Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 73
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
FASEB J ; 38(15): e23880, 2024 Aug 15.
Artículo en Inglés | MEDLINE | ID: mdl-39132919

RESUMEN

Long noncoding RNAs (lncRNAs) are strongly associated with glucose homeostasis, but their roles remain largely unknown. In this study, the potential role of lncRNA-Snhg3 in glucose metabolism was evaluated both in vitro and in vivo. Here, we found a positive relationship between Snhg3 and hepatic glycogenesis. Glucose tolerance improved in hepatocyte-specific Snhg3 knock-in (Snhg3-HKI) mice, while it worsened in hepatocyte-specific Snhg3 knockout (Snhg3-HKO) mice. Furthermore, hepatic glycogenesis had shown remarkable increase in Snhg3-HKI mice and reduction in Snhg3-HKO mice, respectively. Mechanistically, Snhg3 increased mRNA and protein expression levels of PPP1R3B through inducing chromatin remodeling and promoting the phosphorylation of protein kinase B. Collectively, these results suggested that lncRNA-Snhg3 plays a critical role in hepatic glycogenesis.


Asunto(s)
Hígado , ARN Largo no Codificante , Animales , ARN Largo no Codificante/genética , ARN Largo no Codificante/metabolismo , Ratones , Hígado/metabolismo , Ratones Noqueados , Glucosa/metabolismo , Masculino , Hepatocitos/metabolismo , Ratones Endogámicos C57BL , Glucógeno Hepático/metabolismo
2.
Cancer Sci ; 115(7): 2286-2300, 2024 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-38680094

RESUMEN

SNHG3, a long noncoding RNA (lncRNA), has been linked to poor outcomes in patients with liver hepatocellular carcinoma (LIHC). In this study, we found that SNHG3 was overexpressed in LIHC and associated with poor outcomes in patients with LIHC. Functional assays, including colony formation, spheroid formation, and in vivo assays showed that SNHG3 promoted stemness of cancer stem cells (CSC) and tumor growth in vivo by interacting with microRNA-502-3p (miR-502-3p). miR-502-3p inhibitor repressed the tumor-suppressing effects of SNHG3 depletion. Finally, by RNA pull-down, dual-luciferase reporter assay, m6A methylation level detection, and m6A-IP-qPCR assays, we found that miR-502-3p targeted YTHDF3 to regulate the translation of integrin alpha-6 (ITGA6) and targeted HBXIP to inhibit the m6A modification of ITGA6 through methyltransferase-like 3 (METTL3). Our study revealed that SNHG3 controls the YTHDF3/ITGA6 and HBXIP/METTL3/ITGA6 pathways by repressing miR-502-3p expression to sustain the self-renewal properties of CSC in LIHC.


Asunto(s)
Carcinoma Hepatocelular , Regulación Neoplásica de la Expresión Génica , Integrina alfa6 , Neoplasias Hepáticas , MicroARNs , Células Madre Neoplásicas , ARN Largo no Codificante , Animales , Femenino , Humanos , Masculino , Ratones , Persona de Mediana Edad , Carcinoma Hepatocelular/genética , Carcinoma Hepatocelular/patología , Carcinoma Hepatocelular/metabolismo , Línea Celular Tumoral , Proliferación Celular/genética , Integrina alfa6/metabolismo , Integrina alfa6/genética , Neoplasias Hepáticas/genética , Neoplasias Hepáticas/patología , Neoplasias Hepáticas/metabolismo , Metiltransferasas/metabolismo , Metiltransferasas/genética , Ratones Desnudos , MicroARNs/genética , MicroARNs/metabolismo , Células Madre Neoplásicas/metabolismo , Células Madre Neoplásicas/patología , ARN Largo no Codificante/genética , ARN Largo no Codificante/metabolismo
3.
Mod Rheumatol ; 2024 May 06.
Artículo en Inglés | MEDLINE | ID: mdl-38722030

RESUMEN

OBJECTIVES: This study evaluated the expression and significance of SNHG3 in Rheumatoid arthritis (RA) aiming to explore a biomarker and regulator for RA. METHODS: The expression of SNHG3 in serum and synovial tissue was compared between RA patients and healthy individuals using PCR. The RA animal models were induced by the porcine type II collagen with Wistar rats and validated by the foot volume and AI score. The human fibroblast-like synoviocytes (H-FLS) were treated with LPS to mimic the injury during RA onset and the cell growth was assessed by CCK8 assay. RESULTS: SNHG3 was significantly downregulated in the serum and synovial tissue of RA patients compared with healthy individuals. Downregulated SNHG3 could discriminate RA patients from healthy individuals with high sensitivity (0.875) and specificity (0.844). Porcine type II collagen induced increasing foot volume and AI scores of rats and SNHG3 was downregulated in RA rats. In LPS-induced H-FLS, SNHG3 negatively regulated miR-128-3p, and the alleviated effect of SNHG3 overexpression on cellular inflammation and oxidative stress was reversed by miR-128-3p upregulation. CONCLUSIONS: Serum SNHG3 was considered a potential diagnostic biomarker for RA from healthy individuals. SNHG3 regulated inflammatory response and oxidative stress via negatively modulating miR-128-3p.

4.
Immunogenetics ; 75(1): 39-51, 2023 02.
Artículo en Inglés | MEDLINE | ID: mdl-36114381

RESUMEN

The involvement of small nucleolar RNA host gene 3 (SNHG3) in cancer regulation has been reported. This study attempted to deeply investigate the molecular regulatory mechanism of SNHG3 on malignant progression of hepatocellular carcinoma (HCC). According to TCGA analysis, high SNHG3 expression was a risk factor for poor prognosis of HCC patients. Therefore, we further detected the mRNA level of SNHG3 in HCC tissue and cells. It was found that SNHG3 was upregulated in HCC tissue and cells. Afterwards, CCK-8 and flow cytometry assays further proved that silencing SNHG3 inhibited HCC cell proliferation while inducing cell apoptosis and G0/G1 phase arrest. It was also attested in vivo experiments that silencing SNHG3 could reduce the volume and weight of tumors and downregulate the Ki-67 expression to suppress HCC tumor growth. Next, it was discovered that SNHG3 increased the binding of E2F1 and NEIL3 promoter region, thereby activating the transcription feature of NEIL3. Lastly, rescue assays indicated that NEIL3 participated in SNHG3-mediated HCC cell cycle, apoptosis and proliferation. All in all, this study revealed the specific regulatory mechanism of SNHG3 in HCC to enable SNHG3 a hopeful marker for HCC diagnosis and treatment.


Asunto(s)
Carcinoma Hepatocelular , Neoplasias Hepáticas , MicroARNs , Humanos , Carcinoma Hepatocelular/genética , Carcinoma Hepatocelular/patología , Factor de Transcripción E2F1/genética , Factor de Transcripción E2F1/metabolismo , Neoplasias Hepáticas/genética , Neoplasias Hepáticas/patología , Proliferación Celular/genética , ARN Mensajero/genética , Regulación Neoplásica de la Expresión Génica , Línea Celular Tumoral , MicroARNs/genética
5.
Biochem Biophys Res Commun ; 639: 36-45, 2023 01 08.
Artículo en Inglés | MEDLINE | ID: mdl-36463759

RESUMEN

Barrier permeability changes of human pulmonary microvascular endothelial cells (HPMVECs) are important in sepsis-related acute lung injury (ALI) pathogenesis. Long non-coding small nucleolar RNA host gene 3 (SNHG3) mediates the cell-biological phenotype of lung cancer cells and affects the progression of lung cancer, but its role in regulating functions of lung non-malignant cells is still rarely reported. Therefore, we evaluated the regulatory effect of SNHG3 on the function of PMVECs in sepsis-related ALI. Small interference RNA (siRNA)-mediated deletion of SNHG3 promoted the proliferation of PMVECs, reduced apoptosis and barrier permeability, and increased the expression of tight junction proteins claudin-5 and ZO-1. Knockdown of SNHG3 increased the miR-186-5p expression, while overexpression of SNHG3 upregulated the level of wnt5a. Through a dual luciferase reporter assay, we confirmed the binding between SNHG3 and miR-186-5p, miR-186-5p and wnt5a. We further found that knockout of miR-186-5p could inhibit cell proliferation, increase apoptosis and barrier permeability, and down-regulate claudin-5 and ZO-1. Importantly, silencing miR-186-5p and activating Wnt signal pathway could eliminate the barrier repair effect caused by down-regulation of SNHG3. To sum up, our results suggested that knockdown of long non-coding RNA SNHG3 repaired the dysfunction of pulmonary microvascular endothelial barrier through the miR-186-5p/Wnt axis.


Asunto(s)
Lesión Pulmonar Aguda , Neoplasias Pulmonares , MicroARNs , ARN Largo no Codificante , Sepsis , Humanos , Lesión Pulmonar Aguda/etiología , Proliferación Celular/genética , Claudina-5/genética , Células Endoteliales/metabolismo , Pulmón/metabolismo , MicroARNs/genética , MicroARNs/metabolismo , ARN Largo no Codificante/genética , ARN Largo no Codificante/metabolismo , Sepsis/complicaciones , Proteínas Wnt/metabolismo
6.
Neurochem Res ; 48(9): 2754-2766, 2023 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-37093343

RESUMEN

BACKGROUND: Early exposure to sevoflurane may cause brain tissue degeneration; however, the mechanism involved in this process has not been explored. In this study, we investigated the role of long non-coding RNA small nucleolar RNA host gene 3 (lncRNA SNHG3) in sevoflurane-induced neuronal injury. METHODS: The injury models of HT22 and primary cultures of neurons were constructed using sevoflurane treatment. The WST-8 reduction was detected by CCK-8 assay, the level of inflammatory factors was detected by enzyme-linked immunosorbent assay (ELISA), and cell pyroptosis was detected by flow cytometry. The expression of genes and proteins was detected by qRT-PCR and Western blot, respectively. The level of ß-tubulin III in primary cultures of hippocampal neurons was analyzed by immunofluorescence. The relationship among SNHG3, PTBP1 and NEK7 was confirmed by RIP assay. RESULTS: The expression of SNHG3 and NEK7 were enhanced in sevoflurane-treated HT22 cells. Sevoflurane inhibited the WST-8 reduction in a concentration-dependent manner, promoted the pyroptosis, and increased pyroptosis-related protein expression. SNHG3 knockdown significantly inhibited sevoflurane-induced pyroptosis and inflammatory injury in HT22 cells and primary cultures of neurons. Furthermore, SNHG3 regulated NEK7 expression by binding to PTBP1. NEK7 knockdown reversed the decrease in WST-8 reduction, inhibited pyroptosis, and decreased the release of inflammatory factors and pyroptosis-related protein expression by inactivation of NLRP3 signaling in sevoflurane-induced HT22 cells. Moreover, NEK7 overexpression attenuated the effect of SNHG3 knockdown on neuronal pyroptosis and inflammation injury. CONCLUSION: Downregulation of SNHG3 attenuates sevoflurane-induced neuronal inflammation and pyroptosis by mediating the NEK7/NLRP3 axis, suggesting that SNHG3 could be a potential target gene for neuronal injury.


Asunto(s)
MicroARNs , ARN Largo no Codificante , Humanos , Proteína con Dominio Pirina 3 de la Familia NLR/metabolismo , ARN Largo no Codificante/genética , ARN Largo no Codificante/metabolismo , Sevoflurano/toxicidad , Inflamación/inducido químicamente , Inflamación/metabolismo , Neuronas/metabolismo , MicroARNs/metabolismo , Ribonucleoproteínas Nucleares Heterogéneas/metabolismo , Proteína de Unión al Tracto de Polipirimidina/metabolismo , Quinasas Relacionadas con NIMA/metabolismo
7.
Cell Mol Biol Lett ; 27(1): 13, 2022 Feb 05.
Artículo en Inglés | MEDLINE | ID: mdl-35123415

RESUMEN

In recent years, morbidity and mortality of prostate cancer (PCa) have increased dramatically, while mechanistic understanding of its onset and progression remains unmet. LncRNA SNHG3 has been proved to stimulate malignant progression of multiple cancers, whereas its functional mechanism in PCa needs to be deciphered. In this study, our analysis in the TCGA database revealed high SNHG3 expression in PCa tissue. Further analysis in starBase, TargetScan, and mirDIP databases identified the SNHG3/miR-152-3p/SLC7A11 regulatory axis. FISH was conducted to assess the distribution of SNHG3 in PCa tissue. Dual-luciferase reporter gene and RIP assays confirmed the relationship among the three objects. Next, qRT-PCR and western blot were conducted to measure expression levels of SNHG3, miR-152-3p, and SLC7A11. CCK-8, colony formation, Transwell, and flow cytometry were carried out to assess proliferation, migration, invasion, methionine dependence, apoptosis, and the cell cycle. It was noted that SNHG3 as a molecular sponge of miR-152-3p stimulated proliferation, migration, and invasion, restrained methionine dependence and apoptosis, and affected the cell cycle of PCa cells via targeting SLC7A11. Additionally, we constructed xenograft tumor models in nude mice and confirmed that knockdown of SNHG3 could restrain PCa tumor growth and elevate methionine dependence in vivo. In conclusion, our investigation improved understanding of the molecular mechanism of SNHG3 modulating PCa progression, thereby generating novel insights into clinical therapy for PCa.


Asunto(s)
MicroARNs , Neoplasias de la Próstata , ARN Largo no Codificante , Animales , Línea Celular Tumoral , Movimiento Celular/genética , Proliferación Celular/genética , Regulación Neoplásica de la Expresión Génica , Humanos , Masculino , Metionina/genética , Metionina/metabolismo , Ratones , Ratones Desnudos , MicroARNs/genética , MicroARNs/metabolismo , Neoplasias de la Próstata/genética , ARN Largo no Codificante/genética , ARN Largo no Codificante/metabolismo
8.
Metab Brain Dis ; 37(6): 2061-2066, 2022 08.
Artículo en Inglés | MEDLINE | ID: mdl-35622264

RESUMEN

mTOR has been shown to be involved in the regulation of immune responses and differentiation of immune cells. This protein is a candidate molecule for unraveling the molecular mechanisms of autoimmune disorders such as multiple sclerosis (MS). We designed the current study to assess expression of MTOR, and four associated long non-coding RNAs (lncRNAs), namely SNHG1, SNHG3, SHNG5 and DANCR in the peripheral blood of patients with MS compared with healthy controls. Analysis of real-time PCR results has shown down-regulation of SNHG5 and DANCR in MS patients compared with controls. Sex of study participants had no significant effect on expression of either genes and the interaction of sex and disease on expression levels of all studied genes were insignificant. There was a significant negative correlation between expression levels of MTOR gene and disease duration. No other significant correlations were detected between genes expressions and clinical/demographic data. SNHG5 and DANCR transcript levels had AUC values of 0.88 and 0.68 in separation of patients with MS from healthy controls, respectively. Taken together, our study suggests participation of two mTOR-related lncRNAs, i.e. SNHG5 and DANCR in the pathophysiology of MS.


Asunto(s)
Esclerosis Múltiple , ARN Largo no Codificante , Diferenciación Celular , Regulación hacia Abajo , Humanos , Esclerosis Múltiple/genética , ARN Largo no Codificante/genética , ARN Largo no Codificante/metabolismo , Serina-Treonina Quinasas TOR/genética , Serina-Treonina Quinasas TOR/metabolismo
9.
Cancer Cell Int ; 21(1): 114, 2021 Feb 17.
Artículo en Inglés | MEDLINE | ID: mdl-33596916

RESUMEN

BACKGROUND: Platinum-based chemotherapy is a mainstay for treating esophageal cancer patients. In this manuscript, we have provided clues for influence of platinum on overall m6A level and further investigated the potential regulatory mechanism. METHODS: qRT-PCR was used to measure SNHG3 and miR-186-5p expression; ELISA and western blot were used to measure the expression of METTL3. CCK8 was used to measure the cell proliferation rate. Caspase 3/7 activity was used to measure the apoptosis rate. Dual luciferase reporter gene assay and RNA pull down assay were used to investigate the potential crosstalk between miR-186-5p and SNHG3; and miR-186-5p and METTL3. RESULTS: m6A level was increased when treated with platinum (CDDP, CPB and L-OHP). Besides, SNHG3 expression was induced and miR-186-5p expression was suppressed by platinum. Furthermore, SNHG3 could promote the m6A level, however miR-186-5p inhibited the m6A level through targeting METTL3. SNHG3 interacts with miR-186-5p to negatively regulate the expression of miR-186-5p; and miR-186-5p might bind to the 3'UTR of METTL3 to regulate its expression. CONCLUSION: Platinum can increase the overall m6A level of esophageal cancer. SNHG3/miR-186-5p, induced by platinum, was involved in regulating m6A level by targeting METTL3. Our manuscript has provided clues that regulating m6A level might be a novel way to enhance the platinum efficacy.

10.
Oral Dis ; 27(5): 1161-1170, 2021 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-32989886

RESUMEN

OBJECTIVE: The potential molecular mechanism underlying the disease progression of oral squamous cell carcinoma (OSCC) remains largely elusive. The purpose of the study is to figure out the role and molecular mechanism of homeobox B8 (HOXB8) in OSCC. MATERIALS AND METHODS: The expression level of HOXB8 in OSCC was validated by RT-qPCR. The functions of HOXB8 in OSCC cells were identified through loss-of-function assays, including CCK-8 assay, colony formation assay, transwell assay, and immunofluorescence (IF). The upstream miRNA that could directly target HOXB8 was searched out through bioinformatics analysis and luciferase reporter assay. Mechanism experiments were further conducted to predict the long non-coding RNA (lncRNA) that could positively regulate HOXB8 and compete for miR-2682-5p with HOXB8. RESULTS: HOXB8 was markedly upregulated in OSCC tissues and cell lines. Furthermore, cell proliferation and migration were inhibited due to the shortage of HOXB8. HOXB8 was targeted by miR-2682-5p that negatively regulated cell proliferation and migration. Small nucleolar RNA host gene 3 (SNHG3) acted as a sponge for miR-2682-5p. Inhibition of miR-2682-5p or the overexpression of HOXB8 rescued the effects of SNHG3 silencing on the proliferation and migration. CONCLUSION: HOXB8 is regulated by SNHG3/miR-2682-5p axis to promote OSCC cell proliferation and migration.


Asunto(s)
Carcinoma de Células Escamosas , Neoplasias de Cabeza y Cuello , MicroARNs , Neoplasias de la Boca , ARN Largo no Codificante , Carcinoma de Células Escamosas/genética , Línea Celular Tumoral , Proliferación Celular/genética , Regulación Neoplásica de la Expresión Génica , Genes Homeobox , Proteínas de Homeodominio/genética , Humanos , MicroARNs/genética , Neoplasias de la Boca/genética , ARN Largo no Codificante/genética , Carcinoma de Células Escamosas de Cabeza y Cuello
11.
Environ Toxicol ; 36(6): 1070-1079, 2021 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-33522089

RESUMEN

Glaucoma is the main reason for irreversible blindness, and pathological increased intraocular pressure is the leading risk factor for glaucoma. It is reported that trabecular meshwork cell injury is closely associated with the elevated intraocular pressure. The current study aimed to investigate the role of small nucleolar RNA host gene 3 (SNHG3) in human trabecular meshwork (HTM) cells under oxidative stress. A series of experiments including real-time quantitative polymerase chain reaction, subcellular fractionation assay, western blot analysis, cell counting kit-8 assay, RNA pull down, flow cytometry analysis, and RNA immunoprecipitation assay were used to explore the biological function and regulatory mechanism of SNHG3 in HTM cells under oxidative stress. First, we observed that H2 O2 induced SNHG3 upregulation in HTM cells. Then, we found that SNHG3 silencing alleviated H2 O2 -induced oxidative damage in HTM cells. Moreover, snail family transcriptional repressor 2 (SNAI2) knockdown alleviated the oxidative damage induced by H2 O2 in HTM cells. Mechanistically, SNHG3 bound with ELAV like RNA binding protein 2 (ELAVL2) to stabilize SNAI2. Finally, SNAI2 overexpression counteracted the effect of SNHG3 silencing on H2 O2 -treated HTM cells. In conclusion, our results demonstrated that SNHG3 cooperated with ELAVL2 to modulate cell apoptosis and extracellular matrix accumulation by stabilizing SNAI2 in HTM cells under oxidative stress.


Asunto(s)
Estrés Oxidativo , Malla Trabecular , Apoptosis , Células Cultivadas , Matriz Extracelular/metabolismo , Humanos , Factores de Transcripción de la Familia Snail/genética , Factores de Transcripción de la Familia Snail/metabolismo , Regulación hacia Arriba
12.
J Cell Mol Med ; 24(16): 9231-9243, 2020 08.
Artículo en Inglés | MEDLINE | ID: mdl-32596993

RESUMEN

Growing evidence suggests that long non-coding RNAs (lncRNAs) are associated with carcinogenesis. LncRNA small nucleolar RNA host gene 3 (SNHG3) is up-regulated in various cancers and positively associated with poor prognosis of these cancers. However, the precise role of lncRNA SNHG3 in bladder cancer (Bca) remains unclear. In our research, we first reported that lncRNA SNHG3 was up-regulated in bladder cancer tissues and positively related to poor clinical prognosis. Moreover, knockdown of lncRNA SNHG3 significantly suppressed the proliferation, migration, invasion and EMT process of Bca cells in vitro and vivo. Mechanistically, we revealed that suppression of SNHG3 evidently enhanced miR-515-5p expression and decreased GINS2 expression at posttranscriptional levels. Moreover, SNHG3 positively regulated GINS2 expression by sponging miR-515-5p under a competing endogenous RNA (ceRNA) mechanism. To sum up, our study suggested lncRNA SNHG3 acted as a microRNA sponge and an oncogenic role in the progression of bladder cancer.


Asunto(s)
Biomarcadores de Tumor/metabolismo , Proteínas Cromosómicas no Histona/metabolismo , Regulación Neoplásica de la Expresión Génica , MicroARNs/genética , ARN Largo no Codificante/genética , Neoplasias de la Vejiga Urinaria/patología , Animales , Apoptosis , Biomarcadores de Tumor/genética , Proliferación Celular , Proteínas Cromosómicas no Histona/genética , Progresión de la Enfermedad , Transición Epitelial-Mesenquimal , Femenino , Humanos , Metástasis Linfática , Masculino , Ratones , Ratones Endogámicos BALB C , Ratones Desnudos , Persona de Mediana Edad , Pronóstico , Tasa de Supervivencia , Células Tumorales Cultivadas , Neoplasias de la Vejiga Urinaria/genética , Neoplasias de la Vejiga Urinaria/metabolismo , Ensayos Antitumor por Modelo de Xenoinjerto
13.
J Cell Physiol ; 235(10): 6615-6624, 2020 10.
Artículo en Inglés | MEDLINE | ID: mdl-32048306

RESUMEN

Small nucleolar RNA host gene 3 (SNHG3) is a long noncoding RNA (lncRNA), which is known to promote oncogenesis in many cancers but its role in human papillary thyroid carcinoma (PTC) remains poorly understood. We therefore assessed SNHG3 expression in PTC tissues via quantitative reverse transcription polymerase chain reaction. We additionally knocked down SNHG3 in PTC cells using short-hairpin RNAs (shRNAs) to explore its functional roles in PTC. The ability of SNHG3 to bind to specific microRNAs (miRNAs) was predicted using a bioinformatics tool, and this binding was confirmed via dual-luciferase reporter and RNA immunoprecipitation (RIP) assays. We then used a tumor xenograft model to assess the relevance of SNHG3 in vivo. We determined SNHG3 expression to be elevated in PTC tissues relative to controls, with advanced tumor-node-metastasis stage and lymph node metastasis being associated with this expression. Knocking down SNHG3 significantly reduced in vitro PTC cell migration, invasion, proliferation, and colony formation, and it further slowed the growth of tumors in vivo. We found that SNHG3 could bind to miR-214-3p as a competing endogenous RNA (ceRNA) for this miRNA, thereby regulating proteasome 26S subunit non-ATPase 10 (PSMD10) expression, a miR-214-3p target. These results thus indicate that SNHG3 is an oncogenic lncRNA in PTC, acting at least in part via the miR-214-3p/PSMD10 axis.


Asunto(s)
MicroARNs/genética , Complejo de la Endopetidasa Proteasomal/genética , Proteínas Proto-Oncogénicas/genética , ARN Largo no Codificante/genética , Cáncer Papilar Tiroideo/genética , Neoplasias de la Tiroides/genética , Animales , Carcinogénesis/genética , Línea Celular , Movimiento Celular/genética , Proliferación Celular/genética , Progresión de la Enfermedad , Femenino , Regulación Neoplásica de la Expresión Génica/genética , Humanos , Masculino , Ratones , Ratones Endogámicos BALB C , Ratones Desnudos , Persona de Mediana Edad , ARN Interferente Pequeño/genética , Cáncer Papilar Tiroideo/patología , Neoplasias de la Tiroides/patología
14.
J Cell Physiol ; 235(3): 2891-2900, 2020 03.
Artículo en Inglés | MEDLINE | ID: mdl-31602642

RESUMEN

Recently, long noncoding RNAs (lncRNAs) have been widely reported to play pivotal roles in the regulation of human cancers. Although the oncogenic property of lncRNA small nucleolar RNA host gene 3 (SNHG3) has been revealed in a variety of cancers, functions and regulatory mechanism of SNHG3 in non-small-cell lung cancer (NSCLC) remain to be investigated. In this study, we detected the upregulated expression of SNHG3 in NSCLC tissues as well as cells through quantitative reverse-transcription polymerase chain reaction (qRT-PCR) analysis. Using Kaplan-Meier analysis, we determined that a high-level of SNHG3 was associated with a low overall survival rate of patients with NSCLC. Through gain and loss of function experiments, we demonstrated that SNHG3 had a significantly positive effect on NSCLC cell proliferation and migration. Mechanistic investigations revealed that SNHG3 was a predicted direct transcriptional target of E2F1. We observed that the transcriptional activation of SNHG3 could be induced by E2F1. To explore the mechanism, rescue experiments were carried out, which revealed that the cotreatment with SB-431542, JSI-124, or JSI-124 + SB-431542 rescued the effects brought by the overexpression of SNHG3 on NSCLC cell proliferation, migration, and epithelial-mesenchymal transition process. Our results suggested that E2F1 activated SNHG3 and promoted cell proliferation and migration in NSCLC via transforming growth factor-ß pathway and interleukin-6/janus-activated kinase 2/signal transducer and activator of transcription 3 pathway, which implied that SNHG3 may be a biomarker for the treatment of patients with NSCLC.


Asunto(s)
Factor de Transcripción E2F1/metabolismo , ARN Largo no Codificante/genética , Transducción de Señal/genética , Factor de Crecimiento Transformador beta/metabolismo , Animales , Carcinoma de Pulmón de Células no Pequeñas/genética , Línea Celular Tumoral , Proliferación Celular/genética , Factor de Transcripción E2F1/genética , Transición Epitelial-Mesenquimal/genética , Regulación Neoplásica de la Expresión Génica/genética , Humanos , Interleucina-6/metabolismo , Janus Quinasa 2/metabolismo , Neoplasias Pulmonares/genética , Ratones , Factor de Transcripción STAT3/genética , Factor de Transcripción STAT3/metabolismo , Regulación hacia Arriba
15.
J Cell Biochem ; 121(3): 2150-2158, 2020 03.
Artículo en Inglés | MEDLINE | ID: mdl-31762107

RESUMEN

Oral squamous cell carcinoma (OSCC) has been reported to be the most common oral carcinoma. Emerging evidence has revealed the key role that long noncoding RNAs (lncRNAs) play in numerous malignancies, including OSCC. LncRNA small nucleolar RNA host gene 3 (SNHG3) has been reported as an oncogenic factor in some cancers. Nonetheless, the role of SNHG3 in OSCC has never been clarified. In this study, we analyzed the expression patterns of SNHG3 in OSCC through quantitative real-time polymerase chain reaction. It was revealed that the expression level of SNHG3 was remarkably elevated in OSCC cell lines compared with the nontumor cell line. It was demonstrated by functional experiments that SNHG3 knockdown notably inhibited cell proliferation and migration in OSCC. RNA immunoprecipitation, RNA pull down, and messenger RNA (mRNA) stability test verified that SNHG3 decoyed ELAV like RNA-binding protein 1 (ELAVL1) and therefore stabilized nuclear transcription factor Y subunit gamma (NFYC) mRNA to upregulate the expression levels of NFYC in OSCC cells. At last, it was confirmed by rescue experiments that the inhibiting impacts of SNHG3 knockdown on OSCC cell proliferation and migration could be partly revived by NFYC overexpression. Besides, we validated that Wnt/ß-catenin pathway was also involved in SNHG3-regulated OSCC progression. In conclusion, SNHG3 might serve as a novel biomarker for OSCC.


Asunto(s)
Biomarcadores de Tumor/metabolismo , Factor de Unión a CCAAT/metabolismo , Carcinoma de Células Escamosas/patología , Proteína 1 Similar a ELAV/metabolismo , Regulación Neoplásica de la Expresión Génica , Neoplasias de la Boca/patología , ARN Largo no Codificante/genética , Apoptosis , Biomarcadores de Tumor/genética , Factor de Unión a CCAAT/química , Factor de Unión a CCAAT/genética , Carcinoma de Células Escamosas/genética , Carcinoma de Células Escamosas/metabolismo , Movimiento Celular , Proliferación Celular , Proteína 1 Similar a ELAV/genética , Humanos , Neoplasias de la Boca/genética , Neoplasias de la Boca/metabolismo , Estabilidad del ARN , Células Tumorales Cultivadas , Vía de Señalización Wnt , beta Catenina/genética , beta Catenina/metabolismo
16.
J Cell Biochem ; 121(2): 1023-1031, 2020 02.
Artículo en Inglés | MEDLINE | ID: mdl-31452272

RESUMEN

Small nucleolar RNA host gene 3 (SNHG3) is a newly identified long non-coding RNA whose dysregulation has been reported in several cancers. However, the details about clinical significances and biological functions of SNHG3 on acute myeloid leukemia (AML) remain covered. In this study, we revealed increased SNHG3 expression in AML samples and cells and its high potential as a prognostic biomarker for AML patients. Likewise, serglycin (SRGN), which plays an important role in granule-mediated apoptosis, was previously verified to be upregulated in AML and confirmed again by the present study, and its upregulation predicted poor outcomes in AML. Furthermore, knockdown of SNHG3 or SRGN inhibited cell proliferation and induced cell apoptosis. Besides, silencing SNHG3 noticeably decreased the expression of SRGN in AML cells. Moreover, we uncovered that SNHG3 modulated SRGN expression by competitively binding with miR-758-3p. Importantly, both miR-758-3p suppression and SRGN overexpression could mitigate the inhibitory effects of SNHG3 depletion on AML cell growth. Intriguingly, the higher SRGN expression in AML samples with a higher SNHG3 level exhibited an enhanced Ki67 level but a reduced caspase 3 level. To sum up, SNHG3 elicits a growth-promoting function in AML via sponging miR-758-3p to regulate SRGN expression, providing a new therapeutic road for AML patients.


Asunto(s)
Biomarcadores de Tumor/metabolismo , Regulación Neoplásica de la Expresión Génica , Leucemia Mieloide Aguda/patología , MicroARNs/genética , Proteoglicanos/metabolismo , ARN Largo no Codificante/genética , Proteínas de Transporte Vesicular/metabolismo , Apoptosis , Biomarcadores de Tumor/genética , Proliferación Celular , Femenino , Humanos , Leucemia Mieloide Aguda/genética , Leucemia Mieloide Aguda/metabolismo , Masculino , Persona de Mediana Edad , Pronóstico , Proteoglicanos/genética , Tasa de Supervivencia , Células Tumorales Cultivadas , Proteínas de Transporte Vesicular/genética
17.
Cancer Cell Int ; 20(1): 536, 2020 Nov 04.
Artículo en Inglés | MEDLINE | ID: mdl-33292213

RESUMEN

Long noncoding RNAs (lncRNAs) are composed of > 200 nucleotides; they lack the ability to encode proteins but play important roles in a variety of human tumors. A large number of studies have shown that dysregulated expression of lncRNAs is related to tumor oncogenesis and progression. Emerging evidence shows that SNHG3 is a novel oncogenic lncRNA that is abnormally expressed in various tumors, including osteosarcoma, liver cancer, lung cancer, etc. SNHG3 primarily competes as a competitive endogenous RNA (ceRNA) that targets tumor suppressor microRNAs (miRNAs) and ceRNA mechanisms that regulate biological processes of tumors. In addition, abnormal expression of SNHG3 is significantly correlated with patient clinical features. Upregulation of SNHG3 contributes to biological functions, including tumor cell proliferation, migration, invasion and EMT. Therefore, SNHG3 may represent a potential diagnostic and prognostic biomarker, as well as a novel therapeutic target.

18.
BMC Cancer ; 20(1): 838, 2020 Sep 03.
Artículo en Inglés | MEDLINE | ID: mdl-32883233

RESUMEN

BACKGROUND: Breast cancer (BC) is a malignant tumor that occurs in the epithelial tissue of the breast gland. Long non-coding RNA (lncRNA) small nucleolar RNA host gene 3 (SNHG3) has been found to promote BC cell proliferation and invasion by regulating the microRNA (miR)-101/zinc-finger enhancer binding axis in BC. Herein, the objective of the present study is to evaluate the effect of lncRNA SNHG3 on BC cell proliferation and metastasis with the Notch signaling pathway. METHODS: Differentially expressed lncRNA in BC tissues and normal breast tissues was analyzed. SNHG3 si-RNA-1 and SNHG3 si-RNA-2 were constructed to detect the mechanism of SNHG3 interference in BC cell proliferation, viability, migration and invasion. Then, dual-luciferase reporter gene assay was utilized to verify the binding relation between SNHG3 and miR-154-3p as well as miR-154-3p and Notch2. Moreover, xenograft transplantation was applied to confirm the in vitro experiments. RESULTS: Highly expressed SNHG3 was observed in BC tissues. The growth of BC cells in vivo and in vitro was evidently repressed after silencing SNHG3. BC cell invasion and migration were inhibited by silencing SNHG3 in vitro. SNHG3 could act as a competing endogenous RNA of miR-154-3p and upregulate the Notch signaling pathway to promote BC cell development. Activation of the Notch signaling pathway can partly reverse the inhibition of cell activity induced by silencing SNHG3. CONCLUSION: Our study demonstrated that interfered lncRNA SNHG3 promoted BC cell proliferation and metastasis by activating the Notch signaling pathway. This investigation may offer new insight for BC treatment.


Asunto(s)
Neoplasias de la Mama/metabolismo , Proliferación Celular/genética , MicroARNs/metabolismo , ARN Largo no Codificante/metabolismo , Receptor Notch2/metabolismo , Transducción de Señal/genética , Animales , Neoplasias de la Mama/patología , Movimiento Celular/genética , Femenino , Regulación Neoplásica de la Expresión Génica , Silenciador del Gen , Humanos , Células MCF-7 , Ratones , Ratones Endogámicos BALB C , Ratones Desnudos , Invasividad Neoplásica/genética , Metástasis de la Neoplasia/genética , ARN Largo no Codificante/genética , ARN Interferente Pequeño/genética , Carga Tumoral/genética , Ensayos Antitumor por Modelo de Xenoinjerto
19.
Exp Mol Pathol ; 112: 104343, 2020 02.
Artículo en Inglés | MEDLINE | ID: mdl-31751562

RESUMEN

Hypoxic-ischemic brain damage (HIBD) is a major cause of morbidity and mortality in the preterm and term infant. However, the precise mechanism of HIBD remains largely elusive. As a newly discovered long non-coding RNA, small nucleolar RNA host gene 3 (Snhg3) has shown its important roles in cell apoptosis, proliferation, and disease development. In this study, we determined the role of Snhg3 in the pathogenesis of HIBD. Snhg3 expression was significantly down-regulated in the neonatal brain and primary hippocampal cells response to hypoxic/ischemic stress. Snhg3 overexpression protected against hypoxic/ischemic-induced brain injury in vivo and hippocampal cell injury in vitro. Snhg3 acted as the sponge of miR-196 in the hippocampal cells by regulating the expression of miR-196 target genes, XIAP and CAAP1. Moreover, Snhg3 overexpression decreased brain infarct size and ameliorated hypoxic-ischemic neonatal brain damage. This study suggests that Snhg3 is a potential target for the treatment of HIBD.


Asunto(s)
Lesiones Encefálicas/genética , Hipoxia-Isquemia Encefálica/genética , MicroARNs/genética , ARN Largo no Codificante/genética , Animales , Animales Recién Nacidos , Apoptosis/genética , Proteínas Reguladoras de la Apoptosis/genética , Encéfalo/metabolismo , Encéfalo/patología , Lesiones Encefálicas/patología , Proliferación Celular/genética , Modelos Animales de Enfermedad , Regulación de la Expresión Génica/genética , Hipocampo/metabolismo , Hipocampo/patología , Humanos , Hipoxia-Isquemia Encefálica/patología , Proteínas Inhibidoras de la Apoptosis/genética , Masculino , Ratones , Neuronas/metabolismo , Neuronas/patología
20.
J Biol Regul Homeost Agents ; 34(6): 2017-2027, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-33225676

RESUMEN

Small nucleolar RNA host genes (SNHGs) as a subset of long non-coding RNAs (lncRNAs) act critical roles in tumor progression. The present study aimed to elucidate the role and mechanisms of SNHG3 in non-small cell lung cancer (NSCLC). The correlation of SNHG3/miR-340-5p/HOXA10 with the clinicopathological features and outcomes in NSCLC was analyzed by TCGA cohort. In vitro and in vivo functional experiments were conducted to assess the role of SNHG3 in NSCLC cells. Bioinformatic analysis and luciferase gene reporter were used to estimate the interaction between miR-340-5p and SNHG3/HOXA10 3'UTR. The effects of SNHG3 and (or) miR-340-5p on HOXA10 expression were detected by qRT-PCR and Western blot analysis. As a consequence, the elevated expression of SNHG3 and HOXA10 or lowered expression of miR-340-5p was related to the lymph node infiltration, distant metastases and unfavorable prognosis in NSCLC. Ectopic expression of SNHG3 boosted the proliferation and invasion of NSCLC cells in vitro and in vivo, whereas downregulation of SNHG3 reversed these effects. Moreover, SNHG3 could bind with miR-340-5p and reduce its expression levels, and miR-340-5p attenuated SNHG3-induced tumor proliferation and HOXA10 expression in NSCLC cells. Our findings unveiled that SNHG3 might be an oncogenic factor in NSCLC by downregulating miR-340-5p.


Asunto(s)
Carcinoma de Pulmón de Células no Pequeñas , Neoplasias Pulmonares , ARN Largo no Codificante , Carcinoma de Pulmón de Células no Pequeñas/genética , Línea Celular Tumoral , Proliferación Celular/genética , Regulación Neoplásica de la Expresión Génica , Proteínas Homeobox A10 , Humanos , Neoplasias Pulmonares/genética , MicroARNs , ARN Largo no Codificante/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA