Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 45
Filtrar
1.
Nucleic Acids Res ; 52(7): e40, 2024 Apr 24.
Artículo en Inglés | MEDLINE | ID: mdl-38499482

RESUMEN

Genome-wide binding assays aspire to map the complete binding pattern of gene regulators. Common practice relies on replication-duplicates or triplicates-and high stringency statistics to favor false negatives over false positives. Here we show that duplicates and triplicates of CUT&RUN are not sufficient to discover the entire activity of transcriptional regulators. We introduce ICEBERG (Increased Capture of Enrichment By Exhaustive Replicate aGgregation), a pipeline that harnesses large numbers of CUT&RUN replicates to discover the full set of binding events and chart the line between false positives and false negatives. We employed ICEBERG to map the full set of H3K4me3-marked regions, the targets of the co-factor ß-catenin, and those of the transcription factor TBX3, in human colorectal cancer cells. The ICEBERG datasets allow benchmarking of individual replicates, comparing the performance of peak calling and replication approaches, and expose the arbitrary nature of strategies to identify reproducible peaks. Instead of a static view of genomic targets, ICEBERG establishes a spectrum of detection probabilities across the genome for a given factor, underlying the intrinsic dynamicity of its mechanism of action, and permitting to distinguish frequent from rare regulation events. Finally, ICEBERG discovered instances, undetectable with other approaches, that underlie novel mechanisms of colorectal cancer progression.


Asunto(s)
Programas Informáticos , Transcripción Genética , Humanos , beta Catenina/metabolismo , beta Catenina/genética , Sitios de Unión , Línea Celular Tumoral , Secuenciación de Inmunoprecipitación de Cromatina , Neoplasias Colorrectales/genética , Neoplasias Colorrectales/metabolismo , Genoma Humano , Histonas/metabolismo , Histonas/genética , Unión Proteica , Proteínas de Dominio T Box/genética , Proteínas de Dominio T Box/metabolismo , Factores de Transcripción/metabolismo , Factores de Transcripción/genética
2.
Genes Dev ; 32(21-22): 1443-1458, 2018 11 01.
Artículo en Inglés | MEDLINE | ID: mdl-30366904

RESUMEN

Bcl9 and Pygopus (Pygo) are obligate Wnt/ß-catenin cofactors in Drosophila, yet their contribution to Wnt signaling during vertebrate development remains unresolved. Combining zebrafish and mouse genetics, we document a conserved, ß-catenin-associated function for BCL9 and Pygo proteins during vertebrate heart development. Disrupting the ß-catenin-BCL9-Pygo complex results in a broadly maintained canonical Wnt response yet perturbs heart development and proper expression of key cardiac regulators. Our work highlights BCL9 and Pygo as selective ß-catenin cofactors in a subset of canonical Wnt responses during vertebrate development. Moreover, our results implicate alterations in BCL9 and BCL9L in human congenital heart defects.


Asunto(s)
Cardiopatías Congénitas/genética , Péptidos y Proteínas de Señalización Intracelular/genética , Factores de Transcripción/genética , Vía de Señalización Wnt , Proteínas de Pez Cebra/genética , Proteínas Adaptadoras Transductoras de Señales , Animales , Corazón/embriología , Ratones , Mutación , Miocardio/metabolismo , Pez Cebra/embriología , Pez Cebra/genética , beta Catenina/metabolismo
3.
J Biol Chem ; 300(4): 107126, 2024 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-38432629

RESUMEN

The forkhead box family transcription factor FOXQ1 is highly induced in several types of carcinomas, where it promotes epithelial-to-mesenchymal transition and tumor metastasis. The molecular mechanisms that lead to FOXQ1 deregulation in cancer are incompletely understood. Here, we used CRISPR-Cas9-based genomic locus proteomics and promoter reporter constructs to discover transcriptional regulators of FOXQ1 and identified the tumor suppressor p53 as a negative regulator of FOXQ1 expression. Chromatin immunoprecipitation followed by quantitative PCR as well as complementary gain and loss-of-function assays in model cell lines indicated that p53 binds close to the transcription start site of the FOXQ1 promoter, and that it suppresses FOXQ1 expression in various cell types. Consistently, pharmacological activation of p53 using nutlin-3 or doxorubicin reduced FOXQ1 mRNA and protein levels in cancer cell lines harboring wildtype p53. Finally, we observed that p53 mutations are associated with increased FOXQ1 expression in human cancers. Altogether, these results suggest that loss of p53 function-a hallmark feature of many types of cancer-derepresses FOXQ1, which in turn promotes tumor progression.


Asunto(s)
Factores de Transcripción Forkhead , Regulación Neoplásica de la Expresión Génica , Proteína p53 Supresora de Tumor , Humanos , Factores de Transcripción Forkhead/genética , Factores de Transcripción Forkhead/metabolismo , Proteína p53 Supresora de Tumor/metabolismo , Proteína p53 Supresora de Tumor/genética , Línea Celular Tumoral , Regiones Promotoras Genéticas , Mutación
4.
Development ; 149(23)2022 12 01.
Artículo en Inglés | MEDLINE | ID: mdl-36355069

RESUMEN

Upon WNT/ß-catenin pathway activation, stabilized ß-catenin travels to the nucleus where it associates with the TCF/LEF transcription factors, constitutively bound to genomic Wnt-responsive elements (WREs), to activate target gene transcription. Discovering the binding profile of ß-catenin is therefore required to unambiguously assign direct targets of WNT signaling. Cleavage under targets and release using nuclease (CUT&RUN) has emerged as prime technique for mapping the binding profile of DNA-interacting proteins. Here, we present a modified version of CUT&RUN, named LoV-U (low volume and urea), that enables the robust and reproducible generation of ß-catenin binding profiles, uncovering direct WNT/ß-catenin target genes in human cells, as well as in cells isolated from developing mouse tissues. CUT&RUN-LoV-U outperforms original CUT&RUN when targeting co-factors that do not bind the DNA, can profile all classes of chromatin regulators and is well suited for simultaneous processing of several samples. We believe that the application of our protocol will allow the detection of the complex system of tissue-specific WNT/ß-catenin target genes, together with other non-DNA-binding transcriptional regulators that act downstream of ontogenetically fundamental signaling cascades.


Asunto(s)
Factores de Transcripción , beta Catenina , Humanos , Ratones , Animales , beta Catenina/genética , beta Catenina/metabolismo , Factores de Transcripción/genética , Factores de Transcripción/metabolismo , Factores de Transcripción TCF/metabolismo , Vía de Señalización Wnt/genética , Endonucleasas/metabolismo , Genómica , Urea , Activación Transcripcional
5.
J Cell Sci ; 135(19)2022 10 01.
Artículo en Inglés | MEDLINE | ID: mdl-36124643

RESUMEN

The forkhead box transcription factor FOXQ1 contributes to the pathogenesis of carcinomas. In colorectal cancers, FOXQ1 promotes tumour metastasis by inducing epithelial-to-mesenchymal transition (EMT) of cancer cells. FOXQ1 may exacerbate cancer by activating the oncogenic Wnt/ß-catenin signalling pathway. However, the role of FOXQ1 in the Wnt pathway remains to be resolved. Here, we report that FOXQ1 is an activator of Wnt-induced transcription and regulator of ß-catenin target gene expression. Upon Wnt pathway activation, FOXQ1 synergises with the ß-catenin nuclear complex to boost the expression of major Wnt targets. In parallel, we find that FOXQ1 controls the differential expression of various Wnt target genes in a ß-catenin-independent manner. Using RNA sequencing of colorectal cancer cell lines, we show that Wnt signalling and FOXQ1 converge on a transcriptional programme linked to EMT and cell migration. Additionally, we demonstrate that FOXQ1 occupies Wnt-responsive elements in ß-catenin target gene promoters and recruits a similar set of co-factors to the ß-catenin-associated transcription factor Tcf7l1. Taken together, our results indicate a multifaceted role of FOXQ1 in Wnt/ß-catenin signalling, which may drive the metastasis of colorectal cancers.


Asunto(s)
Neoplasias Colorrectales , beta Catenina , Carcinogénesis/genética , Línea Celular Tumoral , Proliferación Celular , Neoplasias Colorrectales/metabolismo , Transición Epitelial-Mesenquimal/genética , Factores de Transcripción Forkhead/metabolismo , Regulación Neoplásica de la Expresión Génica , Humanos , Vía de Señalización Wnt/genética , beta Catenina/genética , beta Catenina/metabolismo
6.
Mol Syst Biol ; 19(5): e11148, 2023 05 09.
Artículo en Inglés | MEDLINE | ID: mdl-36938679

RESUMEN

Early-life stress can result in life-long effects that impact adult health and disease risk, but little is known about how such programming is established and maintained. Here, we show that such epigenetic memories can be initiated in the Drosophila embryo before the major wave of zygotic transcription, and higher-order chromatin structures are established. An early short heat shock results in elevated levels of maternal miRNA and reduced levels of a subgroup of zygotic genes in stage 5 embryos. Using a Dicer-1 mutant, we show that the stress-induced decrease in one of these genes, the insulator-binding factor Elba1, is dependent on functional miRNA biogenesis. Reduction in Elba1 correlates with the upregulation of early developmental genes and promotes a sustained weakening of heterochromatin in the adult fly as indicated by an increased expression of the PEV wm4h reporter. We propose that maternal miRNAs, retained in response to an early embryonic heat shock, shape the subsequent de novo heterochromatin establishment that occurs during early development via direct or indirect regulation of some of the earliest expressed genes, including Elba1.


Asunto(s)
Proteínas de Drosophila , MicroARNs , Animales , Drosophila/genética , Drosophila/metabolismo , Heterocromatina/genética , Heterocromatina/metabolismo , MicroARNs/genética , MicroARNs/metabolismo , Embrión no Mamífero/metabolismo , Desarrollo Embrionario/genética , Proteínas de Drosophila/genética , Proteínas de Drosophila/metabolismo , Regulación del Desarrollo de la Expresión Génica , Drosophila melanogaster/metabolismo
7.
Exp Cell Res ; 429(2): 113646, 2023 08 15.
Artículo en Inglés | MEDLINE | ID: mdl-37271249

RESUMEN

Wnt signaling drives nuclear translocation of ß-catenin and its subsequent association with the DNA-bound TCF/LEF transcription factors, which dictate target gene specificity by recognizing Wnt responsive elements across the genome. ß-Catenin target genes are therefore thought to be collectively activated upon Wnt pathway stimulation. However, this appears in contrast with the non-overlapping patterns of Wnt target gene expression in several contexts, including early mammalian embryogenesis. Here we followed Wnt target gene expression in human embryonic stem cells after Wnt pathway stimulation at a single-cell resolution. Cells changed gene expression program over time consistent with three key developmental events: i) loss of pluripotency, ii) induction of Wnt target genes, and iii) mesoderm specification. Contrary to our expectation, not all cells displayed equal amplitude of Wnt target gene activation; rather, they distributed in a continuum from strong to weak responders when ranked based on the expression of the target AXIN2. Moreover, high AXIN2 did not always correspond to elevated expression of other Wnt targets, which were activated in different proportions in individual cells. The uncoupling of Wnt target gene expression was also identified in single cell transcriptomics profiling of other Wnt-responding cell types, including HEK293T, murine developing forelimbs, and human colorectal cancer. Our finding underlines the necessity to identify additional mechanisms that explain the heterogeneity of the Wnt/ß-catenin-mediated transcriptional outputs in single cells.


Asunto(s)
Vía de Señalización Wnt , beta Catenina , Ratones , Humanos , Animales , Vía de Señalización Wnt/genética , beta Catenina/genética , beta Catenina/metabolismo , Células HEK293 , Factores de Transcripción TCF/metabolismo , Expresión Génica , Mamíferos/genética
8.
Proc Natl Acad Sci U S A ; 118(34)2021 08 24.
Artículo en Inglés | MEDLINE | ID: mdl-34408016

RESUMEN

During malignant progression, epithelial cancer cells dissolve their cell-cell adhesion and gain invasive features. By virtue of its dual function, ß-catenin contributes to cadherin-mediated cell-cell adhesion, and it determines the transcriptional output of Wnt signaling: via its N terminus, it recruits the signaling coactivators Bcl9 and Pygopus, and via the C terminus, it interacts with the general transcriptional machinery. This duality confounds the simple loss-of-function analysis of Wnt signaling in cancer progression. In many cancer types including breast cancer, the functional contribution of ß-catenin's transcriptional activities, as compared to its adhesion functions, to tumor progression has remained elusive. Employing the mouse mammary tumor virus (MMTV)-PyMT mouse model of metastatic breast cancer, we compared the complete elimination of ß-catenin with the specific ablation of its signaling outputs in mammary tumor cells. Notably, the complete lack of ß-catenin resulted in massive apoptosis of mammary tumor cells. In contrast, the loss of ß-catenin's transcriptional activity resulted in a reduction of primary tumor growth, tumor invasion, and metastasis formation in vivo. These phenotypic changes were reflected by stalled cell cycle progression and diminished epithelial-mesenchymal transition (EMT) and cell migration of breast cancer cells in vitro. Transcriptome analysis revealed subsets of genes which were specifically regulated by ß-catenin's transcriptional activities upon stimulation with Wnt3a or during TGF-ß-induced EMT. Our results uncouple the signaling from the adhesion function of ß-catenin and underline the importance of Wnt/ß-catenin-dependent transcription in malignant tumor progression of breast cancer.


Asunto(s)
Adhesión Celular/fisiología , Neoplasias Mamarias Animales/metabolismo , Transducción de Señal/fisiología , Proteína Wnt3A/metabolismo , beta Catenina/metabolismo , Animales , Apoptosis , Ciclo Celular , Movimiento Celular , Transición Epitelial-Mesenquimal/efectos de los fármacos , Femenino , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Neoplasias Mamarias Animales/genética , Ratones , Ratones Transgénicos , Invasividad Neoplásica , Metástasis de la Neoplasia , Transcriptoma , Factor de Crecimiento Transformador beta/farmacología , Proteína Wnt3A/genética , beta Catenina/genética
9.
EMBO J ; 38(2)2019 01 15.
Artículo en Inglés | MEDLINE | ID: mdl-30425074

RESUMEN

During canonical Wnt signalling, the activity of nuclear ß-catenin is largely mediated by the TCF/LEF family of transcription factors. To challenge this view, we used the CRISPR/Cas9 genome editing approach to generate HEK 293T cell clones lacking all four TCF/LEF genes. By performing unbiased whole transcriptome sequencing analysis, we found that a subset of ß-catenin transcriptional targets did not require TCF/LEF factors for their regulation. Consistent with this finding, we observed in a genome-wide analysis that ß-catenin occupied specific genomic regions in the absence of TCF/LEF Finally, we revealed the existence of a transcriptional activity of ß-catenin that specifically appears when TCF/LEF factors are absent, and refer to this as ß-catenin-GHOST response. Collectively, this study uncovers a previously neglected modus operandi of ß-catenin that bypasses the TCF/LEF transcription factors.


Asunto(s)
Perfilación de la Expresión Génica/métodos , Factores de Transcripción TCF/genética , Transcripción Genética , beta Catenina/metabolismo , Sistemas CRISPR-Cas , Edición Génica , Regulación de la Expresión Génica , Células HEK293 , Humanos , Factores de Transcripción TCF/metabolismo , Secuenciación del Exoma/métodos , Vía de Señalización Wnt
10.
Mol Psychiatry ; 27(12): 4893-4904, 2022 12.
Artículo en Inglés | MEDLINE | ID: mdl-36127428

RESUMEN

Excessive fear is a hallmark of anxiety disorders, a major cause of disease burden worldwide. Substantial evidence supports a role of prefrontal cortex-amygdala circuits in the regulation of fear and anxiety, but the molecular mechanisms that regulate their activity remain poorly understood. Here, we show that downregulation of the histone methyltransferase PRDM2 in the dorsomedial prefrontal cortex enhances fear expression by modulating fear memory consolidation. We further show that Prdm2 knock-down (KD) in neurons that project from the dorsomedial prefrontal cortex to the basolateral amygdala (dmPFC-BLA) promotes increased fear expression. Prdm2 KD in the dmPFC-BLA circuit also resulted in increased expression of genes involved in synaptogenesis, suggesting that Prdm2 KD modulates consolidation of conditioned fear by modifying synaptic strength at dmPFC-BLA projection targets. Consistent with an enhanced synaptic efficacy, we found that dmPFC Prdm2 KD increased glutamatergic release probability in the BLA and increased the activity of BLA neurons in response to fear-associated cues. Together, our findings provide a new molecular mechanism for excessive fear responses, wherein PRDM2 modulates the dmPFC -BLA circuit through specific transcriptomic changes.


Asunto(s)
Amígdala del Cerebelo , Complejo Nuclear Basolateral , Amígdala del Cerebelo/fisiología , Complejo Nuclear Basolateral/fisiología , Corteza Prefrontal/metabolismo , Miedo/fisiología , Epigénesis Genética
11.
Stem Cells ; 39(8): 1107-1119, 2021 08.
Artículo en Inglés | MEDLINE | ID: mdl-33739574

RESUMEN

The Sox2 transcription factor is necessary for the long-term self-renewal of neural stem cells (NSCs). Its mechanism of action is still poorly defined. To identify molecules regulated by Sox2, and acting in mouse NSC maintenance, we transduced, into Sox2-deleted NSC, genes whose expression is strongly downregulated following Sox2 loss (Fos, Jun, Egr2), individually or in combination. Fos alone rescued long-term proliferation, as shown by in vitro cell growth and clonal analysis. Furthermore, pharmacological inhibition by T-5224 of FOS/JUN AP1 complex binding to its targets decreased cell proliferation and expression of the putative target Suppressor of cytokine signaling 3 (Socs3). Additionally, Fos requirement for efficient long-term proliferation was demonstrated by the reduction of NSC clones capable of long-term expansion following CRISPR/Cas9-mediated Fos inactivation. Previous work showed that the Socs3 gene is strongly downregulated following Sox2 deletion, and its re-expression by lentiviral transduction rescues long-term NSC proliferation. Fos appears to be an upstream regulator of Socs3, possibly together with Jun and Egr2; indeed, Sox2 re-expression in Sox2-deleted NSC progressively activates both Fos and Socs3 expression; in turn, Fos transduction activates Socs3 expression. Based on available SOX2 ChIPseq and ChIA-PET data, we propose a model whereby Sox2 is a direct activator of both Socs3 and Fos, as well as possibly Jun and Egr2; furthermore, we provide direct evidence for FOS and JUN binding on Socs3 promoter, suggesting direct transcriptional regulation. These results provide the basis for developing a model of a network of interactions, regulating critical effectors of NSC proliferation and long-term maintenance.


Asunto(s)
Células-Madre Neurales , Proteínas Proto-Oncogénicas c-fos , Factores de Transcripción SOXB1 , Animales , Proliferación Celular/genética , Autorrenovación de las Células/genética , Regulación de la Expresión Génica , Redes Reguladoras de Genes , Ratones , Células-Madre Neurales/metabolismo , Proteínas Proto-Oncogénicas c-fos/genética , Proteínas Proto-Oncogénicas c-fos/metabolismo , Factores de Transcripción SOXB1/genética , Factores de Transcripción SOXB1/metabolismo , Proteína 3 Supresora de la Señalización de Citocinas/genética , Proteína 3 Supresora de la Señalización de Citocinas/metabolismo
12.
Genes Dev ; 28(17): 1879-84, 2014 Sep 01.
Artículo en Inglés | MEDLINE | ID: mdl-25184676

RESUMEN

Bcl9 and Bcl9l (Bcl9/9l) encode Wnt signaling components that mediate the interaction between ß-catenin and Pygopus (Pygo) via two evolutionarily conserved domains, HD1 and HD2, respectively. We generated mouse strains lacking these domains to probe the ß-catenin-dependent and ß-catenin-independent roles of Bcl9/9l and Pygo during mouse development. While lens development is critically dependent on the presence of the HD1 domain, it is not affected by the lack of the HD2 domain, indicating that Bcl9/9l act in this context in a ß-catenin-independent manner. Furthermore, we uncover a new regulatory circuit in which Pax6, the master regulator of eye development, directly activates Bcl9/9l transcription.


Asunto(s)
Proteínas del Ojo/metabolismo , Regulación del Desarrollo de la Expresión Génica , Proteínas de Homeodominio/metabolismo , Cristalino/embriología , Proteínas de Neoplasias/genética , Proteínas de Neoplasias/metabolismo , Factores de Transcripción Paired Box/metabolismo , Proteínas Represoras/metabolismo , Animales , Células Cultivadas , Proteínas del Ojo/genética , Técnicas de Sustitución del Gen , Proteínas de Homeodominio/genética , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Ratones , Ratones Endogámicos C57BL , Factor de Transcripción PAX6 , Factores de Transcripción Paired Box/genética , Estructura Terciaria de Proteína/genética , Proteínas Represoras/genética , Transducción de Señal , Factores de Transcripción/metabolismo , Proteínas Wnt/metabolismo , beta Catenina/genética , beta Catenina/metabolismo
13.
Haematologica ; 106(2): 474-482, 2021 02 01.
Artículo en Inglés | MEDLINE | ID: mdl-32107331

RESUMEN

The human fetal γ-globin gene is repressed in the adult stage through complex regulatory mechanisms involving transcription factors and epigenetic modifiers. Reversing γ-globin repression, or maintaining its expression by manipulating regulatory mechanisms, has become a major clinical goal in the treatment of ß-hemoglobinopathies. Here, we identify the orphan nuclear receptor Coup-TFII (NR2F2/ARP-1) as an embryonic/fetal stage activator of γ-globin expression. We show that Coup-TFII is expressed in early erythropoiesis of yolk sac origin, together with embryonic/fetal globins. When overexpressed in adult cells (including peripheral blood cells from human healthy donors and ß039 thalassemic patients) Coup-TFII activates the embryonic/fetal globins genes, overcoming the repression imposed by the adult erythroid environment. Conversely, the knock-out of Coup-TFII increases the ß/γ+ß globin ratio. Molecular analysis indicates that Coup-TFII binds in vivo to the ß-locus and contributes to its conformation. Overall, our data identify Coup-TFII as a specific activator of the γ-globin gene.


Asunto(s)
Receptores Nucleares Huérfanos , gamma-Globinas , Factor de Transcripción COUP II/genética , Factor de Transcripción COUP II/metabolismo , Proteínas Portadoras/genética , Humanos , Regiones Promotoras Genéticas , gamma-Globinas/genética
14.
PLoS Biol ; 15(3): e2000698, 2017 03.
Artículo en Inglés | MEDLINE | ID: mdl-28350803

RESUMEN

Strategies for promoting neural regeneration are hindered by the difficulty of manipulating desired neural fates in the brain without complex genetic methods. The subventricular zone (SVZ) is the largest germinal zone of the forebrain and is responsible for the lifelong generation of interneuron subtypes and oligodendrocytes. Here, we have performed a bioinformatics analysis of the transcriptome of dorsal and lateral SVZ in early postnatal mice, including neural stem cells (NSCs) and their immediate progenies, which generate distinct neural lineages. We identified multiple signaling pathways that trigger distinct downstream transcriptional networks to regulate the diversity of neural cells originating from the SVZ. Next, we used a novel in silico genomic analysis, searchable platform-independent expression database/connectivity map (SPIED/CMAP), to generate a catalogue of small molecules that can be used to manipulate SVZ microdomain-specific lineages. Finally, we demonstrate that compounds identified in this analysis promote the generation of specific cell lineages from NSCs in vivo, during postnatal life and adulthood, as well as in regenerative contexts. This study unravels new strategies for using small bioactive molecules to direct germinal activity in the SVZ, which has therapeutic potential in neurodegenerative diseases.


Asunto(s)
Linaje de la Célula , Ventrículos Laterales/metabolismo , Transcriptoma , Animales , Simulación por Computador , Bases de Datos Genéticas , Regulación de la Expresión Génica , Redes Reguladoras de Genes , Genómica/métodos , Ratones , Regeneración Nerviosa , Células-Madre Neurales/citología , Células-Madre Neurales/metabolismo , Neurogénesis/genética , Oligodendroglía/citología , Oligodendroglía/metabolismo , Transducción de Señal , Bibliotecas de Moléculas Pequeñas
16.
Development ; 140(11): 2377-86, 2013 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-23637336

RESUMEN

Pygopus has been discovered as a fundamental Wnt signaling component in Drosophila. The mouse genome encodes two Pygopus homologs, Pygo1 and Pygo2. They serve as context-dependent ß-catenin coactivators, with Pygo2 playing the more important role. All Pygo proteins share a highly conserved plant homology domain (PHD) that allows them to bind di- and trimethylated lysine 4 of histone H3 (H3K4me2/3). Despite the structural conservation of this domain, the relevance of histone binding for the role of Pygo2 as a Wnt signaling component and as a reader of chromatin modifications remains speculative. Here we generate a knock-in mouse line, homozygous for a Pygo2 mutant defective in chromatin binding. We show that even in the absence of the potentially redundant Pygo1, Pygo2 does not require the H3K4me2/3 binding activity to sustain its function during mouse development. Indeed, during tissue homeostasis, Wnt/ß-catenin-dependent transcription is largely unaffected. However, the Pygo2-chromatin interaction is relevant in testes, where, importantly, Pygo2 binds in vivo to the chromatin in a PHD-dependent manner. Its presence on regulatory regions does not affect the transcription of nearby genes; rather, it is important for the recruitment of the histone acetyltransferase Gcn5 to chromatin, consistent with a testis-specific and Wnt-unrelated role for Pygo2 as a chromatin remodeler.


Asunto(s)
Proteínas de Drosophila/metabolismo , Regulación del Desarrollo de la Expresión Génica , Histonas/metabolismo , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Vía de Señalización Wnt , Animales , Ensamble y Desensamble de Cromatina , Proteínas de Drosophila/genética , Drosophila melanogaster/genética , Drosophila melanogaster/fisiología , Femenino , Fertilidad , Técnicas de Sustitución del Gen , Genotipo , Péptidos y Proteínas de Señalización Intracelular/genética , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Dominios y Motivos de Interacción de Proteínas , Testículo/metabolismo , Factores de Transcripción p300-CBP/metabolismo
17.
Stem Cells ; 33(1): 170-82, 2015 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-25182747

RESUMEN

The fate of neural progenitor cells (NPCs) is determined by a complex interplay of intrinsic programs and extrinsic signals, very few of which are known. ß-Catenin transduces extracellular Wnt signals, but also maintains adherens junctions integrity. Here, we identify for the first time the contribution of ß-catenin transcriptional activity as opposed to its adhesion role in the development of the cerebral cortex by combining a novel ß-catenin mutant allele with conditional inactivation approaches. Wnt/ß-catenin signaling ablation leads to premature NPC differentiation, but, in addition, to a change in progenitor cell cycle kinetics and an increase in basally dividing progenitors. Interestingly, Wnt/ß-catenin signaling affects the sequential fate switch of progenitors, leading to a shortened neurogenic period with decreased number of both deep and upper-layer neurons and later, to precocious astrogenesis. Indeed, a genome-wide analysis highlighted the premature activation of a corticogenesis differentiation program in the Wnt/ß-catenin signaling-ablated cortex. Thus, ß-catenin signaling controls the expression of a set of genes that appear to act downstream of canonical Wnt signaling to regulate the stage-specific production of appropriate progenitor numbers, neuronal subpopulations, and astroglia in the forebrain.


Asunto(s)
Corteza Cerebral/citología , Células-Madre Neurales/citología , Neuronas/citología , Vía de Señalización Wnt , beta Catenina/metabolismo , Animales , Diferenciación Celular/fisiología , Proliferación Celular/fisiología , Corteza Cerebral/metabolismo , Ratones , Ratones Endogámicos C57BL , Células-Madre Neurales/metabolismo , Neuronas/metabolismo , Transducción de Señal
18.
Stem Cells ; 32(5): 1301-12, 2014 May.
Artículo en Inglés | MEDLINE | ID: mdl-24449255

RESUMEN

In the postnatal and adult central nervous system (CNS), the subventricular zone (SVZ) of the forebrain is the main source of neural stem cells (NSCs) that generate olfactory neurons and oligodendrocytes (OLs), the myelinating cells of the CNS. Here, we provide evidence of a primary role for canonical Wnt/ß-catenin signaling in regulating NSC fate along neuronal and oligodendroglial lineages in the postnatal SVZ. Our findings demonstrate that glutamatergic neuronal precursors (NPs) and oligodendrocyte precursors (OPs) are derived strictly from the dorsal SVZ (dSVZ) microdomain under the control of Wnt/ß-catenin, whereas GABAergic NPs are derived mainly from the lateral SVZ (lSVZ) microdomain independent of Wnt/ß-catenin. Transcript analysis of microdissected SVZ microdomains revealed that canonical Wnt/ß-catenin signaling was more pronounced in the dSVZ microdomain. This was confirmed using the ß-catenin-activated Wnt-reporter mouse and by pharmacological stimulation of Wnt/ß-catenin by infusion of the specific glycogen synthase kinase 3ß inhibitor, AR-A014418, which profoundly increased the generation of cycling cells. In vivo genetic/pharmacological stimulation or inhibition of Wnt/ß-catenin, respectively, increased and decreased the differentiation of dSVZ-NSCs into glutamatergic NPs, and had a converse effect on GABAergic NPs. Activation of Wnt/ß-catenin dramatically stimulated the generation of OPs, but its inhibition had no effect, indicating other factors act in concert with Wnt/ß-catenin to fine tune oligodendrogliogenesis in the postnatal dSVZ. These results demonstrate a role for Wnt/ß-catenin signaling within the dorsal microdomain of the postnatal SVZ, in regulating the genesis of glutamatergic neurons and OLs.


Asunto(s)
Ventrículos Laterales/metabolismo , Células-Madre Neurales/metabolismo , Neuronas/metabolismo , Oligodendroglía/metabolismo , Proteína Wnt3/metabolismo , beta Catenina/metabolismo , Animales , Western Blotting , Proliferación Celular/efectos de los fármacos , Ácido Glutámico/metabolismo , Glucógeno Sintasa Quinasa 3/antagonistas & inhibidores , Proteínas Fluorescentes Verdes/genética , Proteínas Fluorescentes Verdes/metabolismo , Ventrículos Laterales/citología , Ratones Transgénicos , Microscopía Confocal , Células-Madre Neurales/citología , Neuronas/citología , Oligodendroglía/citología , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Transducción de Señal/efectos de los fármacos , Transducción de Señal/genética , Tiazoles/farmacología , Urea/análogos & derivados , Urea/farmacología , Proteína Wnt3/genética , beta Catenina/genética
19.
Sci Rep ; 14(1): 10287, 2024 05 04.
Artículo en Inglés | MEDLINE | ID: mdl-38704454

RESUMEN

The identification of regulatory networks contributing to fetal/adult gene expression switches is a major challenge in developmental biology and key to understand the aberrant proliferation of cancer cells, which often reactivate fetal oncogenes. One key example is represented by the developmental gene LIN28B, whose aberrant reactivation in adult tissues promotes tumor initiation and progression. Despite the prominent role of LIN28B in development and cancer, the mechanisms of its transcriptional regulation are largely unknown. Here, by using quantitative RT-PCR and single cell RNA sequencing data, we show that in erythropoiesis the expression of the transcription factor SOX6 matched a sharp decline of LIN28B mRNA during human embryo/fetal to adult globin switching. SOX6 overexpression repressed LIN28B not only in a panel of fetal-like erythroid cells (K562, HEL and HUDEP1; ≈92% p < 0.0001, 54% p = 0.0009 and ≈60% p < 0.0001 reduction, respectively), but also in hepatoblastoma HepG2 and neuroblastoma SH-SY5H cells (≈99% p < 0.0001 and ≈59% p < 0.0001 reduction, respectively). SOX6-mediated repression caused downregulation of the LIN28B/Let-7 targets, including MYC and IGF2BP1, and rapidly blocks cell proliferation. Mechanistically, Lin28B repression is accompanied by SOX6 physical binding within its locus, suggesting a direct mechanism of LIN28B downregulation that might contribute to the fetal/adult erythropoietic transition and restrict cancer proliferation.


Asunto(s)
Proteínas de Unión al ARN , Factores de Transcripción SOXD , Humanos , Factores de Transcripción SOXD/genética , Factores de Transcripción SOXD/metabolismo , Proteínas de Unión al ARN/metabolismo , Proteínas de Unión al ARN/genética , Línea Celular Tumoral , Regulación del Desarrollo de la Expresión Génica , Eritropoyesis/genética , MicroARNs/genética , MicroARNs/metabolismo , Células Hep G2 , Células K562 , Regulación Neoplásica de la Expresión Génica , Células Eritroides/metabolismo
20.
Blood ; 117(13): 3669-79, 2011 Mar 31.
Artículo en Inglés | MEDLINE | ID: mdl-21263153

RESUMEN

Sox6 belongs to the Sry (sex-determining region Y)-related high-mobility-group-box family of transcription factors, which control cell-fate specification of many cell types. Here, we explored the role of Sox6 in human erythropoiesis by its overexpression both in the erythroleukemic K562 cell line and in primary erythroid cultures from human cord blood CD34+ cells. Sox6 induced significant erythroid differentiation in both models. K562 cells underwent hemoglobinization and, despite their leukemic origin, died within 9 days after transduction; primary erythroid cultures accelerated their kinetics of erythroid maturation and increased the number of cells that reached the final enucleation step. Searching for direct Sox6 targets, we found SOCS3 (suppressor of cytokine signaling-3), a known mediator of cytokine response. Sox6 was bound in vitro and in vivo to an evolutionarily conserved regulatory SOCS3 element, which induced transcriptional activation. SOCS3 overexpression in K562 cells and in primary erythroid cells recapitulated the growth inhibition induced by Sox6, which demonstrates that SOCS3 is a relevant Sox6 effector.


Asunto(s)
Células Precursoras Eritroides/fisiología , Eritropoyesis/genética , Factores de Transcripción SOXD/fisiología , Animales , Antígenos CD34/metabolismo , Diferenciación Celular/genética , Procesos de Crecimiento Celular/genética , Células Cultivadas , Ensayo de Unidades Formadoras de Colonias , Células Precursoras Eritroides/metabolismo , Eritropoyesis/fisiología , Regulación de la Expresión Génica/fisiología , Humanos , Células K562 , Ratones , Modelos Biológicos , Factores de Transcripción SOXD/genética , Factores de Transcripción SOXD/metabolismo , Proteína 3 Supresora de la Señalización de Citocinas , Proteínas Supresoras de la Señalización de Citocinas/genética , Proteínas Supresoras de la Señalización de Citocinas/metabolismo , Proteínas Supresoras de la Señalización de Citocinas/fisiología , Transfección
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA