Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 43
Filter
Add more filters










Publication year range
1.
Int J Biol Macromol ; 227: 1-9, 2023 Feb 01.
Article in English | MEDLINE | ID: mdl-36528139

ABSTRACT

Polysaccharide from Laminaria japonica (LJPS) exhibits multiple biological functions. However, we found that crude LJPS doesn't show good anti-lung cancer activity in this study. We therefore used tangential flow filtration (TFF) system to optimize the anticancer activity of LJPS. We divided the crude LJPS into two fractions by TFF system with a 10 kDa filter and denoted as retentate (10K-R) and filtration (10K-F). The chemical assay revealed that the main molecular mass of 10K-R and 10K-F is about 985 and 3 kDa, respectively. The main components of 10K-R include fucose (19.3 %), and glucose (59.5 %); while glucose (88.6 %) is a major component of 10K-F. Biological functions showed that 10K-R but not 10K-F inhibited the viability and mobility of cancer cells. 10K-R downregulated expressions of transforming growth factor ß receptor and Slug, and inhibited intracellular signaling molecules, including FAK, AKT, ERK1/2, and Smad2. This study is the first concept to purify the polysaccharide by TFF system and showed the potential mechanism of 10K-R inhibited cancer cells.


Subject(s)
Laminaria , Neoplasms , Humans , Laminaria/chemistry , Polysaccharides/chemistry , Signal Transduction , Glucose
2.
Int J Biol Macromol ; 165(Pt A): 1604-1613, 2020 Dec 15.
Article in English | MEDLINE | ID: mdl-33011264

ABSTRACT

WSG is a water soluble polysaccharides isolated from Ganoderma lucidum. In this study, we showed that WSG, a glucose-rich polysaccharide with an average molecular mass of approximately 1000 kDa, effectively inhibited cell viability and mobility of lung cancer cells. Functional studies revealed that WSG reduced phosphorylation of ERK1/2 in cells upon either EGF or TGFß stimulation. WSG also inhibited phosphorylation of multiple intracellular signaling molecules such as FAK, AKT and Smad2. Mechanistically, we demonstrated that WSG induced degradation of TGFß and EGF receptors via proteasome and lysosome, respectively. Moreover, we found that WSG significantly suppressed lung tumor growth, reduced the size of metastatic nodules in the lungs and prolonged the survival of LLC1-bearing mice. Our findings suggested that WSG may have potential as a therapeutic intervention for treatment of lung cancer.


Subject(s)
Cell Proliferation/drug effects , Lung Neoplasms/drug therapy , Polysaccharides/chemistry , Reishi/chemistry , Animals , Cell Cycle/drug effects , Cell Line, Tumor , Gene Expression Regulation, Neoplastic/drug effects , Humans , Lung Neoplasms/pathology , MAP Kinase Signaling System/drug effects , Mice , Neoplasm Proteins/genetics , Polysaccharides/pharmacology , Xenograft Model Antitumor Assays
3.
Int J Biol Macromol ; 153: 796-805, 2020 Jun 15.
Article in English | MEDLINE | ID: mdl-32169452

ABSTRACT

Gefitinib is a first tyrosine kinase inhibitor (TKI) designed with an EGFR tyrosine kinase for lung cancer targeted therapy. However, some lung cancer patients with wild-type EGFR (wtEGFR) or acquired secondary EGFR mutation showed lower response rate of gefitinib. In this study, we examined the efficacy of fucoidan on altering gefitinib-sensitivity on TKI-resistant lung cancer A549 and H1975 cells. We found that the simultaneous administration of fucoidan and gefitinib synergistically inhibited lung cancer cell viability via activating apoptotic response. Moreover, we found that fucoidan effectively downregulated expressions of mesenchymal-like molecules. Mechanistically, we demonstrated that fucoidan altered the gefitinib-inhibitory rate may result from induction of proteasome-dependent Slug degradation. Abolishment of TGFß signaling enhanced gefitinib-inhibited cell viability and reduced N-cadherin, Twist and Slug levels. Moreover, knockdown of Slug contributed the increasing the gefitinib-sensitivity of H1975 cells. Our study is the first to find that fucoidan alters the gefitinib-sensitive of TKI-resistant cells by reduction of TGFß receptor-mediated expressions of mesenchymal-like molecules and induction of Slug degradation. Together, our current results indicate that combination of fucoidan and gefitinib may be a potential and effective therapeutic strategy in gefitinib non-sensitive lung cancer.


Subject(s)
Drug Resistance, Neoplasm/drug effects , Gefitinib/pharmacology , Gene Expression Regulation, Neoplastic/drug effects , Lung Neoplasms , Neoplasm Proteins/metabolism , Polysaccharides/pharmacology , Snail Family Transcription Factors/biosynthesis , Transforming Growth Factor beta/metabolism , A549 Cells , Animals , Humans , Lung Neoplasms/drug therapy , Lung Neoplasms/genetics , Lung Neoplasms/metabolism , Male , Mice
4.
Clin Transl Med ; 8(1): 15, 2019 May 01.
Article in English | MEDLINE | ID: mdl-31041568

ABSTRACT

The chemical composition of fucoidan, a kind of sulfated polysaccharide mainly derived from brown seaweed, includes a substantial percentage of L-fucose. Fucoidan has various biological and pharmacological activities, such as anti-cancer/anti-tumor, anti-proliferation, anti-inflammatory and immune-modulatory functions, and fucoidan-related dietary supplements and nutraceuticals have recently drawn considerable attention. In this review, we aim to provide a current view of different aspects of fucoidan biological activity, with a focus on the anti-cancer regulatory effects of fucoidan on growth signaling mechanisms. First, we discuss historical aspects of fucoidan and fucoidan products, as well as the anti-cancer effects of fucoidan on various cancer cells. Second, we discuss fucoidan's biological activities and induction of cell death in cancer cells, including multiple mechanisms and signal transduction pathways related to its anti-cancer effects. Next, we focus on fucoidan and fucoidan-derived products that have been marketed as dietary supplements or nutraceuticals for cancer, including anti-cancer effects of fucoidan when combined as an adjuvant with clinical drugs. Finally, case studies of fucoidan in complementary therapy and as an alternative medicine in animal and mouse models and human clinical trials to alleviate side effects of anti-cancer chemotherapy are discussed. Combining fucoidan with clinical therapeutic agents in the treatment of cancer patients, dissecting the related signal transduction pathways and investigating their dynamic interactions may reveal potential molecular targets in cancer prevention, therapies and key obstacles in the current development of anti-cancer strategies.

5.
Biochem Biophys Res Commun ; 506(4): 927-931, 2018 12 02.
Article in English | MEDLINE | ID: mdl-30392907

ABSTRACT

Rubiadin-1-methyl ether (RBM) is a natural anthraquinone compound isolated from the root of Morinda officinalis How. In our previous study, RBM was found to have inhibitory effects on the TRAP activity of osteoclasts, which means that RBM may be a candidate for therapy of bone diseases characterized by enhanced bone resorption. However, the further effect of RBM on osteoclasts and the underlying mechanism remain unclear. In the present study, we investigated the effects of RBM isolated from Morinda officinalis How. on osteoclasts derived from bone marrow macrophages (BMMs) and the underlying mechanism in vitro. RBM at the dose that did not affect the viability of cells significantly inhibited RANKL-induced osteoclastogenesis and actin ring formation of osteoclast, while RBM performed a stronger effect at the early stage. In addition, RBM downregulated the expression of osteoclast-related proteins, including nuclear factor of activated T cells cytoplasmic 1 (NFATc1), cellular oncogene Fos (c-Fos), matrix metallopeptidase 9 (MMP-9) and cathepsin K (CtsK) as shown by Western blot. Furthermore, RBM inhibited the phosphorylation of NF-κB p65 and the degradation of IκBα as well as decreased the nuclear translocation of p65. Collectively, the results suggest that RBM inhibit osteoclastic bone resorption through blocking NF-κB pathway and may be a promising agent for the prevention and treatment of bone diseases characterized by excessive bone resorption.


Subject(s)
Anthraquinones/pharmacology , Morinda/chemistry , NF-kappa B/metabolism , Osteoclasts/metabolism , Osteogenesis/drug effects , RANK Ligand/pharmacology , Signal Transduction , Actins/metabolism , Animals , Anthraquinones/chemistry , Biomarkers/metabolism , Cell Differentiation/drug effects , Mice, Inbred C57BL , NFATC Transcription Factors/metabolism , Osteoclasts/drug effects , Proto-Oncogene Proteins c-fos/metabolism , Signal Transduction/drug effects , Tartrate-Resistant Acid Phosphatase/metabolism
6.
BMC Complement Altern Med ; 18(1): 288, 2018 Oct 24.
Article in English | MEDLINE | ID: mdl-30355303

ABSTRACT

BACKGROUND: Iridoid glycosides (IGs), including monotropein (MON) and deacetyl asperulosidic acid (DA) as the main ingredients, are the major chemical components in Morinda officinalis How. (MO) root, possessing various pharmacological properties including anti-osteoporosis, anti-inflammation and anti-rheumatism activities.The aim of the present study was to further elucidate the pharmacological actions of MO by investigating the pharmacokinetics and tissue distribution of IGs in MO. METHODS: An ultra high performance liquid chromatography-tandem mass spectrometry (UHPLC-MS) method was developed and validated for simultaneous determination of MON and DA levels in plasma and various tissues of Wistar rats. MON, DA and acetaminophen (ACE) as the internal standard (IS) were extracted from rat plasma and tissue samples by direct deproteinization with methanol. The rats were administered orally at 1650 mg/kg MO and 25, 50 and 100 mg/kg MO iridoid glycosides (MOIGs) or intravenously at MOIG 25 mg/kg for pharmacokinetic study of MON and DA. In addition, 100 mg/kg MOIG was administered orally for tissue distribution study of MON and DA. Non-compartmental pharmacokinetic profiles were constructed. Tissue distributions were calculated according to the validated methods. RESULTS: Significant differences in the pharmacokinetic parameters were observed in male and female rats. The AUC0-t, Cmax and bioavailability of MON and DA in female rats were higher than those in male rats. MON and DA mainly distributed in the intestine and stomach after oral administration, and noteworthily high concentrations of MON and DA were detected in the rat hypothalamus. CONCLUSION: The results of the present study may shed new lights on the biological behavior of MOIGs in vivo, help explain their pharmacological actions, and provide experimental clues for rational clinical use of these IGs extracted from the MO root.


Subject(s)
Drugs, Chinese Herbal/pharmacokinetics , Glycosides/pharmacokinetics , Iridoids/pharmacokinetics , Morinda/chemistry , Administration, Oral , Animals , Drugs, Chinese Herbal/administration & dosage , Drugs, Chinese Herbal/chemistry , Female , Glycosides/administration & dosage , Glycosides/chemistry , Iridoid Glycosides/administration & dosage , Iridoid Glycosides/chemistry , Iridoid Glycosides/pharmacokinetics , Iridoids/administration & dosage , Iridoids/chemistry , Male , Molecular Structure , Plant Roots/chemistry , Rats , Rats, Wistar , Tandem Mass Spectrometry , Tissue Distribution
7.
Chem Biol Interact ; 291: 128-136, 2018 Aug 01.
Article in English | MEDLINE | ID: mdl-29908987

ABSTRACT

Estrogen deficiency and inflammation are known to play important roles in bone metabolism and occurrence of osteoporosis. Monotropein as an iridoid glycoside is reported to decrease estrogen deficiency-induced bone loss and inhibit inflammatory response in LPS-induced RAW 264.7 macrophages. However, the effect of monotropein on bone loss in chronic inflammatory conditions remains unclear. It was found in the present study that monotropein significantly inhibited bone mass reduction and improved bone micro-architectures by enhancing bone formation and blocking increased secretion of inflammatory cytokines in osteoporotic mice induced by combined ovariectomy and LPS. Our in vitro experiment further demonstrated that monotropein was able to increase the proliferation and activity of alkaline phosphatase (ALP), bone matrix mineralization and the expression of bone matrix protein osteopontin (OPN) in osteoblastic MC3T3-E1 cells injured by LPS. In addition, monotropein significantly decreased the production of IL-6 and IL-1ß, inhibited the nuclear translocation of p65 and NF-κB P50, and down-regulated the phosphorylation of NF-κB p65 and IKK, indicating that monotropein could attenuate inflammatory impairment to MC3T3-E1 cells by suppressing the activation of NF-κB pathway. All these results suggest that monotropein may prove to be a promising candidate for the prevention and treatment of inflammatory bone loss.


Subject(s)
Bone Resorption/chemically induced , Bone Resorption/drug therapy , Inflammation/pathology , Iridoids/therapeutic use , NF-kappa B/metabolism , Osteoblasts/pathology , Ovariectomy , Signal Transduction/drug effects , Alkaline Phosphatase/blood , Animals , Bone Density/drug effects , Bone Matrix/drug effects , Bone Matrix/metabolism , Bone Resorption/blood , Calcification, Physiologic/drug effects , Cell Cycle/drug effects , Cell Line , Cell Proliferation/drug effects , Female , Femur/diagnostic imaging , Femur/drug effects , Inflammation/complications , Interleukin-1beta/metabolism , Interleukin-6/metabolism , Iridoids/chemistry , Iridoids/pharmacology , Lipopolysaccharides , Mice, Inbred C57BL , Osteoblasts/drug effects , Osteoblasts/metabolism , Osteocalcin/metabolism , Osteoporosis/blood , Osteoporosis/pathology , X-Ray Microtomography
8.
Cancer Lett ; 432: 112-120, 2018 09 28.
Article in English | MEDLINE | ID: mdl-29746926

ABSTRACT

Cisplatin-based therapy is a traditional, clinical treatment for cancers, including lung cancer. In this study, we found that sequential therapy, i.e., cisplatin followed by fucoidan, reduced tumor volume in an LLC1-bearing C57BL/6 mouse model. Using a series of combined therapeutic experiments, we found that the inhibition rate of the sequential treatment (cisplatin→fucoidan) was 50-75%. However, the inhibition rate of the sequential treatment, with fucoidan pretreatment, was increased to 75-85%. Moreover, we found that the simultaneous administration of fucoidan and cisplatin synergistically inhibited lung cancer cell viability via inducing apoptotic responses, including upregulating cleaved caspase-3 and poly (ADP ribose) polymerase (PARP) expression. Mechanistically, we demonstrated that the fucoidan-induced, TLR4-mediated endoplasmic reticulum stress molecule CHOP promoted caspase-3 activation, which was further stimulated by the cisplatin-induced DNA damage responses, and CHOP shRNA eliminated fucoidan-induced caspase-3 cleavage but did not affect cisplatin-mediated apoptotic molecules. In addition, we observed an increasing number of clinical results that suggest combined cisplatin and fucoidan exerts a greater anti-tumorigenic effect in patients with lung cancer in Taiwan. Together, our current results support the potential of combined fucoidan and cisplatin treatment as an effective therapeutic strategy in lung cancer.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/pharmacology , Carcinoma, Lewis Lung/pathology , Caspase 3/metabolism , Lung Neoplasms/pathology , Poly (ADP-Ribose) Polymerase-1/metabolism , Toll-Like Receptor 4/metabolism , Transcription Factor CHOP/metabolism , Animals , Apoptosis , Carcinoma, Lewis Lung/drug therapy , Carcinoma, Lewis Lung/metabolism , Caspase 3/genetics , Cell Cycle , Cell Proliferation , Cisplatin/administration & dosage , Drug Synergism , Female , Gene Expression Regulation, Neoplastic , Humans , Lung Neoplasms/drug therapy , Lung Neoplasms/metabolism , Male , Mice , Mice, Inbred C57BL , Poly (ADP-Ribose) Polymerase-1/genetics , Polysaccharides/administration & dosage , Toll-Like Receptor 4/genetics , Transcription Factor CHOP/genetics , Tumor Cells, Cultured
9.
Carbohydr Polym ; 167: 229-239, 2017 Jul 01.
Article in English | MEDLINE | ID: mdl-28433158

ABSTRACT

A sulfated 1,4-ß-d-galactoglucan (B86-III) with 1,6-branches was isolated and identified from Antrodia cinnamomea. The repeating unit of B86-III was proposed based on one-dimensional 1D (1H, 13C and DEPT-135) and 2D (DQF-COSY, TOCSY, HSQC and HMBC) NMR spectra. The conformation of the sugars was hypothesized to be a rare boat form instead of a 4C1 chair form. The sulfate substitutions were suggested to be on the C-2 and C-3 positions, resulting in the following structure: B86-III inhibited the viability of H1975 lung cancer cells via cell apoptosis, including the activation of caspase 3 and PARP. Transforming growth factor ß receptor (TGFR) and its downstream signaling FAK and Slug are involved in lung tumorigenesis. B86-III downregulated TGFR I protein levels and inhibited FAK phosphorylation, resulting in inhibition of Slug expression and migration. This study is the first to characterize sulfated polysaccharides with a rare boat-form conformation and identify the mechanism of inhibition lung cancer cell.


Subject(s)
Antineoplastic Agents/chemistry , Antrodia/chemistry , Focal Adhesion Kinase 1/metabolism , Galactans/chemistry , Glucans/chemistry , Polysaccharides, Bacterial/chemistry , Snail Family Transcription Factors/metabolism , Transforming Growth Factor beta/metabolism , Apoptosis , Cell Line, Tumor , Cell Survival , Humans , Lung Neoplasms/pathology , Signal Transduction
10.
Sci Rep ; 7: 44990, 2017 03 23.
Article in English | MEDLINE | ID: mdl-28332554

ABSTRACT

Fucoidan, a sulfated polysaccharide extracted from brown algae, exhibits anti-cancer activity. However, the effects and mechanism of fucoidan-induced apoptosis via endoplasmic reticulum (ER) stress is unclear. In this study, we demonstrated that fucoidan prevents tumorigenesis and reduces tumor size in LLC1-xenograft male C57BL/6 mice. Fucoidan induces an ER stress response by activating the PERK-ATF4-CHOP pathway, resulting in apoptotic cell death in vitro and in vivo. Furthermore, ATF4 knockdown abolishes fucoidan-induced CHOP expression and rescues cell viability. Specifically, fucoidan increases intracellular reactive oxygen species (ROS), which increase ATF4 and CHOP in lung cancer cells. Using the ROS scavenger N-acetyl-l-cysteine (NAC), we found that ROS generation is involved in fucoidan-induced ER stress-mediated apoptosis. Moreover, via Toll-like receptor 4 (TLR4) knockdown, we demonstrated that fucoidan-induced ROS and CHOP expression were attenuated. Our study is the first to identify a novel mechanism for the antitumor activity of fucoidan. We showed that fucoidan inhibits tumor viability by activating the TLR4/ROS/ER stress axis and the downstream PERK-ATF4-CHOP pathway, leading to apoptosis and suppression of lung cancer cell progression. Together, these results indicate that fucoidan is a potential preventive and therapeutic agent for lung cancer that acts via activation of ROS-dependent ER stress pathways.


Subject(s)
Antineoplastic Agents/pharmacology , Apoptosis/drug effects , Endoplasmic Reticulum Stress/drug effects , Lung Neoplasms/metabolism , Polysaccharides/pharmacology , Reactive Oxygen Species/metabolism , Toll-Like Receptor 4/metabolism , Activating Transcription Factor 4/genetics , Activating Transcription Factor 4/metabolism , Animals , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Survival/drug effects , Disease Models, Animal , Gene Expression Regulation, Neoplastic , Humans , Lung Neoplasms/drug therapy , Lung Neoplasms/genetics , Lung Neoplasms/pathology , Mice , Phosphorylation , Proto-Oncogene Proteins c-akt/metabolism , RNA, Small Interfering/genetics , Transcription Factor CHOP/genetics , Transcription Factor CHOP/metabolism , Tumor Burden/drug effects , Xenograft Model Antitumor Assays
11.
Int J Cancer ; 140(11): 2596-2607, 2017 06 01.
Article in English | MEDLINE | ID: mdl-28198003

ABSTRACT

Activating mutation of epidermal growth factor receptor (EGFR) is correlated with malignant lung tumor. In our study, we demonstrated that recombinant LZ-8 (rLZ-8), a medicinal mushroom Ganoderma lucidum protein, induced cell cycle arrest and apoptosis by downregulating the expression of wild-type and mutated EGFR and inhibiting EGFR downstream effectors, AKT and ERK1/2 in lung cancer cells. We showed that rLZ-8 effectively inhibited lung cancer progression and suppressed EGFR expression of lung tumor lesions in mouse model. Functional studies revealed that rLZ-8 reduced the amount of EGFR in cell membranes by altering EGFR localization to enhance the EGF-induced degradation of EGFR. Mechanistically, we demonstrated that rLZ-8 bound to EGFR to induce EGFR autophosphorylation at tyrosine1045 and trigger ubiquitination by inducing the formation of EGFR/Cbl complexes, resulting in the degradation of EGFR; however, Cbl-shRNA abolished rLZ-8-induced EGFR degradation. We provide the first evidence showing that rLZ-8 inhibits growth and induces apoptosis of lung cancer cells by promoting EGFR degradation. The current findings therefore suggest a novel anti-cancer function of rLZ-8 that targeting EGFR overexpression or mutation as well as EGFR-dependent processes in cancer cells.


Subject(s)
Antineoplastic Agents/pharmacology , ErbB Receptors/genetics , Fungal Proteins/pharmacology , Lung Neoplasms/drug therapy , Lung Neoplasms/genetics , Oncogene Protein v-cbl/genetics , Recombinant Proteins/pharmacology , A549 Cells , Agaricales/genetics , Animals , Apoptosis/drug effects , Cell Cycle Checkpoints/drug effects , Cell Cycle Checkpoints/genetics , Cell Proliferation/drug effects , Down-Regulation/drug effects , Fungal Proteins/economics , Humans , Male , Mice , Mice, Inbred C57BL , Mutation/drug effects , Mutation/genetics , Reishi/genetics , Signal Transduction/drug effects , Ubiquitination/drug effects
12.
Int J Biol Macromol ; 95: 1144-1152, 2017 Feb.
Article in English | MEDLINE | ID: mdl-27818294

ABSTRACT

A sulfated polysaccharide of edible mushroom Antrodia cinnamomea (SPS) has been identified as a novel immunomodulatory agent. We examined the anti-cancer effects of SPS by conducting a series of in vitro studies. We found that SPS inhibits the growth of A549 and LLC1 lung cancer cells via the induction of cell cycle arrest and activation of caspase 3 and PARP. By contrast, we found that a non-sulfated polysaccharide of A. cinnamomea (NSPS) does not inhibit lung cancer cell viability. Moreover, NSPS does not induce changes in cell cycle distribution or activate apoptosis-related molecules in both A549 and LLC1 cells. High expression of transforming growth factor ß (TGFß) and TGFß receptors (TGFRs) is correlated with lung tumorigenesis. SPS suppresses TGFß-induced intracellular signaling events, including phosphorylation of Smad2/3, FAK, Akt, and cell migration. By contrast, non-sulfated polysaccharide (NSPS) does not exhibit the similar biological functions in both A549 and LLC1 cells. Mechanistically, we demonstrated SPS effectively reduces TGFR protein levels via induction of proteasome-dependent degradation pathway. Our study is the first to identify the pivotal role of SPS in the induction of TGFR degradation and activation of Caspase 3 and PARP, which leads to suppress viability and migration of lung cancer cells.


Subject(s)
Antineoplastic Agents/pharmacology , Antrodia/chemistry , Epithelial Cells/drug effects , Fungal Polysaccharides/pharmacology , Gene Expression Regulation, Neoplastic , Immunologic Factors/pharmacology , Proteasome Endopeptidase Complex/drug effects , Antineoplastic Agents/isolation & purification , Caspase 3/genetics , Caspase 3/metabolism , Cell Cycle Checkpoints/drug effects , Cell Line, Tumor , Cell Movement/drug effects , Cell Proliferation/drug effects , Epithelial Cells/metabolism , Epithelial Cells/pathology , Focal Adhesion Kinase 1/genetics , Focal Adhesion Kinase 1/metabolism , Fruiting Bodies, Fungal/chemistry , Fungal Polysaccharides/isolation & purification , Humans , Immunologic Factors/isolation & purification , Poly(ADP-ribose) Polymerases/genetics , Poly(ADP-ribose) Polymerases/metabolism , Proteasome Endopeptidase Complex/metabolism , Proteolysis/drug effects , Proto-Oncogene Proteins c-akt/genetics , Proto-Oncogene Proteins c-akt/metabolism , Receptors, Transforming Growth Factor beta/genetics , Receptors, Transforming Growth Factor beta/metabolism , Respiratory Mucosa/drug effects , Respiratory Mucosa/metabolism , Respiratory Mucosa/pathology , Signal Transduction , Smad2 Protein/genetics , Smad2 Protein/metabolism , Smad3 Protein/genetics , Smad3 Protein/metabolism , Sulfates/chemistry , Transforming Growth Factor beta/genetics , Transforming Growth Factor beta/metabolism
13.
Sci Rep ; 6: 36563, 2016 11 10.
Article in English | MEDLINE | ID: mdl-27830743

ABSTRACT

Ganoderma lucidum exerts antitumor activity, but the mechanism of G. lucidum polysaccharides on cancer is unclear. Here, we demonstrated that a fucose-containing fraction of Ling-Zhi (FFLZ) reduced tumor size and suppressed metastasis in vivo. Furthermore, FFLZ inhibited breast cancer cell migration and altered the epithelial-to-mesenchymal transition (EMT) phenotype. Transforming growth factor-ß receptor (TGFR) pathways act as key mediators to promote tumor progression and metastasis. We found that FFLZ down-regulated TGFR and downstream signaling pathways, including the phosphorylation of Smad2/3 and the expression of Smad4. In an investigation of the underlying mechanisms, we found that FFLZ enhanced the Smurf2-dependent ubiquitination of TGFR by disrupting the balance of the lipid rafts, promoted the "re-localization" of the TGFR to the caveolae, and facilitated the degradation of TGFR. Together, our data indicated that FFLZ is associated with the inhibition of EMT and the prevention of metastasis by promoting ubiquitination-dependent TGFR degradation and abolishing TGFR signaling pathways. Moreover, the combination of FFLZ and trastuzumab synergistically inhibited the viability of certain trastuzumab-resistant human breast cancer cells. In summary, our current findings indicate that FFLZ is a potential therapeutic or dietary supplemental agent for cancer patients and that it functions via the caveolin-1/Smad7/Smurf2-dependent ubiquitin-mediated degradation of TGFR.


Subject(s)
Breast Neoplasms/metabolism , Fucose , Mammary Neoplasms, Animal/metabolism , Neoplasm Proteins/metabolism , Plant Extracts/pharmacology , Proteolysis/drug effects , Receptors, Transforming Growth Factor beta/metabolism , Ubiquitination/drug effects , Animals , Breast Neoplasms/genetics , Breast Neoplasms/pathology , Female , Humans , Mammary Neoplasms, Animal/genetics , Mammary Neoplasms, Animal/pathology , Mice, Inbred BALB C , Neoplasm Proteins/genetics , Plant Extracts/chemistry , Receptors, Transforming Growth Factor beta/genetics
14.
Cancer Lett ; 375(2): 340-348, 2016 Jun 01.
Article in English | MEDLINE | ID: mdl-26992741

ABSTRACT

We recently reported that recombinant Ling Zhi-8 (rLZ-8), a medicinal mushroom Ganoderma lucidum recombinant protein, effectively prevents lung cancer cells proliferation in vivo mice model. In our current study, we demonstrated that rLZ-8 suppressed tumor metastasis and increased the survival rate in Lewis lung carcinoma cell-bearing mice. The epithelial to mesenchymal transition (EMT) process is regarded as the critical event in tumor metastasis. Herein, we showed that rLZ-8 effectively induced changes in EMT by interfering with cell adhesion and focal adhesion kinase (FAK) functions in lung cancer cells. Slug, a transcription factor, represses E-cadherin transcription and is regarded as a critical event in EMT and tumor metastasis. Functional studies revealed that downregulation of Slug as a result of rLZ-8-induced FAK inactivation enhanced E-cadherin expression and repressed cancer cell mobility. Moreover, we found that rLZ-8 enhanced the ubiquitination proteasome pathway (UPP)-mediated degradation of Slug in CL1-5 cells. Mechanistically, we demonstrated that rLZ-8 promoted the interaction between MDM2 and Slug, resulting in Slug degradation; however, MDM2-shRNA abolished rLZ-8-enhanced Slug degradation. This study is the first to determine anti-metastatic activity of rLZ-8 and its potential mechanism, with how the regulation of EMT and cell mobility is via the negative modulation of FAK, and thereby leading to the ubiquitination and degradation of Slug. Our findings suggest that the targets of FAK play a key role in metastasis. Moreover, rLZ-8 may be useful as a chemotherapeutic agent for treating lung cancer.


Subject(s)
Focal Adhesion Kinase 1/genetics , Fungal Proteins/administration & dosage , Lung Neoplasms/genetics , Proto-Oncogene Proteins c-mdm2/genetics , Transcription Factors/genetics , Animals , Cell Line, Tumor , Cell Movement/drug effects , Cell Proliferation/drug effects , Epithelial-Mesenchymal Transition/drug effects , Focal Adhesions/drug effects , Fungal Proteins/genetics , Gene Expression Regulation, Neoplastic/drug effects , Humans , Lung Neoplasms/drug therapy , Lung Neoplasms/pathology , Mice , Neoplasm Metastasis , Proteolysis/drug effects , Signal Transduction/drug effects , Snail Family Transcription Factors , Transcription Factors/metabolism
15.
Oncotarget ; 5(17): 7870-85, 2014 Sep 15.
Article in English | MEDLINE | ID: mdl-25149540

ABSTRACT

Fucoidan, a polysaccharide extracted from brown seaweeds, reduces tumor cell proliferation. In this study, we demonstrate that fucoidan reduces tumor size in LLC1-xenograft male C57BL/6 mice. Moreover, we found that LLC1-bearing mice continuously fed fucoidan showed greater antitumor activity than mice with discontinuous feeding. Fucoidan inhibited the in vitro growth of lung cancer cells. Transforming growth factor ß (TGFß) receptors (TGFRs) play important roles in the regulation of proliferation and progression, and high TGFRI expression in lung cancer specimens is associated with a worse prognosis. Herein, using lung cancer cells, we found that fucoidan effectively reduces TGFRI and TGFRII protein levels in vivo and in vitro. Moreover, fucoidan reduces TGFR downstream signaling events, including those in Smad2/3 and non-Smad pathways: Akt, Erk1/2, and FAK phosphorylation. Furthermore, fucoidan suppresses lung cancer cell mobility upon TGFß stimulation. To elucidate how fucoidan decreases TGFR proteins in lung cancer cells, we found that fucoidan enhances the ubiquitination proteasome pathway (UPP)-mediated degradation of TGFRs in A549 and CL1-5 cells. Mechanistically, fucoidan promotes Smurf2 and Smad7 to conjugate TGFRs, resulting in TGF degradation; however, Smurf2-shRNA abolishes fucoidan-enhanced UPP-mediated TGFR degradation. Our study is the first to identify a novel mechanism for the antitumor activity of fucoidan, namely decreasing tumor growth by modulating the TGFR/Smad7/Smurf2-dependent axis, leading to TGFR protein degradation and inhibition of lung cancer cell progression in vitro and in vivo. Our current findings indicate that fucoidan is a potential therapeutic agent or dietary supplementation for lung cancer, acting via the Smurf2-dependent ubiquitin degradation of TGFß receptors.


Subject(s)
Antineoplastic Agents/pharmacology , Lung Neoplasms/metabolism , Polysaccharides/pharmacology , Receptors, Transforming Growth Factor beta/metabolism , Signal Transduction/drug effects , Ubiquitin-Protein Ligases/metabolism , Animals , Blotting, Western , Carcinoma, Non-Small-Cell Lung/metabolism , Cell Line, Tumor , Humans , Immunoprecipitation , In Vitro Techniques , Male , Mice , Mice, Inbred C57BL , Proteasome Endopeptidase Complex/drug effects , Proteasome Endopeptidase Complex/metabolism , RNA, Small Interfering , Transfection , Ubiquitin/metabolism , Xenograft Model Antitumor Assays
16.
Article in English | MEDLINE | ID: mdl-24723958

ABSTRACT

The expression of inflammatory cytokines on vascular walls is a critical event in vascular diseases and inflammation. The aim of the present study was to examine the effects of an extract of Ganoderma lucidum (Reishi) polysaccharides (EORPs), which is effective against immunological disorders, on interleukin- (IL-) 1 ß expression by human aortic smooth muscle cells (HASMCs) and the underlying mechanism. The lipopolysaccharide- (LPS-) induced IL-1 ß expression was significantly reduced when HASMCs were pretreated with EORP by Western blot and immunofluorescent staining. Pretreatment with 10 µ g/mL EORP decreased LPS-induced ERK, p38, JNK, and Akt phosphorylation. But the increase in IL-1 ß expression with LPS treatment was only inhibited by pretreatment with the ERK1/2 inhibitor, while the JNK and p38 inhibitors had no effect. In addition, EORP reduced the phosphorylation and nuclear translocation of nuclear factor- (NF-) κ B p65 in LPS-treated HASMCs. Furthermore, in vivo, IL-1 ß expression was strongly expressed in thoracic aortas in LPS-treated mice. Oral administration of EORP decreased IL-1 ß expression. The level of IL-1 ß expression in LPS-treated or in LPS/EORP-treated group was very low and was similar to that of the saline-treated group in toll-like receptor 4-deficient (TLR4(-/-)) mice. These findings suggest that EORP has the anti-inflammatory property and could prove useful in the prevention of vascular diseases and inflammatory responses.

17.
Proc Natl Acad Sci U S A ; 110(34): 13809-14, 2013 Aug 20.
Article in English | MEDLINE | ID: mdl-23908400

ABSTRACT

Carbohydrate-based vaccines have shown therapeutic efficacy for infectious disease and cancer. The mushroom Ganoderma lucidum (Reishi) containing complex polysaccharides has been used as antitumor supplement, but the mechanism of immune response has rarely been studied. Here, we show that the mice immunized with a l-fucose (Fuc)-enriched Reishi polysaccharide fraction (designated as FMS) induce antibodies against murine Lewis lung carcinoma cells, with increased antibody-mediated cytotoxicity and reduced production of tumor-associated inflammatory mediators (in particular, monocyte chemoattractant protein-1). The mice showed a significant increase in the peritoneal B1 B-cell population, suggesting FMS-mediated anti-glycan IgM production. Furthermore, the glycan microarray analysis of FMS-induced antisera displayed a high specificity toward tumor-associated glycans, with the antigenic structure located in the nonreducing termini (i.e., Fucα1-2Galß1-3GalNAc-R, where Gal, GalNAc, and R represent, respectively, D-galactose, D-N-acetyl galactosamine, and reducing end), typically found in Globo H and related tumor antigens. The composition of FMS contains mainly the backbone of 1,4-mannan and 1,6-α-galactan and through the Fucα1-2Gal, Fucα1-3/4Man, Fucα1-4Xyl, and Fucα1-2Fuc linkages (where Man and Xyl represent d-mannose and d-xylose, respectively), underlying the molecular basis of the FMS-induced IgM antibodies against tumor-specific glycans.


Subject(s)
Antibodies, Neoplasm/immunology , Antigens, Tumor-Associated, Carbohydrate/immunology , Carcinoma, Lewis Lung/immunology , Fucose/immunology , Fungal Polysaccharides/immunology , Reishi/chemistry , Animals , B-Lymphocytes/immunology , Chemokine CCL2/metabolism , Fungal Polysaccharides/metabolism , Immunization , Immunoglobulin M/blood , Immunoglobulin M/metabolism , Mice , Mice, Inbred C57BL , Microarray Analysis
18.
Article in English | MEDLINE | ID: mdl-23864893

ABSTRACT

LZ-8, an immunomodulatory protein isolated from Ganoderma lucidum (also known as Ling-Zhi or Reishi), has been shown to promote cell proliferation and IL-2 production in T cells. In this study, we show that LZ-8 induces the expansion of both murine and human CD4(+) T cells into FOXP3(+) regulatory T (Treg) cells. LZ-8 treatment was found to stimulate a 4-fold and a 10-fold expansion in the Treg populations of murine and human primary CD4(+) T cells, respectively. In addition, the expression of CTLA-4 and IL-10 was induced in LZ-8-treated CD4(+) T cells. Using neutralizing antibodies and gene-deficient T-cell lines, we also found that LZ-8 promotes Treg expansion through a CD45-mediated signaling pathway and that the CD18-dependent induction of IL-2 was involved in Treg formation and IL-10 production. The suppressive activity of LZ-8 was confirmed using a murine model of DSS-induced colitis; the disease was alleviated by the adoptive transfer of LZ-8-treated CD4(+) T cells. In conclusion, a new regulatory function for LZ-8 was identified, and the molecular mechanisms underlying this function were elucidated.

19.
Carcinogenesis ; 34(4): 874-84, 2013 Apr.
Article in English | MEDLINE | ID: mdl-23275155

ABSTRACT

Fucoidan, a polysaccharide extracted from brown seaweeds, reduces tumor cell proliferation. Fucoidan inhibits the growth of breast cancer cells such as 4T1 and MDA-MB-231 and decreases their cell colony formation. Moreover, fucoidan reduces metastatic lung nodules in 4T1 xenograft female Balb/c mice. The molecular network of transforming growth factor ß (TGFß) receptors (TGFRs) plays an important role in the regulation of the epithelial to mesenchymal transition (EMT) in cancer cells. Using 4T1 and MDA-MB-231 cells, we found that fucoidan effectively reverses TGFR-induced EMT morphological changes, upregulates epithelial markers, downregulates mesenchymal markers and decreases the expression of transcriptional repressors Snail, Slug and Twist. Moreover, fucoidan inhibits migration and invasion during the EMT, suggesting the involvement of TGFR-mediated signaling in breast cancer cells. Fucoidan decreases TGFRI and TGFRII proteins and affects downstream signaling molecules, including Smad2/3 phosphorylation and Smad4 expression. In order to elucidate how fucoidan decreases TGFRI and TGFRII proteins in MDA-MB-231 cells, we investigated ubiquitination activity downregulation of TGFRs. It was found that fucoidan enhances proteasome-mediated degradation/ubiquitination of TGFR. This study is the first to identify a novel mechanism for fucoidan antitumor activity, namely regulation of the EMT via modulation of TGFR/Smad-dependent signaling, which leads to an inhibition of breast cancer cell growth in vitro and in vivo. Our current findings indicate that fucoidan is a potential therapeutic agent for breast cancer and acts via an ubiquitin-dependent degradation pathway that affects the TGFR/Smad/Snail, Slug, Twist and EMT axes.


Subject(s)
Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Epithelial-Mesenchymal Transition/drug effects , Neoplasm Metastasis/drug therapy , Polysaccharides/pharmacology , Receptors, Transforming Growth Factor beta/metabolism , Animals , Biomarkers, Tumor , Cell Line, Tumor , Cell Movement/drug effects , Cell Proliferation/drug effects , Cell Transformation, Neoplastic/drug effects , Female , Lung Neoplasms/drug therapy , Lung Neoplasms/secondary , MCF-7 Cells , Mice , Mice, Inbred BALB C , Neoplasm Invasiveness , Neoplasm Transplantation , Signal Transduction/drug effects , Smad Proteins/metabolism , Snail Family Transcription Factors , Transcription Factors/biosynthesis , Transplantation, Heterologous , Twist-Related Protein 1/biosynthesis , Ubiquitin/metabolism , Ubiquitination/drug effects
20.
J Cell Physiol ; 227(8): 3063-71, 2012 Aug.
Article in English | MEDLINE | ID: mdl-22021110

ABSTRACT

Ganoderma lucidum is used in traditional Chinese medicine to prevent or treat a variety of diseases, including cardiovascular disorders. We previously demonstrated that a glucan-containing extract of Reishi polysaccharides (EORP) has the potent anti-inflammatory action of reducing ICAM-1 expression in lipopolysaccharide (LPS)-treated human aortic smooth muscle cells (HASMCs) and LPS-treated mice. In the present study, we examined whether EORP inhibited platelet-derived growth factor-BB (PDGF)-stimulated HASMC proliferation and the mechanism involved. EORP dose-dependently reduced cell numbers and DNA synthesis of PDGF-treated HASMCs in vitro. EORP also arrested cell cycle progression in the G0/G1 phase, and this was associated with decreased expression of cyclin D1, cyclin E, CDK2, CDK4, and p21(Cip1) and upregulation of the cyclin-dependent kinase inhibitor p27(Kip1). The anti-proliferative effect of EORP was partly mediated by downregulation of PDGF-induced JNK phosphorylation. In in vivo studies, the femoral artery of C57BL/6 mice was endothelial-denuded and the mice were fed a diet containing 100 mg/kg/day of EORP. On day 14, both cell proliferation (proliferating cell nuclear antigen-positive cells) in the neointima and the neointima/media area ratio (0.67 ± 0.03 vs. 1.46 ± 0.30) were significantly reduced. Our data show that EORP interferes with the mitogenic activation of JNK, preventing entry of HASMCs into the cell cycle in vitro and reducing cell proliferation in the neointima and decreasing the neointimal area in vivo. Thus, EORP may represent a safe and effective novel approach to the prevention and treatment of vascular proliferative diseases.


Subject(s)
Cell Proliferation/drug effects , Drugs, Chinese Herbal/pharmacology , Myocytes, Smooth Muscle , Neointima , Polysaccharides/pharmacology , Reishi , Animals , Aorta/cytology , Cell Cycle/drug effects , Cell Death/drug effects , Drugs, Chinese Herbal/chemistry , Gene Knockdown Techniques , Humans , Lipopolysaccharides/pharmacology , MAP Kinase Kinase 4/genetics , MAP Kinase Kinase 4/metabolism , Male , Mice , Mice, Inbred C57BL , Myocytes, Smooth Muscle/drug effects , Myocytes, Smooth Muscle/pathology , Phosphorylation/drug effects , Platelet-Derived Growth Factor/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL
...