Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 20 de 2.972
Filtrer
1.
Chem Pharm Bull (Tokyo) ; 72(8): 751-761, 2024.
Article de Anglais | MEDLINE | ID: mdl-39143008

RÉSUMÉ

Gout is the second largest metabolic disease worldwide after diabetes, with acute gouty arthritis as most common symptom. Xanthine oxidase (XOD) and the NOD like receptor-3 (NLRP3) inflammasome are the key targets for acute gout treatment. Chlorogenic acid has been reported with a good anti-inflammatory activity, and Apigenin showed an excellent potential in XOD inhibition. Therefore, a series of chlorogenic acid-apigenin (CA) conjugates with varying linkers were designed and synthesized as dual XOD/NLRP3 inhibitors, and their activities both in XOD and NLRP3 inhibition were evaluated. An in vitro study of XOD inhibitory activity revealed that the majority of CA conjugates exhibited favorable XOD inhibitory activity. Particularly, the effects of compounds 10c and 10d, with an alkyl linker on the apigenin moiety, were stronger than that of allopurinol. The selected CA conjugates also demonstrated a favorable anti-inflammatory activity in RAW264.7 cells. Furthermore, compound 10d, which showed the optimal activity both in XOD inhibition and anti-inflammatory, was chosen and its inhibitory ability on NLRP3 and related proinflammatory cytokines was further tested. Compound 10d effectively reduced NLRP3 expression and the secretion of interluekin-1ß (IL-1ß) and tumor necrosis factor-α (TNF-α) with an activity stronger than the positive control isoliquiritigenin (ISL). Based on these findings, compound 10d exhibits dual XOD/NLRP3 inhibitory activity and, therefore, the therapeutic effects on acute gout is worthy of further study.


Sujet(s)
Apigénine , Acide chlorogénique , Antigoutteux , Protéine-3 de la famille des NLR contenant un domaine pyrine , Animaux , Souris , Apigénine/pharmacologie , Apigénine/composition chimique , Apigénine/synthèse chimique , Protéine-3 de la famille des NLR contenant un domaine pyrine/antagonistes et inhibiteurs , Protéine-3 de la famille des NLR contenant un domaine pyrine/métabolisme , Cellules RAW 264.7 , Acide chlorogénique/pharmacologie , Acide chlorogénique/composition chimique , Acide chlorogénique/synthèse chimique , Antigoutteux/pharmacologie , Antigoutteux/synthèse chimique , Antigoutteux/composition chimique , Antigoutteux/usage thérapeutique , Relation structure-activité , Xanthine oxidase/antagonistes et inhibiteurs , Xanthine oxidase/métabolisme , Structure moléculaire , Goutte/traitement médicamenteux , Relation dose-effet des médicaments , Antienzymes/synthèse chimique , Antienzymes/pharmacologie , Antienzymes/composition chimique , Anti-inflammatoires/synthèse chimique , Anti-inflammatoires/pharmacologie , Anti-inflammatoires/composition chimique
2.
BMC Infect Dis ; 24(1): 695, 2024 Jul 12.
Article de Anglais | MEDLINE | ID: mdl-38997656

RÉSUMÉ

BACKGROUND: Sepsis is a life-threatening organ dysfunction, which seriously threatens human health. The clinical and experimental results have confirmed that Traditional Chinese medicine (TCM), such as Scutellariae Radix, has anti-inflammatory effects. This provides a new idea for the treatment of sepsis. This study systematically analyzed the mechanism of Scutellariae Radix treatment in sepsis based on network pharmacology, RNA sequencing and molecular docking. METHODS: Gene expression analysis was performed using Bulk RNA sequencing on sepsis patients and healthy volunteers. After quality control of the results, the differentially expressed genes (DEGs) were analyzed. The active ingredients and targets of Scutellariae Radix were identified using The Traditional Chinese Medicine Systems Pharmacology Database and Analysis Platform (TCMSP). Gene Ontology (GO) and Protein-Protein Interaction (PPI) analysis were performed for disease-drug intersection targets. With the help of GEO database, Survival analysis and Meta-analysis was performed on the cross-targets to evaluate the prognostic value and screen the core targets. Subsequently, single-cell RNA sequencing was used to determine where the core targets are located within the cell. Finally, in this study, molecular docking experiments were performed to further clarify the interrelationship between the active components of Scutellariae Radix and the corresponding targets. RESULTS: There were 72 active ingredients of Scutellariae Radix, and 50 common targets of drug and disease. GO and PPI analysis showed that the intersection targets were mainly involved in response to chemical stress, response to oxygen levels, response to drug, regulation of immune system process. Survival analysis showed that PRKCD, EGLN1 and CFLAR were positively correlated with sepsis prognosis. Meta-analysis found that the three genes were highly expressed in sepsis survivor, while lowly in non-survivor. PRKCD was mostly found in Macrophages, while EGLN1 and CFLAR were widely expressed in immune cells. The active ingredient Apigenin regulates CFLAR expression, Baicalein regulates EGLN1 expression, and Wogonin regulates PRKCD expression. Molecular docking studies confrmed that the three active components of astragalus have good binding activities with their corresponding targets. CONCLUSIONS: Apigenin, Baicalein and Wogonin, important active components of Scutellaria Radix, produce anti-sepsis effects by regulating the expression of their targets CFLAR, EGLN1 and PRKCD.


Sujet(s)
Médicaments issus de plantes chinoises , Simulation de docking moléculaire , Scutellaria baicalensis , Sepsie , Analyse de séquence d'ARN , Humains , Sepsie/traitement médicamenteux , Scutellaria baicalensis/composition chimique , Médicaments issus de plantes chinoises/usage thérapeutique , Médicaments issus de plantes chinoises/pharmacologie , Médicaments issus de plantes chinoises/composition chimique , Médecine traditionnelle chinoise , Flavanones/usage thérapeutique , Flavanones/pharmacologie , Cartes d'interactions protéiques , Apigénine/usage thérapeutique , Apigénine/pharmacologie , Analyse de profil d'expression de gènes , Gene Ontology , Pharmacologie des réseaux
3.
Int J Mol Sci ; 25(14)2024 Jul 13.
Article de Anglais | MEDLINE | ID: mdl-39062932

RÉSUMÉ

Flavonoids, a class of natural compounds with anticancer activity, exhibit varying biological activities and potencies based on their structural differences. Acylation, including acetylation of flavonoids, generally increases their structural diversity, which is closely related to the diversity of bioactivity within this group of compounds. However, it remains largely unknown how acetylation affects the bioactivity of many flavonoids. Based on our previous findings that O-acetylation enhances quercetin's bioactivity against various cancer cells, we synthesized 12 acetylated flavonoids, including seven novel compounds, to investigate their anticancer activities in the MDA-MB-231, HCT-116, and HepG2 cell lines. Our results showed that acetylation notably enhanced the cell proliferation inhibitory effect of quercetin and kaempferol across all cancer cell lines tested. Interestingly, while the 5,7,4'-O-triacetate apigenin (3Ac-A) did not show an enhanced the effect of inhibition of cell proliferation through acetylation, it exhibited significantly strong anti-migration activity in MDA-MB-231 cells. In contrast, the 7,4'-O-diacetate apigenin (2Ac-Q), which lacks acetylation at the 5-position hydroxy group, showed enhanced cell proliferation inhibitory effect but had weaker anti-migration effects compared to 3Ac-A. These results indicated that acetylated flavonoids, especially quercetin, kaempferol, and apigenin derivatives, are promising for anticancer applications, with 3Ac-A potentially having unique anti-migration pathways independent of apoptosis induction. This study highlights the potential application of flavonoids in novel chemopreventive strategies for their anti-cancer activity.


Sujet(s)
Prolifération cellulaire , Flavonoïdes , Humains , Acétylation/effets des médicaments et des substances chimiques , Flavonoïdes/pharmacologie , Flavonoïdes/composition chimique , Prolifération cellulaire/effets des médicaments et des substances chimiques , Lignée cellulaire tumorale , Mouvement cellulaire/effets des médicaments et des substances chimiques , Quercétine/pharmacologie , Quercétine/composition chimique , Kaempférols/pharmacologie , Kaempférols/composition chimique , Tumeurs/traitement médicamenteux , Tumeurs/métabolisme , Tumeurs/anatomopathologie , Tumeurs/prévention et contrôle , Antinéoplasiques/pharmacologie , Antinéoplasiques/composition chimique , Apoptose/effets des médicaments et des substances chimiques , Cellules HepG2 , Apigénine/pharmacologie , Apigénine/composition chimique
4.
Biomed Pharmacother ; 177: 117075, 2024 Aug.
Article de Anglais | MEDLINE | ID: mdl-38964181

RÉSUMÉ

Obesity is a growing epidemic among reproductive-age men, which can cause and exacerbate male infertility by means of associated comorbidities, endocrine abnormalities, and direct effects on the fidelity and throughput of spermatogenesis. A prominent consequence of male obesity is a reduction in testosterone levels. Natural products have shown tremendous potential anti-obesity effects in metabolic diseases. This study aimed to investigate the potential of apigenin (AP) to alleviate testicular dysfunction induced by a high-fat diet (HFD) and to investigate the underlying mechanisms, focusing on endoplasmic reticulum stress (ERS) and testosterone synthesis. A murine model of obesity was established using HFD-fed mice. The effects of AP on obesity, lipid metabolism, testicular dysfunction, and ERS were assessed through various physiological, histological, and molecular techniques. Administration of AP (10 mg/kg) ameliorated HFD-induced obesity and testicular dysfunction in a mouse model, as evidenced by decreased body weight, improved lipid profiles and testicular pathology, and restored protein levels related to testosterone. Furthermore, in vitro studies demonstrated that AP relieved ERS and recovered testosterone synthesis in murine Leydig cells (TM3) treated with free fatty acids (FFAs). It was also observed that AP rescued testosterone synthesis enzymes in TM3 cells, similar to that observed with the inhibitor of the PERK pathway (GSK2606414). In addition, ChIP, qPCR, and gene silencing showed that the C/EBP homologous protein (CHOP) bound directly to the promoter region of steroidogenic STAR and negatively modulated its expression. Collectively, AP has remarkable potential to alleviate HFD-induced obesity and testicular dysfunction. Its protective effects are attributable partly to mitigating ERS and restoring testosterone synthesis in Leydig cells.


Sujet(s)
Apigénine , Alimentation riche en graisse , Stress du réticulum endoplasmique , Cellules de Leydig , Souris de lignée C57BL , Obésité , Testicule , Testostérone , Animaux , Mâle , Stress du réticulum endoplasmique/effets des médicaments et des substances chimiques , Apigénine/pharmacologie , Souris , Alimentation riche en graisse/effets indésirables , Obésité/traitement médicamenteux , Obésité/métabolisme , Cellules de Leydig/effets des médicaments et des substances chimiques , Cellules de Leydig/métabolisme , Testicule/effets des médicaments et des substances chimiques , Testicule/métabolisme , Testicule/anatomopathologie , Lignée cellulaire , Métabolisme lipidique/effets des médicaments et des substances chimiques
5.
Exp Cell Res ; 441(1): 114150, 2024 Aug 01.
Article de Anglais | MEDLINE | ID: mdl-38971519

RÉSUMÉ

Despite significant advances in the treatment of colorectal cancer (CRC), identification of novel targets and treatment options are imperative for improving its prognosis and survival rates. The mitochondrial SIRT3 and SHMT2 have key roles in metabolic reprogramming and cell proliferation. This study investigated the potential use of the natural product apigenin in CRC treatment employing both in vivo and in vitro models and explored the role of SIRT3 and SHMT2 in apigenin-induced CRC apoptosis. The role of SHMT2 in CRC patients' survival was verified using TCGA database. In vivo, apigenin treatment restored the normal colon appearance. On the molecular level, apigenin augmented the immunohistochemical expression of cleaved caspase-3 and attenuated SIRT3 and SHMT2 mRNA expression CRC patients with decreased SHMT2 expression had improved overall and disease-free survival rates. In vitro, apigenin reduced the cell viability in a time-dependent manner, induced G0/G1 cell cycle arrest, and increased the apoptotic cell population compared to the untreated control. Mechanistically, apigenin treatment mitigated the expression of SHMT2, SIRT3, and its upstream long intergenic noncoding RNA LINC01234 in CRC cells. Conclusively, apigenin induces caspase-3-dependent apoptosis in CRC through modulation of SIRT3-triggered mitochondrial pathway suggesting it as a promising therapeutic agent to improve patient outcomes.


Sujet(s)
Apigénine , Apoptose , Prolifération cellulaire , Tumeurs colorectales , Sirtuine-3 , Apigénine/pharmacologie , Humains , Tumeurs colorectales/traitement médicamenteux , Tumeurs colorectales/anatomopathologie , Tumeurs colorectales/métabolisme , Tumeurs colorectales/génétique , Sirtuine-3/métabolisme , Sirtuine-3/génétique , Apoptose/effets des médicaments et des substances chimiques , Prolifération cellulaire/effets des médicaments et des substances chimiques , Animaux , Souris , Régulation de l'expression des gènes tumoraux/effets des médicaments et des substances chimiques , Souris nude , Lignée cellulaire tumorale , Transduction du signal/effets des médicaments et des substances chimiques , Survie cellulaire/effets des médicaments et des substances chimiques , Tests d'activité antitumorale sur modèle de xénogreffe , ARN long non codant/génétique , ARN long non codant/métabolisme , Glycine hydroxymethyltransferase
6.
Eur J Pharmacol ; 978: 176800, 2024 Sep 05.
Article de Anglais | MEDLINE | ID: mdl-38950835

RÉSUMÉ

Adiponectin plays key roles in energy metabolism and ameliorates inflammation, oxidative stress, and mitochondrial dysfunction via its primary receptors, adiponectin receptors -1 and 2 (AdipoR1 and AdipoR2). Systemic depletion of adiponectin causes various metabolic disorders, including MASLD; however adiponectin supplementation is not yet achievable owing to its large size and oligomerization-associated complexities. Small-molecule AdipoR agonists, thus, may provide viable therapeutic options against metabolic disorders. Using a novel luciferase reporter-based assay here, we have identified Apigenin-6-C-glucoside (ACG), but not apigenin, as a specific agonist for the liver-rich AdipoR isoform, AdipoR2 (EC50: 384 pM) with >10000X preference over AdipoR1. Immunoblot analysis in HEK-293 overexpressing AdipoR2 or HepG2 and PLC/PRF/5 liver cell lines revealed rapid AMPK, p38 activation and induction of typical AdipoR targets PGC-1α and PPARα by ACG at a pharmacologically relevant concentration of 100 nM (reported cMax in mouse; 297 nM). ACG-mediated AdipoR2 activation culminated in a favorable modulation of key metabolic events, including decreased inflammation, oxidative stress, mitochondrial dysfunction, de novo lipogenesis, and increased fatty acid ß-oxidation as determined by immunoblotting, QRT-PCR and extracellular flux analysis. AdipoR2 depletion or AMPK/p38 inhibition dampened these effects. The in vitro results were recapitulated in two different murine models of MASLD, where ACG at 10 mg/kg body weight robustly reduced hepatic steatosis, fibrosis, proinflammatory macrophage numbers, and increased hepatic glycogen content. Together, using in vitro experiments and rodent models, we demonstrate a proof-of-concept for AdipoR2 as a therapeutic target for MASLD and provide novel chemicobiological insights for the generation of translation-worthy pharmacological agents.


Sujet(s)
Apigénine , Glucosides , Récepteurs à l'adiponectine , Récepteurs à l'adiponectine/agonistes , Récepteurs à l'adiponectine/métabolisme , Animaux , Humains , Souris , Apigénine/pharmacologie , Apigénine/usage thérapeutique , Glucosides/pharmacologie , Glucosides/usage thérapeutique , Mâle , Cellules HepG2 , Cellules HEK293 , Modèles animaux de maladie humaine , Souris de lignée C57BL , Stress oxydatif/effets des médicaments et des substances chimiques , Foie/effets des médicaments et des substances chimiques , Foie/métabolisme , Foie/anatomopathologie , AMP-Activated Protein Kinases/métabolisme
7.
J Pharm Biomed Anal ; 248: 116325, 2024 Sep 15.
Article de Anglais | MEDLINE | ID: mdl-38959755

RÉSUMÉ

The high prevalence of cancer and detrimental side effects associated with many cancer treatments necessitate the search for effective alternative therapies. Natural products are increasingly being recognized and investigated for their potential therapeutic benefits. Scutellaria barbata D. Don (SBD), a plant with potent antitumor properties, has attracted significant interest from oncology researchers. Its primary flavonoid components-scutellarin and luteolin-which have limited oral bioavailability due to poor absorption. This hinders its application for cancer treatment. The gut microbiota, which is considered a metabolic organ, can modulate the biotransformation of compounds, thereby altering their bioavailability and efficacy. In this study, we employed liquid chromatography tandem mass spectrometry (LC-MS/MS 8060) and ion trap-time of flight (LC-MSn-IT-TOF) analysis to investigate the ex vivo metabolism of scutellarin and luteolin by the gut microbiota. Five metabolites and one potential metabolite were identified. We summarized previous studies on their antitumor effects and performed in vitro tumor cell line studies to prove their antitumor activities. The possible key pathway of gut microbiota metabolism in vitro was validated using molecular docking and pure enzyme metabolic experiments. In addition, we explored the antitumor mechanisms of the two components of SBD through network pharmacology, providing a basis for subsequent target identification. These findings expand our understanding of the antitumor mechanisms of SBD. Notably, this study contributes to the existing body of knowledge regarding flavonoid biotransformation by the gut microbiota, highlighting the therapeutic potential of SBD in cancer treatment. Moreover, our results provide a theoretical basis for future in vivo pharmacokinetic studies, aiming to optimize the clinical efficacy of SBD in oncological applications.


Sujet(s)
Apigénine , Microbiome gastro-intestinal , Glucuronates , Lutéoline , Scutellaria , Spectrométrie de masse en tandem , Microbiome gastro-intestinal/effets des médicaments et des substances chimiques , Lutéoline/pharmacologie , Lutéoline/métabolisme , Lutéoline/pharmacocinétique , Scutellaria/composition chimique , Apigénine/pharmacologie , Glucuronates/métabolisme , Humains , Spectrométrie de masse en tandem/méthodes , Lignée cellulaire tumorale , Animaux , Simulation de docking moléculaire , Extraits de plantes/pharmacologie , Chromatographie en phase liquide/méthodes , Antinéoplasiques d'origine végétale/pharmacologie , Antinéoplasiques d'origine végétale/pharmacocinétique , Biodisponibilité , Mâle , Biotransformation , Antinéoplasiques/pharmacologie , Antinéoplasiques/pharmacocinétique
8.
J Ethnopharmacol ; 334: 118518, 2024 Nov 15.
Article de Anglais | MEDLINE | ID: mdl-38964628

RÉSUMÉ

ETHNOPHARMACOLOGICAL RELEVANCE: Salvia miltiorrhiza Bunge (S. miltiorrhiza) is an important Traditional Chinese herbal Medicine (TCM) used to treat cardio-cerebrovascular diseases. Based on the pharmacodynamic substance of S. miltiorrhiza, the aim of present study was to investigate the underlying mechanism of S. miltiorrhiza against cardiac fibrosis (CF) through a systematic network pharmacology approach, molecular docking and dynamics simulation as well as experimental investigation in vitro. MATERIALS AND METHODS: A systematic pharmacological analysis was conducted using the Traditional Chinese Medicine Pharmacology (TCMSP) database to screen the effective chemical components of S. miltiorrhiza, then the corresponding potential target genes of the compounds were obtained by the Swiss Target Prediction and TCMSP databases. Meanwhile, GeneCards, DisGeNET, OMIM, and TTD disease databases were used to screen CF targets, and a protein-protein interaction (PPI) network of drug-disease targets was constructed on S. miltiorrhiza/CF targets by Search Tool for the Retrieval of Interacting Genes/Proteins (STING) database. After that, the component-disease-target network was constructed by software Cytoscape 3.7. Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) enrichment analysis were performed for the intersection targets between drug and disease. The relationship between active ingredient of S. miltiorrhiza and disease targets of CF was assessed via molecular docking and molecular dynamics simulation. Subsequently, the underlying mechanism of the hub compound on CF was experimentally investigated in vitro. RESULTS: 206 corresponding targets to effective chemical components from S. miltiorrhiza were determined, and among them, there were 82 targets that overlapped with targets of CF. Further, through PPI analysis, AKT1 and GSK3ß were the hub targets, and which were both enriched in the PI3K/AKT signaling pathway, it was the sub-pathways of the lipid and atherosclerosis pathway. Subsequently, compound-disease-genes-pathways diagram is constructed, apigenin (APi) was a top ingredients and AKT1 (51) and GSK3ß (22) were the hub genes according to the degree value. The results of molecular docking and dynamics simulation showed that APi has strong affinities with AKT and GSK3ß. The results of cell experiments showed that APi inhibited cells viability, proliferation, proteins expression of α-SMA and collagen I/III, phosphorylation of AKT1 and GSK3ß in MCFs induced by TGFß1. CONCLUSION: Through a systematic network pharmacology approach, molecular docking and dynamics simulation, and confirmed by in vitro cell experiments, these results indicated that APi interacts with AKT and GSK3ß to disrupt the phosphorylation of AKT and GSK3ß, thereby inhibiting the proliferation and differentiation of MCFs induced by TGFß1, which providing new insights into the pharmacological mechanism of S. miltiorrhiza in the treatment of CF.


Sujet(s)
Apigénine , Différenciation cellulaire , Prolifération cellulaire , Glycogen synthase kinase 3 beta , Simulation de docking moléculaire , Protéines proto-oncogènes c-akt , Transduction du signal , Protéines proto-oncogènes c-akt/métabolisme , Transduction du signal/effets des médicaments et des substances chimiques , Prolifération cellulaire/effets des médicaments et des substances chimiques , Glycogen synthase kinase 3 beta/métabolisme , Animaux , Apigénine/pharmacologie , Apigénine/composition chimique , Différenciation cellulaire/effets des médicaments et des substances chimiques , Fibroblastes/effets des médicaments et des substances chimiques , Fibroblastes/métabolisme , Cartes d'interactions protéiques , Rats , Pharmacologie des réseaux , Simulation de dynamique moléculaire , Lignée cellulaire , Humains
9.
Int Immunopharmacol ; 139: 112710, 2024 Sep 30.
Article de Anglais | MEDLINE | ID: mdl-39029229

RÉSUMÉ

PANoptosis is manifested with simultaneous activation of biomarkers for both pyroptotic, apoptotic and necroptotic signaling via the molecular platform PANoptosome and it is involved in pathologies of various inflammatory diseases including hemophagocytic lymphohistiocytosis (HLH). Scutellarin is a flavonoid isolated from herbal Erigeron breviscapus (Vant.) Hand.-Mazz. and has been shown to possess multiple pharmacological effects, but it is unknown whether scutellarin has any effects on PANoptosis and related inflammatory diseases. In this study, we found that scutellarin inhibited cell death in bone marrow-derived macrophages (BMDMs) and J774A.1 cells treated with TGF-ß-activated kinase 1 (TAK1) inhibitor 5Z-7-oxozeaenol (OXO) plus lipopolysaccharide (LPS), which has been commonly used to induce PANoptosis. Western blotting showed that scutellarin dose-dependently inhibited the activation biomarkers for pyroptotic (Caspase-1p10 and GSDMD-NT), apoptotic (cleaved Casp3/8/9 and GSDME-NT), and necroptotic (phosphorylated MLKL) signaling. The inhibitory effect of scutellarin was unaffected by NLRP3 or Caspase-1 deletion. Interestingly, scutellarin blocked the assembly of PANoptosome that encompasses ASC, RIPK3, Caspase-8 and ZBP1, suggesting its action on upstream signaling. Consistent with this, scutellarin inhibited mitochondrial damage and mitochondrial reactive oxygen species (mtROS) generation in cells treated with OXO+LPS. Further, mito-TEMPO that can scavenge mtROS significantly inhibited OXO+LPS-induced PANoptotic cell death. In line with the in vitro results, scutellarin markedly alleviated systemic inflammation, multiple organ injury, and activation of PANoptotic biomarkers in mice with HLH. Collectively, our data suggest that scutellarin can inhibit PANoptosis by suppressing mitochondrial damage and mtROS generation and thereby mitigating multiple organ injury in mice with inflammatory disorders.


Sujet(s)
Apigénine , Glucuronates , Lipopolysaccharides , Souris de lignée C57BL , Mitochondries , Espèces réactives de l'oxygène , Apigénine/pharmacologie , Apigénine/usage thérapeutique , Glucuronates/pharmacologie , Glucuronates/usage thérapeutique , Animaux , Espèces réactives de l'oxygène/métabolisme , Mitochondries/effets des médicaments et des substances chimiques , Mitochondries/métabolisme , Souris , Lignée cellulaire , Macrophages/effets des médicaments et des substances chimiques , Macrophages/métabolisme , Macrophages/immunologie , Anti-inflammatoires/pharmacologie , Anti-inflammatoires/usage thérapeutique , Nécroptose/effets des médicaments et des substances chimiques , Mâle , MAP Kinase Kinase Kinases/métabolisme , Inflammation/traitement médicamenteux , Transduction du signal/effets des médicaments et des substances chimiques , Zéaralénone/administration et posologie , Lactones , Résorcinol
10.
FASEB J ; 38(13): e23769, 2024 Jul 15.
Article de Anglais | MEDLINE | ID: mdl-38958951

RÉSUMÉ

Renal ischemia-reperfusion injury (IRI) is an integral process in renal transplantation, which results in compromised graft survival. Macrophages play an important role in both the early inflammatory period and late fibrotic period in response to IRI. In this study, we investigated whether scutellarin (SCU) could protect against renal IRI by regulating macrophage polarization. Mice were given SCU (5-50 mg/kg) by gavage 1 h earlier, followed by a unilateral renal IRI. Renal function and pathological injury were assessed 24 h after reperfusion. The results showed that administration of 50 mg/kg SCU significantly improved renal function and renal pathology in IRI mice. In addition, SCU alleviated IRI-induced apoptosis. Meanwhile, it reduced macrophage infiltration and inhibited pro-inflammatory macrophage polarization. Moreover, in RAW 264.7 cells and primary bone marrow-derived macrophages (BMDMs) exposed to SCU, we found that 150 µM SCU inhibited these cells to polarize to an inflammatory phenotype induced by lipopolysaccharide (LPS) and interferon-γ (IFN-γ). However, SCU has no influence on anti-inflammatory macrophage polarization in vivo and in vitro induced by in interleukin-4 (IL-4). Finally, we explored the effect of SCU on the activation of the mitogen-activated protein kinase (MAPK) pathway both in vivo and in vitro. We found that SCU suppressed the activation of the MAPK pathway, including the extracellular signal-regulated kinase (ERK), Jun N-terminal kinase (JNK), and p38. Our results demonstrated that SCU protects the kidney against IRI by inhibiting macrophage infiltration and polarization toward pro-inflammatory phenotype via the MAPK pathway, suggesting that SCU may be therapeutically important in treatment of IRI.


Sujet(s)
Apigénine , Glucuronates , Système de signalisation des MAP kinases , Macrophages , Lésion d'ischémie-reperfusion , Animaux , Mâle , Souris , Apigénine/pharmacologie , Apoptose/effets des médicaments et des substances chimiques , Glucuronates/pharmacologie , Glucuronates/usage thérapeutique , Inflammation/traitement médicamenteux , Inflammation/métabolisme , Inflammation/prévention et contrôle , Inflammation/anatomopathologie , Rein/métabolisme , Rein/effets des médicaments et des substances chimiques , Rein/anatomopathologie , Macrophages/effets des médicaments et des substances chimiques , Macrophages/métabolisme , Système de signalisation des MAP kinases/effets des médicaments et des substances chimiques , Souris de lignée C57BL , Cellules RAW 264.7 , Lésion d'ischémie-reperfusion/traitement médicamenteux , Lésion d'ischémie-reperfusion/métabolisme
11.
Molecules ; 29(11)2024 Jun 01.
Article de Anglais | MEDLINE | ID: mdl-38893482

RÉSUMÉ

Apigenin, a naturally derived flavonoid, is increasingly being acknowledged for its potential therapeutic applications, especially in oncology. This research explores apigenin's capacity to modulate cancer cell viability, emphasizing its roles beyond its minimal antioxidant activity attributed to its basic molecular structure devoid of hydroxyl groups. We investigated apigenin's effects on two breast cancer cell lines, estrogen-dependent MCF-7 and non-estrogen-dependent MDA-MB-231 cells. Our findings reveal that apigenin exerts a dose-dependent cytotoxic and anti-migratory impact on these cells. Interestingly, both apigenin and doxorubicin-a standard chemotherapeutic agent-induced lipid droplet accumulation in a dose-dependent manner in MDA-MB-231 cells. This phenomenon was absent in MCF-7 cells and not evident when doxorubicin and apigenin were used concurrently, suggesting distinct cellular responses to these treatments that imply that their synergistic effects might be mediated through mechanisms unrelated to lipid metabolism. A further chemoinformatics analysis indicated that apigenin and doxorubicin might interact primarily at the level of ATP-binding cassette (ABC) transporter proteins, with potential indirect influences from the AKT and MYC signaling pathways. These results highlight the importance of understanding the nuanced interactions between apigenin and conventional chemotherapeutic drugs, as they could lead to more effective strategies for cancer treatment. This study underscores apigenin's potential as a modulator of cancer cell dynamics through mechanisms independent of its direct antioxidant effects, thereby contributing to the development of flavonoid-based adjunct therapies in cancer management.


Sujet(s)
Apigénine , Tumeurs du sein , Doxorubicine , Humains , Apigénine/pharmacologie , Apigénine/composition chimique , Doxorubicine/pharmacologie , Tumeurs du sein/traitement médicamenteux , Tumeurs du sein/métabolisme , Tumeurs du sein/anatomopathologie , Femelle , Lignée cellulaire tumorale , Survie cellulaire/effets des médicaments et des substances chimiques , Cellules MCF-7 , Mouvement cellulaire/effets des médicaments et des substances chimiques , Transduction du signal/effets des médicaments et des substances chimiques , Prolifération cellulaire/effets des médicaments et des substances chimiques , Synergie des médicaments
12.
Nat Commun ; 15(1): 5039, 2024 Jun 12.
Article de Anglais | MEDLINE | ID: mdl-38866775

RÉSUMÉ

Urate, the physiological form of uric acid and a potent antioxidant in serum, plays a pivotal role in scavenging reactive oxygen species. Yet excessive accumulation of urate, known as hyperuricemia, is the primary risk factor for the development of gout. The high-capacity urate transporter GLUT9 represents a promising target for gout treatment. Here, we present cryo-electron microscopy structures of human GLUT9 in complex with urate or its inhibitor apigenin at overall resolutions of 3.5 Å and 3.3 Å, respectively. In both structures, GLUT9 exhibits an inward open conformation, wherein the substrate binding pocket faces the intracellular side. These structures unveil the molecular basis for GLUT9's substrate preference of urate over glucose, and show that apigenin acts as a competitive inhibitor by occupying the substrate binding site. Our findings provide critical information for the development of specific inhibitors targeting GLUT9 as potential therapeutics for gout and hyperuricemia.


Sujet(s)
Apigénine , Cryomicroscopie électronique , Transporteurs de glucose par diffusion facilitée , Acide urique , Humains , Transporteurs de glucose par diffusion facilitée/métabolisme , Transporteurs de glucose par diffusion facilitée/antagonistes et inhibiteurs , Transporteurs de glucose par diffusion facilitée/composition chimique , Acide urique/métabolisme , Acide urique/composition chimique , Apigénine/pharmacologie , Apigénine/composition chimique , Sites de fixation , Liaison aux protéines , Hyperuricémie/traitement médicamenteux , Hyperuricémie/métabolisme , Modèles moléculaires , Goutte/traitement médicamenteux , Goutte/métabolisme , Cellules HEK293
13.
Sci Rep ; 14(1): 13430, 2024 06 11.
Article de Anglais | MEDLINE | ID: mdl-38862696

RÉSUMÉ

Previous studies have shown that scutellarin inhibits the excessive activation of microglia, reduces neuronal apoptosis, and exerts neuroprotective effects. However, whether scutellarin regulates activated microglia-mediated neuronal apoptosis and its mechanisms remains unclear. This study aimed to investigate whether scutellarin can attenuate PC12 cell apoptosis induced by activated microglia via the JAK2/STAT3 signalling pathway. Microglia were cultured in oxygen-glucose deprivation (OGD) medium, which acted as a conditioning medium (CM) to activate PC12 cells, to investigate the expression of apoptosis and JAK2/STAT3 signalling-related proteins. We observed that PC12 cells apoptosis in CM was significantly increased, the expression and fluorescence intensity of the pro-apoptotic protein Bax and apoptosis-related protein cleaved caspase-3 were increased, and expression of the anti-apoptotic protein B-cell lymphoma-2 (Bcl-2) was decreased. Phosphorylation levels and fluorescence intensity of the JAK2/STAT3 signalling pathway-related proteins JAK2 and STAT3 decreased. After treatment with scutellarin, PC12 cells apoptosis as well as cleaved caspase-3 and Bax protein expression and fluorescence intensity decreased. The expression and fluorescence intensity of Bcl-2, phosphorylated JAK2, and STAT3 increased. AG490, a specific inhibitor of the JAK2/STAT3 signalling pathway, was used. Our findings suggest that AG490 attenuates the effects of scutellarin. Our study revealed that scutellarin inhibited OGD-activated microglia-mediated PC12 cells apoptosis which was regulated via the JAK2/STAT3 signalling pathway.


Sujet(s)
Apigénine , Apoptose , Glucuronates , Kinase Janus-2 , Microglie , Facteur de transcription STAT-3 , Transduction du signal , Animaux , Apigénine/pharmacologie , Facteur de transcription STAT-3/métabolisme , Kinase Janus-2/métabolisme , Glucuronates/pharmacologie , Cellules PC12 , Apoptose/effets des médicaments et des substances chimiques , Microglie/effets des médicaments et des substances chimiques , Microglie/métabolisme , Transduction du signal/effets des médicaments et des substances chimiques , Rats , Souris , Caspase-3/métabolisme , Glucose/métabolisme , Neuroprotecteurs/pharmacologie , Phosphorylation/effets des médicaments et des substances chimiques , Protéine Bax/métabolisme , Tyrphostines/pharmacologie
14.
Cancer Lett ; 596: 216961, 2024 Aug 01.
Article de Anglais | MEDLINE | ID: mdl-38823764

RÉSUMÉ

Extracellular vesicles are essential for intercellular communication and are involved in tumor progression. Inhibiting the direct release of extracellular vesicles seems to be an effective strategy in inhibiting tumor progression, but lacks of investigation. Here, we report a natural flavonoid compound, apigenin, could significantly inhibit the growth of hepatocellular carcinoma by preventing microvesicle secretion. Mechanistically, apigenin primarily targets the guanine nucleotide exchange factor ARHGEF1, inhibiting the activity of small G protein Cdc42, which is essential in regulating the release of microvesicles from tumor cells. In turn, this inhibits tumor angiogenesis related to VEGF90K transported on microvesicles, ultimately impeding tumor progression. Collectively, these findings highlight the therapeutic potential of apigenin and shed light on its anticancer mechanisms through inhibiting microvesicle biogenesis, providing a solid foundation for the refinement and practical application of apigenin.


Sujet(s)
Apigénine , Carcinome hépatocellulaire , Microparticules membranaires , Tumeurs du foie , Néovascularisation pathologique , Rho guanine nucleotide exchange factors , Humains , Néovascularisation pathologique/traitement médicamenteux , Néovascularisation pathologique/métabolisme , Animaux , Apigénine/pharmacologie , Tumeurs du foie/traitement médicamenteux , Tumeurs du foie/anatomopathologie , Tumeurs du foie/métabolisme , Rho guanine nucleotide exchange factors/métabolisme , Rho guanine nucleotide exchange factors/génétique , Microparticules membranaires/métabolisme , Microparticules membranaires/effets des médicaments et des substances chimiques , Carcinome hépatocellulaire/traitement médicamenteux , Carcinome hépatocellulaire/anatomopathologie , Carcinome hépatocellulaire/métabolisme , Carcinome hépatocellulaire/vascularisation , Souris , Lignée cellulaire tumorale , Protéine G cdc42/métabolisme , Prolifération cellulaire/effets des médicaments et des substances chimiques , Tests d'activité antitumorale sur modèle de xénogreffe , Cellules endothéliales de la veine ombilicale humaine/métabolisme , Cellules endothéliales de la veine ombilicale humaine/effets des médicaments et des substances chimiques , Cellules HepG2 , Souris nude ,
15.
Biomed Pharmacother ; 177: 116986, 2024 Aug.
Article de Anglais | MEDLINE | ID: mdl-38906017

RÉSUMÉ

AIM: Apigenin, a natural bioflavonoid, is reported as an anti-diabetic agent since it possesses the ability to inhibit α-glucosidase activity, cause stimulation of insulin action and secretion, manage ROS, and prevent diabetes complications. Apigenin was identified as a new insulin secretagogue that enhances glucose-stimulated insulin secretion and seems like a better antidiabetic drug candidate. Here we explored the insulinotropic mechanism(s) of apigenin in vitro in mice islets and in vivo in diabetic rats. METHODS: Size-matched pancreatic islets were divided into groups and incubated in the presence or absence of apigenin and agonists or antagonists of major insulin signaling pathways. The secreted insulin was measured by ELISA. The intracellular cAMP was estimated by cAMP acetylation assay. The acute and chronic effects of apigenin were evaluated in diabetic rats. RESULTS: apigenin dose-dependently enhanced insulin secretion in isolated mice islets, and its insulinotropic effect was exerted at high glucose concentrations distinctly different from glibenclamide. Furthermore, apigenin amplified glucose-induced insulin secretion in depolarized and glibenclamide-treated islets. Apigenin showed no effect on intracellular cAMP concentration; however, an additive effect was observed by apigenin in both forskolin and IBMX-induced insulin secretion. Interestingly, H89, a PKA inhibitor, and U0126, a MEK kinase inhibitor, significantly inhibited apigenin-induced insulin secretion; however, no significant effect was observed by using ESI-05, an epac2 inhibitor. Apigenin improved glucose tolerance and increased glucose-stimulated plasma insulin levels in diabetic rats. Apigenin also lowered blood glucose in diabetic rats upon chronic treatment. CONCLUSION: Apigenin exerts glucose-stimulated insulin secretion by modulating the PKA-MEK kinase signaling cascade independent of K-ATP channels.


Sujet(s)
Apigénine , Cyclic AMP-Dependent Protein Kinases , Diabète expérimental , Glucose , Sécrétion d'insuline , Insuline , Animaux , Apigénine/pharmacologie , Sécrétion d'insuline/effets des médicaments et des substances chimiques , Mâle , Cyclic AMP-Dependent Protein Kinases/métabolisme , Diabète expérimental/traitement médicamenteux , Diabète expérimental/métabolisme , Glucose/métabolisme , Insuline/métabolisme , Insuline/sang , Souris , Rats , Transduction du signal/effets des médicaments et des substances chimiques , Canaux KATP/métabolisme , Système de signalisation des MAP kinases/effets des médicaments et des substances chimiques , AMP cyclique/métabolisme , Hypoglycémiants/pharmacologie , Ilots pancréatiques/effets des médicaments et des substances chimiques , Ilots pancréatiques/métabolisme , Souris de lignée C57BL , Rat Wistar , Glycémie/métabolisme , Glycémie/effets des médicaments et des substances chimiques
16.
Food Funct ; 15(13): 6988-7002, 2024 Jul 01.
Article de Anglais | MEDLINE | ID: mdl-38855818

RÉSUMÉ

A high-fat diet (HFD) is a major risk factor for cardiovascular disease. However, the specific effects of a HFD on vascular inflammation and the protective role of vitexin, a bioactive compound derived from food, require further research. This study investigated the protective effects of vitexin intervention against HFD-induced vascular inflammation and its underlying mechanism. The results demonstrated that vitexin intervention significantly reduced body weight, serum total cholesterol, and low-density lipoprotein cholesterol levels in HFD-fed mice. Vitexin also improved vascular pathological changes and the inflammatory status in the mice. Furthermore, vitexin intervention reduced serum TMAO levels in HFD-fed mice by altering the gut microbiota composition. The HFD significantly increased N6-methyladenosine (m6A) levels in aorta tissues, while vitexin intervention reversed this abnormal m6A level. Through metabolite affinity responsive target fluorescence quenching and molecular docking assays, it was found that vitexin could directly bind to fat mass and obesity-associated protein (FTO), potentially promoting m6A demethylation. The dose-response relationship between TMAO and inflammation/m6A was further validated in HUVEC cells and in vivo mouse experiments. Specifically, TMAO increased m6A levels and inflammation, while vitexin inhibited TMAO-mediated m6A modification, exhibiting anti-inflammatory effects. In conclusion, this study demonstrates the protective role of vitexin against HFD-induced vascular inflammation by inhibiting TMAO-mediated RNA m6A modification, laying the foundation for the development of functional foods.


Sujet(s)
Apigénine , Alimentation riche en graisse , Méthylamines , Souris de lignée C57BL , Animaux , Souris , Apigénine/pharmacologie , Mâle , Alimentation riche en graisse/effets indésirables , Humains , Inflammation/traitement médicamenteux , Cellules endothéliales de la veine ombilicale humaine , ARN/métabolisme , Endothélium vasculaire/effets des médicaments et des substances chimiques , Endothélium vasculaire/métabolisme ,
17.
Asian Pac J Cancer Prev ; 25(6): 2069-2075, 2024 Jun 01.
Article de Anglais | MEDLINE | ID: mdl-38918669

RÉSUMÉ

OBJECTIVE: Molecular docking is a key tool in structural molecular biology and computer-assisted drug design. Oral carcinogenesis is a complex, multistep process in which genetic events within signal transduction pathways governing normal cellular physiology are quantitatively or qualitatively altered. There are various molecular targets like Cyclin D and PI3k- alpha Ras Binding Domain receptor protein involved in the pathogenesis of Oral Squamous Cell Carcinoma. The aim of the study is to demonstrate the computer aided drug design to identify a potent natural molecule for targeting cyclin D4 and PI3K RAS binding protein. MATERIALS AND METHODS: Target selection (Cyclin D1 and PI3K-alpha Ras Binding Domain receptor) was done and structures were derived from protein data bank. Ligands (Apigenin, Chrysoeriol and Luteolin) selection was done and structure derived. Final docking was performed by Autodock. RESULTS: From the docking results it can be seen that luteolin has the highest binding energy (-5.45) with the Cyclin D receptor molecule followed by Chrysoeriol (-4.99) and Apigenin (-4.96). The binding energies of the ligands against PI3K-alpha Ras Binding Domain receptors were Apigenin (-4.51), Chrysoeriol (-4.6) and Luteolin (-4.56). CONCLUSION: The study concludes that all the three selected ligands possess high binding energy with both the target proteins involved in carcinogenesis with highest binding energy possessed by Luteolin against the Cyclin D receptor and by Chrysoeriol against PI3K-RAS binding protein. Thus their activity can be utilized to derive potential Anti-cancer therapeutic drugs.


Sujet(s)
Simulation de docking moléculaire , Tumeurs de la bouche , Composés phytochimiques , Humains , Tumeurs de la bouche/traitement médicamenteux , Tumeurs de la bouche/métabolisme , Tumeurs de la bouche/anatomopathologie , Composés phytochimiques/pharmacologie , Composés phytochimiques/composition chimique , Ligands , Marqueurs biologiques tumoraux/métabolisme , Marqueurs biologiques tumoraux/génétique , Cycline D1/métabolisme , Apigénine/pharmacologie , Apigénine/composition chimique , Carcinome épidermoïde/traitement médicamenteux , Carcinome épidermoïde/métabolisme , Carcinome épidermoïde/anatomopathologie , Lutéoline/pharmacologie , Lutéoline/composition chimique , Simulation numérique
18.
J Transl Med ; 22(1): 447, 2024 May 13.
Article de Anglais | MEDLINE | ID: mdl-38741132

RÉSUMÉ

BACKGROUND: Retinal ischemia/reperfusion (RIR) is implicated in various forms of optic neuropathies, yet effective treatments are lacking. RIR leads to the death of retinal ganglion cells (RGCs) and subsequent vision loss, posing detrimental effects on both physical and mental health. Apigenin (API), derived from a wide range of sources, has been reported to exert protective effects against ischemia/reperfusion injuries in various organs, such as the brain, kidney, myocardium, and liver. In this study, we investigated the protective effect of API and its underlying mechanisms on RGC degeneration induced by retinal ischemia/reperfusion (RIR). METHODS: An in vivo model was induced by anterior chamber perfusion following intravitreal injection of API one day prior to the procedure. Meanwhile, an in vitro model was established through 1% oxygen and glucose deprivation. The neuroprotective effects of API were evaluated using H&E staining, spectral-domain optical coherence tomography (SD-OCT), Fluoro-Gold retrograde labeling, and Photopic negative response (PhNR). Furthermore, transmission electron microscopy (TEM) was employed to observe mitochondrial crista morphology and integrity. To elucidate the underlying mechanisms of API, the terminal deoxynucleotidyl transferase dUTP nick end labeling (TUNEL) assay, flow cytometry assay, western blot, cell counting kit-8 (CCK-8) assay, lactate dehydrogenase (LDH) assay, JC-1 kit assay, dichlorofluorescein-diacetate (DCFH-DA) assay, as well as TMRE and Mito-tracker staining were conducted. RESULTS: API treatment protected retinal inner plexiform layer (IPL) and ganglion cell complex (GCC), and improved the function of retinal ganglion cells (RGCs). Additionally, API reduced RGC apoptosis and decreased lactate dehydrogenase (LDH) release by upregulating Bcl-2 and Bcl-xL expression, while downregulating Bax and cleaved caspase-3 expression. Furthermore, API increased mitochondrial membrane potential (MMP) and decreased extracellular reactive oxygen species (ROS) production. These effects were achieved by enhancing mitochondrial function, restoring mitochondrial cristae morphology and integrity, and regulating the expression of OPA1, MFN2, and DRP1, thereby regulating mitochondrial dynamics involving fusion and fission. CONCLUSION: API protects RGCs against RIR injury by modulating mitochondrial dynamics, promoting mitochondrial fusion and fission.


Sujet(s)
Apigénine , Dynamique mitochondriale , Neuroprotecteurs , Lésion d'ischémie-reperfusion , Cellules ganglionnaires rétiniennes , Cellules ganglionnaires rétiniennes/effets des médicaments et des substances chimiques , Cellules ganglionnaires rétiniennes/anatomopathologie , Cellules ganglionnaires rétiniennes/métabolisme , Apigénine/pharmacologie , Apigénine/usage thérapeutique , Animaux , Lésion d'ischémie-reperfusion/traitement médicamenteux , Lésion d'ischémie-reperfusion/anatomopathologie , Neuroprotecteurs/pharmacologie , Neuroprotecteurs/usage thérapeutique , Dynamique mitochondriale/effets des médicaments et des substances chimiques , Mâle , Apoptose/effets des médicaments et des substances chimiques , Mitochondries/effets des médicaments et des substances chimiques , Mitochondries/métabolisme , Espèces réactives de l'oxygène/métabolisme , Modèles biologiques , Souris de lignée C57BL
19.
Biomolecules ; 14(5)2024 May 13.
Article de Anglais | MEDLINE | ID: mdl-38785983

RÉSUMÉ

BACKGROUND: Peroxynitrite (ONOO-) is an oxidant linked with several human pathologies. Apigenin, a natural flavonoid known for its health benefits, remains unexplored in relation to ONOO- effects. This study investigated the potential of apigenin to structurally protect fibrinogen, an essential blood clotting factor, from ONOO--induced damage. METHODS: Multi-approach analyses were carried out where fibrinogen was exposed to ONOO- generation while testing the efficacy of apigenin. The role of apigenin against ONOO--induced modifications in fibrinogen was investigated using UV spectroscopy, tryptophan or tyrosine fluorescence, protein hydrophobicity, carbonylation, and electrophoretic analyses. RESULTS: The findings demonstrate that apigenin significantly inhibits ONOO--induced oxidative damage in fibrinogen. ONOO- caused reduced UV absorption, which was reversed by apigenin treatment. Moreover, ONOO- diminished tryptophan and tyrosine fluorescence, which was effectively restored by apigenin treatment. Apigenin also reduced the hydrophobicity of ONOO--damaged fibrinogen. Moreover, apigenin exhibited protective effects against ONOO--induced protein carbonylation. SDS-PAGE analyses revealed that ONOO-treatment eliminated bands corresponding to fibrinogen polypeptide chains Aα and γ, while apigenin preserved these changes. CONCLUSIONS: This study highlights, for the first time, the role of apigenin in structural protection of human fibrinogen against peroxynitrite-induced nitrosative damage. Our data indicate that apigenin offers structural protection to all three polypeptide chains (Aα, Bß, and γ) of human fibrinogen. Specifically, apigenin prevents the dislocation or breakdown of the amino acids tryptophan, tyrosine, lysine, arginine, proline, and threonine and also prevents the exposure of hydrophobic sites in fibrinogen induced by ONOO-.


Sujet(s)
Apigénine , Fibrinogène , Stress nitrosatif , Acide peroxynitreux , Fibrinogène/métabolisme , Fibrinogène/composition chimique , Apigénine/pharmacologie , Apigénine/composition chimique , Humains , Acide peroxynitreux/composition chimique , Stress nitrosatif/effets des médicaments et des substances chimiques , Interactions hydrophobes et hydrophiles , Carbonylation des protéines/effets des médicaments et des substances chimiques , Tyrosine/composition chimique , Tyrosine/métabolisme , Stress oxydatif/effets des médicaments et des substances chimiques
20.
Int J Mol Sci ; 25(10)2024 May 20.
Article de Anglais | MEDLINE | ID: mdl-38791608

RÉSUMÉ

Due to its propensity to metastasize, cancer remains one of the leading causes of death worldwide. Thanks in part to their intrinsic low cytotoxicity, the effects of the flavonoid family in the prevention and treatment of various human cancers, both in vitro and in vivo, have received increasing attention in recent years. It is well documented that Apigenin (4',5,7-trihydroxyflavone), among other flavonoids, is able to modulate key signaling molecules involved in the initiation of cancer cell proliferation, invasion, and metastasis, including JAK/STAT, PI3K/Akt/mTOR, MAPK/ERK, NF-κB, and Wnt/ß-catenin pathways, as well as the oncogenic non-coding RNA network. Based on these premises, the aim of this review is to emphasize some of the key events through which Apigenin suppresses cancer proliferation, focusing specifically on its ability to target key molecular pathways involved in angiogenesis, epithelial-to-mesenchymal transition (EMT), maintenance of cancer stem cells (CSCs), cell cycle arrest, and cancer cell death.


Sujet(s)
Apigénine , Transition épithélio-mésenchymateuse , Tumeurs , Apigénine/pharmacologie , Apigénine/usage thérapeutique , Humains , Tumeurs/traitement médicamenteux , Tumeurs/métabolisme , Tumeurs/anatomopathologie , Animaux , Transition épithélio-mésenchymateuse/effets des médicaments et des substances chimiques , Transduction du signal/effets des médicaments et des substances chimiques , Prolifération cellulaire/effets des médicaments et des substances chimiques , Cellules souches tumorales/effets des médicaments et des substances chimiques , Cellules souches tumorales/métabolisme , Néovascularisation pathologique/traitement médicamenteux , Néovascularisation pathologique/métabolisme
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE