Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 79
Filtrar
Mais filtros

País/Região como assunto
Tipo de documento
País de afiliação
Intervalo de ano de publicação
1.
Mol Med ; 29(1): 133, 2023 10 03.
Artigo em Inglês | MEDLINE | ID: mdl-37789274

RESUMO

BACKGROUND: Rab-interacting lysosomal protein (RILP) contains an alpha-helical coil with an unexplored biological function in osteosarcoma. This study investigated the expression of RILP in osteosarcoma cells and tissues to determine the effect of RILP on the biological behaviors of osteosarcoma cells and the underlying mechanism. METHODS: Tumor Immune Estimation Resource (TIMER) database, The Cancer Genome Atlas (TCGA) database and Gene Expression Omnibus (GEO) database were used for bioinformatic analysis. Co-immunoprecipitation experiment was used to determine whether the two proteins were interacting. In functional tests, cell counting kit-8 (CCK-8) assay, colony formation assay, wound healing assay, transwell invasion assay, Immunofluorescence (IF) assay and immunohistochemical (IHC) assay were performed. RESULTS: Overexpression of RILP significantly inhibited proliferation and impaired metastasis ability of osteosarcoma cells, while silencing of RILP showed the opposite trend. RNA-seq data analysis was applied in 143B cells and pathway enrichment analysis revealed that differentially expressed genes were mainly enriched in the PI3K/AKT pathway. We further verified that overexpression of RILP restrained the PI3K/AKT/mTOR signaling pathway and induced autophagy in osteosarcoma cells, while the opposite trend was observed when PI3K pathway activator 740Y-P was used. 3-Methyladenine (3-MA), a selective autophagy inhibitor, partially attenuated the inhibitory effect of RILP on the migration and invasion ability of osteosarcoma cells, suggesting the involvement of autophagy in epithelial-mesenchymal transition regulation in osteosarcoma cells. Growth factor receptor binding protein-10 (Grb10), an adaptor protein, was confirmed as a potential target of RILP to restrain the PI3K/AKT signaling pathway. We subcutaneously injected stably overexpressing 143B osteosarcoma cells into nude mice and observed that overexpression of RILP inhibited tumor growth by inhibiting the PI3K/AKT/mTOR pathway. CONCLUSION: Our study revealed that the expression of RILP was associated with favorable prognosis of osteosarcoma and RILP inhibits proliferation, migration, and invasion and promotes autophagy in osteosarcoma cells via Grb10-mediated inhibition of the PI3K/AKT/mTOR signaling pathway. In the future, targeting RILP may be a potential strategy for osteosarcoma treatment.


Assuntos
Neoplasias Ósseas , Osteossarcoma , Animais , Camundongos , Apoptose , Neoplasias Ósseas/metabolismo , Linhagem Celular Tumoral , Movimento Celular , Proliferação de Células , Proteína Adaptadora GRB10/metabolismo , Proteína Adaptadora GRB10/farmacologia , Camundongos Nus , Osteossarcoma/metabolismo , Fosfatidilinositol 3-Quinases/metabolismo , Proteínas Proto-Oncogênicas c-akt/metabolismo , Serina-Treonina Quinases TOR/metabolismo , Humanos
2.
BMC Cancer ; 23(1): 1179, 2023 Dec 01.
Artigo em Inglês | MEDLINE | ID: mdl-38041020

RESUMO

BACKGROUND: Osteosarcoma (OS) is the most common primary malignancy of bone tumors. More and more ARHGAP family genes have been confirmed are to the occurrence, development, and invasion of tumors. However, its significance in osteosarcoma remains unclear. In this study, we aimed to identify the relationship between ARHGAP family genes and prognosis in patients with OS. METHODS: OS samples were retrieved from the TCGA and GEO databases. We then performed LASSO regression analysis and multivariate COX regression analysis to select ARHGAP family genes to construct a risk prognosis model. We then validated this prognostic model. We utilized ESTIMATE and CIBERSORT algorithms to calculate the stroma and immune scores of samples, as well as the proportions of tumor infiltrating immune cells (TICs). Finally, we conducted in vivo and in vitro experiments to investigate the effect of ARHGAP28 on osteosarcoma. RESULTS: We selected five genes to construct a risk prognosis model. Patients were divided into high- and low-risk groups and the survival time of the high-risk group was lower than that of the low-risk group. The high-risk group in the prognosis model constructed had relatively poor immune function. GSEA and ssGSEA showed that the low-risk group had abundant immune pathway infiltration. The overexpression of ARHGAP28 can inhibit the proliferation, migration, and invasion of osteosarcoma cells and tumor growth in mice, and IHC showed that overexpression of ARHGAP28 could inhibit the proliferation of tumor cells. CONCLUSION: We constructed a risk prognostic model based on five ARHGAP family genes, which can predict the overall survival of patients with osteosarcoma, to better assist us in clinical decision-making and individualized treatment.


