Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Resultados 1 - 20 de 89
Filtrar
1.
Cell ; 187(2): 360-374.e19, 2024 01 18.
Artículo en Inglés | MEDLINE | ID: mdl-38176410

RESUMEN

The very-low-density lipoprotein receptor (VLDLR) comprises eight LDLR type A (LA) domains and supports entry of distantly related alphaviruses, including Eastern equine encephalitis virus (EEEV) and Semliki Forest virus (SFV). Here, by resolving multiple cryo-electron microscopy structures of EEEV-VLDLR complexes and performing mutagenesis and functional studies, we show that EEEV uses multiple sites (E1/E2 cleft and E2 A domain) to engage more than one LA domain simultaneously. However, no single LA domain is necessary or sufficient to support efficient EEEV infection. Whereas all EEEV strains show conservation of two VLDLR-binding sites, the EEEV PE-6 strain and a few other EEE complex members feature a single amino acid substitution that enables binding of LA domains to an additional site on the E2 B domain. These structural and functional analyses informed the design of a minimal VLDLR decoy receptor that neutralizes EEEV infection and protects mice from lethal challenge.


Asunto(s)
Microscopía por Crioelectrón , Virus de la Encefalitis Equina del Este , Encefalomielitis Equina , Receptores de LDL , Animales , Ratones , Alphavirus/fisiología , Virus de la Encefalitis Equina del Este/fisiología , Virus de la Encefalitis Equina del Este/ultraestructura , Encefalomielitis Equina/metabolismo , Caballos , Unión Proteica , Receptores de LDL/ultraestructura
2.
Cell ; 186(22): 4818-4833.e25, 2023 10 26.
Artículo en Inglés | MEDLINE | ID: mdl-37804831

RESUMEN

MXRA8 is a receptor for chikungunya (CHIKV) and other arthritogenic alphaviruses with mammalian hosts. However, mammalian MXRA8 does not bind to alphaviruses that infect humans and have avian reservoirs. Here, we show that avian, but not mammalian, MXRA8 can act as a receptor for Sindbis, western equine encephalitis (WEEV), and related alphaviruses with avian reservoirs. Structural analysis of duck MXRA8 complexed with WEEV reveals an inverted binding mode compared with mammalian MXRA8 bound to CHIKV. Whereas both domains of mammalian MXRA8 bind CHIKV E1 and E2, only domain 1 of avian MXRA8 engages WEEV E1, and no appreciable contacts are made with WEEV E2. Using these results, we generated a chimeric avian-mammalian MXRA8 decoy-receptor that neutralizes infection of multiple alphaviruses from distinct antigenic groups in vitro and in vivo. Thus, different alphaviruses can bind MXRA8 encoded by different vertebrate classes with distinct engagement modes, which enables development of broad-spectrum inhibitors.


Asunto(s)
Alphavirus , Animales , Humanos , Fiebre Chikungunya , Virus Chikungunya/química , Mamíferos , Receptores Virales/metabolismo
3.
Cell ; 184(17): 4414-4429.e19, 2021 08 19.
Artículo en Inglés | MEDLINE | ID: mdl-34416146

RESUMEN

Alphaviruses are emerging, mosquito-transmitted pathogens that cause musculoskeletal and neurological disease in humans. Although neutralizing antibodies that inhibit individual alphaviruses have been described, broadly reactive antibodies that protect against both arthritogenic and encephalitic alphaviruses have not been reported. Here, we identify DC2.112 and DC2.315, two pan-protective yet poorly neutralizing human monoclonal antibodies (mAbs) that avidly bind to viral antigen on the surface of cells infected with arthritogenic and encephalitic alphaviruses. These mAbs engage a conserved epitope in domain II of the E1 protein proximal to and within the fusion peptide. Treatment with DC2.112 or DC2.315 protects mice against infection by both arthritogenic (chikungunya and Mayaro) and encephalitic (Venezuelan, Eastern, and Western equine encephalitis) alphaviruses through multiple mechanisms, including inhibition of viral egress and monocyte-dependent Fc effector functions. These findings define a conserved epitope recognized by weakly neutralizing yet protective antibodies that could be targeted for pan-alphavirus immunotherapy and vaccine design.


Asunto(s)
Alphavirus/inmunología , Anticuerpos Antivirales/inmunología , Secuencia Conservada/inmunología , Epítopos/inmunología , Proteínas Virales/inmunología , Infecciones por Alphavirus/inmunología , Infecciones por Alphavirus/virología , Secuencia de Aminoácidos , Animales , Anticuerpos Monoclonales/inmunología , Fiebre Chikungunya/inmunología , Fiebre Chikungunya/virología , Virus Chikungunya/inmunología , Chlorocebus aethiops , Mapeo Epitopo , Epítopos/química , Humanos , Masculino , Ratones Endogámicos C57BL , Modelos Biológicos , Monocitos/metabolismo , Células Vero , Proteínas Virales/química , Liberación del Virus
4.
Cell ; 184(17): 4430-4446.e22, 2021 08 19.
Artículo en Inglés | MEDLINE | ID: mdl-34416147

RESUMEN

Alphaviruses cause severe arthritogenic or encephalitic disease. The E1 structural glycoprotein is highly conserved in these viruses and mediates viral fusion with host cells. However, the role of antibody responses to the E1 protein in immunity is poorly understood. We isolated E1-specific human monoclonal antibodies (mAbs) with diverse patterns of recognition for alphaviruses (ranging from Eastern equine encephalitis virus [EEEV]-specific to alphavirus cross-reactive) from survivors of natural EEEV infection. Antibody binding patterns and epitope mapping experiments identified differences in E1 reactivity based on exposure of epitopes on the glycoprotein through pH-dependent mechanisms or presentation on the cell surface prior to virus egress. Therapeutic efficacy in vivo of these mAbs corresponded with potency of virus egress inhibition in vitro and did not require Fc-mediated effector functions for treatment against subcutaneous EEEV challenge. These studies reveal the molecular basis for broad and protective antibody responses to alphavirus E1 proteins.


Asunto(s)
Alphavirus/inmunología , Anticuerpos Antivirales/inmunología , Reacciones Cruzadas/inmunología , Proteínas Virales/inmunología , Liberación del Virus/fisiología , Animales , Anticuerpos Monoclonales/inmunología , Anticuerpos Monoclonales/aislamiento & purificación , Anticuerpos Neutralizantes/inmunología , Antígenos Virales/inmunología , Línea Celular , Virus Chikungunya/inmunología , Virus de la Encefalitis Equina del Este/inmunología , Encefalomielitis Equina/inmunología , Encefalomielitis Equina/virología , Mapeo Epitopo , Femenino , Caballos , Humanos , Concentración de Iones de Hidrógeno , Articulaciones/patología , Masculino , Ratones Endogámicos C57BL , Modelos Biológicos , Unión Proteica , ARN Viral/metabolismo , Receptores Fc/metabolismo , Temperatura , Virión/metabolismo , Internalización del Virus
5.
Cell ; 183(7): 1884-1900.e23, 2020 12 23.
Artículo en Inglés | MEDLINE | ID: mdl-33301709

RESUMEN

Eastern equine encephalitis virus (EEEV) is one of the most virulent viruses endemic to North America. No licensed vaccines or antiviral therapeutics are available to combat this infection, which has recently shown an increase in human cases. Here, we characterize human monoclonal antibodies (mAbs) isolated from a survivor of natural EEEV infection with potent (<20 pM) inhibitory activity of EEEV. Cryo-electron microscopy reconstructions of two highly neutralizing mAbs, EEEV-33 and EEEV-143, were solved in complex with chimeric Sindbis/EEEV virions to 7.2 Å and 8.3 Å, respectively. The mAbs recognize two distinct antigenic sites that are critical for inhibiting viral entry into cells. EEEV-33 and EEEV-143 protect against disease following stringent lethal aerosol challenge of mice with highly pathogenic EEEV. These studies provide insight into the molecular basis for the neutralizing human antibody response against EEEV and can facilitate development of vaccines and candidate antibody therapeutics.


Asunto(s)
Aerosoles/administración & dosificación , Anticuerpos Monoclonales/inmunología , Anticuerpos Antivirales/inmunología , Virus de la Encefalitis Equina del Este/inmunología , Encefalomielitis Equina/inmunología , Encefalomielitis Equina/prevención & control , Adulto , Animales , Anticuerpos Monoclonales/aislamiento & purificación , Anticuerpos Neutralizantes/inmunología , Antígenos Virales/inmunología , Microscopía por Crioelectrón , Modelos Animales de Enfermedad , Virus de la Encefalitis Equina del Este/ultraestructura , Encefalomielitis Equina/virología , Epítopos/química , Femenino , Glicoproteínas/inmunología , Humanos , Ratones , Modelos Moleculares , Mutagénesis/genética , Pruebas de Neutralización , Unión Proteica , Dominios Proteicos , Proteínas Recombinantes/inmunología , Virus Sindbis/inmunología , Virión/inmunología , Virión/ultraestructura , Internalización del Virus
6.
Nature ; 588(7837): 308-314, 2020 12.
Artículo en Inglés | MEDLINE | ID: mdl-33208938

RESUMEN

Venezuelan equine encephalitis virus (VEEV) is a neurotropic alphavirus transmitted by mosquitoes that causes encephalitis and death in humans1. VEEV is a biodefence concern because of its potential for aerosol spread and the current lack of sufficient countermeasures. The host factors that are required for VEEV entry and infection remain poorly characterized. Here, using a genome-wide CRISPR-Cas9-based screen, we identify low-density lipoprotein receptor class A domain-containing 3 (LDLRAD3)-a highly conserved yet poorly characterized member of the scavenger receptor superfamily-as a receptor for VEEV. Gene editing of mouse Ldlrad3 or human LDLRAD3 results in markedly reduced viral infection of neuronal cells, which is restored upon complementation with LDLRAD3. LDLRAD3 binds directly to VEEV particles and enhances virus attachment and internalization into host cells. Genetic studies indicate that domain 1 of LDLRAD3 (LDLRAD3(D1)) is necessary and sufficient to support infection by VEEV, and both anti-LDLRAD3 antibodies and an LDLRAD3(D1)-Fc fusion protein block VEEV infection in cell culture. The pathogenesis of VEEV infection is abrogated in mice with deletions in Ldlrad3, and administration of LDLRAD3(D1)-Fc abolishes disease caused by several subtypes of VEEV, including highly virulent strains. The development of a decoy-receptor fusion protein suggests a strategy for the prevention of severe VEEV infection and associated disease in humans.


Asunto(s)
Virus de la Encefalitis Equina Venezolana/metabolismo , Receptores de LDL/metabolismo , Receptores Virales/metabolismo , Animales , Sistemas CRISPR-Cas/genética , Línea Celular , Virus de la Encefalitis Equina Venezolana/patogenicidad , Encefalomielitis Equina Venezolana/metabolismo , Encefalomielitis Equina Venezolana/prevención & control , Encefalomielitis Equina Venezolana/virología , Femenino , Prueba de Complementación Genética , Humanos , Masculino , Ratones , Ratones Endogámicos C57BL , Unión Proteica , Receptores de LDL/deficiencia , Receptores de LDL/genética , Receptores Virales/genética , Acoplamiento Viral , Internalización del Virus
7.
Nature ; 586(7830): 509-515, 2020 10.
Artículo en Inglés | MEDLINE | ID: mdl-32967005

RESUMEN

Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) is the aetiological agent of coronavirus disease 2019 (COVID-19), an emerging respiratory infection caused by the introduction of a novel coronavirus into humans late in 2019 (first detected in Hubei province, China). As of 18 September 2020, SARS-CoV-2 has spread to 215 countries, has infected more than 30 million people and has caused more than 950,000 deaths. As humans do not have pre-existing immunity to SARS-CoV-2, there is an urgent need to develop therapeutic agents and vaccines to mitigate the current pandemic and to prevent the re-emergence of COVID-19. In February 2020, the World Health Organization (WHO) assembled an international panel to develop animal models for COVID-19 to accelerate the testing of vaccines and therapeutic agents. Here we summarize the findings to date and provides relevant information for preclinical testing of vaccine candidates and therapeutic agents for COVID-19.


Asunto(s)
Infecciones por Coronavirus/tratamiento farmacológico , Infecciones por Coronavirus/prevención & control , Modelos Animales de Enfermedad , Pandemias/prevención & control , Neumonía Viral/tratamiento farmacológico , Neumonía Viral/prevención & control , Animales , Betacoronavirus/efectos de los fármacos , Betacoronavirus/inmunología , COVID-19 , Vacunas contra la COVID-19 , Infecciones por Coronavirus/inmunología , Hurones/virología , Humanos , Mesocricetus/virología , Ratones , Neumonía Viral/inmunología , Primates/virología , SARS-CoV-2 , Vacunas Virales/inmunología
8.
Proc Natl Acad Sci U S A ; 119(30): e2114119119, 2022 07 26.
Artículo en Inglés | MEDLINE | ID: mdl-35867819

RESUMEN

Alphaviruses can cause severe human arthritis and encephalitis. During virus infection, structural changes of viral glycoproteins in the acidified endosome trigger virus-host membrane fusion for delivery of the capsid core and RNA genome into the cytosol to initiate virus translation and replication. However, mechanisms by which E1 and E2 glycoproteins rearrange in this process remain unknown. Here, we investigate prefusion cryoelectron microscopy (cryo-EM) structures of eastern equine encephalitis virus (EEEV) under acidic conditions. With models fitted into the low-pH cryo-EM maps, we suggest that E2 dissociates from E1, accompanied by a rotation (∼60°) of the E2-B domain (E2-B) to expose E1 fusion loops. Cryo-EM reconstructions of EEEV bound to a protective antibody at acidic and neutral pH suggest that stabilization of E2-B prevents dissociation of E2 from E1. These findings reveal conformational changes of the glycoprotein spikes in the acidified host endosome. Stabilization of E2-B may provide a strategy for antiviral agent development.


Asunto(s)
Virus de la Encefalitis Equina del Este , Proteínas del Envoltorio Viral , Antivirales/química , Antivirales/farmacología , Microscopía por Crioelectrón , Virus de la Encefalitis Equina del Este/química , Concentración de Iones de Hidrógeno , Conformación Proteica , Estabilidad Proteica/efectos de los fármacos , Proteínas del Envoltorio Viral/química
9.
J Neuroinflammation ; 21(1): 24, 2024 Jan 17.
Artículo en Inglés | MEDLINE | ID: mdl-38233868

RESUMEN

BACKGROUND: Venezuelan Equine Encephalitis virus (VEEV) may enter the central nervous system (CNS) within olfactory sensory neurons (OSN) that originate in the nasal cavity after intranasal exposure. While it is known that VEEV has evolved several mechanisms to inhibit type I interferon (IFN) signaling within infected cells, whether this inhibits virologic control during neuroinvasion along OSN has not been studied. METHODS: We utilized an established murine model of intranasal infection with VEEV and a repository of scRNAseq data from IFN-treated OSN to assess the cellular targets and IFN signaling responses after VEEV exposure. RESULTS: We found that immature OSN, which express higher levels of the VEEV receptor LDLRAD3 than mature OSN, are the first cells infected by VEEV. Despite rapid VEEV neuroinvasion after intranasal exposure, olfactory neuroepithelium (ONE) and olfactory bulb (OB) IFN responses, as assessed by evaluation of expression of interferon signaling genes (ISG), are delayed for up to 48 h during VEEV neuroinvasion, representing a potential therapeutic window. Indeed, a single intranasal dose of recombinant IFNα triggers early ISG expression in both the nasal cavity and OB. When administered at the time of or early after infection, IFNα treatment delayed onset of sequelae associated with encephalitis and extended survival by several days. VEEV replication after IFN treatment was also transiently suppressed in the ONE, which inhibited subsequent invasion into the CNS. CONCLUSIONS: Our results demonstrate a critical and promising first evaluation of intranasal IFNα for the treatment of human encephalitic alphavirus exposures.


Asunto(s)
Virus de la Encefalitis Equina Venezolana , Neuronas Receptoras Olfatorias , Humanos , Ratones , Animales , Virus de la Encefalitis Equina Venezolana/genética , Sistema Nervioso Central , Replicación Viral
10.
PLoS Pathog ; 18(6): e1009946, 2022 06.
Artículo en Inglés | MEDLINE | ID: mdl-35696423

RESUMEN

Venezuelan equine encephalitis virus (VEEV) is a positively-stranded RNA arbovirus of the genus Alphavirus that causes encephalitis in humans. Cynomolgus macaques are a relevant model of the human disease caused by VEEV and are useful in exploring pathogenic mechanisms and the host response to VEEV infection. Macaques were exposed to small-particle aerosols containing virus derived from an infectious clone of VEEV strain INH-9813, a subtype IC strain isolated from a human infection. VEEV-exposed macaques developed a biphasic fever after infection similar to that seen in humans. Maximum temperature deviation correlated with the inhaled dose, but fever duration did not. Neurological signs, suggestive of virus penetration into the central nervous system (CNS), were predominantly seen in the second febrile period. Electroencephalography data indicated a statistically significant decrease in all power bands and circadian index during the second febrile period that returned to normal after fever resolved. Intracranial pressure increased late in the second febrile period. On day 6 post-infection macaques had high levels of MCP-1 and IP-10 chemokines in the CNS, as well as a marked increase of T lymphocytes and activated microglia. More than four weeks after infection, VEEV genomic RNA was found in the brain, cerebrospinal fluid and cervical lymph nodes. Pro-inflammatory cytokines & chemokines, infiltrating leukocytes and pathological changes were seen in the CNS tissues of macaques euthanized at these times. These data are consistent with persistence of virus replication and/or genomic RNA and potentially, inflammatory sequelae in the central nervous system after resolution of acute VEEV disease.


Asunto(s)
Virus de la Encefalitis Equina Venezolana , Encefalomielitis Equina Venezolana , Animales , Sistema Nervioso Central , Virus de la Encefalitis Equina Venezolana/genética , Caballos/genética , Inflamación , Macaca fascicularis , ARN Viral/genética
11.
Nature ; 557(7706): 570-574, 2018 05.
Artículo en Inglés | MEDLINE | ID: mdl-29769725

RESUMEN

Arthritogenic alphaviruses comprise a group of enveloped RNA viruses that are transmitted to humans by mosquitoes and cause debilitating acute and chronic musculoskeletal disease 1 . The host factors required for alphavirus entry remain poorly characterized 2 . Here we use a genome-wide CRISPR-Cas9-based screen to identify the cell adhesion molecule Mxra8 as an entry mediator for multiple emerging arthritogenic alphaviruses, including chikungunya, Ross River, Mayaro and O'nyong nyong viruses. Gene editing of mouse Mxra8 or human MXRA8 resulted in reduced levels of viral infection of cells and, reciprocally, ectopic expression of these genes resulted in increased infection. Mxra8 bound directly to chikungunya virus particles and enhanced virus attachment and internalization into cells. Consistent with these findings, Mxra8-Fc fusion protein or anti-Mxra8 monoclonal antibodies blocked chikungunya virus infection in multiple cell types, including primary human synovial fibroblasts, osteoblasts, chondrocytes and skeletal muscle cells. Mutagenesis experiments suggest that Mxra8 binds to a surface-exposed region across the A and B domains of chikungunya virus E2 protein, which are a speculated site of attachment. Finally, administration of the Mxra8-Fc protein or anti-Mxra8 blocking antibodies to mice reduced chikungunya and O'nyong nyong virus infection as well as associated foot swelling. Pharmacological targeting of Mxra8 could form a strategy for mitigating infection and disease by multiple arthritogenic alphaviruses.


Asunto(s)
Virus Chikungunya/metabolismo , Inmunoglobulinas/metabolismo , Proteínas de la Membrana/metabolismo , Virus O'nyong-nyong/metabolismo , Receptores Virales/metabolismo , Células 3T3 , Animales , Anticuerpos Bloqueadores/inmunología , Sistemas CRISPR-Cas/genética , Virus Chikungunya/patogenicidad , Condrocitos/metabolismo , Fibroblastos/metabolismo , Humanos , Inmunoglobulinas/inmunología , Masculino , Proteínas de la Membrana/antagonistas & inhibidores , Proteínas de la Membrana/inmunología , Ratones , Músculo Esquelético/citología , Músculo Esquelético/metabolismo , Virus O'nyong-nyong/patogenicidad , Osteoblastos/metabolismo , Receptores Fc/metabolismo , Receptores Virales/deficiencia , Receptores Virales/genética
12.
PLoS Pathog ; 17(2): e1009308, 2021 02.
Artículo en Inglés | MEDLINE | ID: mdl-33534855

RESUMEN

Aerosol exposure to eastern equine encephalitis virus (EEEV) can trigger a lethal viral encephalitis in cynomolgus macaques which resembles severe human disease. Biomarkers indicative of central nervous system (CNS) infection by the virus and lethal outcome of disease would be useful in evaluating potential medical countermeasures, especially for therapeutic compounds. To meet requirements of the Animal Rule, a better understanding of the pathophysiology of EEEV-mediated disease in cynomolgus macaques is needed. In this study, macaques given a lethal dose of clone-derived EEEV strain V105 developed a fever between 2-3 days post infection (dpi) and succumbed to the disease by 6 dpi. At the peak of the febrile phase, there was a significant increase in the delta electroencephalography (EEG) power band associated with deep sleep as well as a sharp rise in intracranial pressure (ICP). Viremia peaked early after infection and was largely absent by the onset of fever. Granulocytosis and elevated plasma levels of IP-10 were found early after infection. At necropsy, there was a one hundred- to one thousand-fold increase in expression of traumatic brain injury genes (LIF, MMP-9) as well as inflammatory cytokines and chemokines (IFN-γ, IP-10, MCP-1, IL-8, IL-6) in the brain tissues. Phenotypic analysis of leukocytes entering the brain identified cells as primarily lymphoid (T, B, NK cells) with lower levels of infiltrating macrophages and activated microglia. Massive amounts of infectious virus were found in the brains of lethally-infected macaques. While no infectious virus was found in surviving macaques, quantitative PCR did find evidence of viral genomes in the brains of several survivors. These data are consistent with an overwhelming viral infection in the CNS coupled with a tremendous inflammatory response to the infection that may contribute to the disease outcome. Physiological monitoring of EEG and ICP represent novel methods for assessing efficacy of vaccines or therapeutics in the cynomolgus macaque model of EEEV encephalitis.


Asunto(s)
Aerosoles/efectos adversos , Biomarcadores/análisis , Encéfalo/inmunología , Encéfalo/patología , Virus de la Encefalitis Equina del Este/patogenicidad , Encefalitis Viral/inmunología , Fiebre/inmunología , Animales , Encéfalo/virología , Citocinas/metabolismo , Modelos Animales de Enfermedad , Encefalitis Viral/patología , Encefalitis Viral/virología , Femenino , Fiebre/patología , Fiebre/virología , Macaca fascicularis , Masculino
13.
Proc Natl Acad Sci U S A ; 117(16): 8890-8899, 2020 04 21.
Artículo en Inglés | MEDLINE | ID: mdl-32245806

RESUMEN

Eastern equine encephalitis virus (EEEV), a mosquito-borne icosahedral alphavirus found mainly in North America, causes human and equine neurotropic infections. EEEV neurovirulence is influenced by the interaction of the viral envelope protein E2 with heparan sulfate (HS) proteoglycans from the host's plasma membrane during virus entry. Here, we present a 5.8-Å cryoelectron microscopy (cryo-EM) structure of EEEV complexed with the HS analog heparin. "Peripheral" HS binding sites were found to be associated with the base of each of the E2 glycoproteins that form the 60 quasi-threefold spikes (q3) and the 20 sites associated with the icosahedral threefold axes (i3). In addition, there is one HS site at the vertex of each q3 and i3 spike (the "axial" sites). Both the axial and peripheral sites are surrounded by basic residues, suggesting an electrostatic mechanism for HS binding. These residues are highly conserved among EEEV strains, and therefore a change in these residues might be linked to EEEV neurovirulence.


Asunto(s)
Diseño de Fármacos , Virus de la Encefalitis Equina del Este/ultraestructura , Encefalomielitis Equina/tratamiento farmacológico , Proteoglicanos de Heparán Sulfato/metabolismo , Heparina/ultraestructura , Animales , Antivirales/farmacología , Antivirales/uso terapéutico , Sitios de Unión/efectos de los fármacos , Línea Celular , Sulfatos de Condroitina/farmacología , Microscopía por Crioelectrón , Virus de la Encefalitis Equina del Este/metabolismo , Encefalomielitis Equina/virología , Proteoglicanos de Heparán Sulfato/análogos & derivados , Heparina/metabolismo , Humanos , Mesocricetus , Estructura Molecular , Relación Estructura-Actividad , Proteínas del Envoltorio Viral/metabolismo , Proteínas del Envoltorio Viral/ultraestructura , Acoplamiento Viral/efectos de los fármacos
14.
J Gen Virol ; 103(2)2022 02.
Artículo en Inglés | MEDLINE | ID: mdl-35191823

RESUMEN

Arboviruses are medically important arthropod-borne viruses that cause a range of diseases in humans from febrile illness to arthritis, encephalitis and hemorrhagic fever. Given their transmission cycles, these viruses face the challenge of replicating in evolutionarily divergent organisms that can include ticks, flies, mosquitoes, birds, rodents, reptiles and primates. Furthermore, their cell attachment receptor utilization may be affected by the opposing needs for generating high and sustained serum viremia in vertebrates such that virus particles are efficiently collected during a hematophagous arthropod blood meal but they must also bind sufficiently to cellular structures on divergent organisms such that productive infection can be initiated and viremia generated. Sulfated polysaccharides of the glycosaminoglycan (GAG) groups, primarily heparan sulfate (HS), have been identified as cell attachment moieties for many arboviruses. Original identification of GAG binding as a phenotype of arboviruses appeared to involve this attribute arising solely as a consequence of adaptation of virus isolates to growth in cell culture. However, more recently, naturally circulating strains of at least one arbovirus, eastern equine encephalitis, have been shown to bind HS efficiently and the GAG binding phenotype continues to be associated with arbovirus infection in published studies. If GAGs are attachment receptors for many naturally circulating arboviruses, this could lead to development of broad-spectrum antiviral therapies through blocking of the virus-GAG interaction. This review summarizes the available data for GAG/HS binding as a phenotype of naturally circulating arbovirus strains emphasizing the importance of avoiding tissue culture amplification and artifactual phenotypes during their isolation.


Asunto(s)
Infecciones por Arbovirus/virología , Arbovirus/inmunología , Heparitina Sulfato/inmunología , Animales , Humanos
15.
PLoS Pathog ; 16(9): e1008903, 2020 09.
Artículo en Inglés | MEDLINE | ID: mdl-32946524

RESUMEN

Vaccines are urgently needed to combat the global coronavirus disease 2019 (COVID-19) pandemic, and testing of candidate vaccines in an appropriate non-human primate (NHP) model is a critical step in the process. Infection of African green monkeys (AGM) with a low passage human isolate of SARS-CoV-2 by aerosol or mucosal exposure resulted in mild clinical infection with a transient decrease in lung tidal volume. Imaging with human clinical-grade 18F-fluoro-2-deoxy-D-glucose positron emission tomography (18F-FDG PET) co-registered with computed tomography (CT) revealed pulmonary lesions at 4 days post-infection (dpi) that resolved over time. Infectious virus was shed from both respiratory and gastrointestinal (GI) tracts in all animals in a biphasic manner, first between 2-7 dpi followed by a recrudescence at 14-21 dpi. Viral RNA (vRNA) was found throughout both respiratory and gastrointestinal systems at necropsy with higher levels of vRNA found within the GI tract tissues. All animals seroconverted simultaneously for IgM and IgG, which has also been documented in human COVID-19 cases. Young AGM represent an species to study mild/subclinical COVID-19 disease and with possible insights into live virus shedding. Future vaccine evaluation can be performed in AGM with correlates of efficacy being lung lesions by PET/CT, virus shedding, and tissue viral load.


Asunto(s)
Betacoronavirus , Infecciones por Coronavirus/diagnóstico por imagen , Tracto Gastrointestinal/virología , Neumonía Viral/diagnóstico por imagen , Esparcimiento de Virus/fisiología , Animales , COVID-19 , Chlorocebus aethiops , Infecciones por Coronavirus/virología , Pulmón/patología , Pulmón/virología , Pandemias , Neumonía Viral/virología , Tomografía Computarizada por Tomografía de Emisión de Positrones/métodos , SARS-CoV-2
16.
PLoS Pathog ; 15(10): e1007867, 2019 10.
Artículo en Inglés | MEDLINE | ID: mdl-31658290

RESUMEN

Eastern equine encephalitis virus (EEEV), a mosquito-borne RNA virus, is one of the most acutely virulent viruses endemic to the Americas, causing between 30% and 70% mortality in symptomatic human cases. A major factor in the virulence of EEEV is the presence of four binding sites for the hematopoietic cell-specific microRNA, miR-142-3p, in the 3' untranslated region (3' UTR) of the virus. Three of the sites are "canonical" with all 7 seed sequence residues complimentary to miR-142-3p while one is "non-canonical" and has a seed sequence mismatch. Interaction of the EEEV genome with miR-142-3p limits virus replication in myeloid cells and suppresses the systemic innate immune response, greatly exacerbating EEEV neurovirulence. The presence of the miRNA binding sequences is also required for efficient EEEV replication in mosquitoes and, therefore, essential for transmission of the virus. In the current studies, we have examined the role of each binding site by point mutagenesis of the seed sequences in all combinations of sites followed by infection of mammalian myeloid cells, mosquito cells and mice. The resulting data indicate that both canonical and non-canonical sites contribute to cell infection and animal virulence, however, surprisingly, all sites are rapidly deleted from EEEV genomes shortly after infection of myeloid cells or mice. Finally, we show that the virulence of a related encephalitis virus, western equine encephalitis virus, is also dependent upon miR-142-3p binding sites.


Asunto(s)
Regiones no Traducidas 3'/genética , Virus de la Encefalitis Equina del Este/genética , Virus de la Encefalitis Equina del Oeste/genética , MicroARNs/genética , Replicación Viral/genética , Aedes , Animales , Sitios de Unión/genética , Línea Celular , Cricetinae , Virus de la Encefalitis Equina del Este/inmunología , Virus de la Encefalitis Equina del Este/patogenicidad , Virus de la Encefalitis Equina del Oeste/inmunología , Virus de la Encefalitis Equina del Oeste/patogenicidad , Encefalomielitis Equina/inmunología , Encefalomielitis Equina/virología , Femenino , Inmunidad Innata/inmunología , Células L , Ratones , Ratones Endogámicos C3H , Ratones Endogámicos C57BL , Células RAW 264.7 , Virulencia/genética
17.
PLoS Pathog ; 15(2): e1007584, 2019 02.
Artículo en Inglés | MEDLINE | ID: mdl-30742691

RESUMEN

Live attenuated vaccines (LAVs), if sufficiently safe, provide the most potent and durable anti-pathogen responses in vaccinees with single immunizations commonly yielding lifelong immunity. Historically, viral LAVs were derived by blind passage of virulent strains in cultured cells resulting in adaptation to culture and a loss of fitness and disease-causing potential in vivo. Mutations associated with these phenomena have been identified but rarely have specific attenuation mechanisms been ascribed, thereby limiting understanding of the attenuating characteristics of the LAV strain and applicability of the attenuation mechanism to other vaccines. Furthermore, the attenuated phenotype is often associated with single nucleotide changes in the viral genome, which can easily revert to the virulent sequence during replication in animals. Here, we have used a rational approach to attenuation of eastern equine encephalitis virus (EEEV), a mosquito-transmitted alphavirus that is among the most acutely human-virulent viruses endemic to North America and has potential for use as an aerosolized bioweapon. Currently, there is no licensed antiviral therapy or vaccine for this virus. Four virulence loci in the EEEV genome were identified and were mutated individually and in combination to abrogate virulence and to resist reversion. The resultant viruses were tested for virulence in mice to examine the degree of attenuation and efficacy was tested by subcutaneous or aerosol challenge with wild type EEEV. Importantly, all viruses containing three or more mutations were avirulent after intracerebral infection of mice, indicating a very high degree of attenuation. All vaccines protected from subcutaneous EEEV challenge while a single vaccine with three mutations provided reproducible, near-complete protection against aerosol challenge. These results suggest that informed mutation of virulence determinants is a productive strategy for production of LAVs even with highly virulent viruses such as EEEV. Furthermore, these results can be directly applied to mutation of analogous virulence loci to create LAVs from other viruses.


Asunto(s)
Virus de la Encefalitis Equina del Este/genética , Virus de la Encefalitis Equina del Este/inmunología , Vacunas Atenuadas/biosíntesis , Animales , Anticuerpos Neutralizantes , Línea Celular , Cricetinae , Virus de la Encefalitis Equina del Este/patogenicidad , Encefalomielitis Equina Oriental/veterinaria , Encefalomielitis Equina Oriental/virología , Femenino , Ingeniería Genética/métodos , Caballos , Ratones , Mutación , América del Norte , Proyectos de Investigación , Vacunas Atenuadas/inmunología , Vacunas Virales/biosíntesis , Virulencia , Factores de Virulencia
18.
Emerg Infect Dis ; 26(9)2020 09.
Artículo en Inglés | MEDLINE | ID: mdl-32568661

RESUMEN

We aerosolized severe acute respiratory syndrome coronavirus 2 and determined that its dynamic aerosol efficiency surpassed those of severe acute respiratory syndrome coronavirus and Middle East respiratory syndrome. Although we performed experiment only once across several laboratories, our findings suggest retained infectivity and virion integrity for up to 16 hours in respirable-sized aerosols.


Asunto(s)
Aerosoles/aislamiento & purificación , Betacoronavirus/aislamiento & purificación , Infecciones por Coronavirus/transmisión , Transmisión de Enfermedad Infecciosa , Neumonía Viral/transmisión , Suspensiones/aislamiento & purificación , COVID-19 , Infecciones por Coronavirus/virología , Humanos , Coronavirus del Síndrome Respiratorio de Oriente Medio/aislamiento & purificación , Pandemias , Neumonía Viral/virología , SARS-CoV-2
19.
J Gen Virol ; 101(11): 1156-1169, 2020 11.
Artículo en Inglés | MEDLINE | ID: mdl-32821033

RESUMEN

Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), the causative agent of coronavirus disease 2019 (COVID-19), emerged at the end of 2019 and by mid-June 2020 the virus had spread to at least 215 countries, caused more than 8 000 000 confirmed infections and over 450 000 deaths, and overwhelmed healthcare systems worldwide. Like severe acute respiratory syndrome coronavirus (SARS-CoV), which emerged in 2002 and caused a similar disease, SARS-CoV-2 is a betacoronavirus. Both viruses use human angiotensin-converting enzyme 2 (hACE2) as a receptor to enter cells. However, the SARS-CoV-2 spike (S) glycoprotein has a novel insertion that generates a putative furin cleavage signal and this has been postulated to expand the host range. Two low-passage (P) strains of SARS-CoV-2 (Wash1 : P4 and Munich : P1) were cultured twice in Vero E6 cells and characterized virologically. Sanger and MinION sequencing demonstrated significant deletions in the furin cleavage signal of Wash1 : P6 and minor variants in the Munich : P3 strain. Cleavage of the S glycoprotein in SARS-CoV-2-infected Vero E6 cell lysates was inefficient even when an intact furin cleavage signal was present. Indirect immunofluorescence demonstrated that the S glycoprotein reached the cell surface. Since the S protein is a major antigenic target for the development of neutralizing antibodies, we investigated the development of neutralizing antibody titres in serial serum samples obtained from COVID-19 human patients. These were comparable regardless of the presence of an intact or deleted furin cleavage signal. These studies illustrate the need to characterize virus stocks meticulously prior to performing either in vitro or in vivo pathogenesis studies.


Asunto(s)
COVID-19/metabolismo , COVID-19/virología , Furina/metabolismo , Interacciones Huésped-Patógeno , SARS-CoV-2/fisiología , Replicación Viral , Adaptación Fisiológica , Animales , Anticuerpos Neutralizantes/inmunología , COVID-19/epidemiología , COVID-19/inmunología , Chlorocebus aethiops , Furina/inmunología , Variación Genética , Hospitalización , Interacciones Huésped-Patógeno/inmunología , Humanos , Inmunoglobulina A/inmunología , Inmunoglobulina G/inmunología , Pruebas de Neutralización , Proteolisis , ARN Viral , Análisis de Secuencia de ARN , Células Vero , Carga Viral
20.
J Virol ; 93(24)2019 12 15.
Artículo en Inglés | MEDLINE | ID: mdl-31578290

RESUMEN

Alphavirus infection of fibroblastic cell types in vitro inhibits host cell translation and transcription, leading to suppression of interferon alpha/beta (IFN-α/ß) production. However, the effect of infection upon myeloid cells, which are often the first cells encountered by alphaviruses in vivo, is unclear. Previous studies demonstrated an association of systemic IFN-α/ß production with myeloid cell infection efficiency. Murine infection with wild-type Venezuelan equine encephalitis virus (VEEV), a highly myeloid-cell-tropic alphavirus, results in secretion of very high systemic levels of IFN-α/ß, suggesting that stress responses in responding cells are active. Here, we infected myeloid cell cultures with VEEV to identify the cellular source of IFN-α/ß, the timing and extent of translation and/or transcription inhibition in infected cells, and the transcription factors responsible for IFN-α/ß induction. In contrast to fibroblast infection, myeloid cell cultures infected with VEEV secreted IFN-α/ß that increased until cell death was observed. VEEV inhibited translation in most cells early after infection (<6 h postinfection [p.i.]), while transcription inhibition occurred later (>6 h p.i.). Furthermore, the interferon regulatory factor 7 (IRF7), but not IRF3, transcription factor was critical for IFN-α/ß induction in vitro and in sera of mice. We identified a subset of infected Raw 264.7 myeloid cells that resisted VEEV-induced translation inhibition and secreted IFN-α/ß despite virus infection. However, in the absence of IFN receptor signaling, the size of this cell population was diminished. These results indicate that IFN-α/ß induction in vivo is IRF7 dependent and arises in part from a subset of myeloid cells that are resistant, in an IFN-α/ß-dependent manner, to VEEV-induced macromolecular synthesis inhibition.IMPORTANCE Most previous research exploring the interaction of alphaviruses with host cell antiviral responses has been conducted using fibroblast lineage cell lines. Previous studies have led to the discovery of virus-mediated activities that antagonize host cell antiviral defense pathways, such as host cell translation and transcription inhibition and suppression of STAT1 signaling. However, their relevance and impact upon myeloid lineage cell types, which are key responders during the initial stages of alphavirus infection in vivo, have not been well studied. Here, we demonstrate the different abilities of myeloid cells to resist VEEV infection compared to nonmyeloid cell types and begin to elucidate the mechanisms by which host antiviral responses are upregulated in myeloid cells despite the actions of virus-encoded antagonists.


Asunto(s)
Infecciones por Alphavirus/metabolismo , Factor 7 Regulador del Interferón/metabolismo , Interferón-alfa/metabolismo , Interferón beta/metabolismo , Sustancias Macromoleculares/metabolismo , Células Mieloides/metabolismo , Alphavirus/fisiología , Animales , Línea Celular , Modelos Animales de Enfermedad , Virus de la Encefalitis Equina Venezolana/fisiología , Fibroblastos/virología , Humanos , Factor 3 Regulador del Interferón/genética , Factor 7 Regulador del Interferón/genética , Ratones , Ratones Noqueados , Células Mieloides/virología , Células RAW 264.7 , Replicación Viral
SELECCIÓN DE REFERENCIAS
Detalles de la búsqueda