Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 17 de 17
Filtrer
Plus de filtres











Gamme d'année
1.
J Mol Cell Biol ; 2023 Oct 03.
Article de Anglais | MEDLINE | ID: mdl-37791390

RÉSUMÉ

Meningioma is one of the most common primary neoplasms in the central nervous system, whereas there is still no specific molecularly targeted therapy that has been approved for the clinical treatment of aggressive meningiomas. There is therefore an urgent demand to decrypt the biological and molecular landscape of malignant meningioma. Here, through the in-silica prescreening and 10-year follow-up of 445 meningioma patients, we uncovered that CBX7 is progressively decreased with malignancy grade and neoplasia stage in meningioma and a high CBX7 expression level predicts a favorable prognosis in meningioma patients. CBX7 restoration significantly induces cell cycle arrest and inhibits meningioma cell proliferation. iTRAQ-based proteomics analysis indicated that CBX7 restoration triggers the metabolic shift from glycolysis to oxidative phosphorylation. The mechanistic study demonstrated that CBX7 promotes the proteasome-dependent degradation of c-MYC proteins by transcriptionally inhibiting the expression of a c-MYC deubiquitinase, USP44, which attenuates c-MYC-mediated transactivation of LDHA transcripts and further inhibits glycolysis and subsequent cellular proliferation. More importantly, the functional role of CBX7 was further confirmed in both subcutaneous and orthotopic meningioma xenografts mouse models and human meningioma patients. Together, our results shed light on the critical role of CBX7 during meningioma malignancy progression and identified the CBX7/USP44/c-MYC/LDHA axis as a promising therapeutic target against meningioma progression.

2.
World Neurosurg ; 176: e501-e514, 2023 Aug.
Article de Anglais | MEDLINE | ID: mdl-37263494

RÉSUMÉ

BACKGROUND: Dysregulation of immune infiltration critically contributes to the tumorigenesis and progression of meningiomas. However, the landscape of immune microenvironment and key genes correlated with immune cell infiltration remains unclear. METHODS: Four Gene Expression Omnibus data sets were included. CIBERSORT algorithm was utilized to analyze the immune cell infiltration in samples. Wilcoxon test, Random Forest algorithm, and Least Absolute Shrinkage and Selection Operator regression were adopted in identifying significantly different infiltrating immune cells and differentially expressed genes (DEGs). Functional enrichment analysis was performed by Kyoto Encyclopedia of Genes and Genomes and Gene Ontology. The correlation between genes and immune cells was evaluated via Spearman's correlation analysis. Receiver Operator Characteristic curve analysis evaluated the markers' diagnostic effectiveness. The mRNA-miRNA and Drug-Gene-Immune cell interaction networks were constructed to identify potential diagnostic and therapeutic targets. RESULTS: Plasma cells, M1 macrophages, M2 macrophages, neutrophils, eosinophils, and activated NK cells were the significantly different infiltrating immune cells in meningioma. A total of 951 DEGs, associated with synaptic function and structure, ion transport regulation, brain function, and immune-related pathways, were identified. Among 11 hub DEGs, RYR2 and TTR were correlated with plasma cells; SNCG was associated with NK cells; ADCY1 exhibited excellent diagnostic effectiveness; and ADCY1, BMX, KCNA5, SLCO4A1, and TTR could be considered as therapeutic targets. CONCLUSIONS: ADCY1 can be identified as a diagnostic marker; ADCY1, BMX, KCNA5, SLCO4A1, and TTR are potential therapeutic targets, and their associations with macrophages, neutrophils, NK cells, and plasma cells might impact the tumorigenesis of meningiomas.


Sujet(s)
Tumeurs des méninges , Méningiome , microARN , Humains , Méningiome/diagnostic , Méningiome/génétique , Méningiome/thérapie , Carcinogenèse , Transformation cellulaire néoplasique , Tumeurs des méninges/diagnostic , Tumeurs des méninges/génétique , Tumeurs des méninges/thérapie , Microenvironnement tumoral/génétique
3.
Pharmaceutics ; 15(1)2023 Jan 03.
Article de Anglais | MEDLINE | ID: mdl-36678784

RÉSUMÉ

Cardiovascular diseases (CVDs) remain the world's leading cause of death despite the best available healthcare and therapy. Emerging as a key mediator of intercellular and inter-organ communication in CVD pathogenesis, extracellular vesicles (EVs) are a heterogeneous group of membrane-enclosed nano-sized vesicles released by virtually all cells, of which their RNA cargo, especially non-coding RNAs (ncRNA), has been increasingly recognized as a promising diagnostic and therapeutic target. Recent evidence shows that ncRNAs, such as small ncRNAs, circular RNAs, and long ncRNAs, can be selectively sorted into EVs or other non-vesicular carriers and modulate various biological processes in recipient cells. In this review, we summarize recent advances in the literature regarding the origin, extracellular carrier, and functional mechanisms of extracellular ncRNAs with a focus on small ncRNAs, circular RNAs, and long ncRNAs. The pathophysiological roles of extracellular ncRNAs in various CVDs, including atherosclerosis, ischemic heart diseases, hypertension, cardiac hypertrophy, and heart failure, are extensively discussed. We also provide an update on recent developments and challenges in using extracellular ncRNAs as biomarkers or therapeutical targets in these CVDs.

4.
Article de Chinois | WPRIM (Pacifique Occidental) | ID: wpr-989347

RÉSUMÉ

Cancer is the second-leading cause of death worldwide. Cancer mortality is largely caused by the absence of recognizable early signs and a poor prognosis. Therefore, developing efficient diagnostic and prognostic biomarkers is crucial to reducing the incidence of cancer and improving its prognostic accuracy. tRNA-derived fragments are a new class of non-coding RNAs with important regulatory roles in cancer biology. In this paper, the research progress of tRNA-derived fragments as biomarkers in tumorigenesis, development, and prognosis was reviewed to provide a theoretical basis for cancer diagnosis and prognostic assessment.

5.
Biochem Biophys Res Commun ; 635: 291-298, 2022 12 20.
Article de Anglais | MEDLINE | ID: mdl-36327916

RÉSUMÉ

Colorectal cancer is one of the most common malignancies causing the majority of cancer-related deaths. There is an urgent need to develop new anticancer modalities. Recently, efforts have been made to turn clinically approved drugs into anticancer agents in specific tumor microenvironments via NPs. Disulfiram (DSF) as an effective copper (Cu2+)-dependent anti-tumour drug, which has been more widely used in antitumor research. Here, we constructed a novel therapeutic nanoplatforms, DSF@CuS, by encapsulating DSF in hollow CuS NPs to enable in situ chemoselective activation of DSF and hyperthermal amplified chemotherapy. The anticancer effect of DSF was enhanced by the thermal energy generated under NIR irradiation through the intrinsic photothermal conversion of CuS. As a result, significant apoptosis was induced in vitro, and tumor elimination was achieved in vivo. Collectively, DSF@CuS combined with photothermal therapy can significantly promote the apoptosis of CT26 colorectal cancer cells both in vitro and in vivo, providing a potential theoretical agent for the treatment of colorectal cancer.


Sujet(s)
Antinéoplasiques , Tumeurs colorectales , Nanoparticules , Humains , Disulfirame/pharmacologie , Cuivre/pharmacologie , Lignée cellulaire tumorale , Nanoparticules/usage thérapeutique , Sulfures/pharmacologie , Antinéoplasiques/pharmacologie , Tumeurs colorectales/traitement médicamenteux , Microenvironnement tumoral
6.
Med Oncol ; 40(1): 21, 2022 Nov 29.
Article de Anglais | MEDLINE | ID: mdl-36445551

RÉSUMÉ

As a type of central nervous system tumor, meningioma usually compresses the nerve center due to its local expansion, further causing neurological deficits. However, there are limited therapeutic approaches for meningiomas. ITF2357, a potent class I and II histone deacetylase inhibitor (HDACi), has been shown to inhibit cell proliferation, promote apoptosis, and block the cell cycle in various sarcoma cells, including glioblastoma and peripheral T-cell lymphoma. Here, we investigated the potential role of ITF2357 on meningioma cancer cells (IOMM-Lee cells). First, we demonstrated that the half-maximal inhibitory concentration (IC50) of ITF2357 was 1.842 µM by MTT assay. In addition, ITF2357 effectively inhibited the proliferation and colonization ability of IOMM-Lee cells. Flow cytometry analysis showed that ITF2357 induced G0/G1 and G2/M phase cell cycle arrest and cell apoptosis. Mechanically, the RNA sequencing data revealed that ITF2357 could affect the PI3K-Akt signaling pathway and the cell cycle progression. Furthermore, the expression levels of Akt, PI3K, p-Akt, and p-PI3K were determined by western blotting. Collectively, our data revealed that ITF2357 induces G0 G1 and G2/M phase arrest and apoptosis by inhibiting hyperactivation of the PI3K-Akt pathway, ultimately inhibiting cell viability and proliferation of meningioma cells, which developed a new approach to the treatment of meningioma.


Sujet(s)
Tumeurs des méninges , Méningiome , Humains , Méningiome/traitement médicamenteux , Phosphatidylinositol 3-kinases , Protéines proto-oncogènes c-akt , Points de contrôle du cycle cellulaire , Apoptose , Points de contrôle de la phase M du cycle cellulaire , Tumeurs des méninges/traitement médicamenteux
7.
Aging Cell ; 21(7): e13657, 2022 07.
Article de Anglais | MEDLINE | ID: mdl-35718942

RÉSUMÉ

With the aging of the global population, accumulating interest is focused on manipulating the fundamental aging-related signaling pathways to delay the physiological aging process and eventually slow or prevent the appearance or severity of multiple aging-related diseases. Recently, emerging evidence has shown that RNA modifications, which were historically considered infrastructural features of cellular RNAs, are dynamically regulated across most of the RNA species in cells and thereby critically involved in major biological processes, including cellular senescence and aging. In this review, we summarize the current knowledge about RNA modifications and provide a catalog of RNA modifications on different RNA species, including mRNAs, miRNAs, lncRNA, tRNAs, and rRNAs. Most importantly, we focus on the regulation and roles of these RNA modifications in aging-related diseases, including neurodegenerative diseases, cardiovascular diseases, cataracts, osteoporosis, and fertility decline. This would be an important step toward a better understanding of fundamental aging mechanisms and thereby facilitating the development of novel diagnostics and therapeutics for aging-related diseases.


Sujet(s)
Vieillissement/anatomopathologie , microARN , ARN long non codant , Vieillissement de la cellule , microARN/composition chimique , ARN long non codant/composition chimique , ARN messager/composition chimique
8.
Int J Nanomedicine ; 17: 2647-2659, 2022.
Article de Anglais | MEDLINE | ID: mdl-35730051

RÉSUMÉ

Background: Natural bioactive substances have been widely studied for their superior anti-tumor activity and low toxicity. However, natural bioactive substances suffer from poor water-solubility and poor stability in the physiological environment. Therefore, to overcome the drawbacks of natural bioactive substances in tumor therapy, there is an urgent need for an ideal nanocarrier to achieve high bioactive substance loading with low toxicity. Materials and Methods: Face-centered cubic hollow mesoporous Prussian Blue (HMPB) NPs were prepared by stepwise hydrothermal method. Among them, PVP served as a protective agent and HCl served as an etching agent. Firstly, MPB NPs were obtained by 0.01 M HCl etching. Then, the highly uniform dispersed HMPB NPs were obtained by further etching with 1 M HCl. Results: In this work, we report a pH-responsive therapeutic nanoplatform based on HMPB NPs. Surprisingly, as-prepared HMPB NPs with ultra-high bioactive substances loading capacity of 329 µg mg-1 owing to the large surface area (131.67 m2 g-1) and wide internal pore size distribution (1.8-96.2 nm). Moreover, with the outstanding photothermal conversion efficiency of HMPB NPs (30.13%), natural bioactive substances were released in the tumor microenvironment (TME). HMPB@PC B2 achieved excellent synergistic therapeutic effects of photothermal therapy (PTT) and chemotherapy (CT) in vivo and in vitro without causing any extraneous side effects. Conclusion: A biocompatible HMPB@PC B2 nanoplatform was constructed by simple physical adsorption. The in vitro and in vivo experiment results demonstrated that the synergy of PTT/CT provided excellent therapeutic efficiency for cervical cancer without toxicity. Altogether, as-designed nanomedicines based on natural bioactive substances may be provide a promising strategy for cancer therapy.


Sujet(s)
Antinéoplasiques , Nanoparticules , Tumeurs , Antinéoplasiques/pharmacologie , Antinéoplasiques/usage thérapeutique , Lignée cellulaire tumorale , Humains , Tumeurs/traitement médicamenteux , Photothérapie/méthodes , Microenvironnement tumoral
9.
Stem Cells ; 40(1): 22-34, 2022 03 03.
Article de Anglais | MEDLINE | ID: mdl-35511866

RÉSUMÉ

The transition of embryonic stem cells from the epiblast stem cells (EpiSCs) to neural progenitor cells (NPCs), called the neural induction process, is crucial for cell fate determination of neural differentiation. However, the mechanism of this transition is unclear. Here, we identified a long non-coding RNA (linc1548) as a critical regulator of neural differentiation of mouse embryonic stem cells (mESCs). Knockout of linc1548 did not affect the conversion of mESCs to EpiSCs, but delayed the transition from EpiSCs to NPCs. Moreover, linc1548 interacts with the transcription factors OCT6 and SOX2 forming an RNA-protein complex to regulate the transition from EpiSCs to NPCs. Finally, we showed that Zfp521 is an important target gene of this RNA-protein complex regulating neural differentiation. Our findings prove how the intrinsic transcription complex is mediated by a lncRNA linc1548 and can better understand the intrinsic mechanism of neural fate determination.


Sujet(s)
Cellules souches embryonnaires , Feuillets embryonnaires , Animaux , Différenciation cellulaire/génétique , Souris , Souris knockout , ARN , ARN long non codant , Facteurs de transcription SOX-B1
10.
Article de Chinois | WPRIM (Pacifique Occidental) | ID: wpr-989275

RÉSUMÉ

Objective:To investigate the regulatory effects of nuclear factor-κB (NF-κB) on dendritic cell (DC) maturation and function through solute carrier family 1 member 2 (Slc1a2).Methods:Mouse bone marrow-derived DCs were transfected with Slc1a2-specific siRNA and an overexpression Slc1a2 eukaryotic expression vector. The real-time fluorescence quantitation (RT-PCR) and Western Blot methods were used to detect knockdown and overexpression efficiency. The expression of surface molecules (CD40, CD80) and major histocompatibility complex Ⅱ (MHCⅡ) of DCs was detected by flow cytometry. ELISA was used to detect the secretion of the cytokines interleukin (IL)-12, IL-6, and transforming growth factor-β (TGF-β). The effects of knockdown of Slc1a2 on DC maturation and function and the effects of overexpression of Slc1a2 on DC maturation and function were reflected by the above assay results. A mixed lymphocyte culture assay was used to investigate the effect of Slc1a2 on T cell proliferation, and an ELISA was used to detect the lavel of IL-17A. Changes in the relative fluorescence intensity of FITC in DCs were analyzed by flow cytometry to investigate the ability of Slc1a2 overexpression on antigen phagocytosis. Finally, DCs were pretreated with an NF-κB inhibitor, toluoylphenylalanine chloromethyl ketone (TPCK), and the effect of TPCK on the expression of Slc1a2 in DCs and DC maturation was examined.Results:Slc1a2 expression was found to be high in DC treated with lipopolysaccharides (LPS) ( P<0.001). The knockdown of Slc1a2 decreased DC maturation and ability to stimulate the proliferation of CD4 + T cells ( P<0.001) and inhibited IL-17 secretion ( P<0.01). Overexpression of Slc1a2 promoted DC maturation and ability to stimulate the proliferation of CD4 + T cells(all P<0.01) Pretreatment of DC with the NF-κB inhibitor TPCK inhibited the expression of Slc1a2 at mRNA and protein levels induced by LPS. Conclusions:NF-κB regulates Slc1a2 expression, which affects the maturation and function of DC.

11.
EMBO Rep ; 21(11): e50283, 2020 11 05.
Article de Anglais | MEDLINE | ID: mdl-33016573

RÉSUMÉ

A microdeletion within human chromosome 5q14.3 has been associated with the occurrence of neurodevelopmental disorders, such as autism and intellectual disability, and MEF2C haploinsufficiency was identified as main cause. Here, we report that a brain-enriched long non-coding RNA, NDIME, is located near the MEF2C locus and is required for normal neural differentiation of mouse embryonic stem cells (mESCs). NDIME interacts with EZH2, the major component of polycomb repressive complex 2 (PRC2), and blocks EZH2-mediated trimethylation of histone H3 lysine 27 (H3K27me3) at the Mef2c promoter, promoting MEF2C transcription. Moreover, the expression levels of both NDIME and MEF2C were strongly downregulated in the hippocampus of a mouse model of autism, and the adeno-associated virus (AAV)-mediated expression of NDIME in the hippocampus of these mice significantly increased MEF2C expression and ameliorated autism-like behaviors. The results of this study reveal an epigenetic mechanism by which NDIME regulates MEF2C transcription and neural differentiation and suggest potential effects and therapeutic approaches of the NDIME/MEF2C axis in autism.


Sujet(s)
Trouble autistique , Animaux , Trouble autistique/génétique , Différenciation cellulaire , Cellules souches embryonnaires/métabolisme , Protéine-2 homologue de l'activateur de Zeste/génétique , Facteurs de transcription MEF2/génétique , Souris , Complexe répresseur Polycomb-2/génétique , Complexe répresseur Polycomb-2/métabolisme , Régions promotrices (génétique)
12.
Stem Cell Reports ; 11(2): 395-409, 2018 08 14.
Article de Anglais | MEDLINE | ID: mdl-30017820

RÉSUMÉ

Although the functional roles of long noncoding RNAs (lncRNAs) have been increasingly identified, few lncRNAs that control the naïve state of embryonic stem cells (ESCs) are known. Here, we report a naïve-state-associated lncRNA, LincU, which is intrinsically activated by Nanog in mESCs. LincU-deficient mESCs exhibit a primed-like pluripotent state and potentiate the transition from the naïve state to the primed state, whereas ectopic LincU expression maintains mESCs in the naïve state. Mechanistically, we demonstrate that LincU binds and stabilizes the DUSP9 protein, an ERK-specific phosphatase, and then constitutively inhibits the ERK1/2 signaling pathway, which critically contributes to maintenance of the naïve state. Importantly, we reveal the functional role of LincU to be evolutionarily conserved in human. Therefore, our findings unveil LincU as a conserved lncRNA that intrinsically restricts MAPK/ERK activity and maintains the naïve state of ESCs.


Sujet(s)
Auto-renouvellement cellulaire , Cellules souches embryonnaires/cytologie , Cellules souches embryonnaires/métabolisme , Extracellular Signal-Regulated MAP Kinases/métabolisme , ARN long non codant/génétique , Animaux , Différenciation cellulaire/génétique , Auto-renouvellement cellulaire/génétique , Dual-specificity phosphatases/génétique , Régulation de l'expression des gènes au cours du développement , Humains , Souris , Modèles biologiques , Interférence par ARN , Stabilité de l'ARN , Transduction du signal
13.
Stem Cell Reports ; 11(1): 88-101, 2018 07 10.
Article de Anglais | MEDLINE | ID: mdl-29910124

RÉSUMÉ

During reprogramming, telomere re-elongation is important for pluripotency acquisition and ensures the high quality of induced pluripotent stem cells (iPSCs), but the regulatory mechanism remains largely unknown. Our study showed that fully reprogrammed mature iPSCs or mouse embryonic stem cells expressed higher levels of miR-590-3p and miR-590-5p than pre-iPSCs. Ectopic expression of either miR-590-3p or miR-590-5p in pre-iPSCs improved telomere elongation and pluripotency. Activin receptor II A (Acvr2a) is the downstream target and mediates the function of miR-590. Downregulation of Acvr2a promoted telomere elongation and pluripotency. Overexpression of miR-590 or inhibition of ACTIVIN signaling increased telomeric repeat binding factor 1 (Terf1) expression. The p-SMAD2 showed increased binding to the Terf1 promoter in pre-iPSCs compared with mature iPSCs. Downregulation of Terf1 blocked miR-590- or shAcvr2a-mediated promotion of telomere elongation and pluripotency in pre-iPSCs. This study elucidated the role of the miR-590/Acvr2a/Terf1 signaling pathway in modulating telomere elongation and pluripotency in pre-iPSCs.


Sujet(s)
Régulation de l'expression des gènes au cours du développement , Cellules souches pluripotentes induites/cytologie , Cellules souches pluripotentes induites/métabolisme , Homéostasie des télomères/génétique , Télomère/génétique , Récepteur activine, type 2/génétique , Récepteur activine, type 2/métabolisme , Animaux , Différenciation cellulaire/génétique , Lignée cellulaire , Auto-renouvellement cellulaire/génétique , Souris , microARN/génétique , Interférence par ARN , Télomère/métabolisme , Protéine-1 se liant aux répétitions télomériques/génétique , Protéine-1 se liant aux répétitions télomériques/métabolisme
14.
Cell Stem Cell ; 22(6): 893-908.e6, 2018 Jun 01.
Article de Anglais | MEDLINE | ID: mdl-29754779

RÉSUMÉ

Large intergenic non-coding RNAs (lincRNAs) play widespread roles in epigenetic regulation during multiple differentiation processes, but little is known about their mode of action in cardiac differentiation. Here, we identified the key roles of a lincRNA, termed linc1405, in modulating the core network of cardiac differentiation by functionally interacting with Eomes. Chromatin- and RNA-immunoprecipitation assays showed that exon 2 of linc1405 physically mediates a complex consisting of Eomes, trithorax group (TrxG) subunit WDR5, and histone acetyltransferase GCN5 binding at the enhancer region of Mesp1 gene and activates its expression during cardiac mesoderm specification of embryonic stem cells. Importantly, linc1405 co-localizes with Eomes, WDR5, and GCN5 at the primitive streak, and linc1405 depletion impairs heart development and function in vivo. In summary, linc1405 mediates a Eomes/WDR5/GCN5 complex that contributes to cardiogenesis, highlighting the critical roles of lincRNA-based complexes in the epigenetic regulation of cardiogenesis in vitro and in vivo.


Sujet(s)
Mésoderme/métabolisme , Myocytes cardiaques/métabolisme , ARN long non codant/métabolisme , Animaux , Différenciation cellulaire , Épigenèse génétique/génétique , Cellules HEK293 , Humains , Mésoderme/cytologie , Souris , Souris de lignée C57BL , Souris knockout , Myocytes cardiaques/cytologie , Cellules NIH 3T3 , ARN long non codant/génétique
15.
Nucleic Acids Res ; 46(12): 6026-6040, 2018 07 06.
Article de Anglais | MEDLINE | ID: mdl-29733394

RÉSUMÉ

Sin3a is a core component of histone-deacetylation-activity-associated transcriptional repressor complex, playing important roles in early embryo development. Here, we reported that down-regulation of Sin3a led to the loss of embryonic stem cell (ESC) self-renewal and skewed differentiation into mesendoderm lineage. We found that Sin3a functioned as a transcriptional coactivator of the critical Nodal antagonist Lefty1 through interacting with Tet1 to de-methylate the Lefty1 promoter. Further studies showed that two amino acid residues (Phe147, Phe182) in the PAH1 domain of Sin3a are essential for Sin3a-Tet1 interaction and its activity in regulating pluripotency. Furthermore, genome-wide analyses of Sin3a, Tet1 and Pol II ChIP-seq and of 5mC MeDIP-seq revealed that Sin3a acted with Tet1 to facilitate the transcription of a set of their co-target genes. These results link Sin3a to epigenetic DNA modifications in transcriptional activation and have implications for understanding mechanisms underlying versatile functions of Sin3a in mouse ESCs.


Sujet(s)
Protéines de liaison à l'ADN/métabolisme , Cellules souches embryonnaires/métabolisme , Protéines proto-oncogènes/métabolisme , Protéines de répression/métabolisme , Activation de la transcription , Animaux , Différenciation cellulaire , Lignée cellulaire , Lignage cellulaire , Cellules souches embryonnaires/cytologie , Facteurs de détermination de l'asymétrie droite-gauche/génétique , Facteurs de détermination de l'asymétrie droite-gauche/métabolisme , Souris , Protéine Nodal/métabolisme , Régions promotrices (génétique) , Motifs et domaines d'intéraction protéique , Protéines de répression/composition chimique , Protéines de répression/génétique , Complexe Sin3-histone désacétylases-corépresseurs
16.
Stem Cells ; 36(3): 325-336, 2018 03.
Article de Anglais | MEDLINE | ID: mdl-29205638

RÉSUMÉ

Clarifying the regulatory mechanisms of embryonic stem cell (ESC) neural differentiation is helpful not only for understanding neural development but also for obtaining high-quality neural progenitor cells required by stem cell therapy of neurodegenerative diseases. Here, we found that long noncoding RNA 1604 (lncRNA-1604) was highly expressed in cytoplasm during neural differentiation, and knockdown of lncRNA-1604 significantly repressed neural differentiation of mouse ESCs both in vitro and in vivo. Bioinformatics prediction and mechanistic analysis revealed that lncRNA-1604 functioned as a novel competing endogenous RNA of miR-200c and regulated the core transcription factors ZEB1 and ZEB2 during neural differentiation. Furthermore, we also demonstrated the critical role of miR-200c and ZEB1/2 in mouse neural differentiation. Either introduction of miR-200c sponge or overexpression of ZEB1/2 significantly reversed the lncRNA-1604 knockdown-induced repression of mouse ESC neural differentiation. Collectively, these findings not only identified a previously unknown role of lncRNA-1604 and ZEB1/2 but also elucidated a new regulatory lncRNA-1604/miR-200c/ZEB axis in neural differentiation. Stem Cells 2018;36:325-336.


Sujet(s)
microARN/métabolisme , Neurones/cytologie , Neurones/métabolisme , ARN long non codant/métabolisme , Facteur de transcription Zeb2/métabolisme , Facteur de transcription Zeb1/métabolisme , Animaux , Différenciation cellulaire/génétique , Différenciation cellulaire/physiologie , Lignée cellulaire , Biologie informatique/méthodes , Transition épithélio-mésenchymateuse/génétique , Transition épithélio-mésenchymateuse/physiologie , Souris , microARN/génétique , ARN long non codant/génétique , Facteur de transcription Zeb2/génétique , Facteur de transcription Zeb1/génétique
17.
Stem Cell Reports ; 8(5): 1270-1286, 2017 05 09.
Article de Anglais | MEDLINE | ID: mdl-28434941

RÉSUMÉ

Environmental stresses are increasingly acknowledged as core causes of abnormal neural induction leading to neural tube defects (NTDs). However, the mechanism responsible for environmental stress-triggered neural induction defects remains unknown. Here, we report that a spectrum of environmental stresses, including oxidative stress, starvation, and DNA damage, profoundly activate SIRT1, an NAD+-dependent lysine deacetylase. Both mouse embryos and in vitro differentiated embryonic stem cells (ESCs) demonstrated a negative correlation between the expression of SIRT1 and that of OCT6, a key neural fate inducer. Activated SIRT1 radically deacetylates OCT6, triggers an OCT6 ubiquitination/degradation cascade, and consequently increases the incidence of NTD-like phenotypes in mice or hinders neural induction in both human and mouse ESCs. Together, our results suggest that early exposure to environmental stresses results in the dysregulation of the SIRT1/OCT6 axis and increases the risk of NTDs.


Sujet(s)
Exposition environnementale , Anomalies du tube neural/métabolisme , Facteur de transcription Oct-6/métabolisme , Stress oxydatif , Sirtuine-1/métabolisme , Animaux , Cellules cultivées , Altération de l'ADN , Cellules souches embryonnaires/cytologie , Cellules souches embryonnaires/métabolisme , Humains , Souris , Cellules souches neurales/cytologie , Cellules souches neurales/métabolisme , Anomalies du tube neural/étiologie , Anomalies du tube neural/génétique , Facteur de transcription Oct-6/génétique , Protéolyse , Sirtuine-1/génétique , Ubiquitination
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE