Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 130
Filter
Add more filters

Country/Region as subject
Publication year range
1.
Int J Med Mushrooms ; 26(2): 11-23, 2024.
Article in English | MEDLINE | ID: mdl-38421693

ABSTRACT

Ganoderic acid A (GAA) is one of the major triterpenoids in Ganoderma lucidum (GL). Accumulating evidence has indicated that GAA demonstrates multiple pharmacological effects and exhibits treatment potential for various neurological disorders. Here, the effects and mechanisms of GAA in the treatment of neurological disorders were evaluated and discussed through previous research results. By summarizing previous research results, we found that GAA may play a neuroprotective role through various mechanisms: anti-inflammatory, anti-oxidative stress, anti-apoptosis, protection of nerve cells, and regulation of nerve growth factor. Therefore, GAA is a promising natural neuroprotective agent and this review would contribute to the future development of GAA as a novel clinical candidate drug for treating neurological diseases.


Subject(s)
Heptanoic Acids , Lanosterol/analogs & derivatives , Nervous System Diseases , Neuroprotective Agents , Humans , Neuroprotective Agents/pharmacology , Neuroprotective Agents/therapeutic use , Lanosterol/pharmacology , Lanosterol/therapeutic use , Nervous System Diseases/drug therapy
2.
Food Funct ; 13(10): 5820-5837, 2022 May 23.
Article in English | MEDLINE | ID: mdl-35543349

ABSTRACT

Alcoholic liver injury is mainly caused by long-term excessive alcohol consumption and has become a global public threat to human health. It is well known that Ganoderma lucidum has excellent beneficial effects on liver function and lipid metabolism. The object of this study was to investigate the hepatoprotective effects of ganoderic acid A (GAA, one of the main triterpenoids in G. lucidum) against alcohol-induced liver injury and reveal the underlying mechanisms of its protective effects. The results showed that oral administration of GAA significantly inhibited the abnormal elevation of the liver index, serum total triglyceride (TG), cholesterol (TC), low-density lipoprotein cholesterol (LDL-C), aspartate aminotransferase (AST) and alanine aminotransferase (ALT) in mice exposed to alcohol intake, and also significantly protected the liver against alcohol-induced excessive lipid accumulation and pathological changes. Besides, alcohol-induced oxidative stress in the liver was significantly ameliorated by the dietary intervention of GAA through decreasing the hepatic levels of lactate dehydrogenase (LDH) and malondialdehyde (MDA), and increasing hepatic activities of catalase (CAT), superoxide dismutase (SOD), alcohol dehydrogenase (ADH), aldehyde dehydrogenase (ALDH), and hepatic levels of glutathione (GSH). In addition, GAA intervention evidently ameliorated intestinal microbial disorder by markedly increasing the abundance of Muribaculaceae, Prevotellaceae, Jeotgalicoccus, Bilophila, Family_XIII_UCG_001, Aerococcus, Ruminococcaceae_UCG_005, Harryflintia, Christensenellaceae, Rumonpcpccaceae, Prevotelaceae_UCG_001, Clostridiales_vadinBB60_group, Parasutterella and Bifidobacterium, but decreasing the proportion of Lactobacillus, Burkholderia_Caballeroria_Paraburkholderia, Escherichia_Shigella and Erysipelatoclostridium. Furthermore, liver metabolomics based on UPLC-QTOF/MS demonstrated that oral administration of GAA had a significant regulatory effect on the composition of liver metabolites in mice exposed to alcohol intake, especially the levels of the biomarkers involved in the metabolic pathways of riboflavin metabolism, glycine, serine and threonine metabolism, pyruvate metabolism, glycolysis/gluconeogenesis, biosynthesis of unsaturated fatty acids, synthesis and degradation of ketone bodies, fructose and mannose metabolism. Moreover, dietary supplementation of GAA significantly regulated the hepatic mRNA levels of lipid metabolism and inflammatory response related genes. Conclusively, these findings demonstrate that GAA has beneficial effects on alleviating alcohol-induced liver injury and is expected to become a new functional food ingredient for the prevention of alcoholic liver injury.


Subject(s)
Chemical and Drug Induced Liver Injury, Chronic , Reishi , Animals , Chemical and Drug Induced Liver Injury, Chronic/metabolism , Cholesterol/metabolism , Ethanol/pharmacology , Heptanoic Acids , Lanosterol/analogs & derivatives , Lanosterol/pharmacology , Lipid Metabolism , Liver/metabolism , Mice , Oxidative Stress
3.
J Ethnopharmacol ; 292: 115146, 2022 Jun 28.
Article in English | MEDLINE | ID: mdl-35304272

ABSTRACT

ETHNOPHARMACOLOGICAL RELEVANCE: Nonalcoholic fatty liver disease (NAFLD) is a prevalent liver disease, but currently has no specific medication in clinic. Antrodia cinnamomea (AC) is a medicinal fungus and it has been shown that AC can inhibit high fat diet (HFD)-induced lipid deposition in mouse livers, but the effective monomer in AC and mechanism against NAFLD remain unclear. It has been reported that aldehyde dehydrogenase 2 (ALDH2) activation shows protective effects on NAFLD. Our previous study demonstrates that AC and its monomer dehydroeburicoic acid (DEA) can upregulate the ALDH2 activity on alcoholic fatty liver disease mouse model, but it is not clear whether the anti-NAFLD effects of AC and DEA are mediated by ALDH2. AIM TO STUDY: To elucidate the active compound in AC against NAFLD, study whether ALDH2 mediates the anti-NAFLD effects of AC and its effective monomer. MATERIALS AND METHODS: WT mice, ALDH2-/- mice and ALDH2-/- mice re-expressed ALDH2 by lentivirus were fed with a methionine-choline deficient (MCD) diet or high fat diet (HFD) to induce NAFLD, and AC at the different doses (200 and/or 500 mg/kg body weight per day) was administrated by gavage at the same time. Primary hepatocytes derived from WT and ALDH2-/-mice were stimulated by oleic acid (OA) to induce lipid deposition, and the cells were treated with AC or DEA in the meantime. Lentivirus-mediated ALDH2-KD or ALDH2-OE were used to knock down or overexpress ALDH2 expression in HepG2 cells, respectively. Finally, the effects of DEA against NAFLD as well as its effects on upregulating liver ALDH2 and removing the harmful aldehyde 4-hydroxynonenal (4-HNE) were studied in the MCD diet-induced NAFLD mouse model. RESULTS: In WT mice fed with a MCD diet or HFD, AC administration reduced hepatic lipid accumulation, upregulated ALDH2 activity in mouse livers, decreased 4-HNE contents both in mouse livers and serum, inhibited lipogenesis, inflammation and oxidative stress and promoted fatty acid ß-oxidation. These effects were abolished in ALDH2 KO mice but could be restored by re-expression of ALDH2 by lentivirus. In primary hepatocytes of WT mice, AC and DEA inhibited OA-induced lipid accumulation and triglyceride (TG) synthesis, promoting the ß-oxidation of fatty acid in the meantime. However, these effects were lost in primary hepatocytes of ALDH2 KO mice. Moreover, the expression level of ALDH2 significantly affected the inhibitory effects of AC and DEA on OA-induced lipid deposition in HepG2 cells. The effects of AC and DEA on suppressing lipid deposition, inhibiting mitochondrial ROS levels, reducing TG synthesis, and promoting ß-oxidation of fatty acid were all enhanced with the overexpression of ALDH2 and reduced with the knockdown of ALDH2 expression. DEA showed dose-dependent effects on inhibiting liver lipid deposition, elevating ALDH2 activity and reducing 4-HNE levels in the livers of MCD diet-induced NAFLD mice. CONCLUSION: DEA is the effective compound in AC against NAFLD. The related anti-NAFLD mechanisms of AC and DEA were through upregulating ALDH2 expression and activity, thus enhancing the elimination of 4-HNE in the livers, and sequentially alleviating oxidative stress and inflammation, promoting fatty acid ß-oxidation and decreasing lipogenesis.


Subject(s)
Non-alcoholic Fatty Liver Disease , Aldehyde Dehydrogenase, Mitochondrial/genetics , Animals , Diet, High-Fat , Fatty Acids/metabolism , Inflammation/drug therapy , Lanosterol/analogs & derivatives , Lanosterol/pharmacology , Lipid Metabolism , Liver , Mice , Mice, Inbred C57BL , Non-alcoholic Fatty Liver Disease/drug therapy , Non-alcoholic Fatty Liver Disease/metabolism , Polyporales
4.
Biomed Pharmacother ; 141: 111909, 2021 Sep.
Article in English | MEDLINE | ID: mdl-34328088

ABSTRACT

Ganoderma lucidum is a legendary traditional Chinese medicine with various bioactivities. This study was conducted (a) to explore the in vitro fermentation of the water extracts of G. lucidum fruiting body with Lactobacillus acidophilus and Bifidobacterium breve and (b) to investigate the effect of fermentation broth (GLFB) on dexamethasone (DEX)-induced immunosuppressed mice. Our results demonstrated that probiotic fermentation of G. lucidum fruiting body extracts underwent structural changing of major ganoderic acid components, such as ganoderic acid A (GA) into GC2, and this fermentation process involves changing of several metabolic pathways in the probiotic strains. GLFB could significantly improve the immunity, intestinal integrity, and gut microbiota dysbiosis in DEX-treated mice, and the immunostimulatory activity of GLFB was found closely related to its direct regulation on the expansion of CD4+ T cells in Peyer's patches of mice. These data implied that probiotic fermentation of G. lucidum fruiting body extracts promoted its immunostimulatory activity via biotransformation of components such as GA. This research provides a theoretical support for the development and application of G. lucidum fermentation by probiotics.


Subject(s)
Adjuvants, Immunologic/pharmacology , Dexamethasone/pharmacology , Fruiting Bodies, Fungal/chemistry , Immunosuppressive Agents/pharmacology , Probiotics/metabolism , Reishi/metabolism , Animals , CD4-Positive T-Lymphocytes/metabolism , Fermentation , Gastrointestinal Microbiome/drug effects , Heptanoic Acids/pharmacology , Intestines/drug effects , Lanosterol/analogs & derivatives , Lanosterol/pharmacology , Lymphocyte Count , Male , Medicine, Chinese Traditional , Mice , Mice, Inbred BALB C , Peyer's Patches/cytology , Peyer's Patches/drug effects , Reishi/chemistry
5.
Chem Biol Interact ; 344: 109529, 2021 Aug 01.
Article in English | MEDLINE | ID: mdl-34029542

ABSTRACT

Ganoderic acid A (GAA), one of the major triterpenoid components extracted from Ganoderma mushroom has been shown to possess numerous important pharmacological activities. The present study was aimed to investigate the mechanisms of GAA on carbon tetrachloride (CCl4)-induced kidney inflammation, fibrosis and oxidative stress in mice. The male mice were treated with 25 and 50 mg/mg GAA after stimulated with CCl4. Our results showed that GAA improved renal damage by decreasing the serum levels of creatinine, urea, uric acid and alleviating kidney fibrosis. GAA ameliorated CCl4-induced indices of inflammation. GAA suppressed oxidative stress by regulating the glutathione antioxidant system and the thioredoxin antioxidant system. GAA increased the activations of thioredoxin reductase (TrxR), Trx, GSH, SOD, GPx. Furthermore, GAA supplementation inhibited the JAK and STAT3 pathway. GAA inhibited the activations of RhoA, ROCK, NF-κB, TGF-ß and Smad3. Thus, this study demonstrated that GAA possesses immune-protective properties through regulating the Trx/TrxR, JAK2/STAT3 and RhoA/ROCK pathways.


Subject(s)
Anti-Inflammatory Agents/therapeutic use , Antioxidants/therapeutic use , Fibrosis/drug therapy , Heptanoic Acids/therapeutic use , Kidney Diseases/drug therapy , Lanosterol/analogs & derivatives , Signal Transduction/drug effects , Animals , Carbon Tetrachloride , Fibrosis/chemically induced , Fibrosis/pathology , Janus Kinase 2/metabolism , Kidney Diseases/chemically induced , Kidney Diseases/pathology , Lanosterol/therapeutic use , Male , Mice, Inbred ICR , Oxidative Stress/drug effects , Smad3 Protein/metabolism , Thioredoxin-Disulfide Reductase/metabolism , Thioredoxins/metabolism , Transforming Growth Factor beta1/metabolism , rho-Associated Kinases/metabolism
6.
J Ethnopharmacol ; 274: 114081, 2021 Jun 28.
Article in English | MEDLINE | ID: mdl-33798660

ABSTRACT

ETHNOPHARMACOLOGICAL RELEVANCE: Chaga mushrooms (Inonotus obliquus) are commonly used in traditional treatments in Eastern Europe and Asia due to their diverse pharmacological effects, including anti-tumor and immunologic effects. Thus, many cancer patients take Chaga mushrooms as a complementary medicine, even during chemotherapy or radiotherapy. However, few studies have investigated the effects or molecular targets of Chaga mushrooms in breast cancer. AIM OF THE STUDY: Herein, we examined the anticancer effects of Chaga mushrooms in different types of breast cancer cell lines, and explored the underlying molecular mechanism to better understand their effects and benefits. MATERIALS AND METHODS: Chaga mushroom extract (CME) was prepared by extracting Chaga mushrooms with 70% ethanol. The cytotoxic effects of CME were assessed by MTT assay and protein expressions were evaluated by western blotting. To evaluate in vivo anti-tumor effects of CME, CME (2 g/kg) was orally administered to 4T1 tumor-bearing BALB/c mice every other day over 30 days (15 administrations), and tumor sizes were measured. Silica gel column chromatography was used to fractionate CME, and major constituents responsible for cytotoxic effects of CME were identified by 1H/13C-NMR and LC-MS. RESULTS: CME inhibited the proliferation of 4T1 mouse breast cancer cells in a dose and time-dependent manner. The expression of LC3 and phosphorylation of AMPK were increased by CME, while the phosphorylation of mTOR, S6, and S6K1 were suppressed, suggesting that CME induced autophagy by activating AMPK and inhibiting mTOR signaling pathways. Consistent with its observed cytotoxic effect in vitro, CME effectively suppressed tumor growth in 4T1 tumor-bearing BALB/c mice. In addition, inotodiol and trametenolic acid were identified as the major constituents responsible for the cytotoxic effects of CME on breast cancer cells. Moreover, inotodiol and trametenolic acid-enriched fractions both exhibited cytotoxic effects regardless of breast cancer cell subtypes and did not interfere with the cytotoxic effects of conventional drugs. CONCLUSIONS: Taken together, Chaga mushroom extract induced autophagy by activating AMPK and inhibiting the mTOR signaling pathway. Our data suggest Chaga mushrooms may be a beneficial complementary medicine for breast cancer patients.


Subject(s)
Agaricales , Antineoplastic Agents/therapeutic use , Breast Neoplasms/drug therapy , Complex Mixtures/therapeutic use , AMP-Activated Protein Kinases/metabolism , Animals , Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacology , Autophagy/drug effects , Breast Neoplasms/metabolism , Cell Line, Tumor , Cell Survival/drug effects , Complex Mixtures/chemistry , Complex Mixtures/pharmacology , Female , Humans , Lanosterol/analogs & derivatives , Lanosterol/analysis , Lanosterol/pharmacology , Mice, Inbred BALB C , Signal Transduction/drug effects , TOR Serine-Threonine Kinases/metabolism , Triterpenes/analysis , Triterpenes/pharmacology
7.
Phytomedicine ; 85: 153546, 2021 May.
Article in English | MEDLINE | ID: mdl-33799221

ABSTRACT

BACKGROUND: Hyperglycemia-induced cardiovascular dysfunction has been linked to oxidative stress and accelerated apoptosis in the diabetic myocardium. While there is currently no treatment for diabetic cardiomyopathy (DCM), studies suggest that the combinational use of anti-hyperglycemic agents and triterpenes could be effective in alleviating DCM. HYPOTHESIS: To investigate the therapeutic effect of methyl-3ß-hydroxylanosta-9,24-dien-21-oate (RA3), in the absence or presence of the anti-diabetic drug, metformin (MET), against hyperglycemia-induced cardiac injury using an in vitro H9c2 cell model. METHODS: To mimic a hyperglycemic state, H9c2 cells were exposed to high glucose (HG, 33 mM) for 24 h. Thereafter, the cells were treated with RA3 (1 µM), MET (1 µM) and the combination of MET (1 µM) plus RA3 (1 µM) for 24 h, to assess the treatments therapeutic effect. RESULTS: Biochemical analysis revealed that RA3, with or without MET, improves glucose uptake via insulin-dependent (IRS-1/PI3K/Akt signaling) and independent (AMPK) pathways whilst ameliorating the activity of antioxidant enzymes in the H9c2 cells. Mechanistically, RA3 was able to alleviate HG-stimulated oxidative stress through the inhibition of reactive oxygen species (ROS) and lipid peroxidation as well as the reduced expression of the PKC/NF-кB cascade through decreased intracellular lipid content. Subsequently, RA3 was able to mitigate HG-induced apoptosis by decreasing the activity of caspase 3/7 and DNA fragmentation in the cardiomyoblasts. CONCLUSION: RA3, in the absence or presence of MET, demonstrated potent therapeutic properties against hyperglycemia-mediated cardiac damage and could be a suitable candidate in the prevention of DCM.


Subject(s)
Apoptosis/drug effects , Energy Metabolism , Hyperglycemia/pathology , Lanosterol/analogs & derivatives , Myocytes, Cardiac/drug effects , Oxidative Stress/drug effects , Animals , Antioxidants/pharmacology , Caspases , Cell Line , Diabetic Cardiomyopathies , Glucose/metabolism , Insulin/metabolism , Lanosterol/pharmacology , Metformin/pharmacology , Phosphatidylinositol 3-Kinases/metabolism , Rats , Reactive Oxygen Species/metabolism , Signal Transduction/drug effects
8.
Eur J Pharmacol ; 890: 173688, 2021 Jan 05.
Article in English | MEDLINE | ID: mdl-33130280

ABSTRACT

The coronavirus disease-19 (COVID-19) outbreak that is caused by a highly contagious severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2) has become a zoonotic pandemic, with approximately 24.5 million positive cases and 8.3 lakhs deaths globally. The lack of effective drugs or vaccine provoked the research for drug candidates that can disrupt the spread and progression of the virus. The identification of drug molecules through experimental studies is time-consuming and expensive, so there is a need for developing alternative strategies like in silico approaches which can yield better outcomes in less time. Herein, we selected transmembrane protease serine 2 (TMPRSS2) enzyme, a potential pharmacological target against SARS-CoV-2, involved in the spread and pathogenesis of the virus. Since 3D structure is not available for this protein, the present study aims at homology modelling and validation of TMPRSS2 using Swiss-model server. Validation of the modelled TMPRSS2 using various online tools confirmed model accuracy, topology and stereochemical plausibility. The catalytic triad consisting of Serine-441, Histidine-296 and Aspartic acid-345 was identified as active binding site of TMPRSS2 using existing ligands. Molecular docking studies of various drugs and phytochemicals against the modelled TMPRSS2 were performed using camostat as a standard drug. The results revealed eight potential drug candidates, namely nafamostat, meloxicam, ganodermanontriol, columbin, myricetin, proanthocyanidin A2, jatrorrhizine and baicalein, which were further studied for ADME/T properties. In conclusion, the study unravelled eight high affinity binding compounds, which may serve as potent antagonists against TMPRSS2 to impact COVID-19 drug therapy.


Subject(s)
Antiviral Agents/pharmacology , Models, Molecular , Serine Endopeptidases/metabolism , Serine Proteinase Inhibitors/pharmacology , Benzamidines , Berberine/analogs & derivatives , Berberine/pharmacology , Binding Sites , Diterpenes/pharmacology , Flavanones/pharmacology , Flavonoids/pharmacology , Guanidines/pharmacology , Lactones/pharmacology , Lanosterol/analogs & derivatives , Lanosterol/pharmacology , Meloxicam/pharmacology , Proanthocyanidins/pharmacology , Protein Binding , SARS-CoV-2 , COVID-19 Drug Treatment
9.
Chin J Nat Med ; 18(12): 934-940, 2020 Dec.
Article in English | MEDLINE | ID: mdl-33357724

ABSTRACT

A contributory role of oxidative stress and protection by antioxidant nutrients have been suspected in cataract formation. Ganoderic acid A (GAA), an effective lanostane triterpene, is widely reported as an antioxidant. The aim of this study is to investigate the potential effects of GAA on cataract formation. After lens epithelial cells (LECs) were exposed to UVB radiation for different periods, cell viability, apoptosis-related protein levels, malondialdehyde (MDA) and superoxide dismutase (SOD) activities were monitored. We found that cell viability, the Bcl-2/Bax ratio and SOD activity were increased, while Cleaved caspase-3 levels and MDA activity were decreased compared with those in UVB-impaired LECs after GAA treated. Furthermore, GAA activated PI3K/AKT in UVB-impaired LECs and effectively delayed the occurrence of lens opacity in vitro. In conclusion, these findings demonstrated that GAA exhibited protective functions in SRA01/04 cells and rat lenses against UVB-evoked impairment through elevating cell viability and antioxidant activity, inhibiting cell apoptosis, activating the PI3K/AKT pathway and delaying lens opacity.


Subject(s)
Cataract/prevention & control , Epithelial Cells/drug effects , Heptanoic Acids/pharmacology , Lanosterol/analogs & derivatives , Lens, Crystalline/cytology , Ultraviolet Rays/adverse effects , Animals , Apoptosis , Cell Line , Cell Survival , Epithelial Cells/radiation effects , Humans , Lanosterol/pharmacology , Lens, Crystalline/radiation effects , Malondialdehyde/metabolism , Rats , Superoxide Dismutase/metabolism
10.
Food Funct ; 11(8): 6818-6833, 2020 Aug 01.
Article in English | MEDLINE | ID: mdl-32686808

ABSTRACT

Ganoderic acid A (GA) is one of the most abundant triterpenoids in Ganoderma lucidum, and has been proved to possess a wide range of beneficial health effects. The aim of the current study is to investigate the amelioration effects and mechanism of GA on improving hyperlipidemia in mice fed a high-fat diet (HFD). The results showed that GA intervention significantly inhibited the abnormal growth of body weight and epididymal white adipose tissue (eWAT), prevented the hypertrophy of epididymal adipocytes, and ameliorated the biochemical parameters of serum and liver related to lipid metabolism in HFD-fed mice. Histological analysis also showed that the excessive accumulation of lipid droplets in the liver induced by HFD-feeding was greatly alleviated by GA intervention. In addition, GA intervention also increased the level of short chain fatty acids (SCFAs) in the intestine and promoted the excretion of bile acids (BAs) through feces. High-throughput sequencing of bacterial full-length 16S rDNA revealed that daily supplementation with GA made significant structural changes in the gut microbial population of mice fed with HFD, in particular modulating the relative abundance of some function related microbial phylotypes. The relationships between lipid metabolic parameters and gut microbial phylotypes were also revealed by correlation analysis based on a heatmap and network. The result showed that 46 key gut microbial phylotypes (OTUs) were markedly correlated with at least one lipid metabolic parameter. Moreover, UPLC-QTOF/MS-based liver metabolomics showed that 111 biomarkers (47 up-regulated metabolites and 64 down-regulated metabolites) were significantly changed after high-dose GA intervention (75 mg kg-1 day-1), compared with the HFD-fed hyperlipidemic mice. Metabolic pathway enrichment analysis of the differential hepatic metabolites demonstrated that GA intervention had significant regulatory effects on primary bile acid biosynthesis, fatty acid biosynthesis, amino sugar and nucleotide sugar metabolism, inositol phosphate metabolism, and so on. In addition, GA intervention regulated the mRNA levels of hepatic genes involved in fatty acid metabolism and bile acid homeostasis. These findings present new evidence supporting that GA from G. lucidum has the potential to alleviate lipid metabolic disorders and ameliorate the imbalance of gut microflora in a positive way.


Subject(s)
Gastrointestinal Microbiome/drug effects , Heptanoic Acids/pharmacology , Hyperlipidemias/therapy , Lanosterol/analogs & derivatives , Lipid Metabolism/drug effects , Reishi/chemistry , Animals , Bile Acids and Salts/metabolism , Diet, High-Fat/adverse effects , Disease Models, Animal , Fatty Acids, Volatile/metabolism , Feces/chemistry , Hyperlipidemias/etiology , Hyperlipidemias/metabolism , Lanosterol/pharmacology , Liver/metabolism , Male , Metabolomics , Mice
11.
Bioorg Med Chem Lett ; 30(14): 127256, 2020 07 15.
Article in English | MEDLINE | ID: mdl-32527555

ABSTRACT

The leaves of the kaffir lime (Citrus hystrix) are commonly used in cuisine and folk medicine. The aim of this study was to isolate a bioactive compound in kaffir lime leaves and characterize its biological activity. The compound was isolated from a hexane fractional extract and identified as agrostophillinol. This is the first report of agrostophillinol isolated from kaffir lime leaves. In terms of cytotoxicity, agrostophillinol exhibited IC50 values of 36.27 ± 7.30 and 53.44 ± 10.63 µg/mL against EoL-1 and HL60 cells, respectively. Agrostophillinol also exhibited potent anti-inflammatory activity, significantly inhibiting IL-6 secretion.


Subject(s)
Anti-Inflammatory Agents/pharmacology , Antineoplastic Agents, Phytogenic/pharmacology , Antioxidants/pharmacology , Citrus/chemistry , Interleukin-6/antagonists & inhibitors , Lanosterol/analogs & derivatives , Plant Extracts/pharmacology , Animals , Anti-Inflammatory Agents/chemistry , Anti-Inflammatory Agents/isolation & purification , Antineoplastic Agents, Phytogenic/chemistry , Antineoplastic Agents, Phytogenic/isolation & purification , Antioxidants/chemistry , Antioxidants/isolation & purification , Biphenyl Compounds/antagonists & inhibitors , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Survival/drug effects , Dose-Response Relationship, Drug , Drug Screening Assays, Antitumor , Humans , Interleukin-6/metabolism , Lanosterol/chemistry , Lanosterol/isolation & purification , Lanosterol/pharmacology , Mice , Molecular Conformation , Picrates/antagonists & inhibitors , Plant Extracts/chemistry , Plant Extracts/isolation & purification , Plant Leaves/chemistry , RAW 264.7 Cells , Structure-Activity Relationship
12.
Int Immunopharmacol ; 81: 106244, 2020 Apr.
Article in English | MEDLINE | ID: mdl-32035309

ABSTRACT

Inotodiol is a lanostane triterpenoid found only in Chaga mushroom. In the previous study investigating anti-allergic effects of fractionated Chaga mushroom extracts, we have found evidence that purified inotodiol holds an activity to suppress the mast cell function in vivo. To address the therapeutic relevance of the finding, in this study, we investigated whether inotodiol could also alleviate allergy symptoms observed in a chicken ovalbumin (cOVA)-induced mouse model of food allergy. Like the crude 70% ethanol extract of Chaga mushroom (320 mg/kg), oral administration of inotodiol (20 mg/kg), regardless of whether that was for preventive or treatment purpose, resulted in a significant improvement in allergic symptoms and inflammatory lesions in the small intestine appearing after repeated oral challenge with cOVA. Despite the results that inotodiol (20 mg/kg) and the Chaga mushroom extract (320 mg/kg) took effect to a similar extent, immunological mechanisms underlying those effects were found to be distinct from each other. That is, the results obtained from several in vivo assays, including mast cell-mediated passive systemic anaphylaxis, activation/proliferation of adoptively transferred antigen-specific T cells and immunoglobulin (IgG1, IgE, IgA) production by antigen-specific B cells, illustrated that inotodiol selectively inhibited the mast cell function without having any noticeable effect on other immune responses while the crude Chaga mushroom extract indiscriminately suppressed diverse immune responses. The strong anti-allergic activity of inotodiol, along with its remarkable selectivity to mast cell, makes it an excellent therapeutic candidate for food allergy with both high efficacy and outstanding safety.


Subject(s)
Anti-Allergic Agents/therapeutic use , Food Hypersensitivity/drug therapy , Lanosterol/analogs & derivatives , Mast Cells/immunology , Allergens/immunology , Animals , Cell Degranulation/drug effects , Disease Models, Animal , Humans , Inonotus/immunology , Lanosterol/chemistry , Lanosterol/therapeutic use , Mast Cells/drug effects , Mice , Mice, Inbred BALB C , Ovalbumin/immunology , Triterpenes/chemistry
13.
Pharmacology ; 105(9-10): 568-575, 2020.
Article in English | MEDLINE | ID: mdl-31940650

ABSTRACT

BACKGROUND: To study the protective effects of ganoderic acid A (GAA) on bleomycin (BLM)-induced pulmonary fibrosis. METHODS: ICR mice were intratracheally instilled with BLM to induce pulmonary fibrosis on day 0. Then the mice were orally given GAA (25, 50 mg/kg) or dexamethasone (2 mg/kg). After treatment for 21 days, the mice were sacrificed. Wet dry weight (W/D) ratio of lung was used to detect pulmonary edema. Myeloperoxidase (MPO), interleukin-1ß (IL-1ß), IL-6, tumor necrosis factor-α (TNF-α), malondialdehyde (MDA), and superoxide dismutase (SOD) were detected by enzyme-linked immunosorbent assay. Hematoxylin and eosin staining was used to evaluate the pathological changes. The levels of transforming growth factor ß (TGF-ß), phosphorylated-smad3 (p-smad3), p-IκB, and p-nuclear factor-kappa B (NF-κB) in lung tissue were detected by western blot. RESULTS: GAA treatment significantly improved MPO activity, W/D ratio, and lung histopathology. The protective effect of GAA may be related to downregulation of TNF-α, IL-1ß, IL-6, MDA and upregulation of SOD. In addition, GAA significantly decreased the levels of TGF-ß, p-smad3, p-IκB, and p-NF-κB, compared with those in BLM group. CONCLUSION: GAA has protective effect on BLM-induced lung injury, and TGF-ß/Smad-3/NF-κB signaling pathway may play an important role in the pathogenesis of BLM-induced lung injury.


Subject(s)
Heptanoic Acids/pharmacology , Lanosterol/analogs & derivatives , Lung/drug effects , Plant Extracts/pharmacology , Protective Agents/pharmacology , Pulmonary Fibrosis/drug therapy , Animals , Bleomycin/toxicity , Cytokines/blood , Heptanoic Acids/therapeutic use , Lanosterol/pharmacology , Lanosterol/therapeutic use , Lung/metabolism , Lung/pathology , Male , Malondialdehyde/blood , Mice , Mice, Inbred ICR , NF-kappa B/metabolism , Peroxidase/metabolism , Phytotherapy , Plant Extracts/administration & dosage , Plant Extracts/therapeutic use , Protective Agents/therapeutic use , Pulmonary Edema/drug therapy , Pulmonary Fibrosis/chemically induced , Smad3 Protein/metabolism , Superoxide Dismutase/blood , Transforming Growth Factor beta/metabolism
14.
Cytokine ; 127: 154917, 2020 03.
Article in English | MEDLINE | ID: mdl-31775117

ABSTRACT

Ganoderma lucidum is a popular medicinal mushroom, which has been used as therapeutic for centuries in traditional Chinese medicine. Although G. lucidum showed strong protective effects in prevention or treatment of a variety of inflammatory diseases, the mechanisms underlying the anti-inflammatory properties of triterpenes of G. lucidum remain undefined. In the current study, we demonstrated that ethanol extract and triterpenes of G. lucidum specifically suppressed LPS-mediated inflammatory responses. Notably, ganodermanontriol inhibited the expressions and interactions of TLR4 and MyD88, NF-κB translocation to nucleus and its DNA binding activity, phosphorylation of p38, ErK1/2 and JNK. In vivo, we showed that ganodermanontriol effectively prevented LPS/D-Galactosamine-induced liver injury by reducing TNF-α and IL-6 production, and decrease of ALT/AST release. Collectively, our results revealed a novel role in inhibition of inflammatory diseases for triterpenes that may act through potential inhibition of TLR4-MyD88-mediated NF-κB and MAPK signaling pathways.


Subject(s)
Chemical and Drug Induced Liver Injury/prevention & control , Inflammation/prevention & control , Lanosterol/analogs & derivatives , Reishi/chemistry , Triterpenes/pharmacology , Animals , Cell Line , Cell Survival/drug effects , Chemical and Drug Induced Liver Injury/etiology , Chemical and Drug Induced Liver Injury/metabolism , Cytokines/metabolism , Female , Inflammation/chemically induced , Lanosterol/chemistry , Lanosterol/pharmacology , Lipopolysaccharides , MAP Kinase Signaling System/drug effects , Macrophages/cytology , Macrophages/drug effects , Macrophages/metabolism , Mice, Inbred BALB C , Molecular Structure , NF-kappa B/metabolism , Protective Agents/chemistry , Protective Agents/pharmacology , Triterpenes/chemistry
15.
Phytother Res ; 34(3): 640-648, 2020 Mar.
Article in English | MEDLINE | ID: mdl-31742778

ABSTRACT

Ganoderic Acid A (GAA) is often applied for healing cardiovascular and cerebrovascular ailments, but the influences in cerebral ischemia injury are still hazy. The research delved into the functions of GAA in hypoxia-triggered impairment in PC12 cells. PC12 cells received hypoxia management for 12 hr, and subsequently, cell viability, migration, apoptosis, and correlative protein levels were assessed. After preprocessing with GAA, above cell behaviors were monitored again. The vector of microRNA (miR)-153 inhibitor was utilized for PC12 cell transfection to further explore the functions of miR-153 in hypoxia-impaired cells. Pathways of phosphatidylinositol 3-kinase (PI3K)/protein kinase B (AKT) and mammalian target of rapamycin (mTOR) were investigated via executing western blot for uncovering the latent mechanism. Results revealed that hypoxia disposition triggered PC12 cells impairment via restraining cell viability and migration and accelerating apoptosis. However, GAA visibly mollified hypoxia-provoked impairment in PC12 cells. Interestingly, the enhancement of miR-153 triggered by GAA was observed in hypoxia-impaired PC12 cells. After miR-153 inhibitor transfection, the protective functions of GAA in hypoxia-impaired PC12 cells were dramatically inversed. Furthermore, GAA caused PI3K/AKT and mTOR activations via enhancement of miR-153 in hypoxia-impaired PC12 cells. The findings evinced that GAA exhibited the protective functions in PC12 cells against hypoxia-evoked impairment through activating PI3K/AKT and mTOR via elevating miR-153.


Subject(s)
Cytoprotection/drug effects , Heptanoic Acids/pharmacology , Lanosterol/analogs & derivatives , MicroRNAs/genetics , Animals , Apoptosis/drug effects , Cell Hypoxia , Cell Survival/drug effects , Lanosterol/pharmacology , Oncogene Protein v-akt/metabolism , PC12 Cells , Phosphatidylinositol 3-Kinases/metabolism , Rats , TOR Serine-Threonine Kinases/metabolism
16.
Phytomedicine ; 60: 152957, 2019 Jul.
Article in English | MEDLINE | ID: mdl-31128995

ABSTRACT

BACKGROUND: Inonotus obliquus, namely as Chaga mushroom, is a medicinal and edible fungus, which is widely used in food and medical fields. Inotodiol, a natural lanostane-type triterpenoid with remarkable pharmacological activities, was isolated from Inonotus obliquus, which its potential anti-tumor molecular mechanism was elaborated poorly. PURPOSE: The aim of the present study was to investigate the effect of Inotodiol on HeLa cell migration, invasion and apoptosis through p53-dependent pathway. STUDY DESIGN AND METHODS: The potential mechanisms of Inotodiol on HeLa cell anti-metastatic and pro-apoptosis via wound healing assay, trans-well invasion assay, flow cytometry, caspase-3 activity assay and western blot analysis were studied, as well as the involvement of p53 signaling pathway in anti-metastatic and pro-apoptosis of Inotodiol. Besides, the function of tumor suppressor p53 was further verified by small interfering RNA. RESULTS: Firstly, the cell viability assay showed that low-concentration of Inotodiol had no cytotoxicity to HeLa cells and whereas the concentration above 25 µM significantly inhibited HeLa cell growth and even induced apoptosis. This result was further demonstrated by cell proliferation and morphology assay. Secondly, in vitro wound healing and trans-well invasion assays reported that low-concentration treatment of Inotodiol significantly inhibited cells migration and invasion in a dose-dependent manner, the western blot analysis of matrix mettalloprotinase-2 (MMP2) and matrix mettalloprotinase-9 (MMP9) levels were also decreased. Moreover, Inotodiol notably induced tumor cell apoptosis by Annexin-V-FITC apoptosis assay, which is associated with activation pro-apoptotic proteins of PARP, cleaved caspase-3 and Bax expression, inhibition anti-apoptotic protein Bcl-2 expression. Finally, the anti-tumor activity of Inotodiol was attenuated by silencing p53 tumor suppressor, the result revealed that pre-treatment with p53-specific small interfering RNA (si-p53) markedly inhibited Intodiol-indeuced HeLa cell apoptosis and decreased the caspase-3 activity. What is more, the inhibitory effect of Inotodiol on tumor migration and invasion was blocked under p53 knockdown. CONCLUSION: To sum up, the present study indicated that Inotodiol possessed the potential to prevent malignant tumor migration and invasion, and it might be a natural active compound candidate for clinical treatment of human cervical cancer.


Subject(s)
Agaricales/chemistry , Antineoplastic Agents/pharmacology , Apoptosis/drug effects , Lanosterol/analogs & derivatives , Tumor Suppressor Protein p53/metabolism , Uterine Cervical Neoplasms/drug therapy , Caspase 3/genetics , Caspase 3/metabolism , Cell Movement/drug effects , Cell Proliferation/drug effects , Cell Survival/drug effects , Female , HeLa Cells , Humans , Lanosterol/pharmacology , Neoplasm Invasiveness , Proto-Oncogene Proteins c-bcl-2/genetics , Proto-Oncogene Proteins c-bcl-2/metabolism , Signal Transduction/drug effects , Tumor Suppressor Protein p53/genetics
17.
Phytother Res ; 33(5): 1448-1456, 2019 May.
Article in English | MEDLINE | ID: mdl-30838711

ABSTRACT

Effects of ganoderic acid A (GAA), a lanostane triterpene, on hypoxia-ischemia encephalopathy (HIE) remain unclear. We aimed to figure out the specific role of GAA in hypoxia-treated neural stem cells (NSCs) as well as the regulatory mechanisms. Primary rat NSCs were incubated under hypoxia to simulate HIE. Viability and apoptosis of hypoxia-injured NSCs were measured by cell counting kit-8 and flow cytometry assays, respectively. Proteins related to apoptosis, autophagy, and the PI3K/AKT/mTOR pathways were evaluated by Western blot analysis. LY294002 and rapamycin were added to inhibit the PI3K/AKT pathway and mTOR pathway, respectively. Enzyme-linked immunosorbent assay was carried out to test the release of proinflammatory cytokines. We found that hypoxia-induced decrease of cell viability, increases of apoptotic cells and autophagy, and the release of IL-6, IL-1ß, and TNF-α were all attenuated by GAA stimulation. Activation of caspases induced by hypoxia was alleviated by GAA. Furthermore, we found that inhibition of the PI3K/AKT pathway eliminated the effects of GAA on apoptosis and proinflammatory cytokines release in hypoxia-injured NSCs. Meanwhile, inhibition of the mTOR pathway abrogated the effects of GAA on cell autophagy in hypoxia-injured NSCs. In conclusion, GAA alleviated hypoxia-induced injury in NSCs might be through activating the PI3K/AKT and mTOR pathways.


Subject(s)
Apoptosis/drug effects , Autophagy/drug effects , Heptanoic Acids/pharmacology , Lanosterol/analogs & derivatives , Neural Stem Cells/drug effects , Animals , Cell Hypoxia/physiology , Cell Survival/drug effects , Inflammation/metabolism , Lanosterol/pharmacology , Phosphatidylinositol 3-Kinases/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Rats , Rats, Sprague-Dawley , Signal Transduction/drug effects , TOR Serine-Threonine Kinases/metabolism
18.
J Nat Med ; 73(3): 597-601, 2019 Jun.
Article in English | MEDLINE | ID: mdl-30706371

ABSTRACT

Chaga mushrooms, the sclerotium of Inonotus obliquus, have been used in Mongolia as a traditional hair shampoo to maintain healthy hair. Bioassay-guided fractionations of the extract of Chaga mushrooms using a proliferation assay on human follicle dermal papilla cells (HFDPCs) gave five lanostane-type triterpenes (1-5), whose structures were identified by spectroscopic evidence. Among these, lanosterol (1), inotodiol (3), lanost-8,24-diene-3ß,21-diol (4), and trametenolic acid (5) demonstrated proproliferative effects on HFDPCs more potent than minoxidil, an anti-alopecia agent, used as the positive control. The lanostane-type triterpenes (1, 3, 4, and 5) appeared to be potential candidates of new agents possibly used for hair-care with a stimulative effect on hair growth.


Subject(s)
Agaricales/chemistry , Androgen Receptor Antagonists/pharmacology , Cell Extracts/pharmacology , Steroids/analysis , Triterpenes/pharmacology , Cell Extracts/chemistry , Cell Proliferation/drug effects , Cells, Cultured , Hair/growth & development , Hair Follicle/cytology , Humans , Lanosterol/analogs & derivatives , Lanosterol/analysis , Mongolia , Receptors, Androgen/drug effects , Triterpenes/analysis , Triterpenes/chemistry
19.
Nutrients ; 12(1)2019 Dec 27.
Article in English | MEDLINE | ID: mdl-31892211

ABSTRACT

Microglia mediated neuronal inflammation has been widely reported to be responsible for neurodegenerative disease. Deacetyl ganoderic acid F (DeGA F) is a triterpenoid isolated from Ganoderma lucidum, which is a famous edible and medicinal mushroom used for treatment of dizziness and insomnia in traditional medicine for a long time. In this study the inhibitory effects and mechanisms of DeGA F against lipopolysaccharide (LPS)-induced inflammation both in vitro and in vivo were investigated. On murine microglial cell line BV-2 cells, DeGA F treatment inhibited LPS-triggered NO production and iNOS expression and affected the secretion and mRNA levels of relative inflammatory cytokines. DeGA F inhibited LPS-induced activation of the NF-κB pathway, as evidenced by decreased phosphorylation of IKK and IκB and the nuclear translocation of P65. In vivo, DeGA F treatment effectively inhibited NO production in zebrafish embryos. Moreover, DeGA F suppressed the serum levels of pro-inflammatory cytokines, including TNF-α and IL-6 in LPS-stimulated mice model. DeGA F reduced inflammatory response by suppressing microglia and astrocytes activation and also suppressed LPS-induced NF-κB activation in mice brains. Taken together, DeGA F exhibited remarkable anti-inflammatory effects and promising therapeutic potential for neural inflammation associated diseases.


Subject(s)
Heptanoic Acids/pharmacology , Inflammation/prevention & control , Lanosterol/analogs & derivatives , Lipopolysaccharides/pharmacology , Microglia/physiology , NF-kappa B/metabolism , Animals , Brain/drug effects , Brain/metabolism , Cell Line , Cytokines/antagonists & inhibitors , Cytokines/metabolism , Gene Expression/drug effects , Inflammation/pathology , Lanosterol/pharmacology , Male , Mice , Mice, Inbred C57BL , Microglia/drug effects , Microglia/pathology , NF-kappa B/drug effects , Neuritis , Neurodegenerative Diseases , Nitric Oxide/biosynthesis , Nitric Oxide Synthase Type II/genetics , Signal Transduction/drug effects , Zebrafish
20.
Spectrochim Acta A Mol Biomol Spectrosc ; 210: 372-380, 2019 Mar 05.
Article in English | MEDLINE | ID: mdl-30502725

ABSTRACT

Ganoderic Acids (GAs) are the major medicinal compounds in Ganoderma lucidum used as traditional Chinese medicine since ancient times. Ganoderic acid A (GAA) is the first discovered ganoderic acids reported in the literature, which is also one of most abundant triterpenoids of Ganoderma lucidum. Especially, GAA has been extensively investigated in recent decades for its positive medicinal activities. However, the vibrational properties of GAs have rarely been studied or reported. In this work, we focused on the typical GAA and studied the infrared (IR) and Raman spectra based on both experiments and DFT calculations. As such, we could not only achieve the assignments of the vibrational modes, but also from the IR and Raman spectra, we found that the spectral region from 1500 cm-1 to 1800 cm-1 is particularly useful for distinguishing different types of GAs. In addition, its dehydrogenated derivative ganoderenic acid A (GOA) was also studied, which could be identified due to its spectral feature of strong IR and Raman bands around 1620 cm-1. This work therefore may facilitate the application of IR and Raman spectroscopies in the inspection and quality control of Ganoderma lucidum.


Subject(s)
Heptanoic Acids/chemistry , Lanosterol/analogs & derivatives , Spectrophotometry, Infrared/methods , Spectrum Analysis, Raman/methods , Cholestanes/chemistry , Density Functional Theory , Lanosterol/chemistry , Molecular Structure , Reishi/chemistry , Spectroscopy, Fourier Transform Infrared , Vibration
SELECTION OF CITATIONS
SEARCH DETAIL