Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 53
Filtrar
1.
FASEB J ; 38(14): e23817, 2024 Jul 31.
Artigo em Inglês | MEDLINE | ID: mdl-39003633

RESUMO

Excessive apoptosis of intestinal epithelial cells leads to intestinal barrier dysfunction, which is not only one of the pathological features of inflammatory bowel disease (IBD) but also a therapeutic target. A natural plant extract, Ginkgetin (GK), has been reported to have anti-apoptotic activity, but its role in IBD is unknown. This study aimed to explore whether GK has anti-colitis effects and related mechanisms. An experimental colitis model induced by dextran sulfate sodium (DSS) was established, and GK was found to relieve colitis in DSS-induced mice as evidenced by improvements in weight loss, colon shortening, Disease Activity Index (DAI), macroscopic and tissue scores, and proinflammatory mediators. In addition, in DSS mice and TNF-α-induced colonic organoids, GK protected the intestinal barrier and inhibited intestinal epithelial cell apoptosis, by improving permeability and inhibiting the number of apoptotic cells and the expression of key apoptotic regulators (cleaved caspase 3, Bax and Bcl-2). The underlying mechanism of GK's protective effect was explored by bioinformatics, rescue experiments and molecular docking, and it was found that GK might directly target and activate EGFR, thereby interfering with PI3K/AKT signaling to inhibit apoptosis of intestinal epithelial cells in vivo and in vitro. In conclusion, GK inhibited intestinal epithelial apoptosis in mice with experimental colitis, at least in part, by activating EGFR and interfering with PI3K/AKT activation, explaining the underlying mechanism for ameliorating colitis, which may provide new options for the treatment of IBD.


Assuntos
Apoptose , Biflavonoides , Colite , Sulfato de Dextrana , Células Epiteliais , Receptores ErbB , Mucosa Intestinal , Camundongos Endogâmicos C57BL , Fosfatidilinositol 3-Quinases , Proteínas Proto-Oncogênicas c-akt , Transdução de Sinais , Animais , Apoptose/efeitos dos fármacos , Camundongos , Proteínas Proto-Oncogênicas c-akt/metabolismo , Colite/induzido quimicamente , Colite/tratamento farmacológico , Colite/metabolismo , Colite/patologia , Receptores ErbB/metabolismo , Fosfatidilinositol 3-Quinases/metabolismo , Transdução de Sinais/efeitos dos fármacos , Mucosa Intestinal/metabolismo , Mucosa Intestinal/efeitos dos fármacos , Mucosa Intestinal/patologia , Sulfato de Dextrana/toxicidade , Células Epiteliais/metabolismo , Células Epiteliais/efeitos dos fármacos , Biflavonoides/farmacologia , Biflavonoides/uso terapêutico , Masculino , Humanos
2.
J Cell Mol Med ; 28(4): e18139, 2024 02.
Artigo em Inglês | MEDLINE | ID: mdl-38334198

RESUMO

Platelets assume a pivotal role in the cardiovascular diseases (CVDs). Thus, targeting platelet activation is imperative for mitigating CVDs. Ginkgetin (GK), from Ginkgo biloba L, renowned for its anticancer and neuroprotective properties, remains unexplored concerning its impact on platelet activation, particularly in humans. In this investigation, we delved into the intricate mechanisms through which GK influences human platelets. At low concentrations (0.5-1 µM), GK exhibited robust inhibition of collagen and arachidonic acid (AA)-induced platelet aggregation. Intriguingly, thrombin and U46619 remained impervious to GK's influence. GK's modulatory effect extended to ATP release, P-selectin expression, intracellular calcium ([Ca2+ ]i) levels and thromboxane A2 formation. It significantly curtailed the activation of various signaling cascades, encompassing phospholipase Cγ2 (PLCγ2)/protein kinase C (PKC), phosphoinositide 3-kinase/Akt/glycogen synthase kinase-3ß and mitogen-activated protein kinases. GK's antiplatelet effect was not reversed by SQ22536 (an adenylate cyclase inhibitor) or ODQ (a guanylate cyclase inhibitor), and GK had no effect on the phosphorylation of vasodilator-stimulated phosphoproteinSer157 or Ser239 . Moreover, neither cyclic AMP nor cyclic GMP levels were significantly increased after GK treatment. In mouse studies, GK notably extended occlusion time in mesenteric vessels, while sparing bleeding time. In conclusion, GK's profound impact on platelet activation, achieved through inhibiting PLCγ2-PKC cascade, culminates in the suppression of downstream signaling and, ultimately, the inhibition of platelet aggregation. These findings underscore the promising therapeutic potential of GK in the CVDs.


Assuntos
Biflavonoides , Nucleotídeos Cíclicos , Fosfolipases , Humanos , Animais , Camundongos , Nucleotídeos Cíclicos/metabolismo , Nucleotídeos Cíclicos/farmacologia , Fosfolipase C gama/metabolismo , Ácido Araquidônico/farmacologia , Ácido Araquidônico/metabolismo , Fosfolipases/metabolismo , Fosfolipases/farmacologia , Fosfatidilinositol 3-Quinases/metabolismo , Inibidores da Agregação Plaquetária/farmacologia , Ativação Plaquetária , Plaquetas/metabolismo , Agregação Plaquetária , Proteína Quinase C/metabolismo , Fosforilação , Colágeno/metabolismo
3.
Mol Biol Rep ; 51(1): 653, 2024 May 11.
Artigo em Inglês | MEDLINE | ID: mdl-38734766

RESUMO

Prostate cancer is a malignant epithelial tumor of the prostate gland and is the most common malignant tumor of the male genitourinary system. Pharmacological therapies, including chemotherapy and androgen deprivation therapy, play a key role in the treatment of prostate cancer. However, drug resistance and side effects limit the use of these drugs and so there is a need for new drug therapies for prostate cancer patients. Flavonoids, with their wide range of sources and diverse biological activities, have attracted much attention in the field of anti-tumor drug screening. In 2016, at least 58 flavonoids were reported to have anti-prostate cancer activity. In recent years, six additional flavonoid compounds have been found to have anti-prostate cancer potential. In this review, we have collected a large amount of evidence on the anti-prostate cancer effects of these six flavonoids, including a large number of cellular experiments and a small number of preclinical animal experiments. In addition, we predicted their drug-forming properties using Schrödinger's QikProp software and ADMETlab due to the lack of in vivo pharmacokinetic data for the six compounds. In conclusion, this review has fully confirmed the anti-prostate cancer effects of these six flavonoids, summarized their mechanisms of action and predicted their druggability. It provides a reference for the further development of these compounds into anti-prostate cancer drugs.


Assuntos
Flavonoides , Neoplasias da Próstata , Masculino , Flavonoides/farmacologia , Flavonoides/uso terapêutico , Neoplasias da Próstata/tratamento farmacológico , Neoplasias da Próstata/patologia , Humanos , Animais , Antineoplásicos/farmacologia , Antineoplásicos/uso terapêutico
4.
Phytother Res ; 38(3): 1367-1380, 2024 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-38217097

RESUMO

Liver fibrosis affects approximately 800 million patients worldwide, with over 2 million deaths each year. Nevertheless, there are no approved medications for treating liver fibrosis. In this study, we investigated the impacts of ginkgetin on liver fibrosis and the underlying mechanisms. The impacts of ginkgetin on liver fibrosis were assessed in mouse models induced by thioacetamide or bile duct ligation. Experiments on human LX-2 cells and primary mouse hepatic stellate cells (HSCs) were performed to explore the underlying mechanisms, which were also validated in the mouse models. Ginkgetin significantly decreased hepatic extracellular matrix deposition and HSC activation in the fibrotic models induced by thioacetamide (TAA) and bile duct ligation (BDL). Beneficial effects also existed in inhibiting hepatic inflammation and improving liver function. In vitro experiments showed that ginkgetin markedly inhibited HSC viability and induced HSC apoptosis dose-dependently. Mechanistic studies revealed that the antifibrotic effects of ginkgetin depend on STAT1 activation, as the effects were abolished in vitro after STAT1 silencing and in vivo after inhibiting STAT1 activation by fludarabine. Moreover, we observed a meaningful cross-talk between HSCs and hepatocytes, in which IL-6, released by ginkgetin-induced apoptotic HSCs, enhanced hepatocyte proliferation by activating STAT3 signaling. Ginkgetin exhibits antifibrotic effects by inducing HSC apoptosis via STAT1 activation and enhances hepatocyte proliferation secondary to HSC apoptosis via the IL-6/STAT3 pathway.


Assuntos
Biflavonoides , Células Estreladas do Fígado , Tioacetamida , Camundongos , Animais , Humanos , Tioacetamida/metabolismo , Tioacetamida/farmacologia , Tioacetamida/uso terapêutico , Interleucina-6/metabolismo , Cirrose Hepática/tratamento farmacológico , Modelos Animais de Doenças , Apoptose , Fígado/metabolismo , Fator de Transcrição STAT1/metabolismo , Fator de Transcrição STAT1/farmacologia
5.
Immunol Invest ; 52(5): 546-560, 2023 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-37154418

RESUMO

BACKGROUND: Apoptosis, inflammation, and the extracellular matrix (ECM) synthesis and catabolism are compromised with intervertebral disc degeneration (IDD). Ginkgetin (GK) has been demonstrated to alleviate several diseases; however, its effect on IDD remains unknown. METHODS: The nucleus pulposus cells (NPCs) were stimulated with interleukin (IL)-1ß to construct the IDD models in vitro. Rats were used for the construction of the IDD models in vivo via the fibrous ring puncture method. The effect and mechanism of GK on IDD were determined by cell counting kit-8 (CCK-8), flow cytometry, western blot, real-time quantitative polymerase chain reaction (RT-qPCR), enzyme­linked immunosorbent assay (ELISA), hematoxylin and eosin (HE) and safranine O staining, and immunohistochemistry (IHC) assays, respectively. RESULTS: GK increased the cell viability and upregulated the expressions of anti-apoptosis and ECM synthesis markers in NPCs treated with IL-1ß. GK also decreased apoptosis rate, and downregulated the expressions of proteins related to pro-apoptosis, ECM catabolism, and inflammation in vitro. Mechanically, GK reduced the expression of nucleotide binding oligomeric domain like receptor protein 3 (NLRP3) inflammasome-related proteins. Overexpression of NLRP3 reversed the effect of GK on the proliferation, apoptosis, inflammation, and ECM degradation in IL-1ß-induced NPCs. Moreover, GK attenuated the pathological manifestations, inflammation, ECM degradation, and NLRP3 inflammasome expression in IDD rats. CONCLUSION: GK suppressed apoptosis, inflammation, and ECM degradation to alleviate IDD via the inactivation of NLRP3 inflammasome.


Assuntos
Inflamassomos , Degeneração do Disco Intervertebral , Ratos , Animais , Inflamassomos/metabolismo , Degeneração do Disco Intervertebral/tratamento farmacológico , Degeneração do Disco Intervertebral/metabolismo , Degeneração do Disco Intervertebral/patologia , Proteína 3 que Contém Domínio de Pirina da Família NLR/genética , Proteína 3 que Contém Domínio de Pirina da Família NLR/metabolismo , Inflamação/metabolismo , Matriz Extracelular/metabolismo
6.
Neurochem Res ; 47(10): 2963-2974, 2022 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-35593977

RESUMO

Neuroinflammation plays an important role in the pathophysiological process of acute cerebral infarction, which may aggravate brain injury and hinder neuro-repair. Microglia are innate immune cells in the brain. Ginkgetin has anti-inflammatory and neuroprotective effects, but the mechanism remains unclear. This study aims to explore the regulatory effects of ginkgetin on microglia polarization in brain ischemia. Oxygen glucose deprivation (OGD) cellular model and middle cerebral artery occlusion (MCAO) animal model was used in this study. We first observed the dynamic process of microglia polarization in ischemic stroke, and then investigated the effect of ginkgetin treatment on microglia polarization. Finally, we studied the role of PPARγ signaling pathway and the blocking effect of PPARγ antagonist GW9662 in this process. OGD and cerebral ischemia polarized microglia mainly to M1 type. However, ginkgetin treatment converted microglia from M1 type to M2 type, inhibited neuroinflammation, and exerted neuronal protective effects. PPARγ signaling pathway was activated during this process. The above effects could be blocked by GW9662. Ginkgetin can promote M2 polarization of microglia through PPARγ signaling pathway, thereby inhibiting neuroinflammation and promoting recovery of neurological functions in ischemic stroke.


Assuntos
Isquemia Encefálica , AVC Isquêmico , Acidente Vascular Cerebral , Anilidas , Animais , Biflavonoides , Isquemia Encefálica/metabolismo , Infarto da Artéria Cerebral Média/metabolismo , Microglia/metabolismo , Neuroproteção , PPAR gama/metabolismo , Acidente Vascular Cerebral/tratamento farmacológico , Acidente Vascular Cerebral/metabolismo
7.
Int J Mol Sci ; 23(15)2022 Aug 08.
Artigo em Inglês | MEDLINE | ID: mdl-35955943

RESUMO

As an opportunistic pathogen, Escherichia coli (E. coli) forms biofilm that increases the virulence of bacteria and antibiotic resistance, posing a serious threat to human and animal health. Recently, ginkgetin (Gin) has been discovered to have antiinflammatory, antioxidant, and antitumor properties. In the present study, we evaluated the antibiofilm and antibacterial synergist of Gin against E. coli. Additionally, Alamar Blue assay combined with confocal laser scanning microscope (CLSM) and crystal violet (CV) staining was used to evaluate the effect of antibiofilm and antibacterial synergist against E. coli. Results showed that Gin reduces biofilm formation, exopolysaccharide (EPS) production, and motility against E. coli without limiting its growth and metabolic activity. Furthermore, we identified the inhibitory effect of Gin on AI-2 signaling molecule production, which showed apparent anti-quorum sensing (QS) properties. The qRT-PCR also indicated that Gin reduced the transcription of curli-related genes (csgA, csgD), flagella-formation genes (flhC, flhD, fliC, fliM), and QS-related genes (luxS, lsrB, lsrK, lsrR). Moreover, Gin showed obvious antibacterial synergism to overcome antibiotic resistance in E. coli with marketed antibiotics, including gentamicin, colistin B, and colistin E. These results suggested the potent antibiofilm and novel antibacterial synergist effect of Gin for treating E. coli infections.


Assuntos
Infecções por Escherichia coli , Proteínas de Escherichia coli , Animais , Antibacterianos/química , Antibacterianos/farmacologia , Biflavonoides , Biofilmes , Proteínas de Transporte , Colistina/farmacologia , Escherichia coli , Infecções por Escherichia coli/tratamento farmacológico , Proteínas de Escherichia coli/farmacologia , Humanos
8.
Pharmacol Res ; 167: 105525, 2021 05.
Artigo em Inglês | MEDLINE | ID: mdl-33667686

RESUMO

Natural biflavonoids, such as amentoflavone, bilobetin, ginkgetin, isoginkgetin, taiwaniaflavone, morelloflavone, delicaflavone, hinokiflavone, and other derivatives (~ 40 biflavonoids), are isolated from Selaginella sp., Ginkgo biloba, Garcinia sp., and several other species of plants. They are able to exert therapeutic benefits by regulating several proteins/enzymes (PPAR-γ, CCAAT/enhancer-binding protein α [C/EBPα], STAT5, pancreatic lipase, PTP1B, fatty acid synthase, α-glucosidase [AG]) and insulin signaling pathways (via PI3K-AKT), which are linked to metabolism, cell growth, and cell survival mechanisms. Deregulated insulin signaling can cause complications of obesity and diabetes, which can lead to cognitive disorders such as Alzheimer's, Parkinson's, and dementia; therefore, the therapeutic benefits of these biflavones in these areas are highlighted. Since biflavonoids have shown potential to regulate metabolism, growth- and survival-related protein/enzymes, their relation to tumor growth and metastasis of cancer associated with angiogenesis are highlighted. The translational role of biflavones in cancer with respect to the inhibition of metabolism-related processes/pathways, enzymes, or proteins, such as STAT3/SHP-1/PTEN, kinesins, tissue kallikreins, aromatase, estrogen, protein modifiers, antioxidant, autophagy, and apoptosis induction mechanisms, are discussed. Finally, considering their observed bioactivity potential, oral bioavailability studies of biflavones and related clinical trials are outlined.


Assuntos
Antineoplásicos Fitogênicos/uso terapêutico , Biflavonoides/uso terapêutico , Doenças Metabólicas/tratamento farmacológico , Neoplasias/tratamento farmacológico , Animais , Antineoplásicos Fitogênicos/farmacologia , Biflavonoides/farmacologia , Humanos , Doenças Metabólicas/metabolismo , Redes e Vias Metabólicas/efeitos dos fármacos , Neoplasias/metabolismo
9.
J Cell Biochem ; 120(5): 8078-8085, 2019 May.
Artigo em Inglês | MEDLINE | ID: mdl-30582212

RESUMO

AIMS: Acute myocardial infarction (AMI) is one of the most threaten disease in the world. Ginkgetin aglycone (GA) was a new kind of Ginkgo biloba, involved in various diseases, including kidney injury and acute pancreatitis. However, the function of GA in AMI remains unknown. The aim of the study was to investigate the characteristics and function of GA in ischemic-reperfusion injury. METHODS: H2 O 2 - and CoCl 2 -treated H9C2 cells were used to analyze the function of GA in vitro. Caspase 3, interleukin-6 (IL-6), and tumor necrosis factor-α were detected to evaluate the apoptosis and inflammation response. Rat AMI was performed to elucidate the function in vivo. RESULTS: We found that GA could reduce the apoptosis and improved cell survival of H2 O 2 -treated H9C2 cardiomyocytes and CoCl 2 -treated H9C2 cells. GA attenuated CoCl 2 -induced inflammatory response and the level of cleaved caspase 33, suggesting that GA could alleviate the cell apoptosis. GA improved the cardiac function and attenuated the inflammatory cell infiltration in vivo. We also found that nuclear factor-kB signaling pathway, which was activated under hypoxia environment, was also suppressed in the GA-treated group. CONCLUSION: We verified the function and mechanism of GA and provide evidence that GA may play a vital role in ischemic-reperfusion injury, and understanding the precise role of GA will undoubtedly shed new light on the clinical treatment.

10.
J Cell Biochem ; 120(10): 18487-18495, 2019 10.
Artigo em Inglês | MEDLINE | ID: mdl-31265179

RESUMO

BACKGROUND: Ginkgetin, extracted from Ginkgo biloba L leaves, has been demonstrated to have potential anti-inflammatory and immune-suppressive properties. But the neuroprotective effect and potential mechanisms of ginkgetin on cerebral ischemia/reperfusion (IR) injury remain unclear. METHODS: In this research, we studied the neuroprotective effect of ginkgetin in the middle part of the middle cerebral artery occlusion/reperfusion rat model, by analyzing the apoptosis of brain tissues harvested from treatment groups and control groups using the terminal deoxynucleotidyl transferase dUTP nick-end labeling and apoptosis assays. In addition, we detected the association of the neuroprotective effect of ginkgetin with apoptosis inhibition via the activation of the phosphatidylinositol-3-kinase/Akt/mammalian target of rapamycin (PI3K/Akt/mTOR) signaling pathway using Western blot analysis. RESULTS: Our results showed that administration of ginkgetin remarkably reduced brain infarction volumes and neurologic deficits; in addition, reducing apoptotic cell numbers, downregulating the levels of cleaved caspase-3 and Bax, and upregulating the level of Bcl-2 in rats subjected to IR injury in a dose-dependent manner. Moreover, high-dose ginkgetin treatment (100 mg/kg) significantly increased the phosphorylations of Akt and mTOR. Blocking of PI3K by LY294002 clearly decreased its antiapoptotic effect and reduced both Akt and mTOR phosphorylation levels. CONCLUSIONS: Taken together, these results for the first time suggest that ginkgetin antagonizes cerebral IR-induced injury by inhibiting apoptosis in rats, and this effect was attenuated by the activation of PI3K/Akt/mTOR signaling pathway.


Assuntos
Biflavonoides/uso terapêutico , Isquemia Encefálica/tratamento farmacológico , Isquemia Encefálica/metabolismo , Fármacos Neuroprotetores/uso terapêutico , Fosfatidilinositol 3-Quinases/metabolismo , Proteínas Proto-Oncogênicas c-akt/metabolismo , Serina-Treonina Quinases TOR/metabolismo , Animais , Apoptose/efeitos dos fármacos , Western Blotting , Cromonas/farmacologia , Inibidores Enzimáticos/farmacologia , Masculino , Morfolinas/farmacologia , Neurônios/efeitos dos fármacos , Neurônios/metabolismo , Distribuição Aleatória , Ratos , Transdução de Sinais/efeitos dos fármacos
11.
Pharmacol Res ; 139: 325-336, 2019 01.
Artigo em Inglês | MEDLINE | ID: mdl-30472463

RESUMO

Adipogenesis involved in hypertrophy and hyperplasia of adipocytes is responsible for expanding the mass of adipose tissues in obese individuals. Peroxisome proliferator-activated receptor γ (PPARγ) and CCAAT/enhancer-binding protein α (C/EBPα) are two principal transcription factors induced by delicate signaling pathways, including signal transducer and activator of transcription 5 (STAT5), in adipogenesis. Here, we demonstrated a novel role of ginkgetin, a biflavone from Ginkgo biloba leaves, as a STAT5 inhibitor that blocks the differentiation of preadipocytes into adipocytes. During the differentiation of 3T3-L1 cells, ginkgetin treatment during the first 2 days markedly inhibited the formation of lipid-bearing adipocytes. PPARγ and C/EBPα expression was decreased in 3T3-L1 cells during adipogenesis following ginkgetin treatment, whereas no change was observed in C/EBPß or C/EBPδ expression. Inhibition of PPARγ and C/EBPα expression by ginkgetin occurred through the prevention of STAT5 activation during the initiation phase of adipogenesis. In addition, ginkgetin-mediated the inhibition of adipogenesis was recapitulated in the differentiation of primary preadipocytes. Lastly, we confirmed the inhibitory effects of ginkgetin on the hypertrophy of white adipose tissues from high-fat diet-fed mice. These results indicate that ginkgetin is a potential anti-adipogenesis and anti-obesity drug.


Assuntos
Adipogenia/efeitos dos fármacos , Fármacos Antiobesidade/farmacologia , Fármacos Antiobesidade/uso terapêutico , Biflavonoides/farmacologia , Biflavonoides/uso terapêutico , Células 3T3-L1 , Animais , Proteína alfa Estimuladora de Ligação a CCAAT/genética , Proteína alfa Estimuladora de Ligação a CCAAT/metabolismo , Dieta Hiperlipídica , Ginkgo biloba , Masculino , Camundongos , Camundongos Endogâmicos C57BL , PPAR gama/genética , PPAR gama/metabolismo , Folhas de Planta , Transdução de Sinais/efeitos dos fármacos
12.
Cell Mol Biol (Noisy-le-grand) ; 65(4): 79-82, 2019 Apr 30.
Artigo em Inglês | MEDLINE | ID: mdl-31078156

RESUMO

Effect of ginkgetin on proliferation of human cervical cancer (HeLa) cells and the underlying mechanism   were investigated. Human cervical cancer (HeLa) cells were cultured at 37 °C in 10 % fetal bovine serum (FBS) supplemented RPMI 1640 medium in a humidified incubator containing 5 % CO2. Cell proliferation was determined using MTT assay, while real-time quantitative polymerase chain reaction (qRT-PCR) and enzyme-linked immunosorbent assay (ELISA) were used to determine the levels of expression of interleukin 1ß (IL-1ß), tumor necrosis factor-α (TNF-α) and interleukin 8 (IL-8). The expressions of p38 mitogen-activated protein kinases (p38 MAPK) and nuclear factor kappa-light-chain-enhancer of activated B cells (NF- κB) were determined using Western blotting. Treatment of HeLa cells with ginkgetin significantly and time- and dose-dependently inhibited their proliferation (p < 0.05). The invasion of the cells were also significantly and dose-dependently decreased, when compared with control cells (p < 0.05). The expressions of p-p38 and p-NF-κB were significantly and dose-dependently down-regulated, relative to control group (p < 0.05). However, the expressions of p38 and NF-κB in ginkgetin-treated cells were not significantly different from those of control group (p > 0.05). The results of qRT-PCR and ELISA showed that the levels of expression of TNF-α, IL-1ß and IL-8 mRNAs were significantly and dose-dependently reduced in HeLa cells after 48 h of treatment with ginkgetin, when compared with the control group (p < 0.05). The anti-proliferative effect of ginkgetin on HeLa cells is exerted via a mechanism involving the p38/NF-κB pathway.


Assuntos
Biflavonoides/farmacologia , NF-kappa B/metabolismo , Transdução de Sinais/efeitos dos fármacos , Proteínas Quinases p38 Ativadas por Mitógeno/metabolismo , Biflavonoides/química , Proliferação de Células/efeitos dos fármacos , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Células HeLa , Humanos , Interleucina-1beta/metabolismo , Interleucina-8/metabolismo , Invasividade Neoplásica , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Fator de Necrose Tumoral alfa/metabolismo
13.
J Appl Microbiol ; 127(5): 1556-1563, 2019 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-31260158

RESUMO

AIMS: Suilysin (SLY), a crucial virulence-related factor, has multiple cytotoxicities that are regarded as playing a key role in several diseases induced by Streptococcus suis. The aim of this study was to identify an effective inhibitor of SLY and to evaluate the potential inhibitory effect of the inhibitor against S. suis virulence. METHODS AND RESULTS: Antibacterial activity experiments and haemolysis tests were used to identify the SLY inhibitor ginkgetin, and Western blot analysis and oligomerization inhibition tests were employed to determine the potential mechanism for its inhibition effect. The potential inhibitory effect of ginkgetin against S. suis virulence was then assessed through a cytotoxicity test and a mouse infection model. In this study, we demonstrated that the natural ingredient ginkgetin can significantly reduce the haemolytic activity of SLY to protect against S. suis-mediated cell injury in vitro by directly binding to SLY to block the oligomerization of the protein and reducing the bacterial burden in vivo. CONCLUSIONS: The results suggest that ginkgetin can start being used as a potential lead drug for the treatment of S. suis infections. SIGNIFICANCE AND IMPACT OF THE STUDY: The prevention and treatment of S. suis infection might be possible through the targeting of SLY by ginkgetin.


Assuntos
Antibacterianos/administração & dosagem , Biflavonoides/administração & dosagem , Proteínas Hemolisinas/metabolismo , Infecções Estreptocócicas/tratamento farmacológico , Streptococcus suis/efeitos dos fármacos , Fatores de Virulência/metabolismo , Animais , Modelos Animais de Doenças , Feminino , Proteínas Hemolisinas/genética , Humanos , Camundongos , Camundongos Endogâmicos C57BL , Infecções Estreptocócicas/microbiologia , Streptococcus suis/genética , Streptococcus suis/metabolismo , Fatores de Virulência/genética
14.
Biosci Biotechnol Biochem ; 83(4): 675-683, 2019 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-30570395

RESUMO

Ginkgo biloba, a natural biflavonoid isolated from Ginkgo biloba leaves, is reported to have strong anti-inflammatory and immunosuppressive properties. The aim of this study is to investigate the potential anti-inflammatory mechanisms of ginkgo flavonoids on cerebral ischemia/reperfusion (I/R) injury. Inflammatory-associated cytokines in cerebral ischemic hemispheres were determined by immunohistochemical staining, Western blot and enzyme-like immunosorbent assay (ELISA). Our results indicated that treatment with Ginkgetin significantly restored rat brain I/R-induced neurological deficit scores. Inducible nitric oxide synthase (iNOS) and cyclooxygenase-2 (COX-2) expression in Ginkgetin treatment group (100 mg/kg) also significantly reduced. The expression inflammation-related protein prostaglandin E2 (PGE2), tumor necrosis factor alpha (TNF-α), interleukin-1ß (IL-1ß), interleukin-6 (IL-6) and interleukin-8 (IL-8) was also decreased in Ginkgetin treatment group. However, the expression of interleukin-10 (IL-10) was remarkably increased. Thus, this study demonstrates that Ginkgetin protects neurons from I/R-induced rat injury by down-regulating pro-inflammatory cytokines and blocking the TLR4/NF-κB pathway.


Assuntos
Anti-Inflamatórios/farmacologia , Biflavonoides/farmacologia , Isquemia Encefálica/tratamento farmacológico , Regulação da Expressão Gênica/efeitos dos fármacos , Ginkgo biloba/química , Fármacos Neuroprotetores/farmacologia , Traumatismo por Reperfusão/tratamento farmacológico , Animais , Anti-Inflamatórios/isolamento & purificação , Biflavonoides/isolamento & purificação , Isquemia Encefálica/genética , Isquemia Encefálica/imunologia , Isquemia Encefálica/patologia , Ciclo-Oxigenase 2/genética , Ciclo-Oxigenase 2/imunologia , Dinoprostona/genética , Dinoprostona/imunologia , Modelos Animais de Doenças , Esquema de Medicação , Interleucina-10/genética , Interleucina-10/imunologia , Interleucina-1beta/genética , Interleucina-1beta/imunologia , Interleucina-6/genética , Interleucina-6/imunologia , Interleucina-8/genética , Interleucina-8/imunologia , Masculino , NF-kappa B/genética , NF-kappa B/imunologia , Fármacos Neuroprotetores/isolamento & purificação , Óxido Nítrico Sintase Tipo II/genética , Óxido Nítrico Sintase Tipo II/imunologia , Extratos Vegetais/química , Ratos , Ratos Sprague-Dawley , Traumatismo por Reperfusão/genética , Traumatismo por Reperfusão/imunologia , Traumatismo por Reperfusão/patologia , Transdução de Sinais , Receptor 4 Toll-Like/genética , Receptor 4 Toll-Like/imunologia , Fator de Necrose Tumoral alfa/genética , Fator de Necrose Tumoral alfa/imunologia
15.
Z Naturforsch C J Biosci ; 72(11-12): 441-447, 2017 10 26.
Artigo em Inglês | MEDLINE | ID: mdl-28902633

RESUMO

Ginkgetin is known to be an anticancer agent in many studies. However, its effectiveness in treating chronic myeloid leukemia [corrected] remains unknown. The present study aimed to evaluate the effects of ginkgetin on the growth of the K562 cell line. The MTT assay was employed to examine the proliferation of K562, and a terminal deoxynucleotidyl transferase-mediated deoxyuridine triphosphate nick end labeling (TUNEL) staining was conducted to detect the apoptotic rates. Furthermore, changes of tumor necrosis factor-α (TNF-α) were detected by Western blot analysis. Ginkgetin inhibited the proliferation of K562 cells in a dose- and time-dependent manner. Concentrations of ginkgetin required to induce 50% death of K562 at 24, 48 and 72 h were 38.9, 31.3 and 19.2 µM, respectively. Moreover, treatment of ginkgetin increased K562 apoptosis in vitro along with increased levels of TNF-α. Interestingly, anti-TNF-α antibody prevented ginkgetin-induced K562 cell apoptosis and growth inhibition via deactivation of caspase-8, caspase-9 and caspase-3. Concomitantly, downregulation of TNF-α by etanercept in vivo attenuated ginkgetin-induced inhibitory effects on the tumor growth in an xenograft mouse model. Our results indicate that ginkgetin effectively inhibits K562 cell proliferation, and TNF-α plays a key role in ginkgetin-induced cell apoptosis.


Assuntos
Antineoplásicos Fitogênicos/farmacologia , Biflavonoides/farmacologia , Proliferação de Células/efeitos dos fármacos , Regulação Leucêmica da Expressão Gênica , Leucemia Mielogênica Crônica BCR-ABL Positiva/tratamento farmacológico , Fator de Necrose Tumoral alfa/genética , Animais , Anticorpos Neutralizantes/farmacologia , Antineoplásicos Fitogênicos/antagonistas & inibidores , Apoptose/efeitos dos fármacos , Biflavonoides/antagonistas & inibidores , Caspase 3/genética , Caspase 3/metabolismo , Caspase 8/genética , Caspase 8/metabolismo , Caspase 9/genética , Caspase 9/metabolismo , Etanercepte/farmacologia , Humanos , Imunossupressores/farmacologia , Concentração Inibidora 50 , Células K562 , Leucemia Mielogênica Crônica BCR-ABL Positiva/genética , Leucemia Mielogênica Crônica BCR-ABL Positiva/metabolismo , Leucemia Mielogênica Crônica BCR-ABL Positiva/patologia , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Nus , Transdução de Sinais , Fator de Necrose Tumoral alfa/antagonistas & inibidores , Fator de Necrose Tumoral alfa/metabolismo , Ensaios Antitumorais Modelo de Xenoenxerto
16.
Phytother Res ; 30(4): 567-76, 2016 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-27059688

RESUMO

Ginkgetin, a biflavone from Ginkgo biloba leaves, is known to exhibit antiinflammatory, antifungal, neuroprotective, and antitumor activities, but its precise mechanism of action has not been fully elucidated. Because the aberrant activation of STAT3 has been linked with regulation of inflammation, proliferation, invasion, and metastasis of tumors, we hypothesized that ginkgetin modulates the activation of STAT3 in tumor cells. We found that ginkgetin clearly suppressed constitutive phosphorylation of STAT3 through inhibition of the activation of upstream JAK1 and c-Src kinases and nuclear translocation of STAT3 on both A549 and FaDu cells. Treatment with sodium pervanadate reversed the ginkgetin-induced down-modulation of STAT3, thereby indicating a critical role for a PTP. We also found that ginkgetin strongly induced the expression of the SHP-1 and PTEN proteins and its mRNAs. Further, deletion of SHP-1 and PTEN genes by siRNA suppressed the induction of SHP-1 and PTEN, and reversed the inhibition of STAT3 activation. Ginkgetin induced apoptosis as characterized by an increased accumulation of cells in subG1 phase, positive Annexin V binding, loss of mitochondrial membrane potential, down-regulation of STAT3-regulated gene products, and cleavage of PARP. Overall, ginkgetin abrogates STAT3 signaling pathway through induction of SHP-1 and PTEN proteins, thus attenuating STAT3 phosphorylation and tumorigenesis.


Assuntos
Apoptose/efeitos dos fármacos , Biflavonoides/farmacologia , PTEN Fosfo-Hidrolase/metabolismo , Proteína Tirosina Fosfatase não Receptora Tipo 6/metabolismo , Fator de Transcrição STAT3/antagonistas & inibidores , Proteína Tirosina Quinase CSK , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Regulação para Baixo , Humanos , Janus Quinase 1/metabolismo , Fosforilação , Fator de Transcrição STAT3/metabolismo , Transdução de Sinais/efeitos dos fármacos , Quinases da Família src/metabolismo
17.
Cancer Sci ; 106(4): 413-20, 2015 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-25611086

RESUMO

Signal transducer and activator of transcription 3 (STAT3) is constitutively activated in human cancers. Therefore, STAT3 is a therapeutic target of cancer drug discovery. We previously reported that natural products inhibited constitutively activated STAT3 in human prostate tumor cells. We used a dual-luciferase assay to screen 200 natural products isolated from herbal medicines and we identified ginkgetin obtained from the leaves of Ginkgo biloba L. as a STAT3 inhibitor. Ginkgetin inhibited both inducible and constitutively activated STAT3 and blocked the nuclear translocation of p-STAT3 in DU-145 prostate cancer cells. Furthermore, ginkgetin selectively inhibited the growth of prostate tumor cells stimulated with activated STAT3. Ginkgetin induced STAT3 dephosphorylation at Try705 and inhibited its localization to the nucleus, leading to the inhibition of expression of STAT3 target genes such as cell survival-related genes (cyclin D1 and survivin) and anti-apoptotic proteins (Bcl-2 and Bcl-xL). Therefore, ginkgetin inhibited the growth of STAT3-activated tumor cells. We also found that ginkgetin inhibited tumor growth in xenografted nude mice and downregulated p-STAT3(Tyr705) and survivin in tumor tissues. This is the first report that ginkgetin exerts antitumor activity by inhibiting STAT3. Therefore, ginkgetin is a good STAT3 inhibitor and may be a useful lead molecule for development of a therapeutic STAT3 inhibitor.


Assuntos
Antineoplásicos/uso terapêutico , Biflavonoides/uso terapêutico , Neoplasias da Próstata/tratamento farmacológico , Fator de Transcrição STAT3/antagonistas & inibidores , Transporte Ativo do Núcleo Celular/efeitos dos fármacos , Animais , Apoptose/efeitos dos fármacos , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Ciclina D1/biossíntese , Ensaios de Seleção de Medicamentos Antitumorais , Feminino , Pontos de Checagem da Fase G1 do Ciclo Celular/efeitos dos fármacos , Ginkgo biloba/metabolismo , Células HCT116 , Humanos , Proteínas Inibidoras de Apoptose/biossíntese , Interleucina-6/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Nus , Fosforilação/efeitos dos fármacos , Próstata/patologia , Neoplasias da Próstata/patologia , Proteínas Proto-Oncogênicas c-bcl-2/biossíntese , Proteínas Repressoras/biossíntese , Fator de Transcrição STAT3/biossíntese , Survivina , Ensaios Antitumorais Modelo de Xenoenxerto
18.
PeerJ ; 12: e17722, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-39006031

RESUMO

Background: Osteoporosis is a disease associated with bone resorption, characterized primarily by the excessive activation of osteoclasts. Ginkgetin is a compound purified from natural ginkgo leaves which has various biological properties, including anti-inflammation, antioxidant, and anti-tumor effects. This study investigated the bone-protective effects of ginkgetin in ovariectomized (OVX) mice and explored their potential signaling pathway in inhibiting osteoclastogenesis in a mouse model of osteoporosis. Methods: Biochemical assays were performed to assess the levels of Ca, ALP, and P in the blood. Micro CT scanning was used to evaluate the impact of ginkgetin on bone loss in mice. RT-PCR was employed to detect the expression of osteoclast-related genes (ctsk, c-fos, trap) in their femoral tissue. Hematoxylin and eosin (H&E) staining was utilized to assess the histopathological changes in femoral tissue due to ginkgetin. The TRAP staining was used to evaluate the impact of ginkgetin osteoclast generation in vivo. Western blot analysis was conducted to investigate the effect of ginkgetin on the expression of p-NF-κB p65 and IκBα proteins in mice. Results: Our findings indicate that ginkgetin may increase the serum levels of ALP and P, while decreasing the serum level of Ca in OVX mice. H&E staining and micro CT scanning results suggest that ginkgetin can inhibit bone loss in OVX mice. The TRAP staining results showed ginkgetin suppresses the generation of osteoclasts in OVX mice. RT-PCR results demonstrate that ginkgetin downregulate the expression of osteoclast-related genes (ctsk, c-fos, trap) in the femoral tissue of mice, and this effect is dose-dependent. Western blot analysis results reveal that ginkgetin can inhibit the expression of p-NF-κB p65 and IκBα proteins in mice. Conclusion: Ginkgetin can impact osteoclast formation and activation in OVX mice by inhibiting the NF-κB/IκBα signaling pathway, thereby attenuating bone loss in mice.


Assuntos
Biflavonoides , NF-kappa B , Osteoclastos , Transdução de Sinais , Animais , Biflavonoides/farmacologia , Biflavonoides/uso terapêutico , Transdução de Sinais/efeitos dos fármacos , Camundongos , NF-kappa B/metabolismo , Feminino , Osteoclastos/efeitos dos fármacos , Osteoclastos/metabolismo , Osteoporose/tratamento farmacológico , Osteoporose/metabolismo , Osteoporose/patologia , Ovariectomia , Modelos Animais de Doenças , Reabsorção Óssea/tratamento farmacológico , Reabsorção Óssea/metabolismo , Reabsorção Óssea/prevenção & controle , Reabsorção Óssea/patologia , Microtomografia por Raio-X , Inibidor de NF-kappaB alfa/metabolismo , Camundongos Endogâmicos C57BL
19.
J Ethnopharmacol ; 331: 118284, 2024 Sep 15.
Artigo em Inglês | MEDLINE | ID: mdl-38735420

RESUMO

ETHNOPHARMACOLOGICAL RELEVANCE: Ginkgo biloba leaf and seed have been traditionally used in ancient China for the treatment of cough and asthma. However, there is limited literature available on the anti-COPD effects and mechanisms of Ginkgo biloba. AIMS OF THE STUDY: The aim of this study was to comprehensively investigate the therapeutic potential of ginkgo extracts in COPD through a combination of in vivo and in vitro functional experiments. Transcriptomic analyses were also employed to uncover novel molecular mechanisms underlying the therapeutic effects of ginkgetin in COPD. MATERIALS AND METHODS: The therapeutic efficacy of ginkgo extracts was assessed in a COPD model. The anti-inflammatory effects of ginkgetin and its underlying molecular mechanisms were examined in A549 cells treated with cigarette smoke extract (CSE). Additionally, transcriptomic analyses were conducted to identify novel molecular pathways influenced by ginkgetin. These findings were further validated using quantitative real-time polymerase chain reaction (qPCR) and Western blot techniques. RESULTS: The ethyl acetate extract of Ginkgo biloba L. seeds and ginkgetin treatment significantly reduced cytokine production in COPD mice. Following drug administration, lung function improved in different groups. The transcriptome data strongly supports the inhibitory effect of ginkgetin on CSE-induced inflammation through the downregulation of the c/EBPß signaling pathway and subsequent inhibition of CCL2 expression. CONCLUSION: Our results demonstrate that ginkgetin, one of the biflavones found in Ginkgo biloba, exhibits inhibitory effects on smoke-induced airway inflammation. This effect is achieved through the downregulation of the c/EBPß signaling pathway and the reduction of CCL2 expression.


Assuntos
Biflavonoides , Quimiocina CCL2 , Regulação para Baixo , Ginkgo biloba , Doença Pulmonar Obstrutiva Crônica , Transdução de Sinais , Animais , Doença Pulmonar Obstrutiva Crônica/tratamento farmacológico , Biflavonoides/farmacologia , Biflavonoides/uso terapêutico , Humanos , Transdução de Sinais/efeitos dos fármacos , Ginkgo biloba/química , Regulação para Baixo/efeitos dos fármacos , Quimiocina CCL2/genética , Quimiocina CCL2/metabolismo , Camundongos , Masculino , Extratos Vegetais/farmacologia , Extratos Vegetais/uso terapêutico , Fumaça/efeitos adversos , Anti-Inflamatórios/farmacologia , Anti-Inflamatórios/uso terapêutico , Células A549 , Camundongos Endogâmicos C57BL , Modelos Animais de Doenças , Pulmão/efeitos dos fármacos , Pulmão/metabolismo , Pulmão/patologia , Extrato de Ginkgo
20.
Environ Sci Pollut Res Int ; 31(6): 9031-9044, 2024 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-38182957

RESUMO

Polystyrene microplastics (PSMPs) have emerged as a ubiquitous environmental toxicant that affects different organs including testes. Ginkgetin (GNG) is a biflavonoid that shows antioxidant properties. The current research was undertaken to evaluate the ameliorative potential of GNG against PSMPs-instigated testicular damages. Forty-eight albino rats (male) were randomly divided into 4 equal groups: control, PSMPs-treated group (0.01 mgkg-1), GNG + PSMPs-exposed group (25 mgkg-1 + 0.01 mgkg-1), and only GNG-supplemented group (25 mgkg-1). After 56 days of treatment, it was revealed that PSMPs significantly reduced the activity of glutathione peroxidase (GPx), catalase (CAT), superoxide dismutase (SOD), and glutathione reductase (GSR), while concurrently augmented the levels of lipid peroxidation marker, i.e., malondialdehyde (MDA) along with reactive oxygen species (ROS). Rats administered with PSMPs showed a significant reduction in the spermatogenic indices (sperm count, viability, and motility), HOS coiled tail sperm along with increased sperm structural deformities, i.e., tail, head, and mid-piece. Additionally, PSMPs exposure decreased the levels of testosterone, luteinizing (LH), and follicle-stimulating hormones (FSH). Besides, administration of PSMPs reduced the steroidogenic enzymes (13ß-HSD, StAR, and 17ß-HSD) and Bcl-2 expression, while augmented the caspase-3 and Bax expression. PSMPs also elevated the levels of inflammatory markers (IL-6, IL-1ß, TNF-α, and NF-κB) and activity of COX-2 in the testes. Furthermore, PSMPs treatment induced various histopathological damages in the testes of rats. Therefore, findings of the current study suggested that GNG effectively mitigated the PSMPs-induced testicular toxicity owing to its chemoprotective potential possibly through its anti-inflammatory, antioxidant, anti-apoptotic, and androgenic properties.


Assuntos
Biflavonoides , Testículo , Ratos , Masculino , Animais , Antioxidantes/metabolismo , Biflavonoides/análise , Biflavonoides/metabolismo , Biflavonoides/farmacologia , Microplásticos/análise , Plásticos/análise , Poliestirenos/análise , Estresse Oxidativo , Ratos Wistar , Sêmen/metabolismo , Testosterona/metabolismo
SELEÇÃO DE REFERÊNCIAS
Detalhe da pesquisa