Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 128
Filter
1.
Viruses ; 14(2)2022 01 18.
Article in English | MEDLINE | ID: mdl-35215769

ABSTRACT

Bats have been recognized as an exceptional viral reservoir, especially for coronaviruses. At least three bat zoonotic coronaviruses (SARS-CoV, MERS-CoV and SARS-CoV-2) have been shown to cause severe diseases in humans and it is expected more will emerge. One of the major features of CoVs is that they are all highly prone to recombination. An extreme example is the insertion of the P10 gene from reoviruses in the bat CoV GCCDC1, first discovered in Rousettus leschenaultii bats in China. Here, we report the detection of GCCDC1 in four different bat species (Eonycteris spelaea, Cynopterus sphinx, Rhinolophus shameli and Rousettus sp.) in Cambodia. This finding demonstrates a much broader geographic and bat species range for this virus and indicates common cross-species transmission. Interestingly, one of the bat samples showed a co-infection with an Alpha CoV most closely related to RsYN14, a virus recently discovered in the same genus (Rhinolophus) of bat in Yunnan, China, 2020. Taken together, our latest findings highlight the need to conduct active surveillance in bats to assess the risk of emerging CoVs, especially in Southeast Asia.


Subject(s)
Chiroptera/virology , Coronaviridae Infections/veterinary , Coronaviridae/classification , Coronaviridae/genetics , Disease Reservoirs/veterinary , Disease Reservoirs/virology , Phylogeography , Recombination, Genetic , Animals , Cambodia/epidemiology , China/epidemiology , Chiroptera/classification , Coronaviridae/isolation & purification , Coronaviridae Infections/epidemiology , Coronaviridae Infections/transmission , Evolution, Molecular , Genome, Viral , Phylogeny
2.
Transbound Emerg Dis ; 69(4): 2384-2389, 2022 Jul.
Article in English | MEDLINE | ID: mdl-33977671

ABSTRACT

The vampire bat (Desmodus rotundus) is a haematophagous animal that feeds exclusively on the blood of domestic mammals. Vampire bat feeding habits enable their contact with mammalian hosts and may enhance zoonotic spillover. Moreover, they may carry several pathogenic organisms, including coronaviruses (CoVs), for which they are important hosts. The human pathogens that cause severe acute respiratory syndrome (SARS-CoV), Middle East respiratory syndrome (MERS-CoV) and possibly coronavirus disease 2019 (SARS-CoV-2) all originated in bats but required bridge hosts to spread into human populations. To monitor the presence of potential zoonotic viruses in bats, the present work evaluated the presence of CoVs in vampire bats from southern Brazil. A total of 101 vampire bats were captured and euthanized between 2017 and 2019 in Rio Grande do Sul state, southern Brazil. The brain, heart, liver, lungs, kidneys and intestines were collected and macerated individually. The samples were pooled and submitted to high-throughput sequencing (HTS) using the Illumina MiSeq platform and subsequently individually screened using a pancoronavirus RT-PCR protocol. We detected CoV-related sequences in HTS, but only two (2/101; 1.98%) animals had CoV detected in the intestines by RT-PCR. Partial sequences of RdRp and spike genes were obtained in the same sample and the RdRp region in the other sample. The sequences were classified as belonging to Alphacoronavirus. The sequences were closely related to alphacoronaviruses detected in vampire bats from Peru. The continuous monitoring of bat CoVs may help to map and predict putative future zoonotic agents with great impacts on human health.


Subject(s)
Chiroptera , Coronaviridae , Animals , Brazil/epidemiology , Chiroptera/virology , Coronaviridae/classification , Coronaviridae/isolation & purification , Phylogeny , RNA-Dependent RNA Polymerase
3.
Transbound Emerg Dis ; 69(4): 1748-1760, 2022 Jul.
Article in English | MEDLINE | ID: mdl-33966351

ABSTRACT

Highly pathogenic coronaviruses, including SARS-CoV-2, SARS-CoV and MERS-CoV, are thought to be transmitted from bats to humans, but the viral genetic signatures that contribute to bat-to-human transmission remain largely obscure. In this study, we identified an identical ribosomal frameshift motif among the three bat-human pairs of viruses and strong purifying selection after jumping from bats to humans. This represents genetic signatures of coronaviruses that are related to bat-to-human transmission. To further trace the early human-to-human transmission of SARS-CoV-2 in North America, a geographically stratified genome-wide association study (North American isolates and the remaining isolates) and a retrospective study were conducted. We determined that the single nucleotide polymorphisms (SNPs) 1,059.C > T and 25,563.G > T were significantly associated with approximately half of the North American SARS-CoV-2 isolates that accumulated largely during March 2020. Retrospectively tracing isolates with these two SNPs was used to reconstruct the early, reliable transmission history of North American SARS-CoV-2, and European isolates (February 26, 2020) showed transmission 3 days earlier than North American isolates and 17 days earlier than Asian isolates. Collectively, we identified the genetic signatures of the three pairs of coronaviruses and reconstructed an early transmission history of North American SARS-CoV-2. We envision that these genetic signatures are possibly diagnosable and predic markers for public health surveillance.


Subject(s)
COVID-19 , Chiroptera , Coronaviridae , Animals , COVID-19/transmission , COVID-19/veterinary , Chiroptera/virology , Coronaviridae/classification , Coronaviridae/genetics , Genome, Viral , Genome-Wide Association Study/veterinary , Humans , North America , Phylogeny , Polymorphism, Single Nucleotide , Retrospective Studies , SARS-CoV-2/genetics
4.
Infect Genet Evol ; 95: 105075, 2021 11.
Article in English | MEDLINE | ID: mdl-34509646

ABSTRACT

T-cell-mediated immunity to SARS-CoV-2-derived peptides in individuals unexposed to SARS-CoV-2 has been previously reported. This pre-existing immunity was suggested to largely derive from prior exposure to 'common cold' endemic human coronaviruses (HCoVs). To test this, we characterised the sequence homology of SARS-CoV-2-derived T-cell epitopes reported in the literature across the full proteome of the Coronaviridae family. 54.8% of these epitopes had no homology to any of the HCoVs. Further, the proportion of SARS-CoV-2-derived epitopes with any level of sequence homology to the proteins encoded by any of the coronaviruses tested is well-predicted by their alignment-free phylogenetic distance to SARS-CoV-2 (Pearson's r = -0.958). No coronavirus in our dataset showed a significant excess of T-cell epitope homology relative to the proportion of expected random matches, given their genetic similarity to SARS-CoV-2. Our findings suggest that prior exposure to human or animal-associated coronaviruses cannot completely explain the T-cell repertoire in unexposed individuals that recognise SARS-CoV-2 cross-reactive epitopes.


Subject(s)
Antibodies, Viral/blood , COVID-19/immunology , Coronaviridae/immunology , Disease Resistance , Immunologic Memory , SARS-CoV-2/immunology , Animals , Antibodies, Viral/genetics , Antibodies, Viral/immunology , Antigens, Viral/genetics , Antigens, Viral/immunology , Asymptomatic Diseases , COVID-19/genetics , COVID-19/pathology , COVID-19/virology , Chiroptera/virology , Coronaviridae/classification , Coronaviridae/genetics , Coronaviridae/pathogenicity , Cross Reactions , Epitopes, T-Lymphocyte/genetics , Epitopes, T-Lymphocyte/immunology , Eutheria/virology , Humans , Immunity, Cellular , Phylogeny , SARS-CoV-2/classification , SARS-CoV-2/genetics , SARS-CoV-2/pathogenicity , Severity of Illness Index , T-Lymphocytes/immunology , T-Lymphocytes/virology
5.
Sci Rep ; 11(1): 16145, 2021 08 09.
Article in English | MEDLINE | ID: mdl-34373516

ABSTRACT

The genetic element s2m has been acquired through horizontal transfer by many distantly related viruses, including the SARS-related coronaviruses. Here we show that s2m is evolutionarily conserved in these viruses. Though several lineages of severe acute respiratory syndrome coronavirus 2 (SARS­CoV­2) devoid of the element can be found, these variants seem to have been short lived, indicating that they were less evolutionary fit than their s2m-containing counterparts. On a species-level, however, there do not appear to be any losses and this pattern strongly suggests that the s2m element is essential to virus replication in SARS-CoV-2 and related viruses. Further experiments are needed to elucidate the function of s2m.


Subject(s)
Coronaviridae/genetics , Interspersed Repetitive Sequences/genetics , RNA, Viral/genetics , SARS-CoV-2/genetics , Virus Replication/genetics , Animals , Base Sequence , COVID-19/virology , Coronaviridae/classification , Evolution, Molecular , Gene Transfer, Horizontal , Humans , Phylogeny , SARS-CoV-2/physiology , Sequence Homology, Nucleic Acid , Species Specificity
6.
Virology ; 563: 20-27, 2021 11.
Article in English | MEDLINE | ID: mdl-34411808

ABSTRACT

Viruses of the subfamily Orthocoronavirinae can cause mild to severe disease in people, including COVID-19, MERS and SARS. Their most common natural hosts are bat and bird species, which are mostly split across four virus genera. Molecular clock analyses of orthocoronaviruses suggested the most recent common ancestor of these viruses might have emerged either around 10,000 years ago or, using models accounting for selection, many millions of years. Here, we reassess the evolutionary history of these viruses. We present time-aware phylogenetic analyses of a RNA-dependent RNA polymerase locus from 123 orthocoronaviruses isolated from birds and bats, including those in New Zealand, which were geographically isolated from other bats around 35 million years ago. We used this age, as well as the age of the avian-mammals split, to calibrate the molecular clocks, under the assumption that these ages are applicable to the analyzed viruses. We found that the time to the most recent ancestor common for all orthocoronaviruses is likely 150 or more million years, supporting clock analyses that account for selection.


Subject(s)
Birds/virology , Chiroptera/virology , Coronaviridae Infections/virology , Coronaviridae , Genome, Viral , Animals , Coronaviridae/classification , Coronaviridae/genetics , Evolution, Molecular , New Zealand/epidemiology
7.
Int Rev Immunol ; 40(1-2): 5-53, 2021.
Article in English | MEDLINE | ID: mdl-32744465

ABSTRACT

Coronavirus infections are responsible for mild, moderate, and severe infections in birds and mammals. These were first isolated in humans as causal microorganisms responsible for common cold. The 2002-2003 SARS epidemic caused by SARS-CoV and 2012 MERS epidemic (64 countries affected) caused by MERS-CoV showed their acute and fatal side. These two CoV infections killed thousands of patients infected worldwide. However, WHO has still reported the MERS case in December 2019 in middle-eastern country (Saudi Arabia), indicating the MERS epidemic has not ended completely yet. Although we have not yet understood completely these two CoV epidemics, a third most dangerous and severe CoV infection has been originated in the Wuhan city, Hubei district of China in December 2019. This CoV infection called COVID-19 or SARS-CoV2 infection has now spread to 210 countries and territories around the world. COVID-19 has now been declared a pandemic by the World Health Organization (WHO). It has infected more than 16.69 million people with more than 663,540 deaths across the world. Thus the current manuscript aims to describe all three (SARS, MERS, and COVID-19) in terms of their causal organisms (SARS-CoV, MERS-CoV, and SARS-CoV2), similarities and differences in their clinical symptoms, outcomes, immunology, and immunopathogenesis, and possible future therapeutic approaches.


Subject(s)
COVID-19/pathology , Coronaviridae/ultrastructure , Middle East Respiratory Syndrome Coronavirus/immunology , SARS-CoV-2/immunology , Severe Acute Respiratory Syndrome/pathology , Severe acute respiratory syndrome-related coronavirus/immunology , Animals , COVID-19/diagnosis , COVID-19/mortality , Camelus/virology , Chiroptera/virology , Coronaviridae/classification , Disease Reservoirs/virology , Disease Susceptibility/virology , Humans , Middle East Respiratory Syndrome Coronavirus/pathogenicity , Severe acute respiratory syndrome-related coronavirus/pathogenicity , SARS-CoV-2/pathogenicity , Severe Acute Respiratory Syndrome/diagnosis , Severe Acute Respiratory Syndrome/mortality , Virus Replication/physiology
8.
Transbound Emerg Dis ; 68(3): 987-992, 2021 May.
Article in English | MEDLINE | ID: mdl-32725795

ABSTRACT

The novel SARS-CoV-2 coronavirus has attracted attention due to the high number of human cases around the world. It has been proposed that this virus originated in bats, possibly transmitted to humans by an intermediate host, making bats a group of great interest during this outbreak. Almost 10% of the world's bat species inhabit Mexico, and 14 previous novel CoVs have been recorded in Mexican bats. However, the phylogenetic relationships between these viruses and the novel coronavirus are unknown. The aim of this communication was therefore to describe the phylogenetic relationships between Mexican bat-CoVs and SARS-CoV-2. We showed that Mexican bat-CoVs sequences are grouped into two genera, Alphacoronavirus and Betacoronavirus, and the new coronavirus is an independent clade within Betacoronavirus. Due to the diversity of CoVs in Mexican bats, the propensity of CoVs to shift hosts, the invasion mechanisms described for this new virus, and previous reports of animals infected by SARS-CoV-2, the risk of possible infection from humans to Mexican bats should not be discarded and warrants further analyses. To avoid future zoonotic infectious diseases and to limit persecution of bats, we urge researchers and the general population to take extreme precautions and avoid manipulation of bats during the current and future similar outbreaks.


Subject(s)
COVID-19/virology , Chiroptera/virology , SARS-CoV-2/genetics , Alphacoronavirus/classification , Alphacoronavirus/genetics , Animals , COVID-19/epidemiology , Communicable Diseases, Emerging/virology , Coronaviridae/classification , Coronaviridae/genetics , Evolution, Molecular , Genome, Viral , Humans , Mexico/epidemiology , Phylogeny , SARS-CoV-2/classification , Zoonoses/epidemiology
9.
Virus Res ; 290: 198175, 2020 12.
Article in English | MEDLINE | ID: mdl-33007342

ABSTRACT

The outbreak and spread of new strains of coronavirus (SARS-CoV-2) remain a global threat with increasing cases in affected countries. The evolutionary tree of SARS-CoV-2 revealed that Porcine Reproductive and Respiratory Syndrome virus 2, which belongs to the Beta arterivirus genus from the Arteriviridae family is possibly the most ancient ancestral origin of SARS-CoV-2 and other Coronaviridae. This review focuses on phylogenomic distribution and evolutionary lineage of zoonotic viral cross-species transmission of the Coronaviridae family and the implications of bat microbiome in zoonotic viral transmission and infection. The review also casts light on the role of the human microbiome in predicting and controlling viral infections. The significance of microbiome-mediated interventions in the treatment of viral infections is also discussed. Finally, the importance of synthetic viruses in the study of viral evolution and transmission is highlighted.


Subject(s)
Biological Evolution , Coronaviridae Infections/transmission , Coronaviridae/genetics , Microbiota , Zoonoses/transmission , Animals , COVID-19/transmission , COVID-19/virology , Chiroptera/virology , Coronaviridae/classification , Coronaviridae/physiology , Coronaviridae Infections/virology , Genome, Viral/genetics , Humans , Phylogeny , SARS-CoV-2/classification , SARS-CoV-2/genetics , SARS-CoV-2/physiology , Zoonoses/virology
10.
Arch Virol ; 165(10): 2341-2348, 2020 Oct.
Article in English | MEDLINE | ID: mdl-32737584

ABSTRACT

Much remains unknown concerning the origin of the novel pandemic coronavirus that has raged across the globe since emerging in Wuhan of Hubei province, near the center of the People's Republic of China, in December of 2019. All current members of the family Coronaviridae have arisen by a combination of incremental adaptive mutations, against the backdrop of many recombinational events throughout the past, rendering each a unique mosaic of RNA sequences from diverse sources. The consensus among virologists is that the base sequence of the novel coronavirus, designated SARS-CoV-2, was derived from a common ancestor of a bat coronavirus, represented by the strain RaTG13, isolated in Yunnan province in 2013. Into that ancestral genetic background, several recombination events have since occurred from other divergent bat-derived coronaviruses, resulting in localized discordance between the two. One such event left SARS-CoV-2 with a receptor binding domain (RBD) capable of binding the human ACE-2 receptor lacking in RaTG13, and a second event uniquely added to SARS-CoV-2 a site specific for furin, capable of efficient endoproteolytic cleavage and activation of the spike glycoprotein responsible for virus entry and cell fusion. This paper demonstrates by bioinformatic analysis that such recombinational events are facilitated by short oligonucleotide "breakpoint sequences", similar to CAGAC, that direct recombination naturally to certain positions in the genome at the boundaries between blocks of RNA code and potentially RNA structure. This "breakpoint sequence hypothesis" provides a natural explanation for the biogenesis of SARS-CoV-2 over time and in the wild.


Subject(s)
Betacoronavirus/genetics , Coronavirus Infections/virology , Inverted Repeat Sequences , Pneumonia, Viral/virology , RNA, Viral/genetics , Amino Acid Sequence , Animals , Base Sequence , Betacoronavirus/classification , COVID-19 , China/epidemiology , Chiroptera/virology , Coronaviridae/classification , Coronaviridae/genetics , Coronavirus Infections/epidemiology , Evolution, Molecular , Genome, Viral , Host Microbial Interactions/genetics , Humans , Pandemics , Phylogeny , Pneumonia, Viral/epidemiology , Recombination, Genetic , SARS-CoV-2 , Sequence Alignment
11.
Med Sci (Paris) ; 36(8-9): 797-802, 2020.
Article in French | MEDLINE | ID: mdl-32755538

ABSTRACT

SARS-CoV-2 (severe acute respiratory syndrome-coronavirus-2, which emerged in China at the end of 2019, is responsible for a global health crisis resulting in the confinement of more than 3 billion people worldwide and the sharp decline of the world economy. In this context, a race against the clock is launched in order to develop a treatment to stop the pandemic as soon as possible. A study published in Nature by the Volker Thiel team reports the development of reverse genetics for SARS-CoV-2 allowing them to recreate the virus in just a few weeks. The perspectives of this work are very interesting since it will allow the genetic manipulation of the virus and thus the development of precious tools which will be useful to fight the infection. Even though this approach represents a technological leap that will improve our knowledge of the virus, it also carries the germ of possible misuse and the creation of the virus for malicious purposes. The advantages and disadvantages of recreating SARS-CoV-2 in this pandemic period are discussed in this mini-synthesis.


TITLE: Une course contre la montre - Création du SARS-CoV-2 en laboratoire, un mois après son émergence ! ABSTRACT: Le SARS-CoV-2 (severe acute respiratory syndrome-coronavirus-2), qui a émergé à la fin de l'année 2019 en République populaire de Chine, est responsable d'une crise sanitaire mondiale qui a entraîné le confinement de plus de 3 milliards d'individus et l'arrêt brutal de l'économie planétaire. Dans ce contexte, une course contre la montre est lancée afin de développer, dans les plus brefs délais, un traitement permettant d'enrayer la pandémie. Une étude de l'équipe de Volker Thiel, parue dans le journal Nature, rapporte la mise au point d'une technique de génétique inverse pour le SARS-CoV-2, leur ayant permis de recréer le virus en seulement quelques semaines. Les perspectives de ces travaux sont très intéressantes puisqu'elles permettent d'envisager la manipulation génétique du virus et ainsi le développement d'outils précieux qui seront utiles pour combattre l'infection. Si la technique représente également un saut technologique qui permettra d'améliorer nos connaissances sur le virus, elle porte aussi en elle le germe d'un possible mésusage et la création d'un virus à des fins malveillantes. Les avantages et inconvénients de recréer le SARS-CoV-2 dans cette période de pandémie sont discutés dans cet article.


Subject(s)
Betacoronavirus/genetics , Coronavirus Infections/virology , Organisms, Genetically Modified , Pandemics , Pneumonia, Viral/virology , Reverse Genetics/methods , Betacoronavirus/pathogenicity , Biohazard Release , COVID-19 , COVID-19 Vaccines , Chromosomes, Artificial, Yeast , Cloning, Molecular/methods , Coronaviridae/classification , Coronaviridae/genetics , Coronaviridae/pathogenicity , Coronavirus Infections/prevention & control , DNA, Complementary/genetics , Host Specificity , Humans , Organisms, Genetically Modified/genetics , Organisms, Genetically Modified/pathogenicity , Pandemics/prevention & control , Pneumonia, Viral/prevention & control , RNA, Viral/genetics , Recombination, Genetic , Risk , SARS-CoV-2 , Viral Vaccines
12.
Biomed Res Int ; 2020: 4389089, 2020.
Article in English | MEDLINE | ID: mdl-32596311

ABSTRACT

The Coronavirus Disease 2019 (COVID-19) is a new viral infection caused by the severe acute respiratory coronavirus 2 (SARS-CoV-2). Genomic analyses have revealed that SARS-CoV-2 is related to Pangolin and Bat coronaviruses. In this report, a structural comparison between the Sars-CoV-2 Envelope and Membrane proteins from different human isolates with homologous proteins from closely related viruses is described. The analyses here reported show the high structural similarity of Envelope and Membrane proteins to the counterparts from Pangolin and Bat coronavirus isolates. However, the comparisons have also highlighted structural differences specific of Sars-CoV-2 proteins which may be correlated to the cross-species transmission and/or to the properties of the virus. Structural modelling has been applied to map the variant sites onto the predicted three-dimensional structure of the Envelope and Membrane proteins.


Subject(s)
Betacoronavirus/chemistry , Coronavirus Infections/virology , Pneumonia, Viral/virology , Viral Envelope Proteins/chemistry , Viral Matrix Proteins/chemistry , Alphacoronavirus/chemistry , Alphacoronavirus/classification , Alphacoronavirus/genetics , Amino Acid Sequence , Animals , Betacoronavirus/classification , Betacoronavirus/genetics , COVID-19 , Chiroptera/virology , Coronaviridae/chemistry , Coronaviridae/classification , Coronaviridae/genetics , Coronavirus Envelope Proteins , Eutheria/virology , Humans , Models, Molecular , Pandemics , Protein Conformation , SARS-CoV-2 , Sequence Homology, Amino Acid , Species Specificity , Structural Homology, Protein , Viral Envelope Proteins/genetics , Viral Matrix Proteins/genetics
13.
Vet Microbiol ; 244: 108693, 2020 May.
Article in English | MEDLINE | ID: mdl-32402329

ABSTRACT

The recent pandemic caused by the novel human coronavirus, referrred to as severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), not only is having a great impact on the health care systems and economies in all continents but it is also causing radical changes of common habits and life styles. The novel coronavirus (CoV) recognises, with high probability, a zoonotic origin but the role of animals in the SARS-CoV-2 epidemiology is still largely unknown. However, CoVs have been known in animals since several decades, so that veterinary coronavirologists have a great expertise on how to face CoV infections in animals, which could represent a model for SARS-CoV-2 infection in humans. In the present paper, we provide an up-to-date review of the literature currently available on animal CoVs, focusing on the molecular mechanisms that are responsible for the emergence of novel CoV strains with different antigenic, biologic and/or pathogenetic features. A full comprehension of the mechanisms driving the evolution of animal CoVs will help better understand the emergence, spreading, and evolution of SARS-CoV-2.


Subject(s)
Coronaviridae/classification , Coronavirus Infections/veterinary , Pneumonia, Viral/epidemiology , Pneumonia, Viral/virology , Animals , Betacoronavirus/classification , Betacoronavirus/genetics , Betacoronavirus/isolation & purification , COVID-19 , Coronaviridae/genetics , Coronavirus Infections/epidemiology , Coronavirus Infections/transmission , Coronavirus Infections/virology , Disease Models, Animal , Evolution, Molecular , Humans , Pandemics , Pneumonia, Viral/transmission , SARS-CoV-2 , Zoonoses/epidemiology , Zoonoses/transmission , Zoonoses/virology
14.
Viruses ; 12(5)2020 05 25.
Article in English | MEDLINE | ID: mdl-32466150

ABSTRACT

This is the first record of a fish nidovirus isolated from a consignment of goldfish at the United Kingdom (UK) border. The full-length viral genome was 25,985 nt, sharing a 97.9% nucleotide identity with the Chinook salmon bafinivirus (CSBV) NIDO with two deletions of 537 and 480 nt on the ORF Ia protein. To assess the potential impact on UK fish species, Atlantic salmon, common carp and goldfish were exposed to the virus via an intraperitoneal (IP) injection and bath challenge. Moribundity was recorded in only 8% of IP-injected goldfish. A high viral load, ≈107 of the CSBV PpIa gene, was measured in the kidney of moribund goldfish. Mild histopathological changes were observed in the kidneys of challenged carps. Ultrastructural observations in renal tubule epithelial cells of goldfish showed cylindrical tubes (≈15 nm in diameter) and tubular structures budding spherical virions (≈200 nm in diameter) with external spike-like structures. Negative staining showed both circular and bacilliform virions. Seroconversion was measured in common carp and goldfish but not in Atlantic salmon. This study reinforces the potential risk of novel and emerging pathogens being introduced to recipient countries via the international ornamental fish trade and the importance of regular full health screens at the border inspection posts to reduce this risk.


Subject(s)
Coronaviridae/isolation & purification , Fish Diseases/virology , Goldfish/virology , Salmon/virology , Animals , Carps/virology , Coronaviridae/classification , Coronaviridae/genetics , Fish Diseases/diagnosis , Fish Diseases/pathology , Genes, Viral/genetics , Genome, Viral , Kidney/pathology , Kidney/virology , Nidovirales , Phylogeny , United Kingdom , Virulence
15.
Front Biosci (Landmark Ed) ; 25(10): 1894-1900, 2020 06 01.
Article in English | MEDLINE | ID: mdl-32472763

ABSTRACT

We analyzed the nucleocapsid and surface proteins from several Coronaviridae viruses using an alignment-free computer program. Three isolates of novel, human coronavirus (SARS0CoV-2) (2019) that are responsible for the current pandemic and older SARS strains of human and animal coronaviruses were examined. The nucleocapsid and glycoprotein sequences are identical for the three novel 2019 human isolates and they are closely related to these sequences in six bat and human SARS coronaviruses. This strongly supports the bat origin of the pandemic, novel coronavirus. One surface glycoprotein fragment of 111 amino acids is the largest, conserved, common permutation in the examined bat SARS-like and human SARS viruses, including the Covid-19 virus. BLAST analysis confirmed that this fragment is conserved only in the human and bat SARS strains. This fragment likely is involved in infectivity and is of interest for vaccine development. Surface glycoprotein and nucleocapsid protein sequence homologies of 58.9% and 82.5%, respectively, between the novel SARS0CoV-2 strains and the human SARS (2018) virus suggest that existing anti-SARS vaccines may provide some protection against the novel coronavirus.


Subject(s)
Betacoronavirus/genetics , Coronaviridae/genetics , Coronavirus Infections , Nucleocapsid Proteins/genetics , Pandemics , Pneumonia, Viral , Spike Glycoprotein, Coronavirus/genetics , Algorithms , Amino Acid Sequence , Animals , COVID-19 , Chiroptera/virology , Coronaviridae/classification , Coronavirus Nucleocapsid Proteins , Genome, Viral/genetics , Humans , Phosphoproteins , SARS-CoV-2 , Software , Species Specificity , Viral Envelope Proteins/genetics
16.
PLoS Pathog ; 16(5): e1008421, 2020 05.
Article in English | MEDLINE | ID: mdl-32407364

ABSTRACT

The outbreak of a novel corona Virus Disease 2019 (COVID-19) in the city of Wuhan, China has resulted in more than 1.7 million laboratory confirmed cases all over the world. Recent studies showed that SARS-CoV-2 was likely originated from bats, but its intermediate hosts are still largely unknown. In this study, we assembled the complete genome of a coronavirus identified in 3 sick Malayan pangolins. The molecular and phylogenetic analyses showed that this pangolin coronavirus (pangolin-CoV-2020) is genetically related to the SARS-CoV-2 as well as a group of bat coronaviruses but do not support the SARS-CoV-2 emerged directly from the pangolin-CoV-2020. Our study suggests that pangolins are natural hosts of Betacoronaviruses. Large surveillance of coronaviruses in pangolins could improve our understanding of the spectrum of coronaviruses in pangolins. In addition to conservation of wildlife, minimizing the exposures of humans to wildlife will be important to reduce the spillover risks of coronaviruses from wild animals to humans.


Subject(s)
Betacoronavirus/classification , Betacoronavirus/genetics , Coronavirus Infections/virology , Disease Reservoirs/virology , Eutheria/virology , Pneumonia, Viral/virology , Animals , COVID-19 , Coronaviridae/classification , Coronaviridae/genetics , Host Specificity , Humans , Pandemics , Phylogeny , SARS-CoV-2 , Sequence Homology, Nucleic Acid , Zoonoses/prevention & control , Zoonoses/virology
17.
Infect Genet Evol ; 81: 104272, 2020 07.
Article in English | MEDLINE | ID: mdl-32142938

ABSTRACT

The seventh novel human infecting Betacoronavirus that causes pneumonia (2019 novel coronavirus, 2019-nCoV) originated in Wuhan, China. The evolutionary relationship between 2019-nCoV and the other human respiratory illness-causing coronavirus is not closely related. We sought to characterize the relationship of the translated proteins of 2019-nCoV with other species of Orthocoronavirinae. A phylogenetic tree was constructed from the genome sequences. A cluster tree was developed from the profiles retrieved from the presence and absence of homologs of ten 2019-nCoV proteins. The combined data were used to characterize the relationship of the translated proteins of 2019-nCoV to other species of Orthocoronavirinae. Our analysis reliably suggests that 2019-nCoV is most closely related to BatCoV RaTG13 and belongs to subgenus Sarbecovirus of Betacoronavirus, together with SARS coronavirus and Bat-SARS-like coronavirus. The phylogenetic profiling cluster of homolog proteins of one annotated 2019-nCoV protein against other genome sequences revealed two clades of ten 2019-nCoV proteins. Clade 1 consisted of a group of conserved proteins in Orthocoronavirinae comprising Orf1ab polyprotein, Nucleocapsid protein, Spike glycoprotein, and Membrane protein. Clade 2 comprised six proteins exclusive to Sarbecovirus and Hibecovirus. Two of six Clade 2 nonstructural proteins, NS7b and NS8, were exclusively conserved among 2019-nCoV, BetaCoV_RaTG, and BatSARS-like Cov. NS7b and NS8 have previously been shown to affect immune response signaling in the SARS-CoV experimental model. Thus, we speculated that knowledge of the functional changes in the NS7b and NS8 proteins during evolution may provide important information to explore the human infective property of 2019-nCoV.


Subject(s)
Betacoronavirus/classification , Betacoronavirus/genetics , Evolution, Molecular , Phylogeny , Viral Nonstructural Proteins/genetics , Coronaviridae/classification , Coronaviridae/genetics , Coronavirus RNA-Dependent RNA Polymerase , SARS-CoV-2
18.
Nat Microbiol ; 5(4): 536-544, 2020 04.
Article in English | MEDLINE | ID: mdl-32123347

ABSTRACT

The present outbreak of a coronavirus-associated acute respiratory disease called coronavirus disease 19 (COVID-19) is the third documented spillover of an animal coronavirus to humans in only two decades that has resulted in a major epidemic. The Coronaviridae Study Group (CSG) of the International Committee on Taxonomy of Viruses, which is responsible for developing the classification of viruses and taxon nomenclature of the family Coronaviridae, has assessed the placement of the human pathogen, tentatively named 2019-nCoV, within the Coronaviridae. Based on phylogeny, taxonomy and established practice, the CSG recognizes this virus as forming a sister clade to the prototype human and bat severe acute respiratory syndrome coronaviruses (SARS-CoVs) of the species Severe acute respiratory syndrome-related coronavirus, and designates it as SARS-CoV-2. In order to facilitate communication, the CSG proposes to use the following naming convention for individual isolates: SARS-CoV-2/host/location/isolate/date. While the full spectrum of clinical manifestations associated with SARS-CoV-2 infections in humans remains to be determined, the independent zoonotic transmission of SARS-CoV and SARS-CoV-2 highlights the need for studying viruses at the species level to complement research focused on individual pathogenic viruses of immediate significance. This will improve our understanding of virus­host interactions in an ever-changing environment and enhance our preparedness for future outbreaks.


Subject(s)
Betacoronavirus/classification , Animals , Betacoronavirus/genetics , COVID-19 , Classification/methods , Coronaviridae/classification , Coronaviridae/genetics , Coronavirus Infections/epidemiology , Coronavirus Infections/transmission , Coronavirus Infections/virology , Genetic Variation , Genome, Viral , Humans , Nidovirales/classification , Nidovirales/genetics , Open Reading Frames , Pandemics , Phylogeny , Pneumonia, Viral/epidemiology , Pneumonia, Viral/transmission , Pneumonia, Viral/virology , Severe acute respiratory syndrome-related coronavirus/classification , Severe acute respiratory syndrome-related coronavirus/genetics , SARS-CoV-2 , Severe Acute Respiratory Syndrome/epidemiology , Severe Acute Respiratory Syndrome/virology , Terminology as Topic , World Health Organization , Zoonoses
19.
Mem Inst Oswaldo Cruz ; 114: e190198, 2019.
Article in English | MEDLINE | ID: mdl-31596309

ABSTRACT

BACKGROUND: In Brazil the implementation of the Sentinel Surveillance System of Influenza began in 2000. Central public health laboratories use reverse transcription-quantitative polymerase chain reaction (RT-qPCR) for diagnosis of respiratory viruses, but this protocol identifies only specific targets, resulted in inconclusive diagnosis for many samples. Thus, high-throughput sequencing (HTS) would be complementary method in the identification of pathogens in inconclusive samples for RT-qPCR or other specific detection protocols. OBJECTIVES: This study aimed to detect unidentified viruses using HTS approach in negative samples of nasopharynx/tracheal secretions by the standard RT-qPCR collected in the Federal District, Brazil. METHODS: Nucleic acids were extracted from samples collected in winter period of 2016 and subjected to HTS. The results were confirmed by the multiplex PR21 RT-qPCR, which identifies 21 respiratory pathogens. FINDINGS: The main viruses identified by HTS were of families Herpesviridae, Coronaviridae, Parvoviridae and Picornaviridae, with the emphasis on rhinoviruses. The presence of respiratory viruses in the samples was confirmed by the PR21 multiplex RT-qPCR. Coronavirus, enterovirus, bocavirus and rhinovirus were found by multiplex RT-qPCR as well as by HTS analyses. MAIN CONCLUSIONS: Wide virus diversity was found by different methodologies and high frequency of rhinovirus occurrence was confirmed in population in winter, showing its relevance for public health.


Subject(s)
Coronaviridae/isolation & purification , Herpesviridae/isolation & purification , Nasopharynx/virology , Parvoviridae/isolation & purification , Picornaviridae/isolation & purification , Trachea/virology , Coronaviridae/classification , Coronaviridae/genetics , DNA, Viral/genetics , Herpesviridae/classification , Herpesviridae/genetics , Humans , Parvoviridae/classification , Parvoviridae/genetics , Picornaviridae/classification , Picornaviridae/genetics , RNA, Viral/genetics , Real-Time Polymerase Chain Reaction , Reverse Transcriptase Polymerase Chain Reaction
20.
Mol Cell Probes ; 47: 101435, 2019 10.
Article in English | MEDLINE | ID: mdl-31415867

ABSTRACT

Currently in China, porcine epidemic diarrhea virus (PEDV), transmissible gastroenteritis virus (TGEV), porcine rotavirus (PoRV), and porcine deltacoronavirus (PDCoV) are the major causes of porcine viral diarrhea, and mixed infections in clinics are common, resulting in significant economic losses in pig industry. Here, a dual priming oligonucleotide (DPO)-based multiplex real-time SYBR Green RT-PCR assay were developed for accurately differentiating PEDV, TGEV, PoRV, and PDCoV in clinical specimens targeting the N gene of TGEV, PEDV, and PDCoV, and the VP7 gene of PoRV. Results showed that the DPO primer allowed a wider annealing temperature range (40-65 °C) and had a higher priming specificity compared to conventional primer, in which more than 3 nucleotides in the 3'- or 5'-segment of DPO primer mismatched with DNA template, PCR amplification efficiency would decrease substantially or extension would not proceed. DPO-based multiplex real-time RT-PCR method had analytical detection limit of 8.63 × 102 copies/µL, 1.92 × 102 copies/µL, 1.74 × 102 copies/µL, and 1.76 × 102 copies/µL for PEDV, TGEV, PoRV, and PDCoV in clinical specimens, respectively. A total of 672 clinical specimens of piglets with diarrheal symptoms were collected in Northeastern China from 2017 to 2018 followed by analysis using the assay, and epidemiological investigation results showed that PEDV, TGEV, PoRV, and PDCoV prevalence was 19.05%, 5.21%, 4.32%, and 3.87%, respectively. The assay developed in this study showed higher detection accuracy than conventional RT-PCR method, suggesting a useful tool for the accurate differentiation of the four major viruses causing porcine viral diarrhea in practice.


Subject(s)
Coronaviridae/classification , DNA Primers/genetics , Diarrhea/veterinary , Real-Time Polymerase Chain Reaction/methods , Swine Diseases/virology , Animals , Coronaviridae/genetics , Coronaviridae/isolation & purification , Coronavirus/genetics , Coronavirus/isolation & purification , Diarrhea/virology , Porcine epidemic diarrhea virus/genetics , Porcine epidemic diarrhea virus/isolation & purification , RNA, Viral/genetics , Rotavirus/genetics , Rotavirus/isolation & purification , Species Specificity , Swine , Transmissible gastroenteritis virus/genetics , Transmissible gastroenteritis virus/isolation & purification
SELECTION OF CITATIONS
SEARCH DETAIL
...