RESUMO
Osteosarcoma (OS) is a malignant bone tumor that commonly affects young individuals. Circular RNAs (circRNAs) are associated with OS progression. In this study, we aimed to determine the role of hsa_circ_0001480 (circ_0001480) in OS development. OS cell invasion, viability, and colony numbers were assessed via transwell, cell counting kit-8, and colony formation assays, respectively. Tumor growth in vivo was also assessed using an OS mouse model. Additionally, targeted associations among the integrin-binding sialoprotein (IBSP), microRNA (miR)-363-3p, and circ_0001480 were evaluated via RNA immunoprecipitation and dual luciferase reporter assays, whereas their expression levels in OS cells and tissues were determined via quantitative reverse transcription-polymerase chain reaction and western blotting. Loss of circ_0001480 or IBSP significantly inhibited the proliferation and invasion of OS cells, but this effect was reversed by miR-363-3p downregulation. Moreover, circ_0001480 knockdown inhibited neoplasm growth in vivo. circ_0001480 directly bound to miR-363-3p, which further modulated IBSP. Both circ_0001480 and IBSP levels were high, whereas miR-363-3p levels were low in OS cells. Furthermore, low miR-363-3p levels attenuated the suppressive effects of circ_0001480 silencing on the proliferation and invasion of OS cells; however, loss of IBSP partially reversed these effects. Overall, our findings revealed circ_0001480 an oncogenic circRNA stimulating OS progression by modulating the miR-363-3p/IBSP pathway, suggesting its potential for OS treatment.
Assuntos
Sialoproteína de Ligação à Integrina , MicroRNAs , Osteossarcoma , RNA Circular , Osteossarcoma/patologia , Osteossarcoma/genética , Osteossarcoma/metabolismo , MicroRNAs/genética , MicroRNAs/metabolismo , Humanos , RNA Circular/genética , RNA Circular/metabolismo , Animais , Camundongos , Sialoproteína de Ligação à Integrina/metabolismo , Sialoproteína de Ligação à Integrina/genética , Proliferação de Células , Neoplasias Ósseas/patologia , Neoplasias Ósseas/genética , Neoplasias Ósseas/metabolismo , Progressão da Doença , Linhagem Celular Tumoral , Camundongos Nus , Camundongos Endogâmicos BALB CRESUMO
BACKGROUND: This study aimed to investigate the expression of serum miR-363-3p in patients with polycystic ovary syndrome (PCOS) and its predictive value for pregnancy after ovulation induction therapy. METHODS: The expression of serum miR-363-3p was detected by Reverse transcription quantitative polymerase chain reaction (RT-qPCR). PCOS patients were treated with ovulation induction therapy, and after the successful pregnancy was confirmed, they were followed up for 1 year in outpatient department to record the pregnancy outcomes of the patients. The Pearson correlation coefficient was used to evaluate the correlation between the expression level of miR-363-3p and biochemical indicators of PCOS patients. Logistic regression analysis was used to analyze the risk factors of pregnancy failure after ovulation induction therapy. RESULTS: The serum level of miR-363-3p in PCOS group was significantly lower than that in control group. Compared with the control group, both pregnant and non-pregnant groups had lower miR-363-3p levels, while the non-pregnant group had a greater reduction in miR-363-3p levels than the pregnant group. Low levels of miR-363-3p showed high accuracy in distinguishing pregnant and non-pregnant patients. Logistic regression analysis showed that high levels of luteinizing hormone, testosterone (T), prolactin (PRL) and low level of miR-363-3p were independent risk factors for pregnancy failure after ovulation induction in PCOS patients. Additionally, compared with pregnancy outcomes of healthy women, the incidence of premature delivery, macrosomia, and gestational diabetes in PCOS patients increased. CONCLUSIONS: The expression of miR-363-3p in PCOS patients was reduced and correlated with abnormal hormone levels, suggesting that miR-363-3p may be involved in the occurrence and development of PCOS.
Assuntos
MicroRNAs , Síndrome do Ovário Policístico , Gravidez , Humanos , Feminino , Síndrome do Ovário Policístico/complicações , Síndrome do Ovário Policístico/genética , Resultado da Gravidez , Indução da Ovulação , Hormônio LuteinizanteRESUMO
BACKGROUND: Prostate cancer (PCa) is a frequent malignant tumor worldwide with high morbidity along with mortality. MicroRNAs (miRNAs) have been identified as key posttranscriptional modulators in diverse cancers. Herein, we purposed to explore the impacts of miR-363-3p on PCa growth, migration, infiltration along with apoptosis. METHODS: The expressions of miR-363-3p along with Dickkopf 3 (DKK3) were assessed in clinical PCa specimens. We adopted the PCa cell line PC3 and transfected it using miR-363-3p repressors or mimic. The relationship of miR-363-3p with DKK3 was verified by a luciferase enzyme reporter assay. Cell viability along with apoptosis were determined by MTT assay coupled with flow cytometry analysis. Cell migration along infiltration were detected via wound healing, as well as Transwell assays. The contents of DKK3, E-cadherin, vimentin along with N-cadherin were analyzed via Western blotting accompanied with qRT-PCR. RESULTS: MiR-363-3p was found to be inversely associated with the content of DKK3 in clinical PCa specimens. Further investigations revealed that DKK3 was miR-363-3p's direct target. Besides, overexpression of miR-363-3p decreased the contents of DKK3, promoted cell viability, migration coupled with infiltration, and reduced cell apoptosis, while silencing of miR-363-3p resulted in opposite influence. Upregulation of miR-363-3p diminished E-cadherin contents but increased vimentin along with N-cadherin protein contents in PC3 cells; in contrast, miR-363-3p downregulation produced the opposite result. CONCLUSION: Our study indicates that miR-363-3p promotes PCa growth, migration coupled with invasion while dampening apoptosis by targeting DKK3.
Assuntos
MicroRNAs , Neoplasias da Próstata , Apoptose/genética , Caderinas/metabolismo , Linhagem Celular Tumoral , Proliferação de Células/genética , Regulação Neoplásica da Expressão Gênica , Humanos , Masculino , MicroRNAs/genética , MicroRNAs/metabolismo , Neoplasias da Próstata/patologia , Vimentina/metabolismoRESUMO
In our previous study, we reported that CELF2 has a tumour-suppressive function in glioma. Here, we performed additional experiments to elucidate better its role in cancer. The expression profile of CELF2 was analysed by the GEPIA database, and Kaplan-Meier curves were used to evaluate the overall survival rates. Four different online databases were used to predict miRNAs targeting CELF2, and the luciferase assay was performed to identify the binding site. The biological effects of miR-363-3p and CELF2 were also investigated in vitro using MTT, Transwell, and flow cytometry assays. Western blotting, qPCR, and TOP/FOP flash dual-luciferase assays were performed to investigate the impact of miR-363-3p and CELF2 on epithelial-to-mesenchymal transition (EMT) and the Wnt/ß-catenin pathway. The effect of miR-363-3p was also tested in vivo using a xenograft mouse model. We observed an abnormal expression pattern of CELF2 in glioma cells, and higher CELF2 expression correlated with better prognosis. We identified miR-363-3p as an upstream regulator of CELF2 and confirmed its direct binding to the 3'-untranslated region of CELF2. Cell function experiments showed that miR-363-3p affected multiple aspects of glioma cells. Suppressing miR-363-3p expression inhibited glioma cell proliferation and invasion, as well as promoted cell death via attenuating EMT and blocking the Wnt/ß-catenin pathway. These effects could be abolished by the downregulation of CELF2. Treatment with ASO-miR-363-3p decreased tumour size and weight in nude mice. In conclusion, miR-363-3p induced the EMT, which resulted in increased migration and invasion and reduced apoptosis in glioma cell lines, via the Wnt/ß-catenin pathway by targeting CELF2.
Assuntos
Proteínas CELF/genética , Transição Epitelial-Mesenquimal/genética , Glioma/genética , Glioma/metabolismo , MicroRNAs/genética , Proteínas do Tecido Nervoso/genética , Via de Sinalização Wnt , Idoso , Animais , Linhagem Celular Tumoral , Biologia Computacional , Bases de Dados Genéticas , Modelos Animais de Doenças , Feminino , Perfilação da Expressão Gênica , Glioma/patologia , Xenoenxertos , Humanos , Masculino , Camundongos , Pessoa de Meia-Idade , Gradação de Tumores , Estadiamento de NeoplasiasRESUMO
Long noncoding RNAs (lncRNAs) have been proven to exert important functions in the various biological processes of human cancers. It has been reported that lncRNA HNF1 homeobox A antisense RNA 1 (HNF1A-AS1) was abnormally expressed and played a role in the initiation and development of various human cancers. In this study, we confirmed that the expression level of HNF1A-AS1 was increased in glioma tissues and cells. Knockdown of HNF1A-AS1 inhibited cell proliferation and promoted cell apoptosis in glioma. Then, we disclosed the downregulation of miR-363-3p in glioma tissues and cell lines. The interaction between HNF1A-AS1 and miR-363-3p was identified in glioma cells. Furthermore, an inverse correlation between HNF1A-AS1 and miR-363-3p was observed in glioma tissues. Afterwards, we recognized that MAP2K4 was a direct target of miR-363-3p. The expression of MAP2K4 was negatively correlated with miR-363-3p while positively related to HNF1A-AS1 in glioma tissues. We also found the regulatory effect of HNF1A-AS1 on the MAP2K4-dependent JNK signaling pathway. All findings indicated that HNF1A-AS1 induces the upregulation of MAP2K4 to activate the JNK signaling pathway to promote glioma cell growth by acting as a miR-363-3p sponge.
Assuntos
Glioma/genética , MAP Quinase Quinase 4/genética , MicroRNAs/genética , RNA Longo não Codificante/genética , Animais , Apoptose/genética , Proliferação de Células/genética , Intervalo Livre de Doença , Feminino , Regulação Neoplásica da Expressão Gênica/genética , Glioma/patologia , Xenoenxertos , Humanos , Estimativa de Kaplan-Meier , Masculino , Camundongos , Transdução de Sinais/genéticaRESUMO
Lung cancer is one of the most common malignant tumors in the world. Non-small cell lung cancer (NSCLC) accounts for about 80% of all lung cancers. About 75% of patients are in the middle and advanced stages at the time of discovery, and the 5-year survival rate is very low. The aim of this study was to investigate the role of long non-coding RNA (lncRNA) NORAD in the pathogenesis of NSCLC. We found that lncRNA NORAD was highly expressed in human NSCLC tissues and cell lines. The CCK-8 assay results showed that lncRNA NORAD had no effect on cell proliferation. The Transwell assay and Western blotting results showed that overexpression of lncRNA NORAD promoted the invasion and epithelial-mesenchymal transition (EMT) of NSCLC cells. Then bioinformatics analysis was used to screen for candidate miRNA bound with lncRNA NORAD and the target gene of miRNA in NSCLC. The luciferase reporter gene assay and RNA pull-down assay were used to verify the relationship. We found that miR-363-3p expression was down-regulated, whereas PEAK1 expression was upregulated in NSCLC cells. We performed gain and loss function test of lncRNA NORAD, miR-363-3p and PEAK1, the results showed that while miR-363-3p-mimic inhibited cell invasion and EMT by targeting PEAK1, lncRNA NORAD acted as a sponge of miR-363-3p and promoted cell invasion and EMT by increasing the expression of PEAK1. In addition, p-ERK expression was detected by Western blotting to observe the effects of lncRNA NORAD, miR-363-3p and PEAK1 on activation of the ERK signaling pathway. Taken together, lncRNA NORAD upregulated the expression of PEAK1 through sponging miR-363-3p, and then activated the ERK signaling pathway, thereby promoting the development of NSCLC.
Assuntos
Carcinoma Pulmonar de Células não Pequenas/metabolismo , Neoplasias Pulmonares/metabolismo , MicroRNAs/metabolismo , Proteínas Tirosina Quinases/metabolismo , Carcinoma Pulmonar de Células não Pequenas/genética , Carcinoma Pulmonar de Células não Pequenas/patologia , Proliferação de Células/fisiologia , Transição Epitelial-Mesenquimal , Humanos , Neoplasias Pulmonares/genética , Neoplasias Pulmonares/patologia , Sistema de Sinalização das MAP Quinases , MicroRNAs/genética , Pessoa de Meia-Idade , Invasividade Neoplásica , Regulação para CimaRESUMO
Retinoblastoma is the most common malignancy in children's eyes with high incidence. Long non-coding RNAs (lncRNAs) play important roles in the progression of retinoblastoma. LncRNA FEZF1 antisense RNA 1 (FEZF1-AS1) has been found to stimulate retinoblastoma. However, the mechanism of FEZF1-AS1 underlying progression of retinoblastoma is still unclear. In current study, FEZF1-AS1 was up-regulated in retinoblastoma tissues and cells. FEZF1-AS1 overexpression enhanced retinoblastoma cell viability, promoted cell cycle, and inhibited apoptosis. Conversely, FEZF1-AS1 knockdown reduced cell viability, cycle, and elevated apoptosis. The interaction between FEZF1-AS1 and microRNA-363-3p (miR-363-3p) was confirmed. FEZF1-AS1 down-regulated miR-363-3p and up-regulated PAX6. PAX6 was a target gene of miR-363-3p. EZF1-AS1 promoted retinoblastoma cell viability and suppressed apoptosis via PAX6. Further, we demonstrated that FEZF1-AS1 contribute to tumor formation in vivo. In conclusion, FEZF1-AS1 elevated growth and inhibited apoptosis by regulating miR-363-3p/PAX6 in retinoblastoma, which provide a new target for retinoblastoma treatment.
Assuntos
Apoptose/genética , Proliferação de Células/genética , MicroRNAs/genética , Fator de Transcrição PAX6/genética , RNA Antissenso/genética , Proteínas Repressoras/genética , Retinoblastoma/genética , Estudos de Casos e Controles , Ciclo Celular/genética , Técnicas de Silenciamento de Genes , Humanos , Reação em Cadeia da Polimerase , Retinoblastoma/patologiaRESUMO
Clopidogrel-induced gastric injury is an important clinical problem. However, the exact mechanism is still unclarified. Increasing evidence indicates that miRNAs may be involved in the pathogenesis of gastric mucosal injury. In this study, the aim was to investigate the role of miR-363-3p in the gastric mucosal injury caused by clopidogrel. MiRNA microarray analysis was performed using paired gastric mucosal in order to find differential expression of miRNAs. The levels of miR-363-3p were examined in gastric mucosal injury caused by clopidogrel. The GES-1 cells were used as a model system, miR-363-3p mimic/inhibitor was transfected into GES-1 cells, then GES-1 cells were treated with clopidogrel. The levels of miR-363-3p and DUSP10 were examined in GES-1 cells using quantitative real-time PCR (qRT-PCR). CCK-8 assay and flow cytometry analysis were used to detect cell proliferation and apoptosis, respectively. Western blot assay was used to measure the protein levels of DUSP10. The interaction between miR-363-3p and DUSP10 was determined by luciferase reporter assay. MiR-363-3p was selected as a differentially expressed miRNA. The expression of miR-363-3p in gastric mucosal injury caused by clopidogrel was higher than that in normal samples. Also, depletion of miR-363-3p increased the proliferation of GES-1 cells and reduced the apoptosis. Luciferase-reporting assay results confirmed that DUSP10 was one of the target genes of miR-363-3p. DUSP10 inhibited apoptosis in GES-1 cells treated by clopidogrel. Moreover, DUSP10 knockdown abrogated the inhibitory effects on apoptosis in GES-1 cells mediated by miR-363-3p inhibitor. Knockdown of miR-363-3p increased the proliferation and reduced the apoptosis by targeting DUSP10 in GES-1 cells treated by clopidogrel, indicating that miR-363-3p may be a potential therapeutic target for gastric mucosal injury caused by clopidogrel.
Assuntos
MicroRNAs , Apoptose , Proliferação de Células , Clopidogrel/toxicidade , Mucosa Gástrica , Regulação Neoplásica da Expressão Gênica , MicroRNAs/genética , Transdução de SinaisRESUMO
miR-363-3p is downregulated in lung adenocarcinoma and can inhibit tumor growth. Here, we aimed to investigate the effect of miR-363-3p on non-small-cell lung cancer (NSCLC) metastasis. In our study, miR-363-3p overexpression inhibited cell migration and invasion via epithelial-mesenchymal transition inhibition, while miR-363-3p knockdown exhibited the opposite effects. Further studies demonstrated that miR-363-3p bound to 3'-untranslated regions of NEDD9 and SOX4, and negatively regulated their levels. Interestingly, NEDD9 or SOX4 knockdown rescued the metastasis-promoting effects of antagomiR-363-3p. The inhibitory effects of agomiR-363-3p were also blocked by NEDD9 or SOX4 overexpression. Moreover, lentivirus particles carrying pre-miR-363 (LV-pre-miR-363) significantly decreased, while LV-miR-363-3p inhibitor increased metastatic nodule numbers and the levels of NEDD9 and SOX4 in lungs. In conclusion, tumor suppressor miR-363-3p may be a potential target in NSCLC therapy.
Assuntos
Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Carcinoma Pulmonar de Células não Pequenas/patologia , Neoplasias Pulmonares/patologia , MicroRNAs/genética , Fatores de Transcrição SOXC/metabolismo , Proteínas Adaptadoras de Transdução de Sinal/genética , Animais , Carcinoma Pulmonar de Células não Pequenas/genética , Carcinoma Pulmonar de Células não Pequenas/metabolismo , Linhagem Celular Tumoral , Movimento Celular/genética , Transição Epitelial-Mesenquimal/genética , Regulação Neoplásica da Expressão Gênica/genética , Xenoenxertos , Humanos , Neoplasias Pulmonares/genética , Neoplasias Pulmonares/metabolismo , Camundongos , Camundongos Endogâmicos NOD , Camundongos SCID , MicroRNAs/metabolismo , Invasividade Neoplásica/genética , Fatores de Transcrição SOXC/genéticaRESUMO
Propofol, a generally used anaesthetic in patients care, has been proven to induce neurotoxicity. Studies have shown that miR-363-3p was closely related to neurological dysfunction, and the up-regulated miR-363-3p was recognized to be participate in propofol-induced neurotoxicity. However, the mechanisms and functions of miR-363-3p in propofol-induced neurotoxicity remain rarely reported. The aim of our research was to clarify the potential effects of miR-363-3p in neurotoxicity induced by propofol. SH-SY5Y cells were treated with propofol, miR-363-3p inhibitor or sh-CREB. quantitative real-time polymerase chain reaction and western blotting were applied to detect the expression of miR-363-3p, CREB, Bax, Bcl-2, cleaved caspase-9 and cleaved caspase-3 at the mRNA and/or protein level, respectively. The levels of lactate dehydrogenase (LDH), superoxide dismutase (SOD) and malondialdehyde (MDA) in cell supernatant were detected using different kits. Flow cytometry and MTT assay were applied for assessing the functions of miR-363-3p and CREB on cell ability in cellular activity and apoptotic rate. In addition, Bioinformatic analysis and luciferase assay verified the relationship between 3'-UTR of CREB and miR-363-3p. Our data indicated that the cell viability decreased with the increasing propofol concentration. Bioinformatic analysis and luciferase assay suggested that 3'-UTR of transcript of CREB might be a binding site of miR-363-3p, and miR-363-3p could negatively regulate the expression of CREB. The changes in reactive oxygen species, LDH, SOD and MDA suggested that propofol mediates oxidative stress and apoptosis via modulating miR-363-3p/CREB axis. Propofol induces oxidative stress and apoptosis via affecting miR-363-3p/CREB axis in SH-SY5Y cells, suggesting miR-363-3p down-regulation may act as a novel strategy to ameliorate the propofol-induced neurotoxicity. Significance of the study: The present study demonstrated that propofol induces oxidative stress and apoptosis via affecting miR-363-3p/CREB axis in SH-SY5Y cells, suggesting miR-363-3p down-regulation may act as a novel strategy to ameliorate the propofol-induced neurotoxicity.
Assuntos
Apoptose/efeitos dos fármacos , Proteína de Ligação ao Elemento de Resposta ao AMP Cíclico/metabolismo , MicroRNAs/metabolismo , Estresse Oxidativo/efeitos dos fármacos , Propofol/farmacologia , Transdução de Sinais/efeitos dos fármacos , Apoptose/genética , Linhagem Celular Tumoral , Humanos , MicroRNAs/genética , Estresse Oxidativo/genética , Transdução de Sinais/genéticaRESUMO
LncRNA MALAT1 is reported to play a potential role in human cancers. Hence, we investigated the effects of MALAT1 on colorectal cancer in vitro and in vivo, and further validated whether MALAT1 affected colorectal cancer development and EZH2 expression via regulating miR-363-3p. The fresh colorectal cancer tissues, adjacent non-tumor tissues, FHC, LOVO, SW620, CL40 and HCT116 cells were analyzed in this study. MALAT1, miR-363-3p and EZH2 expression levels were assessed using qRT-PCR and Western blot. Cell proliferation, migration and invasion were also measured. Binding effects between MALAT1 and miR-363-3p, or miR-363-3p and EZH2 3'UTR were detected by dual luciferase assay. We observed that MALAT1 was highly expressed in colorectal cancer tissues and cells, and MALAT1 knockdown inhibited cell proliferation as well as expression levels of EZH2 by upregulated miR-363-3p in cell models and in vivo. Moreover, miR-363-3p functions as a downstream target of MALAT1, meanwhile EZH2 was a target of miR-363-3p, suggesting MALAT1 might regulate miR-363-3p and/or EZH2 expression. Collectively, we concluded that MALAT1 functioned as a ceRNA to promote colorectal cancer development and EZH2 expression through sponging miR-363-3p in vitro and in vivo.
Assuntos
Neoplasias Colorretais/patologia , Proteína Potenciadora do Homólogo 2 de Zeste/genética , MicroRNAs/genética , RNA Longo não Codificante/genética , Linhagem Celular Tumoral , Neoplasias Colorretais/genética , Regulação Neoplásica da Expressão Gênica , HumanosRESUMO
BACKGROUND: Deregulation of microtubules and centrosome integrity is response for the initiation and progression of human cancers. Sperm-associated antigen 5 (SPAG5) is essential for the spindle apparatus organization and chromosome segregation, but its role in hepatocellular carcinoma (HCC) remains undefined. METHODS: The expression of SPAG5 in HCC were examined in a large cohort of patients by RT-PCR, western blot and IHC. The clinical significance of SPAG5 was next determined by statistical analyses. The biological function of SPAG5 in HCC and the underlying mechanisms were investigated, using in vitro and in vivo models. RESULTS: Here, we demonstrated that SPAG5 exhibited pro-HCC activities via the activation of PI3K/AKT signaling pathway. SPAG5 expression was increased in HCC and correlated with poor outcomes in two independent cohorts containing 670 patients. High SPAG5 expression was associated with poor tumor differentiation, larger tumor size, advanced TNM stage, tumor vascular invasion and lymph node metastasis. In vitro and in vivo data showed that SPAG5 overexpression promoted tumor growth and metastasis, whereas SPAG5 knockdown led to the opposite phenotypes. SPAG5 interacted with centrosomal protein CEP55 to trigger the phosphorylation of AKT at Ser473. Inhibition of PI3K/AKT signaling markedly attenuated SPAG5-mediated cell growth. Furthermore, SPAG5 expression was suppressed by miR-363-3p which inhibited the activity of SPAG5 mRNA 3'UTR. Ectopic expression of SPAG5 partly abolished the miR-363-3p-caused cell cycle arrest and suppression of cell proliferation and migration. CONCLUSIONS: Collectively, these findings indicate that SPAG5 serves a promising prognostic factor in HCC and functions as an oncogene via CEP55-mediated PI3K/AKT pathway. The newly identified miR-363-3p/SPAG5/CEP55 axis may represent a potential therapeutic target for the clinical intervention of HCC.
Assuntos
Carcinoma Hepatocelular/metabolismo , Proteínas de Ciclo Celular/genética , Proteínas de Ciclo Celular/metabolismo , Neoplasias Hepáticas/metabolismo , Proteínas Nucleares/metabolismo , Animais , Carcinoma Hepatocelular/genética , Carcinoma Hepatocelular/patologia , Linhagem Celular Tumoral , Movimento Celular , Feminino , Regulação Neoplásica da Expressão Gênica , Humanos , Neoplasias Hepáticas/genética , Neoplasias Hepáticas/patologia , Masculino , Camundongos , MicroRNAs/genética , Estadiamento de Neoplasias , Transplante de Neoplasias , Fosfatidilinositol 3-Quinases/metabolismo , Fosforilação , Prognóstico , Proteínas Proto-Oncogênicas c-akt/metabolismo , Transdução de Sinais , Análise de SobrevidaRESUMO
Gallbladder carcinoma (GBC) is an aggressive neoplasm, and the treatment options for advanced GBC are limited. Recently, long non-coding RNAs (lncRNAs) have emerged as new gene regulators and prognostic markers in several cancers. In this study, we found that metastasis-associated lung adenocarcinoma transcript 1 (MALAT1) expression was up-regulated in GBC tissues (P < 0.05). Luciferase reporter assays and RNA pull down assays showed that MALAT1 is a target of miR-363-3p. Real-time quantitative PCR and Western blot analysis indicated that MALAT1 regulated Myeloid cell leukaemia-1 (MCL-1) expression as a competing endogenous RNA (ceRNA) for miR-363-3p in GBC cells. Furthermore, MALAT1 silencing decreased GBC cell proliferation and the S phase cell population and induced apoptosis in vitro. In vivo, tumour volumes were significantly decreased in the MALAT1 silencing group compared with those in the control group. These data demonstrated that the MALAT1/miR-363-3p/MCL-1 regulatory pathway controls the progression of GBC. Inhibition of MALAT1 expression may be to a novel therapeutic strategy for gallbladder cancer.
Assuntos
Neoplasias da Vesícula Biliar/genética , Regulação Neoplásica da Expressão Gênica , MicroRNAs/metabolismo , Proteína de Sequência 1 de Leucemia de Células Mieloides/genética , RNA Longo não Codificante/metabolismo , Animais , Apoptose/genética , Sequência de Bases , Ciclo Celular/genética , Linhagem Celular Tumoral , Proliferação de Células/genética , Regulação para Baixo/genética , Neoplasias da Vesícula Biliar/patologia , Técnicas de Silenciamento de Genes , Masculino , Camundongos Nus , MicroRNAs/genética , Proteína de Sequência 1 de Leucemia de Células Mieloides/metabolismo , RNA Longo não Codificante/genética , Carga Tumoral/genéticaRESUMO
Epithelial-to-mesenchymal transition (EMT) plays an essential role in cancer invasion and metastasis and is associated with tumor recurrence in colorectal cancer (CRC). However, the mechanism that contributes to EMT have not been fully understood in CRC. In the present study, we showed that miR-363-3p was frequently down-regulated in CRC tissue specimens with lymph node metastasis. Moreover, we demonstrated that down-regulation of miR-363-3p promoted CRC cell migration and invasion, and induced EMT in vitro and in vivo, revealing that miR-363-3p playes a potential tumor-suppressive role in CRC. Analysis of the underlying mechanisms revealed that miR-363-3p could regulate Sox4 expression by directly targeting its 3'untranslated region. Down-regulation of miR-363-3p increased Sox4 expression and induced EMT, while overexpression of miR-363-3p decreased Sox4 expression. Taken together, our findings indicate that miR-363-3p is involved in CRC metastasis and functions as a tumor suppressor via negatively regulating Sox4. Therefore, the up-regulation of miR-363-3p in human CRC may have therapeutic benefits.
Assuntos
Neoplasias Colorretais/genética , Neoplasias Colorretais/secundário , Transição Epitelial-Mesenquimal/genética , MicroRNAs/genética , Fatores de Transcrição SOXC/genética , Animais , Linhagem Celular Tumoral , Movimento Celular/genética , Neoplasias Colorretais/patologia , Regulação para Baixo , Genes Supressores de Tumor , Humanos , Metástase Linfática , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Nus , Invasividade Neoplásica/genética , Células Tumorais CultivadasRESUMO
OBJECTIVE: Adenocarcinoma (ACA) of the uterine cervix is increasing in incidence and currently accounts for approximately 20% of all cervical malignancies. MicroRNAs (miRNAs) have been investigated as potential biomarkers of cervical cancer; however, their role in ACA remains unknown. Here, we characterized miRNA expression profiles and investigated miRNAs as diagnostic and prognostic factors in ACA. METHODS: Evaluation of genome-wide miRNA expression profiles in ACA by microarray led to the identification of ten candidate miRNAs, whose expression patterns were validated by qRT-PCR in 45 ACA, 10 normal control, and 15 squamous cell carcinoma samples. The association between miRNA expression and prognosis was analyzed in patients with ACA. RESULTS: Microarray analysis identified 86 miRNAs that were dysregulated more than 2.0-fold (p<0.05) in ACA relative to normal tissues of the uterine cervix. Five most over- and underexpressed miRNAs were selected respectively and their expression patterns were confirmed in the validation set. MiR-135b, miR-192, and miR194 were overexpressed in ACA, and miR-363-3p, miR-195 and miR-199b were significantly associated with conventional prognostic factors. Overexpression of miR-363-3p by more than 2.5-fold relative to the normal control was a strong predictor of favorable prognosis (hazard ratio, 0.1; 95% confidence interval, 0.009-0.779) after adjusting for confounders. CONCLUSIONS: MiR-135b, miR-192, and miR-194 are altered in uterine cervical ACA, and miR-363-3p is an independent favorable prognostic factor in ACA. These miRNAs could be of value as biomarkers for the diagnosis and prognosis of ACA.
Assuntos
Adenocarcinoma/genética , MicroRNAs/biossíntese , Neoplasias do Colo do Útero/genética , Adenocarcinoma/metabolismo , Adulto , Feminino , Genótipo , Humanos , MicroRNAs/genética , Análise em Microsséries , Pessoa de Meia-Idade , Prognóstico , Neoplasias do Colo do Útero/metabolismoRESUMO
PURPOSE: Colorectal cancer (CRC) refers to high-mortality tumors arising in the colon or rectum with a high rate of recurrence. The involvement of long non-coding RNAs (lncRNAs) contributes to the treatment and prognosis evaluation of CRC, and brings a new direction for the radical cure of patients. To identify the pathological mechanism and regulation of lncRNA LINC01128 (LINC01128) on CRC cells, and analyze its potential prognostic value. METHODS: LINC01128 level in tissue and cell specimens from 122 CRC patients was evaluated by RT-qPCR. The clinical significance and prognostic value of LINC01128 in CRC were analyzed via Kaplan-Meier and Cox analysis. CCK8 and Transwell assays were used to study the function of LINC01128 in vitro. The relationship between LINC01128 and miR-363-3p was confirmed by luciferase reporter gene assay. RESULTS: The overexpression of LINC01128 is associated with TNM stage and lymph node metastasis in CRC patients. Silencing LINC01128 inhibited the proliferation and metastasis of CRC cells. In addition, LINC01128 directly targeted and negatively regulated the miR-363-3p expression, while miR-363-3p inhibitor restored the inhibitory function of LINC01128. CONCLUSION: As an independent prognostic factor of CRC, upregulation of LINC01128 predicts poor prognosis and accelerates tumor deterioration through miR-363-3p.
Assuntos
Proliferação de Células , Neoplasias Colorretais , MicroRNAs , RNA Longo não Codificante , Regulação para Cima , Humanos , Neoplasias Colorretais/genética , Neoplasias Colorretais/patologia , RNA Longo não Codificante/genética , MicroRNAs/genética , Prognóstico , Masculino , Proliferação de Células/genética , Feminino , Pessoa de Meia-Idade , Regulação Neoplásica da Expressão Gênica , Linhagem Celular Tumoral , Biomarcadores Tumorais/genética , Biomarcadores Tumorais/metabolismo , Metástase Linfática/genética , IdosoRESUMO
Nonalcoholic fatty liver disease (NAFLD) constitutes one of major worldwide health problem which typically progressively results in nonalcoholic steatohepatitis (NASH) and eventually cirrhosis and liver cancer. Liver-specific deletion of INSIG1 promotes SREBP1 nuclear translocation to activate downstream lipogenic genes expression, leading to lipid accumulation. However, the underlying pathogenesis of NAFLD, and particularly involved in miRNA participation are still to be thoroughly explored. Here, we found that miR-363-3p was significantly overexpressed in high-fat, high-cholesterol (HFHC) diet mice liver tissue and fatty acid-induced steatosis cells. miR-363-3p directly targets INSIG1 to inhibit its expression, thereby facilitating the cleavage of SREBP and nuclear translocation to activate subsequent transcription of lipogenic genes in vitro and in vivo. In addition, we identified apigenin, a natural flavonoid compound, inhibited miR-363-3p expression to up-regulate INSIG1 and suppress nuclear translocation of SREBP1, thereby down-regulated lipogenic genes expression in steatosis cells and HFHC diet mice liver tissues. Taken together, our results demonstrated that miR-363-3p as a key regulator of hepatic lipid homeostasis targeted INSIG1, and apigenin alleviated NAFLD through the miR-363-3p/INSIG1/SREBP1 pathway. This indicates that reduction of miR-363-3p levels as a possible treatment of hepatic steatosis and provides a potential new therapeutic strategy for targeting miRNA to ameliorate NAFLD.
RESUMO
BACKGROUND: Genistein (GEN) is one of the most well-known phytoestrogens identified in various legumes. Although increasing evidence shows GEN has a potential use in phytotherapy to regulate lipid metabolism, its therapeutic mechanisms have not yet been completely elucidated, especially epigenetic alterations of miRNAs to alleviate lipid accumulation in the liver remains unknown. PURPOSE: To clarify how GEN modulates the miRNA profile in HepG2 cells and investigate molecular mechanisms of the modulated miRNA on regulating hepatic lipid metabolism. METHODS: The miRNA microarray was performed to compare the miRNAs expression patterns, followed by determining principal miRNA and its target gene associated with hepatic lipid metabolism modulated by GEN. miR-363-3p mimics (mi) and phosphatase and tensin homolog (PTEN)-siRNA were transfected into HepG2 cells and GEN was further treated with the cells for 24 h RESULTS: GEN induced downregulation of miR-363-3p and upregulation of PTEN, which was a target mRNA of miR-363-3p. The miR-363-3p mi led to an upregulation of sterol-regulatory element-binding protein-1c (SREBP-1c) and its downstream lipid synthesis-related factors in HepG2 cells. In addition, the inhibition of PTEN led to an increase of lipogenesis, which was associated with the AKT/mTOR signal regulation. However, GEN treatment could abrogate the lipogenic effects of miR-363-3p mi or PTEN siRNA. The modulation was associated with estrogen receptor ß (ERß). CONCLUSION: We discerned a new mechanism that GEN regulated hepatic lipid metabolism by inhibiting miR-363-3p, which could be mediated via ERß and by targeting PTEN in HepG2 cells. Additionally, GEN reduced hepatic lipid accumulation by regulating PTEN-AKT/mTOR signal. It implicated a protective role of GEN by elucidating its epigenetic modification of the miRNA modulated by ERß on improving hepatic lipid metabolism and provided novel evidence of the mechanism on targeting miR-363-3p/PTEN in treating hepatic lipid disorders.
Assuntos
Metabolismo dos Lipídeos , MicroRNAs , Humanos , Receptor beta de Estrogênio/genética , Receptor beta de Estrogênio/metabolismo , Transdução de Sinais , Proteínas Proto-Oncogênicas c-akt/metabolismo , Genisteína/farmacologia , Células Hep G2 , MicroRNAs/genética , MicroRNAs/metabolismo , Serina-Treonina Quinases TOR/metabolismo , PTEN Fosfo-Hidrolase/genética , PTEN Fosfo-Hidrolase/metabolismo , RNA Interferente Pequeno/metabolismo , LipídeosRESUMO
Objective: This study aimed to elucidate the prognostic implications of deviant expressions of long non-coding RNA (lncRNA) p53 upregulated regulator of p53 levels (PURPL), microRNA-363-3p (miR-363-3p), and ADAM metallopeptidase domain 10 (ADAM10) in patients diagnosed with ovarian serous cystadenocarcinoma (OSC). Methods: To predict and refine the targeted miRNAs and downstream target genes for PURPL, we utilized open medical databases. Through the employment of real-time RT-PCR, we conducted tissue analysis to discern the expressions of PURPL, miR-363-3p, and ADAM10 in both OSC and control tissues. The pathological correlations in the clinic and the prognostic implications of deviant expressions of PURPL, miR-363-3p, and ADAM10 in OSC patients were analyzed independently. Results: Database inquiries revealed that PURPL might target miR-363-3p, and in turn, miR-363-3p could target ADAM10. Differential expression of PURPL, miR-363-3p, and ADAM10 was observed between OSC and paired tissues. The premature version of miR-363-3p, miR-363, correlated with overall survival (OS), while ADAM10 corresponded with progression-free survival (PFS) in ovarian cancer patients. Tissue detection displayed significantly elevated expressions of PURPL and ADAM10, and conspicuously diminished expressions of miR-363-3p in OSC tissues compared to the control tissues (P<0.05). A negative correlation was observed between the expressions of PURPL and miR-363-3p, and miR-363-3p and ADAM10, while a positive correlation was found between PURPL and ADAM10 in different ovarian tissues (P<0.05). In OSC tissues, upregulation of PURPL was associated with an advanced clinical stage, TP53 mutation, and lymph node metastasis (P<0.05), downregulation of miR-363-3p was associated with a more advanced clinical stage and lymph node metastasis (P<0.05), and overexpression of ADAM10 correlated with a more advanced FIGO stage. High expressions of PURPL and ADAM10, and low expression of miR-363-3p, were linked with poor PFS and OS in OSC patients, respectively (P<0.05). In addition, OSC patients with elevated PURPL and reduced miR-363-3p, patients with elevated PURPL and ADAM10, and patients with reduced miR-363-3p and elevated ADAM10 also demonstrated worse PFS and OS, respectively (P<0.05). Conclusions: The anomalous expressions of PURPL, miR-363-3p, and ADAM10 might contribute to the pathogenesis of OSC via up-down stream regulation, and these abnormal expressions could serve as potential prognostic indicators for OSC patients.
RESUMO
This study attempted to explore whether miR-363-3p play a role in the isoflurane (ISO)-mediated protective effect of cardiomyocyte injury induced by hypoxia/reoxygenation (H/R). A myocardial cell injury model was established, and the different preconditioning ISO concentrations were screened and determined. The miR-363-3p level was detected by RT-qPCR. The effects of miR-363-3p on proliferation and apoptosis of H9c2 cells were detected by CCK-8 assay and flow cytometry. Myocardial injury indexes were determined by enzyme-linked immunosorbent assay (ELISA). The interaction of miR-363-3p with the 3'-UTR of the KLF2 gene was confirmed by luciferase reporter gene assay. ISO pretreatment can reduce the up-regulation of miR-363-3p after H/R injury. ISO pretreatment reduces the inhibition of cell viability and the promotion of cell apoptosis induced by H/R stimuli, while the overexpression of miR-363-3p counteracts the protective effect of ISO pretreatment. Meanwhile, ISO pretreatment also reduced the level of markers of H/R-induced myocardial injury. Moreover, luciferase reporter analysis showed that KLF2 was the downstream target gene of miR-363-3p. ISO pretreatment may alleviate H/R-induced cardiomyocyte injury by regulating miR-363-3p.