Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 10 de 10
Filtrer
Plus de filtres










Base de données
Gamme d'année
1.
J Exp Clin Cancer Res ; 43(1): 25, 2024 Jan 22.
Article de Anglais | MEDLINE | ID: mdl-38246990

RÉSUMÉ

BACKGROUND: Extensive local invasion of glioblastoma (GBM) cells within the central nervous system (CNS) is one factor that severely limits current treatments. The aim of this study was to uncover genes involved in the invasion process, which could also serve as therapeutic targets. For the isolation of invasive GBM cells from non-invasive cells, we used a three-dimensional organotypic co-culture system where glioma stem cell (GSC) spheres were confronted with brain organoids (BOs). Using ultra-low input RNA sequencing (ui-RNA Seq), an invasive gene signature was obtained that was exploited in a therapeutic context. METHODS: GFP-labeled tumor cells were sorted from invasive and non-invasive regions within co-cultures. Ui-RNA sequencing analysis was performed to find a gene cluster up-regulated in the invasive compartment. This gene cluster was further analyzed using the Connectivity MAP (CMap) database. This led to the identification of SKF83566, an antagonist of the D1 dopamine receptor (DRD1), as a candidate therapeutic molecule. Knockdown and overexpression experiments were performed to find molecular pathways responsible for the therapeutic effects of SKF83566. Finally, the effects of SKF83566 were validated in orthotopic xenograft models in vivo. RESULTS: Ui-RNA seq analysis of three GSC cell models (P3, BG5 and BG7) yielded a set of 27 differentially expressed genes between invasive and non-invasive cells. Using CMap analysis, SKF83566 was identified as a selective inhibitor targeting both DRD1 and DRD5. In vitro studies demonstrated that SKF83566 inhibited tumor cell proliferation, GSC sphere formation, and invasion. RNA sequencing analysis of SKF83566-treated P3, BG5, BG7, and control cell populations yielded a total of 32 differentially expressed genes, that were predicted to be regulated by c-Myc. Of these, the UHRF1 gene emerged as the most downregulated gene following treatment, and ChIP experiments revealed that c-Myc binds to its promoter region. Finally, SKF83566, or stable DRD1 knockdown, inhibited the growth of orthotopic GSC (BG5) derived xenografts in nude mice. CONCLUSIONS: DRD1 contributes to GBM invasion and progression by regulating c-Myc entry into the nucleus that affects the transcription of the UHRF1 gene. SKF83566 inhibits the transmembrane protein DRD1, and as such represents a candidate small therapeutic molecule for GBMs.


Sujet(s)
Antagonistes de la dopamine , Glioblastome , Gliome , Protéines proto-oncogènes c-myc , Animaux , Humains , Souris , Encéphale , Protéines liant les séquences stimulatrices de type CCAAT/effets des médicaments et des substances chimiques , Protéines liant les séquences stimulatrices de type CCAAT/métabolisme , Dopamine , Glioblastome/traitement médicamenteux , Glioblastome/génétique , Souris nude , Famille multigénique , Récepteur dopamine D1/antagonistes et inhibiteurs , Ubiquitin-protein ligases/effets des médicaments et des substances chimiques , Ubiquitin-protein ligases/métabolisme , Antagonistes de la dopamine/métabolisme , Antagonistes de la dopamine/pharmacologie , Protéines proto-oncogènes c-myc/effets des médicaments et des substances chimiques , Protéines proto-oncogènes c-myc/métabolisme
2.
J Transl Med ; 21(1): 147, 2023 02 24.
Article de Anglais | MEDLINE | ID: mdl-36829235

RÉSUMÉ

BACKGROUND: Valtrate, a natural compound isolated from the root of Valeriana, exhibits antitumor activity in many cancers through different mechanisms. However, its efficacy for the treatment of glioblastoma (GBM), a tumor type with a poor prognosis, has not yet been rigorously investigated. METHODS: GBM cell lines were treated with valtrate and CCK-8, colony formation and EdU assays, flow cytometry, and transwell, 3D tumor spheroid invasion and GBM-brain organoid co-culture invasion assays were performed to assess properties of proliferation, viability, apoptosis and invasion/migration. RNA sequencing analysis on valtrate-treated cells was performed to identify putative target genes underlying the antitumor activity of the drug in GBM cells. Western blot analysis, immunofluorescence and immunohistochemistry were performed to evaluate protein levels in valtrate-treated cell lines and in samples obtained from orthotopic xenografts. A specific activator of extracellular signal-regulated kinase (ERK) was used to identify the pathways mediating the effect. RESULTS: Valtrate significantly inhibited the proliferation of GBM cells in vitro by inducing mitochondrial apoptosis and suppressed invasion and migration of GBM cells by inhibiting levels of proteins associated with epithelial mesenchymal transition (EMT). RNA sequencing analysis of valtrate-treated GBM cells revealed platelet-derived growth factor receptor A (PDGFRA) as a potential target downregulated by the drug. Analysis of PDGFRA protein and downstream mediators demonstrated that valtrate inhibited PDGFRA/MEK/ERK signaling. Finally, treatment of tumor-bearing nude mice with valtrate led to decreased tumor volume (fivefold difference at day 28) and enhanced survival (day 27 vs day 36, control vs valtrate-treated) relative to controls. CONCLUSIONS: Taken together, our study demonstrated that the natural product valtrate elicits antitumor activity in GBM cells through targeting PDGFRA and thus provides a candidate therapeutic compound for the treatment of GBM.


Sujet(s)
Tumeurs du cerveau , Glioblastome , Valeriana , Souris , Animaux , Humains , Extracellular Signal-Regulated MAP Kinases/métabolisme , Valeriana/métabolisme , Souris nude , Prolifération cellulaire , Glioblastome/anatomopathologie , Transduction du signal , Iridoïdes/pharmacologie , Iridoïdes/usage thérapeutique , Mitogen-Activated Protein Kinase Kinases/métabolisme , Mitogen-Activated Protein Kinase Kinases/pharmacologie , Mitogen-Activated Protein Kinase Kinases/usage thérapeutique , Lignée cellulaire tumorale , Tumeurs du cerveau/génétique , Mouvement cellulaire
3.
J Exp Clin Cancer Res ; 41(1): 336, 2022 Dec 06.
Article de Anglais | MEDLINE | ID: mdl-36471347

RÉSUMÉ

BACKGROUND: The tripartite motif (TRIM) family of proteins plays a key role in the developmental growth and therapeutic resistance of many tumors. However, the regulatory mechanisms and biological functions of TRIM proteins in human glioblastoma (GBM) are not yet fully understood. In this study, we focused on TRIM56, which emerged as the most differentially expressed TRIM family member with increased expression in GBM. METHODS: Western blot, real-time quantitative PCR (qRT-PCR), immunofluorescence (IF) and immunohistochemistry (IHC) were used to study the expression levels of TRIM56 and cIAP1 in GBM cell lines. Co-immunoprecipitation (co-IP) was used to explore the specific binding between target proteins and TRIM56. A xenograft animal model was used to verify the tumor promoting effect of TRIM56 on glioma in vivo. RESULTS: We observed elevated expression of TRIM56 in malignant gliomas and revealed that TRIM56 promoted glioma progression in vitro and in a GBM xenograft model in nude mice. Analysis of the Human Ubiquitin Array and co-IPs showed that cIAP1 is a protein downstream of TRIM56. TRIM56 deubiquitinated cIAP1, mainly through the zinc finger domain (amino acids 21-205) of TRIM56, thereby reducing the degradation of cIAP1 and thus increasing its expression. TRIM56 also showed prognostic significance in overall survival of glioma patients. CONCLUSIONS: TRIM56-regulated post-translational modifications may contribute to glioma development through stabilization of cIAP1. Furthermore, TRIM56 may serve as a novel prognostic indicator and therapeutic molecular target for GBM.


Sujet(s)
Tumeurs du cerveau , Glioblastome , Protéines à motif tripartite , Ubiquitin-protein ligases , Animaux , Humains , Souris , Tumeurs du cerveau/génétique , Tumeurs du cerveau/anatomopathologie , Lignée cellulaire tumorale , Prolifération cellulaire , Régulation de l'expression des gènes tumoraux , Glioblastome/génétique , Glioblastome/anatomopathologie , Protéines IAP/métabolisme , Souris nude , Protéines à motif tripartite/génétique , Protéines à motif tripartite/métabolisme , Ubiquitin-protein ligases/génétique , Ubiquitin-protein ligases/métabolisme
4.
Front Pharmacol ; 13: 855626, 2022.
Article de Anglais | MEDLINE | ID: mdl-35656311

RÉSUMÉ

Resibufogenin (RB) is a major active ingredient in the traditional Chinese medicine Chansu and has garnered considerable attention for its efficacy in the treatment of cancer. However, the anticancer effects and underlying mechanisms of RB on glioblastoma (GBM) remain unknown. Here, we found that RB induced G2/M phase arrest and inhibited invasion in a primary GBM cell line, P3#GBM, and two GBM cell lines, U251 and A172. Subsequently, we demonstrated that RB-induced G2/M phase arrest occurred through downregulation of CDC25C and upregulation of p21, which was caused by activation of the MAPK/ERK pathway, and that RB inhibited GBM invasion by elevating intercellular Ca2+ to suppress the Src/FAK/Paxillin focal adhesion pathway. Intriguingly, we confirmed that upon RB binding to ATP1A1, Na+-K+-ATPase was activated as a receptor and then triggered the intracellular MAPK/ERK pathway and Ca2+-mediated Src/FAK/Paxillin focal adhesion pathway, which led to G2/M phase arrest and inhibited the invasion of GBM cells. Taken together, our findings reveal the antitumor mechanism of RB by targeting the ATP1A1 signaling cascade and two key signaling pathways and highlight the potential of RB as a new class of promising anticancer agents.

5.
Cell Cycle ; 21(17): 1842-1855, 2022 09.
Article de Anglais | MEDLINE | ID: mdl-35532155

RÉSUMÉ

Nucleolar and spindle associated protein 1 (NUSAP1), an indispensable mitotic regulator, has been reported to be involved in the development, progression, and metastasis of several types of cancer. Here, we investigated the expression and biological function of NUSAP1 in human glioblastoma (GBM), an aggressive brain tumor type with largely ineffective treatment options. Analysis of the molecular data in CGGA, TCGA and Rembrandt datasets demonstrated that NUSAP1 was significantly upregulated in GBM relative to low grade gliomas and non-neoplastic brain tissue samples. Kaplan-Meier analysis indicated that patients with tumors showing high NUSAP1 expression exhibited significantly poorer survival in both CGGA (P = 0.002) and Rembrandt cohorts (P = 0.017). Analysis of RNA sequencing data from P3-cells with stable knockdown of NUSAP1 revealed topoisomerase 2A (TOP2A) as a possible molecule downregulated by the loss of NUSAP1. Molecular analysis of the CGGA data revealed a strong correlation between NUSAP1 and TOP2A expression in primary gliomas and recurrent gliomas samples. SiRNA knockdown of either NUSAP1 or TOP2A in U251, T98 and GBM derived patient P3 cells inhibited GBM cell proliferation and invasion, and induced cell apoptosis. Finally, stable knockdown of NUSAP1 with shRNA led to decreased tumor growth in an orthotopic xenograft model of GBM in mice. Taken together, NUSAP1 gene silencing induced apoptosis possibly through the downregulation of the candidate downstream molecule TOP2A. Interference with the expression of NUSAP1 might therefore inhibit malignant progression in GBM, and NUSAP1 might thus serve as a promising molecular target for GBM treatment.


Sujet(s)
Tumeurs du cerveau , ADN topoisomérases de type II , Glioblastome , Gliome , Protéines associées aux microtubules , Protéines liant le poly-adp-ribose , Animaux , Tumeurs du cerveau/anatomopathologie , Lignée cellulaire tumorale , Prolifération cellulaire , ADN topoisomérases de type II/génétique , Régulation négative/génétique , Régulation de l'expression des gènes tumoraux , Glioblastome/anatomopathologie , Gliome/génétique , Humains , Souris , Protéines associées aux microtubules/génétique , Protéines liant le poly-adp-ribose/génétique , Petit ARN interférent/génétique
6.
J Pharmacol Exp Ther ; 380(1): 63-75, 2022 01.
Article de Anglais | MEDLINE | ID: mdl-34750208

RÉSUMÉ

Thiabendazole (TBZ), approved by the US Food and Drug Administration (FDA) for human oral use, elicits a potential anticancer activity on cancer cells in vitro and in animal models. Here, we evaluated the efficacy of TBZ in the treatment of human glioblastoma multiforme (GBM). TBZ reduced the viability of GBM cells (P3, U251, LN229, A172, and U118MG) relative to controls in a dose- and time-dependent manner. However, normal human astrocytes (NHA) exhibited a greater IC50 than tumor cell lines and were thus more resistant to its cytotoxic effects. 5-Ethynyl-2'-deoxyuridine (EdU)-positive cells and the number of colonies formed were decreased in TBZ-treated cells (at 150 µM, P < 0.05 and at 150 µM, P < 0.001, respectively). This decrease in proliferation was associated with a G2/M arrest as assessed with flow cytometry, and the downregulation of G2/M check point proteins. In addition, TBZ suppressed GBM cell invasion. Analysis of RNA sequencing data comparing TBZ-treated cells with controls yielded a group of differentially expressed genes, the functions of which were associated with the cell cycle and DNA replication. The most significantly downregulated gene in TBZ-treated cells was mini-chromosome maintenance protein 2 (MCM2). SiRNA knockdown of MCM2 inhibited proliferation, causing a G2/M arrest in GBM cell lines and suppressed invasion. Taken together, our results demonstrated that TBZ inhibited proliferation and invasion in GBM cells through targeting of MCM2. SIGNIFICANCE STATEMENT: TBZ inhibits the proliferation and invasion of glioblastoma cells by downregulating the expression of MCM2. These results support the repurposing of TBZ as a possible therapeutic drug in the treatment of GBM.


Sujet(s)
Anthelminthiques/usage thérapeutique , Antinéoplasiques/pharmacologie , Tumeurs du cerveau/traitement médicamenteux , Prolifération cellulaire/effets des médicaments et des substances chimiques , Glioblastome/traitement médicamenteux , Composant-2 du complexe de maintenance des minichromosomes/métabolisme , Tiabendazol/pharmacologie , Animaux , Anthelminthiques/pharmacologie , Antinéoplasiques/usage thérapeutique , Tumeurs du cerveau/métabolisme , Lignée cellulaire , Lignée cellulaire tumorale , Mouvement cellulaire/effets des médicaments et des substances chimiques , Cellules cultivées , Repositionnement des médicaments , Glioblastome/métabolisme , Humains , Souris , Souris nude , Tiabendazol/usage thérapeutique
7.
Oncogene ; 40(8): 1425-1439, 2021 02.
Article de Anglais | MEDLINE | ID: mdl-33420375

RÉSUMÉ

Dysregulated iron metabolism is a hallmark of many cancers, including glioblastoma (GBM). However, its role in tumor progression remains unclear. Herein, we identified coatomer protein complex subunit zeta 1 (COPZ1) as a therapeutic target candidate which significantly dysregulated iron metabolism in GBM cells. Overexpression of COPZ1 was associated with increasing tumor grade and poor prognosis in glioma patients based on analysis of expression data from the publicly available database The Cancer Genome Atlas (P < 0.001). Protein levels of COPZ1 were significantly increased in GBM compared to non-neoplastic brain tissue samples in immunohistochemistry and western blot analysis. SiRNA knockdown of COPZ1 suppressed proliferation of U87MG, U251 and P3#GBM in vitro. Stable expression of a COPZ1 shRNA construct in U87MG inhibited tumor growth in vivo by ~60% relative to controls at day 21 after implantation (P < 0.001). Kaplan-Meier analysis of the survival data demonstrated that the overall survival of tumor bearing animals increased from 20.8 days (control) to 27.8 days (knockdown, P < 0.05). COPZ1 knockdown also led to the increase in nuclear receptor coactivator 4 (NCOA4), resulting in the degradation of ferritin, and a subsequent increase in the intracellular levels of ferrous iron and ultimately ferroptosis. These data demonstrate that COPZ1 is a critical mediator in iron metabolism. The COPZ1/NCOA4/FTH1 axis is therefore a novel therapeutic target for the treatment of human GBM.


Sujet(s)
Protéine du coatomère/génétique , Ferritines/génétique , Glioblastome/génétique , Coactivateurs de récepteurs nucléaires/génétique , Oxidoreductases/génétique , Apoptose/génétique , Autophagie/génétique , Lignée cellulaire tumorale , Prolifération cellulaire/génétique , Femelle , Ferroptose/génétique , Humains , Estimation de Kaplan-Meier , Mâle , Adulte d'âge moyen , Petit ARN interférent/génétique
8.
J Biosci ; 452020.
Article de Anglais | MEDLINE | ID: mdl-32345773

RÉSUMÉ

Mounting evidence has reported that microRNAs (miRNAs) play irreplaceable roles in the development of keloid fibrosis. miR-4417 has been reported to contribute to nickel chloride-promoted lung epithelial cell fibrogenesis and tumorigenesis. However, whether miR-4417 is involved in keloid fibrogenesis as well as its underlying mechanisms remain largely elusive. In this study, the expression levels of miR-4417 and CyclinD1 in keloid tissues and fibroblasts were examined by qRT-PCR. Cell proliferation was determined by CCK assay. Western blot and flow cytometry were performed to evaluate cell apoptosis. Cell migration and invasion were measured by Transwell assay. Luciferase reporter assay was used to confirm the relationship between miR4417 and CyclinD1. As a result, we found that miR-4417 was significantly down-regulated in keloid tissues and fibroblasts. miR-4417 up-regulation led to the suppression of proliferation, migration, and invasion, while induced cell apoptosis in keloid fibroblasts. However, miR-4417 depletion exerted an opposite effect. CyclinD1 harbored the binding sites with miR-4417. Besides, the expression of CyclinD1 was evidently decreased in keloid tissues and fibroblasts. Meanwhile, miR-4417 was negatively correlated with CyclinD1 in keloid tissue. The effect of CyclinD1 knockdown on keloid fibroblasts was similar to that of miR-4417 overexpression. Furthermore, the elevated of CyclinD1 expression rescued the effect of miR-4417 up-regulation on keloid fibroblasts. miR-4417/CyclinD1 axis was required for cell proliferation, apoptosis, migration, and invasion in keloid fibroblasts. In conclusion, miR-4417 and CyclinD1 may be potential therapeutic targets for the treatment of keloid.


Sujet(s)
Cycline D1/métabolisme , Fibroblastes/métabolisme , Fibroblastes/anatomopathologie , Chéloïde/métabolisme , Chéloïde/anatomopathologie , microARN/métabolisme , Apoptose , Mouvement cellulaire , Prolifération cellulaire , Cellules cultivées , Cycline D1/génétique , Régulation négative , Fibrose , Régulation de l'expression des gènes , Humains , Chéloïde/génétique
9.
J Cell Mol Med ; 24(5): 2847-2856, 2020 03.
Article de Anglais | MEDLINE | ID: mdl-31778016

RÉSUMÉ

Proteolipid protein 2 (PLP2) is an integral ion channel membrane protein of the endoplasmic reticulum. The protein has been shown to be highly expressed in many cancer types, but its importance in glioma progression is poorly understood. Using publicly available datasets (Rembrandt, TCGA and CGGA), we found that the expression of PLP2 was significantly higher in high-grade gliomas than in low-grade gliomas. We confirmed these results at the protein level through IHC staining of high-grade (n = 56) and low-grade glioma biopsies (n = 16). Kaplan-Meier analysis demonstrated that increased PLP2 expression was associated with poorer patient survival. In functional experiments, siRNA and shRNA PLP2 knockdown induced ER stress and increased apoptosis and autophagy in U87 and U251 glioma cell lines. Inhibition of autophagy with chloroquine augmented apoptotic cell death in U87- and U251-siPLP2 cells. Finally, intracranial xenografts derived from U87- and U251-shPLP2 cells revealed that loss of PLP2 reduced glioma growth in vivo. Our results therefore indicate that increased PLP2 expression promotes GBM growth and that PLP2 represents a potential future therapeutic target.


Sujet(s)
Apoptose/génétique , Autophagie/génétique , Tumeurs du cerveau/génétique , Stress du réticulum endoplasmique/génétique , Régulation de l'expression des gènes tumoraux , Glioblastome/génétique , Glioblastome/anatomopathologie , Protéines à domaine MARVEL/génétique , Protéolipides/génétique , Animaux , Tumeurs du cerveau/anatomopathologie , Tumeurs du cerveau/ultrastructure , Lignée cellulaire tumorale , Prolifération cellulaire/génétique , Régulation négative/génétique , Techniques de knock-down de gènes , Glioblastome/ultrastructure , Humains , Protéines à domaine MARVEL/métabolisme , Mâle , Souris , Pronostic , Protéolipides/métabolisme , Facteur de transcription CHOP/métabolisme
10.
Biomed Pharmacother ; 75: 51-7, 2015 Oct.
Article de Anglais | MEDLINE | ID: mdl-26463631

RÉSUMÉ

miR-425 is a potential tumor suppressor in cancer, but its role in melanoma is still unknown. We aim to investigate miR-425 expression in melanoma tissues and cell lines. Next, cell proliferation, cell cycle, apoptosis and metastasis will be studied using lentivirus-mediated gain-of-function studies. The predicted results are stable miR-425 inhibits cell proliferation and metastasis and induced cell apoptosis. It is predicted that IGF-1 is a potential target gene of miR-495 by bioinformatics analysis. Then luciferase assay analysis identifies IGF-1 as a new direct target gene of miR-425 and miR-425 inhibits melanoma cancer progression via IGF-1. Collectively, our findings suggested that miR-425 may function as a tumor suppressor in melanoma by targeting IGF-1.


Sujet(s)
Facteur de croissance IGF-I/métabolisme , Mélanome/enzymologie , Mélanome/génétique , microARN/génétique , Phosphatidylinositol 3-kinase/métabolisme , Protéines proto-oncogènes c-akt/métabolisme , Tumeurs cutanées/enzymologie , Tumeurs cutanées/génétique , Régions 3' non traduites , Apoptose , Sites de fixation , Lignée cellulaire tumorale , Mouvement cellulaire , Prolifération cellulaire , Régulation négative , Régulation de l'expression des gènes tumoraux , Gènes suppresseurs de tumeur , Humains , Facteur de croissance IGF-I/génétique , Mélanome/secondaire , microARN/métabolisme , Interférence par ARN , Transduction du signal , Tumeurs cutanées/anatomopathologie , Facteurs temps , Transfection
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE