Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 66
Filter
Add more filters

Country/Region as subject
Publication year range
1.
Int J Mol Sci ; 25(6)2024 Mar 14.
Article in English | MEDLINE | ID: mdl-38542261

ABSTRACT

MicroRNAs (miRNA) are involved in the process of carcinogenesis, including the development of endometrial cancer (EC). This study aimed to investigate the association between the expression of three miRNAs (miR-21-5p, miR-205-5p, and miR-222-3p) in endometrial cancer tissues. In addition, the stability of expression of SNORD48 and U6, which were initially planned to be used as reference miRNAs for normalization, was investigated. Endometrial tissue was obtained from 111 patients with EC during hysterectomy and from 19 patients undergoing surgery for uterine fibroids or pelvic organ prolapse as a control group without neoplastic changes. Our study was based on calculations made with a digital PCR method (Qiagen, Hilden, Germany) to measure the absolute expression. In the endometrial cancer tissue, miR-205-5p was upregulated, while miR-222-3p and SNORD48 were downregulated compared to the control group. We detected statistically significant correlation of miR-205-5p, U6, and SNORD48 expression with different histological grades; the expression of miR-205-5p increases with the histopathological grade advancement (intraepithelial neoplasia- EIN = 1590, G1 = 3367.2, G2 = 8067 and G3 = 20,360), while U6 and SNORD expression decreases from EIN to G2 and increases again in the G3 grade (U6: EIN = 19,032, G1 = 16,482.4, G2 = 13,642.4, G3 = 133,008; SNORD48: EIN = 97,088, G1 = 59,520, G2 = 43,544, G3 = 227,200). Our study suggests that upregulation of miR-205-5p and downregulation of miR-222-3p and SNORD48 may influence development of endometrial cancer. Moreover, miR-205-5p, U6, and SNORD48 expression changes may be associated with progression of endometrial cancer. The results also indicate that SNORD48 and U6, commonly used as internal references, may influence endometrial cancer development and progression; therefore, they should not be used as references. However, it is important to note that further research is required to understand their role in endometrial cancer.


Subject(s)
Endometrial Neoplasms , MicroRNAs , Female , Humans , Gene Expression Regulation, Neoplastic , MicroRNAs/genetics , MicroRNAs/metabolism , Endometrial Neoplasms/genetics , Down-Regulation/genetics , Polymerase Chain Reaction
2.
J Cell Biochem ; 124(9): 1379-1390, 2023 09.
Article in English | MEDLINE | ID: mdl-37565526

ABSTRACT

Numerous studies have revealed the profound impact of microRNAs on regulating skeletal muscle development and regeneration. However, the biological function and regulation mechanism of miR-222-3p in skeletal muscle remains largely unknown. In this study, miR-222-3p was found to be abundantly expressed in the impaired skeletal muscles, indicating that it might have function in the development and regeneration process of the skeletal muscle. MiR-222-3p overexpression impeded C2C12 myoblast proliferation and myogenic differentiation, whereas inhibition of miR-222-3p got the opposite results. The dual-luciferase reporter assay showed that insulin receptor substrate-1 (IRS-1) was the target gene of miR-222-3p. We next found that knockdown of IRS-1 could obviously suppress C2C12 myoblast proliferation and differentiation. Additionally, miR-222-3p-induced repression of myoblast proliferation and differentiation was verified to be associated with a decrease in phosphoinositide 3-kinase (PI3K)-Akt signaling. Overall, we demonstrated that miR-222-3p inhibited C2C12 cells myogenesis via IRS-1/PI3K/Akt pathway. Therefore, miR-222-3p may be used as a therapeutic target for alleviating muscle loss caused by inherited and nonhereditary diseases.


Subject(s)
MicroRNAs , Proto-Oncogene Proteins c-akt , Animals , Mice , Cell Differentiation/genetics , Cell Proliferation/genetics , Insulin Receptor Substrate Proteins/genetics , Insulin Receptor Substrate Proteins/metabolism , MicroRNAs/metabolism , Muscle Development/genetics , Myoblasts/metabolism , Phosphatidylinositol 3-Kinase/metabolism , Phosphatidylinositol 3-Kinases/genetics , Phosphatidylinositol 3-Kinases/metabolism , Proto-Oncogene Proteins c-akt/genetics , Proto-Oncogene Proteins c-akt/metabolism
3.
J Neuroinflammation ; 20(1): 156, 2023 Jun 30.
Article in English | MEDLINE | ID: mdl-37391774

ABSTRACT

BACKGROUND: Macrophage in the spinal cord injury (SCI) area imparts a chronic pro-inflammation effect that challenges the recovery of SCI. Previously, endothelial progenitor cell-produced exosomes (EPC-EXOs) have been noticed to facilitate revascularization and inflammation control after SCI. However, their effects on macrophage polarization remained unclear. This study aimed to investigate the EPC-EXOs' role in macrophage polarization and reveal its underlying mechanism. METHODS: We extracted the macrophages and EPC from the bone marrow suspension of C57BL/L mice by centrifugation. After cell identification, the EPC-EXOs were collected by ultra-high-speed centrifugation and exosome extraction kits and identified by transmission electron microscopy and nanoparticle tracking analysis. Then, macrophages were cultured with EPC-EXOs in different concentrations. We labeled the exosome to confirm its internalization by macrophage and detected the macrophage polarization marker level both in vitro and in vivo. We further estimated EPC-EXOs' protective effects on SCI by mice spinal cord tissue H&E staining and motor behavior evaluation. Finally, we performed RT-qPCR to identify the upregulated miRNA in EPC-EXOs and manipulate its expression to estimate its role in macrophage polarization, SOCS3/JAK2/STAT3 pathway activation, and motor behavior improvement. RESULTS: We found that EPC-EXOs decreased the macrophages' pro-inflammatory marker expression and increased their anti-inflammatory marker expression on the 7 and 14 days after SCI. The spinal cord H&E staining results showed that EPC-EXOs raised the tissue-sparing area rate significantly after 28 days of SCI and the motor behavior evaluation indicated an increased BMS score and motor-evoked potential by EPC-EXOs treatment after SCI. The RT-qPCR assay identified that miR-222-3P upregulated in EPC-EXOs and its miRNA-mimic also decreased the pro-inflammatory macrophages and increased the anti-inflammatory macrophages. Additionally, miR-222-3P mimic activated the SOCS3/JAK2/STAT3 pathway, and SOCS3/JAK2/STAT3 pathway inhibition blocked miR-2223P's effects on macrophage polarization and mouse motor behavior. CONCLUSION: Comprehensively, we discovered that EPC-EXOs-derived miR-222-3p affected macrophage polarization via SOCS3/JAK2/STAT3 pathway and promoted mouse functional repair after SCI, which reveals EPC-EXOs' role in modulation of macrophage phenotype and will provide a novel interventional strategy to induce post-SCI recovery.


Subject(s)
Endothelial Progenitor Cells , Exosomes , MicroRNAs , Spinal Cord Injuries , Animals , Mice , Mice, Inbred C57BL , Anti-Inflammatory Agents , Spinal Cord Injuries/therapy , Inflammation , Macrophages , MicroRNAs/genetics
4.
Mol Carcinog ; 62(12): 1817-1831, 2023 Dec.
Article in English | MEDLINE | ID: mdl-37606187

ABSTRACT

The atypical cadherin FAT1 function either as a pro or antitumorigenic in tumors of different tissue origins. Our group previously demonstrated the protumorigenic nature of FAT1 signaling in glioblastoma (GBM). In this study, we investigated how FAT1 influences the expression of clustered oncomiRs (miR-221-3p/miR-222-3p) and their downstream effects in GBM. Through several experiments involving the measurement of specific gene/microRNA expression, gene knockdowns, protein and cellular assays, we have demonstrated a novel oncogenic signaling pathway mediated by FAT1 in glioma. These results have been verified using antimiRs and miR-mimic assays. Initially, in glioma-derived cell lines (U87MG and LN229), we observed FAT1 as a novel up-regulator of the transcription factor NFκB-RelA. RelA then promotes the expression of the clustered-oncomiRs, miR-221-3p/miR-222-3p, which in turn suppresses the expression of the tumor suppressor gene (TSG), PDCD10 (Programmed cell death protein10). The suppression of PDCD10, and other known TSG targets (PTEN/PUMA), by miR-221-3p/miR-222-3p, leads to increased clonogenicity, migration, and invasion of glioma cells. Consistent with our in-vitro findings, we observed a positive expression correlation of FAT1 and miR-221-3p, and an inverse correlation of FAT1 and the miR-targets (PDCD10/PTEN/PUMA), in GBM tissue-samples. These findings were also supported by publicly available GBM databases (The Cancer Genome Atlas [TCGA] and The Repository of Molecular Brain Neoplasia Data [Rembrandt]). Patients with tumors displaying high levels of FAT1 and miR-221-3p expression (50% and 65% respectively) experienced shorter overall survival. Similar results were observed in the TCGA-GBM database. Thus, our findings show a novel FAT1/RelA/miR-221/miR-222 oncogenic-effector pathway that downregulates the TSG, PDCD10, in GBM, which could be targeted therapeutically in a specific manner.


Subject(s)
Brain Neoplasms , Glioblastoma , Glioma , MicroRNAs , Humans , Glioblastoma/metabolism , Cadherins/genetics , Cadherins/metabolism , Apoptosis Regulatory Proteins/genetics , Apoptosis Regulatory Proteins/metabolism , Glioma/genetics , Brain Neoplasms/genetics , Brain Neoplasms/metabolism , MicroRNAs/genetics , MicroRNAs/metabolism , Cell Line, Tumor , Gene Expression Regulation, Neoplastic , Cell Proliferation/genetics , Cell Movement/genetics , Membrane Proteins/genetics , Proto-Oncogene Proteins/genetics
5.
J Biochem Mol Toxicol ; 37(11): e23475, 2023 Nov.
Article in English | MEDLINE | ID: mdl-37462336

ABSTRACT

Ankylosing spondylitis (AS) is a chronic rheumatic disease, and some microRNAs (miRNAs) in AS have been identified. This study aimed to measure miR-222-3p expression in AS patients, investigate the association of miR-222-3p with AS disease activity, and explore the clinical value of miR-222-3p in diagnosing AS and predicting therapeutic efficacy of nonsteroidal anti-inflammatory drugs (NSAIDs) on AS patients. This study included 96 patients with AS, 58 patients with rheumatoid arthritis (RA), and 90 healthy controls. miR-222-3p expression was detected by reverse-transcription quantitative polymerase chain reaction (PCR). The ability of miR-222-3p to discriminate between different groups was evaluated by receiver operating characteristic analysis. The predictive value of miR-222-3p on the efficacy of NSAID treatment for AS was assessed by logistic regression analysis. AS patients treated with oral NSAIDs diclofenac sodium were divided into response (n = 76) and no-response (n = 20) groups after 16 weeks of treatment. miR-222-3p in AS patients was higher than that in healthy subjects and RA patients. miR-222-3p had high diagnostic value in distinguishing patients with AS from RA patients and healthy controls. miR-222-3p, increased in active AS patients, had the ability to screen active AS patients from inactive AS patients. miR-222-3p was decreased in the response group, and had high accuracy in predicting the therapeutic efficiency of NSAIDs. The findings indicate that increased miR-222-3p in AS patients may function as a diagnostic biomarker for AS, and predictive biomarker for the therapeutic efficacy of NSAIDs in patients with AS. In addition, miR-222-3p is associated with AS disease activity.


Subject(s)
Arthritis, Rheumatoid , MicroRNAs , Spondylitis, Ankylosing , Humans , Spondylitis, Ankylosing/drug therapy , Spondylitis, Ankylosing/genetics , MicroRNAs/genetics , MicroRNAs/metabolism , Arthritis, Rheumatoid/drug therapy , Anti-Inflammatory Agents, Non-Steroidal/therapeutic use , Biomarkers/metabolism
6.
J Nanobiotechnology ; 21(1): 128, 2023 Apr 12.
Article in English | MEDLINE | ID: mdl-37046252

ABSTRACT

Chronic non-healing wounds, a prevalent complication of diabetes, are associated with increased mortality in diabetic patients. Excessive accumulation of M1 macrophages in diabetic wounds promotes inflammation and results in dysregulated tissue repair. Adipose tissue macrophages (ATMs) derived from healthy lean donors have the ability to improve glucose tolerance and insulin sensitivity, as well as modulate inflammation. MicroRNAs (miRs), which can be packaged into exosomes (Exos) and secreted from cells, serve as essential regulators of macrophage polarization. Here, we revealed that ATMs isolated from lean mice secrete miRs-containing Exos, which modulate macrophage polarization and promote rapid diabetic wound healing when administered to diabetes-prone db/db mice. The miRs sequence of tissue samples from wounds treated with Exos secreted by lean ATMs (ExosLean) revealed that miR-222-3p was up-regulated. Further analyses showed that inhibiting miR-222-3p using a miR inhibitor impaired the macrophage-reprogramming effect of ExosLean. In the excisional skin wound mouse model, locally inhibiting miR-222-3p disrupted healing dynamics and failed to modulate macrophage polarization. Mechanistic studies revealed a connection between miR-222-3p, Bcl2l11/Bim, an inflammatory response effector, macrophage polarization, and diabetic wound healing. In summary, ExosLean act as positive regulators of macrophage polarization by regulating miR levels in wounds and accelerating wound healing, and thus have important implications for wound management in diabetes.


Subject(s)
Diabetes Mellitus , Exosomes , MicroRNAs , Mice , Animals , Adipose Tissue , MicroRNAs/genetics , MicroRNAs/pharmacology , Inflammation , Macrophages , Wound Healing
7.
Cell Mol Life Sci ; 79(5): 254, 2022 Apr 22.
Article in English | MEDLINE | ID: mdl-35451651

ABSTRACT

Previous studies in our laboratory have reported that miR-222-3p was a tumor-suppressive miRNA in OC. This study aims to further understand the regulatory role of miR-222-3p in OC and provide a new mechanism for its prevention and treatment. We first found that miR-222-3p inhibited the migration and proliferation of OC cells. Then, we observed CDK19 was highly expressed in OC and inversely correlated with miR-222-3p. Besides, we observed that miR-222-3p directly binds to the 3'-UTR of CDK19 and inhibits CDK19 translation, thus inhibiting OC cell migration and proliferation in vitro and repressed tumor growth in vivo. We also observed the inhibitory effect of Hotair on miR-222-3p in OC. In addition, Hotair could promote the proliferation and migration of OC cells in vitro and facilitate the growth and metastasis of tumors in vivo. Moreover, Hotair was positively correlated with CDK19 expression. These results suggest Hotair indirectly up-regulates CDK19 through sponging miR-222-3p, which enhances the malignant behavior of OC. This provides a further understanding of the mechanism of the occurrence and development of OC.


Subject(s)
Cyclin-Dependent Kinases , MicroRNAs , Ovarian Neoplasms , RNA, Long Noncoding , 3' Untranslated Regions , Cell Line, Tumor , Cell Proliferation/genetics , Cyclin-Dependent Kinases/genetics , Female , Humans , MicroRNAs/genetics , Ovarian Neoplasms/genetics , RNA, Long Noncoding/genetics
8.
Int J Mol Sci ; 24(24)2023 Dec 13.
Article in English | MEDLINE | ID: mdl-38139277

ABSTRACT

The progression of obesity and type 2 diabetes (T2D) is intricately linked with adipose tissue (AT) angiogenesis. Despite an established network of microRNAs (miRNAs) regulating AT function, the specific role of angiogenic miRNAs remains less understood. The miR-221/222 cluster has recently emerged as being associated with antiangiogenic activity. However, no studies have explored its role in human AT amidst the concurrent development of obesity and T2D. Therefore, this study aims to investigate the association between the miR-221-3p/222-3p cluster in human AT and its regulatory network with obesity and T2D. MiR-221-3p/222-3p and their target gene (TG) expression levels were quantified through qPCR in visceral (VAT) and subcutaneous (SAT) AT from patients (n = 33) categorized based on BMI as normoweight (NW) and obese (OB) and by glycemic status as normoglycemic (NG) and type 2 diabetic (T2D) subjects. In silico analyses of miR-221-3p/222-3p and their TGs were conducted to identify pertinent signaling pathways. The results of a multivariate analysis, considering the simultaneous expression of miR-221-3p and miR-222-3p as dependent variables, revealed statistically significant distinctions when accounting for variables such as tissue depot, obesity, sex, and T2D as independent factors. Furthermore, both miRNAs and their TGs exhibited differential expression patterns based on obesity severity, glycemic status, sex, and type of AT depot. Our in silico analysis indicated that miR-221-3p/222-3p cluster TGs predominantly participate in angiogenesis, WNT signaling, and apoptosis pathways. In conclusion, these findings underscore a promising avenue for future research, emphasizing the miR-221-3p/222-3p cluster and its associated regulatory networks as potential targets for addressing obesity and related metabolic disorders.


Subject(s)
Diabetes Mellitus, Type 2 , MicroRNAs , Humans , Diabetes Mellitus, Type 2/metabolism , MicroRNAs/genetics , MicroRNAs/metabolism , Obesity/metabolism , Adipose Tissue/metabolism
9.
Int Ophthalmol ; 43(5): 1611-1628, 2023 May.
Article in English | MEDLINE | ID: mdl-36319884

ABSTRACT

BACKGROUND: Age-related cataract (ARC) is a leading cause of blindness worldwide with multiple pathogenic factors. Oxidative damage of lens epithelium cells (LECs) is one of the well-accepted pathogenesis of ARC which can be regulated by DNA repair genes (DRGs). The present research aimed to clarify the regulatory mechanism of exosomal microRNAs (miRNAs) on DRGs in LECs. METHODS: The LECs oxidative damage model was established by UVB-irradiation on SRA01/04 (human lens epithelium cell line). Exosomes from UVB-irradiated cells (UVB-exo) and exosomes from normal control cells (NC-exo) were collected from the culture medium. To explore the functions of LECs exosomes, SRA01/04 were incubated with UVB-exo/NC-exo. Then, we detected SRA01/04 proliferation, viability and apoptosis respectively using 5'-ethynyl-2'-deoxyuridine (EdU), cell-counting kit-8 (CCK-8) and TdT-mediated dUTP Nick-End Labeling (TUNEL) assay. Next, the miRNA expression profiles of UVB-exo and NC-exo were identified by miRNA microarrays. RNA expression in exosomes, cells, and clinical samples was verified by qRT-PCR. The location and expression of MGMT and CD63 proteins were detected by immunofluorescence and western blot. The 3'UTR regulation of miR-222-3p to MGMT was verified by luciferase analyses. RESULTS: MGMT down-regulated while miR-222-3p up-regulated in LECs sub-central anterior capsule from ARC lenses. MGMT and miR-222-3p expressions in central and peripheral LECs from anterior lens capsules were differential. UVB-exo can transport the up-regulated miR-222-3p from oxidative-damaged LECs to normal LECs, which could suppress MGMT expression and increase UVB sensitivity of LECs. CONCLUSIONS: Findings on exosomal miRNA functions provided novel insights into pathogenesis of ARC. Exosomal miR-222-3p can be a potential target for prevention and cure of ARC.


Subject(s)
Cataract , Lens, Crystalline , MicroRNAs , Humans , Cataract/metabolism , Cell Proliferation , DNA Modification Methylases/metabolism , DNA Repair Enzymes/genetics , DNA Repair Enzymes/metabolism , Epithelial Cells/pathology , Epithelium/pathology , Lens, Crystalline/pathology , MicroRNAs/genetics , MicroRNAs/metabolism , Tumor Suppressor Proteins/genetics , Ultraviolet Rays
10.
J Gene Med ; 24(12): e3459, 2022 12.
Article in English | MEDLINE | ID: mdl-36279183

ABSTRACT

BACKGROUND: Although studies suggest that miR222-3p is dysregulated in prostate cancer (PC) cells and tissues, the possible changes in the level of miR222-3p in the plasma samples of PC patients remained unclear. The present study aimed to evaluate the diagnostic value of the plasma miR222-3p expression level as a potential biomarker in PC, benign prostatic hyperplasia (BPH) and healthy people. METHODS: Blood samples were collected from 100 adult males (54 patients with PC, 27 patients with BPH and 19 healthy individuals) referred to our affiliated hospital. The expression level of miR222-3p was evaluated using a quantitative reverse transcription-polymerase chain reaction. Receiver operating characteristic curves were used to evaluate miR222-3p diagnostic accuracy for discriminating between the PC, BPH and healthy individuals. RESULTS: The expression level of miR222-3p was significantly higher in PC patients compared to healthy individuals as a fold change of 5.3 (p = 0.009), but not for BPH individuals. The diagnostic value of the plasma miR222-3p for discrimination of the PC patients from healthy individuals was reasonable [cut-off value (fold change relative to miR16-5p) = 1.69, area under the curve = 0.73, sensitivity = 0.75 and specificity = 0.74]. CONCLUSIONS: Circulating plasma miR-222-3p significantly upregulated in PC patients, but not in BPH ones. Besides these preliminary results showed that miR222-3p has the potential to discriminate PC patients from healthy ones. Addittional studies with a larger sample size are required to confirm these data.


Subject(s)
MicroRNAs , Prostatic Neoplasms , Humans , Male , Prostatic Neoplasms/diagnosis , Prostatic Neoplasms/genetics , MicroRNAs/genetics
11.
BMC Womens Health ; 22(1): 398, 2022 10 01.
Article in English | MEDLINE | ID: mdl-36182923

ABSTRACT

OBJECTIVE: microRNAs (miRNAs) play pivotal roles in polycystic ovary syndrome (PCOS), an endocrine and metabolic disorder that commonly occurs in women of childbearing age. This paper aimed to measure miR-222-3p expression in sera of PCOS patients and to explore its clinical value on PCOS diagnosis and prediction of diabetic and cardiovascular complications. METHODS: Totally 111 PCOS patients and 94 healthy people were recruited and assigned to the overweight (ow) group and non-overweight (non-ow) group, followed by determination of serum miR-222-3p expression. The diagnostic efficiency of miR-222-3p on PCOS ow and non-ow patients was analyzed. Correlations between miR-222-3p and glycolipid metabolic indicators and diabetic and cardiovascular complications in PCOS were analyzed. The downstream target of miR-222-3p was predicted and their binding relationship was verified. The correlation between PGC-1α and miR-222-3p was analyzed. RESULTS: miR-222-3p was highly-expressed in PCOS patients (p < 0.001), in especially PCOS ow patients. The area under the curve (AUC) of miR-222-3p diagnosing PCOS non-ow patients was 0.9474 and cut-off value was 1.290 (89.06% sensitivity, 98.11% specificity), indicating that non-ow people with serum miR-222-3p > 1.290 could basically be diagnosed with PCOS. AUC of miR-222-3p diagnosing PCOS ow patients was 0.9647 and cut-off value was 2.425 (85.11% sensitivity, 100% specificity), suggesting that ow people with serum miR-222-3p > 2.425 could basically be diagnosed with PCOS. miR-222-3p was positively-correlated with fasting plasma glucose (FPG), fasting insulin (FINS), homeostatic model assessment-insulin resistance (HOMA-IR), and low-density lipoprotein cholesterol (LDL-C) and negatively-correlated with high-density lipoprotein cholesterol (HDL-C). miR-222-3p was independently-correlated with diabetic and cardiovascular complications in PCOS (p < 0.05). High expression of miR-222-3p predicted high risks of diabetic and cardiovascular complications in PCOS. miR-222-3p targeted PGC-1α and was negatively-correlated with PGC-1α (r = - 0.2851, p = 0.0224; r = - 0.3151, p = 0.0310). CONCLUSION: High expression of miR-222-3p assisted PCOS diagnosis and predicted increased risks of diabetic and cardiovascular complications. miR-222-3p targeted PGC-1α and was negatively-correlated with PGC-1α.


Subject(s)
Insulin Resistance , MicroRNAs , Polycystic Ovary Syndrome , Blood Glucose , Cholesterol, HDL , Cholesterol, LDL/metabolism , Correlation of Data , Female , Glucose , Glycolipids , Humans , Insulin/metabolism , Lipid Metabolism , Obesity/complications , Overweight/complications , Polycystic Ovary Syndrome/complications
12.
Immunopharmacol Immunotoxicol ; 44(4): 548-555, 2022 Aug.
Article in English | MEDLINE | ID: mdl-35379056

ABSTRACT

BACKGROUND: Medulloblastoma (MB) is an embryonal tumor of the cerebellum, which commonly occurs in childhood. Herein, we investigated the effects of miR-222-3p on the formation of MB stem-like cells via the Notch2/c-myc pathway. METHODS: Quantitative real-time PCR (qRT-PCR) or western blotting was performed to determine the expression of miR-222-3p and Notch2, c-myc, proliferating cell nuclear antigen (PCNA), and caspase-3. Luciferase reporter gene, RNA immunoprecipitation (RIP), and RNA pull-down assay were applied to confirm the interaction between miR-222-3p and Notch2. Cell growth was examined by Cell Counting Kit-8. Cell cycle distribution and the number of stem cell marker CD133+ cells were examined using flow cytometry. The sphere formation assay was performed. RESULTS: miR-222-3p expression was decreased and Notch2 expression was increased in human medulloblastoma cells. miR-222-3p overexpression inhibited cell viability and the sphere formation, induced cell cycle arrest, decreased the number of CD133+ cells, and up-regulated caspase-3 expression and down-regulated PCNA, Notch2, and c-myc expression. However, Notch2 overexpression counteracted these effects of miR-222-3p overexpression. Simultaneous overexpression of Notch2 and miR-222-3p increased the c-myc promoter-luciferase activity which was decreased by miR-222-3p overexpression. Luciferase reporter gene, RIP, and RNA pull-down assay revealed that miR-222-3p targeted Notch2. CONCLUSION: MiR-222-3p suppressed cell viability, altered cell cycle distribution, and inhibited the formation of MB stem-like cells via the Notch2/c-myc pathway.


Subject(s)
Cerebellar Neoplasms , Medulloblastoma , MicroRNAs , Proto-Oncogene Proteins c-myc , Receptor, Notch2 , Apoptosis/genetics , Caspase 3/metabolism , Cell Line, Tumor , Cell Proliferation , Cerebellar Neoplasms/genetics , Humans , Medulloblastoma/genetics , MicroRNAs/genetics , Neoplastic Stem Cells , Proliferating Cell Nuclear Antigen , Proto-Oncogene Proteins c-myc/genetics , Receptor, Notch2/genetics , Signal Transduction
13.
Int J Mol Sci ; 23(8)2022 Apr 12.
Article in English | MEDLINE | ID: mdl-35457094

ABSTRACT

Gestational diabetes mellitus (GDM) causes both maternal and fetal adverse outcomes. The deregulation of microRNAs (miRNAs) in GDM suggests their involvement in GDM pathogenesis and complications. Exosomes are extracellular vesicles (EVs) of endosomal origin, released via exocytosis into the extracellular compartment. Through EVs, miRNAs are delivered in distant target cells and are able to affect gene expression. In this study, miRNA expression was analyzed to find new miRNAs that could improve GDM classification and molecular characterization. MiRNA were profiled in total plasma and EVs in GDM patients and normal glucose tolerance (NGT) women. Samples were collected at third trimester of gestation from two diabetes centers. MiRNA expression was profiled in a discovery cohort using the multiplexed NanoString nCounter Human v3 miRNA. Validation analysis was performed in a second independent cohort using RT-qPCR. A set of miRNAs resulted to be differentially expressed (DE) in total plasma and EVs in GDM. Among them, total plasma miR-222-3p and miR-409-3p were validated in the independent cohort. MiR-222-3p levels correlated with fasting plasma glucose (FPG) (p < 0.001) and birth weight (p = 0.012), whereas miR-409-3p expression correlated with FPG (p < 0.001) and inversely with gestational age (p = 0.001). The major validated target genes of the deregulated miRNAs were consistently linked to type 2 diabetes and GDM pathophysiology. MiR-222-3p and miR-409-3p are two circulating biomarkers that could improve GDM classification power and act in the context of the molecular events leading to the metabolic alterations observed in GDM.


Subject(s)
Diabetes Mellitus, Type 2 , Diabetes, Gestational , MicroRNAs , Biomarkers , Diabetes, Gestational/genetics , Female , Homeostasis/genetics , Humans , MicroRNAs/metabolism , Pregnancy
14.
Biochem Biophys Res Commun ; 553: 126-133, 2021 05 14.
Article in English | MEDLINE | ID: mdl-33770577

ABSTRACT

Circular RNA (circRNA) homeodomain-interacting protein kinase 3 (circ_HIPK3) has recently reported as regulator in spinal cord injury (SCI). The regulatory mechanism of circ_HIPK3 in SCI was further researched in this study. Circ_HIPK3 expression was inhibited by CoCl2 in AGE1.HN cells. The CoCl2-induced cell cycle arrest, cell proliferation inhibition and apoptosis promotion were mitigated by overexpression of circ_HIPK3. Circ_HIPK3 could target miR-222-3p and circ_HIPK3 repressed the CoCl2-induced neuronal cell injury by sponging miR-222-3p. DUSP19 was a target gene of miR-222-3p and circ_HIPK3 affected the expression of DUSP19 via binding to miR-222-3p. The regulation of circ_HIPK3 in CoCl2-induced injury of AGE1.HN cells was associated with the upregulation of DUSP19. Circ_HIPK3 acted as a pathogenic inhibitor in the progression of SCI via the miR-222-3p-mediated DUSP19 upregulation.


Subject(s)
Apoptosis/drug effects , Cobalt/pharmacology , Dual-Specificity Phosphatases/genetics , MicroRNAs/genetics , Neurons/drug effects , Neurons/pathology , RNA, Circular/genetics , Base Sequence , Cell Line , Dual-Specificity Phosphatases/biosynthesis , Dual-Specificity Phosphatases/deficiency , Dual-Specificity Phosphatases/metabolism , Humans , RNA, Circular/deficiency
15.
Microvasc Res ; 135: 104134, 2021 05.
Article in English | MEDLINE | ID: mdl-33428882

ABSTRACT

BACKGROUND: Clinical data show that aneurysm rupture causes high mortality in aged men. MicroRNAs (miRNAs) were reported to regulate endothelial progenitor cells (EPCs) which play a vital role in repairing endothelial damage and maintaining vascular integrity. This study identified a novel miRNA regulator for the functions of EPCs in aneurysm repair. METHODS: Abdominal aortic aneurysm (AAA) model was established on Sprague-Dawley rats which later underwent antagomiR-222 treatment. The histopathological changes of AAA rats were examined by hematoxylin-eosin staining. Flow cytometry was performed to quantify EPCs in peripheral blood and identify EPCs isolated from the rat femur. The potential target of miR-222-3p was predicted by TargetScan v7.2 and validated by Dual-luciferase reporter assay. The effects of miR-222-3p and ADIPOR1 on the migration, invasion and tube formation of EPCs were evaluated by wound healing, Transwell and tube formation assays. The expressions of miR-222-3p and ADIPOR1 in aortic aneurysm tissues and EPCs were assessed by qRT-PCR or Western blot. RESULTS: AAA exhibited histopathological abnormality, a decreased number of EPCs in the peripheral blood and an increased miR-222-3p expression. AntagomiR-222 injection reversed all these phenomena in AAA rats. Upregulating miR-222-3p expression inhibited the migration, invasion, and tube formation of EPCs, and the expressions of ADIPOR1 and phosphorylated-AMKP, while downregulating miR-222-3p expression exerted opposite effects in EPCs. ADIPOR1 was identified as a target gene of miR-222-3p. Overexpressing ADIPOR1 abrogated the effects of miR-222-3p upregulation on EPCs. CONCLUSION: Downregulated miR-222-3p prompted the migration, invasion and recruitment of EPCs by targeting ADIPOR1-induced AMKP activation.


Subject(s)
AMP-Activated Protein Kinases/metabolism , Aorta, Abdominal/enzymology , Aortic Aneurysm, Abdominal/enzymology , Cell Movement , Endothelial Progenitor Cells/enzymology , MicroRNAs/metabolism , Neovascularization, Physiologic , Receptors, Adiponectin/metabolism , Animals , Antagomirs/genetics , Antagomirs/metabolism , Aorta, Abdominal/pathology , Aortic Aneurysm, Abdominal/genetics , Aortic Aneurysm, Abdominal/pathology , Cells, Cultured , Disease Models, Animal , Down-Regulation , Endothelial Progenitor Cells/pathology , Enzyme Activation , Humans , Male , MicroRNAs/genetics , Phosphorylation , Rats, Sprague-Dawley , Receptors, Adiponectin/genetics , Signal Transduction
16.
Cell Biol Int ; 45(8): 1757-1767, 2021 Aug.
Article in English | MEDLINE | ID: mdl-33851769

ABSTRACT

Circular RNAs (circRNAs) play important roles in the pathogenesis of age-related cataract (ARC). CircRNA zinc finger protein 292 (circZNF292, hsa_circ_0004058) is downregulated in ARC lens capsules. Here, we focused on its precise roles in oxidative stress underlying the pathogenesis of ARC. CircZNF292, microRNA (miR)-222-3p, and E2F transcription factor 3 (E2F3) were quantified by quantitative real-time polymerase chain reaction or western blot. Cell viability was assessed by the cell counting kit-8 assay. Cell cycle distribution and apoptosis were detected by flow cytometry. The activities of superoxide dismutase, catalase, and malondialdehyde were measured using the corresponding assay kit. Targeted correlations among circZNF292, miR-222-3p, and E2F3 were verified by the dual-luciferase reporter, RNA immunoprecipitation and RNA pull-down assays. Our data showed that circZNF292 was downregulated in ARC tissues and H2 O2 -treated human lens epithelial B3 (HLE-B3) cells. Increased expression of circZNF292 alleviated H2 O2 -induced cell viability suppression, apoptosis promotion, and oxidative stress enhancement. Mechanistically, circZNF292 directly targeted miR-222-3p, and circZNF292 regulated E2F3 expression through miR-222-3p. MiR-222-3p was a functional mediator of circZNF292 in modulating H2 O2 -induced injury in HLE-B3 cells. Furthermore, reduced level of miR-222-3p ameliorated H2 O2 -induced HLE-B3 cell damage by upregulating E2F3. Our present study demonstrated that increased expression of circZNF292 ameliorated H2 O2 -induced injury in HLE-B3 cells at least in part through the miR-222-3p/E2F3 axis, highlighting a novel insight into the involvement of circRNAs in the pathogenesis of ARC.


Subject(s)
Carrier Proteins/biosynthesis , E2F3 Transcription Factor/biosynthesis , Epithelial Cells/metabolism , Hydrogen Peroxide/toxicity , Lens, Crystalline/metabolism , MicroRNAs/biosynthesis , Nerve Tissue Proteins/biosynthesis , Aged , Cell Line , Dose-Response Relationship, Drug , Epithelial Cells/drug effects , Female , Humans , Lens, Crystalline/drug effects , Lens, Crystalline/injuries , Male , Middle Aged , RNA, Circular/biosynthesis
17.
Metab Brain Dis ; 36(8): 2521-2534, 2021 12.
Article in English | MEDLINE | ID: mdl-34146216

ABSTRACT

Circular RNAs (circRNAs) have been widely implicated in multiple diseases, including ischemic stroke. This study aimed to explore the function and functional mechanism of circ_0006768 in oxygen-glucose deprivation/reoxygenation (OGD/R)-induced brain injury models of ischemic stroke. Human brain microvascular endothelial cells (HBMECs) were induced by OGD/R to mimic ischemic stroke models in vitro. The expression of circ_0006768, microRNA-222-3p (miR-222-3p) and vascular endothelial zinc finger 1 (VEZF1) was detected by quantitative real-time PCR (qPCR). Cell viability, angiogenesis ability and cell migration were assessed by cell counting kit-8 (CCK-8) assay, tube formation assay and wound healing assay, respectively. The releases of pro-inflammatory factors were determined by commercial enzyme-linked immunosorbent assay (ELISA) kits. The protein levels of vascular endothelial growth factor A (VEGFA), N-cadherin and VEZF1 were detected by western blot. The putative relationship between miR-222-3p and circ_0006768 or VEZF1 was validated by dual-luciferase reporter assay, RNA Immunoprecipitation (RIP) assay and pull-down assay. Circ_0006768 was poorly expressed in ischemic stroke plasma and OGD/R-induced HBMECs. OGD/R inhibited cell viability, angiogenesis and cell migration and promoted the releases of pro-inflammatory factors, while circ_0006768 overexpression or miR-222-3p inhibition partially abolished the effects of OGD/R. MiR-222-3p was targeted by circ_0006768, and VEZF1 was a target of miR-222-3p. Circ_0006768 enriched the expression of VEZF1 via mediating miR-222-3p inhibition. Rescue experiments presented that the effects of circ_0006768 overexpression were reversed by miR-222-3p restoration or VEZF1 knockdown. Circ_0006768 overexpression attenuates OGD/R-induced HBMEC injuries by upregulating VEZF1 via miR-222-3p inhibition.


Subject(s)
Brain Injuries , MicroRNAs , Brain/metabolism , Brain Injuries/metabolism , DNA-Binding Proteins/genetics , Endothelial Cells/metabolism , Glucose/metabolism , Humans , MicroRNAs/genetics , MicroRNAs/metabolism , Oxygen/metabolism , Transcription Factors/genetics , Up-Regulation , Vascular Endothelial Growth Factor A/metabolism
18.
Drug Dev Res ; 82(7): 959-968, 2021 11.
Article in English | MEDLINE | ID: mdl-33543488

ABSTRACT

Glucocorticoids (GCs) can effectively control airway inflammation, but can also cause airway epithelial injury. Tectorigenin, a type of isoflavone isolated from various medicinal plants, has hypolipidemic activity, hepatoprotective, and antioxidant effects. We aimed to investigate whether Tectorigenin can repair GCs-induced airway epithelial injury. Airway epithelial cell line (9HTE cells) were treated with dexamethasone (Dex), Tectorigenin, or further transfected, then cell viability, migration, and invasion were examined by Cell Counting Kit (CCK-8), wound healing, and Transwell assays. The expressions of potential miRNAs related to the effect of Tectorigenin were detected by quantitative polymerase chain reaction (qPCR). Expressions of poptosis-related proteins Bcl-2-associated protein-X (Bax), B-cell lymphoma-2 (Bcl-2), Cleaved Caspase-3, and related to Mitorgen-activated protein kinase (MAPK) signaling pathway serine/threonine kinase (Raf1), extracellular signal-regulated kinase kinase 1/2 (MEK1/2), and extracellular signal-regulated kinase 1/2 (ERK1/2) were detected by Western blot. Dex inhibited the cell viability, migration and invasion by promoting Bax and Cleaved Caspase-3 expressions (p <.001) and by inhibiting the expressions of Bcl-2 and miR-222-3p (p <.001). Then, 10 µmol/L Tectorigenin itself did not affect cell viability but could inhibit the effect of Dex on cell viability, migration, and invasion. Tectorigenin up-regulated the expressions of miR-222-3p, Bcl-2, p-Raf1, p-MEK1/2, and p-ERK1/2 (p <.01), but down-regulated the expressions of Bax and Cleaved Caspase-3 (p <.05) in Dex-induced cells. MiR-222-3p inhibitor reversed the antagonistic effect of Tectorigenin on Dex. The study demonstrates that Tectorigenin inhibits apoptosis of Dex-induced 9HTE cells by up-regulating the expression of miR-222-3p, which involves with the MAPK pathway.


Subject(s)
Apoptosis , Epithelial Cells , Isoflavones , MicroRNAs , Cell Movement , Cell Proliferation , Dexamethasone/pharmacology , Epithelial Cells/cytology , Epithelial Cells/drug effects , Humans , Isoflavones/pharmacology , MicroRNAs/genetics , MicroRNAs/metabolism
19.
J Recept Signal Transduct Res ; 40(4): 301-312, 2020 Aug.
Article in English | MEDLINE | ID: mdl-32202184

ABSTRACT

GILZ expression is induced by glucocorticoids (GCs) and is involved in the mechanism of airway epithelial cell repair in patients with asthma. The present study aimed to investigate the role of miR-222-3p/GILZ pathway in treatment of airway epithelial cell repair by GCs. 9HTE cells were treated by 10 µmol/L dexamethasone (Dex) for 6, 12, and 24 hours (h). MiR-222-3p mimic and GILZ were used for cell transfection. Cell vitality, migration, and invasion were detected by methyl-thiazolyl tetrazolium (MTT), wound healing, and Transwell. The targeting relationship between miR-222-3p and GILZ was predicted by TargetScan and further confirmed by dual-luciferase reporter assay. The expressions of relative mRNAs or proteins were detected by Western blot and quantitative polymerase chain reaction (qPCR). The results showed that Dex treatment up-regulated the GILZ expression level but inhibited the levels of p-Raf1, p-MEK1/2, p-ERK1/2, and miR-222-3p of the cells, moreover, it also inhibited cell activity, migration, and invasion in a time-dependent manner. MiR-222-3p specifically targeted GILZ. MiR-222-3p mimic ameliorated the cell viability, migration, and invasion reduced by Dex treatment, increased the expression levels of p-Raf1 and p-MEK1/2, p-ERK1/2, and partially reversed the effects of GILZ overexpression on the above indexes. Moreover, GILZ showed the opposite effects to miR-222-3p. MiR-222-3p activated MAPK signaling pathway through inhibiting the GILZ expression, thus promoting the cell viability, migration, and invasion previously reduced by Dex.


Subject(s)
Asthma/drug therapy , Glucocorticoids/pharmacology , MicroRNAs/genetics , Transcription Factors/genetics , Asthma/genetics , Asthma/pathology , Cell Line , Cell Movement/drug effects , Cell Proliferation/drug effects , Cell Survival/drug effects , Dexamethasone/pharmacology , Epithelial Cells/drug effects , Gene Expression Regulation/drug effects , Humans , MAP Kinase Signaling System/genetics , Signal Transduction/drug effects
20.
Mol Cell Probes ; 51: 101513, 2020 06.
Article in English | MEDLINE | ID: mdl-31968218

ABSTRACT

AIM: This study aims to explore the role and mechanism of exosomes derived from human bone marrow mesenchymal stem cells (hBM-MSCs-Exo) in regulating proliferation and apoptosis of acute myeloid leukemia (AML) cell line THP-1. METHODS: hBM-MSCs-Exo was isolated by ultra-centrifugation and administered into THP-1 cells to elucidate the effects of exosomes in THP-1 cells. Cell proliferation and apoptosis were examined by CCK-8 assay and flow cytometry, respectively. The expression of miR-222-3p, IRF2, and INPP4B were measured by qRT-PCR and western blot. The interaction between miR-222-3p and IRF2 was analyzed by luciferase reporter assay. RESULTS: Lower cell viability rate, higher apoptosis ratio, higher miR-222-3p expression, and lower IRF1/INPP4B expression were observed in THP-1 cells exposed to BM-MSCs-Exo. The proliferation-inhibitory and pro-apoptotic effects of BM-MSCs-Exo on THP-1 cells were markedly compromised when miR-222-3p expression in BM-MSCs-Exo was inhibited. Furthermore, miR-222-3p directly targeted IRF2 and negatively regulated IRF2/INPP4B signaling in THP-1 cells. Moreover, overexpression of either IRF2 or INPP4B counteracted the proliferation-inhibitory and pro-apoptotic effects mediated by BM-MSCs-Exo. CONCLUSION: BM-MSCs delivered miR-222-3p via exosomes to inhibit cell proliferation and promote cell apoptosis by targeting IRF2 and negatively regulating IRF2/INPP4B signaling in THP-1 cells.


Subject(s)
Apoptosis/genetics , Cell Proliferation/genetics , Exosomes/metabolism , Interferon Regulatory Factor-2/metabolism , Leukemia, Myeloid, Acute/metabolism , Mesenchymal Stem Cells/metabolism , MicroRNAs/metabolism , Phosphoric Monoester Hydrolases/metabolism , Cell Line, Tumor , Cell Survival/genetics , Exosomes/genetics , Humans , Interferon Regulatory Factor-2/genetics , Leukemia, Myeloid, Acute/genetics , MicroRNAs/genetics , Phosphoric Monoester Hydrolases/genetics , Protein Binding , Signal Transduction/genetics , Up-Regulation
SELECTION OF CITATIONS
SEARCH DETAIL