Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 71
Filtrar
1.
BMC Genomics ; 25(1): 526, 2024 May 28.
Artigo em Inglês | MEDLINE | ID: mdl-38807051

RESUMO

BACKGROUND: Alzheimer's disease (AD) is a complicated neurodegenerative disease. Neuron-glial cell interactions are an important but not fully understood process in the progression of AD. We used bioinformatic methods to analyze single-nucleus RNA sequencing (snRNA-seq) data to investigate the cellular and molecular biological processes of AD. METHOD: snRNA-seq data were downloaded from Gene Expression Omnibus (GEO) datasets and reprocessed to identify 240,804 single nuclei from healthy controls and patients with AD. The cellular composition of AD was further explored using Uniform Manifold Approximation and Projection (UMAP). Enrichment analysis for the functions of the DEGs was conducted and cell development trajectory analyses were used to reveal underlying cell fate decisions. iTALK was performed to identify ligand-receptor pairs among various cell types in the pathological ecological microenvironment of AD. RESULTS: Six cell types and multiple subclusters were identified based on the snRNA-seq data. A subcluster of neuron and glial cells co-expressing lncRNA-SNHG14, myocardin-related transcription factor A (MRTFA), and MRTFB was found to be more abundant in the AD group. This subcluster was enriched in mitogen-activated protein kinase (MAPK)-, immune-, and apoptosis-related pathways. Through molecular docking, we found that lncRNA-SNHG14 may bind MRTFA and MRTFB, resulting in an interaction between neurons and glial cells. CONCLUSIONS: The findings of this study describe a regulatory relationship between lncRNA-SNHG14, MRTFA, and MRTFB in the six main cell types of AD. This relationship may contribute to microenvironment remodeling in AD and provide a theoretical basis for a more in-depth analysis of AD.


Assuntos
Doença de Alzheimer , Neuroglia , Neurônios , Análise de Célula Única , Doença de Alzheimer/genética , Doença de Alzheimer/patologia , Doença de Alzheimer/metabolismo , Humanos , Neuroglia/metabolismo , Neuroglia/patologia , Neurônios/metabolismo , Microambiente Celular/genética , Biologia Computacional/métodos
2.
IUBMB Life ; 2024 Sep 12.
Artigo em Inglês | MEDLINE | ID: mdl-39266460

RESUMO

The functional role and molecular mechanisms of small-nucleolar RNA host gene 14 (SNHG14) in triple-negative breast cancer (TNBC) progression remain unclear. The expression levels of SNHG14 in breast cancer samples and cell lines were determined using real-time quantitative polymerase chain reaction. Cell proliferation, migration, and invasion abilities were detected using MTS and transwell assays. By RNA sequencing, differentially expressed genes were identified between the SNHG14 siRNA and the negative control group. Gene Ontology and Kyoto Encyclopedia of Genes and Genomes pathway analyses were used to predict the targets and pathways regulated by SNHG14. pRAF, pMEK, and pERK expression were measured by western blot. The xenograft model was constructed to access the biological function of SNHG14 in vivo. A minimal patient-derived xenograft model was established to evaluate the sensitivity to chemotherapy drugs. Our data indicated that SNHG14 expression was increased in TNBC tissues and cell lines. SNHG14 knockdown attenuated the proliferation, migration, and invasion abilities of TNBC cells both in vivo and in vitro. High SNHG14 expression was associated with lymph node metastasis and a high Ki67 index. The targets of SNHG14 were mainly enriched in the MAPK signaling pathway. pRAF, pMEK, and pERK expression were downregulated after being transfected with SNHG14 siRNA. Compared with the negative control group, the expression of CACNA1I, DUSP8, FGF17, FGFR4, FOS, PDGFRB, and DDIT3 was increased, and the expression of MKNK1 was decreased in the SNHG14 siRNA group. Minimal patient-derived xenograft model demonstrated that knockdown of SNHG14 enhanced the sensitivity to Docetaxel in vivo. Compared with the DMSO group, the proliferation of Docetaxel-resistant MDA-MB-231 cells was decreased in Dabrafenib, PD184352, and FR180204 treatment groups. SNHG14 knockdown inhibits TNBC progression by regulating the ERK/MAPK signaling pathway, which provides evidence for SNHG14 as a potential target for TNBC therapy.

3.
Neurochem Res ; 49(5): 1212-1225, 2024 May.
Artigo em Inglês | MEDLINE | ID: mdl-38381247

RESUMO

Parkinson's disease (PD) is a neurodegenerative disorder characterized by the loss of dopaminergic neurons. LncRNA small nucleolar RNA host gene 14 (SNHG14) was found to promote neuron injury in PD. Here, we investigated the mechanisms of SNHG14 in PD process. In vivo or in vitro PD model was established by using 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP)-induced mice or 1-methyl-4-phenylpyridinium (MPP +)-stimulated SK-N-SH cells. The expression of genes and proteins was measured by qRT-PCR and Western blot. In vitro assays were conducted using ELISA, CCK-8, colony formation, EdU, flow cytometry, and Western blot assays, respectively. The oxidative stress was evaluated by determining the production of superoxide dismutase (SOD) and malondialdehyde (MDA). The direct interactions between miR-375-3p and NFAT5 (Nuclear factor of activated T-cells 5) or SNHG14 was verified using dual-luciferase reporter and RNA immunoprecipitation (RIP) assays. SNHG14 and NFAT5 were elevated, while miR-375-3p was decreased in MPTP-mediated PD mouse model and MPP + -induced SK-N-SH cells. Knockdown of SNHG14 or NFAT5, or overexpression of miR-375-3p reversed MPP + -induced neuronal apoptosis, inflammation, and oxidative stress. Mechanistically, SNHG14 directly bound to miR-375, which targeted NFAT5. Inhibition of miR-375-3p abolished the inhibitory activity of SNHG14 knockdown on MPP + -evoked neuronal damage. Besides that, NFAT5 up-regulation counteracted the effects of miR-375-3p on MPP + -mediated neuronal damage. SNHG14 contributed to MPP + -induced neuronal injury by miR-375/NFAT5 axis, suggesting a new insight into the pathogenesis of PD.


Assuntos
Neurônios Dopaminérgicos , MicroRNAs , Doença de Parkinson , RNA Longo não Codificante , Animais , Camundongos , 1-Metil-4-fenilpiridínio , Apoptose/efeitos dos fármacos , Apoptose/genética , Linhagem Celular Tumoral , Neurônios Dopaminérgicos/metabolismo , Inflamação/induzido quimicamente , Inflamação/genética , Inflamação/metabolismo , MicroRNAs/genética , MicroRNAs/metabolismo , Estresse Oxidativo , Doença de Parkinson/genética , Doença de Parkinson/metabolismo , RNA Longo não Codificante/genética , RNA Longo não Codificante/metabolismo , Fatores de Transcrição/genética , Fatores de Transcrição/metabolismo
4.
J Biochem Mol Toxicol ; 37(6): e23343, 2023 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-37009739

RESUMO

Obesity is a metabolic disease with excess weight. LncRNA SNHG14 is abnormally expressed in numerous diseases. This research aimed to enucleate the lncRNA SNHG14 role in obesity. Adipocytes were treated with free fatty acid (FFA) to establish an in vitro model for obesity. Mice were fed a high-fat diet to construct an in vivo model. Gene levels were determined using quantitative real-time PCR (RT-PCR). The protein level was checked by western blot. The lncRNA SNHG14 role in obesity was assessed using western blot and enzyme-linked immunosorbent assay. The mechanism was estimated by Starbase, dual-luciferase reporter gene assay, and RNA pull-down. LncRNA SNHG14 function in obesity was estimated using mouse xenograft models, RT-PCR, western blot, and enzyme-linked immunosorbent assay. LncRNA SNHG14 and BACE1 levels were increased, but the miR-497a-5p level was decreased in FFA-induced adipocytes. Interference with lncRNA SNHG14 reduced endoplasmic reticulum (ER) stress-related molecules GRP78 and CHOP expressions in FFA-induced adipocytes, and decreased IL-1ß, IL-6, and TNF-α expressions, indicating that lncRNA SNHG14 knockdown mitigated FFA-induced ER stress and inflammation in adipocytes. Mechanistically, lncRNA SNHG14 combined with miR-497a-5p, and miR-497a-5p targeted BACE1. Meanwhile, lncRNA SNHG14 knockdown reduced levels of GRP78, CHOP, IL-1ß, IL-6, and TNF-α, while cotransfection with anti-miR-497a-5p or pcDNA-BACE1 abolished these trends. Rescue assays illustrated that lncRNA SNHG14 knockdown relieved FFA-induced adipocyte ER stress and inflammation through miR-497a-5p/BACE1. Meanwhile, lncRNA SNHG14 knockdown restrained adipose inflammation and ER stress caused by obesity in vivo. LncRNA SNHG14 mediated obesity-induced adipose inflammation and ER stress through miR-497a-5p/BACE1.


Assuntos
MicroRNAs , RNA Longo não Codificante , Humanos , Camundongos , Animais , MicroRNAs/genética , MicroRNAs/metabolismo , RNA Longo não Codificante/genética , RNA Longo não Codificante/metabolismo , Fator de Necrose Tumoral alfa/metabolismo , Chaperona BiP do Retículo Endoplasmático , Secretases da Proteína Precursora do Amiloide/genética , Interleucina-6 , Ácido Aspártico Endopeptidases , Obesidade/genética , Estresse do Retículo Endoplasmático , Inflamação/genética , Apoptose
5.
Dig Dis Sci ; 67(3): 936-946, 2022 03.
Artigo em Inglês | MEDLINE | ID: mdl-33782806

RESUMO

OBJECTIVE: To explore how lncRNA SNHG14 modulates the biological features of hepatocellular carcinoma (HCC) cells by regulating SOX9 via mediating miR-206. METHODS: HCC tissues were collected to perform the quantitative reverse transcriptase polymerase chain reaction to determine the expressions of SNHG14, miR-206, and SOX9. HCC cell line SMCC7721 was selected for co-transfection by si-SNHG14/miR-206 inhibitor/si-SOX9, followed by the measurement of cell proliferation using Cell Count Kit-8 (CCK-8) assay and clone formation assay. The migration and invasion were evaluated by wound healing test and Transwell assay. The apoptotic rate was determined by flow cytometry. Levels of the apoptosis-related proteins were measured through Western blotting. RESULTS: SNHG14 and SOX9 were up-regulated in HCC tumor tissues compared with adjacent normal tissues, with decreased miR-206 expression. Moreover, SNHG14 expression was significantly associated with the TNM stage, lymphatic metastasis, and histological differentiation of HCC patients. Besides, inverse correlations between SNHG14 and miR-206, as well as between miR-206 and SOX9, were noted. The dual luciferase reporter gene assay, RIP, and RNA pull-down experiments also revealed the targeting relationship between SNHG14 and miR-206 or between miR-206 and SOX9. Silencing SNHG14 and SOX9 inhibited the proliferation, invasion, and migration of HCC cells, with increased apoptosis, which was all abolished by silencing miR-206. CONCLUSION: Inhibition of SNHG14 suppresses SOX9 by up-regulating miR-206, to further inhibit the proliferation, migration, and invasion of HCC cells with the promoted apoptosis, which is a novel target for the treatment of HCC.


Assuntos
Carcinoma Hepatocelular , Neoplasias Hepáticas , MicroRNAs , RNA Longo não Codificante , Apoptose , Carcinoma Hepatocelular/patologia , Linhagem Celular , Linhagem Celular Tumoral , Movimento Celular , Proliferação de Células , Regulação Neoplásica da Expressão Gênica , Humanos , Neoplasias Hepáticas/patologia , MicroRNAs/genética , MicroRNAs/metabolismo , RNA Longo não Codificante/genética , RNA Longo não Codificante/metabolismo , Fatores de Transcrição SOX9/genética , Fatores de Transcrição SOX9/metabolismo
6.
Metab Brain Dis ; 37(7): 2363-2373, 2022 10.
Artigo em Inglês | MEDLINE | ID: mdl-35781593

RESUMO

BACKGROUND: Parkinson's disease (PD) is a neurodegenerative disease resulted from the loss of dopaminergic neurons. Here, we analyzed the role of long noncoding RNA (lncRNA) small nucleolar RNA host gene 14 (SNHG14) in PD using 1-methyl-4-phenyl pyridine (MPP+)-induced PD cell model. METHODS: Reverse transcription-quantitative polymerase chain reaction (RT-qPCR) and Western blot assay were performed to determine RNA and protein expression, respectively. 3-(4,5-Dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide (MTT) assay and flow cytometry (FCM) analysis were conducted to analyze cell viability and apoptosis. Enzyme-Linked Immunosorbent Assay (ELISA) was conducted to analyze the release of inflammatory cytokines. Cytotoxicity was assessed using reactive oxygen species (ROS) assay kit, superoxide dismutase (SOD) activity assay kit and lactate dehydrogenase (LDH) activity assay kit. Dual-luciferase reporter assay and RNA immunoprecipitation (RIP) assay were performed to confirm the interaction between microRNA-135b-5p (miR-135b-5p) and SNHG14 or karyopherin subunit alpha 4 (KPNA4). RESULTS: MPP+ treatment elevated the expression of SNHG14 in SK-N-SH cells in a dose and time-dependent manner. SNHG14 knockdown alleviated MPP+-induced apoptosis, inflammation, and cytotoxicity in SK-N-SH cells. SNHG14 interacted with miR-135b-5p, and SNHG14 silencing-mediated effects were partly overturned by miR-135b-5p knockdown in PD cell model. Besides, miR-135b-5p interacted with the 3' untranslated region (3'UTR) of KPNA4, and KPNA4 overexpression partly reversed miR-135b-5p overexpression-induced effects in PD cell model. SNHG14 knockdown reduced the protein level of KPNA4 partly by up-regulating miR-135b-5p in SK-N-SH cells. CONCLUSION: SNHG14 promoted MPP+-induced neuro injury in PD cell model through mediating miR-135b-5p/KPNA4 axis.


Assuntos
MicroRNAs , Doenças Neurodegenerativas , Doença de Parkinson , RNA Longo não Codificante , Humanos , RNA Longo não Codificante/genética , RNA Longo não Codificante/metabolismo , Espécies Reativas de Oxigênio/metabolismo , Regiões 3' não Traduzidas , Doença de Parkinson/genética , Doença de Parkinson/metabolismo , MicroRNAs/genética , MicroRNAs/metabolismo , RNA Nucleolar Pequeno/farmacologia , Apoptose , Superóxido Dismutase/metabolismo , Piridinas/farmacologia , Citocinas/metabolismo , Lactato Desidrogenases/genética , Lactato Desidrogenases/metabolismo , Carioferinas/genética , Carioferinas/farmacologia , alfa Carioferinas/genética
7.
Metab Brain Dis ; 37(6): 2005-2016, 2022 08.
Artigo em Inglês | MEDLINE | ID: mdl-35678981

RESUMO

BACKGROUND: The deregulation of long non-coding RNA (lncRNA) is associated with diverse human disorders, including cerebral ischemia/reperfusion injury (CI/RI). LncRNA SNHG14 was reported to function in CI/RI. Whereas, molecular mechanisms regulated by SNHG14 are not fully unveiled. METHODS: Mice subjected to middle cerebral artery occlusion/reperfusion (MCAO/R) were used as CI/RI animal models. Neuro-2a (N2A) cells subjected to oxygen glucose deprivation/reoxygenation (OGD/R) were used as CI/RI cell models. The expression of SNHG14, miR-98-5p and BCL2 like 13 (BCL2L13) was examined using quantitative real-time PCR (qPCR) or western blot. Apoptosis was monitored by flow cytometry assay. Apoptosis-related markers and endoplasmic reticulum (ER) stress-related markers were quantified by western blot. Inflammatory factors and oxidative stress were detected using matched commercial kits. The predicted relationship between miR-98-5p and SNHG14 or BCL2L13 was validated by dual-luciferase reporter assay, RIP assay and pull-down assay. RESULTS: The high expression of SNHG14 was monitored in MCAO/R-treated mice and OGD/R-treated N2A cells. OGD/R-induced N2A cell apoptosis, ER stress, inflammation and oxidative stress were attenuated by SNHG14 knockdown. SNHG14 targeted miR-98-5p to positively regulate BCL2L13 expression. Inhibition of miR-98-5p recovered cell apoptosis, ER stress, inflammation and oxidative stress that were repressed by SNHG14 knockdown. Overexpression of BCL2L13 enhanced cell apoptosis, ER stress, inflammation and oxidative stress that were repressed by miR-98-5p enrichment. CONCLUSIONS: SNHG14 knockdown alleviated OGD/induced N2A cell apoptosis, ER stress, inflammation and oxidative stress by depleting BCL2L13 via increasing miR-98-5p.


Assuntos
Isquemia Encefálica , Hipóxia , MicroRNAs , RNA Longo não Codificante , Traumatismo por Reperfusão , Animais , Apoptose/fisiologia , Isquemia Encefálica/genética , Glucose/metabolismo , Humanos , Hipóxia/metabolismo , Infarto da Artéria Cerebral Média/genética , Inflamação , Camundongos , MicroRNAs/genética , MicroRNAs/metabolismo , Oxigênio/metabolismo , Proteínas Proto-Oncogênicas c-bcl-2/genética , Proteínas Proto-Oncogênicas c-bcl-2/metabolismo , RNA Longo não Codificante/genética , RNA Longo não Codificante/metabolismo , Traumatismo por Reperfusão/genética , Traumatismo por Reperfusão/metabolismo , Regulação para Cima
8.
Zygote ; 30(2): 183-193, 2022 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-34380584

RESUMO

Long non-coding RNAs (lncRNAs) exert vital functions in the occurrence and development of various tumours. The aim of this study was to examine the regulatory effect and underlying molecular mechanism of lncRNA small nucleolar RNA host gene 14 (SNHG14) on the proliferation, invasion and migration of thyroid tumour cells. The expression of SNHG14 in thyroid tumour cell lines was determined using qRT-PCR. CCK-8 and western blot were used to detect the effects of SNHG14 on proliferation and apoptosis of thyroid tumour cells. The effect of SNHG14 on the migration and invasion of thyroid tumour cells was analyzed using immunofluorescence, wound-healing and transwell assays. A targeting relationship between SNHG14 and miR-93-5p was determined using bioinformatics software and luciferase reporter assays. In addition, CCK-8, immunofluorescence, wound-healing and transwell assays were applied to demonstrate that SNHG14 promoted the proliferation, migration and invasion of thyroid tumour cells by targeting miR-93-5p. The biological function of SNHG14 in vivo was explored through a xenograft model and immunohistochemistry. SNHG14 was upregulated in thyroid tumour cells compared with normal cells. Downregulation of SNHG14 effectively reduced the proliferation, migration and invasion of TPC-1 cells, and induced cell apoptosis. Moreover, SNHG14 directly targeted miR-93-5p and there was a negative correlation between them. Further functional experiments illustrated that miR-93-5p overexpression dramatically reversed the promoting role of SNHG14 in proliferation, migration and invasion of TPC-1 cells. Our results demonstrated that SNHG14 promotes the proliferation, invasion and migration of thyroid tumour cells by downregulating miR-93-5p.


Assuntos
MicroRNAs , RNA Longo não Codificante , Neoplasias da Glândula Tireoide , Movimento Celular/genética , Proliferação de Células/genética , Humanos , MicroRNAs/genética , RNA Longo não Codificante/genética , RNA Longo não Codificante/metabolismo , Neoplasias da Glândula Tireoide/genética
9.
Arch Gynecol Obstet ; 305(6): 1507-1516, 2022 06.
Artigo em Inglês | MEDLINE | ID: mdl-34783894

RESUMO

PURPOSE: Dysregulation of long non-coding RNAs (lncRNAs) is being found to have relevance to human cancers, including breast cancer (BC). The aim of this study was to further explore the functional role and molecular mechanisms of small nucleolar RNA host gene 14 (SNHG14) on BC progression. METHODS: The expression levels of SNHG14, miR-543, and krüppel-like factor 7 (KLF7) mRNA were determined by quantitative real-time PCR. Western blot analysis was used to evaluate KLF7 protein level. Cell proliferation, apoptosis, and migration and invasion abilities were detected by Cell Counting kit-8 assay, flow cytometry, and transwell assay, respectively. The direct interactions between miR-543 and SNHG14 or KLF7 were confirmed using dual-luciferase reporter assays. RESULTS: Our data indicated that SNHG14 expression was increased in BC tissues and cells, and SNHG14 knockdown mitigated the proliferation, migration, and invasion and facilitated apoptosis of BC cells. SNHG14 directly interacted with miR-543. MiR-543 mediated the regulatory effects of SNHG14 silencing on BC cell behaviors. Moreover, KLF7 was a direct target of miR-543. Overexpressed miR-543-mediated anti-proliferation, anti-migration, anti-invasion, and pro-apoptosis effects were mediated by KLF7. Furthermore, SNHG14 modulated KFL7 expression through acting as a competing endogenous RNA (ceRNA) of miR-543 in BC cells. CONCLUSION: Our study suggested that SNHG14 knockdown hindered BC progression in vitro at least partly through acting as a ceRNA of miR-543 and modulating KLF7 expression, providing evidence for SNHG14 as a potential target for BC therapy.


Assuntos
Neoplasias da Mama , Fatores de Transcrição Kruppel-Like , MicroRNAs , RNA Longo não Codificante , Apoptose/genética , Neoplasias da Mama/genética , Proliferação de Células/genética , Feminino , Humanos , Fatores de Transcrição Kruppel-Like/genética , MicroRNAs/genética , RNA Longo não Codificante/genética
10.
Int J Neurosci ; 132(1): 77-88, 2022 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-33045891

RESUMO

OBJECTIVE: Microglial activation is an essential pathological mechanism of spinal cord ischemia-reperfusion injury (SCIRI). Previous studies showed dexmedetomidine (DEX) could alleviate SCIRI while the mechanism was not clear. This study aims to investigate the role of DEX in microglial activation and clarify the underlying mechanism. METHODS: The motion function of mice was quantified using the Basso Mouse Scale for Locomotion. The expression of long non-coding RNA (lncRNA) small nucleolar RNA host gene 14 (SNHG14) was determined by qRT-PCR. The expression of high-mobility group box 1 (HMGB1) was measured by western blot. The activation of microglia was evaluated by the expression of ED-1 and the levels of TNF-α and IL-6. The interplay between SNHG14 and HMGB1 was confirmed with RNA pull-down and RIP assay. The stability of HMGB1 was measured by ubiquitination assay and cycloheximide-chase assay. RESULTS: DEX inhibited microglial activation and down-regulated SNHG14 expression in SCIRI mice and oxygen and glucose deprivation/reoxygenation (OGD/R)-treated primary microglia. Functionally, SNHG14 overexpression reversed the inhibitory effect of DEX on OGD/R-induced microglial activation. Further investigation confirmed that SNHG14 bound to HMGB1, positively regulated HMGB1 expression by enhancing its stability. In addition, the silence of HMGB1 eliminated the pro-activation impact of SNHG14 overexpression on DEX-treated microglia under the OGD/R condition. Finally, in vivo experiments showed SNHG14 overexpression abrogated the therapeutic effect of DEX on SCIRI mice by up-regulating HMGB1. CONCLUSION: DEX accelerated HMGB1 degradation via down-regulating SNHG14, thus inhibiting microglial activation in SCIRI mice.


Assuntos
Dexmedetomidina/farmacologia , Proteína HMGB1/efeitos dos fármacos , Microglia/efeitos dos fármacos , RNA Longo não Codificante/efeitos dos fármacos , Traumatismo por Reperfusão/tratamento farmacológico , Doenças Vasculares da Medula Espinal/tratamento farmacológico , Animais , Comportamento Animal , Modelos Animais de Doenças , Locomoção/efeitos dos fármacos , Camundongos , Transdução de Sinais/efeitos dos fármacos
11.
Neurochem Res ; 46(5): 1280-1290, 2021 May.
Artigo em Inglês | MEDLINE | ID: mdl-33609254

RESUMO

BACKGROUND: Ischemic stroke is the leading cause of disability worldwide. Long noncoding RNAs (lncRNAs) play important roles in the pathogenesis of cerebral ischemia. This study aimed to investigate the role and mechanism of lncRNA small nucleolar RNA host gene 14 (SNHG14) in ischemic brain injury. METHODS: Cerebral ischemia was induced by middle cerebral artery occlusion (MCAO) in mice. The expression of SNHG14 in MCAO mouse model was detected by quantitative real-time PCR (qRT-PCR). The levels of SNHG14, microRNA-199b (miR-199b) and aquaporin 4 (AQP4) in oxygen-glucose deprivation (OGD)-stimulated BV2 cells were determined by qRT-PCR or western blot assay. Cell proliferation and apoptosis were assessed by Cell Counting Kit-8 (CCK-8) assay and flow cytometry. The levels of pro-inflammatory cytokines were measured by enzyme-linked immunosorbent assay (ELISA). The levels of oxidative stress markers were examined using commercial kits. The relationships among SNHG14, miR-199b and AQP4 were confirmed by dual-luciferase reporter assay, RNA immunoprecipitation assay and RNA pull-down assay. RESULTS: SNHG14 was up-regulated in MCAO mouse model. Depletion of SNHG14 lessened cerebral ischemia in MCAO mouse model. SNHG14 silencing inhibited inflammation and oxidative stress in OGD-exposed BV2 cells. MiR-199b level was decreased, while AQP4 level was increased in OGD-treated BV2 cells. Knockdown of miR-199b reversed the effect of SNHG14 knockdown on ischemic damage in OGD-stimulated BV2 cells. Moreover, AQP4 overexpression abolished the effect of miR-199b on ischemic injury in OGD-treated BV2 cells. Furthermore, SNHG14 indirectly regulate AQP4 expression by sponging miR-199b. CONCLUSIONS: Knockdown of SNHG14 attenuated ischemic brain injury by inhibiting inflammation and oxidative stress through the miR-199b/AQP4 axis.


Assuntos
Aquaporina 4/metabolismo , Isquemia Encefálica/metabolismo , Infarto da Artéria Cerebral Média/metabolismo , MicroRNAs/metabolismo , RNA Longo não Codificante/metabolismo , Animais , Isquemia Encefálica/etiologia , Linhagem Celular , Técnicas de Silenciamento de Genes , Infarto da Artéria Cerebral Média/complicações , Inflamação/etiologia , Inflamação/metabolismo , Masculino , Camundongos Endogâmicos C57BL , Microglia , Estresse Oxidativo/fisiologia , RNA Longo não Codificante/genética
12.
J Surg Res ; 263: 140-150, 2021 07.
Artigo em Inglês | MEDLINE | ID: mdl-33652176

RESUMO

BACKGROUND: Emerging evidence show that long noncoding RNAs (lncRNAs) are crucial regulators in pathophysiology of acute lung injury (ALI). Small nucleolar RNA host gene 14 (SNHG14) is a novel oncogenic lncRNA, and has been associated with inflammation-related cell injuries. Thus, we wondered the role and mechanism of SNHG14 in lipopolysaccharides (LPS)-induced ALI cell model. METHODS: Expression of SNHG14, miRNA (miR)-124-3p, and transforming growth factor ß type 2 receptor (TGFBR2) was detected by RT-qPCR and western blotting. Cell apoptosis was determined by methyl thiazolyl tetrazolium assay, flow cytometry, western blotting, and lactate dehydrogenase activity kit. Inflammation was measured by enzyme-linked immunosorbent assay. The interaction among SNHG14, miR-124-3p, and TGFBR2 was validated by dual-luciferase reporter assay and RNA immunoprecipitation. RESULTS: LPS administration attenuated human lung epithelial cell viability and B-cell lymphoma-2 expression, but augmented apoptosis rate, cleaved-caspase-3 expression, lactate dehydrogenase activity, and secretions of tumor necrosis factor-α, interleukin-1ß, and IL-6 in A549 cells. Thus, LPS induced A549 cells apoptosis and inflammation, wherein SNHG14 was upregulated and miR-124-3p was downregulated. However, silencing SNHG14 could suppress LPS-induced apoptosis and inflammation depending on upregulating miR-124-3p via target binding. Similarly, overexpressing miR-124-3p attenuated LPS-induced A549 cells injury through inhibiting its downstream target TGFBR2. Furthermore, SNHG14 knockdown could also affect TGFBR2 expression via miR-124-3p. CONCLUSIONS: SNHG14 knockdown prevents A549 cells from LPS-induced apoptosis and inflammation through regulating miR-124-3p and TGFBR2, suggesting a novel SNHG14/miR-124-3p/TGFBR2 circuit in alveolar epithelial cells on the set of ALI.


Assuntos
Lesão Pulmonar Aguda/genética , MicroRNAs/metabolismo , RNA Longo não Codificante/metabolismo , Receptor do Fator de Crescimento Transformador beta Tipo II/genética , Células A549 , Lesão Pulmonar Aguda/imunologia , Apoptose/genética , Apoptose/imunologia , Sobrevivência Celular/genética , Sobrevivência Celular/imunologia , Regulação para Baixo/imunologia , Perfilação da Expressão Gênica , Técnicas de Silenciamento de Genes , Humanos , Lipopolissacarídeos/imunologia , MicroRNAs/agonistas , MicroRNAs/genética , RNA Longo não Codificante/genética , Regulação para Cima/imunologia
13.
J Biol Regul Homeost Agents ; 35(2): 605-615, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-33928771

RESUMO

Osteogenic differentiation of human bone marrow-derived mesenchymal stem cells (hBMSCs) is vital for bone formation, and its dysfunction is linked to osteoporosis (OP). In this work, we explored the function of long non-coding RNA (lncRNA) small nucleolar RNA host gene 14 (SNHG14) in regulating osteogenic differentiation of hBMSCs. In the present study, the expression of SNHG14 in hBMSCs obtained from OP patients was measured by quantitative real-time polymerase chain reaction (qRT-PCR). SNHG14 was over-expressed or knocked down in hBMSCs, and the expression levels of OP-related genes (ALP, OCN, and OPN) in hBMSCs were detected by qRT-PCR and Western blot. StarBase database and miRanda database were used to predict the binding sites between SNHG14 and miR-185-5p, and between miR-185-5p and 3'UTR of WNT1 inducible signaling pathway protein 2 (WISP2), respectively. Luciferase reporter gene assay was used to validate the binding relationship between SNHG14 and miR-185-5p, and miR-185-5p and 3'UTR of WISP2, respectively. Here, we report that SNHG14 was significantly down-regulated in hBMSCs obtained from patients with OP. Overexpression of SNHG14 promoted osteogenic differentiation, while knockdown of SNHG14 worked oppositely. Mechanistically, miR-185-5p was demonstrated to be a target of SNHG14, and could reverse the function of SNHG14. Additionally, WISP2 was identified as a target gene of miR-185-5p in hBMSCs and could be indirectly regulated by SNHG14. Taken together, down-regulation of SNHG14 in hBMSCs accelerated the progression of OP via regulating miR-185-5p/WISP2 axis.


Assuntos
Células-Tronco Mesenquimais , MicroRNAs , RNA Longo não Codificante , Medula Óssea , Diferenciação Celular , Células Cultivadas , Humanos , MicroRNAs/genética , Osteogênese/genética , RNA Longo não Codificante/genética
14.
Acta Biochim Biophys Sin (Shanghai) ; 53(6): 719-728, 2021 May 21.
Artigo em Inglês | MEDLINE | ID: mdl-33856026

RESUMO

Acute kidney injury (AKI) is a complex syndrome with an abrupt decrease of kidney function, which is associated with high morbidity and mortality. Sepsis is the common cause of AKI. Mounting evidence has demonstrated that long non-coding RNAs (lncRNAs) play critical roles in the development and progression of sepsis-induced AKI. In this study, we aimed to illustrate the function and mechanism of lncRNA SNHG14 in lipopolysaccharide (LPS)-induced AKI. We found that SNHG14 was highly expressed in the plasma of sepsis patients with AKI. SNHG14 inhibited cell proliferation and autophagy and promoted cell apoptosis and inflammatory cytokine production in LPS-stimulated HK-2 cells. Functionally, SNHG14 acted as a competing endogenous RNA (ceRNA) to negatively regulate miR-495-3p expression in HK-2 cells. Furthermore, we identified that HIPK1 is a direct target of miR-495-3p in HK-2 cells. We also revealed that the SNHG14/miR-495-3p/HIPK1 interaction network regulated HK-2 cell proliferation, apoptosis, autophagy, and inflammatory cytokine production upon LPS stimulation. In addition, we demonstrated that the SNHG14/miR-495-3p/HIPK1 interaction network regulated the production of inflammatory cytokines (TNF-α, IL-6, and IL-1ß) via modulating NF-κB/p65 signaling in LPS-challenged HK-2 cells. In conclusion, our findings suggested a novel therapeutic axis of SNHG14/miR-495-3p/HIPK1 to treat sepsis-induced AKI.


Assuntos
Injúria Renal Aguda/sangue , Injúria Renal Aguda/induzido quimicamente , Células Epiteliais/metabolismo , Lipopolissacarídeos/efeitos adversos , MicroRNAs/metabolismo , Proteínas Serina-Treonina Quinases/metabolismo , RNA Longo não Codificante/sangue , Sepse/sangue , Transdução de Sinais/genética , Apoptose/genética , Autofagia/genética , Estudos de Casos e Controles , Linhagem Celular Transformada , Proliferação de Células/genética , Citocinas/biossíntese , Células Epiteliais/efeitos dos fármacos , Técnicas de Silenciamento de Genes , Humanos , Túbulos Renais/citologia , MicroRNAs/genética , Proteínas Serina-Treonina Quinases/genética , RNA Longo não Codificante/genética , Sepse/complicações , Transfecção
15.
Zygote ; 29(2): 108-117, 2021 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-33161910

RESUMO

Preeclampsia (PE), a pregnancy-specific disease, has become one of the leading causes of maternal and neonatal morbidity and mortality. Pathogenesis of PE has still not been fully addressed and there is a great need to develop early diagnosis markers and effective therapy. This study aimed to determine if lncRNA SNHG14 has a protective effect on placental trophoblast and prevents PE. SNHG14 levels in the peripheral blood from patients with PE or from women with healthy pregnancies were detected using RT-qPCR. The relationship between SNHG14 and miR-330-5p was determined using a dual-luciferase reporter assay. In addition, cell proliferation and cell cycle were evaluated by performing CCK8 assays and flow-cytometric analysis, respectively. Wound-healing and transwell assays were performed to assess cell migration and invasion ability. lncRNA SNHG14 was downregulated in PE patients; it was involved in trophoblast proliferation and regulated cell proliferation during G1/S transition. In addition, lncRNA SNHG14 promoted migration, invasion and epithelial-mesenchymal transition (EMT) in HTR-8/SVneo cells. Luciferase reporter assay indicated that lncRNA SNHG14 served as a molecular sponge for miR-330-5p and negatively regulated miR-330-5p expression in PE. Furthermore, the effects of silenced SNHG14 on trophoblast proliferation, migration, invasion and EMT were reversed by addition of miR-330-5p inhibitor, suggesting that in PE lncRNA SNHG14 functions by competitively binding to miR-330-5p. Taken together, the current study demonstrated that in PE lncRNA SNHG14 is a vital regulator by binding to miR-330-5p. SNHG14 might serve as a therapeutic application in PE progression.


Assuntos
MicroRNAs , Pré-Eclâmpsia , RNA Longo não Codificante , Movimento Celular , Proliferação de Células , Transição Epitelial-Mesenquimal , Feminino , Humanos , Recém-Nascido , Placenta , Gravidez , Trofoblastos
16.
Environ Toxicol ; 36(12): 2484-2492, 2021 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-34529319

RESUMO

Rheumatoid arthritis (RA) is a widespread autoimmune disorder of the joints. Long noncoding RNAs (lncRNAs) have been reported to participate in the pathogenesis of RA by serving as competitive endogenous RNAs. LncRNA small nucleolar RNA host gene 14 (SNHG14) is involved in the development of various diseases. Here, we found that high expression of SNHG14 in RA was closely related to the disease activity. Functional assays indicated that SNHG14 knockdown obviously hampered phorbol myristate acetate-activated THP-1 (pTHP-1) cell proliferation and proinflammatory cytokines production. In mechanism, SNHG14 served as a sponge of microRNA-17-5p (miR-17-5p), and misshapen like kinase 1 (MINK1) was a target of miR-17-5p. SNHG14 depletion-induced inhibitory effects on cell proliferation and inflammatory response were reversed by MINK1 overexpression in macrophages. Moreover, SNHG14 promoted the jun N-terminal kinase (JNK) signaling via the miR-17-5p/MINK1 axis. Overall, SNHG14 boosted the process of RA by MINK1 activating the JNK pathway.


Assuntos
Artrite Reumatoide , MicroRNAs , Proteínas Serina-Treonina Quinases/metabolismo , RNA Longo não Codificante , Artrite Reumatoide/genética , Proliferação de Células , Citocinas/genética , Humanos , Sistema de Sinalização das MAP Quinases , MicroRNAs/genética , MicroRNAs/metabolismo , RNA Longo não Codificante/genética , RNA Longo não Codificante/metabolismo
17.
J Cell Mol Med ; 24(13): 7115-7126, 2020 07.
Artigo em Inglês | MEDLINE | ID: mdl-32436661

RESUMO

Cardiac hypertrophy (CH) is a common cardiac disease and is closely associated with heart failure. Protocadherin 17 (PCDH17) was reported to aggravate myocardial infarction. Present study was designed to illustrate the impact of PCDH17 and the mechanism of PCDH17 expression regulation in CH. CH model in vivo and in vitro was established by transverse aortic constriction (TAC) and Ang-II treatment. Hypertrophy was evaluated in PMC and H9c2 cells by examining cell surface area and hypertrophic markers. Results demonstrated that PCDH17 was up-regulated in CH in vivo and in vitro. PCDH17 knock-down alleviated hypertrophic response in Ang-II-induced cardiomyocytes. By means of ENCORI database and a series of mechanism assays, miR-322-5p and miR-384-5p were identified to interact with and inhibit PCDH17. Next, lncRNA SNHG14 (small nucleolar RNA host gene 14) was validated to sponge both miR-322-5p and miR-384-5p to elevate PCDH17 level. The subsequent rescue assays revealed that miR-322-5p and miR-384-5p restored SNHG14 depletion-mediated suppression on hypertrophy in Ang-II-induced cardiomyocytes. Besides, Sp1 transcription factor (SP1) was verified as the transcription factor activating both SNHG14 and PCDH17. Both SNHG14 and PCDH17 reversed SP1 knock-down-mediated repression on hypertrophy in Ang-II-induced cardiomyocytes. Together, present study first uncovered ceRNA network of SNHG14/miR-322-5p/miR-384-5p/PCDH17 in Ang-II-induced cardiomyocytes.


Assuntos
Caderinas/genética , Cardiomegalia/genética , RNA Longo não Codificante/genética , Fator de Transcrição Sp1/metabolismo , Regulação para Cima/genética , Angiotensina II , Animais , Sequência de Bases , Caderinas/metabolismo , Inativação Gênica , Masculino , Camundongos Endogâmicos C57BL , MicroRNAs/genética , MicroRNAs/metabolismo , Modelos Biológicos , Miócitos Cardíacos/metabolismo , Protocaderinas , RNA Longo não Codificante/metabolismo
18.
J Cell Biochem ; 121(2): 1998-2008, 2020 02.
Artigo em Inglês | MEDLINE | ID: mdl-31692034

RESUMO

Ample evidence have demonstrated that long noncoding RNAs small nucleolus RNA host gene 14 (SNHG14) serves as a master regulator in various cancers. However, the exact mechanism of SNHG14 in colorectal cancer (CRC) remains unknown. In the present study, we concentrate on the potential function of SNHG14 in the pathogenesis of CRC. From the quantitative reverse transcription-polymerase chain reaction results, SNHG14 was found to be downregulated in CRC tissues compared with the normal mucous samples, and its low expression was significantly correlated with poor clinical outcomes. Overexpression of SNHG14 inhibited cell growth, induced cell apoptosis, suppressed migration and invasion by inhibiting epithelial-mesenchymal transition process. Furthermore, mechanistic studies revealed that miR-92b-3p could rescue the CRC progress induced by SNHG14. Consequently, SNHG14 exhibited low expression in CRC tissues and involved in CRC progression and metastasis by competing for miR-92b-3p, and SNHG14 could be used as a valuable biomarker and therapeutic target for CRC.


Assuntos
Biomarcadores Tumorais/genética , Neoplasias Colorretais/patologia , Regulação Neoplásica da Expressão Gênica , MicroRNAs/genética , RNA Longo não Codificante/genética , Apoptose , Biomarcadores Tumorais/metabolismo , Proliferação de Células , Neoplasias Colorretais/genética , Neoplasias Colorretais/metabolismo , Transição Epitelial-Mesenquimal , Feminino , Humanos , Metástase Linfática , Masculino , Pessoa de Meia-Idade , Prognóstico , Células Tumorais Cultivadas
19.
Biochem Biophys Res Commun ; 523(3): 766-772, 2020 03 12.
Artigo em Inglês | MEDLINE | ID: mdl-31948764

RESUMO

Long noncoding RNA small nucleolus RNA host gene 14 (SNHG14) has been shown to exert oncogenic functions in seceral cancers, but its role in osteosarcoma still unclear. In this present study, we found that SNHG14 was significantly upregulated in osteosarcoma tissues and cell lines. Knockdown SNHG14 expression significantly inhibited osteosarcoma cell proliferation through inducing apoptosis. Further functional assays revealed that SNHG14 knockdown dramatically suppressed cell migration and invasion. Bioinformatics analysis and luciferase assays identified that microRNA-433-3p (miR-433-3p) was a direct target of SNHG14, and directly targeted F-box only protein 22 (FBXO22). Mechanistic analysis demonstrated that SNHG14 acted as a ceRNA in modulating osteosarcoma proliferation, migration and invasion by decoying miR-433-3p to upregulate FBXO22 expression. We also observed that knockdown of FBXO22 and SNHG14 and overexpression of miR-433-3p both dramatically suppressed osteosarcoma cell proliferation, migration and invasion, but induced cell apoptosis. Moreover, the suppresive effect of SNHG14 knockdown on osteosarcoma cell proliferation, invasion and migration was attenuated by miR-433-3p inhibitor. Our findings demonstrated that SNHG14 promoted osteosarcoma progression by acting as a ceRNA for miR-433-3p to regulate FBXO22 expression, suggesting that SNHG14 may serve as a potential therapeutic target in osteosarcoma patients.


Assuntos
Neoplasias Ósseas/genética , Proteínas F-Box/genética , MicroRNAs/genética , Osteossarcoma/genética , RNA Longo não Codificante/genética , Receptores Citoplasmáticos e Nucleares/genética , Neoplasias Ósseas/patologia , Linhagem Celular Tumoral , Proliferação de Células , Progressão da Doença , Regulação Neoplásica da Expressão Gênica , Humanos , Invasividade Neoplásica/genética , Invasividade Neoplásica/patologia , Osteossarcoma/patologia
20.
Biol Res ; 53(1): 38, 2020 Sep 10.
Artigo em Inglês | MEDLINE | ID: mdl-32912324

RESUMO

BACKGROUND: Long non-coding RNA (lncRNA) small nucleolar RNA host gene 14 (SNHG14) is associated with cerebral ischemia-reperfusion (CI/R) injury. This work aims to explore the role of SNHG14 in CI/R injury. METHODS: HT22 (mouse hippocampal neuronal cells) cell model was established by oxygen-glucose deprivation/reoxygenation (OGD/R) treatment. The interaction among SNHG14, miR-182-5p and BNIP3 was verified by luciferase reporter assay. Flow cytometry, western blot and quantitative real-time PCR were performed to examine apoptosis, the expression of genes and proteins. RESULTS: SNHG14 and BNIP3 were highly expressed, and miR-182-5p was down-regulated in the OGD/R-induced HT22 cells. OGD/R-induced HT22 cells exhibited an increase in apoptosis. SNHG14 overexpression promoted apoptosis and the expression of cleaved-caspase-3 and cleaved-caspase-9 in the OGD/R-induced HT22 cells. Moreover, SNHG14 up-regulation enhanced the expression of BNIP3, Beclin-1, and LC3II/LC3I in the OGD/R-induced HT22 cells. Furthermore, SNHG14 regulated BNIP3 expression by sponging miR-182-5p. MiR-182-5p overexpression or BNIP3 knockdown repressed apoptosis in OGD/R-induced HT22 cells, which was abolished by SNHG14 up-regulation. CONCLUSION: Our study demonstrates that lncRNA SNHG14 promotes OGD/R-induced neuron injury by inducing excessive mitophagy via miR-182-5p/BINP3 axis in HT22 mouse hippocampal neuronal cells. Thus, SNHG14/miR-182-5p/BINP3 axis may be a valuable target for CI/R injury therapies.


Assuntos
Hipocampo/citologia , Proteínas de Membrana/genética , MicroRNAs/genética , Proteínas Mitocondriais/genética , Neurônios/patologia , RNA Longo não Codificante , Traumatismo por Reperfusão/genética , Animais , Linhagem Celular , Camundongos , Mitofagia , RNA Longo não Codificante/genética
SELEÇÃO DE REFERÊNCIAS
Detalhe da pesquisa