Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 28
Filter
1.
J Ovarian Res ; 16(1): 8, 2023 Jan 10.
Article in English | MEDLINE | ID: mdl-36627652

ABSTRACT

BACKGROUND: Chemotherapy is among the most common treatment methods for ovarian cancer (OC). However, chemoresistance limits the effectiveness of chemotherapy and leads to treatment failure. We herein investigate the biological effect of forkhead box D3 (FOXD3) in the chemoresistance of OC cells. METHODS: Expression of FOXD3, miR-335 and disheveled-associated activator of morphogenesis 1 (DAAM1) was detected in OC cells and tissues. The regulatory network of FOXD3/miR-335/DAAM1 was validated by dual-luciferase reporter and ChIP assays in vitro. After ectopic expression and depletion experiments in carboplatin/paclitaxel (CP)-resistant (A2780CP) or sensitive (A2780S) OC cells, cell viability, colony formation and apoptosis were tested by CCK-8 assay, colony formation assay and flow cytometry respectively. Effects of FOXD3 on the chemoresistance of OC cells in vivo were evaluated in OC xenografts in nude mice. RESULTS: Overexpression of FOXD3 impaired the proliferation and chemoresistance of OC cells, which was related to the promotion of the miR-335 expression. Functionally, DAAM1 was a putative target of miR-335. Silencing of DAAM1 was responsible for the inhibition of myosin II activation, consequently leading to suppressed OC cell proliferation and chemoresistance. In vivo results further showed that FOXD3 weakened the chemoresistance of OC cells to CP. CONCLUSION: Taken together, we unveil a novel FOXD3/miR-335/DAAM1/myosin II axis that regulates the chemoresistance of OC both in vitro and in vivo.


Subject(s)
Forkhead Transcription Factors , MicroRNAs , Ovarian Neoplasms , Animals , Female , Humans , Mice , Cell Line, Tumor , Cell Proliferation , Forkhead Transcription Factors/genetics , Gene Expression Regulation, Neoplastic , Mice, Nude , Microfilament Proteins , MicroRNAs/genetics , MicroRNAs/metabolism , Ovarian Neoplasms/drug therapy , Ovarian Neoplasms/genetics , Ovarian Neoplasms/metabolism , Paclitaxel/pharmacology , Paclitaxel/therapeutic use , rho GTP-Binding Proteins/metabolism , rho GTP-Binding Proteins/therapeutic use
2.
Article in Chinese | WPRIM (Western Pacific) | ID: wpr-989357

ABSTRACT

Objective:To investigate the clinical value of combined detection of serum immunoglobulin G (IgG), T-frame protein 21 (TBX21), and microRNA-335 (miR-335) in the diagnosis of lupus nephritis (LN).Methods:Ninety-five patients with LN treated in our hospital from January 2021 to January 2023 were selected as the observation group, while ninety-five healthy individuals were selected as the control group. Based on the systemic lupus erythematosus disease activity index (SLEDAI) score at admission, the LN patients were divided into two subgroups: the active LN group (51 cases, SLEDAI score > 4) and the stable LN group (44 cases, SLEDAI score = 0 - 4). The levels of serum IgG, TBX21, and miR-335 were compared between the two groups, and the levels of serum IgG, TBX21, miR-335, blood urea nitrogen (BUN), serum creatinine (Scr), complement C3, complement C4, and SLEDAI score were compared between the two groups. The correlations of serum IgG, PTX3, and miR-335 levels with BUN, Scr, complement C3, complement C4, and SLEDAI scores were analyzed. The diagnostic value of serum IgG, TBX21, and miR-335 in LN was evaluated.Results:Compared with the control group, the levels of serum IgG, TBX21, and miR-335 in the observation group were higher on admission (all P < 0.05). The serum levels of IgG, TBX21, and miR-335 in patients with the active stage were higher than those in patients with the stable stage on admission (all P < 0.05). On admission, the BUN, Scr, and SLEDAI scores of patients in the active stage were higher than those in the stable stage, and the levels of complement C3 and C4 were lower than those in the stable stage (all P < 0.05). The levels of serum IgG, TBX21, and miR-335 on admission were positively correlated with BUN, Scr, and SLEDAI scores and negatively correlated with complement C3 and C4 levels (all P < 0.05). The area under the curve (AUC) of the combination of serum IgG, TBX21, and miR-335 levels in the diagnosis of LN was greater than that of single detection ( P < 0.05). Conclusions:Serum IgG, TBX21, and miR-335 are closely related to disease activity and can be used as reference indicators for the diagnosis and prediction of LN in clinical practice, enabling the development of early targeted treatment plans.

3.
Int J Oncol ; 61(4)2022 Oct.
Article in English | MEDLINE | ID: mdl-36004482

ABSTRACT

Following the publication of this paper, it was drawn to the Editors' attention by a concerned reader that various panels showing the data from flow cytometry experiments in Figs. 2C, 4C and 6C, and certain of the tumor images featured in Fig. 9A, were strikingly similar to data appearing in different form in other articles by different authors. Owing to the fact that the contentious data in the above article had already been published elsewhere, or were already under consideration for publication, prior to its submission to International Journal of Oncology, the Editor has decided that this paper should be retracted from the Journal. The authors were asked for an explanation to account for these concerns, but the Editorial Office did not receive a reply. The Editor apologizes to the readership for any inconvenience caused. [International Journal of Oncology 55: 1110­1124, 2019; DOI: 10.3892/ijo.2019.4875].

4.
Cytokine ; 157: 155914, 2022 09.
Article in English | MEDLINE | ID: mdl-35809451

ABSTRACT

BACKGROUND: Bronchopulmonary dysplasia (BPD) is a common chronic lung disease in premature infants, and its pathogenesis has not been clarified. Long non-coding RNAs (lncRNA) have important functions in cell bioactivity. However, their role in developmental lung disease remains unclear. OBJECTIVE: The aim of this study was to demonstrate the role of lncRNA SNHG6 (SNHG6) in BPD and its underlying mechanisms. METHODS: The blood of patients with BPD were collected, and BPD model of BEAS-2B cells was established by hyperoxia method. SNHG6, miR-335 and KLF5 mRNA expression were detected by RT-qPCR. Western blot was conducted to measure the levels of apoptosis-related proteins' expression and NF-κB pathway related proteins. BEAS-2B cell viability and apoptosis were assessed by CCK-8 and flow cytometry, respectively. Assay Kit was applied to detect ROS, MDA and SOD levels, respectively. ELISA was performed to assess the levels of inflammatory factors. The binding site of miR-335 with SNHG6 or KLF5 were predicted by using DIANA or TargetScan, and which was verified by double luciferase reporter assay. RESULTS: Firstly, SNHG6 was highly expressed and miR-335 was lowly expressed in BPD model, SNHG6 knockdown and miR-335 mimics both alleviated hyperoxia-induced lung cell injury, and SNHG6 targeted miR-335. Subsequently, KLF5 was targeted by miR-335, and KLF5 promoted lung cell injury via activating NF-κB pathway. Furthermore, SNHG6 mediated lung cell injury via regulating the miR-335/KLF5/NF-κB pathway. CONCLUSION: Our research confirmed that SNHG6 mediated hyperoxia-induced lung cell injury via regulating the miR-335/KLF5/NF-κB pathway. These findings suggest that SNHG6 serves as promising targets for the treatment of newborns with BPD.


Subject(s)
Hyperoxia , Lung Diseases , MicroRNAs , RNA, Long Noncoding/genetics , Humans , Hyperoxia/genetics , Infant, Newborn , Kruppel-Like Transcription Factors/genetics , Lung/metabolism , MicroRNAs/genetics , MicroRNAs/metabolism , NF-kappa B , RNA, Long Noncoding/metabolism
5.
Ann Transl Med ; 9(20): 1577, 2021 Oct.
Article in English | MEDLINE | ID: mdl-34790783

ABSTRACT

BACKGROUND: Esophagus squamous cell carcinoma (ESCC) is a sort of cancer that occurs in the esophageal epithelial tissue. This study performed integrated bioinformatics analysis of Gene Expression Omnibus (GEO) datasets GSE32424, GSE29968, and GSE130078. Collagen type XI alpha 1 (COL11A1) was identified as the hub gene in ESCC progression. The involvement of COL11A1 in ESCC development was next determined using in vitro functional tests. METHODS: Hub genes were identified through integrated bioinformatics analysis. The real-time reverse transcription-polymerase chain reaction was implemented for detecting the expression of COL11A1 mRNA in esophageal cancer cells. KYSE-30 cells were transfected using a vector encoding COL11A1. The proliferation of cells was determined using the Cell Counting Kit-8 (CCK-8) assay. Detection of the cell migration and invasion was made through making use of the transwell test. The development of ESCC cells in vivo was evaluated in naked mice. The interplay among COL11A1 and microRNA-335-5p (miR-335-5p) was discovered using a luciferase reporter experiment. RESULTS: In vitro studies showed the upregulation of COL11A1 in ESCC cell lines obtained from ESCC patients and upregulation of COL11A1 was correlated with poor disease-free survival of ESCC patients, thereby implying an oncogenic involvement of COL11A1 in ESCC. Overexpression of COL11A1 enhanced the proliferation of ESCC cells, invasion, and migration; whereas COL11A1 knockdown impeded the proliferation of ESCC cells, invasion, and migration. Additionally, miRNA pathway analysis in combination with TargetScan's online prediction and the luciferase reporter assay suggested miR-335-5p targeting and negatively regulating the COL11A1 3' untranslated region (3'UTR) within ESCC cells. MiR-335-5p overexpression diminished the development of ESCC cells. Additionally, co-expression of COL11A1 ameliorated the repressive influence of miR-335-5p overexpression on the growth and metastasis of ESCC cells. CONCLUSIONS: Using comprehensive bioinformatics analysis, the current study identified COL11A1 as an oncogene in ESCC. The mechanistic studies indicated that COL11A1 promoted ESCC cell progression and that miR-335-5p negatively regulated the expression of COL11A1 in ESCC.

6.
Mol Med Rep ; 24(5)2021 11.
Article in English | MEDLINE | ID: mdl-34542164

ABSTRACT

Septicemia is associated with excessive inflammation, oxidative stress and apoptosis, causing myocardial injury that results in high mortality and disability rates worldwide. The abnormal expression of multiple microRNAs (miRNAs/miRs) is associated with more severe sepsis­induced myocardial injury (SIMI) and miR­335 has been shown to protect cardiomyocytes from oxidative stress. The present study aimed to investigate the role of miR­335 in SIMI. An SIMI model was established by cecal ligation and puncture (CLP) in mice. An miRNA­335 precursor (pre­miR­335) was transfected to accelerate miR­335 expression and an miR­335 inhibitor (anti­miR­335) was used to inhibit miR­335 expression. CLP or sham surgery was performed on pre­miR­335, anti­miR­335 and wild­type mice and miR­335 expression was determined by reverse transcription­quantitative PCR. Inflammatory factors (TNF­α, IL­6 and IL­10) and troponin (cTNI), brain natriuretic peptide (BNP), creatine kinase (CK), lactate dehydrogenase (LDH) and aspartate aminotransferase (AST) were assessed using commercial kits. Apoptosis was detected by flow cytometry and cardiac function was assessed using a Langendorff isolated cardiac perfusion system. miR­335 expression was upregulated and an elevation in inflammatory factors and cTNI, BNP, CK, LDH and AST was observed. Compared with the wild­type control group, pre­miR­335 mice treated with CLP exhibited significantly reduced left ventricular development pressure, maximum pressure increased reduction rates, as well as decreased levels of TNF­α, IL­6 and IL­10, myocardial injury and apoptosis; by contrast, these features were amplified in CLP­treated anti­miR­335 mice. In conclusion, the upregulation of miR­335 exerted ameliorative effects on myocardial injury following sepsis and may indicate a novel therapeutic intervention for SIMI.


Subject(s)
Disease Susceptibility , Gene Expression Regulation , Heart Diseases/diagnosis , Heart Diseases/etiology , MicroRNAs/genetics , Sepsis/complications , Animals , Apoptosis , Biomarkers , Cytokines/metabolism , Disease Models, Animal , Immunophenotyping , Inflammation Mediators , Mice , Sepsis/etiology
7.
Zhongguo Ying Yong Sheng Li Xue Za Zhi ; 37(4): 402-406, 2021 Jul.
Article in Chinese | MEDLINE | ID: mdl-34374261

ABSTRACT

Objective: To investigate the effects of miR-335-5p targeting glucose-6-phosphate dehydrogenase (G6PD) on the proliferation and apoptosis of colon cancer cells. Methods: Normal colon cell group, blank control group, NC group and miRNA-335-5p mimic group were set up. Colonic epithelial cells (IEC) and human colon cancer cells SW480 were cultured in vitro, and the cells in the NC group and miRNA-335-5p mimic group cells were transfected. RT-qPCR was used to detect the expression levels of miR-335-5p and G6PD mRNA in each group of cells. The targeting effect of miR-335-5p on G6PD was verified by Double Luciferase Report experiment. MTT assay was used to detect cell proliferation. Flow cytometry was used to detect the apoptosis rate. The expressions of G6PD, Bax, Bcl-2 and caspase-3 were detected by Western blot. Results: Compared with normal colon cells, the relative expression levels of miR-335-5p in SW480 cells of colon cancer in the blank control group and NC group were decreased, and the relative expression level of G6PD mRNA was increased (P<0.05); compared with the blank control group and NC group, the expression level of miR-335-5p in miR-335-5p mimic group was increased significantly, and the expression of G6PD mRNA was decreased significantly (P<0.05). Compared with the blank control group and NC group, the proliferative activity of colon cancer SW480 cells in miR-335-5p mimic group was decreased significantly, and the apoptosis rate was increased significantly (P<0.05). The relative activity of luciferase in miR-335-5p mimic + WT-G6PD 3 '- UTR group was lower than that in miR-335-5p NC + WT-G6PD 3' - UTR group (P<0.05). Compared with the blank control group, the relative expression levels of G6PD and bcl-2 protein in miR-335-5p mimic group were decreased significantly, and the expression levels of Bax and caspase-3 protein were increased significantly (P<0.05). Conclusion: MiR-335-5p may inhibit the proliferation and promote apoptosis of colon cancer cells by targeting G6PD.


Subject(s)
Colonic Neoplasms , MicroRNAs , Apoptosis , Cell Line, Tumor , Cell Proliferation , Colonic Neoplasms/genetics , Glucosephosphate Dehydrogenase/genetics , Humans , MicroRNAs/genetics
8.
Mol Med Rep ; 24(3)2021 Sep.
Article in English | MEDLINE | ID: mdl-34278455

ABSTRACT

Following the publication of this paper, it was drawn to the Editors' attention by a concerned reader that the cell cycle assay data shown in Fig. 4A, and the western blotting assay data shown in Fig. 4B, were strikingly similar to data appearing in different form in other articles by different authors; furthermore, there were other possible anomalies associated with these data. Owing to the fact that the contentious data in the above article had already been published elsewhere, or were already under consideration for publication, prior to its submission to Molecular Medicine Reports, the Editor has decided that this paper should be retracted from the Journal. The authors were asked for an explanation to account for these concerns, but the Editorial Office did not receive any reply. The Editor apologizes to the readership for any inconvenience caused. [the original article was published in Molecular Medicine Reports 11: 379­385, 2015; DOI: 10.3892/mmr.2014.2684].

9.
Mol Med Rep ; 24(3)2021 Sep.
Article in English | MEDLINE | ID: mdl-34278488

ABSTRACT

Following the publication of this paper, it was drawn to the Editors' attention by a concerned reader that the western blotting data shown in Fig. 1B were strikingly similar to data appearing in different form in other articles by different authors. Owing to the fact that the contentious data in the above article had already been published elsewhere, or were already under consideration for publication, prior to its submission to Molecular Medicine Reports, the Editor has decided that this paper should be retracted from the Journal. After having been in contact with the authors, they agreed with the decision to retract the paper. The Editor apologizes to the readership for any inconvenience caused. [the original article was published in Molecular Medicine Reports 10: 399­404, 2014; DOI: 10.3892/mmr.2014.2150].

10.
Mol Med Rep ; 24(2)2021 Aug.
Article in English | MEDLINE | ID: mdl-34080033

ABSTRACT

Current therapies for retinoblastoma (RB) are unsatisfactory and there is an urgent need for the development of new treatment modalities. Small nucleolar RNA host gene 20 (SNHG20) has been reported to serve a key oncogenic role in the development of various types of cancer, but its role in RB tumorigenesis remains to be fully determined. The present study aimed to investigate the expression patterns and biological roles of SNHG20 in RB. The expression levels of SNHG20 were measured via reverse transcription­quantitative PCR in RB tissues and cell lines. The impact of SNHG20 status on cell proliferation, survival, migration and invasion was determined using small interfering RNA and a range of established experimental assays. The SNHG20/microRNA (miR)­335­5p/E2F transcription factor 3 (E2F3) signaling axis was further investigated using a dual­luciferase activity reporter system and an RNA pull­down assay combined with bioinformatics analyses. SNHG20 expression was significantly increased in RB tissues and cell lines. Silencing of SNHG20 in RB cells was shown to inhibit cell proliferation, clonogenic survival, migration and invasion. Moreover, mechanistic investigations demonstrated that SNHG20 could enhance the expression of E2F3 by sponging of miR­335­5p. These data suggested that the long non­coding RNA SNHG20 may promote cell proliferation, migration and invasion in RB via the miR­335­5p/E2F3 axis.


Subject(s)
E2F3 Transcription Factor/metabolism , MicroRNAs/metabolism , RNA, Long Noncoding/genetics , Retinal Neoplasms/genetics , Retinal Neoplasms/metabolism , Retinoblastoma/genetics , Retinoblastoma/metabolism , Adult , Cell Line , Cell Movement/genetics , Cell Proliferation/genetics , Female , Gene Expression Regulation, Neoplastic/genetics , Gene Silencing , Humans , Male , MicroRNAs/genetics , RNA, Long Noncoding/metabolism , Signal Transduction/genetics , Young Adult
11.
Mol Med Rep ; 24(2)2021 Aug.
Article in English | MEDLINE | ID: mdl-34165173

ABSTRACT

Long non­coding (lnc)RNAs serve a role in a number of diseases, including different types of cancer and acute myocardial infarction. The aim of the present study was to investigate the protective role of lncRNA small nucleolar RNA host gene 8 (SNHG8) in hypoxia­ischemia­reoxygenation (HI/R)­induced myocardial injury and its potential mechanism of action. Cell viability, proliferation, creatine kinase myocardial band, cell apoptosis and protein expression levels were determined by Cell Counting Kit­8 assay, EdU assay, ELISA, flow cytometry and western blotting, respectively. The association between SNHG8 and microRNA (miR)­335 was confirmed using a dual­luciferase reporter gene assay. The effects of the miR­335 inhibitor transfections had on increasing apoptosis and decreasing H9C2 cell viability were reversed in cells co­transfected with SNHG8 small interfering (si)RNA. Furthermore, it was found that miR­335 could regulate RAS p21 protein activator 1 (RASA1) expression and that transfection with SNHG8 siRNA downregulated RASA1 expression. Silencing of RASA1 protected against HI/R­induced H9C2 cell injury. However, SNHG8 siRNA did not further reduce apoptosis, demonstrating that SNHG8 may act through RASA1, and RASA1 may mediate the protection of SNHG8 siRNA in HI/R myocardial injury. Thus, inhibition of lncRNA SNHG8 alleviated HI/R­induced myocardial damage by regulating miR­335 and RASA1.


Subject(s)
Hypoxia/metabolism , Ischemia/metabolism , MicroRNAs/metabolism , RNA, Long Noncoding/metabolism , p120 GTPase Activating Protein/metabolism , Animals , Apoptosis , Cell Hypoxia/genetics , Cell Line, Tumor , Cell Survival , Gene Expression Regulation, Neoplastic , Hypoxia/genetics , Hypoxia/therapy , Ischemia/genetics , Ischemia/therapy , MicroRNAs/genetics , Myocardial Infarction , Myocardial Ischemia/metabolism , Myocardial Ischemia/therapy , Myocytes, Cardiac/metabolism , RNA, Long Noncoding/genetics , RNA, Small Interfering/metabolism , Rats , p120 GTPase Activating Protein/genetics
12.
Stem Cell Res Ther ; 12(1): 169, 2021 03 10.
Article in English | MEDLINE | ID: mdl-33691791

ABSTRACT

BACKGROUND: New mechanistic insights into the self-renewal ability and multipotent properties of neural stem cells (NSCs) are currently under active investigation for potential use in the treatment of neurological diseases. In this study, NSCs were isolated from the forebrain of fetal rats and cultured to induce NSC differentiation, which was associated with low expression of the non-coding RNA microRNA-335-3p (miR-335-3p). METHODS: Loss- and gain-of-function experiments were performed in NSCs after induction of differentiation. RESULTS: Overexpression of miR-335-3p or FoxM1 and inhibition of the Fmr1 or p53 signaling pathways facilitated neurosphere formation, enhanced proliferation and cell cycle entry of NSCs, but restricted NSC differentiation. Mechanistically, FoxM1 positively regulated miR-335-3p by binding to its promoter region, while miR-335-3p targeted and negatively regulated Fmr1. Additionally, the promotive effect of miR-335-3p on NSC self-renewal occurred via p53 signaling pathway inactivation. CONCLUSION: Taken together, miR-335-3p activated by FoxM1 could suppress NSC differentiation and promote NSC self-renewal by inactivating the p53 signaling pathway via Fmr1.


Subject(s)
MicroRNAs , Neural Stem Cells , Animals , Cell Proliferation , Fragile X Mental Retardation Protein/genetics , MicroRNAs/genetics , Rats , Signal Transduction , Tumor Suppressor Protein p53/genetics
13.
Endocr J ; 68(6): 713-727, 2021 Jun 28.
Article in English | MEDLINE | ID: mdl-33716239

ABSTRACT

The circular RNA PRKCI (circ-PRKCI; ID: hsa_circ_0122683) is highly expressed in human papillary thyroid cancer (PTC) tumors according to GSE93522 dataset. However, its role in PTC tumorigenesis remains to be documented. Here, quantitative real-time PCR showed that expression of circ-PRKCI was abnormally upregulated in human PTC patients' tumors and cells, and higher circ-PRKCI might predict lymph node metastasis and recurrence. Functionally, cell behaviors were measured by 3-(4,5-dimethylthiazolyl-2)-2,5-diphenyltetrazolium bromide assay, colony formation assay, fluorescence-activated cell sorting method, scratch wound assay, transwell assay, western blotting, and assay kits for glucose and lactate. As a result, circ-PRKCI knockdown could suppress cell cycle progression of PTC cells and restrain the abilities of cell proliferation, colony formation, wound closure, invasion, glucose consumption and lactate production, accompanied with decreased levels of matrix metalloproteinase-2 (MMP2), MMP9 and Snail. Moreover, above-mentioned inhibition could be imitated by overexpressing microRNA-335-5p (miR-335). Molecularly, circ-PRKCI functioned as a sponge for miR-335 and miR-335 could further targeted E2F transcription factor-3 (E2F3), according to dual-luciferase reporter assay and RNA immunoprecipitation. However, downregulating miR-335 diminished the effects of circ-PRKCI role on cell growth, metastasis and glycolysis in PTC cells; besides, there was a counteractive effect between miR-335 upregulation and E2F3 upregulation in PTC cells as well. Furthermore, xenograft experiment revealed that silencing circ-PRKCI could retard tumor growth of PTC cells in vivo. Collectively, circ-PRKCI exerted oncogenic role in PTC by antagonizing cell progression and glycolysis via regulating miR-335/E2F3 axis, suggesting circ-PRKCI was a potential biomarker and target for PTC.


Subject(s)
Cell Cycle/genetics , Glycolysis/genetics , Isoenzymes/genetics , Protein Kinase C/genetics , RNA, Circular/genetics , Thyroid Cancer, Papillary/genetics , Thyroid Neoplasms/genetics , Adult , Cell Line, Tumor , Cell Proliferation/genetics , Female , Gene Expression Regulation, Neoplastic , Gene Silencing , Humans , Male , MicroRNAs/genetics , Middle Aged , Signal Transduction/genetics , Thyroid Cancer, Papillary/metabolism , Thyroid Cancer, Papillary/pathology , Thyroid Neoplasms/metabolism , Thyroid Neoplasms/pathology
14.
J Clin Lab Anal ; 35(2): e23608, 2021 Feb.
Article in English | MEDLINE | ID: mdl-33277957

ABSTRACT

BACKGROUND: Acute coronary syndrome (ACS) may induce cardiovascular death. The correlation of mast cells related microRNAs (miRs) with risk of ACS has been investigated. We explored regulatory mechanism of miR-335-5p on macrophage innate immune response, atherosclerotic vulnerable plaque formation, and revascularization in ACS in relation to Notch signaling. METHODS: ACS-related gene microarray was collected from Gene Expression Omnibus database. After different agomir or antagomir, or inhibitor of Notch signaling treatment, IL-6, IL-1ß, TNF-α, MCP-1, ICAM-1, and VCAM-1 levels were tested in ACS mice. Additionally, Notch signaling-related genes and matrix metalloproteinases (MMPs) were measured after miR-335-5p interference. Finally, mouse atherosclerosis, lipid accumulation, and the collagen/vessel area ratio of plaque were determined. RESULTS: miR-335-5p targeted JAG1 and mediated Notch signaling in ACS. miR-335-5p up-regulation and Notch signaling inhibition reduced expression of JAG1, Notch pathway-related genes, IL-6, IL-1ß, TNF-α, MCP-1, ICAM-1, VCAM-1, and MMPs, but promote TIMP1 and TIMP2 expression. Additionally, vulnerable plaques were decreased and collagen fiber contents were observed to increase after miR-335-5p overexpression and Notch signaling inhibition. CONCLUSIONS: Overexpression of miR-335-5p inhibited innate immune response of macrophage, reduced atherosclerotic vulnerable plaque formation, and promoted revascularization in ACS mice targeting JAG1 through Notch signaling.


Subject(s)
Acute Coronary Syndrome/genetics , MicroRNAs/genetics , Plaque, Atherosclerotic/genetics , Receptors, Notch/metabolism , 3' Untranslated Regions , Acute Coronary Syndrome/drug therapy , Acute Coronary Syndrome/physiopathology , Animals , Antagomirs/genetics , Antagomirs/pharmacology , Collagen/genetics , Collagen/metabolism , Diamines/pharmacology , Gene Expression , Humans , Jagged-1 Protein/genetics , Jagged-1 Protein/metabolism , Lipids/blood , Lipids/genetics , Male , Mice, Inbred C57BL , Plaque, Atherosclerotic/physiopathology , Receptors, Notch/genetics , Thiazoles/pharmacology
15.
Cell Signal ; 70: 109596, 2020 06.
Article in English | MEDLINE | ID: mdl-32156642

ABSTRACT

Chronic rhinosinusitis (CRS) is featured with chronic symptoms of inflammation or infection in the nasal and sinus tissues. MicroRNAs (miRNAs/miRs), such as dysregulated expression of miR-125b and miR-26a, has been previously demonstrated to be related to CRS. The present study is intended to define the role of miR-335-5p in inflammation and the related mechanism in a mouse model of CRS. The differentially expressed genes associated with CRS were screened by microarray analysis. The targeting relationship between miR-335-5p and TPX2 was analyzed by target prediction program and dual luciferase reporter gene assay. The mouse model of CRS was established, and mice were introduced with miR-335-5p mimics, miR-335-5p inhibitors, or siRNA against TPX2 to explore the regulatory functions of miR-335-5p. The regulatory effect of miR-335-5p on inflammation with the involvement of the AKT signaling pathway was also analyzed with the expression of inflammatory cytokines and AKT signaling pathway-related factors measured. It was indicated that miR-335-5p regulated the TPX2 gene-mediated AKT signaling pathway. TPX2 was identified as a target gene of miR-335-5p, and miR-335-5p elevation inhibited the activation of the AKT signaling pathway. In mice with CRS, up-regulation of miR-335-5p or silence of TPX2 inhibited the inflammation, as evidenced by decreased levels of TNF-α, IL-6 and IL-8, and higher levels of GSK3ß and IL-10. Collectively, miR-335-5p inhibits the activation of AKT signaling pathway by negatively mediating TPX2, which may confer anti-inflammatory protection in CRS.


Subject(s)
Cell Cycle Proteins/metabolism , Inflammation/metabolism , MicroRNAs/physiology , Microtubule-Associated Proteins/metabolism , Nuclear Proteins/metabolism , Rhinitis/metabolism , Sinusitis/metabolism , Animals , Chronic Disease , Glycogen Synthase Kinase 3 beta/metabolism , Mice , Mice, Inbred C57BL , Proto-Oncogene Proteins c-akt/metabolism , Signal Transduction
16.
Mol Med Rep ; 21(3): 1208-1216, 2020 03.
Article in English | MEDLINE | ID: mdl-31894323

ABSTRACT

Differential expression of microRNA (miR)­335­5p, a key tumor suppressor, has been detected in pre­eclampsia (PE) placentas. However, the role of miR­335­5p in the pathogenesis of PE and the factor modulating its aberrant expression remain unknown. The present study used JEG­3 cells in vitro to investigate these mechanisms. The role of miR­335­5p in proliferation, apoptosis and migration of JEG­3 cells was investigated using MTT, Annexin V­FITC/PI, Transwell migration and wound healing assays, respectively. miR­335­5p expression levels were analyzed using reverse transcription­quantitative PCR. The expression levels of E­cadherin, N­cadherin, Snail, specificity protein 1 (Sp1) and p53 were assessed using western blot analysis. Cell viability analysis was performed using the Cell Counting Kit­8 assay. The intracellular reactive oxygen species (ROS) levels were detected using a 2,7­dichlorodihydrofluorescein diacetate assay. The present results suggested that miR­335­5p did not affect the proliferation or apoptotic rate of JEG­3 cells. Overexpression of miR­335­5p significantly inhibited the migration of JEG­3 cells, decreased the expression levels of Sp1, N­cadherin and Snail, and increased E­cadherin expression. Sp1 silencing produced similar results in JEG­3 cells. H2O2 significantly increased the intracellular ROS levels and miR­335­5p expression, whereas N­acetyl­cysteine pretreatment prior to H2O2 treatment reversed the increases in miR­335­5p expression. Knockdown of p53 significantly decreased the expression levels of miR­335­5p in JEG­3 cells and in H2O2­treated cells. The present results suggested that miR­335­5p expression levels in trophoblast cells could be increased by ROS in a p53­dependent manner, leading to the downregulation of Sp1 and subsequent inhibition of epithelial to mesenchymal transition and cell migration. The present results may provide novel evidence on the etiology of PE.


Subject(s)
Epithelial-Mesenchymal Transition/genetics , MicroRNAs/genetics , Reactive Oxygen Species/metabolism , Transcription Factor AP-1/metabolism , Tumor Suppressor Protein p53/metabolism , Apoptosis/genetics , Cell Line, Tumor , Cell Movement/genetics , Humans , Oxidative Stress
17.
Am J Transl Res ; 12(12): 7899-7907, 2020.
Article in English | MEDLINE | ID: mdl-33437368

ABSTRACT

Several studies have proved the tumor-suppressive effects of miR-335 but its role in colon cancer via regulation of the Raf/MEK/ERK signalling pathway is yet unknown. As such the main motive of conducting the present study was to elucidate the role of miR-335 in colon cancer via regulation of Raf/MEK/ERK signalling pathway and to explore its therapeutic potential. The results revealed significant (P < 0.05) downregulation of miR-335 in colon cancer and its overexpression led to a significant (P < 0.05) decline in viability of the HT-29 and SW948 cells. The TUNNEL assay showed miR-335 promotes apoptosis in the HT-29 and SW948 colon cancer cells and is also associated with increase in Bax and decrease in Bcl-2 expression. The results also revealed that miR-335 overexpression enhances the sensitivity of the HT-29 and SW948 cells to the apoptotic effects of cisplatin. From the transwell assays, it was found that the migration of the HT-29 and SW948 cells was decreased by 53% and 45% and while as invasion was decreased by 49% and 42% respectively (P < 0.05). Finally, western blot analysis showed that miR-335 blocks the Raf/MEK/ERK signalling pathway in HT-29 colon cancer cells. The results of in vivo study showed that miR-335 also exhibits tumor-suppressive effects on xenografted tumors. Taken together, it is concluded that miR-335 acts as tumor-suppressor in colon cancer and may exhibit therapeutic implications in its treatment.

18.
Am J Physiol Gastrointest Liver Physiol ; 318(1): G10-G22, 2020 01 01.
Article in English | MEDLINE | ID: mdl-31433213

ABSTRACT

Gastric cancer (GC) is one of the most common cancers in the world and remains a heavy burden of health worldwide. Adenylate cyclase 3 (ADCY3) is a widely expressed membrane-associated protein in human tissues and has been identified to be a new molecular target of GC. Long noncoding RNAs have a substantial influence on tumorigenesis and progression of tumors by binding to microRNAs. Therefore, this study is to clarify the mechanism by which LINC00319 sponges micro RNA-335-5p (miR-335-5p) to influence the development of GC. Initially, microarray analysis identified GC-related differentially expressed LINC00319 and ADCY3 for this study. The interaction was confirmed that LINC00319 interacted with miR-335-5p to regulate ADCY3. Next, SGC-7901 cells presenting with the lowest LINC00319 expression and the highest miR-335-5p expression were transfected with LINC00319, miR-335-5p inhibitor, or ADCY3 vector to examine their roles in growth and metastasis of GC cells, which was further ascertained by in vivo experiments. LINC00319 was upregulated and miR-335-5p was downregulated in GC cells. LINC00319 overexpression, miR-335-5p inhibitor, or ADCY3 overexpression was shown to significantly elevate the expression of cyclin-dependent kinase 4 and metastasis associated 1, decrease that of growth arrest-specific 1, and promote tumor growth and metastasis by increasing proliferation and migration and reducing cell apoptosis. Importantly, it was found that overexpressed miR-335-5p exerted its tumor suppressive role in GC through downregulating ADCY3. Collectively, LINC00319 expedited growth and metastasis of GC by upregulating miR-335-5p-mediated ADCY3.NEW & NOTEWORTHY This study is carried out based on in vivo and in vitro studies in mice and gastric cancer (GC) cells with the aim of clarifying the role of LINC00319 on GC growth and metastasis, which associated with micro RNA-335-5p-mediated adenylate cyclase 3. Altogether, we identified LINC00319 to be a potential therapy to treat GC.


Subject(s)
Adenylyl Cyclases/metabolism , Cell Movement , Cell Proliferation , MicroRNAs/metabolism , RNA, Long Noncoding/metabolism , Stomach Neoplasms/enzymology , 3' Untranslated Regions , Adenylyl Cyclases/genetics , Animals , Apoptosis , Binding Sites , Cell Line, Tumor , Cyclin-Dependent Kinase 4/genetics , Cyclin-Dependent Kinase 4/metabolism , Databases, Genetic , Gene Expression Regulation, Enzymologic , Gene Expression Regulation, Neoplastic , Humans , Male , Mice, Nude , MicroRNAs/genetics , Neoplasm Metastasis , RNA, Long Noncoding/genetics , Repressor Proteins/genetics , Repressor Proteins/metabolism , Signal Transduction , Stomach Neoplasms/genetics , Stomach Neoplasms/pathology , Trans-Activators/genetics , Trans-Activators/metabolism , Tumor Burden , Up-Regulation
19.
J Cell Physiol ; 234(12): 21937-21946, 2019 12.
Article in English | MEDLINE | ID: mdl-31054187

ABSTRACT

Accumulating evidence suggests that noncoding RNAs play a vital role in cancer biology. Circular RNAs (circRNAs), a newly defined class of endogenously widespread noncoding RNAs, have been intensively reported to influence cell function and development, and even cancer prognosis by sponging microRNAs in various types of cancer. Nevertheless, the circRNAs research in hepatocellular carcinoma (HCC) still remains far insufficient. Herein, we investigated the role of a newly defined circRNAs, circ_0005075, in HCC development. We found circ_0005075 was upregulated in HCC tissues. HCC progression was suppressed by downregulation of circ_0005075 in vitro and in vivo, and the suppression was partially reversed by inhibition of microRNA-335 (miR-335) expression. Further, we found the expression of mitogen-activated protein kinase 1 (MAPK1) was substantially regulated by circ_0005075 and miR-335. Mechanically, it was demonstrated that circ_0005075 could directly bind to miR-335 and miR-335 could bind to MAPK1. Our data provide evidence that circ_0005705 promotes the HCC progression by sponging miR-335 and further regulating MAPK1 expression.


Subject(s)
Carcinoma, Hepatocellular/genetics , Gene Expression Regulation, Neoplastic/genetics , Liver Neoplasms/genetics , MicroRNAs/genetics , Mitogen-Activated Protein Kinase 1/biosynthesis , RNA, Circular/genetics , Animals , Disease Progression , Heterografts , Humans , Liver Neoplasms/pathology , Mice , Mitogen-Activated Protein Kinase 1/genetics
20.
J Cell Physiol ; 234(5): 6654-6666, 2019 05.
Article in English | MEDLINE | ID: mdl-30341900

ABSTRACT

Multiple studies have reported different methods in treating gestational diabetes mellitus (GDM); however, the relationship between miR-335-5p and GDM still remains unclear. Here, this study explores the effect of miR-335-5p on insulin resistance and pancreatic islet ß-cell secretion via activation of the TGFß signaling pathway by downregulating VASH1 expression in GDM mice. The GDM mouse model was established and mainly treated with miR-335-5p mimic, miR-335-5p inhibitor, si-VASH1, and miR-335-5p inhibitor + si-VASH1. Oral glucose tolerance test (OGTT) was conducted to detect fasting blood glucose (FBG) fasting insulin (FINS). The OGTT was also used to calculate a homeostasis model assessment of insulin resistance (HOMA-IR). A hyperglycemic clamp was performed to measure the glucose infusion rate (GIR), which estimated ß-cell function. Expressions of miR-335-5p, VASH1, TGF-ß1, and c-Myc in pancreatic islet ß-cells were determined by RT-qPCR, western blot analysis, and insulin release by ELISA. The miR-335-5p mimic and si-VASH1 groups showed elevated blood glucose levels, glucose area under the curve (GAUC), and HOMA-IR, but a reduced GIR and positive expression of VASH1. Overexpression of miR-335-5p and inhibition of VASH1 contributed to activated TGFß1 pathway, higher c-Myc, and lower VASH1 expressions, in addition to downregulated insulin and insulin release levels. These findings provided evidence that miR-335-5p enhanced insulin resistance and suppressed pancreatic islet ß-cell secretion by inhibiting VASH1, eventually activating the TGF-ß pathway in GDM mice, which provides more clinical insight on the GDM treatment.


Subject(s)
Blood Glucose/genetics , Diabetes, Gestational/genetics , Insulin Resistance/genetics , MicroRNAs/genetics , Transforming Growth Factor beta/deficiency , Animals , Blood Glucose/metabolism , Cell Cycle Proteins/genetics , Cell Cycle Proteins/metabolism , Female , Glucose Tolerance Test/methods , Insulin/metabolism , Insulin Resistance/physiology , Insulin-Secreting Cells/metabolism , Male , Mice , Pregnancy , Transforming Growth Factor beta/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...