Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 15 de 15
Filter
Add more filters











Publication year range
1.
BMC Cancer ; 22(1): 727, 2022 Jul 04.
Article in English | MEDLINE | ID: mdl-35787261

ABSTRACT

BACKGROUND: The PD-L1 antibody is an immune checkpoint inhibitor (ICI) attracting attention. The third-generation anticancer drug has been proven to be very effective due to fewer side effects and higher tumor-specific reactions than conventional anticancer drugs. However, as tumors produce additional resistance in the host immune system, the effectiveness of ICI is gradually weakening. Therefore, it is very important to develop a combination therapy that increases the anticancer effect of ICI by removing anticancer resistance factors present around the tumor. METHODS: The syngeneic model was used (n = 6) to investigate the enhanced anti-tumor effect of PD-L1 antibody with the addition of PLAG. MB49 murine urothelial cancer cells were implanted into the C57BL/6 mice subcutaneously. PLAG at different dosages (50/100 mpk) was daily administered orally for another 4 weeks with or without 5 mpk PD-L1 antibody (10F.9G2). PD-L1 antibody was delivered via IP injection once a week. RESULTS: The aPD-L1 monotherapy group inhibited tumor growth of 56% compared to the positive group, while the PLAG and aPD-L1 co-treatment inhibited by 89%. PLAG treatment effectively reduced neutrophils infiltrating localized in tumor and converted to a tumor microenvironment with anti-tumor effective T-cells. PLAG increased tumor infiltration of CD8 positive cytotoxic T-cell populations while effectively inhibiting the infiltration of neoplastic T-cells such as CD4/FoxP3. Eventually, neutrophil-induced tumor ICI resistance was resolved by restoring the neutrophil-to-lymphocyte ratio to the normal range. In addition, regulation of cytokine and chemokine factors that inhibit neutrophil infiltration and increase the killing activity of cytotoxic T cells was observed in the tumors of mice treated with PLAG + aPD-L1. CONCLUSIONS: PLAG effectively turned the tumor-promoting microenvironment into a tumor-suppressing microenvironment. As a molecule that increases the anti-tumor effectiveness of aPD-L1, PLAG has the potential to be an essential and effective ICI co-therapeutic agent.


Subject(s)
Carcinoma, Transitional Cell , Urinary Bladder Neoplasms , Animals , Mice , B7-H1 Antigen , Carcinoma, Transitional Cell/drug therapy , Mice, Inbred C57BL , Neutrophil Infiltration , Tumor Microenvironment , Urinary Bladder Neoplasms/drug therapy
2.
Biochem Biophys Res Commun ; 619: 110-116, 2022 09 03.
Article in English | MEDLINE | ID: mdl-35753218

ABSTRACT

Chemotherapy induces tumor cell death and inhibits tumor progression, but the accompanying immune responses in the surrounding dying tissue cause significant inflammation. These responses, such as excessive neutrophil infiltration into tumor tissue, are the main causes of resistance to anticancer treatment. The development of drugs that reduce neutrophil infiltration into tumors is necessary to increase the anticancer effect of chemotherapy. Here, we show that the antitumor effect of the chemotherapy AC regimen (Adriamycin and cyclophosphamide) was increased by 1-palmitoyl-2-linoleoyl-3-acetyl-rac-glycerol (PLAG) cotreatment in the MDA-MB-231 triple-negative breast cancer xenograft mouse model. Tumor growth was inhibited up to 56% in mice treated with AC and inhibited up to 94% in mice cotreated with AC and PLAG. Side effects of chemotherapy, such as a reduction in body weight, were alleviated in mice cotreated with AC and PLAG. Excessive neutrophil infiltration caused by the AC regimen was successfully cleared in mice cotreated with AC and PLAG. We conclude that PLAG inhibits excessive neutrophil infiltration that aids tumor growth. Reduced neutrophils and increased lymphocytes in PLAG-treated mice can maximize the antitumor effect of the AC regimen and inhibit tumor growth.


Subject(s)
Doxorubicin , Triple Negative Breast Neoplasms , Animals , Cell Line, Tumor , Cyclophosphamide/therapeutic use , Disease Models, Animal , Doxorubicin/therapeutic use , Heterografts , Humans , Mice , Triple Negative Breast Neoplasms/drug therapy , Xenograft Model Antitumor Assays
3.
Neoplasia ; 31: 100815, 2022 09.
Article in English | MEDLINE | ID: mdl-35728512

ABSTRACT

Extracellular adenosine in the tumor microenvironment plays a vital role in cancer development. Specifically, activation of adenosine receptors affects tumor cell growth and adenosine release. We examined the anti-tumor efficacy of 1-palmitoyl-2-linoleoyl-3-acetyl-rac-glycerol (PLAG) in animal models, revealing the role of PLAG in inhibiting tumor progression by promoting the degradation of adenosine 2B receptors (A2BRs) in tumors. PLAG induced the expression of thioredoxin-interacting protein (TXNIP), a type of α-arrestin that accelerates A2BR internalization by interacting with A2BR complexes containing ß-arrestin. Engulfed receptors bound to TXNIP were rapidly degraded after E3 ligase recruitment and ubiquitination, resulting in early termination of intracellular signals that promote tumor overgrowth. However, in control cancer cells, A2BRs bound to protein phosphatase 2A and were returned to the cell membrane instead of being degraded, resulting in continuous receptor-mediated signaling by pathways including the Raf-Erk axis, which promotes tumor proliferation. A TXNIP-silenced cell-implanted mouse model and TXNIP knockout (KO) mice were used to verify that PLAG-mediated suppression of tumor progression is dependent on TXNIP expression. Increased tumor growth was observed in TXNIP-silenced cell-implanted mice, and the anti-tumor effects of PLAG, including delayed tumor overgrowth, were greatly reduced. However, the anti-tumor effects of PLAG were observed in cancer cell-implanted TXNIP-KO mice, which indicates that PLAG produces anti-tumor effects by enhancing TXNIP expression in tumor cells. These essential functions of PLAG, including delaying tumor growth via A2BR degradation, suggest innovative directions for anticancer drug development.


Subject(s)
Carcinoma, Lewis Lung , Carcinoma, Non-Small-Cell Lung , Carrier Proteins , Lung Neoplasms , Receptors, Purinergic P1 , Thioredoxins , Adenosine/pharmacology , Animals , Carcinoma, Lewis Lung/drug therapy , Carcinoma, Lewis Lung/metabolism , Carcinoma, Non-Small-Cell Lung/drug therapy , Carcinoma, Non-Small-Cell Lung/metabolism , Carrier Proteins/metabolism , Diglycerides/metabolism , Diglycerides/pharmacology , Mice , Receptors, Purinergic P1/drug effects , Receptors, Purinergic P1/metabolism , Thioredoxins/metabolism , Tumor Microenvironment
4.
Transl Oncol ; 20: 101398, 2022 Jun.
Article in English | MEDLINE | ID: mdl-35339890

ABSTRACT

Chemotherapy-induced cachexia has been a significant challenge to the successful treatment of cancer patients. Chemotherapy leads to loss of muscle, loss of appetite, and excessive weight loss, which makes these necessary treatments intolerable for most patients. Therefore, it is necessary to alleviate cachexia to successfully treat cancer patients. In this study, tumor-implanted mouse models administered cisplatin showed rapid weight loss and reduced feeding rate by the second week of treatment, and 1-palmitoyl-2-linoleoyl-3-acetyl-rac-glycerol (PLAG) effectively alleviated cisplatin-induced cachexia. In mice treated with cisplatin on a sacrificial day after 6 weeks, the weight of the two major leg muscles (quadriceps femoris and gastrocnemius) were reduced by up to 70%, but this muscle reduction was successfully prevented in the PLAG co-treatment group. The distribution and size of muscle fibers that appear in small units in cisplatin-treated mice were restored to normal levels by PLAG co-treatment. Furthermore, myostatin expression levels were upregulated by cisplatin, whereas myostatin decreased to normal levels with muscle recovery in the PLAG co-treated group. Tumor necrosis factor alpha (TNF-α) and interleukin-6 (IL-6), which are commonly expressed in cachexia, were significantly increased in cisplatin-treated mice but were reduced to normal levels in PLAG co-treated mice. Glucose absorption, an indicator of muscle tissue activity, decreased with cisplatin treatment and recovered to normal levels with PLAG co-treatment. Overall, PLAG effectively alleviated cisplatin-induced cachexia symptoms and reduced tumor growth in tumor-implanted mice. These findings suggest PLAG may be a promising drug to alleviate cachexia in cancer patients receiving chemotherapy.

5.
Oncol Rep ; 44(1): 241-251, 2020 07.
Article in English | MEDLINE | ID: mdl-32377695

ABSTRACT

Activated epidermal growth factor receptors (EGFRs) are crucial for inducing metastasis in cancer cells by promoting matrix metalloproteinase (MMP) expression. The present study was designed to investigate the effects of 1­palmitoyl­2­linoleoyl­3­acetyl­rac­glycerol (PLAG) on MMP expression in epidermal growth factor (EGF)­stimulated breast cancer cells in vitro. EGF stimulation induced internalization of its cognate receptor, EGFR, for stimulus­desensitization. These internalized receptors, complexed with the ubiquitin ligase c­Cbl and EGFR pathway substrate 15 (EPS15) (for degradation), were evaluated by confocal microscopy at 5­90 min time intervals. During intracellular trafficking of EGFRs, EGF­induced signaling cascades were analyzed by examining EGFR and SHC phosphorylation. Modulation of MMP expression was assessed by evaluating the activity of transcription factor AP­1 using a luciferase assay. PLAG accelerated the assembly of EGFRs with c­Cbl and EPS15 and promoted receptor degradation. This faster intracellular EGFR degradation reduced AP­1­mediated MMP expression. PLAG stimulation upregulated thioredoxin­interacting protein (TXNIP) expression, and this mediated the accelerated receptor internalization. This PLAG­induced increase in EGFR trafficking was blocked in TXNIP­silenced cells. By downregulating MMP expression, PLAG effectively attenuated EGF­induced mobility and invasiveness in these cancer cells. These data suggest that PLAG may be a potential therapeutic agent for blocking metastasis.


Subject(s)
Brain Neoplasms/metabolism , Epidermal Growth Factor/pharmacology , Glycerides/pharmacology , Matrix Metalloproteinase 9/metabolism , Adaptor Proteins, Vesicular Transport/metabolism , Brain Neoplasms/drug therapy , Cell Line, Tumor , ErbB Receptors/metabolism , Gene Expression Regulation, Neoplastic/drug effects , Humans , Proteolysis , Proto-Oncogene Proteins c-cbl/metabolism , Signal Transduction/drug effects
6.
Cancers (Basel) ; 12(3)2020 Feb 28.
Article in English | MEDLINE | ID: mdl-32121107

ABSTRACT

The effectiveness of chemotherapy and radiotherapy to treat lung cancer is limited because of highly metastatic nature. Novel strategies and drugs to attenuate metastatic activity are urgently required. In this study, red fluorescence proteins (RFP)-labeled A549 human lung cancer cells were orthotopically implantation, where they developed primary tumors. Metastasis in brain and intestines were reduced by up to 80% by treatment with 100 mpk 1-palmitoyl-2-linoleoyl-3-acetyl-rac-glycerol (PLAG) compared with that in control mice. PLAG treatment also reduced the migration of the primary tumors. Interestingly, substantial neutrophil infiltration was observed in the tumors in control mice. The neutrophil contribution to A549 cell metastatic activity was examined in in vitro co-culture system. Metastatic activity could be achieved in the A549 cells through epidermal growth factor receptor (EGFR) transactivation mediated by protease activating receptor 2 (PAR2) receptor. Neutrophil elastase secreted from tumor-infiltrating neutrophils stimulated PAR2 and induced EGFR transactivation. However, this transactivation was inhibited by inducing PAR2 degradation following PLAG treatment and metastatic activity was effectively inhibited. PLAG attenuated cancer metastatic activity via modulated PAR2/EGFR transactivation by accelerating PAR2 degradation. These results suggest PLAG as potential therapeutic agent to combat tumor metastasis via regulating the activation signal pathway of PAR2 by tumor infiltrate-neutrophils.

7.
FEBS J ; 286(24): 5016-5029, 2019 12.
Article in English | MEDLINE | ID: mdl-31714686

ABSTRACT

Neutrophil activity, including trapping of damage-associated molecular patterns by neutrophil extracellular traps (NETs), is an important response to microbial infection. Most activated neutrophils commit to apoptosis and are removed by activated macrophages in the process of efferocytosis. Improper clearance of apoptotic neutrophils often causes an unnecessary and exaggerated immune response and subsequent chronic inflammation. Effective macrophage mobility toward activated neutrophils, which is triggered by binding of 'find-me' signals to receptors such as P2Y2, is a crucial step for the timely clearance of apoptotic neutrophils. In this paper, we investigated the effect of 1-palmitoyl-2-linoleoyl-3-acetyl-rac-glycerol (PLAG) on efferocytosis and the underlying molecular mechanisms. In a coculture of apoptotic neutrophils with macrophages, PLAG treatment increased levels of efferocytosis of apoptotic neutrophils. PLAG induced faster translocation of P2Y2 from lipid rafts to nonlipid raft plasma membrane domains in macrophages. This repositioning of P2Y2 enables the polarization of the cytoskeleton by association of the receptor with cytoskeletal proteins such as α-tubulin and actin to improve the mobility of macrophages. The formation of vesicular, chylomicron-like structures by PLAG was a prerequisite for the induction of this macrophage activity, as none of these effects was seen when the vesicle receptor GPIHBP1 was absent. Taken together, these data showed that PLAG is a powerful immune resolvent that triggers the prompt clearance of apoptotic neutrophils by enhanced efferocytosis activity. PLAG could therefore be an effective lipid-based efferocytosis enhancer for use as a therapeutic drug to prevent inflammatory disease caused by uncontrolled immune responses.


Subject(s)
Macrophages/metabolism , Neutrophils/metabolism , Receptors, Purinergic P2Y2/metabolism , Apoptosis/genetics , Apoptosis/physiology , Cell Line , Cytoskeleton/metabolism , HL-60 Cells , Humans , Immunoprecipitation , Macrophages/cytology , Microscopy, Confocal
8.
Mol Med Rep ; 17(2): 2572-2580, 2018 Feb.
Article in English | MEDLINE | ID: mdl-29207130

ABSTRACT

Cnidium monnieri (L.) Cusson is a frequently used traditional Chinese medicine that treats gynecological diseases and carbuncles. However, the mechanism of action of C. monnieri remains to be fully elucidated. The present study examined the cell cycle arrest and apoptotic effects resulting from ethanol extract of C. monnieri (CME) in HepG2 (wild­type p53) and Hep3B (p53­null) hepatocellular carcinoma cells. An MTT assay was used to confirm the anti­proliferative effect of CME. The cells were stained with Hoechst 33342 or propidium iodide. It was demonstrated that proliferation of HepG2 cells was suppressed by CME. Cell cycle arrest occurred in the G1 phase following treatment with CME and the number of apoptotic bodies was increased. The expression levels of cell cycle­associated proteins, including protein kinase B (Akt), glycogen synthase kinase­3ß (GSK­3ß), p53, cyclin E and cyclin­dependent kinase 2 (CDK2) were determined by western blot analysis. The protein levels of phosphorylated (p)­Akt, p­GSK­3ß, p­MDM2 and cyclin E were decreased, whereas the protein levels of p53, p21 and p­CDK2 (Thr14/Tyr15) were increased following treatment with CME. Furthermore, treatment or co­treatment with LY294002 (phosphoinositide­3­kinase/Akt inhibitor) or Pifithrin­α (p53 inhibitor) with CME resulted in CME­induced G1 arrest which occurred through the p53­independent signaling pathway in hepatocellular carcinoma cells. In conclusion, CME induces G1 arrest and apoptosis via the Akt/GSK­3ß signaling pathway which is regulated by MDM2­induced degradation of p21, rather than p53.


Subject(s)
Apoptosis/drug effects , Cell Cycle Checkpoints/drug effects , Cnidium/chemistry , Plant Extracts/pharmacology , Signal Transduction/drug effects , Tumor Suppressor Protein p53/metabolism , Carcinoma, Hepatocellular/metabolism , Cell Proliferation/drug effects , Cyclin-Dependent Kinase Inhibitor p21/metabolism , Glycogen Synthase Kinase 3 beta/metabolism , Hep G2 Cells , Humans , Liver Neoplasms/metabolism , Proto-Oncogene Proteins c-akt/metabolism
9.
Oncol Rep ; 38(2): 1206-1212, 2017 Aug.
Article in English | MEDLINE | ID: mdl-28677807

ABSTRACT

The number of patients who die from lung cancer is steadily increasing. According to the 2012 statistics, lung cancer accounts for the highest percentage of death from cancer in both sexes. Many research studies found that lung cancer can be caused not only by smoking but by outdoor pollution, and it leads to over-activation of various surface proteins in cancer cells. The over-activity of epidermal growth factor receptor (EGFR) is implicated as a crucial factor in inducing abnormal metastasis of lung cancer cells. In this study, we investigated the inhibitory effect of Torilis japonica extract (TJE) major fraction compound in A549 lung cancer cells by inhibiting EGFR activity. We confirmed that inhibitory effect of TJE on the abnormal metastasis using invasion assay and 3D cell culture method, as well as the inhibition of EGFR signaling pathway, co-binding with Stat3 and dimer formation for translocation to the nucleus. We confirmed the EGFR targets inhibition of TJE when compared with EGFR knockdown group using siRNA transfection. The CAM assay confirmed once again the efficacy of the TJE. We suggest that TJE is a new potential reagent for EGFR-targeted therapy and anti-abnormal metastasis in A549 lung cancer cells.


Subject(s)
Apiaceae/chemistry , Carcinoma, Non-Small-Cell Lung/drug therapy , ErbB Receptors/antagonists & inhibitors , Gene Expression Regulation, Neoplastic/drug effects , Lung Neoplasms/drug therapy , Plant Extracts/pharmacology , Apoptosis/drug effects , Carcinoma, Non-Small-Cell Lung/metabolism , Carcinoma, Non-Small-Cell Lung/secondary , Cell Movement/drug effects , Cell Proliferation/drug effects , Humans , Lung Neoplasms/metabolism , Lung Neoplasms/pathology , Tumor Cells, Cultured
10.
BMC Complement Altern Med ; 17(1): 236, 2017 Apr 28.
Article in English | MEDLINE | ID: mdl-28454566

ABSTRACT

BACKGROUND: The extracts from Artemisia annua Linné (AAE) has been known to possess various functions including anti-bacterial, anti-virus and anti-oxidant effects. However, the mechanism of those effects of AAE is not well known. Pursuantly, we determined the apoptotic effects of extract of AAE in HCT116 cell. In this study, we suggested that AAE may exert cancer cell apoptosis through PTEN/PDK1/Akt/p53signal pathway and mitochondria-mediated apoptotic proteins. METHODS: We measured 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide (MTT) assay, lactate dehydrogenase (LDH) assay, Hoechst 33342 staining, Annexin V-PI staining, Mitopotential assay, immunofluorescence (IF) and Western blotting. Accordingly, our study showed that AAE treatment to HCT116 cells resulted in inhibition of PDK1, Akt, MDM2, Bcl-2, and pro-caspase 3 as well as activation of PTEN, p53-upregulated modulator of apoptosis (PUMA), Bax and Bak expression. Also we measured in vivo assay that xenograft model, H&E assay, TUNEL assay and IHC. RESULTS: AAE induced apoptosis via PTEN/p53/PDK1/Akt signal pathways through PTEN/p53-independent manner. AAE inhibit cell viability and increase LDH release in HCT116 colon cancer cell. Also, AAE increase apoptotic bodies, caspase -3,7 activation and reduces mitochondria membrane potential. AAE regulates cytochrome c translocation to the cytoplasm and Bax translocation to the mitochondrial membrane in an Immunofluorescence staining and increase PTEN and p53 expression in an in vivo tumor xenograft model. To elucidate the role of the PTEN/p53/PDK1/Akt signal pathways in cancer control, we conditionally inactivated PTEN/p53/PDK1/Akt signal pathways. We used inhibitors of PTEN, p53, PDK1, Akt. In consequence, these results indicate that AAE induced apoptosis by means of a mitochondrial event through the regulation of proteins such as Bax, Bak and cytochrome c in PDK1/Akt signaling pathways via PTEM/p53-independent manner. CONCLUSIONS: We confirmed the apoptotic effect of extracts of AAE by Modulating PTEN/p53/PDK1/Akt/Signal Pathways through PTEN/p53-independent pathwaysin HCT116 colon cancer cell.


Subject(s)
Apoptosis/drug effects , Artemisia annua , Colonic Neoplasms/metabolism , Plant Extracts/pharmacology , Protein Serine-Threonine Kinases/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Tumor Suppressor Proteins/metabolism , Antineoplastic Agents, Phytogenic/pharmacology , Antineoplastic Agents, Phytogenic/therapeutic use , Apoptosis Regulatory Proteins/metabolism , Colonic Neoplasms/drug therapy , HCT116 Cells , Humans , Membrane Potential, Mitochondrial/drug effects , Mitochondria/drug effects , Mitochondria/metabolism , PTEN Phosphohydrolase/metabolism , Phytotherapy , Plant Extracts/therapeutic use , Proto-Oncogene Proteins/metabolism , Pyruvate Dehydrogenase Acetyl-Transferring Kinase , Signal Transduction , Tumor Suppressor Protein p53/metabolism
11.
Int J Oncol ; 49(3): 1088-98, 2016 Sep.
Article in English | MEDLINE | ID: mdl-27314881

ABSTRACT

Torilis japonica extract (TJE) has been reported to possess diverse medicinal properties including anti­inflammatory and antibacterial activities. However, the precise mechanism of its anticancer effect is not understood. Thus, we evaluated the apoptotic effects of TJE and examined its underlying molecular mechanisms in HCT116 colorectal cancer cells. Our results show that TJE induces apoptosis through the generation of intracellular reactive oxygen species (ROS), and that it regulates the mitochondrial outer membrane potential via the AMPK/p38 MAPK signaling pathway. Importantly, ~50% of cancer cells have p53 mutations. Thus, the ability to induce apoptosis in a p53-independent manner would be of great value in cancer treatment. Our results show that not only does TJE regulate the AMPK/p38 signaling pathway, but it induces apoptosis in cells in which p53 has been knocked down using siRNA. Moreover, as in in vitro studies, TJE induced apoptosis and regulated apoptosis related-proteins in an HCT 116 xenograft model. Taken together, our results demonstrate that TJE, a natural compound that may provide a substitute for chemotherapeutic drugs, has potential as an anticancer agent.


Subject(s)
Antineoplastic Agents/administration & dosage , Colonic Neoplasms/drug therapy , MAP Kinase Signaling System/drug effects , Membrane Potential, Mitochondrial/drug effects , Plant Extracts/administration & dosage , Tracheophyta/chemistry , Animals , Antineoplastic Agents/pharmacology , Apoptosis , Cell Survival/drug effects , Colonic Neoplasms/genetics , Colonic Neoplasms/metabolism , HCT116 Cells , Humans , Mice , Mutation , Plant Extracts/pharmacology , Reactive Oxygen Species/metabolism , Tumor Suppressor Protein p53/genetics , Xenograft Model Antitumor Assays
12.
Mol Med Rep ; 13(6): 4681-8, 2016 Jun.
Article in English | MEDLINE | ID: mdl-27082059

ABSTRACT

Colon cancer, a common malignancy, can occur due to poor eating habits and increasing age. Consequently, careful regulation of eating habits may serve as a possible method for preventing the occurrence or progression of colon cancer. Extracts of the fruit of Cnidium monnieri (L.) Cusson are well­known as an effective herbal medicine for the treatment of pain in female genitalia and carbuncle. However, there have been no studies on the apoptotic effects of Cnidium monnieri (L.) Cusson (CME). Adenosine monophosphate­activated protein kinase (AMPK), the major regulator of energy metabolism, is activated by metabolic stress, including hypoxia and glucose deprivation. Activation of AMPK inhibits cell proliferation and induces apoptosis through the inhibition of phosphorylated (p)­Akt and control of B­cell lymphoma 2 (Bcl­2) family members. The pro­apoptotic proteins Bcl­2­associated X protein (Bax) and Bcl­2­homologous antagonist killer (Bak), are activated by their translocation to mitochondria from the cytosol. Translocation of Bax/Bak induces outer membrane permeabilization and is likely to lead to apoptosis through cytochrome C release and caspase activity. In the present study, the apoptotic effects and influence on mitochondria­mediated apoptotic proteins of CME in HCT116 cells were assessed. We hypothesized that CME may have an effect on the inhibition of p­Akt in an AMPK­independent pathway. The present study demonstrated that CME induced the release of LDH and apoptosis through its inhibition of p­Akt to control Bcl­2 and activate Bax and Bak. Co­treatment with CME and AMPK inhibitors showed that CME­induced apoptosis does not occurr through a AMPK­dependent pathway. Therefore, the present study determined, for the first time, that CME induced apoptosis as a result of causing metabolic stresses due to directly regulation of the de­phosphorylation of Akt, whereas it did not control the AMPK-dependent pathway in HCT116 colon cancer cells.


Subject(s)
AMP-Activated Protein Kinases/metabolism , Antineoplastic Agents, Phytogenic/pharmacology , Apoptosis/drug effects , Cnidium/chemistry , Colonic Neoplasms/drug therapy , Signal Transduction/drug effects , Antineoplastic Agents, Phytogenic/chemistry , Colon/drug effects , Colon/metabolism , Colon/pathology , Colonic Neoplasms/metabolism , Colonic Neoplasms/pathology , HCT116 Cells , Humans , Phosphorylation/drug effects , Proto-Oncogene Proteins c-akt/metabolism , Proto-Oncogene Proteins c-bcl-2/metabolism , bcl-2 Homologous Antagonist-Killer Protein/metabolism , bcl-2-Associated X Protein
13.
Int J Oncol ; 45(4): 1673-9, 2014 Oct.
Article in English | MEDLINE | ID: mdl-25051266

ABSTRACT

Abnormal metastasis of carcinoma is associated with the loss of epithelial features and the acquisition of a mesenchymal phenotype. The stimulation of cells with epidermal growth factor (EGF) resulted in morphological changes and induced epithelial-mesenchymal transition (EMT). EGF stimulation resulted in increased mobility along with upregulated actin polarization related proteins, E-cadherin regulators and the mesenchymal markers. Treatment with Torilis japonica extract (TJE) along with stimulation by EGF prevented changes in cell morphology, mobility, expression of actin polarization proteins and EMT markers. Using specific inhibitors and siEGFR, it was demonstrated that TJE suppressed EMT through EGFR inactivation and regulation of its downstream signaling pathways. We suggest that TJE is a new potential reagent for EGFR-targeted therapy and anti-abnormal metastasis in MCF-7 breast cancer.


Subject(s)
Apiaceae/chemistry , Breast Neoplasms/metabolism , Epithelial-Mesenchymal Transition/drug effects , ErbB Receptors/genetics , ErbB Receptors/metabolism , Plant Extracts/pharmacology , Signal Transduction/drug effects , Breast Neoplasms/pathology , Cell Movement/drug effects , Cell Proliferation/drug effects , Female , Gene Expression Regulation, Neoplastic/drug effects , Gene Knockdown Techniques , Humans , MCF-7 Cells , Neoplasm Metastasis , RNA, Small Interfering/metabolism
14.
Int J Mol Med ; 33(4): 863-9, 2014 Apr.
Article in English | MEDLINE | ID: mdl-24535669

ABSTRACT

The induction of apoptosis in cancer cells is a therapeutic strategy for the treatment of cancer. In the present study, we investigated the regulatory mechanisms responsible for quercetin-induced apoptosis, mamely the increased expression of sestrin 2 and the activation of the 5' AMP-activated protein kinase (AMPK)/p38 MAPK signaling pathway. Our results revealed that quercetin induced apoptosis by generating the production of intracellular reactive oxygen species (ROS) and increasing the expression of sestrin 2. The induction of apoptosis by quercetin occurred through the activation of the AMPK/p38 signaling pathway and was dependent on sestrin 2. However, the silencing of sestrin 2 using small interfering RNA (siRNA) targeting sestrin 2 revealed that quercetin did not regulate AMPK or p38 phosphorylation in the cells in which sestrin 2 was silenced. On the other hand, it has been previously reported that sestrin 2 expression is not dependent on p53 expression under hypoxic conditions, whereas DNA damage is dependent on p53. We demonstrate that the increase in the expression of sestrin 2 by quercetin-generated intracellular ROS is p53-independent. The increased expression of sestrin 2 induced apoptosis through the AMPK/p38 signaling pathway in the HT-29 colon cancer cells, which are p53 mutant, treated with quercetin. Thus, our data suggest that quercetin induces apoptosis by reducing mitochondrial membrane potential, generating intracellular ROS production and increasing sestrin 2 expression through the AMPK/p38 pathway. In addition, p53 is not a necessary element for an apoptotic event induced by sestrin 2.


Subject(s)
AMP-Activated Protein Kinases/metabolism , Apoptosis/drug effects , MAP Kinase Signaling System/drug effects , Nuclear Proteins/metabolism , Quercetin/pharmacology , Reactive Oxygen Species/metabolism , Tumor Suppressor Protein p53/metabolism , Acetylcysteine/pharmacology , Benzothiazoles/pharmacology , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Survival/drug effects , Enzyme Activation/drug effects , Humans , Imidazoles/pharmacology , Intracellular Space/drug effects , Intracellular Space/metabolism , Pyridines/pharmacology , RNA, Small Interfering/metabolism , Toluene/analogs & derivatives , Toluene/pharmacology , Transfection , bcl-2-Associated X Protein/metabolism , p38 Mitogen-Activated Protein Kinases/metabolism
15.
J Cancer Prev ; 18(3): 264-70, 2013 Sep.
Article in English | MEDLINE | ID: mdl-25337554

ABSTRACT

BACKGROUND: The suppression of abnormal cell proliferation is therapeutic strategies for the treatment of cancer. In this study, we investigated the regulatory mechanism of quercetin-induced apoptosis through regulation of Sestrin 2 and AMPK signaling pathway. METHODS: After treatment of quercetin to colon cancer cells, intracellular ROS was detected using by DCFH-DA. To examine how quercetin and H2O2 induced apoptosis, we analyzed the change of Sestrin 2, p53 expression and p-AMPKα1, p-mTOR levels by Western blotting. To evaluate the effect of intracellular ROS generated by quercetin on colon cancer cells, NAC, anti-oxidative agent, was co-treated. RESULTS: Quercetin increased apoptotic cell death though generating intracellular reactive oxygen species (ROS), and it was responsible for Sestrin 2 expression. Increased Sestrin 2 expression was accompanied by AMPK activation. Interestingly, mTOR activity by Sestirn 2 expression was dependent on AMPK phosphorylation. On the other hand, the expression of Sestrin 2 by quercetin-generated intracellular ROS was independent of p53. CONCLUSIONS: We suggested that quercetin-induced apoptosis involved Sestrin 2/AMPK/mTOR pathway, which was regulated by increased intracellular ROS by quercetin.

SELECTION OF CITATIONS
SEARCH DETAIL