Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 143
Filter
1.
Nat Commun ; 13(1): 1178, 2022 03 04.
Article in English | MEDLINE | ID: mdl-35246509

ABSTRACT

Recently emerged variants of SARS-CoV-2 contain in their surface spike glycoproteins multiple substitutions associated with increased transmission and resistance to neutralising antibodies. We have examined the structure and receptor binding properties of spike proteins from the B.1.1.7 (Alpha) and B.1.351 (Beta) variants to better understand the evolution of the virus in humans. Spikes of both variants have the same mutation, N501Y, in the receptor-binding domains. This substitution confers tighter ACE2 binding, dependent on the common earlier substitution, D614G. Each variant spike has acquired other key changes in structure that likely impact virus pathogenesis. The spike from the Alpha variant is more stable against disruption upon binding ACE2 receptor than all other spikes studied. This feature is linked to the acquisition of a more basic substitution at the S1-S2 furin site (also observed for the variants of concern Delta, Kappa, and Omicron) which allows for near-complete cleavage. In the Beta variant spike, the presence of a new substitution, K417N (also observed in the Omicron variant), in combination with the D614G, stabilises a more open spike trimer, a conformation required for receptor binding. Our observations suggest ways these viruses have evolved to achieve greater transmissibility in humans.


Subject(s)
Angiotensin-Converting Enzyme 2/metabolism , COVID-19/metabolism , Mutation, Missense , SARS-CoV-2/genetics , Spike Glycoprotein, Coronavirus/genetics , Angiotensin-Converting Enzyme 2/chemistry , Angiotensin-Converting Enzyme 2/ultrastructure , Binding Sites/genetics , COVID-19/transmission , COVID-19/virology , Cryoelectron Microscopy , Cytopathogenic Effect, Viral/genetics , Evolution, Molecular , Host-Pathogen Interactions , Humans , Kinetics , Models, Molecular , Protein Binding , Protein Domains , SARS-CoV-2/metabolism , SARS-CoV-2/physiology , Spike Glycoprotein, Coronavirus/chemistry , Spike Glycoprotein, Coronavirus/metabolism
2.
Virology ; 567: 34-46, 2022 02.
Article in English | MEDLINE | ID: mdl-34953294

ABSTRACT

The bovine viral diarrhea virus 1 (BVDV-1), belonging to the Pestivirus genus, is characterized by the presence of two biotypes, cytopathogenic (cp) or non-cytopathogenic (ncp). For a better understanding of the host pathogen interactions, we set out to identify transcriptomic signatures of bovine lung primary cells (BPCs) infected with a cp or a ncp strain. For this, we used both a targeted approach by reverse transcription droplet digital PCR and whole genome approach using RNAseq. Data analysis showed 3571 differentially expressed transcripts over time (Fold Change >2) and revealed that the most deregulated pathways for cp strain are signaling pathways involved in responses to viral infection such as inflammatory response or apoptosis pathways. Interestingly, our data analysis revealed a deregulation of Wnt signaling pathway, a pathway described in embryogenesis, that was specifically seen with the BVDV-1 cp but not the ncp suggesting a role of this pathway in viral replication.


Subject(s)
Apoptosis Regulatory Proteins/genetics , Bovine Virus Diarrhea-Mucosal Disease/genetics , Cytopathogenic Effect, Viral/genetics , Diarrhea Virus 1, Bovine Viral/genetics , Transcriptome , Wnt Signaling Pathway/genetics , Animals , Apoptosis/genetics , Apoptosis Regulatory Proteins/metabolism , Bovine Virus Diarrhea-Mucosal Disease/metabolism , Bovine Virus Diarrhea-Mucosal Disease/pathology , Bovine Virus Diarrhea-Mucosal Disease/virology , Cattle , Diarrhea Virus 1, Bovine Viral/metabolism , Diarrhea Virus 1, Bovine Viral/pathogenicity , Epithelial Cells/metabolism , Epithelial Cells/virology , Gene Expression Profiling , Gene Expression Regulation , Host-Pathogen Interactions/genetics , Interleukins/genetics , Interleukins/metabolism , Lung/metabolism , Lung/virology , Membrane Potential, Mitochondrial , Mitochondria/genetics , Mitochondria/metabolism , Mitochondria/virology , NF-kappa B/genetics , NF-kappa B/metabolism , Primary Cell Culture , Respiratory Mucosa/metabolism , Respiratory Mucosa/virology , Toll-Like Receptors/genetics , Toll-Like Receptors/metabolism , Virus Replication
3.
Arch Virol ; 166(3): 921-927, 2021 Mar.
Article in English | MEDLINE | ID: mdl-33486628

ABSTRACT

Replication of Newcastle disease virus (NDV) is regulated by various host mechanisms, but the role of the extracellular signal-regulated kinase (ERK) pathway in regulating NDV replication is an open question. In this study, the relationship between the ERK pathway and NDV replication was investigated. NDV activated the ERK signaling in chicken embryo fibroblasts at the late stage of infection, correlating to expression of viral proteins. Specific blockage of the ERK pathway activation significantly decreased the transcription and translation levels of viral genes as well as virus replication and the cytopathogenic effect caused by NDV. Our results demonstrate that activation of the ERK pathway is required for NDV replication.


Subject(s)
Extracellular Signal-Regulated MAP Kinases/metabolism , MAP Kinase Signaling System/physiology , Newcastle Disease/pathology , Newcastle disease virus/genetics , Virus Replication/physiology , Animals , Cell Line , Chick Embryo , Chickens/virology , Cytopathogenic Effect, Viral/genetics , Enzyme Activation , Newcastle Disease/virology , Newcastle disease virus/growth & development
4.
J Cutan Pathol ; 48(7): 908-910, 2021 Jul.
Article in English | MEDLINE | ID: mdl-33345376

ABSTRACT

Rett syndrome (RTT) is a progressive neurological disorder, affecting females with mutations in the X-linked gene methyl-CpG-binding protein 2 (MECP2). While MECP2 has been implicated in cancers of the breast, colon, and prostrate, cancer in patients with RTT is rare. We present a case of malignant melanoma in a patient with RTT, which additionally, displayed hitherto undescribed nuclear features, resembling herpes simplex virus cytopathic effects.


Subject(s)
Cytopathogenic Effect, Viral/genetics , Melanoma/pathology , Rett Syndrome/pathology , Simplexvirus/metabolism , Skin Neoplasms/pathology , Adult , Disease Progression , Female , Humans , Melanoma/diagnosis , Melanoma/metabolism , Melanoma/surgery , Methyl-CpG-Binding Protein 2/genetics , Mutation , Rett Syndrome/complications , Rett Syndrome/genetics , Skin Neoplasms/diagnosis , Skin Neoplasms/metabolism , Skin Neoplasms/surgery , Melanoma, Cutaneous Malignant
5.
Viruses ; 12(10)2020 09 24.
Article in English | MEDLINE | ID: mdl-32987930

ABSTRACT

Salmonid alphavirus (SAV) is the cause of pancreas disease and sleeping disease in farmed salmonid fish in Europe. The spread of these diseases has been difficult to control with biosecurity and current vaccination strategies, and increased understanding of the viral pathogenesis could be beneficial for the development of novel vaccine strategies. N-glycosylation of viral envelope proteins may be crucial for viral virulence and a possible target for its purposed attenuation. In this study, we mutated the N-glycosylation consensus motifs of the E1 and E2 glycoproteins of a SAV3 infectious clone using site-directed mutagenesis. Mutation of the glycosylation motif in E1 gave a complete inactivation of the virus as no viral replication could be detected in cell culture and infectious particles could not be rescued. In contrast, infectious virus particles could be recovered from the SAV3 E2 mutants (E2319Q, E2319A), but not if they were accompanied by lack of N-glycosylation in E1. Compared to the non-mutated infectious clone, the SAV3-E2319Q and SAV3-E2319A recombinant viruses produced less cytopathic effects in cell culture and lower amounts of infectious viral particles. In conclusion, the substitution in the N-linked glycosylation site in E2 attenuated SAV3 in cell culture. The findings could be useful for immunization strategies using live attenuated vaccines and testing in fish will be desirable to study the clone's properties in vivo.


Subject(s)
Alphavirus/genetics , Alphavirus/pathogenicity , Salmon/virology , Trout/virology , Viral Envelope Proteins/genetics , Animals , Cell Line , Cytopathogenic Effect, Viral/genetics , Fish Diseases/virology , Glycosylation , Mutation/genetics , Vaccines, Attenuated , Viral Envelope Proteins/metabolism , Virulence/genetics
6.
J Transl Med ; 18(1): 362, 2020 09 23.
Article in English | MEDLINE | ID: mdl-32967693

ABSTRACT

BACKGROUND: Since the first outbreak of SARS-CoV-2, the clinical characteristics of the Coronavirus Disease 2019 (COVID-19) have been progressively changed. Data reporting a viral intra-host and inter-host evolution favouring the appearance of mild SARS-CoV-2 strains are since being accumulating. To better understand the evolution of SARS-CoV-2 pathogenicity and its adaptation to the host, it is therefore crucial to investigate the genetic and phenotypic characteristics of SARS-CoV-2 strains circulating lately in the epidemic. METHODS: Nasopharyngeal swabs have been analyzed for viral load in the early (March 2020) and late (May 2020) phases of epidemic in Brescia, Italy. Isolation of SARS-CoV-2 from 2 high viral load specimens identified on March 9 (AP66) and on May 8 (GZ69) was performed on Vero E6 cells. Amount of virus released was assessed by quantitative PCR. Genotypic characterization of AP66 and GZ69 was performed by next generation sequencing followed by an in-depth in silico analysis of nucleotide mutations. RESULTS: The SARS-CoV-2 GZ69 strain, isolated in May from an asymptomatic healthcare worker, showed an unprecedented capability of replication in Vero E6 cells in the absence of any evident cytopathic effect. Vero E6 subculturing, up to passage 4, showed that SARS-CoV-2 GZ69 infection was as productive as the one sustained by the cytopathic strain AP66. Whole genome sequencing of the persistently replicating SARS-CoV-2 GZ69 has shown that this strain differs from the early AP66 variant in 9 nucleotide positions (C2939T; C3828T; G21784T; T21846C; T24631C; G28881A; G28882A; G28883C; G29810T) which lead to 6 non-synonymous substitutions spanning on ORF1ab (P892S; S1188L), S (K74N; I95T) and N (R203K, G204R) proteins. CONCLUSIONS: Identification of the peculiar SARS-CoV-2 GZ69 strain in the late Italian epidemic highlights the need to better characterize viral variants circulating among asymptomatic or paucisymptomatic individuals. The current approach could unravel the ways for future studies aimed at analyzing the selection process which favours viral mutations in the human host.


Subject(s)
Betacoronavirus/genetics , Coronavirus Infections/virology , Genetic Variation , Pneumonia, Viral/virology , Amino Acid Substitution , Animals , Betacoronavirus/isolation & purification , Betacoronavirus/physiology , COVID-19 , Chlorocebus aethiops , Coronavirus Infections/epidemiology , Cytopathogenic Effect, Viral/genetics , Cytopathogenic Effect, Viral/physiology , Genome, Viral , Humans , Italy/epidemiology , Mutation , Pandemics , Phylogeny , Pneumonia, Viral/epidemiology , Polymorphism, Single Nucleotide , SARS-CoV-2 , Translational Research, Biomedical , Vero Cells , Viral Proteins/genetics , Viral Proteins/physiology , Virus Cultivation/methods , Virus Replication/genetics , Virus Replication/physiology , Whole Genome Sequencing
7.
Front Immunol ; 11: 367, 2020.
Article in English | MEDLINE | ID: mdl-32194570

ABSTRACT

Epstein-Barr virus (EBV) is an oncogenic human herpes virus that was discovered in 1964. Viral non-coding RNAs, such as BamHI-A rightward fragment-derived microRNAs (BART miRNAs) or BamHI-H rightward fragment 1-derived miRNAs (BHRF1 miRNA) in EBV-infected cells have been recently reported. Host miRNAs are also upregulated upon EBV infection. Viral and host miRNAs are important in maintaining viral infection and evasion of host immunity. Although miRNAs in EBV-infected cells often promote cell proliferation by targeting apoptosis or cell cycle, this review focuses on the regulation of the recognition of the host immune system. This review firstly describes the location and organization of two clusters of viral miRNAs, then describes evasion from host immune surveillance systems by modulating viral gene expression or inhibiting innate and acquired immunity by viral miRNAs as well as host miRNAs. Another topic is the enigmatic depletion of viral miRNAs in several types of EBV-infected tumor cells. Finally, this review introduces the strong correlation of nasopharyngeal cancer cases with a newly identified single nucleotide polymorphism that enhances BART miRNA promoter activity.


Subject(s)
Epstein-Barr Virus Infections/immunology , Gene Expression Regulation, Viral , Herpesvirus 4, Human/genetics , Host-Pathogen Interactions/immunology , MicroRNAs/immunology , RNA, Viral/immunology , Adaptive Immunity/genetics , Alternative Splicing , Carcinoma/genetics , Carcinoma/immunology , Carcinoma/virology , Cytopathogenic Effect, Viral/genetics , Epithelial Cells/virology , Epstein-Barr Virus Infections/genetics , Herpesvirus 4, Human/immunology , Herpesvirus 4, Human/physiology , Host-Pathogen Interactions/genetics , Humans , Immune Evasion/genetics , Immunity, Innate/genetics , Immunologic Surveillance , Lymphoma/genetics , Lymphoma/immunology , Lymphoma/virology , MicroRNAs/genetics , Nasopharyngeal Neoplasms/genetics , Nasopharyngeal Neoplasms/immunology , Nasopharyngeal Neoplasms/virology , Polymorphism, Single Nucleotide , Promoter Regions, Genetic , RNA, Viral/genetics , Viral Matrix Proteins/physiology , Viral Proteins/biosynthesis , Viral Proteins/genetics , Viral Proteins/immunology , Virus Latency/genetics , Virus Latency/immunology
9.
Cells ; 9(2)2020 02 18.
Article in English | MEDLINE | ID: mdl-32085644

ABSTRACT

Enterovirus 71 (EV71) infection is an endemic disease in Southeast Asia and China. We have previously shown that EV71 virus causes functional changes in mitochondria. It is speculative whether EV71 virus alters the host cell metabolism to its own benefit. Using a metabolomics approach, we demonstrate that EV71-infected Vero cells had significant changes in metabolism. Glutathione and its related metabolites, and several amino acids, such as glutamate and aspartate, changed significantly with the infectious dose of virus. Other pathways, including glycolysis and tricarboxylic acid cycle, were also altered. A change in glutamine/glutamate metabolism is critical to the viral infection. The presence of glutamine in culture medium was associated with an increase in viral replication. Dimethyl α-ketoglutarate treatment partially mimicked the effect of glutamine supplementation. In addition, the immunoblot analysis revealed that the expression of glutamate dehydrogenase (GDH) and trifunctional carbamoyl-phosphate synthetase 2, aspartate transcarbamylase, and dihydroorotase (CAD) increased during infection. Knockdown of expression of glutaminase (GLS), GDH and CAD drastically reduced the cytopathic effect (CPE) and viral replication. Furthermore, we found that CAD bound VP1 to promote the de novo pyrimidine synthesis. Our findings suggest that virus may induce metabolic reprogramming of host cells to promote its replication through interactions between viral and host cell proteins.


Subject(s)
Dihydroorotase/metabolism , Enterovirus A, Human/physiology , Enterovirus Infections/metabolism , Glutamate Dehydrogenase/metabolism , Glutaminase/metabolism , Host-Pathogen Interactions/genetics , Virus Replication/drug effects , Virus Replication/genetics , Animals , Chlorocebus aethiops , Cytopathogenic Effect, Viral/drug effects , Cytopathogenic Effect, Viral/genetics , Dihydroorotase/genetics , Enterovirus Infections/virology , Gene Knockdown Techniques , Glutamate Dehydrogenase/genetics , Glutamic Acid/metabolism , Glutaminase/genetics , Glutamine/metabolism , Glutamine/pharmacology , Glycolysis/genetics , Ketoglutaric Acids/pharmacology , RNA Interference , Transfection , Vero Cells
10.
PLoS One ; 14(4): e0215822, 2019.
Article in English | MEDLINE | ID: mdl-31013314

ABSTRACT

Human metapneumovirus (HMPV) has been a notable etiological agent of acute respiratory infection in humans, but it was not discovered until 2001, because HMPV replicates only in a limited number of cell lines and the cytopathic effect (CPE) is often mild. To promote the study of HMPV, several groups have generated green fluorescent protein (GFP)-expressing recombinant HMPV strains (HMPVGFP). However, the growing evidence has complicated the understanding of cell line specificity of HMPV, because it seems to vary notably among HMPV strains. In addition, unique A2b clade HMPV strains with a 180-nucleotide duplication in the G gene (HMPV A2b180nt-dup strains) have recently been detected. In this study, we re-evaluated and compared the cell line specificity of clinical isolates of HMPV strains, including the novel HMPV A2b180nt-dup strains, and six recombinant HMPVGFP strains, including the newly generated recombinant HMPV A2b180nt-dup strain, MG0256-EGFP. Our data demonstrate that VeroE6 and LLC-MK2 cells generally showed the highest infectivity with any clinical isolates and recombinant HMPVGFP strains. Other human-derived cell lines (BEAS-2B, A549, HEK293, MNT-1, and HeLa cells) showed certain levels of infectivity with HMPV, but these were significantly lower than those of VeroE6 and LLC-MK2 cells. Also, the infectivity in these suboptimal cell lines varied greatly among HMPV strains. The variations were not directly related to HMPV genotypes, cell lines used for isolation and propagation, specific genome mutations, or nucleotide duplications in the G gene. Thus, these variations in suboptimal cell lines are likely intrinsic to particular HMPV strains.


Subject(s)
Cell Line/virology , Cytopathogenic Effect, Viral/genetics , Metapneumovirus/growth & development , Respiratory Tract Infections/virology , A549 Cells , Green Fluorescent Proteins/genetics , HEK293 Cells , HeLa Cells , Humans , Metapneumovirus/genetics , Metapneumovirus/pathogenicity , Respiratory Tract Infections/genetics , Respiratory Tract Infections/prevention & control
11.
J Virol ; 93(12)2019 06 15.
Article in English | MEDLINE | ID: mdl-30944176

ABSTRACT

Zika virus (ZIKV) is an emerging mosquito-borne flavivirus. Recent ZIKV outbreaks have produced serious human disease, including neurodevelopmental malformations (congenital Zika syndrome) and Guillain-Barré syndrome. These outcomes were not associated with ZIKV infection prior to 2013, raising the possibility that viral genetic changes could contribute to new clinical manifestations. All contemporary ZIKV isolates encode an N-linked glycosylation site in the envelope (E) protein (N154), but this glycosylation site is absent in many historical ZIKV isolates. Here, we investigated the role of E protein glycosylation in ZIKV pathogenesis using two contemporary Asian-lineage strains (H/PF/2013 and PRVABC59) and the historical African-lineage strain (MR766). We found that glycosylated viruses were highly pathogenic in Ifnar1-/- mice. In contrast, nonglycosylated viruses were attenuated, producing lower viral loads in the serum and brain when inoculated subcutaneously but remaining neurovirulent when inoculated intracranially. These results suggest that E glycosylation is advantageous in the periphery but not within the brain. Accordingly, we found that glycosylation facilitated infection of cells expressing the lectins dendritic cell-specific intercellular adhesion molecule-3-grabbing nonintegrin (DC-SIGN) or DC-SIGN-related (DC-SIGNR), suggesting that inefficient infection of lectin-expressing leukocytes could contribute to the attenuation of nonglycosylated ZIKV in mice.IMPORTANCE It is unclear why the ability of Zika virus (ZIKV) to cause serious disease, including Guillain-Barré syndrome and birth defects, was not recognized until recent outbreaks. One contributing factor could be genetic differences between contemporary ZIKV strains and historical ZIKV strains. All isolates from recent outbreaks encode a viral envelope protein that is glycosylated, whereas many historical ZIKV strains lack this glycosylation. We generated nonglycosylated ZIKV mutants from contemporary and historical strains and evaluated their virulence in mice. We found that nonglycosylated viruses were attenuated and produced lower viral loads in serum and brains. Our studies suggest that envelope protein glycosylation contributes to ZIKV pathogenesis, possibly by facilitating attachment to and infection of lectin-expressing leukocytes.


Subject(s)
Cytopathogenic Effect, Viral/genetics , Viral Envelope Proteins/genetics , Zika Virus/metabolism , A549 Cells , Animals , Cell Adhesion Molecules/metabolism , Chlorocebus aethiops , Female , Flavivirus/metabolism , Glycosylation , Humans , Lectins, C-Type/metabolism , Male , Mice , Mice, Inbred C57BL , Receptors, Cell Surface/metabolism , Vero Cells , Viral Envelope Proteins/metabolism , Virulence , Virus Replication , Zika Virus/genetics , Zika Virus/pathogenicity , Zika Virus Infection/virology
12.
PLoS One ; 14(3): e0214016, 2019.
Article in English | MEDLINE | ID: mdl-30893357

ABSTRACT

Zika virus (ZIKV) has emerged globally as an important pathogen, since it has been recognized as a cause of microcephaly and other neurologic processes and sequalae in newborns. The virus shares homology with Hepaciviruses and therefore may be a cause of hepatitis. We sought to characterize ZIKV replication in hepatocyte-derived cell lines. Huh7.5 and HepG2 cells were infected with ZIKV and replication potential was evaluated by multiple methods including plaque assay, qRT-PCR, negative-strand ZIKV RNA production, and ZIKV NS1 protein production. Growth curves in cells and supernatant were compared to replicative capacity in Vero cells. Overall, viral replication in both hepatocyte lines approximated that observed in the Vero cells. Cell cytopathology was observed after 3 days of infection and apoptosis markers increased. Transmission electron microscopy revealed evidence of viral capsids in cells and negative staining revealed ZIKV particles in the supernatant. Conclusions: Hepatocyte-derived cell lines are permissive for ZIKV replication and produce an overt cytopathic effect consistent with development of an acute viral hepatitis. Further evaluation of replication and injury is warranted.


Subject(s)
Liver/virology , Zika Virus Infection/virology , Zika Virus/genetics , Animals , Cell Line , Cell Line, Tumor , Chlorocebus aethiops , Cytopathogenic Effect, Viral/genetics , Hep G2 Cells , Hepatocytes/virology , Humans , Vero Cells , Viral Load/genetics , Viral Nonstructural Proteins/genetics , Virion/genetics , Virus Replication/genetics
13.
J Virol ; 93(4)2019 02 15.
Article in English | MEDLINE | ID: mdl-30487275

ABSTRACT

Alphavirus infections are characterized by global inhibition of cellular transcription and rapid induction of a cytopathic effect (CPE) in cells of vertebrate origin. Transcriptional shutoff impedes the cellular response to alphavirus replication and prevents establishment of an antiviral state. Chikungunya virus (CHIKV) is a highly pathogenic alphavirus representative, and its nonstructural protein 2 (nsP2) plays critical roles in both inhibition of transcription and CPE development. Previously, we have identified a small peptide in Sindbis virus (SINV) nsP2 (VLoop) that determined the protein's transcriptional inhibition function. It is located in the surface-exposed loop of the carboxy-terminal domain of nsP2 and exhibits high variability between members of different alphavirus serocomplexes. In this study, we found that SINV-specific mutations could not be directly applied to CHIKV. However, by using a new selection approach, we identified a variety of new VLoop variants that made CHIKV and its replicons incapable of inhibiting cellular transcription and dramatically less cytopathic. Importantly, the mutations had no negative effect on RNA and viral replication rates. In contrast to parental CHIKV, the developed VLoop mutants were unable to block induction of type I interferon. Consequently, they were cleared from interferon (IFN)-competent cells without CPE development. Alternatively, in murine cells that have defects in type I IFN production or signaling, the VLoop mutants established persistent, noncytopathic replication. The mutations in nsP2 VLoop may be used for development of new vaccine candidates against alphavirus infections and vectors for expression of heterologous proteins.IMPORTANCE Chikungunya virus is an important human pathogen which now circulates in both the Old and New Worlds. As in the case of other Old World alphaviruses, CHIKV nsP2 not only has enzymatic functions in viral RNA replication but also is a critical inhibitor of the antiviral response and one of the determinants of CHIKV pathogenesis. In this study, we have applied a new strategy to select a variety of CHIKV nsP2 mutants that no longer exhibited transcription-inhibitory functions. The designed CHIKV variants became potent type I interferon inducers and acquired a less cytopathic phenotype. Importantly, they demonstrated the same replication rates as the parental CHIKV. Mutations in the same identified peptide of nsP2 proteins derived from other Old World alphaviruses also abolished their nuclear functions. Such mutations can be further exploited for development of new attenuated alphaviruses.


Subject(s)
Chikungunya virus/metabolism , Viral Nonstructural Proteins/genetics , Animals , Antiviral Agents , Cell Line , Chikungunya Fever/genetics , Chikungunya Fever/metabolism , Chikungunya virus/genetics , Chikungunya virus/physiology , Cytopathogenic Effect, Viral/genetics , DNA Viruses/genetics , Humans , Interferon Type I/genetics , Mice , Mutation , NIH 3T3 Cells , RNA, Viral/metabolism , Replicon , Signal Transduction , Sindbis Virus/genetics , Sindbis Virus/physiology , Viral Nonstructural Proteins/metabolism , Virus Replication/genetics
14.
Nat Microbiol ; 4(2): 216-225, 2019 02.
Article in English | MEDLINE | ID: mdl-30420785

ABSTRACT

Cell motility is essential for viral dissemination1. Vaccinia virus (VACV), a close relative of smallpox virus, is thought to exploit cell motility as a means to enhance the spread of infection1. A single viral protein, F11L, contributes to this by blocking RhoA signalling to facilitate cell retraction2. However, F11L alone is not sufficient for VACV-induced cell motility, indicating that additional viral factors must be involved. Here, we show that the VACV epidermal growth factor homologue, VGF, promotes infected cell motility and the spread of viral infection. We found that VGF secreted from early infected cells is cleaved by ADAM10, after which it acts largely in a paracrine manner to direct cell motility at the leading edge of infection. Real-time tracking of cells infected in the presence of EGFR, MAPK, FAK and ADAM10 inhibitors or with VGF-deleted and F11-deleted viruses revealed defects in radial velocity and directional migration efficiency, leading to impaired cell-to-cell spread of infection. Furthermore, intravital imaging showed that virus spread and lesion formation are attenuated in the absence of VGF. Our results demonstrate how poxviruses hijack epidermal growth factor receptor-induced cell motility to promote rapid and efficient spread of infection in vitro and in vivo.


Subject(s)
Cell Movement , Host-Pathogen Interactions , Peptides/metabolism , Signal Transduction , Vaccinia virus/physiology , Vaccinia/virology , ADAM10 Protein/antagonists & inhibitors , ADAM10 Protein/metabolism , Amyloid Precursor Protein Secretases/antagonists & inhibitors , Amyloid Precursor Protein Secretases/metabolism , Animals , Cell Line , Cell Movement/drug effects , Cytopathogenic Effect, Viral/genetics , Enzyme Inhibitors/pharmacology , ErbB Receptors/antagonists & inhibitors , ErbB Receptors/metabolism , Gene Deletion , HeLa Cells , Humans , Intercellular Signaling Peptides and Proteins/metabolism , Membrane Proteins/antagonists & inhibitors , Membrane Proteins/metabolism , Mice , Peptides/deficiency , Peptides/genetics , Signal Transduction/drug effects , Vaccinia/metabolism , Vaccinia/pathology , Vaccinia virus/genetics , Vaccinia virus/growth & development , Vaccinia virus/metabolism , Viral Proteins/genetics , Viral Proteins/metabolism
15.
PLoS One ; 13(9): e0203331, 2018.
Article in English | MEDLINE | ID: mdl-30192813

ABSTRACT

Zika virus (ZIKV), an arthropod-borne virus, has emerged as a major human pathogen. Prolonged or persistent ZIKV infection of human cells and tissues may serve as a reservoir for the virus and present serious challenges to the safety of public health. Human hematopoietic cell lines with different developmental properties revealed differences in susceptibility and outcomes to ZIKV infection. In three separate studies involving the prototypic MR 766 ZIKV strain and the human monocytic leukemia U937 cell line, ZIKV initially developed only a low-grade infection at a slow rate. After continuous culture for several months, persistently ZIKV-infected cell lines were observed with most, if not all, cells testing positive for ZIKV antigen. The infected cultures produced ZIKV RNA (v-RNA) and infectious ZIKVs persistently ("persistent ZIKVs") with distinct infectivity and pathogenicity when tested using various kinds of host cells. When the genomes of ZIKVs from the three persistently infected cell lines were compared with the genome of the prototypic MR 766 ZIKV strain, distinct sets of mutations specific to each cell line were found. Significantly, all three "persistent ZIKVs" were capable of infecting fresh U937 cells with high efficiency at rapid rates, resulting in the development of a new set of persistently ZIKV-infected U937 cell lines. The genomes of ZIKVs from the new set of persistently ZIKV-infected U937 cell lines were further analyzed for their different mutations. The 2nd generation of persistent ZIKVs continued to possess most of the distinct sets of mutations specific to the respective 1st generation of persistent ZIKVs. We anticipate that the study will contribute to the understanding of the fundamental biology of adaptive mutations and selection during viral persistence. The persistently ZIKV-infected human cell lines that we developed will also be useful to investigate critical molecular pathways of ZIKV persistence and to study drugs or countermeasures against ZIKV infections and transmission.


Subject(s)
Cytopathogenic Effect, Viral/genetics , Virulence/genetics , Zika Virus Infection/transmission , Zika Virus Infection/virology , Zika Virus/genetics , Zika Virus/pathogenicity , B-Lymphocytes/virology , Cell Line , Disease Reservoirs/virology , Genome, Viral , Genomics , Host Microbial Interactions/genetics , Humans , Monocytes/virology , Mutation , T-Lymphocytes/virology , U937 Cells
16.
Int J Mol Sci ; 19(3)2018 Mar 02.
Article in English | MEDLINE | ID: mdl-29498634

ABSTRACT

Influenza virus remains a major health concern worldwide, and there have been continuous efforts to develop effective antivirals despite the use of annual vaccination programs. The purpose of this study was to determine the anti-influenza activity of Bax inhibitor-1 (BI-1). Madin-Darby Canine Kidney (MDCK) cells expressing wild type BI-1 and a non-functional BI-1 mutant, BI-1 ∆C (with the C-terminal 14 amino acids deleted) were prepared and infected with A/PR/8/34 influenza virus. BI-1 overexpression led to the suppression of virus-induced cell death and virus production compared to control Mock or BI-1 ∆C overexpression. In contrast to BI-1 ∆C-overexpressing cells, BI-1-overexpressing cells exhibited markedly reduced virus-induced expression of several viral genes, accompanied by a substantial decrease in ROS production. We found that treatment with a ROS scavenging agent, N-acetyl cysteine (NAC), led to a dramatic decrease in virus production and viral gene expression in control MDCK and BI-1 ∆C-overexpressing cells. In contrast, NAC treatment resulted in the slight additional suppression of virus production and viral gene expression in BI-1-overexpressing cells but was statistically significant. Moreover, the expression of heme oxygenase-1 (HO-1) was also significantly increased following virus infection in BI-1-overexpressing cells compared to control cells. Taken together, our data suggest that BI-1 may act as an anti-influenza protein through the suppression of ROS mediated cell death and upregulation of HO-1 expression in influenza virus infected MDCK cells.


Subject(s)
Apoptosis Regulatory Proteins/metabolism , Heme Oxygenase-1/genetics , Host-Pathogen Interactions , Influenza A virus/physiology , Membrane Proteins/metabolism , Reactive Oxygen Species/metabolism , Animals , Apoptosis Regulatory Proteins/genetics , Cell Death/genetics , Cell Line , Cells, Cultured , Cytopathogenic Effect, Viral/genetics , Dogs , Gene Expression Regulation , Gene Expression Regulation, Viral , Gene Order , Genetic Vectors/genetics , Heme Oxygenase-1/metabolism , Humans , Influenza, Human/genetics , Influenza, Human/metabolism , Influenza, Human/virology , Madin Darby Canine Kidney Cells , Membrane Proteins/genetics , Models, Biological , Virus Replication
17.
Biochem Biophys Res Commun ; 495(2): 1871-1877, 2018 01 08.
Article in English | MEDLINE | ID: mdl-29223400

ABSTRACT

We tested usefulness of a target enrichment system SureSelect, a comprehensive viral nucleic acid detection method, for rapid identification of viral pathogens in feces samples of cattle, pigs and goats. This system enriches nucleic acids of target viruses in clinical/field samples by using a library of biotinylated RNAs with sequences complementary to the target viruses. The enriched nucleic acids are amplified by PCR and subjected to next generation sequencing to identify the target viruses. In many samples, SureSelect target enrichment method increased efficiencies for detection of the viruses listed in the biotinylated RNA library. Furthermore, this method enabled us to determine nearly full-length genome sequence of porcine parainfluenza virus 1 and greatly increased Breadth, a value indicating the ratio of the mapping consensus length in the reference genome, in pig samples. Our data showed usefulness of SureSelect target enrichment system for comprehensive analysis of genomic information of various viruses in field samples.


Subject(s)
Chromosome Mapping/veterinary , Cytopathogenic Effect, Viral/genetics , Genome, Viral/genetics , High-Throughput Nucleotide Sequencing/veterinary , Livestock/virology , Viruses/genetics , Viruses/isolation & purification , Animals , Cattle , Chromosome Mapping/methods , High-Throughput Nucleotide Sequencing/methods , Swine , Viruses/pathogenicity
18.
Oncotarget ; 8(1): 1213-1225, 2017 Jan 03.
Article in English | MEDLINE | ID: mdl-27901484

ABSTRACT

Pexa-Vec (pexastimogene devacirpvec; JX-594) has emerged as an attractive tool in oncolytic virotherapy. Pexa-Vec demonstrates oncolytic and immunotherapeutic mechanisms of action. But the determinants of resistance to Pexa-Vec are mostly unknown. We treated hemoatologic malignant cells with Pexa-Vec and examined the gene-expression pattern of sensitive and resistant cells. Human myeloid malignant cell lines (RPMI-8226, IM-9, K562, THP-1) and lymphoid cancer cell lines (MOLT4, CCRF-CEM, Ramos, U937) were treated with Pexa-Vec. Pexa-Vec was cytotoxic on myeloid cell lines in a dose-dependent manner, and fluorescent imaging and qPCR revealed that Pexa-Vec expression was low in RAMOS than IM-9 after 24 hrs and 48 hrs of infection. Gene expression profiles between two groups were analyzed by microarray. Genes with at least 2-fold increase or decrease in their expression were identified. A total of 660 genes were up-regulated and 776 genes were down-regulated in lymphoid cancer cell lines. The up- and down-regulated genes were categorized into 319 functional gene clusters. We identified the top 10 up-regulated genes in lymphoid cells. Among them three human genes (LEF1, STAMBPL1, and SLFN11) strongly correlated with viral replication. Up-regulation of PVRIG, LPP, CECR1, Arhgef6, IRX3, IGFBP2, CD1d were related to resistant to Pexa-Vec. In conclusion, lymphoid malignant cells are resistant to Pexa-Vec and displayed up-regulated genes associated with resistance to oncolytic viral therapy. These data provide potential targets to overcome resistance, and suggest that molecular assays may be useful in selecting patients for further clinical trials with Pexa-Vec.


Subject(s)
Gene Expression Profiling , Gene Expression Regulation, Neoplastic , Hematologic Neoplasms/genetics , Biomarkers, Tumor , Cell Line, Tumor , Cytopathogenic Effect, Viral/genetics , Genetic Vectors/genetics , Hematologic Neoplasms/therapy , Humans , Oncolytic Virotherapy , Oncolytic Viruses/genetics , Transcriptome , Vaccinia virus/genetics
19.
Virology ; 487: 50-8, 2016 Jan.
Article in English | MEDLINE | ID: mdl-26517396

ABSTRACT

Small ruminant lentiviruses infect goats and sheep, inducing clinical disease in a minority of infected animals. Following an eradication campaign, clinical cases may disappear in a population. The complete elimination of these lentiviruses is however difficult to achieve and the spreading of less virulent strains often parallels the elimination of their virulent counterparts. Here, we characterized three such strains isolated from a flock in the post-eradication phase. We completely sequenced their genomes, showing that one of the isolates was most probably the product of a recombination event between the other two viruses. By comparing the sequences of these isolates with those of virulent strains, we found evidence that particular LTR mutations may explain their attenuated phenotype. Finally, we constructed an infectious molecular clone representative of these viruses, analyzing its replication characteristics in different target cells. This clone will permit us to explore the molecular correlates of cytopathogenicity and virulence.


Subject(s)
Arthritis-Encephalitis Virus, Caprine/genetics , Cloning, Molecular/methods , Lentivirus Infections/virology , RNA, Viral/genetics , Visna-maedi virus/genetics , Animals , Arthritis-Encephalitis Virus, Caprine/isolation & purification , Arthritis-Encephalitis Virus, Caprine/pathogenicity , Base Sequence , Cells, Cultured , Cytopathogenic Effect, Viral/genetics , Goat Diseases/virology , Goats , Macrophages/virology , Molecular Sequence Data , Sequence Alignment , Sequence Analysis, RNA , Sheep , Sheep Diseases/virology , Visna-maedi virus/isolation & purification , Visna-maedi virus/pathogenicity
20.
Biomed Res Int ; 2014: 285607, 2014.
Article in English | MEDLINE | ID: mdl-25313356

ABSTRACT

Entamoeba histolytica is a parasite which presents capacity to degrade tissues and therefore has a pathogenic behavior. As this behavior is not shown by all strains, there have been several studies investigating molecular basis of the cytotoxicity process. Using the suppression subtractive hybridization (SSH) technique, differential gene expressions of two E. histolytica strains, one virulent (EGG) and one nonvirulent (452), have been analyzed with the purpose of isolating genes which may be involved with amoebic virulence. Nine cDNA fragments presenting high homology with E. histolytica previously sequenced genes were subtracted. Of these, four genes were confirmed by RT-PCR. Two coding for hypothetical proteins, one for a cysteine-rich protein, expressed only in the virulent strain, EGG and another one, coding for grainin 2 protein, exclusive from 452 strain. This study provided new insight into the proteins differences in the virulent and nonvirulent E. histolytica strains. We believe that further studies with these proteins may prove association of them with tissue damage, providing new perceptions to improve treatment or diagnosis of the invasive disease.


Subject(s)
Entamoeba histolytica/genetics , Entamoeba histolytica/pathogenicity , Gene Expression Profiling , Gene Expression Regulation , Subtractive Hybridization Techniques/methods , Animals , CHO Cells , Cricetinae , Cricetulus , Cytopathogenic Effect, Viral/genetics , Electrophoresis, Agar Gel , Electrophoresis, Polyacrylamide Gel , Humans , Liver/parasitology , Male , Trophozoites/physiology , Virulence/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...