Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 24
Filter
Add more filters










Publication year range
1.
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
2.
Chem Biol Interact ; 329: 109209, 2020 Sep 25.
Article in English | MEDLINE | ID: mdl-32750325

ABSTRACT

Kinetic modeling of the behavior of complex chemical and biochemical systems is an effective approach to study of the mechanisms of the process. A kinetic model of coronaviral infection development with a description of the dynamic behavior of the main variables, including the concentration of viral particles, affected cells, and pathogenic microflora, is proposed. Changes in the concentration of hydrogen ions in the lungs and the pH -dependence of carbonic anhydrase activity (a key breathing enzyme) are critical. A significant result is the demonstration of an acute bifurcation transition that determines life or system collapse. This transition is connected with exponential growth of concentrations of the process participants and with functioning of the key enzyme carbonic anhydrase in development of toxic effects. Physical and chemical interpretations of the therapeutic effects of the body temperature rise and the potential therapeutic effect of "thermoheliox" (respiration with a thermolized mixture of helium and oxygen) are given. The phenomenon of "thermovaccination" is predicted, which involves stimulation of the immune response by "thermoheliox".


Subject(s)
Coronaviridae Infections/metabolism , Helium/chemistry , Oxygen/chemistry , Adaptive Immunity , Body Temperature , Carbonic Anhydrases/metabolism , Coronaviridae Infections/pathology , Coronaviridae Infections/therapy , Helium/therapeutic use , Humans , Hydrogen-Ion Concentration , Kinetics , Lung/metabolism , Models, Theoretical , Oxygen/therapeutic use
3.
Int J Mol Sci ; 21(11)2020 May 28.
Article in English | MEDLINE | ID: mdl-32481719

ABSTRACT

Mucociliary clearance, mediated by a coordinated function of cilia bathing in the airway surface liquid (ASL) on the surface of airway epithelium, protects the host from inhaled pathogens and is an essential component of the innate immunity. ASL is composed of the superficial mucus layer and the deeper periciliary liquid. Ion channels, transporters, and pumps coordinate the transcellular and paracellular movement of ions and water to maintain the ASL volume and mucus hydration. microRNA (miRNA) is a class of non-coding, short single-stranded RNA regulating gene expression by post-transcriptional mechanisms. miRNAs have been increasingly recognized as essential regulators of ion channels and transporters responsible for ASL homeostasis. miRNAs also influence the airway host defense. We summarize the most up-to-date information on the role of miRNAs in ASL homeostasis and host-pathogen interactions in the airway and discuss concepts for miRNA-directed therapy.


Subject(s)
Coronaviridae Infections/metabolism , Host-Pathogen Interactions , MicroRNAs/genetics , Respiratory Mucosa/metabolism , Respiratory Tract Absorption , Animals , Coronaviridae Infections/genetics , Coronaviridae Infections/virology , Homeostasis , Humans , MicroRNAs/metabolism , Respiratory Mucosa/virology
4.
J Virol ; 92(4)2018 02 15.
Article in English | MEDLINE | ID: mdl-29093093

ABSTRACT

Coronaviruses recently emerged as major human pathogens causing outbreaks of severe acute respiratory syndrome and Middle East respiratory syndrome. They utilize the spike (S) glycoprotein anchored in the viral envelope to mediate host attachment and fusion of the viral and cellular membranes to initiate infection. The S protein is a major determinant of the zoonotic potential of coronaviruses and is also the main target of the host humoral immune response. We report here the 3.5-Å-resolution cryo-electron microscopy structure of the S glycoprotein trimer from the pathogenic porcine deltacoronavirus (PDCoV), which belongs to the recently identified Deltacoronavirus genus. Structural and glycoproteomics data indicate that the glycans of PDCoV S are topologically conserved compared with the human respiratory coronavirus NL63 S, resulting in similar surface areas being shielded from neutralizing antibodies and implying that both viruses are under comparable immune pressure in their respective hosts. The structure further reveals a shortened S2' activation loop, containing a reduced number of basic amino acids, which participates in rendering the spike largely protease resistant. This property distinguishes PDCoV S from recently characterized betacoronavirus S proteins and suggests that the S protein of enterotropic PDCoV has evolved to tolerate the protease-rich environment of the small intestine and to fine-tune its fusion activation to avoid premature triggering and reduction of infectivity.IMPORTANCE Coronaviruses use transmembrane S glycoprotein trimers to promote host attachment and fusion of the viral and cellular membranes. We determined a near-atomic-resolution cryo-electron microscopy structure of the S ectodomain trimer from the pathogenic PDCoV, which is responsible for diarrhea in piglets and has had devastating consequences for the swine industry worldwide. Structural and glycoproteomics data reveal that PDCoV S is decorated with 78 N-linked glycans obstructing the protein surface to limit accessibility to neutralizing antibodies in a way reminiscent of what has recently been described for a human respiratory coronavirus. PDCoV S is largely protease resistant, which distinguishes it from most other characterized coronavirus S glycoproteins and suggests that enteric coronaviruses have evolved to fine-tune fusion activation in the protease-rich environment of the small intestine of infected hosts.


Subject(s)
Coronaviridae Infections/virology , Coronaviridae/physiology , Membrane Fusion , Polysaccharides/metabolism , Spike Glycoprotein, Coronavirus/chemistry , Spike Glycoprotein, Coronavirus/metabolism , Virus Attachment , Animals , Coronaviridae/classification , Coronaviridae Infections/metabolism , Drosophila melanogaster/metabolism , Drosophila melanogaster/virology , Humans , Polysaccharides/chemistry , Protein Conformation , Swine , Swine Diseases/pathology , Swine Diseases/virology
5.
Virology ; 502: 33-38, 2017 02.
Article in English | MEDLINE | ID: mdl-27984784

ABSTRACT

Porcine deltacoronavirus (PDCoV) causes acute enteric disease and mortality in seronegative neonatal piglets. Previously we have demonstrated that PDCoV infection suppresses the production of interferon-beta (IFN-ß), while the detailed mechanisms are poorly understood. Here, we demonstrate that nonstructural protein 5 (nsp5) of PDCoV, the 3C-like protease, significantly inhibits Sendai virus (SEV)-induced IFN-ß production by targeting the NF-κB essential modulator (NEMO), confirmed by the diminished function of NEMO cleaved by PDCoV. The PDCoV nsp5 cleavage site in the NEMO protein was identified as glutamine 231, and was identical to the porcine epidemic diarrhea virus nsp5 cleavage site, revealing the likelihood of a common target in NEMO for coronaviruses. Furthermore, this cleavage impaired the ability of NEMO to activate the IFN response and downstream signaling. Taken together, our findings reveal PDCoV nsp5 to be a newly identified IFN antagonist and enhance the understanding of immune evasion by deltacoronaviruses.


Subject(s)
Coronaviridae Infections/veterinary , Coronaviridae/enzymology , Cysteine Endopeptidases/metabolism , I-kappa B Kinase/metabolism , Interferon-beta/metabolism , Swine Diseases/enzymology , Viral Nonstructural Proteins/metabolism , Animals , Coronaviridae/genetics , Coronaviridae Infections/enzymology , Coronaviridae Infections/metabolism , Coronaviridae Infections/virology , Coronavirus 3C Proteases , Cysteine Endopeptidases/genetics , Host-Pathogen Interactions , I-kappa B Kinase/genetics , Interferon-beta/genetics , Protein Processing, Post-Translational , Swine , Swine Diseases/genetics , Swine Diseases/metabolism , Swine Diseases/virology , Viral Nonstructural Proteins/genetics
6.
Virology ; 497: 185-197, 2016 10.
Article in English | MEDLINE | ID: mdl-27479465

ABSTRACT

Coronavirus spike proteins mediate host-cell-attachment and virus entry. Virus replication takes place within the host cell cytosol, whereas assembly and budding occur at the endoplasmic reticulum-Golgi intermediate compartment. In this study we demonstrated that the last 39 amino acid stretches of Alphacoronavirus spike cytoplasmic domains of the human coronavirus 229E, NL63, and the porcine transmissible gastroenteritis virus TGEV interact with tubulin alpha and beta chains. In addition, a partial co-localization of TGEV spike proteins with authentic host cell ß-tubulin was observed. Furthermore, drug-induced microtubule depolymerization led to changes in spike protein distribution, a reduction in the release of infectious virus particles and less amount of spike protein incorporated into virions. These data demonstrate that interaction of Alphacoronavirus spike proteins with tubulin supports S protein transport and incorporation into virus particles.


Subject(s)
Coronaviridae Infections/metabolism , Coronaviridae Infections/virology , Coronaviridae/physiology , Spike Glycoprotein, Coronavirus/metabolism , Tubulin/metabolism , Virus Assembly , Virus Replication , Animals , Cell Line , Coronaviridae/drug effects , Gastroenteritis, Transmissible, of Swine/metabolism , Gastroenteritis, Transmissible, of Swine/virology , Humans , Intracellular Space/metabolism , Intracellular Space/virology , Nocodazole/pharmacology , Protein Binding , Protein Interaction Domains and Motifs , Protein Transport , Spike Glycoprotein, Coronavirus/chemistry , Swine , Virus Assembly/drug effects , Virus Release , Virus Replication/drug effects
7.
Virology ; 450-451: 64-70, 2014 Feb.
Article in English | MEDLINE | ID: mdl-24503068

ABSTRACT

Coronaviruses encode papain-like proteases (PLpro) that are often multifunctional enzymes with protease activity to process the viral replicase polyprotein and deubiquitinating (DUB)/deISGylating activity, which is hypothesized to modify the innate immune response to infection. Here, we investigate the predicted DUB activity of the PLpro domain of the recently described Middle East Respiratory Syndrome Coronavirus (MERS-CoV). We found that expression of MERS-CoV PLpro reduces the levels of ubiquitinated and ISGylated host cell proteins; consistent with multifunctional PLpro activity. Further, we compared the ability of MERS-CoV PLpro and Severe Acute Respiratory Syndrome Coronavirus (SARS-CoV) PLpro to block innate immune signaling of proinflammatory cytokines. We show that expression of SARS-CoV and MERS-CoV PLpros blocks upregulation of cytokines CCL5, IFN-ß and CXCL10 in stimulated cells. Overall these results indicate that the PLpro domains of MERS-CoV and SARS-CoV have the potential to modify the innate immune response to viral infection and contribute to viral pathogenesis.


Subject(s)
Coronaviridae Infections/virology , Coronaviridae/enzymology , Papain/metabolism , Viral Proteins/metabolism , Amino Acid Sequence , Cell Line , Coronaviridae/chemistry , Coronaviridae/genetics , Coronaviridae Infections/genetics , Coronaviridae Infections/metabolism , Cytokines/genetics , Cytokines/metabolism , Glycosylation , Humans , Molecular Sequence Data , Papain/chemistry , Papain/genetics , Protein Structure, Tertiary , Severe acute respiratory syndrome-related coronavirus/enzymology , Severe acute respiratory syndrome-related coronavirus/genetics , Ubiquitination , Ubiquitins/genetics , Ubiquitins/metabolism , Viral Proteins/chemistry , Viral Proteins/genetics
8.
J Gen Virol ; 95(Pt 3): 614-626, 2014 Mar.
Article in English | MEDLINE | ID: mdl-24362959

ABSTRACT

The emerging Middle East respiratory syndrome coronavirus (MERS-CoV) causes severe pulmonary disease in humans and represents the second example of a highly pathogenic coronavirus (CoV) following severe acute respiratory syndrome coronavirus (SARS-CoV). Genomic studies revealed that two viral proteases, papain-like protease (PLpro) and 3C-like protease (3CLpro), process the polyproteins encoded by the MERS-CoV genomic RNA. We previously reported that SARS-CoV PLpro acts as both deubiquitinase (DUB) and IFN antagonist, but the function of the MERS-CoV PLpro was poorly understood. In this study, we characterized MERS-CoV PLpro, which is a protease and can recognize and process the cleavage sites (CS) of nsp1-2, nsp2-3 and nsp3-4. The LXGG consensus cleavage sites in the N terminus of pp1a/1ab, which is generally essential for CoV PLpro-mediated processing, were also characterized in MERS-CoV. MERS-CoV PLpro, like human SARS-CoV PLpro and NL63-CoV PLP2, is a viral deubiquitinating enzyme. It acts on both K48- and K63-linked ubiquitination and ISG15-linked ISGylation. We confirmed that MERS-CoV PLpro acts as an IFN antagonist through blocking the phosphorylation and nuclear translocation of IFN regulatory factor 3 (IRF3). These findings indicate that MERS-CoV PLpro acts as a viral DUB and suppresses production of IFN-ß by an interfering IRF3-mediated signalling pathway, in addition to recognizing and processing the CS at the N terminus of replicase polyprotein to release the non-structural proteins. The characterization of proteolytic processing, DUB and IFN antagonist activities of MERS-CoV PLpro would reveal the interactions between MERS-CoV and its host, and be applicable to develop strategies targeting PLpro for the effective control of MERS-CoV infection.


Subject(s)
Coronaviridae Infections/metabolism , Coronaviridae/enzymology , Interferon-beta/antagonists & inhibitors , Papain/metabolism , Ubiquitin-Specific Proteases/metabolism , Viral Proteins/metabolism , Amino Acid Sequence , Catalytic Domain , Coronaviridae/chemistry , Coronaviridae/genetics , Coronaviridae Infections/virology , Humans , Interferon Regulatory Factor-3/metabolism , Interferon-beta/metabolism , Molecular Sequence Data , Papain/chemistry , Papain/genetics , Phosphorylation , Polyproteins/genetics , Polyproteins/metabolism , Protein Processing, Post-Translational , Proteolysis , Sequence Alignment , Ubiquitin , Ubiquitin-Specific Proteases/chemistry , Ubiquitin-Specific Proteases/genetics , Viral Proteins/chemistry , Viral Proteins/genetics
9.
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
10.
J Immunol ; 186(6): 3642-52, 2011 Mar 15.
Article in English | MEDLINE | ID: mdl-21317392

ABSTRACT

Acute viral encephalitis requires rapid pathogen elimination without significant bystander tissue damage. In this article, we show that IL-10, a potent anti-inflammatory cytokine, is produced transiently at the peak of infection by CD8 T cells in the brains of coronavirus-infected mice. IL-10(+)CD8 and IL-10(-)CD8 T cells interconvert during acute disease, possibly based on recent Ag exposure. Strikingly, IL-10(+)CD8 T cells were more highly activated and cytolytic than IL-10(-)CD8 T cells, expressing greater levels of proinflammatory cytokines and chemokines, as well as cytotoxic proteins. Even though these cells are highly proinflammatory, IL-10 expressed by these cells was functional. Furthermore, IL-10 produced by CD8 T cells diminished disease severity in mice with coronavirus-induced acute encephalitis, suggesting a self-regulatory mechanism that minimizes immunopathological changes.


Subject(s)
Cytotoxicity, Immunologic , Encephalomyelitis, Acute Disseminated/immunology , Interleukin-10/biosynthesis , Lymphocyte Activation/immunology , T-Lymphocytes, Cytotoxic/immunology , T-Lymphocytes, Cytotoxic/metabolism , Adoptive Transfer , Animals , Cell Line, Tumor , Cells, Cultured , Coronaviridae Infections/immunology , Coronaviridae Infections/metabolism , Coronaviridae Infections/prevention & control , Encephalomyelitis, Acute Disseminated/pathology , Encephalomyelitis, Acute Disseminated/prevention & control , Inflammation Mediators/metabolism , Interleukin-10/deficiency , Interleukin-10/physiology , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Murine hepatitis virus/immunology , T-Lymphocytes, Cytotoxic/transplantation
11.
Res Vet Sci ; 81(3): 310-5, 2006 Dec.
Article in English | MEDLINE | ID: mdl-16759679

ABSTRACT

Brush border membrane-bound digestive enzymes such as disaccharidases (lactase, sucrase, and maltase), leucine aminopeptidase N, and alkaline phosphatase were measured in jejunum from pigs experimentally infected with porcine epidemic diarrhea virus (PEDV). Three piglets from the infected and control groups were euthanized by electrocution and subjected to necropsy at 24, 36, 48, 60, and 72 hours post-inoculation (hpi). The infection of PEDV to jejunum resulted in significant decreases in brush border membrane-bound digestive enzymes such as disaccharidases (lactase, sucrase, and maltase), leucine aminopeptidase N, and alkaline phosphatase. PEDV replication results in massive destruction of villous enterocytes leading to a marked reduction of intestinal epithelial surface and brush border membrane-bound digestive enzyme activity. Reduced enzymatic activity and villous atrophy in the small intestine is thought to result in a maldigestive and malabsorptive diarrhea.


Subject(s)
Coronaviridae Infections/veterinary , Coronaviridae/pathogenicity , Jejunum/enzymology , Microvilli/enzymology , Swine Diseases/metabolism , Alkaline Phosphatase/metabolism , Animals , Coronaviridae Infections/metabolism , Disaccharidases/metabolism , Leucyl Aminopeptidase/metabolism , Swine
12.
Can J Vet Res ; 57(3): 152-8, 1993 Jul.
Article in English | MEDLINE | ID: mdl-8395329

ABSTRACT

In preliminary studies feeding the poorly absorbed carbohydrate sorbitol at 2.3 g/kg body weight as an indication of maximal fermentative capacity failed to produce the expected large increase in breath hydrogen excretion but did produce a transient diarrhea in five out of six control calves. Twelve healthy control and eighteen diarrheic calves were fed lactose or D-xylose on consecutive days at 1.15 g/kg body weight and a concentration of 46 g/L. Breath and blood samples were collected at 1 h intervals from 0 to 7 h. After administration of lactose, there was a significant increase in breath hydrogen excretion in diarrheic versus control calves. The increase in plasma glucose concentrations was delayed in diarrheic calves but the area under the absorption curve was similar in control and diarrheic calves. After administration of D-xylose, breath hydrogen excretion did not increase significantly but plasma D-xylose concentrations were significantly reduced in diarrheic calves. The pathogens commonly isolated from the feces were Cryptosporidium species, rotavirus and coronavirus. The number of pathogens and the severity of the calves' acid-base deficit were not related to the severity of carbohydrate malabsorption. Decreased absorption of lactose and D-xylose may be the result of intestinal villous atrophy caused by viral or parasite infection. It was concluded that carbohydrate malabsorption rather than a specific lactose maldigestion is a significant problem in diarrheic calves. Diarrheic calves appear to digest and absorb lactose when fed in small amounts.


Subject(s)
Cattle Diseases/metabolism , Diarrhea/veterinary , Lactose Intolerance/veterinary , Malabsorption Syndromes/veterinary , Xylose/metabolism , Animals , Blood Glucose/analysis , Breath Tests , Cattle , Coronaviridae Infections/metabolism , Coronaviridae Infections/veterinary , Cryptosporidiosis/metabolism , Diarrhea/metabolism , Female , Fermentation , Intestinal Absorption , Lactose Intolerance/metabolism , Malabsorption Syndromes/metabolism , Male , Regression Analysis , Rotavirus Infections/metabolism , Rotavirus Infections/veterinary , Sorbitol/metabolism , Time Factors , Xylose/blood
13.
J Gen Virol ; 73 ( Pt 4): 901-6, 1992 Apr.
Article in English | MEDLINE | ID: mdl-1321878

ABSTRACT

The importance of N-acetyl-9-O-acetylneuraminic acid (Neu5,9Ac2) as a receptor determinant for bovine coronavirus (BCV) on cultured cells was analysed. Pretreatment of MDCK I (Madin Darby canine kidney) cells with neuraminidase or acetylesterase rendered the cells resistant to infection by BCV. The receptors on a human (CaCo-2) and a porcine (LLC-PK1) epithelial cell line were also found to be sensitive to neuraminidase treatment. The susceptibility to infection by BCV was restored after resialylation of asialo-MDCK I cells with Neu5,9Ac2. Transfer of sialic acid lacking a 9-O-acetyl group was ineffective in this respect. These results demonstrate that 9-O-acetylated sialic acid is used as a receptor determinant by BCV to infect cultured cells. The possibility is discussed that the initiation of a BCV infection involves the recognition of different types of receptors, a first receptor for primary attachment and a second receptor to mediate the fusion between the viral envelope and the cellular membrane.


Subject(s)
Cattle/microbiology , Coronaviridae Infections/metabolism , Coronaviridae/pathogenicity , Sialic Acids/chemistry , Sialic Acids/metabolism , Acetylesterase/pharmacology , Animals , Binding Sites , Cell Membrane/drug effects , Cells, Cultured , Coronaviridae/metabolism , N-Acetylneuraminic Acid , Neuraminidase/pharmacology , Species Specificity
14.
J Nutr ; 122(2): 333-9, 1992 Feb.
Article in English | MEDLINE | ID: mdl-1310111

ABSTRACT

The effect of infection with infectious bronchitis virus (IBV) and reovirus (RV) on vitamin A status was investigated in chickens with a normal or marginal intake of vitamin A. At the age of 4 wk, chickens were infected with either IBV or RV, primarily affecting the respiratory or intestinal tract, respectively. Both viruses lowered plasma retinol levels significantly. The effect was more pronounced in chickens fed a diet marginally deficient in vitamin A than in those fed a diet adequate in vitamin A. Concentrations of retinol-binding protein, transthyretin and albumin in RV-infected chickens were also significantly lower than in noninfected chickens fed the same diets; in chickens infected with IBV, there was no effect. These results suggest that the reduced vitamin A status of IBV-infected chickens could be attributed to increased rate of utilization by tissues. In RV infection, this mechanism could be involved but impaired absorption of nutrients (including vitamin A) and direct loss of nutrients via the intestinal tract could also be important.


Subject(s)
Chickens , Coronaviridae Infections/veterinary , Poultry Diseases/metabolism , Reoviridae Infections/veterinary , Vitamin A/analysis , Animals , Coronaviridae Infections/metabolism , Female , Infectious bronchitis virus , Liver/chemistry , Prealbumin/analysis , Reoviridae Infections/metabolism , Retinol-Binding Proteins/analysis , Retinol-Binding Proteins, Plasma , Serum Albumin/analysis , Vitamin A/blood , Vitamin A Deficiency/complications , Vitamin A Deficiency/veterinary
15.
Invest Ophthalmol Vis Sci ; 33(1): 60-7, 1992 Jan.
Article in English | MEDLINE | ID: mdl-1309730

ABSTRACT

Inoculation of the neurotropic coronavirus mouse hepatitis virus strain JHM intravitreally or into the anterior chamber causes acute infection of the retinal pigment epithelium (RPE) and neural retina. Weeks later, many retinas have foci of moderate to severe atrophy. The effect of coronavirus infection (after intravitreal inoculation) was examined on interphotoreceptor retinoid-binding protein (IRBP), the glycolipoprotein in the interphotoreceptor matrix (IPM) thought to transport retinoids between the photoreceptors and the RPE. Changes in IRBP distribution accompanied virus-associated retinal pathology, including photoreceptor loss and RPE abnormalities. Immunohistochemistry on days 3 and 6 showed that IRBP had diffused into the neural retina away from the IPM. The IRBP became localized abnormally in the same areas as virus-induced lesions, shown by staining adjacent sections with a monoclonal antibody specific for the viral nucleocapsid protein. Moreover, the level of IRBP in isolated retinas, measured in an immunoslot-blot assay, decreased significantly by day 3 and remained low through day 23. This decrease was confirmed in eyecups isolated on day 6. It may be caused in part by loss of photoreceptors and diffusion of IRBP through the retina into the vitreous. These studies show that a virus may induce an acute, limited infection in the retina that can be cleared by the host. However, the infection initiated a series of events resulting in long-term reduction and redistribution of a critical photoreceptor protein.


Subject(s)
Coronaviridae Infections/metabolism , Eye Infections, Viral/metabolism , Eye Proteins/metabolism , Retinal Diseases/metabolism , Retinol-Binding Proteins/metabolism , Animals , Antigens, Viral/analysis , Coronaviridae Infections/immunology , Coronaviridae Infections/pathology , Eye Infections, Viral/immunology , Eye Infections, Viral/pathology , Male , Mice , Mice, Inbred BALB C , Pigment Epithelium of Eye/pathology , Retinal Diseases/immunology , Retinal Diseases/microbiology , Retinal Diseases/pathology
16.
J Med Virol ; 35(2): 142-9, 1991 Oct.
Article in English | MEDLINE | ID: mdl-1662706

ABSTRACT

Cellular susceptibility to fusion mediated by murine coronavirus (mouse hepatitis virus, MHV strain A59) was separated into lipid-dependent and lipid-independent mechanisms with the use of subclones and selected mutants of mouse L-2 fibroblasts. Fusion-resistant L-2 cell mutants had similar cholesterol and fatty acid composition as did their fusion-susceptible parent subclone, and were presumably deficient in a genetically mutable non-lipid, host cell factor (e.g., fusion protein receptor). On the other hand, cellular sensitivity to virus fusion, which is known to be influenced by cell cholesterol content [Daya et al., 1988], was shown further to be modulated by homeostatic alterations in fatty acid metabolism. Cholesterol supplementation of mouse L-2 fibroblasts or of peritoneal macrophages from MHV-susceptible mice elevated susceptibility to viral fusion. Increased fusion susceptibility occurred in cholesterol-supplemented L-2 cells in the absence of any detectable alterations in host cell fatty acid composition, thus demonstrating fusion enhancement by cholesterol alone. L-2 cells cloned by limiting dilution in normal (not cholesterol-supplemented) medium were found to be heterogeneous in cholesterol content. Interestingly, high cholesterol-containing subclones had increased levels of C-18:0, C-18:2, C-20:4, and C-22:6 and markedly reduced levels of C-18:1 fatty acids when compared to low cholesterol-containing subclones. High cholesterol-containing subclones did not show enhanced susceptibility to viral fusion, suggesting that homeostatic alteration of fatty acid metabolism compensated for the increased cholesterol levels and countered the normally fusion-enhancing effect of cholesterol alone.(ABSTRACT TRUNCATED AT 250 WORDS)


Subject(s)
Cholesterol/metabolism , Coronaviridae/growth & development , Fatty Acids/metabolism , Animals , Cell Fusion , Cells, Cultured , Cholesterol, Dietary/pharmacology , Chromatography, Thin Layer , Coronaviridae Infections/metabolism , Coronaviridae Infections/microbiology , Fibroblasts/microbiology , Homeostasis , Macrophages/microbiology , Mice , Mice, Inbred Strains
17.
Virus Res ; 20(1): 45-58, 1991 Jun.
Article in English | MEDLINE | ID: mdl-1656623

ABSTRACT

Six variant viruses of the JHMV strain of murine coronavirus with large (cl-2, CNSV, DL and DS) or small (sp-4 and JHM-X) S proteins were compared in terms of their relative neurovirulence in weanling Lewis rats. Inoculation of various doses of the variants revealed that the cl-2 and CNSV were highly virulent and DL and DS were low-virulent, while sp-4 and JHM-X were avirulent. Pathological examination of rats infected with variants cl-2, DL and sp-4 showed that the cl-2 and DL induced severe and mild acute encephalomyelitis, respectively, while no lesions were observed in the central nervous system of rats infected with sp-4. Virus growth and distribution of antigen in rat brains correlated strongly with neurovirulence. These results suggest that S protein plays a role in neurovirulence in rats. In addition, these variant viruses were shown to be useful tools for further analysis of JHMV neurovirulence in animals as well as in cultured cells.


Subject(s)
Coronaviridae Infections/etiology , Coronaviridae/pathogenicity , Encephalomyelitis/etiology , Animals , Antigens, Viral , Coronaviridae/genetics , Coronaviridae/metabolism , Coronaviridae Infections/metabolism , Coronaviridae Infections/pathology , Encephalomyelitis/metabolism , Encephalomyelitis/pathology , Genetic Variation , Rats , Rats, Inbred Lew , Viral Proteins/genetics , Viral Proteins/metabolism , Virulence/genetics
18.
Am J Vet Res ; 51(5): 726-33, 1990 May.
Article in English | MEDLINE | ID: mdl-2159739

ABSTRACT

Seventy-four cats (52 treated and 22 untreated) were evaluated in efficacy studies of interferon (IFN), Propionibacterium acnes, or a combination of these drugs against experimentally induced feline infectious peritonitis (FIP). Cats were given doses of recombinant human leukocyte (alpha) IFN (rHuIFN-alpha), feline fibroblastic (beta) IFN (FIFN-beta) or P acnes at regular intervals before and after inoculation of virulent FIP virus (FIPV). Prophylactic and therapeutic administration of high doses (10(6) U/kg of body weight) or moderate doses (10(4) U/kg) of rHuIFN-alpha, FIFN-beta (10(3) u/kg), or P acnes (0.4 or 4 mg) did not significantly reduce mortality in treated vs untreated cats. However, the mean survival time in cats treated with 10(6) U of rHuIFN-alpha-/kg alone or combined with doses of P acnes was significantly (P = 0.03) increased after inoculation of highly lethal amounts (200 LD100) of FIPV vs survival time in untreated cats. Although P acnes alone was ineffective, there was some indication that a combination of P acnes and high doses of rHuIFN-alpha was more effective than rHuIFN-alpha alone. Seemingly, the efficacy of rHuIFn-alpha treatment was improved in cats challenge-exposed with less FIPV; in 1 trial, 4 of 5 cats (80%) treated with high doses of rHuIFN-alpha survived after inoculation of minimal lethal amounts (0.6 LD100) of FIPV, whereas only 2 of 5 untreated cats (40%) survived. Pretreatment of cats with 10(6) U of rHuIFN-alpha/kg resulted in detectable serum IFN activity 24 hours later; serum IFN activity was not detected in cats pretreated with P acnes, FIFN-beta, or 10(4) U of rHuIFn-alpha/kg.(ABSTRACT TRUNCATED AT 250 WORDS)


Subject(s)
Adjuvants, Immunologic/pharmacology , Cat Diseases/prevention & control , Coronaviridae Infections/prevention & control , Interferon Type I/pharmacology , Peritonitis/veterinary , Propionibacterium acnes , Animals , Antibody Formation , Cat Diseases/metabolism , Cat Diseases/microbiology , Cat Diseases/mortality , Cats , Coronaviridae , Coronaviridae Infections/metabolism , Coronaviridae Infections/microbiology , Coronaviridae Infections/mortality , Humans , Interferon Type I/blood , Peritonitis/metabolism , Peritonitis/microbiology , Peritonitis/mortality , Random Allocation , Specific Pathogen-Free Organisms , Time Factors
19.
Glia ; 2(5): 318-29, 1989.
Article in English | MEDLINE | ID: mdl-2478465

ABSTRACT

C57BI/6N mice develop a CNS demyelinating disease when inoculated intracranially at 4 weeks of age with the A59 strain of mouse hepatitis virus (MHV-A59). In order to explore the virus-host interactions, the histological features of the demyelinating disease were correlated with the spatial and temporal distribution of viral transcripts and the expression of oligodendrocyte-specific genes (myelin basic protein, proteolipid protein, myelin-associated glycoprotein, and 2',3' cyclic nucleotide 3'-phosphohydrolase) in the spinal cord of diseased mice. Three distinct phases in the disease were identified. In the first phase, 1 week postinfection (1 WPI), virus replication was widespread in both gray and white matter but was preferentially occurring in glial cells. In the ventral and dorsal root zones where viral transcripts were most abundant, all myelin gene transcripts were decreased before demyelination was seen. During the second phase of the disease (2-3 WPI), viral transcripts decreased in abundance and became restricted to the white matter. Numerous demyelinating lesions were observed and were characterized by inflammatory cells, paucity of oligodendrocytes, and a profound decrease of all myelin gene transcripts. In the third phase of the disease (4-6 WPI) no viral transcripts were detected, and remyelination began. In the lesions and the tissue surrounding them, transcripts of all myelin genes increased to levels above normal. The increased expression of myelin gene transcripts occurred in a synchronized manner and with a cellular distribution reminiscent of that seen in developmental myelination. These molecular events correlated with efficient remyelination and clinical recovery in this murine demyelinating disease.


Subject(s)
Coronaviridae Infections/metabolism , Demyelinating Diseases/genetics , Gene Expression Regulation, Viral , Gene Expression Regulation , Myelin Basic Protein/genetics , RNA, Messenger/metabolism , Animals , Demyelinating Diseases/microbiology , Demyelinating Diseases/pathology , Mice , Mice, Inbred C57BL , Myelin Basic Protein/metabolism , Spinal Cord/metabolism , Spinal Cord/ultrastructure
20.
Poult Sci ; 67(9): 1250-63, 1988 Sep.
Article in English | MEDLINE | ID: mdl-2847130

ABSTRACT

Experiments were designed to evaluate the effects of Gray strain infectious bronchitis virus (IBV) and high dietary calcium (Ca), alone and in combination, on renal function in pullets. Eight hundred female Single Comb White Leghorn chicks were raised on starter ration. Five hundred chicks were inoculated intravenously with Gray strain IBV at 4 wk of age; the remaining chicks were not exposed to IBV. At 6 wk of age, IBV-inoculated and uninoculated chicks were randomly divided into two diet treatment groups. Half the chicks were fed commercial grower ration (approximately 1.0% Ca, .6% available P) and half were fed commercial layer ration (approximately 3.25% Ca, .5% available P). Birds remained on their respective diets until 18 wk of age. Kidney function studies were conducted on anesthetized birds at 6 wk of age prior to initiation of the diet treatments, at 10 wk of age, and at 18 wk of age. The layer ration increased Ca excretion, decreased inorganic phosphate excretion, and decreased urine hydrogen ion concentration in 10-wk-old pullets in comparison with the grower ration. These diet effects on kidney function were attenuated when the pullets reached 18 wk of age. The layer ration also caused an 11.5% incidence of urolithiasis, and significantly increased kidney asymmetry in 18-wk-old pullets relative to the effects of the grower ration. Gray strain IBV exposure significantly increased kidney asymmetry in 18-wk-old pullets, but had no gross effect on kidney function clearly related to the etiology of urolithiasis. Gray strain IBV did not enhance the incidence of urolithiasis in any of the age groups.


Subject(s)
Calcium, Dietary/pharmacology , Chickens/microbiology , Coronaviridae Infections/veterinary , Kidney/physiopathology , Poultry Diseases/physiopathology , Age Factors , Animals , Calcium, Dietary/administration & dosage , Coronaviridae Infections/metabolism , Coronaviridae Infections/physiopathology , Female , Infectious bronchitis virus , Kidney/drug effects , Poultry Diseases/metabolism , Poultry Diseases/microbiology
SELECTION OF CITATIONS
SEARCH DETAIL
...