Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 37
Filter
1.
In Vivo ; 38(2): 665-673, 2024.
Article in English | MEDLINE | ID: mdl-38418101

ABSTRACT

BACKGROUND/AIM: Particular matter 2.5 (PM2.5) pollution is associated with senescence induction. Since the impact of PM2.5 on stem cell senescence and potential compounds capable of reversing this process are largely unknown, this study aimed to examine the senescence effects of PM2.5 on dermal papilla (DP) stem cells. Additionally, we explored the reversal of these effects using natural product-derived substances, such as resveratrol (Res) or Emblica fruits, soybean, and Thunbergia Laurifolia (EST) extract. MATERIALS AND METHODS: Cell senescence was determined using the ß-Galactosidase (SA-ß-gal) assay. The senescence-associated secretory phenotype (SASP) was detected using real-time RT-PCR. For senescence markers, the mRNA and protein levels of p21 and p16 were measured using real-time RT-PCR and immunofluorescence analysis. RESULTS: Subtoxic concentration of PM2.5 (50 µg/ml) induced senescence in DP cells. Resveratrol (50, 100 µM) and plant extracts (400, 800 µg/ml) reversed PM2.5-induced cell senescence. Treatment with Res or EST significantly decreased SA-ß-gal staining in PM2.5-treated cells. Furthermore, Res and EST decreased the mRNA levels of SASP, including IL1α, IL7, IL8, and CXCL1. DP cells exposed to PM2.5 exhibited an increase in p21 and p16 mRNA and protein levels, which could be reversed by the addition of Res or EST. Res and EST could reduce p21 and p16 in senescent cells approximately 3- and 2-fold, respectively, compared to untreated senescent cells. CONCLUSION: PM2.5 induced senescence in human DP stem cells. Res and EST extract potentially reverse the senescence phenotypes of such cells.


Subject(s)
Cellular Senescence , Plant Extracts , Humans , Resveratrol/pharmacology , Cellular Senescence/genetics , RNA, Messenger/genetics , Plant Extracts/pharmacology , Particulate Matter
2.
Antioxidants (Basel) ; 12(9)2023 Sep 18.
Article in English | MEDLINE | ID: mdl-37760080

ABSTRACT

Non-small cell lung cancer (NSCLC) poses a significant global health burden with unsatisfactory survival rates, despite advancements in diagnostic and therapeutic modalities. Novel therapeutic approaches are urgently required to improve patient outcomes. Pharmacological ascorbate (P-AscH-; ascorbate at millimolar concentration in plasma) emerged as a potential candidate for cancer therapy for recent decades. In this present study, we explore the anti-cancer effects of P-AscH- on NSCLC and elucidate its underlying mechanisms. P-AscH- treatment induces formation of cellular oxidative distress; disrupts cellular bioenergetics; and leads to induction of apoptotic cell death and ultimately reduction in clonogenic survival. Remarkably, DNA and DNA damage response machineries are identified as vulnerable targets for P-AscH- in NSCLC therapy. Treatments with P-AscH- increase the formation of DNA damage and replication stress markers while inducing mislocalization of DNA repair machineries. The cytotoxic and genotoxic effects of P-AscH- on NSCLC were reversed by co-treatment with catalase, highlighting the roles of extracellular hydrogen peroxide in anti-cancer activities of P-AscH-. The data from this current research advance our understanding of P-AscH- in cancer treatment and support its potential clinical use as a therapeutic option for NSCLC therapy.

4.
BMC Complement Med Ther ; 22(1): 139, 2022 May 18.
Article in English | MEDLINE | ID: mdl-35585532

ABSTRACT

BACKGROUND: Crinum latifolium L. (Amaryllidaceae) has been used in Southeast Asian traditional medicine to alleviate the symptoms of benign prostatic hyperplasia (BPH). The pathological mechanism of BPH is associated with the induction of prostate stromal cell proliferation through transforming growth factor-beta (TGF-ß). Standardization as well as investigation of the potential anti-BPH activity of C. latifolium extract could benefit the further development of BPH-related analyses and provide evidence to support the application of this extract for BPH treatment. This study aimed to standardize and investigate the antiproliferative activity of the ethanolic extract of C. latifolium leaves. The major alkaloids isolated from C. latifolium were also explored for their potential use as bioactive markers. METHODS: Two major alkaloids were isolated from the ethanolic extract of C. latifolium leaves by chromatographic techniques, identified by NMR and MS, and quantified by a validated UHPLC method. Their antiproliferative activity was studied in human prostate stromal cells (WPMY-1) induced by TGF-ß. The synergistic effect of combining the two major isolated alkaloids was analyzed by the zero interaction potency (ZIP) model. RESULTS: Two alkaloids, lycorine (1) and 6α-hydroxybuphanidrine (2), were isolated from the ethanolic leaf extract of C. latifolium. A UHPLC method for the quantification of (1) and (2) was developed and validated in terms of linearity, precision, and accuracy. The C. latifolium leaf extract contained 0.279 ± 0.003% (1) and 0.232 ± 0.004% (2). The crude extract was more potent than either (1) and (2) alone against TGF-ß-treated WPMY-1 cell proliferation. The drug combination study revealed that the greatest synergistic effect of (1) and (2) was achieved at a 1:1 ratio. CONCLUSIONS: The results of this study support the anti-BPH activity of C. latifolium in traditional medicine and suggest that these the two isolated alkaloids may promote the efficacy of the C. latifolium extract. Additionally, major alkaloids (1) and (2) can be used as bioactive markers for the standardization of C. latifolium extracts.


Subject(s)
Alkaloids , Crinum , Prostatic Hyperplasia , Alkaloids/pharmacology , Crinum/chemistry , Humans , Male , Plant Extracts/pharmacology , Plant Extracts/therapeutic use , Prostate/pathology , Prostatic Hyperplasia/drug therapy , Reference Standards , Stromal Cells/pathology , Transforming Growth Factor beta
5.
Article in English | MEDLINE | ID: mdl-34349823

ABSTRACT

Cancer stem-like cells (CSCs) are key mediators driving tumor initiation, metastasis, therapeutic failure, and subsequent cancer relapse. Thus, targeting CSCs has recently emerged as a potential strategy to improve chemotherapy. In this study, the anticancer activity and stemness-regulating capacity of 4,5,4'-trihydroxy-3,3'-dimethoxybibenzyl (TDB), a bibenzyl extracted from Dendrobium ellipsophyllum, are revealed in CSCs of various human lung cancer cells. Culture with TDB (5-10 µM) strongly abolished tumor-initiating cells in lung cancer H460, H23, and A549 cells in both anchorage-dependent and anchorage-independent colony formation assays. Through the 3D single-spheroid formation model, attenuation of self-renewal capacity was observed in CSC-enriched populations treated with 1-10 µM TDB for 7 days. Flow cytometry analysis confirmed the attenuation of %cell overexpressing CD133, a CSC biomarker, in TDB-treated lung cancer spheroids. TDB at 5-10 µM remarkably suppressed regulatory signals of p-Akt/Akt, p-GSK3ß/GSK3ß, and ß-catenin corresponding to the downregulated mRNA level of stemness transcription factors including Nanog, Oct4, and Sox2. Moreover, the antiapoptosis Bcl-2 and Mcl-1 proteins, which are downstream molecules of Akt signaling, were evidently decreased in CSC-enriched spheroids after culture with TDB (1-10 µM) for 24 h. Interestingly, the diminution of Akt expression by specific siAkt effectively reversed suppressive activity of TDB targeting on the CSC phenotype in human lung cancer cells. These findings provide promising evidence of the inhibitory effect of TDB against lung CSCs via suppression of Akt/GSK3ß/ß-catenin cascade and related proteins, which would facilitate the development of this bibenzyl natural compound as a novel CSC-targeted therapeutic approach for lung cancer treatment.

6.
J Nat Med ; 75(4): 949-966, 2021 Sep.
Article in English | MEDLINE | ID: mdl-34287745

ABSTRACT

The incidence of metastasis stage crucially contributes to high recurrence and mortality rate in lung cancer patients. Unfortunately, no available treatment inhibits migration, a key metastasis process in lung cancer. In this study, the effect of 22-O-(N-Boc-L-glycine) ester of renieramycin M (22-Boc-Gly-RM), a semi-synthetic amino ester derivative of bistetrahydroisoquinolinequinone alkaloid isolated from Xestospongia sp., on migratory behavior of human lung cancer cells was investigated. Following 24 h of treatment, 22-Boc-Gly-RM at non-toxic concentrations (0.5-1 µM) effectively restrained motility of human lung cancer H460 cells assessed through wound healing, transwell migration, and multicellular spheroid models. The capability to invade through matrix component was also repressed in H460 cells cultured with 0.1-1 µM 22-Boc-Gly-RM. The dose-dependent reduction of phalloidin-stained actin stress fibers corresponded with the downregulated Rac1-GTP level presented via western blot analysis in 22-Boc-Gly-RM-treated cells. Treatment with 0.1-1 µM of 22-Boc-Gly-RM obviously caused suppression of p-FAK/p-Akt signal and consequent inhibition of epithelial-to-mesenchymal transition (EMT), which was evidenced with augmented level of E-cadherin and reduction of N-cadherin expression. The alteration of invasion-related proteins in 22-Boc-Gly-RM-treated H460 cells was indicated by the diminution of matrix metalloproteinases (MT1-MMP, MMP-2, MMP-7, and MMP-9), as well as the upregulation of tissue inhibitors of metalloproteinases (TIMP), TIMP2, and TIMP3. Thus, 22-Boc-Gly-RM is a promising candidate for anti-metastasis treatment in lung cancer through inhibition of migratory features associated with suppression on EMT.


Subject(s)
Epithelial-Mesenchymal Transition , Lung Neoplasms , Cell Line, Tumor , Cell Movement , Cell Survival , Esters , Glycine/pharmacology , Humans , Lung Neoplasms/drug therapy , Tetrahydroisoquinolines
7.
Phytomedicine ; 85: 153534, 2021 May.
Article in English | MEDLINE | ID: mdl-33773191

ABSTRACT

BACKGROUND: Lung cancer is a leading fatal malignancy due to the high incidence of treatment failure. Dysfunction of the tumor suppressor p53 contributes to cancer initiation, progression, and therapeutic resistance. Targeting MDM2, a negative regulator of p53, has recently attracted interest in cancer drug research as it may restore tumor suppressive function. PURPOSE: The present study aimed to investigate the effect of 3,4-dihydroxy-5,4'-dimethoxybibenzyl (DS-1) on targeting MDM2 and restoring p53 function in lung cancer cells. METHODS: The efficacy of DS-1 alone or in combination with cisplatin in lung cancer cells was determined by MTT, nuclear staining, and annexin V/PI assay. The expression of apoptosis-related proteins was determined by western blot analysis. To evaluate the role of DS-1 on the stabilization and degradation of p53, cycloheximide chasing assay and immunoprecipitation were conducted, and the active form of p53 was investigated by immunofluorescent staining assay. To confirm and demonstrate the site interaction between DS-1 and the MDM2 protein, in silico computational analysis was performed. RESULTS: DS-1 exhibited a cytotoxic effect and sensitized lung cancer cells to cisplatin-induced apoptosis. DS-1 caused a significant increase in the cellular level of p53 protein, while the active form of p53 (phosphorylation at Ser15) was unaltered. DS-1 treatment in combination with cisplatin could enhance activated p-p53 (Ser15) and p53 downstream signaling (Bax, Bcl-2, and Akt), leading to a higher level of apoptosis. Immunoprecipitation analysis revealed that DS-1 decreased the p53-ubiquitin complex, a prerequisite step in p53 proteasomal degradation. Molecular docking simulation further evidenced that DS-1 interacts with MDM2 within the p53-binding domain by carbon-hydrogen bond interaction at Lys27, π-alkyl interactions at Ile37 and Leu30, and van der Waals interactions at Ile75, Val51, Val69, Phe67, Met38, Tyr43, Gly34, and Phe31. Treatment by DS-1 and cisplatin in patient-derivated primary lung cancer cells showed consistent effects by increasing cisplatin sensitivity. CONCLUSIONS: Our findings provide evidence that DS-1 is an MDM2 inhibitor and its underlying mechanism involves MDM2 binding and p53 induction, which may benefit the development of this compound for lung cancer treatment.


Subject(s)
Antineoplastic Agents/pharmacology , Apoptosis/drug effects , Bibenzyls/pharmacology , Lung Neoplasms/pathology , Proto-Oncogene Proteins c-mdm2/antagonists & inhibitors , Tumor Suppressor Protein p53/metabolism , Adult , Aged , Cell Line, Tumor , Cisplatin/pharmacology , Female , Humans , Male , Middle Aged , Molecular Docking Simulation , Proto-Oncogene Proteins c-mdm2/metabolism , Signal Transduction/drug effects
8.
Sci Rep ; 11(1): 4060, 2021 02 18.
Article in English | MEDLINE | ID: mdl-33603033

ABSTRACT

The limitations of cisplatin, a standard chemotherapy for lung cancer, have been documented with serious adverse effects and drug resistance. To address the need for novel therapy, this study firstly reveals the potential of peptide from Lentinus squarrosulus (Mont.) as a chemotherapeutic adjuvant for cisplatin treatment. The purified peptide from L. squarrosulus aqueous extracts was obtained after eluting with 0.4 M NaCl through FPLC equipped with anion exchange column. Preincubation for 24 h with 5 µg/mL of the peptide at prior to treatment with 5 µM cisplatin significantly diminished %cell viability in various human lung cancer cells but not in human dermal papilla and proximal renal cells. Flow cytometry indicated the augmentation of cisplatin-induced apoptosis in lung cancer cells pretreated with peptide from L. squarrosulus. Preculture with the peptide dramatically inhibited colony formation in lung cancer cells derived after cisplatin treatment. Strong suppression on integrin-mediated survival was evidenced with the diminution of integrins (ß1, ß3, ß5, α5, αV) and down-stream signals (p-FAK/FAK, p-Src/Src, p-Akt/Akt) consequence with alteration of p53, Bax, Blc-2 and Mcl-1 in cisplatin-treated lung cancer cells preincubated with peptide from L. squarrosulus. These results support the development of L. squarrosulus peptide as a novel combined chemotherapy with cisplatin for lung cancer treatment.


Subject(s)
Antineoplastic Agents/pharmacology , Apoptosis/drug effects , Cisplatin/pharmacology , Lentinula/chemistry , Lung Neoplasms/drug therapy , Peptides/therapeutic use , Plant Extracts/therapeutic use , Adjuvants, Pharmaceutic/pharmacology , Adjuvants, Pharmaceutic/therapeutic use , Blotting, Western , Cell Line, Tumor , Drug Synergism , Flow Cytometry , Humans , Peptides/isolation & purification , Peptides/pharmacology , Plant Extracts/pharmacology
9.
BMC Complement Med Ther ; 20(1): 46, 2020 Feb 11.
Article in English | MEDLINE | ID: mdl-32046712

ABSTRACT

BACKGROUND: Cyanidin-3-glucoside (C3G), a major anthocyanin present in berries, exhibits a strong antioxidant and has been shown to possess a neuroprotection. Prolonged exposure to glutamate will lead to oxidative damage and endoplasmic reticulum stress which could play a key detrimental role in the development of neurodegenerative disorders (NDs). In the present study, we investigated the neuroprotective effect and underlying mechanisms of C3G on the reduction of oxidative/ER stress-induced apoptosis by glutamate in HT22 mouse hippocampal neuronal cells. METHOD: Cells were pre-treated with C3G in various concentrations, followed by glutamate. Cell viability and toxicity were examined using MTT and LDH assays. The apoptotic and necrotic cell death were carried out by Annexin V-FITC/propidium iodide co-staining assays. Generation of intracellular reactive oxygen species (ROS) in cells was measured by flow cytometry using DCFH-DA probe. Expression of antioxidant genes was evaluated by Real-time polymerase chain reaction analysis. The possible signaling pathways and proteins involved were subsequently demonstrated by Western blot analysis. RESULT: The pretreatment of the HT22 cells with C3G protected cell death from oxidative toxicity induced by glutamate. We demonstrated that treatment cells with glutamate caused several radical forms of ROS formation, and they were abolished by specific ROS inhibitors. Interestingly, C3G directly scavenged radical activity and inhibited intracellular ROS generation in our cell-based system. In addition, C3G pretreatment suppressed the up-regulation of specific ER proteins namely calpain, caspase-12 and C/EBP homologous proteins (CHOP) induced by glutamate-mediated oxidative and ER stress signal by up-regulating the expressions of survival proteins, including extracellular regulated protein kinase (ERK) and nuclear factor E2-related factor 2 (Nrf2). Furthermore, dramatically activated gene expression of endogenous antioxidant enzymes (i.e. superoxide dismutases (SODs), catalase (CAT) and glutathione peroxidase (GPx)), and phase II enzymes (glutathione-S-transferases (GSTs)) was found in C3G-treated with cells. CONCLUSIONS: Our finding suggest that C3G could be a promising neuroprotectant via inhibition of glutamate-induced oxidative and ER stress signal and activation of ERK/Nrf2 antioxidant mechanism pathways.


Subject(s)
Anthocyanins/pharmacology , Endoplasmic Reticulum Stress/drug effects , Glucosides/pharmacology , NF-E2-Related Factor 2/metabolism , Neurons/drug effects , Neuroprotective Agents/pharmacology , Plant Extracts/pharmacology , Animals , Antioxidants/metabolism , Cell Line , Flow Cytometry , Glutamic Acid , Hippocampus/cytology , Mice , Molecular Structure
10.
Phytomedicine ; 62: 152932, 2019 Sep.
Article in English | MEDLINE | ID: mdl-31100681

ABSTRACT

BACKGROUND: Cancer stem cells (CSCs) are well-recognized as a majority cause of treatment failure and can give rise to relapse. The discovery of compounds attenuating CSCs' properties is crucial for enabling advances in novel therapeutics to limit recurrence. CSCs' features in lung cancer are regulated through a reduction in Src-STAT3-c-Myc, which drives cancer progression, drug resistance, and metastasis. METHODS: The effect of lusianthridin suppresses CSC-like phenotypes was determined by 3D culture and anchorage independent growth. The expression of CSC markers and associated proteins were determined by Western blot analyses. Protein ubiquitination and degradation were assessed using immunoprecipitation. RESULTS: Herein, we report that lusianthridin, a pure compound from Dendrobium venustum, dramatically suppressed CSCs in lung cancer cells as verified by several CSC phenotype assessments and CSC markers. The CSC phenotypes in lusianthridin-treated cells were suppressed through downregulation of Src-STAT3-c-Myc pathways. Ectopic Src introduced by the transfection augmented CSC phenotypes in lung cancer cells through STAT3 (increased active p-STAT3Tyr705) and c-Myc signals, while the ShRNA-Src transfection or Src inhibitor dasatinib exhibited opposite results. Treatment of the Src-overexpressing cells with lusianthridin resulted in the reversal of active STAT3 (p-STAT3Tyr705) and c-Myc as well as the CSC marker CD133. Importantly, we confirmed the CSC-targeted activity of lusianthridin in CSC-rich primary lung cancer cells. The compound dramatically inhibited the formation of tumor spheres of primary lung cancer cells. Finally, we demonstrated that after CSC-attenuation by lusianthridin, the lung cancer cells exhibited significantly higher susceptibility to chemotherapeutic drugs. Such a sensitizing effect caused by pro-survival suppression and pro-apoptotic induction together with the abolishment of stemness indicated by the decrease in CSC markers CD133, ABCG2, and ALDH1A1. CONCLUSION: These findings revealed a novel pharmacological action and the underlying mechanism of lusianthridin in negatively regulating CSC-like phenotypes and sensitizing resistant cancer cells to cemetery.


Subject(s)
Carcinoma, Non-Small-Cell Lung/drug therapy , Lung Neoplasms/drug therapy , Neoplastic Stem Cells/drug effects , Phenanthrenes/pharmacology , STAT3 Transcription Factor/metabolism , src-Family Kinases/metabolism , AC133 Antigen/metabolism , ATP Binding Cassette Transporter, Subfamily G, Member 2/metabolism , Aldehyde Dehydrogenase 1 Family/metabolism , Antineoplastic Agents, Phytogenic/pharmacology , Carcinoma, Non-Small-Cell Lung/metabolism , Carcinoma, Non-Small-Cell Lung/pathology , Cell Line, Tumor , Cisplatin/pharmacology , Dasatinib/pharmacology , Down-Regulation/drug effects , Humans , Lung Neoplasms/metabolism , Lung Neoplasms/pathology , Neoplasm Proteins/metabolism , Neoplastic Stem Cells/metabolism , Neoplastic Stem Cells/pathology , Protein Kinase Inhibitors/pharmacology , Proto-Oncogene Proteins c-myc/metabolism , Retinal Dehydrogenase/metabolism , STAT3 Transcription Factor/antagonists & inhibitors , Spheroids, Cellular/drug effects , src-Family Kinases/antagonists & inhibitors , src-Family Kinases/genetics
11.
Phytomedicine ; 58: 152888, 2019 May.
Article in English | MEDLINE | ID: mdl-30901662

ABSTRACT

BACKGROUND: A Lung cancer death account for approximately 1 in 5 of all cancer-related deaths and is particularly virulent due to its enhanced metastasis and resistance to chemotherapy. Chrysotobibenzyl has been reported to decrease cell metastasis, according to the results of an anchorage-independent growth assay; however, its underlying mechanism has not been investigated yet. PURPOSE: The aim of this study was to investigate the effect of chrysotobibenzyl on lung cancer cell migration and drug sensitization and its mechanism. METHODS: Cell viability, cell proliferation and drug sensitization were determined by MTT assay. Cell migration was analyzed using a wound-healing assay. Transwell migration and invasion were analyzed using Boyden chamber assay. Mechanisms of chrysotobibenzyl against metastasis including cell migration, invasion, and epithelial to mesenchymal transition (EMT) were evaluated by Western blot analysis and immunofluorescence. RESULTS: Treatment with chrysotobibenzyl was applied at concentrations of 0-50 µM and the results showed non-cytotoxicity in human lung cancer cells (H460, H292, A549, and H23) and other non-cancerous human cells (HCT116, primary DP1 and primary DP2). However, 50 µM of chrysotobibenzyl significantly altered cell proliferation in H292 cells at 48 h. In addition, 1-50 µM of chrysotobibenzyl significantly inhibited H460 and H292 cell migration, invasion, filopodia formation, and decreased EMT in a dose-dependent manner at 48 h, which were correlated with reduced protein levels of integrins ß1, ß3, and αν, p-FAK, p-AKT, Cdc42, and Cav-1. We also established shRNA-Cav-1-transfected (shCav-1) H460 and H292 cells. shCav-1 transfected cells can decrease cell migration and downregulate the expression of integrins ß1, ß3, and αν when compared with the control. Moreover, chrysotobibenzyl was shown to suppress EMT indicated by the reduction of EMT markers (Vimentin, Snail, and Slug), and sensitize lung cancer cells to cisplatin-mediated apoptosis. CONCLUSION: Treatment with chrysotobibenzyl inhibited lung cancer cell migration via Cav-1, integrins ß1, ß3, and αν, and EMT suppressions. The downregulation of integrins in response to the compound not only inhibited cell metastasis, but also sensitized lung cancer cells to cisplatin-mediated apoptosis.


Subject(s)
Antineoplastic Agents/pharmacology , Bibenzyls/pharmacology , Caveolin 1/metabolism , Integrins/metabolism , Lung Neoplasms/drug therapy , Antineoplastic Agents, Phytogenic/pharmacology , Apoptosis/drug effects , Cell Line, Tumor , Cell Movement/drug effects , Cell Proliferation/drug effects , Cell Survival/drug effects , Cisplatin/pharmacology , Down-Regulation/drug effects , Drug Interactions , Epithelial-Mesenchymal Transition/drug effects , Humans , Lung Neoplasms/pathology , Pseudopodia/drug effects
12.
Anticancer Res ; 39(2): 541-548, 2019 Feb.
Article in English | MEDLINE | ID: mdl-30711928

ABSTRACT

Integrins are cell-matrix adhesion molecules providing both mechanical engagement of cell to extracellular matrix, and generation of cellular signals that are implicated in cancer malignancies. The concept that integrins play important roles in cell survival, proliferation, motility, differentiation, and ensuring appropriate cell localization, leads to the hypothesis that inhibition of certain integrins would benefit cancer therapy. In lung cancer, integrins αv, α5, ß1, ß3, and ß5 have been shown to augment survival and metastatic potential of cancer cells. This review presents data suggesting integrins as molecular targets for anti-cancer approaches, and the mechanisms through which integrins confer resistance of lung cancer to chemotherapeutics and metastasis. The better understanding of these key molecules may benefit the discovery of anti-cancer drugs and strategies.


Subject(s)
Antineoplastic Agents/therapeutic use , Integrins/metabolism , Lung Neoplasms/drug therapy , Cell Adhesion , Cell Differentiation , Cell Proliferation , Cell Survival , Curcumin/pharmacology , Disease Progression , Extracellular Matrix/metabolism , Gene Expression Profiling , Gene Expression Regulation, Neoplastic , Humans , Neoplasm Metastasis , Ouabain/pharmacology , Phloretin/pharmacology , Xanthones/pharmacology
13.
J Nat Med ; 72(3): 615-625, 2018 Jun.
Article in English | MEDLINE | ID: mdl-29488156

ABSTRACT

Failure of current chemotherapeutic drugs leads to the recurrence of tumor pathology and mortality in lung cancer patients. This study aimed to evaluate the anticancer activity and related mechanisms of 4,5,4'-trihydroxy-3,3'-dimethoxybibenzyl (TDB), a bibenzyl extracted from Dendrobium ellipsophyllum Tang and Wang, in human lung cancer cells. Cytotoxicity of TDB (0-300 µM) in different types of human lung cancer cells (H460, H292 and H23) and human dermal papilla cells (DPCs) was evaluated via MTT viability assay. Selective anticancer activity of TDB against human lung cancer cells was demonstrated with a high IC50 (approximately > 300 µM) in DPCs, while IC50 in human lung cancer H460, H292 and H23 cells was approximately 100 ± 5.18, 100 ± 8.73 and 188.89 ± 8.30 µM, respectively. After treatment with 50 µM of TDB for 24 h, flow cytometry analysis revealed the significant increase of early and late apoptosis with absence of necrosis cell death in human lung cancer cells. The up-regulation of p53, a tumor-suppressor protein, was elucidated in human lung cancer cells treated with 10-50 µM of TDB. Alteration to down-stream signaling of p53 including activation of pro-apoptosis protein (Bcl-2-associated X protein; Bax), reduction of anti-apoptosis (B cell lymphoma 2; Bcl-2 and myeloid cell leukemia 1; Mcl-1) and suppression on protein kinase B (Akt) survival pathway were notified in TDB-treated lung cancer cells. The information obtained from this study strengthens the potential development of TDB as an anticancer compound with a favorable human safety profile and high efficacy.


Subject(s)
Bibenzyls/chemistry , Dendrobium/chemistry , Lung Neoplasms/drug therapy , Apoptosis , Bibenzyls/pharmacology , Humans
14.
BMC Complement Altern Med ; 17(1): 553, 2017 Dec 29.
Article in English | MEDLINE | ID: mdl-29284478

ABSTRACT

BACKGROUND: The conversion of the epithelial phenotype of cancer cells into cells with a mesenchymal phenotype-so-called epithelial-mesenchymal transition (EMT)-has been shown to enhance the capacity of the cells to disseminate throughout the body. EMT is therefore becoming a potential target for anti-cancer drug discovery. Here, we showed that phoyunnanin E, a compound isolated from Dendrobium venustum, possesses anti-migration activity and addressed its mechanism of action. METHODS: The cytotoxic and proliferative effects of phoyunnanin E on human non-small cell lung cancer-derived H460, H292, and A549 cells and human keratinocyte HaCaT cells were investigated by MTT assay. The effect of phoyunnanin E on EMT was evaluated by determining the colony formation and EMT markers. The migration and invasion of H460, H292, A549 and HaCaT cells was evaluated by wound healing assay and transwell invasion assay, respectively. EMT markers, integrins and migration-associated proteins were examined by western blot analysis. RESULTS: Phoyunnanin E at the concentrations of 5 and 10 µM, which are non-toxic to H460, H292, A549 and HaCaT cells showed good potential to inhibit the migratory activity of three types of human lung cancer cells. The anti-migration effect of phoyunnanin E was shown to relate to the suppressed EMT phenotypes, including growth in anchorage-independent condition, cell motility, and EMT-specific protein markers (N-cadherin, vimentin, slug, and snail). In addition to EMT suppression, we found that phoyunnanin E treatment with 5 and 10 µM could decrease the cellular level of integrin αv and integrin ß3, these integrins are frequently up-regulated in highly metastatic tumor cells. We further characterized the regulatory proteins in cell migration and found that the cells treated with phoyunnanin E exhibited a significantly lower level of phosphorylated focal adhesion kinase (p-FAK) and phosphorylated ATP-dependent tyrosine kinase (p-AKT), and their downstream effectors (including Ras-related C3 botulinum (Rac-GTP); Cell division cycle 42 (Cdc42); and Ras homolog gene family, member A (Rho-GTP)) in comparison to those of the non-treated control. CONCLUSIONS: We have determined for the first time that phoyunnanin E could inhibit the motility of lung cancer cells via the suppression of EMT and metastasis-related integrins. This new information could support further development of this compound for anti-metastasis approaches.


Subject(s)
Carcinoma, Non-Small-Cell Lung/drug therapy , Cell Movement/drug effects , Epithelial-Mesenchymal Transition/drug effects , Integrins/metabolism , Lung Neoplasms/drug therapy , Phenanthrenes/pharmacology , Plant Extracts/pharmacology , A549 Cells , Carcinoma, Non-Small-Cell Lung/pathology , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Survival/drug effects , Dendrobium/chemistry , Humans , Lung Neoplasms/pathology , Phenanthrenes/chemistry , Plant Extracts/chemistry
15.
Food Chem Toxicol ; 103: 279-288, 2017 May.
Article in English | MEDLINE | ID: mdl-28315776

ABSTRACT

Oxidative and endoplasmic reticulum (ER) stresses cause neuronal damage leading to neurodegenerative disorders. Cleistocalyx nervosum var. paniala (CNP) berry fruit has been shown to possess powerful antioxidant properties. Here, we investigated the neuroprotective effect of CNP extract against glutamate-mediated oxidative/ER stress-induced cell death in mouse hippocampal neuronal HT22 cells. CNP extract was clarified for its radical scavenging activities, total phenolic and anthocyanin contents. The key anthocyanin cyanidin-3-glucoside was used as a marker to standardize the extract used in the study. We found that pretreated cells with CNP extract (0.05-1 µg/ml) prevented neuronal cell death in response to 5 mM glutamate evaluated by cell viability MTT, LDH and apoptosis/necrosis Annexin V/propidium iodide co-staining assays. For mechanistic approach, glutamate-induced cell death through reactive oxygen species (ROS)-mediated ER stress pathways, indicating the increase of ROS and ER stress signature molecules including calpain, caspases-12 and C/EBP homologous proteins (CHOP). CNP extract inhibited ROS production. Moreover, the extract also suppressed the specific-ER stress apoptotic proteins level in glutamate-induced cells by upregulating the gene expression of cellular antioxidant enzymes (SODs, CAT, GPx and GSTs). Taken together, our results provide information about and the molecular mechanism of CNP extract as a promising neuroprotectant and antioxidant.


Subject(s)
Apoptosis/drug effects , Endoplasmic Reticulum Stress/drug effects , Neurons/drug effects , Plant Extracts/pharmacology , Syzygium/chemistry , Animals , Anthocyanins/analysis , Anthocyanins/pharmacology , Antioxidants/analysis , Antioxidants/pharmacology , Apoptosis/physiology , Cell Line , Enzymes/genetics , Enzymes/metabolism , Fruit/chemistry , Gene Expression Regulation/drug effects , Glutamic Acid/adverse effects , Hippocampus/cytology , Mice , Neurons/metabolism , Neurons/pathology , Neurotoxicity Syndromes/prevention & control , Reactive Oxygen Species/metabolism
16.
Int J Oncol ; 50(4): 1341-1351, 2017 Apr.
Article in English | MEDLINE | ID: mdl-28259926

ABSTRACT

Cancer stem cells (CSCs) have been reported as a major cause of cancer metastasis and the failure of cancer treatment. Cumulative studies have indicated that protein kinase B (Akt) and its downstream signaling pathway, including CSC markers, play a critical role in the aggressive behavior of this cancer. In this study, we investigated whether vanillin, a major component in Vanilla planifolia seed, could suppress cancer stemness phenotypes and related proteins in the human non-small cell lung cancer NCI­H460 cell line. A non-toxic concentration of vanillin suppressed spheroid and colony formation, two hallmarks of the cancer stemness phenotype, in vitro in NCI­H460 cells. Western blot analysis revealed that the CSC markers CD133 and ALDH1A1 and the associated transcription factors, Oct4 and Nanog, were extensively downregulated by vanillin. Vanillin also attenuated the expression and activity of Akt, a transcription regulator upstream of CSCs, an action that was confirmed by treatment with the Akt inhibitor perifosine. Furthermore, the ubiquitination of Akt was elevated in response to vanillin treatment prior to proteasomal degradation. This finding indicates that vanillin can inhibit cancer stem cell-like behavior in NCI­H460 cells through the induction of Akt-proteasomal degradation and reduction of downstream CSC transcription factors. This inhibitory effect of vanillin may be an alternative approach in the treatment against lung cancer metastasis and its resistance to chemotherapy.


Subject(s)
Benzaldehydes/therapeutic use , Carcinoma, Non-Small-Cell Lung/drug therapy , Lung Neoplasms/drug therapy , Neoplastic Stem Cells/drug effects , Proto-Oncogene Proteins c-akt/metabolism , Signal Transduction/drug effects , AC133 Antigen/metabolism , Aldehyde Dehydrogenase/metabolism , Aldehyde Dehydrogenase 1 Family , Cell Line, Tumor , Colony-Forming Units Assay , Humans , Nanog Homeobox Protein/metabolism , Octamer Transcription Factor-3/metabolism , Phosphorylcholine/analogs & derivatives , Phosphorylcholine/pharmacology , Proteasome Endopeptidase Complex/metabolism , Proto-Oncogene Proteins c-akt/antagonists & inhibitors , Retinal Dehydrogenase , Spheroids, Cellular/drug effects , Vanilla/chemistry
17.
Article in English | MEDLINE | ID: mdl-27651818

ABSTRACT

Lung cancer remains a leading public health problem as evidenced by its increasing death rate. The main cause of death in lung cancer patients is cancer metastasis. The metastatic behavior of lung cancer cells becomes enhanced when cancer cells undergo epithelial to mesenchymal transition (EMT). Gigantol, a bibenzyl compound extracted from the Thai orchid, Dendrobium draconis, has been shown to have promising therapeutic potential against cancer cells, which leads to the hypothesis that gigantol may be able to inhibit the fundamental EMT process in cancer cells. This study has demonstrated for the first time that gigantol possesses the ability to suppress EMT in non-small cell lung cancer H460 cells. Western blot analysis has revealed that gigantol attenuates the activity of ATP-dependent tyrosine kinase (AKT), thereby inhibiting the expression of the major EMT transcription factor, Slug, by both decreasing its transcription and increasing its degradation. The inhibitory effects of gigantol on EMT result in a decrease in the level of migration in H460 lung cancer cells. The results of this study emphasize the potential of gigantol for further development against lung cancer metastasis.

18.
J Nat Med ; 70(1): 18-27, 2016 Jan.
Article in English | MEDLINE | ID: mdl-26384689

ABSTRACT

Metastasis in lung cancer has been recognized as an important cause of high mortality. Resistance to anoikis and the epithelial-to-mesenchymal transition (EMT) are critical factors for the successful spread of cancer cells. Compounds that suppress these features of cancer cells should be potentially active for anti-metastasis approaches. We have demonstrated for the first time that moscatilin, at its non-toxic concentrations to lung cancer cells and human normal keratinocytes, significantly decreases lung cancer cell survival in the detached condition, and suppresses the formation of tumors in an anchorage-independent growth assay. Furthermore, we found that moscatilin significantly decreased the activated level of survival proteins, namely ERK and Akt. In addition, moscatilin down-regulated cavelolin-1 (Cav-1), leading to a reduction in anti-apoptotic Mcl-1 protein. In terms of EMT, treatment of the cells with moscatilin significantly suppressed mesenchymal cell markers, namely vimentin, Slug, and Snail. These results indicate that moscatilin inhibited anoikis resistance in lung cancer cells via survival suppression, Cav-1 down-regulation, and inhibition of EMT. The compound could therefore be beneficial for the treatment and prevention of lung cancer metastasis.


Subject(s)
Anoikis/drug effects , Benzyl Compounds/pharmacology , Caveolin 1/biosynthesis , Cell Survival/drug effects , Epithelial-Mesenchymal Transition/drug effects , Lung Neoplasms/pathology , Vimentin/biosynthesis , Cell Line, Tumor , Cell Movement , Cell Proliferation/drug effects , Down-Regulation , Humans , Myeloid Cell Leukemia Sequence 1 Protein/metabolism , Neoplasm Metastasis , Snail Family Transcription Factors , Transcription Factors/biosynthesis
19.
Phytomedicine ; 22(14): 1269-78, 2015 Dec 15.
Article in English | MEDLINE | ID: mdl-26626191

ABSTRACT

BACKGROUND: Although the growth of unwanted hair or hirsutism is a harmless condition, many people find it bothersome and embarrassing. Maintaining stem cell features of dermal papilla cells is a critical biological process that keeps the high rate of hair growth. Glycyrrhizic acid has been reported to impair hair growth in some studies; however, its underlying mechanism has not yet been investigated. PURPOSE: This study aimed to explore the effect and underlying mechanism of glycyrrhizic acid on stemness of human dermal papilla cells. STUDY DESIGN/METHODS: The stem cell molecular markers, epithelial to mesenchymal markers and Wnt/ß-catenin-associated proteins of human dermal papilla cell line and primary human dermal papilla cells were analysed by western blot analysis and immunocytochemistry. RESULTS: The present study demonstrated that glycyrrhizic acid significantly depressed the stemness of dermal papilla cells in dose- and time-dependent manners. Clonogenicity and stem cell markers in the glycyrrhizic acid-treated cells were found to gradually decrease in the culture in a time-dependent manner. Our results demonstrated that glycyrrhizic acid exerted the stem cell suppressing effects through the interruption of ATP-dependent tyrosine kinase/glycogen synthase kinase3ß-dependent mechanism which in turn down-regulated the ß-catenin signalling pathway, coupled with decreased its down-stream epithelial-mesenchymal transition and self-renewal transcription factors, namely, Oct-4, Nanog, Sox2, ZEB1 and Snail. The effect of glycyrrhizic acid on the reduction of stem cell features was also observed in the primary dermal papilla cells directly obtained from human hair follicles. CONCLUSION: These results revealed a novel molecular mechanism of glycyrrhizic acid in regulation of dermal papilla cells and provided the evidence supporting the use of this compound in suppressing the growth of unwanted hair.


Subject(s)
Epidermal Cells , Glycyrrhizic Acid/pharmacology , Hair Follicle/cytology , Stem Cells/drug effects , Cell Differentiation , Cells, Cultured , Epidermis/drug effects , Epithelial-Mesenchymal Transition/drug effects , Glycogen Synthase Kinase 3/metabolism , Glycogen Synthase Kinase 3 beta , Hair Follicle/drug effects , Humans , Phenotype , Stem Cells/cytology , Transcription Factors/metabolism , Wnt Signaling Pathway/drug effects
20.
Article in English | MEDLINE | ID: mdl-26339272

ABSTRACT

As cancer stem cells (CSCs) contribute to malignancy, metastasis, and relapse of cancers, potential of compound in inhibition of CSCs has garnered most attention in the cancer research as well as drug development fields recently. Herein, we have demonstrated for the first time that gigantol, a pure compound isolated from Dendrobium draconis, dramatically suppressed stem-like phenotypes of human lung cancer cells. Gigantol at nontoxic concentrations significantly reduced anchorage-independent growth and survival of the cancer cells. Importantly, gigantol significantly reduced the ability of the cancer cells to form tumor spheroids, a critical hallmark of CSCs. Concomitantly, the treatment of the compound was shown to reduce well-known lung CSCs markers, including CD133 and ALDH1A1. Moreover, we revealed that gigantol decreased stemness in the cancer cells by suppressing the activation of protein kinase B (Akt) signal which in turn decreased the cellular levels of pluripotency and self-renewal factors Oct4 and Nanog. In conclusion, gigantol possesses CSCs suppressing activity which may facilitate the development of this compound for therapeutic approaches by targeting CSCs.

SELECTION OF CITATIONS
SEARCH DETAIL