1
|
Thöny B, Ng J, Kurian MA, Mills P, Martinez A. Mouse models for inherited monoamine neurotransmitter disorders. J Inherit Metab Dis 2024; 47:533-550. [PMID: 38168036 DOI: 10.1002/jimd.12710] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/27/2023] [Revised: 11/07/2023] [Accepted: 12/19/2023] [Indexed: 01/05/2024]
Abstract
Several mouse models have been developed to study human defects of primary and secondary inherited monoamine neurotransmitter disorders (iMND). As the field continues to expand, current defects in corresponding mouse models include enzymes and a molecular co-chaperone involved in monoamine synthesis and metabolism (PAH, TH, PITX3, AADC, DBH, MAOA, DNAJC6), tetrahydrobiopterin (BH4) cofactor synthesis and recycling (adGTPCH1/DRD, arGTPCH1, PTPS, SR, DHPR), and vitamin B6 cofactor deficiency (ALDH7A1), as well as defective monoamine neurotransmitter packaging (VMAT1, VMAT2) and reuptake (DAT). No mouse models are available for human DNAJC12 co-chaperone and PNPO-B6 deficiencies, disorders associated with recessive variants that result in decreased stability and function of the aromatic amino acid hydroxylases and decreased neurotransmitter synthesis, respectively. More than one mutant mouse is available for some of these defects, which is invaluable as different variant-specific (knock-in) models may provide more insights into underlying mechanisms of disorders, while complete gene inactivation (knock-out) models often have limitations in terms of recapitulating complex human diseases. While these mouse models have common phenotypic traits also observed in patients, reflecting the defective homeostasis of the monoamine neurotransmitter pathways, they also present with disease-specific manifestations with toxic accumulation or deficiency of specific metabolites related to the specific gene affected. This review provides an overview of the currently available models and may give directions toward selecting existing models or generating new ones to investigate novel pathogenic mechanisms and precision therapies.
Collapse
Affiliation(s)
- Beat Thöny
- Division of Metabolism and Children's Research Center, University Children's Hospital Zurich, Zürich, Switzerland
| | - Joanne Ng
- Genetic Therapy Accelerator Centre, University College London, Queen Square Institute of Neurology, London, UK
| | - Manju A Kurian
- Zayed Centre for Research into Rare Disease in Children, GOS Institute of Child Health, University College London, London, UK
- Department of Neurology, Great Ormond Street Hospital, London, UK
| | - Philippa Mills
- Great Ormond Street Institute of Child Health, University College London, London, UK
| | - Aurora Martinez
- Department of Biomedicine and Center for Translational Research in Parkinson's Disease, University of Bergen, Bergen, Norway
- Neuro-SysMed, Department of Neurology, Haukeland University Hospital, Bergen, Norway
| |
Collapse
|
2
|
Singh P, Zhou L, Shah DA, Cejas RB, Crossman DK, Jouni M, Magdy T, Wang X, Sharafeldin N, Hageman L, McKenna DE, Horvath S, Armenian SH, Balis FM, Hawkins DS, Keller FG, Hudson MM, Neglia JP, Ritchey AK, Ginsberg JP, Landier W, Burridge PW, Bhatia S. Identification of novel hypermethylated or hypomethylated CpG sites and genes associated with anthracycline-induced cardiomyopathy. Sci Rep 2023; 13:12683. [PMID: 37542143 PMCID: PMC10403495 DOI: 10.1038/s41598-023-39357-2] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2023] [Accepted: 07/24/2023] [Indexed: 08/06/2023] Open
Abstract
Anthracycline-induced cardiomyopathy is a leading cause of late morbidity in childhood cancer survivors. Aberrant DNA methylation plays a role in de novo cardiovascular disease. Epigenetic processes could play a role in anthracycline-induced cardiomyopathy but remain unstudied. We sought to examine if genome-wide differential methylation at 'CpG' sites in peripheral blood DNA is associated with anthracycline-induced cardiomyopathy. This report used participants from a matched case-control study; 52 non-Hispanic White, anthracycline-exposed childhood cancer survivors with cardiomyopathy were matched 1:1 with 52 survivors with no cardiomyopathy. Paired ChAMP (Chip Analysis Methylation Pipeline) with integrated reference-based deconvolution of adult peripheral blood DNA methylation was used to analyze data from Illumina HumanMethylation EPIC BeadChip arrays. An epigenome-wide association study (EWAS) was performed, and the model was adjusted for GrimAge, sex, interaction terms of age at enrollment, chest radiation, age at diagnosis squared, and cardiovascular risk factors (CVRFs: diabetes, hypertension, dyslipidemia). Prioritized genes were functionally validated by gene knockout in human induced pluripotent stem cell cardiomyocytes (hiPSC-CMs) using CRISPR/Cas9 technology. DNA-methylation EPIC array analyses identified 32 differentially methylated probes (DMP: 15 hyper-methylated and 17 hypo-methylated probes) that overlap with 23 genes and 9 intergenic regions. Three hundred and fifty-four differential methylated regions (DMRs) were also identified. Several of these genes are associated with cardiac dysfunction. Knockout of genes EXO6CB, FCHSD2, NIPAL2, and SYNPO2 in hiPSC-CMs increased sensitivity to doxorubicin. In addition, EWAS analysis identified hypo-methylation of probe 'cg15939386' in gene RORA to be significantly associated with anthracycline-induced cardiomyopathy. In this genome-wide DNA methylation profile study, we observed significant differences in DNA methylation at the CpG level between anthracycline-exposed childhood cancer survivors with and without cardiomyopathy, implicating differential DNA methylation of certain genes could play a role in pathogenesis of anthracycline-induced cardiomyopathy.
Collapse
Affiliation(s)
- Purnima Singh
- Institute for Cancer Outcomes and Survivorship, University of Alabama at Birmingham, Birmingham, AL, USA
- Department of Pediatrics, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Liting Zhou
- Institute for Cancer Outcomes and Survivorship, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Disheet A Shah
- Department of Pharmacology, Northwestern University, Chicago, IL, USA
| | - Romina B Cejas
- Department of Pharmacology, Northwestern University, Chicago, IL, USA
| | - David K Crossman
- Department of Genetics, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Mariam Jouni
- Department of Pharmacology, Northwestern University, Chicago, IL, USA
| | - Tarek Magdy
- Department of Pharmacology, Northwestern University, Chicago, IL, USA
- Department of Pathology and Translational Pathobiology and Feist-Weiller Cancer Center, Louisiana State University Health Sciences Center-Shreveport, Shreveport, LA, USA
| | - Xuexia Wang
- Department of Biostatistics, Florida International University, Miami, FL, USA
| | - Noha Sharafeldin
- Institute for Cancer Outcomes and Survivorship, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Lindsey Hageman
- Institute for Cancer Outcomes and Survivorship, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Donald E McKenna
- Department of Pharmacology, Northwestern University, Chicago, IL, USA
| | - Steve Horvath
- Department of Human Genetics, David Geffen School of Medicine, University of California, Los Angeles, CA, USA
| | - Saro H Armenian
- Department of Population Sciences, City of Hope, Duarte, CA, USA
| | - Frank M Balis
- Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | | | - Frank G Keller
- Children's Healthcare of Atlanta, Emory University, Atlanta, GA, USA
| | | | | | - A Kim Ritchey
- Children's Hospital of Pittsburgh of UPMC, Pittsburgh, PA, USA
| | | | - Wendy Landier
- Institute for Cancer Outcomes and Survivorship, University of Alabama at Birmingham, Birmingham, AL, USA
- Department of Pediatrics, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Paul W Burridge
- Department of Pharmacology, Northwestern University, Chicago, IL, USA
| | - Smita Bhatia
- Institute for Cancer Outcomes and Survivorship, University of Alabama at Birmingham, Birmingham, AL, USA.
- Department of Pediatrics, University of Alabama at Birmingham, Birmingham, AL, USA.
| |
Collapse
|
3
|
Nuñez NA, Salgado MF, Frye MA. Stimulants in Bipolar Depression: Risks and Benefits. Psychiatr Ann 2023. [DOI: 10.3928/00485713-20230201-01] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/17/2023]
|
4
|
The regulatory role of AP-2β in monoaminergic neurotransmitter systems: insights on its signalling pathway, linked disorders and theragnostic potential. Cell Biosci 2022; 12:151. [PMID: 36076256 PMCID: PMC9461128 DOI: 10.1186/s13578-022-00891-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2022] [Accepted: 08/28/2022] [Indexed: 11/10/2022] Open
Abstract
AbstractMonoaminergic neurotransmitter systems play a central role in neuronal function and behaviour. Dysregulation of these systems gives rise to neuropsychiatric and neurodegenerative disorders with high prevalence and societal burden, collectively termed monoamine neurotransmitter disorders (MNDs). Despite extensive research, the transcriptional regulation of monoaminergic neurotransmitter systems is not fully explored. Interestingly, certain drugs that act on these systems have been shown to modulate central levels of the transcription factor AP-2 beta (AP-2β, gene: TFAP2Β). AP-2β regulates multiple key genes within these systems and thereby its levels correlate with monoamine neurotransmitters measures; yet, its signalling pathways are not well understood. Moreover, although dysregulation of TFAP2Β has been associated with MNDs, the underlying mechanisms for these associations remain elusive. In this context, this review addresses AP-2β, considering its basic structural aspects, regulation and signalling pathways in the controlling of monoaminergic neurotransmitter systems, and possible mechanisms underpinning associated MNDS. It also underscores the significance of AP-2β as a potential diagnostic biomarker and its potential and limitations as a therapeutic target for specific MNDs as well as possible pharmaceutical interventions for targeting it. In essence, this review emphasizes the role of AP-2β as a key regulator of the monoaminergic neurotransmitter systems and its importance for understanding the pathogenesis and improving the management of MNDs.
Collapse
|
5
|
Liu MM, Zhou N, Jiang N, Lu KM, Wu CF, Bao JK. Neuroprotective Effects of Oligosaccharides From Periplaneta Americana on Parkinson’s Disease Models In Vitro and In Vivo. Front Pharmacol 2022; 13:936818. [PMID: 35924055 PMCID: PMC9340460 DOI: 10.3389/fphar.2022.936818] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2022] [Accepted: 05/31/2022] [Indexed: 11/22/2022] Open
Abstract
Parkinson’s disease (PD) is one of the neurodegenerative diseases that is characterized by obvious motor and some nonmotor symptoms. Various therapeutics failed in the effective treatment of PD because of impaired neurological function in the brain and various complications. Periplaneta Americana oligosaccharides (OPA), the main active ingredients extracted from the medicine residues of Periplaneta Americana (P. Americana), have been reported to exert anti-inflammatory effects. The purpose of this study was to evaluate the possible mechanisms of OPA against 1-methyl-4-phenylpyridinium (MPP+)-induced apotosis in SH-SY5Y cells and its potential neuroprotective effects in 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP)-induced PD subacute model mice. The data demonstrated that OPA significantly reversed the MPP+-induced decrease in SH-SY5Y cell viability, reduced the proportion of apoptotic cells, and protected SH-SY5Y cells from apoptosis in a dose-dependent manner by regulating the expression of apoptosis-related genes. Furthermore, OPA also alleviated the motor dysfunction of PD model mice, prevented the loss of tyrosine hydroxylase positive cells, suppressed the apoptosis of substantia nigra cells, and improved the dysbiosis of gut microbiota in vivo, suggesting that OPA demonstrated a significantly neuroprotective effect on PD model mice. These results indicated that OPA might be the possibility of PD therapeutics with economic utility and high safety.
Collapse
Affiliation(s)
- Miao-Miao Liu
- Key Laboratory of Bio-Resource and Eco-Environment of Ministry of Education, College of Life Sciences, Sichuan University, Chengdu, China
| | - Nan Zhou
- Key Laboratory of Bio-Resource and Eco-Environment of Ministry of Education, College of Life Sciences, Sichuan University, Chengdu, China
| | - Na Jiang
- Key Laboratory of Bio-Resource and Eco-Environment of Ministry of Education, College of Life Sciences, Sichuan University, Chengdu, China
| | - Kai-Min Lu
- Key Laboratory of Bio-Resource and Eco-Environment of Ministry of Education, College of Life Sciences, Sichuan University, Chengdu, China
- Pharmacy Research Center, Binzhou Medical University, Yantai, China
| | - Chuan-Fang Wu
- Key Laboratory of Bio-Resource and Eco-Environment of Ministry of Education, College of Life Sciences, Sichuan University, Chengdu, China
- *Correspondence: Chuan-Fang Wu, ; Jin-Ku Bao,
| | - Jin-Ku Bao
- Key Laboratory of Bio-Resource and Eco-Environment of Ministry of Education, College of Life Sciences, Sichuan University, Chengdu, China
- *Correspondence: Chuan-Fang Wu, ; Jin-Ku Bao,
| |
Collapse
|
6
|
Wal P, Dwivedi J, Wal A, Vig H, Singh Y. Detailed insight into the pathophysiology and the behavioral complications associated with the Parkinson's disease and its medications. FUTURE JOURNAL OF PHARMACEUTICAL SCIENCES 2022. [DOI: 10.1186/s43094-022-00425-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022] Open
Abstract
Abstract
Background
The loss of dopamine neurons in the substantia nigra, as well as other mostly catecholaminergic neurons, causes many of the motor symptoms that define Parkinson's disease. Parkinson's disease is commonly thought of as a movement disorder, the significant prevalence of psychiatric complications such as cognitive impairment, and psychosis suggests it should be considered a neuropsychiatric illness, and all behavioral complications are linked to growing disability and the medication.
Main body
Apart from the disease-induced abnormalities, there are several other side effects of the disease and also from the medication used to prevent the disease. This article focuses on the pathogenesis of Parkinson’s disease and also the behavioral abnormalities caused by the disease and its medication. The study's data were gathered by searching several review articles and research papers from a variety of sources, including Elsevier, PubMed, Research Gate, Journal of Pharmaceutical Science, etc., from the year 1985 to 2021. Parkinson's disease is a neurodegenerative disease caused by a variety of complex processes. It is responsible not just for motor symptoms, but also for a variety of behavioral symptoms that can arise as a result of the disease and/or medication.
Conclusion
Only symptomatic drugs are available; thus, finding treatments that directly address the disease mechanisms causing Parkinson’s disease is essential. To alleviate the disease's burden on patients and their families, better treatments for the neuropsychiatric repercussions of Parkinson's disease are required.
Graphical Abstract
Collapse
|
7
|
Sheardown E, Mech AM, Petrazzini MEM, Leggieri A, Gidziela A, Hosseinian S, Sealy IM, Torres-Perez JV, Busch-Nentwich EM, Malanchini M, Brennan CH. Translational relevance of forward genetic screens in animal models for the study of psychiatric disease. Neurosci Biobehav Rev 2022; 135:104559. [PMID: 35124155 PMCID: PMC9016269 DOI: 10.1016/j.neubiorev.2022.104559] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2021] [Revised: 12/10/2021] [Accepted: 02/01/2022] [Indexed: 12/16/2022]
Abstract
Psychiatric disorders represent a significant burden in our societies. Despite the convincing evidence pointing at gene and gene-environment interaction contributions, the role of genetics in the etiology of psychiatric disease is still poorly understood. Forward genetic screens in animal models have helped elucidate causal links. Here we discuss the application of mutagenesis-based forward genetic approaches in common animal model species: two invertebrates, nematodes (Caenorhabditis elegans) and fruit flies (Drosophila sp.); and two vertebrates, zebrafish (Danio rerio) and mice (Mus musculus), in relation to psychiatric disease. We also discuss the use of large scale genomic studies in human populations. Despite the advances using data from human populations, animal models coupled with next-generation sequencing strategies are still needed. Although with its own limitations, zebrafish possess characteristics that make them especially well-suited to forward genetic studies exploring the etiology of psychiatric disorders.
Collapse
Affiliation(s)
- Eva Sheardown
- School of Biological and Behavioural Sciences, Queen Mary University of London, Mile End Road, London E1 4NS, England, UK
| | - Aleksandra M Mech
- School of Biological and Behavioural Sciences, Queen Mary University of London, Mile End Road, London E1 4NS, England, UK
| | | | - Adele Leggieri
- School of Biological and Behavioural Sciences, Queen Mary University of London, Mile End Road, London E1 4NS, England, UK
| | - Agnieszka Gidziela
- School of Biological and Behavioural Sciences, Queen Mary University of London, Mile End Road, London E1 4NS, England, UK
| | - Saeedeh Hosseinian
- School of Biological and Behavioural Sciences, Queen Mary University of London, Mile End Road, London E1 4NS, England, UK
| | - Ian M Sealy
- Cambridge Institute of Therapeutic Immunology & Infectious Disease (CITIID), Department of Medicine, University of Cambridge, Cambridge, UK
| | - Jose V Torres-Perez
- UK Dementia Research Institute at Imperial College London and Department of Brain Sciences, Imperial College London, 86 Wood Lane, London W12 0BZ, UK
| | - Elisabeth M Busch-Nentwich
- School of Biological and Behavioural Sciences, Queen Mary University of London, Mile End Road, London E1 4NS, England, UK
| | - Margherita Malanchini
- School of Biological and Behavioural Sciences, Queen Mary University of London, Mile End Road, London E1 4NS, England, UK
| | - Caroline H Brennan
- School of Biological and Behavioural Sciences, Queen Mary University of London, Mile End Road, London E1 4NS, England, UK.
| |
Collapse
|
8
|
Baronio D, Chen YC, Decker AR, Enckell L, Fernández-López B, Semenova S, Puttonen HAJ, Cornell RA, Panula P. Vesicular monoamine transporter 2 (SLC18A2) regulates monoamine turnover and brain development in zebrafish. Acta Physiol (Oxf) 2022; 234:e13725. [PMID: 34403568 DOI: 10.1111/apha.13725] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2020] [Revised: 08/09/2021] [Accepted: 08/13/2021] [Indexed: 01/22/2023]
Abstract
AIM We aimed at identifying potential roles of vesicular monoamine transporter 2, also known as Solute Carrier protein 18 A2 (SLC18A2) (hereafter, Vmat2), in brain monoamine regulation, their turnover, behaviour and brain development using a novel zebrafish model. METHODS A zebrafish strain lacking functional Vmat2 was generated with the CRISPR/Cas9 system. Larval behaviour and heart rate were monitored. Monoamines and their metabolites were analysed with high-pressure liquid chromatography. Amine synthesising and degrading enzymes, and genes essential for brain development, were analysed with quantitative PCR, in situ hybridisation and immunocytochemistry. RESULTS The 5-bp deletion in exon 3 caused an early frameshift and was lethal within 2 weeks post-fertilisation. Homozygous mutants (hereafter, mutants) displayed normal low locomotor activity during night-time but aberrant response to illumination changes. In mutants dopamine, noradrenaline, 5-hydroxytryptamine and histamine levels were reduced, whereas levels of dopamine and 5-hydroxytryptamine metabolites were increased, implying elevated monoamine turnover. Consistently, there were fewer histamine, 5-hydroxytryptamine and dopamine immunoreactive cells. Cellular dopamine immunostaining, in wild-type larvae more prominent in tyrosine hydroxylase 1 (Th1)-expressing than in Th2-expressing neurons, was absent in mutants. Despite reduced dopamine levels, mutants presented upregulated dopamine-synthesising enzymes. Further, in mutants the number of histidine decarboxylase-expressing neurons was increased, notch1a and pax2a were downregulated in brain proliferative zones. CONCLUSION Lack of Vmat2 increases monoamine turnover and upregulates genes encoding amine-synthesising enzymes, including histidine decarboxylase. Notch1a and pax2a, genes implicated in stem cell development, are downregulated in mutants. The zebrafish vmat2 mutant strain may be a useful model to study how monoamine transport affects brain development and function, and for use in drug screening.
Collapse
Affiliation(s)
- Diego Baronio
- Department of Anatomy, University of Helsinki, Helsinki, Finland
| | - Yu-Chia Chen
- Department of Anatomy, University of Helsinki, Helsinki, Finland
| | - Amanda R Decker
- Department of Anatomy and Cell Biology, University of Iowa, Iowa City, USA
- Iowa Neuroscience Institute, University of Iowa, Iowa City, USA
| | - Louise Enckell
- Department of Anatomy, University of Helsinki, Helsinki, Finland
| | | | | | | | - Robert A Cornell
- Department of Anatomy and Cell Biology, University of Iowa, Iowa City, USA
| | - Pertti Panula
- Department of Anatomy, University of Helsinki, Helsinki, Finland
| |
Collapse
|
9
|
Hor SL, Teoh SL, Lim WL. Plant Polyphenols as Neuroprotective Agents in Parkinson's Disease Targeting Oxidative Stress. Curr Drug Targets 2021; 21:458-476. [PMID: 31625473 DOI: 10.2174/1389450120666191017120505] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2019] [Revised: 09/26/2019] [Accepted: 09/26/2019] [Indexed: 12/15/2022]
Abstract
Parkinson's disease (PD) is the second most prevalent progressive neurodegenerative disorder characterized by the degeneration of dopaminergic neurons in the human midbrain. Various ongoing research studies are competing to understand the pathology of PD and elucidate the mechanisms underlying neurodegeneration. Current pharmacological treatments primarily focused on improving dopamine metabolism in PD patients, despite the side effects of long-term usage. In recent years, it is recognized that oxidative stress-mediated pathways lead to neurodegeneration in the brain, which is associated with the pathophysiology of PD. The importance of oxidative stress is often less emphasized when developing potential therapeutic approaches. Natural plant antioxidants have been shown to mediate the oxidative stress-induced effects in PD, which has gained considerable attention in both in vitro and in vivo studies. Yet, clinical trials on natural polyphenol compounds are limited, restricting the potential use of these compounds as an alternative treatment for PD. Therefore, this review provides an understanding of the oxidative stress-induced effects in PD by elucidating the underlying events contributing to oxidative stress and explore the potential use of polyphenols in improving the oxidative status in PD. Preclinical findings have supported the potential of polyphenols in providing neuroprotection against oxidative stress-induced toxicity in PD. However, limiting factors, such as safety and bioavailability of polyphenols, warrant further investigations so as to make them the potential target for clinical applications in the treatment and management of PD.
Collapse
Affiliation(s)
- Suet Lee Hor
- Department of Biological Sciences, School of Science and Technology, Sunway University, 47500 Selangor, Malaysia
| | - Seong Lin Teoh
- Department of Anatomy, Universiti Kebangsaan Malaysia Medical Centre, 56000 Kuala Lumpur, Malaysia
| | - Wei Ling Lim
- Department of Biological Sciences, School of Science and Technology, Sunway University, 47500 Selangor, Malaysia
| |
Collapse
|
10
|
Chang KH, Chen CM. The Role of Oxidative Stress in Parkinson's Disease. Antioxidants (Basel) 2020; 9:antiox9070597. [PMID: 32650609 PMCID: PMC7402083 DOI: 10.3390/antiox9070597] [Citation(s) in RCA: 125] [Impact Index Per Article: 31.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2020] [Revised: 07/03/2020] [Accepted: 07/06/2020] [Indexed: 12/24/2022] Open
Abstract
Parkinson’s disease (PD) is caused by progressive neurodegeneration of dopaminergic (DAergic) neurons with abnormal accumulation of α-synuclein in substantia nigra (SN). Studies have suggested the potential involvement of dopamine, iron, calcium, mitochondria and neuroinflammation in contributing to overwhelmed oxidative stress and neurodegeneration in PD. Function studies on PD-causative mutations of SNCA, PRKN, PINK1, DJ-1, LRRK2, FBXO7 and ATP13A2 further indicate the role of oxidative stress in the pathogenesis of PD. Therefore, it is reasonable that molecules involved in oxidative stress, such as DJ-1, coenzyme Q10, uric acid, 8-hydroxy-2’-deoxyguanosin, homocysteine, retinoic acid/carotenes, vitamin E, glutathione peroxidase, superoxide dismutase, xanthine oxidase and products of lipid peroxidation, could be candidate biomarkers for PD. Applications of antioxidants to modulate oxidative stress could be a strategy in treating PD. Although a number of antioxidants, such as creatine, vitamin E, coenzyme Q10, pioglitazone, melatonin and desferrioxamine, have been tested in clinical trials, none of them have demonstrated conclusive evidence to ameliorate the neurodegeneration in PD patients. Difficulties in clinical studies may be caused by the long-standing progression of neurodegeneration, lack of biomarkers for premotor stage of PD and inadequate drug delivery across blood–brain barrier. Solutions for these challenges will be warranted for future studies with novel antioxidative treatment in PD patients.
Collapse
Affiliation(s)
| | - Chiung-Mei Chen
- Correspondence: ; Tel.: +886-3-3281200 (ext. 8347); Fax: +886-3-3288849
| |
Collapse
|
11
|
Uhl GR. Dopamine compartmentalization, selective dopaminergic vulnerabilities in Parkinson's disease and therapeutic opportunities. Ann Clin Transl Neurol 2019; 6:406-415. [PMID: 30847375 PMCID: PMC6389739 DOI: 10.1002/acn3.707] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2018] [Revised: 09/29/2018] [Accepted: 11/05/2018] [Indexed: 12/13/2022] Open
Abstract
Progressive depletion of selected dopamine neurons is central to much Parkinson's disease (PD) disability. Although symptomatic treatments can ameliorate the disabilities that this neuronal depletion causes, no current strategy is documented to slow these losses. There is substantial evidence that dopamine in intracytoplasmic/extravesicular neuronal compartments can be toxic. Here, I review evidence that supports roles for dopamine compartmentalization, mediated largely by serial actions of plasma membrane SLC6A3/DAT and vesicular SLC18A2/VMAT2 transporters, in the selective patterns of dopamine neuronal loss found in PD brains. This compartmentalization hypothesis for the dopamine cell type specificity of PD lesions nominates available drugs for amelioration of damage arising from miscompartmentalized dopamine and raises cautions in using other drugs.
Collapse
Affiliation(s)
- George R. Uhl
- Neurology and Research ServicesNew Mexico VA HealthCare SystemAlbuquerqueNew Mexico87108
- Biomedical Research Institute of New MexicoAlbuquerqueNew Mexico87108
- Departments of Neurology, Neuroscience and Molecular Genetics and MicrobiologyUniversity of New MexicoAlbuquerqueNew Mexico87108
- Departments of Neurology, Neuroscience and Mental HealthJohns Hopkins Medical InstitutionsBaltimoreMaryland21287
| |
Collapse
|
12
|
Lohoff FW, Carr GV, Brookshire B, Ferraro TN, Lucki I. Deletion of the vesicular monoamine transporter 1 (vmat1/slc18a1) gene affects dopamine signaling. Brain Res 2019; 1712:151-157. [PMID: 30685272 DOI: 10.1016/j.brainres.2019.01.029] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2018] [Revised: 01/18/2019] [Accepted: 01/23/2019] [Indexed: 10/27/2022]
Abstract
The vesicular monoamine transporter is involved in presynaptic catecholamine storage and neurotransmission. Two isoforms of the transporter exist, VMAT1 and VMAT2, and both are expressed in the brain, though VMAT2 expression is more robust and has been more widely studied. In this study we investigated the role of VMAT1 KO on markers of dopaminergic function and neurotransmission, and dopamine-related behaviors. Null-mutant VMAT1 mice were studied behaviorally using the tail suspension test, elevated zero maze and locomotor activity assessments. Tissue monoamines were measured both ex vivo and by using in vivo microdialysis. Protein expression of tyrosine hydroxylase and D2 dopamine receptors was measured using western blot analysis. Results show that VMAT1 KO mice have decreased dopamine levels in the frontal cortex, increased postsynaptic D2 expression, and lower frontal cortex tyrosine hydroxylase expression compared to WT mice. VMAT1 KO mice also show an exaggerated behavioral locomotor response to acute amphetamine treatment. We conclude that dopaminergic signaling is robustly altered in the frontal cortex of VMAT1 null-mutant mice and suggest that VMAT1 may be relevant to the pathogenesis and/or treatment of psychiatric illnesses including schizophrenia and bipolar disease.
Collapse
Affiliation(s)
- Falk W Lohoff
- Section on Clinical Genomics and Experimental Therapeutics, National Institute on Alcohol Abuse and Alcoholism, National Institutes of Health, Bethesda, MD, USA.
| | - Gregory V Carr
- Lieber Institute for Brain Development, Baltimore, MD, USA; Department of Pharmacology and Molecular Sciences, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Bethany Brookshire
- Center for Neurobiology and Behavior, Department of Psychiatry, University of Pennsylvania School of Medicine Translational Research Laboratories, Philadelphia, PA, USA
| | - Thomas N Ferraro
- Department of Biomedical Sciences, Rowan University, Camden, NJ, USA
| | - Irwin Lucki
- Department of Pharmacology, Uniformed Services University, Bethesda, MD, USA
| |
Collapse
|
13
|
Pifl C, Rajput A, Reither H, Blesa J, Cavada C, Obeso JA, Rajput AH, Hornykiewicz O. Is Parkinson's disease a vesicular dopamine storage disorder? Evidence from a study in isolated synaptic vesicles of human and nonhuman primate striatum. J Neurosci 2014; 34:8210-8. [PMID: 24920625 PMCID: PMC6608236 DOI: 10.1523/jneurosci.5456-13.2014] [Citation(s) in RCA: 128] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2013] [Revised: 05/06/2014] [Accepted: 05/07/2014] [Indexed: 12/21/2022] Open
Abstract
The cause of degeneration of nigrostriatal dopamine (DA) neurons in idiopathic Parkinson's disease (PD) is still unknown. Intraneuronally, DA is largely confined to synaptic vesicles where it is protected from metabolic breakdown. In the cytoplasm, however, free DA can give rise to formation of cytotoxic free radicals. Normally, the concentration of cytoplasmic DA is kept at a minimum by continuous pumping activity of the vesicular monoamine transporter (VMAT)2. Defects in handling of cytosolic DA by VMAT2 increase levels of DA-generated oxy radicals ultimately resulting in degeneration of DAergic neurons. Here, we isolated for the first time, DA storage vesicles from the striatum of six autopsied brains of PD patients and four controls and measured several indices of vesicular DA storage mechanisms. We found that (1) vesicular uptake of DA and binding of the VMAT2-selective label [(3)H]dihydrotetrabenazine were profoundly reduced in PD by 87-90% and 71-80%, respectively; (2) after correcting for DA nerve terminal loss, DA uptake per VMAT2 transport site was significantly reduced in PD caudate and putamen by 53 and 55%, respectively; (3) the VMAT2 transport defect appeared specific for PD as it was not present in Macaca fascicularis (7 MPTP and 8 controls) with similar degree of MPTP-induced nigrostriatal neurodegeneration; and (4) DA efflux studies and measurements of acidification in the vesicular preparations suggest that the DA storage impairment was localized at the VMAT2 protein itself. We propose that this VMAT2 defect may be an early abnormality promoting mechanisms leading to nigrostriatal DA neuron death in PD.
Collapse
Affiliation(s)
- Christian Pifl
- Center for Brain Research, Medical University of Vienna, A-1090 Vienna, Austria,
| | - Alex Rajput
- Movement Disorders Program Saskatchewan, Royal University Hospital, University of Saskatchewan, Saskatoon, Saskatchewan, SK S7N OW8, Canada
| | - Harald Reither
- Center for Brain Research, Medical University of Vienna, A-1090 Vienna, Austria
| | - Javier Blesa
- Movement Disorders Group, Neurosciences Division, CIMA, and Department of Neurology and Neurosurgery, Clinica Universidad de Navarra, E31008 Pamplona, Spain, and
| | - Carmen Cavada
- Departamento de Anatomía, Histología y Neurociencia, Facultad de Medicina, Universidad Autónoma de Madrid, E28049 Madrid, Spain
| | - José A Obeso
- Movement Disorders Group, Neurosciences Division, CIMA, and Department of Neurology and Neurosurgery, Clinica Universidad de Navarra, E31008 Pamplona, Spain, and
| | - Ali H Rajput
- Movement Disorders Program Saskatchewan, Royal University Hospital, University of Saskatchewan, Saskatoon, Saskatchewan, SK S7N OW8, Canada
| | - Oleh Hornykiewicz
- Center for Brain Research, Medical University of Vienna, A-1090 Vienna, Austria
| |
Collapse
|
14
|
Taylor TN, Alter SP, Wang M, Goldstein DS, Miller GW. Reduced vesicular storage of catecholamines causes progressive degeneration in the locus ceruleus. Neuropharmacology 2014; 76 Pt A:97-105. [PMID: 24025942 PMCID: PMC4049095 DOI: 10.1016/j.neuropharm.2013.08.033] [Citation(s) in RCA: 45] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2013] [Revised: 08/23/2013] [Accepted: 08/26/2013] [Indexed: 10/26/2022]
Abstract
Parkinson's disease (PD) is the most common neurodegenerative motor disease. Pathologically, PD is characterized by Lewy body deposition and subsequent death of dopamine neurons in the substantia nigra pars compacta. PD also consistently features degeneration of the locus ceruleus, the main source of norepinephrine in the central nervous system. We have previously reported a mouse model of dopaminergic neurodegeneration based on reduced expression of the vesicular monoamine transporter (VMAT2 LO). To determine if reduced vesicular storage can also cause noradrenergic degeneration, we examined indices of damage to the catecholaminergic systems in brain and cardiac tissue of VMAT2 LO mice. At two months of age, neurochemical analyses revealed substantial reductions in striatal dopamine (94%), cortical dopamine (57%) and norepinephrine (54%), as well as cardiac norepinephrine (97%). These losses were accompanied by increased conversion of dopamine and norepinephrine to their deaminated metabolites. VMAT2 LO mice exhibited loss of noradrenergic innervation in the cortex, as determined by norepinephrine transporter immunoreactivity and (3)H-nisoxetine binding. Using unbiased stereological techniques, we observed progressive degeneration in the locus ceruleus that preceded degeneration of the substantia nigra pars compacta. In contrast, the ventral tegmental area, which is spared in human PD, remained unaffected. The coordinate loss of dopamine and norepinephrine neurons in VMAT2 LO mice parallels the pattern of neurodegeneration that occurs in human PD, and demonstrates that insufficient catecholamine storage can cause spontaneous degeneration in susceptible neurons, underscoring cytosolic catecholamine catabolism as a determinant of neuronal susceptibility in PD. This article is part of the Special Issue entitled 'The Synaptic Basis of Neurodegenerative Disorders'.
Collapse
Affiliation(s)
- Tonya N. Taylor
- Center for Neurodegenerative Disease, Emory University, Atlanta, GA
- Department of Environmental Health, Emory University, Atlanta, GA
| | - Shawn P. Alter
- Center for Neurodegenerative Disease, Emory University, Atlanta, GA
- Department of Environmental Health, Emory University, Atlanta, GA
| | - Minzheng Wang
- Center for Neurodegenerative Disease, Emory University, Atlanta, GA
- Department of Environmental Health, Emory University, Atlanta, GA
| | | | - Gary W. Miller
- Center for Neurodegenerative Disease, Emory University, Atlanta, GA
- Department of Environmental Health, Emory University, Atlanta, GA
| |
Collapse
|
15
|
Hall FS, Drgonova J, Jain S, Uhl GR. Implications of genome wide association studies for addiction: are our a priori assumptions all wrong? Pharmacol Ther 2013; 140:267-79. [PMID: 23872493 DOI: 10.1016/j.pharmthera.2013.07.006] [Citation(s) in RCA: 49] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2013] [Accepted: 07/11/2013] [Indexed: 11/24/2022]
Abstract
Substantial genetic contributions to addiction vulnerability are supported by data from twin studies, linkage studies, candidate gene association studies and, more recently, Genome Wide Association Studies (GWAS). Parallel to this work, animal studies have attempted to identify the genes that may contribute to responses to addictive drugs and addiction liability, initially focusing upon genes for the targets of the major drugs of abuse. These studies identified genes/proteins that affect responses to drugs of abuse; however, this does not necessarily mean that variation in these genes contributes to the genetic component of addiction liability. One of the major problems with initial linkage and candidate gene studies was an a priori focus on the genes thought to be involved in addiction based upon the known contributions of those proteins to drug actions, making the identification of novel genes unlikely. The GWAS approach is systematic and agnostic to such a priori assumptions. From the numerous GWAS now completed several conclusions may be drawn: (1) addiction is highly polygenic; each allelic variant contributing in a small, additive fashion to addiction vulnerability; (2) unexpected, compared to our a priori assumptions, classes of genes are most important in explaining addiction vulnerability; (3) although substantial genetic heterogeneity exists, there is substantial convergence of GWAS signals on particular genes. This review traces the history of this research; from initial transgenic mouse models based upon candidate gene and linkage studies, through the progression of GWAS for addiction and nicotine cessation, to the current human and transgenic mouse studies post-GWAS.
Collapse
Affiliation(s)
- F Scott Hall
- Molecular Neurobiology Branch, National Institute on Drug Abuse, Intramural Research Program, Baltimore, MD 21224, United States.
| | | | | | | |
Collapse
|
16
|
Ohara A, Kasahara Y, Yamamoto H, Hata H, Kobayashi H, Numachi Y, Miyoshi I, Hall FS, Uhl GR, Ikeda K, Sora I. Exclusive expression of VMAT2 in noradrenergic neurons increases viability of homozygous VMAT2 knockout mice. Biochem Biophys Res Commun 2013; 432:526-32. [PMID: 23410751 DOI: 10.1016/j.bbrc.2013.02.014] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2013] [Accepted: 02/04/2013] [Indexed: 11/26/2022]
Abstract
The vesicular monoamine transporter 2 (VMAT2) translocates monoamine neurotransmitters from the neuronal cytoplasm into synaptic vesicles. Since VMAT2-/- mice die within a few days of birth, it is difficult to analyze the detailed VMAT2 functions using these mice. In this study, we generated human VMAT2 transgenic mice that expressed VMAT2 in noradrenergic neurons with the aim to rescue the lethality of VMAT2 deletion. The expression of human VMAT2 in noradrenergic neurons extended the life of VMAT2-/- mice for up to three weeks, and these mice showed severe growth deficiency compared with VMAT2+/+ mice. These results may indicate that VMAT2 expressed in noradrenergic neurons has crucial roles in survival during the first several weeks after birth, and VMAT2 functions in other monoaminergic systems could be required for further extended survival. Although VMAT2 rescue in noradrenergic neurons did not eliminate the increased morbidity and lethality associated with VMAT2 deletion, the extension of the lifespan in VMAT2 transgenic mice will enable behavioral, pharmacological and pathophysiological studies of VMAT2 function.
Collapse
Affiliation(s)
- Arihisa Ohara
- Department of Biological Psychiatry, Tohoku University Graduate School of Medicine, Sendai, Japan
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
17
|
Dias V, Junn E, Mouradian MM. The role of oxidative stress in Parkinson's disease. JOURNAL OF PARKINSON'S DISEASE 2013; 3:461-91. [PMID: 24252804 PMCID: PMC4135313 DOI: 10.3233/jpd-130230] [Citation(s) in RCA: 1096] [Impact Index Per Article: 99.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Oxidative stress plays an important role in the degeneration of dopaminergic neurons in Parkinson's disease (PD). Disruptions in the physiologic maintenance of the redox potential in neurons interfere with several biological processes, ultimately leading to cell death. Evidence has been developed for oxidative and nitrative damage to key cellular components in the PD substantia nigra. A number of sources and mechanisms for the generation of reactive oxygen species (ROS) are recognized including the metabolism of dopamine itself, mitochondrial dysfunction, iron, neuroinflammatory cells, calcium, and aging. PD causing gene products including DJ-1, PINK1, parkin, alpha-synuclein and LRRK2 also impact in complex ways mitochondrial function leading to exacerbation of ROS generation and susceptibility to oxidative stress. Additionally, cellular homeostatic processes including the ubiquitin-proteasome system and mitophagy are impacted by oxidative stress. It is apparent that the interplay between these various mechanisms contributes to neurodegeneration in PD as a feed forward scenario where primary insults lead to oxidative stress, which damages key cellular pathogenetic proteins that in turn cause more ROS production. Animal models of PD have yielded some insights into the molecular pathways of neuronal degeneration and highlighted previously unknown mechanisms by which oxidative stress contributes to PD. However, therapeutic attempts to target the general state of oxidative stress in clinical trials have failed to demonstrate an impact on disease progression. Recent knowledge gained about the specific mechanisms related to PD gene products that modulate ROS production and the response of neurons to stress may provide targeted new approaches towards neuroprotection.
Collapse
Affiliation(s)
- Vera Dias
- Center for Neurodegenerative and Neuroimmunologic Diseases, Department of Neurology, Rutgers - Robert Wood Johnson Medical School, Piscataway, NJ, USA
| | - Eunsung Junn
- Center for Neurodegenerative and Neuroimmunologic Diseases, Department of Neurology, Rutgers - Robert Wood Johnson Medical School, Piscataway, NJ, USA
| | - M. Maral Mouradian
- Center for Neurodegenerative and Neuroimmunologic Diseases, Department of Neurology, Rutgers - Robert Wood Johnson Medical School, Piscataway, NJ, USA
| |
Collapse
|
18
|
Multani PK, Hodge R, Estévez MA, Abel T, Kung H, Alter M, Brookshire B, Lucki I, Nall AH, Talbot K, Doyle GA, Lohoff FW. VMAT1 deletion causes neuronal loss in the hippocampus and neurocognitive deficits in spatial discrimination. Neuroscience 2012. [PMID: 23201251 DOI: 10.1016/j.neuroscience.2012.11.023] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
Vesicular monoamine transporters (VMAT) are involved in presynaptic storage and release of neurotransmitters. While it was thought initially that only VMAT2 is brain expressed and VMAT1 is present only in the periphery, recent data have challenged the exclusive expression of VMAT2 in the brain. To further elucidate the role of VMAT1 brain expression and its potential role in neuropsychiatric disorders, we have investigated mice lacking VMAT1. Comparison of wildtype and knock-out (KO) mice using qPCR and immunohistochemistry documents the expression of VMAT1 in the brain. Deletion of VMAT1 leads to increased hippocampal apoptosis and reduced neurogenesis as assessed by caspase-3-labeling and 5-bromo-deoxy-uridine-labeling. Behavioral data show that mice lacking VMAT1 have neurocognitive deficits. VMAT2 expression is not altered in VMAT1 KO mice, suggesting a distinct role of VMAT1. Our data support VMAT1 brain expression and suggest that VMAT1 plays a key role in survival of hippocampal neurons and thus might contribute to neurocognitive deficits observed in neuropsychiatric disorders.
Collapse
Affiliation(s)
- P K Multani
- Center for Neurobiology and Behavior, Department of Psychiatry, University of Pennsylvania School of Medicine, Translational Research Laboratories, Philadelphia, PA, USA
| | - R Hodge
- Center for Neurobiology and Behavior, Department of Psychiatry, University of Pennsylvania School of Medicine, Translational Research Laboratories, Philadelphia, PA, USA
| | - M A Estévez
- Department of Biology, University of Pennsylvania, Philadelphia, PA, USA
| | - T Abel
- Center for Neurobiology and Behavior, Department of Psychiatry, University of Pennsylvania School of Medicine, Translational Research Laboratories, Philadelphia, PA, USA; Department of Biology, University of Pennsylvania, Philadelphia, PA, USA
| | - H Kung
- Department of Radiology, University of Pennsylvania School of Medicine, Philadelphia, PA, USA
| | - M Alter
- Center for Neurobiology and Behavior, Department of Psychiatry, University of Pennsylvania School of Medicine, Translational Research Laboratories, Philadelphia, PA, USA
| | - B Brookshire
- Center for Neurobiology and Behavior, Department of Psychiatry, University of Pennsylvania School of Medicine, Translational Research Laboratories, Philadelphia, PA, USA
| | - I Lucki
- Center for Neurobiology and Behavior, Department of Psychiatry, University of Pennsylvania School of Medicine, Translational Research Laboratories, Philadelphia, PA, USA; Department of Pharmacology, University of Pennsylvania School of Medicine, Philadelphia, PA, USA
| | - A H Nall
- Center for Neurobiology and Behavior, Department of Psychiatry, University of Pennsylvania School of Medicine, Translational Research Laboratories, Philadelphia, PA, USA
| | - K Talbot
- Center for Neurobiology and Behavior, Department of Psychiatry, University of Pennsylvania School of Medicine, Translational Research Laboratories, Philadelphia, PA, USA
| | - G A Doyle
- Center for Neurobiology and Behavior, Department of Psychiatry, University of Pennsylvania School of Medicine, Translational Research Laboratories, Philadelphia, PA, USA
| | - F W Lohoff
- Center for Neurobiology and Behavior, Department of Psychiatry, University of Pennsylvania School of Medicine, Translational Research Laboratories, Philadelphia, PA, USA.
| |
Collapse
|
19
|
Lev N, Barhum Y, Pilosof NS, Ickowicz D, Cohen HY, Melamed E, Offen D. DJ-1 protects against dopamine toxicity: implications for Parkinson's disease and aging. J Gerontol A Biol Sci Med Sci 2012; 68:215-25. [PMID: 22887838 DOI: 10.1093/gerona/gls147] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023] Open
Abstract
Parkinson's disease (PD) is a common age-related neurodegenerative disorder. Dopamine neurotoxicity, mediated through oxidative stress, is implicated in disease pathogenesis. The vesicular monoamine transporter-2 (VMAT2) transfers dopamine into synaptic vesicles preparing it for exocytotic release and preventing its cytoplasmic oxidation. DJ-1 mutations cause early-onset familial PD. Here, we show that DJ-1 protects dopaminergic neurons and controls the vesicular sequestration of dopamine by upregulating VMAT2. Overexpression of DJ-1 protected cells against dopamine toxicity, reduced oxidative stress, and increased VMAT2 expression and function. Reduced DJ-1 levels resulted in opposite effects. Dopamine vesicular sequestration and its release upon depolarization were dependent on DJ-1 levels. Transcriptional regulation of VMAT2 expression by DJ-1 was confirmed by chromatin immunoprecipitation assay. The results were corroborated in vivo using 6-hydroxydopamine hemiparkinsonian mouse model and transgenic DJ-1 knockout mice. Our experimental data point to a novel potential protective function of DJ-1, which could be used as a therapeutic tool.
Collapse
Affiliation(s)
- Nirit Lev
- Department of Neurology and Laboratory of Neuroscience, Felsenstein Medical Research Centre, Tel Aviv University, Rabin Medical Centre, Campus Beilinson, Petah-Tikva, 49100, Israel.
| | | | | | | | | | | | | |
Collapse
|
20
|
Sadasivan S, Pond BB, Pani AK, Qu C, Jiao Y, Smeyne RJ. Methylphenidate exposure induces dopamine neuron loss and activation of microglia in the basal ganglia of mice. PLoS One 2012; 7:e33693. [PMID: 22470460 PMCID: PMC3312333 DOI: 10.1371/journal.pone.0033693] [Citation(s) in RCA: 73] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2011] [Accepted: 02/20/2012] [Indexed: 12/21/2022] Open
Abstract
Background Methylphenidate (MPH) is a psychostimulant that exerts its pharmacological effects via preferential blockade of the dopamine transporter (DAT) and the norepinephrine transporter (NET), resulting in increased monoamine levels in the synapse. Clinically, methylphenidate is prescribed for the symptomatic treatment of ADHD and narcolepsy; although lately, there has been an increased incidence of its use in individuals not meeting the criteria for these disorders. MPH has also been misused as a “cognitive enhancer” and as an alternative to other psychostimulants. Here, we investigate whether chronic or acute administration of MPH in mice at either 1 mg/kg or 10 mg/kg, affects cell number and gene expression in the basal ganglia. Methodology/Principal Findings Through the use of stereological counting methods, we observed a significant reduction (∼20%) in dopamine neuron numbers in the substantia nigra pars compacta (SNpc) following chronic administration of 10 mg/kg MPH. This dosage of MPH also induced a significant increase in the number of activated microglia in the SNpc. Additionally, exposure to either 1 mg/kg or 10 mg/kg MPH increased the sensitivity of SNpc dopaminergic neurons to the parkinsonian agent 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP). Unbiased gene screening employing Affymetrix GeneChip® HT MG-430 PM revealed changes in 115 and 54 genes in the substantia nigra (SN) of mice exposed to 1 mg/kg and 10 mg/kg MPH doses, respectively. Decreases in the mRNA levels of gdnf, dat1, vmat2, and th in the substantia nigra (SN) were observed with both acute and chronic dosing of 10 mg/kg MPH. We also found an increase in mRNA levels of the pro-inflammatory genes il-6 and tnf-α in the striatum, although these were seen only at an acute dose of 10 mg/kg and not following chronic dosing. Conclusion Collectively, our results suggest that chronic MPH usage in mice at doses spanning the therapeutic range in humans, especially at prolonged higher doses, has long-term neurodegenerative consequences.
Collapse
Affiliation(s)
- Shankar Sadasivan
- Department of Developmental Neurobiology, St. Jude Children's Research Hospital, Memphis, Tennessee, United States of America
| | - Brooks B. Pond
- Department of Developmental Neurobiology, St. Jude Children's Research Hospital, Memphis, Tennessee, United States of America
- Department of Pharmaceutical Sciences, Bill Gatton College of Pharmacy, East Tennessee State University, Johnson City, Tennessee, United States of America
| | - Amar K. Pani
- Department of Developmental Neurobiology, St. Jude Children's Research Hospital, Memphis, Tennessee, United States of America
| | - Chunxu Qu
- Department of Information Sciences, St. Jude Children's Research Hospital, Memphis, Tennessee, United States of America
| | - Yun Jiao
- Department of Developmental Neurobiology, St. Jude Children's Research Hospital, Memphis, Tennessee, United States of America
| | - Richard J. Smeyne
- Department of Developmental Neurobiology, St. Jude Children's Research Hospital, Memphis, Tennessee, United States of America
- * E-mail:
| |
Collapse
|
21
|
Hart AB, de Wit H, Palmer AA. Genetic factors modulating the response to stimulant drugs in humans. Curr Top Behav Neurosci 2012; 12:537-77. [PMID: 22261702 PMCID: PMC3388157 DOI: 10.1007/7854_2011_187] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Individuals vary in their responses to stimulant drugs, and several lines of evidence suggest that the basis for this variation is at least partially genetic in origin. Association studies have examined the effects of polymorphisms in specific genes on acute and chronic responses to stimulant drugs. Several of these genetic polymorphisms are also associated with other psychiatric dimensions and disorders.This chapter examines the evidence for genetic associations between the genes that have been most carefully examined for their influence on the response to stimulant drugs.
Collapse
|
22
|
Zhou Z, Kim J, Insolera R, Peng X, Fink DJ, Mata M. Rho GTPase regulation of α-synuclein and VMAT2: implications for pathogenesis of Parkinson's disease. Mol Cell Neurosci 2011; 48:29-37. [PMID: 21699982 PMCID: PMC3163163 DOI: 10.1016/j.mcn.2011.06.002] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2011] [Revised: 05/06/2011] [Accepted: 06/01/2011] [Indexed: 12/15/2022] Open
Abstract
Accumulation of α-synuclein (Asyn) in neuronal perikarya and dystrophic neurites is characteristic of idiopathic and familial Parkinson's disease. In this study, we investigated the relationship between α-synuclein expression and neurite outgrowth-maturation using MN9D dopaminergic cells and demonstrated key features of Asyn regulation in hippocampal neurons. Neurite elongation elicited by inhibition of Rho GTPase activity with C3 transferase or by db-cAMP treatment was associated with marked reduction of α-synuclein mRNA and protein expression. Rho inhibition resulted in reduction of transcription factor SRF in the nuclear fraction and retention of MKL-1 - the SRF co-transactivator of SRE - in cytosol, indicating that these effects of Rho inhibition may be mediated though reduction of SRF-SRE transcription. Inhibition of Rho GTPase activity led to decreased nuclear localization of GATA2, a key regulator of α-synuclein promoter activity. Rho inhibition-induced neurite extension was associated with increased VMAT2 and SNARE proteins synaptophysin and synapsin I. These results indicate that in the MN9D dopaminergic cell line, α-synuclein transcription and levels of synaptic vesicle associated proteins are inversely correlated with neurite growth. We confirm that in mature hippocampal neurons inhibition of RhoA and knock down of SRF by siRNA also lead to decrease GATA2 and Asyn. The results suggest that RhoA signaling may be potential therapeutic target for the treatment of synucleinopathies.
Collapse
Affiliation(s)
- Zhigang Zhou
- Department of Neurology, University of Michigan and VA Ann Arbor Healthcare System, 5031 BSRB, 109 Zina Pitcher Place, Ann Arbor, MI, 48109
| | - Jeeyong Kim
- Department of Neurology, University of Michigan and VA Ann Arbor Healthcare System, 5031 BSRB, 109 Zina Pitcher Place, Ann Arbor, MI, 48109
| | - Ryan Insolera
- Department of Neurology, University of Michigan and VA Ann Arbor Healthcare System, 5031 BSRB, 109 Zina Pitcher Place, Ann Arbor, MI, 48109
| | - Xiangmin Peng
- Department of Neurology, University of Michigan and VA Ann Arbor Healthcare System, 5031 BSRB, 109 Zina Pitcher Place, Ann Arbor, MI, 48109
| | - David J. Fink
- Department of Neurology, University of Michigan and VA Ann Arbor Healthcare System, 5031 BSRB, 109 Zina Pitcher Place, Ann Arbor, MI, 48109
| | - Marina Mata
- Department of Neurology, University of Michigan and VA Ann Arbor Healthcare System, 5031 BSRB, 109 Zina Pitcher Place, Ann Arbor, MI, 48109
| |
Collapse
|
23
|
Eiden LE, Weihe E. VMAT2: a dynamic regulator of brain monoaminergic neuronal function interacting with drugs of abuse. Ann N Y Acad Sci 2011; 1216:86-98. [PMID: 21272013 DOI: 10.1111/j.1749-6632.2010.05906.x] [Citation(s) in RCA: 117] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
The monoaminergic neuron, in particular the dopaminergic neuron, is central to mediating the hedonic and addictive properties of drugs of abuse. The effects of amphetamine (AMPH) and cocaine (COC), for example, depend on the ability to increase dopamine in the synapse, by effects on either the plasma membrane transporter DAT or the vesicular transporter for monoamine storage, VMAT2. The potential role of DAT as a target for AMPH and COC has been reviewed extensively. Here, we present VMAT2 as a target that enables the rewarding and addictive actions of these drugs, based on imaging, neurochemical, biochemical, cell biological, genetic, and immunohistochemical evidence. The presence of VMAT2 in noradrenergic, serotoninergic, histaminergic, and potentially trace aminergic neurons invites consideration of a wider role for aminergic neurotransmission in AMPH and COC abuse and addiction.
Collapse
Affiliation(s)
- Lee E Eiden
- Section on Molecular Neuroscience, Laboratory of Cellular and Molecular Regulation, National Institute of Mental Health, National Institutes of Health, Bethesda, Maryland, USA.
| | | |
Collapse
|
24
|
Tammimäki A, Männistö PT. Effect of genetic modifications in the synaptic dopamine clearance systems on addiction-like behaviour in mice. Basic Clin Pharmacol Toxicol 2010; 108:2-8. [PMID: 21118356 DOI: 10.1111/j.1742-7843.2010.00647.x] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
During the last 15 years, genetically modified mouse lines have proved to be a valuable research tool. This review summarizes research that studied addiction-like behaviour in mice that had a targeted mutation in the genes of the synaptic dopamine removal systems, i.e. in the dopamine transporter (DAT), a vesicular monoamine transporter 2 (VMAT2) or two dopamine-metabolizing enzymes (monoamine oxidase, MAO, mainly MAO-A isoenzyme, and catechol-O-methyltransferase, COMT). Majority of the mice are knockouts but also some knock-in and knock down mouse lines are included. Most studies have explored DAT, and it has been shown to be the critical target in addiction to psychostimulants. Its role in the development of addiction-like behaviour to nicotine, opioids or ethanol is less clear. VMAT2 also seems to be linked to psychostimulant addiction. MAO-A and COMT have a minor role in addiction-like behaviour that is further complicated by a sexual dimorphism.
Collapse
Affiliation(s)
- Anne Tammimäki
- Division of Pharmacology and Toxicology, Faculty of Pharmacy, University of Helsinki, Finland.
| | | |
Collapse
|
25
|
Sora I, Li B, Igari M, Hall FS, Ikeda K. Transgenic mice in the study of drug addiction and the effects of psychostimulant drugs. Ann N Y Acad Sci 2010; 1187:218-46. [PMID: 20201856 DOI: 10.1111/j.1749-6632.2009.05276.x] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023]
Abstract
The first transgenic models used to study addiction were based upon a priori assumptions about the importance of particular genes in addiction, including the main target molecules of morphine, amphetamine, and cocaine. This consequently emphasized the importance of monoamine transporters, opioid receptors, and monoamine receptors in addiction. Although the effects of opiates were largely eliminated by mu opioid receptor gene knockout, the case for psychostimulants was much more complex. Research using transgenic models supported the idea of a polygenic basis for psychostimulant effects and has associated particular genes with different behavioral consequences of psychostimulants. Phenotypic analysis of transgenic mice, especially gene knockout mice, has been instrumental in identifying the role of specific molecular targets of addictive drugs in their actions. In this article, we summarize studies that have provided insight into the polygenic determination of drug addiction phenotypes in ways that are not possible with other methods, emphasizing research into the effects of psychostimulant drugs in gene knockouts of the monoamine transporters and monoamine receptors.
Collapse
Affiliation(s)
- Ichiro Sora
- Department of Biological Psychiatry, Tohoku University Graduate School of Medicine, Sendai, Japan.
| | | | | | | | | |
Collapse
|
26
|
Sass MB, Lorenz AN, Green RL, Coleman RA. A pragmatic approach to biochemical systems theory applied to an α-synuclein-based model of Parkinson's disease. J Neurosci Methods 2009; 178:366-77. [DOI: 10.1016/j.jneumeth.2008.12.014] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2008] [Revised: 11/24/2008] [Accepted: 12/11/2008] [Indexed: 10/21/2022]
|
27
|
Brunk I, Blex C, Sanchis-Segura C, Sternberg J, Perreau-Lenz S, Bilbao A, Hörtnagl H, Baron J, Juranek J, Laube G, Birnbaumer L, Spanagel R, Ahnert-Hilger G. Deletion of Go2alpha abolishes cocaine-induced behavioral sensitization by disturbing the striatal dopamine system. FASEB J 2008; 22:3736-46. [PMID: 18606864 DOI: 10.1096/fj.08-111245] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
The alpha-subunits of the trimeric Go class of GTPases, comprising the splice variants Go1alpha and Go2alpha, are abundantly expressed in brain and reside on both plasma membrane and synaptic vesicles. Go2alpha is involved in the vesicular storage of monoamines but its physiological relevance is still obscure. We now show that genetic depletion of Go2alpha reduces motor activity induced by dopamine-enhancing drugs like cocaine, as repeated injections of cocaine fail to provoke behavioral sensitization in Go2alpha(-/-) mice. In Go2alpha(-/-) mice, D1 receptor signaling in the striatum is attenuated due to a reduced expression of Golf alpha and Gs alpha. Following cocaine treatment, Go2alpha(-/-) mice have lower D1 and higher D2 receptor amounts compared to wild-type mice. The lack of behavioral sensitization correlates with reduced dopamine levels in the striatum and decreased expression of tyrosine hydroxylase. One reason for the neurochemical changes may be a reduced uptake of monoamines by synaptic vesicles from Go2alpha(-/-) mice as a consequence of a lowered set point for filling. We conclude that Go2alpha optimizes vesicular filling which is instrumental for normal dopamine functioning and for the development of drug-induced behavioral sensitization.
Collapse
Affiliation(s)
- Irene Brunk
- Institute for Integrative Neuroanatomy, Center for Anatomy, Charité-Universitätsmedizin Berlin, Berlin, Germany
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
28
|
Sora I, Igari M, Yamamoto H, Ikeda K. Monoamine transporter as a target molecule for psychostimulants. Nihon Yakurigaku Zasshi 2008; 130:450-4. [PMID: 18079593 DOI: 10.1254/fpj.130.450] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/22/2022]
|
29
|
A metabolomic study of brain tissues from aged mice with low expression of the vesicular monoamine transporter 2 (VMAT2) gene. Neurochem Res 2007; 33:292-300. [PMID: 18041582 DOI: 10.1007/s11064-007-9542-3] [Citation(s) in RCA: 16] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2007] [Accepted: 11/01/2007] [Indexed: 10/22/2022]
Abstract
The vesicular monoamine transporter 2 (VMAT2) sequesters monoamines into synaptic vesicles in preparation for neurotransmission. Samples of cerebellum, cortex, hippocampus, substantia nigra and striatum from VMAT2-deficient mice were compared to age-matched control mice. Multivariate statistical analyses of (1)H NMR spectral profiles separated VMAT2-deficient mice from controls for all five brain regions. Although the data show that metabolic alterations are region- and age-specific, in general, analyses indicated decreases in the concentrations of taurine and creatine/phosphocreatine and increases in glutamate and N-acetyl aspartate in VMAT2-deficient mouse brain tissues. This study demonstrates the efficacy of metabolomics as a functional genomics phenotyping tool for mouse models of neurological disorders, and indicates that mild reductions in the expression of VMAT2 affect normal brain metabolism.
Collapse
|
30
|
Gallardo TD, John GB, Shirley L, Contreras CM, Akbay EA, Haynie JM, Ward SE, Shidler MJ, Castrillon DH. Genomewide discovery and classification of candidate ovarian fertility genes in the mouse. Genetics 2007; 177:179-94. [PMID: 17660561 PMCID: PMC2013718 DOI: 10.1534/genetics.107.074823] [Citation(s) in RCA: 69] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2007] [Accepted: 07/13/2007] [Indexed: 11/18/2022] Open
Abstract
Female infertility syndromes are among the most prevalent chronic health disorders in women, but their genetic basis remains unknown because of uncertainty regarding the number and identity of ovarian factors controlling the assembly, preservation, and maturation of ovarian follicles. To systematically discover ovarian fertility genes en masse, we employed a mouse model (Foxo3) in which follicles are assembled normally but then undergo synchronous activation. We developed a microarray-based approach for the systematic discovery of tissue-specific genes and, by applying it to Foxo3 ovaries and other samples, defined a surprisingly large set of ovarian factors (n = 348, approximately 1% of the mouse genome). This set included the vast majority of known ovarian factors, 44% of which when mutated produce female sterility phenotypes, but most were novel. Comparative profiling of other tissues, including microdissected oocytes and somatic cells, revealed distinct gene classes and provided new insights into oogenesis and ovarian function, demonstrating the utility of our approach for tissue-specific gene discovery. This study will thus facilitate comprehensive analyses of follicle development, ovarian function, and female infertility.
Collapse
Affiliation(s)
- Teresa D Gallardo
- Department of Pathology and Simmons Comprehensive Cancer Center, University of Texas Southwestern Medical Center, Dallas, Texas 75390-9072, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
31
|
Fukushima S, Shen H, Hata H, Ohara A, Ohmi K, Ikeda K, Numachi Y, Kobayashi H, Hall FS, Uhl GR, Sora I. Methamphetamine-induced locomotor activity and sensitization in dopamine transporter and vesicular monoamine transporter 2 double mutant mice. Psychopharmacology (Berl) 2007; 193:55-62. [PMID: 17377774 DOI: 10.1007/s00213-007-0749-4] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/06/2006] [Accepted: 02/19/2007] [Indexed: 10/23/2022]
Abstract
RATIONALE The dopamine transporter (DAT) and the vesicular monoamine transporter 2 (VMAT2) play pivotal roles in the action of methamphetamine (MAP), including acute locomotor effects and behavioral sensitization. However, the relative impact of heterozygous DAT and VMAT2 knockouts (KOs) on the behavioral effects of MAP remains unknown. OBJECTIVES To evaluate the roles of DAT and VMAT2 in MAP-induced locomotor behavior, we examined locomotor activity and sensitization in heterozygous DAT KO (DAT+/-), heterozygous VMAT2 KO (VMAT2+/-), double heterozygous DAT/VMAT2 KO (DAT+/-VMAT2+/-), and wild-type (WT) mice. RESULTS Acute 1 mg/kg MAP injection induced significant locomotor increases in WT and VMAT2+/- mice but not in DAT+/- and DAT+/-VMAT2+/- mice. Acute 2 mg/kg MAP significantly increased locomotor activity in all genotypes. Repeated 1 mg/kg MAP injections revealed a delayed and attenuated development of sensitization in DAT+/- and DAT+/-VMAT2+/- mice compared to WT mice and delayed development in VMAT2+/- mice. In repeated 2 mg/kg MAP injections, DAT+/- and DAT+/-VMAT2+/- mice showed delayed but not attenuated development of sensitization, while there was no difference in the onset of sensitization between VMAT2+/- and WT mice. In DAT+/-VMAT2+/- mice, all of MAP-induced behavioral responses were similar to those in DAT+/- but not VMAT2+/- mice. CONCLUSIONS Heterozygous deletion of DAT attenuates the locomotor effects of MAP and may play larger role in behavioral responses to MAP compared to heterozygous deletion of VMAT2.
Collapse
Affiliation(s)
- Setsu Fukushima
- Department of Biological Psychiatry, Tohoku University Graduate School of Medicine, 1-1 Seiryo-machi, Sendai, 980-8574, Japan
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
32
|
Gutiérrez B, Rosa A, Papiol S, Arrufat FJ, Catalán R, Salgado P, Peralta V, Cuesta MJ, Fañanás L. Identification of two risk haplotypes for schizophrenia and bipolar disorder in the synaptic vesicle monoamine transporter gene (SVMT). Am J Med Genet B Neuropsychiatr Genet 2007; 144B:502-7. [PMID: 17427184 DOI: 10.1002/ajmg.b.30499] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
The synaptic vesicular monoamine transporter (SVMT) plays a key role in monoaminergic neurotransmission determining the size of neurotransmitter vesicular pools available for exocytotic release. Recently, several lines of evidence have suggested that altered functions of SVMT may be involved in the pathogenesis of certain neuropsychiatric diseases, including psychotic and mood disorders. In the present study, we tested the potential involvement of SVMT gene variants in the etiology of schizophrenia and bipolar disorder. Five different SNPs (T440G, C1368T, T2666C, A2683C, and A745G) were included in the analysis covering a region of about 35 kb along the SVMT gene. Analyses were performed in a case-control sample consisting of 88 bipolar patients, 107 subjects with schizophrenia, and 164 controls. Two risk haplotypes for both schizophrenia and bipolar disorder in SVMT gene were identified. Particularly, 2666T-2683A-745G (TAG) and 2666C-2683C-745A (CCA) combinations were significantly more frequent in both bipolar and schizophrenic patients than in controls. UNPHASED package estimated haplotype effects for all patients yielded relative risks of 4.1 (95%CI: 1.83-9.21) for TAG combination and 2.336 (95%CI: 1.28-4.26) for CCA haplotype. Conversely, 2666T-2683C-745A (TCA) and 2666C-2683A-745G (CAG) haplotypes seemed to protect against these mental disorders, since the estimated frequency in control chromosomes was 12% whilst such haplotypes were not observed in any bipolar or schizophrenic subject (P < 0.0000). Our results strongly suggest that SVMT gene or certain regions of it may constitute a genetic substrate of susceptibility for both schizophrenia and bipolar disorder.
Collapse
Affiliation(s)
- Blanca Gutiérrez
- Unitat d'Antropologia, Facultat de Biologia, Universitat de Barcelona, Barcelona, Spain.
| | | | | | | | | | | | | | | | | |
Collapse
|
33
|
Sora I, Fukushima S. [An update on animal model studies of attention-deficit/hyperactivity disorder]. Nihon Yakurigaku Zasshi 2006; 128:8-12. [PMID: 16940693 DOI: 10.1254/fpj.128.8] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/24/2022]
|
34
|
Glatt CE, Wahner AD, White DJ, Ruiz-Linares A, Ritz B. Gain-of-function haplotypes in the vesicular monoamine transporter promoter are protective for Parkinson disease in women. Hum Mol Genet 2005; 15:299-305. [PMID: 16339215 PMCID: PMC3643966 DOI: 10.1093/hmg/ddi445] [Citation(s) in RCA: 82] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
The vesicular monoamine transporter can protect against toxins that induce an acute parkinsonian syndrome. It has been hypothesized that cytoplasmic dopamine has subacute toxic effects in Parkinson Disease (PD) leading to neuronal death and clinical symptoms. Regulatory polymorphisms in the brain form of the vesicular monoamine transporter (VMAT2) which affect its quantitative expression might therefore serve as genetic risk factors for PD. We have screened the promoter region of the gene for VMAT2 (SLC18A2) and identified several novel polymorphisms that form discrete haplotypes. We have tested the common halpotypes in SLC18A2 for functional effects in reporter gene assays and found that there are several gain-of-function haplotypes that display significantly increased transcriptional activity from the reference element. These gain-of-function haplotypes were tested for association with PD and found to confer a protective effect that was selective for females. This finding is consistent with the prediction that increased sequestration of dopamine in secretory vesicles by VMAT2 is protective for PD.
Collapse
Affiliation(s)
- Charles E Glatt
- Department of Psychiatry, Weill Medical College of Cornell University, New York, NY 10028, USA.
| | | | | | | | | |
Collapse
|
35
|
Schwab SG, Franke PE, Hoefgen B, Guttenthaler V, Lichtermann D, Trixler M, Knapp M, Maier W, Wildenauer DB. Association of DNA polymorphisms in the synaptic vesicular amine transporter gene (SLC18A2) with alcohol and nicotine dependence. Neuropsychopharmacology 2005; 30:2263-8. [PMID: 15988470 DOI: 10.1038/sj.npp.1300809] [Citation(s) in RCA: 33] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
The brain synaptic vesicular amine transporter SLCA18A2 is a key component for the uptake of monoamines like dopamine or serotonin into vesicles. We have analyzed seven DNA polymorphisms located in the genomic region of SLC18A2 for association with alcohol- and nicotine dependence, using a family-based design. Our sample comprised 131 families with alcohol-dependent offspring and 96 families with at least one nicotine-dependent offspring. For the alcohol-dependent sample, we found statistical significant association for two single markers (rs363387, P=0.03; rs363333, P=0.0066) as well as for several haplotypes (minimal P=0.0038). When the sample with alcohol dependence was stratified according to gender, we observed increased association for the male subgroup (rs363387, P=0.0011). None of the markers showed association in the sample of families with nicotine dependence. However, analysis of a combined sample of alcohol and nicotine-dependent families resulted in single markers as well as several haplotypes showing statistical significant association with substance dependence (minimal P=0.0044). We conclude that DNA polymorphisms located in SLC18A2 might contribute to the development of substance dependence.
Collapse
Affiliation(s)
- Sibylle G Schwab
- School of Psychiatry and Clinical Neurosciences, University of Western Australia, Perth, WA, Australia.
| | | | | | | | | | | | | | | | | |
Collapse
|
36
|
Caudle WM, Richardson JR, Wang M, Miller GW. Perinatal heptachlor exposure increases expression of presynaptic dopaminergic markers in mouse striatum. Neurotoxicology 2005; 26:721-728. [PMID: 16112329 PMCID: PMC4755341 DOI: 10.1016/j.neuro.2004.09.003] [Citation(s) in RCA: 49] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2004] [Accepted: 09/21/2004] [Indexed: 11/29/2022]
Abstract
Although banned in the 1970s, significant levels of the organochlorine pesticide heptachlor are still present in the environment raising concern over potential human exposure. In particular, organochlorine pesticides have been linked to an increased risk of Parkinson's disease. Studies from our laboratory and others have demonstrated that exposure of laboratory animals to heptachlor alters the levels and function of the dopamine transporter (DAT), an integral component of dopaminergic neurotransmission and a gateway for the dopaminergic neurotoxin MPTP. In this study, we examined the effects of developmental exposure to heptachlor on DAT, and other key components of the dopaminergic system, including the vesicular monoamine transporter 2 (VMAT2), tyrosine hydroxylase (TH), and aromatic amino acid decarboxylase (AADC). Female C57BL/6J mice received 0 or 3mg/kg heptachlor in peanut butter every 3 days for 2 weeks prior to breeding and throughout gestation and lactation until the offspring were weaned on postnatal day (PND) 21. On postnatal day 28, DAT, VMAT2, and TH levels were increased by 100, 70, and 30%, respectively, with no change in AADC levels or total dopamine levels. The ratio of DAT:VMAT2 was increased 29%. Since an increase in the DAT:VMAT2 ratio appears to predict susceptibility of brain regions to Parkinson's disease (PD) and results in increased toxicity of MPTP, these results suggest that alterations of the dopaminergic system by developmental heptachlor exposure may increase the susceptibility of dopamine neurons to toxic insult.
Collapse
Affiliation(s)
- W. Michael Caudle
- Center for Neurodegenerative Disease, Emory University, Whitehead Biomedical Research Building 505, 615 Michael Street, Atlanta, GA 30322, USA
- Department of Environmental and Occupational Health, Rollins School of Public Health, Emory University, Atlanta, GA 30322, USA
| | - Jason R. Richardson
- Center for Neurodegenerative Disease, Emory University, Whitehead Biomedical Research Building 505, 615 Michael Street, Atlanta, GA 30322, USA
- Department of Environmental and Occupational Health, Rollins School of Public Health, Emory University, Atlanta, GA 30322, USA
| | - Minzheng Wang
- Center for Neurodegenerative Disease, Emory University, Whitehead Biomedical Research Building 505, 615 Michael Street, Atlanta, GA 30322, USA
- Department of Environmental and Occupational Health, Rollins School of Public Health, Emory University, Atlanta, GA 30322, USA
| | - Gary W. Miller
- Center for Neurodegenerative Disease, Emory University, Whitehead Biomedical Research Building 505, 615 Michael Street, Atlanta, GA 30322, USA
- Department of Environmental and Occupational Health, Rollins School of Public Health, Emory University, Atlanta, GA 30322, USA
- Corresponding author. Fax: +1 404 727 3728. (G.W. Miller)
| |
Collapse
|
37
|
Anlauf M, Schäfer MKH, Schwark T, von Wurmb-Schwark N, Brand V, Sipos B, Horny HP, Parwaresch R, Hartschuh W, Eiden LE, Klöppel G, Weihe E. Vesicular monoamine transporter 2 (VMAT2) expression in hematopoietic cells and in patients with systemic mastocytosis. J Histochem Cytochem 2005; 54:201-13. [PMID: 16116033 DOI: 10.1369/jhc.5a6739.2005] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Uptake of monoamines into secretory granules is mediated by the vesicular monoamine transporters VMAT1 and VMAT2. In this study, we analyzed their expression in inflammatory and hematopoietic cells and in patients suffering from systemic mastocytosis (SM) and chronic myelogenous leukemia (CML). Normal human and monkey tissue specimens and tissues from patients suffering from SM and CML were analyzed by means of immunohistochemistry, radioactive in situ hybridization, real time RT-PCR, double fluorescence confocal laser scanning microscopy, and immunoelectron microscopy. In normal tissue specimens, VMAT2, but not VMAT1, was expressed in mast cells, megakaryocytes, thrombocytes, basophil granulocytes, and cutaneous Langerhans cells. Further hematopoietic and lymphoid cells showed no expression of VMATs. VMAT2 was expressed in all types of SM, as indicated by coexpression with the mast cell marker tryptase. In CML, VMAT2 expression was retained in neoplastic megakaryocytes and basophil granulocytes. In conclusion, the identification of VMAT2 in mast cells, megakaryocytes, thrombocytes, basophil granulocytes, and cutaneous Langerhans cells provides evidence that these cells possess molecular mechanisms for monoamine storage and handling. VMAT2 identifies normal and neoplastic mast cells, megakaryocytes, and basophil granulocytes and may therefore become a valuable tool for the diagnosis of mastocytosis and malignant systemic diseases involving megakaryocytes and basophil granulocytes.
Collapse
MESH Headings
- Animals
- Basophils/metabolism
- Biomarkers, Tumor/biosynthesis
- Blood Platelets/metabolism
- Bone Marrow Cells/metabolism
- Hematopoiesis
- Humans
- Immunohistochemistry
- In Situ Hybridization
- Langerhans Cells/metabolism
- Leukemia, Myelogenous, Chronic, BCR-ABL Positive/blood
- Leukemia, Myelogenous, Chronic, BCR-ABL Positive/metabolism
- Leukemia, Myelogenous, Chronic, BCR-ABL Positive/pathology
- Macaca mulatta
- Mast Cells/metabolism
- Mastocytosis, Systemic/blood
- Mastocytosis, Systemic/metabolism
- Mastocytosis, Systemic/pathology
- Megakaryocytes/metabolism
- Microscopy, Confocal
- Microscopy, Immunoelectron
- Organ Specificity
- RNA, Messenger/biosynthesis
- Reverse Transcriptase Polymerase Chain Reaction
- Vesicular Monoamine Transport Proteins/biosynthesis
- Vesicular Monoamine Transport Proteins/genetics
Collapse
Affiliation(s)
- Martin Anlauf
- Department of Pathology, University of Kiel, Michaelisstr. 11, 24105 Kiel, Germany.
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
38
|
Chen CXQ, Huang SY, Zhang L, Liu YJ. Synaptophysin enhances the neuroprotection of VMAT2 in MPP+-induced toxicity in MN9D cells. Neurobiol Dis 2005; 19:419-26. [PMID: 16023584 DOI: 10.1016/j.nbd.2005.01.014] [Citation(s) in RCA: 30] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2004] [Revised: 01/10/2005] [Accepted: 01/20/2005] [Indexed: 10/25/2022] Open
Abstract
The use of the potent neurotoxin MPTP in producing a model for Parkinson's disease (PD) has allowed us to dissect the cellular processes responsible for both selective neuronal vulnerability and neuroprotection in idiopathic PD. It has been suggested that vesicular monoamine transporters (VMATs) play a critical neuroprotective role in MPP+ toxicity. However, little is known about how this detoxificative sequestration in dopaminergic (DAergic) neurons is regulated at the molecular and cellular levels. Using the DAergic cell line MN9D as an in vitro model, we found that overexpression of VMAT2 (a neuronal isoform of VMATs) protects the transformants from MPP+-induced toxicity, consistent with the previous work on fibroblastic CHO cells. We further found that the MN9D cells displayed lower expression levels of secretory vesicle proteins such as synaptophysin. Overexpression of synaptophysin in MN9D cells can significantly increase the resistance of the transformants to MPP+ toxicity. The co-expression of VMAT2 and synaptophysin has shown synergistic protection for the transformants, suggesting a role of synaptophysin in the biogenesis of secretory vesicles and in influencing the targeting of VMAT2 to these vesicles. Our work indicates that both the expression level of VMAT2 and capacity of vesicular packaging of DA are important in protecting DAergic cells from MPP+ toxicity.
Collapse
Affiliation(s)
- Carol X-Q Chen
- Department of Neurology, University of Pittsburgh School of Medicine, W958 Biomedical Science Tower, 200 Lothrop Street, Pittsburgh, PA 15213, USA
| | | | | | | |
Collapse
|
39
|
Kariya S, Hirano M, Takahashi N, Furiya Y, Ueno S. Lack of association between polymorphic microsatellites of the VMAT2 gene and Parkinson's disease in Japan. J Neurol Sci 2005; 232:91-4. [PMID: 15850588 DOI: 10.1016/j.jns.2005.02.005] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2004] [Revised: 01/03/2005] [Accepted: 02/08/2005] [Indexed: 10/25/2022]
Abstract
The etiology of Parkinson's disease (PD) remains unclear; however, generation of reactive oxygen species during oxidation of dopamine (DA) could be one of the factors leading to selective loss of nigral dopaminergic neurons in PD. Vesicular monoamine transporter type 2 (VMAT2) proteins in nerve terminals uptake and partition DA from neuronal cytoplasm into synaptic vesicles. Therefore, alterations of VMAT2 function may cause cytoplasmic accumulation of free DA, toxic to dopaminergic neurons. Upstream of a putative promoter region of the VMAT2 gene, there exist polymorphic sequences consisting of two microsatellites, (CA)n and (GA)n. We performed a case-control study of this polymorphic region to determine whether the VMAT2 gene is related to PD. We found six genotypes; however, there was no significant difference in the allele frequencies between patients with PD and control subjects. Our data suggest that the polymorphic region of the VMAT2 gene studied here is not closely related to PD.
Collapse
Affiliation(s)
- Shingo Kariya
- Department of Neurology, Nara Medical University, Kashihara, Nara 634-8522, Japan.
| | | | | | | | | |
Collapse
|
40
|
Miller GM, Verrico CD, Jassen A, Konar M, Yang H, Panas H, Bahn M, Johnson R, Madras BK. Primate trace amine receptor 1 modulation by the dopamine transporter. J Pharmacol Exp Ther 2005; 313:983-94. [PMID: 15764732 DOI: 10.1124/jpet.105.084459] [Citation(s) in RCA: 91] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023] Open
Abstract
Recently identified trace amine receptors are potential direct targets for drugs of abuse, including amphetamine and 3,4-methylenedioxymethamphetamine (MDMA). We cloned full-length rhesus monkey trace amine receptor 1 (rhTA(1)) that was 96% homologous to human TA(1). The trace amines tyramine and beta-phenylethylamine (PEA) and the monoamine transporter substrates (+/-)-amphetamine and (+/-)-MDMA stimulated cAMP accumulation in rhTA(1)-expressing cell lines, as measured by a cAMP response element-luciferase assay. Cocaine did not stimulate cAMP accumulation in rhTA(1) cells, but it blocked [(3)H]PEA transport mediated by the dopamine transporter. Cotransfection with the human dopamine transporter enhanced PEA-, amphetamine-, and MDMA-mediated rhTA(1) receptor activation, but it diminished tyramine activation of rhTA(1). Because TA(1) (EGFP-rhTA(1) chimera) was largely intracellular, conceivably the dopamine transporter can facilitate access of specific agonists to intracellular TA(1). rhTA(1) mRNA expression was detected in rhesus monkey substantia nigra, implying that TA(1) may be colocalized with the dopamine transporter in dopamine neurons. In summary, primate TA(1) receptors are direct targets of trace amines, amphetamine, and MDMA. These receptors could also be indirect targets of amphetamine, MDMA, and cocaine through modification of monoamine transporter function. Conceivably, rhTA(1) receptors may be located on pre- or postsynaptic membranes. Interference with the carrier function of monoamine transporters with a consequent rise of extracellular levels of trace amines could activate these receptors. The cloning of a highly homologous TA(1) from rhesus monkey and demonstration that rhTA(1) receptors are activated by drugs of abuse, indicate that nonhuman primates may serve to model physiological and pharmacological TA(1)-mediated responses in humans.
Collapse
Affiliation(s)
- Gregory M Miller
- Division of Neurochemistry, New England Primate Research Center, Harvard Medical School, Southborough, MA 01772, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
41
|
Ide S, Minami M, Satoh M, Sora I, Ikeda K. [Distinct mechanisms underlying pleasure and analgesia]. Nihon Yakurigaku Zasshi 2005; 125:11-5. [PMID: 15738616 DOI: 10.1254/fpj.125.11] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/25/2022]
|
42
|
Burman J, Tran CH, Glatt C, Freimer NB, Edwards RH. The effect of rare human sequence variants on the function of vesicular monoamine transporter 2. ACTA ACUST UNITED AC 2005; 14:587-94. [PMID: 15475732 DOI: 10.1097/00008571-200409000-00003] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
The extent to which genetic variation in a population contributes to phenotypic variation depends on the frequency of sequence polymorphisms and the effect of these polymorphisms on function. The frequency of polymorphisms might also reflect the severity of their effects on function. We therefore examined the effect of very rare single nucleotide polymorphisms (SNPs) on the activity of the vesicular monoamine transporter 2 (VMAT2, SLC18A2), a gene implicated in neuropsychiatric disease. Of the two rare SNPs identified in an ethnically diverse population, neither eliminates transport, but one that involves replacement of a highly conserved residue with a very similar amino acid impairs substrate recognition. This variant, and another affecting an unconserved residue, also affect inhibition by the clinically used drug reserpine. Because VMAT2 influences a form of toxicity similar to Parkinson's disease, we extended the analysis to two SNPs identified in a population with Parkinson's disease. These two SNPs have no detectable effect on most aspects of VMAT2 function, but one that affects a highly conserved residue may increase sensitivity to the inhibitor tetrabenazine. The results illustrate the relationship between conservation of the affected residue, the nature of the substitution and effects on substrate versus inhibitor interaction.
Collapse
Affiliation(s)
- Jonathon Burman
- Department of Neurology, UCSF School of Medicine, San Francisco, CA 94143-2140, USA
| | | | | | | | | |
Collapse
|
43
|
Liu HC, Lin SK, Liu SK, Chen SL, Hu CJ, Chang JG, Leu SJ. DAT polymorphism and diverse clinical manifestations in methamphetamine abusers. Psychiatr Genet 2004; 14:33-7. [PMID: 15091313 DOI: 10.1097/00041444-200403000-00005] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
The clinical outcome for methamphetamine (MAP) abusers is variable. MAP exerts its biological activity through rapid conversion to amphetamine (AP) and MAP itself. The dopamine transporter (DAT) is the main modulator of MAP/AP-induced dopamine release and dopamine neurotoxicity, and is also the major regulator of dopamine level in the brain. We tested for an association between a DAT-gene polymorphism and clinical variations in MAP abusers. A total of 146 MAP abusers were enrolled in the study and classified into three clinically distinct groups: MAP dependence (n = 30), MAP psychosis (n = 88) and chronic MAP psychosis (n = 28). Patients with schizophrenia (n = 79) and healthy controls (n = 72) were also recruited for the study. The 40 base pair variable number tandem repeat polymorphism in the 3'-untranslated region of the DAT was the focus of the investigation. The subjects were all Chinese residents of Taiwan. The respective allelic frequencies for DAT repeats 11, 10 and 9 were 0.067, 0.833 and 0.083 for the MAP-dependence group, 0.006, 0.864 and 0.119 for the MAP psychosis group, 0.018, 0.893 and 0.089 for the chronic MAP psychosis group, 0.019, 0.911 and 0.07 for the schizophrenic controls, and 0.021, 0.889 and 0.083 for the healthy controls. No significant associations were demonstrated between this DAT polymorphism in genotype and allele frequency and the clinical outcome of MAP abusers. The biological relevance of the variable number tandem repeat polymorphism in the 3'-untranslated region of DAT in MAP abusers was not demonstrated in this study.
Collapse
Affiliation(s)
- Hsing-Cheng Liu
- Department of Addiction Science and Molecular Psychiatry Laboratory, Taipei City Psychiatric Center, Taipei, Taiwan
| | | | | | | | | | | | | |
Collapse
|
44
|
Usera PC, Vincent S, Robertson D. Human phenotypes and animal knockout models of genetic autonomic disorders. J Biomed Sci 2004; 11:4-10. [PMID: 14730204 DOI: 10.1007/bf02256543] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2003] [Accepted: 08/04/2003] [Indexed: 11/28/2022] Open
Abstract
Norepinephrine (NE) is a crucial neurotransmitter involved in autonomic regulation of blood pressure. Dopamine beta-hydroxylase (DBH), the norepinephrine transporter (NET), and the vesicular monoamine transporter subtype 2 catalyze intracellular NE biosynthesis, NE reuptake from the synapse, and vesicular transport, respectively. Genetic disorders in humans have been identified that render DBH, and the NET dysfunctional and result in cardiovascular and neurological abnormalities. Vesicular monoamine transporter subtype 2 (VMAT2) activity protects against neurotoxins, and reduced VMAT2 expression is implicated in drug addiction. Further investigation of the consequences of these genetic abnormalities has been achieved by the construction of mice strains deficient in the genes encoding DBH, NET, and VMAT2.
Collapse
Affiliation(s)
- Phillip C Usera
- Autonomic Dysfunction Center, Department of Clinical Pharmacology, Vanderbilt University School of Medicine, AA-3228 Medical Center North, 1161 21st Avenue South, Nashville, TN 37232, USA
| | | | | |
Collapse
|
45
|
Shen HW, Hagino Y, Kobayashi H, Shinohara-Tanaka K, Ikeda K, Yamamoto H, Yamamoto T, Lesch KP, Murphy DL, Hall FS, Uhl GR, Sora I. Regional differences in extracellular dopamine and serotonin assessed by in vivo microdialysis in mice lacking dopamine and/or serotonin transporters. Neuropsychopharmacology 2004; 29:1790-9. [PMID: 15226739 DOI: 10.1038/sj.npp.1300476] [Citation(s) in RCA: 161] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Cocaine conditioned place preference (CPP) is intact in dopamine transporter (DAT) knockout (KO) mice and enhanced in serotonin transporter (SERT) KO mice. However, cocaine CPP is eliminated in double-KO mice with no DAT and either no or one SERT gene copy. To help determine mechanisms underlying these effects, we now report examination of baselines and drug-induced changes of extracellular dopamine (DAex) and serotonin (5-HT(ex)) levels in microdialysates from nucleus accumbens (NAc), caudate putamen (CPu), and prefrontal cortex (PFc) of wild-type, homozygous DAT- or SERT-KO and heterozygous or homozygous DAT/SERT double-KO mice, which are differentially rewarded by cocaine. Cocaine fails to increase DAex in NAc of DAT-KO mice. By contrast, systemic cocaine enhances DAex in both CPu and PFc of DAT-KO mice though local cocaine fails to affect DAex in CPu. Adding SERT to DAT deletion attenuates the cocaine-induced DAex increases found in CPu, but not those found in PFc. The selective SERT blocker fluoxetine increases DAex in CPu of DAT-KO mice, while cocaine and the selective DAT blocker GBR12909 increase 5-HT(ex) in CPu of SERT-KO mice. These data provide evidence that (a) cocaine increases DAex in PFc independently of DAT and that (b), in the absence of SERT, CPu levels of 5-HT(ex) can be increased by blocking DAT. Cocaine-induced alterations in CPu DA levels in DAT-, SERT-, and DAT/SERT double-KO mice appear to provide better correlations with cocaine CPP than cocaine-induced DA level alterations in NAc or PFc.
Collapse
Affiliation(s)
- Hao-Wei Shen
- Department of Molecular Psychiatry, Tokyo Institute of Psychiatry, Japan
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
46
|
Sidhu A, Wersinger C, Vernier P. Does alpha-synuclein modulate dopaminergic synaptic content and tone at the synapse? FASEB J 2004; 18:637-47. [PMID: 15054086 DOI: 10.1096/fj.03-1112rev] [Citation(s) in RCA: 127] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
alpha-Synuclein is a key component of the pathological process of neurodegeneration in Parkinson's disease. Although its contributions to normal physiological conditions remain elusive, converging observations suggest that a primary function of this protein in dopaminergic neurons may be the regulation of dopamine content and synaptic tone at the synapse. We review here cumulative evidence that demonstrates the participation of alpha-synuclein in the life cycle of dopamine from its synthesis, storage, release, and reuptake. The regulatory role of alpha-synuclein on dopamine metabolism is assessed by discussing the experimental evidence supporting each of these observations in the healthy physiological maintenance of dopaminergic neurons, as well as showing how disruption of these events can initiate the observed neurotoxicity of alpha-synuclein and the genesis of the degenerative processes associated with Parkinson's disease.
Collapse
Affiliation(s)
- Anita Sidhu
- Department of Pediatrics, Georgetown University, Washington, DC, USA.
| | | | | |
Collapse
|
47
|
Eiden LE, Schäfer MKH, Weihe E, Schütz B. The vesicular amine transporter family (SLC18): amine/proton antiporters required for vesicular accumulation and regulated exocytotic secretion of monoamines and acetylcholine. Pflugers Arch 2004; 447:636-40. [PMID: 12827358 DOI: 10.1007/s00424-003-1100-5] [Citation(s) in RCA: 129] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2003] [Revised: 04/28/2003] [Accepted: 04/28/2003] [Indexed: 12/25/2022]
Abstract
The vesicular amine transporters (VATs) are expressed as integral proteins of the lipid bilayer membrane of secretory vesicles in neuronal and endocrine cells. Their function is to allow the transport of acetylcholine (by the vesicular acetylcholine transporter VAChT; SLC18A3) and biogenic amines (by the vesicular monoamine transporters VMAT1 and VMAT2; SLC18A1 and SLC18A2) into secretory vesicles, which then discharge them into the extracellular space by exocytosis. Transport of positively charged amines by members of the SLC18 family in all cases utilizes an electrochemical gradient across the vesicular membrane established by proton pumping into the vesicle via a vacuolar ATPase; the amine is accumulated in the vesicle at the expense of the proton gradient, at a ratio of one translocated amine per two translocated protons. The members of the SLC18 family have become important histochemical markers for chemical coding in neuroendocrine tissues and cells. The structural basis of their remarkable ability to transport positively charged amines against a very large concentration gradient, as well as potential disease association with impaired transporter function and expression, are under intense investigation.
Collapse
Affiliation(s)
- Lee E Eiden
- Section on Molecular Neuroscience, Laboratory of Cellular and Molecular Regulation, National Institute of Mental Health, Building 36, Room 2A-11, 9000 Rockville Pike, Bethesda, MD 20892, USA.
| | | | | | | |
Collapse
|
48
|
Hall FS, Sora I, Uhl GR. Sex-dependent modulation of ethanol consumption in vesicular monoamine transporter 2 (VMAT2) and dopamine transporter (DAT) knockout mice. Neuropsychopharmacology 2003; 28:620-8. [PMID: 12655306 DOI: 10.1038/sj.npp.1300070] [Citation(s) in RCA: 42] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Several lines of evidence suggest that monoaminergic systems, especially dopaminergic and serotoninergic systems, modulate ethanol consumption. Humans display significant differences in expression of the vesicular and plasma membrane monoamine transporters important for monoaminergic functions, including the vesicular monoamine transporter (VMAT2, SLC18A2) and dopamine transporter (DAT, SLC6A3). In addition, many ethanol effects differ by sex in both humans and animal models. Therefore, ethanol consumption and preference were compared in male and female wild-type mice, and knockout (KO) mice with deletions of genes for DAT and VMAT2. Voluntary ethanol (2-32% v/v) and water consumption were compared in two-bottle preference tests in wild-type (+/+) vs heterozygous VMAT2 KO mice (+/-) and in wild-type (+/+) vs heterozygous (+/-) or homozygous (-/-) DAT KO mice. Deletions of either the DAT or VMAT2 genes increased ethanol consumption in male KO mice, although these effects were highly dependent on ethanol concentration, while female DAT KO mice had higher ethanol preferences. Thus, lifetime reductions in the expression of either DAT or VMAT2 increase ethanol consumption, dependent on sex.
Collapse
Affiliation(s)
- F Scott Hall
- Molecular Neurobiology Branch, NIDA-IRP, NIH, DHHS, Baltimore, MD, USA
| | | | | |
Collapse
|
49
|
Abstract
Recent findings in molecular research suggest that the outcome of cardiovascular surgery is at least partly determined by the individual patient's genetic predisposition to react to surgical trauma and extracorporal circulation. The activation of cellular as well as humoral cascades occurs in the perioperative period, and influences the extent of pro- and anticoagulation and pro- and anti-inflammation. These events contribute to the incidence and severity of perioperative ischaemia or organ dysfunction, and thus determine adverse outcomes in patients undergoing cardiac surgery. Candidate genes that are possibly involved in the development of adverse outcomes not only consist of genes relevant to the field of coagulation and inflammation, but also genes functioning in lipid metabolism, ion channels, membrane integrity and others. Genomic variations may prove to serve as future diagnostic tools for the risk stratification of patients undergoing cardiovascular surgery.
Collapse
Affiliation(s)
- Frank Stüber
- Klinik und Poliklinik für Anästhesiologie und Spezielle Intensivmedizin, Universitätsklinkum Bonn, Rheinische Friedrich-Wilhelms-Universität Bonn, Bonn, Germany.
| | | |
Collapse
|
50
|
Eiden LE. The vesicular neurotransmitter transporters: current perspectives and future prospects. FASEB J 2000; 14:2396-400. [PMID: 11099457 DOI: 10.1096/fj.00-0817rev] [Citation(s) in RCA: 63] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Affiliation(s)
- L E Eiden
- Section on Molecular Neuroscience, Laboratory of Cellular and Molecular Regulation, National Institute of Mental Health, Bethesda, Maryland 20892, USA.
| |
Collapse
|