Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 92
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Nat Immunol ; 23(1): 40-49, 2022 01.
Artículo en Inglés | MEDLINE | ID: mdl-34937928

RESUMEN

SARS-CoV-2 infection is generally mild or asymptomatic in children but a biological basis for this outcome is unclear. Here we compare antibody and cellular immunity in children (aged 3-11 years) and adults. Antibody responses against spike protein were high in children and seroconversion boosted responses against seasonal Beta-coronaviruses through cross-recognition of the S2 domain. Neutralization of viral variants was comparable between children and adults. Spike-specific T cell responses were more than twice as high in children and were also detected in many seronegative children, indicating pre-existing cross-reactive responses to seasonal coronaviruses. Importantly, children retained antibody and cellular responses 6 months after infection, whereas relative waning occurred in adults. Spike-specific responses were also broadly stable beyond 12 months. Therefore, children generate robust, cross-reactive and sustained immune responses to SARS-CoV-2 with focused specificity for the spike protein. These findings provide insight into the relative clinical protection that occurs in most children and might help to guide the design of pediatric vaccination regimens.


Asunto(s)
Anticuerpos Antivirales/inmunología , Coronavirus Humano 229E/inmunología , Coronavirus Humano OC43/inmunología , Protección Cruzada/inmunología , SARS-CoV-2/inmunología , Glicoproteína de la Espiga del Coronavirus/inmunología , Inmunidad Adaptativa/inmunología , Adulto , Anticuerpos Neutralizantes/inmunología , COVID-19/inmunología , Vacunas contra la COVID-19/inmunología , Niño , Preescolar , Reacciones Cruzadas/inmunología , Humanos
2.
Emerg Infect Dis ; 30(4): 836-838, 2024 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-38526202

RESUMEN

We conducted a cross-sectional study of Crimean-Congo hemorrhagic fever virus (CCHFV) in northern Tanzania. CCHFV seroprevalence in humans and ruminant livestock was high, as were spatial heterogeneity levels. CCHFV could represent an unrecognized human health risk in this region and should be included as a differential diagnosis for febrile illness.


Asunto(s)
Virus de la Fiebre Hemorrágica de Crimea-Congo , Humanos , Animales , Ganado , Estudios Transversales , Estudios Seroepidemiológicos , Tanzanía/epidemiología
3.
J Gen Virol ; 105(6)2024 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-38861287

RESUMEN

Increased human-to-human transmission of monkeypox virus (MPXV) is cause for concern, and antibodies directed against vaccinia virus (VACV) are known to confer cross-protection against Mpox. We used 430 serum samples derived from the Scottish patient population to investigate antibody-mediated cross-neutralization against MPXV. By combining electrochemiluminescence immunoassays with live-virus neutralization assays, we show that people born when smallpox vaccination was routinely offered in the United Kingdom have increased levels of antibodies that cross-neutralize MPXV. Our results suggest that age is a risk factor of Mpox infection, and people born after 1971 are at higher risk of infection upon exposure.


Asunto(s)
Anticuerpos Neutralizantes , Anticuerpos Antivirales , Monkeypox virus , Mpox , Vacuna contra Viruela , Humanos , Anticuerpos Antivirales/sangre , Anticuerpos Antivirales/inmunología , Vacuna contra Viruela/inmunología , Vacuna contra Viruela/administración & dosificación , Adulto , Persona de Mediana Edad , Monkeypox virus/inmunología , Adulto Joven , Anticuerpos Neutralizantes/sangre , Anticuerpos Neutralizantes/inmunología , Mpox/inmunología , Mpox/prevención & control , Femenino , Adolescente , Anciano , Masculino , Protección Cruzada/inmunología , Escocia , Factores de Edad , Pruebas de Neutralización , Niño , Vacunación , Viruela/prevención & control , Viruela/inmunología , Preescolar , Reacciones Cruzadas , Anciano de 80 o más Años
4.
EMBO Rep ; 23(10): e54322, 2022 10 06.
Artículo en Inglés | MEDLINE | ID: mdl-35999696

RESUMEN

The emergence of SARS-CoV-2 variants has exacerbated the COVID-19 global health crisis. Thus far, all variants carry mutations in the spike glycoprotein, which is a critical determinant of viral transmission being responsible for attachment, receptor engagement and membrane fusion, and an important target of immunity. Variants frequently bear truncations of flexible loops in the N-terminal domain (NTD) of spike; the functional importance of these modifications has remained poorly characterised. We demonstrate that NTD deletions are important for efficient entry by the Alpha and Omicron variants and that this correlates with spike stability. Phylogenetic analysis reveals extensive NTD loop length polymorphisms across the sarbecoviruses, setting an evolutionary precedent for loop remodelling. Guided by these analyses, we demonstrate that variations in NTD loop length, alone, are sufficient to modulate virus entry. We propose that variations in NTD loop length act to fine-tune spike; this may provide a mechanism for SARS-CoV-2 to navigate a complex selection landscape encompassing optimisation of essential functionality, immune-driven antigenic variation and ongoing adaptation to a new host.


Asunto(s)
COVID-19 , SARS-CoV-2 , COVID-19/genética , Humanos , Filogenia , SARS-CoV-2/genética , Glicoproteína de la Espiga del Coronavirus/genética
5.
Emerg Infect Dis ; 29(6): 1223-1227, 2023 06.
Artículo en Inglés | MEDLINE | ID: mdl-37141617

RESUMEN

Anthropogenic transmission of SARS-CoV-2 to pet cats highlights the importance of monitoring felids for exposure to circulating variants. We tested cats in the United Kingdom for SARS-CoV-2 antibodies; seroprevalence peaked during September 2021-February 2022. The variant-specific response in cats trailed circulating variants in humans, indicating multiple human-to-cat transmissions over a prolonged period.


Asunto(s)
COVID-19 , SARS-CoV-2 , Humanos , Gatos , Animales , Estudios Seroepidemiológicos , COVID-19/epidemiología , COVID-19/veterinaria , Anticuerpos Antivirales , Reino Unido/epidemiología
6.
PLoS Pathog ; 17(12): e1010022, 2021 12.
Artículo en Inglés | MEDLINE | ID: mdl-34855916

RESUMEN

Vaccines are proving to be highly effective in controlling hospitalisation and deaths associated with SARS-CoV-2 infection but the emergence of viral variants with novel antigenic profiles threatens to diminish their efficacy. Assessment of the ability of sera from vaccine recipients to neutralise SARS-CoV-2 variants will inform the success of strategies for minimising COVID19 cases and the design of effective antigenic formulations. Here, we examine the sensitivity of variants of concern (VOCs) representative of the B.1.617.1 and B.1.617.2 (first associated with infections in India) and B.1.351 (first associated with infection in South Africa) lineages of SARS-CoV-2 to neutralisation by sera from individuals vaccinated with the BNT162b2 (Pfizer/BioNTech) and ChAdOx1 (Oxford/AstraZeneca) vaccines. Across all vaccinated individuals, the spike glycoproteins from B.1.617.1 and B.1.617.2 conferred reductions in neutralisation of 4.31 and 5.11-fold respectively. The reduction seen with the B.1.617.2 lineage approached that conferred by the glycoprotein from B.1.351 (South African) variant (6.29-fold reduction) that is known to be associated with reduced vaccine efficacy. Neutralising antibody titres elicited by vaccination with two doses of BNT162b2 were significantly higher than those elicited by vaccination with two doses of ChAdOx1. Fold decreases in the magnitude of neutralisation titre following two doses of BNT162b2, conferred reductions in titre of 7.77, 11.30 and 9.56-fold respectively to B.1.617.1, B.1.617.2 and B.1.351 pseudoviruses, the reduction in neutralisation of the delta variant B.1.617.2 surpassing that of B.1.351. Fold changes in those vaccinated with two doses of ChAdOx1 were 0.69, 4.01 and 1.48 respectively. The accumulation of mutations in these VOCs, and others, demonstrate the quantifiable risk of antigenic drift and subsequent reduction in vaccine efficacy. Accordingly, booster vaccines based on updated variants are likely to be required over time to prevent productive infection. This study also suggests that two dose regimes of vaccine are required for maximal BNT162b2 and ChAdOx1-induced immunity.


Asunto(s)
Anticuerpos Neutralizantes/inmunología , Anticuerpos Antivirales/inmunología , Vacuna BNT162 , COVID-19 , Inmunización Secundaria , SARS-CoV-2/inmunología , Eficacia de las Vacunas , Deriva y Cambio Antigénico/inmunología , Vacuna BNT162/administración & dosificación , Vacuna BNT162/inmunología , COVID-19/inmunología , COVID-19/mortalidad , COVID-19/prevención & control , Células HEK293 , Humanos
7.
PLoS Pathog ; 17(9): e1009929, 2021 09.
Artículo en Inglés | MEDLINE | ID: mdl-34534263

RESUMEN

Remdesivir (RDV), a broadly acting nucleoside analogue, is the only FDA approved small molecule antiviral for the treatment of COVID-19 patients. To date, there are no reports identifying SARS-CoV-2 RDV resistance in patients, animal models or in vitro. Here, we selected drug-resistant viral populations by serially passaging SARS-CoV-2 in vitro in the presence of RDV. Using high throughput sequencing, we identified a single mutation in RNA-dependent RNA polymerase (NSP12) at a residue conserved among all coronaviruses in two independently evolved populations displaying decreased RDV sensitivity. Introduction of the NSP12 E802D mutation into our SARS-CoV-2 reverse genetics backbone confirmed its role in decreasing RDV sensitivity in vitro. Substitution of E802 did not affect viral replication or activity of an alternate nucleoside analogue (EIDD2801) but did affect virus fitness in a competition assay. Analysis of the globally circulating SARS-CoV-2 variants (>800,000 sequences) showed no evidence of widespread transmission of RDV-resistant mutants. Surprisingly, we observed an excess of substitutions in spike at corresponding sites identified in the emerging SARS-CoV-2 variants of concern (i.e., H69, E484, N501, H655) indicating that they can arise in vitro in the absence of immune selection. The identification and characterisation of a drug resistant signature within the SARS-CoV-2 genome has implications for clinical management and virus surveillance.


Asunto(s)
Adenosina Monofosfato/análogos & derivados , Alanina/análogos & derivados , Antivirales/farmacología , Tratamiento Farmacológico de COVID-19 , ARN Polimerasa Dependiente de ARN de Coronavirus/genética , Farmacorresistencia Microbiana/genética , SARS-CoV-2/efectos de los fármacos , Adenosina Monofosfato/farmacología , Alanina/farmacología , Animales , Evolución Biológica , Chlorocebus aethiops , Humanos , Mutación , SARS-CoV-2/genética , Glicoproteína de la Espiga del Coronavirus/metabolismo , Células Vero
8.
Proc Natl Acad Sci U S A ; 117(50): 31954-31962, 2020 12 15.
Artículo en Inglés | MEDLINE | ID: mdl-33229566

RESUMEN

Canine distemper virus (CDV) has recently emerged as an extinction threat for the endangered Amur tiger (Panthera tigris altaica). CDV is vaccine-preventable, and control strategies could require vaccination of domestic dogs and/or wildlife populations. However, vaccination of endangered wildlife remains controversial, which has led to a focus on interventions in domestic dogs, often assumed to be the source of infection. Effective decision making requires an understanding of the true reservoir dynamics, which poses substantial challenges in remote areas with diverse host communities. We carried out serological, demographic, and phylogenetic studies of dog and wildlife populations in the Russian Far East to show that a number of wildlife species are more important than dogs, both in maintaining CDV and as sources of infection for tigers. Critically, therefore, because CDV circulates among multiple wildlife sources, dog vaccination alone would not be effective at protecting tigers. We show, however, that low-coverage vaccination of tigers themselves is feasible and would produce substantive reductions in extinction risks. Vaccination of endangered wildlife provides a valuable component of conservation strategies for endangered species.


Asunto(s)
Moquillo/prevención & control , Especies en Peligro de Extinción/economía , Tigres/virología , Vacunación/economía , Vacunas Virales/administración & dosificación , Animales , Animales Salvajes/virología , Toma de Decisiones en la Organización , Reservorios de Enfermedades/veterinaria , Reservorios de Enfermedades/virología , Moquillo/epidemiología , Moquillo/transmisión , Moquillo/virología , Virus del Moquillo Canino/genética , Virus del Moquillo Canino/inmunología , Perros/sangre , Perros/virología , Estudios de Factibilidad , Femenino , Masculino , Modelos Económicos , Filogenia , Estudios Seroepidemiológicos , Siberia , Tigres/sangre , Vacunación/métodos , Cobertura de Vacunación/economía , Cobertura de Vacunación/métodos , Cobertura de Vacunación/organización & administración , Vacunas Virales/economía
9.
J Infect Dis ; 227(1): 40-49, 2022 12 28.
Artículo en Inglés | MEDLINE | ID: mdl-35920058

RESUMEN

Since the emergence of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), humans have been exposed to distinct SARS-CoV-2 antigens, either by infection with different variants, and/or vaccination. Population immunity is thus highly heterogeneous, but the impact of such heterogeneity on the effectiveness and breadth of the antibody-mediated response is unclear. We measured antibody-mediated neutralization responses against SARS-CoV-2Wuhan, SARS-CoV-2α, SARS-CoV-2δ, and SARS-CoV-2ο pseudoviruses using sera from patients with distinct immunological histories, including naive, vaccinated, infected with SARS-CoV-2Wuhan, SARS-CoV-2α, or SARS-CoV-2δ, and vaccinated/infected individuals. We show that the breadth and potency of the antibody-mediated response is influenced by the number, the variant, and the nature (infection or vaccination) of exposures, and that individuals with mixed immunity acquired by vaccination and natural exposure exhibit the broadest and most potent responses. Our results suggest that the interplay between host immunity and SARS-CoV-2 evolution will shape the antigenicity and subsequent transmission dynamics of SARS-CoV-2, with important implications for future vaccine design.


Neutralizing antibodies provide protection against viruses and are generated because of vaccination or prior infections. The main target of anti-SARS-CoV-2 neutralizing antibodies is a protein called spike, which decorates the viral particle and mediates viral entry into cells. As SARS-CoV-2 evolves, mutations accumulate in the spike protein, allowing the virus to escape antibody-mediated immunity and decreasing vaccine effectiveness. Multiple SARS-CoV-2 variants have appeared since the start of the COVID-19 pandemic, causing various waves of infection through the population and infecting­in some cases­people that had been previously infected or vaccinated. Because the antibody response is highly specific, individuals infected with different variants are likely to have different repertoires of neutralizing antibodies. We studied the breadth and potency of the antibody-mediated response against different SARS-CoV-2 variants using sera from vaccinated people as well as from people infected with different variants. We show that potency of the antibody response against different SARS-CoV-2 variants depends on the particular variant that infected each person, the exposure type (infection or vaccination) and the number and order of exposures. Our study provides insight into the interplay between virus evolution and immunity, as well as important information for the development of better vaccination strategies.


Asunto(s)
COVID-19 , SARS-CoV-2 , Humanos , Anticuerpos , Vacunación , Anticuerpos Antivirales , Anticuerpos Neutralizantes , Glicoproteína de la Espiga del Coronavirus
10.
J Virol ; 95(13): e0017821, 2021 06 10.
Artículo en Inglés | MEDLINE | ID: mdl-33762419

RESUMEN

As the hosts of lentiviruses, almost 40 species of felids (family Felidae) are distributed around the world, and more than 20 feline species test positive for feline immunodeficiency virus (FIV), a lineage of lentiviruses. These observations suggest that FIVs globally infected a variety of feline species through multiple cross-species transmission events during a million-year history. Cellular restriction factors potentially inhibit lentiviral replication and limit cross-species lentiviral transmission, and cellular APOBEC3 deaminases are known as a potent restriction factor. In contrast, lentiviruses have evolutionary-acquired viral infectivity factor (Vif) to neutralize the APOBEC3-mediated antiviral effect. Because the APOBEC3-Vif interaction is strictly specific for viruses and their hosts, a comprehensive investigation focusing on Vif-APOBEC3 interplay can provide clues that will elucidate the roles of this virus-host interplay on cross-species transmission of lentiviruses. Here, we performed a comprehensive investigation with 144 patterns of a round robin test using 18 feline APOBEC3Z3 genes, an antiviral APOBEC3 gene in felid, and 8 FIV Vifs and derived a matrix showing the interplay between feline APOBEC3Z3 and FIV Vif. We particularly focused on the interplay between the APOBEC3Z3 of three felids (domestic cat, ocelot, and Asian golden cat) and an FIV Vif (strain Petaluma), and revealed that residues 65 and 66 of the APOBEC3Z3 protein of multiple felids are responsible for the counteraction triggered by FIV Petaluma Vif. Altogether, our findings can be a clue to elucidate not only the scenarios of the cross-species transmissions of FIVs in felids but also the evolutionary interaction between mammals and lentiviruses. IMPORTANCE Most of the emergences of new virus infections originate from the cross-species transmission of viruses. The fact that some virus infections are strictly specific for the host species indicates that certain "species barriers" in the hosts restrict cross-species jump of viruses, while viruses have evolutionary acquired their own "arms" to overcome/antagonize/neutralize these hurdles. Therefore, understanding of the molecular mechanism leading to successful cross-species viral transmission is crucial for considering the menus of the emergence of novel pathogenic viruses. In the field of retrovirology, APOBEC3-Vif interaction is a well-studied example of the battles between hosts and viruses. Here, we determined the sequences of 11 novel feline APOBEC3Z3 genes and demonstrated that all 18 different feline APOBEC3Z3 proteins tested exhibit anti-feline immunodeficiency virus (FIV) activity. Our comprehensive investigation focusing on the interplay between feline APOBEC3 and FIV Vif can be a clue to elucidate the scenarios of the cross-species transmissions of FIVs in felids.


Asunto(s)
Desaminasas APOBEC-1/metabolismo , Productos del Gen vif/metabolismo , Virus de la Inmunodeficiencia Felina/metabolismo , Infecciones por Lentivirus/transmisión , Animales , Gatos , Línea Celular , Células HEK293 , Especificidad del Huésped/fisiología , Interacciones Huésped-Patógeno/fisiología , Humanos , Infecciones por Lentivirus/patología , Panthera , Replicación Viral/fisiología
11.
J Infect Dis ; 223(6): 971-980, 2021 03 29.
Artículo en Inglés | MEDLINE | ID: mdl-33367847

RESUMEN

Identifying drivers of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) exposure and quantifying population immunity is crucial to prepare for future epidemics. We performed a serial cross-sectional serosurvey throughout the first pandemic wave among patients from the largest health board in Scotland. Screening of 7480 patient serum samples showed a weekly seroprevalence ranging from 0.10% to 8.23% in primary and 0.21% to 17.44% in secondary care, respectively. Neutralization assays showed that highly neutralizing antibodies developed in about half of individuals who tested positive with enzyme-linked immunosorbent assay, mainly among secondary care patients. We estimated the individual probability of SARS-CoV-2 exposure and quantified associated risk factors. We show that secondary care patients, male patients, and 45-64-year-olds exhibit a higher probability of being seropositive. The identification of risk factors and the differences in virus neutralization activity between patient populations provided insights into the patterns of virus exposure during the first pandemic wave and shed light on what to expect in future waves.


Asunto(s)
COVID-19/inmunología , SARS-CoV-2/inmunología , Adolescente , Adulto , Anciano , Anticuerpos Neutralizantes/sangre , Anticuerpos Antivirales/sangre , COVID-19/diagnóstico , COVID-19/epidemiología , Línea Celular , Estudios Transversales , Atención a la Salud , Demografía , Ensayo de Inmunoadsorción Enzimática , Femenino , Humanos , Inmunidad , Masculino , Persona de Mediana Edad , Pandemias , Factores de Riesgo , Escocia/epidemiología , Estudios Seroepidemiológicos , Adulto Joven
12.
J Gen Virol ; 102(4)2021 04.
Artículo en Inglés | MEDLINE | ID: mdl-33739251

RESUMEN

The enveloped morbilliviruses utilise conserved proteinaceous receptors to enter host cells: SLAMF1 or Nectin-4. Receptor binding is initiated by the viral attachment protein Haemagglutinin (H), with the viral Fusion protein (F) driving membrane fusion. Crystal structures of the prototypic morbillivirus measles virus H with either SLAMF1 or Nectin-4 are available and have served as the basis for improved understanding of this interaction. However, whether these interactions remain conserved throughout the morbillivirus genus requires further characterisation. Using a random mutagenesis approach, based on error-prone PCR, we targeted the putative receptor binding site for SLAMF1 interaction on peste des petits ruminants virus (PPRV) H, identifying mutations that inhibited virus-induced cell-cell fusion. These data, combined with structural modelling of the PPRV H and ovine SLAMF1 interaction, indicate this region is functionally conserved across all morbilliviruses. Error-prone PCR provides a powerful tool for functionally characterising functional domains within viral proteins.


Asunto(s)
Moléculas de Adhesión Celular/metabolismo , Hemaglutininas Virales/metabolismo , Virus de la Peste de los Pequeños Rumiantes/metabolismo , Reacción en Cadena de la Polimerasa/métodos , Miembro 1 de la Familia de Moléculas Señalizadoras de la Activación Linfocitaria/metabolismo , Proteínas Virales de Fusión/metabolismo , Animales , Interacciones Microbiota-Huesped , Fusión de Membrana , Ovinos
14.
Proc Biol Sci ; 286(1899): 20182772, 2019 03 27.
Artículo en Inglés | MEDLINE | ID: mdl-30914008

RESUMEN

Understanding multi-host pathogen maintenance and transmission dynamics is critical for disease control. However, transmission dynamics remain enigmatic largely because they are difficult to observe directly, particularly in wildlife. Here, we investigate the transmission dynamics of canine parvovirus (CPV) using state-space modelling of 20 years of CPV serology data from domestic dogs and African lions in the Serengeti ecosystem. We show that, although vaccination reduces the probability of infection in dogs, and despite indirect enhancement of population seropositivity as a result of vaccine shedding, the vaccination coverage achieved has been insufficient to prevent CPV from becoming widespread. CPV is maintained by the dog population and has become endemic with approximately 3.5-year cycles and prevalence reaching approximately 80%. While the estimated prevalence in lions is lower, peaks of infection consistently follow those in dogs. Dogs exposed to CPV are also more likely to become infected with a second multi-host pathogen, canine distemper virus. However, vaccination can weaken this coupling, raising questions about the value of monovalent versus polyvalent vaccines against these two pathogens. Our findings highlight the need to consider both pathogen- and host-level community interactions when seeking to understand the dynamics of multi-host pathogens and their implications for conservation, disease surveillance and control programmes.


Asunto(s)
Enfermedades de los Perros/transmisión , Leones , Infecciones por Parvoviridae/veterinaria , Parvovirus Canino/fisiología , Animales , Teorema de Bayes , Enfermedades de los Perros/epidemiología , Perros , Ecosistema , Modelos Biológicos , Infecciones por Parvoviridae/epidemiología , Infecciones por Parvoviridae/transmisión , Prevalencia , Estudios Seroepidemiológicos , Tanzanía/epidemiología
15.
J Virol ; 92(23)2018 12 01.
Artículo en Inglés | MEDLINE | ID: mdl-30232185

RESUMEN

Morbilliviruses infect a broad range of mammalian hosts, including ruminants, carnivores, and humans. The recent eradication of rinderpest virus (RPV) and the active campaigns for eradication of the human-specific measles virus (MeV) have raised significant concerns that the remaining morbilliviruses may emerge in so-called vacated ecological niches. Seeking to assess the zoonotic potential of nonhuman morbilliviruses within human populations, we found that peste des petits ruminants virus (PPRV)-the small-ruminant morbillivirus-is restricted at the point of entry into human cells due to deficient interactions with human SLAMF1-the immune cell receptor for morbilliviruses. Using a structure-guided approach, we characterized a single amino acid change, mapping to the receptor-binding domain in the PPRV hemagglutinin (H) protein, which overcomes this restriction. The same mutation allowed escape from some cross-protective, human patient, anti-MeV antibodies, raising concerns that PPRV is a pathogen with zoonotic potential. Analysis of natural variation within human and ovine SLAMF1 also identified polymorphisms that could correlate with disease resistance. Finally, the mechanistic nature of the PPRV restriction was also investigated, identifying charge incompatibility and steric hindrance between PPRV H and human SLAMF1 proteins. Importantly, this research was performed entirely using surrogate virus entry assays, negating the requirement for in situ derivation of a human-tropic PPRV and illustrating alternative strategies for identifying gain-of-function mutations in viral pathogens.IMPORTANCE A significant proportion of viral pandemics occur following zoonotic transmission events, where animal-associated viruses jump species into human populations. In order to provide forewarnings of the emergence of these viruses, it is necessary to develop a better understanding of what determines virus host range, often at the genetic and structural levels. In this study, we demonstrated that the small-ruminant morbillivirus, a close relative of measles, is unable to use human receptors to enter cells; however, a change of a single amino acid in the virus is sufficient to overcome this restriction. This information will be important for monitoring this virus's evolution in the field. Of note, this study was undertaken in vitro, without generation of a fully infectious virus with this phenotype.


Asunto(s)
Anticuerpos Antivirales/inmunología , Glicoproteínas/metabolismo , Mutación , Peste de los Pequeños Rumiantes/virología , Virus de la Peste de los Pequeños Rumiantes/patogenicidad , Miembro 1 de la Familia de Moléculas Señalizadoras de la Activación Linfocitaria/metabolismo , Replicación Viral , Secuencia de Aminoácidos , Animales , Chlorocebus aethiops , Glicoproteínas/química , Glicoproteínas/genética , Glicoproteínas/inmunología , Humanos , Modelos Teóricos , Mutagénesis Sitio-Dirigida , Peste de los Pequeños Rumiantes/patología , Peste de los Pequeños Rumiantes/transmisión , Virus de la Peste de los Pequeños Rumiantes/genética , Virus de la Peste de los Pequeños Rumiantes/inmunología , Conformación Proteica , Homología de Secuencia , Ovinos , Miembro 1 de la Familia de Moléculas Señalizadoras de la Activación Linfocitaria/química , Miembro 1 de la Familia de Moléculas Señalizadoras de la Activación Linfocitaria/genética , Miembro 1 de la Familia de Moléculas Señalizadoras de la Activación Linfocitaria/inmunología , Células Vero
16.
Proc Natl Acad Sci U S A ; 112(5): 1464-9, 2015 Feb 03.
Artículo en Inglés | MEDLINE | ID: mdl-25605919

RESUMEN

Morbilliviruses cause many diseases of medical and veterinary importance, and although some (e.g., measles and rinderpest) have been controlled successfully, others, such as canine distemper virus (CDV), are a growing concern. A propensity for host-switching has resulted in CDV emergence in new species, including endangered wildlife, posing challenges for controlling disease in multispecies communities. CDV is typically associated with domestic dogs, but little is known about its maintenance and transmission in species-rich areas or about the potential role of domestic dog vaccination as a means of reducing disease threats to wildlife. We address these questions by analyzing a long-term serological dataset of CDV in lions and domestic dogs from Tanzania's Serengeti ecosystem. Using a Bayesian state-space model, we show that dynamics of CDV have changed considerably over the past three decades. Initially, peaks of CDV infection in dogs preceded those in lions, suggesting that spill-over from dogs was the main driver of infection in wildlife. However, despite dog-to-lion transmission dominating cross-species transmission models, infection peaks in lions became more frequent and asynchronous from those in dogs, suggesting that other wildlife species may play a role in a potentially complex maintenance community. Widespread mass vaccination of domestic dogs reduced the probability of infection in dogs and the size of outbreaks but did not prevent transmission to or peaks of infection in lions. This study demonstrates the complexity of CDV dynamics in natural ecosystems and the value of long-term, large-scale datasets for investigating transmission patterns and evaluating disease control strategies.


Asunto(s)
Animales Domésticos , Animales Salvajes , Virus del Moquillo Canino/patogenicidad , Morbillivirus/patogenicidad , Animales , Teorema de Bayes , Moquillo/transmisión , Moquillo/virología , Virus del Moquillo Canino/fisiología , Perros , Leones , Morbillivirus/fisiología
17.
J Gen Virol ; 98(11): 2635-2644, 2017 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-29022862

RESUMEN

Peste des petits ruminants virus (PPRV) is a significant pathogen of small ruminants and is prevalent in much of Africa, the Near and Middle East and Asia. Despite the availability of an efficacious and cheap live-attenuated vaccine, the virus has continued to spread, with its range stretching from Morocco in the west to China and Mongolia in the east. Some of the world's poorest communities rely on small ruminant farming for subsistence and the continued endemicity of PPRV is a constant threat to their livelihoods. Moreover, PPRV's effects on the world's population are felt broadly across many economic, agricultural and social situations. This far-reaching impact has prompted the Food and Agriculture Organization of the United Nations (FAO) and the World Organisation for Animal Health (OIE) to develop a global strategy for the eradication of this virus and its disease. PPRV is a morbillivirus and, given the experience of these organizations in eradicating the related rinderpest virus, the eradication of PPRV should be feasible. However, there are many critical areas where basic and applied virological research concerning PPRV is lacking. The purpose of this review is to highlight areas where new research could be performed in order to guide and facilitate the eradication programme. These areas include studies on disease transmission and epidemiology, the existence of wildlife reservoirs and the development of next-generation vaccines and diagnostics. With the support of the international virology community, the successful eradication of PPRV can be achieved.


Asunto(s)
Transmisión de Enfermedad Infecciosa/veterinaria , Peste de los Pequeños Rumiantes/epidemiología , Peste de los Pequeños Rumiantes/prevención & control , África/epidemiología , Animales , Asia/epidemiología , Erradicación de la Enfermedad/organización & administración , Transmisión de Enfermedad Infecciosa/prevención & control , Medio Oriente/epidemiología , Medicina Veterinaria/organización & administración , Organización Mundial de la Salud
18.
J Virol ; 90(10): 5152-5162, 2016 05 15.
Artículo en Inglés | MEDLINE | ID: mdl-26984722

RESUMEN

UNLABELLED: Although rinderpest virus (RPV) has been eradicated in the wild, efforts are still continuing to restrict the extent to which live virus is distributed in facilities around the world and to prepare for any reappearance of the disease, whether through deliberate or accidental release. In an effort to find an alternative vaccine which could be used in place of the traditional live attenuated RPV strains, we have determined whether cattle can be protected from rinderpest by inoculation with vaccine strains of the related morbillivirus, peste des petits ruminants virus (PPRV). Cattle were vaccinated with wild-type PPRV or either of two established PPRV vaccine strains, Nigeria/75/1 or Sungri/96. All animals developed antibody and T cell immune responses to the inoculated PPRV. However, only the animals given wild-type PPRV were protected from RPV challenge. Animals given PPRV/Sungri/96 were only partially protected, and animals given PPRV/Nigeria/75/1 showed no protection against RPV challenge. While sera from animals vaccinated with the vaccine strain of RPV showed cross-neutralizing ability against PPRV, none of the sera from animals vaccinated with any strain of PPRV was able to neutralize RPV although sera from animals inoculated with wild-type PPRV were able to neutralize RPV-pseudotyped vesicular stomatitis virus. IMPORTANCE: Rinderpest virus has been eradicated, and it is only the second virus for which this is so. Significant efforts are still required to ensure preparedness for a possible escape of RPV from a laboratory or its deliberate release. Since RPV vaccine protects sheep and goats from PPRV, it is important to determine if the reverse is true as this would provide a non-RPV vaccine for dealing with suspected RPV outbreaks. This is probably the last in vivo study with live RPV that will be approved.


Asunto(s)
Enfermedades de los Bovinos/prevención & control , Virus de la Peste de los Pequeños Rumiantes/inmunología , Virus de la Peste Bovina/inmunología , Peste Bovina/prevención & control , Vacunas Virales/inmunología , Animales , Anticuerpos Antivirales/inmunología , Bovinos , Enfermedades de los Bovinos/virología , Reacciones Cruzadas , Ensayo de Inmunoadsorción Enzimática , Peste de los Pequeños Rumiantes/inmunología , Virus de la Peste de los Pequeños Rumiantes/genética , Virus de la Peste de los Pequeños Rumiantes/patogenicidad , Peste Bovina/virología , Vacunación/veterinaria , Vacunas Atenuadas/administración & dosificación , Vacunas Atenuadas/inmunología , Vacunas Virales/administración & dosificación
19.
J Gen Virol ; 96(Pt 3): 671-680, 2015 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-25395594

RESUMEN

Neutralizing antibodies (NAbs) are believed to comprise an essential component of the protective immune response induced by vaccines against feline immunodeficiency virus (FIV) and human immunodeficiency virus (HIV) infections. However, relatively little is known about the role of NAbs in controlling FIV infection and subsequent disease progression. Here, we present studies where we examined the neutralization of HIV-luciferase pseudotypes bearing homologous and heterologous FIV envelope proteins (n = 278) by sequential plasma samples collected at 6 month intervals from naturally infected cats (n = 38) over a period of 18 months. We evaluated the breadth of the NAb response against non-recombinant homologous and heterologous clade A and clade B viral variants, as well as recombinants, and assessed the results, testing for evidence of an association between the potency of the NAb response and the duration of infection, CD4(+) T lymphocyte numbers, health status and survival times of the infected cats. Neutralization profiles varied significantly between FIV-infected cats and strong autologous neutralization, assessed using luciferase-based in vitro assays, did not correlate with the clinical outcome. No association was observed between strong NAb responses and either improved health status or increased survival time of infected animals, implying that other protective mechanisms were likely to be involved. Similarly, no correlation was observed between the development of autologous NAbs and the duration of infection. Furthermore, cross-neutralizing antibodies were evident in only a small proportion (13 %) of cats.


Asunto(s)
Anticuerpos Neutralizantes/fisiología , Especificidad de Anticuerpos , Virus de la Inmunodeficiencia Felina/inmunología , Infecciones por Lentivirus/veterinaria , Animales , Linfocitos T CD4-Positivos , Gatos , Clonación Molecular , Femenino , Regulación Viral de la Expresión Génica/fisiología , Productos del Gen env/genética , Productos del Gen env/metabolismo , Células HEK293 , Humanos , Infecciones por Lentivirus/inmunología , Infecciones por Lentivirus/virología , Masculino , Pruebas de Neutralización
20.
J Gen Virol ; 96(Pt 4): 893-903, 2015 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-25535323

RESUMEN

Analysing the evolution of feline immunodeficiency virus (FIV) at the intra-host level is important in order to address whether the diversity and composition of viral quasispecies affect disease progression. We examined the intra-host diversity and the evolutionary rates of the entire env and structural fragments of the env sequences obtained from sequential blood samples in 43 naturally infected domestic cats that displayed different clinical outcomes. We observed in the majority of cats that FIV env showed very low levels of intra-host diversity. We estimated that env evolved at a rate of 1.16×10(-3) substitutions per site per year and demonstrated that recombinant sequences evolved faster than non-recombinant sequences. It was evident that the V3-V5 fragment of FIV env displayed higher evolutionary rates in healthy cats than in those with terminal illness. Our study provided the first evidence that the leader sequence of env, rather than the V3-V5 sequence, had the highest intra-host diversity and the highest evolutionary rate of all env fragments, consistent with this region being under a strong selective pressure for genetic variation. Overall, FIV env displayed relatively low intra-host diversity and evolved slowly in naturally infected cats. The maximum evolutionary rate was observed in the leader sequence of env. Although genetic stability is not necessarily a prerequisite for clinical stability, the higher genetic stability of FIV compared with human immunodeficiency virus might explain why many naturally infected cats do not progress rapidly to AIDS.


Asunto(s)
Productos del Gen env/genética , Genes env , Virus de la Inmunodeficiencia Felina/genética , Secuencia de Aminoácidos , Animales , Secuencia de Bases , Evolución Biológica , Gatos , Variación Genética , Datos de Secuencia Molecular , Filogenia
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA