Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 7 de 7
Filtrar
1.
J Food Sci Technol ; 55(7): 2758-2764, 2018 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-30042592

RESUMO

Phyllanthus emblica L. is a tropical deciduous tree producing edible berries with potential medicinal value. In this study, a novel water-soluble phyllanthus emblica polysaccharide (PEP) from the berries was isolated by precipitation and purification, and analyzed for its structure features. The results showed that PEP was a α-pyran acidic heteropolysaccharide with a molecular weight of 1.31 × 105 Da, which included galacturonic acid, galactose, rhamnose, and arabinose with a molar ratio of 3.21:6.59:1:0.23. Furthermore, the antioxidant activities of PEP were determined and showed remarkable antioxidant capacities in DPPH, superoxide anion- and hydroxyl-radical scavenging, ferric-reducing antioxidant power, and lipid peroxidation inhibition. This work indicated that PEP as a natural antioxidant agent from the berries of Phyllanthus emblica L. had potential application for developing valuable nutraceutical in food industry.

2.
Biochem Biophys Res Commun ; 469(1): 55-61, 2016 Jan 01.
Artigo em Inglês | MEDLINE | ID: mdl-26592661

RESUMO

Accumulating evidence has demonstrated that arsenic trioxide (ATO) exhibits its anti-cancer activities in a variety of human malignancies. Recent studies have revealed that ATO regulated multiple microRNAs (miRNAs) in human cancers. However, the exact mechanism of ATO-mediated tumor suppressive function has not been fully elucidated. In the present study, we explore whether ATO governed oncogenic miR-27a in breast cancer cells by multiple methods such as MTT assay, RT-PCR, Wound healing assay, Western blotting analysis, migration, Transwell invasion assay, and transfection. Our results showed that ATO inhibited cell growth, migration, invasion, and induced cell apoptosis in breast cancer cells. Further molecular analysis dissected that ATO inhibited miR-27a expression in breast cancer cells. Moreover, inhibition of miR-27a suppressed cell growth, migration, invasion, and trigged cell apoptosis, whereas overexpression of miR-27a enhanced cell growth, motility, and inhibited apoptosis in breast cancer cells. Notably, we found that miR-27a inhibitor treatment potentiates ATO-induced breast cancer cell growth inhibition, apoptosis and motility inhibition. However, overexpression of miR-27a partly abrogated ATO-mediated anti-tumor activity. Our findings provide a novel anti-tumor mechanism of ATO involved in miR-27a for the treatment of breast cancer.


Assuntos
Arsenicais/administração & dosagem , Neoplasias da Mama/tratamento farmacológico , Neoplasias da Mama/genética , Movimento Celular/genética , Proliferação de Células/genética , MicroRNAs/genética , Óxidos/administração & dosagem , Antineoplásicos/administração & dosagem , Apoptose/efeitos dos fármacos , Apoptose/genética , Trióxido de Arsênio , Neoplasias da Mama/patologia , Linhagem Celular Tumoral , Movimento Celular/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Relação Dose-Resposta a Droga , Humanos , Invasividade Neoplásica , Resultado do Tratamento
3.
BMC Pharmacol Toxicol ; 23(1): 18, 2022 03 25.
Artigo em Inglês | MEDLINE | ID: mdl-35337381

RESUMO

BACKGROUND: Propofol and dexmedetomidine (DEX) are widely used in general anesthesia, and exert toxic and protective effects on hippocampal neurons, respectively. The study sought to investigate the molecular mechanisms of DEX-mediated neuroprotection against propofol-induced hippocampal neuron injury in mouse brains. METHODS: Hippocampal neurons of mice and HT22 cells were treated with propofol, DEX, and propofol+DEX. In addition, transfection of miR-377-5p mimics or inhibitors was performed in HT22 cells. Neuronal apoptosis was evaluated by a means of terminal deoxynucleotidyl transferase (TdT)-mediated dUTP nick end labeling (TUNEL) or Hochest 33,258 staining; Arc positive expression in hippocampus tissues was detected using a microscope in immunohistochemistry assays; miRNA-377-5p expression was quantified by RT-qPCR; the protein levels of Arc, DNMT3A, and DNMT3B were determined using western blot; Cell Counting Kit-8 (CCK-8) assay was used to detect the viability and apoptotic rate of the neurons; methylation analysis in the miR-377-5p promoter was performed through methylated DNA immunoprecipitation (MeDIP) assay; dual luciferase reporter assay was performed to confirm whether Arc was under targeted regulation of miR-377-5p. RESULTS: In the current study, both in vitro and in vivo, propofol treatment induced hippocampal neuron apoptosis and suppressed cell viability. DNMT3A and DNMT3B expression levels were decreased following propofol treatment, resulting in lowered methylation in the miR-377-5p promoter region and then enhanced expression of miR-377-5p, leading to a decrease in the expression of downstream Arc. Conversely, the expression levels of DNMT3A and DNMT3B were increased following DEX treatment, thus methylation in miR-377-5p promoter region was improved, and miR-377-5p expression was decreased, leading to an increase in the expression of downstream Arc. Eventually, DEX pretreatment protected hippocampal neurons against propofol-induced neurotoxicity by recovering the expression levels of DNMT3A, miR-377-5p, and Arc to the normal levels. Additionally, DNMT3A knockdown improved miR-377-5p expression but reduced Arc expression, and DNMT3A overexpression exerted the opposite effects. Dual luciferase reporter assay revealed a binding target between miR-377-5p and Arc 3'UTR. The neuroprotective effect of DEX against propofol-induced neuronal apoptosis was diminished after Arc knockdown. Silencing Arc independently triggered the apoptosis of HT22 cells, which was alleviated through transfection of miR-377-5p inhibitors. CONCLUSIONS: DEX reduced propofol-induced hippocampal neuron injury via the miR-377-5p/Arc signaling pathway.


Assuntos
Dexmedetomidina , MicroRNAs , Propofol , Animais , Apoptose , Proteínas do Citoesqueleto , Dexmedetomidina/farmacologia , Hipocampo , Camundongos , MicroRNAs/genética , MicroRNAs/metabolismo , Proteínas do Tecido Nervoso , Neurônios , Propofol/metabolismo , Propofol/farmacologia
4.
Iran J Pharm Res ; 20(2): 515-522, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34567178

RESUMO

Postoperative cognitive dysfunction is a common postoperative neurological complication in elderly patients, and has some relationship with neuroinflammation. some studies have shown ability of dexmedetomidine to improve cognitive performance in elderly individuals who underwent thoracic surgery. Therefore, our study hypothesized that dexmedetomidine treatment may reduce the incidence of POCD in elderly patients.In addition,this study detected the antineuroinflammatory effects of dexmedetomidine by ß-amyloid aggregation inhibitors and release of cytokines in elderly patients . The results show that dexmedetomidine used during operation can inhibit the postoperative release of Aß and cytokines in elderly patients, and dexmedetomidine used during operation can reduce the incidence of postoperative cognitive dysfunction, with dose-dependence. These results provide a clinical application direction for clinical anesthesiologists and ICU physicians.

5.
EXCLI J ; 17: 302-311, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-29743866

RESUMO

NOD-like receptor 3 (NLRP3) plays critical roles in the initiation of inflammasome-mediated inflammation in microglia, thus becomes an important therapeutic target of Alzheimer's disease (AD). Dexmedetomidine (Dex), a new type of clinical anesthetic agent, shows anti-inflammatory properties and inhibits postoperative cognitive dysfunction in AD patients. The present study was aimed to investigate effect of Dex on NLRP3 activity in activated microglia and reveal the underlying mechanisms. The human microglia clone 3 (HMC3) cells were exposed to 100 ng/ml LPS and 5 mM ATP, in the presence and absence of doses of Dex. Data from ELISA and Western blot assays showed that Dex abrogated the promoting effects of LPS/ATP on the release of pro-inflammatory cytokines including IL-1ß and IL-18 in the cell medium and the expression of NLRP3 and its downstream target caspase-1 in HMC3 cells. Furthermore, the present study found that exposure of HMC3 cells to LPS/ATP increased nuclear protein levels of transcription factor c-Fos, but treatment with Dex reversed the increase in c-Fos, as indicated by Western blot and immunofluorescence measures. Luciferase reported assay revealed that c-Fos can bind to the promoter region of NLRP3 gene and positively regulate the expression. These results suggest that Dex inhibiting c-Fos nuclear protein levels promoted by LPS/ATP blocks the up-regulation of NLRP3. This suggestion is supported by co-immunoprecipitation and PCR studies, in which Dex decreased the amount of c-Fos that binds to NLRP3 under the stimulation of LPS/ATP. The present study revealed that Dex inhibits inflammation in microglia cells under stimulation of LPS and ATP by c-Fos/NLRP3/caspase-1 cascades, which adds new understanding of the anti-inflammatory mechanism of Dex.

6.
J Huazhong Univ Sci Technolog Med Sci ; 36(3): 428-433, 2016 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-27376816

RESUMO

This study aimed to establish a new propofol target-controlled infusion (TCI) model in animals so as to study the general anesthetic mechanism at multi-levels in vivo. Twenty Japanese white rabbits were enrolled and propofol (10 mg/kg) was administrated intravenously. Artery blood samples were collected at various time points after injection, and plasma concentrations of propofol were measured. Pharmacokinetic modeling was performed using WinNonlin software. Propofol TCI within the acquired parameters integrated was conducted to achieve different anesthetic depths in rabbits, monitored by narcotrend. The pharmacodynamics was analyzed using a sigmoidal inhibitory maximal effect model for narcotrend index (NI) versus effect-site concentration. The results showed the pharmacokinetics of propofol in Japanese white rabbits was best described by a two-compartment model. The target plasma concentrations of propofol required at light anesthetic depth was 9.77±0.23 µg/mL, while 12.52±0.69 µg/mL at deep anesthetic depth. NI was 76.17±4.25 at light anesthetic depth, while 27.41±5.77 at deep anesthetic depth. The effect-site elimination rate constant (ke0) was 0.263/min, and the propofol dose required to achieve a 50% decrease in the NI value from baseline was 11.19 µg/mL (95% CI, 10.25-13.67). Our results established a new propofol TCI animal model and proved the model controlled the anesthetic depth accurately and stably in rabbits. The study provides a powerful method for exploring general anesthetic mechanisms at different anesthetic depths in vivo.


Assuntos
Anestésicos Intravenosos/farmacocinética , Modelos Estatísticos , Propofol/farmacocinética , Anestésicos Intravenosos/sangue , Animais , Monitoramento de Medicamentos , Infusões Intravenosas , Propofol/sangue , Coelhos , Software
7.
Oncotarget ; 7(14): 18247-61, 2016 Apr 05.
Artigo em Inglês | MEDLINE | ID: mdl-26919096

RESUMO

MiR-206 is low expression in lung cancers and associated with cancer metastasis. However, the roles of miR-206 in epithelial-mesenchymal transition (EMT) and angiogenesis in lung cancer remain unknown. In this study, we find that hepatocyte growth factor (HGF) induces EMT, invasion and migration in A549 and 95D lung cancer cells, and these processes could be markedly inhibited by miR-206 overexpression. Moreover, we demonstrate that miR-206 directly targets c-Met and inhibits its downstream PI3k/Akt/mTOR signaling pathway. In contrast, miR-206 inhibitors promote the expression of c-Met and activate the PI3k/Akt/mTOR signaling, and this effect could be attenuated by the PI3K inhibitor. Moreover, c-Met overexpression assay further confirms the significant inhibitory effect of miR-206 on HGF-induced EMT, cell migration and invasion. Notably, we also find that miR-206 effectively inhibits HGF-induced tube formation and migration of human umbilical vein endothelial cells (HUVECs), and the mechanism is also related to inhibition of PI3k/Akt/mTOR signaling. Finally, we reveal the inhibitory effect of miR-206 on EMT and angiogenesis in xenograft tumor mice model. Taken together, miR-206 inhibits HGF-induced EMT and angiogenesis in lung cancer by suppressing c-Met/PI3k/Akt/mTOR signaling. Therefore, miR-206 might be a potential target for the therapeutic strategy against EMT and angiogenesis of lung cancer.


Assuntos
Carcinoma Pulmonar de Células não Pequenas/patologia , Transição Epitelial-Mesenquimal/genética , Fator de Crescimento de Hepatócito/metabolismo , Neoplasias Pulmonares/patologia , MicroRNAs/genética , Neovascularização Patológica/patologia , Fosfatidilinositol 3-Quinases/metabolismo , Proteínas Proto-Oncogênicas c-akt/metabolismo , Proteínas Proto-Oncogênicas c-met/metabolismo , Serina-Treonina Quinases TOR/metabolismo , Células A549 , Animais , Linhagem Celular Tumoral , Movimento Celular/fisiologia , Células Endoteliais da Veia Umbilical Humana/metabolismo , Humanos , Pulmão/irrigação sanguínea , Pulmão/patologia , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Nus , MicroRNAs/metabolismo , Invasividade Neoplásica/patologia , Transplante de Neoplasias , Inibidores de Fosfoinositídeo-3 Quinase , Transdução de Sinais , Transplante Heterólogo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA