Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 8 de 8
Filtrar
1.
Nucleic Acids Res ; 49(8): 4456-4471, 2021 05 07.
Artigo em Inglês | MEDLINE | ID: mdl-33823555

RESUMO

Kaposi's sarcoma-associated herpesvirus (KSHV) expresses miRNAs during latency. However, regulation of viral miRNAs remains largely unknown. Our prior studies demonstrated that MCPIP1 regulates KSHV miRNA biogenesis by degrading most KSHV pre-miRNAs through its RNase activity. Some viral pre-miRNAs are partially resistant to degradation by MCPIP1. Here, we further characterized MCPIP1 substrate specificity and its antiviral potential against KSHV infection. In vitro cleavage assays and binding assays showed that MCPIP1 cleavage efficiency is related to binding affinity. Motif-based sequence analysis identified that KSHV pre-miRNAs that are well degraded by MCPIP1 have a 5-base motif (M5 base motif) within their terminal loops and this motif region consists of multiple pyrimidine-purine-pyrimidine (YRY) motifs. We further demonstrated that mutation of this M5 base motif within terminal loop of pre-miRNAs inhibited MCPIP1-mediated RNA degradation. We also revealed that MCPIP1 has an antiviral effect against KSHV infection. MCPIP1 can reduce the expression of Dicer, which in turn restricts KSHV infection. Conclusively, our findings demonstrated that MCPIP1 inhibited KSHV infection and suppressed viral miRNA biogenesis by directly degrading KSHV pre-miRNAs and altering the expression of miRNA biogenesis factors.


Assuntos
Infecções por Herpesviridae/metabolismo , Herpesvirus Humano 8/metabolismo , MicroRNAs/metabolismo , RNA Viral/metabolismo , Ribonucleases/metabolismo , Fatores de Transcrição/metabolismo , Linhagem Celular , RNA Helicases DEAD-box/genética , RNA Helicases DEAD-box/metabolismo , Técnicas de Silenciamento de Genes , Infecções por Herpesviridae/genética , Infecções por Herpesviridae/virologia , Herpesvirus Humano 8/genética , Humanos , MicroRNAs/genética , Motivos de Nucleotídeos , Ligação Proteica , Estabilidade de RNA/genética , RNA Viral/genética , Ribonuclease III/genética , Ribonuclease III/metabolismo
2.
Semin Cell Dev Biol ; 111: 135-147, 2021 03.
Artigo em Inglês | MEDLINE | ID: mdl-32631785

RESUMO

Oncogenic DNA viruses establish lifelong infections in humans, and they cause cancers, often in immunocompromised patients, despite anti-viral immune surveillance targeted against viral antigens. High-throughput sequencing techniques allowed the field to identify novel viral non-coding RNAs (ncRNAs). ncRNAs are ideal factors for DNA viruses to exploit; they are non-immunogenic to T cells, thus viral ncRNAs can manipulate host cells without evoking adaptive immune responses. Viral ncRNAs may still trigger the host innate immune response, but many viruses encode decoys/inhibitors to counter-act and evade recognition. In addition, ncRNAs can be secreted to the extracellular space and influence adjacent cells to create a pro-viral microenvironment. In this review, we present recent progress in understanding interactions between oncoviruses and ncRNAs including small and long ncRNAs, microRNAs, and recently identified viral circular RNAs. In addition, potential clinical applications for ncRNA will be discussed. Extracellular ncRNAs are suggested to be diagnostic and prognostic biomarkers and, with the realization of the importance of viral ncRNAs in tumorigenesis, approaches to target critical viral ncRNAs are emerging. Further understanding of viral utilization of ncRNAs will advance anti-viral therapeutics beyond conventional medication and vaccination.


Assuntos
Evasão da Resposta Imune/genética , MicroRNAs/genética , Neoplasias/genética , RNA Circular/genética , RNA Longo não Codificante/genética , RNA Viral/genética , Viroses/genética , Alphapapillomavirus/genética , Alphapapillomavirus/crescimento & desenvolvimento , Alphapapillomavirus/patogenicidade , Antivirais/uso terapêutico , Carcinogênese/genética , Carcinogênese/imunologia , Carcinogênese/patologia , Regulação da Expressão Gênica , Herpesvirus Humano 4/genética , Herpesvirus Humano 4/crescimento & desenvolvimento , Herpesvirus Humano 4/patogenicidade , Herpesvirus Humano 8/genética , Herpesvirus Humano 8/crescimento & desenvolvimento , Herpesvirus Humano 8/patogenicidade , Vírus Linfotrópico T Tipo 1 Humano/genética , Vírus Linfotrópico T Tipo 1 Humano/crescimento & desenvolvimento , Vírus Linfotrópico T Tipo 1 Humano/patogenicidade , Humanos , Imunidade Inata , MicroRNAs/antagonistas & inibidores , MicroRNAs/imunologia , Neoplasias/imunologia , Neoplasias/terapia , Neoplasias/virologia , Oligonucleotídeos Antissenso/uso terapêutico , RNA Circular/imunologia , RNA Longo não Codificante/imunologia , RNA Viral/imunologia , Transdução de Sinais , Viroses/imunologia , Viroses/terapia , Viroses/virologia
3.
J Virol ; 92(17)2018 09 01.
Artigo em Inglês | MEDLINE | ID: mdl-29899098

RESUMO

Following productive infection, bovine herpesvirus 1 (BoHV-1) establishes latency in sensory neurons. As in other alphaherpesviruses, expression of BoHV-1 immediate early (IE) genes is regulated by an enhancer complex containing the viral IE activator VP16, the cellular transcription factor Oct-1, and transcriptional coactivator HCF-1, which is assembled on an IE enhancer core element (TAATGARAT). Expression of the IE transcription unit that encodes the viral IE activators bICP0 and bICP4 may also be induced by the activated glucocorticoid receptor (GR) via two glucocorticoid response elements (GREs) located upstream of the enhancer core. Strikingly, lytic infection and reactivation from latency are consistently enhanced by glucocorticoid treatment in vivo As the coactivator HCF-1 is essential for IE gene expression of alphaherpesviruses and recruited by multiple transcription factors, we tested whether HCF-1 is required for glucocorticoid-induced IE gene expression. Depletion of HCF-1 reduced GR-mediated activation of the IE promoter in mouse neuroblastoma cells (Neuro-2A). More importantly, HCF-1-mediated GR activation of the promoter was dependent on the presence of GRE sites but independent of the TAATGARAT enhancer core element. HCF-1 was also recruited to the GRE region of a promoter lacking the enhancer core, consistent with a direct role of the coactivator in mediating GR-induced transcription. Similarly, during productive lytic infection, HCF-1 and GR occupied the IE region containing the GREs. These studies indicate HCF-1 is critical for GR activation of the viral IE genes and suggests that glucocorticoid induction of viral reactivation proceeds via an HCF-1-GR mechanism in the absence of the viral IE activator VP16.IMPORTANCE BoHV-1 transcription is rapidly activated during stress-induced reactivation from latency. The immediate early transcription unit 1 (IEtu1) promoter is regulated by the GR via two GREs. The IEtu1 promoter regulates expression of two viral transcriptional regulatory proteins, infected cell proteins 0 and 4 (bICP0 and bICP4), and thus must be stimulated during reactivation. This study demonstrates that activation of the IEtu1 promoter by the synthetic corticosteroid dexamethasone requires HCF-1. Interestingly, the GRE sites, but not the IE enhancer core element (TAATGARAT), were required for HCF-1-mediated GR promoter activation. The GR and HCF-1 were recruited to the IEtu1 promoter in transfected and infected cells. Collectively, these studies indicate that HCF-1 is critical for GR activation of the viral IE genes and suggest that an HCF-1-GR complex can stimulate the IEtu1 promoter in the absence of the viral IE activator VP16.


Assuntos
Regulação Viral da Expressão Gênica , Genes Precoces , Glucocorticoides/metabolismo , Herpesvirus Bovino 1/fisiologia , Fator C1 de Célula Hospedeira/metabolismo , Receptores de Glucocorticoides/metabolismo , Transcrição Gênica , Animais , Linhagem Celular , Técnicas de Silenciamento de Genes , Fator C1 de Célula Hospedeira/genética , Camundongos , Neurônios/virologia
4.
Virus Res ; 222: 106-112, 2016 08 15.
Artigo em Inglês | MEDLINE | ID: mdl-27297663

RESUMO

Serum and glucocorticoid-regulated protein kinases (SGK) are serine/threonine protein kinases that contain a catalytic domain resembling other protein kinases: AKT/protein kinase B, protein kinase A, and protein kinase C-Zeta for example. Unlike these constitutively expressed protein kinases, SGK1 RNA and protein levels are increased by growth factors and corticosteroids. Stress can directly stimulate SGK1 levels as well as stimulate bovine herpesvirus 1 (BoHV-1) and herpes simplex virus 1 (HSV-1) productive infection and reactivation from latency suggesting SGK1 can stimulate productive infection. For the first time, we provide evidence that a specific SGK inhibitor (GSK650394) significantly reduced BoHV-1 and HSV-1 replication in cultured cells. Proteins encoded by the three BoHV-1 immediate early genes (bICP0, bICP4, and bICP22) and two late proteins (VP16 and gE) were consistently reduced by GSK650394 during early stages of productive infection. In summary, these studies suggest SGK may stimulate viral replication following stressful stimuli.


Assuntos
Infecções por Herpesviridae/metabolismo , Infecções por Herpesviridae/virologia , Herpesvirus Bovino 1/fisiologia , Herpesvirus Humano 1/fisiologia , Proteínas Imediatamente Precoces/metabolismo , Proteínas Serina-Treonina Quinases/metabolismo , Replicação Viral , Animais , Benzoatos/farmacologia , Compostos Bicíclicos Heterocíclicos com Pontes/farmacologia , Bovinos , Linhagem Celular , Chlorocebus aethiops , Relação Dose-Resposta a Droga , Regulação Viral da Expressão Gênica/efeitos dos fármacos , Glucocorticoides/farmacologia , Herpesvirus Bovino 1/efeitos dos fármacos , Herpesvirus Humano 1/efeitos dos fármacos , Humanos , Proteínas Imediatamente Precoces/antagonistas & inibidores , Camundongos , Regiões Promotoras Genéticas , Biossíntese de Proteínas/efeitos dos fármacos , Proteínas Serina-Treonina Quinases/antagonistas & inibidores , Células Vero , Replicação Viral/efeitos dos fármacos
5.
Virology ; 484: 377-385, 2015 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-26226582

RESUMO

The primary site for life-long latency of bovine herpesvirus 1 (BHV-1) is sensory neurons. The synthetic corticosteroid dexamethasone consistently induces reactivation from latency; however the mechanism by which corticosteroids mediate reactivation is unclear. In this study, we demonstrate for the first time that dexamethasone stimulates productive infection, in part, because the BHV-1 genome contains more than 100 potential glucocorticoid receptor (GR) response elements (GREs). Immediate early transcription unit 1 (IEtu1) promoter activity, but not IEtu2 or VP16 promoter activity, was stimulated by dexamethasone. Two near perfect consensus GREs located within the IEtu1 promoter were necessary for dexamethasone-mediated stimulation. Electrophoretic mobility shift assays and chromatin immunoprecipitation studies demonstrated that the GR interacts with IEtu1 promoter sequences containing the GREs. Although we hypothesize that DEX-mediated stimulation of IEtu1 promoter activity is important during productive infection and perhaps reactivation from latency, stress likely has pleiotropic effects on virus-infected cells.


Assuntos
Corticosteroides/metabolismo , Dexametasona/metabolismo , Herpesvirus Bovino 1/efeitos dos fármacos , Receptores de Glucocorticoides/metabolismo , Elementos de Resposta , Transcrição Gênica/efeitos dos fármacos , Ativação Viral/efeitos dos fármacos , Animais , Linhagem Celular , Imunoprecipitação da Cromatina , Ensaio de Desvio de Mobilidade Eletroforética , Herpesvirus Bovino 1/fisiologia , Regiões Promotoras Genéticas , Ligação Proteica
6.
J Neurovirol ; 21(5): 585-91, 2015 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-25860382

RESUMO

Bovine herpesvirus 1 (BHV-1) establishes latency in sensory neurons. The synthetic corticosteroid dexamethasone consistently induces reactivation from latency. Within 90 min after latently infected calves are treated with dexamethasone, two BHV-1 regulatory proteins, BHV-1-infected cell protein 0 (bICP0) and viral protein 16 (VP16), are expressed in the same neuron. In this study, we demonstrate that VP16 and bICP0 can be detected at 22 and 33 min after dexamethasone (DEX) treatment of latently infected calves. However, we were unable to discern whether VP16 or bICP0 was expressed at early times after reactivation. VP16+ neurons consistently express the glucocorticoid receptor suggesting corticosteroid-mediated activation of its receptor rapidly stimulates reactivation from latency.


Assuntos
Proteína Vmw65 do Vírus do Herpes Simples/biossíntese , Infecções por Herpesviridae/virologia , Herpesvirus Bovino 1/fisiologia , Transativadores/biossíntese , Ubiquitina-Proteína Ligases/biossíntese , Ativação Viral/fisiologia , Latência Viral/fisiologia , Animais , Apoptose/fisiologia , Apoptose/efeitos da radiação , Bovinos , Doenças dos Bovinos/virologia , Dexametasona/farmacologia , Glucocorticoides/farmacologia , Imuno-Histoquímica , Marcação In Situ das Extremidades Cortadas , Células Receptoras Sensoriais/virologia , Gânglio Trigeminal/virologia , Ativação Viral/efeitos dos fármacos , Latência Viral/efeitos dos fármacos
7.
J Virol ; 87(20): 11214-22, 2013 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-23926348

RESUMO

Bovine herpesvirus 1 (BHV-1) establishes a lifelong latent infection in sensory neurons following acute infection. Increased corticosteroid levels, due to stress, increases the incidence of reactivation from latency. Within minutes, corticosteroids activate the glucocorticoid receptor and transcription of promoters containing a glucocorticoid receptor element. A single intravenous injection of the synthetic corticosteroid dexamethasone consistently induces reactivation from latency in calves. Lytic cycle viral gene expression is detected within 6 h after dexamethasone treatment of calves latently infected with BHV-1. Cellular transcription factors are induced by dexamethasone in trigeminal ganglionic neurons within 1.5 h after dexamethasone treatment, suggesting they promote viral gene expression during the early phases of reactivation from latency, which we operationally defined as the escape from latency. In this study, immunohistochemistry was utilized to examine viral protein expression during the escape from latency. Within 1.5 h after dexamethasone treatment, bICP0 and a late protein (VP16) were consistently detected in a subset of trigeminal ganglionic neurons. Most neurons expressing bICP0 also expressed VP16. Additional studies revealed that neurons expressing the glucocorticoid receptor also expressed bICP0 or VP16 at 1.5 h after dexamethasone treatment. Two other late proteins, glycoprotein C and D, were not detected until 6 h after dexamethasone treatment and were detected in only a few neurons. These studies provide evidence that VP16 and the promiscuous viral trans-activator (bICP0) are expressed during the escape from latency, suggesting they promote the production of infectious virus in a small subset of latently infected neurons.


Assuntos
Cistos Glanglionares/virologia , Proteína Vmw65 do Vírus do Herpes Simples/metabolismo , Herpesvirus Bovino 1/fisiologia , Neurônios/virologia , Receptores de Glucocorticoides/biossíntese , Transativadores/metabolismo , Ubiquitina-Proteína Ligases/metabolismo , Ativação Viral , Animais , Bovinos , Dexametasona/administração & dosagem , Perfilação da Expressão Gênica , Imuno-Histoquímica , Fatores Imunológicos/administração & dosagem , Fatores de Tempo
8.
Plant Cell ; 25(3): 985-98, 2013 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-23512853

RESUMO

Small RNAs (sRNAs; ∼20 to 30 nucleotides in length) play important roles in gene regulation as well as in defense responses against transposons and viruses in eukaryotes. Their biogenesis and modes of action have attracted great attention in recent years. However, many aspects of sRNA function, such as the mechanism(s) of translation repression at postinitiation steps, remain poorly characterized. In the unicellular green alga Chlamydomonas reinhardtii, sRNAs derived from genome-integrated inverted repeat transgenes, perfectly complementary to the 3' untranslated region of a target transcript, can inhibit protein synthesis without or with only minimal mRNA destabilization. Here, we report that the sRNA-repressed transcripts are not altered in their polyadenylation status and they remain associated with polyribosomes, indicating inhibition at a postinitiation step of translation. Interestingly, ribosomes associated with sRNA-repressed transcripts show reduced sensitivity to translation inhibition by some antibiotics, such as cycloheximide, both in ribosome run-off assays and in in vivo experiments. Our results suggest that sRNA-mediated repression of protein synthesis in C. reinhardtii may involve alterations to the function/structural conformation of translating ribosomes. Additionally, sRNA-mediated translation inhibition is now known to occur in a number of phylogenetically diverse eukaryotes, suggesting that this mechanism may have been a feature of an ancestral RNA interference machinery.


Assuntos
Chlamydomonas reinhardtii/efeitos dos fármacos , Cicloeximida/farmacologia , Polirribossomos/efeitos dos fármacos , Biossíntese de Proteínas/efeitos dos fármacos , RNA de Plantas/metabolismo , RNA Interferente Pequeno/metabolismo , Chlamydomonas reinhardtii/classificação , Chlamydomonas reinhardtii/genética , Chlamydomonas reinhardtii/metabolismo , Meios de Cultura/metabolismo , Genes de Plantas , Higromicina B/farmacologia , Organismos Geneticamente Modificados/genética , Organismos Geneticamente Modificados/metabolismo , Filogenia , Polirribossomos/genética , Polirribossomos/metabolismo , Estabilidade de RNA , RNA de Plantas/genética , RNA Interferente Pequeno/genética , Relação Estrutura-Atividade
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA