Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 15 de 15
Filter
1.
Int Arch Allergy Immunol ; 182(12): 1143-1154, 2021.
Article in English | MEDLINE | ID: mdl-34649239

ABSTRACT

BACKGROUND: Asthma animal models provide valuable information about the pathogenesis and the treatment of asthma. An ovalbumin (OVA)/complete Freund's adjuvant (CFA)-sensitized model was developed to induce neutrophil-dominant asthma and to investigate whether fungal immunomodulatory peptide-fve (FIP-fve) could improve asthma features in the OVA/CFA-sensitized model. METHODS: We used female BALB/c mice and sensitized them intraperitoneally with OVA/CFA on days 1, 2, and 3. On days 14, 17, 21, 24, and 27, they were challenged with intranasal OVA. The airway hyper-responsiveness (AHR) was detected by BUXCO, inflammatory cells were stained with Liu's stain, the cytokines were detected using ELISA, and the airway inflammation was analyzed with hematoxylin and eosin stain. RESULTS: According to the results, OVA/CFA sensitization could induce AHR, high levels of IgE, and inflammatory cells especially neutrophils infiltration in the lung and airway inflammation. IL-4, IL-5, IL-6, IL-8, IL-10, IL-13, IL-17, IL-25, IL-33, and transforming growth factor-ß (TGF-ß) increased in the OVA/CFA-sensitized mice. OVA/CFA-sensitized mice treated with FIP-fve not only increased IL-12 and IFN-γ but also decreased IL-4, IL-5, IL-6, IL-8, IL-13, IL-17, IL-25, IL-33, and TGF-ß in the bronchoalveolar lavage fluid. Moreover, FIP-fve significantly decreased neutrophil infiltration in the lung. CONCLUSION: The OVA/CFA model induced neutrophilic asthma successfully, and FIP-fve improved neutrophil-dominant asthma.


Subject(s)
Anti-Asthmatic Agents/therapeutic use , Asthma/drug therapy , Fungal Proteins/therapeutic use , Neutrophils/drug effects , Animals , Anti-Asthmatic Agents/pharmacology , Asthma/immunology , Asthma/pathology , Biomarkers/metabolism , Cytokines/metabolism , Enzyme-Linked Immunosorbent Assay , Female , Freund's Adjuvant/immunology , Fungal Proteins/pharmacology , Immunoglobulin E/metabolism , Mice , Mice, Inbred BALB C , Neutrophil Infiltration/drug effects , Neutrophil Infiltration/immunology , Neutrophils/immunology , Ovalbumin/immunology , Real-Time Polymerase Chain Reaction , Treatment Outcome
2.
Nutrients ; 13(8)2021 Aug 15.
Article in English | MEDLINE | ID: mdl-34444952

ABSTRACT

Cisplatin-induced nephrotoxicity is associated with gut microbiota disturbance. The present study aimed to investigate whether supplementation of Lactobacillus reuteri and Clostridium butyricum (LCs) had a protective effect on cisplatin-induced nephrotoxicity through reconstruction of gut microbiota. Wistar rats were given different treatments: control, cisplatin (Cis), cisplatin + C. butyricum and L. reuteri (Cis+LCs), and C. butyricum and L. reuteri (LCs). We observed that cisplatin-treated rats supplemented with LCs exhibited significantly decreased renal inflammation (KIM-1, F4/80, and MPO), oxidative stress, fibrosis (collagen IV, fibronectin, and a-SMA), apoptosis, concentration of blood endotoxin and indoxyl sulfate, and increased fecal butyric acid production compared with those without supplementation. In addition, LCs improved the cisplatin-induced microbiome dysbiosis by maintaining a healthy gut microbiota structure and diversity; depleting Escherichia-Shigella and the Enterobacteriaceae family; and enriching probiotic Bifidobacterium, Ruminococcaceae, Ruminiclostridium_9, and Oscillibacter. Moreover, the LCs intervention alleviated the cisplatin-induced intestinal epithelial barrier impairment. This study indicated LCs probiotic serves as a mediator of the gut-kidney axis in cisplatin-induced nephrotoxicity to restore the intestinal microbiota composition, thereby suppressing uremic toxin production and enhancing butyrate production. Furthermore, the renoprotective effect of LCs is partially mediated by increasing the anti-inflammatory effects and maintaining the integrity of the intestinal barrier.


Subject(s)
Clostridium butyricum , Gastrointestinal Microbiome , Limosilactobacillus reuteri , Nephritis/microbiology , Probiotics/administration & dosage , Animals , Butyric Acid/metabolism , Cisplatin/toxicity , Disease Models, Animal , Inflammation , Kidney/microbiology , Nephritis/chemically induced , Nephritis/therapy , Rats , Rats, Wistar
3.
J Cell Physiol ; 236(2): 1148-1157, 2021 02.
Article in English | MEDLINE | ID: mdl-32686156

ABSTRACT

Saracatinib is an oral Src-kinase inhibitor and has been studied in preclinical models and clinical trials of cancer therapy. GMI, a fungal immunomodulatory protein from Ganoderma microsporum, possesses antitumor capacity. The aim of this study is to evaluate the cytotoxic effect of combination treatment with saracatinib and GMI on parental and pemetrexed-resistant lung cancer cells. Cotreatment with saracatinib and GMI induced synergistic and additive cytotoxic effect in A549 and A400 cells by annexin V/propidium iodide assay and combination index. Using western blot assay, saracatinib, and GMI combined treatment synergistically induced caspase-7 activation in A549 cells. Different from A549 cells, saracatinib and GMI cotreatment markedly increased LC3B-II in A400 cells. ATG5 silencing abolished the caspase-7 activation and reduced cell death in A549 cells after cotreatment. This is the first study to provide a novel strategy of treating lung cancer with or without drug resistance via combination treatment with GMI and saracatinib.


Subject(s)
Autophagy-Related Protein 5/genetics , Benzodioxoles/pharmacology , Caspase 7/genetics , Enzyme Inhibitors/pharmacology , Lung Neoplasms/drug therapy , Quinazolines/pharmacology , src-Family Kinases/genetics , A549 Cells , Animals , Antineoplastic Combined Chemotherapy Protocols/pharmacology , Apoptosis/drug effects , Autophagy/drug effects , Autophagy-Related Protein 5/antagonists & inhibitors , Cell Proliferation/drug effects , Fungal Proteins/chemistry , Fungal Proteins/pharmacology , Ganoderma/chemistry , Humans , Immunologic Factors/pharmacology , Lung Neoplasms/genetics , Lung Neoplasms/pathology , Mice , Synthetic Lethal Mutations/drug effects , Xenograft Model Antitumor Assays , src-Family Kinases/antagonists & inhibitors
4.
Neurogastroenterol Motil ; 32(6): e13803, 2020 06.
Article in English | MEDLINE | ID: mdl-31989744

ABSTRACT

BACKGROUND: Cisplatin is a widely used antineoplastic drug. However, cisplatin-induced dyspepsia syndromes, including delayed gastric emptying, gastric distension, early satiety, nausea, and vomiting, often force patients to take doses lower than those prescribed or even refuse treatment. D-methionine has an appetite-enhancing effect and alleviates weight loss during cisplatin treatment. METHODS: This work established a model of anorexia and dyspepsia symptoms with intraperitoneal injection of cisplatin (5 mg/kg) once a week for three cycles. Presupplementation with or without D-methionine (300 mg/kg) was performed. Orexigenic and anorexigenic hormones (ghrelin, leptin, and glucagon-like peptide-1), tryptophan hydroxylase 1 (TPH1), 5-hydroxytryptamine receptors (5-HT2C and 5-HT3 ), and hypothalamic feeding-related peptides were measured by immunohistochemistry staining, enzyme-linked immunosorbent assay, and real-time PCR assay. KEY RESULTS: Cisplatin administration caused marked decrease in appetite and body weight, promoted adipose and fat tissue atrophy, and delayed gastric emptying and gastric distension, and D-methionine preadministration prior to cisplatin administration significantly ameliorated these side effects. Besides, cisplatin induced an evident increase in serum ghrelin level, TPH1 activity, and 5-HT3 receptor expression in the intestine and decreased plasma leptin levels and gastric ghrelin mRNA gene expression levels. D-methionine supplementation recovered these changes. The expression of orexigenic neuropeptide Y/agouti-related peptide and anorexigenic cocaine- and amphetamine-regulated transcript proopiomelanocortin neurons were altered by D-methionine supplementation in cisplatin-induced anorexia rats. CONCLUSIONS AND INFERENCES: D-methionine supplementation prevents cisplatin-induced anorexia and dyspepsia syndrome possibly by attenuating intestinal tryptophan hydroxylase 1 activity and increasing plasma leptin concentration. Therefore, D-methionine can be used as an adjuvant therapy for treating cisplatin-induced adverse effects.


Subject(s)
Anorexia/chemically induced , Antineoplastic Agents/administration & dosage , Cisplatin/administration & dosage , Dyspepsia/chemically induced , Intestinal Mucosa/drug effects , Leptin/blood , Methionine/administration & dosage , Tryptophan Hydroxylase/metabolism , Animals , Ghrelin/blood , Hypothalamus/drug effects , Hypothalamus/metabolism , Intestinal Mucosa/metabolism , Male , Rats, Wistar , Receptors, Serotonin, 5-HT3/metabolism
5.
Integr Cancer Ther ; 18: 1534735419833795, 2019.
Article in English | MEDLINE | ID: mdl-30879354

ABSTRACT

5-Fluorouracil (5-FU) is used in the treatment of head and neck cancer patients. However, adverse effects experienced such as mucositis and poor appetite may lead to interruption in chemotherapy. The aim of this study is to evaluate the efficacy of GMI, one fungal immunomodulatory protein found in Ganoderma microsporum, for mucositis induced by 5-FU in a mouse model. Mice were administered 5-FU intraperitoneally for 4 days per cycle for a total of 2 chemotherapy cycles. In addition, mice were pretreated with GMI or phosphate-buffered saline 3 days before 5-FU intraperitoneal injection and daily until day 14. On histological analysis, GMI prevented 5-FU-induced damage to the intestinal mucosa and tongue epithelium. We also demonstrated that GMI enhanced the cytotoxicity of 5-FU in 2 oral cancer cell lines, while GMI could not promote this effect in an oral normal cell. In conclusion, GMI alleviates 5-FU-induced damage and decelerates cell death in normal alimentary tract tissue.


Subject(s)
Antimetabolites, Antineoplastic/adverse effects , Fluorouracil/adverse effects , Ganoderma/metabolism , Immunologic Factors/pharmacology , Intestinal Mucosa/drug effects , Mucositis/chemically induced , Mucositis/drug therapy , Animals , Apoptosis/drug effects , Disease Models, Animal , Male , Mice , Mice, Inbred BALB C , Microsporum/drug effects
6.
Integr Cancer Ther ; 18: 1534735419828832, 2019.
Article in English | MEDLINE | ID: mdl-30789014

ABSTRACT

Cisplatin induces anorexia, weight loss, loss of adipose tissue, skeletal muscle atrophy, and serious adverse effects that can cause premature termination of chemotherapy. The aim of this study was to use an animal model to assess cisplatin therapy (3 cycles) with and without d-methionine to investigate its protective effects on cisplatin-induced anorexia and skeletal muscle wasting. Wistar rats were divided into 3 groups and treated as follows: saline as control (group 1), intraperitoneal cisplatin once a week for 3 weeks (group 2), and intraperitoneal cisplatin once a week for 3 weeks plus oral administration of d-methionine (group 3). Tissue somatic index (TSI), gastric emptying index (GEI), and feeding efficiency were measured. Both hepatic lipid metabolism and muscle atrophy-related gene expressions and C2C12 myotubes were determined by polymerase chain reaction. Micro-computed tomography (micro-CT) was used to conduct assessment of bone microarchitecture indices. Pathological changes of the gastric mucosa were assessed by hematoxylin and eosin staining after euthanizing the animals. d-Methionine increased food intake, weight gain, gastric emptying, and feeding efficiency, as well as decrease stomach contents, after cisplatin injections. Cisplatin caused shortening of myofibers. Cisplatin-induced muscle mass wasting was mediated by the elevation of mRNA expressions of MAFbx and MuRF-1 in ubiquitin ligases in muscle tissue homogenate. The mRNA expressions of MyoD and myogenin, markers of muscle differentiation, declined following cisplatin administration. The administration of d-methionine not only led to significant improvements in myofiber diameter and cross-sectional fiber areas but also reversed muscle atrophy-related gene expression. However, there were no significant changes in stomach histology or microarchitecture of trabecular bone among the study groups. The results indicate that d-methionine has an appetite-enhancing effect and ameliorates cisplatin-induced adipose and muscle tissue loss during cisplatin-based chemotherapy.


Subject(s)
Cisplatin/adverse effects , Cisplatin/pharmacology , Methionine/pharmacology , Muscle, Skeletal/drug effects , Muscular Atrophy/chemically induced , Muscular Atrophy/drug therapy , Animals , Anorexia/chemically induced , Anorexia/drug therapy , Cell Line , Male , Mice , RNA, Messenger/metabolism , Rats , Rats, Wistar , Weight Loss/drug effects
7.
Phytomedicine ; 56: 94-102, 2019 Mar 15.
Article in English | MEDLINE | ID: mdl-30668358

ABSTRACT

BACKGROUND: Epigenetic therapy is a promising popular treatment modality for various cancers. Histone modification and miRNA should not be underestimated in lung cancer. This study aimed to investigate whether chidamide, a histone deacetylase inhibitor (HDACi), which inhibits telomerase activity and induces cell cycle arrest, influences ROS and miRNA production in non-small cell lung cancer (NSCLC) cells. METHODS: H1355 and A549 were treated with chidamide. The analysis of DNA content was measured by FACSCalibur equipped with a 488 nm laser. H1355 cells were transfected with miR-129-3p mimic by Lipofectamine2000. Telomerase activity was performed on the telomeric repeat amplification protocol (TRAP) assay. Detection of thymidylate synthase (TS), p21, p53, pRB, and ß-actin, were performed by western blot analysis. RESULTS: Our data showed that expression of TS, p21, and pRB were altered in the presence of chidamide by PCR and western blot. Using BrdU-incorporation analysis, we found that chidamide induced G1 arrest through the regulation of the TS gene by miR-129-3p. Chidamide was shown to suppress telomerase activity in the TRAP assay and reduced the expression of human telomerase reverse transcriptase (hTERT) by PCR and q-PCR in H1355 and A549 cells. Chidamide increased the generation of reactive oxygen species (ROS) by flow cytometry. N-acetyl cysteine (NAC), a ROS scavenger, attenuated chidamide-induced telomerase activity inhibition. CONCLUSION: Chidamide repressed telomerase activity through ROS accumulation and cell cycle arrest by miR-129-3p upregulation in both H1355 and A549 cells. This is the first study to demonstrate that chidamide induces miR-129-3p upregulation and ROS accumulation, leading to cell cycle arrest.


Subject(s)
Aminopyridines/pharmacology , Antineoplastic Agents/pharmacology , Benzamides/pharmacology , Carcinoma, Non-Small-Cell Lung/drug therapy , Lung Neoplasms/drug therapy , MicroRNAs/genetics , A549 Cells , Carcinoma, Non-Small-Cell Lung/genetics , Cell Cycle Checkpoints/drug effects , Cell Cycle Checkpoints/genetics , Cell Line, Tumor , Gene Expression Regulation, Neoplastic/drug effects , Histone Deacetylase Inhibitors/pharmacology , Humans , Lung Neoplasms/genetics , Reactive Oxygen Species/metabolism , Telomerase/genetics
8.
Phytomedicine ; 51: 39-47, 2018 Dec 01.
Article in English | MEDLINE | ID: mdl-30466626

ABSTRACT

BACKGROUND: In terms of melanoma, recent advances have been made in target therapies and immune checkpoint inhibitors, but durable remission is rare. Ganoderma immunomodulatory proteins (GMI) induce a cytotoxic effect in cancer cells via autophagy. However, the role of GMI in melanoma is not clear. PURPOSE: The aims of this study are to investigate the inhibiting effects of GMI combined with chidamide on survival and metastases of melanoma cells via integrin-related signaling pathway and to propose strategies for combining GMI and chidamide using animal model. METHODS: Cell viability was measured by cell CCK-8. The activities of apoptosis- and migration-related proteins were detected on Western blot. Flow cytometry was used to analyze cell cycle distribution and sub-G1 fraction in treated melanoma cells. To evaluate the activity of combination GMI and chidamide treatment, an in vivo anti-tumor metastasis study was performed. RESULTS: GMI combined with chidamide additively induced apoptosis. GMI inhibited the expressions of Integrin α5, αV, ß1, and ß3. The level of p-FAK was inhibited by GMI. Combination treatment of GMI and chidamide decreased survivin and increased cleaved caspase-7 and LC3 II/I. Integrin-αV overexpression activated p-FAK pathways in A375.S2 cells. GMI significantly inhibited cell growth and migration of A375.S2 cells on wound healing assay. In vivo, GMI combined with chidamide suppressed distal tumor metastasis. CONCLUSION: GMI inhibits the migration and growth of melanoma cells via integrin-related signaling pathway. GMI and chidamide induces apoptosis. In vivo, GMI and chidamide additively reduce distant metastases. GMI and chidamide are potential immunotherapeutic adjuvant for metastatic melanoma.


Subject(s)
Aminopyridines/pharmacology , Apoptosis/drug effects , Benzamides/pharmacology , Ganoderma/chemistry , Melanoma, Experimental/drug therapy , Signal Transduction/drug effects , Animals , Cell Cycle , Cell Line, Tumor , Cell Movement/drug effects , Cell Proliferation/drug effects , Cell Survival/drug effects , Down-Regulation , Human Umbilical Vein Endothelial Cells , Humans , Integrin alphaV/metabolism , Male , Melanoma, Experimental/pathology , Mice, Inbred C57BL , Neoplasm Metastasis
9.
Phytomedicine ; 46: 193-198, 2018 Jul 15.
Article in English | MEDLINE | ID: mdl-30097118

ABSTRACT

BACKGROUND: Hepatitis C virus (HCV) is a globally prevalent pathogen and a leading cause of death and morbidity. Traditional therapy with pegylated interferon- and ribavirin has had only limited success, with some adverse effects. Direct-acting antivirals (DAAs) are effective in suppressing HCV replication, but are expensive. PURPOSE: Hypericin has been reported to be a good antiviral agent for inhibiting HCV replication, however, little is known about its mechanisms of action. The aim of this study is to elucidate the mode of action of hypericin in Ava5 human hepatoma cell line (Huh7 derivative) harboring HCV subgenomic replicon RNA. METHODS: To determine the non-structure protein 5A (NS5A) mRNA and NS3 protein expression levels, real-time PCR and Western blot analysis were performed, respectively. To investigate how hypericin inhibits HCV replication, 5-aza-2'-deoxycytidine (5-Aza-dC) and chidamide were used for determining histone modification. Furthermore, shRNA was applied to confirm the role of heme oxygenase (HO-1) in HCV repression. RESULTS: Hypericin in experiment were tested and showed no cytotoxicity. Hypericin reduced HO-1 and NS5A in a time- and dose- dependent manner. Chidamide, but not 5-Aza-dc, restored hypericin-induced reduction in HCV NS3 expression and reversed HO-1 expression in Ava5 cells. LY294002 inhibited HCV replication via HO-1 down-regulation. Constitutive expressed p-AKT was not involved in hypericin-induced reduction in HCV replication. In addition, shHO-1 inhibited HCV replication. CONCLUSION: In conclusion, hypericin inhibits HCV replication via down-regulation of HO-1 expression and deacetylation.


Subject(s)
Antiviral Agents/pharmacology , Hepacivirus/drug effects , Perylene/analogs & derivatives , Virus Replication/drug effects , Anthracenes , Cell Line, Tumor , Down-Regulation , Heme Oxygenase-1/metabolism , Hepacivirus/physiology , Humans , Perylene/pharmacology , Viral Nonstructural Proteins/metabolism
10.
Integr Cancer Ther ; 17(3): 813-824, 2018 09.
Article in English | MEDLINE | ID: mdl-29430988

ABSTRACT

D-methionine is a sulfur-containing amino acid that can act as a potent antioxidant. Anorexia and nephrotoxicity are side effects of cisplatin. The protective effects of D-methionine on cisplatin-induced anorexia and renal injury were investigated. The model of chronic cisplatin administration (5 mg/kg body weight) involved intraperitoneal injection on days 1, 8, and 15 and oral D-methionine (300 mg/kg body weight) coadministration daily for 20 days. On the 21st day of treatment, food intake and body weight in the cisplatin-treated group significantly decreased by 52% and 31%, respectively, when compared with a control group. D-methionine coadministration with cisplatin decreased food intake and body weight by 29% and 8%, respectively. In cisplatin-treated rats, white blood cell, mean corpuscular volume, and platelet values significantly decreased, while mean corpuscular hemoglobin concentration significantly increased by 8.6% when compared with control rats. Cisplatin administration resulted in significantly decreased feeding efficiency, elevated renal oxidative stress, and reduced antioxidative activity. Leukocyte infiltration, tubule vacuolization, tubular expansion, and swelling were observed in the kidneys of cisplatin-treated rats. Oral D-methionine exhibited an antianorexic effect, with improvement in food intake, feeding efficiency, and hematological toxicities, as well as a protective effect against nephrotoxicity by elevated antioxidative activity. D-methionine may serve as a chemoprotectant in patients receiving cisplatin as part of a chemotherapy regimen.


Subject(s)
Anorexia/chemically induced , Anorexia/prevention & control , Cisplatin/adverse effects , Methionine/pharmacology , Renal Insufficiency, Chronic/chemically induced , Renal Insufficiency, Chronic/prevention & control , Weight Loss/drug effects , Animals , Anorexia/blood , Appetite/drug effects , Blood Cell Count , Blood Cells/drug effects , Blood Cells/pathology , Cisplatin/administration & dosage , Eating/drug effects , Hemoglobins/drug effects , Hemoglobins/metabolism , Male , Methionine/administration & dosage , Protective Agents/administration & dosage , Protective Agents/pharmacology , Rats , Rats, Wistar , Renal Insufficiency, Chronic/blood
11.
J Microbiol Immunol Infect ; 50(3): 297-306, 2017 Jun.
Article in English | MEDLINE | ID: mdl-26427878

ABSTRACT

BACKGROUND/PURPOSE: House dust mite (HDM) is well known as one of the major indoor allergens that trigger allergic inflammation, especially asthma, and accounts for 85% of all cases. So far, asthma has been thought of as a condition of imbalance between T helper (Th)1 and Th2. Fungal immunomodulatory protein-Flammulina velutipes (FIP-fve) has been seemingly demonstrated to modulate the response to Th1 cytokine production. The aim of this study was to investigate if the oral administration of FIP-fve can inhibit HDM-induced asthma inflammation in the mouse model. METHODS: We divided the mice (female BALB/c, 4-6 weeks) into four groups: the prevention group, which consisted of mice sensitized by HDM (intraperitoneally on Day 1, Day 7, and Day 14, and intranasally on Day 14, Day 17, Day 21, Day 24, and Day 27) fed with FIP-fve from Day 1 to Day 14; the treatment group, which comprised mice that received treatment from Day 14 to Day 28; the positive control (PC, sensitized by HDM fed without FIP-fve) group; and the negative control group (NC, nonsensitized). Airway hyperresponsiveness induced by methacholine challenge was determined using whole-body barometric plethysmography. In addition, cytokines were analyzed from bronchoalveolar lavage fluid and serum. Histopathological studies and Liu's staining method in mice lungs were also performed. RESULTS: The results showed that both pre- and posttreated FIP-fve groups had significantly reduced airway hyperresponsiveness compared with the PC group after methacholine challenge. In addition, a significantly decreased level of HDM-specific immunoglobulin E in serum and decreased production of Th2 cytokines in bronchoalveolar lavage fluid and serum were observed in these two FIP-fve fed groups. Moreover, more decreased amounts of infiltrating inflammatory cells were present in the lungs of FIP-fve fed groups than those of the PC group. CONCLUSION: Oral FIP-fve had an anti-inflammatory effect on the acute phase of the airway inflammatory process induced by HDM in the mouse model and might have a potentially therapeutic role for allergic airway diseases.


Subject(s)
Asthma/drug therapy , Flammulina/chemistry , Fungal Proteins/administration & dosage , Immunologic Factors/administration & dosage , Administration, Oral , Animals , Disease Models, Animal , Female , Fungal Proteins/isolation & purification , Histocytochemistry , Immunologic Factors/isolation & purification , Mice, Inbred BALB C , Plethysmography , Pyroglyphidae/immunology , Treatment Outcome
12.
Phytomedicine ; 23(13): 1566-1573, 2016 Dec 01.
Article in English | MEDLINE | ID: mdl-27823620

ABSTRACT

BACKGROUND: Chloroquine, a lysosomal inhibitor, is used for malaria, rheumatoid arthritis, and lupus erythematosus therapy. In our previous study, FIP-gts, an immunomodulatory protein from Ganoderma tsugae, inhibited cell viability in lung cancer cells and urothelial cancer cells. Urothelial carcinoma is the most common type of bladder cancer. Cisplatin resistance is an important issue in urothelial carcinoma therapy. PURPOSE: The aim of this study is to investigate the effect of combination treatment with FIP-gts and chloroquine on cytotoxicity to resensitize the cisplatin-resistant cells. METHODS: FIP-gts and chloroquine cytotoxicity were determined by evaluating CCK-8 assay. Cell death pathways, ROS and cell cycle arrested were analysed through flow cytometry and Western blot. ShRNA targeting to autophagy-related genes were tested to evaluate their autophagic cell death for resistant urothelial cells. RESULTS: Using CCK-8 assay, chloroquine increased FIP-gts-induced cytotoxicity in parental and cisplatin-resistant urothelial cancer cell lines. On flow cytometry, chloroquine enhanced FIP-gts-mediated sub-G1 accumulation, annexin V positive signal and mitochondrial membrane potential loss. Caspase-3/PARP cascade and z-VAD-fmk were performed to prove that FIP-gts and chloroquine induced caspase-independent cell death. Using H2DCFDA staining and flow cytometry, FIP-gts and chloroquine did not induce ROS production. N-acetyl cysteine, a ROS scavenger, inhibited the cytotoxicity and LC3-II accumulation in FIP-gts and chloroquine-treated N/P cells. To elucidate the role of autophagy in caspase-independent cell death by FIP-gts and chloroquine, LC3 shRNA were used to inhibit autophagy in N/P cells. The capabilities of FIP-gts and chloroquine to induce cytotoxicity and sub-G1 phase accumulation were abolished in autophagy-defective cells. This is the first study to reveal the novel function of FIP-gts in triggering caspase-independent cell death in cisplatin-resistant urothelial cancer cells. CONCLUSION: Chloroquine enhanced FIP-gts-induced autophagy dependent caspase-independent cell death via abundant autophagosome accumulation. Combination treatment with FIP-gts and chloroquine may provide a new strategy for urothelial cancer therapy.


Subject(s)
Autophagy/drug effects , Chloroquine/pharmacology , Cisplatin/pharmacology , Fungal Proteins/pharmacology , Immunologic Factors/pharmacology , Urologic Neoplasms/drug therapy , Amino Acid Chloromethyl Ketones/pharmacology , Antineoplastic Combined Chemotherapy Protocols/pharmacology , Apoptosis/drug effects , Caspases/metabolism , Cell Cycle/drug effects , Cell Line, Tumor , Cell Survival/drug effects , Chloroquine/administration & dosage , Drug Resistance, Neoplasm/drug effects , Fungal Proteins/administration & dosage , Ganoderma/chemistry , Humans , Membrane Potential, Mitochondrial/drug effects , Urologic Neoplasms/pathology
13.
PLoS One ; 10(5): e0125774, 2015.
Article in English | MEDLINE | ID: mdl-25946033

ABSTRACT

Chemoresistance in cancer therapy is an unfavorable prognostic factor in non-small cell lung cancer (NSCLC). Elevation of intracellular calcium level in multidrug resistant (MDR) sublines leads to sensitization of MDR sublines to cell death. We demonstrated that a fungal protein from Ganoderma microsporum, GMI, elevates the intracellular calcium level and reduces the growth of MDR subline via autophagy and apoptosis, regardless of p-glycoprotein (P-gp) overexpression, in mice xenograft tumors. In addition, we examined the roles of autophagy in the death of MDR A549 lung cancer sublines by GMI, thapsigargin (TG) and tunicamycin (TM) in vitro. Cytotoxicity of TG was inhibited by overexpressed P-gp. However, TM-induced death of MDR sublines was independent of P-gp level. Combinations of TG and TM with either docetaxel or vincristine showed no additional cytotoxic effects on MDR sublines. TG- and TM-mediated apoptosis of MDR sublines was demonstrated on Annexin-V assay and Western blot and repressed by pan-caspase inhibitor (Z-VAD-FMK). Treatment of MDR sublines with TG and TM also augmented autophagy with accumulation of LC3-II proteins, breakdown of p62 and formation of acidic vesicular organelles (AVOs). Inhibition of ATG5 by shRNA silencing significantly reduced autophagy and cell death but not apoptosis following TG or TM treatment. GMI treatment inhibited the phosphorylation of Akt/S473 and p70S6K/T389. Interestingly, the phosphorylation of ERK was not associated with GMI-induced autophagy. We conclude that autophagy plays a pro-death role in acquired MDR and upregulation of autophagy by GMI via Akt/mTOR inhibition provides a potential strategy for overcoming MDR in the treatment of lung cancers.


Subject(s)
Antineoplastic Agents/therapeutic use , Apoptosis/drug effects , Autophagy/drug effects , Carcinoma, Non-Small-Cell Lung/drug therapy , Fungal Proteins/therapeutic use , Lung Neoplasms/drug therapy , ATP Binding Cassette Transporter, Subfamily B, Member 1/metabolism , Amino Acid Chloromethyl Ketones/pharmacology , Animals , Autophagy-Related Protein 5 , Cell Line, Tumor , Docetaxel , Drug Resistance, Multiple , Drug Resistance, Neoplasm , Ganoderma , Humans , Male , Medicine, Chinese Traditional , Mice , Mice, Inbred NOD , Microtubule-Associated Proteins/genetics , Proto-Oncogene Proteins c-akt/antagonists & inhibitors , RNA Interference , RNA, Small Interfering , Ribosomal Protein S6 Kinases, 70-kDa/antagonists & inhibitors , TOR Serine-Threonine Kinases/antagonists & inhibitors , Taxoids/therapeutic use , Thapsigargin/therapeutic use , Tunicamycin/therapeutic use , Vincristine/therapeutic use , Xenograft Model Antitumor Assays
14.
Food Chem Toxicol ; 50(3-4): 558-66, 2012 Mar.
Article in English | MEDLINE | ID: mdl-22210353

ABSTRACT

Oral cancer mortality has increased during the last decade due to the difficulties in treating related metastasis. Dioscorea nipponica Makino, a popular folk medicine, exerts anti-obesity and anti-inflammation properties. However, the effect of this folk medicine on metastasis of oral cancer has yet to be fully elucidated. The present study demonstrates that D. nipponica extracts (DNE), at a range of concentrations (0-50 µg/mL), concentration-dependently inhibited migration/invasion capacities of human oral cancer cells, HSC-3, without cytotoxic effects. The anti-migration effect of DNE was also observed in two other OSCC cell lines, Ca9-22 and Cal-27. Zymography, real time PCR, and Western blotting analyses revealed that DNE inhibited matrix metalloproteinase-2 (MMP-2) enzyme activity, and RNA and protein expression. The inhibitory effects of DNE on MMP-2 proceeded by up-regulating tissue inhibitor of metalloproteinase-2 (TIMP-2), as well as suppressing nuclear translocation and DNA binding activity of cAMP response element-binding (CREB) and activating protein-1 (AP-1) on the MMP-2 promoter in HSC-3 cells. In conclusion, DNE inhibited the invasion of oral cancer cells and may have potential use as a chemopreventive agent against oral cancer metastasis.


Subject(s)
Cyclic AMP Response Element-Binding Protein/metabolism , Dioscorea/chemistry , Matrix Metalloproteinase Inhibitors , Mouth Neoplasms/pathology , Plant Extracts/pharmacology , Transcription Factor AP-1/metabolism , Transcription, Genetic , Base Sequence , Cell Line, Tumor , Chromatin Immunoprecipitation , DNA Primers , Flow Cytometry , Humans , Matrix Metalloproteinase 2/genetics , Mouth Neoplasms/enzymology , Real-Time Polymerase Chain Reaction
15.
Pharm Biol ; 48(11): 1302-9, 2010 Nov.
Article in English | MEDLINE | ID: mdl-20738166

ABSTRACT

CONTEXT: Tianhua (TH-R) is extracted from Trichosanthes kirilowii Maxim (Cucurbitaceae) containing trichosanthin, a traditional Chinese medicine, which has been locally reported to have good anticancer effects in vivo in both animal and human models. However, there have been several reports that trichosanthin has an anticancer effect involving apoptosis. OBJECTIVE: To investigate other anticancer effects of TH-R, various tumorigenesis parameters were verified. MATERIALS AND METHODS: Telomerase activity, anti-apoptosis, anti-migration and immunomodulatory activity were estimated by telomeric repeat amplification protocol assay (TRAP), flow cytometry, Boyden chamber assay and ELISA assay, respectively. RESULTS: In our studies, we are the first to find that TH-R had a cytotoxic effect on lung cancer cells in MTS assays; it could change the cell cycle distribution of human lung cancer cells (A549 cell line) and induce apoptosis. Further anti-telomerase effects in human lung adenocarcinoma A549 cells using the TRAP assay were noted. TH-R also had an aggregation effect on peripheral blood lymphocytes, but no effect on stimulating peripheral lymphocytes to produce human interferon-γ(IFN-γ). TH-R could inhibit the migration, or metastatic ability, of A549 cells by Boyden chamber assay. In the oral feeding therapy of an in vivo mouse model, there was an initial inhibition of A549 cancer cell growth, but no statistical difference after one month of therapy. DISCUSSION AND CONCLUSION: It has been proven that medicinal herbs such as Tianhua have positive effects against cancer through preventing or inhibiting the process of lung tumorigenesis.


Subject(s)
Antineoplastic Agents, Phytogenic/pharmacology , Immunologic Factors/pharmacology , Plant Extracts/pharmacology , Trichosanthin/pharmacology , Adenocarcinoma/drug therapy , Adenocarcinoma/immunology , Adenocarcinoma/pathology , Administration, Oral , Animals , Apoptosis/drug effects , Cell Line, Tumor , Cell Migration Inhibition/drug effects , Cell Survival/drug effects , Humans , Immunomodulation/drug effects , Interferon-gamma/biosynthesis , Lung Neoplasms/drug therapy , Lung Neoplasms/immunology , Lung Neoplasms/pathology , Male , Mice , Mice, Inbred BALB C , Mice, Nude , Neoplasm Transplantation , Telomerase/metabolism , Transplantation, Heterologous
SELECTION OF CITATIONS
SEARCH DETAIL