Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 20 de 94
Filtrer
1.
PLoS Pathog ; 18(1): e1010159, 2022 01.
Article de Anglais | MEDLINE | ID: mdl-34995322

RÉSUMÉ

The clinical impact of rhinovirus C (RV-C) is well-documented; yet, the viral life cycle remains poorly defined. Thus, we characterized RV-C15 replication at the single-cell level and its impact on the human airway epithelium (HAE) using a physiologically-relevant in vitro model. RV-C15 replication was restricted to ciliated cells where viral RNA levels peaked at 12 hours post-infection (hpi), correlating with elevated titers in the apical compartment at 24hpi. Notably, infection was associated with a loss of polarized expression of the RV-C receptor, cadherin-related family member 3. Visualization of double-stranded RNA (dsRNA) during RV-C15 replication revealed two distinct replication complex arrangements within the cell, likely corresponding to different time points in infection. To further define RV-C15 replication sites, we analyzed the expression and colocalization of giantin, phosphatidylinositol-4-phosphate, and calnexin with dsRNA. Despite observing Golgi fragmentation by immunofluorescence during RV-C15 infection as previously reported for other RVs, a high ratio of calnexin-dsRNA colocalization implicated the endoplasmic reticulum as the primary site for RV-C15 replication in HAE. RV-C15 infection was also associated with elevated stimulator of interferon genes (STING) expression and the induction of incomplete autophagy, a mechanism used by other RVs to facilitate non-lytic release of progeny virions. Notably, genetic depletion of STING in HAE attenuated RV-C15 and -A16 (but not -B14) replication, corroborating a previously proposed proviral role for STING in some RV infections. Finally, RV-C15 infection resulted in a temporary loss in epithelial barrier integrity and the translocation of tight junction proteins while a reduction in mucociliary clearance indicated cytopathic effects on epithelial function. Together, our findings identify both shared and unique features of RV-C replication compared to related rhinoviruses and define the impact of RV-C on both epithelial cell organization and tissue functionality-aspects of infection that may contribute to pathogenesis in vivo.


Sujet(s)
Réticulum endoplasmique/virologie , Enterovirus/physiologie , Muqueuse respiratoire/virologie , Réplication virale/physiologie , Cellules cultivées , Effet cytopathogène viral/physiologie , Humains
2.
Sci Rep ; 10(1): 15234, 2020 09 17.
Article de Anglais | MEDLINE | ID: mdl-32943650

RÉSUMÉ

Enterovirus A71 (EV-A71), which belongs to the family Picornaviridae, can invade the central nervous system (CNS) and cause severe CNS complications or death. The EV-A71 antigen has been detected in the neurons in the brains of humans who died from EV-A71 infection. However, the effect of EV-A71 infection on human neuronal cells remains poorly understood. Human neural stem cells (NSCs) and IMR-32 neuroblastoma cells were differentiated into neuronal cells for this study. Although the neuronal cells were permissive to EV-A71 infection, EV-A71 infection did not induce an obvious cytopathic effect on the neuronal cells. EV-A71 infection did not induce apoptosis in neuronal cells. However, autophagy and autophagic flux were induced in EV-A71-infected neuronal cells. The production of autophagosomes was shown to be important for EV-A71 viral RNA (vRNA) replication in neuronal cells.


Sujet(s)
Entérovirus humain A/physiologie , Entérovirus humain A/pathogénicité , Neurones/virologie , Autophagosomes/virologie , Autophagie/physiologie , Caspases/métabolisme , Différenciation cellulaire , Cellules cultivées , Effet cytopathogène viral/physiologie , Entérovirus humain A/génétique , Activation enzymatique , Interactions hôte-microbes/physiologie , Humains , Cellules souches neurales/métabolisme , Cellules souches neurales/anatomopathologie , Cellules souches neurales/virologie , Neurones/métabolisme , Neurones/anatomopathologie , ARN viral/biosynthèse , ARN viral/génétique , Réplication virale/physiologie
3.
J Transl Med ; 18(1): 362, 2020 09 23.
Article de Anglais | MEDLINE | ID: mdl-32967693

RÉSUMÉ

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.


Sujet(s)
Betacoronavirus/génétique , Infections à coronavirus/virologie , Variation génétique , Pneumopathie virale/virologie , Substitution d'acide aminé , Animaux , Betacoronavirus/isolement et purification , Betacoronavirus/physiologie , COVID-19 , Chlorocebus aethiops , Infections à coronavirus/épidémiologie , Effet cytopathogène viral/génétique , Effet cytopathogène viral/physiologie , Génome viral , Humains , Italie/épidémiologie , Mutation , Pandémies , Phylogenèse , Pneumopathie virale/épidémiologie , Polymorphisme de nucléotide simple , SARS-CoV-2 , , Cellules Vero , Protéines virales/génétique , Protéines virales/physiologie , Culture virale/méthodes , Réplication virale/génétique , Réplication virale/physiologie , Séquençage du génome entier
4.
Transbound Emerg Dis ; 67(4): 1574-1584, 2020 Jul.
Article de Anglais | MEDLINE | ID: mdl-31975574

RÉSUMÉ

There are four major porcine reproductive and respiratory syndrome virus 2 (PRRSV2) lineages circulating in China based on classification system, including lineages 1 (NADC30-like), 3 (QYYZ-like), 5.1 (VR2332-like) and 8 (JXA1-like/CH-1a-Like), which leads to the potential recombination. In the present study, a novel variant of PRRSV2 strain named JS18-3 was isolated from piglets suffering severe breathing difficulties in Jiangsu Province of China in 2018. Full-length genome analysis indicated that JS18-3 shared 86.5%, 87.9%, 84.2%, 82.2% and 86.4% nucleotide similarity with PRRSVs CH-1a, JXA1, VR2332, QYYZ and NADC30, respectively. 4871-6635 of JS18-3 shared the highest identity of 99.3% in nucleotide sequence with HP-PRRSV representative strain JXA1 indicating ongoing evolution to HP-PRRSV. JS18-3 was classified into classical lineage 8 of PRRSV2 based on phylogenetic analysis of complete genome and ORF5. Genomic break points in structural (ORF3) and non-structural (NSP2, NSP3) regions of genomes were detected in recombination analysis. JS18-3 is a recombinant isolate from lineages 8, 1 and 3. Replication enhancement and severe cytopathic effects caused by JS18-3 were observed in Marc-145 cells and porcine alveolar macrophages (PAMs) as compared to JX07, a typical strain of lineage 8. Pathogenicity results indicated that piglets inoculated with JS18-3 presented persistent fever, dyspnoea, serious microscopic lung lesions and lymph node congestion. The study suggests that lineage 8 of PRRSV2 is involved in continuing evolution by genetic recombination and mutation leading to outbreaks in vaccinated pigs in China.


Sujet(s)
Génome viral/génétique , Syndrome dysgénésique et respiratoire porcin/virologie , Virus du syndrome respiratoire et reproducteur porcin/isolement et purification , Virus du syndrome respiratoire et reproducteur porcin/physiologie , Recombinaison génétique , Maladies des porcs/virologie , Réplication virale/physiologie , Séquence d'acides aminés , Animaux , Séquence nucléotidique , Chine/épidémiologie , Effet cytopathogène viral/physiologie , Variation génétique , Génomique , Macrophages alvéolaires/virologie , Phylogenèse , Syndrome dysgénésique et respiratoire porcin/épidémiologie , Suidae , Maladies des porcs/épidémiologie
5.
J Virol ; 93(22)2019 11 15.
Article de Anglais | MEDLINE | ID: mdl-31484749

RÉSUMÉ

Phase-separated biomolecular condensates of proteins and nucleic acids form functional membrane-less organelles (e.g., stress granules and P-bodies) in the mammalian cell cytoplasm and nucleus. In contrast to the long-standing belief that interferon (IFN)-inducible human myxovirus resistance protein A (MxA) associated with the endoplasmic reticulum (ER) and Golgi apparatus, we report that MxA formed membraneless metastable (shape-changing) condensates in the cytoplasm. In our studies, we used the same cell lines and methods as those used by previous investigators but concluded that wild-type MxA formed variably sized spherical or irregular bodies, filaments, and even a reticulum distinct from that of ER/Golgi membranes. Moreover, in Huh7 cells, MxA structures associated with a novel cytoplasmic reticular meshwork of intermediate filaments. In live-cell assays, 1,6-hexanediol treatment led to rapid disassembly of green fluorescent protein (GFP)-MxA structures; FRAP revealed a relative stiffness with a mobile fraction of 0.24 ± 0.02 within condensates, consistent with a higher-order MxA network structure. Remarkably, in intact cells, GFP-MxA condensates reversibly disassembled/reassembled within minutes of sequential decrease/increase, respectively, in tonicity of extracellular medium, even in low-salt buffers adjusted only with sucrose. Condensates formed from IFN-α-induced endogenous MxA also displayed tonicity-driven disassembly/reassembly. In vesicular stomatitis virus (VSV)-infected Huh7 cells, the nucleocapsid (N) protein, which participates in forming phase-separated viral structures, associated with spherical GFP-MxA condensates in cells showing an antiviral effect. These observations prompt comparisons with the extensive literature on interactions between viruses and stress granules/P-bodies. Overall, the new data correct a long-standing misinterpretation in the MxA literature and provide evidence for membraneless MxA biomolecular condensates in the uninfected cell cytoplasm.IMPORTANCE There is a long-standing belief that interferon (IFN)-inducible human myxovirus resistance protein A (MxA), which displays antiviral activity against several RNA and DNA viruses, associates with the endoplasmic reticulum (ER) and Golgi apparatus. We provide data to correct this misinterpretation and further report that MxA forms membraneless metastable (shape-changing) condensates in the cytoplasm consisting of variably sized spherical or irregular bodies, filaments, and even a reticulum. Remarkably, MxA condensates showed the unique property of rapid (within 1 to 3 min) reversible disassembly and reassembly in intact cells exposed sequentially to hypotonic and isotonic conditions. Moreover, GFP-MxA condensates included the VSV nucleocapsid (N) protein, a protein previously shown to form liquid-like condensates. Since intracellular edema and ionic changes are hallmarks of cytopathic effects of a viral infection, the tonicity-driven regulation of MxA condensates may reflect a mechanism for modulation of MxA function during viral infection.


Sujet(s)
Cytoplasme/virologie , Protéines de résistance aux myxovirus/métabolisme , Lignée cellulaire , Effet cytopathogène viral/physiologie , Cytoplasme/métabolisme , Humains , Orthomyxoviridae/métabolisme , Protéines/métabolisme , Virus de la stomatite vésiculeuse de type Indiana/métabolisme , Maladies virales/métabolisme , Virus/métabolisme
6.
J Virol ; 93(18)2019 09 15.
Article de Anglais | MEDLINE | ID: mdl-31270223

RÉSUMÉ

The Zika virus (ZIKV) life cycle involves multiple steps and requires interactions with host factors. However, the inability to systematically identify host regulatory factors for ZIKV has hampered antiviral development and our understanding of pathogenicity. Here, using a bioactive compound library with 2,659 small molecules, we applied a high-throughput and imaging-based screen to identify host factors that modulate ZIKV infection. The screen yielded hundreds of hits that markedly inhibited or potentiated ZIKV infection in SNB-19 glioblastoma cells. Among the hits, URMC-099, a mixed-lineage kinase 3 (MLK3) inhibitor, significantly facilitated ZIKV replication in both SNB-19 cells and the neonatal mouse brain. Using gene silencing and overexpression, we further confirmed that MLK3 was a host restriction factor against ZIKV. Mechanistically, MLK3 negatively regulated ZIKV replication through induction of the inflammatory cytokines interleukin-6 (IL-6), IL-8, tumor necrosis factor alpha (TNF-α), and monocyte chemoattractant protein 1 (MCP-1) but did not modulate host interferon-related pathways. Importantly, ZIKV activated the MLK3/MKK7/Jun N-terminal protein kinase (JNK) pathway in both SNB-19 cells and neonatal mouse brain. Together, these findings reveal a critical role for MLK3 in regulating ZIKV infection and facilitate the development of anti-ZIKV therapeutics by providing a number of screening hits.IMPORTANCE Zika fever, an infectious disease caused by the Zika virus (ZIKV), normally results in mild symptoms. Severe infection can cause Guillain-Barré syndrome in adults and birth defects, including microcephaly, in newborns. Although ZIKV was first identified in Uganda in 1947 in rhesus monkeys, a widespread epidemic of ZIKV infection in South and Central America in 2015 and 2016 raised major concerns. To date, there is no vaccine or specific medicine for ZIKV. The significance of our research is the systematic discovery of small molecule candidates that modulate ZIKV infection, which will allow the development of antiviral therapeutics. In addition, we identified MLK3, a key mediator of host signaling pathways that can be activated during ZIKV infection and limits virus replication by inducing multiple inflammatory cytokines. These findings broaden our understanding of ZIKV pathogenesis.


Sujet(s)
MAP Kinase Kinase Kinases/métabolisme , Infection par le virus Zika/métabolisme , Virus Zika/métabolisme , Animaux , Antiviraux/pharmacologie , Lignée cellulaire , Effet cytopathogène viral/effets des médicaments et des substances chimiques , Effet cytopathogène viral/physiologie , Tests de criblage à haut débit/méthodes , Interactions hôte-pathogène/effets des médicaments et des substances chimiques , Humains , Interférons/pharmacologie , MAP Kinase Kinase Kinases/génétique , Souris , Souris de lignée BALB C , Microcéphalie , Virus respiratoire syncytial humain , Virus Sendai , Transduction du signal/effets des médicaments et des substances chimiques , Réplication virale/effets des médicaments et des substances chimiques , Infection par le virus Zika/virologie ,
8.
Vector Borne Zoonotic Dis ; 16(11): 722-727, 2016 11.
Article de Anglais | MEDLINE | ID: mdl-27710208

RÉSUMÉ

BACKGROUND: Alkhumra hemorrhagic fever virus (AHFV) is a flavivirus that was discovered in 1995 in Saudi Arabia. Clinical manifestations of AHFV infection include hemorrhagic fever, hepatitis, and encephalitis with a reported mortality rate as high as 25%. There are no published data on the growth characteristics of AHFV in mammalian cell lines. The objective of this study was to examine the ability of AHFV to grow and propagate in four of the commonly used mammalian cell culture lines and to determine the virus growth curve characteristics in each. MATERIALS AND METHODS: Human epidermoid carcinoma (HEp-2), LLC-MK2, Madin-Darby canine kidney (MDCK), and Vero cell lines were inoculated with AHFV. The virus production by each cell line was determined by growth curve studies. Mean titers were calculated and expressed as median tissue culture infective dose per mL (TCID50/mL). RESULTS: AHFV grew and propagated to variable titers in the employed cell lines. The highest mean titers were observed in the LLC-MK2, followed by the MDCK, Vero, and HEP-2, in descending order. CONCLUSIONS: The growth curve studies showed that AHFV can propagate in the four types of cell lines to variable titers. LLC-MK2 cells are superior to MDCK, Vero, and HEP-2 for propagation of AHFV.


Sujet(s)
Virus de l'encéphalite à tiques (sous-groupe)/physiologie , Culture virale/méthodes , Réplication virale/physiologie , Animaux , Lignée cellulaire , Chlorocebus aethiops , Effet cytopathogène viral/physiologie , Chiens , Humains
10.
Med Sci (Paris) ; 31(5): 506-14, 2015 May.
Article de Français | MEDLINE | ID: mdl-26059301

RÉSUMÉ

Following primary infections HSV-1 replicates productively in epithelial cells and enters sensory neurons via nerve termini. After retrograde transport the virus genome is delivered into the cell nucleus, where it establishes lifelong latent infections. During latency, the virus genome remains as a chromatinized episome expressing only a set of latency-associated transcripts (LAT) and a group of microRNAs that inhibit expression of key lytic viral functions. Periodically the virus can reactivate to reinitiate lytic, secondary infections at peripheral tissues. The ability to establish both lytic and latent infections relies on the coexistence in the virus genome of two alternative gene expression programs, under the control of epigenetic mechanisms. Latency is an adaptive phenotype that allows the virus to escape immune host responses and to reactivate and disseminate to other hosts upon recognizing danger signals such as stress, neurologic trauma or growth factor deprivation.


Sujet(s)
Herpès/virologie , Herpèsvirus humain de type 1/physiologie , Activation virale/physiologie , Latence virale/physiologie , Capside/physiologie , Effet cytopathogène viral/physiologie , Épigenèse génétique , Cellules épithéliales/virologie , Régulation de l'expression des gènes viraux , Gènes viraux , Herpès/immunologie , Herpèsvirus humain de type 1/génétique , Herpèsvirus humain de type 1/immunologie , Interactions hôte-pathogène , Humains , microARN/génétique , microARN/physiologie , ARN viral/physiologie , Cellules réceptrices sensorielles/virologie , Stress physiologique , Protéines virales/génétique , Protéines virales/physiologie , Pénétration virale , Libération de particules virales/physiologie , Réplication virale/physiologie
11.
BMC Res Notes ; 7: 158, 2014 Mar 18.
Article de Anglais | MEDLINE | ID: mdl-24642084

RÉSUMÉ

BACKGROUND: Respiratory syncytial virus (RSV) is a common respiratory pathogen that can cause severe pneumonia. In vivo studies of RSV can be difficult due to variation in viral infection and disease severity in some animal models. Factors that may contribute to the variation are decreases in viral titer due to preparation and storage and method of virus administration. Nebulization is one method of RSV administration that provides even distribution of virus to all lung lobes; however, the exact quantity of the virus killed by nebulization is not defined. To test the hypothesis that sucrose enhances RSV stability and infectivity, a series of in vitro experiments were conducted with RSV strain Memphis 37 stored at varying concentrations (0%, 3%, 5%, 8%, 10%, 15%, and 20%) of sucrose as a possible cryo- and nebulization protectant. The optimal in vitro concentration was then assessed in vivo in a lamb model. METHODS: Prior to titering the virus on HEp-2 cells, the various virus solutions were subjected to one freeze-thaw cycle and one nebulization cycle. Forty-eight hours after viral plating, infectious foci were detected and counted using immunofluorescent imaging. Titers were determined after freeze-thaw and after freeze-thaw followed by nebulization, then compared to the stock titers (before freezing) as well as to one another to determine the loss of infectivity. To further test this in vivo, lambs 2 to 3-days-old were infected via nebulization with RSV using inoculate containing either 20% sucrose or no sucrose followed by assessments of infection severity. RESULTS: Nebulization of virus in 0% sucrose resulted in a 0.580 log reduction in infectivity while virus in 20% sucrose exhibited a 0.297 log reduction. In vivo studies demonstrated that 20% sucrose enhanced RSV lesions and antigen distribution. CONCLUSIONS: The data suggests that both nebulization and freeze-thawing of RSV in the absence of sucrose cause unacceptable losses in viral infectivity and that sucrose acts as a RSV protectant in both regards.


Sujet(s)
Effet cytopathogène viral/effets des médicaments et des substances chimiques , Poumon/effets des médicaments et des substances chimiques , Infections à virus respiratoire syncytial/virologie , Virus respiratoires syncytiaux/effets des médicaments et des substances chimiques , Saccharose/pharmacologie , Animaux , Animaux nouveau-nés , Antigènes viraux/immunologie , Antigènes viraux/métabolisme , Lignée cellulaire tumorale , Effet cytopathogène viral/physiologie , Modèles animaux de maladie humaine , Relation dose-effet des médicaments , Femelle , Congélation , Interactions hôte-pathogène/effets des médicaments et des substances chimiques , Humains , Poumon/virologie , Mâle , Microscopie de fluorescence , Nébuliseurs et vaporisateurs , Virus respiratoires syncytiaux/génétique , Virus respiratoires syncytiaux/immunologie , RT-PCR , Ovis , Protéines virales/génétique , Protéines virales/métabolisme
12.
J Vet Diagn Invest ; 26(2): 221-5, 2014 Mar.
Article de Anglais | MEDLINE | ID: mdl-24569225

RÉSUMÉ

A cell line was established from whole 6-8-week-old central bearded dragon (Pogona vitticeps) embryos. Cells were mid-sized and showed an elongated and polymorphic form. The cell line grew in a monolayer and has been serially passaged for 17 passages at time of publication. This cell line has been used with samples from adenovirus polymerase chain reaction (PCR)-positive bearded dragons, and 2 virus isolates have been obtained so far. The isolates show a clear cytopathic effect in inoculated cells. Both virus isolates have been serially passaged on this cell line, and have been identified by PCR amplification and sequencing of a portion of the DNA-dependent DNA polymerase gene and show 100% nucleotide identity to the corresponding region of an agamid adenovirus. Electron microscopic examination of supernatant from infected cells demonstrated the presence of nonenveloped particles, with a diameter of approximately 80 nm in both virus isolates.


Sujet(s)
Atadenovirus/isolement et purification , Lézards/virologie , Culture virale/méthodes , Animaux , Atadenovirus/physiologie , Lignée cellulaire , Effet cytopathogène viral/physiologie , Lézards/embryologie , Passages en série , Spécificité d'espèce
13.
J Immunol ; 192(3): 1171-83, 2014 Feb 01.
Article de Anglais | MEDLINE | ID: mdl-24391214

RÉSUMÉ

Semliki Forest virus (SFV) requires RNA replication and Bax/Bak for efficient apoptosis induction. However, cells lacking Bax/Bak continue to die in a caspase-dependent manner. In this study, we show in both mouse and human cells that this Bax/Bak-independent pathway involves dsRNA-induced innate immune signaling via mitochondrial antiviral signaling (MAVS) and caspase-8. Bax/Bak-deficient or Bcl-2- or Bcl-xL-overexpressing cells lacking MAVS or caspase-8 expression are resistant to SFV-induced apoptosis. The signaling pathway triggered by SFV does neither involve death receptors nor the classical MAVS effectors TNFR-associated factor-2, IRF-3/7, or IFN-ß but the physical interaction of MAVS with caspase-8 on mitochondria in a FADD-independent manner. Consistently, caspase-8 and -3 activation are reduced in MAVS-deficient cells. Thus, after RNA virus infection MAVS does not only elicit a type I antiviral response but also recruits caspase-8 to mitochondria to mediate caspase-3 activation and apoptosis in a Bax/Bak-independent manner.


Sujet(s)
Protéines adaptatrices de la transduction du signal/physiologie , Apoptose/physiologie , Caspase 8/physiologie , Effet cytopathogène viral/physiologie , Mitochondries/physiologie , Virus de la forêt de Semliki/physiologie , Animaux , Caspase-3/métabolisme , DEAD-box RNA helicases/physiologie , Activation enzymatique , Protéine à domaine de mort associée à Fas/physiologie , Fibroblastes/virologie , Cellules HEK293/virologie , Cellules HeLa/virologie , Humains , Hélicase IFIH1 inductrice de l'interféron , Souris , Mitochondries/enzymologie , Protéines proto-oncogènes c-bcl-2/métabolisme , Interférence par ARN , ARN viral/génétique , Transduction du signal , Réplication virale , Protéine bcl-X/métabolisme
14.
J Vet Med Sci ; 76(12): 1635-9, 2014 Dec.
Article de Anglais | MEDLINE | ID: mdl-25649948

RÉSUMÉ

Field isolates of BVDV which do not show the exaltation of Newcastle disease virus (END) phenomenon (END(-)) are rarely reported. In this study, 45 BVDV field isolates from cattle in Hokkaido prefecture in Japan were analyzed by the reverse plaque formation method, the END method and observation of cytopathic effects. END(-) virus was detected in 34 of 45 isolates (75.6%), although 35 of 45 field isolates contained END phenomenon positive virus as the predominant virus population. We propose that END(-) viruses are widely distributed in the field and that it is possible that the mixture of biologically distinct BVDV correlates with the appearance of disease in infected animals.


Sujet(s)
Diarrhée virale bovine-maladie des muqueuses/épidémiologie , Diarrhée virale bovine-maladie des muqueuses/virologie , Maladies des bovins/épidémiologie , Maladies des bovins/virologie , Virus de la diarrhée virale bovine/génétique , Virus de la diarrhée virale bovine/pathogénicité , Animaux , Séquence nucléotidique , Bovins , Cellules cultivées , Effet cytopathogène viral/physiologie , Japon/épidémiologie , Mâle , Données de séquences moléculaires , Virus de la maladie de Newcastle/croissance et développement , Oligonucléotides/génétique , Analyse de séquence d'ADN/médecine vétérinaire , Spécificité d'espèce , Testicule/cytologie
15.
J Theor Biol ; 345: 32-42, 2014 Mar 21.
Article de Anglais | MEDLINE | ID: mdl-24361326

RÉSUMÉ

Marine viruses shape the structure of the microbial community. They are, thus, a key determinant of the most important biogeochemical cycles in the planet. Therefore, a correct description of the ecological and evolutionary behavior of these viruses is essential to make reliable predictions about their role in marine ecosystems. The infection cycle, for example, is indistinctly modeled in two very different ways. In one representation, the process is described including explicitly a fixed delay between infection and offspring release. In the other, the offspring are released at exponentially distributed times according to a fixed release rate. By considering obvious quantitative differences pointed out in the past, the latter description is widely used as a simplification of the former. However, it is still unclear how the dichotomy "delay versus rate description" affects long-term predictions of host-virus interaction models. Here, we study the ecological and evolutionary implications of using one or the other approaches, applied to marine microbes. To this end, we use mathematical and eco-evolutionary computational analysis. We show that the rate model exhibits improved competitive abilities from both ecological and evolutionary perspectives in steady environments. However, rate-based descriptions can fail to describe properly long-term microbe-virus interactions. Moreover, additional information about trade-offs between life-history traits is needed in order to choose the most reliable representation for oceanic bacteriophage dynamics. This result affects deeply most of the marine ecosystem models that include viruses, especially when used to answer evolutionary questions.


Sujet(s)
Évolution biologique , Effet cytopathogène viral/physiologie , Modèles biologiques , Latence virale/physiologie , Virus/pathogénicité , Animaux , Organismes aquatiques/virologie , Bactériophages/physiologie , Interactions hôte-pathogène/physiologie , Étapes du cycle de vie/physiologie , Océans et mers , Maladies virales/virologie
16.
Vopr Virusol ; 58(4): 20-8, 2013.
Article de Russe | MEDLINE | ID: mdl-24354061

RÉSUMÉ

The propagation of the pandemic influenza virus H1N1 in cultures of bronchial (Calu-3) and intestinal (Caco-2) differentiated epithelial cells of human origin was studied. The canine epithelial cell lines, MDCK-H and MDCK-2, were comparatively tested. The two human cell lines were found to be highly sensitive to the influenza pandemic strains A/Hamburg/05/09 and A/Moscow/501/2011 and maintained their replication without addition of trypsin to culture medium. Virus strains of seasonal influenza H1N1, such as A/Moscow/450/2003, A/Memphis/14/96, and laboratory strain A/PR/8/34, multiplied in these human cells in similar manner. The intracellular cleavage HA0-->HA1+HA2 by the host virus-activating protease (IAP) occurred in both human cell lines under infection with each influenza virus H1N1 including pandemic ones. Comparatively, this cleavage of all influenza H1N1 virus strains appeared to be either undetectable or low-detectible in MDCK-H and MDCK-2, respectively, thereby implying low levels of active IAP in these cells. Multiplication of pandemic and seasonal influenza H1N1 viruses in Calu-3 and Caco-2 cells caused cytopathic effect, which was accompanied with low autophagy and apoptosis events. These data allow recommending human cell lines, Calu-3 and Caco-2, for optimized isolation and passaging of clinical strains of Influenza pandemic viruses H1N1.


Sujet(s)
Sous-type H1N1 du virus de la grippe A/physiologie , Grippe humaine/épidémiologie , Grippe humaine/métabolisme , Pandémies , Réplication virale/physiologie , Animaux , Cellules Caco-2 , Effet cytopathogène viral/physiologie , Chiens , Humains , Cellules rénales canines Madin-Darby
17.
Methods Mol Biol ; 1030: 383-406, 2013.
Article de Anglais | MEDLINE | ID: mdl-23821284

RÉSUMÉ

Mechanism of action studies can be used to demonstrate an inhibitor's ability to specifically inhibit viral replication via a virus-specific or host cell target. A well-characterized mechanism of action is useful in evaluating potential off-target toxicities (e.g., a viral polymerase inhibitor may be assessed for inhibition of host polymerase) and in designing studies to monitor the development of resistance. Several methods can be used to elucidate the mechanism of action of an anti-influenza inhibitor. The first group of methods establishes that the activity of an inhibitor occurs at concentrations that do not cause cytotoxicity, investigates the selective inhibition of influenza, and indicates that inhibition is virus specific in nature. The second group of methods establishes the site of action, typically a target protein, and includes genotypic and phenotypic analysis of variants selected under inhibitor pressure. Finally, methods for measuring virion associated activities and their inhibition are described.


Sujet(s)
Antiviraux/pharmacologie , Virus de la grippe A/effets des médicaments et des substances chimiques , Virus de la grippe A/physiologie , Animaux , Survie cellulaire , Effet cytopathogène viral/effets des médicaments et des substances chimiques , Effet cytopathogène viral/physiologie , Résistance virale aux médicaments/génétique , Génotype , Techniques de génotypage , Glycoprotéine hémagglutinine du virus influenza/génétique , Humains , Tests de sensibilité microbienne/méthodes , Mutation , Sialidase/génétique , Protéines virales/génétique , Virion/effets des médicaments et des substances chimiques , Virion/isolement et purification , Virion/physiologie
18.
J Virol ; 87(17): 9953-8, 2013 Sep.
Article de Anglais | MEDLINE | ID: mdl-23824802

RÉSUMÉ

The newly emerged Middle East respiratory syndrome coronavirus (MERS-CoV) infects human bronchial epithelial Calu-3 cells. Unlike severe acute respiratory syndrome (SARS)-CoV, which exclusively infects and releases through the apical route, this virus can do so through either side of polarized Calu-3 cells. Infection results in profound apoptosis within 24 h irrespective of its production of titers that are lower than those of SARS-CoV. Together, our results provide new insights into the dissemination and pathogenesis of MERS-CoV and may indicate that the virus differs markedly from SARS-CoV.


Sujet(s)
Bronches/virologie , Coronavirus/physiologie , Coronavirus/pathogénicité , Apoptose , Bronches/anatomopathologie , Lignée cellulaire , Polarité de la cellule , Effet cytopathogène viral/physiologie , Cellules épithéliales/anatomopathologie , Cellules épithéliales/virologie , Humains , Virus du SRAS/pathogénicité , Virus du SRAS/physiologie , Spécificité d'espèce , Pénétration virale , Libération de particules virales/physiologie
19.
Prikl Biokhim Mikrobiol ; 49(1): 72-81, 2013.
Article de Russe | MEDLINE | ID: mdl-23662454

RÉSUMÉ

The results of macrophage metabolism studies at their infection by viruses differing in the level of virulence are presented. With the purpose of optimizing the estimation of viral cytopathogenic effects on macrophages, an index of cell reactions, which allows one to reveal the degree of virus influence in standard units, is offered. Generally, the application of high-sensitivity methods for functional activity determination and identification of the correlative communication between its changes and morphological features of cells can be prescribed to objective identification methods of not only viral reproduction, but also differentiation of types and the degree of their cytopathogenic effects.


Sujet(s)
Virus de l'encéphalite à tiques (sous-groupe)/physiologie , Encéphalites à tiques/métabolisme , Encéphalites à tiques/anatomopathologie , Macrophages/anatomopathologie , Macrophages/virologie , Réplication virale/physiologie , Animaux , Cellules cultivées , Effet cytopathogène viral/physiologie , Souris
20.
Braz J Infect Dis ; 16(1): 19-26, 2012.
Article de Anglais | MEDLINE | ID: mdl-22358351

RÉSUMÉ

OBJECTIVE: Epstein-Barr virus (EBV) is a ubiquitous human γ-herpes virus, which can adapt and evade host immune defense. Dendritic cells (DCs) play a pivotal role in the initiation and maintenance of immune responses. This study investigated the effects of EBV on cord blood monocytes derived DCs (CBDC). METHODS: Monocytes were isolated from cord blood and cultured in medium containing recombinant IL-4 and GM-CSF to induce DCs development. B95-8 supernatant was added in monocytes culture medium for EBV infection at day 0. Phenotypic characterization of DCs, apoptotic cells, and mitochondrial membrane potential (MMP) were detected by flow cytometry. The morphology was observed by Hoechst 33258 staining and TUNEL staining, the expression of X-linked inhibitor of apoptosis protein (XIAP) was detected by Western blotting assay and caspase 3, 8 and 9 activity was measured. RESULTS: Phenotypic characterization of DCs was changed in EBV-treated group. Chromatin condensation and DNA fragmentation were observed in EBV induced CBDC apoptosis. In addition, caspase 3, caspase 8, and caspase 9 activation were enhanced in the EBV-treated group. This was accompanied by the loss of MMP. Furthermore, XIAP expression was down-regulated in the EBV-treated group and compared to mock-infected group. CONCLUSION: These results suggested that EBV could inhibit CBDC phenotypic differentiation, and induce CBDC apoptosis in caspase-dependent manner with involvement of the mitochondrial pathway. This might help EBV to evade host immune responses to establish persistent infection.


Sujet(s)
Apoptose/physiologie , Effet cytopathogène viral/physiologie , Cellules dendritiques/anatomopathologie , Sang foetal/cytologie , Herpèsvirus humain de type 4/physiologie , Monocytes/anatomopathologie , Technique de Western , Caspases/immunologie , Différenciation cellulaire , Cellules dendritiques/virologie , Cytométrie en flux , Herpèsvirus humain de type 4/immunologie , Humains , Interleukine-4/immunologie , Monocytes/cytologie , Monocytes/virologie , Phénotype , Protéine inhibitrice de l'apoptose liée au chromosome X/immunologie
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE
...