Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 19 de 19
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
Acta Neurol Belg ; 124(2): 549-557, 2024 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-37814093

RESUMO

OBJECTIVE: Studies suggest that LncRNA maternally expressed 8, small nucleolar RNA host gene (MEG8) contributes to inflammatory regulation, while the function and potential mechanisms of MEG8 in Parkinson's disease (PD) are unknown. This study aimed to assess the clinical value and biological function of MEG8 in PD. METHODS: One hundred and two PD patients, eighty-six AD patients, and eighty healthy controls were enrolled in this study. Lipopolysaccharide (LPS)-induced microglia BV2 constructs an in vitro cell model. RT-qPCR was conducted to quantify the levels of MEG8, miR-485-3p, and FBXO45 in serum and cells. ROC curve was employed to examine the diagnostic value of MEG8 in PD. Serum and cellular pro-inflammatory factor secretion were quantified by ELISA. Dual-luciferase reporter and RIP assay to validate the targeting relationship between miR-485-3p and FBXO45. RESULTS: MEG8 and FBXO45 were significantly decreased in the serum of PD patients and LPS-induced bv2, while miR-485-3p was increased (P < 0.05). ROC curve confirmed that serum MEG8 has high sensitivity and specificity to identify PD patients from healthy controls and AD patients, respectively. Elevated MEG8 alleviated LPS-induced inflammatory factor overproduction compared with LPS-induced BV2 (P < 0.05), but this alleviating effect was eliminated by miR-485-3p (P < 0.05). The LPS-induced inflammatory response was suppressed by the low expression of miR-485-3p but significantly reversed by silencing of FBXO45. MEG8 was a sponge for miR-485-3p and inhibited its levels and promoted FBXO45 expression (P < 0.05). CONCLUSION: Elevated MEG8 is a potential diagnostic biomarker for PD and may mitigate inflammatory damage in PD via the miR-485-3p/FBXO45 axis.


Assuntos
Proteínas F-Box , MicroRNAs , Doença de Parkinson , RNA Longo não Codificante , Humanos , RNA Longo não Codificante/genética , Lipopolissacarídeos/farmacologia , Doença de Parkinson/genética , Inflamação , MicroRNAs/genética , Apoptose
2.
Oral Dis ; 30(5): 3351-3362, 2024 07.
Artigo em Inglês | MEDLINE | ID: mdl-38155397

RESUMO

OBJECTIVES: Periodontal ligament stem cells (PDLSCs) are essential for the treatment of bone diseases because of its great potential to differentiate into osteoblasts. Remarkably, increasing long-non-coding RNAs (lncRNAs) have been reported to be involved in the osteogenic differentiation of PDLSCs. Maternally expressed 8, small nucleolar RNA host gene (MEG8) is implicated in multiple diseases. This study intended to unearth the potential role of MEG8 and unveil the mechanism in PDLSCs undergoing osteoblastic differentiation. MATERIALS AND METHODS: MEG8 expression was measured by quantitative real-time PCR (RT-qPCR) during osteogenic differentiation of PDLSCs into bone cells. Functional assays were used to uncover the biological function of MEG8. Besides, RNA pulldown, RNA-binding protein immunoprecipitation (RIP), and luciferase reporter assays were used to explore the molecular mechanism of MEG8. RESULTS: MEG8 was apparently overexpressed in osteogenically differentiated PDLSCs. Moreover, MEG8 deficiency suppressed the osteoblastic differentiation of PDLSCs. Furthermore, MEG8 modulated the expression of transcription factor 4 (TCF4) by scavenging microRNA-495-3p (miR-495-3p) and microRNA-485-3p (miR-485-3p) through the competing endogenous RNA (ceRNA) mechanism, further stimulating the Wnt/ß-catenin pathway. CONCLUSION: MEG8 stimulates the capacity of PDLSCs for osteogenic differentiation through a ceRNA mode.


Assuntos
Diferenciação Celular , MicroRNAs , Osteogênese , Ligamento Periodontal , RNA Longo não Codificante , Células-Tronco , Ligamento Periodontal/citologia , Ligamento Periodontal/metabolismo , Humanos , Osteogênese/genética , Diferenciação Celular/genética , Células-Tronco/metabolismo , MicroRNAs/genética , MicroRNAs/metabolismo , RNA Longo não Codificante/genética , RNA Longo não Codificante/metabolismo , Osteoblastos/metabolismo , Via de Sinalização Wnt/genética , Células Cultivadas , Fator de Transcrição 4/genética , RNA Nucleolar Pequeno/genética
3.
Genes (Basel) ; 14(6)2023 06 14.
Artigo em Inglês | MEDLINE | ID: mdl-37372444

RESUMO

Meg8-DMR is the first maternal methylated DMR to be discovered in the imprinted Dlk1-Dio3 domain. The deletion of Meg8-DMR enhances the migration and invasion of MLTC-1 depending on the CTCF binding sites. However, the biological function of Meg8-DMR during mouse development remains unknown. In this study, a CRISPR/Cas9 system was used to generate 434 bp genomic deletions of Meg8-DMR in mice. High-throughput and bioinformatics profiling revealed that Meg8-DMR is involved in the regulation of microRNA: when the deletion was inherited from the mother (Mat-KO), the expression of microRNA was unchanged. However, when the deletion occurred from the father (Pat-KO) and homozygous (Homo-KO), the expression was upregulated. Then, differentially expressed microRNAs (DEGs) were identified between WT with Pat-KO, Mat-KO, and Homo-KO, respectively. Subsequently, these DEGs were subjected to the Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway and Gene Ontology (GO) term enrichment analysis to explore the functional roles of these genes. In total, 502, 128, and 165 DEGs were determined. GO analysis showed that these DEGs were mainly enriched in axonogenesis in Pat-KO and Home-KO, while forebrain development was enriched in Mat-KO. Finally, the methylation levels of IG-DMR, Gtl2-DMR, and Meg8-DMR, and the imprinting status of Dlk1, Gtl2, and Rian were not affected. These findings suggest that Meg8-DMR, as a secondary regulatory region, could regulate the expression of microRNAs while not affecting the normal embryonic development of mice.


Assuntos
MicroRNAs , Animais , Feminino , Camundongos , Gravidez , Proteínas de Ligação ao Cálcio/genética , Metilação de DNA/genética , Desenvolvimento Embrionário/genética , Impressão Genômica , Peptídeos e Proteínas de Sinalização Intercelular/genética , MicroRNAs/genética , Sequências Reguladoras de Ácido Nucleico
4.
Am J Med Genet A ; 191(8): 2225-2231, 2023 08.
Artigo em Inglês | MEDLINE | ID: mdl-37222159

RESUMO

Kagami-Ogata syndrome and Temple syndrome are imprinting disorders caused by the abnormal expression of genes in an imprinted cluster on chromosome 14q32. Here, we report a female with mild features of the Kagami-Ogata syndrome phenotype with polyhydramnios, neonatal hypotonia, feeding difficulties, abnormal foot morphology, patent foramen ovale, distal arthrogryposis, normal facial profile, and a bell-shaped thorax without coat hanger ribs. The single nucleotide polymorphism array revealed the interstitial deletion of chromosome 14q32.2-q32.31 (117 kb in size), involving the RTL1as and MEG8 genes, and other small nucleolar RNAs and microRNAs. The differentially methylated regions (DMRs) appeared unaltered. The RTL1as gene deletion and the normal methylation pattern of the MEG3 gene loci were confirmed by methylation-specific multiplex ligation-dependent probe amplification. Deletions of the 14q32 region without involving DMRs, and encompassing only the RTL1as and MEG8 genes, are poorly described in the literature. The mother's chromosomal microarray also confirmed the identical 14q32.2 deletion, although she presented a normal phenotype. The maternally inherited 14q32 deletion was responsible for Kagami-Ogata syndrome in our patient. It was not sufficient, however, to produce Temple syndrome or any other pathogenic phenotype in the patient's mother.


Assuntos
Anormalidades Múltiplas , Transtornos Cromossômicos , Recém-Nascido , Gravidez , Humanos , Feminino , Transtornos Cromossômicos/genética , Impressão Genômica , Herança Materna , Fenótipo , Anormalidades Múltiplas/genética , Dissomia Uniparental , Cromossomos Humanos Par 14/genética
5.
Nephrology (Carlton) ; 27(12): 994-1002, 2022 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-36164928

RESUMO

AIM: Gestational diabetes mellitus (GDM) is the most common complication in pregnancy. This study aimed to investigate the potential mechanism and effects of long-noncoding RNA maternally expressed 8 (lncRNA-MEG8) in GDM. METHODS: Targeted interactions involving lncRNA-MEG8 and miR-296-3p were initially predicted using starBase software and then confirmed using dual-luciferase reporter gene analysis. The expression levels of lncRNA-MEG8 and miR-296-3p in peripheral blood samples from patients with GDM were measured using reverse transcription-quantitative polymerase chain reaction. Enzyme-linked immunosorbent assay was used to evaluate the overall levels of insulin and insulin secretion. Additionally, MTT and flow cytometric methods were used to detect cell viability and apoptosis. Cell apoptosis-associated proteins were determined by western blotting. RESULTS: Our results indicated that lncRNA-MEG8 is a potential target of miR-296-3p. lncRNA-MEG8 level was higher, whereas that of miR-296-3p was lower in patients with GDM than in healthy individuals. LncRNA-MEG8-siRNA promoted insulin content and secretion. Furthermore, MEG8-siRNA increased cell viability and decreased apoptosis. However, these changes were reversed by an miR-296-3p inhibitor. Moreover, a miR-296-3p mimic had the same effect on INS-1 cells as MEG8-siRNA, as evidenced by enhanced insulin secretion, cell viability, and reduced apoptosis. CONCLUSION: LncRNA-MEG8-siRNA promotes pancreatic ß-cell function by upregulating miR-296-3p.


Assuntos
Diabetes Gestacional , Insulinas , MicroRNAs , RNA Longo não Codificante , Feminino , Gravidez , Humanos , RNA Longo não Codificante/genética , RNA Longo não Codificante/metabolismo , Diabetes Gestacional/genética , MicroRNAs/genética , MicroRNAs/metabolismo , RNA Interferente Pequeno/farmacologia , Proliferação de Células , Apoptose , Insulinas/farmacologia
6.
Cancer Cell Int ; 22(1): 285, 2022 Sep 16.
Artigo em Inglês | MEDLINE | ID: mdl-36114498

RESUMO

Maternally expressed 8 (MEG8) is a long non-coding RNA which is expressed in the nucleus. It is highly expressed in adrenal, placenta and brain. Recent studies have shown contribution of MEG8 in different disorders ranging from neoplastic ones to diabetic nephropathy, atherosclerosis, ischemic stroke, trophoblast dysfunction and abortion, Henoch-Schonlein purpura and osteoarthritis. It has an oncogenic role in the development of lung, pancreatic and liver cancer. In the current review, we summarize the role of this lncRNA in mentioned disorders, based on the evidence obtained from in vitro, in vivo and human studies.

7.
Int J Mol Sci ; 23(15)2022 Aug 08.
Artigo em Inglês | MEDLINE | ID: mdl-35955961

RESUMO

The Dlk1-Dio3 imprinted domain on mouse chromosome 12 contains three well-characterized paternally methylated differentially methylated regions (DMRs): IG-DMR, Gtl2-DMR, and Dlk1-DMR. These DMRs control the expression of many genes involved in embryonic development, inherited diseases, and human cancer in this domain. The first maternal methylation DMR discovered in this domain was the Meg8-DMR, the targets and biological function of which are still unknown. Here, using an enhancer-blocking assay, we first dissected the functional parts of the Meg8-DMR and showed that its insulator activity is dependent on the CCCTC-binding factor (CTCF) in MLTC-1. Results from RNA-seq showed that the deletion of the Meg8-DMR and its compartment CTCF binding sites, but not GGCG repeats, lead to the downregulation of numerous genes on chromosome 12, in particular the drastically reduced expression of Dlk1 and Rtl1 in the Dlk1-Dio3 domain, while differentially expressed genes are enriched in the MAPK pathway. In vitro assays revealed that the deletion of the Meg8-DMR and CTCF binding sites enhances cell migration and invasion by decreasing Dlk1 and activating the Notch1-Rhoc-MAPK/ERK pathway. These findings enhance research into gene regulation in the Dlk1-Dio3 domain by indicating that the Meg8-DMR functions as a long-range regulatory element which is dependent on CTCF binding sites and affects multiple genes in this domain.


Assuntos
Impressão Genômica , RNA Longo não Codificante , Animais , Sítios de Ligação , Proteínas de Ligação ao Cálcio/genética , Metilação de DNA , Feminino , Humanos , Peptídeos e Proteínas de Sinalização Intercelular/genética , Camundongos , Gravidez , RNA Longo não Codificante/genética
8.
Curr Pharm Des ; 28(20): 1688-1694, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-35578848

RESUMO

BACKGROUND: As a member of long non-coding RNAs (lncRNAs), maternally expressed gene 8 (MEG8) has been found involving in the progression of a variety of cancers and playing a regulatory role. Therefore, MEG8 may turn into a new therapeutic target for cancer in the future. The purpose of this review is to illustrate the molecular mechanism and physiological function of MEG8 in various cancers. METHODS: We retrieved and analyzed related articles about MEG8, lncRNAs, and cancers, and then summarize the pathophysiological mechanisms of MEG8 in cancer development. RESULTS: LncRNA MEG8 participates in various cancers progression, thus influencing the proliferation, migration, and invasion of cancers. However, the expression of MEG8 is abnormally upregulated in non-small cell lung cancer (NSCLC), pancreatic cancer (PC), liver cancer (HCC), pituitary adenoma (PA) and hemangioma (HA), and inhibited in colorectal cancer (CRC), ovarian cancer (OC) and giant cell tumor (GCT), suggesting its clinical value in cancer therapy. CONCLUSION: LncRNA MEG8 is expected to be a new therapeutic target or biomarker for a wide range of cancers in the future.


Assuntos
Neoplasias/metabolismo , RNA Longo não Codificante , Proliferação de Células , Regulação Neoplásica da Expressão Gênica , Humanos , RNA Longo não Codificante/genética , RNA Longo não Codificante/metabolismo
9.
Pathol Res Pract ; 229: 153734, 2022 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-35030351

RESUMO

Clear cell renal cell carcinoma (ccRCC) is recognized as one of the most lethal malignancies among the urological system, with constantly increasing mortality. While the molecular mechanisms underlying ccRCC progression are still poorly understood, the molecular and functional role of lncRNA in multiple diseases has been well demonstrated. In this study, we hypothesized that lncRNA MEG8 might participate in ccRCC development. At first, we found that MEG8 expression was increased in ccRCC tumor tissues and cells. Next, we demonstrated that MEG8 knockdown suppressed cell viability, migration, and invasion in vitro and inhibited tumor growth in vivo. Subsequently, we utilized bioinformatics analysis, ChIP, and luciferase assays, and we found that PLAG1 could transcriptionally regulate MEG8 in ccRCC cells. Furthermore, MEG8 promoted G3BP1 expression to aggravate ccRCC tumorigenic properties through sponging miR-495-3p. Our study identified a novel PLAG1/MEG8/miR-495-3p/G3BP1 network in ccRCC development, which might be a promising direction for developing new diagnoses or therapeutic agents for ccRCC.


Assuntos
Carcinoma de Células Renais/genética , DNA Helicases/fisiologia , Proteínas de Ligação a DNA/fisiologia , Neoplasias Renais/genética , MicroRNAs/fisiologia , Proteínas de Ligação a Poli-ADP-Ribose/fisiologia , RNA Helicases/fisiologia , Proteínas com Motivo de Reconhecimento de RNA/fisiologia , RNA Longo não Codificante , Humanos , Células Tumorais Cultivadas
10.
Front Mol Biosci ; 8: 697273, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34790697

RESUMO

It has been recognized that rebalancing the abnormal proliferation and migration of vascular smooth muscle cells (VSMCs) helps relieve vascular injury. Presently, we aim to investigate whether long non-coding RNA (lncRNA) maternally expressed 8 (MEG8) plays a role in affecting the excessive proliferation and migration of VSMCs following hypoxia stimulation. A percutaneous transluminal angioplasty balloon dilatation catheter was adopted to establish vascular intimal injury, the levels of MEG8 and miR-195-5p in the carotid artery were tested by quantitative reverse transcription-polymerase chain reaction (qRT-PCR). Hypoxia was used to stimulate VSMCs, then the cell counting kit-8 (CCK-8) assay, Transnwell assay, and wound healing assay were conducted to evaluate the proliferation, and migration of VSMCs. The protein levels of RECK (reversion inducing cysteine rich protein with kazal motifs), MMP (matrix metalloproteinase) 3/9/13, COX2 (cytochrome c oxidase subunit II), macrophage inflammatory protein (MIP)-1beta, VCAM-1 (vascular cell adhesion molecule 1), ICAM-1 (intercellular adhesion molecule 1), and HIF-1α (hypoxia inducible factor 1 subunit alpha) were determined by western blot or cellular immunofluorescence. As the data showed, MEG8 was down-regulated in the carotid artery after balloon injury in rats and hypoxia-treated VSMCs, and miR-195-5p was overexpressed. Forced MEG8 overexpression or inhibiting miR-195-5p attenuated hypoxia-promoted cell proliferation and migration of VSMCs. In addition, miR-195-5p up-regulation reversed MEG8-mediated effects. Hypoxia hindered the RECK expression while boosted MMP3/9/13 levels, and the effect was markedly reversed with MEG8 up-regulation or miR-195-5p down-regulation. Mechanistically, MEG8 functioned as a competitive endogenous (ceRNA) by sponging miR-195-5p which targeted RECK. Moreover, the HIF-1α inhibitor PX478 prevented hypoxia-induced proliferation, and migration of VSMCs, upregulated MEG8, and restrained miR-195-5p expression. Overall, lncRNA MEG8 participated in hypoxia-induced excessive proliferation, inflammation and migration of VSMCs through the miR-195-5p/RECK axis.

11.
Aging (Albany NY) ; 13(10): 14342-14354, 2021 05 19.
Artigo em Inglês | MEDLINE | ID: mdl-34016788

RESUMO

There are few studies on the mechanism of pituitary adenoma (PA) destroying bone. The current study aimed to investigate the role of MEG8/miR-454-3p/TNF-α in bone-invasive pituitary adenomas (BIPAs). In this study, we report that lncRNA MEG8 and TNF-α are upregulated in BIPA tissues while miR-454-3p is downregulated, which is associated with poor progression-free survival (PFS). Functional assays revealed the role of up-regulated MEG8 and down-regulated miR-454-3p in promoting bone destruction. Mechanistically, MEG8 promotes TNF-α expression by sponging miR-454-3p, which ultimately leads to the occurrence of bone destruction. The mechanism is confirmed in vivo and in vitro. Therefore, our data illustrated a new regulatory mechanism of MEG8/miR-454-3p/TNF-α in BIPAs. It may provide a useful strategy for diagnosis and treatment for BIPA patients.


Assuntos
Adenoma/genética , Adenoma/patologia , Osso e Ossos/patologia , MicroRNAs/metabolismo , Neoplasias Hipofisárias/genética , Neoplasias Hipofisárias/patologia , RNA Longo não Codificante/metabolismo , Fator de Necrose Tumoral alfa/metabolismo , Adulto , Idoso , Animais , Sequência de Bases , Linhagem Celular Tumoral , Progressão da Doença , Regulação para Baixo/genética , Feminino , Regulação Neoplásica da Expressão Gênica , Humanos , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Nus , MicroRNAs/genética , Pessoa de Meia-Idade , Modelos Biológicos , Invasividade Neoplásica , Prognóstico , Células RAW 264.7 , RNA Longo não Codificante/genética , Regulação para Cima/genética
12.
Biochem Biophys Res Commun ; 556: 72-78, 2021 06 04.
Artigo em Inglês | MEDLINE | ID: mdl-33839417

RESUMO

Even though long non-coding RNA (lncRNA) MEG8 plays vital roles in carcinogenesis of malignances, its roles and mechanisms in hemangioma remain unknown. Therefore, we evaluate the oncogenic roles of MEG8 in hemangioma. Small interfering RNA (siRNA)-mediated depletion of MEG8 inhibited the proliferation and increased MDA level in human hemangioma endothelial cells (HemECs). The inhibitors of ferroptosis (ferrostatin-1 and liproxstatin-1) abolished the MEG8 silence induced cell viability loss. Knockdown of MEG8 increased the miR-497-5p expression and reduced the mRNA and protein levels of NOTCH2. Using a dual-luciferase assay, we confirmed the binding between MEG8 and miR-497-5p, and between the miR-497-5p and 3'UTR of NOTCH2. We further found that silencing MEG8 significantly decreased the expressions of SLC7A11 and GPX4 both in mRNA and protein level and had no effect on the level of AIFM2. Importantly, blocking miR-497-5p abrogated the effects of MEG8 loss on cell viability, MDA level and expression levels of NOTCH2, SLC7A11 and GPX4 in HemECs. Taken together, our results suggested that knockdown of long non-coding RNA MEG8 inhibited the proliferation and induced the ferroptosis of hemangioma endothelial cells by regulating miR-497-5p/NOTCH2 axis.


Assuntos
Células Endoteliais/metabolismo , Ferroptose/genética , Inativação Gênica , Hemangioma/genética , MicroRNAs/genética , RNA Longo não Codificante/genética , Receptor Notch2/genética , Sistema y+ de Transporte de Aminoácidos/genética , Sistema y+ de Transporte de Aminoácidos/metabolismo , Proteínas Reguladoras de Apoptose/genética , Proteínas Reguladoras de Apoptose/metabolismo , Sequência de Bases , Proliferação de Células/efeitos dos fármacos , Proliferação de Células/genética , Cicloexilaminas/farmacologia , Regulação para Baixo , Células Endoteliais/patologia , Ferroptose/efeitos dos fármacos , Inativação Gênica/efeitos dos fármacos , Humanos , MicroRNAs/metabolismo , Proteínas Mitocondriais/genética , Proteínas Mitocondriais/metabolismo , Fenilenodiaminas/farmacologia , Fosfolipídeo Hidroperóxido Glutationa Peroxidase/genética , Fosfolipídeo Hidroperóxido Glutationa Peroxidase/metabolismo , Quinoxalinas/farmacologia , RNA Longo não Codificante/antagonistas & inibidores , RNA Interferente Pequeno/genética , Receptor Notch2/biossíntese , Receptor Notch2/metabolismo , Compostos de Espiro/farmacologia
13.
Cancer Cell Int ; 21(1): 84, 2021 Feb 01.
Artigo em Inglês | MEDLINE | ID: mdl-33526036

RESUMO

BACKGROUND: Non-small cell lung cancer (NSCLC) is the most common tumor with severe morbidity and high mortality. Long non-coding RNAs (lncRNAs) as crucial regulators participate in multiple cancer progressions. However, the role of lncRNA MEG8 in the development of NSCLC remains unclear. Here, we aimed to investigate the effect of lncRNA MEG8 on the progression of NSCLC and the underlying mechanism. METHODS: Cell proliferation was analyzed by EdU assays. The impacts of lncRNA MEG8, miR-15a-5p, and miR-15b-5p on cell invasion and migration of NSCLC were assessed by transwell assay. The luciferase reporter gene assay was performed using the Dual-luciferase Reporter Assay System. The effect of lncRNA MEG8, miR-15a-5p, and miR-15b-5p on tumor growth was evaluated in nude mice of Balb/c in vivo. RESULTS: We revealed that the expression levels of MEG8 were elevated in the NSCLC patient tissues compared to that in adjacent normal tissues. The expression of MEG8 was negatively relative to that of miR-15a-5p and miR-15b-5p in the NSCLC patient tissues. The expression of MEG8 was upregulated, while miR-15a-5p and miR-15b-5p were downregulated in NSCLC cell lines. The depletion of MEG8 inhibited NSCLC cell proliferation, migration, and invasion in vitro. MEG8 contributed to NSCLC progression by targeting miR-15a-5p/miR-15b-5p in vitro. LncRNA MEG8 contributes to tumor growth of NSCLC via the miR-15a/b-5p/PSAT1 axis in vivo. Thus, we concluded that lncRNA MEG8 promotes NSCLC progression by modulating the miR-15a/b-5p/PSAT1 axis. CONCLUSIONS: Our findings demonstrated that lncRNA MEG8 plays a critical role in NSCLC development. LncRNA MEG8, miR-15a-5p, miR-15b-5p, and PSAT1 may serve as potential targets for NSCLC therapy.

14.
Cell Mol Neurobiol ; 41(6): 1311-1324, 2021 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-32627090

RESUMO

MEG8 is involved in ischemia stroke, however, its role in ischemia stroke remains unknown. The current research aimed to investigate the effects and mechanisms of MEG8 in ischemic stroke. Mouse brain microvascular endothelial cells (BMECs) were treated by oxygen-glucose deprivation (OGD). Then, the expressions of MEG8 and miR-130a-5p were detected by quantitative reverse transcription-polymerase chain reaction (q-PCR). Cell counting kit-8 (CCK-8), wound-healing, tube formation, Western blot, and q-PCR assays were performed to detect the effects of MEG8 and miR-130a-5p on cell viability, migration, and angiogenesis and VEGFA expression. Bioinformatics, dual-luciferase reporter assay, and RNA immunoprecipitation analysis were carried out to investigate the targeting relationship between MEG8 and miR-130a-5p, and between miR-130a-5p and VEGFA. Then, rat middle cerebral artery occlusion (MCAO) model and MEG8 overexpression MCAO model were established, and neurological deficit and infarct volume of the model rats were evaluated. Finally, Western blot and q-PCR were carried out to detect the expressions of MEG8, miR-130a-5p, and VEGFA. MEG8 was upregulated and miR-130a-5p was downregulated in OGD-treated BMECs. MiR-130a-5p was found to be a target of MEG8, and VEGFA was predicted to be a potential target of miR-130a-5p. Downregulation of MEG8 inhibited the cell viability, migration, and angiogenesis and the expression of VEGFA via negatively regulating miR-130a-5p of BMECs treated by OGD/non-OGD. In addition, MEG8 reduced cerebral ischemia, neurological score and miR-130a-5p expression, and increased VEGFA expression of MCAO rat. Our findings proved that MEG8 regulates angiogenesis and attenuates cerebral ischemia after ischemic stroke via miR-130a-5p/VEGFA signaling.


Assuntos
Isquemia Encefálica/metabolismo , AVC Isquêmico/metabolismo , MicroRNAs/biossíntese , Proteínas Tirosina Fosfatases não Receptoras/biossíntese , Fator A de Crescimento do Endotélio Vascular/biossíntese , Animais , Isquemia Encefálica/prevenção & controle , Células Cultivadas , Técnicas de Silenciamento de Genes/métodos , AVC Isquêmico/prevenção & controle , Masculino , Camundongos , MicroRNAs/antagonistas & inibidores , Ratos , Ratos Sprague-Dawley , Transdução de Sinais/fisiologia
15.
J Diabetes Complications ; 35(1): 107749, 2021 01.
Artigo em Inglês | MEDLINE | ID: mdl-33189541

RESUMO

LncRNA MEG8 can be induced by high glucose, indicating the potential role of this lncRNA in high glucose-induced diseases, such as gestational diabetes mellitus (GDM). In this study, a 6-year follow-up was performed on 400 females who had a plan for pregnancy. It was observed that patients with high pre-pregnancy plasma level of MEG8 showed high incidence rate of GDM during pregnancy. The patients with GDM had significantly higher levels of MEG8 in plasma. Plasm levels of MEG8 at one month before the diagnosis of GDM was sufficient to distinguish GDM patients from healthy controls. In addition, this study revealed that GDM patients who had higher level of MEG8 (on the day of discharge) showed significantly higher incidence of kidney injury. Therefore, these results suggest that MEG8 is upregulated in GDM and predicted kidney injury.


Assuntos
Diabetes Gestacional , Glicemia , Diabetes Gestacional/diagnóstico , Diabetes Gestacional/epidemiologia , Diabetes Gestacional/genética , Feminino , Glucose , Humanos , Incidência , Rim , Gravidez , RNA Longo não Codificante/genética , Fatores de Risco
16.
Diabetes Metab Syndr Obes ; 13: 2477-2483, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-32765026

RESUMO

BACKGROUND: It has been reported that lncRNA MEG8 can be induced by glucose in mice model of kidney injury, indicating its role in diabetic nephropathy (DN). This study was carried out to explore the role of MEG8 in DN. MATERIALS AND METHODS: The expression of MEG8 and miR-770-5p in plasma samples from DN patients (n = 66), diabetic patients (DM patients with no complications, n = 66) and healthy controls (n = 66) was detected by RT-qPCR. The interaction between MEG8 and miR-770-5p in podocyte cells was evaluated by transient transfections. Cell apoptosis under high-glucose treatment was detected by cell apoptosis assay. RESULTS: MEG8 and miR-770-5p were upregulated in plasma of DM patients and were further upregulated in DN patients. MEG8 was positively correlated with miR-770-5p. In podocyte cells, high-glucose treatment resulted in increased expression levels of MEG8 and miR-770-5p. In podocyte cells, overexpression of MEG8 resulted in upregulated expression of miR-770-5p and decreased methylation of the miR-770-5p gene. Cell apoptosis analysis showed that overexpression of MEG8 and miR-770-5p resulted in increased cell apoptotic rate under glucose treatment. In addition, combined overexpression of MEG8 and miR-770-5p showed stronger effects. CONCLUSION: MEG8 may upregulate miR-770-5p through methylation to promote DN by promoting cell apoptosis.

17.
J Cell Biochem ; 120(10): 17378-17390, 2019 10.
Artigo em Inglês | MEDLINE | ID: mdl-31265183

RESUMO

Long noncoding RNAs (lncRNAs) are a group of noncoding RNAs whose nucleotides are longer than 200 bp. Previous studies have shown that they play an important regulatory role in many developmental processes and biological pathways. However, the contributions of lncRNAs to placental development are largely unknown. Here, our study aimed to investigate the lncRNA expression signatures in placental development by performing a microarray lncRNA screen. Placental samples were obtained from pregnant C57BL/6 female mice at three key developmental time points (embryonic day E7.5, E13.5, and E19.5). Microarrays were used to analyze the differential expression of lncRNAs during placental development. In addition to the genomic imprinting region and the dynamic DNA methylation status during placental development, we screened imprinted lncRNAs whose expression was controlled by DNA methylation during placental development. We found that the imprinted lncRNA Rian may play an important role during placental development. Its homologous sequence lncRNA MEG8 (RIAN) was abnormally highly expressed in human spontaneous abortion villi. Upregulation of MEG8 expression in trophoblast cell lines decreased cell proliferation and invasion, whereas downregulation of MEG8 expression had the opposite effect. Furthermore, DNA methylation results showed that the methylation of the MEG8 promoter region was increased in spontaneous abortion villi. There was dynamic spatiotemporal expression of imprinted lncRNAs during placental development. The imprinted lncRNA MEG8 is involved in the regulation of early trophoblast cell function. Promoter methylation abnormalities can cause trophoblastic cell defects, which may be one of the factors that occurs in early unexplained spontaneous abortion.


Assuntos
Aborto Espontâneo/etiologia , Impressão Genômica , Placenta/patologia , RNA Longo não Codificante/genética , Trofoblastos/patologia , Aborto Espontâneo/patologia , Animais , Apoptose , Proliferação de Células , Células Cultivadas , Metilação de DNA , Feminino , Camundongos , Camundongos Endogâmicos C57BL , Placenta/metabolismo , Gravidez , Trofoblastos/metabolismo
18.
Biochem Biophys Res Commun ; 510(1): 171-176, 2019 02 26.
Artigo em Inglês | MEDLINE | ID: mdl-30670309

RESUMO

Atherosclerosis is the leading risk factor for cardiovascular disease, in which vascular smooth muscle cell (VSMC) proliferation and apoptosis play an important role. Research has demonstrated that long non-coding RNAs (lncRNAs) are critical regulatory factors for VSMC function, however, the molecular mechanism leading to this pathology is still not fully understood. To explore this further, an ox-LDL induced VSMC model was established, in which lncRNA MEG8 expression was suppressed, while miR-181a-5p was enhanced in a dose- and time-dependent manner. Enhanced MEG8 expression suppressed the proliferation and migration ability of VSMCs, and induced apoptosis. Mechanically, MEG8 was found to promote the PPARα protein via sponging miR-181a-5p. Rescue experiments demonstrated that MEG8 and PPARα collectively regulate the proliferation, migration and apoptosis of VSMCs. Overall, this research illustrates that MEG8 regulates the proliferation and migration of VSMCs via the MEG8/miR-181a/PPARα axis.


Assuntos
MicroRNAs/metabolismo , Músculo Liso Vascular/citologia , PPAR alfa/antagonistas & inibidores , RNA Longo não Codificante/fisiologia , Apoptose , Aterosclerose/patologia , Movimento Celular , Proliferação de Células , Células Cultivadas , Humanos , Lipoproteínas LDL/farmacologia , Modelos Teóricos , PPAR alfa/metabolismo , RNA Longo não Codificante/metabolismo
19.
Epigenomics ; 7(7): 1089-97, 2015 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-26541061

RESUMO

AIM: To investigate the DNA-methylation levels in the newly described MEG8 differentially methylated region (DMR) in the imprinted cluster in 14q32 in patients with Temple syndrome. PATIENTS & METHODS: We included three patients with Temple syndrome which were studied by Infinium HumanMethylation450 BeadChips, locus-specific bisulfite-pyrosequencing, methylation-specific-MLPA and microsatellite analyses. The tag-CpG of the MEG8-DMR was investigated using the Infinium HumanMethylation450 BeadChip. RESULTS: In all three patients, the identical pattern of DNA-hypermethylation of the MEG8-DMR was observed along with DNA-hypomethylation of the IG-DMR and MEG3-DMR. CONCLUSION: Based on the observed MEG8-DMR DNA-hypermethylation and previously published data, we conclude that DNA-methylation of the MEG3- and MEG8-DMR is functionally dependent on the DNA-methylation pattern of the IG-DMR. The observed combination of epimutations is predicted to be associated with bi-allelic MEG3 and MEG8 expression in individuals with Temple syndrome.


Assuntos
Cromossomos Humanos Par 14 , Deficiências do Desenvolvimento/genética , Epigênese Genética , Isoformas de RNA/genética , RNA Longo não Codificante/genética , Dissomia Uniparental/genética , Adolescente , Criança , Pré-Escolar , Ilhas de CpG , Metilação de DNA , Deficiências do Desenvolvimento/patologia , Feminino , Impressão Genômica , Sequenciamento de Nucleotídeos em Larga Escala , Humanos , Masculino , Repetições de Microssatélites , Reação em Cadeia da Polimerase , Dissomia Uniparental/patologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA