RESUMEN
The epigenetic processes that regulate antibody-secreting plasma cells are not well understood. Here, analysis of plasma cell differentiation revealed DNA hypomethylation of 10% of CpG loci that were overrepresented at enhancers. Inhibition of DNA methylation enhanced plasma cell commitment in a cell-division-dependent manner. Analysis of B cells differentiating in vivo stratified by cell division revealed a fivefold increase in mRNA transcription coupled to DNA hypomethylation. Demethylation occurred first at binding motifs for the transcription factors NF-κB and AP-1 and later at those for the transcription factors IRF and Oct-2 and was coincident with activation and differentiation gene-expression programs in a cell-division-dependent manner. These data provide mechanistic insight into cell-division-coupled transcriptional and epigenetic reprogramming and suggest that DNA hypomethylation reflects the cis-regulatory history of plasma cell differentiation.
Asunto(s)
Linfocitos B/fisiología , Metilación de ADN , FN-kappa B/metabolismo , Células Plasmáticas/fisiología , Factor de Transcripción AP-1/metabolismo , Animales , Sitios de Unión/genética , Diferenciación Celular/genética , División Celular/genética , Células Cultivadas , Islas de CpG/genética , Epigénesis Genética , Femenino , Regulación de la Expresión Génica , Inmunidad Humoral/genética , Factores Reguladores del Interferón/genética , Factores Reguladores del Interferón/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , FN-kappa B/genética , Factor 2 de Transcripción de Unión a Octámeros/genética , Factor 2 de Transcripción de Unión a Octámeros/metabolismo , Factor de Transcripción AP-1/genéticaRESUMEN
BOB.1/OBF.1 is a lymphocyte-specific transcriptional co-activator of octamer-dependent transcription. It regulates the expression of genes important for lymphocyte physiology together with the Oct-1 and Oct-2 transcription factors. So far, BOB.1/OBF.1 has been studied in conventional knockout mice, whereby a function of BOB.1/OBF.1 in B but also in T cells was described. The main characteristic of BOB.1/OBF.1-deficient mice is the complete absence of germinal centers. However, it is entirely unsolved at which stage of B-cell development BOB.1/OBF.1 expression is essential for germinal center formation. Still, it is not known whether defects observed late in B-cell development of BOB.1/OBF.1-deficient mice are merely a consequence of defective early B-cell development. To answer the question, whether BOB.1/OBF.1 expression is required before or during the process of germinal center formation, we established a mouse system, which allows the conditional deletion of BOB.1/OBF.1 at different stages of B-cell development. Our data reveal a requirement for BOB.1/OBF.1 during both early antigen-independent and late antigen-dependent B-cell development, and further a requirement for efficient germinal center reaction during complete B-cell ontogeny. By specifically deleting BOB.1/OBF.1 in germinal center B cells, we provide evidence that the failure to form germinal centers is a germinal center B-cell intrinsic defect and not exclusively a consequence of defective early B-cell maturation.
Asunto(s)
Linfocitos B , Centro Germinal , Transactivadores/metabolismo , Animales , Linfocitos B/metabolismo , Diferenciación Celular , Activación de Linfocitos , Ratones , Factor 2 de Transcripción de Unión a Octámeros/metabolismo , Factores de Transcripción/metabolismoRESUMEN
The mammalian retina contains a variety of functionally distinct cell types that are generated by progenitor cells in a specific chronological order. A new paper in Development probes the role of the POU-homeodomain factors Pou2f1 and Pou2f2 in the timely generation of cone photoreceptors in mice. We caught up with first author and PhD student Awais Javed and his supervisor Michel Cayouette (Director of the Cellular Neurobiology Research Unit at the Montreal Clinical Research Institute, Professor at the Université de Montréal and Adjunct Professor at McGill University) to hear more about their work.
Asunto(s)
Retina/metabolismo , Células Fotorreceptoras Retinianas Conos/metabolismo , Células Madre/metabolismo , Animales , Ratones , Factor 1 de Transcripción de Unión a Octámeros/metabolismo , Factor 2 de Transcripción de Unión a Octámeros/metabolismoRESUMEN
Multipotent retinal progenitor cells (RPCs) generate various cell types in a precise chronological order, but how exactly cone photoreceptor production is restricted to early stages remains unclear. Here, we show that the POU-homeodomain factors Pou2f1/Pou2f2, the homologs of Drosophila temporal identity factors nub/pdm2, regulate the timely production of cones in mice. Forcing sustained expression of Pou2f1 or Pou2f2 in RPCs expands the period of cone production, whereas misexpression in late-stage RPCs triggers ectopic cone production at the expense of late-born fates. Mechanistically, we report that Pou2f1 induces Pou2f2 expression, which binds to a POU motif in the promoter of the rod-inducing factor Nrl to repress its expression. Conversely, conditional inactivation of Pou2f2 in RPCs increases Nrl expression and reduces cone production. Finally, we provide evidence that Pou2f1 is part of a cross-regulatory cascade with the other temporal identity factors Ikzf1 and Casz1. These results uncover Pou2f1/2 as regulators of the temporal window for cone genesis and, given their widespread expression in the nervous system, raise the possibility of a general role in temporal patterning.This article has an associated 'The people behind the papers' interview.
Asunto(s)
Proteínas del Ojo/metabolismo , Factor 1 de Transcripción de Unión a Octámeros/metabolismo , Factor 2 de Transcripción de Unión a Octámeros/metabolismo , Retina/metabolismo , Células Fotorreceptoras Retinianas Conos/metabolismo , Animales , Drosophila/metabolismo , Femenino , Ratones , Ratones Endogámicos C57BL , Regiones Promotoras Genéticas/genética , Células Fotorreceptoras Retinianas Bastones/metabolismo , Células Madre/metabolismoRESUMEN
OBF1 is a specific coactivator of the POU family transcription factors OCT1 and OCT2. OBF1 and OCT2 are B cell-specific and indispensable for germinal center (GC) formation, but their mechanism of action is unclear. Here, we show by chromatin immunoprecipitation-sequencing that OBF1 extensively colocalizes with OCT1 and OCT2. We found that these factors also often colocalize with transcription factors of the ETS family. Furthermore, we showed that OBF1, OCT2, and OCT1 bind widely to the promoters or enhancers of genes involved in GC formation in mouse and human GC B cells. Short hairpin RNA knockdown experiments demonstrated that OCT1, OCT2, and OBF1 regulate each other and are essential for proliferation of GC-derived lymphoma cell lines. OBF1 downregulation disrupts the GC transcriptional program: genes involved in GC maintenance, such as BCL6, are downregulated, whereas genes related to exit from the GC program, such as IRF4, are upregulated. Ectopic expression of BCL6 does not restore the proliferation of GC-derived lymphoma cells depleted of OBF1 unless IRF4 is also depleted, indicating that OBF1 controls an essential regulatory node in GC differentiation.
Asunto(s)
Centro Germinal/metabolismo , Factor 1 de Transcripción de Unión a Octámeros/fisiología , Factor 2 de Transcripción de Unión a Octámeros/uso terapéutico , Transactivadores/uso terapéutico , Transcripción Genética/genética , Animales , Linfocitos B/efectos de los fármacos , Linfocitos B/metabolismo , Línea Celular Tumoral , Inmunoprecipitación de Cromatina , Ontología de Genes , Células HEK293 , Humanos , Lipopolisacáridos/farmacología , Linfoma no Hodgkin/genética , Linfoma no Hodgkin/patología , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Factor 1 de Transcripción de Unión a Octámeros/deficiencia , Factor 1 de Transcripción de Unión a Octámeros/genética , Factor 2 de Transcripción de Unión a Octámeros/deficiencia , Factor 2 de Transcripción de Unión a Octámeros/genética , Proteína Proto-Oncogénica c-ets-1/análisis , Interferencia de ARN , ARN Interferente Pequeño/genética , Proteínas Recombinantes/metabolismo , Transactivadores/deficiencia , Transactivadores/genéticaRESUMEN
Cerebral ischemia reperfusion injury (CIRI) is the commonest cause of brain dysfunction. Up-regulation of POU domain class 2 transcription factor 2 (POU2F2) has been reported in patients with cerebral ischemia, while the role of POU2F2 in CIRI remains elusive. Middle cerebral artery occlusion/reperfusion (MCAO/R) in mice and oxygen and glucose deprivation/reperfusion (OGD/R) in mouse primary cortical neurons were used as models of CIRI injury in vivo and in vitro. Lentivirus-mediated POU2F2 knockdown further impaired CIRI induced by MCAO/R in mice, which was accompanied by increased-neurological deficits, cerebral infarct volume and neuronal loss. Our evidence suggested that POU2F2 deficiency deteriorated oxidative stress and ferroptosis according to the phenomenon such as the abatement of SOD, GSH, glutathione peroxidase 4 (GPX4) activity and accumulation of ROS, lipid ROS, 4-hydroxynonenal (4-HNE) and MDA. In vivo, primary cortical neurons with POU2F2 knockdown also showed worse neuronal damage, oxidative stress and ferroptosis. Sestrin2 (Sesn2) was reported as a neuroprotection gene and involved in ferroptosis mechanism. Up-regulation of Sesn2 was observed in the ischemic penumbra and OGD/R-induced neuronal cells. Further, we proved that POU2F2, as a transcription factor, could bind to Sesn2 promoter and positively regulate its expression. Sesn2 overexpression relieved oxidative stress and ferroptosis induced by POU2F2 knockdown in OGD/R-treated neurons. This research demonstrated that CIRI induced a compensatory increase of POU2F2 and Sesn2. Down-regulated POU2F2 exacerbated CIRI through the acceleration of oxidative stress and ferroptosis possibly by decreasing Sesn2 expression, which offers new sights into therapeutic mechanisms for CIRI.
Asunto(s)
Isquemia Encefálica , Ferroptosis , Factor 2 de Transcripción de Unión a Octámeros , Daño por Reperfusión , Sestrinas , Animales , Ratones , Isquemia Encefálica/metabolismo , Infarto de la Arteria Cerebral Media/metabolismo , Especies Reactivas de Oxígeno/metabolismo , Daño por Reperfusión/metabolismo , Factores de Transcripción , Factor 2 de Transcripción de Unión a Octámeros/metabolismo , Sestrinas/metabolismoRESUMEN
Bladder cancer (BC) is one of the most common malignant tumors of the urinary system worldwide. To date, immune checkpoint inhibitors (including PD-1/PD-L1) have been applied to treat patients with bladder cancer in the clinic and achieved the promising outcome. Further improvement of the anticancer efficiency of these immune therapies is crucial for bladder cancer. Our previous RNA-seq data on CBX7 (GSE185630) suggested that CBX7 might repress PD-L1 expression and PD-1 checkpoint pathway in cancer. In this study, we revealed that CBX7 downregulated the expression of POU2F2 that indirectly repressed the PD-L1 in BC cells. Depletion of CBX7 resulted in resistance to PD-1 blockade in bladder cancer. Collectively, our results suggested that the CBX7/POU2F2/PD-L1 axis plays an important role in determining the antitumor effect of PD-1 blockade in bladder cancer.
Asunto(s)
Antígeno B7-H1 , Factor 2 de Transcripción de Unión a Octámeros , Complejo Represivo Polycomb 1 , Neoplasias de la Vejiga Urinaria , Antígeno B7-H1/antagonistas & inhibidores , Antígeno B7-H1/inmunología , Antígeno B7-H1/metabolismo , Humanos , Inhibidores de Puntos de Control Inmunológico/farmacología , Factor 2 de Transcripción de Unión a Octámeros/inmunología , Complejo Represivo Polycomb 1/inmunología , Receptor de Muerte Celular Programada 1/inmunología , Neoplasias de la Vejiga Urinaria/inmunología , Neoplasias de la Vejiga Urinaria/patologíaRESUMEN
BACKGROUND: Emerging research has demonstrated that long non-coding RNAs (lncRNAs) attach great importance to the progression of cervical cancer (CC). LncRNA ARAP1-AS1 was involved in the development of several cancers; however, its role in CC is far from being elucidated. METHODS: Real-time PCR (RT-PCR) was employed to detect ARAP1-AS1 and miR-149-3p expression in CC samples. CC cell lines (HeLa and C33A cells) were regarded as the cell models. The biological effect of ARAP1-AS1 on cancer cells was measured using CCK-8 assay, colony formation assay, flow cytometry, Transwell assay and wound healing assay in vitro, and subcutaneous xenotransplanted tumor model and tail vein injection model in vivo. Furthermore, interactions between ARAP1-AS1 and miR-149-3p, miR-149-3p and POU class 2 homeobox 2 (POU2F2) were determined by bioinformatics analysis, qRT-PCR, Western blot, luciferase reporter and RNA immunoprecipitation assay, respectively. RESULTS: The expression of ARAP1-AS1 was enhanced in CC samples, while miR-149-3p was markedly suppressed. Additionally, ARAP1-AS1 overexpression enhanced the viability, migration, and invasion of CC cells. ARAP1-AS1 downregulated miR-149-3p via sponging it. ARAP1-AS1 and miR-149-3p exhibited a negative correlation in CC samples. On the other hand, ARAP1-AS1 enhanced the expression of POU2F2, which was validated as a target gene of miR-149-3p. CONCLUSION: ARAP1-AS1 was abnormally upregulated in CC tissues and indirectly modulated the POU2F2 expression via reducing miR-149-3p expression. Our study identified a novel axis, ARAP1-AS1/miR-149-3p/POU2F2, in CC tumorigenesis.
Asunto(s)
Proteínas Portadoras/genética , Proteínas Activadoras de GTPasa/genética , Regulación Neoplásica de la Expresión Génica , MicroARNs/genética , Factor 2 de Transcripción de Unión a Octámeros/genética , Neoplasias del Cuello Uterino/genética , Animales , Línea Celular Tumoral , Proliferación Celular , Femenino , Células HeLa , Humanos , Ratones , Ratones Endogámicos BALB C , Ratones Desnudos , Transducción de Señal , Trasplante HeterólogoRESUMEN
BACKGROUND: To detect and investigate the expression of POU domain class 2 transcription factor 2 (POU2F2) in human lung cancer tissues, its role in lung cancer progression, and the potential mechanisms. METHODS: Immunohistochemical (IHC) assays were conducted to assess the expression of POU2F2 in human lung cancer tissues. Immunoblot assays were performed to assess the expression levels of POU2F2 in human lung cancer tissues and cell lines. CCK-8, colony formation, and transwell-migration/invasion assays were conducted to detect the effects of POU2F2 and AGO1 on the proliferaion and motility of A549 and H1299 cells in vitro. CHIP and luciferase assays were performed for the mechanism study. A tumor xenotransplantation model was used to detect the effects of POU2F2 on tumor growth in vivo. RESULTS: We found POU2F2 was highly expressed in human lung cancer tissues and cell lines, and associated with the lung cancer patients' prognosis and clinical features. POU2F2 promoted the proliferation, and motility of lung cancer cells via targeting AGO1 in vitro. Additionally, POU2F2 promoted tumor growth of lung cancer cells via AGO1 in vivo. CONCLUSION: We found POU2F2 was highly expressed in lung cancer cells and confirmed the involvement of POU2F2 in lung cancer progression, and thought POU2F2 could act as a potential therapeutic target for lung cancer.
Asunto(s)
Proteínas Argonautas/metabolismo , Factores Eucarióticos de Iniciación/metabolismo , Neoplasias Pulmonares/genética , Neoplasias Pulmonares/metabolismo , Factor 2 de Transcripción de Unión a Octámeros/metabolismo , Células A549 , Proteínas Argonautas/genética , Línea Celular Tumoral , Movimiento Celular , Proliferación Celular/genética , Factores Eucarióticos de Iniciación/genética , Regulación Neoplásica de la Expresión Génica , Humanos , Neoplasias Pulmonares/patología , Factor 2 de Transcripción de Unión a Octámeros/genética , PronósticoRESUMEN
To elucidate the role of POU2F2 (POU class 2 homeobox 2) in fracture healing, 30 rats with femoral fracture were randomly grouped into three groups: FF group, LV-POU2F2 group and LV-scramble group. Rats were injected with PBS, lentivirus expressing POU2F2 or scramble lentivirus once a week for 4 weeks. Results showed that overexpressing of POU2F2 promoted fracture healing and callus growth. Besides, overexpressing of POU2F2 promoted protein and mRNA expression of Col10a1, Runx2, Osterix, and Osteocalcin. High Mobility Group AT-hook 1 (HMGA1) is a non-histone protein participating in chromatin remodeling of cells. Western blotting manifested HMGA1/Wnt/ß-catenin pathway was activated in POU2F2 group. Moreover, in-vitro study of hMSCs cells supported the above data. In conclusion, POU2F2 promotes fracture healing via activating the HMGA1/Wnt/ß-catenin signaling pathway.
Asunto(s)
Curación de Fractura , Proteína HMGA1a/metabolismo , Factor 2 de Transcripción de Unión a Octámeros/metabolismo , Transducción de Señal , Vía de Señalización Wnt , beta Catenina/metabolismo , Animales , Biomarcadores/metabolismo , Humanos , RatasRESUMEN
The assembly of individual protein subunits into large-scale symmetrical structures is widespread in nature and confers new biological properties. Engineered protein assemblies have potential applications in nanotechnology and medicine; however, a major challenge in engineering assemblies de novo has been to design interactions between the protein subunits so that they specifically assemble into the desired structure. Here we demonstrate a simple, generalizable approach to assemble proteins into cage-like structures that uses short de novo designed coiled-coil domains to mediate assembly. We assembled eight copies of a C3-symmetric trimeric esterase into a well-defined octahedral protein cage by appending a C4-symmetric coiled-coil domain to the protein through a short, flexible linker sequence, with the approximate length of the linker sequence determined by computational modeling. The structure of the cage was verified using a combination of analytical ultracentrifugation, native electrospray mass spectrometry, and negative stain and cryoelectron microscopy. For the protein cage to assemble correctly, it was necessary to optimize the length of the linker sequence. This observation suggests that flexibility between the two protein domains is important to allow the protein subunits sufficient freedom to assemble into the geometry specified by the combination of C4 and C3 symmetry elements. Because this approach is inherently modular and places minimal requirements on the structural features of the protein building blocks, it could be extended to assemble a wide variety of proteins into structures with different symmetries.
Asunto(s)
Pliegue de Proteína , Multimerización de Proteína , Estructura Secundaria de Proteína , Proteínas/química , Secuencia de Aminoácidos , Microscopía por Crioelectrón , Espectrometría de Masas/métodos , Microscopía Electrónica de Transmisión , Modelos Moleculares , Factor 2 de Transcripción de Unión a Octámeros/química , Factor 2 de Transcripción de Unión a Octámeros/ultraestructura , Factor 3 de Transcripción de Unión a Octámeros/química , Factor 3 de Transcripción de Unión a Octámeros/ultraestructura , Proteínas/ultraestructuraRESUMEN
Circular ribonucleic acids are non-coding ribonucleic acids that can be identified from genome sequencing studies. Although they can be readily detected, their regulation and functional role in human diseases such as cancer are unknown. Using a systematic approach, we analyzed ribonucleic acid-sequencing data from a well-characterized cohort of intrahepatic cholangiocarcinoma to identify genetic pathways related to circular ribonucleic acids. Although the expression of most circular ribonucleic acids was similar in both the cancer and non-cancer tissues, expression of circ2174 was significantly increased in cancer tissues. Network analysis of co-related genes identified several pathways associated with circ2174, and common regulatory mediators between genes in these pathways and circ2174. Among these, alterations in several genes involved in interleukin-16 signaling responses such Lck, interleukin-16, and macrophage inflammatory protein-1-beta were the most prominent. Octamer transcription factor (Oct)-2 was identified as a signal transducer that was common to both circ2174 and interleukin-16. Circ2174 has sequence complementarity to miR149 which can target Oct-2. These data suggest a mechanism whereby circ2174 can act as a sponge to regulate the expression of miR149, and thereby modulate Oct-2 and interleukin-16 signaling pathways in cholangiocarcinoma.
Asunto(s)
Colangiocarcinoma/genética , MicroARNs/genética , Factor 2 de Transcripción de Unión a Octámeros/genética , ARN/genética , Colangiocarcinoma/patología , Regulación Neoplásica de la Expresión Génica/genética , Redes Reguladoras de Genes , Humanos , Interleucina-16/genética , ARN Circular , Transducción de SeñalRESUMEN
The expression of Oct-1 and -2 and their binding to the octamer motif in the mammary gland are developmentally and hormonally regulated, consistent with the expression of milk proteins. Both of these transcription factors constitutively bind to the proximal promoter of the milk protein gene ß-casein and might be involved in the inhibition or activation of promoter activity via interactions with other transcription factors or cofactors at different developmental stages. In particular, the lactogenic hormone prolactin and glucocorticoids induce Oct-1 and Oct-2 binding and interaction with both the signal transducer and activator of transcription 5 (STAT5) and the glucocorticoid receptor on the ß-casein promoter to activate ß-casein expression. In addition, increasing evidence has shown the involvement of another Oct factor, Oct-3/4, in mammary tumorigenesis, making Oct-3/4 an emerging prognostic marker of breast cancer and a molecular target for the gene-directed therapeutic intervention, prevention and treatment of breast cancer. This article is part of a Special Issue entitled: The Oct Transcription Factor Family, edited by Dr. Dean Tantin.
Asunto(s)
Neoplasias de la Mama/genética , Carcinogénesis/genética , Regulación Neoplásica de la Expresión Génica , Factor 1 de Transcripción de Unión a Octámeros/genética , Factor 2 de Transcripción de Unión a Octámeros/genética , Secuencia de Aminoácidos , Animales , Neoplasias de la Mama/metabolismo , Neoplasias de la Mama/patología , Carcinogénesis/metabolismo , Carcinogénesis/patología , Femenino , Humanos , Lactancia/genética , Glándulas Mamarias Humanas/metabolismo , Glándulas Mamarias Humanas/patología , Datos de Secuencia Molecular , Factor 1 de Transcripción de Unión a Octámeros/metabolismo , Factor 2 de Transcripción de Unión a Octámeros/metabolismo , Isoformas de Proteínas/genética , Isoformas de Proteínas/metabolismo , Transducción de Señal , Activación TranscripcionalRESUMEN
UNLABELLED: Epstein-Barr virus (EBV) establishes latent infections as multicopy episomes with complex patterns of viral gene transcription and chromatin structure. The EBV origin of plasmid replication (OriP) has been implicated as a critical control element for viral transcription, as well as viral DNA replication and episome maintenance. Here, we examine cellular factors that bind OriP and regulate histone modification, transcription regulation, and episome maintenance. We found that OriP is enriched for histone H3 lysine 4 (H3K4) methylation in multiple cell types and latency types. Host cell factor 1 (HCF1), a component of the mixed-lineage leukemia (MLL) histone methyltransferase complex, and transcription factor OCT2 (octamer-binding transcription factor 2) bound cooperatively with EBNA1 (Epstein-Barr virus nuclear antigen 1) at OriP. Depletion of OCT2 or HCF1 deregulated latency transcription and histone modifications at OriP, as well as the OriP-regulated latency type-dependent C promoter (Cp) and Q promoter (Qp). HCF1 depletion led to a loss of histone H3K4me3 (trimethylation of histone H3 at lysine 4) and H3 acetylation at Cp in type III latency and Qp in type I latency, as well as an increase in heterochromatic H3K9me3 at these sites. HCF1 depletion resulted in the loss of EBV episomes from Burkitt's lymphoma cells with type I latency and reactivation from lymphoblastoid cells (LCLs) with type III latency. These findings indicate that HCF1 and OCT2 function at OriP to regulate viral transcription, histone modifications, and episome maintenance. As HCF1 is best known for its function in herpes simplex virus 1 (HSV-1) immediate early gene transcription, our findings suggest that EBV latency transcription shares unexpected features with HSV gene regulation. IMPORTANCE: EBV latency is associated with several human cancers. Viral latent cycle gene expression is regulated by the epigenetic control of the OriP enhancer region. Here, we show that cellular factors OCT2 and HCF1 bind OriP in association with EBNA1 to maintain elevated histone H3K4me3 and transcriptional enhancer function. HCF1 is known as a transcriptional coactivator of herpes simplex virus (HSV) immediate early (IE) transcription, suggesting that OriP enhancer shares aspects of HSV IE transcription control.
Asunto(s)
Antígenos Nucleares del Virus de Epstein-Barr/metabolismo , Herpesvirus Humano 4/genética , Factor C1 de la Célula Huésped/metabolismo , Factor 2 de Transcripción de Unión a Octámeros/metabolismo , Plásmidos , Latencia del Virus/genética , Antígenos Nucleares del Virus de Epstein-Barr/genética , Regulación de la Expresión Génica , Herpesvirus Humano 4/fisiología , Histonas/genética , Histonas/metabolismo , Factor C1 de la Célula Huésped/deficiencia , Factor C1 de la Célula Huésped/genética , Proteína de Factor 1 del Huésped/genética , Proteína de Factor 1 del Huésped/metabolismo , Humanos , Metilación , Factor 2 de Transcripción de Unión a Octámeros/genética , Origen de RéplicaRESUMEN
Oct-1 (POU2f1) and Oct-2 (POU2f2) are members of the POU family of transcription factors. They recognize the same DNA sequence but fulfil distinct functions: Oct-1 is ubiquitous and regulates a variety of genes while Oct-2 is restricted to B-cells and neurones. Here we examine the interplay and regulatory mechanisms of these factors to control the inducible nitric oxide synthase (iNOS, NOS2). Using two breast cancer cell lines as a comparative model, we found that MCF-7 express iNOS upon cytokine stimulation while MDA-MB-231 do not. Oct-1 is present in both cell lines but MDA-MB-231 also express high levels of Oct-2. Manipulation of Oct-2 expression in these cell lines demonstrates that it is directly responsible for the repression of iNOS in MDA-MB-231. In MCF-7 cells Oct-1 binds the iNOS promoter, recruits RNA PolII and triggers initiation of transcription. In MDA-MB-231 cells, both Oct-1 and Oct-2 bind the iNOS promoter, forming a higher-order complex which fails to recruit RNA PolII, and as a consequence iNOS transcription does not proceed. Unravelling the mechanisms of transcription factor activity is paramount to the understanding of gene expression patterns that determine cell behaviour.
Asunto(s)
Regulación Neoplásica de la Expresión Génica , Óxido Nítrico Sintasa de Tipo II/genética , Factor 1 de Transcripción de Unión a Octámeros/metabolismo , Factor 2 de Transcripción de Unión a Octámeros/metabolismo , Regiones Promotoras Genéticas , ARN Polimerasa III/metabolismo , Proteínas Represoras/metabolismo , Neoplasias de la Mama/genética , Neoplasias de la Mama/metabolismo , Línea Celular Tumoral , Femenino , Humanos , Células MCF-7 , Transcripción GenéticaRESUMEN
PURPOSE: We tested whether mitochondrial electron transport chain electron carrier coenzyme Q10 (CoQ10) increases ATP during bovine IVM and increases %M2 oocytes, mitochondrial polarization/mass, and Oct4, and decreases pAMPK and oocyte death. METHODS: Bovine oocytes were aspirated from ovaries and cultured in IVM media for 24 h with 0, 20, 40, or 60 µM CoQ10. Oocytes were assayed for ATP by luciferase-based luminescence. Oocyte micrographs were quantitated for Oct4, pAMPK (i.e., activity), polarization by JC1 staining, and mitochondrial mass by MitoTracker Green staining. RESULTS: CoQ10 at 40 µM was optimal. Oocytes at 40 µM enabled 1.9-fold more ATP than 0 µM CoQ10. There was 4.3-fold less oocyte death, 1.7-fold more mitochondrial charge polarization, and 3.1-fold more mitochondrial mass at 40 µM than at 0 µM CoQ10. Increased ATP was associated with 2.2-fold lower AMPK thr172P activation and 2.1-fold higher nuclear Oct4 stemness/potency protein at 40 µM than at 0 µM CoQ10. CoQ10 is hydrophobic, and at all doses, 50% was lost from media into oil by ~ 12 h. Replenishing CoQ10 at 12 h did not significantly diminish dead oocytes. CONCLUSIONS: The data suggest that CoQ10 improves mitochondrial function in IVM where unwanted stress, higher AMPK activity, and Oct4 potency loss are induced.
Asunto(s)
Adenosina Trifosfato/metabolismo , Apoptosis/efectos de los fármacos , Técnicas de Maduración In Vitro de los Oocitos/métodos , Mitocondrias/metabolismo , Factores de Transcripción de Octámeros/metabolismo , Oocitos/patología , Proteínas Quinasas/metabolismo , Ubiquinona/análogos & derivados , Quinasas de la Proteína-Quinasa Activada por el AMP , Animales , Bovinos , Células Cultivadas , Metabolismo Energético/efectos de los fármacos , Femenino , Mitocondrias/efectos de los fármacos , Factor 2 de Transcripción de Unión a Octámeros , Oocitos/efectos de los fármacos , Oocitos/metabolismo , Ubiquinona/farmacología , Vitaminas/farmacologíaRESUMEN
Neuronal differentiation is a complex process that involves a plethora of regulatory steps. To identify transcription factors that influence neuronal differentiation we developed a high throughput screen using embryonic stem (ES) cells. Seven-hundred human transcription factor clones were stably introduced into mouse ES (mES) cells and screened for their ability to induce neuronal differentiation of mES cells. Twenty-four factors that are capable of inducing neuronal differentiation were identified, including four known effectors of neuronal differentiation, 11 factors with limited evidence of involvement in regulating neuronal differentiation, and nine novel factors. One transcription factor, Oct-2, was studied in detail and found to be a bifunctional regulator: It can either repress or induce neuronal differentiation, depending on the particular isoform. Ectopic expression experiments demonstrate that isoform Oct-2.4 represses neuronal differentiation, whereas Oct-2.2 activates neuron formation. Consistent with a role in neuronal differentiation, Oct-2.2 expression is induced during differentiation, and cells depleted of Oct-2 and its homolog Oct-1 have a reduced capacity to differentiate into neurons. Our results reveal a number of transcription factors potentially important for mammalian neuronal differentiation, and indicate that Oct-2 may serve as a binary switch to repress differentiation in precursor cells and induce neuronal differentiation later during neuronal development.
Asunto(s)
Células Madre Embrionarias/citología , Células Madre Embrionarias/metabolismo , Neurogénesis/fisiología , Neuronas/citología , Factor 2 de Transcripción de Unión a Octámeros/metabolismo , Animales , Línea Celular , Humanos , Ratones , Sistemas de Lectura Abierta , Transportador 1 de Catión Orgánico/metabolismo , Isoformas de Proteínas/metabolismo , Empalme de Proteína , Proteínas Represoras/metabolismo , TransgenesRESUMEN
BACKGROUND AND AIMS: Aberrant upregulation of POU2F2 expression has been discovered in metastatic gastric cancer (GC). However, the mechanisms underlying the aberrant upregulation and the potential functions of POU2F2 remain uncertain. DESIGN: The role and mechanism of POU2F2 in GC metastasis were investigated in gastric epithelial cells, GC cell lines and an experimental metastasis animal model by gain of function and loss of function. Upstream and downstream targets of POU2F2 were selected by bioinformatics and identified by luciferase reporter assay, electrophoretic mobility shift assay and chromatin immunoprecipitation PCR. The influence of miR-218 on its putative target genes (POU2F2, ROBO1 and IKK-ß) and GC metastasis was further explored via in vitro and in vivo approaches. RESULTS: Increased POU2F2 expression was detected in metastatic GC cell lines and patient samples. POU2F2 was induced by the activation of nuclear factor (NF)-κB and, in turn, regulated ROBO1 transcription, thus functionally contributing to GC metastasis. Finally, miR-218 was found to suppress GC metastasis by simultaneously mediating multiple molecules in the POU2F2-oriented network. CONCLUSIONS: This study demonstrated that NF-κB and the SLIT2/ROBO1 interaction network with POU2F2 as the central part may exert critical effects on tumour metastasis. Blocking the activation of the POU2F2-oriented metastasis network using miR-218 precursors exemplified a promising approach that sheds light on new strategies for GC treatment.
Asunto(s)
Péptidos y Proteínas de Señalización Intercelular/metabolismo , MicroARNs , Metástasis de la Neoplasia/genética , Proteínas del Tejido Nervioso/metabolismo , Factor 2 de Transcripción de Unión a Octámeros/genética , Receptores Inmunológicos/metabolismo , Neoplasias Gástricas , Animales , Línea Celular Tumoral , Movimiento Celular , Modelos Animales de Enfermedad , Regulación Neoplásica de la Expresión Génica , Humanos , Ratones , MicroARNs/genética , MicroARNs/metabolismo , FN-kappa B/metabolismo , Neoplasias Gástricas/genética , Neoplasias Gástricas/metabolismo , Neoplasias Gástricas/patología , Regulación hacia Arriba , Proteínas RoundaboutRESUMEN
Follicular lymphoma (FL) constitutes the second most common non-Hodgkin lymphoma in the western world. FL carries characteristic recurrent structural genomic aberrations. However, information regarding the coding genome in FL is still evolving. Here, we describe the results of massively parallel exome sequencing and single nucleotide polymorphism 6.0 array genomic profiling of 11 highly purified FL cases, and 1 transformed FL case and the validation of selected mutations in 102 FL cases. We report the identification of 15 novel recurrently mutated genes in FL. These include frequent mutations in the linker histone genes HIST1H1 B-E (27%) and mutations in OCT2 (also known as POU2F2; 8%), IRF8 (6%), and ARID1A (11%). A subset of the mutations in HIST1H1 B-E affected binding to DNMT3B, and mutations in HIST1H1 B-E and in EZH2 or ARID1A were largely mutually exclusive, implicating HIST1H1 B-E in epigenetic deregulation in FL. Mutations in OCT2 (POU2F2) affected its transcriptional and functional properties as measured through luciferase assays, the biological analysis of stably transduced cell lines, and global expression profiling. Finally, multiple novel mutated genes located within regions of acquired uniparental disomy in FL are identified. In aggregate, these data substantially broaden our understanding of the genomic pathogenesis of FL.
Asunto(s)
Histonas/genética , Factores Reguladores del Interferón/genética , Linfoma Folicular/genética , Mutación , Proteínas Nucleares/genética , Factor 2 de Transcripción de Unión a Octámeros , Secuencia de Aminoácidos , Proteína de Unión a CREB/genética , Análisis Mutacional de ADN , Proteínas de Unión al ADN/genética , Proteína Potenciadora del Homólogo Zeste 2 , Epigénesis Genética , Exoma , Secuenciación de Nucleótidos de Alto Rendimiento , Histonas/química , Humanos , Datos de Secuencia Molecular , Proteínas de Neoplasias/genética , Complejo Represivo Polycomb 2/genética , Polimorfismo de Nucleótido Simple , Proteínas Proto-Oncogénicas c-bcl-2/genética , Miembro 14 de Receptores del Factor de Necrosis Tumoral/genética , Alineación de Secuencia , Factores de Transcripción , Activación TranscripcionalRESUMEN
MicroRNAs (MiRs) are small, noncoding RNAs that regulate gene expression posttranscriptionally. In this study, we show that MiR-210 is induced by Oct-2, a key transcriptional mediator of B cell activation. Germline deletion of MiR-210 results in the development of autoantibodies from 5 mo of age. Overexpression of MiR-210 in vivo resulted in cell autonomous expansion of the B1 lineage and impaired fitness of B2 cells. Mice overexpressing MiR-210 exhibited impaired class-switched Ab responses, a finding confirmed in wild-type B cells transfected with a MiR-210 mimic. In vitro studies demonstrated defects in cellular proliferation and cell cycle entry, which were consistent with the transcriptomic analysis demonstrating downregulation of genes involved in cellular proliferation and B cell activation. These findings indicate that Oct-2 induction of MiR-210 provides a novel inhibitory mechanism for the control of B cells and autoantibody production.