Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 16 de 16
Filter
Add more filters










Publication year range
1.
Mol Biotechnol ; 66(3): 442-453, 2024 Mar.
Article in English | MEDLINE | ID: mdl-37199885

ABSTRACT

Myocardial infarction (MI) is an extremely severe cardiovascular disease, which ranks as the leading cause of sudden death worldwide. Studies have proved that cardiac injury following MI can cause cardiomyocyte apoptosis and myocardial fibrosis. Bilobalide (Bilo) from Ginkgo biloba leaves have been widely reported to possess excellent cardioprotective effects. However, concrete roles of Bilo in MI have not been investigated yet. We here designed both in vitro and in vivo experiments to explore the effects of Bilo on MI-induced cardiac injury and the underlying mechanisms of its action. We conducted in vitro experiments using oxygen-glucose deprivation (OGD)-treated H9c2 cells. Cell apoptosis in H9c2 cells was assessed by conducting flow cytometry assay and evaluating apoptosis-related proteins with western blotting. MI mouse model was established by performing left anterior descending artery (LAD) ligation. Cardiac function of MI mice was determined by assessing ejection fraction (EF), fractional shortening (FS), left ventricular end-systolic diameter (LVESD), and left ventricular end-diastolic diameter (LVEDD). Histological changes were analyzed, infarct size and myocardial fibrosis were measured by hematoxylin and eosin (H&E) and Masson staining in cardiac tissues from the mice. The apoptosis of cardiomyocytes in MI mice was assessed by TUNEL staining. Western blotting was applied to detect the effect of Bilo on c-Jun N-terminal kinase (JNK)/p38 mitogen-activated protein kinases (p38 MAPK) signaling both in vitro and in vivo. Bilo inhibited OGD-induced cell apoptosis and lactate dehydrogenase (LDH) release in H9c2 cells. The protein levels of p-JNK and p-p38 were significantly downregulated by Bilo treatment. SB20358 (inhibitor of p38) and SP600125 (inhibitor of JNK) suppressed OGD-induced cell apoptosis as Bilo did. In MI mouse model, Bilo improved the cardiac function and significantly reduced the infarct size and myocardial fibrosis. Bilo inhibited MI-induced cardiomyocytes apoptosis in mice. Bilo suppressed the protein levels of p-JNK and p-p38 in cardiac tissues from MI mice. Bilo alleviated OGD-induced cell apoptosis in H9c2 cells and suppressed MI-induced cardiomyocyte apoptosis and myocardial fibrosis in mice via the inactivation of JNK/p38 MAPK signaling pathways. Thus, Bilo may be an effective anti-MI agent.


Subject(s)
Bilobalides , Myocardial Infarction , Mice , Animals , Bilobalides/pharmacology , Myocardial Infarction/drug therapy , Myocardial Infarction/metabolism , Myocytes, Cardiac/metabolism , p38 Mitogen-Activated Protein Kinases/genetics , p38 Mitogen-Activated Protein Kinases/metabolism , p38 Mitogen-Activated Protein Kinases/pharmacology , Apoptosis , Fibrosis
2.
Zhongguo Zhong Yao Za Zhi ; 48(15): 4201-4207, 2023 Aug.
Article in Chinese | MEDLINE | ID: mdl-37802788

ABSTRACT

This study aims to explore the neuroprotective effect of bilobalide(BB) and the mechanisms such as inhibiting inflammatory response in macrophage/microglia, promoting neurotrophic factor secretion, and interfering with the activation and differentiation of peripheral CD4~+ T cells. BB of different concentration(12.5, 25, 50, 100 µg·mL~(-1)) was used to treat the RAW264.7 and BV2 cells for 24 h. The 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide(MTT) assay and cell counting kit-8(CCK-8) were employed to detect the cytotoxicity of BB and appropriate concentration was selected for further experiment. Lipopolysaccharide(LPS) was applied to elicit inflammation in RAW264.7 and BV2 cells, mouse bone marrow-derived macrophages(BMDMs), and primary microglia, respectively. The effect of BB on cell proliferation and secretion of inflammatory cytokines and neurotrophic factors was detected by enzyme-linked immunosorbent assay(ELISA). Spleen monocytes of C57BL/6 female mice(7-8 weeks old) were isolated, and CD4~+ T cells were separated by magnetic beads under sterile conditions. Th17 cells were induced by CD3/CD28 and the conditioned medium for eliciting the inflammation in BMDMs. The content of IL-17 cytokines in the supernatant was detected by ELISA to determine the effect on the activation and differentiation of CD4~+ T cells. In addition, PC12 cells were incubated with the conditioned medium for eliciting inflammation in BMDMs and primary microglia and the count and morphology of cells were observed. The cytoto-xicity was determined by lactate dehydrogenase(LDH) assay. The result showed that BB with the concentration of 12.5-100 µg·mL~(-1) had no toxicity to RAW264.7 and BV2 cells, and had no significant effect on the activity of cell model with low inflammation. The 50 µg·mL~(-1) BB was selected for further experiment, and the results indicated that BB inhibited LPS-induced secretion of inflammatory cytokines. The experiment on CD4~+ T cells showed that the conditioned medium for LPS-induced inflammation in BMDMs promoted the activation and differentiation of CD4~+ T cells, while the conditioned medium of the experimental group with BB intervention reduced the activation and differentiation of CD4~+ T cells. In addition, BB also enhanced the release of neurotrophic factors from BMDMs and primary microglia. The conditioned medium after BB intervention can significantly reduce the death of PC12 neurons, inhibit neuronal damage, and protect neurons. To sum up, BB plays a neuroprotective role by inhibiting macrophage and microglia-mediated inflammatory response and promoting neurotrophic factors.


Subject(s)
Bilobalides , Female , Rats , Mice , Animals , Bilobalides/pharmacology , Neuroprotection , Lipopolysaccharides/toxicity , Culture Media, Conditioned/metabolism , Culture Media, Conditioned/pharmacology , Mice, Inbred C57BL , Macrophages/metabolism , Microglia , Cytokines/metabolism , Nerve Growth Factors/metabolism , Nerve Growth Factors/pharmacology , Inflammation/metabolism
3.
Article in Chinese | WPRIM (Western Pacific) | ID: wpr-1008616

ABSTRACT

This study aims to explore the neuroprotective effect of bilobalide(BB) and the mechanisms such as inhibiting inflammatory response in macrophage/microglia, promoting neurotrophic factor secretion, and interfering with the activation and differentiation of peripheral CD4~+ T cells. BB of different concentration(12.5, 25, 50, 100 μg·mL~(-1)) was used to treat the RAW264.7 and BV2 cells for 24 h. The 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide(MTT) assay and cell counting kit-8(CCK-8) were employed to detect the cytotoxicity of BB and appropriate concentration was selected for further experiment. Lipopolysaccharide(LPS) was applied to elicit inflammation in RAW264.7 and BV2 cells, mouse bone marrow-derived macrophages(BMDMs), and primary microglia, respectively. The effect of BB on cell proliferation and secretion of inflammatory cytokines and neurotrophic factors was detected by enzyme-linked immunosorbent assay(ELISA). Spleen monocytes of C57BL/6 female mice(7-8 weeks old) were isolated, and CD4~+ T cells were separated by magnetic beads under sterile conditions. Th17 cells were induced by CD3/CD28 and the conditioned medium for eliciting the inflammation in BMDMs. The content of IL-17 cytokines in the supernatant was detected by ELISA to determine the effect on the activation and differentiation of CD4~+ T cells. In addition, PC12 cells were incubated with the conditioned medium for eliciting inflammation in BMDMs and primary microglia and the count and morphology of cells were observed. The cytoto-xicity was determined by lactate dehydrogenase(LDH) assay. The result showed that BB with the concentration of 12.5-100 μg·mL~(-1) had no toxicity to RAW264.7 and BV2 cells, and had no significant effect on the activity of cell model with low inflammation. The 50 μg·mL~(-1) BB was selected for further experiment, and the results indicated that BB inhibited LPS-induced secretion of inflammatory cytokines. The experiment on CD4~+ T cells showed that the conditioned medium for LPS-induced inflammation in BMDMs promoted the activation and differentiation of CD4~+ T cells, while the conditioned medium of the experimental group with BB intervention reduced the activation and differentiation of CD4~+ T cells. In addition, BB also enhanced the release of neurotrophic factors from BMDMs and primary microglia. The conditioned medium after BB intervention can significantly reduce the death of PC12 neurons, inhibit neuronal damage, and protect neurons. To sum up, BB plays a neuroprotective role by inhibiting macrophage and microglia-mediated inflammatory response and promoting neurotrophic factors.


Subject(s)
Female , Rats , Mice , Animals , Bilobalides/pharmacology , Neuroprotection , Lipopolysaccharides/toxicity , Culture Media, Conditioned/pharmacology , Mice, Inbred C57BL , Macrophages/metabolism , Microglia , Cytokines/metabolism , Nerve Growth Factors/pharmacology , Inflammation/metabolism
4.
Arthritis Res Ther ; 24(1): 197, 2022 08 18.
Article in English | MEDLINE | ID: mdl-35982488

ABSTRACT

BACKGROUND: Uncoupled extracellular matrix (ECM) causes cartilage degeneration and osteoarthritis (OA) by suppressing the synthesis and activating the degradation of ECM components. Gingko biloba is a natural Chinese herb with a variety of biological functions; however, the extent to which it can protect against OA and the mechanisms involved are unknown. METHODS: In our study, using bioinformatics tools, we were able to identify an important lactone, bilobalide (BB), from Gingko biloba. In vitro experiments were performed to evaluate the potential therapeutic effects of BB on ECM homeostasis. In vivo experiments were conducted to assess the protection of systemic administration of BB on cartilage degeneration. Molecular mechanisms underlying BB-regulated anti-arthritic role were further explored. RESULTS: In interleukin-1ß-incubated human chondrocytes, in vitro treatment with BB increased the expression of cartilage anabolic proteins, while inhibiting the activities of ECM degrading enzymes. In a mice model, systemic administration of BB, in vivo, prevented post-traumatic cartilage erosion and attenuated the formation of abnormal osteophytes in the subchondral bone. Mechanistically, the activation of the adenosine 5'-monophosphate-activated protein kinase (AMPK)-sirtuin 1 (SIRT1) signaling pathway was involved in the anti-arthritic effects of BB. In vitro, blocking BB's chondroprotection with the AMPK-specific inhibitor Compound C abrogated it. CONCLUSIONS: These results demonstrated that BB extracted from Gingko biloba regulates ECM balance to prevent OA by activating the AMPK-SIRT1 signaling pathway. This study proposed the monomer BB, a traditional Chinese medicine, as a de novo therapeutic insight for OA. Schematic representation of the experimental design. Based on the bioinformatic analysis, bilobalide (BB), a natural herb Gingko biloba-derived ingredient, was identified as a candidate for treating osteoarthritis. In vitro, BB treatment not only facilitates cartilage extracellular matrix synthesis but also inhibits proteolytic enzyme activities. In vivo intraperitoneal injection of BB improves cartilage degeneration and subchondral bone sclerosis. BB, in particular, had anti-arthritic effects by activating the AMPK-SIRT1 signaling pathway.


Subject(s)
Bilobalides , Lactones , Osteoarthritis , AMP-Activated Protein Kinases/metabolism , Animals , Bilobalides/pharmacology , Chondrocytes/metabolism , Ginkgo biloba/chemistry , Humans , Lactones/pharmacology , Mice , Osteoarthritis/drug therapy , Osteoarthritis/metabolism , Osteoarthritis/prevention & control , Signal Transduction , Sirtuin 1/metabolism
5.
Appl Biochem Biotechnol ; 194(12): 6407-6422, 2022 Dec.
Article in English | MEDLINE | ID: mdl-35932369

ABSTRACT

Diabetic retinopathy (DR) is a diabetes mellitus (DM) complication that causes visual acuity impairment and loss of sight in the working population, mainly in developed countries. According to the WHO, DR accounts for 5% of the world's 37 million blind people. The prevalence of diabetic retinopathy was highest in Africa, followed by North America and the Caribbean and South and Central America. Hyperglycemia can generate excessive ROS that activates multiple pathways, which can damage the cells. Oxidative stress and inflammatory process are intricate in the DR pathological mechanism. Bilobalide is the main bioactive compound isolated from the Ginkgo biloba, a plant utilized in folklore medicine. Bilobalide, a sesquiterpene trilactone, exhibits excellent antioxidant activity. But the molecular mechanisms associated with such effects, especially the antioxidant-related mechanism, have not been documented. Hence, this investigation explored whether bilobalide may attenuate DR in streptozotocin (STZ)-prompted diabetic rats. The effects of bilobalide on parameters of antioxidant content, oxidative stress, and inflammatory factors in the retinal tissues were evaluated by ELISA, RT-PCR, and immunohistochemistry methods. Bilobalide improved caloric management by reducing food consumption and increasing body weight. Furthermore, the administration of bilobalide decreases the blood glucose level and glycosylated (HbA1c) hemoglobin. The anti-retinopathy activity of bilobalide was established by the increase in the total retina thickness (TRT), inner nuclear layer (INL), and outer nuclear layer (ONL) in diabetic rats. Additionally, the serum level of MDA was decreased. In contrast, the antioxidant enzyme (SOD and CAT) levels were increased with TAC plus lower Keap1 and higher Nrf2 expression in the retina when associated with the DM rats. Moreover, bilobalide increased the nuclear factor erythroid 2-related factor 2 (Nrf2) and Heme oxygenase-1 (HO-1) expression level and inflammatory mediators (NF-κß p65, TNF-α, IL-1ß, and VEGF), thus inhibiting oxidative stress. Bilobalide can be effective against DR, and the possible mechanism may be relatively elucidated by decreasing oxidative stress and anti-inflammatory activities. But the further investigation should be directed to expose the precise mechanism.


Subject(s)
Bilobalides , Diabetes Mellitus, Experimental , Diabetic Retinopathy , Rats , Animals , Streptozocin/adverse effects , NF-E2-Related Factor 2/metabolism , Diabetic Retinopathy/drug therapy , Diabetic Retinopathy/prevention & control , Diabetic Retinopathy/complications , Antioxidants/pharmacology , Diabetes Mellitus, Experimental/complications , Diabetes Mellitus, Experimental/drug therapy , Kelch-Like ECH-Associated Protein 1/metabolism , Bilobalides/pharmacology , Rats, Sprague-Dawley , Oxidative Stress , Retina/metabolism , Retina/pathology , Inflammation/drug therapy
6.
Food Funct ; 12(14): 6226-6239, 2021 Jul 21.
Article in English | MEDLINE | ID: mdl-34096560

ABSTRACT

Dysfunction of the intestinal epithelial barrier and intestinal microbiota dysbiosis can drive the onset or aggravation of ulcerative colitis (UC). Bilobalide (BI) is an extract of Ginkgo biloba that has been shown to exhibit a range of anti-inflammatory properties. Herein, we explored functional and mechanistic effects of BI treatment in a rodent model of DSS-induced UC. These analyses revealed that BI treatment was sufficient to reduce disease severity, increase colon length, and normalize colon histological characteristics relative to those observed in DSS-treated model mice. BI also enhanced the expression of tight junction proteins associated with intestinal barrier integrity including ZO-1, Occludin, and Claudin-3. Through 16S rDNA sequencing analyses, BI was also found to influence the overall richness of the intestinal microbiome, promoting the proliferation of probiotic species including Lactobacillus. Consistent with these in vivo findings, BI treatment protected RAW264.7 cells against lipopolysaccharide (LPS)-induced inflammatory damage, suppressing the activation of the AKT/NF-κB p65 and MAPK signaling pathways in this experimental context. In summary, these findings revealed that BI can suppress MAPK and AKT/NF-κB p65 signaling, thereby suppressing the production of inflammatory cytokines including IL-1ß, IL-6, and TNF-α, while additionally alleviating UC severity by facilitating repair of the intestinal epithelial barrier and the remodeling of intestinal microbial communities.


Subject(s)
Anti-Inflammatory Agents/pharmacology , Bilobalides/pharmacology , Colitis, Ulcerative/drug therapy , Ginkgo biloba/chemistry , Plant Extracts/pharmacology , Animals , Colitis, Ulcerative/metabolism , Colon/metabolism , Cytokines/metabolism , Disease Models, Animal , Dysbiosis/drug therapy , Dysbiosis/metabolism , Gastrointestinal Microbiome/drug effects , Lipopolysaccharides/adverse effects , Mice , Mice, Inbred C57BL , NF-kappa B/metabolism , RAW 264.7 Cells , Signal Transduction/drug effects , Tight Junction Proteins/metabolism
7.
Biomed Res Int ; 2021: 8835408, 2021.
Article in English | MEDLINE | ID: mdl-33959665

ABSTRACT

This study was aimed at examining the effect and underlying mechanisms of bilobalide (BB) on hepatic injury in streptozotocin- (STZ-) induced diabetes mellitus (DM) in immature rats. Immature rats (one day old) were randomly divided into five groups: group I, control nondiabetic rats; group II, STZ-induced, untreated diabetic rats; groups III/IV/V, STZ-induced and BB-treated diabetic rats, which were intraperitoneally injected with BB (2.5 mg/kg, 5 mg/kg, or 10 mg/kg) after 3 days followed by STZ treatment. We observed that BB improved the histopathological changes and maintained normal glucose metabolism, blood lipid, and liver function indicators, such as fasting blood glucose, obesity index, HbA1c, HOMA-IR, fast serum insulin, adiponectin, total cholesterol (TC), triglyceride (TG), high-density lipoprotein (HDL), low-density lipoprotein (LDL), aspartate transaminase (AST), and alanine transaminase (ALT) in STZ-induced DM in immature rats by a biochemical analyzer or ELISA. Meanwhile, Western blot analysis showed that in STZ-induced DM immature rats, BB decreased the expression of apoptosis-related proteins Bax, cleaved caspase-3, and cleaved caspase-9 while enhancing the Bcl-2 expression; BB downregulated the expression of ACC related to fat anabolism, while upregulating the expression of CPT-1 related to fat catabolism. Strikingly, treatment with BB significantly increased the expression of AMPKα1 as well as inhibited HMGB1, TLR4, and p-P65 expression in hepatic tissues of immature DM rats. AMPK inhibitor (compound C, CC) cotreated with BB undermined the protective effect of BB on the liver injury. The results of the present study suggested BB may have a significant role in alleviating liver damage in the STZ-induced immature DM rats.


Subject(s)
AMP-Activated Protein Kinases/metabolism , Bilobalides/pharmacology , Diabetes Mellitus, Experimental/metabolism , Liver Diseases/metabolism , Signal Transduction/drug effects , Animals , Apoptosis/drug effects , Arabidopsis Proteins/metabolism , Blood Glucose/drug effects , DNA-Binding Proteins/metabolism , Lipid Metabolism/drug effects , Liver/drug effects , Liver/metabolism , Male , NF-kappa B/metabolism , Rats , Rats, Wistar , Toll-Like Receptor 4/metabolism
8.
J Biochem Mol Toxicol ; 35(5): e22723, 2021 May.
Article in English | MEDLINE | ID: mdl-33511709

ABSTRACT

Gastric carcinoma is one of the most aggressive types of cancer that ranks fifth among all cancer incidences and third in cancer mortality. As it exhibits a prolonged asymptomatic condition and high recurrence rate, it is a great challenge to treat gastric cancer. Traditional medicine that utilizes herbal phytochemicals to treat various diseases is a potent alternative for current allopathic treatment. Hence, we evaluated the potency of a phytochemical bilobalide for treating gastric cancer in in vitro and in vivo models. Bilobalide, a sesquiterpenoid, is present in the Ginkgo biloba plant that belongs to the family of Ginkgoaceae. The cytotoxicity effect of bilobalide was evaluated in both gastric cancer (AGS) cells and normal gastric epithelial cells. Apoptosis-inducing property of bilobalide against the AGS cell line was analyzed with different fluorescent staining techniques and terminal deoxynucleotidyl transferase dUTP nick-end labeling assay, and cell cycle analysis was carried out by flow cytometry. The in vivo studies were assessed with N-methyl-N-nitrosourea (MNU)-induced gastric cancer in rats. Serum-specific gastric markers were quantified and histopathological analysis of stomach tissue was performed. The expression of target-signaling molecules was analyzed by a reverse-transcription polymerase chain reaction. The in vitro results proved that bilobalide effectively suppressed the AGS cell growth and induced cell death by nuclear damage and apoptosis induction. The bilobalide treatment effectively arrested the cell cycle of AGS cells via inhibiting the PI3K-signaling pathway. Our in vivo results also confirmed that the bilobalide persuasively inhibited the MNU-induced gastric carcinoma via inhibiting the thioredoxin-fold family proteins and inflammatory markers' expression. Overall, our results authentically prove that bilobalide possesses therapeutic potency to cure gastric carcinoma.


Subject(s)
Apoptosis/drug effects , Bilobalides/pharmacology , Cell Proliferation/drug effects , Neoplasms, Experimental/drug therapy , Plant Extracts/chemistry , Stomach Neoplasms/drug therapy , Animals , Bilobalides/chemistry , Cell Line, Tumor , Ginkgo biloba , Humans , Male , Methylnitrosourea/toxicity , Neoplasms, Experimental/chemically induced , Neoplasms, Experimental/metabolism , Neoplasms, Experimental/pathology , Rats , Rats, Wistar , Stomach Neoplasms/chemically induced , Stomach Neoplasms/metabolism , Stomach Neoplasms/pathology
9.
Biochem Biophys Res Commun ; 526(4): 1077-1084, 2020 06 11.
Article in English | MEDLINE | ID: mdl-32312522

ABSTRACT

Bilobalide, one of the key bioactive components of Ginkgo biloba leaves, exerts prominent neuroprotective properties in central nervous system (CNS) disease. However, the effect of bilobalide on blood-brain barrier (BBB) permeability remains unknown. In this study, we investigated the effect of bilobalide on BBB permeability and its potential mechanism involved. Both the in vitro and in vivo results showed that significant enhancement of BBB permeability was found following bilobalide treatment, evidenced by the reduced transendothelial electrical resistance (TEER), the increased fluorescein sodium (Na-F) penetration rate in vitro and the leakage of FITC-dextran in vivo. Transmission electron microscope (TEM) images demonstrated that bilobalide modulated BBB permeability by changing the ultrastructure of tight junctions (TJs). In addition, actin-binding proteins ezrin, radixin and moesin (ERM) and Myosin light chain (MLC) phosphorylation was observed following bilobalide treatment. Moreover, the effect of bilobalide on TEER reduction and ERM/MLC phosphorylation was counteracted by adenosine A1 receptor (A1R) siRNA. The current findings suggested that bilobalide might reversibly modulate BBB permeability by the alteration of TJs ultrastructure through A1R-mediated phosphorylation of actin-binding proteins.


Subject(s)
Bilobalides/pharmacology , Blood-Brain Barrier/metabolism , Microfilament Proteins/metabolism , Receptor, Adenosine A1/metabolism , Animals , Blood-Brain Barrier/drug effects , Dextrans/metabolism , Fluorescein-5-isothiocyanate/analogs & derivatives , Fluorescein-5-isothiocyanate/metabolism , Humans , Male , Mice , Molecular Weight , Permeability/drug effects , Phosphorylation/drug effects , Signal Transduction/drug effects , Tight Junction Proteins/metabolism
10.
Molecules ; 24(19)2019 Sep 27.
Article in English | MEDLINE | ID: mdl-31569605

ABSTRACT

Bilobalide, the only sesquiterpene compound from Ginkgo biloba leaf, exhibits various beneficial pharmaceutical activities, such as antioxidant, anti-inflammation, and protective effects for the central nervous system. Several bioactive components extracted from Ginkgo biloba extract reportedly have the potential to attenuate lipid metabolism. However, the effect of bilobalide on lipid metabolism remains unclear. In this study, we used 3T3-L1 cells as the cell model to investigate the effect of bilobalide on adipogenesis. The results showed that bilobalide inhibited 3T3-L1 preadipocyte differentiation and intracellular lipid accumulation. Quantitative real-time PCR and western blotting results indicated that several specific adipogenic transcription factors and a few important adipogenesis-related genes were significantly down regulated on both mRNA and protein levels in bilobalide treatment groups. By contrast, the expression of some lipolytic genes, such as adipose triglyceride lipase, hormone-sensitive lipase (HSL), and carnitine palmitoyltransferase-1α, were all up-regulated by bilobalide treatment, and the phosphorylation of AMP-activated protein kinase (AMPK), acetyl-CoA carboxylase 1, and HSL were stimulated. Furthermore, bilobalide treatment partially restored AMPK activity following its blockade by compound C (dorsomorphin). These results suggested that bilobalide inhibited adipogenesis and promoted lipolysis in 3T3-L1 cells by activating the AMPK signaling pathway.


Subject(s)
Adipocytes/drug effects , Adipocytes/metabolism , Adipogenesis/drug effects , Bilobalides/pharmacology , MAP Kinase Signaling System/drug effects , 3T3-L1 Cells , Adipogenesis/genetics , Animals , Bilobalides/chemistry , Cell Survival/drug effects , Gene Expression Regulation/drug effects , Ginkgo biloba , Lipolysis/drug effects , Lipolysis/genetics , Mice , Molecular Structure , Plant Extracts/chemistry , Plant Extracts/pharmacology , Transcription Factors/genetics , Transcription Factors/metabolism
11.
Biomed Pharmacother ; 96: 792-797, 2017 Dec.
Article in English | MEDLINE | ID: mdl-29054095

ABSTRACT

The accumulation of aggregated forms of the α-Synuclein (α-Syn) is associated with the pathogenesis of Parkinson's disease (PD), a chronic progressive neurodegenerative disorder. Extensive evidences have shown the promising effects of Ginkgo biloba consumption on motor activity after PD. However, the mechanisms underline the α-Syn-induced cell damage and whether ginkgolides exert neuroprotection against this injury are unclear. Here we showed that aggregated recombinant human α-Syn, but not α-Syn monomers, triggered cell injury in cultured human neuroblastoma cell line SH-SY5Y in an apoptosis way by using flow cytometry and western blot assay. Moreover, pre-treatment with the Ginkgolide B (GB) or Bilobalide (BB) protected SH-SY5Y cells against α-Syn-induced cell viability decreases, and reduced cell apoptosis after aggregated α-Syn stimulation. Together, we firstly find that aggregated α-Syn induced cell apoptosis and GB and BB may attenuate aggregated α-Syn-induced cell apoptosis, which gives us an insight into the novel therapy for PD in future.


Subject(s)
Apoptosis/drug effects , Bilobalides/pharmacology , Ginkgolides/pharmacology , Lactones/pharmacology , Neurons/drug effects , Plant Extracts/pharmacology , alpha-Synuclein/metabolism , Cell Line, Tumor , Cell Survival/drug effects , Ginkgo biloba , Humans , Neuroblastoma/drug therapy , Neuroblastoma/metabolism , Neuroprotective Agents/pharmacology
12.
Eur J Pharmacol ; 777: 136-46, 2016 Apr 15.
Article in English | MEDLINE | ID: mdl-26953225

ABSTRACT

The terpenoid lactones from Ginkgo biloba, bilobalide and ginkgolides, have been shown to act as negative modulators at α1ß2γ2L GABAA receptors. They have structural features similar to those of the chloride channel blocker picrotoxinin. Unlike picrotoxinin, however they are not known to produce convulsant effects. Using two-electrode voltage clamp electrophysiology, this study compared the effect of mutation of 2', 6' and 15' pore facing M2 domain residues to cysteine on the action of picrotoxinin, bilobalide and ginkgolides at α1ß2γ2L GABAA receptors expressed in Xenopus oocytes. Picrotoxinin was affected by mutation differently from the ginkgo terpenoid lactones. Although some of these compounds were affected by the mutation at same position and/or subunit, the changes in their potency were found to be dissimilar. The results suggest that the intracellular pore binding site for picrotoxinin, bilobalide, ginkgolide A, ginkgolide B and ginkgolide C is comprised of 2'ß-6'ß6'γ, 2'α2'ß-6'α6'ß, 2'α2'ß2'γ-6'ß6'γ, 2'α, 2'ß2'γ-6'ß and 2'α2'ß, respectively. Unlike bilobalide and ginkgolides, the inhibitory action of picrotoxinin was not affected by mutations at 15' position. It is proposed that 15'α15'ß, 15'ß, 15'α15'ß and 15'α15'ß15'γ forms an extracellular pore binding site for bilobalide, ginkgolide A, ginkgolide B and ginkgolide C, respectively. The lack of convulsant effects of bilobalide, and ginkgolide A and B may be associated in part with their different binding locations within the chloride channel.


Subject(s)
Bilobalides/pharmacology , Cysteine , GABA-A Receptor Antagonists/pharmacology , Ginkgo biloba/chemistry , Ginkgolides/pharmacology , Mutation , Receptors, GABA-A/metabolism , Animals , Base Sequence , Bilobalides/metabolism , Binding Sites , Dose-Response Relationship, Drug , GABA-A Receptor Antagonists/metabolism , Ginkgolides/metabolism , Humans , Protein Binding , Receptors, GABA-A/chemistry , Receptors, GABA-A/genetics , Sulfhydryl Compounds/metabolism , gamma-Aminobutyric Acid/pharmacology
13.
Cell Mol Neurobiol ; 34(6): 913-23, 2014 Aug.
Article in English | MEDLINE | ID: mdl-24838256

ABSTRACT

Bilobalide, a natural product extracted from Ginkgo biloba leaf, is known to exhibit a number of pharmacological activities. So far, whether it could affect embryonic stem cell differentiation is still unknown. The main aim of this study was to investigate the effect of bilobalide on P19 embryonic carcinoma cells differentiation and the underlying mechanisms. Our results showed that bilobalide induced P19 cells differentiation into neurons in a concentration- and time-dependent manner. We also found that bilobalide promoted neuronal differentiation through activation of Wnt/ß-catenin signaling pathway. Exposure to bilobalide increased inactive GSK-3ß phosphorylation, further induced the nuclear accumulation of ß-catenin, and also up-regulated the expression of Wnt ligands Wnt1 and Wnt7a. Neuronal differentiation induced by bilobalide was totally abolished by XAV939, an inhibitor of Wnt/ß-catenin pathway. These results revealed a novel role of bilobalide in neuronal differentiation from P19 embryonic cells acting through Wnt/ß-catenin signaling pathway, which would provide a better insight into the beneficial effects of bilobalide in brain diseases.


Subject(s)
Bilobalides/pharmacology , Cell Differentiation , Embryonal Carcinoma Stem Cells/drug effects , Embryonic Stem Cells/cytology , Neurons/cytology , Wnt Signaling Pathway/drug effects , beta Catenin/metabolism , Animals , Cell Differentiation/drug effects , Cell Proliferation/drug effects , Embryonal Carcinoma Stem Cells/cytology , Glycogen Synthase Kinase 3/drug effects , Glycogen Synthase Kinase 3/metabolism , Glycogen Synthase Kinase 3 beta , Mice , Neurons/metabolism
14.
Rev. bras. plantas med ; 16(4): 819-825, oct.-dic. 2014. graf
Article in English | LILACS | ID: lil-729889

ABSTRACT

Electrocardiographic effects produced by Ginkgo biloba extract (EGb) and by ginkgolides A (GA) and B (GB), and bilobalide (BB) were investigated in guinea pig heart mounted in Langendorff apparatus (Tyrode, 34 ± 0.1 ºC, 95% O2, 5% CO2). Electrocardiographic parameters were evaluated in the conditions: 1) control with Tyrode and DMSO, 2) EGb (n=4), GA (n=5), GB (n=5) or BB (n=6), and 3) washout. The results showed that 0.1 and 1.0 mg/ml of EGb do not change the electrocardiographic parameters. However, 10 mg/ml of EGb increased the PR interval (PRi) at 21% (p<0.001). This increase was also observed for 50 mM GA (20%, p<0.001) and 70 mM BB (13%, p<0.001), which indicates Ca2+ channel block. However, the 50 mM GB reduced the PRi at 11 % (p<0.001). The GA (23%, p<0.001), GB (16%, p<0.001), and BB (40%, p<0.001) reduced the QT interval (QTi), which suggests the activation of the potassium channel. However, EGb increased QTi (6%, p<0.001). The EGb (28%, p<0.05) and GB (13%, p<0.05) reduced the heart rate. Atrioventricular (AV) block was observed with EGb, GA, and BB. We can conclude that EGb and its terpenoids alter the ECG parameters inducing AV block, which indicates possible arrhythmogenic potential.


Os efeitos eletrocardiográficos produzidos pelo extrato de Ginkgo biloba (EGb) e gingkolídeos A (GA) e B (GB), e bilobalide (BB) foram investigados em coração de cobaia montado sistema de Langendorff (Tyrode, 34 ± 0.1 ºC, 95% O2, 5% CO2). Os parâmetros do ECG foram avaliados nas condições: 1) Tyrode e DMSO, 2) EGb (n=4), GA (n=5), GB (n=5) ou BB (n=6) diluídos em DMSO e 3) washout. Os resultados demonstram que 0,1 e 1,0 mg/mL de EGb não alteraram os parâmetros eletrocardiográficos. Entretanto, 10 mg/ml de EGb aumentaram o intervalo PR (PRi) em 21% (p<0.001). Esse aumento também foi observado com GA a 50µM (20%, p<0,001) e BB a 70 mM (13%, p<0,001) indicando bloqueio de canais de cálcio. Por outro lado, GB reduziu o PRi (11%, p<0,001). O intervalo QT (QTi) foi reduzido por GA (23%, p<0,001), GB (16%, p<0,001) e BB (40%, p < 0.001) sugerindo uma ativação de canais de potássio. Entretanto, EGb aumentou o QTi (6%, p<0.001). A frequência cardíaca foi reduzida por EGb (28%, p<0.05) e GB (13%, p<0.05). Bloqueios átrio-ventriculares (BAV) foram observados com EGb, GA e BB. Podemos concluir que EGb e os terpenos alteram parâmetros eletrocardiográficos induzindo BAV e demonstrando possível potencial arritmogênico.


Subject(s)
Guinea Pigs , Terpenes/analysis , Plant Extracts/antagonists & inhibitors , Ginkgo biloba/adverse effects , Electrocardiography , Ginkgolides/analysis , Bilobalides/pharmacology , Heart/drug effects
15.
Neurosci Lett ; 435(2): 147-51, 2008 Apr 18.
Article in English | MEDLINE | ID: mdl-18329806

ABSTRACT

Bilobalide, a major bioactive component of Ginkgo biloba herbal extracts, exhibits neuroprotective and anti-ischaemic activity. However, its therapeutic potential is limited because of its instability. Attempts to synthesise a more stable analogue culminated in the development of NV-31. This compound recapitulates some aspects of bilobalide pharmacology. However, although bilobalide inhibits recombinant glycine receptor Cl channels (GlyRs), NV-31 potentiates hippocampal neuron GlyRs. Because of the possible therapeutic relevance of this effect, the present study investigated the molecular mechanism and subunit specificity of NV-31 actions at recombinantly expressed alpha1, alpha1beta, alpha2 and alpha3 GlyRs. NV-31 potentiated alpha1 GlyRs by approximately 135% with an EC50 near 170 nM. Its potentiating effect was observed only at low (EC10) glycine concentrations. The magnitude of its potentiating effect was reduced at alpha1beta GlyRs and it had no effect at all at alpha2 and alpha3 GlyRs. NV-31 was unlikely to bind at the bilobalide pore-binding site as its efficacy was not affected by the alpha1 subunit G2'A and T6'S mutations. However, the S15'C mutation to the alcohol-binding site abolished its effects, suggesting that NV-31 modulates the GlyR via a specific (steric or allosteric) interaction with S15'. GlyRs are potential therapeutic targets for chronic anti-inflammatory pain and movement disorders. NV-31, as a positive modulator of these receptors, thus remains viable as a therapeutic candidate for these disorders.


Subject(s)
Bilobalides/pharmacology , Ginkgo biloba/chemistry , Protein Subunits/metabolism , Pyrans/pharmacology , Receptors, Glycine/agonists , Bilobalides/chemistry , Cell Line, Transformed , Dose-Response Relationship, Drug , Dose-Response Relationship, Radiation , Electric Stimulation/methods , GABA Antagonists/pharmacology , Glycine/pharmacology , Humans , Membrane Potentials/drug effects , Membrane Potentials/physiology , Membrane Potentials/radiation effects , Mutation/physiology , Patch-Clamp Techniques , Picrotoxin/pharmacology , Protein Subunits/genetics , Receptors, Glycine/genetics , Transfection/methods
16.
Zhonghua Yi Xue Za Zhi ; 87(7): 485-8, 2007 Feb 13.
Article in Chinese | MEDLINE | ID: mdl-17459230

ABSTRACT

OBJECTIVE: To investigate the effect of extract of Ginkgo biloba leaves (EGb50) on the expression of inducible nitric oxide synthase (iNOS) after sciatic nerve injury. METHODS: 156 male SD rats were randomly divided into 3 groups: experiment group (n=72), undergoing section and anastomosis of sciatic nerve and then gastric perfusion of EGb50 200 mg.kg-1.d-1; injury control group (n=72), undergoing section and anastomosis of sciatic nerve and then gastric perfusion of normal saline daily; and sham operation group (n=12), undergoing sham operation and gastric perfusion of normal saline daily. The rats were killed at different time points: 1, 3, 7, 14, 21, and 28 days after the operation. The sciatic nerve distant to the anastomotic sites with a length of 0.5 cm was cut out to undergo immunohistochemistry and RT-PCR to detect the protein and mRNA expression of iNOS. RESULTS: No obvious iNOS protein and mRNA expression was seen in the sham operation group. 3, 7, 14, and 21 days after operation the mean values of absorbance of iNOS in the sciatic nerve specimens of the experiment group were all significantly lower than those of the injury control group (P<0.05, P<0.01). High mRNA expression of iNOS began to be shown in the sciatic nerve specimens since 1day after the operation and remained at a high level till 7 days after and then began to decrease in the injury control group. The levels of mRNA expression of iNOS in the sciatic nerve specimens 1, 3, and 7 days after the operation of the experiment group were all significantly lower than those of the injury control group (all P<0.01), and there was no significant difference in the levels mRNA expression of iNOS 14, 21, and 28 days after the operation between the experiment group and injury control group (all>0.05). CONCLUSION: The extract of Gingko biloba promotes the regeneration of nervous tissues, probably by inhibiting the expression of iNOS.


Subject(s)
Ginkgo biloba/chemistry , Nitric Oxide Synthase Type II/biosynthesis , Plant Extracts/pharmacology , Plant Leaves/chemistry , Sciatic Nerve/drug effects , Animals , Bilobalides/pharmacology , Immunohistochemistry , Male , Nitric Oxide Synthase Type II/genetics , Plant Extracts/chemistry , RNA, Messenger/genetics , RNA, Messenger/metabolism , Random Allocation , Rats , Rats, Sprague-Dawley , Reverse Transcriptase Polymerase Chain Reaction , Sciatic Nerve/injuries , Sciatic Nerve/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...