Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 726
Filter
1.
Sci Rep ; 14(1): 13790, 2024 06 14.
Article in English | MEDLINE | ID: mdl-38877061

ABSTRACT

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.


Subject(s)
Biomarkers, Tumor , Bone Neoplasms , Osteosarcoma , Tumor Microenvironment , Osteosarcoma/genetics , Osteosarcoma/mortality , Osteosarcoma/immunology , Osteosarcoma/pathology , Humans , Biomarkers, Tumor/genetics , Biomarkers, Tumor/metabolism , Prognosis , Tumor Microenvironment/immunology , Tumor Microenvironment/genetics , Bone Neoplasms/genetics , Bone Neoplasms/mortality , Bone Neoplasms/pathology , Bone Neoplasms/immunology , Male , Female , Gene Expression Regulation, Neoplastic , Cyclic Nucleotide Phosphodiesterases, Type 1/genetics , Cyclic Nucleotide Phosphodiesterases, Type 1/metabolism
2.
BMC Cancer ; 24(1): 703, 2024 Jun 07.
Article in English | MEDLINE | ID: mdl-38849717

ABSTRACT

Immunodeficient murine models are usually used as the preclinical models of osteosarcoma. Such models do not effectively simulate the process of tumorigenesis and metastasis. Establishing a suitable animal model for understanding the mechanism of osteosarcoma and the clinical translation is indispensable. The UMR-106 cell suspension was injected into the marrow cavity of Balb/C nude mice. Tumor masses were harvested from nude mice and sectioned. The tumor fragments were transplanted into the marrow cavities of SD rats immunosuppressed with cyclosporine A. Through muti-rounds selection in SD rats, we constructed orthotopic osteosarcoma animal models using rats with intact immune systems. The primary tumor cells were cultured in-vitro to obtain the immune-tolerant cell line. VX2 tumor fragments were transplanted into the distal femur and parosteal radius of New Zealand white rabbit to construct orthotopic osteosarcoma animal models in rabbits. The rate of tumor formation in SD rats (P1 generation) was 30%. After four rounds of selection and six rounds of acclimatization in SD rats with intact immune systems, we obtained immune-tolerant cell lines and established the orthotopic osteosarcoma model of the distal femur in SD rats. Micro-CT images confirmed tumor-driven osteolysis and the bone destruction process. Moreover, the orthotopic model was also established in New Zealand white rabbits by implanting VX2 tumor fragments into rabbit radii and femurs. We constructed orthotopic osteosarcoma animal models in rats with intact immune systems through muti-rounds in-vivo selection and the rabbit osteosarcoma model.


Subject(s)
Bone Neoplasms , Disease Models, Animal , Osteosarcoma , Animals , Osteosarcoma/pathology , Osteosarcoma/immunology , Rabbits , Rats , Bone Neoplasms/pathology , Bone Neoplasms/immunology , Cell Line, Tumor , Mice , Mice, Nude , Rats, Sprague-Dawley , X-Ray Microtomography , Mice, Inbred BALB C , Immunocompetence , Humans , Neoplasm Transplantation , Femur/pathology , Femur/diagnostic imaging , Male
3.
Sci Rep ; 14(1): 13401, 2024 06 11.
Article in English | MEDLINE | ID: mdl-38862526

ABSTRACT

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.


Subject(s)
Biomarkers, Tumor , Bone Neoplasms , Osteosarcoma , Osteosarcoma/genetics , Osteosarcoma/immunology , Osteosarcoma/mortality , Osteosarcoma/pathology , Humans , Prognosis , Bone Neoplasms/genetics , Bone Neoplasms/mortality , Bone Neoplasms/immunology , Bone Neoplasms/pathology , Biomarkers, Tumor/genetics , Female , Male , Gene Expression Regulation, Neoplastic , Neoplasm Metastasis , Kaplan-Meier Estimate , Gene Expression Profiling , Transcriptome , Proportional Hazards Models , Risk Factors , Algorithms
4.
J Orthop Surg Res ; 19(1): 356, 2024 Jun 15.
Article in English | MEDLINE | ID: mdl-38879525

ABSTRACT

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.


Subject(s)
Bone Neoplasms , Osteosarcoma , Osteosarcoma/genetics , Osteosarcoma/pathology , Osteosarcoma/immunology , Osteosarcoma/mortality , Humans , Prognosis , Bone Neoplasms/genetics , Bone Neoplasms/pathology , Bone Neoplasms/immunology , Bone Neoplasms/mortality , Male , Female , Biomarkers, Tumor/genetics , Ubiquitin-Protein Ligases/genetics , Ubiquitin/genetics
5.
Virchows Arch ; 484(6): 925-937, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38748263

ABSTRACT

High-grade osteosarcoma, a primary malignant bone tumour, is experiencing a global increase in reported incidence with varied prevalence. Despite advances in management, which include surgery and neoadjuvant chemotherapy often an unsatisfactory outcome is found due to poor or heterogeneous response to chemotherapy. Our study delved into chemotherapy responses in osteosarcoma patients and associated molecular expressions, focusing on CD95 receptor (CD95R), interferon (IFN)-γ, catalase, heat-shock protein (Hsp)70, and vascular endothelial growth factor (VEGF). Employing immunohistochemistry and Huvos grading of post-chemo specimens, we analysed formalin-fixed paraffin-embedded (FFPE) osteosarcoma tissue of resected post-chemotherapy specimens from Dr. Soetomo General Academic Hospital in Surabaya, Indonesia (DSGAH), spanning from 2016 to 2020. Results revealed varied responses (poor 40.38%, moderate 48.08%, good 11.54%) and distinct patterns in CD95R, IFN-γ, catalase, Hsp70, and VEGF expression. Significant differences among response groups were observed in CD95R and IFN-γ expression in tumour-infiltrating lymphocytes. The trend of diminishing CD95R expression from poor to good responses, accompanied by an increase in IFN-γ, implied a reduction in the count of viable osteosarcoma cells with the progression of Huvos grading. Catalase expression in osteosarcoma cells was consistently elevated in the poor response group, while Hsp70 expression was highest. VEGF expression in macrophages was significantly higher in the good response group. In conclusion, this study enhances our understanding of immune-chemotherapy interactions in osteosarcoma and identifies potential biomarkers for targeted interventions.


Subject(s)
Bone Neoplasms , Catalase , HSP70 Heat-Shock Proteins , Interferon-gamma , Osteosarcoma , Vascular Endothelial Growth Factor A , fas Receptor , Osteosarcoma/pathology , Osteosarcoma/drug therapy , Osteosarcoma/metabolism , Osteosarcoma/immunology , Humans , Vascular Endothelial Growth Factor A/metabolism , Female , Bone Neoplasms/pathology , Bone Neoplasms/drug therapy , Bone Neoplasms/metabolism , Bone Neoplasms/immunology , Male , HSP70 Heat-Shock Proteins/metabolism , Catalase/metabolism , Young Adult , Adult , fas Receptor/metabolism , fas Receptor/analysis , Adolescent , Biomarkers, Tumor/metabolism , Biomarkers, Tumor/analysis , Neoplasm Grading , Child , Treatment Outcome , Immunohistochemistry , Lymphocytes, Tumor-Infiltrating/immunology , Lymphocytes, Tumor-Infiltrating/drug effects , Middle Aged
6.
Int J Mol Sci ; 25(10)2024 May 14.
Article in English | MEDLINE | ID: mdl-38791381

ABSTRACT

Osteosarcoma is a type of bone cancer that primarily affects children and young adults. The overall 5-year survival rate for localized osteosarcoma is 70-75%, but it is only 20-30% for patients with relapsed or metastatic tumors. To investigate potential glycan-targeting structures for immunotherapy, we stained primary osteosarcomas with recombinant C-type lectin CD301 (MGL, CLEC10A) and observed moderate to strong staining on 26% of the tumors. NK92 cells expressing a CD301-CAR recognized and eliminated osteosarcoma cells in vitro. Cytotoxic activity assays correlated with degranulation and cytokine release assays. Combination with an inhibitory antibody against the immune checkpoint TIGIT (T-cell immunoreceptor with lg and ITIM domains) showed promising additional effects. Overall, this study showed, for the first time, the expression of CD301 ligands in osteosarcoma tissue and demonstrated their use as potential target structures for lectin-based immunotherapy.


Subject(s)
Bone Neoplasms , Immunotherapy , Lectins, C-Type , Osteosarcoma , Polysaccharides , Receptors, Chimeric Antigen , Osteosarcoma/therapy , Osteosarcoma/immunology , Osteosarcoma/metabolism , Osteosarcoma/pathology , Humans , Bone Neoplasms/immunology , Bone Neoplasms/therapy , Bone Neoplasms/metabolism , Bone Neoplasms/pathology , Immunotherapy/methods , Lectins, C-Type/metabolism , Polysaccharides/metabolism , Polysaccharides/chemistry , Receptors, Chimeric Antigen/metabolism , Receptors, Chimeric Antigen/immunology , Cell Line, Tumor , Female , Male , Child , Adolescent , Receptors, Immunologic/metabolism
7.
J Cell Mol Med ; 28(10): e18395, 2024 May.
Article in English | MEDLINE | ID: mdl-38774995

ABSTRACT

Tumour-associated macrophages (TAMs), encompassing M1 and M2 subtypes, exert significant effects on osteosarcoma (OS) progression and immunosuppression. However, the impacts of TAM-derived biomarkers on the progression of OS remains limited. The GSE162454 profile was subjected to single-cell RNA (scRNA) sequencing analysis to identify crucial mediators between TAMs and OS cells. The clinical features, effects and mechanisms of these mediators on OS cells and tumour microenvironment were evaluated via biological function experiments and molecular biology experiments. Phosphodiesterase 4C (PDE4C) was identified as a pivotal mediator in the communication between M2 macrophages and OS cells. Elevated levels of PDE4C were detected in OS tissues, concomitant with M2 macrophage level, unfavourable prognosis and metastasis. The expression of PDE4C was observed to increase during the conversion process of THP-1 cells to M2 macrophages, which transferred the PDE4C mRNA to OS cells through exosome approach. PDE4C increased OS cell proliferation and mobility via upregulating the expression of collagens. Furthermore, a positive correlation was observed between elevated levels of PDE4C and increased TIDE score, decreased response rate following immune checkpoint therapy, reduced TMB and diminished PDL1 expression. Collectively, PDE4C derived from M2 macrophages has the potential to enhance the proliferation and mobility of OS cells by augmenting collagen expression. PDE4C may serve as a valuable biomarker for prognosticating patient outcomes and response rates following immunotherapy.


Subject(s)
Bone Neoplasms , Cyclic Nucleotide Phosphodiesterases, Type 4 , Immunotherapy , Macrophages , Osteosarcoma , Tumor Microenvironment , Humans , Biomarkers, Tumor/metabolism , Biomarkers, Tumor/genetics , Bone Neoplasms/immunology , Bone Neoplasms/pathology , Bone Neoplasms/genetics , Bone Neoplasms/metabolism , Cell Line, Tumor , Cell Movement , Cell Proliferation , Cyclic Nucleotide Phosphodiesterases, Type 4/metabolism , Cyclic Nucleotide Phosphodiesterases, Type 4/genetics , Gene Expression Regulation, Neoplastic , Immunotherapy/methods , Macrophages/metabolism , Macrophages/immunology , Neoplasm Metastasis , Osteosarcoma/pathology , Osteosarcoma/immunology , Osteosarcoma/genetics , Osteosarcoma/metabolism , Osteosarcoma/therapy , Prognosis , Tumor Microenvironment/immunology , Tumor-Associated Macrophages/immunology , Tumor-Associated Macrophages/metabolism
8.
Sci Transl Med ; 16(749): eadg9814, 2024 May 29.
Article in English | MEDLINE | ID: mdl-38809963

ABSTRACT

T cell-based cancer immunotherapy has typically relied on membrane-bound cytotoxicity enhancers such as chimeric antigen receptors expressed in autologous αß T cells. These approaches are limited by tonic signaling of synthetic constructs and costs associated with manufacturing. γδ T cells are an emerging alternative for cellular therapy, having innate antitumor activity, potent antibody-dependent cellular cytotoxicity, and minimal alloreactivity. We present an immunotherapeutic platform technology built around the innate properties of the Vγ9Vδ2 T cell, harnessing specific characteristics of this cell type and offering an allocompatible cellular therapy that recruits bystander immunity. We engineered γδ T cells to secrete synthetic tumor-targeting opsonins in the form of an scFv-Fc fusion protein and a mitogenic IL-15Rα-IL-15 fusion protein (stIL15). Using GD2 as a model antigen, we show that GD2-specific opsonin-secreting Vγ9Vδ2 T cells (stIL15-OPS-γδ T cells) have enhanced cytotoxicity and promote bystander activity of other lymphoid and myeloid cells. Secretion of stIL-15 abrogated the need for exogenous cytokine supplementation and further mediated activation of bystander natural killer cells. Compared with unmodified γδ T cells, stIL15-OPS-γδ T cells exhibited superior in vivo control of subcutaneous tumors and persistence in the blood. Moreover, stIL15-OPS-γδ T cells were efficacious against patient-derived osteosarcomas in animal models and in vitro, where efficacy could be boosted with the addition of zoledronic acid. Together, the data identify stIL15-OPS-γδ T cells as a candidate allogeneic cell therapy platform combining direct cytolysis with bystander activation to promote tumor control.


Subject(s)
Osteosarcoma , Receptors, Antigen, T-Cell, gamma-delta , Animals , Osteosarcoma/therapy , Osteosarcoma/immunology , Osteosarcoma/pathology , Humans , Receptors, Antigen, T-Cell, gamma-delta/metabolism , Receptors, Antigen, T-Cell, gamma-delta/immunology , Cell Line, Tumor , Cytotoxicity, Immunologic , Mice , T-Lymphocytes/immunology , Zoledronic Acid/pharmacology , Bystander Effect , Interleukin-15 , Cell Engineering
9.
Front Biosci (Landmark Ed) ; 29(5): 193, 2024 May 17.
Article in English | MEDLINE | ID: mdl-38812298

ABSTRACT

BACKGROUND: Osteosarcoma (OS) is the most prevalent orthopedic malignancy with a dismal prognosis. Disulfidptosis-related lncRNAs (DRLncs) may be related to the progression of OS, but their potential molecular regulatory role is still unclear. METHODS: Based on the data collected from The Cancer Genome Atlas (TCGA), we conducted correlation analysis and the univariate Cox analysis to screen prognosis-related DRLncs, followed by developing genotyping patterns and corresponding classifier. Subsequently, the survival analysis, enrichment analysis, drug sensitivity analysis and immune infiltration analysis were performed. Afterward, multivariate Cox regression was used to construct a risk model, which was further validated by the receiver operating characteristic (ROC) curve. The aberrant expression of hub DRLncs in OS was validated using the Reverse Transcription Polymerase Chain Reaction (RT-qPCR) assay. RESULTS: We identified 262 DRLncs and eleven prognosis-related DRLncs through filtering. We then constructed two distinct expression patterns of prognosis-related DRLncs and developed a classifier. We obtained 393 differentially expressed genes (DEGs) between different subtypes, which were significantly enriched in biological processes related to the extracellular matrix, integrin binding, focal adhesion, and Wnt signaling pathways. Through immune infiltration analysis, the activated CD4 memory T cells, resting natural killer (NK) cells, M1 macrophages, and resting dendritic cells (DC) were observed to exhibit different abundance in distinct subtypes. In the drug sensitivity analysis, tamoxifen showed a promising effect for drug-resistant OS. Furthermore, we identified five hub DRLncs and constructed a risk model. The RT-qPCR confirmed the aberrant expression of five hub DRLncs in OS. CONCLUSIONS: The present study identified DRLncs in OS, and conducted a comprehensive investigation of DRLncs-related expression patterns, survival status, immune landscape and drug sensitivity to reveal the difference in prognostic, pharmacological and immunological phenotype characteristics between distinct subtypes. Additionally, we developed a risk model to predict the prognosis, and constructed a genotyping classifier to predict the above phenotype characteristics in OS.


Subject(s)
Bone Neoplasms , Gene Expression Regulation, Neoplastic , Osteosarcoma , RNA, Long Noncoding , Humans , Osteosarcoma/genetics , Osteosarcoma/immunology , RNA, Long Noncoding/genetics , Prognosis , Bone Neoplasms/genetics , Bone Neoplasms/immunology , Biomarkers, Tumor/genetics , Biomarkers, Tumor/immunology , Genotype , Gene Expression Profiling , Tumor Microenvironment/immunology , Tumor Microenvironment/genetics , Female , Male
10.
J Cell Mol Med ; 28(9): e18286, 2024 May.
Article in English | MEDLINE | ID: mdl-38742843

ABSTRACT

Osteosarcoma, the primary bone cancer in adolescents and young adults, is notorious for its aggressive growth and metastatic potential. Our study delved into the prognostic impact of inflammasome-related gene signatures in osteosarcoma patients, employing comprehensive genetic profiling to uncover signatures linked with patient outcomes. We identified three patient subgroups through consensus clustering, with one showing worse survival rates correlated with high FGFR3 and RARB expressions. Immune profiling revealed significant immune cell infiltration differences among these subgroups, affecting survival. Utilising advanced machine learning, including StepCox and gradient boosting machine algorithms, we developed a prognostic model with a notable c-index of 0.706, highlighting CD36 and MYD88 as key genes. Higher inflammasome risk scores from our model were associated with poorer survival, corroborated across datasets. In vitro experiments validated CD36 and MYD88's roles in promoting osteosarcoma cell proliferation, invasion and migration, emphasising their therapeutic potential. This research offers new insights into inflammasomes' role in osteosarcoma, introducing novel biomarkers for risk assessment and potential therapeutic targets. Our findings suggest a pathway towards personalised treatment strategies, potentially improving patient outcomes in osteosarcoma.


Subject(s)
Biomarkers, Tumor , Bone Neoplasms , Gene Expression Regulation, Neoplastic , Inflammasomes , Osteosarcoma , Humans , Osteosarcoma/genetics , Osteosarcoma/pathology , Osteosarcoma/immunology , Osteosarcoma/mortality , Inflammasomes/metabolism , Inflammasomes/genetics , Biomarkers, Tumor/genetics , Prognosis , Bone Neoplasms/genetics , Bone Neoplasms/pathology , Bone Neoplasms/mortality , Bone Neoplasms/immunology , Bone Neoplasms/diagnosis , Gene Expression Profiling , Female , Male , Transcriptome/genetics , Cell Line, Tumor , Cell Proliferation/genetics , Adolescent , Myeloid Differentiation Factor 88/genetics , Myeloid Differentiation Factor 88/metabolism
11.
Clin Cancer Res ; 30(11): 2582-2597, 2024 Jun 03.
Article in English | MEDLINE | ID: mdl-38578281

ABSTRACT

PURPOSE: To explore the cellular cross-talk of tumor-resident mast cells (MC) in controlling the activity of cancer-associated fibroblasts (CAF) to overcome tumor microenvironment (TME) abnormalities, enhancing the efficacy of immune-checkpoint inhibitors in sarcoma. EXPERIMENTAL DESIGN: We used a coculture system followed by further validation in mouse models of fibrosarcoma and osteosarcoma with or without administration of the MC stabilizer and antihistamine ketotifen. To evaluate the contribution of ketotifen in sensitizing tumors to therapy, we performed combination studies with doxorubicin chemotherapy and anti-PD-L1 (B7-H1, clone 10F.9G2) treatment. We investigated the ability of ketotifen to modulate the TME in human sarcomas in the context of a repurposed phase II clinical trial. RESULTS: Inhibition of MC activation with ketotifen successfully suppressed CAF proliferation and stiffness of the extracellular matrix accompanied by an increase in vessel perfusion in fibrosarcoma and osteosarcoma as indicated by ultrasound shear wave elastography imaging. The improved tissue oxygenation increased the efficacy of chemoimmunotherapy, supported by enhanced T-cell infiltration and acquisition of tumor antigen-specific memory. Importantly, the effect of ketotifen in reducing tumor stiffness was further validated in sarcoma patients, highlighting its translational potential. CONCLUSIONS: Our study suggests the targeting of MCs with clinically administered drugs, such as antihistamines, as a promising approach to overcome resistance to immunotherapy in sarcomas.


Subject(s)
B7-H1 Antigen , Immune Checkpoint Inhibitors , Mast Cells , Tumor Microenvironment , Humans , Mice , Animals , Mast Cells/drug effects , Mast Cells/immunology , Tumor Microenvironment/drug effects , Tumor Microenvironment/immunology , Immune Checkpoint Inhibitors/pharmacology , Immune Checkpoint Inhibitors/therapeutic use , B7-H1 Antigen/antagonists & inhibitors , Sarcoma/drug therapy , Sarcoma/pathology , Sarcoma/immunology , Ketotifen/pharmacology , Ketotifen/therapeutic use , Cell Line, Tumor , T-Lymphocytes/immunology , T-Lymphocytes/drug effects , Xenograft Model Antitumor Assays , Lymphocytes, Tumor-Infiltrating/immunology , Lymphocytes, Tumor-Infiltrating/drug effects , Female , Cancer-Associated Fibroblasts/drug effects , Cancer-Associated Fibroblasts/pathology , Doxorubicin/pharmacology , Doxorubicin/therapeutic use , Osteosarcoma/drug therapy , Osteosarcoma/pathology , Osteosarcoma/immunology
12.
Cancer Lett ; 591: 216893, 2024 Jun 01.
Article in English | MEDLINE | ID: mdl-38636892

ABSTRACT

The oncogenic properties of Nucleobindin2 (NUCB2) have been observed in various cancer types. Nevertheless, the precise understanding of the biological functions and regulatory mechanisms of NUCB2 in osteosarcoma remains limited. This investigation reported that NUCB2 was significantly increased upon glucose deprivation-induced metabolic stress. Elevated NUCB2 suppressed glucose deprivation-induced cell death and reactive oxygen species (ROS) increase. Depletion of NUCB2 resulted in a reduction in osteosarcoma cell proliferation as well as metastatic potential in vitro and in vivo. Mechanically, NUCB2 ablation suppressed C-X-C Motif Chemokine Ligand 8 (CXCL8) expression which then reduced programmed cell death 1 ligand 1 (PD-L1) expression and stimulated anti-tumor immunity mediated through cytotoxic T cells. Importantly, a combination of NUCB2 depletion with anti-PD-L1 treatment improved anti-tumor T-cell immunity in vivo. Moreover, we further demonstrated that NUCB2 interacted with NUCKS1 to inhibit its degradation, which is responsible for the transcriptional regulation of CXCL8 expression. Altogether, the outcome emphasizes the function of NUCB2 in osteosarcoma and indicates that NUCB2 elevates osteosarcoma progression and immunosuppressive microenvironment through the NUCKS1/CXCL8 pathway.


Subject(s)
Bone Neoplasms , Calcium-Binding Proteins , Disease Progression , Interleukin-8 , Osteosarcoma , Tumor Microenvironment , Osteosarcoma/immunology , Osteosarcoma/pathology , Osteosarcoma/metabolism , Osteosarcoma/genetics , Humans , Animals , Cell Line, Tumor , Interleukin-8/metabolism , Interleukin-8/genetics , Mice , Bone Neoplasms/immunology , Bone Neoplasms/pathology , Bone Neoplasms/metabolism , Bone Neoplasms/genetics , Calcium-Binding Proteins/metabolism , Calcium-Binding Proteins/genetics , Tumor Microenvironment/immunology , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Cell Proliferation , Nerve Tissue Proteins/metabolism , Nerve Tissue Proteins/genetics , Gene Expression Regulation, Neoplastic , B7-H1 Antigen/metabolism , B7-H1 Antigen/genetics , T-Lymphocytes, Cytotoxic/immunology , Signal Transduction , Reactive Oxygen Species/metabolism
13.
Commun Biol ; 7(1): 496, 2024 Apr 24.
Article in English | MEDLINE | ID: mdl-38658617

ABSTRACT

Osteosarcoma (OS) is a heterogeneous, aggressive malignancy of the bone that disproportionally affects children and adolescents. Therapeutic interventions for OS are limited, which is in part due to the complex tumor microenvironment (TME). As such, we used single-cell RNA sequencing (scRNA-seq) to describe the cellular and molecular composition of the TME in 6 treatment-naïve dogs with spontaneously occurring primary OS. Through analysis of 35,310 cells, we identified 41 transcriptomically distinct cell types including the characterization of follicular helper T cells, mature regulatory dendritic cells (mregDCs), and 8 tumor-associated macrophage (TAM) populations. Cell-cell interaction analysis predicted that mregDCs and TAMs play key roles in modulating T cell mediated immunity. Furthermore, we completed cross-species cell type gene signature homology analysis and found a high degree of similarity between human and canine OS. The data presented here act as a roadmap of canine OS which can be applied to advance translational immuno-oncology research.


Subject(s)
Bone Neoplasms , Dog Diseases , Osteosarcoma , Sequence Analysis, RNA , Single-Cell Analysis , Tumor Microenvironment , Dogs , Animals , Osteosarcoma/genetics , Osteosarcoma/veterinary , Osteosarcoma/immunology , Osteosarcoma/pathology , Sequence Analysis, RNA/veterinary , Bone Neoplasms/genetics , Bone Neoplasms/veterinary , Bone Neoplasms/immunology , Bone Neoplasms/pathology , Dog Diseases/genetics , Dog Diseases/immunology , Dog Diseases/pathology , Tumor Microenvironment/immunology , Tumor Microenvironment/genetics , Transcriptome , Female , Gene Expression Regulation, Neoplastic , Male
14.
Sci Rep ; 14(1): 9769, 2024 04 29.
Article in English | MEDLINE | ID: mdl-38684858

ABSTRACT

As a highly aggressive bone malignancy, osteosarcoma poses a significant therapeutic challenge, especially in the setting of metastasis or recurrence. This study aimed to investigate the potential of CD8-Tex cell-associated genes as prognostic biomarkers to reveal the immunogenomic profile of osteosarcoma and guide therapeutic decisions. mRNA expression data and clinical details of osteosarcoma patients were obtained from the TCGA database (TARGET-OS dataset). The GSE21257 dataset (from the GEO database) was used as an external validation set to provide additional information on osteosarcoma specimens. 84 samples from the TARGET-OS dataset were used as the training set, and 53 samples from the GSE21257 dataset served as the external validation cohort. Univariate Cox regression analysis was utilized to identify CD8 Tex cell genes associated with prognosis. The LASSO algorithm was performed for 1000 iterations to select the best subset to form the CD8 Tex cell gene signature (TRS). Final genes were identified using the multivariate Cox regression model of the LASSO algorithm. Risk scores were calculated to categorize patients into high- and low-risk groups, and clinical differences were explored by Kaplan-Meier survival analysis to assess model performance. Prediction maps were constructed to estimate 1-, 3-, and 5 year survival rates for osteosarcoma patients, including risk scores for CD8 Texcell gene markers and clinicopathologic factors. The ssGSEA algorithm was used to assess the differences in immune function between TRS-defined high- and low-risk groups. TME and immune cell infiltration were further assessed using the ESTIMATE and CIBERSORT algorithms. To explore the relationship between immune checkpoint gene expression levels and the two risk-defined groups. A CD8 Tex cell-associated gene signature was extracted from the TISCH database and prognostic markers including two genes were developed. The high-risk group showed lower survival, and model performance was validated by ROC curves and C-index. Predictive plots were constructed to demonstrate survival estimates, combining CD8 Tex cell gene markers and clinical factors. This study provides valuable insights into the molecular and immune characteristics of osteosarcoma and offers potential avenues for advances in therapeutic approaches.


Subject(s)
Biomarkers, Tumor , Bone Neoplasms , CD8-Positive T-Lymphocytes , Osteosarcoma , Osteosarcoma/genetics , Osteosarcoma/immunology , Osteosarcoma/mortality , Osteosarcoma/pathology , Humans , Prognosis , Bone Neoplasms/genetics , Bone Neoplasms/mortality , Bone Neoplasms/immunology , Bone Neoplasms/pathology , Male , Female , Biomarkers, Tumor/genetics , CD8-Positive T-Lymphocytes/immunology , Gene Expression Regulation, Neoplastic , Transcriptome , Kaplan-Meier Estimate , Adult , Gene Expression Profiling , Adolescent
15.
Cancer Biomark ; 39(4): 299-312, 2024.
Article in English | MEDLINE | ID: mdl-38250759

ABSTRACT

BACKGROUND: Osteosarcoma (OS) is a relatively rare malignant bone tumor in teenagers; however, its molecular mechanisms are not yet understood comprehensively. OBJECTIVE: The study aimed to use necroptosis-related genes (NRGs) and their relationships with immune-related genes to construct a prognostic signature for OS. METHODS: TARGET-OS was used as the training dataset, and GSE 16091 and GSE 21257 were used as the validation datasets. Univariate regression, survival analysis, and Kaplan-Meier curves were used to screen for hub genes. The immune-related targets were screened using immune infiltration assays and immune checkpoints. The results were validated using nomogram and decision curve analyses (DCA). RESULTS: Using univariate Cox regression analysis, TNFRSF1A was screened from 14 NRGs as an OS prognostic signature. Functional enrichment was analyzed based on the median expression of TNFRSF1A. The prognosis of the TNFRSF1A low-expression group in the Kaplan-Meier curve was notably worse. Immunohistochemistry analysis showed that the number of activated T cells and tumor purity increased considerably. Furthermore, the immune checkpoint lymphocyte activation gene 3 (LAG-3) is a possible target for intervention. The nomogram accurately predicted 1-, 3-, and 5-year survival rates. DCA validated the model (C = 0.669). Conclusion: TNFRSF1A can be used to elucidate the potential relationship between the immune microenvironment and NRGs in OS pathogenesis.


Subject(s)
Biomarkers, Tumor , Bone Neoplasms , Osteosarcoma , Receptors, Tumor Necrosis Factor, Type I , Humans , Osteosarcoma/genetics , Osteosarcoma/mortality , Osteosarcoma/immunology , Osteosarcoma/pathology , Receptors, Tumor Necrosis Factor, Type I/genetics , Receptors, Tumor Necrosis Factor, Type I/metabolism , Biomarkers, Tumor/genetics , Bone Neoplasms/genetics , Bone Neoplasms/pathology , Bone Neoplasms/mortality , Bone Neoplasms/immunology , Prognosis , Female , Male , Nomograms , Adolescent , Kaplan-Meier Estimate , Gene Expression Regulation, Neoplastic , Tumor Microenvironment/immunology , Tumor Microenvironment/genetics
16.
Biochem Genet ; 61(6): 2363-2381, 2023 Dec.
Article in English | MEDLINE | ID: mdl-37041365

ABSTRACT

Little progress has been made in the treatment and prognosis of osteosarcoma in the past 40 years. Tumor microenvironment (TME) plays a critical role in the progression of osteosarcoma. This study aims to determine immune-associated prognostic biomarkers for osteosarcoma patients. With the help of analytical tools including ESTIMATE, differential gene expression, LASSO, and univariate cox and multivariate cox regression analysis, osteosarcoma gene expression data from Gene Expression Omnibus (GEO) databases were investigated. Following the establishment of a prognostic risk score model, internal and external validations using the GEO and TARGET databases were carried out. A total of 44 and 55 samples respectively in the GSE21257 and the TARGET databases were included. Our analysis found 93 differentially expressed genes (DEGs) between the high and low-ImmuneScore groups. Through univariate cox and LASSO analysis, ALOX5AP was identified as an indicator of TME in osteosarcomas. ALOX5AP was then used to construct a prognostic risk model. Internal and external verification revealed that higher expression of ALOX5AP was correlated with lower risk. Through the CIBERSORT algorithm, the level of CD8 T cells was found to negatively correlate with the risk score. This study revealed that ALOX5AP is an indicator for predicting high CD8 lymphocyte infiltration and "hot" tumor microenvironment in osteosarcomas. Thus, ALOX5AP has the potential to act as a biomarker for effective immunotherapies in osteosarcoma patients.


Subject(s)
5-Lipoxygenase-Activating Proteins , CD8-Positive T-Lymphocytes , Osteosarcoma , Tumor Microenvironment , 5-Lipoxygenase-Activating Proteins/genetics , Humans , CD8-Positive T-Lymphocytes/immunology , Osteosarcoma/genetics , Osteosarcoma/immunology , Computational Biology , Risk Factors , Biomarkers, Tumor/genetics , Lymphocytes, Tumor-Infiltrating
17.
Zhongguo Yi Xue Ke Xue Yuan Xue Bao ; 44(1): 110-117, 2022 Feb.
Article in Chinese | MEDLINE | ID: mdl-35300772

ABSTRACT

Objective To screen the potential key genes of osteosarcoma by bioinformatics methods and analyze their immune infiltration patterns. Methods The gene expression profiles GSE16088 and GSE12865 associated with osteosarcoma were obtained from the Gene Expression Omnibus(GEO),and the differentially expressed genes(DEGs)related to osteosarcoma were screened by bioinformatics tools.Gene Ontology(GO)annotation,Kyoto Encyclopedia of Genes and Genomes(KEGG)pathway enrichment,and analysis of immune cell infiltration were then carried out for the DEGs.The potential Hub genes of osteosarcoma were identified by protein-protein interaction network,and the expression of Hub genes in osteosarcoma and normal tissue samples was verified via the Cancer Genome Atlas(TCGA). Results A total of 108 DEGs were screened out.GO annotation and KEGG pathway enrichment revealed that the DEGs were mainly involved in integrin binding,extracellular matrix (ECM) structural components,ECM receptor interactions,and phosphatidylinositol 3-kinase/protein kinase B(PI3K/Akt)signaling pathway.Macrophages were the predominant infiltrating immune cells in osteosarcoma.Secreted phosphoprotein 1(SPP1),matrix metallopeptidase 2(MMP2),lysyl oxidase(LOX),collagen type V alpha(II)chain(COL5A2),and melanoma cell adhesion molecule(MCAM)presented differential expression between osteosarcoma and normal tissue samples(all P<0.05). Conclusions SPP1,MMP2,LOX,COL5A2,and MCAM are all up-regulated in osteosarcoma,which may serve as potential biomarkers of osteosarcoma.Macrophages are the key infiltrating immune cells in osteosarcoma,which may provide new perspectives for the treatment of osteosarcoma.


Subject(s)
Bone Neoplasms , Osteosarcoma , Tumor-Associated Macrophages , Bone Neoplasms/genetics , Bone Neoplasms/immunology , Computational Biology/methods , Gene Expression Profiling/methods , Humans , Osteosarcoma/genetics , Osteosarcoma/immunology , Phosphatidylinositol 3-Kinases/genetics , Tumor-Associated Macrophages/immunology
18.
Proc Natl Acad Sci U S A ; 119(6)2022 02 08.
Article in English | MEDLINE | ID: mdl-35101971

ABSTRACT

Metastatic osteosarcoma has a poor prognosis with a 2-y, event-free survival rate of ∼15 to 20%, highlighting the need for the advancement of efficacious therapeutics. Chimeric antigen receptor (CAR) T-cell therapy is a potent strategy for eliminating tumors by harnessing the immune system. However, clinical trials with CAR T cells in solid tumors have encountered significant challenges and have not yet demonstrated convincing evidence of efficacy for a large number of patients. A major bottleneck for the success of CAR T-cell therapy is our inability to monitor the accumulation of the CAR T cells in the tumor with clinical-imaging techniques. To address this, we developed a clinically translatable approach for labeling CAR T cells with iron oxide nanoparticles, which enabled the noninvasive detection of the iron-labeled T cells with magnetic resonance imaging (MRI), photoacoustic imaging (PAT), and magnetic particle imaging (MPI). Using a custom-made microfluidics device for T-cell labeling by mechanoporation, we achieved significant nanoparticle uptake in the CAR T cells, while preserving T-cell proliferation, viability, and function. Multimodal MRI, PAT, and MPI demonstrated homing of the T cells to osteosarcomas and off-target sites in animals administered with T cells labeled with the iron oxide nanoparticles, while T cells were not visualized in animals infused with unlabeled cells. This study details the successful labeling of CAR T cells with ferumoxytol, thereby paving the way for monitoring CAR T cells in solid tumors.


Subject(s)
Bone Neoplasms , Ferrosoferric Oxide/pharmacology , Immunotherapy, Adoptive , Magnetic Resonance Imaging , Nanoparticles/therapeutic use , Neoplasms, Experimental , Osteosarcoma , Receptors, Chimeric Antigen/immunology , T-Lymphocytes/immunology , Animals , Bone Neoplasms/diagnostic imaging , Bone Neoplasms/immunology , Bone Neoplasms/therapy , Mice , Neoplasms, Experimental/diagnostic imaging , Neoplasms, Experimental/immunology , Neoplasms, Experimental/therapy , Osteosarcoma/diagnostic imaging , Osteosarcoma/immunology , Osteosarcoma/therapy
19.
Cytotherapy ; 24(6): 567-576, 2022 06.
Article in English | MEDLINE | ID: mdl-35193828

ABSTRACT

Osteosarcoma (OS) is one of the most common malignancies in children and adolescents. Multimodal chemotherapy and aggressive surgical resection have improved the prognosis of patients with osteosarcoma. However, the prognosis of OS patients with unresectable advanced tumors, distant metastasis or chemotherapy is still poor. Chimeric antigen receptor (CAR) T cells have achieved remarkable success in the treatment of hematologic malignancies, injecting new vitality into the field of adoptive cell therapy. However, the efficacy in solid tumors has been largely limited. The reason for the poor curative effect of solid tumors is mainly the heterogeneity of solid tumor antigen, immune escape, tumor microenvironment barrier, resistance of immunosuppressive cells and inhibitory factors, which lead to the obstruction of CAR T cell infiltration and the aggravation of failure. Potential antigenic targets for osteosarcoma CAR T cell therapy are under continuous exploration. Some of the antigenic targets, such as anti-HER2-CAR T cells, have achieved good results in preclinical studies, and some of them have entered clinical studies and achieved certain clinical effects. In this review, we discuss the research progress of potential antigen targets and osteosarcoma microenvironment of CAR T cells in the treatment of osteosarcoma.


Subject(s)
Bone Neoplasms , Immunotherapy, Adoptive , Osteosarcoma , Receptors, Chimeric Antigen , Adolescent , Bone Neoplasms/immunology , Bone Neoplasms/therapy , Child , Humans , Immunotherapy, Adoptive/methods , Osteosarcoma/immunology , Osteosarcoma/therapy , Receptors, Chimeric Antigen/immunology , Tumor Microenvironment
20.
Bioengineered ; 13(2): 3751-3759, 2022 02.
Article in English | MEDLINE | ID: mdl-35081874

ABSTRACT

Osteosarcoma (OS) is solid tumors with high malignancy and incidence starting in the bones. OS pathogenesis has been proved to be closely associated with immune imbalance, and follicular helper T cells (Tfh) significantly affect host humoral immune homeostasis. However, there are few reports on the effect of Tfh cell activation on the prognosis of OS patients. Hence, this investigation on the changes in the proportion of peripheral blood Tfh cells in OS patients, and the relationship between their activity level and OS prognosis. We collected peripheral blood from patients with OS, benign bone tumor (BT group) and healthy subjects (Healthy group), respectively. The number of CD4+CXCR5+ Tfh cell in peripheral blood was measured by flow cytometry and correlation analysis between its activity and OS clinicopathological characteristics was carried out. The data showed that in comparison with the BT and Healthy groups, higher proportion and activation level of peripheral blood CD4+CXCR5+ Tfh cells in CD4+ T cells were found in the OS group. In OS patients, increases of the proportion and activity level of Tfh cells were associated with poorly differentiated OS and tumor metastasis. Additionally, Kaplan-Meier and Cox regression analysis showed a longer overall survival in patients with low proportion of peripheral blood CD4+CXCR5+ Tfh cells in CD4+ T cells, and their activation level may be a prognostic factor in OS patients. In conclusion, peripheral blood CD4+CXCR5+ Tfh cell activation in OS patients was associated with a poor prognosis. This study provided ideas for improving the clinical treatment of OS patients.


Subject(s)
Bone Neoplasms , Osteosarcoma , T Follicular Helper Cells , Adolescent , Adult , Bone Neoplasms/blood , Bone Neoplasms/immunology , Bone Neoplasms/mortality , Disease-Free Survival , Female , Flow Cytometry , Follow-Up Studies , Humans , Male , Osteosarcoma/blood , Osteosarcoma/immunology , Osteosarcoma/mortality , Survival Rate , T Follicular Helper Cells/immunology , T Follicular Helper Cells/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...