to extended conformations, with allosteric communication between the distant FcεRI and CD23 binding sites. We report the structure of an anti-IgE antibody Fab (8D6) bound to IgE-Fc through a mixed protein-carbohydrate epitope, revealing further flexibility and a novel extended conformation with potential relevance to that of membrane-bound IgE in the B cell receptor for antigen. Unlike the earlier, clinically approved anti-IgE antibody omalizumab, 8D6 inhibits binding to FcεRI but not CD23; the structure reveals how this discrimination is achieved through both orthosteric and allosteric mechanisms, supporting therapeutic strategies that retain the benefits of CD23 binding.


Subject(s)
Antibodies, Anti-Idiotypic/chemistry , Antibodies, Anti-Idiotypic/metabolism , Immunoglobulin E/chemistry , Immunoglobulin E/metabolism , Receptors, IgE/metabolism , B-Lymphocytes/immunology , Crystallography, X-Ray , HEK293 Cells , Humans , Immunoglobulin Fab Fragments/chemistry , Immunoglobulin Fab Fragments/metabolism , Immunoglobulin Fc Fragments/chemistry , Immunoglobulin Fc Fragments/metabolism , Mast Cells/immunology , Protein Binding , Protein Conformation
19.
Mol Immunol ; 93: 216-222, 2018 01.
Article in English | MEDLINE | ID: mdl-29216544

ABSTRACT

In allergic disease, mast cell activation is conventionally triggered by allergen-mediated cross-linking of receptor-bound IgE on the cell surface. In addition to its diverse range of intracellular roles in apoptosis, cell proliferation and cancer, Histamine-Releasing Factor (HRF) also activates mast cells and basophils. A subset of IgE antibodies bind HRF through their Fab regions, and two IgE binding sites on HRF have been mapped. HRF can form dimers, and a disulphide-linked dimer is critical for activity. The current model for the activity of HRF in mast cell activation involves cross-linking of receptor-bound IgE by dimeric HRF, mediated by HRF/Fab interactions. HRF crystal and solution structures have provided little insight into either the formation of disulphide-linked HRF dimers or the ability of HRF to activate mast cells. We report the first crystal structure of murine HRF (mHRF) to 4.0Å resolution, revealing a conserved fold. We also solved the structure of human HRF (hHRF) in two new crystal forms, one at the highest resolution (1.4Å) yet reported. The high resolution hHRF structure reveals a disulphide-linked dimer, in which the two molecules are closely associated, and provides a model for the role of both human and murine HRF in mast cell activation.


Subject(s)
Biomarkers, Tumor/chemistry , Mast Cells/physiology , Animals , Biomarkers, Tumor/physiology , Crystallography, X-Ray , Cysteine/chemistry , Cystine/chemistry , Dimerization , Humans , Hypersensitivity/immunology , Immunoglobulin E/immunology , Immunoglobulin Fab Fragments/immunology , Mice , Models, Immunological , Models, Molecular , Protein Conformation , Protein Folding , Receptors, IgE/immunology , Tumor Protein, Translationally-Controlled 1
20.
Biochim Biophys Acta Proteins Proteom ; 1865(11 Pt A): 1336-1347, 2017 Nov.
Article in English | MEDLINE | ID: mdl-28844738

ABSTRACT

Immunoglobulin E (IgE) is the antibody that plays a central role in the mechanisms of allergic diseases such as asthma. Interactions with its receptors, FcεRI on mast cells and CD23 on B cells, are mediated by the Fc region, a dimer of the Cε2, Cε3 and Cε4 domains. A sub-fragment lacking the Cε2 domains, Fcε3-4, also binds to both receptors, although receptor binding almost exclusively involves the Cε3 domains. This domain also contains the N-linked glycosylation site conserved in other isotypes. We report here the crystal structures of IgE-Fc and Fcε3-4 at the highest resolutions yet determined, 1.75Šand 2.0Šrespectively, revealing unprecedented detail regarding the carbohydrate and its interactions with protein domains. Analysis of the crystallographic B-factors of these, together with all earlier IgE-Fc and Fcε3-4 structures, shows that the Cε3 domains exhibit the greatest intrinsic flexibility and quaternary structural variation within IgE-Fc. Intriguingly, both well-ordered carbohydrate and disordered polypeptide can be seen within the same Cε3 domain. A simplified method for comparing the quaternary structures of the Cε3 domains in free and receptor-bound IgE-Fc structures is presented, which clearly delineates the FcεRI and CD23 bound states. Importantly, differential scanning fluorimetric analysis of IgE-Fc and Fcε3-4 identifies Cε3 as the domain most susceptible to thermally-induced unfolding, and responsible for the characteristically low melting temperature of IgE.


Subject(s)
Immunoglobulin E/chemistry , Immunoglobulin Fc Fragments/chemistry , Receptors, IgE/chemistry , Amino Acid Motifs , Binding Sites , Carbohydrate Sequence , Crystallography, X-Ray , Gene Expression , Glycosylation , Humans , Immunoglobulin E/genetics , Immunoglobulin E/immunology , Immunoglobulin Fc Fragments/genetics , Immunoglobulin Fc Fragments/immunology , Models, Molecular , Phase Transition , Protein Binding , Protein Interaction Domains and Motifs , Protein Multimerization , Protein Stability , Protein Structure, Quaternary , Protein Structure, Secondary , Protein Unfolding , Receptors, IgE/genetics , Receptors, IgE/immunology , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Recombinant Proteins/immunology , Temperature
SELECTION OF CITATIONS
SEARCH DETAIL
...