Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 915
Filtrar
1.
Food Funct ; 15(13): 7093-7107, 2024 Jul 01.
Artigo em Inglês | MEDLINE | ID: mdl-38873879

RESUMO

Heat stress can impair the male reproductive function. L-Theanine and dihydromyricetin have biological activities against heat stress; however, their effects on reproductive function in heat-stressed males are unclear. In this study, male mice were given L-theanine, dihydromyricetin, or a combination of both for 28 days, followed by 2 h of heat stress daily for 7 days. All interventions alleviated heat stress-induced testicular damage, improving the testicular organ index, sperm density, acrosome integrity, sperm deformity rate, and hormone levels. Treatment increased the antioxidant enzyme activity and decreased the markers of oxidative and inflammatory stress in the testes. A combination dose of 200 + 200 mg kg-1 d-1 showed the best protective effect. The potential mechanism involves the regulation of HSP27 and HSP70, which regulate the levels of reproductive hormones through the StAR/Cyp11a1/Hsd3b1/Cyp17a1/Hsd17b3 pathway, alleviate inflammation and oxidative stress through the P38/NF-κB/Nrf2/HO-1 pathway, and regulate the Bcl-2/Fas/Caspase3 apoptotic pathway. Overall, L-theanine and dihydromyricetin may play a protective role against heat stress-induced reproductive dysfunction, suggesting their potential use in heat stress-resistant foods.


Assuntos
Flavonóis , Glutamatos , Estresse Oxidativo , Testículo , Animais , Masculino , Flavonóis/farmacologia , Camundongos , Testículo/efeitos dos fármacos , Testículo/metabolismo , Glutamatos/farmacologia , Estresse Oxidativo/efeitos dos fármacos , Resposta ao Choque Térmico/efeitos dos fármacos , Espermatozoides/efeitos dos fármacos , Reprodução/efeitos dos fármacos , Antioxidantes/farmacologia , Substâncias Protetoras/farmacologia , Proteínas de Choque Térmico HSP70/metabolismo , Apoptose/efeitos dos fármacos
2.
J Inorg Biochem ; 258: 112637, 2024 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-38876026

RESUMO

Wet synthesis approach afforded four new heteroleptic mononuclear neutral diamagnetic oxidovanadium(V) complexes, comprising salicylaldehyde-based 2-furoic acid hydrazones and a flavonol coligand of the general composition [VO(fla)(L-ONO)]. The complexes were comprehensively characterized, including chemical analysis, conductometry, infrared, electronic, and mass spectroscopy, as well as 1D 1H and proton-decoupled 13C(1H) NMR spectroscopy, alongside extensive 2D 1H1H COSY, 1H13C HMQC, and 1H13C HMBC NMR analyses. Additionally, the quantum chemical properties of the complexes were studied using Gaussian at the B3LYP, HF, and M062X levels on the 6-31++g(d,p) basis sets. The interaction of these hydrolytically inert vanadium complexes and the BSA was investigated through spectrofluorimetric titration, synchronous fluorimetry, and FRET analysis in a temperature-dependent manner, providing valuable thermodynamic insights into van der Waals interactions and hydrogen bonding. Molecular docking was conducted to gain further understanding of the specific binding sites of the complexes to BSA. Complex 2, featuring a 5-chloro-substituted salicylaldehyde component of the hydrazone, was extensively examined for its biological activity in vivo. The effects of complex administration on biochemical and hematological parameters were evaluated in both healthy and diabetic Wistar rats, revealing antihyperglycemic activity at millimolar concentration. Furthermore, histopathological analysis and bioaccumulation studies of the complex in the brain, kidneys, and livers of healthy and diabetic rats revealed the potential for further development of vanadium(V) hydrazone complexes as antidiabetic and insulin-mimetic agents.


Assuntos
Complexos de Coordenação , Diabetes Mellitus Experimental , Hidrazonas , Hipoglicemiantes , Simulação de Acoplamento Molecular , Vanádio , Animais , Hidrazonas/química , Hidrazonas/síntese química , Hidrazonas/farmacologia , Ratos , Diabetes Mellitus Experimental/tratamento farmacológico , Hipoglicemiantes/química , Hipoglicemiantes/síntese química , Hipoglicemiantes/farmacologia , Complexos de Coordenação/síntese química , Complexos de Coordenação/farmacologia , Complexos de Coordenação/química , Vanádio/química , Flavonóis/farmacologia , Flavonóis/química , Flavonóis/síntese química , Masculino , Ratos Wistar , Soroalbumina Bovina/química , Soroalbumina Bovina/metabolismo , Aldeídos
3.
Int J Mol Sci ; 25(11)2024 May 21.
Artigo em Inglês | MEDLINE | ID: mdl-38891769

RESUMO

Staphylea, also called bladdernuts, is a genus of plants belonging to the family Staphyleaceae, widespread in tropical or temperate climates of America, Europe, and the Far East. Staphylea spp. produce bioactive metabolites with antioxidant properties, including polyphenols which have not been completely investigated for their phytotherapeutic potential, even though they have a long history of use for food. Here, we report the isolation of six flavonol glycosides from the hydroalcoholic extract of aerial parts of Staphylea pinnata L., collected in Italy, using a solid-phase extraction technique. They were identified using spectroscopic, spectrometric, and optical methods as three quercetin and three isorhamnetin glycosides. Among the flavonol glycosides isolated, isoquercetin and quercetin malonyl glucoside showed powerful antioxidant, antimicrobial, and wound healing promoting activity and thus are valuable as antiaging ingredients for cosmeceutical applications and for therapeutic applications in skin wound repair.


Assuntos
Antioxidantes , Flavonóis , Glicosídeos , Extratos Vegetais , Glicosídeos/farmacologia , Glicosídeos/química , Glicosídeos/isolamento & purificação , Flavonóis/farmacologia , Flavonóis/química , Flavonóis/isolamento & purificação , Antioxidantes/farmacologia , Antioxidantes/química , Antioxidantes/isolamento & purificação , Extratos Vegetais/química , Extratos Vegetais/farmacologia , Cicatrização/efeitos dos fármacos , Quercetina/farmacologia , Quercetina/química , Quercetina/análogos & derivados , Quercetina/isolamento & purificação , Humanos , Anti-Infecciosos/farmacologia , Anti-Infecciosos/química , Anti-Infecciosos/isolamento & purificação , Animais
4.
Nanomedicine ; 59: 102752, 2024 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-38740358

RESUMO

Fisetin has displayed potential as an anticonvulsant in preclinical studies yet lacks clinical validation. Challenges like low solubility and rapid metabolism may limit its efficacy. This study explores fisetin-loaded chitosan nanoparticles (NP) to address these issues. Using a murine model of pilocarpine-induced temporal lobe epilepsy, we evaluated the anticonvulsant and neuroprotective effects of fisetin NP. Pilocarpine-induced seizures and associated neurobehavioral deficits were assessed after administering subtherapeutic doses of free fisetin and fisetin NP. Changes in ROS, inflammatory cytokines, and NLRP3/IL-18 expression in different brain regions were estimated. The results demonstrate that the fisetin NP exerts protection against seizures and associated depression-like behavior and memory impairment. Furthermore, biochemical, and histological examinations supported behavioral findings suggesting attenuation of ROS/TNF-α-NLRP3 inflammasome pathway as a neuroprotective mechanism of fisetin NP. These findings highlight the improved pharmacodynamics of fisetin using fisetin NP against epilepsy, suggesting a promising therapeutic approach against epilepsy and associated behavioral deficits.


Assuntos
Quitosana , Epilepsia do Lobo Temporal , Flavonóis , Inflamassomos , Proteína 3 que Contém Domínio de Pirina da Família NLR , Nanopartículas , Pilocarpina , Espécies Reativas de Oxigênio , Fator de Necrose Tumoral alfa , Animais , Epilepsia do Lobo Temporal/tratamento farmacológico , Epilepsia do Lobo Temporal/induzido quimicamente , Epilepsia do Lobo Temporal/patologia , Epilepsia do Lobo Temporal/metabolismo , Quitosana/química , Quitosana/farmacologia , Flavonóis/farmacologia , Camundongos , Proteína 3 que Contém Domínio de Pirina da Família NLR/metabolismo , Inflamassomos/metabolismo , Inflamassomos/efeitos dos fármacos , Nanopartículas/química , Masculino , Espécies Reativas de Oxigênio/metabolismo , Fator de Necrose Tumoral alfa/metabolismo , Flavonoides/farmacologia , Flavonoides/administração & dosagem , Comportamento Animal/efeitos dos fármacos , Anticonvulsivantes/farmacologia , Fármacos Neuroprotetores/farmacologia
5.
Molecules ; 29(9)2024 Apr 28.
Artigo em Inglês | MEDLINE | ID: mdl-38731532

RESUMO

A series of flavanols were synthesized to assess their biological activity against human non-small cell lung cancer cells (A549). Among the sixteen synthesized compounds, it was observed that compounds 6k (3.14 ± 0.29 µM) and 6l (0.46 ± 0.02 µM) exhibited higher potency compared to 5-fluorouracil (5-Fu, 4.98 ± 0.41 µM), a clinical anticancer drug which was used as a positive control. Moreover, compound 6l (4'-bromoflavonol) markedly induced apoptosis of A549 cells through the mitochondrial- and caspase-3-dependent pathways. Consequently, compound 6l might be developed as a candidate for treating or preventing lung cancer.


Assuntos
Antineoplásicos , Apoptose , Flavonóis , Humanos , Flavonóis/farmacologia , Flavonóis/síntese química , Flavonóis/química , Antineoplásicos/farmacologia , Antineoplásicos/síntese química , Antineoplásicos/química , Apoptose/efeitos dos fármacos , Células A549 , Caspase 3/metabolismo , Proliferação de Células/efeitos dos fármacos , Relação Estrutura-Atividade , Estrutura Molecular , Fluoruracila/farmacologia , Mitocôndrias/efeitos dos fármacos , Mitocôndrias/metabolismo , Ensaios de Seleção de Medicamentos Antitumorais , Neoplasias Pulmonares/tratamento farmacológico , Neoplasias Pulmonares/patologia , Neoplasias Pulmonares/metabolismo , Linhagem Celular Tumoral
6.
Biomolecules ; 14(5)2024 May 14.
Artigo em Inglês | MEDLINE | ID: mdl-38785985

RESUMO

Aronia melanocarpa berries contain many compounds with potential benefits for human health. The food flavonoids quercetin and rutin, found in significant amounts in the fruits of A. melanocarpa, are known to have favourable effects on animal and human organisms. However, data on the effect of flavonols isolated from black chokeberry on immune functions during immunosuppression are not available in the literature. Thus, the aim of this study was to evaluate the effect of flavonol fraction isolated from A. melanocarpa fruits, in comparison with pure quercetin and rutin substances, on the dysfunctional state of rat thymus and spleen in immunodeficiency. The study was performed on Wistar rats. The animals were orally administered solutions of the investigated substances for 7 days: water, a mixture of quercetin and rutin and flavonol fraction of A. melanocarpa. For induction of immunosuppression, the animals were injected once intraperitoneally with cyclophosphamide. Substance administration was then continued for another 7 days. The results showed that under the influence of flavonols, there was a decrease in cyclophosphamide-mediated reaction of lipid peroxidation enhancement and stimulation of proliferation of lymphocytes of thymus and spleen in rats. At that, the effect of the flavonol fraction of aronia was more pronounced.


Assuntos
Ciclofosfamida , Flavonóis , Frutas , Photinia , Ratos Wistar , Baço , Timo , Animais , Photinia/química , Ciclofosfamida/farmacologia , Ratos , Frutas/química , Timo/efeitos dos fármacos , Flavonóis/farmacologia , Flavonóis/química , Baço/efeitos dos fármacos , Masculino , Extratos Vegetais/farmacologia , Extratos Vegetais/química , Terapia de Imunossupressão , Quercetina/farmacologia , Quercetina/química , Peroxidação de Lipídeos/efeitos dos fármacos , Imunossupressores/farmacologia , Proliferação de Células/efeitos dos fármacos , Rutina/farmacologia , Rutina/química
7.
Eur J Pharmacol ; 976: 176670, 2024 Aug 05.
Artigo em Inglês | MEDLINE | ID: mdl-38795755

RESUMO

INTRODUCTION: Ischemia/reperfusion is a pathological condition by the restoration of perfusion and oxygenation following a period of restricted blood flow to an organ. To address existing uncertainty in the literature regarding the effects of 3', 4'-dihydroxy flavonol (DiOHF) on cerebral ischemia/reperfusion injury, our study aims to investigate the impact of DiOHF on neurological parameters, apoptosis (Caspase-3), aquaporin 4 (AQP4), and interleukin-10 (IL-10) levels in an experimental rat model of brain ischemia-reperfusion injury. MATERIALS/METHODS: A total of 28 Wistar-albino male rats were used in this study. Experimental groups were formed as 1-Control, 2-Sham, 3-Ischemia-reperfusion, 4-Ischemia-reperfusion + DiOHF (10 mg/kg). The animals were anaesthetized, and the carotid arteries were ligated (ischemia) for 30 min, followed by reperfusion for 30 min. Following reperfusion, DiOHF was administered intraperitoneally to the animals at a dose of 10 mg/kg for 1 week. During the one-week period neurological scores and new object recognition tests were performed. Then, caspase 3 and AQP4 levels were determined by PCR method and IL-10 by ELISA method in hippocampus tissue samples taken from animals sacrificed under anaesthesia. RESULTS: Brain ischemia reperfusion significantly increased both caspase 3 and AQP4 values in the hippocampus tissue, while decreasing IL-10 levels. However, 1-week DiOHF supplementation significantly suppressed increased caspase 3 and AQP4 levels and increased IL-10 values. While I/R also increased neurological score values, it suppressed the ability to recognize new objects, and the administered treatment effectively ameliorated the adverse effects observed, resulting in a positive outcome. CONCLUSIONS: The results of the study show that brain ischemia caused by bilateral carotid occlusion in rats and subsequent reperfusion causes tissue damage, but 1-week DiOHF application has a healing effect on both hippocampus tissue and neurological parameters.


Assuntos
Aquaporina 4 , Caspase 3 , Cognição , Flavonóis , Interleucina-10 , Ratos Wistar , Traumatismo por Reperfusão , Animais , Masculino , Traumatismo por Reperfusão/tratamento farmacológico , Traumatismo por Reperfusão/patologia , Traumatismo por Reperfusão/metabolismo , Flavonóis/farmacologia , Flavonóis/uso terapêutico , Ratos , Cognição/efeitos dos fármacos , Caspase 3/metabolismo , Aquaporina 4/metabolismo , Interleucina-10/metabolismo , Fármacos Neuroprotetores/farmacologia , Fármacos Neuroprotetores/uso terapêutico , Hipocampo/efeitos dos fármacos , Hipocampo/metabolismo , Hipocampo/patologia , Neurônios/efeitos dos fármacos , Neurônios/patologia , Neurônios/metabolismo , Isquemia Encefálica/tratamento farmacológico , Isquemia Encefálica/metabolismo , Modelos Animais de Doenças , Apoptose/efeitos dos fármacos
8.
J Food Sci ; 89(6): 3569-3576, 2024 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-38745380

RESUMO

This study aims to investigate the impact of four key factors, namely, temperature, water source, metal ion, and pH, on the stability of molecular chirality of dihydromyricetin (DMY) and proposed effective strategies for configuration protection. The findings reveal that temperatures exceeding 80°C could accelerate the racemization process of DMY, with a significant increase in racemization observed at 100°C. In addition, DMY exhibited heightened stability in ultrapure water as compared to various water sources, including pure water-1, pure water-2, mineral water, and running water. Notably, the presence of Fe2+ displayed an inhibitory effect on the racemization of DMY, whereas Mg2+, Ca2+, and Mn2+ showed a substantial promotional effect. Additionally, acidic conditions (pH < 5.0) were found to be protective for maintaining the stability of DMY, whereas alkaline conditions (pH > 9.0) were observed to be detrimental. Meanwhile, we first identified the presence of another pair of DMY isomers in this work.


Assuntos
Flavonóis , Flavonóis/farmacologia , Flavonóis/química , Concentração de Íons de Hidrogênio , Estereoisomerismo , Água/química , Temperatura , Isomerismo , Chá/química
9.
Phytomedicine ; 130: 155768, 2024 Jul 25.
Artigo em Inglês | MEDLINE | ID: mdl-38815408

RESUMO

BACKGROUND: Polymyxin E is widely recognized as a last resort for treating multidrug-resistant gram-negative bacteria. Unfortunately, the effectiveness of polymyxin E is significantly reduced when treating life-threatening bacterial infections due to plasmid-mediated polymyxin E resistance. The synergistic effect of applying a polymyxin E adjuvant is a promising strategy for overcoming the growing threat of antibiotic-resistant pathogens. PURPOSE: To evaluate the synergistic effect of fisetin and polymyxin E on S. typhimurium infections in vivo and further elucidate the underlying mechanism of this effect. METHODS: The effect of combining fisetin and polymyxin E to treat mobilized colistin resistance-1-positive (MCR-1-positive) gram-negative bacteria in vitro was examined using various methods, such as checkerboard assays, growth curves and time‒kill curves. To elucidate the mechanism by which fisetin affects MCR-1, we employed ultraviolet (UV) absorption spectroscopy, thin layer chromatography (TLC), and western blot analysis to investigate its effect at the protein level. Subsequently, molecular dynamics simulations (MDS) and metabolomics analysis were utilized to determine the site of interaction between fisetin and MCR-1 as well as the potential pathways and mechanisms involved. A new nanoemulsion of fisetin was produced using high-pressure homogenization, and its stability was tested. Finally, two animal models of S. typhimurium HYM2 infection were established to evaluate the synergistic effect of polymyxin E and fisetin in vivo. RESULTS: Our study revealed that fisetin exhibited a synergistic effect when combined with polymyxin E against MCR-1-positive S. typhimurium. The TLC results demonstrated that fisetin could inhibit the phosphoethanolamine (PEA) transfer of the MCR-1 protein, thereby restoring the activity of polymyxin E in strains against MCR-1. The MDS analysis indicated robust and immediate binding between fisetin and the MCR-1 protein, with both hydrophobic and polar effects playing significant roles in determining the binding energy of the former. Metabolomic studies demonstrated that the addition of fisetin significantly modulated bacterial metabolites. Moreover, it effectively inhibited the activity of ABC transporters in bacteria, thereby mitigating bacterial drug resistance and enhancing the therapeutic efficacy of polymyxin E. Furthermore, in mouse and chick models of infection, intragastric administration of the fisetin nanoemulsion together with polymyxin E resulted in significant therapeutic benefits, including increased survival rates, reduced bacterial colonization, and decreased levels of inflammatory factors. CONCLUSION: Fisetin, an MCR-1 inhibitor and a promising synergistic partner of polymyxin E, has significant potential for clinical application in the treatment of S. typhimurium infections, particularly those resulting extensively from drug-resistant MCR-1-positive strains.


Assuntos
Antibacterianos , Colistina , Flavonoides , Flavonóis , Salmonella typhimurium , Flavonóis/farmacologia , Animais , Colistina/farmacologia , Antibacterianos/farmacologia , Salmonella typhimurium/efeitos dos fármacos , Flavonoides/farmacologia , Emulsões , Sinergismo Farmacológico , Camundongos , Testes de Sensibilidade Microbiana , Feminino , Farmacorresistência Bacteriana , Farmacorresistência Bacteriana Múltipla/efeitos dos fármacos , Simulação de Dinâmica Molecular , Camundongos Endogâmicos BALB C
10.
Phytomedicine ; 130: 155754, 2024 Jul 25.
Artigo em Inglês | MEDLINE | ID: mdl-38820662

RESUMO

BACKGROUND: Gouty arthritis (GA), a common inflammatory condition triggered by monosodium urate crystal accumulation, often necessitates safer treatment alternatives due to the limitations of current therapies. Astilbin, a flavonoid from Smilax glabra Roxb, has demonstrated potential in traditional Chinese medicine for its anti-inflammatory properties. However, the anti-GA effect and its underlying mechanism have not been fully elucidated. PURPOSE: This study aimed to investigate the therapeutic potential of astilbin in GA, focusing on its effects on neutrophil extracellular traps (NETs), as well as the potential molecular target of GA both in vitro and in vivo. STUDY DESIGN: Firstly, astilbin inhibited the citrullinated histone H3 (Cit h3) protein levels and reduced the NETs formation in neutrophils stimulated by monosodium urate (MSU). Secondly, we wondered the effect of astilbin on migration of neutrophils and dimethyl-sulfoxide (DMSO)-differentiated HL-60 (dHL-60) cells under the stimulation of MSU. Then, the effect of astilbin on suppressing NETs through purinergic P2Y6 receptor (P2Y6R) and Interlukin-8 (IL-8)/ CXC chemokine receptor 2 (CXCR2) pathway was investigated. Also, the relationship between P2Y6R and IL-8/CXCR2 was explored in dHL-60 cells under stimulation of MSU. Finally, we testified the effect of astilbin on reducing NETs in GA through suppressing P2Y6R and then down-regulating IL-8/CXCR2 pathway. METHODS: MSU was used to induce NETs in neutrophils and dHL-60 cells. Real-time formation of NETs and migration of neutrophils were monitored by cell living imaging with or without MSU. Then, the effect of astilbin on NETs formation, P2Y6R and IL-8/CXCR2 pathway were detected by immunofluorescence (IF) and western blotting. P2Y6R knockdown dHL-60 cells were established by small interfering RNA to investigate the association between P2Y6R and IL-8/CXCR2 pathway. Also, plasmid of P2Y6R was used to overexpress P2Y6R in dHL-60 cells, which was employed to explore the role of P2Y6R in astilbin inhibiting NETs. Within the conditions of knockdown and overexpression of P2Y6R, migration and NETs formation were assessed by transmigration assay and IF staining, respectively. In vivo, MSU-induced GA mice model was established to assess the effect of astilbin on inflammation by haematoxylin-eosin and ELISA. Additionally, the effects of astilbin on neutrophils infiltration, NETs, P2Y6R and IL-8/CXCR2 pathway were analyzed by IF, ELISA, immunohistochemistry (IHC) and western blotting. RESULTS: Under MSU stimulation, astilbin significantly suppressed the level of Cit h3 and NETs formation including the fluorescent expressions of Cit h3, neutrophils elastase, myeloperoxidase, and intra/extracellular DNA. Also, results showed that MSU caused NETs release in neutrophils as well as a trend towards recruitment of dHL-60 cells to MSU. Astilbin could markedly decrease expressions of P2Y6R and IL-8/CXCR2 pathway which were upregulated by MSU. By silencing P2Y6R, the expression of IL-8/CXCR2 pathway and migration of dHL-60 cells were inhibited, leading to the suppression of NETs. These findings indicated the upstream role of P2Y6R in the IL-8/CXCR2 pathway. Moreover, overexpression of P2Y6R was evidently inhibited by astilbin, causing a downregulation in IL-8/CXCR2 pathway, migration of dHL-60 cells and NETs formation. These results emphasized that astilbin inhibited the IL-8/CXCR2 pathway primarily through P2Y6R. In vivo, astilbin administration led to marked reductions in ankle swelling, inflammatory infiltration as well as neutrophils infiltration. Expressions of P2Y6R and IL-8/CXCR2 pathway were evidently decreased by astilbin and P2Y6R inhibitor MRS2578 either alone or in combination. Also, astilbin and MRS2578 showed notable effect on reducing MSU-induced NETs formation and IL-8/CXCR2 pathway whether used alone or in combination, parallelly demonstrating that astilbin decreased NETs formation mainly through P2Y6R. CONCLUSION: This study revealed that astilbin suppressed NETs formation via downregulating P2Y6R and subsequently the IL-8/CXCR2 pathway, which evidently mitigated GA induced by MSU. It also highlighted the potential of astilbin as a promising natural therapeutic for GA.


Assuntos
Artrite Gotosa , Armadilhas Extracelulares , Flavonóis , Interleucina-8 , Neutrófilos , Receptores Purinérgicos P2 , Armadilhas Extracelulares/efeitos dos fármacos , Humanos , Interleucina-8/metabolismo , Receptores Purinérgicos P2/metabolismo , Neutrófilos/efeitos dos fármacos , Neutrófilos/metabolismo , Artrite Gotosa/tratamento farmacológico , Células HL-60 , Flavonóis/farmacologia , Animais , Ácido Úrico/farmacologia , Receptores de Interleucina-8B/metabolismo , Masculino , Histonas/metabolismo , Anti-Inflamatórios/farmacologia , Camundongos
11.
Int J Mol Sci ; 25(10)2024 May 11.
Artigo em Inglês | MEDLINE | ID: mdl-38791295

RESUMO

To achieve the environmentally friendly and rapid green synthesis of efficient and stable AgNPs for drug-resistant bacterial infection, this study optimized the green synthesis process of silver nanoparticles (AgNPs) using Dihydromyricetin (DMY). Then, we assessed the impact of AgNPs on zebrafish embryo development, as well as their therapeutic efficacy on zebrafish infected with Methicillin-resistant Staphylococcus aureus (MRSA). Transmission electron microscopy (TEM) and dynamic light-scattering (DLS) analyses revealed that AgNPs possessed an average size of 23.6 nm, a polymer dispersity index (PDI) of 0.197 ± 0.0196, and a zeta potential of -18.1 ± 1.18 mV. Compared to other published green synthesis products, the optimized DMY-AgNPs exhibited smaller sizes, narrower size distributions, and enhanced stability. Furthermore, the minimum concentration of DMY-AgNPs required to affect zebrafish hatching and survival was determined to be 25.0 µg/mL, indicating the low toxicity of DMY-AgNPs. Following a 5-day feeding regimen with DMY-AgNP-containing food, significant improvements were observed in the recovery of the gills, intestines, and livers in MRSA-infected zebrafish. These results suggested that optimized DMY-AgNPs hold promise for application in aquacultures and offer potential for further clinical use against drug-resistant bacteria.


Assuntos
Antibacterianos , Flavonóis , Química Verde , Nanopartículas Metálicas , Staphylococcus aureus Resistente à Meticilina , Prata , Peixe-Zebra , Animais , Staphylococcus aureus Resistente à Meticilina/efeitos dos fármacos , Nanopartículas Metálicas/química , Prata/química , Prata/farmacologia , Flavonóis/farmacologia , Flavonóis/química , Química Verde/métodos , Antibacterianos/farmacologia , Antibacterianos/química , Antibacterianos/síntese química , Infecções Estafilocócicas/tratamento farmacológico , Testes de Sensibilidade Microbiana
12.
Eur Rev Med Pharmacol Sci ; 28(8): 3112-3119, 2024 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-38708470

RESUMO

OBJECTIVE: Methotrexate (MTX), a widely used chemotherapeutic and immunosuppressive agent, is associated with hepatotoxicity, leading to liver fibrosis and cirrhosis. This study explores the regenerative and reparative effects of fisetin, a flavonoid with known antioxidant and anti-inflammatory properties, on MTX-induced liver fibrosis in a rat model. MATERIALS AND METHODS: Thirty-six male Wistar albino rats were divided into normal, MTX and saline, and MTX and fisetin. Liver injury was induced in the latter two groups using a single intraperitoneal dose of MTX (20 mg/kg). Fisetin (50 mg/kg/day) or saline was administered intraperitoneally for ten days. After sacrifice, liver tissues were subjected to histopathological evaluation and biochemical analyses, including Transforming Growth Factor-ß1 (TGF-beta), sirtuins-1 (SIRT-1), malondialdehyde (MDA), cytokeratin 18, thrombospondin 1, and alanine transaminase (ALT) levels. RESULTS: MTX administration significantly increased liver injury markers, including TGF-beta, MDA, cytokeratin 18, thrombospondin 1, and ALT, while reducing SIRT-1 levels. Fisetin treatment attenuated these effects, demonstrating its potential therapeutic impact. Histopathological analysis confirmed that fisetin mitigated MTX-induced hepatocyte necrosis, fibrosis, and cellular infiltration. CONCLUSIONS: This study proves that fisetin administration can alleviate MTX-induced liver damage in rats. The reduction in oxidative stress, inflammation, and apoptosis, along with the histological improvements, suggests fisetin's potential as a therapeutic agent against MTX-induced hepatotoxicity. Further investigations and clinical studies are warranted to validate these findings and assess fisetin's translational potential in human cases of MTX-induced liver damage.


Assuntos
Flavonóis , Cirrose Hepática , Metotrexato , Ratos Wistar , Sirtuína 1 , Metotrexato/efeitos adversos , Animais , Masculino , Ratos , Cirrose Hepática/induzido quimicamente , Cirrose Hepática/tratamento farmacológico , Cirrose Hepática/patologia , Cirrose Hepática/metabolismo , Flavonóis/farmacologia , Flavonoides/farmacologia , Fígado/efeitos dos fármacos , Fígado/patologia , Fígado/metabolismo , Antioxidantes/farmacologia
13.
Pharmacology ; 109(3): 169-179, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-38583431

RESUMO

INTRODUCTION: Fisetin has been demonstrated to inhibit the occurrence of atherosclerosis; however, the mechanism of fisetin suppressing atherosclerosis remains elusive. METHODS: The function of fisetin in the inhibition of atherosclerosis was evaluated by hematoxylin and eosin and Oil Red O staining in ApoE-/- mice. Molecular biomarkers of atherosclerosis progression were detected by Western blot and qPCR. Moreover, the inhibition of atherosclerosis on oxidative stress and ferroptosis was evaluated by immunofluorescence staining, qPCR, and Western blot assays. RESULTS: The obtained results showed that serum lipid was attenuated and consequentially the formation of atherosclerosis was also suppressed by fisetin in ApoE-/- mice. Exploration of the mechanism revealed that molecular biomarkers of atherosclerosis were decreased under fisetin treatment. The level of reactive oxygen species and malondialdehyde declined, while the activity of superoxide dismutases and glutathione peroxidase was increased under the fisetin treatment. Additionally, the suppressor of ferroptosis, glutathione peroxidase 4 proteins, was elevated. The ferritin was decreased in the aortic tissues treated with fisetin. CONCLUSIONS: In summary, fisetin attenuated the formation of atherosclerosis through the inhibition of oxidative stress and ferroptosis in the aortic tissues of ApoE-/- mice.


Assuntos
Apolipoproteínas E , Aterosclerose , Ferroptose , Flavonóis , Estresse Oxidativo , Animais , Flavonóis/farmacologia , Aterosclerose/metabolismo , Aterosclerose/prevenção & controle , Aterosclerose/tratamento farmacológico , Estresse Oxidativo/efeitos dos fármacos , Ferroptose/efeitos dos fármacos , Camundongos , Masculino , Apolipoproteínas E/genética , Camundongos Knockout , Espécies Reativas de Oxigênio/metabolismo , Camundongos Endogâmicos C57BL , Flavonoides/farmacologia , Camundongos Knockout para ApoE , Aorta/efeitos dos fármacos , Aorta/metabolismo , Aorta/patologia , Modelos Animais de Doenças , Glutationa Peroxidase/metabolismo
14.
Nutrients ; 16(8)2024 Apr 18.
Artigo em Inglês | MEDLINE | ID: mdl-38674891

RESUMO

The complex and multi-stage processes of carcinogenesis are accompanied by a number of phenomena related to the potential involvement of various chemopreventive factors, which include, among others, compounds of natural origin such as flavonols. The use of flavonols is not only promising but also a recognized strategy for cancer treatment. The chemopreventive impact of flavonols on cancer arises from their ability to act as antioxidants, impede proliferation, promote cell death, inhibit angiogenesis, and regulate the immune system through involvement in diverse forms of cellular death. So far, the molecular mechanisms underlying the regulation of apoptosis, autophagy, necroptosis, pyroptosis, ferroptosis, and cuproptosis occurring with the participation of flavonols have remained incompletely elucidated, and the results of the studies carried out so far are ambiguous. For this reason, one of the therapeutic goals is to initiate the death of altered cells through the use of quercetin, kaempferol, myricetin, isorhamnetin, galangin, fisetin, and morin. This article offers an extensive overview of recent research on these compounds, focusing particularly on their role in combating cancer and elucidating the molecular mechanisms governing apoptosis, autophagy, necroptosis, pyroptosis, ferroptosis, and cuproptosis. Assessment of the mechanisms underlying the anticancer effects of compounds in therapy targeting various types of cell death pathways may prove useful in developing new therapeutic regimens and counteracting resistance to previously used treatments.


Assuntos
Apoptose , Autofagia , Ferroptose , Flavonóis , Necroptose , Neoplasias , Piroptose , Humanos , Flavonóis/farmacologia , Neoplasias/tratamento farmacológico , Neoplasias/patologia , Ferroptose/efeitos dos fármacos , Autofagia/efeitos dos fármacos , Piroptose/efeitos dos fármacos , Apoptose/efeitos dos fármacos , Necroptose/efeitos dos fármacos , Animais , Morte Celular/efeitos dos fármacos
15.
Ecotoxicol Environ Saf ; 277: 116364, 2024 Jun 01.
Artigo em Inglês | MEDLINE | ID: mdl-38657461

RESUMO

The purpose of this study was to investigate the effect of Treg/Th1 imbalance in cadmium-induced lung injury and the potential protective effect of astilbin against cadmium-induced lung injury in chicken. Cadmium exposure significantly decreased T-AOC and GSH-Px levels and SOD activity in the chicken lung tissues. In contrast, it significantly increased the MDA and NO levels. These results indicate that cadmium triggers oxidative stress in lungs. Histopathological analysis revealed that cadmium exposure further induced infiltration of lymphocytes in the chicken lungs, indicating that cadmium causes pulmonary damage. Further analysis revealed that cadmium decreased the expression of IL-4 and IL-10 but increased those of IL-17, Foxp3, TNF-α, and TGF-ß, indicating that the exposure of cadmium induced the imbalance of Treg/Th1. Moreover, cadmium adversely affected chicken lung function by activating the NF-kB pathway and inducing expression of genes downstream to these pathways (COX-2, iNOS), associated with inflammatory injury in the lung tissue. Astilbin reduced cadmium-induced oxidative stress and inflammation in the lungs by increasing antioxidant enzyme activities and restoring Treg/Th1 balance. In conclusion, our results suggest that astilbin treatment alleviated the effects of cadmium-mediated lung injury in chickens by restoring the Treg/Th1 balance.


Assuntos
Cádmio , Galinhas , Flavonóis , Lesão Pulmonar , Pulmão , Estresse Oxidativo , Transdução de Sinais , Linfócitos T Reguladores , Animais , Cádmio/toxicidade , Estresse Oxidativo/efeitos dos fármacos , Pulmão/efeitos dos fármacos , Pulmão/patologia , Transdução de Sinais/efeitos dos fármacos , Linfócitos T Reguladores/efeitos dos fármacos , Flavonóis/farmacologia , Lesão Pulmonar/induzido quimicamente , Lesão Pulmonar/tratamento farmacológico
16.
Int Immunopharmacol ; 130: 111742, 2024 Mar 30.
Artigo em Inglês | MEDLINE | ID: mdl-38452414

RESUMO

BACKGROUND: Cerebral ischemia/reperfusion injury (IRI) is pathologically associated with protein damage. The flavonoid fisetin has good therapeutic effects on cerebral IRI. However, the role of fisetin in regulating protein damage during cerebral IRI development remains unclear. This study investigated the pharmacological effects of fisetin on protein damage during cerebral IRI progression and defined the underlying mechanism of action. METHODS: In vivo and in vitro models of cerebral IRI were established by middle cerebral artery occlusion/reperfusion (MACO/R) and oxygen-glucose deprivation/reperfusion (OGD/R) treatment, respectively. Triphenyl tetrazolium chloride staining was performed to detect cerebral infarct size, and the modified neurologic severity score was used to examine neurological deficits. LDH activity and protein damage were assessed using kits. HT22 cell vitality and apoptosis were examined using CCK-8 assay and TUNEL staining, respectively. Interactions between Foxc1, Ubqln1, Sirt1, and Ezh2 were analyzed using CoIP, ChIP and/or dual-luciferase reporter gene assays. RESULTS: Fisetin alleviated protein damage and ubiquitinated protein aggregation and neuronal death caused by MCAO/R and OGD/R. Ubqln1 knockdown abrogated the inhibitory effect of fisetin on OGD/R-induced protein damage, ubiquitinated protein aggregation, and neuronal death in HT22 cells. Further experiments demonstrated that Foxc1 functions as a transcriptional activator of Ubqln1 and that Sirt1 promotes Foxc1 expression by deacetylating Ezh2 and inhibiting its activity. Furthermore, Sirt1 knockdown abrogated fisetin-mediated biological effects on OGD/R-treated HT22 cells. CONCLUSION: Fisetin improved proteostasis during cerebral IRI by regulating the Sirt1/Foxc1/Ubqln1 signaling axis. Our findings strongly suggest that fisetin-mediated inhibition of protein damage after ischemic stroke is a part of the mechanism through which fisetin is neuroprotective in cerebral IRI.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal , Proteínas Relacionadas à Autofagia , Isquemia Encefálica , Flavonóis , Fatores de Transcrição Forkhead , Proteostase , Traumatismo por Reperfusão , Sirtuína 1 , Apoptose , Isquemia Encefálica/tratamento farmacológico , Flavonóis/farmacologia , Flavonóis/uso terapêutico , Infarto da Artéria Cerebral Média/tratamento farmacológico , Agregados Proteicos , Proteostase/efeitos dos fármacos , Traumatismo por Reperfusão/tratamento farmacológico , Sirtuína 1/metabolismo , Masculino , Animais , Camundongos , Camundongos Endogâmicos C57BL , Fatores de Transcrição Forkhead/metabolismo , Proteínas Relacionadas à Autofagia/metabolismo , Proteínas Adaptadoras de Transdução de Sinal/metabolismo
17.
Int J Pharm ; 654: 123971, 2024 Apr 10.
Artigo em Inglês | MEDLINE | ID: mdl-38452832

RESUMO

Lymphoma and leukemia are both hematological system tumors with complex etiology, and mainly treated with chemotherapeutic drugs. However, therapeutic drugs can interrupt curative effect due to different side effects. Therefore, it is worthwhile to develop a novel therapeutic for providing insights for clinical tumor treatment. In this study, we developed a fisetin nanoparticles (Fisetin NPs) through a self-assembled method, and investigated the activity and potential mechanism of Fisetin NPs against lymphoma and leukemia. The spherical and uniformly distributed Fisetin NPs effectively inhibited both tumor cells proliferation, arrested EL4 cells G0/G1 phase and K562 cells G2/M phase, and induced apoptosis in vitro. In vivo, Fisetin NPs exhibited excellent tumor growth inhibition, effective inhibition of cell proliferation and angiogenesis, significant induction of apoptosis and ideal safety. Mechanically, fisetin upregulated genes (Fas, Pidd, Puma, Apaf1, and p21) in the p53 signaling pathway and bound to N-acetyltransferase 10 (NAT10), ribosomal protein L34 (RPL34) and GTP binding protein 4 (GTPBP4). Collectively, Fisetin NPs have promising therapeutic effects on lymphoma and leukemia, which are of great significant for clinical implications.


Assuntos
Leucemia , Linfoma , Humanos , Flavonoides/farmacologia , Flavonóis/farmacologia , Apoptose , Proliferação de Células , Leucemia/tratamento farmacológico , Linfoma/tratamento farmacológico , Linhagem Celular Tumoral , Proteínas Nucleares/farmacologia , Proteínas de Ligação ao GTP/farmacologia , Acetiltransferases N-Terminal
18.
ChemMedChem ; 19(11): e202300682, 2024 Jun 03.
Artigo em Inglês | MEDLINE | ID: mdl-38369675

RESUMO

Carbon monoxide (CO) delivery molecules are of significant current interest as potential therapeutics, including for anticancer applications. A recent approach toward generating new types of materials-based anticancer agents involves combining the Fenton reactivity of a redox active metal ion with CO delivery. However, small molecule examples of these types of entities have not been systematically studied to evaluate the combined effect on cellular toxicity. Herein we describe a Cu(II) flavonolato complex which produces anticancer effects through a combination of copper-mediated reactive oxygen species (ROS) generation and light-induced flavonol CO release. Confocal microscopy studies provide evidence of enhanced flavonol uptake in the copper flavonolato system relative to the free flavonol, which leads to an increased amount of CO delivery within cells. Importantly, this work demonstrates that a metal flavonolato species can be used to produce enhanced toxicity effects resulting from both metal ion-induced Fenton reactivity and increased cellular uptake of a flavonol CO donor.


Assuntos
Monóxido de Carbono , Complexos de Coordenação , Cobre , Flavonóis , Espécies Reativas de Oxigênio , Monóxido de Carbono/química , Cobre/química , Cobre/farmacologia , Humanos , Complexos de Coordenação/química , Complexos de Coordenação/farmacologia , Complexos de Coordenação/síntese química , Espécies Reativas de Oxigênio/metabolismo , Flavonóis/química , Flavonóis/farmacologia , Antineoplásicos/química , Antineoplásicos/farmacologia , Sobrevivência Celular/efeitos dos fármacos , Estrutura Molecular , Relação Dose-Resposta a Droga , Linhagem Celular Tumoral
19.
Anticancer Res ; 44(3): 901-910, 2024 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-38423634

RESUMO

BACKGROUND/AIM: Fisetin is a yellow-coloring flavonoid that can be found in a wide variety of plants, vegetables, and fruits, such as strawberries, apples, and grapes. It has been shown to have biological activity by targeting different pathways regulating survival and death and to bear antioxidant and anti-inflammatory activity. Fisetin was shown to be cytotoxic on different cancer cell lines and has the ability to kill therapy-induced senescent cancer cells. The aim of the study was to investigate the DNA damaging and cytotoxic potential of fisetin and its ability to enhance the killing effect of temozolomide on glioblastoma cells. MATERIALS AND METHODS: We used LN229 glioblastoma cells and measured survival and apoptosis by flow cytometry, DNA strand breaks by the alkaline comet and γH2AX assay, and the DNA damage response by western blot analysis. RESULTS: Fisetin was cytotoxic on glioblastoma cells, inducing apoptosis. In the dose range of 40-80 µM it also induced DNA damage, as measured by the alkaline comet and γH2AX assay, and triggered DNA damage response, as revealed by p53 activation. Furthermore, fisetin enhanced the genotoxic effect of methyl methanesulfonate, presumably due to inhibition of DNA repair processes. When administered together with temozolomide, the first-line therapeutic for glioblastoma, it enhanced cell death, reduced the yield of senescent cells following treatment and exhibited senolytic activity on glioblastoma cells. CONCLUSION: Data show that high-dose fisetin has a genotoxic potential and suggest that, harnessing the cytotoxic and senolytic activity of the flavonoid, it may enhance the effect of anticancer drugs and eliminate therapy-induced senescent cells. Therefore, it may be useful for adjuvant cancer therapy, including glioblastoma, which is worth to be studied in clinical trials.


Assuntos
Antineoplásicos , Glioblastoma , Humanos , Temozolomida/farmacologia , Temozolomida/uso terapêutico , Glioblastoma/tratamento farmacológico , Glioblastoma/metabolismo , Senoterapia , Flavonóis/farmacologia , Flavonóis/uso terapêutico , Antineoplásicos/farmacologia , Flavonoides/farmacologia , Apoptose , Dano ao DNA , Linhagem Celular Tumoral , DNA
20.
J Agric Food Chem ; 72(7): 3695-3706, 2024 Feb 21.
Artigo em Inglês | MEDLINE | ID: mdl-38324412

RESUMO

Novel N-ethy-2-pyrrolidinone-substituted flavonols, myricetin alkaloids A-C (1-3), quercetin alkaloids A-C (4a, 4b, and 5), and kaempferol alkaloids A and B (6 and 7), were prepared from thermal reaction products of myricetin, quercetin, kaempferol─l-theanine, respectively. We used HPLC-ESI-HRMS/MS to detect 1-7 in 14 cultivars of green tea and found that they were all present in "Shuchazao," "Longjing 43", "Fudingdabai", and "Zhongcha 108" green teas. The structures of 1-4 and 6 were determined by extensive 1D and 2D NMR spectroscopies. These flavonol alkaloids along with their skeletal flavonols were assessed for anti-Alzheimer's disease effect based on molecular docking, acetylcholinesterase inhibition, and the transgenic Caenorhabditis elegans CL4176 model. Compound 7 strongly binds to the protein amyloid ß (Aß1-42) through hydrogen bonds (BE: -9.5 kcal/mol, Ki: 114.3 nM). Compound 3 (100 µM) is the strongest one in significantly extending the mean lifespan (13.4 ± 0.5 d, 43.0% promotion), delaying the Aß1-42-induced paralysis (PT50: 40.7 ± 1.9 h, 17.1% promotion), enhancing the locomotion (140.0% promotion at 48 h), and alleviating glutamic acid (Glu)-induced neurotoxicity (153.5% promotion at 48 h) of CL4176 worms (p < 0.0001).


Assuntos
Alcaloides , Doença de Alzheimer , Animais , Chá/química , Peptídeos beta-Amiloides/genética , Peptídeos beta-Amiloides/farmacologia , Caenorhabditis elegans/genética , Quercetina/farmacologia , Acetilcolinesterase , Simulação de Acoplamento Molecular , Alcaloides/farmacologia , Alcaloides/química , Doença de Alzheimer/tratamento farmacológico , Doença de Alzheimer/genética , Flavonóis/farmacologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...