Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 24
Filter
1.
Molecules ; 24(23)2019 Nov 22.
Article in English | MEDLINE | ID: mdl-31766654

ABSTRACT

Impaired glutathione (GSH) synthesis and dopaminergic transmission are important factors in the pathophysiology of schizophrenia. Our research aimed to assess the effects of l-buthionine-(S,R)-sulfoximine (BSO), a GSH synthesis inhibitor, and GBR 12909, a dopamine reuptake inhibitor, administered alone or in combination, to Sprague-Dawley rats during early postnatal development (p5-p16), on the levels of GSH, sulfur amino acids, global DNA methylation, and schizophrenia-like behavior. GSH, methionine (Met), homocysteine (Hcy), and cysteine (Cys) contents were determined in the liver, kidney, and in the prefrontal cortex (PFC) and hippocampus (HIP) of 16-day-old rats. DNA methylation in the PFC and HIP and schizophrenia-like behavior were assessed in adulthood (p90-p93). BSO caused the tissue-dependent decreases in GSH content and alterations in Met, Hcy, and Cys levels in the peripheral tissues and in the PFC and HIP. The changes in these parameters were accompanied by alterations in the global DNA methylation in the studied brain structures. Parallel to changes in the global DNA methylation, deficits in the social behaviors and cognitive functions were observed in adulthood. Only BSO + GBR 12909-treated rats exhibited behavioral alterations resembling positive symptoms in schizophrenia patients. Our results suggest the usefulness of this neurodevelopmental model for research on the pathomechanism of schizophrenia.


Subject(s)
Amino Acids, Sulfur/deficiency , Buthionine Sulfoximine/adverse effects , Glutathione/deficiency , Piperazines/adverse effects , Schizophrenia/chemically induced , Animals , DNA Methylation/drug effects , Disease Models, Animal , Homeostasis , Male , Rats , Rats, Sprague-Dawley , Schizophrenia/genetics , Schizophrenia/metabolism
2.
Antioxid Redox Signal ; 31(3): 211-226, 2019 07 20.
Article in English | MEDLINE | ID: mdl-30827134

ABSTRACT

Aims: Adaptation to low oxygen of hematopoietic stem cells (HSCs) in the bone marrow has been demonstrated to depend on the activation of hypoxia-inducible factor (HIF)-1α as well as the limited production of reactive oxygen species (ROS). In this study, we aimed at determining whether HIF-1α is involved in protecting HSCs from ROS. Results: Oxidative stress was induced by DL-buthionine-(S,R)-sulfoximine (BSO)-treatment, which increases the mitochondrial ROS level. Hypoxia rescued Lineage-Sca-1+c-kit+ (LSK) cells from BSO-induced apoptosis, whereas cells succumbed to apoptosis in normoxia. Apoptosis in normoxia was inhibited with the antioxidant N-acetyl-L-cysteine or by overexpression of anti-apoptotic BCL-2. Moreover, stabilized expression of oxygen-insensitive HIFs could not protect LSK cells from oxidative stress-induced apoptosis at normoxia, neither could short hairpin RNA to Hif-1α inhibit the protective effects by hypoxia in LSK cells. Likewise, BSO treatment of LSK cells from Hif-1α knockout mice did not suppress the effects seen in hypoxia. Microarray analysis identified the nuclear factor-kappa B (NF-κB) pathway as a pathway induced by hypoxia. By using NF-κB lentiviral construct and DNA-binding assay, we found increased NF-κB activity in cells cultured in hypoxia compared with normoxia. Using an inhibitor against NF-κB activation, we could confirm the involvement of NF-κB signaling as BSO-mediated cell death was significantly increased in hypoxia after adding the inhibitor. Innovation: HIF-1α is not involved in protecting HSCs and progenitors to elevated levels of ROS on glutathione depletion during hypoxic conditions. Conclusion: The study proposes a putative role of NF-κB signaling as a hypoxia-induced regulator in early hematopoietic cells.


Subject(s)
Buthionine Sulfoximine/adverse effects , Hematopoietic Stem Cells/cytology , Hypoxia-Inducible Factor 1, alpha Subunit/genetics , NF-kappa B/metabolism , Acetylcysteine/pharmacology , Animals , Apoptosis , Cell Hypoxia , Cells, Cultured , Gene Knockout Techniques , Hematopoietic Stem Cells/metabolism , Humans , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Mice , Oxidative Stress , Proto-Oncogene Proteins c-bcl-2/genetics , Reactive Oxygen Species/metabolism
3.
Mol Cell Biochem ; 428(1-2): 67-77, 2017 Apr.
Article in English | MEDLINE | ID: mdl-28070834

ABSTRACT

Resveratrol is a dietary polyphenol that displays neuroprotective properties in several in vivo and in vitro experimental models, by modulating oxidative and inflammatory responses. Glutathione (GSH) is a key antioxidant in the central nervous system (CNS) that modulates several cellular processes, and its depletion is associated with oxidative stress and inflammation. Therefore, this study sought to investigate the protective effects of resveratrol against GSH depletion pharmacologically induced by buthionine sulfoximine (BSO) in C6 astroglial cells, as well as its underlying cellular mechanisms. BSO exposure resulted in several detrimental effects, decreasing glutamate-cysteine ligase (GCL) activity, cystine uptake, GSH intracellular content and the activities of the antioxidant enzymes glutathione peroxidase (GPx) and glutathione reductase (GR). Moreover, BSO increased reactive oxygen/nitrogen species (ROS/RNS) levels and pro-inflammatory cytokine release. Resveratrol prevented these effects by protecting astroglial cells against BSO-induced cytotoxicity, by modulating oxidative and inflammatory responses. Additionally, we observed that pharmacological inhibition of heme oxygenase 1 (HO-1), an essential cellular defense against oxidative and inflammatory injuries, abolished all the protective effects of resveratrol. These observations suggest HO-1 pathway as a cellular effector in the mechanism by which resveratrol protects astroglial cells against GSH depletion, a condition that may be associated to neurodegenerative diseases.


Subject(s)
Astrocytes/enzymology , Buthionine Sulfoximine/adverse effects , Glutathione/metabolism , Heme Oxygenase-1/metabolism , Stilbenes/pharmacology , Animals , Astrocytes/drug effects , Astrocytes/immunology , Cell Line , Cell Survival/drug effects , Cytokines/metabolism , Male , Oxidative Stress/drug effects , Rats , Resveratrol , Signal Transduction/drug effects
4.
Pediatr Blood Cancer ; 63(8): 1349-56, 2016 08.
Article in English | MEDLINE | ID: mdl-27092812

ABSTRACT

BACKGROUND: Myeloablative therapy for high-risk neuroblastoma commonly includes melphalan. Increased cellular glutathione (GSH) can mediate melphalan resistance. Buthionine sulfoximine (BSO), a GSH synthesis inhibitor, enhances melphalan activity against neuroblastoma cell lines, providing the rationale for a Phase 1 trial of BSO-melphalan. PROCEDURES: Patients with recurrent/resistant high-risk neuroblastoma received BSO (3 gram/m(2) bolus, then 24 grams/m(2) /day infusion days -4 to -2), with escalating doses of intravenous melphalan (20-125 mg/m(2) ) days -3 and -2, and autologous stem cells day 0 using 3 + 3 dose escalation. RESULTS: Among 28 patients evaluable for dose escalation, one dose-limiting toxicity occurred at 20 mg/m(2) melphalan (grade 3 aspartate aminotransferase/alanine aminotransferase) and one at 80 mg/m(2) (streptococcal bacteremia, grade 4 hypotension/pulmonary/hypocalcemia) without sequelae. Among 25 patients evaluable for response, there was one partial response (PR) and two mixed responses (MRs) among eight patients with prior melphalan exposure; one PR and three MRs among 16 patients without prior melphalan; one stable disease with unknown melphalan history. Melphalan pharmacokinetics with BSO were similar to reports for melphalan alone. Melphalan Cmax for most patients was below the 10 µM concentration that showed neuroblastoma preclinical activity with BSO. CONCLUSIONS: BSO (75 gram/m(2) ) with melphalan (125 mg/m(2) ) is tolerable with stem cell support and active in recurrent/refractory neuroblastoma. Further dose escalation is feasible and may increase responses.


Subject(s)
Antimetabolites, Antineoplastic/therapeutic use , Antineoplastic Agents, Alkylating/therapeutic use , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Buthionine Sulfoximine/therapeutic use , Hematopoietic Stem Cell Transplantation , Melphalan/therapeutic use , Myeloablative Agonists/therapeutic use , Neuroblastoma/drug therapy , Adolescent , Buthionine Sulfoximine/adverse effects , Child , Child, Preschool , Drug Synergism , Female , Glutamate-Cysteine Ligase/antagonists & inhibitors , Glutathione/therapeutic use , Hematopoietic Stem Cells/metabolism , Humans , Male , Melphalan/adverse effects , Melphalan/pharmacokinetics , Neoplasm Recurrence, Local/drug therapy
5.
Nutr Cancer ; 68(3): 507-17, 2016.
Article in English | MEDLINE | ID: mdl-27008382

ABSTRACT

The induction of reactive oxygen species (ROS) to selectively kill cancer cells is an important feature of radiotherapy and various chemotherapies. Depletion of glutathione can induce apoptosis in cancer cells or sensitize them to anticancer treatments intended to modulate ROS levels. In contrast, antioxidants protect cancer cells from oxidative stress-induced cell death by scavenging ROS. The role of exogenous antioxidants in cancer cells under oxidative insults remains controversial and unclear. This study aimed to identify protective pathways modulated by γ-tocotrienol (γT3), an isomer of vitamin E, in human neuroblastoma SH-SY5Y cells under oxidative stress. Using buthionine sulfoximine (BSO) as an inhibitor of glutathione synthesis, we found that BSO treatment reduced the viability of SH-SY5Y cells. BSO induced cell death by increasing apoptosis, decreased the level of reduced glutathione (GSH), and increased ROS levels in SH-SY5Y cells. Addition of γT3 increased the viability of BSO-treated cells, suppressed apoptosis, and decreased the ROS level induced by BSO, while the GSH level was unaffected. These results suggest that decreasing GSH levels by BSO increased ROS levels, leading to apoptosis in SH-SY5Y cells. γT3 attenuated the BSO-induced cell death by scavenging free radicals.


Subject(s)
Buthionine Sulfoximine/adverse effects , Chromans/pharmacology , Free Radical Scavengers/pharmacology , Neuroblastoma/drug therapy , Vitamin E/analogs & derivatives , Vitamin E/pharmacology , Antioxidants/pharmacology , Cell Death/drug effects , Cell Line, Tumor/drug effects , Enzyme Inhibitors/pharmacology , Glutathione/metabolism , Humans , Imidazoles/pharmacology , MAP Kinase Signaling System/drug effects , Neuroblastoma/metabolism , Neuroblastoma/pathology , Protective Agents/pharmacology , Protein Kinase C-delta/metabolism , Pyridines/pharmacology , Reactive Oxygen Species/metabolism
6.
Pediatr Blood Cancer ; 62(10): 1739-46, 2015 Oct.
Article in English | MEDLINE | ID: mdl-26153194

ABSTRACT

PURPOSE: To evaluate BSO-mediated glutathione (GSH) depletion in combination with L-PAM for children with recurrent or refractory high-risk neuroblastoma (NB) as a means to enhance alkylator sensitivity. PROCEDURE: This pilot study (NCI #T95-0092) administered L-S,R-buthionine sulfoximine (BSO) as a bolus followed by 72 hr continuous infusion of either 0.75 g/m(2)/hr (level 1) or 1.0 g/m(2)/hr (level 2) and melphalan (L-PAM) (15 mg/m(2) bolus at hour 48 of BSO infusion). GSH in blood mononuclear cells and bone marrow was measured by enzymatic assay, BSO in plasma by HPLC. RESULTS: Thirty two patients received 58 courses of therapy (median 1, range 1-4 courses). Blood mononuclear cell GSH decreased (48 hr) to 47% ± 15.7%. Level 2 mean steady-state concentration (Css) for BSO = 524 ± 207 µM and peak L-PAM concentration = 3.32 ± 1.2 µM. Grade 3-4 leukopenia and thrombocytopenia were common. There were two deaths from CNS toxicity and acute tubular necrosis; one had a large, intracranial mass, both were receiving cephalosporin antibiotics. No other significant toxicities were seen. There were six responses (five partial and, one mixed) representing an 18% response rate; four/six responses occurred in patients that relapsed following myeloablative therapy and included a 98% reduction in volume (cm(3)) of a pelvic mass, and three/five patients with >3 log reduction of tumor in marrow as measured by immunocytology (sensitivity 1/10(5)). CONCLUSIONS: BSO/L-PAM has activity against recurrent high-risk NB. Exclusion of cephalosporin antibiotics in future clinical trials of BSO may diminish the potential for serious renal and CNS toxicity.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/administration & dosage , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Neoplasm Recurrence, Local/drug therapy , Neuroblastoma/drug therapy , Antineoplastic Combined Chemotherapy Protocols/adverse effects , Buthionine Sulfoximine/administration & dosage , Buthionine Sulfoximine/adverse effects , Child , Chromatography, High Pressure Liquid , Female , Glutathione/analysis , Glutathione/drug effects , Humans , Infusions, Intravenous , Injections, Intravenous , Male , Melphalan/administration & dosage , Melphalan/adverse effects , Pilot Projects
7.
J Toxicol Sci ; 39(4): 537-44, 2014 Aug.
Article in English | MEDLINE | ID: mdl-25056778

ABSTRACT

Generally, reactive metabolites are detoxified by conjugation with glutathione (GSH). A GSH-depleted model was prepared by administering L-buthionine-(S,R)-sulfoximine (BSO), which can be used to detect hepatic damage by reactive metabolites. However, BSO may cause adverse effects on other organs, such as renal damage by reactive metabolites because it depletes GSH in the whole body. The present study was designed to examine whether it was possible to specifically detect hepatic damage by reactive metabolites without reducing renal GSH levels by administering BSO in a time course when hepatic GSH levels are naturally reduced. Male BALB/c mice were administered reverse osmosis (RO) water or 20 mmol/l BSO in drinking water for 4 days. Subsequently, animals in the RO water group were orally administered 500 mg/kg acetaminophen (APAP) at 9:00 or 15:00 and in the BSO group at 9:00 for 4 days. As a result, severe hepatic damage and necrosis of the renal proximal tubules were observed in the BSO/APAP administration at 9:00 group, and all animals died on 1 or 2 days after APAP administration. Hepatic damage was clearly increased in the RO water/APAP administration at 15:00 group compared with the RO water/APAP administration at 9:00 group. However, renal damage and deaths were not observed. This BSO administration model may detect renal damage induced by reactive metabolites. Using an administration time course, whereby hepatic GSH levels were naturally reduced, hepatic damage by reactive metabolites can be detected without secondary renal effects.


Subject(s)
Acetaminophen/adverse effects , Chemical and Drug Induced Liver Injury/etiology , Circadian Rhythm/physiology , Glutathione/deficiency , Glutathione/metabolism , Liver/drug effects , Liver/metabolism , Acetaminophen/administration & dosage , Acetaminophen/metabolism , Administration, Oral , Animals , Buthionine Sulfoximine/administration & dosage , Buthionine Sulfoximine/adverse effects , Disease Models, Animal , Male , Mice, Inbred BALB C , Osmosis/physiology , Time Factors , Water/administration & dosage
8.
J Agric Food Chem ; 61(17): 4072-8, 2013 May 01.
Article in English | MEDLINE | ID: mdl-23573869

ABSTRACT

Oxidative stress state such as depletion of the intracellular glutathione (GSH) is associated with the development of cancer. Some dietary phytochemicals have been shown to possess a cancer preventive effect, although the understanding of the involved mechanisms is still limited. Recent study has shown that glyceollins, phytoalexins derived from soybean by biotic elicitor, might have a cancer preventive effect through induction of detoxifying/antioxidant enzymes. The objective of this study was to investigate the effects of glyceollins on the Nrf2 signaling pathway under excessive oxidative stress induced by GSH depletion. In mouse hepatoma cells (Hepa1c1c7) subjected to the buthionine sulfoximine (BSO), an inhibitor of γ-glutamylcysteine synthetase (γGCS), the intracellular GSH content was significantly lowered. On the other hand, incubation with glyceollins in the presence of BSO increased the level of GSH, expression of γGCS, and nuclear translocation of NF-E2-related factor-2 (Nrf2), compared to the cells treated with BSO only. Nrf2-antioxidant responsive element (ARE)-reporter activity assay in HepG2-C8 showed that BSO increased the ARE-reporter activity in a dose-dependent manner, compared to vehicle-treated cells, whereas cotreatment with glyceollins caused further increase in reporter luciferase activity relative to BSO alone. Taken together, glyceollins synergistically activated the Nrf2 signaling pathway and subsequently the expression of phase 2/antioxidant enzymes in the presence of BSO, suggesting that BSO-induced oxidative stress and that glyceollins regulate the expression of phase 2/antioxidant enzymes through different mechanisms from each other.


Subject(s)
Anticarcinogenic Agents/pharmacology , NF-E2-Related Factor 2/metabolism , Oxidative Stress/drug effects , Pterocarpans/pharmacology , Signal Transduction , Animals , Antioxidants/pharmacology , Buthionine Sulfoximine/adverse effects , Cell Line , Genes, Reporter , Glutamate-Cysteine Ligase/antagonists & inhibitors , Glutamate-Cysteine Ligase/metabolism , Glutathione/metabolism , Hep G2 Cells , Humans , Hypoxia-Inducible Factor-Proline Dioxygenases/genetics , Hypoxia-Inducible Factor-Proline Dioxygenases/metabolism , Mice , NF-E2-Related Factor 2/genetics , Neoplasms/prevention & control , Phytochemicals/pharmacology , Glycine max/chemistry
9.
Food Chem Toxicol ; 56: 425-35, 2013 Jun.
Article in English | MEDLINE | ID: mdl-23500778

ABSTRACT

Here, we investigated whether hirsutenone, a compound isolated from Alnus japonica, was able to attenuate oxidative stress-induced death in transformed retinal ganglion (RGC-5) cells. Hirsutenone effectively protected RGC-5 cells from oxidative insult induced by, l-buthionine-(S,R)-sulfoximine (BSO) plus glutamate in a concentration-dependent manner, as demonstrated by propidium iodide (PI)/Hoechst 33342 double staining, flow cytometry, and MTT assays. Moreover, hirsutenone inhibited the increase in apoptotic protein expression resulting from BSO plus glutamate. Hirsutenone also effectively inhibited sodium nitroprusside (SNP)-induced lipid peroxidation in rat brain homogenates. To investigate the effects of hirsutenone in vivo, we used N-methyl-d-aspartate (NMDA) as a negative insult on the retinas of rats. NMDA affects the thinning of the inner plexiform layer (IPL) and causes an increase in the number of terminal deoxynucleotidyl transferase-mediated dUTP nick-end labeling (TUNEL)-positive ganglion cells in the ganglion cell layer (GCL). Hirsutenone treatment led to a reduction in NMDA-induced IPL and TUNEL staining of the GCL. In conclusion, hirsutenone isolated from A. japonica may act as neuroprotective agent for conditions such as glaucoma.


Subject(s)
Alnus/chemistry , Neuroprotective Agents/pharmacology , Oxidative Stress/drug effects , Plant Extracts/pharmacology , Retinal Ganglion Cells/drug effects , Animals , Apoptosis/drug effects , Buthionine Sulfoximine/adverse effects , Buthionine Sulfoximine/metabolism , Catechols/pharmacology , Cell Survival/drug effects , DNA Nucleotidylexotransferase/metabolism , Diarylheptanoids/pharmacology , Glaucoma/drug therapy , Glaucoma/prevention & control , Glutamic Acid/adverse effects , Glutamic Acid/metabolism , In Situ Nick-End Labeling , Lipid Peroxidation/drug effects , Male , N-Methylaspartate/adverse effects , N-Methylaspartate/antagonists & inhibitors , Nitroprusside/adverse effects , Nitroprusside/metabolism , Propidium/metabolism , Rats , Rats, Sprague-Dawley , Retina/drug effects , Retina/metabolism , Retinal Ganglion Cells/cytology , Retinal Ganglion Cells/metabolism , Thiobarbituric Acid Reactive Substances/chemistry
10.
Mol Nutr Food Res ; 57(2): 249-59, 2013 Feb.
Article in English | MEDLINE | ID: mdl-23255485

ABSTRACT

SCOPE: Flavonoids have well-known antioxidant, anti-inflammatory, and anti-cancer activities. Isoflavone genistein is considered a potent antioxidant agent against oxidative stress. Although several mechanisms have been proposed, a clear antioxidant mechanism of genistein is still remained to be answered. METHODS AND RESULTS: In this study, we focused on the concerted effects on expression of Nrf2 and phase II enzyme pathway components. Transient transfection assays, RT-PCR and immunoblot analysis were performed to study its molecular mechanisms of action. In Caco-2 cells, treatment with genistein markedly attenuated H(2)O(2) -induced peroxide formation; this amelioration was reversed by buthionine sulfoximine(GCLC inhibitor) and zinc protoporphyrin(HO-1 inhibitor). Genistein increased HO-1 and GCLC mRNA and protein expression. Genistein treatment activated the ERK1/2 and PKC signaling pathway; therefore increased Nrf2 mRNA and protein expression. The roles of the ERK1/2 and PKC signaling pathway were determined using PD98059 (ERK1/2 inhibitor) and GF109203X (PKC inhibitor) and RNA interference directed against Nrf2. Both inhibitors and siNrf2 abolished genistein-induced HO-1 and GCLC protein expression. These results suggest the involvement of ERK1/2, PKC, and Nrf2 in inducing HO-1 and GCLC by genistein. CONCLUSION: Our studies show that genistein up-regulated HO-1 and GCLC expression through the EKR1/2 and PKC /Nrf2 pathways during oxidative stress.


Subject(s)
Antioxidants/pharmacology , Genistein/pharmacology , MAP Kinase Signaling System/drug effects , Metabolic Detoxication, Phase II/physiology , NF-E2-Related Factor 2/metabolism , Oxidative Stress/drug effects , Buthionine Sulfoximine/adverse effects , Caco-2 Cells , Flavonoids/pharmacology , Gene Expression/drug effects , Glutamate-Cysteine Ligase/antagonists & inhibitors , Glutamate-Cysteine Ligase/metabolism , Heme Oxygenase-1/antagonists & inhibitors , Heme Oxygenase-1/metabolism , Humans , Hydrogen Peroxide/metabolism , NF-E2-Related Factor 2/genetics , Protein Kinase C/antagonists & inhibitors , Protein Kinase C/metabolism , Protoporphyrins/adverse effects , RNA, Messenger/genetics , RNA, Messenger/metabolism , Up-Regulation
11.
J Appl Toxicol ; 32(10): 823-33, 2012 Oct.
Article in English | MEDLINE | ID: mdl-22407903

ABSTRACT

Drug-induced liver injury (DILI) is a major safety concern in drug development and clinical practice. The pathogenesis of DILI usually involves the participation of the parent drug or metabolites that either affect cellular function or elicit an immune response. However, the mechanisms leading to DILI are unknown in most cases. Methimazole (MTZ) is used as an antithyroid drug and is well known to have induced liver injuries such as cholestatic hepatitis in a small number of human cases. Immune-mediated reactions were also suggested to play a role in MTZ-induced acute liver injury, but the mechanism underlying this process has not been elucidated. To address this issue, we measured plasma aspartate aminotransferase (AST) and alanine aminotransferase (ALT) levels, hepatic glutathione levels, hepatic expression of CD4⁺ Th cell-related transcriptional factors, cytokines and chemokines, plasma interleukin (IL)-4 levels and histopathological changes in the liver following MTZ (450 mg kg⁻¹ , p.o.) administration in mice. The hepatic expression of mRNA for Th2 cell-related factors, such as GATA-binding protein, macrophage inflammatory protein-2 (MIP-2) and plasma IL-4 levels, as well as plasma AST and ALT levels, was significantly increased in mice treated with MTZ. These changes were markedly enhanced by pre-treatment with L-buthionine sulfoximine (3 mmol kg⁻¹, i.p.) and MTZ (15 mg kg⁻¹, p.o.). Neutralization of IL-4 using a monoclonal anti-mouse IL-4 antibody (100 µg/mouse, single i.p.) suppressed the hepatotoxic effect of MTZ. In conclusion, this report is the first to demonstrate that Th2 cytokine-mediated immune responses are involved in MTZ-induced acute liver injury in mice.


Subject(s)
Antithyroid Agents/adverse effects , Chemical and Drug Induced Liver Injury/immunology , Cytokines/metabolism , Liver/drug effects , Methimazole/adverse effects , Th2 Cells/drug effects , Animals , Antibodies, Monoclonal/therapeutic use , Antibodies, Neutralizing/therapeutic use , Antimetabolites/adverse effects , Antithyroid Agents/administration & dosage , Buthionine Sulfoximine/adverse effects , Chemical and Drug Induced Liver Injury/metabolism , Chemical and Drug Induced Liver Injury/physiopathology , Chemical and Drug Induced Liver Injury/prevention & control , Cytokines/genetics , Dose-Response Relationship, Drug , Female , GATA3 Transcription Factor/genetics , GATA3 Transcription Factor/metabolism , Gene Expression Regulation/drug effects , Glutathione/metabolism , Interleukin-4/antagonists & inhibitors , Interleukin-4/blood , Interleukin-4/metabolism , Liver/immunology , Liver/pathology , Liver/physiopathology , Methimazole/administration & dosage , Mice , Mice, Inbred BALB C , RNA, Messenger/metabolism , Th2 Cells/immunology , Th2 Cells/metabolism
12.
Biochim Biophys Acta ; 1822(6): 980-7, 2012 Jun.
Article in English | MEDLINE | ID: mdl-22330094

ABSTRACT

Multidrug resistance-associated protein 2 (MRP2) is a member of a family of efflux transporters that are involved in biliary excretion of organic anions from hepatocytes. Disrupted canalicular localization and decreased protein expression of MRP2 have been observed in patients with chronic cholestatic disorder and hepatic failure without a change in its mRNA expression. We have previously demonstrated that post-transcriptional regulation of the rapid retrieval of rat MRP2 from the canalicular membrane to the intracelluar compartment occurs under conditions of acute (~30min) oxidative stress. However, it is unclear whether MRP2 expression is decreased during its sustained internalization during chronic oxidative stress. The present study employed buthionine sulfoximine (BSO) to induce chronic oxidative stress in the livers of Sprague-Dawley rats and then examined the protein expression and localization of MRP2. Canalicular MRP2 localization was altered by BSO treatment for 2h without changing the hepatic protein expression of MRP2. While the 8h after exposure to BSO, hepatic MRP2 protein expression was decreased, and the canalicular localization of MRP2 was disrupted without changing the mRNA expression of MRP2. The BSO-induced reduction in MRP2 protein expression was suppressed by pretreatment with N-benzyloxycarbonyl (Cbz)-Leu-Leu-leucinal ( MG-132), a proteasomal inhibitor. Furthermore, the modification of MRP2 by small ubiquitin-relatedmodifier 1 (SUMO-1) was impaired in BSO-treated rat liver,while that by ubiquitin (Ub) and MRP2 was enhanced. Taken together, the results of this study suggest the sustained periods of low GSH content coupled with altered modification of MRP2 by Ub/SUMO-1 were accompanied by proteasomal degradation of MRP2.


Subject(s)
Glutathione/metabolism , Hepatocytes/metabolism , Liver/metabolism , Multidrug Resistance-Associated Proteins/metabolism , Proteasome Endopeptidase Complex/metabolism , Animals , Buthionine Sulfoximine/adverse effects , Buthionine Sulfoximine/pharmacology , Cholestasis, Intrahepatic/genetics , Cholestasis, Intrahepatic/metabolism , Cholestasis, Intrahepatic/pathology , Leupeptins/pharmacology , Liver Failure/pathology , Male , Membrane Transport Proteins/genetics , Membrane Transport Proteins/metabolism , Multidrug Resistance-Associated Protein 2 , Multidrug Resistance-Associated Proteins/genetics , Oxidative Stress , Proteasome Inhibitors , RNA, Messenger/genetics , RNA, Messenger/metabolism , Rats , Rats, Sprague-Dawley , SUMO-1 Protein/metabolism , Ubiquitin/metabolism
13.
Am J Hypertens ; 25(6): 629-35, 2012 Jun.
Article in English | MEDLINE | ID: mdl-22223042

ABSTRACT

Oxidative stress contributes to the development of several cardiovascular diseases, including diabetes, renal insufficiency, and arterial hypertension. Animal studies have evidenced the association between higher blood pressure (BP) and increased oxidative stress, and treatment with antioxidants has been shown to reduce BP, while BP reduction due to antihypertensive drugs is associated with reduced oxidative stress. In 2000, it was first reported that oxidative stress and arterial hypertension were produced in normal Sprague-Dawley rats by oral administration of buthionine sulfoximine (BSO), which induces glutathione (GSH) depletion, indicating that oxidative stress may induce hypertension. The contribution of several potential pathogenic factors has been evaluated in the BSO rat model, the prototype of oxidative stress-induced hypertension, including vascular reactivity, endothelium-derived factors, renin-angiotensin system activity, TXA(2)-PGH(2) production, sodium sensitivity, renal dopamine-induced natriuresis, and sympathetic tone. This review summarizes the main factors implicated in the pathogenesis of BSO-induced hypertension and the alterations associated with GSH depletion that are related to renal function or BP control.


Subject(s)
Buthionine Sulfoximine/pharmacology , Cardiovascular System/physiopathology , Glutathione/deficiency , Kidney/physiopathology , Oxidative Stress/drug effects , Animals , Antioxidants/pharmacology , Blood Pressure/drug effects , Blood Pressure/physiology , Buthionine Sulfoximine/adverse effects , Disease Models, Animal , Glutathione/drug effects , Glutathione/metabolism , Hypertension/chemically induced , Hypertension/metabolism , Hypertension/physiopathology , Mice , Oxidative Stress/physiology , Rats , Rats, Sprague-Dawley
14.
Free Radic Biol Med ; 52(1): 160-6, 2012 Jan 01.
Article in English | MEDLINE | ID: mdl-22041456

ABSTRACT

SDHD mutations are associated with human cancers but the mechanisms that may contribute to transformation are unknown. The hypothesis that mutations in SDHD increase levels of superoxide leading to genomic instability was tested using site-directed mutagenesis to generate a truncated SDHD cDNA that was expressed in Chinese hamster fibroblasts. Stable expression of mutant SDHD resulted in 2-fold increases in steady-state levels of superoxide that were accompanied by a significantly increased mutation rate as well as a 70-fold increase in mutation frequency at the hprt locus. Overexpression of MnSOD or treatment with polyethylene glycol conjugated (PEG)-catalase suppressed mutation frequency in SDHD mutant cells by 50% (P<0.05). Simultaneous treatment with PEG-catalase and PEG-SOD suppressed mutation frequency in SDHD mutant cells by 90% (P<0.0005). Finally, 95% depletion of glutathione using l-buthionine-[S,R]-sulfoximine (BSO) in SDHD mutant cells caused a 4-fold increase in mutation frequency (P<0.05). These results demonstrate that mutations in SDHD cause increased steady-state levels of superoxide which significantly contributed to increases in mutation rates and frequency mediated by superoxide and hydrogen peroxide. These results support the hypothesis that mutations in SDHD may contribute to carcinogenesis by increasing genomic instability mediated by increased steady-state levels of reactive oxygen species.


Subject(s)
Cell Transformation, Neoplastic/metabolism , Fibroblasts/metabolism , Hydrogen Peroxide/adverse effects , Neoplasms/enzymology , Protein Subunits/metabolism , Succinate Dehydrogenase/metabolism , Superoxides/adverse effects , Animals , Buthionine Sulfoximine/adverse effects , Catalase/genetics , Catalase/metabolism , Cell Transformation, Neoplastic/genetics , Cricetinae , Fibroblasts/cytology , Gene Expression , Genomic Instability , Glutathione/deficiency , Humans , Mutagenesis, Site-Directed , Mutation Rate , Neoplasms/genetics , Neoplasms/pathology , Plasmids , Point Mutation , Polyethylene Glycols/metabolism , Protein Subunits/genetics , Succinate Dehydrogenase/genetics , Superoxide Dismutase/genetics , Superoxide Dismutase/metabolism , Superoxides/metabolism , Transfection
15.
Chem Res Toxicol ; 24(11): 1836-44, 2011 Nov 21.
Article in English | MEDLINE | ID: mdl-21827172

ABSTRACT

Many cellular proteins with reactive thiols form covalent bonds with electrophiles, thereby modifying their structures and activities. Here, we describe the recovery of a glycolytic protein, glyceraldehyde-3-phosphate dehydrogenase (GAPDH), from such an electrophilic attack by 1,2-napthoquinone (1,2-NQ). GAPDH readily formed a covalent bond with 1,2-NQ through Cys152 at a low concentration (0.2 µM) in a cell-free system, but when human epithelial A549 cells were exposed to this quinone at 20 µM, only minimal binding was observed although extensive binding to numerous other cellular proteins occurred. Depletion of cellular glutathione (GSH) with buthionine sulfoximine (BSO) resulted in some covalent modification of cellular GAPDH by 1,2-NQ and a significant reduction of GAPDH activity in the cells. Incubation of native, but not boiled, human GAPDH that had been modified by 1,2-NQ with GSH resulted in a concentration-dependent removal of 1,2-NQ from the GAPDH conjugate, accompanied by partial recovery of lost catalytic activity and formation of a 1,2-NQ-GSH adduct (1,2-NQ-SG). While GAPDH is recognized as a multifunctional protein, our results show that GAPDH also has a unique ability to recover from electrophilic modification by 1,2-NQ through a GSH-dependent S-transarylation reaction.


Subject(s)
Epithelial Cells/enzymology , Glutathione/metabolism , Glyceraldehyde-3-Phosphate Dehydrogenases/metabolism , Naphthoquinones/metabolism , Recombinant Proteins/metabolism , Sulfhydryl Compounds/metabolism , Buthionine Sulfoximine/adverse effects , Buthionine Sulfoximine/pharmacology , Cell Line , Cell-Free System , Cloning, Molecular , Epithelial Cells/drug effects , Escherichia coli , Glyceraldehyde-3-Phosphate Dehydrogenases/chemistry , Glyceraldehyde-3-Phosphate Dehydrogenases/genetics , Glycolysis/drug effects , Glycolysis/genetics , Humans , Mutation , Naphthoquinones/chemistry , Oxidation-Reduction/drug effects , Plasmids , Protein Denaturation , Protein Processing, Post-Translational , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Sulfhydryl Compounds/chemistry , Transformation, Bacterial
16.
Brain Res ; 1404: 63-71, 2011 Aug 02.
Article in English | MEDLINE | ID: mdl-21704983

ABSTRACT

Previously, we have published that pharmacological induction of oxidative stress causes anxiety-like behavior in rats and also is associated with hypertension in these animals. Here, we report that sub-chronic induction of oxidative stress via pharmacological induction leads to i) reduction in glyoxalase (GLO)-1 and glutathione reductase (GSR)-1 expression; ii) calpain mediated reduction of brain derived neurotrophic factor (BDNF) levels; iii) NFκB mediated upregulation of proinflammatory factors interleukin (IL)-6 and tumor necrosis factor (TNF)-α and elevated angiotensin (AT)-1 receptor levels in hippocampus, amygdala and locus coeruleus regions of the brain. Acute oxidative stress has opposite effects. We speculate that regulation of GLO1, GSR1, BDNF, NFκB and AT-1 receptor may contribute to anxiety-like behavior and hypertension in rats.


Subject(s)
Anxiety/pathology , Brain/metabolism , Gene Expression Regulation/physiology , Hypertension/pathology , Inflammation/complications , Oxidative Stress/physiology , Analysis of Variance , Animals , Anxiety/chemically induced , Brain-Derived Neurotrophic Factor/metabolism , Buthionine Sulfoximine/adverse effects , Calpain/metabolism , Disease Models, Animal , Enzyme-Linked Immunosorbent Assay/methods , Gene Expression Regulation/drug effects , Glutathione Reductase/metabolism , Hypertension/chemically induced , Interleukin-6/metabolism , Lactoylglutathione Lyase/metabolism , Male , Oxidative Stress/drug effects , Rats , Rats, Sprague-Dawley , Receptor, Angiotensin, Type 1/metabolism , Signal Transduction/drug effects , Time Factors , Tumor Necrosis Factor-alpha/metabolism , Xanthine/adverse effects , Xanthine Oxidase/adverse effects
17.
J Inorg Biochem ; 104(8): 858-62, 2010 Aug.
Article in English | MEDLINE | ID: mdl-20427090

ABSTRACT

Tetrathiomolybdate (TTM) is a powerful and selective copper (Cu) chelator that is used as a therapeutic agent for Wilson disease. TTM is the sole agent that can remove Cu bound to metallothionein (MT) in the livers of Long-Evans rats with a cinnamon-like coat color (LEC rats). However, the administration of excess TTM causes the deposition of Cu and molybdenum (Mo) in the liver. In the present study, the effect of hepatic glutathione (GSH) depletion on the removal of Cu from the livers of LEC rats was evaluated to establish an effective therapy by TTM. Pretreatment with l-buthionine sulfoximine (BSO), a depletor of GSH in vivo, reduced the amounts of Cu and Mo excreted into both the bile and the bloodstream, and increased the amounts of Cu and Mo deposited in the livers of LEC rats in the form of an insoluble complex 4h after the TTM injection. The results suggest that GSH depletion creates an oxidative environment in the livers of LEC rats, and the oxidative environment facilitates the insolubilization of Cu and Mo in the livers of LEC rats after the TTM injection. Therefore, the effect of TTM on the removal of Cu from the liver was reduced in the oxidized condition. Wilson disease patients and LEC rats develop liver injury caused by oxidative damage. From a clinical viewpoint, increasing in the GSH concentration is expected to enhance the effect of TTM.


Subject(s)
Chelating Agents/pharmacology , Copper/metabolism , Glutathione/metabolism , Hepatolenticular Degeneration/drug therapy , Hepatolenticular Degeneration/metabolism , Liver/metabolism , Molybdenum/pharmacology , Animals , Buthionine Sulfoximine/adverse effects , Buthionine Sulfoximine/pharmacology , Chelating Agents/adverse effects , Enzyme Inhibitors/adverse effects , Enzyme Inhibitors/pharmacology , Humans , Male , Metallothionein/metabolism , Molybdenum/adverse effects , Molybdenum/metabolism , Oxidation-Reduction/drug effects , Rats , Rats, Inbred LEC , Rats, Wistar , Time Factors
18.
Clin Calcium ; 17(6): 887-91, 2007 Jun.
Article in Japanese | MEDLINE | ID: mdl-17548928

ABSTRACT

The increasing incidence of steroid-induced osteonecrosis of the femoral head is a recent and emerging problem. In-depth pathogenesis, however, has not been well understood. We focused on oxidative stress, which was reported to be involved in many diseases, and conducted animal studies to investigate it. Our results from the rabbit model of steroid-induced osteonecrosis demonstrated that administration of steroids caused oxidative damage in bones, and the administration of glutathione reduced the incidence of osteonecrosis. We were also the first to have successfully induced osteonecrosis in rats by administering buthionine sulfoximine (BSO) . The results indicated the involvement of oxidative stress in the development of osteonecrosis and may contribute to elucidating the underlying mechanisms and prevention of the pathogenesis of steroid-induced osteonecrosis.


Subject(s)
Osteonecrosis/etiology , Oxidative Stress/physiology , Animals , Antioxidants/therapeutic use , Buthionine Sulfoximine/adverse effects , Disease Models, Animal , Female , Glutathione/therapeutic use , Humans , Osteonecrosis/prevention & control , Rabbits , Rats , Rats, Wistar
19.
Neurosci Lett ; 417(3): 266-70, 2007 May 07.
Article in English | MEDLINE | ID: mdl-17400377

ABSTRACT

Oxidative stress affects numerous intracellular macromolecules, and may result in cell death unless precisely regulated. Unregulated oxidative stress can be controlled by various cellular defense mechanisms such as glutathione (GSH) which can critically counteract the damaging effects of oxidative stress in mammalian cells. We determined the effects of unregulated oxidative stress induced by GSH depletion on cells in mouse retina. Mice were intraperitoneally injected with buthionine sulphoximine (BSO) at 1.5 g/kg. After 0, 1, 4, and 7 days of BSO administration, retinas were excised and sections were subjected to GSH assay and terminal uridine deoxynucleotidyl nick end labeling (TUNEL) analysis. After 4 days of BSO administration, the number of TUNEL positive cells was significantly increased. However, after 7 days, TUNEL positive cells returned to the basal level. The retinal region most affected by the BSO treatment appeared to be the outer nuclear layer where the photoreceptor cells reside. Different from cells in other regions, retinal cells in the inner nuclear layer increased in their apoptosis even after the first day of BSO injection, and the increase was further potentiated after 4 days. Taken together, our studies suggested that GSH depletion may cause unregulated oxidative stress to the cells in the retina and indeed increased cell death in the retina. The cells in the inner nuclear layer seemed to be affected earlier than the cells in other layers of the retina. The GSH level in the retina may be a crucial therapeutic target in preventing blindness.


Subject(s)
Apoptosis/physiology , Glutathione/deficiency , Neurons/metabolism , Oxidative Stress/physiology , Retina/metabolism , Animals , Antimetabolites/adverse effects , Apoptosis/drug effects , Buthionine Sulfoximine/adverse effects , Cell Count , In Situ Nick-End Labeling , Male , Mice , Mice, Inbred C57BL , Neurons/drug effects , Oxidative Stress/drug effects , Photoreceptor Cells/drug effects , Photoreceptor Cells/metabolism , Photoreceptor Cells/physiopathology , Retina/drug effects , Retina/physiopathology , Retinal Degeneration/metabolism , Retinal Degeneration/physiopathology , Retinal Degeneration/prevention & control , Up-Regulation/drug effects , Up-Regulation/physiology
20.
J Pharm Pharmacol ; 56(1): 91-9, 2004 Jan.
Article in English | MEDLINE | ID: mdl-14983899

ABSTRACT

Antioxidant therapy can improve the protection and metabolic activity of cells and tissues. In this study, the effect of vitamin E administration on buthionine sulfoximine (BSO)-induced glutathione (GSH) depletion in the rat lung and liver was investigated. Hepatic GSH was depleted by intraperitoneal administration of BSO (4 mmol kg(-1)), twice a day, for 30 days to rats. We also investigated whether the lung and liver mitochondrial GSH contents were influenced by BSO administration and whether an extracellular supply of vitamin E could prevent the changes caused by BSO-mediated GSH depletion. Glutathione levels in lung and liver tissues were depleted by 47% and 60%, respectively. Depletion of hepatic and pulmonary GSH in turn causes decline in the levels of mitochondrial GSH, leading to impaired antioxidant defence function of mitochondria. Both the cytosolic and mitochondrial glutathione disulfides (GSSG) were altered during BSO treatment, and led to drastic increase in GSSG/GSH redox status. One of the experimental groups was given vitamin E (65 mg (kg diet)(-1)) mixed with rat feed. The rats fed with vitamin E were found to have partially restored GSH levels in liver and lung, diminished levels of TBARS and minimized tissue damage. The current findings suggest that the impaired glutathione and glutathione-dependent enzyme status may be correlated with the elevated lipid peroxidation and mitochondrial membrane damage and that vitamin E therapy to the BSO-administered rats prevents the above changes. However, vitamin E did not have any effect on the activity of gamma-glutamyl cysteine synthetase (gamma-GCS).


Subject(s)
Antimetabolites, Antineoplastic/adverse effects , Antioxidants/therapeutic use , Buthionine Sulfoximine/adverse effects , alpha-Tocopherol/therapeutic use , Animals , Antioxidants/metabolism , Glutathione/deficiency , Glutathione/metabolism , Lipid Peroxidation/drug effects , Liver/drug effects , Liver/pathology , Liver/ultrastructure , Lung/drug effects , Lung/pathology , Lung/ultrastructure , Male , Mitochondria/drug effects , Mitochondria/metabolism , Rats , Thiobarbituric Acid Reactive Substances/metabolism , alpha-Tocopherol/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...