Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 42
Filter
Add more filters










Publication year range
1.
Adv Clin Exp Med ; 28(12): 1609-1614, 2019 Dec.
Article in English | MEDLINE | ID: mdl-31851788

ABSTRACT

BACKGROUND: During cerebral ischemia, energy restoration through the regulation of glucose transporters and antioxidant defense mechanisms is essential to maintain cell viability. Antioxidant therapy has been considered effective to attenuate brain damage; moreover, the regulation of transcription factors that positively regulate the expression of glucose transporters is associated with this therapy. Recently, it has been reported that the use of antioxidants such as S-allylcysteine (SAC), a component of aged garlic extract (AGE), improves survival in experimental models of cerebral ischemia. OBJECTIVES: The aim of this study was to determine the effect of AGE and SAC on the level of mRNA expression of the main neuronal glucose transporter (GLUT3) and the glutamate cysteine ligase catalytic subunit (GCLC) in rats with transient focal cerebral ischemia. MATERIAL AND METHODS: Cerebral ischemia was induced in male Wistar rats by middle cerebral artery occlusion (MCAO) for 2 h. The animals were sacrificed after different reperfusion times (0-48 h). Animals injected with AGE (360 mg/kg, intraperitoneally (i.p.)) and SAC (300 mg/kg, i.p.) at the beginning of reperfusion were sacrificed after 2 h. The mRNA expression level was analyzed in the fronto-parietal cortex using quantitative polymerase chain reaction (qPCR). RESULTS: Two major increases in GLUT3 expression at 1 h and 24 h of reperfusion were found. Both treatments increased GLUT3 and GCLC mRNA levels in control and under ischemic/reperfusion injury animals. CONCLUSIONS: This data suggests that SAC and AGE might induce neuroprotection, while controlling reactive oxygen species (ROS) levels, as indicated by the increase in GCLC expression, and regulating the energy content of the cell by increasing glucose transport mediated by GLUT3.


Subject(s)
Brain Ischemia , Garlic , Glucose Transport Proteins, Facilitative/metabolism , Glutamate-Cysteine Ligase/metabolism , Neuroprotective Agents , Animals , Antioxidants/metabolism , Antioxidants/pharmacology , Brain Ischemia/metabolism , Cysteine/analogs & derivatives , Cysteine/pharmacology , Garlic/chemistry , Glucose Transport Proteins, Facilitative/drug effects , Glutamate-Cysteine Ligase/drug effects , Male , Neuroprotective Agents/therapeutic use , Plant Extracts/pharmacology , Rats , Rats, Wistar , Reperfusion Injury/metabolism
2.
J Biol Chem ; 292(13): 5532-5545, 2017 03 31.
Article in English | MEDLINE | ID: mdl-28202547

ABSTRACT

Neuroinflammation and oxidative stress are hallmarks of various neurological diseases. However, whether and how the redox processes control neuroinflammation is incompletely understood. We hypothesized that increasing cellular glutathione (GSH) levels would inhibit neuroinflammation. A series of thiol compounds were identified to elevate cellular GSH levels by a novel approach (i.e. post-translational activation of glutamate cysteine ligase (GCL), the rate-limiting enzyme in GSH biosynthesis). These small thiol-containing compounds were examined for their ability to increase intracellular GSH levels in a murine microglial cell line (BV2), of which dimercaprol (2,3-dimercapto-1-propanol (DMP)) was found to be the most effective compound. DMP increased GCL activity and decreased LPS-induced production of pro-inflammatory cytokines and inducible nitric-oxide synthase induction in BV2 cells in a concentration-dependent manner. The ability of DMP to elevate GSH levels and attenuate LPS-induced pro-inflammatory cytokine production was inhibited by buthionine sulfoximine, an inhibitor of GCL. DMP increased the expression of GCL holoenzyme without altering the expression of its subunits or Nrf2 target proteins (NQO1 and HO-1), suggesting a post-translational mechanism. DMP attenuated LPS-induced MAPK activation in BV2 cells, suggesting the MAPK pathway as the signaling mechanism underlying the effect of DMP. Finally, the ability of DMP to increase GSH via GCL activation was observed in mixed cerebrocortical cultures and N27 dopaminergic cells. Together, the data demonstrate a novel mechanism of GSH elevation by post-translational activation of GCL. Post-translational activation of GCL offers a novel targeted approach to control inflammation in chronic neuronal disorders associated with impaired adaptive responses.


Subject(s)
Dimercaprol/pharmacology , Glutamate-Cysteine Ligase/metabolism , Inflammation/prevention & control , Animals , Cell Line , Cytokines/antagonists & inhibitors , Enzyme Activation/drug effects , Glutamate-Cysteine Ligase/drug effects , Glutathione/metabolism , MAP Kinase Signaling System , Mice , Nervous System/pathology , Oxidation-Reduction , Rats , Sulfhydryl Compounds/metabolism
3.
Int J Pediatr Otorhinolaryngol ; 92: 61-66, 2017 Jan.
Article in English | MEDLINE | ID: mdl-28012535

ABSTRACT

OBJECTIVE: Cisplatin is commonly used to treat solid tumors. However, permanent hearing loss is a major side effect of cisplatin chemotherapy and often results in dose reduction of the cisplatin chemotherapy. Peanut sprouts show cytoprotective properties owing to their antioxidant activities. This study was designed to investigate the effect of peanut sprout extract (PSE) on cisplatin-induced ototoxicity in an auditory cell line, HEI-OC1 cells. METHODS: Cells were exposed to cisplatin for 24 h, with or without pre-treatment with PSE, cell viability was examined using the MTT assay. Apoptotic cells were identified by double staining with Hoechst 33258 and propidium iodide. Western blot analysis was performed to examine apoptotic proteins including C-PARP and C-caspase, anti-apoptotic protein Bcl-2, and Nrf2 redox system activation. Mitochondrial reactive oxygen species (ROS) were investigated to examine whether PSE could scavenge cisplatin-induced ROS. Real-time PCR analyses were performed to investigate the mRNA levels of antioxidant enzymes including NQO1, HO-1, GPx2, Gclc, and catalase. RESULTS: The cisplatin-treated group showed reduced cell viability, increased apoptotic properties and markers, and increased ROS levels. PSE pre-treatment before cisplatin exposure significantly increased cell viability and reduced apoptotic properties and ROS production. These effects resulted from the up-regulation of antioxidant genes, including NQO1, HO-1, GPx2, Gclc, and catalase through Akt phosphorylation and Nrf2 activation. CONCLUSION: Our results demonstrate that PSE protects from cisplatin-induced cytotoxicity by activating the antioxidant effects via the Akt/Nrf-2 pathway in this auditory cell line, and indicate that PSE may provide novel treatment to prevent cisplatin-induced ototoxicity.


Subject(s)
Antineoplastic Agents/toxicity , Apoptosis/drug effects , Arachis , Cisplatin/toxicity , Hair Cells, Auditory/drug effects , Labyrinth Supporting Cells/drug effects , Plant Extracts/pharmacology , Seedlings , Animals , Blotting, Western , Caspases/drug effects , Caspases/metabolism , Catalase/drug effects , Catalase/genetics , Cell Line , Cell Survival/drug effects , Glutamate-Cysteine Ligase/drug effects , Glutamate-Cysteine Ligase/genetics , Glutathione Peroxidase/drug effects , Glutathione Peroxidase/genetics , Heme Oxygenase-1/drug effects , Heme Oxygenase-1/genetics , In Vitro Techniques , Membrane Proteins/drug effects , Membrane Proteins/genetics , Mice , Mitochondria/drug effects , Mitochondria/metabolism , NAD(P)H Dehydrogenase (Quinone)/drug effects , NAD(P)H Dehydrogenase (Quinone)/genetics , NF-E2-Related Factor 2/drug effects , NF-E2-Related Factor 2/metabolism , Oxidation-Reduction/drug effects , Poly(ADP-ribose) Polymerases/drug effects , Poly(ADP-ribose) Polymerases/metabolism , Proto-Oncogene Proteins c-bcl-2/drug effects , Proto-Oncogene Proteins c-bcl-2/metabolism , Reactive Oxygen Species/metabolism , Real-Time Polymerase Chain Reaction , Up-Regulation
4.
Eur J Pharmacol ; 771: 99-106, 2016 Jan 15.
Article in English | MEDLINE | ID: mdl-26687633

ABSTRACT

Glutathione is a key regulator of oxidative balance in all mammals, especially in the central nervous system. The first step of glutathione synthesis is catalyzed by glutamate-cysteine ligase (GCL), which is composed of catalytic and modifier subunits (GCLC and GCLM, respectively). In non-neural cells and tissues, insulin and insulin-like growth factor 1 (IGF-1) have been found to stimulate transcription of GCLC gene. Here we found that treatment of human neuroblastoma SH-SY5Y cells with insulin or IGF-1 increased mRNA level of GCLM, but not of GCLC, in a concentration- and time-dependent manner. In contrast, insulin did not increase GCL expression in rat C6 glioma cells. We also confirmed that IGF-1 increased protein level of GCLM and cellular glutathione content in SH-SY5Y cells. In addition, IGF-1 increased nuclear factor erythroid 2-related factor 2 (Nrf2) protein in the nuclear fraction of SH-SY5Y cells. siRNA-mediated knockdown of Nrf2 protein expression abrogated IGF-1-induced up-regulation of GCLM mRNA expression. Finally, IGF-1-induced increase in nuclear Nrf2 protein and GCLM mRNA expression was abolished by LY294002, a phosphoinositide 3-kinase inhibitor. These results indicate that insulin and IGF-1 have the ability to enhance glutathione biosynthesis in neuronal cells via specific up-regulation of GCLM expression.


Subject(s)
Glutamate-Cysteine Ligase/biosynthesis , Glutathione/biosynthesis , Insulin-Like Growth Factor I/pharmacology , 3-Phosphoinositide-Dependent Protein Kinases/antagonists & inhibitors , Animals , Cell Line, Tumor , Chromones/pharmacology , Gene Expression/drug effects , Glutamate-Cysteine Ligase/drug effects , Glutamate-Cysteine Ligase/genetics , Humans , Morpholines/pharmacology , NF-E2-Related Factor 2/biosynthesis , NF-E2-Related Factor 2/genetics , Neurons/drug effects , Neurons/metabolism , Neuroprotective Agents/pharmacology , RNA, Small Interfering/pharmacology , Rats , Up-Regulation/drug effects
5.
J Med Assoc Thai ; 97 Suppl 8: S57-63, 2014 Aug.
Article in English | MEDLINE | ID: mdl-25518294

ABSTRACT

BACKGROUND: Cymbopogon citratus, Stapf(CCS) is commonly known as lemon grass. Previous studies showed that it has a strong antioxidant property and have been traditionally used as analgesic, antipyretic, antiseptic in SoutheastAsia. However, the effect of CCS on antioxidant defense system has not been demonstrated. OBJECTIVE: The present study was conducted to investigate the effects of CCS water extract on rat antioxidant defense system, especially on the expression of y-glutamylcysteine ligase (γ-GCL) and heme oxygenase-1 (HO-1). MATERIAL AND METHOD: The CCS water extract was screenedfor its phytochemical contents and antioxidant activity in vitro. Moreover, the extract was studied in rats to evaluate its effects in vivo. Male Sprague-Dawley rats aged eight weeks (250±20 g) were orally administered with CCS at 250, 500 and 1,000 mg/kg/day for one month. RESULTS: The extract contained flavonoids (496.17 mg gallic acid/g CCS extract) and phenolic compounds (4,020.18 mg catechin/g CCS extract). The scavenging activity (DPPH assay) of the extract was demonstrated by EC50 of 917.76±86.89 µg/ ml whereas the EC50 of the potent antioxidant, vitamin C was 31.22±1.84 µg/ml. In the animals, the protein expression of antioxidant enzymes, γGCL and HO-1 was significantly increased in the high dose-treated animals (1,000 mg/kg/day). This was consistent with elevation ofserum total antioxidant capacity. CONCLUSION: Taken together the present study provides evidence that CCS water extract exhibits antioxidant activity and antioxidant enzymes induction in vivo.


Subject(s)
Antioxidants/pharmacology , Cymbopogon , Glutamate-Cysteine Ligase/drug effects , Heme Oxygenase-1/drug effects , Plant Extracts/pharmacology , Animals , Ascorbic Acid/pharmacology , Flavonoids/pharmacology , Glutamate-Cysteine Ligase/metabolism , Heme Oxygenase-1/metabolism , Male , Phenols/pharmacology , Rats , Rats, Sprague-Dawley
6.
J. pediatr. (Rio J.) ; 90(5): 493-499, Sep-Oct/2014. graf
Article in English | LILACS | ID: lil-723171

ABSTRACT

Objective: To explore the effect of erythromycin on hyperoxia-induced lung injury. Methods: One-day-old preterm offspring Sprague-Dawley (SD) rats were randomly divided into four groups: group 1, air + sodium chloride; group 2, air + erythromycin;group 3, hyperoxia + sodium chloride; and group 4, hyperoxia + erythromycin. At one, seven, and 14 days of exposure, glutathione (GSH) and interleukin-1 beta (IL-1 beta) were detected by double-antibody sandwich enzyme-linked immunosorbent assay (ELISA), and bicinchoninic acid (BCA) was used to detect GSH protein. γ-glutamine-cysteine synthetase (γ-GCS) mRNA was detected by reverse transcription-polymerase chain reaction (RT-PCR). Results: Compared with group 1, expressions of GSH and γ-GCS mRNA in group 3 were significantly increased at one and seven days of exposure (p < 0.05), but expression of γ-GCS mRNA was significantly reduced at 14 days; expression of IL-1 beta in group 3 was significantly increased at seven days of exposure (p < 0.05), and was significantly reduced at 14 days. Compared with group 3, expressions of GSH and γ-GCS mRNA in group 4 were significantly increased at one, seven, and 14 days of exposure (p < 0.05), but expressions of GSH showed a downward trend at 14 days; expression of IL-1 beta in group 4 was significantly reduced at one and seven days of exposure (p < 0.05). Conclusions: Changes in oxidant-mediated IL-1 beta and GSH are involved in the development of hyperoxia-induced lung injury. Erythromycin may up-regulate the activity of γ-GCS, increasing the expression of GSH, inhibiting the levels of oxidant-mediated IL-1 beta and alleviating hyperoxia-induced lung injury via an antioxidant effect. .


Objetivo: Explorar o efeito da eritromicina sobre lesões pulmonares induzidas por hiperóxia. Métodos: Uma prole de ratos Sprague-Dawley (SD) prematuros com um dia de vida foi dividida aleatoriamente em quatro grupos: grupo 1 ar + cloreto de sódio, grupo 2 ar + eritromicina, grupo 3 hiperóxia + cloreto de sódio e grupo 4 hiperóxia + eritromicina. Com um, sete e 14 dias de exposição, foram detectadas Glutationa (GSH) e Interleucina-1 beta (IL-1 beta) pelo ensaio imunossorvente ligado à enzima (ELISA), e o ácido bicinconinico (BCA) foi utilizado para detectar a proteína GSH. O mRNA da γ-glutamil-cisteina-sintetase (γ-GCS) foi detectado por reação em cadeia da polimerase via transcriptase reversa (RT-PCR). Resultados: Comparadas ao grupo 1, as expressões do mRNA da GSH e da γ-GCS no grupo 3 aumentaram significativamente com um e sete dias de exposição (p < 0,05), porém a expressão de mRNA da γ-GCS diminuiu significativamente aos 14 dias; a expressão de IL-1 beta no grupo 3 aumentou significativamente aos 7 dias de exposição (p < 0,05) e diminuiu significativamente aos 14 dias. Comparadas ao grupo 3, as expressões do mRNA da GSH e da γ-GCS no grupo 4 aumentaram significativamente com um, sete e 14 dias de exposição (p < 0,05), porém as expressões de GSH mostraram uma tendência de queda aos 14 dias; a expressão de IL-1 beta no grupo 4 foi reduzida significativamente com um e sete dias de exposição (p < 0,05). Conclusões: As variações de IL-1 beta e GSH mediadas por oxidantes estão envolvidas no desenvolvimento de lesão pulmonar induzida por hiperóxia. A eritromicina poderá regular positivamente a atividade da γ-GCS, aumentando a expressão de GSH, inibindo os níveis de interleucina-1beta mediada por ...


Subject(s)
Animals , Female , Male , Erythromycin/pharmacology , Glutamate-Cysteine Ligase/drug effects , Hyperoxia/metabolism , Interleukin-1beta/drug effects , Lung/drug effects , Protein Synthesis Inhibitors/pharmacology , Animals, Newborn , Disease Models, Animal , Enzyme-Linked Immunosorbent Assay , Glutathione/metabolism , Interleukin-1beta/metabolism , Lung Injury/metabolism , Oxygen/metabolism , Oxygen/pharmacology , Protein Synthesis Inhibitors/metabolism , Random Allocation , Rats, Sprague-Dawley , Reverse Transcriptase Polymerase Chain Reaction
7.
J Pediatr (Rio J) ; 90(5): 493-9, 2014.
Article in English | MEDLINE | ID: mdl-24878007

ABSTRACT

OBJECTIVE: To explore the effect of erythromycin on hyperoxia-induced lung injury. METHODS: One-day-old preterm offspring Sprague-Dawley (SD) rats were randomly divided into four groups: group 1, air + sodium chloride; group 2, air + erythromycin;group 3, hyperoxia + sodium chloride; and group 4, hyperoxia + erythromycin. At one, seven, and 14 days of exposure, glutathione (GSH) and interleukin-1 beta (IL-1 beta) were detected by double-antibody sandwich enzyme-linked immunosorbent assay (ELISA), and bicinchoninic acid (BCA) was used to detect GSH protein. γ-glutamine-cysteine synthetase (γ-GCS) mRNA was detected by reverse transcription-polymerase chain reaction (RT-PCR). RESULTS: Compared with group 1, expressions of GSH and γ-GCS mRNA in group 3 were significantly increased at one and seven days of exposure (p < 0.05), but expression of γ-GCS mRNA was significantly reduced at 14 days; expression of IL-1 beta in group 3 was significantly increased at seven days of exposure (p < 0.05), and was significantly reduced at 14 days. Compared with group 3, expressions of GSH and γ-GCS mRNA in group 4 were significantly increased at one, seven, and 14 days of exposure (p < 0.05), but expressions of GSH showed a downward trend at 14 days; expression of IL-1 beta in group 4 was significantly reduced at one and seven days of exposure (p < 0.05). CONCLUSIONS: Changes in oxidant-mediated IL-1 beta and GSH are involved in the development of hyperoxia-induced lung injury. Erythromycin may up-regulate the activity of γ-GCS, increasing the expression of GSH, inhibiting the levels of oxidant-mediated IL-1 beta and alleviating hyperoxia-induced lung injury via an antioxidant effect.


Subject(s)
Erythromycin/pharmacology , Glutamate-Cysteine Ligase/drug effects , Hyperoxia/metabolism , Interleukin-1beta/drug effects , Lung/drug effects , Protein Synthesis Inhibitors/pharmacology , Animals , Animals, Newborn , Disease Models, Animal , Enzyme-Linked Immunosorbent Assay , Female , Glutathione/metabolism , Interleukin-1beta/metabolism , Lung Injury/metabolism , Male , Oxygen/metabolism , Oxygen/pharmacology , Protein Synthesis Inhibitors/metabolism , Random Allocation , Rats, Sprague-Dawley , Reverse Transcriptase Polymerase Chain Reaction
8.
J Nutr Biochem ; 24(11): 1911-9, 2013 Nov.
Article in English | MEDLINE | ID: mdl-24011717

ABSTRACT

Chlorogenic acid (CGA) is one of the most abundant dietary polyphenols, possessing well-known antioxidant capacity. The present study is designed to observe the protection provided by CGA against acetaminophen (AP)-induced liver injury in mice in vivo and the underlying mechanisms engaged in this process. Serum transaminases analysis and liver histological evaluation demonstrated the protection of CGA against AP-induced liver injury. CGA treatment decreased the increased number of liver apoptotic cells induced by AP in a dose-dependent manner. CGA also inhibited AP-induced cleaved activation of caspase-3, 7. Moreover, CGA reversed AP-decreased liver reduced glutathione (GSH) levels, glutamate-cysteine ligase (GCL) and glutathione reductase activity. Further results showed that CGA increased mRNA and protein expression of the catalytic subunit of GCL (GCLC), thioredoxin (Trx) 1/2 and thioredoxin reductase (TrxR) 1. Furthermore, CGA abrogated AP-induced phospholyated activation of ERK1/2, c-Jun N-terminal kinase (JNK), p38 kinases and molecular signals upstream. The results of this study demonstrate that CGA counteracts AP-induced liver injury at various levels by preventing apoptosis and oxidative stress damage, and more specifically, both the GSH and Trx antioxidant systems and the mitogen-activated protein kinase (MAPK) signaling cascade appear to be engaged in this protective mechanism.


Subject(s)
Acetaminophen/toxicity , Chemical and Drug Induced Liver Injury/prevention & control , Chlorogenic Acid/therapeutic use , Animals , Apoptosis/drug effects , Caspase 3/metabolism , Glutamate-Cysteine Ligase/drug effects , Glutathione/drug effects , Glutathione/metabolism , Liver/pathology , Male , Mice , Mice, Inbred ICR , Mitogen-Activated Protein Kinases/metabolism
9.
Drug Discov Ther ; 7(2): 78-83, 2013 Apr.
Article in English | MEDLINE | ID: mdl-23715506

ABSTRACT

Acetaminophen (AP) is widely used as the antipyretic and analgesic drug in clinic, and it can induce serious liver injury in the case of excessive abuse. The present study showed that AP (400 mg/kg) induced obvious liver injury, while in male mice the hepatotoxicity induced by AP was much more serious than in female mice as indicated by the results of alanine aminotransferase (ALT) activity and reduced glutathione (GSH) amount. Further, the enzymatic activity and protein expression of glutamate-cysteine ligase (GCL) and glutathione peroxidase (GPx) were all higher in female mice liver than in male after the administration of AP (200 mg/kg). Meanwhile, AP (10 mM) decreased GCL and GPx activity in isolated mouse hepatocytes in the time-dependent manner, while the inhibitors of GCL and GPx can augment AP induced-cytotoxicity. Taken together, our results demonstrate the gender-related liver injury induced by AP and the important role of GCL and GPx in regulating such hepatotoxicity.


Subject(s)
Acetaminophen/toxicity , Analgesics, Non-Narcotic/toxicity , Chemical and Drug Induced Liver Injury/metabolism , Glutamate-Cysteine Ligase/drug effects , Glutathione Peroxidase/drug effects , Liver/metabolism , Animals , Chemical and Drug Induced Liver Injury/etiology , Female , Glutamate-Cysteine Ligase/metabolism , Glutathione/metabolism , Glutathione Peroxidase/metabolism , Liver/drug effects , Male , Mice , Mice, Inbred ICR , Sex Factors
10.
J Periodontol ; 80(8): 1281-8, 2009 Aug.
Article in English | MEDLINE | ID: mdl-19656028

ABSTRACT

BACKGROUND: This study examined the effects of nicotine on osteoblastic differentiation and the osteoclastogenesis regulatory molecules receptor activator of nuclear factor-kappa B ligand (RANKL) and osteoprotegerin (OPG). In addition, we investigated the mechanism by which nicotine induced antioxidant defense enzyme expression as a protective response. METHODS: The expression of osteoblast markers, RANKL, OPG, and antioxidant defense enzymes were examined in nicotine-treated human periodontal ligament (PDL) cells by reverse transcription-polymerase chain reaction and Western blotting. RESULTS: Nicotine treatment concomitantly downregulated the expression of OPG and osteoblastic differentiation markers, such as alkaline phosphatase, osteocalcin, and osteopontin, and upregulated the expression of RANKL. Nicotine induced the synthesis of the transcription factor NF-E2-related factor-2 (Nrf2) as well as a number of cellular antioxidants and phase II enzymes, such as heme oxygenase-1. Pretreatment with antioxidants inhibited the upregulation of RANKL, the downregulation of OPG expression, and cytotoxicity by nicotine in PDL cells. CONCLUSIONS: Nicotine upregulated RANKL and antioxidant defense enzymes. These data suggest that Nrf2-mediated induction of cellular antioxidants and phase II enzymes could contribute to the cellular defense against nicotine-induced cytotoxicity and osteoclastic differentiation in PDL cells.


Subject(s)
Antioxidants/metabolism , Nicotine/pharmacology , Nicotinic Agonists/pharmacology , Periodontal Ligament/drug effects , RANK Ligand/drug effects , Alkaline Phosphatase/drug effects , Cell Differentiation/drug effects , Cell Line , Cell Survival/drug effects , Down-Regulation , Ferritins/drug effects , Glutamate-Cysteine Ligase/drug effects , Glutathione Peroxidase/drug effects , Glutathione Transferase/drug effects , Heme Oxygenase-1/drug effects , Humans , Mitogen-Activated Protein Kinases , NAD(P)H Dehydrogenase (Quinone)/drug effects , NF-E2-Related Factor 2/drug effects , NF-kappa B/drug effects , Nicotine/toxicity , Nicotinic Agonists/toxicity , Osteoblasts/drug effects , Osteocalcin/drug effects , Osteoclasts/drug effects , Osteopontin/drug effects , Osteoprotegerin/drug effects , Periodontal Ligament/cytology , Phosphatidylinositol 3-Kinases/drug effects , Up-Regulation
11.
J Periodontal Res ; 44(5): 673-82, 2009 Oct.
Article in English | MEDLINE | ID: mdl-19364390

ABSTRACT

BACKGROUND AND OBJECTIVE: Arecoline, an arecanut alkaloid present in the saliva of betel quid chewers, has been implicated in the pathogenesis of a variety of inflammatory oral diseases, including oral submucous fibrosis and periodontitis. To understand the molecular basis of arecoline action in epithelial changes associated with these diseases, we investigated the effects of arecoline on human keratinocytes with respect to cell growth regulation and the expression of stress-responsive genes. MATERIAL AND METHODS: Human keratinocyte cells (of the HaCaT cell line) were treated with arecoline, following which cell viability was assessed using the Trypan Blue dye-exclusion assay, cell growth and proliferation were analyzed using the MTT (3-[4,5-dimethylthiazol-2-yl]-2,5-diphenyltetrazolium bromide) and 5-bromo-2-deoxyuridine incorporation assays, cell cycle arrest and generation of reactive oxygen species were examined using flow cytometry, and gene expression changes were investigated using the reverse transcription-polymerase chain reaction technique. The role of oxidative stress, muscarinic acetylcholine receptor and mitogen-activated protein kinase (MAPK) pathways were studied using specific inhibitors. Western blot analysis was performed to study p38 MAPK activation. RESULTS: Arecoline induced the generation of reactive oxygen species and cell cycle arrest at the G1/G0 phase in HaCaT cells without affecting the expression of p21/Cip1. Arecoline-induced epithelial cell death at higher concentrations was caused by oxidative trauma without eliciting apoptosis. Sublethal concentrations of arecoline upregulated the expression of the following stress-responsive genes: heme oxygenase-1; ferritin light chain; glucose-6-phosphate dehydrogenase; glutamate-cysteine ligase catalytic subunit; and glutathione reductase. Additionally, there was a dose-dependent induction of interleukin-1alfa mRNA by arecoline via oxidative stress and p38 MAPK activation. CONCLUSION: Our data highlight the role of oxidative stress in arecoline-mediated cell death, gene regulation and inflammatory processes in human keratinocytes.


Subject(s)
Arecoline/pharmacology , Cholinergic Agonists/pharmacology , Keratinocytes/drug effects , Oxidative Stress/drug effects , Apoferritins , Cell Cycle/drug effects , Cell Death/drug effects , Cell Line , Cell Proliferation/drug effects , Cell Survival/drug effects , Coloring Agents , Cyclin-Dependent Kinase Inhibitor p21/drug effects , Dose-Response Relationship, Drug , Enzyme Activation/drug effects , Gene Expression Regulation/drug effects , Glucosephosphate Dehydrogenase/drug effects , Glutamate-Cysteine Ligase/drug effects , Glutathione Reductase/drug effects , Heme Oxygenase-1/drug effects , Humans , Interleukin-1alpha/biosynthesis , Mitogen-Activated Protein Kinases/drug effects , Oxidative Stress/genetics , Protein Kinase Inhibitors/metabolism , Reactive Oxygen Species/analysis , Receptors, Muscarinic/drug effects , p38 Mitogen-Activated Protein Kinases/drug effects
12.
Toxicol In Vitro ; 22(8): 1832-9, 2008 Dec.
Article in English | MEDLINE | ID: mdl-18801422

ABSTRACT

The objective was to investigate the regulation of glutathione (GSH) turnover in porcine aortic endothelial cells (PAECs) treated with sodium arsenite (NaAsO(2)), arsenic trioxide (As(2)O(3)) or sodium arsenate (Na(2)HAsO(4)) up to 72 hr at 0, 1, 5, and 10 microM, respectively. Intracellular GSH and glutathione disulfide (GSSG) contents, as well as the activities and mRNA levels of glutamate-cysteine lyase (GCL; gamma-glutamylcysteine synthetase) and gamma-glutamyl transpeptidase (GGT), were examined. The trivalent arsenic compounds increased GSH and GSSG contents in PAECs. An increase in GCL activity was observed at 24hr whereas an increase in GCL mRNA level was observed at 72 hr. The increase in GGT activity was only observed at 72 hr. In addition, a tendency of increase in GGT mRNA level was observed. Na(2)HAsO(4) treatment did not affect GSH content and the turnover-related enzymes. A differential GSH modulation in PAECs by trivalent arsenic compounds was found. The regulatory mechanism responsible for the As(2)O(3)-induced GSH increase is related to the GSH-turnover enzymes, GCL and GGT, while that for the NaAsO(2)-induced GSH increase may not be related to expression of GSH-turnover enzymes.


Subject(s)
Arsenates/toxicity , Arsenites/toxicity , Glutathione/drug effects , Oxides/toxicity , Sodium Compounds/toxicity , Animals , Aorta, Thoracic/cytology , Aorta, Thoracic/drug effects , Arsenic Trioxide , Arsenicals , Cells, Cultured , Endothelium, Vascular/cytology , Endothelium, Vascular/drug effects , Glutamate-Cysteine Ligase/drug effects , Glutamate-Cysteine Ligase/metabolism , Glutathione/metabolism , Glutathione Disulfide/drug effects , Glutathione Disulfide/metabolism , RNA, Messenger/drug effects , RNA, Messenger/metabolism , Swine , Time Factors , gamma-Glutamyltransferase/drug effects , gamma-Glutamyltransferase/metabolism
13.
Chem Biol Interact ; 176(2-3): 173-8, 2008 Nov 25.
Article in English | MEDLINE | ID: mdl-18765236

ABSTRACT

The chemoprotective effect of hydroxytyrosol (HT), a strong antioxidant compound from extra virgin olive oil, against acrylamide (AA)-induced genotoxicity was investigated in a human hepatoma cell line, HepG2. The micronucleus test (MNT) assay was used to monitor genotoxicity. In MNT, we found that HT at all tested concentrations (12.5-50 microM) significantly reduced the micronuclei frequencies in a concentration-dependent manner caused by AA. In order to clarify the underlying mechanisms we measured the intracellular reactive oxygen species (ROS) formation using 2,7-dichlorofluorescein diacetate (DCFH-DA) as a fluorescent probe. Intracellular glutathione (GSH) level was estimated by fluorometric methods. The rate-limiting enzyme in GSH synthesis is gamma-glutamylcysteine synthetase (gamma-GCS) and gamma-GCS was measured using Western blotting. The results showed that HT significantly concentration-dependent reduced the genotoxicity caused by AA. Furthermore, HT was able to reduce intracellular ROS formation and attenuate GSH depletion caused by AA in a concentration-dependent manner. It was also found that HT enhanced the expression of gamma-GCS in HepG2 cells treated with 10 mM AA using immunoblotting in a concentration-dependent manner. The results showed that HT reduced the AA-induced genotoxicity by decreasing the ROS level and increasing the GSH level. The data strongly suggest that HT have significant protective ability against AA-induced genotoxicity in vitro.


Subject(s)
Acrylamide/toxicity , Antioxidants/pharmacology , Liver/pathology , Phenylethyl Alcohol/analogs & derivatives , Blotting, Western , Cell Line, Tumor , Dose-Response Relationship, Drug , Fluoresceins , Fluorescent Dyes , Glutamate-Cysteine Ligase/biosynthesis , Glutamate-Cysteine Ligase/drug effects , Glutathione/analysis , Glutathione/drug effects , Glutathione/metabolism , Humans , Micronucleus Tests , Phenylethyl Alcohol/pharmacology , Reactive Oxygen Species/metabolism
14.
Int J Cancer ; 123(6): 1262-8, 2008 Sep 15.
Article in English | MEDLINE | ID: mdl-18566994

ABSTRACT

Lycopene can be cleaved by carotene 9',10'-oxygenase at its 9',10' double bond to form apo-10'-lycopenoids, including apo-10'-lycopenal, -lycopenol and -lycopenoic acid. The latter has been recently shown to inhibit lung carcinogenesis both in vivo and in vitro, however, the mechanism(s) underlying this protection is not well defined. In the present study, we report that treatment with apo-10'-lycopenoic acid, in a time- and dose-dependent manner, results in the nuclear accumulation of transcription factor Nrf2 (nuclear factor E(2)-related factor 2) protein in BEAS-2B human bronchial epithelial cells. The activation of Nrf2 by apo-10'-lycopenoic acid is associated with the induction of phase II detoxifying/antioxidant enzymes including heme oxygenase-1, NAD(P)H:quinone oxidoreductase 1, glutathione S-transferases, and glutamate-cysteine ligases in BEAS-2B cells. Furthermore, apo-10'-lycopenoic acid treatment increased total intracellular glutathione levels and suppressed both endogenous reactive oxygen species generation and H(2)O(2)-induced oxidative damage in BEAS-2B cells. In addition, both apo-10'-lycopenol and apo-10'-lycopenal induced heme oxygenase-1 gene expression in BEAS-2B cells. These data strongly suggest that the anti-carcinogenic and antioxidant functions of lycopene may be mediated by apo-10'-lycopenoids via activating Nrf2 and inducing phase II detoxifying/antioxidant enzymes.


Subject(s)
Anticarcinogenic Agents/pharmacology , Bronchi/drug effects , Carotenoids/pharmacology , NF-E2-Related Factor 2/drug effects , Respiratory Mucosa/drug effects , Anticarcinogenic Agents/metabolism , Blotting, Western , Bronchi/enzymology , Carotenoids/metabolism , Cells, Cultured , Gene Expression/drug effects , Glutamate-Cysteine Ligase/drug effects , Glutamate-Cysteine Ligase/metabolism , Glutathione Transferase/drug effects , Glutathione Transferase/metabolism , Heme Oxygenase-1/drug effects , Heme Oxygenase-1/metabolism , Humans , Lycopene , NAD(P)H Dehydrogenase (Quinone)/drug effects , NAD(P)H Dehydrogenase (Quinone)/metabolism , Oxidative Stress/drug effects , Reactive Oxygen Species/metabolism , Respiratory Mucosa/enzymology , Reverse Transcriptase Polymerase Chain Reaction
15.
Toxicol Appl Pharmacol ; 229(1): 44-55, 2008 May 15.
Article in English | MEDLINE | ID: mdl-18299140

ABSTRACT

Due to its unique physicochemical and optical properties, C60 has raised interest in commercialization for a variety of products. While several reports have determined this nanomaterial to act as a powerful antioxidant, many other studies have demonstrated a strong oxidative potential through photoactivation. To directly address the oxidative potential of C60, the effects of light and chemical supplementation and depletion of glutathione (GSH) on C60-induced toxicity were evaluated. Embryonic zebrafish were used as a model organism to examine the potential of C60 to elicit oxidative stress responses. Reduced light during C60 exposure significantly decreased mortality and the incidence of fin malformations and pericardial edema at 200 and 300 ppb C60. Embryos co-exposed to the glutathione precursor, N-acetylcysteine (NAC), also showed reduced mortality and pericardial edema; however, fin malformations were not reduced. Conversely, co-exposure to the GSH synthesis inhibitors, buthionine sulfoximine (BSO) and diethyl maleate (DEM), increased the sensitivity of zebrafish to C60 exposure. Co-exposure of C60 or its hydroxylated derivative, C60(OH)(24), with H2O2 resulted in increased mortality along the concentration gradient of H2O2 for both materials. Microarrays were used to examine the effects of C60 on the global gene expression at two time points, 36 and 48 h post fertilization (hpf). At both life stages there were alterations in the expression of several key stress response genes including glutathione-S-transferase, glutamate cysteine ligase, ferritin, alpha-tocopherol transport protein and heat shock protein 70. These results support the hypothesis that C60 induces oxidative stress in this model system.


Subject(s)
Fullerenes/toxicity , Gene Expression Regulation/drug effects , Oxidative Stress/drug effects , Acetylcysteine/pharmacology , Animals , Buthionine Sulfoximine/pharmacology , Carrier Proteins/drug effects , Carrier Proteins/metabolism , Embryo, Nonmammalian/drug effects , Embryo, Nonmammalian/metabolism , Ferritins/drug effects , Ferritins/metabolism , Glutamate-Cysteine Ligase/drug effects , Glutamate-Cysteine Ligase/metabolism , Glutathione Transferase/drug effects , Glutathione Transferase/metabolism , HSP70 Heat-Shock Proteins/drug effects , HSP70 Heat-Shock Proteins/metabolism , Hydrogen Peroxide/metabolism , Light , Maleates/pharmacology , Microarray Analysis , Models, Animal , Pericardial Effusion/chemically induced , Time Factors , Zebrafish
16.
Cancer Immunol Immunother ; 57(3): 347-58, 2008 Mar.
Article in English | MEDLINE | ID: mdl-17668203

ABSTRACT

We recently reported that inhibition of Cyclooxygenase-2 (Cox-2) reduced human B-CLL proliferation and survival. Herein, we investigated the mechanisms whereby small molecule Cox-2 selective inhibitors, SC-58125 (a Celebrex analog) and CAY10404 blunt survival of human B-cell lymphomas and chronic lymphocytic leukemia B-cells. SC-58125 and OSU03012 (a Celebrex analog that lacks Cox-2 inhibitory activity) both decreased intracellular glutathione (GSH) content in malignant human B-cells, as well as in Cox-2 deficient mouse B-cells. This new finding supports Cox-2 independent effects of SC-58125. Interestingly, SC-58125 also significantly increased B-cell reactive oxygen species (ROS) production, suggesting that ROS are a pathway that reduces malignant cell survival. Addition of GSH ethyl ester protected B lymphomas from the increased mitochondrial membrane permeability and reduced survival induced by SC-58125. Moreover, the SC-58125-mediated GSH depletion resulted in elevated steady-state levels of the glutamate cysteine ligase catalytic subunit mRNA and protein. These new findings of increased ROS and diminished GSH levels following SC-58125 exposure support novel mechanisms whereby a Cox-2 selective inhibitor reduces malignant B-cell survival. These observations also support the concept that certain Cox-2 selective inhibitors may have therapeutic value in combination with other drugs to kill malignant B lineage cells.


Subject(s)
B-Lymphocytes/drug effects , Cyclooxygenase 2 Inhibitors/pharmacology , Cyclooxygenase 2/drug effects , Glutathione/metabolism , Leukemia, Lymphocytic, Chronic, B-Cell/drug therapy , Lymphoma, B-Cell/drug therapy , Oxidative Stress/drug effects , Animals , B-Lymphocytes/metabolism , Catalytic Domain/drug effects , Catalytic Domain/genetics , Cell Proliferation/drug effects , Cell Survival/drug effects , Cells, Cultured , Cyclooxygenase 2/deficiency , Cyclooxygenase 2/metabolism , Dose-Response Relationship, Drug , Glutamate-Cysteine Ligase/drug effects , Glutamate-Cysteine Ligase/genetics , Glutathione/analogs & derivatives , Glutathione/antagonists & inhibitors , Glutathione/pharmacology , Humans , Isoxazoles/pharmacology , Leukemia, Lymphocytic, Chronic, B-Cell/metabolism , Lymphoma, B-Cell/metabolism , Mice , Mice, Knockout , Pyrazoles/pharmacology , RNA, Messenger/drug effects , RNA, Messenger/genetics , Reactive Oxygen Species/metabolism , Reverse Transcriptase Polymerase Chain Reaction/methods , Sulfonamides/pharmacology , Sulfones/pharmacology
17.
Free Radic Biol Med ; 43(3): 444-53, 2007 Aug 01.
Article in English | MEDLINE | ID: mdl-17602960

ABSTRACT

On liver injury, quiescent hepatic stellate cells (HSC), the most relevant cell type for hepatic fibrogenesis, become active, characterized by enhanced cell growth and overproduction of extracellular matrix (ECM). Oxidative stress facilitates HSC activation and the pathogenesis of hepatic fibrosis. Glutathione (GSH) is the most important intracellular antioxidant. We previously showed that curcumin, the yellow pigment in curry from turmeric, significantly inhibited HSC activation. The aim of this study is to elucidate the underlying mechanisms. It is hypothesized that curcumin might inhibit HSC activation mainly by its antioxidant capacity. Results from this study demonstrate that curcumin dose and time dependently attenuates oxidative stress in passaged HSC demonstrated by scavenging reactive oxygen species and reducing lipid peroxidation. Curcumin elevates the level of cellular GSH and induces de novo synthesis of GSH in HSC by stimulating the activity and gene expression of glutamate-cysteine ligase (GCL), a key rate-limiting enzyme in GSH synthesis. Depletion of cellular GSH by the inhibition of GCL activity using L-buthionine sulfoximine evidently eliminates the inhibitory effects of curcumin on HSC activation. Taken together, our results demonstrate, for the first time, that the antioxidant property of curcumin mainly results from increasing the level of cellular GSH by inducing the activity and gene expression of GCL in activated HSC in vitro. De novo synthesis of GSH is a prerequisite for curcumin to inhibit HSC activation. These results provide novel insights into the mechanisms of curcumin as an antifibrogenic candidate in the prevention and treatment of hepatic fibrosis.


Subject(s)
Antioxidants/pharmacology , Curcumin/pharmacology , Glutathione/biosynthesis , Hepatocytes/metabolism , Hepatocytes/physiology , Animals , Cells, Cultured , Gene Expression/drug effects , Glutamate-Cysteine Ligase/drug effects , Glutamate-Cysteine Ligase/metabolism , Lipid Peroxides/metabolism , Male , Rats , Rats, Sprague-Dawley , Reactive Oxygen Species/metabolism
18.
Redox Rep ; 12(1): 101-6, 2007.
Article in English | MEDLINE | ID: mdl-17263920

ABSTRACT

4-Hydroxynonenal (HNE), a major electrophilic product of lipid peroxidation, is regarded as both a marker of oxidative stress and a mediator of oxidative damage. At subtoxic concentrations, however, this compound has been shown to be a signalling molecule that can induce the expression of various antioxidant/detoxification enzymes, including glutamate-cysteine ligase (GCL), the rate-limiting enzyme in the de novo synthesis of glutathione. GCL consists of a catalytic (GCLC) and modulatory (GCLM) subunit, which are encoded by separate genes. Here, we investigated the effect of submicromolar concentrations of HNE on the expression of the GCL genes and the transcription factors involved. We demonstrated that submicromolar concentrations of HNE (as little as 0.3 muM) could increase the expression of both GCLC and GCLM. We also found that the induction of GCL expression was abrogated by siRNA for Nrf2. Our data suggest that a submicromolar concentration of HNE, as found in human plasma under physiological conditions, can induce GCL transcription in cultured cells implying that 'basal' expression of GCL is under regulation by lipid peroxidation that occurs under physiological conditions. Moreover, this induction is mediated through the EpRE-Nrf2 signalling pathway thought to be predominantly active only during stress.


Subject(s)
Aldehydes/toxicity , Lipid Peroxidation/physiology , Animals , Glutamate-Cysteine Ligase/drug effects , Glutamate-Cysteine Ligase/metabolism , Glutathione/metabolism , Kinetics , Signal Transduction/drug effects , Signal Transduction/physiology
19.
Brain Res ; 1117(1): 101-8, 2006 Oct 30.
Article in English | MEDLINE | ID: mdl-16949561

ABSTRACT

Glucocorticoids have been widely used as a therapeutic drug for sudden sensorineural hearing loss. However, very little is known about the mechanism(s) underlying the protective effect of glucocorticoids against hearing loss. As an approach toward elucidating the mechanism(s), we evaluated the effects of dexamethasone (DEX) treatment on the biosynthesis of GSH in the mouse cochlea in vivo. The systemic administration of DEX led to a significant increase in the total GSH level in the cochlea 2 to 24 h later. This DEX-induced increase in GSH occurred selectively in the spiral ganglion, but not significantly in the lateral wall tissues or in the organ of Corti. Furthermore, RT-PCR analysis revealed that DEX treatment resulted in enhanced expression of gamma-glutamylcysteine synthetase (gamma-GCS), which is the rate-limiting enzyme for de novo GSH synthesis, 1 to 24 h after the treatment. In addition to enhancing GSH biosynthesis, DEX treatment was effective in reducing lipid peroxidation in the cochlea. Taken together, DEX has the ability to facilitate GSH biosynthesis through enhanced expression of gamma-GCS in the cochlear spiral ganglion.


Subject(s)
Dexamethasone/pharmacology , Glutathione/biosynthesis , Glutathione/drug effects , Neuroprotective Agents/pharmacology , Spiral Ganglion/drug effects , Spiral Ganglion/metabolism , Animals , Disease Models, Animal , Glucocorticoids/pharmacology , Glutamate-Cysteine Ligase/drug effects , Glutamate-Cysteine Ligase/metabolism , Hearing Loss, Sensorineural/drug therapy , Hearing Loss, Sensorineural/physiopathology , Hearing Loss, Sensorineural/prevention & control , Lipid Peroxidation/drug effects , Lipid Peroxidation/physiology , Male , Mice , Neurons, Afferent/drug effects , Neurons, Afferent/metabolism , Oxidative Stress/drug effects , Oxidative Stress/physiology , Reactive Nitrogen Species/metabolism , Up-Regulation/drug effects , Up-Regulation/physiology
20.
Biometals ; 18(5): 519-28, 2005 Oct.
Article in English | MEDLINE | ID: mdl-16333752

ABSTRACT

Gamma-glutamylcysteine synthetase (gamma-GCS) catalyzes the first, rate-limiting step in the biosynthesis of glutathione (GSH). To evaluate the protective role of cellular GSH against arsenic-induced oxidative stress in Caenorhabditis elegans (C. elegans), we examined the effect of the C. elegans ortholog of GCS(h), gcs-1, in response to inorganic arsenic exposure. We have evaluated the responses of wild-type and gcs-1 mutant nematodes to both inorganic arsenite (As(III)) and arsenate (As(V)) ions and found that gcs-1 mutant nematodes are more sensitive to arsenic toxicity than that of wild-type animals. The amount of metal ion required to kill half of the population of worms falls in the order of wild-type/As(V)>gcs-1/As(V)> wild-type/As(III)>gcs-1/As(III). gcs-1 mutant nematodes also showed an earlier response to the exposure of As(III) and As(V) than that of wild-type animals. Pretreatment with GSH significantly raised the survival rate of gcs-1 mutant worms compared to As(III)- or As(V)-treated worms alone. These results indicate that GCS-1 is essential for the synthesis of intracellular GSH in C. elegans and consequently that the intracellular GSH status plays a critical role in protection of C. elegans from arsenic-induced oxidative stress.


Subject(s)
Arsenates/antagonists & inhibitors , Arsenites/antagonists & inhibitors , Caenorhabditis elegans Proteins/metabolism , Caenorhabditis elegans/metabolism , Glutamate-Cysteine Ligase/metabolism , Oxidative Stress , Animals , Arsenates/toxicity , Arsenites/toxicity , Caenorhabditis elegans/drug effects , Caenorhabditis elegans Proteins/drug effects , Caenorhabditis elegans Proteins/genetics , Dose-Response Relationship, Drug , Drug Resistance, Bacterial , Glutamate-Cysteine Ligase/drug effects , Glutamate-Cysteine Ligase/genetics , Glutathione/biosynthesis , Glutathione/pharmacology , Mutation , Time Factors
SELECTION OF CITATIONS
SEARCH DETAIL
...