Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 19.672
Filtrar
Más filtros

Intervalo de año de publicación
1.
Cell ; 186(9): 2040-2040.e1, 2023 04 27.
Artículo en Inglés | MEDLINE | ID: mdl-37116474

RESUMEN

Farmed mammals may act as hosts for zoonotic viruses that can cause disease outbreaks in humans. This SnapShot shows which farmed mammals, and to what extent, are of particular risk of harboring and spreading viruses from viral families that are commonly associated with zoonotic disease. It also discusses genome surveillance methods and biosafety measures. To view this SnapShot, open or download the PDF.


Asunto(s)
Virus , Zoonosis , Animales , Humanos , Mamíferos , Brotes de Enfermedades , Medición de Riesgo
2.
Cell ; 186(4): 688-690, 2023 02 16.
Artículo en Inglés | MEDLINE | ID: mdl-36803601

RESUMEN

Trafficking of live mammals is considered a major risk for emergence of zoonotic viruses. SARS-CoV-2-related coronaviruses have previously been identified in pangolins, the world's most smuggled mammal. A new study identifies a MERS-related coronavirus in trafficked pangolins with broad mammalian tropism and a newly acquired furin cleavage site in Spike.


Asunto(s)
Coronavirus , Pangolines , Animales , Humanos , Quirópteros , COVID-19 , Coronavirus del Síndrome Respiratorio de Oriente Medio , Filogenia , SARS-CoV-2 , Coronavirus/fisiología , Zoonosis
3.
Cell ; 185(7): 1117-1129.e8, 2022 03 31.
Artículo en Inglés | MEDLINE | ID: mdl-35298912

RESUMEN

Game animals are wildlife species traded and consumed as food and are potential reservoirs for SARS-CoV and SARS-CoV-2. We performed a meta-transcriptomic analysis of 1,941 game animals, representing 18 species and five mammalian orders, sampled across China. From this, we identified 102 mammalian-infecting viruses, with 65 described for the first time. Twenty-one viruses were considered as potentially high risk to humans and domestic animals. Civets (Paguma larvata) carried the highest number of potentially high-risk viruses. We inferred the transmission of bat-associated coronavirus from bats to civets, as well as cross-species jumps of coronaviruses from bats to hedgehogs, from birds to porcupines, and from dogs to raccoon dogs. Of note, we identified avian Influenza A virus H9N2 in civets and Asian badgers, with the latter displaying respiratory symptoms, as well as cases of likely human-to-wildlife virus transmission. These data highlight the importance of game animals as potential drivers of disease emergence.


Asunto(s)
Animales Salvajes/virología , Enfermedades Transmisibles Emergentes/virología , Reservorios de Enfermedades , Mamíferos/virología , Viroma , Animales , China , Filogenia , Zoonosis
4.
Cell ; 184(8): 1960-1961, 2021 04 15.
Artículo en Inglés | MEDLINE | ID: mdl-33831378

RESUMEN

The events of the past year have underscored the serious and rapid threat that emerging viruses pose to global health. However, much of the rapid progress in understanding and combating SARS-CoV-2 was made possible because of the decades of important groundwork laid from researchers studying other emergent infectious diseases. The 2021 John Dirks Canada Gairdner Global Health award recognizes the contributions of Joseph Sriyal Malik Peiris and Yi Guan toward understanding the origins and options for control of newly emerging infectious disease outbreaks in Asia, notably zoonotic influenza and severe acute respiratory syndrome (SARS). Cell's Nicole Neuman corresponded with Yi Guan about his path to becoming a viral infection sleuth and the challenges of understanding emerging pathogens and their origins. Excerpts of their exchange are included here.


Asunto(s)
COVID-19 , Enfermedades Transmisibles Emergentes , Brotes de Enfermedades , Gripe Humana , Zoonosis , Animales , Asia , COVID-19/epidemiología , COVID-19/transmisión , Enfermedades Transmisibles Emergentes/epidemiología , Enfermedades Transmisibles Emergentes/historia , Enfermedades Transmisibles Emergentes/transmisión , Brotes de Enfermedades/historia , Salud Global , Historia del Siglo XXI , Humanos , Gripe Humana/epidemiología , Gripe Humana/historia , Gripe Humana/transmisión , Zoonosis/epidemiología , Zoonosis/transmisión
5.
Cell ; 184(19): 4848-4856, 2021 09 16.
Artículo en Inglés | MEDLINE | ID: mdl-34480864

RESUMEN

Since the first reports of a novel severe acute respiratory syndrome (SARS)-like coronavirus in December 2019 in Wuhan, China, there has been intense interest in understanding how severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) emerged in the human population. Recent debate has coalesced around two competing ideas: a "laboratory escape" scenario and zoonotic emergence. Here, we critically review the current scientific evidence that may help clarify the origin of SARS-CoV-2.


Asunto(s)
SARS-CoV-2/fisiología , Animales , Evolución Biológica , COVID-19/virología , Humanos , Laboratorios , SARS-CoV-2/genética , Zoonosis/virología
6.
Cell ; 181(2): 223-227, 2020 Apr 16.
Artículo en Inglés | MEDLINE | ID: mdl-32220310

RESUMEN

The ongoing pandemic of a new human coronavirus, SARS-CoV-2, has generated enormous global concern. We and others in China were involved in the initial genome sequencing of the virus. Herein, we describe what genomic data reveal about the emergence SARS-CoV-2 and discuss the gaps in our understanding of its origins.


Asunto(s)
Betacoronavirus/genética , Quirópteros/virología , Infecciones por Coronavirus/virología , Reservorios de Enfermedades/virología , Neumonía Viral/virología , Animales , Animales Salvajes , COVID-19 , China , Infecciones por Coronavirus/transmisión , ADN Ambiental , Genoma Viral , Pandemias , Neumonía Viral/transmisión , SARS-CoV-2 , Análisis de Secuencia de ARN , Zoonosis/virología
7.
Cell ; 167(4): 1079-1087.e5, 2016 11 03.
Artículo en Inglés | MEDLINE | ID: mdl-27814505

RESUMEN

The 2013-2016 outbreak of Ebola virus (EBOV) in West Africa was the largest recorded. It began following the cross-species transmission of EBOV from an animal reservoir, most likely bats, into humans, with phylogenetic analysis revealing the co-circulation of several viral lineages. We hypothesized that this prolonged human circulation led to genomic changes that increased viral transmissibility in humans. We generated a synthetic glycoprotein (GP) construct based on the earliest reported isolate and introduced amino acid substitutions that defined viral lineages. Mutant GPs were used to generate a panel of pseudoviruses, which were used to infect different human and bat cell lines. These data revealed that specific amino acid substitutions in the EBOV GP have increased tropism for human cells, while reducing tropism for bat cells. Such increased infectivity may have enhanced the ability of EBOV to transmit among humans and contributed to the wide geographic distribution of some viral lineages.


Asunto(s)
Evolución Biológica , Ebolavirus/fisiología , Fiebre Hemorrágica Ebola/virología , Especificidad del Huésped , África Occidental/epidemiología , Animales , Quirópteros/virología , Brotes de Enfermedades , Ebolavirus/clasificación , Ebolavirus/genética , Ebolavirus/patogenicidad , Fiebre Hemorrágica Ebola/epidemiología , Fiebre Hemorrágica Ebola/transmisión , Humanos , Mutación , Filogenia , Proteínas del Envoltorio Viral/genética , Proteínas del Envoltorio Viral/metabolismo , Zoonosis
8.
Cell ; 160(1-2): 20-35, 2015 Jan 15.
Artículo en Inglés | MEDLINE | ID: mdl-25533784

RESUMEN

Zoonotic viruses, such as HIV, Ebola virus, coronaviruses, influenza A viruses, hantaviruses, or henipaviruses, can result in profound pathology in humans. In contrast, populations of the reservoir hosts of zoonotic pathogens often appear to tolerate these infections with little evidence of disease. Why are viruses more dangerous in one species than another? Immunological studies investigating quantitative and qualitative differences in the host-virus equilibrium in animal reservoirs will be key to answering this question, informing new approaches for treating and preventing zoonotic diseases. Integrating an understanding of host immune responses with epidemiological, ecological, and evolutionary insights into viral emergence will shed light on mechanisms that minimize fitness costs associated with viral infection, facilitate transmission to other hosts, and underlie the association of specific reservoir hosts with multiple emerging viruses. Reservoir host studies provide a rich opportunity for elucidating fundamental immunological processes and their underlying genetic basis, in the context of distinct physiological and metabolic constraints that contribute to host resistance and disease tolerance.


Asunto(s)
Fenómenos Fisiológicos de los Virus , Zoonosis/virología , Animales , Enfermedades Transmisibles Emergentes/inmunología , Enfermedades Transmisibles Emergentes/transmisión , Enfermedades Transmisibles Emergentes/virología , Reservorios de Enfermedades , Interacciones Huésped-Patógeno , Humanos , Virosis , Zoonosis/inmunología , Zoonosis/transmisión
9.
Cell ; 162(4): 738-50, 2015 Aug 13.
Artículo en Inglés | MEDLINE | ID: mdl-26276630

RESUMEN

The 2013-2015 West African epidemic of Ebola virus disease (EVD) reminds us of how little is known about biosafety level 4 viruses. Like Ebola virus, Lassa virus (LASV) can cause hemorrhagic fever with high case fatality rates. We generated a genomic catalog of almost 200 LASV sequences from clinical and rodent reservoir samples. We show that whereas the 2013-2015 EVD epidemic is fueled by human-to-human transmissions, LASV infections mainly result from reservoir-to-human infections. We elucidated the spread of LASV across West Africa and show that this migration was accompanied by changes in LASV genome abundance, fatality rates, codon adaptation, and translational efficiency. By investigating intrahost evolution, we found that mutations accumulate in epitopes of viral surface proteins, suggesting selection for immune escape. This catalog will serve as a foundation for the development of vaccines and diagnostics. VIDEO ABSTRACT.


Asunto(s)
Genoma Viral , Fiebre de Lassa/virología , Virus Lassa/genética , ARN Viral/genética , África Occidental/epidemiología , Animales , Evolución Biológica , Reservorios de Enfermedades , Ebolavirus/genética , Variación Genética , Glicoproteínas/genética , Fiebre Hemorrágica Ebola/virología , Humanos , Fiebre de Lassa/epidemiología , Fiebre de Lassa/transmisión , Virus Lassa/clasificación , Virus Lassa/fisiología , Murinae/genética , Mutación , Nigeria/epidemiología , Proteínas Virales/genética , Zoonosis/epidemiología , Zoonosis/virología
10.
Trends Immunol ; 45(3): 198-210, 2024 03.
Artículo en Inglés | MEDLINE | ID: mdl-38453576

RESUMEN

Bats are among the most diverse mammalian species, representing over 20% of mammalian diversity. The past two decades have witnessed a disproportionate spillover of viruses from bats to humans compared with other mammalian hosts, attributed to the viral richness within bats, their phylogenetic likeness to humans, and increased human contact with wildlife. Unique evolutionary adaptations in bat genomes, particularly in antiviral protection and immune tolerance genes, enable bats to serve as reservoirs for pandemic-inducing viruses. Here, we discuss current limitations and advances made in understanding the role of bats as drivers of pandemic zoonoses. We also discuss novel technologies that have revealed spatial, dynamic, and physiological factors driving virus and host coevolution.


Asunto(s)
Quirópteros , Virus , Animales , Humanos , Reservorios de Enfermedades , Filogenia , Zoonosis , Mamíferos
11.
Proc Natl Acad Sci U S A ; 121(4): e2312556121, 2024 Jan 23.
Artículo en Inglés | MEDLINE | ID: mdl-38227655

RESUMEN

Hemorrhagic fever with renal syndrome (HFRS) is a zoonotic disease caused by the rodent-transmitted orthohantaviruses (HVs), with China possessing the most cases globally. The virus hosts in China are Apodemus agrarius and Rattus norvegicus, and the disease spread is strongly influenced by global climate dynamics. To assess and predict the spatiotemporal trends of HFRS from 2005 to 2098, we collected historical HFRS data in mainland China (2005-2020), historical and projected climate and population data (2005-2098), and spatial variables including biotic, environmental, topographical, and socioeconomic. Spatiotemporal predictions and mapping were conducted under 27 scenarios incorporating multiple integrated representative concentration pathway models and population scenarios. We identify the type of magistral HVs host species as the best spatial division, including four region categories. Seven extreme climate indices associated with temperature and precipitation have been pinpointed as key factors affecting the trends of HFRS. Our predictions indicate that annual HFRS cases will increase significantly in 62 of 356 cities in mainland China. Rattus regions are predicted to be the most active, surpassing Apodemus and Mixed regions. Eighty cities are identified as at severe risk level for HFRS, each with over 50 reported cases annually, including 22 new cities primarily located in East China and Rattus regions after 2020, while 6 others develop new risk. Our results suggest that the risk of HFRS will remain high through the end of this century, with Rattus norvegicus being the most active host, and that extreme climate indices are significant risk factors. Our findings can inform evidence-based policymaking regarding future risk of HFRS.


Asunto(s)
Fiebre Hemorrágica con Síndrome Renal , Ratas , Animales , Fiebre Hemorrágica con Síndrome Renal/epidemiología , Fiebre Hemorrágica con Síndrome Renal/etiología , Clima , Zoonosis , China/epidemiología , Murinae , Incidencia
12.
Q Rev Biophys ; 57: e2, 2024 03 13.
Artículo en Inglés | MEDLINE | ID: mdl-38477116

RESUMEN

Zoonoses are infectious agents that are transmissible between animals and humans. Up to 60% of known infectious diseases and 75% of emergent diseases are zoonotic. Genomic variation between homeostatic populations provides a novel window into the effect of environmental pathogens on allelic distributions within the populations. Genodynamics is a biophysical approach utilizing developed metrics on biallelic single-nucleotide polymorphisms (SNPs) that can be used to quantify the adaptive influences due to pathogens. A genomic free energy that is minimized when overall population health is optimized describes the influence of environmental agents upon genomic variation. A double-blind exploration of over 100 thousand SNPs searching for smooth functional dependencies upon four zoonotic pathogens carried by four possible hosts amidst populations that live in their ancestral environments has been conducted. Exemplars that infectious agents can have significant adaptive influence on human populations are presented. One discussed SNP is likely associated with both adaptive and innate immune regulation. The adaptive response of another SNP suggests an intriguing connection between zoonoses and human cancers. The adaptive forces of the presented pathogens upon the human genome have been quantified.


Asunto(s)
Genómica , Zoonosis , Animales , Humanos , Zoonosis/epidemiología , Polimorfismo de Nucleótido Simple , Ensayos Clínicos Controlados Aleatorios como Asunto
13.
Annu Rev Med ; 75: 159-175, 2024 Jan 29.
Artículo en Inglés | MEDLINE | ID: mdl-37788486

RESUMEN

Mpox, previously known as monkeypox, is caused by an Orthopoxvirus related to the variola virus that causes smallpox. Prior to 2022, mpox was considered a zoonotic disease endemic to central and west Africa. Since May 2022, more than 86,000 cases of mpox from 110 countries have been identified across the world, predominantly in men who have sex with men, most often acquired through close physical contact or during sexual activity. The classical clinical presentation of mpox is a prodrome including fever, lethargy, and lymphadenopathy followed by a characteristic vesiculopustular rash. The recent 2022 outbreak included novel presentations of mpox with a predominance of anogenital lesions, mucosal lesions, and other features such as anorectal pain, proctitis, oropharyngeal lesions, tonsillitis, and multiphasic skin lesions. We describe the demographics and clinical spectrum of classical and novel mpox, outlining the potential complications and management.


Asunto(s)
Mpox , Minorías Sexuales y de Género , Masculino , Animales , Humanos , Homosexualidad Masculina , Zoonosis , Brotes de Enfermedades
14.
N Engl J Med ; 388(9): 792-803, 2023 03 02.
Artículo en Inglés | MEDLINE | ID: mdl-36856615

RESUMEN

BACKGROUND: The appropriate antibiotic treatment for severe scrub typhus, a neglected but widespread reemerging zoonotic infection, is unclear. METHODS: In this multicenter, double-blind, randomized, controlled trial, we compared the efficacy of intravenous doxycycline, azithromycin, or a combination of both in treating severe scrub typhus. Patients who were 15 years of age or older with severe scrub typhus with at least one organ involvement were enrolled. The patients were assigned to receive a 7-day course of intravenous doxycycline, azithromycin, or both (combination therapy). The primary outcome was a composite of death from any cause at day 28, persistent complications at day 7, and persistent fever at day 5. RESULTS: Among 794 patients (median age, 48 years) who were included in the modified intention-to-treat analysis, complications included those that were respiratory (in 62%), hepatic (in 54%), cardiovascular (in 42%), renal (in 30%), and neurologic (in 20%). The use of combination therapy resulted in a lower incidence of the composite primary outcome than the use of doxycycline (33% and 47%, respectively), for a risk difference of -13.3 percentage points (95% confidence interval [CI], -21.6 to -5.1; P = 0.002). The incidence with combination therapy was also lower than that with azithromycin (48%), for a risk difference of -14.8 percentage points (95% CI, -23.1 to -6.5; P<0.001). No significant difference was seen between the azithromycin and doxycycline groups (risk difference, 1.5 percentage points; 95% CI, -7.0 to 10.0; P = 0.73). The results in the per-protocol analysis were similar to those in the primary analysis. Adverse events and 28-day mortality were similar in the three groups. CONCLUSIONS: Combination therapy with intravenous doxycycline and azithromycin was a better therapeutic option for the treatment of severe scrub typhus than monotherapy with either drug alone. (Funded by the India Alliance and Wellcome Trust; INTREST Clinical Trials Registry-India number, CTRI/2018/08/015159.).


Asunto(s)
Antibacterianos , Azitromicina , Doxiciclina , Tifus por Ácaros , Animales , Humanos , Persona de Mediana Edad , Antibacterianos/administración & dosificación , Antibacterianos/efectos adversos , Antibacterianos/uso terapéutico , Azitromicina/administración & dosificación , Azitromicina/efectos adversos , Azitromicina/uso terapéutico , Doxiciclina/administración & dosificación , Doxiciclina/uso terapéutico , Tifus por Ácaros/tratamiento farmacológico , Zoonosis , Método Doble Ciego , Quimioterapia Combinada , Administración Intravenosa
15.
PLoS Biol ; 21(9): e3002268, 2023 09.
Artículo en Inglés | MEDLINE | ID: mdl-37676899

RESUMEN

The management of future pandemic risk requires a better understanding of the mechanisms that determine the virulence of emerging zoonotic viruses. Meta-analyses suggest that the virulence of emerging zoonoses is correlated with but not completely predictable from reservoir host phylogeny, indicating that specific characteristics of reservoir host immunology and life history may drive the evolution of viral traits responsible for cross-species virulence. In particular, bats host viruses that cause higher case fatality rates upon spillover to humans than those derived from any other mammal, a phenomenon that cannot be explained by phylogenetic distance alone. In order to disentangle the fundamental drivers of these patterns, we develop a nested modeling framework that highlights mechanisms that underpin the evolution of viral traits in reservoir hosts that cause virulence following cross-species emergence. We apply this framework to generate virulence predictions for viral zoonoses derived from diverse mammalian reservoirs, recapturing trends in virus-induced human mortality rates reported in the literature. Notably, our work offers a mechanistic hypothesis to explain the extreme virulence of bat-borne zoonoses and, more generally, demonstrates how key differences in reservoir host longevity, viral tolerance, and constitutive immunity impact the evolution of viral traits that cause virulence following spillover to humans. Our theoretical framework offers a series of testable questions and predictions designed to stimulate future work comparing cross-species virulence evolution in zoonotic viruses derived from diverse mammalian hosts.


Asunto(s)
Quirópteros , Zoonosis , Animales , Humanos , Quirópteros/virología , Filogenia , Virulencia/genética , Zoonosis/virología
16.
Cell ; 147(6): 1209-11, 2011 Dec 09.
Artículo en Inglés | MEDLINE | ID: mdl-22153064
17.
Nature ; 584(7821): 398-402, 2020 08.
Artículo en Inglés | MEDLINE | ID: mdl-32759999

RESUMEN

Land use change-for example, the conversion of natural habitats to agricultural or urban ecosystems-is widely recognized to influence the risk and emergence of zoonotic disease in humans1,2. However, whether such changes in risk are underpinned by predictable ecological changes remains unclear. It has been suggested that habitat disturbance might cause predictable changes in the local diversity and taxonomic composition of potential reservoir hosts, owing to systematic, trait-mediated differences in species resilience to human pressures3,4. Here we analyse 6,801 ecological assemblages and 376 host species worldwide, controlling for research effort, and show that land use has global and systematic effects on local zoonotic host communities. Known wildlife hosts of human-shared pathogens and parasites overall comprise a greater proportion of local species richness (18-72% higher) and total abundance (21-144% higher) in sites under substantial human use (secondary, agricultural and urban ecosystems) compared with nearby undisturbed habitats. The magnitude of this effect varies taxonomically and is strongest for rodent, bat and passerine bird zoonotic host species, which may be one factor that underpins the global importance of these taxa as zoonotic reservoirs. We further show that mammal species that harbour more pathogens overall (either human-shared or non-human-shared) are more likely to occur in human-managed ecosystems, suggesting that these trends may be mediated by ecological or life-history traits that influence both host status and tolerance to human disturbance5,6. Our results suggest that global changes in the mode and the intensity of land use are creating expanding hazardous interfaces between people, livestock and wildlife reservoirs of zoonotic disease.


Asunto(s)
Biodiversidad , Especificidad del Huésped , Zoonosis/microbiología , Zoonosis/parasitología , Zoonosis/virología , Animales , Aves/microbiología , Aves/parasitología , Aves/virología , Humanos , Mamíferos/microbiología , Mamíferos/parasitología , Mamíferos/virología , Especificidad de la Especie , Zoonosis/transmisión
18.
Nature ; 583(7815): 286-289, 2020 07.
Artículo en Inglés | MEDLINE | ID: mdl-32380510

RESUMEN

The current outbreak of coronavirus disease-2019 (COVID-19) poses unprecedented challenges to global health1. The new coronavirus responsible for this outbreak-severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2)-shares high sequence identity to SARS-CoV and a bat coronavirus, RaTG132. Although bats may be the reservoir host for a variety of coronaviruses3,4, it remains unknown whether SARS-CoV-2 has additional host species. Here we show that a coronavirus, which we name pangolin-CoV, isolated from a Malayan pangolin has 100%, 98.6%, 97.8% and 90.7% amino acid identity with SARS-CoV-2 in the E, M, N and S proteins, respectively. In particular, the receptor-binding domain of the S protein of pangolin-CoV is almost identical to that of SARS-CoV-2, with one difference in a noncritical amino acid. Our comparative genomic analysis suggests that SARS-CoV-2 may have originated in the recombination of a virus similar to pangolin-CoV with one similar to RaTG13. Pangolin-CoV was detected in 17 out of the 25 Malayan pangolins that we analysed. Infected pangolins showed clinical signs and histological changes, and circulating antibodies against pangolin-CoV reacted with the S protein of SARS-CoV-2. The isolation of a coronavirus from pangolins that is closely related to SARS-CoV-2 suggests that these animals have the potential to act as an intermediate host of SARS-CoV-2. This newly identified coronavirus from pangolins-the most-trafficked mammal in the illegal wildlife trade-could represent a future threat to public health if wildlife trade is not effectively controlled.


Asunto(s)
Betacoronavirus/genética , Betacoronavirus/aislamiento & purificación , Euterios/virología , Evolución Molecular , Genoma Viral/genética , Homología de Secuencia de Ácido Nucleico , Animales , Betacoronavirus/clasificación , COVID-19 , China , Quirópteros/virología , Chlorocebus aethiops , Proteínas de la Envoltura de Coronavirus , Infecciones por Coronavirus/epidemiología , Infecciones por Coronavirus/patología , Infecciones por Coronavirus/transmisión , Infecciones por Coronavirus/veterinaria , Infecciones por Coronavirus/virología , Proteínas M de Coronavirus , Proteínas de la Nucleocápside de Coronavirus , Reservorios de Enfermedades/virología , Genómica , Especificidad del Huésped , Humanos , Pulmón/patología , Pulmón/virología , Malasia , Proteínas de la Nucleocápside/genética , Pandemias , Fosfoproteínas , Filogenia , Neumonía Viral/epidemiología , Neumonía Viral/transmisión , Neumonía Viral/virología , Reacción en Cadena de la Polimerasa , Recombinación Genética , SARS-CoV-2 , Alineación de Secuencia , Análisis de Secuencia de ARN , Glicoproteína de la Espiga del Coronavirus/genética , Células Vero , Proteínas del Envoltorio Viral/genética , Proteínas de la Matriz Viral/genética , Zoonosis/transmisión , Zoonosis/virología
19.
Nature ; 586(7831): 776-778, 2020 10.
Artículo en Inglés | MEDLINE | ID: mdl-32408337

RESUMEN

Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) was first detected in Wuhan in December 2019 and caused coronavirus disease 2019 (COVID-19)1,2. In 2003, the closely related SARS-CoV had been detected in domestic cats and a dog3. However, little is known about the susceptibility of domestic pet mammals to SARS-CoV-2. Here, using PCR with reverse transcription, serology, sequencing the viral genome and virus isolation, we show that 2 out of 15 dogs from households with confirmed human cases of COVID-19 in Hong Kong were found to be infected with SARS-CoV-2. SARS-CoV-2 RNA was detected in five nasal swabs collected over a 13-day period from a 17-year-old neutered male Pomeranian. A 2.5-year-old male German shepherd was positive for SARS-CoV-2 RNA on two occasions and virus was isolated from nasal and oral swabs. Antibody responses were detected in both dogs using plaque-reduction-neutralization assays. Viral genetic sequences of viruses from the two dogs were identical to the virus detected in the respective human cases. The dogs remained asymptomatic during quarantine. The evidence suggests that these are instances of human-to-animal transmission of SARS-CoV-2. It is unclear whether infected dogs can transmit the virus to other animals or back to humans.


Asunto(s)
Betacoronavirus/aislamiento & purificación , Infecciones por Coronavirus/transmisión , Infecciones por Coronavirus/veterinaria , Enfermedades de los Perros/transmisión , Enfermedades de los Perros/virología , Pandemias/veterinaria , Neumonía Viral/transmisión , Neumonía Viral/veterinaria , Zoonosis/transmisión , Zoonosis/virología , Enzima Convertidora de Angiotensina 2 , Animales , Anticuerpos Neutralizantes/inmunología , Anticuerpos Antivirales/inmunología , Betacoronavirus/genética , COVID-19 , Infecciones por Coronavirus/epidemiología , Infecciones por Coronavirus/virología , Perros , Femenino , Hong Kong/epidemiología , Humanos , Masculino , Persona de Mediana Edad , Peptidil-Dipeptidasa A/metabolismo , Filogenia , Neumonía Viral/epidemiología , Neumonía Viral/virología , Receptores Virales/metabolismo , SARS-CoV-2 , Factores de Tiempo
20.
Nature ; 581(7807): 221-224, 2020 05.
Artículo en Inglés | MEDLINE | ID: mdl-32225175

RESUMEN

A novel severe acute respiratory syndrome (SARS)-like coronavirus (SARS-CoV-2) recently emerged and is rapidly spreading in humans, causing COVID-191,2. A key to tackling this pandemic is to understand the receptor recognition mechanism of the virus, which regulates its infectivity, pathogenesis and host range. SARS-CoV-2 and SARS-CoV recognize the same receptor-angiotensin-converting enzyme 2 (ACE2)-in humans3,4. Here we determined the crystal structure of the receptor-binding domain (RBD) of the spike protein of SARS-CoV-2 (engineered to facilitate crystallization) in complex with ACE2. In comparison with the SARS-CoV RBD, an ACE2-binding ridge in SARS-CoV-2 RBD has a more compact conformation; moreover, several residue changes in the SARS-CoV-2 RBD stabilize two virus-binding hotspots at the RBD-ACE2 interface. These structural features of SARS-CoV-2 RBD increase its ACE2-binding affinity. Additionally, we show that RaTG13, a bat coronavirus that is closely related to SARS-CoV-2, also uses human ACE2 as its receptor. The differences among SARS-CoV-2, SARS-CoV and RaTG13 in ACE2 recognition shed light on the potential animal-to-human transmission of SARS-CoV-2. This study provides guidance for intervention strategies that target receptor recognition by SARS-CoV-2.


Asunto(s)
Betacoronavirus/química , Peptidil-Dipeptidasa A/química , Peptidil-Dipeptidasa A/metabolismo , Receptores Virales/química , Receptores Virales/metabolismo , Glicoproteína de la Espiga del Coronavirus/química , Glicoproteína de la Espiga del Coronavirus/metabolismo , Zoonosis/virología , Enzima Convertidora de Angiotensina 2 , Animales , Betacoronavirus/efectos de los fármacos , Betacoronavirus/metabolismo , Sitios de Unión , COVID-19 , China/epidemiología , Quirópteros/virología , Coronavirus/química , Coronavirus/aislamiento & purificación , Infecciones por Coronavirus/tratamiento farmacológico , Infecciones por Coronavirus/epidemiología , Infecciones por Coronavirus/transmisión , Infecciones por Coronavirus/virología , Cristalización , Cristalografía por Rayos X , Reservorios de Enfermedades/virología , Euterios/virología , Humanos , Modelos Moleculares , Pandemias , Neumonía Viral/tratamiento farmacológico , Neumonía Viral/epidemiología , Neumonía Viral/transmisión , Neumonía Viral/virología , Unión Proteica , Dominios Proteicos , Estabilidad Proteica , Coronavirus Relacionado al Síndrome Respiratorio Agudo Severo/química , SARS-CoV-2 , Glicoproteína de la Espiga del Coronavirus/genética , Zoonosis/epidemiología , Zoonosis/transmisión
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA