Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 56
Filtrar
Mais filtros

Bases de dados
Tipo de documento
Intervalo de ano de publicação
1.
Arch Microbiol ; 203(6): 3163-3169, 2021 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-33821299

RESUMO

In this study, aimed at investigating and characterizing river sediment bacteria, we isolated a Gram-stain-positive, rod-shaped, obligate anaerobic bacterium, strain CBA3637T, from the sediment of the Geum River. This strain grew at 10-40 °C (optimum, 30 °C), 0-1% NaCl (optimum, 0%), and pH 7-8 (optimum, pH 7). The 16S rRNA gene sequence comparison revealed Aminipila butyrica DSM 103574T to be the closest relative of strain CBA3637T (96.6-96.7% similarity); and both strains clustered together in phylogenetic analysis. The genome of strain CBA3637T was found to consist of a single chromosome (3.51 Mbp; 36.98% G + C content). Comparative genomic analysis of the strain CBA3637T with A. butyrica DSM 103574T revealed that strain CBA3637T possessed five unique pathways related to polyamine biosynthesis, lipopolysaccharide metabolism, pyrimidine metabolism, and cofactor and vitamin metabolism. Strain CBA3637T contained C14:0, C16:0, and C18:1 ω9c as the major fatty acids, and diphosphatidylglycerol as the major polar lipid. No respiratory quinone was observed. Biochemical, chemotaxonomic, and genotypic data revealed that the strain CBA3637T is a representative of a novel species within the genus Aminipila, for which the name Aminipila terrae is proposed. The type strain is CBA3637T (= KACC 21651T = DSM 110662T).


Assuntos
Clostridiales , Sedimentos Geológicos , Fosfolipídeos , Rios , Anaerobiose , Composição de Bases , Clostridiales/classificação , Clostridiales/genética , Clostridiales/isolamento & purificação , Ácidos Graxos/análise , Sedimentos Geológicos/microbiologia , Fosfolipídeos/análise , Filogenia , RNA Ribossômico 16S/genética , Rios/microbiologia
2.
Antonie Van Leeuwenhoek ; 114(5): 507-513, 2021 May.
Artigo em Inglês | MEDLINE | ID: mdl-33730291

RESUMO

Strain CBA3638T was isolated from the Geum River sediment, Republic of Korea. The cells of strain CBA3638T were Gram-stain-positive, strictly anaerobic, rod-shaped, and 0.5-1.0 µm wide, and 4.0-4.5 µm long. Optimal growth occurred at 37 °C, pH 7.0, and 1.0% (w/v) NaCl. Based on the 16S rRNA gene sequence, the phylogenetic analysis showed that strain CBA3638T belongs to the genus Anaerocolumna in the family Lachnospiraceae, and is most closely related to Anaerocolumna cellulosilytica (94.6-95.0%). The DDH value with A. cellulosilytica SN021T showed 15.0% relatedness. The genome of strain CBA3638T consisted of one circular chromosome that is 5,500,435 bp long with a 36.7 mol% G + C content. The genome contained seven 16S-5S-23S rRNA operons and one antibiotic resistance-related transporter gene (mefA). Quinones were not detected. The predominant cellular fatty acids were C16:0 and C14:0 and the polar lipids were diphosphatidylglycerol, phosphatidylcholine, and uncharacterised polar lipids. Based on the polyphasic taxonomic analysis, we propose strain CBA3638T as a novel species in the genus Anaerocolumna, with the name Anaerocolumna sedimenticola sp. nov. The type strain is CBA3638T (= KACC 21652T = DSM 110663T).


Assuntos
Água Doce , Fosfolipídeos , Técnicas de Tipagem Bacteriana , Composição de Bases , Clostridiales , DNA Bacteriano/genética , Ácidos Graxos/análise , Fosfolipídeos/análise , Filogenia , RNA Ribossômico 16S/genética , Análise de Sequência de DNA
3.
J Med Virol ; 90(4): 655-662, 2018 04.
Artigo em Inglês | MEDLINE | ID: mdl-29106738

RESUMO

Ten Yucatan miniature piglets were challenged with the human norovirus (NoV) GII.12/GII.3 CAU140599 strain and five piglets were used as negative controls. Stool, serum, and organs were collected and processed from two NoV-infected piglets and one negative piglet at 1, 2, 3, 5, and 7 days post-inoculation (dpi). NoV was detected in stool and serum samples by real-time RT-PCR. Mild diarrhea was observed at 1-3 dpi. Fecal shedding and viremia were detected intermittently at 1, 3, and 7 dpi. While interferon-α was significantly elevated at 2-3 dpi, interferon-γ was not changed. Immunohistochemistry demonstrated that the NoV capsid antigen was present in macrophages, lymphocytes, and dendritic cells of the stomach, intestines, lymph nodes, spleen, and tonsils. Intestinal epithelium did not exhibit a positive signal for NoV. In addition, negative-sense viral RNA was confirmed in immune cells by fluorescence in situ hybridization. Therefore, NoV might be associated with macrophages and lymphocytes in gastrointestinal tract and immune organs of experimentally infected miniature piglets.


Assuntos
Infecções por Caliciviridae/patologia , Modelos Animais de Doenças , Genótipo , Norovirus/patogenicidade , Estruturas Animais/patologia , Estruturas Animais/virologia , Animais , Animais Recém-Nascidos , Diarreia/patologia , Fezes/virologia , Imuno-Histoquímica , Linfócitos/virologia , Macrófagos/virologia , Norovirus/classificação , Norovirus/genética , Norovirus/isolamento & purificação , Reação em Cadeia da Polimerase em Tempo Real , Suínos , Porco Miniatura , Fatores de Tempo , Eliminação de Partículas Virais
4.
Biofouling ; 34(10): 1079-1092, 2018 11.
Artigo em Inglês | MEDLINE | ID: mdl-30698028

RESUMO

The effects of dual species interactions on biofilm formation by Aeromonas hydrophila in the presence of Pseudomonas aeruginosa, Pseudomonas fluorescens, Pectobacterium carotovorum, Salmonella Typhimurium, and Listeria monocytogenes were examined. High-performance liquid chromatography and liquid-chromatography-mass spectrometry were performed to identify N-acyl homoserine lactone (AHL) molecules secreted by monocultures and dual cultures grown in crab broth. Field emission scanning electron microscopy was performed to observe attachment and biofilm formation. P. aeruginosa and P. fluorescens inhibited biofilm formation by A. hydrophila on the crab surface, without affecting their own biofilm-forming abilities. Dual biofilms of S. Typhimurium, L. monocytogenes, or P. carotovorum did not affect A. hydrophila biofilm formation. Exoprotease, AHL, and AI-2 levels were significantly reduced in dual cultures of P. aeruginosa and P. fluorescens with A. hydrophila, supporting the relationship between quorum sensing and biofilm formation. Dual-species biofilms were studied in their natural environment and in the laboratory.


Assuntos
Aeromonas hydrophila/crescimento & desenvolvimento , Biofilmes/crescimento & desenvolvimento , Braquiúros/microbiologia , Exopeptidases/metabolismo , Microbiota/fisiologia , Percepção de Quorum/fisiologia , Alimentos Marinhos/microbiologia , Acil-Butirolactonas/metabolismo , Aeromonas hydrophila/enzimologia , Aeromonas hydrophila/fisiologia , Animais , Aderência Bacteriana/fisiologia , Técnicas de Cocultura
5.
J Neuroinflammation ; 13(1): 133, 2016 06 01.
Artigo em Inglês | MEDLINE | ID: mdl-27250711

RESUMO

BACKGROUND: Chronic infection with Theiler's murine encephalomyelitis virus (TMEV) in susceptible SJL/J mice induces an immune-mediated demyelinating disease and has extensively been used as a relevant infectious model for multiple sclerosis (MS). Infection of the host with many other viruses also leads to acute or chronic inflammatory diseases in the central nervous system (CNS). Levels of viral load in the host often play a critical role in the pathogenesis of virus-induced diseases. Thus, the inhibition of viral replication in the host against a broad spectrum of similar viruses is critically important for preventing the viral pathogenicity. METHODS: P2/P3-expressing transgenic (B6 X SJL)F1 founders were generated and bred onto the C57BL/6 and SJL/J backgrounds. Differences in the development of demyelinating disease were compared. Viral persistence, cytokine production, and immune responses in the CNS of infected control and P2/P3-Tg mice were analyzed after infection using quantitative PCR, ELISA, and flow cytometry. Various cell types from the control and P2/P3-Tg mice, as well as cells transfected in vitro with the P2 and/or P3 regions, were also analyzed for viral replication and innate cytokine production. RESULTS: P2/P3-transgenic (P2/P3-Tg) mice carrying the viral non-structural protein genes displayed significantly reduced virus-specific T cell responses in the CNS against both the structural and non-structural proteins. Consequently, viral loads in the CNS were greater in the Tg mice during the chronic infection. However, P2/P3-Tg SJL mice exhibited reduced disease incidence and less severe clinical symptoms than did their non-transgenic littermates. Interestingly, P2/P3-Tg mice showed low viral loads in the CNS at a very early period after infection (1-3 days) with TMEV and related EMCV but not unrelated VSV. Cells from P2/P3-Tg mice and cells transfected with the P2 and/or P3 regions in vitro yielded also lower viral replication but higher IFN-α/ß production. CONCLUSIONS: This study demonstrates that the expression of viral non-structural genes in mice inhibits initial viral replication and suppresses sustaining pathogenic anti-viral immune responses to broad viral determinants. It appears that the elevation of innate immune cytokines produced in the cells expressing the non-structural viral genes upon viral infection is responsible for the inhibitions. The inhibition is partially virus-specific as it is more efficient for a related virus compared to an unrelated virus, suggesting a role for the similarity in the viral genome structures. Therefore, the expression of viral non-structural genes may serve as a useful new method to prevent a broadly virus-specific pathogenesis in the hosts.


Assuntos
Doenças Desmielinizantes/genética , Doenças Desmielinizantes/metabolismo , Regulação Viral da Expressão Gênica , Theilovirus/genética , Theilovirus/metabolismo , Replicação Viral/fisiologia , Animais , Células Cultivadas , Feminino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Linfócitos T/fisiologia
6.
J Virol ; 89(9): 4918-31, 2015 May.
Artigo em Inglês | MEDLINE | ID: mdl-25694600

RESUMO

UNLABELLED: The ORF45 protein of Kaposi's sarcoma-associated herpesvirus (KSHV) is a gammaherpesvirus-specific immediate-early tegument protein. Our previous studies have revealed its crucial roles in both early and late stages of KSHV infection. In this study, we surveyed the interactome of ORF45 using a panel of monoclonal antibodies. In addition to the previously identified extracellular regulated kinase (ERK) and p90 ribosomal S6 kinase (RSK) proteins, we found several other copurified proteins, including prominent ones of ∼38 kDa and ∼130 kDa. Mass spectrometry revealed that the 38-kDa protein is viral ORF33 and the 130-kDa protein is cellular USP7 (ubiquitin-specific protease 7). We mapped the ORF33-binding domain to the highly conserved carboxyl-terminal 19 amino acids (aa) of ORF45 and the USP7-binding domain to the reported consensus motif in the central region of ORF45. Using immunofluorescence staining, we observed colocalization of ORF45 with ORF33 or USP7 both under transfected conditions and in KSHV-infected cells. Moreover, we noticed ORF45-dependent relocalization of a portion of ORF33/USP7 from the nucleus to the cytoplasm. We found that ORF45 caused an increase in ORF33 protein accumulation that was abolished if either the ORF33- or USP7-binding domain in ORF45 was deleted. Furthermore, deletion of the conserved carboxyl terminus of ORF45 in the KSHV genome drastically reduced the level of ORF33 protein in KSHV-infected cells and abolished production of progeny virions. Collectively, our results not only reveal new components of the ORF45 interactome, but also demonstrate that the interactions among these proteins are crucial for KSHV lytic replication. IMPORTANCE: Kaposi's sarcoma-associated herpesvirus (KSHV) is the causative agent of several human cancers. KSHV ORF45 is a multifunctional protein that is required for KSHV lytic replication, but the exact mechanisms by which ORF45 performs its critical functions are unclear. Our previous studies revealed that all ORF45 protein in cells exists in high-molecular-weight complexes. We therefore sought to characterize the interactome of ORF45 to provide insights into its roles during lytic replication. Using a panel of monoclonal antibodies, we surveyed the ORF45 interactome in KSHV-infected cells. We identified two new binding partners of ORF45: the viral protein ORF33 and cellular ubiquitin-specific protease 7 (USP7). We further demonstrate that the interaction between ORF45 and ORF33 is crucial for the efficient production of KSHV viral particles, suggesting that the targeted interference with this interaction may represent a novel strategy to inhibit KSHV lytic replication.


Assuntos
Proteínas do Capsídeo/metabolismo , Herpesvirus Humano 8/fisiologia , Proteínas Imediatamente Precoces/metabolismo , Mapeamento de Interação de Proteínas , Ubiquitina Tiolesterase/metabolismo , Replicação Viral , Proteínas do Capsídeo/química , Linhagem Celular , Humanos , Espectrometria de Massas , Peso Molecular , Ubiquitina Tiolesterase/química , Peptidase 7 Específica de Ubiquitina
7.
J Virol ; 89(1): 195-207, 2015 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-25320298

RESUMO

UNLABELLED: We have previously shown that ORF45, an immediate-early and tegument protein of Kaposi's sarcoma-associated herpesvirus (KSHV), causes sustained activation of p90 ribosomal S6 kinases (RSKs) and extracellular regulated kinase (ERK) (E. Kuang, Q. Tang, G. G. Maul, and F. Zhu, J Virol 82:1838-1850, 2008, http://dx.doi.org/10.1128/JVI.02119-07). We now have identified the critical region of ORF45 that is involved in RSK interaction and activation. Alanine scanning mutagenesis of this region revealed that a single F66A point mutation abolished binding of ORF45 to RSK or ERK and, consequently, its ability to activate the kinases. We introduced the F66A mutation into BAC16 (a bacterial artificial chromosome clone containing the entire infectious KSHV genome), producing BAC16-45F66A. In parallel, we also repaired the mutation and obtained a revertant, BAC16-45A66F. The reconstitution of these mutants in iSLK cells demonstrated that the ORF45-F66A mutant failed to cause sustained ERK and RSK activation during lytic reactivation, resulting in dramatic differences in the phosphoproteomic profile between the wild-type virus-infected cells and the mutant virus-infected cells. ORF45 mutation or deletion also was accompanied by a noticeable decreased in viral gene expression during lytic reactivation. Consequently, the ORF45-F66A mutant produced significantly fewer infectious progeny virions than the wild type or the revertant. These results suggest a critical role for ORF45-mediated RSK activation in KSHV lytic replication. IMPORTANCE: KSHV is the causative agent of three human malignancies. KSHV pathogenesis is intimately linked to its ability to modulate the host cell microenvironment and to facilitate efficient production of progeny viral particles. We previously described the mechanism by which the KSHV lytic protein ORF45 activates the cellular kinases ERK and RSK. We now have mapped the critical region of ORF45 responsible for binding and activation of ERK/RSK to a single residue, F66. We mutated this amino acid of ORF45 (F66A) and introduced the mutation into a newly developed bacterial artificial chromosome containing the KSHV genome (BAC16). This system has provided us with a useful tool to characterize the functions of ORF45-activated RSK upon KSHV lytic reactivation. We show that viral gene expression and virion production are significantly reduced by F66A mutation, indicating a critical role for ORF45-activated RSK during KSHV lytic replication.


Assuntos
Herpesvirus Humano 8/fisiologia , Interações Hospedeiro-Patógeno , Proteínas Imediatamente Precoces/metabolismo , Proteínas Quinases S6 Ribossômicas 90-kDa/metabolismo , Replicação Viral , Linhagem Celular , Análise Mutacional de DNA , Ativação Enzimática , Humanos , Proteínas Imediatamente Precoces/genética , Mutagênese Sítio-Dirigida , Proteínas Mutantes/genética , Proteínas Mutantes/metabolismo , Ligação Proteica , Mapeamento de Interação de Proteínas
8.
PLoS Pathog ; 10(1): e1003863, 2014 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-24453968

RESUMO

Nuclear domain 10 (ND10) components are restriction factors that inhibit herpesviral replication. Effector proteins of different herpesviruses can antagonize this restriction by a variety of strategies, including degradation or relocalization of ND10 proteins. We investigated the interplay of Kaposi's Sarcoma-Associated Herpesvirus (KSHV) infection and cellular defense by nuclear domain 10 (ND10) components. Knock-down experiments in primary human cells show that KSHV-infection is restricted by the ND10 components PML and Sp100, but not by ATRX. After KSHV infection, ATRX is efficiently depleted and Daxx is dispersed from ND10, indicating that these two ND10 components can be antagonized by KSHV. We then identified the ORF75 tegument protein of KSHV as the viral factor that induces the disappearance of ATRX and relocalization of Daxx. ORF75 belongs to a viral protein family (viral FGARATs) that has homologous proteins in all gamma-herpesviruses. Isolated expression of ORF75 in primary cells induces a relocalization of PML and dispersal of Sp100, indicating that this viral effector protein is able to influence multiple ND10 components. Moreover, by constructing a KSHV mutant harboring a stop codon at the beginning of ORF75, we could demonstrate that ORF75 is absolutely essential for viral replication and the initiation of viral immediate-early gene expression. Using recombinant viruses either carrying Flag- or YFP-tagged variants of ORF75, we could further corroborate the role of ORF75 in the antagonization of ND10-mediated intrinsic immunity, and show that it is independent of the PML antagonist vIRF3. Members of the viral FGARAT family target different ND10 components, suggesting that the ND10 targets of viral FGARAT proteins have diversified during evolution. We assume that overcoming ND10 intrinsic defense constitutes a critical event in the replication of all herpesviruses; on the other hand, restriction of herpesviral replication by ND10 components may also promote latency as the default outcome of infection.


Assuntos
Infecções por Herpesviridae/imunologia , Herpesvirus Humano 8/fisiologia , Imunidade Inata , Proteínas Nucleares/imunologia , Proteínas Estruturais Virais/imunologia , Replicação Viral/fisiologia , Proteínas Adaptadoras de Transdução de Sinal/genética , Proteínas Adaptadoras de Transdução de Sinal/imunologia , Células Cultivadas , Proteínas Correpressoras , Códon de Terminação/genética , Códon de Terminação/imunologia , DNA Helicases/genética , DNA Helicases/imunologia , Técnicas de Silenciamento de Genes , Infecções por Herpesviridae/genética , Infecções por Herpesviridae/patologia , Humanos , Masculino , Chaperonas Moleculares , Mutação , Proteínas Nucleares/genética , Proteínas Estruturais Virais/genética , Proteína Nuclear Ligada ao X
9.
Arch Virol ; 161(4): 779-88, 2016 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-26780772

RESUMO

Human norovirus (hNoV) infections cause acute gastroenteritis, accounting for millions of disease cases and more than 200,000 deaths annually. However, the lack of in vitro infection models and robust small-animal models has posed barriers to the development of virus-specific therapies and preventive vaccines. Promising recent progress in the development of a norovirus infection model is reviewed in this article, as well as attempts and efforts made since the discovery of hNoV more than 40 years ago. Because suitable experimental animal models for human norovirus are lacking, attractive alternatives are also discussed.


Assuntos
Infecções por Caliciviridae/virologia , Norovirus/fisiologia , Animais , Infecções por Caliciviridae/patologia , Técnicas de Cultura de Células , Linhagem Celular , Humanos
10.
J Gen Virol ; 96(12): 3635-3645, 2015 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-26467721

RESUMO

Kaposi's sarcoma-associated herpesvirus (KSHV) belongs to the human gammaherpesvirus subfamily and is associated with malignancies of endothelial origin (Kaposi's sarcoma, KS) and B-cell origin [primary effusion lymphoma (PEL) and multicentric Castleman's disease (MCD)]. Viral lytic replication is known to be required for KS and MCD. As KSHV-related tumours mostly develop in human subjects when the immune system is compromised by immunosuppressive regimen, human immunodeficiency virus infection or some genetic deficiencies, KSHV-specific immune responses are believed to be important in the control of KSHV replication. However, analysis of the roles of immune cells in viral pathogenesis has been difficult due to the lack of an adequate animal model. Recently, congenital OX40 deficiency, as determined by genome-wide exome sequencing, was shown to be associated with aggressive childhood KS in a patient, suggesting that disrupted OX40­OX40L interactions might be implicated in disease development. Here, we report that interaction of recombinant OX40 protein with OX40L expressed on endothelial cells severely impaired KSHV lytic replication. Furthermore, 4-1BB­4-1BBL interactions were also capable of efficiently inhibiting viral replication in B-cells and endothelial cells. To the best of our knowledge, this is the first direct evidence that ligation of tumour necrosis factor superfamily members and their cognate receptors is important for the control of viral lytic replication. These data are likely to pave the way for the development of KSHV-specific therapies for KS and MCD, in which viral lytic replication is a disease-determining factor.


Assuntos
Ligante 4-1BB/metabolismo , Linfócitos B/virologia , Células Endoteliais/virologia , Herpesvirus Humano 8/fisiologia , Receptores OX40/metabolismo , Replicação Viral/fisiologia , Ligante 4-1BB/genética , Linfócitos B/fisiologia , Linfócitos T CD4-Positivos/fisiologia , Linfócitos T CD4-Positivos/virologia , Regulação para Baixo , Células Endoteliais/fisiologia , Regulação da Expressão Gênica/fisiologia , Humanos , Tonsila Palatina/citologia , Receptores OX40/genética , Transdução de Sinais/fisiologia
11.
J Virol ; 86(24): 13717-28, 2012 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-23055563

RESUMO

Theiler's virus-induced demyelinating disease has been extensively investigated as a model for persistent viral infection and multiple sclerosis (MS). However, the role of CD8(+) T cells in the development of disease remains unclear. To assess the role of virus-specific CD8(+) T cells in the pathogenesis of demyelinating disease, a single amino acid substitution was introduced into the predominant viral epitope (VP3 from residues 159 to 166 [VP3(159-166)]) and/or a subdominant viral epitope (VP3(173-181)) of susceptible SJL/J mice by site-directed mutagenesis. The resulting variant viruses (N160V, P179A, and N160V/P179A) failed to induce CD8(+) T cell responses to the respective epitopes. Surprisingly, mice infected with N160V or N160V/P179A virus, which lacks CD8(+) T cells against VP3(159-166), did not develop demyelinating disease, in contrast to wild-type virus or P179A virus lacking VP3(173-181)-specific CD8(+) T cells. Our findings clearly show that the presence of VP3(159-166)-specific CD8(+) T cells, rather than viral persistence itself, is strongly correlated with disease development. VP3(173-181)-specific CD8(+) T cells in the central nervous system (CNS) of these virus-infected mice expressed higher levels of transforming growth factor ß, forkhead box P3, interleukin-22 (IL-22), and IL-17 mRNA but caused minimal cytotoxicity compared to that caused by VP3(159-166)-specific CD8(+) T cells. VP3(159-166)-specific CD8(+) T cells exhibited high functional avidity for gamma interferon production, whereas VP3(173-181)-specific CD8(+) T cells showed low avidity. To our knowledge, this is the first report indicating that the induction of the IL-17-producing CD8(+) T cell type is largely epitope specific and that this specificity apparently plays a differential role in the pathogenicity of virus-induced demyelinating disease. These results strongly advocate for the careful consideration of CD8(+) T cell-mediated intervention of virus-induced inflammatory diseases.


Assuntos
Linfócitos T CD8-Positivos/imunologia , Epitopos/imunologia , Esclerose Múltipla/imunologia , Viroses/imunologia , Animais , Linhagem Celular , Cricetinae , Modelos Animais de Doenças , Camundongos , Esclerose Múltipla/virologia , Reação em Cadeia da Polimerase em Tempo Real , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Replicação Viral
12.
J Virol ; 86(18): 9708-20, 2012 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-22740391

RESUMO

Efficient genetic modification of herpesviruses such as Kaposi's sarcoma-associated herpesvirus (KSHV) has come to rely on bacterial artificial chromosome (BAC) technology. In order to facilitate this approach, we generated a new KSHV BAC clone, called BAC16, derived from the rKSHV.219 virus, which stems from KSHV and Epstein-Barr virus-coinfected JSC1 primary effusion lymphoma (PEL) cells. Restriction enzyme and complete sequencing data demonstrate that the KSHV of JSC1 PEL cells showed a minimal level of sequence variation across the entire viral genome compared to the complete genomic sequence of other KSHV strains. BAC16 not only stably propagated in both Escherichia coli and mammalian cells without apparent genetic rearrangements, but also was capable of robustly producing infectious virions (∼5 × 10(7)/ml). We also demonstrated the utility of BAC16 by generating deletion mutants of either the K3 or K5 genes, whose products are E3 ligases of the membrane-associated RING-CH (MARCH) family. While previous studies have shown that individual expression of either K3 or K5 results in efficient downregulation of the surface expression of major histocompatibility complex class I (MHC-I) molecules, we found that K5, but not K3, was the primary factor critical for the downregulation of MHC-I surface expression during KSHV lytic reactivation or following de novo infection. The data presented here demonstrate the utility of BAC16 for the generation and characterization of KSHV knockout and mutant recombinants and further emphasize the importance of functional analysis of viral genes in the context of the KSHV genome besides the study of individual gene expression.


Assuntos
Cromossomos Artificiais Bacterianos/genética , Herpesvirus Humano 8/genética , Animais , Sequência de Bases , Linhagem Celular Tumoral , Chlorocebus aethiops , Clonagem Molecular , DNA Viral/genética , Escherichia coli/genética , Deleção de Genes , Regulação Viral da Expressão Gênica , Genoma Viral , Herpesvirus Humano 8/patogenicidade , Herpesvirus Humano 8/fisiologia , Antígenos de Histocompatibilidade Classe I/metabolismo , Interações Hospedeiro-Patógeno , Humanos , Proteínas Imediatamente Precoces/genética , Proteínas Imediatamente Precoces/fisiologia , Linfoma de Efusão Primária/virologia , Dados de Sequência Molecular , Mutação , Plasmídeos/genética , Células Vero , Proteínas Virais/genética , Proteínas Virais/fisiologia
13.
PLoS Pathog ; 7(10): e1002300, 2011 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-22022268

RESUMO

During the lytic phase of infection, the gamma herpesvirus Kaposi's Sarcoma-Associated Herpesvirus (KSHV) expresses a highly abundant, 1.1 kb nuclear noncoding RNA of unknown function. We observe that this polyadenylated nuclear (PAN) RNA avidly binds host poly(A)-binding protein C1 (PABPC1), which normally functions in the cytoplasm to bind the poly(A) tails of mRNAs, regulating mRNA stability and translation efficiency. During the lytic phase of KSHV infection, PABPC1 is re-localized to the nucleus as a consequence of expression of the viral shutoff exonuclease (SOX) protein; SOX also mediates the host shutoff effect in which host mRNAs are downregulated while viral mRNAs are selectively expressed. We show that whereas PAN RNA is not required for the host shutoff effect or for PABPC1 re-localization, SOX strongly upregulates the levels of PAN RNA in transient transfection experiments. This upregulation is destroyed by the same SOX mutation that ablates the host shutoff effect and PABPC1 nuclear re-localization or by removal of the poly(A) tail of PAN. In cells induced into the KSHV lytic phase, depletion of PAN RNA using RNase H-targeting antisense oligonucleotides reveals that it is necessary for the production of late viral proteins from mRNAs that are themselves polyadenylated. Our results add to the repertoire of functions ascribed to long noncoding RNAs and suggest a mechanism of action for nuclear noncoding RNAs in gamma herpesvirus infection.


Assuntos
Regulação Viral da Expressão Gênica , Herpesvirus Humano 8/genética , Herpesvirus Humano 8/metabolismo , Proteína I de Ligação a Poli(A)/metabolismo , RNA não Traduzido/metabolismo , RNA Viral/metabolismo , Linhagem Celular , Núcleo Celular/metabolismo , Células HEK293 , Humanos , Poli A/metabolismo , Interferência de RNA , RNA Mensageiro/metabolismo , RNA Interferente Pequeno , RNA Viral/genética , Ribonuclease H/metabolismo , Fatores de Transcrição SOX , Replicação Viral
14.
J Microbiol Biotechnol ; 33(12): 1587-1295, 2023 Dec 28.
Artigo em Inglês | MEDLINE | ID: mdl-37915256

RESUMO

Since its first report in 2019, severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) has posed a grave threat to public health. Virus-specific countermeasures, such as vaccines and therapeutics, have been developed and have contributed to the control of the viral pandemic, which has become endemic. Nonetheless, new variants continue to emerge and could cause a new pandemic. Consequently, it is important to comprehensively understand viral evolution and the roles of mutations in viral infectivity and transmission. SARS-CoV-2 beta variant encode mutations (D614G, N501Y, E484K, and K417N) in the spike which are frequently found in other variants as well. While their individual role in viral infectivity has been elucidated against various therapeutic antibodies, it still remains unclear whether those mutations may act additively or synergistically when combined. Here, we report that N501Y mutation shows differential effect on two therapeutic antibodies tested. Interestingly, the relative importance of E484K and K417N mutations in antibody evasion varies depending on the antibody type. Collectively, these findings suggest that continuous efforts to develop effective antibody therapeutics and combinatorial treatment with multiple antibodies are more rational and effective forms of treatment.


Assuntos
Anticorpos , COVID-19 , SARS-CoV-2 , Glicoproteína da Espícula de Coronavírus , Humanos , Anticorpos/metabolismo , Anticorpos Neutralizantes , COVID-19/imunologia , Mutação , Ligação Proteica , SARS-CoV-2/genética , SARS-CoV-2/imunologia , Glicoproteína da Espícula de Coronavírus/genética
15.
J Biol Chem ; 286(48): 41171-41182, 2011 Dec 02.
Artigo em Inglês | MEDLINE | ID: mdl-21994950

RESUMO

Open reading frame 45 (ORF45) of Kaposi sarcoma-associated herpesvirus (KSHV) causes sustained activation of p90 ribosomal S6 kinase (RSK), which is crucial for KSHV lytic replication, but the exact functional roles remain to be determined. To characterize the biological consequence of persistent RSK activation by ORF45, we screened known cellular substrates of RSK. We found that ORF45 induced phosphorylation of eukaryotic translation initiation factor 4B (eIF4B), increased its assembly into translation initiation complex, and subsequently facilitated protein translation. The ORF45/RSK-mediated eIF4B phosphorylation was distinguishable from that caused by the canonical AKT/mammalian target of rapamycin/ribosomal S6 kinase and MEK/ERK/RSK pathways because it was resistant to both rapamycin (an mammalian target of rapamycin inhibitor) and U1026 (an MEK inhibitor). The rapamycin and U1026 doubly insensitive eIF4B phosphorylation was induced during KSHV reactivation but was abolished if either ORF45 or RSK1/2 were ablated by siRNA, a pattern that is correlated with reduced lytic gene expression as we observed previously. Ectopic expression of eIF4B but not its phosphorylation-deficient mutant form increased KSHV lytic gene expression and production of progeny viruses. Together, these results indicated that ORF45/RSK axis-induced eIF4B phosphorylation is involved in translational regulation and is required for optimal KSHV lytic replication.


Assuntos
Fatores de Iniciação em Eucariotos/metabolismo , Herpesvirus Humano 8/fisiologia , Proteínas Imediatamente Precoces/metabolismo , Sistema de Sinalização das MAP Quinases , Biossíntese de Proteínas , Proteínas Quinases S6 Ribossômicas 90-kDa/metabolismo , Replicação Viral/fisiologia , Linhagem Celular Tumoral , Fatores de Iniciação em Eucariotos/genética , Regulação Viral da Expressão Gênica/efeitos dos fármacos , Regulação Viral da Expressão Gênica/fisiologia , Células HEK293 , Humanos , Proteínas Imediatamente Precoces/genética , Imunossupressores/farmacologia , Mutação , Fosforilação/efeitos dos fármacos , Fosforilação/genética , Proteínas Proto-Oncogênicas c-akt/antagonistas & inibidores , Proteínas Proto-Oncogênicas c-akt/genética , Proteínas Proto-Oncogênicas c-akt/metabolismo , Proteínas Quinases S6 Ribossômicas 90-kDa/genética , Sirolimo/farmacologia , Serina-Treonina Quinases TOR/antagonistas & inibidores , Serina-Treonina Quinases TOR/genética , Serina-Treonina Quinases TOR/metabolismo , Replicação Viral/efeitos dos fármacos
16.
J Virol ; 85(19): 9767-77, 2011 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-21795352

RESUMO

Kaposi's sarcoma-associated herpesvirus (KSHV) displays strong lymphotropism in vivo, but paradoxically, established B cell lines have largely been refractory to infection by soluble KSHV virions. Here we show that this block can be overcome by exposure to cell-associated virus. Doxycycline-inducible recombinant KSHV.219 (rKSHV.219)-harboring SLK (iSLK.219) cells were employed as KSHV donors. Cocultivation of lymphoid cell lines with reactivated iSLK.219 cells resulted in readily demonstrable viral entry into each cell line; similar observations were made in primary tonsillar B cell cultures. Moreover, infected lymphoid cells were able to outgrow upon puromycin selection, indicating development of persistent infection. Infected BJAB cells display signatures of latent infection, including classical latency-associated transcripts, a punctate pattern of LANA expression, and episomal maintenance of the KSHV genome. However, when lytically activated by various chemical stimuli, infected BJAB cells were able to produce only low levels of infectious virions. These data demonstrate that (i) cell-associated viruses can bypass viral entry blocks in most lymphoid cell lines, (ii) the determinants of cell-associated virus entry differ from those of soluble virion infection, and (iii) immortalized lymphoblastoid lines have partial postentry blocks to efficient lytic reactivation.


Assuntos
Linfócitos B/virologia , Herpesvirus Humano 8/patogenicidade , Internalização do Vírus , Linhagem Celular , Humanos , Ativação Viral , Latência Viral , Replicação Viral
17.
J Microbiol ; 60(3): 235-237, 2022 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-35235176

RESUMO

Unprecedented. This is the closest and most appropriate word to describe the COVID-19 pandemic, which the world has been experiencing with pain and fear. The first case of pneumonia-like symptoms of unknown etiology appeared presumably in November 2019, with the subsequent official report to the WHO by the Chinese authorities on December 31, 2019. China's first confirmed death from the virus occurred on January 11, 2020, when a 61-year-old male resident of Hubei, the capital of Wuhan Province, died. Within a month, the COVID-19 death toll surpassed 1,000 (February 10, 2020). Accordingly, just 30 days after the initial report, the coronavirus outbreak was called a "public health emergency of international concern" by the WHO, the organization's highest alert level. Unfortunately, the WHO soon declared the COVID-19 outbreak a pandemic (March 11, 2020). Within a year of viral emergence, and by December 2020, more than 80 million confirmed cases had been reported worldwide. Infections increased exponentially over the following year. As of February 11, 2022, over 400 million cases have been reported, with nearly 6 million deaths, an unprecedented rate of spread across borders.


Assuntos
COVID-19 , Pandemias , COVID-19/epidemiologia , Surtos de Doenças , Humanos , Masculino , Pessoa de Meia-Idade , Saúde Pública , SARS-CoV-2
18.
J Microbiol ; 60(3): 276-289, 2022 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-35157219

RESUMO

SARS-CoV-2, the causative agent of COVID-19, first emerged in 2019. Antibody responses against SARS-CoV-2 have been given a lot of attention. However, the armamentarium of humoral and T cells may have differing roles in different viral infections. Though the exact role of T cells in COVID-19 remains to be elucidated, prior experience with human coronavirus has revealed an essential role of T cells in the outcomes of viral infections. Moreover, an increasing body of evidence suggests that T cells might be effective against SARS-CoV-2. This review summarizes the role of T cells in mouse CoV, human pathogenic respiratory CoV in general and SARS-CoV-2 in specific.


Assuntos
COVID-19 , SARS-CoV-2 , Animais , Anticorpos Antivirais , Humanos , Camundongos , Linfócitos T
19.
Viruses ; 14(3)2022 03 04.
Artigo em Inglês | MEDLINE | ID: mdl-35336937

RESUMO

In the past 20 years, coronaviruses (CoVs), including SARS-CoV-1, MERS-CoV, and SARS-CoV-2, have rapidly evolved and emerged in the human population. The innate immune system is the first line of defense against invading pathogens. Multiple host cellular receptors can trigger the innate immune system to eliminate invading pathogens. However, these CoVs have acquired strategies to evade innate immune responses by avoiding recognition by host sensors, leading to impaired interferon (IFN) production and antagonizing of the IFN signaling pathways. In contrast, the dysregulated induction of inflammasomes, leading to uncontrolled production of IL-1 family cytokines (IL-1ß and IL-18) and pyroptosis, has been associated with COVID-19 pathogenesis. This review summarizes innate immune evasion strategies employed by SARS-CoV-1 and MERS-CoV in brief and SARS-CoV-2 in more detail. In addition, we outline potential mechanisms of inflammasome activation and evasion and their impact on disease prognosis.


Assuntos
COVID-19 , SARS-CoV-2 , Citocinas/metabolismo , Humanos , Evasão da Resposta Imune , Imunidade Inata
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA