Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 8 de 8
Filter
1.
J Gene Med ; 22(11): e3248, 2020 11.
Article in English | MEDLINE | ID: mdl-32621625

ABSTRACT

BACKGROUND: Numerous studies have shown that aberrant expression of long non-coding RNAs (lncRNAs) is associated with the development and metastasis of osteosarcoma (OS). However, the role and function of LINC00319 with respect to regulating OS progression is unknown. The present study aimed to reveal the function and related mechanism of LINC00319 in OS. METHODS: The expression of LINC00319, miR-455-3p and nuclear factor IB (NFIB) in OS cells and tissues was determined using a reverse transcriptase-polymerase chain reaction (PCR). The sublocalization of LINC00319 was predicted by the lncATLAS database (http://lncatlas.crg.eu) and RNA fluorescence in situ hybridization (FISH) was further performed to detect the subcellular localization of LINC00319. LINC00319, miR-455-3p and NFIB target sites were predicted by StarBase (http://starbase.sysu.edu.cn/index.php) and validated using a dual luciferase reporter gene assay. We subsequently performed LINC00319 gain- and loss-of-function studies to define the role of LINC00319 in OS cell migration. RESULTS: PCR results showed that lncRNA LINC00319 exhibited high expression in tumor cells and tissue. Moreover, LINC00319 was positioned in the cytoplasm, which was identified by FISH. Knockdown of lncRNA LINC00319/NFIB or overexpression of miR-455-3p blocked the migration of OS cells. In addition, the inhibitory effect of migration with the knockdown of lncRNA LINC00319 was partially blocked by administration of miR-455-3p inhibitor. CONCLUSIONS: lncRNA LINC00319 may promote OS progression by regulating the miR-455-3p/NFIB axis, which probably serves as an innovative potential indicator of prognosis and a target of therapy for OS.


Subject(s)
Biomarkers, Tumor/metabolism , Bone Neoplasms/pathology , Gene Expression Regulation, Neoplastic , MicroRNAs/genetics , NFI Transcription Factors/metabolism , Osteosarcoma/pathology , RNA, Long Noncoding/genetics , Apoptosis , Biomarkers, Tumor/genetics , Bone Neoplasms/genetics , Bone Neoplasms/metabolism , Bone Neoplasms/surgery , Cell Movement , Cell Proliferation , Disease Progression , Humans , NFI Transcription Factors/genetics , Osteosarcoma/genetics , Osteosarcoma/metabolism , Osteosarcoma/surgery , Prognosis , Tumor Cells, Cultured
2.
Mediators Inflamm ; 2020: 4092762, 2020.
Article in English | MEDLINE | ID: mdl-32908449

ABSTRACT

MicroRNA-155 (miRNA-155) is abundant in fibroblast-like synoviocytes (FLS) in rheumatoid arthritis (RA). Lysine-specific demethylase 1 (LSD1) has been found that it can ameliorate the severity of RA. Tumor necrosis factor-alpha, interleukin-1 beta, and interleukin-6 are key proinflammatory cytokines implicated in the pathogenesis of RA. In our study, we investigated whether miRNA-155 participates in the expression of LSD1 and proinflammatory cytokines in rheumatoid synovial cells. First of all, flow cytometry and cell counting kit-8 analysis were employed to explore the apoptosis and proliferation of FLS, respectively. Subsequently, reverse transcription-quantitative polymerase chain reaction (RT-qPCR) was applied to probe into the level of miRNA-155 in FLS when stimulated by miRNA-155 molecules. Moreover, RT-qPCR was used to explore the relative LSD1 miRNA expression in FLS when stimulated by miRNA-155 molecules, and Western blot and immunofluorescence assay were applied to probe into the expression level of LSD1. Finally, enzyme-linked immunosorbent assay was employed to analyze the secreting level of proinflammatory cytokines in FLS when stimulated by miRNA-155 molecules. RA-FLS showed a higher apoptosis rate than normal FLS. The cell proliferation of both HFLS and MH7A cells was promoted by miRNA-155 upregulation. Meanwhile, the expression of LSD1 and proinflammatory cytokines in the FLS of RA was also changed by miRNA-155 regulation. In conclusion, miRNA-155 participates in the expression of LSD1 and proinflammatory cytokines in rheumatoid synovial cells. These findings imply a potential function and interaction of miRNA-155 and LSD1.


Subject(s)
Cytokines/metabolism , Histone Demethylases/biosynthesis , MicroRNAs/biosynthesis , Synovial Membrane/metabolism , Synoviocytes/metabolism , Apoptosis , Arthritis, Rheumatoid/metabolism , Cell Line, Tumor , Cell Proliferation , Cells, Cultured , Enzyme-Linked Immunosorbent Assay , Fibroblasts/metabolism , Flow Cytometry , Gene Expression Profiling , Humans , Inflammation , Tumor Necrosis Factor-alpha/metabolism , Up-Regulation
3.
Int J Syst Evol Microbiol ; 69(8): 2388-2394, 2019 Aug.
Article in English | MEDLINE | ID: mdl-31150324

ABSTRACT

A facultatively anaerobic, Gram-stain-negative, non-motile and straight to slightly curved long rod-shaped bacterial strain, designated XSD2T, was isolated from coastal seawater of Xiaoshi Island, PR China. The cells were catalase-positive, oxidase-negative and non-flagellated. Strain XSD2T was found to grow at 20-40 °C (optimum, 33 °C), at pH 6.0-8.5 (pH 7.0-7.5) and with 1-5 % (w/v) NaCl (3 %). Carotenoid pigments were produced. The major cellular fatty acids (>10.0 %) were iso-C15 : 0, iso-C16 : 0 3-OH and C17 : 1ω6c and the major polar lipids were phosphatidylethanolamine, one unidentified aminolipid and three unidentified polar lipids. The sole respiratory quinone was MK-7 and the genomic DNA G+C content was 44.1 mol%. The result of the 16S rRNA gene sequence analysis confirmed the affiliation of this organism to the order Marinilabiliales, family Prolixibacteraceae, with Mariniphaga sediminis SY21T as its closest relative with only 93.6 % sequence similarity. On the basis of physiological, biochemical and chemotaxonomic characteristics, we propose that strain XSD2T (=KCTC 62994T=MCCC 1H00347T) represents a novel species of a novel genus in the family Prolixibacteraceae, for which the name Maribellus luteus gen. nov., sp. nov. is proposed.


Subject(s)
Bacteroidetes/classification , Phylogeny , Seawater/microbiology , Bacterial Typing Techniques , Bacteroidetes/isolation & purification , Base Composition , China , DNA, Bacterial/genetics , Fatty Acids/chemistry , Islands , Phosphatidylethanolamines/chemistry , Pigmentation , RNA, Ribosomal, 16S/genetics , Sequence Analysis, DNA , Vitamin K 2/analogs & derivatives , Vitamin K 2/chemistry
4.
Immunol Res ; 2024 May 09.
Article in English | MEDLINE | ID: mdl-38722530

ABSTRACT

Rheumatoid arthritis (RA) is a chronic autoimmune disease characterized by synovial hyperplasia, pannus formation, and cartilage and bone destruction. Lysine-specific demethylase 1 (LSD1), an enzyme involved in transcriptional regulation, has an unclear role in synovial inflammation, fibroblast-like synoviocytes migration, and invasion during RA pathogenesis. In this study, we observed increased LSD1 expression in RA synovial tissues and in TNF-α-stimulated MH7A cells. SP2509, an LSD1 antagonist, directly reduced LSD1 expression and reversed the elevated levels of proteins associated with inflammation, apoptosis, proliferation, and autophagy induced by TNF-α. Furthermore, SP2509 inhibited the migratory capacity of MH7A cells, which was enhanced by TNF-α. In CIA models, SP2509 treatment ameliorated RA development, reducing the expression of pro-inflammatory cytokines and alleviating joint pathological symptoms. These findings underscore the significance of LSD1 in RA and propose the therapeutic potential of SP2509.

5.
Exp Ther Med ; 23(2): 148, 2022 Feb.
Article in English | MEDLINE | ID: mdl-35069829

ABSTRACT

Fibroblast-like synoviocytes (FLSs) have functions in the pathogenesis of rheumatoid arthritis (RA) through the onset of synovitis, the growth of pannus and the destruction of cartilage and bone. The significant increase in the proliferation, migration and invasion of FLSs induces the onset and advancement of RA. To date, the exact function of corepressor element-1 silencing transcription factor (CoREST) in RA remains unclear, but its expression has been determined in RA synovial tissues. In this study, the effects of CoREST were investigated in a TNF-α-induced FLS activation model. Following the silencing of CoREST expression with small interfering (si)RNA, the viability and migration of FLSs were evaluated. Furthermore, the possible molecular mechanisms were explored by detecting the expression of key factors, including matrix metalloproteinases (MMPs), lysine-specific histone demethylase 1 (LSD1) and associated cytokines, via reverse transcription-quantitative PCR and western blotting. CoREST expression increased not only in the RA synovial tissues, but also in the TNF-α-induced FLS activation model. Following the silencing of CoREST in the FLSs treated with TNF-α, cell viability was inhibited, and the migratory capacity of FLSs was suppressed, which was accompanied by the reduced expression of MMP-3 and MMP-9. The expression of LSD1 was also downregulated. There was a notable decrease in the synthesis of interferon-γ and interleukin (IL)-17, while IL-10 expression was increased. The knockdown of CoREST inhibited the viability and migration of FLSs stimulated with TNF-α. Thus, the suppression of CoREST may have crucial roles in the occurrence and development of RA.

6.
Bioengineered ; 12(2): 9991-9999, 2021 12.
Article in English | MEDLINE | ID: mdl-34637672

ABSTRACT

Osteosarcoma is known to have a high metastatic potential, which is closely related to angiogenesis. circRNAs are closely associated with osteosarcoma metastasis. This study aims to investigate the role of Circular RNA circFOXP1 in angiogenesis in osteosarcoma. We detected circFOXP1 expression in osteosarcoma, as well as its prognostic value. Tube formation assay and immunohistochemistry staining were conducted to determine the condition of tube formation. RT-qPCR was performed to explore targeted genes. Luciferase reporter assays were carried out to explore the interaction between miR-127-5p, ircFOXP1, and CDKN2AIP, respectively. In vivo studies further confirmed the relationship between circFOXP1 and tumor angiogenesis in osteosarcoma. We found that circFOXP1 expression was increased in osteosarcoma, and could promote angiogenesis in osteosarcoma through upregulating CDKN2AIP expression. Moreover, circFOXP1 could directly bind to miR-127-5p, which further targets CDKN2AIP directly. In conclusion, circFOXP1 promoted angiogenesis by regulating miR-127-5p/CDKN2AIP signaling pathway in osteosarcoma.


Subject(s)
Apoptosis Regulatory Proteins/metabolism , MicroRNAs/metabolism , Neovascularization, Pathologic/genetics , RNA, Circular/metabolism , RNA-Binding Proteins/metabolism , Adult , Aged , Animals , Apoptosis Regulatory Proteins/genetics , Base Sequence , Cell Line, Tumor , Cell Proliferation/genetics , Gene Expression Regulation, Neoplastic , Humans , Mice, Inbred BALB C , Mice, Nude , MicroRNAs/genetics , Middle Aged , Osteosarcoma/genetics , Osteosarcoma/mortality , Osteosarcoma/pathology , RNA, Circular/genetics , RNA-Binding Proteins/genetics , Signal Transduction , Survival Rate
7.
J Orthop Surg Res ; 16(1): 272, 2021 Apr 20.
Article in English | MEDLINE | ID: mdl-33879199

ABSTRACT

BACKGROUND: The purpose of the current study was to explore the role and underlying mechanism of cellular retinoic acid binding protein 2 (CRABP2) in dexamethasone (DEX)-induced apoptosis in human osteoblast cells. METHODS: GSE10311 was downloaded from the Gene Expression Omnibus (GEO) database to identify the differentially expressed genes (DEGs) by the limma/R package. Primary human osteoblast was isolated and treated with different concentration of DEX (0, 10-8, 10-7, 10-6, 10-5, and 10-4 mol/L), and cell viability and flow cytometry were used to detect cell proliferation and apoptosis. A CRABP2 overexpression plasmid (oe-CRABP2) was used to overexpress CRABP2, and western blotting was conducted to detect protein expression. RESULTS: We found that CRABP2 was downregulated in the DEX-treated group. Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway analyses indicated that DEGs were associated with PI3K/Akt signaling pathway. DEX downregulated CRABP2 gene and protein expression, inhibited viability, and induced human osteoblast apoptosis. Overexpression of CRABP2 reversed DEX-induced apoptosis in human osteoblast. Moreover, overexpression of CRABP2 delayed the progression of DEX-induced osteonecrosis of the femoral head (ONFH) animal model. CONCLUSION: In conclusion, CRABP2 is effective at inhibiting DEX-induced human osteoblast apoptosis and delayed ONFH progression.


Subject(s)
Apoptosis/drug effects , Apoptosis/genetics , Dexamethasone/adverse effects , Gene Expression/genetics , Gene Expression/physiology , Glucocorticoids/adverse effects , Osteoblasts/physiology , Receptors, Retinoic Acid/genetics , Receptors, Retinoic Acid/physiology , Cell Survival/drug effects , Cell Survival/genetics , Cells, Cultured , Down-Regulation/drug effects , Down-Regulation/genetics , Femur Head Necrosis/chemically induced , Femur Head Necrosis/genetics , Femur Head Necrosis/pathology , Humans , Phosphatidylinositol 3-Kinases/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Receptors, Retinoic Acid/metabolism , Signal Transduction/genetics , Signal Transduction/physiology
8.
J Orthop Surg Res ; 15(1): 162, 2020 Apr 25.
Article in English | MEDLINE | ID: mdl-32334633

ABSTRACT

BACKGROUND: This article reports the effects of proenkephalin (PENK) on osteosarcoma (OS) cell migration. METHODS: A Gene Expression Omnibus (GEO) dataset was used to identify differentially expressed genes (DEGs) in OS tumor samples and normal human osteoblasts. Tumor tissue and adjacent normal tissue were collected from 40 OS patients. MG63 cells were transfected with si-PENK. Transwell migration assays and wound healing assays were performed to compare the effect of PENK on migration. Moreover, LY294002 was used to identify the potential mechanism. Gene expression was examined via qRT-PCR and Western blotting. RESULTS: Bioinformatic analysis revealed that PENK was downregulated in OS tumor samples compared with normal human osteoblasts. Moreover, PENK was identified as the hub gene of the DEGs. The PI3K/Akt signaling pathway was significantly enriched in the DEGs. Moreover, PENK was downregulated in OS and MG63 cells compared with the corresponding control cells. Silencing PENK promoted MG63 cell migration; however, treatment with LY294002 partially attenuated PENK silencing-induced OS cell migration. CONCLUSION: PENK inhibits OS cell migration by activating the PI3K/Akt signaling pathway.


Subject(s)
Bone Neoplasms/metabolism , Cell Movement , Enkephalins/physiology , Osteosarcoma/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Protein Precursors/physiology , Proto-Oncogene Proteins c-akt/metabolism , Signal Transduction , Adult , Blotting, Western , Bone Neoplasms/pathology , Cell Line, Tumor , Cell Movement/physiology , Enkephalins/metabolism , Female , Gene Expression Regulation, Neoplastic , Humans , Male , Osteoblasts/metabolism , Osteosarcoma/pathology , Protein Precursors/metabolism , Real-Time Polymerase Chain Reaction , Young Adult
SELECTION OF CITATIONS
SEARCH DETAIL