Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 99
Filtrar
1.
J Med Virol ; 96(4): e29580, 2024 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-38566572

RESUMEN

A persistent infection with human papillomavirus (HPV) can induce precancerous lesions of the cervix that may ultimately develop into cancer. Cervical cancer development has been linked to altered microRNA (miRNA) expression, with miRNAs regulating anchorage-independent growth being particularly important for the progression of precancerous lesions to cancer. In this study, we set out to identify and validate targets of miR-129-5p, a previously identified tumor suppressive miRNA involved in anchorage-independent growth and HPV-induced carcinogenesis. We predicted 26 potential miR-129-5p targets using online databases, followed by KEGG pathway enrichment analysis. RT-qPCR and luciferase assays confirmed that 3'UTR regions of six genes (ACTN1, BMPR2, CAMK4, ELK4, EP300, and GNAQ) were targeted by miR-129-5p. Expressions of ACTN1, CAMK4, and ELK4 were inversely correlated to miR-129-5p expression in HPV-transformed keratinocytes, and their silencing reduced anchorage-independent growth. Concordantly, miR-129-5p overexpression decreased protein levels of ACTN1, BMPR2, CAMK4 and ELK4 in anchorage-independent conditions. Additionally, c-FOS, a downstream target of ELK4, was downregulated upon miR-129-5p overexpression, suggesting regulation through the ELK4/c-FOS axis. ACTN1 and ELK4 expression was also upregulated in high-grade precancerous lesions and cervical cancers, supporting their clinical relevance. In conclusion, we identified six targets of miR-129-5p involved in the regulation of anchorage-independent growth, with ACTN1, BMPR2, ELK4, EP300, and GNAQ representing novel targets for miR-129-5p. For both ACTN1 and ELK4 functional and clinical relevance was confirmed, indicating that miR-129-5p-regulated ACTN1 and ELK4 expression contributes to HPV-induced carcinogenesis.


Asunto(s)
MicroARNs , Infecciones por Papillomavirus , Lesiones Precancerosas , Neoplasias del Cuello Uterino , Femenino , Humanos , Virus del Papiloma Humano , Infecciones por Papillomavirus/genética , Infecciones por Papillomavirus/patología , Línea Celular Tumoral , MicroARNs/genética , MicroARNs/metabolismo , Queratinocitos/metabolismo , Queratinocitos/patología , Carcinogénesis/genética , Carcinogénesis/patología , Lesiones Precancerosas/patología , Proliferación Celular/genética , Proteína Elk-4 del Dominio ets , Actinina/genética
2.
J Transl Med ; 19(1): 342, 2021 08 09.
Artículo en Inglés | MEDLINE | ID: mdl-34372882

RESUMEN

BACKGROUND: We tried to elaborate the molecular mechanism of ETS-like transcription factor 4 (ELK4) affecting gastric cancer (GC) progression through M2 polarization of macrophages mediated by lysine-specific demethylase 5A (KDM5A)-Praja2 (PJA2)-kinase suppressor of ras 1 (KSR1) axis. METHODS: GC expression dataset was obtained from GEO database, and the downstream regulatory mechanism of ELK4 was predicted. Tumor-associated macrophages (TAMs) were isolated from GC tissues. The interaction among ELK4, KDM5A, PJA2 and KSR1 was analyzed by dual luciferase reporter gene, ChIP and Co-IP assays. The stability of KSR1 protein was detected by cycloheximide (CHX) treatment. After TAMs were co-cultured with HGC-27 cells, HGC-27 cell biological processes were assessed through gain- and loss-of function assays. Tumorigenicity was detected by tumorigenicity test in nude mice. RESULTS: In GC and TAMs, ELK4, KDM5A and KSR1 were highly expressed, while PJA2 was lowly expressed. M2 polarization of macrophages promoted the development of GC. ELK4 activated KDM5A by transcription and promoted macrophage M2 polarization. KDM5A inhibited the expression of PJA2 by removing H3K4me3 of PJA2 promoter, which promoted M2 polarization of macrophages. PJA2 reduced KSR1 by ubiquitination. ELK4 promoted the proliferative, migrative and invasive potentials of GC cells as well as the growth of GC xenografts by regulating KSR1. CONCLUSION: ELK4 may reduce the PJA2-dependent inhibition of KSR1 by transcriptional activation of KDM5A to promote M2 polarization of macrophages, thus promoting the development of GC.


Asunto(s)
Neoplasias Gástricas , Animales , Línea Celular Tumoral , Humanos , Activación de Macrófagos , Macrófagos , Ratones , Ratones Desnudos , Proteína 2 de Unión a Retinoblastoma , Neoplasias Gástricas/genética , Activación Transcripcional , Ubiquitina-Proteína Ligasas , Proteína Elk-4 del Dominio ets
3.
Prostate ; 80(2): 198-208, 2020 02.
Artículo en Inglés | MEDLINE | ID: mdl-31794091

RESUMEN

BACKGROUND: Both hormone-sensitive and castration- and enzalutamide-resistant prostate cancers (PCa) depend on the ternary complex factor (TCF) protein ELK1 to serve as a tethering protein for the androgen receptor (AR) to activate a critical set of growth genes. The two sites in ELK1 required for AR binding are conserved in other members of the TCF subfamily, ELK3 and ELK4. Here we examine the potential utility of the three proteins as prognosticators of disease recurrence in PCa. METHODS: Transcriptional activity assays; Retrospective analysis of PCa recurrence using data on 501 patients in The Cancer Genome Atlas (TCGA) database; Unpaired Wilcoxon rank-sum test and multiple comparison correction using the Holm's method; Spearman's correlations; Kaplan-Meier methods; Univariable and multivariable Cox regression analyses; LASSO-based penalized Cox regression models; Time-dependent area under the receiver operating characteristic (ROC) curve. RESULTS: ELK4 but not ELK3 was coactivated by AR similar to ELK1. Tumor expression of neither ELK3 nor ELK4 was associated with disease-free survival (DFS). ELK1 was associated with higher clinical T-stage, pathology T-stage, Gleason score, prognostic grade, and positive lymph node status. ELK1 was a negative prognosticator of DFS, independent of ELK3, ELK4, clinical T-stage, pathology T-stage, prognostic grade, lymph node status, age, and race. Inclusion of ELK1 increased the abilities of the Oncotype DX and Prolaris gene panels to predict disease recurrence, correctly predicting disease recurrence in a unique subset of patients. CONCLUSIONS: ELK1 is a strong, independent prognosticator of disease recurrence in PCa, underscoring its unique role in PCa growth. Inclusion of ELK1 may enhance the utility of currently used prognosticators for clinical decision making in prostate cancer.


Asunto(s)
Recurrencia Local de Neoplasia/genética , Neoplasias de la Próstata/genética , Proteína Elk-1 con Dominio ets/genética , Adulto , Anciano , Análisis por Conglomerados , Supervivencia sin Enfermedad , Células HeLa , Humanos , Masculino , Persona de Mediana Edad , Recurrencia Local de Neoplasia/patología , Pronóstico , Neoplasias de la Próstata/patología , Proteínas Proto-Oncogénicas c-ets/genética , Receptores Androgénicos/genética , Estudios Retrospectivos , Activación Transcripcional , Proteína Elk-4 del Dominio ets/genética
4.
RNA Biol ; 17(9): 1293-1308, 2020 09.
Artículo en Inglés | MEDLINE | ID: mdl-32372707

RESUMEN

The blood-brain barrier (BBB) plays a pivotal role in the maintenance and regulation of the neural microenvironment. The BBB breakdown is a pathological change in early Alzheimer's disease (AD). RNA-binding proteins (RBPs) and long non-coding RNAs (lncRNAs) are involved in the regulation of BBB permeability. Our study demonstrates the role of TRA2A/LINC00662/ELK4 axis in regulating BBB permeability in AD microenvironment. In Aß1-42-incubated microvascular endothelial cells (ECs) of the BBB model in vitro, TRA2A and LINC00662 were enriched. TRA2A increased the stability of LINC00662 by binding with it. The knockdown of either TRA2A or LINC00662 decreased BBB permeability due to increased expression of tight junction-related proteins. ELK4 was less expressed in the BBB model in AD microenvironment in vitro. LINC00662 mediated the degradation of ELK4 mRNA by SMD pathway. Downregulation of ELK4 increased BBB permeability by increasing the tight junction-related protein expression.TRA2A/LINC00662/ELK4 axis plays a crucial role in the regulation of BBB permeability in AD microenvironment, which may provide a novel target for the therapy of AD.


Asunto(s)
Barrera Hematoencefálica/metabolismo , Microambiente Celular/genética , Regulación de la Expresión Génica , ARN Largo no Codificante/metabolismo , Proteínas de Unión al ARN/metabolismo , Proteína Elk-4 del Dominio ets/genética , Enfermedad de Alzheimer/etiología , Enfermedad de Alzheimer/metabolismo , Enfermedad de Alzheimer/patología , Péptidos beta-Amiloides/metabolismo , Biomarcadores , Células Endoteliales/metabolismo , Técnicas de Silenciamiento del Gen , Humanos , Fragmentos de Péptidos/metabolismo , Permeabilidad , Regiones Promotoras Genéticas , Unión Proteica , Interferencia de ARN , Estabilidad del ARN , ARN Largo no Codificante/genética , Proteínas de Uniones Estrechas/genética , Proteínas de Uniones Estrechas/metabolismo
5.
J Immunol ; 201(6): 1681-1691, 2018 09 15.
Artículo en Inglés | MEDLINE | ID: mdl-30068599

RESUMEN

In mouse thymocyte development, signaling by the TCR through the ERK pathway is required for positive selection of conventional naive T cells. The Ets transcription factor ELK4 (SAP-1), an ERK-regulated cofactor of the SRF transcription factor, plays an important role in positive selection by activating immediate-early genes such as the Egr transcription factor family. The role of ELK4-SRF signaling in development of other T cell types dependent on ERK signaling has been unclear. In this article, we show that ELK4, and its close relative ELK1, act cell autonomously in the thymus to control the generation of innate-like αß CD8+ T cells with memory-like characteristics. Mice lacking ELK4 and ELK1 develop increased numbers of innate-like αß CD8+ T cells, which populate the periphery. These cells develop cell autonomously rather than through expansion of PLZF+ thymocytes and concomitantly increased IL-4 signaling. Their development is associated with reduced TCR-mediated activation of ELK4-SRF target genes and can be partially suppressed by overexpression of the ELK4-SRF target gene EGR2. Consistent with this, partial inhibition of ERK signaling in peripheral CD8+T cells promotes the generation of cells with innate-like characteristics. These data establish that low-level ERK signaling through ELK4 (and ELK1) promotes innate-like αß CD8+ T cell differentiation, tuning conventional versus innate-like development.


Asunto(s)
Linfocitos T CD8-positivos/inmunología , Diferenciación Celular/inmunología , Sistema de Señalización de MAP Quinasas/inmunología , Timo/inmunología , Proteína Elk-1 con Dominio ets/inmunología , Proteína Elk-4 del Dominio ets/inmunología , Animales , Linfocitos T CD8-positivos/citología , Diferenciación Celular/genética , Inmunidad Innata , Sistema de Señalización de MAP Quinasas/genética , Ratones , Ratones Noqueados , Receptores de Antígenos de Linfocitos T/genética , Receptores de Antígenos de Linfocitos T/inmunología , Timo/citología , Proteína Elk-1 con Dominio ets/genética , Proteína Elk-4 del Dominio ets/genética
6.
J Biol Chem ; 292(32): 13296-13311, 2017 08 11.
Artículo en Inglés | MEDLINE | ID: mdl-28655758

RESUMEN

Sirtuin 7 (SIRT7), a member of the NAD+-dependent class III histone deacetylases, is involved in the regulation of various cellular processes and in resisting various stresses, such as hypoxia, low glucose levels, and DNA damage. Interestingly, SIRT7 is linked to the control of glycolysis, suggesting a role in glucose metabolism. Given the important roles of SIRT7, it is critical to clarify how SIRT7 activity is potentially regulated. It has been reported that some transcriptional and post-transcriptional regulatory mechanisms are involved. However, little is known how SIRT7 is regulated by the post-translational modifications. Here, we identified ubiquitin-specific peptidase 7 (USP7), a deubiquitinase, as a negative regulator of SIRT7. We showed that USP7 interacts with SIRT7 both in vitro and in vivo, and we further demonstrated that SIRT7 undergoes endogenous Lys-63-linked polyubiquitination, which is removed by USP7. Although the USP7-mediated deubiquitination of SIRT7 had no effect on its stability, the deubiquitination repressed its enzymatic activity. We also showed that USP7 coordinates with SIRT7 to regulate the expression of glucose-6-phosphatase catalytic subunit (G6PC), a gluconeogenic gene. USP7 depletion by RNA interference increased both G6PC expression and SIRT7 enzymatic activity. Moreover, SIRT7 targeted the G6PC promoter through the transcription factor ELK4 but not through forkhead box O1 (FoxO1). In summary, SIRT7 is a USP7 substrate and has a novel role as a regulator of gluconeogenesis. Our study may provide the basis for new clinical approaches to treat metabolic disorders related to glucose metabolism.


Asunto(s)
Regulación Enzimológica de la Expresión Génica , Glucosa-6-Fosfatasa/metabolismo , Regiones Promotoras Genéticas , Procesamiento Proteico-Postraduccional , Sirtuinas/metabolismo , Ubiquitina Tiolesterasa/metabolismo , Proteína Elk-4 del Dominio ets/metabolismo , Sustitución de Aminoácidos , Línea Celular Tumoral , Eliminación de Gen , Gluconeogénesis , Glucosa-6-Fosfatasa/antagonistas & inhibidores , Glucosa-6-Fosfatasa/genética , Células HEK293 , Humanos , Hidrólisis , Lisina/metabolismo , Mutación , Fragmentos de Péptidos/antagonistas & inhibidores , Fragmentos de Péptidos/genética , Fragmentos de Péptidos/metabolismo , Dominios y Motivos de Interacción de Proteínas , Subunidades de Proteína/genética , Subunidades de Proteína/metabolismo , Interferencia de ARN , Proteínas Recombinantes de Fusión/química , Proteínas Recombinantes de Fusión/metabolismo , Sirtuinas/antagonistas & inhibidores , Sirtuinas/genética , Especificidad por Sustrato , Ubiquitina Tiolesterasa/antagonistas & inhibidores , Ubiquitina Tiolesterasa/genética , Peptidasa Específica de Ubiquitina 7 , Ubiquitinación , Proteína Elk-4 del Dominio ets/genética
7.
Nature ; 487(7405): 114-8, 2012 Jul 05.
Artículo en Inglés | MEDLINE | ID: mdl-22722849

RESUMEN

Sirtuin proteins regulate diverse cellular pathways that influence genomic stability, metabolism and ageing. SIRT7 is a mammalian sirtuin whose biochemical activity, molecular targets and physiological functions have been unclear. Here we show that SIRT7 is an NAD(+)-dependent H3K18Ac (acetylated lysine 18 of histone H3) deacetylase that stabilizes the transformed state of cancer cells. Genome-wide binding studies reveal that SIRT7 binds to promoters of a specific set of gene targets, where it deacetylates H3K18Ac and promotes transcriptional repression. The spectrum of SIRT7 target genes is defined in part by its interaction with the cancer-associated E26 transformed specific (ETS) transcription factor ELK4, and comprises numerous genes with links to tumour suppression. Notably, selective hypoacetylation of H3K18Ac has been linked to oncogenic transformation, and in patients is associated with aggressive tumour phenotypes and poor prognosis. We find that deacetylation of H3K18Ac by SIRT7 is necessary for maintaining essential features of human cancer cells, including anchorage-independent growth and escape from contact inhibition. Moreover, SIRT7 is necessary for a global hypoacetylation of H3K18Ac associated with cellular transformation by the viral oncoprotein E1A. Finally, SIRT7 depletion markedly reduces the tumorigenicity of human cancer cell xenografts in mice. Together, our work establishes SIRT7 as a highly selective H3K18Ac deacetylase and demonstrates a pivotal role for SIRT7 in chromatin regulation, cellular transformation programs and tumour formation in vivo.


Asunto(s)
Transformación Celular Neoplásica/metabolismo , Histona Desacetilasas/metabolismo , Histonas/metabolismo , Lisina/metabolismo , Sirtuinas/metabolismo , Acetilación , Proteínas E1A de Adenovirus/genética , Proteínas E1A de Adenovirus/metabolismo , Animales , Secuencia de Bases , Sitios de Unión , Línea Celular Tumoral , Proliferación Celular , Transformación Celular Neoplásica/genética , Transformación Celular Neoplásica/patología , Cromatina/metabolismo , Inhibición de Contacto , Progresión de la Enfermedad , Humanos , Ratones , Trasplante de Neoplasias , Motivos de Nucleótidos , Fenotipo , Regiones Promotoras Genéticas , Proteínas Represoras/metabolismo , Sirtuinas/deficiencia , Sirtuinas/genética , Transcripción Genética , Trasplante Heterólogo , Proteína Elk-4 del Dominio ets/metabolismo
8.
Biochem J ; 474(9): 1509-1528, 2017 04 19.
Artículo en Inglés | MEDLINE | ID: mdl-28275114

RESUMEN

The adapter protein Dok-4 (downstream of kinase-4) has been reported as both an activator and inhibitor of Erk and Elk-1, but lack of knowledge about the identity of its partner molecules has precluded any mechanistic insight into these seemingly conflicting properties. We report that Dok-4 interacts with the transactivation domain of Elk-4 through an atypical phosphotyrosine-binding domain-mediated interaction. Dok-4 possesses a nuclear export signal and can relocalize Elk-4 from nucleus to cytosol, whereas Elk-4 possesses two nuclear localization signals that restrict interaction with Dok-4. The Elk-4 protein, unlike Elk-1, is highly unstable in the presence of Dok-4, through both an interaction-dependent mechanism and a pleckstrin homology domain-dependent but interaction-independent mechanism. This is reversed by proteasome inhibition, depletion of endogenous Dok-4 or lysine-to-arginine mutation of putative Elk-4 ubiquitination sites. Finally, Elk-4 transactivation is potently inhibited by Dok-4 overexpression but enhanced by Dok-4 knockdown in MDCK renal tubular cells, which correlates with increased basal and EGF-induced expression of Egr-1, Fos and cylcinD1 mRNA, and cell proliferation despite reduced Erk activation. Thus, Dok-4 can target Elk-4 activity through multiple mechanisms, including binding of the transactivation domain, nuclear exclusion and protein destabilization, without a requirement for inhibition of Erk.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/genética , Señales de Exportación Nuclear/genética , Señales de Localización Nuclear/genética , Proteína Elk-4 del Dominio ets/genética , Transporte Activo de Núcleo Celular/genética , Proteínas Adaptadoras Transductoras de Señales/metabolismo , Secuencia de Aminoácidos , Animales , Células COS , Proliferación Celular/genética , Perros , Regulación de la Expresión Génica , Células HEK293 , Humanos , Immunoblotting , Células de Riñón Canino Madin Darby , Ratones , Microscopía Confocal , Unión Proteica , Interferencia de ARN , Homología de Secuencia de Aminoácido , Técnicas del Sistema de Dos Híbridos , Proteína Elk-4 del Dominio ets/metabolismo
9.
Genomics ; 109(2): 75-82, 2017 03.
Artículo en Inglés | MEDLINE | ID: mdl-28189763

RESUMEN

DNA adenine methyltransferase identification (DamID) is an enzymatic technology for detecting DNA regions targeted by chromatin-associated proteins. Proteins are fused to bacterial DNA adenine methyltransferase (Dam) and expressed in cultured cells or whole organisms. Here, we used DamID to detect DNA regions bound by the cardiac-restricted transcription factors (TFs) NKX2-5 and SRF, and ubiquitously-expressed co-factors ELK1 and ELK4. We compared targets bound by these TFs as N- and C-terminal fusions with Dam, for both wild type (WT) NKX2-5 and mutant proteins mimicking those found in congenital heart disease. Overall, DamID is highly robust: while the orientation of WT Dam fusions can affect the size of the target sets, their signatures remained largely reproducible. Furthermore, a severe NKX2-5 mutant lacking the homeodomain showed strong steric effects negatively impacting target discovery. The extent of steric effect is likely to be dependent on the protein in question and the orientation of Dam fusion.


Asunto(s)
Cromatina/metabolismo , Regulación de la Expresión Génica , Técnicas Genéticas , Cardiopatías Congénitas/metabolismo , Metiltransferasa de ADN de Sitio Específico (Adenina Especifica) , Animales , ADN/metabolismo , Cardiopatías Congénitas/genética , Proteína Homeótica Nkx-2.5/metabolismo , Humanos , Ratones , Factor de Respuesta Sérica/metabolismo , Proteína Elk-1 con Dominio ets/metabolismo , Proteína Elk-4 del Dominio ets/metabolismo
10.
Pharmazie ; 72(11): 687-693, 2017 Nov 01.
Artículo en Inglés | MEDLINE | ID: mdl-29442044

RESUMEN

We aimed to elucidate the roles and regulatory mechanism of miR-3188 in oxidized low-density lipoprotein (ox-LDL)-induced cell injury in THP-1 derived macrophages, thus providing a new insight for the treatment of atherosclerosis (AS). A total of 85 AS patients and 45 healthy controls were enrolled. The levels of miR-3188 and lipoprotein-associated phospholipase A2 (Lp-PLA2) in AS patients and healthy controls were detected. Then ox-LDL was used to treat human THP-1 derived macrophages. The effects of overexpression and suppression of miR-3188 on regulating ox-LDL-induced cell injury in THP-1 derived macrophages were investigated. Additionally, the potential target of miR-3188 was identified, which was verified by luciferase reporter assay. Besides, the relationship between miR-3188 and RhoA/ROCK pathway was explored. miR-3188 was downregulated in AS patients, while the levels of Lp-PLA2 in AS patients were increased. Ox-LDL significantly induced cell injury by decreasing cell viability, inducing cell apoptosis and increasing the production of inflammatory cytokines, including IL-1ß, IL-6, MCP-1 and TNF-α. In addition, miR-3188 was significantly downregulated after ox-LDL treatment. Overexpression of miR-3188 alleviated ox-LDL-induced cell injury, while inhibition of miR-3188 had opposite effects. ETS-domain protein 4 (ELK4) was a target of miR-3188. The effects of miR-3188 inhibition on ox-LDL-induced cell injury were markedly reversed by knockdown of ELK4. Besides, inhibition of miR-3188 enhanced ox-LDL-activated RhoA/ROCK pathway, while knockdown of ELK4 suppressed this pathway. Downregulation of miR-3188 may contribute to AS development via negatively regulating Lp-PLA2, targeting ELK4 and activating RhoA/ROCK pathway. miR-3188 may serve as a target for AS treatment.


Asunto(s)
Aterosclerosis/patología , MicroARNs/fisiología , Proteína Elk-4 del Dominio ets/metabolismo , Quinasas Asociadas a rho/metabolismo , Proteína de Unión al GTP rhoA/metabolismo , 1-Alquil-2-acetilglicerofosfocolina Esterasa/metabolismo , Apoptosis/fisiología , Aterosclerosis/genética , Estudios de Casos y Controles , Supervivencia Celular/fisiología , Citocinas/metabolismo , Regulación hacia Abajo , Femenino , Técnicas de Silenciamiento del Gen , Humanos , Lipoproteínas LDL/metabolismo , Macrófagos/patología , Masculino , MicroARNs/genética , Persona de Mediana Edad , Proteína Elk-4 del Dominio ets/genética
11.
Am J Hum Genet ; 92(4): 489-503, 2013 Apr 04.
Artículo en Inglés | MEDLINE | ID: mdl-23540573

RESUMEN

Analysis of 4,405 variants in 89,050 European subjects from 41 case-control studies identified three independent association signals for estrogen-receptor-positive tumors at 11q13. The strongest signal maps to a transcriptional enhancer element in which the G allele of the best candidate causative variant rs554219 increases risk of breast cancer, reduces both binding of ELK4 transcription factor and luciferase activity in reporter assays, and may be associated with low cyclin D1 protein levels in tumors. Another candidate variant, rs78540526, lies in the same enhancer element. Risk association signal 2, rs75915166, creates a GATA3 binding site within a silencer element. Chromatin conformation studies demonstrate that these enhancer and silencer elements interact with each other and with their likely target gene, CCND1.


Asunto(s)
Neoplasias de la Mama/genética , Cromosomas Humanos Par 11/genética , Ciclina D1/genética , Elementos de Facilitación Genéticos/genética , Polimorfismo de Nucleótido Simple/genética , Sitios de Unión , Estudios de Casos y Controles , Línea Celular Tumoral , Cromatina/química , Cromatina/genética , Inmunoprecipitación de Cromatina , Ciclina D1/metabolismo , Ensayo de Cambio de Movilidad Electroforética , Femenino , Factor de Transcripción GATA3/antagonistas & inhibidores , Factor de Transcripción GATA3/genética , Factor de Transcripción GATA3/metabolismo , Regulación Neoplásica de la Expresión Génica , Humanos , Luciferasas/metabolismo , Regiones Promotoras Genéticas/genética , ARN Mensajero/genética , ARN Interferente Pequeño/genética , Reacción en Cadena en Tiempo Real de la Polimerasa , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Elementos Silenciadores Transcripcionales/genética , Proteína Elk-4 del Dominio ets/antagonistas & inhibidores , Proteína Elk-4 del Dominio ets/genética , Proteína Elk-4 del Dominio ets/metabolismo
12.
Nature ; 460(7256): 705-10, 2009 Aug 06.
Artículo en Inglés | MEDLINE | ID: mdl-19578358

RESUMEN

MicroRNAs (miRNAs) are regulators of myriad cellular events, but evidence for a single miRNA that can efficiently differentiate multipotent stem cells into a specific lineage or regulate direct reprogramming of cells into an alternative cell fate has been elusive. Here we show that miR-145 and miR-143 are co-transcribed in multipotent murine cardiac progenitors before becoming localized to smooth muscle cells, including neural crest stem-cell-derived vascular smooth muscle cells. miR-145 and miR-143 were direct transcriptional targets of serum response factor, myocardin and Nkx2-5 (NK2 transcription factor related, locus 5) and were downregulated in injured or atherosclerotic vessels containing proliferating, less differentiated smooth muscle cells. miR-145 was necessary for myocardin-induced reprogramming of adult fibroblasts into smooth muscle cells and sufficient to induce differentiation of multipotent neural crest stem cells into vascular smooth muscle. Furthermore, miR-145 and miR-143 cooperatively targeted a network of transcription factors, including Klf4 (Kruppel-like factor 4), myocardin and Elk-1 (ELK1, member of ETS oncogene family), to promote differentiation and repress proliferation of smooth muscle cells. These findings demonstrate that miR-145 can direct the smooth muscle fate and that miR-145 and miR-143 function to regulate the quiescent versus proliferative phenotype of smooth muscle cells.


Asunto(s)
Linaje de la Célula , MicroARNs/metabolismo , Miocitos del Músculo Liso/citología , Miocitos del Músculo Liso/metabolismo , Animales , Diferenciación Celular , Proliferación Celular , Femenino , Regulación de la Expresión Génica , Proteína Homeótica Nkx-2.5 , Proteínas de Homeodominio/metabolismo , Factor 4 Similar a Kruppel , Masculino , Ratones , Ratones Transgénicos , MicroARNs/genética , Modelos Biológicos , Miocardio/metabolismo , Factores de Transcripción/metabolismo , Transcripción Genética , Enfermedades Vasculares/metabolismo , Proteína Elk-4 del Dominio ets/metabolismo
13.
BMC Genomics ; 15: 301, 2014 Apr 23.
Artículo en Inglés | MEDLINE | ID: mdl-24758171

RESUMEN

BACKGROUND: Serum response factor (SRF) is a widely expressed transcription factor involved in multiple regulatory programs. It is believed that SRF can toggle between disparate programs of gene expression through association with different cofactors. However, the direct evidence as to how these factors function on a genome-wide level is still lacking. RESULTS: In the present study, I explored the functions of SRF and its representative cofactors, megakaryoblastic leukemia 1/2 (MKL1/2) and ETS-domain protein 4 (ELK4), during fungal infection challenge in macrophages. The knockdown study, combined with gene expression array analysis, revealed that MKL1/2 regulated SRF-dependent genes were related to actin cytoskeleton organization, while ELK4 regulated SRF-dependent genes were related to external stimulus responses. Subsequent chromatin immunoprecipitation coupled with massively parallel sequencing (ChIP-seq) suggested that many of these regulations were mediated directly in cis. CONCLUSIONS: I conclude that SRF utilizes MKL1/2 to fulfill steady state cellular functions, including cytoskeletal organization, and utilizes ELK4 to facilitate acute responses to external infection. Together, these findings indicate that SRF, along with its two cofactors, are important players in both cellular homeostasis and stress responses in macrophages.


Asunto(s)
Regulación de la Expresión Génica , Macrófagos/metabolismo , Factor de Respuesta Sérica/genética , Transactivadores/metabolismo , Factores de Transcripción/metabolismo , Transcripción Genética , Proteína Elk-4 del Dominio ets/metabolismo , Animales , Sitios de Unión , Línea Celular , Regulación de la Expresión Génica/efectos de los fármacos , Técnicas de Silenciamiento del Gen , Macrófagos/efectos de los fármacos , Masculino , Ratones , Motivos de Nucleótidos , Posición Específica de Matrices de Puntuación , Regiones Promotoras Genéticas , Unión Proteica , Transporte de Proteínas , Reproducibilidad de los Resultados , Transactivadores/genética , Factores de Transcripción/genética , Zimosan/farmacología , Proteína Elk-4 del Dominio ets/genética
14.
Int J Dev Neurosci ; 84(6): 581-593, 2024 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-38923578

RESUMEN

Glioma, a formidable form of brain cancer, poses significant challenges in terms of treatment and prognosis. Circular RNA nucleoporin 98 (circNUP98) has emerged as a potential regulator in various cancers, yet its role in glioma remains unclear. Here, we elucidate the functional role of circNUP98 in glioma cell proliferation, invasion, and migration, shedding light on its therapeutic implications. Glioma cells were subjected to si-NUP98 transfection, followed by assessments of cell viability, proliferation, invasion, and migration. Subcellular localization of circNUP98 was determined, and its downstream targets were identified. We delineated the binding relationships between circNUP98 and microRNA (miR)-520f-3p, as well as between miR-520f-3p and ETS transcription factor ELK4 (ELK4). The expression levels of circNUP98/miR-520f-3p/ELK4 were quantified. Our findings demonstrated that circNUP98 was upregulated in glioma cells, and its inhibition significantly attenuated glioma cell proliferation, invasion, and migration. Mechanistically, circNUP98 functioned as a sponge for miR-520f-3p, thereby relieving the inhibitory effect of miR-520f-3p on ELK4. Moreover, inhibition of miR-520f-3p or overexpression of ELK4 partially rescued the suppressive effect of circNUP98 knockdown on glioma cell behaviors. In summary, our study unveils that circNUP98 promotes glioma cell progression via the miR-520f-3p/ELK4 axis, offering novel insights into the therapeutic targeting of circNUP98 in glioma treatment.


Asunto(s)
Neoplasias Encefálicas , Movimiento Celular , Proliferación Celular , Glioma , MicroARNs , ARN Circular , MicroARNs/metabolismo , MicroARNs/genética , Humanos , Glioma/metabolismo , Glioma/genética , Glioma/patología , ARN Circular/genética , ARN Circular/metabolismo , Movimiento Celular/genética , Movimiento Celular/fisiología , Línea Celular Tumoral , Neoplasias Encefálicas/genética , Neoplasias Encefálicas/metabolismo , Neoplasias Encefálicas/patología , Proteína Elk-4 del Dominio ets/genética , Proteína Elk-4 del Dominio ets/metabolismo , Invasividad Neoplásica/genética , Proteínas de Complejo Poro Nuclear/genética , Proteínas de Complejo Poro Nuclear/metabolismo , Regulación Neoplásica de la Expresión Génica , Animales
15.
Nat Commun ; 14(1): 6117, 2023 09 30.
Artículo en Inglés | MEDLINE | ID: mdl-37777542

RESUMEN

The proline synthesis is importantly involved in tumor growth under hypoxia, while the underlying mechanism remains to be further investigated. Here we show that pyrroline-5-carpoxylate reductase-1 (PYCR1), displaying a constant nuclear localization, is phosphorylated by nuclear IGF1R at Tyrosine 135 under hypoxia; this phosphorylation promotes the binding of PYCR1 to ELK4 and thus PYCR1 recruitment to ELK4-targeted genes promoter. Under hypoxia, ELK4-binding ability and enzymatic activity of PYCR1 are both required for ELK4-Sirt7-mediated transcriptional repression and cell growth maintenance, in which PYCR1-catalyzed NAD+ production stimulates the deacetylation activity of Sirt7 on H3K18ac that restrains genes transcription. Functionally, PYCR1 Tyr-135 phosphorylation exerts supportive effect on tumor growth under hypoxia, and the level of PYCR1 Tyr-135 phosphorylation is associated with malignancy of colorectal cancer (CRC). These data uncover the relationship between the compartmentally metabolic activity of PYCR1 and genes transcription regulation, and highlight the oncogenic role of PYCR1 during CRC development.


Asunto(s)
Neoplasias , Oxidorreductasas , Humanos , Línea Celular Tumoral , Proteína Elk-4 del Dominio ets/metabolismo , Hipoxia/genética , Neoplasias/genética , Receptor IGF Tipo 1/genética , delta-1-Pirrolina-5-Carboxilato Reductasa
16.
Adv Sci (Weinh) ; 10(32): e2303378, 2023 11.
Artículo en Inglés | MEDLINE | ID: mdl-37786278

RESUMEN

Although the MAPK/MEK/ERK pathway is prevalently activated in colorectal cancer (CRC), MEK/ERK inhibitors show limited efficiency in clinic. As a downstream target of MAPK, ELK4 is thought to work primarily by forming a complex with SRF. Whether ELK4 can serve as a potential therapeutic target is unclear and the transcriptional regulatory mechanism has not been systemically analyzed. Here, it is shown that ELK4 promotes CRC tumorigenesis. Integrated genomics- and proteomics-based approaches identified SP1 and SP3, instead of SRF, as cooperative functional partners of ELK4 at genome-wide level in CRC. Serum-induced phosphorylation of ELK4 by MAPKs facilitated its interaction with SP1/SP3. The pathological neoangiogenic factor LRG1 is identified as a direct target of the ELK4-SP1/SP3 complex. Furthermore, targeting the ELK4-SP1/SP3 complex by combination treatment with MEK/ERK inhibitor and the relatively specific SP1 inhibitor mithramycin A (MMA) elicited a synergistic antitumor effect on CRC. Clinically, ELK4 is a marker of poor prognosis in CRC. A 9-gene prognostic model based on the ELK4-SP1/3 complex-regulated gene set showed robust prognostic accuracy. The results demonstrate that ELK4 cooperates with SP1 and SP3 to transcriptionally regulate LRG1 to promote CRC tumorigenesis in an SRF-independent manner, identifying the ELK4-SP1/SP3 complex as a potential target for rational combination therapy.


Asunto(s)
Neoplasias Colorrectales , Regulación de la Expresión Génica , Humanos , Regiones Promotoras Genéticas , Neoplasias Colorrectales/genética , Carcinogénesis/genética , Quinasas de Proteína Quinasa Activadas por Mitógenos/genética , Proteína Elk-4 del Dominio ets/genética , Glicoproteínas
17.
Mol Biotechnol ; 65(10): 1608-1618, 2023 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-36715861

RESUMEN

Increasing evidences indicate the crucial role of circRNAs in tumorigenesis, but little is understood about their molecular mechanism in retinoblastoma (RB). This paper was designed for exploring the circ_0119412 function in cases with RB and the potential mechanism. RT-qPCR was utilized to ascertain circ_0119412 and miR-186-5p levels in RB tissues and cells, and western blotting was used to quantify ELK4 in RB cells. In addition, CCK-8 and scratch assays were carried out for evaluation of cell proliferation and migration, respectively. Apoptosis-related proteins levels (Bax and Bcl-2) were measure by western blotting. Tumor growth in vivo was detected utilizing xenograft tumor experiment. The targeting relationship between circ_0119412, miR-186-5p, and ELK4 was validated using a dual-luciferase reporter assay and an RNA immunoprecipitation (RIP) assay. In RB tissues and cells, Circ_0119412 and ELK4 expression were upregulated, while miR-186-5p expression was downregulated. In vitro assay revealed that downregulating circ_0119412 accelerated the cell apoptosis of RB cells and slowed down their migration and proliferation, and the in vivo assay indicated that circ_0119412 downregulation reduced the weight and volume of tumor in nude mice. In addition, miR-186-5p interference promoted the malignant behavior of RB cells, while ELK4 silencing showed an opposite trend. Mechanically, circ_0119412 can promote RB malignant phenotypes via miR-186-5p/ELK4 axis. Circ_0119412 was found to be upregulated in RB, and could accelerate the progression of RB via the miR-186-5p/ELK4 axis, indicating circ_0119412 may serve a promising clinical therapeutic target of RB.


Asunto(s)
MicroARNs , Neoplasias de la Retina , Retinoblastoma , Humanos , Animales , Ratones , Retinoblastoma/genética , Ratones Desnudos , Apoptosis/genética , Proliferación Celular/genética , Neoplasias de la Retina/genética , MicroARNs/genética , Línea Celular Tumoral , Movimiento Celular/genética , Proteína Elk-4 del Dominio ets
18.
Balkan Med J ; 40(6): 409-414, 2023 10 20.
Artículo en Inglés | MEDLINE | ID: mdl-37519006

RESUMEN

Background: Cervical cancer (CC) is a prevalent gynecological carcinoma, and patients infected with human papillomavirus (HPV) have a higher morbidity rate. Aims: To explore the effects of ETS-like transcription factor 4 (ELK4) in patients with HPV+ CC. Study design: In vitro cell lines and human-sample study. Methods: The ELK4 levels in human tissue (65 HPV+ CC tissue and 25 HPV− normal cervical tissue) and cell lines (human cervical epithelial immortalized cell line H8 and CC cell lines HeLa [HPV18], CaSki [HPV16], and SiHa [HPV−]) were quantified using qRT-PCR and western blot assay. ELK4 knockdown transfection was effective and confirmed by western blotting. The MTT and EDU assays were used to evaluate cell viability and proliferation, respectively. Flow cytometry was used to detect the CC cell cycle stage. Stem cell markers, such as cluster of differentiation 133 (CD133), CD44, and aldehyde dehydrogenase 1, and the cervicospheres formed were measured. ChIP-qPCR and luciferase activity experiments were used to assess the bond between ELK4 and F-box protein 22 (FBXO22). Results: ELK4 was highly expressed in the HPV+ CC tissue. CC cells with ELK4 knockdown had lower viability and proliferation than the control cells. ELK4 knockdown blocked the progression of the cell cycle from G1 to S phase. ELK4 knockdown suppressed the stem cell-like characteristics of the HPV+ CC cells. ELK4 bonded with the FBXO22 promoter, inhibiting the levels of phosphatase and tensin homolog (PTEN). Conclusion: ELK4 facilitated cell cycle progression and stem cell-like characteristics by regulating the FBXO22/PTEN axis. Thus, ELK4 could be a potential therapeutic target to arrest the progress of HPV-associated CC.


Asunto(s)
Proteínas F-Box , Infecciones por Papillomavirus , Neoplasias del Cuello Uterino , Femenino , Humanos , Neoplasias del Cuello Uterino/metabolismo , Virus del Papiloma Humano , Línea Celular Tumoral , Proliferación Celular , Células Madre/metabolismo , Células Madre/patología , Ciclo Celular , Fosfohidrolasa PTEN/metabolismo , Fosfohidrolasa PTEN/farmacología , Proteína Elk-4 del Dominio ets/metabolismo , Receptores Citoplasmáticos y Nucleares/metabolismo , Receptores Citoplasmáticos y Nucleares/farmacología , Proteínas F-Box/farmacología
19.
RNA ; 16(6): 1156-66, 2010 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-20406994

RESUMEN

In humans, more than 30,000 chimeric transcripts originating from 23,686 genes have been identified. The mechanisms and association of chimeric transcripts arising from chromosomal rearrangements with cancer are well established, but much remains unknown regarding the biogenesis and importance of other chimeric transcripts that arise from nongenomic alterations. Recently, a SLC45A3-ELK4 chimera has been shown to be androgen-regulated, and is overexpressed in metastatic or high-grade prostate tumors relative to local prostate cancers. Here, we characterize the expression of a KLK4 cis sense-antisense chimeric transcript, and show other examples in prostate cancer. Using non-protein-coding microarray analyses, we initially identified an androgen-regulated antisense transcript within the 3' untranslated region of the KLK4 gene in LNCaP cells. The KLK4 cis-NAT was validated by strand-specific linker-mediated RT-PCR and Northern blotting. Characterization of the KLK4 cis-NAT by 5' and 3' rapid amplification of cDNA ends (RACE) revealed that this transcript forms multiple fusions with the KLK4 sense transcript. Lack of KLK4 antisense promoter activity using reporter assays suggests that these transcripts are unlikely to arise from a trans-splicing mechanism. 5' RACE and analyses of deep sequencing data from LNCaP cells treated +/-androgens revealed six high-confidence sense-antisense chimeras of which three were supported by the cDNA databases. In this study, we have shown complex gene expression at the KLK4 locus that might be a hallmark of cis sense-antisense chimeric transcription.


Asunto(s)
ADN sin Sentido/genética , Variación Genética , Calicreínas/genética , Neoplasias de la Próstata/genética , Transcripción Genética , Antígenos de Neoplasias/genética , Quimera/genética , Mapeo Cromosómico , ADN de Neoplasias/genética , Exones , Reordenamiento Génico , Estudio de Asociación del Genoma Completo , Humanos , Masculino , Proteínas de Transporte de Membrana/genética , Neoplasias de la Próstata/patología , Proteína Elk-4 del Dominio ets/genética
20.
J Immunol ; 185(2): 1082-92, 2010 Jul 15.
Artículo en Inglés | MEDLINE | ID: mdl-20554967

RESUMEN

The ternary complex factors (TCFs; SAP-1, Elk-1, and Net) are serum response factor cofactors that share many functional properties and are coexpressed in many tissues. SAP-1, the predominant thymus TCF, is required for thymocyte positive selection. In this study, we assessed whether the different TCFs are functionally equivalent. Elk-1 deletion, but not the hypomorphic Net(delta) mutation, exacerbated the SAP-1 positive selection phenotype, but triply deficient thymocytes were no more defective than SAP-1(-/-) Elk-1(-/-) cells. Inactivation of the other TCFs did not affect SAP-1-independent processes, including beta-selection, regulatory T cell selection, and negative selection, although reduced marginal zone B cells were observed in SAP-1(-/-) Elk-1(-/-) animals. Ectopic expression of Elk-1, but not Net, rescued positive selection of SAP-1(-/-) thymocytes; thus, SAP-1 and Elk-1 are functionally equivalent in this system, and the SAP-1 null selection phenotype reflects only its high expression in the thymus. Array analysis of TCR-stimulated double-positive cells identified SAP-1-dependent inducible genes whose transcription was further impaired in SAP-1(-/-) Elk-1(-/-) cells; thus, these genes, which include Egr-1 and Egr-2, represent candidate mediators of positive selection. Chromatin immunoprecipitation revealed subtly different promoter targeting between the different TCFs. Ectopic expression of Egr-1 restored positive selection in SAP-1 null thymocytes, establishing it (and possibly other Egr family members) as the major effector for ERK-SAP-1 signaling in thymocyte positive selection.


Asunto(s)
Proteínas Proto-Oncogénicas c-ets/inmunología , Timo/inmunología , Proteína Elk-1 con Dominio ets/inmunología , Proteína Elk-4 del Dominio ets/inmunología , Animales , Diferenciación Celular/genética , Diferenciación Celular/inmunología , Células Cultivadas , Inmunoprecipitación de Cromatina , Femenino , Citometría de Flujo , Perfilación de la Expresión Génica , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Análisis de Secuencia por Matrices de Oligonucleótidos , Proteínas Proto-Oncogénicas c-ets/genética , Proteínas Proto-Oncogénicas c-ets/metabolismo , Retroviridae/genética , Timo/citología , Timo/metabolismo , Transducción Genética , Proteína Elk-1 con Dominio ets/genética , Proteína Elk-1 con Dominio ets/metabolismo , Proteína Elk-4 del Dominio ets/genética , Proteína Elk-4 del Dominio ets/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA