Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 7 de 7
Filter
Add more filters










Database
Language
Publication year range
1.
Int J Mol Med ; 41(4): 1817-1825, 2018 Apr.
Article in English | MEDLINE | ID: mdl-29393357

ABSTRACT

Oxidative stress is increasingly recognized as a central event contributing to the degeneration of dopaminergic neurons in the pathogenesis of Parkinson's disease (PD). Although reactive oxygen species (ROS) production is implicated as a causative factor in PD, the cellular and molecular mechanisms linking oxidative stress with dopaminergic neuron death are complex and not well characterized. The primary insults cause the greatest production of ROS, which contributes to oxidative damage by attacking all macromolecules, including lipids, proteins and nucleic acids, leading to defects in their physiological function. Consequently, the defects in these macromolecules result in mitochondrial dysfunction and neuroinflammation, which subsequently enhance the production of ROS and ultimately neuronal damage. The interaction between these various mechanisms forms a positive feedback loop that drives the progressive loss of dopaminergic neurons in PD, and oxidative stress­mediated neuron damage appears to serve a central role in the neurodegenerative process. Thus, understanding the cellular and molecular mechanisms by which oxidative stress contributes to the loss of dopaminergic neurons may provide a promising therapeutic approach in PD treatment.


Subject(s)
Dopaminergic Neurons/pathology , Mitochondria/pathology , Oxidative Stress , Parkinson Disease/pathology , Animals , Cell Death , Dopaminergic Neurons/metabolism , Humans , Mitochondria/metabolism , Mitochondrial Membrane Transport Proteins/metabolism , Mitochondrial Permeability Transition Pore , Parkinson Disease/metabolism , Reactive Oxygen Species/metabolism
2.
Int J Mol Med ; 41(2): 615-623, 2018 Feb.
Article in English | MEDLINE | ID: mdl-29207041

ABSTRACT

Mitochondria are important organelles in virtually all eukaryotic cells, and are involved in a wide range of physiological and pathophysiological processes. Besides the generation of cellular energy in the form of adenosine triphosphate, mitochondria are also involved in calcium homeostasis, reactive oxygen species production and the activation of the intrinsic cell death pathway, thus determining cell survival and death. Mitochondrial abnormalities have been implicated in a wide range of disorders, including neurodegenerative disease such as Parkinson's disease (PD), and considered as a primary cause and central event responsible for the progressive loss of dopaminergic neurons in PD. Thus, reversion or attenuation of mitochondrial dysfunction should alleviate the severity or progression of the disease. The present review systematically summarizes the possible mechanisms associated with mitochondria­mediated dopaminergic neuron damage in PD, in an attempt to elucidate the requirement for further studies for the development of effective PD treatments.


Subject(s)
Dopaminergic Neurons/metabolism , Mitochondria/genetics , Parkinson Disease/genetics , Calcium/metabolism , Cell Death/genetics , Dopaminergic Neurons/pathology , Humans , Mitochondria/metabolism , Mitochondria/pathology , Parkinson Disease/metabolism , Parkinson Disease/pathology , Reactive Oxygen Species/metabolism , Signal Transduction/genetics
3.
Int J Mol Med ; 37(2): 429-35, 2016 Feb.
Article in English | MEDLINE | ID: mdl-26677001

ABSTRACT

Oxidative stress is widely considered as a central event in the pathogenesis of Parkinson's disease (PD). The mechanisms underlying the oxidative damage-mediated loss of dopaminergic neurons in PD are not yet fully understood. Accumulating evidence has indicated that oxidative DNA damage plays a crucial role in programmed neuronal cell death, and is considered to be at least partly responsible for the degeneration of dopaminergic neurons in PD. This process involves a number of signaling cascades and molecular proteins. Proliferating cell nuclear antigen (PCNA) is a pleiotropic protein affecting a wide range of vital cellular processes, including chromatin remodelling, DNA repair and cell cycle control, by interacting with a number of enzymes and regulatory proteins. In the present study, the exposure of PC12 cells to 1-methyl-4-phenylpyridinium (MPP+) led to the loss of cell viability and decreased the expression levels of PCNA in a dose- and time-dependent manner, indicating that this protein may be involved in the neurotoxic actions of MPP+ in dopaminergic neuronal cells. In addition, a significant upregulation in p53 expression was also observed in this cellular model of PD. p53 is an upstream inducer of PCNA and it has been recognized as a key contributor responsible for dopaminergic neuronal cell death in mouse models of 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP)-induced PD. This indicates that MPP+-induced oxidative damage is mediated by the downregulation of PCNA through the p53 pathway in a cellular model of PD. Thus, our results may provide some novel insight into the molecular mechanisms responsible for the development of PD and provide new possible therapeutic targets for the treatment of PD.


Subject(s)
Parkinson Disease/genetics , Proliferating Cell Nuclear Antigen/biosynthesis , Tumor Suppressor Protein p53/biosynthesis , 1-Methyl-4-phenylpyridinium/administration & dosage , Animals , DNA Damage/genetics , Disease Models, Animal , Dopaminergic Neurons/metabolism , Dopaminergic Neurons/pathology , Gene Expression Regulation/drug effects , Humans , Mice , Oxidative Stress/drug effects , Oxidative Stress/genetics , PC12 Cells , Parkinson Disease/pathology , Proliferating Cell Nuclear Antigen/genetics , Rats , Tumor Suppressor Protein p53/genetics
4.
Mol Med Rep ; 12(6): 7941-8, 2015 Dec.
Article in English | MEDLINE | ID: mdl-26497173

ABSTRACT

Endothelial dysfunction and injury are central events in the pathogenesis of ischemic vascular disorders. Endothelial progenitor cells (EPCs) are mobilized from the bone marrow into the peripheral circulation, where they locate to sites of injured endothelium and are involved in endothelial repair and vascular regeneration. During these processes, EPCs are exposed to oxidative stress, a crucial pathological condition, which occurs during vascular injury and limits the efficacy of EPCs in the repair of injured endothelium. Statins are effective inhibitors of 3­hydroxy­3­methylglutaryl coenzyme A reductase, and are commonly used to manage and prevent ischemic vascular disease by reducing plasma cholesterol levels. In addition to lowering cholesterol, statins have also been reported to exert pleiotropic actions, including anti­inflammatory and anti­oxidative activities. The present study aimed to investigate the ability of atorvastatin to protect endothelial colony­forming cells (ECFCs), a homogeneous subtype of EPCs, from hydrogen peroxide (H2O2)­induced oxidative damage, and to determine the mechanism underlying this protective action. MTT assay, acridine orange/ethidium bromide staining, reactive oxygen species assay, western blot analysis and tube formation assay were employed. The results demonstrated that H2O2 induced cell death and decreased the tube­forming ability of the ECFCs, in a concentration­dependent manner; however, these effects were partially attenuated following administration of atorvastatin. The reversion of the quantitative and qualitative impairment of the H2O2­treated ECFCs appeared to be mediated by the regulation of annexin A2, as the expression levels of annexin A2 were decreased following treatment with H2O2 and increased following treatment with atorvastatin. These results indicated that annexin A2 may be involved in the H2O2­induced damage of ECFCs, and in the protective activities of atorvastatin in response to oxidative stress.


Subject(s)
Annexin A2/genetics , Atorvastatin/pharmacology , Endothelial Cells/drug effects , Oxidative Stress/drug effects , Animals , Annexin A2/metabolism , Annexin A2/physiology , Apoptosis/drug effects , Gene Expression Regulation/drug effects , Humans , Hydrogen Peroxide , Rats
5.
Int J Mol Med ; 34(5): 1358-64, 2014 Nov.
Article in English | MEDLINE | ID: mdl-25174304

ABSTRACT

The mitochondria are the most important cytoplasmic organelles in determining cell survival and death. Mitochondrial dysfunction leads to a wide range of disorders, including neurodegenerative diseases. The central events in the mitochondrial­dependent cell death pathway are the activation of the mitochodrial permeability transition pore (mPTP) and the disruption of mitochondrial membrane potential, which cause the release of apoptogenic molecules and finally lead to cell death. This is thought to be at least partly responsible for the loss of dopaminergic neurons in Parkinson's disease (PD); thus, the attenuation of mitochondrial dysfunction may contribute to alleviating the severity and progression of this disease. Guanosine is a pleiotropic molecule affecting multiple cellular processes, including cellular growth, differentiation and survival. Its protective effects on the central nervous system and and on several cell types by inhibiting apoptosis have been shown in a number of pathological conditions. This study aimed to analyze the ability of guanosine to protect neuronal PC12 cells from the toxicity induced by 1-methyl-4-phenylpyridinium (MPP+), the active metabolite of 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP), which mediates selective damage to dopaminergic neurons and causes irreversible Parkinson-like symptoms in humans and primates. Our results demonstrated that the apoptosis of PC12 cells induced by MPP+ was significantly prevented by pre-treatment for 3 h with guanosine. In addition, guanosine attenuated the MPP+-induced collapse of mitochondrial transmembrane potential and prevented the sebsequent activation of caspase-3, thereby protecting dopaminergic neurons against mitochondrial stress-induced damage.


Subject(s)
Guanosine/pharmacology , Mitochondria/drug effects , Neuroprotective Agents/pharmacology , Parkinson Disease/drug therapy , 1-Methyl-4-phenyl-1,2,3,6-tetrahydropyridine/adverse effects , 1-Methyl-4-phenylpyridinium/adverse effects , Animals , Apoptosis/drug effects , Caspase 3/genetics , Caspase 3/metabolism , Cell Survival/drug effects , Dopaminergic Neurons/drug effects , Dopaminergic Neurons/metabolism , Gene Expression Regulation , Membrane Potential, Mitochondrial/drug effects , Mitochondria/metabolism , Oxidative Stress/drug effects , PC12 Cells , Rats , Reactive Oxygen Species/metabolism , bcl-2-Associated X Protein/genetics , bcl-2-Associated X Protein/metabolism
6.
Mol Med Rep ; 9(6): 2043-50, 2014 Jun.
Article in English | MEDLINE | ID: mdl-24681994

ABSTRACT

Glycogen synthase kinase-3 (GSK-3) is a pleiotropic serine/threonine protein kinase found in almost all eukaryotes. It is structurally highly conserved and has been identified as a multifaceted enzyme affecting a wide range of biological functions, including gene expression and cellular processes. There are two closely related isoforms of GSK-3; GSK-3α and GSK-3ß. The latter appears to play crucial roles in regulating the pathogenesis of diverse diseases, including neurodegenerative disease. The present review focuses on the involvement of this protein in Parkinson's disease (PD), a common neurodegenerative disorder characterized by the gradually progressive and selective loss of dopaminergic neurons, and by intracellular inclusions known as Lewy bodies (LBs) expressed in surviving neurons of the substantia nigra (SN). GSK-3ß is involved in multiple signaling pathways and has several phosphorylation targets. Numerous apoptotic conditions can be facilitated by the GSK-3ß signaling pathways. Studies have shown that GSK-3ß inhibition protects the dopaminergic neurons from various stress-induced injuries, indicating the involvement of GSK-3ß in PD pathogenesis. However, the underlying mechanisms of the protective effect of GSK-3ß inhibition on dopaminergic neurons in PD is not completely understood. Multiple pathological events have been recognized to be responsible for the loss of dopaminergic neurons in PD, including mitochondrial dysfunction, oxidative stress, protein aggregation and neuroinflammation. The present review stresses the regulatory roles of GSK-3ß in these events and in dopaminergic neuron degeneration, in an attempt to gain an improved understanding of the underlying mechanisms and to provide a potential effective therapeutic target for PD.


Subject(s)
Glycogen Synthase Kinase 3/metabolism , Parkinson Disease/enzymology , Animals , Apoptosis , Electron Transport Complex I/metabolism , Fetal Proteins/genetics , Fetal Proteins/metabolism , Gene Expression Regulation , Glycogen Synthase Kinase 3/genetics , Glycogen Synthase Kinase 3 beta , Humans , Inflammation/genetics , Inflammation/metabolism , Mitochondria/metabolism , Parkinson Disease/genetics , Parkinson Disease/metabolism , Reactive Oxygen Species/metabolism , Signal Transduction , T-Box Domain Proteins/genetics , T-Box Domain Proteins/metabolism , alpha-Synuclein/genetics , alpha-Synuclein/metabolism
7.
Int J Mol Med ; 32(1): 108-14, 2013 Jul.
Article in English | MEDLINE | ID: mdl-23615851

ABSTRACT

Reactive oxygen species (ROS) elicited by oxidative stress are widely recognized as a major initiator in the dege-neration of dopaminergic neurons distinctive of Parkinson's disease (PD). The interaction of ROS with mitochondria triggers sequential events in the mitochondrial cell death pathway, which is thought to be responsible for ROS-mediated neurodegeneration in PD. α-lipoic acid (LA) is a pleiotropic compound with potential pharmacotherapeutic value against a range of pathophysiological insults. Its protective actions against oxidative damage by scavenging ROS and reducing production of free radicals have been reported in various in vitro and in vivo systems. This study analyzed the ability of LA to protect PC12 neuronal cells from toxicity of 1-methyl-4-phenylpyridinium (MPP+), the neurotoxic metabolite of 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP) which is known to kill dopaminergic neurons selectively and to cause severe parkinsonism-like symptoms in humans and primate animals. Our results demonstrate that the apoptosis of PC12 cells elicited by MPP+ could be significantly prevented by pretreatment with LA for 1 h. In addition, LA inhibits intercellular ROS levels and the mitochondrial transmembrane permeability, the key players in the pathogenesis of PD, thereby protecting dopaminergic neuronal cells against oxidative damage.


Subject(s)
1-Methyl-4-phenylpyridinium/toxicity , Apoptosis/drug effects , Dopaminergic Neurons/drug effects , Dopaminergic Neurons/metabolism , Neuroprotective Agents/pharmacology , Reactive Oxygen Species/metabolism , Thioctic Acid/pharmacology , Animals , Caspase 3/metabolism , Cell Survival/drug effects , Membrane Potential, Mitochondrial/drug effects , PC12 Cells , Proto-Oncogene Proteins c-bcl-2/metabolism , Rats , bcl-2-Associated X Protein/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...