Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 20 de 4.809
Filtrer
1.
Neoplasma ; 71(3): 279-288, 2024 Jun.
Article de Anglais | MEDLINE | ID: mdl-38958715

RÉSUMÉ

Osteosarcoma (OS) is a common primary bone tumor in children and adolescents. Circular RNA (circRNA)-IARS acts as an oncogene in multiple human tumors. However, the circ-IARS function in OS is unclear. This research aimed to elucidate the roles and mechanisms of circ-IARS in OS. In this study, circ-IARS expressions were raised in OS tissues and cells. circ-IARS expressions were closely related to clinical stage and distant metastasis. Furthermore, overall survival rates were reduced in OS patients with high circ-IARS levels. Also, silencing circ-IARS weakened OS cell proliferation and invasion, yet enhanced cell ferroptosis. Mechanistically, circ-IARS targeted miR-188-5p to regulate RAB14 expressions in OS cells. Moreover, circ-IARS knockdown repressed OS cell proliferation, invasion, and induced ferroptosis, yet these impacts were abolished by co-transfection with anti-miR-188-5p or pcDNA-RAB14. Meanwhile, interference with circ-IARS reduced OS cell proliferation, and decreased RAB14 (a member of the RAS oncogene family), GPX4, and xCT (crucial ferroptosis regulators) expressions in vivo. In conclusion, circ-IARS facilitated OS progression via miR-188-5p/RAB14.


Sujet(s)
Tumeurs osseuses , Prolifération cellulaire , Ferroptose , microARN , Ostéosarcome , ARN circulaire , Protéines G rab , Humains , Ostéosarcome/génétique , Ostéosarcome/anatomopathologie , Ostéosarcome/métabolisme , microARN/génétique , ARN circulaire/génétique , Protéines G rab/génétique , Protéines G rab/métabolisme , Ferroptose/génétique , Tumeurs osseuses/génétique , Tumeurs osseuses/anatomopathologie , Tumeurs osseuses/métabolisme , Mâle , Lignée cellulaire tumorale , Femelle , Évolution de la maladie , Souris , Animaux , Régulation de l'expression des gènes tumoraux
2.
Exp Biol Med (Maywood) ; 249: 10161, 2024.
Article de Anglais | MEDLINE | ID: mdl-38966281

RÉSUMÉ

Osteosarcoma is a form of bone cancer that predominantly impacts osteoblasts, the cells responsible for creating fresh bone tissue. Typical indications include bone pain, inflammation, sensitivity, mobility constraints, and fractures. Utilising imaging techniques such as X-rays, MRI scans, and CT scans can provide insights into the size and location of the tumour. Additionally, a biopsy is employed to confirm the diagnosis. Analysing genes with distinct expression patterns unique to osteosarcoma can be valuable for early detection and the development of effective treatment approaches. In this research, we comprehensively examined the entire transcriptome and pinpointed genes with altered expression profiles specific to osteosarcoma. The study mainly aimed to identify the molecular fingerprint of osteosarcoma. In this study, we processed 90 FFPE samples from PathWest with an almost equal number of osteosarcoma and healthy tissues. RNA was extracted from Paraffin-embedded tissue; RNA was sequenced, the sequencing data was analysed, and gene expression was compared to the healthy samples of the same patients. Differentially expressed genes in osteosarcoma-derived samples were identified, and the functions of those genes were explored. This result was combined with our previous studies based on FFPE and fresh samples to perform a meta-analysis. We identified 1,500 identical differentially expressed genes in PathWest osteosarcoma samples compared to normal tissue samples of the same patients. Meta-analysis with combined fresh tissue samples identified 530 differentially expressed genes. IFITM5, MMP13, PANX3, and MAGEA6 were some of the most overexpressed genes in osteosarcoma samples, while SLC4A1, HBA1, HBB, AQP7 genes were some of the top downregulated genes. Through the meta-analysis, 530 differentially expressed genes were identified to be identical among FFPE (105 FFPE samples) and 36 fresh bone samples. Deconvolution analysis with single-cell RNAseq data confirmed the presence of specific cell clusters in FFPE samples. We propose these 530 DEGs as a molecular fingerprint of osteosarcoma.


Sujet(s)
Tumeurs osseuses , Analyse de profil d'expression de gènes , Ostéosarcome , Ostéosarcome/génétique , Ostéosarcome/anatomopathologie , Humains , Analyse de profil d'expression de gènes/méthodes , Tumeurs osseuses/génétique , Tumeurs osseuses/anatomopathologie , Tumeurs osseuses/métabolisme , Inclusion en paraffine , Transcriptome/génétique , Régulation de l'expression des gènes tumoraux , Fixation tissulaire , Formaldéhyde
3.
Sci Rep ; 14(1): 15557, 2024 Jul 05.
Article de Anglais | MEDLINE | ID: mdl-38969706

RÉSUMÉ

Metastasis is driven by extensive cooperation between a tumor and its microenvironment, resulting in the adaptation of molecular mechanisms that evade the immune system and enable pre-metastatic niche (PMN) formation. Little is known of the tumor-intrinsic factors that regulate these mechanisms. Here we show that expression of the transcription factor interferon regulatory factor 5 (IRF5) in osteosarcoma (OS) and breast carcinoma (BC) clinically correlates with prolonged survival and decreased secretion of tumor-derived extracellular vesicles (t-dEVs). Conversely, loss of intra-tumoral IRF5 establishes a PMN that supports metastasis. Mechanistically, IRF5-positive tumor cells retain IRF5 transcripts within t-dEVs that contribute to altered composition, secretion, and trafficking of t-dEVs to sites of metastasis. Upon whole-body pre-conditioning with t-dEVs from IRF5-high or -low OS and BC cells, we found increased lung metastatic colonization that replicated findings from orthotopically implanted cancer cells. Collectively, our findings uncover a new role for IRF5 in cancer metastasis through its regulation of t-dEV programming of the PMN.


Sujet(s)
Tumeurs du sein , Vésicules extracellulaires , Facteurs de régulation d'interféron , Métastase tumorale , Microenvironnement tumoral , Vésicules extracellulaires/métabolisme , Facteurs de régulation d'interféron/métabolisme , Facteurs de régulation d'interféron/génétique , Humains , Animaux , Souris , Lignée cellulaire tumorale , Tumeurs du sein/anatomopathologie , Tumeurs du sein/génétique , Tumeurs du sein/métabolisme , Femelle , Ostéosarcome/anatomopathologie , Ostéosarcome/génétique , Ostéosarcome/métabolisme , Tumeurs du poumon/secondaire , Tumeurs du poumon/anatomopathologie , Tumeurs du poumon/génétique , Tumeurs du poumon/métabolisme , Régulation de l'expression des gènes tumoraux
4.
Aging (Albany NY) ; 16(12): 10462-10476, 2024 Jun 17.
Article de Anglais | MEDLINE | ID: mdl-38889378

RÉSUMÉ

BACKGROUND: Osteosarcoma is a highly malignant bone tumor that exhibits rapid growth and early metastasis. Hypoxia plays a pivotal role in promoting the proliferation and metastasis of osteosarcoma through a series of molecular events, which are partially mediated and regulated by HIF-1α. However, the regulatory network associated with HIF-1α in osteosarcoma remains limited. Therefore, the objective of this study was to identify critical hypoxia-associated genes and investigate their effects and molecular mechanisms in osteosarcoma cells. METHODS: Through bioinformatics analysis, matrilin-4 (MATN4) was identified as a crucial gene associated with hypoxia. The expression of MATN4 and HIF-1α was assessed using immunohistochemistry, RT-qPCR, and western blotting. The proliferative capacity of osteosarcoma cells was assessed through the utilization of CCK-8, EDU staining, and colony formation assays. The effects of MATN4 on the mobility of OS cells were evaluated using wound-healing assays and transwell assays. The interaction between MATN4 and HIF-1α was detected through chromatin immunoprecipitation. RESULTS: MATN4 is overexpressed in osteosarcoma tissue and cells, particularly in osteosarcoma cells with high metastatic potential. Knockdown of MATN4 inhibits the proliferation, migration, and invasion abilities of osteosarcoma cells and reverses the promoting effects of hypoxia on these functions. Additionally, HIF-1α binds to MATN4 and upregulates its expression. Interestingly, knockdown of HIF-1α reduces the stimulatory effects of MATN4 overexpression on the proliferation, migration, and invasion of osteosarcoma cells under hypoxic conditions. CONCLUSIONS: Taken together, our results suggest that MATN4 is regulated by HIF-1α and confers a more aggressive phenotype on OS cells. This evidence suggests that MATN4 may act as a potential target for OS diagnosis and treatment.


Sujet(s)
Tumeurs osseuses , Prolifération cellulaire , Régulation de l'expression des gènes tumoraux , Sous-unité alpha du facteur-1 induit par l'hypoxie , Ostéosarcome , Ostéosarcome/génétique , Ostéosarcome/anatomopathologie , Ostéosarcome/métabolisme , Humains , Sous-unité alpha du facteur-1 induit par l'hypoxie/métabolisme , Sous-unité alpha du facteur-1 induit par l'hypoxie/génétique , Prolifération cellulaire/génétique , Tumeurs osseuses/génétique , Tumeurs osseuses/anatomopathologie , Tumeurs osseuses/métabolisme , Lignée cellulaire tumorale , Mouvement cellulaire/génétique , Métastase tumorale
5.
J Cell Mol Med ; 28(11): e18462, 2024 Jun.
Article de Anglais | MEDLINE | ID: mdl-38847478

RÉSUMÉ

Osteosarcoma (OS) is the most common primary malignant bone tumour in children and young adults. Account for 80% of all OS cases, conventional OS are characterized by the presence of osteoblastic, chondroblastic and fibroblastic cell types. Despite this heterogeneity, therapeutic treatment and prognosis of OS are essentially the same for all OS subtypes. Here, we report that DEC2, a transcriptional repressor, is expressed at higher levels in chondroblastic OS compared with osteoblastic OS. This difference suggests that DEC2 is disproportionately involved in the progression of chondroblastic OS, and thus, DEC2 may represent a possible molecular target for treating this type of OS. In the human chondroblastic-like OS cell line MNNG/HOS, we found that overexpression of DEC2 affects the proliferation of the cells by activating the VEGFC/VEGFR2 signalling pathway. Enhanced expression of DEC2 increased VEGFR2 expression, as well as increased the phosphorylation levels at sites Y951 and Y1175 of VEGFR2. On the one hand, activation of VEGFR2Y1175 enhanced cell proliferation through VEGFR2Y1175-PLCγ1-PKC-SPHK-MEK-ERK signalling. On the other hand, activation of VEGFR2Y951 decreased mitochondria-dependent apoptosis rate through VEGFR2Y951-VARP-PI3K-AKT signalling. Activation of these two signalling pathways resulted in enhanced progression of chondroblastic OS. In conclusion, DEC2 plays a pivotal role in cell proliferation and apoptosis-resistance in chondroblastic OS via the VEGFC/VEGFR2 signalling pathway. These findings lay the groundwork for developing focused treatments that target specific types of OS.


Sujet(s)
Tumeurs osseuses , Prolifération cellulaire , Régulation de l'expression des gènes tumoraux , Ostéosarcome , Transduction du signal , Facteur de croissance endothéliale vasculaire de type C , Récepteur-2 au facteur croissance endothéliale vasculaire , Humains , Ostéosarcome/métabolisme , Ostéosarcome/anatomopathologie , Ostéosarcome/génétique , Récepteur-2 au facteur croissance endothéliale vasculaire/métabolisme , Récepteur-2 au facteur croissance endothéliale vasculaire/génétique , Lignée cellulaire tumorale , Tumeurs osseuses/métabolisme , Tumeurs osseuses/anatomopathologie , Tumeurs osseuses/génétique , Facteur de croissance endothéliale vasculaire de type C/métabolisme , Facteur de croissance endothéliale vasculaire de type C/génétique , Animaux , Apoptose/génétique , Phosphorylation
6.
Biol Direct ; 19(1): 44, 2024 Jun 07.
Article de Anglais | MEDLINE | ID: mdl-38849910

RÉSUMÉ

BACKGROUND: The molecular mechanisms of osteosarcoma (OS) are complex. In this study, we focused on the functions of melanoma cell adhesion molecule (MCAM), methyltransferase 3 (METTL3) and insulin like growth factor 2 mRNA binding protein 1 (IGF2BP1) in OS development. METHODS: qRT-PCR assay and western blot assay were performed to determine mRNA and protein expression of MCAM, METTL3, IGF2BP1 and YY1. MTT assay and colony formation assay were conducted to assess cell proliferation. Cell apoptosis, invasion and migration were evaluated by flow cytometry analysis, transwell assay and wound-healing assay, respectively. Methylated RNA Immunoprecipitation (MeRIP), dual-luciferase reporter, Co-IP, RIP and ChIP assays were performed to analyze the relationships of MCAM, METTL3, IGF2BP1 and YY1. The functions of METTL3 and MCAM in tumor growth were explored through in vivo experiments. RESULTS: MCAM was upregulated in OS, and MCAM overexpression promoted OS cell growth, invasion and migration and inhibited apoptosis. METTL3 and IGF2BP1 were demonstrated to mediate the m6A methylation of MCAM. Functionally, METTL3 or IGF2BP1 silencing inhibited OS cell progression, while MCAM overexpression ameliorated the effects. Transcription factor YY1 promoted the transcription level of METTL3 and regulated METTL3 expression in OS cells. Additionally, METTL3 deficiency suppressed tumor growth in vivo, while MCAM overexpression abated the effect. CONCLUSION: YY1/METTL3/IGF2BP1/MCAM axis aggravated OS development, which might provide novel therapy targets for OS.


Sujet(s)
Adénosine , Methyltransferases , Ostéosarcome , Protéines de liaison à l'ARN , Ostéosarcome/génétique , Ostéosarcome/métabolisme , Methyltransferases/métabolisme , Methyltransferases/génétique , Humains , Protéines de liaison à l'ARN/génétique , Protéines de liaison à l'ARN/métabolisme , Adénosine/analogues et dérivés , Adénosine/métabolisme , Adénosine/génétique , Lignée cellulaire tumorale , Animaux , Souris , Prolifération cellulaire , Tumeurs osseuses/génétique , Tumeurs osseuses/métabolisme , Évolution de la maladie , Souris nude , Apoptose , Mouvement cellulaire , Régulation de l'expression des gènes tumoraux
7.
Biomed Pharmacother ; 176: 116924, 2024 Jul.
Article de Anglais | MEDLINE | ID: mdl-38876052

RÉSUMÉ

Noncoding RNAs (ncRNAs) do not participate in protein-coding. Ferroptosis is a newly discovered form of cell death mediated by reactive oxygen species and lipid peroxidation. Recent studies have shown that ncRNAs such as microRNAs, long noncoding RNAs, circular RNAs, and ferroptosis are involved in the occurrence and development of osteosarcoma (OS). Studies have confirmed that ncRNAs participate in the development of OS by regulating the ferroptosis. However, systematic summary on this topic are still lacking. This review summarises the potential role of ncRNAs in the diagnosis, treatment, drug resistance, and prognosis of OS and the basis for diagnosing, preventing, and treating clinical OS and developing effective drugs. This review summarises the latest research progress on ncRNAs that regulate ferroptosis in OS, attempts to clarify the molecular mechanisms by which ncRNAs regulate ferroptosis in the pathogenesis of OS, and elaborates on the involvement of ferroptosis in OS from the perspective of ncRNAs.


Sujet(s)
Tumeurs osseuses , Ferroptose , microARN , Ostéosarcome , ARN circulaire , ARN long non codant , Ferroptose/génétique , Ostéosarcome/génétique , Ostéosarcome/anatomopathologie , Humains , microARN/génétique , microARN/métabolisme , ARN long non codant/génétique , ARN long non codant/métabolisme , ARN circulaire/génétique , ARN circulaire/métabolisme , Tumeurs osseuses/génétique , Tumeurs osseuses/anatomopathologie , Tumeurs osseuses/métabolisme , Animaux , Régulation de l'expression des gènes tumoraux
8.
Sci Rep ; 14(1): 13790, 2024 06 14.
Article de Anglais | MEDLINE | ID: mdl-38877061

RÉSUMÉ

PDE1B had been found to be involved in various diseases, including tumors and non-tumors. However, little was known about the definite role of PDE1B in osteosarcoma. Therefore, we mined public data on osteosarcoma to reveal the prognostic values and immunological roles of the PDE1B gene. Three osteosarcoma-related datasets from online websites were utilized for further data analysis. R 4.3.2 software was utilized to conduct difference analysis, prognostic analysis, gene set enrichment analysis (GSEA), nomogram construction, and immunological evaluations, respectively. Experimental verification of the PDE1B gene in osteosarcoma was conducted by qRT-PCR and western blot, based on the manufacturer's instructions. The PDE1B gene was discovered to be lowly expressed in osteosarcoma, and its low expression was associated with poor OS (all P < 0.05). Experimental verifications by qRT-PCR and western blot results remained consistent (all P < 0.05). Univariate and multivariate Cox regression analyses indicated that the PDE1B gene had independent abilities in predicting OS in the TARGET osteosarcoma dataset (both P < 0.05). GSEA revealed that PDE1B was markedly linked to the calcium, cell cycle, chemokine, JAK STAT, and VEGF pathways. Moreover, PDE1B was found to be markedly associated with immunity (all P < 0.05), and the TIDE algorithm further shed light on that patients with high-PDE1B expression would have a better immune response to immunotherapies than those with low-PDE1B expression, suggesting that the PDE1B gene could prevent immune escape from osteosarcoma. The PDE1B gene was found to be a tumor suppressor gene in osteosarcoma, and its high expression was related to a better OS prognosis, suppressing immune escape from osteosarcoma.


Sujet(s)
Marqueurs biologiques tumoraux , Tumeurs osseuses , Ostéosarcome , Microenvironnement tumoral , Ostéosarcome/génétique , Ostéosarcome/mortalité , Ostéosarcome/immunologie , Ostéosarcome/anatomopathologie , Humains , Marqueurs biologiques tumoraux/génétique , Marqueurs biologiques tumoraux/métabolisme , Pronostic , Microenvironnement tumoral/immunologie , Microenvironnement tumoral/génétique , Tumeurs osseuses/génétique , Tumeurs osseuses/mortalité , Tumeurs osseuses/anatomopathologie , Tumeurs osseuses/immunologie , Mâle , Femelle , Régulation de l'expression des gènes tumoraux , Cyclic Nucleotide Phosphodiesterases, Type 1/génétique , Cyclic Nucleotide Phosphodiesterases, Type 1/métabolisme
9.
Head Neck Pathol ; 18(1): 48, 2024 Jun 17.
Article de Anglais | MEDLINE | ID: mdl-38884816

RÉSUMÉ

BACKGROUND: Craniofacial osteosarcomas (CFOS) are uncommon malignant neoplasms of the head and neck with different clinical presentation, biological behavior and prognosis from conventional osteosarcomas of long bones. Very limited genetic data have been published on CFOS. METHODS: In the current study, we performed comprehensive genomic studies in 15 cases of high-grade CFOS by SNP array and targeted next generation sequencing. RESULT: Our study shows high-grade CFOS demonstrate highly complex and heterogenous genomic alterations and harbor frequently mutated tumor suppressor genes TP53, CDKN2A/B, and PTEN, similar to conventional osteosarcomas. Potentially actionable gene amplifications involving CCNE1, AKT2, MET, NTRK1, PDGFRA, KDR, KIT, MAP3K14, FGFR1, and AURKA were seen in 43% of cases. GNAS hotspot activating mutations were also identified in a subset of CFOS cases, with one case representing malignant transformation from fibrous dysplasia, suggesting a role for GNAS mutation in the development of CFOS. CONCLUSION: High-grade CFOS demonstrate highly complex and heterogenous genomic alterations, with amplification involving receptor tyrosine kinase genes, and frequent mutations involving tumor suppressor genes.


Sujet(s)
Variations de nombre de copies de segment d'ADN , Séquençage nucléotidique à haut débit , Ostéosarcome , Humains , Femelle , Mâle , Adulte , Ostéosarcome/génétique , Ostéosarcome/anatomopathologie , Adulte d'âge moyen , Adolescent , Mutation , Enfant , Jeune adulte , Sujet âgé , Tumeurs osseuses/génétique , Tumeurs osseuses/anatomopathologie , Tumeurs du crâne/génétique , Tumeurs du crâne/anatomopathologie , Analyse de mutations d'ADN
10.
Sci Rep ; 14(1): 12934, 2024 06 05.
Article de Anglais | MEDLINE | ID: mdl-38839983

RÉSUMÉ

Osteosarcoma is a primary malignant tumor that commonly affects children and adolescents, with a poor prognosis. The existence of tumor heterogeneity leads to different molecular subtypes and survival outcomes. Recently, lipid metabolism has been identified as a critical characteristic of cancer. Therefore, our study aims to identify osteosarcoma's lipid metabolism molecular subtype and develop a signature for survival outcome prediction. Four multicenter cohorts-TARGET-OS, GSE21257, GSE39058, and GSE16091-were amalgamated into a unified Meta-Cohort. Through consensus clustering, novel molecular subtypes within Meta-Cohort patients were delineated. Subsequent feature selection processes, encompassing analyses of differentially expressed genes between subtypes, univariate Cox analysis, and StepAIC, were employed to pinpoint biomarkers related to lipid metabolism in TARGET-OS. We selected the most effective algorithm for constructing a Lipid Metabolism-Related Signature (LMRS) by utilizing four machine-learning algorithms reconfigured into ten unique combinations. This selection was based on achieving the highest concordance index (C-index) in the test cohort of GSE21257, GSE39058, and GSE16091. We identified two distinct lipid metabolism molecular subtypes in osteosarcoma patients, C1 and C2, with significantly different survival rates. C1 is characterized by increased cholesterol, fatty acid synthesis, and ketone metabolism. In contrast, C2 focuses on steroid hormone biosynthesis, arachidonic acid, and glycerolipid and linoleic acid metabolism. Feature selection in the TARGET-OS identified 12 lipid metabolism genes, leading to a model predicting osteosarcoma patient survival. The LMRS, based on the 12 identified genes, consistently accurately predicted prognosis across TARGET-OS, testing cohorts, and Meta-Cohort. Incorporating 12 published signatures, LMRS showed robust and significantly superior predictive capability. Our results offer a promising tool to enhance the clinical management of osteosarcoma, potentially leading to improved clinical outcomes.


Sujet(s)
Tumeurs osseuses , Métabolisme lipidique , Apprentissage machine , Ostéosarcome , Ostéosarcome/génétique , Ostéosarcome/mortalité , Ostéosarcome/métabolisme , Ostéosarcome/anatomopathologie , Humains , Métabolisme lipidique/génétique , Pronostic , Tumeurs osseuses/génétique , Tumeurs osseuses/mortalité , Tumeurs osseuses/métabolisme , Tumeurs osseuses/anatomopathologie , Marqueurs biologiques tumoraux/génétique , Marqueurs biologiques tumoraux/métabolisme , Femelle , Mâle , Régulation de l'expression des gènes tumoraux , Adolescent , Analyse de profil d'expression de gènes/méthodes , Enfant
11.
Medicine (Baltimore) ; 103(23): e38470, 2024 Jun 07.
Article de Anglais | MEDLINE | ID: mdl-38847690

RÉSUMÉ

Osteosarcoma (OS) is the most common primary malignant bone tumor occurring in children and adolescents. Improvements in our understanding of the OS pathogenesis and metastatic mechanism on the molecular level might lead to notable advances in the treatment and prognosis of OS. Biomarkers related to OS metastasis and prognosis were analyzed and identified, and a prognostic model was established through the integration of bioinformatics tools and datasets in multiple databases. 2 OS datasets were downloaded from the Gene Expression Omnibus database for data consolidation, standardization, batch effect correction, and identification of differentially expressed genes (DEGs); following that, gene ontology and Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway enrichment analyses were performed on the DEGs; the STRING database was subsequently used for protein-protein interaction (PPI) network construction and identification of hub genes; hub gene expression was validated, and survival analysis was conducted through the employment of the TARGET database; finally, a prognostic model was established and evaluated subsequent to the screening of survival-related genes. A total of 701 DEGs were identified; by gene ontology and KEGG pathway enrichment analyses, the overlapping DEGs were enriched for 249 biological process terms, 13 cellular component terms, 35 molecular function terms, and 4 KEGG pathways; 13 hub genes were selected from the PPI network; 6 survival-related genes were identified by the survival analysis; the prognostic model suggested that 4 genes were strongly associated with the prognosis of OS. DEGs related to OS metastasis and survival were identified through bioinformatics analysis, and hub genes were further selected to establish an ideal prognostic model for OS patients. On this basis, 4 protective genes including TPM1, TPM2, TPM3, and TPM4 were yielded by the prognostic model.


Sujet(s)
Tumeurs osseuses , Biologie informatique , Ostéosarcome , Cartes d'interactions protéiques , Ostéosarcome/génétique , Ostéosarcome/mortalité , Ostéosarcome/anatomopathologie , Humains , Biologie informatique/méthodes , Pronostic , Tumeurs osseuses/génétique , Tumeurs osseuses/mortalité , Tumeurs osseuses/anatomopathologie , Cartes d'interactions protéiques/génétique , Marqueurs biologiques tumoraux/génétique , Régulation de l'expression des gènes tumoraux , Analyse de profil d'expression de gènes/méthodes , Gene Ontology , Bases de données génétiques , Analyse de survie , Métastase tumorale/génétique
12.
J Biomed Sci ; 31(1): 59, 2024 Jun 05.
Article de Anglais | MEDLINE | ID: mdl-38835012

RÉSUMÉ

Osteosarcoma (OS) is the most prevalent and fatal type of bone tumor. It is characterized by great heterogeneity of genomic aberrations, mutated genes, and cell types contribution, making therapy and patients management particularly challenging. A unifying picture of molecular mechanisms underlying the disease could help to transform those challenges into opportunities.This review deeply explores the occurrence in OS of large-scale RNA regulatory networks, denominated "competing endogenous RNA network" (ceRNET), wherein different RNA biotypes, such as long non-coding RNAs, circular RNAs and mRNAs can functionally interact each other by competitively binding to shared microRNAs. Here, we discuss how the unbalancing of any network component can derail the entire circuit, driving OS onset and progression by impacting on cell proliferation, migration, invasion, tumor growth and metastasis, and even chemotherapeutic resistance, as distilled from many studies. Intriguingly, the aberrant expression of the networks components in OS cells can be triggered also by the surroundings, through cytokines and vesicles, with their bioactive cargo of proteins and non-coding RNAs, highlighting the relevance of tumor microenvironment. A comprehensive picture of RNA regulatory networks underlying OS could pave the way for the development of innovative RNA-targeted and RNA-based therapies and new diagnostic tools, also in the perspective of precision oncology.


Sujet(s)
Ostéosarcome , Humains , Ostéosarcome/génétique , Ostéosarcome/thérapie , ARN long non codant/génétique , ARN long non codant/métabolisme , Tumeurs osseuses/génétique , Tumeurs osseuses/thérapie , Tumeurs osseuses/métabolisme , Tumeurs osseuses/anatomopathologie , Réseaux de régulation génique , ARN circulaire/génétique , microARN/génétique , microARN/métabolisme , Régulation de l'expression des gènes tumoraux
13.
Diagn Pathol ; 19(1): 79, 2024 Jun 11.
Article de Anglais | MEDLINE | ID: mdl-38863002

RÉSUMÉ

BACKGROUND: Osteosarcoma is a bone tumor that is characterized by high malignancy and a high mortality rate, and that originates from primitive osteoblastic mesenchymal cells and is most common in rapidly growing long bones. PSMD14, also known as RPN11 or POH1, is a member of the JAMM isopeptidase family, which is able to remove the substrate protein ubiquitination label, thereby regulating the stability and function of the substrate protein. In this study, we explored the expression and potential biological significance of the PSMD14 deubiquitinating enzyme in osteosarcoma. METHODS: Immunohistochemical methods were used to detect the expression of PSMD14 in biopsies of 91 osteosarcoma patients, and the specimens were classified into high and low PSMD14 expression groups. The correlation between PSMD14 expression and clinical indicators and prognosis was compared.SiRNA was used to downregulate PSMD14 in two osteosarcoma cell lines (HOS and SJSA-1), and the effects of downregulation of PSMD14 on the viability, proliferation, and invasion ability of osteosarcoma cells were analyzed. RESULTS: We identified significant differences in recurrence, metastasis, and survival time of the osteosarcoma patients on the basis of PSMD14 expression. High expression of PSMD14 in osteosarcoma patients was associated with a low survival rate and high risk of metastasis and recurrence. Down-regulation of PSMD14 inhibited the viability, proliferation, and invasiveness of osteosarcoma cell lines. CONCLUSIONS: PSMD14 may be a new prognostic marker and therapeutic target for osteosarcoma.


Sujet(s)
Marqueurs biologiques tumoraux , Tumeurs osseuses , Ostéosarcome , Ostéosarcome/anatomopathologie , Ostéosarcome/mortalité , Ostéosarcome/métabolisme , Ostéosarcome/génétique , Humains , Tumeurs osseuses/anatomopathologie , Tumeurs osseuses/mortalité , Tumeurs osseuses/métabolisme , Tumeurs osseuses/génétique , Mâle , Marqueurs biologiques tumoraux/métabolisme , Marqueurs biologiques tumoraux/analyse , Femelle , Pronostic , Lignée cellulaire tumorale , Adulte , Prolifération cellulaire , Proteasome endopeptidase complex/métabolisme , Adolescent , Jeune adulte , Adulte d'âge moyen , Invasion tumorale , Transactivateurs
14.
Sci Rep ; 14(1): 13401, 2024 06 11.
Article de Anglais | MEDLINE | ID: mdl-38862526

RÉSUMÉ

This study aimed to determine an immune-related RNA signature as a prognostic marker, in this study, we developed a risk score model for predicting the prognosis of osteosarcoma metastasis. We first downloaded the clinical information and expression data of osteosarcoma samples from the UCSC Xena and GEO databases, of which the former was the training set and the latter was the validation set. Immune infiltration was assessed using the ssGSEA and ESTIMATE algorithms, and the osteosarcoma samples were divided into the Immunity_L and Immunity_H groups. Then, eleven RNAs were identified as the optimal prognostic RNA signatures using LASSO Cox regression analysis for establishing a risk score (RS) model. Kaplan-Meier approach indicated the high-risk group exhibited a shorter survival. Furthermore, we analyzed the tumor metastasis, age, and RS model status were determined to be independent clinical prognostic factors using Cox regression analysis. Decision curve analysis (DCA) indicated that the prognostic factor + RS model had the best net benefit. Finally, nine tumor-infiltrating immune cells (TIICs) showed significant differences in abundance between high- and low-risk groups via CIBERSORT deconvolution algorithm. In conclusion, the immune-related eleven-RNA signature be could served as a potential prognostic biomarker for osteosarcoma metastasis.


Sujet(s)
Marqueurs biologiques tumoraux , Tumeurs osseuses , Ostéosarcome , Ostéosarcome/génétique , Ostéosarcome/immunologie , Ostéosarcome/mortalité , Ostéosarcome/anatomopathologie , Humains , Pronostic , Tumeurs osseuses/génétique , Tumeurs osseuses/mortalité , Tumeurs osseuses/immunologie , Tumeurs osseuses/anatomopathologie , Marqueurs biologiques tumoraux/génétique , Femelle , Mâle , Régulation de l'expression des gènes tumoraux , Métastase tumorale , Estimation de Kaplan-Meier , Analyse de profil d'expression de gènes , Transcriptome , Modèles des risques proportionnels , Facteurs de risque , Algorithmes
15.
BMC Musculoskelet Disord ; 25(1): 437, 2024 Jun 04.
Article de Anglais | MEDLINE | ID: mdl-38835052

RÉSUMÉ

BACKGROUND: Osteosarcoma (OS) is the most common bone malignant tumor in children, and its prognosis is often poor. Anoikis is a unique mode of cell death.However, the effects of Anoikis in OS remain unexplored. METHOD: Differential analysis of Anoikis-related genes was performed based on the metastatic and non-metastatic groups. Then LASSO logistic regression and SVM-RFE algorithms were applied to screen out the characteristic genes. Later, Univariate and multivariate Cox regression was conducted to identify prognostic genes and further develop the Anoikis-based risk score. In addition, correlation analysis was performed to analyze the relationship between tumor microenvironment, drug sensitivity, and prognostic models. RESULTS: We established novel Anoikis-related subgroups and developed a prognostic model based on three Anoikis-related genes (MAPK1, MYC, and EDIL3). The survival and ROC analysis results showed that the prognostic model was reliable. Besides, the results of single-cell sequencing analysis suggested that the three prognostic genes were closely related to immune cell infiltration. Subsequently, aberrant expression of two prognostic genes was identified in osteosarcoma cells. Nilotinib can promote the apoptosis of osteosarcoma cells and down-regulate the expression of MAPK1. CONCLUSIONS: We developed a novel Anoikis-related risk score model, which can assist clinicians in evaluating the prognosis of osteosarcoma patients in clinical practice. Analysis of the tumor immune microenvironment and chemotherapeutic drug sensitivity can provide necessary insights into subsequent mechanisms. MAPK1 may be a valuable therapeutic target for neoadjuvant chemotherapy in osteosarcoma.


Sujet(s)
Anoïkis , Tumeurs osseuses , Mitogen-Activated Protein Kinase 1 , Traitement néoadjuvant , Ostéosarcome , Microenvironnement tumoral , Ostéosarcome/traitement médicamenteux , Ostéosarcome/génétique , Humains , Anoïkis/effets des médicaments et des substances chimiques , Anoïkis/génétique , Tumeurs osseuses/génétique , Tumeurs osseuses/traitement médicamenteux , Mitogen-Activated Protein Kinase 1/métabolisme , Mitogen-Activated Protein Kinase 1/génétique , Microenvironnement tumoral/effets des médicaments et des substances chimiques , Pronostic , Mâle , Femelle , Lignée cellulaire tumorale , Régulation de l'expression des gènes tumoraux , Enfant , Adolescent
16.
J Orthop Surg Res ; 19(1): 356, 2024 Jun 15.
Article de Anglais | MEDLINE | ID: mdl-38879525

RÉSUMÉ

BACKGROUND: Ubiquitin/ubiquitin-like (Ub/UBL)-related genes have been reported to be associated with the survival of osteosarcoma patients but have not yet been systematically explored. METHODS: The prognostic value of Ub/UBL-related genes, immune cell infiltration and clinicopathological features of patients were explored by Cox and LASSO regression analyses. A prognostic model was established and then validated in the GSE21257 dataset. The differential expression of hub genes in osteosarcoma was confirmed by qRT-PCR, western blotting and immunohistochemistry. RESULTS: Tripartite Motif Containing 8 (TRIM8) and Ubiquitin Like With PHD And Ring Finger Domains 2 (UHRF2) were screened as genes with prognostic value in osteosarcoma. Kaplan-Meier analysis and scatter plots indicated that patients in the high gene significance score group tended to have a worse prognosis. The concordance index, calibration analysis and receiver operating characteristic analysis suggested that the model had good prediction accuracy and high sensitivity and specificity. Decision curve analysis revealed that patients could obtain greater net benefit from this model. Functional analyses of the differentially expressed genes indicated that they were involved in important functions and pathways. TRIM8 and UHRF2 were confirmed to be highly expressed in osteosarcoma cell lines and tissues. CONCLUSIONS: TRIM8 and UHRF2 are potential prognostic genes in osteosarcoma, and these results provide insights into the roles of these genes and their implications for patient outcomes.


Sujet(s)
Tumeurs osseuses , Ostéosarcome , Ostéosarcome/génétique , Ostéosarcome/anatomopathologie , Ostéosarcome/immunologie , Ostéosarcome/mortalité , Humains , Pronostic , Tumeurs osseuses/génétique , Tumeurs osseuses/anatomopathologie , Tumeurs osseuses/immunologie , Tumeurs osseuses/mortalité , Mâle , Femelle , Marqueurs biologiques tumoraux/génétique , Ubiquitin-protein ligases/génétique , Ubiquitine/génétique
17.
Anticancer Res ; 44(7): 2787-2792, 2024 Jul.
Article de Anglais | MEDLINE | ID: mdl-38925854

RÉSUMÉ

BACKGROUND/AIM: Methotrexate (MTX) resistance in osteosarcoma leads to a very poor prognosis. In the present study, in order to further understand the basis and ramifications of MTX resistance in osteosarcoma, we selected an osteosarcoma cell line that has a 5,500-fold-increased MTX IC50 Materials and Methods: The super MTX-resistant 143B osteosarcoma cells (143B-MTXSR) were selected from MTX-sensitive parental human 143B osteosarcoma cells (143B-P) by continuous culture with step-wise increased amounts of MTX. To compare the malignancy of 143B-MTXSR and 143B-P, colony-formation capacity was compared with clonogenic assays on plastic and in soft agar. In addition, tumor growth was compared with orthotopic xenograft mouse models of osteosarcoma. Expression of dihydrofolate reductase (DHFR), phosphoinositide 3-kinase (PI3K)/protein kinase B (AKT)/mammalian target of rapamycin (mTOR), and myelocytomatosis oncogene (MYC) was examined with western immunoblotting and compared in 143B-MTXSR and 143B-P cells. RESULTS: 143B-MTXSR had a 5,500-fold increase in the MTX IC50 compared to the parental 143B-P cells. Expression of DHFR was increased 10-fold in 143B-MTXSR compared to 143B-P (p<0.01). 143B-MTXSR cells had reduced colony-formation capacity on plastic (p=0.032) and in soft agar (p<0.01) compared to 143B-P and reduced tumor growth in orthotopic xenograft mouse models (p<0.001). These results demonstrate that 143B-MTXSR had reduced malignancy. 143B-MTXSR also showed an increased expression of PI3K (p<0.01), phosphorylated (activated) AKT (p=0.031), phosphorylated mTOR (p=0.043), and c-MYC (p=0.024) compared to 143B-P. CONCLUSION: The present study demonstrates that the increased expression of DHFR, PI3K/AKT/mTOR and c-MYC appears to be linked to super MTX resistance and, paradoxically, to reduced malignancy. The present results suggest that DHFR may be a powerful tumor suppressor when highly amplified.


Sujet(s)
Résistance aux médicaments antinéoplasiques , Méthotrexate , Ostéosarcome , Phosphatidylinositol 3-kinases , Protéines proto-oncogènes c-akt , Protéines proto-oncogènes c-myc , Sérine-thréonine kinases TOR , Dihydrofolate reductase , Ostéosarcome/traitement médicamenteux , Ostéosarcome/anatomopathologie , Ostéosarcome/métabolisme , Ostéosarcome/génétique , Méthotrexate/pharmacologie , Humains , Dihydrofolate reductase/métabolisme , Dihydrofolate reductase/génétique , Animaux , Résistance aux médicaments antinéoplasiques/effets des médicaments et des substances chimiques , Sérine-thréonine kinases TOR/métabolisme , Protéines proto-oncogènes c-akt/métabolisme , Lignée cellulaire tumorale , Phosphatidylinositol 3-kinases/métabolisme , Protéines proto-oncogènes c-myc/métabolisme , Protéines proto-oncogènes c-myc/génétique , Souris , Tests d'activité antitumorale sur modèle de xénogreffe , Tumeurs osseuses/traitement médicamenteux , Tumeurs osseuses/anatomopathologie , Tumeurs osseuses/métabolisme , Tumeurs osseuses/génétique , Amplification de gène , Transduction du signal/effets des médicaments et des substances chimiques , Souris nude , Antimétabolites antinéoplasiques/pharmacologie
18.
Front Biosci (Landmark Ed) ; 29(6): 207, 2024 May 30.
Article de Anglais | MEDLINE | ID: mdl-38940027

RÉSUMÉ

BACKGROUND: Osteosarcoma (OS) is a primary malignant bone tumor in the pediatric and adolescent populations. Long non-coding RNAs (LncRNAs), such as plasma-cytoma variant translocation 1 (PVT1), have emerged as significant regulators of OS metastasis. Recent studies have indicated that activation of signal transducer and activator of transcription 3 (STAT3) signaling, which might be controlled by PVT1, inhibits ferroptosis to promote the malignant progression of cancer. Therefore, the present study aimed to determine the role of PVT1 in OS pathogenesis and investigate whether PVT1 affects OS progression by regulating STAT3/GPX4 pathway-mediated ferroptosis. METHODS: The human OS cell line MG63 were transfected with sh-PVT1 plasmid to inhibit PVT1 expression, with or without co-transfection with a STAT3 overexpression plasmid. The expression of PVT1 was determined by real-time quantitative polymerase chain reaction (RT-qPCR). The proliferation, migration, invasion, and apoptosis of MG63 cells were determined using the cell counting kit-8 (CCK8), Transwell assay, and flow cytometry. The levels of malondialdehyde (MDA), Fe2+, and glutathione (GSH) were determined by ELISA kits, whereas reactive oxygen species (ROS) level was determined by immunofluorescence. The protein expression levels of STAT3, p-STAT3, and glutathione peroxidase 4 (GPX4) were detected by western blot (WB). RESULTS: PVT1 expression was significantly increased in MG63 cells. When knocking down PVT1 with sh-PVT1 plasmid, the proliferation, migration, and invasion of MG63 cells were markedly inhibited, while the rate of apoptosis was upregulated. Further investigation revealed that MG63 cells with PVT1 knockdown exhibited elevated levels of MDA, Fe2+, and ROS. In addition, the inhibition of PVT1 expression resulted in decreased levels of GSH and inhibited expression of p-STAT3 and GPX4. When sh-PVT1 was co-transfected with STAT3 overexpression plasmid in MG63 cells, the increased levels of MDA, Fe2+, and ROS were downregulated, and the decreased expressions of GSH, p-STAT3, and GPX4 were upregulated. CONCLUSION: PVT1 promotes OS metastasis by activating the STAT3/GPX4 pathway to inhibit ferroptosis. Targeting PVT1 might be a novel therapeutic strategy for OS treatment.


Sujet(s)
Tumeurs osseuses , Ferroptose , Ostéosarcome , Phospholipid hydroperoxide glutathione peroxidase , ARN long non codant , Facteur de transcription STAT-3 , Humains , Ostéosarcome/génétique , Ostéosarcome/métabolisme , Ostéosarcome/anatomopathologie , ARN long non codant/génétique , ARN long non codant/métabolisme , Ferroptose/génétique , Facteur de transcription STAT-3/métabolisme , Facteur de transcription STAT-3/génétique , Lignée cellulaire tumorale , Tumeurs osseuses/génétique , Tumeurs osseuses/métabolisme , Tumeurs osseuses/anatomopathologie , Phospholipid hydroperoxide glutathione peroxidase/métabolisme , Phospholipid hydroperoxide glutathione peroxidase/génétique , Prolifération cellulaire/génétique , Espèces réactives de l'oxygène/métabolisme , Transduction du signal , Mouvement cellulaire/génétique , Évolution de la maladie , Apoptose/génétique , Régulation de l'expression des gènes tumoraux
19.
Cancer Biol Ther ; 25(1): 2343450, 2024 Dec 31.
Article de Anglais | MEDLINE | ID: mdl-38742566

RÉSUMÉ

The potential function and mechanism of circRNAs in regulating malignant performances of Osteosarcoma (OS) cells have not been well investigated. The expression level of CircLMO7, miR-21-5p and ARHGAP24 were detected by RT-qPCR. The relationship between miR-21-5p and circ-LMO7, as well as between miR-21-5p and ARHGAP24, was predicted and examined through bioinformatics analysis and luciferase reporter gene experiments. Moreover, OS cell growth, invasion, migration, and apoptosis were detected using the cell counting kit-8 (CCK-8), transwell and flow cytometry assays, respectively. ARHGAP24 protein level was measured using western blotting. In present study, we choose to investigate the role and mechanism of circ-LOM7 on OS cell proliferation, migration and invasion. circ-LOM7 was found to be down-regulated in OS tissues and cell lines. Enforced expression of circ-LOM7 suppressed the growth, invasion, and migration of OS cells. In contrast, decreasing circ-LMO7 expression had opposite effects. Furthermore, miR-21-5p was predicted to be sponged by circ-LMO7, and had an opposite role of circ-LMO7 in OS. Moreover, ARHGAP24 served as miR-21-5p's downstream target. Mechanistically, circ-LMO7 was packed in exosomes and acted as a cancer-suppresser on OS by sponging miR-21-5p and upregulating the expression of ARHGAP24. The exosomal circ-LMO7 expression was significantly decreased in OS cell exosomes, and co-culture experiments showed that exosomal circ-LMO7 suppressed the proliferation ability of OS cells. Circ-LMO7 exerts as a tumor suppressor in OS, and the circ-LMO7/miR-21-5P/ARHGAP24 axis is involved in OS progression.


Sujet(s)
Évolution de la maladie , Exosomes , Protéines d'activation de la GTPase , microARN , Ostéosarcome , ARN circulaire , Ostéosarcome/génétique , Ostéosarcome/anatomopathologie , Ostéosarcome/métabolisme , Humains , microARN/génétique , microARN/métabolisme , ARN circulaire/génétique , ARN circulaire/métabolisme , Exosomes/métabolisme , Exosomes/génétique , Protéines d'activation de la GTPase/génétique , Protéines d'activation de la GTPase/métabolisme , Prolifération cellulaire , Souris , Animaux , Lignée cellulaire tumorale , Mouvement cellulaire/génétique , Apoptose/génétique , Tumeurs osseuses/génétique , Tumeurs osseuses/anatomopathologie , Tumeurs osseuses/métabolisme , Régulation de l'expression des gènes tumoraux , Mâle , Femelle
20.
Tissue Cell ; 88: 102409, 2024 Jun.
Article de Anglais | MEDLINE | ID: mdl-38781792

RÉSUMÉ

BACKGROUND: Osteosarcoma is originated from skeletal system. Recombinant human proteoglycan 4 (rhPRG4) can inhibit cell proliferation and migration in multiple cancers. This research is designed to dig out the role and mechanism of PRG4 in osteosarcoma. METHODS: Human osteosarcoma cell lines, MG63 and 143B, were transfected with programmed death 1 (PD-L1) overexpression vectors and/or treated with 20, 50, and 100 µg/mL rhPRG4, followed by the determination of cell viability, colony formation, sphere formation, invasion, migration, apoptosis, and the expressions of matrix metalloproteinases (MMPs), PD-L1 and apoptosis-related proteins. Tumor-bearing mouse models were constructed by injection of 143B cells and treatment of anti-PD-L1 antibody and/or adenovirus PRG4 (AdPRG4). Tumor volume was monitored, and immunohistochemical location of Ki67 was performed. Expressions of MMPs, transforming growth factor-ß (TGF-ß), PD-L1, and epithelial mesenchymal transition (EMT)-related proteins were measured in tumors. RESULTS: RhPRG4 (20, 50, and 100 µg/mL) inhibited the viability, colony formation, sphere formation, invasion, migration, and the expressions of MMP2, MMP9 and Bcl2 in osteosarcoma cells, while promoting cell apoptosis as well as Bax and c-caspase3 expressions, at a dose-dependent manner; by contrast, PD-L1 overexpression reversed the above effects of 100 µg/mL rhPRG4. AdPRG4 or anti-PD-L1 antibody decreased tumor volume, number of pulmonary metastasis nodule, Ki67 location, and expressions of TGF-ß, PD-L1, MMP2, MMP9, Vimentin, and Snail, but increased E-cadherin expression in tumor cells. Moreover, anti-PD-L1 antibody and AdPRG4 together functioned more effectively than them alone in reducing tumor burden. CONCLUSION: PRG4 represses the genesis and metastasis of osteosarcoma via inhibiting PD-L1 expression, and AdPRG4 enhances the effectiveness of anti-PD-L1 therapy.


Sujet(s)
Antigène CD274 , Ostéosarcome , Protéoglycanes , Animaux , Humains , Souris , Apoptose/effets des médicaments et des substances chimiques , Antigène CD274/métabolisme , Antigène CD274/génétique , Tumeurs osseuses/anatomopathologie , Tumeurs osseuses/métabolisme , Tumeurs osseuses/génétique , Lignée cellulaire tumorale , Mouvement cellulaire/effets des médicaments et des substances chimiques , Prolifération cellulaire/effets des médicaments et des substances chimiques , Transition épithélio-mésenchymateuse/effets des médicaments et des substances chimiques , Transition épithélio-mésenchymateuse/génétique , Régulation de l'expression des gènes tumoraux/effets des médicaments et des substances chimiques , Souris nude , Métastase tumorale , Ostéosarcome/anatomopathologie , Ostéosarcome/métabolisme , Ostéosarcome/génétique , Protéoglycanes/métabolisme
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE
...