Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 148
Filter
1.
Viruses ; 13(10)2021 09 23.
Article in English | MEDLINE | ID: mdl-34696338

ABSTRACT

Diverse coronavirus (CoV) strains can infect both humans and animals and produce various diseases. CoVs have caused three epidemics and pandemics in the last two decades, and caused a severe impact on public health and the global economy. Therefore, it is of utmost importance to understand the emergence and evolution of endemic and emerging CoV diversity in humans and animals. For diverse bird species, the Infectious Bronchitis Virus is a significant one, whereas feline enteric and canine coronavirus, recombined to produce feline infectious peritonitis virus, infects wild cats. Bovine and canine CoVs have ancestral relationships, while porcine CoVs, especially SADS-CoV, can cross species barriers. Bats are considered as the natural host of diverse strains of alpha and beta coronaviruses. Though MERS-CoV is significant for both camels and humans, humans are nonetheless affected more severely. MERS-CoV cases have been reported mainly in the Arabic peninsula since 2012. To date, seven CoV strains have infected humans, all descended from animals. The severe acute respiratory syndrome coronaviruses (SARS-CoV and SARS-CoV-2) are presumed to be originated in Rhinolopoid bats that severely infect humans with spillover to multiple domestic and wild animals. Emerging alpha and delta variants of SARS-CoV-2 were detected in pets and wild animals. Still, the intermediate hosts and all susceptible animal species remain unknown. SARS-CoV-2 might not be the last CoV to cross the species barrier. Hence, we recommend developing a universal CoV vaccine for humans so that any future outbreak can be prevented effectively. Furthermore, a One Health approach coronavirus surveillance should be implemented at human-animal interfaces to detect novel coronaviruses before emerging to humans and to prevent future epidemics and pandemics.


Subject(s)
Coronavirus Infections/epidemiology , Coronavirus Infections/genetics , Epidemics/prevention & control , Animals , Animals, Domestic/virology , Animals, Wild/virology , Coronaviridae/metabolism , Coronaviridae/pathogenicity , Genome, Viral/genetics , Humans , Middle East Respiratory Syndrome Coronavirus/genetics , Pandemics/prevention & control , Phylogeny , Severe acute respiratory syndrome-related coronavirus/genetics , SARS-CoV-2/genetics , Viral Zoonoses/epidemiology , Viral Zoonoses/transmission
2.
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
3.
Biomed Res Int ; 2021: 8856018, 2021.
Article in English | MEDLINE | ID: mdl-34239932

ABSTRACT

Coronaviruses (CoVs) are enveloped nonsegmented positive-sense RNA viruses belonging to the family Coronaviridae that contain the largest genome among RNA viruses. Their genome encodes 4 major structural proteins, and among them, the Spike (S) protein plays a crucial role in determining the viral tropism. It mediates viral attachment to the host cell, fusion to the membranes, and cell entry using cellular proteases as activators. Several in vitro models have been developed to study the CoVs entry, pathogenesis, and possible therapeutic approaches. This article is aimed at summarizing the current knowledge about the use of relevant methodologies and cell lines permissive for CoV life cycle studies. The synthesis of this information can be useful for setting up specific experimental procedures. We also discuss different strategies for inhibiting the binding of the S protein to the cell receptors and the fusion process which may offer opportunities for therapeutic intervention.


Subject(s)
Antiviral Agents , Coronaviridae , Models, Biological , Viral Tropism , Virus Internalization , Antiviral Agents/chemistry , Antiviral Agents/pharmacology , COVID-19 , Cells, Cultured , Coronaviridae/drug effects , Coronaviridae/metabolism , Coronaviridae/pathogenicity , Coronaviridae/physiology , Coronaviridae Infections , Humans , SARS-CoV-2 , Spike Glycoprotein, Coronavirus/chemistry , Spike Glycoprotein, Coronavirus/metabolism
4.
PLoS Pathog ; 17(4): e1009149, 2021 04.
Article in English | MEDLINE | ID: mdl-33878118

ABSTRACT

The COVID-19 pandemic has demonstrated the serious potential for novel zoonotic coronaviruses to emerge and cause major outbreaks. The immediate animal origin of the causative virus, SARS-CoV-2, remains unknown, a notoriously challenging task for emerging disease investigations. Coevolution with hosts leads to specific evolutionary signatures within viral genomes that can inform likely animal origins. We obtained a set of 650 spike protein and 511 whole genome nucleotide sequences from 222 and 185 viruses belonging to the family Coronaviridae, respectively. We then trained random forest models independently on genome composition biases of spike protein and whole genome sequences, including dinucleotide and codon usage biases in order to predict animal host (of nine possible categories, including human). In hold-one-out cross-validation, predictive accuracy on unseen coronaviruses consistently reached ~73%, indicating evolutionary signal in spike proteins to be just as informative as whole genome sequences. However, different composition biases were informative in each case. Applying optimised random forest models to classify human sequences of MERS-CoV and SARS-CoV revealed evolutionary signatures consistent with their recognised intermediate hosts (camelids, carnivores), while human sequences of SARS-CoV-2 were predicted as having bat hosts (suborder Yinpterochiroptera), supporting bats as the suspected origins of the current pandemic. In addition to phylogeny, variation in genome composition can act as an informative approach to predict emerging virus traits as soon as sequences are available. More widely, this work demonstrates the potential in combining genetic resources with machine learning algorithms to address long-standing challenges in emerging infectious diseases.


Subject(s)
Biological Evolution , Coronaviridae Infections/diagnosis , Coronaviridae Infections/virology , Coronaviridae/pathogenicity , Genome, Viral , Machine Learning , Spike Glycoprotein, Coronavirus/metabolism , Animals , Coronaviridae Infections/genetics , Coronaviridae Infections/metabolism , Phylogeny , Spike Glycoprotein, Coronavirus/genetics
5.
Arch Immunol Ther Exp (Warsz) ; 68(6): 35, 2020 Nov 13.
Article in English | MEDLINE | ID: mdl-33185755

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
6.
Clin Immunol ; 220: 108588, 2020 11.
Article in English | MEDLINE | ID: mdl-32905851

ABSTRACT

Though recent reports link SARS-CoV-2 infections with hyper-inflammatory states in children, most children experience no/mild symptoms, and hospitalization and mortality rates are low in the age group. As symptoms are usually mild and seroconversion occurs at low frequencies, it remains unclear whether children significantly contribute to community transmission. Several hypotheses try to explain age-related differences in disease presentation and severity. Possible reasons for milder presentations in children as compared to adults include frequent contact to seasonal coronaviruses, presence of cross-reactive antibodies, and/or co-clearance with other viruses. Increased expression of ACE2 in young people may facilitate virus infection, while limiting inflammation and reducing the risk of severe disease. Further potential factors include recent vaccinations and a more diverse memory T cell repertoire. This manuscript reviews age-related host factors that may protect children from COVID-19 and complications associated, and addresses the confusion around seropositivity and immunity.


Subject(s)
Antibodies, Viral/blood , Betacoronavirus/pathogenicity , Coronaviridae Infections/prevention & control , Coronaviridae/pathogenicity , Coronavirus Infections/prevention & control , Pandemics/prevention & control , Pneumonia, Viral/prevention & control , Adaptive Immunity/drug effects , Adolescent , Asymptomatic Diseases , Betacoronavirus/drug effects , Betacoronavirus/immunology , COVID-19 , Child , Coronaviridae/drug effects , Coronaviridae/immunology , Coronaviridae Infections/epidemiology , Coronaviridae Infections/immunology , Coronaviridae Infections/virology , Coronavirus Infections/epidemiology , Coronavirus Infections/immunology , Coronavirus Infections/virology , Cross Protection , Female , Humans , Immune Evasion/genetics , Immune Evasion/immunology , Immunity, Innate/drug effects , Male , Pneumonia, Viral/epidemiology , Pneumonia, Viral/immunology , Pneumonia, Viral/virology , SARS-CoV-2 , T-Lymphocyte Subsets/immunology , T-Lymphocyte Subsets/virology , United Kingdom/epidemiology , Vaccination , Young Adult
8.
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
9.
Med Sci (Paris) ; 36(8-9): 775-782, 2020.
Article in French | MEDLINE | ID: mdl-32755537

ABSTRACT

The recent emergence of a new coronavirus, SARS-CoV-2, responsible for COVID-19, is a new warning of the risk to public health represented by viral zoonoses and in particular by coronaviruses. Mainly described as being able to infect the upper and lower respiratory tract, coronaviruses can also infect the central and peripheral nervous systems as many other respiratory viruses, such as influenza or respiratory syncytial virus. Viral infections of the nervous system are a major public health concern as they can cause devastating illnesses up to death, especially when they occur in the elderly, who are more susceptible to these infections. Knowledge concerning the pathophysiology of recently emerging coronaviruses (MERS-CoV, SARS-CoV and SARS-CoV-2) and how they reach the central nervous system are very sketchy and the work in progress aims in particular to better understand their biology and the mechanisms associated with neurological damage. In this review we will discuss the current state of knowledge on the neurotropism of human coronaviruses and the associated mechanisms by developing in particular the latest data concerning SARS-CoV-2.


TITLE: Les atteintes neurologiques liées au SARS-CoV-2 et autres coronavirus humains. ABSTRACT: L'émergence récente d'un nouveau coronavirus, le SARS-CoV-2, responsable de la maladie appelée COVID-19, est un nouvel avertissement du risque pour la santé publique représenté par les zoonoses virales et notamment par les coronavirus. Principalement connus pour leur capacité à infecter les voies respiratoires supérieures et inférieures, les coronavirus peuvent également affecter le système nerveux central et périphérique, comme c'est le cas pour de nombreux virus respiratoires, tels que les virus influenza ou le virus respiratoire syncytial. Les infections du système nerveux sont un problème important de santé publique car elles peuvent provoquer des atteintes dévastatrices allant jusqu'au décès du patient, en particulier lorsqu'elles surviennent chez les personnes fragilisées ou âgées plus sensibles à ce type d'infection. Les connaissances de la physiopathologie des infections par les coronavirus émergents (MERS-CoV, SARS-CoV et SARS-CoV-2) et leurs moyens d'accéder au système nerveux central sont, pour l'heure, très sommaires. Les travaux en cours visent notamment à mieux appréhender les mécanismes associés aux atteintes neurologiques observées. Dans cette revue nous aborderons l'état des connaissances actuelles sur le neurotropisme des coronavirus humains et les mécanismes associés en développant tout particulièrement les dernières données concernant le SARS-CoV-2.


Subject(s)
Betacoronavirus/pathogenicity , Coronavirus Infections/complications , Nervous System Diseases/etiology , Pandemics , Pneumonia, Viral/complications , Animals , Biological Transport , COVID-19 , COVID-19 Testing , Clinical Laboratory Techniques , Communicable Diseases, Emerging , Coronaviridae/pathogenicity , Coronaviridae/physiology , Coronaviridae/ultrastructure , Coronaviridae Infections/complications , Coronavirus Infections/diagnosis , Coronavirus Infections/drug therapy , Coronavirus Infections/physiopathology , Humans , Nervous System/virology , Nervous System Diseases/diagnosis , Nervous System Diseases/therapy , Nervous System Diseases/virology , Organ Specificity , Pneumonia, Viral/diagnosis , Pneumonia, Viral/physiopathology , SARS-CoV-2 , Viral Tropism , Virulence , Virus Replication , Zoonoses , COVID-19 Drug Treatment
10.
Cell Physiol Biochem ; 54(4): 767-790, 2020 Aug 25.
Article in English | MEDLINE | ID: mdl-32830930

ABSTRACT

The pandemic of the severe acute respiratory syndrome coronavirus (SARS-CoV)-2 at the end of 2019 marked the third outbreak of a highly pathogenic coronavirus affecting the human population in the past twenty years. Cross-species zoonotic transmission of SARS-CoV-2 has caused severe pathogenicity and led to more than 655,000 fatalities worldwide until July 28, 2020. Outbursts of this virus underlined the importance of controlling infectious pathogens across international frontiers. Unfortunately, there is currently no clinically approved antiviral drug or vaccine against SARS-CoV-2, although several broad-spectrum antiviral drugs targeting multiple RNA viruses have shown a positive response and improved recovery in patients. In this review, we compile our current knowledge of the emergence, transmission, and pathogenesis of SARS-CoV-2 and explore several features of SARS-CoV-2. We emphasize the current therapeutic approaches used to treat infected patients. We also highlight the results of in vitro and in vivo data from several studies, which have broadened our knowledge of potential drug candidates for the successful treatment of patients infected with and discuss possible virus and host-based treatment options against SARS-CoV-2.


Subject(s)
Betacoronavirus , Coronavirus Infections , Pandemics , Pneumonia, Viral , Animals , Antiviral Agents/pharmacology , Antiviral Agents/therapeutic use , Betacoronavirus/drug effects , Betacoronavirus/genetics , Betacoronavirus/physiology , COVID-19 , COVID-19 Vaccines , Coronaviridae/pathogenicity , Coronaviridae Infections/epidemiology , Coronaviridae Infections/virology , Coronavirus Infections/drug therapy , Coronavirus Infections/epidemiology , Coronavirus Infections/prevention & control , Coronavirus Infections/therapy , Coronavirus Infections/transmission , Cytokine Release Syndrome/etiology , Cytokine Release Syndrome/prevention & control , Cytokines/antagonists & inhibitors , Drug Delivery Systems , Endocytosis/drug effects , Forecasting , Genome, Viral , Global Health , Humans , Immunity, Herd , Immunization, Passive , Pandemics/prevention & control , Peptide Hydrolases/pharmacology , Peptide Hydrolases/therapeutic use , Pneumonia, Viral/drug therapy , Pneumonia, Viral/epidemiology , Pneumonia, Viral/prevention & control , Pneumonia, Viral/transmission , RNA, Viral/genetics , Receptors, Coronavirus , Receptors, Virus/antagonists & inhibitors , Receptors, Virus/metabolism , SARS-CoV-2 , Spike Glycoprotein, Coronavirus/antagonists & inhibitors , Spike Glycoprotein, Coronavirus/metabolism , Viral Vaccines , Virus Internalization/drug effects , Virus Replication/drug effects , Zoonoses , COVID-19 Drug Treatment , COVID-19 Serotherapy
11.
J Neurol Sci ; 414: 116884, 2020 07 15.
Article in English | MEDLINE | ID: mdl-32464367

ABSTRACT

A comprehensive review of the neurological disorders reported during the current COVID-19 pandemic demonstrates that infection with SARS-CoV-2 affects the central nervous system (CNS), the peripheral nervous system (PNS) and the muscle. CNS manifestations include: headache and decreased responsiveness considered initial indicators of potential neurological involvement; anosmia, hyposmia, hypogeusia, and dysgeusia are frequent early symptoms of coronavirus infection. Respiratory failure, the lethal manifestation of COVID-19, responsible for 264,679 deaths worldwide, is probably neurogenic in origin and may result from the viral invasion of cranial nerve I, progressing into rhinencephalon and brainstem respiratory centers. Cerebrovascular disease, in particular large-vessel ischemic strokes, and less frequently cerebral venous thrombosis, intracerebral hemorrhage and subarachnoid hemorrhage, usually occur as part of a thrombotic state induced by viral attachment to ACE2 receptors in endothelium causing widespread endotheliitis, coagulopathy, arterial and venous thromboses. Acute hemorrhagic necrotizing encephalopathy is associated to the cytokine storm. A frontal hypoperfusion syndrome has been identified. There are isolated reports of seizures, encephalopathy, meningitis, encephalitis, and myelitis. The neurological diseases affecting the PNS and muscle in COVID-19 are less frequent and include Guillain-Barré syndrome; Miller Fisher syndrome; polyneuritis cranialis; and rare instances of viral myopathy with rhabdomyolysis. The main conclusion of this review is the pressing need to define the neurology of COVID-19, its frequency, manifestations, neuropathology and pathogenesis. On behalf of the World Federation of Neurology we invite national and regional neurological associations to create local databases to report cases with neurological manifestations observed during the on-going pandemic. International neuroepidemiological collaboration may help define the natural history of this worldwide problem.


Subject(s)
Betacoronavirus , Cerebrovascular Disorders/etiology , Coronavirus Infections/complications , Nervous System Diseases/etiology , Neuromuscular Diseases/etiology , Pandemics , Pneumonia, Viral/complications , Registries , Adult , Angiotensin-Converting Enzyme 2 , Animals , COVID-19 , Cerebrovascular Disorders/physiopathology , Communicable Diseases, Emerging/epidemiology , Communicable Diseases, Emerging/virology , Coronaviridae/pathogenicity , Coronaviridae/physiology , Coronaviridae/ultrastructure , Coronavirus Infections/epidemiology , Coronavirus Infections/physiopathology , Coronavirus Infections/veterinary , Coronavirus Infections/virology , Cytokine Release Syndrome/etiology , Cytokine Release Syndrome/physiopathology , Endothelium, Vascular/pathology , Endothelium, Vascular/virology , Humans , Models, Animal , Nervous System Diseases/physiopathology , Neuromuscular Diseases/physiopathology , Organ Specificity , Peptidyl-Dipeptidase A/physiology , Pneumonia, Viral/physiopathology , SARS-CoV-2 , Thrombophilia/etiology , Thrombophilia/physiopathology , Viral Tropism
12.
Trends Microbiol ; 26(7): 598-610, 2018 07.
Article in English | MEDLINE | ID: mdl-29268982

ABSTRACT

Viruses from the Coronaviridae, Togaviridae, and Hepeviridae families ​all contain genes that encode a conserved protein domain, called a macrodomain; however, the role of this domain during infection has remained enigmatic. The recent discovery that mammalian macrodomain proteins enzymatically remove ADP-ribose, a common post-translation modification, from proteins has led to an outburst of studies describing both the enzymatic activity and function of viral macrodomains. These new studies have defined these domains as de-ADP-ribosylating enzymes, which indicates that these viruses have evolved to counteract antiviral ADP-ribosylation, likely mediated by poly-ADP-ribose polymerases (PARPs). Here, we comprehensively review this rapidly expanding field, describing the structures and enzymatic activities of viral macrodomains, and discussing their roles in viral replication and pathogenesis.


Subject(s)
Protein Domains , Viral Nonstructural Proteins/chemistry , Virus Replication , Viruses/genetics , Viruses/pathogenicity , Adenosine Diphosphate Ribose/metabolism , Coronaviridae/genetics , Coronaviridae/pathogenicity , Hepevirus/genetics , Hepevirus/pathogenicity , Histones , Poly(ADP-ribose) Polymerases , Protein Processing, Post-Translational , Togaviridae/genetics , Togaviridae/pathogenicity , Viral Nonstructural Proteins/metabolism , Viruses/enzymology
13.
J Vet Diagn Invest ; 28(5): 486-97, 2016 Sep.
Article in English | MEDLINE | ID: mdl-27578872

ABSTRACT

Porcine deltacoronavirus (PDCoV) is a newly identified virus that has been detected in swine herds of North America associated with enteric disease. The aim of this study was to demonstrate the pathogenicity, course of infection, virus kinetics, and aerosol transmission of PDCoV using 87 conventional piglets and their 9 dams, including aerosol and contact controls to emulate field conditions. Piglets 2-4 days of age and their dams were administered an oronasal PDCoV inoculum with a quantitative real-time reverse transcription (qRT)-PCR quantification cycle (Cq) value of 22 that was generated from a field sample having 100% nucleotide identity to USA/Illinois121/2014 determined by metagenomic sequencing and testing negative for other enteric disease agents using standard assays. Serial samples of blood, serum, oral fluids, nasal and fecal swabs, and tissues from sequential autopsy, conducted daily on days 1-8 and regular intervals thereafter, were collected throughout the 42-day study for qRT-PCR, histopathology, and immunohistochemistry. Diarrhea developed in all inoculated and contact control pigs, including dams, by 2 days post-inoculation (dpi) and in aerosol control pigs and dams by 3-4 dpi, with resolution occurring by 12 dpi. Mild to severe atrophic enteritis with PDCoV antigen staining was observed in the small intestine of affected piglets from 2 to 8 dpi. Mesenteric lymph node and small intestine were the primary sites of antigen detection by immunohistochemistry, and virus RNA was detected in these tissues to the end of the study. Virus RNA was detectable in piglet fecal swabs to 21 dpi, and dams to 14-35 dpi.


Subject(s)
Coronaviridae/pathogenicity , Coronavirus Infections/veterinary , Swine Diseases/virology , Animals , Animals, Newborn , Coronaviridae/genetics , Coronavirus Infections/virology , Diarrhea/veterinary , Female , Immunohistochemistry/veterinary , RNA, Viral/genetics , Real-Time Polymerase Chain Reaction/veterinary , Survival Analysis , Swine , Swine Diseases/mortality , Swine Diseases/pathology
15.
Proc Natl Acad Sci U S A ; 113(11): 3048-53, 2016 Mar 15.
Article in English | MEDLINE | ID: mdl-26976607

ABSTRACT

Outbreaks from zoonotic sources represent a threat to both human disease as well as the global economy. Despite a wealth of metagenomics studies, methods to leverage these datasets to identify future threats are underdeveloped. In this study, we describe an approach that combines existing metagenomics data with reverse genetics to engineer reagents to evaluate emergence and pathogenic potential of circulating zoonotic viruses. Focusing on the severe acute respiratory syndrome (SARS)-like viruses, the results indicate that the WIV1-coronavirus (CoV) cluster has the ability to directly infect and may undergo limited transmission in human populations. However, in vivo attenuation suggests additional adaptation is required for epidemic disease. Importantly, available SARS monoclonal antibodies offered success in limiting viral infection absent from available vaccine approaches. Together, the data highlight the utility of a platform to identify and prioritize prepandemic strains harbored in animal reservoirs and document the threat posed by WIV1-CoV for emergence in human populations.


Subject(s)
Chiroptera/virology , Communicable Diseases, Emerging/virology , Coronaviridae Infections/virology , Coronaviridae/pathogenicity , Angiotensin-Converting Enzyme 2 , Animals , Antibodies, Monoclonal/immunology , Antibodies, Neutralizing/immunology , Antibodies, Viral/immunology , Cells, Cultured , Chlorocebus aethiops , Coronaviridae/genetics , Coronaviridae/immunology , Coronaviridae/isolation & purification , Coronaviridae/physiology , Coronaviridae Infections/prevention & control , Coronaviridae Infections/transmission , Coronaviridae Infections/veterinary , Cross Reactions , Encephalitis, Viral/virology , Epithelial Cells/virology , Host Specificity , Humans , Lung/cytology , Mice , Mice, Inbred BALB C , Mice, Transgenic , Models, Molecular , Peptidyl-Dipeptidase A/genetics , Peptidyl-Dipeptidase A/physiology , Point Mutation , Protein Conformation , Receptors, Virus/genetics , Receptors, Virus/physiology , Recombinant Fusion Proteins/metabolism , Severe acute respiratory syndrome-related coronavirus/immunology , Species Specificity , Spike Glycoprotein, Coronavirus/genetics , Spike Glycoprotein, Coronavirus/physiology , Vero Cells , Virus Replication , Zoonoses
16.
Virology ; 482: 51-9, 2015 Aug.
Article in English | MEDLINE | ID: mdl-25817405

ABSTRACT

Porcine deltacoronavirus (PDCoV) was first identified in Hong Kong in 2009-2010 and reported in United States swine for the first time in February 2014. However, diagnostic tools other than polymerase chain reaction for PDCoV detection were lacking and Koch's postulates had not been fulfilled to confirm the pathogenic potential of PDCoV. In the present study, PDCoV peptide-specific rabbit antisera were developed and used in immunofluorescence and immunohistochemistry assays to assist PDCoV diagnostics. The pathogenicity and pathogenesis of PDCoV was investigated following orogastric inoculation of 5-day-old piglets with a plaque-purified PDCoV cell culture isolate (3 × 10(4) TCID50 per pig). The PDCoV-inoculated piglets developed mild to moderate diarrhea, shed increasing amount of virus in rectal swabs from 2 to 7 days post inoculation, and developed macroscopic and microscopic lesions in small intestines with viral antigen confirmed by immunohistochemistry staining. This study experimentally confirmed PDCoV pathogenicity and characterized PDCoV pathogenesis in neonatal piglets.


Subject(s)
Coronaviridae/isolation & purification , Coronaviridae/pathogenicity , Coronavirus Infections/pathology , Coronavirus Infections/virology , Swine Diseases/pathology , Swine Diseases/virology , Animals , Antigens, Viral/analysis , Cell Culture Techniques , Coronaviridae/genetics , Diarrhea/pathology , Diarrhea/virology , Disease Models, Animal , Immunohistochemistry , Intestine, Small/pathology , Molecular Sequence Data , RNA, Viral/chemistry , RNA, Viral/genetics , Sequence Analysis, DNA , Swine , Virus Cultivation , Virus Shedding
17.
Virus Res ; 202: 120-34, 2015 Apr 16.
Article in English | MEDLINE | ID: mdl-25445340

ABSTRACT

Coronaviruses are a large group of enveloped, single-stranded positive-sense RNA viruses that infect a wide range of avian and mammalian species, including humans. The emergence of deadly human coronaviruses, severe acute respiratory syndrome coronavirus (SARS-CoV), and Middle East respiratory syndrome coronavirus (MERS-CoV) have bolstered research in these viral and often zoonotic pathogens. While coronavirus cell and tissue tropism, host range, and pathogenesis are initially controlled by interactions between the spike envelope glycoprotein and host cell receptor, it is becoming increasingly apparent that proteolytic activation of spike by host cell proteases also plays a critical role. Coronavirus spike proteins are the main determinant of entry as they possess both receptor binding and fusion functions. Whereas binding to the host cell receptor is an essential first step in establishing infection, the proteolytic activation step is often critical for the fusion function of spike, as it allows for controlled release of the fusion peptide into target cellular membranes. Coronaviruses have evolved multiple strategies for proteolytic activation of spike, and a large number of host proteases have been shown to proteolytically process the spike protein. These include, but are not limited to, endosomal cathepsins, cell surface transmembrane protease/serine (TMPRSS) proteases, furin, and trypsin. This review focuses on the diversity of strategies coronaviruses have evolved to proteolytically activate their fusion protein during spike protein biosynthesis and the critical entry step of their life cycle, and highlights important findings on how proteolytic activation of coronavirus spike influences tissue and cell tropism, host range and pathogenicity.


Subject(s)
Coronaviridae/physiology , Coronaviridae/pathogenicity , Host-Pathogen Interactions , Peptide Hydrolases/metabolism , Spike Glycoprotein, Coronavirus/metabolism , Viral Tropism , Host Specificity , Humans , Proteolysis
18.
J Virol ; 88(10): 5209-12, 2014 May.
Article in English | MEDLINE | ID: mdl-24600003

ABSTRACT

The identification of Middle East respiratory syndrome coronavirus (MERS-CoV) in 2012 reaffirmed the importance of understanding how coronaviruses emerge, infect, and cause disease. By comparing what is known about severe acute respiratory syndrome coronavirus (SARS-CoV) to what has recently been found for MERS-CoV, researchers are discovering similarities and differences that may be important for pathogenesis. Here we discuss what is known about each virus and what gaps remain in our understanding, especially concerning MERS-CoV.


Subject(s)
Communicable Diseases, Emerging/virology , Coronaviridae/isolation & purification , Coronavirus Infections/virology , Biomedical Research/trends , Coronaviridae/pathogenicity , Humans
19.
J Gen Virol ; 95(Pt 2): 408-412, 2014 Feb.
Article in English | MEDLINE | ID: mdl-24197535

ABSTRACT

The Middle East respiratory syndrome coronavirus (MERS-CoV) is a newly emerging highly pathogenic virus causing almost 50 % lethality in infected individuals. The development of a small-animal model is critical for the understanding of this virus and to aid in development of countermeasures against MERS-CoV. We found that BALB/c, 129/SvEv and 129/SvEv STAT1 knockout mice are not permissive to MERS-CoV infection. The lack of infection may be due to the low level of mRNA and protein for the MERS-CoV receptor, dipeptidyl peptidase 4 (DPP4), in the lungs of mice. The low level of DPP4 in the lungs likely contributes to the lack of viral replication in these mouse models and suggests that a transgenic mouse model expressing DPP4 to higher levels is necessary to create a mouse model for MERS-CoV.


Subject(s)
Coronaviridae/pathogenicity , Disease Resistance , Animals , Disease Models, Animal , Humans , Mice , Mice, Inbred BALB C , Mice, Knockout , Mice, SCID
20.
EMBO J ; 32(23): 3055-65, 2013 Nov 27.
Article in English | MEDLINE | ID: mdl-24169568

ABSTRACT

The IFNL4 gene is a recently discovered type III interferon, which in a significant fraction of the human population harbours a frameshift mutation abolishing the IFNλ4 ORF. The expression of IFNλ4 is correlated with both poor spontaneous clearance of hepatitis C virus (HCV) and poor response to treatment with type I interferon. Here, we show that the IFNL4 gene encodes an active type III interferon, named IFNλ4, which signals through the IFNλR1 and IL-10R2 receptor chains. Recombinant IFNλ4 is antiviral against both HCV and coronaviruses at levels comparable to IFNλ3. However, the secretion of IFNλ4 is impaired compared to that of IFNλ3, and this impairment is not due to a weak signal peptide, which was previously believed. We found that IFNλ4 gets N-linked glycosylated and that this glycosylation is required for secretion. Nevertheless, this glycosylation is not required for activity. Together, these findings result in the paradox that IFNλ4 is strongly antiviral but a disadvantage during HCV infection.


Subject(s)
Antiviral Agents/pharmacology , Coronaviridae Infections/prevention & control , Hepatitis C/prevention & control , Interleukins/metabolism , Receptors, Interferon/metabolism , Receptors, Interleukin/metabolism , Amino Acid Sequence , Blotting, Western , Cell Proliferation , Cells, Cultured , Coronaviridae/pathogenicity , Coronaviridae Infections/metabolism , Coronaviridae Infections/virology , Glycosylation , Hepacivirus/pathogenicity , Hepatitis C/metabolism , Hepatitis C/virology , Humans , Immunoenzyme Techniques , Interferon-gamma/metabolism , Interleukins/chemistry , Interleukins/genetics , Molecular Sequence Data , Protein Conformation , Protein Sorting Signals/physiology , RNA, Messenger/genetics , Real-Time Polymerase Chain Reaction , Receptors, Interferon/genetics , Receptors, Interleukin/genetics , Respiratory System/cytology , Respiratory System/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Sequence Homology, Amino Acid , Virus Replication , Interferon gamma Receptor
SELECTION OF CITATIONS
SEARCH DETAIL
...