Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 8 de 8
Filtrar
Mais filtros

Base de dados
Tipo de documento
Intervalo de ano de publicação
1.
J Virol ; 92(2)2018 01 15.
Artigo em Inglês | MEDLINE | ID: mdl-29093088

RESUMO

Prior studies have found that HIV, through the Vpr protein, promotes genome reduplication (polyploidy) in infection-surviving epithelial cells within renal tissue. However, the temporal progression and molecular regulation through which Vpr promotes polyploidy have remained unclear. Here we define a sequential progression to Vpr-mediated polyploidy in human renal tubule epithelial cells (RTECs). We found that as in many cell types, Vpr first initiates G2 cell cycle arrest in RTECs. We then identified a previously unreported cascade of Vpr-dependent events that lead to renal cell survival and polyploidy. Specifically, we found that a fraction of G2-arrested RTECs reenter the cell cycle. Following this cell cycle reentry, two distinct outcomes occur. Cells that enter complete mitosis undergo mitotic cell death due to extra centrosomes and aberrant division. Conversely, cells that abort mitosis undergo endoreplication to become polyploid. We further show that multiple small-molecule inhibitors of the phosphatidylinositol 3-kinase-related kinase (PIKK) family, including those that target ATR, ATM, and mTOR, indirectly prevent Vpr-mediated polyploidy by preventing G2 arrest. In contrast, an inhibitor that targets DNA-dependent protein kinase (DNA-PK) specifically blocks the Vpr-mediated transition from G2 arrest to polyploidy. These findings outline a temporal, molecularly regulated path to polyploidy in HIV-positive renal cells.IMPORTANCE Current cure-focused efforts in HIV research aim to elucidate the mechanisms of long-term persistence of HIV in compartments. The kidney is recognized as one such compartment, since viral DNA and mRNA persist in the renal tissues of HIV-positive patients. Further, renal disease is a long-term comorbidity in the setting of HIV. Thus, understanding the regulation and impact of HIV infection on renal cell biology will provide important insights into this unique HIV compartment. Our work identifies mechanisms that distinguish between HIV-positive cell survival and death in a known HIV compartment, as well as pharmacological agents that alter these outcomes.


Assuntos
Células Epiteliais/metabolismo , Células Epiteliais/virologia , HIV-1/fisiologia , Interações Hospedeiro-Patógeno , Mitose , Poliploidia , Produtos do Gene vpr do Vírus da Imunodeficiência Humana/metabolismo , Morte Celular , Linhagem Celular , Sobrevivência Celular , Proteína Quinase Ativada por DNA/antagonistas & inibidores , Imunofluorescência , Pontos de Checagem da Fase G2 do Ciclo Celular , Humanos , Túbulos Renais/citologia , Túbulos Renais/virologia , Microscopia de Fluorescência , Modelos Biológicos , Inibidores de Proteínas Quinases/farmacologia , Transdução de Sinais
2.
PLoS Biol ; 14(11): e2000998, 2016 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-27893764

RESUMO

Kaposi's sarcoma-associated herpesvirus (KSHV), the causative agent of Kaposi's sarcoma, encodes 25 mature viral miRNAs. MCP-1-induced protein-1 (MCPIP1), a critical regulator of immune homeostasis, has been shown to suppress miRNA biosynthesis via cleavage of precursor miRNAs through its RNase domain. We demonstrate that MCPIP1 can directly cleave KSHV and EBV precursor miRNAs and that MCPIP1 expression is repressed following de novo KSHV infection. In addition, repression with siRNAs to MCPIP1 in KSHV-infected cells increased IL-6 and KSHV miRNA expression, supporting a role for MCPIP1 in IL-6 and KSHV miRNA regulation. We also provide evidence that KSHV miRNAs repress MCPIP1 expression by targeting the 3'UTR of MCPIP1. Conversely, expression of essential miRNA biogenesis components Dicer and TRBP is increased following latent KSHV infection. We propose that KSHV infection inhibits a negative regulator of miRNA biogenesis (MCPIP1) and up-regulates critical miRNA processing components to evade host mechanisms that inhibit expression of viral miRNAs. KSHV-mediated alterations in miRNA biogenesis represent a novel mechanism by which KSHV interacts with its host and a new mechanism for the regulation of viral miRNA expression.


Assuntos
Herpesvirus Humano 8/fisiologia , MicroRNAs/fisiologia , Ribonucleases/fisiologia , Fatores de Transcrição/fisiologia , Humanos , RNA Interferente Pequeno/genética , Ribonucleases/genética , Fatores de Transcrição/genética
3.
J Virol ; 89(8): 4249-61, 2015 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-25631082

RESUMO

UNLABELLED: MicroRNAs (miRNAs) are small, ∼ 22-nucleotide-long RNAs that regulate gene expression posttranscriptionally. Kaposi's sarcoma-associated herpesvirus (KSHV) encodes 12 pre-miRNAs during latency, and the functional significance of these microRNAs during KSHV infection and their cellular targets have been emerging recently. Using a previously reported microarray profiling analysis, we identified breakpoint cluster region mRNA (Bcr) as a cellular target of the KSHV miRNA miR-K12-6-5p (miR-K6-5). Bcr protein levels were repressed in human umbilical vein endothelial cells (HUVECs) upon transfection with miR-K6-5 and during KSHV infection. Luciferase assays wherein the Bcr 3' untranslated region (UTR) was cloned downstream of a luciferase reporter showed repression in the presence of miR-K6-5, and mutation of one of the two predicted miR-K6-5 binding sites relieved this repression. Furthermore, inhibition or deletion of miR-K6-5 in KSHV-infected cells showed increased Bcr protein levels. Together, these results show that Bcr is a direct target of the KSHV miRNA miR-K6-5. To understand the functional significance of Bcr knockdown in the context of KSHV-associated disease, we hypothesized that the knockdown of Bcr, a negative regulator of Rac1, might enhance Rac1-mediated angiogenesis. We found that HUVECs transfected with miR-K6-5 had increased Rac1-GTP levels and tube formation compared to HUVECs transfected with control miRNAs. Knockdown of Bcr in latently KSHV-infected BCBL-1 cells increased the levels of viral RTA, suggesting that Bcr repression by KSHV might aid lytic reactivation. Together, our results reveal a new function for both KSHV miRNAs and Bcr in KSHV infection and suggest that KSHV miRNAs, in part, promote angiogenesis and lytic reactivation. IMPORTANCE: Kaposi's sarcoma (KS)-associated herpesvirus (KSHV) infection is linked to multiple human cancers and lymphomas. KSHV encodes small nucleic acids (microRNAs) that can repress the expression of specific human genes, the biological functions of which are still emerging. This report uses a variety of approaches to show that a KSHV microRNA represses the expression of the human gene called breakpoint cluster region (Bcr). Repression of Bcr correlated with the activation of a protein previously shown to cause KS-like lesions in mice (Rac1), an increase in KS-associated phenotypes (tube formation in endothelial cells and vascular endothelial growth factor [VEGF] synthesis), and modification of the life cycle of the virus (lytic replication). Our results suggest that KSHV microRNAs suppress host proteins and contribute to KS-associated pathogenesis.


Assuntos
Regulação da Expressão Gênica/fisiologia , Infecções por Herpesviridae/metabolismo , Herpesvirus Humano 8/genética , MicroRNAs/genética , Neovascularização Fisiológica/fisiologia , Proteínas Proto-Oncogênicas c-bcr/metabolismo , Proteínas rac1 de Ligação ao GTP/metabolismo , Western Blotting , Clonagem Molecular , Primers do DNA/genética , Imunofluorescência , Regulação da Expressão Gênica/genética , Infecções por Herpesviridae/genética , Células Endoteliais da Veia Umbilical Humana , Humanos , Luciferases , Mutagênese Sítio-Dirigida , Neovascularização Fisiológica/genética , Proteínas Proto-Oncogênicas c-bcr/genética , Reação em Cadeia da Polimerase em Tempo Real , Proteínas rac1 de Ligação ao GTP/genética
4.
J Virol ; 86(21): 11663-74, 2012 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-22896623

RESUMO

Kaposi's sarcoma (KS)-associated herpesvirus (KSHV) is the causative agent of KS, an important AIDS-associated malignancy. KSHV expresses at least 18 different mature microRNAs (miRNAs). We identified interleukin-1 receptor (IL-1R)-associated kinase 1 (IRAK1) as a potential target of miR-K12-9 (miR-K9) in an array data set examining changes in cellular gene expression levels in the presence of KSHV miRNAs. Using 3'-untranslated region (3'UTR) luciferase reporter assays, we confirmed that miR-K9 and other miRNAs inhibit IRAK1 expression. In addition, IRAK1 expression is downregulated in cells transfected with miR-K9 and during de novo KSHV infection. IRAK1 is an important component of the Toll-like receptor (TLR)/IL-1R signaling cascade. The downregulation of IRAK1 by miR-K9 resulted in the decreased stimulation of NF-κB activity in endothelial cells treated with IL-1α and in B cells treated with a TLR7/8 agonist. Interestingly, miR-K9 had a greater effect on NF-κB activity than did a small interfering RNA (siRNA) targeting IRAK1 despite the more efficient downregulation of IRAK1 expression with the siRNA. We hypothesized that KSHV miRNAs may also be regulating a second component of the TLR/IL-1R signaling cascade, resulting in a stronger phenotype. Reanalysis of the array data set identified myeloid differentiation primary response protein 88 (MYD88) as an additional potential target. 3'UTR luciferase reporter assays and Western blot analysis confirmed the targeting of MYD88 by miR-K5. The presence of miR-K9 and miR-K5 inhibited the production of IL-6 and IL-8 upon the IL-1α stimulation of endothelial cells. These results demonstrate KSHV-encoded miRNAs regulating the TLR/IL-1R signaling cascade at two distinct points and suggest the importance of these pathways during viral infection.


Assuntos
Citocinas/antagonistas & inibidores , Herpesvirus Humano 8/imunologia , Herpesvirus Humano 8/patogenicidade , Quinases Associadas a Receptores de Interleucina-1/antagonistas & inibidores , MicroRNAs/metabolismo , Fator 88 de Diferenciação Mieloide/antagonistas & inibidores , Transdução de Sinais , Fusão Gênica Artificial , Western Blotting , Linhagem Celular , Perfilação da Expressão Gênica , Inativação Gênica , Genes Reporter , Humanos , Evasão da Resposta Imune , Quinases Associadas a Receptores de Interleucina-1/genética , Interleucinas/imunologia , Interleucinas/metabolismo , Luciferases/análise , Luciferases/genética , MicroRNAs/genética , Análise em Microsséries , Fator 88 de Diferenciação Mieloide/genética , Receptores Toll-Like/imunologia , Receptores Toll-Like/metabolismo
5.
J Virol ; 85(1): 305-14, 2011 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-20980522

RESUMO

Gag orchestrates the assembly and release of human immunodeficiency virus type 1 (HIV-1) particles. We explored here the potential of anti-Gag RNA aptamers to inhibit HIV-1 replication. In vitro, RNA aptamers raised against an HIV-1 Gag protein, lacking the N-terminal myristate and the C-terminal p6 (DP6-Gag), could bind to matrix protein (MA), nucleocapsid protein (NC), or entire DP6-Gag protein. Upon cotransfection with pNL4-3.Luc molecular clone into 293T cells, six of the aptamers caused mild inhibition (2- to 3-fold) in the extracellular capsid levels, and one aptamer displayed 20-fold inhibition. The reduction was not due to a release defect but reflected Gag mRNA levels. We hypothesized that the aptamers influence genomic RNA levels via perturbation of specific Gag-genomic RNA interactions. Binding studies revealed that the "NC-binders" specifically compete with the packaging signal (ψ) of HIV-1 for binding to DP6-Gag. Therefore, we tested the ability of two NC-binders to inhibit viruses containing ψ-region deletions (ΔSL1 or ΔSL3) and found that the NC-binders were no longer able to inhibit Gag synthesis. The inability of these aptamers to inhibit ψ-deleted viruses correlated with the absence of competition with the corresponding ψ transcripts lacking SL1 or SL3 for binding DP6-Gag in vitro. These results indicate that the NC-binding aptamers disrupt Gag-genomic RNA interaction and negatively affect genomic RNA transcription, processing, or stability. Our results reveal an essential interaction between HIV-1 Gag and the ψ-region that may be distinct from that which occurs during the encapsidation of genomic RNA. Thus, anti-Gag aptamers can be an effective tool to perturb Gag-genomic RNA interactions.


Assuntos
Aptâmeros de Nucleotídeos/metabolismo , Produtos do Gene gag/metabolismo , Antígenos HIV/metabolismo , HIV-1/efeitos dos fármacos , Proteínas do Nucleocapsídeo/metabolismo , Produtos do Gene gag do Vírus da Imunodeficiência Humana/metabolismo , Aptâmeros de Nucleotídeos/farmacologia , Linhagem Celular , Produtos do Gene gag/química , Antígenos HIV/química , HIV-1/química , HIV-1/genética , HIV-1/metabolismo , Humanos , Proteínas do Nucleocapsídeo/química , Ligação Proteica , Montagem de Vírus/efeitos dos fármacos , Replicação Viral/efeitos dos fármacos , Produtos do Gene gag do Vírus da Imunodeficiência Humana/química
6.
Sci Rep ; 7: 40813, 2017 01 19.
Artigo em Inglês | MEDLINE | ID: mdl-28102325

RESUMO

Kaposi's sarcoma-associated herpesvirus (KSHV) encodes 12 pre-microRNAs during latency that are processed to yield ~25 mature microRNAs (miRNAs). We were interested in identifying cellular networks that were targeted by KSHV-miRNAs and employed network building strategies using validated KSHV miRNA targets. Here, we report the identification of a gene network centering on the transcription factor- signal transducer and activator of transcription 3 (STAT3) that is targeted by KSHV miRNAs. KSHV miRNAs suppressed STAT3 and STAT5 activation and inhibited STAT3-dependent reporter activation upon IL6-treatment. KSHV miRNAs also repressed the induction of antiviral interferon-stimulated genes upon IFNα- treatment. Finally, we observed increased lytic reactivation of KSHV from latently infected cells upon STAT3 repression with siRNAs or a small molecule inhibitor. Our data suggest that treatment of infected cells with a STAT3 inhibitor and a viral replication inhibitor, ganciclovir, represents a possible strategy to eliminate latently infected cells without increasing virion production. Together, we show that KSHV miRNAs suppress a network of targets associated with STAT3, deregulate cytokine-mediated gene activation, suppress an interferon response, and influence the transition into the lytic phase of viral replication.


Assuntos
Interferons/metabolismo , Fator de Transcrição STAT3/metabolismo , Regiões 3' não Traduzidas , Antagomirs , Sequência de Bases , Redes Reguladoras de Genes , Vetores Genéticos , Herpesvirus Humano 8/genética , Células Endoteliais da Veia Umbilical Humana , Humanos , Proteínas Inibidoras de Apoptose/genética , Proteínas Inibidoras de Apoptose/metabolismo , Interleucina-6/farmacologia , MicroRNAs/antagonistas & inibidores , MicroRNAs/genética , MicroRNAs/metabolismo , Interferência de RNA , RNA Interferente Pequeno/metabolismo , Receptores da Eritropoetina/genética , Receptores da Eritropoetina/metabolismo , Fator de Transcrição STAT3/antagonistas & inibidores , Fator de Transcrição STAT3/genética , Alinhamento de Sequência , Transdução de Sinais/efeitos dos fármacos , Survivina
7.
PLoS One ; 10(8): e0135560, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-26263384

RESUMO

Kaposi's sarcoma (KS) is characterized by highly vascularized spindle-cell tumors induced after infection of endothelial cells by Kaposi's sarcoma-associated herpesvirus (KSHV). In KS tumors, KSHV expresses only a few latent proteins together with 12 pre-microRNAs. Previous microarray and proteomic studies predicted that multiple splice variants of the tumor suppressor protein tropomyosin 1 (TPM1) were targets of KSHV microRNAs. Here we show that at least two microRNAs of KSHV, miR-K2 and miR-K5, repress protein levels of specific isoforms of TPM1. We identified a functional miR-K5 binding site in the 3' untranslated region (UTR) of one TPM1 isoform. Furthermore, the inhibition or loss of miR-K2 or miR-K5 restores expression of TPM1 in KSHV-infected cells. TPM1 protein levels were also repressed in KSHV-infected clinical samples compared to uninfected samples. Functionally, miR-K2 increases viability of unanchored human umbilical vein endothelial cells (HUVEC) by inhibiting anoikis (apoptosis after cell detachment), enhances tube formation of HUVECs, and enhances VEGFA expression. Taken together, KSHV miR-K2 and miR-K5 may facilitate KSHV pathogenesis.


Assuntos
Células Endoteliais/metabolismo , Herpesvirus Humano 8/genética , MicroRNAs/genética , Interferência de RNA , RNA Viral , Tropomiosina/genética , Regiões 3' não Traduzidas , Anoikis/genética , Linhagem Celular , Células Endoteliais/virologia , Regulação da Expressão Gênica , Ordem dos Genes , Células Endoteliais da Veia Umbilical Humana , Humanos , Peso Molecular , Isoformas de Proteínas , RNA Mensageiro/genética , Tropomiosina/química , Tropomiosina/metabolismo
8.
Viruses ; 4(9): 1687-710, 2012 09.
Artigo em Inglês | MEDLINE | ID: mdl-23170179

RESUMO

EBV and KSHV are both gamma-herpesviruses which express multiple viral microRNAs. Various methods have been used to investigate the functions of these microRNAs, largely through identification of microRNA target genes. Surprisingly, these related viruses do not share significant sequence homology in their microRNAs. A number of reports have described functions of EBV and KSHV microRNA targets, however only three experimentally validated target genes have been shown to be targeted by microRNAs from both viruses. More sensitive methods to identify microRNA targets have predicted approximately 60% of host targets could be shared by EBV and KSHV microRNAs, but by targeting different sequences in the host targets. In this review, we explore the similarities of microRNA functions and targets of these related viruses.


Assuntos
Herpesvirus Humano 4/patogenicidade , Herpesvirus Humano 8/patogenicidade , MicroRNAs/metabolismo , RNA Viral/metabolismo , Fatores de Virulência/metabolismo , Regulação da Expressão Gênica , Herpesvirus Humano 4/genética , Herpesvirus Humano 8/genética , Humanos , MicroRNAs/genética , RNA Viral/genética , Fatores de Virulência/genética
SELEÇÃO DE REFERÊNCIAS
Detalhe da pesquisa