Your browser doesn't support javascript.
Show: 20 | 50 | 100
Results 1 - 20 de 31
Filter
1.
J Gen Virol ; 104(4)2023 04.
Article in English | MEDLINE | ID: covidwho-2305800

ABSTRACT

The family Coronaviridae includes viruses with positive-sense RNA genomes of 22-36 kb that are expressed through a nested set of 3' co-terminal subgenomic mRNAs. Members of the subfamily Orthocoronavirinae are characterized by 80-160 nm diameter, enveloped virions with spike projections. The orthocoronaviruses, severe acute respiratory syndrome coronavirus and Middle East respiratory syndrome-related coronavirus are extremely pathogenic for humans and in the last two decades have been responsible for the SARS and MERS epidemics. Another orthocoronavirus, severe acute respiratory syndrome coronavirus 2, was responsible for the recent global COVID-19 pandemic. This is a summary of the International Committee on Taxonomy of Viruses (ICTV) Report on the family Coronaviridae which is available at www.ictv.global/report/coronaviridae.


Subject(s)
COVID-19 , Coronaviridae , Viruses , Humans , Coronaviridae/genetics , Pandemics , Viruses/genetics , Virion/genetics , Genome, Viral , Virus Replication
2.
Infect Genet Evol ; 108: 105399, 2023 03.
Article in English | MEDLINE | ID: covidwho-2165704

ABSTRACT

Bats remains as reservoirs for highly contagious and pathogenic viral families including the Coronaviridae, Filoviridae, Paramyxoviruses, and Rhabdoviridae. Spill over of viral species (SARS-CoV, MERS-CoV & SARS-CoV2) from bats (as a possible potential reservoirs) have recently caused worst outbreaks. Early detection of viral species of pandemic potential in bats is of great importance. We detected beta coronaviruses in the studied bats population (positive samples from Rousettus leschenaultia) and performed the evolutionary analysis, amino acid sequence alignment, and analysed the 3-Dimentional protein structure. We detected the coronaviruses for the first time in bats from Pakistan. Our analysis based on RdRp partial gene sequencing suggest that the studied viral strains are closely related to MERS-CoV-like viruses as they exhibit close structure similarities (with few substitutions) and also observed a substitution in highly conserved SDD in the palm subdomain of motif C to ADD, when compared with earlier reported viral strains. It could be concluded from our study that coronaviruses are circulating among the bat's population in Pakistan. Based on the current findings, we suggest large scale screening procedures of bat virome across the country to detect potential pathogenic viral species.


Subject(s)
COVID-19 , Chiroptera , Coronaviridae , Middle East Respiratory Syndrome Coronavirus , Viruses , Humans , Animals , RNA, Viral , Pakistan/epidemiology , Phylogeny , COVID-19/genetics , SARS-CoV-2/genetics , Coronaviridae/genetics , Viruses/genetics , Middle East Respiratory Syndrome Coronavirus/genetics , Genome, Viral
3.
Viruses ; 13(8)2021 08 05.
Article in English | MEDLINE | ID: covidwho-1390162

ABSTRACT

Nucleotidylylation is a post-transcriptional modification important for replication in the picornavirus supergroup of RNA viruses, including members of the Caliciviridae, Coronaviridae, Picornaviridae and Potyviridae virus families. This modification occurs when the RNA-dependent RNA polymerase (RdRp) attaches one or more nucleotides to a target protein through a nucleotidyl-transferase reaction. The most characterized nucleotidylylation target is VPg (viral protein genome-linked), a protein linked to the 5' end of the genome in Caliciviridae, Picornaviridae and Potyviridae. The nucleotidylylation of VPg by RdRp is a critical step for the VPg protein to act as a primer for genome replication and, in Caliciviridae and Potyviridae, for the initiation of translation. In contrast, Coronaviridae do not express a VPg protein, but the nucleotidylylation of proteins involved in replication initiation is critical for genome replication. Furthermore, the RdRp proteins of the viruses that perform nucleotidylylation are themselves nucleotidylylated, and in the case of coronavirus, this has been shown to be essential for viral replication. This review focuses on nucleotidylylation within the picornavirus supergroup of viruses, including the proteins that are modified, what is known about the nucleotidylylation process and the roles that these modifications have in the viral life cycle.


Subject(s)
Nucleotides/metabolism , Positive-Strand RNA Viruses/metabolism , RNA-Dependent RNA Polymerase/metabolism , Viral Proteins/metabolism , Caliciviridae/genetics , Caliciviridae/metabolism , Coronaviridae/genetics , Coronaviridae/metabolism , Genome, Viral , Nidovirales/genetics , Nidovirales/metabolism , Picornaviridae/genetics , Picornaviridae/metabolism , Positive-Strand RNA Viruses/genetics , Potyviridae/genetics , Potyviridae/metabolism , RNA, Viral/genetics , RNA, Viral/metabolism , Virus Replication
4.
Viruses ; 14(7)2022 06 25.
Article in English | MEDLINE | ID: covidwho-1911654

ABSTRACT

Coronaviruses are well known as a diverse family of viruses that affect a wide range of hosts. Since the outbreak of severe acute respiratory syndrome, a variety of bat-associated coronaviruses have been identified in many countries. However, they do not represent all the specific geographic locations of their hosts. In this study, full-length genomes representing newly identified bat coronaviruses in South Korea were obtained using an RNA sequencing approach. The analysis, based on genome structure, conserved replicase domains, spike gene, and nucleocapsid genes revealed that bat Alphacoronaviruses are from three different viral species. Among them, the newly identified B20-97 strain may represent a new putative species, closely related to PEDV. In addition, the newly-identified MERS-related coronavirus exhibited shared genomic nucleotide identities of less than 76.4% with other Merbecoviruses. Recombination analysis and multiple alignments of spike and RBD amino acid sequences suggested that this strain underwent recombination events and could possibly use hDPP4 molecules as its receptor. The bat SARS-related CoV B20-50 is unlikely to be able to use hACE2 as its receptor and lack of an open reading frame in ORF8 gene region. Our results illustrate the diversity of coronaviruses in Korean bats and their evolutionary relationships. The evolution of the bat coronaviruses related ORF8 accessory gene is also discussed.


Subject(s)
Alphacoronavirus , Chiroptera , Coronaviridae , Coronavirus Infections , Middle East Respiratory Syndrome Coronavirus , Severe acute respiratory syndrome-related coronavirus , Alphacoronavirus/genetics , Animals , Betacoronavirus/genetics , Coronaviridae/genetics , Genome, Viral , Genomics , Middle East Respiratory Syndrome Coronavirus/genetics , Phylogeny , Severe acute respiratory syndrome-related coronavirus/genetics
5.
Virology ; 570: 123-133, 2022 05.
Article in English | MEDLINE | ID: covidwho-1764025

ABSTRACT

The current outbreak of coronavirus disease-2019 (COVID-19) caused by SARS-CoV-2 poses unparalleled challenges to global public health. SARS-CoV-2 is a Betacoronavirus, one of four genera belonging to the Coronaviridae subfamily Orthocoronavirinae. Coronaviridae, in turn, are members of the order Nidovirales, a group of enveloped, positive-stranded RNA viruses. Here we present a systematic phylogenetic and evolutionary study based on protein domain architecture, encompassing the entire proteomes of all Orthocoronavirinae, as well as other Nidovirales. This analysis has revealed that the genomic evolution of Nidovirales is associated with extensive gains and losses of protein domains. In Orthocoronavirinae, the sections of the genomes that show the largest divergence in protein domains are found in the proteins encoded in the amino-terminal end of the polyprotein (PP1ab), the spike protein (S), and many of the accessory proteins. The diversity among the accessory proteins is particularly striking, as each subgenus possesses a set of accessory proteins that is almost entirely specific to that subgenus. The only notable exception to this is ORF3b, which is present and orthologous over all Alphacoronaviruses. In contrast, the membrane protein (M), envelope small membrane protein (E), nucleoprotein (N), as well as proteins encoded in the central and carboxy-terminal end of PP1ab (such as the 3C-like protease, RNA-dependent RNA polymerase, and Helicase) show stable domain architectures across all Orthocoronavirinae. This comprehensive analysis of the Coronaviridae domain architecture has important implication for efforts to develop broadly cross-protective coronavirus vaccines.


Subject(s)
COVID-19 , Coronaviridae , Nidovirales , Coronaviridae/genetics , Evolution, Molecular , Humans , Membrane Proteins/genetics , Nidovirales/genetics , Phylogeny , SARS-CoV-2/genetics
6.
Vopr Virusol ; 67(1): 69-76, 2022 03 15.
Article in Russian | MEDLINE | ID: covidwho-1744341

ABSTRACT

INTRODUCTION: The novel coronavirus infection COVID-19 is a major public health problem worldwide. Several publications show the presence of gastrointestinal (GI) symptoms (nausea, vomiting, and diarrhea) in addition to respiratory disorders.The aim of this study was the monitoring of RNA of COVID-19 pathogen, coronavirus SARS-CoV-2 (Coronaviridae: Coronavirinae: Betacoronavirus; Sarbecovirus) in children hospitalized with acute intestinal infection (AII), with following molecular-genetic characterization of detected strains. MATERIAL AND METHODS: Fecal samples of children with AII hospitalized in infectious hospital of Nizhny Novgorod (Russia) in the period from 01.07.2020 to 31.10.2021 were used as material for the study. Viral RNA detection was performed by real-time polymerase chain reaction (RT-PCR). The nucleotide sequence of S-protein gene fragment was determined by Sanger sequencing. RESULTS AND DISCUSSION: SARS-CoV-2 genetic material was detected in 45 out of 2476 fecal samples. The maximum number of samples containing RNA of the virus occurred in November 2020 (detection rate of 12.2%). In 20.0% of cases, SARS-CoV-2 RNA was detected in combination with rota-, noro-, and adenoviruses. 28 nucleotide sequences of S-protein gene fragment complementary DNA (cDNA) were determined. Phylogenetic analysis showed that the studied SARS-CoV-2 strains belonged to two variants. Analysis of the S-protein amino acid sequence of the strains studied showed the absence of the N501Y mutation in the 2020 samples, which is a marker for variants with a high epidemic potential, called variants of concern (VOC) according to the World Health Organization (WHO) definition (lines Alpha B.1.1.7, Beta B.1.351, Gamma P.1). Delta line variant B.1.617.2 was identified in two samples isolated in September 2021. CONCLUSION: The detection of SARS-CoV-2 RNA in the fecal samples of children with AII, suggesting that the fecal-oral mechanism of pathogen transmission may exist, determines the necessity to optimize its monitoring and to develop an algorithm of actions with patients with signs of AII under the conditions of a novel coronavirus infection pandemic.


Subject(s)
COVID-19 , Coronaviridae , COVID-19/diagnosis , COVID-19/epidemiology , Child , Coronaviridae/genetics , Humans , Phylogeny , RNA, Viral/genetics , SARS-CoV-2/genetics
7.
Viruses ; 14(2)2022 01 18.
Article in English | MEDLINE | ID: covidwho-1625960

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
8.
Nucleic Acids Res ; 50(D1): D765-D770, 2022 01 07.
Article in English | MEDLINE | ID: covidwho-1462428

ABSTRACT

The COVID-19 pandemic has seen unprecedented use of SARS-CoV-2 genome sequencing for epidemiological tracking and identification of emerging variants. Understanding the potential impact of these variants on the infectivity of the virus and the efficacy of emerging therapeutics and vaccines has become a cornerstone of the fight against the disease. To support the maximal use of genomic information for SARS-CoV-2 research, we launched the Ensembl COVID-19 browser; the first virus to be encompassed within the Ensembl platform. This resource incorporates a new Ensembl gene set, multiple variant sets, and annotation from several relevant resources aligned to the reference SARS-CoV-2 assembly. Since the first release in May 2020, the content has been regularly updated using our new rapid release workflow, and tools such as the Ensembl Variant Effect Predictor have been integrated. The Ensembl COVID-19 browser is freely available at https://covid-19.ensembl.org.


Subject(s)
COVID-19/virology , Databases, Genetic , SARS-CoV-2/genetics , Web Browser , Coronaviridae/genetics , Genetic Variation , Genome, Viral , Humans , Molecular Sequence Annotation
9.
Science ; 374(6567): eabj3624, 2021 Oct 29.
Article in English | MEDLINE | ID: covidwho-1440797

ABSTRACT

Inherited genetic factors can influence the severity of COVID-19, but the molecular explanation underpinning a genetic association is often unclear. Intracellular antiviral defenses can inhibit the replication of viruses and reduce disease severity. To better understand the antiviral defenses relevant to COVID-19, we used interferon-stimulated gene (ISG) expression screening to reveal that 2'-5'-oligoadenylate synthetase 1 (OAS1), through ribonuclease L, potently inhibits severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2). We show that a common splice-acceptor single-nucleotide polymorphism (Rs10774671) governs whether patients express prenylated OAS1 isoforms that are membrane-associated and sense-specific regions of SARS-CoV-2 RNAs or if they only express cytosolic, nonprenylated OAS1 that does not efficiently detect SARS-CoV-2. In hospitalized patients, expression of prenylated OAS1 was associated with protection from severe COVID-19, suggesting that this antiviral defense is a major component of a protective antiviral response.


Subject(s)
2',5'-Oligoadenylate Synthetase/genetics , 2',5'-Oligoadenylate Synthetase/metabolism , COVID-19/genetics , COVID-19/physiopathology , RNA, Double-Stranded/metabolism , RNA, Viral/metabolism , SARS-CoV-2/physiology , 5' Untranslated Regions , A549 Cells , Animals , COVID-19/enzymology , COVID-19/immunology , Chiroptera/genetics , Chiroptera/virology , Coronaviridae/enzymology , Coronaviridae/genetics , Coronaviridae/physiology , Endoribonucleases/metabolism , Humans , Interferons/immunology , Isoenzymes/genetics , Isoenzymes/metabolism , Phosphoric Diester Hydrolases/genetics , Phosphoric Diester Hydrolases/metabolism , Polymorphism, Single Nucleotide , Protein Prenylation , RNA, Double-Stranded/chemistry , RNA, Double-Stranded/genetics , RNA, Viral/chemistry , RNA, Viral/genetics , Retroelements , SARS-CoV-2/genetics , Severity of Illness Index , Virus Replication
10.
Int J Biol Sci ; 17(14): 3717-3727, 2021.
Article in English | MEDLINE | ID: covidwho-1417291

ABSTRACT

SARS-CoV-2 belongs to the coronavirus family. Comparing genomic features of viral genomes of coronavirus family can improve our understanding about SARS-CoV-2. Here we present the first pan-genome analysis of 3,932 whole genomes of 101 species out of 4 genera from the coronavirus family. We found that a total of 181 genes in the pan-genome of coronavirus family, among which only 3 genes, the S gene, M gene and N gene, are highly conserved. We also constructed a pan-genome from 23,539 whole genomes of SARS-CoV-2. There are 13 genes in total in the SARS-CoV-2 pan-genome. All of the 13 genes are core genes for SARS-CoV-2. The pan-genome of coronaviruses shows a lower level of diversity than the pan-genomes of other RNA viruses, which contain no core gene. The three highly conserved genes in coronavirus family, which are also core genes in SARS-CoV-2 pan-genome, could be potential targets in developing nucleic acid diagnostic reagents with a decreased possibility of cross-reaction with other coronavirus species.


Subject(s)
Coronaviridae/genetics , Genome, Viral , Phylogeny
11.
Infect Genet Evol ; 95: 105075, 2021 11.
Article in English | MEDLINE | ID: covidwho-1401708

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
12.
Transbound Emerg Dis ; 69(4): 1748-1760, 2022 Jul.
Article in English | MEDLINE | ID: covidwho-1388406

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
13.
Virology ; 563: 20-27, 2021 11.
Article in English | MEDLINE | ID: covidwho-1356482

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
14.
Sci Rep ; 11(1): 16145, 2021 08 09.
Article in English | MEDLINE | ID: covidwho-1349686

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
15.
Viruses ; 13(5)2021 05 18.
Article in English | MEDLINE | ID: covidwho-1234835

ABSTRACT

The ongoing coronavirus disease 2019 (COVID-19) pandemic has had devastating health and socio-economic impacts. Human activities, especially at the wildlife interphase, are at the core of forces driving the emergence of new viral agents. Global surveillance activities have identified bats as the natural hosts of diverse coronaviruses, with other domestic and wildlife animal species possibly acting as intermediate or spillover hosts. The African continent is confronted by several factors that challenge prevention and response to novel disease emergences, such as high species diversity, inadequate health systems, and drastic social and ecosystem changes. We reviewed published animal coronavirus surveillance studies conducted in Africa, specifically summarizing surveillance approaches, species numbers tested, and findings. Far more surveillance has been initiated among bat populations than other wildlife and domestic animals, with nearly 26,000 bat individuals tested. Though coronaviruses have been identified from approximately 7% of the total bats tested, surveillance among other animals identified coronaviruses in less than 1%. In addition to a large undescribed diversity, sequences related to four of the seven human coronaviruses have been reported from African bats. The review highlights research gaps and the disparity in surveillance efforts between different animal groups (particularly potential spillover hosts) and concludes with proposed strategies for improved future biosurveillance.


Subject(s)
Coronavirus Infections/epidemiology , Epidemiological Monitoring/veterinary , Africa/epidemiology , Animals , Animals, Wild/virology , COVID-19/epidemiology , Chiroptera/virology , Coronaviridae/genetics , Coronavirus/pathogenicity , Ecosystem , Genetic Variation , Genome, Viral , Pandemics , Phylogeny , SARS-CoV-2/pathogenicity , Spike Glycoprotein, Coronavirus/genetics
16.
Arch Immunol Ther Exp (Warsz) ; 68(6): 35, 2020 Nov 13.
Article in English | MEDLINE | ID: covidwho-983490

ABSTRACT

The COVID-19 pandemic developing rapidly in 2020 is triggered by the emergence of a new human virus-SARS-CoV-2. The emergence of a new virus is not an unexpected phenomenon and has been predicted for many years. Since the virus has spread all over the world, it will be very difficult or even impossible to eradicate it. A necessary condition for complete or partial elimination of the virus is to have an effective vaccine. It is possible that SARS-CoV-2 will become milder in the next few years and COVID-19 will then only threaten individuals from risk groups.


Subject(s)
Betacoronavirus/physiology , Coronavirus Infections/epidemiology , Pandemics , Pneumonia, Viral/epidemiology , Animals , Betacoronavirus/pathogenicity , Biological Evolution , COVID-19 , Communicable Disease Control/organization & administration , Communicable Diseases, Emerging , Coronaviridae/genetics , Coronaviridae/pathogenicity , Coronavirus Infections/transmission , Coronavirus Infections/veterinary , Coronavirus Infections/virology , Disease Eradication , Disease Susceptibility , Forecasting , Host Specificity , Humans , Pandemics/prevention & control , Pneumonia, Viral/transmission , Pneumonia, Viral/virology , SARS-CoV-2 , Selection, Genetic , Virulence , Zoonoses
17.
Virus Res ; 290: 198175, 2020 12.
Article in English | MEDLINE | ID: covidwho-811767

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
18.
Chem Biol Interact ; 331: 109226, 2020 Nov 01.
Article in English | MEDLINE | ID: covidwho-778555

ABSTRACT

Presence of Simple Sequence Repeats (SSRs), both in genic and intergenic regions, have been widely studied in eukaryotes, prokaryotes, and viruses. In the current study, we undertook a survey to analyze the frequency and distribution of microsatellites or SSRs in multiple genomes of Coronaviridae members. We successfully identified 919 SSRs with length ≥12 bp across 55 reference genomes majority of which (838 SSRs) were found abundant in genic regions. The in-silico analysis further identified the preferential abundance of hexameric SSRs than any other size-based motif class. Our analysis shows that the genome size and GC content of the genome had a weak influence on SSR frequency and density. However, we find a positive correlation of SSRs GC content with genomic GC content. We also report relatively low abundances of all theoretically possible 501 repeat motif classes in all the genomes of Coronaviridae. The majority of SSRs were AT-rich. Overall, we see an underrepresentation of SSRs across the genomes of Coronaviridae. Besides, our integrative study highlights the presence of SSRs in ORF1ab (nsp3, nsp4, nsp5A_3CLpro and nsp5B_3CLpro, nsp6, nsp10, nsp12, nsp13, & nsp15 domains), S, ORF3a, ORF7a, N & 3' UTR regions of SARS-CoV-2 and harbours multiple mutations (3'UTR and ORF1ab SSRs serving as major mutational hotspots). This indicates the genic SSRs are under selection pressure against mutations that might alter the reading frame and at the same time responsible for rapid protein evolution. Our preliminary results indicate the significance of the limited repertoire of SSRs in the genomes of Coronaviridae.


Subject(s)
Coronaviridae/genetics , Microsatellite Repeats/genetics , 3' Untranslated Regions , Base Composition , Base Sequence , Betacoronavirus/genetics , Evolution, Molecular , Genome, Viral , Humans , Mutation , Polyproteins , SARS-CoV-2 , Viral Proteins/genetics
19.
BMC Res Notes ; 13(1): 398, 2020 Aug 27.
Article in English | MEDLINE | ID: covidwho-733023

ABSTRACT

OBJECTIVE: In December 2019 a novel coronavirus (SARS-CoV-2) that is causing the current COVID-19 pandemic was identified in Wuhan, China. Many questions have been raised about its origin and adaptation to humans. In the present work we performed a genetic analysis of the Spike glycoprotein (S) of SARS-CoV-2 and other related coronaviruses (CoVs) isolated from different hosts in order to trace the evolutionary history of this protein and the adaptation of SARS-CoV-2 to humans. RESULTS: Based on the sequence analysis of the S gene, we suggest that the origin of SARS-CoV-2 is the result of recombination events between bat and pangolin CoVs. The hybrid SARS-CoV-2 ancestor jumped to humans and has been maintained by natural selection. Although the S protein of RaTG13 bat CoV has a high nucleotide identity with the S protein of SARS-CoV-2, the phylogenetic tree and the haplotype network suggest a non-direct parental relationship between these CoVs. Moreover, it is likely that the basic function of the receptor-binding domain (RBD) of S protein was acquired by the SARS-CoV-2 from the MP789 pangolin CoV by recombination and it has been highly conserved.


Subject(s)
Betacoronavirus/genetics , Coronaviridae/genetics , Recombination, Genetic , Spike Glycoprotein, Coronavirus/genetics , Adaptation, Biological/genetics , Angiotensin-Converting Enzyme 2 , Animals , Binding Sites/genetics , Chiroptera/virology , Eutheria/virology , Evolution, Molecular , Furin/metabolism , Host Specificity , Humans , Peptidyl-Dipeptidase A/metabolism , Phylogeny , SARS-CoV-2 , Selection, Genetic , Spike Glycoprotein, Coronavirus/metabolism
20.
Transbound Emerg Dis ; 68(3): 987-992, 2021 May.
Article in English | MEDLINE | ID: covidwho-722280

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
SELECTION OF CITATIONS
SEARCH DETAIL