Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 12 de 12
Filtrer
Plus de filtres










Base de données
Gamme d'année
1.
J Equine Vet Sci ; 126: 104538, 2023 07.
Article de Anglais | MEDLINE | ID: mdl-37150233

RÉSUMÉ

The introduction of microfluidic card technology has opened the field for rapid point-of-care (POC) molecular assays, including fecal and environmental Salmonella spp. testing. The purpose of this study was to evaluate a novel POC PCR assay for the detection of Salmonella spp. in feces and environmental samples. A total of 143 fecal samples and 132 environmental samples were collected for POC PCR Salmonella spp. testing as well as qPCR testing. Each sample was inoculated into selenite broth and incubated for 18 to 24 hours. For the POC PCR assay, 14 µl of selenite broth were mixed with 126 µl of PCR reaction mix and pipetted into a microfluidic test card targeting the invA and ttrC gene of Salmonella enterica. For qPCR analysis, 200 µl of the selenite broth were processed for DNA purification and Salmonella spp. testing targeting the invA gene. The overall agreement between the POC PCR Salmonella spp. assay and qPCR assay was 88.1% for feces and 97.0% for environmental samples. Strong agreement and short turn-around-time make the POC device the first molecular diagnostic platform allowing detection of Salmonella spp. in a hospital setting without having to ship out samples to a veterinary diagnostic laboratory. The availability of an accurate POC PCR assay for the detection of Salmonella spp. will enhance the diagnostic capability of equine veterinarians to timely support a diagnosis of salmonellosis and also monitor the environment in order to reduce the risk of nosocomial infections.


Sujet(s)
Systèmes automatisés lit malade , Salmonella , Animaux , Equus caballus/génétique , Réaction de polymérisation en chaîne/médecine vétérinaire , Salmonella/génétique , Fèces
2.
Open Forum Infect Dis ; 8(7): ofab129, 2021 Jul.
Article de Anglais | MEDLINE | ID: mdl-34327248

RÉSUMÉ

Together with protective measures, routine screening for severe acute respiratory syndrome coronavirus 2 infection helps provide a safe working environment. We evaluated a pooled nucleic acid testing strategy in a research laboratory. It allowed lab activity to be maintained and would save 25 920 person-hours and $1 684 800/year by increasing the margin of safety for returning to work.

3.
J Virol ; 80(23): 11432-46, 2006 Dec.
Article de Anglais | MEDLINE | ID: mdl-17005646

RÉSUMÉ

Kaposi's sarcoma-associated herpesvirus (KSHV) (human herpesvirus 8) binds to adherent target cell surface heparan sulfate molecules via its envelope glycoproteins gB and gpK8.1A, to integrins via gB, to the transporter CD98/xCT complex, and possibly to another molecule(s). This is followed by virus entry overlapping with the induction of preexisting host cell signal pathways, such as focal adhesion kinase, Src, phosphatidylinositol 3-kinase (PI3-K), Rho-GTPases, protein kinase C-zeta, and extracellular signal-regulated kinase 1/2. Here, using hemagglutinin-tagged plasmids expressing wild-type, dominant-positive, and dominant-negative forms of RhoA in HEK (human embryonic kidney) 293 cells, we investigated the role of RhoA-GTPase in virus entry. The dominant-negative form of RhoA GTPase and treatment of target cells with Clostridium difficile toxin B (CdTxB), a specific inactivator of Rho-GTPases, significantly blocked KSHV entry. KSHV infection induced closely similar levels of FAK and PI3-K in all three cell types. In contrast, very strong Src activation was observed in KSHV-infected dominant-positive RhoA cells compared to wild-type cells, and only moderate Src activation was seen in dominant-negative cells. Inhibition of Src activation by CdTxB and reduction of RhoA activation by Src inhibitors suggest that KSHV-induced Src is involved in RhoA activation, which in turn is involved in a feedback-sustained activation of Src. Since the decreased entry in RhoA dominant-negative cells may be due to inefficient signaling downstream of RhoA, we examined the induction of RhoA-activated Dia-2, which is also known to induce Src. Dia-2 coimmunoprecipitated with activated Src, which was inhibited by Src inhibitors, in the infected cells. Together with the reduced virus entry in RhoA dominant-negative cells, these results suggest that activated RhoA-dependent Dia-2 probably functions as a link between RhoA and Src in KSHV-infected cells, mediating the sustained Src activation, and that KSHV-induced Src and RhoA play roles in facilitating entry into adherent target cells.


Sujet(s)
Adhérence cellulaire/physiologie , Herpèsvirus humain de type 8/physiologie , Protéines G rho/physiologie , src-Family kinases/métabolisme , Lignée cellulaire , Régulation de l'expression des gènes viraux , Herpèsvirus humain de type 8/génétique , Humains , Transduction du signal
4.
J Virol ; 80(13): 6534-52, 2006 Jul.
Article de Anglais | MEDLINE | ID: mdl-16775340

RÉSUMÉ

Infection of human dermal microvascular endothelial (HMVEC-d) cells and human foreskin fibroblast (HFF) cells in vitro by Kaposi's sarcoma-associated herpesvirus (KSHV) provides an excellent in vitro model system to study viral latency. KSHV infection is characterized by the induction of preexisting host signal cascades; sustained expression of the latency-associated open reading frame 73 (ORF73) (LANA-1), ORF72, and K13 genes; transient expression of a limited number of lytic genes, including the lytic cycle switch ORF50 (replication and transcription activator) gene; and reprogramming of host transcriptional machinery regulating a variety of cellular processes, including several proinflammatory responses. The cyclooxygenase 2 (COX-2) gene was one of the host cell genes that was highly up-regulated at 2 and 4 h postinfection (p.i.) of HMVEC-d and HFF cells (P. P. Naranatt, H. H. Krishnan, S. R. Svojanovsky, C. Bloomer, S. Mathur, and B. Chandran, Cancer Res. 64:72-84, 2004). Since COX-2 is an important mediator of inflammatory and angiogenic responses, here, using real-time PCR, Western blot, and immunofluorescence assays, we characterized the COX-2 stimulation and its role in KSHV infection. KSHV induced a robust COX-2 expression, which reached a maximum at 2 h p.i. in HMVEC-d cells and at 8 h p.i. in HFF cells, and significantly higher levels were continuously detected for up to 72 h p.i. Constitutive COX-1 protein levels were not modulated by KSHV infection. Moderate levels of COX-2 were also induced by UV-irradiated KSHV and by envelope glycoproteins gB and gpK8.1A; however, viral gene expression appears to be essential for the increased COX-2 induction. High levels of prostaglandin E(2) (PGE(2)), a COX-2 product, were released in the culture supernatant medium of infected cells. PGE(2) synthase, catalyzing the biosynthesis of PGE(2), also increased upon infection and inhibition of COX-2 by NS-398, and indomethacin drastically reduced the levels of PGE(2) and PGE(2) synthase. COX-2 inhibition did not affect KSHV binding, internalization of virus, or the trafficking to the infected cell nuclei. However, latent ORF73 gene expression and ORF73 promoter activity were significantly reduced by COX-2 inhibitors, and this inhibition was relieved by exogenous supplementation with PGE(2). In contrast, lytic ORF50 gene expression and ORF50 promoter activity were unaffected. These studies demonstrate that COX-2 and PGE(2) play roles in facilitating latent viral gene expression and the establishment and maintenance of latency and suggest that KSHV has evolved to utilize the inflammatory responses induced during infection of endothelial cells for the maintenance of viral latent gene expression.


Sujet(s)
Cyclooxygenase 2/biosynthèse , Régulation de l'expression des gènes viraux/physiologie , Infections à Herpesviridae/enzymologie , Herpèsvirus humain de type 8/physiologie , Protéines membranaires/biosynthèse , Protéines de l'enveloppe virale/métabolisme , Latence virale/physiologie , Lignée cellulaire , Cyclooxygenase 1/biosynthèse , Inhibiteurs des cyclooxygénases/pharmacologie , Dinoprostone/biosynthèse , Cellules endothéliales/enzymologie , Cellules endothéliales/virologie , Fibroblastes/enzymologie , Fibroblastes/virologie , Régulation de l'expression des gènes codant pour des enzymes/effets des médicaments et des substances chimiques , Régulation de l'expression des gènes codant pour des enzymes/physiologie , Régulation de l'expression des gènes codant pour des enzymes/effets des radiations , Régulation de l'expression des gènes viraux/effets des médicaments et des substances chimiques , Régulation de l'expression des gènes viraux/effets des radiations , Humains , Indométacine/pharmacologie , Inflammation/enzymologie , Inflammation/virologie , Nitrobenzènes/pharmacologie , Cadres ouverts de lecture/physiologie , Régions promotrices (génétique)/physiologie , Sulfonamides/pharmacologie , Transcription génétique/effets des médicaments et des substances chimiques , Transcription génétique/physiologie , Transcription génétique/effets des radiations , Rayons ultraviolets , Régulation positive/effets des médicaments et des substances chimiques , Régulation positive/physiologie , Régulation positive/effets des radiations , Latence virale/effets des médicaments et des substances chimiques , Latence virale/effets des radiations , Réplication virale/effets des médicaments et des substances chimiques , Réplication virale/physiologie , Réplication virale/effets des radiations
5.
J Virol ; 80(3): 1167-80, 2006 Feb.
Article de Anglais | MEDLINE | ID: mdl-16414994

RÉSUMÉ

Kaposi's sarcoma-associated herpesvirus/human herpesvirus 8 (KSHV/HHV-8) interacts with cell surface alpha3beta1 integrin early during in vitro infection of human endothelial cells and fibroblasts and activates the focal adhesion kinase (FAK) that is immediately downstream in the outside-in signaling pathway by integrins, leading to the activation of several downstream signaling molecules. In this study, using real-time DNA and reverse transcription-PCR assays to measure total internalized viral DNA, viral DNA associated with infected nuclei, and viral gene expression, we examined the stage of infection at which FAK plays the most significant role. Early during KSHV infection, FAK was phosphorylated in FAK-positive Du17 mouse embryonic fibroblasts. The absence of FAK in Du3 (FAK(-/-)) cells resulted in about 70% reduction in the internalization of viral DNA, suggesting that FAK plays a role in KSHV entry. Expression of FAK in Du3 (FAK(-/-)) cells via an adenovirus vector augmented the internalization of viral DNA. Expression of the FAK dominant-negative mutant FAK-related nonkinase (FRNK) in Du17 cells significantly reduced the entry of virus. Virus entry in Du3 cells, albeit in reduced quantity, delivery of viral DNA to the infected cell nuclei, and expression of KSHV genes suggested that in the absence of FAK, another molecule(s) may be partially compensating for FAK function. Infection of Du3 cells induced the phosphorylation of the FAK-related proline-rich tyrosine kinase (Pyk2) molecule, which has been shown to complement some of the functions of FAK. Expression of an autophosphorylation site mutant of Pyk2 in which Y402 is mutated to F (F402 Pyk2) reduced viral entry in Du3 cells, suggesting that Pyk2 facilitates viral entry moderately in the absence of FAK. These results suggest a critical role for KSHV infection-induced FAK in the internalization of viral DNA into target cells.


Sujet(s)
Focal adhesion protein-tyrosine kinases/physiologie , Herpèsvirus humain de type 8/physiologie , Herpèsvirus humain de type 8/pathogénicité , Animaux , Cellules cultivées , ADN viral/génétique , ADN viral/métabolisme , Focal adhesion kinase 2/antagonistes et inhibiteurs , Focal adhesion kinase 2/génétique , Focal adhesion kinase 2/métabolisme , Focal adhesion protein-tyrosine kinases/composition chimique , Focal adhesion protein-tyrosine kinases/déficit , Focal adhesion protein-tyrosine kinases/génétique , Herpèsvirus humain de type 8/génétique , Humains , Système de signalisation des MAP kinases , Souris , Souris knockout , Modèles biologiques , Phosphorylation , Protein-tyrosine kinases/génétique , Virulence
6.
J Virol ; 79(16): 10308-29, 2005 Aug.
Article de Anglais | MEDLINE | ID: mdl-16051824

RÉSUMÉ

Kaposi's sarcoma-associated herpesvirus (KSHV) in vitro target cell infection is characterized by the expression of the latency-associated genes ORF 73 (LANA-1), ORF 72, and K13 and by the transient expression of a very limited number of lytic genes such as lytic cycle switch gene ORF 50 (RTA) and the immediate early (IE) lytic K5, K8, and v-IRF2 genes. During the early stages of infection, several overlapping multistep complex events precede the initiation of viral gene expression. KSHV envelope glycoprotein gB induces the FAK-Src-PI3K-RhoGTPase (where FAK is focal adhesion kinase) signaling pathway. As early as 5 min postinfection (p.i.), KSHV induced the extracellular signal-regulated kinase 1 and 2 (ERK1/2) via the PI3K-PKCzeta-MEK pathway. In addition, KSHV modulated the transcription of several host genes of primary human dermal microvascular endothelial cells (HMVEC-d) and fibroblast (HFF) cells by 2 h and 4 h p.i. Neutralization of virus entry and infection by PI-3K and other cellular tyrosine kinase inhibitors suggested a critical role for signaling molecules in KSHV infection of target cells. Here we investigated the induction of ERK1/2 by KSHV and KSHV envelope glycoproteins gB and gpK8.1A and the role of induced ERK in viral and host gene expression. Early during infection, significant ERK1/2 induction was observed even with low multiplicity of infection of live and UV-inactivated KSHV in serum-starved cells as well as in the presence of serum. Entry of UV-inactivated virus and the absence of viral gene expression suggested that ERK1/2 induction is mediated by the initial signal cascade induced by KSHV binding and entry. Purified soluble gpK8.1A induced the MEK1/2 dependent ERK1/2 but not ERK5 and p38 mitogen-activated protein kinase (MAPK) in HMVEC-d and HFF. Moderate ERK induction with soluble gB was seen only in HMVEC-d. Preincubation of gpK8.1A with heparin or anti-gpK8.1A antibodies inhibited the ERK induction. U0126, a selective inhibitor for MEK/ERK blocked the gpK8.1A- and KSHV-induced ERK activation. ERK1/2 inhibition did not block viral DNA internalization and had no significant effect on nuclear delivery of KSHV DNA during de novo infection. Analyses of viral gene expression by quantitative real-time reverse transcriptase PCR revealed that pretreatment of cells with U0126 for 1 h and during the 2-h infection with KSHV significantly inhibited the expression of ORF 73, ORF 50 (RTA), and the IE-K8 and v-IRF2 genes. However, the expression of lytic IE-K5 gene was not affected significantly. Expression of ORF 73 in BCBL-1 cells was also significantly inhibited by preincubation with U0126. Inhibition of ERK1/2 also inhibited the transcription of some of the vital host genes such as DUSP5 (dual specificity phosphatase 5), ICAM-1 (intercellular adhesion molecule 1), heparin binding epidermal growth factor, and vascular endothelial growth factor that were up-regulated early during KSHV infection. Several MAPK-regulated host transcription factors such as c-Jun, STAT1alpha, MEF2, c-Myc, ATF-2 and c-Fos were induced early during infection, and ERK inhibition significantly blocked the c-Fos, c-Jun, c-Myc, and STAT1alpha activation in the infected cells. AP1 transcription factors binding to the RTA promoter in electrophoretic mobility shift assays were readily detected in the infected cell nuclear extracts which were significantly reduced by ERK inhibition. Together, these results suggest that very early during de novo infection, KSHV induces the ERK1/2 to modulate the initiation of viral gene expression and host cell genes, which further supports our hypothesis that beside the conduit for viral DNA delivery into the cytoplasm, KSHV interactions with host cell receptor(s) create an appropriate intracellular environment facilitating infection.


Sujet(s)
Régulation de l'expression des gènes viraux , Infections à Herpesviridae/enzymologie , Herpèsvirus humain de type 8/génétique , MAP Kinase Kinase 1/physiologie , MAP Kinase Kinase 2/physiologie , Mitogen-Activated Protein Kinase 1/physiologie , Mitogen-Activated Protein Kinase 3/physiologie , Transport nucléaire actif , ADN/métabolisme , Activation enzymatique , Humains , Protéines précoces immédiates/métabolisme , Système de signalisation des MAP kinases , Mitogen-Activated Protein Kinase 7/métabolisme , Cadres ouverts de lecture , Transactivateurs/métabolisme , Facteur de transcription AP-1/métabolisme , Protéines virales/métabolisme , p38 Mitogen-Activated Protein Kinases/métabolisme
7.
J Virol ; 79(2): 1191-206, 2005 Jan.
Article de Anglais | MEDLINE | ID: mdl-15613346

RÉSUMÉ

Human herpesvirus 8 (HHV-8; also called Kaposi's sarcoma-associated herpesvirus), which is implicated in the pathogenesis of Kaposi's sarcoma (KS) and lymphoproliferative disorders, infects a variety of target cells both in vivo and in vitro. HHV-8 binds to several in vitro target cells via cell surface heparan sulfate and utilizes the alpha3beta1 integrin as one of its entry receptors. Interactions with cell surface molecules induce the activation of host cell signaling cascades and cytoskeletal changes (P. P. Naranatt, S. M. Akula, C. A. Zien, H. H. Krishnan, and B. Chandran, J. Virol. 77:1524-1539, 2003). However, the mechanism by which the HHV-8-induced signaling pathway facilitates the complex events associated with the internalization and nuclear trafficking of internalized viral DNA is as yet undefined. Here we examined the role of HHV-8-induced cytoskeletal dynamics in the infectious process and their interlinkage with signaling pathways. The depolymerization of microtubules did not affect HHV-8 binding and internalization, but it inhibited the nuclear delivery of viral DNA and infection. In contrast, the depolymerization of actin microfilaments did not have any effect on virus binding, entry, nuclear delivery, or infection. Early during infection, HHV-8 induced the acetylation of microtubules and the activation of the RhoA and Rac1 GTPases. The inactivation of Rho GTPases by Clostridium difficile toxin B significantly reduced microtubular acetylation and the delivery of viral DNA to the nucleus. In contrast, the activation of Rho GTPases by Escherichia coli cytotoxic necrotizing factor significantly augmented the nuclear delivery of viral DNA. Among the Rho GTPase-induced downstream effector molecules known to stabilize the microtubules, the activation of RhoA-GTP-dependent diaphanous 2 was observed, with no significant activation in the Rac- and Cdc42-dependent PAK1/2 and stathmin molecules. The nuclear delivery of viral DNA increased in cells expressing a constitutively active RhoA mutant and decreased in cells expressing a dominant-negative mutant of RhoA. HHV-8 capsids colocalized with the microtubules, as observed by confocal microscopic examination, and the colocalization was abolished by the destabilization of microtubules with nocodazole and by the phosphatidylinositol 3-kinase inhibitor affecting the Rho GTPases. These results suggest that HHV-8 induces Rho GTPases, and in doing so, modulates microtubules and promotes the trafficking of viral capsids and the establishment of infection. This is the first demonstration of virus-induced host cell signaling pathways in the modulation of microtubule dynamics and in the trafficking of viral DNA to the infected cell nucleus. These results further support our hypothesis that HHV-8 manipulates the host cell signaling pathway to create an appropriate intracellular environment that is conducive to the establishment of a successful infection.


Sujet(s)
Protéines de transport/physiologie , Noyau de la cellule/virologie , ADN viral/métabolisme , Herpèsvirus humain de type 8/physiologie , Microtubules/physiologie , Transduction du signal , Protéine G RhoA/physiologie , Acétylation , Cellules cultivées , Formines , Humains , Protéine G rac1/physiologie
8.
J Virol ; 78(8): 4207-23, 2004 Apr.
Article de Anglais | MEDLINE | ID: mdl-15047836

RÉSUMÉ

Human herpesvirus 8 (HHV-8; Kaposi's sarcoma-associated herpesvirus) envelope glycoprotein gB possesses an RGD motif, interacts with alpha 3 beta 1 integrin, and uses it as one of the entry receptors. HHV-8 induces the integrin-dependent focal adhesion kinase (FAK), a critical step in the outside-in signaling pathways necessary for the subsequent phosphorylation of other cellular kinases, cytoskeletal rearrangements, and other functions. As an initial step toward deciphering the role of HHV-8 gB-integrin interaction in infection, signal pathways induced by gB were examined. A truncated form of gB without the transmembrane and carboxyl domains (gB Delta TM), a gB Delta TM mutant form (gB Delta TM-RGA) with an RGD-to-RGA mutation, and inhibitors of cellular kinases were used. HHV-8 gB Delta TM, but not gB Delta TM-RGA, induced FAK phosphorylation in target cells, which was in part dependent on the presence of alpha 3 beta 1 integrin. FAK was critical for the subsequent phosphorylation of Src by gB Delta TM, and Src induction was essential for the phosphorylation of phosphatidylinositol 3-kinase (PI-3K). HHV-8 gB Delta TM-induced PI-3K was essential for the induction of RhoA and Cdc42 Rho GTPases that was accompanied by the cytoskeletal rearrangements. These gB-induced morphological changes were inhibited by the PI-3K inhibitors. Ezrin, one of the essential elements required to cross-link the actin cytoskeleton with the plasma membrane and to induce the morphological changes, was induced by the Rho GTPases. Inhibition of cellular tyrosine kinases by the brief treatment of cells with 4',5,7-trihydroxyisoflavone (genistein) blocked the entry of HHV-8 into target cells. These findings suggest that, independently of other viral glycoproteins and via its RGD motif, HHV-8 gB induces integrin-dependent pre-existing FAK-Src-PI-3K-Rho GTPase kinases. Since these signal pathways play vital roles in host cell endocytosis and movement of particulate materials in the cytoplasm, the early stages of HHV-8 gB interaction with host cells may provide a very conducive environment for the successful infection of target cells.


Sujet(s)
Herpèsvirus humain de type 8/physiologie , Herpèsvirus humain de type 8/pathogénicité , Protéines de l'enveloppe virale/physiologie , Animaux , Cellules CHO , Lignée cellulaire , Cricetinae , Cytosquelette/métabolisme , Cytosquelette/ultrastructure , Antienzymes/pharmacologie , Focal adhesion kinase 1 , Focal adhesion protein-tyrosine kinases , Génistéine/pharmacologie , Herpèsvirus humain de type 8/effets des médicaments et des substances chimiques , Herpèsvirus humain de type 8/génétique , Humains , Intégrine alpha3 bêta1/métabolisme , Cinétique , Modèles biologiques , Phosphatidylinositol 3-kinases/métabolisme , Phosphorylation , Protein-tyrosine kinases/métabolisme , Délétion de séquence , Transduction du signal , Protéines de l'enveloppe virale/composition chimique , Protéines de l'enveloppe virale/génétique , Virulence/effets des médicaments et des substances chimiques , Protéines G rho/métabolisme , src-Family kinases/métabolisme
9.
J Virol ; 78(7): 3601-20, 2004 Apr.
Article de Anglais | MEDLINE | ID: mdl-15016882

RÉSUMÉ

Kaposi's sarcoma-associated herpesvirus (KSHV) infection of in vitro target cells is characterized by the expression of the latency-associated open reading frame (ORF) 73 gene (LANA-1) and the absence of progeny virus production. This default latent infection can be switched into lytic cycle by phorbol ester and by the lytic cycle ORF 50 (RTA) protein. In this study, the kinetics of latent and lytic gene expression immediately following KSHV infection of primary human dermal microvascular endothelial (HMVEC-d) and foreskin fibroblast (HFF) cells were examined by real-time reverse transcriptase PCR and whole-genome array. Within 2 h postinfection (p.i.), high levels of ORF 50 transcripts were detected in both cell types, which declined sharply by 24 h p.i. In contrast, comparatively low levels of ORF 73 expression were detected within 2 h p.i., increased subsequently, were maintained at a steady state, and declined slowly by 120 h p.i. The RTA and LANA-1 proteins were detected in the majority of infected cells by immunoperoxidase assays. In genome array, only 29 of 94 (31%) KSHV genes were expressed, which included 11 immediate-early/early, 8 early, and 5 late lytic genes and 4 latency-associated genes. While the expression of latent ORF 72, 73, and K13 genes continued, nearly all of the lytic genes declined or were undetectable by 8 and 24 h p.i. in HMVEC-d and HFF cells, respectively. Only a limited number of RTA-activated KSHV genes were expressed briefly, and the majority of KSHV genes involved in viral DNA synthesis and structural proteins were not expressed. However, early during infection, the lytic K2, K4, K5, K6, and vIRF2 genes with immune modulation functions and the K7 gene with antiapoptotic function were expressed. Expression of K5 was detected for up to 5 days of observation, and vIRF2 was expressed up to 24 h p.i. The full complement of lytic cycle genes were expressed when 12-O-tetradecanoylphorbol-13-acetate was added to the HMVEC-d cells after 48 h p.i. These data suggest that in contrast to alpha- and betaherpesviruses and some members of gammaherpesviruses, gamma-2 KSHV in vitro infection is characterized by the concurrent expression of latent and a limited number of lytic genes immediately following infection and a subsequent decline and/or absence of lytic gene expression with the persistence of latent genes. Expression of its limited lytic cycle genes could be a "strategy" that evolved in KSHV allowing it to evade the immune system and to provide the necessary factors and time to establish and/or maintain latency during the initial phases of infection. These are unique observations among in vitro herpesvirus infections and may have important implications in KSHV biology and pathogenesis.


Sujet(s)
Apoptose , Cellules endothéliales/virologie , Fibroblastes/virologie , Régulation de l'expression des gènes viraux , Herpèsvirus humain de type 8/physiologie , Activation virale/génétique , Latence virale/génétique , Antigènes viraux , Cellules cultivées , Gènes viraux/génétique , Herpèsvirus humain de type 8/génétique , Herpèsvirus humain de type 8/immunologie , Humains , Protéines précoces immédiates/génétique , Protéines précoces immédiates/métabolisme , Cinétique , Protéines nucléaires/génétique , Protéines nucléaires/métabolisme , Séquençage par oligonucléotides en batterie , Cadres ouverts de lecture/génétique , RT-PCR , Facteurs temps , Transactivateurs/génétique , Transactivateurs/métabolisme , Protéines virales/génétique , Protéines virales/immunologie , Protéines virales/métabolisme
10.
Cancer Res ; 64(1): 72-84, 2004 Jan 01.
Article de Anglais | MEDLINE | ID: mdl-14729610

RÉSUMÉ

Kaposi's sarcoma-associated herpesvirus (KSHV/HHV-8) is etiologically linked to the endothelial tumor Kaposi's sarcoma and with two lymphoproliferatve disorders, primary effusion lymphoma and multicentric Castleman's disease. HHV-8 infects a variety of target cells both in vivo and in vitro, binds to the in vitro target cells via cell surface heparan sulfate, and uses the alpha(3)beta(1) integrin as one of the entry receptors. Within minutes of infection, HHV-8 induced the integrin-mediated signaling pathways and morphological changes in the target cells (S. M. Akula et al., Cell, 108: 407-419, 2002; P. P. Naranatt et al., J. Virol., 77: 1524-1539, 2003). As an initial step toward understanding the role of host genes in HHV-8 infection and pathogenesis, modulation of host cell gene expression immediately after infection was examined. To reflect HHV-8's broad cellular tropism, mRNAs collected at 2 and 4 h after infection of primary human endothelial [human adult dermal microvascular endothelial cells (HMVECd)] and foreskin fibroblast [human foreskin fibroblast (HFF)] cells and human B cell line (BJAB) were analyzed by oligonucleotide array with approximately 22,000 human transcripts. With a criteria of >2-fold gene induction as significant, approximately 1.72% of the genes were differentially expressed, of which, 154 genes were shared by at least two cells and 33 genes shared by all three cells. HHV-8-induced transcriptional profiles in the endothelial and fibroblast cells were closely similar, with substantial differences in the B cells. In contrast to the antiapoptotic regulators induced in HMVECd and HFF cells, proapoptotic regulators were induced in the B cells. A robust increase in the expression of IFN-induced genes suggestive of innate immune response induction was observed in HMVECd and HFF cells, whereas there was a total lack of immunity related protein inductions in B cells. These striking cell type-specific behaviors suggest that HHV-8-induced host cell gene modulation events in B cells may be different compared with the adherent endothelial and fibroblast target cells. Functional clustering of modulated genes identified several host molecules hitherto unknown to HHV-8 infection. These results indicate that early during infection, HHV-8 reprograms the host transcriptional machinery regulating a variety of cellular processes including apoptosis, transcription, cell cycle regulation, signaling, inflammatory response, and angiogenesis, all of which may play important roles in the biology and pathogenesis of HHV-8.


Sujet(s)
Lymphocytes B/virologie , Endothélium vasculaire/virologie , Fibroblastes/virologie , Régulation de l'expression des gènes viraux , Herpèsvirus humain de type 8/génétique , Transcription génétique/génétique , Adulte , Cellules cultivées , Analyse de regroupements , Enzymes/génétique , Analyse de profil d'expression de gènes/méthodes , Marqueurs génétiques , Herpèsvirus humain de type 8/pathogénicité , Humains , Nouveau-né , Mâle , Microcirculation , Sondes oligonucléotidiques , Protéines/génétique , RT-PCR , Sarcome de Kaposi/génétique , Sarcome de Kaposi/virologie , Peau/vascularisation , Activation de la transcription
11.
J Virol ; 77(14): 7978-90, 2003 Jul.
Article de Anglais | MEDLINE | ID: mdl-12829837

RÉSUMÉ

Kaposi's sarcoma (KS)-associated herpesvirus or human herpesvirus 8 (HHV-8) DNA and transcripts have been detected in the B cells, macrophages, keratinocytes, and endothelial and epithelial cells of KS patients. In vitro, HHV-8 infects human B, endothelial, epithelial, and fibroblast cells, as well as animal cells, and the infection is characterized by (i) absence of lytic replication by the input virus and (ii) latent infection. For its initial binding to target cells, HHV-8 uses ubiquitous heparan sulfate molecules via its envelope-associated glycoproteins gB and gpK8.1A. HHV-8 also interacts with the alpha3beta1 integrin via its glycoprotein gB, and virus binding studies suggest that alpha3beta1 is one of the HHV-8 entry receptors (S. M. Akula, N. P. Pramod, F. Z. Wang, and B. Chandran, Cell 108:407-419, 2002). In this study, morphological and biochemical techniques were used to examine the entry of HHV-8 into human foreskin fibroblasts (HFF). HHV-8 was detected in coated vesicles and in large, smooth-surfaced endocytic vesicles. Fusion of viral envelope with the vesicle wall was also observed. In immune electron microscopy, anti-HHV-8 gB antibodies colocalized with virus-containing endocytic vesicles. In fluorescence microscopic analyses, transferrin was colocalized with HHV-8. HHV-8 infection was significantly inhibited by preincubation of cells with chlorpromazine HCl, which blocks endocytosis via clathrin-coated pits, but not by nystatin and cholera toxin B, which blocks endocytosis via caveolae and induces the dissociation of lipid rafts, respectively. Infection was also inhibited by blocking the acidification of endosomes by NH(4)Cl and bafilomycin A. Inhibition of HHV-8 open reading frame 73 gene expression by chlorpromazine HCl, bafilomycin A, and NH(4)Cl demonstrated that the virions in the vesicles could proceed to cause an infection. Taken together, these findings suggest that for its infectious entry into HFF, HHV-8 uses clathrin-mediated endocytosis and a low-pH intracellular environment.


Sujet(s)
Endocytose , Fibroblastes/virologie , Herpèsvirus humain de type 8/pathogénicité , Lignée cellulaire , Cellules cultivées , Clathrine/antagonistes et inhibiteurs , Clathrine/métabolisme , Endosomes/virologie , Humains , Concentration en ions d'hydrogène , Microscopie électronique , Microscopie immunoélectronique , Sarcome de Kaposi/virologie , Transferrine/métabolisme
12.
J Virol ; 77(2): 1524-39, 2003 Jan.
Article de Anglais | MEDLINE | ID: mdl-12502866

RÉSUMÉ

Human herpesvirus 8 (HHV-8) is implicated in the pathogenesis of Kaposi's sarcoma. HHV-8 envelope glycoprotein B (gB) possesses the RGD motif known to interact with integrin molecules, and HHV-8 infectivity was inhibited by RGD peptides, by antibodies against alpha3 and beta1 integrins, and by soluble alpha3beta1 integrin (S. M. Akula, N. P. Pramod, F.-Z. Wang, and B. Chandran, Cell 108:407-419, 2002). Anti-gB antibodies immunoprecipitated the virus alpha3 and beta1 complexes, and virus-binding studies suggest a role for alpha3beta1 in HHV-8 entry. HHV-8 infection induced the integrin-mediated activation of focal adhesion kinase (FAK), implicating a role for integrin and the associated signaling pathways in HHV-8 entry into the target cells. Immediately after infection, target cells exhibited morphological changes and cytoskeletal rearrangements, suggesting the induction of signal pathways. As early as 5 min postinfection, HHV-8 activated the MEK-ERK1/2 pathway. The focal adhesion components phosphatidylinositol 3-kinase (PI 3-kinase) and protein kinase C-zeta (PKC-zeta) were recruited as upstream mediators of the HHV-8-induced ERK pathway. Anti-HHV-8 gB-neutralizing antibodies and soluble alpha3beta1 integrin inhibited the virus-induced signaling pathways. Early kinetics of the cellular signaling pathway and its activation by UV-inactivated HHV-8 suggest a role for virus binding and/or entry but not viral gene expression in this induction. Studies with human alpha3 integrin-transfected Chinese hamster ovary cells and FAK-negative mouse DU3 cells suggest that the alpha3beta1 integrin and FAK play roles in the HHV-8 mediated signal induction. Inhibitors specific for PI 3-kinase, PKC-zeta, MEK, and ERK significantly reduced the virus infectivity without affecting virus binding to the target cells. Examination of viral DNA entry suggests a role for PI 3-kinase in HHV-8 entry into the target cells and a role for PKC-zeta, MEK, and ERK at a post-viral entry stage of infection. These findings implicate a critical role for integrin-associated mitogenic signaling in HHV-8's infection of target cells and suggest that, by orchestrating the signal cascade, HHV-8 may create an appropriate intracellular environment to facilitate the infection.


Sujet(s)
Herpèsvirus humain de type 8/physiologie , Système de signalisation des MAP kinases/physiologie , Phosphatidylinositol 3-kinases/métabolisme , Protéine kinase C/métabolisme , Actines/métabolisme , Séquence nucléotidique , Biopolymères/métabolisme , Amorces ADN , Antienzymes/pharmacologie , Herpèsvirus humain de type 8/pathogénicité , Humains , Inhibiteurs des phosphoinositide-3 kinases , Virulence
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE
...