Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 44
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.
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
3.
Nat Commun ; 14(1): 2192, 2023 04 25.
Article in English | MEDLINE | ID: mdl-37185332

ABSTRACT

Outcomes for half of patients with melanoma remain poor despite standard-of-care checkpoint inhibitor therapies. The prevalence of the melanoma-associated antigen chondroitin sulfate proteoglycan 4 (CSPG4) expression is ~70%, therefore effective immunotherapies directed at CSPG4 could benefit many patients. Since IgE exerts potent immune-activating functions in tissues, we engineer a monoclonal IgE antibody with human constant domains recognizing CSPG4 to target melanoma. CSPG4 IgE binds to human melanomas including metastases, mediates tumoricidal antibody-dependent cellular cytotoxicity and stimulates human IgE Fc-receptor-expressing monocytes towards pro-inflammatory phenotypes. IgE demonstrates anti-tumor activity in human melanoma xenograft models engrafted with human effector cells and is associated with enhanced macrophage infiltration, enriched monocyte and macrophage gene signatures and pro-inflammatory signaling pathways in the tumor microenvironment. IgE prolongs the survival of patient-derived xenograft-bearing mice reconstituted with autologous immune cells. No ex vivo activation of basophils in patient blood is measured in the presence of CSPG4 IgE. Our findings support a promising IgE-based immunotherapy for melanoma.


Subject(s)
Melanoma , Proteoglycans , Humans , Mice , Animals , Proteoglycans/metabolism , Antigens , Chondroitin Sulfate Proteoglycans , Melanoma/metabolism , Antibodies, Monoclonal/pharmacology , Immunoglobulin E , Tumor Microenvironment
4.
Structure ; 31(6): 677-688.e4, 2023 06 01.
Article in English | MEDLINE | ID: mdl-37015227

ABSTRACT

Carboxysomes are proteinaceous bacterial microcompartments that sequester the key enzymes for carbon fixation in cyanobacteria and some proteobacteria. They consist of a virus-like icosahedral shell, encapsulating several enzymes, including ribulose 1,5-bisphosphate carboxylase/oxygenase (RuBisCO), responsible for the first step of the Calvin-Benson-Bassham cycle. Despite their significance in carbon fixation and great bioengineering potentials, the structural understanding of native carboxysomes is currently limited to low-resolution studies. Here, we report the characterization of a native α-carboxysome from a marine cyanobacterium by single-particle cryoelectron microscopy (cryo-EM). We have determined the structure of its RuBisCO enzyme, and obtained low-resolution maps of its icosahedral shell, and of its concentric interior organization. Using integrative modeling approaches, we have proposed a complete atomic model of an intact carboxysome, providing insight into its organization and assembly. This is critical for a better understanding of the carbon fixation mechanism and toward repurposing carboxysomes in synthetic biology for biotechnological applications.


Subject(s)
Cyanobacteria , Ribulose-Bisphosphate Carboxylase , Cryoelectron Microscopy , Ribulose-Bisphosphate Carboxylase/chemistry , Ribulose-Bisphosphate Carboxylase/metabolism , Organelles/metabolism , Photosynthesis , Bacterial Proteins/metabolism
5.
PLoS One ; 18(3): e0282831, 2023.
Article in English | MEDLINE | ID: mdl-36913398

ABSTRACT

Imiquimod, a Toll-like receptor 7 (TLR7) agonist is routinely used for topical administration in basal cell carcinoma and stage zero melanoma. Similarly, the TLR agonist Bacillus Calmette-Guérin is used for the local treatment of bladder cancer and clinical trials showed treatment efficacy of intratumoral injections with TLR9 agonists. However, when administered systemically, endosomal TLR agonists cause adverse responses due to broad immune activation. Hence, strategies for targeted delivery of TLR agonists to the tumor tissue are needed to enable the widespread use of endosomal TLR agonists in the context of tumor immunotherapy. One strategy for targeted delivery of TLR agonist is their conjugation to tumor antigen-specific therapeutic antibodies. Such antibody-TLR agonist conjugates act synergistically by inducing local TLR-mediated innate immune activation which complements the anti-tumor immune mechanisms induced by the therapeutic antibody. In this study, we explored different conjugation strategies for TLR9 agonists to immunoglobulin G (IgG). We evaluated biochemical conjugation of immunostimulatory CpG oligodesoxyribonucleotides (ODN) to the HER2-specific therapeutic antibody Trastuzumab with different cross-linkers comparing stochastic with site-specific conjugation. The physiochemical make-up and biological activities of the generated Trastuzumab-ODN conjugates were characterized in vitro and demonstrated that site-specific conjugation of CpG ODN is crucial for maintaining the antigen-binding capabilities of Trastuzumab. Furthermore, site-specific conjugate was effective in promoting anti-tumor immune responses in vivo in a pseudo-metastasis mouse model with engineered human HER2-transgenic tumor cells. In this in vivo model, co-delivery of Trastuzumab and CpG ODN in form of site-specific conjugates was superior to co-injection of unconjugated Trastuzumab, CpG ODN or stochastic conjugate in promoting T cell activation and expansion. Thereby, this study highlights that site-specific conjugation of CpG ODN to therapeutic antibodies targeting tumor markers is a feasible and more reliable approach for generation of conjugates which retain and combine the functional properties of the adjuvant and the antibody.


Subject(s)
Immunoconjugates , Neoplasms , Animals , Mice , Humans , Toll-Like Receptor 9/agonists , Adjuvants, Immunologic/pharmacology , Antigens , Immunoconjugates/chemistry , Neoplasms/drug therapy , Immunoglobulin G , Trastuzumab , Oligodeoxyribonucleotides , Toll-Like Receptor 7/agonists
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.
J Biol Chem ; 296: 100513, 2021.
Article in English | MEDLINE | ID: mdl-33676896

ABSTRACT

The C1q and TNF related 4 (C1QTNF4) protein is a structurally unique member of the C1QTNF family, a family of secreted proteins that have structural homology with both complement C1q and the tumor necrosis factor superfamily. C1QTNF4 has been linked to the autoimmune disease systemic lupus erythematosus through genetic studies; however, its role in immunity and inflammation remains poorly defined and a cell surface receptor of C1QTNF4 has yet to be identified. Here we report identification of nucleolin as a cell surface receptor of C1QTNF4 using mass spectrometric analysis. Additionally, we present evidence that the interaction between C1QTNF4 and nucleolin is mediated by the second C1q-like domain of C1QTNF4 and the C terminus of nucleolin. We show that monocytes and B cells are target cells of C1QTNF4 and observe extensive binding to dead cells. Imaging flow cytometry experiments in monocytes show that C1QTNF4 becomes actively internalized upon cell binding. Our results suggest that nucleolin may serve as a docking molecule for C1QTNF4 and act in a context-dependent manner through coreceptors. Taken together, these findings further our understanding of C1QTNF4's function in the healthy immune system and how dysfunction may contribute to the development of systemic lupus erythematosus.


Subject(s)
B-Lymphocytes/immunology , Cytokines/metabolism , Immunity, Innate/immunology , Inflammation/immunology , Monocytes/immunology , Phosphoproteins/metabolism , RNA-Binding Proteins/metabolism , Receptors, Cell Surface/metabolism , Amino Acid Sequence , B-Lymphocytes/cytology , B-Lymphocytes/metabolism , Cytokines/genetics , Humans , Inflammation/metabolism , Inflammation/pathology , Monocytes/cytology , Monocytes/metabolism , Phosphoproteins/genetics , RNA-Binding Proteins/genetics , Receptors, Cell Surface/genetics , Nucleolin
8.
Biophys J ; 120(2): 254-269, 2021 01 19.
Article in English | MEDLINE | ID: mdl-33345902

ABSTRACT

Förster resonance energy transfer (FRET) is a powerful tool to investigate the interaction between proteins in living cells. Fluorescence proteins, such as the green fluorescent protein (GFP) and its derivatives, are coexpressed in cells linked to proteins of interest. Time-resolved fluorescence anisotropy is a popular tool to study homo-FRET of fluorescent proteins as an indicator of dimerization, in which its signature consists of a very short component at the beginning of the anisotropy decay. In this work, we present an approach to study GFP homo-FRET via a combination of time-resolved fluorescence anisotropy, the stretched exponential decay model, and molecular dynamics simulations. We characterize a new, to our knowledge, FRET standard formed by two enhanced GFPs (eGFPs) and a flexible linker of 15 aminoacids (eGFP15eGFP) with this protocol, which is validated by using an eGFP monomer as a reference. An excellent agreement is found between the FRET efficiency calculated from the fit of the eGFP15eGFP fluorescence anisotropy decays with a stretched exponential decay model (〈EFRETexp〉 = 0.25 ± 0.05) and those calculated from the molecular dynamics simulations (〈EFRETMD〉 = 0.18 ± 0.14). The relative dipole orientation between the GFPs is best described by the orientation factors 〈κ2〉 = 0.17 ± 0.16 and 〈|κ|〉 = 0.35 ± 0.20, contextualized within a static framework in which the linker hinders the free rotation of the fluorophores and excludes certain configurations. The combination of time- and polarization-resolved fluorescence spectroscopy with molecular dynamics simulations is shown to be a powerful tool for the study and interpretation of homo-FRET.


Subject(s)
Fluorescence Resonance Energy Transfer , Molecular Dynamics Simulation , Fluorescence Polarization , Green Fluorescent Proteins/genetics , Microscopy, Fluorescence
9.
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
10.
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
11.
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
12.
Proc Natl Acad Sci U S A ; 114(52): 13738-13743, 2017 12 26.
Article in English | MEDLINE | ID: mdl-29229862

ABSTRACT

The microtubule motor kinesin-1 interacts via its cargo-binding domain with both microtubules and organelles, and hence plays an important role in controlling organelle transport and microtubule dynamics. In the absence of cargo, kinesin-1 is found in an autoinhibited conformation. The molecular basis of how cargo engagement affects the balance between kinesin-1's active and inactive conformations and roles in microtubule dynamics and organelle transport is not well understood. Here we describe the discovery of kinesore, a small molecule that in vitro inhibits kinesin-1 interactions with short linear peptide motifs found in organelle-specific cargo adaptors, yet activates kinesin-1's function of controlling microtubule dynamics in cells, demonstrating that these functions are mechanistically coupled. We establish a proof-of-concept that a microtubule motor-cargo interface and associated autoregulatory mechanism can be manipulated using a small molecule, and define a target for the modulation of microtubule dynamics.


Subject(s)
Enzyme Activators , Kinesins , Microtubules , Amino Acid Motifs , Enzyme Activators/chemistry , Enzyme Activators/pharmacology , HeLa Cells , Humans , Kinesins/chemistry , Kinesins/genetics , Kinesins/metabolism , Microtubules/chemistry , Microtubules/genetics , Microtubules/metabolism
13.
Front Immunol ; 8: 1112, 2017.
Article in English | MEDLINE | ID: mdl-28959256

ABSTRACT

Monoclonal antibodies find broad application as therapy for various types of cancer by employing multiple mechanisms of action against tumors. Manipulating the Fc-mediated functions of antibodies that engage immune effector cells, such as NK cells, represents a strategy to influence effector cell activation and to enhance antibody potency and potentially efficacy. We developed a novel approach to generate and ascertain the functional attributes of Fc mutant monoclonal antibodies. This entailed coupling single expression vector (pVitro1) antibody cloning, using polymerase incomplete primer extension (PIPE) polymerase chain reaction, together with simultaneous Fc region point mutagenesis and high yield transient expression in human mammalian cells. Employing this, we engineered wild type, low (N297Q, NQ), and high (S239D/I332E, DE) FcR-binding Fc mutant monoclonal antibody panels recognizing two cancer antigens, HER2/neu and chondroitin sulfate proteoglycan 4. Antibodies were generated with universal mutagenic primers applicable to any IgG1 pVitro1 constructs, with high mutagenesis and transfection efficiency, in small culture volumes, at high yields and within 12 days from design to purified material. Antibody variants conserved their Fab-mediated recognition of target antigens and their direct anti-proliferative effects against cancer cells. Fc mutations had a significant impact on antibody interactions with Fc receptors (FcRs) on human NK cells, and consequently on the potency of NK cell activation, quantified by immune complex-mediated calcium mobilization and by antibody-dependent cellular cytotoxicity (ADCC) of tumor cells. This strategy for manipulation and testing of Fc region engagement with cognate FcRs can facilitate the design of antibodies with defined effector functions and potentially enhanced efficacy against tumor cells.

14.
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
15.
Sci Rep ; 7(1): 8164, 2017 08 15.
Article in English | MEDLINE | ID: mdl-28811536

ABSTRACT

Degranulation of mast cells and basophils, with release of agents of the allergic response, ensues when multivalent antigens bind to and cross-link the cells' receptor-bound IgE antibodies. A widely used commercial monoclonal IgE antibody, SPE-7 IgE from Sigma, was found to possess the radically anomalous property, termed "cytokinergic", of inducing basophil degranulation without the intervention of an antigen. We show here that the IgE monomer, freed of protein contaminants, is devoid of this activity, and that the source of the anomaly is a trace impurity, identified as a dissociation-resistant IgE trimer. Possible models for the formation of IgE trimers and the manner in which they cross-link cell surface receptors are suggested herein.


Subject(s)
Antibodies, Monoclonal/chemistry , Antibodies, Monoclonal/immunology , Basophils/immunology , Cell Degranulation/immunology , Immunoglobulin E/chemistry , Immunoglobulin E/immunology , Protein Multimerization , Animals , Antibodies, Monoclonal/isolation & purification , Antibodies, Monoclonal/metabolism , Basophils/metabolism , Cell Line , Humans , Immunoglobulin E/isolation & purification , Immunoglobulin E/metabolism , Mice , Protein Binding , Receptors, IgE/metabolism
16.
J Biol Chem ; 292(24): 9975-9987, 2017 06 16.
Article in English | MEDLINE | ID: mdl-28438838

ABSTRACT

Immunoglobulin E and its interactions with receptors FcϵRI and CD23 play a central role in allergic disease. Omalizumab, a clinically approved therapeutic antibody, inhibits the interaction between IgE and FcϵRI, preventing mast cell and basophil activation, and blocks IgE binding to CD23 on B cells and antigen-presenting cells. We solved the crystal structure of the complex between an omalizumab-derived Fab and IgE-Fc, with one Fab bound to each Cϵ3 domain. Free IgE-Fc adopts an acutely bent structure, but in the complex it is only partially bent, with large-scale conformational changes in the Cϵ3 domains that inhibit the interaction with FcϵRI. CD23 binding is inhibited sterically due to overlapping binding sites on each Cϵ3 domain. Studies of omalizumab Fab binding in solution demonstrate the allosteric basis for FcϵRI inhibition and, together with the structure, reveal how omalizumab may accelerate dissociation of receptor-bound IgE from FcϵRI, exploiting the intrinsic flexibility and allosteric potential of IgE.


Subject(s)
Anti-Asthmatic Agents/pharmacology , Immunoglobulin E/metabolism , Models, Molecular , Omalizumab/pharmacology , Receptors, IgE/antagonists & inhibitors , Allosteric Site , Amino Acid Substitution , Crystallography, X-Ray , Fluorescence Resonance Energy Transfer , Humans , Immunoglobulin E/chemistry , Immunoglobulin E/genetics , Immunoglobulin Fab Fragments/chemistry , Immunoglobulin Fab Fragments/genetics , Immunoglobulin Fab Fragments/metabolism , Immunoglobulin Fab Fragments/pharmacology , Immunoglobulin Fc Fragments/chemistry , Immunoglobulin Fc Fragments/genetics , Immunoglobulin Fc Fragments/metabolism , Immunoglobulin Fc Fragments/pharmacology , Omalizumab/chemistry , Omalizumab/genetics , Omalizumab/metabolism , Pliability , Point Mutation , Protein Conformation , Protein Interaction Domains and Motifs , Protein Refolding , Receptors, IgE/chemistry , Receptors, IgE/metabolism , Recombinant Fusion Proteins/chemistry , Recombinant Fusion Proteins/metabolism , Recombinant Fusion Proteins/pharmacology , Recombinant Proteins/chemistry , Recombinant Proteins/metabolism , Recombinant Proteins/pharmacology , Solubility , Surface Plasmon Resonance
17.
Sci Rep ; 7: 45533, 2017 03 31.
Article in English | MEDLINE | ID: mdl-28361904

ABSTRACT

The antibody IgE plays a central role in allergic disease mechanisms. Its effector functions are controlled through interactions between the Fc region and two principal cell surface receptors FcεRI and CD23. The interaction with FcεRI is primarily responsible for allergic sensitization and the inflammatory response, while IgE binding to CD23 is involved in the regulation of IgE synthesis and allergen transcytosis. Here we present the crystal structure of a CD23/IgE-Fc complex and conduct isothermal titration calorimetric binding studies. Two lectin-like "head" domains of CD23 bind to IgE-Fc with affinities that differ by more than an order of magnitude, but the crystal structure reveals only one head bound to one of the two identical heavy-chains in the asymmetrically bent IgE-Fc. These results highlight the subtle interplay between receptor binding sites in IgE-Fc and their affinities, the understanding of which may be exploited for therapeutic intervention in allergic disease.


Subject(s)
B-Lymphocytes/immunology , Binding Sites/immunology , Immunoglobulin E/immunology , Receptors, IgE/immunology , Allergens/immunology , Humans , Immunoglobulin Fc Fragments/immunology , Protein Binding/immunology , Transcytosis/immunology
18.
Cancer Res ; 77(5): 1127-1141, 2017 03 01.
Article in English | MEDLINE | ID: mdl-28096174

ABSTRACT

IgE antibodies are key mediators of antiparasitic immune responses, but their potential for cancer treatment via antibody-dependent cell-mediated cytotoxicity (ADCC) has been little studied. Recently, tumor antigen-specific IgEs were reported to restrict cancer cell growth by engaging high-affinity Fc receptors on monocytes and macrophages; however, the underlying therapeutic mechanisms were undefined and in vivo proof of concept was limited. Here, an immunocompetent rat model was designed to recapitulate the human IgE-Fcε receptor system for cancer studies. We also generated rat IgE and IgG mAbs specific for the folate receptor (FRα), which is expressed widely on human ovarian tumors, along with a syngeneic rat tumor model expressing human FRα. Compared with IgG, anti-FRα IgE reduced lung metastases. This effect was associated with increased intratumoral infiltration by TNFα+ and CD80+ macrophages plus elevated TNFα and the macrophage chemoattractant MCP-1 in lung bronchoalveolar lavage fluid. Increased levels of TNFα and MCP-1 correlated with IgE-mediated tumor cytotoxicity by human monocytes and with longer patient survival in clinical specimens of ovarian cancer. Monocytes responded to IgE but not IgG exposure by upregulating TNFα, which in turn induced MCP-1 production by monocytes and tumor cells to promote a monocyte chemotactic response. Conversely, blocking TNFα receptor signaling abrogated induction of MCP-1, implicating it in the antitumor effects of IgE. Overall, these findings show how antitumor IgE reprograms monocytes and macrophages in the tumor microenvironment, encouraging the clinical use of IgE antibody technology to attack cancer beyond the present exclusive reliance on IgG. Cancer Res; 77(5); 1127-41. ©2017 AACR.


Subject(s)
Antibodies, Anti-Idiotypic/immunology , Folate Receptor 1/immunology , Macrophages/immunology , Ovarian Neoplasms/immunology , Tumor Necrosis Factor-alpha/immunology , Animals , Cell Line, Tumor , Female , Folate Receptor 1/antagonists & inhibitors , Humans , Ovarian Neoplasms/drug therapy , Rats , Rats, Wistar , Signal Transduction , Tumor Necrosis Factor-alpha/biosynthesis
19.
Sci Rep ; 4: 5885, 2014 Jul 30.
Article in English | MEDLINE | ID: mdl-25073855

ABSTRACT

Over the last four decades, molecular cloning has evolved tremendously. Efficient products allowing assembly of multiple DNA fragments have become available. However, cost-effective tools for engineering antibodies of different specificities, isotypes and species are still needed for many research and clinical applications in academia. Here, we report a method for one-step assembly of antibody heavy- and light-chain DNAs into a single mammalian expression vector, starting from DNAs encoding the desired variable and constant regions, which allows antibodies of different isotypes and specificity to be rapidly generated. As a proof of principle we have cloned, expressed and characterized functional recombinant tumor-associated antigen-specific chimeric IgE/κ and IgG1/κ, as well as recombinant grass pollen allergen Phl p 7 specific fully human IgE/λ and IgG4/λ antibodies. This method utilizing the antibody expression vectors, available at Addgene, has many applications, including the potential to support simultaneous processing of antibody panels, to facilitate mechanistic studies of antigen-antibody interactions and to conduct early evaluations of antibody functions.


Subject(s)
Cloning, Molecular/methods , Genetic Vectors/metabolism , Immunoglobulin E/biosynthesis , Reagent Kits, Diagnostic , Recombinant Fusion Proteins/biosynthesis , Animals , Antigens, Plant/biosynthesis , Antigens, Plant/genetics , Base Sequence , Calcium-Binding Proteins/biosynthesis , Calcium-Binding Proteins/genetics , DNA Primers/chemical synthesis , Epitopes/chemistry , Epitopes/immunology , Escherichia coli/genetics , Escherichia coli/metabolism , Gene Expression , Genetic Vectors/chemistry , Humans , Immunoglobulin E/genetics , Immunoglobulin G/biosynthesis , Immunoglobulin G/genetics , Immunoglobulin kappa-Chains/biosynthesis , Immunoglobulin kappa-Chains/genetics , Immunoglobulin lambda-Chains/biosynthesis , Immunoglobulin lambda-Chains/genetics , Molecular Sequence Data , Plant Proteins/biosynthesis , Plant Proteins/genetics , Polymerase Chain Reaction , Recombinant Fusion Proteins/genetics
20.
Acta Crystallogr F Struct Biol Commun ; 70(Pt 3): 305-9, 2014 Mar.
Article in English | MEDLINE | ID: mdl-24598915

ABSTRACT

The antibody IgE plays a central role in allergic disease, functioning principally through two cell-surface receptors: FcℇRI and CD23. FcℇRI on mast cells and basophils mediates the immediate hypersensitivity response, whilst the interaction of IgE with CD23 on B cells regulates IgE production. Crystal structures of the lectin-like `head' domain of CD23 alone and bound to a subfragment of IgE consisting of the dimer of Cℇ3 and Cℇ4 domains (Fcℇ3-4) have recently been determined, revealing flexibility in the IgE-binding site of CD23. Here, a new crystal form of the CD23-Fcℇ3-4 complex with different molecular-packing constraints is reported, which together with the earlier results demonstrates that conformational variability at the interface extends additionally to the IgE Fc and the quaternary structure of its domains.


Subject(s)
Immunoglobulin E/chemistry , Immunoglobulin Fc Fragments/chemistry , Receptors, IgE/chemistry , Crystallography, X-Ray , Humans , Hydrogen Bonding , Models, Molecular , Protein Binding , Protein Interaction Domains and Motifs , Protein Structure, Quaternary , Protein Structure, Secondary
SELECTION OF CITATIONS
SEARCH DETAIL
...