Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 24
Filter
Add more filters










Publication year range
1.
Chem Biol Interact ; 363: 110003, 2022 Aug 25.
Article in English | MEDLINE | ID: mdl-35661739

ABSTRACT

Excessive occupational, medical, and environmental exposure of zinc oxide nanoparticles (ZnONPs) caused its accumulation in the nervous system and raised global concerns over its detrimental effects. However, very few researches had been conducted on the impact of mitochondrial quality control process on central nervous system (CNS) after ZnONPs administration, including mitochondrial fission, fusion, biogenesis, and autophagy. In present study, mitochondrial dysfunction and apoptosis were triggered in ZnONPs-exposed human neuroblastoma SH-SY5Y cells. Upregulation of mitochondrial biogenesis regulator (PGC-1α) and fission proteins (Drp1) and downregulation of fusion proteins (OPA1 and Mfn2) were observed in 3 and 6 µg/mL ZnONPs-treated cells. Meanwhile, loss of mitochondrial dynamics and biogenesis was observed in the severe impaired cells (treated with 12 µg/mL ZnONPs). More, autophagy and mitophagy were significantly activated in ZnONPs-treated cells. The increased Beclin1 and LC3 II proteins, decreases of p62 protein, and activated PINK1/Parkin signaling were quantified. The autophagy agonist (Rapamycin), inhibitor (3-MA), and mitophagy inhibitor (Cyclosporine A, CsA) were employed to verify the roles of autophagy and mitophagy in ZnONPs-treated cells. Consequently, mitochondrial dysfunction and apoptosis were aggravated by the blockage of autophagy and mitophagy. Our research could be used to evaluate the risk assessment of ZnONPs exposure in CNS neurons so as to provide a crucial guideline for their future biological applications.


Subject(s)
Nanoparticles , Neuroblastoma , Zinc Oxide , Autophagy , Humans , Mitochondria/metabolism , Nanoparticles/toxicity , Neuroblastoma/metabolism , Zinc Oxide/pharmacology
2.
J Trace Elem Med Biol ; 73: 127006, 2022 Sep.
Article in English | MEDLINE | ID: mdl-35660560

ABSTRACT

BACKGROUND: Astragalus Polysaccharides (APS) had been reported to exhibit antitumor activities. Given that nanoparticles possessed unique advantages in cancer treatment, APS was used as the modifier to prepare gold, silver and selenium nanoparticles (APS-Au, APS-Ag and APS-Se NPs) in the present study. METHODS: The three nanoparticles were synthesized via a green approach and characterized by DLS, TEM, XRD, FT-IR and UV-Vis. The inhibitory effects of these nanoparticles on various tumor cells proliferation were examined by MTT assay in vitro. Reactive oxygen species (ROS), mitochondrial membrane potential (MMP) and the expression of apoptosis and autophagy-related proteins were also detected. RESULTS: Among these, APS-Se NPs displayed the most potent antitumor activities against MCF-7 cells in vitro. Flow cytometric analysis suggested that after cells were exposed to elevated concentrations of APS-Se NPs (10, 20 and 40 µmol/L), the rate of apoptosis was increasing (16.63 ± 0.89, 38.60 ± 3.46 and 44.38 ± 2.62%, respectively). Further analysis by immunofluorescence revealed an increase in intracellular ROS and a loss of MMP. This was accompanied by increased LC3-I to LC3-II conversion. Also, western blot analysis demonstrated that the ratios of Bax/Bcl-2 and cleaved caspase9/caspase 9 rose, and LC3-II and p62 protein levels increased. The addition of chloroquine, an inhibitor of autophagy, further enhanced protein expression of p62 and LC3-II. CONCLUSION: APS-Se NPs exerted their cytotoxic activity in MCF-7 cells by blocking autophagy and facilitating mitochondrial pathway-mediated apoptosis.


Subject(s)
Astragalus Plant , Nanoparticles , Selenium , Apoptosis , Astragalus Plant/metabolism , Autophagy , Humans , MCF-7 Cells , Polysaccharides/pharmacology , Reactive Oxygen Species/metabolism , Selenium/pharmacology , Spectroscopy, Fourier Transform Infrared
3.
Chem Biol Drug Des ; 97(2): 200-209, 2021 02.
Article in English | MEDLINE | ID: mdl-32915501

ABSTRACT

In an effort to discover new agents with high anti-inflammatory activity, 22 new 4-sulfonyloxy/alkoxy benzoxazolone derivatives were synthesized, characterized, and evaluated for their anti-inflammatory activities against lipopolysaccharide (LPS)-induced nitric oxide (NO) production and TNF-α expression in RAW 264.7 cells in vitro. Most of these compounds displayed greater inhibitory ability against NO production than the lead compound 4-o-methyl-benzenesulfonyl benzoxazolone, and the most active compound 2h exhibited the strongest inhibitory activity against NO, IL-1ß, and IL-6 production with IC50 values 17.67, 20.07, and 8.61 µΜ, respectively. The effects of 2h were comparable or stronger than those of the positive control celecoxib. Compound 2h also displayed higher activity in vivo than celecoxib in a mouse model of xylene-induced ear edema, based on their inhibitory rates of 42.69% and 30.87%, respectively. Further molecular analysis revealed that compound 2h significantly reduced the iNOS levels in cell supernatant and suppressed the protein expression of iNOS, p-p38, p-ERK, and nuclear NF-κB. The results indicated that the anti-inflammatory effect of 2h might be realized through the regulation of ERK- and p38-mediated mitogen-activated protein kinase (MAPK)-NF-κB/iNOS signaling, thereby reducing the excessive release of NO, IL-1ß, and IL-6. Our findings demonstrated that compound 2h, a new benzoxazolone derivative, could inhibit activation of the MAPK-NF-κB/iNOS pathway, supporting its potential as a novel anti-inflammatory agent.


Subject(s)
Anti-Inflammatory Agents/pharmacology , Benzoxazoles/chemistry , Signal Transduction/drug effects , Animals , Anti-Inflammatory Agents/chemical synthesis , Anti-Inflammatory Agents/therapeutic use , Benzoxazoles/pharmacology , Benzoxazoles/therapeutic use , Binding Sites , Down-Regulation/drug effects , Edema/chemically induced , Edema/drug therapy , Extracellular Signal-Regulated MAP Kinases/metabolism , Lipopolysaccharides/pharmacology , Macrophages/cytology , Macrophages/drug effects , Macrophages/metabolism , Mice , Molecular Docking Simulation , NF-kappa B/metabolism , Nitric Oxide/metabolism , Nitric Oxide Synthase Type II/chemistry , Nitric Oxide Synthase Type II/metabolism , RAW 264.7 Cells , Structure-Activity Relationship , Tumor Necrosis Factor-alpha/metabolism , p38 Mitogen-Activated Protein Kinases/metabolism
4.
Mol Cell Biochem ; 475(1-2): 249-260, 2020 Dec.
Article in English | MEDLINE | ID: mdl-32840737

ABSTRACT

Farrerol, a dihydroflavone isolated from Rhododendron dauricum L., can inhibit vascular smooth muscle cell (VSMC) proliferation and exert a protective effect on H2O2-induced vascular endothelial cells injury. In this study, we investigated the effects of farrerol on VSMC phenotypic modulation and balloon injury-induced vascular neointimal formation and explored the underlying mechanisms. Serum-starved rat thoracic aorta SMCs (RASMCs) were first pretreated with farrerol (3, 10, and 30 µM, respectively), U0126 (a MEK kinase inhibitor), and SB203580 (a p38 kinase inhibitor), and followed by treatment with serum (10% FBS). The expression of several VSMC-specific markers, including α-SMA, SM22α, and OPN, were analyzed by western blot. Phosphorylation of extracellular signal-regulated protein kinase 1/2 (ERK 1/2) and p38 mitogen-activated protein kinase (MAPK) was also investigated. Farrerol inhibited the serum-induced transition of RASMCs from the contractile to the synthetic phenotype, and this was associated with a decrease in α-SMA and SM22α expression, and an increase in OPN expression. Farrerol also inhibited serum-induced phosphorylation of ERK1/2 and p38MAPK in RASMCs. Moreover, U0126 and SB203580 both inhibited the serum-induced phenotypic transition of RASMCs. These findings indicate that farrerol can maintain the contractile phenotype of VSMCs partly via inactivating the ERK1/2 and p38 MAPK signaling pathways. Using a rat model of carotid artery balloon injury, inhibition of VSMC phenotypic transition and suppression of neointimal formation were confirmed in vivo following the perivascular application of farrerol. Our results suggested that farrerol could be a promising lead compound for the treatment of vascular proliferative diseases.


Subject(s)
Chromones/pharmacology , Mitogen-Activated Protein Kinase 1/antagonists & inhibitors , Mitogen-Activated Protein Kinase 3/antagonists & inhibitors , Muscle, Smooth, Vascular/drug effects , Vascular System Injuries/drug therapy , p38 Mitogen-Activated Protein Kinases/antagonists & inhibitors , Animals , Aorta/drug effects , Aorta/metabolism , Cells, Cultured , Disease Models, Animal , Male , Muscle Contraction/drug effects , Muscle, Smooth, Vascular/metabolism , Neointima/metabolism , Phosphorylation , Phytochemicals/pharmacology , Rats , Rats, Sprague-Dawley , Signal Transduction , Vascular System Injuries/metabolism , Vascular System Injuries/pathology
5.
Oxid Med Cell Longev ; 2020: 1364147, 2020.
Article in English | MEDLINE | ID: mdl-32724488

ABSTRACT

Astragalus membranaceus (AM), used in traditional Chinese medicine, has been shown to enhance immune functions, and recently, its anti-inflammatory effects were identified. However, the mechanisms of action remain unclear. Most studies have shown that autophagy might be involved in the immune response of the body, including inflammation. Here, we developed an inflammatory model by stimulating macrophages with lipopolysaccharides (LPS) to explore the anti-inflammatory effect and mechanisms of AM injection from the perspective of the regulation of autophagy. Immunoblot, immunofluorescence, and ELISA were used to determine the effects of AM injection on the production of interleukin-6 (IL-6) and alterations of autophagy markers. It was found that AM injection reduced the expression of IL-6 in LPS-stimulated macrophages and reversed the LPS-induced inhibition of cellular autophagy. After treatment with inhibitors of signaling pathways, it was shown that LPS downregulated autophagy and upregulated the production of IL-6 in macrophages via the protein kinase B (Akt)/mammalian target of rapamycin (mTOR) pathway. AM injection reversed the effects of LPS by activating the AMP-activated protein kinase (AMPK) instead of inhibiting Akt. These results were further confirmed by testing activators and siRNA silencing of AMPK. Hence, these 2 distinct signaling molecules appear to exert opposite effects on mTOR, which integrates information from multiple upstream signaling pathways, negatively regulating autophagy. In addition, we demonstrated that autophagy might play a key role in regulating the production of IL-6 by testing activators and inhibitors of autophagy and siRNA silencing of ATG5. These findings showed that AM injection might enhance autophagy by activating AMPK and might further play a repressive effect on the LPS-stimulated expression of IL-6. This study explored the relationship between autophagy, signaling pathways, and the production of inflammatory factors in a model of endotoxin infection and treatment with AM injection.


Subject(s)
AMP-Activated Protein Kinases/metabolism , Astragalus Plant/chemistry , Interleukin-6/metabolism , Lipopolysaccharides/metabolism , Macrophages/metabolism , Medicine, Chinese Traditional/methods , Animals , Injections , Mice , Transfection
6.
Oxid Med Cell Longev ; 2020: 5967434, 2020.
Article in English | MEDLINE | ID: mdl-32082480

ABSTRACT

Oxidative stress-mediated endothelial injury is considered to be involved in the pathogenesis of various cardiovascular diseases. Farrerol, a typical natural flavanone from the medicinal plant Rhododendron dauricum L., has been reported to show protective effects against oxidative stress-induced endothelial injuries in our previous study. However, its action molecular mechanisms and targets are still unclear. In the present study, we determined whether farrerol can interact with glycogen synthase kinase 3ß- (GSK-3ß-) nuclear factor erythroid 2-related factor 2- (Nrf2-) antioxidant response element (ARE) signaling, which is critical in defense against oxidative stress. Our results demonstrated that farrerol could specifically target Nrf2 negative regulator GSK-3ß and inhibit its kinase activity. Mechanistic studies proved that farrerol could induce an inhibitory phosphorylation of GSK-3ß at Ser9 without affecting the expression level of total GSK-3ß protein and promote the nuclear translocation of Nrf2 as well as the mRNA and protein expression of its downstream target genes heme oxygenase-1 (HO-1) and NAD(P)H: quinone oxidoreductase 1 (NQO1) in EA.hy926 cells. Further studies performed with GSK-3ß siRNA and specific inhibitor lithium chloride (LiCl) confirmed that GSK-3ß inhibition was involved in farrerol-mediated endothelial protection and Nrf2 signaling activation. Moreover, molecular docking and molecular dynamics studies revealed that farrerol could bind to the ATP pocket of GSK-3ß, which is consistent with the ATP-competitive kinetic behavior. Collectively, our results firstly demonstrate that farrerol could attenuate endothelial oxidative stress by specifically targeting GSK-3ß and further activating the Nrf2-ARE signaling pathway.


Subject(s)
Antioxidant Response Elements/genetics , Chromones/pharmacology , Endothelial Cells/drug effects , Glycogen Synthase Kinase 3 beta/antagonists & inhibitors , NF-E2 Transcription Factor/metabolism , Oxidative Stress/drug effects , Signal Transduction/drug effects , Antioxidants/pharmacology , Cell Line , Cell Nucleus/metabolism , Cell Survival/drug effects , Cell Survival/genetics , Chromones/chemistry , Endothelial Cells/enzymology , Endothelial Cells/metabolism , Endothelium/drug effects , Endothelium/enzymology , Endothelium/metabolism , Glycogen Synthase Kinase 3 beta/chemistry , Glycogen Synthase Kinase 3 beta/genetics , Glycogen Synthase Kinase 3 beta/metabolism , Heme Oxygenase-1/metabolism , Humans , Kinetics , Lithium Chloride/pharmacology , Molecular Docking Simulation , NAD(P)H Dehydrogenase (Quinone)/metabolism , NF-E2 Transcription Factor/genetics , Oxidative Stress/genetics , Phosphorylation , RNA, Small Interfering , Signal Transduction/genetics
8.
Int J Biol Macromol ; 137: 829-835, 2019 Sep 15.
Article in English | MEDLINE | ID: mdl-31284007

ABSTRACT

Selenium nanoparticles (SeNPs) have been attracting increasing attention as potential cancer therapeutic agents. In the present study, laminarin polysaccharides (LP) decorated selenium nanoparticles (LP-SeNPs) with an average diameter of ca. 60 nm were synthesized. Transmission electron microscope (TEM), laser particle analyzer, UV-visible spectrometer and Energy dispersive X-ray (EDX) spectrometer were applied to characterize the prepared SeNPs. The cytotoxicity, apoptosis, and autophagy were examined using a series of cellular assays. The results revealed that LP-SeNPs exhibited cytotoxicity against HepG2 cells with IC50 value was 23.4 ±â€¯2.7 µM. After cells were treated with various concentrations of LP-SeNPs (10, 20 and 40 µM) for 24h, the total apoptosis rate increased to 17.4 ±â€¯1.6, 20.9 ±â€¯1.3 and 30.9 ±â€¯1.2%, respectively. Additionally, treatment of LP-SeNPs increased the expression of Bax and cleaved caspase-9 but decreased the level of Bcl-2. This suggested that LP-SeNPs induced mitochondria-mediated apoptosis. Further, exposure of cells to LP-SeNPs for 12 h induced the upregulation of LC3-II and p62. Treatment of chloroquine (CQ), the inhibitors of the autophagosome, resulted in further accumulation of p62 and LC3-II. These results demonstrated that LP-SeNPs induced the activation of early autophagy, but blocked the late phase of autophagy. Inhibition of late phase of autophagy resulted in the damaged organelles cannot be cleared and aggravating apoptosis. In conclusion, these results indicated that LP-SeNPs exerted its cytotoxicity in HepG2 cells by inhibiting autophagy and inducing apoptosis.


Subject(s)
Apoptosis/drug effects , Autophagy/drug effects , Glucans/chemistry , Glucans/pharmacology , Nanoparticles/chemistry , Selenium/chemistry , Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacology , Hep G2 Cells , Humans , Signal Transduction/drug effects , Solutions
9.
Pharm Biol ; 56(1): 528-534, 2018 Dec.
Article in English | MEDLINE | ID: mdl-30387372

ABSTRACT

CONTEXT: Selenium nanoparticles (SeNPs) have attracted worldwide attention due to their unique properties and potential bioactivities. Considering that hawthorn is both a traditional medicine and a common edible food, hawthorn fruit extract (HE) was chosen as a reductant to prepare SeNPs. OBJECTIVE: SeNPs were synthesized by using an aqueous HE as a reductant and stabilizer. The antitumor activities and potential mechanisms of SeNPs were explored by using a series of cellular assays. MATERIALS AND METHODS: The HE mediated SeNPs (HE-SeNPs) were examined using various characterisation methods. The cytotoxicity was measured against HepG2 cells after treated with 0, 5, 10 and 20 µg/mL of HE-SeNPs for 24 h. Annexin V-FITC/PI staining analysis was performed to observe the apoptosis of HepG2 cells. Additionally, mitochondrial membrane potential (MMP), intracellular reactive oxygen species (ROS) levels were evaluated. Finally, the protein expression levels of caspase-9 and Bcl-2 were identified by Western blot. RESULTS: The mono-dispersed and stable SeNPs were prepared with an average size of 113 nm. HE-SeNPs showed obvious antitumor activities towards HepG2 cells with an IC50 of 19.22 ± 5.3 µg/mL. Results from flow cytometry revealed that both early and total apoptosis rates increased after treating with HE-SeNPs. After cells were treated with various concentrations of HE-SeNPs (5, 10 and 20 µg/mL) for 24 h, the total rate increased to 7.3 ± 0.5, 9.7 ± 1.7 and 19.2 ± 1.6%, respectively. Meanwhile, treatment of HE-SeNPs up-regulated intracellular ROS levels and reduced the MMP. In addition, HE-SeNPs induced the up-regulation of caspase-9 and down-regulation of Bcl-2. DISCUSSION AND CONCLUSIONS: HE-SeNPs induced intracellular oxidative stress and mitochondrial dysfunction to initiate HepG2 cell apoptosis through the mitochondrial pathway. Therefore, HE-SeNPs may be a candidate for further evaluation as a chemotherapeutic agent for human liver cancer.


Subject(s)
Apoptosis/drug effects , Crataegus/chemistry , Nanoparticles/chemistry , Selenium/chemistry , Selenium/pharmacology , Caspase 9/metabolism , Cell Line , Cell Survival/drug effects , Fruit/chemistry , Green Chemistry Technology , Hep G2 Cells , Humans , Liver/cytology , Liver/drug effects , Liver/metabolism , Liver Neoplasms/drug therapy , Liver Neoplasms/metabolism , Liver Neoplasms/pathology , Membrane Potential, Mitochondrial/drug effects , Mitochondria/drug effects , Mitochondria/metabolism , Nanoparticles/administration & dosage , Oxidation-Reduction/drug effects , Oxidative Stress/drug effects , Plant Extracts/chemistry , Plant Extracts/isolation & purification , Plant Extracts/pharmacology , Proto-Oncogene Proteins c-bcl-2/metabolism , Reactive Oxygen Species/metabolism
10.
Mater Sci Eng C Mater Biol Appl ; 90: 104-112, 2018 Sep 01.
Article in English | MEDLINE | ID: mdl-29853073

ABSTRACT

Selenium nanoparticles (Se NPs) attract a lot of attention as potential cancer therapeutic agents. However, the antitumor activities of pure Se NPs are poor, and some modifiers are needed to enhance the activities. In the present study, we prepared Ferulic Acid (FA)-modified selenium nanoparticles in a facile synthetic approach. The obtained FA-Se NPs were characterized using transmission electron microscope (TEM), dynamic light scattering (DLS), ultraviolet-visible spectrophotometer (UV-VIS), Fourier transform infrared spectroscopy (FTIR), X-ray diffraction (XRD) and Energy dispersive X-ray (EDX) spectroscopy. In vitro antitumor effects of FA, Se NPs and FA-Se NPs in HepG-2 cells were examined by methyl thiazolyl tetrazolium (MTT) assay. It showed that FA-Se NPs effectively inhibited the growth of HepG-2 cells with IC50 value of 11.57 ±â€¯3.6 µg/ml, while the value of Se NPs was >100 µg/ml. In addition, FA behaves no obvious antitumor effects at high concentrations up to 100 µg/ml. In order to investigate the antitumor mechanism of FA-Se NPs, fluorescence morphological examination and Annexin V-FITC/PI staining analysis were performed to observe the apoptosis of HepG-2 cells induced by FA-Se NPs. Meanwhile, mitochondrial membrane potential (MMP), intracellular reactive oxygen species (ROS) levels and caspase-3 and -9 activities were detected. The results revealed that FA-Se NPs induced intracellular ROS generation and MMP disruption by finally activating caspase-3/9 to trigger HepG-2 cells apoptosis through mitochondrial pathway. Further investigation on the interactions of FA-Se NPs with calf thymus DNA (ctDNA) indicated that the antitumor activities may be associated with the DNA-binding properties of FA-Se NPs.


Subject(s)
Coumaric Acids/chemistry , Nanoparticles/chemistry , Selenium/chemistry , Hep G2 Cells , Humans , Membrane Potential, Mitochondrial/drug effects , Microscopy, Electron, Transmission , Mitochondria/drug effects , Reactive Oxygen Species/metabolism , Selenium/pharmacology , Spectroscopy, Fourier Transform Infrared , X-Ray Diffraction
11.
Pharmacol Rep ; 70(3): 558-564, 2018 Jun.
Article in English | MEDLINE | ID: mdl-29677526

ABSTRACT

BACKGROUND: Great attention has been paid to the development of novel anti-inflammatory drugs to overcome the adverse reactions of traditional drugs. Recently, a new compound 4-o-methyl-benzenesulfonyl benzoxazolone (MBB) we have prepared attracted our attention for its promising anti-inflammatory activity and low toxicity. The present study aimed to further investigate the anti-inflammatory effects of MBB both in vivo and in vitro in order to determine its potential as a novel NSAIDs lead compound. METHODS: The anti-inflammatory effects in vivo were evaluated using acetic acid-induced mice writhing, xylene-induced mice ear edema and collagen-induced rat arthritis. NO, TNF-α, IL-6, IL-1ß and iNOS productions by LPS-stimulated RAW264.7 cells were determined to investigate the basis of anti-inflammatory effects. Finally, the COX inhibition effect was tested in vitro using COX inhibitor screening assay kit. RESULTS: MBB could significantly decrease the writhing and ear swelling in a dose-dependent manner, and it also had a moderate anti-arthritic potential associated with an attenuation of arthritis index score, arthritis swelling, and inhibition of TNF-α and IL-1ß. MBB could inhibit the activity of NO, TNF-α, IL-6, IL-1ß and iNOS to perform its activity in vitro, but it had no effect against COX-1 and COX-2. The anti-inflammation effect may be mediated via the inhibition of iNOS to reduce the production of inflammatory mediators which should be further confirmed. CONCLUSIONS: The compound MBB displayed anti-inflammatory and anti-arthritic effect, and it could be considered as a new NSAIDs lead compound for the further structure modification to develop novel anti-inflammatory drugs.


Subject(s)
Anti-Inflammatory Agents, Non-Steroidal/pharmacology , Anti-Inflammatory Agents/pharmacology , Benzoxazoles/pharmacology , Animals , Arthritis, Experimental/drug therapy , Arthritis, Experimental/metabolism , Cyclooxygenase 1/metabolism , Cyclooxygenase 2/metabolism , Edema/drug therapy , Edema/metabolism , Female , Inflammation/drug therapy , Inflammation/metabolism , Interleukin-1beta/metabolism , Interleukin-6/metabolism , Male , Mice , Nitric Oxide/metabolism , Nitric Oxide Synthase Type II/metabolism , RAW 264.7 Cells , Rats , Rats, Sprague-Dawley , Tumor Necrosis Factor-alpha/metabolism
12.
Drug Des Devel Ther ; 11: 1891-1904, 2017.
Article in English | MEDLINE | ID: mdl-28694689

ABSTRACT

This study deals with the design and synthesis of a series of novel 4-methoxy-substituted and 5-methyl-substituted (3'S,4'S)-(-)-cis-khellactones. The newly synthesized compounds were characterized by 1H nuclear magnetic resonance (NMR), 13C-NMR, mass spectrometry, and elemental analysis. All the derivatives were subjected to in vitro cytotoxicity screening against HEPG-2 (human liver carcinoma), SGC-7901 (human gastric carcinoma), and LS174T (human colon carcinoma), by using the MTT assay. The results revealed that several of the 4-methoxy-substituted compounds exhibited potent cytotoxicity. Among these, compound 12e showed the highest activity against cancer cells which 50% inhibitory concentration (IC50) values were in the range of 6.1-9.2 µM with low toxicity on normal human hepatocyte. Preliminary investigation of possible mechanisms of action of compound 12e against HEPG-2 cells indicated possible induction of apoptosis, as determined by morphological observations and Annexin V/propidium iodide (PI) double staining, in addition to apparent dissipation of mitochondrial membrane potential (MMP), as measured by 5,5',6,6'-tetrachloro-1,1',3,3'-tetraethyl-imidacarbocyanine iodide (JC-1) staining in combination with the activation of caspase-9 and caspase-3 by Western blot analysis. Overall, the data suggest that compound 12e may be a promising potential anti-cancer agent that could act primarily by inducing apoptosis through the mitochondria-mediated intrinsic pathway in human hepatoma cells.


Subject(s)
Antineoplastic Agents, Phytogenic/chemical synthesis , Antineoplastic Agents, Phytogenic/pharmacology , Apoptosis/drug effects , Coumarins/chemical synthesis , Coumarins/pharmacology , Caspases/drug effects , Cell Line, Tumor , Drug Screening Assays, Antitumor , Enzyme Activation , Humans , Magnetic Resonance Spectroscopy , Membrane Potential, Mitochondrial/drug effects , Signal Transduction/drug effects , Structure-Activity Relationship , Tandem Mass Spectrometry , Tetrazolium Salts , Thiazoles
13.
Carbohydr Polym ; 151: 996-1005, 2016 Oct 20.
Article in English | MEDLINE | ID: mdl-27474647

ABSTRACT

Dibutyltin (DBT) which was widely used as biocide and plastic stabilizer has been described as a potent neurotoxicant. Chitosan (CS), a natural nontoxic biopolymer, possesses a variety of biological activities including antibacterial, antifungal, free radical scavenging and neuroprotective activities. The present study was undertaken to investigate the protective effects of CS against DBT-induced apoptosis in rat pheochromocytoma (PC12) cells and the underlying mechanisms in vitro. Our results demonstrated that pretreatment with CS significantly increased the cell viability and decreased lactate dehydrogenase (LDH) release induced by DBT in a dose-dependent manner. Meanwhile, DBT-induced cell apoptosis, mitochondrial membrane potential (MMP) disruption, and generation of intracellular reactive oxygen species (ROS) were attenuated by CS. Real-time PCR assay showed that DBT markedly enhanced the mRNA levels of Bax, Bad, cytochrome-c and Apaf-1, reduced the Bcl-2 and Bcl-xL mRNA levels, while these genes expression alteration could be partially reversed by CS treatment. Furthermore, CS also inhibited the DBT-inducted activation of caspase-9, and -3 at mRNA and protein expression levels. Taken together, these results suggested that CS could protect the PC12 cells from apoptosis induced by DBT through inhibition of the mitochondria-dependent pathway.


Subject(s)
Apoptosis/drug effects , Chitosan/pharmacology , Mitochondria/drug effects , Neurotoxins/toxicity , Organotin Compounds/toxicity , Animals , Cytoprotection/drug effects , Gene Expression Regulation/drug effects , Membrane Potential, Mitochondrial/drug effects , PC12 Cells , Rats , Reactive Oxygen Species/metabolism
14.
Eur J Cancer Prev ; 25(6): 490-9, 2016 11.
Article in English | MEDLINE | ID: mdl-26656929

ABSTRACT

Current cancer treatment is partly limited by chemotherapy-induced vascular toxicity associated with damage to vascular endothelial cells. In this study, the cytotoxicity of farrerol against SGC7901 gastric cancer cells and human umbilical vein endothelial cells (HUVECs) in vitro was investigated along with the underlying mechanisms of its growth-inhibitory effect against SGC7901 cells. MTT assays showed that farrerol inhibited SGC7901 cell growth, but exerted no cytotoxicity against HUVECs. Flow cytometry showed that treatment of SGC7901 cells with farrerol (5, 40, or 160 µmol/l) for 24 h caused G0/G1 cell cycle arrest in a concentration-dependent manner. Western blotting indicated that exposure of SGC7901 cells to farrerol resulted in significant upregulation of p27KIP1 (p27), accompanied by sustained activation of ERK1/2 and p38 MAPK instead of JNK. Farrerol-stimulated p27 expression, p38 MAPK activation, and cell growth inhibition were attenuated by pretreatment with U0126, an MEK1/2 inhibitor. In conclusion, this study indicates the selective cytotoxicity of farrerol against SGC7901 cells, but not HUVECs. Furthermore, it provides the first evidence that farrerol could induce cancer cell growth inhibition by G0/G1-phase cell-cycle arrest mediated by sustained ERK activation. The findings show the potential of farrerol as a chemotherapeutic agent without vascular toxicity for use against gastric cancer.


Subject(s)
Cell Cycle Checkpoints/drug effects , Cell Proliferation/drug effects , Chromones/pharmacology , G1 Phase/drug effects , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3/metabolism , Resting Phase, Cell Cycle/drug effects , Stomach Neoplasms/pathology , Apoptosis/drug effects , Humans , Stomach Neoplasms/drug therapy , Stomach Neoplasms/metabolism , Tumor Cells, Cultured
15.
Eur J Pharmacol ; 769: 211-8, 2015 Dec 15.
Article in English | MEDLINE | ID: mdl-26593430

ABSTRACT

Farrerol, a typical natural flavanone, is the major active component of Rhododendron dauricum L. The objective of this study was to evaluate the attenuation effect of farrerol against the aortic lesions in spontaneously hypertensive rats (SHR) for the first time. Twelve-week-old male SHR were orally administered with farrerol (50mg/kg/day), verapamil (50mg/kg/day, positive control), or vehicle for 8 weeks (n=10 in each group). Age-matched Wistar-Kyoto rats (WKY) served as normal controls (n=10). Our results revealed that farrerol significantly reduced the systolic blood pressure in SHR (from 177±4mmHg to 158±5mmHg) and also dramatically attenuated the aortic lesion, which is characterized by decreased media thickness, wall area, media-lumen ratio, nuclei size and an increased nuclei number (P<0.05). Moreover, the levels of O2(-) along with NAD(P)H oxidase activity were reduced (P<0.05), while the activity of endothelial nitric oxide synthase (eNOS) was elevated (P<0.05) in aortic homogenates after the intervention of farrerol. Furthermore, farrerol upregulated the expression of eNOS in both of mRNA and protein levels, accompanied by the downregulated mRNA and protein expression of p22(phox) (P<0.05), an essential subunit for NADPH oxidase activity. Our findings indicated that farrerol has a significant protective effect against the aortic lesion in SHR, which may be related to the enhanced eNOS activity and reduced NAD(P)H oxidase activity.


Subject(s)
Aorta/drug effects , Aorta/metabolism , Chromones/pharmacology , NADPH Oxidases/metabolism , Nitric Oxide Synthase Type III/genetics , Nitric Oxide Synthase Type III/metabolism , Up-Regulation/drug effects , Animals , Aorta/pathology , Aorta/physiology , Blood Pressure/drug effects , Gene Expression Regulation, Enzymologic/drug effects , Male , NADPH Oxidases/genetics , Nitrites/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism , Rats , Rats, Inbred SHR , Superoxides/metabolism
16.
Eur J Cancer Prev ; 24(5): 365-72, 2015 Sep.
Article in English | MEDLINE | ID: mdl-26061993

ABSTRACT

Farrerol, a typical flavanone isolated from the Chinese medicinal plant Rhododendron dauricum L., has been found to show various biological activities. However, to the best of our knowledge, its inhibitory actions against cancer cells have not been reported as yet. Therefore, the present study aimed to investigate the cytotoxic and apoptotic effects of farrerol on human gastric cancer SGC-7901 cells. Farrerol showed a 50% inhibition of SGC-7901 cell growth at a concentration of 40.4 µmol/l for 24 h according to MTT assays. The cell morphology results indicated that SGC-7901 cells treated with farrerol showed several features of apoptotic cell death, which was also confirmed by the Annexin-V FITC/PI double-staining assay. Further studies showed that farrerol treatment induced the attenuation of mitochondrial membrane potential, accompanied by the release of Cyt-c and the activation of caspase-9 and caspase-3. Furthermore, farrerol decreased the gene expression of Bcl-2, whereas the gene expression level of Bax was found to increase after farrerol treatment. These combined results indicated that farrerol can induce apoptosis through a mitochondrial-mediated pathway.


Subject(s)
Antineoplastic Agents, Phytogenic/pharmacology , Apoptosis/drug effects , Chromones/pharmacology , Membrane Potential, Mitochondrial/drug effects , Mitochondria/pathology , Signal Transduction/drug effects , Stomach Neoplasms/pathology , Blotting, Western , Caspase 3/genetics , Caspase 3/metabolism , Caspase 9/genetics , Caspase 9/metabolism , Cell Proliferation/drug effects , Humans , Mitochondria/drug effects , Mitochondria/metabolism , RNA, Messenger/genetics , Real-Time Polymerase Chain Reaction , Reverse Transcriptase Polymerase Chain Reaction , Stomach Neoplasms/drug therapy , Stomach Neoplasms/metabolism , Tumor Cells, Cultured , bcl-2-Associated X Protein/genetics , bcl-2-Associated X Protein/metabolism
17.
Biomed Pharmacother ; 71: 119-27, 2015 Apr.
Article in English | MEDLINE | ID: mdl-25960226

ABSTRACT

Two novel diorganotin (IV) complexes, based on 4-nitro-N-phthaloyl-glycine (HL), namely {4-NO2C6H3(CO)2NCH2COO}2Sn(n-Bu)2 (1) and {4-NO2C6H3(CO)2NCH2COO}2SnMe2 (2), were synthesized and characterized by elemental analysis, FT-IR, (1)H- and (13)C-NMR spectroscopic techniques. In vitro antitumor activities of both complexes were evaluated by the 3-(4,5-dimethylthiazoly-2-yl)-2,5-diphenyltetrazolium bromide (MTT) assay against three human cancer cell lines: HepG-2 (human liver carcinoma), SGC-7901 (human gastric carcinoma) and LS174T (human colon carcinoma). Complex 1 exhibited strong antitumor activity with IC50 values of 1.51±0.41, 1.80±0.63, and 2.48±0.96 µM, respectively; while complex 2 had no obvious effects on the three selected cancer cell lines at high concentrations up to 100 µM. Complex 1-induced apoptosis was further confirmed by morphological observations and annexin V-FITC/PI staining flow cytometry analysis in HepG-2 cells. Cell cycle analysis revealed that complex 1 caused cell cycle arrest at G2/M phase. Molecular mechanism studies suggested that the apoptosis was mediated through the mitochondrial pathway with intracellular reactive oxygen species (ROS) promotion and mitochondrial membrane potential (MMP) disruption by finally activating effector caspase-3/9 to trigger cell apoptosis. Moreover, the interactions of both complexes with calf thymus DNA (CT-DNA) were investigated by using UV-Vis titration and fluorometric competition measurements. The DNA-binding constants Kb (intrinsic binding constant) and K(sv) (quenching constant) had been obtained in the order: 1>2, consisted with the antitumor activity results. Taken together, complex 1 exhibited excellent antitumor activity suggesting that it may be a potential candidate for further chemical optimization and cancer therapy.


Subject(s)
Antineoplastic Agents/chemical synthesis , Antineoplastic Agents/pharmacology , Coordination Complexes/chemical synthesis , Coordination Complexes/pharmacology , DNA, Neoplasm/metabolism , Glycine/chemical synthesis , Glycine/pharmacology , Organotin Compounds/chemical synthesis , Organotin Compounds/pharmacology , Phthalimides/chemical synthesis , Phthalimides/pharmacology , Animals , Antineoplastic Agents/chemistry , Apoptosis/drug effects , Caspase 3/metabolism , Caspase 9/metabolism , Cattle , Cell Cycle Checkpoints/drug effects , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Shape/drug effects , Coordination Complexes/chemistry , DNA/metabolism , Enzyme Activation/drug effects , Ethidium/metabolism , Flow Cytometry , Glycine/chemistry , Humans , Membrane Potential, Mitochondrial/drug effects , Mitochondria/drug effects , Mitochondria/metabolism , Organotin Compounds/chemistry , Phthalimides/chemistry , Proton Magnetic Resonance Spectroscopy , Reactive Oxygen Species/metabolism , Spectrometry, Fluorescence , Spectrophotometry, Ultraviolet , Spectroscopy, Fourier Transform Infrared
18.
Int J Mol Sci ; 15(4): 6641-56, 2014 Apr 17.
Article in English | MEDLINE | ID: mdl-24747597

ABSTRACT

Farrerol, isolated from Rhododendron dauricum L., has been proven to be an important multifunctional physiologically active component, but its vasoactive mechanism is not clear. The present study was performed to observe the vasoactive effects of farrerol on rat aorta and to investigate the possible underlying mechanisms. Isolated aortic rings of rat were mounted in an organ bath system and the myogenic effects stimulated by farrerol were studied. Intracellular Ca2+ ([Ca2+]in) was measured by molecular probe fluo-4-AM and the activities of L-type voltage-gated Ca2+ channels (LVGC) were studied with whole-cell patch clamp in cultured vascular smooth muscle cells (VSMCs). The results showed that farrerol significantly induced dose-dependent relaxation on aortic rings, while this vasorelaxation was not affected by NG-nitro-l-arginine methylester ester or endothelium denudation. In endothelium-denuded aortas, farrerol also reduced Ca2+-induced contraction on the basis of the stable contraction induced by KCl or phenylephrine (PE) in Ca2+-free solution. Moreover, after incubation with verapamil, farrerol can induce relaxation in endothelium-denuded aortas precontracted by PE, and this effect can be enhanced by ruthenium red, but not by heparin. With laser scanning confocal microscopy method, the farrerol-induced decline of [Ca2+]in in cultured VSMCs was observed. Furthermore, we found that farrerol could suppress Ca2+ influx via LVGC by patch clamp technology. These findings suggested that farrerol can regulate the vascular tension and could be developed as a practicable vasorelaxation drug.


Subject(s)
Aorta/drug effects , Calcium/metabolism , Chromones/pharmacology , Muscle, Smooth, Vascular/metabolism , Animals , Aorta/cytology , Aorta/physiology , Calcium Channels, L-Type/metabolism , Cells, Cultured , Chromones/chemistry , Chromones/isolation & purification , Male , Muscle Contraction/drug effects , Muscle, Smooth, Vascular/cytology , Patch-Clamp Techniques , Phenylephrine/pharmacology , Rats , Rats, Sprague-Dawley , Rhododendron/chemistry , Rhododendron/metabolism , Vasodilation/drug effects , Verapamil/pharmacology
19.
Food Chem Toxicol ; 60: 302-8, 2013 Oct.
Article in English | MEDLINE | ID: mdl-23927876

ABSTRACT

As ubiquitous environmental toxicants, organotin (IV) compounds (OTC) accumulate in the food chain and potential effects on human health are disquieting. The present study compared the cytotoxicity of three diorganotins, namely, dimethyltin (DMT), dibutyltin (DBT) and diphenyltin (DPT), in rat pheochromocytoma (PC12) cells, and the molecular mechanisms responsible for their cytotoxic effects were also explored. Twenty-four hours exposure of PC12 cells to DBT and DPT resulted in a concentration-dependent decrease in cell viability with median lethal concentration (LC50) of 2.97 µM and 7.24 µM, respectively. However, DMT at concentrations up to 128 µM had no obvious effect on cell viability. The mechanistic study revealed that the extent of apoptosis was greater for DBT than that for DPT, followed by DMT, as evidenced by acridine orange/ethidium bromide (AO/EB) fluorescent staining method and annexin V-FITC/PI staining flow cytometry analysis, as well as generation of intracellular reactive oxygen species (ROS), mitochondrial membrane potential (MMP) disruption, release of cytochrome c (Cyt c), and consequent activation of caspase-9, and -3. These investigations suggested that the cytotoxic potency of three diorganotins in PC12 cells was in the order of DBT>DPT≫DMT, and these compounds could induce PC12 cells apoptosis through ROS mediated mitochondrial pathway.


Subject(s)
Apoptosis/drug effects , Organotin Compounds/toxicity , Animals , Caspase 3/genetics , Caspase 3/metabolism , Caspase 9/genetics , Caspase 9/metabolism , Cell Line, Tumor , Cell Survival/drug effects , Cytochromes c/metabolism , Lethal Dose 50 , Membrane Potential, Mitochondrial/drug effects , PC12 Cells , Rats , Reactive Oxygen Species/metabolism
20.
Molecules ; 18(4): 4158-69, 2013 Apr 08.
Article in English | MEDLINE | ID: mdl-23567363

ABSTRACT

An asymmetric synthesis of a series of novel 4-methyl-(3'S,4'S)-cis-khellactone derivatives 3a-o is reported for the first time. Their structures were confirmed by 1H-NMR, 13C-NMR and MS. Their cytotoxic activity was evaluated by the MTT assay against three selected human cancer cell lines: HEPG-2 (human liver carcinoma), SGC-7901 (human gastric carcinoma), LS174T (human colon carcinoma). Some compounds showed high inhibitory activity against these human cancer cell lines. Among them, compound 3a exhibited strong cytotoxicity, with IC50 values ranging from 8.51 to 29.65 µM. The results showed that 4-methyl-cis-khellactone derivatives with S,S configuration could be a potential antitumor agents.


Subject(s)
Antineoplastic Agents/chemical synthesis , Antineoplastic Agents/pharmacology , Coumarins/chemical synthesis , Coumarins/pharmacology , Cell Line, Tumor , Cell Proliferation/drug effects , Hep G2 Cells , Humans , Inhibitory Concentration 50 , Molecular Structure , Structure-Activity Relationship
SELECTION OF CITATIONS
SEARCH DETAIL
...