1
|
Allboani A, Kar S, Kavdia M. Computational modeling of neuronal nitric oxide synthase biochemical pathway: A mechanistic analysis of tetrahydrobiopterin and oxidative stress. Free Radic Biol Med 2024; 222:625-637. [PMID: 39004235 DOI: 10.1016/j.freeradbiomed.2024.07.011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/19/2024] [Revised: 06/14/2024] [Accepted: 07/11/2024] [Indexed: 07/16/2024]
Abstract
Neuronal cell dysfunction plays an important role in neurodegenerative diseases. Oxidative stress can disrupt the redox balance within neuronal cells and may cause neuronal nitric oxide synthase (nNOS) to uncouple, contributing to the neurodegenerative processes. Experimental studies and clinical trials using nNOS cofactor tetrahydrobiopterin (BH4) and antioxidants in neuronal cell dysfunction have shown inconsistent results. A better mechanistic understanding of complex interactions of nNOS activity and oxidative stress in neuronal cell dysfunction is needed. In this study, we developed a computational model of neuronal cell using nNOS biochemical pathways to explore several key mechanisms that are known to influence neuronal cell redox homeostasis. We studied the effects of oxidative stress and BH4 synthesis on nNOS nitric oxide production and biopterin ratio (BH4/total biopterin). Results showed that nNOS remained coupled and maintained nitric oxide production for oxidative stress levels less than 230 nM/s. The results showed that neuronal oxidative stress above 230 nM/s increased the degree of nNOS uncoupling and introduced instability in the nitric oxide production. The nitric oxide production did not change irrespective of initial biopterin ratio of 0.05-0.99 for a given oxidative stress. Oxidative stress resulted in significant reduction in BH4 levels even when nitric oxide production was not affected. Enhancing BH4 synthesis or supplementation improved nNOS coupling, however the degree of improvement was determined by the levels of oxidative stress and BH4 synthesis. The results of our mechanistic analysis indicate that there is a potential for significant improvement in neuronal dysfunction by simultaneously increasing BH4 levels and reducing cellular oxidative stress.
Collapse
Affiliation(s)
- Amnah Allboani
- Department of Biomedical Engineering, Wayne State University, Detroit, MI, 48202, USA
| | - Saptarshi Kar
- College of Engineering and Technology, American University of the Middle East, Kuwait
| | - Mahendra Kavdia
- Department of Biomedical Engineering, Wayne State University, Detroit, MI, 48202, USA.
| |
Collapse
|
2
|
De Simone G, Mazza B, Vellucci L, Barone A, Ciccarelli M, de Bartolomeis A. Schizophrenia Synaptic Pathology and Antipsychotic Treatment in the Framework of Oxidative and Mitochondrial Dysfunction: Translational Highlights for the Clinics and Treatment. Antioxidants (Basel) 2023; 12:antiox12040975. [PMID: 37107350 PMCID: PMC10135787 DOI: 10.3390/antiox12040975] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2023] [Revised: 04/05/2023] [Accepted: 04/13/2023] [Indexed: 04/29/2023] Open
Abstract
Schizophrenia is a worldwide mental illness characterized by alterations at dopaminergic and glutamatergic synapses resulting in global dysconnectivity within and between brain networks. Impairments in inflammatory processes, mitochondrial functions, energy expenditure, and oxidative stress have been extensively associated with schizophrenia pathophysiology. Antipsychotics, the mainstay of schizophrenia pharmacological treatment and all sharing the common feature of dopamine D2 receptor occupancy, may affect antioxidant pathways as well as mitochondrial protein levels and gene expression. Here, we systematically reviewed the available evidence on antioxidants' mechanisms in antipsychotic action and the impact of first- and second-generation compounds on mitochondrial functions and oxidative stress. We further focused on clinical trials addressing the efficacy and tolerability of antioxidants as an augmentation strategy of antipsychotic treatment. EMBASE, Scopus, and Medline/PubMed databases were interrogated. The selection process was conducted in respect of the Preferred Reporting Items for Systematic Reviews and Meta-Analyses (PRISMA) criteria. Several mitochondrial proteins involved in cell viability, energy metabolism, and regulation of oxidative systems were reported to be significantly modified by antipsychotic treatment with differences between first- and second-generation drugs. Finally, antioxidants may affect cognitive and psychotic symptoms in patients with schizophrenia, and although the evidence is only preliminary, the results indicate that further studies are warranted.
Collapse
Affiliation(s)
- Giuseppe De Simone
- Section of Psychiatry, Laboratory of Translational and Molecular Psychiatry and Unit of Treatment-Resistant Psychosis, Department of Neuroscience, Reproductive Sciences, and Dentistry, University Medical School of Naples "Federico II", Via Pansini 5, 80131 Naples, Italy
| | - Benedetta Mazza
- Section of Psychiatry, Laboratory of Translational and Molecular Psychiatry and Unit of Treatment-Resistant Psychosis, Department of Neuroscience, Reproductive Sciences, and Dentistry, University Medical School of Naples "Federico II", Via Pansini 5, 80131 Naples, Italy
| | - Licia Vellucci
- Section of Psychiatry, Laboratory of Translational and Molecular Psychiatry and Unit of Treatment-Resistant Psychosis, Department of Neuroscience, Reproductive Sciences, and Dentistry, University Medical School of Naples "Federico II", Via Pansini 5, 80131 Naples, Italy
| | - Annarita Barone
- Section of Psychiatry, Laboratory of Translational and Molecular Psychiatry and Unit of Treatment-Resistant Psychosis, Department of Neuroscience, Reproductive Sciences, and Dentistry, University Medical School of Naples "Federico II", Via Pansini 5, 80131 Naples, Italy
| | - Mariateresa Ciccarelli
- Section of Psychiatry, Laboratory of Translational and Molecular Psychiatry and Unit of Treatment-Resistant Psychosis, Department of Neuroscience, Reproductive Sciences, and Dentistry, University Medical School of Naples "Federico II", Via Pansini 5, 80131 Naples, Italy
| | - Andrea de Bartolomeis
- Section of Psychiatry, Laboratory of Translational and Molecular Psychiatry and Unit of Treatment-Resistant Psychosis, Department of Neuroscience, Reproductive Sciences, and Dentistry, University Medical School of Naples "Federico II", Via Pansini 5, 80131 Naples, Italy
- UNESCO Chair on Health Education and Sustainable Development, University of Naples "Federico II", 80131 Naples, Italy
| |
Collapse
|
3
|
Mueller NPF, Carloni P, Alfonso-Prieto M. Molecular determinants of acrylamide neurotoxicity through covalent docking. Front Pharmacol 2023; 14:1125871. [PMID: 36937867 PMCID: PMC10018202 DOI: 10.3389/fphar.2023.1125871] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2022] [Accepted: 02/17/2023] [Indexed: 03/06/2023] Open
Abstract
Acrylamide (ACR) is formed during food processing by Maillard reaction between sugars and proteins at high temperatures. It is also used in many industries, from water waste treatment to manufacture of paper, fabrics, dyes and cosmetics. Unfortunately, cumulative exposure to acrylamide, either from diet or at the workplace, may result in neurotoxicity. Such adverse effects arise from covalent adducts formed between acrylamide and cysteine residues of several neuronal proteins via a Michael addition reaction. The molecular determinants of acrylamide reactivity and its impact on protein function are not completely understood. Here we have compiled a list of acrylamide protein targets reported so far in the literature in connection with neurotoxicity and performed a systematic covalent docking study. Our results indicate that acrylamide binding to cysteine is favored in the presence of nearby positively charged amino acids, such as lysines and arginines. For proteins with more than one reactive Cys, docking scores were able to discriminate between the primary ACR modification site and secondary sites modified only at high ACR concentrations. Therefore, docking scores emerge as a potential filter to predict Cys reactivity against acrylamide. Inspection of the ACR-protein complex structures provides insights into the putative functional consequences of ACR modification, especially for non-enzyme proteins. Based on our study, covalent docking is a promising computational tool to predict other potential protein targets mediating acrylamide neurotoxicity.
Collapse
Affiliation(s)
- Nicolas Pierre Friedrich Mueller
- Institute for Advanced Simulations IAS-5, Institute of Neuroscience and Medicine INM-9, Computational Biomedicine, Forschungszentrum Jülich, Jülich, Germany
- Faculty of Mathematics and Natural Sciences, Heinrich Heine University Düsseldorf, Düsseldorf, Germany
| | - Paolo Carloni
- Institute for Advanced Simulations IAS-5, Institute of Neuroscience and Medicine INM-9, Computational Biomedicine, Forschungszentrum Jülich, Jülich, Germany
- Department of Physics, RWTH Aachen University, Aachen, Germany
| | - Mercedes Alfonso-Prieto
- Institute for Advanced Simulations IAS-5, Institute of Neuroscience and Medicine INM-9, Computational Biomedicine, Forschungszentrum Jülich, Jülich, Germany
- Cécile and Oskar Vogt Institute for Brain Research, Medical Faculty, University Hospital Düsseldorf, Heinrich Heine University Düsseldorf, Düsseldorf, Germany
| |
Collapse
|
4
|
Nepal B, Das S, Reith ME, Kortagere S. Overview of the structure and function of the dopamine transporter and its protein interactions. Front Physiol 2023; 14:1150355. [PMID: 36935752 PMCID: PMC10020207 DOI: 10.3389/fphys.2023.1150355] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2023] [Accepted: 02/21/2023] [Indexed: 03/06/2023] Open
Abstract
The dopamine transporter (DAT) plays an integral role in dopamine neurotransmission through the clearance of dopamine from the extracellular space. Dysregulation of DAT is central to the pathophysiology of numerous neuropsychiatric disorders and as such is an attractive therapeutic target. DAT belongs to the solute carrier family 6 (SLC6) class of Na+/Cl- dependent transporters that move various cargo into neurons against their concentration gradient. This review focuses on DAT (SCL6A3 protein) while extending the narrative to the closely related transporters for serotonin and norepinephrine where needed for comparison or functional relevance. Cloning and site-directed mutagenesis experiments provided early structural knowledge of DAT but our contemporary understanding was achieved through a combination of crystallization of the related bacterial transporter LeuT, homology modeling, and subsequently the crystallization of drosophila DAT. These seminal findings enabled a better understanding of the conformational states involved in the transport of substrate, subsequently aiding state-specific drug design. Post-translational modifications to DAT such as phosphorylation, palmitoylation, ubiquitination also influence the plasma membrane localization and kinetics. Substrates and drugs can interact with multiple sites within DAT including the primary S1 and S2 sites involved in dopamine binding and novel allosteric sites. Major research has centered around the question what determines the substrate and inhibitor selectivity of DAT in comparison to serotonin and norepinephrine transporters. DAT has been implicated in many neurological disorders and may play a role in the pathology of HIV and Parkinson's disease via direct physical interaction with HIV-1 Tat and α-synuclein proteins respectively.
Collapse
Affiliation(s)
- Binod Nepal
- Department of Microbiology and Immunology, Drexel University College of Medicine, Philadelphia, PA, United States
| | - Sanjay Das
- Department of Microbiology and Immunology, Drexel University College of Medicine, Philadelphia, PA, United States
| | - Maarten E. Reith
- Department of Psychiatry, New York University School of Medicine, New York City, NY, United States
| | - Sandhya Kortagere
- Department of Microbiology and Immunology, Drexel University College of Medicine, Philadelphia, PA, United States
- *Correspondence: Sandhya Kortagere,
| |
Collapse
|
5
|
Murray AJ, Rogers JC, Katshu MZUH, Liddle PF, Upthegrove R. Oxidative Stress and the Pathophysiology and Symptom Profile of Schizophrenia Spectrum Disorders. Front Psychiatry 2021; 12:703452. [PMID: 34366935 PMCID: PMC8339376 DOI: 10.3389/fpsyt.2021.703452] [Citation(s) in RCA: 59] [Impact Index Per Article: 14.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/30/2021] [Accepted: 06/28/2021] [Indexed: 12/12/2022] Open
Abstract
Schizophrenia is associated with increased levels of oxidative stress, as reflected by an increase in the concentrations of damaging reactive species and a reduction in anti-oxidant defences to combat them. Evidence has suggested that whilst not the likely primary cause of schizophrenia, increased oxidative stress may contribute to declining course and poor outcomes associated with schizophrenia. Here we discuss how oxidative stress may be implicated in the aetiology of schizophrenia and examine how current understanding relates associations with symptoms, potentially via lipid peroxidation induced neuronal damage. We argue that oxidative stress may be a good target for future pharmacotherapy in schizophrenia and suggest a multi-step model of illness progression with oxidative stress involved at each stage.
Collapse
Affiliation(s)
- Alex J. Murray
- Institute for Mental Health, University of Birmingham, Birmingham, United Kingdom
| | - Jack C. Rogers
- Institute for Mental Health, University of Birmingham, Birmingham, United Kingdom
| | - Mohammad Zia Ul Haq Katshu
- Institute of Mental Health, Division of Mental Health and Neurosciences University of Nottingham, Nottingham, United Kingdom
- Nottinghamshire Healthcare National Health Service Foundation Trust, Nottingham, United Kingdom
| | - Peter F. Liddle
- Institute of Mental Health, Division of Mental Health and Neurosciences University of Nottingham, Nottingham, United Kingdom
| | - Rachel Upthegrove
- Institute for Mental Health, University of Birmingham, Birmingham, United Kingdom
- Early Intervention Service, Birmingham Women's and Children's National Health Service Foundation Trust, Centre for Human Brain Health, University of Birmingham, Birmingham, United Kingdom
| |
Collapse
|
6
|
Wu M, Su H, Zhao M. The Role of α-Synuclein in Methamphetamine-Induced Neurotoxicity. Neurotox Res 2021; 39:1007-1021. [PMID: 33555547 DOI: 10.1007/s12640-021-00332-2] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2020] [Revised: 12/19/2020] [Accepted: 01/06/2021] [Indexed: 12/28/2022]
Abstract
Methamphetamine (METH), a highly addictive psychostimulant, is the second most widely used illicit drug. METH produces damage dopamine neurons and apoptosis via multiple inter-regulating mechanisms, including dopamine overload, hyperthermia, oxidative stress, mitochondria dysfunction, endoplasmic reticulum stress, protein degradation system dysfunction, and neuroinflammation. Increasing evidence suggests that chronic METH abuse is associated with neurodegenerative changes in the human brain and an increased risk of Parkinson's disease (PD). METH use and PD may share some common steps in causing neurotoxicity. Accumulation of α-synuclein, a presynaptic protein, is the pathological hallmark of PD. Intriguingly, α-synuclein upregulation and aggregation are also found in dopaminergic neurons in the substantia nigra in chronic METH users. This suggests α-synuclein may play a role in METH-induced neurotoxicity. The mechanism of α-synuclein cytotoxicity in PD has attracted considerable attention; however, how α-synuclein affects METH-induced neurotoxicity has not been reviewed. In this review, we summarize the relationship between METH use and PD, interdependent mechanisms that are involved in METH-induced neurotoxicity and the significance of α-synuclein upregulation in response to METH use. The identification of α-synuclein overexpression and aggregation as a contributor to METH-induced neurotoxicity may provide a novel therapeutic target for the treatment of the deleterious effect of this drug and drug addiction.
Collapse
Affiliation(s)
- Manqing Wu
- Shanghai Mental Health Center, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Hang Su
- Shanghai Mental Health Center, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Min Zhao
- Shanghai Mental Health Center, Shanghai Jiao Tong University School of Medicine, Shanghai, China.
- Shanghai Key Laboratory of Psychotic Disorders, Shanghai, China.
- Shanghai Clinical Research Center for Mental Health, Shanghai, China.
- CAS Center for Excellence in Brain Science and Intelligence Technology (CEBSIT), Chinese Academy of Sciences, Shanghai, China.
| |
Collapse
|
7
|
Triningsih D, Yang JH, Sim KH, Lee C, Lee YJ. Acrylamide and its metabolite induce neurotoxicity via modulation of protein kinase C and AMP-activated protein kinase pathways. Toxicol In Vitro 2021; 72:105105. [PMID: 33545342 DOI: 10.1016/j.tiv.2021.105105] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2020] [Revised: 01/26/2021] [Accepted: 01/27/2021] [Indexed: 11/25/2022]
Abstract
Acrylamide is known as a neurotoxicant found in commonly consumed food as well as in human body. However, the underlying mechanisms involved in neurotoxicity by acrylamide and its metabolite, glycidamide remain largely unknown. In this study, we have examined the interplay between CYP2E1, AMPK, ERK and PKC in acrylamide-induced neurotoxicity associated with autophagy in PC12 cells. Acrylamide-induced cell death was mediated by CYP2E1 expression and the activation of ERK, PKC-ɑ and PKC-δ, whereas AMPK knockdown exacerbated the acrylamide-induced neurotoxic effects. PKC-ɑ, but not PKC-δ, plays an upstream regulator of ERK and AMPK. Moreover, AMPK activation suppressed ERK, and CYP2E1 and AMPK bilaterally inhibit each other. Furthermore, acrylamide increased autophagy with impaired autophagic flux, evidenced by the increased beclin-1, LC3-II and p62 protein. Acrylamide-induced neuronal death was ameliorated by 3-methyladenine, an autophagy inhibitor, whereas neuronal death was exacerbated by chloroquine, a lysosomal inhibitor. Interestingly, PKC-δ siRNA, but not PKC-ɑ siRNA, dramatically reduced acrylamide-induced beclin-1 and LC3-II levels, whereas AMPK siRNA further increased beclin-1, LC3-II and p62 protein levels. Glycidamide, a major metabolite, mimicked acrylamide only with a higher potency. Taken together, acrylamide- and glycidamide-induced neurotoxicity may involve cytotoxic autophagy, which is mediated by interplay between PKCs and AMPK pathways.
Collapse
Affiliation(s)
- Dahlia Triningsih
- Department of Pharmacology, School of Medicine, Catholic University of Daegu, Daegu, Republic of Korea
| | - Jae-Ho Yang
- Department of Pharmacology, School of Medicine, Catholic University of Daegu, Daegu, Republic of Korea
| | - Kyeong Hwa Sim
- Department of Pharmacology, School of Medicine, Catholic University of Daegu, Daegu, Republic of Korea
| | - Chuhee Lee
- Department of Biochemistry and Molecular Biology, School of Medicine, Yeungnam University, Daegu, Republic of Korea
| | - Youn Ju Lee
- Department of Pharmacology, School of Medicine, Catholic University of Daegu, Daegu, Republic of Korea.
| |
Collapse
|
8
|
Kohut SJ, Kaufman MJ. Magnetic resonance spectroscopy studies of substance use disorders: Current landscape and potential future directions. Pharmacol Biochem Behav 2020; 200:173090. [PMID: 33333132 DOI: 10.1016/j.pbb.2020.173090] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/10/2020] [Revised: 11/13/2020] [Accepted: 12/08/2020] [Indexed: 12/25/2022]
Abstract
Over 200 in vivo magnetic resonance spectroscopy (MRS) studies of substance use and related disorders (SUD) were published this past decade. The large majority of this work used proton (1H)-MRS to characterize effects of acute and chronic exposures to drugs of abuse on human brain metabolites including N-acetylaspartate, choline-containing metabolites, creatine plus phosphocreatine, glutamate, and GABA. Some studies used phosphorus (31P)-MRS to quantify biomarkers of cerebral metabolism including phosphocreatine and adenosine triphosphate. A few studies used carbon (13C)-MRS to quantify intermediary metabolism. This Mini-review discusses select studies that illustrate how MRS can complement neurocircuitry research including by use of multimodal imaging strategies that combine MRS with functional MRI (fMRI) and/or diffusion tensor imaging (DTI). Additionally, magnetic resonance spectroscopic imaging (MRSI), which enables simultaneous multivoxel MRS acquisitions, can be used to better understand and interpret whole-brain functional or structural connectivity data. The review discusses some limitations in MRS methodology and then highlights important knowledge gaps and areas for potential future investigation, including the use of 1H- and 31P-MRS to quantify cerebral metabolism, oxidative stress, inflammation, and brain temperature, all of which are associated with SUD and all of which can influence neurocircuitry and behavior.
Collapse
Affiliation(s)
- Stephen J Kohut
- Behavioral Biology Research Program, McLean Hospital, Belmont, MA 02478, USA; McLean Imaging Center, McLean Hospital, Belmont, MA 02478, USA; Harvard Medical School, Department of Psychiatry, Belmont, MA 02478, USA
| | - Marc J Kaufman
- McLean Imaging Center, McLean Hospital, Belmont, MA 02478, USA; Harvard Medical School, Department of Psychiatry, Belmont, MA 02478, USA.
| |
Collapse
|
9
|
Abalenikhina YV, Kosmachevskaya OV, Topunov AF. Peroxynitrite: Toxic Agent and Signaling Molecule (Review). APPL BIOCHEM MICRO+ 2020. [DOI: 10.1134/s0003683820060022] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
|
10
|
Jeon M, Lin G, Stephen ZR, Vechey JE, Singh M, Revia R, Newman AH, Martinez D, Zhang M. Cocaine analogue conjugated magnetic nanoparticles for labeling and imaging dopaminergic neurons. Biomater Sci 2020; 8:4166-4175. [PMID: 32515443 DOI: 10.1039/d0bm00546k] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Molecular imaging of the dopamine transporter (DAT) with Positron Emission Tomography (PET) or Single Photon Emission Computed Tomography (SPECT) has been widely used in studies of neurological and psychiatric disorders. Nevertheless, there is a great interest in expanding molecular imaging to include magnetic resonance technology, because of the superior spatial resolution this technology may provide. Here we present a magnetic nanoparticle (NP) that specifically targets dopaminergic neurons and allows DAT imaging with magnetic resonance imaging (MRI). The nanoparticle (namely, NP-DN) is composed of an iron oxide core and a polyethylene glycol (PEG) coating to which a DAT specific dopaminergic neurolabeler (DN) is conjugated. NP-DN displayed long-term stability with favorable hydrodynamic size and surface charge suitable for in vivo application. In vitro studies showed NP-DN was non-toxic, displayed specificity towards DAT-expressing neurons, and demonstrated a 3-fold increase in DAT labeling over non-targeted NP. Our study shows NP-DN provides excellent contrast enhancement for MRI and demonstrates great potential for neuroimaging.
Collapse
Affiliation(s)
- Mike Jeon
- Department of Material Sciences and Engineering, University of Washington, Seattle, WA 98195, USA.
| | | | | | | | | | | | | | | | | |
Collapse
|
11
|
Functions and dysfunctions of nitric oxide in brain. Biochim Biophys Acta Mol Basis Dis 2019; 1865:1949-1967. [DOI: 10.1016/j.bbadis.2018.11.007] [Citation(s) in RCA: 97] [Impact Index Per Article: 16.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2018] [Revised: 10/29/2018] [Accepted: 11/11/2018] [Indexed: 02/06/2023]
|
12
|
Yang B, Xu J, Yuan ZH, Zheng DJ, He ZX, Jiao QC, Zhu HL. A new selective fluorescence probe with a quinoxalinone structure (QP-1) for cysteine and its application in live-cell imaging. Talanta 2018; 189:629-635. [DOI: 10.1016/j.talanta.2018.07.064] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2018] [Revised: 07/15/2018] [Accepted: 07/19/2018] [Indexed: 01/05/2023]
|
13
|
Weng M, Xie X, Liu C, Lim KL, Zhang CW, Li L. The Sources of Reactive Oxygen Species and Its Possible Role in the Pathogenesis of Parkinson's Disease. PARKINSON'S DISEASE 2018; 2018:9163040. [PMID: 30245802 PMCID: PMC6139203 DOI: 10.1155/2018/9163040] [Citation(s) in RCA: 57] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/22/2018] [Revised: 06/29/2018] [Accepted: 07/24/2018] [Indexed: 02/07/2023]
Abstract
Parkinson's disease (PD) is the second most common neurodegenerative disorder characterized by progressive loss of dopaminergic neurons in the substantia nigra. The precise mechanism underlying pathogenesis of PD is not fully understood, but it has been widely accepted that excessive reactive oxygen species (ROS) are the key mediator of PD pathogenesis. The causative factors of PD such as gene mutation, neuroinflammation, and iron accumulation all could induce ROS generation, and the later would mediate the dopaminergic neuron death by causing oxidation protein, lipids, and other macromolecules in the cells. Obviously, it is of mechanistic and therapeutic significance to understand where ROS are derived and how ROS induce dopaminergic neuron damage. In the present review, we try to summarize and discuss the main source of ROS in PD and the key pathways through which ROS mediate DA neuron death.
Collapse
Affiliation(s)
- Minrui Weng
- Key Laboratory of Flexible Electronics (KLOFE) & Institute of Advanced Materials (IAM), Jiangsu National Synergetic Innovation Center for Advanced Materials (SICAM), Nanjing Tech University (Nanjing Tech), 30 South Puzhu Road, Nanjing 211816, China
| | - Xiaoji Xie
- Key Laboratory of Flexible Electronics (KLOFE) & Institute of Advanced Materials (IAM), Jiangsu National Synergetic Innovation Center for Advanced Materials (SICAM), Nanjing Tech University (Nanjing Tech), 30 South Puzhu Road, Nanjing 211816, China
| | - Chao Liu
- Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117593
| | - Kah-Leong Lim
- Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117593
| | - Cheng-wu Zhang
- Key Laboratory of Flexible Electronics (KLOFE) & Institute of Advanced Materials (IAM), Jiangsu National Synergetic Innovation Center for Advanced Materials (SICAM), Nanjing Tech University (Nanjing Tech), 30 South Puzhu Road, Nanjing 211816, China
| | - Lin Li
- Key Laboratory of Flexible Electronics (KLOFE) & Institute of Advanced Materials (IAM), Jiangsu National Synergetic Innovation Center for Advanced Materials (SICAM), Nanjing Tech University (Nanjing Tech), 30 South Puzhu Road, Nanjing 211816, China
| |
Collapse
|
14
|
Morris G, Walder K, McGee SL, Dean OM, Tye SJ, Maes M, Berk M. A model of the mitochondrial basis of bipolar disorder. Neurosci Biobehav Rev 2017; 74:1-20. [PMID: 28093238 DOI: 10.1016/j.neubiorev.2017.01.014] [Citation(s) in RCA: 120] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2016] [Revised: 01/08/2017] [Accepted: 01/10/2017] [Indexed: 12/11/2022]
|
15
|
Current understanding of methamphetamine-associated dopaminergic neurodegeneration and psychotoxic behaviors. Arch Pharm Res 2017; 40:403-428. [DOI: 10.1007/s12272-017-0897-y] [Citation(s) in RCA: 52] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2016] [Accepted: 12/02/2016] [Indexed: 12/21/2022]
|
16
|
Bermingham DP, Blakely RD. Kinase-dependent Regulation of Monoamine Neurotransmitter Transporters. Pharmacol Rev 2016; 68:888-953. [PMID: 27591044 PMCID: PMC5050440 DOI: 10.1124/pr.115.012260] [Citation(s) in RCA: 73] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Modulation of neurotransmission by the monoamines dopamine (DA), norepinephrine (NE), and serotonin (5-HT) is critical for normal nervous system function. Precise temporal and spatial control of this signaling in mediated in large part by the actions of monoamine transporters (DAT, NET, and SERT, respectively). These transporters act to recapture their respective neurotransmitters after release, and disruption of clearance and reuptake has significant effects on physiology and behavior and has been linked to a number of neuropsychiatric disorders. To ensure adequate and dynamic control of these transporters, multiple modes of control have evolved to regulate their activity and trafficking. Central to many of these modes of control are the actions of protein kinases, whose actions can be direct or indirectly mediated by kinase-modulated protein interactions. Here, we summarize the current state of our understanding of how protein kinases regulate monoamine transporters through changes in activity, trafficking, phosphorylation state, and interacting partners. We highlight genetic, biochemical, and pharmacological evidence for kinase-linked control of DAT, NET, and SERT and, where applicable, provide evidence for endogenous activators of these pathways. We hope our discussion can lead to a more nuanced and integrated understanding of how neurotransmitter transporters are controlled and may contribute to disorders that feature perturbed monoamine signaling, with an ultimate goal of developing better therapeutic strategies.
Collapse
Affiliation(s)
- Daniel P Bermingham
- Department of Pharmacology (D.P.B., R.D.B.) and Psychiatry (R.D.B.), Vanderbilt University Medical Center, Nashville, Tennessee; and Department of Biomedical Sciences, Charles E. Schmidt College of Medicine and Brain Institute, Florida Atlantic University, Jupiter, Florida (R.D.B.)
| | - Randy D Blakely
- Department of Pharmacology (D.P.B., R.D.B.) and Psychiatry (R.D.B.), Vanderbilt University Medical Center, Nashville, Tennessee; and Department of Biomedical Sciences, Charles E. Schmidt College of Medicine and Brain Institute, Florida Atlantic University, Jupiter, Florida (R.D.B.)
| |
Collapse
|
17
|
Reddy IA, Pino JA, Weikop P, Osses N, Sørensen G, Bering T, Valle C, Bluett RJ, Erreger K, Wortwein G, Reyes JG, Graham D, Stanwood GD, Hackett TA, Patel S, Fink-Jensen A, Torres GE, Galli A. Glucagon-like peptide 1 receptor activation regulates cocaine actions and dopamine homeostasis in the lateral septum by decreasing arachidonic acid levels. Transl Psychiatry 2016; 6:e809. [PMID: 27187231 PMCID: PMC5070047 DOI: 10.1038/tp.2016.86] [Citation(s) in RCA: 54] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/18/2015] [Revised: 02/19/2016] [Accepted: 03/30/2016] [Indexed: 12/12/2022] Open
Abstract
Agonism of the glucagon-like peptide 1 (GLP-1) receptor (GLP-1R) has been effective at treating aspects of addictive behavior for a number of abused substances, including cocaine. However, the molecular mechanisms and brain circuits underlying the therapeutic effects of GLP-1R signaling on cocaine actions remain elusive. Recent evidence has revealed that endogenous signaling at the GLP-1R within the forebrain lateral septum (LS) acts to reduce cocaine-induced locomotion and cocaine conditioned place preference, both considered dopamine (DA)-associated behaviors. DA terminals project from the ventral tegmental area to the LS and express the DA transporter (DAT). Cocaine acts by altering DA bioavailability by targeting the DAT. Therefore, GLP-1R signaling might exert effects on DAT to account for its regulation of cocaine-induced behaviors. We show that the GLP-1R is highly expressed within the LS. GLP-1, in LS slices, significantly enhances DAT surface expression and DAT function. Exenatide (Ex-4), a long-lasting synthetic analog of GLP-1 abolished cocaine-induced elevation of DA. Interestingly, acute administration of Ex-4 reduces septal expression of the retrograde messenger 2-arachidonylglycerol (2-AG), as well as a product of its presynaptic degradation, arachidonic acid (AA). Notably, AA reduces septal DAT function pointing to AA as a novel regulator of central DA homeostasis. We further show that AA oxidation product γ-ketoaldehyde (γ-KA) forms adducts with the DAT and reduces DAT plasma membrane expression and function. These results support a mechanism in which postsynaptic septal GLP-1R activation regulates 2-AG levels to alter presynaptic DA homeostasis and cocaine actions through AA.
Collapse
Affiliation(s)
- I A Reddy
- Neuroscience Program, Vanderbilt University School of Medicine, Nashville, TN, USA
| | - J A Pino
- Instituto de Química, Facultad de Ciencias, Pontificia Universidad Católica de Valparaíso, Valparaíso, Chile
- Department of Pharmacology and Therapeutics, College of Medicine, University of Florida, Gainesville, FL, USA
| | - P Weikop
- Laboratory of Neuropsychiatry, Department of Neuroscience and Pharmacology, University of Copenhagen, Copenhagen, Denmark
- Psychiatric Centre Copenhagen, University Hospital Copenhagen, Copenhagen, Denmark
| | - N Osses
- Instituto de Química, Facultad de Ciencias, Pontificia Universidad Católica de Valparaíso, Valparaíso, Chile
| | - G Sørensen
- Laboratory of Neuropsychiatry, Department of Neuroscience and Pharmacology, University of Copenhagen, Copenhagen, Denmark
- Psychiatric Centre Copenhagen, University Hospital Copenhagen, Copenhagen, Denmark
| | - T Bering
- Laboratory of Neuropsychiatry, Department of Neuroscience and Pharmacology, University of Copenhagen, Copenhagen, Denmark
| | - C Valle
- Departamento de Ciencias Básicas, Universidad de Viña del Mar, Viña del Mar, Chile
| | - R J Bluett
- Neuroscience Program, Vanderbilt University School of Medicine, Nashville, TN, USA
- Department of Psychiatry, Vanderbilt University Medical Center, Nashville, TN, USA
| | - K Erreger
- Department of Molecular Physiology and Biophysics, Vanderbilt University Medical Center, Nashville, TN, USA
| | - G Wortwein
- Laboratory of Neuropsychiatry, Department of Neuroscience and Pharmacology, University of Copenhagen, Copenhagen, Denmark
- Department of Public Health, University of Copenhagen, Copenhagen, Denmark
| | - J G Reyes
- Instituto de Química, Facultad de Ciencias, Pontificia Universidad Católica de Valparaíso, Valparaíso, Chile
| | - D Graham
- Department of Biomedical Sciences and Center for Brain Repair, Florida State University, Tallahassee, FL, USA
| | - G D Stanwood
- Department of Biomedical Sciences and Center for Brain Repair, Florida State University, Tallahassee, FL, USA
| | - T A Hackett
- Neuroscience Program, Vanderbilt University School of Medicine, Nashville, TN, USA
- Department of Hearing and Speech Sciences, Vanderbilt University Medical Center, Nashville, TN, USA
| | - S Patel
- Department of Psychiatry, Vanderbilt University Medical Center, Nashville, TN, USA
| | - A Fink-Jensen
- Laboratory of Neuropsychiatry, Department of Neuroscience and Pharmacology, University of Copenhagen, Copenhagen, Denmark
- Psychiatric Centre Copenhagen, University Hospital Copenhagen, Copenhagen, Denmark
| | - G E Torres
- Department of Pharmacology and Therapeutics, College of Medicine, University of Florida, Gainesville, FL, USA
| | - A Galli
- Neuroscience Program, Vanderbilt University School of Medicine, Nashville, TN, USA
- Department of Molecular Physiology and Biophysics, Vanderbilt University Medical Center, Nashville, TN, USA
| |
Collapse
|
18
|
Comparative Proteomic Analysis of Carbonylated Proteins from the Striatum and Cortex of Pesticide-Treated Mice. PARKINSONS DISEASE 2015; 2015:812532. [PMID: 26345149 PMCID: PMC4546751 DOI: 10.1155/2015/812532] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/29/2015] [Revised: 05/27/2015] [Accepted: 05/30/2015] [Indexed: 01/28/2023]
Abstract
Epidemiological studies indicate exposures to the herbicide paraquat (PQ) and fungicide maneb (MB) are associated with increased risk of Parkinson's disease (PD). Oxidative stress appears to be a premier mechanism that underlies damage to the nigrostriatal dopamine system in PD and pesticide exposure. Enhanced oxidative stress leads to lipid peroxidation and production of reactive aldehydes; therefore, we conducted proteomic analyses to identify carbonylated proteins in the striatum and cortex of pesticide-treated mice in order to elucidate possible mechanisms of toxicity. Male C57BL/6J mice were treated biweekly for 6 weeks with saline, PQ (10 mg/kg), MB (30 mg/kg), or the combination of PQ and MB (PQMB). Treatments resulted in significant behavioral alterations in all treated mice and depleted striatal dopamine in PQMB mice. Distinct differences in 4-hydroxynonenal-modified proteins were observed in the striatum and cortex. Proteomic analyses identified carbonylated proteins and peptides from the cortex and striatum, and pathway analyses revealed significant enrichment in a variety of KEGG pathways. Further analysis showed enrichment in proteins of the actin cytoskeleton in treated samples, but not in saline controls. These data indicate that treatment-related effects on cytoskeletal proteins could alter proper synaptic function, thereby resulting in impaired neuronal function and even neurodegeneration.
Collapse
|
19
|
Lohr KM, Stout KA, Dunn AR, Wang M, Salahpour A, Guillot TS, Miller GW. Increased Vesicular Monoamine Transporter 2 (VMAT2; Slc18a2) Protects against Methamphetamine Toxicity. ACS Chem Neurosci 2015; 6:790-9. [PMID: 25746685 DOI: 10.1021/acschemneuro.5b00010] [Citation(s) in RCA: 57] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022] Open
Abstract
The psychostimulant methamphetamine (METH) is highly addictive and neurotoxic to dopamine terminals. METH toxicity has been suggested to be due to the release and accumulation of dopamine in the cytosol of these terminals. The vesicular monoamine transporter 2 (VMAT2; SLC18A2) is a critical mediator of dopamine handling. Mice overexpressing VMAT2 (VMAT2-HI) have an increased vesicular capacity to store dopamine, thus augmenting striatal dopamine levels and dopamine release in the striatum. Based on the altered compartmentalization of intracellular dopamine in the VMAT2-HI mice, we assessed whether enhanced vesicular function was capable of reducing METH-induced damage to the striatal dopamine system. While wildtype mice show significant losses in striatal levels of the dopamine transporter (65% loss) and tyrosine hydroxylase (46% loss) following a 4 × 10 mg/kg METH dosing regimen, VMAT2-HI mice were protected from this damage. VMAT2-HI mice were also spared from the inflammatory response that follows METH treatment, showing an increase in astroglial markers that was approximately one-third of that of wildtype animals (117% vs 36% increase in GFAP, wildtype vs VMAT2-HI). Further analysis also showed that elevated VMAT2 level does not alter the ability of METH to increase core body temperature, a mechanism integral to the toxicity of the drug. Finally, the VMAT2-HI mice showed no difference from wildtype littermates on both METH-induced conditioned place preference and in METH-induced locomotor activity (1 mg/kg METH). These results demonstrate that elevated VMAT2 protects against METH toxicity without enhancing the rewarding effects of the drug. Since the VMAT2-HI mice are protected from METH despite higher basal dopamine levels, this study suggests that METH toxicity depends more on the proper compartmentalization of synaptic dopamine than on the absolute amount of dopamine in the brain.
Collapse
Affiliation(s)
| | | | | | | | - Ali Salahpour
- Department
of Pharmacology and Toxicology, University of Toronto, Toronto, Ontario M5S 1A8, Canada
| | | | | |
Collapse
|
20
|
Extracellular dopamine and alterations on dopamine transporter are related to reserpine toxicity in Caenorhabditis elegans. Arch Toxicol 2015; 90:633-45. [PMID: 25579234 DOI: 10.1007/s00204-015-1451-7] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2014] [Accepted: 01/06/2015] [Indexed: 10/24/2022]
Abstract
Reserpine is used as an animal model of parkinsonism. We hypothesized that the involuntary movements induced by reserpine in rodents are induced by dopaminergic toxicity caused by extracellular dopamine accumulation. The present study tested the effects of reserpine on the dopaminergic system in Caenorhabditis elegans. Reserpine was toxic to worms (decreased the survival, food intake, development and changed egg laying and defecation cycles). In addition, reserpine increased the worms' locomotor rate on food and decreased dopamine levels. Morphological evaluations of dopaminergic CEP neurons confirmed neurodegeneration characterized by decreased fluorescence intensity and the number of worms with intact CEP neurons, and increased number of shrunken somas per worm. These effects were unrelated to reserpine's effect on decreased expression of the dopamine transporter, dat-1. Interestingly, the locomotor rate on food and the neurodegenerative parameters fully recovered to basal conditions upon reserpine withdrawal. Furthermore, reserpine decreased survival in vesicular monoamine transporter and dat-1 loss-of-function mutant worms. In addition, worms pre-exposed to dopamine followed by exposure to reserpine had decreased survival. Reserpine activated gst-4, which controls a phase II detoxification enzymes downstream of nuclear factor (erythroid-derived-2)-like 2. Our findings establish that the dopamine transporter, dat-1, plays an important role in reserpine toxicity, likely by increasing extracellular dopamine concentrations.
Collapse
|
21
|
Navarro-Yepes J, Zavala-Flores L, Anandhan A, Wang F, Skotak M, Chandra N, Li M, Pappa A, Martinez-Fong D, Del Razo LM, Quintanilla-Vega B, Franco R. Antioxidant gene therapy against neuronal cell death. Pharmacol Ther 2014; 142:206-30. [PMID: 24333264 PMCID: PMC3959583 DOI: 10.1016/j.pharmthera.2013.12.007] [Citation(s) in RCA: 97] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2013] [Accepted: 11/26/2013] [Indexed: 12/21/2022]
Abstract
Oxidative stress is a common hallmark of neuronal cell death associated with neurodegenerative disorders such as Alzheimer's disease, Parkinson's disease, as well as brain stroke/ischemia and traumatic brain injury. Increased accumulation of reactive species of both oxygen (ROS) and nitrogen (RNS) has been implicated in mitochondrial dysfunction, energy impairment, alterations in metal homeostasis and accumulation of aggregated proteins observed in neurodegenerative disorders, which lead to the activation/modulation of cell death mechanisms that include apoptotic, necrotic and autophagic pathways. Thus, the design of novel antioxidant strategies to selectively target oxidative stress and redox imbalance might represent important therapeutic approaches against neurological disorders. This work reviews the evidence demonstrating the ability of genetically encoded antioxidant systems to selectively counteract neuronal cell loss in neurodegenerative diseases and ischemic brain damage. Because gene therapy approaches to treat inherited and acquired disorders offer many unique advantages over conventional therapeutic approaches, we discussed basic research/clinical evidence and the potential of virus-mediated gene delivery techniques for antioxidant gene therapy.
Collapse
Affiliation(s)
- Juliana Navarro-Yepes
- Redox Biology Center, University of Nebraska-Lincoln, Lincoln, NE 68583, United States; School of Veterinary Medicine and Biomedical Sciences, University of Nebraska-Lincoln, Lincoln, NE 68583, United States; Department of Toxicology, CINVESTAV-IPN, Mexico City, Mexico
| | - Laura Zavala-Flores
- Redox Biology Center, University of Nebraska-Lincoln, Lincoln, NE 68583, United States; School of Veterinary Medicine and Biomedical Sciences, University of Nebraska-Lincoln, Lincoln, NE 68583, United States
| | - Annadurai Anandhan
- Redox Biology Center, University of Nebraska-Lincoln, Lincoln, NE 68583, United States; School of Veterinary Medicine and Biomedical Sciences, University of Nebraska-Lincoln, Lincoln, NE 68583, United States
| | - Fang Wang
- Department of Mechanical and Materials Engineering, University of Nebraska-Lincoln, Lincoln, NE 68583, United States
| | - Maciej Skotak
- Department of Mechanical and Materials Engineering, University of Nebraska-Lincoln, Lincoln, NE 68583, United States
| | - Namas Chandra
- Department of Mechanical and Materials Engineering, University of Nebraska-Lincoln, Lincoln, NE 68583, United States
| | - Ming Li
- Department of Psychology, University of Nebraska-Lincoln, Lincoln, NE 68583, United States
| | - Aglaia Pappa
- Department of Molecular Biology and Genetics, Democritus University of Thrace, University Campus, Dragana, Alexandroupolis, Greece
| | - Daniel Martinez-Fong
- Department of Physiology, Biophysics and Neurosciences, CINVESTAV-IPN, Mexico City, Mexico
| | | | | | - Rodrigo Franco
- Redox Biology Center, University of Nebraska-Lincoln, Lincoln, NE 68583, United States; School of Veterinary Medicine and Biomedical Sciences, University of Nebraska-Lincoln, Lincoln, NE 68583, United States.
| |
Collapse
|
22
|
Stetler RA, Leak RK, Gan Y, Li P, Zhang F, Hu X, Jing Z, Chen J, Zigmond MJ, Gao Y. Preconditioning provides neuroprotection in models of CNS disease: paradigms and clinical significance. Prog Neurobiol 2014; 114:58-83. [PMID: 24389580 PMCID: PMC3937258 DOI: 10.1016/j.pneurobio.2013.11.005] [Citation(s) in RCA: 151] [Impact Index Per Article: 13.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2012] [Revised: 11/18/2013] [Accepted: 11/18/2013] [Indexed: 12/14/2022]
Abstract
Preconditioning is a phenomenon in which brief episodes of a sublethal insult induce robust protection against subsequent lethal injuries. Preconditioning has been observed in multiple organisms and can occur in the brain as well as other tissues. Extensive animal studies suggest that the brain can be preconditioned to resist acute injuries, such as ischemic stroke, neonatal hypoxia/ischemia, surgical brain injury, trauma, and agents that are used in models of neurodegenerative diseases, such as Parkinson's disease and Alzheimer's disease. Effective preconditioning stimuli are numerous and diverse, ranging from transient ischemia, hypoxia, hyperbaric oxygen, hypothermia and hyperthermia, to exposure to neurotoxins and pharmacological agents. The phenomenon of "cross-tolerance," in which a sublethal stress protects against a different type of injury, suggests that different preconditioning stimuli may confer protection against a wide range of injuries. Research conducted over the past few decades indicates that brain preconditioning is complex, involving multiple effectors such as metabolic inhibition, activation of extra- and intracellular defense mechanisms, a shift in the neuronal excitatory/inhibitory balance, and reduction in inflammatory sequelae. An improved understanding of brain preconditioning should help us identify innovative therapeutic strategies that prevent or at least reduce neuronal damage in susceptible patients. In this review, we focus on the experimental evidence of preconditioning in the brain and systematically survey the models used to develop paradigms for neuroprotection, and then discuss the clinical potential of brain preconditioning.
Collapse
Affiliation(s)
- R Anne Stetler
- State Key Laboratory of Medical Neurobiology and Institute of Brain Sciences, Fudan University, Shanghai Medical College, Shanghai 200032, China; Department of Neurology and Center of Cerebrovascular Disease Research, University of Pittsburgh, School of Medicine, Pittsburgh, PA 15213, USA; Geriatric Research, Educational and Clinical Center, Veterans Affairs Pittsburgh Health Care System, Pittsburgh, PA 15261, USA
| | - Rehana K Leak
- Division of Pharmaceutical Sciences, Mylan School of Pharmacy, Duquesne University, Pittsburgh, PA 15282, USA
| | - Yu Gan
- State Key Laboratory of Medical Neurobiology and Institute of Brain Sciences, Fudan University, Shanghai Medical College, Shanghai 200032, China; Department of Neurology and Center of Cerebrovascular Disease Research, University of Pittsburgh, School of Medicine, Pittsburgh, PA 15213, USA
| | - Peiying Li
- State Key Laboratory of Medical Neurobiology and Institute of Brain Sciences, Fudan University, Shanghai Medical College, Shanghai 200032, China; Department of Neurology and Center of Cerebrovascular Disease Research, University of Pittsburgh, School of Medicine, Pittsburgh, PA 15213, USA
| | - Feng Zhang
- State Key Laboratory of Medical Neurobiology and Institute of Brain Sciences, Fudan University, Shanghai Medical College, Shanghai 200032, China; Department of Neurology and Center of Cerebrovascular Disease Research, University of Pittsburgh, School of Medicine, Pittsburgh, PA 15213, USA; Geriatric Research, Educational and Clinical Center, Veterans Affairs Pittsburgh Health Care System, Pittsburgh, PA 15261, USA
| | - Xiaoming Hu
- Department of Neurology and Center of Cerebrovascular Disease Research, University of Pittsburgh, School of Medicine, Pittsburgh, PA 15213, USA; Geriatric Research, Educational and Clinical Center, Veterans Affairs Pittsburgh Health Care System, Pittsburgh, PA 15261, USA
| | - Zheng Jing
- Department of Neurology and Center of Cerebrovascular Disease Research, University of Pittsburgh, School of Medicine, Pittsburgh, PA 15213, USA; Geriatric Research, Educational and Clinical Center, Veterans Affairs Pittsburgh Health Care System, Pittsburgh, PA 15261, USA
| | - Jun Chen
- State Key Laboratory of Medical Neurobiology and Institute of Brain Sciences, Fudan University, Shanghai Medical College, Shanghai 200032, China; Department of Neurology and Center of Cerebrovascular Disease Research, University of Pittsburgh, School of Medicine, Pittsburgh, PA 15213, USA; Geriatric Research, Educational and Clinical Center, Veterans Affairs Pittsburgh Health Care System, Pittsburgh, PA 15261, USA
| | - Michael J Zigmond
- State Key Laboratory of Medical Neurobiology and Institute of Brain Sciences, Fudan University, Shanghai Medical College, Shanghai 200032, China; Department of Neurology and Center of Cerebrovascular Disease Research, University of Pittsburgh, School of Medicine, Pittsburgh, PA 15213, USA
| | - Yanqin Gao
- State Key Laboratory of Medical Neurobiology and Institute of Brain Sciences, Fudan University, Shanghai Medical College, Shanghai 200032, China.
| |
Collapse
|
23
|
Snider GW, Ruggles E, Khan N, Hondal RJ. Selenocysteine confers resistance to inactivation by oxidation in thioredoxin reductase: comparison of selenium and sulfur enzymes. Biochemistry 2013; 52:5472-81. [PMID: 23865454 DOI: 10.1021/bi400462j] [Citation(s) in RCA: 78] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Mammalian thioredoxin reductase (TR) is a selenocysteine (Sec)-containing homodimeric pyridine nucleotide oxidoreductase which catalyzes the reduction of oxidized thioredoxin. We have previously demonstrated the full-length mitochondrial mammalian TR (mTR3) enzyme to be resistant to inactivation from exposure to 50 mM H2O2. Because a Sec residue oxidizes more rapidly than a cysteine (Cys) residue, it has been previously thought that Sec-containing enzymes are "sensitive to oxidation" compared to Cys-orthologues. Here we show for the first time a direct comparison of the abilities of Sec-containing mTR3 and the Cys-orthologue from D. melanogaster (DmTR) to resist inactivation by oxidation from a variety of oxidants including H2O2, hydroxyl radical, peroxynitrite, hypochlorous acid, hypobromous acid, and hypothiocyanous acid. The results show that the Sec-containing TR is far superior to the Cys-orthologue TR in resisting inactivation by oxidation. To further test our hypothesis that the use of Sec confers strong resistance to inactivation by oxidation, we constructed a chimeric enzyme in which we replaced the active site Cys nucleophile of DmTR with a Sec residue using semisynthesis. The chimeric Sec-containing enzyme has similar ability to resist inactivation by oxidation as the wild type Sec-containing TR from mouse mitochondria. The use of Sec in the chimeric enzyme "rescued" the enzyme from oxidant-induced inactivation for all of the oxidants tested in this study, in direct contrast to previous understanding. We discuss two possibilities for this rescue effect from inactivation under identical conditions of oxidative stress: (i) Sec resists overoxidation and inactivation, whereas a Cys residue can be permanently overoxidized to the sulfinic acid form, and (ii) Sec protects the body of the enzyme from harmful oxidation by allowing the enzyme to metabolize (turnover) various oxidants much better than a Cys-containing TR.
Collapse
Affiliation(s)
- Gregg W Snider
- Department of Biochemistry, University of Vermont, 89 Beaumont Ave, Given Building Room B413, Burlington, VT 05405, USA
| | | | | | | |
Collapse
|
24
|
Kim HK, Andreazza AC. The relationship between oxidative stress and post-translational modification of the dopamine transporter in bipolar disorder. Expert Rev Neurother 2012; 12:849-59. [PMID: 22853792 DOI: 10.1586/ern.12.64] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Bipolar disorder (BD) has been consistently associated with altered levels of oxidative stress markers, although the cause and consequences of these alterations remain to be elucidated. One of the main hypotheses regarding the pathogenesis of mania involves increased dopaminergic transmission. In this review, the authors aim to discuss a potential mechanism by which increased oxidative stress inhibits the uptake of dopamine through the post-translational modification of the dopamine transporter and its implications for BD. Within the next 5 years, the authors believe that the mechanisms of dopamine transporter oxidation and its impact on the pathophysiology of BD will be elucidated, which may open avenues for the development of more specific interventions for the treatment of this debilitating illness.
Collapse
|
25
|
N-Acetyl Cysteine Protects against Methamphetamine-Induced Dopaminergic Neurodegeneration via Modulation of Redox Status and Autophagy in Dopaminergic Cells. PARKINSONS DISEASE 2012; 2012:424285. [PMID: 23056996 PMCID: PMC3465903 DOI: 10.1155/2012/424285] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/19/2012] [Accepted: 08/27/2012] [Indexed: 01/12/2023]
Abstract
Methamphetamine- (MA-) induced neurotoxicity is associated with mitochondrial dysfunction and enhanced oxidative stress. Our previous study demonstrated that MA induces autophagy in a dopaminergic neuronal cell model (N27 cells). The cellular mechanisms underlying MA-induced autophagy and apoptosis remain poorly characterized. In the present study we sought to investigate the importance of GSH redox status in MA-induced neurotoxicity using a thiol antioxidant, N-acetylcysteine (NAC). Morphological and biochemical analysis revealed that MA-induced autophagy in N27 dopaminergic cells was associated with pronounced depletion of GSH levels. Moreover, pretreatment with NAC reduced MA-induced GSH depletion and autophagy, while depletion of GSH using L-buthionine sulfoximine (L-BSO) enhanced autophagy. Furthermore, treatment with NAC significantly attenuated MA-induced apoptotic cell death as well as oxidative stress markers, namely, 3-nitrotyrosine (3-NT) and 4-hydroxynonenal (4-HNE). Together, these results suggest that NAC exhibits significant protective effects against MA-induced dopaminergic cell death, presumably via modulation of the GSH level and autophagy. Collectively, our data provide mechanistic insights into the role of cellular GSH redox status in MA-induced autophagy and apoptotic cell death, and additional studies are needed to determine the therapeutic effectiveness of cellular redox modifiers in attenuating dopaminergic neurodegeneration in vivo.
Collapse
|
26
|
Maeng HJ, Bang YJ, Chung SJ. Functional impairment of P-glycoprotein by sodium nitroprusside pretreatment in mouse brain capillary endothelial cells. Arch Pharm Res 2012; 35:1215-21. [PMID: 22864744 DOI: 10.1007/s12272-012-0712-8] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2012] [Revised: 02/27/2012] [Accepted: 03/13/2012] [Indexed: 11/29/2022]
Abstract
We examined whether pretreatment of mouse brain blood vessel endothelial cell clone 4 (MBEC4) cells with sodium nitroprusside (SNP), a NO(x) donor, as an in vitro model of the bloodbrain barrier could affect P-glycoprotein (P-gp) functional activity. Uptake into the cells and MBEC4 plasma membrane vesicles (MPMVs) in the presence or absence of SNP pretreatment was used to investigate functional changes. Increased accumulation of [(3)H]vincristine, a widely used substrate for P-gp, into MBEC4 was observed upon SNP pretreatment, likely due to impaired P-gp function. To better understand the mechanism of the impairment, MPMVs were prepared and characterized in terms of purity and Na(+)-dependent glucose uptake. [(3)H]daunomycin uptake into MPMVs was diminished after SNP pretreatment in the presence of an ATP-regenerating system, indicating that the functional activity of P-gp was impaired after exposure to SNP. Under conditions of excess ATP, daunomycin uptake into the vesicles was still decreased after SNP pretreatment, indicating that SNP interacted directly with the transport system, but not with the ATP-regenerating system. Together, these results suggest that NO or NO(x) functionally impairs P-gp in the in vitro blood-brain barrier model with SNP pretreatment.
Collapse
Affiliation(s)
- Han-Joo Maeng
- College of Pharmacy, Inje University, Gimhae 621-749, Korea
| | | | | |
Collapse
|
27
|
Jang EY, Park KA, Lee JR, Yang CH, Hwang M. Protective Effect of Sauchinone on Methamphetamine-Induced Neurotoxicity in Mice. J Pharmacol Sci 2012; 118:531-6. [DOI: 10.1254/jphs.11207sc] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/28/2022] Open
|
28
|
Meulendyke KA, Pletnikov MV, Engle EL, Tarwater PM, Graham DR, Zink MC. Early minocycline treatment prevents a decrease in striatal dopamine in an SIV model of HIV-associated neurological disease. J Neuroimmune Pharmacol 2011; 7:454-64. [PMID: 22198699 DOI: 10.1007/s11481-011-9332-1] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2011] [Accepted: 12/01/2011] [Indexed: 12/13/2022]
Abstract
HIV-infected individuals, even with antiretroviral therapy, often display cognitive, behavioral and motor abnormalities and have decreased dopamine (DA) levels. Minocycline prevents encephalitis and neurodegeneration in SIV models, suggesting that it might also protect against nigrostriatal dopaminergic system dysfunction. Using an SIV/macaque model of HIV-associated CNS disease, we demonstrated that striatal levels of DA were significantly lower in macaques late in infection and that levels of the metabolite DOPAC also tended to be lower. DA levels declined more than its metabolites, indicating a dysregulation of DA production or catabolism. Minocycline treatment beginning at 12 but not 21 days postinoculation prevented striatal DA loss. DA decline was not due to direct loss of dopaminergic projections to the basal ganglia as there was no difference in tyrosine hydroxylase, dopamine transporter, vesicular monoamine transporter 2 or synaptophysin between minocycline-treated and untreated macaques. SIV-infected macaques had significantly higher monoamine oxidase (MAO) activity than uninfected macaques, although MAO activity was not affected by minocycline. Oxidative/nitrosative stress was examined by nitrotyrosine staining in the deep white matter and was lower in SIV-infected, minocycline-treated macaques compared with untreated macaques. These data suggest that minocycline, which has antioxidant activity, has a protective effect on DA homeostasis when administered at an appropriate time in SIV neuropathogenesis.
Collapse
Affiliation(s)
- Kelly A Meulendyke
- Department of Molecular and Comparative Pathobiology, Johns Hopkins University School of Medicine, 733 North Broadway Street, Baltimore, MD 21205, USA
| | | | | | | | | | | |
Collapse
|
29
|
Peroxynitrite and Nitroxidative Stress: Detection Probes and Micro-Sensors. A Case of a Nanostructured Catalytic Film. ACTA ACUST UNITED AC 2011. [DOI: 10.1021/bk-2011-1083.ch011] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/07/2023]
|
30
|
El Ayadi A, Zigmond MJ. Low concentrations of methamphetamine can protect dopaminergic cells against a larger oxidative stress injury: mechanistic study. PLoS One 2011; 6:e24722. [PMID: 22022363 PMCID: PMC3192034 DOI: 10.1371/journal.pone.0024722] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2010] [Accepted: 08/19/2011] [Indexed: 11/19/2022] Open
Abstract
Mild stress can protect against a larger insult, a phenomenon termed preconditioning or tolerance. To determine if a low intensity stressor could also protect cells against intense oxidative stress in a model of dopamine deficiency associated with Parkinson disease, we used methamphetamine to provide a mild, preconditioning stress, 6-hydroxydopamine (6-OHDA) as a source of potentially toxic oxidative stress, and MN9D cells as a model of dopamine neurons. We observed that prior exposure to subtoxic concentrations of methamphetamine protected these cells against 6-OHDA toxicity, whereas higher concentrations of methamphetamine exacerbated it. The protection by methamphetamine was accompanied by decreased uptake of both [(3)H] dopamine and 6-OHDA into the cells, which may have accounted for some of the apparent protection. However, a number of other effects of methamphetamine exposure suggest that the drug also affected basic cellular survival mechanisms. First, although methamphetamine preconditioning decreased basal pERK1/2 and pAkt levels, it enhanced the 6-OHDA-induced increase in these phosphokinases. Second, the apparent increase in pERK1/2 activity was accompanied by increased pMEK1/2 levels and decreased activity of protein phosphatase 2. Third, methamphetamine upregulated the pro-survival protein Bcl-2. Our results suggest that exposure to low concentrations of methamphetamine cause a number of changes in dopamine cells, some of which result in a decrease in their vulnerability to subsequent oxidative stress. These observations may provide insights into the development of new therapies for prevention or treatment of PD.
Collapse
Affiliation(s)
- Amina El Ayadi
- Pittsburgh Institute for Neurodegenerative Diseases, University of Pittsburgh, Pittsburgh, Pennsylvania, United States of America
- Department of Neuroscience and Cell Biology, University of Texas Medical Branch, Galveston, Texas, United States of America
| | - Michael J. Zigmond
- Pittsburgh Institute for Neurodegenerative Diseases, University of Pittsburgh, Pittsburgh, Pennsylvania, United States of America
| |
Collapse
|
31
|
Pubill D, Garcia-Ratés S, Camarasa J, Escubedo E. Neuronal Nicotinic Receptors as New Targets for Amphetamine-Induced Oxidative Damage and Neurotoxicity. Pharmaceuticals (Basel) 2011. [PMCID: PMC4055958 DOI: 10.3390/ph4060822] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
Amphetamine derivatives such as methamphetamine (METH) and 3,4-methylenedioxymethamphetamine (MDMA, “ecstasy”) are widely abused drugs in a recreational context. This has led to concern because of the evidence that they are neurotoxic in animal models and cognitive impairments have been described in heavy abusers. The main targets of these drugs are plasmalemmal and vesicular monoamine transporters, leading to reverse transport and increased monoamine efflux to the synapse. As far as neurotoxicity is concerned, increased reactive oxygen species (ROS) production seems to be one of the main causes. Recent research has demonstrated that blockade of α7 nicotinic acetylcholine receptors (nAChR) inhibits METH- and MDMA-induced ROS production in striatal synaptosomes which is dependent on calcium and on NO-synthase activation. Moreover, α7 nAChR antagonists (methyllycaconitine and memantine) attenuated in vivo the neurotoxicity induced by METH and MDMA, and memantine prevented the cognitive impairment induced by these drugs. Radioligand binding experiments demonstrated that both drugs have affinity to α7 and heteromeric nAChR, with MDMA showing lower Ki values, while fluorescence calcium experiments indicated that MDMA behaves as a partial agonist on α7 and as an antagonist on heteromeric nAChR. Sustained Ca increase led to calpain and caspase-3 activation. In addition, modulatory effects of MDMA on α7 and heteromeric nAChR populations have been found.
Collapse
Affiliation(s)
- David Pubill
- Author to whom correspondence should be addressed; E-Mails: ; Tel.: +34-93-402-4531; Fax: +34-93-403-5982
| | | | | | | |
Collapse
|
32
|
Northrop NA, Smith LP, Yamamoto BK, Eyerman DJ. Regulation of glutamate release by α7 nicotinic receptors: differential role in methamphetamine-induced damage to dopaminergic and serotonergic terminals. J Pharmacol Exp Ther 2011; 336:900-7. [PMID: 21159748 PMCID: PMC3061539 DOI: 10.1124/jpet.110.177287] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2010] [Accepted: 12/14/2010] [Indexed: 11/22/2022] Open
Abstract
Regulation of glutamate release is an important underlying mechanism in mediating excitotoxic events such as damage to dopamine (DA) and serotonin (5-HT) neurons observed after exposure to methamphetamine (Meth). One way to regulate glutamate release may be through the modulation of α7 nicotinic acetylcholine (nACh) receptors. Meth administration is known to increase acetylcholine release; however, it is unknown whether Meth increases glutamate release and causes long-term damage to both DA and 5-HT terminals through the activation of α7 nACh receptors. To test this hypothesis, the α7 nACh receptor antagonist, methyllycaconitine (MLA), was administered before the administration of repeated doses of Meth while simultaneously monitoring extracellular striatal glutamate with in vivo microdialysis. In addition, the subsequent long-term decreases in markers of dopaminergic and serotonergic terminals, including DA reuptake transporter (DAT), serotonin reuptake transporter (SERT), vesicular monoamine transporter-2, vesicular DA, and vesicular 5-HT content in the rat striatum, were measured. The results show that MLA pretreatment prevented Meth-induced increases in striatal glutamate and protected against the subsequent long-term decreases in striatal DAT and vesicular DA content without affecting the hyperthermia produced by Meth. In contrast, the Meth-induced decreases in striatal SERT immunoreactivity and vesicular 5-HT content were not affected by MLA. This suggests that the α7 nACh receptor differentially mediates glutamate-dependent damage to DA but not 5-HT terminals in a manner that is independent of hyperthermia. Furthermore, antagonism of α7 nACh receptors may be a possible therapeutic strategy for decreasing extracellular glutamate and preventing the excitotoxic damage observed in other DA-related neurodegenerative disorders.
Collapse
Affiliation(s)
- Nicole A Northrop
- Department of Pharmacology and Experimental Therapeutics, Boston University School of Medicine, Boston, Massachusetts, USA
| | | | | | | |
Collapse
|
33
|
Escubedo E, Abad S, Torres I, Camarasa J, Pubill D. Comparative neurochemical profile of 3,4-methylenedioxymethamphetamine and its metabolite alpha-methyldopamine on key targets of MDMA neurotoxicity. Neurochem Int 2011; 58:92-101. [DOI: 10.1016/j.neuint.2010.11.001] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2010] [Revised: 08/31/2010] [Accepted: 11/03/2010] [Indexed: 10/18/2022]
|
34
|
The effects of 3,4-methylenedioxymethamphetamine (MDMA) on nicotinic receptors: Intracellular calcium increase, calpain/caspase 3 activation, and functional upregulation. Toxicol Appl Pharmacol 2010; 244:344-53. [DOI: 10.1016/j.taap.2010.01.014] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2009] [Revised: 01/25/2010] [Accepted: 01/26/2010] [Indexed: 01/15/2023]
|
35
|
Lu P, Mamiya T, Lu L, Mouri A, Niwa M, Kim HC, Zou LB, Nagai T, Yamada K, Ikejima T. Silibinin attenuates cognitive deficits and decreases of dopamine and serotonin induced by repeated methamphetamine treatment. Behav Brain Res 2010; 207:387-93. [DOI: 10.1016/j.bbr.2009.10.024] [Citation(s) in RCA: 59] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2009] [Revised: 10/14/2009] [Accepted: 10/16/2009] [Indexed: 10/20/2022]
|
36
|
Cocaine-induced status epilepticus and death generate oxidative stress in prefrontal cortex and striatum of mice. Neurochem Int 2009; 56:183-7. [PMID: 19822180 DOI: 10.1016/j.neuint.2009.10.001] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2009] [Revised: 09/21/2009] [Accepted: 10/01/2009] [Indexed: 11/21/2022]
Abstract
Oxidative stress (OS) has been related to cocaine's actions and also to numerous nervous system pathologies, including seizures. The purpose of this work was to determine the alterations in glutathione (GSH) content, nitrite/nitrate and MDA levels after cocaine-induced toxicity. Male Swiss mice were injected (i.p.) with cocaine 90 mg/kg and observed during 1h. After this cocaine overdose some animals presented status epilepticus (SE) while some died after seizures. These animals were divided in two groups, SE and death. A group with an association of the antioxidant Vitamin E (Vit E, 400mg/kg, i.p.) plus Coc 90 (Vit E plus Coc 90) was undertaken to assess the neuroprotective effect of Vit E. Neurochemical analyses were carried out in prefrontal cortex (PFC) and striatum (ST). GSH levels increased only after cocaine-induced death in both areas studied. Cocaine-induced SE has increased nitrite/nitrate content in PFC and ST, while after death the increase was only in PFC. MDA (the lipid peroxidation marker) was elevated after SE and death in ST and only after death in PFC. Antioxidant treatment significantly reduced the GSH, nitrite/nitrate in ST and MDA levels. Only nitrite/nitrate content in PFC has not been decreased by Vit E pretreatment. The results relate that oxidative stress occurs after cocaine-induced toxicity mainly after death indicating that probably the increase of OS in the animal's brain leads to seizures and death, also showing a protective effect of Vit E in this process. Together with previous results this study contributes to the knowledge of cocaine-induced toxicity and possible in the near future to the use of antioxidants in the prevention of cocaine-induced CNS toxicity.
Collapse
|
37
|
Benedetto A, Au C, Aschner M. Manganese-Induced Dopaminergic Neurodegeneration: Insights into Mechanisms and Genetics Shared with Parkinson’s Disease. Chem Rev 2009; 109:4862-84. [DOI: 10.1021/cr800536y] [Citation(s) in RCA: 98] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Affiliation(s)
- Alexandre Benedetto
- Department of Pediatrics, Center for Molecular Neuroscience, Department of Pharmacology, and the Kennedy Center for Research on Human Development, Vanderbilt University Medical Center, Nashville, Tennessee 37232-0414
| | - Catherine Au
- Department of Pediatrics, Center for Molecular Neuroscience, Department of Pharmacology, and the Kennedy Center for Research on Human Development, Vanderbilt University Medical Center, Nashville, Tennessee 37232-0414
| | - Michael Aschner
- Department of Pediatrics, Center for Molecular Neuroscience, Department of Pharmacology, and the Kennedy Center for Research on Human Development, Vanderbilt University Medical Center, Nashville, Tennessee 37232-0414
| |
Collapse
|
38
|
Dreiem A, Shan M, Okoniewski RJ, Sanchez-Morrissey S, Seegal RF. Methylmercury inhibits dopaminergic function in rat pup synaptosomes in an age-dependent manner. Neurotoxicol Teratol 2009; 31:312-7. [PMID: 19464365 DOI: 10.1016/j.ntt.2009.05.001] [Citation(s) in RCA: 40] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2009] [Revised: 05/14/2009] [Accepted: 05/16/2009] [Indexed: 11/28/2022]
Abstract
Methylmercury (MeHg) is an environmental neurotoxicant that is especially harmful during brain development. Previously, we found greater sensitivity to MeHg-induced oxidative stress and greater loss of mitochondrial membrane potential in synaptosomes from early postnatal rats than in synaptosomes from older rat pups and adults. Here, we determine whether MeHg exposure also leads to greater changes in dopamine (DA) levels and dopamine transporter (DAT) function in synaptosomes from early postnatal rats. We report that MeHg exposure leads to DAT inhibition, and increases the levels of released DA compared to control; further, the effects are much greater in synaptosomes prepared from postnatal day (PND) 7 rats than in synaptosomes from PND 14 or PND 21 animals. In addition to the effects of MeHg in young rats, we observed age-dependent differences in dopaminergic function in unexposed synaptosomes: synaptosomal DA levels increased with age, whereas medium (released) DA levels were high at PND 7 and were lower in PND 14 and PND 21 synaptosomes. DAT activity increased slightly from PND 7 to PND 14 and then increased more strongly to PND 21, suggesting that higher DA release, in addition to the lower DAT activity seen in PND 7 animals, was responsible for the age differences in levels of released DA. These results demonstrate that MeHg affects the dopaminergic system during early development; it thus may contribute to the neurobehavioral effects seen in MeHg-exposed children.
Collapse
Affiliation(s)
- Anne Dreiem
- Wadsworth Center, New York State Department of Health, Empire State Plaza, Albany, NY 12201, USA
| | | | | | | | | |
Collapse
|
39
|
Thomas DM, Francescutti-Verbeem DM, Kuhn DM. Increases in cytoplasmic dopamine compromise the normal resistance of the nucleus accumbens to methamphetamine neurotoxicity. J Neurochem 2009; 109:1745-55. [PMID: 19457119 DOI: 10.1111/j.1471-4159.2009.06094.x] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/01/2022]
Abstract
Methamphetamine (METH) is a neurotoxic drug of abuse that damages the dopamine (DA) neuronal system in a highly delimited manner. The brain structure most affected by METH is the caudate-putamen (CPu) where long-term DA depletion and microglial activation are most evident. Even damage within the CPu is remarkably heterogenous with lateral and ventral aspects showing the greatest deficits. The nucleus accumbens (NAc) is largely spared of the damage that accompanies binge METH intoxication. Increases in cytoplasmic DA produced by reserpine, L-DOPA or clorgyline prior to METH uncover damage in the NAc as evidenced by microglial activation and depletion of DA, tyrosine hydroxylase (TH), and the DA transporter. These effects do not occur in the NAc after treatment with METH alone. In contrast to the CPu where DA, TH, and DA transporter levels remain depleted chronically, DA nerve ending alterations in the NAc show a partial recovery over time. None of the treatments that enhance METH toxicity in the NAc and CPu lead to losses of TH protein or DA cell bodies in the substantia nigra or the ventral tegmentum. These data show that increases in cytoplasmic DA dramatically broaden the neurotoxic profile of METH to include brain structures not normally targeted for damage by METH alone. The resistance of the NAc to METH-induced neurotoxicity and its ability to recover reveal a fundamentally different neuroplasticity by comparison to the CPu. Recruitment of the NAc as a target of METH neurotoxicity by alterations in DA homeostasis is significant in light of the important roles played by this brain structure.
Collapse
Affiliation(s)
- David M Thomas
- Department of Pharmaceutical Sciences, Eugene Applebaum College of Pharmacy and Health Sciences, Wayne State University, Detroit, Michigan, USA.
| | | | | |
Collapse
|
40
|
Escubedo E, Camarasa J, Chipana C, García-Ratés S, Pubill D. Involvement of nicotinic receptors in methamphetamine- and MDMA-induced neurotoxicity: pharmacological implications. INTERNATIONAL REVIEW OF NEUROBIOLOGY 2009; 88:121-66. [PMID: 19897077 DOI: 10.1016/s0074-7742(09)88006-9] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
During the last years, we have focused on the study of the neurotoxic effects of 3,4-methylenedioxymethamphetamine (MDMA) and methamphetamine (METH) on the central nervous system (CNS) and their pharmacological prevention methods. In the process of this research, we have used a semipurified synaptosomal preparation from striatum of mice or rats as a reliable in vitro model to study reactive oxygen species (ROS) production by these amphetamine derivatives, which is well-correlated with their dopaminergic injury in in vivo models. Using this preparation, we have demonstrated that blockade of alpha7 nicotinic receptors with methyllycaconitine (MLA) prevents ROS production induced by MDMA and METH. Consequently, in vivo, MLA significantly prevents MDMA- and METH-induced neurotoxicity at dopaminergic level (mouse striatum), without affecting hyperthermia induced by these amphetamines. Additionally, when neuroprotection was assayed with memantine (MEM), a dual antagonist of NMDA and alpha7 receptors, an effective neuroprotection was obtained also ahead of serotonergic injury induced by MDMA in rats. MEM also prevents MDMA effect on serotonin transporter functionality and METH effect on dopamine transporter (DAT), suggesting that behavioral effects of these psychostimulants can also be modulated by MEM. Finally, we have demonstrated that MEM prevents the impaired memory function induced by MDMA, and also, using binding studies with radioligands, we have characterized the interaction of these substances with nicotinic receptors. Studies at molecular level showed that both MDMA and METH displaced competitively the binding of radioligands with homomeric alpha7 and heteromeric nicotinic acetylcholine receptors (nAChRs), indicating that they can directly interact with them. In all the cases, MDMA displayed higher affinity than METH and it was higher for heteromeric than for alpha7 subtype. Pre-incubation of differentiated PC12 cells with MDMA or METH induces nAChR upregulation in a concentration- and time-dependent manner, as many nicotinic ligands do, supporting their functional interaction with nAChRs. Such interaction expands the pharmacological profile of amphetamines and can account for some of their effects.
Collapse
Affiliation(s)
- E Escubedo
- Unitat de Farmacologia i Farmacognósia, Facultat de Farmácia, Universitat de Barcelona, Barcelona 08028, Spain
| | | | | | | | | |
Collapse
|
41
|
Kuhn DM, Francescutti-Verbeem DM, Thomas DM. Dopamine disposition in the presynaptic process regulates the severity of methamphetamine-induced neurotoxicity. Ann N Y Acad Sci 2008; 1139:118-26. [PMID: 18991856 DOI: 10.1196/annals.1432.026] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022]
Abstract
Methamphetamine (METH) is well known for its ability to cause damage to dopamine (DA) nerve endings of the striatum. The mechanisms by which METH causes neurotoxicity are not fully understood, but likely candidates are increased oxidative and nitrosative stress and mitochondrial dysfunction. Microglial activation is also emerging as an important element of the METH neurotoxic cascade, and it appears that extensive cross-talk between these cells and DA nerve endings is an early event in this process. It may seem paradoxical, but DA itself is also thought to be an essential factor in the neuronal damaging effects of METH, but issues relating to its precise role in this regard remain unanswered. We present in this overview a summary of studies that tested how alterations in the disposition of presynaptic DA (injections of reserpine, L-DOPA, or clorgyline) modulate METH neurotoxicity. In all cases, these drugs significantly increased the magnitude of microglial activation as well as the severity of damage to striatal DA nerve endings caused by METH. The enhancement of METH effects in striatum by reserpine, L-DOPA, and clorgyline persisted for 14 days and showed no evidence of recovery. These data establish that subtle shifts in the newly synthesized pool of DA can cause substantial changes in the severity of METH-induced neurotoxicity. DA released into the synapse by METH is very likely the source of downstream reactants that provoke microglial activation and the ensuing damage to DA nerve endings.
Collapse
Affiliation(s)
- Donald M Kuhn
- Department of Psychiatry and Behavioral Neurosciences, Wayne State University School of Medicine, Detroit, Michigan, USA.
| | | | | |
Collapse
|
42
|
LoPachin RM, Gavin T, Barber DS. Type-2 alkenes mediate synaptotoxicity in neurodegenerative diseases. Neurotoxicology 2008; 29:871-82. [DOI: 10.1016/j.neuro.2008.04.016] [Citation(s) in RCA: 41] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2008] [Revised: 04/22/2008] [Accepted: 04/23/2008] [Indexed: 01/23/2023]
|
43
|
Pinthong M, Black SAG, Ribeiro FM, Pholpramool C, Ferguson SSG, Rylett RJ. Activity and subcellular trafficking of the sodium-coupled choline transporter CHT is regulated acutely by peroxynitrite. Mol Pharmacol 2008; 73:801-12. [PMID: 17971421 DOI: 10.1124/mol.107.040881] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/14/2025] Open
Abstract
Excess formation of nitric oxide and superoxide by-products (peroxynitrite, reactive oxygen, and reactive nitrogen species) attenuates cholinergic transmission potentially having a role in Alzheimer disease pathogenesis. In this study, we investigated mechanisms by which acute exposure to peroxynitrite impairs function of the sodium-dependent hemicholinium-3 (HC-3)-sensitive choline transporter (CHT) that provides substrate for acetylcholine synthesis. The peroxynitrite generator 3-morpholinosydnonimine (SIN-1) acutely inhibited choline uptake in cells stably expressing FLAG-tagged rat CHT in a dose- and time-dependent manner, with an IC(50) = 0.9 +/- 0.14 mM and t((1/2)) = 4 min. SIN-1 significantly reduced V(max) of choline uptake without altering the K(m). This correlated with a SIN-1-induced decrease in cell surface CHT protein, observed as lowered levels of HC-3 binding and biotinylated CHT at the plasma membrane. It is noteworthy that short-term exposure of cells to SIN-1 accelerated the rate of internalization of CHT from the plasma membrane, but it did not alter return of CHT back to the cell surface. SIN-1 did not disrupt cell membrane integrity or cause cell death. Thus, the inhibitory effect of SIN-1 on choline uptake activity and HC-3 binding was related to enhanced internalization of CHT proteins from the plasma membrane to subcellular organelles.
Collapse
Affiliation(s)
- Metta Pinthong
- Department of Physiology and Pharmacology, Schulich School of Medicine and Dentistry, University of Western Ontario, London, ON, Canada
| | | | | | | | | | | |
Collapse
|
44
|
Modulation of dopamine uptake by nitric oxide in cultured mesencephalic neurons. Brain Res 2008; 1198:27-33. [DOI: 10.1016/j.brainres.2007.12.054] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2007] [Revised: 12/11/2007] [Accepted: 12/15/2007] [Indexed: 12/29/2022]
|
45
|
Chipana C, García-Ratés S, Camarasa J, Pubill D, Escubedo E. Different oxidative profile and nicotinic receptor interaction of amphetamine and 3,4-methylenedioxy-methamphetamine. Neurochem Int 2008; 52:401-10. [PMID: 17716785 DOI: 10.1016/j.neuint.2007.07.016] [Citation(s) in RCA: 31] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2007] [Revised: 07/10/2007] [Accepted: 07/18/2007] [Indexed: 11/20/2022]
Abstract
d-Amphetamine (AMPH) and MDMA increased intracellular production of reactive oxygen species (ROS) in isolated mouse striatal synaptosomes. MDMA showed a maximal oxidative effect at 50-100 microM. However, for AMPH a double maximum was obtained, the first between 0.1 and 1 microM and the second at 1mM. No oxidative effect was present in synaptosomes from reserpinized mice. Cocaine and l-deprenyl inhibited MDMA and AMPH (0.1 microM) ROS production but not that of AMPH at a higher concentration (1mM). When this high concentration was used, its oxidative effect was abolished by a phospholipase A(2) inhibitor. Delta(9)-Tetrahydrocannabinol fully prevented the oxidative effect of AMPH and MDMA, by a CB(1) receptor-independent mechanism, as did it NPC 15437 and genistein. The pro-oxidative effect induced by AMPH and MDMA showed a strong dependence on calcium (extracellular and from internal stores) and also was inhibited by nicotinic receptor (nAChR) antagonists dihydro-beta-erythroidine, methyllycaconitine (MLA) and alpha-bungarotoxin. MDMA displaced [(3)H]epibatidine and [(3)H]MLA binding with higher affinity than AMPH. Both amphetamines competitively displaced [(3)H]epibatidine from heteromeric receptors but results obtained from [(3)H]MLA binding demonstrated a non-competitive profile. Preincubation of PC12 cells with AMPH or MDMA reduced [(3)H]dopamine uptake. For MDMA, this effect was prevented by MLA. To summarize, comparing AMPH and MDMA we have demonstrated that these drugs induce an oxidative effect dependent on drug concentration and also reduce dopamine uptake. Processes that are known to affect dopamine transporter functionality also seem to modulate amphetamine derivatives-induced ROS production. For MDMA, acute effects tested are blocked by nAChR antagonists, which points to the possibility that these antagonists could be used to treat some of the adverse effects described in MDMA abusers. Conversely, no implication of nicotinic receptors has been proved for AMPH-induced effects at concentrations achievable in CNS after its administration.
Collapse
Affiliation(s)
- C Chipana
- Unitat de Farmacologia i Farmacognòsia, Facultat de Farmàcia, Nucli Universitari de Pedralbes, Universitat de Barcelona, 08028 Barcelona, Spain
| | | | | | | | | |
Collapse
|
46
|
LoPachin RM, Barber DS, Gavin T. Molecular mechanisms of the conjugated alpha,beta-unsaturated carbonyl derivatives: relevance to neurotoxicity and neurodegenerative diseases. Toxicol Sci 2007; 104:235-49. [PMID: 18083715 DOI: 10.1093/toxsci/kfm301] [Citation(s) in RCA: 125] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Conjugated alpha,beta-unsaturated carbonyl derivatives such acrylamide, acrolein, and 4-hydroxy-2-nonenal (HNE) are members of a large class of chemicals known as the type-2 alkenes. Human exposure through diet, occupation, and pollution is pervasive and has been linked to toxicity in most major organs. Evidence suggests that these soft electrophiles produce toxicity by a common mechanism involving the formation of Michael-type adducts with nucleophilic sulfhydryl groups. In this commentary, the adduct chemistry of the alpha,beta-unsaturated carbonyls and possible protein targets will be reviewed. We also consider how differences in electrophilic reactivity among the type-2 alkenes impact corresponding toxicokinetics and toxicological expression. Whereas these concepts have mechanistic implications for the general toxicity of type-2 alkenes, this commentary will focus on the ability of these chemicals to produce presynaptic damage via protein adduct formation. Given the ubiquitous environmental presence of the conjugated alkenes, discussions of molecular mechanisms and possible neurotoxicological risks could be important. Understanding the neurotoxicodynamic of the type-2 alkenes might also provide mechanistic insight into neurodegenerative conditions where neuronal oxidative stress and presynaptic dysfunction are presumed initiating events. This is particularly germane to a recent proposal that lipid peroxidation and the subsequent liberation of acrolein and HNE in oxidatively stressed neurons mediate synaptotoxicity in brains of Alzheimer's disease patients. This endogenous neuropathogenic process could be accelerated by environmental type-2 alkene exposure because common nerve terminal proteins are targeted by alpha,beta-unsaturated carbonyl derivatives. Thus, the protein adduct chemistry of the conjugated type-2 alkenes offers a mechanistic explanation for the environmental toxicity induced by these chemicals and might provide insight into the pathogenesis of certain human neurodegenerative diseases.
Collapse
Affiliation(s)
- Richard M LoPachin
- Department of Anesthesiology, Albert Einstein College of Medicine, Montefiore Medical Center, Bronx, New York 10467-2490, USA.
| | | | | |
Collapse
|
47
|
Thomas DM, Francescutti-Verbeem DM, Kuhn DM. The newly synthesized pool of dopamine determines the severity of methamphetamine-induced neurotoxicity. J Neurochem 2007; 105:605-16. [PMID: 18088364 DOI: 10.1111/j.1471-4159.2007.05155.x] [Citation(s) in RCA: 100] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
Abstract
The neurotransmitter dopamine (DA) has long been implicated as a participant in the neurotoxicity caused by methamphetamine (METH), yet, its mechanism of action in this regard is not fully understood. Treatment of mice with the tyrosine hydroxylase (TH) inhibitor alpha-methyl-p-tyrosine (AMPT) lowers striatal cytoplasmic DA content by 55% and completely protects against METH-induced damage to DA nerve terminals. Reserpine, by disrupting vesicle amine storage, depletes striatal DA by more than 95% and accentuates METH-induced neurotoxicity. l-DOPA reverses the protective effect of AMPT against METH and enhances neurotoxicity in animals with intact TH. Inhibition of MAO-A by clorgyline increases pre-synaptic DA content and enhances METH striatal neurotoxicity. In all conditions of altered pre-synaptic DA homeostasis, increases or decreases in METH neurotoxicity paralleled changes in striatal microglial activation. Mice treated with AMPT, l-DOPA, or clorgyline + METH developed hyperthermia to the same extent as animals treated with METH alone, whereas mice treated with reserpine + METH were hypothermic, suggesting that the effects of alterations in cytoplasmic DA on METH neurotoxicity were not strictly mediated by changes in core body temperature. Taken together, the present data reinforce the notion that METH-induced release of DA from the newly synthesized pool of transmitter into the extracellular space plays an essential role in drug-induced striatal neurotoxicity and microglial activation. Subtle alterations in intracellular DA content can lead to significant enhancement of METH neurotoxicity. Our results also suggest that reactants derived from METH-induced oxidation of released DA may serve as neuronal signals that lead to microglial activation early in the neurotoxic process associated with METH.
Collapse
Affiliation(s)
- David M Thomas
- Department of Psychiatry & Behavioral Neurosciences, Wayne State University School of Medicine, Detroit, Michigan, USA.
| | | | | |
Collapse
|
48
|
Barber DS, Stevens S, LoPachin RM. Proteomic analysis of rat striatal synaptosomes during acrylamide intoxication at a low dose rate. Toxicol Sci 2007; 100:156-67. [PMID: 17698512 DOI: 10.1093/toxsci/kfm210] [Citation(s) in RCA: 63] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
Abstract
We have hypothesized that acrylamide (ACR) intoxication causes cumulative nerve terminal damage by forming adducts with nucleophilic cysteine sulfhydryl groups on critical presynaptic proteins. To determine the cumulative effects of ACR on the cysteine-containing proteome of nerve terminal, we employed cleavable isotope-coded affinity tagging (ICAT) and liquid chromatography-tandem mass spectrometry. ICAT analysis uses a sulfhydryl-specific tag to identify and quantitate cysteine-containing proteins. Synaptosomes were prepared from striatum of ACR-intoxicated rats (21 mg/kg/day x 7, 14, or 21 days) and their age-matched controls. The synaptosomal proteins of each experimental group were labeled with either light (12C9--control) or heavy (13C9--ACR) ICAT reagent. Results show that ACR intoxication caused a progressive reduction in the ICAT labeling of many nerve terminal proteins. A label-free mass spectrometric approach (multidimensional protein identification) was used to show that the observed reductions in ICAT incorporation were not due to general changes in protein abundance and that ACR formed adducts with cysteine residues on peptides which also exhibited reduced ICAT incorporation. The decrease in labeling was temporally correlated to the development of neurological toxicity and confirmed previous findings that cysteine adducts of ACR accumulate as a function of exposure. The accumulation of adduct is consistent with the cumulative neurotoxicity induced by ACR and suggests that cysteine adduct formation is a necessary neuropathogenic step. Furthermore, our analyses identified specific proteins (e.g., v-ATPase, dopamine transporter, N-ethylmaleimide-sensitive factor) that were progressively and significantly adducted by ACR and might, therefore, be neurotoxicologically relevant targets.
Collapse
Affiliation(s)
- David S Barber
- Center for Environmental and Human Toxicology, University of Florida, Building 471, Mowry Road, Gainesville, Florida 32611-0885, USA
| | | | | |
Collapse
|
49
|
Eyerman DJ, Yamamoto BK. A rapid oxidation and persistent decrease in the vesicular monoamine transporter 2 after methamphetamine. J Neurochem 2007; 103:1219-27. [PMID: 17683483 DOI: 10.1111/j.1471-4159.2007.04837.x] [Citation(s) in RCA: 60] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Methamphetamine (METH) produces long-term decreases in markers of dopamine (DA) terminals in animals and humans. A decrease in the function of the vesicular monoamine transporter 2 (VMAT2) has been associated with damage to striatal DA terminals caused by METH; however, a possible mechanism for this decrease in VMAT2 function has not been defined. The current study showed that METH caused a rapid decrease to 68% of controls in VMAT2 protein immunoreactivity of the vesicular fraction from striatal synaptosomes within 1 h after a repeated high-dose administration regimen of METH. This decrease was associated with a 75% increase in nitrosylation of VMAT2 protein in the synaptosomal fraction as measured by nitrosocysteine immunoreactivity of VMAT2 protein. The rapid decreases in VMAT2 persisted when evaluated 7 days later and were illustrated by decreases in VMAT2 immunoreactivity and DA content of the vesicular fraction to 34% and 51% of control values, respectively. The decreases were blocked or attenuated by prior injections of the neuronal nitric oxide synthase inhibitor, S-methyl-l-thiocitrulline. These studies demonstrate that METH causes a rapid neuronal nitric oxide synthase-dependent oxidation of VMAT2 and long-term decreases in VMAT2 protein and function. The results also suggest that surviving DA terminals after METH exposure may have a compromised capacity to buffer cytosolic DA concentrations and DA-derived oxidative stress.
Collapse
Affiliation(s)
- David J Eyerman
- Laboratory of Neurochemistry, Department of Pharmacology and Experimental Therapeutics, Boston University School of Medicine, Boston, Massachusetts, USA
| | | |
Collapse
|
50
|
Jung T, Engels M, Klotz LO, Kröncke KD, Grune T. Nitrotyrosine and protein carbonyls are equally distributed in HT22 cells after nitrosative stress. Free Radic Biol Med 2007; 42:773-86. [PMID: 17320760 DOI: 10.1016/j.freeradbiomed.2006.11.029] [Citation(s) in RCA: 34] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/18/2006] [Revised: 11/22/2006] [Accepted: 11/28/2006] [Indexed: 11/24/2022]
Abstract
The generation of reactive oxygen and nitrogen species is an inevitable result of cellular metabolism and environmental influence. Such oxidation processes are always combined with the formation of various protein oxidation products. Environmental oxidants might either be activated inside the cell or act by themselves. Therefore, differences in the localization of oxidant formation might change the major compartment of oxidant action. Therefore, we employed NO donors (SNOC, DETA/NO, and Spe/NO) alone or in combination with the redox-cycling bipyridinium compound paraquat, the superoxide- and NO-releasing compound SIN-1, the relatively more lipophilic oxidants tert-butyl and cumene hydroperoxide, and peroxynitrite itself to test the ability of these compounds to generate oxidized and nitrated proteins in various cellular compartments. Combined treatment with oxidants and nitrating compounds led to the formation of protein carbonyls and nitrotyrosine with a severalfold higher concentration in the cytosol, compared to the nucleus. In fluorescence microscopy studies, the resulting protein modifications show a similar distribution of oxidized proteins and nitrotyrosine with highest concentrations in the perinuclear area. Studying the time- and concentration-dependent formation and degradation of protein carbonyls and nitrated proteins large similarities could be measured. Therefore, it can be concluded that formation, localization, and kinetics of protein carbonyl and nitrotyrosine formation parallel each other depending on the stress-inducing agent.
Collapse
Affiliation(s)
- Tobias Jung
- Research Institute of Environmental Medicine, Heinrich Heine University, Auf'm Hennekamp 50, 40225 Duesseldorf, Germany
| | | | | | | | | |
Collapse
|