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.
Artigo em Inglês | MEDLINE | ID: mdl-34890603

RESUMO

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.


Assuntos
Via de Sinalização Hippo/fisiologia , Células-Tronco Neoplásicas/patologia , Neoplasias da Retina/patologia , Retinoblastoma/patologia , Fatores de Transcrição SOXB1/fisiologia , Proteínas de Sinalização YAP/fisiologia , Proteínas Adaptadoras de Transdução de Sinal/genética , Animais , Western Blotting , Feminino , Humanos , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Nus , Proteínas de Neoplasias/metabolismo , Células-Tronco Neoplásicas/metabolismo , Reação em Cadeia da Polimerase em Tempo Real , Neoplasias da Retina/metabolismo , Retinoblastoma/metabolismo , Fatores de Transcrição/genética , Transplante Heterólogo , Células Tumorais Cultivadas
3.
Brain Res ; 1752: 147265, 2021 02 01.
Artigo em Inglês | MEDLINE | ID: mdl-33422527

RESUMO

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.


Assuntos
Células Amácrinas/fisiologia , Retina/crescimento & desenvolvimento , Células Bipolares da Retina/fisiologia , Fatores de Transcrição SOXB1/fisiologia , Animais , Animais Recém-Nascidos , Feminino , Regulação da Expressão Gênica no Desenvolvimento , Masculino , Camundongos
4.
Int J Mol Sci ; 23(1)2021 Dec 26.
Artigo em Inglês | MEDLINE | ID: mdl-35008655

RESUMO

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.


Assuntos
Diferenciação Celular , Desenvolvimento Embrionário , Neurônios/metabolismo , Fatores de Transcrição SOXB1/metabolismo , Núcleo Supraquiasmático/metabolismo , Animais , Ritmo Circadiano , Camundongos , Neurônios/fisiologia , Fatores de Transcrição SOXB1/fisiologia , Núcleo Supraquiasmático/crescimento & desenvolvimento , Núcleo Supraquiasmático/fisiologia
5.
J Neurosci Res ; 99(2): 679-698, 2021 02.
Artigo em Inglês | MEDLINE | ID: mdl-33099767

RESUMO

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.


Assuntos
Audição/fisiologia , Neuroglia/fisiologia , Gânglio Espiral da Cóclea/citologia , Potenciais de Ação , Animais , Bário/farmacologia , Células Cultivadas , Nervo Coclear/fisiologia , Desipramina/farmacologia , Feminino , Transporte de Íons , Masculino , Potenciais da Membrana , Camundongos , Camundongos Endogâmicos C57BL , Bainha de Mielina/fisiologia , Neuritos/ultraestrutura , Neurônios Aferentes/fisiologia , Técnicas de Patch-Clamp , Canais de Potássio Corretores do Fluxo de Internalização/fisiologia , Fatores de Transcrição SOXB1/fisiologia
6.
J Hematol Oncol ; 13(1): 119, 2020 09 03.
Artigo em Inglês | MEDLINE | ID: mdl-32883331

RESUMO

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.


Assuntos
Carcinoma/secundário , Neoplasias Colorretais/patologia , Neoplasias Hepáticas/secundário , Modelos Biológicos , Invasividade Neoplásica/patologia , Organoides , Animais , Biomarcadores Tumorais , Carcinoma/patologia , Progressão da Doença , Perfilação da Expressão Gênica , Regulação Neoplásica da Expressão Gênica , Xenoenxertos , Humanos , Neoplasias Hepáticas/patologia , Camundongos , Proteínas de Neoplasias/biossíntese , Proteínas de Neoplasias/genética , Proteínas de Neoplasias/fisiologia , Organoides/efeitos dos fármacos , Organoides/patologia , Organoides/transplante , Fatores de Transcrição SOXB1/biossíntese , Fatores de Transcrição SOXB1/genética , Fatores de Transcrição SOXB1/fisiologia
7.
Science ; 369(6502): 397-403, 2020 07 24.
Artigo em Inglês | MEDLINE | ID: mdl-32703873

RESUMO

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.


Assuntos
Diferenciação Celular , Autofagia Mediada por Chaperonas , Células-Tronco Embrionárias/fisiologia , Animais , Linhagem Celular , Epigênese Genética , Histonas/fisiologia , Ácidos Cetoglutáricos/metabolismo , Camundongos , Fator 3 de Transcrição de Octâmero/fisiologia , Fatores de Transcrição SOXB1/fisiologia
8.
Cell Rep ; 31(2): 107511, 2020 04 14.
Artigo em Inglês | MEDLINE | ID: mdl-32294450

RESUMO

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.


Assuntos
Meduloblastoma/etiologia , Meduloblastoma/metabolismo , Fatores de Transcrição SOXB1/metabolismo , Animais , Linhagem da Célula/genética , Células Cultivadas , Neoplasias Cerebelares/patologia , Cerebelo/embriologia , Feminino , Proteínas Hedgehog/metabolismo , Humanos , Masculino , Camundongos Knockout , Camundongos Transgênicos , Recidiva Local de Neoplasia/patologia , Células-Tronco Neurais/metabolismo , Neurogênese , Neurônios/metabolismo , Fatores de Transcrição SOXB1/fisiologia , Transdução de Sinais/fisiologia , Análise de Célula Única/métodos
9.
Proc Natl Acad Sci U S A ; 117(12): 6942-6950, 2020 03 24.
Artigo em Inglês | MEDLINE | ID: mdl-32139605

RESUMO

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.


Assuntos
Diferenciação Celular , Células-Tronco Embrionárias/fisiologia , Regulação da Expressão Gênica no Desenvolvimento , Redes Reguladoras de Genes , MicroRNAs/genética , Animais , Proteínas Argonautas/fisiologia , Células-Tronco Embrionárias/citologia , Perfilação da Expressão Gênica , Camundongos , Camundongos Knockout , Proteína Homeobox Nanog/fisiologia , Proteínas de Ligação a RNA/fisiologia , Fatores de Transcrição SOXB1/fisiologia , Transdução de Sinais
10.
Cancer Res ; 80(8): 1644-1655, 2020 04 15.
Artigo em Inglês | MEDLINE | ID: mdl-32094299

RESUMO

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.


Assuntos
Neoplasias Encefálicas/patologia , Sobrevivência Celular , Proteína Forkhead Box O1/genética , Genes Supressores de Tumor , Glioblastoma/patologia , MicroRNAs/metabolismo , Proteínas de Neoplasias/fisiologia , PTEN Fosfo-Hidrolase/genética , Fatores de Transcrição SOXB1/fisiologia , Animais , Proteína 11 Semelhante a Bcl-2/metabolismo , Neoplasias Encefálicas/genética , Neoplasias Encefálicas/metabolismo , Neoplasias Encefálicas/radioterapia , Morte Celular , Desdiferenciação Celular/genética , Linhagem Celular Tumoral , Reprogramação Celular/fisiologia , Repressão Epigenética , Feminino , Regulação Neoplásica da Expressão Gênica , Glioblastoma/genética , Glioblastoma/metabolismo , Glioblastoma/radioterapia , Xenoenxertos , Humanos , Camundongos , Camundongos Nus , MicroRNAs/administração & dosagem , MicroRNAs/antagonistas & inibidores , MicroRNAs/genética , Nanopartículas/administração & dosagem , Proteínas de Neoplasias/genética , Células-Tronco Neoplásicas/metabolismo , Células-Tronco Neoplásicas/patologia , Células-Tronco Neoplásicas/efeitos da radiação , Células-Tronco Neurais , Fator 3 de Transcrição de Octâmero/metabolismo , Tolerância a Radiação , Distribuição Aleatória , Fatores de Transcrição SOXB1/genética , Fatores de Transcrição SOXB1/metabolismo , Transfecção/métodos , Carga Tumoral , Ensaio Tumoral de Célula-Tronco/métodos , Regulação para Cima
11.
Dev Biol ; 461(2): 160-171, 2020 05 15.
Artigo em Inglês | MEDLINE | ID: mdl-32059837

RESUMO

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.


Assuntos
Autorrenovação Celular/fisiologia , Galinhas/genética , Células-Tronco Neurais/citologia , Tubo Neural/embriologia , Neurulação/fisiologia , Fatores de Transcrição SOXB1/fisiologia , Cauda/embriologia , Animais , Linhagem da Célula , Embrião de Galinha , Genes Reporter , Mesoderma/citologia , Tubo Neural/citologia , Neurulação/genética , Fatores de Transcrição SOXB1/antagonistas & inibidores , Fatores de Transcrição SOXB1/genética , Cauda/citologia
12.
Pathol Oncol Res ; 26(2): 693-706, 2020 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-30712195

RESUMO

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.


Assuntos
Algoritmos , Biologia Computacional , Fatores de Transcrição SOXB1/fisiologia , Humanos
13.
Neuroscience ; 422: 146-160, 2019 12 01.
Artigo em Inglês | MEDLINE | ID: mdl-31678344

RESUMO

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.


Assuntos
Envelhecimento/fisiologia , Diferenciação Celular/fisiologia , Células Ciliadas Vestibulares/fisiologia , Fatores de Transcrição SOXB1/fisiologia , Animais , Contagem de Células , Linhagem da Célula/fisiologia , Camundongos , Camundongos Knockout , Camundongos Transgênicos , Fatores de Transcrição SOXB1/genética , Sáculo e Utrículo/citologia
14.
Oncogene ; 38(38): 6507-6520, 2019 09.
Artigo em Inglês | MEDLINE | ID: mdl-31358900

RESUMO

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.


Assuntos
Neoplasias da Próstata/genética , Neoplasias da Próstata/patologia , Receptores Androgênicos/fisiologia , Fatores de Transcrição SOXB1/fisiologia , Proteína Supressora de Tumor p53/genética , Animais , Transformação Celular Neoplásica/genética , Progressão da Doença , Deleção de Genes , Regulação Neoplásica da Expressão Gênica , Humanos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Invasividade Neoplásica , Receptores Androgênicos/genética , Transdução de Sinais/genética , Transcriptoma
15.
Development ; 146(13)2019 06 21.
Artigo em Inglês | MEDLINE | ID: mdl-31152002

RESUMO

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.


Assuntos
Cóclea/embriologia , Orelha Interna/embriologia , Células Ciliadas Auditivas/fisiologia , Morfogênese/genética , Fatores de Transcrição SOXB1/fisiologia , Animais , Padronização Corporal/genética , Diferenciação Celular/genética , Cóclea/citologia , Orelha Interna/crescimento & desenvolvimento , Embrião de Mamíferos , Desenvolvimento Embrionário/genética , Feminino , Células Ciliadas Auditivas/citologia , Masculino , Camundongos , Camundongos Transgênicos , Gravidez , Fatores de Transcrição SOXB1/genética , Fatores de Tempo
16.
Genes Cells ; 24(6): 436-448, 2019 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-31038803

RESUMO

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.


Assuntos
Fatores de Ribosilação do ADP/genética , Fatores de Ribosilação do ADP/fisiologia , Crista Neural/embriologia , Animais , Regulação da Expressão Gênica no Desenvolvimento/genética , Lisossomos/genética , Lisossomos/fisiologia , Camundongos/embriologia , Camundongos Endogâmicos C57BL , Proteínas Monoméricas de Ligação ao GTP/metabolismo , Crista Neural/metabolismo , Tubo Neural/embriologia , Tubo Neural/metabolismo , Fatores de Transcrição SOXB1/fisiologia , Transdução de Sinais
17.
Exp Gerontol ; 120: 21-27, 2019 06.
Artigo em Inglês | MEDLINE | ID: mdl-30822486

RESUMO

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.


Assuntos
Senescência Celular , Células Endoteliais da Veia Umbilical Humana/fisiologia , Inflamação/etiologia , Células-Tronco Mesenquimais/fisiologia , Fatores de Transcrição SOXB1/fisiologia , Proliferação de Células , Células Cultivadas , Humanos , Interleucina-6/biossíntese , Fator de Necrose Tumoral alfa/biossíntese
18.
Exp Anim ; 68(1): 35-47, 2019 Feb 26.
Artigo em Inglês | MEDLINE | ID: mdl-30089733

RESUMO

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.


Assuntos
Diferenciação Celular/genética , Diferenciação Celular/fisiologia , Miócitos Cardíacos , Células-Tronco Pluripotentes/citologia , Células-Tronco Pluripotentes/fisiologia , Coelhos , Fosfatase Alcalina/fisiologia , Animais , Linhagem Celular , Fator 4 Semelhante a Kruppel , Fatores de Transcrição Kruppel-Like/fisiologia , Proteína Homeobox Nanog/fisiologia , Fator 3 de Transcrição de Octâmero/fisiologia , Proteínas Proto-Oncogênicas c-myc/fisiologia , Fatores de Transcrição SOXB1/fisiologia , Antígenos Embrionários Estágio-Específicos/fisiologia
19.
Protein Cell ; 10(5): 347-364, 2019 05.
Artigo em Inglês | MEDLINE | ID: mdl-30588557

RESUMO

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.


Assuntos
Estradiol/metabolismo , Fertilidade/fisiologia , Folículo Ovariano/crescimento & desenvolvimento , Ovário/fisiologia , Fatores de Transcrição SOXB1/fisiologia , Peixe-Zebra/crescimento & desenvolvimento , Animais , Apoptose , Aromatase/metabolismo , Feminino , Fertilidade/genética , Mutação com Perda de Função , Fatores de Transcrição SOXB1/genética , Peixe-Zebra/genética , Proteínas de Peixe-Zebra/metabolismo
20.
Dev Cell ; 47(3): 331-347.e5, 2018 11 05.
Artigo em Inglês | MEDLINE | ID: mdl-30399335

RESUMO

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.


Assuntos
Planárias/metabolismo , Fatores de Transcrição SOXB1/metabolismo , Células Receptoras Sensoriais/metabolismo , Animais , Diferenciação Celular/genética , Proliferação de Células , Cílios/fisiologia , Regeneração Nervosa/genética , Regeneração Nervosa/fisiologia , Planárias/fisiologia , Interferência de RNA , Regeneração/fisiologia , Fatores de Transcrição SOXB1/genética , Fatores de Transcrição SOXB1/fisiologia , Células Receptoras Sensoriais/fisiologia , Células-Tronco/citologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...