Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 9 de 9
Filter
Add more filters










Database
Language
Publication year range
1.
Orthop Surg ; 15(4): 1060-1071, 2023 Apr.
Article in English | MEDLINE | ID: mdl-36782343

ABSTRACT

OBJECTIVE: Osteosarcoma (OS) is regarded as one of the most common malignant bone tumors, mainly occurring in children and adolescents with high mortality. The dysregulation of lncRNAs is reported to regulate tumor development and be closely related to patient prognosis. Nevertheless, the role of long noncoding RNAs (lncRNAs) prostate-specific transcript 1 (PCGEM1) in OS remains uncharacterized. The current study aimed to explore the role of PCGEM1 in OS. METHODS: Reverse transcription-quantitative polymerase chain reaction (RT-qPCR) was performed to examine the expression of PCGEM1 in OS cell lines. CCK-8, colony formation, Transwell, and western blotting analyses were applied to measure OS cell viability, proliferation, migration, invasion, and epithelial-mesenchymal transition (EMT) after PCGEM1 downregulation. Nuclear-cytoplasmic fractionation, RNA pulldown, RNA immunoprecipitation (RIP), luciferase reporter assays were performed to verify the relationship among PCGEM1, miR-433-3p. and OMA1 in OS. The xenograft tumor models were established to evaluate the effect of PCGEM1 on tumor growth of OS. RESULTS: In this study, we discovered that PCGEM1 knockdown inhibited cell proliferation, migration, invasion and EMT in OS (P < 0.05). Additionally, PCGEM1 directly bound to miR-433-3p (P < 0.01). OMA1 was confirmed to be a target gene of miR-433-3p (P < 0.05), positively regulated by PCGEM1 but negatively regulated by miR-433-3p. Rescue assays further verified that overexpression of OMA1 reversed the PCGEM1 knockdown-mediated inhibitory effect on the malignant phenotype in OS cells (P < 0.05). Moreover, knockdown of PCGEM1 inhibited tumor growth and metastasis in vivo (P < 0.05). CONCLUSIONS: Overall, PCGEM1 mediated tumor growth and metastasis of OS by sponging miR-433-3p and regulating OMA1, which might provide an innovative strategy for OS diagnosis or treatment.


Subject(s)
Bone Neoplasms , Metalloendopeptidases , MicroRNAs , Osteosarcoma , RNA, Long Noncoding , Humans , Male , Bone Neoplasms/genetics , Bone Neoplasms/pathology , Cell Line, Tumor , Cell Movement , Cell Proliferation , Gene Expression Regulation, Neoplastic , MicroRNAs/genetics , MicroRNAs/metabolism , Osteosarcoma/genetics , Osteosarcoma/pathology , RNA, Long Noncoding/genetics , RNA, Long Noncoding/metabolism , Metalloendopeptidases/metabolism
2.
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
3.
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
4.
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
5.
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
6.
Neuroreport ; 30(15): 1016-1024, 2019 10 16.
Article in English | MEDLINE | ID: mdl-31503208

ABSTRACT

3,4-Dihydroxyphenylethanol (DOPET) is a potent antioxidant polyphenolic compound. In this study, our objective was to investigate the underlying mechanism of the neuroprotective role of DOPET in attenuating spinal cord injury (SCI). Initially, SCI was induced by performing surgical laminectomy on the rats at T10-T12 level. Then, the neurological function-dependent locomotion was measured using Basso Beattie Bresnahan score, which declined in the SCI-induced group. Increased antioxidant levels such as superoxide dismutase, glutathione peroxidase, and glutathione along with other parameters such as increased lipid peroxidation (LPO) and myeloperoxidase (MPO) activities were all observed in the SCI group. Levels of proinflammatory cytokines such as tumor necrosis factor-α and interleukin-1ß were upregulated in the serum and spinal cord tissue as observed on the immunoblot. Interestingly, protein levels of apoptotic markers such as Bax, cleaved caspase 3 and RT-PCR analysis-based mRNA level of pro-inflammatory cytokine, nuclear factor- κ activated B cells (NF-κB) were significantly upregulated in the spinal cord tissue. Nonetheless, antiapoptotic factor such as B-cell lymphoma 2 (Bcl-2) protein expression was downregulated in the same group. However, on administering 10 mg/kg of DOPET, the neuronal function was rescued, antioxidants were restored back to the normal levels, LPO and MPO activities were reduced in conjunction with downregulated levels of proinflammatory cytokines and apoptotic markers in the SCI group. These findings show that DOPET could potentially target multiple signalling pathways to combat SCI.


Subject(s)
Antioxidants/therapeutic use , Inflammation/pathology , Inflammation/prevention & control , Oxidative Stress/drug effects , Phenylethyl Alcohol/analogs & derivatives , Spinal Cord Injuries/pathology , Animals , Antioxidants/metabolism , Apoptosis Regulatory Proteins/metabolism , Cytokines/metabolism , Lipid Peroxidation/drug effects , Locomotion , Male , Peroxidase/metabolism , Phenylethyl Alcohol/therapeutic use , Rats , Rats, Sprague-Dawley , Signal Transduction/drug effects , Spinal Cord Injuries/metabolism
7.
Oncol Res ; 26(4): 655-664, 2018 May 07.
Article in English | MEDLINE | ID: mdl-29191257

ABSTRACT

Although cisplatin has been shown to be an integral part of chemotherapy regimen in osteosarcoma (OS) treatment, toxicity issues and chemoresistance have hindered therapeutic development for OS. Exploring novel combination therapy methods is needed to circumvent the limitations of cisplatin alone. The proteasome inhibitor MG132 has shown antitumor effects in many solid tumors. However, little is known about its effects in combination with cisplatin in OS cells. In this study, we examined the effects of MG132 in combination with cisplatin in human OS cells (MG-63 and HOS). MG132 and cisplatin were applied to OS cells, respectively or jointly. The results demonstrated that MG132 markedly inhibited cell viability in a dose- and time-dependent manner, whereas viability of osteoblast cells was not affected, suggesting a selective toxicity of MG132 to cancerous cells. Mechanistically, MG132 arrested cells in the G2/M phase in association with increased p21waf1 and induced cell apoptosis, which was accompanied by cleaved PARP. In addition to its apoptotic effect alone, MG132 significantly enhanced cisplatin-induced apoptosis in OS cells. Furthermore, cell viability of the combined application of 10 µM MG132 and 5 µg/ml cisplatin was markedly inhibited compared to that of the individual application. These events were accompanied by the downregulation of NF-κB, mitochondrial antiapoptotic protein Bcl-xL, and PI3K/Akt, which play a key role in cell survival. Finally, combination treatment of MG132 and cisplatin showed more antiproliferative effect than the single treatment in OS xenograft models. In summary, we concluded that MG132 interacted synergistically with cisplatin, which raised the possibility that combining the two drugs may represent a novel strategy in OS.


Subject(s)
Antineoplastic Agents/pharmacology , Cell Proliferation/drug effects , Cisplatin/pharmacology , Leupeptins/pharmacology , Proteasome Inhibitors/pharmacology , Animals , Antineoplastic Agents/administration & dosage , Bone Neoplasms/drug therapy , Bone Neoplasms/metabolism , Bone Neoplasms/pathology , Cell Line, Tumor , Cell Survival/drug effects , Cisplatin/administration & dosage , Cisplatin/therapeutic use , Dose-Response Relationship, Drug , Drug Synergism , Humans , Leupeptins/administration & dosage , Leupeptins/therapeutic use , Mice, Inbred BALB C , Mice, Nude , Osteosarcoma/drug therapy , Osteosarcoma/metabolism , Osteosarcoma/pathology , Proteasome Inhibitors/administration & dosage , Xenograft Model Antitumor Assays
8.
Chin Med J (Engl) ; 123(21): 3067-73, 2010 Nov.
Article in English | MEDLINE | ID: mdl-21162957

ABSTRACT

BACKGROUND: There are few reports of a biological role for glycosyltransferases in the infiltration of osteoarthritic synovitis. The aim of this research was to investigate the expression and cellular location of ß-1,4-galactosyltransferase I (ß-1,4-GalT-I) in a surgically-induced rat model of knee osteoarthritis (OA), and explore the role of ß-1,4-GalT-I in the pathogenesis of OA. METHODS: Male Sprague-Dawley rats were randomly divided into three groups: OA group, sham group and normal group. The model of OA was established in the right knees of rats by anterior cruciate ligament transaction (ACLT) with partial medial meniscectomy. Fibroblast-like synoviocytes (FLSs) obtained from normal rat synovial tissue were cultured. The expression of ß-1,4-GalT-I mRNA in the synovial tissue, articular cartilage and FLSs treated with tumor necrosis factor-α (TNF-α) were assayed by real-time PCR. Western-blotting and immunohistochemisty were used to observe the expression of ß-1,4-GalT-I at the protein level. Double immunofluorescent staining was used to define the location of the ß-1,4-GalT-I with macrophage-like synoviocytes, FLSs, neutrophils, and TNF-α in the OA synovium. The alteration of TNF-α in FLSs which were treated with lipopolysaccharide (LPS) and ß-1,4-GalT-I-Ab were detected by enzyme-linked immunosorbent assay (ELISA). RESULTS: The mRNA and protein expression of ß-1,4-GalT-I increased in synovial tissue of the OA group compared with the normal and sham groups at two and four weeks after the surgery, however, no significant difference appeared in the articular cartilage. Immunohistochemistry also indicated that the ß-1,4-GalT-I expression in OA synovium at four weeks after surgery increased sharply compared with the control group. ß-1,4-GalT-I co-localized with macrophage-like synoviocytes, FLSs, neutrophils and TNF-α in rat OA synovitis. Moreover, in vitro ß-1,4-GalT-I mRNA in FLSs was affected in a dose- and time-dependent manner in response to TNF-α stimulation. ELISA revealed that the expression of TNF-α was attenuated in FLSs in vitro when treated with anti ß-1,4-GalT-I antibody. CONCLUSION: ß-1,4-GalT-I may play an important role in the inflammation process of rat OA synovial tissue which would provide the foundation for further researching into the concrete mechanism of ß-1,4-GalT-I in OA synovitis.


Subject(s)
Galactosyltransferases/metabolism , Knee Joint/enzymology , Osteoarthritis, Knee/enzymology , Synovitis/enzymology , Animals , Blotting, Western , Cells, Cultured , Enzyme-Linked Immunosorbent Assay , Galactosyltransferases/genetics , Immunohistochemistry , Knee Joint/pathology , Knee Joint/surgery , Male , Osteoarthritis, Knee/genetics , Osteoarthritis, Knee/pathology , Polymerase Chain Reaction , Rats , Rats, Sprague-Dawley , Synovial Membrane/enzymology , Synovitis/etiology
SELECTION OF CITATIONS
SEARCH DETAIL
...