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.
Am J Chin Med ; 48(5): 1179-1202, 2020.
Article in English | MEDLINE | ID: mdl-32668972

ABSTRACT

Over-expression of calpains in tumor tissues can be associated with cancer progression. Thus, inhibition of calpain activity using specific inhibitors has become a novel approach to control tumor growth. In this study, the anticancer potential of cryptotanshinone in combination with calpain inhibitor had been investigated in colon cancer cells and tumor xenograft. Cryptotanshinone elicited an initial endoplasmic reticular (ER) stress response, whereas prolonged stress would result in the promotion of apoptosis. It was then discovered that cryptotanshinone could cause rapid and sustained increase in cytosolic calcium in colon cancer cells accompanied by early GRP78 overexpression, which could be attenuated by pre-treatment of the calcium chelator BAPTA-AM. Cryptotanshinone also facilitated an early increase in calpain activity, which could be blocked by BAPTA-AM or the calpain inhibitor PD150606. A dynamic interaction between GRP78 and calpain during the action of cryptotanshinone was unveiled. This together with the altered NF-[Formula: see text]B signaling could be abolished by calpain inhibitor. GRP78 knockdown increased the sensitivity of cancer cells to cryptotanshinone-evoked apoptosis and reduction of cancer cell colony formation. Such sensitization of drug action had been confirmed to be p53-dependent by using p53-mutated (HT-29) and p53-deficient (HCT116 p53-∕-) cells. The synergistic antitumor effect of cryptotanshinone and calpain inhibitor was further exhibited in vivo. Taken together, findings in this study exemplify a new chemotherapeutic regimen comprising cryptotanshinone and calpain inhibitor by regulation of calpain and calcium homeostasis. This has provided us with new insights in the search of a potential target-specific neoadjuvant therapy against colon cancer.


Subject(s)
Antineoplastic Agents, Phytogenic , Apoptosis/drug effects , Apoptosis/genetics , Calcium/metabolism , Calpain/metabolism , Colonic Neoplasms/metabolism , Colonic Neoplasms/pathology , Homeostasis/drug effects , Phenanthrenes/pharmacology , Phenanthrenes/therapeutic use , Phytotherapy , Tumor Suppressor Protein p53/metabolism , Animals , Calpain/genetics , Colonic Neoplasms/genetics , Endoplasmic Reticulum Chaperone BiP , Endoplasmic Reticulum Stress/drug effects , Gene Expression/drug effects , Heat-Shock Proteins/genetics , Heat-Shock Proteins/metabolism , Humans , Mice, Nude , Tumor Cells, Cultured
2.
Phytomedicine ; 68: 153177, 2020 Mar.
Article in English | MEDLINE | ID: mdl-32106002

ABSTRACT

BACKGROUND: Calycosin is a bioactive isoflavonoid of the medicinal plant Astragalus membranaceus that exhibits a wide range of pharmacological properties. In the present study, we have attempted to explore the anti-tumorigenic potential of calycosin in pancreatic cancer. METHODS: MTT assay was used to determine cancer cell viability. Cell cycle analysis and detection of apoptosis were performed using flow cytometry. A wound healing assay was employed to study the migratory activity of cancer cells. Western blotting and RT-PCR were used to explore the mechanism by assessing the target proteins and genes. An orthotopic tumor xenograft mouse model was also used to study the drug effects in vivo. RESULTS: Calycosin inhibited the growth of pancreatic cancer cells by inducing p21Waf1/Cip1-induced cell cycle arrest and caspase-dependent apoptosis. Alternatively, it also promoted MIA PaCa-2 cell migration by eliciting epithelial-mesenchymal transition (EMT) and matrix metalloproteinase activation. In vivo study has confirmed that calycosin would provoke the pro-invasive and angiogenic drive and subsequent EMT in pancreatic tumors. Further mechanistic study suggests that induction of the Raf/MEK/ERK pathway and facilitated polarization of M2 tumor-associated macrophage in the tumor microenvironment both contribute to the pro-metastatic potential of calycosin. These events appear to be associated with increased expression of TGF-ß1 at both transcriptional and post-translational levels, which may explain the paradoxical drug actions since TGF-ß has been implicated to play dual roles as both tumor suppressor and tumor promoter in pancreatic cancer development. CONCLUSION: Findings of this study provide innovative insights about the impact of calycosin in pancreatic cancer progression through induction of cell cycle arrest and apoptosis while possessing certain tumor-promoting property by modulation of the tumor microenvironment.


Subject(s)
Isoflavones/pharmacology , Pancreatic Neoplasms/drug therapy , Pancreatic Neoplasms/pathology , Transforming Growth Factor beta/metabolism , Animals , Apoptosis/drug effects , Cell Cycle Checkpoints/drug effects , Cell Line, Tumor , Cell Movement/drug effects , Cell Survival/drug effects , Epithelial-Mesenchymal Transition/drug effects , Humans , Isoflavones/adverse effects , Male , Mice , Mice, Inbred C57BL , Pancreatic Neoplasms/metabolism , RAW 264.7 Cells , Transforming Growth Factor beta1/genetics , Transforming Growth Factor beta1/metabolism , Tumor Microenvironment/drug effects , Xenograft Model Antitumor Assays
3.
Phytomedicine ; 74: 152761, 2020 Aug.
Article in English | MEDLINE | ID: mdl-31005370

ABSTRACT

BACKGROUND: Ginseng has therapeutic potential for treating obesity and the associated gut microbiota dysbiosis. However, whether white ginseng and red ginseng, the two kinds of commonly used processed ginseng, possess different anti-obesity effects remains unknown. PURPOSE: Anti-obesity effects of water extracts of white ginseng and red ginseng (WEWG and WERG) were compared, and the potential mechanisms were discussed. METHODS: Chemical profiles of WEWG and WERG were characterized by ultra-high performance liquid chromatography-tandem triple quadrupole mass spectrometry (UHPLC-QqQ-MS/MS) and high performance liquid chromatography coupled with evaporative light scattering detector (HPLC-ELSD). Anti-obesity effects of WEWG/WERG were examined by determining fat accumulation, systemic inflammation, enteric metabolic disorders and gut microbiota dysbiosis in high-fat diet (HFD)-fed obese mice. RESULTS: Both WEWG and WERG exerted anti-obesity effects, with WEWG stronger than WERG. Compared to WERG, WEWG contained less contents of carbohydrates (polysaccharides, oligosaccharides, free monosaccharides) and ginsenosides, but chemical structures or compositions of these components in WEWG were characteristic, i.e. narrower molecular weight distribution and higher molar ratios of glucose residues of polysaccharides; higher content ratios of oligosaccharides DP2-3 (di-/tri-saccharides)-to-oligosaccharides DP4-7 (tetra-/penta-/hexa-/hepta-saccharides), sucrose-to-melibiose, maltose-to-trehalose and high-polar-to-low-polar ginsenosides. WEWG better ameliorated fat accumulation, enteric metabolic disorders and gut microbiota dysbiosis in HFD-fed obese mice than WERG. CONCLUSION: The stronger anti-obesity effect of white ginseng appears to correlate with differences in its chemical profile as compared to red ginseng. The carbohydrates and ginsenosides in WEWG potentially present more structural and compositional specificity to the obesity-associated gut bacteria, allowing more beneficial effects of WEWG on the gut microbiota dysbiosis. This consequently better alleviates the enteric metabolic disorders and systemic inflammation, thereby contributing to the stronger anti-obesity effect of WEWG as compared to WERG.


Subject(s)
Anti-Obesity Agents/pharmacology , Dysbiosis/drug therapy , Gastrointestinal Microbiome/drug effects , Obesity/drug therapy , Panax/chemistry , Animals , Anti-Obesity Agents/chemistry , Carbohydrates/analysis , Chromatography, High Pressure Liquid , Diet, High-Fat/adverse effects , Dysbiosis/microbiology , Gastrointestinal Microbiome/physiology , Ginsenosides/analysis , Male , Mice, Inbred BALB C , Obesity/microbiology , Plant Extracts/chemistry , Plant Extracts/pharmacology , Polysaccharides/analysis , Species Specificity , Tandem Mass Spectrometry
4.
Cell Physiol Biochem ; 36(6): 2433-46, 2015.
Article in English | MEDLINE | ID: mdl-26279445

ABSTRACT

BACKGROUND: Eruberin A (2, 3-dehydroflavonoid), a flavanol glycoside isolated from Pronephrium penangianum, has been used as a blood-nourishing folk medicine for centuries; however, it indeed possesses a variety of other health-promoting benefits including anti-fibrotic bioactivity. Activation of pancreatic stellate cells (PSCs) is the key initiating step in pancreatic fibrosis, which is a characteristic feature associated with chronic pancreatitis and pancreatic adenocarcinoma. METHODS: The anti-fibrotic effect of eruberin A and the underlying mechanisms of its anti-fibrotic action in LTC-14 cells, which retained essential characteristics and morphological features of primary PSCs, were examined by means of real-time polymerase chain reactions, Western blotting and immunostaining. RESULTS: The application of eruberin A (20 µg/ml) effectively inhibited the expression levels of fibrotic mediators namely alpha-smooth muscle actin, fibronectin and type I-collagen, so as the sonic hedgehog signaling pathway components post transforming growth factor-beta (5 ng/ml) stimulation. Eruberin A treatment also led to a notable decrease in the activation of nuclear factor-kappaB (NF-κB) and the phosphorylation of phosphoinositide 3-kinase (PI3K)/serine-threonine kinase (AKT). CONCLUSION: Our results demonstrated that eruberin A significantly suppressed the expression levels of fibrotic mediators in PSCs, and we suggest that its anti-fibrotic mechanism was associated with an attenuation of the PI3K/AKT/NF-κB signaling pathway.


Subject(s)
Anthocyanins/pharmacology , Flavonols/pharmacology , Glycosides/pharmacology , Pancreatic Stellate Cells/pathology , Animals , Anthocyanins/chemistry , Cell Death/drug effects , Cell Line , Down-Regulation/drug effects , Fibrosis , Flavonols/chemistry , Glycosides/chemistry , Hedgehog Proteins/metabolism , Humans , Magnetic Resonance Spectroscopy , NF-kappa B/metabolism , Pancreatic Stellate Cells/drug effects , Phosphatidylinositol 3-Kinases/metabolism , Rats , Signal Transduction/drug effects , Transforming Growth Factor beta/pharmacology
5.
Oncol Rep ; 28(6): 2188-94, 2012 Dec.
Article in English | MEDLINE | ID: mdl-23023137

ABSTRACT

Formononetin is a novel herbal isoflavonoid isolated from Astragalus membranaceus, a medicinal plant that possesses antitumorigenic properties. Our previous findings demonstrated that formononetin initiates growth-inhibitory and pro-apoptotic activities in human colon cancer cells. In the present study, we aimed to further examine the potential of formononetin in controlling angiogenesis and tumor cell invasiveness in human colon cancer cells and tumor xenografts. The results showed that formononetin downregulated the expression of the key pro-angiogenic factors, including vascular endothelial growth factor (VEGF) and matrix metalloproteinases. We also discovered that the invasiveness of metastatic colon cancer cells was alleviated following drug treatment. The potential anti-angiogenic effect of formononetin was examined in nude mouse xenografts. The tumor size and the number of proliferating cells were reduced in the tumor tissues obtained from the formononetin-treated group. The serum VEGF level was also reduced in the drug-treated animals when compared to the controls. These findings suggest that formononetin inhibits angiogenesis and tumor cell invasion, and thus support its use in the treatment of advanced and metastatic colon cancers.


Subject(s)
Colonic Neoplasms/blood supply , Colonic Neoplasms/drug therapy , Isoflavones/pharmacology , Neovascularization, Pathologic/drug therapy , Phytoestrogens/pharmacology , Vascular Endothelial Growth Factor A/blood , Animals , Astragalus propinquus/chemistry , Cell Line, Tumor , Cell Movement/drug effects , Cell Proliferation/drug effects , Colonic Neoplasms/metabolism , Colonic Neoplasms/pathology , Disease Progression , Female , Humans , Mice , Mice, Inbred BALB C , Mice, Nude , Neoplasm Invasiveness , Plant Extracts/pharmacology , Plant Roots/chemistry , Vascular Endothelial Growth Factor A/biosynthesis , Xenograft Model Antitumor Assays
6.
Int J Mol Med ; 26(3): 341-9, 2010 Sep.
Article in English | MEDLINE | ID: mdl-20664949

ABSTRACT

We have previously demonstrated that the total saponins of Astragalus membranaceus (AST) possess potential anti-tumorigenic effects in human colon cancer cells and tumor xenografts. In the present study, the proapoptotic effects of AST were investigated in native and cytokine-induced HT-29 cells to further unveil its mechanism of action. Growth-inhibitory action of AST (60 microg/ml) was demonstrated in native HT-29 cells, which was exaggerated in tumor necrosis factor (TNF) (5 ng/ml)-induced cells. These were accompanied by caspase 3 activation, cleavage of poly(ADP-ribose) polymerase and a subsequent increase in apoptotic cell numbers. Furthermore, activation of procaspase 8 indicates that the extrinsic apoptotic pathway was involved, while cleavage of Bid into t-Bid implicates cross-talk with the intrinsic apoptotic pathway. Alternatively, AST caused S and G2/M phase arrest, while in cytokine-induced cells S phase arrest was predominant. Further adding to our recent suggestion on its correlation with phosphatidylinositol 3-kinase (PI3K)-Akt signaling, we have now revealed that AST caused overexpression of PTEN and down-regulation of mammalian target of rapamycin (mTOR) expression. Nevertheless, these events were preceded by a decrease in nuclear factor-kappaB (NF-kappaB)/DNA binding activity with continuous ERK 1/2 activation. Some of these effects became more intense in cytokine-induced cells. Our findings in this study suggest that AST induces the extrinsic apoptotic cascade and causes cell cycle arrest in HT-29 cells by modulation of both mTOR and ERK signaling pathways, of which inhibition of NF-kappaB is important in the latter mechanism. Most of the above processes are more pronounced in cytokine-induced cells.


Subject(s)
Apoptosis/drug effects , Astragalus Plant/chemistry , Intracellular Signaling Peptides and Proteins/metabolism , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3/metabolism , Protein Serine-Threonine Kinases/metabolism , Saponins/pharmacology , Signal Transduction/drug effects , Caspase 8/metabolism , Enzyme Activation , HT29 Cells , Humans , NF-kappa B/antagonists & inhibitors , NF-kappa B/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Protein Precursors/metabolism , Saponins/chemistry , Signal Transduction/physiology , TOR Serine-Threonine Kinases , Tumor Necrosis Factor-alpha/metabolism
7.
Invest New Drugs ; 28(1): 1-13, 2010 Feb.
Article in English | MEDLINE | ID: mdl-19139819

ABSTRACT

Formononetin is a novel herbal isoflavonoid isolated from Astragalus membranaceus, a medicinal plant that possesses antitumorigenic property. We attempted to compare the anticarcinogenic mechanism of formononetin with that of the known proapoptotic flavonoid isoliquiritigenin (ISL) in human cancer cells. We first evaluated the effects of formononetin and ISL on HCT 116 colon cancer cell viability. Immunofluorescence staining was then performed to observe the morphological changes of cancer cells undergoing apoptosis, which had been substantiated using Annexin V-FITC/propidium iodide staining. Western immunoblotting and flow cytometry were also employed to study parameters associated with apoptosis and cell proliferation. Our data show that formononetin and ISL both inhibited the growth of colon cancer cells and promoted apoptosis. These processes were accompanied by caspase activation and downregulation of the antiapoptotic proteins Bcl-2 and Bcl-x(L). Besides, the novel proapoptotic protein NSAID-activated gene (NAG-1) and its upstream regulator were overexpressed in drug-treated cells. Nevertheless, only ISL was found to induce a G2 arrest. These findings exemplify that both formononetin and ISL could cause growth inhibition and facilitate apoptosis in colon cancer cells, while only ISL is capable of inducing phase-specific cell cycle arrest. This suggests that the anticarcinogenic activities of different herbal flavonoids may involve both common and differential mechanisms of action, which could be developed as potential anticancer drugs.


Subject(s)
Apoptosis/drug effects , Astragalus propinquus/chemistry , Cell Cycle/drug effects , Chalcones/pharmacology , Colonic Neoplasms/pathology , Flavonoids/pharmacology , Isoflavones/pharmacology , Apoptosis Regulatory Proteins/metabolism , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Shape/drug effects , Chalcones/chemistry , Down-Regulation/drug effects , Drug Screening Assays, Antitumor , Flavonoids/chemistry , G2 Phase/drug effects , Humans , Isoflavones/chemistry , Phytotherapy
8.
Int J Mol Med ; 24(4): 571-7, 2009 Oct.
Article in English | MEDLINE | ID: mdl-19724899

ABSTRACT

Conventional chemotherapy of liver cancer fails to provide satisfactory remission and may cause serious side effects, thus it is crucial to derive alternative treatments that effectively inhibit cancer cell growth with known mechanisms of action. In the present study, we investigated the anti-carcinogenic effects of Coptis chinensis and its major constituent, berberine, in HepG2 hepatocellular carcinoma (HCC) cells and attempted to elucidate the underlying mechanism, including involvement of the nonsteroidal anti-inflammatory drug (NSAID)-activated gene (NAG-1). Inhibition of cell proliferation, induction of apoptosis and cell cycle arrest at the G2/M phase were observed in HepG2 cells treated with Coptis chinensis or berberine. The pro-apoptotic effects were associated with corresponding down-regulation of Bcl-2, activation of procaspase-3 and -9 as well as cleavage of poly (ADP-ribose) polymerase. We further demonstrated the involvement of NAG-1 in the pro-apoptotic events following prior activation of its upstream transcriptional factor early growth response gene (Egr-1). This was confirmed by increased NAG-1 promoter activity preceded by the elevation of Egr-1/ DNA binding activity. Our results suggest that both Coptis chinensis and berberine are potential anti-carcinogenic agents in treating HCC by inducing cell cycle arrest and promotion of apoptosis, while NAG-1 is a molecular target during the drug-induced pro-apoptotic action in HepG2 cells.


Subject(s)
Anticarcinogenic Agents/therapeutic use , Berberine/therapeutic use , Carcinoma, Hepatocellular/drug therapy , Coptis/chemistry , Drugs, Chinese Herbal/therapeutic use , Growth Differentiation Factor 15/metabolism , Liver Neoplasms/drug therapy , Anticarcinogenic Agents/chemistry , Berberine/chemistry , Blotting, Western , Cell Cycle/drug effects , Cell Line, Tumor , Cell Proliferation/drug effects , Chromatography, High Pressure Liquid , Drugs, Chinese Herbal/chemistry , Early Growth Response Protein 1/metabolism , Electrophoretic Mobility Shift Assay , Flow Cytometry , Humans , Poly(ADP-ribose) Polymerases/metabolism , Proto-Oncogene Proteins c-bcl-2/metabolism
9.
Int J Mol Med ; 23(2): 189-96, 2009 Feb.
Article in English | MEDLINE | ID: mdl-19148542

ABSTRACT

Astragalus membranaceus has been used to ameliorate the side effects of anti-neoplastic drugs. We recently reported that total Astragalus saponins (AST) possess anti-tumor properties in human colon cancer cells and tumor xenografts. Nevertheless, the precise mechanism of action has not been fully elucidated. The present study aimed to unveil the anti-carcinogenic potential of AST in HepG2 human hepatocellular carcinoma (HCC) cells and to clarify the signaling pathway. We demonstrated here that AST downregulated expression of the HCC tumor marker alpha-fetoprotein and suppressed HepG2 cell growth by inducing apoptosis. AST also caused caspase activation, poly(ADP-ribose) polymerase (PARP) cleavage, nuclear chromatin condensation, with downregulation of the anti-apoptotic proteins bcl-2 and bcl-xL and decreased nuclear factor-kappa B (NF-kappaB)/DNA-binding activity. Concomitantly, expression of the phosphorylated form of the extracellular signal-regulated protein kinase (ERK) was prominently increased. Nevertheless, pretreatment of ERK inhibitor PD98059 did not attenuate AST-induced PARP cleavage. Taken together, these results exemplify that AST induced growth inhibition and promoted apoptosis in HepG2 cells through modulation of an ERK-independent NF-kappaB signaling pathway.


Subject(s)
Apoptosis , Astragalus Plant/chemistry , Extracellular Signal-Regulated MAP Kinases/metabolism , NF-kappa B/metabolism , Saponins/toxicity , Signal Transduction , Carcinoma, Hepatocellular/metabolism , Cell Line, Tumor , Down-Regulation , Humans , Liver Neoplasms/metabolism , Nitric Oxide Synthase Type II/metabolism , alpha-Fetoproteins/genetics , alpha-Fetoproteins/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL