Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 19 de 19
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
BMC Vet Res ; 20(1): 187, 2024 May 10.
Artículo en Inglés | MEDLINE | ID: mdl-38730463

RESUMEN

BACKGROUND: Porcine epidemic diarrhea virus (PEDV), a type of coronavirus, is one of the main pathogens that can infect pigs of all ages. It causes diarrhea and acute death of newborn piglets, resulting in massive economic losses to the worldwide swine industry. While vaccination remains the primary approach in combating PEDV, it often fails to address all the challenges posed by the infection, particularly in light of the emergence of evolving mutant strains. Therefore, there is a critical need to identify potent antiviral drugs that can effectively safeguard pigs against PEDV infection. RESULTS: In this study, the antiviral efficacy of SP2509, a specific antagonist of Lysine-specific demethylase 1(LSD1), was evaluated in vitro. The RT-qPCR, Western blot, TCID50, and IFA showed that at a concentration of 1µmol/L, SP2509 significantly inhibited PEDV infection. Additionally, viral life cycle assays showed that SP2509 operates by impeding PEDV internalization and replication rather than attachment and release. Regarding mechanism, in Huh-7 cells, knockdowns LSD1 can suppress PEDV replication. This indicated that the inhibition effect of SP2509 on PEDV largely depends on the activity of its target protein, LSD1. CONCLUSION: Our results in vitro show that SP2509 can inhibit PEDV infection during the internalization and replication stage and revealed a role of LSD1 as a restriction factor for PEDV. These imply that LSD1 might be a target for interfering with the viral infection, and SP2509 could be developed as an effective anti-PEDV agent.


Asunto(s)
Antivirales , Histona Demetilasas , Virus de la Diarrea Epidémica Porcina , Replicación Viral , Virus de la Diarrea Epidémica Porcina/efectos de los fármacos , Animales , Antivirales/farmacología , Replicación Viral/efectos de los fármacos , Histona Demetilasas/antagonistas & inhibidores , Porcinos , Chlorocebus aethiops , Enfermedades de los Porcinos/virología , Enfermedades de los Porcinos/tratamiento farmacológico , Infecciones por Coronavirus/veterinaria , Infecciones por Coronavirus/tratamiento farmacológico , Infecciones por Coronavirus/virología , Células Vero
2.
Elife ; 122024 Feb 19.
Artículo en Inglés | MEDLINE | ID: mdl-38372724

RESUMEN

Epigenetic regulators present novel opportunities for both ischemic stroke research and therapeutic interventions. While previous work has implicated that they may provide neuroprotection by potentially influencing coordinated sets of genes and pathways, most of them remain largely uncharacterized in ischemic conditions. In this study, we used the oxygen-glucose deprivation (OGD) model in the immortalized mouse hippocampal neuronal cell line HT-22 and carried out an RNAi screen on epigenetic regulators. PRMT5 was identified as a novel negative regulator of neuronal cell survival after OGD, which presented a phenotype of translocation from the cytosol to the nucleus upon oxygen and energy depletion both in vitro and in vivo. PRMT5 bound to the chromatin and a large number of promoter regions to repress downstream gene expression. Silencing Prmt5 significantly dampened the OGD-induced changes for a large-scale of genes, and gene ontology analysis showed that PRMT5-target genes were highly enriched for Hedgehog signaling. Encouraged by the above observation, mice were treated with middle cerebral artery occlusion with the PRMT5 inhibitor EPZ015666 and found that PRMT5 inhibition sustains protection against neuronal death in vivo. Together, these findings revealed a novel epigenetic mechanism of PRMT5 in cerebral ischemia and uncovered a potential target for neuroprotection.


Asunto(s)
Isquemia Encefálica , Proteínas Hedgehog , Proteína-Arginina N-Metiltransferasas , Animales , Ratones , Isquemia Encefálica/genética , Glucosa , Neuroprotección/genética , Oxígeno , Fenotipo , Proteína-Arginina N-Metiltransferasas/genética
3.
World J Microbiol Biotechnol ; 40(2): 78, 2024 Jan 23.
Artículo en Inglés | MEDLINE | ID: mdl-38253730

RESUMEN

Efforts to curtail the escalating health threat posed by methicillin-resistant Staphylococcus aureus (MRSA), a formidable superbug, necessitate the development of innovative treatment strategies. Leveraging potential compounds from natural sources in tandem with antibiotics has emerged as a promising approach against MRSA. These strategies should enhance the antibiotic efficacy, reduce dosage and toxicity, and bypass MRSA resistance. In this study, we used a checkerboard assay to illustrate the significant synergistic anti-MRSA effect of shikimic acid (SA), a naturally occurring compound, and ceftiofur (CF). Time-kill curves further revealed that a combination of 1/4 of the minimum inhibitory concentration (MIC) of SA and 1/8 MIC of the sodium CF eradicated MRSA within 2 h, with no noticeable toxicity observed with these concentrations. In vivo experiments confirmed that this combination therapy demonstrated robust antimicrobial activity against MRSA-induced bacteremia in mice, significantly reducing bacterial loads in the kidneys, liver, and spleen, attenuating inflammatory cell infiltration, and alleviating pathological damage. This study not only offers a compelling strategy, capitalizing on the synergistic potential of SA and CF, to rapidly address antibiotic resistance but also contributes significantly to the refinement of antimicrobial therapeutic strategies.


Asunto(s)
Staphylococcus aureus Resistente a Meticilina , Animales , Ratones , Ácido Shikímico/farmacología , Cefalosporinas/farmacología , Antibacterianos/farmacología
4.
Ecotoxicol Environ Saf ; 269: 115747, 2024 Jan 01.
Artículo en Inglés | MEDLINE | ID: mdl-38070415

RESUMEN

Microplastics (MPs) are global environmental pollutants with potential toxicity concerns, and their effects on the reproductive system have attracted increasing attention. This study investigated the interaction between MPs and mammalian biomolecules, focusing on the relationship between the testosterone adsorption behavior of MPs and male reproductive health. The adsorption capacity of different types of MPs for testosterone was evaluated in vitro experiments. Polyamide (PA)-MPs exhibited stronger adsorption, while polymethyl methacrylate (PMMA)-MPs displayed the weakest adsorption. Sorption equilibrium between PA-MPs and testosterone was achieved within 6 h, fitting the Pseudo-2nd-order model and Langmuir isotherm. The effects of MPs on male reproduction in mice was determined in vivo experiments. Male mice were treated with 0.1 and 0.5 mg/d PA-MPs/PMMA-MPs by gavage once per day for 28 days. The results showed that only 0.5 mg/d PA-MP exposure induced decreased serum testosterone levels, increased testicular testosterone levels compared to the control, and more severe damage to seminiferous tubule structure, sperm motility and sperm morphology compared to the PMMA-MPs group. Meanwhile, PA-MPs could reduce intracellular nuclear translocation of androgen receptor (AR) mediated by testosterone, while PMMA-MPs had no impact. The study revealed that PA-MP adsorption reduced testosterone bioavailability and caused sperm quality to decline, offering new insights into the combined toxicity mechanism of MPs in male mammals.


Asunto(s)
Microplásticos , Contaminantes Químicos del Agua , Masculino , Animales , Ratones , Microplásticos/toxicidad , Plásticos/toxicidad , Plásticos/química , Nylons , Testosterona , Adsorción , Disponibilidad Biológica , Polimetil Metacrilato , Salud Reproductiva , Semen/química , Motilidad Espermática , Contaminantes Químicos del Agua/análisis , Mamíferos
5.
Antibiotics (Basel) ; 12(7)2023 Jul 12.
Artículo en Inglés | MEDLINE | ID: mdl-37508274

RESUMEN

Non-typhoidal Salmonella (NTS) is a foodborne pathogen and a prevalent causative agent for disease outbreaks globally. The Salmonella enterica serovar 4,[5],12:i:- (S.4,[5],12:i:-) belongs to the monophasic variant of Salmonella typhimurium, which is of current global concern. In this study, the epidemiology and genomic characterization of S. 4,[5],12:i:- isolates from 17 livestock farms in Hunan Province between 2019 and 2020, as well as their susceptibility to 14 antimicrobial agents, were profiled. Twelve Salmonella serotypes were identified using the White-Kauffmann-Le Minor scheme, and whole-genome sequencing analyses were conducted based on these isolates. Overall, 107 Salmonella strains were isolated, of which 73% (78/107) were multidrug resistant. Resistance to tetracycline (85.05%) was found to be the most prevalent, followed by the oqxAB and aac(6')-Ib-cr genes. S. typhimurium (monophasic) 4,[5],12:i:- was the most common serotype, followed by S. typhimurium and S. derby. Most antimicrobial-resistant strains were isolated from pigs, indicating that they could be important reservoirs of resistant non-typhoidal Salmonella strains. The presence of similar genetic environments in S. 4,[5],12:i:- indicates both vertical and horizontal transmission of resistance plasmids, which may promote the spread of drug resistance genes. Appropriate measures should be taken to curb the prevalence of S. 4,[5],12:i:-.

6.
Stem Cell Rev Rep ; 19(5): 1540-1553, 2023 07.
Artículo en Inglés | MEDLINE | ID: mdl-36971904

RESUMEN

BACKGROUND: RAP1 interacting factor 1 (Rif1) is highly expressed in mice embryos and mouse embryonic stem cells (mESCs). It plays critical roles in telomere length homeostasis, DNA damage, DNA replication timing and ERV silencing. However, whether Rif1 regulates early differentiation of mESC is still unclear. METHODS: In this study, we generated a Rif1 conditional knockout mouse embryonic stem (ES) cell line based on Cre-loxP system. Western blot, flow cytometry, quantitative real-time polymerase chain reaction (qRT-PCR), RNA high-throughput sequencing (RNA-Seq), chromatin immunoprecipitation followed high-throughput sequencing (ChIP-Seq), chromatin immunoprecipitation quantitative PCR (ChIP-qPCR), immunofluorescence, and immunoprecipitation were employed for phenotype and molecular mechanism assessment. RESULTS: Rif1 plays important roles in self-renewal and pluripotency of mESCs and loss of Rif1 promotes mESC differentiation toward the mesendodermal germ layers. We further show that Rif1 interacts with histone H3K27 methyltransferase EZH2, a subunit of PRC2, and regulates the expression of developmental genes by directly binding to their promoters. Rif1 deficiency reduces the occupancy of EZH2 and H3K27me3 on mesendodermal gene promoters and activates ERK1/2 activities. CONCLUSION: Rif1 is a key factor in regulating the pluripotency, self-renewal, and lineage specification of mESCs. Our research provides new insights into the key roles of Rif1 in connecting epigenetic regulations and signaling pathways for cell fate determination and lineage specification of mESCs.


Asunto(s)
Fibrinógeno , Células Madre Embrionarias de Ratones , Animales , Ratones , Fibrinógeno/metabolismo , Diferenciación Celular/genética , Línea Celular , Estratos Germinativos/metabolismo , Proteínas de Unión a Telómeros/genética , Proteínas de Unión a Telómeros/metabolismo
7.
iScience ; 25(10): 105049, 2022 Oct 21.
Artículo en Inglés | MEDLINE | ID: mdl-36124234

RESUMEN

Lysine-specific demethylase 1 (LSD1) is well-known for its role in decommissioning enhancers during mouse embryonic stem cell (ESC) differentiation. Its role in gene promoters remains poorly understood despite its widespread presence at these sites. Here, we report that LSD1 promotes RNA polymerase II (RNAPII) pausing, a rate-limiting step in transcription regulation, in ESCs. We found the knockdown of LSD1 preferentially affects genes with higher RNAPII pausing. Next, we demonstrate that the co-localization sites of LSD1 and MYC, a factor known to regulate pause-release, are enriched for other RNAPII pausing factors. We show that LSD1 and MYC directly interact and MYC recruitment to genes co-regulated with LSD1 is dependent on LSD1 but not vice versa. The co-regulated gene set is significantly enriched for housekeeping processes and depleted of transcription factors compared to those bound by LSD1 alone. Collectively, our integrative analysis reveals a pleiotropic role of LSD1 in promoting RNAPII pausing.

8.
Cell Regen ; 11(1): 25, 2022 Aug 02.
Artículo en Inglés | MEDLINE | ID: mdl-35915272

RESUMEN

Mouse embryonic stem cells (mESCs) cycle in and out of a transient 2-cell (2C)-like totipotent state, driven by a complex genetic circuit involves both the coding and repetitive sections of the genome. While a vast array of regulators, including the multi-functional protein Rif1, has been reported to influence the switch of fate potential, how they act in concert to achieve this cellular plasticity remains elusive. Here, by modularizing the known totipotency regulatory factors, we identify an unprecedented functional connection between Rif1 and the non-canonical polycomb repressive complex PRC1.6. Downregulation of the expression of either Rif1 or PRC1.6 subunits imposes similar impacts on the transcriptome of mESCs. The LacO-LacI induced ectopic colocalization assay detects a specific interaction between Rif1 and Pcgf6, bolstering the intactness of the PRC1.6 complex. Chromatin immunoprecipitation followed by sequencing (ChIP-seq) analysis further reveals that Rif1 is required for the accurate targeting of Pcgf6 to a group of genomic loci encompassing many genes involved in the regulation of the 2C-like state. Depletion of Rif1 or Pcgf6 not only activates 2C genes such as Zscan4 and Zfp352, but also derepresses a group of the endogenous retroviral element MERVL, a key marker for totipotency. Collectively, our findings discover that Rif1 can serve as a novel auxiliary component in the PRC1.6 complex to restrain the genetic circuit underlying totipotent fate potential, shedding new mechanistic insights into its function in regulating the cellular plasticity of embryonic stem cells.

9.
Nat Commun ; 13(1): 3501, 2022 06 17.
Artículo en Inglés | MEDLINE | ID: mdl-35715442

RESUMEN

Transposable elements (TEs) through evolutionary exaptation have become an integral part of the human genome, offering ample regulatory sequences and shaping chromatin 3D architecture. While the functional impacts of TE-derived sequences on early embryogenesis have been recognized, their roles in malignancy are only starting to emerge. Here we show that many TEs, especially the pluripotency-related human endogenous retrovirus H (HERVH), are abnormally activated in colorectal cancer (CRC) samples. Transcriptional upregulation of HERVH is associated with mutations of several tumor suppressors, particularly ARID1A. Knockout of ARID1A in CRC cells leads to increased transcription at several HERVH loci, which involves compensatory contribution by ARID1B. Suppression of HERVH in CRC cells and patient-derived organoids impairs tumor growth. Mechanistically, HERVH transcripts colocalize with nuclear BRD4 foci, modulating their dynamics and co-regulating many target genes. Altogether, we uncover a critical role for ARID1A in restraining HERVH, whose abnormal activation can promote tumorigenesis by stimulating BRD4-dependent transcription.


Asunto(s)
Retrovirus Endógenos , Factores de Transcripción , Proteínas de Ciclo Celular/genética , Cromatina/genética , Elementos Transponibles de ADN , Proteínas de Unión al ADN/genética , Retrovirus Endógenos/genética , Retrovirus Endógenos/metabolismo , Humanos , Proteínas Nucleares/genética , Proteínas Nucleares/metabolismo , Factores de Transcripción/genética
10.
Cell Discov ; 6: 58, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32884836

RESUMEN

Previous studies have implicated an essential role for UHRF1-mediated histone H3 ubiquitination in recruiting DNMT1 to replication sites for DNA maintenance methylation during S phase of the cell cycle. However, the regulatory mechanism on UHRF1-mediated histone ubiquitination is not clear. Here we present evidence that UHRF1 and USP7 oppositely control ubiquitination of histones H3 and H2B in S phase of the cell cycle and that DNMT1 binds both ubiquitinated H3 and H2B. USP7 knockout markedly increased the levels of ubiquitinated H3 and H2B in S phase, the association of DNMT1 with replication sites and importantly, led to a progressive increase of global DNA methylation shown with increased cell passages. Using DNMT3A/DNMT3B/USP7 triple knockout cells and various DNA methylation analyses, we demonstrated that USP7 knockout led to an overall elevation of DNA methylation levels. Mechanistic study demonstrated that USP7 suppresses DNMT1 recruitment and DNA methylation through its deubiquitinase activity and the interaction with DNMT1. Altogether our study provides evidence that USP7 is a negative regulator of global DNA methylation and that USP7 protects the genome from excessive DNA methylation by attenuating histone ubiquitination-dependent DNMT1 recruitment.

11.
Cell Rep ; 31(12): 107794, 2020 06 23.
Artículo en Inglés | MEDLINE | ID: mdl-32579921

RESUMEN

As a core component of the mitotic checkpoint complex, BubR1 has a modular organization of molecular functions, with KEN box and other motifs at the N terminus inhibiting the anaphase-promoting complex/cyclosome, and a kinase domain at the C terminus, whose function remains unsettled, especially at organismal levels. We generate knock-in BubR1 mutations in the Drosophila genome to separately disrupt the KEN box and the kinase domain. All of the mutants are homozygously viable and fertile and show no defects in mitotic progression. The mutants without kinase activity have an increased lifespan and phenotypic changes associated with attenuated insulin signaling, including reduced InR on the cell membrane, weakened PI3K and AKT activity, and elevated expression of dFoxO targets. The BubR1 kinase-dead mutants have a reduced cap cell number in female germaria, which can be rescued by expressing a constitutively active InR. We conclude that one major physiological role of BubR1 kinase in Drosophila is to modulate insulin signaling.


Asunto(s)
Proteínas de Ciclo Celular/metabolismo , Proteínas de Drosophila/metabolismo , Drosophila melanogaster/citología , Drosophila melanogaster/enzimología , Insulina/metabolismo , Transducción de Señal , Células Madre/citología , Células Madre/enzimología , Animales , Proteínas de Ciclo Celular/genética , Proteínas de Drosophila/genética , Drosophila melanogaster/embriología , Embrión no Mamífero/metabolismo , Femenino , Sitios Genéticos , Homeostasis , Mitosis/genética , Mutación Puntual/genética , Somatomedinas/metabolismo
12.
Nucleic Acids Res ; 45(22): 12723-12738, 2017 Dec 15.
Artículo en Inglés | MEDLINE | ID: mdl-29040764

RESUMEN

Transposable elements, including endogenous retroviruses (ERVs), constitute a large fraction of the mammalian genome. They are transcriptionally silenced during early development to protect genome integrity and aberrant transcription. However, the mechanisms that control their repression are not fully understood. To systematically study ERV repression, we carried out an RNAi screen in mouse embryonic stem cells (ESCs) and identified a list of novel regulators. Among them, Rif1 displays the strongest effect. Rif1 depletion by RNAi or gene deletion led to increased transcription and increased chromatin accessibility at ERV regions and their neighboring genes. This transcriptional de-repression becomes more severe when DNA methylation is lost. On the mechanistic level, Rif1 directly occupies ERVs and is required for repressive histone mark H3K9me3 and H3K27me3 assembly and DNA methylation. It interacts with histone methyltransferases and facilitates their recruitment to ERV regions. Importantly, Rif1 represses ERVs in human ESCs as well, and the evolutionally-conserved HEAT-like domain is essential for its function. Finally, Rif1 acts as a barrier during somatic cell reprogramming, and its depletion significantly enhances reprogramming efficiency. Together, our study uncovered many previously uncharacterized repressors of ERVs, and defined an essential role of Rif1 in the epigenetic defense against ERV activation.


Asunto(s)
Cromatina/genética , Retrovirus Endógenos/genética , Proteínas de Unión a Telómeros/genética , Activación Viral , Animales , Línea Celular , Células Cultivadas , Cromatina/metabolismo , Metilación de ADN , Células Madre Embrionarias/metabolismo , Retrovirus Endógenos/fisiología , Células HEK293 , Histona Metiltransferasas , N-Metiltransferasa de Histona-Lisina/metabolismo , Histonas/metabolismo , Humanos , Metilación , Ratones , Interferencia de ARN , Proteínas de Unión a Telómeros/metabolismo
13.
Cell Discov ; 2: 16007, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-27462454

RESUMEN

Global DNA hypomethylation is a most common epigenetic alteration in cancer, but the mechanism remains elusive. Previous studies demonstrate that UHRF1 but not UHRF2 is required for mediating DNA maintenance methylation by DNMT1. Here we report unexpectedly a conserved function for UHRF1 and UHRF2: inhibiting de novo DNA methylation by functioning as E3 ligases promoting DNMT3A degradation. UHRF1/2 are frequently overexpressed in cancers and we present evidence that UHRF1/2 overexpression downregulates DNMT3A proteins and consequently leads to DNA hypomethylation. Abrogating this negative regulation on DNMT3A or overexpression of DNMT3A leads to increased DNA methylation and impaired tumor growth. We propose a working model that UHRF1/2 safeguards the fidelity of DNA methylation and suggests that UHRF1/2 overexpression is likely a causal factor for widespread DNA hypomethylation in cancer via suppressing DNMT3A.

14.
Genes Dev ; 30(12): 1440-53, 2016 06 15.
Artículo en Inglés | MEDLINE | ID: mdl-27340176

RESUMEN

Superenhancers (SEs) are large genomic regions with a high density of enhancer marks. In cancer, SEs are found near oncogenes and dictate cancer gene expression. However, how oncogenic SEs are regulated remains poorly understood. Here, we show that INO80, a chromatin remodeling complex, is required for SE-mediated oncogenic transcription and tumor growth in melanoma. The expression of Ino80, the SWI/SNF ATPase, is elevated in melanoma cells and patient melanomas compared with normal melanocytes and benign nevi. Furthermore, Ino80 silencing selectively inhibits melanoma cell proliferation, anchorage-independent growth, tumorigenesis, and tumor maintenance in mouse xenografts. Mechanistically, Ino80 occupies >90% of SEs, and its occupancy is dependent on transcription factors such as MITF and Sox9. Ino80 binding reduces nucleosome occupancy and facilitates Mediator recruitment, thus promoting oncogenic transcription. Consistently, genes co-occupied by Ino80 and Med1 are selectively expressed in melanomas compared with melanocytes. Together, our results reveal an essential role of INO80-dependent chromatin remodeling in SE function and suggest a novel strategy for disrupting SEs in cancer treatment.


Asunto(s)
Carcinogénesis/genética , Proteínas de Ciclo Celular/metabolismo , Elementos de Facilitación Genéticos/fisiología , Regulación Neoplásica de la Expresión Génica/genética , Melanoma/genética , Melanoma/fisiopatología , Proteínas Nucleares/metabolismo , Animales , Proteínas de Ciclo Celular/genética , Proliferación Celular/genética , Ensamble y Desensamble de Cromatina/genética , Silenciador del Gen , Xenoinjertos , Humanos , Subunidad 1 del Complejo Mediador/genética , Melanocitos/metabolismo , Melanoma/enzimología , Ratones , Proteínas Nucleares/genética , Unión Proteica , Factores de Transcripción/metabolismo
15.
J Biol Chem ; 290(22): 14181-91, 2015 May 29.
Artículo en Inglés | MEDLINE | ID: mdl-25900245

RESUMEN

The underlying mechanism for the establishment and maintenance of differential DNA methylation in imprinted genes is largely unknown. Previous studies using Dnmt1 knock-out embryonic stem (ES) cells demonstrated that, although re-expression of DNMT1 restored DNA methylation in the non-imprinted regions, the methylation patterns of imprinted genes could be restored only through germ line passage. Knock-out of Uhrf1, an accessory factor essential for DNMT1-mediated DNA methylation, in mouse ES cells also led to impaired global DNA methylation and loss of genomic imprinting. Here, we demonstrate that, although re-expression of UHRF1 in Uhrf1(-/-) ES cells restored DNA methylation for the bulk genome but not for most of the imprinted genes, it did rescue DNA methylation for the imprinted H19, Nnat, and Dlk1 genes. Analysis of histone modifications at the differential methylated regions of the imprinted genes by ChIP assays revealed that for the imprinted genes whose DNA methylation could be restored upon re-expression of UHRF1, the active histone markers (especially H3K4me3) were maintained at considerably low levels, and low levels were maintained even in Uhrf1(-/-) ES cells. In contrast, for the imprinted genes whose DNA methylation could not be restored upon UHRF1 re-expression, the active histone markers (especially H3K4me3) were relatively high and became even higher in Uhrf1(-/-) ES cells. Our study thus supports a role for histone modifications in determining the establishment of imprinting-related DNA methylation and demonstrates that mouse ES cells can be a valuable model for mechanistic study of the establishment and maintenance of differential DNA methylation in imprinted genes.


Asunto(s)
Metilación de ADN , Impresión Genómica , Proteínas Nucleares/genética , Alelos , Animales , Proteínas Potenciadoras de Unión a CCAAT , Proteínas de Unión al Calcio , Inmunoprecipitación de Cromatina , Cromatografía Líquida de Alta Presión , Islas de CpG , Células Madre Embrionarias/citología , Histonas/metabolismo , Péptidos y Proteínas de Señalización Intercelular/metabolismo , Proteínas de la Membrana/metabolismo , Ratones , Ratones Transgénicos , Proteínas del Tejido Nervioso/metabolismo , Proteínas Nucleares/metabolismo , Análisis de Secuencia por Matrices de Oligonucleótidos , ARN Largo no Codificante/metabolismo , Sulfitos/química , Factores de Transcripción/genética , Factores de Transcripción/metabolismo , Ubiquitina-Proteína Ligasas
16.
J Biol Chem ; 289(9): 5986-96, 2014 Feb 28.
Artículo en Inglés | MEDLINE | ID: mdl-24394411

RESUMEN

The ten-eleven translocation (TET) family of dioxygenases (TET1/2/3) converts 5-methylcytosine to 5-hydroxymethylcytosine and provides a vital mechanism for DNA demethylation. However, how TET proteins are regulated is largely unknown. Here we report that the O-linked ß-GlcNAc (O-GlcNAc) transferase (OGT) is not only a major TET3-interacting protein but also regulates TET3 subcellular localization and enzymatic activity. OGT catalyzes the O-GlcNAcylation of TET3, promotes TET3 nuclear export, and, consequently, inhibits the formation of 5-hydroxymethylcytosine catalyzed by TET3. Although TET1 and TET2 also interact with and can be O-GlcNAcylated by OGT, neither their subcellular localization nor their enzymatic activity are affected by OGT. Furthermore, we show that the nuclear localization and O-GlcNAcylation of TET3 are regulated by glucose metabolism. Our study reveals the differential regulation of TET family proteins by OGT and a novel link between glucose metabolism and DNA epigenetic modification.


Asunto(s)
Núcleo Celular/enzimología , Metilación de ADN/fisiología , Dioxigenasas/metabolismo , N-Acetilglucosaminiltransferasas/metabolismo , 5-Metilcitosina/análogos & derivados , Transporte Activo de Núcleo Celular/fisiología , Núcleo Celular/genética , Citosina/análogos & derivados , Citosina/metabolismo , Dioxigenasas/genética , Glucosa/genética , Glucosa/metabolismo , Células HeLa , Humanos , N-Acetilglucosaminiltransferasas/genética
17.
Cell Rep ; 4(4): 817-29, 2013 Aug 29.
Artículo en Inglés | MEDLINE | ID: mdl-23972994

RESUMEN

Transcription has been linked to DNA damage. How the most highly transcribed mammalian ribosomal (rDNA) genes maintain genome integrity in the absence of transcription-coupled DNA damage repair is poorly understood. Here, we report that ABH2/ALKBH2, a DNA alkylation repair enzyme, is highly enriched in the nucleolus. ABH2 interacts with DNA repair proteins Ku70 and Ku80 as well as nucleolar proteins nucleolin, nucleophosmin 1, and upstream binding factor (UBF). ABH2 associates with and promotes rDNA transcription through its DNA repair activity. ABH2 knockdown impairs rDNA transcription and leads to increased single-stranded and double-stranded DNA breaks that are more pronounced in the rDNA genes, whereas ABH2 overexpression protects cells from methyl-methanesulfonate-induced DNA damage and inhibition of rDNA transcription. In response to massive alkylation damage, ABH2 rapidly redistributes from the nucleolus to nucleoplasm. Our study thus reveals a critical role of ABH2 in maintaining rDNA gene integrity and transcription and provides insight into the ABH2 DNA repair function.


Asunto(s)
Enzimas Reparadoras del ADN/metabolismo , Dioxigenasas/metabolismo , ARN Ribosómico/genética , Reparación del ADN por Recombinación , Transcripción Genética , Transporte Activo de Núcleo Celular , Dioxigenasa Dependiente de Alfa-Cetoglutarato, Homólogo 2 de AlkB , Alquilación , Antígenos Nucleares/metabolismo , Nucléolo Celular/enzimología , Nucléolo Celular/metabolismo , Roturas del ADN de Doble Cadena , Enzimas Reparadoras del ADN/genética , Proteínas de Unión al ADN/metabolismo , Dioxigenasas/genética , Células HEK293 , Células HeLa , Humanos , Autoantígeno Ku , Proteínas Nucleares/metabolismo , Nucleofosmina , Fosfoproteínas/metabolismo , Proteínas del Complejo de Iniciación de Transcripción Pol1/metabolismo , Unión Proteica , ARN Ribosómico/metabolismo , Proteínas de Unión al ARN/metabolismo , Nucleolina
18.
Nat Commun ; 4: 1563, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23463006

RESUMEN

Epigenetic inheritance of DNA methylation in mammals requires a multifunctional protein UHRF1, which is believed to recruit DNMT1 to DNA replication forks through a unique hemi-methylated CpG-binding activity. Here we demonstrate that the UHRF1 mutants deficient in binding either hemi-methylated CpG or H3K9me2/3, but not both, are able to associate with pericentric heterochromatin, recruit Dnmt1 and partially rescue DNA methylation defects in mouse Uhrf1 null ES cells. Furthermore, we present evidence that the flip out of the methylated cytosine induced by UHRF1 binding is unlikely essential for subsequent DNA methylation by DNMT1. Together, our study demonstrates that UHRF1 can target DNMT1 for DNA maintenance methylation through binding either H3K9me2/3 or hemi-methylated CpG, and that the presence of both binding activities ensures high fidelity DNA maintenance methylation. In addition, our study indicates that UHRF1 mediates cross-talk between H3K9 methylation and DNA methylation at the level of DNA methylation maintenance.


Asunto(s)
ADN (Citosina-5-)-Metiltransferasas/metabolismo , Metilación de ADN , Histonas/metabolismo , Lisina/metabolismo , Proteínas Nucleares/metabolismo , 5-Metilcitosina/metabolismo , Animales , Proteínas Potenciadoras de Unión a CCAAT , Islas de CpG/genética , ADN (Citosina-5-)-Metiltransferasa 1 , Replicación del ADN , Células Madre Embrionarias/citología , Células Madre Embrionarias/metabolismo , Células HeLa , Heterocromatina/metabolismo , Humanos , Ratones , Modelos Biológicos , Proteínas Mutantes/metabolismo , Mutación/genética , Células 3T3 NIH , Proteínas Nucleares/química , Proteínas Nucleares/deficiencia , Antígeno Nuclear de Célula en Proliferación/metabolismo , Unión Proteica , Estructura Terciaria de Proteína , Transporte de Proteínas , Fracciones Subcelulares/metabolismo , Ubiquitina-Proteína Ligasas
19.
Cell Res ; 21(12): 1723-39, 2011 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-22064703

RESUMEN

Recent studies demonstrate that UHRF1 is required for DNA methylation maintenance by targeting DNMT1 to DNA replication foci, presumably through its unique hemi-methylated DNA-binding activity and interaction with DNMT1. UHRF2, another member of the UHRF family proteins, is highly similar to UHRF1 in both sequence and structure, raising questions about its role in DNA methylation. In this study, we demonstrate that, like UHRF1, UHRF2 also binds preferentially to methylated histone H3 lysine 9 (H3K9) through its conserved tudor domain and hemi-methylated DNA through the SET and Ring associated domain. Like UHRF1, UHRF2 is enriched in pericentric heterochromatin. The heterochromatin localization depends to large extent on its methylated H3K9-binding activity and to less extent on its methylated DNA-binding activity. Coimmunoprecipitation experiments demonstrate that both UHRF1 and UHRF2 interact with DNMT1, DNMT3a, DNMT3b and G9a. Despite all these conserved functions, we find that UHRF2 is not able to rescue the DNA methylation defect in Uhrf1 null mouse embryonic stem cells. This can be attributed to the inability for UHRF2 to recruit DNMT1 to replication foci during S phase of the cell cycle. Indeed, we find that while UHRF1 interacts with DNMT1 in an S phase-dependent manner in cells, UHRF2 does not. Thus, our study demonstrates that UHRF2 and UHRF1 are not functionally redundant in DNA methylation maintenance and reveals the cell-cycle-dependent interaction between UHRF1 and DNMT1 as a key regulatory mechanism targeting DNMT1 for DNA methylation.


Asunto(s)
ADN (Citosina-5-)-Metiltransferasas/metabolismo , Metilación de ADN , Proteínas Nucleares/metabolismo , Ubiquitina-Proteína Ligasas/metabolismo , Animales , Proteínas Potenciadoras de Unión a CCAAT , Células Cultivadas , ADN (Citosina-5-)-Metiltransferasa 1 , Células Madre Embrionarias/metabolismo , Heterocromatina/metabolismo , Histonas/metabolismo , Humanos , Ratones , Proteínas Nucleares/genética , Unión Proteica , Fase S
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...