Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 16 de 16
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Cell ; 187(10): 2536-2556.e30, 2024 May 09.
Artículo en Inglés | MEDLINE | ID: mdl-38653237

RESUMEN

Cysteine-focused chemical proteomic platforms have accelerated the clinical development of covalent inhibitors for a wide range of targets in cancer. However, how different oncogenic contexts influence cysteine targeting remains unknown. To address this question, we have developed "DrugMap," an atlas of cysteine ligandability compiled across 416 cancer cell lines. We unexpectedly find that cysteine ligandability varies across cancer cell lines, and we attribute this to differences in cellular redox states, protein conformational changes, and genetic mutations. Leveraging these findings, we identify actionable cysteines in NF-κB1 and SOX10 and develop corresponding covalent ligands that block the activity of these transcription factors. We demonstrate that the NF-κB1 probe blocks DNA binding, whereas the SOX10 ligand increases SOX10-SOX10 interactions and disrupts melanoma transcriptional signaling. Our findings reveal heterogeneity in cysteine ligandability across cancers, pinpoint cell-intrinsic features driving cysteine targeting, and illustrate the use of covalent probes to disrupt oncogenic transcription-factor activity.


Asunto(s)
Cisteína , Neoplasias , Animales , Humanos , Ratones , Línea Celular Tumoral , Cisteína/metabolismo , Cisteína/química , Ligandos , Melanoma/metabolismo , Neoplasias/tratamiento farmacológico , Neoplasias/metabolismo , FN-kappa B/química , FN-kappa B/metabolismo , Oxidación-Reducción , Transducción de Señal , Factores de Transcripción SOXE/química , Factores de Transcripción SOXE/metabolismo
2.
Dermatol Online J ; 26(1)2020 Jan 15.
Artículo en Inglés | MEDLINE | ID: mdl-32155034

RESUMEN

Sox10 immunostaining is used for the diagnosis and margin evaluation of melanocytic lesions. Sox10 was initially thought not to stain fibrohistiocytic processes. Consequently, it was believed to reliably distinguish desmoplastic melanoma from scar. However, recent data from formalin sections suggest Sox10 is less specific than previously thought. In this report, we demonstrate that Sox10-stained Mohs sections commonly show strong, fractional staining of scar. When using Sox10 with frozen section immunohistochemistry, Mohs practitioners should recognize the potential of this marker to stain scar to avoid overdiagnosis of desmoplastic melanoma.


Asunto(s)
Cicatriz/patología , Colorantes/efectos adversos , Melanoma/diagnóstico , Factores de Transcripción SOXE/metabolismo , Neoplasias Cutáneas/diagnóstico , Secciones por Congelación , Humanos , Inmunohistoquímica , Melanoma/cirugía , Cirugía de Mohs , Factores de Transcripción SOXE/química , Neoplasias Cutáneas/cirugía , Coloración y Etiquetado , Melanoma Cutáneo Maligno
3.
Nucleic Acids Res ; 47(13): 6917-6931, 2019 07 26.
Artículo en Inglés | MEDLINE | ID: mdl-31194875

RESUMEN

SOX8, SOX9 and SOX10 compose the SOXE transcription factor group. They govern cell fate and differentiation in many lineages, and mutations impairing their activity cause severe diseases, including campomelic dysplasia (SOX9), sex determination disorders (SOX8 and SOX9) and Waardenburg-Shah syndrome (SOX10). However, incomplete knowledge of their modes of action limits disease understanding. We here uncover that the proteins share a bipartite transactivation mechanism, whereby a transactivation domain in the middle of the proteins (TAM) synergizes with a C-terminal one (TAC). TAM comprises amphipathic α-helices predicted to form a protein-binding pocket and overlapping with minimal transactivation motifs (9-aa-TAD) described in many transcription factors. One 9-aa-TAD sequence includes an evolutionarily conserved and functionally required EΦ[D/E]QYΦ motif. SOXF proteins (SOX7, SOX17 and SOX18) contain an identical motif, suggesting evolution from a common ancestor already harboring this motif, whereas TAC and other transactivating SOX proteins feature only remotely related motifs. Missense variants in this SOXE/SOXF-specific motif are rare in control individuals, but have been detected in cancers, supporting its importance in development and physiology. By deepening understanding of mechanisms underlying the central transactivation function of SOXE proteins, these findings should help further decipher molecular networks essential for development and health and dysregulated in diseases.


Asunto(s)
Factor de Transcripción SOX9/química , Factores de Transcripción SOXE/química , Activación Transcripcional/fisiología , Secuencias de Aminoácidos , Línea Celular , Secuencia Conservada , Evolución Molecular , Humanos , Mutación Missense , Dominios Proteicos , Proteínas Recombinantes de Fusión/química , Alineación de Secuencia , Homología de Secuencia de Aminoácido
4.
PLoS One ; 13(1): e0190834, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-29315345

RESUMEN

The transcription factor SOX10 plays an important role in vertebrate neural crest development, including the establishment and maintenance of the melanocyte lineage. SOX10 is also highly expressed in melanoma tumors, and SOX10 expression increases with tumor progression. The suppression of SOX10 in melanoma cells activates TGF-ß signaling and can promote resistance to BRAF and MEK inhibitors. Since resistance to BRAF/MEK inhibitors is seen in the majority of melanoma patients, there is an immediate need to assess the underlying biology that mediates resistance and to identify new targets for combinatorial therapeutic approaches. Previously, we demonstrated that SOX10 protein is required for tumor initiation, maintenance and survival. Here, we present data that support phosphorylation as a mechanism employed by melanoma cells to tightly regulate SOX10 expression. Mass spectrometry identified eight phosphorylation sites contained within SOX10, three of which (S24, S45 and T240) were selected for further analysis based on their location within predicted MAPK/CDK binding motifs. SOX10 mutations were generated at these phosphorylation sites to assess their impact on SOX10 protein function in melanoma cells, including transcriptional activation on target promoters, subcellular localization, and stability. These data further our understanding of SOX10 protein regulation and provide critical information for identification of molecular pathways that modulate SOX10 protein levels in melanoma, with the ultimate goal of discovering novel targets for more effective combinatorial therapeutic approaches for melanoma patients.


Asunto(s)
Melanoma/metabolismo , Factores de Transcripción SOXE/metabolismo , Secuencia de Aminoácidos , Línea Celular Tumoral , Cromatografía Liquida , Humanos , Fosforilación , Procesamiento Proteico-Postraduccional , Factores de Transcripción SOXE/química , Espectrometría de Masas en Tándem
5.
Sci Rep ; 7: 41513, 2017 01 27.
Artículo en Inglés | MEDLINE | ID: mdl-28128317

RESUMEN

Waardenburg syndrome type 4 (WS4) or Waardenburg-Shah syndrome is a rare genetic disorder with a prevalence of <1/1,000,000 and characterized by the association of congenital sensorineural hearing loss, pigmentary abnormalities, and intestinal aganglionosis. There are three types of WS4 (WS4A-C) caused by mutations in endothelin receptor type B, endothelin 3, and SRY-box 10 (SOX10), respectively. This study investigated a genetic mutation in a Chinese family with one WS4 patient in order to improve genetic counselling. Genomic DNA was extracted, and mutation analysis of the three WS4 related genes was performed using Sanger sequencing. We detected a de novo heterozygous deletion mutation [c.1333delT (p.Ser445Glnfs*57)] in SOX10 in the patient; however, this mutation was absent in the unaffected parents and 40 ethnicity matched healthy controls. Subsequent phylogenetic analysis and three-dimensional modelling of the SOX10 protein confirmed that the c.1333delT heterozygous mutation was pathogenic, indicating that this mutation might constitute a candidate disease-causing mutation.


Asunto(s)
Pueblo Asiatico/genética , Enfermedad de Hirschsprung/genética , Mutación/genética , Factores de Transcripción SOXE/genética , Síndrome de Waardenburg/genética , Secuencia de Bases , Familia , Haplotipos/genética , Humanos , Lactante , Masculino , Paternidad , Polimorfismo de Nucleótido Simple/genética , Factores de Transcripción SOXE/química
6.
Semin Cell Dev Biol ; 63: 13-22, 2017 03.
Artículo en Inglés | MEDLINE | ID: mdl-27481580

RESUMEN

In mammals, sex determination defines the differentiation of the bipotential genital ridge into either testes or ovaries. Sry, the mammalian Y-chromosomal testis-determining gene, is a master regulator of male sex determination. It acts to switch the undifferentiated genital ridge towards testis development, triggering the adoption of a male fate. Sry initiates a cascade of gene networks through the direct regulation of Sox9 expression and promotes supporting cell differentiation, Leydig cell specification, vasculature formation and testis cord development. In the absence of Sry, alternative genetic cascades, including female sex-determining genes RSPO1, Wnt4/ß-catenin and Foxl2, are involved in the formation of female genitalia and the maintenance of female ovarian development. The mutual antagonisms between male and female sex-determining pathways are crucial in not just the initiation but also the maintenance of the somatic sex of the gonad throughout the organism's lifetime. Any imbalances in above sex-determining genes can cause disorders of sex development in humans and mice. In this review, we provide a detailed summary of the expression profiles, biochemical properties and developmental functions of Sry and SoxE genes in embryonic testis development and adult gonadal development. We also briefly summarize the dedicate balances between male and female sex-determining genes in mammalian sex development, with particular highlights on the molecular actions of Sry and Sox9 transcription factors.


Asunto(s)
Gónadas/metabolismo , Mamíferos/genética , Factores de Transcripción SOXE/genética , Procesos de Determinación del Sexo/genética , Proteína de la Región Y Determinante del Sexo/genética , Animales , Modelos Biológicos , Factores de Transcripción SOXE/química , Proteína de la Región Y Determinante del Sexo/química
7.
BMC Genomics ; 17(1): 887, 2016 11 07.
Artículo en Inglés | MEDLINE | ID: mdl-27821050

RESUMEN

BACKGROUND: The transcription factor SOX10 is essential for all stages of Schwann cell development including myelination. SOX10 cooperates with other transcription factors to activate the expression of key myelin genes in Schwann cells and is therefore a context-dependent, pro-myelination transcription factor. As such, the identification of genes regulated by SOX10 will provide insight into Schwann cell biology and related diseases. While genome-wide studies have successfully revealed SOX10 target genes, these efforts mainly focused on myelinating stages of Schwann cell development. We propose that less-biased approaches will reveal novel functions of SOX10 outside of myelination. RESULTS: We developed a stringent, computational-based screen for genome-wide identification of SOX10 response elements. Experimental validation of a pilot set of predicted binding sites in multiple systems revealed that SOX10 directly regulates a previously unreported alternative promoter at SOX6, which encodes a transcription factor that inhibits glial cell differentiation. We further explored the utility of our computational approach by combining it with DNase-seq analysis in cultured Schwann cells and previously published SOX10 ChIP-seq data from rat sciatic nerve. Remarkably, this analysis enriched for genomic segments that map to loci involved in the negative regulation of gliogenesis including SOX5, SOX6, NOTCH1, HMGA2, HES1, MYCN, ID4, and ID2. Functional studies in Schwann cells revealed that: (1) all eight loci are expressed prior to myelination and down-regulated subsequent to myelination; (2) seven of the eight loci harbor validated SOX10 binding sites; and (3) seven of the eight loci are down-regulated upon repressing SOX10 function. CONCLUSIONS: Our computational strategy revealed a putative novel function for SOX10 in Schwann cells, which suggests a model where SOX10 activates the expression of genes that inhibit myelination during non-myelinating stages of Schwann cell development. Importantly, the computational and functional datasets we present here will be valuable for the study of transcriptional regulation, SOX protein function, and glial cell biology.


Asunto(s)
Diferenciación Celular , Neuroglía/citología , Neuroglía/metabolismo , Factores de Transcripción SOXE/metabolismo , Secuencia de Bases , Diferenciación Celular/genética , Secuencia de Consenso , Secuencia Conservada , Exones , Genómica/métodos , Secuenciación de Nucleótidos de Alto Rendimiento , Regiones Promotoras Genéticas , Elementos Reguladores de la Transcripción , Elementos de Respuesta , Factores de Transcripción SOXE/química , Factores de Transcripción SOXE/genética , Células de Schwann/metabolismo
8.
Hum Mol Genet ; 24(19): 5433-50, 2015 Oct 01.
Artículo en Inglés | MEDLINE | ID: mdl-26206884

RESUMEN

SOX10 is required for melanocyte development and maintenance, and has been linked to melanoma initiation and progression. However, the molecular mechanisms by which SOX10 guides the appropriate gene expression programs necessary to promote the melanocyte lineage are not fully understood. Here we employ genetic and epigenomic analysis approaches to uncover novel genomic targets and previously unappreciated molecular roles of SOX10 in melanocytes. Through global analysis of SOX10-binding sites and epigenetic characteristics of chromatin states, we uncover an extensive catalog of SOX10 targets genome-wide. Our findings reveal that SOX10 predominantly engages 'open' chromatin regions and binds to distal regulatory elements, including novel and previously known melanocyte enhancers. Integrated chromatin occupancy and transcriptome analysis suggest a role for SOX10 in both transcriptional activation and repression to regulate functionally distinct classes of genes. We demonstrate that distinct epigenetic signatures and cis-regulatory sequence motifs predicted to bind putative co-regulatory transcription factors define SOX10-activated and SOX10-repressed target genes. Collectively, these findings uncover a central role of SOX10 as a global regulator of gene expression in the melanocyte lineage by targeting diverse regulatory pathways.


Asunto(s)
Perfilación de la Expresión Génica/métodos , Redes Reguladoras de Genes , Melanocitos/metabolismo , Análisis de Secuencia por Matrices de Oligonucleótidos/métodos , Factores de Transcripción SOXE/metabolismo , Animales , Sitios de Unión , Línea Celular , Cromatina/genética , Cromatina/metabolismo , Epigenómica/métodos , Melanocitos/citología , Ratones , Factores de Transcripción SOXE/química , Factores de Transcripción SOXE/genética
9.
Sci Rep ; 5: 10398, 2015 May 27.
Artículo en Inglés | MEDLINE | ID: mdl-26013289

RESUMEN

The SOXE transcription factors SOX8, SOX9 and SOX10 are master regulators of mammalian development directing sex determination, gliogenesis, pancreas specification and neural crest development. We identified a set of palindromic SOX binding sites specifically enriched in regulatory regions of melanoma cells. SOXE proteins homodimerize on these sequences with high cooperativity. In contrast to other transcription factor dimers, which are typically rigidly spaced, SOXE group proteins can bind cooperatively at a wide range of dimer spacings. Using truncated forms of SOXE proteins, we show that a single dimerization (DIM) domain, that precedes the DNA binding high mobility group (HMG) domain, is sufficient for dimer formation, suggesting that DIM : HMG rather than DIM:DIM interactions mediate the dimerization. All SOXE members can also heterodimerize in this fashion, whereas SOXE heterodimers with SOX2, SOX4, SOX6 and SOX18 are not supported. We propose a structural model where SOXE-specific intramolecular DIM:HMG interactions are allosterically communicated to the HMG of juxtaposed molecules. Collectively, SOXE factors evolved a unique mode to combinatorially regulate their target genes that relies on a multifaceted interplay between the HMG and DIM domains. This property potentially extends further the diversity of target genes and cell-specific functions that are regulated by SOXE proteins.


Asunto(s)
Factores de Transcripción SOXE/metabolismo , Secuencia de Aminoácidos , Sitios de Unión , Línea Celular Tumoral , Dicroismo Circular , ADN/química , ADN/metabolismo , Dimerización , Ensayo de Cambio de Movilidad Electroforética , Dominios HMG-Box , Humanos , Datos de Secuencia Molecular , Mutagénesis , Motivos de Nucleótidos , Unión Proteica , Proteínas Recombinantes/biosíntesis , Proteínas Recombinantes/química , Proteínas Recombinantes/aislamiento & purificación , Factor de Transcripción SOX9/química , Factor de Transcripción SOX9/metabolismo , Factores de Transcripción SOXE/química , Factores de Transcripción SOXE/genética , Alineación de Secuencia
10.
Clin Genet ; 88(4): 352-9, 2015 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-25256313

RESUMEN

Ninety genes have been identified to date that are involved in non-syndromic hearing loss, and more than 300 different forms of syndromic hearing impairment have been described. Mutations in SOX10, one of the genes contributing to syndromic hearing loss, induce a large range of phenotypes, including several subtypes of Waardenburg syndrome and Kallmann syndrome with deafness. In addition, rare mutations have been identified in patients with isolated signs of these diseases. We used the recent characterization of temporal bone imaging aspects in patients with SOX10 mutations to identify possible patients with isolated hearing loss due to SOX10 mutation. We selected 21 patients with isolated deafness and temporal bone morphological defects for mutational screening. We identified two SOX10 mutations and found that both resulted in a non-functional protein in vitro. Re-evaluation of the two affected patients showed that both had previously undiagnosed olfactory defects. Diagnosis of anosmia or hyposmia in young children is challenging, and particularly in the absence of magnetic resonance imaging (MRI), SOX10 mutations can mimic non-syndromic hearing impairment. MRI should complete temporal bones computed tomographic scan in the management of congenital deafness as it can detect brain anomalies, cochlear nerve defects, and olfactory bulb malformation in addition to inner ear malformations.


Asunto(s)
Pérdida Auditiva/genética , Mutación , Factores de Transcripción SOXE/genética , Hueso Temporal/patología , Adolescente , Adulto , Anciano , Secuencia de Aminoácidos , Secuencia de Bases , Niño , Análisis Mutacional de ADN , Diagnóstico Diferencial , Oído Interno/anomalías , Femenino , Estudios de Asociación Genética , Pérdida Auditiva Sensorineural/genética , Humanos , Imagen por Resonancia Magnética , Masculino , Datos de Secuencia Molecular , Fenotipo , Factores de Transcripción SOXE/química , Síndrome de Waardenburg/genética
11.
Dev Biol ; 382(1): 330-43, 2013 Oct 01.
Artículo en Inglés | MEDLINE | ID: mdl-23644063

RESUMEN

The SOX10 transcription factor is a characteristic marker for migratory multipotent neural crest (NC) progenitors as well as several of their differentiated derivatives. The involvement of SOX10 in Waardenburg-Hirschsprung disease (pigmentation defects, deafness and intestinal aganglionosis) and studies of mutant animal models have contributed significantly to the understanding of its function in neural crest cells (NCC) in general and in the melanocytes and enteric nervous system (ENS) in particular. Cell-based studies have further demonstrated the important roles of this transcription factor in maintaining the NC progenitor cell number and in determining glial cell fate. Phenotypic variability observed among patients presenting with SOX10 mutations is in agreement with molecular genetics and animal model studies, which revealed that SOX10 cooperates with different partner factors; a number of genetic modifiers of SOX10 have been identified. This study reviews the expression, regulation, and function of SOX10 in normal development of the ENS and in disease conditions, as well as the genetic and molecular interactions of SOX10 with other ENS genes/factors. We also discuss future research areas. Further understanding of SOX10 function will benefit from genomic and cell biological studies that integrate the cell-intrinsic molecular mechanisms and the interactions of the enteric NCC with the niche environment.


Asunto(s)
Sistema Nervioso Entérico/embriología , Sistema Nervioso Entérico/metabolismo , Factores de Transcripción SOXE/metabolismo , Animales , Secuencia de Bases , Regulación del Desarrollo de la Expresión Génica , Humanos , Modelos Animales , Datos de Secuencia Molecular , Mutación/genética , Cresta Neural/citología , Factores de Transcripción SOXE/química , Factores de Transcripción SOXE/genética
12.
J Genet ; 89(2): 183-92, 2010 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-20861569

RESUMEN

The Sox9 gene attracts a lot of attention because of its connection with gonadal development and differentiation. However, Sox8, belonging to the same subgroup SoxE, has rarely been studied. To investigate the function as well as the evolutionary origin of SOXE subgroup, we amplified the genomic DNA of Paramisgurnus dabryanu using a pair of degenerate primers. Using rapid amplification of the cDNA ends (RACE), it was discovered that P. dabryanu has two duplicates: Sox8a and Sox8b. Each has an intron of different length in the conserved HMG-box region. The overall sequence similarity of the deduced amino acid of PdSox8a and PdSox8b was 46.26%, and only two amino acids changed in the HMG-box. This is the first evidence showing that there are two distinct duplications of Sox8 genes in Cypriniformes. Southern blot analysis showed only one hybrid band, with lengths 7.4 or 9.2 kb. Both semi-quantitative RT-PCR and real-time quantitative PCR assay displayed that both PdSox8a and PdSox8b are downregulated during early embryonic development. In adult tissues, the two Sox8 genes expressed ubiquitously, and expression levels are particularly high in the gonads and brain. In gonads, both PdSox8a and PdSox8b are expressed at a higher level in the tesis than in the ovary. PdSox8a and PdSox8b may have functional overlaps and are essential for the neuronal development and differentiation of gonads.


Asunto(s)
Factores de Transcripción SOXE/genética , Factores de Transcripción SOXE/metabolismo , Tilapia/genética , Secuencia de Aminoácidos , Animales , Secuencia de Bases , Clonación Molecular , Desarrollo Embrionario/genética , Femenino , Expresión Génica , Regulación del Desarrollo de la Expresión Génica , Masculino , Factores de Transcripción SOXE/química , Alineación de Secuencia , Análisis de Secuencia de ADN , Homología de Secuencia de Aminoácido , Tilapia/embriología
13.
Hum Mol Genet ; 19(12): 2409-20, 2010 Jun 15.
Artículo en Inglés | MEDLINE | ID: mdl-20308050

RESUMEN

The HMG-domain containing transcription factor Sox10 is essential for neural crest (NC) development and for oligodendrocyte differentiation. Heterozygous SOX10 mutations in humans lead to corresponding defects in several NC-derived lineages and to leukodystrophies. Disease phenotypes range from Waardenburg syndrome and Waardenburg-Hirschsprung disease to Peripheral demyelinating neuropathy, Central dysmyelination, Waardenburg syndrome and Hirschsprung disease (PCWH). The phenotypic variability can partly be explained by the action of modifier genes, but is also influenced by the mutation that leads to haploinsufficiency in some and to mutant SOX10 proteins with altered properties in other cases. Here, we used in ovo electroporation in the developing neural tube of chicken to determine which regions and properties of SOX10 are required for early NC development. We found a strict reliance on the DNA-binding activity and the presence of the C-terminal transactivation domain and a lesser influence of the dimerization function and a conserved domain in the center of the protein. Intriguingly, dominant-negative effects on early NC development were mostly observed for truncated SOX10 proteins whose production in patients is probably prevented by nonsense-mediated decay. In contrast, mutant SOX10 proteins that occur in patients were usually inactive. Any dominant negative activity which some of these mutants undoubtedly possess must, therefore, be restricted to single NC-derived cell lineages or oligodendrocytes at later times. This contributes to the phenotypic variability of human SOX10 mutations.


Asunto(s)
Cresta Neural/anomalías , Defectos del Tubo Neural/genética , Tubo Neural/metabolismo , Factores de Transcripción SOXE/genética , Animales , Apoptosis/genética , Embrión de Pollo , Enfermedades Desmielinizantes/genética , Electroporación , Enfermedad de Hirschsprung/genética , Humanos , Mutación , Tubo Neural/crecimiento & desarrollo , Tubo Neural/patología , Defectos del Tubo Neural/patología , Estructura Terciaria de Proteína , Factores de Transcripción SOXE/química , Factores de Transcripción SOXE/metabolismo , Médula Espinal/embriología , Médula Espinal/metabolismo , Síndrome de Waardenburg/genética
14.
Int J Biochem Cell Biol ; 42(3): 433-6, 2010 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-19647095

RESUMEN

The group E SOX proteins consist of SOX8, SOX9 and SOX10. These transcription factors contain, besides a DNA-binding HMG domain and a transactivation domain, a DNA-dependent dimerization domain, unique among SOX proteins. Among these three SOX E proteins, which are all expressed during mammalian testis development, SOX9 stands out in importance. It is SOX9 that becomes activated by SRY in pre-Sertoli cells, executing SRY's role as a testis-determining factor by inducing Sertoli cell and testis cord differentiation. However, Sox9 is dispensable during subsequent embryonic and postnatal testis development, since ablation of Sox9 at embryonic day 14.0, after the sex determination stage, only leads to late-onset sterility at about 5 months. A similar late male sterility phenotype occurs in constitutive Sox8 null mutants. In the combined absence of Sox9 and Sox8, primary male infertility evolves, revealing functional redundancy. Loss of Sox10 has no effect on testis development.


Asunto(s)
Mamíferos/embriología , Mamíferos/genética , Factores de Transcripción SOXE/genética , Testículo/embriología , Animales , Regulación del Desarrollo de la Expresión Génica , Humanos , Masculino , Factor de Transcripción SOX9/química , Factor de Transcripción SOX9/genética , Factor de Transcripción SOX9/metabolismo , Factores de Transcripción SOXE/química , Factores de Transcripción SOXE/metabolismo , Proteína de la Región Y Determinante del Sexo/genética , Proteína de la Región Y Determinante del Sexo/metabolismo , Testículo/metabolismo
15.
J Neurochem ; 112(3): 744-54, 2010 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-19922439

RESUMEN

The high-mobility group domain transcription factor Sox10 is believed to influence myelination in Schwann cells by directly activating myelin genes and by inducing Krox20 as a pivotal regulator of peripheral myelination. Krox20 induction at this stage is thought to be mediated by the myelinating Schwann cell element 35 kb downstream of the Krox20 transcriptional start site and requires cooperation with Oct6. Here, we prove for the first time in vivo that Schwann cell-specific Krox20 expression indeed depends on Sox10. We also provide evidence that Sox10 functions through multiple, mostly monomeric binding sites in the myelinating Schwann cell element in a manner that should render the enhancer exquisitely sensitive to Sox10 levels. Synergistic activation of the enhancer by Sox10 and Oct6 furthermore does not involve cooperative binding to closely spaced binding sites in defined composite elements. Nevertheless, the POU domain of Oct6 and the high-mobility group domain of Sox10 as the two DNA-binding domains were both essential indicating that each transcription factor has to bind independently to DNA. Whereas the POU domain was the only important region of Oct6, two further Sox10 domains were required for synergistic Krox20 activation. These were the carboxyterminal transactivation domain and the conserved K2 domain in the central portion of Sox10. All required regions are conserved in several closely related POU and Sox proteins thus explaining why Oct6 and Sox10 can be replaced by their relatives during Krox20 induction in myelinating Schwann cells.


Asunto(s)
Proteína 2 de la Respuesta de Crecimiento Precoz/metabolismo , Regulación del Desarrollo de la Expresión Génica/fisiología , Factores de Transcripción SOXE/metabolismo , Células de Schwann/metabolismo , Animales , Sitios de Unión/genética , Línea Celular Transformada , Proteína 2 de la Respuesta de Crecimiento Precoz/genética , Ensayo de Cambio de Movilidad Electroforética/métodos , Embrión de Mamíferos , Regulación del Desarrollo de la Expresión Génica/genética , Humanos , Ratones , Ratones Transgénicos , Mutación/genética , Proteínas de Transporte de Catión Orgánico/genética , Proteínas de Transporte de Catión Orgánico/metabolismo , Unión Proteica/genética , Estructura Terciaria de Proteína/genética , Factores de Transcripción SOXE/química , Factores de Transcripción SOXE/genética , Transfección/métodos
16.
Mol Biol Cell ; 19(4): 1575-86, 2008 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-18272785

RESUMEN

Sry-box (Sox)8, Sox9, and Sox10 are all strongly expressed in the neural crest. Here, we studied the influence of these closely related transcription factors on the developing adrenal medulla as one prominent neural crest derivative. Whereas Sox9 was not expressed, both Sox8 and Sox10 occurred widely in neural crest cells migrating to the adrenal gland and in the gland itself, and they were down-regulated in cells expressing catecholaminergic traits. Sox10-deficient mice lacked an adrenal medulla. The adrenal anlage was never colonized by neural crest cells, which failed to specify properly at the dorsal aorta and died apoptotically during migration. Furthermore, mutant neural crest cells did not express Sox8. Strong adrenal phenotypes were also observed when the Sox10 dimerization domain was inactivated or when a transactivation domain in the central portion was deleted. Sox8 in contrast had only minimal influence on adrenal gland development. Phenotypic consequences became only visible in Sox8-deficient mice upon additional deletion of one Sox10 allele. Replacement of Sox10 by Sox8, however, led to significant rescue of the adrenal medulla, indicating that functional differences between the two related Sox proteins contribute less to the different adrenal phenotypes of the null mutants than dependence of Sox8 expression on Sox10.


Asunto(s)
Glándulas Suprarrenales/embriología , Glándulas Suprarrenales/metabolismo , Factor de Transcripción SOX9/metabolismo , Factores de Transcripción SOXE/metabolismo , Glándulas Suprarrenales/inervación , Médula Suprarrenal/embriología , Médula Suprarrenal/metabolismo , Alelos , Animales , Células Cromafines/citología , Células Cromafines/metabolismo , Secuencia Conservada , Ganglios Simpáticos/embriología , Ganglios Simpáticos/metabolismo , Regulación del Desarrollo de la Expresión Génica , Proteínas de Homeodominio/metabolismo , Ratones , Ratones Endogámicos C3H , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Transgénicos , Cresta Neural/embriología , Cresta Neural/metabolismo , Estructura Terciaria de Proteína , Factor de Transcripción SOX9/química , Factor de Transcripción SOX9/genética , Factores de Transcripción SOXE/química , Factores de Transcripción SOXE/deficiencia , Factores de Transcripción SOXE/genética , Factores de Transcripción/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...