Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 13 de 13
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
NAR Cancer ; 5(3): zcad050, 2023 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-37746636

RESUMEN

SET/TAF-Iß, a subunit of the inhibitor of acetyltransferases (INHAT) complex, exhibits transcriptional repression activity by inhibiting histone acetylation. We find that SET/TAF-Iß regulates mono-ubiquitination of histone H2A at lysine 119 (H2AK119ub), which is involved in polycomb-mediated transcriptional repression, in HCT116 cells. In this report, we demonstrate that SET/TAF-Iß acts as an E2 ubiquitin-conjugating enzyme for PRC1-independent H2AK119ub. Furthermore, we identify that MIB1 is the E3 ligase partner for SET/TAF-Iß using LC-MS/MS and in vitro ubiquitination assays. Transcriptome analysis reveals that SET/TAF-Iß and MIB1 regulate the expression of genes related to DNA replication and cell cycle progression in HCT116 cells, and knockdown of either protein reduces proliferation of HCT116 cells by impeding cell cycle progression. Together, our study reveals a novel PRC1-independent epigenetic regulatory mechanism for H2AK119ub by SET/TAF-Iß and MIB1 in colon cancer.

2.
Nat Cell Biol ; 25(9): 1369-1383, 2023 09.
Artículo en Inglés | MEDLINE | ID: mdl-37696949

RESUMEN

Oxidative stress contributes to tumourigenesis by altering gene expression. One accompanying modification, 8-oxoguanine (o8G) can change RNA-RNA interactions via o8G•A base pairing, but its regulatory roles remain elusive. Here, on the basis of o8G-induced guanine-to-thymine (o8G > T) variations featured in sequencing, we discovered widespread position-specific o8Gs in tumour microRNAs, preferentially oxidized towards 5' end seed regions (positions 2-8) with clustered sequence patterns and clinically associated with patients in lower-grade gliomas and liver hepatocellular carcinoma. We validated that o8G at position 4 of miR-124 (4o8G-miR-124) and 4o8G-let-7 suppress lower-grade gliomas, whereas 3o8G-miR-122 and 4o8G-let-7 promote malignancy of liver hepatocellular carcinoma by redirecting the target transcriptome to oncogenic regulatory pathways. Stepwise oxidation from tumour-promoting 3o8G-miR-122 to tumour-suppressing 2,3o8G-miR-122 occurs and its specific modulation in mouse liver effectively attenuates diethylnitrosamine-induced hepatocarcinogenesis. These findings provide resources and insights into epitranscriptional o8G regulation of microRNA functions, reprogrammed by redox changes, implicating its control for cancer treatment.


Asunto(s)
Carcinoma Hepatocelular , Glioma , Neoplasias Hepáticas , MicroARNs , Animales , Ratones , Carcinoma Hepatocelular/inducido químicamente , Carcinoma Hepatocelular/genética , MicroARNs/genética , Carcinogénesis/genética , Guanina , Oxidación-Reducción , Neoplasias Hepáticas/inducido químicamente , Neoplasias Hepáticas/genética
3.
Exp Mol Med ; 54(10): 1626-1642, 2022 10.
Artículo en Inglés | MEDLINE | ID: mdl-36266447

RESUMEN

In pathophysiology, reactive oxygen species control diverse cellular phenotypes by oxidizing biomolecules. Among these, the guanine base in nucleic acids is the most vulnerable to producing 8-oxoguanine, which can pair with adenine. Because of this feature, 8-oxoguanine in DNA (8-oxo-dG) induces a G > T (C > A) mutation in cancers, which can be deleterious and thus actively repaired by DNA repair pathways. 8-Oxoguanine in RNA (o8G) causes problems in aberrant quality and translational fidelity, thereby it is subjected to the RNA decay pathway. In addition to oxidative damage, 8-oxo-dG serves as an epigenetic modification that affects transcriptional regulatory elements and other epigenetic modifications. With the ability of o8G•A in base pairing, o8G alters structural and functional RNA-RNA interactions, enabling redirection of posttranscriptional regulation. Here, we address the production, regulation, and function of 8-oxo-dG and o8G under oxidative stress. Primarily, we focus on the epigenetic and epitranscriptional roles of 8-oxoguanine, which highlights the significance of oxidative modification in redox-mediated control of gene expression.


Asunto(s)
Reparación del ADN , Guanina , 8-Hidroxi-2'-Desoxicoguanosina , Guanina/química , Guanina/metabolismo , Estrés Oxidativo , Daño del ADN , Epigénesis Genética , ARN/genética , ARN/metabolismo
4.
Genes Genomics ; 44(11): 1353-1361, 2022 11.
Artículo en Inglés | MEDLINE | ID: mdl-35951156

RESUMEN

BACKGROUND: Ubiquitin-like with PHD and RING finger domains 1 (UHRF1) is upregulated in colon cancer cells and associated with silencing tumor suppressor genes (TSGs) to promote colon cancer cell proliferation. OBJECTIVE: To investigate epigenetic modification of UHRF1 by TIP60. Whether UHRF1 acetylation by TIP60 can induce cell proliferation in colon cancer cells. METHODS: Acetylation sites of UHRF1 by TIP60 was predicted by ASEB (Acetylation Set Enrichment Based) method and identified by immunoprecipitation assay using anti-pan-acetyl lysine antibody and in vitro acetylation assay. Based on this method, UHRF1 acetylation-deficient mimic 4KR (K644R, K646R, K648R, K650R) mutant was generated to investigate effects of UHRF1 acetylation by TIP60. shRNA system was used to generate stable knockdown cell line of UHRF1. With transient transfection of UHRF1 WT and 4KR, the effects of UHRF1 4KR mutant on Jun dimerization protein 2 (JDP2) gene expression, cell proliferation and cell cycle were investigated by RT-qPCR and FACS analysis in shUHRF1 colon cancer cell line. RESULTS: Downregulation of TIP60-mediated UHRF1 acetylation is correlated with suppressed cell cycle progression. Acetylation-deficient mimic of UHRF1 showed poor cell growth through increased expression of JDP2 gene. CONCLUSIONS: Acetylation of UHRF1 4K residues by TIP60 is important for colon cancer cell growth. Furthermore, upregulated JDP2 expression by acetylation-deficient mutant of UHRF1 might be an important epigenetic target for colon cancer cell proliferation.


Asunto(s)
Proteínas Potenciadoras de Unión a CCAAT , Neoplasias del Colon , Lisina Acetiltransferasa 5 , Ubiquitina-Proteína Ligasas , Acetilación , Proteínas Potenciadoras de Unión a CCAAT/genética , Proteínas Potenciadoras de Unión a CCAAT/metabolismo , Proliferación Celular/genética , Neoplasias del Colon/genética , Metilación de ADN , Humanos , Lisina/genética , Lisina/metabolismo , Lisina Acetiltransferasa 5/genética , Lisina Acetiltransferasa 5/metabolismo , ARN Interferente Pequeño , Ubiquitina-Proteína Ligasas/genética , Ubiquitina-Proteína Ligasas/metabolismo , Ubiquitinas/genética , Ubiquitinas/metabolismo
5.
BMB Rep ; 55(11): 541-546, 2022 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-35880433

RESUMEN

The repair of DNA double-strand breaks (DSBs) by homologous recombination (HR) is crucial for maintaining genomic integrity and is involved in numerous fundamental biological processes. Post-translational modifications by proteins play an important role in regulating DNA repair. Here, we report that the methyltransferase SET7 regulates HR-mediated DSB repair by methylating TIP60, a histone acetyltransferase and tumor suppressor involved in gene expression and protein stability. We show that SET7 targets TIP60 for methylation at K137, which facilitates DSB repair by promoting HR and determines cell viability against DNA damage. Interestingly, TIP60 demethylation is catalyzed by LSD1, which affects HR efficiency. Taken together, our findings reveal the importance of TIP60 methylation status by SET7 and LSD1 in the DSB repair pathway. [BMB Reports 2022; 55(11): 541-546].


Asunto(s)
Roturas del ADN de Doble Cadena , Histonas , Metilación , Histonas/metabolismo , Reparación del ADN , Procesamiento Proteico-Postraduccional , ADN/metabolismo , Histona Demetilasas/metabolismo
6.
Biochem Biophys Res Commun ; 561: 120-127, 2021 07 05.
Artículo en Inglés | MEDLINE | ID: mdl-34023776

RESUMEN

Epigenetic dysregulation has been strongly implicated in carcinogenesis and is one of the mechanisms that contribute to the development of lung cancer. Using genome-wide CRISPR/Cas9 library screening, we showed SET domain-containing protein 1A (SETD1A) is an essential epigenetic modifier of the proliferation of NSCLC H1299 cells. Depletion of SETD1A strikingly inhibited the proliferation of NSCLC cells. IHC staining and bioinformatics showed that SETD1A is upregulated in lung cancer. Kaplan-Meier survival analysis indicated that high expression of SETD1A is associated with poor prognosis of patients with NSCLC. We revealed that loss of SETD1A inhibits DNA replication and induces replication stress accompanied by impaired fork progression. In addition, transcription of CDC7 and TOP1, which are involved in replication origin activation and fork progression, respectively, was significantly reduced by knockdown of SETD1A. Taken together, these findings demonstrated SETD1A is a critical epigenetic modifier of NSCLC cell proliferation by promoting the transcription of a subset of DNA replication-associated genes.


Asunto(s)
Carcinoma de Pulmón de Células no Pequeñas/enzimología , Carcinoma de Pulmón de Células no Pequeñas/patología , N-Metiltransferasa de Histona-Lisina/metabolismo , Neoplasias Pulmonares/enzimología , Neoplasias Pulmonares/patología , Sistemas CRISPR-Cas , Carcinoma de Pulmón de Células no Pequeñas/genética , Carcinoma de Pulmón de Células no Pequeñas/metabolismo , Proteínas de Ciclo Celular/metabolismo , Línea Celular Tumoral , Proliferación Celular , Células Cultivadas , Biología Computacional/métodos , Replicación del ADN , Bases de Datos Genéticas , Regulación Neoplásica de la Expresión Génica , N-Metiltransferasa de Histona-Lisina/antagonistas & inhibidores , N-Metiltransferasa de Histona-Lisina/genética , Humanos , Neoplasias Pulmonares/genética , Neoplasias Pulmonares/metabolismo , Proteínas Serina-Treonina Quinasas/metabolismo , Fase S
7.
Cell Prolif ; 53(11): e12920, 2020 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-33029857

RESUMEN

OBJECTIVES: The level of histone H3 lysine 79 methylation is regulated by the cell cycle and involved in cell proliferation. KDM2B is an H3K79 demethylase. Proliferating cell nuclear antigen (PCNA) is a component of the DNA replication machinery. This study aimed at elucidating a molecular link between H3K79me recognition of PCNA and cell cycle control. MATERIALS AND METHODS: We generated KDM2B-depleted 293T cells and histone H3-K79R mutant-expressing 293T cells. Western blots were primarily utilized to examine the H3K79me level and its effect on subsequent PCNA dissociation from chromatin. We applied IP, peptide pull-down, isothermal titration calorimetry (ITC) and ChIP experiments to show the PCNA binding towards methylated H3K79 and DNA replication origins. Flow cytometry, MTT, iPOND and DNA fibre assays were used to assess the necessity of KDM2B for DNA replication and cell proliferation. RESULTS: We revealed that KDM2B-mediated H3K79 demethylation regulated cell cycle progression. We found that PCNA bound chromatin in an H3K79me-dependent manner during S phase. KDM2B was responsible for the timely dissociation of PCNA from chromatin, allowing to efficient DNA replication. Depletion of KDM2B aberrantly enriched chromatin with PCNA and caused slow dissociation of residual PCNA, leading to a negative effect on cell proliferation. CONCLUSIONS: We suggested a novel interaction between PCNA and H3K79me. Thus, our findings provide a new mechanism of KDM2B in regulation of DNA replication and cell proliferation.


Asunto(s)
Replicación del ADN , Proteínas F-Box/metabolismo , Histonas/metabolismo , Histona Demetilasas con Dominio de Jumonji/metabolismo , Antígeno Nuclear de Célula en Proliferación/metabolismo , Ciclo Celular , Proliferación Celular , Cromatina , Desmetilación , Células HEK293 , Humanos , Fase S
8.
Commun Biol ; 3(1): 462, 2020 08 21.
Artículo en Inglés | MEDLINE | ID: mdl-32826945

RESUMEN

The human myelogenous leukemic cell line, K562 undergoes erythroid differentiation by exposure to hemin. Here, we uncovered NSD2 as an innate erythroid differentiation-related factor through a genome-wide CRISPR library screen and explored the regulatory role of NSD2 during myeloid leukemia cell differentiation. We found that NSD2 stability was disrupted by poly-ubiquitination in differentiated K562 cells. Proteomic analysis revealed an interaction between NSD2 and an E3 ubiquitin ligase, BRCA1, which ubiquitylates NSD on K292. Depletion of BRCA1 stabilized NSD2 protein and suppressed K562 cell differentiation. Furthermore, BRCA1 protein level was decreased in bone marrow tumor, while NSD2 level was elevated. Surprisingly, among BRCA1 mutation(s) discovered in lymphoma patients, BRCA1 K1183R prevented its translocation into the nucleus, failed to reduce NSD2 protein levels in hemin-treated K562 cells and eventually disrupted cell differentiation. Our results indicate the regulation of NSD2 stability by BRCA1-mediated ubiquitination as a potential therapeutic target process in multiple myeloma.


Asunto(s)
Proteína BRCA1/metabolismo , N-Metiltransferasa de Histona-Lisina/metabolismo , Leucemia/metabolismo , Complejo de la Endopetidasa Proteasomal/metabolismo , Proteínas Represoras/metabolismo , Biomarcadores , Diferenciación Celular , Línea Celular Tumoral , Células Cultivadas , Epigénesis Genética , Regulación Leucémica de la Expresión Génica , Hemina/metabolismo , N-Metiltransferasa de Histona-Lisina/genética , Humanos , Células K562 , Leucemia/etiología , Leucemia/patología , Clasificación del Tumor , Unión Proteica , Proteolisis , Proteínas Represoras/genética , Ubiquitina-Proteína Ligasas/metabolismo , Ubiquitinación
9.
Cell Rep ; 32(4): 107958, 2020 07 28.
Artículo en Inglés | MEDLINE | ID: mdl-32726623

RESUMEN

UHRF1 is a key regulator in DNA methylation maintenance. It binds histone H3K9me2/3 and hemi-methylated DNA and recruits DNMT1 to DNA replication forks during S phase. However, the regulatory mechanism of hemi-methylated DNA binding activity of UHRF1 remains unknown. In this study, we reveal that acetylation of UHRF1 is regulated by PCAF and HDAC1. We show that UHRF1 acetylation at K490 attenuates its binding affinity to hemi-methylated DNA. We analyze genome-wide DNA methylation and gene-expression patterns using stable cell lines and discover that cells where the endogenous UHRF1 is replaced with an acetyl-mimetic (UHRF1 K490Q) mutant show deficiencies in inherited DNA methylation and show different gene-expression patterns in genes related to cell survival. These results reveal that precise regulation of UHRF1 acetylation is required to maintain DNA methylation during cell division and control cell survival.


Asunto(s)
Proteínas Potenciadoras de Unión a CCAAT/metabolismo , Metilación de ADN/fisiología , Ubiquitina-Proteína Ligasas/metabolismo , Acetilación , Proteínas Potenciadoras de Unión a CCAAT/fisiología , ADN/metabolismo , ADN (Citosina-5-)-Metiltransferasa 1/metabolismo , ADN (Citosina-5-)-Metiltransferasas/metabolismo , Metilación de ADN/genética , Proteínas de Unión al ADN/genética , Proteínas de Unión al ADN/metabolismo , Células HCT116 , Células HEK293 , Histona Desacetilasa 1/metabolismo , Histonas/metabolismo , Humanos , Células K562 , Proteínas Nucleares/metabolismo , Unión Proteica , Procesamiento Proteico-Postraduccional , Ubiquitina-Proteína Ligasas/fisiología , Factores de Transcripción p300-CBP/metabolismo
10.
BMB Rep ; 53(2): 112-117, 2020 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-31964471

RESUMEN

A recent study suggested that methylation of ubiquitin-like with PHD and RING finger domain 1 (UHRF1) is regulated by SET7 and lysine-specific histone demethylase 1A (LSD1) and is essential for homologous recombination (HR). The study demonstrated that SET7-mediated methylation of UHRF1 promotes polyubiquitination of proliferating cell nuclear antigen (PCNA), inducing HR. However, studies on mediators that interact with and recruit UHRF1 to damaged lesions are needed to elucidate the mechanism of UHRF1 methylationinduced HR. Here, we identified that poly [ADP-ribose] polymerase 1 (PARP1) interacts with damage-induced methylated UHRF1 specifically and mediates UHRF1 to induce HR progression. Furthermore, cooperation of UHRF1-PARP1 is essential for cell viability, suggesting the importance of the interaction of UHRF1-PARP1 for damage tolerance in response to damage. Our data revealed that PARP1 mediates the HR mechanism, which is regulated by UHRF1 methylation. The data also indicated the significant role of PARP1 as a mediator of UHRF1 methylation-correlated HR pathway. [BMB Reports 2020; 53(2): 112-117].


Asunto(s)
Proteínas Potenciadoras de Unión a CCAAT/metabolismo , Daño del ADN/genética , Recombinación Homóloga/genética , Poli(ADP-Ribosa) Polimerasa-1/metabolismo , Ubiquitina-Proteína Ligasas/metabolismo , Proteínas Potenciadoras de Unión a CCAAT/química , Proteínas Potenciadoras de Unión a CCAAT/genética , Supervivencia Celular/genética , Daño del ADN/efectos de los fármacos , Metilación de ADN/efectos de los fármacos , Puntos de Control de la Fase G2 del Ciclo Celular/efectos de los fármacos , Puntos de Control de la Fase G2 del Ciclo Celular/genética , Células HCT116 , Células HEK293 , Humanos , Peróxido de Hidrógeno/farmacología , Poli(ADP-Ribosa) Polimerasa-1/genética , Unión Proteica , Ubiquitina-Proteína Ligasas/química , Ubiquitina-Proteína Ligasas/genética
11.
Nucleic Acids Res ; 47(1): 184-196, 2019 01 10.
Artículo en Inglés | MEDLINE | ID: mdl-30357346

RESUMEN

Ubiquitin-like with PHD and RING finger domains 1 (UHRF1) is a key epigenetic regulator of DNA methylation maintenance and heterochromatin formation. The roles of UHRF1 in DNA damage repair also have been emphasized in recent years. However, the regulatory mechanism of UHRF1 remains elusive. In this study, we showed that UHRF1 is methylated by SET7 and demethylation is catalyzed by LSD1. In addition, methylation of UHRF1 is induced in response to DNA damage and its phosphorylation in S phase is a prerequisite for interaction with SET7. Furthermore, UHRF1 methylation catalyzes the conjugation of polyubiquitin chains to PCNA and promotes homologous recombination for DNA repair. SET7-mediated UHRF1 methylation is also shown to be essential for cell viability against DNA damage. Our data revealed the regulatory mechanism underlying the UHRF1 methylation status by SET7 and LSD1 in double-strand break repair pathway.


Asunto(s)
Proteínas Potenciadoras de Unión a CCAAT/genética , Roturas del ADN de Doble Cadena , Metilación de ADN/genética , Histona Demetilasas/genética , N-Metiltransferasa de Histona-Lisina/genética , Daño del ADN/genética , Reparación del ADN/genética , Heterocromatina/genética , Humanos , Fosforilación , Antígeno Nuclear de Célula en Proliferación/genética , Unión Proteica/genética , Fase S/genética , Ubiquitina-Proteína Ligasas
12.
FASEB J ; 32(10): 5737-5750, 2018 10.
Artículo en Inglés | MEDLINE | ID: mdl-29763382

RESUMEN

The methylation of histone H3 lysine 79 (H3K79) is an active chromatin marker and is prominent in actively transcribed regions of the genome; however, demethylase of H3K79 remains unknown despite intensive research. Here, we show that KDM2B, also known as FBXL10 and a member of the Jumonji C family of proteins known for its histone H3K36 demethylase activity, is a di- and trimethyl H3K79 demethylase. We demonstrate that KDM2B induces transcriptional repression of HOXA7 and MEIS1 via occupancy of promoters and demethylation of H3K79. Furthermore, genome-wide analysis suggests that H3K79 methylation levels increase when KDM2B is depleted, which indicates that KDM2B functions as an H3K79 demethylase in vivo. Finally, stable KDM2B-knockdown cell lines exhibit displacement of NAD+-dependent deacetylase sirtuin-1 (SIRT1) from chromatin, with concomitant increases in H3K79 methylation and H4K16 acetylation. Our findings identify KDM2B as an H3K79 demethylase and link its function to transcriptional repression via SIRT1-mediated chromatin silencing.-Kang, J.-Y., Kim, J.-Y., Kim, K.-B., Park, J. W., Cho, H., Hahm, J. Y., Chae, Y.-C., Kim, D., Kook, H., Rhee, S., Ha, N.-C., Seo, S.-B. KDM2B is a histone H3K79 demethylase and induces transcriptional repression via sirtuin-1-mediated chromatin silencing.


Asunto(s)
Cromatina/metabolismo , Proteínas F-Box/metabolismo , Silenciador del Gen , Proteínas de Homeodominio/biosíntesis , Histona Demetilasas con Dominio de Jumonji/metabolismo , Proteína 1 del Sitio de Integración Viral Ecotrópica Mieloide/biosíntesis , Sirtuina 1/metabolismo , Transcripción Genética , Cromatina/genética , Proteínas F-Box/genética , Proteínas de Homeodominio/genética , Humanos , Histona Demetilasas con Dominio de Jumonji/genética , Células K562 , Proteína 1 del Sitio de Integración Viral Ecotrópica Mieloide/genética , Sirtuina 1/genética
13.
Nucleic Acids Res ; 43(7): 3509-23, 2015 Apr 20.
Artículo en Inglés | MEDLINE | ID: mdl-25765655

RESUMEN

Histone H3K9 methyltransferase (HMTase) G9a-mediated transcriptional repression is a major epigenetic silencing mechanism. UHRF1 (ubiquitin-like with PHD and ring finger domains 1) binds to hemimethylated DNA and plays an essential role in the maintenance of DNA methylation. Here, we provide evidence that UHRF1 is transcriptionally downregulated by H3K9 HMTase G9a. We found that increased expression of G9a along with transcription factor YY1 specifically represses UHRF1 transcription during TPA-mediated leukemia cell differentiation. Using ChIP analysis, we found that UHRF1 was among the transcriptionally silenced genes during leukemia cell differentiation. Using a DNA methylation profiling array, we discovered that the UHRF1 promoter was hypomethylated in samples from leukemia patients, further supporting its overexpression and oncogenic activity. Finally, we showed that G9a regulates UHRF1-mediated H3K23 ubiquitination and proper DNA replication maintenance. Therefore, we propose that H3K9 HMTase G9a is a specific epigenetic regulator of UHRF1.


Asunto(s)
Proteínas Potenciadoras de Unión a CCAAT/genética , Diferenciación Celular , Metilasas de Modificación del ADN/metabolismo , Regulación de la Expresión Génica , Leucemia/patología , Transcripción Genética , Línea Celular , Inmunoprecipitación de Cromatina , Citometría de Flujo , Humanos , Leucemia/genética , Ubiquitina-Proteína Ligasas
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA