BACKGROUND Fanlian Huazhuo Formula(FLHZF)has the functions of invigorating spleen and resolving phlegm,clearing heat and purging turbidity.It has been identified to have therapeutic effects on type 2 diabetes mellitus...BACKGROUND Fanlian Huazhuo Formula(FLHZF)has the functions of invigorating spleen and resolving phlegm,clearing heat and purging turbidity.It has been identified to have therapeutic effects on type 2 diabetes mellitus(T2DM)in clinical application.Non-alcoholic fatty liver disease(NAFLD)is frequently diagnosed in patients with T2DM.However,the therapeutic potential of FLHZF on NAFLD and the underlying mechanisms need further investigation.AIM To elucidate the effects of FLHZF on NAFLD and explore the underlying hepatoprotective mechanisms in vivo and in vitro.METHODS HepG2 cells were treated with free fatty acid for 24 hours to induce lipid accumulation cell model.Subsequently,experiments were conducted with the different concentrations of freeze-dried powder of FLHZF for 24 hours.C57BL/6 mice were fed a high-fat diet for 8-week to establish a mouse model of NAFLD,and then treated with the different concentrations of FLHZF for 10 weeks.RESULTS FLHZF had therapeutic potential against lipid accumulation and abnormal changes in biochemical indicators in vivo and in vitro.Further experiments verified that FLHZF alleviated abnormal lipid metabolism might by reducing oxidative stress,regulating the AMPKα/SREBP-1C signaling pathway,activating autophagy,and inhibiting hepatocyte apoptosis.CONCLUSION FLHZF alleviates abnormal lipid metabolism in NAFLD models by regulating reactive oxygen species,autophagy,apoptosis,and lipid synthesis signaling pathways,indicating its potential for clinical application in NAFLD.展开更多
OBJECTIVE Mu-Xiang-You-Fang(MXYF)is a classic prescription of Hui medicine,composed of five herbs,which has been used to treat ischemic stroke for many years.However,the potential pharmacological mecha⁃nisms of MXYF r...OBJECTIVE Mu-Xiang-You-Fang(MXYF)is a classic prescription of Hui medicine,composed of five herbs,which has been used to treat ischemic stroke for many years.However,the potential pharmacological mecha⁃nisms of MXYF remain unclear.The present research is to investigate the neuroprotective effect of MXYF and its role in modulating autophagy via AMPK/mTOR signaling pathway in the PC12 oxygen-glucose deprivation and reperfusion(OGD/R)injury model.METHODS MXYF was extracted by supercritical CO2 fluid extraction apparatus.PC12 OGD/R injury model was established by oxygen-glucose deprivation for 2 h and reperfusion for 24 h.The effects of MXYF on the viability and cytotoxicity of PC12 cells were determined through cell counting kit(CCK-8)assay.Colorimetric method was performed to determine the LDH leakage rate.The calcium concentration was determined by chemical fluorescence method and the mitochondrial membrane potential was determined through flow cytometry.Monodansylcadaverine(MDC)staining was conducted to detect autophagosome formation.The expression of LC3,Beclin1,p62,p-AMPK,ULK1,p-mTOR and p-p70s6k proteins were determined by immunofluorescence and Western blotting analyses.RESULTS MXYF(1,2 and 4 mg·L^-1)could significantly increase the cell viability and mitochondrial membrane potential,while decreased the release of lactate dehydrogenase(LDH)and calcium concentration in PC12 cells.Mechanistic studies showed that MXYF reduced the LC3-II/LC3-I ratio and inhibited the expression of beclin1,p-AMPK and ULK1.In comparison,the expres⁃sion of p-mTOR,p-p70s6k and p62 were significantly enhanced.CONCLUSION MXYF inhibits autophagy after OGD/Rinduced PC12 cell injury through AMPK-mTOR pathway,thus MXYF might have therapeutic potential for treating the ischemic stroke.展开更多
Puerarin suppresses autophagy to alleviate cerebral ischemia/reperfusion injury, and accumulating evidence indicates that the AMPKm TOR signaling pathway regulates the activation of the autophagy pathway through the c...Puerarin suppresses autophagy to alleviate cerebral ischemia/reperfusion injury, and accumulating evidence indicates that the AMPKm TOR signaling pathway regulates the activation of the autophagy pathway through the coordinated phosphorylation of ULK1. In this study, we investigated the mechanisms underlying the neuroprotective effect of puerarin and its role in modulating autophagy via the AMPK-m TOR-ULK1 signaling pathway in the rat middle cerebral artery occlusion model of cerebral ischemia/reperfusion injury. Rats were intraperitoneally injected with puerarin, 50 or 100 mg/kg, daily for 7 days. Then, 30 minutes after the final administration, rats were subjected to transient middle cerebral artery occlusion for 90 minutes. Then, after 24 hours of reperfusion, the Longa score and infarct volume were evaluated in each group. Autophagosome formation was observed by transmission electron microscopy. LC3, Beclin-1 p62, AMPK, m TOR and ULK1 protein expression levels were examined by immunofluorescence and western blot assay. Puerarin substantially reduced the Longa score and infarct volume, and it lessened autophagosome formation in the hippocampal CA1 area following cerebral ischemia/reperfusion injury in a dose-dependent manner. Pretreatment with puerarin(50 or 100 mg/kg) reduced Beclin-1 expression and the LC3-II/LC3-I ratio, as well as p-AMPK and p S317-ULK1 levels. In comparison, it increased p62 expression. Furthermore, puerarin at 100 mg/kg dramatically increased the levels of p-m TOR and p S757-ULK1 in the hippocampus on the ischemic side. Our findings suggest that puerarin alleviates autophagy by activating the APMK-m TOR-ULK1 signaling pathway. Thus, puerarin might have therapeutic potential for treating cerebral ischemia/reperfusion injury.展开更多
Objective Pituitary adrenocorticotropic hormone(ACTH)-secreting adenoma is a relatively intractable endocrine adenoma that can cause a range of severe metabolic disorders and pathological changes involving multiple sy...Objective Pituitary adrenocorticotropic hormone(ACTH)-secreting adenoma is a relatively intractable endocrine adenoma that can cause a range of severe metabolic disorders and pathological changes involving multiple systems.Previous studies have shown that celastrol has antitumor effects on a variety of tumor cells via the AKT/mTOR signaling.However,whether celastrol has pronounced antitumor effects on pituitary ACTH-secreting adenoma is unclear.This study aimed to identify a new effective therapeutic drug for pituitary ACTH-secreting adenoma.Methods Mouse pituitary ACTH-secreting adenoma cells(AtT20 cells)were used as an experimental model in vitro and to establish a xenograft tumor model in mice.Cells and animals were administered doses of celastrol at various levels.The effects of celastrol on cell viability,migration,apoptosis and autophagy were then examined.Finally,the potential involvement of AKT/mTOR signaling in celastrol’s mechanism was assessed.Results Celastrol inhibited the proliferation and migration of pituitary adenoma cells in a time-and concentration-dependent manner.It blocked AtT20 cells in the G0/G1 phase,and induced apoptosis and autophagy by downregulating the AKT/mTOR signaling pathway.Similar results were obtained in mice.Conclusion Celastrol exerts potent antitumor effects on ACTH-secreting adenoma by downregulating the AKT/mTOR signaling in vitro and in vivo.展开更多
Wilson disease(WD),known as hepatolenticular degeneration(HLD),is a treatable autosomal recessive disorder of copper metabolism.Because copper deposits in the liver first,the liver is not only the original defective o...Wilson disease(WD),known as hepatolenticular degeneration(HLD),is a treatable autosomal recessive disorder of copper metabolism.Because copper deposits in the liver first,the liver is not only the original defective organ but also the most affected organ.The liver injury is also one of the main causes of death throughout the course of the disease.Therefore,the treatment of liver injury is the main task of WD treatment,which is of great significance to improve the prognosis of patients.Autophagy is a process that promotes cell survival through degradation,recycling,and absorption in order to maintain the normal physiological function of cells,while excessive autophagy can aggravate cell death.In view of the abnormal damage of liver cells in patients with WD,which may be related to the change of autophagy level,in this study,we established an animal model of WD through toxic milk(TX)mice,observed the change of autophagy level in the liver,and observed the change of liver damage in mice after treatment with autophagy inhibitors.It was found that the mTOR signaling pathway was activated and autophagy was inhibited in Wilson mouse liver.After treatment with rapamycin,the autophagy level of mice liver was upregulated,and the copper content of mice liver was reduced,and the damage was alleviated.TX mouse hepatocytes were isolated,after using siRNA to interfere with mTOR expression,the copper accumulation was significantly reduced,which was the same with RAPA treatment.The results showed that in TX mice,the damage caused by copper accumulation in the liver may be related to the decrease of autophagy level caused by the activation of the mTOR signaling pathway.Our findings suggested that RAPA or the use of siRNA targeting mTOR may have potential applications in the treatment of Wilson’s disease.展开更多
BACKGROUND Altered miR-188-3p expression has been observed in various human cancers.AIM To investigate the miR-188-3p expression,its roles,and underlying molecular events in gastric cancer.METHODS Fifty gastric cancer...BACKGROUND Altered miR-188-3p expression has been observed in various human cancers.AIM To investigate the miR-188-3p expression,its roles,and underlying molecular events in gastric cancer.METHODS Fifty gastric cancer and paired normal tissues were collected to analyze miR-188-3p and CBL expression.Normal and gastric cancer cells were used to manipulate miR-188-3p and CBL expression through different assays.The relationship between miR-188-3p and CBL was predicted bioinformatically and confirmed using a luciferase gene reporter assay.A Kaplan-Meier analysis was used to associate miR-188-3p or CBL expression with patient survival.A nude mouse tumor cell xenograft assay was used to confirm the in vitro data.RESULTS MiR-188-3p was found to be lower in the plasma of gastric cancer patients,tissues,and cell lines compared to their healthy counterparts.It was associated with overall survival of gastric cancer patients(P<0.001),tumor differentiation(P<0.001),lymph node metastasis(P=0.033),tumor node metastasis stage(I/II vs III/IV,P=0.024),and American Joint Committee on Cancer stage(I/II vs III/IV,P=0.03).Transfection with miR-188-3p mimics reduced tumor cell growth and invasion while inducing apoptosis and autophagy.CBL was identified as a direct target of miR-188-3p,with its expression antagonizing the effects of miR-188-3p on gastric cancer(GC)cell proliferation by inducing tumor cell apoptosis and autophagy through the inactivation of the Akt/mTOR signaling pathway.The in vivo data confirmed antitumor activity via CBL downregulation in gastric cancer.CONCLUSION The current data provides ex vivo,in vitro,and in vivo evidence that miR-188-3p acts as a tumor suppressor gene or possesses antitumor activity in GC.展开更多
基金Supported by Basic and Applied Basic Research Found of Guangdong Province,No.2022A1515011307。
文摘BACKGROUND Fanlian Huazhuo Formula(FLHZF)has the functions of invigorating spleen and resolving phlegm,clearing heat and purging turbidity.It has been identified to have therapeutic effects on type 2 diabetes mellitus(T2DM)in clinical application.Non-alcoholic fatty liver disease(NAFLD)is frequently diagnosed in patients with T2DM.However,the therapeutic potential of FLHZF on NAFLD and the underlying mechanisms need further investigation.AIM To elucidate the effects of FLHZF on NAFLD and explore the underlying hepatoprotective mechanisms in vivo and in vitro.METHODS HepG2 cells were treated with free fatty acid for 24 hours to induce lipid accumulation cell model.Subsequently,experiments were conducted with the different concentrations of freeze-dried powder of FLHZF for 24 hours.C57BL/6 mice were fed a high-fat diet for 8-week to establish a mouse model of NAFLD,and then treated with the different concentrations of FLHZF for 10 weeks.RESULTS FLHZF had therapeutic potential against lipid accumulation and abnormal changes in biochemical indicators in vivo and in vitro.Further experiments verified that FLHZF alleviated abnormal lipid metabolism might by reducing oxidative stress,regulating the AMPKα/SREBP-1C signaling pathway,activating autophagy,and inhibiting hepatocyte apoptosis.CONCLUSION FLHZF alleviates abnormal lipid metabolism in NAFLD models by regulating reactive oxygen species,autophagy,apoptosis,and lipid synthesis signaling pathways,indicating its potential for clinical application in NAFLD.
基金National Natural Science Foundation of China(8166070081260679)Ningxia College FirstClass Discipline Construction Project(Chinese Medicine)Funded Project(NXYLXK2017A06)
文摘OBJECTIVE Mu-Xiang-You-Fang(MXYF)is a classic prescription of Hui medicine,composed of five herbs,which has been used to treat ischemic stroke for many years.However,the potential pharmacological mecha⁃nisms of MXYF remain unclear.The present research is to investigate the neuroprotective effect of MXYF and its role in modulating autophagy via AMPK/mTOR signaling pathway in the PC12 oxygen-glucose deprivation and reperfusion(OGD/R)injury model.METHODS MXYF was extracted by supercritical CO2 fluid extraction apparatus.PC12 OGD/R injury model was established by oxygen-glucose deprivation for 2 h and reperfusion for 24 h.The effects of MXYF on the viability and cytotoxicity of PC12 cells were determined through cell counting kit(CCK-8)assay.Colorimetric method was performed to determine the LDH leakage rate.The calcium concentration was determined by chemical fluorescence method and the mitochondrial membrane potential was determined through flow cytometry.Monodansylcadaverine(MDC)staining was conducted to detect autophagosome formation.The expression of LC3,Beclin1,p62,p-AMPK,ULK1,p-mTOR and p-p70s6k proteins were determined by immunofluorescence and Western blotting analyses.RESULTS MXYF(1,2 and 4 mg·L^-1)could significantly increase the cell viability and mitochondrial membrane potential,while decreased the release of lactate dehydrogenase(LDH)and calcium concentration in PC12 cells.Mechanistic studies showed that MXYF reduced the LC3-II/LC3-I ratio and inhibited the expression of beclin1,p-AMPK and ULK1.In comparison,the expres⁃sion of p-mTOR,p-p70s6k and p62 were significantly enhanced.CONCLUSION MXYF inhibits autophagy after OGD/Rinduced PC12 cell injury through AMPK-mTOR pathway,thus MXYF might have therapeutic potential for treating the ischemic stroke.
基金supported by the National Natural Science Foundation of China,No.81202625the Open Fund of Key Laboratory of Cardiovascular and Cerebrovascular Diseases Translational Medicine,China Three Gorges University,China,No.2016xnxg101
文摘Puerarin suppresses autophagy to alleviate cerebral ischemia/reperfusion injury, and accumulating evidence indicates that the AMPKm TOR signaling pathway regulates the activation of the autophagy pathway through the coordinated phosphorylation of ULK1. In this study, we investigated the mechanisms underlying the neuroprotective effect of puerarin and its role in modulating autophagy via the AMPK-m TOR-ULK1 signaling pathway in the rat middle cerebral artery occlusion model of cerebral ischemia/reperfusion injury. Rats were intraperitoneally injected with puerarin, 50 or 100 mg/kg, daily for 7 days. Then, 30 minutes after the final administration, rats were subjected to transient middle cerebral artery occlusion for 90 minutes. Then, after 24 hours of reperfusion, the Longa score and infarct volume were evaluated in each group. Autophagosome formation was observed by transmission electron microscopy. LC3, Beclin-1 p62, AMPK, m TOR and ULK1 protein expression levels were examined by immunofluorescence and western blot assay. Puerarin substantially reduced the Longa score and infarct volume, and it lessened autophagosome formation in the hippocampal CA1 area following cerebral ischemia/reperfusion injury in a dose-dependent manner. Pretreatment with puerarin(50 or 100 mg/kg) reduced Beclin-1 expression and the LC3-II/LC3-I ratio, as well as p-AMPK and p S317-ULK1 levels. In comparison, it increased p62 expression. Furthermore, puerarin at 100 mg/kg dramatically increased the levels of p-m TOR and p S757-ULK1 in the hippocampus on the ischemic side. Our findings suggest that puerarin alleviates autophagy by activating the APMK-m TOR-ULK1 signaling pathway. Thus, puerarin might have therapeutic potential for treating cerebral ischemia/reperfusion injury.
基金This work was supported by the National Natural Science Youth Foundation of China(No.81602204).
文摘Objective Pituitary adrenocorticotropic hormone(ACTH)-secreting adenoma is a relatively intractable endocrine adenoma that can cause a range of severe metabolic disorders and pathological changes involving multiple systems.Previous studies have shown that celastrol has antitumor effects on a variety of tumor cells via the AKT/mTOR signaling.However,whether celastrol has pronounced antitumor effects on pituitary ACTH-secreting adenoma is unclear.This study aimed to identify a new effective therapeutic drug for pituitary ACTH-secreting adenoma.Methods Mouse pituitary ACTH-secreting adenoma cells(AtT20 cells)were used as an experimental model in vitro and to establish a xenograft tumor model in mice.Cells and animals were administered doses of celastrol at various levels.The effects of celastrol on cell viability,migration,apoptosis and autophagy were then examined.Finally,the potential involvement of AKT/mTOR signaling in celastrol’s mechanism was assessed.Results Celastrol inhibited the proliferation and migration of pituitary adenoma cells in a time-and concentration-dependent manner.It blocked AtT20 cells in the G0/G1 phase,and induced apoptosis and autophagy by downregulating the AKT/mTOR signaling pathway.Similar results were obtained in mice.Conclusion Celastrol exerts potent antitumor effects on ACTH-secreting adenoma by downregulating the AKT/mTOR signaling in vitro and in vivo.
基金supported by Natural Science Foundation of Anhui Province(1908085MH266)National Natural Science Foundation of China(81673948).
文摘Wilson disease(WD),known as hepatolenticular degeneration(HLD),is a treatable autosomal recessive disorder of copper metabolism.Because copper deposits in the liver first,the liver is not only the original defective organ but also the most affected organ.The liver injury is also one of the main causes of death throughout the course of the disease.Therefore,the treatment of liver injury is the main task of WD treatment,which is of great significance to improve the prognosis of patients.Autophagy is a process that promotes cell survival through degradation,recycling,and absorption in order to maintain the normal physiological function of cells,while excessive autophagy can aggravate cell death.In view of the abnormal damage of liver cells in patients with WD,which may be related to the change of autophagy level,in this study,we established an animal model of WD through toxic milk(TX)mice,observed the change of autophagy level in the liver,and observed the change of liver damage in mice after treatment with autophagy inhibitors.It was found that the mTOR signaling pathway was activated and autophagy was inhibited in Wilson mouse liver.After treatment with rapamycin,the autophagy level of mice liver was upregulated,and the copper content of mice liver was reduced,and the damage was alleviated.TX mouse hepatocytes were isolated,after using siRNA to interfere with mTOR expression,the copper accumulation was significantly reduced,which was the same with RAPA treatment.The results showed that in TX mice,the damage caused by copper accumulation in the liver may be related to the decrease of autophagy level caused by the activation of the mTOR signaling pathway.Our findings suggested that RAPA or the use of siRNA targeting mTOR may have potential applications in the treatment of Wilson’s disease.
基金Supported by the National Natural Science Funds of China,No.81974448Guangdong Medical Research Foundation,No.B2019126Shenzhen Science and Technology Innovation Commission,No.JCYJ20210324135005013.
文摘BACKGROUND Altered miR-188-3p expression has been observed in various human cancers.AIM To investigate the miR-188-3p expression,its roles,and underlying molecular events in gastric cancer.METHODS Fifty gastric cancer and paired normal tissues were collected to analyze miR-188-3p and CBL expression.Normal and gastric cancer cells were used to manipulate miR-188-3p and CBL expression through different assays.The relationship between miR-188-3p and CBL was predicted bioinformatically and confirmed using a luciferase gene reporter assay.A Kaplan-Meier analysis was used to associate miR-188-3p or CBL expression with patient survival.A nude mouse tumor cell xenograft assay was used to confirm the in vitro data.RESULTS MiR-188-3p was found to be lower in the plasma of gastric cancer patients,tissues,and cell lines compared to their healthy counterparts.It was associated with overall survival of gastric cancer patients(P<0.001),tumor differentiation(P<0.001),lymph node metastasis(P=0.033),tumor node metastasis stage(I/II vs III/IV,P=0.024),and American Joint Committee on Cancer stage(I/II vs III/IV,P=0.03).Transfection with miR-188-3p mimics reduced tumor cell growth and invasion while inducing apoptosis and autophagy.CBL was identified as a direct target of miR-188-3p,with its expression antagonizing the effects of miR-188-3p on gastric cancer(GC)cell proliferation by inducing tumor cell apoptosis and autophagy through the inactivation of the Akt/mTOR signaling pathway.The in vivo data confirmed antitumor activity via CBL downregulation in gastric cancer.CONCLUSION The current data provides ex vivo,in vitro,and in vivo evidence that miR-188-3p acts as a tumor suppressor gene or possesses antitumor activity in GC.