Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 36
Filtrar
1.
J Mol Med (Berl) ; 102(2): 273-284, 2024 02.
Artículo en Inglés | MEDLINE | ID: mdl-38153509

RESUMEN

EN1 encodes a homeodomain-containing transcription factor and is a determinant of bone density and fracture. Previous powerful genome-wide association studies (GWASs) have identified multiple single-nucleotide polymorphisms (SNPs) near EN1 at 2q14.2 locus for osteoporosis, but the causal SNPs and functional mechanisms underlying these associations are poorly understood. The target genes regulated by the transcription factor EN1 are also unclear. In this study, we identified rs188303909, a functional CpG-SNP, as a causal SNP for osteoporosis at 2q14.2 through the integration of functional and epigenomic analyses. Functional experiments demonstrated that unmethylated rs188303909 acted as a strong allele-specific distal enhancer to regulate EN1 expression by modifying the binding of transcription factor E2F6, but rs188303909 methylation attenuated the active effect of E2F6 on EN1 expression. Importantly, transcription factor EN1 could differentially bind osteoporosis GWAS lead SNPs rs4869739-T and rs4355801-G to upregulate CCDC170 and COLEC10 expression, thus promoting bone formation. Our study provided a mechanistic insight into expression regulation of the osteoporosis susceptibility gene EN1, which could be a potential therapeutic target for osteoporosis precision medicine. KEY MESSAGES: CpG-SNP rs188303909 is a causal SNP at the osteoporosis susceptibility locus 2q14.2. Rs188303909 distally regulates EN1 expression by modulating DNA methylation and E2F6 binding. EN1 upregulates CCDC170 and COLEC10 expression through osteoporosis GWAS lead SNPs rs4869739 and rs4355801.


Asunto(s)
Osteoporosis , Polimorfismo de Nucleótido Simple , Humanos , Estudio de Asociación del Genoma Completo , Metilación de ADN , Osteoporosis/genética , Factores de Transcripción/genética , Predisposición Genética a la Enfermedad , Colectinas/genética , Factor de Transcripción E2F6/genética , Proteínas de Homeodominio/genética
2.
Appl Biochem Biotechnol ; 195(5): 3096-3108, 2023 May.
Artículo en Inglés | MEDLINE | ID: mdl-36525235

RESUMEN

Endometrial cancer (EC) ranks fourth among the most common gynecologic malignancies. Despite advances in medical technology, the pathogenesis is still unclear. Numerous reports have identified the involvement of lncRNA in the malignant progression of endometrial cancer. The aim of the study was to investigate the expression level of lncRNA ENST00000585827 (lncRNA E27) in endometrial cancer and the molecular mechanism that regulates the development of endometrial cancer. Combined with the results of the previous study, PCR analysis confirmed that lncRNA E27 was significantly upregulated in endometrial cancer cell lines. The results of CCK-8, wound healing assay, and transwell experiments showed that lncRNA E27 could significantly inhibit cell proliferation, migration, and invasion. Flow cytometry results confirmed that lncRNA E27 could promote apoptosis. Furthermore, based on bioinformatics predictions, dual-luciferase assay and RT-qPCR analysis confirmed that miR-424, as its downstream molecule, competitively regulates the expression of E2F6/E2F7. Rescue experiments further supported that lncRNA E27 inhibited proliferation, migration, invasion, and promoted apoptosis of endometrial cancer through miR-424/E2F6/E2F7 signaling axis. Conclusively, our findings revealed the role of lncRNA E27 in regulating the miR-424/E2F6/E2F7 signaling axis during EC progression, opening up new strategies for the treatment of endometrial cancer.


Asunto(s)
Neoplasias Endometriales , MicroARNs , ARN Largo no Codificante , Humanos , Femenino , MicroARNs/genética , MicroARNs/metabolismo , ARN Largo no Codificante/genética , ARN Largo no Codificante/metabolismo , Línea Celular Tumoral , Proliferación Celular/genética , Neoplasias Endometriales/genética , Neoplasias Endometriales/metabolismo , Neoplasias Endometriales/patología , Regulación Neoplásica de la Expresión Génica , Factor de Transcripción E2F7/genética , Factor de Transcripción E2F7/metabolismo , Factor de Transcripción E2F6/genética , Factor de Transcripción E2F6/metabolismo
3.
Int J Biol Sci ; 18(10): 4071-4087, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35844791

RESUMEN

Centromere protein U (CENPU), a centromere-binding protein required for cellular mitosis, has been reported to be closely associated with carcinogenesis in multiple malignancies; however, the role of CENPU in hepatocellular carcinoma (HCC) is still unclear. Herein, we investigated its biological role and molecular mechanism in the development of HCC. High CENPU expression in HCC tissue was observed and correlated positively with a poor prognosis in HCC patients. CENPU knockdown inhibited the proliferation, metastasis, and G1/S transition of HCC cells in vivo and in vitro, while ectopic expression of CENPU exerted the opposite effects. Mechanistically, CENPU physically interacted with E2F6 and promoted its ubiquitin-mediated degradation, thus affecting the transcription level of E2F1 and further accelerating the G1/S transition to promote HCC cell proliferation. E2F1 directly binds to the CENPU promoter and increases the transcription of CENPU, thereby forming a positive regulatory loop. Collectively, our findings indicate a crucial role for CENPU in E2F1-mediated signalling for cell cycle progression and reveal a role for CENPU as a predictive biomarker and therapeutic target for HCC patients.


Asunto(s)
Carcinoma Hepatocelular , Factor de Transcripción E2F6/metabolismo , Neoplasias Hepáticas , Carcinoma Hepatocelular/metabolismo , Línea Celular Tumoral , Proliferación Celular/genética , Factor de Transcripción E2F1/genética , Factor de Transcripción E2F1/metabolismo , Factor de Transcripción E2F6/genética , Retroalimentación , Regulación Neoplásica de la Expresión Génica/genética , Humanos , Neoplasias Hepáticas/genética , Neoplasias Hepáticas/metabolismo , Metástasis de la Neoplasia , Ubiquitinación/genética
4.
Sci China Life Sci ; 65(9): 1840-1854, 2022 09.
Artículo en Inglés | MEDLINE | ID: mdl-35266112

RESUMEN

Liver cancer is highly heterogeneous, and the tumor tissue harbors a variety of cell types. Liver tumor initiating cells (TICs) well contribute to tumor heterogeneity and account for tumor initiation and metastasis, but the molecular mechanisms of liver TIC self-renewal are elusive. Here, we identified a functional read-through rt-circRNA, termed rtcisE2F, that is highly expressed in liver cancer and liver TICs. rtcisE2F plays essential roles in the self-renewal and activities of liver TICs. rtcisE2F targets E2F6 and E2F3 mRNAs, attenuates mRNA turnover, and increases E2F6/E2F3 expression. Mechanistically, rtcisE2F functions as a scaffold of N-methyladenosine (m6A) reader IGF2BP2 and E2F6/E2F3 mRNA. rtcisE2F promotes the association of E2F6/E2F3 mRNAs with IGF2BP2, and inhibits their association with another m6A reader, YTHDF2. IGF2BP2 inhibits E2F6/E2F3 mRNA decay, whereas YTHDF2 promotes E2F6/E2F3 mRNA decay. By switching m6A readers, rtcisE2F enhances E2F6/E2F3 mRNA stability. E2F6 and E2F3 are both required for liver TIC self-renewal and Wnt/ß-catenin activation, and inhibition of these pathways is a potential strategy for preventing liver tumorigenesis and metastasis. In conclusion, the rtcisE2F-IGF2BP2/YTHDF2-E2F6/E2F3-Wnt/ß-catenin axis drives liver TIC self-renewal and initiates liver tumorigenesis and metastasis, and may provide a strategy to eliminate liver TICs.


Asunto(s)
Neoplasias Hepáticas , ARN Largo no Codificante , Adenosina/análogos & derivados , Carcinogénesis/metabolismo , Línea Celular Tumoral , Factor de Transcripción E2F3 , Factor de Transcripción E2F6/genética , Factor de Transcripción E2F6/metabolismo , Regulación Neoplásica de la Expresión Génica , Humanos , Neoplasias Hepáticas/metabolismo , Células Madre Neoplásicas/metabolismo , Estabilidad del ARN , ARN Largo no Codificante/genética , ARN Mensajero/genética , ARN Mensajero/metabolismo , Proteínas de Unión al ARN/genética , Proteínas de Unión al ARN/metabolismo , Factores de Transcripción/metabolismo , beta Catenina/genética , beta Catenina/metabolismo
5.
Nat Commun ; 12(1): 7020, 2021 12 02.
Artículo en Inglés | MEDLINE | ID: mdl-34857746

RESUMEN

Silencing of a subset of germline genes is dependent upon DNA methylation (DNAme) post-implantation. However, these genes are generally hypomethylated in the blastocyst, implicating alternative repressive pathways before implantation. Indeed, in embryonic stem cells (ESCs), an overlapping set of genes, including germline "genome-defence" (GGD) genes, are upregulated following deletion of the H3K9 methyltransferase SETDB1 or subunits of the non-canonical PRC1 complex PRC1.6. Here, we show that in pre-implantation embryos and naïve ESCs (nESCs), hypomethylated promoters of germline genes bound by the PRC1.6 DNA-binding subunits MGA/MAX/E2F6 are enriched for RING1B-dependent H2AK119ub1 and H3K9me3. Accordingly, repression of these genes in nESCs shows a greater dependence on PRC1.6 than DNAme. In contrast, GGD genes are hypermethylated in epiblast-like cells (EpiLCs) and their silencing is dependent upon SETDB1, PRC1.6/RING1B and DNAme, with H3K9me3 and DNAme establishment dependent upon MGA binding. Thus, GGD genes are initially repressed by PRC1.6, with DNAme subsequently engaged in post-implantation embryos.


Asunto(s)
Factores de Transcripción Básicos con Cremalleras de Leucinas y Motivos Hélice-Asa-Hélice/genética , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/genética , Factor de Transcripción E2F6/genética , Regulación del Desarrollo de la Expresión Génica , N-Metiltransferasa de Histona-Lisina/genética , Histonas/genética , Proteínas del Grupo Polycomb/genética , Animales , Factores de Transcripción Básicos con Cremalleras de Leucinas y Motivos Hélice-Asa-Hélice/metabolismo , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/metabolismo , Metilación de ADN , Factor de Transcripción E2F6/metabolismo , Implantación del Embrión , Embrión de Mamíferos , Epigénesis Genética , Femenino , Silenciador del Gen , Genes Reporteros , Proteínas Fluorescentes Verdes/genética , Proteínas Fluorescentes Verdes/metabolismo , N-Metiltransferasa de Histona-Lisina/metabolismo , Histonas/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Células Madre Embrionarias de Ratones/citología , Células Madre Embrionarias de Ratones/metabolismo , Complejo Represivo Polycomb 1/genética , Complejo Represivo Polycomb 1/metabolismo , Proteínas del Grupo Polycomb/metabolismo , Subunidades de Proteína/genética , Subunidades de Proteína/metabolismo , Transducción de Señal , Ubiquitina-Proteína Ligasas/genética , Ubiquitina-Proteína Ligasas/metabolismo
6.
Cancer Lett ; 518: 266-277, 2021 10 10.
Artículo en Inglés | MEDLINE | ID: mdl-34339800

RESUMEN

Dysregulated ubiquitination of tumor-related proteins plays a critical role in tumor development and progression. The deubiquitinase USP22 is aberrantly expressed in certain types of cancer and contributes to aggressive tumor progression. However, the precise mechanism underlying the pro-tumorigenic function of USP22 in hepatocellular carcinoma (HCC) remains unclear. Here, we report that E2F6, a pocket protein-independent transcription repressor, is essential for HCC cell growth, and that its activities are controlled by USP22-mediated deubiquitination. USP22 interacts with and stabilizes E2F6, resulting in the transcriptional repression of phosphatase DUSP1. Moreover, the process involving DUSP1 repression by E2F6 strengthens AKT activation in HCC cells. Therefore, these findings provide mechanistic insights into the USP22-mediated control of oncogenic AKT signaling, emphasizing the importance of USP22-E2F6 regulation in HCC development.


Asunto(s)
Carcinoma Hepatocelular/genética , Proliferación Celular/genética , Factor de Transcripción E2F6/genética , Neoplasias Hepáticas/genética , Proteínas Proto-Oncogénicas c-akt/genética , Ubiquitina Tiolesterasa/genética , Ubiquitinación/genética , Animales , Carcinoma Hepatocelular/patología , Línea Celular , Línea Celular Tumoral , Expresión Génica/genética , Células HEK293 , Células Hep G2 , Humanos , Neoplasias Hepáticas/patología , Masculino , Ratones , Ratones Desnudos , Monoéster Fosfórico Hidrolasas/genética , Transducción de Señal/genética , Transcripción Genética/genética
7.
Oncol Rep ; 46(2)2021 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-34165171

RESUMEN

Long noncoding RNA SLC9A3 antisense RNA 1 (SLC9A3­AS1) plays a central role in lung cancer; yet, its functions in nasopharyngeal carcinoma (NPC) have not been elucidated. The present study revealed the roles of SLC9A3­AS1 in NPC and dissected the mechanisms downstream of SLC9A3­AS1. SLC9A3­AS1 levels in NPC were assessed by applying RT­qPCR. The modulatory role of SLC9A3­AS1 interference on NPC cells was examined using numerous functional experiments. High expression of SLC9A3­AS1 was observed in NPC samples. Patients with NPC with a high level of SLC9A3­AS1 experienced a shorter overall survival than those with a low SLC9A3­AS1 level. Loss of SLC9A3­AS1 reduced NPC cell proliferation, colony formation, migration, and invasion but induced cell apoptosis in vitro. Animal experiments further revealed that the depletion of SLC9A3­AS1 hindered NPC tumour growth in vivo. As a competitive endogenous RNA, SLC9A3­AS1 sponged microRNA­486­5p (miR­486­5p), consequently upregulating E2F transcription factor 6 (E2F6). Finally, the effects of SLC9A3­AS1 silencing on NPC cells were reversed by inhibiting miR­486­5p or overexpressing E2F6. In summary, SLC9A3­AS1 exerted carcinogenic effects on NPC cells by adjusting the miR­486­5p/E2F6 axis. Accordingly, the newly identified SLC9A3­AS1/miR­486­5p/E2F6 pathway may offer attractive therapeutic targets for future development.


Asunto(s)
Factor de Transcripción E2F6/genética , MicroARNs/genética , Carcinoma Nasofaríngeo/patología , Neoplasias Nasofaríngeas/patología , ARN Largo no Codificante/genética , Regulación hacia Arriba , Adulto , Animales , Estudios de Casos y Controles , Línea Celular Tumoral , Movimiento Celular , Proliferación Celular , Femenino , Regulación Neoplásica de la Expresión Génica , Humanos , Masculino , Ratones , Persona de Mediana Edad , Carcinoma Nasofaríngeo/genética , Neoplasias Nasofaríngeas/genética , Trasplante de Neoplasias , Análisis de Supervivencia
8.
Nat Commun ; 12(1): 3582, 2021 06 11.
Artículo en Inglés | MEDLINE | ID: mdl-34117224

RESUMEN

In mouse development, long-term silencing by CpG island DNA methylation is specifically targeted to germline genes; however, the molecular mechanisms of this specificity remain unclear. Here, we demonstrate that the transcription factor E2F6, a member of the polycomb repressive complex 1.6 (PRC1.6), is critical to target and initiate epigenetic silencing at germline genes in early embryogenesis. Genome-wide, E2F6 binds preferentially to CpG islands in embryonic cells. E2F6 cooperates with MGA to silence a subgroup of germline genes in mouse embryonic stem cells and in embryos, a function that critically depends on the E2F6 marked box domain. Inactivation of E2f6 leads to a failure to deposit CpG island DNA methylation at these genes during implantation. Furthermore, E2F6 is required to initiate epigenetic silencing in early embryonic cells but becomes dispensable for the maintenance in differentiated cells. Our findings elucidate the mechanisms of epigenetic targeting of germline genes and provide a paradigm for how transient repression signals by DNA-binding factors in early embryonic cells are translated into long-term epigenetic silencing during mouse development.


Asunto(s)
Islas de CpG/genética , Factor de Transcripción E2F6/genética , Factor de Transcripción E2F6/metabolismo , Desarrollo Embrionario/genética , Epigénesis Genética , Células Germinativas/metabolismo , Animales , Sitios de Unión , Sistemas CRISPR-Cas , Diferenciación Celular , Metilación de ADN , Silenciador del Gen , Ratones , Ratones Noqueados , Células Madre Embrionarias de Ratones , Complejo Represivo Polycomb 1/metabolismo , ARN Interferente Pequeño
9.
Bioengineered ; 12(1): 927-936, 2021 12.
Artículo en Inglés | MEDLINE | ID: mdl-33734009

RESUMEN

Countless studies have demonstrated that Circular RNAs (circRNAs) exert vital effects in regulating tumorigenesis of various cancers. CircRNA ZNF609 (circ-ZNF609) has been reported as an oncogene in various human cancers. Nevertheless, its regulating effect in cervical cancer (CC) remains to be further explored. RT-qPCR was adopted to measure circ-ZNF609, miR-197-3p and E2F6 levels. CC cell proliferation, migration and invasion were analyzed via CCK-8 and transwell assays. Dual-luciferase reporter assay was adopted to confirm the interaction between miR-197-3p and circ-ZNF609 or E2F6. In the present study, it was found that circ-ZNF609 was elevated in CC tissues and cell lines, and circ-ZNF609 deletion repressed cell viability, migration and invasion in CC. Moreover, circ-ZNF609 was identified to negatively regulate miR-197-3p expression in CC cells. The inhibition of miR-197-3p abrogated the inhibitory effect on CC cell proliferation, migration and invasion induced by circ-ZNF609 knockdown. Additionally, we further demonstrated that circ-ZNF609 upregulated E2F6 by interacting with miR-197-3p. Finally, rescue assays indicated that E2F6 overexpression upended the suppression of CC progression induced by circ-ZNF609 deletion. In conclusion, circ-ZNF609 promoted CC progression through modulating the miR-197-3p/E2F6 axis as an oncogene. This finding offers a unique insight into CC molecular mechanism and suggests a potential target for CC therapy.


Asunto(s)
Factor de Transcripción E2F6 , MicroARNs , ARN Circular , Neoplasias del Cuello Uterino , Animales , Línea Celular Tumoral , Proliferación Celular , Progresión de la Enfermedad , Factor de Transcripción E2F6/genética , Factor de Transcripción E2F6/metabolismo , Femenino , Técnicas de Silenciamiento del Gen , Humanos , Masculino , Ratones , Ratones Endogámicos BALB C , Ratones Desnudos , MicroARNs/genética , MicroARNs/metabolismo , ARN Circular/genética , ARN Circular/metabolismo , Reacción en Cadena en Tiempo Real de la Polimerasa , Neoplasias del Cuello Uterino/genética , Neoplasias del Cuello Uterino/metabolismo , Neoplasias del Cuello Uterino/patología
10.
FASEB J ; 34(9): 11382-11394, 2020 09.
Artículo en Inglés | MEDLINE | ID: mdl-32692467

RESUMEN

Ovarian cancer remains the sixth most frequently occurring cancer in women worldwide. Long noncoding RNAs (lncRNAs) are capable of regulating gene expression, and thus, participating in a wide range of biological functions and disease processes including cancer development. Our work suggests that lncRNA TMPO antisense RNA 1 (TMPO-AS1) represents an oncogenic lncRNA in ovarian cancer and presents a novel mechanism involving transcription factor E2F transcription factor 6 (E2F6) and lipocalin-2 (LCN2). We identified upregulated lncRNA TMPO-AS1 in ovarian cancer tissues and cells. siRNA-mediated silencing of lncRNA TMPO-AS1 restrained the aggressiveness of ovarian cancer cells and their pro-angiogenic ability, and reduced the expression of LCN2. LncRNA TMPO-AS1 was found to interact with E2F6, a transcriptional repressor that could bind to the promoter region of LCN2 gene. In addition, silencing of E2F6 or overexpression of LCN2 restored the aggressiveness of ovarian cancer cells and their pro-angiogenic ability following siRNA-mediated silencing of lncRNA TMPO-AS1. Taken together, we demonstrated lncRNA TMPO-AS1 could potentially promote LCN2 transcriptional activity by binding to transcription factor E2F6, and thus, stimulated the progression of ovarian cancer. These findings underscore a possible alternative therapeutic strategy for ovarian cancer treatment.


Asunto(s)
Carcinogénesis/genética , Regulación Neoplásica de la Expresión Génica , Lipocalina 2/genética , Neoplasias Ováricas/genética , ARN Largo no Codificante/genética , Animales , Línea Celular Tumoral , Factor de Transcripción E2F6/genética , Factor de Transcripción E2F6/metabolismo , Femenino , Humanos , Lipocalina 2/metabolismo , Ratones Endogámicos BALB C , Ratones Desnudos , Neoplasias Ováricas/metabolismo , Interferencia de ARN , Carga Tumoral/genética , Ensayos Antitumor por Modelo de Xenoinjerto/métodos
11.
Med Oncol ; 37(8): 68, 2020 Jul 24.
Artículo en Inglés | MEDLINE | ID: mdl-32710193

RESUMEN

CREB signaling is known for several decades, but how it regulates both positive and negative regulators of cell proliferation is not well understood. On the other hand functions of major epigenetic repressors such as DNMT3B, EZH2 and CUL4B for their repressive epigenetic modifications on chromatin have also been well studied. However, there is very limited information available on how these repressors are regulated at their transcriptional level. Here, using computational tools and molecular techniques including site directed mutagenesis, promoter reporter assay, chromatin immunoprecipitation (ChIP), we identified that CREB acts as a common transcription factor for DNMT3B, EZH2, CUL4B and E2F6. ChIP assay revealed that pCREB binds to promoters of these repressors at CREs and induce their transcription. As expected, the expression of these repressors and their associated repressive marks particularly H3K27me3 and H2AK119ub are increased and decreased upon CREB overexpression and knock-down conditions respectively in the cancer cells indicating that CREB regulates the functions of these repressors by activating their transcription. Since CREB and these epigenetic repressors are overexpressed in various cancer types, our findings showed the molecular relationship between them and indicate that CREB is an important therapeutic target for cancer therapy.


Asunto(s)
Proteínas Cullin/metabolismo , Proteína de Unión a Elemento de Respuesta al AMP Cíclico/metabolismo , ADN (Citosina-5-)-Metiltransferasas/metabolismo , Proteína Potenciadora del Homólogo Zeste 2/metabolismo , Neoplasias/metabolismo , Línea Celular , Línea Celular Tumoral , Biología Computacional/métodos , Proteínas Cullin/genética , Proteína de Unión a Elemento de Respuesta al AMP Cíclico/genética , ADN (Citosina-5-)-Metiltransferasas/genética , Factor de Transcripción E2F6/genética , Factor de Transcripción E2F6/metabolismo , Proteína Potenciadora del Homólogo Zeste 2/genética , Epigénesis Genética , Humanos , Neoplasias/genética , Neoplasias/patología , Regiones Promotoras Genéticas , Transducción de Señal , ADN Metiltransferasa 3B
12.
Mol Cell Biol ; 40(10)2020 04 28.
Artículo en Inglés | MEDLINE | ID: mdl-32094308

RESUMEN

Recently, abundant evidence has clarified that long noncoding RNAs (lncRNAs) play an oncogenic or anticancer role in the tumorigenesis and development of diverse human cancers. Described as a crucial regulator in some cancers, MIR22HG has not yet been studied in laryngocarcinoma and therefore the underlying regulatory role of MIR22HG in laryngocarcinoma is worth detecting. In this study, MIR22HG expression in laryngocarcinoma cells was confirmed to be downregulated, and upregulated MIR22HG expression led to suppressive effects on laryngocarcinoma cell proliferation and migration. Molecular mechanism assays revealed that MIR22HG sponges miR-5000-3p in laryngocarcinoma cells. Besides, decreased expression of miR-5000-3p suppressed laryngocarcinoma cell proliferation and migration. Moreover, the FBXW7 gene was reported to be a downstream target gene of miR-5000-3p in laryngocarcinoma cells. More importantly, rescue assays verified that FBXW7 depletion or miR-5000-3p upregulation countervailed the repressive effects of MIR22HG overexpression on laryngocarcinoma progression. In addition, E2F6 was proved to be capable of inhibiting MIR22HG transcription in laryngocarcinoma cells. To sum up, E2F6-induced downregulation of MIR22HG promotes laryngocarcinoma progression through the miR-5000-3p/FBXW7 axis.


Asunto(s)
Factor de Transcripción E2F6/genética , Proteína 7 que Contiene Repeticiones F-Box-WD/genética , Neoplasias Laríngeas/genética , MicroARNs/genética , Línea Celular Tumoral , Proliferación Celular , Progresión de la Enfermedad , Regulación hacia Abajo , Regulación Neoplásica de la Expresión Génica , Humanos , Neoplasias Laríngeas/patología , Invasividad Neoplásica/genética , Invasividad Neoplásica/patología
13.
Biosci Rep ; 40(1)2020 01 31.
Artículo en Inglés | MEDLINE | ID: mdl-31682716

RESUMEN

Cervical cancer (CC) is a prevalent gynecological cancer, and the patients with CC usually suffer from dismal prognosis. Long non-coding RNAs (lncRNAs) are demonstrated to serve as promising biological targets in human cancers. Gastric carcinoma proliferation enhancing transcript 1 (GHET1) has been revealed to function as an oncogene in several cancers, but it has never been investigated in CC. We proposed to examine the biological role of GHET1 in CC and the underlying mechanism and validated the up-regulated expression of GHET1 in CC cell lines. Loss-of-function assays demonstrated that down-regulation of GHET1 inhibited cell growth, migration and epithelial-to-mesenchymal transition (EMT) in CC. Furthermore, we validated that GHET1 down-regulation could inactivate AKT/mTOR and Wnt/ß-catenin pathways, and that respective activation of these two pathways abrogated the inhibitive effect of GHET1 knockdown on CC cell growth, migration and EMT. Moreover, we unfolded a preliminary investigation on the modulation of GHET1 on AKT/mTOR and Wnt/ß-catenin pathways. We found that GHET1 stabilized E2F6 mRNA through interacting with IGF2BP2, so as to regulate the activity of AKT/mTOR and Wnt/ß-catenin pathways. Rescue assays also proved that GHET1 regulated these two pathways and CC cell growth, migration and EMT through E2F6. In conclusion, we revealed that down-regulation of GHET1 suppresses cervical cancer progression through regulating AKT/mTOR and Wnt/ß-catenin signaling pathways, indicating GHET1 as a promising molecular biomarker for CC treatment improvement.


Asunto(s)
Proteínas Proto-Oncogénicas c-akt/metabolismo , ARN Largo no Codificante/metabolismo , Serina-Treonina Quinasas TOR/metabolismo , Neoplasias del Cuello Uterino/enzimología , Vía de Señalización Wnt , Movimiento Celular , Proliferación Celular , Progresión de la Enfermedad , Factor de Transcripción E2F6/genética , Factor de Transcripción E2F6/metabolismo , Transición Epitelial-Mesenquimal , Femenino , Regulación Neoplásica de la Expresión Génica , Células HeLa , Humanos , Invasividad Neoplásica , ARN Largo no Codificante/genética , Proteínas de Unión al ARN/genética , Proteínas de Unión al ARN/metabolismo , Neoplasias del Cuello Uterino/genética , Neoplasias del Cuello Uterino/patología
14.
Mol Cell ; 74(5): 1037-1052.e7, 2019 06 06.
Artículo en Inglés | MEDLINE | ID: mdl-31029542

RESUMEN

Polycomb repressive complexes 1 and 2 (PRC1 and PRC2) control cell identity by establishing facultative heterochromatin repressive domains at common sets of target genes. PRC1, which deposits H2Aub1 through the E3 ligases RING1A/B, forms six biochemically distinct subcomplexes depending on the assembled PCGF protein (PCGF1-PCGF6); however, it is yet unclear whether these subcomplexes have also specific activities. Here we show that PCGF1 and PCGF2 largely compensate for each other, while other PCGF proteins have high levels of specificity for distinct target genes. PCGF2 associates with transcription repression, whereas PCGF3 and PCGF6 associate with actively transcribed genes. Notably, PCGF3 and PCGF6 complexes can assemble and be recruited to several active sites independently of RING1A/B activity (therefore, of PRC1). For chromatin recruitment, the PCGF6 complex requires the combinatorial activities of its MGA-MAX and E2F6-DP1 subunits, while PCGF3 requires an interaction with the USF1 DNA binding transcription factor.


Asunto(s)
Complejo Represivo Polycomb 1/genética , Transcripción Genética , Ubiquitina-Proteína Ligasas/genética , Animales , Factores de Transcripción Básicos con Cremalleras de Leucinas y Motivos Hélice-Asa-Hélice/genética , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico , Cromatina/genética , Proteínas de Unión al ADN/genética , Factor de Transcripción E2F6/genética , Heterocromatina/genética , Ratones , Células Madre Embrionarias de Ratones/metabolismo , Complejo Represivo Polycomb 2/genética , Proteínas del Grupo Polycomb/genética , Proteínas Represoras/genética , Factor de Transcripción DP1/genética , Factores de Transcripción/genética , Factores Estimuladores hacia 5'/genética
15.
Biochem Biophys Res Commun ; 513(3): 560-566, 2019 06 04.
Artículo en Inglés | MEDLINE | ID: mdl-30981507

RESUMEN

Both type 1 and type 2 diabetes are associated with loss of functional beta cell mass, and strategies to restore beta cells are urgently needed. We reported previously that overexpression of the nuclear receptor TLX induces beta cell proliferation, but the underlying molecular mechanism has not been defined. Here, we identified direct targets of TLX in beta cells at the genome-wide level by ChIP-Seq. These targets include a cadre of regulators that are known to be critical for proliferation. Among these ChIP targets, E2F6 was tightly associated with the cell cycle modules, and thus, we further analyzed E2F6 expression and function in beta cells. We showed that E2F6 is strongly downregulated by TLX, and its expression inhibits beta cell proliferation. Moreover, coexpression of E2F6 with TLX partially abrogated the proliferative effects of TLX. These results strongly suggest that TLX acts through E2F6 to regulate beta cell proliferation. Together, the results of this study reveal a direct interaction between TLX and E2F6 and suggest new targets for the expansion of functional beta cell mass.


Asunto(s)
Factor de Transcripción E2F6/metabolismo , Células Secretoras de Insulina/metabolismo , Receptores Citoplasmáticos y Nucleares/metabolismo , Animales , Línea Celular , Proliferación Celular , Factor de Transcripción E2F6/genética , Factor de Transcripción E2F6/fisiología , Regulación de la Expresión Génica , Genoma , Células Secretoras de Insulina/citología , Ratones , Regiones Promotoras Genéticas
16.
Cancer Sci ; 110(3): 1085-1095, 2019 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-30582655

RESUMEN

Ovarian cancer is the most lethal cancer of the female reproductive system. In that regard, several epidemiological studies suggest that long-term exposure to estrogen could increase ovarian cancer risk, although its precise role remains controversial. To decipher a mechanism for this, we previously generated a mathematical model of how estrogen-mediated upregulation of the transcription factor, E2F6, upregulates the ovarian cancer stem/initiating cell marker, c-Kit, by epigenetic silencing the tumor suppressor miR-193a, and a competing endogenous (ceRNA) mechanism. In this study, we tested that previous mathematical model, showing that estrogen treatment of immortalized ovarian surface epithelial cells upregulated both E2F6 and c-KIT, but downregulated miR-193a. Luciferase assays further confirmed that microRNA-193a targets both E2F6 and c-Kit. Interestingly, ChIP-PCR and bisulphite pyrosequencing showed that E2F6 also epigenetically suppresses miR-193a, through recruitment of EZH2, and by a complex ceRNA mechanism in ovarian cancer cell lines. Importantly, cell line and animal experiments both confirmed that E2F6 promotes ovarian cancer stemness, whereas E2F6 or EZH2 depletion derepressed miR-193a, which opposes cancer stemness, by alleviating DNA methylation and repressive chromatin. Finally, 118 ovarian cancer patients with miR-193a promoter hypermethylation had poorer survival than those without hypermethylation. These results suggest that an estrogen-mediated E2F6 ceRNA network epigenetically and competitively inhibits microRNA-193a activity, promoting ovarian cancer stemness and tumorigenesis.


Asunto(s)
Factor de Transcripción E2F6/genética , Células Madre Neoplásicas/patología , Neoplasias Ováricas/genética , ARN/genética , Transcripción Genética/genética , Animales , Línea Celular Tumoral , Metilación de ADN/efectos de los fármacos , Metilación de ADN/genética , Epigénesis Genética/efectos de los fármacos , Epigénesis Genética/genética , Células Epiteliales/efectos de los fármacos , Células Epiteliales/patología , Estrógenos/efectos adversos , Femenino , Genes Supresores de Tumor/fisiología , Humanos , Ratones , Ratones Endogámicos BALB C , Ratones Desnudos , Ratones SCID , MicroARNs/genética , Células Madre Neoplásicas/efectos de los fármacos , Neoplasias Ováricas/etiología , Transcripción Genética/efectos de los fármacos , Regulación hacia Arriba/efectos de los fármacos , Regulación hacia Arriba/genética
17.
Eur Rev Med Pharmacol Sci ; 22(19): 6300-6307, 2018 10.
Artículo en Inglés | MEDLINE | ID: mdl-30338798

RESUMEN

OBJECTIVE: To investigate the effect of miR-425 on the proliferation and apoptosis of clear cell renal carcinoma (ccRCA) cells, and to explore the underlying mechanism. PATIENTS AND METHODS: A total of 80 pairs of human clear cell renal carcinoma (ccRCA) and cancer-adjacent normal tissue samples were collected in this study. Human ccRCA cell line (786-O) and normal human kidney cell line (HK-2) were used in cellular research. The expression level of miR-425 was detected in ccRCA tissues and cells, respectively. Target genes of miR-425 were predicted by bioinformatics and verified by luciferase reporter gene assay. Moreover, the role of miR-425 in regulating E2F6 as well as its effect on the proliferation and apoptosis of ccRCA cells were detected. RESULTS: Quantitative reverse transcriptase-polymerase chain reaction (qRT-PCR) results showed that the expression of miR-425 was significantly decreased in ccRCA tissues and cells. The proliferation ability and cell cycle of 786-O cells were significantly inhibited after miR-425 overexpression. The percentage of cells in G0/G1 phase was remarkably increased, while the percentage of cells in S and G2/M phases was significantly decreased. Besides, the number of apoptotic cells was significantly increased in the miR-425 intervention group. On-line target gene prediction software indicated that E2F6 was the potential downstream target gene of miR-425. RT-PCR, Western blotting and luciferase reporter gene assay demonstrated that the expression of E2F6 was negatively regulated by miR-425. In addition, subsequent experiments showed that the up-regulation of E2F6 could suppress the inhibitory effect of miR-425 on the proliferation and apoptosis of ccRCA cells. CONCLUSIONS: Our research demonstrated the inhibitory function of miR-425 in ccRCA. Therefore, the miR-425/E2F6 axis was expected to be one of the targets of ccRCA targeted therapy.


Asunto(s)
Carcinoma de Células Renales/genética , Factor de Transcripción E2F6/genética , Neoplasias Renales/genética , MicroARNs/genética , Apoptosis , Ciclo Celular , Línea Celular Tumoral , Proliferación Celular , Progresión de la Enfermedad , Regulación hacia Abajo , Regulación Neoplásica de la Expresión Génica , Humanos
18.
PLoS Genet ; 14(1): e1007193, 2018 01.
Artículo en Inglés | MEDLINE | ID: mdl-29381691

RESUMEN

Diverse Polycomb repressive complexes 1 (PRC1) play essential roles in gene regulation, differentiation and development. Six major groups of PRC1 complexes that differ in their subunit composition have been identified in mammals. How the different PRC1 complexes are recruited to specific genomic sites is poorly understood. The Polycomb Ring finger protein PCGF6, the transcription factors MGA and E2F6, and the histone-binding protein L3MBTL2 are specific components of the non-canonical PRC1.6 complex. In this study, we have investigated their role in genomic targeting of PRC1.6. ChIP-seq analysis revealed colocalization of MGA, L3MBTL2, E2F6 and PCGF6 genome-wide. Ablation of MGA in a human cell line by CRISPR/Cas resulted in complete loss of PRC1.6 binding. Rescue experiments revealed that MGA recruits PRC1.6 to specific loci both by DNA binding-dependent and by DNA binding-independent mechanisms. Depletion of L3MBTL2 and E2F6 but not of PCGF6 resulted in differential, locus-specific loss of PRC1.6 binding illustrating that different subunits mediate PRC1.6 loading to distinct sets of promoters. Mga, L3mbtl2 and Pcgf6 colocalize also in mouse embryonic stem cells, where PRC1.6 has been linked to repression of germ cell-related genes. Our findings unveil strikingly different genomic recruitment mechanisms of the non-canonical PRC1.6 complex, which specify its cell type- and context-specific regulatory functions.


Asunto(s)
Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/fisiología , ADN/metabolismo , Factor de Transcripción E2F6/fisiología , Proteínas Nucleares/fisiología , Complejo Represivo Polycomb 1/metabolismo , Factores de Transcripción/fisiología , Animales , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/genética , Células Cultivadas , Factor de Transcripción E2F6/genética , Regulación de la Expresión Génica , Técnicas de Silenciamiento del Gen , Células HEK293 , Células Madre Embrionarias Humanas/fisiología , Humanos , Ratones , Células Madre Embrionarias de Ratones/fisiología , Proteínas Nucleares/genética , Unión Proteica/genética , Factores de Transcripción/genética
19.
Oncogene ; 37(3): 332-340, 2018 01 18.
Artículo en Inglés | MEDLINE | ID: mdl-28925402

RESUMEN

MDM2, an E3 ubiquitin ligase, is a potent inhibitor of the p53 tumor suppressor and is elevated in many human cancers that retain wild-type p53. MDM2 SNP309G is a functional polymorphism that results in elevated levels of MDM2 (due to enhanced SP1 binding to the MDM2 promoter) thus decreasing p53 activity. Mdm2SNP309G/G mice are more prone to spontaneous tumor formation than Mdm2SNP309T/T mice, providing direct evidence for the impact of this SNP in tumor development. We asked whether environmental factors impact SNP309G function and show that SNP309G cooperates with ionizing radiation to exacerbate tumor development. Surprisingly, ultraviolet B light or Benzo(a)pyrene exposure of skin shows that SNP309G allele actually protects against squamous cell carcinoma susceptibility. These contrasting differences led us to interrogate the mechanism by which Mdm2 SNP309 regulates tumor susceptibility in a tissue-specific manner. Although basal Mdm2 levels were significantly higher in most tissues in Mdm2SNP309G/G mice compared with Mdm2SNP309T/T mice, they were significantly lower in Mdm2SNP309G/G keratinocytes, the cell-type susceptible to squamous cell carcinoma. The assessment of potential transcriptional regulators in ENCODE ChIP-seq database identified transcriptional repressor E2F6 as a possible negative regulator of MDM2 expression. Our data show that E2F6 suppresses Mdm2 expression in cells harboring the SNP309G allele but not the SNP309T allele. Thus, Mdm2 SNP309G exhibits tissue-specific regulation and differentially impacts cancer risk.


Asunto(s)
Carcinoma de Células Escamosas/genética , Factor de Transcripción E2F6/metabolismo , Predisposición Genética a la Enfermedad , Proteínas Proto-Oncogénicas c-mdm2/genética , Neoplasias Cutáneas/genética , Alelos , Animales , Carcinógenos/toxicidad , Carcinoma de Células Escamosas/etiología , Carcinoma de Células Escamosas/mortalidad , Carcinoma de Células Escamosas/patología , Supervivencia sin Enfermedad , Factor de Transcripción E2F6/genética , Femenino , Queratinocitos , Masculino , Ratones , Ratones Endogámicos C57BL , Neoplasias Experimentales/etiología , Neoplasias Experimentales/genética , Fenotipo , Polimorfismo de Nucleótido Simple , Cultivo Primario de Células , Factores Sexuales , Piel/citología , Piel/efectos de los fármacos , Piel/patología , Piel/efectos de la radiación , Neoplasias Cutáneas/etiología , Neoplasias Cutáneas/mortalidad , Neoplasias Cutáneas/patología , Rayos Ultravioleta/efectos adversos
20.
Cell Signal ; 40: 230-238, 2017 12.
Artículo en Inglés | MEDLINE | ID: mdl-28964969

RESUMEN

The E2F/Rb pathway regulates cell growth, differentiation, and death. In particular, E2F1 promotes apoptosis in all cells including those of the heart. E2F6, which represses E2F activity, was found to induce dilated cardiomyopathy in the absence of apoptosis in murine post-natal heart. Here we evaluate the anti-apoptotic potential of E2F6 in neonatal cardiomyocytes (NCM) from E2F6-Tg hearts which showed significantly less caspase-3 cleavage, a lower Bax/Bcl2 ratio, and improved cell viability in response to CoCl2 exposure. This correlated with a decrease in the pro-apoptotic E2F3 protein levels. In contrast, no difference in apoptotic markers or cell viability was observed in response to Doxorubicin (Dox) treatment between Wt and Tg-NCM. Dox caused a rapid and dramatic loss of the E2F6 protein in Tg-NCM within 6h and was undetectable after 12h. The level of e2f6 transcript was unchanged in Wt NCM, but was dramatically decreased in Tg cells in response to both Dox and CoCl2. This was related to an impact of the drugs on the α-myosin heavy chain promoter used to drive the E2F6 transgene. By comparison in HeLa, Dox induced apoptosis through upregulation of endogenous E2F1 involving post-transcriptional mechanisms, while E2F6 was down regulated with induction of the Checkpoint kinase-1 and proteasome degradation. These data imply that E2F6 serves to modulate E2F activity and protect cells including cardiomyocytes from apoptosis and improve survival. Strategies to modulate E2F6 levels may be therapeutically useful to mitigate cell death associated disorders.


Asunto(s)
Apoptosis/genética , Factor de Transcripción E2F1/genética , Factor de Transcripción E2F6/genética , Miocitos Cardíacos/metabolismo , Animales , Animales Recién Nacidos , Apoptosis/efectos de los fármacos , Cardiomiopatía Dilatada , Caspasa 3/genética , Diferenciación Celular/genética , Proliferación Celular/genética , Doxorrubicina/administración & dosificación , Factor de Transcripción E2F3/genética , Regulación del Desarrollo de la Expresión Génica/efectos de los fármacos , Células HeLa , Humanos , Ratones , Miocitos Cardíacos/efectos de los fármacos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...