Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 163
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Cell ; 2024 Sep 12.
Artículo en Inglés | MEDLINE | ID: mdl-39293445

RESUMEN

Filoviruses, including the Ebola and Marburg viruses, cause hemorrhagic fevers with up to 90% lethality. The viral nucleocapsid is assembled by polymerization of the nucleoprotein (NP) along the viral genome, together with the viral proteins VP24 and VP35. We employed cryo-electron tomography of cells transfected with viral proteins and infected with model Ebola virus to illuminate assembly intermediates, as well as a 9 Å map of the complete intracellular assembly. This structure reveals a previously unresolved third and outer layer of NP complexed with VP35. The intrinsically disordered region, together with the C-terminal domain of this outer layer of NP, provides the constant width between intracellular nucleocapsid bundles and likely functions as a flexible tether to the viral matrix protein in the virion. A comparison of intracellular nucleocapsids with prior in-virion nucleocapsid structures reveals that the nucleocapsid further condenses vertically in the virion. The interfaces responsible for nucleocapsid assembly are highly conserved and offer targets for broadly effective antivirals.

2.
Cell ; 185(6): 995-1007.e18, 2022 03 17.
Artículo en Inglés | MEDLINE | ID: mdl-35303429

RESUMEN

Several ebolaviruses cause outbreaks of severe disease. Vaccines and monoclonal antibody cocktails are available to treat Ebola virus (EBOV) infections, but not Sudan virus (SUDV) or other ebolaviruses. Current cocktails contain antibodies that cross-react with the secreted soluble glycoprotein (sGP) that absorbs virus-neutralizing antibodies. By sorting memory B cells from EBOV infection survivors, we isolated two broadly reactive anti-GP monoclonal antibodies, 1C3 and 1C11, that potently neutralize, protect rodents from disease, and lack sGP cross-reactivity. Both antibodies recognize quaternary epitopes in trimeric ebolavirus GP. 1C11 bridges adjacent protomers via the fusion loop. 1C3 has a tripartite epitope in the center of the trimer apex. One 1C3 antigen-binding fragment anchors simultaneously to the three receptor-binding sites in the GP trimer, and separate 1C3 paratope regions interact differently with identical residues on the three protomers. A cocktail of both antibodies completely protected nonhuman primates from EBOV and SUDV infections, indicating their potential clinical value.


Asunto(s)
Anticuerpos Neutralizantes , Anticuerpos Antivirales , Ebolavirus , Fiebre Hemorrágica Ebola , Animales , Epítopos , Glicoproteínas/química , Subunidades de Proteína
3.
Cell ; 183(1): 1-3, 2020 10 01.
Artículo en Inglés | MEDLINE | ID: mdl-33007260

RESUMEN

The pandemic has impacted every scientist differently. Many negative impacts are frequently discussed. Here we highlight unexpected positives that we have found and hope will persist: improved access to experts; deeper and broader human engagement among colleagues, collaborators, and competitors; and significant democratization of research.


Asunto(s)
COVID-19/psicología , Pandemias/ética , Humanos , Optimismo/psicología , SARS-CoV-2/patogenicidad
4.
Cell ; 182(3): 786-786.e1, 2020 08 06.
Artículo en Inglés | MEDLINE | ID: mdl-32763187

RESUMEN

In order to initiate successful infection, viruses have to transmit and deliver their genome from one host cell or organism to another. To achieve this, enveloped viruses must first fuse their membrane with those of the target host cell. Here, we describe the sequence of events leading to the entry of representative enveloped viruses, highlighting the strategies they use to gain access to the host cell cytosol.


Asunto(s)
Endocitosis , Endosomas/virología , Fusión de Membrana , Internalización del Virus , Virus/metabolismo , Animales , Endosomas/metabolismo , Humanos , Virosis/enzimología , Virosis/metabolismo , Virus/genética
5.
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
6.
Cell ; 174(4): 938-952.e13, 2018 08 09.
Artículo en Inglés | MEDLINE | ID: mdl-30096313

RESUMEN

Antibodies are promising post-exposure therapies against emerging viruses, but which antibody features and in vitro assays best forecast protection are unclear. Our international consortium systematically evaluated antibodies against Ebola virus (EBOV) using multidisciplinary assays. For each antibody, we evaluated epitopes recognized on the viral surface glycoprotein (GP) and secreted glycoprotein (sGP), readouts of multiple neutralization assays, fraction of virions left un-neutralized, glycan structures, phagocytic and natural killer cell functions elicited, and in vivo protection in a mouse challenge model. Neutralization and induction of multiple immune effector functions (IEFs) correlated most strongly with protection. Neutralization predominantly occurred via epitopes maintained on endosomally cleaved GP, whereas maximal IEF mapped to epitopes farthest from the viral membrane. Unexpectedly, sGP cross-reactivity did not significantly influence in vivo protection. This comprehensive dataset provides a rubric to evaluate novel antibodies and vaccine responses and a roadmap for therapeutic development for EBOV and related viruses.


Asunto(s)
Anticuerpos Monoclonales/inmunología , Anticuerpos Monoclonales/aislamiento & purificación , Ebolavirus/inmunología , Epítopos/inmunología , Fiebre Hemorrágica Ebola/prevención & control , Glicoproteínas de Membrana/inmunología , Animales , Anticuerpos Monoclonales/administración & dosificación , Femenino , Fiebre Hemorrágica Ebola/inmunología , Fiebre Hemorrágica Ebola/virología , Inmunización , Ratones , Ratones Endogámicos BALB C , Resultado del Tratamiento
7.
Immunity ; 55(5): 738-748, 2022 05 10.
Artículo en Inglés | MEDLINE | ID: mdl-35545026

RESUMEN

The brutal toll of another viral pandemic can be blunted by investing now in research that uncovers mechanisms of broad-based immunity so we may have vaccines and therapeutics at the ready. We do not know exactly what pathogen may trigger the next wave or next pandemic. We do know, however, that the human immune system must respond and must be bolstered with effective vaccines and other therapeutics to preserve lives and livelihoods. These countermeasures must focus on features conserved among families of pathogens in order to be responsive against something yet to emerge. Here, we focus on immunological approaches to mitigate the impact of the next emerging virus pandemic by developing vaccines that elicit both broadly protective antibodies and T cells. Identifying human immune mechanisms of broad protection against virus families with pandemic potential will be our best defense for humanity in the future.


Asunto(s)
COVID-19 , Vacunas , Anticuerpos Antivirales , Humanos , Pandemias/prevención & control , SARS-CoV-2 , Linfocitos T
8.
Cell ; 164(3): 392-405, 2016 Jan 28.
Artículo en Inglés | MEDLINE | ID: mdl-26806128

RESUMEN

Recent studies have suggested that antibody-mediated protection against the Ebolaviruses may be achievable, but little is known about whether or not antibodies can confer cross-reactive protection against viruses belonging to diverse Ebolavirus species, such as Ebola virus (EBOV), Sudan virus (SUDV), and Bundibugyo virus (BDBV). We isolated a large panel of human monoclonal antibodies (mAbs) against BDBV glycoprotein (GP) using peripheral blood B cells from survivors of the 2007 BDBV outbreak in Uganda. We determined that a large proportion of mAbs with potent neutralizing activity against BDBV bind to the glycan cap and recognize diverse epitopes within this major antigenic site. We identified several glycan cap-specific mAbs that neutralized multiple ebolaviruses, including SUDV, and a cross-reactive mAb that completely protected guinea pigs from the lethal challenge with heterologous EBOV. Our results provide a roadmap to develop a single antibody-based treatment effective against multiple Ebolavirus infections.


Asunto(s)
Anticuerpos Monoclonales/inmunología , Anticuerpos Neutralizantes/inmunología , Ebolavirus/inmunología , Fiebre Hemorrágica Ebola/inmunología , Sobrevivientes , Animales , Reacciones Cruzadas , Modelos Animales de Enfermedad , Mapeo Epitopo , Cobayas , Humanos , Ratones , Ratones Endogámicos BALB C , Microscopía Electrónica , Modelos Moleculares , Mutagénesis , Uganda
9.
Nat Immunol ; 19(11): 1169-1178, 2018 11.
Artículo en Inglés | MEDLINE | ID: mdl-30333617

RESUMEN

Recent Ebola virus disease epidemics have highlighted the need for effective vaccines and therapeutics to prevent future outbreaks. Antibodies are clearly critical for control of this deadly disease; however, the specific mechanisms of action of protective antibodies have yet to be defined. In this Perspective we discuss the antibody features that correlate with in vivo protection during infection with Ebola virus, based on the results of a systematic and comprehensive study of antibodies directed against this virus. Although neutralization activity mediated by the Fab domains of the antibody is strongly correlated with protection, recruitment of immune effector functions by the Fc domain has also emerged as a complementary, and sometimes alternative, route to protection. For a subset of antibodies, Fc-mediated clearance and killing of infected cells seems to be the main driver of protection after exposure and mirrors observations in vaccination studies. Continued analysis of antibodies that achieve protection partially or wholly through Fc-mediated functions, the precise functions required, the intersection with specificity and the importance of these functions in different animal models is needed to identify and begin to capitalize on Fc-mediated protection in vaccines and therapeutics alike.


Asunto(s)
Anticuerpos Antivirales/inmunología , Ebolavirus/inmunología , Fiebre Hemorrágica Ebola/inmunología , Animales , Anticuerpos Neutralizantes/inmunología , Humanos
10.
Immunity ; 54(4): 815-828.e5, 2021 04 13.
Artículo en Inglés | MEDLINE | ID: mdl-33852832

RESUMEN

Protective Ebola virus (EBOV) antibodies have neutralizing activity and induction of antibody constant domain (Fc)-mediated innate immune effector functions. Efforts to enhance Fc effector functionality often focus on maximizing antibody-dependent cellular cytotoxicity, yet distinct combinations of functions could be critical for antibody-mediated protection. As neutralizing antibodies have been cloned from EBOV disease survivors, we sought to identify survivor Fc effector profiles to help guide Fc optimization strategies. Survivors developed a range of functional antibody responses, and we therefore applied a rapid, high-throughput Fc engineering platform to define the most protective profiles. We generated a library of Fc variants with identical antigen-binding fragments (Fabs) from an EBOV neutralizing antibody. Fc variants with antibody-mediated complement deposition and moderate natural killer (NK) cell activity demonstrated complete protective activity in a stringent in vivo mouse model. Our findings highlight the importance of specific effector functions in antibody-mediated protection, and the experimental platform presents a generalizable resource for identifying correlates of immunity to guide therapeutic antibody design.


Asunto(s)
Ebolavirus/inmunología , Fiebre Hemorrágica Ebola/inmunología , Fragmentos Fab de Inmunoglobulinas/inmunología , Fragmentos Fc de Inmunoglobulinas/inmunología , Animales , Anticuerpos Neutralizantes/inmunología , Anticuerpos Antivirales/inmunología , Formación de Anticuerpos/inmunología , Citotoxicidad Celular Dependiente de Anticuerpos/inmunología , Femenino , Células HEK293 , Fiebre Hemorrágica Ebola/virología , Humanos , Inmunoglobulina G/inmunología , Ratones Endogámicos BALB C , Receptores Fc/inmunología
11.
Cell ; 163(5): 1053-1054, 2015 Nov 19.
Artículo en Inglés | MEDLINE | ID: mdl-26590413

RESUMEN

Chikungunya virus recently caused large outbreaks world-wide. In this issue of Cell, Fox et al. describe several potently neutralizing antibodies against multiple alphaviruses. The structure of the virus in complex with one of the antibodies reveals the antibody-induced rearrangement and crosslinking of the viral surface proteins that result in neutralization.


Asunto(s)
Infecciones por Alphavirus/inmunología , Alphavirus/inmunología , Anticuerpos Monoclonales/inmunología , Anticuerpos Neutralizantes/inmunología , Anticuerpos Antivirales/inmunología , Epítopos , Proteínas del Envoltorio Viral/inmunología , Animales , Humanos
12.
Cell ; 160(5): 904-912, 2015 Feb 26.
Artículo en Inglés | MEDLINE | ID: mdl-25723165

RESUMEN

The filoviruses, including Marburg and Ebola, express a single glycoprotein on their surface, termed GP, which is responsible for attachment and entry of target cells. Filovirus GPs differ by up to 70% in protein sequence, and no antibodies are yet described that cross-react among them. Here, we present the 3.6 Å crystal structure of Marburg virus GP in complex with a cross-reactive antibody from a human survivor, and a lower resolution structure of the antibody bound to Ebola virus GP. The antibody, MR78, recognizes a GP1 epitope conserved across the filovirus family, which likely represents the binding site of their NPC1 receptor. Indeed, MR78 blocks binding of the essential NPC1 domain C. These structures and additional small-angle X-ray scattering of mucin-containing MARV and EBOV GPs suggest why such antibodies were not previously elicited in studies of Ebola virus, and provide critical templates for development of immunotherapeutics and inhibitors of entry.


Asunto(s)
Anticuerpos Neutralizantes/química , Anticuerpos Neutralizantes/inmunología , Marburgvirus/química , Proteínas del Envoltorio Viral/química , Secuencia de Aminoácidos , Animales , Anticuerpos Monoclonales/química , Anticuerpos Monoclonales/metabolismo , Anticuerpos Neutralizantes/aislamiento & purificación , Anticuerpos Neutralizantes/metabolismo , Anticuerpos Antivirales/química , Anticuerpos Antivirales/inmunología , Anticuerpos Antivirales/metabolismo , Complejo Antígeno-Anticuerpo/química , Línea Celular , Reacciones Cruzadas , Cristalografía por Rayos X , Drosophila , Ebolavirus/química , Humanos , Fragmentos Fab de Inmunoglobulinas/química , Fragmentos Fab de Inmunoglobulinas/metabolismo , Enfermedad del Virus de Marburg/inmunología , Marburgvirus/genética , Marburgvirus/inmunología , Modelos Moleculares , Datos de Secuencia Molecular , Mucinas/química , Alineación de Secuencia , Proteínas del Envoltorio Viral/metabolismo
13.
Cell ; 160(5): 893-903, 2015 Feb 26.
Artículo en Inglés | MEDLINE | ID: mdl-25723164

RESUMEN

The mechanisms by which neutralizing antibodies inhibit Marburg virus (MARV) are not known. We isolated a panel of neutralizing antibodies from a human MARV survivor that bind to MARV glycoprotein (GP) and compete for binding to a single major antigenic site. Remarkably, several of the antibodies also bind to Ebola virus (EBOV) GP. Single-particle EM structures of antibody-GP complexes reveal that all of the neutralizing antibodies bind to MARV GP at or near the predicted region of the receptor-binding site. The presence of the glycan cap or mucin-like domain blocks binding of neutralizing antibodies to EBOV GP, but not to MARV GP. The data suggest that MARV-neutralizing antibodies inhibit virus by binding to infectious virions at the exposed MARV receptor-binding site, revealing a mechanism of filovirus inhibition.


Asunto(s)
Anticuerpos Neutralizantes/química , Anticuerpos Neutralizantes/inmunología , Complejo Antígeno-Anticuerpo/ultraestructura , Enfermedad del Virus de Marburg/inmunología , Marburgvirus/química , Proteínas del Envoltorio Viral/química , Adulto , Animales , Anticuerpos Monoclonales/química , Anticuerpos Monoclonales/metabolismo , Anticuerpos Neutralizantes/aislamiento & purificación , Anticuerpos Neutralizantes/metabolismo , Anticuerpos Antivirales/química , Anticuerpos Antivirales/inmunología , Anticuerpos Antivirales/metabolismo , Linfocitos B/inmunología , Femenino , Humanos , Fragmentos Fab de Inmunoglobulinas/química , Fragmentos Fab de Inmunoglobulinas/metabolismo , Marburgvirus/genética , Marburgvirus/inmunología , Modelos Moleculares , Mutación , Estructura Terciaria de Proteína , Proteínas del Envoltorio Viral/metabolismo
14.
Nature ; 632(8025): 630-636, 2024 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-39085605

RESUMEN

The upper airway is an important site of infection, but immune memory in the human upper airway is poorly understood, with implications for COVID-19 and many other human diseases1-4. Here we demonstrate that nasal and nasopharyngeal swabs can be used to obtain insights into these challenging problems, and define distinct immune cell populations, including antigen-specific memory B cells and T cells, in two adjacent anatomical sites in the upper airway. Upper airway immune cell populations seemed stable over time in healthy adults undergoing monthly swabs for more than 1 year, and prominent tissue resident memory T (TRM) cell and B (BRM) cell populations were defined. Unexpectedly, germinal centre cells were identified consistently in many nasopharyngeal swabs. In subjects with SARS-CoV-2 breakthrough infections, local virus-specific BRM cells, plasma cells and germinal centre B cells were identified, with evidence of local priming and an enrichment of IgA+ memory B cells in upper airway compartments compared with blood. Local plasma cell populations were identified with transcriptional profiles of longevity. Local virus-specific memory CD4+ TRM cells and CD8+ TRM cells were identified, with diverse additional virus-specific T cells. Age-dependent upper airway immunological shifts were observed. These findings provide new understanding of immune memory at a principal mucosal barrier tissue in humans.


Asunto(s)
Memoria Inmunológica , Células B de Memoria , Células T de Memoria , Mucosa Nasal , Nasofaringe , SARS-CoV-2 , Adulto , Humanos , Linfocitos T CD4-Positivos/inmunología , Linfocitos T CD4-Positivos/citología , Linfocitos T CD8-positivos/inmunología , Linfocitos T CD8-positivos/citología , COVID-19/inmunología , COVID-19/virología , Centro Germinal/inmunología , Centro Germinal/citología , Inmunoglobulina A/inmunología , Memoria Inmunológica/inmunología , Células B de Memoria/inmunología , Células T de Memoria/inmunología , Mucosa Nasal/inmunología , Mucosa Nasal/virología , Nasofaringe/virología , Nasofaringe/inmunología , Células Plasmáticas/inmunología , Células Plasmáticas/citología , SARS-CoV-2/inmunología
15.
Immunity ; 52(2): 388-403.e12, 2020 02 18.
Artículo en Inglés | MEDLINE | ID: mdl-32023489

RESUMEN

Structural principles underlying the composition of protective antiviral monoclonal antibody (mAb) cocktails are poorly defined. Here, we exploited antibody cooperativity to develop a therapeutic mAb cocktail against Ebola virus. We systematically analyzed the antibody repertoire in human survivors and identified a pair of potently neutralizing mAbs that cooperatively bound to the ebolavirus glycoprotein (GP). High-resolution structures revealed that in a two-antibody cocktail, molecular mimicry was a major feature of mAb-GP interactions. Broadly neutralizing mAb rEBOV-520 targeted a conserved epitope on the GP base region. mAb rEBOV-548 bound to a glycan cap epitope, possessed neutralizing and Fc-mediated effector function activities, and potentiated neutralization by rEBOV-520. Remodeling of the glycan cap structures by the cocktail enabled enhanced GP binding and virus neutralization. The cocktail demonstrated resistance to virus escape and protected non-human primates (NHPs) against Ebola virus disease. These data illuminate structural principles of antibody cooperativity with implications for development of antiviral immunotherapeutics.


Asunto(s)
Anticuerpos Monoclonales/inmunología , Anticuerpos Neutralizantes/inmunología , Anticuerpos Antivirales/inmunología , Ebolavirus/inmunología , Glicoproteínas/inmunología , Fiebre Hemorrágica Ebola/inmunología , Animales , Anticuerpos Monoclonales/uso terapéutico , Anticuerpos Neutralizantes/uso terapéutico , Anticuerpos Antivirales/uso terapéutico , Línea Celular , Modelos Animales de Enfermedad , Quimioterapia Combinada , Epítopos , Femenino , Glicoproteínas/química , Fiebre Hemorrágica Ebola/prevención & control , Humanos , Fragmentos Fab de Inmunoglobulinas/inmunología , Macaca mulatta , Masculino , Ratones , Ratones Endogámicos BALB C , Imitación Molecular , Conformación Proteica
16.
Cell ; 154(4): 763-74, 2013 Aug 15.
Artículo en Inglés | MEDLINE | ID: mdl-23953110

RESUMEN

Proteins, particularly viral proteins, can be multifunctional, but the mechanisms behind multifunctionality are not fully understood. Here, we illustrate through multiple crystal structures, biochemistry, and cellular microscopy that VP40 rearranges into different structures, each with a distinct function required for the ebolavirus life cycle. A butterfly-shaped VP40 dimer traffics to the cellular membrane. Once there, electrostatic interactions trigger rearrangement of the polypeptide into a linear hexamer. These hexamers construct a multilayered, filamentous matrix structure that is critical for budding and resembles tomograms of authentic virions. A third structure of VP40, formed by a different rearrangement, is not involved in virus assembly but instead uniquely binds RNA to regulate viral transcription inside infected cells. These results provide a functional model for ebolavirus matrix assembly and the other roles of VP40 in the virus life cycle and demonstrate how a single wild-type, unmodified polypeptide can assemble into different structures for different functions.


Asunto(s)
Ebolavirus/metabolismo , Proteínas de la Matriz Viral/química , Proteínas de la Matriz Viral/metabolismo , Cristalografía por Rayos X , Dimerización , Ebolavirus/química , Ebolavirus/clasificación , Ebolavirus/genética , Modelos Moleculares , Mutagénesis , Mutación Puntual , Proteínas de la Matriz Viral/genética , Ensamble de Virus , Liberación del Virus
17.
Immunity ; 49(2): 363-374.e10, 2018 08 21.
Artículo en Inglés | MEDLINE | ID: mdl-30029854

RESUMEN

Ebolaviruses cause severe disease in humans, and identification of monoclonal antibodies (mAbs) that are effective against multiple ebolaviruses are important for therapeutics development. Here we describe a distinct class of broadly neutralizing human mAbs with protective capacity against three ebolaviruses infectious for humans: Ebola (EBOV), Sudan (SUDV), and Bundibugyo (BDBV) viruses. We isolated mAbs from human survivors of ebolavirus disease and identified a potent mAb, EBOV-520, which bound to an epitope in the glycoprotein (GP) base region. EBOV-520 efficiently neutralized EBOV, BDBV, and SUDV and also showed protective capacity in relevant animal models of these infections. EBOV-520 mediated protection principally by direct virus neutralization and exhibited multifunctional properties. This study identified a potent naturally occurring mAb and defined key features of the human antibody response that may contribute to broad protection. This multifunctional mAb and related clones are promising candidates for development as broadly protective pan-ebolavirus therapeutic molecules.


Asunto(s)
Anticuerpos Monoclonales/inmunología , Anticuerpos Monoclonales/farmacología , Anticuerpos Neutralizantes/inmunología , Anticuerpos Neutralizantes/farmacología , Anticuerpos Antivirales/inmunología , Anticuerpos Antivirales/farmacología , Ebolavirus/inmunología , Glicoproteínas/inmunología , Fiebre Hemorrágica Ebola/inmunología , Células 3T3 , Adulto , Animales , Células CHO , Línea Celular , Chlorocebus aethiops , Cricetulus , Modelos Animales de Enfermedad , Drosophila , Femenino , Hurones , Cobayas , Fiebre Hemorrágica Ebola/prevención & control , Fiebre Hemorrágica Ebola/virología , Humanos , Inmunoglobulina G/inmunología , Células Jurkat , Masculino , Ratones , Ratones Endogámicos BALB C , Ratones Noqueados , Células THP-1 , Células Vero
18.
Proc Natl Acad Sci U S A ; 119(10): e2119676119, 2022 03 08.
Artículo en Inglés | MEDLINE | ID: mdl-35235462

RESUMEN

Lymphocytic choriomeningitis virus (LCMV) is a rodent-borne zoonotic arenavirus that causes congenital abnormalities and can be fatal for transplant recipients. Using a genome-wide loss-of-function screen, we identify host factors required for LCMV entry into cells. We identify the lysosomal mucin CD164, glycosylation factors, the heparan sulfate biosynthesis machinery, and the known receptor alpha-dystroglycan (α-DG). Biochemical analysis revealed that the LCMV glycoprotein binds CD164 at acidic pH and requires a sialylated glycan at residue N104. We demonstrate that LCMV entry proceeds by the virus switching binding from heparan sulfate or α-DG at the plasma membrane to CD164 prior to membrane fusion, thus identifying additional potential targets for therapeutic intervention.


Asunto(s)
Virus de la Coriomeningitis Linfocítica/fisiología , Internalización del Virus , Células A549 , Sistemas CRISPR-Cas , Endolina/fisiología , Edición Génica , Células HEK293 , Células HeLa , Interacciones Huésped-Patógeno , Humanos , Concentración de Iones de Hidrógeno , Virus de la Coriomeningitis Linfocítica/patogenicidad , Fusión de Membrana , Factores de Virulencia
19.
Proc Natl Acad Sci U S A ; 119(30): e2201208119, 2022 07 26.
Artículo en Inglés | MEDLINE | ID: mdl-35858434

RESUMEN

Completion of the Lassa virus (LASV) life cycle critically depends on the activities of the virally encoded, RNA-dependent RNA polymerase in replication and transcription of the viral RNA genome in the cytoplasm of infected cells. The contribution of cellular proteins to these processes remains unclear. Here, we applied proximity proteomics to define the interactome of LASV polymerase in cells under conditions that recreate LASV RNA synthesis. We engineered a LASV polymerase-biotin ligase (TurboID) fusion protein that retained polymerase activity and successfully biotinylated the proximal proteome, which allowed the identification of 42 high-confidence LASV polymerase interactors. We subsequently performed a small interfering RNA (siRNA) screen to identify those interactors that have functional roles in authentic LASV infection. As proof of principle, we characterized eukaryotic peptide chain release factor subunit 3a (eRF3a/GSPT1), which we found to be a proviral factor that physically associates with LASV polymerase. Targeted degradation of GSPT1 by a small-molecule drug candidate, CC-90009, resulted in strong inhibition of LASV infection in cultured cells. Our work demonstrates the feasibility of using proximity proteomics to illuminate and characterize yet-to-be-defined host-pathogen interactome, which can reveal new biology and uncover novel targets for the development of antivirals against highly pathogenic RNA viruses.


Asunto(s)
Acetamidas , Antivirales , Isoindoles , Virus Lassa , Factores de Terminación de Péptidos , Piperidonas , ARN Polimerasa Dependiente del ARN , Proteínas Virales , Acetamidas/farmacología , Acetamidas/uso terapéutico , Antivirales/farmacología , Antivirales/uso terapéutico , Línea Celular Tumoral , Humanos , Isoindoles/farmacología , Isoindoles/uso terapéutico , Fiebre de Lassa/tratamiento farmacológico , Virus Lassa/efectos de los fármacos , Factores de Terminación de Péptidos/metabolismo , Piperidonas/metabolismo , Piperidonas/farmacología , Piperidonas/uso terapéutico , Mapas de Interacción de Proteínas/efectos de los fármacos , Proteolisis/efectos de los fármacos , Proteoma , Proteómica , ARN Polimerasa Dependiente del ARN/metabolismo , Proteínas Virales/metabolismo
20.
J Infect Dis ; 228(Suppl 6): S359-S375, 2023 10 18.
Artículo en Inglés | MEDLINE | ID: mdl-37849403

RESUMEN

Lassa virus (LASV), Junin virus (JUNV), and several other members of the Arenaviridae family are capable of zoonotic transfer to humans and induction of severe viral hemorrhagic fevers. Despite the importance of arenaviruses as potential pandemic pathogens, numerous gaps exist in scientific knowledge pertaining to this diverse family, including gaps in understanding replication, immunosuppression, receptor usage, and elicitation of neutralizing antibody responses, that in turn complicates development of medical countermeasures. A further challenge to the development of medical countermeasures for arenaviruses is the requirement for use of animal models at high levels of biocontainment, where each model has distinct advantages and limitations depending on, availability of space, animals species-specific reagents, and most importantly the ability of the model to faithfully recapitulate human disease. Designation of LASV and JUNV as prototype pathogens can facilitate progress in addressing the public health challenges posed by members of this important virus family.


Asunto(s)
Arenaviridae , Virus Junin , Animales , Humanos , Replicación Viral , Virus Junin/fisiología , Virus Lassa , Modelos Animales
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA