Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 5 de 5
Filter
Add more filters










Publication year range
1.
Biomedicines ; 12(5)2024 Apr 23.
Article in English | MEDLINE | ID: mdl-38790894

ABSTRACT

Long noncoding RNAs (lncRNAs) are RNA molecules of 200 nucleotides or more in length that are not translated into proteins. Their expression is tissue-specific, with the vast majority involved in the regulation of cellular processes and functions. Many human diseases, including cancer, have been shown to be associated with deregulated lncRNAs, rendering them potential therapeutic targets and biomarkers for differential diagnosis. The expression of lncRNAs in the nervous system varies in different cell types, implicated in mechanisms of neurons and glia, with effects on the development and functioning of the brain. Reports have also shown a link between changes in lncRNA molecules and the etiopathogenesis of brain neoplasia, including glioblastoma multiforme (GBM). GBM is an aggressive variant of brain cancer with an unfavourable prognosis and a median survival of 14-16 months. It is considered a brain-specific disease with the highly invasive malignant cells spreading throughout the neural tissue, impeding the complete resection, and leading to post-surgery recurrences, which are the prime cause of mortality. The early diagnosis of GBM could improve the treatment and extend survival, with the lncRNA profiling of biological fluids promising the detection of neoplastic changes at their initial stages and more effective therapeutic interventions. This review presents a systematic overview of GBM-associated deregulation of lncRNAs with a focus on lncRNA fingerprints in patients' blood.

2.
Bioengineered ; 13(4): 8617-8630, 2022 04.
Article in English | MEDLINE | ID: mdl-35322735

ABSTRACT

Hepatocellular carcinoma (HCC) is a common malignancy in the world, with high mortality and poor prognosis. Hepatitis B virus (HBV) is one of the key factors implicated in the occurrence of HCC. Increasing evidence suggests that miRNAs play important roles in the development and metastasis of HBV-associated HCC (HBV-HCC). Here, we performed CCK8 (Cell count kit-8), EdU (5-ethynyl-2'-deoxyuridine) incorporation assay, wound-healing assay, transwell assay to study the changes in the cellular phenotype. Luciferase reporter assay, RNA pull-down experiment, RT-qPCR and western blotting were employed to study molecular mechanism. In addition, we also constructed a mouse HCC xenograft model to verify the functional role of HMMR-AS1/miR-627-3p/HMGA2 signal axis in vivo. Our study demonstrated that HMMR-AS1 was highly expressed in HCC tissues and cell lines, suggesting its implication in the progression of HCC. In addition, in vitro experiments showed that high HMMR-AS1 expression facilitated the migration, invasion, and proliferation of HCC cells. We further revealed that HMMR-AS1 promoted the malignant phenotype of HCC cells by regulating miR-627-3p/HMGA2 axis. Together, our data suggest that HMMR-AS1 regulates HBV-HCC progression via miR-627-3p/HMGA2 axis.


Subject(s)
Carcinoma, Hepatocellular , Liver Neoplasms , MicroRNAs , RNA, Long Noncoding , Animals , Carcinoma, Hepatocellular/metabolism , Cell Line, Tumor , Cell Proliferation/genetics , Extracellular Matrix Proteins , Hepatitis B virus/genetics , Humans , Hyaluronan Receptors , Liver Neoplasms/metabolism , Mice , MicroRNAs/genetics , MicroRNAs/metabolism , RNA, Antisense , RNA, Long Noncoding/genetics , RNA, Long Noncoding/metabolism
3.
Bioengineered ; 12(1): 7893-7906, 2021 12.
Article in English | MEDLINE | ID: mdl-34719318

ABSTRACT

Sevoflurane (Sev) is a volatile anesthetic that can inhibit tumor malignancy. Glioma is a main brain problem, but the mechanism of Sev in glioma progression is largely unclear. This study aims to explore a potential regulatory network of long noncoding RNA (lncRNA)/microRNA (miRNA)/mRNA associated with the function of Sev in glioma progression. LncRNA HMMR antisense RNA 1 (HMMR-AS1), miR-7 and cyclin-dependent kinase 4 (CDK4) abundances were examined via quantitative reverse transcription polymerase chain reaction and western blot. Cell viability, invasion, and colony formation ability were analyzed via cell counting kit-8, transwell analysis, and colony formation. The target association was analyzed via dual-luciferase reporter analysis and RNA pull-down. The in vivo function of Sev was investigated by xenograft model. HMMR-AS1 abundance was increased in glioma tissues and cells, and reduced via Sev. Sev constrained cell viability, invasion, and colony formation ability via decreasing HMMR-AS1 in glioma cells. miR-7 expression was decreased in glioma, and was targeted via HMMR-AS1. HMMR-AS1 silence restrained cell viability, invasion, and colony formation ability by up-regulating miR-7 in glioma cells. Sev increases miR-7 abundance via decreasing HMMR-AS1. CDK4 was targeted via miR-7, and highly expressed in glioma. miR-7 overexpression inhibited cell viability, invasion, and colony formation ability via reducing CDK4 in glioma cells. CDK4 expression was reduced by Sev via HMMR-AS1/miR-7 axis. Sev suppressed cell growth in glioma by regulating HMMR-AS1. Sev represses glioma cell progression by regulating HMMR-AS1/miR-7/CDK4 axis.


Subject(s)
Brain Neoplasms/metabolism , Glioma/metabolism , MicroRNAs/genetics , Sevoflurane/pharmacology , Animals , Cell Line, Tumor , Cell Survival/drug effects , Cyclin-Dependent Kinase 4/genetics , Cyclin-Dependent Kinase 4/metabolism , Disease Progression , Male , Mice , Mice, Inbred BALB C , MicroRNAs/metabolism , Neoplasm Invasiveness/genetics , RNA, Long Noncoding/genetics , RNA, Long Noncoding/metabolism
4.
Aging (Albany NY) ; 11(10): 3041-3054, 2019 05 25.
Article in English | MEDLINE | ID: mdl-31128573

ABSTRACT

PURPOSE: Long noncoding RNAs (lncRNA) play critical roles in cancer development. In this study, we aimed to explore the function and possible molecular mechanism of HMMR-AS1 involved in lung adenocarcinoma (LUAD). EXPERIMENTAL DESIGN: Firstly, we analyzed HMMR-AS1 expression in LUAD tissues with the sequencing data from The Cancer Genome Atlas (TCGA). Next, we evaluated the effects of HMMR-AS1 on LUAD cell proliferation and apoptosis, and its regulation of miR-138 by acting as a ceRNA. The animal model was used to support the in vitro experimental findings. RESULTS: HMMR-AS1 expression was significantly upregulated in LUAD tissues and was associated with larger tumor diameter, advanced TNM stage, lymph node metastasis, and shorter survival. Knockdown of HMMR-AS1 induced apoptosis and growth arrest in vitro and inhibited tumorigenesis in mouse xenografts. Mechanistically, HMMR-AS1 functioned as a ceRNA of miR-138, thereby leading to repression of its endogenous target sirt6. Moreover, knockdown of HMMR-AS1 dramatically inhibited tumor growth and metastasis of LUAD in vivo. CONCLUSIONS: Taken together, HMMR-AS1 is significantly over-expressed in LUAD, and HMMR-AS1-miR-138-sirt6 axis play a critical role in LUAD tumorigenesis. Our findings highlight an oncogenic role of HMMR-AS1 in LUAD.


Subject(s)
Adenocarcinoma/metabolism , Lung Neoplasms/metabolism , RNA, Long Noncoding/metabolism , A549 Cells , Case-Control Studies , Extracellular Matrix Proteins/genetics , Humans , Hyaluronan Receptors/genetics , Lung Neoplasms/genetics , MicroRNAs/metabolism , RNA, Long Noncoding/genetics , Sirtuins/metabolism
5.
Article in Chinese | WPRIM (Western Pacific) | ID: wpr-821736

ABSTRACT

Objective@#The purpose of this study is to explore the biological function of long non-coding RNA (lncRNA) HMMR-AS1 in proliferation and metastasis of lung adenocarcinoma (LUAD). @*Methods@#Real-time fluorescence quantitative polymerase chain reaction (RT-qPCR) was used to detect the expression of HMMR-AS1 and its sense strand HMMR in LUAD cell lines. Then we knock down the HMMR-AS1 expression through small interfering RNA and evaluate the transfection efficiency and its effect on the expression of HMMR. CCK-8 (cell counting kit), clone formation, flow cytometric analysis, wound scratch assay and transwell assay were used to assess the biological function of A549 and H1299 cells. Western blot was used to detect the protein expression of HMMR in the two cell lines after transfection with si-HMMR-AS1. @*Results@#The expression of HMMR-AS1 in A549 and H1299 cells of LUAD cell line was markedly higher than that in normal lung epithelial cell BEAS-2A by upregulating approximately 3.06 and 5.02 folds (P<0.05), respectively. After transfection with si-HMMR-AS1, the expression of HMMR-AS1 markedly reduced in both levels of transcription and protein (P<0.05). Furthermore, knocking down of HMMR-AS1 significantly inhibited the proliferation, migration and invasion abilities, and increased the apoptosis rates of A549 and H1299 cells. @*Conclusion@#LncRNA HMMR-AS1 could promote malignant progression of LUAD cells through enhancing the growth, migration and invasion ability of LUAD cells.

SELECTION OF CITATIONS
SEARCH DETAIL