Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 30
Filter
Add more filters










Publication year range
1.
Chem Pharm Bull (Tokyo) ; 67(6): 599-603, 2019.
Article in English | MEDLINE | ID: mdl-31155566

ABSTRACT

The PF-543 is known as a potent and selective inhibitor of sphingosine kinase (SK) 1 amongst all the SK inhibitors known to date. In a recently reported study by Pfizer on the synthesis of PF-543 derivatives and the SK inhibitory effects, the introduction of propyl moiety into sulfonyl group of PF-543 in the case of 26b revealed an excellent result of 1.7 nM of IC50 of SK1, suggesting the potential substitution of chain structure for benzenesulfonyl structure. In the present work, we aimed for identification of antitumor activity and inhibitory effects of PF-543 derivative containing aliphatic long chain (similar to known SK inhibitors) on SK1. The synthesized compound 2 exhibited an inhibitory effect on SK1 in a manner similar to that of PF-543; the PF-543 derivative manifested similar antitumor activity on HT29, HCT116 (colorectal cancer cell line), and AGS (gastric cancer cell line) cells. Also, from the docking study conducted with PF-543 and compound 2, it was apparent that the aliphatic chain in compound 2 could probably replace benzenesulfonyl structure of PF-543.


Subject(s)
Antineoplastic Agents/chemical synthesis , Pyrrolidines/chemistry , Sulfones/chemistry , Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacology , Binding Sites , Catalytic Domain , Cell Line, Tumor , Cell Survival/drug effects , Humans , Inhibitory Concentration 50 , Methanol , Molecular Docking Simulation , Phosphotransferases (Alcohol Group Acceptor)/antagonists & inhibitors , Phosphotransferases (Alcohol Group Acceptor)/metabolism , Pyrrolidines/chemical synthesis , Pyrrolidines/pharmacology , Structure-Activity Relationship , Sulfones/chemical synthesis , Sulfones/pharmacology
2.
J Med Food ; 22(6): 602-613, 2019 Jun.
Article in English | MEDLINE | ID: mdl-31045470

ABSTRACT

The fruits, leaves, and roots of Cudrania tricuspidata have been reported to contain large amounts of vitamin B, vitamin C, and flavonoids. They exhibit various physiological activities such as antitumor and anti-inflammatory effects. However, the hepatoprotective effects of C. tricuspidata extracts against oxidative stress-mediated liver injury have not yet been investigated. We thus examined whether C. tricuspidata leaf extracts (CTEs) protect against oxidative stress-mediated liver injury in vitro and in vivo and elucidated the underlying mechanism. The cytoprotective effects of CTE through the NF-E2-related factor 2 (Nrf2)/antioxidant response element (ARE) activation were presented and measured by biochemical analysis in HepG2 cells. To assess the protective effects of CTE in vivo, mice were administered with CTE (250 and 500 mg/kg; 5 days; p.o.) before a single dose of acetaminophen (APAP) (300 mg/kg; 24 h; i.p.). CTE increased ARE luciferase activity when compared with extracts of other parts of C. tricuspidata. CTE upregulated nuclear translocation of Nrf2 and its target gene expression. In addition, CTE inhibited the generation of reactive oxygen species (ROS) and cell death induced by arachidonic acid (AA) and iron (Fe) treatment in primary hepatocytes or HepG2 cells. The cytoprotective effects of CTE against oxidative stress might be due to kaempferol, the major flavonoid present in CTE. Kaempferol pretreatment blocked AA+Fe-induced ROS production and reversed glutathione depletion, which in turn led to decreased cell death. Furthermore, the protective effects of CTE against liver injury induced by excess APAP in mice or primary hepatocytes were observed. CTE could be a promising therapeutic candidate against oxidative stress-induced liver injury.


Subject(s)
Liver Diseases/drug therapy , Liver/injuries , Moraceae/chemistry , Plant Extracts/administration & dosage , Animals , Glutathione/metabolism , Hepatocytes/drug effects , Hepatocytes/metabolism , Humans , Kaempferols/administration & dosage , Kaempferols/analysis , Liver/drug effects , Liver/metabolism , Liver Diseases/genetics , Liver Diseases/metabolism , Male , Mice , Mice, Inbred ICR , NF-E2-Related Factor 2/genetics , NF-E2-Related Factor 2/metabolism , Oxidative Stress/drug effects , Plant Extracts/chemistry , Reactive Oxygen Species/metabolism
3.
Antioxid Redox Signal ; 31(3): 243-259, 2019 07 20.
Article in English | MEDLINE | ID: mdl-30909713

ABSTRACT

Aims: Hepatic fibrosis results from chronic liver injury and inflammatory responses. Sestrin 2 (Sesn2), an evolutionarily conserved antioxidant enzyme, reduces the severities of acute hepatitis and metabolic liver diseases. However, the role of Sesn2 in the pathogenesis of liver fibrosis remains obscure. Here, we used cultured hepatic stellate cells (HSCs) and chronic carbon tetrachloride (CCl4) and bile duct ligation (BDL) murine models to investigate the effects of Sesn2 on fibrogenesis. Results: Sesn2 protein and mRNA levels were upregulated in activated primary HSCs, and by increasing transcription, transforming growth factor-ß (TGF-ß) also increased Sesn2 expression in HSCs. Furthermore, Smad activation was primarily initiated by TGF-ß signaling, and Smad3 activation increased Sesn2 luciferase activity. In silico analysis of the 5' upstream region of the Sesn2 gene revealed a putative Smad-binding element (SBE), and its deletion demonstrated that the SBE between -964 and -956 bp within human Sesn2 promoter was critically required for TGF-ß-mediated response. Moreover, ectopic expression of Sesn2 reduced gene expressions associated with HSC activation, and this was accompanied by marked decreases in SBE luciferase activity and Smad phosphorylation. Infection of recombinant adenovirus Sesn2 reduced hepatic injury severity, as evidenced by reductions in CCl4- or BDL-induced alanine aminotransferase and aspartate aminotransferase, and inhibited collagen accumulation. Furthermore, HSC-specific lentiviral delivery of Sesn2 prevented CCl4-induced liver fibrosis. Finally, Sesn2 expression was downregulated in the livers of patients with liver cirrhosis and in mouse models of hepatic fibrosis. Innovation and Conclusion: Our findings suggest that Sesn2 has the potential to inhibit HSC activation and hepatic fibrosis.


Subject(s)
Hepatic Stellate Cells/cytology , Liver Cirrhosis/metabolism , Nuclear Proteins/genetics , Nuclear Proteins/metabolism , Smad3 Protein/metabolism , Animals , Binding Sites , Carbon Tetrachloride/adverse effects , Cells, Cultured , Disease Models, Animal , Down-Regulation , Hepatic Stellate Cells/drug effects , Hepatic Stellate Cells/metabolism , Humans , Liver Cirrhosis/genetics , Male , Mice , Nuclear Proteins/chemistry , Promoter Regions, Genetic , Transforming Growth Factor beta/pharmacology
4.
Chem Pharm Bull (Tokyo) ; 66(10): 1015-1018, 2018.
Article in English | MEDLINE | ID: mdl-30270236

ABSTRACT

FTY720 is employed for the treatment of multiple sclerosis and exerts apoptotic effects on various cancers through protein phosphatase 2A (PP2A) activation. In compound 4, the dihydroxy head group of FTY720 was modified into dihydroxy phenyl group. The cell survival in compound 4 treated colorectal and gastric cancer cells was significantly reduced as compared with control, 34.6 and 25.1%, respectively. The docking study of compound 4 showed that the aromatic head group effectively binds to PP2A.


Subject(s)
Antineoplastic Agents/pharmacology , Fingolimod Hydrochloride/pharmacology , Antineoplastic Agents/chemical synthesis , Antineoplastic Agents/chemistry , Cell Proliferation/drug effects , Cell Survival/drug effects , Dose-Response Relationship, Drug , Drug Screening Assays, Antitumor , Fingolimod Hydrochloride/chemical synthesis , Fingolimod Hydrochloride/chemistry , Humans , Molecular Docking Simulation , Molecular Structure , Structure-Activity Relationship , Tumor Cells, Cultured
5.
Molecules ; 23(11)2018 Oct 24.
Article in English | MEDLINE | ID: mdl-30355990

ABSTRACT

FTY720 inhibits various cancers through PP2A activation. The structure of FTY720 is also used as a basic structure for the design of sphingosine kinase (SK) inhibitors. We have synthesized derivatives using an amide chain in FTY720 with a phenyl backbone, and then compounds were screened by an MTT cell viability assay. The PP2A activity of compound 7 was examined. The phosphorylation levels of AKT and ERK, downstream targets of PP2A, in the presence of compound 7, were determined. Compound 7 may exhibit anticancer effects through PP2A activation rather than the mechanism by inhibition of SK1 in cancer cells. In the docking study of compound 7 and PP2A, the amide chain of compound 7 showed an interaction with Asn61 that was different from FTY720, which is expected to affect the activity of the compound.


Subject(s)
Antineoplastic Agents/chemical synthesis , Antineoplastic Agents/pharmacology , Enzyme Inhibitors/chemical synthesis , Enzyme Inhibitors/pharmacology , Fingolimod Hydrochloride/chemical synthesis , Fingolimod Hydrochloride/pharmacology , Protein Phosphatase 2/antagonists & inhibitors , Cell Line, Tumor , Cell Survival/drug effects , Extracellular Signal-Regulated MAP Kinases/metabolism , Fingolimod Hydrochloride/analogs & derivatives , Humans , Inhibitory Concentration 50 , Models, Molecular , Molecular Conformation , Molecular Structure , Phosphorylation , Proto-Oncogene Proteins c-akt/metabolism , Structure-Activity Relationship
6.
Toxicol Res ; 33(3): 219-224, 2017 Jul.
Article in English | MEDLINE | ID: mdl-28744353

ABSTRACT

Lipin1 was identified as a phosphatidate phosphatase enzyme, and it plays a key role in lipid metabolism. Since free radicals contribute to metabolic diseases in the liver, this study investigated the effects of free radicals on the regulation of Lipin1 expression in Huh7 and AML12 cells. Hydrogen peroxide induced mRNA and protein expression of Lipin1 in Huh7 cells, which was assayed by quantitative RT-PCR and immunoblotting, respectively. Induction of Lipin1 by hydrogen peroxide was confirmed in AML12 cells. Hydrogen peroxide treatment significantly increased expression of sterol regulatory element-binding protein (SREBP)-2, but not SREBP-1. Moreover, nuclear translocation of SREBP-2 was detected after hydrogen peroxide treatment. Hydrogen peroxide-induced Lipin1 or SREBP-2 expression was significantly reduced by N-acetyl-l-cysteine treatment, indicating that reactive oxygen species (ROS) were implicated in Lipin1 expression. Next, we investigated whether the hypoxic environments that cause endogenous ROS production in mitochondria in metabolic diseases affect the expression of Lipin1. Exposure to hypoxia also increased Lipin1 expression. In contrast, pretreatment with antioxidants attenuated hypoxia-induced Lipin1 expression. Collectively, our results show that ROS activate SREBP-2, which induces Lipin1 expression.

7.
Arch Pharm Res ; 40(2): 231-239, 2017 Feb.
Article in English | MEDLINE | ID: mdl-28028695

ABSTRACT

5-Fluorouracil (5-FU) is a chemotherapeutic agent used in the treatment of colorectal cancer. In this study, we investigated whether 5-FU induces Sestrin2 (SESN2), an antioxidant enzyme, and the role of SESN2 in 5-FU action in colon cancer cells. We found that 5-FU upregulated SESN2 protein expression in both HCT116 and HT29 cells. It also increased transcripts of SESN1 and SESN2, but not of SESN3. Furthermore, we investigated whether production of reactive oxygen species (ROS) was involved in 5-FU-induced SESN2 expression. 5-FU did not increase ROS production nor affect Nrf2 phosphorylation and expression levels. Moreover, SESN2 upregulation by 5-FU was not prevented by pretreatment with antioxidants. Next, we investigated p53 levels after 5-FU treatment to elucidate the regulation of SESN2 by 5-FU. An increase in p53 levels was detected following 5-FU treatment; pifithrin-α, an inhibitor of p53 activation, reversed 5-FU-induced SESN2 expression. 5-FU prevented serum-induced in vitro cell migration, but knockdown of SESN2 or treatment with pifithrin-α reversed a 5-FU-mediated decrease in cell migration. Taken together, our results suggest that 5-FU increases SESN2 levels via a p53-dependent pathway, which contributes to inhibition of cancer cell migration in vitro.


Subject(s)
Antimetabolites, Antineoplastic/pharmacology , Cell Movement/drug effects , Colorectal Neoplasms/metabolism , Fluorouracil/pharmacology , Nuclear Proteins/metabolism , Tumor Suppressor Protein p53/metabolism , Antioxidants/pharmacology , Benzothiazoles/pharmacology , Gene Knockdown Techniques , HCT116 Cells , HT29 Cells , Heat-Shock Proteins/genetics , Humans , NF-E2-Related Factor 2/metabolism , Nuclear Proteins/genetics , Phosphorylation , RNA Interference , RNA, Small Interfering , Reactive Oxygen Species/metabolism , Signal Transduction , Toluene/analogs & derivatives , Toluene/pharmacology , Transcriptional Activation , Tumor Suppressor Protein p53/antagonists & inhibitors , Up-Regulation
8.
Free Radic Biol Med ; 101: 511-523, 2016 12.
Article in English | MEDLINE | ID: mdl-27840318

ABSTRACT

Sestrin2 (SESN2) is an antioxidant protein that modulates cellular redox homeostasis through regeneration of peroxiredoxins. It has beneficial effects in oxidative or metabolic stress conditions as an upstream regulator of AMP-activated protein kinase (AMPK). Since hypoxia causes oxidative and metabolic stress, this study investigated the effect of SESN2 on signaling pathways altered by hypoxia in colon cancer cells. SESN2 overexpression in HEK293 cells inhibited hypoxia-inducible factor-1α (HIF-1α), which plays a crucial role in tumor growth and development in hypoxia. Moreover, infection with adenovirus-SESN2 (Ad-SESN2) decreased hypoxia or CoCl2-induced HIF-1α accumulation in colorectal cancer cells. Ad-SESN2 also reduced CoCl2-induced hypoxia response element (HRE)-luciferase activity and mRNA level of HIF-1α-driven genes. Furthermore, Ad-SESN2 infected cells showed anti-metastatic effects in serum-induced cell migration and invasion in vitro. Ad-SESN2 facilitated the ubiquitination of HIF-1α protein and increased hydroxyl-HIF-1α (OH-HIF-1α) level. In contrast, treatment with dimethyloxalylglycine (DMOG), an inhibitor of prolyl hydroxylase (PHD), reversed Ad-SESN2-induced OH-HIF-1α and subsequently suppressed HIF-1α level. The inhibitory effects of SESN2 on the serum-induced in vitro cell migration and invasion were also abrogated by DMOG treatment. Furthermore, knockdown of AMPKα reversed Ad-SESN2-mediated increase of OH-HIF-1α and inhibition of HIF-1α. Dominant-negative form of AMPK also restored the Ad-SESN2 mediated decrease in HIF-1α accumulation. Lastly, Ad-SESN2 suppressed tumor growth in a mouse xenograft model. Taken together, these results suggest that SESN2 increases degradation of HIF-1α via AMPK-PHD regulation that contributes to inhibition of in vitro and in vivo tumorigenesis.


Subject(s)
AMP-Activated Protein Kinases/genetics , Colorectal Neoplasms/therapy , Gene Expression Regulation, Neoplastic , Hypoxia-Inducible Factor 1, alpha Subunit/genetics , Nuclear Proteins/genetics , Prolyl Hydroxylases/genetics , AMP-Activated Protein Kinases/metabolism , Adenoviridae/genetics , Adenoviridae/metabolism , Amino Acids, Dicarboxylic/pharmacology , Animals , Cell Hypoxia/genetics , Cell Movement/drug effects , Colorectal Neoplasms/metabolism , Colorectal Neoplasms/pathology , Genetic Vectors/chemistry , Genetic Vectors/metabolism , HCT116 Cells , HEK293 Cells , HT29 Cells , Humans , Hydroxylation , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Mice , Mice, Inbred BALB C , Neoplasm Transplantation , Nuclear Proteins/metabolism , Prolyl Hydroxylases/metabolism , Response Elements , Signal Transduction , Ubiquitination
9.
Biol Pharm Bull ; 39(11): 1830-1838, 2016.
Article in English | MEDLINE | ID: mdl-27803454

ABSTRACT

Isorhamnetin is a flavonoid metabolite of quercetin and isolated from water dropwort (Oenanthe javanica, Umbelliferae). It has been reported that isorhamnetin exerts beneficial effects including antioxidant, anti-inflammatory, and anti-proliferative activities. The present study investigated whether the antioxidant activity of isorhamnetin is correlated with its anti-cancer effects on colorectal cancer cells. Isorhamnetin significantly repressed cobalt chloride (CoCl2)- or hypoxia-induced hypoxia inducible factor-1α (HIF-1α) accumulation in HCT116 and HT29 cells. When compared with quercetin, isorhamnetin showed potent inhibition of HIF-1α. Moreover, it inhibited CoCl2-induced activity of hypoxia response element reporter gene and HIF-1α-dependent transcription of genes such as glucose transporter 1, lactate dehydrogenase A, carbonic anhydrase-IX, and pyruvate dehydrogenase kinase 1. Isorhamnetin also blocked hydrogen peroxide (H2O2)-induced HIF-1α accumulation. The antioxidant effects of isorhamnetin were confirmed by observation of CoCl2- or H2O2-induced reactive oxygen species (ROS) production. Consistently, overexpressed HIF-1α was decreased by isorhamnetin or N-acetyl-L-cysteine in HEK293 cells. In vitro migration and invasion assay further confirmed the inhibitory effects of isorhamnetin on cancer cells. Collectively, these results demonstrate that isorhamnetin inhibits ROS-mediated HIF-1α accumulation, which contributes to its anti-metastatic efficacy.


Subject(s)
Antineoplastic Agents/pharmacology , Hypoxia-Inducible Factor 1, alpha Subunit/antagonists & inhibitors , Quercetin/analogs & derivatives , Cell Movement/drug effects , Cobalt , HCT116 Cells , HEK293 Cells , HT29 Cells , Humans , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Quercetin/pharmacology , Reactive Oxygen Species/metabolism
10.
Toxicol Sci ; 153(1): 28-38, 2016 09.
Article in English | MEDLINE | ID: mdl-27345520

ABSTRACT

The adipogenic transcriptional regulation was reported to inhibit transdifferentiation of hepatic stellate cells (HSCs), which constitute the main fibrogenic cell type in the liver. Lipin-1 exhibits a dual function: an enzyme that catalyzes the conversion of phosphatidate to diacylglycerol and a transcriptional regulator. However, the involvement of Lipin-1 in the regulation of transforming growth factor-ß (TGF-ß) signaling and fibrogenesis in HSCs is not fully understood. Here, we showed that Lipin-1 was downregulated in activated primary HSCs and TGF-ß-treated LX-2 cells, immortalized human HSC cell lines. The downregulation of Lipin-1 by TGF-ß was not dependent on altered mRNA stability but rather on protein stability. Treatment of LX-2 cells with the proteasome inhibitor led to the accumulation of Lipin-1. Moreover, we observed a significant increase in Lipin-1 polyubiquitination. Overexpression of Lipin-1 attenuated TGF-ß-induced fibrogenic gene expression. In addition, Lipin-1 inhibited TGF-ß-mediated activation of Sma and Mad-related family (SMAD), a major transcription factor that transduces intracellular signals from TGF-ß. Resveratrol, a well-known natural polyphenolic antioxidant, is known to inhibit liver fibrosis, although its mechanism of action remains unknown. Our data showed that resveratrol significantly increased the levels of Lipin-1 protein and mRNA in HSCs. Further investigation revealed that resveratrol blocked the polyubiquitination of Lipin-1. Resveratrol inhibited TGF-ß-induced fibrogenic gene expression. TGF-ß-induced SMAD binding element-luciferase reporter activity was significantly diminished by resveratrol with a simultaneous decrease in SMAD3 phosphorylation. Consistently, knockdown of the Lipin-1 gene using siRNA abolished the inhibitory effect of resveratrol. We conclude that Lipin-1 can antagonize HSC activation through the inhibition of TGF-ß/SMAD signaling and that resveratrol may affect Lipin-1 gene induction and contribute to the inhibition of TGF-ß-mediated hepatic fibrogenesis.


Subject(s)
Hepatic Stellate Cells/metabolism , Liver Cirrhosis/physiopathology , Phosphatidate Phosphatase/physiology , Signal Transduction , Transforming Growth Factor beta/metabolism , Cell Line, Transformed , Hepatic Stellate Cells/pathology , Humans , Liver Cirrhosis/metabolism , Phosphatidate Phosphatase/metabolism , Resveratrol , Stilbenes/pharmacology
11.
Biol Pharm Bull ; 39(5): 799-806, 2016.
Article in English | MEDLINE | ID: mdl-27150150

ABSTRACT

Compound C is a widely used chemical inhibitor that down-regulates AMP-activated protein kinase (AMPK) activity. However, it has been suggested that compound C exerts AMPK-independent effects in various cells. Here, we investigated whether compound C induces Sestrin2 (SESN2), an antioxidant enzyme induced by diverse stress. In addition, the mechanism responsible for SESN2 induction by compound C was determined. Our results showed that compound C increased SESN2 protein expression in HepG2 cells in a concentration- and time-dependent manner. The induction of SESN2 mRNA was also observed in cells treated with compound C. Increase of SESN2 luciferase activity confirmed transcriptional regulation by compound C and this substance also increased nuclear factor erythroid 2 (NF-E2)-related factor-2 (Nrf2) phosphorylation, which implies that Nrf2 was involved in SESN2 induction. Next, we sought to demonstrate whether production of reactive oxygen species (ROS) accompanied SESN2 expression. Compound C increased ROS production, but this effect was prevented by pretreatment with antioxidants or the mitochondrial complex I inhibitor. Moreover, cyclosporin A, an inhibitor of pore formation in the mitochondrial membrane, attenuated compound C-induced SESN2 induction. However, overexpression of a constitutively active form of AMPK was not able to abolish SESN2 induction by compound C, which implies that its action is independent of AMPK inhibition. In conclusion, this is the first study demonstrating that compound C alters mitochondrial function and induces ROS production, which ultimately leads to phosphorylation of Nrf2 and induction of SESN2.


Subject(s)
AMP-Activated Protein Kinases/antagonists & inhibitors , Mitochondria/drug effects , Nuclear Proteins/metabolism , Pyrazoles/pharmacology , Pyrimidines/pharmacology , Reactive Oxygen Species/metabolism , Hep G2 Cells , Humans , Luciferases/genetics , Luciferases/metabolism , Membrane Potential, Mitochondrial/drug effects , Mitochondria/metabolism , Mitochondria/physiology , Nuclear Proteins/genetics , RNA, Messenger/metabolism
12.
Biol Pharm Bull ; 38(7): 966-70, 2015.
Article in English | MEDLINE | ID: mdl-26133704

ABSTRACT

Sestrin2 (Sesn2), a highly conserved antioxidant protein, is induced by various stresses, including oxidative and energetic stress, and protects cells against those stresses. In normal physiological conditions, redox-homeostasis plays an essential role in cell survival and performs the cellular functions to protect the cells against oxidative damage. The liver is susceptible to oxidative stress, since it is responsible for xenobiotic detoxification and energy metabolism. For this reason, oxidative stress is associated with the pathogenesis of liver diseases. Recently, the role of Sesn2 has been investigated in liver injury and related diseases. In this paper, we review the role of Sesn2 in the pathophysiology of liver diseases and the potential clinical applications of Sesn2 as a therapeutic target to prevent/treat liver diseases. This article promotes our understanding of liver disease progression and advances the development of strategies for pharmacological intervention.


Subject(s)
Liver Diseases/metabolism , Nuclear Proteins/metabolism , Animals , Homeostasis , Humans , Hydrogen Peroxide/metabolism , Oxidation-Reduction , Signal Transduction
13.
Cell Signal ; 27(7): 1533-43, 2015 Jul.
Article in English | MEDLINE | ID: mdl-25778901

ABSTRACT

Sestrin2 (SESN2) regulates redox-homeostasis and apoptosis in response to various stresses. Although the antioxidant effects of SESN2 have been well established, the roles of SESN2 in mitochondrial function and metabolic stress have not yet been elucidated. In this study, we investigated the role of SESN2 in mitochondrial dysfunction under glucose deprivation and related signaling mechanisms. Glucose deprivation significantly upregulated SESN2 expression in hepatocyte-derived cells. Antioxidant treatments repressed SESN2 induction under glucose deprivation, this result suggested that reactive oxygen species (ROS) production was involved in SESN2 induction. Moreover, NF-E2-related factor-2 (Nrf2) phosphorylation was accompanied in induction of SESN2 by glucose deprivation. To elucidate the functional role of SESN2, we examined cells that stably overexpressed SESN2. Overexpression of SESN2 inhibited glucose deprivation-induced ROS production and cell death. In addition, under glucose deprivation, the changes in mitochondrial membrane potential, ADP/ATP ratio, and mitochondrial DNA content were significantly restored in SESN2-overexpressing cells. Moreover, siRNA knockdown of SESN2 failed to prevent mitochondrial permeability transition by glucose depletion. Mechanistic investigation showed that glucose deprivation significantly increased AMP-activated protein kinase (AMPK) activation. The recovery of mitochondrial function under glucose deprivation in SESN2-overexpressing cells was not seen in SESN2-overexpressing cells transfected with a dominant-negative AMPK; this result suggested that AMPK activation was responsible for SESN2-mediated mitochondrial protection against glucose deprivation. Treatment with 5-aminoimidazole-4-carboxamide-1-ß-D-ribofuranoside (AICAR, an AMPK activator) also provided cytoprotective effects against glucose deprivation. Our findings provide evidence for the functional importance of SESN2-AMPK activation in the protection of mitochondria and cells against glucose deprivation-induced metabolic stress.


Subject(s)
AMP-Activated Protein Kinases/metabolism , Glucose/pharmacology , Mitochondria/drug effects , Nuclear Proteins/metabolism , Reactive Oxygen Species/metabolism , AMP-Activated Protein Kinases/chemistry , Aminoimidazole Carboxamide/analogs & derivatives , Aminoimidazole Carboxamide/pharmacology , Animals , Apoptosis/drug effects , Cell Membrane Permeability/drug effects , Cells, Cultured , Hep G2 Cells , Hepatocytes/cytology , Hepatocytes/drug effects , Hepatocytes/metabolism , Humans , Hydrogen Peroxide/metabolism , Male , Mice , Mice, Inbred ICR , Mitochondria/metabolism , NF-E2-Related Factor 2/metabolism , Nuclear Proteins/antagonists & inhibitors , Nuclear Proteins/genetics , RNA Interference , RNA, Small Interfering/metabolism , Ribonucleotides/pharmacology , Up-Regulation/drug effects
14.
Toxicol Sci ; 144(2): 425-35, 2015 Apr.
Article in English | MEDLINE | ID: mdl-25637945

ABSTRACT

The Sestrin2 (Sesn2) is an evolutionary conserved enzyme that scavenges reactive oxygen species and regulates autophagy through the AMPK-mTOR pathway. The present study was aimed at determining whether Toll-like receptor (TLR) signaling regulates Sesn2 expression and identifying the underlying molecular mechanism. Lipopolysaccharide (LPS), a representative TLR4 ligand, significantly increased the levels of Sesn2 protein in macrophages. LPS also increased Sesn2 mRNA levels and luciferase reporter activity; however, the mRNA levels of Sesn1 were not affected by LPS. Moreover, treatment of macrophages with other TLR ligands (eg, polyI:C or peptidoglycan) also induced Sesn2 expression. We found that LPS-mediated Sesn2 induction was transcriptionally regulated by AP-1 and Nrf2, and that overexpression of c-Jun or Nrf2 increased Sesn2 protein levels and Sesn2 promoter-driven luciferase reporter activity. Moreover, deletion of the antioxidant response element (ARE) in the Sesn2 promoter or Nrf2 knockout abolished LPS-mediated induction of Sesn2. LPS induced Sesn2 gene expression through p38 and PI3K activation. Surprisingly, treatment with the proteasome inhibitor MG132, but not the lysosomal inhibitor chloroquine, caused Sesn2 to accumulate in the cells. In the presence of MG132, we observed that Sesn2 was ubiquitinated. However, LPS treatment attenuated Sesn2 ubiquitination induced by MG132, which resulted in Sesn2 accumulation. Mice treated with D-galactosamine (Gal)/LPS exhibited enhanced Sesn2 expression in the liver. Moreover, infection with a recombinant adenovirus encoding Sens2 markedly reduced the number of Gal/LPS-induced TUNEL-positive cells. Our results suggest that TLR-mediated Sesn2 induction is dependent on AP-1, Nrf2, and the inhibition of ubiquitin-mediated degradation of Sesn2 and might protect cells against endotoxin toxicity.


Subject(s)
Macrophages/metabolism , NF-E2-Related Factor 2/physiology , Nuclear Proteins/biosynthesis , Proteasome Endopeptidase Complex/metabolism , Toll-Like Receptors/physiology , Transcription Factor AP-1/physiology , Ubiquitin/metabolism , Animals , Cell Line , Gene Expression , Lipopolysaccharides/pharmacology , Macrophages/drug effects , Mice , Nuclear Proteins/genetics , Peroxidases , Phosphatidylinositol 3-Kinases/metabolism , p38 Mitogen-Activated Protein Kinases/metabolism
15.
Free Radic Biol Med ; 78: 156-67, 2015 Jan.
Article in English | MEDLINE | ID: mdl-25463278

ABSTRACT

Sestrins (Sesns) are conserved antioxidant proteins that accumulate in cells in response to various stresses. However, the regulatory roles of Sesn2 in the immune system and in inflammatory responses remain obscure. In the present study, we investigated whether Sesn2 regulates Toll like receptor (TLR)-mediated inflammatory signaling and sought to identify the molecular mechanism responsible. In cells expressing Sesn2, it was found that Sesn2 almost completely inhibited lipopolysaccharide (LPS)-induced NO release and iNOS expression. A gene knockdown experiment confirmed the role of Sesn2 in LPS-activated RAW264.7 cells. Consistently, proinflammatory cytokine (e.g., TNF-α, IL-6, and IL-1ß) release and expression were inhibited in Sesn2-expressing cells. Furthermore, Sesn2 prevented LPS-elicited cell death and ROS production via inhibition of NADPH oxidase. NF-κB and AP-1 are redox-sensitive transcription factors that regulate the expressions of diverse inflammatory genes. Surprisingly, Sesn2 specifically inhibited AP-1 luciferase activity and its DNA binding, but not those of NF-κB. AP-1 inhibition by Sesn2 was found to be due to a lack of JNK, p38, and c-Jun phosphorylation. Next, we investigated whether Sesn2 protects galactosamine (Gal)/LPS-induced liver injury in mice infected with a recombinant adenovirus Sesn2 (Ad-Sesn2). Ad-Sesn2 present less severe hepatic injury as supported by decreases in the ALT, AST, and hepatocyte degeneration. Moreover, Ad-Sesn2 attenuated Gal/LPS-induced proinflammatory gene expression in mice. The study shows that Sesn2 inhibits TLR-induced proinflammatory signaling and protects cells by inhibiting JNK- or p38-mediated c-Jun phosphorylation.


Subject(s)
Inflammation Mediators/metabolism , Inflammation/immunology , Macrophages/immunology , Nuclear Proteins/metabolism , Signal Transduction , Animals , Antioxidants/metabolism , Blotting, Western , Cells, Cultured , Cytokines/genetics , Cytokines/metabolism , Electrophoretic Mobility Shift Assay , Immunoenzyme Techniques , Inflammation/chemically induced , Inflammation/metabolism , Inflammation/pathology , Lipopolysaccharides/adverse effects , Macrophages/cytology , Macrophages/metabolism , Mice , NF-kappa B/genetics , NF-kappa B/metabolism , Nitric Oxide/metabolism , Nuclear Proteins/antagonists & inhibitors , Nuclear Proteins/genetics , Peroxidases , Phosphorylation , RNA, Messenger/genetics , RNA, Small Interfering/genetics , Reactive Oxygen Species/metabolism , Real-Time Polymerase Chain Reaction , Reverse Transcriptase Polymerase Chain Reaction , Transcription Factor AP-1/genetics , Transcription Factor AP-1/metabolism
16.
Toxicol Res ; 30(3): 193-8, 2014 Sep.
Article in English | MEDLINE | ID: mdl-25343013

ABSTRACT

Degradation of glucose is aberrantly increased in hyperglycemia, which causes various harmful effects on the liver. Methylglyoxal is produced during glucose degradation and the levels of methylglyoxal are increased in diabetes patients. In this study we investigated whether methylglyoxal induces mitochondrial impairment and apoptosis in HepG2 cells and induces liver toxicity in vivo. Methylglyoxal caused apoptotic cell death in HepG2 cells. Moreover, methylglyoxal significantly promoted the production of reactive oxygen species (ROS) and depleted glutathione (GSH) content. Pretreatment with antioxidants caused a marked decrease in methylglyoxal-induced apoptosis, indicating that oxidant species are involved in the apoptotic process. Methylglyoxal treatment induced mitochondrial permeability transition, which represents mitochondrial impairment. However, pretreatment with cyclosporin A, an inhibitor of the formation of the permeability transition pore, partially inhibited methylglyoxal-induced cell death. Furthermore, acute treatment of mice with methylglyoxal increased the plasma levels of alanine aminotransferase (ALT) and aspartate aminotransferase (AST), indicating liver toxicity. Collectively, our results showed that methylglyoxal increases cell death and induces liver toxicity, which results from ROS-mediated mitochondrial dysfunction and oxidative stress.

17.
Toxicol Appl Pharmacol ; 280(2): 314-22, 2014 Oct 15.
Article in English | MEDLINE | ID: mdl-25151220

ABSTRACT

Methylglyoxal is found in high levels in the blood and other tissues of diabetic patients and exerts deleterious effects on cells and tissues. Previously, we reported that resveratrol, a polyphenol in grapes, induced the expression of Sestrin2 (SESN2), a novel antioxidant protein, and inhibited hepatic lipogenesis. This study investigated whether resveratrol protects cells from the methylglyoxal-induced toxicity via SESN2 induction. Methylglyoxal significantly induced cell death in HepG2 cells. However, cells pretreated with resveratrol were rescued from methylglyoxal-induced apoptosis. Resveratrol attenuated glutathione (GSH) depletion and ROS production promoted by methylglyoxal. Moreover, mitochondrial damage was observed by methylglyoxal treatment, but resveratrol restored mitochondrial function, as evidenced by the observed lack of mitochondrial permeability transition and increased ADP/ATP ratio. Resveratrol treatment inhibited SESN2 depletion elicited by methylglyoxal. SESN2 overexpression repressed methylglyoxal-induced mitochondrial dysfunction and apoptosis. Likewise, rotenone-induced cytotoxicity was not observed in SESN2 overexpressed cells. Furthermore, siRNA knockdown of SESN2 reduced the ability of resveratrol to prevent methylglyoxal-induced mitochondrial permeability transition. In addition, when mice were exposed to methylglyoxal after infection of Ad-SESN2, the plasma levels of alanine aminotransferase (ALT) and aspartate aminotransferase (AST) and GSH depletion by methylglyoxal in liver was reduced in Ad-SESN2 infected mice. Our results demonstrated that resveratrol is capable of protecting cells from methylglyoxal-induced mitochondrial dysfunction and oxidative stress via SESN2 induction.


Subject(s)
Apoptosis/drug effects , Mitochondria/drug effects , Nuclear Proteins/physiology , Pyruvaldehyde/toxicity , Stilbenes/pharmacology , Adenoviridae/genetics , Animals , Glycation End Products, Advanced/metabolism , Hep G2 Cells , Humans , Male , Mice , Mice, Inbred ICR , Mitochondria/physiology , Nuclear Proteins/genetics , Reactive Oxygen Species/metabolism , Resveratrol
18.
Inflammation ; 37(3): 712-22, 2014 Jun.
Article in English | MEDLINE | ID: mdl-24337631

ABSTRACT

Previously, we reported that isorhamnentin, a 3'-O-methylated metabolite of quercetin, reduced inducible nitric oxide synthase (iNOS) expression and NO production. The present study further investigated the underlying mechanism of anti-inflammatory and antioxidant effects of isorhamnentin. Administration of isorhamnetin decreased the number of cyclooxygenase-2 (COX-2) positive cells in rats with carrageenan-induced paw edema. Isorhamnetin also suppressed lipopolysaccharide (LPS)-induced expression of COX-2 in cells. It is well known that LPS-induced reactive oxygen species (ROS) production leads to COX-2 induction. Isorhamnetin decreased LPS-induced ROS production and apoptosis. In addition, the basal expression of heme oxygenase-1 (HO-1) was increased by isorhamnetin treatment in agreement with the increase in nuclear translocation of NF-E2-related factor-2 (Nrf2), an essential transcription factor for the regulation of HO-1 expression. Moreover, pretreatment of tin protoporphyrin IX (SnPP), a chemical inhibitor of HO-1, reversed the ability of isothamnetin to inhibit COX-2 expression. These results demonstrate that induction of HO-1 by isorhamnetin leads to a reduction in ROS production and its antioxidant property might contribute to the inhibition of COX-2 expression in response to inflammation.


Subject(s)
Antioxidants/pharmacology , Cyclooxygenase 2 Inhibitors/pharmacology , Cyclooxygenase 2/biosynthesis , Heme Oxygenase (Decyclizing)/metabolism , Quercetin/analogs & derivatives , Active Transport, Cell Nucleus/drug effects , Animals , Anti-Inflammatory Agents/pharmacology , Apoptosis/drug effects , Cell Line , Edema/chemically induced , Edema/drug therapy , Heme Oxygenase (Decyclizing)/antagonists & inhibitors , Heme Oxygenase (Decyclizing)/biosynthesis , Inflammation/immunology , L-Lactate Dehydrogenase/metabolism , Lipopolysaccharides , Macrophages/enzymology , Macrophages/immunology , Metalloporphyrins/pharmacology , Mice , NF-E2-Related Factor 2/metabolism , Nitric Oxide/biosynthesis , Nitric Oxide Synthase Type II/biosynthesis , Protoporphyrins/pharmacology , Quercetin/pharmacology , Rats , Rats, Sprague-Dawley , Reactive Oxygen Species/metabolism
19.
Toxicol Appl Pharmacol ; 274(2): 293-301, 2014 Jan 15.
Article in English | MEDLINE | ID: mdl-24211276

ABSTRACT

Isorhamentin is a 3'-O-methylated metabolite of quercetin, and has been reported to have anti-inflammatory and anti-proliferative effects. However, the effects of isorhamnetin on Nrf2 activation and on the expressions of its downstream genes in hepatocytes have not been elucidated. Here, we investigated whether isorhamnetin has the ability to activate Nrf2 and induce phase II antioxidant enzyme expression, and to determine the protective role of isorhamnetin on oxidative injury in hepatocytes. In HepG2 cells, isorhamnetin increased the nuclear translocation of Nrf2 in a dose- and time-dependent manner, and consistently, increased antioxidant response element (ARE) reporter gene activity and the protein levels of hemeoxygenase (HO-1) and of glutamate cysteine ligase (GCL), which resulted in intracellular GSH level increases. The specific role of Nrf2 in isorhamnetin-induced Nrf2 target gene expression was verified using an ARE-deletion mutant plasmid and Nrf2-knockout MEF cells. Deletion of the ARE in the promoter region of the sestrin2 gene, which is recently identified as the Nrf2 target gene by us, abolished the ability of isorhamnetin to increase luciferase activity. In addition, Nrf2 deficiency completely blocked the ability of isorhamnetin to induce HO-1 and GCL. Furthermore, isorhamnetin pretreatment blocked t-BHP-induced ROS production and reversed GSH depletion by t-BHP and consequently, due to reduced ROS levels, decreased t-BHP-induced cell death. In addition isorhamnetin increased ERK1/2, PKCδ and AMPK phosphorylation. Finally, we showed that Nrf2 deficiency blocked the ability of isorhamnetin to protect cells from injury induced by t-BHP. Taken together, our results demonstrate that isorhamnetin is efficacious in protecting hepatocytes against oxidative stress by Nrf2 activation and in inducing the expressions of its downstream genes.


Subject(s)
Anti-Inflammatory Agents/pharmacology , NF-E2-Related Factor 2/metabolism , Oxidative Stress/drug effects , Quercetin/analogs & derivatives , AMP-Activated Protein Kinases/genetics , AMP-Activated Protein Kinases/metabolism , Antioxidant Response Elements/drug effects , Antioxidants/pharmacology , Cell Proliferation/drug effects , Cell Survival/drug effects , Glutamate-Cysteine Ligase/genetics , Glutamate-Cysteine Ligase/metabolism , Glutathione/metabolism , Heme Oxygenase-1/genetics , Heme Oxygenase-1/metabolism , Hep G2 Cells , Hepatocytes/drug effects , Hepatocytes/metabolism , Humans , MAP Kinase Signaling System/drug effects , NF-E2-Related Factor 2/genetics , Phosphorylation , Protein Kinase C-delta/genetics , Protein Kinase C-delta/metabolism , Quercetin/pharmacology , tert-Butylhydroperoxide/toxicity
20.
Oxid Med Cell Longev ; 2013: 763257, 2013.
Article in English | MEDLINE | ID: mdl-23766860

ABSTRACT

The liver is a central organ that performs a wide range of functions such as detoxification and metabolic homeostasis. Since it is a metabolically active organ, liver is particularly susceptible to oxidative stress. It is well documented that liver diseases including hepatitis, fibrosis, cirrhosis, and hepatocellular carcinoma are highly associated with antioxidant capacity. NF-E2-related factor-2 (Nrf2) is an essential transcription factor that regulates an array of detoxifying and antioxidant defense genes expression in the liver. It is activated in response to electrophiles and induces its target genes by binding to the antioxidant response element (ARE). Therefore, the roles of the Nrf2-ARE pathway in liver diseases have been extensively investigated. Studies from several animal models suggest that the Nrf2-ARE pathway collectively exhibits diverse biological functions against viral hepatitis, alcoholic and nonalcoholic liver disease, fibrosis, and cancer via target gene expression. In this review, we will discuss the role of the Nrf2-ARE pathway in liver pathophysiology and the potential application of Nrf2 as a therapeutic target to prevent and treat liver diseases.


Subject(s)
Antioxidant Response Elements/genetics , Liver Diseases/metabolism , NF-E2-Related Factor 2/metabolism , Signal Transduction , Animals , Humans , Liver Regeneration , Protective Agents/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...