Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 108
Filter
1.
Biomed Pharmacother ; 130: 110539, 2020 Oct.
Article in English | MEDLINE | ID: mdl-32768880

ABSTRACT

Side effects of chemotherapy are burning questions for physicians and patients involved in cancers. Ganoderma lucidum is a widely consumed traditional Chinese medicine and edible mushroom with multiple functional properties. The present study aims to investigate the potential of polysaccharides from spore of G. lucidum (SGP) on small intestinal barrier function recovery against paclitaxel (PTX) challenge in a breast cancer mice model and IEC-6 cell line. The 4T1 tumor-bearing mice were treated with PTX together with four-week daily oral administration of SGP. Results indicated that combination of PTX and SGP reversed body weight lost and remolded the histology of small intestine, accompanied with promoted proliferation but suppressed apoptosis in intestinal cells. Intestinal barrier function was enhanced by the combination as indicated by reduced endotoxemia and the up-regulation of tight junction proteins, including Zonula occludens-1 (ZO-1), E-cadherin, ß-catenin and Occludin. The protection of SGP was further confirmed in IEC-6 cells affected by PTX in vitro. The combination treatment prevented PTX-induced apoptosis in IEC-6 by inhibiting microtubule polymerization, and the aforementioned tight junction proteins were also upregulated. These findings suggest a promising protective effect of SGP against small intestinal barrier injury caused by PTX, highlighting its clinical implication against the chemotherapy side effects.


Subject(s)
Antineoplastic Agents, Phytogenic/toxicity , Apoptosis/drug effects , Intestinal Mucosa/drug effects , Microtubules/drug effects , Paclitaxel/toxicity , Polysaccharides/pharmacology , Reishi/chemistry , Spores, Fungal/chemistry , Animals , Antineoplastic Agents, Phytogenic/antagonists & inhibitors , Cell Line, Tumor , Cell Proliferation/drug effects , Epithelial Cells/drug effects , Female , Humans , Intestinal Mucosa/cytology , Mice , Mice, Inbred BALB C , Paclitaxel/antagonists & inhibitors , Polysaccharides/chemistry , Tight Junction Proteins/biosynthesis , Weight Loss/drug effects
2.
Biomed Pharmacother ; 129: 110441, 2020 Sep.
Article in English | MEDLINE | ID: mdl-32580047

ABSTRACT

Caffeine (1,3,7-trimethylxanthine) is a xanthine alkaloid found in a number of dietary products consumed worldwide, such as coffee, tea, and soft beverages, and is known to act as a modifying agent for cytotoxic chemotherapeutic drugs. Studies have shown that caffeine reduces the cytotoxic effects of paclitaxel and inhibits paclitaxel-induced apoptosis; however, the underlying mechanism remains unclear. Here, we investigated whether caffeine inhibits the antitumor activity of paclitaxel via down-regulation of α-tubulin acetylation. In vitro studies, involving MTT assay, wound-healing assay, cell apoptosis assay, and western blotting analysis of A549 and HeLa cells, were performed. A549 and HeLa cell-based xenografts were established, and western blotting and immunohistochemical staining were performed for in vivo studies. The results showed that caffeine promoted the growth of cancer cells treated with paclitaxel. Additionally, caffeine enhanced migration ability, inhibited apoptosis, and decreased the acetylation of α-tubulin in paclitaxel-treated cancer cells. Furthermore, caffeine decreased the inhibitory effect of paclitaxel on tumor growth through down-regulation of α-tubulin acetylation in vivo. Taken together, these findings demonstrate that caffeine inhibits the anticancer activity of paclitaxel via down-regulation of α-tubulin acetylation, suggesting that patients receiving treatment with taxanes, such as paclitaxel, should avoid consuming caffeinated beverages or foods.


Subject(s)
Antineoplastic Agents, Phytogenic/antagonists & inhibitors , Caffeine/pharmacology , Lung Neoplasms/drug therapy , Paclitaxel/antagonists & inhibitors , Tubulin/metabolism , Uterine Cervical Neoplasms/drug therapy , A549 Cells , Acetylation , Animals , Antineoplastic Agents, Phytogenic/pharmacology , Apoptosis/drug effects , Cell Movement/drug effects , Cell Proliferation/drug effects , Drug Interactions , Female , HeLa Cells , Humans , Lung Neoplasms/metabolism , Lung Neoplasms/pathology , Mice, Inbred BALB C , Mice, Nude , Paclitaxel/pharmacology , Protein Processing, Post-Translational , Signal Transduction , Tumor Burden/drug effects , Uterine Cervical Neoplasms/metabolism , Uterine Cervical Neoplasms/pathology , Xenograft Model Antitumor Assays
3.
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
4.
Apoptosis ; 22(11): 1473-1484, 2017 Nov.
Article in English | MEDLINE | ID: mdl-28913568

ABSTRACT

Cholangiocarcinoma (CCA) is an aggressive, metastatic bile duct cancer. CCA is difficult to diagnose, and responds poorly to current radio- and chemo-therapy. Piperlongumine (PL) is a naturally-occurring small molecule selectively toxic to cancer cells by targeting reactive oxygen species (ROS). In this study, we demonstrated the potential anticancer activity of PL in CCA. PL markedly induced death in CCA cell lines in a dose- and time-dependent manner through the activation of caspase-3 and PARP. PL also stimulated ROS accumulation in CCA. Co-exposure of PL with the ROS scavenger N-acetyl-L-cysteine or GSH completely blocked PL-induced apoptosis in CCA cell lines. Increased p21 via the p53-independent pathway in PL-treated CCA cells led to G2/M phase arrest and cell apoptosis. In addition, the study showed that PL trigger CCA cell lines death through JNK-ERK activation. Furthermore, the different antioxidant capacity of CCA cell lines also indicates the susceptibility of the cells to PL treatment. Our findings reveal that PL exhibits anti-tumor activity and has potential to be used as a chemotherapeutic agent against CCA.


Subject(s)
Antineoplastic Agents, Phytogenic/pharmacology , Dioxolanes/pharmacology , G2 Phase Cell Cycle Checkpoints/drug effects , Gene Expression Regulation, Neoplastic/drug effects , Piper/chemistry , Reactive Oxygen Species/agonists , Acetylcysteine/pharmacology , Antineoplastic Agents, Phytogenic/antagonists & inhibitors , Antineoplastic Agents, Phytogenic/isolation & purification , Apoptosis/drug effects , Bile Ducts/drug effects , Bile Ducts/metabolism , Bile Ducts/pathology , Caspase 3/genetics , Caspase 3/metabolism , Cell Line, Tumor , Cyclin-Dependent Kinase Inhibitor p21/genetics , Cyclin-Dependent Kinase Inhibitor p21/metabolism , Dioxolanes/antagonists & inhibitors , Dioxolanes/isolation & purification , Epithelial Cells/drug effects , Epithelial Cells/metabolism , Epithelial Cells/pathology , G2 Phase Cell Cycle Checkpoints/genetics , Glutathione/pharmacology , Humans , MAP Kinase Signaling System/drug effects , Poly(ADP-ribose) Polymerases/genetics , Poly(ADP-ribose) Polymerases/metabolism , Proto-Oncogene Proteins c-bcl-2/antagonists & inhibitors , Proto-Oncogene Proteins c-bcl-2/genetics , Proto-Oncogene Proteins c-bcl-2/metabolism , Reactive Oxygen Species/metabolism , Tumor Suppressor Protein p53/genetics , Tumor Suppressor Protein p53/metabolism , bcl-2-Associated X Protein/agonists , bcl-2-Associated X Protein/genetics , bcl-2-Associated X Protein/metabolism
5.
Z Naturforsch C J Biosci ; 72(11-12): 441-447, 2017 10 26.
Article in English | MEDLINE | ID: mdl-28902633

ABSTRACT

Ginkgetin is known to be an anticancer agent in many studies. However, its effectiveness in treating chronic myeloid leukemia [corrected] remains unknown. The present study aimed to evaluate the effects of ginkgetin on the growth of the K562 cell line. The MTT assay was employed to examine the proliferation of K562, and a terminal deoxynucleotidyl transferase-mediated deoxyuridine triphosphate nick end labeling (TUNEL) staining was conducted to detect the apoptotic rates. Furthermore, changes of tumor necrosis factor-α (TNF-α) were detected by Western blot analysis. Ginkgetin inhibited the proliferation of K562 cells in a dose- and time-dependent manner. Concentrations of ginkgetin required to induce 50% death of K562 at 24, 48 and 72 h were 38.9, 31.3 and 19.2 µM, respectively. Moreover, treatment of ginkgetin increased K562 apoptosis in vitro along with increased levels of TNF-α. Interestingly, anti-TNF-α antibody prevented ginkgetin-induced K562 cell apoptosis and growth inhibition via deactivation of caspase-8, caspase-9 and caspase-3. Concomitantly, downregulation of TNF-α by etanercept in vivo attenuated ginkgetin-induced inhibitory effects on the tumor growth in an xenograft mouse model. Our results indicate that ginkgetin effectively inhibits K562 cell proliferation, and TNF-α plays a key role in ginkgetin-induced cell apoptosis.


Subject(s)
Antineoplastic Agents, Phytogenic/pharmacology , Biflavonoids/pharmacology , Cell Proliferation/drug effects , Gene Expression Regulation, Leukemic , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/drug therapy , Tumor Necrosis Factor-alpha/genetics , Animals , Antibodies, Neutralizing/pharmacology , Antineoplastic Agents, Phytogenic/antagonists & inhibitors , Apoptosis/drug effects , Biflavonoids/antagonists & inhibitors , Caspase 3/genetics , Caspase 3/metabolism , Caspase 8/genetics , Caspase 8/metabolism , Caspase 9/genetics , Caspase 9/metabolism , Etanercept/pharmacology , Humans , Immunosuppressive Agents/pharmacology , Inhibitory Concentration 50 , K562 Cells , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/genetics , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/metabolism , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/pathology , Male , Mice , Mice, Inbred BALB C , Mice, Nude , Signal Transduction , Tumor Necrosis Factor-alpha/antagonists & inhibitors , Tumor Necrosis Factor-alpha/metabolism , Xenograft Model Antitumor Assays
6.
Biochem Biophys Res Commun ; 491(2): 530-536, 2017 09 16.
Article in English | MEDLINE | ID: mdl-28389245

ABSTRACT

Thymol is a phenolic compound with various pharmacological activities such as anti-inflammatory, anti-bacterial and anti-tumor effects. However, the effect of thymol on bladder cancer cell growth is still elusive. The purpose of this study is to investigate the efficacy of thymol in bladder cancer cells and its underlying mechanism. Thymol inhibited bladder cancer cell proliferation in a dose and time-dependent manner. We also observed cell cycle arrest at the G2/M phase after the treatment of thymol. Moreover, thymol could induce apoptosis in bladder cancer cells via the intrinsic pathway along with caspase-3/9 activation, release of cytochrome c and down-regulation of anti-apoptotic Bcl-2 family proteins. The activation of JNK and p38 was also critical for thymol-induced apoptosis since it was abrogated by the treatment of JNK inhibitor (SP600125), and p38 inhibitor (SB203580) but not ERK inhibitor (SCH772984). Furthermore, the generation of ROS (reactive oxygen species) was detected after the treatment of thymol. ROS scavenger NAC (N-acetyl cysteine) could block the thymol-triggered apoptosis and activation of MAPKs. These findings offer a novel therapeutic approach for bladder cancer.


Subject(s)
Antineoplastic Agents, Phytogenic/pharmacology , Apoptosis/drug effects , Epithelial Cells/drug effects , G2 Phase Cell Cycle Checkpoints/drug effects , Gene Expression Regulation, Neoplastic , Thymol/pharmacology , Acetylcysteine/pharmacology , Anthracenes/pharmacology , Antineoplastic Agents, Phytogenic/antagonists & inhibitors , Caspase 3/genetics , Caspase 3/metabolism , Caspase 9/genetics , Caspase 9/metabolism , Cell Line, Transformed , Cell Line, Tumor , Cell Proliferation/drug effects , Cytochromes c/metabolism , Epithelial Cells/metabolism , Epithelial Cells/pathology , Free Radical Scavengers/pharmacology , Humans , Imidazoles/pharmacology , MAP Kinase Kinase 4/genetics , MAP Kinase Kinase 4/metabolism , Protein Kinase Inhibitors/pharmacology , Proto-Oncogene Proteins c-bcl-2/genetics , Proto-Oncogene Proteins c-bcl-2/metabolism , Pyridines/pharmacology , Reactive Oxygen Species/agonists , Reactive Oxygen Species/antagonists & inhibitors , Reactive Oxygen Species/metabolism , Thymol/antagonists & inhibitors , Urothelium/drug effects , Urothelium/metabolism , Urothelium/pathology , p38 Mitogen-Activated Protein Kinases/genetics , p38 Mitogen-Activated Protein Kinases/metabolism
7.
Tumour Biol ; 37(8): 10653-64, 2016 Aug.
Article in English | MEDLINE | ID: mdl-26867767

ABSTRACT

Colchicine is a well-known and potent microtubule targeting agent, but the therapeutic value of colchicine against cancer is limited by its toxicity against normal cells. But, there is no report of its cytotoxic potential against lung cancer cell, at clinically permissible or lower concentrations, minimally toxic to non-cancerous cells. Hence, in the present study, we investigated the possible mechanism by which the efficacy of colchicine against lung cancer cells at less toxic dose could be enhanced. Colchicine at clinically admissible concentration of 2.5 nM had no cytotoxic effect and caused no G2/M arrest in A549 cells. However, at this concentration, colchicine strongly hindered the reformation of cold depolymerised interphase and spindle microtubule. Colchicine induced senescence and reactive oxygen species mediated autophagy in A549 cells at this concentration. Autophagy inhibitor 3-methyladenine (3-MA) sensitised the cytotoxicity of colchicine in A549 cells by switching senescence to apoptotic death, and this combination had reduced cytotoxicity to normal lung fibroblast cells (WI38). Together, these findings indicated the possible use of colchicine at clinically relevant dose along with autophagy inhibitor in cancer therapy.


Subject(s)
Antineoplastic Agents, Phytogenic/pharmacology , Autophagy/drug effects , Cellular Senescence/drug effects , Colchicine/pharmacology , Lung Neoplasms/pathology , A549 Cells , Adenine/analogs & derivatives , Adenine/pharmacology , Antineoplastic Agents, Phytogenic/administration & dosage , Antineoplastic Agents, Phytogenic/antagonists & inhibitors , Apoptosis/drug effects , Cell Cycle/drug effects , Cell Line , Colchicine/administration & dosage , Colchicine/antagonists & inhibitors , Dose-Response Relationship, Drug , Fibroblasts , Humans , Membrane Potential, Mitochondrial/drug effects , Microtubules/drug effects , Reactive Oxygen Species/metabolism , Spindle Apparatus/drug effects , Tubulin Modulators/administration & dosage , Tubulin Modulators/antagonists & inhibitors , Tubulin Modulators/pharmacology
8.
Pharmazie ; 71(12): 727-732, 2016 12 01.
Article in English | MEDLINE | ID: mdl-29442003

ABSTRACT

Methylsulfinylmethane (dimethyl sulfoxide; DMSO) is widely used in clinical treatment and bioresearch. Moreover, there is bioconversion between methylsulfanylmethane (dimethyl sulfide; DMS), DMSO, and methylsulfonylmethane (DMSO2) in mammalian metabolism. Due to the real-time detection limits for volatile compounds, most research has focused on DMSO2 as a stable byproduct of DMSO. Therefore, details about the production of DMS as a byproduct of DMSO metabolism remain to be elucidated. Here, we report the characterization of trace-level volatile organic compounds (VOCs) produced following DMSO treatment of cultured human cells using an ultrasensitive vacuum ultraviolet photoionization mass spectrometer (VUV-PIMS). Using this approach, 24 h after DMSO treatment we detected 16.9 and 21 parts per billion by volume (ppbv) DMS in the atmosphere above the cells (headspace) within HeLa and 293T tissue culture flasks, respectively. When simultaneously exposed to 50 nM paclitaxel (PTX), 17.6 and 22.3 ppbv DMS were detected in the headspace of HeLa and 293T culture flasks, respectively. Nevertheless, at doses of PTX more or less than 50 nM, the detectable levels of DMS were reduced to as low as 8.4 ppbv. Our experimental results demonstrate that by co-administering 5 to 10 nM PTX with DMSO, it is possible to moderate the production of DMS considerably. However, at higher doses of PTX, increased apoptosis was observed that likely contributed to higher DMS production by cells.


Subject(s)
Cell Survival/drug effects , Dimethyl Sulfoxide/pharmacology , Protective Agents/pharmacology , Volatile Organic Compounds/metabolism , Antineoplastic Agents, Phytogenic/antagonists & inhibitors , Antineoplastic Agents, Phytogenic/toxicity , Cells, Cultured , HEK293 Cells , HeLa Cells , Humans , Paclitaxel/antagonists & inhibitors , Paclitaxel/toxicity , Sulfones
9.
J Neurosci ; 35(39): 13487-500, 2015 Sep 30.
Article in English | MEDLINE | ID: mdl-26424893

ABSTRACT

Peripheral neuropathy is dose limiting in paclitaxel cancer chemotherapy and can result in both acute pain during treatment and chronic persistent pain in cancer survivors. The hypothesis tested was that paclitaxel produces these adverse effects at least in part by sensitizing transient receptor potential vanilloid subtype 1 (TRPV1) through Toll-like receptor 4 (TLR4) signaling. The data show that paclitaxel-induced behavioral hypersensitivity is prevented and reversed by spinal administration of a TRPV1 antagonist. The number of TRPV1(+) neurons is increased in the dorsal root ganglia (DRG) in paclitaxel-treated rats and is colocalized with TLR4 in rat and human DRG neurons. Cotreatment of rats with lipopolysaccharide from the photosynthetic bacterium Rhodobacter sphaeroides (LPS-RS), a TLR4 inhibitor, prevents the increase in numbers of TRPV1(+) neurons by paclitaxel treatment. Perfusion of paclitaxel or the archetypal TLR4 agonist LPS activated both rat DRG and spinal neurons directly and produced acute sensitization of TRPV1 in both groups of cells via a TLR4-mediated mechanism. Paclitaxel and LPS sensitize TRPV1 in HEK293 cells stably expressing human TLR4 and transiently expressing human TRPV1. These physiological effects also are prevented by LPS-RS. Finally, paclitaxel activates and sensitizes TRPV1 responses directly in dissociated human DRG neurons. In summary, TLR4 was activated by paclitaxel and led to sensitization of TRPV1. This mechanism could contribute to paclitaxel-induced acute pain and chronic painful neuropathy. Significance statement: In this original work, it is shown for the first time that paclitaxel activates peripheral sensory and spinal neurons directly and sensitizes these cells to transient receptor potential vanilloid subtype 1 (TRPV1)-mediated capsaicin responses via Toll-like receptor 4 (TLR4) in multiple species. A direct functional interaction between TLR4 and TRPV1 is shown in rat and human dorsal root ganglion neurons, TLR4/TRPV1-coexpressing HEK293 cells, and in both rat and mouse spinal cord slices. Moreover, this is the first study to show that this interaction plays an important role in the generation of behavioral hypersensitivity in paclitaxel-related neuropathy. The key translational implications are that TLR4 and TRPV1 antagonists may be useful in the prevention and treatment of chemotherapy-induced peripheral neuropathy in humans.


Subject(s)
Antineoplastic Agents, Phytogenic/pharmacology , Paclitaxel/pharmacology , Sensory Receptor Cells/drug effects , TRPV Cation Channels/antagonists & inhibitors , Toll-Like Receptor 4/drug effects , Animals , Antineoplastic Agents, Phytogenic/antagonists & inhibitors , Calcium/metabolism , Excitatory Postsynaptic Potentials/drug effects , Ganglia, Spinal/cytology , Ganglia, Spinal/drug effects , HEK293 Cells , Humans , Hyperalgesia/chemically induced , Hyperalgesia/physiopathology , Male , Mice , Mice, Inbred C57BL , Paclitaxel/antagonists & inhibitors , Pain Measurement/drug effects , Patch-Clamp Techniques , Rats , Rats, Sprague-Dawley , Signal Transduction/drug effects , Spinal Cord/drug effects , Toll-Like Receptor 4/antagonists & inhibitors
10.
PLoS One ; 10(10): e0140935, 2015.
Article in English | MEDLINE | ID: mdl-26496199

ABSTRACT

High levels of intracellular reactive oxygen species (ROS) in cells is recognized as one of the major causes of cancer cell apoptosis and has been developed into a promising therapeutic strategy for cancer therapy. However, whether apoptosis associated biophysical properties of cancer cells are related to intracellular ROS functions is still unclear. Here, for the first time, we determined the changes of biophysical properties associated with the ROS-mediated oesophageal cancer KYSE-150 cell apoptosis using high resolution atomic force microscopy (AFM). Oridonin was proved to induce ROS-mediated KYSE-150 cell apoptosis in a dose dependent manner, which could be reversed by N-acetylcysteine (NAC) pretreatment. Based on AFM imaging, the morphological damage and ultrastructural changes of KYSE-150 cells were found to be closely associated with ROS-mediated oridonin-induced KYSE-150 cell apoptosis. The changes of cell stiffness determined by AFM force measurement also demonstrated ROS-dependent changes in oridonin induced KYSE-150 cell apoptosis. Our findings not only provided new insights into the anticancer effects of oridonin, but also highlighted the use of AFM as a qualitative and quantitative nanotool to detect ROS-mediated cancer cell apoptosis based on cell biophysical properties, providing novel information of the roles of ROS in cancer cell apoptosis at nanoscale.


Subject(s)
Antineoplastic Agents, Phytogenic/pharmacology , Apoptosis/drug effects , Diterpenes, Kaurane/pharmacology , Epithelial Cells/drug effects , Reactive Oxygen Species/metabolism , Acetylcysteine/pharmacology , Antineoplastic Agents, Phytogenic/antagonists & inhibitors , Cell Line, Tumor , Diterpenes, Kaurane/antagonists & inhibitors , Dose-Response Relationship, Drug , Epithelial Cells/metabolism , Epithelial Cells/ultrastructure , Esophagus/drug effects , Esophagus/metabolism , Esophagus/ultrastructure , Free Radical Scavengers/pharmacology , Humans , Membrane Potential, Mitochondrial/drug effects , Microscopy, Atomic Force , Mitochondria/drug effects , Mitochondria/metabolism , Mitochondria/pathology , Oxidative Stress , Reactive Oxygen Species/agonists , Reactive Oxygen Species/antagonists & inhibitors
11.
Food Funct ; 6(6): 1847-54, 2015 Jun.
Article in English | MEDLINE | ID: mdl-25940566

ABSTRACT

Mango fruit contain many bioactive compounds, some of which are transcription factor regulators. Estrogen receptor alpha (ERα) and beta (ERß) are two regulators of gene transcription that are important in a variety of physiological processes and also in diseases including breast cancer. We examined the ability of the mango constituents quercetin, mangiferin, and the aglycone form of mangiferin, norathyriol, to activate both isoforms of the estrogen receptor. Quercetin and norathyriol decreased the viability of MCF-7 breast cancer cells whereas mangiferin had no effect on MCF-7 cells. We also determined that quercetin and mangiferin selectively activated ERα whereas norathyriol activated both ERα and ERß. Despite quercetin, mangiferin and norathyriol having similar polyphenolic structural motifs, only norathyriol activated ERß, showing that bioactive agents in mangoes have very specific biological effects. Such specificity may be important given the often-opposing roles of ERα and ERß in breast cancer proliferation and other cellular processes.


Subject(s)
Antineoplastic Agents, Phytogenic/pharmacology , Breast Neoplasms/drug therapy , Estrogen Receptor alpha/agonists , Phytoestrogens/pharmacology , Quercetin/pharmacology , Xanthenes/pharmacology , Xanthones/pharmacology , Animals , Antineoplastic Agents, Phytogenic/antagonists & inhibitors , Antineoplastic Agents, Phytogenic/metabolism , Breast Neoplasms/metabolism , COS Cells , Cell Survival/drug effects , Chlorocebus aethiops , Estrogen Receptor Antagonists/pharmacology , Estrogen Receptor alpha/antagonists & inhibitors , Estrogen Receptor alpha/genetics , Estrogen Receptor alpha/metabolism , Estrogen Receptor beta/agonists , Estrogen Receptor beta/antagonists & inhibitors , Estrogen Receptor beta/genetics , Estrogen Receptor beta/metabolism , Female , Fruit/chemistry , Genes, Reporter/drug effects , Humans , MCF-7 Cells , Mangifera/chemistry , Neoplasm Proteins/agonists , Neoplasm Proteins/antagonists & inhibitors , Neoplasm Proteins/genetics , Neoplasm Proteins/metabolism , Phytoestrogens/antagonists & inhibitors , Phytoestrogens/metabolism , Quercetin/antagonists & inhibitors , Recombinant Proteins/chemistry , Recombinant Proteins/metabolism , Response Elements/drug effects , Transcriptional Activation/drug effects , Xanthenes/antagonists & inhibitors , Xanthenes/metabolism , Xanthones/antagonists & inhibitors , Xanthones/metabolism
12.
Pharm Biol ; 53(6): 838-48, 2015 Jun.
Article in English | MEDLINE | ID: mdl-25429779

ABSTRACT

CONTEXT: Curcumin exhibits a wide spectrum of biological activities which include neuroprotective, antinociceptive, anti-inflammatory, and antioxidant activity. OBJECTIVE: The present study evaluates the effect of curcumin in vincristine-induced neuropathy in a mice model. MATERIALS AND METHODS: Vincristine sulfate (0.1 mg/kg, i.p. for 10 consecutive days) was administered to mice to induce neuropathy. Pain behavior was assessed at different days, i.e., 0, 7, 10, and 14 d. Sciatic nerve total calcium, superoxide dismutase (SOD), catalase (CAT), glutathione peroxidase (GPx), reduced glutathione (GSH), nitric oxide (NO), and lipid peroxidation (LPO) were also estimated after the 14th day of study. Pregabalin (10 mg/kg, p.o.) and curcumin (15, 30, and 60 mg/kg, p.o.) were administered for 14 consecutive days. RESULTS: Curcumin at 60 mg/kg significantly attenuated the vincristine-induced neuropathic pain manifestations in terms of thermal hyperalgesia (p < 0.001) and allodynia (p < 0.001); mechanical hyperalgesia (p < 0.001); functional loss (p < 0.001); and in the delayed phase of formalin test (p < 0.001). Curcumin at 30 and 60 mg/kg exhibited significant changes (p < 0.001) in antioxidant levels and in total calcium levels in vincristine-injected mice. CONCLUSION: Curcumin at 30 and 60 mg/kg dose levels significantly attenuated vincristine-induced neuropathy which may be due to its multiple actions including antinociceptive, calcium inhibitory, and antioxidant effect.


Subject(s)
Antineoplastic Agents, Phytogenic/antagonists & inhibitors , Antineoplastic Agents, Phytogenic/toxicity , Curcumin/pharmacology , Neuroprotective Agents/pharmacology , Peripheral Nervous System Diseases/drug therapy , Vincristine/antagonists & inhibitors , Vincristine/toxicity , Animals , Antioxidants/pharmacology , Behavior, Animal/drug effects , Calcium/metabolism , Calcium Channels, N-Type/drug effects , Male , Mice , Neuralgia/chemically induced , Neuralgia/drug therapy , Pain Measurement/drug effects , Peripheral Nervous System Diseases/chemically induced , Postural Balance/drug effects , Sciatica/drug therapy
13.
Toxicol Appl Pharmacol ; 281(3): 285-93, 2014 Dec 15.
Article in English | MEDLINE | ID: mdl-25448047

ABSTRACT

Beta-lapachone (beta-Lp) derived from the Lapacho tree is a potentially novel anticancer agent currently under clinical trials. Previous studies suggested that redox activation of beta-Lp catalyzed by NAD(P)H: quinone oxidoreductase 1 (NQO1) accounted for its killing of cancer cells. However, the exact mechanisms of this effect remain largely unknown. Using chemiluminescence and electron paramagnetic resonance (EPR) spin-trapping techniques, this study for the first time demonstrated the real-time formation of ROS in the redox activation of beta-lapachone from cancer cells mediated by mitochondria and NQO1 in melanoma B16-F10 and hepatocellular carcinoma HepG2 cancer cells. ES936, a highly selective NQO1 inhibitor, and rotenone, a selective inhibitor of mitochondrial electron transport chain (METC) complex I were found to significantly block beta-Lp meditated redox activation in B16-F10 cells. In HepG2 cells ES936 inhibited beta-Lp-mediated oxygen radical formation by ~80% while rotenone exerted no significant effect. These results revealed the differential contribution of METC and NQO1 to beta-lapachone-induced ROS formation and cancer cell killing. In melanoma B16-F10 cells that do not express high NQO1 activity, both NOQ1 and METC play a critical role in beta-Lp redox activation. In contrast, in hepatocellular carcinoma HepG2 cells expressing extremely high NQO1 activity, redox activation of beta-Lp is primarily mediated by NQO1 (METC plays a minor role). These findings will contribute to our understanding of how cancer cells are selectively killed by beta-lapachone and increase our ability to devise strategies to enhance the anticancer efficacy of this potentially novel drug while minimizing its possible adverse effects on normal cells.


Subject(s)
Antineoplastic Agents, Phytogenic/metabolism , Electron Transport Complex I/metabolism , NAD(P)H Dehydrogenase (Quinone)/metabolism , Naphthoquinones/metabolism , Neoplasm Proteins/metabolism , Neoplasms/enzymology , Prodrugs/metabolism , Activation, Metabolic/drug effects , Animals , Antineoplastic Agents, Phytogenic/antagonists & inhibitors , Antineoplastic Agents, Phytogenic/pharmacology , Cell Line, Tumor , Electron Transport Complex I/antagonists & inhibitors , Enzyme Inhibitors/pharmacology , Humans , Indolequinones/pharmacology , Mice , Mitochondria/drug effects , Mitochondria/enzymology , Mitochondria/metabolism , NAD(P)H Dehydrogenase (Quinone)/antagonists & inhibitors , Naphthoquinones/antagonists & inhibitors , Naphthoquinones/pharmacology , Neoplasm Proteins/antagonists & inhibitors , Neoplasms/drug therapy , Neoplasms/metabolism , Oxidation-Reduction/drug effects , Prodrugs/chemistry , Prodrugs/pharmacology , Reactive Oxygen Species/metabolism , Reverse Transcriptase Inhibitors/chemistry , Reverse Transcriptase Inhibitors/metabolism , Reverse Transcriptase Inhibitors/pharmacology , Rotenone/pharmacology
14.
Sci Rep ; 4: 7041, 2014 Nov 13.
Article in English | MEDLINE | ID: mdl-25391369

ABSTRACT

Reactive oxygen species (ROS) and cellular oxidant stress are considered inducers of carcinogenesis. However, the association of ROS with cancer is both complex and, at times, paradoxical. We assessed the effects of dihydromyricetin (DHM) on the induction of ROS accumulation and on the activation of the mitochondrial signaling pathway in human hepatoma HepG2 cells. The results indicated that DHM could reduce ROS accumulation in a concentration-dependent manner. Additionally, with increasing concentrations of DHM, the expression of proteins that participate in the cell apoptosis program increased in a concentration-dependent manner. Furthermore, we found that a low dose of H2O2 (10 nM) could reverse DHM-induced cell apoptosis. We observed the following critical issues: first, the cellular redox balance is vital in DHM-induced apoptosis of human hepatocellular carcinoma (HCC) cells, and second, ROS could function as a redox-active signaling messenger to determine DHM-induced cell apoptosis. In this study, we demonstrated that low levels of ROS are also critical for the function of HCC cells.


Subject(s)
Antineoplastic Agents, Phytogenic/pharmacology , Apoptosis/drug effects , Flavonols/pharmacology , Mitochondria/drug effects , Reactive Oxygen Species/metabolism , Antineoplastic Agents, Phytogenic/antagonists & inhibitors , Cell Line, Tumor , Cell Proliferation , Dose-Response Relationship, Drug , Flavonols/antagonists & inhibitors , Hep G2 Cells , Humans , Hydrogen Peroxide/pharmacology , Mitochondria/metabolism , Signal Transduction
15.
Oncotarget ; 5(20): 9650-63, 2014 Oct 30.
Article in English | MEDLINE | ID: mdl-25333260

ABSTRACT

In this study, we compared the blood serum levels of circulating cell-free and exosomal microRNAs, and their involvement in the molecular subtypes of breast cancer patients. Our analyses on cell-free miR-101, miR-372 and miR-373 were performed in preoperative blood serum of 168 patients with invasive breast cancer, 19 patients with benign breast diseases and 28 healthy women. MicroRNAs were additionally quantified in exosomes of 50 cancer patients and 12 healthy women from the same cohort. Relative concentrations were measured by quantitative TaqMan MicroRNA assays and correlated to clinicopathological risk factors. The concentrations of cell-free miR-101 (p=0.013) and miR-373 (p=0.024) were significantly different between patients with breast cancer and benign tumors. A prevalence of miR-101, miR-372 and miR-373 were found in exosomes. The levels of circulating exosomal (but not cell-free) miR-373 were higher in triple negative than luminal carcinomas (p=0.027). Also, estrogen-negative (p=0.021) and progesterone-negative (p=0.01) tumors displayed higher concentrations of exosomal miR-373 than patients with hormone-receptor positive tumors. Overexpression of miR-373 by transfection of MCF-7 cells showed downregulated protein expression of the estrogen receptor, and inhibition of apoptosis induced by camptothecin. Our data indicate that serum levels of exosomal miR-373 are linked to triple negative and more aggressive breast carcinomas.


Subject(s)
Exosomes/metabolism , MicroRNAs/blood , Triple Negative Breast Neoplasms/blood , Adult , Aged , Aged, 80 and over , Antineoplastic Agents, Phytogenic/antagonists & inhibitors , Antineoplastic Agents, Phytogenic/pharmacology , Apoptosis/drug effects , Apoptosis/genetics , Biomarkers, Tumor/blood , Biomarkers, Tumor/genetics , Breast Diseases/blood , Breast Diseases/genetics , Camptothecin/antagonists & inhibitors , Camptothecin/pharmacology , Case-Control Studies , Cell Line, Tumor , Cell Movement , Cohort Studies , Exosomes/genetics , Female , Gene Expression Profiling , Gene Expression Regulation, Neoplastic , Humans , MCF-7 Cells , Middle Aged , Receptors, Estrogen/biosynthesis , Receptors, Estrogen/genetics , Triple Negative Breast Neoplasms/drug therapy , Triple Negative Breast Neoplasms/genetics , Triple Negative Breast Neoplasms/pathology
16.
PLoS One ; 9(10): e109630, 2014.
Article in English | MEDLINE | ID: mdl-25299698

ABSTRACT

BACKGROUND: In this study we investigated the in vitro and in vivo anticancer effect of carnosol, a naturally occurring polyphenol, in triple negative breast cancer. RESULTS: We found that carnosol significantly inhibited the viability and colony growth induced G2 arrest in the triple negative MDA-MB-231. Blockade of the cell cycle was associated with increased p21/WAF1 expression and downregulation of p27. Interestingly, carnosol was found to induce beclin1-independent autophagy and apoptosis in MDA-MB-231 cells. The coexistence of both events, autophagy and apoptosis, was confirmed by electron micrography. Induction of autophagy was found to be an early event, detected within 3 h post-treatment, which subsequently led to apoptosis. Carnosol treatment also caused a dose-dependent increase in the levels of phosphorylated extracellular signal-regulated kinase 1 and 2 (pERK1/2). Moreover, we show that carnosol induced DNA damage, reduced the mitochondrial potential and triggered the activation of the intrinsic and extrinsic apoptotic pathway. Furthermore, we found that carnosol induced a dose-dependent generation of reactive oxygen species (ROS) and inhibition of ROS by tiron, a ROS scavenger, blocked the induction of autophagy and apoptosis and attenuated DNA damage. To our knowledge, this is the first report to identify the induction of autophagy by carnosol. CONCLUSION: In conclusion our findings provide strong evidence that carnosol may be an alternative therapeutic candidate against the aggressive form of breast cancer and hence deserves more exploration.


Subject(s)
Abietanes/pharmacology , Antineoplastic Agents, Phytogenic/pharmacology , Apoptosis Regulatory Proteins/genetics , Membrane Proteins/genetics , Reactive Oxygen Species/metabolism , 1,2-Dihydroxybenzene-3,5-Disulfonic Acid Disodium Salt/pharmacology , Abietanes/antagonists & inhibitors , Antineoplastic Agents, Phytogenic/antagonists & inhibitors , Apoptosis/drug effects , Apoptosis Regulatory Proteins/metabolism , Autophagy/drug effects , Beclin-1 , Cell Line, Tumor , Cell Proliferation/drug effects , Cyclin-Dependent Kinase Inhibitor p21/genetics , Cyclin-Dependent Kinase Inhibitor p21/metabolism , Cyclin-Dependent Kinase Inhibitor p27/genetics , Cyclin-Dependent Kinase Inhibitor p27/metabolism , DNA Damage , Female , Free Radical Scavengers/pharmacology , G2 Phase Cell Cycle Checkpoints/drug effects , Humans , Membrane Potential, Mitochondrial/drug effects , Membrane Proteins/metabolism , Mitogen-Activated Protein Kinase 1/genetics , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3/genetics , Mitogen-Activated Protein Kinase 3/metabolism , Phosphorylation/drug effects , Triple Negative Breast Neoplasms/genetics , Triple Negative Breast Neoplasms/metabolism , Triple Negative Breast Neoplasms/pathology
17.
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
18.
Br J Pharmacol ; 171(3): 636-45, 2014 Feb.
Article in English | MEDLINE | ID: mdl-24117398

ABSTRACT

BACKGROUND AND PURPOSE: Paclitaxel (PAC) is associated with chemotherapy-induced neuropathic pain (CIPN) that can lead to the cessation of treatment in cancer patients even in the absence of alternate therapies. We previously reported that chronic administration of the non-psychoactive cannabinoid cannabidiol (CBD) prevents PAC-induced mechanical and thermal sensitivity in mice. Hence, we sought to determine receptor mechanisms by which CBD inhibits CIPN and whether CBD negatively effects nervous system function or chemotherapy efficacy. EXPERIMENTAL APPROACH: The ability of acute CBD pretreatment to prevent PAC-induced mechanical sensitivity was assessed, as was the effect of CBD on place conditioning and on an operant-conditioned learning and memory task. The potential interaction of CBD and PAC on breast cancer cell viability was determined using the MTT assay. KEY RESULTS: PAC-induced mechanical sensitivity was prevented by administration of CBD (2.5 - 10 mg·kg⁻¹) in female C57Bl/6 mice. This effect was reversed by co-administration of the 5-HT(1A) antagonist WAY 100635, but not the CB1 antagonist SR141716 or the CB2 antagonist SR144528. CBD produced no conditioned rewarding effects and did not affect conditioned learning and memory. Also, CBD + PAC combinations produce additive to synergistic inhibition of breast cancer cell viability. CONCLUSIONS AND IMPLICATIONS: Our data suggest that CBD is protective against PAC-induced neurotoxicity mediated in part by the 5-HT(1A) receptor system. Furthermore, CBD treatment was devoid of conditioned rewarding effects or cognitive impairment and did not attenuate PAC-induced inhibition of breast cancer cell viability. Hence, adjunct treatment with CBD during PAC chemotherapy may be safe and effective in the prevention or attenuation of CIPN.


Subject(s)
Cannabidiol/therapeutic use , Neuralgia/prevention & control , Neurons/drug effects , Neuroprotective Agents/therapeutic use , Paclitaxel/antagonists & inhibitors , Receptor, Serotonin, 5-HT1A/metabolism , Serotonin 5-HT1 Receptor Agonists/therapeutic use , Animals , Antineoplastic Agents, Phytogenic/adverse effects , Antineoplastic Agents, Phytogenic/agonists , Antineoplastic Agents, Phytogenic/antagonists & inhibitors , Antineoplastic Agents, Phytogenic/pharmacology , Behavior, Animal/drug effects , Brain/drug effects , Brain/metabolism , Breast Neoplasms/drug therapy , Cannabidiol/adverse effects , Cannabidiol/antagonists & inhibitors , Cell Line, Tumor , Cell Survival/drug effects , Conditioning, Operant/drug effects , Drug Synergism , Female , Humans , Memory/drug effects , Mice , Mice, Inbred C57BL , Neuralgia/chemically induced , Neuralgia/metabolism , Neuralgia/physiopathology , Neurons/metabolism , Neuroprotective Agents/adverse effects , Neuroprotective Agents/antagonists & inhibitors , Paclitaxel/adverse effects , Paclitaxel/agonists , Paclitaxel/pharmacology , Receptor, Serotonin, 5-HT1A/chemistry , Serotonin 5-HT1 Receptor Agonists/adverse effects , Serotonin 5-HT1 Receptor Agonists/chemistry , Serotonin 5-HT1 Receptor Antagonists/pharmacology
19.
Toxicol In Vitro ; 28(2): 248-57, 2014 Mar.
Article in English | MEDLINE | ID: mdl-24239945

ABSTRACT

Cisplatin, a cancer chemotherapy drug, is nephrotoxic. The aim of this study was to investigate whether resveratrol (RES) reduced cisplatin cytotoxicity and oxidative stress. Rat renal cortical slices were pre-incubated 30min with 0 (VEH, ethanol) or 30µg/ml RES followed by 60, 90 or 120min co-incubation with 0, 75, or 150µg/ml cisplatin. Lactate dehydrogenase (LDH) leakage was unchanged at 60 and 90min by cisplatin. Cisplatin increased (p<0.05) LDH leakage at 120min which was protected by RES. Cisplatin induced oxidative stress prior to LDH leakage as cisplatin depressed glutathione peroxidase and superoxide dismutase (SOD) activity, increased lipid peroxidation, protein carbonyls and 4-hydroxynonenal (4-HNE) adducted proteins within 60min. RES failed to reverse glutathione (GSH) depression by cisplatin. In order to eliminated an extracellular interaction between RES and cisplatin, additional studies (RINSE studies) allowed a 30min RES uptake into slices, transfer of slices to buffer lacking RES, followed by 120min cisplatin incubation. RES in the RINSE studies prevented LDH leakage by cisplatin indicating that RES protection was not via a physical interaction with cisplatin in the media. These findings indicate that RES diminished cisplatin in vitro renal toxicity and prevented the development of oxidative stress.


Subject(s)
Antineoplastic Agents, Phytogenic/antagonists & inhibitors , Antineoplastic Agents, Phytogenic/toxicity , Antineoplastic Agents/toxicity , Antioxidants/pharmacology , Cisplatin/antagonists & inhibitors , Cisplatin/toxicity , Kidney Diseases/pathology , Oxidative Stress/drug effects , Stilbenes/pharmacology , Aldehydes/metabolism , Animals , Cell Survival/drug effects , Glutathione/metabolism , Glutathione Peroxidase/metabolism , Kidney/pathology , Kidney Diseases/chemically induced , L-Lactate Dehydrogenase/metabolism , Lipid Peroxidation/drug effects , Male , Oxidation-Reduction , Protein Carbonylation/drug effects , Rats , Rats, Inbred F344 , Resveratrol , Superoxide Dismutase/metabolism , Tumor Necrosis Factor-alpha/biosynthesis
20.
Anticancer Drugs ; 24(9): 928-36, 2013 Oct.
Article in English | MEDLINE | ID: mdl-23965728

ABSTRACT

Previous studies have reported that vitexins induce cytotoxic effects. In the present study, we investigate a new native lignan vitexin 6 (VB6) in vitro to determine the molecular mechanism underlying its cytotoxicity. We screened and cultured several tumor cell lines and subsequently analyzed VB6 cytotoxicity against 14 different tumor cell lines using a 3-(4,5-dimethythiazol-2-yl)-2,5-diphenyl tetrazolium bromide assay. The expression of proteins that regulate apoptosis and autophagy was determined using western blot analysis. VB6 showed an excellent cytotoxic effect against various cancer cell lines in vitro. It induced apoptosis and autophagy of cancer cells. VB6-induced apoptosis showed a time-dependent and concentration-dependent relationship with cleaved poly (ADP-ribose) polymerase, cleaved caspase-3, Bax upregulation, and Bcl-2 downregulation. The levels of Beclin-1 and LC3-II, which are markers for cell autophagy, gradually increased after VB6 treatment. Jun N-terminal kinase (JNK) phosphorylation was increased after VB6 treatment, accompanied by upregulation of P-Bcl-2 and P-C-Jun expression. Cotreatment with a JNK inhibitor significantly decreased VB6-induced cell death and downregulated P-Bcl-2, and cleaved PARP and Beclin-1 expression. The new native lignan VB6 inhibits cancer cell proliferation by activating the JNK pathway. We believe that VB6 could be a valuable chemotherapeutic drug after further evaluation.


Subject(s)
Antineoplastic Agents, Phytogenic/pharmacology , Apigenin/pharmacology , Apoptosis/drug effects , Autophagy/drug effects , JNK Mitogen-Activated Protein Kinases/metabolism , Lignans/pharmacology , MAP Kinase Signaling System/drug effects , Antineoplastic Agents, Phytogenic/antagonists & inhibitors , Apigenin/antagonists & inhibitors , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Survival/drug effects , Drugs, Chinese Herbal/chemistry , Ethnopharmacology , Humans , Inhibitory Concentration 50 , JNK Mitogen-Activated Protein Kinases/antagonists & inhibitors , JNK Mitogen-Activated Protein Kinases/chemistry , Kinetics , Lignans/antagonists & inhibitors , Neoplasm Proteins/agonists , Neoplasm Proteins/antagonists & inhibitors , Neoplasm Proteins/metabolism , Neoplasms/drug therapy , Neoplasms/metabolism , Neoplasms/pathology , Protein Kinase Inhibitors/pharmacology , Seeds/chemistry , Vitex/chemistry
SELECTION OF CITATIONS
SEARCH DETAIL
...