Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 17 de 17
Filtrar
1.
Mol Cancer ; 23(1): 40, 2024 02 22.
Artículo en Inglés | MEDLINE | ID: mdl-38383439

RESUMEN

Finding effective therapeutic targets to treat NRAS-mutated melanoma remains a challenge. Long non-coding RNAs (lncRNAs) recently emerged as essential regulators of tumorigenesis. Using a discovery approach combining experimental models and unbiased computational analysis complemented by validation in patient biospecimens, we identified a nuclear-enriched lncRNA (AC004540.4) that is upregulated in NRAS/MAPK-dependent melanoma, and that we named T-RECS. Considering potential innovative treatment strategies, we designed antisense oligonucleotides (ASOs) to target T-RECS. T-RECS ASOs reduced the growth of melanoma cells and induced apoptotic cell death, while having minimal impact on normal primary melanocytes. Mechanistically, treatment with T-RECS ASOs downregulated the activity of pro-survival kinases and reduced the protein stability of hnRNPA2/B1, a pro-oncogenic regulator of MAPK signaling. Using patient- and cell line- derived tumor xenograft mouse models, we demonstrated that systemic treatment with T-RECS ASOs significantly suppressed the growth of melanoma tumors, with no noticeable toxicity. ASO-mediated T-RECS inhibition represents a promising RNA-targeting approach to improve the outcome of MAPK pathway-activated melanoma.


Asunto(s)
Melanoma , ARN Largo no Codificante , Humanos , Ratones , Animales , Melanoma/patología , ARN Largo no Codificante/genética , Apoptosis/genética , Oligonucleótidos Antisentido/genética , Oligonucleótidos Antisentido/uso terapéutico , Línea Celular Tumoral , Proteínas de la Membrana/genética , GTP Fosfohidrolasas/genética , GTP Fosfohidrolasas/metabolismo
2.
Nature ; 514(7521): 257-61, 2014 Oct 09.
Artículo en Inglés | MEDLINE | ID: mdl-25119036

RESUMEN

Homeodomain proteins, described 30 years ago, exert essential roles in development as regulators of target gene expression; however, the molecular mechanisms underlying transcriptional activity of homeodomain factors remain poorly understood. Here investigation of a developmentally required POU-homeodomain transcription factor, Pit1 (also known as Pou1f1), has revealed that, unexpectedly, binding of Pit1-occupied enhancers to a nuclear matrin-3-rich network/architecture is a key event in effective activation of the Pit1-regulated enhancer/coding gene transcriptional program. Pit1 association with Satb1 (ref. 8) and ß-catenin is required for this tethering event. A naturally occurring, dominant negative, point mutation in human PIT1(R271W), causing combined pituitary hormone deficiency, results in loss of Pit1 association with ß-catenin and Satb1 and therefore the matrin-3-rich network, blocking Pit1-dependent enhancer/coding target gene activation. This defective activation can be rescued by artificial tethering of the mutant R271W Pit1 protein to the matrin-3 network, bypassing the pre-requisite association with ß-catenin and Satb1 otherwise required. The matrin-3 network-tethered R271W Pit1 mutant, but not the untethered protein, restores Pit1-dependent activation of the enhancers and recruitment of co-activators, exemplified by p300, causing both enhancer RNA transcription and target gene activation. These studies have thus revealed an unanticipated homeodomain factor/ß-catenin/Satb1-dependent localization of target gene regulatory enhancer regions to a subnuclear architectural structure that serves as an underlying mechanism by which an enhancer-bound homeodomain factor effectively activates developmental gene transcriptional programs.


Asunto(s)
Elementos de Facilitación Genéticos/genética , Regulación del Desarrollo de la Expresión Génica , Proteínas de Homeodominio/metabolismo , Proteínas Asociadas a Matriz Nuclear/metabolismo , Proteínas de Unión al ARN/metabolismo , Transcripción Genética , Animales , Células Cultivadas , Proteínas de Homeodominio/genética , Humanos , Proteínas de Unión a la Región de Fijación a la Matriz/metabolismo , Ratones , Hipófisis/embriología , Hipófisis/metabolismo , Unión Proteica , Factor de Transcripción Pit-1/genética , Factor de Transcripción Pit-1/metabolismo , Transcripción Genética/genética , beta Catenina/metabolismo
3.
BMC Biol ; 17(1): 104, 2019 12 13.
Artículo en Inglés | MEDLINE | ID: mdl-31830989

RESUMEN

BACKGROUND: Polarity is necessary for epithelial cells to perform distinct functions at their apical and basal surfaces. Oral epithelial cell-derived ameloblasts at secretory stage (SABs) synthesize large amounts of enamel matrix proteins (EMPs), largely amelogenins. EMPs are unidirectionally secreted into the enamel space through their apical cytoplasmic protrusions, or Tomes' processes (TPs), to guide the enamel formation. Little is known about the transcriptional regulation underlying the establishment of cell polarity and unidirectional secretion of SABs. RESULTS: The higher-order chromatin architecture of eukaryotic genome plays important roles in cell- and stage-specific transcriptional programming. A genome organizer, special AT-rich sequence-binding protein 1 (SATB1), was discovered to be significantly upregulated in ameloblasts compared to oral epithelial cells using a whole-transcript microarray analysis. The Satb1-/- mice possessed deformed ameloblasts and a thin layer of hypomineralized and non-prismatic enamel. Remarkably, Satb1-/- ameloblasts at the secretory stage lost many morphological characteristics found at the apical surface of wild-type (wt) SABs, including the loss of Tomes' processes, defective inter-ameloblastic adhesion, and filamentous actin architecture. As expected, the secretory function of Satb1-/- SABs was compromised as amelogenins were largely retained in cells. We found the expression of epidermal growth factor receptor pathway substrate 8 (Eps8), a known regulator for actin filament assembly and small intestinal epithelial cytoplasmic protrusion formation, to be SATB1 dependent. In contrast to wt SABs, EPS8 could not be detected at the apical surface of Satb1-/- SABs. Eps8 expression was greatly reduced in small intestinal epithelial cells in Satb1-/- mice as well, displaying defective intestinal microvilli. CONCLUSIONS: Our data show that SATB1 is essential for establishing secretory ameloblast cell polarity and for EMP secretion. In line with the deformed apical architecture, amelogenin transport to the apical secretory front and secretion into enamel space were impeded in Satb1-/- SABs resulting in a massive cytoplasmic accumulation of amelogenins and a thin layer of hypomineralized enamel. Our studies strongly suggest that SATB1-dependent Eps8 expression plays a critical role in cytoplasmic protrusion formation in both SABs and in small intestines. This study demonstrates the role of SATB1 in the regulation of amelogenesis and the potential application of SATB1 in ameloblast/enamel regeneration.


Asunto(s)
Ameloblastos/fisiología , Amelogénesis , Polaridad Celular , Esmalte Dental/crecimiento & desarrollo , Proteínas de Unión a la Región de Fijación a la Matriz/genética , Animales , Diferenciación Celular , Humanos , Proteínas de Unión a la Región de Fijación a la Matriz/metabolismo , Ratones
4.
Proc Natl Acad Sci U S A ; 109(47): 19071-8, 2012 Nov 20.
Artículo en Inglés | MEDLINE | ID: mdl-23144223

RESUMEN

Neurons within each layer in the mammalian cortex have stereotypic projections. Four genes-Fezf2, Ctip2, Tbr1, and Satb2-regulate these projection identities. These genes also interact with each other, and it is unclear how these interactions shape the final projection identity. Here we show, by generating double mutants of Fezf2, Ctip2, and Satb2, that cortical neurons deploy a complex genetic switch that uses mutual repression to produce subcortical or callosal projections. We discovered that Tbr1, EphA4, and Unc5H3 are critical downstream targets of Satb2 in callosal fate specification. This represents a unique role for Tbr1, implicated previously in specifying corticothalamic projections. We further show that Tbr1 expression is dually regulated by Satb2 and Ctip2 in layers 2-5. Finally, we show that Satb2 and Fezf2 regulate two disease-related genes, Auts2 (Autistic Susceptibility Gene2) and Bhlhb5 (mutated in Hereditary Spastic Paraplegia), providing a molecular handle to investigate circuit disorders in neurodevelopmental diseases.


Asunto(s)
Redes Reguladoras de Genes , Neocórtex/crecimiento & desarrollo , Neocórtex/metabolismo , Neuronas/metabolismo , Proteínas Represoras/metabolismo , Fosfatasa Alcalina/metabolismo , Animales , Axones/enzimología , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/metabolismo , Moléculas de Adhesión Celular Neuronal/metabolismo , Corteza Cerebral/metabolismo , Proteínas del Citoesqueleto , Proteínas de Unión al ADN/genética , Proteínas de Unión al ADN/metabolismo , Proteínas Ligadas a GPI/metabolismo , Regulación del Desarrollo de la Expresión Génica , Sitios Genéticos/genética , Isoenzimas/metabolismo , Ratones , Mutación/genética , Proteínas del Tejido Nervioso/metabolismo , Receptores de Netrina , Proteínas Nucleares/metabolismo , Unión Proteica , Receptor EphA4/metabolismo , Receptores de Superficie Celular/metabolismo , Proteínas Represoras/genética , Proteínas de Dominio T Box , Tálamo/metabolismo , Factores de Transcripción , Proteínas Supresoras de Tumor/metabolismo
5.
Semin Cancer Biol ; 23(2): 72-9, 2013 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-22771615

RESUMEN

When cells change functions or activities (such as during differentiation, response to extracellular stimuli, or migration), gene expression undergoes large-scale reprogramming, in cell type- and function-specific manners. Large changes in gene regulation require changes in chromatin architecture, which involve recruitment of chromatin remodeling enzymes and epigenomic modification enzymes to specific genomic loci. Transcription factors must also be accurately assembled at these loci. SATB1 is a genome organizer protein that facilitates these processes, providing a nuclear architectural platform that anchors hundreds of genes, through its interaction with specific genomic sequences; this activity allows expression of all these genes to be regulated in parallel, and enables cells to thereby alter their function. We review and describe future perspectives on SATB1 function in higher-order chromatin structure and gene regulation, and its role in metastasis of breast cancer and other tumor types.


Asunto(s)
Ensamble y Desensamble de Cromatina/genética , Proteínas de Unión a la Región de Fijación a la Matriz/fisiología , Neoplasias/genética , Animales , Neoplasias de la Mama/genética , Neoplasias de la Mama/patología , Cromatina/química , Cromatina/metabolismo , Progresión de la Enfermedad , Epigénesis Genética/genética , Femenino , Genoma Humano/fisiología , Humanos , Proteínas de Unión a la Región de Fijación a la Matriz/genética , Modelos Biológicos
6.
Nature ; 452(7184): 187-93, 2008 Mar 13.
Artículo en Inglés | MEDLINE | ID: mdl-18337816

RESUMEN

Mechanisms underlying global changes in gene expression during tumour progression are poorly understood. SATB1 is a genome organizer that tethers multiple genomic loci and recruits chromatin-remodelling enzymes to regulate chromatin structure and gene expression. Here we show that SATB1 is expressed by aggressive breast cancer cells and its expression level has high prognostic significance (P < 0.0001), independent of lymph-node status. RNA-interference-mediated knockdown of SATB1 in highly aggressive (MDA-MB-231) cancer cells altered the expression of >1,000 genes, reversing tumorigenesis by restoring breast-like acinar polarity and inhibiting tumour growth and metastasis in vivo. Conversely, ectopic SATB1 expression in non-aggressive (SKBR3) cells led to gene expression patterns consistent with aggressive-tumour phenotypes, acquiring metastatic activity in vivo. SATB1 delineates specific epigenetic modifications at target gene loci, directly upregulating metastasis-associated genes while downregulating tumour-suppressor genes. SATB1 reprogrammes chromatin organization and the transcription profiles of breast tumours to promote growth and metastasis; this is a new mechanism of tumour progression.


Asunto(s)
Neoplasias de la Mama/genética , Neoplasias de la Mama/patología , Regulación Neoplásica de la Expresión Génica/genética , Proteínas de Unión a la Región de Fijación a la Matriz/metabolismo , Metástasis de la Neoplasia/genética , Animales , Biomarcadores de Tumor/análisis , Neoplasias de la Mama/diagnóstico , Línea Celular , Línea Celular Tumoral , Polaridad Celular , Progresión de la Enfermedad , Epigénesis Genética/genética , Perfilación de la Expresión Génica , Humanos , Neoplasias Pulmonares/metabolismo , Neoplasias Pulmonares/patología , Metástasis Linfática/diagnóstico , Metástasis Linfática/genética , Metástasis Linfática/patología , Proteínas de Unión a la Región de Fijación a la Matriz/deficiencia , Proteínas de Unión a la Región de Fijación a la Matriz/genética , Ratones , Ratones Desnudos , Metástasis de la Neoplasia/diagnóstico , Metástasis de la Neoplasia/patología , Trasplante de Neoplasias , Fenotipo , Pronóstico , Interferencia de ARN
7.
Methods ; 58(3): 243-54, 2012 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-22782115

RESUMEN

Mammalian genomes are organized into multiple layers of higher-order chromatin structure, and in this organization chromatin looping is a striking and crucial feature that brings together distal genomic loci into close spatial proximity. Such three-dimensional organization of chromatin has been suggested to be functionally important in gene regulation. Many important questions need to be addressed, such as what types of nuclear proteins are responsible for folding chromatin into loops, whether there are any genomic marks that serve as the core sites of chromatin folding events, how distal genomic sites are brought together, and what are the biological consequences for interactions between distal genomic loci. In order to address these fundamental questions, it is essential to devise and employ methods that can capture higher-order structures formed by specific nuclear proteins at high resolution. In this article, in order to describe methods of analyzing protein-mediated chromatin interactions, we will use as an example a global genome-organizer protein, SATB1, which mediates chromatin looping.


Asunto(s)
Cromatina/genética , Proteínas de Unión a la Región de Fijación a la Matriz/fisiología , Animales , Secuencia de Bases , Sitios de Unión , Inmunoprecipitación de Cromatina/métodos , Mapeo Cromosómico/métodos , Reactivos de Enlaces Cruzados/química , Formaldehído/química , Humanos , Modelos Moleculares , Datos de Secuencia Molecular , Conformación de Ácido Nucleico
8.
Res Sq ; 2023 Dec 01.
Artículo en Inglés | MEDLINE | ID: mdl-38077055

RESUMEN

Finding effective therapeutic targets to treat NRAS-mutated melanoma remains a challenge. Long non-coding RNAs (lncRNAs) recently emerged as essential regulators of tumorigenesis. Using a discovery approach combining experimental models and unbiased computational analysis complemented by validation in patient biospecimens, we identified a nuclear-enriched lncRNA (AC004540.4) that is upregulated in NRAS/MAPK-dependent melanoma, and that we named T-RECS. Considering potential innovative treatment strategies, we designed antisense oligonucleotides (ASOs) to target T-RECS. T-RECS ASOs reduced the growth of melanoma cells and induced apoptotic cell death, while having minimal impacton normal primary melanocytes. Mechanistically, treatment with T-RECS ASOs downregulated the activity of pro-survival kinases and reduced the protein stability of hnRNPA2/B1, a pro-oncogenic regulator of MAPK signaling. Using patient- and cell line- derived tumor xenograft mouse models, we demonstrated that systemic treatment with T-RECS ASOs significantly suppressed the growth of melanoma tumors, with no noticeable toxicity. ASO-mediated T-RECS inhibition represents a promising RNA-targeting approach to improve the outcome of MAPK pathway-activated melanoma.

9.
Nat Commun ; 13(1): 5554, 2022 09 22.
Artículo en Inglés | MEDLINE | ID: mdl-36138028

RESUMEN

CD4+ and CD8+ double-positive (DP) thymocytes play a crucial role in T cell development in the thymus. DP cells rearrange the T cell receptor gene Tcra to generate T cell receptors with TCRß. DP cells differentiate into CD4 or CD8 single-positive (SP) thymocytes, regulatory T cells, or invariant nature kill T cells (iNKT) in response to TCR signaling. Chromatin organizer SATB1 is highly expressed in DP cells and is essential in regulating Tcra rearrangement and differentiation of DP cells. Here we explored the mechanism of SATB1 orchestrating gene expression in DP cells. Single-cell RNA sequencing shows that Satb1 deletion changes the cell identity of DP thymocytes and down-regulates genes specifically and highly expressed in DP cells. Super-enhancers regulate the expressions of DP-specific genes, and our Hi-C data show that SATB1 deficiency in thymocytes reduces super-enhancer activity by specifically decreasing interactions among super-enhancers and between super-enhancers and promoters. Our results reveal that SATB1 plays a critical role in thymocyte development to promote the establishment of DP cell identity by globally regulating super-enhancers of DP cells at the chromatin architectural level.


Asunto(s)
Proteínas de Unión a la Región de Fijación a la Matriz , Timocitos , Antígenos CD4/genética , Antígenos CD4/metabolismo , Linfocitos T CD4-Positivos/metabolismo , Antígenos CD8/genética , Antígenos CD8/metabolismo , Linfocitos T CD8-positivos/metabolismo , Diferenciación Celular/genética , Cromatina/metabolismo , Proteínas de Unión a la Región de Fijación a la Matriz/genética , Proteínas de Unión a la Región de Fijación a la Matriz/metabolismo , Receptores de Antígenos de Linfocitos T/metabolismo , Receptores de Antígenos de Linfocitos T alfa-beta/genética , Timo/metabolismo
10.
Cell Rep ; 19(6): 1176-1188, 2017 05 09.
Artículo en Inglés | MEDLINE | ID: mdl-28494867

RESUMEN

T cell receptor (TCR) signaling by MHC class I and II induces thymocytes to acquire cytotoxic and helper fates via the induction of Runx3 and ThPOK transcription factors, respectively. The mechanisms by which TCR signaling is translated into transcriptional programs for each cell fate remain elusive. Here, we show that, in post-selection thymocytes, a genome organizer, SATB1, activates genes for lineage-specifying factors, including ThPOK, Runx3, CD4, CD8, and Treg factor Foxp3, via regulating enhancers in these genes in a locus-specific manner. Indeed, SATB1-deficient thymocytes are partially re-directed into inappropriate T lineages after both MHC class I- and II-mediated selection, and they fail to generate NKT and Treg subsets. Despite its essential role in activating enhancers for the gene encoding ThPOK in TCR-signaled thymocytes, SATB1 becomes dispensable for maintaining ThPOK in CD4+ T cells. Collectively, our findings demonstrate that SATB1 shapes the primary T cell pool by directing lineage-specific transcriptional programs in the thymus.


Asunto(s)
Linfopoyesis , Proteínas de Unión a la Región de Fijación a la Matriz/metabolismo , Subgrupos de Linfocitos T/citología , Animales , Antígenos CD4/genética , Antígenos CD4/metabolismo , Antígenos CD8/genética , Antígenos CD8/metabolismo , Linaje de la Célula , Subunidad alfa 3 del Factor de Unión al Sitio Principal/genética , Subunidad alfa 3 del Factor de Unión al Sitio Principal/metabolismo , Elementos de Facilitación Genéticos , Factores de Transcripción Forkhead/genética , Factores de Transcripción Forkhead/metabolismo , Proteínas de Unión a la Región de Fijación a la Matriz/genética , Ratones , Subgrupos de Linfocitos T/metabolismo , Timo/citología , Timo/metabolismo , Factores de Transcripción/genética , Factores de Transcripción/metabolismo , Activación Transcripcional
11.
Neuron ; 91(4): 763-776, 2016 Aug 17.
Artículo en Inglés | MEDLINE | ID: mdl-27478017

RESUMEN

Motor behaviors such as walking or withdrawing the limb from a painful stimulus rely upon integrative multimodal sensory circuitry to generate appropriate muscle activation patterns. Both the cellular components and the molecular mechanisms that instruct the assembly of the spinal sensorimotor system are poorly understood. Here we characterize the connectivity pattern of a sub-population of lamina V inhibitory sensory relay neurons marked during development by the nuclear matrix and DNA binding factor Satb2 (ISR(Satb2)). ISR(Satb2) neurons receive inputs from multiple streams of sensory information and relay their outputs to motor command layers of the spinal cord. Deletion of the Satb2 transcription factor from ISR(Satb2) neurons perturbs their cellular position, molecular profile, and pre- and post-synaptic connectivity. These alterations are accompanied by abnormal limb hyperflexion responses to mechanical and thermal stimuli and during walking. Thus, Satb2 is a genetic determinant that mediates proper circuit development in a core sensory-to-motor spinal network.


Asunto(s)
Extremidades/fisiología , Proteínas de Unión a la Región de Fijación a la Matriz/fisiología , Vías Nerviosas/fisiología , Dolor/fisiopatología , Células Receptoras Sensoriales/fisiología , Médula Espinal/fisiología , Factores de Transcripción/fisiología , Caminata/fisiología , Animales , Interneuronas/fisiología , Proteínas de Unión a la Región de Fijación a la Matriz/genética , Ratones , Ratones Noqueados , Mutación , Reflejo/fisiología , Factores de Transcripción/genética
12.
J Exp Med ; 212(5): 809-24, 2015 May 04.
Artículo en Inglés | MEDLINE | ID: mdl-25847946

RESUMEN

Rag1 and Rag2 gene expression in CD4(+)CD8(+) double-positive (DP) thymocytes depends on the activity of a distant anti-silencer element (ASE) that counteracts the activity of an intergenic silencer. However, the mechanistic basis for ASE activity is unknown. Here, we show that the ASE physically interacts with the distant Rag1 and Rag2 gene promoters in DP thymocytes, bringing the two promoters together to form an active chromatin hub. Moreover, we show that the ASE functions as a classical enhancer that can potently activate these promoters in the absence of the silencer or other locus elements. In thymocytes lacking the chromatin organizer SATB1, we identified a partial defect in Tcra gene rearrangement that was associated with reduced expression of Rag1 and Rag2 at the DP stage. SATB1 binds to the ASE and Rag promoters, facilitating inclusion of Rag2 in the chromatin hub and the loading of RNA polymerase II to both the Rag1 and Rag2 promoters. Our results provide a novel framework for understanding ASE function and demonstrate a novel role for SATB1 as a regulator of Rag locus organization and gene expression in DP thymocytes.


Asunto(s)
Diferenciación Celular/inmunología , Cromatina/inmunología , Proteínas de Unión al ADN/inmunología , Proteínas de Homeodominio/inmunología , Proteínas de Unión a la Región de Fijación a la Matriz/inmunología , Elementos de Respuesta/inmunología , Timocitos/inmunología , Animales , Diferenciación Celular/genética , Cromatina/genética , Proteínas de Unión al ADN/genética , Regulación de la Expresión Génica/genética , Proteínas de Homeodominio/genética , Proteínas de Unión a la Región de Fijación a la Matriz/genética , Ratones , Ratones Noqueados , Timocitos/citología
13.
Mol Cell Biol ; 32(2): 333-47, 2012 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-22064485

RESUMEN

Complex behaviors, such as learning and memory, are associated with rapid changes in gene expression of neurons and subsequent formation of new synaptic connections. However, how external signals are processed to drive specific changes in gene expression is largely unknown. We found that the genome organizer protein Satb1 is highly expressed in mature neurons, primarily in the cerebral cortex, dentate hilus, and amygdala. In Satb1-null mice, cortical layer morphology was normal. However, in postnatal Satb1-null cortical pyramidal neurons, we found a substantial decrease in the density of dendritic spines, which play critical roles in synaptic transmission and plasticity. Further, we found that in the cerebral cortex, Satb1 binds to genomic loci of multiple immediate early genes (IEGs) (Fos, Fosb, Egr1, Egr2, Arc, and Bdnf) and other key neuronal genes, many of which have been implicated in synaptic plasticity. Loss of Satb1 resulted in greatly alters timing and expression levels of these IEGs during early postnatal cerebral cortical development and also upon stimulation in cortical organotypic cultures. These data indicate that Satb1 is required for proper temporal dynamics of IEG expression. Based on these findings, we propose that Satb1 plays a critical role in cortical neurons to facilitate neuronal plasticity.


Asunto(s)
Encéfalo/crecimiento & desarrollo , Espinas Dendríticas/metabolismo , Regulación del Desarrollo de la Expresión Génica , Genes Inmediatos-Precoces , Proteínas de Unión a la Región de Fijación a la Matriz/genética , Proteínas de Unión a la Región de Fijación a la Matriz/metabolismo , Animales , Encéfalo/citología , Encéfalo/metabolismo , ADN/metabolismo , Espinas Dendríticas/genética , Eliminación de Gen , Sitios Genéticos , Masculino , Proteínas de Unión a la Región de Fijación a la Matriz/análisis , Ratones , Neuronas/citología , Neuronas/metabolismo , Unión Proteica , Factores de Transcripción/análisis , Factores de Transcripción/genética , Factores de Transcripción/metabolismo
14.
PLoS One ; 7(12): e51786, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-23251624

RESUMEN

SATB1 drives metastasis when expressed in breast tumor cells by radically reprogramming gene expression. Here, we show that SATB1 also has an oncogenic activity to transform certain non-malignant breast epithelial cell lines. We studied the non-malignant MCF10A cell line, which is used widely in the literature. We obtained aliquots from two different sources (here we refer to them as MCF10A-1 and MCF10A-2), but found them to be surprisingly dissimilar in their responses to oncogenic activity of SATB1. Ectopic expression of SATB1 in MCF10A-1 induced tumor-like morphology in three-dimensional cultures, led to tumor formation in immunocompromised mice, and when injected into tail veins, led to lung metastasis. The number of metastases correlated positively with the level of SATB1 expression. In contrast, SATB1 expression in MCF10A-2 did not lead to any of these outcomes. Yet DNA copy-number analysis revealed that MCF10A-1 is indistinguishable genetically from MCF10A-2. However, gene expression profiling analysis revealed that these cell lines have significantly divergent signatures for the expression of genes involved in oncogenesis, including cell cycle regulation and signal transduction. Above all, the early DNA damage-response kinase, ATM, was greatly reduced in MCF10A-1 cells compared to MCF10A-2 cells. We found the reason for reduction to be phenotypic drift due to long-term cultivation of MCF10A. ATM knockdown in MCF10A-2 and two other non-malignant breast epithelial cell lines, 184A1 and 184B4, enabled SATB1 to induce malignant phenotypes similar to that observed for MCF10A-1. These data indicate a novel role for ATM as a suppressor of SATB1-induced malignancy in breast epithelial cells, but also raise a cautionary note that phenotypic drift could lead to dramatically different functional outcomes.


Asunto(s)
Mama/patología , Proteínas de Ciclo Celular/metabolismo , Transformación Celular Neoplásica/patología , Proteínas de Unión al ADN/metabolismo , Progresión de la Enfermedad , Células Epiteliales/metabolismo , Células Epiteliales/patología , Proteínas de Unión a la Región de Fijación a la Matriz/metabolismo , Proteínas Serina-Treonina Quinasas/metabolismo , Proteínas Supresoras de Tumor/metabolismo , Animales , Proteínas de la Ataxia Telangiectasia Mutada , Neoplasias de la Mama/genética , Neoplasias de la Mama/patología , Línea Celular Tumoral , Proliferación Celular , Transformación Celular Neoplásica/genética , Transición Epitelial-Mesenquimal/genética , Femenino , Perfilación de la Expresión Génica , Regulación Neoplásica de la Expresión Génica , Humanos , Pulmón/patología , Ratones , Ratones Desnudos , Células 3T3 NIH , Invasividad Neoplásica , Fenotipo
15.
J Cell Biol ; 194(6): 825-39, 2011 Sep 19.
Artículo en Inglés | MEDLINE | ID: mdl-21930775

RESUMEN

During development, multipotent progenitor cells establish tissue-specific programs of gene expression. In this paper, we show that p63 transcription factor, a master regulator of epidermal morphogenesis, executes its function in part by directly regulating expression of the genome organizer Satb1 in progenitor cells. p63 binds to a proximal regulatory region of the Satb1 gene, and p63 ablation results in marked reduction in the Satb1 expression levels in the epidermis. Satb1(-/-) mice show impaired epidermal morphology. In Satb1-null epidermis, chromatin architecture of the epidermal differentiation complex locus containing genes associated with epidermal differentiation is altered primarily at its central domain, where Satb1 binding was confirmed by chromatin immunoprecipitation-on-chip analysis. Furthermore, genes within this domain fail to be properly activated upon terminal differentiation. Satb1 expression in p63(+/-) skin explants treated with p63 small interfering ribonucleic acid partially restored the epidermal phenotype of p63-deficient mice. These data provide a novel mechanism by which Satb1, a direct downstream target of p63, contributes in epidermal morphogenesis via establishing tissue-specific chromatin organization and gene expression in epidermal progenitor cells.


Asunto(s)
Ensamble y Desensamble de Cromatina/genética , Cromatina/metabolismo , Epidermis/embriología , Regulación del Desarrollo de la Expresión Génica , Proteínas de Unión a la Región de Fijación a la Matriz/genética , Fosfoproteínas/genética , Transactivadores/genética , Animales , Diferenciación Celular , Células Epidérmicas , Genoma , Hibridación Fluorescente in Situ , Proteínas de Unión a la Región de Fijación a la Matriz/metabolismo , Ratones , Ratones Endogámicos C57BL , Fosfoproteínas/metabolismo , Transactivadores/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA