Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 37
Filtrar
1.
Transbound Emerg Dis ; 69(6): 3896-3905, 2022 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-36379049

RESUMO

RNA sequence data from SARS CoV2 patients helps to construct a gene network related to this disease. A detailed analysis of the human host response to SARS CoV2 with expression profiling by high-throughput sequencing has been accomplished with primary human lung epithelial cell lines. Using this data, the clustered gene annotation and gene network construction are performed with the help of the String database. Among the four clusters identified, only 1 with 44 genes could be annotated. Interestingly, this corresponded to basal cells with p = 1.37e - 05, which is relevant for respiratory tract infection. Functional enrichment analysis of genes present in the gene network has been completed using the String database and the Network Analyst tool. Among three types of cell-cell communication, only the anchoring junction between the basal cell membrane and the basal lamina in the host cell is involved in the virus transmission. In this junction point, a hemidesmosome structure plays a vital role in virus spread from one cell to basal lamina in the respiratory tract. In this protein complex structure, different integrin protein molecules of the host cell are used to promote the spread of virus infection into the extracellular matrix. So, small molecular blockers of different anchoring junction proteins, such as integrin alpha 3, integrin beta 1, can provide efficient protection against this deadly viral disease. ORF8 from SARS CoV2 virus can interact with both integrin proteins of human host. By using molecular docking technique, a ternary complex of these three proteins is modelled. Several oligopeptides are predicted as modulators for this ternary complex. In silico analysis of these modulators is very important to develop novel therapeutics for the treatment of SARS CoV2.


Assuntos
COVID-19 , Síndrome Respiratória Aguda Grave , Humanos , Animais , COVID-19/veterinária , SARS-CoV-2/genética , Simulação de Acoplamento Molecular , Síndrome Respiratória Aguda Grave/veterinária , Comunicação Celular , Integrinas
2.
Viruses ; 13(11)2021 10 29.
Artigo em Inglês | MEDLINE | ID: mdl-34834994

RESUMO

In the last two decades, several coronavirus (CoV) interspecies jumping events have occurred between bats and other animals/humans, leading to major epidemics/pandemics and high fatalities. The SARS epidemic in 2002/2003 had a ~10% fatality. The discovery of SARS-related CoVs in horseshoe bats and civets and genomic studies have confirmed bat-to-civet-to-human transmission. The MERS epidemic that emerged in 2012 had a ~35% mortality, with dromedaries as the reservoir. Although CoVs with the same genome organization (e.g., Tylonycteris BatCoV HKU4 and Pipistrellus BatCoV HKU5) were also detected in bats, there is still a phylogenetic gap between these bat CoVs and MERS-CoV. In 2016, 10 years after the discovery of Rhinolophus BatCoV HKU2 in Chinese horseshoe bats, fatal swine disease outbreaks caused by this virus were reported in southern China. In late 2019, an outbreak of pneumonia emerged in Wuhan, China, and rapidly spread globally, leading to >4,000,000 fatalities so far. Although the genome of SARS-CoV-2 is highly similar to that of SARS-CoV, patient zero and the original source of the pandemic are still unknown. To protect humans from future public health threats, measures should be taken to monitor and reduce the chance of interspecies jumping events, either occurring naturally or through recombineering experiments.


Assuntos
COVID-19/virologia , Quirópteros/virologia , Infecções por Coronavirus/virologia , Coronavirus/fisiologia , Adaptação ao Hospedeiro , Síndrome Respiratória Aguda Grave/virologia , Alphacoronavirus/genética , Alphacoronavirus/fisiologia , Animais , COVID-19/transmissão , Coronavirus/genética , Infecções por Coronavirus/epidemiologia , Infecções por Coronavirus/transmissão , Infecções por Coronavirus/veterinária , Especificidade de Hospedeiro , Humanos , Coronavírus da Síndrome Respiratória do Oriente Médio/genética , Coronavírus da Síndrome Respiratória do Oriente Médio/fisiologia , Coronavírus Relacionado à Síndrome Respiratória Aguda Grave/genética , Coronavírus Relacionado à Síndrome Respiratória Aguda Grave/fisiologia , SARS-CoV-2/genética , SARS-CoV-2/fisiologia , Síndrome Respiratória Aguda Grave/epidemiologia , Síndrome Respiratória Aguda Grave/transmissão , Síndrome Respiratória Aguda Grave/veterinária
3.
Viruses ; 12(11)2020 11 19.
Artigo em Inglês | MEDLINE | ID: mdl-33228210

RESUMO

An outbreak of winter dysentery, complicated by severe respiratory syndrome, occurred in January 2020 in a high production dairy cow herd located in a hilly area of the Calabria region. Of the 52 animals belonging to the farm, 5 (9.6%) died with severe respiratory distress, death occurring 3-4 days after the appearance of the respiratory signs (caught and gasping breath). Microbiological analysis revealed absence of pathogenic bacteria whilst Real-time PCR identified the presence of RNA from Bovine Coronavirus (BCoV) in several organs: lungs, small intestine (jejunum), mediastinal lymph nodes, liver and placenta. BCoV was therefore hypothesized to play a role in the lethal pulmonary infection. Like the other CoVs, BCoV is able to cause different syndromes. Its role in calf diarrhea and in mild respiratory disease is well known: we report instead the involvement of this virus in a severe and fatal respiratory disorder, with symptoms and disease evolution resembling those of Severe Acute Respiratory Syndromes (SARS).


Assuntos
Doenças dos Bovinos/mortalidade , Infecções por Coronavirus/veterinária , Coronavirus Bovino/patogenicidade , Doenças Pulmonares Intersticiais/veterinária , Animais , Bovinos , Doenças dos Bovinos/virologia , Infecções por Coronavirus/mortalidade , Coronavirus Bovino/genética , Diarreia/virologia , Surtos de Doenças , Fezes/virologia , Feminino , Itália/epidemiologia , Doenças Pulmonares Intersticiais/mortalidade , Doenças Pulmonares Intersticiais/virologia , Filogenia , RNA Viral/genética , Infecções Respiratórias/mortalidade , Infecções Respiratórias/virologia , Síndrome Respiratória Aguda Grave/mortalidade , Síndrome Respiratória Aguda Grave/veterinária , Síndrome Respiratória Aguda Grave/virologia
4.
Zool Res ; 41(5): 503-516, 2020 Sep 18.
Artigo em Inglês | MEDLINE | ID: mdl-32772513

RESUMO

As of June 2020, Coronavirus Disease 2019 (COVID-19) has killed an estimated 440 000 people worldwide, 74% of whom were aged ≥65 years, making age the most significant risk factor for death caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection. To examine the effect of age on death, we established a SARS-CoV-2 infection model in Chinese rhesus macaques ( Macaca mulatta) of varied ages. Results indicated that infected young macaques manifested impaired respiratory function, active viral replication, severe lung damage, and infiltration of CD11b + and CD8 + cells in lungs at one-week post infection (wpi), but also recovered rapidly at 2 wpi. In contrast, aged macaques demonstrated delayed immune responses with a more severe cytokine storm, increased infiltration of CD11b + cells, and persistent infiltration of CD8 + cells in the lungs at 2 wpi. In addition, peripheral blood T cells from aged macaques showed greater inflammation and chemotaxis, but weaker antiviral functions than that in cells from young macaques. Thus, the delayed but more severe cytokine storm and higher immune cell infiltration may explain the poorer prognosis of older aged patients suffering SARS-CoV-2 infection.


Assuntos
Envelhecimento/imunologia , Betacoronavirus/imunologia , Infecções por Coronavirus/imunologia , Citocinas/imunologia , Macaca mulatta/imunologia , Pneumonia Viral/imunologia , Linfócitos T/imunologia , Fatores Etários , Envelhecimento/metabolismo , Animais , Betacoronavirus/fisiologia , COVID-19 , Infecções por Coronavirus/veterinária , Infecções por Coronavirus/virologia , Citocinas/metabolismo , Inflamação/imunologia , Inflamação/veterinária , Inflamação/virologia , Pulmão/imunologia , Pulmão/patologia , Pulmão/virologia , Macaca mulatta/virologia , Doenças dos Macacos/imunologia , Doenças dos Macacos/virologia , Pandemias/veterinária , Pneumonia Viral/veterinária , Pneumonia Viral/virologia , SARS-CoV-2 , Síndrome Respiratória Aguda Grave/imunologia , Síndrome Respiratória Aguda Grave/veterinária , Síndrome Respiratória Aguda Grave/virologia , Linfócitos T/metabolismo , Linfócitos T/patologia , Carga Viral/imunologia , Carga Viral/veterinária , Replicação Viral/imunologia
5.
Open Vet J ; 10(2): 164-177, 2020 08.
Artigo em Inglês | MEDLINE | ID: mdl-32821661

RESUMO

Viruses are having great time as they seem to have bogged humans down. Severe acute respiratory syndrome (SARS), Middle East respiratory syndrome (MERS), and novel coronavirus (COVID-19) are the three major coronaviruses of present-day global human and animal health concern. COVID-19 caused by SARS-CoV-2 is identified as the newest disease, presumably of bat origin. Different theories on the evolution of viruses are in circulation, yet there is no denying the fact that the animal source is the skeleton. The whole world is witnessing the terror of the COVID-19 pandemic that is following the same path of SARS and MERS, and seems to be more severe. In addition to humans, several species of animals are reported to have been infected with these life-threatening viruses. The possible routes of transmission and their zoonotic potentialities are the subjects of intense research. This review article aims to overview the link of all these three deadly coronaviruses among animals along with their phylogenic evolution and cross-species transmission. This is essential since animals as pets or food are said to pose some risk, and their better understanding is a must in order to prepare a possible plan for future havoc in both human and animal health. Although COVID-19 is causing a human health hazard globally, its reporting in animals are limited compared to SARS and MERS. Non-human primates and carnivores are most susceptible to SARS-coronavirus and SARS-CoV-2, respectively, whereas the dromedary camel is susceptible to MERS-coronavirus. Phylogenetically, the trio viruses are reported to have originated from bats and have special capacity to undergo mutation and genomic recombination in order to infect humans through its reservoir or replication host. However, it is difficult to analyze how the genomic pattern of coronaviruses occurs. Thus, increased possibility of new virus-variants infecting humans and animals in the upcoming days seems to be the biggest challenge for the future of the world. One health approach is portrayed as our best way ahead, and understanding the animal dimension will go a long way in formulating such preparedness plans.


Assuntos
Betacoronavirus/classificação , Infecções por Coronavirus/veterinária , Coronavírus da Síndrome Respiratória do Oriente Médio/classificação , Pandemias/veterinária , Pneumonia Viral/veterinária , Síndrome Respiratória Aguda Grave/veterinária , Coronavírus Relacionado à Síndrome Respiratória Aguda Grave/classificação , Animais , Animais Selvagens , Betacoronavirus/genética , COVID-19 , Camelídeos Americanos/virologia , Camelus/virologia , Gatos , Quirópteros/virologia , Infecções por Coronavirus/imunologia , Infecções por Coronavirus/transmissão , Suscetibilidade a Doenças/veterinária , Cães , Eutérios/virologia , Furões/virologia , Humanos , Leões/virologia , Coronavírus da Síndrome Respiratória do Oriente Médio/genética , Filogenia , Pneumonia Viral/imunologia , Pneumonia Viral/transmissão , Primatas/virologia , Cães Guaxinins/virologia , Coronavírus Relacionado à Síndrome Respiratória Aguda Grave/genética , SARS-CoV-2 , Síndrome Respiratória Aguda Grave/imunologia , Síndrome Respiratória Aguda Grave/transmissão , Serpentes/virologia , Tigres/virologia , Viverridae/virologia
6.
Vet Res Commun ; 44(3-4): 101-110, 2020 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-32651761

RESUMO

The outbreak of the SARS-CoV-2 in mainland China with subsequent human to human transmission worldwide had taken up the shape of a devastating pandemic. The ability of the virus to infect multiple species other than humans has currently been reported in experimental conditions. Non-human primates, felines, ferrets, rodents and host of other animals could previously be infected in experimental conditions with SARS-CoV and recently with SARS-CoV-2, both virus using Angiotensin-converting-enzyme 2 receptor for cellular entry. The variations in sequence homology of ACE2 receptor across species is identified as one of the factors determining virulence and pathogenicity in animals. The infection in experimental animals with SARS-CoV or SARS-CoV-2 on most occasions are asymptomatic, however, the virus could multiply within the respiratory tract and extra-pulmonary organs in most of the species. Here, we discuss about the pathogenicity, transmission, variations in angiotensin-converting-enzyme 2 receptor-binding across species and host pathogen interactions of SARS and SARS-CoV-2 in laboratory animals used in research.


Assuntos
Betacoronavirus/patogenicidade , Infecções por Coronavirus/veterinária , Interações Hospedeiro-Patógeno , Pandemias/veterinária , Pneumonia Viral/veterinária , Síndrome Respiratória Aguda Grave/veterinária , Coronavírus Relacionado à Síndrome Respiratória Aguda Grave/patogenicidade , Animais , COVID-19 , Callithrix/virologia , Gatos/virologia , Galinhas/virologia , Quirópteros/virologia , Chlorocebus aethiops/virologia , Infecções por Coronavirus/transmissão , Infecções por Coronavirus/virologia , Cricetinae/virologia , Furões/virologia , Macaca fascicularis/virologia , Macaca mulatta/virologia , Camundongos , Camundongos Endogâmicos/virologia , Pneumonia Viral/transmissão , Pneumonia Viral/virologia , Roedores/virologia , SARS-CoV-2 , Síndrome Respiratória Aguda Grave/transmissão , Síndrome Respiratória Aguda Grave/virologia , Suínos/virologia
7.
Infez Med ; 28(suppl 1): 71-83, 2020 Jun 01.
Artigo em Inglês | MEDLINE | ID: mdl-32532942

RESUMO

INTRODUCTION: Coronaviruses are zoonotic viruses that include human epidemic pathogens such as the Middle East Respiratory Syndrome virus (MERS-CoV), and the Severe Acute Respiratory Syndrome virus (SARS-CoV), among others (e.g., COVID-19, the recently emerging coronavirus disease). The role of animals as potential reservoirs for such pathogens remains an unanswered question. No systematic reviews have been published on this topic to date. METHODS: We performed a systematic literature review with meta-analysis, using three databases to assess MERS-CoV and SARS-CoV infection in animals and its diagnosis by serological and molecular tests. We performed a random-effects model meta-analysis to calculate the pooled prevalence and 95% confidence interval (95%CI). RESULTS: 6,493articles were retrieved (1960-2019). After screening by abstract/title, 50 articles were selected for full-text assessment. Of them, 42 were finally included for qualitative and quantitative analyses. From a total of 34 studies (n=20,896 animals), the pool prevalence by RT-PCR for MERS-CoV was 7.2% (95%CI 5.6-8.7%), with 97.3% occurring in camels, in which pool prevalence was 10.3% (95%CI 8.3-12.3). Qatar was the country with the highest MERS-CoV RT-PCR pool prevalence: 32.6% (95%CI 4.8-60.4%). From 5 studies and 2,618 animals, for SARS-CoV, the RT-PCR pool prevalence was 2.3% (95%CI 1.3-3.3). Of those, 38.35% were reported on bats, in which the pool prevalence was 14.1% (95%CI0.0-44.6%). DISCUSSION: A considerable proportion of infected animals tested positive, particularly by nucleic acid amplification tests (NAAT). This essential condition highlights the relevance of individual animals as reservoirs of MERS-CoV and SARS-CoV. In this meta-analysis, camels and bats were found to be positive by RT-PCR in over 10% of the cases for both; thus, suggesting their relevance in the maintenance of wild zoonotic transmission.


Assuntos
Animais Selvagens/virologia , Camelus/virologia , Quirópteros/virologia , Infecções por Coronavirus/veterinária , Coronavírus da Síndrome Respiratória do Oriente Médio/isolamento & purificação , Síndrome Respiratória Aguda Grave/veterinária , Coronavírus Relacionado à Síndrome Respiratória Aguda Grave/isolamento & purificação , Animais , Animais Domésticos/virologia , Anticorpos Antivirais/sangue , Infecções por Coronavirus/epidemiologia , Infecções por Coronavirus/transmissão , Estudos Transversais , Reservatórios de Doenças , Especificidade de Hospedeiro , Humanos , Coronavírus da Síndrome Respiratória do Oriente Médio/genética , Coronavírus da Síndrome Respiratória do Oriente Médio/imunologia , Prevalência , Doenças dos Primatas/epidemiologia , Doenças dos Primatas/virologia , Primatas/virologia , RNA Viral/sangue , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Doenças dos Roedores/epidemiologia , Doenças dos Roedores/virologia , Roedores/virologia , Coronavírus Relacionado à Síndrome Respiratória Aguda Grave/genética , Coronavírus Relacionado à Síndrome Respiratória Aguda Grave/imunologia , Testes Sorológicos , Síndrome Respiratória Aguda Grave/epidemiologia , Síndrome Respiratória Aguda Grave/transmissão , Zoonoses
9.
J Virol ; 93(24)2019 12 15.
Artigo em Inglês | MEDLINE | ID: mdl-31554686

RESUMO

Outbreaks of severe diarrhea in neonatal piglets in Guangdong, China, in 2017 resulted in the isolation and discovery of a novel swine enteric alphacoronavirus (SeACoV) derived from the species Rhinolophus bat coronavirus HKU2 (Y. Pan, X. Tian, P. Qin, B. Wang, et al., Vet Microbiol 211:15-21, 2017). SeACoV was later referred to as swine acute diarrhea syndrome CoV (SADS-CoV) by another group (P. Zhou, H. Fan, T. Lan, X.-L. Yang, et al., Nature 556:255-258, 2018). The present study was set up to investigate the potential species barriers of SADS-CoV in vitro and in vivo We first demonstrated that SADS-CoV possesses a broad species tropism and is able to infect cell lines from diverse species, including bats, mice, rats, gerbils, hamsters, pigs, chickens, nonhuman primates, and humans. Trypsin contributes to but is not essential for SADS-CoV propagation in vitro Furthermore, C57BL/6J mice were inoculated with the virus via oral or intraperitoneal routes. Although the mice exhibited only subclinical infection, they supported viral replication and prolonged infection in the spleen. SADS-CoV nonstructural proteins and double-stranded RNA were detected in splenocytes of the marginal zone on the edge of lymphatic follicles, indicating active replication of SADS-CoV in the mouse model. We identified that splenic dendritic cells (DCs) are the major targets of virus infection by immunofluorescence and flow cytometry approaches. Finally, we demonstrated that SADS-CoV does not utilize known CoV receptors for cellular entry. The ability of SADS-CoV to replicate in various cells lines from a broad range of species and the unexpected tropism for murine DCs provide important insights into the biology of this bat-origin CoV, highlighting its possible ability to cross interspecies barriers.IMPORTANCE Infections with bat-origin coronaviruses (CoVs) (severe acute respiratory syndrome CoV [SARS-CoV] and Middle East respiratory syndrome CoV [MERS-CoV]) have caused severe illness in humans after "host jump" events. Recently, a novel bat-HKU2-like CoV named swine acute diarrhea syndrome CoV (SADS-CoV) has emerged in southern China, causing lethal diarrhea in newborn piglets. It is important to assess the species barriers of SADS-CoV infection since the animal hosts (other than pigs and bats) and zoonotic potential are still unknown. An in vitro susceptibility study revealed a broad species tropism of SADS-CoV, including various rodent and human cell lines. We established a mouse model of SADS-CoV infection, identifying its active replication in splenic dendritic cells, which suggests that SADS-CoV has the potential to infect rodents. These findings highlight the potential cross-species transmissibility of SADS-CoV, although further surveillance in other animal populations is needed to fully understand the ecology of this bat-HKU2-origin CoV.


Assuntos
Alphacoronavirus/fisiologia , Quirópteros/virologia , Infecções por Coronavirus/transmissão , Infecções por Coronavirus/virologia , Infecção Hospitalar/virologia , Células Dendríticas/virologia , Diarreia/virologia , Síndrome Respiratória Aguda Grave/virologia , Alphacoronavirus/genética , Alphacoronavirus/patogenicidade , Animais , Linhagem Celular , Células Cultivadas , Galinhas , China/epidemiologia , Infecções por Coronavirus/epidemiologia , Diarreia/veterinária , Modelos Animais de Doenças , Feminino , Humanos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Coronavírus da Síndrome Respiratória do Oriente Médio/genética , Ratos , Coronavírus Relacionado à Síndrome Respiratória Aguda Grave/genética , Coronavírus Relacionado à Síndrome Respiratória Aguda Grave/crescimento & desenvolvimento , Síndrome Respiratória Aguda Grave/patologia , Síndrome Respiratória Aguda Grave/transmissão , Síndrome Respiratória Aguda Grave/veterinária , Especificidade da Espécie , Baço/patologia , Baço/virologia , Suínos , Internalização do Vírus , Replicação Viral
10.
Nature ; 556(7700): 255-258, 2018 04.
Artigo em Inglês | MEDLINE | ID: mdl-29618817

RESUMO

Cross-species transmission of viruses from wildlife animal reservoirs poses a marked threat to human and animal health 1 . Bats have been recognized as one of the most important reservoirs for emerging viruses and the transmission of a coronavirus that originated in bats to humans via intermediate hosts was responsible for the high-impact emerging zoonosis, severe acute respiratory syndrome (SARS) 2-10 . Here we provide virological, epidemiological, evolutionary and experimental evidence that a novel HKU2-related bat coronavirus, swine acute diarrhoea syndrome coronavirus (SADS-CoV), is the aetiological agent that was responsible for a large-scale outbreak of fatal disease in pigs in China that has caused the death of 24,693 piglets across four farms. Notably, the outbreak began in Guangdong province in the vicinity of the origin of the SARS pandemic. Furthermore, we identified SADS-related CoVs with 96-98% sequence identity in 9.8% (58 out of 591) of anal swabs collected from bats in Guangdong province during 2013-2016, predominantly in horseshoe bats (Rhinolophus spp.) that are known reservoirs of SARS-related CoVs. We found that there were striking similarities between the SADS and SARS outbreaks in geographical, temporal, ecological and aetiological settings. This study highlights the importance of identifying coronavirus diversity and distribution in bats to mitigate future outbreaks that could threaten livestock, public health and economic growth.


Assuntos
Alphacoronavirus/isolamento & purificação , Alphacoronavirus/patogenicidade , Doenças dos Animais/epidemiologia , Doenças dos Animais/virologia , Quirópteros/virologia , Infecções por Coronavirus/veterinária , Diarreia/veterinária , Suínos/virologia , Alphacoronavirus/classificação , Alphacoronavirus/genética , Doenças dos Animais/transmissão , Animais , Biodiversidade , China/epidemiologia , Infecções por Coronavirus/epidemiologia , Infecções por Coronavirus/transmissão , Diarreia/patologia , Diarreia/virologia , Reservatórios de Doenças/veterinária , Reservatórios de Doenças/virologia , Genoma Viral/genética , Humanos , Jejuno/patologia , Jejuno/virologia , Filogenia , Síndrome Respiratória Aguda Grave/epidemiologia , Síndrome Respiratória Aguda Grave/veterinária , Síndrome Respiratória Aguda Grave/virologia , Análise Espaço-Temporal , Zoonoses/epidemiologia , Zoonoses/transmissão , Zoonoses/virologia
12.
Zoonoses Public Health ; 63(8): 608-615, 2016 12.
Artigo em Inglês | MEDLINE | ID: mdl-27178103

RESUMO

Bats have been demonstrated to be natural reservoirs of severe acute respiratory syndrome coronavirus (SARS CoV) and Middle East respiratory syndrome (MERS) CoV. Faecal samples from 248 individuals of 20 bat species were tested for partial RNA-dependent RNA polymerase gene of CoV and 57 faecal samples from eight bat species were tested positive. The highest detection rate of 44% for Scotophilus kuhlii, followed by 30% for Rhinolophus monoceros. Significantly higher detection rates of coronaviral RNA were found in female bats and Scotophilus kuhlii roosting in palm trees. Phylogenetic analysis classified the positive samples into SARS-related (SARSr) CoV, Scotophilus bat CoV 512 close to those from China and Philippines, and Miniopterus bat CoV 1A-related lineages. Coronaviral RNA was also detected in bat guano from Scotophilus kuhlii and Myotis formosus flavus on the ground and had potential risk for human exposure. Diverse bat CoV with zoonotic potential could be introduced by migratory bats and maintained in the endemic bat population in Taiwan.


Assuntos
Quirópteros/virologia , Síndrome Respiratória Aguda Grave/veterinária , Coronavírus Relacionado à Síndrome Respiratória Aguda Grave/isolamento & purificação , Animais , Fezes/virologia , Feminino , Masculino , Filogenia , RNA Viral/isolamento & purificação , RNA Polimerase Dependente de RNA/genética , Fatores de Risco , Coronavírus Relacionado à Síndrome Respiratória Aguda Grave/classificação , Coronavírus Relacionado à Síndrome Respiratória Aguda Grave/genética , Síndrome Respiratória Aguda Grave/epidemiologia , Especificidade da Espécie , Taiwan , Zoonoses
13.
Ecohealth ; 13(1): 72-82, 2016 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-27048154

RESUMO

Following the SARS outbreak, extensive surveillance was undertaken globally to detect and identify coronavirus diversity in bats. This study sought to identify the diversity and prevalence of coronaviruses in bats in the Australasian region. We identified four different genotypes of coronavirus, three of which (an alphacoronavirus and two betacoronaviruses) are potentially new species, having less than 90% nucleotide sequence identity with the most closely related described viruses. We did not detect any SARS-like betacoronaviruses, despite targeting rhinolophid bats, the putative natural host taxa. Our findings support the virus-host co-evolution hypothesis, with the detection of Miniopterus bat coronavirus HKU8 (previously reported in Miniopterus species in China, Hong Kong and Bulgaria) in Australian Miniopterus species. Similarly, we detected a novel betacoronavirus genotype from Pteropus alecto which is most closely related to Bat coronavirus HKU9 identified in other pteropodid bats in China, Kenya and the Philippines. We also detected possible cross-species transmission of bat coronaviruses, and the apparent enteric tropism of these viruses. Thus, our findings are consistent with a scenario wherein the current diversity and host specificity of coronaviruses reflects co-evolution with the occasional host shift.


Assuntos
Quirópteros/virologia , Infecções por Coronavirus/virologia , Coronavirus/isolamento & purificação , Animais , Australásia/epidemiologia , Sequência de Bases , Coronavirus/genética , Infecções por Coronavirus/epidemiologia , Infecções por Coronavirus/veterinária , Genoma Viral , Genótipo , Filogenia , Reação em Cadeia da Polimerase , Prevalência , RNA Viral/genética , Síndrome Respiratória Aguda Grave/veterinária , Taiwan/epidemiologia
14.
Virol Sin ; 31(1): 31-40, 2016 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-26920708

RESUMO

Since the 2002-2003 severe acute respiratory syndrome (SARS) outbreak prompted a search for the natural reservoir of the SARS coronavirus, numerous alpha- and betacoronaviruses have been discovered in bats around the world. Bats are likely the natural reservoir of alpha- and betacoronaviruses, and due to the rich diversity and global distribution of bats, the number of bat coronaviruses will likely increase. We conducted a surveillance of coronaviruses in bats in an abandoned mineshaft in Mojiang County, Yunnan Province, China, from 2012-2013. Six bat species were frequently detected in the cave: Rhinolophus sinicus, Rhinolophus affinis, Hipposideros pomona, Miniopterus schreibersii, Miniopterus fuliginosus, and Miniopterus fuscus. By sequencing PCR products of the coronavirus RNA-dependent RNA polymerase gene (RdRp), we found a high frequency of infection by a diverse group of coronaviruses in different bat species in the mineshaft. Sequenced partial RdRp fragments had 80%-99% nucleic acid sequence identity with well-characterized Alphacoronavirus species, including BtCoV HKU2, BtCoV HKU8, and BtCoV1, and unassigned species BtCoV HKU7 and BtCoV HKU10. Additionally, the surveillance identified two unclassified betacoronaviruses, one new strain of SARS-like coronavirus, and one potentially new betacoronavirus species. Furthermore, coronavirus co-infection was detected in all six bat species, a phenomenon that fosters recombination and promotes the emergence of novel virus strains. Our findings highlight the importance of bats as natural reservoirs of coronaviruses and the potentially zoonotic source of viral pathogens.


Assuntos
Quirópteros/virologia , Infecções por Coronavirus/virologia , Coronavirus/isolamento & purificação , Sequência de Aminoácidos , Animais , Sequência de Bases , China/epidemiologia , Coronavirus/genética , Infecções por Coronavirus/epidemiologia , Infecções por Coronavirus/veterinária , Genoma Viral , Filogenia , Reação em Cadeia da Polimerase/métodos , RNA Viral/genética , RNA Polimerase Dependente de RNA/genética , Coronavírus Relacionado à Síndrome Respiratória Aguda Grave/genética , Coronavírus Relacionado à Síndrome Respiratória Aguda Grave/isolamento & purificação , Análise de Sequência de DNA , Síndrome Respiratória Aguda Grave/epidemiologia , Síndrome Respiratória Aguda Grave/genética , Síndrome Respiratória Aguda Grave/veterinária
16.
Nature ; 503(7477): 535-8, 2013 Nov 28.
Artigo em Inglês | MEDLINE | ID: mdl-24172901

RESUMO

The 2002-3 pandemic caused by severe acute respiratory syndrome coronavirus (SARS-CoV) was one of the most significant public health events in recent history. An ongoing outbreak of Middle East respiratory syndrome coronavirus suggests that this group of viruses remains a key threat and that their distribution is wider than previously recognized. Although bats have been suggested to be the natural reservoirs of both viruses, attempts to isolate the progenitor virus of SARS-CoV from bats have been unsuccessful. Diverse SARS-like coronaviruses (SL-CoVs) have now been reported from bats in China, Europe and Africa, but none is considered a direct progenitor of SARS-CoV because of their phylogenetic disparity from this virus and the inability of their spike proteins to use the SARS-CoV cellular receptor molecule, the human angiotensin converting enzyme II (ACE2). Here we report whole-genome sequences of two novel bat coronaviruses from Chinese horseshoe bats (family: Rhinolophidae) in Yunnan, China: RsSHC014 and Rs3367. These viruses are far more closely related to SARS-CoV than any previously identified bat coronaviruses, particularly in the receptor binding domain of the spike protein. Most importantly, we report the first recorded isolation of a live SL-CoV (bat SL-CoV-WIV1) from bat faecal samples in Vero E6 cells, which has typical coronavirus morphology, 99.9% sequence identity to Rs3367 and uses ACE2 from humans, civets and Chinese horseshoe bats for cell entry. Preliminary in vitro testing indicates that WIV1 also has a broad species tropism. Our results provide the strongest evidence to date that Chinese horseshoe bats are natural reservoirs of SARS-CoV, and that intermediate hosts may not be necessary for direct human infection by some bat SL-CoVs. They also highlight the importance of pathogen-discovery programs targeting high-risk wildlife groups in emerging disease hotspots as a strategy for pandemic preparedness.


Assuntos
Quirópteros/virologia , Peptidil Dipeptidase A/metabolismo , Coronavírus Relacionado à Síndrome Respiratória Aguda Grave/isolamento & purificação , Coronavírus Relacionado à Síndrome Respiratória Aguda Grave/metabolismo , Enzima de Conversão de Angiotensina 2 , Animais , China , Chlorocebus aethiops , Reservatórios de Doenças/virologia , Fezes/virologia , Imunofluorescência , Genoma Viral/genética , Especificidade de Hospedeiro , Humanos , Dados de Sequência Molecular , Pandemias/prevenção & controle , Pandemias/veterinária , Peptidil Dipeptidase A/genética , Reação em Cadeia da Polimerase em Tempo Real , Receptores Virais/genética , Receptores Virais/metabolismo , Coronavírus Relacionado à Síndrome Respiratória Aguda Grave/genética , Coronavírus Relacionado à Síndrome Respiratória Aguda Grave/ultraestrutura , Síndrome Respiratória Aguda Grave/prevenção & controle , Síndrome Respiratória Aguda Grave/transmissão , Síndrome Respiratória Aguda Grave/veterinária , Síndrome Respiratória Aguda Grave/virologia , Especificidade da Espécie , Glicoproteína da Espícula de Coronavírus/química , Glicoproteína da Espícula de Coronavírus/metabolismo , Células Vero , Vírion/isolamento & purificação , Vírion/ultraestrutura , Internalização do Vírus , Viverridae/metabolismo
17.
J Virol ; 85(8): 4025-30, 2011 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-21289121

RESUMO

The shedding of severe acute respiratory syndrome coronavirus (SARS-CoV) into saliva droplets plays a critical role in viral transmission. The source of high viral loads in saliva, however, remains elusive. Here we investigate the early target cells of infection in the entire array of respiratory tissues in Chinese macaques after intranasal inoculations with a single-cycle pseudotyped virus and a pathogenic SARS-CoV. We found that angiotensin-converting enzyme 2-positive (ACE2(+)) cells were widely distributed in the upper respiratory tract, and ACE2(+) epithelial cells lining salivary gland ducts were the early target cells productively infected. Our findings also have implications for SARS-CoV early diagnosis and prevention.


Assuntos
Células Epiteliais/virologia , Ductos Salivares/virologia , Síndrome Respiratória Aguda Grave/veterinária , Síndrome Respiratória Aguda Grave/virologia , Coronavírus Relacionado à Síndrome Respiratória Aguda Grave/isolamento & purificação , Enzima de Conversão de Angiotensina 2 , Animais , Imuno-Histoquímica , Macaca mulatta , Microscopia , Peptidil Dipeptidase A/análise , Receptores Virais/análise , Coronavírus Relacionado à Síndrome Respiratória Aguda Grave/patogenicidade , Carga Viral
18.
Emerg Infect Dis ; 15(3): 482-5, 2009 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-19239771
19.
J Virol ; 82(14): 6984-91, 2008 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-18448527

RESUMO

It is believed that a novel coronavirus, severe acute respiratory syndrome coronavirus (SARS-CoV), was passed from palm civets to humans and caused the epidemic of SARS in 2002 to 2003. The major species barriers between humans and civets for SARS-CoV infections are the specific interactions between a defined receptor-binding domain (RBD) on a viral spike protein and its host receptor, angiotensin-converting enzyme 2 (ACE2). In this study a chimeric ACE2 bearing the critical N-terminal helix from civet and the remaining peptidase domain from human was constructed, and it was shown that this construct has the same receptor activity as civet ACE2. In addition, crystal structures of the chimeric ACE2 complexed with RBDs from various human and civet SARS-CoV strains were determined. These structures, combined with a previously determined structure of human ACE2 complexed with the RBD from a human SARS-CoV strain, have revealed a structural basis for understanding the major species barriers between humans and civets for SARS-CoV infections. They show that the major species barriers are determined by interactions between four ACE2 residues (residues 31, 35, 38, and 353) and two RBD residues (residues 479 and 487), that early civet SARS-CoV isolates were prevented from infecting human cells due to imbalanced salt bridges at the hydrophobic virus/receptor interface, and that SARS-CoV has evolved to gain sustained infectivity for human cells by eliminating unfavorable free charges at the interface through stepwise mutations at positions 479 and 487. These results enhance our understanding of host adaptations and cross-species infections of SARS-CoV and other emerging animal viruses.


Assuntos
Glicoproteínas de Membrana/química , Glicoproteínas de Membrana/metabolismo , Peptidil Dipeptidase A/química , Peptidil Dipeptidase A/metabolismo , Síndrome Respiratória Aguda Grave/virologia , Coronavírus Relacionado à Síndrome Respiratória Aguda Grave/crescimento & desenvolvimento , Proteínas do Envelope Viral/química , Proteínas do Envelope Viral/metabolismo , Viverridae/virologia , Enzima de Conversão de Angiotensina 2 , Animais , Cristalografia por Raios X , Humanos , Glicoproteínas de Membrana/genética , Modelos Moleculares , Peptidil Dipeptidase A/genética , Estrutura Quaternária de Proteína , Receptores Virais/química , Receptores Virais/genética , Receptores Virais/metabolismo , Recombinação Genética , Síndrome Respiratória Aguda Grave/veterinária , Especificidade da Espécie , Glicoproteína da Espícula de Coronavírus , Proteínas do Envelope Viral/genética , Ligação Viral
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA