Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 12 de 12
Filtrar
Mais filtros

Base de dados
Tipo de documento
Intervalo de ano de publicação
1.
Nature ; 564(7735): 278-282, 2018 12.
Artigo em Inglês | MEDLINE | ID: mdl-30487602

RESUMO

The entry of foreign DNA into many mammalian cell types triggers the innate immune system, a complex set of responses to prevent infection by pathogens. One aspect of the response is the potent epigenetic silencing of incoming viral DNAs1, including the extrachromosomal DNAs that are formed immediately after infection by retroviruses. These unintegrated viral DNAs are very poorly transcribed in all cells, even in permissive cells, in contrast to the robust expression that is observed after viral integration2-5. The factors that are responsible for this low expression have not yet been identified. Here we performed a genome-wide CRISPR-Cas9 screen for genes that are required for silencing an integrase-deficient MLV-GFP reporter virus to explore the mechanisms responsible for repression of unintegrated viral DNAs in human cells. Our screen identified the DNA-binding protein NP220, the three proteins (MPP8, TASOR and PPHLN1) that comprise the HUSH complex-which silences proviruses in heterochromatin6 and retrotransposons7,8-the histone methyltransferase SETDB1, and other host factors that are required for silencing. Further tests by chromatin immunoprecipitation showed that NP220 is the key protein that recruits the HUSH complex, SETDB1 and the histone deacetylases HDAC1 and HDAC4 to silence the unintegrated retroviral DNA. Knockout of NP220 accelerates the replication of retroviruses. These experiments identify the molecular machinery that silences extrachromosomal retroviral DNA.


Assuntos
DNA Viral/genética , Proteínas de Ligação a DNA/metabolismo , Inativação Gênica , Complexos Multiproteicos/metabolismo , Proteínas Nucleares/metabolismo , Retroviridae/genética , Antígenos de Neoplasias/metabolismo , Proteína 9 Associada à CRISPR/genética , Proteína 9 Associada à CRISPR/metabolismo , Imunoprecipitação da Cromatina , Repetições Palindrômicas Curtas Agrupadas e Regularmente Espaçadas/genética , Histona Desacetilase 1/metabolismo , Histona Desacetilases/metabolismo , Histona-Lisina N-Metiltransferase , Humanos , Fosfoproteínas/metabolismo , Proteínas Metiltransferases/metabolismo , Provírus/genética , Proteínas de Ligação a RNA , Proteínas Repressoras/metabolismo , Fatores de Transcrição , Integração Viral
2.
Proc Natl Acad Sci U S A ; 117(31): 18701-18710, 2020 08 04.
Artigo em Inglês | MEDLINE | ID: mdl-32690679

RESUMO

Yin Yang 1 (YY1) is a DNA-binding transcription factor that either activates or represses gene expression. YY1 has previously been implicated in the transcriptional silencing of many retroviruses by binding to DNA sequences in the U3 region of the viral long terminal repeat (LTR). We here show that YY1 overexpression leads to profound activation, rather than repression, of human T lymphotropic virus type 1 (HTLV-1) expression, while YY1 down-regulation reduces HTLV-1 expression. The YY1 responsive element mapped not to YY1 DNA-binding sites in the HTLV-1 LTR but to the R region. The HTLV-1 R sequence alone is sufficient to provide YY1 responsiveness to a nonresponsive promoter, but only in the sense orientation and only when included as part of the mRNA. YY1 binds to the R region of HTLV-1 RNA in vitro and in vivo, leading to increased transcription initiation and elongation. The findings indicate that YY1 is a potent transactivator of HTLV-1 gene expression acting via binding viral RNA, rather than DNA.


Assuntos
Regulação Viral da Expressão Gênica/genética , Vírus Linfotrópico T Tipo 1 Humano , RNA/metabolismo , Sequências Repetidas Terminais/genética , Fator de Transcrição YY1 , Células HEK293 , Vírus Linfotrópico T Tipo 1 Humano/genética , Vírus Linfotrópico T Tipo 1 Humano/metabolismo , Humanos , Células Jurkat , Ligação Proteica/genética , RNA/genética , Ativação Transcricional/genética , Fator de Transcrição YY1/genética , Fator de Transcrição YY1/metabolismo
3.
J Virol ; 95(15): e0049521, 2021 07 12.
Artigo em Inglês | MEDLINE | ID: mdl-34011543

RESUMO

During retrovirus infection, a histone-free DNA copy of the viral RNA genome is synthesized and rapidly loaded with nucleosomes de novo upon nuclear entry. The potential role of viral accessory proteins in histone loading onto retroviral DNAs has not been extensively investigated. The p12 protein of Moloney murine leukemia virus (MMLV) is a virion protein that is critical for tethering the incoming viral DNA to host chromatin in the early stages of infection. Infection by virions containing a mutant p12 (PM14) defective in chromatin tethering results in the formation of viral DNAs that do not accumulate in the nucleus. In this report, we show that viral DNAs of these mutants are not loaded with histones. Moreover, the DNA genomes delivered by mutant p12 show prolonged association with viral structural proteins nucleocapsid (NC) and capsid (CA). The histone-poor viral DNA genomes do not become associated with the host RNA polymerase II machinery. These findings provide insights into fundamental aspects of retroviral biology, indicating that tethering to host chromatin by p12 and retention in the nucleus are required to allow loading of histones onto the viral DNA. IMPORTANCE Incoming retroviral DNAs are rapidly loaded with nucleosomal histones upon entry into the nucleus and before integration into the host genome. The entry of murine leukemia virus DNA into the nucleus occurs only upon dissolution of the nuclear membrane in mitosis, and retention in the nucleus requires the action of a viral protein, p12, which tethers the DNA to host chromatin. Data presented here show that the tethering activity of p12 is required for the loading of histones onto the viral DNA. p12 mutants lacking tethering activity fail to acquire histones, retain capsid and nucleocapsid proteins, and are poorly transcribed. The work defines a new requirement for a viral protein to allow chromatinization of viral DNA.


Assuntos
Proteínas do Capsídeo/metabolismo , Produtos do Gene gag/genética , Histonas/metabolismo , Vírus da Leucemia Murina de Moloney/crescimento & desenvolvimento , Vírus da Leucemia Murina de Moloney/metabolismo , Capsídeo/metabolismo , Linhagem Celular Tumoral , Cromatina/metabolismo , DNA Viral/metabolismo , Genoma Viral/genética , Células HEK293 , Células HeLa , Humanos , Vírus da Leucemia Murina de Moloney/genética , Montagem de Vírus/genética
4.
J Virol ; 91(1)2017 Jan 01.
Artigo em Inglês | MEDLINE | ID: mdl-27795446

RESUMO

Embryonic carcinoma (EC) cells are malignant counterparts of embryonic stem (ES) cells and serve as useful models for investigating cellular differentiation and human embryogenesis. Though the susceptibility of murine EC cells to retroviral infection has been extensively analyzed, few studies of retrovirus infection of human EC cells have been performed. We tested the susceptibility of human EC cells to transduction by retroviral vectors derived from three different retroviral genera. We show that human EC cells efficiently express reporter genes delivered by vectors based on human immunodeficiency virus type 1 (HIV-1) and Mason-Pfizer monkey virus (M-PMV) but not Moloney murine leukemia virus (MLV). In human EC cells, MLV integration occurs normally, but no viral gene expression is observed. The block to MLV expression of MLV genomes is relieved upon cellular differentiation. The lack of gene expression is correlated with transcriptional silencing of the MLV promoter through the deposition of repressive histone marks as well as DNA methylation. Moreover, depletion of SETDB1, a histone methyltransferase, resulted in a loss of transcriptional silencing and upregulation of MLV gene expression. Finally, we provide evidence showing that the lack of MLV gene expression may be attributed in part to the lack of MLV enhancer function in human EC cells. IMPORTANCE: Human embryonic carcinoma (EC) cells are shown to restrict the expression of murine leukemia virus genomes but not retroviral genomes of the lentiviral or betaretroviral families. The block occurs at the level of transcription and is accompanied by the deposition of repressive histone marks and methylation of the integrated proviral DNA. The host machinery required for silencing in human EC cells is distinct from that in murine EC cell lines: the histone methyltransferase SETDB1 is required, but the widely utilized corepressor TRIM28/Kap1 is not. A transcriptional enhancer element from the Mason-Pfizer monkey virus can override the silencing and promote transcription of chimeric proviral DNAs. The findings reveal novel features of human EC gene regulation not present in their murine counterparts.


Assuntos
Inativação Gênica , Genoma Viral , HIV-1/genética , Células-Tronco Embrionárias Humanas/imunologia , Vírus dos Macacos de Mason-Pfizer/genética , Vírus da Leucemia Murina de Moloney/genética , Células-Tronco Neoplásicas/imunologia , Animais , Diferenciação Celular , Metilação de DNA , Genes Reporter , HIV-1/metabolismo , Histona-Lisina N-Metiltransferase , Histonas/genética , Histonas/imunologia , Especificidade de Hospedeiro , Células-Tronco Embrionárias Humanas/virologia , Humanos , Vírus dos Macacos de Mason-Pfizer/metabolismo , Camundongos , Vírus da Leucemia Murina de Moloney/metabolismo , Células-Tronco Neoplásicas/virologia , Regiões Promotoras Genéticas , Proteínas Metiltransferases/antagonistas & inibidores , Proteínas Metiltransferases/genética , Proteínas Metiltransferases/imunologia , RNA Interferente Pequeno/genética , RNA Interferente Pequeno/metabolismo , Especificidade da Espécie , Transcrição Gênica
5.
J Biol Chem ; 290(36): 21890-900, 2015 Sep 04.
Artigo em Inglês | MEDLINE | ID: mdl-26198631

RESUMO

Yin Yang 1 (YY1) is a member of the GLI-Krüppel class of DNA and RNA binding transcription factors that can either activate or repress gene expression during cell growth, differentiation, and embryogenesis. Although much is known about YY1 interacting proteins and the target promoters regulated by YY1, much less is known about YY1 regulation through post-translational modifications. In this study we show that YY1 is tyrosine-phosphorylated in multiple cell types. Using a combination of pharmacological inhibition, kinase overexpression, and kinase knock-out studies, we demonstrate that YY1 is a target of multiple Src family kinases in vitro and in vivo. Moreover, we have identified multiple sites of YY1 phosphorylation and analyzed the effect of phosphorylation on the activity of YY1-responsive retroviral and cellular promoters. Phosphorylation of tyrosine 383 interferes with DNA and RNA binding, leading to the down-regulation of YY1 activity. Finally, we provide the first evidence that YY1 is a downstream target of epidermal growth factor receptor signaling in vivo. Taken together, the identification of YY1 as a target of Src family kinases provide key insights into the inhibitory role of tyrosine kinases in modulating YY1 activity.


Assuntos
Tirosina/metabolismo , Fator de Transcrição YY1/metabolismo , Quinases da Família src/metabolismo , Animais , Sítios de Ligação/genética , Western Blotting , Linhagem Celular , Linhagem Celular Tumoral , Fator de Crescimento Epidérmico/farmacologia , Expressão Gênica/efeitos dos fármacos , Células HEK293 , Células HeLa , Humanos , Mesilato de Imatinib/farmacologia , Indóis/farmacologia , Células Jurkat , Mutação , Fosforilação/efeitos dos fármacos , Regiões Promotoras Genéticas/genética , Ligação Proteica/genética , Inibidores de Proteínas Quinases/farmacologia , Proteínas Proto-Oncogênicas c-fos/genética , Interferência de RNA , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Sulfonamidas/farmacologia , Tirosina/genética , Fator de Transcrição YY1/genética , Quinases da Família src/antagonistas & inibidores , Quinases da Família src/genética
6.
J Virol ; 89(5): 2813-9, 2015 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-25540373

RESUMO

UNLABELLED: Mason-Pfizer monkey virus (M-PMV) is a prototypical betaretrovirus responsible for simian AIDS (SAIDS) in rhesus macaques. It has been shown previously that mouse cells are resistant to infection by HIV-1 and other primate lentiviruses. However, the susceptibility of mouse cells to primate retroviruses such as M-PMV remains unexplored. In the present study, using single-round green fluorescent protein (GFP) reporter viruses, we showed that various mouse cell lines are unable to support the early stages of M-PMV replication. The block to infection occurs postentry and is independent of the viral envelope. Using quantitative real-time PCR, we showed that the block to infection occurs after reverse transcription but before formation of circular DNA or proviral DNA. Finally, we showed that the M-PMV block in mouse cells is not attributable to the previously characterized mouse restriction factor Fv1. Overall, these findings suggest that mouse cells exhibit a previously uncharacterized block to M-PMV infection. IMPORTANCE: Here we document a novel postentry restriction to M-PMV infection in mouse cells. The block occurs after reverse transcription but before the formation of circular or proviral DNA and is independent of the previous characterized mouse restriction factor Fv1.


Assuntos
Vírus dos Macacos de Mason-Pfizer/imunologia , Vírus dos Macacos de Mason-Pfizer/fisiologia , Integração Viral , Animais , Linhagem Celular , Genes Reporter , Proteínas de Fluorescência Verde/análise , Camundongos , Reação em Cadeia da Polimerase em Tempo Real , Coloração e Rotulagem
7.
Circ Res ; 114(3): 421-33, 2014 Jan 31.
Artigo em Inglês | MEDLINE | ID: mdl-24297735

RESUMO

RATIONALE: Mitochondrial oxidative stress (mitoOS) has been shown to correlate with the progression of human atherosclerosis. However, definitive cell type-specific causation studies in vivo are lacking, and the molecular mechanisms of potential proatherogenic effects remain to be determined. OBJECTIVE: Our aims were to assess the importance of macrophage mitoOS in atherogenesis and to explore the underlying molecular mechanisms. METHODS AND RESULTS: We first validated Western diet-fed Ldlr(-/-) mice as a model of human mitoOS-atherosclerosis association by showing that non-nuclear oxidative DNA damage, a marker of mitoOS in lesional macrophages, correlates with aortic root lesion development. To investigate the importance of macrophage mitoOS, we used a genetic engineering strategy in which the OS suppressor catalase was ectopically expressed in mitochondria (mCAT) in macrophages. MitoOS in lesional macrophages was successfully suppressed in these mice, and this led to a significant reduction in aortic root lesional area. The mCAT lesions had less monocyte-derived cells, less Ly6c(hi) monocyte infiltration into lesions, and lower levels of monocyte chemotactic protein-1. The decrease in lesional monocyte chemotactic protein-1 was associated with the suppression of other markers of inflammation and with decreased phosphorylation of RelA (NF-κB p65), indicating decreased activation of the proinflammatory NF-κB pathway. Using models of mitoOS in cultured macrophages, we showed that mCAT suppressed monocyte chemotactic protein-1 expression by decreasing the activation of the IκB-kinase ß-RelA NF-κB pathway. CONCLUSIONS: MitoOS in lesional macrophages amplifies atherosclerotic lesion development by promoting NF-κB-mediated entry of monocytes and other inflammatory processes. In view of the mitoOS-atherosclerosis link in human atheromata, these findings reveal a potentially new therapeutic target to prevent the progression of atherosclerosis.


Assuntos
Aterosclerose/metabolismo , Macrófagos/metabolismo , Mitocôndrias/metabolismo , NF-kappa B/genética , Estresse Oxidativo , Animais , Aterosclerose/patologia , Células Cultivadas , Feminino , Inflamação/genética , Inflamação/metabolismo , Inflamação/patologia , Macrófagos/patologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Camundongos Transgênicos , Mitocôndrias/genética , Mitocôndrias/patologia , Estresse Oxidativo/genética
8.
J Virol ; 88(3): 1825-9, 2014 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-24227866

RESUMO

Mouse embryonic cells are unable to support the replication of Moloney murine leukemia virus (MLV). The integrated viral DNA is transcriptionally silenced, largely due to binding of host transcriptional repressors to the primer binding site (PBS) of the provirus. We have previously shown that a PBS DNA-binding repressor complex contains ZFP809 and TRIM28. Here, we identified ErbB3-binding protein 1 (EBP1) to be a novel component of the ZFP809-TRIM28 silencing complex and show that EBP1 depletion reduces PBS-mediated retroviral silencing.


Assuntos
Primers do DNA/genética , Inativação Gênica , Leucemia/veterinária , Vírus da Leucemia Murina de Moloney/genética , Proteínas Nucleares/metabolismo , Doenças dos Roedores/metabolismo , Animais , Sítios de Ligação , Linhagem Celular Tumoral , Primers do DNA/metabolismo , Proteínas de Ligação a DNA/genética , Proteínas de Ligação a DNA/metabolismo , Leucemia/embriologia , Leucemia/genética , Leucemia/metabolismo , Leucemia/virologia , Camundongos , Vírus da Leucemia Murina de Moloney/química , Vírus da Leucemia Murina de Moloney/fisiologia , Proteínas Nucleares/genética , Ligação Proteica , RNA Viral/química , RNA Viral/genética , RNA Viral/metabolismo , Proteínas de Ligação a RNA , Proteínas Repressoras/genética , Proteínas Repressoras/metabolismo , Doenças dos Roedores/embriologia , Doenças dos Roedores/genética , Doenças dos Roedores/virologia , Proteína 28 com Motivo Tripartido , Replicação Viral
9.
Nat Commun ; 13(1): 1474, 2022 03 18.
Artigo em Inglês | MEDLINE | ID: mdl-35304442

RESUMO

Retroviruses utilize the viral integrase (IN) protein to integrate a DNA copy of their genome into host chromosomal DNA. HIV-1 integration sites are highly biased towards actively transcribed genes, likely mediated by binding of the IN protein to specific host factors, particularly LEDGF, located at these gene regions. We here report a substantial redirection of integration site distribution induced by a single point mutation in HIV-1 IN. Viruses carrying the K258R IN mutation exhibit a high frequency of integrations into centromeric alpha satellite repeat sequences, as assessed by deep sequencing, a more than 10-fold increase over wild-type. Quantitative PCR and in situ immunofluorescence assays confirm this bias of the K258R mutant virus for integration into centromeric DNA. Immunoprecipitation studies identify host factors binding to IN that may account for the observed bias for integration into centromeres. Centromeric integration events are known to be enriched in the latent reservoir of infected memory T cells, as well as in elite controllers who limit viral replication without intervention. The K258R point mutation in HIV-1 IN is also present in databases of latent proviruses found in patients, and may reflect an unappreciated aspect of the establishment of viral latency.


Assuntos
Integrase de HIV , Mutação Puntual , Centrômero/genética , Centrômero/metabolismo , Integrase de HIV/genética , Integrase de HIV/metabolismo , Humanos , Provírus/genética , Integração Viral/genética , Replicação Viral
10.
Cell Host Microbe ; 20(6): 798-809, 2016 Dec 14.
Artigo em Inglês | MEDLINE | ID: mdl-27866901

RESUMO

Chromosomal structure of nuclear DNA is usually maintained by insertion of nucleosomes into preexisting chromatin, both on newly synthesized DNA at replication forks and at sites of DNA damage. But during retrovirus infection, a histone-free DNA copy of the viral genome is synthesized that must be loaded with nucleosomes de novo. Here, we show that core histones are rapidly loaded onto unintegrated Moloney murine leukemia virus DNAs. Loading of nucleosomes requires nuclear entry, but does not require viral DNA integration. The histones associated with unintegrated DNAs become marked by covalent modifications, with a delay relative to the time of core histone loading. Expression from unintegrated DNA can be enhanced by modulation of the histone-modifying machinery. The data show that histone loading onto unintegrated DNAs occurs very rapidly after nuclear entry and does not require prior establishment of an integrated provirus.


Assuntos
DNA Viral , Código das Histonas , Histonas/metabolismo , Retroviridae/genética , Internalização do Vírus , Animais , Ciclo Celular , Cromatina , Imunoprecipitação da Cromatina , Dano ao DNA , Replicação do DNA/genética , DNA Viral/análise , Epigenômica , Células HEK293/virologia , Humanos , Camundongos , Vírus da Leucemia Murina de Moloney/genética , Células NIH 3T3/virologia , Nucleossomos/genética , Infecções por Retroviridae/virologia , Integração Viral
11.
Cell Rep ; 4(1): 50-8, 2013 Jul 11.
Artigo em Inglês | MEDLINE | ID: mdl-23810560

RESUMO

Embryonic cells transcriptionally repress the expression of endogenous and exogenous retroelements. Trim28, a key player in this silencing, is known to act in a large DNA-bound complex, but the other components of the complex are not fully characterized. Here, we show that the zinc finger protein Yin Yang 1 (YY1) is one such component. YY1 binds to the long terminal repeat (LTR) region of both exogenous and endogenous retroviruses (ERVs). Deletion of the YY1-binding site from the retroviral genome leads to a major loss of silencing in embryonic cells and a coordinated loss of repressive histone marks from the proviral chromatin. Depletion of YY1 protein results in marked upregulation of expression of exogenous viruses and of selected ERVs. Finally, we report an embryonic cell-specific interaction between YY1 and Trim28. Our results suggest a major role for YY1 in the silencing of both exogenous retroviruses and ERVs in embryonic cells.


Assuntos
Células-Tronco Embrionárias/metabolismo , Inativação Gênica , Provírus/genética , Fator de Transcrição YY1/metabolismo , Animais , Sítios de Ligação , Cromatina/metabolismo , DNA Viral/genética , DNA Viral/metabolismo , Células-Tronco Embrionárias/virologia , Deleção de Genes , Células HEK293 , Histonas/metabolismo , Humanos , Camundongos , Vírus da Leucemia Murina de Moloney/genética , Vírus da Leucemia Murina de Moloney/metabolismo , Células NIH 3T3 , Proteínas Nucleares/metabolismo , Provírus/metabolismo , Proteínas Repressoras/metabolismo , Sequências Repetidas Terminais , Transcrição Gênica , Proteína 28 com Motivo Tripartido , Fator de Transcrição YY1/genética
12.
Eur J Neurosci ; 18(8): 2403-7, 2003 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-14622203

RESUMO

Neurotransmitter release sites at the freeze-fractured frog neuromuscular junction are composed of inner and outer paired rows of large membrane particles, the putative calcium channels, anchored by the ribs of an underlying protein scaffold. We analysed the locations of the release site particles as a reflection of the scaffold structure, comparing particle distributions in secreting terminals with those where secretion was blocked with botulinum toxin A, which cleaves a small segment off SNAP-25, or botulinum toxin C1, which cleaves the cytoplasmic domain of syntaxin. In the idle terminal the inner and outer paired rows were located approximately 25 and approximately 44 nm, respectively, from the release site midline. However, adjacent to vesicular fusion sites both particle rows were displaced towards the midline by approximately 25%. The intervals between the particles along each row were examined by a nearest-neighbour approach. In control terminals the peak interval along the inner row was approximately 17 nm, consistent with previous reports and the spacing of the scaffold ribs. While the average distance between particles in the outer row was also approximately 17 nm, a detailed analysis revealed short 'linear clusters' with a approximately 14 nm interval. These clusters were enriched at vesicle fusion sites, suggesting an association with the docking sites, and were eliminated by botulinum C1, but not A. Our findings suggest, first, that the release site scaffold ribs undergo a predictable, and possibly active, shortening during exocytosis and, second, that at the vesicle docking site syntaxin plays a role in the cross-linking of the rib tips to form the vesicle docking sites.


Assuntos
Toxinas Botulínicas/farmacologia , Exocitose/efeitos dos fármacos , Junção Neuromuscular/metabolismo , Neurotransmissores/metabolismo , Vesículas Transportadoras/efeitos dos fármacos , Animais , Anuros , Toxinas Botulínicas Tipo A/farmacologia , Técnica de Fratura por Congelamento/métodos , Proteínas de Membrana/metabolismo , Proteínas do Tecido Nervoso/metabolismo , Junção Neuromuscular/efeitos dos fármacos , Junção Neuromuscular/ultraestrutura , Distribuição Normal , Proteínas Qa-SNARE , Vesículas Sinápticas , Proteína 25 Associada a Sinaptossoma , Sinaptossomos/efeitos dos fármacos , Sinaptossomos/metabolismo , Sinaptossomos/ultraestrutura , Vesículas Transportadoras/metabolismo , Vesículas Transportadoras/ultraestrutura
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA