Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 5 de 5
Filter
Add more filters










Database
Language
Publication year range
1.
Pharm Biol ; 61(1): 1298-1309, 2023 Dec.
Article in English | MEDLINE | ID: mdl-37606265

ABSTRACT

CONTEXT: Danggui Buxue Decoction (DBD), a traditional Chinese medicine formula, has the potential to enhance the antitumor effect of gemcitabine in non-small cell lung cancer (NSCLC) treatment by increasing gemcitabine's active metabolites. However, whether gemcitabine affects the pharmacokinetics of DBD's major components remains unclear. OBJECTIVE: This study evaluates the herb-drug interaction between DBD's major components and gemcitabine and validates the underlying pharmacokinetic mechanism. MATERIALS AND METHODS: The pharmacokinetics of 3.6 g/kg DBD with and without a single-dose administration of 50 mg/kg gemcitabine was investigated in Sprague-Dawley rats. The effects of gemcitabine on intestinal permeability, hepatic microsomal enzymes in rat tissues, and CYP3A overexpressing HepG2 cells were determined using western blot analysis. RESULTS: The combination of gemcitabine significantly altered the pharmacokinetic profiles of DBD's major components in rats. The Cmax and AUC of calycosin-7-O-ß-d-glucoside notably increased through sodium-glucose transporter 1 (SGLT-1) expression promotion. The AUC of ligustilide and ferulic acid was also significantly elevated with the elimination half-life (t1/2) prolonged by 2.4-fold and 7.8-fold, respectively, by down-regulating hepatic CYP3A, tight junction proteins zonula occludens-1 (ZO-1) and occludin expression. DISCUSSION AND CONCLUSIONS: Gemcitabine could modulate the pharmacokinetics of DBD's major components by increasing intestinal permeability, enhancing transporter expression, and down-regulating CYP3A. These findings provide critical information for clinical research on DBD as an adjuvant for NSCLC with gemcitabine and help make potential dosage adjustments more scientifically and rationally.


Subject(s)
Carcinoma, Non-Small-Cell Lung , Lung Neoplasms , Rats , Animals , Carcinoma, Non-Small-Cell Lung/drug therapy , Gemcitabine , Cytochrome P-450 CYP3A , Down-Regulation , Rats, Sprague-Dawley , Lung Neoplasms/drug therapy
2.
Front Psychol ; 10: 2856, 2019.
Article in English | MEDLINE | ID: mdl-31993003

ABSTRACT

OBJECTIVES: To explore the influence of different background factors on middle school PE teachers' self-efficacy, work input and creative teaching, and to reveal the relationship between teaching self-efficacy and work input on creative teaching. METHODS: By means of self-efficacy, work engagement and creative teaching scale, a questionnaire survey was conducted among middle school PE teachers, and the data were processed and modeled by SPSS and AMOS statistical analysis software. RESULTS: Physical education (PE) teachers' self-teaching effectiveness was influenced by background factors such as gender, age, teaching age, full-time or part-time work and educational level. Work input was only affected by age, teaching experience and educational level, while creative teaching seemed to be only related to background factors such as educational background and full-time or part-time work; PE teachers' general teaching effectiveness and personal teaching effectiveness had significant positive effects on energy input, concentration input, dedication input, cognitive creativity, skill creativity and emotional creativity; Concentration input had a significant positive impact on the three-dimensional of creative teaching, while energy input and dedication input had no impact on the three-dimensional of creative teaching; Work input as an intermediary variable of self-efficacy's influence on creative teaching had been verified, but the real intermediary role was not the whole work input, but the concentration input in its structure. CONCLUSION: Both general and individual teaching effectiveness had positive effects on work input and creative teaching, but the energetic and dedicated input in work input cannot promote teachers' creative teaching effectively. Therefore, the professional ethics training of PE teachers in their enthusiasm and dedication to work should be strengthened.

3.
Mol Neurobiol ; 54(6): 4292-4302, 2017 08.
Article in English | MEDLINE | ID: mdl-27339879

ABSTRACT

Caspase family has been recognized to be involved in dopaminergic (DA) neuronal death and to exert an unfavorable role in Parkinson's disease (PD) pathology. Our previous study has revealed that caspase-1, as an important component of NLRP3 inflammasome, induces microglia-mediated neuroinflammation in the pathogenesis of PD. However, the role of caspase-1 in DA neuronal degeneration in the onset of PD remains unclear. Here, we showed that caspase-1 knockout ameliorated DA neuronal loss and dyskinesia in 1-methyl-4-phenyl-1, 2, 3, 6-tetrahydropyridine/probenecid (MPTP/p)-induced PD model mice. We further found that caspase-1 knockout decreased MPTP/p-induced caspase-7 cleavage, subsequently inhibited nuclear translocation of poly (ADP-ribose) polymerase 1 (PARP1), and reduced the release of apoptosis-inducing factor (AIF). Consistently, we demonstrated that caspase-1 inhibitor suppressed caspase-7/PARP1/AIF-mediated apoptosis pathway by 1-methyl-4-phenylpyridinium ion (MPP+) stimulation in SH-SY5Y cells. Caspase-7 overexpression reduced the protective effects of caspase-1 inhibitor on SH-SY5Y cell apoptosis. Collectively, our results have revealed that caspase-1 regulates DA neuronal death in the pathogenesis of PD in mice via caspase-7/PARP1/AIF pathway. These findings will shed new insight into the potential of caspase-1 as a target for PD therapy.


Subject(s)
Apoptosis Inducing Factor/metabolism , Caspase 1/deficiency , Caspase 7/metabolism , Dopaminergic Neurons/pathology , Parkinson Disease/enzymology , Parkinson Disease/pathology , 1-Methyl-4-phenyl-1,2,3,6-tetrahydropyridine , Amino Acid Chloromethyl Ketones/pharmacology , Animals , Caspase 1/metabolism , Cell Death/drug effects , Cell Line, Tumor , Disease Models, Animal , Dopaminergic Neurons/metabolism , Down-Regulation/drug effects , Enzyme Activation/drug effects , Humans , Mice, Knockout , Models, Biological , Motor Activity/drug effects , Poly(ADP-ribose) Polymerases/metabolism
4.
CNS Neurosci Ther ; 22(9): 729-39, 2016 09.
Article in English | MEDLINE | ID: mdl-27306494

ABSTRACT

AIM: Ginkgolide B (GB) has shown neuroprotective effect in treating ischemic stroke, related to its property of anti-inflammation. Nevertheless, it is unclear whether GB is able to modulate microglia/macrophage polarization, which has recently been proven to be vital in the pathology of ischemic stroke. METHODS: We performed transient middle cerebral artery occlusion (tMCAO) on C57BL/6J male mice and induced cultured BV2 microglia and primary bone marrow-derived macrophages to be M1/2 phenotype by LPS+ interferon-γ and IL-4, respectively. Immunofluorescence and flow cytometry were used for detecting the specialized protein expression of M1/2, such as CD206 and CD16/32. qPCR was utilized to detect the signature gene change of M1/2. RESULTS: GB significantly reduced cerebral ischemic damage and ameliorated the neurological deficits of mice after tMCAO. More importantly, our experiments proved that GB promoted microglia/macrophage transferring from inflammatory M1 phenotype to a protective, anti-inflammatory M2 phenotype in vivo or vitro. CV3988 and silencing the platelet activator factor (PAF) receptor by siRNA demonstrated that PAF receptor was involved in the modulation of microglia/macrophage polarization. CONCLUSION: Our results reveal a novel pharmacological effect of GB in modulating microglia/macrophage polarization after tMCAO, thus deepening our understanding of neuroprotective mechanisms of GB in treatment of ischemic stroke. Furthermore, this new mechanism may allow GB to be used in many other microglia/macrophage polarization-related inflammatory diseases.


Subject(s)
Cell Polarity/drug effects , Ginkgolides/pharmacology , Ginkgolides/therapeutic use , Infarction, Middle Cerebral Artery/drug therapy , Lactones/pharmacology , Lactones/therapeutic use , Microglia/drug effects , Animals , Brain Edema/drug therapy , Brain Edema/etiology , Cell Hypoxia/drug effects , Cells, Cultured , Cerebral Cortex/cytology , Culture Media, Conditioned/pharmacology , Cytokines/genetics , Cytokines/metabolism , Disease Models, Animal , Enzyme Inhibitors/pharmacology , Gene Expression Regulation/drug effects , Glucose/deficiency , Infarction, Middle Cerebral Artery/pathology , Lipopolysaccharides/pharmacology , Male , Mice , Mice, Inbred C57BL , Motor Activity/drug effects , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/metabolism
5.
Article in English | MEDLINE | ID: mdl-27207922

ABSTRACT

BACKGROUND: Emerging evidence indicates that NLRP3 inflammasome-induced inflammation plays a crucial role in the pathogenesis of depression. Thus, inhibition of NLRP3 inflammasome activation may offer a therapeutic benefit in the treatment of depression. Fluoxetine, a widely used antidepressant, has been shown to have potential antiinflammatory activity, but the underlying mechanisms remain obscure. METHODS: We used a chronic mild stress model and cultured primary macrophage/microglia to investigate the effects of fluoxetine on NLRP3 inflammasome and its underlying mechanisms. RESULTS: We demonstrated that fluoxetine significantly suppressed NLRP3 inflammasome activation, subsequent caspase-1 cleavage, and interleukin-1ß secretion in both peripheral macrophages and central microglia. We further found that fluoxetine reduced reactive oxygen species production, attenuated the phosphorylation of double-stranded RNA-dependent protein kinase, and inhibited the association of protein kinase with NLRP3. These data indicate that fluoxetine inhibits the activation of NLRP3 inflammasome via downregulating reactive oxygen species-protein kinase-NLRP3 signaling pathway. Correspondingly, in vivo data showed that fluoxetine also suppressed NLRP3 inflammasome activation in hippocampus and macrophages of chronic mild stress mice and alleviated chronic mild stress-induced depression-like behavior. CONCLUSIONS: Our findings reveal that fluoxetine confers an antidepressant effect partly through inhibition of peripheral and central NLRP3 inflammasome activation and suggest the potential clinical use of fluoxetine in NLRP3 inflammasome-driven inflammatory diseases such as depression.

SELECTION OF CITATIONS
SEARCH DETAIL