Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 18 de 18
Filter
1.
Int Immunopharmacol ; 121: 110506, 2023 Aug.
Article in English | MEDLINE | ID: mdl-37343371

ABSTRACT

Osteoarthritis (OA) is a prevalent disease among elderly individuals that is caused by cartilage degeneration. Chondrocyte senescence involved in the development of OA, and antisenescence therapies have been proposed for OA treatment. In our study, we identified the role of a microRNA, miR-33-5p, in promoting chondrocyte senescence and OA progression. miR-33-5p expression was upregulated under senescence conditions. miR-33-5p-mimic transfection can induce cellular senescence, while transfection of a miR-33-5p-inhibitor in chondrocytes alleviated senescence induced by IL-1ß. Moreover, SIRT6 expression was downregulated under IL-1ß treatment, and could be restored by miR-33-5p-inhibitor transfection. Luciferase assays revealed that miR-33-5p targeted the SIRT6 mRNA 3' UTR. In addition, SIRT6 mRNA expression showed negative correlations with senescence and OA degree in human cartilage. Bioinformatic analysis also confirmed the pro-senescence effect of miR-33-5p. Furthermore, periodic intraarticular injection of agomiR-33-5p induced cartilage loss and OA-like cartilage changes. To conclude, we revealed the pro-senescence and cartilage-destructive effect of miR-33-5p, whose expression was elevated under various senescence conditions, and showed that SIRT6 was one of its targets. Therefore, miR-33-5p is a potential therapeutic target for treating OA.


Subject(s)
MicroRNAs , Osteoarthritis , Sirtuins , Humans , Aged , Chondrocytes , MicroRNAs/metabolism , Osteoarthritis/metabolism , Interleukin-1beta/metabolism , Apoptosis , RNA, Messenger/metabolism , Sirtuins/metabolism
2.
Int J Oncol ; 60(5)2022 05.
Article in English | MEDLINE | ID: mdl-35302171

ABSTRACT

Chronic myeloid leukemia (CML) is a hematological disease, and imatinib (IM) resistance represents a major problem for its clinical treatment. In the present study, the role of tribbles pseudokinase 2 (TRIB2) in IM resistance of CML and the possible mechanism were investigated. It was found that TRIB2 was highly expressed in IM­resistant patients with CML through the Oncomine database and this conclusion was confirmed using reverse transcription­quantitative PCR and western blot experiments. Knockdown of TRIB2 was found to increase the drug sensitivity of KG cells to IM using Cell­Counting Kit­8 (CCK­8) assays, and the low­expression TRIB2 mice were further found to be more sensitive to the IM and have a higher survival rate in leukemia model mice. Moreover, using western blot and luciferase experiments, it was found that TRIB2 could regulate c­Fos through the ERK signaling pathway, and c­Fos suppressed the transcriptional activity and the expression of miR­33a­5p. Further investigation identified that the binding site for c­Fos to function on miR­33a­5p was the ­958­965 region. Finally, CCK­8 assays and western blot experiments demonstrated that miR­33a­5p could inhibit the proliferation of KG cells and reduce IM resistance by suppressing the expression of HMGA2. In conclusion, it was demonstrated that TRIB2 regulates miR­33a­5p to reverse IM resistance in CML, which may help identify novel targets and therapeutic strategies for the clinical treatment of IM resistance.


Subject(s)
Gene Expression Regulation, Neoplastic/drug effects , Imatinib Mesylate/pharmacology , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/drug therapy , MAP Kinase Signaling System/drug effects , Animals , Calcium-Calmodulin-Dependent Protein Kinases/metabolism , Cell Line, Tumor/drug effects , Drug Resistance, Neoplasm/drug effects , Humans , Imatinib Mesylate/therapeutic use , Mice , MicroRNAs/metabolism , Signal Transduction/drug effects
3.
Cell Cycle ; 21(6): 572-584, 2022.
Article in English | MEDLINE | ID: mdl-35090377

ABSTRACT

Long non-coding RNA tumor protein 53 target gene 1 (TP53TG1) has been unraveled to exert regulatory effects on cancer progression, while the regulatory function of TP53TG1 on cervical cancer (CC) via regulating microRNA (miR)-33a-5p/Forkhead box K2 (FOXK2) axis remains rarely explored. This study aims to unearth the regulatory mechanism of TP53TG1/miR-33a-5p/FOXK2 axis in CC. The CC clinical samples were collected, and CC cells were cultured. TP53TG1, miR-33a-5p and FOXK2 levels were examined in CC tissues and cells. The CC cells were transfected with high- or low-expressed TP53TG1, FOXK2 or miR-33a-5p to determine the changes of CC cell biological activities and the status of phosphatidylinositol 3-kinase/protein kinase B/mammalian target of rapamycin (PI3K/AKT/mTOR) pathway. The tumorigenesis in nude mice was conducted. The relationship among TP53TG1, miR-33a-5p and FOXK2 was validated. TP53TG1 and FOXK2 expression levels were increased and miR-33a-5p expression level was reduced in CC cells and tissues. The silenced TP53TG1 or FOXK2, or elevated miR-33a-5p decelerated the CC cell development and restrained the activation of PI3K/AKT/mTOR signaling pathway. The depleted FOXK2 or elevated miR-33a-5p reversed the effects of decreased TP53TG1 on CC cell progression. TP53TG1 sponged miR-33a-5p, which targeted FOXK2. The experiment in vivo validated the outcomes of the experiment in vitro. TP53TG1 accelerates the CC development via regulating miR-33a-5p to target FOXK2 with the involvement of PI3K/AKT/mTOR signaling pathway. This study provides novel theory basis and distinct therapeutic targets for CC treatment.


Subject(s)
DNA-Binding Proteins , Forkhead Transcription Factors , MicroRNAs , RNA, Long Noncoding , Uterine Cervical Neoplasms , Animals , Cell Line, Tumor , Cell Proliferation/genetics , DNA-Binding Proteins/genetics , Female , Forkhead Transcription Factors/genetics , Gene Expression Regulation, Neoplastic , Humans , Mammals/metabolism , Mice , Mice, Nude , MicroRNAs/genetics , MicroRNAs/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Proto-Oncogene Proteins c-akt/metabolism , RNA, Long Noncoding/genetics , RNA, Long Noncoding/metabolism , TOR Serine-Threonine Kinases/metabolism , Uterine Cervical Neoplasms/pathology
4.
Article in Chinese | WPRIM (Western Pacific) | ID: wpr-955378

ABSTRACT

Objective:To investigate the correlation between plasma microRNA (miR)-122, miR-33a and the severity of coronary artery disease in patients with type 2 diabetes mellitus (T2DM) and coronary heart disease.Methods:The clinical data of 196 patients with T2DM from January 2019 to October 2021 in Xuzhou First People′s Hospital were retrospectively analyzed. Among them, 81 cases were complicated with coronary heart disease (combined group), 115 cases were not complicated with coronary heart disease (control group). The plasma levels of miR-122 and miR-33a were detected by real-time fluorescence quantitative reverse transcription polymerase chain reaction, the plasma level of N-terminal B-type natriuretic peptide precursor (NT-proBNP) was detected by enzyme-linked immunosorbent assay. In combined group, the number of coronary artery lesions was determined according to the results of coronary angiography, and Gensini score was evaluated. Linear regression model was used to analyze the relationship between plasma miR-122, miR-33a and NT-proBNP levels with the incidence of coronary heart disease in patients with T2DM. Receiver operating characteristic (ROC) curve was used to analyze the plasma miR-122 and miR-33a in predicting efficiency of coronary heart disease in patients with T2DM. In combined group, Spearman correlation method was used to analyze the relationship between plasma miR-122, miR-33a and the number of coronary artery lesions, and Pearson correlation method was used to analyze the relationship between plasma miR-122, miR-33a and plasma NT proBNP, Gensini score.Results:The plasma miR-122, miR-33a and NT-proBNP in combined group were significantly higher than those in control group: 5.76 ± 1.35 vs. 1.18 ± 0.33, 1.39 ± 0.37 vs. 0.65 ± 0.11 and (786.87 ± 156.39) ng/L vs. (103.45 ± 19.27) ng/L respectively, and there were statistical differences ( P<0.01). Linear regression result showed that plasma miR-122, miR-33a, and NT-proBNP were positive correlation with occurrence of coronary heart disease in patients with T2DM ( P<0.01); ROC curve analysis result showed that the area under curve of plasma miR-122, miR-33a and combination in predicting coronary heart disease in patients with T2DM were 0.816, 0.845 and 0.912 respectively (95% CI 0.744 to 0.865, 0.768 to 0.892 and 0.836 to 0.967). Coronary angiography result showed that there were 46 cases of single vessel lesions, 25 cases of double vessel lesions and 10 cases of three vessel lesions. The plasma miR-122, miR-33a, NT-proBNP and Gensini score in patients with three vessel lesions were significantly higher than those in patients with double vessel lesions and patients with single vessel lesions: 6.52 ± 0.96 vs. 4.95 ± 0.85 and 3.74 ± 0.52, 1.45 ± 0.31 vs. 1.06 ± 0.25 and 0.81 ± 0.13, (829.78 ± 62.59) ng/L vs. (627.48 ± 47.12) and (502.64 ± 38.24) ng/L, (63.89 ± 12.71) scores vs. (42.18 ± 6.03) and (22.36 ± 2.41) scores, the indexes in patients with double vessel lesions were significantly higher than those in patients with single vessel lesions, and there were statistical differences ( P<0.05). In combined group, Spearman correlation analysis result showed that the plasma miR-122 and miR-33a were positive correlation with the number of coronary artery lesions ( r = 0.879 and 0.825, P<0.05); Pearson correlation analysis result showed that the plasma miR-122 and miR-33a were positive correlation with the plasma NT-proBNP and Gensini score (miR-122: r = 0.896 and 0.788, miR-33a: r = 0.871 and 0.765; P<0.05). Conclusions:The plasma levels of miR-122 and miR-33a are related to the occurrence of coronary heart disease and severity of coronary artery disease in patients with T2DM, which may be used to guide the prevention and treatment of coronary heart disease in patients with T2DM.

5.
Oncol Lett ; 21(6): 489, 2021 Jun.
Article in English | MEDLINE | ID: mdl-33968205

ABSTRACT

Several chemotherapeutic options are available for patients with metastatic colorectal cancer (mCRC), making it important to individualize treatment regimens. Individualization requires the clinical application of biomarkers for regimen selection, which is presently insufficient. miRNAs serve an important role in the control of biological processes in several types of cancer, acting as plasma biomarkers. The current study aimed to evaluate novel plasma microRNAs for predicting chemo-resistance in chemotherapy for patients with colorectal cancer (CRC) by employing a Toray 3D-Gene microRNA array-based approach, which compared plasma content before and during treatment. Specific miRNAs that acted as biomarkers of the fluoropyrimidine (FP) + oxaliplatin (OX) + bevacizumab (BEV) regime, a common first-line treatment for mCRC, were searched. The plasma samples of 110 patients with mCRC who had received the FP+OX+BEV regimen were subjected to microarray analyses using the 3D-Gene miRNA microarray platform, after which miRNAs levels were quantified via reverse transcription- quantitative PCR. Patients exhibiting complete response, partial response (PR) and reduced stable disease (SD) were defined as responders. Patients with extended SD and progression disease (PD) were defined as non-responders. Following microarray analysis, miR-33a-5p was selected as the candidate miRNA as it was upregulated in non-responder plasma samples. The expression of miR-33a-5p was upregulated in the non-responders (n=15) compared with the responders (n=95) (P=0.032). The high expression group demonstrated significantly poor progression-free survival (P<0.01). To evaluate whether miR-33a-5p can serve as a marker of chemo-resistance, miR-33a-5p expression levels were assessed at the following three time-points: Pre-point (before chemotherapy); PR-point (3-months after chemotherapy began); and PD-point (the time at which recurrence or progression was recorded). The results revealed that expression levels were significantly increased at the PD-point when compared with that at the pre-point (P=0.024). The current study determined that the miR-33a-5p expression level in the plasma may serve as a predictive marker of efficacy and as a biomarker of chemo-resistance.

6.
Exp Ther Med ; 21(3): 205, 2021 Mar.
Article in English | MEDLINE | ID: mdl-33500698

ABSTRACT

The present study aimed to detect the levels of microRNA (miR)-33a-5p in the renal tissue, serum and urine of patients with primary IgA nephropathy (IgAN), thereby preliminarily exploring the association between the levels of miR-33a-5p and the condition of primary IgAN to provide evidence for the expression of miR-33a-5p in the serum and urine of IgAN patients as a clinical marker. Reverse-transcription quantitative PCR was performed to evaluate the level of miR-33a-5p in IgAN patients according to severity and pathological classification. The results suggested that the levels of miR-33a-5p in the serum, urine and kidney tissues of patients with IgAN were lower than those of the control tissues obtained from cancer patients (0.28±0.25 vs. 1.00±0.45, P<0.05; 0.34±0.28 vs. 1.00±0.53, P<0.05; 0.47±0.27 vs. 1.00±0.38, P<0.05, respectively). Receiver operating characteristic curve analysis suggested that the serum and urine levels of miR-33a-5p may be used as a marker to differentiate renal injury in IgAN patients from healthy individuals. At the same time, according to the estimated glomerular filtration rate (eGFR) and Lee classification of nephropathy, it was determined that with the progression of renal failure and the increase of the pathological grade of kidney tissue, the relative level of miR-33a-5p in kidney tissue also decreased (eGFR <50 ml/min vs. eGFR ≥50 ml/min/1.73 m2 group: 0.38±0.27 vs. 1.00±0.34, P<0.001; Lee grade ≤3 group vs. Lee grade >3: 1.00±0.48 vs. 0.38±0.45, P<0.05). This result suggested that the levels of miR-33a-5p in serum, urine and kidney tissues decreased with the severity of renal injury and the progression of renal failure in patients with IgAN. Hence, miR-33a-5p detected in the serum and urine may be used as a non-invasive biomarker to reflect the progression of renal injury and renal failure in patients with IgAN.

7.
Oncol Lett ; 18(6): 5986-5994, 2019 Dec.
Article in English | MEDLINE | ID: mdl-31788073

ABSTRACT

Drug resistance is a significant obstacle when treating triple-negative breast cancer (TNBC). Several studies have demonstrated that microRNAs (miRNAs) have essential roles in regulating drug resistance in different types of cancer. miR-33a-5p has previously been reported to be a tumor suppressor in several types of cancer. However, its role in breast cancer remains unknown. The present study aimed to investigate the role of miR-33a-5p in the chemoresistance of TNBC and uncover its potential molecular mechanisms. Cell Counting Kit-8 assay was used to examine cell proliferation, reverse transcription-quantitative PCR analysis was used to examine miR-33a levels, and western blotting and immunofluorescence assays were used to examine the expression of epithelial-mesenchymal transition (EMT)-associated proteins and of eukaryotic translation initiation factor 5A2 (eIF5A2). The results indicated that miR-33a-5p expression was lower in TNBC cells compared with non-TNBC cells. miR-33a-5p overexpression significantly improved the doxorubicin (Dox) sensitivity of TNBC cells, but not that of non-TNBC cells. It was then observed that Dox treatment inhibited miR-33a-5p expression and induced EMT in TNBC cells, by increasing the expression levels of vimentin, while decreasing the expression levels of E-cadherin. Furthermore, it was revealed that forced expression of miR-33a-5p attenuated Dox-induced EMT. eIF5A2 was identified as a potential target of miR-33a-5p, and miR-33a-5p overexpression inhibited the expression of eIF5A2. eIF5A2 inhibition, via its inhibitor GC7, sensitized TNBC cells to Dox and reversed Dox-induced EMT. Overall, the present study demonstrated that miR-33a-5p enhanced the sensitivity of TNBC cells to Dox, by suppressing eIF5A2 expression and reversing Dox-induced EMT, providing a potential therapeutic target for treating drug-resistant TNBC.

8.
Fitoterapia ; 139: 104393, 2019 Nov.
Article in English | MEDLINE | ID: mdl-31669721

ABSTRACT

The extract of Dioscorea zingiberensis C.H. Wright rhizomes is found to be effective in the therapy of cardiovascular disease. Steroidal saponins make substantial contribution. Previous study has proposed that methylprotodioscin (MP) may promote cholesterol efflux by increasing ABCA1 expression. But the other main saponins ingredients are not referred to. The aim of the present work was to reveal the effect and mechanism of protodioscin (PD), MP and pseudoprotodioscin (PPD) on the synthesis-related gene expression of cholesterol and triglycerides. MTT assay apoptosis assay with annexin AV-APC and 7-AAD double staining were performed. MicroRNA assay and qRT-PCR were used to analyze the gene expression which regulates synthesis of cholesterol and triglycerides. Western blot was to demonstrate the levels of target proteins. Cholesterol efflux assay was executed to study the stimulative effect of saponins on cholesterol efflux. In Hep G2 cells, PPD increased ABCA1 protein and mRNA levels, and promoted the effluxion of ApoA-1-mediated cholesterol. The underlying mechanisms involved that PPD inhibited SREBP1c and SREBP2 transcription by decreasing microRNA 33a/b levels. This procedure reciprocally led to the increase of ABCA1 levels. In THP-1 macrophages, PPD showed the similar effect, which reduced HMGCR, FAS and ACC mRNA levels and promoted low density lipoprotein receptor by decreasing the PCSK9 levels. These studies demonstrated that PPD is a potential agent for cholesterol efflux, SREBPs and microRNA 33a/b inhibition, which related to the gene expression for the synthesis of cholesterol and triglycerides.


Subject(s)
Cholesterol/biosynthesis , Diosgenin/analogs & derivatives , MicroRNAs/antagonists & inhibitors , Saponins/pharmacology , Sterol Regulatory Element Binding Protein 1/antagonists & inhibitors , Sterol Regulatory Element Binding Protein 2/antagonists & inhibitors , Triglycerides/biosynthesis , ATP Binding Cassette Transporter 1/metabolism , Dioscorea/chemistry , Diosgenin/pharmacology , Hep G2 Cells , Humans , MicroRNAs/genetics , Plant Extracts/pharmacology , Proprotein Convertase 9/metabolism , Receptors, LDL/metabolism , Rhizome/chemistry , THP-1 Cells
9.
Cell Cycle ; 18(19): 2524-2537, 2019 Oct.
Article in English | MEDLINE | ID: mdl-31416393

ABSTRACT

At present, some researches have revealed the participation of long noncoding RNAs (lncRNAs) in liver cancer, but few of them have mentioned the role of CRNDE in drug resistance of liver cancer. Hence, this study is conducted to understand the role of CRNDE on liver cancer by regulating microRNA-33a (miR-33a) and high mobility group protein A2 (HMGA2) in liver cancer. First, drug-resistance model (HepG2 and BEL-7402) of human liver cancer cells was established. Then, CRNDE expression in drug-resistant cell lines (HepG2/adriamycin [ADM], BEL-7402/ADM) and parental cell lines (HepG2, BEL-7402) was detected. Furthermore, HepG2/ADM and BEL-7402/ADM cell lines with poor CRNDE expression or miR-33a overexpression was constructed. Next, drug-resistance index was calculated, and cell proliferation, apoptosis, migration, and invasion were detected, respectively. Then, the growth of tumor was observed in nude mice. Finally, the binding relationship between CRNDE and miR-33a and the targeting relationship between miR-33a and HMGA2 were verified. LncRNA CRNDE expressed highly in drug-resistant cells of liver cancer. Downregulated CRNDE and upregulated miR-33a-inhibited cells drug-resistance and promoted their apoptosis in liver cancer drug-resistant cells. CRNDE adsorbing and inhibiting miR-33a to promote HMGA2 in liver cancer drug-resistant cells by acting as a ceRNA. Silencing CRNDE or up-regulating miR-33a inhibited tumor growth of liver cancer in vivo. Our study provides evidence that downregulated CRNDE could upregulate miR-33a and inhibit HMGA2 expression, thus significantly promotes apoptosis of liver cancer cells and inhibiting its proliferation, migration, invasion and drug resistance.


Subject(s)
Antineoplastic Agents/pharmacology , Drug Resistance, Neoplasm/genetics , HMGA2 Protein/metabolism , Liver Neoplasms/metabolism , MicroRNAs/metabolism , RNA, Long Noncoding/metabolism , Animals , Apoptosis/drug effects , Apoptosis/genetics , Cell Line, Tumor , Cell Movement/drug effects , Cell Movement/genetics , Cell Proliferation/drug effects , Cell Proliferation/genetics , Down-Regulation , Gene Expression Regulation, Neoplastic/genetics , Gene Silencing , HMGA2 Protein/genetics , Humans , Liver Neoplasms/genetics , Mice , Mice, Inbred BALB C , Mice, Nude , MicroRNAs/genetics , RNA, Long Noncoding/genetics , Transplantation, Heterologous , Up-Regulation
10.
J Cell Biochem ; 120(12): 19868-19877, 2019 12.
Article in English | MEDLINE | ID: mdl-31318088

ABSTRACT

The importance of long noncoding RNAs (lncRNAs) has been certified in malignant melanoma. Nonetheless, the functions of lncRNA paternally expressed gene 10 (PEG10) in malignant melanoma remain uninvestigated. This research discloses the influence of PEG10 in the biological actions of malignant melanoma cells. The sh-PEG10 plasmid was transfected into A375 cells; meanwhile, the effects of declined PEG10 on cell viability, apoptosis, migration, invasion, and the correlative protein levels were probed. The miR-33a expression in sh-PEG10-transfected cells was examined, and the above biological processes were studied again in miR-33a inhibitor-transfected A375 cells. Phosphoinositide 3-kinase/protein kinase B (PI3K/AKT) and mechanistic target of rapamycin (mTOR) pathways were delved via Western blot. We found that the enhancement of PEG10 was discovered in melanoma tissues compared to related nonmelanoma tissues. Declination of PEG10 frustrated cell viability, repressed cyclinD1 and CDK4 expression, and triggered apoptosis, as well as suppressed migration and invasion in A375 cells. A negative correction between PEG10 and miR-33a was confirmed, and repressed miR-33a inverted the functions of PEG10 repression in A375 cells. In addition, PEG10 repression discouraged the activation of PI3K/AKT and mTOR pathways via elevation of miR-33a. These results indicated that declination of PEG10 restrained A375 cell growth, migration, and invasion via adjusting miR-33a and PI3K/AKT and mTOR pathways.


Subject(s)
Melanoma/genetics , Melanoma/pathology , MicroRNAs/genetics , RNA, Long Noncoding/genetics , Adult , Cell Line, Tumor , Cell Movement/genetics , Cell Proliferation/genetics , Female , Gene Expression Regulation, Neoplastic , Gene Knockdown Techniques , Humans , Male , Middle Aged , Phosphatidylinositol 3-Kinases/genetics , Phosphatidylinositol 3-Kinases/metabolism , Proto-Oncogene Proteins c-akt/genetics , Proto-Oncogene Proteins c-akt/metabolism , TOR Serine-Threonine Kinases/genetics , TOR Serine-Threonine Kinases/metabolism , Up-Regulation
11.
Clin Exp Pharmacol Physiol ; 46(10): 928-936, 2019 10.
Article in English | MEDLINE | ID: mdl-31209892

ABSTRACT

Colorectal cancer (CRC) is one of the most common malignancies with high levels of invasiveness, drug resistance and mortality, but the internal mechanisms of CRC are largely unknown. MicroRNAs (miRs) have been reported to be involved in the development of CRC, and numerous studies have demonstrated that the abnormal expression of miR-33a-5p might be associated with CRC. However, the function and downstream mechanism of miR-33a-5p in colorectal cancer (CRC) remains unclear. Methylenetetrahydrofolate dehydrogenase 2 (MTHFD2), a mitochondrial enzyme involved in folic acid metabolism, interestingly was confirmed to be one of the target genes of miR-33a-5p in the present study. We first confirmed that miR-33a-5p in CRC tissues and cell lines were downregulated (P < 0.05), and that the proliferation, clone formation capacities, G1/S progression, and migration capacities of the two CRC cell lines HCT116 cells and HT29 were suppressed by miR-33a-5p overexpression in vitro (P < 0.05). Ctrl HCT116 and miR-33a-5p-overexpressing HCT116 were injected into nude mice. In vivo tumour formation was significantly suppressed by miR-33a-5p overexpression (P < 0.05) as well as the proliferation marker Ki67 (P < 0.05). Additionally, MTHFD2 protein expression was significantly enhanced in CRC tissues. From bioinformatics predictions and a luciferase report analysis, MTHFD2 was confirmed to be one of the target genes of miR-33a-5p. In contrast to the role of miR-33a-5p overexpression, MTHFD2 overexpression promoted the proliferation and migration of HCT116 and HT29 cells (P < 0.05), which confirmed that MTHFD2 was a functional target gene of miR-33a-5p. In conclusion, miR-33a-5p inhibits the growth and migration of CRC by targeting MTHFD2.


Subject(s)
Aminohydrolases/genetics , Colorectal Neoplasms/genetics , Colorectal Neoplasms/pathology , Methylenetetrahydrofolate Dehydrogenase (NADP)/genetics , MicroRNAs/genetics , Multifunctional Enzymes/genetics , Animals , Base Sequence , Cell Line, Tumor , Cell Movement/genetics , Cell Proliferation/genetics , Female , G1 Phase Cell Cycle Checkpoints/genetics , Gene Expression Regulation, Neoplastic , Humans , Male , Mice , Middle Aged , S Phase Cell Cycle Checkpoints/genetics
12.
Am J Physiol Gastrointest Liver Physiol ; 317(2): G147-G160, 2019 08 01.
Article in English | MEDLINE | ID: mdl-30943047

ABSTRACT

Invasion and metastasis are responsible for the majority of deaths in gastric cancer (GC). microRNA-33a (miR-33a) might function as a tumor suppressor in multiple cancers. Here, we describe the regulation and function of miR-33a in GC and mechanisms involved in epithelial-mesenchymal transition (EMT) and metastasis. First, GC tissues and adjacent normal tissues were collected. miR-33a upregulation or SNAI2 depletion on GC cells were introduced to assess the detailed regulatory mechanism of them. We assessed the expression of miR-33a, SNAI2, Snail/Slug signaling pathway-related genes, and EMT-related markers in GC tissues and cells. miR-33a distribution in GC tissues and adjacent normal tissues was measured. Cell proliferation, migration and invasion, and cell cycle distribution were assessed. In nude mice, GC tumor growth and lymph node metastasis were observed. Furthermore, the predicative value of miR-33a in the prognosis of GC patients was evaluated. The obtained results indicated that lowly expressed miR-33a, highly expressed SNAI2, activated Snail/Slug, and increased EMT were identified in GC tissues. miR-33a was located mainly in the cytoplasm. miR-33a targeted and negatively regulated SNAI2. MKN-45 and MKN-28 cell lines were selected for in vitro experiments. Upregulated miR-33a expression or siRNA-mediated silencing of SNAI2 suppressed the activation of Snail/Slug, whereby GC cell proliferation, invasion and migration, EMT, tumor growth, and lymph node metastasis were inhibited. High expression of miR-33a was a protective factor influencing the prognosis of GC. This study suggests that miR-33a inhibited EMT, invasion, and metastasis of GC through the Snail/Slug signaling pathway by modulating SNAI2 expression.NEW & NOTEWORTHY miR-33a targets and inhibits the expression of SNAI2, overexpression of SNAI2 activates the Snail/Slug signaling pathway, the Snail/Slug signaling pathway promotes GC cell proliferation, invasion, and metastasis, and overexpression of miR-33a inhibits cell proliferation, invasion, and metastasis. This study provides a new therapeutic target for the treatment of GC.


Subject(s)
MicroRNAs/metabolism , Snail Family Transcription Factors/metabolism , Stomach Neoplasms , Animals , Cell Line, Tumor , Cell Proliferation , Epithelial-Mesenchymal Transition , Gene Expression Regulation, Neoplastic , Humans , Mice , Mice, Nude , Neoplasm Invasiveness , Neoplasm Metastasis , Signal Transduction , Stomach Neoplasms/metabolism , Stomach Neoplasms/pathology
13.
J Cell Mol Med ; 22(12): 6285-6293, 2018 12.
Article in English | MEDLINE | ID: mdl-30324697

ABSTRACT

Emerging evidence suggests that microRNAs (miRNAs) are essential for metabolic haemostasis of liver tissues. Among them, miR-33a is supposed to modulate the cholesterol export and fatty acid oxidation, but whether miR-33a involves in the process of fatty liver disease is unclear. To disclose the hypothesis, we utilized miR-33a mimic and antisense to explore their effects in primary hepatocytes or high-fat diet (HFD)-fed mice. Treatment with palmitic acid (PA) or HFD significantly increased the expression of miR-33a in hepatocytes or liver tissues. In primary hepatocytes, miR-33a mimic decreased mitochondrial function, including reduction of ATP production and oxygen consumption, whereas miR-33a inhibition protected PA-induced mitochondrial dysfunction. Interestingly, miR-33a selectively suppressed mitochondrial complex I activity and protein expression, but not other complexes. Through bioinformatics prediction, we found miR-33a directly targeted on the 3'-UTR of NDUFA5. Dual-luciferase reporter analysis further confirmed the direct suppression of miR-33a on NDUFA5 expression. More importantly, administration of miR-33a antisense could effectively restore HFD-induced mitochondrial dysfunction through up-regulation of NDUFA5 levels. Mice treated with miR-33a antisense also exhibited improved liver function and structural disorders under obese status. Taken together, miR-33a was an important mediator of hepatocyte mitochondrial function, and the therapeutic benefits implied miR-33a antisense had the potential clinical application in combating the fatty liver disease.


Subject(s)
Fatty Liver/genetics , MicroRNAs/genetics , Mitochondria, Liver/genetics , NADH Dehydrogenase/genetics , Obesity/genetics , Adenosine Triphosphate/genetics , Animals , Diet, High-Fat , Disease Models, Animal , Fatty Liver/metabolism , Fatty Liver/pathology , Gene Expression Regulation/genetics , Hepatocytes/metabolism , Hepatocytes/pathology , Humans , Liver/metabolism , Liver/pathology , Mice , Mitochondria, Liver/metabolism , Mitochondria, Liver/pathology , Obesity/metabolism , Obesity/pathology , Oxygen Consumption/genetics , Palmitic Acid/metabolism , Primary Cell Culture
14.
Hepatol Res ; 48(5): 397-407, 2018 Apr.
Article in English | MEDLINE | ID: mdl-29243365

ABSTRACT

AIM: Liver fibrosis is a life-threatening disorder for which no approved therapy is available. Recently, we reported that mouse hepatic stellate cell (HSC) activation increased free cholesterol (FC) accumulation, partly by enhancing signaling through sterol regulatory element-binding protein 2 (SREBP2) and microRNA-33a (miR-33a), which resulted in HSC sensitization to transforming growth factor-ß (TGFß)-induced activation in a "vicious cycle" of liver fibrosis. METHODS: Human HSCs were isolated from surgical liver specimens from control patients and patients with liver fibrosis. C57BL/6 mice were treated with carbon tetrachloride for 4 weeks and concurrently given SREBP2-siRNA- or anti-miR-33a-bearing vitamin A-coupled liposomes. RESULTS: In human activated HSCs obtained from patients with liver fibrosis, FC accumulation was enhanced independently of serum cholesterol levels through increased signaling by both SREBP2 and miR-33a. This increased FC accumulation enhanced Toll-like receptor 4 (TLR4) protein levels and lowered the TGFß-pseudoreceptor Bambi (bone morphogenetic protein and activin membrane-bound inhibitor) mRNA levels in HSCs. Notably, in a mouse liver fibrosis model, reduction of FC accumulation, specifically in activated HSCs by suppression of SREBP2 or miR-33a expression using SREBP2-siRNA- or anti-miR-33a-bearing vitamin A-coupled liposomes, downregulated TLR4 signaling, increased Bambi expression, and consequently ameliorated liver fibrosis. CONCLUSIONS: Our results suggest that FC accumulation in HSCs, as an intracellular mediator promoting HSC activation, contributes to a vicious cycle of HSC activation in human and mouse liver fibrosis independent of serum cholesterol levels. Targeting FC accumulation-related molecules in HSCs through a vitamin A-coupled liposomal system represents a favorable therapeutic strategy for liver fibrosis.

15.
Exp Ther Med ; 13(5): 2507-2514, 2017 May.
Article in English | MEDLINE | ID: mdl-28565872

ABSTRACT

MicroRNA-33a (miR-33a) is dysregulated in a number of human cancers, where it functions as an oncogenic miRNA. However, the clinical significance of miR-33a and its underlying molecular pathways regarding the progression of hepatocellular carcinoma (HCC) are currently unknown. In the present study, it was observed that the level of miR-33a expression was significantly increased in HCC tissues, relative to adjacent non-tumor tissues. Increased miR-33a expression was significantly correlated with poor prognostic features of HCC, including larger tumor size, higher Edmondson-Steiner grading and higher tumor-node-metastasis tumor stage. Furthermore, high levels of miR-33a expression were associated with decreases in the 5-year overall survival rate and recurrence-free survival of patients with HCC. In addition, functional experiments indicated that overexpression of miR-33a led to increased proliferation and reduced apoptosis of the HCC cell line Huh7, while knockdown of miR-33a decreased proliferation and induced apoptosis in the HCC cell line HepG2. Furthermore, peroxisome proliferator activated receptor alpha (PPARα) was identified as a direct target of miR-33a in HCC. Upregulation of miR-33a was found to reduce the levels of PPARα expression in Huh7 cells, while inhibition of miR-33a lead to a downregulation in PPARα expression in HepG2 cells. Collectively, these results suggest that miR-33a regulates the proliferation and apoptosis of HCC cells, and is a potential prognostic marker of HCC.

16.
Oncol Lett ; 10(4): 2135-2141, 2015 Oct.
Article in English | MEDLINE | ID: mdl-26622808

ABSTRACT

A body of evidence has indicated that microRNAs (miRNAs) may have significant roles in cancer. Aberrant expression of miRNAs has frequently been observed in various human malignancies, including osteosarcoma (OS). However, the roles of miRNAs in OS remain poorly understood. In the present study, high-throughput deep sequencing was performed to screen for deregulated miRNAs in OS. Screening identified 310 miRNAs which were significantly overexpressed and 41 miRNAs which were significantly downregulated (>2-fold) in OS samples, compared with adjacent non-tumor bone tissues. Among these miRNAs, miR-33a-5p was notably downregulated. TaqMan reverse transcription-polymerase chain reaction analysis further verified that miR-33a-5p expression was significantly reduced in a large cohort of human OS samples. Enhancing miR-33a-5p expression via transfection with miR-33a-5p precursor significantly inhibited OS cell growth, suggesting potential antitumor properties of miR-33a-5p. The results of the present study provide novel insights into the miRNAs involved in OS, and suggest that miR-33a-5p may function as a tumor suppressor in OS. Therefore, miR-33a-5p may be able to serve as a diagnostic and therapeutic target for OS treatment.

17.
Cancer Biol Ther ; 16(6): 846-55, 2015.
Article in English | MEDLINE | ID: mdl-25891797

ABSTRACT

BACKGROUND: Our previous findings showed that miR-33 expressed abnormally in clinical specimens of melanoma, but the exact molecular mechanism has not been elucidated. OBJECT: To determine miR-33's roles in melanoma and confirm whether HIF-1α is a direct target gene of miR-33a. METHODS: First miR-33a/b expression levels were detected in HM, WM35, WM451, A375 and SK-MEL-1. Then lentiviral vectors were constructed to intervene miR-33a expression in melanoma cells. Cell proliferation, invasion and metastasis were detected. A375 cells mice model was performed to test the tumorigenesis of melanoma in vivo. Finally the dual reporter gene assay was carried out to confirm whether HIF-1α is a direct target gene of miR-33a. RESULTS: MiR-33a/b exhibited a lower expression in WM35, WM451, A375 and SK-MEL-1 of the metastatic skin melanoma cell lines than that in HM. Then inhibition of miR-33a expression in WM35 and WM451 cell lines could promote cell proliferation, invasion and metastasis. Conversely, increased expression of miR-33a in A375 cells could inhibit cellproliferation, invasion and metastasis. In vivo tests also confirmed that overexpression of miR-33a in A375 cells significantly inhibited melanoma tumorigenesis. Finally, we confirmed that HIF-1α is a direct target gene of miR-33a. CONCLUSION: The newly identified miR-33a/HIF-1α axis might provide a new strategy for the treatment of melanoma.


Subject(s)
Genes, Tumor Suppressor , Hypoxia-Inducible Factor 1, alpha Subunit/genetics , Melanoma/genetics , MicroRNAs/genetics , RNA Interference , Animals , Cell Line, Tumor , Cell Proliferation , Disease Models, Animal , Down-Regulation , Gene Expression , Humans , Melanoma/pathology , Mice , Xenograft Model Antitumor Assays
18.
Ther Apher Dial ; 18(1): 44-50, 2014 Feb.
Article in English | MEDLINE | ID: mdl-24499083

ABSTRACT

Atherosclerosis and accompanying cardiovascular disease are the first causes of mortality in patients undergoing maintenance hemodialysis. Anti-atherosclerotic effects of hemodiafiltration (HDF) have been reported. Our study aimed to investigate the effect of serum derived from a healthy group (n = 23), before and after hemodialysis (HD) therapy (n = 23), and before and after HDF therapy (n = 17) on the expression of microRNA-33a and its target genes adenosine triphosphate-binding cassette transporter A1,G1 (ABCA1, ABCG1) in THP-1 macrophages. Meanwhile, blood lipids and high-sensitivity C-reactive protein (hs-CRP) were measured in these groups. Our data showed that the expression of miRNA-33a was lower (P < 0.05) and ABCA1 and ABCG1 were higher (P < 0.05) in the healthy group than pre-HD and pre-HDF. miR-33a was significantly decreased (P < 0.05) but ABCA1, ABCG1 was significantly increased (P < 0.05) in post-HDF compared with pre-HDF, while these parameters in pre- and post- HD groups did not show any significant change (P > 0.05). High density lipoprotein cholesterol (HDL-C) was higher and hs-CRP was lower in the healthy group than pre-HD and pre-HDF groups. Moreover, a significant increase of HDL-C (P < 0.05) and decrease (P < 0.05) of hs-CRP was shown in post-HDF compared with pre-HDF, but HD appeared to have no significant change in these subjects. HDF therapy can downregulate miR-33a expression, and then result in ABCA1, ABCG1 upregulation and an increase in circulating HDL-C, leading to a possible anti-atherosclerosis effect to some extent.


Subject(s)
ATP Binding Cassette Transporter 1/genetics , ATP-Binding Cassette Transporters/genetics , MicroRNAs/genetics , Renal Dialysis/methods , ATP Binding Cassette Transporter, Subfamily G, Member 1 , Aged , Atherosclerosis/prevention & control , C-Reactive Protein/metabolism , Case-Control Studies , Cell Line , Cholesterol, HDL/blood , Down-Regulation , Female , Hemodiafiltration/methods , Humans , Macrophages/metabolism , Male , Middle Aged , Up-Regulation
SELECTION OF CITATIONS
SEARCH DETAIL
...