Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 9 de 9
Filtrer
Plus de filtres











Base de données
Gamme d'année
1.
Life (Basel) ; 13(11)2023 Nov 20.
Article de Anglais | MEDLINE | ID: mdl-38004374

RÉSUMÉ

GSK3ß is a promising target for treating various disease conditions, including myocardial ischemia-reperfusion injury (IR). This study investigated the potential of GSK3ß as a novel drug for managing IR in rats exposed to PM2.5 for 1 day and up to 21 days. Female Wistar rats were exposed to PM2.5 at a concentration of 250 µg/m3 for 3 h daily for either a single day or 21 days. After exposure, the isolated rat hearts underwent 30 min of ischemia followed by 60 min of reperfusion. GSK3ß inhibition effectively reduced IR injury in rat hearts from animals exposed to PM2.5 for 1 day but not in those exposed for 21 days. PM2.5 exposure disrupted the redox balance in mitochondria and reduced the gene expression of antioxidants (glutaredoxin and peroxiredoxin) and NRF2, which protects against oxidative stress. PM2.5 also impaired mitochondrial bioenergetics, membrane potential, and quality control, leading to mitochondrial stress. Importantly, PM2.5 increased the translocation of GSK3ß into mitochondria and compromised the overall mitochondrial function, particularly in the 21-day-exposed rat myocardium. The results indicate that extended exposure to PM2.5 leads to oxidative stress that disrupts mitochondrial function and diminishes the effectiveness of GSK3ß inhibitors in offering cardio-protection through mitochondria.

2.
Cells ; 12(16)2023 08 15.
Article de Anglais | MEDLINE | ID: mdl-37626874

RÉSUMÉ

Circulatory GSK3ß is recognized as a biomarker and therapeutic target for diseases, including myocardial diseases. However, its potential as a target for myocardial ischemia-reperfusion injury (IR) in the presence of PM2.5 exposure is unclear. Wistar rats underwent IR following either a 21-day or single exposure to PM2.5 at a concentration of 250 µg/m3. The effects of GSK3ß inhibitor on cardiac physiology, tissue injury, mitochondrial function, and the PI3K/AKT/GSK3ß signalling axis were examined. The inhibitor was not effective in improving hemodynamics or reducing IR-induced infarction in the myocardium exposed to PM2.5 for 21 days. However, for a single-day exposure, the inhibitor showed potential in mitigating cardiac injury. In normal hearts undergoing IR, the inhibitor activated the PI3K/AKT signalling pathway, improved mitochondrial function, and reduced oxidative stress. These positive effects were not observed in PM2.5-exposed rats. Furthermore, the inhibitor stimulated autophagy in hearts exposed to PM2.5 for 21 days and subjected to IR, resulting in increased mTOR expression and decreased AMPK expression. In normal hearts and those exposed to a single dose of PM2.5, the inhibitor effectively activated the PI3K/Akt/AMPK axis. These findings suggest that GSK3ß may not be a reliable therapeutic target for IR in the presence of chronic PM2.5 exposure.


Sujet(s)
Lésion de reperfusion myocardique , Rats , Animaux , Lésion de reperfusion myocardique/traitement médicamenteux , Glycogen synthase kinase 3 beta , Rat Wistar , AMP-Activated Protein Kinases , Phosphatidylinositol 3-kinases , Protéines proto-oncogènes c-akt , Matière particulaire/toxicité
3.
Life Sci ; 289: 120231, 2022 Jan 15.
Article de Anglais | MEDLINE | ID: mdl-34921867

RÉSUMÉ

AIMS: To investigate the role of Skp2 and JunB on acute promyelocytic leukemia (APL) progression and the related mechanism. MATERIALS AND METHODS: The expression of Skp2 in NB4 cell line was depleted to explore its effect on proliferation and differentiation both in vitro and in vivo assays. Western blot and quantitative RT-PCR analysis were performed to explore Skp2-regulated downstream target genes. Luciferase and co-immunoprecipitation analysis indicated that PML-RARα inhibited the transactivation of JunB by interacting with the PU.1 protein. The western blot analysis confirmed that Skp2 could maintain the stability of PML-RARα. KEY FINDINGS: We report that the progression of APL and the attenuation of APL sensitivity to ATRA are positively associated with Skp2. Elevated Skp2 expression promotes APL progression by decreasing the expression of lncRNA HOTAIRM1 and inactivation of GSK3ß, causing autophagy inhibition followed by the suppression of PML-RARα ubiquitylation and degradation, which represses JunB transcriptional activation through PU.1/PML-RARα transcriptional complex to block cell differentiation. Coupled with ATRA or GSK3ß inhibitor treatment, genetic or pharmacological inhibition of Skp2 strikingly induces JunB expression by accelerating the degradation of PML-RARα, which contributes to the eradication of APL. Additionally, the expressions of Skp2 and JunB are negatively correlated in mice subcutaneous leukemia xenograft tumors. SIGNIFICANCE: Collectively, this study uncovers the roles of Skp2 in PML-RARα stabilization and in APL oncogenic functions. We reveal a novel mechanism of PML-RARα degradation and JunB regulation that constitute an important signaling network of Skp2-GSK3ß-PML/RARα-JunB.


Sujet(s)
Régulation de l'expression des gènes dans la leucémie , Leucémie aiguë promyélocytaire/métabolisme , Protéines de fusion oncogènes/métabolisme , Protéines associées aux kinases de la phase S/métabolisme , Facteurs de transcription/biosynthèse , Activation de la transcription , Animaux , Cellules HEK293 , Humains , Leucémie aiguë promyélocytaire/traitement médicamenteux , Leucémie aiguë promyélocytaire/génétique , Leucémie aiguë promyélocytaire/anatomopathologie , Souris , Souris de lignée NOD , Souris SCID , Protéines de fusion oncogènes/génétique , Stabilité protéique , Protéines associées aux kinases de la phase S/génétique , Facteurs de transcription/génétique , Cellules U937 , Tests d'activité antitumorale sur modèle de xénogreffe
4.
ACS Chem Neurosci ; 12(8): 1328-1342, 2021 04 21.
Article de Anglais | MEDLINE | ID: mdl-33797877

RÉSUMÉ

Multitarget-directed ligands (MTDLs) are considered a promising therapeutic strategy to address the multifactorial nature of Alzheimer's disease (AD). Novel MTDLs have been designed as inhibitors of human acetylcholinesterases/butyrylcholinesterases, monoamine oxidase A/B, and glycogen synthase kinase 3ß and as calcium channel antagonists via the Biginelli multicomponent reaction. Among these MTDLs, (±)-BIGI-3h was identified as a promising new hit compound showing in vitro balanced activities toward the aforementioned recognized AD targets. Additional in vitro studies demonstrated antioxidant effects and brain penetration, along with the ability to inhibit the aggregation of both τ protein and ß-amyloid peptide. The in vivo studies have shown that (±)-BIGI-3h (10 mg/kg intraperitoneally) significantly reduces scopolamine-induced cognitive deficits.


Sujet(s)
Maladie d'Alzheimer , Maladie d'Alzheimer/traitement médicamenteux , Inhibiteurs des canaux calciques/pharmacologie , Inhibiteurs des canaux calciques/usage thérapeutique , Canaux calciques , Anticholinestérasiques/pharmacologie , Anticholinestérasiques/usage thérapeutique , Glycogen synthase kinase 3 beta , Humains , Ligands , Monoamine oxidase/métabolisme
5.
Oncol Rep ; 44(6): 2373-2385, 2020 12.
Article de Anglais | MEDLINE | ID: mdl-33125126

RÉSUMÉ

Glycogen synthase kinase 3ß (GSK 3ß), a multifunctional serine and threonine kinase, plays a critical role in a variety of cellular activities, including signaling transduction, protein and glycogen metabolism, cell proliferation, cell differentiation, and apoptosis. Therefore, aberrant regulation of GSK 3ß results in a broad range of human diseases, such as tumors, diabetes, inflammation and neurodegenerative diseases. Accumulating evidence has suggested that GSK 3ß is correlated with tumorigenesis and progression. However, GSK 3ß is controversial due to its bifacial roles of tumor suppression and activation. In addition, overexpression of GSK 3ß is involved in tumor growth, whereas it contributes to the cell sensitivity to chemotherapy. However, the underlying regulatory mechanisms of GSK 3ß in tumorigenesis remain obscure and require further in­depth investigation. In this review, we comprehensively summarize the roles of GSK 3ß in tumorigenesis and oncotherapy, and focus on its potentials as an available target in oncotherapy.


Sujet(s)
Protocoles de polychimiothérapie antinéoplasique/usage thérapeutique , Carcinogenèse/anatomopathologie , Glycogen synthase kinase 3 beta/métabolisme , Tumeurs/anatomopathologie , Facteurs de transcription/métabolisme , Animaux , Antinéoplasiques immunologiques/pharmacologie , Antinéoplasiques immunologiques/usage thérapeutique , Protocoles de polychimiothérapie antinéoplasique/pharmacologie , Apoptose/effets des médicaments et des substances chimiques , Carcinogenèse/effets des médicaments et des substances chimiques , Carcinogenèse/génétique , Modèles animaux de maladie humaine , Évolution de la maladie , Régulation de l'expression des gènes tumoraux/effets des médicaments et des substances chimiques , Régulation de l'expression des gènes tumoraux/immunologie , Glycogen synthase kinase 3 beta/antagonistes et inhibiteurs , Humains , Lymphocytes/effets des médicaments et des substances chimiques , Lymphocytes/enzymologie , Lymphocytes/immunologie , Souris , Tumeurs/traitement médicamenteux , Tumeurs/génétique , Tumeurs/immunologie , Phosphorylation/effets des médicaments et des substances chimiques , Phosphorylation/génétique , Phosphorylation/immunologie , Inhibiteurs de protéines kinases/pharmacologie , Inhibiteurs de protéines kinases/usage thérapeutique , Protéolyse/effets des médicaments et des substances chimiques , Transduction du signal/effets des médicaments et des substances chimiques
6.
Cancer Chemother Pharmacol ; 85(2): 379-390, 2020 02.
Article de Anglais | MEDLINE | ID: mdl-31832810

RÉSUMÉ

PURPOSE: 6-Gingerol, a major biochemical and pharmacological active ingredient of ginger, has shown anti-inflammatory and antitumor activities against various cancers. Searching for natural products with fewer side effects for developing adjunctive therapeutic options is necessary. METHODS: The effects of 6-gingerol on proliferation, colony formation, and cell cycle in RCC cells were detected by a 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide (MTT) assay, colony formation assay, and propidium iodide (PI) staining, respectively. Western blotting, an immunofluorescence assay, and immunohistochemical staining were performed to assess the expression of relevant proteins. A subcutaneous tumor model was set up to investigate the 6-gingerol effects on tumor growth in vivo, and the pharmacokinetics of 6-gingerol in mice were detected by LC/MS assays. RESULTS: 6-Gingerol treatment exerted time- and dose-dependent inhibition of the growth and colony formation of ACHN, 786-O, and 769-P cells, leading to a concomitant induction of cell-cycle G1-phase arrest and decrease in Ki-67 expression in the cell nucleus. Western-blotting results showed that 6-gingerol reduces phosphorylation of protein kinase B (AKT) Ser 473, cyclin-dependent kinases (CDK4), and cyclin D1 and, meanwhile, increases glycogen synthase kinase (GSK 3ß) protein amount. Furthermore, the efficacy of 6-gingerol was demonstrated in an in vivo murine model of 786-O. CONCLUSION: The above results indicate that 6-gingerol can induce cell-cycle arrest and cell-growth inhibition through the AKT-GSK 3ß-cyclin D1 signaling pathway in vitro and in vivo, suggesting that 6-gingerol should be useful for renal-cell carcinoma treatment.


Sujet(s)
Néphrocarcinome/traitement médicamenteux , Catéchols/pharmacologie , Cycline D1/métabolisme , Alcools gras/pharmacologie , Points de contrôle de la phase G1 du cycle cellulaire/effets des médicaments et des substances chimiques , Glycogen synthase kinase 3 beta/métabolisme , Tumeurs du rein/traitement médicamenteux , Protéines proto-oncogènes c-akt/métabolisme , Animaux , Néphrocarcinome/métabolisme , Lignée cellulaire tumorale , Prolifération cellulaire/effets des médicaments et des substances chimiques , Humains , Tumeurs du rein/métabolisme , Souris , Souris de lignée BALB C , Souris nude , Phosphorylation/effets des médicaments et des substances chimiques , Transduction du signal/effets des médicaments et des substances chimiques , bêta-Caténine/métabolisme
7.
BMC Cancer ; 18(1): 1168, 2018 Nov 26.
Article de Anglais | MEDLINE | ID: mdl-30477461

RÉSUMÉ

BACKGROUND: Wnt signaling has been linked with P-glycoprotein (P-gp) overexpression and which was mainly mediated by ß-catenin nuclear translocation. Flavonoids have already been reported as modulators of the Wnt/ß-catenin pathway and hence they may serve as promising agents in the reversal of P-gp mediated cancer multi drug resistance (MDR). METHODS: In this study, we screened selected flavonoids against Wnt/ß-catenin signaling molecules. The binding interaction of flavonoids (theaflavin, quercetin, rutin, epicatechin 3 gallate and tamarixetin) with GSK 3ß was determined by molecular docking. Flavonoids on P-gp expression and the components of Wnt signaling in drug-resistant KBCHR8-5 cells were analyzed by western blotting and qRT-PCR. The MDR reversal potential of these selected flavonoids against P-gp mediated drug resistance was analyzed by cytotoxicity assay in KBCHR8-5 and MCF7/ADR cell lines. The chemosensitizing potential of flavonoids was further analyzed by observing cell cycle arrest in KBCHR8-5 cells. RESULTS: In this study, we observed that the components of Wnt/ß-catenin pathway such as Wnt and GSK 3ß were activated in multidrug resistant KBCHR8-5 cell lines. All the flavonoids selected in this study significantly decreased the expression of Wnt and GSK 3ß in KBCHR8-5 cells and subsequently modulates P-gp overexpression in this drug-resistant cell line. Further, we observed that these flavonoids considerably decreased the doxorubicin resistance in KBCHR8-5 and MCF7/ADR cell lines. The MDR reversal potential of flavonoids were found to be in the order of theaflavin > quercetin > rutin > epicatechin 3 gallate > tamarixetin. Moreover, we observed that flavonoids pretreatment significantly induced the doxorubicin-mediated arrest at the phase of G2/M. Further, the combinations of doxorubicin with flavonoids significantly modulate the expression of drug response genes in KBCHR8-5 cells. CONCLUSION: The present findings illustrate that the studied flavonoids significantly enhances doxorubicin-mediated cell death through modulating P-gp expression pattern by targeting Wnt/ß-catenin signaling in drug-resistant KBCHR8-5 cells.


Sujet(s)
Glycoprotéine P/génétique , Multirésistance aux médicaments/génétique , Résistance aux médicaments antinéoplasiques/génétique , Flavonoïdes/pharmacologie , Expression des gènes , Voie de signalisation Wnt/effets des médicaments et des substances chimiques , Glycoprotéine P/composition chimique , Apoptose/effets des médicaments et des substances chimiques , Apoptose/génétique , Sites de fixation , Lignée cellulaire tumorale , Flavonoïdes/composition chimique , Glycogen synthase kinase 3 beta/composition chimique , Glycogen synthase kinase 3 beta/métabolisme , Humains , Modèles biologiques , Conformation moléculaire , Liaison aux protéines , Relation structure-activité
8.
Neurotox Res ; 34(3): 463-476, 2018 Oct.
Article de Anglais | MEDLINE | ID: mdl-29687202

RÉSUMÉ

Hesperidin, a flavanoglycone abundantly present in citrus fruits, is reported to have antioxidant, anti-inflammatory, and neuroprotective properties. Previous reports from our laboratory indicated the neuroprotective effect of hesperidin against aluminum chloride (AlCl3)-induced memory loss, acetylcholine esterase hyperactivity, oxidative stress, and enhanced expression of amyloid ß protein biosynthesis-related markers. However, their role on AlCl3-induced inflammation, caspase activation, Tau pathology, altered Akt/GSK 3ß signaling pathway, and Aß clearance marker has not yet been fully elucidated. Intraperitonial injection of AlCl3 (100 mg/kg body weight) for 60 days significantly elevated the expressions of insulin-degrading enzyme (IDE), cyclin-dependent kinase 5 (CDK 5), and phosphoTau (pTau); inflammatory markers such as glial fibrillary acidic protein (GFAP), ionized calcium-binding adapter molecule 1 (Iba-1), NF-kB, cyclooxygenase-2 (COX-2), interleukin (IL)-1ß, IL-4, IL-6, tumor necrosis factor-alpha (TNF-α), inducible nitric oxide synthase (iNOS); and apoptotic markers including cytosolic cytochrome c (cyto c), caspase-3, caspase-8, and caspase-9, and lowered expressions of mitochondrial cyto c, phospho-Akt (pAkt) and phospho-glycogen synthase kinase-3ß (pGSK-3ß) in the hippocampus and cortex. Co-administration of hesperidin to AlCl3 rats for 60 days significantly ameliorated the aluminum-induced pathological changes. The behavioral studies also supported the above findings. Our results imply that treatment with hesperidin might be a potent option for treating the symptoms of cognitive impairment in Alzheimer's disease by targeting its most prominent hallmarks.


Sujet(s)
Anti-inflammatoires/usage thérapeutique , Caspases/métabolisme , Encéphalite/traitement médicamenteux , Hespéridine/usage thérapeutique , Protéine oncogène v-akt/métabolisme , Transduction du signal/effets des médicaments et des substances chimiques , Facteurs âges , Chlorure d'aluminium , Composés de l'aluminium/toxicité , Animaux , Chlorures/toxicité , Cytokines/métabolisme , Modèles animaux de maladie humaine , Encéphalite/induit chimiquement , Mâle , Apprentissage du labyrinthe/effets des médicaments et des substances chimiques , Troubles de la mémoire/induit chimiquement , Troubles de la mémoire/traitement médicamenteux , Troubles mentaux/induit chimiquement , Troubles mentaux/traitement médicamenteux , Activité motrice/effets des médicaments et des substances chimiques , Rats , Rat Wistar , /effets des médicaments et des substances chimiques
9.
Open Biochem J ; 2: 67-76, 2008.
Article de Anglais | MEDLINE | ID: mdl-18949077

RÉSUMÉ

Modification of the Small Ubiquitin-like Modifier (SUMO) (SUMOylation) appears to regulate diverse cellular processes, including nuclear transport, signal transduction, apoptosis, autophagy, cell cycle control, ubiquitin-dependent degradation and gene transcription. Glycogen synthase kinase 3beta (GSK 3beta) is a serine/threonine kinase that is thought to contribute to a variety of biological events, including embryonic development, metabolism, tumorigenesis, and cell death. GSK 3beta is a constitutively active kinase that regulates many intracellular signaling pathways by phosphorylating substrates such as beta-catenin. We noticed that the putative SUMOylation sites are localized on K(292 )residueof (291)FKFPQ(295) in GSK 3beta based on analysis of the SUMOylation consensus sequence. In this report, we showed that the SUMOylation of GSK 3beta occurs on its K(292) residue, and this modification promotes its nuclear localization in COS-1. Additionally, our data showed that the GSK 3beta SUMO mutant (K292R) decreased its kinase activity and protein stability, affecting cell death. Therefore, our observations at first time suggested that SUMOylation on the K(292) residue of GSK 3beta might be a GSK 3beta regulation mechanism for its kinase activation, subcellular localization, protein stability, and cell apoptosis.

SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE