Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 4.868
Filter
1.
Zhong Nan Da Xue Xue Bao Yi Xue Ban ; 49(6): 870-877, 2024 Jun 28.
Article in English, Chinese | MEDLINE | ID: mdl-39311783

ABSTRACT

OBJECTIVES: Osteosarcoma is the most common malignant bone tumor in children and adolescents, characterized by a high potential for proliferation and metastasis. Patients with osteosarcoma who have distant metastases generally have a poor prognosis. Challenges in treatment include incomplete resection of tumor and chemotherapy resistance, with no effective cure currently available. Recent studies suggest that ß-1,4-N-acetyl-galactosaminyltransferase 1 (B4GALNT1) plays a role in the progression of various malignant tumors. However, the function of B4GALNT1 in osteosarcoma cells has not been reported. This study aims to investigate the expression of B4GALNT1 in osteosarcoma tissues compared to normal tissues and to explore its effects on the proliferation, migration, and invasion of osteosarcoma cells, thereby providing new theoretical foundations and directions for the treatment of osteosarcoma patients. METHODS: Tumor tissues and corresponding normal tissue samples were collected from 16 osteosarcoma patients who underwent tumor resection at the Second Xiangya Hospital of Central South University. The patients' ages ranged from 8 to 17 years (median age 12 years). The expression of B4GALNT1 mRNA in osteosarcoma tissues, corresponding normal tissues, 3 osteosarcoma cell lines (MG63, Saos-2, and U2OS), and human fetal osteoblastic cells (hFOB) was detected using real-time reverse transcription PCR (real-time RT-PCR). The effects of B4GALNT1 knockdown on the proliferation of osteosarcoma cells Saos-2 and U2OS were analyzed using cell counting kit-8 (CCK-8) assays and colony formation assays. The effects of B4GALNT1 knockdown on the migration and invasion abilities of Saos-2 and U2OS cells were evaluated using Transwell migration and invasion assays. Western blotting analysis was performed to assess the impact of B4GALNT1 knockdown on the expression of epithelial-mesenchymal transition (EMT) and invasion-related proteins in Saos-2 and U2OS cells. RESULTS: Real-time RT-PCR results showed that B4GALNT1 mRNA expression levels were significantly higher in osteosarcoma tissues and the 3 osteosarcoma cell lines compared to normal tissues and hFOB cells (all P<0.01). CCK-8 and colony formation assays indicated that B4GALNT1 knockdown significantly reduced the proliferation rate of osteosarcoma cells compared to the control group (all P<0.05). Transwell migration and invasion assays demonstrated that B4GALNT1 knockdown significantly decreased the number of migrating and invading osteosarcoma cells (all P<0.01). Western blotting analysis revealed that B4GALNT1 knockdown inhibited the expression of N-cadherin, Snail, Vimentin, and matrix metalloproteinase 9 (MMP9) compared to the control group (all P<0.01). CONCLUSIONS: B4GALNT1 is upregulated in osteosarcoma tissues and cell lines, and its knockdown suppresses the malignant phenotype of osteosarcoma cells. B4GALNT1 may function as an oncogene in the proliferation and metastasis of osteosarcoma cells.


Subject(s)
Bone Neoplasms , Cell Movement , Cell Proliferation , Down-Regulation , N-Acetylgalactosaminyltransferases , Osteosarcoma , Humans , Osteosarcoma/pathology , Osteosarcoma/genetics , Osteosarcoma/metabolism , N-Acetylgalactosaminyltransferases/genetics , N-Acetylgalactosaminyltransferases/metabolism , Cell Proliferation/genetics , Child , Bone Neoplasms/genetics , Bone Neoplasms/pathology , Bone Neoplasms/metabolism , Adolescent , Cell Line, Tumor , Cell Movement/genetics , Male , Female , Neoplasm Invasiveness , Polypeptide N-acetylgalactosaminyltransferase , Neoplasm Metastasis , RNA, Small Interfering/genetics , RNA, Messenger/genetics , RNA, Messenger/metabolism
2.
Cell Physiol Biochem ; 58(5): 510-526, 2024 Sep 15.
Article in English | MEDLINE | ID: mdl-39305132

ABSTRACT

BACKGROUND/AIMS: Osteosarcoma is a prevalent and aggressive primary malignant bone tumor affecting children and adolescents. Despite advancements in sequencing technologies, there remains a lack of reliable prognostic biomarkers and effective targeted therapies for osteosarcoma. This study focuses on identifying key prognostic genes, particularly the role of GNAS, in osteosarcoma progression. METHODS: Bioinformatics analyses were performed on osteosarcoma datasets from the Gene Expression Omnibus (GEO). Differential gene expression analysis, weighted correlation network analysis (WGCNA), and survival analysis identified potential prognostic hub genes. The expression and function of these genes were validated through immunohistochemistry and animal experiments. Specifically, the role of GNAS was investigated through siRNA-mediated knockdown in osteosarcoma cell lines and nude mice models. RESULTS: Five hub genes (PROP1, GNAS, CYP4F2, LHX3, CNGB1) were identified as significantly related to osteosarcoma prognosis. Among these, GNAS was found to be highly expressed in osteosarcoma tissues compared to normal tissues. Immunohistochemical analysis confirmed the elevated expression of GNAS in osteosarcoma samples. GNAS mutation analysis revealed a low mutation rate in osteosarcoma, suggesting its oncogenic role is independent of mutational status. Animal experiments demonstrated that knocking down GNAS significantly inhibited tumor growth and induced apoptosis in osteosarcoma cells. CONCLUSION: GNAS is highly expressed in osteosarcoma and associated with poor prognosis, acting as an oncogene in osteosarcoma progression. Targeting GNAS could be a potential therapeutic strategy for osteosarcoma. Further studies on GNAS-related signaling pathways may provide deeper insights into the molecular mechanisms driving osteosarcoma malignancy.


Subject(s)
Bone Neoplasms , Chromogranins , GTP-Binding Protein alpha Subunits, Gs , Mice, Nude , Osteosarcoma , Osteosarcoma/genetics , Osteosarcoma/pathology , Osteosarcoma/metabolism , Humans , GTP-Binding Protein alpha Subunits, Gs/genetics , GTP-Binding Protein alpha Subunits, Gs/metabolism , Animals , Chromogranins/genetics , Chromogranins/metabolism , Cell Line, Tumor , Bone Neoplasms/genetics , Bone Neoplasms/pathology , Bone Neoplasms/metabolism , Prognosis , Mice , Male , Female , Mutation , RNA, Small Interfering/metabolism , Cell Proliferation , RNA Interference , Apoptosis , Mice, Inbred BALB C , Carcinogenesis/genetics , Carcinogenesis/pathology
3.
Aging (Albany NY) ; 16(17): 12239-12251, 2024 Sep 06.
Article in English | MEDLINE | ID: mdl-39248711

ABSTRACT

The relationship between metastasis-associated protein 2 (MTA2) overexpression and tumor growth and metastasis has been extensively studied in a variety of tumor cells but not in human osteosarcoma cells. This study aims to elucidate the clinical significance, underlying molecular mechanisms, and biological functions of MTA2 in human osteosarcoma in vitro and in vivo. Our results show that MTA2 was elevated in osteosarcoma cell lines and osteosarcoma tissues and was associated with tumor stage and overall survival of osteosarcoma patients. Knockdown of MTA2 inhibited osteosarcoma cell migration and invasion by reducing the expression of urokinase-type plasminogen activator (uPA). Bioinformatic analysis demonstrated that high levels of uPA in human osteosarcoma tissues correlated positively with MTA2 expression. Furthermore, treatment with recombinant human uPA (Rh-uPA) caused significant restoration of OS cell migration and invasion in MTA2 knockdown osteosarcoma cells. We found that ERK1/2 depletion increased the expression of uPA, facilitating osteosarcoma cell migration and invasion. Finally, MTA2 depletion significantly reduced tumor metastasis and the formation of lung nodules in vivo. Overall, our study suggests that MTA2 knockdown suppresses osteosarcoma cell metastasis by decreasing uPA expression via ERK signaling. This finding provides new insight into potential treatment strategies against osteosarcoma metastasis by targeting MTA2.


Subject(s)
Bone Neoplasms , Cell Movement , Gene Knockdown Techniques , Histone Deacetylases , Osteosarcoma , Repressor Proteins , Urokinase-Type Plasminogen Activator , Osteosarcoma/genetics , Osteosarcoma/pathology , Osteosarcoma/metabolism , Humans , Urokinase-Type Plasminogen Activator/genetics , Urokinase-Type Plasminogen Activator/metabolism , Cell Line, Tumor , Repressor Proteins/genetics , Repressor Proteins/metabolism , Cell Movement/genetics , Bone Neoplasms/genetics , Bone Neoplasms/metabolism , Bone Neoplasms/pathology , Animals , Histone Deacetylases/metabolism , Histone Deacetylases/genetics , Male , Female , Mice , Gene Expression Regulation, Neoplastic , Neoplasm Invasiveness/genetics , Neoplasm Metastasis , Mice, Nude , MAP Kinase Signaling System/genetics
4.
Cancer Med ; 13(18): e70239, 2024 Sep.
Article in English | MEDLINE | ID: mdl-39300969

ABSTRACT

BACKGROUND: Osteosarcoma (OS) survival rates and outcome have not improved in 50 years since the advent of modern chemotherapeutics. Thus, there is a critical need for an improved understanding of the tumor microenvironment to identify better therapies. Extracellular matrix (ECM) deposition and hypoxia are known to abrogate the efficacy of various chemical and cell-based therapeutics. Here, we aim to mechanistically investigate the combinatorial effects of hypoxia and matrix deposition with the use of OS spheroids. METHODS: We use two murine OS cell lines with differential metastatic potential to form spheroids. We form spheroids of two sizes, use ascorbate-2-phosphate supplementation to enhance ECM deposition, and study cell response under standard (21% O2) and physiologic (5% O2) oxygen tensions. Finally, we examine chemotherapeutic responses to doxorubicin treatment. RESULTS: ECM production and oxygen tension are key determinants of spheroid size through cell organization based on nutrient and oxygen distribution. Interestingly, highly metastatic OS is more susceptible to chemotherapeutics compared to less metastatic OS when matrix production increases. Together, these data suggest that dynamic interactions between ECM production and oxygen diffusion may result in distinct chemotherapeutic responses despite inherent tumor aggressiveness. CONCLUSION: This work establishes OS spheroids as a valuable tool for early OS tumor formation investigation and holds potential for novel therapeutic target and prognostic indicator discovery.


Subject(s)
Extracellular Matrix , Osteosarcoma , Oxygen , Spheroids, Cellular , Tumor Microenvironment , Osteosarcoma/drug therapy , Osteosarcoma/metabolism , Osteosarcoma/pathology , Spheroids, Cellular/drug effects , Extracellular Matrix/metabolism , Animals , Mice , Oxygen/metabolism , Cell Line, Tumor , Doxorubicin/pharmacology , Doxorubicin/therapeutic use , Bone Neoplasms/drug therapy , Bone Neoplasms/metabolism , Bone Neoplasms/pathology , Humans , Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use
5.
Folia Biol (Praha) ; 70(2): 113-122, 2024.
Article in English | MEDLINE | ID: mdl-39231319

ABSTRACT

Recent studies have highlighted the significant role of 5-hydroxymethylcytosine (5hmC) in carcinogenesis. However, the specific role of 5hmC in osteosarcoma (OS) remains largely unexplored. The-re-fore, this study aimed to investigate the function of 5hmC and TET3 in OS. In this study, we found a decreased total level of 5hmC in OS tissues. The expression of the TET3 protein was also decreased in OS. Importantly, the decreased levels of TET3 were associated with a decreased disease-free survival (DFS) rate in patients. To investigate the role of TET3 and 5hmC in OS, we manipulated the levels of TET3 in MG-63 cells. Silencing TET3 in these cells resulted in a twofold increase in proliferation. Additio-nally, the level of 5hmC decreased in these cells. Con-versely, over-expression of TET3 in MG-63 cells led to the expected inhibition of proliferation and invasion, accompanied by an increase in 5hmC levels. In conclusion, both 5hmC and TET3 protein levels were decreased in OS. Additionally, the over-expression of TET3 inhibited the proliferation of MG-63 cells, while the suppression of TET3 had the opposite effect. These findings suggest that decreased levels of 5hmC and TET3 may serve as potential markers for OS.


Subject(s)
5-Methylcytosine , Cell Proliferation , DNA Demethylation , Dioxygenases , Epigenesis, Genetic , Female , Humans , Male , 5-Methylcytosine/analogs & derivatives , 5-Methylcytosine/metabolism , Bone Neoplasms/genetics , Bone Neoplasms/metabolism , Bone Neoplasms/pathology , Cell Line, Tumor , Dioxygenases/metabolism , Gene Expression Regulation, Neoplastic , Osteosarcoma/genetics , Osteosarcoma/metabolism , Osteosarcoma/pathology , Proto-Oncogene Proteins/metabolism , Proto-Oncogene Proteins/genetics
6.
Cell Commun Signal ; 22(1): 433, 2024 Sep 09.
Article in English | MEDLINE | ID: mdl-39252029

ABSTRACT

Bone sarcomas are malignant tumors of mesenchymal origin. Complete surgical resection is the cornerstone of multidisciplinary treatment. However, advanced, unresectable forms remain incurable. A crucial step towards addressing this challenge involves comprehending the molecular mechanisms underpinning tumor progression and metastasis, laying the groundwork for innovative precision medicine-based interventions. We previously showed that tyrosine kinase receptor Ephrin Type-A Receptor 2 (EphA2) is overexpressed in bone sarcomas. EphA2 is a key oncofetal protein implicated in metastasis, self-renewal, and chemoresistance. Molecular, genetic, biochemical, and pharmacological approaches have been developed to target EphA2 and its signaling pathway aiming to interfere with its tumor-promoting effects or as a carrier for drug delivery. This review synthesizes the main functions of EphA2 and their relevance in bone sarcomas, providing strategies devised to leverage this receptor for diagnostic and therapeutic purposes, with a focus on its applicability in the three most common bone sarcoma histotypes: osteosarcoma, chondrosarcoma, and Ewing sarcoma.


Subject(s)
Bone Neoplasms , Receptor, EphA2 , Signal Transduction , Humans , Receptor, EphA2/metabolism , Receptor, EphA2/genetics , Bone Neoplasms/metabolism , Bone Neoplasms/pathology , Bone Neoplasms/genetics , Animals , Osteosarcoma/pathology , Osteosarcoma/metabolism , Osteosarcoma/genetics , Molecular Targeted Therapy , Sarcoma/metabolism , Sarcoma/genetics , Sarcoma/pathology
7.
Cell Rep Med ; 5(9): 101728, 2024 Sep 17.
Article in English | MEDLINE | ID: mdl-39293390

ABSTRACT

Despite advances in treatment, the prognosis of patients with osteosarcoma remains unsatisfactory, and searching for potential targets is imperative. Here, we identify N4-acetylcytidine (ac4C) acetyltransferase 10 (NAT10) as a candidate therapeutic target in osteosarcoma through functional screening. NAT10 overexpression is correlated with a poor prognosis, and NAT10 knockout inhibits osteosarcoma progression. Mechanistically, NAT10 enhances mRNA stability of activating transcription factor 4 (ATF4) through ac4C modification. ATF4 induces the transcription of asparagine synthetase (ASNS), which catalyzes asparagine (Asn) biosynthesis, facilitating osteosarcoma progression. Utilizing virtual screening, we identify paliperidone and AG-401 as potential NAT10 inhibitors, and both inhibitors are found to bind to NAT10 proteins. Inhibiting NAT10 suppresses osteosarcoma progression in vivo. Combined treatment using paliperidone and AG-401 produces synergistic inhibition for osteosarcoma in patient-derived xenograft (PDX) models. Our findings demonstrate that NAT10 facilitates osteosarcoma progression through the ATF4/ASNS/Asn axis, and pharmacological inhibition of NAT10 may be a feasible therapeutic approach for osteosarcoma.


Subject(s)
Activating Transcription Factor 4 , Asparagine , Aspartate-Ammonia Ligase , Osteosarcoma , Humans , Osteosarcoma/drug therapy , Osteosarcoma/pathology , Osteosarcoma/metabolism , Osteosarcoma/genetics , Activating Transcription Factor 4/metabolism , Activating Transcription Factor 4/genetics , Animals , Cell Line, Tumor , Aspartate-Ammonia Ligase/metabolism , Aspartate-Ammonia Ligase/genetics , Aspartate-Ammonia Ligase/antagonists & inhibitors , Mice , Asparagine/metabolism , Disease Progression , Xenograft Model Antitumor Assays , Bone Neoplasms/pathology , Bone Neoplasms/drug therapy , Bone Neoplasms/metabolism , Bone Neoplasms/genetics , Gene Expression Regulation, Neoplastic/drug effects , Cell Proliferation/drug effects , Mice, Nude , Male , Female
8.
Int J Biol Sci ; 20(11): 4114-4127, 2024.
Article in English | MEDLINE | ID: mdl-39247831

ABSTRACT

Osteosarcoma is the most prevalent form of primary malignant bone tumor, primarily affecting children and adolescents. The nerve growth factors (NGF) referred to as neurotrophins have been associated with cancer-induced bone pain; however, the role of NGF in osteosarcoma has yet to be elucidated. In osteosarcoma samples from the Genomic Data Commons data portal, we detected higher levels of NGF and M2 macrophage markers, but not M1 macrophage markers. In cellular experiments, NGF-stimulated osteosarcoma conditional medium was shown to facilitate macrophage polarization from the M0 to the M2 phenotype. NGF also enhanced VCAM-1-dependent monocyte adhesion within the osteosarcoma microenvironment by down-regulating miR-513c-5p levels through the FAK and c-Src cascades. In in vivo xenograft models, the overexpression of NGF was shown to enhance tumor growth, while the oral administration of the TrK inhibitor larotrectinib markedly antagonized NGF-promoted M2 macrophage expression and tumor progression. These results suggest that larotrectinib could potentially be used as a therapeutic agent aimed at mitigating NGF-mediated osteosarcoma progression.


Subject(s)
Monocytes , Nerve Growth Factor , Osteosarcoma , Tumor Microenvironment , Vascular Cell Adhesion Molecule-1 , Osteosarcoma/metabolism , Osteosarcoma/drug therapy , Osteosarcoma/pathology , Humans , Nerve Growth Factor/metabolism , Animals , Tumor Microenvironment/drug effects , Monocytes/metabolism , Monocytes/drug effects , Vascular Cell Adhesion Molecule-1/metabolism , Mice , Cell Adhesion/drug effects , Cell Line, Tumor , Bone Neoplasms/metabolism , Bone Neoplasms/drug therapy , Bone Neoplasms/pathology , Macrophages/metabolism , Pyrimidines/pharmacology , Pyrimidines/therapeutic use , Pyrazoles/pharmacology , Pyrazoles/therapeutic use , Mice, Nude
9.
J Biochem Mol Toxicol ; 38(9): e23833, 2024 Sep.
Article in English | MEDLINE | ID: mdl-39243199

ABSTRACT

Osteosarcoma (OS) is the most frequent bone malignancy in humans. Previous evidence suggest that circ_0032463 is an oncogenic circular RNA (circRNA) in various cancers, including OS. However, the molecular mechanism of circ_0032463 involved in OS is still unclear. Circ_0032463, microRNA-145-5p (miR-145-5p), GDNF receptor alpha 1 (GFRA1), and Wilms tumor 1-associated protein (WTAP) levels were determined using real-time quantitative polymerase chain reaction (RT-qPCR). Cell proliferation, apoptosis, migration, invasion, and angiogenesis were analyzed using 5-ethynyl-2'-deoxyuridine (EdU), flow cytometry, transwell, and tube formation assays. Western blot analysis was performed to measure matrix metalloproteinase 2 (MMP2), MMP9, GFRA1, and WTAP protein levels. Binding between miR-145-5p and circ_0032463 or GFRA1 was confirmed using a dual-luciferase reporter and pull-down assay. The biological role of circ_0032463 on OS cell growth was also analyzed using a xenograft tumor model in vivo. Methylated RNA immunoprecipitation assay validated the interaction between WTAP and circ_0032463. Circ_0032463, GFRA1, and WTAP levels were increased, and miR-145-5p was decreased in OS tissues and cells. Circ_0032463 deficiency might hinder OS cell proliferation, migration, invasion, angiogenesis, and promote apoptosis in vitro. Mechanically, circ_0032463 worked as a miR-145-5p sponge to increase GFRA1 expression. Repression of circ_0032463 knockdown on tumor cell growth was proved in vivo. Besides, N6-methyladenosine (m6A) modification facilitates the biogenesis of circ_0032463. Taken together, m6A-mediated biogenesis of circ_0032463 facilitates OS cell malignant biological behavior partly via regulating the miR-145-5p/GFRA1 axis, suggesting a promising molecular marker for OS treatment.


Subject(s)
Bone Neoplasms , Glial Cell Line-Derived Neurotrophic Factor Receptors , MicroRNAs , Osteosarcoma , RNA, Circular , MicroRNAs/genetics , MicroRNAs/metabolism , Humans , Osteosarcoma/genetics , Osteosarcoma/pathology , Osteosarcoma/metabolism , RNA, Circular/genetics , RNA, Circular/metabolism , Glial Cell Line-Derived Neurotrophic Factor Receptors/genetics , Glial Cell Line-Derived Neurotrophic Factor Receptors/metabolism , Animals , Cell Line, Tumor , Mice , Bone Neoplasms/genetics , Bone Neoplasms/metabolism , Bone Neoplasms/pathology , Gene Expression Regulation, Neoplastic , RNA Splicing Factors/genetics , RNA Splicing Factors/metabolism , Mice, Nude , Male , Mice, Inbred BALB C , Cell Proliferation/genetics , Disease Progression , Female , RNA, Neoplasm/genetics , RNA, Neoplasm/metabolism , Adenosine/analogs & derivatives , Cell Cycle Proteins
10.
J Biochem Mol Toxicol ; 38(9): e23846, 2024 Sep.
Article in English | MEDLINE | ID: mdl-39243204

ABSTRACT

As a subclass of noncoding RNAs, circular RNA play an important role in tumour development. The aim of this study was to investigate the role of circ_0004674 in osteosarcoma glycolysis and the molecular mechanism of its regulation. We examined the expression of circ_0004674, miR-140-3p, TCF4 and glycolysis-related proteins (including HK2, PKM2, GLUT1 and LDHA) in osteosarcoma cells and tissues by quantitative reverse transcription-polymerase chain reaction and immunoblotting (Western blot analysis). The role of circ_0004674, miR-140-3p and TCF4 in the proliferation, apoptosis, migration and invasion of OS cells was examined using CCK8 assay, Apoptosis assay, Wound healing assay, Transwell migration and Matrigel invasion assay. The interaction of circ_0004674/miR-140-3p and miR-1543/TCF4 was also analysed using a dual luciferase reporter assay. Finally, the glycolytic process was assessed by glucose uptake assays and lactate production measurements. The results showed that the expression of circ_0004674 and TCF4 was significantly higher in MG63 and U2OS cells compared to hFOB1.19 cells, while the expression of miR-140-3p was downregulated. Silencing of circ_0004674 gene significantly inhibited the proliferation, migration and invasion of cancer cells and promoted apoptosis of cancer cells. Experiments such as dual luciferase reporter analysis showed that circ_0004674 regulates the expression of glycolysis-related proteins through the miR-140-3p/TCF4 pathway, and inhibition of this gene attenuated the depletion of glucose content and the production of lactate in cancer cells. Furthermore, inhibition of miR-140-3p or overexpression of TCF could reverse the phenotypic changes in cancer cells induced by circ_0004674 silencing. In summary, this study elucidated the specific function and potential mechanisms of circ_0004674 in osteosarcoma glycolysis. The findings demonstrate that miR-140-3p and TCF4 function respectively as a tumor suppressor gene and an oncogene in osteosarcoma. Notably, they influence glycolysis and associated pathways, regulating osteosarcoma proliferation. Therefore, circ_0004674 promotes osteosarcoma glycolysis and proliferation through the miR-140-3p/TCF4 pathway, enhancing the malignant behaviour of tumours, and it is expected to be a potential molecular target for osteosarcoma treatment.


Subject(s)
Cell Proliferation , Glycolysis , MicroRNAs , Osteosarcoma , RNA, Circular , Transcription Factor 4 , Humans , Osteosarcoma/genetics , Osteosarcoma/metabolism , Osteosarcoma/pathology , MicroRNAs/genetics , MicroRNAs/metabolism , Cell Line, Tumor , RNA, Circular/genetics , RNA, Circular/metabolism , Transcription Factor 4/metabolism , Transcription Factor 4/genetics , Bone Neoplasms/genetics , Bone Neoplasms/metabolism , Bone Neoplasms/pathology , Cell Movement , Gene Expression Regulation, Neoplastic , Apoptosis/genetics , Signal Transduction
11.
Int J Mol Med ; 54(5)2024 11.
Article in English | MEDLINE | ID: mdl-39219279

ABSTRACT

Metastasis is the leading cause of cancer­related death in osteosarcoma (OS). OS stem cells (OSCs) and anoikis resistance are considered to be essential for tumor metastasis formation. However, the underlying mechanisms involved in the maintenance of a stem­cell phenotype and anoikis resistance in OS are mostly unknown. Fos­like antigen 1 (FOSL1) is important in maintaining a stem­like phenotype in various cancers; however, its role in OSCs and anoikis resistance remains unclear. In the present study, the dynamic expression patterns of FOSL1 were investigated during the acquisition of cancer stem­like properties using RNA sequencing, PCR, western blotting and immunofluorescence. Flow cytometry, tumor­sphere formation, clone formation assays, anoikis assays, western blotting and in vivo xenograft and metastasis models were used to further investigate the responses of the stem­cell phenotype and anoikis resistance to FOSL1 overexpression or silencing in OS cell lines. The underlying molecular mechanisms were evaluated, focusing on whether SOX2 is crucially involved in FOSL1­mediated stemness and anoikis in OS. FOSL1 expression was observed to be upregulated in OSCs and promoted tumor­sphere formation, clone formation and tumorigenesis in OS cells. FOSL1 expression correlated positively with the expression of stemness­related factors (SOX2, NANOG, CD117 and Stro1). Moreover, FOSL1 facilitated OS cell anoikis resistance and promoted metastases by regulating the expression of apoptosis related proteins BCL2 and BAX. Mechanistically, FOSL1 upregulated SOX2 expression by interacting with the SOX2 promoter and activating its transcription. The results also showed that SOX2 is critical for FOSL1­mediated stem­like properties and anoikis resistance. The current findings indicated that FOSL1 is an important regulator that promotes a stem cell­like phenotype and anoikis resistance to facilitate tumorigenesis and metastasis in OS by regulating the transcription of SOX2. Thus, FOSL1 might represent an attractive target for therapeutic interventions in OS.


Subject(s)
Anoikis , Carcinogenesis , Gene Expression Regulation, Neoplastic , Neoplastic Stem Cells , Osteosarcoma , Proto-Oncogene Proteins c-fos , SOXB1 Transcription Factors , Osteosarcoma/pathology , Osteosarcoma/genetics , Osteosarcoma/metabolism , Humans , Proto-Oncogene Proteins c-fos/metabolism , Proto-Oncogene Proteins c-fos/genetics , SOXB1 Transcription Factors/metabolism , SOXB1 Transcription Factors/genetics , Anoikis/genetics , Animals , Neoplastic Stem Cells/metabolism , Neoplastic Stem Cells/pathology , Cell Line, Tumor , Mice , Carcinogenesis/genetics , Carcinogenesis/pathology , Neoplasm Metastasis , Bone Neoplasms/pathology , Bone Neoplasms/genetics , Bone Neoplasms/metabolism , Bone Neoplasms/secondary , Mice, Nude , Male , Female , Mice, Inbred BALB C
12.
Cells ; 13(17)2024 Aug 28.
Article in English | MEDLINE | ID: mdl-39273017

ABSTRACT

PAK4 and PD-L1 have been suggested as novel therapeutic targets in human cancers. Moreover, PAK4 has been suggested to be a molecule closely related to the immune evasion of cancers. Therefore, this study evaluated the roles of PAK4 and PD-L1 in the progression of osteosarcomas in 32 osteosarcomas and osteosarcoma cells. In human osteosarcomas, immunohistochemical positivity for the expression of PAK4 (overall survival, p = 0.028) and PD-L1 (relapse-free survival, p = 0.002) were independent indicators for the survival of patients in a multivariate analysis. In osteosarcoma cells, the overexpression of PAK4 increased proliferation and invasiveness, while the knockdown of PAK4 suppressed proliferation and invasiveness. The expression of PAK4 was associated with the expression of the molecules related to cell cycle regulation, invasion, and apoptosis. PAK4 was involved in resistance to apoptosis under a treatment regime with doxorubicin for osteosarcoma. In U2OS cells, PAK4 was involved in the stabilization of PD-L1 from ubiquitin-mediated proteasomal degradation and the in vivo infiltration of immune cells such as regulatory T cells and PD1-, CD4-, and CD8-positive cells in mice tumors. In conclusion, this study suggests that PAK4 is involved in the progression of osteosarcoma by promoting proliferation, invasion, and resistance to doxorubicin and stabilized PD-L1 from proteasomal degradation.


Subject(s)
B7-H1 Antigen , Cell Proliferation , Doxorubicin , Drug Resistance, Neoplasm , Osteosarcoma , p21-Activated Kinases , Osteosarcoma/pathology , Osteosarcoma/drug therapy , Osteosarcoma/metabolism , Osteosarcoma/genetics , Humans , B7-H1 Antigen/metabolism , Female , Drug Resistance, Neoplasm/drug effects , Drug Resistance, Neoplasm/genetics , Doxorubicin/pharmacology , Doxorubicin/therapeutic use , Animals , p21-Activated Kinases/metabolism , p21-Activated Kinases/genetics , Male , Cell Line, Tumor , Cell Proliferation/drug effects , Mice , Apoptosis/drug effects , Bone Neoplasms/metabolism , Bone Neoplasms/pathology , Bone Neoplasms/drug therapy , Bone Neoplasms/genetics , Adult , Adolescent , Protein Stability/drug effects , Mice, Nude , Young Adult , Gene Expression Regulation, Neoplastic/drug effects , Neoplasm Invasiveness
13.
Lab Invest ; 104(9): 102122, 2024 Sep.
Article in English | MEDLINE | ID: mdl-39098628

ABSTRACT

The assessment of chemotherapy response in osteosarcoma (OS) based on the average percentage of viable cells is limited, as it overlooks the spatial heterogeneity of tumor cell response (foci of resistant cells), immune microenvironment, and bone microarchitecture. Despite the resulting positive classification for response to chemotherapy, some patients experience early metastatic recurrence, demonstrating that our conventional tools for evaluating treatment response are insufficient. We studied the interactions between tumor cells, immune cells (lymphocytes, histiocytes, and osteoclasts), and bone extracellular matrix (ECM) in 18 surgical resection samples of OS using multiplex and conventional immunohistochemistry (IHC: CD8, CD163, CD68, and SATB2), combined with multiscale characterization approaches in territories of good and poor response (GRT/PRT) to treatment. GRT and PRT were defined as subregions with <10% and ≥10% of viable tumor cells, respectively. Local correlations between bone ECM porosity and density of immune cells were assessed in these territories. Immune cell density was then correlated to overall patient survival. Two patterns were identified for histiocytes and osteoclasts. In poor responder patients, CD68 osteoclast density exceeded that of CD163 histiocytes but was not related to bone ECM load. Conversely, in good responder patients, CD163 histiocytes were more numerous than CD68 osteoclasts. For both of them, a significant negative local correlation with bone ECM porosity was found (P < .01). Moreover, in PRT, multinucleated osteoclasts were rounded and intermingled with tumor cells, whereas in GRT, they were elongated and found in close contact with bone trabeculae. CD8 levels were always low in metastatic patients, and those initially considered good responders rapidly died from their disease. The specific recruitment of histiocytes and osteoclasts within the bone ECM, and the level of CD8 represent new features of OS response to treatment. The associated prognostic signatures should be integrated into the therapeutic stratification algorithm of patients after surgery.


Subject(s)
Bone Neoplasms , Extracellular Matrix , Osteosarcoma , Tumor Microenvironment , Humans , Osteosarcoma/immunology , Osteosarcoma/pathology , Osteosarcoma/therapy , Osteosarcoma/metabolism , Bone Neoplasms/immunology , Bone Neoplasms/pathology , Female , Male , Extracellular Matrix/metabolism , Extracellular Matrix/pathology , Adult , Adolescent , Bone Matrix/metabolism , Young Adult , Child , Antigens, CD/metabolism
14.
Sci Rep ; 14(1): 19349, 2024 08 20.
Article in English | MEDLINE | ID: mdl-39164287

ABSTRACT

Inhibition of translation initiation using eIF4A inhibitors like (-)-didesmethylrocaglamide [(-)-DDR] and (-)-rocaglamide [(-)-Roc] is a potential cancer treatment strategy as they simultaneously diminish multiple oncogenic drivers. We showed that human and dog osteosarcoma cells expressed higher levels of eIF4A1/2 compared with mesenchymal stem cells. Genetic depletion of eIF4A1 and/or 2 slowed osteosarcoma cell growth. To advance preclinical development of eIF4A inhibitors, we demonstrated the importance of (-)-chirality in DDR for growth-inhibitory activity. Bromination of DDR at carbon-5 abolished growth-inhibitory activity, while acetylating DDR at carbon-1 was tolerated. Like (-)-DDR, (±)-DDR, and (-)-Roc, (±)-DDR-acetate increased γH2A.X levels and induced G2/M arrest and apoptosis. Consistent with translation inhibition, these rocaglates decreased the levels of several mitogenic kinases, the STAT3 transcription factor, and the stress-activated protein kinase p38. However, phosphorylated p38 was greatly enhanced in treated cells, suggesting activation of stress response pathways. RNA sequencing identified RHOB as a top upregulated gene in both (-)-DDR- and (-)-Roc-treated osteosarcoma cells, but the Rho inhibitor Rhosin did not enhance the growth-inhibitory activity of (-)-DDR or (-)-Roc. Nonetheless, these rocaglates potently suppressed tumor growth in a canine osteosarcoma patient-derived xenograft model. These results suggest that these eIF4A inhibitors can be leveraged to treat both human and dog osteosarcomas.


Subject(s)
Eukaryotic Initiation Factor-4A , Osteosarcoma , Dogs , Animals , Humans , Osteosarcoma/drug therapy , Osteosarcoma/pathology , Osteosarcoma/metabolism , Cell Line, Tumor , Eukaryotic Initiation Factor-4A/antagonists & inhibitors , Eukaryotic Initiation Factor-4A/metabolism , Mice , Antineoplastic Agents/pharmacology , Apoptosis/drug effects , Bone Neoplasms/drug therapy , Bone Neoplasms/pathology , Bone Neoplasms/metabolism , Bone Neoplasms/genetics , Xenograft Model Antitumor Assays , Cell Proliferation/drug effects , Benzofurans/pharmacology
15.
J Orthop Surg Res ; 19(1): 467, 2024 Aug 08.
Article in English | MEDLINE | ID: mdl-39118123

ABSTRACT

BACKGROUND: Osteosarcoma is a soft tissue neoplasm with elevated recurrence risk and highly metastatic potential. Metal response element binding transcriptional factor 2 (MTF2) has been revealed to exert multiple activities in human tissues. The present research was conducted to explore the functions and related response mechanism of MTF2 in osteosarcoma which have not been introduced yet. METHODS: Bioinformatics tools identified the differential MTF2 expression in osteosarcoma tissues. MTF2 expression in osteosarcoma cells was examined with Western blot. Cell Counting Kit-8 (CCK-8) assay, 5-Ethynyl-2'-deoxyuridine (EDU) staining, wound healing as well as transwell assays measured cell proliferation, migration and invasion, respectively. Flow cytometry assay detected the cellular apoptotic level. Western blot also measured the expressions of proteins associated with epithelial mesenchymal transition (EMT), apoptosis and enhancer of zeste homolog 2 (EZH2)/secreted frizzled-related protein 1 (SFRP1)/Wnt signaling. Co-immunoprecipitation (Co-IP) assay confirmed MTF2-EZH2 interaction. RESULTS: MTF2 expression was increased in osteosarcoma tissues and cells. MTF2 interference effectively inhibited the proliferation, migration and invasion of osteosarcoma cells and promoted the cellular apoptotic rate. MTF2 directly bound to EZH2 and MTF2 silence reduced EZH2 expression, activated SFRP1 expression and blocked Wnt signaling in osteosarcoma cells. EZH2 upregulation or SFRP1 antagonist WAY-316606 partly counteracted the impacts of MTF2 down-regulation on the SFRP1/Wnt signaling and the biological phenotypes of osteosarcoma cells. CONCLUSIONS: MTF2 might down-regulate SFRP1 to activate Wnt signaling and drive the progression of osteosarcoma via interaction with EZH2 protein.


Subject(s)
Bone Neoplasms , Cell Proliferation , Enhancer of Zeste Homolog 2 Protein , Osteosarcoma , Wnt Signaling Pathway , Humans , Apoptosis/physiology , Bone Neoplasms/metabolism , Bone Neoplasms/pathology , Bone Neoplasms/genetics , Cell Line, Tumor , Cell Movement/physiology , Cell Proliferation/physiology , Disease Progression , Enhancer of Zeste Homolog 2 Protein/metabolism , Enhancer of Zeste Homolog 2 Protein/genetics , Gene Expression Regulation, Neoplastic , Intercellular Signaling Peptides and Proteins/metabolism , Intercellular Signaling Peptides and Proteins/genetics , Membrane Proteins/metabolism , Membrane Proteins/genetics , Osteosarcoma/metabolism , Osteosarcoma/pathology , Osteosarcoma/genetics , Transcription Factors/metabolism , Transcription Factors/genetics , Wnt Signaling Pathway/physiology , Polycomb Repressive Complex 2/genetics , Polycomb Repressive Complex 2/metabolism
16.
Cancer Rep (Hoboken) ; 7(8): e2159, 2024 Aug.
Article in English | MEDLINE | ID: mdl-39118262

ABSTRACT

BACKGROUND: Ring finger protein 135 (RNF135) is an E3 ubiquitin ligase that has been implicated in the tumorigenesis of multiple human malignancies. However, whether RNF135 plays a role in the development of human osteosarcoma (OS) remains unknown. METHODS: RNF135 expression in 20 human OS and 20 human osteochondroma specimens were evaluated by means of immunohistochemistry staining. The effects of shRNA-mediated RNF135 knockdown on human OS cell growth and apoptosis were evaluated through a panel of in vitro studies on cell proliferation, colony formation, exposure of phosphatidylserine on the cell surface, and caspase 3/7 activation. The protein levels of PI3K, AKT, and p-AKT were determined by western blot analysis. RESULTS: We detected significantly higher RNF135 levels in human OS tissues than human osteochondroma tissues. In in vitro studies, shRNA-mediated RNF135 knockdown in human OS cells inhibited proliferation and induced apoptosis. In addition, RNF135 knockdown reduced PI3K and p-AKT protein levels and activated caspase 3 and 7. CONCLUSIONS: These results supported that RNF135 contributes to human OS development through PI3K/AKT-dependent mechanisms. Targeting RNF135 may provide a new therapeutic approach for treating this human malignancy.


Subject(s)
Apoptosis , Bone Neoplasms , Cell Proliferation , Osteosarcoma , Phosphatidylinositol 3-Kinases , Proto-Oncogene Proteins c-akt , Signal Transduction , Ubiquitin-Protein Ligases , Female , Humans , Male , Bone Neoplasms/pathology , Bone Neoplasms/genetics , Bone Neoplasms/metabolism , Cell Line, Tumor , Gene Expression Regulation, Neoplastic , Gene Knockdown Techniques , Osteochondroma/pathology , Osteochondroma/genetics , Osteochondroma/metabolism , Osteosarcoma/pathology , Osteosarcoma/genetics , Osteosarcoma/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Ubiquitin-Protein Ligases/genetics , Ubiquitin-Protein Ligases/metabolism , Up-Regulation
17.
J Cell Mol Med ; 28(16): e70021, 2024 Aug.
Article in English | MEDLINE | ID: mdl-39153212

ABSTRACT

Accumulating evidence has demonstrated that F-box protein 22 (FBXO22) participates in tumour development and progression in various types of human malignancies. However, the functions and detailed molecular mechanisms of FBXO22 in osteosarcoma tumorigenesis and progression remain elusive. In this study, we aimed to determine the effects of FBXO22 on the cell proliferation, migration and invasion of osteosarcoma cells using cell counting kit-8 and Matrigel Transwell approaches. Moreover, we explored the molecular mechanisms by which FBXO22 mediated oncogenesis and progression in osteosarcoma via Western blotting, immunoprecipitation and ubiquitination. We found that FBXO22 depletion inhibited the proliferation, migration and invasion of osteosarcoma cells, whereas FBXO22 overexpression increased the proliferation and motility of osteosarcoma cells. Mechanistically, FBXO22 promoted the ubiquitination and degradation of FoxO1 in osteosarcoma cells. FBXO22 depletion reduced cell proliferation and motility via regulation of FoxO1. Taken together, our findings provide new insight into FBXO22-induced osteosarcoma tumorigenesis. The inhibition of FBXO22 could be a promising strategy for the treatment of osteosarcoma.


Subject(s)
Cell Movement , Cell Proliferation , F-Box Proteins , Forkhead Box Protein O1 , Gene Expression Regulation, Neoplastic , Osteosarcoma , Ubiquitination , Osteosarcoma/metabolism , Osteosarcoma/pathology , Osteosarcoma/genetics , Humans , Forkhead Box Protein O1/metabolism , Forkhead Box Protein O1/genetics , F-Box Proteins/metabolism , F-Box Proteins/genetics , Cell Movement/genetics , Cell Line, Tumor , Proteolysis , Disease Progression , Bone Neoplasms/metabolism , Bone Neoplasms/pathology , Bone Neoplasms/genetics , Neoplasm Invasiveness , Carcinogenesis/genetics , Carcinogenesis/metabolism , Carcinogenesis/pathology , Receptors, Cytoplasmic and Nuclear
18.
Sci Rep ; 14(1): 18720, 2024 08 12.
Article in English | MEDLINE | ID: mdl-39134572

ABSTRACT

ARHGAP25, a member of the ARHGAP family, encodes a negative regulator of Rho-GTPase that is important for actin remodeling, cell polarity, and cell migration. ARHGAP25 is down-regulated in a variety of solid tumors and promotes cancer cell growth, migration, and invasion. However, nothing is understood about ARHGAP25's biological function in osteosarcoma. This work used qPCR and WB to confirm the expression of ARHGAP25 in osteosarcoma following the initial analysis of its expression in pan-cancer. For GO and KEGG analysis, we have chosen 300 genes from the TARGET osteosarcoma data that had the strongest positive correlation with ARHGAP25, and we created nomogram and calibration charts. We simultaneously overexpressed ARHGAP25 in osteosarcoma cells to examine its impact on apoptosis and proliferation. By using MSP, we determined their methylation status in osteosarcoma cells and normal bone cells. We observed that ARHGAP25 was significantly downregulated in a range of malignancies, including osteosarcoma, and was associated with poor patient outcomes. The decrease of ARHGAP25 expression in osteosarcoma is related to DNA methylation. Overexpression of ARHGAP25 induced apoptosis and inhibited the proliferation of osteosarcoma cells in vitro. In addition, ARHGAP25 is also associated with immune-related pathways in osteosarcoma. These findings suggest that ARHGAP25 is a valuable prognostic biomarker in osteosarcoma patients.


Subject(s)
Apoptosis , Bone Neoplasms , Cell Proliferation , Computational Biology , DNA Methylation , GTPase-Activating Proteins , Gene Expression Regulation, Neoplastic , Osteosarcoma , Osteosarcoma/genetics , Osteosarcoma/pathology , Osteosarcoma/metabolism , Humans , GTPase-Activating Proteins/genetics , GTPase-Activating Proteins/metabolism , Computational Biology/methods , Bone Neoplasms/genetics , Bone Neoplasms/pathology , Bone Neoplasms/metabolism , Bone Neoplasms/mortality , Apoptosis/genetics , Cell Line, Tumor , Cell Proliferation/genetics , Prognosis , Male , Female , Biomarkers, Tumor/genetics , Biomarkers, Tumor/metabolism , Clinical Relevance
19.
Oncotarget ; 15: 535-540, 2024 Aug 02.
Article in English | MEDLINE | ID: mdl-39102216

ABSTRACT

WNT signaling regulates osteosarcoma proliferation. However, there is controversy in the field of osteosarcoma as to whether WNT signaling is pro- or anti-tumorigenic. WNT-targeting therapeutics, both activators and inhibitors, are compared. WNT5B, a ß-catenin-independent ligand, and WNT10B, a ß-catenin-dependent WNT ligand, are each expressed in osteosarcomas, but they are not expressed in the same tumors. Furthermore, WNT10B and WNT5B regulate different histological subtypes of osteosarcomas. Using WNT signaling modulators as therapeutics may depend on the WNT ligand and/or the activated signaling pathway.


Subject(s)
Bone Neoplasms , Osteosarcoma , Wnt Proteins , Wnt Signaling Pathway , Osteosarcoma/metabolism , Osteosarcoma/pathology , Osteosarcoma/drug therapy , Humans , Wnt Proteins/metabolism , Wnt Proteins/antagonists & inhibitors , Bone Neoplasms/metabolism , Bone Neoplasms/pathology , Bone Neoplasms/drug therapy , Wnt Signaling Pathway/drug effects , Animals , Proto-Oncogene Proteins/metabolism , Proto-Oncogene Proteins/genetics , Molecular Targeted Therapy , Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use , beta Catenin/metabolism , Gene Expression Regulation, Neoplastic
20.
BMC Cancer ; 24(1): 928, 2024 Aug 01.
Article in English | MEDLINE | ID: mdl-39090568

ABSTRACT

BACKGROUND: Osteosarcoma (OS) is one of the most common primary malignant tumors of bone in children, which develops from osteoblasts and typically occurs during the rapid growth phase of the bone. Recently, Super-Enhancers(SEs)have been reported to play a crucial role in osteosarcoma growth and metastasis. Therefore, there is an urgent need to identify specific targeted inhibitors of SEs to assist clinical therapy. This study aimed to elucidate the role of BRD4 inhibitor GNE-987 targeting SEs in OS and preliminarily explore its mechanism. METHODS: We evaluated changes in osteosarcoma cells following treatment with a BRD4 inhibitor GNE-987. We assessed the anti-tumor effect of GNE-987 in vitro and in vivo by Western blot, CCK8, flow cytometry detection, clone formation, xenograft tumor size measurements, and Ki67 immunohistochemical staining, and combined ChIP-seq with RNA-seq techniques to find its anti-tumor mechanism. RESULTS: In this study, we found that extremely low concentrations of GNE-987(2-10 nM) significantly reduced the proliferation and survival of OS cells by degrading BRD4. In addition, we found that GNE-987 markedly induced cell cycle arrest and apoptosis in OS cells. Further study indicated that VHL was critical for GNE-987 to exert its antitumor effect in OS cells. Consistent with in vitro results, GNE-987 administration significantly reduced tumor size in xenograft models with minimal toxicity, and partially degraded the BRD4 protein. KRT80 was identified through analysis of the RNA-seq and ChIP-seq data. U2OS HiC analysis suggested a higher frequency of chromatin interactions near the KRT80 binding site. The enrichment of H3K27ac modification at KRT80 was significantly reduced after GNE-987 treatment. KRT80 was identified as playing an important role in OS occurrence and development. CONCLUSIONS: This research revealed that GNE-987 selectively degraded BRD4 and disrupted the transcriptional regulation of oncogenes in OS. GNE-987 has the potential to affect KRT80 against OS.


Subject(s)
Apoptosis , Bone Neoplasms , Cell Cycle Proteins , Cell Proliferation , Osteosarcoma , Transcription Factors , Animals , Humans , Mice , Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use , Apoptosis/drug effects , Bone Neoplasms/drug therapy , Bone Neoplasms/pathology , Bone Neoplasms/genetics , Bone Neoplasms/metabolism , Bromodomain Containing Proteins , Cell Cycle Proteins/metabolism , Cell Cycle Proteins/genetics , Cell Cycle Proteins/antagonists & inhibitors , Cell Line, Tumor , Cell Proliferation/drug effects , Enhancer Elements, Genetic , Gene Expression Regulation, Neoplastic/drug effects , Mice, Nude , Osteosarcoma/drug therapy , Osteosarcoma/pathology , Osteosarcoma/genetics , Osteosarcoma/metabolism , Transcription Factors/metabolism , Transcription Factors/antagonists & inhibitors , Transcription Factors/genetics , Xenograft Model Antitumor Assays
SELECTION OF CITATIONS
SEARCH DETAIL