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.
Artículo en Inglés | MEDLINE | ID: mdl-37350633

RESUMEN

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.


Asunto(s)
Antineoplásicos , Complemento C1q , Humanos , Complemento C1q/metabolismo , Proteínas del Sistema Complemento , Activación de Complemento , Línea Celular Tumoral
4.
Methods Mol Biol ; 2227: 249-264, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-33847947

RESUMEN

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.


Asunto(s)
Complemento C4/química , Complemento C4/aislamiento & purificación , Proteómica/métodos , Biología/métodos , Cromatografía en Gel/métodos , Cristalografía por Rayos X/métodos , Humanos , Intercambio Iónico , Microscopía Electrónica/métodos , Coloración Negativa/métodos , Conformación Proteica , Dispersión del Ángulo Pequeño , Difracción de Rayos X/métodos
5.
Biomolecules ; 11(2)2021 02 17.
Artículo en Inglés | MEDLINE | ID: mdl-33671302

RESUMEN

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.


Asunto(s)
Activación de Complemento , Proteínas del Sistema Complemento , Nanomedicina/métodos , Anticuerpos de Dominio Único/química , Animales , Anticuerpos Monoclonales Humanizados/uso terapéutico , Síndrome Hemolítico Urémico Atípico/tratamiento farmacológico , Síndrome Hemolítico Urémico Atípico/inmunología , Complemento C1/química , Convertasas de Complemento C3-C5/química , Epítopos/química , Hemoglobinuria Paroxística/tratamiento farmacológico , Hemoglobinuria Paroxística/inmunología , Humanos , Inmunidad Innata , Inmunoglobulina G/inmunología , Inflamación , Conformación Molecular , Unión Proteica , Proteolisis
6.
J Immunol ; 205(8): 2287-2300, 2020 10 15.
Artículo en Inglés | MEDLINE | ID: mdl-32938727

RESUMEN

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.


Asunto(s)
Complemento C3b/inmunología , C5 Convertasa de la Vía Alternativa del Complemento/inmunología , Vía Alternativa del Complemento/inmunología , Anticuerpos de Dominio Único/inmunología , Animales , Células HEK293 , Humanos , Ratones , Dominios Proteicos
7.
Front Immunol ; 11: 1504, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32849513

RESUMEN

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.


Asunto(s)
Complemento C1q/inmunología , Receptores de Reconocimiento de Patrones/metabolismo , Anticuerpos de Dominio Único/metabolismo , Afinidad de Anticuerpos , Células Cultivadas , Activación de Complemento , Complemento C1q/antagonistas & inhibidores , Vía Clásica del Complemento , Cristalografía por Rayos X , Humanos , Inmunoglobulina G/metabolismo , Inmunoglobulina M/metabolismo , Unión Proteica , Conformación Proteica , Receptores de Reconocimiento de Patrones/genética , Anticuerpos de Dominio Único/química , Anticuerpos de Dominio Único/genética , Relación Estructura-Actividad
8.
Nat Commun ; 11(1): 3797, 2020 07 30.
Artículo en Inglés | MEDLINE | ID: mdl-32732998

RESUMEN

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.


Asunto(s)
Lipopolisacáridos/metabolismo , Lotus/microbiología , Lotus/fisiología , Mesorhizobium/metabolismo , Proteínas de Plantas/metabolismo , Secuencia de Aminoácidos , Mesorhizobium/genética , Fijación del Nitrógeno/fisiología , Proteínas de Plantas/genética , Pliegue de Proteína , Nódulos de las Raíces de las Plantas/microbiología , Nódulos de las Raíces de las Plantas/fisiología , Simbiosis/fisiología
9.
Mol Immunol ; 124: 200-210, 2020 08.
Artículo en Inglés | MEDLINE | ID: mdl-32599335

RESUMEN

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.


Asunto(s)
Anticuerpos Biespecíficos/inmunología , Activación de Complemento/inmunología , Vía Alternativa del Complemento/fisiología , Properdina/inmunología , Anticuerpos de Dominio Único/inmunología , Animales , Línea Celular Tumoral , Cricetinae , Receptores ErbB/inmunología , Humanos
10.
J Biol Chem ; 295(26): 8746-8758, 2020 06 26.
Artículo en Inglés | MEDLINE | ID: mdl-32376685

RESUMEN

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.


Asunto(s)
Activación de Complemento/efectos de los fármacos , Complemento C3/antagonistas & inhibidores , Anticuerpos de Dominio Único/farmacología , Animales , Complemento C3/inmunología , Convertasas de Complemento C3-C5/antagonistas & inhibidores , Convertasas de Complemento C3-C5/inmunología , Hemólisis/efectos de los fármacos , Humanos , Ratones , Modelos Moleculares , Ovinos
11.
Bioconjug Chem ; 31(5): 1295-1300, 2020 05 20.
Artículo en Inglés | MEDLINE | ID: mdl-32320218

RESUMEN

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.


Asunto(s)
Aldehídos/química , Anticuerpos de Dominio Único/química , Coloración y Etiquetado/métodos , Colorantes Fluorescentes/química , Inmunoconjugados/química
12.
Front Immunol ; 10: 2007, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-31507604

RESUMEN

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.


Asunto(s)
Convertasas de Complemento C3-C5/química , Proteínas del Sistema Complemento/química , Properdina/química , Multimerización de Proteína , Sitios de Unión , Convertasas de Complemento C3-C5/metabolismo , Células HEK293 , Humanos , Modelos Moleculares , Mutación , Properdina/metabolismo , Unión Proteica , Conformación Proteica , Procesamiento Proteico-Postraduccional , Proteolisis , Relación Estructura-Actividad
13.
Science ; 362(6414): 598-602, 2018 11 02.
Artículo en Inglés | MEDLINE | ID: mdl-30385580

RESUMEN

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.


Asunto(s)
Anticuerpos Neutralizantes/inmunología , Anticuerpos Antivirales/inmunología , Camélidos del Nuevo Mundo/inmunología , Glicoproteínas Hemaglutininas del Virus de la Influenza/inmunología , Virus de la Influenza A/inmunología , Virus de la Influenza B/inmunología , Vacunas contra la Influenza/inmunología , Infecciones por Orthomyxoviridae/prevención & control , Animales , Anticuerpos Neutralizantes/química , Anticuerpos Neutralizantes/ultraestructura , Anticuerpos Antivirales/química , Anticuerpos Antivirales/ultraestructura , Cristalografía por Rayos X , Perros , Femenino , Epítopos Inmunodominantes/química , Epítopos Inmunodominantes/genética , Epítopos Inmunodominantes/inmunología , Células de Riñón Canino Madin Darby , Ratones , Ratones Endogámicos BALB C , Pruebas de Neutralización , Biblioteca de Péptidos , Proteínas Recombinantes/química , Proteínas Recombinantes/genética , Proteínas Recombinantes/inmunología , Anticuerpos de Dominio Único
14.
J Biol Chem ; 293(17): 6269-6281, 2018 04 27.
Artículo en Inglés | MEDLINE | ID: mdl-29497000

RESUMEN

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.


Asunto(s)
Complejo Antígeno-Anticuerpo/química , Complemento C3/química , Vía Alternativa del Complemento , Anticuerpos de Dominio Único/química , Animales , Complejo Antígeno-Anticuerpo/inmunología , Camélidos del Nuevo Mundo , Complemento C3/inmunología , Cristalografía por Rayos X , Humanos , Ratones , Estructura Cuaternaria de Proteína , Anticuerpos de Dominio Único/inmunología
15.
Nat Commun ; 9(1): 7, 2018 01 02.
Artículo en Inglés | MEDLINE | ID: mdl-29295972

RESUMEN

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.


Asunto(s)
Epítopos/química , Inmunoglobulina E/química , Receptores de IgE/química , Anticuerpos Antiidiotipos/química , Anticuerpos Antiidiotipos/metabolismo , Sitios de Unión , Cristalografía por Rayos X , Epítopos/metabolismo , Humanos , Inmunoglobulina E/metabolismo , Fragmentos Fc de Inmunoglobulinas/química , Fragmentos Fc de Inmunoglobulinas/metabolismo , Modelos Moleculares , Unión Proteica , Conformación Proteica , Receptores de IgE/metabolismo , Anticuerpos de Dominio Único/química , Anticuerpos de Dominio Único/metabolismo
16.
Science ; 349(6254): 1301-6, 2015 Sep 18.
Artículo en Inglés | MEDLINE | ID: mdl-26303961

RESUMEN

The identification of human broadly neutralizing antibodies (bnAbs) targeting the hemagglutinin (HA) stem revitalized hopes of developing a universal influenza vaccine. Using a rational design and library approach, we engineered stable HA stem antigens ("mini-HAs") based on an H1 subtype sequence. Our most advanced candidate exhibits structural and bnAb binding properties comparable to those of full-length HA, completely protects mice in lethal heterologous and heterosubtypic challenge models, and reduces fever after sublethal challenge in cynomolgus monkeys. Antibodies elicited by this mini-HA in mice and nonhuman primates bound a wide range of HAs, competed with human bnAbs for HA stem binding, neutralized H5N1 viruses, and mediated antibody-dependent effector activity. These results represent a proof of concept for the design of HA stem mimics that elicit bnAbs against influenza A group 1 viruses.


Asunto(s)
Glicoproteínas Hemaglutininas del Virus de la Influenza/química , Glicoproteínas Hemaglutininas del Virus de la Influenza/inmunología , Subtipo H1N1 del Virus de la Influenza A/inmunología , Subtipo H5N1 del Virus de la Influenza A/inmunología , Vacunas contra la Influenza/inmunología , Gripe Humana/prevención & control , Animales , Anticuerpos Neutralizantes/inmunología , Anticuerpos Antivirales/inmunología , Humanos , Ratones , Multimerización de Proteína , Estructura Secundaria de Proteína
17.
J Virol ; 87(22): 12471-80, 2013 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-24027321

RESUMEN

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.


Asunto(s)
Anticuerpos Antivirales/inmunología , Antígenos Virales/inmunología , Glicoproteínas Hemaglutininas del Virus de la Influenza/inmunología , Subtipo H1N1 del Virus de la Influenza A/inmunología , Gripe Humana/inmunología , Pandemias , Receptores Virales/inmunología , Anticuerpos Antivirales/metabolismo , Afinidad de Anticuerpos , Antígenos Virales/metabolismo , Sitios de Unión de Anticuerpos , Cristalografía por Rayos X , Glicoproteínas Hemaglutininas del Virus de la Influenza/química , Glicoproteínas Hemaglutininas del Virus de la Influenza/metabolismo , Humanos , Gripe Humana/epidemiología , Gripe Humana/virología , Modelos Moleculares , Conformación Proteica , Receptores Virales/metabolismo , Estados Unidos/epidemiología
18.
Antiviral Res ; 98(3): 476-83, 2013 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-23583287

RESUMEN

Despite available antivirals and vaccines, influenza continues to be a major cause of mortality worldwide. Vaccination generally induces an effective, but strain-specific antibody response. As the virus continually evolves, new vaccines have to be administered almost annually when a novel strain becomes dominant. Furthermore, the sporadic emerging resistance to neuraminidase inhibitors among circulating strains suggests an urgent need for new therapeutic agents. Recently, several cross-reactive antibodies have been described, which neutralize an unprecedented spectrum of influenza viruses. These broadly neutralizing antibodies generally target conserved functional regions on the major influenza surface glycoprotein hemagglutinin (HA). The characterization of their neutralization breadth and epitopes on HA could stimulate the development of new antibody-based antivirals and broader influenza vaccines. This article forms part of a symposium in Antiviral Research on "Treatment of influenza: targeting the virus or the host."


Asunto(s)
Anticuerpos Neutralizantes/inmunología , Anticuerpos Antivirales/inmunología , Gripe Humana/prevención & control , Anticuerpos Neutralizantes/química , Anticuerpos Antivirales/química , Afinidad de Anticuerpos , Complejo Antígeno-Anticuerpo , Diseño de Fármacos , Epítopos/química , Epítopos/inmunología , Glicoproteínas Hemaglutininas del Virus de la Influenza/química , Glicoproteínas Hemaglutininas del Virus de la Influenza/inmunología , Humanos , Vacunas contra la Influenza/inmunología , Gripe Humana/inmunología , Pruebas de Neutralización , Orthomyxoviridae/inmunología , Conformación Proteica , Vacunación
19.
Proc Natl Acad Sci U S A ; 109(38): 15425-30, 2012 Sep 18.
Artículo en Inglés | MEDLINE | ID: mdl-22949645

RESUMEN

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.


Asunto(s)
Complemento C4/química , Sitios de Unión , Cristalografía/métodos , Células HEK293 , Humanos , Inmunidad Innata , Mananos/química , Serina Proteasas Asociadas a la Proteína de Unión a la Manosa/química , Conformación Molecular , Mutación , Unión Proteica , Conformación Proteica , Estructura Terciaria de Proteína , Proteínas/química , Proteolisis , Proteínas Recombinantes/química , Electricidad Estática , Especificidad por Sustrato
20.
Science ; 337(6100): 1343-8, 2012 Sep 14.
Artículo en Inglés | MEDLINE | ID: mdl-22878502

RESUMEN

Identification of broadly neutralizing antibodies against influenza A viruses has raised hopes for the development of monoclonal antibody-based immunotherapy and "universal" vaccines for influenza. However, a substantial part of the annual flu burden is caused by two cocirculating, antigenically distinct lineages of influenza B viruses. Here, we report human monoclonal antibodies, CR8033, CR8071, and CR9114, that protect mice against lethal challenge from both lineages. Antibodies CR8033 and CR8071 recognize distinct conserved epitopes in the head region of the influenza B hemagglutinin (HA), whereas CR9114 binds a conserved epitope in the HA stem and protects against lethal challenge with influenza A and B viruses. These antibodies may inform on development of monoclonal antibody-based treatments and a universal flu vaccine for all influenza A and B viruses.


Asunto(s)
Anticuerpos Monoclonales/inmunología , Glicoproteínas Hemaglutininas del Virus de la Influenza/inmunología , Epítopos Inmunodominantes/inmunología , Virus de la Influenza B/inmunología , Vacunas contra la Influenza/inmunología , Infecciones por Orthomyxoviridae/prevención & control , Secuencia de Aminoácidos , Animales , Anticuerpos Monoclonales/química , Anticuerpos Neutralizantes/química , Anticuerpos Neutralizantes/inmunología , Secuencia Conservada , Humanos , Epítopos Inmunodominantes/química , Ratones , Datos de Secuencia Molecular , Pruebas de Neutralización , Conformación Proteica
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA