Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 20
Filter
1.
Phytother Res ; 37(8): 3495-3507, 2023 Aug.
Article in English | MEDLINE | ID: mdl-37125528

ABSTRACT

Effective amelioration of ischemia/reperfusion (I/R)-induced intestinal injury and revealing its mechanisms remain the challenges in both preclinic and clinic. Potential mechanisms of naringin in ameliorating I/R-induced intestinal injury remain unknown. Based on pre-experiments, I/R-injured rat intestine in vivo and hypoxia-reoxygenation (H/R)-injured IEC-6 cells in vitro were used to verify that naringin-alleviated I/R-induced intestinal injury was mediated via deactivating cGAS-STING signaling pathway. Naringin improved intestinal damage using hematoxylin and eosin staining and decreased alanine aminotransferase and aspartate aminotransferase contents in plasma. Naringin decreased inflammation characterized by reducing IL-6, IL-1ß, TNF-α, and IFN-ß contents in both plasma and IEC-6 cells. Naringin mitigated oxidative stress via recovering superoxide dismutase, glutathione, and malondialdehyde levels in the I/R-injured intestine. Naringin reduced the expression of apoptotic proteins, including Bax, caspase-3, and Bcl-2, and reduced terminal deoxynucleotidyl transferase-mediated dUTP-biotin nick-end labeling-positive cells both in vivo and in vitro, and decreased Hoechst 33342 signals in vitro. cGAS, STING, p-TBK1, p-IRF3, and NF-κB expressions were up-regulated both in vivo and in vitro respectively and the up-regulated indexes were reversed by naringin. Transfection of cGAS-siRNA and cGAS-cDNA significantly down-regulated and up-regulated cGAS-STING signaling-related protein expressions, respectively, and partially weakened naringin-induced amelioration on these indexes, suggesting that deactivation of cGAS-STING signaling is the crucial target for naringin-induced amelioration on I/R-injured intestine.


Subject(s)
Intestines , Reperfusion Injury , Rats , Animals , Signal Transduction , Inflammation/drug therapy , Nucleotidyltransferases/metabolism , Reperfusion Injury/drug therapy , Apoptosis
2.
Phytother Res ; 36(11): 4244-4262, 2022 Nov.
Article in English | MEDLINE | ID: mdl-35820659

ABSTRACT

Vancomycin (VCM)'s nephrotoxicity limits its application and therapeutic efficiency. The aim of this study was to determine the protective effect of rhein against VCM-induced nephrotoxicity (VIN). VIN models were established in rats and NRK-52E cells. Rhein up-regulated the expressions of renal organic anion transporter (Oat) 1, Oat3, organic cation transporter 2 (Oct2), multidrug resistance-associated protein 2 (Mrp2), mammal multidrug and toxin extrusion proteins 1 (Mate 1) and P-glycoprotein (P-gp) to facilitate the efflux of plasma creatinine, blood urea nitrogen (BUN), and plasma indoxyl sulfate. Rhein increased the expression of nuclear factor erythroid 2-related factor 2 (Nrf2) to regulate the expression of Mrp2, P-gp, and Mate 1. The increased level of superoxide dismutase (SOD), decreased level of malondialdehyde (MDA) and reduced number of apoptosis cells were observed after treatment of rhein. Rhein decreased the number of apoptosis cells as well as increased the expression of B-cell lymphoma-2 (Bcl-2) and decreased expressions of Bcl-2-like protein 4 (Bax). ML385, as a typical inhibitor of Nrf2, reversed the protective effects of rhein in cells. Rhein oriented itself in the site of Keap1, inhibiting the Keap1-Nrf2 interaction. Rhein ameliorated VIN mainly through regulating the expressions of renal transporters and acting on Nrf2 pathway.


Subject(s)
NF-E2-Related Factor 2 , Vancomycin , Rats , Animals , Vancomycin/pharmacology , Kelch-Like ECH-Associated Protein 1/metabolism , NF-E2-Related Factor 2/metabolism , Kidney , Proto-Oncogene Proteins c-bcl-2/metabolism , Oxidative Stress , Mammals/metabolism
3.
Phytother Res ; 36(2): 899-913, 2022 Feb.
Article in English | MEDLINE | ID: mdl-35041255

ABSTRACT

Nonalcoholic fatty liver disease (NAFLD), a metabolic disease, has received wide attention worldwide. However, there is no approved effective drug for NAFLD treatment. In the study, H&E and Oil Red O staining were employed to detect liver histopathological changes and the accumulation of lipid droplets. Quantitative real-time PCR, Western blot, bioinformatics, luciferase assay, immunofluorescence staining, reactive oxygen species (ROS), and siRNA were used to further elucidate the mechanism of isoliquiritigenin (ISL) against NAFLD. The results showed that ISL significantly reduced the liver-to-body weight ratios and biochemical index. And the staining results showed that ISL remarkedly ameliorated liver histopathological changes of NAFLD. Furthermore, ISL significantly increased the levels of PPARα, CPT1α, and ACADS, which were involved in lipid metabolism, and inhibited the ROS, TNF-α, IL-1ß, and IL-6 expression by activating PGC-1α. Bioinformatics and luciferase assay analysis confirmed that miR-138-5p might bind to PGC-1α mRNA in NAFLD. Importantly, the expression of miR-138-5p was increased in the NAFLD, which was significantly decreased by ISL. In addition, the miR-138-5p inhibitor also promoted lipid metabolism and inhibited inflammatory response in NAFLD via PGC-1α activation. The above results demonstrate that ISL alleviates NAFLD through modulating miR-138-5p/PGC-1α-mediated lipid metabolism and inflammatory reaction in vivo and in vitro.


Subject(s)
Chalcones , MicroRNAs , Non-alcoholic Fatty Liver Disease , Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha/metabolism , Animals , Chalcones/pharmacology , Down-Regulation , Humans , Liver , Male , Mice, Inbred C57BL , MicroRNAs/metabolism , Non-alcoholic Fatty Liver Disease/drug therapy , Non-alcoholic Fatty Liver Disease/metabolism
4.
Oxid Med Cell Longev ; 2021: 5147069, 2021.
Article in English | MEDLINE | ID: mdl-34630849

ABSTRACT

Intestinal ischemia-reperfusion (I/R) may induce cell/tissue injuries, leading to multiple organ failure. Based on our preexperiments, we proposed that sesamin could protect against and ameliorate intestinal I/R injuries and related disorders with involvement of activating Nrf2 signaling pathway. This proposal was evaluated using SD intestinal I/R injury rats in vivo and hypoxia/reoxygenation- (H/R-) injured rat small intestinal crypt epithelial cell line (IEC-6 cells) in vitro. Sesamin significantly alleviated I/R-induced intestinal histopathological injuries and significantly reduced serum biochemical indicators ALT and AST, alleviating I/R-induced intestinal injury in rats. Sesamin also significantly reversed I/R-increased TNF-α, IL-6, IL-1ß, and MPO activity in serum and MDA in tissues and I/R-decreased GSH in tissues and SOD in both tissues and IEC-6 cells, indicating its anti-inflammatory and antioxidative stress effects. Further, sesamin significantly decreased TUNEL-positive cells, downregulated the increased Bax and caspase-3 protein expression, upregulated the decreased protein expression of Bcl-2 in I/R-injured intestinal tissues, and significantly reversed H/R-reduced IEC-6 cell viability as well as reduced the number of apoptotic cells among H/R-injured IEC-6 cell, showing antiapoptotic effects. Activation of Nrf2 is known to ameliorate tissue/cell injuries. Consistent with sesamin-induced ameliorations of both intestinal I/R injuries and H/R injuries, transfection of Nrf2 cDNA significantly upregulated the expression of Nrf2, HO-1, and NQO1, respectively. On the contrary, either Nrf2 inhibitor (ML385) or Nrf2 siRNA transfection significantly decreased the expression of these proteins. Our results suggest that activation of the Nrf2/HO-1/NQO1 signaling pathway is involved in sesamin-induced anti-inflammatory, antioxidative, and antiapoptotic effects in protection against and amelioration of intestinal I/R injuries.


Subject(s)
Anti-Inflammatory Agents/administration & dosage , Antioxidants/administration & dosage , Dioxoles/administration & dosage , Heme Oxygenase (Decyclizing)/metabolism , Intestinal Diseases/drug therapy , Intestinal Diseases/metabolism , Lignans/administration & dosage , NAD(P)H Dehydrogenase (Quinone)/metabolism , NF-E2-Related Factor 2/metabolism , Phytotherapy/methods , Plant Extracts/administration & dosage , Reperfusion Injury/drug therapy , Reperfusion Injury/metabolism , Sesamum/chemistry , Signal Transduction/drug effects , Animals , Apoptosis/drug effects , Cell Line , Disease Models, Animal , Epithelial Cells/drug effects , Epithelial Cells/metabolism , Intestinal Mucosa/cytology , Male , NF-E2-Related Factor 2/genetics , Oxidative Stress/drug effects , Rats , Rats, Sprague-Dawley , Signal Transduction/genetics , Transfection , Treatment Outcome
5.
Phytomedicine ; 90: 153629, 2021 Sep.
Article in English | MEDLINE | ID: mdl-34304130

ABSTRACT

BACKGROUND: Alcoholic liver disease (ALD) is a progressive disease beginning with simple steatosis but can progress to alcoholic steatohepatitis, fibrosis, cirrhosis, and even hepatocellular carcinoma. The morbidity of ALD is on the rise and has been a large burden on global healthcare system. It is unfortunately that there are currently no approved therapeutic drugs against ALD. Hence, it is of utmost urgency to develop the efficacious therapies. The ability of many molecular targets against ALD is under investigation. Farnesoid X receptor (FXR), a member of the ligand-activated transcription factor superfamily, has been recently demonstrated to have a crucial role in the pathogenesis and progression of ALD. PURPOSE: The purpose of the study is to determine whether Yangonin (YAN), a FXR agonist previously demonstrated by us, exerts the hepatoprotective effects against ALD and further to clarify the mechanisms in vitro and in vivo. STUDY DESIGN: The alcoholic liver disease model induced by Lieber-Decarli liquid diet was established with or without Yan treatment. METHODS: We determined the liver to body weight ratios, the body weight, serum and hepatic biochemical indicators. The alleviation of the liver histopathological progression was evaluated by H&E and immunohistochemical staining. Western blot and quantitative real-time PCR were used to demonstrate YAN treatment-mediated alleviation mechanisms of ALD. RESULTS: The data indicated that YAN existed hepatoprotective activity against ALD via FXR activation. YAN improved the lipid homeostasis by decreasing hepatic lipogenesis and increasing fatty acid ß-oxidation and lipoprotein lipolysis through modulating the related protein. Also, YAN ameliorated ethanol-induced cholestasis via inhibiting bile acid uptake transporter Ntcp and inducing bile acid efflux transporter Bsep and Mrp2 expression. Besides, YAN improved bile acid homeostasis via inducing Sult2a1 expression and inhibiting Cyp7a1 and Cyp8b1 expression. Furthermore, YAN attenuated ethanol-triggered hepatocyte damage by inhibiting cellular senescence marker P16, P21 and Hmga1 expression. Also, YAN alleviated ethanol-induced inflammation by down-regulating the inflammation-related gene IL-6, IL-1ß and TNF-α expression. Notably, the protective effects of YAN were cancelled by FXR siRNA in vitro and FXR antagonist GS in vivo. CONCLUSIONS: YAN exerted significant hepatoprotective effects against liver injury triggered by ethanol via FXR-mediated target gene modulation.


Subject(s)
Cellular Senescence , Cholestasis , Lipid Metabolism , Liver Diseases, Alcoholic , Pyrones/pharmacology , Receptors, Cytoplasmic and Nuclear/metabolism , Animals , Bile Acids and Salts , Homeostasis , Liver , Liver Diseases, Alcoholic/drug therapy , Mice , Mice, Inbred C57BL
6.
Phytother Res ; 35(3): 1658-1668, 2021 Mar.
Article in English | MEDLINE | ID: mdl-33141989

ABSTRACT

Puerarin is an isoflavone isolated from Pueraria lobata (Willd.) Ohwi. In the present study, reversal effect and underlying mechanisms of puerarin on multidrug resistance (MDR) were investigated in K562/ADR cells. K562/ADR cells exhibited adriamycin (ADR) resistance and higher levels of MDR1 expression compared with K562 cells. Puerarin enhanced the chemosensitivity of K562/ADR cells and increased the ADR accumulation in K562/ADR cells. The expression levels of MDR1 were down-regulated by puerarin in K562/ADR cells. Luciferase reporter assay further demonstrated the inhibitory effect of puerarin on TNF-α-induced NF-κB activation. The phosphorylation of IκB-α was significantly suppressed by puerarin. In silico docking analyses suggested that puerarin well matched with the active sites of IκB-α. Moreover, a large number of autophagosomes were found in the cytoplasm of K562/ADR cells after puerarin treatment. The significant increase in LC3-II and beclin-1 was also observed, indicating autophagy induction by puerarin in K562/ADR cells. Puerarin induced cell cycle arrest and apoptosis in K562/ADR cells. Finally, puerarin inhibited phosphorylation of Akt and JNK. In conclusion, puerarin-sensitized K562/ADR cells by downregulating MDR1 expression via inhibition of NF-κB pathway and autophagy induction via Akt inhibition.


Subject(s)
Autophagy/drug effects , Isoflavones/therapeutic use , K562 Cells/metabolism , NF-kappa B/metabolism , Vasodilator Agents/therapeutic use , Humans , Isoflavones/pharmacology , Transfection , Vasodilator Agents/pharmacology
7.
Phytother Res ; 34(11): 2998-3010, 2020 Nov.
Article in English | MEDLINE | ID: mdl-32468621

ABSTRACT

Imipenem (Imp) is a widely used broad-spectrum antibiotic. However, renal adverse effects limit its clinical application. We previously reported that organic anion transporters (OATs) facilitated the renal transport of Imp and contributed its nephrotoxicity. Natural flavonoids exhibited renal protective effect. Here, we aimed to develop potent OAT inhibitors from traditional Chinese medicines (TCMs) and to evaluate its protective effect against Imp-induced nephrotoxicity. Among 50 TCMs, Tribuli Fructus, Platycladi Cacumen, and Lycopi Herba exhibited potent inhibition on OAT1/3. After screening their main components, Apigenin strongly inhibited Imp uptake by OAT1/3-HEK293 cells with IC50 values of 1.98 ± 0.36 µM (OAT1) and 2.29 ± 0.88 µM (OAT3). Moreover, Imp exhibited OAT1/3-dependent cytotoxicity, which was alleviated by Apigenin. Furthermore, Apigenin ameliorated Imp-induced nephrotoxicity in rabbits, and reduced the renal secretion of Imp. Apigenin inhibited intracellular accumulation of Imp and sequentially decreased the nephrocyte toxicity in rabbit primary proximal tubule cells (rPTCs). Apigenin, a flavone widely distributed in TCMs, was a potent OAT1/3 inhibitor. Through OAT inhibition, at least in part, Apigenin decreased the renal exposure of Imp and consequently protected against the nephrotoxicity of Imp. Apigenin can be used as a promising agent to reduce the renal adverse reaction of Imp in clinic.


Subject(s)
Apigenin/therapeutic use , Imipenem/adverse effects , Kidney Diseases/chemically induced , Kidney Diseases/drug therapy , Medicine, Chinese Traditional/methods , Organic Anion Transporters/therapeutic use , Animals , Apigenin/pharmacology , Humans , Kidney Tubules, Proximal/drug effects , Kidney Tubules, Proximal/metabolism , Male , Rabbits , Transfection
8.
Oxid Med Cell Longev ; 2020: 7691352, 2020.
Article in English | MEDLINE | ID: mdl-32308808

ABSTRACT

Saccharides from Arctium lappa. L. root (ALR-S) is a high-purity fructosaccharide separated from the medicinal plant Arctium lappa. L. root. These compounds showed many pharmacological effects in previous studies. In the present study, the antithrombotic effects of ALR-S in arterial thrombosis via inhibiting platelet adhesion and rebalancing thrombotic and antithrombotic factor expression and secretion were found in rats and human aortic endothelial cells (HAECs). This study also showed that inhibition of oxidative stress (OS), which is closely involved in the expression of coagulation- and thrombosis-related proteins, was involved in the antithrombotic effects of ALR-S. Furthermore, studies using FeCl3-treated HAECs showed that ALR-S induced the abovementioned effects at least partly by blocking the ERK/NF-κB pathway. Moreover, U0126, a specific inhibitor of ERK, exhibited the same effects with ALR-S on a thrombotic process in FeCl3-injured HAECs, suggesting the thrombotic role of the ERK/NF-κB pathway and the antithrombotic role of blocking the ERK/NF-κB pathway by ALR-S. In conclusion, our study revealed that the ERK/NF-κB pathway is a potential therapeutic target in arterial thrombosis and that ALR-S has good characteristics for the cure of arterial thrombosis via regulating the ERK/NF-κB signaling pathway.


Subject(s)
Arctium/chemistry , MAP Kinase Signaling System/physiology , NF-kappa B/metabolism , Plant Roots/chemistry , Plants, Medicinal/chemistry , Animals , Humans , Male , Rats , Rats, Sprague-Dawley , Signal Transduction , Thrombosis
9.
Adv Healthc Mater ; 8(24): e1901005, 2019 12.
Article in English | MEDLINE | ID: mdl-31738019

ABSTRACT

A great challenge still remains to explore the facile approaches to construct multifunctional nanoparticles for acquiring precise cancer theranostics. Herein, a biocompatible theranostic nanoplatform capable of simultaneous cancer imaging and therapy is attempted by loading of paclitaxel (PTX) and indocyanine green (ICG) molecules into the matrix of Gd2 O3 @human serum albumin (HSA) nanoparticles (PIGH NPs) via hydrophobic interaction. The subsequent in vitro investigations reveal that the PIGH NPs afford uniform particle size, sustained drug release profile, strong longitudinal relaxivity, potent photothermal effect, effective singlet oxygen generation, and ideal resistance to photobleaching. Moreover, the PIGH NPs achieve high cellular uptake, efficient cytoplasmic drug translocation based on singlet oxygen-triggered endolysosomal disruption and prominent cytotoxicity effect against 4T1 cells under 808 nm near-infrared (NIR) irradiation in contrast to PTX/ICG-loaded HSA nanoparticles (PIH NPs) and free PTX/ICG. After intravenous injection, the PIGH NPs exhibit preferable tumor accumulation and achieve effective tumor ablation in 4T1 tumor bearing mouse model with excellent dual near-infrared fluorescence/magnetic resonance (NIRF/MR) imaging guided synergistic chemo-phototherapy. Hence, the PIGH NPs can be utilized as potential theranostic nanosystem for simultaneous cancer imaging and therapy.


Subject(s)
Gadolinium/chemistry , Nanoparticles/chemistry , Phototherapy/methods , Serum Albumin, Human/chemistry , Animals , Cell Line, Tumor , Drug Delivery Systems/methods , Endocytosis/physiology , Female , Indocyanine Green/chemistry , Mice , Temperature , Theranostic Nanomedicine
10.
Int Immunopharmacol ; 65: 458-469, 2018 Dec.
Article in English | MEDLINE | ID: mdl-30390593

ABSTRACT

Development of active compounds to control inflammation against systemic inflammatory response syndrome (SIRS) is critical important. Dioscin shows anti-inflammatory effects in our previous works. However, the action of the compound on SIRS still remained unknown. In the present paper, zymosan induced generalized inflammation (ZIGI) models in mice and rats, and PMA-differentiated THP­1 cells stimulated by lipopolysaccharide (LPS) and Pam3-Cys-Ser-Lys4 (Pam3CSK4) were used. The results showed that dioscin significantly inhibited the proliferation of THP­1 cells stimulated by LPS and Pam3CSK4, obviously reduced the soakage of inflammatory cells and necrosis in liver, kidney and intestine of rats and mice, and reduced peritoneal ascites fluid compared with ZIGI model groups. In addition, dioscin significantly declined the levels of alanine transaminase (ALT), aspartate transaminase (AST), creatinine (Cr), blood urea nitrogen (BUN), malondialdehyde (MDA) and myeloperoxidase (MPO), increased the levels of superoxide dismutase (SOD) in rats and mice. The migration of macrophages in tissues was also suppressed by dioscin. Mechanism investigation showed that dioscin significantly inhibited the expression levels of TLR2, MyD88, NF­κb, HMGB­1, increased the expression levels of IKBα, and decreased the mRNA levels of interleukin­1 beta (IL­1ß), interleukin­6 (IL­6) and tumor necrosis factor­alpha (TNF­α) in liver, kidney, intestine tissues of rats and mice, and in PMA-differentiated THP­1 cells, which were further confirmed by TLR2 siRNA silencing in vitro. In conclusion, our data confirmed that dioscin exhibited protective effects against SIRS via adjusting TLR2/MyD88 signal pathway, which should be developed as one potent candidate to treat SIRS in the future.


Subject(s)
Anti-Inflammatory Agents/therapeutic use , Diosgenin/analogs & derivatives , Macrophages/drug effects , Animals , Dioscorea/immunology , Diosgenin/therapeutic use , Humans , Macrophages/physiology , Male , Mice , Mice, Inbred C57BL , Myeloid Differentiation Factor 88/metabolism , NF-kappa B/metabolism , Rats , Rats, Sprague-Dawley , Signal Transduction/drug effects , Systemic Inflammatory Response Syndrome/drug therapy , THP-1 Cells , Toll-Like Receptor 2/metabolism , Zymosan/immunology
11.
Regul Toxicol Pharmacol ; 98: 58-68, 2018 Oct.
Article in English | MEDLINE | ID: mdl-30030101

ABSTRACT

Acetaminophen (APAP) is a worldwide used drug for treating fever and pain. However, APAP overdose is the leading cause of drug-induced liver injury. The purpose of the current study is to evaluate the hepatoprotective effect of ginsenoside Rg1 (Rg1), the main pharmacologically active compounds of Panax ginseng, against APAP-induced acute liver injury, and further to elucidate the involvement of Nrf2 signaling pathway by in vivo and in vitro experiments. Male C57BL/6 mice were treated with Rg1 for 3 days before injection of APAP. Serum and liver tissue samples were collected 6 h later. The results indicated that Rg1 significantly attenuated APAP-induced hepatotoxicity and oxidative stress in a dose-dependent manner. Rg1 effectively enhanced antioxidant and detoxification capacity, which is largely dependent on up-regulating Nrf2 nuclear translocation, reducing Keap1 protein expression and up-regulating Nrf2 target genes including GCLC, GCLM, HO-1, NQO1, Ugt1a1, Ugt1a6, Ugt2b1, Sult2a1, Mrp2, Mrp3 and Mrp4. Furthermore, Rg1 repressed the activities of Cyp2e1, Cyp3a11, Cyp1a2, which are important enzymes in the formation of APAP toxic metabolite N-acetyl-p-benzoquinone imine. However, the changes in transporters and enzymes, as well as ameliorative liver histology induced by Rg1 were abrogated by Nrf2 antagonist all-transretinoic acid in vivo and Nrf2 siRNA in vitro. In conclusion, Rg1 produced hepatoprotective effects against APAP-induced acute liver injury via Nrf2 signaling pathway. Rg1 might be an effective approach for the prevention against acute liver injury.


Subject(s)
Chemical and Drug Induced Liver Injury/drug therapy , Ginsenosides/pharmacology , Ginsenosides/therapeutic use , NF-E2-Related Factor 2/metabolism , Protective Agents/pharmacology , Protective Agents/therapeutic use , Acetaminophen , Animals , Cells, Cultured , Chemical and Drug Induced Liver Injury/metabolism , Cytochrome P-450 Enzyme System/genetics , Gene Expression Regulation, Enzymologic/drug effects , Hepatocytes/drug effects , Hepatocytes/metabolism , Male , Membrane Transport Proteins/genetics , Mice, Inbred C57BL , NF-E2-Related Factor 2/genetics , Oxidative Stress/drug effects , RNA, Small Interfering/genetics , Signal Transduction/drug effects
12.
Environ Toxicol ; 33(10): 1050-1060, 2018 Oct.
Article in English | MEDLINE | ID: mdl-29964319

ABSTRACT

Oxidative stress and inflammatory response are well known to be involved in the pathogenesis of acute liver injury. This study was performed to examine the hepatoprotective effect of ginsenoside Rg1 (Rg1) against CCl4 -induced acute liver injury, and further to elucidate the involvement of Nrf2 signaling pathway in vivo and in vitro. Mice were orally administered Rg1 (15, 30, and 60 mg/kg) or sulforaphane (SFN) once daily for 1 week prior to 750 µL/kg CCl4 injection. The results showed that Rg1 markedly altered relative liver weights, promoted liver repair, increased the serum level of TP and decreased the serum levels of ALT, AST and ALP. Hepatic oxidative stress was inhibited by Rg1, as evidenced by the decrease in MDA, and increases in GSH, SOD, and CAT in the liver. Further research demonstrated that Rg1 suppressed liver inflammation response through repressing the expression levels of inflammation-related genes including TNF-α, IL-1ß, IL-6, COX-2, and iNOS. In addition, Rg1 enhanced antioxidative stress and liver detoxification abilities by up-regulating Nrf2 and its target-genes such as GCLC, GCLM, HO-1, NQO1, Besp, Mrp2, Mrp3, Mrp4, and down-regulating Cyp2e1. However, the changes in Nrf2 target-genes, as well as ameliorative liver histology induced by Rg1 were abrogated by Nrf2 antagonist all-transretinoic acid in vivo and Nrf2 siRNA in vitro. Overall, the findings indicated that Rg1 might be an effective approach for the prevention against acute liver injury by activating Nrf2 signaling pathway.


Subject(s)
Chemical and Drug Induced Liver Injury/pathology , Ginsenosides/pharmacology , Panax/chemistry , Protective Agents/pharmacology , Signal Transduction/drug effects , Alanine Transaminase/blood , Animals , Antioxidants/metabolism , Aspartate Aminotransferases/blood , Carbon Tetrachloride/toxicity , Chemical and Drug Induced Liver Injury/metabolism , Chemical and Drug Induced Liver Injury/prevention & control , Cytochrome P-450 Enzyme System/genetics , Cytochrome P-450 Enzyme System/metabolism , Cytokines/metabolism , Ginsenosides/chemistry , Ginsenosides/therapeutic use , Liver/drug effects , Liver/pathology , Male , Mice , Mice, Inbred C57BL , NF-E2-Related Factor 2/antagonists & inhibitors , NF-E2-Related Factor 2/genetics , NF-E2-Related Factor 2/metabolism , Oxidative Stress/drug effects , Panax/metabolism , Protective Agents/therapeutic use , RNA Interference , RNA, Small Interfering/metabolism
13.
Int Immunopharmacol ; 61: 266-276, 2018 Aug.
Article in English | MEDLINE | ID: mdl-29902710

ABSTRACT

Acute liver injury (ALI) is a dramatic liver disease characterized by large areas of inflammation in the liver. This study aimed to investigate the protective effects of ginsenoside Rg1 (Rg1), a biologically active component in Panax ginseng, on lipopolysaccharide/d-galactosamine (LPS/D-GalN)-induced ALI in mice, and meanwhile explore the molecular mechanism in vivo and in vitro. Mice were pretreated with Rg1 for three days prior to LPS (40 µg/kg)/D-GalN (700 mg/kg) administration. The results showed that Rg1 improved the survival rate and reduced the liver to body weight ratios in mice. Rg1 also reduced the production of oxidative markers such as MDA and MPO induced by LPS/D-GalN. In addition, Rg1 significantly decreased the production of inflammatory cytokines including TNF-α, IL-6, IL-1ß, Mip-2, Mcp-1, iNOS, and increased the activity of anti-inflammatory cytokine IL-10. Moreover, Rg1 inhibited the protein expression of TLR4 and its downstream genes including NF-κB and MAPKs, which are involved in inflammatory response. Rg1 dramatically reduced oxidative stress by regulating the expression of efflux transporters Mrp2 and various enzymes including GCLC, GCLM, HO-1 and NQO1. However, the changes in these genes and protein induced by Rg1 were abrogated by TLR4 antagonist TAK-242 in vitro. In conclusion, Rg1 had hepatoprotective effect on LPS/D-GalN-induced ALI in mice. The protection may be associated with the inhibition of TLR4. These findings suggest that Rg1 may be a promising agent for prevention against ALI.


Subject(s)
Anti-Inflammatory Agents/therapeutic use , Chemical and Drug Induced Liver Injury/drug therapy , Ginsenosides/therapeutic use , Animals , Cells, Cultured , Cytokines/metabolism , Disease Models, Animal , Galactosamine/immunology , Humans , Inflammation Mediators/metabolism , Lipopolysaccharides/immunology , Male , Mice , Mice, Inbred C57BL , Oxidative Stress/drug effects , Panax/immunology , Signal Transduction , Toll-Like Receptor 4/metabolism
14.
Eur J Pharmacol ; 824: 64-71, 2018 Apr 05.
Article in English | MEDLINE | ID: mdl-29427579

ABSTRACT

Accumulation of toxic bile acids in liver could cause cholestasis and liver injury. The purpose of the current study is to evaluate the hepatoprotective effect of yangonin, a product isolated from an edible botanical Kava against lithocholic acid (LCA)-induced cholestasis, and further to elucidate the involvement of farnesoid X receptor (FXR) in the anticholestatic effect using in vivo and in vitro experiments. The cholestatic liver injury model was established by intraperitoneal injections of LCA in C57BL/6 mice. Serum biomarkers and H&E staining were used to identify the amelioration of cholestasis after yangonin treatment. Mice hepatocytes culture, gene silencing experiment, real-time PCR and Western blot assay were used to elucidate the mechanisms underlying yangonin hepatoprotection. The results indicated that yangonin promoted bile acid efflux and reduced hepatic uptake via an induction in FXR-target genes Bsep, Mrp2 expression and an inhibition in Ntcp, all of which are responsible for bile acid transport. Furthermore, yangonin reduced bile acid synthesis through repressing FXR-target genes Cyp7a1 and Cyp8b1, and increased bile acid metabolism through an induction in gene expression of Sult2a1, which are involved in bile acid synthesis and metabolism. In addition, yangonin suppressed liver inflammation through repressing inflammation-related gene NF-κB, TNF-α and IL-1ß. In vitro evidences showed that the changes in transporters and enzymes induced by yangonin were abrogated when FXR was silenced. In conclusions, yangonin produces protective effect against LCA-induced hepatotoxity and cholestasis due to FXR-mediated regulation. Yangonin may be an effective approach for the prevention against cholestatic liver diseases.


Subject(s)
Cholestasis/chemically induced , Cholestasis/prevention & control , Kava/chemistry , Lithocholic Acid/toxicity , Liver/drug effects , Liver/pathology , Pyrones/pharmacology , Animals , Cell Line , Cholestasis/metabolism , Cholestasis/pathology , Cytoprotection/drug effects , Hepatocytes/drug effects , Hepatocytes/metabolism , Hepatocytes/pathology , Homeostasis/drug effects , Lithocholic Acid/metabolism , Male , Mice , Mice, Inbred C57BL , Pyrones/isolation & purification , Receptors, Cytoplasmic and Nuclear/metabolism
15.
Phytomedicine ; 25: 83-92, 2017 Feb 15.
Article in English | MEDLINE | ID: mdl-28190475

ABSTRACT

BACKGROUND: Non-alcoholic steatohepatitis (NASH) represents the more severe end of hepatic steatosis and is associated with progressive liver disease. Calycosin, derived from the root of Radix Astragali, has been demonstrated to have favorable efficacy on acute liver injury. PURPOSE: The present study was to investigate the hepatoprotective effect of calycosin on attenuating triglyceride accumulation and hepatic fibrosis, as well as explore the potential mechanism in murine model of NASH. STUDY DESIGN: The C57BL/6 male mice were fed with methionine choline deficient (MCD) diet for 4 weeks to induce NASH and treated with or without calycosin by oral gavage for 4 weeks. METHODS: The body weight, liver weight and the liver to body weight ratios were measured. Serum ALT, AST, TG, TC, FFA, MCP-1 and mKC levels were accessed by biochemical methods. H&E staining and Oil red O staining were used to identify the amelioration of liver histopathology. Immunohistochemistry of a-SMA, Masson trichrome staining and Sirius red staining were used to identify the amelioration of hepatic fibrosis. The quantitative real-time-PCR and Western blot were applied to observe the expression changes of key factors involved in triglyceride synthesis, free fatty acid ß-oxidation and hepatic fibrosis. RESULTS: Calycosin significantly inhibited body weight loss induced by MCD diet, decreased the ALT and AST activities, MCP-1 and mKC in a dose-dependent manner. The H&E and Oil red O staining indicated calycosin effectively improved hepatic steatosis, improved the degree of triglyceride accumulation. Masson trichrome and Sirius red staining indicated that calycosin treatment remarkably attenuated the degree of hepatic fibrosis. Immunohistochemistry of a-SMA demonstrated that calycosin attenuated hepatic fibrosis by inhibiting hepatic stellate cell activation. Further, calycosin inhibited the expression of SREBP-1c, FASN, ACC and SCD1 involved in triglyceride synthesis, promoted the expression of PPARa, CPT1, Syndecan-1 and LPL involved in free fatty acid ß-oxidation. The above effects of calycosin were attributed to FXR activation. CONCLUSION: Calycosin attenuates triglyceride accumulation and hepatic fibrosis to protect against NASH via FXR activation.


Subject(s)
Astragalus Plant/chemistry , Drugs, Chinese Herbal/chemistry , Isoflavones/pharmacology , Liver/drug effects , Non-alcoholic Fatty Liver Disease/metabolism , Receptors, Cytoplasmic and Nuclear/agonists , Triglycerides/metabolism , Animals , Chemokine CCL2/metabolism , Diet/adverse effects , Disease Models, Animal , Isoflavones/therapeutic use , Liver/metabolism , Liver/pathology , Liver Cirrhosis/metabolism , Male , Mice, Inbred C57BL , Non-alcoholic Fatty Liver Disease/drug therapy , Non-alcoholic Fatty Liver Disease/etiology , Non-alcoholic Fatty Liver Disease/pathology , Phytotherapy , Receptors, Cytoplasmic and Nuclear/metabolism , Sterol Regulatory Element Binding Protein 1/metabolism
16.
Acta Pharmacol Sin ; 38(1): 69-79, 2017 Jan.
Article in English | MEDLINE | ID: mdl-27773935

ABSTRACT

Alisol B 23-acetate (AB23A) is a natural triterpenoid isolated from the traditional Chinese medicine rhizoma alismatis, which exhibits a number of pharmacological activities, including anti-hepatitis virus, anti-cancer and antibacterial effects. In this study we examined whether AB23A protected against non-alcoholic steatohepatitis (NASH) in mice, and the mechanisms underlying the protective effects. NASH was induced in mice fed a methionine and choline-deficient (MCD) diet for 4 weeks. The mice were simultaneously treated with AB23A (15, 30, and 60 mg·kg-1·d-1, ig) for 4 weeks. On the last day, blood samples and livers were collected. Serum liver functional enzymes, inflammatoru markers were assessed. The livers were histologically examined using H&E, Oil Red O, Masson's trichrome and Sirius Red staining. Mouse primary hepatocytes were used for in vitro experiments. The mechanisms underlying AB23A protection were analyzed using siRNA, qRT-PCR, and Western blot assays. AB23A treatment significantly and dose-dependently decreased the elevated levels of serum ALT and AST in MCD diet-fed mice. Furthermore, AB23A treatment significantly reduced hepatic triglyceride accumulation, inflammatory cell infiltration and hepatic fibrosis in the mice. AB23A-induced decreases in serum and hepatic lipids were related to decreased hepatic lipogenesis through decreasing hepatic levels of SREBP-1c, FAS, ACC1 and SCD1 and increased lipid metabolism via inducing PPARα, CPT1α, ACADS and LPL. The reduction in inflammatory cell infiltration corresponded to deceased serum levels of mKC and MCP-1 and decreased hepatic gene expression of MCP-1 and VCAM-1. The reduction in hepatic fibrosis was correlated with decreased hepatic gene expression of fibrosis markers. The protective effects of AB23A were FXR-dependent, because treatment with the FXR agonist CDCA mimicked AB23A-induced hepato-protection in the mice, whereas co-administration of FXR antagonist guggulsterone abrogated AB23A-induced hepato-protection. In mouse primary hepatocytes, FXR gene silencing abrogated AB23A-induced changes in gene expression of Apo C-II, CPT1α, ACADS and LPL. AB23A produces protective effects against NASH in mice via FXR activation.


Subject(s)
Cholestenones/pharmacology , Non-alcoholic Fatty Liver Disease/prevention & control , Receptors, Cytoplasmic and Nuclear/agonists , Animals , Chenodeoxycholic Acid/pharmacology , Cholestenones/antagonists & inhibitors , Choline Deficiency , Dose-Response Relationship, Drug , Fibrosis/pathology , Gene Expression/drug effects , Hepatocytes/metabolism , Lipid Metabolism/drug effects , Lipogenesis/drug effects , Liver/enzymology , Liver/metabolism , Liver/pathology , Male , Methionine/deficiency , Mice , Pregnenediones/pharmacology , Primary Cell Culture , Protective Agents/pharmacology , Receptors, Cytoplasmic and Nuclear/antagonists & inhibitors
17.
Food Funct ; 7(9): 3716-23, 2016 Sep 14.
Article in English | MEDLINE | ID: mdl-27487733

ABSTRACT

Non-alcoholic steatohepatitis (NASH) is a syndrome with simultaneous severe hepatic steatosis, lobular inflammation and pericelluar fibrosis. The purpose of the present study is to investigate the protective effect of glycyrrhizic acid (GA), a natural triterpene glycoside from edible botanical glycyrrhiza glabra, on NASH induced by a methionine and choline-deficient (MCD) diet in mice, and further to elucidate the mechanisms of GA protection. Serum ALT and AST assay and H&E staining were used to identify the amelioration of the liver histopathological changes. Serum and hepatic lipid assay and Oil Red O staining were used to measure lipid accumulation. Hepatic inflammatory and fibrosis gene determination, as well as Mason Trichrome and Sirius Red staining were used to determine the reduction of hepatic inflammation and pericelluar fibrosis. Quantitative real-time PCR and Western blot assays were used to elucidate the mechanisms underlying GA protection. The results indicated that GA treatment reduced hepatic lipogenesis through a decrease in hepatic levels of SREBP-1c, FAS, ACC1 and SCD1, and increased lipid metabolism through an induction of PPARα, CPT1α, ACADS and LPL. GA also reduced hepatic inflammation via a decrease in the expression of the hepatic inflammatory genes MCP-1 and VCAM-1. In addition, GA reduced liver fibrosis through limiting HSC activation and collagen deposition. In conclusion, GA produces a markedly protective effect against NASH induced by a methionine and choline-deficient (MCD) diet in mice.


Subject(s)
Glycyrrhiza/chemistry , Glycyrrhizic Acid/therapeutic use , Non-alcoholic Fatty Liver Disease/drug therapy , Alanine Transaminase/blood , Animals , Aspartate Aminotransferases/blood , Biomarkers/blood , Diet , Fatty Acids/metabolism , Inflammation/blood , Lipid Metabolism , Lipogenesis , Liver/pathology , Liver/physiopathology , Male , Methionine/deficiency , Mice , Mice, Inbred C57BL , Non-alcoholic Fatty Liver Disease/etiology , Non-alcoholic Fatty Liver Disease/pathology , Triglycerides/metabolism
18.
Pharm Res ; 32(2): 538-48, 2015 Feb.
Article in English | MEDLINE | ID: mdl-25143196

ABSTRACT

PURPOSE: Investigating the hepatoprotective effect of calycosin against acute liver injury in association with FXR activation and STAT3 phosphorylation. METHODS: The acute liver injury model was established by intraperitoneal injection of CCl4 in C57BL/6 mice. Serum alanine aminotransferase, aspartate aminotransferase, HE staining and TUNEL assay were used to identify the amelioration of the liver histopathological changes and hepatocytes apoptosis after calycosin treatment. ELISA kit and 5-bromo-2-deoxyuridine immunohistochemistry were used to measure the liver bile acid concentration and hepatocyte mitotic rate in vivo. The relation between calycosin and activation of FXR and STAT3 was comfirmed using the Luciferase assay, Molecular docking, Real-time PCR and Western Blot in vitro. RESULTS: The liver histopathological changes, hepatocytes apoptosis, liver bile acid overload and hepatocyte mitosis showed significant changes after calycosin treatment. Calycosin promoted the expression of FXR target genes such as FoxM1B and SHP but the effect was reversed by FXR suppressor guggulsterone. Molecular docking results indicated that calycosin could be embedded into the binding pocket of FXR, thereby increasing the expressions of STAT3 tyrosine phosphorylation and its target genes, Bcl-xl and SOCS3. CONCLUSIONS: Calycosin plays a critical role in hepatoprotection against liver injury in association with FXR activation and STAT3 phosphorylation.


Subject(s)
Carbon Tetrachloride/toxicity , Chemical and Drug Induced Liver Injury/metabolism , Chemical and Drug Induced Liver Injury/prevention & control , Isoflavones/therapeutic use , Receptors, Cytoplasmic and Nuclear/metabolism , STAT3 Transcription Factor/metabolism , Animals , Drugs, Chinese Herbal/therapeutic use , Hep G2 Cells , Humans , Male , Mice , Mice, Inbred C57BL , Protective Agents/therapeutic use
19.
Biol Pharm Bull ; 33(8): 1438-42, 2010.
Article in English | MEDLINE | ID: mdl-20686245

ABSTRACT

The effects of Sho-seiryu-to (TJ-19), an ethical Kampo formulation, on bleomycin (BLM)-induced pulmonary fibrosis in rats was examined. Pulmonary fibrosis was induced by intratracheal instillation of a single dose of BLM (5 mg/kg). The TJ-19 used consisted of at least 21 constituents, as determined by three-dimensional HPLC analysis, and was administered orally twice a day at a dose of 1.5 g/kg until the end of the study period. Changes in general appearance and body weight were monitored. Twenty-eight days after BLM instillation, the animals were sacrificed and the study parameters were measured. TJ-19 attenuated the loss in body weight, increase in lung/body weight ratio and concentration of hydroxyproline and malondialdehyde in the lung tissues induced by BLM administration. TJ-19 also prevented BLM-induced fibrotic changes in the lung histology. These protective effects of TJ-19 were observed when administration was started 1 week before and simultaneously with the instillation of BLM. These results suggest that TJ-19 has prophylactic potential against BLM-induced pulmonary fibrosis, and may therefore be a promising drug candidate and medicinal resource for preventing BLM-induced and idiopathic pulmonary fibrosis.


Subject(s)
Drugs, Chinese Herbal/therapeutic use , Idiopathic Pulmonary Fibrosis/prevention & control , Lung/drug effects , Medicine, Kampo , Animals , Bleomycin , Chromatography, High Pressure Liquid , Collagen/metabolism , Disease Models, Animal , Drugs, Chinese Herbal/administration & dosage , Idiopathic Pulmonary Fibrosis/chemically induced , Idiopathic Pulmonary Fibrosis/metabolism , Idiopathic Pulmonary Fibrosis/pathology , Lipid Peroxidation/drug effects , Lipid Peroxides/metabolism , Lung/metabolism , Lung/pathology , Male , Rats , Rats, Sprague-Dawley
20.
Acta Med Okayama ; 62(5): 319-25, 2008 Oct.
Article in English | MEDLINE | ID: mdl-18985092

ABSTRACT

The vascular effects of an aqueous extract prepared from the leaves of Eucommia ulmoides Oliv. (ELE), a medicinal herb commonly used in antihypertensive herbal prescriptions in China, were investigated in rat mesenteric resistance arteries. The mesenteric vascular bed was perfused with Krebs solution and the perfusion pressure was measured with a pressure transducer. In preparations with an intact endothelium and precontracted with 7 microM methoxamine, perfusion of ELE (107102 mg/ml for 15 min) caused a concentration-dependent vasodilatation, which was abolished by chemical removal of the endothelium. The ELE-induced vasodilatation was inhibited by neither indomethacin (INDO, a cyclooxygenase inhibitor) nor NG-nitro-L-arginine-methyl ester (L-NAME, a nitric oxide inhibitor). The ELE-induced vasodilatation was significantly inhibited by tetraethylammonium (TEA, a K channel blocker) and 18alpha-glycyrrhetinic acid (18alpha-GA, a gap-junction inhibitor), and abolished by high K-containing Krebs' solution. Atropine (a muscarinic acetylcholine receptor antagonist) significantly inhibited the vasodilatation induced by ELE at high concentrations. These results suggest that the ELE-induced vasodilatation is endothelium-dependent but nitric oxide (NO)- and prostaglandin I2 (PGI2)-independent, and is mainly mediated by the endothelium-derived hyperpolarizing factor (EDHF) in the mesenteric resistance arteries. Furthermore, the ELE-induced EDHF-mediated response involves the activation of K-channels and gap junctions.


Subject(s)
Biological Factors/metabolism , Drugs, Chinese Herbal/pharmacology , Endothelium, Vascular/metabolism , Eucommiaceae , Mesenteric Arteries/drug effects , Vascular Resistance/drug effects , Vasodilation/drug effects , Acetylcholine/pharmacology , Animals , Atropine/pharmacology , Dose-Response Relationship, Drug , Endothelium, Vascular/drug effects , Enzyme Inhibitors/pharmacology , Indomethacin/pharmacology , Male , Mesenteric Arteries/physiology , NG-Nitroarginine Methyl Ester/pharmacology , Plant Leaves , Potassium Channel Blockers/pharmacology , Rats , Rats, Wistar , Vascular Resistance/physiology , Vasodilation/physiology , Vasodilator Agents/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL