Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 145
Filtrar
1.
Exp Eye Res ; 214: 108887, 2022 01.
Artículo en Inglés | MEDLINE | ID: mdl-34890603

RESUMEN

PURPOSE: To explore the mechanisms underlying stemness maintenance of retinoblastoma (RB) stem cells (RSCs). METHODS: The retinoblastoma stem-like cells (RSLCs) were isolated by single cell cloning in combination of examination of sphere-forming capacities. The stemness of the cells were characterized by the sphere-forming capacity and the expression levels of RSCs markers. Gene manipulation was performed by lentivirus system. Transcriptional regulation was identified by qRT-PCR, luciferase reporter, nuclear run-on and DNA pull-down assay. Spearman analysis was employed for correlation analysis of genes in tumor tissues of RB patients. RESULTS: The isolated RSLCs exhibited enhanced sphere-forming capacity and constantly higher levels of CD44, ABCG2, SOX2 and PAX6, but not CD133. SOX2 positively regulated the stemness of RSLCs. SOX2 directly binds to the promoters of WWTR1 and YAP and transcriptionally activates WWTR1 and YAP. Knockdown of WWTR1 or YAP partially abolished the effect of SOX2 on the stemness of RSLCs. CONCLUSIONS: SOX2, as a key deriver, maintains RB stemness by activating Hippo/YAP signaling. Inhibition of Hippo/YAP signaling would be an effective strategy for human RB caused by SOX2 upregulation.


Asunto(s)
Vía de Señalización Hippo/fisiología , Células Madre Neoplásicas/patología , Neoplasias de la Retina/patología , Retinoblastoma/patología , Factores de Transcripción SOXB1/fisiología , Proteínas Señalizadoras YAP/fisiología , Proteínas Adaptadoras Transductoras de Señales/genética , Animales , Western Blotting , Femenino , Humanos , Ratones , Ratones Endogámicos BALB C , Ratones Desnudos , Proteínas de Neoplasias/metabolismo , Células Madre Neoplásicas/metabolismo , Reacción en Cadena en Tiempo Real de la Polimerasa , Neoplasias de la Retina/metabolismo , Retinoblastoma/metabolismo , Factores de Transcripción/genética , Trasplante Heterólogo , Células Tumorales Cultivadas
3.
Brain Res ; 1752: 147265, 2021 02 01.
Artículo en Inglés | MEDLINE | ID: mdl-33422527

RESUMEN

Transcription factor Sox2 is widely recognized for its critical roles in the nervous system, including the neural retina. Here, we aimed to reveal the function of Sox2 in the process of mouse postnatal development. After the suppression of Sox2 at P0, there was an increase number in bipolar cells but a decrease in amacrine cells. Inhibited Sox2 expression also led to decreased visual function. Furthermore, we found a distinctive type of retinal cells expressing the characteristic proteins of both bipolar cells and amacrine cells at P6, which may be an intermediate state in which amacrine cells were transforming into bipolar cells. Transcription factors associated with the development of bipolar cells and amacrine cells also support those changes. Our work indicated that inhibition of Sox2 could change cell fate by affecting transcription factors in the development of bipolar cells and amacrine cells, may provide new directions for the study and treatment of retinal genetic diseases and retinal dysplasia.


Asunto(s)
Células Amacrinas/fisiología , Retina/crecimiento & desarrollo , Células Bipolares de la Retina/fisiología , Factores de Transcripción SOXB1/fisiología , Animales , Animales Recién Nacidos , Femenino , Regulación del Desarrollo de la Expresión Génica , Masculino , Ratones
4.
Int J Mol Sci ; 23(1)2021 Dec 26.
Artículo en Inglés | MEDLINE | ID: mdl-35008655

RESUMEN

In mammals, the hypothalamic suprachiasmatic nucleus (SCN) functions as the central circadian pacemaker, orchestrating behavioral and physiological rhythms in alignment to the environmental light/dark cycle. The neurons that comprise the SCN are anatomically and functionally heterogeneous, but despite their physiological importance, little is known about the pathways that guide their specification and differentiation. Here, we report that the stem/progenitor cell transcription factor, Sex determining region Y-box 2 (Sox2), is required in the embryonic SCN to control the expression of SCN-enriched neuropeptides and transcription factors. Ablation of Sox2 in the developing SCN leads to downregulation of circadian neuropeptides as early as embryonic day (E) 15.5, followed by a decrease in the expression of two transcription factors involved in SCN development, Lhx1 and Six6, in neonates. Thymidine analog-retention assays revealed that Sox2 deficiency contributed to reduced survival of SCN neurons during the postnatal period of cell clearance, but did not affect progenitor cell proliferation or SCN specification. Our results identify SOX2 as an essential transcription factor for the proper differentiation and survival of neurons within the developing SCN.


Asunto(s)
Diferenciación Celular , Desarrollo Embrionario , Neuronas/metabolismo , Factores de Transcripción SOXB1/metabolismo , Núcleo Supraquiasmático/metabolismo , Animales , Ritmo Circadiano , Ratones , Neuronas/fisiología , Factores de Transcripción SOXB1/fisiología , Núcleo Supraquiasmático/crecimiento & desarrollo , Núcleo Supraquiasmático/fisiología
5.
J Neurosci Res ; 99(2): 679-698, 2021 02.
Artículo en Inglés | MEDLINE | ID: mdl-33099767

RESUMEN

Spiral ganglion neurons (SGNs) are the primary afferent neurons of the auditory system, and together with their attendant glia, form the auditory nerve. Within the cochlea, satellite glial cells (SGCs) encapsulate the cell body of SGNs, whereas Schwann cells (SCs) wrap their peripherally- and centrally-directed neurites. Despite their likely importance in auditory nerve function and homeostasis, the physiological properties of auditory glial cells have evaded description. Here, we characterized the voltage-activated membrane currents of glial cells from the mouse cochlea. We identified a prominent weak inwardly rectifying current in SGCs within cochlear slice preparations (postnatal day P5-P6), which was also present in presumptive SGCs within dissociated cultures prepared from the cochleae of hearing mice (P14-P15). Pharmacological block by Ba2+ and desipramine suggested that channels belonging to the Kir4 family mediated the weak inwardly rectifying current, and post hoc immunofluorescence implicated the involvement of Kir4.1 subunits. Additional electrophysiological profiles were identified for glial cells within dissociated cultures, suggesting that glial subtypes may have specific membrane properties to support distinct physiological roles. Immunofluorescence using fixed cochlear sections revealed that although Kir4.1 is restricted to SGCs after the onset of hearing, these channels are more widely distributed within the glial population earlier in postnatal development (i.e., within both SGCs and SCs). The decrease in Kir4.1 immunofluorescence during SC maturation was coincident with a reduction of Sox2 expression and advancing neurite myelination. The data suggest a diversification of glial properties occurs in preparation for sound-driven activity in the auditory nerve.


Asunto(s)
Audición/fisiología , Neuroglía/fisiología , Ganglio Espiral de la Cóclea/citología , Potenciales de Acción , Animales , Bario/farmacología , Células Cultivadas , Nervio Coclear/fisiología , Desipramina/farmacología , Femenino , Transporte Iónico , Masculino , Potenciales de la Membrana , Ratones , Ratones Endogámicos C57BL , Vaina de Mielina/fisiología , Neuritas/ultraestructura , Neuronas Aferentes/fisiología , Técnicas de Placa-Clamp , Canales de Potasio de Rectificación Interna/fisiología , Factores de Transcripción SOXB1/fisiología
6.
J Hematol Oncol ; 13(1): 119, 2020 09 03.
Artículo en Inglés | MEDLINE | ID: mdl-32883331

RESUMEN

Tumor metastasis accounts for the majority of cancer-related deaths; it is therefore important to develop preclinical models that faithfully recapitulate disease progression. Here, we generated paired organoids derived from primary tumors and matched liver metastases in the same colorectal cancer (CRC) patients. Despite the fact that paired organoids exhibit comparable gene expression and cell morphology, organoids from metastatic lesions demonstrate more aggressive phenotypes, tumorigenesis, and metastatic capacity than those from primary lesions. Transcriptional analyses of the paired organoids reveal signature genes and pathways altered during the progression of CRC, including SOX2. Further study shows that inducible knockdown of SOX2 attenuated invasion, proliferation, and liver metastasis outgrowth. Taken together, we use patient-derived paired primary and metastatic cancer organoids to model CRC metastasis and illustrate that SOX2 is associated with CRC progression and may serve as a potential prognostic biomarker and therapeutic target of CRC.


Asunto(s)
Carcinoma/secundario , Neoplasias Colorrectales/patología , Neoplasias Hepáticas/secundario , Modelos Biológicos , Invasividad Neoplásica/patología , Organoides , Animales , Biomarcadores de Tumor , Carcinoma/patología , Progresión de la Enfermedad , Perfilación de la Expresión Génica , Regulación Neoplásica de la Expresión Génica , Xenoinjertos , Humanos , Neoplasias Hepáticas/patología , Ratones , Proteínas de Neoplasias/biosíntesis , Proteínas de Neoplasias/genética , Proteínas de Neoplasias/fisiología , Organoides/efectos de los fármacos , Organoides/patología , Organoides/trasplante , Factores de Transcripción SOXB1/biosíntesis , Factores de Transcripción SOXB1/genética , Factores de Transcripción SOXB1/fisiología
7.
Science ; 369(6502): 397-403, 2020 07 24.
Artículo en Inglés | MEDLINE | ID: mdl-32703873

RESUMEN

Embryonic stem cells can propagate indefinitely in a pluripotent state, able to differentiate into all types of specialized cells when restored to the embryo. What sustains their pluripotency during propagation remains unclear. Here, we show that core pluripotency factors OCT4 and SOX2 suppress chaperone-mediated autophagy (CMA), a selective form of autophagy, until the initiation of differentiation. Low CMA activity promotes embryonic stem cell self-renewal, whereas its up-regulation enhances differentiation. CMA degrades isocitrate dehydrogenases IDH1 and IDH2 and reduces levels of intracellular α-ketoglutarate, an obligatory cofactor for various histone and DNA demethylases involved in pluripotency. These findings suggest that CMA mediates the effect of core pluripotency factors on metabolism, shaping the epigenetic landscape of stem cells and governing the balance between self-renewal and differentiation.


Asunto(s)
Diferenciación Celular , Autofagia Mediada por Chaperones , Células Madre Embrionarias/fisiología , Animales , Línea Celular , Epigénesis Genética , Histonas/fisiología , Ácidos Cetoglutáricos/metabolismo , Ratones , Factor 3 de Transcripción de Unión a Octámeros/fisiología , Factores de Transcripción SOXB1/fisiología
8.
Cell Rep ; 31(2): 107511, 2020 04 14.
Artículo en Inglés | MEDLINE | ID: mdl-32294450

RESUMEN

Medulloblastoma (MB) is a neoplasm linked to dysregulated cerebellar development. Previously, we demonstrated that the Sonic Hedgehog (SHH) subgroup grows hierarchically, with Sox2+ cells at the apex of tumor progression and relapse. To test whether this mechanism is rooted in a normal developmental process, we studied the role of Sox2 in cerebellar development. We find that the external germinal layer (EGL) is derived from embryonic Sox2+ precursors and that the EGL maintains a rare fraction of Sox2+ cells during the first postnatal week. Through lineage tracing and single-cell analysis, we demonstrate that these Sox2+ cells are within the Atoh1+ lineage, contribute extensively to adult granule neurons, and resemble Sox2+ tumor cells. Critically, constitutive activation of the SHH pathway leads to their aberrant persistence in the EGL and rapid tumor onset. We propose that failure to eliminate this rare but potent developmental population is the tumor initiation mechanism in SHH-subgroup MB.


Asunto(s)
Meduloblastoma/etiología , Meduloblastoma/metabolismo , Factores de Transcripción SOXB1/metabolismo , Animales , Linaje de la Célula/genética , Células Cultivadas , Neoplasias Cerebelosas/patología , Cerebelo/embriología , Femenino , Proteínas Hedgehog/metabolismo , Humanos , Masculino , Ratones Noqueados , Ratones Transgénicos , Recurrencia Local de Neoplasia/patología , Células-Madre Neurales/metabolismo , Neurogénesis , Neuronas/metabolismo , Factores de Transcripción SOXB1/fisiología , Transducción de Señal/fisiología , Análisis de la Célula Individual/métodos
9.
Proc Natl Acad Sci U S A ; 117(12): 6942-6950, 2020 03 24.
Artículo en Inglés | MEDLINE | ID: mdl-32139605

RESUMEN

Pluripotent embryonic stem cells (ESCs) contain the potential to form a diverse array of cells with distinct gene expression states, namely the cells of the adult vertebrate. Classically, diversity has been attributed to cells sensing their position with respect to external morphogen gradients. However, an alternative is that diversity arises in part from cooption of fluctuations in the gene regulatory network. Here we find ESCs exhibit intrinsic heterogeneity in the absence of external gradients by forming interconverting cell states. States vary in developmental gene expression programs and display distinct activity of microRNAs (miRNAs). Notably, miRNAs act on neighborhoods of pluripotency genes to increase variation of target genes and cell states. Loss of miRNAs that vary across states reduces target variation and delays state transitions, suggesting variable miRNAs organize and propagate variation to promote state transitions. Together these findings provide insight into how a gene regulatory network can coopt variation intrinsic to cell systems to form robust gene expression states. Interactions between intrinsic heterogeneity and environmental signals may help achieve developmental outcomes.


Asunto(s)
Diferenciación Celular , Células Madre Embrionarias/fisiología , Regulación del Desarrollo de la Expresión Génica , Redes Reguladoras de Genes , MicroARNs/genética , Animales , Proteínas Argonautas/fisiología , Células Madre Embrionarias/citología , Perfilación de la Expresión Génica , Ratones , Ratones Noqueados , Proteína Homeótica Nanog/fisiología , Proteínas de Unión al ARN/fisiología , Factores de Transcripción SOXB1/fisiología , Transducción de Señal
10.
Cancer Res ; 80(8): 1644-1655, 2020 04 15.
Artículo en Inglés | MEDLINE | ID: mdl-32094299

RESUMEN

Glioblastoma multiforme (GBM) and other solid malignancies are heterogeneous and contain subpopulations of tumor cells that exhibit stem-like features. Our recent findings point to a dedifferentiation mechanism by which reprogramming transcription factors Oct4 and Sox2 drive the stem-like phenotype in glioblastoma, in part, by differentially regulating subsets of miRNAs. Currently, the molecular mechanisms by which reprogramming transcription factors and miRNAs coordinate cancer stem cell tumor-propagating capacity are unclear. In this study, we identified miR-486-5p as a Sox2-induced miRNA that targets the tumor suppressor genes PTEN and FoxO1 and regulates the GBM stem-like cells. miR-486-5p associated with the GBM stem cell phenotype and Sox2 expression and was directly induced by Sox2 in glioma cell lines and patient-derived neurospheres. Forced expression of miR-486-5p enhanced the self-renewal capacity of GBM neurospheres, and inhibition of endogenous miR-486-5p activated PTEN and FoxO1 and induced cell death by upregulating proapoptotic protein BIM via a PTEN-dependent mechanism. Furthermore, delivery of miR-486-5p antagomirs to preestablished orthotopic GBM neurosphere-derived xenografts using advanced nanoparticle formulations reduced tumor sizes in vivo and enhanced the cytotoxic response to ionizing radiation. These results define a previously unrecognized and therapeutically targetable Sox2:miR-486-5p axis that enhances the survival of GBM stem cells by repressing tumor suppressor pathways. SIGNIFICANCE: This study identifies a novel axis that links core transcriptional drivers of cancer cell stemness to miR-486-5p-dependent modulation of tumor suppressor genes that feeds back to regulate glioma stem cell survival.


Asunto(s)
Neoplasias Encefálicas/patología , Supervivencia Celular , Proteína Forkhead Box O1/genética , Genes Supresores de Tumor , Glioblastoma/patología , MicroARNs/metabolismo , Proteínas de Neoplasias/fisiología , Fosfohidrolasa PTEN/genética , Factores de Transcripción SOXB1/fisiología , Animales , Proteína 11 Similar a Bcl2/metabolismo , Neoplasias Encefálicas/genética , Neoplasias Encefálicas/metabolismo , Neoplasias Encefálicas/radioterapia , Muerte Celular , Desdiferenciación Celular/genética , Línea Celular Tumoral , Reprogramación Celular/fisiología , Represión Epigenética , Femenino , Regulación Neoplásica de la Expresión Génica , Glioblastoma/genética , Glioblastoma/metabolismo , Glioblastoma/radioterapia , Xenoinjertos , Humanos , Ratones , Ratones Desnudos , MicroARNs/administración & dosificación , MicroARNs/antagonistas & inhibidores , MicroARNs/genética , Nanopartículas/administración & dosificación , Proteínas de Neoplasias/genética , Células Madre Neoplásicas/metabolismo , Células Madre Neoplásicas/patología , Células Madre Neoplásicas/efectos de la radiación , Células-Madre Neurales , Factor 3 de Transcripción de Unión a Octámeros/metabolismo , Tolerancia a Radiación , Distribución Aleatoria , Factores de Transcripción SOXB1/genética , Factores de Transcripción SOXB1/metabolismo , Transfección/métodos , Carga Tumoral , Ensayo de Tumor de Célula Madre/métodos , Regulación hacia Arriba
11.
Dev Biol ; 461(2): 160-171, 2020 05 15.
Artículo en Inglés | MEDLINE | ID: mdl-32059837

RESUMEN

In amniotes, unlike primary neurulation in the anterior body, secondary neurulation (SN) proceeds along with axial elongation by the mesenchymal-to-epithelial transition of SN precursors in the tail bud. It has been under debate whether the SN is generated by neuromesodermal common progenitor cells (NMPs) or neural restricted lineage. Our direct cell labeling and serial transplantations identify uni-fated (neural) precursors in the early tail bud. The uni-fated SN precursor territory is further divided into two subpopulations, neural-differentiating and self-renewing cells, which are regulated by high- and low levels of Sox2, respectively. Unexpectedly, uni-fated SN precursors change their fate at later stages to produce both SN and mesoderm. Thus, chicken embryos adopt a previously unappreciated prolonged phase with uni-fated SN stem cells in the early tail bud, which is absent or very limited in mouse embryos.


Asunto(s)
Autorrenovación de las Células/fisiología , Pollos/genética , Células-Madre Neurales/citología , Tubo Neural/embriología , Neurulación/fisiología , Factores de Transcripción SOXB1/fisiología , Cola (estructura animal)/embriología , Animales , Linaje de la Célula , Embrión de Pollo , Genes Reporteros , Mesodermo/citología , Tubo Neural/citología , Neurulación/genética , Factores de Transcripción SOXB1/antagonistas & inhibidores , Factores de Transcripción SOXB1/genética , Cola (estructura animal)/citología
12.
Pathol Oncol Res ; 26(2): 693-706, 2020 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-30712195

RESUMEN

Sex determining region Y-box 2 (Sox2) is a transcription factor that is essential for maintaining self-renewal or pluripotency of undifferentiated embryonic stem cells. The expression and distribution of Sox2 in tumor tissues have been extensively recorded, which are related to the progression and metastasis of tumor. However, a complete mechanistic understanding of Sox2 regulation and function remains to be studied. Herein, we show new potential properties of Sox2 regulation and functions from bioinformatics analysis. We use numerous algorithms to characterize the Sox2 gene promoter elements and the Sox2 protein structure, physio-chemical, localization properties and its evolutionary relationships. The expression of Sox2 is regulated by a diverse set of transcription factors and associated with the levels of methylation of CpG Islands in promoters. The structural properties of Sox2 indicate that Sox2 expresses as a stem cell marker in a variety of stem cells. Sox2 together with other transcription factors or proteins regulate the expression of downstream target genes, which makes a great difference to the biological function of stem cells. Not only stem cells, Sox2 also play an important role in tumor cells. In conclusion, this information from bioinformatics analysis will help to understand Sox2 regulation and functions better in future attempts.


Asunto(s)
Algoritmos , Biología Computacional , Factores de Transcripción SOXB1/fisiología , Humanos
13.
Neuroscience ; 422: 146-160, 2019 12 01.
Artículo en Inglés | MEDLINE | ID: mdl-31678344

RESUMEN

The vestibular system of the inner ear contains Type I and Type II hair cells (HCs) generated from sensory progenitor cells; however, little is known about how the HC subtypes are formed. Sox2 (encoding SRY-box 2) is expressed in Type II, but not in Type I, HCs. The present study aimed to investigate the role of SOX2 in cell fate determination in Type I vs. Type II HCs. First, we confirmed that Type I HCs developed from Sox2-expressing cells through lineage tracing of Sox2-positive cells using a CAG-tdTomato reporter mouse crossed with a Sox2-CreER mouse. Then, Sox2 loss of function was induced in HCs, using Sox2flox transgenic mice crossed with a Gfi1-Cre driver mouse. Knockout of Sox2 in HCs increased the number of Type I HCs and decreased the number of Type II HCs, while the total number of HCs and Sox2-positive supporting cells did not change. In addition, the effect of Sox2-knockout persisted into adulthood, resulting in an increased number of Type I HCs. These results demonstrate that SOX2 plays a critical role in the determination of Type II vs. Type I HC fate. The results suggested that Sox2 is a potential target for generating Type I HCs, which may be important for regenerative strategies for balance disorders.


Asunto(s)
Envejecimiento/fisiología , Diferenciación Celular/fisiología , Células Ciliadas Vestibulares/fisiología , Factores de Transcripción SOXB1/fisiología , Animales , Recuento de Células , Linaje de la Célula/fisiología , Ratones , Ratones Noqueados , Ratones Transgénicos , Factores de Transcripción SOXB1/genética , Sáculo y Utrículo/citología
14.
Oncogene ; 38(38): 6507-6520, 2019 09.
Artículo en Inglés | MEDLINE | ID: mdl-31358900

RESUMEN

Recent genome analysis of human prostate cancers demonstrated that both AR gene amplification and TP53 mutation are among the most frequently observed alterations in advanced prostate cancer. However, the biological role of these dual genetic alterations in prostate tumorigenesis is largely unknown. In addition, there are no biologically relevant models that can be used to assess the molecular mechanisms for these genetic abnormalities. Here, we report a novel mouse model, in which elevated transgenic AR expression and Trp53 deletion occur simultaneously in mouse prostatic epithelium to mimic human prostate cancer cells. These compound mice developed an earlier onset of high-grade prostatic intraepithelial neoplasia and accelerated prostate tumors in comparison with mice harboring only the AR transgene. Histological analysis showed prostatic sarcomatoid and basaloid carcinomas with massive squamous differentiation in the above compound mice. RNA-sequencing analyses identified a robust enrichment of the signature genes for human prostatic basal cell carcinomas in the above prostate tumors. Master regulator analysis revealed SOX2 as a transcriptional regulator in prostatic basal cell tumors. Elevated expression of SOX2 and its downstream target genes were detected in prostatic tumors of the compound mice. Chromatin immunoprecipitation analyses implicate a coregulatory role of AR and SOX2 in the expression of prostatic basal cell signature genes. Our data demonstrate a critical role of SOX2 in prostate tumorigenesis and provide mechanistic insight into prostate tumor aggressiveness and progression mediated by aberrant AR and p53 signaling pathways.


Asunto(s)
Neoplasias de la Próstata/genética , Neoplasias de la Próstata/patología , Receptores Androgénicos/fisiología , Factores de Transcripción SOXB1/fisiología , Proteína p53 Supresora de Tumor/genética , Animales , Transformación Celular Neoplásica/genética , Progresión de la Enfermedad , Eliminación de Gen , Regulación Neoplásica de la Expresión Génica , Humanos , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Invasividad Neoplásica , Receptores Androgénicos/genética , Transducción de Señal/genética , Transcriptoma
15.
Development ; 146(13)2019 06 21.
Artículo en Inglés | MEDLINE | ID: mdl-31152002

RESUMEN

The transcription factor sex determining region Y-box 2 (SOX2) is required for the formation of hair cells and supporting cells in the inner ear and is a widely used sensory marker. Paradoxically, we demonstrate via fate mapping that, initially, SOX2 primarily marks nonsensory progenitors in the mouse cochlea, and is not specific to all sensory regions until late otic vesicle stages. SOX2 fate mapping reveals an apical-to-basal gradient of SOX2 expression in the sensory region of the cochlea, reflecting the pattern of cell cycle exit. To understand SOX2 function, we undertook a timed-deletion approach, revealing that early loss of SOX2 severely impaired morphological development of the ear, whereas later deletions resulted in sensory disruptions. During otocyst stages, SOX2 shifted dramatically from a lateral to medial domain over 24-48 h, reflecting the nonsensory-to-sensory switch observed by fate mapping. Early loss or gain of SOX2 function led to changes in otic epithelial volume and progenitor proliferation, impacting growth and morphological development of the ear. Our study demonstrates a novel role for SOX2 in early otic morphological development, and provides insights into the temporal and spatial patterns of sensory specification in the inner ear.


Asunto(s)
Cóclea/embriología , Oído Interno/embriología , Células Ciliadas Auditivas/fisiología , Morfogénesis/genética , Factores de Transcripción SOXB1/fisiología , Animales , Tipificación del Cuerpo/genética , Diferenciación Celular/genética , Cóclea/citología , Oído Interno/crecimiento & desarrollo , Embrión de Mamíferos , Desarrollo Embrionario/genética , Femenino , Células Ciliadas Auditivas/citología , Masculino , Ratones , Ratones Transgénicos , Embarazo , Factores de Transcripción SOXB1/genética , Factores de Tiempo
16.
Genes Cells ; 24(6): 436-448, 2019 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-31038803

RESUMEN

Lysosomes are acidic organelles responsible for degrading both exogenous and endogenous materials. The small GTPase Arl8 localizes primarily to lysosomes and is involved in lysosomal function. In the present study, using Arl8b gene-trapped mutant (Arl8b-/- ) mice, we show that Arl8b is required for the development of dorsal structures of the neural tube, including the thalamus and hippocampus. In embryonic day (E) 10.5 Arl8b-/- embryos, Sox1 (a neuroepithelium marker) was ectopically expressed in the roof plate, whereas the expression of Gdf7 and Msx1 (roof plate markers) was reduced in the dorsal midline of the midbrain. Ectopic expression of Sox1 in Arl8b-/- embryos was detected also at E9.0 in the neural fold, which gives rise to the roof plate. In addition, the levels of Bmp receptor IA and phosphorylated Smad 1/5/8 (downstream of BMP signaling) were increased in the neural fold of E9.0 Arl8b-/- embryos. These results suggest that Arl8b is involved in the development of the neural fold and the subsequently formed roof plate, possibly via control of BMP signaling.


Asunto(s)
Factores de Ribosilacion-ADP/genética , Factores de Ribosilacion-ADP/fisiología , Cresta Neural/embriología , Animales , Regulación del Desarrollo de la Expresión Génica/genética , Lisosomas/genética , Lisosomas/fisiología , Ratones/embriología , Ratones Endogámicos C57BL , Proteínas de Unión al GTP Monoméricas/metabolismo , Cresta Neural/metabolismo , Tubo Neural/embriología , Tubo Neural/metabolismo , Factores de Transcripción SOXB1/fisiología , Transducción de Señal
17.
Exp Gerontol ; 120: 21-27, 2019 06.
Artículo en Inglés | MEDLINE | ID: mdl-30822486

RESUMEN

Human aging is a physiological process characterized by a chronic low-grade inflammation. Senescence may affect endothelial cells, subsequently involved in the most common age-related diseases (ARDs), as well as mesenchymal stem cells (MSCs) with an impairment of their properties in tissues regeneration. Endothelial cells seem to be able to exert a paracrine effect on BM-MSCs through the secretion of pro-inflammatory factors. This work is aimed to evaluate if the senescent status of human umbilical vein endothelial cells (HUVECs) could affect bone marrow derived MSCs (BM-MSCs) proliferative ability and stemness. HUVECs were cultured until the senescence status. Young (passage 3) and senescent HUVECs (passage 13) were indirectly co-cultured with BM-MSCs for 8 days in order to evaluate the effect of their senescence status on proliferative ability and stemness of MSCs. The co-culture of senescent HUVECs with BM-MSCs was associated with a reduced proliferative ability of BM-MSCs, an enforced pro-inflammatory phenotype of BM-MSCs (increased synthesis of proinflammatory cytokines such as IL-6 and TNF-α) and an increased expression of miR-126a-3p, in association with a significant decrease of SOX2, a stemmness- associated gene, targeted by miR-126a-3p. A more general IPA analysis, revealed as miR-126a-3p also modulates the expression of IRS1, IRS2, IL6ST and PIK3R2, all targets that enforce the hypothesis that senescent endothelial cells may reduce the proliferative ability and the stemness phenotype of bone marrow-derived mesenchymal stem cells.


Asunto(s)
Senescencia Celular , Células Endoteliales de la Vena Umbilical Humana/fisiología , Inflamación/etiología , Células Madre Mesenquimatosas/fisiología , Factores de Transcripción SOXB1/fisiología , Proliferación Celular , Células Cultivadas , Humanos , Interleucina-6/biosíntesis , Factor de Necrosis Tumoral alfa/biosíntesis
18.
Exp Anim ; 68(1): 35-47, 2019 Feb 26.
Artículo en Inglés | MEDLINE | ID: mdl-30089733

RESUMEN

Stem cells are promising cell source for treatment of multiple diseases as well as myocardial infarction. Rabbit model has essentially used for cardiovascular diseases and regeneration but information on establishment of induced pluripotent stem cells (iPSCs) and differentiation potential is fairly limited. In addition, there is no report of cardiac differentiation from iPSCs in the rabbit model. In this study, we generated rabbit iPSCs by reprogramming rabbit fibroblasts using the 4 transcription factors (OCT3/4, SOX2, KLF4, and c-Myc). Three iPSC lines were established. The iPSCs from all cell lines expressed genes (OCT3/4, SOX2, KLF4 and NANOG) and proteins (alkaline phosphatase, OCT-3/4 and SSEA-4) essentially described for pluripotency (in vivo and in vitro differentiation). Furthermore, they also had ability to form embryoid body (EB) resulting in three-germ layer differentiation. However, ability of particular cell lines and cell numbers at seeding markedly influenced on EB formation and also their diameters. The cell density at 20,000 cells per EB was selected for cardiac differentiation. After plating, the EBs attached and cardiac-like beating areas were seen as soon as 11 days of culture. The differentiated cells expressed cardiac progenitor marker FLK1 (51 ± 1.48%) on day 5 and cardiac troponin-T protein (10.29 ± 1.37%) on day 14. Other cardiac marker genes (cardiac ryanodine receptors (RYR2), α-actinin and PECAM1) were also expressed. This study concluded that rabbit iPSCs remained their in vitro pluripotency with capability of differentiation into mature-phenotype cardiomyocytes. However, the efficiency of cardiac differentiation is still restricted.


Asunto(s)
Diferenciación Celular/genética , Diferenciación Celular/fisiología , Miocitos Cardíacos , Células Madre Pluripotentes/citología , Células Madre Pluripotentes/fisiología , Conejos , Fosfatasa Alcalina/fisiología , Animales , Línea Celular , Factor 4 Similar a Kruppel , Factores de Transcripción de Tipo Kruppel/fisiología , Proteína Homeótica Nanog/fisiología , Factor 3 de Transcripción de Unión a Octámeros/fisiología , Proteínas Proto-Oncogénicas c-myc/fisiología , Factores de Transcripción SOXB1/fisiología , Antígenos Embrionarios Específico de Estadio/fisiología
19.
Protein Cell ; 10(5): 347-364, 2019 05.
Artículo en Inglés | MEDLINE | ID: mdl-30588557

RESUMEN

Folliculogenesis is essential for production of female gametes in vertebrates. However, the molecular mechanisms underlying follicle development, particularly apoptosis regulation in ovary, remain elusive. Here, we generated sox3 knockout zebrafish lines using CRISPR/Cas9. sox3 knockout led to follicle development retardation and a reduced fecundity in females. Comparative analysis of transcriptome between sox3-/- and wild-type ovaries revealed that Sox3 was involved in pathways of ovarian steroidogenesis and apoptosis. Knockout of sox3 promoted follicle apoptosis and obvious apoptosis signals were detected in somatic cells of stages III and IV follicles of sox3-/- ovaries. Moreover, Sox3 can bind to and activate the promoter of cyp19a1a. Up-regulation of Cyp19a1a expression promoted 17ß-estradiol synthesis, which inhibited apoptosis in follicle development. Thus, Sox3 functions as a regulator of Cyp19a1a expression, via 17ß-E2 linking apoptosis suppression, which is implicated in improving female fecundity.


Asunto(s)
Estradiol/metabolismo , Fertilidad/fisiología , Folículo Ovárico/crecimiento & desarrollo , Ovario/fisiología , Factores de Transcripción SOXB1/fisiología , Pez Cebra/crecimiento & desarrollo , Animales , Apoptosis , Aromatasa/metabolismo , Femenino , Fertilidad/genética , Mutación con Pérdida de Función , Factores de Transcripción SOXB1/genética , Pez Cebra/genética , Proteínas de Pez Cebra/metabolismo
20.
Dev Cell ; 47(3): 331-347.e5, 2018 11 05.
Artículo en Inglés | MEDLINE | ID: mdl-30399335

RESUMEN

SoxB1 genes play fundamental roles in neurodevelopmental processes and maintaining stem cell multipotency, but little is known about their function in regeneration. We addressed this question by analyzing the activity of the SoxB1 homolog soxB1-2 in the planarian Schmidtea mediterranea. Expression and functional analysis revealed that soxB1-2 marks ectodermal-lineage progenitors, and its activity is required for differentiation of subsets of ciliated epidermal and neuronal cells. Moreover, we show that inhibiting soxB1-2 or its candidate target genes leads to abnormal sensory neuron regeneration that causes planarians to display seizure-like movements or phenotypes associated with the loss of sensory modalities. Our analyses highlight soxB1-2-regulated genes that are expressed in sensory neurons and are homologous to factors implicated in epileptic disorders in humans and animal models of epilepsy, indicating that planarians can serve as a complementary model to investigate genetic causes of epilepsy.


Asunto(s)
Planarias/metabolismo , Factores de Transcripción SOXB1/metabolismo , Células Receptoras Sensoriales/metabolismo , Animales , Diferenciación Celular/genética , Proliferación Celular , Cilios/fisiología , Regeneración Nerviosa/genética , Regeneración Nerviosa/fisiología , Planarias/fisiología , Interferencia de ARN , Regeneración/fisiología , Factores de Transcripción SOXB1/genética , Factores de Transcripción SOXB1/fisiología , Células Receptoras Sensoriales/fisiología , Células Madre/citología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA