Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 23
Filtrar
1.
J Immunol ; 211(3): 403-413, 2023 08 01.
Artigo em Inglês | MEDLINE | ID: mdl-37350633

RESUMO

Activation of the complement system represents an important effector mechanism of endogenous and therapeutic Abs. However, efficient complement activation is restricted to a subset of Abs due to the requirement of multivalent interactions between the Ab Fc regions and the C1 complex. In the present study, we demonstrate that Fc-independent recruitment of C1 by modular bispecific single-domain Abs that simultaneously bind C1q and a surface Ag can potently activate the complement system. Using Ags from hematological and solid tumors, we show that these bispecific Abs are cytotoxic to human tumor cell lines that express the Ag and that the modular design allows a functional exchange of the targeting moiety. Direct comparison with clinically approved Abs demonstrates a superior ability of the bispecific Abs to induce complement-dependent cytotoxicity. The efficacy of the bispecific Abs to activate complement strongly depends on the epitope of the C1q binding Ab, demonstrating that the spatial orientation of the C1 complex upon Ag engagement is a critical factor for efficient complement activation. Collectively, our data provide insight into the mechanism of complement activation and provide a new platform for the development of immunotherapies.


Assuntos
Antineoplásicos , Complemento C1q , Humanos , Complemento C1q/metabolismo , Proteínas do Sistema Complemento , Ativação do Complemento , Linhagem Celular Tumoral
2.
J Immunol ; 205(8): 2287-2300, 2020 10 15.
Artigo em Inglês | MEDLINE | ID: mdl-32938727

RESUMO

The complement system is an intricate cascade of the innate immune system and plays a key role in microbial defense, inflammation, organ development, and tissue regeneration. There is increasing interest in developing complement regulatory and inhibitory agents to treat complement dysfunction. In this study, we describe the nanobody hC3Nb3, which is specific for the C-terminal C345c domain of human and mouse complement component C3/C3b/C3c and potently inhibits C3 cleavage by the alternative pathway. A high-resolution structure of the hC3Nb3-C345c complex explains how the nanobody blocks proconvertase assembly. Surprisingly, although the nanobody does not affect classical pathway-mediated C3 cleavage, hC3Nb3 inhibits classical pathway-driven hemolysis, suggesting that the C-terminal domain of C3b has an important function in classical pathway C5 convertase activity. The hC3Nb3 nanobody binds C3 with low nanomolar affinity in an SDS-resistant complex, and the nanobody is demonstrated to be a powerful reagent for C3 detection in immunohistochemistry and flow cytometry. Overall, the hC3Nb3 nanobody represents a potent inhibitor of both the alternative pathway and the terminal pathway, with possible applications in complement research, diagnostics, and therapeutics.


Assuntos
Complemento C3b/imunologia , C5 Convertase da Via Alternativa do Complemento/imunologia , Via Alternativa do Complemento/imunologia , Anticorpos de Domínio Único/imunologia , Animais , Células HEK293 , Humanos , Camundongos , Domínios Proteicos
3.
J Biol Chem ; 295(26): 8746-8758, 2020 06 26.
Artigo em Inglês | MEDLINE | ID: mdl-32376685

RESUMO

The complement system is a tightly controlled proteolytic cascade in the innate immune system, which tags intruding pathogens and dying host cells for clearance. An essential protein in this process is complement component C3. Uncontrolled complement activation has been implicated in several human diseases and disorders and has spurred the development of therapeutic approaches that modulate the complement system. Here, using purified proteins and several biochemical assays and surface plasmon resonance, we report that our nanobody, hC3Nb2, inhibits C3 deposition by all complement pathways. We observe that the hC3Nb2 nanobody binds human native C3 and its degradation products with low nanomolar affinity and does not interfere with the endogenous regulation of C3b deposition mediated by Factors H and I. Using negative stain EM analysis and functional assays, we demonstrate that hC3Nb2 inhibits the substrate-convertase interaction by binding to the MG3 and MG4 domains of C3 and C3b. Furthermore, we notice that hC3Nb2 is cross-reactive and inhibits the lectin and alternative pathway in murine serum. We conclude that hC3Nb2 is a potent, general, and versatile inhibitor of the human and murine complement cascades. Its cross-reactivity suggests that this nanobody may be valuable for analysis of complement activation within animal models of both acute and chronic diseases.


Assuntos
Ativação do Complemento/efeitos dos fármacos , Complemento C3/antagonistas & inibidores , Anticorpos de Domínio Único/farmacologia , Animais , Complemento C3/imunologia , Convertases de Complemento C3-C5/antagonistas & inibidores , Convertases de Complemento C3-C5/imunologia , Hemólise/efeitos dos fármacos , Humanos , Camundongos , Modelos Moleculares , Ovinos
4.
Nat Immunol ; 9(7): 753-60, 2008 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-18536718

RESUMO

To provide insight into the structural and functional properties of human complement component 5 (C5), we determined its crystal structure at a resolution of 3.1 A. The core of C5 adopted a structure resembling that of C3, with the domain arrangement at the position corresponding to the C3 thioester being very well conserved. However, in contrast to C3, the convertase cleavage site in C5 was ordered and the C345C domain flexibly attached to the core of C5. Binding of the tick C5 inhibitor OmCI to C5 resulted in stabilization of the global conformation of C5 but did not block the convertase cleavage site. The structure of C5 may render possible a structure-based approach for the design of new selective complement inhibitors.


Assuntos
Complemento C5/química , Complemento C5/metabolismo , Proteínas de Insetos/metabolismo , Estrutura Quaternária de Proteína , Animais , Proteínas de Artrópodes , Sítios de Ligação , Proteínas de Transporte , Complemento C3 , Cristalografia por Raios X , Humanos , Proteínas de Insetos/química , Ressonância de Plasmônio de Superfície
5.
Bioconjug Chem ; 31(5): 1295-1300, 2020 05 20.
Artigo em Inglês | MEDLINE | ID: mdl-32320218

RESUMO

Chemically modified antigen-binding proteins are widely applied for their targeting abilities in the fields of biotechnology, medicine, and diagnostics. However, the production of site-selectively modified proteins remains a challenge. Here, we have designed a chemical probe for the introduction of a reactive aldehyde on nanobodies by metal-complex-guided conjugation. The probe design allows for purification of the conjugates, and the aldehyde constitutes an efficient handle for further modification of the nanobodies. In vitro experiments confirmed the binding activity and selectivity of fluorescent conjugates toward the native antigen. Furthermore, the modification strategy allowed for production of a nanobody-drug conjugate that was active in vitro.


Assuntos
Aldeídos/química , Anticorpos de Domínio Único/química , Coloração e Rotulagem/métodos , Corantes Fluorescentes/química , Imunoconjugados/química
7.
J Biol Chem ; 293(17): 6269-6281, 2018 04 27.
Artigo em Inglês | MEDLINE | ID: mdl-29497000

RESUMO

The complement system is a complex, carefully regulated proteolytic cascade for which suppression of aberrant activation is of increasing clinical relevance, and inhibition of the complement alternative pathway is a subject of intense research. Here, we describe the nanobody hC3Nb1 that binds to multiple functional states of C3 with subnanomolar affinity. The nanobody causes a complete shutdown of alternative pathway activity in human and murine serum when present in concentrations comparable with that of C3, and hC3Nb1 is shown to prevent proconvertase assembly, as well as binding of the C3 substrate to C3 convertases. Our crystal structure of the C3b-hC3Nb1 complex and functional experiments demonstrate that proconvertase formation is blocked by steric hindrance between the nanobody and an Asn-linked glycan on complement factor B. In addition, hC3Nb1 is shown to prevent factor H binding to C3b, rationalizing its inhibition of factor I activity. Our results identify hC3Nb1 as a versatile, inexpensive, and powerful inhibitor of the alternative pathway in both human and murine in vitro model systems of complement activation.


Assuntos
Complexo Antígeno-Anticorpo/química , Complemento C3/química , Via Alternativa do Complemento , Anticorpos de Domínio Único/química , Animais , Complexo Antígeno-Anticorpo/imunologia , Camelídeos Americanos , Complemento C3/imunologia , Cristalografia por Raios X , Humanos , Camundongos , Estrutura Quaternária de Proteína , Anticorpos de Domínio Único/imunologia
9.
EMBO J ; 30(3): 606-16, 2011 Feb 02.
Artigo em Inglês | MEDLINE | ID: mdl-21217642

RESUMO

Complement acts as a danger-sensing system in the innate immune system, and its activation initiates a strong inflammatory response and cleavage of the proteins C3 and C5 by proteolytic enzymes, the convertases. These contain a non-catalytic substrate contacting subunit (C3b or C4b) in complex with a protease subunit (Bb or C2a). We determined the crystal structures of the C3b homologue cobra venom factor (CVF) in complex with C5, and in complex with C5 and the inhibitor SSL7 at 4.3 Å resolution. The structures reveal a parallel two-point attachment between C5 and CVF, where the presence of SSL7 only slightly affects the C5-CVF interface, explaining the IgA dependence for SSL7-mediated inhibition of C5 cleavage. CVF functions as a relatively rigid binding scaffold inducing a conformational change in C5, which positions its cleavage site in proximity to the serine protease Bb. A general model for substrate recognition by the convertases is presented based on the C5-CVF and C3b-Bb-SCIN structures. Prior knowledge concerning interactions between the endogenous convertases and their substrates is rationalized by this model.


Assuntos
Convertases de Complemento C3-C5/metabolismo , Complemento C5/metabolismo , Venenos Elapídicos/metabolismo , Exotoxinas/metabolismo , Modelos Moleculares , Complexos Multiproteicos/metabolismo , Conformação Proteica , Convertases de Complemento C3-C5/química , Complemento C5/química , Cristalografia , Venenos Elapídicos/química , Exotoxinas/química , Humanos , Complexos Multiproteicos/química
10.
Proc Natl Acad Sci U S A ; 109(38): 15425-30, 2012 Sep 18.
Artigo em Inglês | MEDLINE | ID: mdl-22949645

RESUMO

An essential aspect of innate immunity is recognition of molecular patterns on the surface of pathogens or altered self through the lectin and classical pathways, two of the three well-established activation pathways of the complement system. This recognition causes activation of the MASP-2 or the C1s serine proteases followed by cleavage of the protein C4. Here we present the crystal structures of the 203-kDa human C4 and the 245-kDa C4·MASP-2 substrate·enzyme complex. When C4 binds to MASP-2, substantial conformational changes in C4 are induced, and its scissile bond region becomes ordered and inserted into the protease catalytic site in a manner canonical to serine proteases. In MASP-2, an exosite located within the CCP domains recognizes the C4 C345C domain 60 Å from the scissile bond. Mutations in C4 and MASP-2 residues at the C345C-CCP interface inhibit the intermolecular interaction and C4 cleavage. The possible assembly of the huge in vivo enzyme-substrate complex consisting of glycan-bound mannan-binding lectin, MASP-2, and C4 is discussed. Our own and prior functional data suggest that C1s in the classical pathway of complement activated by, e.g., antigen-antibody complexes, also recognizes the C4 C345C domain through a CCP exosite. Our results provide a unified structural framework for understanding the early and essential step of C4 cleavage in the elimination of pathogens and altered self through two major pathways of complement activation.


Assuntos
Complemento C4/química , Sítios de Ligação , Cristalografia/métodos , Células HEK293 , Humanos , Imunidade Inata , Mananas/química , Serina Proteases Associadas a Proteína de Ligação a Manose/química , Conformação Molecular , Mutação , Ligação Proteica , Conformação Proteica , Estrutura Terciária de Proteína , Proteínas/química , Proteólise , Proteínas Recombinantes/química , Eletricidade Estática , Especificidade por Substrato
11.
J Virol ; 87(22): 12471-80, 2013 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-24027321

RESUMO

Influenza virus is a global health concern due to its unpredictable pandemic potential. This potential threat was realized in 2009 when an H1N1 virus emerged that resembled the 1918 virus in antigenicity but fortunately was not nearly as deadly. 5J8 is a human antibody that potently neutralizes a broad spectrum of H1N1 viruses, including the 1918 and 2009 pandemic viruses. Here, we present the crystal structure of 5J8 Fab in complex with a bacterially expressed and refolded globular head domain from the hemagglutinin (HA) of the A/California/07/2009 (H1N1) pandemic virus. 5J8 recognizes a conserved epitope in and around the receptor binding site (RBS), and its HCDR3 closely mimics interactions of the sialic acid receptor. Electron microscopy (EM) reconstructions of 5J8 Fab in complex with an HA trimer from a 1986 H1 strain and with an engineered stabilized HA trimer from the 2009 H1 pandemic virus showed a similar mode of binding. As for other characterized RBS-targeted antibodies, 5J8 uses avidity to extend its breadth and affinity against divergent H1 strains. 5J8 selectively interacts with HA insertion residue 133a, which is conserved in pandemic H1 strains and has precluded binding of other RBS-targeted antibodies. Thus, the RBS of divergent HAs is targeted by 5J8 and adds to the growing arsenal of common recognition motifs for design of therapeutics and vaccines. Moreover, consistent with previous studies, the bacterially expressed H1 HA properly refolds, retaining its antigenic structure, and presents a low-cost and rapid alternative for engineering and manufacturing candidate flu vaccines.


Assuntos
Anticorpos Antivirais/imunologia , Antígenos Virais/imunologia , Glicoproteínas de Hemaglutininação de Vírus da Influenza/imunologia , Vírus da Influenza A Subtipo H1N1/imunologia , Influenza Humana/imunologia , Pandemias , Receptores Virais/imunologia , Anticorpos Antivirais/metabolismo , Afinidade de Anticorpos , Antígenos Virais/metabolismo , Sítios de Ligação de Anticorpos , Cristalografia por Raios X , Glicoproteínas de Hemaglutininação de Vírus da Influenza/química , Glicoproteínas de Hemaglutininação de Vírus da Influenza/metabolismo , Humanos , Influenza Humana/epidemiologia , Influenza Humana/virologia , Modelos Moleculares , Conformação Proteica , Receptores Virais/metabolismo , Estados Unidos/epidemiologia
12.
Proc Natl Acad Sci U S A ; 107(8): 3681-6, 2010 Feb 23.
Artigo em Inglês | MEDLINE | ID: mdl-20133685

RESUMO

Staphylococcus aureus secretes the SSL7 protein as part of its immune evasion strategy. The protein binds both complement C5 and IgA, yet it is unclear whether SSL7 cross-links these two proteins and, if so, what purpose this serves the pathogen. We have isolated a stable IgA-SSL7-C5 complex, and our crystal structure of the C5-SSL7 complex confirms that binding to C5 occurs exclusively through the C-terminal beta-grasp domain of SSL7 leaving the OB domain free to interact with IgA. SSL7 interacts with C5 >70 A from the C5a cleavage site without inducing significant conformational changes in C5, and efficient inhibition of convertase cleavage of C5 is shown to be IgA dependent. Inhibition of C5a production and bacteriolysis are all shown to require C5 and IgA binding while inhibition of hemolysis is achieved by the C5 binding SSL7 beta-grasp domain alone. These results provide a conceptual and structural basis for the development of a highly specific complement inhibitor preventing only the formation of the lytic membrane attack complex without affecting the important signaling functions of C5a.


Assuntos
Complemento C5/antagonistas & inibidores , Complemento C5/química , Exotoxinas/imunologia , Staphylococcus aureus/imunologia , Animais , Complemento C5/imunologia , Cristalografia por Raios X , Humanos , Mutação , Estrutura Terciária de Proteína , Coelhos , Staphylococcus aureus/patogenicidade
13.
Methods Mol Biol ; 2227: 249-264, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-33847947

RESUMO

Activated complement component C4 (C4b) is the nonenzymatic component of the classical pathway (CP) convertases of the complement system. Preparation of C4 and C4b samples suitable for structural biology studies is challenging due to low yields and complexity of recombinant C4 production protocols reported so far and heterogeneity of C4 in native sources. Here we present a purification protocol for human C4 and describe sample preparation methods for structural investigation of C4 and its complexes by crystallography, small angle X-ray scattering, and electron microscopy.


Assuntos
Complemento C4/química , Complemento C4/isolamento & purificação , Proteômica/métodos , Biologia/métodos , Cromatografia em Gel/métodos , Cristalografia por Raios X/métodos , Humanos , Troca Iônica , Microscopia Eletrônica/métodos , Coloração Negativa/métodos , Conformação Proteica , Espalhamento a Baixo Ângulo , Difração de Raios X/métodos
14.
Biomolecules ; 11(2)2021 02 17.
Artigo em Inglês | MEDLINE | ID: mdl-33671302

RESUMO

The complement system is part of the innate immune response, where it provides immediate protection from infectious agents and plays a fundamental role in homeostasis. Complement dysregulation occurs in several diseases, where the tightly regulated proteolytic cascade turns offensive. Prominent examples are atypical hemolytic uremic syndrome, paroxysmal nocturnal hemoglobinuria and Alzheimer's disease. Therapeutic intervention targeting complement activation may allow treatment of such debilitating diseases. In this review, we describe a panel of complement targeting nanobodies that allow modulation at different steps of the proteolytic cascade, from the activation of the C1 complex in the classical pathway to formation of the C5 convertase in the terminal pathway. Thorough structural and functional characterization has provided a deep mechanistic understanding of the mode of inhibition for each of the nanobodies. These complement specific nanobodies are novel powerful probes for basic research and offer new opportunities for in vivo complement modulation.


Assuntos
Ativação do Complemento , Proteínas do Sistema Complemento , Nanomedicina/métodos , Anticorpos de Domínio Único/química , Animais , Anticorpos Monoclonais Humanizados/uso terapêutico , Síndrome Hemolítico-Urêmica Atípica/tratamento farmacológico , Síndrome Hemolítico-Urêmica Atípica/imunologia , Complemento C1/química , Convertases de Complemento C3-C5/química , Epitopos/química , Hemoglobinúria Paroxística/tratamento farmacológico , Hemoglobinúria Paroxística/imunologia , Humanos , Imunidade Inata , Imunoglobulina G/imunologia , Inflamação , Conformação Molecular , Ligação Proteica , Proteólise
15.
Mol Immunol ; 124: 200-210, 2020 08.
Artigo em Inglês | MEDLINE | ID: mdl-32599335

RESUMO

The complement system represents a powerful part of the innate immune system capable of removing pathogens and damaged host cells. Nevertheless, only a subset of therapeutic antibodies are capable of inducing complement dependent cytotoxicity, which has fuelled the search for new strategies to potentiate complement activation. Properdin (FP) functions as a positive complement regulator by stabilizing the alternative pathway C3 convertase. Here, we explore a novel strategy for direct activation of the alternative pathway of complement using bi-specific single domain antibodies (nanobodies) that recruit endogenous FP to a cell surface. As a proof-of-principle, we generated bi-specific nanobodies with specificity toward FP and the validated cancer antigen epidermal growth factor receptor (EGFR) and tested their ability to activate complement onto cancer cell lines expressing EGFR. Treatment led to recruitment of FP, complement activation and significant deposition of C3 fragments on the cells in a manner sensitive to the geometry of FP recruitment. The bi-specific nanobodies induced complement dependent lysis of baby hamster kidney cells expressing human EGFR but were unable to lyse human tumour cells due to the presence of complement regulators. Our results confirm that FP can function as a surface bound focal point for initiation of complement activation independent of prior C3b deposition. However, recruitment of FP by bi-specific nanobodies appears insufficient for overcoming the inhibitory action of the negative complement regulators overexpressed by many human tumour cell lines. Our data provide general information on the efficacy of properdin as an initiator of complement but suggest that properdin recruitment on its own may have limited utility as a platform for potent complement activation on regulated cell surfaces.


Assuntos
Anticorpos Biespecíficos/imunologia , Ativação do Complemento/imunologia , Via Alternativa do Complemento/fisiologia , Properdina/imunologia , Anticorpos de Domínio Único/imunologia , Animais , Linhagem Celular Tumoral , Cricetinae , Receptores ErbB/imunologia , Humanos
16.
Front Immunol ; 11: 1504, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-32849513

RESUMO

The classical pathway of complement is important for protection against pathogens and in maintaining tissue homeostasis, but excessive or aberrant activation is directly linked to numerous pathologies. We describe the development and in vitro characterization of C1qNb75, a single domain antibody (nanobody) specific for C1q, the pattern recognition molecule of the classical pathway. C1qNb75 binds to the globular head modules of human C1q with sub-nanomolar affinity and impedes classical pathway mediated hemolysis by IgG and IgM. Crystal structure analysis revealed that C1qNb75 recognizes an epitope primarily located in the C1q B-chain that overlaps with the binding sites of IgG and IgM. Thus, C1qNb75 competitively prevents C1q from binding to IgG and IgM causing blockade of complement activation by the classical pathway. Overall, C1qNb75 represents a high-affinity nanobody-based inhibitor of IgG- and IgM-mediated activation of the classical pathway and may serve as a valuable reagent in mechanistic and functional studies of complement, and as an efficient inhibitor of complement under conditions of excessive CP activation.


Assuntos
Complemento C1q/imunologia , Receptores de Reconhecimento de Padrão/metabolismo , Anticorpos de Domínio Único/metabolismo , Afinidade de Anticorpos , Células Cultivadas , Ativação do Complemento , Complemento C1q/antagonistas & inibidores , Via Clássica do Complemento , Cristalografia por Raios X , Humanos , Imunoglobulina G/metabolismo , Imunoglobulina M/metabolismo , Ligação Proteica , Conformação Proteica , Receptores de Reconhecimento de Padrão/genética , Anticorpos de Domínio Único/química , Anticorpos de Domínio Único/genética , Relação Estrutura-Atividade
17.
Nat Commun ; 11(1): 3797, 2020 07 30.
Artigo em Inglês | MEDLINE | ID: mdl-32732998

RESUMO

Receptor-mediated perception of surface-exposed carbohydrates like lipo- and exo-polysaccharides (EPS) is important for non-self recognition and responses to microbial associated molecular patterns in mammals and plants. In legumes, EPS are monitored and can either block or promote symbiosis with rhizobia depending on their molecular composition. To establish a deeper understanding of receptors involved in EPS recognition, we determined the structure of the Lotus japonicus (Lotus) exopolysaccharide receptor 3 (EPR3) ectodomain. EPR3 forms a compact structure built of three putative carbohydrate-binding modules (M1, M2 and LysM3). M1 and M2 have unique ßαßß and ßαß folds that have not previously been observed in carbohydrate binding proteins, while LysM3 has a canonical ßααß fold. We demonstrate that this configuration is a structural signature for a ubiquitous class of receptors in the plant kingdom. We show that EPR3 is promiscuous, suggesting that plants can monitor complex microbial communities though this class of receptors.


Assuntos
Lipopolissacarídeos/metabolismo , Lotus/microbiologia , Lotus/fisiologia , Mesorhizobium/metabolismo , Proteínas de Plantas/metabolismo , Sequência de Aminoácidos , Mesorhizobium/genética , Fixação de Nitrogênio/fisiologia , Proteínas de Plantas/genética , Dobramento de Proteína , Nódulos Radiculares de Plantas/microbiologia , Nódulos Radiculares de Plantas/fisiologia , Simbiose/fisiologia
18.
Front Immunol ; 10: 2007, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-31507604

RESUMO

Properdin (FP) is a positive regulator of the immune system stimulating the activity of the proteolytically active C3 convertase C3bBb in the alternative pathway of the complement system. Here we present two crystal structures of FP and two structures of convertase bound FP. A structural core formed by three thrombospondin repeats (TSRs) and a TB domain harbors the convertase binding site in FP that mainly interacts with C3b. Stabilization of the interaction between the C3b C-terminus and the MIDAS bound Mg2+ in the Bb protease by FP TSR5 is proposed to underlie FP convertase stabilization. Intermolecular contacts between FP and the convertase subunits suggested by the structure were confirmed by binding experiments. FP is shown to inhibit C3b degradation by FI due to a direct competition for a common binding site on C3b. FP oligomers are held together by two sets of intermolecular contacts, where the first is formed by the TB domain from one FP molecule and TSR4 from another. The second and largest interface is formed by TSR1 and TSR6 from the same two FP molecules. Flexibility at four hinges between thrombospondin repeats is suggested to enable the oligomeric, polydisperse, and extended architecture of FP. Our structures rationalize the effects of mutations associated with FP deficiencies and provide a structural basis for the analysis of FP function in convertases and its possible role in pattern recognition.


Assuntos
Convertases de Complemento C3-C5/química , Proteínas do Sistema Complemento/química , Properdina/química , Multimerização Proteica , Sítios de Ligação , Convertases de Complemento C3-C5/metabolismo , Células HEK293 , Humanos , Modelos Moleculares , Mutação , Properdina/metabolismo , Ligação Proteica , Conformação Proteica , Processamento de Proteína Pós-Traducional , Proteólise , Relação Estrutura-Atividade
19.
Nat Commun ; 9(1): 7, 2018 01 02.
Artigo em Inglês | MEDLINE | ID: mdl-29295972

RESUMO

Anti-IgE therapeutics interfere with the ability of IgE to bind to its receptors on effector cells. Here we report the crystal structure of an anti-IgE single-domain antibody in complex with an IgE Fc fragment, revealing how the antibody inhibits interactions between IgE and the two receptors FcεRI and CD23. The epitope overlaps only slightly with the FcεRI-binding site but significantly with the CD23-binding site. Solution scattering studies of the IgE Fc reveal that antibody binding induces a half-bent conformation in between the well-known bent and extended IgE Fc conformations. The antibody acts as functional homolog of CD23 and induces a closed conformation of IgE Fc incompatible with FcεRI binding. Notably the antibody displaces IgE from both CD23 and FcεRI, and abrogates allergen-mediated basophil activation and facilitated allergen binding. The inhibitory mechanism might facilitate strategies for the future development of anti-IgE therapeutics for treatment of allergic diseases.


Assuntos
Epitopos/química , Imunoglobulina E/química , Receptores de IgE/química , Anticorpos Anti-Idiotípicos/química , Anticorpos Anti-Idiotípicos/metabolismo , Sítios de Ligação , Cristalografia por Raios X , Epitopos/metabolismo , Humanos , Imunoglobulina E/metabolismo , Fragmentos Fc das Imunoglobulinas/química , Fragmentos Fc das Imunoglobulinas/metabolismo , Modelos Moleculares , Ligação Proteica , Conformação Proteica , Receptores de IgE/metabolismo , Anticorpos de Domínio Único/química , Anticorpos de Domínio Único/metabolismo
20.
Science ; 362(6414): 598-602, 2018 11 02.
Artigo em Inglês | MEDLINE | ID: mdl-30385580

RESUMO

Broadly neutralizing antibodies against highly variable pathogens have stimulated the design of vaccines and therapeutics. We report the use of diverse camelid single-domain antibodies to influenza virus hemagglutinin to generate multidomain antibodies with impressive breadth and potency. Multidomain antibody MD3606 protects mice against influenza A and B infection when administered intravenously or expressed locally from a recombinant adeno-associated virus vector. Crystal and single-particle electron microscopy structures of these antibodies with hemagglutinins from influenza A and B viruses reveal binding to highly conserved epitopes. Collectively, our findings demonstrate that multidomain antibodies targeting multiple epitopes exhibit enhanced virus cross-reactivity and potency. In combination with adeno-associated virus-mediated gene delivery, they may provide an effective strategy to prevent infection with influenza virus and other highly variable pathogens.


Assuntos
Anticorpos Neutralizantes/imunologia , Anticorpos Antivirais/imunologia , Camelídeos Americanos/imunologia , Glicoproteínas de Hemaglutininação de Vírus da Influenza/imunologia , Vírus da Influenza A/imunologia , Vírus da Influenza B/imunologia , Vacinas contra Influenza/imunologia , Infecções por Orthomyxoviridae/prevenção & controle , Animais , Anticorpos Neutralizantes/química , Anticorpos Neutralizantes/ultraestrutura , Anticorpos Antivirais/química , Anticorpos Antivirais/ultraestrutura , Cristalografia por Raios X , Cães , Feminino , Epitopos Imunodominantes/química , Epitopos Imunodominantes/genética , Epitopos Imunodominantes/imunologia , Células Madin Darby de Rim Canino , Camundongos , Camundongos Endogâmicos BALB C , Testes de Neutralização , Biblioteca de Peptídeos , Proteínas Recombinantes/química , Proteínas Recombinantes/genética , Proteínas Recombinantes/imunologia , Anticorpos de Domínio Único
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA