Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 5 de 5
Filter
1.
Article | IMSEAR | ID: sea-210740

ABSTRACT

Alpha-mangostin is the major component in Mangosteen (Garcinia mangostana Linn) pericarp having severalpharmacological activities including reducing blood pressure, antidiabetic, anticancer, and antioxidants. The objectiveof this study was to develop Fourier transform infrared spectroscopy-multivariate calibration of partial least square(PLS) for quantitative analysis of alpha-mangostin and to classify mangosteen pericarp using principal componentanalysis. Mangosteen pericarps from different locations (Java provinces and South Sulawesi, Republic of Indonesia)were extracted using ethanol and were subjected to high performance liquid chromatography (HPLC) for the analysisof alpha-mangostin and Fourier transform infrared (FTIR) spectroscopy measurements. HPLC was used to determinethe levels of alpha-mangostin and used as actual values during FTIR spectroscopy analysis. The prediction of alphamangostin was obtained from the correlation between actual values and FTIR predicted values and facilitated withthe PLS model. The results showed that the wavenumbers region of 3,825–937 cm−1 offered a reliable model with acoefficient correlation (r) value of 0.9927 and root mean square error of calibration of 0.0831%. The validation modelsalso exhibited the accurate and precise results for the prediction of alpha-mangostin with an r-value of 0.9754 androot mean square error of prediction value of 0.174%. Furthermore, the chemometrics of principal component analysisusing variables of absorbances at selected fingerprint (1,000–800 cm−1) could classify mangosteen pericarp fromdifferent regions. FTIR spectroscopy combined with chemometrics offered a reliable method for quality assurance ofmangosteen pericarp

2.
Asian Pacific Journal of Tropical Biomedicine ; (12): 325-332, 2020.
Article in Chinese | WPRIM | ID: wpr-823947

ABSTRACT

Objective: To investigate the antibiofilm activity of alpha-mangostin (AMG) loaded nanoparticle (nanoAMG) against dental caries pathogen Streptococcus mutans. Methods: AMG was isolated from the peels of Garcinia mangostana L. using silica gel columns and chemically analysed by high performance liquid chromatography and nuclear magnetic resonance. NanoAMG was prepared using the solvent evaporation method combined with high-speed homogenization. The nanoparticles were characterized using dynamic light scattering, field emission scanning electron microscopy (FE-SEM) and Fourier transform infrared spectroscopy (FTIR). The toxicity of nanoAMG in fibroblast NIH/3T3 cell line was determined using MTT method. The antibiofilm effect of nanoAMG was determined through the evaluation of biofilm formation by Streptococcus mutans using a 96-well plate. Biofilm biomass was quantified using crystal violet. Cell viability was observed under confocal microscopy using LIVE/DEAD BacLight staining. Moreover, gene expression was determined by quantitative real-time PCR and membrane permeabilization activity by measuring the uptake of o-nitrophenol-β-D-galactoside. Results: NanoAMG size was in a range of 10-50 nm with a polydispersity index of < 0.3 and zeta potential value of -35.2 mV. The size and the incorporation of AMG in the nanoparticles were confirmed by FE-SEM and FTIR analyses. The IC50 values of the test agents on NIH/3T3 cells were (9.80 ± 0.63) μg/mL for AMG and (8.70 ± 0.81) μg/mL for nanoAMG, while no toxicity was generated from excipients used to prepare nanoparticles. In the early stage of biofilm formation, treatment with 6.25 μmol/L nanoAMG caused a reduction in biofilm biomass up to 49.1%, compared to 33.4% for AMG. In contrast, biofilms at the late stage were more resistant to the test agents. At 96 μmol/L (= 10 × MIC), nanoAMG reduced only 20.7% of biofilm biomass while AMG did not show any effect. Expressions of gtfB and gtfC genes involved in biofilm formation were down-regulated 3.3 and 12.5 folds, respectively, compared to AMG (2.4 and 7.6 folds, respectively). LIVE/DEAD BacLight fluorescence staining and microscopy observation indicated that biofilm cells were killed by both nanoAMG andAMG at 48 μmol/L (= 5 × MIC). In addition, membrane permeabilization activity was increased in a time dependent manner and higher in nanoAMG treated cells compared toAMG.Conclusions: AMG coated nanoparticle can enhance AMG bioactivity and can be used as a new and promising antibiofilm agent.

3.
Asian Pacific Journal of Tropical Biomedicine ; (12): 325-332, 2020.
Article in Chinese | WPRIM | ID: wpr-950291

ABSTRACT

Objective: To investigate the antibiofilm activity of alpha- mangostin (AMG) loaded nanoparticle (nanoAMG) against dental caries pathogen Streptococcus mutans. Methods: AMG was isolated from the peels of Garcinia mangostana L. using silica gel columns and chemically analysed by high performance liquid chromatography and nuclear magnetic resonance. NanoAMG was prepared using the solvent evaporation method combined with high-speed homogenization. The nanoparticles were characterized using dynamic light scattering, field emission scanning electron microscopy (FE-SEM) and Fourier transform infrared spectroscopy (FTIR). The toxicity of nanoAMG in fibroblast NIH/3T3 cell line was determined using MTT method. The antibiofilm effect of nanoAMG was determined through the evaluation of biofilm formation by Streptococcus mutans using a 96-well plate. Biofilm biomass was quantified using crystal violet. Cell viability was observed under confocal microscopy using LIVE/DEAD BacLight staining. Moreover, gene expression was determined by quantitative real-time PCR and membrane permeabilization activity by measuring the uptake of o-nitrophenol- β-D-galactoside. Results: NanoAMG size was in a range of 10-50 nm with a polydispersity index of < 0.3 and zeta potential value of -35.2 mV The size and the incorporation of AMG in the nanoparticles were confirmed by FE-SEM and FTIR analyses. The IC

4.
Int J Pharm Pharm Sci ; 2019 Jun; 11(6): 45-49
Article | IMSEAR | ID: sea-205909

ABSTRACT

Objective: This study assessed the effects of alpha-mangostin (AM) and citronella oil (CO) working alone or in combination against Propionibacterium acnes (P. acnes) and Staphylococcus aureus (S. aureus). Methods: The screening for antibacterial activity of AM and CO against P. acnes and S. aureus was carried out using the disk diffusion method. The minimum inhibitory concentrations (MICs) and minimum bactericidal concentrations (MBCs) of these two substances were determined using the broth microdilution method. The fractional inhibitory concentration indices (FICI) of a combination of AM and CO were obtained by checkerboard dilution assay. Results: The results showed that alpha-mangostin and citronella oil do indeed fight against P. acnes and S. aureus. The MICs and MBCs of AM against P. acnes and S. aureus were the same at 6.25 and 50 µg/ml, respectively. Both the MIC and the MBC of CO against P. acnes were 27.81µg/ml. The MIC and the MBC of CO against S. aureus were 112.13 and 224.25 µg/ml, respectively. The FICI of a combination of AM and CO against P. acnes and S. aureus were 2.00, indicating indifferent interaction with no additional inhibitory effect. Conclusion: AM and CO are very effective against P. acnes and S. aureus, nevertheless their effect when used together was indifferent from using alone. Further research may find that either or both of these substances combined with yet a different natural agent could provide synergy againstP. acnes and S. aureus.

5.
Immune Network ; : 253-260, 2012.
Article in English | WPRIM | ID: wpr-20067

ABSTRACT

alpha-Mangostin is a xanthon derivative contained in the fruit hull of mangosteen (Garcinia mangostana L.), and the administration of alpha-Mangostin inhibited the growth of transplanted colon cancer, Her/CT26 cells which expressed Her-2/neu as tumor antigen. Although alpha-Mangostin was reported to have inhibitory activity against sarco/endoplasmic reticulum Ca2+ ATPase like thapsigargin, it showed different activity for autophagy regulation. In the current study, we found that alpha-Mangostin induced autophagy activation in mouse intestinal epithelial cells, as GFP-LC3 transgenic mice were orally administered with 20 mg/kg of alpha-Mangostin daily for three days. However, the activation of autophagy by alpha-Mangostin did not significantly increase OVA-specific T cell proliferation. As we assessed ER stress by using XBP-1 reporter system and phosphorylation of eIF2alpha, thapsigargin-induced ER stress was significantly reduced by alpha-Mangostin. However, coadministration of thapsigargin with alpha-Mangostin completely blocked the antitumor activity of alpha-Mangostin, suggesting ER stress with autophagy blockade accelerated tumor growth in mouse colon cancer model. Thus the antitumor activity of alpha-Mangostin can be ascribable to the autophagy activation rather than ER stress induction.


Subject(s)
Animals , Mice , Autophagy , Calcium-Transporting ATPases , Cell Proliferation , Colonic Neoplasms , Epithelial Cells , Fruit , Garcinia mangostana , Mice, Transgenic , Phosphorylation , Reticulum , Thapsigargin , Transplants , Xanthones
SELECTION OF CITATIONS
SEARCH DETAIL