1
|
Kambey PA, Wu J, Liu W, Su M, Buberwa W, Tang C. Targeting serum response factor (SRF) deactivates ΔFosB and mitigates Levodopa-induced dyskinesia in a mouse model of Parkinson's disease. Gene Ther 2024; 31:614-624. [PMID: 39384937 DOI: 10.1038/s41434-024-00492-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2023] [Revised: 09/18/2024] [Accepted: 09/26/2024] [Indexed: 10/11/2024]
Abstract
L-3,4-dihydroxyphenylalanine (L-DOPA) is currently the preferred treatment for Parkinson's Disease (PD) and is considered the gold standard. However, prolonged use of L-DOPA in patients can result in involuntary movements known as Levodopa-induced dyskinesia (LID), which includes uncontrollable dystonia affecting the trunk, limbs, and face. The role of ΔFosB protein, a truncated splice variant of the FosB gene, in LID has been acknowledged, but its underlying mechanism has remained elusive. Here, using a mouse model of Parkinson's disease treated with chronic levodopa we demonstrate that serum response factor (SRF) binds to the FosB promoter, thereby activating FosB expression and levodopa induced-dyskinetic movements. Western blot analysis demonstrates a significant increase in SRF expression in the dyskinetic group compared to the control group. Knocking down SRF significantly reduced abnormal involuntary movements (AIMS) and ΔFosB expression compared to the control. Conversely, overexpression of SRF led to an increase in ΔFosB expression and worsened levodopa-induced dyskinesia. To shed light on the regulatory role of the Akt signaling pathway in this phenomenon, we administered the Akt agonist SC79 to PD mouse models via intraperitoneal injection, followed by L-DOPA administration. The expression of SRF, ΔFosB, and phosphorylated Akt (p-Akt) significantly increased in this group compared to the group receiving normal saline to signify that these happen through Akt signaling pathway. Collectively, our findings identify a promising therapeutic target for addressing levodopa-induced dyskinesia.
Collapse
Affiliation(s)
- Piniel Alphayo Kambey
- Xuzhou Key Laboratory of Neurobiology, Department of Neurobiology and Anatomy, Xuzhou Medical University, 209 Tongshan Road, Xuzhou, Jiangsu, 221004, China.
- Key Laboratory of Regenerative Biology, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, 190 Kaiyuan Avenue, Guangzhou Science Park, Huangpu District, Guangzhou, China.
| | - Jiao Wu
- Xuzhou Key Laboratory of Neurobiology, Department of Neurobiology and Anatomy, Xuzhou Medical University, 209 Tongshan Road, Xuzhou, Jiangsu, 221004, China
| | - WenYa Liu
- Xuzhou Key Laboratory of Neurobiology, Department of Neurobiology and Anatomy, Xuzhou Medical University, 209 Tongshan Road, Xuzhou, Jiangsu, 221004, China
| | - Mingyu Su
- Xuzhou Key Laboratory of Neurobiology, Department of Neurobiology and Anatomy, Xuzhou Medical University, 209 Tongshan Road, Xuzhou, Jiangsu, 221004, China
| | - Wokuheleza Buberwa
- Department of Neurology, The second affiliated hospital of Xi'an Jiaotong University, 710049, Xi'an, China
| | - Chuanxi Tang
- Xuzhou Key Laboratory of Neurobiology, Department of Neurobiology and Anatomy, Xuzhou Medical University, 209 Tongshan Road, Xuzhou, Jiangsu, 221004, China.
| |
Collapse
|
2
|
Mantas I, Flais I, Masarapu Y, Ionescu T, Frapard S, Jung F, Le Merre P, Saarinen M, Tiklova K, Salmani BY, Gillberg L, Zhang X, Chergui K, Carlén M, Giacomello S, Hengerer B, Perlmann T, Svenningsson P. Claustrum and dorsal endopiriform cortex complex cell-identity is determined by Nurr1 and regulates hallucinogenic-like states in mice. Nat Commun 2024; 15:8176. [PMID: 39289358 PMCID: PMC11408527 DOI: 10.1038/s41467-024-52429-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2023] [Accepted: 09/04/2024] [Indexed: 09/19/2024] Open
Abstract
The Claustrum/dorsal endopiriform cortex complex (CLA) is an enigmatic brain region with extensive glutamatergic projections to multiple cortical areas. The transcription factor Nurr1 is highly expressed in the CLA, but its role in this region is not understood. By using conditional gene-targeted mice, we show that Nurr1 is a crucial regulator of CLA neuron identity. Although CLA neurons remain intact in the absence of Nurr1, the distinctive gene expression pattern in the CLA is abolished. CLA has been hypothesized to control hallucinations, but little is known of how the CLA responds to hallucinogens. After the deletion of Nurr1 in the CLA, both hallucinogen receptor expression and signaling are lost. Furthermore, functional ultrasound and Neuropixel electrophysiological recordings revealed that the hallucinogenic-receptor agonists' effects on functional connectivity between prefrontal and sensorimotor cortices are altered in Nurr1-ablated mice. Our findings suggest that Nurr1-targeted strategies provide additional avenues for functional studies of the CLA.
Collapse
Affiliation(s)
- Ioannis Mantas
- Department of Clinical Neuroscience, Karolinska Institutet, Stockholm, Sweden.
- Department of Neuroscience, Karolinska Institutet, Stockholm, Sweden.
| | - Ivana Flais
- Department of Clinical Neuroscience, Karolinska Institutet, Stockholm, Sweden
- CNSDR, Boehringer Ingelheim Pharma GmbH & Co. KG, Biberach, Germany
- Department of Neuroimaging King's College London, London, UK
| | - Yuvarani Masarapu
- Science for Life Laboratory, Department of Gene Technology, KTH Royal Institute of Technology, Stockholm, Sweden
| | - Tudor Ionescu
- CNSDR, Boehringer Ingelheim Pharma GmbH & Co. KG, Biberach, Germany
| | - Solène Frapard
- Science for Life Laboratory, Department of Gene Technology, KTH Royal Institute of Technology, Stockholm, Sweden
| | - Felix Jung
- Department of Neuroscience, Karolinska Institutet, Stockholm, Sweden
| | - Pierre Le Merre
- Department of Neuroscience, Karolinska Institutet, Stockholm, Sweden
| | - Marcus Saarinen
- Department of Clinical Neuroscience, Karolinska Institutet, Stockholm, Sweden
| | - Katarina Tiklova
- Department of Cell and Molecular Biology, Karolinska Institutet, Stockholm, Sweden
| | | | - Linda Gillberg
- Department of Cell and Molecular Biology, Karolinska Institutet, Stockholm, Sweden
| | - Xiaoqun Zhang
- Department of Clinical Neuroscience, Karolinska Institutet, Stockholm, Sweden
| | - Karima Chergui
- Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden
| | - Marie Carlén
- Department of Neuroscience, Karolinska Institutet, Stockholm, Sweden
| | - Stefania Giacomello
- Science for Life Laboratory, Department of Gene Technology, KTH Royal Institute of Technology, Stockholm, Sweden
| | - Bastian Hengerer
- CNSDR, Boehringer Ingelheim Pharma GmbH & Co. KG, Biberach, Germany
| | - Thomas Perlmann
- Department of Cell and Molecular Biology, Karolinska Institutet, Stockholm, Sweden
| | - Per Svenningsson
- Department of Clinical Neuroscience, Karolinska Institutet, Stockholm, Sweden
| |
Collapse
|
3
|
Barros VN, Barros EMN, da Silva C, Lacerda S, Mello LE. c‐Jun expression after cerebral hyperstimulation differs between rats and marmosets. J Neurosci Res 2019; 97:760-771. [DOI: 10.1002/jnr.24399] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2018] [Revised: 01/28/2019] [Accepted: 01/28/2019] [Indexed: 11/12/2022]
Affiliation(s)
- Vanessa Novaes Barros
- Department of Physiology Escola Paulista de Medicina, Universidade Federal de São Paulo—UNIFESP São Paulo Brazil
| | - Elisa Maria Novaes Barros
- Department of Physiology Escola Paulista de Medicina, Universidade Federal de São Paulo—UNIFESP São Paulo Brazil
| | - Clivandir da Silva
- Department of Physiology Escola Paulista de Medicina, Universidade Federal de São Paulo—UNIFESP São Paulo Brazil
| | | | - Luiz E. Mello
- Department of Physiology Escola Paulista de Medicina, Universidade Federal de São Paulo—UNIFESP São Paulo Brazil
- D’Or Institute for Research and Education, IDOR Rio de Janeiro Brazil
| |
Collapse
|
4
|
Keil MF, Briassoulis G, Stratakis CA. The Role of Protein Kinase A in Anxiety Behaviors. Neuroendocrinology 2016; 103:625-39. [PMID: 26939049 DOI: 10.1159/000444880] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/27/2015] [Accepted: 02/19/2016] [Indexed: 11/19/2022]
Abstract
This review focuses on the genetic and other evidence supporting the notion that the cyclic AMP (cAMP) signaling pathway and its mediator, the protein kinase A (PKA) enzyme, which respond to environmental stressors and regulate stress responses, are central to the pathogenesis of disorders related to anxiety. We describe the PKA pathway and review in vitro animal studies (mouse) and other evidence that support the importance of PKA in regulating behaviors that lead to anxiety. Since cAMP signaling and PKA have been pharmacologically exploited since the 1940s (even before the identification of cAMP as a second messenger with PKA as its mediator) for a number of disorders from asthma to cardiovascular diseases, there is ample opportunity to develop therapies using this new knowledge about cAMP, PKA, and anxiety disorders.
Collapse
Affiliation(s)
- Margaret F Keil
- Section on Endocrinology and Genetics, Program in Developmental Endocrinology and Genetics, Eunice Kennedy Shriver National Institute of Child Health and Human Development (NICHD), National Institutes of Health, Bethesda, Md., USA
| | | | | |
Collapse
|
5
|
Yun JW, Ahn JB, Kang BC. Modeling Parkinson's disease in the common marmoset (Callithrix jacchus): overview of models, methods, and animal care. Lab Anim Res 2015; 31:155-65. [PMID: 26755918 PMCID: PMC4707143 DOI: 10.5625/lar.2015.31.4.155] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2015] [Revised: 11/04/2015] [Accepted: 11/11/2015] [Indexed: 12/14/2022] Open
Abstract
The common marmoset (Callithrix jacchus) is a small-bodied, popular New World monkey and is used widely in reproductive biology, neuroscience, and drug development, due to its comparative ease of handling, high reproductive efficiency, and its unique behavioral characters. In this review, we discuss the marmoset models in Parkinson's disease (PD), which is a neurological movement disorder primarily resulting from a degeneration of dopaminergic neurons with clinical features of tremor, rigidity, postural instability, and akinesia. The most common PD models involve the administration of 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP) or 6-hydroxydopamine to study the pathogenesis and to evaluate novel therapies. Following the systemic or local administration of these neurotoxins, the marmosets with very severe Parkinson's symptoms are recommended to be placed in an intensive care unit with artificial feeding to increase survival rate. All procedures with MPTP should be conducted in a special room with enclosed cages under negative-pressure by trained researchers with personal protection. Behavioral tests are conducted to provide an external measure of the brain pathology. Along with several biomarkers, including α-synuclein and DJ-1, non-invasive neuroimaging techniques such as positron emission tomography and magnetic resonance imaging are used to evaluate the functional changes associated with PD. With the recent growing interest in potential and novel therapies such as stem cell and gene therapy for PD in Korea, the marmoset can be considered as a suitable non-human primate model in PD research to bridge the gap between rodent studies and clinical applications.
Collapse
Affiliation(s)
- Jun-Won Yun
- Department of Experimental Animal Research, Biomedical Research Institute, Seoul National University Hospital, Seoul, Korea
| | - Jae-Bum Ahn
- Department of Experimental Animal Research, Biomedical Research Institute, Seoul National University Hospital, Seoul, Korea
- Graduate School of Translational Medicine, Seoul National University College of Medicine, Seoul, Korea
| | - Byeong-Cheol Kang
- Department of Experimental Animal Research, Biomedical Research Institute, Seoul National University Hospital, Seoul, Korea
- Graduate School of Translational Medicine, Seoul National University College of Medicine, Seoul, Korea
- Designed Animal Research Center, Institute of GreenBio Science Technology, Seoul National University, Pyeongchang-gun, Gangwon, Korea
| |
Collapse
|
6
|
Majláth Z, Vécsei L. NMDA antagonists as Parkinson’s disease therapy: disseminating the evidence. Neurodegener Dis Manag 2014; 4:23-30. [DOI: 10.2217/nmt.13.77] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
SUMMARY Oral levodopa is the current baseline therapy in the management of Parkinson’s disease, but nonmotor complications and therapy-related dyskinesias pose an important challenge for clinicians. Glutamate receptors have been implicated in the neurodegenerative process of Parkinson’s disease and also in the development of levodopa-induced dyskinesias. This article discusses the role of NMDA receptors in Parkinson’s disease and their modulation as a possible therapeutic approach.
Collapse
Affiliation(s)
- Zsófia Majláth
- Department of Neurology, University of Szeged, Semmelweis utca 6, H-6725 Szeged, Hungary
| | - László Vécsei
- Neuroscience Research Group of the Hungarian Academy of Sciences & University of Szeged, Semmelweis utca 6, H-6725 Szeged, Hungary
| |
Collapse
|
7
|
Imbalanced Dopaminergic Transmission Mediated by Serotonergic Neurons in L-DOPA-Induced Dyskinesia. PARKINSONS DISEASE 2011; 2012:323686. [PMID: 22007343 PMCID: PMC3191743 DOI: 10.1155/2012/323686] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/01/2011] [Accepted: 08/02/2011] [Indexed: 11/23/2022]
Abstract
L-DOPA-induced dyskinesias (LIDs) are one of the main motor side effects of L-DOPA therapy in Parkinson's disease. The review will consider the biochemical evidence indicating that the serotonergic neurons are involved in the dopaminergic effects of L-DOPA in the brain. The consequences are an ectopic and aberrant release of dopamine that follows the serotonergic innervation of the brain. After mid- to long-term treatment with L-DOPA, the pattern of L-DOPA-induced dopamine release is modified. In several brain regions, its effect is dramatically reduced while, in the striatum, its effect is quite preserved. LIDs could appear when the dopaminergic effects of L-DOPA fall in brain areas such as the cortex, enhancing the subcortical impact of dopamine and promoting aberrant motor responses. The consideration of the serotonergic system in the core mechanism of action of L-DOPA opens an important reserve of possible strategies to limit LIDs.
Collapse
|
8
|
Ahmed I, Bose SK, Pavese N, Ramlackhansingh A, Turkheimer F, Hotton G, Hammers A, Brooks DJ. Glutamate NMDA receptor dysregulation in Parkinson's disease with dyskinesias. ACTA ACUST UNITED AC 2011; 134:979-86. [PMID: 21371994 DOI: 10.1093/brain/awr028] [Citation(s) in RCA: 156] [Impact Index Per Article: 11.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
Levodopa-induced dyskinesias are a common complication of long-term therapy in Parkinson's disease. Although both pre- and post-synaptic mechanisms seem to be implicated in their development, the precise physiopathology of these disabling involuntary movements remains to be fully elucidated. Abnormalities in glutamate transmission (over expression and phosphorylation of N-methyl-D-aspartate receptors) have been associated with the development of levodopa-induced dyskinesias in animal models of Parkinsonism. The role of glutamate function in dyskinetic patients with Parkinson's disease, however, is unclear. We used (11)C-CNS 5161 [N-methyl-3(thyomethylphenyl)cyanamide] positron emission tomography, a marker of activated N-methyl-D-aspartate receptor ion channels, to compare in vivo glutamate function in parkinsonian patients with and without levodopa-induced dyskinesias. Each patient was assessed with positron emission tomography twice, after taking and withdrawal from levodopa. Striatal and cortical tracer uptake was calculated using a region of interest approach. In the 'OFF' state withdrawn from levodopa, dyskinetic and non-dyskinetic patients had similar levels of tracer uptake in basal ganglia and motor cortex. However, when positron emission tomography was performed in the 'ON' condition, dyskinetic patients had higher (11)C-CNS 5161 uptake in caudate, putamen and precentral gyrus compared to the patients without dyskinesias, suggesting that dyskinetic patients may have abnormal glutamatergic transmission in motor areas following levodopa administration. These findings are consistent with the results of animal model studies indicating that increased glutamatergic activity is implicated in the development and maintenance of levodopa-induced dyskinesias. They support the hypothesis that blockade of glutamate transmission may have a place in the management of disabling dyskinesias in Parkinson's disease.
Collapse
Affiliation(s)
- Imtiaz Ahmed
- Centre for Neuroscience, Imperial College, London, UK
| | | | | | | | | | | | | | | |
Collapse
|
9
|
Jackson MJ, Jenner P. The MPTP-Treated Primate, with Specific Reference to the Use of the Common Marmoset (Callithrix jacchus). NEUROMETHODS 2011. [DOI: 10.1007/978-1-61779-298-4_20] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
|
10
|
Abstract
Gene expression changes in neuropsychiatric and neurodegenerative disorders, and gene responses to therapeutic drugs, provide new ways to identify central nervous system (CNS) targets for drug discovery. This review summarizes gene and pathway targets replicated in expression profiling of human postmortem brain, animal models, and cell culture studies. Analysis of isolated human neurons implicates targets for Alzheimer's disease and the cognitive decline associated with normal aging and mild cognitive impairment. In addition to tau, amyloid-beta precursor protein, and amyloid-beta peptides (Abeta), these targets include all three high-affinity neurotrophin receptors and the fibroblast growth factor (FGF) system, synapse markers, glutamate receptors (GluRs) and transporters, and dopamine (DA) receptors, particularly the D2 subtype. Gene-based candidates for Parkinson's disease (PD) include the ubiquitin-proteosome system, scavengers of reactive oxygen species, brain-derived neurotrophic factor (BDNF), its receptor, TrkB, and downstream target early growth response 1, Nurr-1, and signaling through protein kinase C and RAS pathways. Increasing variability and decreases in brain mRNA production from middle age to old age suggest that cognitive impairments during normal aging may be addressed by drugs that restore antioxidant, DNA repair, and synaptic functions including those of DA to levels of younger adults. Studies in schizophrenia identify robust decreases in genes for GABA function, including glutamic acid decarboxylase, HINT1, glutamate transport and GluRs, BDNF and TrkB, numerous 14-3-3 protein family members, and decreases in genes for CNS synaptic and metabolic functions, particularly glycolysis and ATP generation. Many of these metabolic genes are increased by insulin and muscarinic agonism, both of which are therapeutic in psychosis. Differential genomic signals are relatively sparse in bipolar disorder, but include deficiencies in the expression of 14-3-3 protein members, implicating these chaperone proteins and the neurotransmitter pathways they support as possible drug targets. Brains from persons with major depressive disorder reveal decreased expression for genes in glutamate transport and metabolism, neurotrophic signaling (eg, FGF, BDNF and VGF), and MAP kinase pathways. Increases in these pathways in the brains of animals exposed to electroconvulsive shock and antidepressant treatments identify neurotrophic and angiogenic growth factors and second messenger stimulation as therapeutic approaches for the treatment of depression.
Collapse
|
11
|
Barnum CJ, Eskow KL, Dupre K, Blandino P, Deak T, Bishop C. Exogenous corticosterone reduces L-DOPA-induced dyskinesia in the hemi-parkinsonian rat: role for interleukin-1beta. Neuroscience 2008; 156:30-41. [PMID: 18687386 DOI: 10.1016/j.neuroscience.2008.07.016] [Citation(s) in RCA: 68] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2008] [Revised: 06/18/2008] [Accepted: 07/08/2008] [Indexed: 10/21/2022]
Abstract
While the etiology of Parkinson's disease (PD) remains unknown, there is overwhelming evidence that neuroinflammation plays a critical role in the progressive loss of dopamine (DA) neurons. Because nearly all persons suffering from PD receive l-DOPA, it is surprising that inflammation has not been examined as a potential contributor to the abnormal involuntary movements (AIMs) that occur as a consequence of chronic l-DOPA treatment. As an initial test of this hypothesis, we examined the effects of exogenously administered corticosterone (CORT), an endogenous anti-inflammatory agent, on the expression and development of l-DOPA-induced dyskinesia (LID) in unilateral DA-depleted rats. To do this, male Sprague-Dawley rats received unilateral medial forebrain bundle 6-hydroxydopamine lesions. Three weeks later, l-DOPA primed rats received acute injections of CORT (0-3.75 mg/kg) prior to l-DOPA to assess the expression of LID. A second group of rats was used to examine the development of LID in l-DOPA naïve rats co-treated with CORT and l-DOPA for 2 weeks. AIMs and rotations were recorded. Exogenous CORT dose-dependently attenuated both the expression and development of AIMs without affecting rotations. Real-time reverse-transcription polymerase chain reaction of striatal tissue implicated a role for interleukin-1 (IL-1) beta in these effects as its expression was increased on the lesioned side in rats treated with l-DOPA (within the DA-depleted striatum) and attenuated with CORT. In the final experiment, interleukin-1 receptor antagonist (IL-1ra) was microinjected into the striatum of l-DOPA-primed rats to assess the impact of IL-1 signaling on LID. Intrastriatal IL-1ra reduced the expression of LID without affecting rotations. These findings indicate a novel role for neuroinflammation in the expression of LID, and may implicate the use of anti-inflammatory agents as a potential adjunctive therapy for the treatment of LID.
Collapse
Affiliation(s)
- C J Barnum
- Behavioral Neuroscience Program, Department of Psychology, State University of New York at Binghamton, 4400 Vestal Parkway East, Binghamton, NY 13902, USA
| | | | | | | | | | | |
Collapse
|
12
|
Zhang X, Andren PE, Svenningsson P. Changes on 5-HT2 receptor mRNAs in striatum and subthalamic nucleus in Parkinson's disease model. Physiol Behav 2007; 92:29-33. [PMID: 17588622 DOI: 10.1016/j.physbeh.2007.05.033] [Citation(s) in RCA: 41] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Abnormal interactions between the serotonin and dopamine systems may underlie the high prevalence of non-motor complications in Parkinson's disease (PD). Here, we demonstrate that the genes encoding serotonin 5-HT2A and 5-HT2C receptors are differently regulated by dopamine in the 6-hydroxydopamine (6-OHDA) rat model of PD. Nigrostriatal cell loss causes an up-regulation of 5-HT2AR mRNA, but a down-regulation of 5-HT2CR mRNA, in striatum. Repeated injections with L-DOPA/benserazide reverse the effect of 6-OHDA lesioning on 5-HT2AR, but not on 5-HT2CR, gene expression. Neither 6-OHDA-lesioning nor L-DOPA/benserazide treatment had any effect on 5-HT2AR mRNA in cortex or on 5-HT2CR mRNA in nucleus subthalamicus. These data suggest that the regulation of 5-HT2AR in striatum, in the 6-OHDA rat model of PD, is mainly dependent upon alterations in dopamine levels. 5-HT2CR, on the other hand, are regulated by nigrostriatal cell loss and by the accompanied reduction of factor(s), other than dopamine, that are normally co-expressed with dopamine. The apparent imbalance between 5-HT2AR and 5-HT2CR levels in this PD model indicates a potential role for these receptors in the pathophysiology of neuropsychiatric symptoms, such as depression and L-DOPA-induced hallucinations, which are co-morbid with PD. The fact that 5-HT2CR are differentially regulated as compared to 5-HT2AR to alterations in the dopamine tone predicts that pharmacological manipulations at 5-HT2CR, but not at 5-HT2AR, will result in similar effects in PD patients whether they are treated or not with dopamine replacement.
Collapse
MESH Headings
- Animals
- Cell Death
- Disease Models, Animal
- Dopamine/physiology
- Gene Expression Regulation
- Male
- Neostriatum/cytology
- Neostriatum/metabolism
- Neurons/cytology
- Neurons/metabolism
- Oxidopamine
- Parkinsonian Disorders/chemically induced
- Parkinsonian Disorders/metabolism
- RNA, Messenger/metabolism
- Rats
- Rats, Sprague-Dawley
- Receptor, Serotonin, 5-HT2A/genetics
- Receptor, Serotonin, 5-HT2A/metabolism
- Receptor, Serotonin, 5-HT2C/genetics
- Receptor, Serotonin, 5-HT2C/metabolism
- Subthalamic Nucleus/cytology
- Subthalamic Nucleus/metabolism
Collapse
Affiliation(s)
- Xiaoqun Zhang
- Karolinska Institutet, Section for Translational Neuropharmacology, Department of Physiology and Pharmacology, S-171 77 Stockholm, Sweden
| | | | | |
Collapse
|
13
|
Långström B, Andrén PE, Lindhe O, Svedberg M, Hall H. In Vitro Imaging Techniques in Neurodegenerative Diseases. Mol Imaging Biol 2007; 9:161-75. [PMID: 17318669 DOI: 10.1007/s11307-007-0088-1] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
Neurodegeneration induces various changes in the brain, changes that may be investigated using neuroimaging techniques. The in vivo techniques are useful for the visualization of major changes, and the progressing abnormalities may also be followed longitudinally. However, to study and quantify minor abnormalities, neuroimaging of postmortem brain tissue is used. These in vitro methods are complementary to the in vivo techniques and contribute to the knowledge of pathophysiology and etiology of the neurodegenerative diseases. In vitro radioligand autoradiography has given great insight in the involvement of different neuronal receptor systems in these diseases. Data on the dopamine and cholinergic systems in neurodegeneration are discussed in this review. Also, the amyloid plaques are studied using in vitro radioligand autoradiography. Using one of the newer methods, imaging matrix-assisted laser desorption ionization mass spectrometry, the distribution of a large number of peptides and proteins may be detected in vitro on brain cryosections. In this overview, we describe in vitro imaging techniques in the neurodegenerative diseases as a complement to in vivo positron emission tomography and single photon emission computed tomography imaging.
Collapse
Affiliation(s)
- Bengt Långström
- Department of Biochemistry and Organic Chemistry, Uppsala University, Uppsala, Sweden.
| | | | | | | | | |
Collapse
|
14
|
Valastro B, Andersson M, Lindgren HS, Cenci MA. Expression pattern of JunD after acute or chronic l-DOPA treatment: Comparison with ΔFosB. Neuroscience 2007; 144:198-207. [PMID: 17055656 DOI: 10.1016/j.neuroscience.2006.09.001] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2006] [Revised: 08/31/2006] [Accepted: 09/01/2006] [Indexed: 11/19/2022]
Abstract
In this study, we have used 6-hydroxydopamine-lesioned rats to examine changes in striatal junD and fosB/deltafosB expression induced by acute and chronic treatment with L-DOPA (5 and 15 days). Changes at the protein levels were studied using Western immunoblotting while mRNA changes were compared using in situ hybridization histochemistry. We observed a significant increase in the level of deltaFosB proteins after chronic treatment with L-DOPA, an effect that was not observed for JunD proteins. In addition, the upregulation of deltaFosB was already present after an acute treatment but increased upon chronic treatment. By contrast, junD and deltafosB mRNA were both upregulated significantly above control levels after an acute injection of L-DOPA. In conclusion, this study suggests a differential expression pattern of junD and deltafosB in a rat model of L-DOPA-induced dyskinesia. The upregulation of deltaFosB protein, but not JunD, is likely to reflect an increased stability of the deltaFosB proteins without ongoing enhanced transcription of the encoding genes.
Collapse
Affiliation(s)
- B Valastro
- Basal Ganglia Pathophysiology Unit, Department of Experimental Medical Science, Lund University, BMC F11, Lund 221 84, Sweden.
| | | | | | | |
Collapse
|
15
|
Nelson M, Foxwell AR, Tyrer P, Dean RT. Protein-bound 3,4-dihydroxy-phenylanine (DOPA), a redox-active product of protein oxidation, as a trigger for antioxidant defences. Int J Biochem Cell Biol 2006; 39:879-89. [PMID: 17098462 DOI: 10.1016/j.biocel.2006.10.004] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2006] [Revised: 10/01/2006] [Accepted: 10/02/2006] [Indexed: 11/20/2022]
Abstract
Protein hydroperoxides and protein-bound 3,4-dihydroxy-phenylanine are amongst the major long-lived redox-active products during free radical attack on proteins. Protein-bound 3,4-dihydroxy-phenylanine can redox cycle between catechol and quinone form, and bind transition metals, whereas hydroperoxides are converted to stable hydroxides. The free amino acid 3,4-dihydroxy-phenylanine is a normal metabolite, an oxidation product of tyrosine, involved in pathways of dopamine and melanin production, and we have shown that it may be incorporated into protein-by-protein synthesis. However, physiological levels of protein-bound 3,4-dihydroxy-phenylanine are very low; yet remarkably elevated levels occur in some pathologies. We propose that, unlike free 3,4-dihydroxy-phenylanine, protein-bound 3,4-dihydroxy-phenylanine is a signal for the activation of cellular defences both against the oxidative fluxes during oxidative stress and against the oxidative damage which sometimes ensues. Unlike free 3,4-dihydroxy-phenylanine, the levels of protein-bound 3,4-dihydroxy-phenylanine can change 5-10-fold during oxidative damage in vivo, an appropriate property for a signalling molecule. We suggest mechanisms by which protein-bound 3,4-dihydroxy-phenylanine might trigger oxidative defences, via NF-kappaB and other transcription factors. Little evidence yet bears directly on this, but we discuss some implications of observations on free 3,4-dihydroxy-phenylanine supply to cells in vitro, to Parkinson's patients, and to animal models of the disease. Several of the effects of 3,4-dihydroxy-phenylanine in these situations may be mediated by the production and actions of protein-bound 3,4-dihydroxy-phenylanine. Some experimental tests of the hypothesis are outlined and some possible therapeutic implications.
Collapse
Affiliation(s)
- Michelle Nelson
- Gadi Research Group, University of Canberra, ACT 2601, Australia
| | | | | | | |
Collapse
|
16
|
Sköld K, Svensson M, Nilsson A, Zhang X, Nydahl K, Caprioli RM, Svenningsson P, Andrén PE. Decreased Striatal Levels of PEP-19 Following MPTP Lesion in the Mouse. J Proteome Res 2006; 5:262-9. [PMID: 16457591 DOI: 10.1021/pr050281f] [Citation(s) in RCA: 92] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
PEP-19 is a neuronal calmodulin-binding protein, and as such, a putative modulator of calcium regulated processes. In the present study, we used proteomics technology approaches such as peptidomics and imaging MALDI mass spectrometry, as well as traditional techniques (immunoblotting and in situ hybridization) to identify PEP-19 and, specifically, to measure PEP-19 mRNA and protein levels in an animal model of Parkinson's disease. 1-Methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP) administration in mice resulted in a significant decrease in striatal PEP-19 mRNA. Capillary nano-flow liquid chromatography electrospray mass spectrometry analysis of striatal tissue revealed a significant decrease of the PEP-19 protein level. Moreover, imaging MALDI mass spectrometry also showed that PEP-19 protein was predominantly localized to the striatum of the brain tissue cross sections. After MPTP administration, PEP-19 levels were significantly reduced by 30%. We conclude that PEP-19 mRNA and protein expression are decreased in the striatum of a common animal model of Parkinson's disease. Further studies are needed to show the specific involvement of PEP-19 in the neurodegeneration seen in MPTP lesioned animals. Finally, this study has shown that the combination of traditional molecular biology techniques with novel, highly specific and sensitive mass spectrometry methods is advantageous in characterizing molecular events of many diseases, including Parkinson's disease.
Collapse
Affiliation(s)
- Karl Sköld
- Laboratory for Biological and Medical Mass Spectrometry, Uppsala University, Box 583 Biomedical Centre, SE-75123 Uppsala, Sweden
| | | | | | | | | | | | | | | |
Collapse
|
17
|
Huzarska M, Zieliński M, Herman ZS. Repeated treatment with antidepressants enhances dopamine D1 receptor gene expression in the rat brain. Eur J Pharmacol 2006; 532:208-13. [PMID: 16499906 DOI: 10.1016/j.ejphar.2005.09.069] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2005] [Revised: 08/23/2005] [Accepted: 09/01/2005] [Indexed: 11/19/2022]
Abstract
Many pharmacological investigations have demonstrated that antidepressant agents profoundly affect serotonergic and noradrenergic neurotransmission. The molecular mechanisms by which these drugs exert their therapeutic action have not been clearly established. In our study, the possibility that antidepressant drug action is associated with dopamine neurotransmission was examined. To this end, the effect of 21-day treatment with 10 mg/kg of amitryptyline, mirtazapine and sertraline on the striatal and nucleus accumbens dopamine receptors was verified. The striatum and nucleus accumbens tissues were dissected 24 h after the last dose of the drug and total RNA was isolated. The expression of dopamine D1 to dopamine D5 receptors using reverse-transcriptase polymerase chain reaction (RT-PCR) procedure was compared to the glyceraldehyde-3-phosphate-dehydrogenase (GAPDH) as constitutive gene activation internal control. Lab Works UV program has analyzed the mean optical density values of RT-PCR products. Statistical comparison of relative optical densities by a one-way analysis of variance (ANOVA) followed by Dunnett's test was performed. Despite their different pharmacological profiles, all three above-presented antidepressants significantly increased dopamine D(1) mRNA content. Our findings indicate that repeated antidepressant administration triggers induction of the brain dopaminergic receptors which is correlated with neuroadaptation of the brain dopaminergic pathway.
Collapse
Affiliation(s)
- Małgorzata Huzarska
- Department of Clinical Pharmacology, Medical University of Silesia, Medyków 18, 40-752 Katowice, Poland.
| | | | | |
Collapse
|
18
|
Liu WG, Chen Y, Li B, Lu GQ, Chen SD. Neuroprotection by Pergolide Against Levodopa-Induced Cytotoxicity of Neural Stem Cells. Neurochem Res 2004; 29:2207-14. [PMID: 15672541 DOI: 10.1007/s11064-004-7027-1] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/01/2022]
Abstract
Neural stem cells (NSCs) are currently considered very hopeful candidates for cell replacement therapy in neurodegenerative pathologies such as Parkinson's disease (PD), but like embryonic neural tissue transplantation, levodopa medication may still be required to improve symptoms even after cell transplantation. The issues of whether levodopa induces cytotoxicity and apoptosis of NSCs following transplantation, as well as the means to prevent these processes from occurring remain to be elucidated. In this study, the possible cytotoxicity of levodopa at different doses on C17.2 neural stem cells and subsequent neuroprotection by pergolide were investigated. The cell viability was determined by the MTT assay. Cell proliferation was assayed by BrdU labeling, while apoptosis was detected by Annexin-V-FLUOS staining and flow cytometry. Levels of p53, Bax, Bcl-2, NFkB, cytochrome c, caspase-3 as well as cleavage of caspase-3 were measured by western blotting. We found levodopa induced a concentration- and time-dependent decrease in cell viability and proliferation. Apoptotic cells were observed at different stages, specifically 12 and 24 h following exposure to levodopa (200 microM). Elevated p53, Bax, cytochrome c, caspase-3 and active fragments of caspase-3 protein were observed in the cells exposed to levodopa. These alterations were partly inhibited by pergolide, a dopamine receptor agonist, while Bcl-2 and NFkB p65 levels remained constant at the various time-points in all the groups examined. These observations indicate that levodopa at high concentrations (> or = 200 microM) was neurotoxic to C17.2 neural stem cells via inhibition of DNA synthesis and cell proliferation. Activation of the mitochondria-dependent pathway and caspase-3 protease may contribute to the mechanism by which levodopa induces apoptosis. Pergolide, an anti-Parkinson drug, has a neuroprotective effect and partly blocks levodopa-induced cytotoxicity.
Collapse
Affiliation(s)
- Wei-Guo Liu
- Department of Neurology, Clinical & Research Center for Parkinson Disease & Movement Disorders, Ruijin Hospital, Shanghai Second Medical University, Shanghai 200025, PR China
| | | | | | | | | |
Collapse
|