Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 20 de 4.241
Filtrer
1.
Cancer Biol Ther ; 25(1): 2385517, 2024 Dec 31.
Article de Anglais | MEDLINE | ID: mdl-39087955

RÉSUMÉ

BACKGROUND: CDK4 is highly expressed and associated with poor prognosis and decreased survival in advanced neuroblastoma (NB). Targeting CDK4 degradation presents a potentially promising therapeutic strategy compared to conventional CDK4 inhibitors. However, the autophagic degradation of the CDK4 protein and its anti-proliferation effect in NB cells has not been mentioned. RESULTS: We identified autophagy as a new pathway for the degradation of CDK4. Firstly, autophagic degradation of CDK4 is critical for NVP-BEZ235-induced G0/G1 arrest, as demonstrated by the overexpression of CDK4, autophagy inhibition, and blockade of autophagy-related genes. Secondly, we present the first evidence that p62 binds to CDK4 and then enters the autophagy-lysosome to degrade CDK4 in a CTSB-dependent manner in NVP-BEZ235 treated NB cells. Similar results regarding the interaction between p62 and CDK4 were observed in the NVP-BEZ235 treated NB xenograft mouse model. CONCLUSIONS: Autophagic degradation of CDK4 plays a pivotal role in G0/G1 cell cycle arrest in NB cells treated with NVP-BEZ235.


Sujet(s)
Autophagie , Kinase-4 cycline-dépendante , Points de contrôle de la phase G1 du cycle cellulaire , Neuroblastome , Kinase-4 cycline-dépendante/métabolisme , Neuroblastome/métabolisme , Neuroblastome/anatomopathologie , Neuroblastome/traitement médicamenteux , Neuroblastome/génétique , Humains , Animaux , Souris , Autophagie/effets des médicaments et des substances chimiques , Points de contrôle de la phase G1 du cycle cellulaire/effets des médicaments et des substances chimiques , Lignée cellulaire tumorale , Tests d'activité antitumorale sur modèle de xénogreffe , Quinoléines/pharmacologie , Phase G0/effets des médicaments et des substances chimiques , Prolifération cellulaire/effets des médicaments et des substances chimiques , Imidazoles/pharmacologie , Souris nude , Protéolyse
2.
Int J Mol Sci ; 25(13)2024 Jun 28.
Article de Anglais | MEDLINE | ID: mdl-39000210

RÉSUMÉ

Neurodegeneration diseases (NDs) are a group of complex diseases primarily characterized by progressive loss of neurons affecting mental function and movement. Oxidative stress is one of the factors contributing to the pathogenesis of NDs, including Alzheimer's disease (AD). These reactive species disturb mitochondrial function and accelerate other undesirable conditions including tau phosphorylation, inflammation, and cell death. Therefore, preventing oxidative stress is one of the imperative methods in the treatment of NDs. To accomplish this, we prepared hexane and ethyl acetate extracts of Anethum graveolens (dill) and identified the major phyto-components (apiol, carvone, and dihydrocarvone) by GC-MS. The extracts and major bioactives were assessed for neuroprotective potential and mechanism in hydrogen peroxide-induced oxidative stress in the SH-SY5Y neuroblastoma cell model and other biochemical assays. The dill (extracts and bioactives) provided statistically significant neuroprotection from 0.1 to 30 µg/mL by mitigating ROS levels, restoring mitochondrial membrane potential, reducing lipid peroxidation, and reviving the glutathione ratio. They moderately inhibited acetylcholine esterase (IC50 dill extracts 400-500 µg/mL; carvone 275.7 µg/mL; apiole 388.3 µg/mL), displayed mild anti-Aß1-42 fibrilization (DHC 26.6%) and good anti-oligomerization activity (>40% by dill-EA, carvone, and apiole). Such multifactorial neuroprotective displayed by dill and bioactives would help develop a safe, low-cost, and small-molecule drug for NDs.


Sujet(s)
Anethum graveolens , Neuroblastome , Neuroprotecteurs , Stress oxydatif , Extraits de plantes , Graines , Humains , Neuroprotecteurs/pharmacologie , Neuroprotecteurs/composition chimique , Lignée cellulaire tumorale , Extraits de plantes/pharmacologie , Extraits de plantes/composition chimique , Neuroblastome/métabolisme , Neuroblastome/traitement médicamenteux , Neuroblastome/anatomopathologie , Stress oxydatif/effets des médicaments et des substances chimiques , Anethum graveolens/composition chimique , Graines/composition chimique , Potentiel de membrane mitochondriale/effets des médicaments et des substances chimiques , Peptides bêta-amyloïdes/métabolisme , Peroxydation lipidique/effets des médicaments et des substances chimiques , Espèces réactives de l'oxygène/métabolisme , Peroxyde d'hydrogène , Composés phytochimiques/pharmacologie , Composés phytochimiques/composition chimique , Survie cellulaire/effets des médicaments et des substances chimiques , Acetylcholinesterase/métabolisme
3.
Theranostics ; 14(9): 3439-3469, 2024.
Article de Anglais | MEDLINE | ID: mdl-38948053

RÉSUMÉ

Rationale: Synergic reprogramming of metabolic dominates neuroblastoma (NB) progression. It is of great clinical implications to develop an individualized risk prognostication approach with stratification-guided therapeutic options for NB based on elucidating molecular mechanisms of metabolic reprogramming. Methods: With a machine learning-based multi-step program, the synergic mechanisms of metabolic reprogramming-driven malignant progression of NB were elucidated at single-cell and metabolite flux dimensions. Subsequently, a promising metabolic reprogramming-associated prognostic signature (MPS) and individualized therapeutic approaches based on MPS-stratification were developed and further validated independently using pre-clinical models. Results: MPS-identified MPS-I NB showed significantly higher activity of metabolic reprogramming than MPS-II counterparts. MPS demonstrated improved accuracy compared to current clinical characteristics [AUC: 0.915 vs. 0.657 (MYCN), 0.713 (INSS-stage), and 0.808 (INRG-stratification)] in predicting prognosis. AZD7762 and etoposide were identified as potent therapeutics against MPS-I and II NB, respectively. Subsequent biological tests revealed AZD7762 substantially inhibited growth, migration, and invasion of MPS-I NB cells, more effectively than that of MPS-II cells. Conversely, etoposide had better therapeutic effects on MPS-II NB cells. More encouragingly, AZD7762 and etoposide significantly inhibited in-vivo subcutaneous tumorigenesis, proliferation, and pulmonary metastasis in MPS-I and MPS-II samples, respectively; thereby prolonging survival of tumor-bearing mice. Mechanistically, AZD7762 and etoposide-induced apoptosis of the MPS-I and MPS-II cells, respectively, through mitochondria-dependent pathways; and MPS-I NB resisted etoposide-induced apoptosis by addiction of glutamate metabolism and acetyl coenzyme A. MPS-I NB progression was fueled by multiple metabolic reprogramming-driven factors including multidrug resistance, immunosuppressive and tumor-promoting inflammatory microenvironments. Immunologically, MPS-I NB suppressed immune cells via MIF and THBS signaling pathways. Metabolically, the malignant proliferation of MPS-I NB cells was remarkably supported by reprogrammed glutamate metabolism, tricarboxylic acid cycle, urea cycle, etc. Furthermore, MPS-I NB cells manifested a distinct tumor-promoting developmental lineage and self-communication patterns, as evidenced by enhanced oncogenic signaling pathways activated with development and self-communications. Conclusions: This study provides deep insights into the molecular mechanisms underlying metabolic reprogramming-mediated malignant progression of NB. It also sheds light on developing targeted medications guided by the novel precise risk prognostication approaches, which could contribute to a significantly improved therapeutic strategy for NB.


Sujet(s)
Évolution de la maladie , Étoposide , Neuroblastome , Microenvironnement tumoral , Neuroblastome/traitement médicamenteux , Neuroblastome/métabolisme , Neuroblastome/anatomopathologie , Microenvironnement tumoral/effets des médicaments et des substances chimiques , Humains , Animaux , Souris , Lignée cellulaire tumorale , Étoposide/pharmacologie , Étoposide/usage thérapeutique , Pronostic , Reprogrammation cellulaire/effets des médicaments et des substances chimiques , Prolifération cellulaire/effets des médicaments et des substances chimiques , Tests d'activité antitumorale sur modèle de xénogreffe , Thérapie moléculaire ciblée/méthodes , Apprentissage machine , Apoptose/effets des médicaments et des substances chimiques ,
4.
BMC Complement Med Ther ; 24(1): 257, 2024 Jul 09.
Article de Anglais | MEDLINE | ID: mdl-38982456

RÉSUMÉ

BACKGROUND: Neuroblastoma, a prevalent solid tumor in children, often manifests with hidden onset sites, rapid growth, and high metastatic potential. The prognosis for children with high-risk neuroblastoma remains poor, highlighting the urgent need for novel prognostic models and therapeutic avenues. In recent years, puerarin, as a kind of small molecule drug extracted from Chinese medicine Pueraria lobata, has demonstrated significant anticancer effects on various cancer cell types. In this study, through bioinformatics analysis and in vitro experiments, the potential and mechanism of puerarin in the treatment of neuroblastoma were investigated, and a prognostic model was established. METHODS: A total of 9 drug-disease related targets were observed by constructing a database of drug targets and disease genes. Besides, GO and KEGG enrichment analysis was performed to explore the potential mechanism of its therapeutic effect. To construct the prognostic model, risk regression analysis and LASSO analysis were carried out for validation. Finally, the prognostic genes were identified. Parachute test and immunofluorescence staining were performed to verify the potential mechanism of puerarin in neuroblastoma treatment. RESULTS: Three prognostic genes, i.e., BIRC5, TIMP2 and CASP9, were identified. In vitro studies verified puerarin's impact on BIRC5, TIMP2, and CASP9 expression, inhibiting proliferation in neuroblastoma SH-SY5Y cells. Puerarin disrupts the cytoskeleton, boosts gap junctional communication, curtailing invasion and migration, and induces mitochondrial damage in SH-SY5Y cells. CONCLUSIONS: Based on network pharmacology and bioinformatics analysis, combined with in vitro experimental verification, puerarin was hereby observed to enhance GJIC in neuroblastoma, destroy cytoskeleton and thus inhibit cell invasion and migration, cause mitochondrial damage of tumor cells, and inhibit cell proliferation. Overall, puerarin, as a natural medicinal compound, does hold potential as a novel therapy for neuroblastoma.


Sujet(s)
Biologie informatique , Isoflavones , Neuroblastome , Neuroblastome/traitement médicamenteux , Isoflavones/pharmacologie , Humains , Lignée cellulaire tumorale , Prolifération cellulaire/effets des médicaments et des substances chimiques
5.
Biomed Pharmacother ; 177: 117040, 2024 Aug.
Article de Anglais | MEDLINE | ID: mdl-38959605

RÉSUMÉ

Invariant natural killer T cell (iNKT) cells produce large amounts of cytokines in response to α-Galactosylceramide (α-GalCer) stimulation. An analog containing two phenyl rings on the acyl chain, C34, was previously found to be more Th1-biased than α-GalCer and triggered greater anticancer activities against breast cancer, melanoma and lung cancer in mice. Since liver is enriched in iNKT cells, we investigated anticancer efficacy of C34 on neuroblastoma with hepatic metastasis. C34 induced Th1-biased cytokine secretions in the liver, significantly suppressed neuroblastoma growth/metastasis and prolonged mouse survival. The anti-tumor efficacy might be attributed to greater expansions of hepatic NKT, NK, CD4+ T, and CD8+ T cells as well as reduction of the number of SSCloGr1intCD11b+ subset of myeloid-derived suppressor cells (MDSCs) in the liver of tumor-bearing mice, as compared to DMSO control group. C34 also upregulated expression of CD1d and CD11c, especially in the SSCloGr1intCD11b+ subset of MDSCs, which might be killed by C34-activated NKT cells, attributing to their reduced number. In addition, C34 also induced expansion of CD4+ T, CD8+ T, and NK cells, which might eliminate neuroblastoma cells. These immune-modulating effects of C34 might act in concert in the local milieu of liver to suppress the tumor growth. Further analysis of database of neuroblastoma revealed that patients with high CD11c expression in the monocytic MDSCs in the tumor had longer survival, suggesting the potential clinical application of C34 for treatment of neuroblastoma.


Sujet(s)
Glycolipides , Tumeurs du foie , Cellules T tueuses naturelles , Neuroblastome , Animaux , Neuroblastome/anatomopathologie , Neuroblastome/traitement médicamenteux , Neuroblastome/immunologie , Cellules T tueuses naturelles/effets des médicaments et des substances chimiques , Cellules T tueuses naturelles/immunologie , Cellules T tueuses naturelles/métabolisme , Tumeurs du foie/traitement médicamenteux , Tumeurs du foie/anatomopathologie , Tumeurs du foie/immunologie , Tumeurs du foie/secondaire , Lignée cellulaire tumorale , Souris , Glycolipides/pharmacologie , Humains , Femelle , Cytokines/métabolisme , Cellules myéloïdes suppressives/effets des médicaments et des substances chimiques , Cellules myéloïdes suppressives/immunologie , Cellules myéloïdes suppressives/métabolisme , Antinéoplasiques/pharmacologie , Galactosylcéramides/pharmacologie
6.
Clinics (Sao Paulo) ; 79: 100434, 2024.
Article de Anglais | MEDLINE | ID: mdl-38959634

RÉSUMÉ

OBJECTIVES: To retrospectively investigate the impact of pre-treatment Extracellular Volume Fraction (ECV) measured by Computed Tomography (CT) on the response of primary lesions to preoperative chemotherapy in abdominal neuroblastoma. METHODS: A total of seventy-five patients with abdominal neuroblastoma were retrospectively included in the study. The regions of interest for the primary lesion and aorta were determined on unenhanced and equilibrium phase CT images before treatment, and their average CT values were measured. Based on patient hematocrit and average CT values, the ECV was calculated. The correlation between ECV and the reduction in primary lesion volume was examined. A receiver operating characteristic curve was generated to assess the predictive performance of ECV for a very good partial response of the primary lesion. RESULTS: There was a negative correlation between primary lesion volume reduction and ECV (r = -0.351, p = 0.002), and primary lesions with very good partial response had lower ECV (p < 0.001). The area under the curve for ECV in predicting the very good partial response of primary lesion was 0.742 (p < 0.001), with a 95 % Confidence Interval of 0.628 to 0.836. The optimal cut-off value was 0.28, and the sensitivity and specificity were 62.07 % and 84.78 %, respectively. CONCLUSIONS: The measurement of pre-treatment ECV on CT images demonstrates a significant correlation with the response of the primary lesion to preoperative chemotherapy in abdominal neuroblastoma.


Sujet(s)
Tumeurs de l'abdomen , Neuroblastome , Tomodensitométrie , Humains , Neuroblastome/imagerie diagnostique , Neuroblastome/traitement médicamenteux , Neuroblastome/chirurgie , Neuroblastome/anatomopathologie , Mâle , Femelle , Études rétrospectives , Tomodensitométrie/méthodes , Enfant d'âge préscolaire , Enfant , Nourrisson , Tumeurs de l'abdomen/imagerie diagnostique , Tumeurs de l'abdomen/traitement médicamenteux , Tumeurs de l'abdomen/anatomopathologie , Tumeurs de l'abdomen/chirurgie , Résultat thérapeutique , Courbe ROC , Valeur prédictive des tests , Adolescent , Charge tumorale/effets des médicaments et des substances chimiques , Sensibilité et spécificité , Valeurs de référence , Protocoles de polychimiothérapie antinéoplasique/usage thérapeutique , Reproductibilité des résultats
7.
Pediatr Blood Cancer ; 71(9): e31175, 2024 Sep.
Article de Anglais | MEDLINE | ID: mdl-38961591

RÉSUMÉ

Atypical hemolytic uremic syndrome (aHUS) is a complement-mediated thrombotic microangiopathy sometimes associated with germline variants in genes of the complement system. Clinical findings of microangiopathic hemolytic anemia, thrombocytopenia, and acute kidney injury arise due to aberrant complement protein activation in the circulation. A 13-month-old boy with metastatic neuroblastoma (NB) developed aHUS during his first cycle of induction chemotherapy with germline testing revealing a complement factor H (CFH) gene mutation, currently classified as a variant of uncertain significance (VUS). Now he is in disease remission after successful complement blockade therapy, thus highlighting a unique presentation of aHUS in a patient with newly diagnosed NB.


Sujet(s)
Syndrome hémolytique et urémique atypique , Chimiothérapie d'induction , Neuroblastome , Humains , Neuroblastome/traitement médicamenteux , Neuroblastome/anatomopathologie , Neuroblastome/génétique , Mâle , Syndrome hémolytique et urémique atypique/traitement médicamenteux , Syndrome hémolytique et urémique atypique/génétique , Syndrome hémolytique et urémique atypique/anatomopathologie , Chimiothérapie d'induction/effets indésirables , Nourrisson , Facteur H du complément/génétique , Protocoles de polychimiothérapie antinéoplasique/effets indésirables , Protocoles de polychimiothérapie antinéoplasique/usage thérapeutique , Mutation germinale
8.
Pediatr Blood Cancer ; 71(9): e31173, 2024 Sep.
Article de Anglais | MEDLINE | ID: mdl-38965702

RÉSUMÉ

BACKGROUND: Tandem high-dose chemotherapy and autologous stem cell transplantation (HDCT/auto-SCT) and incorporation of 131I-metaiodobenzylguanidine (131I-MIBG) treatment have shown positive outcomes in high-risk neuroblastoma. However, more optimized treatment strategies are still needed. PROCEDURE: The NB-2014 study was a nonrandomized, prospective trial that examined survival outcomes in metastatic high-risk neuroblastoma patients using response-adapted consolidation therapy. We used post-induction residual 123I-MIBG status at metastatic sites as a treatment response marker. Patients achieving complete resolution of MIBG uptake at metastatic sites underwent a reduced first HDCT/auto-SCT with a 20% dose reduction in HDCT. After the first HDCT/auto-SCT, patients with remaining MIBG uptake received dose-escalated (18 mCi/kg) 131I-MIBG treatment. In contrast, those with complete resolution of MIBG at metastatic sites received a standard dose (12 mCi/kg) of 131I-MIBG. We compared survival and toxicity outcomes with a historical control group from the NB-2009. RESULTS: Of 65 patients treated, 63% achieved complete resolution of MIBG uptake at metastatic sites following induction chemotherapy, while 29% of patients still had MIBG uptake at metastatic sites after the first HDCT/auto-SCT. The 3-year event-free survival (EFS) and overall survival (OS) rates were 68.2% ± 6.0% and 86.5% ± 4.5%, respectively. Compared to NB-2009, EFS was similar (p = .855); however, NB-2014 had a higher OS (p = .031), a lower cumulative incidence of treatment-related mortality (p = .036), and fewer acute and late toxicities. CONCLUSIONS: Our results suggest that response-adaptive consolidation therapy based on chemotherapy response at metastatic sites facilitates better treatment tailoring, and appears promising for patients with metastatic high-risk neuroblastoma.


Sujet(s)
3-Iodobenzyl-guanidine , Chimiothérapie de consolidation , Neuroblastome , Humains , Neuroblastome/thérapie , Neuroblastome/mortalité , Neuroblastome/anatomopathologie , Neuroblastome/traitement médicamenteux , Femelle , Mâle , Enfant d'âge préscolaire , Nourrisson , Enfant , 3-Iodobenzyl-guanidine/usage thérapeutique , Études prospectives , Taux de survie , Protocoles de polychimiothérapie antinéoplasique/usage thérapeutique , Adolescent , Études de suivi , Transplantation autologue , Pronostic , Transplantation de cellules souches hématopoïétiques , Radiopharmaceutiques
9.
Sci Rep ; 14(1): 16636, 2024 Jul 18.
Article de Anglais | MEDLINE | ID: mdl-39025941

RÉSUMÉ

In therapies, curcumin is now commonly formulated in liposomal form, administered through injections or creams. This enhances its concentration at the cellular level compared to its natural form ingestion. Due to its hydrophobic nature, curcumin is situated in the lipid part of the membrane, thereby modifying its properties and influencing processes The aim of the research was to investigate whether the toxicity of specific concentrations of curcumin, assessed through biochemical tests for the SK-N-SH and H-60 cell lines, is related to structural changes in the membranes of these cells, caused by the localization of curcumin in their hydrophobic regions. Biochemical tests were performed using spectrophotometric methods. Langmuir technique were used to evaluate the interaction of the curcumin with the studied lipids. Direct introduction of curcumin into the membranes alters their physicochemical parameters. The extent of these changes depends on the initial properties of the membrane. In the conducted research, it has been demonstrated that curcumin may exhibit toxicity to human cells. The mechanism of this toxicity is related to its localization in cell membranes, leading to their dysfunction. The sensitivity of cells to curcumin presence depends on the saturation level of their membranes; the more rigid the membrane, the lower the concentration of curcumin causes its disruption.


Sujet(s)
Membrane cellulaire , Curcumine , Neuroblastome , Curcumine/pharmacologie , Curcumine/composition chimique , Humains , Membrane cellulaire/effets des médicaments et des substances chimiques , Membrane cellulaire/métabolisme , Neuroblastome/traitement médicamenteux , Neuroblastome/métabolisme , Neuroblastome/anatomopathologie , Lignée cellulaire tumorale , Leucémies/traitement médicamenteux , Leucémies/métabolisme , Leucémies/anatomopathologie , Antinéoplasiques/pharmacologie , Antinéoplasiques/composition chimique , Interactions hydrophobes et hydrophiles
10.
Med Oncol ; 41(8): 194, 2024 Jul 03.
Article de Anglais | MEDLINE | ID: mdl-38958814

RÉSUMÉ

Neuroblastoma is a common nervous system tumor in childhood, and current treatments are not adequate. HSP90 is a molecular chaperone protein that plays a critical role in the regulation of cancer-related proteins. HSP90 inhibition may exert anticancer effects by targeting cancer-related processes such as tumor growth, cell proliferation, metastasis, and apoptosis. Therefore, HSP90 inhibition is a promising strategy in the treatment of various types of cancer, and the development of next-generation inhibitors could potentially lead to more effective and safer treatments. XL-888 and Debio0932 is a next-generation HSP90 inhibitor and can inhibit the correct folding and stabilization of client proteins that cancer-associated HSP90 helps to fold correctly. In this study, we aimed to investigate the comprehensive molecular pathways of the anticancer activity of XL-888 and Debio0932 in human neuroblastoma cells SH-SY5Y. The cytotoxic effects of XL-888 and Debio0932 on the neuroblastoma cell line SH-SY5Y cells were evaluated by MTT assay. Then, the effect of these HSP90 inhibitors on the expression of important genes in cancer was revealed by Quantitative Real Time Polymerase Chain Reaction (qRT-PCR) method. The qRT-PCR data were evaluated using Kyoto Encyclopedia of Genes and Genomes (KEGG) and Gene Ontology (GO) biological process tools. Finally, the effect of HSP90 inhibitors on HSP27, HSP70 and HSP90 protein expression was investigated by Western blotting analysis. The results revealed that XL-888 and Debio0932 had a role in regulating many cancer-related pathways such as migration, invasion, metastasis, angiogenesis, and apoptosis in SH-SY5Y cells. In conclusion, it shows that HSP90 inhibitors can be considered as a promising candidate in the treatment of neuroblastoma and resistance to chemotherapy.


Sujet(s)
Antinéoplasiques , Protéines du choc thermique HSP90 , Neuroblastome , Humains , Protéines du choc thermique HSP90/antagonistes et inhibiteurs , Neuroblastome/traitement médicamenteux , Neuroblastome/anatomopathologie , Neuroblastome/métabolisme , Lignée cellulaire tumorale , Antinéoplasiques/pharmacologie , Apoptose/effets des médicaments et des substances chimiques , Prolifération cellulaire/effets des médicaments et des substances chimiques
11.
PeerJ ; 12: e17732, 2024.
Article de Anglais | MEDLINE | ID: mdl-39035166

RÉSUMÉ

Alzheimer's disease (AD) is a neurodegenerative disease that causes physical damage to neuronal connections, leading to brain atrophy. This disruption of synaptic connections results in mild to severe cognitive impairments. Unfortunately, no effective treatment is currently known to prevent or reverse the symptoms of AD. The aim of this study was to investigate the effects of three synthetic peptides, i.e., KLVFF, RGKLVFFGR and RIIGL, on an AD in vitro model represented by differentiated SH-SY5Y neuroblastoma cells exposed to retinoic acid (RA) and brain-derived neurotrophic factor (BDNF). The results demonstrated that RIIGL peptide had the least significant cytotoxic activity to normal SH-SY5Y while exerting high cytotoxicity against the differentiated cells. The mechanism of RIIGL peptide in the differentiated SH-SY5Y was investigated based on changes in secretory proteins compared to another two peptides. A total of 380 proteins were identified, and five of them were significantly detected after treatment with RIIGL peptide. These secretory proteins were found to be related to microtubule-associated protein tau (MAPT) and amyloid-beta precursor protein (APP). RIIGL peptide acts on differentiated SH-SY5Y by regulating amyloid-beta formation, neuron apoptotic process, ceramide catabolic process, and oxidative phosphorylation and thus has the potentials to treat AD.


Sujet(s)
Maladie d'Alzheimer , Peptides bêta-amyloïdes , Facteur neurotrophique dérivé du cerveau , Différenciation cellulaire , Neuroblastome , Protéines tau , Humains , Maladie d'Alzheimer/métabolisme , Maladie d'Alzheimer/anatomopathologie , Maladie d'Alzheimer/traitement médicamenteux , Peptides bêta-amyloïdes/métabolisme , Peptides bêta-amyloïdes/pharmacologie , Neuroblastome/anatomopathologie , Neuroblastome/métabolisme , Neuroblastome/traitement médicamenteux , Lignée cellulaire tumorale , Différenciation cellulaire/effets des médicaments et des substances chimiques , Facteur neurotrophique dérivé du cerveau/métabolisme , Facteur neurotrophique dérivé du cerveau/pharmacologie , Protéines tau/métabolisme , Trétinoïne/pharmacologie , Précurseur de la protéine bêta-amyloïde/métabolisme , Précurseur de la protéine bêta-amyloïde/génétique
12.
Nutrients ; 16(11)2024 Jun 06.
Article de Anglais | MEDLINE | ID: mdl-38892725

RÉSUMÉ

Xanthohumol (Xn) is an antioxidant flavonoid mainly extracted from hops (Humulus lupulus), one of the main ingredients of beer. As with other bioactive compounds, their therapeutic potential against different diseases has been tested, one of which is Alzheimer's disease (AD). Adenosine is a neuromodulatory nucleoside that acts through four different G protein-coupled receptors: A1 and A3, which inhibit the adenylyl cyclases (AC) pathway, and A2A and A2B, which stimulate this activity, causing either a decrease or an increase, respectively, in the release of excitatory neurotransmitters such as glutamate. This adenosinergic pathway, which is altered in AD, could be involved in the excitotoxicity process. Therefore, the aim of this work is to describe the effect of Xn on the adenosinergic pathway using cell lines. For this purpose, two different cellular models, rat glioma C6 and human neuroblastoma SH-SY5Y, were exposed to a non-cytotoxic 10 µM Xn concentration. Adenosine A1 and A2A, receptor levels, and activities related to the adenosine pathway, such as adenylate cyclase, protein kinase A, and 5'-nucleotidase, were analyzed. The adenosine A1 receptor was significantly increased after Xn exposure, while no changes in A2A receptor membrane levels or AC activity were reported. Regarding 5'-nucleotidases, modulation of their activity by Xn was noted since CD73, the extracellular membrane attached to 5'-nucleotidase, was significantly decreased in the C6 cell line. In conclusion, here we describe a novel pathway in which the bioactive flavonoid Xn could have potentially beneficial effects on AD as it increases membrane A1 receptors while modulating enzymes related to the adenosine pathway in cell cultures.


Sujet(s)
Adénosine , Flavonoïdes , Gliome , Humulus , Neuroblastome , Propiophénones , Récepteur A1 à l'adénosine , Humains , Flavonoïdes/pharmacologie , Rats , Propiophénones/pharmacologie , Animaux , Adénosine/métabolisme , Adénosine/pharmacologie , Lignée cellulaire tumorale , Humulus/composition chimique , Neuroblastome/métabolisme , Neuroblastome/traitement médicamenteux , Gliome/métabolisme , Gliome/traitement médicamenteux , Récepteur A1 à l'adénosine/métabolisme , Transduction du signal/effets des médicaments et des substances chimiques , Adenylate Cyclase/métabolisme , Récepteur A2A à l'adénosine/métabolisme
13.
J Physiol Investig ; 67(3): 103-106, 2024 May 01.
Article de Anglais | MEDLINE | ID: mdl-38857206

RÉSUMÉ

A recent study investigated the correlation between telmisartan (TEL) exposure and Alzheimer's disease (AD) risk among African Americans (AAs) and European Americans. Their findings indicated that moderate-to-high TEL exposure was linked to a decreased incidence of AD among AAs. These results suggest a potential association between TEL and a reduced risk of AD specifically within the AA population. Here, we investigated the effects of TEL, either alone or in combination with ranolazine (Ran) or dapagliflozin (Dapa), on voltage-gated Na + currents ( INa ) in Neuro-2a cells. TEL, primarily used for treating hypertension and cardiovascular disorders, showed a stimulatory effect on INa , while Ran and Dapa reversed this stimulation. In Neuro-2a cells, we demonstrated that with exposure to TEL, the transient ( INa(T) ) and late ( INa(L) ) components of INa were differentially stimulated with effective EC 50 's of 16.9 and 3.1 µM, respectively. The research implies that TEL's impact on INa might be associated with enhanced neuronal excitability. This study highlights the complex interplay between TEL, Ran, and Dapa on INa and their potential implications for AD, emphasizing the need for further investigation to understand the mechanisms involved.


Sujet(s)
Acétanilides , Composés benzhydryliques , Benzimidazoles , Benzoates , Glucosides , Neuroblastome , Pipérazines , Ranolazine , Telmisartan , Telmisartan/pharmacologie , Telmisartan/usage thérapeutique , Glucosides/pharmacologie , Glucosides/usage thérapeutique , Benzimidazoles/pharmacologie , Benzimidazoles/usage thérapeutique , Ranolazine/pharmacologie , Ranolazine/usage thérapeutique , Benzoates/pharmacologie , Benzoates/usage thérapeutique , Neuroblastome/traitement médicamenteux , Neuroblastome/anatomopathologie , Lignée cellulaire tumorale , Animaux , Acétanilides/pharmacologie , Pipérazines/pharmacologie , Pipérazines/usage thérapeutique , Souris , Composés benzhydryliques/pharmacologie , Composés benzhydryliques/usage thérapeutique , Neurones/effets des médicaments et des substances chimiques , Neurones/métabolisme , Ouverture et fermeture des portes des canaux ioniques/effets des médicaments et des substances chimiques
14.
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
16.
Biomed Pharmacother ; 177: 116993, 2024 Aug.
Article de Anglais | MEDLINE | ID: mdl-38889643

RÉSUMÉ

AIM: Neuroblastoma (NB) is, in spite of current intensive therapy with severe side effects, still not cured so new therapies are needed. Recently, we showed combining phosphoinositide 3-kinase (PI3K) (BYL719), fibroblast growth factor receptor (FGFR) (JNJ-42756493) and cyclin-dependent kinase 4/6 (CDK4/6) (PD-0332991) inhibitors, in vitro in NB cell lines grown as monolayers had synergistic effects. However, there were variations depending on the combinations used and the targeted NB cell lines. To obtain further information and to mimic more natural circumstances, we investigated the effects of single and combined administrations of the above inhibitors in spheroid NB-cultures. MATERIAL AND METHODS: Spheroid cultures of NB cell lines SK-N-AS, SK-N-BE(2)-C, SK-N-FI and SK-N-SH were established and treated with single and combined administrations of BYL719, JNJ-42756493, and PD-0332991 and followed for growth, viability, proliferation, cytotoxicity and migration. KEY FINDINGS: Single inhibitor administrations gave dose dependent responses with regard to growth and viability and their combinations were efficient and resulted in a range of additive and synergistic effects. The responses to individual drugs and their various combinations were predominantly alike regardless of whether the cells were cultivated in monolayer or D spheroid NB models. However, in general, slightly higher drug concentrations were necessary in spheroidcultures. SIGNIFICANCE: This study provides pre-clinical evidence that single PI3K, FGFR, and CDK4/6, inhibitors exhibit promising anti-NB activity and when combined lower doses of the drugs could be also used in spheroid NB-cultures, supporting the pursuit of further in vitro and in vivo studies in preparation for future potential clinical use.


Sujet(s)
Kinase-4 cycline-dépendante , Kinase-6 cycline-dépendante , Synergie des médicaments , Neuroblastome , Inhibiteurs des phosphoinositide-3 kinases , Inhibiteurs de protéines kinases , Récepteur facteur croissance fibroblaste , Sphéroïdes de cellules , Neuroblastome/traitement médicamenteux , Neuroblastome/anatomopathologie , Humains , Kinase-4 cycline-dépendante/antagonistes et inhibiteurs , Kinase-4 cycline-dépendante/métabolisme , Sphéroïdes de cellules/effets des médicaments et des substances chimiques , Lignée cellulaire tumorale , Inhibiteurs des phosphoinositide-3 kinases/pharmacologie , Récepteur facteur croissance fibroblaste/antagonistes et inhibiteurs , Récepteur facteur croissance fibroblaste/métabolisme , Inhibiteurs de protéines kinases/pharmacologie , Kinase-6 cycline-dépendante/antagonistes et inhibiteurs , Kinase-6 cycline-dépendante/métabolisme , Prolifération cellulaire/effets des médicaments et des substances chimiques , Pyridines/pharmacologie , Thérapie moléculaire ciblée , Phosphatidylinositol 3-kinases/métabolisme , Survie cellulaire/effets des médicaments et des substances chimiques , Pipérazines/pharmacologie , Pipérazines/administration et posologie , Relation dose-effet des médicaments
18.
Int Immunopharmacol ; 133: 112145, 2024 May 30.
Article de Anglais | MEDLINE | ID: mdl-38691920

RÉSUMÉ

Treatment strategies for paediatric neuroblastoma as well as many other cancers are limited by the unfavourable tumour microenvironment (TME). In this study, the TMEs of neuroblastoma were grouped by their genetic signatures into four distinct subtypes: immune enriched, immune desert, non-proliferative and fibrotic. An Immune Score and a Proliferation Score were constructed based on the molecular features of the subtypes to quantify the immune microenvironment or malignancy degree of cancer cells in neuroblastoma, respectively. The Immune Score correlated with a patient's response to immunotherapy; the Proliferation Score was an independent prognostic biomarker for neuroblastoma and proved to be more accurate than the existing clinical predictors. This double scoring system was further validated and the conserved molecular pattern associated with immune landscape and malignancy degree was confirmed. Axitinib and BI-2536 were confirmed as candidate drugs for neuroblastoma by the double scoring system. Both in vivo and in vitro experiments demonstrated that axitinib-induced pyroptosis of neuroblastoma cells activated anti-tumour immunity and inhibited tumour growth; BI-2536 induced cell cycle arrest at the S phase in neuroblastoma cells. The comprehensive double scoring system of neuroblastoma may predict prognosis and screen for therapeutic strategies which could provide personalized treatments.


Sujet(s)
Axitinib , Immunothérapie , Neuroblastome , Microenvironnement tumoral , Neuroblastome/immunologie , Neuroblastome/thérapie , Neuroblastome/anatomopathologie , Neuroblastome/traitement médicamenteux , Humains , Microenvironnement tumoral/immunologie , Pronostic , Animaux , Immunothérapie/méthodes , Lignée cellulaire tumorale , Axitinib/usage thérapeutique , Enfant , Mâle , Femelle , Enfant d'âge préscolaire , Souris , Nourrisson , Tests d'activité antitumorale sur modèle de xénogreffe , Prolifération cellulaire/effets des médicaments et des substances chimiques
19.
Molecules ; 29(10)2024 May 14.
Article de Anglais | MEDLINE | ID: mdl-38792167

RÉSUMÉ

Methylglyoxal-induced ROS elevation is the primary cause of neuronal damage. Metformin is a traditional hypoglycemic drug that has been reported to be beneficial to the nervous system. In this study, flavonoids were found to enhance the protective effect of metformin when added at a molar concentration of 0.5%. The structure-activity relationship (SAR) analysis indicated that ortho- substitution in the B ring, and the absence of double bonds between the 2 and 3 position combined with the gallate substitution with R configuration at the 3 position in the C ring played crucial roles in the synergistic effects, which could be beneficial for designing a combination of the compounds. Additionally, the mechanism study revealed that a typical flavonoid, EGCG, enhanced ROS scavenging and anti-apoptotic ability via the BCL2/Bax/Cyto C/Caspase-3 pathway, and synergistically inhibited the expression of GSK-3ß, BACE-1, and APP in PC-12 cells when used in combination with metformin. The dose of metformin used in the combination was only 1/4 of the conventional dose when used alone. These results suggested that ROS-mediated apoptosis and the pathways related to amyloid plaques (Aß) formation can be the targets for the synergistic neuroprotective effects of flavonoids and metformin.


Sujet(s)
Apoptose , Synergie des médicaments , Flavonoïdes , Metformine , Méthylglyoxal , Espèces réactives de l'oxygène , Metformine/pharmacologie , Metformine/composition chimique , Rats , Flavonoïdes/pharmacologie , Flavonoïdes/composition chimique , Cellules PC12 , Animaux , Relation structure-activité , Apoptose/effets des médicaments et des substances chimiques , Espèces réactives de l'oxygène/métabolisme , Neuroblastome/métabolisme , Neuroblastome/anatomopathologie , Neuroblastome/traitement médicamenteux , Neuroprotecteurs/pharmacologie , Neuroprotecteurs/composition chimique , Transduction du signal/effets des médicaments et des substances chimiques
20.
FASEB J ; 38(10): e23689, 2024 May 31.
Article de Anglais | MEDLINE | ID: mdl-38785406

RÉSUMÉ

Neuroblastoma, a prevalent extracranial solid tumor in children, arises from undifferentiated nerve cells. While tumor vasculature, often characterized by increased permeability, influences metastasis and recurrence, the direct impact of blood-borne molecules on tumor progression remains unclear. In the present study, we focused on the effect of exposure to albumin, one of the most abundant proteins in the serum, on human neuroblastoma cells. Albumin exposure elevated oxidative stress and led to mitochondria dysfunction via the activation of TGFß and PI3K pathways, accompanied by an increase in the metastatic and invasive properties of neuroblastoma cells. Proteins relevant to the induction of autophagy were upregulated in response to prolonged albumin exposure. Additionally, pre-exposure to albumin before treatment resulted in increased resistance to paclitaxel. Two valeriana-type iridoid glycosides, patrisophoroside and patrinalloside, recently isolated from Nardostachys jatamansi significantly mitigated the effect of albumin on oxidative stress, cell invasiveness, and chemoresistance. These findings illuminate the potential role of blood-borne molecules, such as albumin, in the progression and metastasis of neuroblastoma, as well as the possible therapeutic implications of valeriana-type iridoid glycosides in anti-cancer treatment.


Sujet(s)
Résistance aux médicaments antinéoplasiques , Glycosides d'iridoïdes , Neuroblastome , Paclitaxel , Humains , Neuroblastome/anatomopathologie , Neuroblastome/métabolisme , Neuroblastome/traitement médicamenteux , Résistance aux médicaments antinéoplasiques/effets des médicaments et des substances chimiques , Paclitaxel/pharmacologie , Glycosides d'iridoïdes/pharmacologie , Lignée cellulaire tumorale , Invasion tumorale , Stress oxydatif/effets des médicaments et des substances chimiques , Antinéoplasiques d'origine végétale/pharmacologie , Valeriana/composition chimique , Sérumalbumine/métabolisme
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE