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










Publication year range
1.
Mol Med Rep ; 19(2): 984-993, 2019 02.
Article in English | MEDLINE | ID: mdl-30569150

ABSTRACT

Tumor necrosis factor­related apoptosis-inducing ligand (TRAIL) is well known as a transmembrane cytokine and has been proposed as one of the most effective anti­cancer therapeutic agents, owing to its efficiency to selectively induce cell death in a variety of tumor cells. Suppression of autophagy flux has been increasingly acknowledged as an effective and novel therapeutic intervention for cancer. The present study demonstrated that the anti­cancer and anti­inflammatory drug celastrol, through its anti­metastatic properties, may initiate TRAIL­mediated apoptotic cell death in lung cancer cells. This sensitization was negatively affected by N­acetyl­l­cysteine, which restored the mitochondrial membrane potential (ΔΨm) and inhibited reactive oxygen species (ROS) generation. Notably, treatment with celastrol caused an increase in microtubule­associated proteins 1A/1B light chain 3B­II and p62 levels, whereas co­treatment of celastrol and TRAIL increased active caspase 3 and 8 levels compared with the control, confirming inhibited autophagy flux. The combined use of TRAIL with celastrol may serve as a safe and adequate therapeutic technique for the treatment of TRAIL­resistant lung cancer, suggesting that celastrol­mediated autophagy flux inhibition sensitized TRAIL­initiated apoptosis via regulation of ROS and ΔΨm.


Subject(s)
Antineoplastic Agents, Phytogenic/pharmacology , Apoptosis/drug effects , Gene Expression Regulation, Neoplastic , Membrane Potential, Mitochondrial/drug effects , Reactive Oxygen Species/agonists , TNF-Related Apoptosis-Inducing Ligand/pharmacology , Triterpenes/pharmacology , A549 Cells , Acetylcysteine/pharmacology , Antineoplastic Agents, Phytogenic/antagonists & inhibitors , Apoptosis/genetics , Autophagy/drug effects , Autophagy/genetics , Caspase 8/genetics , Caspase 8/metabolism , Cell Line, Tumor , Chloroquine/pharmacology , Drug Combinations , Drug Synergism , Humans , Microtubule-Associated Proteins/genetics , Microtubule-Associated Proteins/metabolism , Mitochondria/drug effects , Mitochondria/metabolism , Pentacyclic Triterpenes , Reactive Oxygen Species/metabolism , Sequestosome-1 Protein/genetics , Sequestosome-1 Protein/metabolism , Signal Transduction , Triterpenes/antagonists & inhibitors
2.
Chin J Nat Med ; 16(1): 10-19, 2018 Jan.
Article in English | MEDLINE | ID: mdl-29425586

ABSTRACT

Metastasis is responsible for the majority of cancer-related deaths and prevention of metastasis remains a big challenge for cancer therapy. Cucurbitacin B (Cuc B) is a natural triterpenoid with potent anticancer activities while its effect on metastasis remains unclear. In the present study, the inhibitory effect and mechanisms of Cuc B on metastasis were investigated in MDA-MB-231 breast cancer cells. The cells were treated with or without Cuc B, and the cytotoxicity was determined by MTT assay. The effect of Cuc B on metastasis was evaluated with wound healing, transwell, and adhesion assays. Furthermore, the adhesion of cancer cells to endothelial cells was determined. The protein expression was determined by Western blotting. Cuc B (< 100 nmol·L-1) showed no obvious cytotoxicity to MDA-MB-231 cells, but significantly inhibited migration, invasion, and adhesion to Matrigel, fibronectin, type I collagen, and endothelial cells. Cuc B dramatically inhibited the phosphorylation of focal adhesion kinase (FAK) and paxillin in dose- and time-dependent manners. Furthermore, Cuc B induced intracellular reactive oxygen species (ROS) generation, which could be reduced by N-acetyl-l-cysteine (NAC). In addition, NAC pretreatment could reverse Cuc B-induced suppression of migration and adhesion, expression of FAK, but showed no effect on paxillin expression. In summary, Cuc B suppressed ROS-dependent metastasis through FAK pathway in breast cancer MDA-MB-231 cells, demonstrating novel mechanisms for the anticancer effects of Cuc B.


Subject(s)
Antineoplastic Agents/pharmacology , Breast Neoplasms/metabolism , Focal Adhesion Kinase 1/metabolism , Neoplasm Metastasis/pathology , Reactive Oxygen Species/metabolism , Triterpenes/pharmacology , Acetylcysteine/pharmacology , Breast Neoplasms/enzymology , Breast Neoplasms/pathology , Breast Neoplasms/physiopathology , Cell Adhesion/drug effects , Cell Line, Tumor , Cell Movement/drug effects , Collagen Type I/metabolism , Dose-Response Relationship, Drug , Down-Regulation/drug effects , Female , Fibronectins/metabolism , Humans , Neoplasm Invasiveness/pathology , Paxillin/metabolism , Phosphorylation/drug effects , Triterpenes/antagonists & inhibitors , Triterpenes/chemistry
3.
Acta Pharmacol Sin ; 39(3): 492-498, 2018 Mar.
Article in English | MEDLINE | ID: mdl-29168472

ABSTRACT

Deubiquitinating protease USP7 is a promising therapeutic target for cancer treatment, and interest in developing USP7 inhibitors has greatly increased. In the present study, we reported a series of natural pentacyclic triterpenes with USP7 inhibitory activity in vitro. Among them, both the ursane triterpenes and oleanane triterpenes were more active than the lupine triterpenes, whereas ursolic acid was the most potent with IC50 of 7.0±1.5 µmol/L. Molecular docking studies showed that ursolic acid might occupy the ubiquitin binding pocket of USP7, with the 17-carboxyl group and 3-hydroxyl group playing a vital role in the USP7-ursolic acid interaction. Using the cellular thermal shift assay, we demonstrated that ursolic acid interacted with USP7 in RPMI8226 human myeloma cells. Ursolic acid dose-dependently inhibited the proliferation of the myeloma cells with IC50 of 6.56 µmol/L, accompanied by reductions in USP7 substrates such as MDM2, UHRF1 and DNMT1. Overexpression of USP7 partially, but significantly attenuated ursolic acid-induced cell death as well as downregulation of MDM2, UHRF1 and DNMT1. In conclusion, we demonstrate for the first time that pentacyclic triterpenes represent a novel scaffold for developing USP7 inhibitors and that USP7 inhibition contributes to the anti-cancer effect of ursolic acid.


Subject(s)
Antineoplastic Agents/pharmacology , Neoplasms/drug therapy , Pentacyclic Triterpenes/pharmacology , Ubiquitin-Specific Peptidase 7/antagonists & inhibitors , CCAAT-Enhancer-Binding Proteins/metabolism , Cell Line, Tumor , Cell Proliferation/drug effects , DNA (Cytosine-5-)-Methyltransferase 1/metabolism , Dose-Response Relationship, Drug , Humans , Molecular Docking Simulation , Neoplasms/pathology , Proto-Oncogene Proteins c-mdm2/metabolism , Structure-Activity Relationship , Triterpenes/antagonists & inhibitors , Triterpenes/pharmacology , Ubiquitin-Protein Ligases , Ubiquitin-Specific Peptidase 7/biosynthesis , Ursolic Acid
4.
J Pharmacol Sci ; 134(1): 22-28, 2017 May.
Article in English | MEDLINE | ID: mdl-28522217

ABSTRACT

Chondrosarcomas (CS) is the second most frequent tumors of cartilage origin. A small compound extracted from Thunder God Vine (Tripterygium wilfordii Hook. F.) called celastrol can directly bound CIP2A protein and effectively inhibit cell proliferation and induce apoptosis in several cancer cells. However, little knowledge is concern about the important role of CIP2A in CS patients and the therapeutic value of celastrol on CS. Our results showed that CIP2A and c-MYC were verified to be oncoproteins by detecting their mRNA and protein expression in 10 human CS tissues by qRT-PCR and Western blots. After treatment of celastrol, the proliferation, migration and invasion were significantly inhibited; whereas the apoptosis was largely induced in human CS cell lines. In addition, celastrol inhibited the expression of CIP2A, c-MYC, and suppressed apoptotic proteins BAX and caspase-8 in human CS cells, on the other hand, it induced the expression of antiapoptotic protein Bcl-2. Finally, knockdown of CIP2A also inhibited the migration and invasion and induced apoptosis of human CS cells. To sum up, we found that celastrol had effects on inhibiting proliferation, migration, invasion and inducing apoptosis through suppression CIP2A/c-MYC signaling pathway in vitro, which may provide a new therapeutic regimen for CS.


Subject(s)
Autoantigens/metabolism , Chondrosarcoma/drug therapy , Membrane Proteins/metabolism , Proto-Oncogene Proteins c-myc/metabolism , Triterpenes/pharmacology , Apoptosis , Autoantigens/genetics , Caspase 8/genetics , Caspase 8/metabolism , Cell Line, Tumor , Cell Movement/drug effects , Cell Proliferation/drug effects , Cell Survival , Chondrosarcoma/metabolism , Humans , Intracellular Signaling Peptides and Proteins , Membrane Proteins/genetics , Neoplasm Invasiveness , Pentacyclic Triterpenes , Proto-Oncogene Proteins c-bcl-2/genetics , Proto-Oncogene Proteins c-bcl-2/metabolism , Proto-Oncogene Proteins c-myc/genetics , RNA, Messenger/genetics , RNA, Small Interfering , Signal Transduction/drug effects , Triterpenes/antagonists & inhibitors , Triterpenes/chemistry , Triterpenes/therapeutic use , bcl-2-Associated X Protein/genetics , bcl-2-Associated X Protein/metabolism
5.
Drug Des Devel Ther ; 11: 1127-1134, 2017.
Article in English | MEDLINE | ID: mdl-28435224

ABSTRACT

BACKGROUND: G-protein-coupled bile acid receptor 1 (GPBAR1, also known as TGR5) has been shown to participate in glucose homeostasis. In animal models, a TGR5 agonist increases incretin secretion to reduce hyperglycemia. Many agonists have been developed for clinical use. However, the effects of TGR5 blockade have not been studied extensively, with the exception of studies using TGR5 knockout mice. Therefore, we investigated the potential effect of triamterene on TGR5. METHODS: We transfected the TGR5 gene into cultured Chinese hamster ovary cells (CHO-K1 cells) to express TGR5. Then, we applied a fluorescent indicator to examine the glucose uptake of these transfected cells. In addition, NCI-H716 cells that secrete incretin were also evaluated. Fura-2, a fluorescence indicator, was applied to determine the changes in calcium concentrations. The levels of cyclic adenosine monophosphate (cAMP) and glucagon-like peptide (GLP-1) were estimated using enzyme-linked immunosorbent assay kits. Moreover, rats with streptozotocin (STZ)-induced type 1-like diabetes were used to investigate the effects in vivo. RESULTS: Triamterene dose dependently inhibits the increase in glucose uptake induced by TGR5 agonists in CHO-K1 cells expressing the TGR5 gene. In cultured NCI-H716 cells, TGR5 activation also increases GLP-1 secretion by increasing calcium levels. Triamterene inhibits the increased calcium levels by TGR5 activation through competitive antagonism. Moreover, the GLP-1 secretion and increased cAMP levels induced by TGR5 activation are both dose dependently reduced by triamterene. However, treatment with KB-R7943 at a dose sufficient to block the Na+/Ca2+ exchanger (NCX) failed to modify the responses to TGR5 activation in NCI-H716 cells or CHO-K1 cells expressing TGR5. Therefore, the inhibitory effects of triamterene on TGR5 activation do not appear to be related to NCX inhibition. Blockade of TGR5 activation by triamterene was further characterized in vivo using the STZ-induced diabetic rats. CONCLUSION: Based on the obtained data, we identified triamterene as a reliable inhibitor of TGR5. Therefore, triamterene can be developed as a clinical inhibitor of TGR5 activation in future studies.


Subject(s)
Diabetes Mellitus, Experimental/drug therapy , Diabetes Mellitus, Type 1/drug therapy , Receptors, G-Protein-Coupled/antagonists & inhibitors , Triamterene/pharmacology , Animals , CHO Cells , Cells, Cultured , Cricetulus , Diabetes Mellitus, Experimental/chemically induced , Diabetes Mellitus, Type 1/chemically induced , Dose-Response Relationship, Drug , Humans , Injections, Intravenous , Lithocholic Acid/antagonists & inhibitors , Lithocholic Acid/pharmacology , Male , Pentacyclic Triterpenes , Rats , Rats, Sprague-Dawley , Receptors, G-Protein-Coupled/genetics , Receptors, G-Protein-Coupled/metabolism , Streptozocin/administration & dosage , Structure-Activity Relationship , Triamterene/administration & dosage , Triterpenes/antagonists & inhibitors , Triterpenes/pharmacology , Betulinic Acid
6.
Acta Pharmacol Sin ; 37(7): 950-62, 2016 Jul.
Article in English | MEDLINE | ID: mdl-27292614

ABSTRACT

AIM: Tubeimoside-1 (TBMS1), a triterpenoid saponin extracted from the Chinese herbal medicine Bolbostemma paniculatum (Maxim) Franquet (Cucurbitaceae), has shown anticancer activities in various cancer cell lines. The aim of this study was to investigate the anticancer activity and molecular targets of TBMS1 in human prostate cancer cells in vitro. METHODS: DU145 and P3 human prostate cancer cells were treated with TBMS1. Cell viability and apoptosis were detected. ROS generation, mitochondrial membrane potential and cell cycle profile were examined. Western blotting was used to measure the expression of relevant proteins in the cells. RESULTS: TBMS1 (5-100 µmol/L) significantly suppressed the viability of DU145 and P3 cells with IC50 values of approximately 10 and 20 µmol/L, respectively. Furthermore, TBMS1 dose-dependently induced apoptosis and cell cycle arrest at G0/G1 phase in DU145 and P3 cells. In DU145 cells, TBMS1 induced mitochondrial apoptosis, evidenced by ROS generation, mitochondrial dysfunction, endoplasmic reticulum stress, modulated Bcl-2 family protein and cleaved caspase-3, and activated ASK-1 and its downstream targets p38 and JNK. The G0/G1 phase arrest was linked to increased expression of p53 and p21 and decreased expression of cyclin E and cdk2. Co-treatment with Z-VAD-FMK (pan-caspase inhibitor) could attenuate TBMS1-induced apoptosis but did not prevent G0/G1 arrest. Moreover, co-treatment with NAC (ROS scavenger), SB203580 (p38 inhibitor), SP600125 (JNK inhibitor) or salubrinal (ER stress inhibitor) significantly attenuated TBMS1-induced apoptosis. CONCLUSION: TBMS1 induces oxidative stress-mediated apoptosis in DU145 human prostate cancer cells in vitro via the mitochondrial pathway.


Subject(s)
Apoptosis/drug effects , Cell Cycle Checkpoints/drug effects , G1 Phase/drug effects , Oxidative Stress/drug effects , Prostatic Neoplasms/pathology , Resting Phase, Cell Cycle/drug effects , Saponins/pharmacology , Triterpenes/pharmacology , Acetylcysteine/pharmacology , Amino Acid Chloromethyl Ketones/pharmacology , Anthracenes/pharmacology , Caspase 3 , Cell Line, Tumor , Cell Survival/drug effects , Cinnamates/pharmacology , Cyclin E/metabolism , Cyclin-Dependent Kinase 2/metabolism , Dose-Response Relationship, Drug , Endoplasmic Reticulum Stress/drug effects , Humans , Imidazoles/pharmacology , JNK Mitogen-Activated Protein Kinases/metabolism , MAP Kinase Kinase Kinase 5/metabolism , Male , Membrane Potential, Mitochondrial/drug effects , Proto-Oncogene Proteins c-bcl-2/metabolism , Pyridines/pharmacology , Reactive Oxygen Species/metabolism , Saponins/antagonists & inhibitors , Thiourea/analogs & derivatives , Thiourea/pharmacology , Triterpenes/antagonists & inhibitors , p38 Mitogen-Activated Protein Kinases/metabolism
7.
Can J Physiol Pharmacol ; 94(5): 542-53, 2016 May.
Article in English | MEDLINE | ID: mdl-27070866

ABSTRACT

Astragaloside IV (ASI), a traditional Chinese medicine, is a main active ingredient of Astragalus membranaceus. Many clinical studies have found that ASI protects cardiomyocytes in cardiovascular diseases, but the underlying mechanisms remain obscure. The aim of this study was to investigate the molecular mechanisms responsible for the protective effects of ASI in cardiomyocytes from anoxia/reoxygenation (A/R) injury. According to the previous studies, we hypothesized that the cardioprotective effects of ASI against A/R injury might be associated with Notch1/Hes1 signaling pathway. In this study, neonatal rat primary cardiomyocytes were preconditioned with ASI prior to A/R injury. Our results showed that ASI effectively increased the cell viability, decreased the content of MDA, decreased the activities of CPK and LDH, increased the activities of GSH-Px and SOD, and reduced the reactive oxygen species (ROS) generation and the loss of mitochondrial membrane potential (Δψm). ASI inhibited the mitochondrial permeability transition pore (mPTP) opening and activation of caspase-3, and finally decreased the cell apoptosis in cardiomyocytes. Furthermore, ASI upregulated Hes1 protein expression. However, pretreatment with DAPT, a Notch1 inhibitor, effectively attenuated the cardioprotective effects of ASI against A/R injury, except MDA, SOD, GSH-Px, and the ROS generation. Taken together, we demonstrated that ASI could protect against A/R injury via the Notch1/Hes1 signaling pathway.


Subject(s)
Cardiotonic Agents/pharmacology , Drugs, Chinese Herbal/pharmacology , Myocardial Reperfusion Injury/prevention & control , Myocytes, Cardiac/drug effects , Saponins/pharmacology , Transcription Factor HES-1/agonists , Triterpenes/pharmacology , Up-Regulation/drug effects , Animals , Animals, Newborn , Apoptosis/drug effects , Astragalus propinquus/chemistry , Cardiotonic Agents/antagonists & inhibitors , Caspase 3/chemistry , Caspase 3/metabolism , Cells, Cultured , Diamines/pharmacology , Drugs, Chinese Herbal/chemistry , Enzyme Activation/drug effects , Lipid Peroxidation/drug effects , Membrane Potential, Mitochondrial/drug effects , Mitochondrial Membrane Transport Proteins/antagonists & inhibitors , Mitochondrial Membrane Transport Proteins/metabolism , Mitochondrial Permeability Transition Pore , Myocardial Reperfusion Injury/metabolism , Myocardial Reperfusion Injury/pathology , Myocytes, Cardiac/cytology , Myocytes, Cardiac/metabolism , Myocytes, Cardiac/pathology , Rats, Sprague-Dawley , Reactive Oxygen Species/antagonists & inhibitors , Reactive Oxygen Species/metabolism , Receptor, Notch1/antagonists & inhibitors , Receptor, Notch1/metabolism , Saponins/antagonists & inhibitors , Signal Transduction/drug effects , Thiazoles/pharmacology , Transcription Factor HES-1/metabolism , Triterpenes/antagonists & inhibitors
8.
Planta Med ; 81(12-13): 1141-5, 2015 Aug.
Article in English | MEDLINE | ID: mdl-26287693

ABSTRACT

The marine sponge-derived fungus Auxarthron reticulatum produces the cannabinoid receptor antagonist amauromine (1). Recultivation of the fungus to obtain further amounts for more detailed pharmacological evaluation of 1 additionally yielded the novel triterpene glycoside auxarthonoside (2), bearing, in nature, a rather rare sugar moiety, i.e., N-acetyl-6-methoxy-glucosamine. Amauromine (1), which inhibited cannabinoid CB1 receptors (Ki 0.178 µM) also showed antagonistic activity at the cannabinoid-like orphan receptor GPR18 (IC50 3.74 µM). The diketopiperazine 1 may thus serve as a lead structure for the development of more potent and selective GPR18 antagonists, which are required to study the orphan receptor's potential as a new drug target. Despite the execution of many biological assays, to date, no bioactivity could be found for auxarthonoside (2).


Subject(s)
Alkaloids/chemistry , Ascomycota/chemistry , Cannabinoid Receptor Antagonists/chemistry , Indoles/chemistry , Porifera/microbiology , Receptors, G-Protein-Coupled/antagonists & inhibitors , Triterpenes/antagonists & inhibitors , Alkaloids/antagonists & inhibitors , Alkaloids/isolation & purification , Animals , Cannabinoid Receptor Antagonists/isolation & purification , Glycosides/antagonists & inhibitors , Humans , Indoles/antagonists & inhibitors , Indoles/isolation & purification , Molecular Structure , Receptor, Cannabinoid, CB1/antagonists & inhibitors , Receptor, Cannabinoid, CB2/antagonists & inhibitors
9.
Eur J Pharmacol ; 750: 167-75, 2015 Mar 05.
Article in English | MEDLINE | ID: mdl-25617794

ABSTRACT

The current study was aimed to investigate the gastroprotective effects of friedelin isolated from the hexane extract of leaves of Azima tetracantha. Ethanol-induced gastric ulcer model was used to investigate the gastroprotective effects of friedelin. Antioxidant enzymes, lipid peroxidation, nitric oxide, gastric vascular permeability, pro and anti-inflammatory cytokines and apoptosis level have been investigated. Ethanol caused severe gastric damage and friedelin pretreatment protected against its deleterious role. Antioxidant enzyme activities, anti-inflammatory cytokines, prostaglandin E2 (PGE2), constitutive nitric oxide synthase (cNOS) and mucus weight have been increased significantly. However, the vascular permeability, pro-inflammatory cytokines, inducible nitric oxide synthase (iNOS), caspase-3 and apoptosis level have significantly been decreased after friedelin ingestion. The present study has clearly demonstrated the anti-ulcer potential of friedelin, these findings suggested that friedelin could be a new useful natural gastroprotective tool against gastric ulcer.


Subject(s)
Cytoprotection/drug effects , Ethanol/adverse effects , Salvadoraceae/chemistry , Stomach Ulcer/chemically induced , Stomach Ulcer/prevention & control , Triterpenes/isolation & purification , Triterpenes/pharmacology , Animals , Apoptosis/drug effects , Capillary Permeability/drug effects , Caspase 3/metabolism , Catalase/metabolism , Cytokines/metabolism , Dinoprostone/metabolism , Female , Gastric Mucosa/drug effects , Gastric Mucosa/enzymology , Gastric Mucosa/metabolism , Gastric Mucosa/pathology , Glutathione Peroxidase/metabolism , Hydrogen-Ion Concentration , Male , Malondialdehyde/metabolism , Nitric Oxide/metabolism , Nitric Oxide Synthase Type II/metabolism , Peroxidase/metabolism , Rats , Rats, Wistar , Stomach Ulcer/metabolism , Stomach Ulcer/pathology , Superoxide Dismutase/metabolism , Triterpenes/antagonists & inhibitors
10.
Toxicol Lett ; 227(1): 65-73, 2014 May 16.
Article in English | MEDLINE | ID: mdl-24680927

ABSTRACT

Ziyuglycoside II, a triterpenoid saponin compound extracted from Sanguisorba officinalis L., has been reported to have a wide range of clinical applications including anti-cancer effect. In this study, the anti-proliferative effect of ziyuglycoside II in two classic human breast cancer cell lines, MCF-7 and MDA-MB-231, was extensively investigated. Our study indicated that ziyuglycoside II could effectively induce G2/M phase arrest and apoptosis in both cell lines. Cell cycle blocking was associated with the down-regulation of Cdc25C, Cdc2, cyclin A and cyclin B1 as well as the up-regulation of p21/WAF1, phospho-Cdc25C and phospho-Cdc2. Ziyuglycoside II treatment also induced reactive oxygen species (ROS) production and apoptosis by activating the extrinsic/Fas/FasL pathway as well as the intrinsic/mitochondrial pathway. More importantly, the c-Jun NH2-terminal kinase (JNK), a downstream target of ROS, was found to be a critical mediator of ziyuglycoside II-induced cell apoptosis. Further knockdown of JNK by siRNA could inhibit ziyuglycoside II-mediated apoptosis with attenuating the up-regulation of Bax and Fas/FasL as well as the down-regulation of Bcl-2. Taken together, the cell death of breast cancer cells in response to ziyuglycoside II was dependent upon cell cycle arrest and cell apoptosis via a ROS-dependent JNK activation pathway. Our findings may significantly contribute to the understanding of the anti-proliferative effect of ziyuglycoside II, in particular to breast carcinoma and provide novel insights into the potential application of such compound in breast cancer therapy.


Subject(s)
Antineoplastic Agents, Phytogenic/pharmacology , Apoptosis/drug effects , Breast Neoplasms/drug therapy , G2 Phase/drug effects , MAP Kinase Signaling System/drug effects , Reactive Oxygen Species/metabolism , Saponins/pharmacology , Antineoplastic Agents, Phytogenic/antagonists & inhibitors , Breast Neoplasms/metabolism , Carcinoma/drug therapy , Carcinoma/metabolism , Cell Line , Cell Line, Tumor , Cell Proliferation/drug effects , Female , Gene Expression Regulation, Neoplastic/drug effects , Gene Silencing , Humans , Mammary Glands, Human/drug effects , Mammary Glands, Human/metabolism , Membrane Potential, Mitochondrial/drug effects , Neoplasm Proteins/agonists , Neoplasm Proteins/antagonists & inhibitors , Neoplasm Proteins/genetics , Neoplasm Proteins/metabolism , RNA, Small Interfering , Receptors, Estrogen/metabolism , Receptors, Progesterone/metabolism , Saponins/adverse effects , Saponins/antagonists & inhibitors , Triterpenes/adverse effects , Triterpenes/antagonists & inhibitors , Triterpenes/pharmacology
11.
PLoS One ; 8(9): e74782, 2013.
Article in English | MEDLINE | ID: mdl-24086374

ABSTRACT

ATP-binding cassette transporter A1 (ABCA1) is critical in exporting cholesterol from macrophages and plays a protective role in the development of atherosclerosis. The purpose of this study was to investigate the effects of betulinic acid (BA), a pentacyclic triterpenoid, on ABCA1 expression and cholesterol efflux, and to further determine the underlying mechanism. BA promoted ABCA1 expression and cholesterol efflux, decreased cellular cholesterol and cholesterol ester content in LPS-treated macrophages. Furthermore, we found that BA promoted ABCA1 expression via down-regulation of miR-33s. The inhibition of LPS-induced NF-κB activation further decreased miR-33s expression and enhanced ABCA1 expression and cholesterol efflux when compared with BA only treatment. In addition, BA suppressed IκB phosphorylation, p65 phosphorylation and nuclear translocation, and the transcription of NF-κB-dependent related gene. Moreover, BA reduced atherosclerotic lesion size, miR-33s levels and NF-κB activation, and promoted ABCA1 expression in apoE(-/-) mice. Taken together, these results reveal a novel mechanism for the BA-mediated ABCA1 expression, which may provide new insights for developing strategies for modulating vascular inflammation and atherosclerosis.


Subject(s)
ATP Binding Cassette Transporter 1/antagonists & inhibitors , Cholesterol/metabolism , Lipopolysaccharides/pharmacology , MicroRNAs/genetics , NF-kappa B/metabolism , Triterpenes/antagonists & inhibitors , Triterpenes/pharmacology , ATP Binding Cassette Transporter 1/metabolism , Animals , Apolipoproteins E/deficiency , Apolipoproteins E/metabolism , Atherosclerosis/blood , Atherosclerosis/metabolism , Atherosclerosis/pathology , Biological Transport/drug effects , Body Weight/drug effects , Cell Line , Cell Nucleus/drug effects , Cell Nucleus/metabolism , Gene Expression Regulation/drug effects , Humans , Lipids/blood , Macrophages/drug effects , Macrophages/metabolism , Male , Mice , MicroRNAs/metabolism , Models, Biological , Pentacyclic Triterpenes , Protein Transport/drug effects , Signal Transduction/drug effects , Signal Transduction/genetics , Betulinic Acid
13.
Anticancer Drugs ; 22(2): 158-65, 2011 Feb.
Article in English | MEDLINE | ID: mdl-20948428

ABSTRACT

Ursolic acid (UA), a pentacyclic triterpenoid compound, is widely distributed in the plant kingdom and has a broad range of biological effects. This study was carried out for the first time to investigate the potential role of UA in the differentiation of human leukemia HL60 cells and the underlying mechanisms in it. UA could induce differentiation of HL60 cells into the monocytic lineage, as assessed by the morphological change, nitroblue tetrazolium reduction assay, and expression of CD14 and CD11b surface antigens. Moreover, UA activated the extracellular signal-regulated kinase (ERK) pathway in both dose-dependent and time-dependent manners. Inhibiting ERK pathway activation with PD98059 could significantly block the differentiation induced by UA. Consistent with the induced differentiation, the upregulation of CCAAT/enhancer-binding protein ß by UA was also eliminated by PD98059. Taken together, the results reported here show that UA can promote the monocytic differentiation of HL60 cells and increase the expression of CCAAT/enhancer-binding protein ß by activating the ERK pathway, suggesting that UA could be a potential candidate as a differentiation-inducing agent for the therapeutic treatment of leukemia.


Subject(s)
Antineoplastic Agents, Phytogenic/pharmacology , CCAAT-Enhancer-Binding Protein-beta/biosynthesis , Extracellular Signal-Regulated MAP Kinases/metabolism , Leukemia, Myeloid, Acute/drug therapy , MAP Kinase Signaling System/drug effects , Triterpenes/pharmacology , CCAAT-Enhancer-Binding Protein-beta/genetics , CCAAT-Enhancer-Binding Protein-beta/metabolism , Cell Differentiation/drug effects , Cell Growth Processes/drug effects , Cell Line, Tumor , Extracellular Signal-Regulated MAP Kinases/antagonists & inhibitors , Flavonoids/pharmacology , HCT116 Cells , HL-60 Cells , Humans , Leukemia, Myeloid, Acute/genetics , Leukemia, Myeloid, Acute/metabolism , Leukemia, Myeloid, Acute/pathology , Phosphorylation/drug effects , Proto-Oncogene Proteins c-myc/biosynthesis , Proto-Oncogene Proteins c-myc/genetics , Triterpenes/antagonists & inhibitors , U937 Cells , Up-Regulation/drug effects , Ursolic Acid
14.
Mar Drugs ; 8(1): 80-90, 2010 Jan 19.
Article in English | MEDLINE | ID: mdl-20161972

ABSTRACT

Geoditin A, an isomalabaricane triterpene isolated from the marine sponge Geodia japonica, has been demonstrated to dissipate mitochondrial membrane potential, activate caspase 3, decrease cytoplasmic proliferating cell nuclear antigen (PCNA), and induce apoptosis of leukemia cells, but the underlying mechanism remains unclear [1]. In this study, we found fragmentation of Golgi structure, suppression of transferrin receptor expression, production of oxidants, and DNA fragmentation in human colon cancer HT29 cells after treatment with geoditin A for 24 h. This apoptosis was not abrogated by chelation of intracellular iron with salicylaldehyde isonicotinoyl hydrazone (SIH), but suppressed by N-acetylcysteine (NAC), a thiol antioxidant and GSH precursor, indicating that the cytotoxic effect of geoditin A is likely mediated by a NAC-inhibitable oxidative stress. Our results provide a better understanding of the apoptotic properties and chemotherapeutical potential of this marine triterpene.


Subject(s)
Antineoplastic Agents/pharmacology , Apoptosis/drug effects , Marine Toxins/toxicity , Oxidative Stress/drug effects , Resorcinols/pharmacology , Triterpenes/pharmacology , Acetylcysteine/pharmacology , Adenocarcinoma/drug therapy , Adenocarcinoma/metabolism , Adenocarcinoma/pathology , Animals , Antineoplastic Agents/antagonists & inhibitors , Cell Proliferation/drug effects , Colonic Neoplasms/drug therapy , Colonic Neoplasms/metabolism , Colonic Neoplasms/pathology , DNA Fragmentation/drug effects , Dose-Response Relationship, Drug , Down-Regulation/drug effects , Endocytosis/drug effects , Free Radical Scavengers/pharmacology , Golgi Apparatus/drug effects , Golgi Apparatus/pathology , HT29 Cells , Humans , Inhibitory Concentration 50 , Iron Chelating Agents/pharmacology , Marine Toxins/antagonists & inhibitors , Oxidants/metabolism , Receptors, Transferrin/metabolism , Resorcinols/antagonists & inhibitors , Triterpenes/antagonists & inhibitors
15.
Biochim Biophys Acta ; 1783(9): 1632-41, 2008 Sep.
Article in English | MEDLINE | ID: mdl-18501196

ABSTRACT

mRNA for four-transmembrane L6 family member 5 (TM4SF5), a homolog of tumor antigen L6, was previously shown to be highly expressed in diverse tumors. We recently found that human hepatocarcinoma tissues also overexpressed TM4SF5 protein, in comparison to normal liver tissues. We also found that tiarellic acid (TA) caused cell detachment-related apoptosis in cells expressing endogenous or stably-overexpressing TM4SF5. When cells expressing TM4SF5 were treated with TA, we observed reduced phosphorylation of focal adhesion kinase, paxillin, and p130Cas, but not c-Src. TA treatment also caused focal adhesion loss and reduced cell adhesion, and increased the numbers of floating cells and apoptotic cells. These effects were blocked by overexpression of focal adhesion molecules, suggesting that treatment with TA mediates anoikis of TM4SF5-expressing cells. However, TM4SF5-null cells were not affected by TA, indicating that these effects occur specifically in TM4SF5-positive cells. TA administration reduced tumor formation in nude mice injected with TM4SF5-expressing cells, presumably through increased apoptosis in TM4SF5-positive tumors. These observations indicate that TM4SF5-positive tumorigenesis can be inhibited by TA via induction of cell detachment-related apoptosis, and suggest that TA may be developed as a putative therapeutic reagent against TM4SF5-positive tumorigenesis.


Subject(s)
Antineoplastic Agents/pharmacology , Membrane Proteins/antagonists & inhibitors , Neoplasms/metabolism , Triterpenes/pharmacology , Animals , Antineoplastic Agents/antagonists & inhibitors , Antineoplastic Agents/chemistry , Apoptosis , Cell Adhesion , Cell Line, Tumor , Crk-Associated Substrate Protein/metabolism , Female , Focal Adhesion Protein-Tyrosine Kinases/metabolism , Focal Adhesions/metabolism , Humans , Membrane Proteins/metabolism , Mice , Mice, Nude , Neoplasms/etiology , Neoplasms/pathology , Paxillin/metabolism , Triterpenes/antagonists & inhibitors , Triterpenes/chemistry
16.
J Pharm Pharmacol ; 57(9): 1221-9, 2005 Sep.
Article in English | MEDLINE | ID: mdl-16105244

ABSTRACT

Axonal regeneration is important for functional recovery following nerve damage. Centella asiatica Urban herb, also known as Hydrocotyle asiatica L., has been used in Ayurvedic medicine for centuries as a nerve tonic. Here, we show that Centella asiatica ethanolic extract (100 microg mL-1) elicits a marked increase in neurite outgrowth in human SH-SY5Y cells in the presence of nerve growth factor (NGF). However, a water extract of Centella was ineffective at 100 microg mL-1. Sub-fractions of Centella ethanolic extract, obtained through silica-gel chromatography, were tested (100 microg mL-1) for neurite elongation in the presence of NGF. Greatest activity was found with a non-polar fraction (GKF4). Relatively polar fractions (GKF10 to GKF13) also showed activity, albeit less than GKF4. Thus, Centella contains more than one active component. Asiatic acid (AA), a triterpenoid compound found in Centella ethanolic extract and GKF4, showed marked activity at 1 microM (microg mL-1). AA was not present in GKF10 to GKF13, further indicating that other active components must be present. Neurite elongation by AA was completely blocked by the extracellular-signal-regulated kinase (ERK) pathway inhibitor PD 098059 (10 microM). Male Sprague-Dawley rats given Centella ethanolic extract in their drinking water (300-330 mg kg-1 daily) demonstrated more rapid functional recovery and increased axonal regeneration (larger calibre axons and greater numbers of myelinated axons) compared with controls, indicating that the axons grew at a faster rate. Taken together, our findings indicate that components in Centella ethanolic extract may be useful for accelerating repair of damaged neurons.


Subject(s)
Administration, Oral , Centella/chemistry , Nerve Regeneration/drug effects , Neurites/drug effects , Animals , Cell Line, Tumor , Disease Models, Animal , Ethanol/chemistry , Flavonoids/pharmacology , Humans , Male , Medicine, Ayurvedic , Nerve Crush , Nerve Regeneration/physiology , Neurites/ultrastructure , Pentacyclic Triterpenes , Plant Extracts/chemistry , Plant Extracts/isolation & purification , Plant Extracts/pharmacology , Rats , Rats, Sprague-Dawley , Sciatic Nerve/drug effects , Sciatic Nerve/pathology , Sciatic Nerve/physiology , Triterpenes/antagonists & inhibitors , Triterpenes/chemistry , Triterpenes/pharmacology
17.
Anticancer Drugs ; 15(6): 619-24, 2004 Jul.
Article in English | MEDLINE | ID: mdl-15205607

ABSTRACT

Betulinic acid (BA) is a pentacyclic triterpene found in a number of medicinal plants and has been shown to cause apoptosis in a number of cell lines. We report here that BA may also have an effect on HL-60 cell differentiation. BA was cytotoxic to HL-60 cells with an IC50 of 5.7 microM after a 72-h treatment. Flow cytometry analysis showed that after exposure to 1-12 microM of BA for 72 h, approximately 10% of viable cells were in the sub-G1, presumably apoptotic, phase. At the same time differentiation was induced in approximately 10% (at 1 microM BA) to a maximum of 20% (at 6 microM BA) of cells as judged by the NBT-reduction test, and the expression of membrane markers CD11b and CD14. On the other hand, at 1 and 5 nM, 1alpha,25-dihydroxyvitamin D3 (DHD3) induced differentiation in approximately 10 and 70% of cells, respectively. At 1 nM DHD3, the addition of 1 microM BA increased differentiated cells from 10 to 43% and with 3 microM BA the increase was to 80%. BA also enhanced the effects of DHD3 in the expansion of the G1 cell population with a concomitant decrease of S phase cells. The effects of DHD3 and BA on CD11b and CD14 expression were inhibited by PD98059, a MEK inhibitor. Our results suggest that BA may enhance the effect of DHD3 in inducing mitogen-activated protein kinase kinase/extracellular signal-regulated protein kinase-mediated HL-60 cell differentiation.


Subject(s)
Calcitriol/pharmacology , HL-60 Cells/cytology , Triterpenes/pharmacology , Apoptosis/drug effects , CD11b Antigen/biosynthesis , CD11b Antigen/genetics , Cell Differentiation/drug effects , Cell Division/drug effects , Dose-Response Relationship, Drug , Drug Synergism , Enzyme Activation/drug effects , Enzyme Activation/genetics , Enzyme Activation/physiology , Flavonoids/pharmacology , G1 Phase/drug effects , G1 Phase/physiology , Gene Expression/drug effects , Gene Expression/genetics , Humans , Lipopolysaccharide Receptors/biosynthesis , Lipopolysaccharide Receptors/genetics , Mitogen-Activated Protein Kinase Kinases/antagonists & inhibitors , Mitogen-Activated Protein Kinase Kinases/drug effects , Mitogen-Activated Protein Kinase Kinases/metabolism , Monocytes/cytology , Monocytes/drug effects , Pentacyclic Triterpenes , Triterpenes/antagonists & inhibitors , Triterpenes/chemistry , Betulinic Acid
18.
Pharmacol Toxicol ; 86(2): 53-8, 2000 Feb.
Article in English | MEDLINE | ID: mdl-10728914

ABSTRACT

This study was designed to investigate the effect of cAMP on ursolic acid-induced apoptosis of HL-60 cells. Ursolic acid decreased the viability of the cells in a dose-dependent manner, which was revealed as an apototic process characterized by ladder-pattern DNA fragmentation in agarose gel electrophoresis and segmented nuclei in DAPI-sulpharhodamin 101 staining. Ursolic acid-induced apoptosis of the cells was markedly inhibited by the addition of cAMP-elevating agents including DB-cAMP, CPT-cAMP, 8-Br-cAMP and forskolin. These results were further evidenced by the fact that inhibitors of cAMP-dependent protein kinase including H89 and KT5720 completely inhibited the cAMP-mediated rescue of HL-60 cells from ursolic acid-induced apoptosis. In addition, differentiating agents of the cells such as dimethyl sulfoxide and retinoic acid did not affect the ursolic acid-induced apoptosis of HL-60 cells. These results suggest that signaling pathway of cAMP-dependent activation of protein kinase A may affect the responsiveness of tumor cells upon ursolic acid.


Subject(s)
Antineoplastic Agents, Phytogenic/antagonists & inhibitors , Apoptosis/drug effects , Carbazoles , Cyclic AMP-Dependent Protein Kinases/metabolism , Cyclic AMP/pharmacology , HL-60 Cells/enzymology , Sulfonamides , Triterpenes/antagonists & inhibitors , Antineoplastic Agents, Phytogenic/pharmacology , Cell Survival/drug effects , Cyclic AMP-Dependent Protein Kinases/antagonists & inhibitors , Dimethyl Sulfoxide/pharmacology , Dose-Response Relationship, Drug , Electrophoresis, Agar Gel , Enzyme Activation , Enzyme Inhibitors/pharmacology , Flow Cytometry , Humans , Indoles/pharmacology , Isoquinolines/pharmacology , Pyrroles/pharmacology , Tretinoin/pharmacology , Triterpenes/pharmacology , Ursolic Acid
19.
J Biol Chem ; 274(36): 25237-44, 1999 Sep 03.
Article in English | MEDLINE | ID: mdl-10464244

ABSTRACT

Correolide, a novel nortriterpene natural product, potently inhibits the voltage-gated potassium channel, K(v)1.3, and [(3)H]dihydrocorreolide (diTC) binds with high affinity (K(d) approximately 10 nM) to membranes from Chinese hamster ovary cells that express K(v)1.3 (Felix, J. P., Bugianesi, R. M., Schmalhofer, W. A., Borris, R., Goetz, M. A., Hensens, O. D., Bao, J.-M., Kayser, F. , Parsons, W. H., Rupprecht, K., Garcia, M. L., Kaczorowski, G. J., and Slaughter, R. S. (1999) Biochemistry 38, 4922-4930). Mutagenesis studies were used to localize the diTC binding site and to design a high affinity receptor in the diTC-insensitive channel, K(v)3.2. Transferring the pore from K(v)1.3 to K(v)3.2 produces a chimera that binds peptidyl inhibitors of K(v)1.3 with high affinity, but not diTC. Transfer of the S(5) region of K(v)1.3 to K(v)3.2 reconstitutes diTC binding at 4-fold lower affinity as compared with K(v)1.3, whereas transfer of the entire S(5)-S(6) domain results in a normal K(v)1.3 phenotype. Substitutions in S(5)-S(6) of K(v)1.3 with nonconserved residues from K(v)3.2 has identified two positions in S(5) and one in S(6) that cause significant alterations in diTC binding. High affinity diTC binding can be conferred to K(v)3.2 after substitution of these three residues with the corresponding amino acids found in K(v)1.3. These results suggest that lack of sensitivity of K(v)3.2 to diTC is a consequence of the presence of Phe(382) and Ile(387) in S(5), and Met(458) in S(6). Inspection of K(v)1.1-1.6 channels indicates that they all possess identical S(5) and S(6) domains. As expected, diTC binds with high affinity (K(d) values 7-21 nM) to each of these homotetrameric channels. However, the kinetics of binding are fastest with K(v)1.3 and K(v)1.4, suggesting that conformations associated with C-type inactivation will facilitate entry and exit of diTC at its binding site. Taken together, these findings identify K(v)1 channel regions necessary for high affinity diTC binding, as well as, reveal a channel conformation that markedly influences the rate of binding of this ligand.


Subject(s)
Potassium Channels/metabolism , Triterpenes/metabolism , Amino Acid Sequence , Animals , Binding Sites , CHO Cells , Cricetinae , Humans , Kinetics , Molecular Sequence Data , Potassium Channels/chemistry , Protein Binding , Recombinant Fusion Proteins/chemistry , Recombinant Fusion Proteins/metabolism , Triterpenes/antagonists & inhibitors
20.
Biochemistry ; 38(16): 4922-30, 1999 Apr 20.
Article in English | MEDLINE | ID: mdl-10213593

ABSTRACT

A novel nortriterpene, termed correolide, purified from the tree Spachea correae, inhibits Kv1.3, a Shaker-type delayed rectifier potassium channel present in human T lymphocytes. Correolide inhibits 86Rb+ efflux through Kv1.3 channels expressed in CHO cells (IC50 86 nM; Hill coefficient 1) and displays a defined structure-activity relationship. Potency in this assay increases with preincubation time and with time after channel opening. Correolide displays marked selectivity against numerous receptors and voltage- and ligand-gated ion channels. Although correolide is most potent as a Kv1.3 inhibitor, it blocks all other members of the Kv1 family with 4-14-fold lower potency. C20-29-[3H]dihydrocorreolide (diTC) was prepared and shown to bind in a specific, saturable, and reversible fashion (Kd = 11 nM) to a single class of sites in membranes prepared from CHO/Kv1.3 cells. The molecular pharmacology and stoichiometry of this binding reaction suggest that one diTC site is present per Kv1.3 channel tetramer. This site is allosterically coupled to peptide and potassium binding sites in the pore of the channel. DiTC binding to human brain synaptic membranes identifies channels composed of other Kv1 family members. Correolide depolarizes human T cells to the same extent as peptidyl inhibitors of Kv1.3, suggesting that it is a candidate for development as an immunosuppressant. Correolide is the first potent, small molecule inhibitor of Kv1 series channels to be identified from a natural product source and will be useful as a probe for studying potassium channel structure and the physiological role of such channels in target tissues of interest.


Subject(s)
Ion Channel Gating/drug effects , Potassium Channel Blockers , Potassium Channels, Voltage-Gated , T-Lymphocytes/metabolism , Triterpenes/chemistry , Triterpenes/pharmacology , Animals , Binding Sites/drug effects , CHO Cells , Cell Line , Charybdotoxin/pharmacology , Cricetinae , Humans , Immunosuppressive Agents/antagonists & inhibitors , Immunosuppressive Agents/chemistry , Immunosuppressive Agents/metabolism , Immunosuppressive Agents/pharmacology , Kv1.3 Potassium Channel , Membrane Potentials/drug effects , Neurotoxins/pharmacology , Potassium Channels/metabolism , Rubidium Radioisotopes/metabolism , Scorpion Venoms/pharmacology , Synaptic Membranes/drug effects , Synaptic Membranes/metabolism , T-Lymphocytes/drug effects , Triterpenes/antagonists & inhibitors , Triterpenes/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...