Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 20
Filter
1.
Toxicon ; 233: 107243, 2023 Sep.
Article in English | MEDLINE | ID: mdl-37567418

ABSTRACT

It is the first report on leaves of Grewia bracteata Roth for its anticancer effect. In this study, three polarity-guided solvent extracts of Grewia bracteata leaves from n-hexane (GLH), ethyl acetate (GLE), and methanol (GLM) were screened for anticancer effects on HeLa, HCT-116, MCF-7, HCT-116 p53-/- and PC-3 cells via methyl thiazoldiphenyltetrazolium bromide (MTT) assay. Based on the results, GLM was fractionated, and the obtained fractions were tested on HCT-116 cells. Further, FT-IR, HPLC analysis, clonogenic assay, wound healing assay, DCFDA, and cell cycle experiments were conducted on HCT-116 cells. The extracts from methanol (GLM) and ethyl-acetate (GLE) demonstrated a more selective and promising inhibition on HCT-116 cells than others. Notably, GLM recorded superior inhibition on HCT-116 p53-/- than GLE. Amongst, the methanol column fraction (GMCF) showed prominent inhibition on HCT-116 (IC50:63.55 ± 0.61 µg/ml) and HCT-116 p53-/- (IC50: 84.51 ± 0.58 µg/ml) cells. Further, the test on normal cells (NKE) revealed minimal toxicity of GMCF. The phytochemical test, FT-IR, HPLC, and LC-HRMS analyses confirmed the high abundance of polyphenolic acid/polyphenols in GMCF. Further, the clonogenic and wound healing assays on HCT-116 cells were also performed. Later, the probable cell death mechanism was identified using DCFDA and cell cycle experiments. These experiments disclosed that GMCF induced HCT-116 cell death was probably due to reactive oxygen species (ROS) upregulation and cells cycle arrest at SubG0 phase. It inferred that the activity is most probably p53 independent, a tumor suppressor gene responsible for drug resistance in colon cancer.


Subject(s)
Colonic Neoplasms , Grewia , Humans , Reactive Oxygen Species/metabolism , Methanol , Tumor Suppressor Protein p53 , Genes, p53 , Spectroscopy, Fourier Transform Infrared , Apoptosis , Colonic Neoplasms/drug therapy , Colonic Neoplasms/metabolism , Colonic Neoplasms/pathology , Cell Line, Tumor , Plant Extracts/pharmacology
2.
Phytochemistry ; 212: 113727, 2023 Aug.
Article in English | MEDLINE | ID: mdl-37207991

ABSTRACT

Ten spirocyclic polycyclic polyprenylated acylphloroglucinols (PPAP), hunascynols A-J (1-10), and 12 known analogs were isolated from the aerial parts of Hypericum ascyron Linn. Compounds 1 and 2, which share a 1,2-seco-spirocyclic PPAP skeleton, could be derived from spirocyclic PPAP, with a common octahydrospiro[cyclohexan-1,5'-indene]-2,4,6-trione core, through a cascade of Retro-Claisen, keto-enol tautomerism, and esterification reactions. Aldolization of normal spirocyclic PPAP yielded 3, which has a caged framework with a 6/5/6/5/6 ring system. The structures of these compounds were determined using spectroscopy and X-ray diffraction. The inhibitory activities of all isolates were tested in three human cancer cell lines and a zebrafish model. Compounds 1 and 2 displayed moderate cytotoxicity against HCT116 cells (IC50 6.87 and 9.86 µM, respectively). The mechanisms of these compounds were evaluated using Western blot assays. Compounds 3 and 5 inhibited the growth of sub-intestinal vessels in zebrafish embryos. Further, the target genes were screened using real-time PCR.


Subject(s)
Hypericum , Humans , Animals , Molecular Structure , Hypericum/chemistry , Zebrafish , Cell Line , Phloroglucinol
3.
J Ethnopharmacol ; 303: 115933, 2023 Mar 01.
Article in English | MEDLINE | ID: mdl-36403742

ABSTRACT

ETHNOPHARMACOLOGICAL RELEVANCE: Wei-Tong-Xin (WTX), derives from the Chinese herbal decoction (CHD) of Wan-Ying-Yuan in ancient China, has been shown to be effective therapeutic herbal decoction for treating gastrointestinal diseases. Present studies have demonstrated that WTX had potential to alleviate the symptoms of gastrointestinal inflammation, gastric ulcer and improve gastric motility. AIM OF THE STUDY: The study primarily focused on exploring the therapeutic effect and possible pharmacological mechanism of WTX on colorectal cancer (CRC) based on network pharmacology, in vitro and in vivo experiments. MATERIALS AND METHODS: Firstly, colorectal cancer and WTX associated with targets were searched from GeneCards database and TCM Systems Pharmacology Database and Analysis Platform (TCMSP) respectively. The protein-protein interaction (PPI) network also was constructed to screening key targets. In addition, the Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) enrichment analyses were applied to predict the underlying biological function and mechanism involving in the anti-colorectal cancer effect of WTX. Next, CCK-8, colony formation and transwell assays were performed to verify the influence of proliferation and metastasizing ability of HCT116 cells after treated with WTX. Cell cycle, apoptosis and reactive oxygen species (ROS) were analysis by flow cytometry. Hoechst 33258 staining was conducted to observe nuclear morphology changes. Protein expression of apoptosis and PI3K/AKT signaling as well as mRNA expression of ferroptosis and apoptosis were determined by Western Blotting and RT-qPCR. The effects of WTX and LY294002 combination on the PI3K/Akt/mTOR signaling pathway were measured by Western Blotting. Finally, the xenograft tumor mouse model was established by subcutaneous injection of CT26 cells to measure tumors volume and weight. Hematoxylin and eosin (HE) staining and immunohistochemical analysis were used to observe the pathological changes and the protein expression in tumor tissues. RESULTS: There were 286 potential treatment targets from 130 bioactive compounds in WTX, 1349 CRC-related targets were identified. Eleven core targets (TP53, AKT1, STAT3, JUN, TNF, HSP90AA1, IL-6, MAPK3, CASP3, EGFR, MYC) were found by PPI network analysis constructed of 142 common targets. The results of KEGG enrichment displayed PI3K/AKT signaling pathway as core pathway. After the treatment of WTX, the inhibitory of viability, metastases and cell cycle arrest at G2/M phase were observed in HCT116 cells. Moreover, WTX induced an increase in the expression of apoptosis proteins (Bak, cytochrome c, cleaved caspase-9/caspase-9 and cleaved caspase-3/caspase-3) and the levels of ROS and MDA, a decrease in the expression of PI3K/AKT signaling related proteins (PI3K, p-PI3K, p-AKT/AKT and p-mTOR/mTOR) and the level of SOD. WTX treatment significantly reduced the tumor weight, increased cleaved caspase-3 positive area and decreased that of ki67 in xenograft mouse model. CONCLUSION: Through a network pharmacology approach and in vitro experiments, we predicted and verified the effect of WTX on colorectal cancer cells mainly depended on the regulation of intrinsic apoptosis via PI3K/AKT signaling pathway, and further animal experiments proved that WTX has a good anti-colon cancer effect in vivo.


Subject(s)
Drugs, Chinese Herbal , Neoplasms , Humans , Animals , Mice , Caspase 3 , Caspase 9 , Network Pharmacology , Phosphatidylinositol 3-Kinases , Proto-Oncogene Proteins c-akt , Reactive Oxygen Species , Apoptosis , TOR Serine-Threonine Kinases , Signal Transduction , Drugs, Chinese Herbal/pharmacology , Drugs, Chinese Herbal/therapeutic use , Molecular Docking Simulation
4.
Metabolites ; 12(11)2022 Oct 31.
Article in English | MEDLINE | ID: mdl-36355130

ABSTRACT

Doxorubicin (DOX) is a chemotherapeutic agent is used for various cancer cells. To characterize the chemical structural components and metabolic inhibition, we applied a DOX to HCT116 colon cancer cells using an independent metabolites profiling approach. Chemical metabolomics has been involved in the new drug delivery systems. Metabolomics profiling of DOX-applied HCT116 colon cancer cellular metabolisms is rare. We used 1H nuclear magnetic resonance (NMR) spectroscopy in this study to clarify how DOX exposure affected HCT116 colon cancer cells. Metabolomics profiling in HCT116 cells detects 50 metabolites. Tracking metabolites can reveal pathway activities. HCT116 colon cancer cells were evenly treated with different concentrations of DOX for 24 h. The endogenous metabolites were identified by comparison with healthy cells. We found that acetate, glucose, glutamate, glutamine, sn-glycero-3-phosphocholine, valine, methionine, and isoleucine were increased. Metabolic expression of alanine, choline, fumarate, taurine, o-phosphocholine, inosine, lysine, and phenylalanine was decreased in HCT116 cancer cells. The metabolic phenotypic expression is markedly altered during a high dose of DOX. It is the first time that there is a metabolite pool and phenotypic expression in colon cancer cells. Targeting the DOX-metabolite axis may be a novel strategy for improving the curative effect of DOX-based therapy for colon cancer cells. These methods facilitate the routine metabolomic analysis of cancer cells.

5.
Molecules ; 27(17)2022 Sep 01.
Article in English | MEDLINE | ID: mdl-36080410

ABSTRACT

The advanced technology for synthesizing nanoparticles utilizes natural resources in an environmentally friendly manner. Additionally, green synthesis is preferred to chemical and physical synthesis because it takes less time and effort. The green synthesis of cobalt oxide nanoparticles has recently risen due to its physico-chemical properties. In this study, many functional groups present in Psidium guajava leaf extracts are used to stabilize the synthesis of cobalt oxide nanoparticles. The biosynthesized cobalt oxide nanoparticles were investigated using UV-visible spectroscopic analysis. Additionally, Fourier-transform infrared spectroscopy revealed the presence of carboxylic acids, hydroxyl groups, aromatic amines, alcohols and phenolic groups. The X-ray diffraction analysis showed various peaks ranging from 32.35 to 67.35°, and the highest intensity showed at 36.69°. The particle size ranged from 26 to 40 nm and confirmed the average particle size is 30.9 nm. The green synthesized P. guajava cobalt oxide nanoparticles contain cobalt as the major abundant element, with 42.26 wt% and 18.75 at% confirmed by the EDAX techniques. SEM images of green synthesized P. guajava cobalt oxide nanoparticles showed agglomerated and non-uniform spherical particles. The anti-bacterial activity of green synthesized P. guajava cobalt oxide nanoparticles was evaluated against Gram-positive Staphylococcus aureus and Gram-negative Escherichia coli with a 7 to 18 mm inhibitory zone. The photocatalytic activity was evaluated using green synthesized P. guajava cobalt oxide nanoparticles and observed 79% of dye degradation. The MTT assay of P. guajava cobalt oxide nanoparticles showed an excellent cytotoxic effect against MCF 7 and HCT 116 cells compared to normal cells. The percentage of cell viability of P. guajava cobalt oxide nanoparticles was observed as 90, 83, 77, 68, 61, 58 and 52% for MCF-7 cells and 82, 70, 63, 51, 43, 40, and 37% for HCT 116 cells at the concentration of 1.53, 3.06, 6.12, 12.24, 24.48, 50, and 100 µg/mL compared to control cells. These results confirmed that green synthesized P. guajava cobalt oxide nanoparticles have a potential photocatalytic and anti-bacterial activity and also reduced cell viability against MCF-7 breast cancer and HCT 116 colorectal cancer cells.


Subject(s)
Metal Nanoparticles , Psidium , Anti-Bacterial Agents/chemistry , Cobalt/metabolism , Green Chemistry Technology/methods , Metal Nanoparticles/chemistry , Microbial Sensitivity Tests , Oxides , Plant Extracts/chemistry , Psidium/chemistry , Spectroscopy, Fourier Transform Infrared , X-Ray Diffraction
6.
Molecules ; 26(19)2021 Sep 25.
Article in English | MEDLINE | ID: mdl-34641357

ABSTRACT

We report the development of in vitro propagation protocols through an adventitious shoot induction pathway for a rare and medicinal Scutellaria havanensis. In vitro propagation studies using nodal explants showed MS medium supplemented with 10 µM 6-Benzylaminopurine induced the highest number of adventitious shoots in a time-dependent manner. A ten-day incubation was optimum for shoot bud induction as longer exposures resulted in hyperhydricity of the explants and shoots induced. We also report preliminary evidence of Agrobacterium tumefaciens EHA105-mediated gene transfer transiently expressing the green fluorescent protein in this species. Transformation studies exhibited amenability of various explant tissues, internode being the most receptive. As the plant has medicinal value, research was carried out to evaluate its potential antioxidant capacity and the efficacy of methanolic leaf extracts in curbing the viability of human colorectal cancer cell line HCT116. Comparative total polyphenol and flavonoid content measurement of fresh and air-dried leaf extract revealed that the fresh leaf extracts contain higher total polyphenol and flavonoid content. The HCT 116 cell viability was assessed by colorimetric assay using a 3-(4,5-dimethyl-thiazol-2-yl)-2,5-diphenyltetrazolium bromide, showed a steady growth inhibition after 24 h of incubation. Scanning electron microscopy of leaf surface revealed a high density of glandular and non-glandular trichomes. This research provides a basis for the conservation of this rare plant and future phytochemical screening and clinical research.


Subject(s)
Antineoplastic Agents/pharmacology , Antioxidants/pharmacology , Colonic Neoplasms/drug therapy , Plant Extracts/pharmacology , Plant Shoots/chemistry , Scutellaria/chemistry , Cell Proliferation , Colonic Neoplasms/pathology , HCT116 Cells , Humans
7.
Mol Biol Rep ; 48(10): 6805-6820, 2021 Oct.
Article in English | MEDLINE | ID: mdl-34468912

ABSTRACT

BACKGROUND: Colorectal cancer (CRC) is major aliment around the word, with a cumulative rate of mortality. Metformin (MT) was recently approved as anticancer drug against solid tumors, such as CRC. Resistance to MT therapy remains to be a challenging matter facing the development of possible anti-cancer strategy. To circumvent this problem, MT nano-encapsulation has been introduced to sensitize resistant cancer cells. The purpose of the current study is to explore the MT's aptitude encapsulated in lecithin (LC) and chitosan (CS) nanoparticles to inhibit CRC proliferation through modulations of long noncoding RNAs (lncRNAs), micro RNAs (miRNAs), and some biochemical markers. METHODS AND RESULTS: Cytotoxic screenings of the newly synthesized MT-based regimens; MT, MT-LC NPs (NP1), MT-CS NPs (NP2), and MT-LC-CS NPs (NP3) against colorectal cancerous Caco-2 and HCT116 cell lines versus normal WI-38 cells were performed. The epigenetic mechanistic effects of these proposed regimens on lncRNAs and miRNAs were investigated. Additionally, some protein levels were assessed in CRC cells upon treatments; YKL-40, PPARγ, E-cadherin (ECN), and VEGF. We resulted that NP1 recorded the highest significant cytotoxic effect on CRC cells. HCT116 cells were more sensitive to the NP1 compared to Caco-2 cells. Intriguingly, it was suggested that NP1 tackled the CRC cells through down-regulation of the H19, HOTTIP, HULC, LINC00641, miR-200, miR-92a, miR-21, YKL-40, PPARγ, and VEGF expressions, as well as up-regulation of the miR-944 and ECN expressions. CONCLUSIONS: We concluded that the NP1 can potentially be cytotoxic to CRC cells in-vitro by modulating noncoding RNA.


Subject(s)
Colorectal Neoplasms/genetics , Epigenesis, Genetic , Gene Expression Regulation, Neoplastic , Lecithins/chemistry , Metformin/pharmacology , Nanoparticles/chemistry , RNA, Long Noncoding/genetics , Antineoplastic Agents/pharmacology , Cadherins/metabolism , Cell Death/drug effects , Cell Line, Tumor , Chitinase-3-Like Protein 1/metabolism , Colorectal Neoplasms/pathology , Drug Liberation , Gene Expression Regulation, Neoplastic/drug effects , Humans , Inhibitory Concentration 50 , MicroRNAs/genetics , MicroRNAs/metabolism , Nanoparticles/ultrastructure , PPAR gamma/metabolism , Particle Size , RNA, Long Noncoding/metabolism , Static Electricity , Vascular Endothelial Growth Factor A/metabolism
8.
Iran Biomed J ; 25(2): 106-16, 2021 03 01.
Article in English | MEDLINE | ID: mdl-33465843

ABSTRACT

Background: To study the anticancer activity of Plantago major, we assessed the effect of ethanolic, methanolic and acetonic extracts of this plant on HCT-116, SW-480, and HEK-293 cell lines as control. Methods: The cytotoxic activity, biocompatibility, and toxicity were evaluated by MTT assay, hemolysis, and Artemia salina-LD50 (on mice) tests, respectively. The analysis of the extracts was performed by GC-MS analysis. Results: The results showed that all the extracts had the most antiproliferative properties on the HCT-116 cell line. The P. major root extract was more effective than the aerial parts, and IC50 values for ethanolic, methanolic and acetonic root extracts were 405.59, 470.16, and 82.26 µg/mL, respectively on HCT-116 cell line at 72 h. Hemolysis degree of the ethanolic extract of aerial and root parts were approximately 1% at 400 µg/mL.. Using the ethanolic extracts, the Artemia survived every concentration, and no toxicity was observed. One week after the oral administration of different parts of P. major extracts, none of the mice died, even those were administered 2000 mg/kg. The results of GC/MS analysis showed that P. major extracts contain potential anticancer compounds, such as stearic acid (8.61%) in aerial parts of methanolic extract and 1,2- Benzenedicarboxylic acid, mono(2-ethylhexyl)ester (88.07% and 40.63%) in aerial and root parts of acetonic extract of P. major. Conclusion: Our findings suggest that the P. major is a source of potential compounds with antiproliferative properties.


Subject(s)
Biocompatible Materials/pharmacology , Plant Extracts/chemistry , Plant Extracts/pharmacology , Plantago/chemistry , Plants, Medicinal/chemistry , Administration, Oral , Animals , Artemia , Body Weight/drug effects , Cell Death/drug effects , Cell Line, Tumor , Cell Proliferation/drug effects , Ethanol , Gas Chromatography-Mass Spectrometry , HEK293 Cells , Hemolysis/drug effects , Humans , Inhibitory Concentration 50 , Mice , Plant Extracts/toxicity , Toxicity Tests, Acute
9.
Curr Pharm Biotechnol ; 22(2): 262-273, 2021.
Article in English | MEDLINE | ID: mdl-32532192

ABSTRACT

BACKGROUND: The anticancer effects of Phyllanthus amarus extract on various cancer cells have been investigated, however, the effects of its major constituents on HCT116 human colorectal cancer cells have not been reported. OBJECTIVE: In the present study, we investigated the cytotoxic effect of 80% ethanol extract of P. amarus and its marker constituents (phyllanthin, hypophyllanthin, gallic acid, niranthin, greraniin, phyltetralin, isolintetralin, corilagin and ellagic acid) on HCT116 and their underlying mechanisms of action. METHODS: Their antiproliferative and apoptotic effects on HCT 116 were performed using MTT assay and flow cytometric analysis, respectively, while caspases 3/7, 8 and 9 activities were examined using the colorimetric method. The expression of cleaved poly ADP ribose polymerase enzyme (PARP) and cytochrome c proteins was investigated by the immune-blot technique. RESULTS AND DISCUSSION: HPLC and LC-MS/MS analyses demonstrated that the extract contained mainly lignans and polyphenols. The plant samples markedly suppressed the growth and expansion of HCT116 cells in a concentration- and time-dependent manner with no toxicity against normal human fibroblast CCD18 Co. P. amarus extract, phyllanthin and gallic acid induced mode of cell death primarily through apoptosis as confirmed by the exteriorization of phosphatidylserine. Caspases 3/7, 8, and 9 activities increased in a concentration-dependent manner following 24h treatment. The expressions of cleaved PARP (Asp 214) and cytochrome c were markedly upregulated. CONCLUSION: P. amarus extract, phyllanthin and gallic acid exhibited an apoptotic effect on HCT116 cells through the caspases-dependent pathway.


Subject(s)
Apoptosis/drug effects , Caspases/metabolism , Colonic Neoplasms/enzymology , Lignans/pharmacology , Phyllanthus , Plant Extracts/pharmacology , Polyphenols/pharmacology , Apoptosis/physiology , Cell Proliferation/drug effects , Cell Proliferation/physiology , Chromatography, Liquid/methods , Colonic Neoplasms/drug therapy , Colonic Neoplasms/pathology , Dose-Response Relationship, Drug , HCT116 Cells , Humans , Lignans/isolation & purification , Lignans/therapeutic use , Plant Extracts/isolation & purification , Plant Extracts/therapeutic use , Polyphenols/isolation & purification , Polyphenols/therapeutic use , Signal Transduction/drug effects , Signal Transduction/physiology , Tandem Mass Spectrometry/methods
10.
Mol Biol Rep ; 47(11): 8831-8840, 2020 Nov.
Article in English | MEDLINE | ID: mdl-33141288

ABSTRACT

The Artemisia absinthium (AA), belongs to the Asteraceae family, is used as a therapeutic agent in traditional medicine in Iran. It is a rich source of biology-active compounds. However, the molecular mechanism of AA contributing to cell proliferation and apoptosis is still unknown. This study aims to assess the anticancer activity of the methanolic extract of A. absinthium (MEAA) against human colorectal cancer HCT-116 cell line. The cytotoxic effects of MEAA on HCT-116 cells was evaluated by 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium (MTT) assay. The expression levels of BAX and BCL-2 in HCT-116 cell line were examined by qRT-PCR. Annexin V/PI-flow cytometry technique was used to detect the cell cycle and apoptosis. MMP was predicted by Rhodamine 123 staining, and caspase 3 activity was analyzed by ELISA. Western blot method was performed to detect the expression level of BAX, Bcl-2 and Caspase-3 proteins. The MTT test revealed MEAA reduced the viability of HCT-116 cells. The mRNA and protein levels of BAX increased, but those of BCL-2 decreased in MEAA-treated cells. MEAA also prompted cell cycle arrest and induced apoptosis. After adding MEAA, the protein level and activity of caspase 3 and MMP destruction significantly increased. MEAA predominantly prompted apoptosis in HCT-116 cells by activating the mitochondrial pathway.


Subject(s)
Apoptosis/drug effects , Caspase 3/metabolism , Cell Cycle Checkpoints/drug effects , Membrane Potential, Mitochondrial/drug effects , Plant Extracts/pharmacology , Proto-Oncogene Proteins c-bcl-2/metabolism , bcl-2-Associated X Protein/metabolism , Animals , Artemisia absinthium/chemistry , Cell Survival/drug effects , Chlorocebus aethiops , Colorectal Neoplasms/genetics , Colorectal Neoplasms/metabolism , Colorectal Neoplasms/pathology , Dose-Response Relationship, Drug , Enzyme Activation/drug effects , Gene Expression Regulation, Neoplastic/drug effects , HCT116 Cells , Humans , Methanol/chemistry , Proto-Oncogene Proteins c-bcl-2/genetics , Vero Cells , bcl-2-Associated X Protein/genetics
11.
Oncol Rep ; 44(5): 1997-2008, 2020 11.
Article in English | MEDLINE | ID: mdl-33000266

ABSTRACT

Curcumin is the main component of the Chinese herbal plant turmeric, which has been demonstrated to possess antitumor and other pharmacological properties. The aim of the present study was to investigate the effects of curcumin on the viability, migration and apoptosis of human colorectal carcinoma HCT­116 cells, and to explore the underlying molecular mechanisms. In addition, it was investigated whether the antitumor effect of curcumin on HCT­116 cells could match that of the chemotherapeutic drug 5­fluorouracil (5­FU). HCT­116 cells were treated with curcumin (10, 20 and 30 µM) and 5­FU (500 µM), and cell viability and proliferation were detected by Cell Counting Kit­8 and colony formation assays, respectively. The migration and invasion of treated cells were determined using Transwell and carboxyfluorescein succinimidyl amino ester fluorescent labeling assays. Cell cycle distribution and apoptosis rates were detected by flow cytometry. Furthermore, cell morphology changes associated with apoptosis were observed by fluorescence microscopy with acridine orange/ethidium bromide dual staining. To investigate the possible underlying molecular mechanisms, the gene and protein levels of Fas, Fas­associated via death domain (FADD), caspase­8, caspase­3, matrix metalloproteinase (MMP)­9, nuclear factor (NF)­κB, E­cadherin and claudin­3 were detected using quantitative PCR analysis, zymography and western blotting. The results revealed that curcumin markedly inhibited the viability and proliferation of HCT­116 cells in a dose­ and time­dependent manner. The migration, aggregation and invasion of HCT­116 cells into the lungs of mice were decreased by curcumin treatment in a dose­dependent manner. S­phase arrest and gradually increased apoptotic rates of HCT­116 cells were observed with increasing curcumin concentrations. Additionally, the mRNA and protein levels of apoptosis­associated proteins (Fas, FADD, caspase­8 and caspase­3) and E­cadherin in HCT­116 cells were upregulated following treatment with curcumin in a dose­dependent manner. By contrast, the expression of migration­associated proteins, including MMP­9, NF­κB and claudin­3, was downregulated with increasing curcumin concentrations. These data suggested that the inhibitory effect of curcumin on HCT­116 cells may match that of 5­FU. Therefore, curcumin induced cell apoptosis and inhibited tumor cell metastasis by regulating the NF­κB signaling pathway, and its therapeutic effect may be comparable to that of 5­FU.


Subject(s)
Colorectal Neoplasms/drug therapy , Curcumin/pharmacology , Animals , Apoptosis/drug effects , Cell Movement/drug effects , Cell Proliferation/drug effects , Colorectal Neoplasms/pathology , Curcumin/therapeutic use , Fluorouracil/pharmacology , Fluorouracil/therapeutic use , HCT116 Cells , Humans , Mice , NF-kappa B/metabolism , Signal Transduction/drug effects , Xenograft Model Antitumor Assays
12.
Chin J Nat Med ; 18(2): 81-89, 2020 Feb.
Article in English | MEDLINE | ID: mdl-32172951

ABSTRACT

Nervosine VII is one of the known saturated pyrrolizidine alkaloids isolated from the plant of Liparis nervosa. This is first study to investigate the antitumor activity of nervosine VII in vitro, and the results indicated that nervosine VII induced autophagy and apoptosis in HCT116 human colorectal cancer cells. Mechanistic studies showed that nervosine VII-induced apoptosis was associated with the intrinsic pathway by the activation of caspase-9, -3 and -7. Autophagy induced by nervosine VII was characteristic with the regulation of autophagic markers including the increase of LC3-II and beclin 1 proteins, and the decrease of p62 protein. Nervosine VII simultaneously induced autophagy and apoptosis by activated MAPKs signaling pathway including JNK, ERK1/2 and p38, suppressing the p53 signaling pathway.


Subject(s)
Antineoplastic Agents/pharmacology , Apoptosis/drug effects , Autophagy/drug effects , Plant Extracts/pharmacology , Pyrrolizidine Alkaloids/pharmacology , HCT116 Cells , Humans , Orchidaceae/chemistry
13.
J Photochem Photobiol B ; 204: 111767, 2020 Mar.
Article in English | MEDLINE | ID: mdl-32006893

ABSTRACT

Colon carcinoma is a recurring type of cancer that affects the intestine epithelial with a poor survival rate. It was already proven the anticancer property of hesperidin in various cancers but the bioavailability hesperidin is poor, which hinders the hesperidin usage. In this investigation we synthesized hesperidin loaded Zn2+@ SA/PCT nanocomposites and assessed its anticancer potential against colon cancer (HCT116) cells. Hesperidin loaded Zn2+@ SA/PCT nanocomposites were characterized using Fourier transform infrared (FTIR), X-ray diffraction (XRD), scanning electron microscope (SEM) and transmission electron microscope (TEM) analysis. The drug releasing capacity and cytotoxic property was assessed via drug releasing assay, MTT assay with HCT116 cells. The anticancer potency of hesperidin nanocomposites were evaluated with TUNEL, DAPI staining, reactive oxygen species (ROS) generation assay and it is confirmed with flow cytometry analysis of MMP disruption in colon cancer (HCT116) cell line. Further the immunoblotting analysis of cysteine proteases Caspases 3, 9, PARP, proapoptotic protein Bax and antiapoptotic protein Bcl2 were performed. The results of FTIR, XRD and electroscopic analyses confirmed the synthesized hesperidin nanocomposites accomplish the properties of potent nanodrug and the MTT assay authentically confirmed that the synthesized hesperidin nanocomposite inhibited the HCT116 cell growth, and the results of fluorescent staining proved that the hesperidin nanocomposite induced the apoptotic mediated cell necrosis via promoting the expression of apoptotic proteins thereby induced the apoptosis in colon cancer (HCT116) cells. Hence, it was concluded that the, hesperidin loaded nanocomposites persuasively inhibited proliferation of colon carcinoma cell and induced apoptosis in in vitro condition.


Subject(s)
Antineoplastic Agents, Phytogenic/pharmacology , Cell Proliferation/drug effects , Hesperidin/chemistry , Nanocomposites/chemistry , Alginates/chemistry , Antineoplastic Agents, Phytogenic/chemistry , Caspase 3/metabolism , Cell Survival/drug effects , Colonic Neoplasms/metabolism , Colonic Neoplasms/pathology , Drug Compounding , Drug Liberation , HCT116 Cells , Hesperidin/pharmacology , Humans , Membrane Potential, Mitochondrial/drug effects , Pectins/chemistry , Proto-Oncogene Proteins c-bcl-2/genetics , Proto-Oncogene Proteins c-bcl-2/metabolism , Reactive Oxygen Species/metabolism , Zinc/chemistry
14.
Acta Pharmacol Sin ; 39(10): 1645-1660, 2018 Oct.
Article in English | MEDLINE | ID: mdl-29849127

ABSTRACT

Escin, a triterpene saponin isolated from horse chestnut seed, has been used to treat encephaledema, tissue swelling and chronic venous insufficiency. Recent studies show that escin induces cell cycle arrest, tumor proliferation inhibition and tumor cell apoptosis. But the relationship between escin-induced DNA damage and cell apoptosis in tumor cells remains unclear. In this study, we investigated whether and how escin-induced DNA damage contributed to escin-induced apoptosis in human colorectal cancer cells. Escin (5-80 µg/mL) dose-dependently inhibited the cell viability and colony formation in HCT116 and HCT8 cells. Escin treatment induced DNA damage, leading to p-ATM and γH2AX upregulation. Meanwhile, escin treatment increased the expression of p62, an adaptor protein, which played a crucial role in controlling cell survival and tumorigenesis, and had a protective effect against escin-induced DNA damage: knockdown of p62 apparently enhanced escin-induced DNA damage, whereas overexpression of p62 reduced escin-induced DNA damage. In addition, escin treatment induced concentration- and time-dependent apoptosis. Similarly, knockdown of p62 significantly increased escin-induced apoptosis in vitro and produced en escin-like antitumor effect in vivo. Overexpression of p62 decreased the rate of apoptosis. Further studies revealed that the functions of p62 in escin-induced DNA damage were associated with escin-induced apoptosis, and p62 knockdown combined with the ATM inhibitor KU55933 augmented escin-induced DNA damage and further increased escin-induced apoptosis. In conclusion, our results demonstrate that p62 regulates ATM/γH2AX pathway-mediated escin-induced DNA damage and apoptosis.


Subject(s)
Antineoplastic Agents/therapeutic use , Apoptosis/drug effects , Colorectal Neoplasms/drug therapy , DNA Damage/drug effects , Escin/therapeutic use , Sequestosome-1 Protein/metabolism , Animals , Antineoplastic Agents/pharmacology , Ataxia Telangiectasia Mutated Proteins/genetics , Ataxia Telangiectasia Mutated Proteins/metabolism , Autophagy/drug effects , Cell Line, Tumor , Cell Proliferation/drug effects , Down-Regulation , Escin/pharmacology , Female , Histones/genetics , Histones/metabolism , Humans , Mice, Nude , Sequestosome-1 Protein/genetics , Signal Transduction/drug effects , Up-Regulation
15.
Phytomedicine ; 40: 140-147, 2018 Feb 01.
Article in English | MEDLINE | ID: mdl-29496166

ABSTRACT

BACKGROUND: Colorectal cancer (CRC) is one of the most common malignancies associated with high mortality rate worldwide. We previously reported that pristimerin inhibits cell growth and induces apoptosis in CRC cells. HYPOTHESIS/PURPOSE: To further understand the molecular mechanism by which pristimerin elicits its anticancer activities on colon cancer cells, we investigated its effect on nuclear factor-κB (NF-κB) signaling pathway. STUDY DESIGN: This study consisted of both in vitro and in vivo experiments involving HCT-116 cell line and xenograft mouse model. Molecular techniques such as qRT-PCR, western blotting and immunofluorescence were used to demonstrate pristimerin in vitro effect on NF-κB signaling pathway; whereas it's in vivo activity was analyzed by western blot and immunohistochemistry on tumor tissues. RESULTS: Our in vitro results on HCT-116 cells showed that pristimerin inhibited IKK phosphorylation, IкB-α degradations and IкB-α phosphorylation in both dose- and time- dependent manners, which caused suppression of NF-кB p65 phosphorylation, nuclear translocation and accumulation of NF-кB. Moreover, pristimerin was found to inhibit both constitutive activated-NF-кB and tumor necrosis factor-α (TNF-α)- and lipopolysaccharide (LPS)-induced activation of NF-кB signaling pathway. Furthermore, our in vivo results on xenograft animal model revealed that pristimerin inhibited tumor growth mainly through suppressing NF-кB activity in tumor tissues. CONCLUSION: Pristimerin antitumor activities were mainly mediated through inhibition of NF-кB signaling pathway in colon tumor cells. These findings further explain that pristimerin has the therapeutic potential for targeting colon cancer.


Subject(s)
Antineoplastic Agents, Phytogenic/pharmacology , Colorectal Neoplasms/drug therapy , NF-kappa B/metabolism , Triterpenes/pharmacology , Animals , Antineoplastic Agents, Phytogenic/administration & dosage , Apoptosis/drug effects , Cell Proliferation/drug effects , Colorectal Neoplasms/metabolism , Colorectal Neoplasms/pathology , Dose-Response Relationship, Drug , Female , HCT116 Cells , Humans , Lipopolysaccharides/pharmacology , Mice, Inbred BALB C , Pentacyclic Triterpenes , Phosphorylation , Signal Transduction/drug effects , Triterpenes/administration & dosage , Tumor Necrosis Factor-alpha/pharmacology , Xenograft Model Antitumor Assays
16.
Ultrastruct Pathol ; 42(3): 255-261, 2018.
Article in English | MEDLINE | ID: mdl-29565703

ABSTRACT

Medicinal plants have recently gained increasing scientific interest as an important source of molecules with different therapeutic potentials. Accordingly, the present study was carried out to investigate ultrastructural changes induced by the aqueous extract of Solanum incanum (SI) fruit on human colorectal carcinoma cell line (HCT 116 cells). Examination of SI-treated HCT 116 cells with transmission electron microscopy (TEM) demonstrated numerous ultrastructural changes in the form of loss of the surface microvilli, mitochondrial damage and dilatation of cristae, and formation of autophagic vacuoles and increasing numbers of lipid droplets. Also, majority of the treated cells showed nuclear shrinkage with chromatin condensation and nucleolar changes. Moreover, some cells showed focal areas of cytoplasmic degeneration associating with formation of myelin figures and fatty globules. In conclusion, TEM was able to verify cytotoxicity of SI aqueous extract against HCT 116 colon cancer cells.


Subject(s)
Antineoplastic Agents/pharmacology , Colonic Neoplasms/ultrastructure , Plant Extracts/pharmacology , Apoptosis/drug effects , Cell Proliferation/drug effects , Fruit , HCT116 Cells , Humans , Microscopy, Electron, Transmission , Solanum
17.
Integr Med Res ; 6(2): 179-189, 2017 Jun.
Article in English | MEDLINE | ID: mdl-28664141

ABSTRACT

BACKGROUND: Dae-Hwang-Mok-Dan-Tang (DHMDT), a traditional Korean medicine, contains five species of medicinal plants and has been used to treat patients with digestive tract cancer for hundreds of years; however, its anticancer mechanism is poorly understood. In the present study, we investigated the proapoptotic effects of DHMDT in human colon cancer HCT-116 cells. METHODS: Cytotoxicity was evaluated using the 3-(4,5-dimethyl-2-thiazolyl)-2,5-diphenyl-2H-tetrazolium bromide assay. Apoptosis was detected using 4,6-diamidino-2-phenyllindile staining, agarose gel electrophoresis, and flow cytometry. The protein levels were determined using Western blot analysis. Caspase activity was measured using a colorimetric assay. RESULTS: Treatment with DHMDT resulted in a growth inhibition coupled with apoptosis induction, which was associated with the downregulation of members of IAP (inhibitor of apoptosis protein) family, including XIAP and survivin, and the activation of caspase-9 and -3 accompanied by proteolytic degradation of poly(ADP-ribose)-polymerase and phospholipase C-γ1. DHMDT treatment also showed a correlation with the translocation of proapoptotic Bax to mitochondria, the loss of mitochondrial membrane permeabilization, and the cytochrome c release from the mitochondria to the cytosol. Moreover, DHMDT increased the levels of death receptor-associated ligands and enhanced activation of caspase-8 and cleavage of its substrate, Bid. However, the pan-caspase inhibitor could reverse DHMDT-induced apoptosis. In addition, DHMDT suppressed the phosphoinositide 3-kinase (PI3K)/Akt pathway, and treatment with a potent inhibitor of PI3K further increased the apoptotic activity of DHMDT. CONCLUSION: Our data showed that DHMDT induces HCT-116 cell apoptosis by activating intrinsic and extrinsic apoptosis pathways and by suppressing the PI3K/Akt signal pathway; however, further studies are needed to identify the active compounds.

18.
Bioorg Med Chem Lett ; 27(14): 3144-3147, 2017 07 15.
Article in English | MEDLINE | ID: mdl-28532669

ABSTRACT

Two new pterosin glycosides, (2S,3S)-pterosin C 3-O-ß-d-(4'-(E)-caffeoyl)-glucopyranoside (1) and (2S,3S)-pterosin C 3-O-ß-d-(6'-(E)-p-coumaroyl)-glucopyranoside (2), were isolated from Pteris multifida (Pteridaceae) roots along with ten known pterosin compounds (3-12). The chemical structures of the isolated compounds were elucidated by extensive analysis of the 1D, 2D NMR, HRESIMS, and CD spectroscopic data. The cytotoxicities of 1-12 against HCT116 human colorectal cancer cell line were evaluated. Among the isolates, compound 1 showed moderate antiproliferative activity in HCT116 cells with an IC50 value of 8.0±1.7µM. Additionally, 1 induced the upregulation of the caspase-9 and procaspase-9 levels in Western blots and increased the annexin V/propidium iodide (PI)-positive cell population in flow cytometry.


Subject(s)
Indans/chemistry , Indans/pharmacology , Pteris/chemistry , Sesquiterpenes/chemistry , Sesquiterpenes/pharmacology , Antineoplastic Agents, Phytogenic/chemistry , Antineoplastic Agents, Phytogenic/isolation & purification , Antineoplastic Agents, Phytogenic/toxicity , Caspase 9/metabolism , Cell Proliferation/drug effects , Circular Dichroism , Colonic Neoplasms , HCT116 Cells , Humans , Indans/isolation & purification , Magnetic Resonance Spectroscopy , Mass Spectrometry , Molecular Conformation , Plant Extracts/chemistry , Plant Roots/chemistry , Plant Roots/metabolism , Pteris/metabolism , Sesquiterpenes/isolation & purification , Sesquiterpenes/toxicity
19.
Int J Mol Sci ; 16(11): 27411-21, 2015 Nov 16.
Article in English | MEDLINE | ID: mdl-26580615

ABSTRACT

Evodiamine (EVO) exhibits strong anti-cancer effects. However, the effect of EVO on the human colorectal cancer cell line HCT-116 has not been explored in detail, and its underlying molecular mechanisms remain unknown. In the present study, cell viability was assessed by Cell Counting Kit-8 (CCK-8). Cell cycle and apoptosis were measured by flow cytometry, and morphological changes in the nucleus were examined by fluorescence microscopy and Hoechst staining. Cell motility was detected by Transwell assay. ELISA was used to assess the protein levels of autocrine motility factor (AMF) in the cell supernatant, and protein expression was determined by Western blotting. Our results showed that EVO inhibited the proliferation of HCT-116 cells, caused accumulation of cells in S and G2/M phases, and reduced the levels of the secreted form of AMF. The protein levels of tumor suppressor protein (p53), Bcl-2 Associated X protein (Bax), B cell CLL/lymphoma-2 (Bcl-2), phosphoglucose isomerase (PGI), phosphorylated signal transducers and activators of transcription 3 (p-STAT3) and matrix metalloproteinase 3 (MMP3) were altered in cells treated with EVO. Taken together, our results suggest that EVO modulates the activity of the p53 signaling pathway to induce apoptosis and downregulate MMP3 expression by inactivating the JAK2/STAT3 pathway through the downregulation of PGI to inhibit migration of HCT-116 human colorectal cancer cells.


Subject(s)
Antineoplastic Agents, Phytogenic/pharmacology , Apoptosis/drug effects , Cell Movement/drug effects , Plant Extracts/pharmacology , Quinazolines/pharmacology , Cell Cycle Checkpoints/drug effects , Cell Cycle Proteins/genetics , Cell Cycle Proteins/metabolism , Cell Proliferation/drug effects , Colorectal Neoplasms/genetics , Colorectal Neoplasms/metabolism , Colorectal Neoplasms/pathology , Gene Expression Regulation, Neoplastic/drug effects , HCT116 Cells , Humans , Janus Kinase 2/antagonists & inhibitors , Protein Kinase Inhibitors/pharmacology , Signal Transduction/drug effects
20.
J Agric Food Chem ; 62(1): 167-72, 2014 Jan 08.
Article in English | MEDLINE | ID: mdl-24308429

ABSTRACT

Sweet potato leaves contain the highest levels of functional polyphenols. In this study the effects of the sweet potato leaf extract and its contents, such as mono (3, 4, and 5)-caffeoylquinic acid (CQA), di-CQA (4,5-diCQA, 3,5-diCQA, and 3,4-diCQA) and caffeic acid (CA), were evaluated on the ß-catenin/Tcf-4 signaling in human colorectal cancer HCT116 cells. The extract and the CQA derivatives inhibited the ß-catenin/Tcf-4 signaling, and the inhibition of the di-CQA (with two caffeoyl groups) was higher than that of the mono-CQA (one-caffeoyl group) and CA, suggesting that the caffeoyl structure in the presence of a catechol group plays a significant role in interfering with the ß-catenin/Tcf-4 signaling. In addition, the CQA derivatives had no effect on the ß-catenin protein expression, but all test compounds inhibited the expression of the Tcf-4 transcription, and the inhibition of the di-CQA derivatives was stronger than those of the mono-CQA derivatives as well as the ß-catenin/Tcf-4 transcriptional activity. These compounds can modulate the downstream Wnt signaling pathway, suggesting that sweet potato leaves can be a protective food for colorectal cancer.


Subject(s)
Basic Helix-Loop-Helix Leucine Zipper Transcription Factors/genetics , Down-Regulation/drug effects , Ipomoea batatas/chemistry , Plant Extracts/pharmacology , Quinic Acid/analogs & derivatives , Transcription Factors/genetics , beta Catenin/metabolism , Basic Helix-Loop-Helix Leucine Zipper Transcription Factors/metabolism , HCT116 Cells , Humans , Plant Leaves/chemistry , Quinic Acid/pharmacology , Signal Transduction/drug effects , Transcription Factor 4 , Transcription Factors/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL