Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 20 de 5.281
Filtrer
1.
Front Immunol ; 15: 1415736, 2024.
Article de Anglais | MEDLINE | ID: mdl-38962012

RÉSUMÉ

Background: Neuroblastoma (NB), characterized by its marked heterogeneity, is the most common extracranial solid tumor in children. The status and functionality of mitochondria are crucial in regulating NB cell behavior. While the significance of mitochondria-related genes (MRGs) in NB is still missing in key knowledge. Materials and methods: This study leverages consensus clustering and machine learning algorithms to construct and validate an MRGs-related signature in NB. Single-cell data analysis and experimental validation were employed to characterize the pivotal role of FEN1 within NB cells. Results: MRGs facilitated the classification of NB patients into 2 distinct clusters with considerable differences. The constructed MRGs-related signature and its quantitative indicators, mtScore and mtRisk, effectively characterize the MRGs-related patient clusters. Notably, the MRGs-related signature outperformed MYCN in predicting NB patient prognosis and was adept at representing the tumor microenvironment (TME), tumor cell stemness, and sensitivity to the chemotherapeutic agents Cisplatin, Topotecan, and Irinotecan. FEN1, identified as the most contributory gene within the MRGs-related signature, was found to play a crucial role in the communication between NB cells and the TME, and in the developmental trajectory of NB cells. Experimental validations confirmed FEN1's significant influence on NB cell proliferation, apoptosis, cell cycle, and invasiveness. Conclusion: The MRGs-related signature developed in this study offers a novel predictive tool for assessing NB patient prognosis, immune infiltration, stemness, and chemotherapeutic sensitivity. Our findings unveil the critical function of FEN1 in NB, suggesting its potential as a therapeutic target.


Sujet(s)
Analyse de profil d'expression de gènes , Neuroblastome , Analyse sur cellule unique , Transcriptome , Humains , Neuroblastome/génétique , Neuroblastome/anatomopathologie , Mitochondries/génétique , Régulation de l'expression des gènes tumoraux , Microenvironnement tumoral/génétique , Lignée cellulaire tumorale , Marqueurs biologiques tumoraux/génétique , Pronostic
2.
Nat Commun ; 15(1): 5585, 2024 Jul 11.
Article de Anglais | MEDLINE | ID: mdl-38992040

RÉSUMÉ

MYCN oncogene amplification is frequently observed in aggressive childhood neuroblastoma. Using an unbiased large-scale mutagenesis screen in neuroblastoma-prone transgenic mice, we identify a single germline point mutation in the transcriptional corepressor Runx1t1, which abolishes MYCN-driven tumorigenesis. This loss-of-function mutation disrupts a highly conserved zinc finger domain within Runx1t1. Deletion of one Runx1t1 allele in an independent Runx1t1 knockout mouse model is also sufficient to prevent MYCN-driven neuroblastoma development, and reverse ganglia hyperplasia, a known pre-requisite for tumorigenesis. Silencing RUNX1T1 in human neuroblastoma cells decreases colony formation in vitro, and inhibits tumor growth in vivo. Moreover, RUNX1T1 knockdown inhibits the viability of PAX3-FOXO1 fusion-driven rhabdomyosarcoma and MYC-driven small cell lung cancer cells. Despite the role of Runx1t1 in MYCN-driven tumorigenesis neither gene directly regulates the other. We show RUNX1T1 forms part of a transcriptional LSD1-CoREST3-HDAC repressive complex recruited by HAND2 to enhancer regions to regulate chromatin accessibility and cell-fate pathway genes.


Sujet(s)
Carcinogenèse , Protéine du proto-oncogène N-Myc , Neuroblastome , Neuroblastome/génétique , Neuroblastome/métabolisme , Neuroblastome/anatomopathologie , Animaux , Protéine du proto-oncogène N-Myc/génétique , Protéine du proto-oncogène N-Myc/métabolisme , Humains , Souris , Carcinogenèse/génétique , Lignée cellulaire tumorale , Régulation de l'expression des gènes tumoraux , Souris transgéniques , Souris knockout , Facteurs de transcription/métabolisme , Facteurs de transcription/génétique , Histone Demethylases/métabolisme , Histone Demethylases/génétique , Protéines corépressives/métabolisme , Protéines corépressives/génétique
3.
Int J Mol Sci ; 25(13)2024 Jul 06.
Article de Anglais | MEDLINE | ID: mdl-39000543

RÉSUMÉ

Human individual differences in brain cytochrome P450 (CYP) metabolism, including induction, inhibition, and genetic variation, may influence brain sensitivity to neurotoxins and thus participate in the onset of neurodegenerative diseases. The aim of this study was to explore the modulation of CYPs in neuronal cells. The experimental approach was focused on differentiating human neuroblastoma SH-SY5Y cells into a phenotype resembling mature dopamine neurons and investigating the effects of specific CYP isoform induction. The results demonstrated that the differentiation protocols using retinoic acid followed by phorbol esters or brain-derived neurotrophic factor successfully generated SH-SY5Y cells with morphological neuronal characteristics and increased neuronal markers (NeuN, synaptophysin, ß-tubulin III, and MAO-B). qRT-PCR and Western blot analysis showed that expression of the CYP 1A1, 3A4, 2D6, and 2E1 isoforms was detectable in undifferentiated cells, with subsequent increases in CYP 2E1, 2D6, and 1A1 following differentiation. Further increases in the 1A1, 2D6, and 2E1 isoforms following ß-naphthoflavone treatment and 1A1 and 2D6 isoforms following ethanol treatment were evident. These results demonstrate that CYP isoforms can be modulated in SH-SY5Y cells and suggest their potential as an experimental model to investigate the role of CYPs in neuronal processes involved in the development of neurodegenerative diseases.


Sujet(s)
Différenciation cellulaire , Cytochrome P-450 enzyme system , Maladies neurodégénératives , Humains , Cytochrome P-450 enzyme system/métabolisme , Cytochrome P-450 enzyme system/génétique , Lignée cellulaire tumorale , Maladies neurodégénératives/métabolisme , Maladies neurodégénératives/anatomopathologie , Trétinoïne/pharmacologie , Trétinoïne/métabolisme , Facteur neurotrophique dérivé du cerveau/métabolisme , Facteur neurotrophique dérivé du cerveau/génétique , Neuroblastome/métabolisme , Neuroblastome/anatomopathologie , Neuroblastome/génétique , Isoenzymes/métabolisme , Isoenzymes/génétique , Neurones dopaminergiques/métabolisme , Neurones/métabolisme
4.
Int J Mol Sci ; 25(11)2024 May 23.
Article de Anglais | MEDLINE | ID: mdl-38891878

RÉSUMÉ

Neuroblastoma is the most common malignant extracranial solid tumor of childhood. Recent studies involving the application of advanced high-throughput "omics" techniques have revealed numerous genomic alterations, including aberrant coding-gene transcript levels and dysfunctional pathways, that drive the onset, growth, progression, and treatment resistance of neuroblastoma. Research conducted in the past decade has shown that long non-coding RNAs, once thought to be transcriptomic noise, play key roles in cancer development. With the recent and continuing increase in the amount of evidence for the underlying roles of long non-coding RNAs in neuroblastoma, the potential clinical implications of these RNAs cannot be ignored. In this review, we discuss their biological mechanisms of action in the context of the central driving mechanisms of neuroblastoma, focusing on potential contributions to the diagnosis, prognosis, and treatment of this disease. We also aim to provide a clear, integrated picture of future research opportunities.


Sujet(s)
Marqueurs biologiques tumoraux , Régulation de l'expression des gènes tumoraux , Neuroblastome , ARN long non codant , Humains , Neuroblastome/génétique , Neuroblastome/thérapie , Neuroblastome/métabolisme , Neuroblastome/anatomopathologie , ARN long non codant/génétique , ARN long non codant/métabolisme , Marqueurs biologiques tumoraux/génétique , Animaux , Pronostic , Thérapie moléculaire ciblée/méthodes
5.
Beijing Da Xue Xue Bao Yi Xue Ban ; 56(3): 495-504, 2024 Jun 18.
Article de Chinois | MEDLINE | ID: mdl-38864136

RÉSUMÉ

OBJECTIVE: To investigate the function and underlying mechanism of cysteine and glycine-rich protein 2 (CSRP2) in neuroblastoma (NB). METHODS: The correlation between the expression level of CSRP2 mRNA and the prognosis of NB children in NB clinical samples was analyzed in R2 Genomics Analysis and Visualization Platform. The small interfering RNA (siRNA) targeting CSRP2 or CSRP2 plasmid were transfected to NB cell lines SK-N-BE(2) and SH-SY5Y. Cell proliferation was observed by crystal violet staining and real-time cellular analysis. The ability of colony formation of NB cells was observed by colony-forming unit assay. Immunofluorescence assay was used to detect the expression of the proliferation marker Ki-67. Flow cytometry analysis for cell cycle proportion was used with cells stained by propidium iodide (PI). Annexin V/7AAD was used to stain cells and analyze the percentage of cell apoptosis. The ability of cell migration was determined by cell wound-healing assay. The level of protein and mRNA expression of CSRP2 in NB primary tumor and NB cell lines were detected by Western blot and quantitative real-time PCR (RT-qPCR). RESULTS: By analyzing the NB clinical sample databases, it was found that the expression levels of CSRP2 in high-risk NB with 3/4 stages in international neuroblastoma staging system (INSS) were significantly higher than that in low-risk NB with 1/2 INSS stages. The NB patients with high expression levels of CSRP2 were shown lower overall survival rate than those with low expression levels of CSRP2. We detected the protein levels of CSRP2 in the NB samples by Western blot, and found that the protein level of CSRP2 in 3/4 INSS stages was significantly higher than that in 1/2 INSS stages. Knockdown of CSRP2 inhibited cell viability and proliferation of NB cells. Overexpression of CSRP2 increased the proliferation of NB cells. Flow cytometry showed that the proportion of sub-G1, G0/G1 and S phase cells and Annexin V positive cells were increased after CSRP2 deficiency. In the cell wound-healing assay, the healing rate of NB cells was significantly attenuated after knockdown of CSRP2. Further mechanism studies showed that the proportion of the proliferation marker Ki-67 and the phosphorylation levels of extracellular signal-regulated kinases 1/2 (ERK1/2) were significantly decreased after CSRP2 knockdown. CONCLUSION: CSRP2 is highly expressed in high-risk NB with 3/4 INSS stages, and the expression levels of CSRP2 are negatively correlated with the overall survival of NB patients. CSRP2 significantly increased the proliferation and cell migration of NB cells and inhibited cell apoptosis via the activation of ERK1/2. All these results indicate that CSRP2 promotes the progression of NB by activating ERK1/2, and this study will provide a potential target for high-risk NB therapy.


Sujet(s)
Apoptose , Mouvement cellulaire , Prolifération cellulaire , Neuroblastome , Humains , Neuroblastome/métabolisme , Neuroblastome/anatomopathologie , Neuroblastome/génétique , Lignée cellulaire tumorale , Petit ARN interférent/génétique , ARN messager/génétique , ARN messager/métabolisme , Pronostic , Cycle cellulaire , Évolution de la maladie , Antigène KI-67/métabolisme , Facteurs d'épissage riches en sérine-arginine/métabolisme , Facteurs d'épissage riches en sérine-arginine/génétique
6.
Lancet Oncol ; 25(7): 922-932, 2024 Jul.
Article de Anglais | MEDLINE | ID: mdl-38936379

RÉSUMÉ

BACKGROUND: Neuroblastoma is the most common extracranial solid tumour in children. Relapsed or refractory neuroblastoma is associated with a poor outcome. We assessed the combination of irinotecan-temozolomide and dasatinib-rapamycin (RIST) in patients with relapsed or refractory neuroblastoma. METHODS: The multicentre, open-label, randomised, controlled, phase 2, RIST-rNB-2011 trial recruited from 40 paediatric oncology centres in Germany and Austria. Patients aged 1-25 years with high-risk relapsed (defined as recurrence of all stage IV and MYCN amplification stages, after response to treatment) or refractory (progressive disease during primary treatment) neuroblastoma, with Lansky and Karnofsky performance status at least 50%, were assigned (1:1) to RIST (RIST group) or irinotecan-temozolomide (control group) by block randomisation, stratified by MYCN status. We compared RIST (oral rapamycin [loading 3 mg/m2 on day 1, maintenance 1 mg/m2 on days 2-4] and oral dasatinib [2 mg/kg per day] for 4 days with 3 days off, followed by intravenous irinotecan [50 mg/m2 per day] and oral temozolomide [150 mg/m2 per day] for 5 days with 2 days off; one course each of rapamycin-dasatinib and irinotecan-temozolomide for four cycles over 8 weeks, then two courses of rapamycin-dasatinib followed by one course of irinotecan-temozolomide for 12 weeks) with irinotecan-temozolomide alone (with identical dosing as experimental group). The primary endpoint of progression-free survival was analysed in all eligible patients who received at least one course of therapy. The safety population consisted of all patients who received at least one course of therapy and had at least one post-baseline safety assessment. This trial is registered at ClinicalTrials.gov, NCT01467986, and is closed to accrual. FINDINGS: Between Aug 26, 2013, and Sept 21, 2020, 129 patients were randomly assigned to the RIST group (n=63) or control group (n=66). Median age was 5·4 years (IQR 3·7-8·1). 124 patients (78 [63%] male and 46 [37%] female) were included in the efficacy analysis. At a median follow-up of 72 months (IQR 31-88), the median progression-free survival was 11 months (95% CI 7-17) in the RIST group and 5 months (2-8) in the control group (hazard ratio 0·62, one-sided 90% CI 0·81; p=0·019). Median progression-free survival in patients with amplified MYCN (n=48) was 6 months (95% CI 4-24) in the RIST group versus 2 months (2-5) in the control group (HR 0·45 [95% CI 0·24-0·84], p=0·012); median progression-free survival in patients without amplified MYCN (n=76) was 14 months (95% CI 9-7) in the RIST group versus 8 months (4-15) in the control group (HR 0·84 [95% CI 0·51-1·38], p=0·49). The most common grade 3 or worse adverse events were neutropenia (54 [81%] of 67 patients given RIST vs 49 [82%] of 60 patients given control), thrombocytopenia (45 [67%] vs 41 [68%]), and anaemia (39 [58%] vs 38 [63%]). Nine serious treatment-related adverse events were reported (five patients given control and four patients given RIST). There were no treatment-related deaths in the control group and one in the RIST group (multiorgan failure). INTERPRETATION: RIST-rNB-2011 demonstrated that targeting of MYCN-amplified relapsed or refractory neuroblastoma with a pathway-directed metronomic combination of a multkinase inhibitor and an mTOR inhibitor can improve progression-free survival and overall survival. This exclusive efficacy in MYCN-amplified, relapsed neuroblastoma warrants further investigation in the first-line setting. FUNDING: Deutsche Krebshilfe.


Sujet(s)
Protocoles de polychimiothérapie antinéoplasique , Dasatinib , Irinotécan , Récidive tumorale locale , Neuroblastome , Sirolimus , Témozolomide , Humains , Témozolomide/administration et posologie , Témozolomide/usage thérapeutique , Irinotécan/administration et posologie , Protocoles de polychimiothérapie antinéoplasique/usage thérapeutique , Protocoles de polychimiothérapie antinéoplasique/effets indésirables , Mâle , Femelle , Neuroblastome/traitement médicamenteux , Neuroblastome/mortalité , Neuroblastome/anatomopathologie , Neuroblastome/génétique , Enfant d'âge préscolaire , Enfant , Dasatinib/administration et posologie , Dasatinib/usage thérapeutique , Dasatinib/effets indésirables , Adolescent , Récidive tumorale locale/traitement médicamenteux , Récidive tumorale locale/anatomopathologie , Nourrisson , Adulte , Sirolimus/administration et posologie , Sirolimus/usage thérapeutique , Jeune adulte , Allemagne , Résistance aux médicaments antinéoplasiques , Survie sans progression
7.
Cell Death Dis ; 15(6): 428, 2024 Jun 18.
Article de Anglais | MEDLINE | ID: mdl-38890285

RÉSUMÉ

Neuroblastoma (NB) is a highly aggressive pediatric cancer that originates from immature nerve cells, presenting significant treatment challenges due to therapy resistance. Despite intensive treatment, approximately 50% of high-risk NB cases exhibit therapy resistance or experience relapse, resulting in poor outcomes often associated with tumor immune evasion. B7-H3 is an immune checkpoint protein known to inhibit immune responses. MicroRNAs (miRNAs) are small non-coding RNAs involved in post-transcriptional gene regulation. Our study aims to explore the impact of miRNAs on B7-H3 regulation, the anti-tumor immune response, and tumorigenicity in NB. Analysis of NB patients and patient-derived xenograft tumors revealed a correlation between higher B7-H3 expression and poorer patient survival. Notably, deceased patients exhibited a depletion of miR-29 family members (miR-29a, miR-29b, and miR-29c), which displayed an inverse association with B7-H3 expression in NB patients. Overexpression and knockdown experiments demonstrated that these miRNAs degrade B7-H3 mRNA, resulting in enhanced NK cell activation and cytotoxicity. In vivo, experiments provided further evidence that miR-29 family members reduce tumorigenicity, macrophage infiltration, and microvessel density, promote infiltration and activation of NK cells, and induce tumor cell apoptosis. These findings offer a rationale for developing more effective combination treatments that leverage miRNAs to target B7-H3 in NB patients.


Sujet(s)
Antigènes B7 , Cellules tueuses naturelles , microARN , Neuroblastome , microARN/métabolisme , microARN/génétique , Humains , Antigènes B7/métabolisme , Antigènes B7/génétique , Neuroblastome/génétique , Neuroblastome/immunologie , Neuroblastome/anatomopathologie , Cellules tueuses naturelles/immunologie , Cellules tueuses naturelles/métabolisme , Animaux , Souris , Lignée cellulaire tumorale , Régulation de l'expression des gènes tumoraux , Souris nude , Femelle , Mâle , Activation des lymphocytes
8.
Biomolecules ; 14(6)2024 May 31.
Article de Anglais | MEDLINE | ID: mdl-38927051

RÉSUMÉ

Manganese (Mn) is an essential heavy metal in the human body, while excess Mn leads to neurotoxicity, as observed in this study, where 100 µM of Mn was administered to the human neuroblastoma (SH-SY5Y) cell model of dopaminergic neurons in neurodegenerative diseases. We quantitated pathway and gene changes in homeostatic cell-based adaptations to Mn exposure. Utilizing the Gene Expression Omnibus, we accessed the GSE70845 dataset as a microarray of SH-SY5Y cells published by Gandhi et al. (2018) and applied statistical significance cutoffs at p < 0.05. We report 74 pathway and 10 gene changes with statistical significance. ReactomeGSA analyses demonstrated upregulation of histones (5 out of 10 induced genes) and histone deacetylases as a neuroprotective response to remodel/mitigate Mn-induced DNA/chromatin damage. Neurodegenerative-associated pathway changes occurred. NF-κB signaled protective responses via Sirtuin-1 to reduce neuroinflammation. Critically, Mn activated three pathways implicating deficits in purine metabolism. Therefore, we validated that urate, a purine and antioxidant, mitigated Mn-losses of viability in SH-SY5Y cells. We discuss Mn as a hypoxia mimetic and trans-activator of HIF-1α, the central trans-activator of vascular hypoxic mitochondrial dysfunction. Mn induced a 3-fold increase in mRNA levels for antioxidant metallothionein-III, which was induced 100-fold by hypoxia mimetics deferoxamine and zinc.


Sujet(s)
Manganèse , Neuroblastome , Humains , Manganèse/toxicité , Neuroblastome/métabolisme , Neuroblastome/anatomopathologie , Neuroblastome/génétique , Lignée cellulaire tumorale , Survie cellulaire/effets des médicaments et des substances chimiques , Neuroprotecteurs/pharmacologie , Marqueurs biologiques/métabolisme
10.
Genome Biol ; 25(1): 161, 2024 06 19.
Article de Anglais | MEDLINE | ID: mdl-38898465

RÉSUMÉ

BACKGROUND: Neuroblastoma is a common pediatric cancer, where preclinical studies suggest that a mesenchymal-like gene expression program contributes to chemotherapy resistance. However, clinical outcomes remain poor, implying we need a better understanding of the relationship between patient tumor heterogeneity and preclinical models. RESULTS: Here, we generate single-cell RNA-seq maps of neuroblastoma cell lines, patient-derived xenograft models (PDX), and a genetically engineered mouse model (GEMM). We develop an unsupervised machine learning approach ("automatic consensus nonnegative matrix factorization" (acNMF)) to compare the gene expression programs found in preclinical models to a large cohort of patient tumors. We confirm a weakly expressed, mesenchymal-like program in otherwise adrenergic cancer cells in some pre-treated high-risk patient tumors, but this appears distinct from the presumptive drug-resistance mesenchymal programs evident in cell lines. Surprisingly, however, this weak-mesenchymal-like program is maintained in PDX and could be chemotherapy-induced in our GEMM after only 24 h, suggesting an uncharacterized therapy-escape mechanism. CONCLUSIONS: Collectively, our findings improve the understanding of how neuroblastoma patient tumor heterogeneity is reflected in preclinical models, provides a comprehensive integrated resource, and a generalizable set of computational methodologies for the joint analysis of clinical and pre-clinical single-cell RNA-seq datasets.


Sujet(s)
Neuroblastome , RNA-Seq , Analyse sur cellule unique , Neuroblastome/génétique , Neuroblastome/anatomopathologie , Humains , Animaux , Analyse sur cellule unique/méthodes , Souris , Lignée cellulaire tumorale , Régulation de l'expression des gènes tumoraux , Résistance aux médicaments antinéoplasiques/génétique , Transcriptome , Analyse de l'expression du gène de la cellule unique
11.
Cancer Res Commun ; 4(6): 1533-1547, 2024 Jun 20.
Article de Anglais | MEDLINE | ID: mdl-38837897

RÉSUMÉ

Acquiring a telomere maintenance mechanism is a hallmark of high-risk neuroblastoma and commonly occurs by expressing telomerase (TERT). Telomerase-negative neuroblastoma has long telomeres and utilizes the telomerase-independent alternative lengthening of telomeres (ALT) mechanism. Conversely, no discernable telomere maintenance mechanism is detected in a fraction of neuroblastoma with long telomeres. Here, we show, unlike most cancers, DNA of the TERT promoter is broadly hypomethylated in neuroblastoma. In telomerase-positive neuroblastoma cells, the hypomethylated DNA promoter is approximately 1.5 kb. The TERT locus shows active chromatin marks with low enrichment for the repressive mark, H3K27me3. MYCN, a commonly amplified oncogene in neuroblstoma, binds to the promoter and induces TERT expression. Strikingly, in neuroblastoma with long telomeres, the hypomethylated region spans the entire TERT locus, including multiple nearby genes with enrichment for the repressive H3K27me3 chromatin mark. Furthermore, subtelomeric regions showed enrichment of repressive chromatin marks in neuroblastomas with long telomeres relative to those with short telomeres. These repressive marks were even more evident at the genic loci, suggesting a telomere position effect (TPE). Inhibiting H3K27 methylation by three different EZH2 inhibitors induced the expression of TERT in cell lines with long telomeres and H3K27me3 marks in the promoter region. EZH2 inhibition facilitated MYCN binding to the TERT promoter in neuroblastoma cells with long telomeres. Taken together, these data suggest that epigenetic regulation of TERT expression differs in neuroblastoma depending on the telomere maintenance status, and H3K27 methylation is important in repressing TERT expression in neuroblastoma with long telomeres. SIGNIFICANCE: The epigenetic landscape of the TERT locus is unique in neuroblastoma. The DNA at the TERT locus, unlike other cancer cells and similar to normal cells, are hypomethylated in telomerase-positive neuroblastoma cells. The TERT locus is repressed by polycomb repressive complex-2 complex in neuroblastoma cells that have long telomeres and do not express TERT. Long telomeres in neuroblastoma cells are also associated with repressive chromatin states at the chromosomal termini, suggesting TPE.


Sujet(s)
Neuroblastome , Régions promotrices (génétique) , Telomerase , Télomère , Neuroblastome/génétique , Neuroblastome/anatomopathologie , Neuroblastome/métabolisme , Telomerase/génétique , Telomerase/métabolisme , Humains , Régions promotrices (génétique)/génétique , Télomère/métabolisme , Télomère/génétique , Lignée cellulaire tumorale , Méthylation de l'ADN/génétique , Protéine du proto-oncogène N-Myc/génétique , Protéine du proto-oncogène N-Myc/métabolisme , Régulation de l'expression des gènes tumoraux , Protéines du groupe Polycomb/génétique , Protéines du groupe Polycomb/métabolisme
12.
Sci Adv ; 10(25): eado1583, 2024 Jun 21.
Article de Anglais | MEDLINE | ID: mdl-38905335

RÉSUMÉ

Neuroblastoma is a childhood developmental cancer; however, its embryonic origins remain poorly understood. Moreover, in-depth studies of early tumor-driving events are limited because of the lack of appropriate models. Herein, we analyzed RNA sequencing data obtained from human neuroblastoma samples and found that loss of expression of trunk neural crest-enriched gene MOXD1 associates with advanced disease and worse outcome. Further, by using single-cell RNA sequencing data of human neuroblastoma cells and fetal adrenal glands and creating in vivo models of zebrafish, chick, and mouse, we show that MOXD1 is a determinate of tumor development. In addition, we found that MOXD1 expression is highly conserved and restricted to mesenchymal neuroblastoma cells and Schwann cell precursors during healthy development. Our findings identify MOXD1 as a lineage-restricted tumor-suppressor gene in neuroblastoma, potentiating further stratification of these tumors and development of novel therapeutic interventions.


Sujet(s)
Régulation de l'expression des gènes tumoraux , Gènes suppresseurs de tumeur , Neuroblastome , Danio zébré , Animaux , Humains , Souris , Lignée cellulaire tumorale , Lignage cellulaire/génétique , Crête neurale/métabolisme , Crête neurale/anatomopathologie , Neuroblastome/génétique , Neuroblastome/anatomopathologie , Neuroblastome/métabolisme , Cellules de Schwann/métabolisme , Cellules de Schwann/anatomopathologie , Danio zébré/génétique
14.
Int J Mol Sci ; 25(9)2024 Apr 27.
Article de Anglais | MEDLINE | ID: mdl-38732012

RÉSUMÉ

Neuroblastoma (NB) is the most commonly diagnosed extracranial solid tumor in children, accounting for 15% of all childhood cancer deaths. Although the 5-year survival rate of patients with a high-risk disease has increased in recent decades, NB remains a challenge in pediatric oncology, and the identification of novel potential therapeutic targets and agents is an urgent clinical need. The RNA-binding protein LIN28B has been identified as an oncogene in NB and is associated with a poor prognosis. Given that LIN28B acts by negatively regulating the biogenesis of the tumor suppressor let-7 miRNAs, we reasoned that selective interference with the LIN28B/let-7 miRNA interaction would increase let-7 miRNA levels, ultimately leading to reduced NB aggressiveness. Here, we selected (-)-epigallocatechin 3-gallate (EGCG) out of 4959 molecules screened as the molecule with the best inhibitory activity on LIN28B/let-7 miRNA interaction and showed that treatment with PLC/PLGA-PEG nanoparticles containing EGCG (EGCG-NPs) led to an increase in mature let-7 miRNAs and a consequent inhibition of NB cell growth. In addition, EGCG-NP pretreatment reduced the tumorigenic potential of NB cells in vivo. These experiments suggest that the LIN28B/let-7 miRNA axis is a good therapeutic target in NB and that EGCG, which can interfere with this interaction, deserves further preclinical evaluation.


Sujet(s)
Catéchine , microARN , Neuroblastome , Protéines de liaison à l'ARN , Catéchine/analogues et dérivés , Catéchine/pharmacologie , Neuroblastome/génétique , Neuroblastome/anatomopathologie , Neuroblastome/métabolisme , Neuroblastome/traitement médicamenteux , microARN/génétique , microARN/métabolisme , Humains , Protéines de liaison à l'ARN/métabolisme , Protéines de liaison à l'ARN/génétique , Animaux , Souris , Lignée cellulaire tumorale , Régulation de l'expression des gènes tumoraux/effets des médicaments et des substances chimiques , Prolifération cellulaire/effets des médicaments et des substances chimiques , Tests d'activité antitumorale sur modèle de xénogreffe , Souris nude
15.
Mol Cell ; 84(11): 2070-2086.e20, 2024 Jun 06.
Article de Anglais | MEDLINE | ID: mdl-38703770

RÉSUMÉ

The MYCN oncoprotein binds active promoters in a heterodimer with its partner protein MAX. MYCN also interacts with the nuclear exosome, a 3'-5' exoribonuclease complex, suggesting a function in RNA metabolism. Here, we show that MYCN forms stable high-molecular-weight complexes with the exosome and multiple RNA-binding proteins. MYCN binds RNA in vitro and in cells via a conserved sequence termed MYCBoxI. In cells, MYCN associates with thousands of intronic transcripts together with the ZCCHC8 subunit of the nuclear exosome targeting complex and enhances their processing. Perturbing exosome function results in global re-localization of MYCN from promoters to intronic RNAs. On chromatin, MYCN is then replaced by the MNT(MXD6) repressor protein, inhibiting MYCN-dependent transcription. RNA-binding-deficient alleles show that RNA-binding limits MYCN's ability to activate cell growth-related genes but is required for MYCN's ability to promote progression through S phase and enhance the stress resilience of neuroblastoma cells.


Sujet(s)
Protéine du proto-oncogène N-Myc , Protéines nucléaires , Protéines oncogènes , Protéines de liaison à l'ARN , Protéine du proto-oncogène N-Myc/métabolisme , Protéine du proto-oncogène N-Myc/génétique , Humains , Protéines de liaison à l'ARN/métabolisme , Protéines de liaison à l'ARN/génétique , Protéines nucléaires/métabolisme , Protéines nucléaires/génétique , Protéines oncogènes/métabolisme , Protéines oncogènes/génétique , Régions promotrices (génétique) , Lignée cellulaire tumorale , Neuroblastome/métabolisme , Neuroblastome/génétique , Neuroblastome/anatomopathologie , Exosomes/métabolisme , Exosomes/génétique , Introns , Liaison aux protéines , Noyau de la cellule/métabolisme , Exosome multienzyme ribonuclease complex/métabolisme , Exosome multienzyme ribonuclease complex/génétique , Régulation de l'expression des gènes tumoraux , ARN/métabolisme , ARN/génétique , Protéines de répression/métabolisme , Protéines de répression/génétique , Prolifération cellulaire
16.
Cell Rep ; 43(5): 114165, 2024 May 28.
Article de Anglais | MEDLINE | ID: mdl-38691450

RÉSUMÉ

The N6-methyladenosine (m6A) RNA modification is an important regulator of gene expression. m6A is deposited by a methyltransferase complex that includes methyltransferase-like 3 (METTL3) and methyltransferase-like 14 (METTL14). High levels of METTL3/METTL14 drive the growth of many types of adult cancer, and METTL3/METTL14 inhibitors are emerging as new anticancer agents. However, little is known about the m6A epitranscriptome or the role of the METTL3/METTL14 complex in neuroblastoma, a common pediatric cancer. Here, we show that METTL3 knockdown or pharmacologic inhibition with the small molecule STM2457 leads to reduced neuroblastoma cell proliferation and increased differentiation. These changes in neuroblastoma phenotype are associated with decreased m6A deposition on transcripts involved in nervous system development and neuronal differentiation, with increased stability of target mRNAs. In preclinical studies, STM2457 treatment suppresses the growth of neuroblastoma tumors in vivo. Together, these results support the potential of METTL3/METTL14 complex inhibition as a therapeutic strategy against neuroblastoma.


Sujet(s)
Différenciation cellulaire , Prolifération cellulaire , Methyltransferases , Neuroblastome , Methyltransferases/métabolisme , Methyltransferases/antagonistes et inhibiteurs , Neuroblastome/anatomopathologie , Neuroblastome/métabolisme , Neuroblastome/traitement médicamenteux , Neuroblastome/génétique , Humains , Différenciation cellulaire/effets des médicaments et des substances chimiques , Prolifération cellulaire/effets des médicaments et des substances chimiques , Lignée cellulaire tumorale , Animaux , Souris , Régulation de l'expression des gènes tumoraux/effets des médicaments et des substances chimiques , Adénosine/analogues et dérivés , Adénosine/métabolisme , Adénosine/pharmacologie
18.
Biochemistry ; 63(12): 1534-1542, 2024 Jun 18.
Article de Anglais | MEDLINE | ID: mdl-38804064

RÉSUMÉ

Zinc Finger MYND (Myeloid, Nervy, and DEAF-1) type containing 8 (ZMYND8) is a crucial epigenetic regulator that plays a multifaceted role in governing a spectrum of vital cellular processes, encompassing proliferation, apoptosis, migration, tumor suppression, and differentiation. It has emerged as a key player in neuronal differentiation by orchestrating the expression of neuronal lineage-committed genes. The present study uncovers the role of ZMYND8 in regulating the Sonic Hedgehog (SHH) signaling axis, which is crucial for neuronal differentiation. Genetic deletion of ZMYND8 leads to a significant reduction in SHH pathway genes, GLI1, and PTCH1 expression during all-trans-retinoic acid (ATRA)-induced differentiation. ZMYND8 and RNA pol II S5P are found to co-occupy the GLI1 and PTCH1 gene promoters, positively impacting their gene transcription upon ATRA treatment. Interestingly, ZMYND8 is found to counteract the inhibitory effects of Cyclopamine that block the upstream SHH pathway protein SMO, resulting in enhanced neurite formation in neuroblastoma cells following their treatment with ATRA. These results indicate that ZMYND8 is an epigenetic regulator of the SHH signaling pathway and has tremendous therapeutic potential in ATRA-mediated differentiation of neuroblastoma.


Sujet(s)
Différenciation cellulaire , Protéines Hedgehog , Neuroblastome , Transduction du signal , Trétinoïne , Protéines Hedgehog/métabolisme , Protéines Hedgehog/génétique , Humains , Différenciation cellulaire/effets des médicaments et des substances chimiques , Trétinoïne/pharmacologie , Transduction du signal/effets des médicaments et des substances chimiques , Neuroblastome/métabolisme , Neuroblastome/anatomopathologie , Neuroblastome/génétique , Lignée cellulaire tumorale , Récepteur Patched-1/métabolisme , Récepteur Patched-1/génétique , Facteurs de transcription/métabolisme , Facteurs de transcription/génétique , Protéine à doigt de zinc GLI1/métabolisme , Protéine à doigt de zinc GLI1/génétique , Régulation de l'expression des gènes tumoraux/effets des médicaments et des substances chimiques , Souris , Animaux , Protéines suppresseurs de tumeurs
19.
Mol Carcinog ; 63(8): 1559-1571, 2024 Aug.
Article de Anglais | MEDLINE | ID: mdl-38780126

RÉSUMÉ

The therapeutic effect of anlotinib on neuroblastoma is still not fully understood. This study aims to explore the differentiation therapeutic effects of anlotinib on neuroblastoma and its potential association with the neural development regulatory protein collapsin response mediator protein 5 (CRMP5), both in vivo and in vitro. A patient-derived xenograft (PDX) model was established to observe the therapeutic effect of anlotinib. Neuroblastoma cell lines SK-N-SH and SK-N-AS were cultured to observe the morphological impact of anlotinib. Transwell assay was used to evaluate the cell invasion, and Western blot analysis and immunohistochemistry were employed to detect the expressions of neuronal differentiation-related proteins. Results indicate that anlotinib effectively inhibited tumor growth in the PDX model, modulated the expressions of neuronal differentiation markers. In vitro, anlotinib treatment induced neurite outgrowth in neuroblastoma cells and inhibited their invasive ability, reflecting a change in neuronal marker expression patterns consistent with the PDX model. Similarly, in the SK-N-AS mouse xenograft model, anlotinib demonstrated comparable tumor-suppressing effects and promoted neuronal-like differentiation. Additionally, anlotinib significantly downregulated CRMP5 expression in neuroblastoma both in vivo and in vitro. Overexpression of CRMP5 significantly reversed the differentiation therapy effect of anlotinib, exacerbating the aggressiveness and reducing the differentiation level of neuroblastoma. These findings highlight the potential of anlotinib as an anti-neuroblastoma agent. It may suppress tumor proliferation and invasion by promoting the differentiation of tumor cells towards a neuronal-like state, and this differentiation therapy effect involves the inhibition of CRMP5 signaling.


Sujet(s)
Différenciation cellulaire , Prolifération cellulaire , Indoles , Protéines de tissu nerveux , Neuroblastome , Quinoléines , Tests d'activité antitumorale sur modèle de xénogreffe , Humains , Neuroblastome/traitement médicamenteux , Neuroblastome/anatomopathologie , Neuroblastome/métabolisme , Neuroblastome/génétique , Animaux , Souris , Quinoléines/pharmacologie , Différenciation cellulaire/effets des médicaments et des substances chimiques , Indoles/pharmacologie , Lignée cellulaire tumorale , Protéines de tissu nerveux/métabolisme , Protéines de tissu nerveux/génétique , Prolifération cellulaire/effets des médicaments et des substances chimiques , Régulation de l'expression des gènes tumoraux/effets des médicaments et des substances chimiques , Régulation négative/effets des médicaments et des substances chimiques , Neurones/effets des médicaments et des substances chimiques , Neurones/métabolisme , Neurones/anatomopathologie , Souris nude , Hydrolases/génétique , Hydrolases/métabolisme , Antinéoplasiques/pharmacologie , Protéines associées aux microtubules
20.
Sci Adv ; 10(22): eadm9449, 2024 May 31.
Article de Anglais | MEDLINE | ID: mdl-38820154

RÉSUMÉ

Pediatric cancers are frequently driven by genomic alterations that result in aberrant transcription factor activity. Here, we used functional genomic screens to identify multiple genes within the transcriptional coactivator Spt-Ada-Gcn5-acetyltransferase (SAGA) complex as selective dependencies for MYCN-amplified neuroblastoma, a disease of dysregulated development driven by an aberrant oncogenic transcriptional program. We characterized the DNA recruitment sites of the SAGA complex in neuroblastoma and the consequences of loss of SAGA complex lysine acetyltransferase (KAT) activity on histone acetylation and gene expression. We demonstrate that loss of SAGA complex KAT activity is associated with reduced MYCN binding on chromatin, suppression of MYC/MYCN gene expression programs, and impaired cell cycle progression. Further, we showed that the SAGA complex is pharmacologically targetable in vitro and in vivo with a KAT2A/KAT2B proteolysis targeting chimeric. Our findings expand our understanding of the histone-modifying complexes that maintain the oncogenic transcriptional state in this disease and suggest therapeutic potential for inhibitors of SAGA KAT activity in MYCN-amplified neuroblastoma.


Sujet(s)
Régulation de l'expression des gènes tumoraux , Protéine du proto-oncogène N-Myc , Neuroblastome , Neuroblastome/génétique , Neuroblastome/métabolisme , Neuroblastome/anatomopathologie , Humains , Protéine du proto-oncogène N-Myc/génétique , Protéine du proto-oncogène N-Myc/métabolisme , Lignée cellulaire tumorale , Histone acetyltransferases/métabolisme , Histone acetyltransferases/génétique , Acétylation , Histone/métabolisme , Animaux , Amplification de gène , Chromatine/métabolisme , Chromatine/génétique , Souris
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE
...