Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 647
Filter
1.
Int J Mol Sci ; 25(11)2024 Jun 04.
Article in English | MEDLINE | ID: mdl-38892366

ABSTRACT

In order to overcome the resistance to radiotherapy in human chondrosarcoma cells, the prevention from efficient DNA repair with a combined treatment with the DNA-dependent protein kinase catalytic subunit (DNA-PKcs) inhibitor AZD7648 was explored for carbon ion (C-ion) as well as reference photon (X-ray) irradiation (IR) using gene expression analysis, flow cytometry, protein phosphorylation, and telomere length shortening. Proliferation markers and cell cycle distribution changed significantly after combined treatment, revealing a prominent G2/M arrest. The expression of the G2/M checkpoint genes cyclin B, CDK1, and WEE1 was significantly reduced by IR alone and the combined treatment. While IR alone showed no effects, additional AZD7648 treatment resulted in a dose-dependent reduction in AKT phosphorylation and an increase in Chk2 phosphorylation. Twenty-four hours after IR, the key genes of DNA repair mechanisms were reduced by the combined treatment, which led to impaired DNA repair and increased radiosensitivity. A time-dependent shortening of telomere length was observed in both cell lines after combined treatment with AZD7648 and 8 Gy X-ray/C-ion IR. Our data suggest that the inhibition of DNA-PKcs may increase sensitivity to X-rays and C-ion IR by impairing its functional role in DNA repair mechanisms and telomere end protection.


Subject(s)
Chondrosarcoma , DNA-Activated Protein Kinase , Heavy Ion Radiotherapy , Telomere , Humans , DNA-Activated Protein Kinase/antagonists & inhibitors , DNA-Activated Protein Kinase/metabolism , DNA-Activated Protein Kinase/genetics , Cell Line, Tumor , Chondrosarcoma/metabolism , Chondrosarcoma/genetics , Chondrosarcoma/radiotherapy , Chondrosarcoma/drug therapy , Telomere/drug effects , Telomere/metabolism , Cell Cycle Checkpoints/drug effects , Cell Cycle Checkpoints/radiation effects , DNA Repair/drug effects , Radiation Tolerance/drug effects , Pyrazoles/pharmacology , Cell Proliferation/drug effects , Bone Neoplasms/metabolism , Bone Neoplasms/genetics , Bone Neoplasms/pathology , Bone Neoplasms/drug therapy , G2 Phase Cell Cycle Checkpoints/drug effects , G2 Phase Cell Cycle Checkpoints/radiation effects
2.
Cells ; 13(11)2024 Jun 04.
Article in English | MEDLINE | ID: mdl-38891109

ABSTRACT

Chondrosarcoma (ChS), a malignant cartilage-producing tumor, is the second most frequently diagnosed osseous sarcoma after osteosarcoma. It represents a very heterogeneous group of malignant chemo- and radiation-resistant neoplasms, accounting for approximately 20% of all bone sarcomas. The majority of ChS patients have a good prognosis after a complete surgical resection, as these tumors grow slowly and rarely metastasize. Conversely, patients with inoperable disease, due to the tumor location, size, or metastases, represent a great clinical challenge. Despite several genetic and epigenetic alterations that have been described in distinct ChS subtypes, very few therapeutic options are currently available for ChS patients. Therefore, new prognostic factors for tumor progression as well as new treatment options have to be explored, especially for patients with unresectable or metastatic disease. Recent studies have shown that a correlation between immune infiltrate composition, tumor aggressiveness, and survival does exist in ChS patients. In addition, the intra-tumor microvessel density has been proven to be associated with aggressive clinical behavior and a high metastatic potential in ChS. This review will provide an insight into the ChS microenvironment, since immunotherapy and antiangiogenic agents are emerging as interesting therapeutic options for ChS patients.


Subject(s)
Chondrosarcoma , Tumor Microenvironment , Humans , Chondrosarcoma/pathology , Chondrosarcoma/genetics , Chondrosarcoma/metabolism , Bone Neoplasms/pathology , Bone Neoplasms/therapy , Bone Neoplasms/metabolism , Bone Neoplasms/genetics , Immunotherapy , Angiogenesis Inhibitors/therapeutic use , Angiogenesis Inhibitors/pharmacology
3.
Int J Mol Sci ; 25(10)2024 May 09.
Article in English | MEDLINE | ID: mdl-38791180

ABSTRACT

Chondrosarcoma is a malignant bone tumor that arises from abnormalities in cartilaginous tissue and is associated with lung metastases. Lymphangiogenesis plays an essential role in cancer metastasis. Visfatin is an adipokine reported to enhance tumor metastasis, but its relationship with VEGF-D generation and lymphangiogenesis in chondrosarcoma remains undetermined. Our results from clinical samples reveal that VEGF-D levels are markedly higher in chondrosarcoma patients than in normal individuals. Visfatin stimulation promotes VEGF-D-dependent lymphatic endothelial cell lymphangiogenesis. We also found that visfatin induces VEGF-D production by activating HIF-1α and reducing miR-2277-3p generation through the Raf/MEK/ERK signaling cascade. Importantly, visfatin controls chondrosarcoma-related lymphangiogenesis in vivo. Therefore, visfatin is a promising target in the treatment of chondrosarcoma lymphangiogenesis.


Subject(s)
Bone Neoplasms , Chondrosarcoma , Hypoxia-Inducible Factor 1, alpha Subunit , Lymphangiogenesis , MicroRNAs , Nicotinamide Phosphoribosyltransferase , Vascular Endothelial Growth Factor D , Humans , Chondrosarcoma/metabolism , Chondrosarcoma/genetics , Chondrosarcoma/pathology , Lymphangiogenesis/genetics , MicroRNAs/genetics , MicroRNAs/metabolism , Nicotinamide Phosphoribosyltransferase/metabolism , Nicotinamide Phosphoribosyltransferase/genetics , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Hypoxia-Inducible Factor 1, alpha Subunit/genetics , Vascular Endothelial Growth Factor D/metabolism , Vascular Endothelial Growth Factor D/genetics , Bone Neoplasms/metabolism , Bone Neoplasms/pathology , Bone Neoplasms/genetics , Animals , Gene Expression Regulation, Neoplastic , Cell Line, Tumor , Mice , Cytokines/metabolism , Male , Female , MAP Kinase Signaling System
4.
J Cancer Res Ther ; 20(2): 522-530, 2024 Apr 01.
Article in English | MEDLINE | ID: mdl-38687921

ABSTRACT

ABSTRACT: Bone sarcomas encompass a group of spontaneous mesenchymal malignancies, among which osteosarcoma, Ewing sarcoma, chondrosarcoma, and chordoma are the most common subtypes. Chondrosarcoma, a relatively prevalent malignant bone tumor that originates from chondrocytes, is characterized by endogenous cartilage ossification within the tumor tissue. Despite the use of aggressive treatment approaches involving extensive surgical resection, chemotherapy, and radiotherapy for patients with osteosarcoma, chondrosarcoma, and chordoma, limited improvements in patient outcomes have been observed. Furthermore, resistance to chemotherapy and radiation therapy has been observed in chondrosarcoma and chordoma cases. Consequently, novel therapeutic approaches for bone sarcomas, including chondrosarcoma, need to be uncovered. Recently, the emergence of immunotherapy and immune checkpoint inhibitors has garnered attention given their clinical success in various diverse types of cancer, thereby prompting investigations into their potential for managing chondrosarcoma. Considering that circumvention of immune surveillance is considered a key factor in the malignant progression of tumors and that immune checkpoints play an important role in modulating antitumor immune effects, blockers or inhibitors targeting these immune checkpoints have become effective therapeutic tools for patients with tumors. One such checkpoint receptor implicated in this process is programmed cell death protein-1 (PD-1). The association between PD-1 and programmed cell death ligand-1 (PD-L1) and cancer progression in humans has been extensively studied, highlighting their remarkable potential as biomarkers for cancer treatment. This review comprehensively examines available studies on current chondrosarcoma treatments and advancements in anti-PD-1/PD-L1 blockade therapy for chondrosarcoma.


Subject(s)
B7-H1 Antigen , Bone Neoplasms , Chondrosarcoma , Immune Checkpoint Inhibitors , Programmed Cell Death 1 Receptor , Humans , Chondrosarcoma/therapy , Chondrosarcoma/pathology , Chondrosarcoma/metabolism , B7-H1 Antigen/antagonists & inhibitors , B7-H1 Antigen/metabolism , Programmed Cell Death 1 Receptor/antagonists & inhibitors , Programmed Cell Death 1 Receptor/metabolism , Bone Neoplasms/pathology , Bone Neoplasms/therapy , Bone Neoplasms/metabolism , Bone Neoplasms/immunology , Bone Neoplasms/drug therapy , Immune Checkpoint Inhibitors/therapeutic use , Immune Checkpoint Inhibitors/pharmacology , Immunotherapy/methods
5.
Mod Pathol ; 37(5): 100464, 2024 May.
Article in English | MEDLINE | ID: mdl-38447752

ABSTRACT

Extraskeletal myxoid chondrosarcoma (EMC) is an uncommon mesenchymal neoplasm characteristically composed of uniform-appearing round to spindle-shaped cells with eosinophilic cytoplasm and abundant myxoid extracellular matrix. Although the majority of cases harbor a pathognomonic t(9;22) translocation that fuses EWSR1 with the orphan nuclear receptor NR4A3, there are less common variants that partner NR4A3 with TAF15, TCF12, or TFG. By immunohistochemistry, EMC has features of both cartilaginous and neuroendocrine differentiation, as evidenced by inconsistent expression of S100 protein and synaptophysin or INSM1, respectively, in a subset of cases. Given the limitations of available immunohistochemical stains for the diagnosis of EMC, we analyzed genome-wide gene expression microarray data to identify candidate biomarkers based on differential expression in EMC in comparison with other mesenchymal neoplasms. This analysis pointed to CHRNA6 as the gene with the highest relative expression in EMC (96-fold; P = 8.2 × 10-26) and the only gene with >50-fold increased expression in EMC compared with other tumors. Using RNA chromogenic in situ hybridization, we observed strong and diffuse expression of CHRNA6 in 25 cases of EMC, including both EWSR1-rearranged and TAF15-rearranged variants. All examined cases of histologic mimics were negative for CHRNA6 overexpression; however, limited CHRNA6 expression, not reaching a threshold of >5 puncta or 1 aggregate of chromogen in >25% of cells, was observed in 69 of 685 mimics (10.1%), spanning an array of mesenchymal tumors. Taken together, these findings suggest that, with careful interpretation and the use of appropriate thresholds, CHRNA6 RNA chromogenic in situ hybridization is a potentially useful ancillary histologic tool for the diagnosis of EMC.


Subject(s)
Biomarkers, Tumor , Chondrosarcoma , In Situ Hybridization , Neoplasms, Connective and Soft Tissue , Humans , Biomarkers, Tumor/genetics , Biomarkers, Tumor/analysis , Chondrosarcoma/genetics , Chondrosarcoma/pathology , Chondrosarcoma/diagnosis , Chondrosarcoma/metabolism , Neoplasms, Connective and Soft Tissue/genetics , Neoplasms, Connective and Soft Tissue/pathology , Neoplasms, Connective and Soft Tissue/diagnosis , Female , Male , Middle Aged , Aged , In Situ Hybridization/methods , Adult , Receptors, Nicotinic/genetics , Receptors, Nicotinic/metabolism , Neoplasms, Connective Tissue/genetics , Neoplasms, Connective Tissue/pathology , Neoplasms, Connective Tissue/diagnosis , Aged, 80 and over , Immunohistochemistry
6.
Int J Radiat Biol ; 100(5): 724-735, 2024.
Article in English | MEDLINE | ID: mdl-38442236

ABSTRACT

PURPOSE: Radiation-induced bystander effect (RIBE) frequently is seen as DNA damage in unirradiated bystander cells, but the repair processes initiated in response to that DNA damage are not well understood. RIBE-mediated formation of micronuclei (MN), a biomarker of persistent DNA damage, was previously observed in bystander normal fibroblast (AG01522) cells, but not in bystander human chondrosarcoma (HTB94) cells. The molecular mechanisms causing this disparity are not clear. Herein, we investigate the role of DNA repair in the bystander responses of the two cell lines. METHODS: Cells were irradiated with X-rays and immediately co-cultured with un-irradiated cells using a trans-well insert system in which they share the same medium. The activation of DNA damage response (DDR) proteins was detected by immunofluorescence staining or Western blotting. MN formation was examined by the cytokinesis-block MN assay, which is a robust method to detect persistent DNA damage. RESULTS: Immunofluorescent foci of γH2AX and 53BP1, biomarkers of DNA damage and repair, revealed a greater capacity for DNA repair in HTB94 cells than in AG01522 cells in both irradiated and bystander populations. Autophosphorylation of ATR at the threonine 1989 site was expressed at a greater level in HTB94 cells compared to AG01522 cells at the baseline and in response to hydroxyurea treatment or exposure to 1 Gy of X-rays. An inhibitor of ATR, but not of ATM, promoted MN formation in bystander HTB94 cells. In contrast, no effect of either inhibitor was observed in bystander AG01522 cells, indicating that ATR signaling might be a pivotal pathway to preventing the MN formation in bystander HTB94 cells. Supporting this idea, we found an ATR-dependent increase in the fractions of bystander HTB94 cells with pRPA2 S33 and RAD51 foci. A blocker of RAD51 facilitated MN formation in bystander HTB94 cells. CONCLUSION: Our results indicate that HTB94 cells were likely more efficient in DNA repair than AG01522 cells, specifically via ATR signaling, which inhibited the bystander signal-induced MN formation. This study highlights the significance of DNA repair efficiency in bystander cell responses.


Subject(s)
Ataxia Telangiectasia Mutated Proteins , Bystander Effect , Chondrosarcoma , DNA Repair , Rad51 Recombinase , Signal Transduction , Humans , Ataxia Telangiectasia Mutated Proteins/metabolism , Bystander Effect/radiation effects , Cell Line, Tumor , Chondrosarcoma/metabolism , Chondrosarcoma/radiotherapy , DNA Damage , Histones/metabolism , Rad51 Recombinase/metabolism
7.
Environ Toxicol ; 39(7): 3790-3798, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38497692

ABSTRACT

Chondrosarcoma is a malignant bone tumor that arises from abnormalities in cartilaginous tissue and is associated with lung metastases. Extracellular vesicles called exosomes are primarily used as mediators of intercellular signal transmission to control tumor metastasis. Visfatin is an adipokine reported to enhance tumor metastasis, but its relationship with exosome generation in chondrosarcoma motility remains undetermined. Our results found that overexpressing visfatin augments the production of exosomes from chondrosarcoma cells. Visfatin-treated chondrosarcoma exosomes educate macrophage polarization towards the M2 but not M1 phenotype. Interestingly, M2 macrophages polarized by exosomes return to chondrosarcoma cells to facilitate cell motility. According to these findings, chondrosarcoma cells emit more exosomes when treated with visfatin. The stimulation of exosome generation by visfatin polarizes M2 macrophages and enhances the motility of chondrosarcoma.


Subject(s)
Cell Movement , Chondrosarcoma , Exosomes , Macrophages , Nicotinamide Phosphoribosyltransferase , Chondrosarcoma/pathology , Chondrosarcoma/metabolism , Exosomes/metabolism , Macrophages/drug effects , Macrophages/metabolism , Humans , Nicotinamide Phosphoribosyltransferase/metabolism , Cell Movement/drug effects , Cell Line, Tumor , Bone Neoplasms/pathology , Bone Neoplasms/metabolism , Cytokines/metabolism
8.
Genes (Basel) ; 15(2)2024 02 14.
Article in English | MEDLINE | ID: mdl-38397231

ABSTRACT

Background: Undifferentiated pleomorphic sarcoma of bone (UPSb) is a rare primary bone sarcoma that lacks a specific line of differentiation. Distinguishing between UPSb and other malignant bone sarcomas, including dedifferentiated chondrosarcoma and osteosarcoma, is challenging due to their overlapping features. We have previously identified that UPSb tumours have elevated mRNA levels of Fibroblast Growth Factor 23 (FGF23) transcripts compared to other sarcomas including osteosarcoma. In the present study, we evaluated the specificity and practicality of FGF23 immunoreactivity as a specific diagnostic tool to differentiate UPSb tumours from osteosarcomas and dedifferentiated chondrosarcomas. Methods: A total of 10 UPSb, 10 osteosarcoma, and 10 dedifferentiated chondrosarcoma cases (all high-grade), were retrieved and immunohistochemistry for FGF23 was performed. Results: FGF23 protein was expressed at high levels in 80-90% of undifferentiated pleomorphic sarcoma of the bone cases, whereas it was expressed at significantly lower levels in dedifferentiated chondrosarcoma and osteosarcoma cases. A semiquantitative analysis, considering the intensity of immunoreactivity, confirmed significantly elevated FGF23 expression levels in UPSb tissues compared to those observed in osteosarcoma and dedifferentiated chondrosarcoma tissues. Conclusions: The results we present here suggest that FGF23 immunohistochemistry may be a useful tool to aid in differentiating UPSb from morphologically similar malignant bone sarcomas, especially in situations where sampling is restricted and there is limited clinical information available.


Subject(s)
Bone Neoplasms , Chondrosarcoma , Fibroblast Growth Factor-23 , Osteosarcoma , Sarcoma , Humans , Bone Neoplasms/diagnosis , Bone Neoplasms/genetics , Chondrosarcoma/diagnosis , Chondrosarcoma/genetics , Chondrosarcoma/metabolism , Osteosarcoma/diagnosis , Osteosarcoma/genetics , Sarcoma/diagnosis , Sarcoma/genetics , Sarcoma/pathology , Fibroblast Growth Factor-23/metabolism
9.
Sci Rep ; 13(1): 16650, 2023 10 03.
Article in English | MEDLINE | ID: mdl-37789008

ABSTRACT

Histological analysis is the gold standard method for cancer diagnosis. However, it is prone to subjectivity and sampling bias. In response to these limitations, we introduce a quantitative bimodal approach that aims to provide non-invasive guidance towards suspicious regions. Light backscattering spectroscopy and quantitative ultrasound techniques were combined to characterize two different bone tumor types from animal models: chondrosarcomas and osteosarcomas. Two different cell lines were used to induce osteosarcoma growth. Histological analyses were conducted to serve as references. Three ultrasound parameters and intensities of the light reflectance profiles showed significant differences between chondrosarcomas and osteosarcomas at the 5% level. Likewise, variations in the same biomarkers were reported for the two types of osteosarcoma, despite their similar morphology observed in the histological examinations. These observations show the sensitivity of our techniques in probing fine tissue properties. Secondly, the ultrasound spectral-based technique identified the mean size of chondrosarcoma cells and nuclei with relative errors of about 22% and 9% respectively. The optical equivalent technique correctly extracted scatterer size distributions that encompass nuclei and cells for chondrosarcomas and osteosarcomas ([Formula: see text] and [Formula: see text] respectively). The optical scattering contributions of nuclei were estimated at 52% for the chondrosarcomas and 69% for the osteosarcomas, probably indicating the abundant and the absent extracellular matrix respectively. Thus, the ultrasound and the optical methods brought complementary parameters. They successfully estimated morphological parameters at the cellular and the nuclear scales, making our bimodal technique promising for tumor characterization.


Subject(s)
Bone Neoplasms , Chondrosarcoma , Osteosarcoma , Sarcoma , Soft Tissue Neoplasms , Animals , Bone Neoplasms/metabolism , Osteosarcoma/pathology , Chondrosarcoma/diagnostic imaging , Chondrosarcoma/metabolism , Spectrum Analysis
10.
Cell Rep ; 42(6): 112578, 2023 06 27.
Article in English | MEDLINE | ID: mdl-37267108

ABSTRACT

Chondrosarcomas are the most common malignancy of cartilage and are associated with somatic mutations in isocitrate dehydrogenase 1 (IDH1) and IDH2 genes. Somatic IDH mutations are also found in its benign precursor lesion, enchondromas, suggesting that IDH mutations are early events in malignant transformation. Human mutant IDH chondrosarcomas and mutant Idh mice that develop enchondromas investigated in our studies display glycogen deposition exclusively in mutant cells from IDH mutant chondrosarcomas and Idh1 mutant murine growth plates. Pharmacologic blockade of glycogen utilization induces changes in tumor cell behavior, downstream energetic pathways, and tumor burden in vitro and in vivo. Mutant IDH1 interacts with hypoxia-inducible factor 1α (HIF1α) to regulate expression of key enzymes in glycogen metabolism. Here, we show a critical role for glycogen in enchondromas and chondrosarcomas, which is likely mediated through an interaction with mutant IDH1 and HIF1α.


Subject(s)
Chondroma , Chondrosarcoma , Isocitrate Dehydrogenase , Animals , Humans , Mice , Bone Neoplasms/metabolism , Cartilage/metabolism , Chondrosarcoma/genetics , Chondrosarcoma/metabolism , Chondrosarcoma/pathology , Isocitrate Dehydrogenase/genetics , Isocitrate Dehydrogenase/metabolism , Mutation/genetics
11.
J Pineal Res ; 75(1): e12872, 2023 Aug.
Article in English | MEDLINE | ID: mdl-37057370

ABSTRACT

Chondrosarcoma has a high propensity to metastasize and responds poorly to chemotherapy and radiation treatment. The enzymatic activity of matrix metalloproteinases (MMPs) is very important in chondrosarcoma metastasis. Melatonin exhibits anticarcinogenic activity in many types of cancers by suppressing the expression of certain MMP family members, but this has not yet been clearly determined in chondrosarcoma. Our study demonstrates that MMP7 plays an essential role in chondrosarcoma cell proliferation, migration, and anoikis resistance. We also found that MMP7 is highly expressed in chondrosarcomas. Our in vitro and in vivo investigations show that melatonin strongly inhibits chondrosarcoma cell proliferation, migration, and anoikis resistance by directly suppressing MMP7 expression. Melatonin reduced MMP7 synthesis by promoting levels of miR-520f-3p expression, which were downregulated in human chondrosarcoma tissue samples. Pharmacological inhibition of miR-520f-3p markedly reversed the effects of melatonin upon chondrosarcoma proliferation and metastasis. Thus, our study suggests that melatonin has therapeutic potential for reducing the tumorigenesis and metastatic potential of chondrosarcoma via the miR-520f-3p/MMP7 axis.


Subject(s)
Chondrosarcoma , Melatonin , MicroRNAs , Humans , MicroRNAs/genetics , Cell Line, Tumor , Melatonin/pharmacology , Matrix Metalloproteinase 7/metabolism , Cell Proliferation , Chondrosarcoma/drug therapy , Chondrosarcoma/genetics , Chondrosarcoma/metabolism , Cell Movement , Gene Expression Regulation, Neoplastic
12.
Histopathology ; 82(6): 937-945, 2023 May.
Article in English | MEDLINE | ID: mdl-36754860

ABSTRACT

AIMS: Extraskeletal myxoid chondrosarcoma (EMC) is a rare form of adult sarcoma with distinct histology and NR4A3 gene fusion. Immunohistochemically, EMCs are variably positive for S100 protein and neuroendocrine markers. Unlike histologically similar soft-tissue myoepithelial tumours, keratin expression is rare. Prompted by two recent EMC cases with diffuse keratin expression, we investigated the expression of epithelial markers in a molecularly confirmed cohort of EMC and identified two additional similar cases. METHODS AND RESULTS: Four keratin-positive EMCs occurred in one man and three women aged 46-59 years. All tumours displayed nonclassic histology with prominent stromal fibrosis, and keratin AE1/AE3 was expressed either diffusely (N = 2) or focally (N = 2). In one tumour, keratin expression was limited to the sclerotic area. All tumours coexpressed epithelial membrane antigen and two additionally expressed S100 protein or glial fibrillary acidic protein. All tumours harboured NR4A3 fusions, including TAF15::NR4A3 (N = 1) and EWSR1::NR4A3 (N = 3). Two cases were initially considered as most consistent with myoepithelial tumours based on widespread stromal fibrosis and keratin expression. DNA methylation analysis classified two tumours tested as EMCs. CONCLUSIONS: We identified a small subset of EMCs characterised by keratin expression and prominent stromal fibrosis. This histological pattern must be recognised in the differential diagnosis of myoepithelial tumours because misclassification may lead to the erroneous prediction of tumour behaviour and may alter patient management. NR4A3 genetic analysis should be considered even in the face of keratin expression and prominent stromal fibrosis.


Subject(s)
Chondrosarcoma , Myoepithelioma , Soft Tissue Neoplasms , Adult , Male , Humans , Female , Myoepithelioma/diagnosis , Myoepithelioma/genetics , Myoepithelioma/pathology , Keratins/metabolism , Calmodulin-Binding Proteins , RNA-Binding Proteins/genetics , Chondrosarcoma/diagnosis , Chondrosarcoma/genetics , Chondrosarcoma/metabolism , Soft Tissue Neoplasms/diagnosis , Soft Tissue Neoplasms/genetics , S100 Proteins , Fibrosis
13.
Int J Mol Sci ; 24(1)2023 Jan 03.
Article in English | MEDLINE | ID: mdl-36614283

ABSTRACT

Chondrosarcoma is the second most common type of bone cancer. Surgical resection is the best choice for clinical treatment. High-grade chondrosarcoma is destructive and is more possible to metastasis, which is difficult to remove using surgery. Doxorubicin (Dox) is the most commonly used chemotherapy drug in the clinical setting; however, drug resistance is a major obstacle to effective treatment. In the present study, we compared Dox-resistant SW1353 cells to their parental cells using RNA sequencing (RNA-Seq). We found that the apelin (APLN) pathway was highly activated in resistant cells. In addition, tissue array analysis also showed that APLN was higher in high-grade tissues compared to low-grade tissues. APLN is a member of the adipokine family, which is a novel secreted peptide with multifunctional and biological activities. Previously, studies have shown that inhibition of the APLN axis may have a therapeutic benefit in cancers. However, the role of APLN in chondrosarcoma is completely unclear, and no related studies have been reported. During in vitro experiments, APLN was also observed to be highly expressed and secreted in Dox-resistant cells. Once APLN was knocked down, it could effectively improve its sensitivity to Dox. We also explored possible upstream regulatory microRNAs (miRNAs) of APLN through bioinformatics tools and the results disclosed that miR-631 was the most likely regulator of APLN. Furthermore, the expression of miR-631 was lower in the resistant cells, but overexpression of miR-631 in the Dox-resistant cell lines significantly increased the Dox sensitivity. These results were also observed in another chondrosarcoma cell line, JJ012 cells. Taken together, these findings will provide rationale for the development of drug resistance biomarkers and therapeutic strategies for APLN pathway inhibitors to improve the survival of patients with chondrosarcoma.


Subject(s)
Apelin , Bone Neoplasms , Chondrosarcoma , Doxorubicin , Drug Resistance, Neoplasm , MicroRNAs , Humans , Apelin/genetics , Apelin/metabolism , Bone Neoplasms/drug therapy , Bone Neoplasms/genetics , Bone Neoplasms/metabolism , Chondrosarcoma/drug therapy , Chondrosarcoma/genetics , Chondrosarcoma/metabolism , Doxorubicin/pharmacology , Doxorubicin/therapeutic use , MicroRNAs/genetics , MicroRNAs/therapeutic use
14.
Cell Cycle ; 22(8): 939-950, 2023 04.
Article in English | MEDLINE | ID: mdl-36636023

ABSTRACT

Peroxisome proliferator-activated receptor gamma (PPARγ) is a nuclear receptor and master transcription factor of adipogenesis-related genes, and has been reported as an antitumor target for chondrosarcomas. Herein, we show that the nonsteroidal anti-inflammatory drug, zaltoprofen, induces the expression of PPARγ at the mRNA and protein levels, following the induction of PPARγ-activating factors, such as Krox20, C/EBPß, and C/EBPα, in human extraskeletal chondrosarcoma H-EMC-SS cells. Upregulation of the cell cycle checkpoint proteins, p21, p27, and p53, was observed upon treatment of H-EMC-SS cells with zaltoprofen, which probably resulted in the inhibition of proliferation of these cells observed in vitro. Zaltoprofen treatment inhibited tumor growth, induced tumor cell apoptosis, and was well tolerated in a mouse model of extraskeletal myxoid chondrosarcoma. Our results provide mechanistic insights into the therapeutic effect of zaltoprofen that should promote further studies on the rational use of this drug for the effective treatment of sarcomas.


Subject(s)
Chondrosarcoma , PPAR gamma , Animals , Humans , Mice , Anti-Inflammatory Agents , Cell Cycle Proteins/metabolism , Chondrosarcoma/drug therapy , Chondrosarcoma/metabolism , Chondrosarcoma/pathology , PPAR gamma/metabolism , Tumor Suppressor Protein p53/genetics , Cyclin-Dependent Kinase Inhibitor p27 , Cyclin-Dependent Kinase Inhibitor p21
15.
Elife ; 122023 01 09.
Article in English | MEDLINE | ID: mdl-36622753

ABSTRACT

Chondrosarcomas are primary cancers of cartilaginous tissue and capable of alteration to highly aggressive, metastatic, and treatment-refractory states, leading to a poor prognosis with a five-year survival rate at 11 months for dedifferentiated subtype. At present, the surgical resection of chondrosarcoma is the only effective treatment, and no other treatment options including targeted therapies, conventional chemotherapies, or immunotherapies are available for these patients. Here, we identify a signal pathway way involving EZH2/SULF1/cMET axis that contributes to malignancy of chondrosarcoma and provides a potential therapeutic option for the disease. A non-biased chromatin immunoprecipitation sequence, cDNA microarray analysis, and validation of chondrosarcoma cell lines identified sulfatase 1 (SULF1) as the top EZH2-targeted gene to regulate chondrosarcoma progression. Overexpressed EZH2 resulted in downregulation of SULF1 in chondrosarcoma cell lines, which in turn activated cMET pathway. Pharmaceutical inhibition of cMET or genetically silenced cMET pathway significantly retards the chondrosarcoma growth and extends mice survival. The regulation of EZH2/SULF1/cMET axis were further validated in patient samples with chondrosarcoma. The results not only established a signal pathway promoting malignancy of chondrosarcoma but also provided a therapeutic potential for further development of effective target therapy to treat chondrosarcoma.


Subject(s)
Bone Neoplasms , Chondrosarcoma , Enhancer of Zeste Homolog 2 Protein , Sulfotransferases , Animals , Mice , Bone Neoplasms/genetics , Bone Neoplasms/metabolism , Bone Neoplasms/pathology , Cartilage/pathology , Chondrosarcoma/genetics , Chondrosarcoma/metabolism , Chondrosarcoma/pathology , Receptor Protein-Tyrosine Kinases/metabolism , Signal Transduction , Sulfotransferases/genetics , Humans , Enhancer of Zeste Homolog 2 Protein/genetics
16.
Hum Cell ; 36(1): 446-455, 2023 Jan.
Article in English | MEDLINE | ID: mdl-36316541

ABSTRACT

Extraskeletal myxoid chondrosarcoma (EMC) is a malignant mesenchymal neoplasm of uncertain differentiation as classified by the WHO Classification of Tumours 2020. Although often associated with pronlonged survival, EMC has high rates of distant recurrences and disease-associated death. EMCs are translocation sarcomas and harbor in > 90% of the cases an NR4A3 rearrangement. The molecular consequences of the NR4A3 gene fusions are not yet fully elucidated as well-characterized ex vivo cell models for EMC are lacking. Patient-derived ex vivo models are important and essential tools for investigating disease mechanisms associated with diseases that are rare, that exhibit poor prognosis and for the identification of potential novel treatment options. We established two novel EMC ex vivo models (USZ20-EMC1 and USZ22-EMC2) for functional testing and research purposes. USZ20-EMC1 and USZ22-EMC2 were established and maintained as sarco-sphere cell models for several months in culture. The cells were molecularly characterized using DNA sequencing and methylation profiling. Both cell models represent their native tumor tissue as confirmed by histomorphology and their molecular profiles, suggesting that native tumor cell function can be recapitulated in the ex vivo models. Using a functional screening approach, novel anti-cancer drug sensitivities including potential synergistic combinations were identified. In conclusion, two novel EMC ex vivo cell models (USZ20-EMC1 and USZ22-EMC2) were successfully established and characterized from native tumor tissues. Both cell models will be useful tools for further investigating disease mechanisms and for answering basic and translational research questions.


Subject(s)
Chondrosarcoma , Neoplasms, Connective and Soft Tissue , Sarcoma , Soft Tissue Neoplasms , Humans , Chondrosarcoma/genetics , Chondrosarcoma/metabolism , Chondrosarcoma/pathology , Neoplasms, Connective and Soft Tissue/genetics , Soft Tissue Neoplasms/genetics
17.
Cancer Treat Res Commun ; 33: 100644, 2022.
Article in English | MEDLINE | ID: mdl-36368296

ABSTRACT

BACKGROUND: Previous studies showed that proline-rich polypeptide (PRP-1) is a ligand for innate immunity toll-like receptors (TLR), and an inhibitor of the mammalian target of rapamycin complex 1 (mTORC1) which induces the death of chondrosarcoma cancer stem cells (CSC). The aim of this study was to investigate the effect of PRP-1 on the regulation of unfolded protein response (UPR) in human chondrosarcoma cells. MATERIALS AND METHODS: Lysates were prepared from a monolayer (bulk or ALDHhigh population), or spheroids chondrosarcoma cell cultures and treated with PRP-1 or control, followed by protein levels quantification by western blotting and mRNA expression by RT-qPCR of protein-RNA-like endoplasmic reticulum kinase (PERK), eukaryotic translation initiation factor 2α (eIF2α), activating transcription factor 4 (ATF4), CCAAT-enhancer-binding protein homologous protein (CHOP), activating transcription factor 6 (ATF6), inositol-requiring enzyme 1 (IRE1α), and X-box binding protein (XBP1). RESULTS: The PRP-1 has been shown to increase the expression of PERK, eIF2α, ATF4, CHOP, ATF6, IRE1α, and XBP1, on both protein and mRNA levels. CONCLUSION: PRP-1 activated UPR branches in monolayer, spheroid, and stem cell populations of human chondrosarcoma.


Subject(s)
Bone Neoplasms , Chondrosarcoma , Toll-Like Receptors , Unfolded Protein Response , Humans , Activating Transcription Factor 4/genetics , Activating Transcription Factor 4/metabolism , Endoribonucleases/genetics , Endoribonucleases/metabolism , Ligands , Protein Serine-Threonine Kinases/genetics , Protein Serine-Threonine Kinases/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism , RNA, Messenger/pharmacology , Signal Transduction , Toll-Like Receptors/genetics , Toll-Like Receptors/metabolism , Unfolded Protein Response/genetics , Unfolded Protein Response/physiology , Chondrosarcoma/genetics , Chondrosarcoma/metabolism , Chondrosarcoma/pathology , Bone Neoplasms/genetics , Bone Neoplasms/metabolism , Bone Neoplasms/pathology
18.
Cancer Biomark ; 35(4): 429-437, 2022.
Article in English | MEDLINE | ID: mdl-36404534

ABSTRACT

INTRODUCTION: The E3 ubiquitin ligase FBXW11 exerts an oncogenic or tumor suppressive function in a cellular context-dependent manner. However, the clinical significance and biological role of FBXW11 in chondrosarcoma have not been clearly characterized. This study focuses on the expression profile, prognostic value and biological function of FBXW11 in chondrosarcoma. METHODS: FBXW11 expression was analyzed by qRT-PCR and Western blot in six cases of chondrosarcoma specimens and the matched adjacent non-tumor tissues. The expression profile and prognostic value of FBXW11 were investigated in sixty-three cases of chondrosarcoma patients. Cell viability, colony formation, migration, invasion and apoptosis assays were further detected in SW1353 chondrosarcoma cells with restored FBXW11 expression. RESULTS: Downregulation of FBXW11 was remarkably detected in human chondrosarcoma specimens compared with the corresponding non-tumor tissues and benign cartilage tumors. Downregulated FBXW11 expression significantly correlated with high-grade chondrosarcoma and poor prognosis. Furthermore, FBXW11 was identified as an independent prognostic factor for the overall survival of chondrosarcoma patients. Restored expression of FBXW11 significantly suppressed chondrosarcoma cell growth and induced apoptosis. CONCLUSIONS: These findings establish that FBXW11 was markedly downregulated and recognized as an independent prognostic factor for patients with chondrosarcoma, and restored FBXW11 expression can suppress chondrosarcoma growth and induce apoptosis, highlighting a novel biological marker and potential therapeutic target against chondrosarcoma.


Subject(s)
Bone Neoplasms , Chondrosarcoma , Humans , Prognosis , Bone Neoplasms/metabolism , Chondrosarcoma/genetics , Chondrosarcoma/metabolism , Apoptosis/genetics , Ubiquitin-Protein Ligases/genetics , Ubiquitin-Protein Ligases/metabolism , Cell Proliferation/genetics , Cell Line, Tumor , beta-Transducin Repeat-Containing Proteins
19.
Cells ; 11(21)2022 11 02.
Article in English | MEDLINE | ID: mdl-36359873

ABSTRACT

New treatments for chondrosarcoma are extremely important. Chondrosarcoma is a primary malignant bone tumor with a very unfavorable prognosis. High-grade chondrosarcoma has a high potential to metastasize to any organ in the body. Platelet-derived growth factor (PDGF) is a potent angiogenic factor that promotes tumor angiogenesis and metastasis. The adipocytokine visfatin promotes metastatic potential of chondrosarcoma; however, the role of visfatin in angiogenesis in human chondrosarcoma is unclear. We report that the levels of PDGF-C expression were positively correlated with tumor stages, significantly higher than the levels of expression in normal cartilage. Visfatin increased PDGF-C expression and endothelial progenitor cell (EPC) angiogenesis through the PI3K/Akt/mTOR signaling pathway, and dose-dependently down-regulated the synthesis of miR-1264, which targets the 3'-UTR of PDGF-C. Additionally, we discovered inhibition of visfatin or PDGF-C in chondrosarcoma tumors significantly reduced tumor angiogenesis and size. Our results indicate that visfatin inhibits miR-1264 production through the PI3K/Akt/mTOR signaling cascade, and thereby promotes PDGF-C expression and chondrosarcoma angiogenesis. Visfatin may be worth targeting in the treatment of chondrosarcoma angiogenesis.


Subject(s)
Bone Neoplasms , Chondrosarcoma , Endothelial Progenitor Cells , MicroRNAs , Humans , Endothelial Progenitor Cells/metabolism , Nicotinamide Phosphoribosyltransferase/genetics , Nicotinamide Phosphoribosyltransferase/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Vascular Endothelial Growth Factor A/metabolism , Chondrosarcoma/metabolism , Neovascularization, Pathologic/metabolism , Platelet-Derived Growth Factor/metabolism , TOR Serine-Threonine Kinases/metabolism , Bone Neoplasms/metabolism , MicroRNAs/genetics , MicroRNAs/metabolism
20.
BMC Cancer ; 22(1): 758, 2022 Jul 12.
Article in English | MEDLINE | ID: mdl-35820864

ABSTRACT

BACKGROUND: Although chondrosarcoma is the second most common primary malignant bone tumor, treatment options are limited due to its extensive resistance to a chemo- and radiation therapy. Since shikonin has shown potent anticancer activity in various types of cancer cells, it represents a promising compound for the development of a new therapeutic approach. METHODS: The dose-relationships of shikonin and its derivatives acetylshikonin and cyclopropylshikonin on two human chondrosarcoma cell lines were measured using the CellTiter-Glo®. The changes in the cell cycle were presented by flow cytometry. Protein phosphorylation and expression apoptotic markers, MAPKs and their downstream targets were analyzed using western blotting and gene expression were evaluated using RT-qPCR. RESULTS: Chondrosarcoma cells showed a dose-dependent inhibition of cell viability after treatment with shikonin and its derivatives, with the strongest effect for shikonin and IC50 values of 1.3 ± 0.2 µM. Flow cytometric measurements revealed a G2/M arrest of the cells after treatment. Protein and gene expression analysis demonstrated a dose-dependent downregulation of survivin and XIAP, and an upregulation of Noxa, γH2AX, cleaved caspase-8, -9, -3, and -PARP. Furthermore, the expression of various death receptors was modulated. As MAPK signaling pathways play a key role in tumor biology, their phosphorylation pattern and their corresponding downstream gene regulation were analyzed. Treatment with shikonin derivatives caused an inhibition of pSTAT3 and an increase of pAKT and the MAPKs pERK, pJNK, and pp38 in a dose-dependent manner. CONCLUSIONS: These data demonstrated the significant anti-tumorigenic effect of shikonin derivatives in chondrosarcoma and encourage further research.


Subject(s)
Bone Neoplasms , Chondrosarcoma , Mitogen-Activated Protein Kinases , Naphthoquinones , Receptors, Death Domain , Apoptosis/drug effects , Bone Neoplasms/drug therapy , Bone Neoplasms/metabolism , Bone Neoplasms/pathology , Cell Cycle Checkpoints/drug effects , Cell Line, Tumor , Chondrosarcoma/drug therapy , Chondrosarcoma/metabolism , Chondrosarcoma/pathology , Humans , Naphthoquinones/pharmacology , Receptors, Death Domain/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...