Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 20 de 2.962
Filtrer
1.
Chem Biol Interact ; 398: 111073, 2024 Aug 01.
Article de Anglais | MEDLINE | ID: mdl-38823538

RÉSUMÉ

Cancer is a complex and multifaceted group of diseases with a high mortality rate characterized by uncontrolled proliferation of abnormal cells. Dysregulation of normal signalling pathways in cancer contributes to the different hallmarks of this disease. The signalling pathway of which phosphatidylinositol 3-kinase (PI3K) is a part is not an exception. In fact, dysregulated activation of PI3K signalling pathways can result in unbridled cellular proliferation and enhanced cell survival, thereby fostering the onset and advancement of cancer. Therefore, there is substantial interest in developing targeted therapies specifically aimed at inhibiting the PI3K enzyme and its associated pathways. Also, the therapeutic interest on pyrazoles and indazoles has been growing due to their various medicinal properties, namely, anticancer activity. Derivatives of these compounds have been studied as PI3K inhibitors, and they showed promising results. There are already some PI3K inhibitors approved by Food and Drug Administration (FDA), such as Idelalisib (Zydelig®) and Alpelisib (Piqray®). In this context, this review aims to address the importance of PI3K in cellular processes and its role in cancer. Additionally, it aims to report a comprehensive literature review of PI3K inhibitors, containing the pyrazole and indazole scaffolds, published in the last fifteen years, focusing on structure-activity relationship aspects, thus providing important insights for the design of novel and more effective PI3K inhibitors.


Sujet(s)
Antinéoplasiques , Indazoles , Tumeurs , Inhibiteurs des phosphoinositide-3 kinases , Pyrazoles , Humains , Indazoles/composition chimique , Indazoles/pharmacologie , Indazoles/usage thérapeutique , Pyrazoles/composition chimique , Pyrazoles/pharmacologie , Pyrazoles/usage thérapeutique , Tumeurs/traitement médicamenteux , Inhibiteurs des phosphoinositide-3 kinases/pharmacologie , Inhibiteurs des phosphoinositide-3 kinases/composition chimique , Inhibiteurs des phosphoinositide-3 kinases/usage thérapeutique , Antinéoplasiques/composition chimique , Antinéoplasiques/pharmacologie , Antinéoplasiques/usage thérapeutique , Inhibiteurs de protéines kinases/composition chimique , Inhibiteurs de protéines kinases/pharmacologie , Inhibiteurs de protéines kinases/usage thérapeutique , Animaux , Phosphatidylinositol 3-kinases/métabolisme , Phosphatidylinositol 3-kinases/composition chimique , Phosphatidylinositol 3-kinase/métabolisme , Transduction du signal/effets des médicaments et des substances chimiques
2.
Medicine (Baltimore) ; 103(24): e38508, 2024 Jun 14.
Article de Anglais | MEDLINE | ID: mdl-38875362

RÉSUMÉ

Breast cancer is currently the most commonly occurring cancer globally. Among breast cancer cases, the human epidermal growth factor receptor 2 (HER2)-positive breast cancer accounts for 15% to 20% and is a crucial focus in the treatment of breast cancer. Common HER2-targeted drugs approved for treating early and/or advanced breast cancer include trastuzumab and pertuzumab, which effectively improve patient prognosis. However, despite treatment, most patients with terminal HER2-positive breast cancer ultimately suffer death from the disease due to primary or acquired drug resistance. The prevalence of aberrantly activated the protein kinase B (AKT) signaling in HER2-positive breast cancer was already observed in previous studies. It is well known that p-AKT expression is linked to an unfavorable prognosis, and the phosphatidylinositol-3-kinase (PI3K)/AKT pathway, as the most common mutated pathway in breast cancer, plays a major role in the mechanism of drug resistance. Therefore, in the current review, we summarize the molecular alterations present in HER2-positive breast cancer, elucidate the relationships between HER2 overexpression and alterations in the PI3K/AKT signaling pathway and the pathways of the alterations in breast cancer, and summarize the resistant mechanism of drugs targeting the HER2-AKT pathway, which will provide an adjunctive therapeutic rationale for subsequent resistance to directed therapy in the future.


Sujet(s)
Tumeurs du sein , Résistance aux médicaments antinéoplasiques , Protéines proto-oncogènes c-akt , Récepteur ErbB-2 , Transduction du signal , Humains , Tumeurs du sein/traitement médicamenteux , Tumeurs du sein/génétique , Tumeurs du sein/métabolisme , Récepteur ErbB-2/métabolisme , Récepteur ErbB-2/génétique , Femelle , Protéines proto-oncogènes c-akt/métabolisme , Résistance aux médicaments antinéoplasiques/génétique , Phosphatidylinositol 3-kinases/métabolisme , Antinéoplasiques/usage thérapeutique , Phosphatidylinositol 3-kinase/métabolisme
3.
Cell Adh Migr ; 18(1): 1-12, 2024 Dec.
Article de Anglais | MEDLINE | ID: mdl-38831518

RÉSUMÉ

In this research, we investigated the role of PIK3R6, a regulatory subunit of PI3Kγ, known for its tumor-promoting properties, in clear cell renal cell carcinoma (CCRCC). Utilizing the UALCAN website, we found PIK3R6 upregulated in CCRCC, correlating with lower survival rates. We compared PIK3R6 expression in CCRCC tumor tissues and adjacent normal tissues using immunohistochemistry. Post RNA interference-induced knockdown of PIK3R6 in 786-O and ACHN cell lines, we performed CCK-8, colony formation, Edu staining, flow cytometry, wound healing, and transwell assays. Results showed that PIK3R6 silencing reduced cell proliferation, migration, and invasion, and induced G0/G1 phase arrest and apoptosis. Molecular analysis revealed decreased CDK4, Cyclin D1, N-cadherin, Vimentin, Bcl-2, p-PI3K and p-AKT, with increased cleaved caspase-3, Bax, and E-cadherin levels in CCRCC cells. Moreover, inhibiting PIK3R6 hindered tumor growth. These findings suggest a significant role for PIK3R6 in CCRCC cell proliferation and metastasis, presenting it as a potential therapeutic target.


Sujet(s)
Apoptose , Néphrocarcinome , Mouvement cellulaire , Prolifération cellulaire , Tumeurs du rein , Animaux , Femelle , Humains , Mâle , Souris , Apoptose/génétique , Néphrocarcinome/génétique , Néphrocarcinome/anatomopathologie , Néphrocarcinome/métabolisme , Lignée cellulaire tumorale , Mouvement cellulaire/génétique , Prolifération cellulaire/génétique , Régulation de l'expression des gènes tumoraux , Techniques de knock-down de gènes , Tumeurs du rein/génétique , Tumeurs du rein/anatomopathologie , Tumeurs du rein/métabolisme , Souris nude , Phosphatidylinositol 3-kinase/métabolisme
4.
Am J Physiol Cell Physiol ; 327(1): C65-C73, 2024 Jul 01.
Article de Anglais | MEDLINE | ID: mdl-38766766

RÉSUMÉ

The blood-brain barrier (BBB) plays a critical role in the development and outcome of subarachnoid hemorrhage (SAH). This study focuses on the potential mechanism by which G-protein-coupled estrogen receptor 30 (GPR30) affects the BBB after SAH. A rat SAH model was established using an intravascular perforation approach. G1 (GPR30 agonist) was administered to investigate the mechanism of BBB damage after SAH. Brain water content, Western blotting, Evans blue leakage, and immunofluorescence staining were performed. Brain microvascular endothelial cells were induced by hemin to establish SAH model in vitro. By adding LY294002 [a phosphatidylinositol 3-kinase (PI3K) blocker] and zinc protoporphyrin IX (ZnPP IX) [a heme oxygenase 1 (HO-1) antagonist], the mechanism of improving BBB integrity through the activation of GPR30 was studied. In vivo, GPR30 activation improved BBB disruption, as evidenced by decreased cerebral edema, downregulated albumin expression, and reduced extravasation of Evans blue and IgG after G1 administration in SAH rats. Moreover, SAH downregulated the levels of tight junction (TJ) proteins, whereas treatment with G1 reversed the effect of SAH. The protective effect of G1 on BBB integrity in vitro was consistent with that in vivo, as evidenced by G1 reducing the impact of hemin on transendothelial electrical resistance (TEER) value, dextran diffusivity, and TJ protein levels in brain microvascular endothelial cells. In addition, G1 activated the PI3K/ protein kinase B (Akt) and nuclear factor erythroid 2-related factor 2 (Nrf2)/HO-1 pathways both in vivo and in vitro. Furthermore, the administration of LY294002 and ZnPP IX partially reversed the protective effect of G1 on BBB integrity in hemin-stimulated cells. We demonstrated that the activation of GPR30, at least partly through the PI3K/Akt and Nrf2/HO-1 pathways, alleviated BBB damage both in vivo and in vitro. This study introduced a novel therapeutic approach for protecting the BBB after SAH.NEW & NOTEWORTHY The PI3K/Akt and Nrf2/HO-1 pathways might be potential mechanisms by which GPR30 protected the integrity of the BBB in SAH models. Therefore, treatment of SAH with GPR30 activator might be a promising therapeutic strategy.


Sujet(s)
Barrière hémato-encéphalique , Facteur-2 apparenté à NF-E2 , Phosphatidylinositol 3-kinases , Protéines proto-oncogènes c-akt , Rat Sprague-Dawley , Récepteurs couplés aux protéines G , Transduction du signal , Hémorragie meningée , Animaux , Facteur-2 apparenté à NF-E2/métabolisme , Barrière hémato-encéphalique/métabolisme , Barrière hémato-encéphalique/effets des médicaments et des substances chimiques , Barrière hémato-encéphalique/anatomopathologie , Protéines proto-oncogènes c-akt/métabolisme , Récepteurs couplés aux protéines G/métabolisme , Récepteurs couplés aux protéines G/agonistes , Hémorragie meningée/métabolisme , Hémorragie meningée/anatomopathologie , Hémorragie meningée/traitement médicamenteux , Hémorragie meningée/complications , Mâle , Rats , Phosphatidylinositol 3-kinases/métabolisme , Cellules endothéliales/métabolisme , Cellules endothéliales/effets des médicaments et des substances chimiques , Heme oxygenase (decyclizing)/métabolisme , Heme oxygenase-1/métabolisme , Modèles animaux de maladie humaine , Phosphatidylinositol 3-kinase/métabolisme , Hémine/pharmacologie
5.
Anticancer Res ; 44(6): 2533-2544, 2024 Jun.
Article de Anglais | MEDLINE | ID: mdl-38821596

RÉSUMÉ

BACKGROUND/AIM: Chemotherapy is mainly used in the clinical treatment of prostate cancer. Different anticancer mechanisms can induce cell death in various cancers. Reactive oxygen species (ROS) play crucial roles in cell proliferation, differentiation, apoptosis, and signal transduction. It is widely accepted that ROS accumulation is closely related to chemical drug-induced cancer cell death. MATERIALS AND METHODS: We utilized the MTT assay to detect changes in cell proliferation. Additionally, colony formation and wound healing assay were conducted to investigate the effect of hispidin on cell colony formation and migration ability. Fluorescence microscopy was used to detect intracellular and mitochondrial ROS levels, while western blot was used for detection of cell apoptosis. RESULTS: Hispidin treatment significantly decreased viability of PC3 and DU145 cancer cells but exhibited no cytotoxicity in WPMY-1 cells. Furthermore, hispidin treatment inhibited cell migration and colony formation and triggered cellular and mitochondrial ROS accumulation, leading to mitochondrial dysfunction and mitochondrion-dependent apoptosis. Moreover, hispidin treatment induced ferroptosis in PC3 cells. Scavenging of ROS with N-acetyl cysteine significantly inhibited hispidin-induced apoptosis by altering the expression of apoptosis-related proteins, such as cleaved caspase-3, 9, Bax, and Bcl2. Furthermore, hispidin treatment dramatically up-regulated MAPK (involving p38, ERK, and JNK proteins) and NF-kB signaling pathways while down-regulating AKT phosphorylation. Hispidin treatment also inhibited ferroptosis signaling pathways (involving P53, Nrf-2, and HO-1 proteins) in PC3 cells. In addition, inhibiting these signaling pathways via treatment with specific inhibitors significantly reversed hispidin-induced apoptosis, cellular ROS levels, mitochondrial dysfunction, and ferroptosis. CONCLUSION: Hispidin may represent a potential candidate for treating prostate cancer.


Sujet(s)
Apoptose , Ferroptose , Tumeurs de la prostate , Espèces réactives de l'oxygène , Humains , Mâle , Ferroptose/effets des médicaments et des substances chimiques , Tumeurs de la prostate/anatomopathologie , Tumeurs de la prostate/métabolisme , Tumeurs de la prostate/traitement médicamenteux , Apoptose/effets des médicaments et des substances chimiques , Espèces réactives de l'oxygène/métabolisme , Lignée cellulaire tumorale , Prolifération cellulaire/effets des médicaments et des substances chimiques , Système de signalisation des MAP kinases/effets des médicaments et des substances chimiques , Mouvement cellulaire/effets des médicaments et des substances chimiques , Transduction du signal/effets des médicaments et des substances chimiques , Mitochondries/effets des médicaments et des substances chimiques , Mitochondries/métabolisme , Pyridones/pharmacologie , Phosphatidylinositol 3-kinase/métabolisme , Pyrones
6.
J Ethnopharmacol ; 332: 118366, 2024 Oct 05.
Article de Anglais | MEDLINE | ID: mdl-38763371

RÉSUMÉ

ETHNOPHARMACOLOGICAL RELEVANCE: Siwu decoction (SWD) is widely used in gynecological diseases, such as peripheral menopause syndrome, premature ovarian failure, and menstrual disorder. However, the mechanism of SWD on postmenopausal osteoporosis (PMOP) remains unclear. AIM OF THE STUDY: To discover the phytoestrogenic osteoprotective effect of SWD on PMOP. MATERIALS AND METHODS: The potential mechanism of SWD on PMOP was filtered through network pharmacology research. The potential mechanism was verified in MC3T3-E1 cell lines in vitro. CCK8 assay was conducted to assess cell proliferation and the expressions of ER/PI3K/AKT pathway were analyzed using Western blot. Female F-344 rats were chosen to set up the PMOP model. The osteoprotective effect of SWD in vivo was evaluated using Hematoxylin-eosin staining, TRAP staining, Goldner staining and DXA. The potential mechanism was verified in vivo through Western blot and immunohistochemistry. RT-qPCR was conducted to unveil the expressions of osteogenesis genes. RESULTS: Network pharmacology research showed that ER/PI3K/AKT pathway may be the potential mechanism of SWD on PMOP. SWD promoted the proliferation of osteoblasts and regulated the protein expressions of ER/PI3K/AKT pathway in vitro. SWD improved the morphological structure, bone mineralization and bone mineral density of femurs and suppressed osteoclastogenesis in PMOP rat model via ER/PI3K/AKT pathway in vivo. In addition, SWD regulated the mRNA expressions of osteogenesis-related genes. CONCLUSIONS: SWD exerts a phytoestrogenic osteoprotective on PMOP by regulating ER/PI3K/AKT pathway, which marks it as a valuable medicine or supplement of PMOP.


Sujet(s)
Prolifération cellulaire , Médicaments issus de plantes chinoises , Ostéoporose post-ménopausique , Phyto-oestrogènes , Protéines proto-oncogènes c-akt , Récepteurs des oestrogènes , Transduction du signal , Animaux , Femelle , Médicaments issus de plantes chinoises/pharmacologie , Ostéoporose post-ménopausique/traitement médicamenteux , Ostéoporose post-ménopausique/prévention et contrôle , Souris , Transduction du signal/effets des médicaments et des substances chimiques , Récepteurs des oestrogènes/métabolisme , Rats , Protéines proto-oncogènes c-akt/métabolisme , Phyto-oestrogènes/pharmacologie , Phyto-oestrogènes/usage thérapeutique , Prolifération cellulaire/effets des médicaments et des substances chimiques , Phosphatidylinositol 3-kinase/métabolisme , Ostéogenèse/effets des médicaments et des substances chimiques , Lignée cellulaire , Humains , Densité osseuse/effets des médicaments et des substances chimiques , Ostéoblastes/effets des médicaments et des substances chimiques , Ostéoblastes/métabolisme , Phosphatidylinositol 3-kinases/métabolisme , Modèles animaux de maladie humaine , Pharmacologie des réseaux
7.
J Ethnopharmacol ; 330: 118264, 2024 Aug 10.
Article de Anglais | MEDLINE | ID: mdl-38692417

RÉSUMÉ

ETHNOPHARMACOLOGICAL RELEVANCE: Optimized New Shengmai Powder (ONSMP) is a sophisticated traditional Chinese medicinal formula renowned for bolstering vital energy, optimizing blood circulation, and mitigating fluid retention. After years of clinical application, ONSMP has shown a significant impact in improving myocardial injury and cardiac function and has a positive effect on treating heart failure. However, many unknowns exist about the molecular biological mechanisms of how ONSMP exerts its therapeutic effects, which require further research and exploration. AIM OF THE STUDY: Exploring the potential molecular biological mechanisms by which ONSMP ameliorates cardiomyocyte apoptosis and ferroptosis in ischemic heart failure (IHF). MATERIALS AND METHODS: First, we constructed a rat model of IHF by inducing acute myocardial infarction through surgery and using echocardiography, organ coefficients, markers of heart failure, antioxidant markers, and histopathological examination to assess the effects of ONSMP on cardiomyocyte apoptosis and ferroptosis in IHF rats. Next, we used bioinformatics analysis techniques to analyze the active components, signaling pathways, and core targets of ONSMP and calculated the interactions between core targets and corresponding elements. Finally, we detected the positive expression of apoptosis and ferroptosis markers and core indicators of signaling pathways by immunohistochemistry; detected the mean fluorescence intensity of core indicators of signaling pathways by immunofluorescence; detected the protein expression of signaling pathways and downstream effector molecules by western blotting; and detected the mRNA levels of p53 and downstream effector molecules by quantitative polymerase chain reaction. RESULTS: ONSMP can activate the Ser83 site of ASK by promoting the phosphorylation of the PI3K/AKT axis, thereby inhibiting the MKK3/6-p38 axis and the MKK4/7-JNK axis signaling to reduce p53 expression, and can also directly target and inhibit the activity of p53, ultimately inhibiting p53-mediated mRNA and protein increases in PUMA, SAT1, PIG3, and TFR1, as well as mRNA and protein decreases in SLC7A11, thereby inhibiting cardiomyocyte apoptosis and ferroptosis, effectively improving cardiac function and ventricular remodeling in IHF rat models. CONCLUSION: ONSMP can inhibit cardiomyocyte apoptosis and ferroptosis through the PI3K/AKT/p53 signaling pathway, delaying the development of IHF.


Sujet(s)
Apoptose , Médicaments issus de plantes chinoises , Ferroptose , Défaillance cardiaque , Myocytes cardiaques , Transduction du signal , Animaux , Mâle , Rats , Apoptose/effets des médicaments et des substances chimiques , Modèles animaux de maladie humaine , Médicaments issus de plantes chinoises/pharmacologie , Ferroptose/effets des médicaments et des substances chimiques , Défaillance cardiaque/traitement médicamenteux , Ischémie myocardique/traitement médicamenteux , Myocytes cardiaques/effets des médicaments et des substances chimiques , Myocytes cardiaques/métabolisme , Phosphatidylinositol 3-kinase/métabolisme , Poudres , Protéines proto-oncogènes c-akt/métabolisme , Rat Sprague-Dawley , Transduction du signal/effets des médicaments et des substances chimiques , Protéine p53 suppresseur de tumeur/métabolisme , Protéine p53 suppresseur de tumeur/génétique
8.
BMC Cardiovasc Disord ; 24(1): 280, 2024 May 29.
Article de Anglais | MEDLINE | ID: mdl-38811893

RÉSUMÉ

BACKGROUND: Myocardial ischemia-reperfusion injury (I/RI) is a major cause of perioperative cardiac-related adverse events and death. Studies have shown that sevoflurane postconditioning (SpostC), which attenuates I/R injury and exerts cardioprotective effects, regulates mitochondrial dynamic balance via HIF-1α, but the exact mechanism is unknown. This study investigates whether the PI3K/AKT pathway in SpostC regulates mitochondrial dynamic balance by mediating HIF-1α, thereby exerting myocardial protective effects. METHODS: The H9C2 cardiomyocytes were cultured to establish the hypoxia-reoxygenation (H/R) model and randomly divided into 4 groups: Control group, H/R group, sevoflurane postconditioning (H/R + SpostC) group and PI3K/AKT blocker (H/R + SpostC + LY) group. Cell survival rate was determined by CCK-8; Apoptosis rate was determined by flow cytometry; mitochondrial membrane potential was evaluated by Mito Tracker™ Red; mRNA expression levels of AKT, HIF-1α, Opa1and Drp1 were detected by quantitative real-time polymerase chain reaction (qRT-PCR); Western Blot assay was used to detect the protein expression levels of AKT, phosphorylated AKT (p-AKT), HIF-1α, Opa1 and Drp1. RESULTS: Compared with the H/R group, the survival rate of cardiomyocytes in the H/R + SpostC group increased, the apoptosis rate decreased and the mitochondrial membrane potential increased. qRT-PCR showed that the mRNA expression of HIF-1α and Opa1 were higher in the H/R + SpostC group compared with the H/R group, whereas the transcription level of Drp1 was lower in the H/R + SpostC group. In the H/R + SpostC + LY group, the mRNA expression of HIF-1α was lower than the H/R + SpostC group. There was no difference in the expression of Opa1 mRNA between the H/R group and the H/R + SpostC + LY group. WB assay results showed that compared with the H/R group, the protein expression levels of HIF-1α, Opa1, P-AKT were increased and Drp1 protein expression levels were decreased in the H/R + SpostC group. HIF-1α, P-AKT protein expression levels were decreased in the H/R + SpostC + LY group compared to the H/R + SpostC group. CONCLUSION: SpostC mediates HIF-1α-regulated mitochondrial fission and fusion-related protein expression to maintain mitochondrial dynamic balance by activating the PI3K/AKT pathway and increasing AKT phosphorylation, thereby attenuating myocardial I/R injury.


Sujet(s)
Apoptose , Sous-unité alpha du facteur-1 induit par l'hypoxie , Potentiel de membrane mitochondriale , Mitochondries du myocarde , Dynamique mitochondriale , Lésion de reperfusion myocardique , Myocytes cardiaques , Phosphatidylinositol 3-kinase , Protéines proto-oncogènes c-akt , Sévoflurane , Transduction du signal , Sous-unité alpha du facteur-1 induit par l'hypoxie/métabolisme , Sous-unité alpha du facteur-1 induit par l'hypoxie/génétique , Protéines proto-oncogènes c-akt/métabolisme , Animaux , Myocytes cardiaques/effets des médicaments et des substances chimiques , Myocytes cardiaques/anatomopathologie , Myocytes cardiaques/métabolisme , Myocytes cardiaques/enzymologie , Sévoflurane/pharmacologie , Lésion de reperfusion myocardique/anatomopathologie , Lésion de reperfusion myocardique/métabolisme , Lésion de reperfusion myocardique/prévention et contrôle , Lésion de reperfusion myocardique/génétique , Lésion de reperfusion myocardique/enzymologie , Dynamique mitochondriale/effets des médicaments et des substances chimiques , Lignée cellulaire , Rats , Apoptose/effets des médicaments et des substances chimiques , Phosphatidylinositol 3-kinase/métabolisme , Mitochondries du myocarde/effets des médicaments et des substances chimiques , Mitochondries du myocarde/métabolisme , Mitochondries du myocarde/anatomopathologie , Mitochondries du myocarde/enzymologie , Potentiel de membrane mitochondriale/effets des médicaments et des substances chimiques , Hypoxie cellulaire , Dynamines/métabolisme , Dynamines/génétique , dGTPases/métabolisme , dGTPases/génétique , Inhibiteurs des phosphoinositide-3 kinases/pharmacologie , Cytoprotection , Postconditionnement ischémique , Phosphorylation
9.
BMC Cardiovasc Disord ; 24(1): 275, 2024 May 28.
Article de Anglais | MEDLINE | ID: mdl-38807081

RÉSUMÉ

BACKGROUND: Autophagy, as a regulator of cell survival, plays an important role in atherosclerosis (AS). Sperm associated antigen 5 (SPAG5) is closely associated with the classical autophagy pathway, PI3K/Akt/mTOR signaling pathway. This work attempted to investigate whether SPAG5 can affect AS development by regulating autophagy. METHODS: Human umbilical vein endothelial cells (HUVECs) were treated with oxidized-low density lipoprotein (ox-LDL) to induce cell damage. ApoE-/- mice were fed a Western diet to establish an AS mouse model. Haematoxylin and eosin (H&E) staining and Oil Red O staining evaluated the pathological changes and in lipid deposition in aortic tissues. CCK-8 and flow cytometry detected cell proliferation and apoptosis. Immunohistochemistry, Enzyme linked immunosorbent assay, qRT-PCR and western blotting assessed the levels of mRNA and proteins. RESULTS: Ox-LDL treatment elevated SPAG5 expression and the expression of autophagy-related proteins, LC3-I, LC3-II, Beclin-1, and p62, in HUVECs. GFP-LC3 dots were increased in ox-LDL-treated HUVECs and LPS-treated HUVECs. SPAG5 knockdown reversed both ox-LDL and LPS treatment-mediated inhibition of cell proliferation and promotion of apoptosis in HUVECs. SPAG5 silencing further elevated autophagy and repressed the expression of PI3K, p-Akt/Akt, and p-mTOR/mTOR in ox-LDL-treated HUVECs. 3-MA (autophagy inhibitor) treatment reversed SPAG5 silencing-mediated increase of cell proliferation and decrease of apoptosis in ox-LDL-treated HUVECs. In vivo, SPAG5 knockdown reduced atherosclerotic plaques in AS mice through activating autophagy and inhibiting PI3K/Akt/mTOR signaling pathway. CONCLUSION: This work demonstrated that SPAG5 knockdown alleviated AS development through activating autophagy. Thus, SPAG5 may be a potential target for AS therapy.


Sujet(s)
Apoptose , Athérosclérose , Autophagie , Prolifération cellulaire , Modèles animaux de maladie humaine , Cellules endothéliales de la veine ombilicale humaine , Souris invalidées pour les gènes ApoE , Plaque d'athérosclérose , Protéines proto-oncogènes c-akt , Transduction du signal , Sérine-thréonine kinases TOR , Animaux , Autophagie/effets des médicaments et des substances chimiques , Cellules endothéliales de la veine ombilicale humaine/métabolisme , Cellules endothéliales de la veine ombilicale humaine/anatomopathologie , Humains , Athérosclérose/anatomopathologie , Athérosclérose/métabolisme , Athérosclérose/génétique , Athérosclérose/prévention et contrôle , Sérine-thréonine kinases TOR/métabolisme , Apoptose/effets des médicaments et des substances chimiques , Protéines proto-oncogènes c-akt/métabolisme , Prolifération cellulaire/effets des médicaments et des substances chimiques , Maladies de l'aorte/anatomopathologie , Maladies de l'aorte/génétique , Maladies de l'aorte/prévention et contrôle , Maladies de l'aorte/métabolisme , Souris de lignée C57BL , Lipoprotéines LDL/métabolisme , Mâle , Cellules cultivées , Protéines associées à l'autophagie/métabolisme , Protéines associées à l'autophagie/génétique , Aorte/anatomopathologie , Aorte/métabolisme , Phosphatidylinositol 3-kinase/métabolisme , Protéines du cycle cellulaire/métabolisme , Protéines du cycle cellulaire/génétique , Souris , Apolipoprotéines E
10.
BMC Cardiovasc Disord ; 24(1): 236, 2024 May 05.
Article de Anglais | MEDLINE | ID: mdl-38705985

RÉSUMÉ

BACKGROUND: This study was designed to investigate the mechanism by which miR-30a-5p mediates cardiomyocyte apoptosis after acute myocardial infarction (AMI) induced by hypoxia/reoxygenation (H/R). METHODS: Differentially expressed miRNAs were analyzed by RNA high-throughput sequencing in acute myocardial infarction (ST-elevation myocardial infarction) patients versus healthy individuals (controls). The H/R model was used to assess the regulatory mechanism of miRNAs in AMI. Lentivirus-associated vectors were used to overexpress or knock down miR-30a-5p in cellular models. The pathological mechanisms of miR-30a-5p regulating the development of acute myocardial infarction were serially explored by qPCR, bioinformatics, target gene prediction, dual luciferase, enzyme-linked immunosorbent assays (ELISAs) and Western blotting. RESULTS: The results showed that the expression of miR-30a-5p was significantly increased in AMI patients and H9C2 cells. Hypoxia decreased cardiomyocyte survival over time, and reoxygenation further reduced cell survival. Bax and Phosphatase and tensin homolog (PTEN)were suppressed, while Bcl-2 was upregulated. Additionally, miR-30a-5p specifically targeted the PTEN gene. According to the GO and KEGG analyses, miR-30a-5p may participate in apoptosis by interacting with PTEN. The miR-30a-5p mimic decreased the expression of apoptosis-related proteins and the levels of the proinflammatory markers IL-1ß, IL-6, and TNF-α by activating the PTEN/PI3K/Akt signaling pathway. Conversely, anti-miR-30a-5p treatment attenuated these effects. Additionally, silencing PTEN and anti-miR-30a-5p had opposite effects on H/R-induced cell apoptosis. CONCLUSIONS: miR-30a-5p plays a crucial role in cardiomyocyte apoptosis after hypoxia-induced acute myocardial infarction. Our findings provide translational evidence that miR-30a-5p is a novel potential therapeutic target for AMI.


Sujet(s)
Apoptose , Hypoxie cellulaire , microARN , Myocytes cardiaques , Phosphohydrolase PTEN , Transduction du signal , Animaux , Femelle , Humains , Mâle , Adulte d'âge moyen , Rats , Études cas-témoins , Lignée cellulaire , Régulation de l'expression des gènes , microARN/génétique , microARN/métabolisme , Infarctus du myocarde/génétique , Infarctus du myocarde/anatomopathologie , Infarctus du myocarde/métabolisme , Lésion de reperfusion myocardique/génétique , Lésion de reperfusion myocardique/anatomopathologie , Lésion de reperfusion myocardique/métabolisme , Lésion de reperfusion myocardique/enzymologie , Myocytes cardiaques/anatomopathologie , Myocytes cardiaques/métabolisme , Myocytes cardiaques/enzymologie , Phosphatidylinositol 3-kinase/métabolisme , Protéines proto-oncogènes c-akt/métabolisme , Phosphohydrolase PTEN/métabolisme , Phosphohydrolase PTEN/génétique
11.
J Physiol Pharmacol ; 75(1)2024 02.
Article de Anglais | MEDLINE | ID: mdl-38583438

RÉSUMÉ

Kinetochore scaffold 1 (KNL1) is indispensable for generating motile micro-tubule attachments and isolating chromosomes. KNL1 is highly expressed in multiple middle-route tissues and promotes tumor development. However, how it functions in non-small cell lung cancer (NSCLC) is unclear. Real-time quantitative PCR (RT-qPCR) and Western blotting (WB) were used to determine KNL1 expression in NSCLC tissues and cells. The sh-KNL1 or oe-KNL1 was transfected into NSCLC cells. The colony formation assay, cell counting kit-8 (CCK-8) assay, and flow cytometry were used to evaluate cell proliferation and apoptosis. A transwell assay was used to monitor invasion and migration. The CCK-8 assay was used to measure NSCLC cell sensitivity to chemotherapy drugs. WB confirmed the protein levels of apoptosis-related proteins, cell cycle-associated proteins, and the phosphatidylinositol 3 kinase (PI3K)/protein kinase B (AKT)/nuclear factor kappaB (NF-κB) pathway. A PI3K/AKT/NF-κB pathway inhibitor was used to intervene in NSCLC cell transfection along with oe-KNL1, thus revealing the function of the pathway in carcinogenicity mediated by KNL1. In result KNL1 expression was substantially increased in NSCLC tissues and cells. High-level KNL1 expression is related to the poor prognosis of NSCLC patients. KNL1 silencing bolstered promoted NSCLC cell apoptosis and inhibited proliferation, cell cycle progression, invasion, and EMT, whereas KNL1 silencing had the opposite effect. KNL1 knockdown increased NSCLC cell sensitivity to chemical drugs. KNL1 promoted PI3K/AKT/NF-κB pathway activation, while PI3K/AKT/NF-κB pathway inhibition weakened the procancer effect mediated by KNL1 overexpression but had little influence on KNL1 levels. We conclude that KNL1 activates the PI3K/AKT/NF-κB pathway to increase NSCLC progression and attenuate NSCLC sensitivity to chemotherapy drugs.


Sujet(s)
Carcinome pulmonaire non à petites cellules , Tumeurs du poumon , Humains , Protéines régulatrices de l'apoptose/métabolisme , Carcinome pulmonaire non à petites cellules/génétique , Carcinome pulmonaire non à petites cellules/métabolisme , Carcinome pulmonaire non à petites cellules/anatomopathologie , Lignée cellulaire tumorale , Prolifération cellulaire , Régulation négative , Kinétochores/métabolisme , Kinétochores/anatomopathologie , Tumeurs du poumon/génétique , Tumeurs du poumon/anatomopathologie , Facteur de transcription NF-kappa B/métabolisme , Phosphatidylinositol 3-kinase/génétique , Phosphatidylinositol 3-kinase/métabolisme , Phosphatidylinositol 3-kinase/pharmacologie , Phosphatidylinositol 3-kinases/métabolisme , Protéines proto-oncogènes c-akt/métabolisme
12.
Arch Esp Urol ; 77(2): 183-192, 2024 Mar.
Article de Anglais | MEDLINE | ID: mdl-38583011

RÉSUMÉ

PURPOSE: This study aimed to determine the influence of miR-1297 on kidney injury in rats with diabetic nephropathy (DN) and its causal role. METHODS: A DN rat model was established through right kidney resection and intraperitoneal injection of streptozotocin (STZ). Sham rats did not undergo right kidney resection or STZ injection. The DN rats were divided into the DN model and antagomiR-1297 treatment groups. Kidney morphology was observed using hematoxylin and eosin staining. Renal function indices, including blood urea nitrogen (BUN), serum creatinine (SCr), and urinary protein, were measured using kits. Levels of tumor necrosis factor-α (TNF-α), interleukin (IL)-6, IL-1ß, superoxide dismutase (SOD), and glutathione peroxidase (GSH-Px) were determined through enzyme-linked immunosorbent assay (ELISA). Fibrin (FN), collagen type I (Col I), and α-smooth muscle actin (α-SMA) were assessed through western blotting and real-time reverse transcription-polymerase chain reaction. Apoptosis was detected using terminal deoxynucleotidyl transferase dUTP nick end labeling staining. miR-1297 targets were predicted using bioinformatic software and verified through luciferase reporter assay. Phosphatase and tensin homolog deleted on chromosome 10 (PTEN)/phosphoinositide 3-kinase (PI3K)/protein kinase B (AKT) pathway expression was analyzed through western blotting. RESULTS: AntagomiR-1297 reduced BUN (p = 0.005), SCr (p = 0.012), and urine protein (p < 0.001) levels and improved kidney tissue morphology. It prevented renal interstitial fibrosis by decreasing FN, Col I, and α-SMA protein levels (all p < 0.001). AntagomiR-1297 increased SOD (p = 0.001) and GSH-Px (p = 0.002) levels. Additionally, it reduced levels of cell inflammatory factors, including TNF-α, IL-6, and IL-1ß (all p < 0.001), and alleviated apoptosis (p < 0.001) in rat kidney tissue with DN. miR-1297 was pinpointed as a target for PTEN. AntagomiR-1297 increased PTEN expression and suppressed PI3K and AKT phosphorylation (all p < 0.001). CONCLUSIONS: AntagomiR-1297 can mitigate renal fibrosis, renal inflammation, apoptosis, and oxidative stress levels through the PTEN/PI3K/AKT pathway.


Sujet(s)
Diabète , Néphropathies diabétiques , microARN , Rats , Animaux , Protéines proto-oncogènes c-akt/génétique , Protéines proto-oncogènes c-akt/métabolisme , Protéines proto-oncogènes c-akt/pharmacologie , Phosphatidylinositol 3-kinase/génétique , Phosphatidylinositol 3-kinase/métabolisme , Phosphatidylinositol 3-kinase/pharmacologie , Néphropathies diabétiques/génétique , Néphropathies diabétiques/métabolisme , Phosphatidylinositol 3-kinases/métabolisme , Phosphatidylinositol 3-kinases/pharmacologie , Transduction du signal , Facteur de nécrose tumorale alpha/métabolisme , Facteur de nécrose tumorale alpha/pharmacologie , Antagomirs/métabolisme , Antagomirs/pharmacologie , Rein , microARN/génétique , Superoxide dismutase/métabolisme , Superoxide dismutase/pharmacologie , Diabète/métabolisme
13.
Cell Biochem Funct ; 42(3): e3998, 2024 Apr.
Article de Anglais | MEDLINE | ID: mdl-38561964

RÉSUMÉ

Breast cancer (BC) is the most commonly diagnosed cancer and the leading cause of cancer mortality in women. As the phosphatidylinositol 3-kinase (PI3K) signaling pathway is involved in a wide range of physiological functions of cells including growth, proliferation, motility, and angiogenesis, any alteration in this axis could induce oncogenic features; therefore, numerous preclinical and clinical studies assessed agents able to inhibit the components of this pathway in BC patients. To the best of our knowledge, this is the first study that analyzed all the registered clinical trials investigating safety and efficacy of the PI3K/AKT/mTOR axis inhibitors in BC. Of note, we found that the trends of PI3K inhibitors in recent years were superior as compared with the inhibitors of either AKT or mTOR. However, most of the trials entering phase III and IV used mTOR inhibitors (majorly Everolimus) followed by PI3K inhibitors (majorly Alpelisib) leading to the FDA approval of these drugs in the BC context. Despite favorable efficacies, our analysis shows that the majority of trials are utilizing PI3K pathway inhibitors in combination with hormone therapy and chemotherapy; implying monotherapy cannot yield huge clinical benefits, at least partly, due to the activation of compensatory mechanisms. To emphasize the beneficial effects of these inhibitors in combined-modal strategies, we also reviewed recent studies which investigated the conjugation of nanocarriers with PI3K inhibitors to reduce harmful toxicities, increase the local concentration, and improve their efficacies in the context of BC therapy.


Sujet(s)
Tumeurs du sein , Phosphatidylinositol 3-kinase , Humains , Femelle , Phosphatidylinositol 3-kinase/métabolisme , Phosphatidylinositol 3-kinase/pharmacologie , Phosphatidylinositol 3-kinase/usage thérapeutique , Tumeurs du sein/traitement médicamenteux , Tumeurs du sein/métabolisme , Protéines proto-oncogènes c-akt/métabolisme , Phosphatidylinositol 3-kinases/métabolisme , Transduction du signal , Sérine-thréonine kinases TOR/métabolisme , Inhibiteurs des phosphoinositide-3 kinases/pharmacologie , Inhibiteurs des phosphoinositide-3 kinases/usage thérapeutique
14.
J Ethnopharmacol ; 328: 118117, 2024 Jun 28.
Article de Anglais | MEDLINE | ID: mdl-38548120

RÉSUMÉ

ETHNOPHARMACOLOGICAL RELEVANCE: Chuanxiong, a plant of the Umbelliferae family, is a genuine medicinal herb from Sichuan Province. Phthalides are one of its main active components and exhibit good protective effect against cerebrovascular diseases. However, the mechanism by which phthalides exert neuroprotective effects is still largely unclear. AIM OF THE STUDY: In this study, we extracted a phthalein component (named as QBT) from Ligusticum Chuanxiong, and investigated its neuroprotective effects against vascular dementia (VaD) rats and the underlying mechanism, focusing on the chemokine 12 (CXCL12)/chemokine (C-X-C motif) receptor 4 (CXCR4) axis. METHODS: A rat model of VaD was established, and treated with QBT. Cognitive dysfunction in VaD rats was assessed using the Y-maze, new object recognition, and Morris water maze tests. Neuronal damage and inflammatory response in VaD rats were examined through Nissl staining, immunofluorescence, enzyme-linked immunospecific assay, and western blotting analysis. Furthermore, the effects of QBT on CXCL12/CXCR4 axis and its downstream signaling pathways, Janus kinase 2 (JAK2)/signal transducers and activators of transcription 3 (STAT3) and phosphatidylinositol 3 kinase (PI3K)/protein kinase B (AKT)/nuclear factor-κB (NF-κB), were investigated in VaD rats and BV2 microglial cells exposed to oxygen glucose deprivation. RESULTS: QBT significantly alleviated cognitive dysfunction and neuronal damage in VaD rats, along with inhibition of VaD-induced over-activation of microglia and astrocytes and inflammatory response. Moreover, QBT exhibited anti-inflammatory effects by inhibiting the CXCL12/CXCR4 axis and its downstream JAK2/STAT3 and PI3K/AKT/NF-κB pathways, thereby attenuating the neuroinflammatory response both in vivo and in vitro. CONCLUSION: QBT effectively mitigated neuronal damage and cognitive dysfunction in VaD rats, exerting neuroprotective effects by suppressing neuroinflammatory response through inhibition of the CXCL12/CXCR4 axis.


Sujet(s)
Dysfonctionnement cognitif , Démence vasculaire , Neuroprotecteurs , Rats , Animaux , Protéines proto-oncogènes c-akt/métabolisme , Facteur de transcription NF-kappa B/métabolisme , Maladies neuro-inflammatoires , Phosphatidylinositol 3-kinases/métabolisme , Rat Sprague-Dawley , Démence vasculaire/traitement médicamenteux , Démence vasculaire/métabolisme , Neuroprotecteurs/pharmacologie , Neuroprotecteurs/usage thérapeutique , Neuroprotecteurs/métabolisme , Phosphatidylinositol 3-kinase/métabolisme , Microglie , Dysfonctionnement cognitif/traitement médicamenteux , Dysfonctionnement cognitif/métabolisme , Chimiokine CXCL12/métabolisme
15.
J Integr Neurosci ; 23(3): 61, 2024 Mar 19.
Article de Anglais | MEDLINE | ID: mdl-38538223

RÉSUMÉ

BACKGROUND: Tanshinone IIA (TSIIA) is an element of the effective ingredients of Salvia miltiorrhiza Bunge (Labiatae), exhibits a significant therapeutic effect in brain neuroprotection. The focus of this study was the examination of synaptic plasticity of in Mg2+-free-induced epileptic hippocampus neurons and how TSIIA protects against it. METHODS: The purity of the primary hippocampal neurons extracted from Sprague Dawley rats was assessed within 24 hours by microtubule-associated protein (MAP2) immunofluorescence staining. A hippocampal neuron model for Mg2+-free-induced spontaneous recurrent epileptiform discharge was developed, five experimental groups were then randomized: blank (Blank), model (Model), TSIIA (TSIIA, 20 µM), LY294002 (LY294002, 25 µM), and TSIIA+LY294002 (TSIIA+LY294002, 20 µM+25 µM). FIJI software was used to examine variations of neurite complexity, total length of hippocampal neurons, number of primary dendrites and density of dendritic spines. Developmental regulation brain protein (Drebrin) and brain-derived neurotrophic factor (BDNF) expression was evaluated using immunofluorescence staining and the relative expression of phospho-protein kinase B (p-Akt)/Akt, BDNF, synaptophysin (SYN) and postsynaptic density 95 (PSD-95) determined by Western blot. RESULTS: In contrast to the model group, TSIIA drastically reduced damage to synaptic plasticity of hippocampal neurons caused by epilepsy (p < 0.05). The TSIIA group showed a significant increase in the relative expression of PSD-95, SYN, BDNF, and p-Akt/Akt (p < 0.01). CONCLUSIONS: TSIIA was effective in reducing harm to the synaptic plasticity of hippocampal neurons induced by persistent status epilepticus, with the possible mechanism being regulation of the phosphatidylinositol 3-kinase 56 (PI3K)/Akt signaling pathway.


Sujet(s)
Abiétanes , Épilepsie , Protéines proto-oncogènes c-akt , Animaux , Rats , Abiétanes/pharmacologie , Facteur neurotrophique dérivé du cerveau/métabolisme , Homologue-4 de la protéine Disks Large/métabolisme , Épilepsie/induit chimiquement , Épilepsie/traitement médicamenteux , Épilepsie/métabolisme , Hippocampe/métabolisme , Plasticité neuronale/physiologie , Neurones/métabolisme , Phosphatidylinositol 3-kinase/métabolisme , Phosphatidylinositol 3-kinases/métabolisme , Protéines proto-oncogènes c-akt/métabolisme , Rat Sprague-Dawley , Transduction du signal
16.
J Ethnopharmacol ; 327: 117946, 2024 Jun 12.
Article de Anglais | MEDLINE | ID: mdl-38447615

RÉSUMÉ

ETHNOPHARMACOLOGICAL RELEVANCE: In China, Xanthoceras sorbifolium Bunge was first documented as "Wen Guan Hua" in the "Jiu Huang Ben Cao" in 1406 A.D. According to the "National Compilation of Chinese Herbal Medicine," X. sorbifolium leaves are sweet and flat in nature and can dispel wind and dampness, suggesting that their extract can be used to treat rheumatoid arthritis. X. sorbifolium Bunge has also been used to treat arteriosclerosis, hyperlipidemia, hypertension, chronic hepatitis, and rheumatism, complications associated with hyperuricemic nephropathy (HN), a condition characterized by kidney damage resulting from high levels of uric acid (UA) in the blood. AIM OF THE STUDY: The purpose of this study was to investigate the effects and underlying mechanisms of a 70% ethanol extract from X. sorbifolium leaves (EX) in alleviating HN. MATERIALS AND METHODS: A mouse model of hyperuricemia was established to initially evaluate the hypouricemic effects and determine the effective dose of EX. Phytochemical analyses were conducted using ultra high-performance liquid chromatography and liquid chromatography-mass spectroscopy. The potential key pathways of EX in the alleviation of HN were inferred using network pharmacology and bioinformatics. An HN rat model was then established, and experiments including biomarker detection, western blotting, reverse transcription quantitative polymerase chain reaction, immunohistochemical and Masson's trichrome staining, and transmission electron microscopy were conducted to evaluate the effect of EX on UA transporter expression in vitro. RESULTS: Network pharmacology and bioinformatics analyses revealed that the phosphoinositide 3-kinase (PI3K)/AKT signaling pathway was the key pathway for the alleviation of HN progression by EX. EX treatment reduced serum biomarkers in HN rats, downregulated the expression of p-PI3K, p-AKT, glucose transporter 9 (GLUT9), urate transporter 1 (URAT1), Collagen I, matrix metalloproteinase (MMP)-2, and MMP-9, and upregulated the expression of ATP binding cassette subfamily G member 2 (ABCG2) to improve renal interstitial fibrosis in HN rats. A high content of both quercitrin and cynaroside were identified in EX; their administration inhibited the increased expression of GLUT9 and URAT1 in damaged HK-2 cells. CONCLUSION: Our study provides evidence that EX alleviates HN. The potential mechanism underlying this effect may be the regulation of UA transporters, such as GLUT9 and URAT1, by limiting the activation of the PI3K/AKT signaling pathway to improve renal injury.


Sujet(s)
Hyperuricémie , Maladies du rein , Souris , Rats , Animaux , Acide urique , Phosphatidylinositol 3-kinases/métabolisme , Protéines proto-oncogènes c-akt/métabolisme , Phosphatidylinositol 3-kinase/métabolisme , Rein , Maladies du rein/métabolisme , Transduction du signal , Marqueurs biologiques/métabolisme
17.
J Virol ; 98(4): e0170123, 2024 Apr 16.
Article de Anglais | MEDLINE | ID: mdl-38451084

RÉSUMÉ

Human adenoviruses (HAdV) are classified as DNA tumor viruses due to their potential to mediate oncogenic transformation in non-permissive mammalian cells and certain human stem cells. To achieve transformation, the viral early proteins of the E1 and E4 regions must block apoptosis and activate proliferation: the former predominantly through modulating the cellular tumor suppressor p53 and the latter by activating cellular pro-survival and pro-metabolism protein cascades, such as the phosphoinositide 3-kinase (PI3K-Akt) pathway, which is activated by HAdV E4orf1. Focusing on HAdV-C5, we show that E4orf1 is necessary and sufficient to stimulate Akt activation through phosphorylation in H1299 cells, which is not only hindered but repressed during HAdV-C5 infection with a loss of E4orf1 function in p53-positive A549 cells. Contrary to other research, E4orf1 localized not only in the common, cytoplasmic PI3K-Akt-containing compartment, but also in distinct nuclear aggregates. We identified a novel inhibitory mechanism, where p53 selectively targeted E4orf1 to destabilize it, also stalling E4orf1-dependent Akt phosphorylation. Co-IP and immunofluorescence studies showed that p53 and E4orf1 interact, and since p53 is bound by the HAdV-C5 E3 ubiquitin ligase complex, we also identified E4orf1 as a novel factor interacting with E1B-55K and E4orf6 during infection; overexpression of E4orf1 led to less-efficient E3 ubiquitin ligase-mediated proteasomal degradation of p53. We hypothesize that p53 specifically subverts the pro-survival function of E4orf1-mediated PI3K-Akt activation to protect the cell from metabolic hyper-activation or even transformation.IMPORTANCEHuman adenoviruses (HAdV) are nearly ubiquitous pathogens comprising numerous subtypes that infect various tissues and organs. Among many encoded proteins that facilitate viral replication and subversion of host cellular processes, the viral E4orf1 protein has emerged as an intriguing yet under-investigated player in the complex interplay between the virus and its host. Nonetheless, E4orf1 has gained attention as a metabolism activator and oncogenic agent, while recent research is showing that E4orf1 may play a more important role in modulating the cellular pathways such as phosphoinositide 3-kinase-Akt-mTOR. Our study reveals a novel and general impact of E4orf1 on host mechanisms, providing a novel basis for innovative antiviral strategies in future therapeutic settings. Ongoing investigations of the cellular pathways modulated by HAdV are of great interest, particularly since adenovirus-based vectors actually serve as vaccine or gene vectors. HAdV constitute an ideal model system to analyze the underlying molecular principles of virus-induced tumorigenesis.


Sujet(s)
Protéines E4 d'adénovirus , Adénovirus humains , Phosphatidylinositol 3-kinase , Protéines proto-oncogènes c-akt , Protéine p53 suppresseur de tumeur , Humains , Protéines E4 d'adénovirus/génétique , Protéines E4 d'adénovirus/métabolisme , Infections humaines à adénovirus/virologie , Adénovirus humains/croissance et développement , Adénovirus humains/métabolisme , Lignée cellulaire tumorale , Cellules HEK293 , Cadres ouverts de lecture/génétique , Phosphatidylinositol 3-kinase/métabolisme , Phosphorylation , Protéines proto-oncogènes c-akt/agonistes , Protéines proto-oncogènes c-akt/métabolisme , Protéine p53 suppresseur de tumeur/métabolisme , Ubiquitin-protein ligases/métabolisme , Réplication virale
18.
Chem Biol Interact ; 394: 110968, 2024 May 01.
Article de Anglais | MEDLINE | ID: mdl-38522564

RÉSUMÉ

Bone metastases caused by breast cancer pose a major challenge to the successful treatment of breast cancer patients. Many researchers have suggested that herbal medicines are extremely effective at preventing and treating cancer-associated osteolysis. Previous studies have revealed that Morusin (MOR) is cytotoxic to many cancer cells ex vivo. Nevertheless, how MOR contributes to osteolysis induced by breast cancer is still unknown, and the potential mechanism of action against osteolysis is worthy of further study. The protective effect and molecular mechanism of MOR in inhibiting breast cancer cell-induced osteolysis were verified by experiments and network pharmacology. Cell function was assessed by cell proliferation, osteoclast (OC) formation, bone resorption, and phalloidin staining. Tumour growth was examined by micro-CT scanning in vivo. To identify potential MOR treatments, the active ingredient-target pathway of breast cancer was screened using network pharmacology and molecular docking approaches. This study is the first to report that MOR can prevent osteolysis induced by breast cancer cells. Specifically, our results revealed that MOR inhibits RANKL-induced osteoclastogenesis and restrains the proliferation, invasion and migration of MDA-MB-231 breast cells through restraining the PI3K/AKT/MTOR signalling pathway. Notably, MOR prevented bone loss caused by breast cancer cell-induced osteolysis in vivo, indicating that MOR inhibited the development of OCs and the resorption of bone, which are essential for cancer cell-associated bone distraction. This study showed that MOR treatment inhibited osteolysis induced by breast cancer in vivo. MOR inhibited OC differentiation and bone resorption ex vivo and in vivo and might be a potential drug candidate for treating breast cancer-induced osteolysis.


Sujet(s)
Tumeurs du sein , Ostéolyse , Phosphatidylinositol 3-kinase , Transduction du signal , Sérine-thréonine kinases TOR , Animaux , Femelle , Humains , Souris , Tumeurs du sein/traitement médicamenteux , Lignée cellulaire tumorale , Mouvement cellulaire/effets des médicaments et des substances chimiques , Prolifération cellulaire/effets des médicaments et des substances chimiques , Souris de lignée BALB C , Souris nude , Simulation de docking moléculaire , Ostéoclastes/effets des médicaments et des substances chimiques , Ostéoclastes/métabolisme , Ostéolyse/métabolisme , Ostéolyse/traitement médicamenteux , Ostéolyse/anatomopathologie , Phosphatidylinositol 3-kinase/métabolisme , Protéines proto-oncogènes c-akt/métabolisme , Ligand de RANK/métabolisme , Transduction du signal/effets des médicaments et des substances chimiques , Sérine-thréonine kinases TOR/métabolisme
19.
PLoS One ; 19(3): e0300051, 2024.
Article de Anglais | MEDLINE | ID: mdl-38527038

RÉSUMÉ

The cytotoxicity of the ethyl acetate fraction of the Calotropis gigantea (L.) Dryand. (C. gigantea) stem bark extract (CGEtOAc) has been demonstrated in many types of cancers. This study examined the improved cancer therapeutic activity of sorafenib when combined with CGEtOAc in HepG2 cells. The cell viability and cell migration assays were applied in HepG2 cells treated with varying concentrations of CGEtOAc, sorafenib, and their combination. Flow cytometry was used to determine apoptosis, which corresponded with a decline in mitochondrial membrane potential and activation of DNA fragmentation. Reactive oxygen species (ROS) levels were assessed in combination with the expression of the phosphatidylinositol-3-kinase (PI3K)/ protein kinase B (Akt)/ mammalian target of rapamycin (mTOR) pathway, which was suggested for association with ROS-induced apoptosis. Combining CGEtOAc at 400 µg/mL with sorafenib at 4 µM, which were their respective half-IC50 concentrations, significantly inhibited HepG2 viability upon 24 h of exposure in comparison with the vehicle and each single treatment. Consequently, CGEtOAc when combined with sorafenib significantly diminished HepG2 migration and induced apoptosis through a mitochondrial-correlation mechanism. ROS production was speculated to be the primary mechanism of stimulating apoptosis in HepG2 cells after exposure to a combination of CGEtOAc and sorafenib, in association with PI3K/Akt/mTOR pathway suppression. Our results present valuable knowledge to support the development of anticancer regimens derived from the CGEtOAc with the chemotherapeutic agent sorafenib, both of which were administered at half-IC50, which may minimize the toxic implications of cancer treatments while improving the therapeutic effectiveness toward future medical applications.


Sujet(s)
Acétates , Calotropis , Tumeurs du foie , Humains , Sorafénib/pharmacologie , Sorafénib/usage thérapeutique , Protéines proto-oncogènes c-akt/métabolisme , Cellules HepG2 , Calotropis/métabolisme , Phosphatidylinositol 3-kinases/métabolisme , Espèces réactives de l'oxygène/métabolisme , Écorce/métabolisme , Sérine-thréonine kinases TOR/métabolisme , Phosphatidylinositol 3-kinase/métabolisme , Apoptose , Lignée cellulaire tumorale , Tumeurs du foie/traitement médicamenteux , Tumeurs du foie/métabolisme
20.
Zhen Ci Yan Jiu ; 49(3): 265-273, 2024 Mar 25.
Article de Anglais, Chinois | MEDLINE | ID: mdl-38500323

RÉSUMÉ

OBJECTIVES: To observe the effects of electroacupuncture (EA) on the phosphatidylinositol-3-kinase (PI3K)/protein kinase B (Akt)/cAMP response element binding protein (CREB) signaling pathway-related proteins and hippocampal neuron apoptosis in diabetic cognitive impairment (DCI) rats, and to explore the mechanisms of EA in treating DCI. METHODS: Adult male SD rats were randomly divided into normal, model, and EA groups, with 12 rats in each group. The animal model of DCI was replicated using a high-fat, high-sugar diet combined with low-dose streptozotocin. The EA group received EA stimulation at "Yishu" (EX-B6), "Zusanli" (ST36), "Baihui" (GV20), and "Dazhui" (GV14). Blood glucose contents of the rats in each group were measured. The Morris water maze test was used to assess the learning and memory abilities of rats. Transmission electron microscopy was used to observe the ultrastructure of hippocampal CA1 neurons. Nissl staining was used to observe the pathological changes in hippocampal CA1 neurons. TUNEL staining was used to detect the apoptosis in hippocampal CA1 neurons. Western blot was used to detect the protein expression levels of p-PI3K/PI3K and p-Akt/Akt, as well as CREB, p-CREB, cysteine aspartate pro-tease (Caspase)-3, B-cell lymphoma-2 (Bcl-2), and Bcl-2 related X protein (Bax) in the hippocampal tissue of rats. RESULTS: Compared with the normal group, the rats' random blood glucose contents were significantly increased (P<0.01), the escape latency prolonged (P<0.01), and the original platform crossing counts reduced (P<0.01) in the model group. Significant damage to hippocampal CA1 neurons, a significantly increased neuronal apoptosis index (P<0.01), decreased ratio of p-PI3K/PI3K and p-Akt/Akt and expression of CREB, p-CREB and Bcl-2 proteins, increased expression of Caspase-3 and Bax proteins (P<0.01) were observed in the hippocampal tissue of rats in the model group. Compared with the model group, the rats in the EA group showed decreased random blood glucose content (P<0.01), shortened escape latency (P<0.01), increased original platform crossing counts (P<0.01), improved quantity and pathological morphology and ultrastructure of hippocampal CA1 neurons, reduced neuronal apoptosis index (P<0.01), increased ratio of p-PI3K/PI3K and p-Akt/Akt, and expression of CREB, p-CREB and Bcl-2 proteins (P<0.05, P<0.01) in the hippocampal tissue, and decreased expression of Caspase-3 and Bax proteins (P<0.01). CONCLUSIONS: EA can improve the learning and memory abilities of rats with DCI, and the mechanism may be related to the regulation of the expression of PI3K/Akt/CREB signaling pathway-related proteins, which attenuates the neuronal apoptosis in the hippocampus of rats, and improves the neural function.


Sujet(s)
Dysfonctionnement cognitif , Diabète , Électroacupuncture , Rats , Mâle , Animaux , Protéines proto-oncogènes c-akt/génétique , Protéines proto-oncogènes c-akt/métabolisme , Rat Sprague-Dawley , Phosphatidylinositol 3-kinases/génétique , Protéine Bax/génétique , Protéine Bax/métabolisme , Caspase-3/métabolisme , Protéine de liaison à l'élément de réponse à l'AMP cyclique/génétique , Protéine de liaison à l'élément de réponse à l'AMP cyclique/métabolisme , Phosphatidylinositol 3-kinase/métabolisme , Glycémie , Transduction du signal , Hippocampe/métabolisme , Apoptose , Dysfonctionnement cognitif/génétique , Dysfonctionnement cognitif/thérapie
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE
...