This study aimed to investigate the effect and mechanisms of Ephedra Herb(EH)extract on adriamycin-induced nephrotic syndrome(NS),providing an experimental basis for the clinical treatment of NS.Hematoxylin and eosin ...This study aimed to investigate the effect and mechanisms of Ephedra Herb(EH)extract on adriamycin-induced nephrotic syndrome(NS),providing an experimental basis for the clinical treatment of NS.Hematoxylin and eosin staining,creatinine,urea nitrogen,and kidn injury molecule-1 were used to evaluate the activities of EH extract on renal function.The levels of inflammatory factors and oxidative stress were detected by kits.The levels of reactive oxygen species,immune cells,and apoptosis were measured by flow cytometry.A network pharmacological approach was used to predict the potential targets and mechanisms of EH extract in the treatment of NS.The protein levels of apoptosis-related proteins and CAMKK2,p-CAMKK2,AMPK,p-AMPK,mTOR and p-mTOR in the kidneys were detected by Western blot.The effective material basis of EH extract was screened by MTT assay.The AMPK pathway inhibitor(compound C,CC)was added to investigate the effect of the potent material basis on adriamycin-induced cell injury.EH extract significantly improved renal injury and relieve inflammation,oxidative stress,and apoptosis in rats.Network pharmacology and Western blot results showed that the effect of EH extract on NS may be associated with the CAMKK2/AMPK/mTOR signaling pathway.Moreover,methylephedrine significantly ameliorated adriamycin-induced NRK-52e cell injury.Methylephedrine also significantly improved the phosphorylation of AMPK and mTOR,which were blocked by CC.In sum,EH extract may ameliorate renal injury via the CAMKK2/AMPK/mTOR signaling pathway.Moreover,methylephedrine may be one of the material bases of EH extract.展开更多
Emerging evidence discloses the involvement of calcium channel protein in the pathological process of liver diseases.Transient receptor potential cation channel subfamily C member 3(TRPC3),a ubiquitously expressed non...Emerging evidence discloses the involvement of calcium channel protein in the pathological process of liver diseases.Transient receptor potential cation channel subfamily C member 3(TRPC3),a ubiquitously expressed non-selective cation channel protein,controls proliferation,inflammation,and immune response via operating calcium influx in various organs.However,our understanding on the biofunction of hepatic TRPC3 is still limited.The present study aims to clarify the role and potential mechanism(s)of TRPC3 in alcohol-associated liver disease(ALD).We recently found that TRPC3 expression plays an important role in the disease process of ALD.Alcohol exposure led to a significant reduction of hepatic TRPC3 in patients with alcohol-related hepatitis(AH)and ALD models.Antioxidants(N-acetylcysteine and mitoquinone)intervention improved alcohol-induced suppression of TRPC3 via a miR-339-5p-involved mechanism.TRPC3 loss robustly aggravated the alcohol-induced hepatic steatosis and liver injury in mouse liver;this was associated with the suppression of Ca^(2+)/calmodulin-dependent protein kinase kinase 2(CAMKK2)/AMP-activated protein kinase(AMPK)and dysregulation of genes related to lipid metabolism.TRPC3 loss also enhanced hepatic inflammation and early fibrosis-like change in mice.Replenishing hepatic TRPC3 effectively reversed chronic alcohol-induced detrimental alterations in ALD mice.Briefly,chronic alcohol exposure-induced TRPC3 reduction contributes to the pathological development of ALD via suppression of the CAMKK2/AMPK pathway.Oxidative stress-stimulated miR-339-5p upregulation contributes to alcohol-reduced TRPC3.TRPC3 is the requisite and a potential target to defend alcohol consumption-caused ALD.展开更多
Background: Autophagy is elevated in metastatic tumors and is often associatedwith active epithelial-to-mesenchymal transition (EMT). However, the extent towhich EMT is dependent on autophagy is largely unknown. This ...Background: Autophagy is elevated in metastatic tumors and is often associatedwith active epithelial-to-mesenchymal transition (EMT). However, the extent towhich EMT is dependent on autophagy is largely unknown. This study aimed toidentify the mechanisms by which autophagy facilitates EMT.Methods: We employed a liquid chromatography-based metabolomic approachwith kirsten rat sarcoma viral oncogene (KRAS) and liver kinase B1 (LKB1)gene co-mutated (KL) cells that represent an autophagy/EMT-coactivatedinvasive lung cancer subtype for the identification of metabolites linked to autophagy-driven EMT activation. Molecular mechanisms of autophagy-drivenEMT activation were further investigated by quantitative real-time polymerasechain reaction (qRT-PCR), Western blotting analysis, immunoprecipitation,immunofluorescence staining, and metabolite assays. The effects of chemicaland genetic perturbations on autophagic flux were assessed by two orthogonalapproaches: microtubule-associated protein 1A/1B-light chain 3 (LC3) turnoveranalysis by Western blotting and monomeric red fluorescent protein-greenfluorescent protein (mRFP-GFP)-LC3 tandem fluorescent protein quenchingassay. Transcription factor EB (TFEB) activity was measured by coordinatedlysosomal expression and regulation (CLEAR) motif-driven luciferase reporterassay. Experimental metastasis (tail vein injection) mouse models were used toevaluate the impact of calcium/calmodulin-dependent protein kinase kinase 2(CAMKK2) or ATP citrate lyase (ACLY) inhibitors on lung metastasis using IVISluciferase imaging system.Results: We found that autophagy in KL cancer cells increased acetyl-coenzymeA (acetyl-CoA), which facilitated the acetylation and stabilization of theEMT-inducing transcription factor Snail. The autophagy/acetyl-CoA/acetylSnail axis was further validated in tumor tissues and in autophagy-activatedpancreatic cancer cells. TFEB acetylation in KL cancer cells sustained prometastatic autophagy in a mammalian target of rapamycin complex 1 (mTORC1)-independent manner. Pharmacological inhibition of this axis via CAMKK2inhibitors or ACLY inhibitors consistently reduced the metastatic capacity of KLcancer cells in vivo.Conclusions: This study demonstrates that autophagy-derived acetyl-CoA promotes Snail acetylation and thereby facilitates invasion and metastasis of KRASLKB1 co-mutated lung cancer cells and that inhibition of the autophagy/acetylCoA/acetyl-Snail axis using CAMKK2 or ACLY inhibitors could be a potentialtherapeutic strategy to suppress metastasis of KL lung cancer.展开更多
基金the National Key Research and Development Project(Nos.2019YFC1708802 and 2017YFC1702800)Henan province high-level personnel special support“ZhongYuan One Thousand People Pla”,Zhongyuan Leading Talent(No.ZYQR201810080)the Major Science and Technology Projects in Henan Province:Study on the key technology for quality control and the key characteristics of Rehmannia glutinosa,Dioscorea opposita Thunb.and Achyranthes bidentata Blume from Henan Province(No.171100310500).
文摘This study aimed to investigate the effect and mechanisms of Ephedra Herb(EH)extract on adriamycin-induced nephrotic syndrome(NS),providing an experimental basis for the clinical treatment of NS.Hematoxylin and eosin staining,creatinine,urea nitrogen,and kidn injury molecule-1 were used to evaluate the activities of EH extract on renal function.The levels of inflammatory factors and oxidative stress were detected by kits.The levels of reactive oxygen species,immune cells,and apoptosis were measured by flow cytometry.A network pharmacological approach was used to predict the potential targets and mechanisms of EH extract in the treatment of NS.The protein levels of apoptosis-related proteins and CAMKK2,p-CAMKK2,AMPK,p-AMPK,mTOR and p-mTOR in the kidneys were detected by Western blot.The effective material basis of EH extract was screened by MTT assay.The AMPK pathway inhibitor(compound C,CC)was added to investigate the effect of the potent material basis on adriamycin-induced cell injury.EH extract significantly improved renal injury and relieve inflammation,oxidative stress,and apoptosis in rats.Network pharmacology and Western blot results showed that the effect of EH extract on NS may be associated with the CAMKK2/AMPK/mTOR signaling pathway.Moreover,methylephedrine significantly ameliorated adriamycin-induced NRK-52e cell injury.Methylephedrine also significantly improved the phosphorylation of AMPK and mTOR,which were blocked by CC.In sum,EH extract may ameliorate renal injury via the CAMKK2/AMPK/mTOR signaling pathway.Moreover,methylephedrine may be one of the material bases of EH extract.
基金supported by grants from the National Natural Science Foundation of China(81973041,82273625,82103839,and 82103838)Zhejiang Natural Science Foundation(LR20H260001 and LZ21H030001)Zhejiang Chinese Medicine University Postgraduate Scientific Research Fund Project(2022YKJ17).
文摘Emerging evidence discloses the involvement of calcium channel protein in the pathological process of liver diseases.Transient receptor potential cation channel subfamily C member 3(TRPC3),a ubiquitously expressed non-selective cation channel protein,controls proliferation,inflammation,and immune response via operating calcium influx in various organs.However,our understanding on the biofunction of hepatic TRPC3 is still limited.The present study aims to clarify the role and potential mechanism(s)of TRPC3 in alcohol-associated liver disease(ALD).We recently found that TRPC3 expression plays an important role in the disease process of ALD.Alcohol exposure led to a significant reduction of hepatic TRPC3 in patients with alcohol-related hepatitis(AH)and ALD models.Antioxidants(N-acetylcysteine and mitoquinone)intervention improved alcohol-induced suppression of TRPC3 via a miR-339-5p-involved mechanism.TRPC3 loss robustly aggravated the alcohol-induced hepatic steatosis and liver injury in mouse liver;this was associated with the suppression of Ca^(2+)/calmodulin-dependent protein kinase kinase 2(CAMKK2)/AMP-activated protein kinase(AMPK)and dysregulation of genes related to lipid metabolism.TRPC3 loss also enhanced hepatic inflammation and early fibrosis-like change in mice.Replenishing hepatic TRPC3 effectively reversed chronic alcohol-induced detrimental alterations in ALD mice.Briefly,chronic alcohol exposure-induced TRPC3 reduction contributes to the pathological development of ALD via suppression of the CAMKK2/AMPK pathway.Oxidative stress-stimulated miR-339-5p upregulation contributes to alcohol-reduced TRPC3.TRPC3 is the requisite and a potential target to defend alcohol consumption-caused ALD.
基金Korea Health Technology R&D Project through the Korea Health Industry Development Institute,Grant/Award Number:HI14C1324National Research Foundation of Korea,Grant/Award Numbers:2020R1A2C3007792,2019R1A2C3004155,2019H1A2A1075632+2 种基金NCI Lung Cancer SPORE,Grant/Award Number:P50CA70907Cancer Prevention and Research Institute of Texas(CPRIT),Grant/Award Number:RP160652“Team Science Award”of Yonsei University College of Medicine,Grant/Award Number:6-2021-0194。
文摘Background: Autophagy is elevated in metastatic tumors and is often associatedwith active epithelial-to-mesenchymal transition (EMT). However, the extent towhich EMT is dependent on autophagy is largely unknown. This study aimed toidentify the mechanisms by which autophagy facilitates EMT.Methods: We employed a liquid chromatography-based metabolomic approachwith kirsten rat sarcoma viral oncogene (KRAS) and liver kinase B1 (LKB1)gene co-mutated (KL) cells that represent an autophagy/EMT-coactivatedinvasive lung cancer subtype for the identification of metabolites linked to autophagy-driven EMT activation. Molecular mechanisms of autophagy-drivenEMT activation were further investigated by quantitative real-time polymerasechain reaction (qRT-PCR), Western blotting analysis, immunoprecipitation,immunofluorescence staining, and metabolite assays. The effects of chemicaland genetic perturbations on autophagic flux were assessed by two orthogonalapproaches: microtubule-associated protein 1A/1B-light chain 3 (LC3) turnoveranalysis by Western blotting and monomeric red fluorescent protein-greenfluorescent protein (mRFP-GFP)-LC3 tandem fluorescent protein quenchingassay. Transcription factor EB (TFEB) activity was measured by coordinatedlysosomal expression and regulation (CLEAR) motif-driven luciferase reporterassay. Experimental metastasis (tail vein injection) mouse models were used toevaluate the impact of calcium/calmodulin-dependent protein kinase kinase 2(CAMKK2) or ATP citrate lyase (ACLY) inhibitors on lung metastasis using IVISluciferase imaging system.Results: We found that autophagy in KL cancer cells increased acetyl-coenzymeA (acetyl-CoA), which facilitated the acetylation and stabilization of theEMT-inducing transcription factor Snail. The autophagy/acetyl-CoA/acetylSnail axis was further validated in tumor tissues and in autophagy-activatedpancreatic cancer cells. TFEB acetylation in KL cancer cells sustained prometastatic autophagy in a mammalian target of rapamycin complex 1 (mTORC1)-independent manner. Pharmacological inhibition of this axis via CAMKK2inhibitors or ACLY inhibitors consistently reduced the metastatic capacity of KLcancer cells in vivo.Conclusions: This study demonstrates that autophagy-derived acetyl-CoA promotes Snail acetylation and thereby facilitates invasion and metastasis of KRASLKB1 co-mutated lung cancer cells and that inhibition of the autophagy/acetylCoA/acetyl-Snail axis using CAMKK2 or ACLY inhibitors could be a potentialtherapeutic strategy to suppress metastasis of KL lung cancer.