Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 109
Filter
1.
Int J Biol Macromol ; : 133164, 2024 Jun 13.
Article in English | MEDLINE | ID: mdl-38878919

ABSTRACT

Obesity is a global health challenge that causes metabolic dysregulation and increases the risk of various chronic diseases. The gut microbiome is crucial in modulating host energy metabolism, immunity, and inflammation and is influenced by dietary factors. Gac fruit (Momordica cochinchinensis), widely consumed in Southeast Asia, has been proven to have various biological activities. However, the composition and effect of crude gac aril polysaccharides (GAP) on obesity and gut microbiota disturbed by high-fat diet (HFD) remain to be elucidated. Compositional analysis showed that GAP contains high oligosaccharides, with an average of 7-8 saccharide units. To mimic clinical obesity, mice were first made obese by feeding HFD for eight weeks. GAP intervention was performed from week 9 to week 20 in HFD-fed mice. Our results showed that GAP inhibited body weight gain, eWAT adipocyte hypertrophy, adipokine derangement, and hyperlipidemia in HFD-induced obese mice. GAP improved insulin sensitivity, impaired glucose tolerance, and hepatic steatosis. GAP modulated the gut microbiota composition and reversed the HFD-induced dysbiosis of at least 20 genera. Taken together, GAP improves metabolic health and modulates the gut microbiome to relieve obesity risk factors, demonstrating the potential of dietary GAP for treating obesity-associated disorders.

2.
Food Chem Toxicol ; 172: 113564, 2023 Feb.
Article in English | MEDLINE | ID: mdl-36563924

ABSTRACT

Head and neck squamous cell carcinoma (HNSCC) is a relatively common malignancy, characterized by lethal morbidity. Herein, we attempted to investigate the autophagy/apoptosis activities of the submerged fermented broths of Antrodia salmonea (AS) in HNSCC Twist-overexpressing (OECM-1 and FaDu-Twist) cells. AS (0-150 µg/mL) effectively reduced cell viability, colony formation, and downregulated Twist expression in OECM-1 and FaDu-Twist cells compared to FaDu cells. AS- induced apoptosis was mainly associated with activation of caspase-3, PARP cleavage, increased expression of VDAC-1 and disproportionation of Bax/Bcl-2. Annexin V/PI staining suggested late apoptosis induction by AS treatment. AS exhibits enhanced autophagy process mediated via LC3-I/II accumulation, increased acidic vesicular organelles (AVOs) formation and p62/SQSTM1 expression feeding into the apoptotic program. However, pre-treatment with autophagy blockers 3-MA and CQ significantly diminished AS-induced cell death. Additionally, suppression of AS-induced ROS release by treatment with antioxidant N-acetylcysteine (NAC) resulted in reduction of apoptotic and autophagic cell death. In vivo studies strengthened the above observations and showed that AS effectively reduced the tumor volume and tumor weight in OECM-1-xenografted nude mice. This study discovered that Antrodia salmonea exhibits a novel anti-cancer mechanism which could be harnessed as a new potent drug for HNSCC treatment.


Subject(s)
Apoptosis , Head and Neck Neoplasms , Animals , Mice , Squamous Cell Carcinoma of Head and Neck/drug therapy , Reactive Oxygen Species/metabolism , Mice, Nude , Autophagy , Cell Line, Tumor , Head and Neck Neoplasms/drug therapy
3.
J Agric Food Chem ; 69(48): 14557-14567, 2021 Dec 08.
Article in English | MEDLINE | ID: mdl-34813306

ABSTRACT

Triple negative breast cancer (TNBC) is one of the leading causes of cancer death in the world and lacks an effective targeted therapy. G-protein-coupled receptor 161 (GPR161) has been demonstrated to perform the functional regulations on TNBC progression and might be a potential new target for TNBC therapy. This study showed the effects of bisdemethoxycurcumin (BDMC) on GPR161 regulation, indicating that BDMC effectively inhibited GPR161 expression and downregulated GPR161-driven signaling. BDMC showed the potent inhibitory effects on TNBC proliferation through suppressing GPR161-mediated mammalian target of rapamycin (mTOR)/70 kDa ribosomal protein S6 kinase (p70S6K) activation. Besides, in this study, we discover the mechanism of GPR161-driven TNBC metastasis, linking to GPR161-mediated twist-related protein 1 (Twist1)/matrix metallopeptidase 9 (MMP9) contributing to the epithelial-mesenchymal transition (EMT). BDMC effectively repressed GPR161-mediated TNBC metastasis via inhibiting Twist1/MMP9-induced EMT. The three-dimensional invasion assay also showed that BDMC significantly inhibited TNBC invasion. The combination treatment of BDMC and rapamycin enhanced the inhibition of TNBC proliferation and metastasis through increasing the blockage of mTOR activation. Furthermore, this study also observed that BDMC activated the caspase 3/9 signaling pathway to induce TNBC apoptosis. Therefore, BDMC could be applicable to anticancer therapy, especially targeting on the GPR161-driven cancer type.


Subject(s)
Triple Negative Breast Neoplasms , Apoptosis , Cell Line, Tumor , Cell Movement , Cell Proliferation , Diarylheptanoids , Epithelial-Mesenchymal Transition , Gene Expression Regulation, Neoplastic , Humans , Receptors, G-Protein-Coupled/genetics , Signal Transduction , Sirolimus , TOR Serine-Threonine Kinases/genetics , TOR Serine-Threonine Kinases/metabolism , Triple Negative Breast Neoplasms/drug therapy , Triple Negative Breast Neoplasms/genetics
4.
Environ Toxicol ; 36(12): 2578-2588, 2021 Dec.
Article in English | MEDLINE | ID: mdl-34599545

ABSTRACT

Tumor necrosis factor-related apoptosis-induced ligand (TRAIL) shows little or no toxicity in most normal cells and preferentially induces apoptosis in a variety of malignant cells. However, patients develop resistance to TRAIL, therefore, sensitizing agents that can sensitize the tumor cells to TRAIL-mediated apoptosis are necessary. In this study, we investigated the effect of 2-(3-hydroxyphenyl)-5-methylnaphthyridin-4-one (CSC-3436), an useful flavonoid, to overcome the TRAIL-resistant triple negative breast cancer (TNBC) cells. We found that CSC-3436 potentiated TRAIL-induced apoptosis in TRAIL-resistant TNBC cells and this correlated with the upregulation of death receptors (DR)-5 and down-regulation of decreased decoy receptor (DcR)-1 expression. When examined for its mechanism, we found that the decreased expression of anti-apoptotic proteins c-FLIPS/L, Bcl-Xl, Bcl-2, Survivin, and XIAP. CSC-3436 would increase the expression of Bax and promoted the cleavage of bid. In addition, the induction of DR5 by CSC-3436 was found to be dependent on the modulation of reactive oxygen species (ROS)/p38/C/EBP-homologous protein (CHOP) signaling pathways. Overall, our results indicated that CSC-3436 could potentiate the apoptotic effects of TRAIL through down-regulation of cell survival proteins and upregulation of DR5 via the ROS-mediated upregulation of CHOP protein.


Subject(s)
Receptors, TNF-Related Apoptosis-Inducing Ligand , Triple Negative Breast Neoplasms , Apoptosis , Cell Line, Tumor , Humans , Ligands , Naphthyridines , Reactive Oxygen Species , Receptors, TNF-Related Apoptosis-Inducing Ligand/genetics , TNF-Related Apoptosis-Inducing Ligand , Transcription Factor CHOP/genetics
5.
Environ Toxicol ; 36(11): 2186-2195, 2021 Nov.
Article in English | MEDLINE | ID: mdl-34291863

ABSTRACT

Bladder cancer is the most common malignancy of the urinary tract and arising from the epithelial lining of the urinary bladder. Resistance to cytotoxic therapies is associated with overexpression of oncogenic proteins; including HER2, and Akt in chemotherapy resistance of bladder cancer. Various studies demonstrated that curcuminoids, the most important active phenolic compounds of turmeric (Curcuma longa), have anti-tumor activities in a wide range of human malignant cell lines. The aim of this study is to evaluate whether curcuminoids (curcumin, demethoxycurcumin (DMC), and bisdemethoxycurcumin) could repress the expression of HER2 in HER2-overexpressing bladder cancer cells. Among the test compounds, DMC significantly suppressed the expression of HER2, and preferentially inhibited cell proliferation and induced apoptosis in HER2-overexpressing bladder cancer cells. DMC decreases HER2 level through inhibiting the interaction of HER2 and Hsp90. Our study also indicated that DMC showed additive activity in combination with chemotherapeutic agents, including paclitaxel and cisplatin. These findings show that DMC should be developed further as a new antitumor drug candidate for treatment of HER2-overexpressing bladder cancer.


Subject(s)
Curcumin , Urinary Bladder Neoplasms , Apoptosis , Cell Line, Tumor , Curcumin/pharmacology , Diarylheptanoids , Humans , Phosphatidylinositol 3-Kinases , Proto-Oncogene Proteins c-akt/genetics , Receptor, ErbB-2 , Urinary Bladder Neoplasms/drug therapy
6.
Spectrochim Acta A Mol Biomol Spectrosc ; 262: 120139, 2021 Dec 05.
Article in English | MEDLINE | ID: mdl-34245971

ABSTRACT

A new efficient Schiff base sensor SB3 for fluorescent and colorimetric "naked-eye" "turn-on" sensing of cyanide anion (CN-) with excellent sensitivity and selectivity was developed. The 4,4'-(perfluoropropane-2,2-diyl)bisphenol group and two phenyl groups were covalently linked by two C = N bonds to extend the conjugation length. The four hydroxyl groups can improve the water solubility of the SB3 sensor. The SB3 sensor exhibited high specificity towards CN- by interrupting its intramolecular charge transfer, resulting in a color change and remarkable "turn-on" green fluorescence emission. The sensing mechanism is caused by the nucleophilic addition of CN- toward imine groups of the SB3 sensor, leading to breaks of the conjugation, fluorescent spectral changes, and color change. It was confirmed by 1H NMR titration and Mass spectra. The detection limits for CN- and Al3+obtained by fluorescence spectrum are 0.80 µM and 0.25 µM, respectively. The SB3 sensor can act as an efficient chemical sensor for detecting the CN- and Al3+ ions under common environmental and physiological conditions (pH 5-12). Besides, the sensor can also detect CN- in food materials (such as sprouting potatoes and cassava flour) and imaging CN-in living cells with strong "turn-on" fluorescence at 490 nm. SB3 is an excellent CN- sensor that exhibits some advantages, including easy synthesis, distinct fluorescence and color change, high selectivity, low detection limit, and good anti-interference ability to analyze solution and food samples, together with fluorescence cell imaging.


Subject(s)
Colorimetry , Cyanides , Aluminum , Fluorescent Dyes , Schiff Bases , Spectrometry, Fluorescence
7.
Free Radic Biol Med ; 173: 151-169, 2021 09.
Article in English | MEDLINE | ID: mdl-34314818

ABSTRACT

3-O-ethyl ascorbic acid (EAA) is an ether-derivative of ascorbic acid, known to inhibit tyrosinase activity, and is widely used in skincare formulations. Nevertheless, the molecular mechanisms underlying the EAA's effects are poorly understood. Here, the anti-melanogenic activity of EAA was demonstrated through Nrf2-mediated α-MSH inhibition in UVA-irradiated keratinocytes (HaCaT) and autophagy induction and inhibition of α-MSH-stimulated melanogenesis in melanocytes (B16F10). EAA pretreatment increased the HaCaT cell viability but suppressed ROS-mediated p53/POMC/α-MSH pathways in UVA-irradiated cells. Further, the conditioned medium from EAA-pretreated and UVA-irradiated HaCaT cells suppressed the MITF-CREB-tyrosinase pathways leading to the inhibition of melanin synthesis in B16F10 cells. EAA treatment increased nuclear Nrf2 translocation via the p38, PKC, and ROS pathways leading to HO-1, γ-GCLC, and NQO-1 antioxidant expression in HaCaT cells. However, Nrf2 silencing reduced the EAA-mediated anti-melanogenic activity, evidenced by impaired antioxidant gene expression and uncontrolled ROS (H202) generation following UVA irradiation. In B16F10 cells, EAA-induced autophagy was shown by enhanced LC3-II levels, AVO formation, Beclin-1 upregulation, and activation of p62/SQSTM1. Further, EAA-induced anti-melanogenic activity was substantially decreased in autophagy inhibitor (3-MA) pretreated or LC3 knockdown B16F10 cells. Notably, transmission electron microscopy data showed increased melanosome-engulfing autophagosomes in EAA-treated B16F10 cells. Moreover, EAA also down-regulated MC1R, TRP-1/-2, tyrosinase expressions, and melanin synthesis by suppressing the cAMP-CREB-mediated MITF expression in B16F10 cells stimulated with α-MSH. In vivo studies on the zebrafish model further confirmed that EAA inhibited tyrosinase expression/activity and endogenous pigmentation. In conclusion, 3-O-ethyl ascorbic acid is an effective skin-whitening agent and could be used as a topical agent for cosmetic purposes.


Subject(s)
Melanins , Melanoma, Experimental , Animals , Ascorbic Acid , Autophagy , Cell Line, Tumor , Keratinocytes , Melanocytes , Melanoma, Experimental/drug therapy , NF-E2-Related Factor 2/genetics , Zebrafish , alpha-MSH
8.
In Vivo ; 35(4): 2047-2057, 2021.
Article in English | MEDLINE | ID: mdl-34182480

ABSTRACT

BACKGROUND: Tetrandrine, a bis-benzylisoquinoline alkaloid, induces apoptosis of many types of human cancer cell. Hydrogen peroxide (H2O2) is a reactive oxygen species inducer; however, there are no reports to show whether pre-treatment of tetrandrine with H2O2 induces more cell apoptosis than H2O2 alone. Thus, the present study investigated the effects of tetrandrine on H2O2-induced cell apoptosis of human keratinocytes, HaCaT, in vitro. MATERIALS AND METHODS: HaCaT cells were pre-treated with and without tetrandrine for 1 h, and then treated with H2O2 for examining cell morphological changes and cell viability using contrast-phase microscopy and propidium iodide (PI) exclusion assay, respectively. Cells were measured apoptotic cell death by using annexin V/PI double staining and further analyzed by flow cytometer. Cells were further assessed for DNA condensation using 2-(4-amidinophenyl)-6-indolecarbamidine staining. Western blotting was used to measure expression of apoptosis-associated proteins and confocal laser microscopy was used to measure the protein expression and nuclear translocation from the cytoplasm to nuclei. RESULTS: Pre-treatment of tetrandrine for 1 h and treatment with H2O2 enhanced H2O2-induced cell morphological changes and reduced cell viability, whilst increasing apoptotic cell death and DNA condensation. Furthermore, tetrandrine significantly increased expression of reactive oxygen species-associated proteins such as superoxide dismutase (Cu/Zn) and superoxide dismutase (Mn) but significantly reduced the level of catalase, which was also confirmed by confocal laser microscopy. It also increased expression of DNA repair-associated proteins ataxia telangiectasia mutated, ataxia-telangectasia and Rad3-related, phospho-P53, P53 and phosphorylated histone H2AX, and of pro-apoptotic proteins BCL2 apoptosis regulator-associated X-protein, caspase-3, caspase-8, caspase-9 and poly ADP ribose polymerase in HaCaT cells. CONCLUSION: These are the first and novel findings showing tetrandrine enhances H2O2-induced apoptotic cell death of HaCaT cells and may provide a potent approach for the treatment of proliferated malignant keratinocytes.


Subject(s)
Benzylisoquinolines , Caspases , Apoptosis , Benzylisoquinolines/pharmacology , Caspases/genetics , Cell Survival , Humans , Hydrogen Peroxide , Keratinocytes , Reactive Oxygen Species
9.
In Vivo ; 35(2): 833-843, 2021.
Article in English | MEDLINE | ID: mdl-33622876

ABSTRACT

BACKGROUND/AIM: Hydrogen peroxide (H2O2) is one of the reactive oxygen species (ROS), which can induce apoptotic cell death in numerous cancer cells. Pterostilbene (PTE), a natural polyphenolic compound, induces cell apoptosis in many human cancer cells. MATERIALS AND METHODS: We investigated whether PTE could enhance H2O2-induced cell apoptosis in human keratinocyte HaCaT cells in vitro. The morphological change of HaCaT cells was observed and photographed under a contrast-phase microscope. The percentage of cell viability was measured by propidium iodide exclusion assay. Cell apoptosis was performed by Annexin V/PI double staining and assayed by flow cytometer. DNA condensation was measured by DAPI staining. The protein expression was determined by western blotting. ROS production-associated proteins were also assayed by confocal laser scanning microscopy. RESULTS: PTE pre-treatment enhanced H2O2 (600 µM)-induced cell morphological changes and reduced the total cell number (cell viability). The decreased cell viability in HaCaT cells was through induction of apoptotic cell death, which was confirmed by Annexin V/PI double staining and DAPI staining. Western blotting studies indicated that HaCaT cells which were pre-treated with PTE (100 µM) and then co-treated with H2O2 (600 µM) for 12 h showed significantly increased levels of SOD (Cu/Zn), SOD (Mn), Bax, caspase-3, caspase-8, caspase-9, PARP, p53, p-p53, and p-H2A.X but decreased levels Bcl-2 and catalase. Results also showed that HaCaT cells pre-treated with PTE and then co-treated with H2O2 had increased expression of SOD (Cu/Zn) and glutathione but decreased catalase. CONCLUSION: These observations suggest that PTE pre-treatment can enhance the H2O2-induced apoptotic cell death in keratinocyte cells and may be an effective candidate for the treatment of proliferative keratinocytes.


Subject(s)
Caspases , Hydrogen Peroxide , Apoptosis , Caspase 3/metabolism , Caspases/metabolism , Cell Survival , Humans , Keratinocytes/metabolism , Reactive Oxygen Species , Signal Transduction , Stilbenes
10.
Environ Toxicol ; 35(9): 911-921, 2020 Sep.
Article in English | MEDLINE | ID: mdl-32270916

ABSTRACT

Leukemia is one of the major diseases causing cancer-related deaths in the young population, and its cure rate is unsatisfying with side effects on patients. Fluorouracil (5-FU) is currently used as an anticancer drug for leukemia patients. Casticin, a natural polymethoxyflavone, exerts anticancer activity against many human cancer cell lines in vitro, but no other reports show 5-FU combined with casticin increased the mouse leukemia cell apoptosis in vitro. Herein, the antileukemia activity of 5-FU combined with casticin in WEHI-3 mouse leukemia cells was investigated in vitro. Treatment of two-drug combination had a higher decrease in cell viability and a higher increase in apoptotic cell death, the level of DNA condensation, and the length of comet tail than that of 5-FU or casticin treatment alone in WEHI-3 cells. In addition, the two-drug combination has a greater production rate of reactive oxygen species but a lower level of Ca2+ release and mitochondrial membrane potential (ΔΨm ) than that of 5-FU alone. Combined drugs also induced higher caspase-3 and caspase-8 activities than that of casticin alone and higher caspase-9 activity than that of 5-FU or casticin alone at 48 hours treatment. Furthermore, 5-FU combined with casticin has a higher expression of Cu/Zn superoxide dismutase (SOD [Cu/Zn]) and lower catalase than that of 5-FU or casticin treatment alone. The combined treatment has higher levels of Bax, Endo G, and cytochrome C of proapoptotic proteins than that of casticin alone and induced lower levels of B-cell lymphoma 2 (BCL-2) and BCL-X of antiapoptotic proteins than that of 5-FU or casticin only. Furthermore, the combined treatment had a higher expression of cleaved poly (ADP-ribose) polymerase (PARP) than that of casticin only. Based on these findings, we may suggest that 5-FU combined with casticin treatment increased apoptotic cell death in WEHI-3 mouse leukemia cells that may undergo mitochondria and caspases signaling pathways in vitro.


Subject(s)
Antineoplastic Agents/pharmacology , Apoptosis/drug effects , Flavonoids/pharmacology , Fluorouracil/pharmacology , Animals , Antineoplastic Agents/administration & dosage , Caspases/metabolism , Cell Line, Tumor , Cell Survival/drug effects , Cytochromes c/metabolism , Drug Synergism , Flavonoids/administration & dosage , Fluorouracil/administration & dosage , Humans , Leukemia/pathology , Membrane Potential, Mitochondrial/drug effects , Mice , Mitochondria/drug effects , Mitochondria/metabolism , Reactive Oxygen Species/metabolism , Signal Transduction/drug effects
11.
Polymers (Basel) ; 12(1)2020 Jan 06.
Article in English | MEDLINE | ID: mdl-31935917

ABSTRACT

Here, staple carbon fiber fabric-reinforced polycarbonate (PC)- and epoxy (EP)-based composites with different impregnating resin levels were fabricated using a modified film stacking process. The effects of surface topographies and resin types on the tribological properties of stable carbon fabric composites (sCFC) were investigated. Friction and wear tests on the carbon composites were conducted under unlubricated sliding using a disk-on-disk wear test machine. Experimental results showed that the coefficient of friction (COF) of the sCFC was dominated by matrix type, followed by peak material portion (Smr1) values, and finalized with core height (Sk) values. The COF of composites decreased by increasing the sliding speed and applied pressure. This also relied on surface topography and temperature generated at the worn surface. However, the specific wear rate was strongly affected by resin impregnation. Partially-impregnated composites showed lower specific wear rate, whereas fully-impregnated composites showed a higher wear rate. This substantially increased by increasing the sliding speed and applied pressure. Scanning electron microscopy observations of the worn surfaces revealed that the primary wear mechanisms were abrasion, adhesion, and fatigue for PC-based composites. For EP-based composites, this was primarily abrasion and fatigue. Results proved that partially-impregnated composites exhibited better tribological properties under severe conditions.

12.
Molecules ; 25(2)2020 Jan 15.
Article in English | MEDLINE | ID: mdl-31952105

ABSTRACT

Casticin was obtained from natural plants, and it has been shown to exert biological functions; however, no report concerns the induction of DNA damage and repair in human lung cancer cells. The objective of this study was to investigate the effects and molecular mechanism of casticin on DNA damage and repair in human lung cancer A549 cells. Cell viability was determined by flow cytometric assay. The DNA damage was evaluated by 4',6-diamidino-2-phenylindole (DAPI) staining and electrophoresis which included comet assay and DNA gel electrophoresis. The protein levels associated with DNA damage and repair were analyzed by western blotting. The expression and translocation of p-H2A.X were observed by confocal laser microscopy. Casticin reduced total viable cell number and induced DNA condensation, fragmentation, and damage in A549 cells. Furthermore, casticin increased p-ATM at 6 h and increased p-ATR and BRCA1 at 6-24 h treatment but decreased p-ATM at 24-48 h, as well as decreased p-ATR and BRCA1 at 48 h. Furthermore, casticin decreased p-p53 at 6-24 h but increased at 48 h. Casticin increased p-H2A.X and MDC1 at 6-48 h treatment. In addition, casticin increased PARP (cleavage) at 6, 24, and 48 h treatment, DNA-PKcs and MGMT at 48 h in A549 cells. Casticin induced the expressions and nuclear translocation of p-H2AX in A549 cells by confocal laser microscopy. Casticin reduced cell number through DNA damage and condensation in human lung cancer A549 cells.


Subject(s)
Apoptosis , DNA Damage , DNA Repair , Flavonoids/pharmacology , Gene Expression Regulation, Neoplastic/drug effects , Lung Neoplasms/pathology , Neoplasm Proteins/metabolism , A549 Cells , Cell Survival , Histones/metabolism , Humans , Lung Neoplasms/drug therapy , Lung Neoplasms/genetics , Lung Neoplasms/metabolism
13.
J Biol Eng ; 13: 86, 2019.
Article in English | MEDLINE | ID: mdl-31754373

ABSTRACT

BACKGROUND: Large gap healing is a difficult issue in the recovery of peripheral nerve injury. The present study provides in vivo trials of silicone rubber chambers filled with collagen containing IFN-γ or IL-4 to bridge a 15 mm sciatic nerve defect in rats. Fillings of NGF and normal saline were used as the positive and negative controls. Neuronal electrophysiology, neuronal connectivity, macrophage infiltration, location and expression levels of calcitonin gene-related peptide and histology of the regenerated nerves were evaluated. RESULTS: At the end of 6 weeks, animals from the groups of NGF and IL-4 had dramatic higher rates of successful regeneration (100 and 80%) across the wide gap as compared to the groups of IFN-γ and saline controls (30 and 40%). In addition, the NGF group had significantly higher NCV and shorter latency compared to IFN-γ group (P < 0.05). The IL-4 group recruited significantly more macrophages in the nerves as compared to the saline controls and the NGF-treated animals (P < 0.05). CONCLUSIONS: The current study demonstrated that NGF and IL-4 show potential growth-promoting capability for peripheral nerve regeneration. These fillings in the bridging conduits may modulate local inflammatory conditions affecting recovery of the nerves.

14.
15.
In Vivo ; 33(4): 1193-1201, 2019.
Article in English | MEDLINE | ID: mdl-31280209

ABSTRACT

BACKGROUND/AIM: Our current study aimed to determine the molecular mechanisms of citronellol-induced cell death and ROS accumulation in non-small cell lung cancer (NCI-H1299 cells) and also compare the anticancer effects of citronellol and EOPC. MATERIALS AND METHODS: ROS measurement and western blotting were performed to detect whether citronellol can induce necroptosis in vitro. Besides, we performed an in vivo analysis of tumourigenesis inhibition by citronellol treatment in BALB/c (nu/nu) nude mice. RESULTS: Necroptosis occured by up-regulating TNF-α, RIP1/RIP3 activities, and down-regulating caspase-3/caspase-8 activities after citronellol treatment in NCI-H1299 cells. Citronellol also resulted in a biphasic increase in ROS production at 1 h and at 12 h in NCI-H1299 cells. Xenograft model experiments showed that citronellol could effectively inhibit subcutaneous tumours produced 4 weeks after intraperitoneal injection of NCI-H1299 in BALB/c nude mice. CONCLUSION: Citronellol induced necroptosis of NCI-H1299 cells via TNF-α pathway and ROS accumulation.


Subject(s)
Acyclic Monoterpenes/pharmacology , Lung Neoplasms/metabolism , Necroptosis/drug effects , Reactive Oxygen Species/metabolism , Signal Transduction/drug effects , Tumor Necrosis Factor-alpha/metabolism , Animals , Cell Line, Tumor , Disease Models, Animal , Dose-Response Relationship, Drug , Humans , Lung Neoplasms/drug therapy , Lung Neoplasms/pathology , Mice , Xenograft Model Antitumor Assays
16.
Am J Chin Med ; 47(3): 657-674, 2019.
Article in English | MEDLINE | ID: mdl-30974966

ABSTRACT

Glioblastoma (GBM) is the most commonly occurring tumor in the cerebral hemispheres. Currently, temozolomide (TMZ), an alkylating agent that induces DNA strand breaks, is considered the frontline chemotherapeutic agent for GBM. Despite its frontline status, GBM patients commonly exhibit resistance to TMZ treatment. We have recently established and characterized TMZ-resistant human glioma cells. The aim of this study is to investigate whether curcumin modulates cell apoptosis through the alternation of the connexin 43 (Cx43) protein level in TMZ-resistant GBM. Overexpression of Cx43, but not ATP-binding cassette transporters (ABC transporters), was observed (approximately 2.2-fold) in TMZ-resistant GBM cells compared to the Cx43 levels in parental GBM cells. Furthermore, at a concentration of 10 µ M, curcumin significantly reduced Cx43 protein expression by about 40%. In addition, curcumin did not affect the expression of other connexins like Cx26 or epithelial-to-mesenchymal transition (EMT) proteins such as ß -catenin or α E-catenin. Curcumin treatment led to an increase in TMZ-induced cell apoptosis from 4% to 8%. Importantly, it did not affect the mRNA expression level of Cx43. Concomitant treatment with the translation inhibitor cycloheximide (CHX) exerted additional effects on Cx43 degradation. Treatment with the autophagy inhibitor 3-MA (methyladenine) did not affect the curcumin-induced Cx43 degradation. Interestingly, treatment with the proteasome inhibitor MG132 (carbobenzoxy-Leu-Leu-leucinal) significantly negated the curcumin-induced Cx43 degradation, which suggests that curcumin-induced Cx43 degradation occurs through the ubiquitin-proteasome pathway.


Subject(s)
Antineoplastic Agents, Alkylating/pharmacology , Apoptosis/drug effects , Connexin 43/metabolism , Curcumin/pharmacology , Glioblastoma/genetics , Glioblastoma/pathology , Proteolysis/drug effects , Temozolomide/pharmacology , Humans , Stimulation, Chemical , Tumor Cells, Cultured
17.
J Cell Physiol ; 234(6): 9118-9129, 2019 06.
Article in English | MEDLINE | ID: mdl-30341909

ABSTRACT

Head and neck squamous cell carcinoma (HNSCC) is one of the leading causes of cancer deaths worldwide, especially in male. With poor prognosis, significant portions of patients with HNSCC die due to cancer recurrence and tumor metastasis after chemotherapy and targeted therapies. The HNSCC FaDu cell ectopic expression of Twist, a key transcriptional factor of epithelial-mesenchymal transition (EMT), which triggers EMT and results in the acquisition of a mesenchymal phenotype, was used as the cell model. Our results demonstrated that treatment with newly synthesized 2-(3-hydroxyphenyl)-5-methylnaphthyridin-4-one (CSC-3436), a flavonoid derivative, elicited changes in its cell morphology, upregulated E-cadherin messenger RNA and protein expression, downregulated N-cadherin, vimentin, and CD133 (a marker associated with tumor-initiating cells) in FaDu-pCDH-Twist cells. Moreover, CSC-3436 exposure reduced B cell-specific Moloney murine leukemia virus integration site 1 (Bmi1) expression regulated by Twist and further suppressed the direct co-regulation of E-cadherin by Twist and Bmi1. Interestingly, CSC-3436 reduced EMT, cancer stemness, and migration/invasion abilities through the inhibition of the Twist/Bmi1-Akt/ß-catenin pathway. Most importantly, our findings provided new evidence that CSC-3436 played a crucial role in therapeutic targeting to Bmi1 and its molecular pathway in HNSCC, and it will be valuable in prognostic prediction and treatment.


Subject(s)
Antineoplastic Agents/pharmacology , Epithelial-Mesenchymal Transition/drug effects , Head and Neck Neoplasms/drug therapy , Naphthyridines/pharmacology , Nuclear Proteins/metabolism , Polycomb Repressive Complex 1/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Squamous Cell Carcinoma of Head and Neck/drug therapy , Twist-Related Protein 1/metabolism , Animals , Antigens, CD/genetics , Antigens, CD/metabolism , Cadherins/genetics , Cadherins/metabolism , Cell Line, Tumor , Cell Movement/drug effects , Female , Gene Expression Regulation, Neoplastic , Head and Neck Neoplasms/enzymology , Head and Neck Neoplasms/genetics , Head and Neck Neoplasms/pathology , Humans , Mice, Inbred NOD , Mice, SCID , Neoplasm Invasiveness , Nuclear Proteins/genetics , Polycomb Repressive Complex 1/genetics , Signal Transduction , Squamous Cell Carcinoma of Head and Neck/enzymology , Squamous Cell Carcinoma of Head and Neck/genetics , Squamous Cell Carcinoma of Head and Neck/secondary , Twist-Related Protein 1/genetics , Xenograft Model Antitumor Assays
18.
Environ Toxicol ; 34(4): 364-374, 2019 Apr.
Article in English | MEDLINE | ID: mdl-30549224

ABSTRACT

Tetrandrine (TET) has been reported to induce anti-cancer activity in many human cancer cells and also to inhibit cancer cell migration and invasion. However, there are no reports to show TET inhibits cell migration and invasion in human brain glioblastoma multiforme GBM 8401 cells. In this study, we investigated the anti-metastasis effects of TET on GBM 8401 cells in vitro. Under sub-lethal concentrations (from 1, 5 up to 10 µM), TET significantly inhibited cell mobility, migration and invasion of GBM 8401 cells that were assayed by wound healing and Transwell assays. Gelatin zymography assay showed that TET inhibited MMP-2 activity in GBM 8401 cells. Western blotting results indicated that TET inhibited several key metastasis-related proteins, such as p-EGFR(Tyr1068) , SOS-1, GRB2, Ras, p-AKT(Ser473) and p-AKT(Thr308) , NF-κB-p65, Snail, E-cadherin, N-cadherin, NF-κB, MMP-2 and MMP-9 that were significant reduction at 24 and 48 hours treatment by TET. TET reduced MAPK signaling associated proteins such as p-JNK1/2 and p-c-Jun in GBM 8401 cells. The electrophoretic mobility shift (EMSA) assay was used to investigate NF-κB and DNA binding was reduced by TET in a dose-dependently. Based on these findings, we suggested that TET could be used in anti-metastasis of human brain glioblastoma multiforme GBM 8401 cells in the future.


Subject(s)
Anticarcinogenic Agents/pharmacology , Benzylisoquinolines/pharmacology , Brain Neoplasms/pathology , Cell Movement/drug effects , Glioblastoma/pathology , Brain Neoplasms/metabolism , Cell Line, Tumor , Glioblastoma/metabolism , Humans , Matrix Metalloproteinase 2/metabolism , Matrix Metalloproteinase 9/metabolism , NF-kappa B/metabolism , Neoplasm Invasiveness , Signal Transduction
19.
Food Chem Toxicol ; 124: 219-230, 2019 Feb.
Article in English | MEDLINE | ID: mdl-30529123

ABSTRACT

Antrodia salonea (AS), a fungus that is indigenous to Taiwan has been well known for its anti-cancer properties. We investigated the anti-metastatic and anti-epithelial-mesenchymal transition (EMT) properties of AS in TNBC cells. To determine their EMT and metastasis levels, in vitro wound healing, wound invasion, Western blotting, RT-PCR, luciferase activity and immunofluorescence assays were performed, while the in vivo anti-metastatic efficacy of AS was evaluated in BALB/c-nu mice through bioluminescence imaging, HE staining, and immunohistochemical staining. MDA-MB-231 cells, when treated with AS concentrations (25-100 µg/mL) resulted in significant reduction of invasion and migration as well as the downregulation of VEGF, uPAR, uPA and MMP-9 (inhibition of PI3K/AKT/NFκB pathways). AS treatment prevented morphological changes and reversed EMT through the upregulation of E-cadherin and the downregulation of N-cadherin, Slug, Twist, and Vimentin. Inhibition of Smad3 signaling pathway, downregulation of ß-catenin pathway and upregulation of GSK3ß expression were also observed while, suppression of metastasis and EMT in TGF-ß1-stimulated non-tumorigenic MCF-10A cells was observed when treated with AS. Histological analysis confirmed that AS reduced tumor metastasis and upregulated E-cadherin expression in biopsied lung tissues. Our results indicated that AS exhibits anti-EMT and anti-metastatic activity, that could contribute to develop anticancer drugs against TNBC.


Subject(s)
Antrodia/chemistry , Biological Products/therapeutic use , Epithelial-Mesenchymal Transition/drug effects , Neoplasm Invasiveness/prevention & control , Triple Negative Breast Neoplasms/drug therapy , Animals , Antigens, CD/genetics , Cadherins/genetics , Cell Line, Tumor , Cell Movement/drug effects , Down-Regulation , Female , Fruiting Bodies, Fungal/chemistry , Humans , Hyphae/chemistry , Lung Neoplasms/secondary , Mice, Inbred BALB C , Transcription Factor RelA/metabolism , Triple Negative Breast Neoplasms/pathology , Up-Regulation , Wnt Signaling Pathway/drug effects , beta Catenin/metabolism
20.
Environ Toxicol ; 34(4): 434-442, 2019 Apr.
Article in English | MEDLINE | ID: mdl-30578657

ABSTRACT

Numerous evidences have shown that chrysin induced cytotoxic effects via induced cell cycle arrest and induction of cell apoptosis in human cancer cell lines, however, no information showed that chrysin inhibited skin cancer cell migration and invasion. In this study, we investigated anti-metastasis mechanisms of chrysin in human melanoma cancer A375.S2 cells in vitro. Under sub-lethal concentrations of chrysin (0, 5, 10, and 15 µM) which inhibits cell mobility, migration and invasion of A375.S2 cells that were assayed by wound healing and Transwell filter. That chrysin inhibited MMP-2 activity in A375.S2 cells was investigated by gelatin zymography assay. Western blotting was used to examine protein expression and results indicated that chrysin inhibited the expression of GRB2, SOS-1, PKC, p-AKT (Thr308), NF-κBp65, and NF-κBp50 at 24 and 48 hours treatment, but only at 10-15 µM of chrysin decreased Ras, PI3K, p-c-Jun, and Snail only at 48 hours treatment and only decrease p-AKT(Ser473) at 24 hours treatment. Furthermore, chrysin (5-15 µM) decreased the expression of uPA, N-cadherin and MMP-1 at 24 and 48 hours treatment but only decreased MMP-2 and VEGF at 48 hours treatment at 10-15 µM and 5-15 µM of chrysin, respectively, however, increased E-cadherin at 5-15 µM treatment. Results of confocal laser microscopy systems indicated that chrysin inhibited expression of NF-κBp65 in A375.S2 cells. Based on these observations, we suggest that chrysin can be used in anti-metastasis of human melanoma cells in the future.


Subject(s)
Cell Movement/drug effects , Flavonoids/pharmacology , MAP Kinase Signaling System/drug effects , Melanoma/pathology , NF-kappa B/metabolism , Skin Neoplasms/pathology , Apoptosis/drug effects , Cell Culture Techniques , Cell Line, Tumor , Cell Survival/drug effects , Humans , Matrix Metalloproteinase 2/metabolism , Melanoma/metabolism , Neoplasm Invasiveness , Skin Neoplasms/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...