Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 11 de 11
Filter
Add more filters










Publication year range
1.
Nutrients ; 14(21)2022 Oct 28.
Article in English | MEDLINE | ID: mdl-36364815

ABSTRACT

Colorectal cancer (CRC) is one of the diseases with the highest rates of prevalence and mortality despite therapeutic methods in the world. In particular, there are not enough methods to treat metastasis of CRC cells to distant organs. Cannabis sativa Linne (C. sativa) is a popular medicinal plant used by humans to treat many diseases. Recently, extracts of C. sativa have shown diverse pharmacological effects as a result of choosing different extraction methods. In this study, we performed experiments to confirm the inhibitory effect and related mechanisms of supercritical extract of C. sativa on metastatic CRC cells. The effect of SEC on the viability of CRC cell lines, CT26 and HCT116, was determined using CCK reagent. Flow cytometry was performed to confirm whether SEC can promote cell cycle arrest and apoptosis. Additionally, SEC reduced proliferation of CT26 and HCT116 cells without causing toxicity to normal colon cell line CCD-18Co cells. SEC treatment reduced colony formation in both CRC cell lines, promoted G0/G1 phase arrest and apoptosis in CT26 and HCT116 cells through AMPK activation and MAPKs such as ERK, JNK, and p38 inactivation. Moreover, oral administration of SEC decreased pulmonary metastasis of CT26 cells. Our research demonstrates the inhibitory effect of SEC on CRC cell proliferation and metastasis. Thus, SEC might have therapeutic potential for CRC treatment.


Subject(s)
Cannabis , Colorectal Neoplasms , Lung Neoplasms , Humans , AMP-Activated Protein Kinases , Cell Line, Tumor , Colorectal Neoplasms/pathology , Cell Cycle Checkpoints , Apoptosis , Lung Neoplasms/pathology , Cell Proliferation
2.
Phytomedicine ; 96: 153809, 2022 Feb.
Article in English | MEDLINE | ID: mdl-34782203

ABSTRACT

BACKGROUND: Despite the rising 5-year survival rate of colorectal cancer (CRC) patients, the survival rate decreases as the stage progress, and a low survival rate is highly associated with metastasis. PURPOSE: The purpose of our study is to investigate the effect of dehydroevodiamine (DHE) on the lung metastasis of CRC and the proliferation of CRC cells. STUDY DESIGN: Cell death was confirmed after DHE treatment on several CRC cell lines. The mechanism of cell cytotoxicity was found using flow cytometry. After that, the expression of the proteins or mRNAs related to the cell cytotoxicity was confirmed. Also, anti-metastatic ability of DHE in CRC cells was measured by checking the expression of Epithelial to Mesenchymal Transition (EMT) markers. Lung metastasis mouse model was established, and DHE was administered orally for 14 days. RESULTS: DHE suppressed the viability of HCT116, CT26, SW480, and LoVo cells. DHE treatment led to G2/M arrest via a reduction of cyclin B1/CDK1 and caspase-dependent apoptosis. It also induced autophagy by regulating LC3-II and beclin-1 expression. Additionally, migration and invasion of CRC cells were decreased by DHE through regulation of the expression of EMT markers. Oral administration of DHE could inhibit the lung metastasis of CT26 cells in an in vivo model. CONCLUSION: Our study demonstrated that DHE has a potential therapeutic effect on colorectal cancer metastasis.


Subject(s)
Colorectal Neoplasms , Lung Neoplasms , Alkaloids , Animals , Apoptosis , Cell Line, Tumor , Cell Movement , Cell Proliferation , Colorectal Neoplasms/drug therapy , Epithelial-Mesenchymal Transition , G2 Phase Cell Cycle Checkpoints , Gene Expression Regulation, Neoplastic , Humans , Lung Neoplasms/drug therapy , Mice , Neoplasm Metastasis
3.
Am J Chin Med ; 49(6): 1535-1555, 2021.
Article in English | MEDLINE | ID: mdl-34247563

ABSTRACT

Colorectal cancer (CRC) is the second most common cause of cancer death in the world, and metastatic CRC is a major cause of cancer death. Gallotannin (GT), a polyphenolic compound, has shown various biological effects such as anti-oxidant, anti-inflammatory, antimicrobial, and antitumor effects. However, the effects of GT on metastatic CRC cells are not completely understood. This study aimed to investigate the anti-metastatic effect of GT and the underlying mechanisms on metastatic CRC cells. Oral administration of GT suppressed the lung metastasis of metastatic CRC cells in the experimental mouse model. GT decreased the viability of metastatic CRC cell lines, including CT26, HCT116, and SW620, by inducing apoptosis through the activation of extrinsic and intrinsic pathways, cell cycle arrest through inactivation of CDK2/cyclin A complex, and autophagic cell death through up-regulation of LC3B and p62 levels. GT regulated PI3K/AKT/mTOR and AMPK signaling pathways, which are critical for the development and maintenance of cancer. Additionally, non-cytotoxic concentrations of GT can suppress migration and invasion of CRC cells by inhibiting the expression and activity of matrix metalloproteinase (MMP)-2 and MMP-9 and epithelial-mesenchymal transition by downregulating the expression of mesenchymal markers including snail, twist, and vimentin. In conclusion, GT prevented colorectal lung metastasis by reducing survival and inhibiting the metastatic phenotypes of CRC cells.


Subject(s)
Apoptosis/drug effects , Autophagy/drug effects , Cell Cycle Checkpoints/drug effects , Colorectal Neoplasms/drug therapy , Hydrolyzable Tannins/pharmacology , Lung Neoplasms/drug therapy , Animals , Cell Line, Tumor , Colorectal Neoplasms/pathology , Hydrolyzable Tannins/chemistry , Lung Neoplasms/secondary , Mice , Molecular Structure
4.
Foods ; 9(11)2020 Nov 02.
Article in English | MEDLINE | ID: mdl-33147777

ABSTRACT

Rubus coreanus Miquel (R. coreanus) is a unripen fruit of black raspberry native to eastern Asia. It is used as traditional oriental medicine and supplementary foods for centuries. Previous studies have shown that the R. coreanus extract (RCE) and its main constitute ellagic acid possess diverse biological activities. However, the effects of RCE on antitumor immunity and T cell function were not fully understood. The present study describes the anti-tumor effect of RCE in humanized PD-1 mice by blocking PD-1/PD-L1 interaction. Competitive enzyme-linked immunosorbent assay (ELISA) and pull down assay were performed to elucidate the binding properties of RCE in vitro. Cellular PD-1/PD-L1 blockade activities were measured by T cell receptor (TCR)-induced nuclear factor of activated T cells-luciferase activity in co-cultured cell models with PD-1/NFAT Jurkat and PD-L1/aAPC CHO-K1 cells. The in vivo efficacy of RCE was confirmed in humanized PD-1 mice bearing MC38 colorectal tumor. RCE and ellagic acid dose-dependently block the binding of PD-1 to PD-L1. Moreover, oral administration of RCE showed the potent anti-tumor activity similar to anti-PD-1 antibody. The present study suggests that RCE possesses potent anti-tumor effect via PD-1/PD-L1 blockade, and ellagic acid is the main compound in RCE. Thus, we provide new aspects of RCE as an immunotherapeutic agent.

5.
Nutrients ; 12(10)2020 Oct 19.
Article in English | MEDLINE | ID: mdl-33086629

ABSTRACT

BACKGROUND: Cachexia induced by cancer is a systemic wasting syndrome and it accompanies continuous body weight loss with the exhaustion of skeletal muscle and adipose tissue. Cancer cachexia is not only a problem in itself, but it also reduces the effectiveness of treatments and deteriorates quality of life. However, effective treatments have not been found yet. Although Arctii Fructus (AF) has been studied about several pharmacological effects, there were no reports on its use in cancer cachexia. METHODS: To induce cancer cachexia in mice, we inoculated CT-26 cells to BALB/c mice through subcutaneous injection and intraperitoneal injection. To mimic cancer cachexia in vitro, we used conditioned media (CM), which was CT-26 colon cancer cells cultured medium. RESULTS: In in vivo experiments, AF suppressed expression of interleukin (IL)-6 and atrophy of skeletal muscle and adipose tissue. As a result, the administration of AF decreased mortality by preventing weight loss. In adipose tissue, AF decreased expression of uncoupling protein 1 (UCP1) by restoring AMP-activated protein kinase (AMPK) activation. In in vitro model, CM increased muscle degradation factors and decreased adipocytes differentiation factors. However, these tendencies were ameliorated by AF treatment in C2C12 myoblasts and 3T3-L1 cells. CONCLUSION: Taken together, our study demonstrated that AF could be a therapeutic supplement for patients suffering from cancer cachexia.


Subject(s)
Adipose Tissue/pathology , Arctium/chemistry , Cachexia/drug therapy , Muscle, Skeletal/pathology , Phytotherapy , Plant Extracts/pharmacology , Plant Extracts/therapeutic use , Weight Loss/drug effects , 3T3-L1 Cells , AMP-Activated Protein Kinases/metabolism , Animals , Atrophy/prevention & control , Cachexia/etiology , Cachexia/genetics , Gene Expression/drug effects , Interleukin-6/genetics , Interleukin-6/metabolism , Male , Mice , Mice, Inbred BALB C , Neoplasms/complications , Plant Extracts/isolation & purification , Tumor Cells, Cultured , Uncoupling Protein 1/genetics , Uncoupling Protein 1/metabolism
6.
Molecules ; 25(9)2020 Apr 27.
Article in English | MEDLINE | ID: mdl-32349276

ABSTRACT

Pancreatic cancer (PC) is one of the most severe cancers, and its incidence and mortality rates have steadily increased in the past decade. In this study, we demonstrate the effect of Angelica gigas Nakai extract on pancreatic ductal adenocarcinoma cells. We prepared A. gigas Nakai ethanol extract (AGE) using roots of A. gigas Nakai and detected its active compound decursin from AGE by ultra-performance liquid chromatography analysis. AGE and decursin significantly decreased viability and colony formation of PANC-1 and MIA PaCa-2 cells. AGE and decursin induced G0/G1 phase arrest through downregulation of cyclin D1 and cyclin-dependent kinase 4 (CDK4). Caspase-3-dependent apoptosis of PANC-1 cells was promoted by AGE and decursin. Additionally, nontoxic concentrations of AGE and decursin treatment could suppress matrix metalloproteinase (MMP)-2 and MMP-9 expression and activity by inhibiting p38 phosphorylation. Taken together, this study demonstrates that AGE and decursin have potential properties to be considered in PC treatment.


Subject(s)
Angelica/chemistry , Antineoplastic Agents/pharmacology , Benzopyrans/pharmacology , Butyrates/pharmacology , Pancreatic Neoplasms/drug therapy , Plant Extracts/pharmacology , Plant Roots/chemistry , Apoptosis/drug effects , Benzopyrans/chemistry , Butyrates/chemistry , Caspase 3/metabolism , Cell Line, Tumor , Cell Proliferation/drug effects , Chromatography, High Pressure Liquid , Cyclin D1/metabolism , Cyclin-Dependent Kinase 4/metabolism , G1 Phase Cell Cycle Checkpoints/drug effects , Humans , Matrix Metalloproteinase 2/metabolism , Matrix Metalloproteinase 9/metabolism , Pancreatic Neoplasms/metabolism , Phosphorylation , Plant Extracts/analysis , p38 Mitogen-Activated Protein Kinases/metabolism
7.
Phytomedicine ; 68: 153147, 2020 Mar.
Article in English | MEDLINE | ID: mdl-32028184

ABSTRACT

BACKGROUND: Gomisin A (G.A), a lignan compound extracted from the fruits of Schisandra chinensis, is known to exert anti-tumor effects on hepatocarcinoma and colorectal cancer cells. Suppression of proliferation and metastatic abilities of cancer cells are some effective cancer treatment methods. PURPOSE: The objective of this study is to investigate the effects of G.A on metastatic melanoma, and the mechanism by which it affects metastatic melanoma. STUDY DESIGN: The anti-proliferative and anti-metastatic effects of G.A were observed in in vitro and in vivo. METHODS: WST assay and flow cytometry were conducted to investigate the effect of G.A on proliferation, cell cycle arrest, and apoptosis in metastatic melanoma cell lines. Migration and invasion abilities of G.A-treated melanoma cells were observed by wound healing and invasion assays. RESULTS: G.A (25-100 µM) decreased the viability of melanoma cells by inducing cell cycle arrest and apoptosis. These anti-proliferative effects of G.A were found to be mediated by AMPK, ERK, and JNK activation. G.A (5-20 µM) decreased the migration and invasion of melanoma cells by suppressing epithelial-mesenchymal transition (EMT). Consequently, G.A (2-50 mg/kg) inhibited lung metastasis by suppressing EMT and inducing cell cycle arrest and apoptosis in melanoma cells. CONCLUSION: These results conclude that G.A has the potential to reduce metastatic melanoma through its anti-proliferative and anti-metastatic effects.


Subject(s)
Antineoplastic Agents, Phytogenic/pharmacology , Cyclooctanes/pharmacology , Dioxoles/pharmacology , Lignans/pharmacology , Melanoma/drug therapy , Melanoma/pathology , AMP-Activated Protein Kinases/antagonists & inhibitors , AMP-Activated Protein Kinases/metabolism , Animals , Apoptosis/drug effects , Cell Cycle Checkpoints/drug effects , Cell Line, Tumor , Cell Movement/drug effects , Cell Proliferation/drug effects , Cell Survival/drug effects , Epithelial-Mesenchymal Transition/drug effects , Extracellular Signal-Regulated MAP Kinases/metabolism , Humans , Lung Neoplasms/drug therapy , Lung Neoplasms/secondary , MAP Kinase Kinase 4/metabolism , Melanoma/metabolism , Mice, Inbred C57BL , Xenograft Model Antitumor Assays
8.
Phytomedicine ; 62: 152952, 2019 Sep.
Article in English | MEDLINE | ID: mdl-31132754

ABSTRACT

BACKGROUND: Although rubrofusarin-6-ß-gentiobioside (RFG), which is a component of Cassiae tora seed, could likely regulate hyperlipidemia, its anti-obesity effect and related mechanism have not been elucidated. PURPOSE: The aim of this study was to examine whether RFG can ameliorate obesity and the mechanism of lipid accumulation regulated by RFG. STUDY DESIGN: In in vitro experiments, we confirmed the anti-adipogenic effect of RFG using 3T3-L1 cells and human adipose mesenchymal stem cells (hAMSCs). To confirm the anti-obesity effect, High-Fat Diet (HFD)-induced obese mice were selected as a model. METHODS: We investigated anti-adipogenic effects of RFG using MTS assay, Oil Red O Staining, real-time RT-PCR, western blot analysis, and immunofluorescence staining. The anti-obesity effect of RFG was confirmed in HFD-induced mice model using hematoxylin and eosin staining and serum analysis. RESULTS: RFG inhibited lipid accumulation in 3T3-L1 cells and hAMSCs by reducing expression of mammalian targets of rapamycin (mTOR), peroxisome proliferator-activated receptor (PPAR)γ, and CCAAT-enhancer binding protein (C/EBP)α. RFG phosphorylated AMP-activated protein kinase (AMPK) in a liver kinase B (LKB) 1-independent manner. Moreover, the anti-adipogenic effect of RFG was blocked by AMPK inhibitor. These results suggest that RFG inhibits lipid accumulation via AMPK signaling. Furthermore, RFG reduced the body weight, size of epididymal white adipose tissue (eWAT), and fatty liver in the mice. RFG also suppressed levels of adipogenic factors PPARγ, C/EBPα, FAS, LPL, and aP2) by activating AMPK in the eWAT and liver. CONCLUSION: RFG can ameliorate obesity, and thus, could be used as a therapeutic agent for treating obesity.


Subject(s)
Anti-Obesity Agents/pharmacology , Chromones/pharmacology , Glucosides/pharmacology , Lipid Metabolism/drug effects , Weight Gain/drug effects , 3T3-L1 Cells , AMP-Activated Protein Kinases/metabolism , Adipocytes/drug effects , Adipogenesis/drug effects , Adipogenesis/physiology , Animals , Body Weight/drug effects , Diet, High-Fat/adverse effects , Humans , Male , Mesenchymal Stem Cells/drug effects , Mesenchymal Stem Cells/metabolism , Mice , Mice, Obese , Obesity/drug therapy , Obesity/etiology , TOR Serine-Threonine Kinases/metabolism
9.
J Ginseng Res ; 43(1): 68-76, 2019 Jan.
Article in English | MEDLINE | ID: mdl-30662295

ABSTRACT

BACKGROUND: In colorectal cancer (CRC), 40-60% of patients develop metastasis. The epithelial-mesenchymal transition (EMT) is a pivotal and intricate process that increases the metastatic potential of CRC. The aim of this study was to investigate the effect of Korean Red Ginseng extract (RGE) on colorectal metastasis through inhibition of EMT and the metastatic abilities of CRC cells. METHODS: To investigate the effect of RGE on the metastatic phenotypes of CRC cells, CT26 and HT29 cells were evaluated by using an adhesion assay, a wound-healing assay, an invasion assay, zymography, and real-time reverse transcription-polymerase chain reaction. Western-blot analysis was conducted to elucidate the molecular mechanisms of RGE, which showed an inhibitory effect on the transforming growth factor-ß1 (TGF-ß1)-induced EMT in HT29 cells. Additionally, the antimetastatic effect of RGE was evaluated in a mouse model of lung metastasis injected with CT26 cells. RESULTS: RGE decreased the adhesion and migration ability of the CT26 cells and TGF-ß1-treated HT29 cells. The invasion ability was also reduced by RGE treatment through the inhibition of matrix metalloproteinase-9 expression and activity. Moreover, RGE suppressed the TGF-ß1-induced EMT via TGF-ß1/Smad-signaling-mediated Snail/E-cadherin expression in HT29 cells and lung tissue in CT26 tumor-bearing mice. CONCLUSION: Our results demonstrated that RGE inhibited colorectal lung metastasis through a reduction in metastatic phenotypes, such as migration, invasion, and the EMT of CRC cells.

10.
Nutrients ; 12(1)2019 Dec 26.
Article in English | MEDLINE | ID: mdl-31887988

ABSTRACT

BACKGROUND: Colorectal cancer (CRC) is one of the diseases with high prevalence and mortality worldwide. In particular, metastatic CRC shows low probability of surgery and lacks proper treatment. In this study, we conducted experiments to investigate the inhibitory effect of betulin against metastatic CRC and related mechanisms. METHODS: Water-soluble tetrazolium assay was used to determine the effect of betulin on metastatic CRC cell viability. Flow cytometry and TUNEL assay were performed to confirm whether betulin can induce apoptosis, autophagy, and cell cycle arrest. A lung metastasis mouse model was employed to estimate the anti-metastatic effect of betulin. RESULTS: betulin decreased viability of metastatic CRC cells, including CT26, HCT116, and SW620 cell lines. Through PI3K/Akt/mTOR inactivation, betulin induced AMPK-mediated G0/G1 phase arrest and autophagy of CT26 and HCT116 cells. In addition, betulin occurred caspase-dependent apoptosis via the mitogen-activated protein kinase signaling pathway in metastatic CRC cells. Moreover, orally administered betulin significantly inhibited metastasis of CT26 cells to the lung. CONCLUSION: Our results demonstrate the anti-metastatic effect and therapeutic potential of betulin in metastatic CRC treatment.


Subject(s)
Autophagy/drug effects , Cell Cycle Checkpoints/drug effects , Colonic Neoplasms/pathology , Lung Neoplasms , Triterpenes/pharmacology , Animals , Apoptosis/drug effects , Cell Line, Tumor , Humans , Lung Neoplasms/prevention & control , Lung Neoplasms/secondary , Mice , Mice, Inbred BALB C , Neoplasm Metastasis
11.
Oncol Rep ; 41(1): 202-212, 2019 Jan.
Article in English | MEDLINE | ID: mdl-30365120

ABSTRACT

Galla Rhois is a commonly used medicine in East Asia for the treatment of several diseases. However, the effects of Galla Rhois on the metastasis of colorectal cancer (CRC) and the underlying molecular mechanisms have not been studied. We investigated the anti­metastatic properties of Galla Rhois water extract (GRWE) on metastatic CRC cells. The effect of GRWE on the viability of colon 26 (CT26) cells was evaluated using WST­8 assay. Annexin V assay and western blot analysis were performed to elucidate the underlying molecular mechanisms involved in apoptosis. GRWE suppressed viability of CT26 cells by inducing apoptosis through the cleavage of caspase­3 and PARP, downregulation of caspase­8, caspase­9, Bcl­2 and Bcl­xL, and upregulation of Bax. Metastatic phenotypes such as epithelial­mesenchymal transition (EMT), migration, and invasion of CRC cells were investigated by real­time reverse transcription polymerase chain reaction, wound healing assay, and matrigel invasion assay, respectively. Non­cytotoxic concentrations of GRWE inhibited EMT in CRC cells by regulating the expression of EMT markers. GRWE attenuated cell migration and invasion through the inhibition of matrix metalloproteinase (MMP)­2 and MMP­9 activity. Moreover, GRWE suppressed colorectal lung metastasis in vivo, suggestive of its potential application for the treatment of colorectal metastasis.


Subject(s)
Adenylate Kinase/metabolism , Biological Products/administration & dosage , Colorectal Neoplasms/drug therapy , Lung Neoplasms/drug therapy , Lung Neoplasms/secondary , Animals , Biological Products/pharmacology , Cell Line, Tumor , Cell Movement/drug effects , Cell Proliferation/drug effects , Cell Survival/drug effects , Colorectal Neoplasms/metabolism , Epithelial-Mesenchymal Transition/drug effects , Gene Expression Regulation, Neoplastic/drug effects , HT29 Cells , Humans , Lung Neoplasms/metabolism , Mice , Xenograft Model Antitumor Assays
SELECTION OF CITATIONS
SEARCH DETAIL
...