Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 31
Filter
Add more filters










Publication year range
1.
Biomater Sci ; 11(13): 4522-4536, 2023 Jun 27.
Article in English | MEDLINE | ID: mdl-37158091

ABSTRACT

Aging involves tissue and cell potential dysfunction characterized by stem cell senescence and extracellular matrix microenvironment (ECM) alteration. Chondroitin sulfate (CS), found in the ECM of normal cells and tissues, aids in maintaining tissue homeostasis. Here, CS-derived biomaterial (CSDB) from sturgeon is extracted to investigate its antiaging effect in senescence-accelerated mouse prone-8 (SAMP8) mice and elucidate the underlying mechanism of its action. Although CSDB has been widely extracted from different sources and used as a scaffold, hydrogel, or drug carrier for the treatment of various pathological diseases, CSDB has not yet been used as a biomaterial for the amelioration of senescence and aging features. In this study, the extracted sturgeon CSDB showed a low molecular weight and comprised 59% 4-sulfated CS and 23% 6-sulfated CS. In an in vitro study, sturgeon CSDB promoted cell proliferation and reduced oxidative stress to inhibit stem cell senescence. In an ex vivo study, after oral CSDB treatment of SAMP8 mice, the stem cells were extracted to analyze the p16Ink4a and p19Arf gene-related pathways, which were inhibited and then SIRT-1 gene expression was upregulated to reprogram stem cells from a senescence state for retarding aging. In an in vivo study, CSDB also restored the aging-phenotype-related bone mineral density and skin morphology to prolong longevity. Thus, sturgeon CSDB may be useful for prolonging healthy longevity as an anti-aging drug.


Subject(s)
Antioxidants , Longevity , Mice , Animals , Chondroitin Sulfates/pharmacology , Aging/genetics , Cellular Senescence , Fishes/genetics , Stem Cells , Gene Expression
2.
Mar Drugs ; 20(8)2022 Jul 27.
Article in English | MEDLINE | ID: mdl-36005485

ABSTRACT

Overexpressed EGFR and mutant K-Ras play vital roles in therapeutic resistance in colorectal cancer patients. To search for an effective therapeutic protocol is an urgent task. A secondary metabolite in the sponge Hippospongia sp., Heteronemin, has been shown to induce anti-proliferation in several types of cancers. A thyroxine-deaminated analogue, tetrac, binds to integrin αvß3 to induce anti-proliferation in different cancers. Heteronemin- and in combination with tetrac-induced antiproliferative effects were evaluated. Tetrac enhanced heteronemin-induced anti-proliferation in HT-29 cells (KRAS WT CRC) and HCT-116 cells (KRAS MT CRC). Heteronemin and tetrac arrested cell cycle in different phases. Combined treatment increased the cell accumulation in sub-G1 and S phases. The combined treatment also induced the inactivation of EGFR signaling and downregulated the phosphorylated ERK1/2 protein in both cell lines. Heteronemin and the combination showed the downregulation of the phosphorylated and total PI3K protein in HT-29 cells (KRAS WT CRC). Results by NanoString technology and RT-qPCR revealed that heteronemin and combined treatment suppressed the expression of EGFR and downstream genes in HCT-116 cells (KRAS MT CRC). Heteronemin or combined treatment downregulated genes associated with cancer progression and decreased cell motility. Heteronemin or the combined treatment suppressed PD-L1 expression in both cancer cell lines. However, only tetrac and the combined treatment inhibited PD-L1 protein accumulation in HT-29 cells (KRAS WT CRC) and HCT-116 cells (KRAS MT CRC), respectively. In summary, heteronemin induced anti-proliferation in colorectal cancer cells by blocking the EGFR-dependent signal transduction pathway. The combined treatment further enhanced the anti-proliferative effect via PD-L1 suppression. It can be an alternative strategy to suppress mutant KRAS resistance for anti-EGFR therapy.


Subject(s)
Colorectal Neoplasms , Thyroxine , B7-H1 Antigen/metabolism , Cell Line, Tumor , Cell Proliferation , Colorectal Neoplasms/metabolism , ErbB Receptors/metabolism , Humans , Mutation , Proto-Oncogene Proteins p21(ras)/genetics , Proto-Oncogene Proteins p21(ras)/metabolism , Proto-Oncogene Proteins p21(ras)/pharmacology , Signal Transduction , Terpenes , Thyroxine/analogs & derivatives
3.
Front Cell Dev Biol ; 10: 829788, 2022.
Article in English | MEDLINE | ID: mdl-35237605

ABSTRACT

Doxycycline, an antibiotic, displays the inhibition of different signal transduction pathways, such as anti-inflammation and anti-proliferation, in different types of cancers. However, the anti-cancer mechanisms of doxycycline via integrin αvß3 are incompletely understood. Integrin αvß3 is a cell-surface anchor protein. It is the target for estrogen, androgen, and thyroid hormone and plays a pivotal role in the proliferation, migration, and angiogenic process in cancer cells. In our previous study, thyroxine hormones can interact with integrin αvß3 to activate the extracellular signal-regulated kinase 1/2 (ERK1/2), and upregulate programmed death-ligand 1 (PD-L1) expression. In the current study, we investigated the inhibitory effects of doxycycline on proliferation in two breast cancer cell lines, MCF-7 and MDA-MB-231 cells. Doxycycline induces concentration-dependent anti-proliferation in both breast cancer cell lines. It regulates gene expressions involved in proliferation, pro-apoptosis, and angiogenesis. Doxycycline suppresses cell cyclin D1 (CCND1) and c-Myc which play crucial roles in proliferation. It also inhibits PD-L1 gene expression. Our findings show that modulation on integrin αvß3 binding activities changed both thyroxine- and doxycycline-induced signal transductions by an integrin αvß3 inhibitor (HSDVHK-NH2). Doxycycline activates phosphorylation of focal adhesion kinase (FAK), a downstream of integrin, but inhibits the ERK1/2 phosphorylation. Regardless, doxycycline-induced FAK phosphorylation is blocked by HSDVHK-NH2. In addition, the specific mechanism of action associated with pERK1/2 inhibition via integrin αvß3 is unknown for doxycycline treatment. On the other hand, our findings indicated that inhibiting ERK1/2 activation leads to suppression of PD-L1 expression by doxycycline treatment. Furthermore, doxycycline-induced gene expressions are disturbed by a specific integrin αvß3 inhibitor (HSDVHK-NH2) or a mitogen-activated protein kinase (MAPK)/extracellular signal-regulated kinases (ERK) kinase (MAPK/ERK, MEK) inhibitor (PD98059). The results imply that doxycycline may interact with integrin αvß3 and inhibits ERK1/2 activation, thereby regulating cell proliferation and downregulating PD-L1 gene expression in estrogen receptor (ER)-negative breast cancer MDA-MB-231 cells.

4.
Polymers (Basel) ; 13(11)2021 May 24.
Article in English | MEDLINE | ID: mdl-34073693

ABSTRACT

Osteoconduction is an important consideration for fabricating bio-active materials for bone regeneration. For years, hydroxyapatite and ß-calcium triphosphate (ß-TCP) have been used to develop bone grafts for treating bone defects. However, this material can be difficult to handle due to filling material sagging. High molecular weight hyaluronic acid (H-HA) can be used as a carrier to address this problem and improve operability. However, the effect of H-HA on bone formation is still controversial. In this study, low molecular weight hyaluronic acid (L-HA) was fabricated using gamma-ray irradiation. The viscoelastic properties and chemical structure of the fabricated hybrids were evaluated by a rheological analysis nuclear magnetic resonance (NMR) spectrum. The L-MH was mixed with H-HA to produce H-HA/L-HA hybrids at ratios of 80:20, 50:50 and 20:80 (w/w). These HA hybrids were then combined with hydroxyapatite and ß-TCP to create a novel bone graft composite. For animal study, artificial bone defects were prepared in rabbit femurs. After 12 weeks of healing, the rabbits were scarified, and the healing statuses were observed and evaluated through micro-computer tomography (CT) and tissue histological images. Our viscoelastic analysis showed that an HA hybrid consisting 20% H-HA is sufficient to maintain elasticity; however, the addition of L-HA dramatically decreases the dynamic viscosity of the HA hybrid. Micro-CT images showed that the new bone formations in the rabbit femur defect model treated with 50% and 80% L-HA were 1.47 (p < 0.05) and 2.26 (p < 0.01) times higher than samples filled with HA free bone graft. In addition, a similar tendency was observed in the results of HE staining. These results lead us to suggest that the material with an H-HA/L-HA ratio of 50:50 exhibited acceptable viscosity and significant new bone formation. Thus, it is reasonable to suggest that it may be a potential candidate to serve as a supporting system for improving the operability of granular bone grafts and enhancing new bone formations.

5.
Int J Nanomedicine ; 16: 3789-3802, 2021.
Article in English | MEDLINE | ID: mdl-34103915

ABSTRACT

INTRODUCTION: It has been reported that low-molecular-weight hyaluronic acid (LMWHA) exhibits a potentially beneficial effect on cancer therapy through targeting of CD44 receptors on tumor cell surfaces. However, its applicability towards tumor detection is still unclear. In this regard, LMWHA-conjugated iron (Fe3O4) nanoparticles (LMWHA-IONPs) were prepared in order to evaluate its application for enhancing the T2* weighted MRI imaging sensitivity for tumor detection. METHODS: LMWHA and Fe3O4 NPs were produced using γ-ray irradiation and chemical co-precipitation methods, respectively. First, LMWHA-conjugated FITC was prepared to confirm the ability of LMWHA to target U87MG cells using fluorescence microscopy. The hydrodynamic size distribution and dispersion of the IONPs and prepared LMWHA-IONPs were analyzed using dynamic light scattering (DLS). In addition, cell viability assays were performed to examine the biocompatibility of LMWHA and LMWHA-IONPs toward U87MG human glioblastoma and NIH3T3 fibroblast cell lines. The ability of LMWHA-IONPs to target tumor cells was confirmed by detecting iron (Fe) ion content using the thiocyanate method. Finally, time-of-flight secondary ion mass spectrometry (TOF-SIMS) imaging and in vitro magnetic resonance imaging (MRI) were performed to confirm the contrast enhancement effect of LMWHA-IONPs. RESULTS: Florescence analysis results showed that LMWHA-FITC successfully targeted the surfaces of both tested cell types. The ability of LMWHA to target U87MG cells was higher than for NIH3T3 cells. Cell viability experiments showed that the fabricated LMWHA-IONPs possessed good biocompatibility for both cell lines. After co-culturing test cells with the LMWHA-IONPs, detected Fe ion content in the U87MG cells was much higher than that of the NIH3T3 cells in both thiocyanate assays and TOF-SIMs images. Finally, the addition of LMWHA-IONPs to the U87MG cells resulted in an obvious improvement in T2* weighted MR image contrast compared to control NIH3T3 cells. DISCUSSION: Overall, the present results suggest that LMWHA-IONPs fabricated in this study provide an effective MRI contrast agent for improving the diagnosis of early stage glioblastoma in MRI examinations.


Subject(s)
Gamma Rays , Glioblastoma/diagnostic imaging , Hyaluronic Acid/chemistry , Iron/chemistry , Magnetic Resonance Imaging , Metal Nanoparticles/chemistry , Animals , Cell Line, Tumor , Cell Proliferation , Cell Survival/drug effects , Glioblastoma/pathology , Humans , Hyaluronic Acid/ultrastructure , Metal Nanoparticles/ultrastructure , Mice , Molecular Weight , NIH 3T3 Cells , Oleic Acid/chemistry , Particle Size
6.
Phytochemistry ; 187: 112776, 2021 Jul.
Article in English | MEDLINE | ID: mdl-33933828

ABSTRACT

Transformation of 15-ene steviol (ent-13-hydroxy-kaur-15-en-19-oic acid) by growth cultures of Aspergillus niger BCRC 32720, Cunninghamella bainieri ATCC 9244, and Mortierella isabellina ATCC 38063 was conducted to generate various derivatives for the development of bioactive compounds. Four previously undescribed compounds along with six known compounds were obtained. The newly identified isolates were characterized using 1D and 2D NMR, IR, and HRESIMS, and three compounds were further confirmed by X-ray crystallographic analyses. Subsequently, the effects of 15-ene steviol and its derivatives on lipopolysaccharide (LPS)-induced cytokine production by THP-1 cells were examined, with dexamethasone used as a positive control. Results indicated that most of the tested compounds showed lower inhibitory effects than those detected in the dexamethasone-treated group, except that 15-ene steviol showed better effects than dexamethasone on the reduction of LPS-induced monocyte chemoattractant protein (MCP)-1, -2, and -3 release. Three specialized products similarly showed better effects than dexamethasone on the inhibition of LPS-induced secretion of regulated on activation, normal T cell expressed and secreted (RANTES). Moreover, none of the tested compounds showed any cytotoxicity or triggered cell apoptosis, and none affected the protein integrity of toll-like receptor 4 (TLR4) or MyD88, suggesting that these compounds may exert the anti-inflammatory activity downstream of membrane-associated TLR4 and MyD88 molecules.


Subject(s)
Cunninghamella , Aspergillus niger , Diterpenes, Kaurane , Fungi
7.
Cells ; 9(8)2020 08 03.
Article in English | MEDLINE | ID: mdl-32756527

ABSTRACT

The property of drug-resistance may attenuate clinical therapy in cancer cells, such as chemoresistance to gefitinib in colon cancer cells. In previous studies, overexpression of PD-L1 causes proliferation and metastasis in cancer cells; therefore, the PD-L1 pathway allows tumor cells to exert an adaptive resistance mechanism in vivo. Nano-diamino-tetrac (NDAT) has been shown to enhance the anti-proliferative effect induced by first-line chemotherapy in various types of cancer, including colorectal cancer (CRC). In this work, we attempted to explore whether NDAT could enhance the anti-proliferative effect of gefitinib in CRC and clarified the mechanism of their interaction. The MTT assay was utilized to detect a reduction in cell proliferation in four primary culture tumor cells treated with gefitinib or NDAT. The gene expression of PD-L1 and other tumor growth-related molecules were quantified by quantitative polymerase chain reaction (qPCR). Furthermore, the identification of PI3K and PD-L1 in treated CRC cells were detected by western blotting analysis. PD-L1 presentation in HCT116 xenograft tumors was characterized by specialized immunohistochemistry (IHC) and the hematoxylin and eosin stain (H&E stain). The correlations between the change in PD-L1 expression and tumorigenic characteristics were also analyzed. (3) The PD-L1 was highly expressed in Colo_160224 rather than in the other three primary CRC cells and HCT-116 cells. Moreover, the PD-L1 expression was decreased by gefitinib (1 µM and 10 µM) in two cells (Colo_150624 and 160426), but 10 µM gefitinib stimulated PD-L1 expression in gefitinib-resistant primary CRC Colo_160224 cells. Inactivated PI3K reduced PD-L1 expression and proliferation in CRC Colo_160224 cells. Gefitinib didn't inhibit PD-L1 expression and PI3K activation in gefitinib-resistant Colo_160224 cells. However, NDAT inhibited PI3K activation as well as PD-L1 accumulation in gefitinib-resistant Colo_160224 cells. The combined treatment of NDAT and gefitinib inhibited pPI3K and PD-L1 expression and cell proliferation. Additionally, NDAT reduced PD-L1 accumulation and tumor growth in the HCT116 (K-RAS mutant) xenograft experiment. (4) Gefitinib might suppress PD-L1 expression but did not inhibit proliferation through PI3K in gefitinib-resistant primary CRC cells. However, NDAT not only down-regulated PD-L1 expression via blocking PI3K activation but also inhibited cell proliferation in gefitinib-resistant CRCs.


Subject(s)
B7-H1 Antigen/genetics , Cell Proliferation/drug effects , Colorectal Neoplasms/pathology , Drug Resistance, Neoplasm/drug effects , Gefitinib/pharmacology , Phosphatidylinositol 3-Kinases/metabolism , Polyglactin 910/pharmacology , Thyroxine/analogs & derivatives , Animals , Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use , B7-H1 Antigen/metabolism , Colorectal Neoplasms/drug therapy , Drug Resistance, Neoplasm/genetics , Gefitinib/therapeutic use , Gene Expression Regulation, Neoplastic/drug effects , HCT116 Cells , HT29 Cells , Humans , Mice , Phosphoinositide-3 Kinase Inhibitors/pharmacology , Phosphoinositide-3 Kinase Inhibitors/therapeutic use , Polyglactin 910/therapeutic use , Thyroxine/pharmacology , Thyroxine/therapeutic use , Tumor Cells, Cultured , Xenograft Model Antitumor Assays
8.
Food Chem Toxicol ; 133: 110808, 2019 Nov.
Article in English | MEDLINE | ID: mdl-31499123

ABSTRACT

The obesity-regulated gene, leptin, is essential for diet. Leptin resistance causes obesity and related diseases. Certain types of diet are able to decrease leptin resistance. However, leptin has been shown to be correlated with inflammation and stimulate proliferation of various cancers. Two synthetic leptin derivatives (mimetics), OB3 and [D-Leu-4]-OB3, show more effective than leptin in reducing obesity and diabetes in mouse models. OB3 inhibits leptin-induced proliferation in ovarian cancer cells. However, effects of these mimetics in hepatocellular carcinoma (HCC) have not been investigated. In the present study, we examined the effects of OB3 and [D-Leu-4]-OB3 on cell proliferation and gene expressions in human HCC cell cultures. In contrast to what was reported for leptin, OB3 and [D-Leu-4]-OB3 reduced cell proliferation in hepatomas. Both OB3 and [D-Leu-4]-OB3 stimulated expression of pro-apoptotic genes. Both compounds also inhibited expressions of pro-inflammatory, proliferative and metastatic genes and PD-L1 expression. In combination with leptin, OB3 inhibited leptin-induced cell proliferation and expressions of pro-inflammation-, and proliferation-related genes. Furthermore, the OB3 peptide inhibited phosphoinositide 3-kinase (PI3K) activation which is essential for leptin-induced proliferation in HCC. These results indicate that OB3 and [D-Leu-4]-OB3 may have the potential to reduce leptin-related inflammation and proliferation in HCC cells.


Subject(s)
Cell Proliferation/drug effects , Gene Expression/drug effects , Leptin/pharmacology , Peptide Fragments/pharmacology , Signal Transduction/drug effects , Cell Line, Tumor , G1 Phase Cell Cycle Checkpoints/drug effects , Humans , Intercellular Signaling Peptides and Proteins/genetics , Intracellular Signaling Peptides and Proteins/genetics , Phosphatidylinositol 3-Kinase/metabolism , Protein Kinase Inhibitors/pharmacology
9.
Food Chem Toxicol ; 132: 110693, 2019 Oct.
Article in English | MEDLINE | ID: mdl-31336132

ABSTRACT

Thyroid hormone, L-thyroxine (T4), induces inflammatory genes expressions and promotes cancer growth. It also induces expression of the checkpoint programmed death-ligand 1 (PD-L1), which plays a vital role in cancer progression. On the other hand, resveratrol inhibits inflammatory genes expressions. Moreover, resveratrol increases nuclear inducible cyclooxygenase (COX)-2 accumulation, complexes with p53, and induces p53-dependent anti-proliferation. In this study, we investigated the effect of T4 on resveratrol-induced anti-proliferation in oral cancer. T4 increased the expression and cytoplasmic accumulation of PD-L1. Increased expressions of pro-inflammatory genes, interleukin (IL)-1ß and transforming growth factor (TGF)-ß1, were shown to stimulate PD-L1 expression. T4 stimulated pro-inflammatory and proliferative genes expressions, and oral cancer cells proliferation. In contrast, resveratrol inhibited those genes and activated anti-proliferative genes. T4 retained resveratrol-induced COX-2 in cytoplasm and prevented COX-2 nuclear accumulation when resveratrol treated cancer cells. A specific signal transducer and activator of transcription 3 (STAT3) inhibitor, S31-201, blocked T4-induced inhibition and restored resveratrol-induced nuclear COX-2 accumulation. By inhibiting the T4-activated STAT3 signal transduction axis with S31-201, resveratrol was able to sequentially reestablish COX-2/p53-dependent gene expressions and anti-proliferation. These findings provide a novel understanding of the inhibitory effects of T4 on resveratrol-induced anticancer properties via the sequential expression of PD-L1 and inflammatory genes.


Subject(s)
Cell Proliferation/drug effects , Cytokines/genetics , Gene Expression/drug effects , Inflammation Mediators/metabolism , Mouth Neoplasms/pathology , Resveratrol/pharmacology , Thyroxine/pharmacology , Cell Line, Tumor , Cell Nucleus/drug effects , Cell Nucleus/enzymology , Cyclooxygenase 2/metabolism , Humans , Mouth Neoplasms/metabolism , STAT3 Transcription Factor/metabolism
10.
Sci Rep ; 8(1): 14587, 2018 10 01.
Article in English | MEDLINE | ID: mdl-30275489

ABSTRACT

Chronic kidney disease is a worldwide problem, and Pb contamination is a potential risk factor. Since current biomarkers are not sensitive for the diagnosis of Pb-induced nephrotoxicity, novel biomarkers are needed. Metformin has both hypoglycaemic effects and reno-protection ability. However, its mechanism of action is unknown. We aimed to discover the early biomarkers for the diagnosis of low-level Pb-induced nephrotoxicity and understand the mechanism of reno-protection of metformin. Male Wistar rats were randomly divided into control, Pb, Pb + ML, Pb + MH and MH groups. Pb (250 ppm) was given daily via drinking water. Metformin (50 or 100 mg/kg/d) was orally administered. Urine was analysed by nuclear magnetic resonance (NMR)-based metabolomics coupled with multivariate statistical analysis, and potential biomarkers were subsequently quantified. The results showed that Pb-induced nephrotoxicity was closely correlated with the elevation of 5-aminolevulinic acid, D-lactate and guanidinoacetic acid in urine. After co-treatment with metformin, 5-aminolevulinic acid and D-lactate were decreased. This is the first demonstration that urinary 5-aminolevulinic acid, D-lactate and guanidinoacetic acid could be early biomarkers of low-level Pb-induced nephrotoxicity in rats. The reno-protection of metformin might be attributable to the reduction of D-lactate excretion.


Subject(s)
Biological Factors/urine , Lead Poisoning/complications , Metabolome , Metformin/administration & dosage , Protective Agents/administration & dosage , Renal Insufficiency/chemically induced , Renal Insufficiency/prevention & control , Administration, Oral , Animals , Disease Models, Animal , Environmental Pollutants/toxicity , Lead/toxicity , Magnetic Resonance Spectroscopy , Male , Rats, Wistar , Treatment Outcome , Urinalysis
11.
Horm Cancer ; 9(6): 420-432, 2018 12.
Article in English | MEDLINE | ID: mdl-30187356

ABSTRACT

Drug resistance complicates the clinical use of gefitinib. Tetraiodothyroacetic acid (tetrac) and nano-diamino-tetrac (NDAT) have been shown in vitro and in xenografts to have antiproliferative/angiogenic properties and to potentiate antiproliferative activity of other anticancer agents. In the current study, we investigated the effects of NDAT on the anticancer activities of gefitinib in human colorectal cancer cells. ß-Galactoside α-2,6-sialyltransferase 1 (ST6Gal1) catalyzes EGFR sialylation that is associated with gefitinib resistance in colorectal cancers, and this was also investigated. Gefitinib inhibited cell proliferation of HT-29 cells (K-ras wild-type), and NDAT significantly enhanced the antiproliferative action of gefitinib. Gefitinib inhibited cell proliferation of HCT116 cells (K-ras mutant) only in high concentration, and this was further enhanced by NDAT. NDAT enhancedd gefitinib-induced antiproliferation in gefitinib-resistant colorectal cancer cells by inhibiting ST6Gal1 activity and PI3K activation. Furthermore, NDAT enhanced gefitinib-induced anticancer activity additively in colorectal cancer HCT116 cell xenograft-bearing nude mice. Results suggest that NDAT may have an application with gefitinib as combination colorectal cancer therapy.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/pharmacology , Colorectal Neoplasms/pathology , Gefitinib/pharmacology , Polyglactin 910/pharmacology , Thyroxine/analogs & derivatives , Animals , Apoptosis/drug effects , Cell Proliferation/drug effects , Enzyme Activation/drug effects , ErbB Receptors/drug effects , ErbB Receptors/metabolism , HCT116 Cells , HT29 Cells , Humans , Mice , Mice, Nude , Phosphatidylinositol 3-Kinases/drug effects , Phosphatidylinositol 3-Kinases/metabolism , Thyroxine/pharmacology , Xenograft Model Antitumor Assays
12.
Front Pharmacol ; 9: 807, 2018.
Article in English | MEDLINE | ID: mdl-30116189

ABSTRACT

Traditional herb medicine, golden thread (Anoectochilus formosanus Hayata) has been used to treat various diseases. Hyperglycemia induces generation of reactive oxygen species (ROS) and enhancement of oxidative stress which are risk factors for cancer progression and metastasis. In this study, we evaluated hypoglycemic effect of A. formosanus extracts (AFEs) in an inducible hyperglycemia animal model and its capacity of free-radical scavenging to establish hyperglycemia-related carcinogenesis. AFE reduced blood glucose in hyperglycemic mice while there was no change in control group. The incremental area under blood glucose response curve was decreased significantly in hyperglycemic mice treated with AFE in a dose-dependent manner. AFE and metformin at the same administrated dose of 50 mg/kg showed similar effect on intraperitoneal glucose tolerance test in hyperglycemic mice. Free-radical scavenger capacity of AFE was concentration dependent and 200 µg/ml of AFE was able to reduce more than 41% of the free radical. Treatment of cancer cells with AFE inhibited constitutive PD-L1 expression and its protein accumulation. It also induced expression of pro-apoptotic genes but inhibited proliferative and metastatic genes. In addition, it induced anti-proliferation in cancer cells. The results suggested that AFE not only reduced blood glucose concentration as metformin but also showed its potential use in cancer immune chemoprevention/therapy via hypoglycemic effect, ROS scavenging and PD-L1 suppression.

13.
J Cell Mol Med ; 22(2): 999-1013, 2018 02.
Article in English | MEDLINE | ID: mdl-29214724

ABSTRACT

CME-1, a novel water-soluble polysaccharide purified from Ophiocordyceps sinensis mycelia, has anti-oxidative, antithrombotic and antitumour properties. In this study, other major attributes of CME-1, namely anti-inflammatory and immunomodulatory properties, were investigated. Treating lipopolysaccharide (LPS)-stimulated RAW 264.7 cells with CME-1 concentration-dependently suppressed nitric oxide formation and inducible nitric oxide synthase (iNOS) expression. In the CME-1-treated RAW 264.7 cells, LPS-induced IκBα degradation and the phosphorylation of p65, Akt and mitogen-activated protein kinases (MAPKs), including extracellular signal-regulated kinase, c-Jun N-terminal kinase and p38, were reduced. Treatment with a protein phosphatase 2A (PP2A)-specific inhibitor, significantly reversed the CME-1-suppressed iNOS expression; IκBα degradation; and p65, Akt and MAPK phosphorylation. PP2A activity up-regulation and PP2A demethylation reduction were also observed in the cells. Moreover, CME-1-induced PP2A activation and its subsequent suppression of LPS-activated RAW 264.7 cells were diminished by the inhibition of ceramide signals. LPS-induced reactive oxygen species (ROS) and hydroxyl radical formation were eliminated by treating RAW 264.7 cells with CME-1. Furthermore, the role of ceramide signalling pathway and anti-oxidative property were also demonstrated in CME-1-mediated inhibition of LPS-activated primary peritoneal macrophages. In conclusion, CME-1 suppressed iNOS expression by up-regulating ceramide-induced PP2A activation and reducing ROS production in LPS-stimulated macrophages. CME-1 is a potential therapeutic agent for treating inflammatory diseases.


Subject(s)
Ceramides/pharmacology , Lipopolysaccharides/pharmacology , Macrophages/enzymology , Nitric Oxide Synthase Type II/metabolism , Polysaccharides/pharmacology , Protein Phosphatase 2/metabolism , Animals , Antioxidants/pharmacology , Cordyceps/chemistry , Enzyme Activation/drug effects , MAP Kinase Signaling System/drug effects , Macrophages/drug effects , Male , Mice , Mice, Inbred C57BL , NF-KappaB Inhibitor alpha/metabolism , NF-kappa B/metabolism , Nitric Oxide/biosynthesis , Proto-Oncogene Proteins c-akt/metabolism , RAW 264.7 Cells , Reactive Oxygen Species/metabolism , Signal Transduction/drug effects
14.
Sci Rep ; 6: 22166, 2016 Feb 26.
Article in English | MEDLINE | ID: mdl-26916099

ABSTRACT

The strength of hydrogen bond (HB) decides water's property and activity. Here we propose the mechanisms on creation and persistence of innovatively prepared liquid water, which is treated by Au nanoparticles (AuNPs) under resonant illumination of green-light emitting diode (LED) to create Au NP-treated (sAuNT) water, with weak HB at room temperature. Hot electron transfer on resonantly illuminated AuNPs, which is confirmed from Au LIII-edge X-ray absorption near edge structure (XANES) spectra, is responsible for the creation of negatively charged sAuNT water with the incorporated energy-reduced hot electron. This unique electronic feature makes it stable at least for one week. Compared to deionized (DI) water, the resulting sAuNT water exhibits many distinct properties at room temperature. Examples are its higher activity revealed from its higher vapor pressure and lower specific heat. Furthermore, Mpemba effect can be successfully explained by our purposed hypothesis based on sAuNT water-derived idea of water energy and HB.

15.
Article in English | MEDLINE | ID: mdl-25738390

ABSTRACT

Objective: CME-1 is a polysaccharide purified from the mycelia of medicinal mushroom Cordyceps sinensis, its molecular weight was determined to be 27.6 kDa by using nuclear magnetic resonance and gas chromatography-mass spectrometry. The initiation of arterial thromboses is relevant to various cardiovascular diseases (CVDs) and is believed to involve platelet activation. Our recent study exhibited that CME-1 has potent antiplatelet activity via the activation of adenylate cyclase/cyclic AMP ex vivo and in vivo. Methods: The aggregometry, and immunoblotting were used in this study. Results: In this study, the mechanisms of CME-1 in platelet activation is further investigated and found that CME-1 inhibited platelet aggregation as well as the ATP-release reaction, relative intracellular [Ca+2] mobilization, and the phosphorylation of phospholipase C (PLC)γ2 and protein kinase C (PKC) stimulated by collagen. CME-1 has no effects on inhibiting either convulxin, an agonist of glycoprotein VI, or aggretin, an agonist of integrin α2ß1 stimulated platelet aggregation. Moreover, this compound markedly diminished thrombin and arachidonic acid (AA) induced phosphorylation of p38 mitogen-activated protein kinase (MAPK), extracellular signal-regulated kinase 2, c-Jun N-terminal kinase 1, and Akt. Treatment with SQ22536, an inhibitor of adenylate cyclase, markedly diminished the CME-1-mediated increasing of cyclic AMP level and reversed prostaglandin E1- or CME-1-mediated inhibition of platelet aggregation and p38 MAPK and Akt phosphorylation stimulated by thrombin or AA. Furthermore, phosphodiesterase activity of human platelets was not altered by CME-1. Conclusion: The crucial finding of this study is that the antiplatelet activity of CME-1 may initially inhibit the PLCγ2-PKC-p47 cascade, and inhibit PI3-kinase/Akt and MAPK phosphorylation through adenylate cyclase/cyclic AMP activation, then inhibit intracellular [Ca+2] mobilization, and, ultimately, inhibit platelet activation. The novel role of CME-1 in antiplatelet activity indicates that this compound exhibits high therapeutic potential for treating or preventing CVDs.

16.
Curr Pharm Biotechnol ; 16(5): 451-61, 2015.
Article in English | MEDLINE | ID: mdl-25751172

ABSTRACT

OBJECTIVE: CME-1 is a polysaccharide purified from the mycelia of medicinal mushroom Cordyceps sinensis, its molecular weight was determined to be 27.6 kDa by using nuclear magnetic resonance and gas chromatography-mass spectrometry. The initiation of arterial thromboses is relevant to various cardiovascular diseases (CVDs) and is believed to involve platelet activation. Our recent study exhibited that CME-1 has potent antiplatelet activity via the activation of adenylate cyclase/cyclic AMP ex vivo and in vivo. METHODS: The aggregometry, and immunoblotting were used in this study. RESULTS: In this study, the mechanisms of CME-1 in platelet activation is further investigated and found that CME-1 inhibited platelet aggregation as well as the ATP-release reaction, relative intracellular [Ca(+2)] mobilization, and the phosphorylation of phospholipase C (PLC)γ2 and protein kinase C (PKC) stimulated by collagen. CME-1 has no effects on inhibiting either convulxin, an agonist of glycoprotein VI, or aggretin, an agonist of integrin α2ß1 stimulated platelet aggregation. Moreover, this compound markedly diminished thrombin and arachidonic acid (AA) induced phosphorylation of p38 mitogen-activated protein kinase (MAPK), extracellular signal-regulated kinase 2, c-Jun N-terminal kinase 1, and Akt. Treatment with SQ22536, an inhibitor of adenylate cyclase, markedly diminished the CME-1-mediated increasing of cyclic AMP level and reversed prostaglandin E1- or CME-1-mediated inhibition of platelet aggregation and p38 MAPK and Akt phosphorylation stimulated by thrombin or AA. Furthermore, phosphodiesterase activity of human platelets was not altered by CME-1. CONCLUSION: The crucial finding of this study is that the antiplatelet activity of CME-1 may initially inhibit the PLCγ2-PKC-p47 cascade, and inhibit PI3-kinase/Akt and MAPK phosphorylation through adenylate cyclase/ cyclic AMP activation, then inhibit intracellular [Ca(+2)] mobilization, and, ultimately, inhibit platelet activation. The novel role of CME-1 in antiplatelet activity indicates that this compound exhibits high therapeutic potential for treating or preventing CVDs.


Subject(s)
Blood Platelets/drug effects , Platelet Activation/drug effects , Platelet Aggregation Inhibitors/pharmacology , Polysaccharides/pharmacology , Blood Platelets/metabolism , Cordyceps , Humans , Mycelium , Phosphatidylinositol 3-Kinases/metabolism , Phospholipase C gamma/metabolism , Protein Kinase C/metabolism , Proto-Oncogene Proteins c-akt/metabolism
17.
Oncotarget ; 5(24): 12891-907, 2014 Dec 30.
Article in English | MEDLINE | ID: mdl-25436977

ABSTRACT

We have used a perfusion bellows cell culture system to investigate resveratrolinduced anti-proliferation/apoptosis in a human estrogen receptor (ER)-negative breast cancer cell line (MDA-MB-231). Using an injection system to perfuse media with stilbene, we showed resveratrol (0.5 - 100 µM) to decrease cell proliferation in a concentration-dependent manner. Comparison of influx and medium efflux resveratrol concentrations revealed rapid disappearance of the stilbene, consistent with cell uptake and metabolism of the agent reported by others. Exposure of cells to 10 µM resveratrol for 4 h daily × 6 d inhibited cell proliferation by more than 60%. Variable extracellular acid-alkaline conditions (pH 6.8 - 8.6) affected basal cell proliferation rate, but did not alter anti-proliferation induced by resveratrol. Resveratrol-induced gene expression, including transcription of the most up-regulated genes and pro-apoptotic p53-dependent genes, was not affected by culture pH changes. The microarray findings in the context of induction of anti-proliferation with brief daily exposure of cells to resveratrol-and rapid disappearance of the compound in the perfusion system-are consistent with existence of an accessible initiation site for resveratrol actions on tumor cells, e.g., the cell surface receptor for resveratrol described on integrin αvß3.


Subject(s)
Breast Neoplasms/drug therapy , Stilbenes/pharmacology , Breast Neoplasms/genetics , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Cell Line, Tumor , Cell Proliferation/drug effects , Female , Gene Expression/drug effects , Humans , Integrin alphaVbeta3/metabolism , Resveratrol , Signal Transduction
18.
Thromb Res ; 134(6): 1301-10, 2014 Dec.
Article in English | MEDLINE | ID: mdl-25294588

ABSTRACT

INTRODUCTION: CME-1, a novel water-soluble polysaccharide, was purified from the mycelia of Cordyceps sinensis, and its chemical structure was characterized to contain mannose and galactose in a ratio of 4:6 (27.6 kDa). CME-1 was originally observed to exert a potent inhibitory effect on tumor migration and a cytoprotective effect against oxidative stress. Activation of platelets caused by arterial thrombosis is relevant to various cardiovascular diseases (CVDs). However, no data are available concerning the effects of CME-1 on platelet activation. Hence, the purpose of this study was to examine the ex vivo and in vivo antithrombotic effects of CME-1 and its possible mechanisms in platelet activation. METHODS: The aggregometry, immunoblotting, flow cytometric analysis and platelet functional analysis were used in this study. RESULTS: CME-1 (2.3-7.6 µM) exhibited highly potent activity in inhibiting human platelet aggregation when stimulated by collagen, thrombin, and arachidonic acid but not by U46619. CME-1 inhibited platelet activation accompanied by inhibiting Akt, mitogen-activated protein kinases (MAPKs), thromboxane B2 (TxB2) and hydroxyl radical (OH(●)) formation. However, CME-1 interrupted neither FITC-triflavin nor FITC-collagen binding to platelets. CME-1 markedly increased cyclic AMP levels, but not cyclic GMP levels, and stimulated vasodilator-stimulated phosphoprotein (VASP) phosphorylation. SQ22536, an inhibitor of adenylate cyclase, but not ODQ, an inhibitor of guanylate cyclase, obviously reversed the CME-1-mediated effects on platelet aggregation and vasodilator-stimulated phosphoprotein (VASP), Akt, p38 MAPK phosphorylation, and TxB2 formation. CME-1 substantially prolonged the closure time of whole blood and the occlusion time of platelet plug formation. CONCLUSION: This study demonstrates for the first time that CME-1 exhibits highly potent antiplatelet activity that may initially activate adenylate cyclase/cyclic AMP and, subsequently, inhibit intracellular signals (such as Akt and MAPKs), ultimately inhibiting platelet activation. This novel role of CME-1 indicates that CME-1 exhibits high potential for application in treating and preventing CVDs.


Subject(s)
Adenylyl Cyclases/metabolism , Cordyceps/chemistry , Cyclic AMP/metabolism , Fungal Polysaccharides/pharmacology , Platelet Activation/drug effects , Platelet Aggregation Inhibitors/pharmacology , Polysaccharides/pharmacology , Thrombosis/drug therapy , Animals , Blood Coagulation/drug effects , Blood Coagulation/physiology , Cells, Cultured , Dose-Response Relationship, Drug , Enzyme Activation , Fungal Polysaccharides/chemistry , Fungal Polysaccharides/isolation & purification , Humans , Mice , Mycelium/chemistry , Platelet Activation/physiology , Platelet Aggregation Inhibitors/chemistry , Platelet Aggregation Inhibitors/isolation & purification , Polysaccharides/chemistry , Polysaccharides/isolation & purification , Signal Transduction/drug effects , Signal Transduction/physiology , Thrombosis/physiopathology , Treatment Outcome
19.
J Cancer Res Ther ; 10(1): 43-9, 2014.
Article in English | MEDLINE | ID: mdl-24762485

ABSTRACT

BACKGROUND: Matrix metalloproteinases (MMPs) play important roles in the invasion and migration of cancer cells. In melanoma, several signaling pathways are constitutively activated. Among these, the mitogen-activated protein kinase (MAPKs) signaling pathways are activated through multiple signal transduction molecules and appear to play major roles in melanoma progression. Therefore, the inhibition of MAPK signaling might be a crucial role for the treatment of melanoma cancer. AIMS: We examined the anticancer effect of CME-1, a novel water-soluble polysaccharide fraction, isolated from Cordyceps sinensis mycelia on B16-F10 melanoma cells. MATERIALS AND METHODS: B16-F10 cells were exposed to different concentrations of CME-1 (250, 500 and 800 µg/ml) for 24 h in 5% CO² incubator at 37°C. Western blot analysis was performed to detect the expression of MMP-1, p-p38 MAPK, p-ERK1/2, and IkB-α in B16-F10 cells. Cell migration test was performed by wound healing migration assay. RESULTS: CME-1 suppresses cell migration in a concentration-dependent manner. Western blotting analysis revealed that CME-1 led to the reduction on the expression levels of MMP-1 and down regulated the expression of phosphorylated extracellular signal-regulated kinase (ERK1/2 and p38 mitogen-activated protein kinase (p38 MAPK). CME-1 restored the IkB-degradation in B16F10 cells. CONCLUSIONS: These results indicate that CME-1 inhibited MMP-1 expressions in B16F10 melanoma cells through either NF-kB or ERK/p38 MAPK down regulation thereby inhibiting B16F10 cell migration. Therefore, we proposed that CME-1 might be developed as a therapeutic potential candidate for the treatment of cancer metastasis.


Subject(s)
Antineoplastic Agents/pharmacology , Cordyceps/chemistry , Fungal Polysaccharides/pharmacology , Mycelium/chemistry , Animals , Cell Line, Tumor , Cell Movement/drug effects , Extracellular Signal-Regulated MAP Kinases/metabolism , Matrix Metalloproteinase 1/metabolism , Melanoma, Experimental/drug therapy , Melanoma, Experimental/metabolism , Mice , Signal Transduction/drug effects , p38 Mitogen-Activated Protein Kinases/metabolism
20.
J Mol Med (Berl) ; 92(6): 603-14, 2014 Jun.
Article in English | MEDLINE | ID: mdl-24626935

ABSTRACT

UNLABELLED: gamma-Aminobutyric acid (GABA) is the major inhibitory neurotransmitter in the central nervous system, and it also appears in peripheral tissues. Platelets are anuclear blood cells that play a central role in hemostatic processes. Although platelets possess a GABA uptake system, the functional activity of GABA in platelets has remained unclear. We determined that GABA is abundantly distributed in the platelets at a concentration of approximately 1.03 ng/10(6) cells. GABA (0.5 µM) specifically inhibited collagen-induced platelet activation accompanied by [Ca(2+)]i mobilization, phospholipase Cγ2, protein kinase C, Akt phosphorylation, and hydroxyl radical formation. In addition, GABA interfered with fluorescein isothiocyanate-collagen binding to platelet membranes and produced a concentration-dependent shift in the collagen concentration-response curve and a Schild plot slope of -0.96 ± 0.11, indicating competitive inhibition. Platelet activation induced by convulxin, a glycoprotein VI agonist, was inhibited by GABA, whereas activation induced by the integrin α(2)ß(1) agonist, aggretin, was not. Immunoprecipitation and surface plasmon resonance revealed that GABA binds directly to glycoprotein VI in human platelets with equilibrium dissociation (binding) constant (K(D)) of 41.4 nM. The closure time of whole blood and the occlusion time of platelet plug formation were significantly prolonged by GABA in vivo. In this study, GABA is a specific inhibitor of collagen glycoprotein VI and may be involved in an endogenous negative feedback mechanism for platelet activation. Thus, GABA may represent a potential target for the development of novel interventions for the treatment of cardiovascular diseases associated with platelet activation, such as stroke and myocardial infarction. KEY MESSAGES: GABA is abundantly distributed in the platelets. GABA inhibited platelet activation stimulated by convulxin. GABA directly associated with glycoprotein VI in platelet membrane. GABA prolonged the closure time of whole blood and the occlusion time of platelet plug formation in vivo.


Subject(s)
Blood Platelets/drug effects , Blood Platelets/metabolism , Platelet Activation/drug effects , gamma-Aminobutyric Acid/metabolism , gamma-Aminobutyric Acid/pharmacology , Crotalid Venoms/pharmacology , Flow Cytometry , Humans , Immunoprecipitation , Lectins, C-Type , Platelet Membrane Glycoproteins/agonists , Surface Plasmon Resonance , Tandem Mass Spectrometry
SELECTION OF CITATIONS
SEARCH DETAIL
...