Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 9 de 9
Filter
Add more filters










Database
Language
Publication year range
1.
Front Pharmacol ; 15: 1382787, 2024.
Article in English | MEDLINE | ID: mdl-38659592

ABSTRACT

Background: Prostate cancer and non-small cell lung cancer (NSCLC) present significant challenges in the development of effective therapeutic strategies. Hormone therapies for prostate cancer target androgen receptors and prostate-specific antigen markers. However, treatment options for prostatic small-cell neuroendocrine carcinoma are limited. NSCLC, on the other hand, is primarily treated with epidermal growth factor receptor (EGFR) tyrosine kinase inhibitors but exhibits resistance. This study explored a novel therapeutic approach by investigating the potential anticancer properties of vitekwangin B, a natural compound derived from Vitex trifolia. Methods: Vitekwangin B was chromatographically isolated from the fruits of V. trifolia. ANO1 protein levels in prostate cancer and NSCLC cells were verified and evaluated again after vitekwangin B treatment. Results: Vitekwangin B did not inhibit anoctamin1 (ANO1) channel function but significantly reduced ANO1 protein levels. These results demonstrate that vitekwangin B effectively inhibited cancer cell viability and induced apoptosis in prostate cancer and NSCLC cells. Moreover, it exhibited minimal toxicity to liver cells and did not affect hERG channel activity, making it a promising candidate for further development as an anticancer drug. Conclusion: Vitekwangin B may offer a new direction for cancer therapy by targeting ANO1 protein, potentially improving treatment outcomes in patients with prostate cancer and NSCLC. Further research is needed to explore its full potential and overcome existing drug resistance challenges.

2.
Chem Biodivers ; 20(11): e202301296, 2023 Nov.
Article in English | MEDLINE | ID: mdl-37842907

ABSTRACT

Vitex trifolia L. is a medicinal plant and widely distributed in the northern mountainous areas of Vietnam. Phytochemical study on the fruits of this plant led to the isolation of nine iridoid derivatives (1-9) including three undescribed compounds (1-3). Their structures were elucidated to be 3''-hydroxyscrophuloside A1 (1), 3''-hydroxycallicoside D (2), 2'-p-hydroxybenzoylaucubin (3), 6'-p-hydroxybenzoylmussaenosidic acid (4), nishindaside (5), agnuside (6), 10-O-vanilloylaucubin (7), 6'-O-p-hydroxybenzoyl-gardoside (8), and buddlejoside B (9) based on extensive analyses of HR-ESI-MS, 1D and 2D NMR spectra. Compounds 1, 2, 4, and 8 significantly posessed anti-barterial activity against Enterococcus faecalis, Staphylococcus aureus, Escherichia coli, Pseudomonas aeruginosa strains with MIC values in range of 16-64 µg/mL. At concentration of 20 µM, compounds 1-9 did not show cytotoxic effects against human lung cancer cells (PC9).


Subject(s)
Anti-Infective Agents , Antineoplastic Agents , Vitex , Humans , Iridoids/chemistry , Vitex/chemistry , Fruit/chemistry , Anti-Infective Agents/pharmacology , Anti-Infective Agents/analysis , Plant Extracts/analysis
3.
Front Pharmacol ; 14: 1163970, 2023.
Article in English | MEDLINE | ID: mdl-37274097

ABSTRACT

Anoctamin 1 (ANO1), a drug target for various cancers, including prostate and oral cancers, is an intracellular calcium-activated chloride ion channel that plays various physiopathological roles, especially in the induction of cancer growth and metastasis. In this study, we tested a novel compound isolated from Schisandra sphenanthera, known as schisandrathera D, for its inhibitory effect on ANO1. Schisandrathera D dose-dependently suppressed the ANO1 activation-mediated decrease in fluorescence of yellow fluorescent protein; however, it did not affect the adenosine triphosphate-induced increase in the intracellular calcium concentration or forskolin-induced cystic fibrosis transmembrane conductance regulator activity. Specifically, schisandrathera D gradually decreased the levels of ANO1 protein and significantly reduced the cell viability in ANO1-expressing cells when compared to those in ANO1-knockout cells. These effects could be attributed to the fact that schisandrathera D displayed better binding capacity to ANO1 protein than the previously known ANO1 inhibitor, Ani9. Finally, schisandrathera D increased the levels of caspase-3 and cleaved poly (ADP-ribose) polymerase 1, thereby indicating that its anticancer effect is mediated through apoptosis. Thus, this study highlights that schisandrathera D, which reduces ANO1 protein levels, has apoptosis-mediated anticancer effects in prostate and oral cancers, and thus, can be further developed into an anticancer agent.

4.
Nat Prod Res ; : 1-7, 2022 Oct 14.
Article in English | MEDLINE | ID: mdl-36239487

ABSTRACT

Using combined chromatographic methods, two new triterpenoid glycosides, bacopasaponin K (1) and bacopasaponin L (2), along with eight known compounds, bacopaside IV (3), bacopaside VII (4), bacopasaponin E (5), bacoside A3 (6), bacopasaponin F (7), bacopasaponin C (8), bacopaside I (9), and bacopaside II (10) were isolated from the methanol extract of the Bacopa monnieri. Their structures were elucidated by 1D-, 2D-NMR spectroscopic analysis, HR-ESI-MS and comparing with the NMR data reported in the literature. All these compounds were evaluated for their cytotoxic activity using the cell counting kit-8 (CCK-8) assay. Compounds 4, 6, 8, and 10 exhibited potential cytotoxic effects against human lung cancer cells (PC9) and human colon cancer cells (SW620).

5.
Biomed Pharmacother ; 153: 113373, 2022 Sep.
Article in English | MEDLINE | ID: mdl-35785700

ABSTRACT

Anoctamin 1 (ANO1) is a calcium-activated chloride channel found in various cell types and is overexpressed in non-small cell lung cancer (NSCLC), a major cause of cancer-related mortality. With the rising interest in development of druggable compounds for NSCLC, there has been a corresponding rise in interest in ANO1, a novel drug target for NSCLC. However, as ANO1 inhibitors that have been discovered simultaneously exhibit both the functions of an inhibition of ANO1 channel as well as a reduction of ANO1 protein levels, it is unclear which of the two functions directly causes the anticancer effect. In this study, verteporfin, a chemical compound that reduces ANO1 protein levels was identified through high-throughput screening. Verteporfin did not inhibit ANO1-induced chloride secretion but reduced ANO1 protein levels in a dose-dependent manner with an IC50 value of ~300 nM. Moreover, verteporfin inhibited neither P2Y receptor-induced intracellular Ca2+ mobilization nor cystic fibrosis transmembrane conductance regulator (CFTR) channel activity, and molecular docking studies revealed that verteporfin bound to specific sites of ANO1 protein. Confirming that verteporfin reduces ANO1 protein levels, we then investigated the molecular mechanisms involved in its effect on NSCLC cells. Interestingly, verteporfin decreased ANO1 protein levels, the EGFR-STAT3 pathway as well as ANO1 mRNA expression. Verteporfin reduced the viability of ANO1-expressing cells (PC9, and gefitinib-resistant PC9) and induced apoptosis by increasing caspase-3 activity and PARP-1 cleavage. However, it did not affect hERG channel activity. These results show that the anticancer mechanism of verteporfin is caused via the down-regulation of ANO1.


Subject(s)
Anoctamin-1 , Carcinoma, Non-Small-Cell Lung , Lung Neoplasms , Neoplasm Proteins , Verteporfin , Anoctamin-1/genetics , Anoctamin-1/metabolism , Calcium/metabolism , Carcinoma, Non-Small-Cell Lung/drug therapy , Chloride Channels/metabolism , Down-Regulation , Humans , Lung Neoplasms/drug therapy , Molecular Docking Simulation , Neoplasm Proteins/genetics , Neoplasm Proteins/metabolism , Verteporfin/pharmacology
6.
Int J Mol Sci ; 22(13)2021 Jul 01.
Article in English | MEDLINE | ID: mdl-34281152

ABSTRACT

Non-small cell lung cancer (NSCLC) is one of the leading causes of cancer-related mortality; thus, therapeutic targets continue to be developed. Anoctamin1 (ANO1), a novel drug target considered for the treatment of NSCLC, is a Ca2+-activated chloride channel (CaCC) overexpressed in various carcinomas. It plays an important role in the development of cancer; however, the role of ANO1 in NSCLC is unclear. In this study, diethylstilbestrol (DES) was identified as a selective ANO1 inhibitor using high-throughput screening. We found that DES inhibited yellow fluorescent protein (YFP) fluorescence reduction caused by ANO1 activation but did not inhibit cystic fibrosis transmembrane conductance regulator channel activity or P2Y activation-related cytosolic Ca2+ levels. Additionally, electrophysiological analyses showed that DES significantly reduced ANO1 channel activity, but it more potently reduced ANO1 protein levels. DES also inhibited the viability and migration of PC9 cells via the reduction in ANO1, phospho-ERK1/2, and phospho-EGFR levels. Moreover, DES induced apoptosis by increasing caspase-3 activity and PARP-1 cleavage in PC9 cells, but it did not affect the viability of hepatocytes. These results suggest that ANO1 is a crucial target in the treatment of NSCLC, and DES may be developed as a potential anti-NSCLC therapeutic agent.


Subject(s)
Anoctamin-1/antagonists & inhibitors , Carcinoma, Non-Small-Cell Lung/drug therapy , Diethylstilbestrol/pharmacology , Neoplasm Proteins/antagonists & inhibitors , Anoctamin-1/metabolism , Apoptosis/drug effects , Carcinoma, Non-Small-Cell Lung/metabolism , Cell Line, Tumor , Cell Proliferation/drug effects , Cystic Fibrosis Transmembrane Conductance Regulator/genetics , Cystic Fibrosis Transmembrane Conductance Regulator/metabolism , Diethylstilbestrol/metabolism , Humans , Lung Neoplasms , Neoplasm Proteins/metabolism , Signal Transduction
7.
Cancer Res ; 81(13): 3539-3553, 2021 07 01.
Article in English | MEDLINE | ID: mdl-33910929

ABSTRACT

Extracellular vesicles (EV) in the tumor microenvironment have emerged as crucial mediators that promote proliferation, metastasis, and chemoresistance. However, the role of circulating small EVs (csEV) in cancer progression remains poorly understood. In this study, we report that csEV facilitate cancer progression and determine its molecular mechanism. csEVs strongly promoted the migration of cancer cells via interaction with phosphatidylserine of csEVs. Among the three TAM receptors, TYRO3, AXL, and MerTK, TYRO3 mainly interacted with csEVs. csEV-mediated TYRO3 activation promoted migration and metastasis via the epithelial-mesenchymal transition and stimulation of RhoA in invasive cancer cells. Additionally, csEV-TYRO3 interaction induced YAP activation, which led to increased cell proliferation and chemoresistance. Combination treatment with gefitinib and KRCT-6j, a selective TYRO3 inhibitor, significantly reduced tumor volume in xenografts implanted with gefitinib-resistant non-small cell lung cancer cells. The results of this study show that TYRO3 activation by csEVs facilitates cancer cell migration and chemoresistance by activation of RhoA or YAP, indicating that the csEV/TYRO3 interaction may serve as a potential therapeutic target for aggressive cancers in the clinic. SIGNIFICANCE: These findings demonstrate that circulating extracellular vesicles are a novel driver in migration and survival of aggressive cancer cells via TYRO3 activation. GRAPHICAL ABSTRACT: http://cancerres.aacrjournals.org/content/canres/81/13/3539/F1.large.jpg.


Subject(s)
Drug Resistance, Neoplasm , Extracellular Vesicles/metabolism , Gefitinib/pharmacology , Liver Neoplasms/secondary , Neoplasms/pathology , Receptor Protein-Tyrosine Kinases/metabolism , Splenic Neoplasms/secondary , Animals , Apoptosis , Biomarkers, Tumor/genetics , Biomarkers, Tumor/metabolism , Cell Proliferation , Gene Expression Regulation, Neoplastic , Humans , Liver Neoplasms/drug therapy , Liver Neoplasms/genetics , Liver Neoplasms/metabolism , Male , Mice , Mice, Inbred BALB C , Mice, Nude , Neoplasms/drug therapy , Neoplasms/genetics , Neoplasms/metabolism , Receptor Protein-Tyrosine Kinases/genetics , Splenic Neoplasms/drug therapy , Splenic Neoplasms/genetics , Splenic Neoplasms/metabolism , Tumor Cells, Cultured , Xenograft Model Antitumor Assays
8.
Mol Cancer Ther ; 17(4): 825-837, 2018 04.
Article in English | MEDLINE | ID: mdl-29437878

ABSTRACT

The most common therapy for estrogen receptor-positive breast cancer is antihormone therapy, such as tamoxifen. However, acquisition of resistance to tamoxifen in one third of patients presents a serious clinical problem. Polo-like kinase 1 (Plk1) is a key oncogenic regulator of completion of G2-M phase of the cell cycle. We assessed Plk1 expression in five chemoresistant cancer cell types and found that Plk1 and its downstream phosphatase Cdc25c were selectively overexpressed in tamoxifen-resistant MCF-7 (TAMR-MCF-7) breast cancer cells. Real-time monitoring of cell proliferation also showed that TAMR-MCF-7 cells were more sensitive to inhibition of cell proliferation by the ATP-competitive Plk1 inhibitor BI2536 than were the parent MCF-7 cells. Moreover, BI2536 suppressed expression of epithelial-mesenchymal transition marker proteins and 3D spheroid formation in TAMR-MCF-7 cells. Using TAMR-MCF-7 cell-implanted xenograft and spleen-liver metastasis models, we showed that BI2536 inhibited tumor growth and metastasis in vivo Our results suggest that Plk1 could be a novel target for the treatment of tamoxifen-resistant breast cancer. Mol Cancer Ther; 17(4); 825-37. ©2018 AACR.


Subject(s)
Biomarkers, Tumor/metabolism , Breast Neoplasms/pathology , Cell Cycle Proteins/metabolism , Drug Resistance, Neoplasm , Liver Neoplasms/secondary , Protein Serine-Threonine Kinases/metabolism , Proto-Oncogene Proteins/metabolism , Splenic Neoplasms/secondary , Tamoxifen/pharmacology , Animals , Antineoplastic Agents, Hormonal , Apoptosis , Biomarkers, Tumor/genetics , Breast Neoplasms/drug therapy , Breast Neoplasms/metabolism , Cell Cycle , Cell Cycle Proteins/antagonists & inhibitors , Cell Cycle Proteins/genetics , Cell Movement , Cell Proliferation , Female , Gene Expression Regulation, Neoplastic , Humans , Liver Neoplasms/drug therapy , Liver Neoplasms/metabolism , Mice , Mice, Inbred BALB C , Mice, Nude , Protein Serine-Threonine Kinases/antagonists & inhibitors , Protein Serine-Threonine Kinases/genetics , Proto-Oncogene Proteins/antagonists & inhibitors , Proto-Oncogene Proteins/genetics , Pteridines/pharmacology , Splenic Neoplasms/drug therapy , Splenic Neoplasms/metabolism , Tumor Cells, Cultured , Xenograft Model Antitumor Assays , Polo-Like Kinase 1
9.
Cancer Lett ; 390: 115-125, 2017 04 01.
Article in English | MEDLINE | ID: mdl-28108315

ABSTRACT

We previously demonstrated that tamoxifen (TAM)-resistant human breast cancer (TAMR-MCF-7) cells showed increased expression of mesenchymal marker proteins compared to the parent MCF-7 cells. Notch is functionally important in the promotion of epithelial-mesenchymal transition (EMT) during both development and tumor progression. Notch1 and Notch4 have been reported as prognostic markers in human breast cancer. Here, we indicated that Notch4, but not Notch1, plays a critical role in the regulation of EMT signaling in TAMR-MCF-7 cells. Notch4 suppression by either Notch inhibitors or Notch4 siRNA attenuated EMT signaling. Tyrosine-phosphorylated STAT3 protein is known as a crucial signaling molecule in the regulation of tumorigenesis and metastasis. We found that TAMR-MCF-7 cells exhibited constitutive STAT3 phosphorylation, and Notch inhibition reduced the level of activated STAT3 in TAMR-MCF-7 cells. An intrasplenic injection model of liver metastases was performed using TAMR-MCF-7 cells. Mice injected with N-[N-(3,5-Difluorophenacetyl)-l-alanyl]-S-phenylglycine t-butyl ester (DAPT, 10 mg/kg) formed smaller splenic tumors and showed a reduced micrometastatic tumor burden in their livers compared with the control group treated with vehicle. To conclude, Notch4 could be a potential target to prevent metastasis in TAM-resistant breast cancer.


Subject(s)
Breast Neoplasms/physiopathology , Epithelial-Mesenchymal Transition/physiology , Proto-Oncogene Proteins/metabolism , Receptors, Notch/metabolism , STAT3 Transcription Factor/metabolism , Signal Transduction/physiology , Tamoxifen/therapeutic use , Animals , Antineoplastic Agents, Hormonal/pharmacology , Antineoplastic Agents, Hormonal/therapeutic use , Breast Neoplasms/drug therapy , Cell Line, Tumor , Cell Proliferation , Drug Resistance, Neoplasm/genetics , Epithelial-Mesenchymal Transition/drug effects , Female , Humans , Immunoblotting , Mice, Nude , Polymerase Chain Reaction , Protein Binding , Proto-Oncogene Proteins/antagonists & inhibitors , Receptor, Notch4 , Receptors, Notch/antagonists & inhibitors , Signal Transduction/drug effects , Tamoxifen/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL
...