Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 20 de 4.469
Filtrer
1.
Sci Rep ; 14(1): 15339, 2024 07 03.
Article de Anglais | MEDLINE | ID: mdl-38961115

RÉSUMÉ

Given the hierarchical nature of bone and bone interfaces, osseointegration, namely the formation of a direct bone-implant contact, is best evaluated using a multiscale approach. However, a trade-off exists between field of view and spatial resolution, making it challenging to image large volumes with high resolution. In this study, we combine established electron microscopy techniques to probe bone-implant interfaces at the microscale and nanoscale with plasma focused ion beam-scanning electron microscopy (PFIB-SEM) tomography to evaluate osseointegration at the mesoscale. This characterization workflow is demonstrated for bone response to an additively manufactured Ti-6Al-4V implant which combines engineered porosity to facilitate bone ingrowth and surface functionalization via genistein, a phytoestrogen, to counteract bone loss in osteoporosis. SEM demonstrated new bone formation at the implant site, including in the internal implant pores. At the nanoscale, scanning transmission electron microscopy and energy-dispersive X-ray spectroscopy confirmed the gradual nature of the bone-implant interface. By leveraging mesoscale analysis with PFIB-SEM tomography that captures large volumes of bone-implant interface with nearly nanoscale resolution, the presence of mineral ellipsoids varying in size and orientation was revealed. In addition, a well-developed lacuno-canalicular network and mineralization fronts directed both towards the implant and away from it were highlighted.


Sujet(s)
Génistéine , Ostéo-intégration , Titane , Ostéo-intégration/effets des médicaments et des substances chimiques , Génistéine/pharmacologie , Génistéine/composition chimique , Titane/composition chimique , Animaux , Matériaux revêtus, biocompatibles/composition chimique , Interface os-implant , Microscopie électronique à balayage , Prothèses et implants , Porosité , Alliages/composition chimique
2.
Drug Deliv ; 31(1): 2372277, 2024 Dec.
Article de Anglais | MEDLINE | ID: mdl-38952058

RÉSUMÉ

Skin melanoma is considered the most dangerous form of skin cancer due to its association with high risk of metastasis, high mortality rate and high resistance to different treatment options. Genistein is a natural isoflavonoid with known chemotherapeutic activity. Unfortunately, it has low bioavailability due to its poor aqueous solubility and excessive metabolism. In the current study, genistein was incorporated into transferosomal hydrogel to improve its bioavailability. The prepared transferosomal formulations were characterized regarding: particle size; polydispersity index; zeta potential; encapsulation efficiency; TEM; FTIR; DSC; XRD; in vitro drug release; viscosity; pH; ex vivo anti-tumor activity on 3D skin melanoma spheroids and 1-year stability study at different storage temperatures. The optimized formulation has high encapsulation efficiency with an excellent particle size that will facilitate its penetration through the skin. The transfersomes have a spherical shape with sustained drug release profile. The anti-tumor activity evaluation of genistein transfersome revealed that genistein is a potent chemotherapeutic agent with enhanced penetration ability through the melanoma spheroids when incorporated into transfersomes. Stability study results demonstrate the high physical and chemical stability of our formulations. All these outcomes provide evidence that our genistein transferosomal hydrogel is a promising treatment option for skin melanoma.


Sujet(s)
Libération de médicament , Génistéine , Hydrogels , Mélanome , Taille de particule , Tumeurs cutanées , Génistéine/administration et posologie , Génistéine/pharmacologie , Génistéine/pharmacocinétique , Mélanome/traitement médicamenteux , Tumeurs cutanées/traitement médicamenteux , Humains , Hydrogels/composition chimique , Systèmes de délivrance de médicaments/méthodes , Lignée cellulaire tumorale , Stabilité de médicament , Antinéoplasiques/administration et posologie , Antinéoplasiques/pharmacologie , Antinéoplasiques/pharmacocinétique , Solubilité , Vecteurs de médicaments/composition chimique , Chimie pharmaceutique , Viscosité , Biodisponibilité , Administration par voie cutanée , Sphéroïdes de cellules/effets des médicaments et des substances chimiques
3.
Mar Drugs ; 22(6)2024 Jun 13.
Article de Anglais | MEDLINE | ID: mdl-38921587

RÉSUMÉ

Deep-sea environments, as relatively unexplored extremes within the Earth's biosphere, exhibit notable distinctions from terrestrial habitats. To thrive in these extreme conditions, deep-sea actinomycetes have evolved unique biochemical metabolisms and physiological capabilities to ensure their survival in this niche. In this study, five actinomycetes strains were isolated and identified from the Mariana Trench via the culture-dependent method and 16S rRNA sequencing approach. The antimicrobial activity of Microbacterium sp. B1075 was found to be the most potent, and therefore, it was selected as the target strain. Molecular networking analysis via the Global Natural Products Social Molecular Networking (GNPS) platform identified 25 flavonoid compounds as flavonoid secondary metabolites. Among these, genistein was purified and identified as a bioactive compound with significant antibacterial activity. The complete synthesis pathway for genistein was proposed within strain B1075 based on whole-genome sequencing data, with the key gene being CHS (encoding chalcone synthase). The expression of the gene CHS was significantly regulated by high hydrostatic pressure, with a consequent impact on the production of flavonoid compounds in strain B1075, revealing the relationship between actinomycetes' synthesis of flavonoid-like secondary metabolites and their adaptation to high-pressure environments at the molecular level. These results not only expand our understanding of deep-sea microorganisms but also hold promise for providing valuable insights into the development of novel pharmaceuticals in the field of biopharmaceuticals.


Sujet(s)
Antibactériens , Génistéine , Génistéine/pharmacologie , Génistéine/métabolisme , Antibactériens/pharmacologie , Antibactériens/biosynthèse , Microbacterium , ARN ribosomique 16S/génétique , Actinobacteria/métabolisme , Actinobacteria/génétique , Métabolisme secondaire , Phylogenèse , Acyltransferases
4.
Int J Mol Sci ; 25(12)2024 Jun 17.
Article de Anglais | MEDLINE | ID: mdl-38928362

RÉSUMÉ

The aryl hydrocarbon receptor (AhR) is a transcription factor that regulates the immune system through complicated transcriptional programs. Genistein, an AhR ligand, exhibits anti-inflammatory properties. However, its role in modulating immune responses via the AhR signaling pathway remains unclear. In this study, 360 male Arbor Acre broilers (1-day-old) were fed a basal diet supplemented with 40 or 80 mg/kg genistein and infected with or without Clostridium perfringens (Cp). Our results demonstrated that genistein ameliorated Cp-induced intestinal damage, as reflected by the reduced intestinal lesion scores and improved intestinal morphology and feed-to-gain ratio. Moreover, genistein increased intestinal sIgA, TGF-ß, and IL-10, along with elevated serum IgG, IgA, and lysozyme levels. Genistein improved intestinal AhR and cytochrome P450 family 1 subfamily A member 1 (CYP1A1) protein levels and AhR+ cell numbers in Cp-challenged broilers. The increased number of AhR+CD163+ cells in the jejunum suggested a potential association between genistein-induced AhR activation and anti-inflammatory effects mediated through M2 macrophage polarization. In IL-4-treated RAW264.7 cells, genistein increased the levels of AhR, CYP1A1, CD163, and arginase (Arg)-1 proteins, as well as IL-10 mRNA levels. This increase was attenuated by the AhR antagonist CH223191. In summary, genistein activated the AhR signaling pathway in M2 macrophages, which enhanced the secretion of anti-inflammatory cytokines and attenuated intestinal damage in Cp-infected broilers Cp.


Sujet(s)
Poulets , Entérite , Génistéine , Macrophages , Récepteurs à hydrocarbure aromatique , Animaux , Récepteurs à hydrocarbure aromatique/métabolisme , Génistéine/pharmacologie , Génistéine/usage thérapeutique , Macrophages/effets des médicaments et des substances chimiques , Macrophages/métabolisme , Souris , Entérite/traitement médicamenteux , Entérite/métabolisme , Mâle , Cellules RAW 264.7 , Maladies de la volaille/traitement médicamenteux , Maladies de la volaille/métabolisme , Intestins/effets des médicaments et des substances chimiques , Intestins/anatomopathologie , Clostridium perfringens , Infections à Clostridium/traitement médicamenteux , Nécrose , Activation des macrophages/effets des médicaments et des substances chimiques , Inflammation/traitement médicamenteux , Inflammation/anatomopathologie , Inflammation/métabolisme , Transduction du signal/effets des médicaments et des substances chimiques
5.
Curr Pharm Biotechnol ; 25(7): 807-824, 2024.
Article de Anglais | MEDLINE | ID: mdl-38902930

RÉSUMÉ

Throughout the past several centuries, herbal constituents have been the subject of scientific interest and the latest research into their therapeutic potential is underway. Genistein is a soy-derived isoflavone found in huge amounts in soy, along with the plants of the Fabaceae family. Scientific studies have demonstrated the beneficial effects of genistein on various health conditions. Genistein presents a broad range of pharmacological activities, including anticancer, neuroprotective, cardioprotective, antiulcer, anti-diabetic, wound healing, anti-bacterial, antiviral, skin, and radioprotective effects. However, the hydrophobic nature of genistein results in constrained absorption and restricts its therapeutic potential. In this review, the number of nanocarriers for genistein delivery has been explored, such as polymeric nanoparticles, nanostructured lipid carriers, solid lipid nanoparticles, liposomes, micelles, transferosomes, and nanoemulsions and nanofibers. These nano-formulations of genistein have been utilized as a potential strategy for various disorders, employing a variety of ex vivo, in vitro, and in vivo models and various administration routes. This review concluded that genistein is a potential therapeutic agent for treating various diseases, including cancer, neurodegenerative disorders, cardiovascular disorders, obesity, diabetes, ulcers, etc., when formulated in suitable nanocarriers.


Sujet(s)
Génistéine , Nanoparticules , Génistéine/pharmacologie , Génistéine/usage thérapeutique , Génistéine/composition chimique , Humains , Animaux , Nanoparticules/composition chimique , Nanotechnologie/méthodes , Vecteurs de médicaments/composition chimique , Systèmes de délivrance de médicaments/méthodes , Tumeurs/traitement médicamenteux
6.
Nutrients ; 16(11)2024 May 24.
Article de Anglais | MEDLINE | ID: mdl-38892535

RÉSUMÉ

Rice bean [Vigna umbellata (Thunb.) Ohwi and Ohashi], an annual legume in the genus Vigna, is a promising crop suitable for cultivation in a changing climate to ensure food security. It is also a medicinal plant widely used in traditional Chinese medicine; however, little is known about the medicinal compounds in rice bean. In this study, we assessed the diuretic effect of rice bean extracts on mice as well as its relationship with the contents of eight secondary metabolites in seeds. Mice gavaged with rice bean extracts from yellow and black seeds had higher urinary output (5.44-5.47 g) and water intake (5.8-6.3 g) values than mice gavaged with rice bean extracts from red seeds. Correlation analyses revealed significant negative correlations between urine output and gallic acid (R = -0.70) and genistein (R = -0.75) concentrations, suggesting that these two polyphenols negatively regulate diuresis. There were no obvious relationships between mice diuresis-related indices (urine output, water intake, and weight loss) and rutin or catechin contents, although the concentrations of both of these polyphenols in rice bean seeds were higher than the concentrations of the other six secondary metabolites. Our study findings may be useful for future research on the diuretic effects of rice bean, but they should be confirmed on the basis of systematic medical trials.


Sujet(s)
Diurétiques , Polyphénols , Graines , Animaux , Souris , Diurétiques/pharmacologie , Graines/composition chimique , Polyphénols/pharmacologie , Polyphénols/analyse , Mâle , Extraits de plantes/pharmacologie , Vigna/composition chimique , Acide gallique/pharmacologie , Génistéine/pharmacologie , Catéchine/pharmacologie , Catéchine/analyse , Rutoside/pharmacologie , Rutoside/analyse , Diurèse/effets des médicaments et des substances chimiques
7.
Int J Biol Macromol ; 270(Pt 1): 132296, 2024 Jun.
Article de Anglais | MEDLINE | ID: mdl-38740159

RÉSUMÉ

Glycerol kinase (GK) and glycerol 3-phosphate dehydrogenase (GPDH) are critical in glucose homeostasis. The role of genistein and metformin on these enzymes and glucose production was investigated in C2C12, HepG2, and 3T3-L1 cells. Enzyme kinetics, Real-Time PCR and western blots were performed to determine enzyme activities and expressions of mRNAs and proteins. Glucose production and uptake were also measured in these cells. siRNAs were used to assess their impact on the enzymes and glucose production. Ki values for the compounds were determined using purified GK and GPDH. Genistein decreased GK activity by ∼45 %, while metformin reduced cGPDH and mGPDH activities by ∼32 % and âˆ¼43 %, respectively. Insignificant changes in expressions (mRNAs and proteins) of the enzymes were observed. The compounds showed dose-dependent alterations in glucose production and uptake in these cells. Genistein non-competitively inhibited His-GK activity (Ki 19.12 µM), while metformin non-competitively inhibited His-cGPDH (Ki 75.52 µM) and mGPDH (Ki 54.70 µM) activities. siRNAs transfection showed ∼50 % and âˆ¼35 % decrease in activities of GK and mGPDH and a decrease in glucose production (0.38-fold and 0.42-fold) in 3T3-L1 cells. Considering the differential effects of the compounds, this study may provide insights into the potential therapeutic strategies for type II diabetes mellitus.


Sujet(s)
Adipocytes , Génistéine , Glucose , Glycerol kinase , Glycerolphosphate dehydrogenase , Hépatocytes , Metformine , Génistéine/pharmacologie , Metformine/pharmacologie , Souris , Animaux , Glycerol kinase/métabolisme , Glycerol kinase/génétique , Hépatocytes/effets des médicaments et des substances chimiques , Hépatocytes/métabolisme , Humains , Adipocytes/effets des médicaments et des substances chimiques , Adipocytes/métabolisme , Glycerolphosphate dehydrogenase/métabolisme , Glycerolphosphate dehydrogenase/génétique , Glucose/métabolisme , Cellules 3T3-L1 , Cellules HepG2 , Glycérophosphate/métabolisme , Glycérophosphate/pharmacologie , Cinétique
8.
Int J Mol Sci ; 25(10)2024 May 20.
Article de Anglais | MEDLINE | ID: mdl-38791595

RÉSUMÉ

The most common malignancy in women is breast cancer. During the development of cancer, oncogenic transcription factors facilitate the overproduction of inflammatory cytokines and cell adhesion molecules. Antiapoptotic proteins are markedly upregulated in cancer cells, which promotes tumor development, metastasis, and cell survival. Promising findings have been found in studies on the cell cycle-mediated apoptosis pathway for medication development and treatment. Dietary phytoconstituents have been studied in great detail for their potential to prevent cancer by triggering the body's defense mechanisms. The underlying mechanisms of action may be clarified by considering the role of polyphenols in important cancer signaling pathways. Phenolic acids, flavonoids, tannins, coumarins, lignans, lignins, naphthoquinones, anthraquinones, xanthones, and stilbenes are examples of natural chemicals that are being studied for potential anticancer drugs. These substances are also vital for signaling pathways. This review focuses on innovations in the study of polyphenol genistein's effects on breast cancer cells and presents integrated chemical biology methods to harness mechanisms of action for important therapeutic advances.


Sujet(s)
Tumeurs du sein , Génistéine , Transduction du signal , Humains , Génistéine/pharmacologie , Tumeurs du sein/métabolisme , Tumeurs du sein/traitement médicamenteux , Tumeurs du sein/anatomopathologie , Femelle , Transduction du signal/effets des médicaments et des substances chimiques , Apoptose/effets des médicaments et des substances chimiques , Animaux , Polyphénols/pharmacologie , Polyphénols/composition chimique
9.
J Biochem Mol Toxicol ; 38(6): e23738, 2024 Jun.
Article de Anglais | MEDLINE | ID: mdl-38764152

RÉSUMÉ

Ulcerative colitis (UC) is a chronic problem of the intestine and relapsing in nature. Biochanin A is a nature-derived isoflavonoid and has numerous bioactivities. However, its role against UC and intestinal inflammation remains obscure. We aimed to comprehensively explore the pharmacological effect of biochanin A in alleviating colitis and to evaluate the potential mechanisms. Initially, we explored the anti-inflammatory action of biochanin A (15, 30, and 60 µM) by employing lipopolysaccharide (LPS)-activated RAW 264.7 cells. In RAW 264.7 cells under LPS stimulation, biochanin A inhibited the elevation of reactive oxygen species (ROS) (p < 0.0001), interleukin (IL)-1ß (p < 0.0001), IL-18 (p < 0.01), and tumor necrosis factor (TNF)-α (p < 0.01) release, nitrite production (p < 0.0001), and the expression of inducible nitric oxide synthase (iNOS) and cyclooxygenase-2 (COX-2) proteins. Next, we studied the effectiveness of biochanin A (20 and 40 mg/kg) in mouse colitis induced with dextran sulfate sodium (DSS) by assessing colon length, disease activity index (DAI) scoring, and performing colonoscopy and histological analysis. The pro-inflammatory cytokines were estimated using ELISA. Western blot studies were performed to assess underlying mechanisms. In mice, biochanin A treatment alleviated DAI score (p < 0.0001), restored colon length (p < 0.05) and morphology, and re-established colon histopathology. Biochanin A affects the phosphorylation of proteins associated with NF-κB (p65) and mitogen-activated protein kinase (MAPK) axis and regulates colonic inflammation by reducing the expression of inflammatory cytokines and myeloperoxidase (MPO) activity. Altogether, our findings support the idea that the anticolitis potential of biochanin A is allied with anti-inflammatory activity by inhibiting the MAPK/NF-κB (p65) axis. Hence, biochanin A may be an alternative option to alleviate the risk of colitis.


Sujet(s)
Rectocolite hémorragique , Génistéine , Facteur de transcription RelA , Animaux , Génistéine/pharmacologie , Souris , Cellules RAW 264.7 , Rectocolite hémorragique/induit chimiquement , Rectocolite hémorragique/traitement médicamenteux , Rectocolite hémorragique/métabolisme , Rectocolite hémorragique/anatomopathologie , Facteur de transcription RelA/métabolisme , Mâle , Système de signalisation des MAP kinases/effets des médicaments et des substances chimiques , Mitogen-Activated Protein Kinases/métabolisme , Inflammation/traitement médicamenteux , Inflammation/métabolisme , Inflammation/anatomopathologie , Sulfate dextran/toxicité
10.
Molecules ; 29(9)2024 Apr 23.
Article de Anglais | MEDLINE | ID: mdl-38731403

RÉSUMÉ

Food supplements have become beneficial as adjuvant therapies for many chronic disorders, including cancer. Genistein, a natural isoflavone enriched in soybeans, has gained potential interest as an anticancer agent for various cancers, primarily by modulating apoptosis, the cell cycle, and angiogenesis and inhibiting metastasis. However, in lung cancer, the exact impact and mechanism of action of genistein still require clarification. To provide more insight into the mechanism of action of genistein, network pharmacology was employed to identify the key targets and their roles in lung cancer pathogenesis. Based on the degree score, the hub genes AKT1, CASP3, EGFR, STAT3, ESR1, SRC, PTGS2, MMP9, PRAG, and AR were significantly correlated with genistein treatment. AKT1, EGFR, and STAT3 were enriched in the non-small cell lung cancer (NSCLC) pathway according to Kyoto Encyclopedia of Genes and Genomes analysis, indicating a significant connection to lung cancer development. Moreover, the binding affinity of genistein to NSCLC target proteins was further verified by molecular docking and molecular dynamics simulations. Genistein exhibited potential binding to AKT1, which is involved in apoptosis, cell migration, and metastasis, thus holding promise for modulating AKT1 function. Therefore, this study aimed to investigate the mechanism of action of genistein and its therapeutic potential for the treatment of NSCLC.


Sujet(s)
Génistéine , Tumeurs du poumon , Simulation de dynamique moléculaire , Pharmacologie des réseaux , Génistéine/pharmacologie , Génistéine/composition chimique , Humains , Tumeurs du poumon/traitement médicamenteux , Tumeurs du poumon/métabolisme , Tumeurs du poumon/anatomopathologie , Simulation de docking moléculaire , Carcinome pulmonaire non à petites cellules/traitement médicamenteux , Carcinome pulmonaire non à petites cellules/métabolisme , Carcinome pulmonaire non à petites cellules/anatomopathologie , Protéines proto-oncogènes c-akt/métabolisme , Régulation de l'expression des gènes tumoraux/effets des médicaments et des substances chimiques
11.
Eur J Pharmacol ; 975: 176636, 2024 Jul 15.
Article de Anglais | MEDLINE | ID: mdl-38729417

RÉSUMÉ

Endothelial cells express multiple receptors mediating estrogen responses; including the G protein-coupled estrogen receptor (GPER). Past studies on nitric oxide (NO) production elicited by estrogens raised the question whether 17-ß-estradiol (E2) and natural phytoestrogens activate equivalent mechanisms. We hypothesized that E2 and phytoestrogens elicit NO production via coupling to distinct intracellular pathways signalling. To this aim, perfusion of E2 and phytoestrogens to the precontracted rat mesentery bed examined vasorelaxation, while fluorescence microscopy on primary endothelial cells cultures quantified single cell NO production determined following 4-amino-5-methylamino-2',7'-difluoroescein diacetate (DAF) incubation. Daidzein (DAI) and genistein (GEN) induced rapid vasodilatation associated to NO production. Multiple estrogen receptor activity was inferred based on the reduction of DAF-NO signals; G-36 (GPER antagonist) reduced 75 % of all estrogen responses, while fulvestrant (selective nuclear receptor antagonist) reduced significantly more the phytoestrogens responses than E2. The joint application of both antagonists abolished the E2 response but not the phytoestrogen-induced DAF-NO signals. Wortmannin or LY-294002 (PI3K inhibitors), reduced by 90% the E2-evoked signal while altering significantly less the DAI-induced response. In contrast, H-89 (PKA inhibitor), elicited a 23% reduction of the E2-induced signal while blocking 80% of the DAI-induced response. Desmethylxestospongin-B (IP3 receptor antagonist), decreased to equal extent the E2 or the DAI-induced signal. Epidermal growth factor (EGF) induced NO production, cell treatment with AG-1478, an EGF receptor kinase inhibitor reduced 90% DAI-induced response while only 53% the E2-induced signals; highlighting GPER induced EGF receptor trans-modulation. Receptor functional selectivity may explain distinct signalling pathways mediated by E2 and phytoestrogens.


Sujet(s)
Cyclic AMP-Dependent Protein Kinases , Récepteurs ErbB , Oestradiol , Monoxyde d'azote , Phosphatidylinositol 3-kinases , Phyto-oestrogènes , Transduction du signal , Vasodilatation , Animaux , Phyto-oestrogènes/pharmacologie , Oestradiol/pharmacologie , Monoxyde d'azote/métabolisme , Rats , Transduction du signal/effets des médicaments et des substances chimiques , Vasodilatation/effets des médicaments et des substances chimiques , Cyclic AMP-Dependent Protein Kinases/métabolisme , Phosphatidylinositol 3-kinases/métabolisme , Récepteurs ErbB/métabolisme , Mâle , Isoflavones/pharmacologie , Cellules endothéliales/effets des médicaments et des substances chimiques , Cellules endothéliales/métabolisme , Génistéine/pharmacologie , Récepteurs des oestrogènes/métabolisme , Rat Wistar
12.
Anticancer Res ; 44(6): 2307-2323, 2024 Jun.
Article de Anglais | MEDLINE | ID: mdl-38821627

RÉSUMÉ

BACKGROUND/AIM: Pancreatic cancer is an aggressive type of cancer, with a dismally low survival rate of <5%. FDA-approved drugs like gemcitabine have shown little therapeutic success, prolonging survival by a mere six months. Isoflavones, such as biochanin A and daidzein, are known to exhibit anti-cancer activity, whereas statins reportedly have anti-proliferative effects. This study investigated the effects of combination treatment of biochanin A and atorvastatin on pancreatic cancer cells. MATERIALS AND METHODS: Pancreatic cancer cells AsPC-1, PANC-1, and MIA PaCa-2 were procured from ATCC. The cell viability studies were carried out using MTT & cell count assays. Flow cytometry was used to study cell apoptosis whereas cell metabolism studies were carried out using the Seahorse Mito stress test and XF-PMP assay. The effects of treatment on cell signaling pathways & cell cycle associated proteins were investigated using western blot whereas invasiveness of cancer cells was evaluated using gelatin zymography. RESULTS: The combination treatment decreased the survival and enhanced pro-apoptotic responses compared to single treatments in the pancreatic cancer cells. In PANC-1 cells, the combination treatment decreased invasiveness, reduced expression of activated STAT3 and expression of critical mediators of cell cycle progression. Furthermore, the combination treatment induced a differential inhibition of respiratory complexes in the pancreatic cancer cells. CONCLUSION: The combination treatment of biochanin A and atorvastatin exerts enhanced anti-cancer effects, inducing apoptosis, down-regulating cell cycle associated proteins and invasiveness in pancreatic cancer cells and merits further investigation for new, improved treatments for pancreatic cancer.


Sujet(s)
Apoptose , Atorvastatine , Points de contrôle du cycle cellulaire , Métabolisme énergétique , Génistéine , Mitochondries , Tumeurs du pancréas , Humains , Génistéine/pharmacologie , Tumeurs du pancréas/traitement médicamenteux , Tumeurs du pancréas/métabolisme , Tumeurs du pancréas/anatomopathologie , Atorvastatine/pharmacologie , Lignée cellulaire tumorale , Mitochondries/effets des médicaments et des substances chimiques , Mitochondries/métabolisme , Points de contrôle du cycle cellulaire/effets des médicaments et des substances chimiques , Apoptose/effets des médicaments et des substances chimiques , Métabolisme énergétique/effets des médicaments et des substances chimiques , Prolifération cellulaire/effets des médicaments et des substances chimiques , Survie cellulaire/effets des médicaments et des substances chimiques , Protocoles de polychimiothérapie antinéoplasique/pharmacologie , Transduction du signal/effets des médicaments et des substances chimiques
13.
Molecules ; 29(8)2024 Apr 10.
Article de Anglais | MEDLINE | ID: mdl-38675531

RÉSUMÉ

Glycolytic overload promotes accumulation of the highly reactive dicarbonyl compounds, resulting in harmful conditions called dicarbonyl stress. Methylglyoxal (MG) is a highly reactive dicarbonyl species and its accumulation plays a crucial pathophysiological role in diabetes and its vascular complications. MG cytotoxicity is mediated by reactive oxygen species (ROS) generation, a key event underlying the intracellular signaling pathways leading to inflammation and apoptosis. The identification of compounds able to inhibit ROS signaling pathways and counteract the MG-induced toxicity is a crucial step for developing new therapeutic strategies in the treatment of diabetic vascular complications. In this study, the effect of genistein, a natural soybean isoflavone, has been evaluated on MG-induced cytotoxicity in human endothelial cells. Our results show that genistein is able to counteract the MG-induced apoptosis by restraining ROS production, thus inhibiting the MAPK signaling pathways and caspase-3 activation. These findings identify a beneficial role for genistein, providing new insights for its potential clinical applications in preserving endothelial function in diabetic vascular complications.


Sujet(s)
Apoptose , Cellules endothéliales , Génistéine , Stress oxydatif , Méthylglyoxal , Espèces réactives de l'oxygène , Génistéine/pharmacologie , Méthylglyoxal/métabolisme , Humains , Apoptose/effets des médicaments et des substances chimiques , Stress oxydatif/effets des médicaments et des substances chimiques , Espèces réactives de l'oxygène/métabolisme , Cellules endothéliales/effets des médicaments et des substances chimiques , Cellules endothéliales/métabolisme , Cellules endothéliales de la veine ombilicale humaine/effets des médicaments et des substances chimiques , Cellules endothéliales de la veine ombilicale humaine/métabolisme , Caspase-3/métabolisme , Transduction du signal/effets des médicaments et des substances chimiques , Système de signalisation des MAP kinases/effets des médicaments et des substances chimiques
14.
Poult Sci ; 103(6): 103670, 2024 Jun.
Article de Anglais | MEDLINE | ID: mdl-38598909

RÉSUMÉ

Aging is associated with alterations in gut function, including intestinal inflammation, leaky gut, and impaired epithelial regeneration. Rejuvenating the aged gut is imperative to extend the laying cycle of aged laying hens. Genistein is known to have beneficial effects on age-related diseases, but its precise role in homeostasis of the aged gut of laying hens remains to be elucidated. In this study, 160 45-wk-old Hyline Brown laying hens were continuously fed a basal diet or a diet supplemented with 40 mg/kg genistein until they reached 100 wk of age. The results revealed that long-term genistein supplementation led to an improvement in the egg production rate and feed conversion ratio, as well as an increase in egg quality. Moreover, the expression levels of senescence markers, such as ß-galactosidase, P16, and P21, were decreased in the gut of genistein-treated aged laying hens. Furthermore, genistein ameliorated gut dysfunctions, such as intestinal inflammation, leaky gut, and impaired epithelial regeneration. Treg cell-derived IL-10 plays a crucial role in the genistein-induced regulation of age-related intestinal inflammation. This study demonstrates that long-term consumption of genistein improves homeostasis in the aged gut and extends the laying cycle of aged laying hens. Moreover, the link between genistein and Treg cells provides a rationale for dietary intervention against age-associated gut dysfunction.


Sujet(s)
Vieillissement , Aliment pour animaux , Poulets , Régime alimentaire , Compléments alimentaires , Génistéine , Homéostasie , Animaux , Génistéine/pharmacologie , Génistéine/administration et posologie , Poulets/physiologie , Poulets/immunologie , Femelle , Homéostasie/effets des médicaments et des substances chimiques , Compléments alimentaires/analyse , Régime alimentaire/médecine vétérinaire , Aliment pour animaux/analyse , Répartition aléatoire
15.
Chem Biol Interact ; 395: 111014, 2024 May 25.
Article de Anglais | MEDLINE | ID: mdl-38648921

RÉSUMÉ

There is an increasing appreciation that colonic barrier function is closely related to the development and progression of colitis. The mucus layer is a crucial component of the colonic barrier, responsible for preventing harmful bacteria from invading the intestinal epithelium and causing inflammation. Furthermore, a defective mucus barrier is also a significant characteristic of ulcerative colitis (UC). Biochanin A (BCA), an isoflavonoid, has garnered increasing interest due to its significant biological activities. However, the impact of BCA on UC has not been reported yet. In this study, we used a dextran sodium sulfate (DSS)-induced ulcerative colitis model and the Muc2 deficient (Muc2-/-) mice spontaneous colitis model to explore the mechanisms of BCA in the treatment of UC. Here, we verified that DSS-induced UC was observably attenuated and spontaneous colitis in Muc2-/- mice was relieved by BCA. Treatment with BCA improved colitis-related symptoms and reduced intestinal permeability by upregulating the levels of goblet cells and tight junction (TJ) proteins. In addition, we confirmed that BCA promotes autophagy through the AMP-activated protein kinase (AMPK)/mammalian target of rapamycin (mTOR)/Unc-51-like kinase 1 (ULK1) pathway, thereby alleviating DSS-induced UC. In addition, the administration of BCA was able to reduce apoptosis and promote proliferation by suppressing Cleaved Caspase-3 (Cleaved Cas-3) expression, and increasing PCNA and Ki67 levels. Further research revealed that BCA treatment ameliorated spontaneous colitis and alleviated epithelial damage in Muc2-/- mice by restoring the intestinal barrier and promoting autophagy. Our results demonstrated that BCA alleviated UC by enhancing intestinal barrier function and promoting autophagy. These findings indicate that BCA may be a novel treatment alternative for UC.


Sujet(s)
Rectocolite hémorragique , Côlon , Sulfate dextran , Génistéine , Mucine-2 , Animaux , Mucine-2/métabolisme , Mucine-2/génétique , Sulfate dextran/toxicité , Rectocolite hémorragique/induit chimiquement , Rectocolite hémorragique/traitement médicamenteux , Rectocolite hémorragique/anatomopathologie , Rectocolite hémorragique/métabolisme , Génistéine/pharmacologie , Génistéine/usage thérapeutique , Souris , Côlon/anatomopathologie , Côlon/effets des médicaments et des substances chimiques , Côlon/métabolisme , Autophagie/effets des médicaments et des substances chimiques , Souris de lignée C57BL , Modèles animaux de maladie humaine , Souris knockout , Apoptose/effets des médicaments et des substances chimiques , Muqueuse intestinale/effets des médicaments et des substances chimiques , Muqueuse intestinale/métabolisme , Muqueuse intestinale/anatomopathologie , Mâle , AMP-Activated Protein Kinases/métabolisme , Sérine-thréonine kinases TOR/métabolisme
16.
Eur J Pharmacol ; 977: 176583, 2024 Aug 15.
Article de Anglais | MEDLINE | ID: mdl-38679123

RÉSUMÉ

Dry eye disease (DED) is a complex disorder driven by several factors like reduced tear production, increased evaporation, or poor tear quality. Oxidative stress plays a key role by exacerbating the inflammatory cycle. Previous studies explored antioxidants for DED treatment due to the link between oxidative damage and inflammation. Biochanin A (BCA) is a bioisoflavone from red clover with potent anti-inflammatory effects. This study investigated BCA's therapeutic potential for DED. Human corneal epithelial cells were cultured under hyperosmotic conditions to mimic DED. BCA treatment increased cell viability and decreased apoptosis and inflammatory cytokine expression. A DED mouse model was developed using female C57BL/6 mice in a controlled low-humidity environment combined with scopolamine injections. Mice received eye drops containing phosphate-buffered saline, low-dose BCA, or high-dose BCA. The effectiveness was evaluated by measuring tear volume, fluorescein staining, eye-closing ratio, corneal sensitivity and PAS staining. The levels of inflammatory components in corneas and conjunctiva were measured to assess DED severity. Maturation of antigen-presenting cells in cervical lymph nodes was analyzed by flow cytometry. BCA eye drops effectively reduced inflammation associated with DED in mice. BCA also decreased oxidative stress levels by reducing reactive oxygen species and enhancing the nuclear translocation of nuclear factor erythroid-2-related factor 2 (Nrf2). These findings demonstrate that BCA ameliorates oxidative stress and ocular surface inflammation, indicating potential as a DED treatment by relieving oxidative damage and mitigating inflammation.


Sujet(s)
Syndromes de l'oeil sec , Génistéine , Souris de lignée C57BL , Facteur-2 apparenté à NF-E2 , Stress oxydatif , Animaux , Syndromes de l'oeil sec/traitement médicamenteux , Syndromes de l'oeil sec/métabolisme , Syndromes de l'oeil sec/anatomopathologie , Génistéine/pharmacologie , Génistéine/usage thérapeutique , Femelle , Humains , Stress oxydatif/effets des médicaments et des substances chimiques , Souris , Facteur-2 apparenté à NF-E2/métabolisme , Modèles animaux de maladie humaine , Espèces réactives de l'oxygène/métabolisme , Anti-inflammatoires/pharmacologie , Anti-inflammatoires/usage thérapeutique , Cytokines/métabolisme , Apoptose/effets des médicaments et des substances chimiques , Inflammation/traitement médicamenteux , Inflammation/métabolisme , Inflammation/anatomopathologie , Solutions ophtalmiques/pharmacologie , Survie cellulaire/effets des médicaments et des substances chimiques , Antioxydants/pharmacologie , Cellules épithéliales/effets des médicaments et des substances chimiques , Cellules épithéliales/métabolisme , Lignée cellulaire , Cornée/effets des médicaments et des substances chimiques , Cornée/métabolisme , Cornée/anatomopathologie
17.
J Biochem Mol Toxicol ; 38(4): e23697, 2024 Apr.
Article de Anglais | MEDLINE | ID: mdl-38578078

RÉSUMÉ

Genistein, an isoflavone has the potential to mimic, augment, or dysregulate the steroid hormone production pathways. We hypothesized that genistein affects the granulosa cell (GCs) functions through a series of biochemical, molecular, and genomic cascades. The present study was conducted to evaluate the impact of genistein exposure on GCs viability, apoptosis, and steroidogenesis. The present study involved 3/5 days of exposure to genistein on GCs collected from abattoir-derived ovine ovaries at doses of 0, 1, 10, 25, 50, and 100 µM. The harvested GCs were used for growth, cytotoxicity, and gene expression studies related to apoptosis, growth, and steroidogenesis. We observed that genistein had both stimulatory at 10 and 25 µM levels as well as inhibitory effects at 50 and 100 µM levels on the growth and proliferation of GCs. Genistein significantly decreased the levels of 17ß-estradiol at higher exposure (50 and 100 µM), whereas the progesterone level increased significantly as the genistein exposure increased. Additionally, genistein could also alter the mRNA expression of the steroidogenic receptor, enzymes, proteins, and growth-related genes suggesting that genistein could potentially alter the steroidogenic pathways. We conclude that genistein can interfere with cell survival and steroidogenesis by exhibiting a dose-dependent biphasic response on the viability, growth-related parameters, and the synthesis of 17ß-estradiol in the cultured GCs.


Sujet(s)
Génistéine , Isoflavones , Femelle , Ovis , Animaux , Génistéine/pharmacologie , Progestérone/métabolisme , Cellules de la granulosa/métabolisme , Oestradiol/pharmacologie , Oestradiol/métabolisme , Isoflavones/pharmacologie , Ovis aries/métabolisme , Cellules cultivées
19.
Shanghai Kou Qiang Yi Xue ; 33(1): 13-21, 2024 Feb.
Article de Chinois | MEDLINE | ID: mdl-38583019

RÉSUMÉ

PURPOSE: To clarify the effect of genistein(GEN) on osteogenic differentiation and explore the effect of GEN loaded by platelet-rich fibrin (PRF) on the repair process of bone defects in obese mice. METHODS: In in vitro experiments, the effect of GEN(0, 0.1, 1, 10, 50 µmol/L) on the proliferation of mouse embryonic osteoblast precursor cells (MC3T3-E1) was determined by CCK 8. Alkaline phosphatase(ALP) staining and quantitative detection of ALP activity were performed to determine the changes of ALP activity in cells; RNA and protein expression levels of ALP, osteopontin (OPN) and osteocalcin (OCN) were detected by quantitative real-time PCR(qRT-PCR) and Western blot. Alizarin red staining was used to define the effect of GEN on mineralization of MC3T3-E1. To verify the feasibility of the PRF drug loading, the ultrastructure of PRF was subsequently observed under SEM. In in vivo experiments, obese C57 mouse models were established by high-fat diet feeding. On this basis, skull defect models with a diameter of 2.8 mm were established, and the prepared GEN/PRF complexes were placed into the bone defect area. The effects of GEN on skull defect repair in obese mice were evaluated by Micro-CT scanning and hematoxylin-eosin(H-E) staining. Statistical analysis was performed with GraphPad Prism 5.0 software package. RESULTS: CCK 8 results showed that 0.1, 1 µmol/L GEN promoted cell proliferation within 7 days(P<0.05); 10 µmol/L GEN had no significant effect on the process of cell proliferation. From the second day, 50 µmol/L GEN significantly inhibited cell growth and showed cytotoxicity(P<0.05). These two concentrations had similar effects in promoting cellular osteogenic differentiation. SEM results showed that PRF presented a 3-dimensional network structure, providing space for loading drug molecules. In in vivo experiments, the body weight of mice in the high-fat diet (HFD) group was 27.7% greater than that in the normal diet group(P<0.05) and had abnormal glucose tolerance (P<0.05). Micro-CT showed that compared with the normal diet group, the number of bone trabeculae in the femur of obese mice was decreased(P<0.05), the distance between bone trabeculae was widened(P<0.05), and the bone density was decreased (P<0.05). In addition, GEN (0.1, 1.0 µmol/L) loaded by PRF increased bone volume fraction in the skull of obese mice (P<0.05). H-E results showed that GEN/PRF promoted the healing of the bone defects. CONCLUSIONS: GEN promotes osteogenic differentiation of MC3T3-E1, and it can effectively accelerate the healing of cranial bone defects after loading with PRF in obese mice.


Sujet(s)
Ostéogenèse , Fibrine riche en plaquettes , Animaux , Souris , Ostéogenèse/génétique , Génistéine/pharmacologie , Souris obèse , Sincalide/pharmacologie , Différenciation cellulaire/génétique , Ostéoblastes
20.
Poult Sci ; 103(6): 103734, 2024 Jun.
Article de Anglais | MEDLINE | ID: mdl-38636201

RÉSUMÉ

Dietary supplementation with bioactive substances that can regulate lipid metabolism is an effective approach for reducing excessive fat deposition in chickens. Genistein (GEN) has the potential to alleviate fat deposition; however, the underlying mechanism of GEN's fat-reduction action in chickens remains unclear. Therefore, the present study aimed to explore the underlying mechanism of GEN on the reduction of fat deposition from a novel perspective: intercellular transmission of adipokine between adipocytes and hepatocytes. The findings showed that GEN enhanced the secretion of adiponectin (APN) in chicken adipocytes, and the enhancement effect of GEN was completely blocked when the cells were pretreated with inhibitors targeting estrogen receptor ß (ERß) or proliferator-activated receptor γ (PPARγ) signals, respectively. Furthermore, the results demonstrated that both co-treatment with GEN and APN or treatment with the medium supernatant (Med SUP) derived from chicken adipocytes treated with GEN significantly decreased the content of triglyceride and increased the protein levels of ERß, Sirtuin 1 (SIRT1) and phosphor-AMP-activated protein kinase (p-AMPK) in chicken hepatocytes compared to the cells treated with GEN or APN alone. Moreover, the increase in the protein levels of SIRT1 and p-AMPK induced by GEN and APN co-treatment or Med SUP treatment were blocked in chicken hepatocytes pretreated with the inhibitor of ERß signals. Importantly, the up-regulatory effect of GEN and APN co-treatment or Med SUP treatment on the protein level of p-AMPK was also blocked in chicken hepatocytes pretreated with a SIRT1 inhibitor; however, the increase in the protein level of SIRT1 induced by GEN and APN co-treatment or Med SUP treatment was not reversed when the hepatocytes were pretreated with an AMPK inhibitor. In conclusion, the present study demonstrated that GEN enhanced APN secretion by activating the ERß-Erk-PPARγ signaling pathway in chicken adipocytes. Subsequently, adipocyte-derived APN synergized with GEN to activate the ERß-mediated SIRT1-AMPK signaling pathway in chicken hepatocytes, ultimately reducing fat deposition. These findings provide substantial evidence from a novel perspective, supporting the potential use of GEN as a dietary supplement to prevent excessive fat deposition in poultry.


Sujet(s)
Adiponectine , Poulets , Récepteur bêta des oestrogènes , Génistéine , Hépatocytes , Transduction du signal , Sirtuine-1 , Animaux , Génistéine/pharmacologie , Génistéine/administration et posologie , Hépatocytes/effets des médicaments et des substances chimiques , Hépatocytes/métabolisme , Sirtuine-1/métabolisme , Récepteur bêta des oestrogènes/métabolisme , Transduction du signal/effets des médicaments et des substances chimiques , Adiponectine/métabolisme , AMP-Activated Protein Kinases/métabolisme , Protéines aviaires/métabolisme , Adipocytes/effets des médicaments et des substances chimiques , Adipocytes/métabolisme , Tissu adipeux/effets des médicaments et des substances chimiques
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE
...