Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 4 de 4
Filtrar
Mais filtros

Base de dados
Tipo de documento
País de afiliação
Intervalo de ano de publicação
1.
Arch Biochem Biophys ; 744: 109691, 2023 08.
Artigo em Inglês | MEDLINE | ID: mdl-37473980

RESUMO

Ferroptosis, an iron-dependent cell death, is caused by lipid peroxidation. Noteworthily, accumulation of iron and lipid peroxidation are found in the proximity of the neuritic plaque, a hallmark of Alzheimer's disease (AD), but the relationship between ferroptosis and neuroinflammation in AD is unclear. Silibinin, extracted from the Silybum marianum, is possibly developed as an agent for AD treatment from its neuroprotective effect, but the effect of silibinin on sporadic AD that accounts for more than 95% of AD remains unclear. To determine whether silibinin alleviates the pathogenesis of sporadic AD and investigate the underlying mechanisms, STZ-treated HT22 murine hippocampal neurons and intracerebroventricular injection of streptozotocin (ICV-STZ) rats, a sporadic AD model, were used in this study. Results show that silibinin not only promotes survival of STZ-treated HT22 cells, but also ameliorates the cognitive impairment and anxiety/depression-like behavior of ICV-STZ rats. We here demonstrate that silibinin evidently inhibits the protein level of p53 as well as upregulates the protein level of cystine/glutamate antiporter SLC7A11 and ferroptosis inhibitor GPX4, but not p21, leading to the protection against STZ-induced ferroptotic damage. Immunofluorescent staining also shows that accumulation of lipid peroxidation induced by ferroptotic damage leads to increased fluorescence of 8-oxo-deoxyguanosine (8-OHDG), a maker of oxidized DNA. The oxidized DNA then leaks to the cytoplasm and upregulates the expression of the stimulator of interferon gene (STING), which triggers the production of IFN-ß and other inflammatory cascades including NF-κB/TNFα and NLRP3/caspase 1/IL-1ß. However, the treatment with silibinin blocks the above pathological changes. Moreover, in HT22 cells with/without STZ treatment, GPX4-knockdown increases the protein level of STING, indicating that the ferroptotic damage leads to the activation of STING signaling pathway. These results imply that silibinin exerts neuroprotective effect on an STZ-induced sporadic AD model by downregulating ferroptotic damage and thus the downstream STING-mediated neuroinflammation.


Assuntos
Doença de Alzheimer , Fármacos Neuroprotetores , Ratos , Camundongos , Animais , Doença de Alzheimer/induzido quimicamente , Doença de Alzheimer/tratamento farmacológico , Doença de Alzheimer/metabolismo , Silibina/farmacologia , Silibina/uso terapêutico , Regulação para Baixo , Doenças Neuroinflamatórias , Fármacos Neuroprotetores/farmacologia , Fármacos Neuroprotetores/uso terapêutico , Estreptozocina/efeitos adversos , Modelos Animais de Doenças
2.
Phytomedicine ; 109: 154594, 2023 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-36610115

RESUMO

BACKGROUND: Over-activation of N-methyl-D-aspartate receptors (NMDARs) is involved in sporadic Alzheimer's disease. Silibinin, a natural flavonoid gained from the seeds of Silybum marianum, exerts neuroprotective effects on sporadic AD models, but its impacts on NMDARs remain unknown. PURPOSE: To study silibinin's regulatory effects on NMDARs pathway in sporadic AD models. METHODS: MTT assay, western blotting, confocal microscopy, flow cytometry, RT-PCR, and siRNA transfection etc. were used for cellular and molecular studies. The direct interactions between silibinin and NMDAR subunits were evaluated by computational molecular docking, drug affinity responsive target stability (DARTS) assay and cellular thermal shift assay (CETSA). Y maze test, novel objects recognition test and Morris water maze test were conducted to examine the learning and memory ability of rats. RESULTS: An in vitro AD model was established by treating HT22 murine hippocampal neurons with streptozotocin (STZ), as evidenced by the amyloid ß (Aß) deposition and hyperphosphorylation of tau proteins. Silibinin shows protection of neurons against STZ-induced cell damage. It is noteworthy that STZ-induced cellular calcium influx is inhibited by silibinin-treatment, indicating the possible modulation of calcium channels. Studies on NMDARs, the most widely distributed calcium channel, by using molecular docking, DARTS and CESTA, reveal that the GluN2B subunit, but not GluN2A, is the potential target of silibinin. Further studies using the pharmacological agonist (NMDA) and the GluN2B-specific inhibitor (Ifenprodil) or siRNA, indicate that the protection by silibinin treatment from STZ-induced cytotoxicity is medicated through interference with GluN2B-containing NMDARs, followed by the upregulation of CaMKIIα/ BDNF/ TrkB signaling pathway and improved levels of synaptic proteins (SYP and PSD-95). The results in vivo using rats intracerebroventricularly injected with STZ (ICV-STZ), a well-established sporadic AD model, confirm that silibinin improves learning and memory ability in association with modulation of the GluN2B/CaMKIIα/ BDNF/TrkB signaling pathway. CONCLUSION: Inhibiting over-activation of GluN2B-containing NMDARs is involved in the neuroprotective effect of silibinin on STZ-induced sporadic AD models.


Assuntos
Doença de Alzheimer , Fármacos Neuroprotetores , Ratos , Camundongos , Animais , Doença de Alzheimer/induzido quimicamente , Doença de Alzheimer/tratamento farmacológico , Receptores de N-Metil-D-Aspartato/metabolismo , Peptídeos beta-Amiloides/metabolismo , Silibina/farmacologia , Fármacos Neuroprotetores/farmacologia , Fármacos Neuroprotetores/uso terapêutico , Estreptozocina , Fator Neurotrófico Derivado do Encéfalo/metabolismo , Simulação de Acoplamento Molecular , Modelos Animais de Doenças
3.
Sci China Life Sci ; 63(1): 68-79, 2020 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-31463739

RESUMO

Mouth ulcer is associated with inflammation and high risk of bacterial infection, which aggravates the patient's condition. Currently, there is no effective treatment for mouth ulcer. Herein, we report that vitamin-modified iron oxide nanoparticles improve the healing of mouth ulcer through anti-inflammation and antibacterial activities. We discovered that vitamin B2 (VB2) modified iron oxide nanoparticles performed enhanced peroxidase-like, catalase-like, and superoxide dismutase (SOD)-like activities, acting as typical iron oxide nanozymes (IONzymes) with triad activities. In particular, VB2 modification significantly improved the SOD-like activity, thus providing a reactive oxygen species (ROS)-scavenging ability. Cellular antioxidant experiments showed that vitamin B2 modified IONzymes (VB2-IONzymes) protect human oral keratinocytes (HOK) and BALB/3T3 cells from hydrogen peroxide (H2O2), and these cells have high biocompatibility to eukaryotic cells. In addition, VB2-IONzymes exerted an antibacterial activity against Streptococcus mutans, Staphylococcus aureus, and Escherichia coli. Importantly, VB2-IONzymes accelerated the recovery of mouth ulcer and reduced the local secretion of inflammatory factors in mouse ulcer model via ROS scavenging and antibacterial activity. Taken together, our work demonstrates that vitamin B2 modification endows iron oxide nanoparticles with enhanced enzyme-like activities and VB2-IONzymes may be a promising reagent in the treatment of mouth ulcer because of their intrinsic anti-inflammation and antibacterial capabilities.


Assuntos
Compostos Férricos/química , Nanopartículas Metálicas/química , Úlceras Orais/tratamento farmacológico , Riboflavina/química , Cicatrização/efeitos dos fármacos , Animais , Antibacterianos/química , Antibacterianos/farmacologia , Antioxidantes/química , Antioxidantes/farmacologia , Células 3T3 BALB , Materiais Biomiméticos/química , Materiais Biomiméticos/farmacologia , Catalase/metabolismo , Linhagem Celular , Humanos , Peróxido de Hidrogênio/metabolismo , Queratinócitos/metabolismo , Camundongos , Peroxidase/metabolismo , Espécies Reativas de Oxigênio/metabolismo , Riboflavina/farmacologia , Superóxido Dismutase/metabolismo
4.
Nat Commun ; 9(1): 3713, 2018 09 13.
Artigo em Inglês | MEDLINE | ID: mdl-30213949

RESUMO

The use of natural substance to ward off microbial infections has a long history. However, the large-scale production of natural extracts often reduces antibacterial potency, thus limiting practical applications. Here we present a strategy for converting natural organosulfur compounds into nano-iron sulfides that exhibit enhanced antibacterial activity. We show that compared to garlic-derived organosulfur compounds nano-iron sulfides exhibit an over 500-fold increase in antibacterial efficacy to kill several pathogenic and drug-resistant bacteria. Furthermore, our analysis reveals that hydrogen polysulfanes released from nano-iron sulfides possess potent bactericidal activity and the release of polysulfanes can be accelerated by the enzyme-like activity of nano-iron sulfides. Finally, we demonstrate that topical applications of nano-iron sulfides can effectively disrupt pathogenic biofilms on human teeth and accelerate infected-wound healing. Together, our approach to convert organosulfur compounds into inorganic polysulfides potentially provides an antibacterial alternative to combat bacterial infections.


Assuntos
Antibacterianos/química , Biofilmes/efeitos dos fármacos , Alho/química , Proteínas Ferro-Enxofre/química , Sulfetos/química , Compostos de Enxofre/química , Células 3T3 , Compostos Alílicos/química , Animais , Antioxidantes/química , Bactérias/efeitos dos fármacos , Infecções Bacterianas/tratamento farmacológico , Cálcio/química , Sobrevivência Celular , Esmalte Dentário/efeitos dos fármacos , Esmalte Dentário/microbiologia , Dentina/química , Farmacorresistência Bacteriana , Fibroblastos/metabolismo , Humanos , Queratinócitos/citologia , Malondialdeído/química , Camundongos , Camundongos Endogâmicos BALB C , Extratos Vegetais/química , Espécies Reativas de Oxigênio , Streptococcus mutans , Dente/efeitos dos fármacos , Dente/microbiologia , Cicatrização
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA