Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 97
Filter
1.
Toxicol Appl Pharmacol ; 486: 116921, 2024 May.
Article in English | MEDLINE | ID: mdl-38582374

ABSTRACT

As a protein kinase inhibitor, cantharidin (CTD) exhibits antitumor activities. However, CTD is highly toxic, thereby limiting clinical applications. Moreover, relatively few studies have investigated CTD-induced reproductive toxicity, thus the underlying mechanism remains unclear. In this study, the toxic effects of CTD on mouse testis were confirmed in vivo and the potential mechanism was predicted by network toxicology (NT) and molecular docking technology. Proteins involved in the signaling pathways and core targets were verified. The results showed that different concentrations of CTD induced weight loss increased the testicular coefficient, and caused obvious pathological damage to testicular cells. The NT results showed that the main targets of CTD-induced testicular injury (TI) included AKT1, Caspase 3, Bcl-2, and Bax. The results of pathway enrichment analysis showed that CTD-induced TI was closely related to apoptosis and the PI3K/AKT and HIF-1 signaling pathways. Molecular docking methods confirmed high affinity between CTD and key targets. Western blot analysis showed that CTD inhibited expression of PI3K, AKT, and the anti-apoptotic protein Bcl-2, while promoting expression of the pro-apoptotic proteins Bax and Caspase 3. These results suggest that CTD-induced TI involves multiple targets and pathways, and the underlying mechanism was associated with inhibition of the apoptosis-related PI3K/AKT signaling pathway.


Subject(s)
Cantharidin , Molecular Docking Simulation , Network Pharmacology , Proto-Oncogene Proteins c-akt , Signal Transduction , Testis , Animals , Male , Mice , Proto-Oncogene Proteins c-akt/metabolism , Testis/drug effects , Testis/metabolism , Testis/pathology , Cantharidin/toxicity , Signal Transduction/drug effects , Apoptosis/drug effects , Phosphatidylinositol 3-Kinases/metabolism
2.
J Appl Toxicol ; 44(7): 990-1004, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38448202

ABSTRACT

Cantharidin (CTD) is the main active component in the traditional Chinese medicine Mylabris and an effective anti-tumor agent. However, it is relatively toxic and exhibits nephrotoxicity, which limits its clinical use. However, its toxic mechanism is not clear. The toxic effects of CTD exposure on the kidney and the protective effect of resveratrol (RES) were studied in a mouse model, by determination of serum biochemical and renal antioxidant indicators, histopathological and ultrastructural observation, and metabonomics. After CTD exposure, serum uric acid, creatinine, and tissue oxidative stress indicators increased, and the renal glomerular and tubular epithelial cells showed clear pathological damage. Ultrastructure observation revealed marked mitochondrial swelling, endoplasmic reticulum dilation, and the presence of autophagy lysosomes in glomerular epithelial cells. RES ameliorated the renal injury induced by CTD. Metabonomics analysis indicated that CTD can induce apoptosis and oxidative damage in kidney cells, mainly by disrupting sphingolipid and glutathione metabolism, increasing sphingosine and sphingomyelin levels, and decreasing glutathione levels. RES counteracts these effects by regulating renal cell proliferation, the inflammatory response, oxidative stress, and apoptosis, by improving the levels of phosphatidylcholine (PC), LysoPC, and lysophosphatidyl glycerol in the glycerophospholipid metabolism pathway, thereby reducing CTD-induced nephrotoxicity. The mechanisms of CTD-induced renal injury and the protective effect of RES were revealed by metabonomics, providing a basis for evaluating clinical treatment regimens to reduce CTD-induced nephrotoxicity.


Subject(s)
Cantharidin , Kidney , Metabolomics , Oxidative Stress , Resveratrol , Animals , Resveratrol/pharmacology , Mice , Male , Cantharidin/toxicity , Oxidative Stress/drug effects , Kidney/drug effects , Kidney/pathology , Kidney/metabolism , Apoptosis/drug effects , Chromatography, Liquid , Antioxidants/pharmacology , Mass Spectrometry
3.
Vet Clin North Am Equine Pract ; 40(1): 113-119, 2024 Apr.
Article in English | MEDLINE | ID: mdl-37716858

ABSTRACT

Cantharidin is the toxic component of blister beetles of the genus Epicauta. Cantharidin is a potent vesicant which causes blisters, erosions, and ulcerations in the gastrointestinal and urinary tracts, and can cause myocardial necrosis. Blister beetles are found over most of North America and specifically contaminate alfalfa at harvest. History of alfalfa feeding, with colic, dysuria, hypocalcemia, and hypomagnesemia are suggestive of blister beetle toxicosis. Myocardial damage causes increased serum cardiac troponin 1. Tentative diagnosis can be made by finding the beetles in feed or ingesta. Definitive diagnosis requires detection of cantharidin in urine or gastric contents. Treatment involves ending exposure, decreasing absorption, controlling pain, using gastroprotectants, and fluids and electrolyte replacement. Prognosis is guarded to poor.


Subject(s)
Coleoptera , Colic , Horse Diseases , Horses , Animals , Cantharidin/toxicity , Horse Diseases/diagnosis , Horse Diseases/etiology , Colic/veterinary , Pain/veterinary
4.
Toxicol Appl Pharmacol ; 465: 116450, 2023 04 15.
Article in English | MEDLINE | ID: mdl-36907384

ABSTRACT

Cantharidin (CTD), extracted from the traditional Chinese medicine mylabris, has shown significant curative effects against a variety of tumors, but its clinical application is limited by its high toxicity. Studies have revealed that CTD can cause toxicity in the kidneys; however, the underlying molecular mechanisms remain unclear. In this study, we investigated the toxic effects in mouse kidneys following CTD treatment by pathological and ultrastructure observations, biochemical index detection, and transcriptomics, and explored the underlying molecular mechanisms by RNA sequencing (RNA-seq). The results showed that after CTD exposure, the kidneys had different degrees of pathological damage, altered uric acid and creatinine levels in serum, and the antioxidant indexes in tissues were significantly increased. These changes were more pronounced at medium and high doses of CTD. RNA-seq analysis revealed 674 differentially expressed genes compared with the control group, of which 131 were upregulated and 543 were downregulated. Gene ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway enrichment analyses showed that many differentially expressed genes were closely related to the stress response, the CIDE protein family, and the transporter superfamily, as well as the MAPK, AMPK, and HIF-1 pathways. The reliability of the RNA-seq results was verified by qRT-PCR of the six target genes. These findings offer insight into the molecular mechanisms of renal toxicity caused by CTD and provide an important theoretical basis for the clinical treatment of CTD-induced nephrotoxicity.


Subject(s)
Cantharidin , Transcriptome , Mice , Animals , Cantharidin/toxicity , Reproducibility of Results , Gene Expression Profiling , Sequence Analysis, RNA
5.
Fish Shellfish Immunol ; 123: 20-35, 2022 Apr.
Article in English | MEDLINE | ID: mdl-35218974

ABSTRACT

Cantharidin is a toxic vesicant terpene used in folk and traditional medicine due to its various therapeutic effects. Since there are no previous data on the effect of cantharidin in fish, this study aimed to investigate the in vitro related-inflammatory effects of cantharidin in gilthead seabream (Sparus aurata L.) head-kidney leucocytes (HKLs). In the first experiment, the HKLs were incubated with 0, 5 and 10 µg mL-1 of cantharidin for 24 h to delimit its possible toxic effects. In a second experiment, leucocytes were incubated with ranging concentrations from 0 to 10 µg mL-1 for 3, 6, or 12 h. Cell viability was higher in acidophilic granulocytes than in monocytes/macrophages and lymphocytes. Cantharidin caused apoptosis as was evidenced by transmission electron microscopy. In addition, cantharidin produced a time- and dose-dependent decrease of respiratory burst and phagocytic activities in HKLs, while their peroxidase activity was increased at 24 h of incubation with 5 and 10 µg mL-1 of cantharidin. Different changes in the gene expression were observed after incubation with cantharidin. While the gene expression of tnfa, il1b and crel was up-regulated in HKLs, the nfkb1 and igmh genes were down-regulated in comparison to the expression found in control HKLs. Present results offer a first view of the possible effects and action mechanisms of cantharidin in HKLs, as well as its implication in the inflammatory process, which could be of interest not only for basic research but also in the aquaculture sector.


Subject(s)
Sea Bream , Animals , Cantharidin/metabolism , Cantharidin/toxicity , Head Kidney , Kidney , Leukocytes , Sea Bream/metabolism
6.
J Appl Toxicol ; 42(6): 970-980, 2022 06.
Article in English | MEDLINE | ID: mdl-34866203

ABSTRACT

Cantharidin (CTD) is an effective antitumor agent. However, it exhibits significant hepatotoxicity, the mechanism of which remains unclear. In this study, biochemical and histopathological analyses complemented with ultra-high-performance liquid chromatography-tandem mass spectrometry (UHPLC-MS/MS)-based targeted metabolomic analysis of bile acids (BAs) were employed to investigate CTD-induced hepatotoxicity in rats. Sixteen male and female Sprague-Dawley rats were randomly divided into two groups: control and CTD (1.0 mg/kg) groups. Serum and liver samples were collected after 28 days of intervention. Biochemical, histopathological, and BA metabolomic analyses were performed for all samples. Further, the key biomarkers of CTD-induced hepatotoxicity were identified via multivariate and metabolic pathway analyses. In addition, metabolite-gene-enzyme network and Kyoto Encyclopedia of Genes and Genomes pathway analyses were used to identify the signaling pathways related to CTD-induced hepatotoxicity. The results revealed significantly increased levels of biochemical indices (alanine aminotransferase, aspartate aminotransferase, and total bile acid). Histopathological analysis revealed that the hepatocytes were damaged. Further, 20 endogenous BAs were quantitated via UHPLC-MS/MS, and multivariate and metabolic pathway analyses of BAs revealed that hyocholic acid, cholic acid, and chenodeoxycholic acid were the key biomarkers of CTD-induced hepatotoxicity. Meanwhile, primary and secondary BA biosynthesis and taurine and hypotaurine metabolism were found to be associated with the mechanism by which CTD induced hepatotoxicity in rats. This study provides useful insights for research on the mechanism of CTD-induced hepatotoxicity.


Subject(s)
Chemical and Drug Induced Liver Injury , Drug-Related Side Effects and Adverse Reactions , Animals , Bile Acids and Salts/metabolism , Biomarkers , Cantharidin/toxicity , Chemical and Drug Induced Liver Injury/pathology , Chromatography, High Pressure Liquid/methods , Drug-Related Side Effects and Adverse Reactions/metabolism , Female , Liver , Male , Metabolomics/methods , Rats , Rats, Sprague-Dawley , Tandem Mass Spectrometry/methods
7.
Pak J Pharm Sci ; 35(6(Special)): 1827-1834, 2022 Nov.
Article in English | MEDLINE | ID: mdl-36861250

ABSTRACT

Cantharidin is a natural compound with cardiotoxicity. Cellular senescence and senescence-associated secretory phenotype (SASP) are implicated in chemotherapy-associated cardiotoxicity. We here investigated how cantharidin induced cardiomyocyte senescence. H9c2 cells were treated with cantharidin. Senescence, mitochondrial functions, SASP, NOD-like receptor thermal protein domain associated protein 3 (NLRP3) signaling and AMP-activated protein kinase (AMPK) phosphorylation were examined. Cantharidin inhibited viability and increased expression of senescence-associated ß--galactosidase (SA-ß-Gal), p16 and p21 in H9c2 cells, suggesting occurrence of senescence. Cantharidin impaired mitochondrial functions evidenced by reduction in basal respiration, ATP levels and spare respiratory capacity. Cantharidin also decreased mitochondrial DNA copy number and down-regulated mRNA levels of cytochrome c oxidase-I, -II and -III. Moreover, cantharidin suppressed activity of mitochondria complex-I and -II. Examinations of SASP showed that cantharidin promoted expression and secretion of SASP cytokines interleukin-1ß-, -6 and -8 and tumor necrosis factor-α, associated with activation of NLRP3/caspase-1 pathway. Finally, cantharidin suppressed AMPK phosphorylation. AMPK activator GSK621 abrogated the up-regulation of SA-ß--Gal, p16 and p21 and counteracted the activation of NLRP3 and caspase-1 in cantharidin-challenged H9c2 cells. In conclusion, cantharidin stimulated senescence and SASP in cardiomyocytes through activation of NLRP3 inflammasome and inhibition of AMPK, providing novel molecular insights into cantharidin-induced cardiotoxicity.


Subject(s)
AMP-Activated Protein Kinases , Myocytes, Cardiac , Humans , Cantharidin/toxicity , Inflammasomes , NLR Family, Pyrin Domain-Containing 3 Protein , Cardiotoxicity , Caspases
8.
BMC Genomics ; 22(1): 808, 2021 Nov 08.
Article in English | MEDLINE | ID: mdl-34749651

ABSTRACT

BACKGROUND: Meloidae (blister beetles) are known to synthetize cantharidin (CA), a toxic and defensive terpene mainly stored in male accessory glands (MAG) and emitted outward through reflex-bleeding. Recent progresses in understanding CA biosynthesis and production organ(s) in Meloidae have been made, but the way in which self-protection is achieved from the hazardous accumulation and release of CA in blister beetles has been experimentally neglected. To provide hints on this pending question, a comparative de novo assembly transcriptomic approach was performed by targeting two tissues where CA is largely accumulated and regularly circulates in Meloidae: the male reproductive tract (MRT) and the haemolymph. Differential gene expression profiles in these tissues were examined in two blister beetle species, Lydus trimaculatus (Fabricius, 1775) (tribe Lyttini) and Mylabris variabilis (Pallas, 1781) (tribe Mylabrini). Upregulated transcripts were compared between the two species to identify conserved genes possibly involved in CA detoxification and transport. RESULTS: Based on our results, we hypothesize that, to avoid auto-intoxication, ABC, MFS or other solute transporters might sequester purported glycosylated CA precursors into MAG, and lipocalins could bind CA and mitigate its reactivity when released into the haemolymph during the autohaemorrhaging response. We also found an over-representation in haemolymph of protein-domains related to coagulation and integument repairing mechanisms that likely reflects the need to limit fluid loss during reflex-bleeding. CONCLUSIONS: The de novo assembled transcriptomes of L. trimaculatus and M. variabilis here provided represent valuable genetic resources to further explore the mechanisms employed to cope with toxicity of CA in blister beetle tissues. These, if revealed, might help conceiving safe and effective drug-delivery approaches to enhance the use of CA in medicine.


Subject(s)
Cantharidin , Coleoptera , Animals , Cantharidin/toxicity , Coleoptera/genetics , Genitalia, Male , Hemolymph , Male , Transcriptome
9.
Biochem Biophys Res Commun ; 553: 141-147, 2021 05 14.
Article in English | MEDLINE | ID: mdl-33770579

ABSTRACT

Cantharidin is a potent anti-cancer drug and is known to exert its cytotoxic effects in several cancer cell lines. Although we have ample knowledge about its mode of action, we still know a little about cantharidin associated drug resistance mechanisms which dictates the efficacy and cytotoxic potential of this drug. In this direction, in the present study we employed Sacharomyces cerevisiae as a model organism and screened mutants of pleiotropic drug resistance network of genes for their susceptibility to cantharidin. We show that growth of pdr1Δ and pdr1Δpdr3Δ was severely reduced in presence of cantharidin whereas that of pdr3Δ remain unaffected when compared to wildtype. Loss of one of the PDR1 target genes PDR5, encoding an ABC membrane efflux pump, rendered the cells hypersensitive whereas overexpression of it conferred resistance. Additionally, cantharidin induced the upregulation of both PDR1 and PDR5 genes. Interestingly, pdr1Δpdr5Δ double deletion mutants were hypersensitive to cantharidin showing a synergistic effect in its cellular detoxification. Furthermore, transcriptional activation of PDR5 post cantharidin treatment was majorly dependent on the presence of Pdr1 and less significantly of Pdr3 transcription factors. Altogether our findings suggest that Pdr1 acts to increase cantharidin resistance by elevating the level of Pdr5 which serves as a major detoxification safeguard under CAN stress.


Subject(s)
ATP-Binding Cassette Transporters/metabolism , Cantharidin/pharmacology , Drug Resistance, Fungal/drug effects , Saccharomyces cerevisiae Proteins/metabolism , Saccharomyces cerevisiae/drug effects , ATP-Binding Cassette Transporters/genetics , Adaptation, Physiological/drug effects , Adaptation, Physiological/genetics , Cantharidin/toxicity , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Drug Resistance, Fungal/genetics , Gene Expression Regulation, Fungal/drug effects , Inactivation, Metabolic/drug effects , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae/metabolism , Saccharomyces cerevisiae Proteins/genetics , Transcription Factors/genetics , Transcription Factors/metabolism , Up-Regulation/drug effects
10.
J Appl Toxicol ; 40(11): 1491-1497, 2020 11.
Article in English | MEDLINE | ID: mdl-32618016

ABSTRACT

The cardiotoxicity of cantharidin has been well characterized, but the understanding of the underlying mechanism(s) is incomplete. To more fully understand the differentially expressed genes (DEGs) in cantharidin-induced myocardial injury, Sprague-Dawley rats were exposed to cantharidin (1.34 mg/kg or 2.67 mg/kg) for 24 h and then the heart was sampled for pathologic changes analysis and RNA-sequencing-based transcriptomic profiling. In addition, serum troponin T (TN-T) levels were also tested using the enzyme-linked immunosorbent assay method. The results showed that cantharidin could cause myocardial damage and elevated serum TN-T levels. The genes with a fold change ≥2 were considered as DEGs and we found 38 DEGs that were mainly enriched in eight pathways revealed by Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway analysis. The cellular component of gene ontology analysis showed that the DEGs were mostly enriched in the extracellular matrix. In conclusion, our present study demonstrated that cantharidin induces myocardial injury by multiple modulatory mechanisms, which provide new insights for further study of the pathophysiologic mechanism of cantharidin-induced myocardial injury.


Subject(s)
Cantharidin/toxicity , Gene Expression Profiling , Heart Diseases/chemically induced , Myocardium/metabolism , RNA-Seq , Transcriptome , Animals , Cardiotoxicity , Gene Regulatory Networks , Heart Diseases/genetics , Heart Diseases/metabolism , Heart Diseases/pathology , Male , Myocardium/pathology , Rats, Sprague-Dawley , Signal Transduction/genetics , Troponin T/blood
11.
J Appl Toxicol ; 40(9): 1259-1271, 2020 09.
Article in English | MEDLINE | ID: mdl-32468647

ABSTRACT

Cantharidin (CTD), a compound secreted from Mylabris species, exhibits strong antitumor properties; however, hepatotoxicity restricts its clinical application. The mechanism by which CTD induces toxicity remains unclear. In the present study, the hepatotoxicity of CTD in the rat was investigated using a metabolomic approach combined with conventional pathology methods. A total of 30 rats were intragastrically treated with two doses of CTD (0.75 and 1.5 mg/kg) for 15 days to evaluate hepatotoxicity. Serum and liver samples were collected for biochemical dynamics analyses, histopathological examination and metabolomic analysis. It was found that liver index and serum biochemical indices were significantly increased. Furthermore, the pathology results showed that hepatocytes and subcellular organelles were damaged. Metabolomics analysis found 4 biomarkers in serum and 15 in the liver that were associated with CTD-induced hepatotoxicity. In addition, these were responsible for CTD hepatotoxicity by glycerophospholipid metabolism, sphingolipid metabolism, and steroid hormone biosynthesis. In conclusion, conventional pathology and metabolomics for exploring hepatotoxicity can provide useful information about the safety and potential risks of CTD.


Subject(s)
Biomarkers/blood , Cantharidin/toxicity , Chemical and Drug Induced Liver Injury/metabolism , Chemical and Drug Induced Liver Injury/physiopathology , Chromatography, High Pressure Liquid/methods , Hepatocytes/drug effects , Metabolomics/methods , Animals , Coleoptera/chemistry , Dose-Response Relationship, Drug , Female , Male , Models, Animal , Rats , Rats, Sprague-Dawley
12.
J Appl Toxicol ; 40(11): 1526-1533, 2020 11.
Article in English | MEDLINE | ID: mdl-32627230

ABSTRACT

Cantharidin is of high medicinal value but has strong toxicity. Nowadays, multiple research has focused on the mechanism of its antitumor activity while research on toxicological profiles associated with cantharidin poisoning is still limited. Its hepatotoxicity has attracted attention recently for the crucial role of the liver in detoxification. Here, we aim to find a potential mechanism for cantharidin-induced acute hepatotoxicity with a view to assisting subsequent research or clinical use or detoxification. Twenty-one male Sprague-Dawley rats were randomly divided into control, low-dose (1.34 mg/kg) and high-dose (2.67 mg/kg) cantharidin exposure groups. We used hematoxylin-eosin to observe pathological changes and used immunofluorescent staining, western blotting and real-time quantitative polymerase chain reaction to detect the expression of the markers. The main pathological changes in livers of cantharidin-treated rats were necrosis, inflammatory infiltration and hemorrhage. We found coexpression of tumor necrosis factor alpha (TNF-α), IkappaB kinase-alpha (IKK-α) and caspase3 by immunofluorescent staining in livers of cantharidin-treated rats. Compared with the control, the levels of TNF-α, IKK-α and caspase3 increased significantly in the experimental groups (P < .05). The ratio of B-cell lymphoma-2 (Bcl-2)/Bax increased in the low-dose group but decreased in the high-dose group (P < .05). Cantharidin exposure raised IKK-α mRNA and caspase3 mRNA levels (P < .05). In conclusion, the participation of TNF-α, IKK-α, Bcl-2, Bax and caspase3 uncovered a novel mechanism underlying cantharidin-induced acute hepatotoxicity, and the mechanism needs to be studied further.


Subject(s)
Cantharidin/toxicity , Caspase 3/metabolism , Chemical and Drug Induced Liver Injury/etiology , I-kappa B Kinase/metabolism , Liver/drug effects , Proto-Oncogene Proteins c-bcl-2/metabolism , Tumor Necrosis Factor-alpha/metabolism , Animals , Caspase 3/genetics , Chemical and Drug Induced Liver Injury/enzymology , Chemical and Drug Induced Liver Injury/pathology , I-kappa B Kinase/genetics , Liver/enzymology , Liver/pathology , Male , Necrosis , Rats, Sprague-Dawley , Signal Transduction
13.
J Appl Toxicol ; 40(8): 1153-1161, 2020 08.
Article in English | MEDLINE | ID: mdl-32162354

ABSTRACT

Early diagnosis of cantharidin-induced myocardial injury is the key to reduce the fatality rate in clinical practice. The purpose of the present study was to explore biomarkers that can be used for the prediction and diagnosis of cantharidin-induced myocardial injury. Of 65 male Sprague-Dawley rats weighing 200-230 g, 25 rats were divided into five groups according to the administration dose of cantharidin (0, 1.34, 2.67, 4 and 5.34 mg/kg; n = 5 per group) and the other 40 rats were treated with 2.67 mg/kg cantharidin and divided into nine groups according to the administration time (0, 1, 2, 4, 6, 8, 12, 24, 48 and 72 hours; n = 4 per group). Pathological changes of hypoxia, necrosis and inflammation were confirmed in heart samples that were exposed to cantharidin by hematoxylin-eosin staining and overall scores of pathological changes among heart samples in cantharidin exposure groups showed an increasing trend compared with in the control group. Coexpression of vascular endothelial growth factor (VEGF), hypoxia inducible factor-1α (HIF-1α) and caspase9 was shown in the myocardium by immunofluorescence staining. Western blotting results showed that expression of VEGF, HIF-1α and caspase9 in cantharidin-treated rat hearts showed an increasing trend compared with in the control group. Results of enzyme-linked immunosorbent assay suggested that plasma levels of troponin T (TN-T), VEGF and HIF-1α were elevated at different intervals after cantharidin administration, and VEGF and HIF-1α had a significant linear relationship with TN-T that was verified by multiple linear regression analysis. Preliminary results serve to illustrate that TN-T, VEGF and HIF-1α might be valuable molecular markers in cantharidin-induced myocardial injury and that diagnostic accuracy needs to be studied further.


Subject(s)
Biomarkers/blood , Cantharidin/toxicity , Cardiomyopathies/chemically induced , Cardiotoxicity/physiopathology , Troponin T/drug effects , Vascular Endothelial Growth Factors/drug effects , Animals , Cardiomyopathies/physiopathology , Dose-Response Relationship, Drug , Hypoxia/chemically induced , Hypoxia/physiopathology , Inflammation/chemically induced , Inflammation/physiopathology , Male , Necrosis/chemically induced , Necrosis/physiopathology , Predictive Value of Tests , Rats , Rats, Sprague-Dawley
14.
J Appl Toxicol ; 40(12): 1622-1635, 2020 12.
Article in English | MEDLINE | ID: mdl-32638414

ABSTRACT

Cantharidin (CTD), an important active compound derived from the traditional Chinese medicine Mylabris (also called Banmao), has been used in the treatment of diseases such as tumors and dermatosis. However, Mylabris has been shown to induce hepatotoxicity in clinical practice and animal experiments, limiting its use. Further, a detailed mechanism underlying CTD-induced hepatotoxicity has not been determined. In the present study, we aimed to explore the effect of endoplasmic reticulum stress (ERS), autophagy, and apoptosis on CTD-induced hepatotoxicity. We found that CTD could inhibit the proliferation of LO2 cells; increase alanine aminotransferase, aspartate aminotransferase, lactate dehydrogenase, and malondialdehyde levels; and reduce glutathione peroxidase and superoxide dismutase activities. Western blotting showed that low concentrations of CTD induced the expressions of ERS-related proteins [GRP78, ATF4, PERK, p-PERK, XBP1-1 s, and CHOP], but high concentrations of CTD inhibited their expressions. Furthermore, high concentrations of CTD activated autophagy (LC3, Beclin-1, Atg3, Atg4A, Atg4B, and Atg7), induced the expressions of apoptotic proteins (Bax/Bcl-2 and caspase-3), and increased LO2 toxicity. Taken together, these results indicated that CTD can induce LO2 cytotoxicity by inhibiting ERS and inducing autophagy and apoptosis, which provides a scientific basis for CTD-induced hepatotoxicity.


Subject(s)
Apoptosis/drug effects , Autophagy/drug effects , Cantharidin/toxicity , Chemical and Drug Induced Liver Injury/etiology , Endoplasmic Reticulum Stress/drug effects , Liver/drug effects , Apoptosis Regulatory Proteins/metabolism , Autophagy-Related Proteins/metabolism , Cell Line , Chemical and Drug Induced Liver Injury/metabolism , Chemical and Drug Induced Liver Injury/pathology , Endoplasmic Reticulum Chaperone BiP , Humans , Liver/metabolism , Liver/ultrastructure , Oxidative Stress/drug effects , Signal Transduction
15.
Chem Res Toxicol ; 32(11): 2204-2213, 2019 11 18.
Article in English | MEDLINE | ID: mdl-31617706

ABSTRACT

Cantharidin is the major bioactive compound extracted from the blister beetle, a traditional Chinese medicine, and has been proved to be a natural component with widely antitumor activity. However, clinical application of cantharidin is relatively restricted due to its potential toxic effects, especially hepatotoxicity. Although cantharidin-induced liver injury has been reported, the underlying molecular mechanisms remain unclear. In the present study, an UPLC-Q-TOF/MS based metabolomics approach combined with blood biochemical analysis, histopathological examination, and cell apoptosis assay were used to investigate the mechanisms of cantharidin-induced hepatotoxicity. A total of 54 significantly changed metabolites and 14 disturbed metabolic pathways were identified in the cantharidin exposed groups. Among them, four metabolites (oxidized glutathione, glutathione, 3-sulfinoalanine, and deoxycholic acid 3-glucuronide) were selected based on their high impact value and potential biological function in the process of liver injury post cantharidin treatment. Our study provides a deeper understanding of the mechanisms of cantharidin-induced hepatotoxicity and may contribute to reduce the liver injury and gain more effective and safe clinical use of cantharidin. In addition, our results also demonstrated that cantharidin could impair multiple biological processes in liver, and future studies will be necessary to reveal the detailed molecular mechanisms of cantharidin-induced hepatotoxicity.


Subject(s)
Antineoplastic Agents/toxicity , Cantharidin/toxicity , Chemical and Drug Induced Liver Injury/metabolism , Animals , Apoptosis/drug effects , Cell Line , Chemical and Drug Induced Liver Injury/pathology , Chromatography, High Pressure Liquid , Humans , Liver/drug effects , Liver/metabolism , Liver/pathology , Male , Mass Spectrometry , Metabolomics/methods , Mice
16.
J Proteome Res ; 17(1): 579-589, 2018 01 05.
Article in English | MEDLINE | ID: mdl-29261316

ABSTRACT

The new strategy for chemical toxicity testing and modeling is to use in vitro human cell-based assays in conjunction with quantitative high-throughput screening (qHTS) technology, to identify molecular mechanisms and predict in vivo responses. Stem cells are more physiologically relevant than immortalized cell lines because of their unique proliferation and differentiation potentials. We established a robust two stem cells-two lineages assay system, encompassing human mesenchymal stem cells (hMSCs) along osteogenesis and human induced pluripotent stem cells (hiPSCs) along hepatogenesis. We performed qHTS phenotypic screening of LOPAC1280 and identified 38 preliminary hits for hMSCs. This was followed by validation of a selected number of hits and determination of their IC50 values and mechanistic studies of idarubicin and cantharidin treatments using proteomics and bioinformatics. In general, hiPSCs were more sensitive than hMSCs to chemicals, and differentiated progenies were less sensitive than their progenitors. We showed that chemical toxicity depends on both stem cell types and their differentiation stages. Proteomics identified and quantified over 3000 proteins for both stem cells. Bioinformatics identified apoptosis and G2/M as the top pathways conferring idarubicin toxicity. Our Omics-based assays of stem cells provide mechanistic insights into chemical toxicity and may help prioritize chemicals for in-depth toxicological evaluations.


Subject(s)
Induced Pluripotent Stem Cells/drug effects , Mesenchymal Stem Cells/drug effects , Proteomics/methods , Toxicity Tests , Apoptosis , Cantharidin/toxicity , Cells, Cultured , Computational Biology/methods , G2 Phase Cell Cycle Checkpoints , Humans , Idarubicin/toxicity , Proteins/analysis
17.
Pestic Biochem Physiol ; 142: 91-101, 2017 Oct.
Article in English | MEDLINE | ID: mdl-29107253

ABSTRACT

To investigate the insecticidal mechanism of cantharidin, a promising biological pesticide substance from blister beetle, on Sf9 cells, a cultured cell line derived from fall armyworm, Spodoptera frugiperda, we preliminary studied the attribution of Bax channel and mitochondrial permeability transition pore on cantharidin-induced mitochondrial apoptosis signal pathway. Changes in cell morphology, activity of mitochondrial dehydrogenases, release of cytochrome C and mitochondrial transmembrane potential were detected when the two channels were blocked by specific inhibitors, Bax channel blocker and cyclosporin A. Results showed that cantharidin-induced apoptotic features, including changes in the cell morphology, release of cytochrome C and decrease in mitochondrial transmembrane potential could be significantly inhibited by Bax channel blocker, while cyclosporin A accelerated the downward trend of mitochondrial dehydrogenases activity and caused a decrease of Ca2+ in mitochondria. In summary, Bax might be necessary but not exclusively for the apoptosis induced by cantharidin and the attribution of these channels seems to be more complexity.


Subject(s)
Apoptosis , Cantharidin/toxicity , Insect Proteins/metabolism , Mitochondria/drug effects , Mitochondrial Membrane Transport Proteins/metabolism , Spodoptera/metabolism , bcl-2-Associated X Protein/metabolism , Animals , Cytochromes c/metabolism , Insect Proteins/genetics , Membrane Potential, Mitochondrial/drug effects , Mitochondria/metabolism , Mitochondrial Membrane Transport Proteins/drug effects , Mitochondrial Permeability Transition Pore , Sf9 Cells , Spodoptera/cytology , Spodoptera/genetics , bcl-2-Associated X Protein/genetics
18.
Environ Toxicol ; 32(3): 723-738, 2017 Mar.
Article in English | MEDLINE | ID: mdl-27113412

ABSTRACT

Cantharidin (CTD), a potential anticancer agent of Traditional Chinese Medicine has cytotxic effects in different human cancer cell lines. The cytotoxic effects of CTD on A431 human skin cancer (epidermoid carcinoma) cells in vitro and in A431 cell xenograft mouse model were examined. In vitro, A431 human skin cell were treated with CTD for 24 and 48 h. Cell phase distribution, ROS production, Ca2+ release, Caspase activity and the level of apoptosis associated proteins were measured. In vivo, A431 cell xenograft mouse model were examined. CTD-induced cell morphological changes and decreased percentage of viable A431 cells via G0/G1 phase arrest and induced apoptosis. CTD-induced G0/G1 phase arrest through the reduction of protein levels of cyclin E, CDK6, and cyclin D in A431 cells. CTD-induced cell apoptosis of A431 cells also was confirm by DNA gel electrophoresis showed CTD-induced DNA fragmentation. CTD reduced the mitochondrial membrane potential and stimulated release of cytochrome c, AIF and Endo G in A431 cells. Flow cytometry demonstrated that CTD increased activity of caspase-8, -9 and -3. However, when cells were pretreated with specific caspase inhibitors activity was reduced and cell viability increased. CTD increased protein levels of death receptors such as DR4, DR5, TRAIL and levels of the active form of caspase-8, -9 and -3 in A431 cells. AIF and Endo G proteins levels were also enhanced by CTD. In vivo studies showed that CTD significantly inhibited A431 cell xenograft tumors in mice. Taken together, these in vitro and in vivo results provide insight into the mechanisms of CTD on cell growth and tumor production. © 2016 Wiley Periodicals, Inc. Environ Toxicol 32: 723-738, 2017.


Subject(s)
Antineoplastic Agents/toxicity , Apoptosis/drug effects , Cantharidin/toxicity , Animals , Antineoplastic Agents/therapeutic use , Cantharidin/therapeutic use , Carcinoma, Squamous Cell/drug therapy , Carcinoma, Squamous Cell/metabolism , Carcinoma, Squamous Cell/pathology , Caspases/genetics , Caspases/metabolism , Cell Line, Tumor , Cell Survival/drug effects , Cyclin D/metabolism , Cytochromes c/metabolism , DNA Fragmentation/drug effects , G1 Phase Cell Cycle Checkpoints/drug effects , Humans , Male , Membrane Potential, Mitochondrial/drug effects , Mice , Mice, Inbred BALB C , Mice, Nude , Reactive Oxygen Species/metabolism , Receptors, Death Domain/metabolism , Signal Transduction/drug effects , Skin Neoplasms/drug therapy , Skin Neoplasms/metabolism , Skin Neoplasms/pathology , TNF-Related Apoptosis-Inducing Ligand/metabolism , Transplantation, Heterologous
19.
Anticancer Drugs ; 26(5): 508-17, 2015 Jun.
Article in English | MEDLINE | ID: mdl-25588161

ABSTRACT

This study investigated the anticancer effects of two newly synthesized norcantharidin analogs, N-farnesyloxy-norcantharimide (NOC15) and N-farnesyl-norcantharimide (NC15), in L1210 cells and in a syngeneic mouse leukemia model (L1210 cell line plus DBA/2 mice). We found that the half-maximal inhibitory concentration (IC50) of NOC15 and NC15 on L1210 cells is 1.56 and 2.62 µmol/l, respectively, and that the IC50 of NOC15 and NC15 on human normal lymphoblast is 207.9 and 2569 µmol/l, respectively. In cell cycle analysis, NOC15 could increase the sub-G1 phase, whereas NC15 could induce G2/M arrest. Annexin-V apoptosis assay indicated that both NOC15 and NC15 could induce cell apoptosis. In the syngeneic mouse leukemia model, both NOC15 and NC15 could increase the survival days of mice and decrease the tumor weight. Moreover, both NOC15 and NC15 could retard the increase in peripheral blood leukocyte count due to L1210 cells. In the subcutaneous (s.c.) group, the treatment with NOC15 could retard the decrease in the weight of the liver and the spleen caused by L1210 cells, whereas the treatment with NC15 could retard the decrease in the weight of the spleen caused by L1210 cells. We conclude that the new compounds NOC15 and NC15 have strong anticancer activity and low toxicity both in vitro and in vivo. NOC15 and NC15 may have the potential to be developed into anticancer agents in the future.


Subject(s)
Antineoplastic Agents/therapeutic use , Cantharidin/analogs & derivatives , Leukemia L1210/drug therapy , Animals , Annexin A5/metabolism , Antineoplastic Agents/toxicity , Apoptosis/drug effects , Cantharidin/therapeutic use , Cantharidin/toxicity , Cell Cycle/drug effects , Cell Line, Tumor , Cell Survival/drug effects , Humans , Leukemia L1210/mortality , Leukemia L1210/pathology , Mice, Inbred DBA
20.
Environ Toxicol ; 30(10): 1135-43, 2015 Sep.
Article in English | MEDLINE | ID: mdl-24639390

ABSTRACT

Cantharidin is one of the major compounds from mylabris and it has cytotoxic effects in many different types of human cancer cells. Previously, we found that cantharidin induced cell death through cell cycle arrest and apoptosis induction in human lung cancer NCI-H460 cells. However, cantharidin-affected DNA damage, repair, and associated protein levels in NCI-H460 cells have not been examined. In this study, we determined whether cantharidin induced DNA damage and condensation and altered levels of proteins in NCI-H460 cells in vitro. Incubation of NCI-H460 cells with 0, 2.5, 5, 10, and 15 µM of cantharidin caused a longer DNA migration smear (comet tail). Cantharidin also increased DNA condensation. These effects were dose-dependent. Cantharidin (5, 10, and 15 µM) treatment of NCI-H460 cells reduced protein levels of ataxia telangiectasia mutated (ATM), breast cancer 1, early onset (BRCA-1), 14-3-3 proteins sigma (14-3-3σ), DNA-dependent serine/threonine protein kinase (DNA-PK), O(6) -methylguanine-DNA methyltransferase (MGMT), and mediator of DNA damage checkpoint protein 1 (MDC1). Protein translocation of p-p53, p-H2A.X (S140), and MDC1 from cytoplasm to nucleus was induced by cantharidin in NCI-H460 cells. Taken together, this study showed that cantharidin caused DNA damage and inhibited levels of DNA repair-associated proteins. These effects may contribute to cantharidin-induced cell death in vitro.


Subject(s)
Cantharidin/toxicity , DNA Damage/drug effects , DNA Repair Enzymes/metabolism , DNA Repair/drug effects , Apoptosis/drug effects , Cell Line, Tumor , Comet Assay , DNA Repair Enzymes/antagonists & inhibitors , Humans , Lung Neoplasms/metabolism , Lung Neoplasms/pathology , Microscopy, Confocal
SELECTION OF CITATIONS
SEARCH DETAIL