Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 18 de 18
1.
Food Chem Toxicol ; 187: 114631, 2024 May.
Article En | MEDLINE | ID: mdl-38570025

Toosendanin (TSN) is the main active compound derived from Melia toosendan Sieb et Zucc with various bioactivities. However, liver injury was observed in TSN limiting its clinical application. Lipid metabolism plays a crucial role in maintaining cellular homeostasis, and its disruption is also essential in TSN-induced hepatotoxicity. This study explored the hepatotoxicity caused by TSN in vitro and in vivo. The lipid droplets were significantly decreased, accompanied by a decrease in fatty acid transporter CD36 and crucial enzymes in the lipogenesis including ACC and FAS after the treatment of TSN. It was suggested that TSN caused lipid metabolism disorder in hepatocytes. TOFA, an allosteric inhibitor of ACC, could partially restore cell survival via blocking malonyl-CoA accumulation. Notably, TSN downregulated the LXRα/Lipin1/SREBP1 signaling pathway. LXRα activation improved cell survival and intracellular neutral lipid levels, while SREBP1 inhibition aggravated the cell damage and caused a further decline in lipid levels. Male Balb/c mice were treated with TSN (5, 10, 20 mg/kg/d) for 7 days. TSN exposure led to serum lipid levels aberrantly decreased. Moreover, the western blotting results showed that LXRα/Lipin1/SREBP1 inhibition contributed to TSN-induced liver injury. In conclusion, TSN caused lipid metabolism disorder in liver via inhibiting LXRα/Lipin1/SREBP1 signaling pathway.


Chemical and Drug Induced Liver Injury , Drugs, Chinese Herbal , Lipid Metabolism Disorders , Triterpenes , Mice , Animals , Male , Lipid Metabolism , Drugs, Chinese Herbal/pharmacology , Chemical and Drug Induced Liver Injury/etiology , Lipids
2.
J Ethnopharmacol ; 330: 118196, 2024 Aug 10.
Article En | MEDLINE | ID: mdl-38631488

ETHNOPHARMACOLOGICAL RELEVANCE: Rosmarinic acid (RA), a natural polyphenol abundant in numerous herbal remedies, has been attracting growing interest owing to its exceptional ability to protect the liver. Toosendanin (TSN), a prominent bioactive compound derived from Melia toosendan Siebold & Zucc., boasts diverse pharmacological properties. Nevertheless, TSN possesses remarkable hepatotoxicity. Intriguingly, the potential of RA to counteract TSN-induced liver damage and its probable mechanisms remain unexplored. AIM OF THE STUDY: This study is aimed at exploring whether RA can alleviate TSN-induced liver injury and the potential mechanisms involved autophagy. MATERIALS AND METHODS: CCK-8 and LDH leakage rate assay were used to evaluate cytotoxicity. Balb/c mice were intraperitoneally administered TSN (20 mg/kg) for 24 h after pretreatment with RA (0, 40, 80 mg/kg) by gavage for 5 days. The autophagic proteins P62 and LC3B expressions were detected using western blot and immunohistochemistry. RFP-GFP-LC3B and transmission electron microscopy were applied to observe the accumulation levels of autophagosomes and autolysosomes. LysoTracker Red and DQ-BSA staining were used to evaluate the lysosomal acidity and degradation ability respectively. Western blot, immunohistochemistry and immunofluorescence staining were employed to measure the expressions of JAK2/STAT3/CTSC pathway proteins. Dual-luciferase reporter gene was used to measure the transcriptional activity of CTSC and RT-PCR was used to detect its mRNA level. H&E staining and serum biochemical assay were employed to determine the degree of damage to the liver. RESULTS: TSN-induced damage to hepatocytes and livers was significantly alleviated by RA. RA markedly diminished the autophagic flux blockade and lysosomal dysfunction caused by TSN. Mechanically, RA alleviated TSN-induced down-regulation of CTSC by activating JAK2/STAT3 signaling pathway. CONCLUSION: RA could protect against TSN-induced liver injury by activating the JAK2/STAT3/CTSC pathway-mediated autophagy and lysosomal function.


Autophagy , Chemical and Drug Induced Liver Injury , Cinnamates , Depsides , Janus Kinase 2 , Lysosomes , Rosmarinic Acid , STAT3 Transcription Factor , Signal Transduction , Animals , Humans , Male , Mice , Autophagy/drug effects , Chemical and Drug Induced Liver Injury/drug therapy , Chemical and Drug Induced Liver Injury/metabolism , Chemical and Drug Induced Liver Injury/pathology , Chemical and Drug Induced Liver Injury/prevention & control , Cinnamates/pharmacology , Depsides/pharmacology , Drugs, Chinese Herbal/pharmacology , Janus Kinase 2/metabolism , Liver/drug effects , Liver/metabolism , Liver/pathology , Lysosomes/drug effects , Lysosomes/metabolism , Mice, Inbred BALB C , Signal Transduction/drug effects , STAT3 Transcription Factor/metabolism
3.
Toxicol Lett ; 394: 102-113, 2024 Apr.
Article En | MEDLINE | ID: mdl-38460807

Toosendanin (TSN) is the main active component in the traditional herb Melia toosendan Siebold & Zucc, which exhibits promising potential for development due to its diverse pharmacological properties. However, the hepatotoxicity associated with TSN needs further investigation. Previous research has implicated autophagy dysregulation in TSN-induced hepatotoxicity, yet the underlying mechanisms remain elusive. In this study, the mechanisms of signal transducer and activator of transcription 3 (STAT3) in TSN-induced autophagy inhibition and liver injury were explored using Stat3 knockout C57BL/6 mice and HepG2 cells. TSN decreased cell viability, increased lactate dehydrogenase (LDH) production in vitro, and elevated serum aspartate transaminase (AST) and alanine aminotransferase (ALT) levels as well as liver lesions in vivo, suggesting TSN had significant hepatotoxicity. TSN inhibited Janus kinase 2 (JAK2)/STAT3 pathway and the expression of cathepsin C (CTSC). Inhibition of STAT3 exacerbated TSN-induced autophagy inhibition and hepatic injury, whereas activation of STAT3 attenuated these effects of TSN. Mechanistically, STAT3 transcriptionally regulated the level of CTSC gene, which in turn affected autophagy and the process of liver injury. TSN-administered Stat3 knockout mice showed more severe hepatotoxicity, CTSC downregulation, and autophagy blockade than wildtype mice. In summary, TSN caused hepatotoxicity by inhibiting STAT3/CTSC axis-dependent autophagy and lysosomal function.


Chemical and Drug Induced Liver Injury , Drugs, Chinese Herbal , Triterpenes , Animals , Mice , STAT3 Transcription Factor/metabolism , Cathepsin C/metabolism , Mice, Inbred C57BL , Drugs, Chinese Herbal/pharmacology , Autophagy
4.
Toxicol Lett ; 377: 51-61, 2023 Mar 15.
Article En | MEDLINE | ID: mdl-36801351

Toosendanin (TSN) is the main active compound of Melia toosendan Sieb et Zucc with various bioactivities. In this study, we investigated the role of ferroptosis in TSN-induced hepatotoxicity. The characteristic indicators of ferroptosis were detected including reactive oxygen species (ROS), lipid-ROS, glutathione (GSH), ferrous ion and the expression of glutathione peroxidase 4 (GPX4), which showed that TSN caused ferroptosis in hepatocytes. The results of qPCR analysis and western blotting assay showed that TSN-induced activation of protein kinase R-like endoplasmic reticulum kinase (PERK)- eukaryotic initiation factor 2 α subunit (eIF2α)- activation transcription factor 4 (ATF4) signaling pathway resulted in increasing activation transcription factor 3 (ATF3) expression, which upregulated the expression of transferrin receptor 1 (TFRC). Furthermore, TFRC mediated iron accumulation leading to ferroptosis in hepatocytes. To clarify whether TSN triggered ferroptosis in vivo, male Balb/c mice were treated with the different doses of TSN. The results of hematoxylin-eosin (H&E) staining, 4-hydroxynonenal (4-HNE) staining, malondialdehyde (MDA) content and the protein expression of GPX4 showed that ferroptosis contributed to TSN-induced hepatotoxicity. Iron homeostasis relative protein and PERK- eIF2α- ATF4 signaling pathway also involved in hepatotoxicity of TSN in vivo.


Chemical and Drug Induced Liver Injury , Drugs, Chinese Herbal , Ferroptosis , Animals , Mice , Male , Eukaryotic Initiation Factor-2/metabolism , Reactive Oxygen Species/metabolism , Transcription Factor 4 , Activating Transcription Factor 4/genetics , Activating Transcription Factor 4/metabolism
5.
Pharmaceuticals (Basel) ; 15(12)2022 Dec 03.
Article En | MEDLINE | ID: mdl-36558960

Toosendanin (TSN) is a triterpenoid from the fruit or bark of Melia toosendan Sieb et Zucc, which has clear antitumor and insecticidal activities, but it possesses limiting hepatotoxicity in clinical application. Autophagy is a degradation and recycling mechanism to maintain cellular homeostasis, and it also plays an essential role in TSN-induced hepatotoxicity. Nevertheless, the specific mechanism of TSN on autophagy-related hepatotoxicity is still unknown. The hepatotoxicity of TSN in vivo and in vitro was explored in this study. It was found that TSN induced the upregulation of the autophagy-marker microtubule-associated proteins 1A/1B light chain 3B (LC3B) and P62, the accumulation of autolysosomes, and the inhibition of autophagic flux. The middle and late stages of autophagy were mainly studied. The data showed that TSN did not affect the fusion of autophagosomes and lysosomes but significantly inhibited the acidity, the degradation capacity of lysosomes, and the expression of hydrolase cathepsin B (CTSB). The activation of autophagy could alleviate TSN-induced hepatocyte damage. TSN inhibited the expression of transcription factor EB (TFEB), which is a key transcription factor for many genes of autophagy and lysosomes, such as CTSB, and overexpression of TFEB alleviated the autophagic flux blockade caused by TSN. In summary, TSN caused hepatotoxicity by inhibiting TFEB-lysosome-mediated autophagic flux and activating autophagy by rapamycin (Rapa), which could effectively alleviate TSN-induced hepatotoxicity, indicating that targeting autophagy is a new strategy to intervene in the hepatotoxicity of TSN.

6.
Toxicol Appl Pharmacol ; 425: 115606, 2021 08 15.
Article En | MEDLINE | ID: mdl-34087332

Triptolide (TP), a primary bioactive ingredient isolated from the traditional Chinese herbal medicine Tripterygium wilfordii Hook. F. (TWHF), has attracted great interest for its therapeutic biological activities in inflammation and autoimmune disease. However, its clinical use is limited by severe testicular toxicity, and the underlying mechanism has not been elucidated. Our preliminary evidence demonstrated that TP disrupted glucose metabolism and caused testicular toxicity. During spermatogenesis, Sertoli cells (SCs) provide lactate as an energy source to germ cells by glycolysis. The transcription factors GATA-binding protein 4 (GATA4) and specificity protein 1 (Sp1) can regulate glycolysis. Based on this evidence, we speculate that TP causes abnormal glycolysis in SCs by influencing the expression of the transcription factors GATA4 and Sp1. The mechanism of TP-induced testicular toxicity was investigated in vitro and in vivo. The data indicated that TP decreased glucose consumption, lactate production, and the mRNA levels of glycolysis-related transporters and enzymes. TP also downregulated the protein expression of the transcription factors GATA4 and Sp1, as well as the glycolytic enzyme phosphofructokinase platelet (PFKP). Phosphorylated GATA4 and nuclear GATA4 protein levels were reduced in a dose- and time-dependent manner after TP incubation. Similar effects were observed in shGata4-treated TM4 cells and BALB/c mice administered 0.4 mg/kg TP for 28 days, and glycolysis was also inhibited. Gata4 knockdown downregulated Sp1 and PFKP expression. Furthermore, the Sp1 inhibitor plicamycin inhibited PFKP protein levels in TM4 cells. In conclusion, TP inhibited GATA4-mediated glycolysis by suppressing Sp1-dependent PFKP expression in SCs and caused testicular toxicity.


Diterpenes/pharmacology , GATA4 Transcription Factor/metabolism , Glycolysis/drug effects , Phenanthrenes/pharmacology , Phosphofructokinase-1, Type C/metabolism , Sertoli Cells/drug effects , Sp1 Transcription Factor/metabolism , Animals , Cell Line , Cell Proliferation , Cell Survival/drug effects , Down-Regulation , Epoxy Compounds/pharmacology , GATA4 Transcription Factor/drug effects , GATA4 Transcription Factor/genetics , Gene Expression Regulation/drug effects , HEK293 Cells , Humans , Male , Mice , Mice, Inbred ICR , Phosphofructokinase-1, Type C/drug effects , Phosphofructokinase-1, Type C/genetics , Sertoli Cells/metabolism , Signal Transduction/drug effects , Sp1 Transcription Factor/drug effects , Sp1 Transcription Factor/genetics
7.
Toxicol Lett ; 318: 1-11, 2020 Jan.
Article En | MEDLINE | ID: mdl-31618665

Triptolide (TP), a principal bioactive component extracted from traditional Chinese medicine Tripterygium wilfordii Hook. F. (TWHF), has attracted wide attention of its therapeutic effects on inflammation and autoimmune diseases. However, the therapeutic application of TP is hindered by severe cardiomyocyte toxicity and narrow therapeutic window. We previously identified that the p53 was an indispensable contributor in TP-induced myocardial injury. p53 has an inhibitory effect on IKKß-NF-κB pathway that regulates glucose transporters (GLUT) expression. Based on these evidences, we speculate that p53 mediates TP-disturbed glucose uptake by blocking IKKß-NF-κB signaling. This study focused on the effect of TP on cardiac glucose uptake and the role of p53 in glucose metabolism in cardiomyocytes, and p53 -/- mice. TP treatment depressed glucose consumption and ATP production resulting in myocardial damage. Incubation with ATP (5 mM) remarkably decreased the cellular damage. Immunoblotting and immunofluorescence identified that TP suppressed glucose uptake by restricting IKKß-NF-κB signaling activation, GLUT1 and GLUT4 expression. p53 inhibition alleviated the cell damage and the compromise of glucose uptake. Mechanistically, p53 antagonist PFTα abolished TP-induced the inhibition of IKKß, IκBα phosphorylation, p65 nuclear translocation, and GLUT1, GLUT4 expression. Consistently, in acute heart injury models, p53 deficiency upregulated IKKß-NF-κB activation and GLUT1, GLUT4 protein levels which was also indicated as amelioration of heart histological injury after 1.2 mg kg-1 TP administration. The present findings indicate that TP-induced p53 overactivation suppresses glucose uptake by inhibiting IKKß-NF-κB pathway and downregulating NF-κB-dependent GLUT1 and GLUT4 expression.


Diterpenes/toxicity , Glucose/metabolism , Heart Diseases/chemically induced , I-kappa B Kinase/metabolism , Myocytes, Cardiac/drug effects , NF-kappa B/metabolism , Phenanthrenes/toxicity , Tumor Suppressor Protein p53/metabolism , Animals , Apoptosis/drug effects , Cardiotoxicity , Cell Line , Energy Metabolism/drug effects , Epoxy Compounds/toxicity , Glucose Transporter Type 1/genetics , Glucose Transporter Type 1/metabolism , Glucose Transporter Type 4/genetics , Glucose Transporter Type 4/metabolism , Heart Diseases/genetics , Heart Diseases/metabolism , Heart Diseases/pathology , Mice, Knockout , Myocytes, Cardiac/metabolism , Myocytes, Cardiac/pathology , Rats, Sprague-Dawley , Signal Transduction/drug effects , Tumor Suppressor Protein p53/deficiency , Tumor Suppressor Protein p53/genetics
8.
Toxicol In Vitro ; 58: 187-194, 2019 Aug.
Article En | MEDLINE | ID: mdl-30926361

Triptolide (TP) isolated from Tripterygium wilfordii Hook F. (TWHF) shows extensive anti-inflammation, immunosuppression and anti-tumor properties. However, its therapeutic potential is limited by its severe side effects, especially the nephrotoxicity. This study intended to explore the role of the GSK-3ß/Fyn pathway in TP-induced oxidative damage and the potential mechanism of Nrf2 protein downregulation. Our data showed that TP induced oxidative stress and cell damage in the rat renal tubular epithelial cell line NRK-52E cells by activation of GSK-3ß and nuclear translocation of Fyn, which resulted in decreased Nrf2 nuclear translocation. Moreover, TP significantly induced Nrf2 degradation by ubiquitination, which was blocked by the proteasome inhibitor MG132. In addition, cotreatment with a typical GSK-3ß inhibitor, lithium chloride, promoted the nuclear translocation of Nrf2 and decreased the nuclear translocation of Fyn, which led to reduced cell damage, LDH leakage, glutathione depletion and cell apoptosis. Collectively, our results indicated that TP induced oxidative damage in NRK-52E cells by facilitating Nrf2 degradation by ubiquitination via the GSK-3ß/Fyn pathway.


Antineoplastic Agents, Alkylating/toxicity , Diterpenes/toxicity , Glycogen Synthase Kinase 3 beta/metabolism , Immunosuppressive Agents/toxicity , NF-E2-Related Factor 2/metabolism , Phenanthrenes/toxicity , Proto-Oncogene Proteins c-fyn/metabolism , Animals , Apoptosis/drug effects , Cell Line , Epoxy Compounds/toxicity , Glutathione/metabolism , Oxidative Stress/drug effects , Rats , Reactive Oxygen Species/metabolism , Signal Transduction/drug effects , Ubiquitination/drug effects
9.
Toxicol Lett ; 300: 105-115, 2019 Jan.
Article En | MEDLINE | ID: mdl-30394310

Triptolide (TP) is the main ingredient of Chinese herb Tripterygium wilfordii Hook f. (TWHF). Despite of its multifunction in pharmaceutics, accumulating evidences showed that TP caused obvious hepatotoxicity in clinic. The current study investigated the role of Notch1 signaling in TP-induced hepatotoxicity. Our data indicated that TP inhibited the protein expression of Notch1 and its active form Notch intracellular domain (NICD) leading to increased PTEN (phosphatase and tensin homolog deleted on chromosome ten) expression. Moreover, PTEN triggered Txnip (thioredoxin-interacting protein) activation by inhibiting Akt phosphorylation, which resulted in reduction of Trx (thioredoxin). In conclusion, TP caused liver injury through initiating oxidative stress in hepatocyte. This study indicated the potency of Notch1 to protect against TP-induced hepatotoxicity.


Diterpenes/toxicity , Hep G2 Cells/drug effects , Hepatocytes/drug effects , PTEN Phosphohydrolase/drug effects , Phenanthrenes/toxicity , Receptor, Notch1/metabolism , Signal Transduction/drug effects , Thioredoxins/drug effects , Epoxy Compounds/toxicity , Hepatocytes/metabolism , Humans , Neurotoxicity Syndromes/metabolism , PTEN Phosphohydrolase/metabolism , Thioredoxins/metabolism , Tripterygium/chemistry
10.
Toxicol Appl Pharmacol ; 355: 269-285, 2018 09 15.
Article En | MEDLINE | ID: mdl-30009776

Triptolide (TP), a major active component of Tripterygium wilfordii Hook f., is widely used in the treatment of inflammation and autoimmune disorders. Its clinical application is limited by severe adverse effects, especially cardiotoxicity. Accumulative evidences indicate that TP induces DNA damage by inhibiting RNA polymerase. Considering the relationship among DNA damage, p53, and the role of p53 in mitochondria-dependent apoptosis, we speculate that TP-induced cardiotoxicity results from p53 activation. In this study, the role of p53 in TP-induced cardiotoxicity was investigated in H9c2 cells, primary cardiomyocytes, and C57BL/6 genetic background p53-/- mice. p53 protein level was elevated by TP in vitro and in acute heart injury models. With TP administration (1.2 mg/kg), p53 deficiency prevented heart histology injury and decreased serum cardiac troponin I (cTn-I) and apoptotic proteins. Mechanistically, immunoblotting and immunofluorescence staining identified that TP-induced toxicity is dependent on p53 nuclear translocation and transactivation of Bcl2 family genes, leading to mitochondrial outer membrane permeabilization (MOMP) and mitochondria dysfunction. Consistently, p53 antagonist PFTα counteracted TP-induced p53 overexpression and regulation of Bcl2 family transcription, which improved mitochondrial membrane integrity and prevented apoptosis. Moreover, Bax antagonist Bax inhibitor peptide (BIP) V5 ameliorated TP-induced apoptosis through suppressing membrane depolarization and ROS accumulation. These results suggest that TP-induced cardiotoxicity is p53-dependent by promoting Bax-induced mitochondria-mediated apoptosis.


Anti-Inflammatory Agents, Non-Steroidal/toxicity , Diterpenes/toxicity , Heart Diseases/chemically induced , Membrane Potential, Mitochondrial/drug effects , Mitochondria, Heart/drug effects , Myocytes, Cardiac/drug effects , Phenanthrenes/toxicity , Tumor Suppressor Protein p53/metabolism , Animals , Apoptosis/drug effects , Cell Line , Diterpenes/antagonists & inhibitors , Epoxy Compounds/antagonists & inhibitors , Epoxy Compounds/toxicity , Heart Diseases/pathology , Mice , Mice, Inbred C57BL , Mice, Knockout , Myocardium/pathology , Phenanthrenes/antagonists & inhibitors , Reactive Oxygen Species/metabolism , Tumor Suppressor Protein p53/deficiency , Tumor Suppressor Protein p53/genetics , bcl-2-Associated X Protein/antagonists & inhibitors
11.
Toxicol Lett ; 292: 136-150, 2018 Aug.
Article En | MEDLINE | ID: mdl-29723566

Triptolide is a major active ingredient of tripterygium glycosides, used for the therapy of immune and inflammatory diseases. However, its clinical applications are limited by severe male fertility toxicity associated with decreased sperm count, mobility and testicular injures. In this study, we determined that triptoide-induced mitochondrial dysfunction triggered reduction of lactate and dysregulation of fatty acid metabolism in mouse Sertoli cells. First, triptolide induced mitochondrial damage through the suppressing of proliferator-activated receptor coactivator-1 alpha (PGC-1α) activity and protein. Second, mitochondrial damage decreased lactate production and dysregulated fatty acid metabolism. Finally, mitochondrial dysfunction was initiated by the inhibition of sirtuin 1 (SIRT1) with the regulation of AMP-activated protein kinase (AMPK) in Sertoli cells after triptolide treatment. Meanwhile, triptolide induced mitochondrial fatty acid oxidation dysregulation by increasing AMPK phosphorylation. Taken together, we provide evidence that the mechanism of triptolide-induced testicular toxicity under mitochondrial injury may involve a metabolic change.


Anti-Inflammatory Agents/toxicity , Diterpenes/toxicity , Energy Metabolism/drug effects , Fatty Acids/metabolism , Immunosuppressive Agents/toxicity , Mitochondria/drug effects , Phenanthrenes/toxicity , Sertoli Cells/drug effects , AMP-Activated Protein Kinases/metabolism , Animals , Cell Line , Dose-Response Relationship, Drug , Energy Metabolism/genetics , Epoxy Compounds/toxicity , Gene Expression Regulation, Enzymologic , Lactic Acid/metabolism , Male , Mice, Inbred ICR , Mitochondria/metabolism , Mitochondria/pathology , Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha/metabolism , Phosphorylation , Sertoli Cells/metabolism , Sertoli Cells/pathology , Signal Transduction/drug effects , Sirtuin 1/genetics , Sirtuin 1/metabolism , Time Factors
12.
Toxicol Appl Pharmacol ; 342: 1-13, 2018 03 01.
Article En | MEDLINE | ID: mdl-29407771

Burkitt's lymphoma (BL) is a highly aggressive B-cell non-Hodgkin lymphoma with rapid growth and dissemination propensity. Triptolide (TP), an active component extracted from Chinese herb Tripterygium wilfordii Hook f., has broad-spectrum anti-tumor activities. This study aimed to explore the in vitro and in vivo anti-cancer effects of TP on BL and the potential molecular mechanisms. In this study, the in vitro anti-tumor activity of TP was determined by CCK-8 and flow cytometry assays in Raji, NAMALWA and Daudi cells. The expression of SIRT3, phosphorylation and acetylation of glycogen synthase kinase-3ß (GSK-3ß) were analyzed by Western blot assay. Moreover, we examined the mitochondrial membrane potential by JC-1 method and measured apoptosis related protein using Western blot assay. BL xenograft model in NOD/SCID mice were established to evaluate the in vivo anti-cancer effect of TP. We discovered that TP inhibited BL cell growth and induced apoptosis in a dose-dependent manner. Loss of SIRT3 provides growth advances for BL cells. However, TP could up-regulate SIRT3 expression, which resulted in suppression of BL cells proliferation. GSK-3ß was activated by SIRT3-mediated deacetylation, which subsequently induced mitochondrial translocation and accumulation of Bax and decrease of mitochondrial membrane potential. Anti-tumor studies in vivo showed that TP (0.36 mg/kg) inhibited the growth of BL xenografts in NOD/SCID mice with an inhibitory rate of 73.13%. Our data revealed that TP triggered mitochondrial apoptotic pathway in BL by increasing SIRT3 expression and activating SIRT3/GSK-3ß/Bax pathway. This study indicated that TP is a potential anti-cancer Chinese herbal medicine against BL.


Antineoplastic Agents/pharmacology , Burkitt Lymphoma/metabolism , Diterpenes/pharmacology , Glycogen Synthase Kinase 3 beta/metabolism , Phenanthrenes/pharmacology , Sirtuin 3/metabolism , Acetylation , Animals , Antineoplastic Agents/therapeutic use , Apoptosis/drug effects , Burkitt Lymphoma/drug therapy , Burkitt Lymphoma/pathology , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Survival/drug effects , Diterpenes/therapeutic use , Epoxy Compounds/pharmacology , Epoxy Compounds/therapeutic use , Humans , Male , Membrane Potential, Mitochondrial/drug effects , Mice, Inbred NOD , Mice, SCID , Mitochondria/drug effects , Mitochondria/physiology , Phenanthrenes/therapeutic use , Tumor Burden/drug effects , bcl-2-Associated X Protein/metabolism
13.
Toxicol Appl Pharmacol ; 313: 195-203, 2016 Dec 15.
Article En | MEDLINE | ID: mdl-27751939

Triptolide (TP), a diterpene triepoxide, is a major active component of Tripterygium wilfordii extracts, which are prepared as tablets and has been used clinically for the treatment of inflammation and autoimmune disorders. However, TP's therapeutic potential is limited by severe adverse effects. In a previous study, we reported that TP induced mitochondria dependent apoptosis in cardiomyocytes. Glycogen synthase kinase-3ß (GSK-3ß) is a multifunctional serine/threonine kinase that plays important roles in the necrosis and apoptosis of cardiomyocytes. Our study aimed to investigate the role of GSK-3ß in TP-induced cardiotoxicity. Inhibition of GSK-3ß activity by SB 216763, a potent and selective GSK-3 inhibitor, prominently ameliorated the detrimental effects in C57BL/6J mice with TP administration, which was associated with a correction of GSK-3ß overactivity. Consistently, in TP-treated H9c2 cells, SB 216763 treatment counteracted GSK-3ß overactivity, improved cell viability, and prevented apoptosis by modulating the expression of Bcl-2 family proteins. Mechanistically, GSK-3ß interacted with and phosphorylated cyclophilin F (Cyp-F), a key regulator of mitochondrial permeability transition pore (mPTP). GSK-3ß inhibition prevented the phosphorylation and activation of Cyp-F, and desensitized mPTP. Our findings suggest that pharmacological targeting of GSK-3ß could represent a promising therapeutic strategy for protecting against cardiotoxicity induced by TP.


Diterpenes/toxicity , Glycogen Synthase Kinase 3 beta/antagonists & inhibitors , Heart Injuries/prevention & control , Mitochondrial Membrane Transport Proteins/drug effects , Phenanthrenes/toxicity , Animals , Cell Line , Epoxy Compounds/toxicity , Heart Injuries/chemically induced , Male , Mice , Mice, Inbred C57BL , Mitochondrial Permeability Transition Pore
14.
Toxicol In Vitro ; 34: 128-137, 2016 Aug.
Article En | MEDLINE | ID: mdl-27064125

Triptolide (TP), an active component of the traditional Chinese herb Tripterygium wilfordii Hook f. (TWHF), has multiple pharmacological effects. However, the severe toxicity of TP greatly restricts its clinical applications. Although TP exposure causes serious heart injury, the mechanism underlying TP-induced cardiotoxicity has rarely been investigated. In previous studies, we found that TP-induced oxidative stress was involved in the mitochondria-dependent apoptosis of cardiomyocytes. Opening of the mitochondrial permeability transition pore (mPTP) is the key to the mitochondrial dysfunction in cardiac toxicity. The aim of this study was to investigate the potential cardioprotective effects of sirtuin 3 (SIRT3) on the mPTP. In the present study, the cytotoxicity of TP was accompanied by the up-regulation of the SIRT3 protein level and its rapid aggregation in nuclei and mitochondria. The SIRT3-FOXO3 signaling pathway was activated simultaneously, resulting in increased transcription of manganese superoxide dismutase (MnSOD) and catalase (CAT) for the elimination of reactive oxygen species (ROS). In addition, augmentation of the SIRT3 level via the overexpression plasmid SIRT3-Flag provided resistance to TP-induced cellular damage, whereas knocking down the SIRT3 level via siRNA accelerated the damage. Because it is an activator of SIRT3, the protective effect of resveratrol was also evaluated in H9c2 cells. In conclusion, the current results suggest that activation of SIRT3 substantially ameliorates the detrimental effects of TP by closing the mPTP.


Diterpenes/toxicity , Mitochondrial Membrane Transport Proteins/metabolism , Myocytes, Cardiac/drug effects , Phenanthrenes/toxicity , Sirtuins/metabolism , Animals , Apoptosis/drug effects , Catalase/metabolism , Cell Line , Cells, Cultured , Epoxy Compounds/toxicity , Forkhead Box Protein O3/metabolism , Membrane Potential, Mitochondrial/drug effects , Mitochondrial Permeability Transition Pore , Myocytes, Cardiac/metabolism , Rats, Sprague-Dawley , Reactive Oxygen Species/metabolism , Superoxide Dismutase/metabolism
15.
Pharmazie ; 71(9): 514-523, 2016 Sep 01.
Article En | MEDLINE | ID: mdl-29441847

Clinical application of triptolide (TP), a main active ingredient of the traditional Chinese herb Tripterygium wilfordii Hook f. (TWHF), is limited by a series of severe toxicities, including cardiotoxicity. In previous studies, we found the activation of sirtuin 3 (SIRT3) attenuated TP-induced toxicity in cardiomyocytes. Resveratrol (RSV), a polyphenol from the skins of grapes and red wine, is an activator of SIRT3. The current study aimed to investigate the protective effect of RSV against TP-induced cardiotoxicity and the underlying mechanisms. Mice were treated with a single dose of TP (2.5 mg/kg) via the intragastric (i.g.) route. After 24 h, TP induced abnormal changes of serum biochemistry, activity decrease of antioxidant enzymes and damage of heart tissue such as myocardial fiber rupture, cell swelling and interstitial congestion. In contrast, administration with RSV (50 mg/kg i.g. 12 h before and 2 h after the administration of TP) attenuated the detrimental effects induced by TP in BALB/c mice. Moreover, the cardiomyocyte protective effects of RSV on TP-induced heart injury were associated with the activation of SIRT3 and its downstream targets. In vitro study also indicated that RSV counteracted TP-induced cardiotoxicity through SIRT3-FOXO3 signaling pathway in H9c2 cells. Collectively, these findings suggest the potential of RSV as a promising agent in protecting heart from TP-induced damage.


Antioxidants/pharmacology , Antioxidants/therapeutic use , Cardiotonic Agents/pharmacology , Cardiotonic Agents/therapeutic use , Cardiotoxicity , Diterpenes/toxicity , Heart Diseases/chemically induced , Heart Diseases/prevention & control , Phenanthrenes/toxicity , Sirtuin 3/drug effects , Stilbenes/pharmacology , Stilbenes/therapeutic use , Animals , Apoptosis/drug effects , Cell Line , Diterpenes/antagonists & inhibitors , Epoxy Compounds/antagonists & inhibitors , Epoxy Compounds/toxicity , Female , Forkhead Box Protein O3/drug effects , Humans , Male , Mice , Mice, Inbred BALB C , Myocardium/enzymology , Myocardium/pathology , Myocytes, Cardiac/drug effects , Phenanthrenes/antagonists & inhibitors , Resveratrol , Signal Transduction/drug effects
16.
PLoS One ; 9(7): e100685, 2014.
Article En | MEDLINE | ID: mdl-24988078

Triptolide, the major active component of Tripterygium wilfordii Hook f. (TWHF), has a wide range of pharmacological activities. However, the toxicities of triptolide, particularly the hepatotoxicity, limit its clinical application. The hepatotoxicity of triptolide has not been well characterized yet. The aim of this study was to investigate the role of NF-E2-related factor 2 (Nrf2) in triptolide-induced toxicity and whether activation of Nrf2 could protect against triptolide-induced hepatotoxicity. The results showed that triptolide caused oxidative stress and cell damage in HepG2 cells, and these toxic effects could be aggravated by Nrf2 knockdown or be counteracted by overexpression of Nrf2. Treatment with a typical Nrf2 agonist, sulforaphane (SFN), attenuated triptolide-induced liver dysfunction, structural damage, glutathione depletion and decrease in antioxidant enzymes in BALB/C mice. Moreover, the hepatoprotective effect of SFN on triptolide-induced liver injury was associated with the activation of Nrf2 and its downstream targets. Collectively, these results indicate that Nrf2 activation protects against triptolide-induced hepatotoxicity.


Antineoplastic Agents, Alkylating/adverse effects , Chemical and Drug Induced Liver Injury/metabolism , Chemical and Drug Induced Liver Injury/prevention & control , Diterpenes/adverse effects , NF-E2-Related Factor 2/metabolism , Phenanthrenes/adverse effects , Animals , Anticarcinogenic Agents/pharmacology , Antineoplastic Agents, Alkylating/pharmacology , Chemical and Drug Induced Liver Injury/pathology , Diterpenes/pharmacology , Epoxy Compounds/adverse effects , Epoxy Compounds/pharmacology , Hep G2 Cells , Humans , Isothiocyanates/pharmacology , Mice , Mice, Inbred BALB C , NF-E2-Related Factor 2/antagonists & inhibitors , Oxidative Stress/drug effects , Phenanthrenes/pharmacology , Sulfoxides
17.
Pharm Biol ; 51(4): 447-54, 2013 Apr.
Article En | MEDLINE | ID: mdl-23363047

CONTEXT: N'-(7-Fluoro-5-N-methyl-10H-indolo[3,2-b]quinolin-5-ium)-N,N-dimethylpropane-1,3-diamine iodide (SYUIQ-FM05) is a semi-synthetic derivative of cryptolepine which is from Cryptolepis sanguinolenta (Lindl.) Schlechter (Periplocaeae). This ligand inhibits telomerase activity by stabilizing the G-quadruplex structure and induces growth arrest in cancer cells. OBJECTIVE: The anticancer activity of SYUIQ-FM05 via inhibiting c-kit transcription was investigated in leukemic cells. MATERIALS AND METHODS: The cytotoxicity of SYUIQ-FM05 in K562 cells was evaluated using a cell viability assay and flow cytometry (FCM) at 0.4, 2.0, 10.0 and 20.0 nM. Under the same concentrations of SYUIQ-FM05 or 100 nM imatinib mesylate (IM), quantitative polymerase chain reaction (Q-PCR) investigated transcription of c-kit and bcl-2, and western blotting analyzed the expression levels of c-Kit, total mitogen-activated protein kinase kinases (MEKs), phospho-MEK (p-MEK), total extracellular regulated protein kinases (ERKs), phospho-ERK (p-ERK), Bcl-2 and Bax. RESULTS: SYUIQ-FM05 inhibited cellular growth with an IC(50) of 10.83 ± 0.05 nM in K562 cells. c-Kit transcription was suppressed 2.69-, 4.39-, 7.71- and 10.52-fold at 0.4, 2.0, 10.0 and 20.0 nM SYUIQ-FM05, respectively, which produced proportional loss of total c-Kit protein except IM. Both SYUIQ-FM05 and IM downregulated p-MEK and p-ERK. Furthermore, bcl-2 transcription was suppressed 1.58- and 1.86-fold at 10.0 and 20.0 nM SYUIQ-FM05, respectively, but 0.4 and 2.0 nM SYUIQ-FM05 had no effect. A decrease in Bcl-2 and an increase in Bax appeared in these treated cells. DISCUSSION AND CONCLUSION: These findings demonstrate that SYUIQ-FM05 could induce apoptosis in a leukemic cell line through inhibiting c-kit transcription, which supports the anticancer potency of SYUIQ-FM05 in c-Kit-positive leukemic cells.


Antineoplastic Agents/pharmacology , Apoptosis/drug effects , Diamines/pharmacology , Enzyme Inhibitors/pharmacology , G-Quadruplexes/drug effects , Indoles/pharmacology , Proto-Oncogene Proteins c-kit/metabolism , Quinolines/pharmacology , Quinolinium Compounds/pharmacology , Telomerase/antagonists & inhibitors , Transcription, Genetic/drug effects , Benzamides/pharmacology , Blotting, Western , Cell Survival/drug effects , Dose-Response Relationship, Drug , Down-Regulation , Extracellular Signal-Regulated MAP Kinases/metabolism , Flow Cytometry , Humans , Imatinib Mesylate , Inhibitory Concentration 50 , K562 Cells , Ligands , Mitogen-Activated Protein Kinase Kinases/metabolism , Phosphorylation , Piperazines/pharmacology , Proto-Oncogene Mas , Proto-Oncogene Proteins c-bcl-2/genetics , Proto-Oncogene Proteins c-bcl-2/metabolism , Proto-Oncogene Proteins c-kit/genetics , Pyrimidines/pharmacology , Real-Time Polymerase Chain Reaction , Reverse Transcriptase Polymerase Chain Reaction , Telomerase/metabolism , bcl-2-Associated X Protein/metabolism
18.
Zhongguo Zhong Yao Za Zhi ; 31(14): 1184-7, 2006 Jul.
Article Zh | MEDLINE | ID: mdl-17048591

OBJECTIVE: To observe the improvement effects of puerarin on glycated brain damages in rat model induced by D-galactose. METHOD: The model rats of protein glycation were induced by intraperitoneal administration of D-galactose (150 mg x kg(-1) x d(-1)) for 8 weeks, and all rats were treated with puerarin (high dose 300 mg x kg(-1), middle dose 150 mg x kg(-1), low dose 75 mg x kg(-1)) for 6 weeks. The activity of aldose reductase in red blood cells, the amount of glycated products (fructosamine in serum, glycohaemoglobin, advanced glycation end-products) and AGEs in brain tissue, calcium ion in brain cells were measured. Moreover, mitochondria in brain hippocampus cells were observed under electronic microscope. RESULT: High dose and middle dose of puerarin can decrease the activity of aldose reductase in red blood cells (P < 0.01), and inhibit the formation of glycation products significantly in model rats induced by D-galactose (P < 0.01). Also, puerarin can decrease the content of AGEs in brain and the level of calcium ions in brain cells (P < 0.05, P < 0.01), and decrease lesions degree in mitochondria in brain hippocampus cells. CONCLUSION: Puerarin can produce the protective effects on glycated brain damages through inhibiting the glycation reaction in rats induced by D-galactose.


Brain/pathology , Isoflavones/pharmacology , Neuroprotective Agents/pharmacology , Pueraria , Aldehyde Reductase/metabolism , Animals , Brain/metabolism , Calcium/metabolism , Erythrocytes/enzymology , Female , Fructosamine/blood , Galactose/antagonists & inhibitors , Glycated Hemoglobin/metabolism , Glycation End Products, Advanced/metabolism , Hippocampus/ultrastructure , Isoflavones/isolation & purification , Male , Mitochondria/ultrastructure , Plants, Medicinal/chemistry , Pueraria/chemistry , Random Allocation , Rats , Rats, Sprague-Dawley
...