Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 24
Filtrar
1.
Genome Res ; 28(6): 836-845, 2018 06.
Artigo em Inglês | MEDLINE | ID: mdl-29728366

RESUMO

Retrotransposons encompass half of the human genome and contribute to the formation of heterochromatin, which provides nuclear structure and regulates gene expression. Here, we asked if the human silencing hub (HUSH) complex is necessary to silence retrotransposons and whether it collaborates with TRIM28 and the chromatin remodeler ATRX at specific genomic loci. We show that the HUSH complex contributes to de novo repression and DNA methylation of an SVA retrotransposon reporter. By using naïve versus primed mouse pluripotent stem cells, we reveal a critical role for the HUSH complex in naïve cells, implicating it in programming epigenetic marks in development. Although the HUSH component FAM208A binds to endogenous retroviruses (ERVs) and long interspersed element-1s (LINE-1s or L1s), it is mainly required to repress evolutionarily young L1s (mouse-specific lineages <5 million years old). TRIM28, in contrast, is necessary to repress both ERVs and young L1s. Genes co-repressed by TRIM28 and FAM208A are evolutionarily young, or exhibit tissue-specific expression, are enriched in young L1s, and display evidence for regulation through LTR promoters. Finally, we demonstrate that the HUSH complex is also required to repress L1 elements in human cells. Overall, these data indicate that the HUSH complex and TRIM28 co-repress young retrotransposons and new genes rewired by retrotransposon noncoding DNA.


Assuntos
Genoma Humano , Proteínas Nucleares/genética , Retroelementos/genética , Proteína 28 com Motivo Tripartido/genética , Animais , Metilação de DNA/genética , Retrovirus Endógenos/genética , Heterocromatina/genética , Humanos , Elementos Nucleotídeos Longos e Dispersos/genética , Camundongos , Regiões Promotoras Genéticas
2.
EMBO Rep ; 20(12): e49262, 2019 12 05.
Artigo em Inglês | MEDLINE | ID: mdl-31621182

RESUMO

Transposon silencing requires the histone methyltransferase SETDB1. In this issue of EMBO Reports, Tsusaka et al [1] and Osumi et al [2] illustrate how the cofactor ATF7IP and its fly homolog Windei (Wde) regulate the methyltransferase function of SETDB1 through its nuclear licensing. The new insight gained from these two articles will shift how we think about epigenetic regulation and its multiple layers of control.


Assuntos
Núcleo Celular , Epigênese Genética , Ubiquitinação
3.
Proc Natl Acad Sci U S A ; 115(15): E3529-E3538, 2018 04 10.
Artigo em Inglês | MEDLINE | ID: mdl-29581310

RESUMO

Adeno-associated virus (AAV) is a small human Dependovirus whose low immunogenicity and capacity for long-term persistence have led to its widespread use as vector for gene therapy. Despite great recent successes in AAV-based gene therapy, further improvements in vector technology may be hindered by an inadequate understanding of various aspects of basic AAV biology. AAV is unique in that its replication is largely dependent on a helper virus and cellular factors. In the absence of helper virus coinfection, wild-type AAV establishes latency through mechanisms that are not yet fully understood. Challenging the currently held model for AAV latency, we show here that the corepressor Krüppel-associated box domain-associated protein 1 (KAP1) binds the latent AAV2 genome at the rep ORF, leading to trimethylation of AAV2-associated histone 3 lysine 9 and that the inactivation of KAP1 repression is necessary for AAV2 reactivation and replication. We identify a viral mechanism for the counteraction of KAP1 in which interference with the KAP1 phosphatase protein phosphatase 1 (PP1) by the AAV2 Rep proteins mediates enhanced phosphorylation of KAP1-S824 and thus relief from KAP1 repression. Furthermore, we show that this phenomenon involves recruitment of the NIPP1 (nuclear inhibitor of PP1)-PP1α holoenzyme to KAP1 in a manner dependent upon the NIPP1 FHA domain, identifying NIPP1 as an interaction partner for KAP1 and shedding light on the mechanism through which PP1 regulates cellular KAP1 activity.


Assuntos
Proteínas de Ligação a DNA/metabolismo , Dependovirus/metabolismo , Receptores de Neuropeptídeo Y/antagonistas & inibidores , Proteína 28 com Motivo Tripartido/metabolismo , Proteínas Virais/metabolismo , Linhagem Celular , Replicação do DNA/fisiologia , DNA Viral/genética , Proteínas de Ligação a DNA/genética , Dependovirus/genética , Epigênese Genética , Genoma Viral , Células HEK293 , Células HeLa , Humanos , Infecções por Parvoviridae/metabolismo , Infecções por Parvoviridae/virologia , Receptores de Neuropeptídeo Y/metabolismo , Proteínas Virais/genética , Vírion/metabolismo , Latência Viral , Replicação Viral/fisiologia
4.
EMBO Rep ; 19(10)2018 10.
Artigo em Inglês | MEDLINE | ID: mdl-30061100

RESUMO

Endogenous retroviruses (ERVs) have accumulated in vertebrate genomes and contribute to the complexity of gene regulation. KAP1 represses ERVs during development by its recruitment to their repetitive sequences through KRAB zinc-finger proteins (KZNFs), but little is known about the regulation of ERVs in adult tissues. We observed that KAP1 repression of HERVK14C was conserved in differentiated human cells and performed KAP1 knockout to obtain an overview of KAP1 function. Our results show that KAP1 represses ERVs (including HERV-T and HERV-S) and ZNF genes, both of which overlap with KAP1 binding sites and H3K9me3 in multiple cell types. Furthermore, this pathway is functionally conserved in adult human peripheral blood mononuclear cells. Cytosine methylation that acts on KAP1 regulated loci is necessary to prevent an interferon response, and KAP1-depletion leads to activation of some interferon-stimulated genes. Finally, loss of KAP1 leads to a decrease in H3K9me3 enrichment at ERVs and ZNF genes and an RNA-sensing response mediated through MAVS signaling. These data indicate that the KAP1-KZNF pathway contributes to genome stability and innate immune control in adult human cells.


Assuntos
Retrovirus Endógenos/genética , Imunidade Inata/genética , Proteínas Repressoras/genética , Proteína 28 com Motivo Tripartido/genética , Sítios de Ligação/genética , Metilação de DNA/genética , Retrovirus Endógenos/imunologia , Retrovirus Endógenos/patogenicidade , Regulação da Expressão Gênica/imunologia , Técnicas de Inativação de Genes , Genoma Humano/imunologia , Histonas/genética , Histonas/imunologia , Humanos , Leucócitos Mononucleares/imunologia , Leucócitos Mononucleares/virologia , Regiões Promotoras Genéticas
5.
Nature ; 487(7405): 57-63, 2012 Jul 05.
Artigo em Inglês | MEDLINE | ID: mdl-22722858

RESUMO

Embryonic stem (ES) cells are derived from blastocyst-stage embryos and are thought to be functionally equivalent to the inner cell mass, which lacks the ability to produce all extraembryonic tissues. Here we identify a rare transient cell population within mouse ES and induced pluripotent stem (iPS) cell cultures that expresses high levels of transcripts found in two-cell (2C) embryos in which the blastomeres are totipotent. We genetically tagged these 2C-like ES cells and show that they lack the inner cell mass pluripotency proteins Oct4 (also known as Pou5f1), Sox2 and Nanog, and have acquired the ability to contribute to both embryonic and extraembryonic tissues. We show that nearly all ES cells cycle in and out of this privileged state, which is partially controlled by histone-modifying enzymes. Transcriptome sequencing and bioinformatic analyses showed that many 2C transcripts are initiated from long terminal repeats derived from endogenous retroviruses, suggesting this foreign sequence has helped to drive cell-fate regulation in placental mammals.


Assuntos
Desdiferenciação Celular/genética , Células-Tronco Embrionárias/citologia , Células-Tronco Embrionárias/metabolismo , Retrovirus Endógenos/genética , Células-Tronco Pluripotentes/citologia , Células-Tronco Totipotentes/citologia , Células-Tronco Totipotentes/metabolismo , Animais , Desdiferenciação Celular/fisiologia , Linhagem da Célula/genética , Quimera/embriologia , Cromatina/genética , Cromatina/metabolismo , Embrião de Mamíferos/citologia , Embrião de Mamíferos/metabolismo , Embrião de Mamíferos/virologia , Células-Tronco Embrionárias/virologia , Epigênese Genética , Feminino , Regulação da Expressão Gênica no Desenvolvimento , Genes Reporter/genética , Histonas/química , Histonas/metabolismo , Células-Tronco Pluripotentes Induzidas/citologia , Células-Tronco Pluripotentes Induzidas/metabolismo , Lisina/química , Lisina/metabolismo , Metilação , Camundongos , Fenótipo , Células-Tronco Pluripotentes/metabolismo , Células-Tronco Pluripotentes/virologia , Sequências Repetidas Terminais/genética , Células-Tronco Totipotentes/virologia , Transcriptoma/genética
6.
Genome Res ; 24(8): 1251-9, 2014 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-24879558

RESUMO

Endogenous retroelements (EREs) account for about half of the mouse or human genome, and their potential as insertional mutagens and transcriptional perturbators is suppressed by early embryonic epigenetic silencing. Here, we asked how ERE control is maintained during the generation of induced pluripotent stem cells (iPSCs), as this procedure involves profound epigenetic remodeling. We found that all EREs tested were markedly up-regulated during the reprogramming of either mouse embryonic fibroblasts, human CD34(+) cells, or human primary hepatocytes. At the iPSC stage, EREs of some classes were repressed, whereas others remained highly expressed, yielding a pattern somewhat reminiscent of that recorded in embryonic stem cells. However, variability persisted between individual iPSC clones in the control of specific ERE integrants. Both during reprogramming and in iPS cells, the up-regulation of specific EREs significantly impacted on the transcription of nearby cellular genes. While transcription triggered by specific ERE integrants at highly precise developmental stages may be an essential step toward obtaining pluripotent cells, the broad and unspecific unleashing of the repetitive genome observed here may contribute to the inefficiency of the reprogramming process and to the phenotypic heterogeneity of iPSCs.


Assuntos
Retrovirus Endógenos/genética , Células-Tronco Pluripotentes Induzidas/fisiologia , Transcriptoma , Animais , Células Cultivadas , Reprogramação Celular , Inativação Gênica , Humanos , Camundongos , Regulação para Cima
7.
Genome Res ; 23(3): 452-61, 2013 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-23233547

RESUMO

TRIM28 is critical for the silencing of endogenous retroviruses (ERVs) in embryonic stem (ES) cells. Here, we reveal that an essential impact of this process is the protection of cellular gene expression in early embryos from perturbation by cis-acting activators contained within these retroelements. In TRIM28-depleted ES cells, repressive chromatin marks at ERVs are replaced by histone modifications typical of active enhancers, stimulating transcription of nearby cellular genes, notably those harboring bivalent promoters. Correspondingly, ERV-derived sequences can repress or enhance expression from an adjacent promoter in transgenic embryos depending on their TRIM28 sensitivity in ES cells. TRIM28-mediated control of ERVs is therefore crucial not just to prevent retrotransposition, but more broadly to safeguard the transcriptional dynamics of early embryos.


Assuntos
Células-Tronco Embrionárias/metabolismo , Proteínas Nucleares/metabolismo , Proteínas Repressoras/metabolismo , Retroelementos , Transcrição Gênica , Animais , Cromatina/genética , Cromatina/metabolismo , Mapeamento Cromossômico , Metilação de DNA , Células-Tronco Embrionárias/citologia , Células-Tronco Embrionárias/virologia , Retrovirus Endógenos/genética , Deleção de Genes , Regulação da Expressão Gênica no Desenvolvimento , Inativação Gênica , Loci Gênicos , Histonas/genética , Histonas/metabolismo , Camundongos , Proteínas Nucleares/genética , Regiões Promotoras Genéticas , Proteínas Repressoras/genética , Análise de Sequência de RNA , Proteína 28 com Motivo Tripartido , Regulação para Cima
8.
Development ; 140(3): 519-29, 2013 Feb 01.
Artigo em Inglês | MEDLINE | ID: mdl-23293284

RESUMO

Endogenous retroviruses (ERVs) undergo de novo DNA methylation during the first few days of mammalian embryogenesis, although the factors that control the targeting of this process are largely unknown. We asked whether KAP1 (KRAB-associated protein 1) is involved in this mechanism because of its previously defined role in maintaining the silencing of ERVs through the histone methyltransferase ESET and histone H3 lysine 9 trimethylation. Here, we demonstrate that introduced ERV sequences are sufficient to direct rapid de novo methylation of a flanked promoter in embryonic stem (ES) cells. This mechanism requires the presence of an ERV sequence-recognizing KRAB zinc-finger protein (ZFP) and both KAP1 and ESET. Furthermore, this process can also take place on a strong cellular promoter and leads to methylation signatures that are subsequently maintained in vivo throughout embryogenesis. Finally, we show that methylation of ERVs residing in the genome is affected by knockout of KAP1 in early embryos. KRAB-ZFPs, KAP1 and ESET are thus likely to be responsible for the early embryonic instatement of stable epigenetic marks at ERV-containing loci.


Assuntos
Metilação de DNA , DNA Viral/metabolismo , Retrovirus Endógenos/metabolismo , Histona-Lisina N-Metiltransferase/metabolismo , Proteínas Nucleares/metabolismo , Proteínas Repressoras/metabolismo , Animais , Animais Geneticamente Modificados , DNA Viral/genética , Embrião de Mamíferos/citologia , Embrião de Mamíferos/metabolismo , Embrião de Mamíferos/virologia , Células-Tronco Embrionárias/citologia , Células-Tronco Embrionárias/metabolismo , Células-Tronco Embrionárias/virologia , Retrovirus Endógenos/enzimologia , Retrovirus Endógenos/genética , Regulação da Expressão Gênica no Desenvolvimento , Técnicas de Inativação de Genes , Inativação Gênica , Técnicas de Transferência de Genes , Vetores Genéticos/genética , Vetores Genéticos/metabolismo , Células HEK293 , Histona-Lisina N-Metiltransferase/genética , Histonas/genética , Histonas/metabolismo , Humanos , Lentivirus/genética , Lentivirus/metabolismo , Camundongos , Proteínas Nucleares/genética , Regiões Promotoras Genéticas , Proteínas Repressoras/genética , Transcriptoma , Transfecção , Proteína 28 com Motivo Tripartido
9.
Nature ; 463(7278): 237-40, 2010 Jan 14.
Artigo em Inglês | MEDLINE | ID: mdl-20075919

RESUMO

More than forty per cent of the mammalian genome is derived from retroelements, of which about one-quarter are endogenous retroviruses (ERVs). Some are still active, notably in mice the highly polymorphic early transposon (ETn)/MusD and intracisternal A-type particles (IAP). ERVs are transcriptionally silenced during early embryogenesis by histone and DNA methylation (and reviewed in ref. 7), although the initiators of this process, which is essential to protect genome integrity, remain largely unknown. KAP1 (KRAB-associated protein 1, also known as tripartite motif-containing protein 28, TRIM28) represses genes by recruiting the histone methyltransferase SETDB1, heterochromatin protein 1 (HP1) and the NuRD histone deacetylase complex, but few of its physiological targets are known. Two lines of evidence suggest that KAP1-mediated repression could contribute to the control of ERVs: first, KAP1 can trigger permanent gene silencing during early embryogenesis, and second, a KAP1 complex silences the retrovirus murine leukaemia virus in embryonic cells. Consistent with this hypothesis, here we show that KAP1 deletion leads to a marked upregulation of a range of ERVs, in particular IAP elements, in mouse embryonic stem (ES) cells and in early embryos. We further demonstrate that KAP1 acts synergistically with DNA methylation to silence IAP elements, and that it is enriched at the 5' untranslated region (5'UTR) of IAP genomes, where KAP1 deletion leads to the loss of histone 3 lysine 9 trimethylation (H3K9me3), a hallmark of KAP1-mediated repression. Correspondingly, IAP 5'UTR sequences can impose in cis KAP1-dependent repression on a heterologous promoter in ES cells. Our results establish that KAP1 controls endogenous retroelements during early embryonic development.


Assuntos
Células-Tronco Embrionárias/metabolismo , Retrovirus Endógenos/genética , Inativação Gênica , Genes de Partícula A Intracisternal/genética , Proteínas Nucleares/metabolismo , Proteínas Repressoras/metabolismo , Regiões 5' não Traduzidas/genética , Acetilação , Animais , Metilação de DNA , Embrião de Mamíferos/metabolismo , Embrião de Mamíferos/virologia , Células-Tronco Embrionárias/virologia , Fibroblastos , Genes Reporter , Proteínas de Fluorescência Verde/genética , Proteínas de Fluorescência Verde/metabolismo , Histonas/metabolismo , Vírus da Leucemia Murina/genética , Vírus da Leucemia Murina/fisiologia , Lisina/metabolismo , Metilação , Camundongos , Proteínas Nucleares/deficiência , Proteínas Nucleares/genética , Regiões Promotoras Genéticas/genética , Proteínas Repressoras/deficiência , Proteínas Repressoras/genética , Proteína 28 com Motivo Tripartido
10.
Retrovirology ; 12: 45, 2015 May 29.
Artigo em Inglês | MEDLINE | ID: mdl-26021318

RESUMO

Over half of our genome is composed of retrotransposons, which are mobile elements that can readily amplify their copy number by replicating through an RNA intermediate. Most of these elements are no longer mobile but still contain regulatory sequences that can serve as promoters, enhancers or repressors for cellular genes. Despite dominating our genetic content, little is known about the precise functions of retrotransposons, which include both endogenous retroviruses (ERVs) and non-LTR elements like long interspersed nuclear element 1 (LINE-1). However, a few recent cutting-edge publications have illustrated how retrotransposons shape species-specific stem cell gene expression by two opposing mechanisms, involving their recruitment of stem cell-enriched transcription factors (TFs): firstly, they can activate expression of genes linked to naïve pluripotency, and secondly, they can induce repression of proximal genes. The paradox that different retrotransposons are active or silent in embryonic stem cells (ESCs) can be explained by differences between retrotransposon families, between individual copies within the same family, and between subpopulations of ESCs. Since they have coevolved with their host genomes, some of them have been co-opted to perform species-specific beneficial functions, while others have been implicated in genetic disease. In this review, we will discuss retrotransposon functions in ESCs, focusing on recent mechanistic advances of how HERV-H has been adopted to preserve human naïve pluripotency and how particular LINE-1, SVA and ERV family members recruit species-specific transcriptional repressors. This review highlights the fine balance between activation and repression of retrotransposons that exists to harness their ability to drive evolution, while minimizing the risk they pose to genome integrity.


Assuntos
Células-Tronco Embrionárias/fisiologia , Regulação da Expressão Gênica , Retroelementos , Humanos
11.
Cell Rep ; 42(6): 112625, 2023 06 27.
Artigo em Inglês | MEDLINE | ID: mdl-37294634

RESUMO

Endogenous retroviruses (ERVs) have rewired host gene networks. To explore the origins of co-option, we employed an active murine ERV, IAPEz, and an embryonic stem cell (ESC) to neural progenitor cell (NPC) differentiation model. Transcriptional silencing via TRIM28 maps to a 190 bp sequence encoding the intracisternal A-type particle (IAP) signal peptide, which confers retrotransposition activity. A subset of "escapee" IAPs (∼15%) exhibits significant genetic divergence from this sequence. Canonical repressed IAPs succumb to a previously undocumented demarcation by H3K9me3 and H3K27me3 in NPCs. Escapee IAPs, in contrast, evade repression in both cell types, resulting in their transcriptional derepression, particularly in NPCs. We validate the enhancer function of a 47 bp sequence within the U3 region of the long terminal repeat (LTR) and show that escapee IAPs convey an activating effect on nearby neural genes. In sum, co-opted ERVs stem from genetic escapees that have lost vital sequences required for both TRIM28 restriction and autonomous retrotransposition.


Assuntos
Retrovirus Endógenos , Proteína 28 com Motivo Tripartido , Animais , Camundongos , Diferenciação Celular , Células-Tronco Embrionárias/metabolismo , Retrovirus Endógenos/genética , Retrovirus Endógenos/metabolismo , Histonas/metabolismo , Proteína 28 com Motivo Tripartido/metabolismo , Sequências Repetidas Terminais/genética
12.
Sci Adv ; 8(43): eabp8085, 2022 Oct 28.
Artigo em Inglês | MEDLINE | ID: mdl-36306355

RESUMO

Mammalian genomes are a battleground for genetic conflict between repetitive elements and KRAB-zinc finger proteins (KZFPs). We asked whether KZFPs can regulate cell fate by using ZFP819, which targets a satellite DNA array, ZP3AR. ZP3AR coats megabase regions of chromosome 7 encompassing genes encoding ZSCAN4, a master transcription factor of totipotency. Depleting ZFP819 in mouse embryonic stem cells (mESCs) causes them to transition to a 2-cell (2C)-like state, whereby the ZP3AR array switches from a poised to an active enhancer state. This is accompanied by a global erosion of heterochromatin roadblocks, which we link to decreased SETDB1 stability. These events result in transcription of active LINE-1 elements and impaired differentiation. In summary, ZFP819 and TRIM28 partner up to close chromatin across Zscan4, to promote exit from totipotency. We propose that satellite DNAs may control developmental fate transitions by barcoding and switching off master transcription factor genes.


Assuntos
DNA Satélite , Proteínas Repressoras , Animais , Camundongos , DNA Satélite/genética , Mamíferos/genética , Análise de Sequência com Séries de Oligonucleotídeos , Proteínas Repressoras/metabolismo , Fatores de Transcrição/genética , Cromossomos
13.
J Virol ; 83(4): 1555-62, 2009 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-19036811

RESUMO

Lentiviral vectors deliver antigens to dendritic cells (DCs) in vivo, but they do not trigger DC maturation. We therefore expressed a viral protein that constitutively activates NF-kappaB, vFLIP from Kaposi's sarcoma-associated herpesvirus (KSHV), in a lentivector to mature DCs. vFLIP activated NF-kappaB in mouse bone marrow-derived DCs in vitro and matured these DCs to a similar extent as lipopolysaccharide; costimulatory markers CD80, CD86, CD40, and ICAM-1 were upregulated and tumor necrosis factor alpha and interleukin-12 secreted. The vFLIP-expressing lentivector also matured DCs in vivo. When we coexpressed vFLIP in a lentivector with ovalbumin (Ova), we found an increased immune response to Ova; up to 10 times more Ova-specific CD8(+) T cells secreting gamma interferon were detected in the spleens of vFLIP_Ova-immunized mice than in the spleens of mice immunized with GFP_Ova. Furthermore, this increased CD8(+) T-cell response correlated with improved tumor-free survival in a tumor therapy model. A single immunization with vFLIP_Ova also reduced the parasite load when mice were challenged with OVA-Leishmania donovani. In conclusion, vFLIP from KSHV is a DC activator, maturing DCs in vitro and in vivo. This demonstrates that NF-kappaB activation is sufficient to induce many aspects of DC maturation and that expression of a constitutive NF-kappaB activator can improve the efficacy of a vaccine vector.


Assuntos
Linfócitos T CD8-Positivos/imunologia , Células Dendríticas/virologia , Lentivirus/genética , NF-kappa B/biossíntese , Proteínas Virais/imunologia , Vacinas Virais/genética , Animais , Vacinas Anticâncer/imunologia , Citocinas/biossíntese , Leishmania donovani/imunologia , Leishmaniose/prevenção & controle , Lentivirus/imunologia , Camundongos , Neoplasias/imunologia , Ovalbumina/imunologia , Receptores Imunológicos/biossíntese , Baço/imunologia , Análise de Sobrevida , Proteínas Virais/genética , Vacinas Virais/imunologia
14.
Mol Ther ; 17(9): 1643-50, 2009 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-19584812

RESUMO

Lentiviral vectors (LVs) are tools for in vivo gene delivery, to correct genetic defects or to deliver antigens for vaccination. It was reported that systemic injection of LVs in mice transduced cells in liver and spleen. Here we describe the reasons for, and consequences of, persistent gene expression in spleen. After 5 days of intravenous injection, a green fluorescence protein (GFP)-expressing LV was detected in lymphocytes, macrophages and all subsets of dendritic cells (DCs) in spleen. In the case of macrophages and DCs, the percentage of transduced cells increased between 5 and 30 days after injection. We used bromodeoxyuridine (BrdU) incorporation to show that the macrophages were largely nondividing, whereas the transduced DCs arose from dividing precursor cells and could be detected in spleen 2 months after injection. Expression of ovalbumin (OVA) in the LV reduced the number of transduced DCs in spleen after 30 days. However, the remaining transduced cells stimulated proliferation and activation of OVA-specific CD8(+) T cells transferred 2 months after LV injection. The mice also maintained cytolytic activity against OVA-pulsed targets. These results show that LVs transduce DC precursors, which maintain transduced DCs in spleen for at least 2 months, leading to prolonged antigen presentation and effective T-cell memory.


Assuntos
Apresentação de Antígeno/imunologia , Células Dendríticas/imunologia , Células Dendríticas/metabolismo , Vetores Genéticos/genética , Imunização/métodos , Lentivirus/genética , Transdução Genética/métodos , Animais , Apresentação de Antígeno/genética , Diferenciação Celular , Células Dendríticas/citologia , Linfócitos/citologia , Linfócitos/imunologia , Linfócitos/metabolismo , Macrófagos/citologia , Macrófagos/imunologia , Macrófagos/metabolismo , Camundongos , Reação em Cadeia da Polimerase , Baço/citologia , Baço/metabolismo
15.
Viruses ; 12(12)2020 12 16.
Artigo em Inglês | MEDLINE | ID: mdl-33339171

RESUMO

As guest editors, we are pleased to present this Special Issue on endogenous retroviruses (ERVs) and their impact on mammalian development and disease [...].


Assuntos
Suscetibilidade a Doenças , Desenvolvimento Embrionário , Retrovirus Endógenos , Animais , Humanos
16.
Viruses ; 12(10)2020 09 26.
Artigo em Inglês | MEDLINE | ID: mdl-32993145

RESUMO

The human genome has been under selective pressure to evolve in response to emerging pathogens and other environmental challenges. Genome evolution includes the acquisition of new genes or new isoforms of genes and changes to gene expression patterns. One source of genome innovation is from transposable elements (TEs), which carry their own promoters, enhancers and open reading frames and can act as 'controlling elements' for our own genes. TEs include LINE-1 elements, which can retrotranspose intracellularly and endogenous retroviruses (ERVs) that represent remnants of past retroviral germline infections. Although once pathogens, ERVs also represent an enticing source of incoming genetic material that the host can then repurpose. ERVs and other TEs have coevolved with host genes for millions of years, which has allowed them to become embedded within essential gene expression programmes. Intriguingly, these host genes are often subject to the same epigenetic control mechanisms that evolved to combat the TEs that now regulate them. Here, we illustrate the breadth of host gene regulation through TEs by focusing on examples of young (The New), ancient (The Old), and disease-causing (The Ugly) TE integrants.


Assuntos
Elementos de DNA Transponíveis/genética , Regulação da Expressão Gênica/genética , Elementos Nucleotídeos Longos e Dispersos/genética , Sequências Reguladoras de Ácido Nucleico/genética , Retrovirus Endógenos/genética , Epigênese Genética/genética , Humanos , Regiões Promotoras Genéticas/genética
17.
Nat Commun ; 11(1): 5387, 2020 11 03.
Artigo em Inglês | MEDLINE | ID: mdl-33144593

RESUMO

The Human Silencing Hub (HUSH) complex is necessary for epigenetic repression of LINE-1 elements. We show that HUSH-depletion in human cell lines and primary fibroblasts leads to induction of interferon-stimulated genes (ISGs) through JAK/STAT signaling. This effect is mainly attributed to MDA5 and RIG-I sensing of double-stranded RNAs (dsRNAs). This coincides with upregulation of primate-conserved LINE-1s, as well as increased expression of full-length hominid-specific LINE-1s that produce bidirectional RNAs, which may form dsRNA. Notably, LTRs nearby ISGs are derepressed likely rendering these genes more responsive to interferon. LINE-1 shRNAs can abrogate the HUSH-dependent response, while overexpression of an engineered LINE-1 construct activates interferon signaling. Finally, we show that the HUSH component, MPP8 is frequently downregulated in diverse cancers and that its depletion leads to DNA damage. These results suggest that LINE-1s may drive physiological or autoinflammatory responses through dsRNA sensing and gene-regulatory roles and are controlled by the HUSH complex.


Assuntos
Epigênese Genética/fisiologia , Regulação Neoplásica da Expressão Gênica , Inativação Gênica/fisiologia , Interferon Tipo I/metabolismo , Elementos Nucleotídeos Longos e Dispersos/fisiologia , Proteína DEAD-box 58/genética , Proteína DEAD-box 58/metabolismo , Dano ao DNA , Regulação para Baixo , Técnicas de Inativação de Genes , Células HEK293 , Células HeLa , Humanos , Inflamação , Helicase IFIH1 Induzida por Interferon/metabolismo , Elementos Nucleotídeos Longos e Dispersos/genética , Fosfoproteínas/metabolismo , RNA de Cadeia Dupla , Receptores Imunológicos , Análise de Sequência de RNA , Transdução de Sinais
18.
Curr Opin Virol ; 25: 28-33, 2017 08.
Artigo em Inglês | MEDLINE | ID: mdl-28675832

RESUMO

Retrotransposons tune immune reactivity in differentiated cells because when they are transcribed, their nucleic acids can be viewed as non-self leading to innate immune sensing. Most retrotransposons, however, are subject to transcriptional regulation by a multitude of epigenetic pathways, which have coevolved with them for millions of years. While a lot is known about the epigenetic control of retrotransposons in germ cells and early embryos, surprisingly little is understood about these pathways in adult tissues, particularly in human cells. Recent evidence suggests that retrotransposon repression persists in differentiated cells and is dynamic. Future insight into this topic may teach us how to reactivate or silence specific retrotransposon families, to promote anti-tumor immunity or dampen autoimmunity through epigenetic modulation.


Assuntos
Diferenciação Celular , Epigênese Genética , Genoma Humano , Imunidade , Retroelementos/genética , Adulto , Animais , Regulação da Expressão Gênica , Humanos , Camundongos , Retroelementos/imunologia , Transcrição Gênica
19.
J Cell Biol ; 216(11): 3429-3431, 2017 11 06.
Artigo em Inglês | MEDLINE | ID: mdl-29066607

RESUMO

Cancer cells thrive on genetic and epigenetic changes that confer a selective advantage but also need strategies to avoid immune recognition. In this issue, Cuellar et al. (2017. J. Cell Biol https://doi.org/10.1083/jcb.201612160) find that the histone methyltransferase SETDB1 enables acute myeloid leukemia cells to evade sensing of retrotransposons by innate immune receptors.


Assuntos
Interferons , Leucemia Mieloide Aguda , Epigênese Genética , Código das Histonas , Histona-Lisina N-Metiltransferase , Humanos , Proteínas Metiltransferases , Retroelementos
20.
Methods Mol Biol ; 1400: 403-17, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-26895067

RESUMO

Deposition of epigenetic marks is an important layer of the transcriptional control of retrotransposons, especially during early embryogenesis. Krüppel-associated box domain zinc finger proteins (KRAB-ZFPs) are one of the largest families of transcription factors, and collectively partake in this process by tethering to thousands of retroelement-containing genomic loci their cofactor KAP1, which acts as a scaffold for a heterochromatin-inducing machinery. However, while the sequence-specific DNA binding potential of the poly-zinc finger-containing KRAB-ZFPs is recognized, very few members of the family have been assigned specific targets. In this chapter, we describe a large-scale functional screen to identify the retroelements bound by individual murine KRAB-ZFPs. Our method is based on the automated transfection of a library of mouse KRAB-ZFP-containing vectors into 293T cells modified to express GFP from a PGK promoter harboring in its immediate vicinity a KAP1-recruiting retroelement-derived sequence. Analysis is then performed by plate reader and flow cytometry fluorescence readout. Such large-scale DNA-centered functional approach can not only help to identify the trans-acting factors responsible for silencing retrotransposons, but also serve as a model for dissecting the transcriptional networks influenced by retroelement-derived cis-acting sequences.


Assuntos
Epigênese Genética , Epigenômica/métodos , Proteínas Repressoras/metabolismo , Retroelementos , Animais , Linhagem Celular , Clonagem Molecular , Regulação da Expressão Gênica , Biblioteca Gênica , Humanos , Camundongos , Ligação Proteica , Proteínas Repressoras/genética , Fatores de Transcrição/genética , Fatores de Transcrição/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA