Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 26
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Cell Rep ; 37(6): 109975, 2021 11 09.
Artículo en Inglés | MEDLINE | ID: mdl-34758317

RESUMEN

Dopamine (DA) neurons in the ventral tier of the substantia nigra pars compacta (SNc) degenerate prominently in Parkinson's disease, while those in the dorsal tier are relatively spared. Defining the molecular, functional, and developmental characteristics of each SNc tier is crucial to understand their distinct susceptibility. We demonstrate that Sox6 expression distinguishes ventrally and dorsally biased DA neuron populations in the SNc. The Sox6+ population in the ventral SNc includes an Aldh1a1+ subset and is enriched in gene pathways that underpin vulnerability. Sox6+ neurons project to the dorsal striatum and show activity correlated with acceleration. Sox6- neurons project to the medial, ventral, and caudal striatum and respond to rewards. Moreover, we show that this adult division is encoded early in development. Overall, our work demonstrates a dual origin of the SNc that results in DA neuron cohorts with distinct molecular profiles, projections, and functions.


Asunto(s)
Cuerpo Estriado/patología , Neuronas Dopaminérgicas/patología , Regulación del Desarrollo de la Expresión Génica , Enfermedad de Parkinson/patología , Factores de Transcripción SOXD/metabolismo , Factores de Transcripción SOXD/fisiología , Sustancia Negra/patología , Anciano , Anciano de 80 o más Años , Animales , Estudios de Casos y Controles , Cuerpo Estriado/metabolismo , Dopamina/metabolismo , Neuronas Dopaminérgicas/metabolismo , Femenino , Humanos , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Persona de Mediana Edad , Enfermedad de Parkinson/genética , Enfermedad de Parkinson/metabolismo , Factores de Transcripción SOXD/genética , Sustancia Negra/metabolismo , Área Tegmental Ventral/metabolismo , Área Tegmental Ventral/patología
2.
Gene ; 767: 145148, 2021 Jan 30.
Artículo en Inglés | MEDLINE | ID: mdl-32949698

RESUMEN

Ischemic stroke is a common clinical cardiovascular disease and often accompanied by central nervous system injury. It often causes paralysis or loss of motor function after central nervous system injury and significantly reduces the patient's quality of life. At present, there is no effective treatment strategy for nerve damage caused by ischemic stroke. Therefore, it is urgently need to explore effective treatment targets. The protein expression of SOX5, VEGF and apoptosis related proteins were measured by western blot. The mRNA expression of SOX5 and VEGF were detected by RT-qPCR. The concentration of S100B and GFAP which are related to nerve damage were detected using ELISA assay. The transcriptional regulation of SOX5 on VEGF was detected using ChIP-PCR and dual luciferase reporter gene assays. The cell apoptosis was measured by TUNEL assay and cell viability was detected by CCK-8 assay. In our study, we found that the expression of SOX5 was significantly reduced when LPS induced apoptosis in PC-12 cells. Overexpression of SOX5 repaired LPS-induced apoptosis. SOX5 promotes VEGF expression as a transcription factor to activate the PI3K/AKT pathway. VEGF also repairs nerve injury and brain tissue injury caused by ischemic stroke. In conclusion, SOX5 transcription regulates the expression of VEGF to activate the PI3K/AKT pathway, which repaired nerve damage caused by ischemic stroke. Therefore, SOX5 could be a new targetto regulate VEGF which can repair nerve injury induced by ischemic stroke.


Asunto(s)
Accidente Cerebrovascular Isquémico/tratamiento farmacológico , Accidente Cerebrovascular Isquémico/metabolismo , Factores de Transcripción SOXD/metabolismo , Animales , Apoptosis/fisiología , Isquemia Encefálica/genética , Isquemia Encefálica/metabolismo , Muerte Celular , Proliferación Celular , Regulación de la Expresión Génica/genética , Fármacos Neuroprotectores/metabolismo , Fármacos Neuroprotectores/uso terapéutico , Células PC12 , Fosfatidilinositol 3-Quinasas/metabolismo , Proteínas Proto-Oncogénicas c-akt/metabolismo , Ratas , Factores de Transcripción SOXD/genética , Factores de Transcripción SOXD/fisiología , Transducción de Señal/genética , Accidente Cerebrovascular/genética , Factor A de Crecimiento Endotelial Vascular/metabolismo , Factores de Crecimiento Endotelial Vascular/metabolismo
3.
Mol Med Rep ; 22(2): 948-956, 2020 08.
Artículo en Inglés | MEDLINE | ID: mdl-32468049

RESUMEN

Dilated cardiomyopathy (DCM) is a disease that can lead to heart expansion and severe heart failure, but the specific pathogenesis remains unclear. Sox5 is a member of the Sox family with a key role in cardiac function. However, the role of Sox5 in DCM remains unclear. In the present study, wild­type mice were intraperitoneally injected with doxorubicin (Dox) to induce DCM, and heart specimens from human patients with DCM were used to investigate the preliminary role of Sox5 in DCM. The present study demonstrated that, compared with control human hearts, the hearts of patients with DCM exhibited high expression levels of Sox5 and activation of the wnt/ß­catenin pathway. This result was consistent with Dox­induced DCM in mice. Furthermore, in Dox­treated mice, apoptosis was activated during the development of DCM. Inflammation and collagen deposition also increased in DCM mice. The results of the present study indicate that Sox5 may be associated with the development of DCM. Sox5 may be a novel potential factor that regulates DCM.


Asunto(s)
Cardiomiopatía Dilatada/metabolismo , Factores de Transcripción SOXD/biosíntesis , Factores de Transcripción SOXD/fisiología , Anciano , Animales , Apoptosis , Cardiomiopatía Dilatada/inducido químicamente , Colágeno/metabolismo , Modelos Animales de Enfermedad , Doxorrubicina/efectos adversos , Femenino , Fibrosis/metabolismo , Humanos , Inflamación/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Persona de Mediana Edad , Miocardio/metabolismo , Miocardio/patología , Proteína Wnt1/metabolismo , beta Catenina/metabolismo
4.
Biomed Pharmacother ; 109: 2128-2135, 2019 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-30551470

RESUMEN

Altered expression of microRNAs (miRNAs) was involved in prostate cancer progression. However, how miRNAs contributed to prostate cancer development remained unknown. Here, we reported that miR-139-5p levels were decreased in prostate cancer tumor tissues and prostate cancer cell lines. Transfection of miR-139-5p mimics reduced cell proliferation and migration ability of prostate cancer cells. Western blotting and RT-qPCR showed that elevation of miR-139-5p greatly inhibited SOX5 expression in prostate cancer cells. Through regulation of SOX5, enhanced expression of miR-139-5p downregulated TWIST, decreased N-cadherin and Vimentin expression, suggesting inhibition of epithelial-mesenchymal transition (EMT) process. The dual luciferase assay validated that SOX5 was a direct target of miR-139-5p. Additionally, a significant negative correlation between SOX5 mRNA levels and miR-139-5p levels were detected in prostate cancer tumor tissues. Our study indicated that miR-139-5p functioned as a tumor suppressor in prostate cancer cells by regulation of SOX5, and it might be a promising target for prostate cancer patients.


Asunto(s)
Progresión de la Enfermedad , Regulación hacia Abajo/fisiología , MicroARNs/biosíntesis , Neoplasias de la Próstata/metabolismo , Factores de Transcripción SOXD/fisiología , Línea Celular , Línea Celular Tumoral , Humanos , Masculino , MicroARNs/genética , Neoplasias de la Próstata/patología
5.
Cell ; 175(2): 372-386.e17, 2018 10 04.
Artículo en Inglés | MEDLINE | ID: mdl-30270042

RESUMEN

Intestinal mesenchymal cells play essential roles in epithelial homeostasis, matrix remodeling, immunity, and inflammation. But the extent of heterogeneity within the colonic mesenchyme in these processes remains unknown. Using unbiased single-cell profiling of over 16,500 colonic mesenchymal cells, we reveal four subsets of fibroblasts expressing divergent transcriptional regulators and functional pathways, in addition to pericytes and myofibroblasts. We identified a niche population located in proximity to epithelial crypts expressing SOX6, F3 (CD142), and WNT genes essential for colonic epithelial stem cell function. In colitis, we observed dysregulation of this niche and emergence of an activated mesenchymal population. This subset expressed TNF superfamily member 14 (TNFSF14), fibroblastic reticular cell-associated genes, IL-33, and Lysyl oxidases. Further, it induced factors that impaired epithelial proliferation and maturation and contributed to oxidative stress and disease severity in vivo. Our work defines how the colonic mesenchyme remodels to fuel inflammation and barrier dysfunction in IBD.


Asunto(s)
Enfermedades Inflamatorias del Intestino/fisiopatología , Mesodermo/fisiología , Animales , Proliferación Celular , Colitis/genética , Colitis/fisiopatología , Colon/fisiología , Células Epiteliales/metabolismo , Fibroblastos/fisiología , Heterogeneidad Genética , Homeostasis , Humanos , Inflamación , Mucosa Intestinal/inmunología , Mucosa Intestinal/fisiología , Intestinos/inmunología , Intestinos/fisiología , Células Madre Mesenquimatosas/fisiología , Mesodermo/metabolismo , Ratones , Ratones Endogámicos C57BL , Miofibroblastos , Pericitos , Células RAW 264.7 , Factores de Transcripción SOXD/fisiología , Análisis de la Célula Individual/métodos , Tromboplastina/fisiología , Miembro 14 de la Superfamilia de Ligandos de Factores de Necrosis Tumoral/genética , Vía de Señalización Wnt/fisiología
6.
Front Immunol ; 9: 749, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-29706965

RESUMEN

Objectives: Fibroblast-like synoviocytes (FLS) exhibit a unique aggressive phenotype in rheumatoid arthritis (RA). Increased FLS migration and subsequent invasion of the extracellular matrix are essential to joint destruction in RA. Our previous research reported that transcription factor SOX5 was highly expressed in RA-FLS. Here, the effects of SOX5 in RA-FLS migration and invasion will be investigated. Methods: The migration and invasion of RA-FLS were evaluated using a transwell chamber assay. The expression of several potential SOX5-targeted genes, including matrix metalloproteinases (MMP-1, 2, 3 and 9), chemokines (CCL4, CCL2, CCR5 and CCR2), and pro-inflammatory cytokines (TNF-α and IL-6), were examined in RA-FLS using SOX5 gain- and loss-of-function study. The molecular mechanisms of SOX5-mediated MMP-9 expressions were assayed by luciferase reporter gene and chromatin immunoprecipitation (ChIP) studies. The in vivo effect of SOX5 on FLS migration and invasion was examined using collagen-induced arthritis (CIA) in DBA/1J mice. Results: Knockdown SOX5 decreased lamellipodium formation, migration, and invasion of RA-FLS. The expression of MMP-9 was the only gene tested to be concomitantly affected by silencing or overexpressing SOX5. ChIP assay revealed that SOX5 was bound to the MMP-9 promoter in RA-FLS. The overexpression of SOX5 markedly enhanced the MMP-9 promoter activity, and specific deletion of a putative SOX5-binding site in MMP-9 promoter diminished this promoter-driven transcription in FLS. Locally knocked down SOX5 inhibited MMP-9 expression in the joint tissue and reduced pannus migration and invasion into the cartilage in CIA mice. Conclusion: SOX5 plays a novel role in mediating migration and invasion of FLS in part by regulating MMP-9 expression in RA.


Asunto(s)
Artritis Experimental/metabolismo , Artritis Reumatoide/metabolismo , Metaloproteinasa 9 de la Matriz/fisiología , Factores de Transcripción SOXD/fisiología , Sinoviocitos/fisiología , Animales , Articulación del Tobillo/patología , Artritis Experimental/patología , Artritis Reumatoide/patología , Movimiento Celular , Células Cultivadas , Humanos , Masculino , Ratones Endogámicos DBA
7.
Cancer Res ; 77(18): 4985-4997, 2017 09 15.
Artículo en Inglés | MEDLINE | ID: mdl-28687615

RESUMEN

Molecular mechanisms preventing self-renewing brain stem cells from oncogenic transformation are poorly defined. We show that the expression levels of SOX5, SOX6, and SOX21 (SOX5/6/21) transcription factors increase in stem cells of the subventricular zone (SVZ) upon oncogenic stress, whereas their expression in human glioma decreases during malignant progression. Elevated levels of SOX5/6/21 promoted SVZ cells to exit the cell cycle, whereas genetic ablation of SOX5/6/21 dramatically increased the capacity of these cells to form glioma-like tumors in an oncogene-driven mouse brain tumor model. Loss-of-function experiments revealed that SOX5/6/21 prevent detrimental hyperproliferation of oncogene expressing SVZ cells by facilitating an antiproliferative expression profile. Consistently, restoring high levels of SOX5/6/21 in human primary glioblastoma cells enabled expression of CDK inhibitors and decreased p53 protein turnover, which blocked their tumorigenic capacity through cellular senescence and apoptosis. Altogether, these results provide evidence that SOX5/6/21 play a central role in driving a tumor suppressor response in brain stem cells upon oncogenic insult. Cancer Res; 77(18); 4985-97. ©2017 AACR.


Asunto(s)
Neoplasias Encefálicas/patología , Transformación Celular Neoplásica/patología , Glioma/patología , Células Madre Neoplásicas/patología , Factores de Transcripción SOXB2/metabolismo , Factores de Transcripción SOXD/fisiología , Animales , Apoptosis , Biomarcadores de Tumor/genética , Biomarcadores de Tumor/metabolismo , Neoplasias Encefálicas/genética , Neoplasias Encefálicas/metabolismo , Proliferación Celular , Transformación Celular Neoplásica/genética , Transformación Celular Neoplásica/metabolismo , Senescencia Celular , Femenino , Glioma/genética , Glioma/metabolismo , Humanos , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Desnudos , Células Madre Neoplásicas/metabolismo , Oncogenes , Factores de Transcripción SOXB2/genética , Células Tumorales Cultivadas , Ensayos Antitumor por Modelo de Xenoinjerto
8.
J BUON ; 22(1): 258-264, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-28365963

RESUMEN

PURPOSE: SOX5 plays important roles in various kinds of cancers. However, the expression and roles of SOX5 in osteosarcoma (OS) have not been investigated well. In the present study we aimed to investigate the mechanism of SOX5 in OS. METHODS: OS and adjacent non-cancerous specimens were obtained from patients with OS. PCR was applied to detect SOX5 mRNA. Then human OS cell lines (U2OS, SoSP-M, SoSP-9607, and MG-63) and one immortalized normal osteoblast hFOB1.19 were investigated. SOX5 knocking with shRNA in U2OS and SOX5 upregulation with recombinant plasmid in MG-63 were applied. Real-time cell monitoring system and invasion assay were used, and Western blot assay was performed to detect the protein level of E-cadherin, N-cadherin, Vimentin and Snail, where Glyceraldehyde3- phosphate dehydrogenase (GAPDH) was presented as control. P<0.05 was considered as statistically significant. RESULTS: Significant upregulation of SOX5 in OS tissues and cell lines was identified. The gain- and loss-of-function studies suggested that OS cell migration and invasion were promoted significantly by SOX5. Additionally, SOX5 promoted epithelial-mesenchymal transition (EMT) by regulation of Snail. CONCLUSION: SOX5 is a novel regulator of EMT in OS, and is a potential target for OS.


Asunto(s)
Neoplasias Óseas/patología , Transición Epitelial-Mesenquimal , Osteosarcoma/patología , Factores de Transcripción SOXD/fisiología , Factores de Transcripción de la Familia Snail/fisiología , Neoplasias Óseas/mortalidad , Línea Celular Tumoral , Movimiento Celular , Quinasas MAP Reguladas por Señal Extracelular/fisiología , Humanos , Invasividad Neoplásica , Osteosarcoma/mortalidad , Proteínas Proto-Oncogénicas c-raf/fisiología
9.
Dev Biol ; 422(1): 47-57, 2017 02 01.
Artículo en Inglés | MEDLINE | ID: mdl-28012818

RESUMEN

The transcription factor Sox10 is a key regulator of vertebrate neural crest development and serves crucial functions in the differentiation of multiple neural crest lineages. In the chick neural crest, two cis-regulatory elements have been identified that mediate Sox10 expression: Sox10E2, which initiates expression in cranial neural crest; Sox10E1 driving expression in vagal and trunk neural crest. Both also mediate Sox10 expression in the otic placode. Here, we have dissected and analyzed the Sox10E1 enhancer element to identify upstream regulatory inputs. Via mutational analysis, we found two critical Sox sites with differential impact on trunk versus otic Sox10E1 mediated reporter expression. Mutation of a combined SoxD/E motif was sufficient to completely abolish neural crest but not ear enhancer activity. However, mutation of both the SoxD/E and another SoxE site eliminated otic Sox10E1 expression. Loss-of-function experiments reveal Sox5 and Sox8 as critical inputs for trunk neural crest enhancer activity, but only Sox8 for its activity in the ear. Finally, we show by ChIP and co-immunoprecipitation that Sox5 directly binds to the SoxD/E site, and that it can interact with Sox8, further supporting their combinatorial role in activation of Sox10E1 in the trunk neural crest. The results reveal important tissue-specific inputs into Sox10 expression in the developing embryo.


Asunto(s)
Elementos de Facilitación Genéticos , Regulación del Desarrollo de la Expresión Génica , Factores de Transcripción SOXD/fisiología , Factores de Transcripción SOXE/genética , Factores de Transcripción SOXE/fisiología , Animales , Sitios de Unión , Embrión de Pollo , Mutación , Cresta Neural/fisiología , Especificidad de Órganos
10.
Oncogene ; 35(13): 1692-702, 2016 Mar 31.
Artículo en Inglés | MEDLINE | ID: mdl-26119940

RESUMEN

Sex-determining region Y box 6 (SOX6) has been described as a tumor-suppressor gene in several cancers. Our previous work has suggested that SOX6 upregulated p21(Waf1/Cip1)(p21) expression in a p53-dependent manner; however, the underlying mechanism has remained elusive. In this study, we confirmed that SOX6 can suppress cell proliferation in vitro and in vivo by stabilizing p53 protein and subsequently upregulating p21. Co-immunoprecipitation and immunocytofluorescence assays demonstrated that SOX6 can promote formation of the p14ARF-HDM2-p53 ternary complex by promoting translocation of p14ARF (p14 alternate reading frame tumor suppressor) to the nucleoplasm, thereby inhibiting HDM2-mediated p53 nuclear export and degradation. Chromatin immunoprecipitation combined with PCR assay proved that SOX6 can bind to a potential binding site in the regulatory region of the c-Myc gene. Furthermore, we confirmed that SOX6 can downregulate the expression of c-Myc, as well as its direct target gene nucleophosmin 1 (NPM1), and that the SOX6-induced downregulation of NPM1 is linked to translocation of p14ARF to the nucleoplasm. Finally, we showed that the highly conserved high-mobility group (HMG) domain of SOX6 is required for SOX6-mediated p53 stabilization and tumor inhibitory activity. Collectively, these results reveal a new mechanism of SOX6-mediated tumor suppression involving p21 upregulation via the p14ARF-HDM2-p53 axis in an HMG domain-dependent manner.


Asunto(s)
Transformación Celular Neoplásica/genética , Proteínas Proto-Oncogénicas c-mdm2/fisiología , Factores de Transcripción SOXD/fisiología , Proteína p14ARF Supresora de Tumor/fisiología , Proteína p53 Supresora de Tumor/fisiología , Proteína p53 Supresora de Tumor/farmacocinética , Animales , Proliferación Celular/genética , Femenino , Genes Supresores de Tumor/fisiología , Células HeLa , Humanos , Ratones , Ratones Endogámicos C57BL , Ratones Desnudos , Nucleofosmina , Estructura Terciaria de Proteína/genética , Factores de Transcripción SOXD/química , Factores de Transcripción SOXD/genética , Transducción de Señal/fisiología , Células Tumorales Cultivadas
11.
Apoptosis ; 21(2): 174-83, 2016 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-26659076

RESUMEN

Sepsis-induced cardiac apoptosis is one of the major pathogenic factors in myocardial dysfunction. As it enhances numerous proinflammatory factors, lipopolysaccharide (LPS) is considered the principal mediator in this pathological process. However, the detailed mechanisms involved are unclear. In this study, we attempted to explore the mechanisms involved in LPS-induced cardiomyocyte apoptosis. We found that LPS stimulation inhibited microRNA (miR)-499 expression and thereby upregulated the expression of SOX6 and PDCD4 in neonatal rat cardiomyocytes. We demonstrate that SOX6 and PDCD4 are target genes of miR-499, and they enhance LPS-induced cardiomyocyte apoptosis by activating the BCL-2 family pathway. The apoptosis process enhanced by overexpression of SOX6 or PDCD4, was rescued by the cardiac-abundant miR-499. Overexpression of miR-499 protected the cardiomyocytes against LPS-induced apoptosis. In brief, our results demonstrate the existence of a miR-499-SOX6/PDCD4-BCL-2 family pathway in cardiomyocytes in response to LPS stimulation.


Asunto(s)
Proteínas Reguladoras de la Apoptosis/fisiología , Apoptosis/inmunología , Lipopolisacáridos/farmacología , MicroARNs/metabolismo , Miocitos Cardíacos/fisiología , Factores de Transcripción SOXD/fisiología , Animales , Células Cultivadas , Células HeLa , Ventrículos Cardíacos/citología , Humanos , MicroARNs/genética , Interferencia de ARN/inmunología , Ratas Sprague-Dawley
12.
Mol Cell Biochem ; 412(1-2): 49-57, 2016 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-26590087

RESUMEN

Sox6 is a transcription factor that induces neuronal differentiation in P19 cells; its suppression not only inhibits neuronal differentiation but also induces retinoic acid (RA)-dependent apoptosis of P19 cells. In the present study, we found that Sox6 suppression-induced apoptosis was mediated by activation of caspase 9 and 3. Moreover, we noted a weak leakage of cytochrome c into the cytoplasm from the mitochondria, indicating that apoptosis occurs through a mitochondrial pathway in Sox6-suppressed P19 (P19[anti-Sox6]) cells. Sox6 suppression in the presence of RA also induced the expression and secretion of bone morphogenetic protein 4 (BMP-4). Addition of an anti-BMP-4 antibody for neutralization increased cell viability and led to RA-dependent death of P19[anti-Sox6] cells. Our results indicate that Sox6 suppression induces RA-dependent cell death of P19 cells, mediated by BMP-4 expression and secretion. Normally, high Sox6 expression leads to RA-mediated neuronal differentiation in P19 cells; however, Sox6 deficiency induces production and secretion of BMP-4, which mediates selective cell death. Our findings suggest that Sox6 contributes to cell survival by suppressing BMP-4 transcription during neuronal differentiation.


Asunto(s)
Apoptosis/efectos de los fármacos , Proteína Morfogenética Ósea 4/metabolismo , Diferenciación Celular , Factores de Transcripción SOXD/fisiología , Tretinoina/farmacología , Animales , Apoptosis/fisiología , Caspasas/metabolismo , Línea Celular , Activación Enzimática , Ratones
14.
Int J Dev Biol ; 59(10-12): 471-8, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-26864488

RESUMEN

Members of the Sox gene family play roles in many biological processes including organogenesis. We carried out comparative in situ hybridization analysis of seventeen sox genes (Sox1-14, 17, 18, 21) during murine odontogenesis from the epithelial thickening to the cytodifferentiation stages. Localized expression of five Sox genes (Sox6, 9, 13, 14 and 21) was observed in tooth bud epithelium. Sox13 showed restricted expression in the primary enamel knots. At the early bell stage, three Sox genes (Sox8, 11, 17 and 21) were expressed in pre-ameloblasts, whereas two others (Sox5 and 18) showed expression in odontoblasts. Sox genes thus showed a dynamic spatio-temporal expression during tooth development.


Asunto(s)
Embrión de Mamíferos/metabolismo , Regulación del Desarrollo de la Expresión Génica , Odontogénesis/fisiología , Factores de Transcripción SOXD/fisiología , Germen Dentario/metabolismo , Animales , Embrión de Mamíferos/citología , Hibridación in Situ , Ratones , Ratones Transgénicos , Germen Dentario/citología
15.
FEBS Lett ; 588(18): 3352-60, 2014 Sep 17.
Artículo en Inglés | MEDLINE | ID: mdl-25101494

RESUMEN

Catsper is a Ca(2+)permeable channel required for sperm hyperactivation. In spite of its central role in male fertility, the transcriptional mechanisms that regulate Catsper1 expression are ill defined. In this work, we describe the identification and characterization of important regulatory elements in the murine Catsper1 gene proximal promoter. Four transcription start sites and three functional Sox-binding sites were identified in the Catsper1 promoter. Interestingly, transcription factors Sox5 and Sox9 caused a significant increase in transactivation of the Catsper1 promoter in heterologous systems, and chromatin immunoprecipitation assays showed that both transcription factors interact with the Catsper1 promoter in vivo. These results provide new insights into the molecular mechanisms that control Catsper channel expression.


Asunto(s)
Canales de Calcio/genética , Regulación de la Expresión Génica , Factor de Transcripción SOX9/fisiología , Factores de Transcripción SOXD/fisiología , Animales , Secuencia de Bases , Sitios de Unión , Canales de Calcio/metabolismo , Células HEK293 , Humanos , Masculino , Ratones , Datos de Secuencia Molecular , Regiones Promotoras Genéticas , Unión Proteica , Espermátides/metabolismo , Espermatocitos/metabolismo , Testículo/citología , Sitio de Iniciación de la Transcripción , Transcripción Genética
16.
Cell Rep ; 8(4): 1018-25, 2014 Aug 21.
Artículo en Inglés | MEDLINE | ID: mdl-25127144

RESUMEN

Distinct midbrain dopamine (mDA) neuron subtypes are found in the substantia nigra pars compacta (SNc) and the ventral tegmental area (VTA), but it is mainly SNc neurons that degenerate in Parkinson's disease. Interest in how mDA neurons develop has been stimulated by the potential use of stem cells in therapy or disease modeling. However, very little is known about how specific dopaminergic subtypes are generated. Here, we show that the expression profiles of the transcription factors Sox6, Otx2, and Nolz1 define subpopulations of mDA neurons already at the neural progenitor cell stage. After cell-cycle exit, Sox6 selectively localizes to SNc neurons, while Otx2 and Nolz1 are expressed in a subset of VTA neurons. Importantly, Sox6 ablation leads to decreased expression of SNc markers and a corresponding increase in VTA markers, while Otx2 ablation has the opposite effect. Moreover, deletion of Sox6 affects striatal innervation and dopamine levels. We also find reduced Sox6 levels in Parkinson's disease patients. These findings identify Sox6 as a determinant of SNc neuron development and should facilitate the engineering of relevant mDA neurons for cell therapy and disease modeling.


Asunto(s)
Neuronas Dopaminérgicas/fisiología , Factores de Transcripción Otx/fisiología , Factores de Transcripción SOXD/fisiología , Sustancia Negra/citología , Área Tegmental Ventral/citología , Animales , Tipificación del Cuerpo , Humanos , Ratones Transgénicos , Especificidad de Órganos , Sustancia Negra/embriología , Sustancia Negra/metabolismo , Área Tegmental Ventral/embriología , Área Tegmental Ventral/metabolismo
17.
J Pharmacol Sci ; 122(4): 318-25, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23883486

RESUMEN

Pre-chondrocytes undergo cellular differentiation stages during chondrogenesis under the influence by different transcription factors such as sry-type high mobility group box-9 (Sox9) and runt-related transcription factor-2 (Runx2). We have shown upregulation by parathyroid hormone (PTH) of the clock gene Period-1 (Per1) through the cAMP/protein kinase A signaling pathway in pre-chondrocytic ATDC5 cells. Here, we investigated the role of Per1 in the suppression of chondrogenic differentiation by PTH. In ATDC5 cells exposed to 10 nM PTH, a drastic but transient increase in Per1 expression was seen only 1 h after addition together with a prolonged decrease in Sox6 levels. However, no significant changes were induced in Sox5 and Runx2 levels in cells exposed to PTH. In stable Per1 transfectants, a significant decrease in Sox6 levels was seen, with no significant changes in Sox5 and Sox9 levels, in addition to the inhibition of gene transactivation by Sox9 allies. Knockdown of Per1 by siRNA significantly increased the Sox6 and type II collagen levels in cells cultured for 24 - 60 h. These results suggest that Per1 plays a role in the suppressed chondrocytic differentiation by PTH through a mechanism relevant to negative regulation of transactivation of the Sox6 gene during chondrogenesis.


Asunto(s)
Diferenciación Celular/genética , Condrocitos/citología , Condrogénesis/genética , Regulación del Desarrollo de la Expresión Génica/genética , Proteínas Circadianas Period/genética , Proteínas Circadianas Period/fisiología , Factores de Transcripción SOXD/genética , Factores de Transcripción SOXD/fisiología , Animales , Diferenciación Celular/efectos de los fármacos , Células Cultivadas , Condrogénesis/efectos de los fármacos , Proteínas Quinasas Dependientes de AMP Cíclico/fisiología , Regulación del Desarrollo de la Expresión Génica/efectos de los fármacos , Ratones , Hormona Paratiroidea/farmacología , Proteínas Circadianas Period/metabolismo , ARN Interferente Pequeño , Factores de Transcripción SOXD/metabolismo , Transducción de Señal/genética , Transducción de Señal/fisiología , Activación Transcripcional/genética
18.
Osteoarthritis Cartilage ; 19(10): 1246-53, 2011 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-21843649

RESUMEN

OBJECTIVE: This study aims to investigate the regulation of expression of Cartilage oligomeric matrix protein (COMP), which is predominately expressed by chondrocytes and functions to organize the extracellular matrix. Mutations in COMP cause two skeletal dysplasias: pseudoachondroplasia and multiple epiphyseal dysplasia. The mechanism controlling COMP expression during chondrocyte differentiation is still poorly understood. DESIGN: Primary human bone marrow-derived stem cells were induced to differentiate into chondrocyte by pellet cultures. We then compared the temporal expression of COMP with the well-characterized cartilage-specific Type II collagen (Col2a1), and their response to transforming growth factor (TGF)ß and Sox trio (Sox5, 6, and 9) stimulation. RESULTS: COMP and Col2a1 expression are differentially regulated by three distinct mechanisms. First, upregulation of COMP mRNA precedes Col2a1 by several days during chondrogenesis. Second, COMP expression is independent of high cell density but requires TGF-ß1. Induction of COMP mRNA by TGF-ß1 is detected within 2h in the absence of protein synthesis and is blocked by specific inhibitors of the TGFß signaling pathway; and therefore, COMP is a primary TFGß-response gene. Lastly, while Col2a1 expression is intimately controlled by the Sox trio, overexpression of Sox trio fails to activate the COMP promoter. CONCLUSION: COMP and Col2a1 expression are regulated differently during chondrogenesis. COMP is a primary response gene of TGFß and its fast induction during chondrogenesis suggests that COMP is suitable for rapidly accessing the chondrogenic potential of stem cells.


Asunto(s)
Células de la Médula Ósea/citología , Condrogénesis/fisiología , Colágeno Tipo II/metabolismo , Proteínas de la Matriz Extracelular/metabolismo , Glicoproteínas/metabolismo , Células Madre Mesenquimatosas/metabolismo , Factor de Crecimiento Transformador beta1/fisiología , Células de la Médula Ósea/metabolismo , Proteína de la Matriz Oligomérica del Cartílago , Regulación de la Expresión Génica , Humanos , Proteínas Matrilinas , Células Madre Mesenquimatosas/citología , Factor de Transcripción SOX9/fisiología , Factores de Transcripción SOXD/fisiología , Transducción de Señal , Regulación hacia Arriba
19.
Dev Dyn ; 240(6): 1311-21, 2011 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-21495113

RESUMEN

Approximately 20,000 genes are encoded in our genome, one tenth of which are thought to be transcription factors. Considering the complexity and variety of cell types generated during development, many transcription factors likely play multiple roles. Uncovering the versatile roles of Sox6 in vertebrate development sheds some light on how an organism efficiently utilizes the limited resources of transcription factors. The structure of the Sox6 gene itself may dictate its functional versatility. First, Sox6 contains no known regulatory domains; instead, it utilizes various cofactors. Second, Sox6 has a long 3'-UTR that contains multiple microRNA targets, thus its protein level is duly adjusted by cell type-specific microRNAs. Just combining these two characteristics alone makes Sox6 extremely versatile. To date, Sox6 has been reported to regulate differentiation of tissues of mesoderm, ectoderm, and endoderm origins, making Sox6 a truly multifaceted transcription factor.


Asunto(s)
Regulación del Desarrollo de la Expresión Génica/genética , Factores de Transcripción SOXD/fisiología , Vertebrados/crecimiento & desarrollo , Vertebrados/genética , Secuencia de Aminoácidos , Animales , Humanos , Modelos Biológicos , Datos de Secuencia Molecular , Conformación Proteica , Factores de Transcripción SOXD/química , Factores de Transcripción SOXD/genética , Factores de Transcripción SOXD/metabolismo , Homología de Secuencia de Aminoácido , Factores de Transcripción/metabolismo , Factores de Transcripción/fisiología , Vertebrados/embriología
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