Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 11 de 11
Filtrar
Más filtros











Intervalo de año de publicación
1.
Science ; 384(6703): eadm8693, 2024 Jun 28.
Artículo en Inglés | MEDLINE | ID: mdl-38935733

RESUMEN

Measles virus (MeV) presents a public health threat that is escalating as vaccine coverage in the general population declines and as populations of immunocompromised individuals, who cannot be vaccinated, increase. There are no approved therapeutics for MeV. Neutralizing antibodies targeting viral fusion are one potential therapeutic approach but have not yet been structurally characterized or advanced to clinical use. We present cryo-electron microscopy (cryo-EM) structures of prefusion F alone [2.1-angstrom (Å) resolution], F complexed with a fusion-inhibitory peptide (2.3-Å resolution), F complexed with the neutralizing and protective monoclonal antibody (mAb) 77 (2.6-Å resolution), and an additional structure of postfusion F (2.7-Å resolution). In vitro assays and examination of additional EM classes show that mAb 77 binds prefusion F, arrests F in an intermediate state, and prevents transition to the postfusion conformation. These structures shed light on antibody-mediated neutralization that involves arrest of fusion proteins in an intermediate state.


Asunto(s)
Anticuerpos Monoclonales , Anticuerpos Neutralizantes , Anticuerpos Antivirales , Microscopía por Crioelectrón , Virus del Sarampión , Proteínas Virales de Fusión , Anticuerpos Neutralizantes/inmunología , Anticuerpos Neutralizantes/química , Virus del Sarampión/inmunología , Virus del Sarampión/química , Proteínas Virales de Fusión/inmunología , Proteínas Virales de Fusión/química , Anticuerpos Monoclonales/inmunología , Anticuerpos Monoclonales/química , Anticuerpos Antivirales/inmunología , Anticuerpos Antivirales/química , Humanos , Conformación Proteica
2.
Front Immunol ; 15: 1329846, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38529279

RESUMEN

Understanding adaptive immunity against SARS-CoV-2 is a major requisite for the development of effective vaccines and treatments for COVID-19. CD4+ T cells play an integral role in this process primarily by generating antiviral cytokines and providing help to antibody-producing B cells. To empower detailed studies of SARS-CoV-2-specific CD4+ T cell responses in mouse models, we comprehensively mapped I-Ab-restricted epitopes for the spike and nucleocapsid proteins of the BA.1 variant of concern via IFNγ ELISpot assay. This was followed by the generation of corresponding peptide:MHCII tetramer reagents to directly stain epitope-specific T cells. Using this rigorous validation strategy, we identified 6 immunogenic epitopes in spike and 3 in nucleocapsid, all of which are conserved in the ancestral Wuhan strain. We also validated a previously identified epitope from Wuhan that is absent in BA.1. These epitopes and tetramers will be invaluable tools for SARS-CoV-2 antigen-specific CD4+ T cell studies in mice.


Asunto(s)
COVID-19 , SARS-CoV-2 , Animales , Ratones , Linfocitos T CD4-Positivos , Epítopos de Linfocito T , Nucleocápside/química , Péptidos/química , SARS-CoV-2/química , Antígenos de Histocompatibilidad Clase II/química , Glicoproteína de la Espiga del Coronavirus/química
3.
Cell Rep ; 39(8): 110841, 2022 05 24.
Artículo en Inglés | MEDLINE | ID: mdl-35613585

RESUMEN

Lassa virus (LASV) is the etiologic agent of Lassa Fever, a hemorrhagic disease that is endemic to West Africa. During LASV infection, LASV glycoprotein (GP) engages with multiple host receptors for cell entry. Neutralizing antibodies against GP are rare and principally target quaternary epitopes displayed only on the metastable, pre-fusion conformation of GP. Currently, the structural features of the neutralizing GPC-A antibody competition group are understudied. Structures of two GPC-A antibodies presented here demonstrate that they bind the side of the pre-fusion GP trimer, bridging the GP1 and GP2 subunits. Complementary biochemical analyses indicate that antibody 25.10C, which is broadly specific, neutralizes by inhibiting binding of the endosomal receptor LAMP1 and also by blocking membrane fusion. The other GPC-A antibody, 36.1F, which is lineage-specific, prevents LAMP1 association only. These data illuminate a site of vulnerability on LASV GP and will guide efforts to elicit broadly reactive therapeutics and vaccines.


Asunto(s)
Fiebre de Lassa , Virus Lassa , Anticuerpos Neutralizantes , Epítopos , Glicoproteínas/metabolismo , Humanos , Fiebre de Lassa/prevención & control , Virus Lassa/metabolismo , Proteínas del Envoltorio Viral
4.
Cell ; 178(4): 1004-1015.e14, 2019 08 08.
Artículo en Inglés | MEDLINE | ID: mdl-31398326

RESUMEN

Lassa virus (LASV) causes hemorrhagic fever and is endemic in West Africa. Protective antibody responses primarily target the LASV surface glycoprotein (GPC), and GPC-B competition group antibodies often show potent neutralizing activity in humans. However, which features confer potent and broadly neutralizing antibody responses is unclear. Here, we compared three crystal structures of LASV GPC complexed with GPC-B antibodies of varying neutralization potency. Each GPC-B antibody recognized an overlapping epitope involved in binding of two adjacent GPC monomers and preserved the prefusion trimeric conformation. Differences among GPC-antibody interactions highlighted specific residues that enhance neutralization. Using structure-guided amino acid substitutions, we increased the neutralization potency and breadth of these antibodies to include all major LASV lineages. The ability to define antibody residues that allow potent and broad neutralizing activity, together with findings from analyses of inferred germline precursors, is critical to develop potent therapeutics and for vaccine design and assessment.


Asunto(s)
Anticuerpos Neutralizantes/inmunología , Anticuerpos Antivirales/inmunología , Células Germinativas/inmunología , Fiebre de Lassa/inmunología , Virus Lassa/inmunología , Glicoproteínas de Membrana/química , Proteínas del Envoltorio Viral/química , Animales , Antígenos Virales/inmunología , Chlorocebus aethiops , Drosophila/citología , Epítopos/química , Epítopos/inmunología , Células HEK293 , Humanos , Fiebre de Lassa/virología , Glicoproteínas de Membrana/inmunología , Estructura Secundaria de Proteína , Células Vero , Proteínas del Envoltorio Viral/inmunología , Vacunas Virales/inmunología
5.
Curr Opin Virol ; 31: 52-58, 2018 08.
Artículo en Inglés | MEDLINE | ID: mdl-29843991

RESUMEN

The structure of a prefusion arenavirus GPC was enigmatic for many years, owing to the metastable and non-covalent nature of the association between the receptor binding and fusion subunits. Recent engineering efforts to stabilize the glycoprotein of the Old World arenavirus Lassa in a native, yet cleaved state, allowed the first structure of any arenavirus prefusion GPC trimer to be determined. Comparison of this structure with the structures of other arenavirus glycoprotein subunits reveals surprising findings: that the receptor binding subunit, GP1, of Lassa virus is conformationally labile, while the GP1 subunit of New World arenaviruses is not, and that the arenavirus GPC adopts a trimeric state unlike other glycoproteins with similar fusion machinery. Structural analysis, combined with recent biochemical data regarding antibody epitopes and receptor binding requirements, provides a basis for rational vaccine design.


Asunto(s)
Epítopos/inmunología , Glicoproteínas/química , Glicoproteínas/metabolismo , Virus Lassa/química , Arenavirus/metabolismo , Humanos , Virus Lassa/metabolismo , Unión Proteica , Estructura Terciaria de Proteína , Proteínas del Envoltorio Viral/metabolismo , Internalización del Virus
6.
mSphere ; 3(3)2018.
Artículo en Inglés | MEDLINE | ID: mdl-29720525

RESUMEN

Arenaviruses pose a major public health threat and cause numerous infections in humans each year. Although most viruses belonging to this family do not cause disease in humans, some arenaviruses, such as Lassa virus and Machupo virus, are the etiological agents of lethal hemorrhagic fevers. The absence of a currently licensed vaccine and the highly pathogenic nature of these viruses both make the necessity of developing viable vaccines and therapeutics all the more urgent. Arenaviruses have a single glycoprotein on the surface of virions, the glycoprotein complex (GPC), and this protein can be used as a target for vaccine development. Here, we describe immunization strategies to generate monoclonal antibodies (MAbs) that cross-react between the glycoprotein complexes of both Old World and New World arenaviruses. Several monoclonal antibodies isolated from immunized mice were highly cross-reactive, binding a range of Old World arenavirus glycoproteins, including that of Lassa virus. One such monoclonal antibody, KL-AV-2A1, bound to GPCs of both New World and Old World viruses, including Lassa and Machupo viruses. These cross-reactive antibodies bound to epitopes present on the glycoprotein 2 subunit of the glycoprotein complex, which is relatively conserved among arenaviruses. Monoclonal antibodies binding to these epitopes, however, did not inhibit viral entry as they failed to neutralize a replication-competent vesicular stomatitis virus pseudotyped with the Lassa virus glycoprotein complex in vitro In addition, no protection from virus challenge was observed in in vivo mouse models. Even so, these monoclonal antibodies might still prove to be useful in the development of clinical and diagnostic assays.IMPORTANCE Several viruses in the Arenaviridae family infect humans and cause severe hemorrhagic fevers which lead to high case fatality rates. Due to their pathogenicity and geographic tropisms, these viruses remain very understudied. As a result, an effective vaccine or therapy is urgently needed. Here, we describe efforts to produce cross-reactive monoclonal antibodies that bind to both New and Old World arenaviruses. All of our MAbs seem to be nonneutralizing and nonprotective and target subunit 2 of the glycoprotein. Due to the lack of reagents such as recombinant glycoproteins and antibodies for rapid detection assays, our MAbs could be beneficial as analytic and diagnostic tools.


Asunto(s)
Anticuerpos Antivirales/inmunología , Arenavirus del Nuevo Mundo/inmunología , Arenavirus del Viejo Mundo/inmunología , Reacciones Cruzadas , Glicoproteínas/inmunología , Proteínas Estructurales Virales/inmunología , Animales , Anticuerpos Monoclonales/inmunología , Anticuerpos Antivirales/aislamiento & purificación , Infecciones por Arenaviridae/inmunología , Infecciones por Arenaviridae/prevención & control , Modelos Animales de Enfermedad , Mapeo Epitopo , Epítopos de Linfocito B/inmunología , Ratones
7.
Cell Host Microbe ; 23(1): 101-109.e4, 2018 Jan 10.
Artículo en Inglés | MEDLINE | ID: mdl-29324225

RESUMEN

Since their first identification 50 years ago, marburgviruses have emerged several times, with 83%-90% lethality in the largest outbreaks. Although no vaccines or therapeutics are available for human use, the human antibody MR191 provides complete protection in non-human primates when delivered several days after inoculation of a lethal marburgvirus dose. The detailed neutralization mechanism of MR191 remains outstanding. Here we present a 3.2 Å crystal structure of MR191 complexed with a trimeric marburgvirus surface glycoprotein (GP). MR191 neutralizes by occupying the conserved receptor-binding site and competing with the host receptor Niemann-Pick C1. The structure illuminates previously disordered regions of GP including the stalk, fusion loop, CX6CC switch, and an N-terminal region of GP2 that wraps about the outside of GP1 to anchor a marburgvirus-specific "wing" antibody epitope. Virus escape mutations mapped far outside the MR191 receptor-binding site footprint suggest a role for these other regions in the GP quaternary structure.


Asunto(s)
Anticuerpos Monoclonales/inmunología , Anticuerpos Neutralizantes/inmunología , Anticuerpos Antivirales/inmunología , Marburgvirus/inmunología , Receptores Virales/inmunología , Receptores Virales/ultraestructura , Proteínas Virales de Fusión/inmunología , Proteínas Virales de Fusión/ultraestructura , Agrobacterium tumefaciens , Animales , Anticuerpos Monoclonales/ultraestructura , Sitios de Unión/inmunología , Proteínas Portadoras/inmunología , Línea Celular , Chlorocebus aethiops , Cristalografía por Rayos X , Drosophila melanogaster , Humanos , Péptidos y Proteínas de Señalización Intracelular , Marburgvirus/metabolismo , Glicoproteínas de Membrana/inmunología , Proteína Niemann-Pick C1 , Nicotiana , Células Vero , Proteínas del Envoltorio Viral/genética , Proteínas del Envoltorio Viral/inmunología , Acoplamiento Viral
8.
Science ; 356(6341): 923-928, 2017 06 02.
Artículo en Inglés | MEDLINE | ID: mdl-28572385

RESUMEN

The arenavirus Lassa causes severe hemorrhagic fever and a significant disease burden in West Africa every year. The glycoprotein, GPC, is the sole antigen expressed on the viral surface and the critical target for antibody-mediated neutralization. Here we present the crystal structure of the trimeric, prefusion ectodomain of Lassa GP bound to a neutralizing antibody from a human survivor at 3.2-angstrom resolution. The antibody extensively anchors two monomers together at the base of the trimer, and biochemical analysis suggests that it neutralizes by inhibiting conformational changes required for entry. This work illuminates pH-driven conformational changes in both receptor-binding and fusion subunits of Lassa virus, illustrates the unique assembly of the arenavirus glycoprotein spike, and provides a much-needed template for vaccine design against these threats to global health.


Asunto(s)
Anticuerpos Neutralizantes/química , Anticuerpos Neutralizantes/metabolismo , Virus Lassa/fisiología , Modelos Moleculares , Proteínas del Envoltorio Viral/química , Proteínas del Envoltorio Viral/metabolismo , Anticuerpos Antivirales/química , Anticuerpos Antivirales/metabolismo , Cristalización , Epítopos/química , Humanos , Concentración de Iones de Hidrógeno , Fiebre de Lassa/inmunología , Fiebre de Lassa/virología , Virus Lassa/química , Virus Lassa/inmunología , Unión Proteica , Conformación Proteica , Multimerización de Proteína , Estabilidad Proteica , Estructura Cuaternaria de Proteína , Internalización del Virus
9.
Nat Commun ; 7: 11544, 2016 05 10.
Artículo en Inglés | MEDLINE | ID: mdl-27161536

RESUMEN

Lassa fever is a severe multisystem disease that often has haemorrhagic manifestations. The epitopes of the Lassa virus (LASV) surface glycoproteins recognized by naturally infected human hosts have not been identified or characterized. Here we have cloned 113 human monoclonal antibodies (mAbs) specific for LASV glycoproteins from memory B cells of Lassa fever survivors from West Africa. One-half bind the GP2 fusion subunit, one-fourth recognize the GP1 receptor-binding subunit and the remaining fourth are specific for the assembled glycoprotein complex, requiring both GP1 and GP2 subunits for recognition. Notably, of the 16 mAbs that neutralize LASV, 13 require the assembled glycoprotein complex for binding, while the remaining 3 require GP1 only. Compared with non-neutralizing mAbs, neutralizing mAbs have higher binding affinities and greater divergence from germline progenitors. Some mAbs potently neutralize all four LASV lineages. These insights from LASV human mAb characterization will guide strategies for immunotherapeutic development and vaccine design.


Asunto(s)
Anticuerpos Monoclonales/inmunología , Anticuerpos Neutralizantes/inmunología , Anticuerpos Antivirales/inmunología , Virus Lassa/inmunología , Especificidad de Anticuerpos , Antígenos Virales/química , Antígenos Virales/genética , Antígenos Virales/inmunología , Arenavirus/inmunología , Reacciones Cruzadas , Mapeo Epitopo , Epítopos/química , Epítopos/genética , Epítopos/inmunología , Humanos , Fiebre de Lassa/inmunología , Fiebre de Lassa/prevención & control , Virus Lassa/genética , Modelos Moleculares , Mutagénesis Sitio-Dirigida , Eliminación de Secuencia , Proteínas del Envoltorio Viral/química , Proteínas del Envoltorio Viral/genética , Proteínas del Envoltorio Viral/inmunología
10.
Nat Struct Mol Biol ; 23(6): 513-521, 2016 06.
Artículo en Inglés | MEDLINE | ID: mdl-27111888

RESUMEN

Arenaviruses exist worldwide and can cause hemorrhagic fever and neurologic disease. A single glycoprotein expressed on the viral surface mediates entry into target cells. This glycoprotein, termed GPC, contains a membrane-associated signal peptide, a receptor-binding subunit termed GP1 and a fusion-mediating subunit termed GP2. Although GPC is a critical target of antibodies and vaccines, the structure of the metastable GP1-GP2 prefusion complex has remained elusive for all arenaviruses. Here we describe the crystal structure of the fully glycosylated prefusion GP1-GP2 complex of the prototypic arenavirus LCMV at 3.5 Å. This structure reveals the conformational changes that the arenavirus glycoprotein must undergo to cause fusion and illustrates the fusion regions and potential oligomeric states.


Asunto(s)
Virus de la Coriomeningitis Linfocítica/química , Glicoproteínas de Membrana/química , Proteínas del Envoltorio Viral/química , Animales , Línea Celular , Cristalografía por Rayos X , Drosophila , Glicosilación , Humanos , Coriomeningitis Linfocítica/metabolismo , Coriomeningitis Linfocítica/virología , Virus de la Coriomeningitis Linfocítica/fisiología , Glicoproteínas de Membrana/metabolismo , Modelos Moleculares , Conformación Proteica , Multimerización de Proteína , Señales de Clasificación de Proteína , Subunidades de Proteína/química , Subunidades de Proteína/metabolismo , Proteínas del Envoltorio Viral/metabolismo , Internalización del Virus
11.
J Virol ; 88(16): 9287-96, 2014 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-24899200

RESUMEN

UNLABELLED: The structure of adenovirus outer capsid was revealed recently at 3- to 4-Å resolution (V. Reddy, S. Natchiar, P. Stewart, and G. Nemerow, Science 329:1071-1075, 2010, http://dx.doi.org/10.1126/science.1187292); however, precise details on the function and biochemical and structural features for the inner core still are lacking. Protein V is one the most important components of the adenovirus core, as it links the outer capsid via association with protein VI with the inner DNA core. Protein V is a highly basic protein that strongly binds to DNA in a nonspecific manner. We report the expression of a soluble protein V that exists in monomer-dimer equilibrium. Using reversible cross-linking affinity purification in combination with mass spectrometry, we found that protein V contains multiple DNA binding sites. The binding sites from protein V mediate heat-stable nucleic acid associations, with some of the binding sites possibly masked in the virus by other core proteins. We also demonstrate direct interaction between soluble proteins V and VI, thereby revealing the bridging of the inner DNA core with the outer capsid proteins. These findings are consistent with a model of nucleosome-like structures proposed for the adenovirus core and encapsidated DNA. They also suggest an additional role for protein V in linking the inner nucleic acid core with protein VI on the inner capsid shell. IMPORTANCE: Scant knowledge exists of how the inner core of adenovirus containing its double-stranded DNA (dsDNA) genome and associated proteins is organized. Here, we report a purification scheme for a recombinant form of protein V that allowed analysis of its interactions with the nucleic acid core region. We demonstrate that protein V exhibits stable associations with dsDNA due to the presence of multiple nucleic acid binding sites identified both in the isolated recombinant protein and in virus particles. As protein V also binds to the membrane lytic protein VI molecules, this core protein may serve as a bridge from the inner dsDNA core to the inner capsid shell.


Asunto(s)
Adenoviridae/genética , Adenoviridae/metabolismo , ADN Viral/metabolismo , Proteínas del Núcleo Viral/genética , Proteínas del Núcleo Viral/metabolismo , Proteínas Virales/genética , Proteínas Virales/metabolismo , Secuencia de Aminoácidos , Secuencia de Bases , Sitios de Unión/genética , Cápside/metabolismo , Proteínas de la Cápside/genética , Proteínas de la Cápside/metabolismo , ADN Viral/genética , Datos de Secuencia Molecular , Unión Proteica/genética , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Virión/genética , Virión/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA