Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 41
Filter
Add more filters










Publication year range
1.
Arch Med Sci ; 20(2): 539-556, 2024.
Article in English | MEDLINE | ID: mdl-38757039

ABSTRACT

Introduction: Long non-coding RNAs (lncRNAs) functioning as competing endogenous RNAs (ceRNAs) play critical roles in tumour progression. However, prognosis-related ceRNA networks in lung adenocarcinoma (LUAD) have not been well characterised. Material and methods: LUAD datasets were downloaded from the TCGA database, and the patients were divided into metastasis and non-metastasis groups. The differential expression of lncRNAs (DELs), miRNAs (DEMs), and mRNAs (DEGs) was analysed using the Limma package. Next, interactions between miRNA, lncRNA, and mRNA were predicted by miRcode, miRTar-Base, miRDB, and TargetScan. The ceRNA network was constructed based on these interactions using Cytoscape software. DEG enrichment analysis was performed by GO and KEGG. After the prognosis analysis, we further screened molecules and constructed the prognosis-related ceRNA network. Moreover, the interactions between lncRNA, miRNA, and mRNA were validated by biological experiments. Results: 854 DELs, 150 DEMs, and 2211 DEGs between metastasis and non-metastasis LUAD patients were identified. Functional enrichment analysis suggested that DEGs were closely related to key biological processes involved in LUAD progression. The prognosis-related ceRNA network included 1 miRNA, 2 lncRNAs, and 4 mRNAs. In this network, MIR155HG and ADAMTS9-AS2 can function as ceRNAs of miR-212 to regulate EPM2AIP1, LAX1, PRICKLE2, and CD226. Moreover, our study confirmed that MIR155HG inhibited the proliferation, migration, and invasion of LUAD cells by sponging miR-212-3p to regulate CD226. Conclusions: This ceRNA network contributes to understanding the pathogenesis of LUAD. Furthermore, the molecules in the network are valuable predictive factors for LUAD prognosis as well as potential therapeutic biomarkers.

2.
Biochem Genet ; 2024 Apr 12.
Article in English | MEDLINE | ID: mdl-38609670

ABSTRACT

The polycystic ovary syndrome (PCOS), a common endocrine disorder, is mainly related to infertility. Moreover, it is characterized by promoted androgen, suppressed ovulation and insulin resistance. Long non-coding RNA X inactive specific transcript (lncRNA XIST), known as an oncogene or a cancer inhabited factor, is involved in several disease. However, the diagnostic mechanisms of lncRNA XIST in PCOS have not been clarified. Our study aimed to explain whether lncRNA XIST regulates KGN cells proliferation and apoptosis via microRNA (miR)-212-3p/RASA1 axis in PCOS. Levels of lncRNA XIST, miR-212-3p and RASA1 in KGN cells were detected through reverse transcription quantitative polymerase chain reaction (RT-qPCR) assay. Fluorescence in situ Hybridization (FISH) was performed to confirm the expression of lncRNA XIST and miR-212-3p in KGN cells. StarBase and dual-luciferase reporter assay were applied for exploring the interaction between miR-212-3p and RASA1. Cell viability, apoptosis, protein expression of Bcl-2 and Bax were assessed by MTT, flow cytometry analysis, RT-qPCR and western blot, respectively. We found that lncRNA XIST was low-expressed, miR-212-3p was over-expressed, and RASA1 was dramatically down-regulated in KGN cells. LncRNA XIST negatively regulated miR-212-3p expression in KGN cells. MiR-212-3p interacted with RASA1 and negatively regulated RASA1 levels in KGN cells. Up-regulation of lncRNA XIST signally decreased cells viability, stimulated more apoptotic cells, enhanced Bax expression, and depressed Bcl-2 level in KGN cells. However, these observations were abolished after miR-212-3p mimic treatment. Furthermore, miR-212-3p inhibitor significantly inhibited cell proliferation, enhanced more apoptotic cells, increased Bax expression, and decreased Bcl-2 level in KGN cells, and these effects were eliminated by RASA1-siRNA transfection. Our observations revealed that lncRNA XIST protects against PCOS through regulating miR-212-3p/RASA1 axis, suggesting that lncRNA XIST may be a promising therapeutic target for PCOS therapy.

3.
Arch Biochem Biophys ; 751: 109827, 2024 01.
Article in English | MEDLINE | ID: mdl-38000494

ABSTRACT

Osteoarthritis (OA) is a common aging-related disease affecting entire joint structures, encompassing articular cartilage and subchondral bone. Although senescence and dysfunction of chondrocytes are considered crucial factors in the occurrence of OA, the exact pathogenesis remains to be investigated. In our study, chondrocytes were incubated with a conditioned medium obtained from osteoclasts at different differentiation stages, suggesting that osteoclasts and osteoclast precursors suppressed anabolism and promoted the catabolism of chondrocytes in vitro. In contrast, the function of osteoclasts was more significant than osteoclast precursors. Further blocking of osteoclast exosome secretion by using GW4869 abolished the effect of osteoclasts on chondrocytes. Functionally, exosomal transfer of osteoclast-derived miR-212-3p inhibited Smad2 to mediate chondrocyte dysfunction, thus accelerating cartilage matrix degradation in OA via TGF-ß1/Smad2 signaling. The mechanism was also confirmed within the articular cartilage in OA patients and surgery-induced OA mice. Our study provides new information on intercellular interactions in the bone microenvironment within articular cartilage and subchondral bone during OA progression. The miR-212-3p/Smad2 axis is a potential target for the prevention and therapy of OA.


Subject(s)
Cartilage, Articular , MicroRNAs , Osteoarthritis , Animals , Humans , Mice , Cartilage, Articular/metabolism , Chondrocytes/metabolism , MicroRNAs/genetics , MicroRNAs/metabolism , Osteoarthritis/metabolism , Osteoclasts/metabolism , Transforming Growth Factor beta1/metabolism
4.
Int J Exp Pathol ; 105(1): 21-32, 2024 Feb.
Article in English | MEDLINE | ID: mdl-38054576

ABSTRACT

Sepsis-induced acute lung injury (ALI) is an inflammatory condition involving the pyroptosis of macrophages. This study investigated the role of circular RNA hsa_circ_0006990 (circVAPA) in regulating macrophage pyroptosis in ALI and the underlying mechanisms. The expression pattern of circVAPA was examined in the mouse model of ALI and in the LPS-treated RAW264.7 macrophage cell line. Lung tissue damage was evaluated by haematoxylin and eosin staining, immunohistochemistry and a myeloperoxidase activity assay. The molecular mechanisms were investigated by luciferase reporter assay, western blot, RT-qPCR and ELISA. circVAPA was down-regulated in the lung tissues of ALI mice and LPS-induced RAW264.7 cells. circVAPA over-expression alleviated lung tissue injury and dampened LPS-induced pyroptosis and Th17-associated inflammatory responses. miR-212-3p was identified as a target of circVAPA, and miR-212-3p negatively regulated the expression of Sirt1. Sirt1 knockdown largely abolished the effect of circVAPA over-expression on pyroptosis. CircVAPA/miR-212-3p/Sirt1 axis also regulates Nrf2 and NLRP3 expression upon LPS challenge. By targeting miR-212-3p, circVAPA over-expression negatively regulates the expression of Sirt1 and pyroptosis-related factors (Nrf2 and NLRP3), which alleviates the inflammatory damages in sepsis-induced ALI.


Subject(s)
Acute Lung Injury , MicroRNAs , Sepsis , Animals , Mice , NF-E2-Related Factor 2/genetics , RNA, Circular/genetics , Sirtuin 1/genetics , Lipopolysaccharides , NLR Family, Pyrin Domain-Containing 3 Protein/genetics , Pyroptosis/genetics , Acute Lung Injury/genetics , Macrophages , Sepsis/complications , Sepsis/genetics , MicroRNAs/genetics
5.
Kaohsiung J Med Sci ; 39(12): 1222-1232, 2023 Dec.
Article in English | MEDLINE | ID: mdl-37987200

ABSTRACT

This study was to probe the protective effects and mechanisms of salvianolic acid A (SAA) on cerebral ischemia-reperfusion injury (CIRI). The middle cerebral artery occlusion model (MCAO) was established in rats. Rats' behavior, neurological deficits, brain injury, inflammation, and apoptosis in the brain tissue were evaluated. The inflammatory response and apoptosis of PC12 cells induced by oxygen glucose deprivation/reperfusion (OGD/R) were detected. SAA-mediated changes in miR-212-3p, SOX7, and Wnt/ß-catenin pathway were determined, and the targeting relationship between miR-212-3p and SOX7 was clarified. SAA alleviated the neurological deficits and brain injury of MCAO rats and inhibited the inflammatory response and apoptosis of OGD/R-conditioned PC-12 cells. SAA upregulated miR-212-3p, Wnt3a, and ß-catenin, whereas inhibited SOX7 levels. Silencing miR-212-3p counteracted the protective effect of SAA in the context of CIRI. SOX7 was a target protein of miR-212-3p. Silencing SOX7 based on SAA and miR-212-3p knockdown suppressed OGD/R-induced inflammation and apoptosis and increased Wnt3a and ß-catenin levels in PC12 cells. SAA can improve the brain and nervous system injury caused by cerebral ischemia-reperfusion by upregulating miR-212-3p, thereby inhibiting SOX7 and activating the Wnt/ßcatenin signaling pathway.


Subject(s)
Brain Injuries , Brain Ischemia , MicroRNAs , Reperfusion Injury , Rats , Animals , beta Catenin/genetics , MicroRNAs/metabolism , Brain Ischemia/metabolism , Reperfusion Injury/drug therapy , Reperfusion Injury/genetics , Brain Injuries/drug therapy , Brain Injuries/genetics , Inflammation/complications , Nerve Regeneration/genetics , Apoptosis/genetics
6.
Aging (Albany NY) ; 15(22): 13411-13421, 2023 11 28.
Article in English | MEDLINE | ID: mdl-38019469

ABSTRACT

To explore the mechanisms of action of micro ribonucleic acid (miR)-212-3p in the secretion of inflammatory factors in monocyte-macrophages and the directed differentiation into osteoclasts (OCs) in ankylosing spondylitis (AS), proteoglycan was used to establish an AS mouse model. The mouse monocyte-macrophages were cultured in vitro, transfected with miR-212-3p mimic, and added with phosphorylated-extracellular signal-regulated kinase (p-ERK)1/2 agonist Ro67-7476 in vitro. After the cells were transfected with the miR-212-3p mimic in each group, the expressions of p-ERK1/2, matrix metalloproteinase-1 (MMP-1), MMP-3, interleukin-1ß (IL-1ß), and tumor necrosis factor-α (TNF-α) significantly declined, whereas those of tartrate-resistant acid phosphatase (TRAP), calcitonin, and p-nuclear factor of activated T cell 1 (NFATC1) significantly rose. After Ro67-7476 was added, the protein expressions of p-ERK1/2, MMP-1, MMP-3, IL-1ß, and TNF-α were significantly increased in each group, but they displayed decreasing trends in cells transfected with the miR-212-3p mimic. In contrast, the protein expressions of TRAP, calcitonin, and p-NFATC1 declined, but they showed increasing trends in cells transfected with the miR-212-3p mimic. miR-212-3p can, through inhibiting the phosphorylation of p-ERK1/2, prevent the aggregation of macrophages and the secretion of inflammatory factors. It also up-regulates the expression of OC marker proteins to facilitate the differentiation and maturation of OCs, ultimately relieving AS-induced inflammation and new bone growth-induced joint neoplasm.


Subject(s)
Cell Differentiation , MicroRNAs , Spondylitis, Ankylosing , Animals , Mice , Calcitonin , Cell Differentiation/genetics , Macrophages/metabolism , Matrix Metalloproteinase 1 , Matrix Metalloproteinase 3 , MicroRNAs/genetics , MicroRNAs/metabolism , Monocytes/metabolism , Osteoclasts/metabolism , Spondylitis, Ankylosing/genetics , Tumor Necrosis Factor-alpha/metabolism
7.
Diabetol Metab Syndr ; 15(1): 119, 2023 Jun 07.
Article in English | MEDLINE | ID: mdl-37280692

ABSTRACT

BACKGROUND: Long noncoding RNAs (LncRNAs) have been identified to play an important role in diabetes. The aim of the present study was to determine the expression and function of small nucleolar RNA host gene 16 (SNHG16) in diabetic inflammation. METHODS: For the in vitro experiments, quantitative real-time PCR (qRT-PCR), Western blotting and immunofluorescence were used to detect LncRNA SNHG16 expression in the high-glucose state. The potential microRNA sponge target of LncRNA SNHG16, miR-212-3p, was detected by dual-luciferase reporter analysis and qRT-PCR. For the in vivo experiments, glucose changes in mice were detected after si-SNHG16 treatment, and SNHG16 and inflammatory factor expression in kidney tissues were detected by qRT-PCR and immunohistochemistry. RESULTS: LncRNA SNHG16 was upregulated in diabetic patients, HG-induced THP-1 cells, and diabetic mice. Silencing SNHG16 inhibited the diabetic inflammatory response and the development of diabetic nephropathy. miR-212-3p was found to be directly dependent on LncRNA SNHG16. miR-212-3p could inhibitor P65 phosphorylation in THP-1 cells. The miR-212-3p inhibitor reversed the action of si-SNHG16 in THP-1 cells and induced an inflammatory response in THP-1 cells. LncRNA SNHG16 was also found to be higher in the peripheral blood of diabetic patients than in the normal person. The area under the ROC curve is 0.813. CONCLUSION: These data suggested that silencing LncRNA SNHG16 suppresses diabetic inflammatory responses by competitively binding miR-212-3p to regulate NF-κB. LncRNA SNHG16 can be used as a novel biomarker for patients with type 2 diabetes.

8.
Int J Mol Sci ; 24(3)2023 Feb 01.
Article in English | MEDLINE | ID: mdl-36769104

ABSTRACT

Hypoxia impairs blood-brain barrier (BBB) structure and function, causing pathophysiological changes in the context of stroke and high-altitude brain edema. Brain microvascular endothelial cells (BMECs) are major structural and functional elements of the BBB, and their exact role in hypoxia remains unknown. Here, we first deciphered the molecular events that occur in BMECs under 24 h hypoxia by whole-transcriptome sequencing assay. We found that hypoxia inhibited BMEC cell cycle progression and proliferation and downregulated minichromosome maintenance complex component 2 (Mcm2) expression. Mcm2 overexpression attenuated the inhibition of cell cycle progression and proliferation caused by hypoxia. Then, we predicted the upstream miRNAs of MCM2 through TargetScan and miRanDa and selected miR-212-3p, whose expression was significantly increased under hypoxia. Moreover, the miR-212-3p inhibitor attenuated the inhibition of cell cycle progression and cell proliferation caused by hypoxia by regulating MCM2. Taken together, these results suggest that the miR-212-3p/MCM2 axis plays an important role in BMECs under hypoxia and provide a potential target for the treatment of BBB disorder-related cerebrovascular disease.


Subject(s)
Endothelial Cells , MicroRNAs , Humans , Endothelial Cells/metabolism , Minichromosome Maintenance Complex Component 2/genetics , Minichromosome Maintenance Complex Component 2/metabolism , Brain/metabolism , MicroRNAs/genetics , MicroRNAs/metabolism , Cell Proliferation/genetics , Cell Division , Hypoxia/genetics , Hypoxia/metabolism , Cell Hypoxia/genetics
9.
Cell Signal ; 102: 110554, 2023 02.
Article in English | MEDLINE | ID: mdl-36476391

ABSTRACT

Emerging evidence indicates that multiple mechanisms are involved in bone loss induced by mechanical unloading. Thus far, few study has established the pathophysiological role of histone modification for osteogenic differentiation under mechanical unloading. Here we demonstrated that the histone H3 lysine 9 (H3K9) methyltransferase Setdb1, which was sensitive to mechanical unloading, was increased during osteogenic differentiation of MC3T3-E1 cells for the first time. Knockdown of Setdb1 significantly blocked osteoblast function in vivo and in vitro. Through bioinformatics analysis of candidate miRNAs regulated by H3K9me3, we further identified that Setdb1 inhibited the expression of miR-212-3p by regulating the formation of H3K9me3 in the promoter region. Mechanically, we revealed that miR-212-3p was upregulated under mechanical unloading and suppressed osteogenic differentiation by directly downregulating High mobility group box 1 protein (Hmgb1) expression. Furthermore, we verified the molecular mechanism of the SETDB1/miR-212-3p/HMGB1 pathway in hFOB cells under mechanical unloading. In summary, these data demonstrate the essential function of the Setdb1/miR-212-3p/Hmgb1 pathway in osteogenic differentiation under mechanical unloading, and present a potential protective strategies against bone loss induced by mechanical unloading.


Subject(s)
HMGB1 Protein , MicroRNAs , Histone Methyltransferases/metabolism , HMGB1 Protein/metabolism , Osteogenesis/genetics , Cell Differentiation , MicroRNAs/genetics , MicroRNAs/metabolism , Osteoblasts/metabolism
10.
Eur J Med Res ; 27(1): 135, 2022 Jul 27.
Article in English | MEDLINE | ID: mdl-35897048

ABSTRACT

BACKGROUND: Circular RNAs (circRNA) are important in mediating tumor progression, but their roles in endometrial carcinoma (EC) are not fully understood yet. Many circRNAs are dysregulated and may contribute to EC progression. The functions of circWDR26 in EC remain unknown. METHODS: The expression of circWDR26 in EC and adjacent normal tissues, and cell lines was determined by qPCR. The proliferation, apoptosis, migration, and invasion of EC cells was examined by CCK-8 assay, flow cytometry, wound healing assay and Transwell assay. The interaction between circWDR26, MSH2 and miR-212-3p was determined by luciferase assay. EC cells were inoculated into nude mice and tumor burden was determined by measuring tumor dimensions, size, and weight. The proliferative marker Ki67 in EC tissue was determined by immunohistochemistry. RESULTS: The expression of circWDR26 in EC tissues or cell lines was higher than in the normal tissue or endometrial epithelial cells. Downregulation of circWDR26 resulted in attenuated proliferation, increased apoptosis, reduced migration and invasion of EC cells. Mechanistically, circWDR26 targeted and suppressed the expression of miR-212-3p. We further found that MSH2 was the novel target of miR-212-3p and was upregulated by circWDR26 via inhibiting miR-212-3p. In vivo experiment demonstrated that circWDR26 was essential for EC tumor growth. CONCLUSION: circWDR26 promoted EC progression by regulating miR-212-3p/MSH2 axis and provided novel insights into anti-cancer treatment.


Subject(s)
Endometrial Neoplasms , MicroRNAs , MutS Homolog 2 Protein , RNA, Circular , Animals , Cell Line, Tumor , Cell Proliferation/genetics , Endometrial Neoplasms/genetics , Endometrial Neoplasms/pathology , Female , Humans , Mice , Mice, Nude , MicroRNAs/genetics , MutS Homolog 2 Protein/genetics , RNA, Circular/genetics
11.
Stem Cell Res Ther ; 13(1): 130, 2022 03 28.
Article in English | MEDLINE | ID: mdl-35346361

ABSTRACT

BACKGROUND: The treatment of bone loss has posed a challenge to clinicians for decades. Thus, it is of great significance to identify more effective methods for bone regeneration. However, the role and mechanisms of long non-coding RNA small nucleolar RNA host gene 5 (SNHG5) during osteogenic differentiation remain unclear. METHODS: We investigated the function of SNHG5, Yin Yang 1 (YY1), miR-212-3p and growth differentiation factor 5 (GDF5) in osteogenic differentiation of human bone marrow mesenchymal stem cells (hBMSCs) in vitro and in vivo. Molecular mechanisms were clarified by chromatin immunoprecipitation assay and dual luciferase reporter assay. RESULTS: We found SNHG5 expression was upregulated during osteogenesis of hBMSCs. Knockdown of SNHG5 in hBMSCs inhibited osteogenic differentiation while overexpression of SNHG5 promoted osteogenesis. Moreover, YY1 transcription factor directly bound to the promoter region of SNHG5 and regulated SNHG5 expression to promote osteogenesis. Dual luciferase reporter assay confirmed that SNHG5 acted as a miR-212-3p sponge and miR-212-3p directly targeted GDF5 and further activated Smad1/5/8 phosphorylation. miR-212-3p inhibited osteogenic differentiation, while GDF5 promoted osteogenic differentiation of hBMSCs. In addition, calvarial defect experiments showed knockdown of SNHG5 and GDF5 inhibited new bone formation in vivo. CONCLUSION: Our results demonstrated that the novel pathway YY1/SNHG5/miR-212-3p/GDF5/Smad regulates osteogenic differentiation of hBMSCs and may serve as a potential target for the treatment of bone loss.


Subject(s)
Mesenchymal Stem Cells , MicroRNAs , Osteogenesis , RNA, Long Noncoding , Growth Differentiation Factor 5/genetics , Growth Differentiation Factor 5/metabolism , Humans , Mesenchymal Stem Cells/cytology , MicroRNAs/genetics , RNA, Long Noncoding/genetics
12.
Bioengineered ; 13(2): 4166-4172, 2022 02.
Article in English | MEDLINE | ID: mdl-35112979

ABSTRACT

Long non-coding RNA (lncRNA) TSPEAR-AS2 (TSPEAR Antisense RNA 2) participates in many human diseases, while its roles in rheumatoid arthritis (RA) are unknown. Plasma expression levels of TSPEAR-AS2 and microRNA (miR)-212-3p in both RA patients and healthy controls were measured by RT-qPCR. Diagnostic potentials of plasma TSPEAR-AS2 and miR-212-3p were assessed by ROC curve analysis. Normalized expression levels of TSPEAR-AS2 and miR-212-3p were subjected to Pearson's correlation coefficient to evaluate their corrections. TSPEAR-AS2 was significantly downregulated in RA patients, while plasma expression levels of miR-212-3p were significantly increased in RA patients. The expression of TSPEAR-AS2 and miR-212-3p showed promising diagnostic value for RA. Plasma expression levels of TSPEAR-AS2 and miR-212-3p were significantly and inversely correlated in RA patients but not in healthy controls. Besides, overexpression of TSPEAR-AS2 decreased the apoptosis of RA HFLSs, while miR-212-3p increased cell apoptosis. In addition, miR-212-3p attenuated the effects of overexpression of TSPEAR-AS2. Overexpression of TSPEAR-AS2 decreased the expression levels of miR-212-3p in HFLS, while overexpression of miR-212-3p did not affect the expression of TSPEAR-AS2. In conclusion, TSPEAR-AS2 is downregulated in RA and its overexpression can decrease the apoptosis of RA HFLSs by downregulating miR-212-3p.


Subject(s)
Apoptosis/genetics , Arthritis, Rheumatoid , MicroRNAs , RNA, Long Noncoding , Synoviocytes/metabolism , Adult , Aged , Arthritis, Rheumatoid/blood , Arthritis, Rheumatoid/genetics , Cells, Cultured , Down-Regulation/genetics , Female , Fibroblasts/metabolism , Humans , Male , MicroRNAs/blood , MicroRNAs/genetics , Middle Aged , RNA, Long Noncoding/blood , RNA, Long Noncoding/genetics
13.
Acta Neuropathol Commun ; 9(1): 195, 2021 12 18.
Article in English | MEDLINE | ID: mdl-34922631

ABSTRACT

Haploinsufficiency of chromosome 17p and c-Myc amplification distinguish group 3 medulloblastomas which are associated with early metastasis, rapid recurrence, and swift mortality. Tumor suppressor genes on this locus have not been adequately characterized. We elucidated the role of miR-212-3p in the pathophysiology of group 3 tumors. First, we learned that miR-212-3p undergoes epigenetic silencing by histone modifications in group 3 tumors. Restoring its expression reduced cancer cell proliferation, migration, colony formation, and wound healing in vitro and attenuated tumor burden and improved survival in vivo. MiR-212-3p also triggered c-Myc destabilization and degradation, leading to elevated apoptosis. We then isolated an oncogenic target of miR-212-3p, i.e. NFIB, a nuclear transcription factor implicated in metastasis and recurrence in various cancers. Increased expression of NFIB was confirmed in group 3 tumors and associated with poor survival. NFIB silencing reduced cancer cell proliferation, migration, and invasion. Concurrently, reduced medullosphere formation and stem cell markers (Nanog, Oct4, Sox2, CD133) were noted. These results substantiate the tumor-suppressive role of miR-212-3p in group 3 MB and identify a novel oncogenic target implicated in metastasis and tumor recurrence.


Subject(s)
Cerebellar Neoplasms/metabolism , Gene Expression Regulation, Neoplastic/genetics , Medulloblastoma/metabolism , MicroRNAs/metabolism , NFI Transcription Factors/metabolism , Animals , Cells, Cultured , Cerebellar Neoplasms/genetics , Disease Models, Animal , Humans , Medulloblastoma/genetics , Mice , MicroRNAs/genetics , NFI Transcription Factors/genetics
14.
J Diabetes Complications ; 35(11): 108020, 2021 11.
Article in English | MEDLINE | ID: mdl-34507876

ABSTRACT

Previous studies found that hypoxic pretreatment of endothelial progenitor cells (EPCs) prior to transplantation had a greater therapeutic effect than untreated EPCs in promoting diabetic wound healing. However, the exact mechanism is uncertain. Here, circRNA expression in EPCs after hypoxic treatment was investigated. High-throughput sequencing was used to assess abnormal expression by EPCs of circular RNAs (circRNAs) following hypoxic pretreatment. Additionally, an in vivo full-thickness skin defect mouse model was used to assess the effects of transplanted EPCs on diabetic wound closure. Subsequently, the regulatory mechanism and targets were studied. The results showed that circ-Klhl8 overexpression suppressed hyper glucose-induced endothelial cell damage by activating autophagy. MiR-212-3p and SIRT5 were identified as the downstream targets of circ-Klhl8. Circ-Klhl8 overexpression promoted skin wound healing by regulating SIRT5-mediated autophagy. In conclusion, the study found that circ-Klhl8 overexpression increased the EPC therapeutic effect in promoting diabetic wound healing by targeting the miR-212-3p/SIRT5 axis.


Subject(s)
Diabetes Mellitus , Endothelial Progenitor Cells , MicroRNAs , RNA, Circular/genetics , Sirtuins , Wound Healing , Animals , Mice , MicroRNAs/genetics , Stem Cell Transplantation , Wound Healing/genetics
15.
Bioengineered ; 12(1): 5655-5663, 2021 12.
Article in English | MEDLINE | ID: mdl-34470571

ABSTRACT

The purpose of this study was to evaluate the function and possible mechanism of miR-212-3p in fetal growth restriction (FGR) and to demonstrate the relationship between miR-212-3p and placental growth factor (PGF). First, we used qRT-PCR to detect the expression of miR-212-3p and PGF in placental tissues of normal delivery (HC group) and FGR, as well as in human trophoblast cell HTR-8/Svneo. The results revealed that miR-212-3p expression was significantly upregulated and PGF was significantly downregulated in placental tissue in the FGR group compared with the HC group. In addition, interference with miR-212-3p expression increased the proliferation, invasion, and migration of HTR-8/SVneo cells and decreased apoptosis of cells. Meanwhile, Western blot results showed that miR-212-3p expression downregulation promoted the phosphorylated protein expression of Phosphoinositide 3-kinase (PI3K) and protein kinase B (AKT), which in turn activated the PI3K/AKT signaling pathway. And the results of dual luciferase reporter further showed that miR-212-3p could target PGF, and the expression of both was negatively correlated in FGR group tissues. In addition, downregulation of miR-212-3p expression reversed the inhibitory effect of PGF downregulation on HTR-8/SVneo cells. In conclusion, miR-212-3p can target and inhibit the PGF expression and regulate the PI3K/AKT signaling pathway to regulate trophoblast cell invasion, migration, proliferation and cell apoptosis. This provides a potential biomarker for the development of FGR.


Subject(s)
Fetal Growth Retardation , MicroRNAs , Trophoblasts/metabolism , Biomarkers/metabolism , Cell Line , Cell Movement/genetics , Cell Proliferation/genetics , Female , Fetal Growth Retardation/genetics , Fetal Growth Retardation/metabolism , Fetal Growth Retardation/pathology , Humans , MicroRNAs/genetics , MicroRNAs/metabolism , Placenta/cytology , Placenta/metabolism , Placenta/pathology , Pregnancy
16.
Brain Res ; 1770: 147622, 2021 11 01.
Article in English | MEDLINE | ID: mdl-34403662

ABSTRACT

BACKGROUND: Alzheimer's Disease (AD) is a neurodegenerative disorder characterized by progressive memory loss and cognition and language impairment. CircRNA lysophosphatidic acid receptor 1 (circLPAR1) was found to be increased in AD patients, however, the potential role of circLPAR1 in AD process remains unclear. METHODS: Beta-amyloid (Aß) 25-35-stimulated CHP-212 and IMR-32 cells were used to perform expression and function analyses. The expression of genes and proteins was determined by qRT-PCR and Western blot. Cell proliferation and apoptosis were analyzed using cell counting kit-8 (CCK-8) assay, flow cytometry, and Western blot, respectively. ELISA analysis was used to detect the levels of interleukin (IL)-1ß, IL-6, and tumor necrosis factor-α (TNF-α). The levels of reactive oxygen species (ROS), lactate dehydrogenase (LDH) and superoxide dismutase (SOD) were detected using commercial kits. The direct interactions between miR-212-3p and ZNF217 (Zinc finger protein 217) or circLPAR1 was verified using dual-luciferase reporter and RNA immunoprecipitation (RIP) assays. RESULTS: CircLPAR1 was highly expressed in AD patients and Aß25-35-stimulated CHP-212 and IMR-32 cells. Knockdown of circLPAR1 suppressed Aß25-35-induced neuronal apoptosis, inflammation, and oxidative stress. Mechanistically, circLPAR1 competitively bound to miR-212-3p to elevate its target ZNF217. Rescue experiments suggested that miR-212-3p inhibition reversed circLPAR1 silencing-evoked inhibition on neuronal injury under Aß25-35 stimulation. Moreover, miR-212-3p re-expression reduced Aß25-35-induced neuronal apoptosis, inflammation, and oxidative stress, which were abolished by ZNF217 up-regulation. CONCLUSION: CircLPAR1 promotes Aß25-35-induced apoptosis, inflammation, and oxidative stress via miR-212-3p/ZNF217 axis, suggesting a new insight into the pathogenesis of AD.


Subject(s)
Alzheimer Disease/metabolism , Amyloid beta-Peptides/pharmacology , MicroRNAs/metabolism , Neurons/metabolism , Peptide Fragments/pharmacology , RNA, Circular/metabolism , Trans-Activators/metabolism , Alzheimer Disease/genetics , Cell Line , Humans , L-Lactate Dehydrogenase/metabolism , MicroRNAs/genetics , Neurons/drug effects , Oxidative Stress/drug effects , RNA, Circular/genetics , Reactive Oxygen Species/metabolism , Superoxide Dismutase/metabolism , Trans-Activators/genetics
17.
Brain Res ; 1768: 147576, 2021 10 01.
Article in English | MEDLINE | ID: mdl-34216580

ABSTRACT

BACKGROUND: Multiple cellular and molecular changes are involved in the etiology of spinal cord injury (SCI) and the recovery from SCI. Accumulating studies showed aberrant expression of microRNAs (miRNAs) after SCI. Here, we established in vivo and in vitro models to analyze the role of miR-212-3p in SCI. METHODS: An in vivo model of SCI was established in Sprague-Dawley rats. SCI-induced histopathological changes of the spinal cord were observed by hematoxylin-eosin staining. Functional recovery of rats with SCI was evaluated using the Basso-Beattie-and-Bresnahan scale. PC12 cells were stimulated by lipopolysaccharide (LPS) to establish SCI model of neuronal apoptosis in vitro. Dual-luciferase reporter assay was performed to validate the potential target of miR-212-3p predicted by TargetScan 7.2. MTT assay and flow cytometry were carried out to measure the viability and apoptosis of PC12 cell, respectively. The expressions of miR-212-3p, PTEN, phosphorylated (p)-AKT, AKT, p-mTOR, mTOR, Cleaved caspase-3 and BCl-2 in spinal cord tissues and PC12 cells were analyzed by qRT-PCR or Western blot. RESULTS: In the spinal cord of rats with SCI, the expressions of miR-212-3p, p-AKT, p-mTOR and BCl-2 were downregulated, whereas those of PTEN and Cleaved caspase-3 were upregulated. BBB scores were low, and there were histopathological changes, which were all reversed after the injection of agomiR-212-3p. MiR-212-3p directly targeted PTEN. Upregulated miR-212-3p in LPS-injured PC12 cells suppressed apoptosis, downregulated the expressions of PTEN and Cleaved caspase-3, promoted viability and upregulated the expressions of p-AKT, p-mTOR and BCl-2, which were all reversed by overexpressed PTEN. CONCLUSION: MiR-212-3p improved functional recovery of SCI rats and inhibited LPS-induced neurocyte apoptosis by targeting PTEN to activate AKT/mTOR pathway.


Subject(s)
MicroRNAs/genetics , Recovery of Function/genetics , Spinal Cord Injuries/genetics , Animals , Apoptosis/genetics , Male , MicroRNAs/metabolism , Neurons/metabolism , PC12 Cells , PTEN Phosphohydrolase/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Rats , Rats, Sprague-Dawley , Recovery of Function/physiology , Spinal Cord/metabolism , TOR Serine-Threonine Kinases/metabolism
18.
Front Oncol ; 11: 657094, 2021.
Article in English | MEDLINE | ID: mdl-33869063

ABSTRACT

Long non-coding RNAs(lncRNAs) play an important role in cancer initiation and progression. However, hub lncRNAs involved in breast cancer still remain underexplored. In this study, integrated bioinformatics analysis was used to define LINC01977 as a key oncogenic driver in breast cancer. Subsequently, in vitro assays showed that LINC01977 could significantly promote breast cancer progression and chemoresistance to doxorubicin. To further investigate its biological mechanism, we performed dual-luciferase reporter assay, real-time PCR, RNA immunoprecipitation (RIP), and rescue assay. Our results indicated that LINC01977 may function as ceRNA to prevent GOLM1 gene from miRNA-mediated repression by sponging miR-212-3p. Overall, LINC01977 can serve as a novel prognostic indicator, and help develop more effective therapeutic approaches for breast cancer patients.

19.
Cancer Manag Res ; 13: 899-912, 2021.
Article in English | MEDLINE | ID: mdl-33564265

ABSTRACT

BACKGROUND: Linc-ROR is a long non-coding RNA, that is found aberrantly expressed in various human cancers. We aim here to unveil the role of Linc-ROR in gastric cancer (GC) progression. METHODS: qPCR was used to determine gene expression. Cell viability was measured by CCK-8 assay. Transwell assays were performed to evaluate the GC cells' migratory and invasive abilities. Xenograft mouse model was conducted to measure tumor growth. RESULTS: We found that Linc-ROR were overexpressed in GC tissues compared to the adjacent tissues. High Linc-ROR predicts poor prognosis of GC patients. The prediction of bioinformatics online revealed that Linc-ROR could bind to miR-212-3p. Further, dual-luciferase reporter assay confirmed a direct interaction between Linc-ROR and miR-212-3p. Overexpression of miR-212-3p facilitated GC cells' migration and invasion, while the silencing of miR-212-3p attenuated GC cell migratory and invasive abilities. Moreover, Linc-ROR knockdown significantly suppressed the proliferation, migration, and invasion of GC cells, whereas miR-212-3p antagomir partially reversed Linc-ROR knockdown-induced phenotypes. Fibroblast growth factor 7 (FGF7), a downstream molecule of miR-212-3p, was overexpressed in GC cells. The recovery of FGF7 expression partially reversed the phenotypes caused by Linc-ROR silencing. Mechanistically, silencing of Linc-ROR contributed to the downregulation of CDK4, CDK6, Cyclin D1, N-Cadherin, Vimentin, MMP-9, MMP-2, but caused the upregulation of P21, P27, E-Cadherin, CK-19 in MGC-803 cells; however, FGF7 treatment could reverse the results induced by Linc-ROR silencing. Results in vivo further suggested that Linc-ROR knockdown repressed GC tumor growth, where the expression of miR-212-3p was up-regulated and FGF7 expression was downregulated in tumor tissues of mice. CONCLUSION: These findings indicated that Linc-ROR/miR-212-3p/FGF7 axis played an important role in gastric cancer progression. Linc-ROR expression level was associated with the prognosis of GC patients.

20.
Neurochem Res ; 46(2): 230-240, 2021 Feb.
Article in English | MEDLINE | ID: mdl-33241432

ABSTRACT

Long non-coding RNA (lncRNA) nuclear-enriched assembly transcript 1 (NEAT1) has been reported to be highly expressed in Parkinson's disease (PD). However, the mechanism of NEAT1 in PD progression has not been fully elucidated. 1-methyl-4-phenyl-1, 2, 3, 6-tetrahydropyridine injection (MPTP) was used to construct PD mouse models in vivo, and 1-methyl-4-phenyl pyridine (MPP+) was used to build PD cell models in vitro. Quantitative real-time polymerase chain reaction (qRT-PCR) was employed to test the expression of NEAT1, microRNA (miR)-212-3p and axis inhibition protein 1 (AXIN1). The viability, apoptosis and inflammation of cells were determined using cell counting kit 8 (CCK8) assay, flow cytometry and enzyme-linked immunosorbent assay (ELISA), respectively. Then, the protein levels of apoptosis-related markers and AXIN1 were measured by western blot (WB) analysis. Furthermore, dual-luciferase reporter assay and RNA immunoprecipitation (RIP) assay were performed to verify the interaction between miR-212-3p and NEAT1 or AXIN1. NEAT1 was upregulated in PD mouse models and cell models. Function experiments confirmed that NEAT1 knockdown could promote the viability, suppress the apoptosis and inflammation of MPP+-stimulated SK-N-SH cells to restrain PD progression. MiR-212-3p was downregulated in PD, and its inhibitor could reverse the suppression effect of NEAT1 knockdown on PD progression. Additionally, AXIN1 was a target of miR-212-3p, and its overexpression could invert the inhibition effect of miR-212-3p mimic on PD progression. Furthermore, AXIN1 expression was inhibited by NEAT1 silencing and promoted by NEAT1 overexpression, while these effect could be recovered by miR-212-3p inhibitor and mimic, respectively. Our results demonstrated that NEAT1 knockdown suppressed PD progression through regulating the miR-212-3p/AXIN1 pathway, indicating that NEAT1 might be a therapeutic target for neuroprotection in PD.


Subject(s)
Axin Protein/metabolism , MicroRNAs/metabolism , Parkinson Disease/metabolism , RNA, Long Noncoding/metabolism , Animals , Apoptosis/physiology , Cell Line, Tumor , Down-Regulation , Gene Knockdown Techniques , Humans , Inflammation/metabolism , Mice, Inbred C57BL , RNA, Long Noncoding/genetics , Up-Regulation
SELECTION OF CITATIONS
SEARCH DETAIL