Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 10 de 10
Filter
Add more filters










Publication year range
1.
Neuromolecular Med ; 19(1): 1-10, 2017 Mar.
Article in English | MEDLINE | ID: mdl-27263112

ABSTRACT

As a key regulator of cell metabolism and survival, mechanistic target of rapamycin (mTOR) emerges as a novel therapeutic target for Parkinson's disease (PD). A growing body of research indicates that restoring perturbed mTOR signaling in PD models can prevent neuronal cell death. Nevertheless, molecular mechanisms underlying mTOR-mediated effects in PD have not been fully understood yet. Here, we review recent progress in characterizing the association of mTOR signaling with PD risk factors and further discuss the potential roles of mTOR in PD.


Subject(s)
Nerve Tissue Proteins/physiology , Parkinson Disease/metabolism , Signal Transduction/physiology , TOR Serine-Threonine Kinases/physiology , Animals , Apoptosis , Autophagy , Dopaminergic Neurons/metabolism , Dopaminergic Neurons/pathology , Genetic Predisposition to Disease , Humans , Mechanistic Target of Rapamycin Complex 1/metabolism , Mechanistic Target of Rapamycin Complex 2/metabolism , Neurotoxins/toxicity , Oxidative Stress , Parkinson Disease/etiology , Parkinson Disease/genetics , Parkinsonian Disorders/chemically induced , Parkinsonian Disorders/metabolism , Risk Factors , alpha-Synuclein/physiology
2.
Neurotox Res ; 30(3): 499-509, 2016 10.
Article in English | MEDLINE | ID: mdl-27435481

ABSTRACT

The involvement of copper in the pathophysiology of neurodegenerative disorders has been documented but remains poorly understood. This study aimed at investigating the molecular mechanism underlying copper-induced neurotoxicity. Human neuroblastoma SH-SY5Y cells were treated with different concentrations of Cu(II) (25-800 µM). The relative levels of AMPKα, phosphorylated (p)-AMPKα were examined by western blotting. The results showed that copper reduced cell viability and enhanced apoptosis of SH-SY5Y cells. Pretreatment with N-acetyl-L-cysteine, a common ROS scavenger, decreased copper-induced cytotoxicity. Furthermore, the levels of p-AMPKα in SH-SY5Y cells were increased by a relatively low concentration of copper and decreased by a relatively high concentration of copper at 24 h. Moreover, inhibition of AMPK with compound C or RNA interference aggravated concentration-dependent cytotoxicity of Cu(II). Taken together, these results indicated that AMPK activity might be important for the neurotoxicity of Cu(II).


Subject(s)
AMP-Activated Protein Kinases/antagonists & inhibitors , Copper/toxicity , Neurons/drug effects , AMP-Activated Protein Kinases/genetics , AMP-Activated Protein Kinases/metabolism , Acetylcysteine/pharmacology , Apoptosis/drug effects , Apoptosis/physiology , Cell Line, Tumor , Cell Survival/drug effects , Cell Survival/physiology , Dose-Response Relationship, Drug , Enzyme Inhibitors/pharmacology , Free Radical Scavengers/pharmacology , Gene Knockdown Techniques , Humans , Neurons/metabolism , Neurons/pathology , RNA Interference , Reactive Oxygen Species/metabolism
3.
Mol Med Rep ; 5(1): 250-5, 2012 Jan.
Article in English | MEDLINE | ID: mdl-21993612

ABSTRACT

The present study investigated whether there is an interaction between reactive oxygen species (ROS) and p38 mitogen-activated protein kinase (MAPK) during chemical hypoxia-induced injury in PC12 cells. The results of the present study showed that cobalt chloride (CoCl2), a chemical hypoxia agent, markedly induced ROS generation and phosphorylation of p38MAPK, as well as neuronal injuries. N-acetylcysteine (NAC), a ROS scavenger, blocked CoCl2-induced phosphorylation of p38MAPK. In addition, SB203580, an inhibitor of p38MAPK attenuated not only CoCl2-induced activation of p38MAPK, but also ROS production. These results suggest that ROS and p38MAPK are capable of interacting positively during chemical hypoxia. Furthermore, NAC and SB203580 markedly prevented CoCl2-induced cytotoxicity, apoptosis and a loss of mitochondrial membrane potential. Taken together, our findings suggest that the positive interaction between CoCl2 induction of ROS and p38MAPK activation may play a significant role in CoCl2-induced neuronal injuries. We provide new insights into the mechanisms responsible for CoCl2-induced injuries in PC12 cells.


Subject(s)
Reactive Oxygen Species/metabolism , p38 Mitogen-Activated Protein Kinases/metabolism , Animals , Apoptosis , Cell Hypoxia/drug effects , Cobalt/toxicity , Enzyme Inhibitors/pharmacology , Imidazoles/pharmacology , Membrane Potential, Mitochondrial , PC12 Cells , Phosphorylation , Pyridines/pharmacology , Rats , Reactive Oxygen Species/antagonists & inhibitors
4.
Mol Cell Biochem ; 362(1-2): 149-57, 2012 Mar.
Article in English | MEDLINE | ID: mdl-22134701

ABSTRACT

Hydrogen sulfide (H(2)S) has been shown to exert cardioprotective effects. However, the roles of extracellular signal-regulated protein kinases 1/2 (ERK1/2) in H(2)S-induced cardioprotection have not been completely elucidated. In this study, cobalt chloride (CoCl(2)), a chemical hypoxia mimetic agent, was applied to treat H9c2 cells to establish a chemical hypoxia-induced cardiomyocyte injury model. The results showed that pretreatment with NaHS (a donor of H(2)S) before exposure to CoCl(2) attenuated the decreased cell viability, the increased apoptosis rate, the loss of mitochondrial membrane potential (ΔΨm), and the intracellular accumulation of reactive oxygen species (ROS) in H9c2 cells. Exposure of H9c2 cells to CoCl(2) or hydrogen peroxide (H(2)O(2)) upregulated expression of phosphorylated (p) ERK1/2, which was reduced by pretreatment with NaHS or N-acetyl-L-cysteine, a ROS scavenger. More importantly, U0126, a selective inhibitor of ERK1/2, mimicked the above cytoprotection of H(2)S against CoCl(2)-induced injury in H9c2 cells. In conclusion, these results indicate that H(2)S protects H9c2 cells against chemical hypoxia-induced injury partially by inhibiting ROS-mediated activation of ERK1/2.


Subject(s)
Cell Hypoxia , Extracellular Signal-Regulated MAP Kinases/metabolism , Hydrogen Sulfide/pharmacology , Oxidative Stress/drug effects , Reactive Oxygen Species/metabolism , Animals , Apoptosis/drug effects , Butadienes/pharmacology , Cardiotonic Agents/pharmacology , Cell Hypoxia/drug effects , Cell Line , Cell Survival/drug effects , Cobalt/pharmacology , Extracellular Signal-Regulated MAP Kinases/antagonists & inhibitors , Hydrogen Peroxide/pharmacology , Membrane Potential, Mitochondrial/drug effects , Nitriles/pharmacology , Rats
5.
Mol Cell Biochem ; 363(1-2): 419-26, 2012 Apr.
Article in English | MEDLINE | ID: mdl-22203419

ABSTRACT

The roles of hydrogen sulfide (H(2)S) and endoplasmic reticulum (ER) stress in doxorubicin (DOX)-induced cardiotoxicity are still unclear. This study aimed to dissect the hypothesis that H(2)S could protect H9c2 cells against DOX-induced cardiotoxicity by inhibiting ER stress. Our results showed that exposure of H9c2 cells to DOX significantly inhibited the expression and activity of cystathionine-γ-lyase (CSE), a synthetase of H(2)S, accompanied by the decreased cell viability and the increased reactive oxygen species (ROS) accumulation. In addition, exposure of cells to H(2)O(2) (an exogenous ROS) mimicked the inhibitory effect of DOX on the expression and activity of CSE. Pretreatment with N-acetyl-L: -cysteine (NAC) (a ROS scavenger) attenuated intracellular ROS accumulation, cytotoxicity, and the inhibition of expression and activity of CSE induced by DOX. Notably, the ER stress-related proteins, including glucose-regulated protein 78 (GRP78) and C/EBP homologous protein (CHOP) were obviously upregulated in DOX-treated H9c2 cells. Pretreatment with sodium hydrosulfide (NaHS, a H(2)S donor) before DOX exposure markedly suppressed DOX-induced overexpressions of GRP78 and CHOP, cytotoxicity and oxidative stress. In conclusion, we have demonstrated that ROS-mediated inhibition of CSE is involved in DOX-induced cytotoxicity in H9c2 cells, and that exogenous H(2)S can confer protection against DOX-induced cardiotoxicity partly through inhibition of ER stress.


Subject(s)
Antibiotics, Antineoplastic/toxicity , Antioxidants/pharmacology , Doxorubicin/toxicity , Endoplasmic Reticulum Stress/drug effects , Hydrogen Sulfide/metabolism , Myocytes, Cardiac/drug effects , Sulfides/pharmacology , Acetylcysteine/pharmacology , Animals , Antioxidants/metabolism , Cell Line , Cell Survival/drug effects , Cystathionine gamma-Lyase/metabolism , Cytoprotection , Dose-Response Relationship, Drug , Free Radical Scavengers/pharmacology , Heat-Shock Proteins/metabolism , Hydrogen Peroxide/toxicity , Myocytes, Cardiac/metabolism , Myocytes, Cardiac/pathology , Oxidants/toxicity , Rats , Reactive Oxygen Species/metabolism , Sulfides/metabolism , Time Factors , Transcription Factor CHOP/metabolism
6.
Nan Fang Yi Ke Da Xue Xue Bao ; 31(11): 1949-53, 2011 Nov.
Article in Chinese | MEDLINE | ID: mdl-22126789

ABSTRACT

OBJECTIVE: To explore the role of bone morphogenetic protein-7 (BMP-7) in strontium ranelate (Sr)-induced osteogenic differentiation of bone marrow mesenchymal stem cells (BMSCs). METHODS: BMSCs were isolated from 4-week-old rats and cultured in vitro. The third or fourth passages of BMSCs were examined using alkaline phosphatase kit for changes in ALP activity in response to treatment with different concentrations of Sr. Calcium nodules in the induced cells were detected using alizarin red staining, and the cellular BMP-7 expression was detected by Western blotting. RESULTS: Within the concentration range of 0.1-3.0 mmol/L, Sr dose-dependently increased ALP activity in rat BMSCs. ALP activity reached the highest level after treatment with 3 mmol/L Sr, which also significantly promoted the formation of calcium nodules. Within the range of 0.1-3.0 mmol/L, Sr dose-dependently enhanced the expression of BMP-7, and its peak expression occurred following 3 mmol/L Sr treatment. Noggin (100 ng/ml), an inhibitor of BMP-7, obviously suppressed Sr-induced over-expression of BMP-7 and reduced ALP activity and calcium nodule formation in the BMSCs. CONCLUSION: Sr promotes osteogenic differentiation of rat BMSCs by increasing the expression of BMP-7.


Subject(s)
Bone Marrow Cells/cytology , Bone Morphogenetic Protein 7/metabolism , Mesenchymal Stem Cells/cytology , Organometallic Compounds/pharmacology , Osteoblasts/cytology , Thiophenes/pharmacology , Animals , Bone Density Conservation Agents/pharmacology , Bone Morphogenetic Protein 7/genetics , Cell Differentiation/drug effects , Cells, Cultured , Female , Male , Mesenchymal Stem Cells/metabolism , Osteogenesis/drug effects , Rats
7.
Brain Inj ; 25(6): 604-13, 2011.
Article in English | MEDLINE | ID: mdl-21534738

ABSTRACT

PRIMARY OBJECTIVE: Recent evidence suggests that delayed hypoxic post-conditioning is neuroprotective. The aim of the present study was to test whether early post-conditioning applied immediately after hypoxia could protect cultured neurons from hypoxia/reoxygenation (H/R)-induced injuries. METHODS: Primary cortical neuronal culture depleted of microglia was exposed to H/R. Post-conditioning started immediately after hypoxia and consisted of three cycles of 15-minutes of reoxygenation and 15-minutes of hypoxia. Cell viability assay was performed using Cell Counting Kit-8 (CCK-8). Apoptosis was evaluated by Hoechst 33258 staining, FITC-Annexin V/PI double staining and Western blot assay (testing the cleaved caspase-3 expression). Reactive oxygen species (ROS), intracellular Ca(2+) and mitochondrial membrane potential (MMP) were examined using confocal laser-scanning microscopy. MAIN RESULTS: H/R significantly reduced cell viability and increased neuronal apoptosis and necrosis. Furthermore, the expression of cleaved caspase-3, ROS production and intracellular Ca(2+) were increased. MMP was attenuated. Injuries induced by H/R were substantially attenuated by early hypoxic post-conditioning. Changes in cleaved caspase-3 expression, ROS production, intracellular Ca(2+) level and MMP in response to H/R were significantly decreased by the post-conditioning. CONCLUSIONS: The findings demonstrated that early hypoxic post-conditioning could protect neurons against H/R-induced injuries independent of microglial cells, possibly by inhibiting ROS over-production and intracellular Ca(2+) accumulation and maintaining MMP.


Subject(s)
Apoptosis/physiology , Cell Hypoxia/physiology , Cell Survival/physiology , Cerebral Cortex/metabolism , Neurons/physiology , Reactive Oxygen Species/metabolism , Animals , Blotting, Western , Cells, Cultured , Cerebral Cortex/physiopathology , Rats , Rats, Sprague-Dawley
8.
Clin Exp Pharmacol Physiol ; 38(1): 42-9, 2011 Jan.
Article in English | MEDLINE | ID: mdl-21083699

ABSTRACT

1. Increasing evidence indicates that hydrogen sulphide (H2S) may serve as an important biological cytoprotective agent. Heat shock protein (Hsp) 90 can attenuate stress-induced injury. However, whether Hsp90 mediates the cytoprotective effect of H2S against chemical hypoxia-induced injury in PC12 cells is not known. 2. In the present study, CoCl2 (a chemical hypoxia mimetic) was used to treat PC12 cells to create a model of chemical hypoxia. To explore the role of Hsp90 in the cytoprotection afforded by H2S against chemical hypoxia-induced injury, 2 µmol/L 17-allylaminogeldanamycin (17-AAG), a selective inhibitor of Hsp90, was administered for 30 min prior to preconditioning with 400 µmol/L NaHS, followed by chemical hypoxia. 3. Cobalt chloride reduced cell viability (by 52.7 ± 1.5%), increased PC12 cell apoptosis (by 42.1 ± 1.5%), induced reactive oxygen species (ROS) by 3.79% compared with control and induced the dissipation of mitochondrial membrane potential (MMP) by 2.56% compared with control. 4. Pretreatment of PC12 cells with 100-400 µmol/L sodium hydrosulphide (NaHS), an H2S donor, for 3 h prior to exposure to 600 µmol/L CoCl2 provided significant, concentration-dependant protection to PC12 cells against CoCl2-induced cytotoxicity. Specifically, pretreatment of PC12 cells with 400 µmol/L NaHS decreased apoptosis to 16.77 ± 1.77% and blocked the CoCl2-induced increase in ROS production and loss of MMP. 5. At 400 µmol/L, NaHS upregulated Hsp90 in a time-dependant manner (over the period 0-180 min). In addition to its effects on Hsp90 expression, NaHS pretreatment of PC12 cells augmented the overexpression of Hsp90 induced by 600 µmol/L CoCl2 by 1.38-fold (P < 0.01). 6. Treatment of PC12 cells with 2 µmol/L 17-AAG for 30 min prior to NaHS pretreatment blocked the overexpression of Hsp90 induced by NaHS preconditioning, as evidenced by decreased cell viability (by 54.2 + 1.2%; P < 0.01), increased PC12 cell apoptosis (by 36.6 ± 1.2%; P < 0.01) and increasing ROS production. 7. The findings of the present study provide novel evidence that Hsp90 mediates H2S-induced neuroprotection against chemical hypoxia-induced injury via anti-oxidant and anti-apoptotic effects.


Subject(s)
Apoptosis/drug effects , Cytoprotection/drug effects , HSP90 Heat-Shock Proteins/physiology , Hydrogen Sulfide/pharmacology , Hypoxia/complications , Animals , Antioxidants/pharmacology , Cell Hypoxia/drug effects , Cobalt , Cytotoxins , Down-Regulation/drug effects , Drug Evaluation, Preclinical , HSP90 Heat-Shock Proteins/metabolism , Hypoxia/chemically induced , Hypoxia/metabolism , Hypoxia/pathology , Membrane Potential, Mitochondrial/drug effects , Neuroprotective Agents/pharmacology , PC12 Cells , Rats , Reactive Oxygen Species/metabolism
9.
Clin Exp Pharmacol Physiol ; 37(3): 316-21, 2010 Mar.
Article in English | MEDLINE | ID: mdl-19769612

ABSTRACT

1. The aim of the present study was to investigate the effect of hydrogen sulphide (H(2)S) on cobalt chloride (CoCl(2))-induced injury in H9c2 embryonic rat cardiac cells. 2. After 36 h incubation in the presence of 600 micromol/L CoCl(2), reduced cell viability of H9c2 cells was observed, as well as the induction of apoptosis. In addition, CoCl(2) (600 micromol/L) enhanced the production of reactive oxygen species (ROS) and the expression of cleaved caspase 3, induced a loss of mitochondrial membrane potential (MMP) and decreased reduced glutathione (GSH) production. These results suggest that CoCl(2) induces similar responses to hypoxia/ischaemia. 3. Pretreatment of cells with 400 micromol/L NaHS (a H(2)S donor) for 30 min prior to exposure to CoCl(2) (600 micromol/L) significantly protected H9c2 cells against CoCl(2)-induced injury. Specifically, increased cell viability and decreased apoptosis were observed. In addition, NaHS pretreatment blocked the CoCl(2)-induced increases in ROS production and cleaved caspase 3 expression, as well as the decreases in GSH production and loss of MMP. 4. Pretreatment of cells with 2000 micromol/L N-acetylcysteine (NAC), a ROS scavenger, for 1 h prior to CoCl(2) exposure significantly protected H9c2 cells against CoCl(2)-induced injury, specifically enhancing cell viability, decreasing ROS production and preventing loss of MMP. 5. The findings of the present study suggest that H(2)S protects H9c2 cells against CoCl(2)-induced injury by suppressing oxidative stress and caspase 3 activation.


Subject(s)
Cobalt/toxicity , Cytoprotection/drug effects , Cytoprotection/physiology , Hydrogen Sulfide/pharmacology , Animals , Cell Hypoxia/drug effects , Cell Hypoxia/physiology , Cell Line , Cell Survival/drug effects , Cell Survival/physiology , Cells, Cultured , Myocardium/cytology , Oxidative Stress/drug effects , Oxidative Stress/physiology , Rats , Reactive Oxygen Species/metabolism
10.
Nan Fang Yi Ke Da Xue Xue Bao ; 29(10): 1977-81, 2009 Oct.
Article in Chinese | MEDLINE | ID: mdl-19861245

ABSTRACT

OBJECTIVE: To investigate the protective effect of reactive oxygen species (ROS) scavenger, N-acetyl-L-cysteine (NAC), against H9c2 cardiomyocytes from injuries induced by chemical hypoxia. METHODS: H9c2 cells were treated with cobalt chloride (CoCl2), a chemical hypoxia-mimetic agent, to establish the chemical hypoxia-induced cardiomyocyte injury model. NAC was added into the cell medium 60 min prior to CoCl2 exposure. The cell viability was evaluated using cell counter kit (CCK-8), and the intercellular ROS level was measured by 2', 7'- dichlorfluorescein-diacetate (DCFH-DA) staining and photofluorography. Mitochondrial membrane potential (MMP) of the cells was observed by Rhodamine123 (Rh123) staining and photofluorography, and the ratio of GSSG/ (GSSG+GSH) was calculated according to detection results of the GSSG kit. RESULTS: Exposure of H9c2 cardiomyocytes to 600 micromol/L CoCl2 for 36 h resulted in significantly reduced cell viability. Pretreatment with NAC at the concentrations ranging from 500 to 2000 micromol/L 60 min before CoCl2 exposure dose-dependently inhibited CoCl2-induced H9c2 cell injuries, and obviously increased the cell viability. NAC at 2000 micromol/L obviously inhibited the oxidative stress induced by CoCl2, decreased the ratio of GSSG/(GSSG+GSH), increased ROS level, and antagonized CoCl2-induced inhibition on MMP. CONCLUSION: NAC offers obvious protective effect on H9c2 cardiomyocytes against injuries induced by chemical hypoxia by decreasing in the ratio of GSSG/(GSSG+GSH) and ROS level and ameliorating MMP.


Subject(s)
Free Radical Scavengers/pharmacology , Myocytes, Cardiac/drug effects , Oxidative Stress/drug effects , Reactive Oxygen Species/metabolism , Animals , Cell Hypoxia/drug effects , Cells, Cultured , Embryo, Mammalian , Myocytes, Cardiac/metabolism , Myocytes, Cardiac/pathology , Rats
SELECTION OF CITATIONS
SEARCH DETAIL
...