Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 12 de 12
Filter
Add more filters










Publication year range
1.
Life (Basel) ; 13(11)2023 Nov 05.
Article in English | MEDLINE | ID: mdl-38004306

ABSTRACT

Hyperglycemia and inflammation are hallmarks of the prediabetes stage, which has the potential to develop into diabetes mellitus. In this stage, lifestyle changes and exercise are recommended and have been shown to be effective. However, there has been insufficient study investigating the impact of Ruesi Dadton (RD) exercise on prediabetes. Therefore, this study aimed to investigate the effect of RD exercise on biomarkers of glycemic level including fasting plasma glucose (FPG), the 2 h oral glucose tolerance test (OGTT), hemoglobin A1C (HbA1C), the biomarkers of inflammation C-reactive protein (CRP) and interleukin 6 (IL-6), and body mass index (BMI) on prediabetes during 12-week RD exercise. A total of 64 participants were randomly assigned into two groups, RD and control (CON), and were tested by measuring their glycemic levels to screen for prediabetes. The RD group was instructed to perform 10 postures of RD exercise in 60 min, three times a week. The CON group received standard lifestyle recommendations that were not pharmacologically managed. The results reveal that the RD group experienced a significant decrease in FPG, OGTT, HbA1C, and IL-6 (p < 0.01), and BMI and CRP (p < 0.05) compared to the CON group. In addition, the CON group had considerably higher glycemic levels, BMI and IL-6 levels (p < 0.01). Our study demonstrates that RD could decrease the biomarkers of glycemic level and inflammation during 12 weeks of RD exercise in prediabetes. These findings suggest that RD exercise is an effective approach for reducing systemic inflammation and controlling glycemic levels in prediabetic patients.

2.
Molecules ; 27(23)2022 Nov 22.
Article in English | MEDLINE | ID: mdl-36500213

ABSTRACT

Lung cancer metastasis is a multifaceted process that accounts for 90% of cancer deaths. According to several studies, the epithelial-mesenchymal transition (EMT) plays an essential role in lung cancer metastasis. Therefore, this study aimed to investigate the potential pharmacological effect of cycloartocarpin on the suppression of metastasis-related behaviors and EMT. An MTT assay was used to examine cell viability. Cell migration was determined using a wound healing assay. Anchorage-independent cell growth was also performed. Western blot analysis was used to identify the key signaling proteins involved in the regulation of EMT and migration. The results found that non-toxic concentrations of cycloartocarpin (10-20 µM) effectively suppressed cell migration and attenuated anchorage-independent growth in H292, A549, and H460 cells. Interestingly, these effects were consistent with the findings of Western blot analysis, which revealed that the level of phosphorylated focal adhesion kinase (p-FAK), phosphorylated ATP-dependent tyrosine kinase (p-AKT), and cell division cycle 42 (Cdc42) were significantly reduced, resulting in the inhibition of the EMT process, as evidenced by decreased N-cadherin, vimentin, and slug expression. Taken together, the results suggest that cycloartocarpin inhibits EMT by suppressing the FAK/AKT signaling pathway, which is involved in Cdc42 attenuation. Our findings demonstrated that cycloartocarpin has antimetastatic potential for further research and development in lung cancer therapy.


Subject(s)
Carcinoma, Non-Small-Cell Lung , Lung Neoplasms , Humans , Carcinoma, Non-Small-Cell Lung/metabolism , Epithelial-Mesenchymal Transition , Lung Neoplasms/metabolism , Cell Line, Tumor , Cell Movement , Signal Transduction
3.
Pharmaceuticals (Basel) ; 14(11)2021 Oct 26.
Article in English | MEDLINE | ID: mdl-34832867

ABSTRACT

Cancer stem cells (CSCs) are an important therapeutic target. The therapeutic agents targeting CSCs should lead to improved clinical outcomes. Here we have demonstrated the CSC-suppressing activity of pongol methyl ether (PME), a pure compound from Millettia erythrocalyx. METHODS: CSC-suppressing effects were evaluated by spheroid formation assay and detection of CSC markers. The related CSC cell signals were evaluated by Western blot, immunofluorescence and molecular docking analysis. Proteins affected by PME treatment were subjected to bioinformatic analysis. Protein-protein interaction (PPI) networks were constructed by the Search Tool for Interactions of Chemicals (STITCH). The Kyoto Encyclopedia of Genes and Genomes (KEGG) mapper were used to confirm the underlying pathways. RESULTS: PME (5-25 µM) significantly suppressed the ability of lung cancer cells to form colonies, grow in an anchorage-independent manner and generate tumour spheroids. PME at 25 µM significantly decreased the CSC markers (CD133 and ALDH1A1) and pluripotent transcription factors (Oct4 and Nanog). Akt, the key upstream signal of CSC control, was significantly decreased by the PME treatment. The molecular docking indicated that PME was bound to Akt-1 with a binding affinity of -9.2 kcal/mol greater than the Akt-1 inhibitor (reference compound; CQW). The STITCH network identified a total of 15 proteins interacted in PPI networks, and Akt-1 was identified as a central protein. The KEGG mapper indicated that the selected CSC markers were mostly involved in the 'signalling pathways regulating pluripotency of stem cells' pathway map and Akt, Oct4 and Nanog were the regulatory proteins in the dominant pathway. In addition, PME (10-25 µM) can suppress spheroid formation and reduce CSC-specific marker expression in patient-derived primary lung cancer cells. CONCLUSIONS: Our study revealed a novel pharmacological effect and the underlying mechanism of PME that can attenuate CSC phenotypes in lung cancer cells and may be developed for lung cancer therapy.

4.
Pharmaceutics ; 13(8)2021 Aug 10.
Article in English | MEDLINE | ID: mdl-34452194

ABSTRACT

Metal nanomaterials can enhance the efficacy of current cancer therapies. Here, we show that Ti0.8O2 nanosheets cause cytotoxicity in several lung cancer cells but not in normal cells. The nanosheet-treated cells showed certain apoptosis characteristics. Protein analysis further indicated the activation of the p53-dependent death mechanism. Transmission electron microscopy (TEM) and scanning electron microscopy (SEM) analyses revealed the cellular uptake of the nanosheets and the induction of cell morphological change. The nanosheets also exhibited a substantial apoptosis effect on drug-resistant metastatic primary lung cancer cells, and it was found that the potency of the nanosheets was dramatically higher than standard drugs. Ti0.8O2 nanosheets induce apoptosis through a molecular mechanism involving peroxynitrite (ONOO-) generation. As peroxynitrite is known to be a potent inducer of S-nitrosylation, we further found that the nanosheets mediated the S-nitrosylation of p53 at C182, resulting in higher protein-protein complex stability, and this was likely to induce the surrounding residues, located in the interface region, to bind more strongly to each other. Molecular dynamics analysis revealed that S-nitrosylation stabilized the p53 dimer with a ΔGbindresidue of <-1.5 kcal/mol. These results provide novel insight on the apoptosis induction effect of the nanosheets via a molecular mechanism involving S-nitrosylation of the p53 protein, emphasizing the mechanism of action of nanomaterials for cancer therapy.

5.
Antioxidants (Basel) ; 10(4)2021 Mar 29.
Article in English | MEDLINE | ID: mdl-33805467

ABSTRACT

Autophagic cell death (ACD) is an alternative death mechanism in resistant malignant cancer cells. In this study, we demonstrated how polyphenol stilbene compound PE5 exhibits potent ACD-promoting activity in lung cancer cells that may offer an opportunity for novel cancer treatment. Cell death caused by PE5 was found to be concomitant with dramatic autophagy induction, as indicated by acidic vesicle staining, autophagosome, and the LC3 conversion. We further confirmed that the main death induction caused by PE5 was via ACD, since the co-treatment with an autophagy inhibitor could reverse PE5-mediated cell death. Furthermore, the defined mechanism of action and upstream regulatory signals were identified using proteomic analysis. Time-dependent proteomic analysis showed that PE5 affected 2142 and 1996 proteins after 12 and 24 h of treatment, respectively. The crosstalk network comprising 128 proteins that control apoptosis and 25 proteins involved in autophagy was identified. Protein-protein interaction analysis further indicated that the induction of ACD was via AKT/mTOR and Bcl-2 suppression. Western blot analysis confirmed that the active forms of AKT, mTOR, and Bcl-2 were decreased in PE5-treated cells. Taken together, we demonstrated the novel mechanism of PE5 in shifting autophagy toward cell death induction by targeting AKT/mTOR and Bcl-2 suppression.

6.
Cancer Genomics Proteomics ; 18(3): 261-272, 2021.
Article in English | MEDLINE | ID: mdl-33893079

ABSTRACT

BACKGROUND/AIM: c-Met (mesenchymal-epithelial transition factor) facilitates cancer progression and is recognized as a promising drug target. The molecular target of gigantol from Dendrobium draconis in suppressing cancer metastasis is largely unknown. MATERIALS AND METHODS: Proteins affected by gigantol treatment were subjected to proteomic and bioinformatic analysis. Protein-Protein interaction (PPI) networks were constructed by the Search Tool for the Retrieval of Interacting Genes (STRING). The Kyoto Encyclopedia of Genes and Genomes (KEGG) database and hub gene were used to enrich the dominant pathways. Western blot analysis and immunofluorescence were used to validate the effect of gigantol on the target protein and signaling. RESULTS: Gigantol down-regulates 41 adhesion proteins and 39-migratory proteins, while it up-regulates 30 adhesion-related proteins and 22 proteins controlling cell migration. The key components of our constructed PPI network comprised 41 proteins of cell adhesion enriched in 40 nodes with 25 edges, 39 proteins of cell migration enriched in 39 nodes with 76 edges in down-regulated proteins, 30 proteins of cell adhesion enriched in 30 nodes with 21 edges, and 22 proteins of cell migration enriched in 22 nodes with 22 edges in up-regulated protein. c-Met was identified as a central protein of the PPI network in the largest degree. KEGG mapper further suggested that c-Met, PI3K, and AKT were the regulatory proteins affected by gigantol. To confirm, the effects of gigantol on c-Met, the p-PI3K, PI3K, p-AKT, and AKT proteins were investigated by western blotting and the results showed a consistent effect of gigantol in the suppression of the c-Met/PI3K/AKT signal. Next, immunofluorescence showed a dramatic decrease in c-Met, PI3K and AKT activation in response to gigantol. CONCLUSION: c-Met is an important target of gigantol treatment in lung cancer cells. Gigantol suppresses metastasis-related cell motility through decreasing c-Met resulting in PI3K/AKT signaling disruption.


Subject(s)
Bibenzyls/pharmacology , Guaiacol/analogs & derivatives , Lung Neoplasms/drug therapy , Proto-Oncogene Proteins c-met/metabolism , Guaiacol/pharmacology , Humans , Lung Neoplasms/metabolism , Lung Neoplasms/pathology , Molecular Targeted Therapy , Neoplasm Metastasis , Protein Interaction Maps/drug effects
7.
Anticancer Res ; 40(9): 4989-4999, 2020 Sep.
Article in English | MEDLINE | ID: mdl-32878787

ABSTRACT

BACKGROUND/AIM: Epithelial to mesenchymal transition (EMT) is a cellular process that facilitates cancer metastasis. Therefore, therapeutic approaches that target EMT have garnered increasing attention. The present study aimed to examine the in vitro effects of ephemeranthol A on cell death, migration, and EMT of lung cancer cells. MATERIALS AND METHODS: Ephemeranthol A was isolated from Dendrobium infundibulum. Non-small cell lung cancer cells H460 were treated with ephemeranthol A and apoptosis was evaluated by Hoechst 33342 staining. Anoikis resistance was determined by soft agar assay. Wound healing assay was performed to test the migration. The regulatory proteins of apoptosis and cell motility were determined by western blot. RESULTS: Treatment with ephemeranthol A resulted in a concentration-dependent cell apoptosis. At non-toxic concentrations, the compound could inhibit anchorage-independent growth of the cancer cells, as indicated by the decreased colony size and number. Ephemeranthol A also exhibited an inhibitory effect on migration. We further found that ephemeranthol A exerts its antimetastatic effects via inhibition of EMT, as indicated by the markedly decrease of N-cadherin, vimentin, and Slug. Furthermore, the compound suppressed the activation of focal adhesion kinase (FAK) and protein kinase B (Akt) proteins, which are key regulators of cell migration. As for the anticancer activity, ephemeranthol A induced apoptosis by decreasing Bcl-2 followed by the activation of caspase 3 and caspase 9. CONCLUSION: The pro-apoptotic and anti-migratory effects of ephemeranthol A on human lung cancer cells support its use for the development of novel anticancer therapies.


Subject(s)
Antineoplastic Agents/pharmacology , Epithelial-Mesenchymal Transition/drug effects , Focal Adhesion Kinase 1/metabolism , Lung Neoplasms/pathology , Phenanthrenes/pharmacology , Proto-Oncogene Proteins c-akt/metabolism , Signal Transduction/drug effects , Antineoplastic Agents/chemistry , Apoptosis/drug effects , Carcinoma, Non-Small-Cell Lung/drug therapy , Carcinoma, Non-Small-Cell Lung/metabolism , Carcinoma, Non-Small-Cell Lung/pathology , Cell Line, Tumor , Cell Movement/drug effects , Cell Proliferation/drug effects , Cell Survival/drug effects , Dendrobium/chemistry , Humans , Lung Neoplasms/drug therapy , Lung Neoplasms/metabolism , Molecular Structure , Phenanthrenes/chemistry , Phenanthrenes/therapeutic use
8.
Cancers (Basel) ; 12(4)2020 Apr 03.
Article in English | MEDLINE | ID: mdl-32260280

ABSTRACT

Myeloid cell leukemia 1 (Mcl-1) and B-cell lymphoma 2 (Bcl-2) proteins are promising targets for cancer therapy. Here, we investigated the structure-activity relationships (SARs) and performed molecular docking analysis of renieramycin T (RT) and its analogues and identified the critical functional groups of Mcl-1 targeting. RT have a potent anti-cancer activity against several lung cancer cells and drug-resistant primary cancer cells. RT mediated apoptosis through Mcl-1 suppression and it also reduced the level of Bcl-2 in primary cells. For SAR study, five analogues of RT were synthesized and tested for their anti-cancer and Mcl-1- and Bcl-2-targeting effects. Only two of them (TM-(-)-18 and TM-(-)-4a) exerted anti-cancer activities with the loss of Mcl-1 and partly reduced Bcl-2, while the other analogues had no such effects. Specific cyanide and benzene ring parts of RT's structure were identified to be critical for its Mcl-1-targeting activity. Computational molecular docking indicated that RT, TM-(-)-18, and TM-(-)-4a bound to Mcl-1 with high affinity, whereas TM-(-)-45, a compound with a benzene ring but no cyanide for comparison, showed the lowest binding affinity. As Mcl-1 helps cancer cells evading apoptosis, these data encourage further development of RT compounds as well as the design of novel drugs for treating Mcl-1-driven cancers.

9.
Anticancer Res ; 39(12): 6981-6988, 2019 Dec.
Article in English | MEDLINE | ID: mdl-31810970

ABSTRACT

BACKGROUND/AIM: Individualized proper chemotherapy using in vitro drug sensitivity testing has been proposed as a novel therapeutic modality and shown to have better efficacy than empiric chemotherapy. However, issues around establishing a patient-derived cell culture or xenograft, the timing of the testing obtained, and the validity of testing represent major limitations to translating the use of such a technique to clinical practice. PATIENTS AND METHODS: In this study, we assessed the feasibility of an in vitro drug sensitivity technique for testing malignant pleural effusion from advanced-stage non-small cell lung cancer. RESULTS: Our technique was able to produce a turnaround time for in vitro drug sensitivity testing of less than 1 week, with a success rate of more than 90% of cases. Correlated with the individual clinical outcome, using the area under the dose response curve (AUC) could define the level of in vitro drug sensitivity as: responsive (AUC>0.25), intermediate response (0.1≤AUC≤0.25), or resistance (AUC<0.1). CONCLUSION: Data obtained from this method of drug testing were correlated with the clinical outcome. The present drug sensitivity evaluation may benefit the development of individual precision chemotherapy.


Subject(s)
Antineoplastic Agents/pharmacology , Carcinoma, Non-Small-Cell Lung/drug therapy , Lung Neoplasms/drug therapy , Pleural Effusion, Malignant/drug therapy , Adult , Aged , Aged, 80 and over , Antineoplastic Agents/therapeutic use , Area Under Curve , Cell Proliferation/drug effects , Cell Survival/drug effects , Drug Screening Assays, Antitumor , Feasibility Studies , Female , Humans , Male , Middle Aged , Precision Medicine , Prospective Studies , Time Factors , Tumor Cells, Cultured
10.
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
11.
Anticancer Res ; 38(5): 2607-2618, 2018 05.
Article in English | MEDLINE | ID: mdl-29715080

ABSTRACT

Throughout the world, breast cancer is among the major causes of cancer-related death and is the most common cancer found in women. The development of cancer molecular knowledge has surpassed the novel concept of cancer biology and unraveled principle targets for anticancer drug developments and treatment strategies. Metastatic breast cancer cells acquire their aggressive features through several mechanisms, including augmentation of survival, proliferation, tumorigenicity, and motility-related cellular pathways. Clearly, natural product-derived compounds have since long been recognized as an important source for anticancer drugs, several of which have been shown to have promising anti-metastasis activities by suppressing key molecular features supporting such cell aggressiveness. This review provides the essential details of breast cancer, the molecular-based insights into metastasis, as well as the effects and mechanisms of potential compounds for breast cancer therapeutic approaches. As the abilities of cancer cells to invade and metastasize are addressed as the hallmarks of cancer, compounds possessing anti-metastatic effects, together with their defined molecular drug action could benefit the development of new drugs as well as treatment strategies.


Subject(s)
Antineoplastic Agents/therapeutic use , Breast Neoplasms/pathology , Neoplasm Metastasis/physiopathology , Angiogenesis Inhibitors/pharmacology , Angiogenesis Inhibitors/therapeutic use , Animals , Antineoplastic Agents/pharmacology , Antineoplastic Agents, Phytogenic/isolation & purification , Antineoplastic Agents, Phytogenic/therapeutic use , Biological Products/therapeutic use , Cell Adhesion , Drug Discovery , Drug Screening Assays, Antitumor , Epithelial-Mesenchymal Transition , Extracellular Matrix/pathology , Female , Humans , Intercellular Signaling Peptides and Proteins/physiology , Molecular Targeted Therapy , Neoplasm Invasiveness , Neoplasm Metastasis/drug therapy , Neoplasm Proteins/physiology , Neovascularization, Pathologic/drug therapy , Neovascularization, Pathologic/physiopathology
12.
Anticancer Res ; 37(11): 6311-6319, 2017 11.
Article in English | MEDLINE | ID: mdl-29061814

ABSTRACT

BACKGROUND/AIM: Metastasis in lung cancer is a major cause of high mortality. Metastasis depends on the potential of cancer cells to migrate and invade. Here we demonstrated the anti-migration and invasion activities of the compound cycloartobiloxanthone from Artocarpus gomezianus Wall. ex Tréc. (Moraceae). MATERIALS AND METHODS: The effect of the compound on viability of human lung cancer H460 cells was investigated by 3-[4,5-dimethylthiazol-2-yl]-2,5diphenyl tetrazoliumbromide (MTT) assay. Migration and invasion assays were performed. Filopodia formation was determined by phalloidin-rhodamine staining. The hallmark signaling proteins in regulation of epithelial to mesenchymal transition (EMT), migration, and integrin α5, αV, ß1 and ß3 were determined by western blot analysis. RESULTS: Cycloartobiloxanthone at concentrations lower than 10 µM has no cytotoxic effects. Regarding cell motility, cycloartobiloxanthone at 5-10 µM and 1-10 µM exhibited anti-migration and anti-invasion activities, respectively. Filopodia were found to be significantly reduced in cycloartobiloxanthone-treated cells. These effects correlated with the results from western blot analysis showing that the phosphorylation of focal adhesion kinase on Try397 (p-FAK (Try397)), and cell division cycle 42 (CDC42) were significantly reduced. Cycloartobiloxanthone significantly suppressed migratory integrins including integrin α5, αV, and ß3, while had no significant effect on integrin ß1. Besides, the compound suppressed epithelial to mesenchymal transition in lung cancer H460 cells indicated by the change in cell morphology form fibroblast-like to epithelial morphology with up-regulation of E-cadherin. CONCLUSION: Cycloartobiloxanthone possesses anti-migration and anti-invasion properties by suppressing several migratory-regulated mechanisms including suppressing migratory FAK and CDC42 signal, reduced filopodia of migrating cells, decreasing integrin α5, αv and ß3, and inhibiting EMT. Our findings demonstrated the potentials of cycloartobiloxanthone for further studies and developments.


Subject(s)
Antineoplastic Agents/pharmacology , Carcinoma, Non-Small-Cell Lung/metabolism , Cell Cycle Proteins/metabolism , Flavonoids/pharmacology , Focal Adhesion Kinase 1/metabolism , Lung Neoplasms/metabolism , Carcinoma, Non-Small-Cell Lung/drug therapy , Cell Line, Tumor , Cell Movement/drug effects , Cell Survival/drug effects , Epithelial-Mesenchymal Transition/drug effects , Gene Expression Regulation, Neoplastic/drug effects , Humans , Lung Neoplasms/drug therapy , Neoplasm Invasiveness , Neoplasm Metastasis , Phosphorylation/drug effects
SELECTION OF CITATIONS
SEARCH DETAIL
...