Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 7 de 7
Filter
Add more filters










Database
Language
Publication year range
1.
Cell Death Dis ; 12(1): 59, 2021 01 11.
Article in English | MEDLINE | ID: mdl-33431838

ABSTRACT

Small nucleolar RNA host gene 6 (SNHG6) is a newly discovered long non-coding RNA (lncRNA), while the regulatory mechanism of SNHG6 in chondrosarcoma is largely unknown. Here we found that SNHG6 expression was upregulated and showed positive correlation with the progression of chondrosarcoma. Functional assays demonstrated that SNHG6 was required for the proliferation, migration, and invasion of chondrosarcoma cells. Mechanistic study revealed that SNHG6 could recruit EZH2 and maintain high level of H3K27me3 to repress the transcription of tumor-suppressor genes, including KLF6. KLF6 was found to bind to the promoter region of SP1 and restrained its transcription, while SP1 could be recruited to the promoter region of SNHG6 and promoted its transcription to form a positive loop. In summary, this study reveals that SP1-induced SNHG6 forms a positive loop to facilitate the carcinogenesis of chondrosarcoma through the suppression of KLF6 by recruiting EZH2, which manifests the oncogenic function of SNHG6 in chondrosarcoma.


Subject(s)
Bone Neoplasms/metabolism , Chondrosarcoma/metabolism , Enhancer of Zeste Homolog 2 Protein/metabolism , Kruppel-Like Factor 6/metabolism , RNA, Long Noncoding/metabolism , Sp1 Transcription Factor/metabolism , Animals , Bone Neoplasms/genetics , Cell Line, Tumor , Cell Movement/physiology , Cell Proliferation/physiology , Chondrosarcoma/genetics , Enhancer of Zeste Homolog 2 Protein/genetics , Humans , Kruppel-Like Factor 6/genetics , Male , Mice , Mice, Inbred BALB C , Mice, Nude , RNA, Long Noncoding/genetics , Sp1 Transcription Factor/genetics , Transfection
2.
BMC Cancer ; 19(1): 638, 2019 Jun 28.
Article in English | MEDLINE | ID: mdl-31253134

ABSTRACT

BACKGROUND: The optimal reconstructive method after diaphyseal malignant bone tumor resection remains controversial. This multicenter clinical study was designed to investigate the clinical value and complications of segmental prosthesis in the repair of diaphyseal defects. METHODS: We present 49 patients from three clinical centers treated with wide resection for primary or metastatic bone tumors involving the diaphysis of the femur, tibia, humerus, or ulna, followed by reconstruction using a modular intramedullary segmental prosthesis. RESULTS: Enrolled patients included 23 men and 26 women with a mean age of 63.3 years. Of these, seven patients had primary bone tumors and 42 patients had metastatic lesions. At the mean follow-up of 13.7 months, 17 patients were alive, 31 patients were deceased due to tumor progression, and one patient was dead of another reason. There were eight nononcologic complications (two with radial nerve injury, three with delayed incision healing, two with aseptic loosening in the proximal humerus prosthetic stem and one with structural failure) and three oncologic complications (three with primary tumor recurrence) among all patients. The incidence of complications in primary tumor patients (4/7, 57.1%) was higher than that in patients with metastatic tumors (7/42, 16.7%) (p = 0.036). Aseptic loosening and mechanical complications were not common for patients with primary tumors, although the reconstruction length difference was statistically significant (p = 0.023). No statistically significant differences were observed in limb function, while the mean musculoskeletal tumor society score was 21.2 in femora, 19.6 in humeri, and 17.8 in tibiae (p = 0.134). CONCLUSIONS: Segmental prostheses represent an optional method for the reconstruction of diaphyseal defects in patients with limited life expectancy. Segmental prostheses in the humerus experienced more complications than those used to treat lesions in the femur.


Subject(s)
Bone Neoplasms/surgery , Diaphyses/surgery , Plastic Surgery Procedures/methods , Prostheses and Implants , Adolescent , Adult , Aged , Aged, 80 and over , Bone Neoplasms/mortality , Bone Neoplasms/pathology , Diaphyses/pathology , Female , Humans , Male , Middle Aged , Prostheses and Implants/adverse effects , Prosthesis Failure , Prosthesis Implantation , Plastic Surgery Procedures/adverse effects , Retrospective Studies , Treatment Outcome , Young Adult
3.
Chem Biol Interact ; 296: 1-8, 2018 Dec 25.
Article in English | MEDLINE | ID: mdl-30125549

ABSTRACT

BACKGROUD/AIMS: Abnormal activation of the Wnt/ß-catenin signaling, which may be antagonized by the members of secreted frizzled-related proteins family (SFRPs), is implicated in tumor occurrence and development. However, the function of SFRP5 relating to Wnt/ß-catenin pathway in chondrosarcoma is not clear yet. This study was undertaken to investigate the potential role of SFRP5 promoter methylation in chondrosarcoma metastasis and invasion through activating canonical Wnt signaling pathway. METHODS AND RESULTS: The results demonstrated that SFRP5 promoter was hypermethylated and SFRP5 expression was significantly reduced in chondrosarcoma cell lines at the mRNA and protein levels. The canonical Wnt/ß-catenin signaling was observably activated with ß-catenin stabilization by dephosphorylation and translocation into the nuclear. 5-Aza-2'-deoxycytidine (5-Aza-dC), the DNA methyltransferase inhibitor, significantly inhibited the proliferation of chondrosarcoma cells by cell cycle arrest through repressing the methylation of SFRP5 and promoting its expression. Both 5-Aza-dC treatment and SFRP5 overexpression could significantly inhibited the metastasis and invasion of chondrosarcoma cells by inactivating Wnt/ß-catenin signaling pathway and promoting chondrosarcoma cells mesenchymal-epithelial transition (MET). 5-Aza-dC also inhibited the xenograft growth and lung metastasis of chondrosarcoma cells in vivo via suppressing SFRP5 promotor methylation, inactivating Wnt/ß-catenin pathway and inducing epithelial markers expression. CONCLUSION: All of our results revealed the epigenetic silencing of SFRP5 by promoter methylation plays pivotal roles in chondrosarcoma development and metastasis through SFRP5/Wnt/ß-catenin signaling axis. Modulation of their levels may serve as potential targets and diagnostic tools for novel therapeutic strategies of chondrosarcoma.


Subject(s)
Chondrosarcoma/genetics , Chondrosarcoma/pathology , Epigenesis, Genetic/genetics , Eye Proteins/genetics , Membrane Proteins/genetics , Neoplasm Invasiveness/genetics , Neoplasm Metastasis/genetics , Wnt Signaling Pathway/genetics , Adaptor Proteins, Signal Transducing , Animals , Antimetabolites, Antineoplastic/chemistry , Antimetabolites, Antineoplastic/pharmacology , Azacitidine/analogs & derivatives , Azacitidine/chemistry , Azacitidine/pharmacology , Cells, Cultured , Chondrosarcoma/drug therapy , Chondrosarcoma/metabolism , Dose-Response Relationship, Drug , Drug Screening Assays, Antitumor , Epigenesis, Genetic/drug effects , Eye Proteins/antagonists & inhibitors , Eye Proteins/metabolism , Humans , Membrane Proteins/antagonists & inhibitors , Membrane Proteins/metabolism , Mice , Mice, Inbred BALB C , Mice, Nude , Neoplasms, Experimental/drug therapy , Neoplasms, Experimental/genetics , Neoplasms, Experimental/metabolism , Structure-Activity Relationship , Wnt Signaling Pathway/drug effects
4.
Oncotarget ; 8(31): 50724-50730, 2017 Aug 01.
Article in English | MEDLINE | ID: mdl-28881598

ABSTRACT

Giant cell tumor of bone (GCT), which frequently occurs in the patients' spine, is relatively prevalent in Chinese population. A group of GCT invades into vessels and appears to be circulating tumor cells (CTCs) responsible for the distal metastasis of the primary tumor. So far the cell surface markers of GCT have not been determined. In the current study, we aimed to identify a novel CTC marker with higher specificity in GCT. TRAIL-R1+ cells were purified from GCT cell lines. The TRAIL-R1+ cells were compared with total GCT cells for tumor sphere formation, chemo-resistance, tumor formation in nude mice, and frequency of developing distal metastases. We found that TRAIL-R1+ GCT cells appeared to be highly enriched for CTCs in GCT. Compared to total GCT cells, TRAIL-R1+ GCT cells generated significantly more tumor spheres in culture, were higher chemo-resistant, and had a higher frequency of being detected in the circulation after subcutaneous transplantation as well as development of distal metastases. Thus, we conclude that TRAIL-R1+ may be a novel CTC marker in GCT. Selective elimination of TRAIL-R1+ GCT cells may improve the current GCT therapy.

5.
Life Sci ; 189: 76-83, 2017 Nov 15.
Article in English | MEDLINE | ID: mdl-28942283

ABSTRACT

AIMS: Edaravone is a strong free radical scavenger most used for treating acute ischemic stroke. In this study we investigated the protective effects and underlying mechanisms of edaravone on compression-induced damage in rat nucleus pulposus (NP) cells. MATERIALS AND METHODS: Cell viability was determined using MTT assay methods. NP cell apoptosis was measured by Hoechst 33,258 staining and Annexin V/PI double staining. Intracellular reactive oxygen species (ROS), mitochondrial membrane potential (MMP), and intracellular calcium ([Ca2+]i) were determined by fluorescent probes DCFH-DA, JC-1 and Fluo-3/AM, respectively. Apoptosis-related proteins (cleaved caspase-3, cytosolic cytochrome c, Bax and Bcl-2) and extracellular matrix proteins (aggrecan and collagen II) were analyzed by western blot. KEY FINDINGS: Edaravone attenuated the compression-induced decrease in viability of NP cells in a dose-dependent manner. 33,258 and Annexin V/PI double staining showed that edaravone protected NP cells from compression-induced apoptosis. Further studies confirmed that edaravone protected NP cells against compression-induced mitochondrial pathway of apoptosis by inhibiting overproduction of ROS, collapse of MMP and overload of [Ca2+]i. In addition, edaravone promoted the expression of aggrecan and collagen II in compression-treated NP cells. SIGNIFICANCE: These results strongly indicate that edaravone ameliorates compression-induced damage in rat nucleus pulposus cells. Edaravone could be a potential new drug for treatment of IDD.


Subject(s)
Antipyrine/analogs & derivatives , Apoptosis/drug effects , Cell Survival/drug effects , Free Radical Scavengers/pharmacology , Nucleus Pulposus/drug effects , Aggrecans/genetics , Animals , Antipyrine/administration & dosage , Antipyrine/pharmacology , Cells, Cultured , Collagen Type II/genetics , Dose-Response Relationship, Drug , Edaravone , Free Radical Scavengers/administration & dosage , Gene Expression Regulation/drug effects , Intervertebral Disc Degeneration/drug therapy , Intervertebral Disc Degeneration/physiopathology , Membrane Potential, Mitochondrial , Nucleus Pulposus/pathology , Rats , Rats, Sprague-Dawley , Reactive Oxygen Species/metabolism
6.
Medicine (Baltimore) ; 96(27): e7245, 2017 Jul.
Article in English | MEDLINE | ID: mdl-28682874

ABSTRACT

BACKGROUND: The aim of the study was to explore the effects of microRNA-107 (miR-107) by targeting Dkk-1 on osteosarcoma (OS) via the Wnt/ß-catenin signaling pathway. METHODS: OS and adjacent tissues were collected from 67 patients diagnosed with OS. Expressions of miR-107, Dkk-1, LRP5, ß-catenin, and c-Myc were detected by the quantitative real-time polymerase chain reaction (qRT-PCR) and Western blotting. The dual-luciferase reporter gene assay was performed to observe the relationship between miR-107 and Dkk-1.Transfected cells were divided into different investigating groups designated as Inhibitor, Mimic, siRNA, Inhibitor + siRNA, negative control (NC), and blank groups. qRT-PCR and Western blotting were used to detect expressions of miR-107, Dkk-1, ß-catenin, Bcl-2, c-Myc, Caspase-3, and PARP. Cell counting kit-8 (CCK-8), flow cytometry (FCM), colony-formation efficiency (CFE), and subcutaneous tumorigenicity assays were all utilized for to determine cell proliferation, apoptosis, colony-forming, and tumorigenic abilities. RESULTS: Dkk-1 is the target gene of miR-107. Decreased expressions of miR-107, LRP5, ß-catenin, and c-Myc, and increased expressions of Dkk-1 were found in OS tissues. The Mimic and siRNA groups exhibited decreased proliferation rates, colony-forming abilities, and tumorigenicity and increased apoptosis rates, whereas the inhibitor group showed opposite trends when compared to the blank group. On the other hand, expressions of miR-107, LRP5, ß-catenin, c-Myc, Caspase-3, and PARP were all elevated in the mimic group, whereas expressions of Dkk-1 and Bcl-2 were reduced; opposite trends were observed in the inhibitor group. CONCLUSION: We conclude that miR-107 is likely to inhibit the occurrence and development of OS by down-regulating Dkk-1 via the Wnt/ß-catenin signaling pathway, providing us with a new therapeutic target for the treatment of OS.


Subject(s)
Intercellular Signaling Peptides and Proteins/metabolism , MicroRNAs/metabolism , Osteosarcoma/metabolism , Wnt Signaling Pathway/physiology , Animals , Apoptosis/physiology , Caspase 3/metabolism , Cell Proliferation/physiology , Cells, Cultured , Humans , Low Density Lipoprotein Receptor-Related Protein-5/metabolism , Mice, Nude , MicroRNAs/antagonists & inhibitors , MicroRNAs/genetics , Neoplasm Transplantation , Poly(ADP-ribose) Polymerases/metabolism , Proto-Oncogene Proteins c-bcl-2/metabolism , Proto-Oncogene Proteins c-myc/metabolism , RNA, Small Interfering , Random Allocation , Wnt Proteins/metabolism , beta Catenin/metabolism
7.
Biochem Biophys Res Commun ; 487(1): 181-188, 2017 May 20.
Article in English | MEDLINE | ID: mdl-28411026

ABSTRACT

Compression-induced programmed cell death of nucleus pulposus (NP) cells is an important contributor to intervertebral disc degeneration (IDD). Dynamin-related protein 1 (Drp1), a crucial mitochondrial fission protein, triggers programmed necrosis upon cellular injury. However, limited information is available about the role of Drp1 in compression-induced programmed necrosis of NP cells. In the present study, we found that compression resulted in upregulation and mitochondrial translocation of Drp1. Inhibition of Drp1 by siRNA or mitochondrial division inhibitor 1 (mdivi-1) effectively prevented the programmed necrosis of NP cells treated with compression. Furthermore, Drp1 promoted mitochondrial translocation of p53 and nuclear translocation of apoptosis-inducing factor (AIF) in compression-treated NP cells. Inhibition of p53 mitochondrial translocation by pifithrin-µ (PFT-µ) and silencing of AIF expression by siRNA significantly alleviated compression-induced NP cell programmed necrosis. These data indicates that Drp1 mediates compression-induced programmed necrosis of NP cells by promoting mitochondrial translocation of p53 and nuclear translocation of AIF.


Subject(s)
Apoptosis Inducing Factor/metabolism , Dynamins/metabolism , Mitochondria/metabolism , Nucleus Pulposus/metabolism , Nucleus Pulposus/pathology , Tumor Suppressor Protein p53/metabolism , Animals , Cell Nucleus/metabolism , Cells, Cultured , Compressive Strength , Intervertebral Disc Degeneration/metabolism , Intervertebral Disc Degeneration/pathology , Necrosis/metabolism , Necrosis/pathology , Protein Transport , Rats , Rats, Sprague-Dawley , Stress, Mechanical
SELECTION OF CITATIONS
SEARCH DETAIL
...