Assuntos
Neoplasias Ósseas , Osteossarcoma , Humanos , Animais , Camundongos , Prognóstico , Osteossarcoma/genética , Fatores de Risco , Algoritmos , Neoplasias Ósseas/genética
3.
Int J Clin Oncol ; 27(12): 1891-1903, 2022 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-36269529

RESUMO

BACKGROUND: Multiple studies have revealed that long non-coding RNA (lncRNA) NR2F2-AS1 plays a role in affecting cancer cell proliferation and metastasis. Here, both in vitro and in vivo experiments were performed for investigating the function and mechanism of NR2F2-AS1 in human osteosarcoma (OS). METHODS: The NR2F2-AS1 level in human OS tissues and adjacent non-tumor tissues was examined by quantitative reverse transcription-polymerase chain reaction (qRT-PCR). The NR2F2-AS1 overexpression model was constructed in OS cells, then cell proliferation, invasion, and apoptosis were monitored. The OS xenograft model was established in nude mice using NR2F2-AS1-overexpressed OS cells. The downstream target genes of NR2F2-AS1 were predicted. qRT-PCR and Western blot were implemented to validate the profiles of miR-425-5p and HMGB2. The targeting link between NR2F2-AS1 and miR-425-5p, miR-425-5p and HMGB2 was further probed by dual-luciferase reporter experiment. RESULTS: In comparison to adjacent non-tumor tissues, OS tissues showed upregulated NR2F2-AS1 expression. Higher NR2F2-AS1 level was predominantly correlated with worse clinical stages. In vivo and in vitro tests corroborated that NR2F2-AS1 overexpression spurred OS cell proliferation, growth, invasion, and choked apoptosis. Mechanistically, NR2F2-AS1 hampered miR-425-5p expression as its competitive endogenous RNA (ceRNA). Thus, NR2F2-AS1 facilitated the HMGB2 expression. However, miR-425-5p inhibited HMGB2 expression by targeting the latter. CONCLUSION: NR2F2-AS1 expedited the evolution of OS by elevating HMGB2 levels through sponging miR-425-5p. The NR2F2-AS1/miR-425-5p/HMGB2 regulatory axis is a promising target in treating human OS.


Assuntos
Neoplasias Ósseas , Proteína HMGB2 , MicroRNAs , Osteossarcoma , RNA Longo não Codificante , Animais , Humanos , Camundongos , Neoplasias Ósseas/patologia , Linhagem Celular Tumoral , Movimento Celular/genética , Proliferação de Células/genética , Fator II de Transcrição COUP/genética , Fator II de Transcrição COUP/metabolismo , Regulação Neoplásica da Expressão Gênica/genética , Proteína HMGB2/genética , Proteína HMGB2/metabolismo , Camundongos Nus , MicroRNAs/genética , MicroRNAs/metabolismo , Osteossarcoma/genética , Osteossarcoma/patologia , RNA Longo não Codificante/genética , Fatores de Transcrição/genética
4.
Biochem Biophys Res Commun ; 521(4): 880-886, 2020 01 22.
Artigo em Inglês | MEDLINE | ID: mdl-31708095

RESUMO

The side effects of chemotherapy, drug resistance, and tumor metastasis hinder the development of treatment for osteosarcoma, leading to poor prognosis of patients with the disease. Proscillaridin A, a kind of cardiac glycoside, has been proven to have anti-proliferative properties in many malignant tumors, but the efficacy of the drug in treating osteosarcoma is unclear. In the present study, we assessed the effects of Proscillaridin A on osteosarcoma and investigated its underlying action mechanism. The cell cytotoxicity assay showed that Proscillaridin A significantly inhibited the proliferation of 143B cells in a dose- and time-dependent manner. Also, flow cytometry and invasion assay revealed that Proscillaridin A induced apoptosis and reduced 143B cell motility. Western blotting and PCR were used to detect the expressions of Bcl-xl and MMP2 and showed that mRNA/protein expression levels decreased significantly in Proscillaridin A-treated osteosarcoma cells. Using a mouse xenograft model, we found that Proscillaridin A treatment significantly inhibited tumor growth and lung metastasis in vivo and decreased the expression levels of Bcl-xl and MMP2. No noticeable side effect was observed in the liver, kidney, and hematological functions. Conclusively, Proscillaridin A suppressed proliferation, induced apoptosis, and inhibited 143B cell metastasis in vitro and in vivo, and these effects could be mediated by downregulating the expressions of Bcl-xl and MMP2.


Assuntos
Neoplasias Ósseas/tratamento farmacológico , Neoplasias Ósseas/patologia , Osteossarcoma/tratamento farmacológico , Osteossarcoma/patologia , Proscilaridina/farmacologia , Animais , Antineoplásicos/efeitos adversos , Antineoplásicos/farmacologia , Apoptose/efeitos dos fármacos , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Humanos , Masculino , Metaloproteinase 2 da Matriz/genética , Metaloproteinase 2 da Matriz/metabolismo , Camundongos Endogâmicos BALB C , Proscilaridina/efeitos adversos , Ensaios Antitumorais Modelo de Xenoenxerto , Proteína bcl-X/genética , Proteína bcl-X/metabolismo
5.
Cancer Cell Int ; 20: 245, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-32549792

RESUMO

BACKGROUND: Osteosarcoma (OS) is the most common primary bone malignancy in children and adolescents, and hyperproliferation of cells is a major problem of OS. FBXO2 belongs to the family of F-box proteins, and is a substrate recognition component of the Skp1-Cul1-F-box protein (SCF) E3 ubiquitin ligase complex with specificity for high-mannose glycoproteins. The aim of the present study was to investigate the critical role of FBXO2 in OS cells. METHODS: The protein and mRNA expression levels of FBXO2 in clinic OS patients were measured by quantitative real time-polymerase chain reaction (qRT-PCR), Western blot and Immunohistochemical (IHC) staining assays, respectively. The FBXO2 overexpression model was constructed by retro-virus transfection in OS cells. FBXO2 knockout (KO) cells were generated by Clustered regularly interspaced short palindromic repeat (CRISPR)-CRISPR-associated protein 9 (Cas9) assay. Cell counting and colony formation assays were used to analyze the effect of FBXO2 on the biological function of OS cells. FBXO2 KO cells were injected into nude mice to observe tumor growth in vivo. The interaction between FBXO2 and IL-6 was detected by immunoprecipitation. Luciferase assay was used to determine the transcriptional activity of STAT3. RESULTS: Here, we show that FBXO2 is significantly up-regulated in clinical OS samples compared to adjacent normal tissues. Ectopic expression of FBXO2 leads to increased OS cell proliferation and colony-forming ability, while FBXO2 knockout by CRISPR-Cas9-based gene editing has the opposite effect. In addition, the glycoprotein recognition activity of FBXO2 is required for its biological function in OS. In vivo experiments showed that FBXO2 knockout greatly impaired the tumorigenicity of OS cells in nude mice. At the molecular level, we found that knocking out FBXO2 can significantly inhibit STAT3 phosphorylation and downstream target gene expression through IL-6R stabilization. CONCLUSION: Together, these results indicate that FBXO2 promotes OS development by activating the STAT3 signaling pathway, suggesting that FBXO2 may be a new target for OS treatment.

6.
Cell Biol Int ; 44(9): 1820-1830, 2020 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-32379372

RESUMO

Transmembrane protein 98 (TMEM98) is a recently discovered gene, the inhibition of which has preliminarily been demonstrated to inhibit progression of several solid cancers in vitro. However, its involvement in tumorigenesis of gastric cancer (GC) has not been reported. Here, we aimed to explore the expression of TMEM98 in GC tissues and cell lines and to determine the role of TMEM98 in GC cell proliferation and invasion. TMEM98 was significantly upregulated in GC tissues, which was associated with low survival rate of GC patients. Interestingly, GC cell proliferation and invasion were promoted by TMEM98 messenger RNA (mRNA) upregulation and inhibited by TMEM98 mRNA downregulation, but not affected by TMEM98 protein. Using RNA-binding protein immunoprecipitation assay and RNA pull-down assay, we demonstrated that TMEM98 mRNA could directly bind with and upregulate nuclear factor 90 (NF90). Similarly, NF90 protein could not only enhance the stability of TMEM98 mRNA but antagonize the suppressive effect of TMEM98-small interfering RNA on proliferation and invasion in MKN-45 cells. Moreover, RNA pull-down assay, with wild-type (WT) and binding-site-mutated biotinylated TMEM98 mRNA transcripts, demonstrated that WT TMEM98 mRNA bound with NF90 protein through an 8-nt motif at the last exon, but the motif mutation abolished the capacity of TMEM98 mRNA binding to NF90 protein. Furthermore, overexpression of the WT last exon of TMEM98 increased NF90 expression and cell proliferation/invasion expectedly, but overexpression of the mutated last exon had no obvious effect. In conclusion, TMEM98 mRNA enhanced the proliferation and invasion of GC cells by interacting with the NF90 protein.


Assuntos
Proteínas de Membrana/metabolismo , Proteínas do Fator Nuclear 90/metabolismo , Neoplasias Gástricas/genética , Linhagem Celular Tumoral , Movimento Celular/genética , Proliferação de Células/genética , China , Expressão Gênica/genética , Regulação Neoplásica da Expressão Gênica/genética , Humanos , Proteínas de Membrana/genética , Invasividade Neoplásica/genética , Proteínas do Fator Nuclear 90/genética , RNA Mensageiro/metabolismo , RNA Interferente Pequeno/metabolismo , Proteínas de Ligação a RNA/metabolismo , Transdução de Sinais , Neoplasias Gástricas/metabolismo
7.
Mol Carcinog ; 58(1): 3-18, 2019 01.
Artigo em Inglês | MEDLINE | ID: mdl-29964327

RESUMO

Overcoming platinum drug resistance represents a major clinical challenge in osteosarcoma (OS) treatment. The high rates and patterns of therapeutic failure seen in patients are consistent with a steady accumulation of drug-resistant cancer stem cells (CSCs). Notch signaling is implicated in regulating CSCs and tumor resistance to platinum. Thus, we attempt to investigate whether inhibiting of Notch pathway could sensitize cisplatin (CDDP) to CDDP-resistant OS cells and the underlying molecular mechanisms. OS cell lines resistant to CDDP were treated with DAPT, CDDP or combination, we present evidences that DAPT enhances the cytotoxic effect of CDDP in resistant OS by inhibiting proliferation, resulting in G0/G1 cell-cycle arrest, inducing apoptosis, and reducing motility. In addition, DAPT targeting depletes OS stem cells (OSCs), thus increasing tumor sensitivity to platinum, which indicating that a dual combination targeting both OSCs and the bulk of tumor cells are needed for tumor eradication. We also found that the combination of CDDP and DAPT exhibit additive suppression on phosphorylated AKT and ERK, contributing to the anti-cancer effects. In animal model, this combination therapy inhibits the growth and metastasis of CDDP resistant tumor xenografts in nude mice to a greater extent than treatment with either reagent alone. Based on these results, we conclude that CDDP plus DAPT was able to sensitize CDDP-resistant human OS cells to CDDP by downregulation of Notch signaling. CDDP and DAPT combination treatment may be effective and promising for advanced OS.


Assuntos
Neoplasias Ósseas/tratamento farmacológico , Cisplatino/farmacologia , Diaminas/farmacologia , Resistencia a Medicamentos Antineoplásicos/efeitos dos fármacos , Sinergismo Farmacológico , Osteossarcoma/tratamento farmacológico , Receptor Notch1/antagonistas & inibidores , Tiazóis/farmacologia , Adolescente , Adulto , Secretases da Proteína Precursora do Amiloide/antagonistas & inibidores , Animais , Antineoplásicos/farmacologia , Apoptose , Biomarcadores Tumorais/genética , Biomarcadores Tumorais/metabolismo , Neoplasias Ósseas/metabolismo , Neoplasias Ósseas/patologia , Estudos de Casos e Controles , Proliferação de Células , Criança , Quimioterapia Combinada , Seguimentos , Humanos , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Nus , Células-Tronco Neoplásicas/efeitos dos fármacos , Células-Tronco Neoplásicas/metabolismo , Células-Tronco Neoplásicas/patologia , Osteossarcoma/metabolismo , Osteossarcoma/patologia , Prognóstico , Receptor Notch1/genética , Receptor Notch1/metabolismo , Transdução de Sinais , Taxa de Sobrevida , Células Tumorais Cultivadas , Ensaios Antitumorais Modelo de Xenoenxerto , Adulto Jovem
8.
Cells Tissues Organs ; 208(1-2): 13-24, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-32045921

RESUMO

OBJECTIVE: The aim of this work was to determine the effect of miR-375 on chondrocyte metabolism and oxidative stress in osteoarthritis (OA) mouse models through the JAK2/STAT3 signaling pathway. METHODS: Chondrocytes were divided into control, IL-1ß, IL-1ß + miR-375 mimic, IL-1ß + miR-375 inhibitor, IL-1ß + miR-NC (negative control), and IL-1ß + miR-375 inhibitor + siJAK2 groups. The chondrocyte proliferation was determined by MTT assay, the superoxide dismutase (SOD) and malondialdehyde (MDA) levels by corresponding kits, and the chondrocyte apoptosis by TUNEL staining. Furthermore, OA mouse models were divided into Sham, OA + miR-NC, and OA + miRNA-375 antagomir groups. The pathological changes were observed, and the expressions of miR-375 and the JAK2/STAT3 pathway were determined by qRT-PCR and Western blotting, respectively. RESULTS: IL-1ß-induced chondrocytes had significant increases in miR-375 and MDA, with decreased proliferation and SOD levels, as compared to the control group. Meanwhile, they also exhibited elevated apoptosis, with upregulations of ADAMTS-5 and MMP-13 and downregulations of COL2A1 and ACAN, as well as decreased p-JAK2/JAK2, p-STAT3/STAT3, and Bcl-2/Bax. However, these changes were significantly improved after transfection with miR-375 inhibitor, but transfection with miR-375 mimic resulted in severer exacerbation. Notably, the improvement of miR-375 inhibitor could be abolished by transfection with siJAK2. Furthermore, miR-375 antagomir significantly alleviated OA progression in OA mice in vivo. CONCLUSION: MiR-375 suppression enhanced the ability of chondrocyte to antagonize the oxidative stress and maintained the homeostasis of extracellular matrix metabolism to protect chondrocytes from OA via activation of the JAK2/STAT3 pathway, indicating that miR-375 is a potential molecular target for OA treatment.


Assuntos
Condrócitos/patologia , MicroRNAs/metabolismo , Osteoartrite , Animais , Apoptose , Modelos Animais de Doenças , Desenvolvimento de Medicamentos , Interleucina-1beta/metabolismo , Janus Quinase 2/metabolismo , Camundongos , Osteoartrite/tratamento farmacológico , Osteoartrite/metabolismo , Estresse Oxidativo , Fator de Transcrição STAT3/metabolismo , Transdução de Sinais
9.
Cell Physiol Biochem ; 46(3): 1134-1147, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-29669353

RESUMO

BACKGROUND/AIMS: Osteosarcoma is a common primary malignant bone tumor that mainly occurs in childhood and adolescence. Despite developments in the diagnosis and treatment of osteosarcoma, the prognosis is still very poor. Cinobufagin is an active component in the anti-tumor Chinese medicine called "Chan Su", and we previously revealed that cinobufagin induced apoptosis and reduced the viability of osteosarcoma cells; however, the underlying mechanism remains to be elucidated. Herein, the present study was undertaken to illuminate the molecular mechanism of cinobufagin-induced apoptosis of osteosarcoma cell. METHODS: U2OS and 143B cells were treated with different concentrations of cinobufagin. Cell viability, colony formation ability and morphological changes were assessed by a CCK-8 assay, a clonogenic assay and light microscopy, respectively. Cell apoptosis was detected by Hoechst 33258 and Annexin V-FITC/PI staining. Reactive oxygen species (ROS) and mitochondrial membrane potential (ΔΨm) were determined by flow cytometry. Glutathione (GSH) levels were detected by a GSH and GSSG assay kit. The levels of apoptosis-related proteins were determined by western blotting, and 143B cells were introduced to establish a xenograft tumor model. The effect of cinobufagin on osteosarcoma was further investigated in vivo. RESULTS: Our results showed that cinobufagin significantly reduced the viability of U2OS and 143B cells in vitro in a dose-and time-dependent manner. In addition, cinobufagin-induced apoptosis in U2OS and 143B cells was concentration-dependent. Moreover, we found that cinobufagin treatment increased the level of intracellular ROS, decreased ΔΨm, reduced GSH and inhibited GSH reductase (GR). The effects of cinobufagin on cell proliferation, apoptosis, ROS generation and ΔΨm loss were dramatically reversed when the cells were pretreated with the thiol-antioxidants NAC or GSH. Moreover, cinobufagin treatment increased the expression of the pro-apoptotic protein Bax and decreased the expression of the anti-apoptitic protein Bcl-2, thus altering the ratio of Bax to Bcl-2. Furthermore, Cinobufagin treatment caused cytochrome c release from the mitochondria to cytoplasm, thus increasing the protein levels of cleaved-caspase family members to induce apoptosis. Ac-DEVD-CHO or Z-LEHD-FMK significantly reduced cinobufagin-induced apoptosis. Finally, a subcutaneous xenograft animal study verified that cinobufagin also significantly suppressed osteosarcoma growth in vivo. CONCLUSIONS: Our present data demonstrated that cinobufagin triggered cell apoptosis in osteosarcoma cells via the intrinsic mitochondria-dependent apoptosis pathway by the accumulation of ROS and the loss of ΔΨm. In an in vivo subcutaneous xenograft model, cinobufagin exhibited excellent tumor inhibitory effects. These results suggest that cinobufagin might potentially be further developed as an anti-tumor candidate for treating osteosarcoma patients in the clinic.


Assuntos
Apoptose/efeitos dos fármacos , Bufanolídeos/toxicidade , Mitocôndrias/efeitos dos fármacos , Animais , Neoplasias Ósseas/tratamento farmacológico , Neoplasias Ósseas/metabolismo , Neoplasias Ósseas/patologia , Bufanolídeos/química , Bufanolídeos/uso terapêutico , Inibidores de Caspase/farmacologia , Linhagem Celular Tumoral , Sobrevivência Celular/efeitos dos fármacos , Glutationa/metabolismo , Glutationa Redutase/antagonistas & inibidores , Glutationa Redutase/metabolismo , Humanos , Masculino , Potencial da Membrana Mitocondrial/efeitos dos fármacos , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Nus , Mitocôndrias/metabolismo , Oligopeptídeos/farmacologia , Osteossarcoma/tratamento farmacológico , Osteossarcoma/metabolismo , Osteossarcoma/patologia , Proteínas Proto-Oncogênicas c-bcl-2/metabolismo , Espécies Reativas de Oxigênio/metabolismo , Transdução de Sinais/efeitos dos fármacos
10.
Biochem Biophys Res Commun ; 505(2): 372-377, 2018 10 28.
Artigo em Inglês | MEDLINE | ID: mdl-30253943

RESUMO

Cisplatin (CDDP) has been shown to be a promising anticancer drug that is effective against many types of cancer, which include osteosarcoma (OS). However, its therapeutic application is restricted by its toxicity in normal tissues, side effects caused in patients, and chemotherapy resistance. Thus, to further improve patients' treatment, the development of novel, more effective and well tolerated therapeutic approaches against OS in clinical is urgent and important. In the present study, nude mice were inoculated subcutaneously with injections of HOS8603 cells, CDDP and docetaxel (DTX) were administered intraperitoneally respectively. The inhibitive effects and the side effects were observed. Tumor weights and volumes were significantly lower and the tumor inhibition rate was significantly higher in the combination group than those of either drug alone or vehicle. The cell density in the tumor tissue was significantly decreased, apoptotic and necrotic cell death was significantly increased in the combination group, as compared with those of either drug alone or vehicle. In addition, there was no obvious side effect happening besides the appearance of erythema and papules in some mice. These results suggest that the combined effects of CDDP and DTX on the growth of human OS in vivo were superior to the single effects. CDDP combined with DTX had synergistic effects at lower concentrations and promoted apoptosis, but did not increase the side effects of chemotherapy.


Assuntos
Protocolos de Quimioterapia Combinada Antineoplásica/uso terapêutico , Cisplatino/uso terapêutico , Docetaxel/uso terapêutico , Osteossarcoma/tratamento farmacológico , Animais , Apoptose/efeitos dos fármacos , Linhagem Celular Tumoral , Sinergismo Farmacológico , Xenoenxertos , Humanos , Camundongos , Camundongos Nus , Carga Tumoral/efeitos dos fármacos
11.
Cancer Cell Int ; 17: 29, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-28239298

RESUMO

BACKGROUND: Evidence is accumulating to link cancer stem cells to the pathogenesis and progression of osteosarcoma. The aim of this study is to investigate the role of miR-335 in osteosarcoma stem cells. METHODS: Tumor spheroid culture and flow cytometry were applied to screen out osteosarcoma stem cells. Real-time quantitative PCR was used to detect the expression level of miR-335 in MG63, U2OS and 143B osteosarcoma stem cells. The relationship of miR-335 expression with osteosarcoma stem cells was then analyzed. Transwell assay and transplantation assay were performed to elucidate biological effects of miR-335 on cell invasion and vivo tumor formation. Western Blot and luciferase assays were executed to investigate the regulation of POU5F1 by miR-335. RESULTS: The expression of miR-335 in osteosarcoma stem cells was lower than their differentiated counterparts. Cells expressing miR-335 possessed decreased stem cell-like properties. Gain or loss of function assays were applied to find that miR-335 antagonist promoted stem cell-like properties as well as invasion. Luciferase report and transfection assay showed that POU5F1 was downregulated by miR-335. Pre-miR-335 resulted in tumor enhanced sensitivity to traditional chemotherapy, whereas anti-miR-335 promoted chemoresistance. Finally, the inhibitory effect of miR-335 on in vivo tumor formation showed that combination of pre-miR-335 with cisplatin further reduced the tumor size, and miR-335 brought down the sphere formation capacity induced by cisplatin. CONCLUSIONS: The current study demonstrates that miR-335 negatively regulates osteosarcoma stem cell-like properties by targeting POU5F1, and miR-335 could target CSCs to synergize with traditional chemotherapeutic agents to overcome osteosarcoma.

12.
Tumour Biol ; 36(5): 3735-43, 2015 May.
Artigo em Inglês | MEDLINE | ID: mdl-25560491

RESUMO

Beyond their established role in the mammalian circadian clock, recent studies have confirmed that the circadian genes have been implicated in tumor onset and progression. Currently, the biological effects of circadian genes on osteosarcoma cells' proliferation and migration are not well understood. Period2 (Per2) is one of the core circadian genes that act as master regulators of development and is frequently dysregulated in several cancers. However, the effects of human Per2 (hPer2) on the biological behavior of osteosarcoma cells are rarely reported. In the present study, to address the expression of hPer2 in osteosarcoma cells, the pEGFP-N1-hPer2 eukaryotic expression vector was constructed and transfected into cultured MG63 cells using Lipofectamine™ 2000. The overexpression of hPer2 in MG63 cells was verified by qRT-PCR and Western blotting, respectively. Finally, we investigated the effects of hPer2 protein overexpression on MG63 cells' viability, cycle, apoptosis, and invasive ability. In conclusion, the recombinant pEGFP-N1-hPer2 plasmid had been constructed successfully and expressed effectively in MG63 cells. Furthermore, results also showed that the viability, proliferation, and invasive abilities were suppressed, and the apoptosis was enhanced in MG63 cells. This preliminary study provides ground work for further research on the roles of circadian gene hPer2 in osteosarcoma cells MG63 and would offer promise for the development of novel therapeutic strategies in the treatment of osteosarcoma.


Assuntos
Neoplasias Ósseas/patologia , Osteossarcoma/patologia , Proteínas Circadianas Period/genética , Apoptose , Ciclo Celular , Linhagem Celular Tumoral , Proliferação de Células , Ritmo Circadiano , Humanos , Microscopia de Fluorescência , Invasividade Neoplásica , Proteínas Circadianas Period/fisiologia , Plasmídeos
13.
Mol Cell Biochem ; 400(1-2): 245-52, 2015 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-25416447

RESUMO

Recent data strongly suggest the important role of miRNAs in various cancer-related processes. Osteosarcoma (OS) is the most common primary cancer of the bone and usually leads to deaths due to its rapid proliferation and metastasis. Here, we demonstrated that compared with noncancerous bone tissues, miR-135b expression is frequently upregulated in OS specimens, inversely correlated with potential target-FOXO1 expression pattern. Bioinformatics analysis combined with experimental confirmation revealed FOXO1 is a direct target of miR-135b in OS. Functionally, miR-135b inhibitor significantly inhibited OS cells proliferation and invasion. Forced expression of FOXO1 showed the opposite effect, and FOXO1 knockdown abolished the effect of miR-135b inhibitor. Taken together, our data provide compelling evidence that miR-135b functions as an onco-miRNA in OS to promote OS cells proliferation and invasion, and its oncogenic effects are mediated chiefly through targeting FOXO1.


Assuntos
Fatores de Transcrição Forkhead/genética , MicroRNAs/biossíntese , Osteossarcoma/genética , Linhagem Celular Tumoral , Proliferação de Células/genética , Proteína Forkhead Box O1 , Fatores de Transcrição Forkhead/biossíntese , Regulação Neoplásica da Expressão Gênica , Humanos , MicroRNAs/genética , Invasividade Neoplásica/genética , Osteossarcoma/patologia
14.
BMC Musculoskelet Disord ; 16: 148, 2015 Jun 16.
Artigo em Inglês | MEDLINE | ID: mdl-26076895

RESUMO

BACKGROUND: The suture bridge (SB) technique and conventional double-row (DR) are both effective in repair of full-thickness rotator cuff tears . However, increasing numbers of scholars believe that the SB technique produces better results than conventional DR because of the higher bone-tendon contact area and pressure. However, The clinical outcomes have been mixed and little direct evidence has been supplied in vivo. This study was designed using the SB and DR techniques to determine which is the better technique. METHODS: Sixty-four New Zealand white rabbits were randomly divided into 2 groups, the SB group and DR group. SB and DR were then used to repair their rotator cuff tears. Rabbits were then sacrificed at the 2(nd), 4(th), or 8(th) week after surgery and a histological comparison was made. The biomechanical comparison was made at the 8(th) week. RESULTS: The load to failure of the SB group was 134.59 ± 17.69 N at the 8(th) postoperative week, and that was significantly higher than in the DR group (103.83 ± 6.62, P = 0.001), but both repair groups remained lower than in the control group (199.25 ± 14.81). Histological evaluation showed that both the SB and DR groups healed at the bone-tendon interface. But there were subtle differences between the two groups in the structure and morphology of collagen fibers and cartilage cells at bone-tendon interface. In general, the collagen fibers of the SB group were more compact than those of the DR group at all times tested. At the 4(th) and 8(th) weeks, the collagen fibers and cartilage cells in the SB group were arranged in a column modality, but those in the DR group were distributed horizontally. CONCLUSION: The SB technique facilitated healing more effectively than the conventional DR technique. The difference in morphology of collagen fibers and cartilage cells may be related to the difference in bone-tendon contact pressure.


Assuntos
Cabeça do Úmero/cirurgia , Manguito Rotador/cirurgia , Técnicas de Sutura , Traumatismos dos Tendões/cirurgia , Animais , Fenômenos Biomecânicos , Modelos Animais de Doenças , Cabeça do Úmero/patologia , Cabeça do Úmero/fisiopatologia , Masculino , Pressão , Coelhos , Manguito Rotador/patologia , Manguito Rotador/fisiopatologia , Lesões do Manguito Rotador , Estresse Mecânico , Traumatismos dos Tendões/patologia , Traumatismos dos Tendões/fisiopatologia , Fatores de Tempo , Cicatrização
15.
Transl Oncol ; 39: 101802, 2024 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-37839175

RESUMO

BACKGROUND: Serpin Family H Member 1 (SERPINH1) may be involved in the regulation of occurrence and development of tumors. However, the role and mechanism of SERPINH1 in osteosarcoma remain poorly understood. The aim of this study is to investigate the expression and role of SRPINH1 in osteosarcoma and to elucidate its underlying mechanisms. METHODS: First, we examined the expression of SERPINH1 in osteosarcoma and analyzed publicly available datasets to investigate whether SERPINH1 expression was associated with the prognosis of osteosarcoma. Then we constructed SERPINH1 overexpression and knockdown systems in osteosarcoma cells, and examined the proliferation, migration and invasion ability of osteosarcoma cells after SERPINH1 expression changes using CCK-8 assay, wound healing assay and transwell invasion assay. In addition, we constructed a subcutaneous xenograft tumor model to study the function of SERPINH1 in vivo. We also examined the downstream pathways of SERPINH1 by functional analysis and performed subsequent validation. RESULTS: SERPINH1 was upregulated and associated with poor survival in patients with osteosarcoma. SERPINH1 promoted the proliferation, migration and invasion of osteosarcoma cells and promotes the growth of osteosarcoma in vivo by activating the PI3K-Akt signaling pathway. CONCLUSION: SERPINH1 partakes in the biological process of osteosarcoma as a tumor promotor and may be an emerging biomarker in osteosarcoma.

16.
Biomedicines ; 12(1)2024 Jan 19.
Artigo em Inglês | MEDLINE | ID: mdl-38275399

RESUMO

Maintaining proper mechanical strength and tissue volume is important for bone growth at the site of a bone defect. In this study, potassium magnesium phosphate hexahydrate (KMgPO4·6H2O, MPC) was applied to gelma-methacrylate hydrogel (GelMA) to prepare GelMA/MPC composites (GMPCs). Among these, 5 GMPC showed the best performance in vivo and in vitro. These combinations significantly enhanced the mechanical strength of GelMA and regulated the degradation and absorption rate of MPC. Considerably better mechanical properties were noted in 5 GMPC compared with other concentrations. Better bioactivity and osteogenic ability were also found in 5 GMPC. Magnesium ions (Mg2+) are bioactive and proven to promote bone tissue regeneration, in which the enhancement efficiency is closely related to Mg2+ concentrations. These findings indicated that GMPCs that can release Mg2+ are effective in the treatment of bone defects and hold promise for future in vivo applications.

17.
Biomedicines ; 12(2)2024 Feb 01.
Artigo em Inglês | MEDLINE | ID: mdl-38397946

RESUMO

In orthopedics, the repair of bone defects remains challenging. In previous research reports, magnesium phosphate cements (MPCs) were widely used because of their excellent mechanical properties, which have been widely used in the field of orthopedic medicine. We built a new k-struvite (MPC) cement obtained from zinc oxide (ZnO) and assessed its osteogenic properties. Zinc-doped magnesium phosphate cement (ZMPC) is a novel material with good biocompatibility and degradability. This article summarizes the preparation method, physicochemical properties, and biological properties of ZMPC through research on this material. The results show that ZMPC has the same strength and toughness (25.3 ± 1.73 MPa to 20.18 ± 2.11 MPa), that meet the requirements of bone repair. Furthermore, the material can gradually degrade (12.27% ± 1.11% in 28 days) and promote osteogenic differentiation (relative protein expression level increased 2-3 times) of rat bone marrow mesenchymal stem cells (rBMSCs) in vitro. In addition, in vivo confirmation revealed increased bone regeneration in a rat calvarial defect model compared with MPC alone. Therefore, ZMPC has broad application prospects and is expected to be an important repair material in the field of orthopedic medicine.

18.
Biol Direct ; 19(1): 28, 2024 Apr 22.
Artigo em Inglês | MEDLINE | ID: mdl-38650011

RESUMO

BACKGROUND: Osteosarcoma is a diverse and aggressive bone tumor. Driver genes regulating osteosarcoma initiation and progression remains incompletely defined. Zinc finger protein 692 (ZNF692), a kind of Krüppel C2H2 zinc finger transcription factor, exhibited abnormal expression in different types of malignancies and showed a correlation with the clinical prognosis of patients as well as the aggressive characteristics of cancer cells. Nevertheless, its specific role in osteosarcoma is still not well understood. METHODS: We investigated the dysregulation and clinical significance of ZNF692 in osteosarcoma through bioinformatic method and experimental validation. A range of in vitro assays, including CCK-8, colony formation, EdU incorporation, wound healing, and transwell invasion tests, were conducted to assess the impact of ZNF692 on cell proliferation, migration, and invasion in osteosarcoma. A xenograft mouse model was established to evaluate the effect of ZNF692 on tumor growth in vivo. Western blot assay was used to measure the protein levels of MEK1/2, P-MEK1/2, ERK1/2, and P-ERK1/2 in cells that had been genetically modified to either reduce or increase the expression of ZNF692. The relationship between ZNF692 and tyrosine kinase non-receptor 2 (TNK2) were validated by qRT-PCR, chromatin immunoprecipitation and luciferase reporter assays. RESULTS: Expression of ZNF692 was increased in both human osteosarcoma tissues and cell lines. Furthermore, the expression of ZNF692 served as an independent predictive biomarker in osteosarcoma. The results of the survival analysis indicated that increased expression of ZNF692 was associated with worse outcome. Downregulation of ZNF692 inhibits the proliferation, migration, and invasion of osteosarcoma cells, whereas upregulation of ZNF692 has the opposite impact. Western blot assay indicates that reducing ZNF692 decreases phosphorylation of MEK1/2 and ERK1/2, whereas increasing ZNF692 expression enhances their phosphorylation. U0126, a potent inhibitor specifically targeting the MEK/ERK signaling pathway, partially counteracts the impact of ZNF692 overexpression on the proliferation, migration, and invasion of osteosarcoma cells. In addition, ZNF692 specifically interacts with the promoter region of TNK2 and stimulates the transcription of TNK2 in osteosarcoma cells. Forcing the expression of TNK2 weakens the inhibitory impact of ZNF692 knockdown on P-MEK1/2 and P-ERK1/2. Similarly, partly inhibiting TNK2 counteracts the enhancing impact of ZNF692 overexpression on the phosphorylation of MEK1/2 and ERK1/2. Functional tests demonstrate that the suppressive effects of ZNF692 knockdown on cell proliferation, migration, and invasion are greatly reduced when TNK2 is overexpressed. In contrast, the reduction of TNK2 hinders the ability of ZNF692 overexpression to enhance cell proliferation, migration, and invasion. CONCLUSION: ZNF692 promotes the proliferation, migration, and invasion of osteosarcoma cells via the TNK2-dependent stimulation of the MEK/ERK signaling pathway. The ZNF692-TNK2 axis might potentially function as a possible predictive biomarker and a promising target for novel therapeutics in osteosarcoma.


Assuntos
Movimento Celular , Proliferação de Células , Sistema de Sinalização das MAP Quinases , Osteossarcoma , Animais , Feminino , Humanos , Camundongos , Neoplasias Ósseas/genética , Neoplasias Ósseas/metabolismo , Linhagem Celular Tumoral , Regulação Neoplásica da Expressão Gênica , Camundongos Nus , Invasividade Neoplásica , Osteossarcoma/genética , Osteossarcoma/metabolismo
19.
Int J Biol Macromol ; 266(Pt 1): 130998, 2024 May.
Artigo em Inglês | MEDLINE | ID: mdl-38521332

RESUMO

Although calcium­magnesium phosphate cements (CMPCs) have been widely applied to treating critical-size bone defects, their repair efficiency is unsatisfactory owing to their weak surface bioactivity and uncontrolled ion release. In this study, we lyophilized alginate sodium (AS) as a coating onto HAp/K-struvite (H@KSv) to develop AS/HAp/K-struvite (AH@KSv), which promotes bone regeneration. The compressive strength and hydrophilicity of AH@KSv significantly improved, leading to enhanced cell adhesion in vitro. Importantly, the SA coating enables continuous ions release of Mg2+ and Ca2+, finally leading to enhanced osteogenesis in vitro/vivo and different patterns of new bone ingrowth in vivo. Furthermore, these composites increased the expression levels of biomarkers of the TRPM7/PI3K/Akt signaling pathway via an equilibrium effect of Mg2+ to Ca2+. In conclusion, our study provides novel insights into the mechanisms of Mg-based biomaterials for bone regeneration.


Assuntos
Alginatos , Cimentos Ósseos , Regeneração Óssea , Fosfatos , Fosfatidilinositol 3-Quinases , Proteínas Proto-Oncogênicas c-akt , Transdução de Sinais , Canais de Cátion TRPM , Regeneração Óssea/efeitos dos fármacos , Canais de Cátion TRPM/metabolismo , Alginatos/química , Alginatos/farmacologia , Fosfatidilinositol 3-Quinases/metabolismo , Proteínas Proto-Oncogênicas c-akt/metabolismo , Transdução de Sinais/efeitos dos fármacos , Animais , Fosfatos/química , Fosfatos/farmacologia , Cimentos Ósseos/química , Cimentos Ósseos/farmacologia , Osteogênese/efeitos dos fármacos , Compostos de Magnésio/química , Compostos de Magnésio/farmacologia , Fosfatos de Cálcio/química , Fosfatos de Cálcio/farmacologia , Adesão Celular/efeitos dos fármacos , Propriedades de Superfície , Camundongos , Ratos , Força Compressiva
20.
Bioact Mater ; 37: 459-476, 2024 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-38698920

RESUMO

Magnesium phosphate bone cements (MPC) have been recognized as a viable alternative for bone defect repair due to their high mechanical strength and biodegradability. However, their poor porosity and permeability limit osteogenic cell ingrowth and vascularization, which is critical for bone regeneration. In the current study, we constructed a novel hierarchically-porous magnesium phosphate bone cement by incorporating extracellular matrix (ECM)-mimicking electrospun silk fibroin (SF) nanofibers. The SF-embedded MPC (SM) exhibited a heterogeneous and hierarchical structure, which effectively facilitated the rapid infiltration of oxygen and nutrients as well as cell ingrowth. Besides, the SF fibers improved the mechanical properties of MPC and neutralized the highly alkaline environment caused by excess magnesium oxide. Bone marrow stem cells (BMSCs) adhered excellently on SM, as illustrated by formation of more pseudopodia. CCK8 assay showed that SM promoted early proliferation of BMSCs. Our study also verified that SM increased the expression of OPN, RUNX2 and BMP2, suggesting enhanced osteogenic differentiation of BMSCs. We screened for osteogenesis-related pathways, including FAK signaing, Wnt signaling and Notch signaling, and found that SM aided in the process of bone regeneration by suppressing the Notch signaling pathway, proved by the downregulation of NICD1, Hes1 and Hey2. In addition, using a bone defect model of rat calvaria, the study revealed that SM exhibited enhanced osteogenesis, bone ingrowth and vascularization compared with MPC alone. No adverse effect was found after implantation of SM in vivo. Overall, our novel SM exhibited promising prospects for the treatment of critical-sized bone defects.

SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA