Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 80
Filter
1.
Sci Total Environ ; 933: 172824, 2024 Jul 10.
Article in English | MEDLINE | ID: mdl-38688370

ABSTRACT

A recently synthesized aminated 3,4-dioxygenated xanthone (Xantifoul2) was found to have promising antifouling (AF) effects against the settlement of the macrofouler Mytilus galloprovincialis larvae. Preliminary assessment indicated that Xantifoul2 has reduced ecotoxicological impacts: e.g., being non-toxic to the marine crustacea Artemia salina (<10 % mortality at 50 µM) and showing low bioconcentration factor in marine organisms. In order to meet the EU Biocidal Product Regulation, a preliminary hazard assessment of this new nature-inspired antifouling (NIAF) agent was conducted in this work. Xantifoul2 did not affect the swimming ability of the planktonic crustacean Daphnia magna, the growth of the diatom Phaeodactylum tricornutum, and the cellular respiration of luminescent Gram-negative bacteria Vibrio fischeri, supporting the low toxicity towards several non-target marine species. Regarding human cytotoxicity, Xantifoul2 did not affect the cell viability of retinal human cells (hTERT-RPE-1) and lipidomic studies revealed depletion of lipids involved in cell death, membrane modeling, lipid storage, and oxidative stress only at a high concentration (10 µM). Accelerated degradation studies in water were conducted under simulated sunlight to allow the understanding of putative transformation products (TPs) that could be generated in the aquatic ecosystems. Both Xantifoul2 and photolytic-treated Xantifoul2 in the aqueous matrix were therefore evaluated on several nuclear receptors (NRs). The results of this preliminary hazard assessment of Xantifoul2, combined with the high degradation rates in water, provide strong evidence of the safety of this AF agent under the evaluated conditions, and provide the support for future validation studies before this compound can be introduced in the market.


Subject(s)
Biofouling , Biofouling/prevention & control , Animals , Water Pollutants, Chemical/toxicity , Aliivibrio fischeri/drug effects , Xanthones/toxicity , Mytilus/drug effects , Mytilus/physiology , Diatoms/drug effects , Humans , Daphnia/drug effects , Daphnia/physiology , Artemia/drug effects
2.
Fitoterapia ; 157: 105127, 2022 Mar.
Article in English | MEDLINE | ID: mdl-35033607

ABSTRACT

One new xanthone, griseophenexanthone A (1), one new benzophenone, digriseophene A (2), and 14 previously reported compounds were isolated from the culture of Penicillium sp. ct-28, an endophytic fungus of Corydlis tomentella. The structures of the isolated compounds were identified by an extensive analysis of HRESIMS, 1D and 2D NMR. MTT assay showed that six xanthones (1 and 3-7) significantly inhibited cell proliferation in four cancer cell lines, with IC50 values ranging from 18.12 ± 2.42 to 85.55 ± 7.66 µM. Our results showed that slight structural changes led to obvious activity differences among these compounds. We also investigated the effects of the six xanthones on cell cycle and apoptosis in human hepatoma HepG2 cells. Compound 7 caused cell cycle arrest at G1 phase, compounds 5 and 6 caused cell cycle arrest at S phase, whereas compounds 1, 3 and 4 had no effects on cell cycle distribution. All six xanthones induced apoptosis in dose-dependent manners in HepG2 cells accompanied by degradation of PARP and activation of caspase 3. The structure-activity relationship analysis revealed that the effects of these xanthones on cell cycle and apoptosis in HepG2 cells were closely related to the substituent groups on their skeleton. Our studies provide novel insights for the structural optimization of xanthones in the development of new anticancer drugs.


Subject(s)
Benzophenones/toxicity , Cell Proliferation/drug effects , Corydalis/microbiology , Penicillium/chemistry , Xanthones/toxicity , Apoptosis/drug effects , Benzophenones/chemistry , Benzophenones/isolation & purification , Cell Cycle/drug effects , Cell Line, Tumor , Dose-Response Relationship, Drug , Hep G2 Cells , Humans , Inhibitory Concentration 50 , Magnetic Resonance Spectroscopy , Molecular Structure , Structure-Activity Relationship , Xanthones/chemistry , Xanthones/isolation & purification
3.
J Ethnopharmacol ; 285: 114796, 2022 Mar 01.
Article in English | MEDLINE | ID: mdl-34740771

ABSTRACT

ETHNOPHARMACOLOGICAL RELEVANCE: Sophora flavescens is a traditional Chinese medicine commonly used in clinical practice, which has the effects of clearing away heat and dampness. Unfortunately, it has been reported that Sophora flavescens and its preparation may cause liver damage to a certain extent, but the exact mechanism is not clear. AIM OF THE STUDY: To assess the safety and risk of Sophora flavescens and to elucidate the relationship between Idiosyncratic drug-induced liver injury (IDILI) and the NOD-like receptor family protein 3 (NLRP3) inflammasome. MATERIALS AND METHODS: Western blot, Caspase-Glo® 1 Inflammasome Assay, ELISA kits, Flow cytometry and FLIPRT Tetra system were used to study the effect of isoxanthohumol (IXN) on the activation of NLRP3 inflammasome and its mechanism. Combined with the lipopolysaccharide-mediated susceptibility IDILI model in mice to evaluate the hepatotoxicity of IXN. RESULTS: IXN facilitates the activation of caspase-1 and secretion of interleukin (IL)-1ß triggered by adenosine triphosphate (ATP), nigericin but not those induced by silicon dioxide and poly (I:C). Furthermore, the activation of NLR-family CARD-containing protein 4 (NLRC4) and the absent in melanoma 2 (AIM2) was not affected by IXN. Mechanistically, IXN promotes NLRP3-dependent apoptosis-associated speck-like protein containing a C-terminal caspase recruitment domain (ASC) oligomerization and the generation of mitochondrial reactive oxygen species (mtROS) triggered by ATP. The in vivo data showed that non-hepatotoxic doses of IXN resulted in increased levels of glutamate-pyruvate transaminase, glutamate-oxaloacetate transaminase, tumor necrosis factor and IL-1ß in the serum and showed increased liver inflammation in the susceptible IDILI model mediated by lipopolysaccharide. CONCLUSIONS: These results show that IXN enhances NLRP3 inflammasome activation by promoting the accumulation of ATP-induced mtROS and ASC oligomerization to cause IDILI, indicating that IXN may be a risk factor for liver injury caused by the clinical use of Sophora flavescens.


Subject(s)
Chemical and Drug Induced Liver Injury , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , Sophora/chemistry , Xanthones , Adenosine Triphosphate/metabolism , Animals , Aspartate Aminotransferases/metabolism , Cells, Cultured , Chemical and Drug Induced Liver Injury/immunology , Chemical and Drug Induced Liver Injury/metabolism , Inflammasomes/metabolism , Medicine, Chinese Traditional/methods , Mice , Mice, Inbred C57BL , Reactive Oxygen Species/metabolism , Xanthones/pharmacology , Xanthones/toxicity
4.
Fitoterapia ; 156: 105092, 2022 Jan.
Article in English | MEDLINE | ID: mdl-34826556

ABSTRACT

Caged-polyprenylated xanthonoids represent a rare class of natural products. This type of compounds is mainly isolated from Genus Garcinia. Phytochemical studies on the leaves and twigs of Garcinia oligantha led to the isolation of four new caged-polyprenylated xanthonoids, oliganthone CF (1-4), and two new simple xanthones (5-6), oliganthaxanthone D and oliganthaxanthone E. Eight known other polyprenylated xanthones (7-14) including five caged-polyprenylated xanthonoids (7-11) were also isolated. Their structures were elucidated based on the analyses of extensive spectroscopic data. All the isolated compounds except for 5, 6 and 14 showed cell viability reducing effect against human lung cancer A549 cells. Compounds 1-3 were proved to be potential apoptosis inducing agents.


Subject(s)
Cytotoxins/toxicity , Garcinia/chemistry , Plant Extracts/toxicity , Xanthones/toxicity , A549 Cells , Apoptosis , Blotting, Western , Cytotoxins/chemistry , Cytotoxins/isolation & purification , Humans , Molecular Structure , Plant Extracts/chemistry , Plant Extracts/isolation & purification , Plant Leaves/chemistry , Plant Stems/chemistry , Spectrophotometry, Infrared , Spectrophotometry, Ultraviolet , Xanthones/chemistry , Xanthones/isolation & purification
5.
Biomed Pharmacother ; 144: 112333, 2021 Dec.
Article in English | MEDLINE | ID: mdl-34678724

ABSTRACT

Diabetes mellitus (DM) is concomitant with significant morbidity and mortality and its prevalence is accumulative in worldwide. The conventional antidiabetic agents are known to mitigate the symptoms of diabetes; however, they may also cause side and adverse effects. There is an imperative necessity to conduct preclinical and clinical trials for the discovery of alternative therapeutic agents that can overcome the drawbacks of current synthetic antidiabetic drugs. This study aimed to investigate the efficacy of lowering blood glucose and underlined mechanism of γ-mangostin, mangosteen (Garcinia mangostana) xanthones. The results showed γ-Mangostin had a antihyperglycemic ability in short (2 h)- and long-term (28 days) administrations to diet-induced diabetic mice. The long-term administration of γ-mangostin attenuated fasting blood glucose of diabetic mice and exhibited no hepatotoxicity and nephrotoxicity. Moreover, AMPK, PPARγ, α-amylase, and α-glucosidase were found to be the potential targets for simulating binds with γ-mangostin after molecular docking. To validate the docking results, the inhibitory potency of γ-mangostin againstα-amylase/α-glucosidase was higher than Acarbose via enzymatic assay. Interestingly, an allosteric relationship between γ-mangostin and insulin was also found in the glucose uptake of VSMC, FL83B, C2C12, and 3T3-L1 cells. Taken together, the results showed that γ-mangostin exerts anti-hyperglycemic activity through promoting glucose uptake and reducing saccharide digestion by inhibition of α-amylase/α-glucosidase with insulin sensitization, suggesting that γ-mangostin could be a new clue for drug discovery and development to treat diabetes.


Subject(s)
AMP-Activated Protein Kinases/metabolism , Blood Glucose/drug effects , Diabetes Mellitus/drug therapy , Garcinia mangostana , Glycoside Hydrolase Inhibitors/pharmacology , Insulin Resistance , PPAR gamma/metabolism , Plant Extracts/pharmacology , Xanthones/pharmacology , 3T3-L1 Cells , Animals , Biomarkers/blood , Blood Glucose/metabolism , Diabetes Mellitus/blood , Diabetes Mellitus/enzymology , Diet, High-Fat , Disease Models, Animal , Down-Regulation , Garcinia mangostana/chemistry , Glycoside Hydrolase Inhibitors/isolation & purification , Glycoside Hydrolase Inhibitors/toxicity , Male , Mice , Mice, Inbred ICR , Plant Extracts/isolation & purification , Plant Extracts/toxicity , Signal Transduction , Time Factors , Xanthones/toxicity , alpha-Amylases/antagonists & inhibitors , alpha-Amylases/metabolism
6.
J Biochem Mol Toxicol ; 35(4): e22721, 2021 Apr.
Article in English | MEDLINE | ID: mdl-33533530

ABSTRACT

Gastric cancer (GC) represents the fifth most human malignant disease and the third-most common cause of cancer-related death. Gambogic acid (GA) is a natural compound with a polyprenylated xanthone structure and possesses remarkable antitumor activity in a variety of cancer cells. However, the mechanism underlying the inhibitory effect of GA in GC is far from being completely understood. The goal of the present study is to investigate whether potential microRNAs are involved in antitumor effect of GA toward GC and to elucidate the possible mechanisms. We identified that miR-26a-5p was significantly increased by GA in GC cell lines and xenograft tumor. Downregulation of miR-26a-5p not only prevented GA-induced inhibition on GC cell growth, but also suppressed GA-induced apoptosis of GC cells. Informatics assay predicted that Wnt5a was regulated by miR-26a-5p and GA-induced downregulation of Wnt5a was prevented by anti-miR-26a-5p. Reporter gene assay showed that miR-26a-5p could negatively regulate Wnt5a through direct binding with 3'-UTR messenger RNA of Wnt5a. Thus, upregulation of Wnt5a exhibited the same action tendency for GA-induced GC cell growth and apoptosis as observed by downregulation of miR-26a-5p. In conclusion, these results indicated that the inhibitory effect of GA on GC was mediated by the upregulation of miR-26a-5p and downregulation of Wnt5a. Our study provided new clues for the potential therapeutic effect of GA against GC and highlighted the importance of miR-26a-5p/Wnt5a pathway in the regulation of GC development.


Subject(s)
MicroRNAs/metabolism , Neoplasm Proteins/metabolism , RNA, Neoplasm/metabolism , Stomach Neoplasms/metabolism , Wnt Signaling Pathway/drug effects , Wnt-5a Protein/metabolism , Xanthones/toxicity , Animals , Cell Line, Tumor , Female , Humans , Mice , Mice, Inbred BALB C , Mice, Nude , MicroRNAs/genetics , Neoplasm Proteins/genetics , RNA, Neoplasm/genetics , Stomach Neoplasms/chemically induced , Stomach Neoplasms/genetics , Wnt-5a Protein/genetics
7.
Molecules ; 25(22)2020 Nov 16.
Article in English | MEDLINE | ID: mdl-33207599

ABSTRACT

We investigated alpha-mangostin (α-mangostin, α-MG), a xanthone natural product extracted from the pericarp of mangosteen (Garcinia mangostana), for its antifungal activities and possible mechanism against Colletotrichum gloeosporioides, which causes mango anthracnose. The results demonstrated that α-MG had a relatively high in vitro inhibitory activity against C. gloeosporioides among 20 plant pathogenic fungi. The median effective concentration (EC50) values of α-MG against mycelial growth were nearly 10 times higher than those of spore germination inhibition for both strains of C. gloeosporioides, the carbendazim-sensitive (CBD-s) and carbendazim-resistant (CBD-r). The results suggested that α-MG exhibited a better inhibitory effect on spore germination than on the mycelial growth of C. gloeosporioides. Further investigation indicated that the protective effect could be superior to the therapeutic effect for mango leaves for scab development. The morphological observations of mycelium showed that α-MG caused the accumulation of dense bodies. Ultrastructural observation further revealed that α-MG caused a decrease in the quantity and shape of the swelling of mitochondria in the mycelium cells of C. gloeosporioides. In addition, bioassays disclosed that the inhibitory activity of α-MG on spore germination was reduced by adding exogenous adenosine triphosphate (ATP). These results suggested that the mode of action of α-MG could be involved in the destruction of mitochondrial energy metabolism. The current study supports α-MG as a natural antifungal agent in crop protection.


Subject(s)
Antifungal Agents/pharmacology , Colletotrichum/drug effects , Xanthones/pharmacology , Adenosine Triphosphate/pharmacology , Antifungal Agents/chemistry , Colletotrichum/ultrastructure , Microbial Sensitivity Tests , Mycelium/drug effects , Mycelium/growth & development , Mycelium/ultrastructure , Plant Leaves/chemistry , Spores, Fungal/drug effects , Xanthones/chemistry , Xanthones/toxicity
8.
Biomolecules ; 10(8)2020 07 30.
Article in English | MEDLINE | ID: mdl-32751491

ABSTRACT

The development of alternative ecological and effective antifouling technologies is still challenging. Synthesis of nature-inspired compounds has been exploited, given the potential to assure commercial supplies of potential ecofriendly antifouling agents. In this direction, the antifouling activity of a series of nineteen synthetic small molecules, with chemical similarities with natural products, were exploited in this work. Six (4, 5, 7, 10, 15 and 17) of the tested xanthones showed in vivo activity toward the settlement of Mytilus galloprovincialis larvae (EC50: 3.53-28.60 µM) and low toxicity to this macrofouling species (LC50 > 500 µM and LC50/EC50: 17.42-141.64), and two of them (7 and 10) showed no general marine ecotoxicity (<10% of Artemia salina mortality) after 48 h of exposure. Regarding the mechanism of action in mussel larvae, the best performance compounds 4 and 5 might be acting by the inhibition of acetylcholinesterase activity (in vitro and in silico studies), while 7 and 10 showed specific targets (proteomic studies) directly related with the mussel adhesive structure (byssal threads), given by the alterations in the expression of Mytilus collagen proteins (PreCols) and proximal thread proteins (TMPs). A quantitative structure-activity relationship (QSAR) model was built with predictive capacity to enable speeding the design of new potential active compounds.


Subject(s)
Biofouling/prevention & control , Mytilus/drug effects , Xanthones/chemistry , Xanthones/toxicity , Animals , Biological Products/chemical synthesis , Biological Products/chemistry , Biological Products/toxicity , Larva/drug effects , Larva/physiology , Mytilus/physiology , Xanthones/chemical synthesis
9.
Planta Med ; 86(15): 1073-1079, 2020 Oct.
Article in English | MEDLINE | ID: mdl-32023632

ABSTRACT

We recently isolated the prenylated xanthones 2-deprenyl-rheediaxanthone B (XB) and 2-deprenyl-7-hydroxy-rheediaxanthone B (OH-XB) from the South American tree fern Metaxya rostrata. This study explores the mechanisms underlying the FoxM1 downregulation induced by both xanthones. Analysis of cell viability and cell-death induction in SW480, HCT116, Caco-2, DLD1 and HT29 exposed to xanthones found cell-loss and activation of caspase in all cell lines except HT29 that do not have high FoxM1 protein levels. To determine the cellular mechanism of xanthone-induced FoxM1 loss, protein stability was analyzed by cycloheximide-chase experiments and showed reduction of FoxM1 stability by XB but not OH-XB. Destabilization was prevented by inhibiting proteasome activity using MG-132 and moderately by the lysosomal inhibitor bafilomycin A1 (baf A1). OH-XB had a stronger impact than XB on FoxM1 mRNA expression by qRT-PCR, and MG-132 positively affected FoxM1 protein level in OH-XB exposed cells even though no decrease in protein abundance had been induced by the xanthone. Additionally, the compound inhibited topoisomerase I causing DNA DSB and early cell cycle arrest. This may reduce FoxM1 gene expression, which may in turn compromise DNA repair and enhance xanthone-induced cell death. With regard to xanthone-induced cell death, MG-132 protected cultures from cell loss induced by both compounds, and baf A1 was active against these XB-induced effects. In summary, both destabilization of FoxM1 protein and topoisomerase I inhibition contribute to both XB and OH-XB cytotoxic activity albeit at different ratios.


Subject(s)
DNA Topoisomerases, Type I , Xanthones , Caco-2 Cells , Cell Cycle Checkpoints , Cell Line, Tumor , Ferns/chemistry , Forkhead Box Protein M1/genetics , Humans , Xanthones/toxicity
10.
J Appl Toxicol ; 40(2): 234-244, 2020 02.
Article in English | MEDLINE | ID: mdl-31633820

ABSTRACT

Thioxanthone and its analogues, 2- or 4-isopropylthioxanthone, 2-chlorothioxanthone, 2,4-diethylthioxanthone (DETX) and xanthone, are used as photoinitiators of ultraviolet (UV) light-initiated curable inks. As these photoinitiators were found in numerous food/beverage products packaged in cartons printed with UV-cured inks, the cytotoxic effects and mechanisms of these compounds were studied in freshly isolated rat hepatocytes. The toxicity of DETX was greater than that of other compounds. DETX elicited not only concentration (0-2.0 mm)- and time (0-3 hours)-dependent cell death accompanied by the depletion of cellular adenosine triphosphate (ATP), and reduced glutathione (GSH) and protein thiol levels, but also the accumulation of GSH disulfide and malondialdehyde. Pretreatment of hepatocytes with either fructose at a concentration of 10 mm or N-acetyl-l-cysteine (NAC) at a concentration of 5.0 mm ameliorated DETX (1 mm)-induced cytotoxicity. Further, the exposure of hepatocytes to DETX resulted in the induction of reactive oxygen species (ROS) and loss of mitochondrial membrane potential, both of which were partially prevented by the addition of NAC. These results indicate that: (1) DETX-induced cytotoxicity is linked to mitochondrial failure and depletion of cellular GSH; (2) insufficient cellular ATP levels derived from mitochondrial dysfunction were, at least in part, ameliorated by the addition of fructose; and (3) GSH loss and/or ROS formation was prevented by NAC. Taken collectively, these results suggest that the onset of toxic effects caused by DETX may be partially attributable to cellular energy stress as well as oxidative stress.


Subject(s)
Cell Death/drug effects , Cell Survival/drug effects , Cells, Cultured/drug effects , Hepatocytes/drug effects , Light , Thioxanthenes/toxicity , Xanthones/toxicity , Animals , Rats , Rats, Inbred F344
11.
Eur J Med Chem ; 177: 362-373, 2019 Sep 01.
Article in English | MEDLINE | ID: mdl-31158750

ABSTRACT

Inhibiting the decomposition of carbohydrates into glucose or promoting glucose conversion is considered to be an effective treatment for type 2 diabetes. Herein, a series of novel xanthone-triazole derivatives were designed, synthesized, and their α-glucosidase inhibitory activities and glucose uptake in HepG2 cells were investigated. Most of the compounds showed better inhibitory activities than the parental compound a (1,3-dihydroxyxanthone, IC50 = 160.8 µM) and 1-deoxynojirimycin (positive control, IC50 = 59.5 µM) towards α-glucosidase. Compound 5e was the most potent inhibitor, with IC50 value of 2.06 µM. The kinetics of enzyme inhibition showed that compounds 5e, 5g, 5h, 6c, 6d, 6g and 6h were noncompetitive inhibitors, and molecular docking results were consistent with the noncompetitive property that these compounds bind to allosteric sites away from the active site (Asp214, Glu276 and Asp349). On the other hand, the glucose uptake assays exhibited that compounds 5e, 6a, 6c and 7g displayed high activities in promoting the glucose uptake. The cytotoxicity assays showed that most compounds were low-toxic to human normal hepatocyte cell line (LO2). These novel xanthone triazole derivatives exhibited dual therapeutic effects of α-glucosidase inhibition and glucose uptake promotion, thus they could be use as antidiabetic agents for developing novel drugs against type 2 diabetes.


Subject(s)
Glucose/metabolism , Glycoside Hydrolase Inhibitors/pharmacology , Hypoglycemic Agents/pharmacology , Triazoles/pharmacology , Xanthones/pharmacology , Binding Sites , Drug Design , Glycoside Hydrolase Inhibitors/chemical synthesis , Glycoside Hydrolase Inhibitors/metabolism , Glycoside Hydrolase Inhibitors/toxicity , Hep G2 Cells , Hepatocytes/drug effects , Humans , Hypoglycemic Agents/chemical synthesis , Hypoglycemic Agents/metabolism , Hypoglycemic Agents/toxicity , Kinetics , Molecular Docking Simulation , Molecular Structure , Protein Binding , Structure-Activity Relationship , Triazoles/chemical synthesis , Triazoles/metabolism , Triazoles/toxicity , Xanthones/chemical synthesis , Xanthones/metabolism , Xanthones/toxicity , alpha-Glucosidases/chemistry , alpha-Glucosidases/metabolism
12.
J Clin Invest ; 129(2): 546-555, 2019 02 01.
Article in English | MEDLINE | ID: mdl-30561388

ABSTRACT

Innate immune activation contributes to the transition from nonalcoholic fatty liver to nonalcoholic steatohepatitis (NASH). Stimulator of IFN genes (STING, also referred to Tmem173) is a universal receptor that recognizes released DNA and triggers innate immune activation. In this work, we investigated the role of STING in the progression of NASH in mice. Both methionine- and choline-deficient diet (MCD) and high-fat diet (HFD) were used to induce NASH in mice. Strikingly, STING deficiency attenuated steatosis, fibrosis, and inflammation in livers in both murine models of NASH. Additionally, STING deficiency increased fasting glucose levels in mice independently of insulin, but mitigated HFD-induced insulin resistance and weight gain and reduced levels of cholesterol, triglycerides, and LDL in serum; it also enhanced levels of HDL. The mitochondrial DNA (mtDNA) from hepatocytes of HFD-fed mice induced TNF-α and IL-6 expression in cultured Kupffer cells (KCs), which was attenuated by STING deficiency or pretreatment with BAY11-7082 (an NF-κB inhibitor). Finally, chronic exposure to 5,6-dimethylxanthenone-4-acetic acid (DMXAA, a STING agonist) led to hepatic steatosis and inflammation in WT mice, but not in STING-deficient mice. We proposed that STING functions as an mtDNA sensor in the KCs of liver under lipid overload and induces NF-κB-dependent inflammation in NASH.


Subject(s)
Dietary Fats/adverse effects , Hepatocytes/metabolism , Insulin Resistance , Kupffer Cells/metabolism , Membrane Proteins/metabolism , Mitochondria, Liver/metabolism , Animals , DNA, Mitochondrial/genetics , DNA, Mitochondrial/metabolism , Dietary Fats/pharmacology , Female , Hepatocytes/pathology , Humans , Inflammation/chemically induced , Inflammation/genetics , Inflammation/metabolism , Inflammation/pathology , Interleukin-6/genetics , Interleukin-6/metabolism , Kupffer Cells/pathology , Membrane Proteins/antagonists & inhibitors , Membrane Proteins/deficiency , Mice , Mice, Mutant Strains , Mitochondria, Liver/genetics , Mitochondria, Liver/pathology , Nitriles/pharmacology , Non-alcoholic Fatty Liver Disease , Oxygen Consumption/genetics , Sulfones/pharmacology , Tumor Necrosis Factor-alpha/genetics , Tumor Necrosis Factor-alpha/metabolism , Xanthones/toxicity
13.
Biomed Pharmacother ; 103: 708-718, 2018 07.
Article in English | MEDLINE | ID: mdl-29680739

ABSTRACT

This article has been retracted: please see Elsevier Policy on Article Withdrawal (https://www.elsevier.com/about/our-business/policies/article-withdrawal). This article has been retracted at the request of the Editor-in-Chief and the corresponding author as panels from Figure 2A appear as similar to panels from Figure 3A of the article published by Xiangyang Dou, Meihua Wang, Tao Zhang and Jiapei Yao in The Anatomical Record (2019) https://doi.org/10.1002/ar.24324 and Figure 2A of the article published by Qiang Wang, Yi Yan, Jie Zhang, Peng Guo, Yuqing Xing, Yong Wang, Fawei Qin and Qingyun Zeng in Biomedicine & Pharmacotherapy 104 (2018) 28-35 https://doi.org/10.1016/j.biopha.2018.05.013. Moreover, panels from Figure 2B appear as similar (after vertical mirroring) to panels from Figure 2A of the article published by Ning Wang, Fang Zhou, Jinhui Guo, Huaiyuan Zhu, Shanshui Luo and Jingjing Cao in Life Sciences 209 (2018) 498-506 https://doi.org/10.1016/j.lfs.2018.08.052. Also, panels from Figure 7C appear as similar to panels from Figure 8D of the article published by Zichao Li, Luying Zhang, Mingquan Gao, Mei Han, Kaili Liu, Zhuang Zhang, Zhi Gong, Lifei Xing, Xianzhou Shi, Kui Lu and Hui Gao in the Journal of Experimental & Clinical Cancer Research 38:8 (2019) https://doi.org/10.1186/s13046-018-1012-z. Although this article was published earlier than the other articles, the Editor decided to retract this article given the concerns on the reliability of the data. The corresponding author acknowledged that some of the TUNEL, Flow Cytometry and Immunohistochemistry (IHC) measurements have been provided by third party entities. One of the conditions of submission of a paper for publication is that authors declare explicitly that their work is original and has not appeared in a publication elsewhere. Re-use of any data should be appropriately cited. As such this article represents a severe abuse of the scientific publishing system. The scientific community takes a very strong view on this matter and apologies are offered to readers of the journal that this was not detected during the submission process.


Subject(s)
Apoptosis/drug effects , Autophagy/drug effects , Ovarian Neoplasms/pathology , Xanthones/toxicity , Animals , Apoptosis/physiology , Autophagy/physiology , Cell Line, Tumor , Cell Survival/drug effects , Cell Survival/physiology , Female , Humans , Male , Mice , Mice, Inbred BALB C , Mice, Nude , Ovarian Neoplasms/drug therapy , Random Allocation , Xanthones/therapeutic use , Xenograft Model Antitumor Assays/methods
14.
Cell Physiol Biochem ; 46(2): 829-846, 2018.
Article in English | MEDLINE | ID: mdl-29627822

ABSTRACT

BACKGROUND/AIMS: Gambogic acid (GA), the main active compound of Gamboge hanburyi, has been reported to be a potential novel antitumor drug. Whether GA inhibits putative cancer stem cells (CSCs), which are considered to be the major cause of cancer treatment failure, remains largely unknown. This study investigated whether GA inhibits the CSCs of colorectal cancer (CRC) and its possible mechanisms. METHODS: We performed CCK8 and tumor sphere formation assays, percentage analysis of both side population and CD133+CD44+ cells, and the detection of stem cells markers, in order to assess the role of GA in inhibiting the stem celllike features of CRC. An mRNA microarray was performed to identify the downstream gene affected by GA and rescue assays were performed to further clarify whether the downstream gene is involved in the GA induced decrease of the stem cell-like CRC population. CRC cells were engineered with a CSC detector vector encoding GFP and luciferase (Luc) under the control of the Nanog promoter, which were utilized to investigate the effect of GA on putative CSC in human tumor xenograft-bearing mice using in vivo bioluminescence imaging. RESULTS: Our results showed that GA significantly reduced tumor sphere formation and the percentages of side population and CD133+CD44+ cells, while also decreasing the expression of stemness and EMT-associated markers in CRC cells in vitro. GA killed stem-like CRC cells by upregulating the expression of ZFP36, which is dependent on the inactivation of the EGFR/ ERK signaling pathway. GFP+ cells harboring the PNanog-GFP-T2A-Luc transgene exhibited CSC characteristics. The in vivo results showed that GA significantly inhibited tumor growth in nude mice, accompanied by a remarkable reduction in the putative CSC number, based on whole-body bioluminescence imaging. CONCLUSION: These findings suggest that GA significantly inhibits putative CSCs of CRC both in vitro and in vivo by inhibiting the activation of the EGFR/ ERK/ZFP36 signaling pathway and may be an effective drug candidate for anticancer therapies.


Subject(s)
Apoptosis/drug effects , Tristetraprolin/metabolism , Up-Regulation/drug effects , Xanthones/toxicity , Animals , Cell Line, Tumor , Cell Proliferation/drug effects , Colorectal Neoplasms/drug therapy , Colorectal Neoplasms/metabolism , Colorectal Neoplasms/pathology , ErbB Receptors/metabolism , Extracellular Signal-Regulated MAP Kinases/metabolism , Fibronectins/genetics , Fibronectins/metabolism , HCT116 Cells , Humans , Mice , Mice, Nude , Nanog Homeobox Protein/genetics , Nanog Homeobox Protein/metabolism , Neoplastic Stem Cells/cytology , Neoplastic Stem Cells/drug effects , Neoplastic Stem Cells/metabolism , SOXB1 Transcription Factors/genetics , SOXB1 Transcription Factors/metabolism , Signal Transduction/drug effects , Transplantation, Heterologous , Tristetraprolin/genetics , Xanthones/therapeutic use
15.
Drug Deliv Transl Res ; 8(3): 617-632, 2018 06.
Article in English | MEDLINE | ID: mdl-29637488

ABSTRACT

The current research work encompasses the development, characterization, and evaluation of self-assembled phospholipidic nano-mixed miceller system (SPNMS) of a poorly soluble BCS Class IV xanthone bioactive, mangiferin (Mgf) functionalized with co-delivery of vitamin E TPGS. Systematic optimization using I-optimal design yielded self-assembled phospholipidic nano-micelles with a particle size of < 60 nm and > 80% of drug release in 15 min. The cytotoxicity and cellular uptake studies performed using MCF-7 and MDA-MB-231 cell lines demonstrated greater kill and faster cellular uptake. The ex vivo intestinal permeability revealed higher lymphatic uptake, while in situ perfusion and in vivo pharmacokinetic studies indicated nearly 6.6- and 3.0-folds augmentation in permeability and bioavailability of Mgf. In a nutshell, vitamin E functionalized SPNMS of Mgf improved the biopharmaceutical performance of Mgf in rats for enhanced anticancer potency.


Subject(s)
Micelles , Nanostructures/administration & dosage , Phospholipids/administration & dosage , Vitamin E/administration & dosage , Xanthones/administration & dosage , ATP Binding Cassette Transporter, Subfamily B, Member 1/metabolism , Animals , Cell Line, Tumor , Cell Survival/drug effects , Drug Liberation , Female , Humans , Jejunum/metabolism , Nanostructures/chemistry , Nanostructures/toxicity , Phospholipids/chemistry , Phospholipids/toxicity , Rats, Sprague-Dawley , Solubility , Vitamin E/chemistry , Vitamin E/toxicity , Xanthones/chemistry , Xanthones/toxicity
16.
Eur J Med Chem ; 148: 268-278, 2018 Mar 25.
Article in English | MEDLINE | ID: mdl-29466776

ABSTRACT

DNA topoisomerase I (Topo I) is an important anticancer drug target, and xanthone dimers are considered to be a new kind of Topo I inhibitor chemotypes. Based on the characteristics of dimeric xanthone structures, five new dimeric xanthones (1-5) and two known SAD isomers (6 and 7) were isolated from the mangrove-derived fungus Aspergillus vericolor. The absolute configurations of compounds 1-7, entailing both central and axial chirality elements, were established by a combination of ECD comparison, chemical conversions, and biogenetic considerations. Compounds 1-7 possessed high structural diversity and exhibited cytotoxicity at different levels. The selected new compounds 1, 2, and 5 showed Topo I inhibition properties and the most potent compound 1, an atropisomer of compound 2, was confirmed to inhibit Topo I-mediated DNA relaxation by targeting Topo I, thereby, arresting the cell cycle process and inducing necrosis in cancer cells. Molecular docking studies showed that compound 1 could bind DNA by π-π interaction and DNA Topo I by hydrogen bonds to form a ternary complex.


Subject(s)
Fungi/chemistry , Topoisomerase I Inhibitors/chemistry , Xanthones/pharmacology , Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacology , Aspergillus/chemistry , Cell Cycle Checkpoints/drug effects , Cell Line, Tumor , Dimerization , Drug Discovery , Drug Screening Assays, Antitumor , Humans , Molecular Docking Simulation , Molecular Structure , Necrosis/chemically induced , Xanthones/chemistry , Xanthones/toxicity
17.
Cell Death Dis ; 9(3): 286, 2018 02 19.
Article in English | MEDLINE | ID: mdl-29459714

ABSTRACT

Mitochondria are cellular organelles with crucial functions in the generation and distribution of ATP, the buffering of cytosolic Ca2+ and the initiation of apoptosis. Compounds that interfere with these functions are termed mitochondrial toxins, many of which are derived from microbes, such as antimycin A, oligomycin A, and ionomycin. Here, we identify the mycotoxin phomoxanthone A (PXA), derived from the endophytic fungus Phomopsis longicolla, as a mitochondrial toxin. We show that PXA elicits a strong release of Ca2+ from the mitochondria but not from the ER. In addition, PXA depolarises the mitochondria similarly to protonophoric uncouplers such as CCCP, yet unlike these, it does not increase but rather inhibits cellular respiration and electron transport chain activity. The respiration-dependent mitochondrial network structure rapidly collapses into fragments upon PXA treatment. Surprisingly, this fragmentation is independent from the canonical mitochondrial fission and fusion mediators DRP1 and OPA1, and exclusively affects the inner mitochondrial membrane, leading to cristae disruption, release of pro-apoptotic proteins, and apoptosis. Taken together, our results suggest that PXA is a mitochondrial toxin with a novel mode of action that might prove a useful tool for the study of mitochondrial ion homoeostasis and membrane dynamics.


Subject(s)
Mitochondria/drug effects , Mitochondrial Membranes/drug effects , Mycotoxins/toxicity , Xanthones/toxicity , Animals , Ascomycota/metabolism , Calcium/metabolism , Cell Line , Electron Transport/drug effects , Electron Transport Chain Complex Proteins/metabolism , Humans , Mice , Mitochondria/metabolism , Mitochondrial Membranes/metabolism , Mycotoxins/metabolism , Xanthones/metabolism
18.
Nat Prod Res ; 32(18): 2147-2151, 2018 Sep.
Article in English | MEDLINE | ID: mdl-28826239

ABSTRACT

Two naturally occurring xanthones, ananixanthone (1) and ß-mangostin (2), were isolated using column chromatographic method from the n-hexane and methanol extracts of Calophyllum teysmannii, respectively. The major constituent, ananixanthone (1), was subjected to structural modifications via acetylation, methylation and benzylation yielding four new xanthone derivatives, ananixanthone monoacetate (3), ananixanthone diacetate (4), 5-methoxyananixanthone (5) and 5-O-benzylananixanthone (6). Compound 1 together with its four new derivatives were subjected to MTT assay against three cancer cell lines; SNU-1, K562 and LS174T. The results indicated that the parent compound has greater cytotoxicity capabilities against SNU-1 and K562 cell lines with IC50 values of 8.97 ± 0.11 and 2.96 ± 0.06 µg/mL, respectively. Compound 5 on the other hand exhibited better cytotoxicity against LS174T cell line with an IC50 value of 5.76 ± 1.07 µg/mL.


Subject(s)
Calophyllum/chemistry , Xanthones/chemistry , Cell Death/drug effects , Cell Line, Tumor , Humans , Plant Extracts , Xanthones/isolation & purification , Xanthones/toxicity
19.
Cell Physiol Biochem ; 44(4): 1460-1470, 2017.
Article in English | MEDLINE | ID: mdl-29190630

ABSTRACT

BACKGROUND/AIMS: α-mangostin has anti-carcinogenic effects against several cancers. We investigated the molecular mechanism of this compound on the metastasis of human renal carcinoma cells. METHODS: Cell viability was measured using the MTT assay, and cell cycle distribution using flow cytometry. A Matrigel-based assay was used to measure in vitro cell migration and invasion. MAPK-related proteins and matrix metalloproteinase (MMP)-9 and MMP-2 expression were measured by western blotting, and MMP2/-9 activities were determined by gelatin zymography. RT-qPCR and a luciferase assay were used to examine the transcriptional activity of MMP-9. RESULTS: α-mangostin inhibited the migration and invasion of RCC cells in a dose-dependent manner, but had no evident cytotoxic effects. Treatment of 786-O cells with α-mangostin inhibited activation of MEK and ERK. Treatment with a specific MEK inhibitor (U0126) enhanced the inhibitory effects of α-mangostin on cell migration and invasion, and the phosphorylation of ERK and MEK. Moreover, α-mangostin inhibited the expression of the MMP-9 mRNA levels as well as the activity of MMP-9 promoter, and these suppressive effects were further enhanced by U0126. CONCLUSIONS: Our results suggest that α-mangostin suppresses cell migration and invasion via MEK/ERK/MMP9 pathway, and might be a promising anti-metastatic agent against human renal cell carcinoma.


Subject(s)
Anticarcinogenic Agents/toxicity , Extracellular Signal-Regulated MAP Kinases/metabolism , MAP Kinase Kinase Kinases/metabolism , Matrix Metalloproteinase 9/metabolism , Signal Transduction/drug effects , Transcription, Genetic/drug effects , Xanthones/toxicity , Anticarcinogenic Agents/chemistry , Butadienes/pharmacology , Cell Line, Tumor , Cell Movement/drug effects , Cell Survival/drug effects , Humans , Kidney Neoplasms/metabolism , Kidney Neoplasms/pathology , Matrix Metalloproteinase 2/metabolism , Matrix Metalloproteinase 9/genetics , Nitriles/pharmacology , Xanthones/chemistry
20.
Cell Physiol Biochem ; 44(4): 1381-1395, 2017.
Article in English | MEDLINE | ID: mdl-29186708

ABSTRACT

BACKGROUND/AIMS: Glioma is the most devastating cancer in the brain and has a poor prognosis in adults. Therefore, there is a critical need for novel therapeutic strategies for the management of glioma patients. Isogambogenic acid, an active compound extracted from the Chinese herb Garcinia hanburyi, induces autophagic cell death. METHODS: Cell viability was detected with MTT assays. Cell proliferation was assessed using the colony formation assay. Morphological changes associated with autophagy and apoptosis were tested by TEM and Hoechst staining, respectively. The apoptosis rate was measured by flow cytometry. Western blot, immunofluorescence and immunohistochemical analyses were used to detect protein expression. U87-derived xenografts were established for the examination of the effect of isogambogenic acid on glioma growth in vivo. RESULTS: Isogambogenic acid induced autophagic death in U87 and U251 cells, and blocking late-stage autophagy markedly enhanced the antiproliferative activities of isogambogenic acid. Moreover, we observed the activation of AMPK-mTOR signalling in isogambogenic acid-treated glioma cells. Furthermore, the activation of AMPK or the inhibition of mTOR augmented isogambogenic acid-induced autophagy. Inhibition of autophagy attenuated apoptosis in isogambogenic acid-treated glioma cells. Finally, isogambogenic acid inhibited the growth of U87 glioma in vivo. CONCLUSION: Isogambogenic acid inhibits the growth of glioma via activation of the AMPK-mTOR signalling pathway, which may provide evidence for future clinical applications in glioma therapy.


Subject(s)
AMP-Activated Protein Kinases/metabolism , Antineoplastic Agents/toxicity , Cell Proliferation/drug effects , TOR Serine-Threonine Kinases/metabolism , Xanthones/toxicity , Adenine/analogs & derivatives , Adenine/pharmacology , Animals , Antineoplastic Agents/chemistry , Antineoplastic Agents/therapeutic use , Apoptosis/drug effects , Autophagy/drug effects , Brain Neoplasms/drug therapy , Brain Neoplasms/pathology , Cell Cycle Checkpoints/drug effects , Cell Line, Tumor , Glioma/drug therapy , Glioma/pathology , Humans , Immunohistochemistry , Mice , Mice, Inbred BALB C , Mice, Nude , Signal Transduction/drug effects , Transplantation, Heterologous , Xanthones/chemistry , Xanthones/therapeutic use
SELECTION OF CITATIONS
SEARCH DETAIL