Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 89
Filtrar
Más filtros

Tipo del documento
Intervalo de año de publicación
1.
Development ; 149(5)2022 03 01.
Artículo en Inglés | MEDLINE | ID: mdl-35262177

RESUMEN

Axonal projections from layer V neurons of distinct neocortical areas are topographically organized into discrete clusters within the pontine nuclei during the establishment of voluntary movements. However, the molecular determinants controlling corticopontine connectivity are insufficiently understood. Here, we show that an intrinsic cortical genetic program driven by Nr2f1 graded expression is directly implicated in the organization of corticopontine topographic mapping. Transgenic mice lacking cortical expression of Nr2f1 and exhibiting areal organization defects were used as model systems to investigate the arrangement of corticopontine projections. By combining three-dimensional digital brain atlas tools, Cre-dependent mouse lines and axonal tracing, we show that Nr2f1 expression in postmitotic neurons spatially and temporally controls somatosensory topographic projections, whereas expression in progenitor cells influences the ratio between corticopontine and corticospinal fibres passing the pontine nuclei. We conclude that cortical gradients of area-patterning genes are directly implicated in the establishment of a topographic somatotopic mapping from the cortex onto pontine nuclei.


Asunto(s)
Mapeo Encefálico , Puente , Animales , Axones , Corteza Cerebral , Ratones , Vías Nerviosas/fisiología , Neuronas , Puente/fisiología
2.
Neurobiol Dis ; 199: 106604, 2024 Jul 11.
Artículo en Inglés | MEDLINE | ID: mdl-39002810

RESUMEN

Mitochondria are essential regulators of cellular energy metabolism and play a crucial role in the maintenance and function of neuronal cells. Studies in the last decade have highlighted the importance of mitochondrial dynamics and bioenergetics in adult neurogenesis, a process that significantly influences cognitive function and brain plasticity. In this review, we examine the mechanisms by which mitochondria regulate adult neurogenesis, focusing on the impact of mitochondrial function on the behavior of neural stem/progenitor cells and the maturation and plasticity of newborn neurons in the adult mouse hippocampus. In addition, we explore the link between mitochondrial dysfunction, adult hippocampal neurogenesis and genes associated with cognitive deficits in neurodevelopmental disorders. In particular, we provide insights into how alterations in the transcriptional regulator NR2F1 affect mitochondrial dynamics and may contribute to the pathophysiology of the emerging neurodevelopmental disorder Bosch-Boonstra-Schaaf optic atrophy syndrome (BBSOAS). Understanding how genes involved in embryonic and adult neurogenesis affect mitochondrial function in neurological diseases might open new directions for therapeutic interventions aimed at boosting mitochondrial function during postnatal life.

3.
Neurobiol Dis ; 193: 106455, 2024 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-38408685

RESUMEN

White matter (WM) tract formation and axonal pathfinding are major processes in brain development allowing to establish precise connections between targeted structures. Disruptions in axon pathfinding and connectivity impairments will lead to neural circuitry abnormalities, often associated with various neurodevelopmental disorders (NDDs). Among several neuroimaging methodologies, Diffusion Tensor Imaging (DTI) is a magnetic resonance imaging (MRI) technique that has the advantage of visualizing in 3D the WM tractography of the whole brain non-invasively. DTI is particularly valuable in unpinning structural tract connectivity defects of neural networks in NDDs. In this study, we used 3D DTI to unveil brain-specific tract defects in two mouse models lacking the Nr2f1 gene, which mutations in patients have been proven to cause an emerging NDD, called Bosch-Boonstra-Schaaf Optic Atrophy (BBSOAS). We aimed to investigate the impact of the lack of cortical Nr2f1 function on WM morphometry and tract microstructure quantifications. We found in both mutant mice partial loss of fibers and severe misrouting of the two major cortical commissural tracts, the corpus callosum, and the anterior commissure, as well as the two major hippocampal efferent tracts, the post-commissural fornix, and the ventral hippocampal commissure. DTI tract malformations were supported by 2D histology, 3D fluorescent imaging, and behavioral analyses. We propose that these interhemispheric connectivity impairments are consistent in explaining some cognitive defects described in BBSOAS patients, particularly altered information processing between the two brain hemispheres. Finally, our results highlight 3DDTI as a relevant neuroimaging modality that can provide appropriate morphometric biomarkers for further diagnosis of BBSOAS patients.


Asunto(s)
Atrofia Óptica , Sustancia Blanca , Humanos , Ratones , Animales , Imagen de Difusión Tensora , Sustancia Blanca/diagnóstico por imagen , Sustancia Blanca/patología , Encéfalo , Imagen por Resonancia Magnética , Atrofia Óptica/patología
4.
EMBO J ; 39(13): e104163, 2020 07 01.
Artículo en Inglés | MEDLINE | ID: mdl-32484994

RESUMEN

The relationships between impaired cortical development and consequent malformations in neurodevelopmental disorders, as well as the genes implicated in these processes, are not fully elucidated to date. In this study, we report six novel cases of patients affected by BBSOAS (Boonstra-Bosch-Schaff optic atrophy syndrome), a newly emerging rare neurodevelopmental disorder, caused by loss-of-function mutations of the transcriptional regulator NR2F1. Young patients with NR2F1 haploinsufficiency display mild to moderate intellectual disability and show reproducible polymicrogyria-like brain malformations in the parietal and occipital cortex. Using a recently established BBSOAS mouse model, we found that Nr2f1 regionally controls long-term self-renewal of neural progenitor cells via modulation of cell cycle genes and key cortical development master genes, such as Pax6. In the human fetal cortex, distinct NR2F1 expression levels encompass gyri and sulci and correlate with local degrees of neurogenic activity. In addition, reduced NR2F1 levels in cerebral organoids affect neurogenesis and PAX6 expression. We propose NR2F1 as an area-specific regulator of mouse and human brain morphology and a novel causative gene of abnormal gyrification.


Asunto(s)
Factor de Transcripción COUP I/metabolismo , Neocórtex/embriología , Células-Madre Neurales/metabolismo , Lóbulo Occipital/embriología , Atrofias Ópticas Hereditarias/embriología , Lóbulo Parietal/embriología , Animales , Factor de Transcripción COUP I/genética , Modelos Animales de Enfermedad , Humanos , Ratones , Neocórtex/patología , Células-Madre Neurales/patología , Lóbulo Occipital/patología , Atrofias Ópticas Hereditarias/genética , Atrofias Ópticas Hereditarias/patología , Factor de Transcripción PAX6/genética , Factor de Transcripción PAX6/metabolismo , Lóbulo Parietal/patología
5.
J Med Virol ; 95(4): e28749, 2023 04.
Artículo en Inglés | MEDLINE | ID: mdl-37185850

RESUMEN

Hepatitis B Virus (HBV) replication has been reported to be restricted by the intrahepatic host restriction factors and antiviral signaling pathways. The intracellular mechanisms underlying the significant viremia difference among different phases of the natural history chronic HBV infection remain elusive. We herein report that the hypoxia-induced gene domain protein-1a (HIGD1A) was highly expressed in the liver of inactive HBV carriers with low viremia. Ectopic expression of HIGD1A in hepatocyte-derived cells significantly inhibited HBV transcription and replication in a dose-dependent manner, while silence of HIGD1A promoted HBV gene expression and replication. Similar results were also observed in both de novo HBV-infected cell culture model and HBV persistence mouse model. Mechanistically, HIGD1A is located on the mitochondrial inner membrane and activates nuclear factor kappa B (NF-κB) signaling pathway through binding to paroxysmal nonkinesigenic dyskinesia (PNKD), which further enhances the expression of a transcription factor NR2F1 to inhibit HBV transcription and replication. Consistently, knockdown of PNKD or NR2F1 and blockage of NF-κB signaling pathway abrogated the inhibitory effect of HIGD1A on HBV replication. Mitochondrial HIGD1A exploits the PNKD-NF-κB-NR2F1 nexus to act as a host restriction factor of HBV infection. Our study thus shed new lights on the regulation of HBV by hypoxia-related genes and related antiviral strategies.


Asunto(s)
Virus de la Hepatitis B , Hepatitis B , Animales , Ratones , Antivirales/farmacología , Virus de la Hepatitis B/fisiología , FN-kappa B/genética , FN-kappa B/metabolismo , Transcripción Viral , Viremia , Replicación Viral , Humanos
6.
Cytokine ; 162: 156105, 2023 02.
Artículo en Inglés | MEDLINE | ID: mdl-36527891

RESUMEN

BACKGROUND: The long noncoding RNAs' (lncRNAs) effect on cancer therapy resistance by targeting microRNA (miRNA) in the regulation of drug resistance genes has attracted more and more attention. This study attempted to explore the mechanism of "lncRNA NR2F1-AS1/miR-483-3p/IGF1″ axis in azacitidine resistance of THP-1 cells. METHODS: THP-1 cells were treated with azacitidine to construct THP1-Aza cells. Cell number and morphological changes were observed by a microscope. CCK8, flow cytometry and transwell were used to detect cell proliferation, apoptosis, cycle, invasion and migration. The targeting relationships between NR2F1-AS1 and miR-483-3p, IGF1 and miR-483-3p were analyzed by dual-luciferase, respectively. RIP assay was applied to verify the interaction between NR2F1-AS1 and miR-483-3p. The relative mRNA expression levels of miR-483-3p, AKT1, PI3K, NR2F1-AS1 and IGF1 were detected by qRT-PCR. PI3K, p-PI3K, AKT, p-AKT and IGF1 protein expression were detected by western blot. RESULTS: Compared with THP-1 cells, NR2F1-AS1 and IGF1 were highly expressed in THP1-Aza cells, and the miR-483-3p expression was significantly decreased in THP1-Aza cells. Knockdown of NR2F1-AS1 increased apoptosis and G1 phase, and reduced cells growth, invasion and migration ability of THP1-Aza cells. Dual-luciferase demonstrated that NR2F1-AS1 could bind to miR-483-3p, and miR-483-3p could bind to IGF1. RIP assay verified the interaction between NR2F1-AS1 and miR-483-3p. Compared with the si-NR2F1-AS1 group, miR-483-3p inhibitor or oe-IGF1 treatment reduced the apoptosis and cell cycle, and increased the cell growth, invasion and migration ability of THP-1-Aza cells. CONCLUSION: LncRNA NR2F1-AS1 affects the sensitivity of THP-1 cells to azacitidine resistance by regulating the miR-483-3p/IGF1 axis, which may be a potential target for the treatment of acute monocytic leukemia.


Asunto(s)
MicroARNs , ARN Largo no Codificante , Humanos , ARN Largo no Codificante/genética , ARN Largo no Codificante/metabolismo , Células THP-1 , Proteínas Proto-Oncogénicas c-akt/metabolismo , Línea Celular Tumoral , MicroARNs/genética , MicroARNs/metabolismo , Proliferación Celular/genética , Fosfatidilinositol 3-Quinasas/genética , Fosfatidilinositol 3-Quinasas/metabolismo , Movimiento Celular/genética , Regulación Neoplásica de la Expresión Génica
7.
Cereb Cortex ; 32(17): 3611-3632, 2022 08 22.
Artículo en Inglés | MEDLINE | ID: mdl-34963132

RESUMEN

The generation and differentiation of cortical projection neurons are extensively regulated by interactive programs of transcriptional factors. Here, we report the cooperative functions of transcription factors Bcl11a and Bcl11b in regulating the development of cortical projection neurons. Among the cells derived from the cortical neural stem cells, Bcl11a is expressed in the progenitors and the projection neurons, while Bcl11b expression is restricted to the projection neurons. Using conditional knockout mice, we show that deficiency of Bcl11a leads to reduced proliferation and precocious differentiation of cortical progenitor cells, which is exacerbated when Bcl11b is simultaneously deleted. Besides defective neuronal production, the differentiation of cortical projection neurons is blocked in the absence of both Bcl11a and Bcl11b: Expression of both pan-cortical and subtype-specific genes is reduced or absent; axonal projections to the thalamus, hindbrain, spinal cord, and contralateral cortical hemisphere are reduced or absent. Furthermore, neurogenesis-to-gliogenesis switch is accelerated in the Bcl11a-CKO and Bcl11a/b-DCKO mice. Bcl11a likely regulates neurogenesis through repressing the Nr2f1 expression. These results demonstrate that Bcl11a and Bcl11b jointly play critical roles in the generation and differentiation of cortical projection neurons and in controlling the timing of neurogenesis-to-gliogenesis switch.


Asunto(s)
Células-Madre Neurales , Factores de Transcripción , Animales , Diferenciación Celular/fisiología , Ratones , Ratones Noqueados , Células-Madre Neurales/metabolismo , Neurogénesis/fisiología , Neuronas/fisiología , Proteínas Represoras/genética , Proteínas Represoras/metabolismo , Factores de Transcripción/genética , Factores de Transcripción/metabolismo , Proteínas Supresoras de Tumor/metabolismo
8.
Int J Med Sci ; 20(9): 1152-1162, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-37575267

RESUMEN

Background: Long noncoding RNAs (lncRNAs) substantially affect tumor metastasis and are aberrantly expressed in various cancers. However, its role in breast cancer (BC) remains unclear. Methods: A microarray assay of differentially expressed lncRNAs in epithelial-mesenchymal transition (EMT) and non-EMT cells was performed. The prognostic value of lnc NR2F1-AS1 expression in patients with BC was analyzed using The Cancer Genome Atlas database. Lnc NR2F1-AS1 expression levels in different BC cell lines were assessed using quantitative real-time PCR. The role of lnc NR2F1-AS1 in BC cell metastasis was investigated in vitro and in vivo. Dual luciferase reporter assay and RNA immunoprecipitation were performed to investigate the relationship between lnc NR2F1-AS1, miR-25-3p, and ZEB2. Results: High levels of lnc NR2F1-AS1 were observed in BC cells undergoing EMT and were closely correlated with adverse prognosis in patients with BC. Lnc NR2F1-AS1 knockdown significantly inhibited BC cell migration, invasiveness in vitro, and metastasis in vivo. Mechanistically, lnc NR2F1-AS1 competitively binds to miR-25-3p to impede ZEB2 degradation, a positive EMT transcription factor in BC. Conclusions: Our study revealed a novel lnc NR2F1-AS1/miR-25-3p/ZEB2 axis in BC metastasis and that lnc NR2F1-AS1 may serve as a potential therapeutic target for BC metastasis.


Asunto(s)
Neoplasias de la Mama , MicroARNs , Neoplasias Primarias Secundarias , ARN Largo no Codificante , Humanos , Femenino , MicroARNs/genética , MicroARNs/metabolismo , Neoplasias de la Mama/patología , ARN Largo no Codificante/genética , ARN Largo no Codificante/metabolismo , Línea Celular Tumoral , Células MCF-7 , Proliferación Celular/genética , Movimiento Celular/genética , Regulación Neoplásica de la Expresión Génica/genética , Factor de Transcripción COUP I/genética , Factor de Transcripción COUP I/metabolismo , Caja Homeótica 2 de Unión a E-Box con Dedos de Zinc/genética , Caja Homeótica 2 de Unión a E-Box con Dedos de Zinc/metabolismo , Melanoma Cutáneo Maligno
9.
Hum Mutat ; 43(2): 128-142, 2022 02.
Artículo en Inglés | MEDLINE | ID: mdl-34837429

RESUMEN

Pathogenic variants of the nuclear receptor subfamily 2 group F member 1 gene (NR2F1) are responsible for Bosch-Boonstra-Schaaf optic atrophy syndrome (BBSOAS), an autosomal dominant disorder characterized by optic atrophy associated with developmental delay and intellectual disability, but with a clinical presentation which appears to be multifaceted. We created the first public locus-specific database dedicated to NR2F1. All variants and clinical cases reported in the literature, as well as new unpublished cases, were integrated into the database using standard nomenclature to describe both molecular and phenotypic anomalies. We subsequently pursued a comprehensive approach based on computed representation and analysis suggesting a refinement of the BBSOAS clinical description with respect to neurological features and the inclusion of additional signs of hypotonia and feeding difficulties. This database is fully accessible for both clinician and molecular biologists and should prove useful in further refining the clinical synopsis of NR2F1 as new data is recorded.


Asunto(s)
Factor de Transcripción COUP I , Bases de Datos Genéticas , Discapacidad Intelectual , Atrofias Ópticas Hereditarias , Atrofia Óptica , Factor de Transcripción COUP I/genética , Humanos , Discapacidad Intelectual/genética , Hipotonía Muscular/genética , Atrofias Ópticas Hereditarias/genética , Atrofia Óptica/diagnóstico , Atrofia Óptica/genética
10.
Biochem Biophys Res Commun ; 604: 123-129, 2022 05 14.
Artículo en Inglés | MEDLINE | ID: mdl-35303678

RESUMEN

Many regulators controlling arterial identity are well described; however, transcription factors that promote vein identity and vascular patterning have remained largely unknown. We previously identified the transcription factors Islet2 (Isl2) and Nr2f1b required for specification of the vein and tip cell identity mediated by notch pathway in zebrafish. However, the interaction between Isl2 and Nr2f1b is not known. In this study, we report that Nr2f2 plays minor roles on vein and intersegmental vessels (ISV) growth and dissect the genetic interactions among the three transcription factors Isl2, Nr2f1b, and Nr2f2 using a combinatorial knockdown strategy. The double knockdown of isl2/nr2f1b, isl2/nr2f2, and nr2f1b/nr2f2 showed the enhanced defects in vasculature including less completed ISV, reduced veins, and ISV cells. We further tested the genetic relationship among these three transcription factors. We found isl2 can regulate the expression of nr2f1b and nr2f2, suggesting a model where Isl2 functions upstream of Nr2f1b and Nr2f2. We hypothsized that Isl2 and Nr2f1b can function together through cis-regulatory binding motifs. In-vitro luciferase assay results, we showed that Isl2 and Nr2f1b can cooperatively enhance gene expression. Moreover, co-immunoprecipitation results indicated that Isl2 and Nr2f1b interact physically. Together, we showed that the interaction of the Nr2f1b and Nr2f2 transcription factors in combination with the Islet2 play coordinated roles in the vascular development of zebrafish.


Asunto(s)
Arterias , Proteínas con Homeodominio LIM , Factores de Transcripción , Proteínas de Pez Cebra , Pez Cebra , Animales , Arterias/crecimiento & desarrollo , Proteínas con Homeodominio LIM/genética , Proteínas con Homeodominio LIM/metabolismo , Factores de Transcripción/genética , Factores de Transcripción/metabolismo , Venas , Pez Cebra/genética , Pez Cebra/crecimiento & desarrollo , Proteínas de Pez Cebra/genética , Proteínas de Pez Cebra/metabolismo
11.
Exp Eye Res ; 219: 109069, 2022 06.
Artículo en Inglés | MEDLINE | ID: mdl-35398206

RESUMEN

This study aimed to investigate whether dysregulation of NR2F1-AS1 is associated with the T2D onset and DR progression. A total of 269 individuals diagnosed with T2D were recruited in this study, including 111 individuals with uncomplicated T2D and 158 individuals with nonproliferative diabetic retinopathy (NPDR). A total of 48 among the NPDR individuals developed into proliferative diabetic retinopathy (PDR), excepting that 6 individuals were dropped out. RT-qPCR was used to determine the expression of NR2F1-AS1 level in blood. The correlations between NR2F1-AS1 expression and other parameters were tested by Pearson correlation. Receiver operating characteristic (ROC) curves were plotted for evaluating the diagnostic value of NR2F1-AS1 as a biochemical indicator detecting T2D and PDR. Kaplan-Meier method and Cox multivariable analysis was used to evaluate the predictive power of NR2F1-AS1 for incidence of PDR. A significant increase in blood NR2F1-AS1 in uncomplicated T2D, NPDR, and PDR patients was found when compared to healthy subjects. Significant correlations between NR2F1-AS1 expression and the level of fasting glucose, 2-h postprandial blood glucose, and HbA1c were revealed. Furthermore, NR2F1-AS1 represented distinguishing ability in T2D individuals from healthy subjects and PDR individuals from NPDR individuals. NR2F1-AS1 also showed strong predictive ability for PDR. The examination of blood NR2F1-AS1 may be used as a noninvasive biomarker for screening of T2D and early diagnosis of DR. NR2F1-AS1 may also be used as a promising novel biomarker for prediction PDR. NR2F1-AS1 may play a role in the progression of DR by moderating EndMT.


Asunto(s)
Diabetes Mellitus Tipo 2 , Retinopatía Diabética , ARN Largo no Codificante , Biomarcadores , Factor de Transcripción COUP I , Diabetes Mellitus Tipo 2/complicaciones , Diabetes Mellitus Tipo 2/diagnóstico , Diabetes Mellitus Tipo 2/genética , Retinopatía Diabética/diagnóstico , Retinopatía Diabética/genética , Humanos , ARN Largo no Codificante/genética , Curva ROC
12.
Am J Med Genet A ; 188(3): 900-906, 2022 03.
Artículo en Inglés | MEDLINE | ID: mdl-34787370

RESUMEN

Bosch-Boonstra-Schaaf optic atrophy syndrome (BBSOAS) is a rare congenital syndrome characterized by a range of phenotypes including optic atrophy and intellectual disability among other features. Pathogenic variants in the NR2F1 (nuclear receptor subfamily 2 group F member 1) gene have been linked to this condition. A recent report has shown that pathogenic variants in the start codon lead to decreased expression of the NR2F1 protein and a relatively mild phenotype, similar to that seen in whole gene deletions, and due to the lack of the dominant negative effect. Here we describe a severe case of BBSOAS with an initiation codon missense variant. The developmental delay, seizures, optic atrophy are in keeping with features observed in this condition, however this is the first report to describe colobomas and septo-optic dysplasia as associated features potentially extending the phenotype linked to BBSOAS. In addition, this is the first description of a severe phenotype linked to a de novo missense variant in the start codon of the NR2F1 gene.


Asunto(s)
Coloboma , Discapacidad Intelectual , Atrofias Ópticas Hereditarias , Atrofia Óptica , Displasia Septo-Óptica , Factor de Transcripción COUP I/genética , Codón Iniciador , Coloboma/genética , Humanos , Discapacidad Intelectual/genética , Atrofias Ópticas Hereditarias/genética , Atrofia Óptica/diagnóstico , Atrofia Óptica/genética , Displasia Septo-Óptica/diagnóstico , Displasia Septo-Óptica/genética
13.
Rev Invest Clin ; 74(4): 181-192, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-36087939

RESUMEN

Background: Cervical cancer (CC), as a serious menace to the health of women, has long been one of the most lethal gynecologic neoplasms throughout the world. Long non-coding RNA (LncRNA) NR2F1-AS1 has been documented to exert crucial functions in many malignant tumors. Nonetheless, the function and molecular mechanism of NR2F1-AS1 in CC remain completely unknown. Objectives: This study aimed to explore the function and molecular mechanism of NR2F1-AS1 in CC. Methods: The expression levels of NR2F1-AS1, miR-642a-3p, NR2F1 in CC tissues, and cell lines were examined by reverse transcription real-time quantitative polymerase chain reaction. Cell viability, proliferation, migration, and invasion were detected by 3-(4,5-dimethyl-2-thiazolyl)-2,5-diphenyl-2-H-tetrazolium bromide, colony formation and Transwell assays. The protein levels of epithelial-mesenchymal transition markers and NR2F1 in CC cells were assessed by Western blot analysis. The correlations among NR2F1-AS1, miR-642a-3p, and NR2F1 were estimated through luciferase reporter and RNA immunoprecipitation assays. Results: NR2F1-AS1 expression was clearly downregulated in CC tissues and cell lines. Molecular mechanistic experiments showed that NR2F1-AS1 overexpression upregulated NR2F1 expression in CC cells by directly binding to miR-642a-3p, and inhibiting by this way cell viability, proliferation, migration, and invasion in CC. Rescue assays showed that NR2F1 knockdown or miR-642a-3p overexpression offset NR2F1-AS1 upregulation-induced inhibition on CC cell malignant phenotypes. Conclusions: These findings revealed that NR2F1-AS1 played a tumor suppressor role in CC by mediating the miR-642a-3p/NR2F1 axis.


Asunto(s)
MicroARNs , ARN Largo no Codificante , Neoplasias del Cuello Uterino , Factor de Transcripción COUP I/genética , Línea Celular Tumoral , Proliferación Celular/genética , Femenino , Humanos , MicroARNs/genética , MicroARNs/metabolismo , ARN Largo no Codificante/genética , ARN Largo no Codificante/metabolismo , Neoplasias del Cuello Uterino/genética
14.
J Gene Med ; 23(7): e3338, 2021 07.
Artículo en Inglés | MEDLINE | ID: mdl-33822440

RESUMEN

BACKGROUND: Long non-coding RNAs (lncRNAs) are vital regulators during the biological processes of melanoma. The present study aimed to uncover biological functions of lncRNA termed NR2F1 antisense RNA 1 (NR2F1-AS1) in melanoma and the potential mechanisms. METHODS: Relative levels of NR2F1-AS1 and miR-493-5p in a total of 137 paired primary melanoma tissues and corresponding non-tumor tissues, as well as three melanoma cell lines, were examined by a real-time polymerase chain reaction. The clinical significance of NR2F1-AS1 expression was analyzed statistically. The STAT3 binding motif in the promoter region of NR2F1-AS1 was identified by JASPAR (http://jaspar.genereg.net). The association between STAT3 and NR2F1-AS1 was determined by dual-luciferase reporter and chromatin immunoprecipitation assays. The effects of NR2F1-AS1 on cell proliferation, migration and were measured by cell counting kit-8 (CCK-8), Edu, transwell and wound healing assays. Dual-luciferase reporter and RNA pull-down assays were applied to validate the interaction among NR2F1-AS1, miR-493-5p and GOLM1. Furthermore, in vivo experiments were conducted to demonstrate the oncogenic role of NR2F1-AS1 in melanoma. RESULTS: Up-regulated NR2F1-AS1 and down-regulated miR-493-5p were detected in melanoma tumors and cells. The overexpression of NR2F1-AS1 was induced by STAT3. High NR2F1-AS1 expression was correlated to advanced tumor stage and poor prognosis of melanoma. Functional studies using CCK-8, Edu, transwell and wound healing assays revealed that the proliferative, migratory and invasive capacities of melanoma cells were attenuated by the by inhibition of NR2F1-AS1. Moreover, NR2F1-AS1 was able to up-regulate GOLM1 through recognizing and binding miR-493-5p. Furthermore, knockdown of miR-493-5p distinctly reversed these inhibitory effects of NR2F1-AS1 down-regulation on the tumorigenesis and progression of melanoma. CONCLUSIONS: Our findings demonstrate a key role for NR2F1-AS1 in melanoma progression via targeting miR-493-5p/GOLM1 axis.


Asunto(s)
Factor de Transcripción COUP I/genética , Melanoma/genética , Melanoma/metabolismo , Proteínas de la Membrana/metabolismo , MicroARNs/metabolismo , ARN Largo no Codificante/genética , Factor de Transcripción STAT3/genética , Factor de Transcripción COUP I/metabolismo , Línea Celular Tumoral , Movimiento Celular , Proliferación Celular , Progresión de la Enfermedad , Regulación Neoplásica de la Expresión Génica , Humanos , ARN Largo no Codificante/metabolismo , Transducción de Señal
15.
Cereb Cortex ; 30(11): 5667-5685, 2020 10 01.
Artículo en Inglés | MEDLINE | ID: mdl-32572460

RESUMEN

The formation of functional cortical maps in the cerebral cortex results from a timely regulated interaction between intrinsic genetic mechanisms and electrical activity. To understand how transcriptional regulation influences network activity and neuronal excitability within the neocortex, we used mice deficient for Nr2f1 (also known as COUP-TFI), a key determinant of primary somatosensory (S1) area specification during development. We found that the cortical loss of Nr2f1 impacts on spontaneous network activity and synchronization of S1 cortex at perinatal stages. In addition, we observed alterations in the intrinsic excitability and morphological features of layer V pyramidal neurons. Accordingly, we identified distinct voltage-gated ion channels regulated by Nr2f1 that might directly influence intrinsic bioelectrical properties during critical time windows of S1 cortex specification. Altogether, our data suggest a tight link between Nr2f1 and neuronal excitability in the developmental sequence that ultimately sculpts the emergence of cortical network activity within the immature neocortex.


Asunto(s)
Factor de Transcripción COUP I/metabolismo , Neurogénesis/fisiología , Células Piramidales/metabolismo , Corteza Somatosensorial/embriología , Corteza Somatosensorial/crecimiento & desarrollo , Animales , Femenino , Regulación del Desarrollo de la Expresión Génica/fisiología , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Corteza Somatosensorial/metabolismo
16.
J Cell Mol Med ; 24(14): 8236-8247, 2020 07.
Artículo en Inglés | MEDLINE | ID: mdl-32548873

RESUMEN

Long non-coding RNAs (lncRNAs) take various effects in cancer mostly through sponging with microRNAs (miRNAs). lncRNA NR2F1-AS1 is found to promote tumour progression in hepatocellular carcinoma, endometrial cancer and thyroid cancer. However, the role of lncRNA NR2F1-AS1 in breast cancer angiogenesis remains unknown. In this study, we found lncRNA NR2F1-AS1 was positively related with CD31 and CD34 in breast cancer through Pearson's correlation analysis, while lncRNA NR2F1-AS1 transfection promoted human umbilical vascular endothelial cell (HUVEC) tube formation. In breast cancer cells, lncRNA NR2F1-AS1 enhanced the HUVEC proliferation, tube formation and migration ability through tumour-conditioned medium (TCM). In zebrafish model, lncRNA NR2F1-AS1 increased the breast cancer cell-related neo-vasculature and subsequently promoted the breast cancer cell metastasis. In mouse model, lncRNA NR2F1-AS1 promoted the tumour vessel formation, increased the micro vessel density (MVD) and then induced the growth of primary tumour. Mechanically, lncRNA NR2F1-AS1 increased insulin-like growth factor-1 (IGF-1) expression through sponging miRNA-338-3p in breast cancer cells and then activated the receptor of IGF-1 (IGF-1R) and extracellular signal-regulated kinase (ERK) pathway in HUVECs. These results indicated that lncRNA NR2F1-AS1 could promote breast cancer angiogenesis through IGF-1/IGF-1R/ERK pathway.


Asunto(s)
Neoplasias de la Mama/genética , Neoplasias de la Mama/metabolismo , Factor de Transcripción COUP I/genética , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Factor I del Crecimiento Similar a la Insulina/metabolismo , ARN Largo no Codificante/genética , Receptor IGF Tipo 1/metabolismo , Animales , Neoplasias de la Mama/patología , Línea Celular Tumoral , Modelos Animales de Enfermedad , Femenino , Regulación Neoplásica de la Expresión Génica , Humanos , Neovascularización Patológica/genética , Neovascularización Patológica/metabolismo , Transducción de Señal , Pez Cebra
17.
J Cell Biochem ; 121(2): 2009-2018, 2020 02.
Artículo en Inglés | MEDLINE | ID: mdl-31692033

RESUMEN

Papillary thyroid carcinoma (PTC) is an aggressive histological subtype of thyroid carcinoma (THCA), whose occurrence rate is high. The participation of long noncoding RNAs in the pathologies of cancers has attracted significant attention during the past decades. The purpose of the current study is to investigate the role of NR2F1 antisense RNA 1 (NR2F1-AS1) in PTC. The expression of NR2F1 in THCA samples was analyzed by bioinformatics tool gene expression profiling interactive analysis. Levels of NR2F1-AS1, microRNA-423-5p (miR-423-5p), and SRY-box 12 (SOX12) were evaluated by a quantitative reverse transcription-polymerase chain reaction and Western blot. The impact of NR2F1-AS1 on PTC cell proliferation and invasion was assessed by Cell Counting Kit-8, EdU, and Transwell invasion assays. The interactions among NR2F1-AS1, miR-423-5p, and SOX12 were determined by RNA immunoprecipitation and luciferase reporter assays. Consequently, we found that NR2F1-AS1 and SOX12 levels were elevated in PTC, whereas miR-423-5p was downregulated in PTC cells. Functionally, NR2F1-AS1 silence led to reduced proliferation and invasion of PTC cells. Mechanistically, NR2F1-AS1 interacted with miR-423-5p to induce SOX12 expression in PTC cells. In conclusion, the present study firstly stated that NR2F1-AS1 regulated miR-423-5p/SOX12 to promote proliferation and invasion of PTC, indicating NR2F1-AS1 as a potential novel target for the molecular-targeted therapy of PTC.


Asunto(s)
Regulación Neoplásica de la Expresión Génica , MicroARNs/genética , ARN sin Sentido/genética , ARN Largo no Codificante/genética , Factores de Transcripción SOXC/metabolismo , Cáncer Papilar Tiroideo/patología , Neoplasias de la Tiroides/patología , Apoptosis , Biomarcadores de Tumor/genética , Biomarcadores de Tumor/metabolismo , Factor de Transcripción COUP I/genética , Proliferación Celular , Humanos , Invasividad Neoplásica , Factores de Transcripción SOXC/genética , Cáncer Papilar Tiroideo/genética , Cáncer Papilar Tiroideo/metabolismo , Neoplasias de la Tiroides/genética , Neoplasias de la Tiroides/metabolismo , Células Tumorales Cultivadas
18.
Development ; 144(11): 2045-2058, 2017 06 01.
Artículo en Inglés | MEDLINE | ID: mdl-28506990

RESUMEN

Development of the dentate gyrus (DG), the primary gateway for hippocampal inputs, spans embryonic and postnatal stages, and involves complex morphogenetic events. We have previously identified the nuclear receptor COUP-TFI as a novel transcriptional regulator in the postnatal organization and function of the hippocampus. Here, we dissect its role in DG morphogenesis by inactivating it in either granule cell progenitors or granule neurons. Loss of COUP-TFI function in progenitors leads to decreased granule cell proliferative activity, precocious differentiation and increased apoptosis, resulting in a severe DG growth defect in adult mice. COUP-TFI-deficient cells express high levels of the chemokine receptor Cxcr4 and migrate abnormally, forming heterotopic clusters of differentiated granule cells along their paths. Conversely, high COUP-TFI expression levels downregulate Cxcr4 expression, whereas increased Cxcr4 expression in wild-type hippocampal cells affects cell migration. Finally, loss of COUP-TFI in postmitotic cells leads to only minor and transient abnormalities, and to normal Cxcr4 expression. Together, our results indicate that COUP-TFI is required predominantly in DG progenitors for modulating expression of the Cxcr4 receptor during granule cell neurogenesis and migration.


Asunto(s)
Factor de Transcripción COUP I/metabolismo , Movimiento Celular , Giro Dentado/citología , Giro Dentado/metabolismo , Mitosis , Receptores CXCR4/genética , Animales , Animales Recién Nacidos , Recuento de Células , Diferenciación Celular/genética , Movimiento Celular/genética , Proliferación Celular/genética , Giro Dentado/embriología , Eliminación de Gen , Regulación del Desarrollo de la Expresión Génica , Proteínas de Homeodominio/metabolismo , Ratones Noqueados , Mitosis/genética , Modelos Biológicos , Células-Madre Neurales/citología , Células-Madre Neurales/metabolismo , Neurogénesis/genética , Neuroglía/metabolismo , Receptores CXCR4/metabolismo , Factores de Transcripción/metabolismo
19.
Development ; 144(15): 2837-2851, 2017 08 01.
Artículo en Inglés | MEDLINE | ID: mdl-28694260

RESUMEN

Distinct cortical interneuron (CIN) subtypes have unique circuit functions; dysfunction in specific subtypes is implicated in neuropsychiatric disorders. Somatostatin- and parvalbumin-expressing (SST+ and PV+) interneurons are the two major subtypes generated by medial ganglionic eminence (MGE) progenitors. Spatial and temporal mechanisms governing their cell-fate specification and differential integration into cortical layers are largely unknown. We provide evidence that Coup-TF1 and Coup-TF2 (Nr2f1 and Nr2f2) transcription factor expression in an arc-shaped progenitor domain within the MGE promotes time-dependent survival of this neuroepithelium and the time-dependent specification of layer V SST+ CINs. Coup-TF1 and Coup-TF2 autonomously repress PV+ fate in MGE progenitors, in part through directly driving Sox6 expression. These results have identified, in mouse, a transcriptional pathway that controls SST-PV fate.


Asunto(s)
Factor de Transcripción COUP II/metabolismo , Factor de Transcripción COUP I/metabolismo , Interneuronas/metabolismo , Neocórtex/citología , Animales , Factor de Transcripción COUP I/genética , Factor de Transcripción COUP II/genética , Células Cultivadas , Inmunoprecipitación de Cromatina , Femenino , Regulación del Desarrollo de la Expresión Génica/genética , Regulación del Desarrollo de la Expresión Génica/fisiología , Inmunohistoquímica , Hibridación in Situ , Masculino , Ratones , Ratones Endogámicos C57BL , Parvalbúminas/genética , Parvalbúminas/metabolismo , Factores de Transcripción SOXD/genética , Factores de Transcripción SOXD/metabolismo , Somatostatina/genética , Somatostatina/metabolismo
20.
Am J Med Genet A ; 182(6): 1426-1437, 2020 06.
Artículo en Inglés | MEDLINE | ID: mdl-32275123

RESUMEN

Bosch-Boonstra-Schaaf Optic Atrophy Syndrome (BBSOAS) is an autosomal dominant neurodevelopmental disorder caused by loss-of-function variants in NR2F1 and characterized by visual impairment, developmental delay, and intellectual disability. Here we report 18 new cases, provide additional clinical information for 9 previously reported individuals, and review an additional 27 published cases to present a total of 54 patients. Among these are 22 individuals with point mutations or in-frame deletions in the DNA-binding domain (DBD), and 32 individuals with other types of variants including whole-gene deletions, nonsense and frameshift variants, and point mutations outside the DBD. We corroborate previously described clinical characteristics including developmental delay, intellectual disability, autism spectrum disorder diagnoses/features thereof, cognitive/behavioral anomalies, hypotonia, feeding difficulties, abnormal brain MRI findings, and seizures. We also confirm a vision phenotype that includes optic nerve hypoplasia, optic atrophy, and cortical visual impairment. Additionally, we expand the vision phenotype to include alacrima and manifest latent nystagmus (fusional maldevelopment), and we broaden the behavioral phenotypic spectrum to include a love of music, an unusually good long-term memory, sleep difficulties, a high pain tolerance, and touch sensitivity. Furthermore, we provide additional evidence for genotype-phenotype correlations, specifically supporting a more severe phenotype associated with DBD variants.


Asunto(s)
Factor de Transcripción COUP I/genética , Discapacidad Intelectual/genética , Atrofias Ópticas Hereditarias/genética , Convulsiones/genética , Codón sin Sentido/genética , Proteínas de Unión al ADN , Femenino , Mutación del Sistema de Lectura/genética , Estudios de Asociación Genética , Humanos , Discapacidad Intelectual/complicaciones , Discapacidad Intelectual/fisiopatología , Masculino , Mutación/genética , Atrofias Ópticas Hereditarias/complicaciones , Atrofias Ópticas Hereditarias/fisiopatología , Mutación Puntual/genética , Convulsiones/complicaciones , Convulsiones/fisiopatología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA