Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 63
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
Int J Mol Sci ; 21(7)2020 Mar 31.
Artigo em Inglês | MEDLINE | ID: mdl-32244497

RESUMO

Paternally expressed gene 10 (PEG10) is a human retrotransposon-derived imprinted gene. The mRNA of PEG10 encodes two protein isoforms: the Gag-like protein (RF1PEG10) is coded by reading frame 1, while the Gag-Pol-like polyprotein (RF1/RF2PEG10) is coded by reading frames 1 and 2. The proteins are translated by a typical retroviral frameshift mechanism. The protease (PR) domain of RF2PEG10 contains an -Asp-Ser-Gly- sequence, which corresponds to the consensus -Asp-Ser/Thr-Gly- active-site motif of retroviral aspartic proteases. The function of the aspartic protease domain of RF2PEG10 remains unclear. To elucidate the function of PEG10 protease (PRPEG10), we designed a frameshift mutant (fsRF1/RF2PEG10) for comparison with the RF1/RF2PEG10 form. To study the effects of PRPEG10 on cellular proliferation and viability, mammalian HEK293T and HaCaT cells were transfected with plasmids coding for either RF1/RF2PEG10, the frameshift mutant (fsRF1/RF2PEG10), or a PR active-site (D370A) mutant fsRF1/RF2PEG10. Our results indicate that fsRF1/RF2PEG10 overexpression results in increased cellular proliferation. Remarkably, transfection with fsRF1/RF2PEG10 had a detrimental effect on cell viability. We hypothesize that PRPEG10 plays an important role in the function of this retroviral remnant, mediating the proliferation of cells and possibly implicating it in the inhibition of apoptosis.


Assuntos
Proteínas Reguladoras de Apoptose/genética , Proteínas Reguladoras de Apoptose/metabolismo , Proteínas de Ligação a DNA/genética , Proteínas de Ligação a DNA/metabolismo , Peptídeo Hidrolases/metabolismo , Proteínas de Ligação a RNA/genética , Proteínas de Ligação a RNA/metabolismo , Retroelementos/fisiologia , Ácido Aspártico Endopeptidases/genética , Proliferação de Células , Sobrevivência Celular , Mutação da Fase de Leitura , Células HEK293 , Células HaCaT , Humanos , Peptídeo Hidrolases/genética , Isoformas de Proteínas , Fases de Leitura , Proteínas Recombinantes , Alinhamento de Sequência , Transfecção
2.
Biochem Pharmacol ; 175: 113904, 2020 05.
Artigo em Inglês | MEDLINE | ID: mdl-32156659

RESUMO

Breast cancer is the most common cancer type in females worldwide. Environmental exposure to pesticides affecting hormonal homeostasis does not necessarily induce DNA mutations but may influence gene expression by disturbances in epigenetic regulation. Expression of long interspersed nuclear element-1 (LINE-1) has been associated with tumorigenesis in several cancers. In nearly all somatic cells, LINE-1 is silenced by DNA methylation in the 5́'UTR and reactivated during disease initiation and/or progression. Strong ligands of aryl hydrocarbon receptor (AhR) activate LINE-1 through the transforming growth factor-ß1 (TGF-ß1)/Smad pathway. Hexachlorobenzene (HCB) and chlorpyrifos (CPF), both weak AhR ligands, promote cell proliferation and migration in breast cancer cells, as well as tumor growth in rat models. In this context, our aim was to examine the effect of these pesticides on LINE-1 expression and ORF1p localization in the triple-negative breast cancer cell line MDA-MB-231 and the non-tumorigenic epithelial breast cell line NMuMG, and to evaluate the role of TGF-ß1 and AhR pathways. Results show that 0.5 µM CPF and 0.005 µM HCB increased LINE-1 mRNA expression through Smad and AhR signaling in MDA-MB-231. In addition, the methylation of the first sites in 5́'UTR of LINE-1 was reduced by pesticide exposure, although the farther sites remained unaffected. Pesticides modulated ORF1p localization in MDA-MB-231: 0.005 µM HCB and 50 µM CPF increased nuclear translocation, while both induced cytoplasmic retention at 0.5 and 5 µM. Moreover, both stimulated double-strand breaks, enhancing H2AX phosphorylation, coincidentally with ORF1p nuclear localization. In NMuMG similar results were observed, since they heighten LINE-1 mRNA levels. CPF effect was through AhR and TGF-ß1 signaling, whereas HCB action depends only of AhR. In addition, both pesticides increase ORF1p expression and nuclear localization. Our results provide experimental evidence that HCB and CPF exposure modify LINE-1 methylation levels and induce LINE-1 reactivation, suggesting that epigenetic mechanisms could contribute to pesticide-induced breast cancer progression.


Assuntos
Fatores de Transcrição Hélice-Alça-Hélice Básicos/metabolismo , Células Epiteliais/metabolismo , Elementos Nucleotídeos Longos e Dispersos/fisiologia , Receptores de Hidrocarboneto Arílico/metabolismo , Retroelementos/fisiologia , Neoplasias de Mama Triplo Negativas/metabolismo , Linhagem Celular Tumoral , Relação Dose-Resposta a Droga , Células Epiteliais/efeitos dos fármacos , Células Epiteliais/patologia , Feminino , Hexaclorobenzeno/metabolismo , Hexaclorobenzeno/toxicidade , Humanos , Ligantes , Elementos Nucleotídeos Longos e Dispersos/efeitos dos fármacos , Retroelementos/efeitos dos fármacos , Neoplasias de Mama Triplo Negativas/patologia
3.
Annu Rev Pathol ; 15: 51-70, 2020 01 24.
Artigo em Inglês | MEDLINE | ID: mdl-31977294

RESUMO

Our genome is a historic record of successive invasions of mobile genetic elements. Like other eukaryotes, we have evolved mechanisms to limit their propagation and minimize the functional impact of new insertions. Although these mechanisms are vitally important, they are imperfect, and a handful of retroelement families remain active in modern humans. This review introduces the intrinsic functions of transposons, the tactics employed in their restraint, and the relevance of this conflict to human pathology. The most straightforward examples of disease-causing transposable elements are germline insertions that disrupt a gene and result in a monogenic disease allele. More enigmatic are the abnormal patterns of transposable element expression in disease states. Changes in transposon regulation and cellular responses to their expression have implicated these sequences in diseases as diverse as cancer, autoimmunity, and neurodegeneration. Distinguishing their epiphenomenal from their pathogenic effects may provide wholly new perspectives on our understanding of disease.


Assuntos
Elementos de DNA Transponíveis/fisiologia , Doença/genética , Alelos , Animais , Autoimunidade/genética , Genoma Humano/fisiologia , Mutação em Linhagem Germinativa/fisiologia , Humanos , Mutagênese Insercional/fisiologia , Neoplasias/genética , Neoplasias/patologia , Degeneração Neural/genética , Degeneração Neural/patologia , Retroelementos/fisiologia
4.
Plant Sci ; 287: 110181, 2019 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-31481212

RESUMO

The flavonoid compound anthocyanin is an important plant metabolite with nutritional and aesthetic value as well as anti-oxidative capacity. MYB transcription factors are key regulators of anthocyanin biosynthesis in plants. In pepper (Capsicum annuum), the CaAn2 gene, encoding an R2R3 MYB transcription factor, regulates anthocyanin biosynthesis. However, no functional study or structural analysis of functional and dysfunctional CaAn2 alleles has been performed. Here, to elucidate the function of CaAn2, we generated transgenic Nicotiana benthamiana and Arabidopsis thaliana plants expressing CaAn2. All of the tissues in these plants were purple. Promoter analysis of CaAn2 in purple C. annuum 'KC00134' plants revealed the insertion of a non-long terminal repeat (LTR) retrotransposon designated Ca-nLTR-A. To determine the promoter activity and functional domain of Ca-nLTR-A, various constructs carrying different domains of Ca-nLTR-A fused with GUS were transformed into N. benthamiana. Promoter analysis showed that the 3' untranslated region (UTR) of the second open reading frame of Ca-nLTR-A is responsible for CaAn2 expression in 'KC00134'. Sequence analysis of Ca-nLTR-A identified transcription factor binding sites known to regulate anthocyanin biosynthesis. This study indicates that insertion of a non-LTR retrotransposon in the promoter may activate expression of CaAn2 by recruiting transcription factors at the 3' UTR and thus provides the first example of exaptation of a non-LTR retrotransposon into a new promoter in plants.


Assuntos
Antocianinas/biossíntese , Capsicum/metabolismo , Proteínas de Plantas/metabolismo , Retroelementos/fisiologia , Fatores de Transcrição/metabolismo , Antocianinas/metabolismo , Arabidopsis , Capsicum/genética , Clonagem Molecular , Regulação da Expressão Gênica de Plantas , Genes de Plantas/genética , Genes de Plantas/fisiologia , Proteínas de Plantas/genética , Plantas Geneticamente Modificadas , Regiões Promotoras Genéticas/genética , Retroelementos/genética , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Nicotiana , Técnicas do Sistema de Duplo-Híbrido
5.
Nat Genet ; 50(2): 186-192, 2018 02.
Artigo em Inglês | MEDLINE | ID: mdl-29335544

RESUMO

Chromosome dosage has substantial effects on reproductive isolation and speciation in both plants and animals, but the underlying mechanisms are largely obscure 1 . Transposable elements in animals can regulate hybridity through maternal small RNA 2 , whereas small RNAs in plants have been postulated to regulate dosage response via neighboring imprinted genes3,4. Here we show that a highly conserved microRNA in plants, miR845, targets the tRNAMet primer-binding site (PBS) of long terminal repeat (LTR) retrotransposons in Arabidopsis pollen, and triggers the accumulation of 21-22-nucleotide (nt) small RNAs in a dose-dependent fashion via RNA polymerase IV. We show that these epigenetically activated small interfering RNAs (easiRNAs) mediate hybridization barriers between diploid seed parents and tetraploid pollen parents (the 'triploid block'), and that natural variation for miR845 may account for 'endosperm balance' allowing the formation of triploid seeds. Targeting of the PBS with small RNA is a common mechanism for transposon control in mammals and plants, and provides a uniquely sensitive means to monitor chromosome dosage and imprinting in the developing seed.


Assuntos
Arabidopsis/genética , Mecanismo Genético de Compensação de Dose/genética , MicroRNAs/fisiologia , RNA de Plantas/genética , Retroelementos/fisiologia , Regulação da Expressão Gênica de Plantas , Genoma de Planta , MicroRNAs/genética , Poliploidia , Sequências Repetidas Terminais/genética
6.
Nucleic Acids Res ; 45(10): 6194-6208, 2017 Jun 02.
Artigo em Inglês | MEDLINE | ID: mdl-28334904

RESUMO

Short interspersed nuclear elements (SINEs) are retrotransposons evolutionarily derived from endogenous RNA Polymerase III RNAs. Though SINE elements have undergone exaptation into gene regulatory elements, how transcribed SINE RNA impacts transcriptional and post-transcriptional regulation is largely unknown. This is partly due to a lack of information regarding which of the loci have transcriptional potential. Here, we present an approach (short interspersed nuclear element sequencing, SINE-seq), which selectively profiles RNA Polymerase III-derived SINE RNA, thereby identifying transcriptionally active SINE loci. Applying SINE-seq to monitor murine B2 SINE expression during a gammaherpesvirus infection revealed transcription from 28 270 SINE loci, with ∼50% of active SINE elements residing within annotated RNA Polymerase II loci. Furthermore, B2 RNA can form intermolecular RNA-RNA interactions with complementary mRNAs, leading to nuclear retention of the targeted mRNA via a mechanism involving p54nrb. These findings illuminate a pathway for the selective regulation of mRNA export during stress via retrotransposon activation.


Assuntos
Regulação da Expressão Gênica/genética , RNA Mensageiro/genética , Elementos Nucleotídeos Curtos e Dispersos/genética , Animais , Transporte Biológico/genética , Proteínas de Ciclo Celular/genética , Técnicas de Silenciamento de Genes , Infecções por Herpesviridae/genética , Camundongos , Células NIH 3T3 , Proteínas Associadas à Matriz Nuclear/metabolismo , Interferência de RNA , RNA Polimerase III/genética , RNA Mensageiro/metabolismo , Proteínas de Ligação a RNA/metabolismo , Retroelementos/fisiologia , Rhadinovirus , Análise de Sequência de DNA , Estresse Fisiológico/genética , Frações Subcelulares/metabolismo , Transcrição Gênica , Infecções Tumorais por Vírus/genética
7.
Adv Immunol ; 132: 47-69, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-27769507

RESUMO

The ability to distinguish between self and nonself is the fundamental basis of the immune system in all organisms. The conceptual distinction between self and nonself, however, breaks down when it comes to endogenous retroviruses and other retroelements. While some retroelements retain the virus-like features including the capacity to replicate and reinvade the host genome, most have become inactive through mutations or host epigenetic silencing. And yet, accumulating evidence suggests that endogenous retroelements, both active and inactive, play important roles not only in pathogenesis of immune disorders, but also in proper functioning of the immune system. This review discusses the recent development in our understanding of the interaction between retroelements and the host innate immune system. In particular, it focuses on the impact of retroelement transcripts on the viral RNA sensors such as Toll-like receptors, RIG-I-like receptors, protein kinase R, and the inflammasomes.


Assuntos
Doenças do Sistema Imunitário/genética , Imunidade Inata , RNA Viral/genética , Retroelementos/fisiologia , Infecções por Retroviridae/imunologia , Retroviridae/fisiologia , Animais , Autoantígenos/imunologia , Epigênese Genética , Interações Hospedeiro-Patógeno , Humanos , Receptores de Reconhecimento de Padrão/metabolismo , Ativação Viral , Latência Viral
8.
Biochemistry (Mosc) ; 81(9): 1023-30, 2016 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-27682174

RESUMO

The telomere is a nucleoprotein complex at the ends of linear chromosomes that protects them from fusion and degradation. The telomere consists of telomeric DNA, a protective protein complex and telomeric RNA. Biogenesis of telomeric transcripts in development is still far from being understood. Drosophila telomeres are elongated by a transposition of specialized telomeric retrotransposons that encode proteins. Using transgenic constructs encoding tagged telomeric protein, we found that transcripts of Drosophila telomeric element HeT-A bind Gag-HeT-A protein encoded by these transcripts. Maternal HeT-A transcripts and Gag-HeT-A form ribonucleoprotein granules around centrosomes, centers of microtubule organization, during blastoderm formation, upon disruption of telomere silencing during oogenesis. The specific localization of HeT-A RNA is dependent on microtubules since disruption of microtubules caused delocalization of HeT-A transcripts. This transgenic system is a valuable model for the study of telomeric RNA biogenesis.


Assuntos
Centrossomo/metabolismo , Proteínas de Drosophila/metabolismo , Embrião não Mamífero/metabolismo , Desenvolvimento Embrionário/fisiologia , Produtos do Gene gag/metabolismo , Retroelementos/fisiologia , Telômero/metabolismo , Animais , Proteínas de Drosophila/genética , Drosophila melanogaster , Feminino , Produtos do Gene gag/genética , Telômero/genética
9.
Uirusu ; 66(1): 11-20, 2016.
Artigo em Japonês | MEDLINE | ID: mdl-28484174

RESUMO

In the human genome, there are approximately 30 LTR retrotransposon-derived genes, such as the sushi-ichi retrotransposon homologues (SIRH) and the paraneoplastic Ma antigen (PNMA) family genes. They are derivatives from the original LTR retrotransposons and each gene seems to have its own unique function. PEG10/SIRH1 as well as PEG11/RTL1/SIRH2 and SIRH7/LDOC1 play essential roles in placenta formation, maintenance of fetal capillaries and the differentiation/maturation of a variety of placental cells, respectively. All of this evidence provides strong support for their contribution to the evolution of viviparity in mammals via their eutherian-specific functions. SIRH11/ZCCHC16 is an X-linked gene that encodes a CCHC type of zinc-finger protein that exhibits high sequence identity to the LTR retrotransposon Gag protein and its deletion causes abnormal behavior related to cognition, including attention, impulsivity and working memory, possibly via the locus coeruleus noradrenaergic system in mice. Therefore, we have suggested that the acquisition of SIRH11/ZCCHC16 was involved in eutherian brain evolution. Interestingly, SIRH11/ZCCHC16 displays lineage-specific structural and putative species-specific functional variations in eutherians, suggesting that it contributed to the diversification of eutherians via increasing evolutionary fitness by these changes.


Assuntos
Encéfalo/fisiologia , Genoma Humano/genética , Retroelementos/genética , Animais , Atenção , Comportamento Animal , Evolução Biológica , Encéfalo/metabolismo , Cognição , Impressão Genômica/genética , Humanos , Camundongos , Camundongos Knockout , Norepinefrina/metabolismo , Retroelementos/fisiologia , Sequências Repetidas Terminais
10.
Genes Dev ; 30(1): 64-77, 2016 Jan 01.
Artigo em Inglês | MEDLINE | ID: mdl-26701264

RESUMO

Throughout the animal kingdom, p53 genes govern stress response networks by specifying adaptive transcriptional responses. The human member of this gene family is mutated in most cancers, but precisely how p53 functions to mediate tumor suppression is not well understood. Using Drosophila and zebrafish models, we show that p53 restricts retrotransposon activity and genetically interacts with components of the piRNA (piwi-interacting RNA) pathway. Furthermore, transposon eruptions occurring in the p53(-) germline were incited by meiotic recombination, and transcripts produced from these mobile elements accumulated in the germ plasm. In gene complementation studies, normal human p53 alleles suppressed transposons, but mutant p53 alleles from cancer patients could not. Consistent with these observations, we also found patterns of unrestrained retrotransposons in p53-driven mouse and human cancers. Furthermore, p53 status correlated with repressive chromatin marks in the 5' sequence of a synthetic LINE-1 element. Together, these observations indicate that ancestral functions of p53 operate through conserved mechanisms to contain retrotransposons. Since human p53 mutants are disabled for this activity, our findings raise the possibility that p53 mitigates oncogenic disease in part by restricting transposon mobility.


Assuntos
Genes p53/genética , Retroelementos/fisiologia , Proteína Supressora de Tumor p53/genética , Proteína Supressora de Tumor p53/metabolismo , Animais , Drosophila/genética , Feminino , Variação Genética , Humanos , Masculino , Camundongos , Mutação/genética , Neoplasias/genética , Retroelementos/genética , Peixe-Zebra/genética
11.
Med Sci (Paris) ; 30(6-7): 659-64, 2014.
Artigo em Francês | MEDLINE | ID: mdl-25014457

RESUMO

Transposable elements (TE) represent around 40% of the human genome. They are endogenous mobile DNA sequences able to jump and duplicate in the host genome. TE have long been considered as "junk" DNA but are now believed to be important regulators of gene expression by participating to the establishment of the DNA methylation profile. Recent advances in genome sequencing reveals a higher transposition frequency and TE driven gene expression in somatic cells than previously thought. As TE propagation is deleterious and may be involved in oncogenic mechanisms, host cells have developed silencing mechanisms mainly described in germinal and embryonic cells. However, somatic cells are also proned to TE transposition and use specific mechanisms involving tumor suppressor proteins including p53, Rb and PLZF. These transcription factors specifically target genomic retrotransposon sequences, histone deacetylase and DNA methylase activities, inducing epigenetic modifications related to gene silencing. Thus, these transcription factors negatively regulate TE expression by the formation of DNA methylation profil in somatic cells possibly associated with oncogenic mechanisms.


Assuntos
Epigênese Genética/genética , Sequências Repetitivas de Ácido Nucleico , Retroelementos/fisiologia , Animais , Metilação de DNA , Regulação da Expressão Gênica , Humanos , Elementos Nucleotídeos Longos e Dispersos , Neoplasias/genética
12.
Nat Neurosci ; 17(7): 923-33, 2014 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-24859200

RESUMO

Neuronal migration disorders such as lissencephaly and subcortical band heterotopia are associated with epilepsy and intellectual disability. DCX, PAFAH1B1 and TUBA1A are mutated in these disorders; however, corresponding mouse mutants do not show heterotopic neurons in the neocortex. In contrast, spontaneously arisen HeCo mice display this phenotype, and our study revealed that misplaced apical progenitors contribute to heterotopia formation. While HeCo neurons migrated at the same speed as wild type, abnormally distributed dividing progenitors were found throughout the cortical wall from embryonic day 13. We identified Eml1, encoding a microtubule-associated protein, as the gene mutated in HeCo mice. Full-length transcripts were lacking as a result of a retrotransposon insertion in an intron. Eml1 knockdown mimicked the HeCo progenitor phenotype and reexpression rescued it. We further found EML1 to be mutated in ribbon-like heterotopia in humans. Our data link abnormal spindle orientations, ectopic progenitors and severe heterotopia in mouse and human.


Assuntos
Coristoma/genética , Proteínas Associadas aos Microtúbulos/genética , Proteínas Associadas aos Microtúbulos/fisiologia , Mutação/fisiologia , Células-Tronco Neurais/fisiologia , Sequência de Aminoácidos , Animais , Bromodesoxiuridina , Ciclo Celular/fisiologia , Movimento Celular/fisiologia , Córtex Cerebral/citologia , Córtex Cerebral/embriologia , Lissencefalias Clássicas e Heterotopias Subcorticais em Banda , Proteína Duplacortina , Eletroporação , Humanos , Imuno-Histoquímica , Malformações Arteriovenosas Intracranianas/patologia , Íntrons/genética , Camundongos , Microscopia Confocal , Microtúbulos/fisiologia , Mitose/fisiologia , Dados de Sequência Molecular , Retroelementos/fisiologia , Fuso Acromático/fisiologia
13.
Cell Mol Life Sci ; 71(19): 3749-66, 2014 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-24854580

RESUMO

Infection of cells with human immunodeficiency virus 1 (HIV-1) is controlled by restriction factors, host proteins that counteract a variety of steps in the life cycle of this lentivirus. These include SAMHD1, APOBEC3G and tetherin, which block reverse transcription, hypermutate viral DNA and prevent progeny virus release, respectively. These and other HIV-1 restriction factors are conserved and have clear orthologues in the mouse. This review summarises studies in knockout mice lacking HIV-1 restriction factors. In vivo experiments in such animals have not only validated in vitro data obtained from cultured cells, but have also revealed new findings about the biology of these proteins. Indeed, genetic ablation of HIV-1 restriction factors in the mouse has provided evidence that restriction factors control retroviruses and other viruses in vivo and has led to new insights into the mechanisms by which these proteins counteract infection. For example, in vivo experiments in knockout mice demonstrate that virus control exerted by restriction factors can shape adaptive immune responses. Moreover, the availability of animals lacking restriction factors opens the possibility to study the function of these proteins in other contexts such as autoimmunity and cancer. Further in vivo studies of more recently identified HIV-1 restriction factors in gene targeted mice are, therefore, justified.


Assuntos
HIV-1/metabolismo , Animais , Membrana Celular/metabolismo , Citidina Desaminase/genética , Citidina Desaminase/metabolismo , Proteínas Ligadas por GPI/genética , Proteínas Ligadas por GPI/metabolismo , Humanos , Camundongos , Camundongos Knockout , Proteínas Monoméricas de Ligação ao GTP/genética , Proteínas Monoméricas de Ligação ao GTP/metabolismo , Retroelementos/fisiologia , Transcrição Reversa/fisiologia , Proteína 1 com Domínio SAM e Domínio HD , Proteínas Virais/metabolismo
14.
Mol Pharmacol ; 85(5): 747-57, 2014 May.
Artigo em Inglês | MEDLINE | ID: mdl-24569088

RESUMO

Canonically, opioids influence cells by binding to a G protein-coupled opioid receptor, initiating intracellular signaling cascades, such as protein kinase, phosphatidylinositol 3-kinase, and extracellular receptor kinase pathways. This results in several downstream effects, including decreased levels of the reduced form of glutathione (GSH) and elevated oxidative stress, as well as epigenetic changes, especially in retrotransposons and heterochromatin, although the mechanism and consequences of these actions are unclear. We characterized the acute and long-term influence of morphine on redox and methylation status (including DNA methylation levels) in cultured neuronal SH-SY5Y cells. Acting via µ-opioid receptors, morphine inhibits excitatory amino acid transporter type 3-mediated cysteine uptake via multiple signaling pathways, involving different G proteins and protein kinases in a temporal manner. Decreased cysteine uptake was associated with decreases in both the redox and methylation status of neuronal cells, as defined by the ratios of GSH to oxidized forms of glutathione and S-adenosylmethionine to S-adenosylhomocysteine levels, respectively. Further, morphine induced global DNA methylation changes, including CpG sites in long interspersed nuclear elements (LINE-1) retrotransposons, resulting in increased LINE-1 mRNA. Together, these findings illuminate the mechanism by which morphine, and potentially other opioids, can influence neuronal-cell redox and methylation status including DNA methylation. Since epigenetic changes are implicated in drug addiction and tolerance phenomenon, this study could potentially extrapolate to elucidate a novel mechanism of action for other drugs of abuse.


Assuntos
Cisteína/antagonistas & inibidores , Metilação de DNA/efeitos dos fármacos , Transportador 3 de Aminoácido Excitatório/antagonistas & inibidores , Morfina/farmacologia , Retroelementos/efeitos dos fármacos , Transcrição Gênica/efeitos dos fármacos , Linhagem Celular Tumoral , Cisteína/metabolismo , Metilação de DNA/fisiologia , Transportador 3 de Aminoácido Excitatório/fisiologia , Humanos , Oxirredução/efeitos dos fármacos , Retroelementos/fisiologia , Transcrição Gênica/fisiologia
15.
PLoS One ; 8(9): e75381, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-24058682

RESUMO

Telomeres from Drosophila appear to be very different from those of other organisms - in size and the mechanism of their maintenance. In the absence of the enzyme telomerase, Drosophila telomeres are maintained by retrotransposition of three elements, HeT-A, TART, and TAHRE, but details of their transposition mechanisms are not known. Here we characterized some biochemical characteristics of the HeT-A Gag protein encoded by the HeT-A element to understand this mechanism. The HeT-A Gag protein when overexpressed in S2 cells was localized to the nucleus but was resistant to high salt, detergents and nuclease extraction treatments. Analysis of the HeT-A Gag protein by tandem mass spectrophotometry revealed that serines 216 and 221 are phosphorylated. Substituting these serines with alanine or aspartic acid by site-directed mutagenesis did not result in any changes in HeT-A Gag translocation across the nucleus, suggesting that phosphorylation of these sites is not associated with HeT-A Gag translocation, but time course experiments showed that these phosphorylation sites are important for Gag-protein stability.


Assuntos
Núcleo Celular/metabolismo , Proteínas de Drosophila/metabolismo , Produtos do Gene gag/metabolismo , Retroelementos/fisiologia , Transporte Ativo do Núcleo Celular/fisiologia , Alanina/genética , Alanina/metabolismo , Substituição de Aminoácidos , Animais , Ácido Aspártico/genética , Ácido Aspártico/metabolismo , Linhagem Celular , Núcleo Celular/genética , Proteínas de Drosophila/genética , Drosophila melanogaster , Produtos do Gene gag/genética , Mutação de Sentido Incorreto , Fosforilação/fisiologia , Estabilidade Proteica , Espectrometria de Massas em Tandem
16.
Curr Biol ; 22(11): R432-7, 2012 Jun 05.
Artigo em Inglês | MEDLINE | ID: mdl-22677280

RESUMO

June 27, 1970 was a significant day for our understanding of both the flow of information in biological systems and the evolution of eukaryotic genomes as this was the day that Nature published back-to-back papers reporting the discovery of an enzyme that copies RNA into DNA. This soon became known as reverse transcriptase and the RNA tumour viruses in which it was detected were renamed retroviruses. The realisation that retroviruses can convert their genomic RNA into DNA provided a route by which they could integrate into the chromosomes of infected cells as Howard Temin and his colleagues had proposed some years earlier. At the time it was thought that the ability to copy RNA into DNA would be confined to retroviruses. One of the more startling outcomes of whole genome DNA sequencing has been the discovery that eukaryotes can have more reverse transcriptase genes than genes coding for any other protein, and that the largest single component of many eukaryotic genomes has been generated by reverse transcription.


Assuntos
Retroelementos/fisiologia , Animais , Evolução Molecular , Humanos , DNA Polimerase Dirigida por RNA/genética , DNA Polimerase Dirigida por RNA/metabolismo , Sequências Repetidas Terminais
17.
Biol Reprod ; 86(3): 88, 2012 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-22116807

RESUMO

The proper functioning of the placenta requires specific patterns of methylation and the appropriate regulation of retroelements, some of which have been co-opted by the genome for placental-specific gene expression. Our inquiry was initiated to determine the causes of the placental defects observed in crosses between two species of mouse, Mus musculus and Mus caroli. M. musculus × M. caroli fetuses are rarely carried to term, possibly as a result of genomic incompatibility in the placenta. Taking into account that placental dysplasia is observed in Peromyscus and other Mus hybrids, and that endogenous retroviruses are expressed in the placental transcriptome, we hypothesized that these placental defects could result, in part, from failure of the genome defense mechanism, DNA methylation, to regulate the expression of retroelements. Hybrid M. musculus × M. caroli embryos were produced by artificial insemination, and dysplastic placentas were subjected to microarray and methylation screens. Aberrant overexpression of an X-linked Mus retroelement in these hybrid placentas is consistent with local demethylation of this retroelement, concomitant with genome instability, disruption of gene regulatory pathways, and dysgenesis. We propose that the placenta is a specific site of control that is disrupted by demethylation and retroelement activation in interspecific hybridization that occur as a result of species incompatibility of methylation machinery. To our knowledge, the present data provide the first report of retroelement activation linked to decreased methylation in a eutherian hybrid system.


Assuntos
Quimera/genética , Metilação de DNA/genética , Camundongos Endogâmicos/genética , Camundongos/genética , Placentação/genética , Retroelementos/genética , Animais , Metilação de DNA/fisiologia , Feminino , Instabilidade Genômica/genética , Hibridização Genética/genética , Cariótipo , Modelos Animais , Placentação/fisiologia , Gravidez , Retroelementos/fisiologia
18.
Carcinogenesis ; 32(10): 1484-92, 2011 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-21828060

RESUMO

Retroelements constitute a large part of the human genome. These sequences are mostly silenced in normal cells, but genome-wide DNA hypomethylation in cancers might lead to their re-expression. Whether this re-expression really occurs in human cancers is largely unkown. We therefore investigated expression and DNA methylation of several classes of retroelements in human prostate cancer tissues and cell lines by quantitative reverse transcription-polymerase chain reaction and pyrosequencing, respectively. The most striking finding was strong and generalized increased expression of the HERV-K_22q11.23 provirus in cancers, including de novo expression of a spliced accessory Np9 transcript in some tumors. In parallel, DNA methylation in the long terminal repeat (LTR) decreased. Conversely, HERVK17 expression was significantly diminished in cancer tissues, but this decrease was unrelated to LTR methylation. Expression of both proviruses was restricted to androgen-responsive prostate cancer cell lines and LTRs sequences containing steroid hormone-responsive elements bound the androgen receptor and conferred androgen responsiveness to reporter constructs. Expression of LINE-1 5'-untranslated region (UTR) and 3'-UTR sequences in prostate cancers rather decreased, despite significant hypomethylation of the internal LINE-1 promoter. Increased expression of the young AluYa5 and AluYb8 families was restricted to individual tumors. Our findings demonstrate a surprising specificity of changes in expression and DNA methylation of retroelements in prostate cancer. In particular, LINE-1 hypomethylation does not lead to generalized overexpression, but specific human endogenous retrovirus-K proviruses display conspicuous changes in their expression hinting at significant functions during prostate carcinogenesis.


Assuntos
Metilação de DNA , Retrovirus Endógenos/genética , Elementos Nucleotídeos Longos e Dispersos/genética , Neoplasias da Próstata/genética , Provírus/genética , Retroelementos/fisiologia , Imunoprecipitação da Cromatina , DNA de Neoplasias/genética , Humanos , Luciferases/metabolismo , Masculino , Reação em Cadeia da Polimerase , Regiões Promotoras Genéticas , Neoplasias da Próstata/metabolismo , Neoplasias da Próstata/patologia , Transcrição Gênica , Células Tumorais Cultivadas
19.
Genetika ; 47(2): 149-58, 2011 Feb.
Artigo em Russo | MEDLINE | ID: mdl-21516786

RESUMO

The rDNA locus of insects and other arthropods contains non-LTR retrotransposons (retroposons) that are specifically inserted into 28S rRNA genes. The most frequent retroposons are R1 and R2, but the mechanism of insertion and the functions of these mobile elements have not been studied in detail. A clone containing a full-length R1 retroposon copy was islated from the cosmid library of Blattella germanica genes and sequenced. The amino acid sequences encoded by ORF1 of the R1 retroposon were subjected to bioinformatic analysis. It was found that ORF1 of this mobile element encodes a protein (ORF1p) belonging to the superfamily of zinc finger (CCHC) retroviral nucleocapsid proteins and contains two conserved RRM domains (RNA-recognizing motifs) identified on the basis of analysis of the secondary structure of this protein. The discovery of RRM domains in ORF1p of R1 retroposons can contribute to the understanding of the mechanisms of their retrotransposition. We revealed a coiled-coil motif in the N-terminal region of R1 ORF1p, which is similar to the coiled-coil domain involved in homo- or heteromultimerization of proteins and in protein-protein interactions. The domain organization of homologous Gag-like proteins of retroposons in some insects and fungi was found to be similar to the structure established by us for R1 ORF1p of B. germanica.


Assuntos
Baratas/química , Proteínas de Insetos/química , Fases de Leitura Aberta/fisiologia , Retroelementos/fisiologia , Motivos de Aminoácidos , Animais , Baratas/genética , Proteínas Fúngicas/química , Proteínas Fúngicas/genética , Fungos/química , Fungos/genética , Proteínas de Insetos/genética , Homologia Estrutural de Proteína , Dedos de Zinco
20.
J Cell Biol ; 192(5): 781-95, 2011 Mar 07.
Artigo em Inglês | MEDLINE | ID: mdl-21383078

RESUMO

The Tudor domain-containing proteins (TDRDs) are an evolutionarily conserved family of proteins involved in germ cell development. We show here that in mice, TDRD5 is a novel component of the intermitochondrial cements (IMCs) and the chromatoid bodies (CBs), which are cytoplasmic ribonucleoprotein granules involved in RNA processing for spermatogenesis. Tdrd5-deficient males are sterile because of spermiogenic arrest at the round spermatid stage, with occasional failure in meiotic prophase. Without TDRD5, IMCs and CBs are disorganized, with mislocalization of their key components, including TDRD1/6/7/9 and MIWI/MILI/MIWI2. In addition, Tdrd5-deficient germ cells fail to repress LINE-1 retrotransposons with DNA-demethylated promoters. Cyclic adenosine monophosphate response element modulator (CREM) and TRF2, key transcription factors for spermiogenesis, are expressed in Tdrd5-deficient round spermatids, but their targets, including Prm1/Prm2/Tnp1, are severely down-regulated, which indicates the importance of IMC/CB-mediated regulation for postmeiotic gene expression. Strikingly, Tdrd5-deficient round spermatids injected into oocytes contribute to fertile offspring, demonstrating that acquisition of a functional haploid genome may be uncoupled from TDRD5 function.


Assuntos
Proteínas/fisiologia , Retroelementos/fisiologia , Espermatogênese/genética , Animais , Linhagem da Célula , Proteínas Cromossômicas não Histona/metabolismo , Modulador de Elemento de Resposta do AMP Cíclico/metabolismo , Metilação de DNA , Feminino , Regulação da Expressão Gênica , Elementos Nucleotídeos Longos e Dispersos/fisiologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Regiões Promotoras Genéticas , Protaminas/metabolismo , Proteínas/análise , Proteínas/genética , Espermátides/metabolismo , Espermatogênese/fisiologia , Proteína 2 de Ligação a Repetições Teloméricas/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA