1
|
Garwood M, Vijayakumar P, Bohnen NI, Koeppe RA, Kotagal V. Serotonin transporter density in isolated rapid eye movement sleep behavioral disorder. FRONTIERS IN SLEEP 2024; 2:1298854. [PMID: 38765701 PMCID: PMC11101191 DOI: 10.3389/frsle.2023.1298854] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Indexed: 05/22/2024]
Abstract
Background/objective The serotoninergic nervous system is known to play a role in the maintenance of rapid eye movement (REM) sleep. Serotoninergic projections are known to be vulnerable in synucleinopathies. To date, positron emission tomography (PET) studies using serotonin-specific tracers have not been reported in isolated REM sleep behavior disorder (iRBD). Methods We conducted a cross-sectional imaging study using serotonin transporter (SERT) 11C-3-amino-4-(2-dimethylaminomethyl-phenylsulfaryl)-benzonitrile (DASB) PET to identify differences in serotonin system integrity between 11 participants with iRBD and 16 older healthy controls. Results Participants with iRBD showed lower DASB distribution volume ratios (DVRs) in the total neocortical mantle [1.13 (SD: 0.07) vs. 1.19 (SD: 0.06); t = 2.33, p = 0.028)], putamen [2.07 (SD: 0.19) vs. 2.25 (SD: 0.18); t = 2.55, p = 0.017], and insula [1.26 (SD: 0.11) vs. 1.39 (SD: 0.09); t = 3.58, p = 0.001]. Paradoxical increases relative to controls were seen in cerebellar hemispheres [0.98 (SD: 0.04) vs. 0.95 (SD: 0.02); t = 2.93, p = 0.007)]. No intergroup differences were seen in caudate, substantia nigra, or other brainstem regions with the exception of the dorsal mesencephalic raphe [3.08 (SD: 0.53) vs. 3.47 (SD: 0.48); t = 2.00, p = 0.056] that showed a non-significant trend toward lower values in iRBD. Conclusions Insular, neocortical, and striatal serotoninergic terminal loss may be common in prodromal synucleinopathies before the onset of parkinsonism or dementia. Given our small sample size, these results should be interpreted as hypothesis-generating/exploratory in nature.
Collapse
Affiliation(s)
- Mark Garwood
- Department of Neurology, University of Michigan, Ann Arbor, MI, United States
| | | | - Nicolaas I. Bohnen
- Department of Neurology, University of Michigan, Ann Arbor, MI, United States
- Division of Nuclear Medicine, Department of Radiology, University of Michigan, Ann Arbor, MI, United States
- Ann Arbor Veterans Affairs Healthcare System, VAAAHS Geriatric Research Education and Clinical Center, Ann Arbor, MI, United States
| | - Robert A. Koeppe
- Division of Nuclear Medicine, Department of Radiology, University of Michigan, Ann Arbor, MI, United States
| | - Vikas Kotagal
- Department of Neurology, University of Michigan, Ann Arbor, MI, United States
- Ann Arbor Veterans Affairs Healthcare System, VAAAHS Geriatric Research Education and Clinical Center, Ann Arbor, MI, United States
| |
Collapse
|
2
|
Woo KA, Joun JH, Yoon EJ, Lee CY, Jeon B, Kim YK, Lee JY. Monoaminergic Degeneration and Ocular Motor Abnormalities in De Novo Parkinson's Disease. Mov Disord 2023; 38:2291-2301. [PMID: 37846885 DOI: 10.1002/mds.29623] [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: 03/07/2023] [Revised: 07/07/2023] [Accepted: 09/12/2023] [Indexed: 10/18/2023] Open
Abstract
BACKGROUND Evaluating eye movements in Parkinson's disease (PD) provides valuable insights into the underlying pathophysiological changes. OBJECTIVE The aim was to investigate the relationship between monoaminergic degeneration and ocular motor abnormalities in de novo PD. METHODS Drug-naive PD patients who underwent N-(3-[18 F]fluoropropyl)-2-carbomethoxy-3-(4-iodophenyl) nortropane positron emission tomography scans and video-oculography at diagnosis were eligible. Measurements of saccadic accuracy, latency, and smooth pursuit gain and square wave jerk frequency were collected. Patients underwent Movement Disorders Society-Unified Parkinson's Disease Rating Scale (MDS-UPDRS) and detailed cognitive tests. We investigated the associations between ocular motor measurements and specific tracer uptake ratios (SUR) in the caudate nucleus, anterior and posterior putamen, thalamus, and dorsal raphe nuclei, along with motor and cognitive symptoms. RESULTS One-hundred twenty-four subjects were included in this study. Saccadic accuracy was positively associated with parkinsonian motor severity expressed as Hoehn and Yahr stages, MDS-UPDRS Part III scores, and subscores for bradykinesia and rigidity but not with tremor scores (PFDR < 0.05). Saccadic accuracy correlated with poor performances in the Rey-Complex-Figure copy, and latency with the Digit Symbol Coding and the Montreal Cognitive Assessment scores (PFDR < 0.05). Prolonged saccadic latency correlated with reduced thalamic SUR, whereas decreased saccadic accuracy correlated with reduced SUR in the anterior and posterior putamen (PFDR < 0.05). Reduced smooth pursuit gain showed associations with reduced SUR in the dorsal raphe, a serotonin-predominant region, but did not correlate with parkinsonism severity scores. CONCLUSION Defective dopaminergic and nondopaminergic neural systems may discretely influence ocular motor function in de novo PD patients. © 2023 International Parkinson and Movement Disorder Society.
Collapse
Affiliation(s)
- Kyung Ah Woo
- Department of Neurology, Seoul Metropolitan Government-Seoul National University Boramae Medical Center, Seoul National University College of Medicine, Seoul, Republic of Korea
| | - Joo Hong Joun
- Department of Neurology, Seoul Metropolitan Government-Seoul National University Boramae Medical Center, Seoul National University College of Medicine, Seoul, Republic of Korea
| | - Eun Jin Yoon
- Department of Nuclear Medicine, Seoul Metropolitan Government-Seoul National University Boramae Medical Center, Seoul National University College of Medicine, Seoul, Republic of Korea
- Memory Network Medical Research Center, Seoul National University, Seoul, Republic of Korea
| | - Chan Young Lee
- Department of Neurology, Ewha Womans University Mokdong Hospital, Ewha Womans University College of Medicine, Seoul, Republic of Korea
| | - Beomseok Jeon
- Department of Neurology, Seoul National University Hospital, Seoul National University College of Medicine, Seoul, Republic of Korea
| | - Yu Kyeong Kim
- Department of Nuclear Medicine, Seoul Metropolitan Government-Seoul National University Boramae Medical Center, Seoul National University College of Medicine, Seoul, Republic of Korea
| | - Jee-Young Lee
- Department of Neurology, Seoul Metropolitan Government-Seoul National University Boramae Medical Center, Seoul National University College of Medicine, Seoul, Republic of Korea
| |
Collapse
|
3
|
Conti Mazza MM, Centner A, Werner DF, Bishop C. Striatal serotonin transporter gain-of-function in L-DOPA-treated, hemi-parkinsonian rats. Brain Res 2023; 1811:148381. [PMID: 37127174 PMCID: PMC10562932 DOI: 10.1016/j.brainres.2023.148381] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2023] [Revised: 04/24/2023] [Accepted: 04/26/2023] [Indexed: 05/03/2023]
Abstract
L-DOPA is the standard treatment for Parkinson's disease (PD), but chronic treatment typically leads to L-DOPA-induced dyskinesia (LID). LID involves a complex interaction between the remaining dopamine (DA) system and the semi-homologous serotonin (5-HT) system. Since serotonin transporters (SERT) have some affinity for DA uptake, they may serve as a functional compensatory mechanism when DA transporters (DAT) are scant. DAT and SERT's functional contributions in the dyskinetic brain have not been well delineated. The current investigation sought to determine how DA depletion and L-DOPA treatment affect DAT and SERT transcriptional processes, translational processes, and functional DA uptake in the 6-hydroxydopamine-lesioned hemi-parkinsonian rat. Rats were counterbalanced for motor impairment into equally lesioned treatment groups then given daily L-DOPA (0 or 6 mg/kg) for 2 weeks. At the end of treatment, the substantia nigra was processed for tyrosine hydroxylase (TH) and DAT gene expression and dorsal raphe was processed for SERT gene expression. The striatum was processed for synaptosomal DAT and SERT protein expression and ex vivo DA uptake. Nigrostriatal DA loss severely reduced DAT mRNA and protein expression in the striatum with minimal changes in SERT. L-DOPA treatment, while not significantly affecting DAT or SERT alone, did increase striatal SERT:DAT protein ratios. Using ex vivo microdialysis, L-DOPA treatment increased DA uptake via SERT when DAT was depleted. Overall, these results suggest that DA loss and L-DOPA treatment uniquely alter DAT and SERT, revealing implications for monoamine transporters as potential biomarkers and therapeutic targets in the hemi-parkinsonian model and dyskinetic PD patients.
Collapse
Affiliation(s)
- Melissa M Conti Mazza
- Behavioral Neuroscience Program, Department of Psychology, Binghamton University, 4400 Vestal Parkway East, Binghamton, NY 13902-6000, USA
| | - Ashley Centner
- Behavioral Neuroscience Program, Department of Psychology, Binghamton University, 4400 Vestal Parkway East, Binghamton, NY 13902-6000, USA
| | - David F Werner
- Behavioral Neuroscience Program, Department of Psychology, Binghamton University, 4400 Vestal Parkway East, Binghamton, NY 13902-6000, USA
| | - Christopher Bishop
- Behavioral Neuroscience Program, Department of Psychology, Binghamton University, 4400 Vestal Parkway East, Binghamton, NY 13902-6000, USA.
| |
Collapse
|
4
|
Pinna A, Parekh P, Morelli M. Serotonin 5-HT 1A receptors and their interactions with adenosine A 2A receptors in Parkinson's disease and dyskinesia. Neuropharmacology 2023; 226:109411. [PMID: 36608814 DOI: 10.1016/j.neuropharm.2023.109411] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2022] [Revised: 12/19/2022] [Accepted: 01/03/2023] [Indexed: 01/05/2023]
Abstract
The dopamine neuronal loss that characterizes Parkinson's Disease (PD) is associated to changes in neurotransmitters, such as serotonin and adenosine, which contribute to the symptomatology of PD and to the onset of dyskinetic movements associated to levodopa treatment. The present review describes the role played by serotonin 5-HT1A receptors and the adenosine A2A receptors on dyskinetic movements induced by chronic levodopa in PD. The focus is on preclinical and clinical results showing the interaction between serotonin 5-HT1A receptors and other receptors such as 5-HT1B receptors and adenosine A2A receptors. 5-HT1A/1B receptor agonists and A2A receptor antagonists, administered in combination, contrast dyskinetic movements induced by chronic levodopa without impairing motor behaviour, suggesting that this drug combination might be a useful therapeutic approach for counteracting the PD motor deficits and dyskinesia associated with chronic levodopa treatment. This article is part of the Special Issue on "The receptor-receptor interaction as a new target for therapy".
Collapse
Affiliation(s)
- Annalisa Pinna
- National Research Council of Italy, Neuroscience Institute, UOS of Cagliari, c/o Department of Biomedical Sciences, Cittadella Universitaria di Monserrato, 09042, Monserrato (CA), Italy.
| | - Pathik Parekh
- Department of Biomedical Sciences, Section of Neuroscience, University of Cagliari, Cittadella Universitaria di Monserrato, 09042, Monserrato (CA), Italy
| | - Micaela Morelli
- National Research Council of Italy, Neuroscience Institute, UOS of Cagliari, c/o Department of Biomedical Sciences, Cittadella Universitaria di Monserrato, 09042, Monserrato (CA), Italy; Department of Biomedical Sciences, Section of Neuroscience, University of Cagliari, Cittadella Universitaria di Monserrato, 09042, Monserrato (CA), Italy.
| |
Collapse
|
5
|
Budrow C, Elder K, Coyle M, Centner A, Lipari N, Cohen S, Glinski J, Kinzonzi N, Wheelis E, McManus G, Manfredsson F, Bishop C. Broad Serotonergic Actions of Vortioxetine as a Promising Avenue for the Treatment of L-DOPA-Induced Dyskinesia. Cells 2023; 12:837. [PMID: 36980178 PMCID: PMC10047495 DOI: 10.3390/cells12060837] [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: 02/09/2023] [Revised: 03/03/2023] [Accepted: 03/05/2023] [Indexed: 03/30/2023] Open
Abstract
Parkinson's Disease (PD) is a neurodegenerative disorder characterized by motor symptoms that result from loss of nigrostriatal dopamine (DA) cells. While L-DOPA provides symptom alleviation, its chronic use often results in the development of L-DOPA-induced dyskinesia (LID). Evidence suggests that neuroplasticity within the serotonin (5-HT) system contributes to LID onset, persistence, and severity. This has been supported by research showing 5-HT compounds targeting 5-HT1A/1B receptors and/or the 5-HT transporter (SERT) can reduce LID. Recently, vortioxetine, a multimodal 5-HT compound developed for depression, demonstrated acute anti-dyskinetic effects. However, the durability and underlying pharmacology of vortioxetine's anti-dyskinetic actions have yet to be delineated. To address these gaps, we used hemiparkinsonian rats in Experiment 1, examining the effects of sub-chronic vortioxetine on established LID and motor performance. In Experiment 2, we applied the 5-HT1A antagonist WAY-100635 or 5-HT1B antagonist SB-224289 in conjunction with L-DOPA and vortioxetine to determine the contributions of each receptor to vortioxetine's effects. The results revealed that vortioxetine consistently and dose-dependently attenuated LID while independently, 5-HT1A and 5-HT1B receptors each partially reversed vortioxetine's effects. Such findings further support the promise of pharmacological strategies, such as vortioxetine, and indicate that broad 5-HT actions may provide durable responses without significant side effects.
Collapse
Affiliation(s)
- Carla Budrow
- Behavioral Neuroscience Program, Department of Psychology, Binghamton University, Binghamton, NY 13902, USA; (C.B.)
| | - Kayla Elder
- Behavioral Neuroscience Program, Department of Psychology, Binghamton University, Binghamton, NY 13902, USA; (C.B.)
| | - Michael Coyle
- Behavioral Neuroscience Program, Department of Psychology, Binghamton University, Binghamton, NY 13902, USA; (C.B.)
| | - Ashley Centner
- Behavioral Neuroscience Program, Department of Psychology, Binghamton University, Binghamton, NY 13902, USA; (C.B.)
| | - Natalie Lipari
- Behavioral Neuroscience Program, Department of Psychology, Binghamton University, Binghamton, NY 13902, USA; (C.B.)
| | - Sophie Cohen
- Behavioral Neuroscience Program, Department of Psychology, Binghamton University, Binghamton, NY 13902, USA; (C.B.)
| | - John Glinski
- Behavioral Neuroscience Program, Department of Psychology, Binghamton University, Binghamton, NY 13902, USA; (C.B.)
| | - N’Senga Kinzonzi
- Behavioral Neuroscience Program, Department of Psychology, Binghamton University, Binghamton, NY 13902, USA; (C.B.)
| | - Emily Wheelis
- Behavioral Neuroscience Program, Department of Psychology, Binghamton University, Binghamton, NY 13902, USA; (C.B.)
| | - Grace McManus
- Behavioral Neuroscience Program, Department of Psychology, Binghamton University, Binghamton, NY 13902, USA; (C.B.)
| | - Fredric Manfredsson
- Department of Neurobiology, Barrow Neurological Institute, Phoenix, AZ 85013, USA
| | - Christopher Bishop
- Behavioral Neuroscience Program, Department of Psychology, Binghamton University, Binghamton, NY 13902, USA; (C.B.)
| |
Collapse
|
6
|
Xu C, Wu Y, Tang L, Liang Y, Zhao Y. Protective effect of cistanoside A on dopaminergic neurons in Parkinson's disease via mitophagy. Biotechnol Appl Biochem 2023; 70:268-280. [PMID: 35420720 DOI: 10.1002/bab.2350] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2021] [Accepted: 03/23/2022] [Indexed: 11/05/2022]
Abstract
One of the main pathological features of Parkinson's disease (PD) is the loss of dopaminergic neurons in the substantia nigra compacta (SNc). Cistanoside A (CA) has a strong neuroprotective effect in PD, but the exact mechanism is unclear. In the present study, the MPTP-stimulated mouse model of PD and MPP+ -treated PD model in the MES23.5 neuronal cell model of PD were used to investigate the neuroprotective effects of CA on PD and its potential mechanism. The in vivo experiment results indicated that CA improved the motor function in mice and increased the number of tyrosine hydroxylase positive cells in SNc. In vitro experiments showed that CA reduced the MPP+ -induced decrease in neurons and mitochondrial membrane potential and promoted the activation of autophagosomes. Furthermore, we found that CA promoted the recruitment of PINK1 and Parkin aggregation to impair mitochondrial membranes and inhibited mitochondrial damage via LC3- and p62-mediated autophagy. In conclusion, CA protects against MPTP-induced neurotoxicity in vivo and MPP+ -induced neurotoxicity in vitro, possibly by promoting the PINK1/Parkin/p62 pathway to accelerate the degradation of damaged mitochondria thereby reducing oxidative stress.
Collapse
Affiliation(s)
- Chengcheng Xu
- Nanjing University of Chinese Medicine, Nanjing, Jiangsu, China
| | - Yao Wu
- Department of Neurology, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, Jiangsu, China
| | - Lili Tang
- Department of Neurology, Nanjing Hospital of Chinese Medicine Affiliated to Nanjing University of Chinese Medicine, Nanjing, Jiangsu, China
| | - Yan Liang
- Department of Neurology, Nanjing Hospital of Chinese Medicine Affiliated to Nanjing University of Chinese Medicine, Nanjing, Jiangsu, China
| | - Yang Zhao
- Department of Neurology, Nanjing Hospital of Chinese Medicine Affiliated to Nanjing University of Chinese Medicine, Nanjing, Jiangsu, China
| |
Collapse
|
7
|
Synergistic effect of serotonin 1A and serotonin 1B/D receptor agonists in the treatment of L-DOPA-induced dyskinesia in 6-hydroxydopamine-lesioned rats. Exp Neurol 2022; 358:114209. [PMID: 35988699 DOI: 10.1016/j.expneurol.2022.114209] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2022] [Revised: 08/01/2022] [Accepted: 08/14/2022] [Indexed: 11/24/2022]
Abstract
BACKGROUND The gold standard for symptomatic relief of Parkinson's disease (PD) is L-DOPA. However, long-term treatment often leads to motor complications such as L-DOPA-induced dyskinesia (LID). While amantadine (Gocovri™) is the only approved therapy for dyskinesia in PD patients on the American market, it is associated with neurological side effects and limited efficacy. Thus, there remains a high unmet need for addressing LID in PD patients worldwide. OBJECTIVE The objective of this study was to evaluate the efficacy, safety and performance compared to approved treatments of the serotonin receptor 1A (5-HT1A) and 5-HT1B/D agonists buspirone and zolmitriptan in the 6-hydroxydopamine unilaterally lesioned rat model for PD. METHODS The hemiparkinsonian 6-OHDA-lesioned rats underwent chronic treatment with L-DOPA to induce dyskinesia and were subsequently used for efficacy testing of buspirone, zolmitriptan and comparison with amantadine, measured as abnormal involuntary movement (AIM) scores after L-DOPA challenge. Safety testing was performed in model and naïve animals using forelimb adjusting, rotarod and open field tests. RESULTS 5-HT1A and 5-HT1B/D agonism effectively reduced AIM scores in a synergistic manner. The drug combination of buspirone and zolmitriptan was safe and did not lead to tolerance development following sub-chronic administration. Head-to-head comparison with amantadine showed superior performance of buspirone and zolmitriptan in the model. CONCLUSIONS The strong anti-dyskinetic effect found with combined 5-HT1A and 5-HT1B/D agonism renders buspirone and zolmitriptan together a meaningful treatment for LID in PD.
Collapse
|
8
|
Cohen SR, Terry ML, Coyle M, Wheelis E, Centner A, Smith S, Glinski J, Lipari N, Budrow C, Manfredsson FP, Bishop C. The multimodal serotonin compound Vilazodone alone, but not combined with the glutamate antagonist Amantadine, reduces l-DOPA-induced dyskinesia in hemiparkinsonian rats. Pharmacol Biochem Behav 2022; 217:173393. [DOI: 10.1016/j.pbb.2022.173393] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/04/2022] [Revised: 04/25/2022] [Accepted: 04/27/2022] [Indexed: 01/06/2023]
|
9
|
Tseng KY, Kuo TT, Wang V, Huang EYK, Ma KH, Olson L, Hoffer BJ, Chen YH. Tetrabenazine Mitigates Aberrant Release and Clearance of Dopamine in the Nigrostriatal System, and Alleviates L-DOPA-Induced Dyskinesia in a Mouse Model of Parkinson's Disease. JOURNAL OF PARKINSON'S DISEASE 2022; 12:1545-1565. [PMID: 35599497 DOI: 10.3233/jpd-223195] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/15/2023]
Abstract
BACKGROUND L-DOPA-induced dyskinesia (LID), occurring with aberrant processing of exogenous L-DOPA in the dopamine-denervated striatum, is a main complication of levodopa treatment in Parkinson's disease. OBJECTIVE To characterize the effects of the vesicular antagonist tetrabenazine (TBZ) on L-DOPA-induced behavior, neurochemical signals, and underlying protein expressions in an animal model of Parkinson's disease. METHODS 20-week-old MitoPark mice were co-treated or separately administered TBZ and L-DOPA for 14 days. Abnormal involuntary movements (AIMs) and locomotor activity were analyzed. To explore dopamine (DA) transmission, fast scan cyclic voltammetry was used to assess presynaptic DA dynamics in striatal slices following treatments. PET imaging with 4-[18F]-PE2I, ADAM and immunoblotting assays were used to detect receptor protein changes in the DA-denervated striatum. Finally, nigrostriatal tissues were collected for HPLC measures of DA, serotonin and their metabolites. RESULTS A single injection of TBZ given in the interval between the two L-DOPA/Carbidopa treatments significantly attenuated L-DOPA-induced AIMs expression and locomotor hyperactivity. TBZ was shown to reduce tonic and phasic release of DA following L-DOPA treatment in DA-denervated striatal tissue. In the DA-depleted striatum, TBZ decreased the expression of L-DOPA-enhanced D1 receptors and the serotonin reuptake transporter. Neurochemical analysis indicated that TBZ attenuated L-DOPA-induced surges of DA levels by promoting DA turnover in the nigrostriatal system. CONCLUSIONS Our findings demonstrate that TBZ diminishes abnormal striatal DA transmission, which involves the ability of TBZ to modulate the presymptomatic dynamics of DA, and then mitigate aberrant release of exogenous L-DOPA from nerve terminals. The results support the potential of repositioning TBZ to counteract LID development.
Collapse
Affiliation(s)
- Kuan-Yin Tseng
- National Defense Medical Center, Taipei, Taiwan, R.O.C
- Department of Neurological Surgery, Tri-Service General Hospital, Taipei, Taiwan, R.O.C
| | - Tung-Tai Kuo
- Department of Neurological Surgery, Tri-Service General Hospital, Taipei, Taiwan, R.O.C
| | - Vicki Wang
- Ph.D. Program in Translational Medicine, National Defense Medical Center and Academia Sinica, Taipei, Taiwan
| | - Eagle Yi-Kung Huang
- Department of Pharmacology, National Defense Medical Center, Taipei, Taiwan, R.O.C
| | - Kuo-Hsing Ma
- Graduate Institute of Biology and Anatomy, National Defense Medical Center, Taipei, Taiwan, R.O.C
| | - Lars Olson
- Department of Neuroscience, Karolinska Institute, Stockholm, Sweden
| | - Barry J Hoffer
- Department of Neurosurgery, University Hospitals of Cleveland, Case Western Reserve University School of Medicine, Cleveland, OH, USA
| | - Yuan-Hao Chen
- National Defense Medical Center, Taipei, Taiwan, R.O.C
- Department of Neurological Surgery, Tri-Service General Hospital, Taipei, Taiwan, R.O.C
| |
Collapse
|
10
|
Palermo G, Giannoni S, Bellini G, Siciliano G, Ceravolo R. Dopamine Transporter Imaging, Current Status of a Potential Biomarker: A Comprehensive Review. Int J Mol Sci 2021; 22:11234. [PMID: 34681899 PMCID: PMC8538800 DOI: 10.3390/ijms222011234] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2021] [Revised: 10/12/2021] [Accepted: 10/13/2021] [Indexed: 11/16/2022] Open
Abstract
A major goal of current clinical research in Parkinson's disease (PD) is the validation and standardization of biomarkers enabling early diagnosis, predicting outcomes, understanding PD pathophysiology, and demonstrating target engagement in clinical trials. Molecular imaging with specific dopamine-related tracers offers a practical indirect imaging biomarker of PD, serving as a powerful tool to assess the status of presynaptic nigrostriatal terminals. In this review we provide an update on the dopamine transporter (DAT) imaging in PD and translate recent findings to potentially valuable clinical practice applications. The role of DAT imaging as diagnostic, preclinical and predictive biomarker is discussed, especially in view of recent evidence questioning the incontrovertible correlation between striatal DAT binding and nigral cell or axon counts.
Collapse
Affiliation(s)
- Giovanni Palermo
- Unit of Neurology, Department of Clinical and Experimental Medicine, University of Pisa, 56126 Pisa, Italy; (G.P.); (S.G.); (G.B.); (G.S.)
| | - Sara Giannoni
- Unit of Neurology, Department of Clinical and Experimental Medicine, University of Pisa, 56126 Pisa, Italy; (G.P.); (S.G.); (G.B.); (G.S.)
- Unit of Neurology, San Giuseppe Hospital, 50053 Empoli, Italy
| | - Gabriele Bellini
- Unit of Neurology, Department of Clinical and Experimental Medicine, University of Pisa, 56126 Pisa, Italy; (G.P.); (S.G.); (G.B.); (G.S.)
| | - Gabriele Siciliano
- Unit of Neurology, Department of Clinical and Experimental Medicine, University of Pisa, 56126 Pisa, Italy; (G.P.); (S.G.); (G.B.); (G.S.)
| | - Roberto Ceravolo
- Unit of Neurology, Department of Clinical and Experimental Medicine, University of Pisa, 56126 Pisa, Italy; (G.P.); (S.G.); (G.B.); (G.S.)
- Center for Neurodegenerative Diseases, Unit of Neurology, Parkinson’s Disease and Movement Disorders, Department of Clinical and Experimental Medicine, University of Pisa, 56126 Pisa, Italy
| |
Collapse
|
11
|
Ferrer-Raventós P, Beyer K. Alternative platelet activation pathways and their role in neurodegenerative diseases. Neurobiol Dis 2021; 159:105512. [PMID: 34537329 DOI: 10.1016/j.nbd.2021.105512] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2021] [Revised: 09/08/2021] [Accepted: 09/14/2021] [Indexed: 12/24/2022] Open
Abstract
PURPOSE OF THE REVIEW The study of platelets in the context of neurodegenerative diseases is intensifying, and increasing evidence suggests that platelets may play an important role in the pathogenesis of neurodegenerative disorders. Therefore, we aim to provide a comprehensive overview of the role of platelets and their diverse activation pathways in the development of these diseases. RECENT FINDINGS Platelets participate in synaptic plasticity, learning, memory, and platelets activated by exercise promote neuronal differentiation in several brain regions. Platelets also contribute to the immune response by modulating their surface protein profile and releasing pro- and anti-inflammatory mediators. In Alzheimer's disease, increased levels of platelet amyloid precursor protein raise the production of amyloid-beta peptides promoting platelet activation, triggering at the same time amyloid-beta fibrillation. In Parkinson's disease, increased platelet α-synuclein is associated with elevated ROS production and mitochondrial dysfunction. SUMMARY In this review, we revise different platelet activation pathways, those classically involved in hemostasis and wound healing, and alternative activation pathways recently described in the context of neurodegenerative diseases, especially in Alzheimer's disease.
Collapse
Affiliation(s)
- Paula Ferrer-Raventós
- Memory Unit, Neurology Department and Sant Pau Biomedical Research Institute, Hospital de la Santa Creu i Sant Pau, Universitat Autònoma de Barcelona, Barcelona, Spain; Network Center for Biomedical Research in Neurodegenerative Diseases (CIBERNED), Madrid, Spain
| | - Katrin Beyer
- Department of Pathology, Germans Trias i Pujol Research Institute (IGTP), Universitat Autònoma de Barcelona (UAB), 08916 Badalona, Barcelona, Spain.
| |
Collapse
|
12
|
Maillet A, Météreau E, Tremblay L, Favre E, Klinger H, Lhommée E, Le Bars D, Castrioto A, Prange S, Sgambato V, Broussolle E, Krack P, Thobois S. Serotonergic and Dopaminergic Lesions Underlying Parkinsonian Neuropsychiatric Signs. Mov Disord 2021; 36:2888-2900. [PMID: 34494685 DOI: 10.1002/mds.28722] [Citation(s) in RCA: 32] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2020] [Revised: 05/29/2021] [Accepted: 06/14/2021] [Indexed: 12/28/2022] Open
Abstract
BACKGROUND Parkinson's disease (PD) is characterized by heterogeneous motor and nonmotor manifestations related to alterations in monoaminergic neurotransmission systems. Nevertheless, the characterization of concomitant dopaminergic and serotonergic dysfunction after different durations of Parkinson's disease, as well as their respective involvement in the expression and severity of neuropsychiatric signs, has gained little attention so far. METHODS To fill this gap, we conducted a cross-sectional study combining clinical and dual-tracer positron emission tomography (PET) neuroimaging approaches, using radioligands of dopamine ([11 C]-N-(3-iodoprop-2E-enyl)-2-beta-carbomethoxy-3-beta-(4-methylphenyl)-nortropane) ([11 C]PE2I) and serotonin ([11 C]-N,N-dimethyl-2-(-2-amino-4-cyanophenylthio)-benzylamine) ([11 C]DASB) reuptake, after different durations of Parkinson's disease (ie, in short-disease duration drug-naive de novo (n = 27, 0-2 years-duration), suffering from apathy (n = 14) or not (n = 13); intermediate-disease duration (n = 15, 4-7 years-duration) and long-disease duration, non-demented (n = 15, 8-10 years-duration) patients). Fifteen age-matched healthy subjects were also enrolled. RESULTS The main findings are threefold: (1) both dopaminergic and serotonergic lesions worsen with the duration of Parkinson's disease, spreading from midbrain/subcortical to cortical regions; (2) the presence of apathy at PD onset is associated with more severe cortical and subcortical serotonergic and dopaminergic disruption, similar to the denervation pattern observed in intermediate-disease duration patients; and (3) the severity of parkinsonian apathy, depression, and trait-anxiety appears primarily related to serotonergic alteration within corticostriatal limbic areas. CONCLUSIONS Altogether, these findings highlight the prominent role of serotonergic degeneration in the expression of several neuropsychiatric symptoms occurring after different durations of Parkinson's disease. © 2021 International Parkinson and Movement Disorder Society.
Collapse
Affiliation(s)
- Audrey Maillet
- CNRS, Institut des Sciences Cognitives Marc Jeannerod, UMR 5229 CNRS, Lyon, France.,Université Claude Bernard Lyon I, Lyon, France
| | - Elise Météreau
- CNRS, Institut des Sciences Cognitives Marc Jeannerod, UMR 5229 CNRS, Lyon, France.,Université Claude Bernard Lyon I, Lyon, France.,Hospices Civils de Lyon, Hôpital Neurologique Pierre Wertheimer, Service de Neurologie C, Lyon, France
| | - Léon Tremblay
- CNRS, Institut des Sciences Cognitives Marc Jeannerod, UMR 5229 CNRS, Lyon, France.,Université Claude Bernard Lyon I, Lyon, France
| | - Emilie Favre
- Hospices Civils de Lyon, Hôpital Neurologique Pierre Wertheimer, Service de Neurologie C, Lyon, France.,Genopsy, Centre Hospitalier Le Vinatier, Lyon, France
| | - Hélène Klinger
- Hospices Civils de Lyon, Hôpital Neurologique Pierre Wertheimer, Service de Neurologie C, Lyon, France
| | - Eugénie Lhommée
- Université Grenoble Alpes, Grenoble Institut des Neurosciences, Inserm U1216, Grenoble, France.,Unité des Troubles du Mouvement, Département de Neurologie, CHU de Grenoble, Grenoble, France
| | - Didier Le Bars
- CERMEP, Imagerie du Vivant, Lyon, France.,Institut de Chimie et de Biochimie Moléculaires et Supramoléculaires, UMR 5246 CNRS, Université Claude Bernard Lyon I, Lyon, France
| | - Anna Castrioto
- Université Grenoble Alpes, Grenoble Institut des Neurosciences, Inserm U1216, Grenoble, France.,Unité des Troubles du Mouvement, Département de Neurologie, CHU de Grenoble, Grenoble, France
| | - Stéphane Prange
- CNRS, Institut des Sciences Cognitives Marc Jeannerod, UMR 5229 CNRS, Lyon, France.,Université Claude Bernard Lyon I, Lyon, France.,Faculté de médecine Lyon Sud Charles Mérieux, Univ Lyon, Université Claude Bernard Lyon I, Lyon, France
| | - Véronique Sgambato
- CNRS, Institut des Sciences Cognitives Marc Jeannerod, UMR 5229 CNRS, Lyon, France.,Université Claude Bernard Lyon I, Lyon, France
| | - Emmanuel Broussolle
- CNRS, Institut des Sciences Cognitives Marc Jeannerod, UMR 5229 CNRS, Lyon, France.,Université Claude Bernard Lyon I, Lyon, France.,Faculté de médecine Lyon Sud Charles Mérieux, Univ Lyon, Université Claude Bernard Lyon I, Lyon, France
| | - Paul Krack
- Université Grenoble Alpes, Grenoble Institut des Neurosciences, Inserm U1216, Grenoble, France.,Unité des Troubles du Mouvement, Département de Neurologie, CHU de Grenoble, Grenoble, France.,Department of Neurology, Center for Parkinson's Disease and Movement Disorders, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland
| | - Stéphane Thobois
- CNRS, Institut des Sciences Cognitives Marc Jeannerod, UMR 5229 CNRS, Lyon, France.,Université Claude Bernard Lyon I, Lyon, France.,Faculté de médecine Lyon Sud Charles Mérieux, Univ Lyon, Université Claude Bernard Lyon I, Lyon, France
| |
Collapse
|
13
|
Ionescu TM, Amend M, Hafiz R, Biswal BB, Maurer A, Pichler BJ, Wehrl HF, Herfert K. Striatal and prefrontal D2R and SERT distributions contrastingly correlate with default-mode connectivity. Neuroimage 2021; 243:118501. [PMID: 34428573 DOI: 10.1016/j.neuroimage.2021.118501] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2021] [Revised: 07/23/2021] [Accepted: 08/20/2021] [Indexed: 11/28/2022] Open
Abstract
Although brain research has taken important strides in recent decades, the interaction and coupling of its different physiological levels is still not elucidated. Specifically, the molecular substrates of resting-state functional connectivity (rs-FC) remain poorly understood. The aim of this study was elucidating interactions between dopamine D2 receptors (D2R) and serotonin transporter (SERT) availabilities in the striatum (CPu) and medial prefrontal cortex (mPFC), two of the main dopaminergic and serotonergic projection areas, and the default-mode network. Additionally, we delineated its interaction with two other prominent resting-state networks (RSNs), the salience network (SN) and the sensorimotor network (SMN). To this extent, we performed simultaneous PET/fMRI scans in a total of 59 healthy rats using [11C]raclopride and [11C]DASB, two tracers used to image quantify D2R and SERT respectively. Edge, node and network-level rs-FC metrics were calculated for each subject and potential correlations with binding potentials (BPND) in the CPu and mPFC were evaluated. We found widespread negative associations between CPu D2R availability and all the RSNs investigated, consistent with the postulated role of the indirect basal ganglia pathway. Correlations between D2Rs in the mPFC were weaker and largely restricted to DMN connectivity. Strikingly, medial prefrontal SERT correlated both positively with anterior DMN rs-FC and negatively with rs-FC between and within the SN, SMN and the posterior DMN, underlining the complex role of serotonergic neurotransmission in this region. Here we show direct relationships between rs-FC and molecular properties of the brain as assessed by simultaneous PET/fMRI in healthy rodents. The findings in the present study contribute to the basic understanding of rs-FC by revealing associations between inter-subject variances of rs-FC and receptor and transporter availabilities. Additionally, since current therapeutic strategies typically target neurotransmitter systems with the aim of normalizing brain function, delineating associations between molecular and network-level brain properties is essential and may enhance the understanding of neuropathologies and support future drug development.
Collapse
Affiliation(s)
- Tudor M Ionescu
- Werner Siemens Imaging Center, Department of Preclinical Imaging and Radiopharmacy, Eberhard Karls University Tuebingen, Tuebingen, Germany
| | - Mario Amend
- Werner Siemens Imaging Center, Department of Preclinical Imaging and Radiopharmacy, Eberhard Karls University Tuebingen, Tuebingen, Germany
| | - Rakibul Hafiz
- Department of Biomedical Engineering, New Jersey Institute of Technology, University Heights, Newark, NJ, USA
| | - Bharat B Biswal
- Department of Biomedical Engineering, New Jersey Institute of Technology, University Heights, Newark, NJ, USA
| | - Andreas Maurer
- Werner Siemens Imaging Center, Department of Preclinical Imaging and Radiopharmacy, Eberhard Karls University Tuebingen, Tuebingen, Germany
| | - Bernd J Pichler
- Werner Siemens Imaging Center, Department of Preclinical Imaging and Radiopharmacy, Eberhard Karls University Tuebingen, Tuebingen, Germany
| | - Hans F Wehrl
- Werner Siemens Imaging Center, Department of Preclinical Imaging and Radiopharmacy, Eberhard Karls University Tuebingen, Tuebingen, Germany
| | - Kristina Herfert
- Werner Siemens Imaging Center, Department of Preclinical Imaging and Radiopharmacy, Eberhard Karls University Tuebingen, Tuebingen, Germany.
| |
Collapse
|
14
|
Zheng C, Zhang F. New insights into pathogenesis of l-DOPA-induced dyskinesia. Neurotoxicology 2021; 86:104-113. [PMID: 34331976 DOI: 10.1016/j.neuro.2021.07.006] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2021] [Revised: 07/25/2021] [Accepted: 07/26/2021] [Indexed: 10/20/2022]
Abstract
Parkinson's disease (PD) is a progressive and self-propelling neurodegenerative disorder, which is characterized by motor symptoms, such as rigidity, tremor, slowness of movement and problems with gait. These symptoms become worse over time. To date, Dopamine (DA) replacement therapy with 3, 4-dihydroxy-l-phenylalanine (L-DOPA) is still the most effective pharmacotherapy for motor symptoms of PD. Unfortunately, motor fluctuations consisting of wearing-off effect actions and dyskinesia tend to occur in a few years of starting l-DOPA. Currently, l-DOPA-induced dyskinesia (LID) is troublesome and the pathogenesis of LID requires further investigation. Importantly, a new intervention for LID is imminent. Thus, this review mainly summarized the clinical features, risk factors and pathogenesis of LID to provide updatefor the development of therapeutic targets and new approaches for the treatment of LID.
Collapse
Affiliation(s)
- Changqing Zheng
- Laboratory Animal Center and Key Laboratory of Basic Pharmacology of Ministry of Education and Joint International Research Laboratory of Ethnomedicine of Ministry of Education, Zunyi Medical University, Zunyi, Guizhou, China
| | - Feng Zhang
- Laboratory Animal Center and Key Laboratory of Basic Pharmacology of Ministry of Education and Joint International Research Laboratory of Ethnomedicine of Ministry of Education, Zunyi Medical University, Zunyi, Guizhou, China.
| |
Collapse
|
15
|
Caligiore D, Montedori F, Buscaglione S, Capirchio A. Increasing Serotonin to Reduce Parkinsonian Tremor. Front Syst Neurosci 2021; 15:682990. [PMID: 34354572 PMCID: PMC8331097 DOI: 10.3389/fnsys.2021.682990] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2021] [Accepted: 06/21/2021] [Indexed: 01/07/2023] Open
Abstract
While current dopamine-based drugs seem to be effective for most Parkinson's disease (PD) motor dysfunctions, they produce variable responsiveness for resting tremor. This lack of consistency could be explained by considering recent evidence suggesting that PD resting tremor can be divided into different partially overlapping phenotypes based on the dopamine response. These phenotypes may be associated with different pathophysiological mechanisms produced by a cortical-subcortical network involving even non-dopaminergic areas traditionally not directly related to PD. In this study, we propose a bio-constrained computational model to study the neural mechanisms underlying a possible type of PD tremor: the one mainly involving the serotoninergic system. The simulations run with the model demonstrate that a physiological serotonin increase can partially recover dopamine levels at the early stages of the disease before the manifestation of overt tremor. This result suggests that monitoring serotonin concentration changes could be critical for early diagnosis. The simulations also show the effectiveness of a new pharmacological treatment for tremor that acts on serotonin to recover dopamine levels. This latter result has been validated by reproducing existing data collected with human patients.
Collapse
Affiliation(s)
- Daniele Caligiore
- Computational and Translational Neuroscience Laboratory, Institute of Cognitive Sciences and Technologies, National Research Council, Rome, Italy
| | - Francesco Montedori
- Computational and Translational Neuroscience Laboratory, Institute of Cognitive Sciences and Technologies, National Research Council, Rome, Italy
| | - Silvia Buscaglione
- Neurophysiology and Neuroengineering of Human-Technology Interaction Research Unit (NeXT), Campus Bio-Medico University, Rome, Italy
| | - Adriano Capirchio
- Computational and Translational Neuroscience Laboratory, Institute of Cognitive Sciences and Technologies, National Research Council, Rome, Italy
| |
Collapse
|
16
|
Walker M, Kuebler L, Goehring CM, Pichler BJ, Herfert K. Imaging SERT Availability in a Rat Model of L-DOPA-Induced Dyskinesia. Mol Imaging Biol 2021; 22:634-642. [PMID: 31392531 DOI: 10.1007/s11307-019-01418-2] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
PURPOSE The development of L-DOPA-induced dyskinesia (LID) is one of the most severe side effects of chronic L-DOPA treatment in Parkinson's disease patients. [11C]DASB positron emission tomography (PET) provides a prominent tool to visualize and quantify serotonin transporter (SERT) pathology in vivo in patients and in animal models. To evaluate the effect of chronic L-DOPA treatment on SERT availability in an animal model of LID, we performed a longitudinal PET study. PROCEDURES Rats received a unilateral 6-hydroxydopamine (6-OHDA) lesion, and striatal and extrastriatal SERT expression levels were studied with [11C]DASB, a marker of SERT availability, before and after daily treatment with L-DOPA. Dyskinesias were evaluated at different time points over a period of 21 days. RESULTS [11C]DASB binding was found to be decreased after 6-OHDA lesions in the striatum, cortex, and hippocampus 5 weeks after 6-OHDA injection in the lesioned hemisphere of the rat brain. Chronic L-DOPA priming resulted in a relative preservation of SERT availability in the lesioned and healthy hemisphere compared to baseline measurements. CONCLUSIONS Our longitudinal PET data support a preservation of SERT availability after the induction of L-DOPA-induced dyskinesia, which is in line with previous reports in dyskinetic PD patients.
Collapse
Affiliation(s)
- Michael Walker
- Werner Siemens Imaging Center, Department of Preclinical Imaging and Radiopharmacy, Eberhard-Karls University of Tuebingen, Röntgenweg 13, Tuebingen, Germany
| | - Laura Kuebler
- Werner Siemens Imaging Center, Department of Preclinical Imaging and Radiopharmacy, Eberhard-Karls University of Tuebingen, Röntgenweg 13, Tuebingen, Germany
| | - Chris Marc Goehring
- Werner Siemens Imaging Center, Department of Preclinical Imaging and Radiopharmacy, Eberhard-Karls University of Tuebingen, Röntgenweg 13, Tuebingen, Germany
| | - Bernd J Pichler
- Werner Siemens Imaging Center, Department of Preclinical Imaging and Radiopharmacy, Eberhard-Karls University of Tuebingen, Röntgenweg 13, Tuebingen, Germany
| | - Kristina Herfert
- Werner Siemens Imaging Center, Department of Preclinical Imaging and Radiopharmacy, Eberhard-Karls University of Tuebingen, Röntgenweg 13, Tuebingen, Germany.
| |
Collapse
|
17
|
Labrador-Espinosa MA, Grothe MJ, Macías-García D, Jesús S, Adarmes-Gómez A, Muñoz-Delgado L, Fernández-Rodríguez P, Martín-Rodríguez JF, Huertas I, García-Solís D, Mir P. Levodopa-Induced Dyskinesia in Parkinson Disease Specifically Associates With Dopaminergic Depletion in Sensorimotor-Related Functional Subregions of the Striatum. Clin Nucl Med 2021; 46:e296-e306. [PMID: 33782308 DOI: 10.1097/rlu.0000000000003609] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
PURPOSE To determine whether the development of levodopa-induced dyskinesia (LID) in Parkinson disease (PD) specifically relates to dopaminergic depletion in sensorimotor-related subregions of the striatum. METHODS Our primary study sample consisted of 185 locally recruited PD patients, of which 73 (40%) developed LID. Retrospective 123I-FP-CIT SPECT data were used to quantify the specific dopamine transporter (DAT) binding ratio within distinct functionally defined striatal subregions related to limbic, executive, and sensorimotor systems. Regional DAT levels were contrasted between patients who developed LID (PD + LID) and those who did not (PD-LID) using analysis of covariance models controlled for demographic and clinical features. For validation of the findings and assessment of the evolution of LID-associated DAT changes from an early disease stage, we also studied serial 123I-FP-CIT SPECT data from 343 de novo PD patients enrolled in the Parkinson Progression Marker's Initiative using mixed linear model analysis. RESULTS Compared with PD-LID, DAT level reductions in PD + LID patients were most pronounced in the sensorimotor striatal subregion (F = 5.99, P = 0.016) and also significant in the executive-related subregion (F = 5.30, P = 0.023). In the Parkinson Progression Marker's Initiative cohort, DAT levels in PD + LID (n = 161, 47%) were only significantly reduced compared with PD-LID in the sensorimotor striatal subregion (t = -2.05, P = 0.041), and this difference was already present at baseline and remained largely constant over time. CONCLUSION Measuring DAT depletion in functionally defined sensorimotor-related striatal regions of interest may provide a more sensitive tool to detect LID-associated dopaminergic changes at an early disease stage and could improve individual prognosis of this common clinical complication in PD.
Collapse
Affiliation(s)
| | | | | | | | | | - Laura Muñoz-Delgado
- Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), Madrid
| | | | | | - Ismael Huertas
- From the Unidad de Trastornos del Movimiento, Servicio de Neurología y Neurofisiología Clínica, Instituto de Biomedicina de Sevilla, Hospital Universitario Virgen del Rocío/CSIC/Universidad de Sevilla, Seville
| | | | | |
Collapse
|
18
|
Fabbrini A, Guerra A. Pathophysiological Mechanisms and Experimental Pharmacotherapy for L-Dopa-Induced Dyskinesia. J Exp Pharmacol 2021; 13:469-485. [PMID: 33953618 PMCID: PMC8092630 DOI: 10.2147/jep.s265282] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2021] [Accepted: 03/30/2021] [Indexed: 12/21/2022] Open
Abstract
L-dopa-induced dyskinesia (LID) is the most frequent motor complication associated with chronic L-dopa treatment in Parkinson’s disease (PD). Recent advances in the understanding of the pathophysiological mechanisms underlying LID suggest that abnormalities in multiple neurotransmitter systems, in addition to dopaminergic nigrostriatal denervation and altered dopamine release and reuptake dynamics at the synaptic level, are involved in LID development. Increased knowledge of neurobiological LID substrates has led to the development of several drug candidates to alleviate this motor complication. However, with the exception of amantadine, none of the pharmacological therapies tested in humans have demonstrated clinically relevant beneficial effects. Therefore, LID management is still one of the most challenging problems in the treatment of PD patients. In this review, we first describe the known pathophysiological mechanisms of LID. We then provide an updated report of experimental pharmacotherapies tested in clinical trials of PD patients and drugs currently under study to alleviate LID. Finally, we discuss available pharmacological LID treatment approaches and offer our opinion of possible issues to be clarified and future therapeutic strategies.
Collapse
Affiliation(s)
- Andrea Fabbrini
- Department of Human Neurosciences, Sapienza University of Rome, Rome, Italy
| | | |
Collapse
|
19
|
Serotonin/dopamine interaction in the induction and maintenance of L-DOPA-induced dyskinesia: An update. PROGRESS IN BRAIN RESEARCH 2021; 261:287-302. [PMID: 33785132 DOI: 10.1016/bs.pbr.2021.01.032] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Ample evidence suggests that the serotonergic system plays a major role in several aspects of Parkinson's disease. In this review, we focus on the interplay between dopamine and serotonin in the appearance of L-DOPA-induced dyskinesia (LID), the most troublesome side effect of L-DOPA therapy. Indeed, while this drug exerts significant amelioration of motor symptoms during the first few years of treatment, eventually, most of patients experience dyskinesias, which limit the use of L-DOPA in advanced stages of disease. Here, we present the mechanisms underlying LID and the role of serotonin neurons, review preclinical and clinical data, and discuss possible therapeutic strategies.
Collapse
|
20
|
Frey KA, Bohnen NILJ. Molecular Imaging of Neurodegenerative Parkinsonism. PET Clin 2021; 16:261-272. [PMID: 33589385 DOI: 10.1016/j.cpet.2020.12.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
Abstract
Advances in molecular PET imaging of neurodegenerative parkinsonism are reviewed with focus on neuropharmacologic radiotracers depicting terminals of selectively vulnerable neurons in these conditions. Degeneration and losses of dopamine, norepinephrine, serotonin, and acetylcholine imaging markers thus far do not differentiate among the parkinsonian conditions. Recent studies performed with [18F]fluorodeoxyglucose PET are limited by the need for automated image analysis tools and by lack of routine coverage for this imaging indication in the United States. Ongoing research engages use of novel molecular modeling and in silico methods for design of imaging ligands targeting these specific proteinopathies.
Collapse
Affiliation(s)
- Kirk A Frey
- Department of Radiology (Nuclear Medicine and Molecular Imaging), University of Michigan, 1500 East Medical Center Drive, Room B1-G505 UH, Ann Arbor, MI 48109-5028, USA; Department of Neurology, University of Michigan, 1500 East Medical Center Drive, Room B1-G505 UH, Ann Arbor, MI 48109-5028, USA.
| | - Nicolaas I L J Bohnen
- Department of Radiology (Nuclear Medicine and Molecular Imaging), University of Michigan, 24 Frank Lloyd Wright Drive, Box 362, Ann Arbor, MI 48105, USA; Department of Neurology, University of Michigan, 24 Frank Lloyd Wright Drive, Box 362, Ann Arbor, MI 48105, USA; Ann Arbor Veterans Administration Medical Center, Ann Arbor, MI, USA
| |
Collapse
|
21
|
de Natale ER, Wilson H, Politis M. Serotonergic imaging in Parkinson's disease. PROGRESS IN BRAIN RESEARCH 2021; 261:303-338. [PMID: 33785134 DOI: 10.1016/bs.pbr.2020.11.001] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Parkinson's disease (PD) is a neurodegenerative disorder characterized by the progressive degeneration of monoaminergic central pathways such as the serotonergic. The degeneration of serotonergic signaling in striatal and extrastriatal brain regions is an early feature of PD and is associated with several motor and non-motor complications of the disease. Molecular imaging techniques with Positron Emission Tomography (PET) have greatly contributed to the investigation of biological changes in vivo and to the understanding of the extent of serotonergic pathology in patients or individuals at risk for PD. Such discoveries provide with opportunities for the identification of new targets that can be used for the development of novel disease-modifying drugs or symptomatic treatments. Future studies of imaging serotonergic molecular targets will better clarify the importance of serotonergic pathology in PD, including progression of pathology, target-identification for pharmacotherapy, and relevance to endogenous synaptic serotonin levels. In this article, we review the current status and understanding of serotonergic imaging in PD.
Collapse
Affiliation(s)
| | - Heather Wilson
- Neurodegeneration Imaging Group, University of Exeter Medical School, London, United Kingdom
| | - Marios Politis
- Neurodegeneration Imaging Group, University of Exeter Medical School, London, United Kingdom.
| |
Collapse
|
22
|
Hinkle JT, Perepezko K, Gonzalez LL, Mills KA, Pontone GM. Apathy and Anxiety in De Novo Parkinson's Disease Predict the Severity of Motor Complications. Mov Disord Clin Pract 2021; 8:76-84. [PMID: 33426161 DOI: 10.1002/mdc3.13117] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2020] [Revised: 10/09/2020] [Accepted: 10/27/2020] [Indexed: 11/11/2022] Open
Abstract
Background Neuropsychiatric and affective symptoms are prevalent in prodromal and clinical Parkinson's disease (PD). Some evidence suggests that they may also signify risk for motor complications (motor fluctuations and dyskinesias) of dopamine replacement therapy (DRT). Objective To determine whether neuropsychiatric symptoms present in de novo PD (ie, before DRT initiation) predict the severity of eventual motor complications of DRT. Methods We used clinical, demographic, neurobehavioral, and neuroimaging data from the Parkinson's Progression Markers Initiative (PPMI), a multicenter observational PD study. Participants were unmedicated at enrollment and 361 initiated DRT during PPMI follow-up. We used Cox proportional hazard and multivariate ordinal mixed-effects regression models to evaluate the relationship between baseline neuropsychiatric symptoms and motor complications as measured by the Movement Disorders Society-revised Unified Parkinson's Disease Rating Scale (MDS-UPDRS). Results The cumulative incidences of dyskinesias and motor fluctuations during follow-up (6.0 ± 1.5 years) were 34.3% and 59.9%, respectively. Both apathy and high trait-anxiety (top quartile) conveyed over two-fold increases in hazard for dyskinesia onset and for adverse impact on activities of daily living caused by both dyskinesias and motor fluctuations. The longitudinal severity of motor fluctuations and dyskinesias was significantly predicted by baseline trait-anxiety and apathy, but not depression. Models were adjusted for dimensionally related symptoms (eg autonomic dysfunction) and potential confounding variables (eg DRT dose). Conclusions Apathy and anxiety levels in de novo PD may be neuropsychiatric biomarkers of vulnerability to earlier and more disabling motor complications of DRT.
Collapse
Affiliation(s)
- Jared T Hinkle
- Medical Scientist Training Program Johns Hopkins University School of Medicine Baltimore Maryland USA
| | - Kate Perepezko
- Department of Psychiatry Johns Hopkins University School of Medicine Baltimore Maryland USA.,Department of Mental Health Johns Hopkins Bloomberg School of Public Health Baltimore Maryland USA
| | - Lorenzo L Gonzalez
- Department of Psychiatry Johns Hopkins University School of Medicine Baltimore Maryland USA
| | - Kelly A Mills
- Department of Neurology Johns Hopkins University School of Medicine Baltimore Maryland USA
| | - Gregory M Pontone
- Department of Psychiatry Johns Hopkins University School of Medicine Baltimore Maryland USA.,Department of Neurology Johns Hopkins University School of Medicine Baltimore Maryland USA
| |
Collapse
|
23
|
Olanow CW, Calabresi P, Obeso JA. Continuous Dopaminergic Stimulation as a Treatment for Parkinson's Disease: Current Status and Future Opportunities. Mov Disord 2020; 35:1731-1744. [DOI: 10.1002/mds.28215] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2020] [Revised: 06/05/2020] [Accepted: 06/15/2020] [Indexed: 12/21/2022] Open
Affiliation(s)
- C. Warren Olanow
- Department of Neurology and Department of Neuroscience Mount Sinai School of Medicine New York New York USA
- Clintrex Research Corporation Sarasota Florida USA
| | - Paolo Calabresi
- Neurology Fondazione Policlinico Universitario Agostino Gemelli IRCCS Rome Italy
- Dipartimento Neuroscienze Università Cattolica del Sacro Cuore Rome Italy
| | - Jose A. Obeso
- CINAC, Hospital Universitario HM Puerta del Sur, Universidad CEU‐San Pablo Móstoles Madrid Spain
- CIBERNED, Instituto de Salud Carlos III Madrid Spain
| |
Collapse
|
24
|
Porter E, Roussakis AA, Lao-Kaim NP, Piccini P. Multimodal dopamine transporter (DAT) imaging and magnetic resonance imaging (MRI) to characterise early Parkinson's disease. Parkinsonism Relat Disord 2020; 79:26-33. [PMID: 32861103 DOI: 10.1016/j.parkreldis.2020.08.010] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/21/2019] [Revised: 08/05/2020] [Accepted: 08/08/2020] [Indexed: 01/12/2023]
Abstract
Idiopathic Parkinson's disease (PD), the second most common neurodegenerative disorder, is characterised by the progressive loss of dopaminergic nigrostriatal terminals. Currently, in early idiopathic PD, dopamine transporter (DAT)-specific imaging assesses the extent of striatal dopaminergic deficits, and conventional magnetic resonance imaging (MRI) of the brain excludes the presence of significant ischaemic load in the basal ganglia as well as signs indicative of other forms of Parkinsonism. In this article, we discuss the use of multimodal DAT-specific and MRI protocols for insight into the early pathological features of idiopathic PD, including: structural MRI, diffusion tensor imaging, nigrosomal iron imaging and neuromelanin-sensitive MRI sequences. These measures may be acquired serially or simultaneously in a hybrid scanner. From current evidence, it appears that both nigrosomal iron imaging and neuromelanin-sensitive MRI combined with DAT-specific imaging are useful to assist clinicians in diagnosing PD, while conventional structural MRI and diffusion tensor imaging protocols are better suited to a research context focused on characterising early PD pathology. We believe that in the future multimodal imaging will be able to characterise prodromal PD and stratify the clinical stages of PD progression.
Collapse
Affiliation(s)
- Eleanor Porter
- Imperial College London, Hammersmith Hospital, Neurology Imaging Unit, London, UK
| | | | - Nicholas P Lao-Kaim
- Imperial College London, Hammersmith Hospital, Neurology Imaging Unit, London, UK
| | - Paola Piccini
- Imperial College London, Hammersmith Hospital, Neurology Imaging Unit, London, UK.
| |
Collapse
|
25
|
Kong Y, Zhang C, Liu K, Wagle Shukla A, Sun B, Guan Y. Imaging of dopamine transporters in Parkinson disease: a meta-analysis of 18 F/ 123 I-FP-CIT studies. Ann Clin Transl Neurol 2020; 7:1524-1534. [PMID: 32794655 PMCID: PMC7480930 DOI: 10.1002/acn3.51122] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2020] [Revised: 05/31/2020] [Accepted: 06/05/2020] [Indexed: 12/12/2022] Open
Abstract
OBJECTIVE 18 F-FP-CIT and 123 I-FP-CIT are widely used radiotracers in molecular imaging for Parkinson's disease (PD) diagnosis. Compared with 123 I-FP-CIT, 18 F-FP-CIT has superior tracer kinetics. We aimed to conduct a meta-analysis to assess the efficacy of using 18 F-FP-CIT positron emission tomography (PET) and 123 I-FP-CIT single-photon emission computed tomography (SPECT) of dopamine transporters in patients with PD in order to provide evidence for clinical decision-making. METHODS We searched the PubMed, Embase, Wanfang Data, and China National Knowledge Infrastructure databases to identify the relevant studies from the time of inception of the databases to 30 April 2020. We identified six PET studies, including 779 patients with PD and 124 healthy controls, which met the inclusion criteria. Twenty-seven SPECT studies with 1244 PD patients and 859 controls were also included in this meta-analysis. RESULTS Overall effect-size analysis indicated that patients with PD showed significantly reduced 18 F-FP-CIT uptake in three brain regions [caudate nucleus: standardized mean difference (SMD) = -1.71, Z = -3.31, P = 0.0009; anterior putamen: SMD = -3.71, Z = -6.26, P < 0.0001; and posterior putamen: SMD = -5.49, Z = -5.97, P < 0.0001]. Significant decreases of 123 I-FP-CIT uptake were also observed in the caudate (SMD = -2.31, Z = -11.49, P < 0.0001) and putamen (SMD = -3.25, Z = -14.79, P < 0.0001). INTERPRETATION In conclusion, our findings indicate that both 18 F-FP-CIT PET and 123 I-FP-CIT SPECT imaging of dopamine transporters can provide viable biomarkers for early PD diagnosis.
Collapse
Affiliation(s)
- Yanyan Kong
- Department of Neurosurgery, Center for Functional Neurosurgery, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China.,PET Center, Huashan Hospital, Fudan University, Shanghai, 200235, China
| | - Chencheng Zhang
- Department of Neurosurgery, Center for Functional Neurosurgery, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Kawai Liu
- Department of Mathematics, The Shanghai SMIC Private School, Shanghai, 200000, China
| | - Aparna Wagle Shukla
- Department of Neurology and Fixel Center for Neurological Diseases and the Program for Movement Disorders and Neurorestoration, University of Florida, Gainesville, FL 32611
| | - Bomin Sun
- Department of Neurosurgery, Center for Functional Neurosurgery, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Yihui Guan
- PET Center, Huashan Hospital, Fudan University, Shanghai, 200235, China
| |
Collapse
|
26
|
Marin C, Bonastre M, Fuentes M, Mullol J. Lack of correlation between dyskinesia and pallidal serotonin transporter expression-induced by L-Dopa and Pramipexole in hemiparkinsonian rats. Pharmacol Biochem Behav 2020; 197:173012. [PMID: 32750392 DOI: 10.1016/j.pbb.2020.173012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/21/2020] [Revised: 07/30/2020] [Accepted: 07/31/2020] [Indexed: 11/30/2022]
Abstract
The role of pallidal serotonergic terminals in the development of L-Dopa-induced dyskinesias (LIDs) in Parkinson's disease (PD) has been recently highlighted correlating pallidal serotonin transporter (SERT) expression levels with dyskinesias severity. However, the role of external globus pallidus (GPe, GP in rodents) serotonergic function in LIDs is still controversial since several studies have shown no differences in GPe serotonin (SER) and SERT levels between dyskinetic and non-dyskinetic PD patients. In addition, the increase in pallidal SERT/dopamine transporter (DAT) binding ratio obtained in positron emission tomography studies has been shown similar in both subtypes of PD patients. Based on these controversial results, further studies are required to clarify the possible involvement of GPe serotonergic activity in LIDs expression. We investigated the pallidal SER and SERT expression changes and the abnormal involuntary movements (AIMs) induced by L-Dopa or the D3/D2 dopamine (DA) agonist, Pramipexole, in partial unilateral 6-hydroxydopamine (6-OHDA)-lesioned rats. L-Dopa treatment led to an increment of axial (p < 0.01), limb (p < 0.01), and orolingual (p < 0.01) AIMs. However, Pramipexole treatment did not induce AIMs. The number of GP SERT-positive axon varicosities was increased in L-Dopa (p < 0.05) and Pramipexole (p < 0.01) treated rats. No differences were observed in the number of GP SERT-positive varicosities between L-Dopa and Pramipexole treatments. Our results indicate a lack of correlation between GP SERT expression levels and the development of AIMs suggesting that pallidal serotonergic fibers are not responsible for LIDs. The possible involvement of the SER system in dyskinesia may include other mechanisms.
Collapse
Affiliation(s)
- Concepció Marin
- INGENIO, IRCE, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Catalonia, Spain.
| | - Mercè Bonastre
- INGENIO, IRCE, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Catalonia, Spain
| | - Mireya Fuentes
- INGENIO, IRCE, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Catalonia, Spain
| | - Joaquim Mullol
- INGENIO, IRCE, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Catalonia, Spain.
| |
Collapse
|
27
|
Jankovic J, Okun MS, Kordower JH. Stem Cells: Scientific and Ethical Quandaries of a Personalized Approach to Parkinson's Disease. Mov Disord 2020; 35:1312-1314. [DOI: 10.1002/mds.28187] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2020] [Revised: 06/05/2020] [Accepted: 06/08/2020] [Indexed: 12/30/2022] Open
Affiliation(s)
- Joseph Jankovic
- Parkinson's Disease Center and Movement Disorders Clinic, Department of Neurology Baylor College of Medicine Houston Texas USA
| | - Michael S. Okun
- Norman Fixel Institute for Neurological Diseases, Department of Neurology University of Florida Health Gainesville Florida USA
| | - Jeffrey H. Kordower
- Department of Neurological Sciences Rush University Medical Center Chicago Illinois USA
| |
Collapse
|
28
|
A Retrospective Imaging Evaluation of Presynaptic Dopaminergic Degeneration in Multiple System Atrophy with Levodopa Induced Dyskinesia. Tremor Other Hyperkinet Mov (N Y) 2020; 10:6. [PMID: 32775020 PMCID: PMC7394229 DOI: 10.5334/tohm.58] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Background: Multiple system atrophy (MSA) may develop levodopa-induced dyskinesia, which is dystonic and predominant in the orofacial region. We aimed to characterize the patterns of presynaptic dopaminergic degeneration in patients with MSA and dyskinesia using 123I-N-x-fluoropropyl-2b-carbo-methoxy-3b-(4-iodophenyl) nortropan single-photon emission computed tomography (123I-FP-CIT SPECT). Methods: A single center cross-sectional retrospective study was conducted using consecutive chart review of patients with probable MSA who underwent 123I-FP-CIT SPECT. The degeneration patterns were compared between the groups with and without dyskinesia via visual assessment of 123I-FP-CIT SPECT images. Results: Twenty-five patients with probable MSA who had undergone dopamine transporter imaging were identified (age [mean ± standard error], 62.5 ± 1.7 years; disease duration, 48.8 ± 7.0 months). Four of them presented dyskinesia and 21 of patients did not. Twenty-five patients with MSA were visually classified into five grades: one Grade 1 (normal), two Grade 2 (eagle wing), three Grade 3 (mixed), nine Grade 4 (egg shape), and ten Grade 5 (burst striatum). All patients with MSA and dyskinesia were classified into Grade 5. Visual grading significantly correlated with disease duration and levodopa responsiveness. Conclusions: Severe presynaptic dopaminergic dysfunction in 123I-FP-CIT SPECT images, higher doses of dopaminergic medication, and longer disease durations were associated with occurrence of levodopa-induced dyskinesia, even in MSA.
Collapse
|
29
|
Fisher R, Hikima A, Morris R, Jackson MJ, Rose S, Varney MA, Depoortere R, Newman-Tancredi A. The selective 5-HT 1A receptor agonist, NLX-112, exerts anti-dyskinetic and anti-parkinsonian-like effects in MPTP-treated marmosets. Neuropharmacology 2020; 167:107997. [PMID: 32057799 PMCID: PMC7103782 DOI: 10.1016/j.neuropharm.2020.107997] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2019] [Revised: 02/03/2020] [Accepted: 02/09/2020] [Indexed: 11/16/2022]
Abstract
l-DOPA is the gold-standard pharmacotherapy for treatment of Parkinson's disease (PD) but can lead to the appearance of troubling dyskinesia which are attributable to 'false neurotransmitter' release of dopamine by serotonergic neurons. Reducing the activity of these neurons diminishes l-DOPA-induced dyskinesia (LID), but there are currently no clinically approved selective, high efficacy 5-HT1A receptor agonists. Here we describe the effects of NLX-112, a highly selective and efficacious 5-HT1A receptor agonist, on LID in 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP)-treated marmosets, a non-human primate model of PD. NLX-112 exhibited modest plasma half-life (~2h) and marked plasma protein binding (96%). When administered to parkinsonian marmosets with l-DOPA (7 mg/kg p.o.), NLX-112 (0.025, 0.1 and 0.4 mg/kg p.o.) reduced LID scores at early time-points after administration, whilst only minimally interfering with the l-DOPA-induced reversal of motor disability. In contrast, the prototypical 5-HT1A receptor agonist, (+)8-OH-DPAT (0.6 and 2 mg/kg p. o.), reduced LID but also abolished l-DOPA's anti-disability activity. Administered by itself, NLX-112 (0.1, 0.2 mg/kg p.o.) produced very little dyskinesia or locomotor activity, but reduced motor disability scores by about half the extent elicited by l-DOPA, suggesting that it may have motor facilitation effects of its own. Both NLX-112 and (+)8-OH-DPAT induced unusual and dose-limiting behaviors in marmoset that resembled 'serotonin behavioral syndrome' observed previously in rat. Overall, the present study showed that NLX-112 has anti-LID activity at the doses tested as well as reducing motor disability. The data suggest that additional investigation of NLX-112 is desirable to explore its potential as a treatment for PD and PD-LID.
Collapse
Affiliation(s)
- Ria Fisher
- Faculty of Life Sciences and Medicine, Hodgkin Building, King's College London, Guy's Campus, London, SE1 1UL, UK
| | - Atsuko Hikima
- Faculty of Life Sciences and Medicine, Hodgkin Building, King's College London, Guy's Campus, London, SE1 1UL, UK
| | - Rebecca Morris
- Faculty of Life Sciences and Medicine, Hodgkin Building, King's College London, Guy's Campus, London, SE1 1UL, UK
| | - Michael J Jackson
- Faculty of Life Sciences and Medicine, Hodgkin Building, King's College London, Guy's Campus, London, SE1 1UL, UK
| | - Sarah Rose
- Faculty of Life Sciences and Medicine, Hodgkin Building, King's College London, Guy's Campus, London, SE1 1UL, UK
| | - Mark A Varney
- Neurolixis SAS, 2 Rue Georges Charpak, 81100, Castres, France
| | | | | |
Collapse
|
30
|
Aljuaid M, Booth S, Hobson DE, Borys A, Williams K, Katako A, Ryner L, Goertzen AL, Ko JH. Blood Flow and Glucose Metabolism Dissociation in the Putamen Is Predictive of Levodopa Induced Dyskinesia in Parkinson's Disease Patients. Front Neurol 2019; 10:1217. [PMID: 31824400 PMCID: PMC6881455 DOI: 10.3389/fneur.2019.01217] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2019] [Accepted: 11/01/2019] [Indexed: 12/28/2022] Open
Abstract
Background: The forefront treatment of Parkinson's disease (PD) is Levodopa. When patients are treated with Levodopa cerebral blood flow is increased while cerebral metabolic rate is decreased in key subcortical regions including the putamen. This phenomenon is especially pronounced in patients with Levodopa-induced dyskinesia (LID). Method: To study the effect of clinically-determined anti-parkinsonian medications, 10 PD patients (5 with LID and 5 without LID) have been scanned with FDG-PET (a probe for glucose metabolism) and perfusion MRI (a probe for cerebral blood flow) both when they are ON and OFF medications. Patients additionally underwent resting state fMRI to detect changes in dopamine-mediated cortico-striatal connectivity. The degree of blood flow-glucose metabolism dissociation was quantified by comparing the FDG-PET and perfusion MRI data. Results: A significant interaction effect (imaging modality × medication; blood flow-glucose metabolism dissociation) has been found in the putamen (p = 0.023). Post-hoc analysis revealed that anti-parkinsonian medication consistently normalized the pathologically hyper-metabolic state of the putamen while mixed effects were observed in cerebral blood flow changes. This dissociation was especially predominant in patients with LID compared to those without. Unlike the prior study, this differentiation was not observed when cortico-striatal functional connectivity was assessed. Conclusion: We confirmed striatal neurovascular dissociation between FDG-PET and perfusion MRI in response to clinically determined anti-parkinsonian medication. We further proposed a novel analytical method to quantify the degree of dissociation in the putamen using only the ON condition scans, Putamen-to-thalamus Hyper-perfusion/hypo-metabolism Index (PHI), which may have the potential to be used as a biomarker for LID (correctly classifying 8 out 10 patients). For wider use of PHI, a larger validation study is warranted.
Collapse
Affiliation(s)
- Maram Aljuaid
- Department of Human Anatomy and Cell Science, Rady Faculty of Health Sciences, Max Rady College of Medicine, University of Manitoba, Winnipeg, MB, Canada.,Neuroscience Research Program, Kleysen Institute for Advanced Medicine, Health Sciences Centre, Winnipeg, MB, Canada
| | - Samuel Booth
- Department of Human Anatomy and Cell Science, Rady Faculty of Health Sciences, Max Rady College of Medicine, University of Manitoba, Winnipeg, MB, Canada.,Neuroscience Research Program, Kleysen Institute for Advanced Medicine, Health Sciences Centre, Winnipeg, MB, Canada
| | - Douglas E Hobson
- Section of Neurology, Department of Internal Medicine, Rady Faculty of Health Sciences, Max Rady College of Medicine, University of Manitoba, Winnipeg, MB, Canada
| | - Andrew Borys
- Section of Neurology, Department of Internal Medicine, Rady Faculty of Health Sciences, Max Rady College of Medicine, University of Manitoba, Winnipeg, MB, Canada
| | - Kelly Williams
- Section of Neurology, Department of Internal Medicine, Rady Faculty of Health Sciences, Max Rady College of Medicine, University of Manitoba, Winnipeg, MB, Canada
| | - Audrey Katako
- Department of Human Anatomy and Cell Science, Rady Faculty of Health Sciences, Max Rady College of Medicine, University of Manitoba, Winnipeg, MB, Canada.,Neuroscience Research Program, Kleysen Institute for Advanced Medicine, Health Sciences Centre, Winnipeg, MB, Canada
| | - Lawrence Ryner
- Department of Radiology, Rady Faculty of Health Sciences, Max Rady College of Medicine, University of Manitoba, Winnipeg, MB, Canada
| | - Andrew L Goertzen
- Department of Radiology, Rady Faculty of Health Sciences, Max Rady College of Medicine, University of Manitoba, Winnipeg, MB, Canada
| | - Ji Hyun Ko
- Department of Human Anatomy and Cell Science, Rady Faculty of Health Sciences, Max Rady College of Medicine, University of Manitoba, Winnipeg, MB, Canada.,Neuroscience Research Program, Kleysen Institute for Advanced Medicine, Health Sciences Centre, Winnipeg, MB, Canada
| |
Collapse
|
31
|
Meadows SM, Conti MM, Gross L, Chambers NE, Avnor Y, Ostock CY, Lanza K, Bishop C. Diverse serotonin actions of vilazodone reduce l-3,4-dihidroxyphenylalanine-induced dyskinesia in hemi-parkinsonian rats. Mov Disord 2019; 33:1740-1749. [PMID: 30485908 DOI: 10.1002/mds.100] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2018] [Revised: 06/08/2018] [Accepted: 06/28/2018] [Indexed: 12/19/2022] Open
Abstract
BACKGROUND The serotonergic system is a well-established modulator of l-dopa-induced dyskinesia. To date, targeting serotonin transporters or serotonin receptor subtype 1A (5-HT1A ) reduces l-dopa-induced dyskinesia in animal models; however, these strategies have failed to translate clinically. Ideally, a compound acting at both known antidyskinetic sites could optimize serotonin-mediated approaches. Vilazodone is a selective serotonin reuptake inhibitor and a partial 5-HT1A agonist approved by the U.S. Food and Drug Administration, situating Vilazodone in a unique position to reduce l-dopa-induced dyskinesia without compromising l-dopa-mediated motor improvements. OBJECTIVES The goal of the present study was to characterize Vilazodone's effects on l-dopa-induced behaviors, neurochemistry and gene expression in unilateral 6-hydroxydopamine-lesioned hemi-parkinsonian rats. METHODS In experiments 1 and 2, l-dopa-naïve and l-dopa-primed animals were coadministered Vilazodone and l-dopa daily for 3 weeks to model subchronic use, and behavioral, neurochemical, and messenger RNA (mRNA) expression changes were measured. In experiment 3, dyskinetic behavior was assessed following 5-HT1A or serotonin receptor subtype 1B blockade prior to Vilazodone-l-dopa coadministration. RESULTS Vilazodone significantly suppressed developing and established l-dopa-induced dyskinesia without compromising the promotor effects of l-dopa therapy. In the dopamine-depleted striatum, Vilazodone-l-dopa cotreatment increased dopamine content, suggesting a normalization of dopamine kinetics in dyskinetic brain, and reduced l-dopa-induced c-Fos and preprodynorphin mRNA overexpression, indicative of attenuated dopamine D1 receptor-mediated direct pathway overactivity. Only 5-HT1A antagonism partially attenuated Vilazodone's antidyskinetic efficacy, suggesting both serotonin transporter-dependent effects and 5-HT1A receptors in Vilazodone's actions. CONCLUSIONS Our findings show Vilazodone has a serotonin-dependent effect on rodent l-dopa-induced dyskinesia and implicate the potential for repositioning Vilazodone against l-dopa-induced dyskinesia development and expression in Parkinson's disease patients. © 2018 International Parkinson and Movement Disorder Society.
Collapse
Affiliation(s)
- Samantha M Meadows
- Behavioral Neuroscience Program, Department of Psychology, Binghamton University, Binghamton, New York, USA
| | - Melissa M Conti
- Behavioral Neuroscience Program, Department of Psychology, Binghamton University, Binghamton, New York, USA
| | - Libby Gross
- Behavioral Neuroscience Program, Department of Psychology, Binghamton University, Binghamton, New York, USA
| | - Nicole E Chambers
- Behavioral Neuroscience Program, Department of Psychology, Binghamton University, Binghamton, New York, USA
| | - Yarden Avnor
- Behavioral Neuroscience Program, Department of Psychology, Binghamton University, Binghamton, New York, USA
| | - Corinne Y Ostock
- Behavioral Neuroscience Program, Department of Psychology, Binghamton University, Binghamton, New York, USA
| | - Kathryn Lanza
- Behavioral Neuroscience Program, Department of Psychology, Binghamton University, Binghamton, New York, USA
| | - Christopher Bishop
- Behavioral Neuroscience Program, Department of Psychology, Binghamton University, Binghamton, New York, USA
| |
Collapse
|
32
|
Okuzumi A, Hatano T, Kamagata K, Hori M, Mori A, Oji Y, Taniguchi D, Daida K, Shimo Y, Yanagisawa N, Nojiri S, Aoki S, Hattori N. Neuromelanin or DaT-SPECT: which is the better marker for discriminating advanced Parkinson's disease? Eur J Neurol 2019; 26:1408-1416. [PMID: 31136060 PMCID: PMC6851628 DOI: 10.1111/ene.14009] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2018] [Accepted: 05/23/2019] [Indexed: 01/10/2023]
Abstract
Background and purpose Whether the neuromelanin‐positive substantia nigra pars compacta area (NM‐SNc) on neuromelanin magnetic resonance imaging (NM‐MRI) and the specific binding ratio (SBR) on 123I‐N‐v‐fluoropropyl‐2b‐carbomethoxy3b‐(4‐iodophenyl)nortropane single photon emission computed tomography (DaT‐SPECT) can be correlated with motor fluctuations (MFs) in advanced Parkinson's disease (PD) was investigated. Methods Thirty‐five PD patients (60 ± 13 years) and 23 healthy individuals as controls (59 ± 19 years) were enrolled. The relationships between NM‐MRI and DaT‐SPECT were prospectively examined in two subgroups divided according to the presence or absence of MFs. Multivariate analysis was performed using the Cox proportional hazard model to screen for association factors. Results The NM‐SNc size was correlated with the SBR (Spearman's ρ = 0.43, P < 0.05). The NM‐SNc size was significantly reduced in PD with MFs compared with the subgroup without (P < 0.001), whereas the SBR did not significantly differ between the groups. NM‐SNc size was a significant association factor for MFs (hazard ratio 0.94, P = 0.04). In receiver operating characteristic analysis of the factors for MF occurrence, the area under the receiver operating characteristic curve of the NM‐SNc size showed a significant difference of 0.89 (P < 0.05) but no significant difference was found in the SBR. Conclusions NM‐SNc size was significantly correlated with the SBR in PD, but several factors in advanced PD were more closely associated with NM‐SNc size than the SBR. NM‐MRI might reflect the status of advanced PD more accurately than DaT‐SPECT. Therefore, NM‐MRI appears to provide a better marker for discriminating advanced PD than DaT‐SPECT.
Collapse
Affiliation(s)
- A Okuzumi
- Department of Neurology, Graduate School of Medicine, Juntendo University, Tokyo, Japan
| | - T Hatano
- Department of Neurology, Graduate School of Medicine, Juntendo University, Tokyo, Japan
| | - K Kamagata
- Department of Radiology, Graduate School of Medicine, Juntendo University, Tokyo, Japan
| | - M Hori
- Department of Radiology, Graduate School of Medicine, Juntendo University, Tokyo, Japan
| | - A Mori
- Department of Neurology, Graduate School of Medicine, Juntendo University, Tokyo, Japan
| | - Y Oji
- Department of Neurology, Graduate School of Medicine, Juntendo University, Tokyo, Japan
| | - D Taniguchi
- Department of Neurology, Graduate School of Medicine, Juntendo University, Tokyo, Japan
| | - K Daida
- Department of Neurology, Graduate School of Medicine, Juntendo University, Tokyo, Japan
| | - Y Shimo
- Department of Neurology, Graduate School of Medicine, Juntendo University, Tokyo, Japan.,Department of Research and Therapeutics for Movement Disorders, Juntendo University Graduate School of Medicine, Tokyo, Japan
| | - N Yanagisawa
- Medical Technology Innovation Center, Clinical Research and Trial Center, Juntendo University Graduate School of Medicine, Tokyo, Japan
| | - S Nojiri
- Medical Technology Innovation Center, Clinical Research and Trial Center, Juntendo University Graduate School of Medicine, Tokyo, Japan
| | - S Aoki
- Department of Radiology, Graduate School of Medicine, Juntendo University, Tokyo, Japan
| | - N Hattori
- Department of Neurology, Graduate School of Medicine, Juntendo University, Tokyo, Japan.,Department of Research and Therapeutics for Movement Disorders, Juntendo University Graduate School of Medicine, Tokyo, Japan
| |
Collapse
|
33
|
Nørgaard M, Ganz M, Svarer C, Feng L, Ichise M, Lanzenberger R, Lubberink M, Parsey RV, Politis M, Rabiner EA, Slifstein M, Sossi V, Suhara T, Talbot PS, Turkheimer F, Strother SC, Knudsen GM. Cerebral serotonin transporter measurements with [ 11C]DASB: A review on acquisition and preprocessing across 21 PET centres. J Cereb Blood Flow Metab 2019; 39:210-222. [PMID: 29651896 PMCID: PMC6365604 DOI: 10.1177/0271678x18770107] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
Positron Emission Tomography (PET) imaging has become a prominent tool to capture the spatiotemporal distribution of neurotransmitters and receptors in the brain. The outcome of a PET study can, however, potentially be obscured by suboptimal and/or inconsistent choices made in complex processing pipelines required to reach a quantitative estimate of radioligand binding. Variations in subject selection, experimental design, data acquisition, preprocessing, and statistical analysis may lead to different outcomes and neurobiological interpretations. We here review the approaches used in 105 original research articles published by 21 different PET centres, using the tracer [11C]DASB for quantification of cerebral serotonin transporter binding, as an exemplary case. We highlight and quantify the impact of the remarkable variety of ways in which researchers are currently conducting their studies, while implicitly expecting generalizable results across research groups. Our review provides evidence that the foundation for a given choice of a preprocessing pipeline seems to be an overlooked aspect in modern PET neuroscience. Furthermore, we believe that a thorough testing of pipeline performance is necessary to produce reproducible research outcomes, avoiding biased results and allowing for better understanding of human brain function.
Collapse
Affiliation(s)
- Martin Nørgaard
- 1 Neurobiology Research Unit, Copenhagen University Hospital, Rigshospitalet, Copenhagen, Denmark.,2 Faculty of Health Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Melanie Ganz
- 1 Neurobiology Research Unit, Copenhagen University Hospital, Rigshospitalet, Copenhagen, Denmark.,3 Department of Computer Science, University of Copenhagen, Copenhagen, Denmark
| | - Claus Svarer
- 1 Neurobiology Research Unit, Copenhagen University Hospital, Rigshospitalet, Copenhagen, Denmark
| | - Ling Feng
- 1 Neurobiology Research Unit, Copenhagen University Hospital, Rigshospitalet, Copenhagen, Denmark
| | - Masanori Ichise
- 4 Department of Functional Brain Imaging Research, National Institute of Radiological Sciences, National Institutes for Quantum and Radiological Science and Technology, Chiba, Japan
| | - Rupert Lanzenberger
- 5 Department of Psychiatry and Psychotherapy, Medical University of Vienna, Vienna, Austria
| | - Mark Lubberink
- 6 Department of Nuclear Medicine and Positron Emission Tomography, Uppsala University, Uppsala, Sweden
| | - Ramin V Parsey
- 7 Department of Psychiatry, School of Medicine, Stony Brook University, Stony Brook, NY, USA
| | - Marios Politis
- 8 Neurodegeneration Imaging Group, Institute of Psychiatry, Psychology and Neuroscience (IoPPN), King's College London, London, UK
| | - Eugenii A Rabiner
- 9 Imanova Limited, London, UK.,10 Centre for Neuroimaging Sciences, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, UK
| | - Mark Slifstein
- 7 Department of Psychiatry, School of Medicine, Stony Brook University, Stony Brook, NY, USA
| | - Vesna Sossi
- 11 Department of Physics and Astronomy, University of British Columbia, Vancouver, BC, Canada
| | - Tetsuya Suhara
- 4 Department of Functional Brain Imaging Research, National Institute of Radiological Sciences, National Institutes for Quantum and Radiological Science and Technology, Chiba, Japan
| | - Peter S Talbot
- 12 Division of Neuroscience and Experimental Psychology, School of Biological Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester Academic Health Science Centre, Manchester, UK
| | | | - Stephen C Strother
- 14 Rotman Research Institute at Baycrest, University of Toronto, Toronto, Canada
| | - Gitte M Knudsen
- 1 Neurobiology Research Unit, Copenhagen University Hospital, Rigshospitalet, Copenhagen, Denmark.,2 Faculty of Health Sciences, University of Copenhagen, Copenhagen, Denmark
| |
Collapse
|
34
|
Sellnow RC, Newman JH, Chambers N, West AR, Steece-Collier K, Sandoval IM, Benskey MJ, Bishop C, Manfredsson FP. Regulation of dopamine neurotransmission from serotonergic neurons by ectopic expression of the dopamine D2 autoreceptor blocks levodopa-induced dyskinesia. Acta Neuropathol Commun 2019; 7:8. [PMID: 30646956 PMCID: PMC6332643 DOI: 10.1186/s40478-018-0653-7] [Citation(s) in RCA: 46] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2018] [Accepted: 12/19/2018] [Indexed: 12/01/2022] Open
Abstract
Levodopa-induced dyskinesias (LID) are a prevalent side effect of chronic treatment with levodopa (L-DOPA) for the motor symptoms of Parkinson’s disease (PD). It has long been hypothesized that serotonergic neurons of the dorsal raphe nucleus (DRN) are capable of L-DOPA uptake and dysregulated release of dopamine (DA), and that this “false neurotransmission” phenomenon is a main contributor to LID development. Indeed, many preclinical studies have demonstrated LID management with serotonin receptor agonist treatment, but unfortunately, promising preclinical data has not been translated in large-scale clinical trials. Importantly, while there is an abundance of convincing clinical and preclinical evidence supporting a role of maladaptive serotonergic neurotransmission in LID expression, there is no direct evidence that dysregulated DA release from serotonergic neurons impacts LID formation. In this study, we ectopically expressed the DA autoreceptor D2Rs (or GFP) in the DRN of 6-hydroxydopamine (6-OHDA) lesioned rats. No negative impact on the therapeutic efficacy of L-DOPA was seen with rAAV-D2Rs therapy. However, D2Rs treated animals, when subjected to a LID-inducing dose regimen of L-DOPA, remained completely resistant to LID, even at high doses. Moreover, the same subjects remained resistant to LID formation when treated with direct DA receptor agonists, suggesting D2Rs activity in the DRN blocked dyskinesogenic L-DOPA priming of striatal neurons. In vivo microdialysis confirmed that DA efflux in the striatum was reduced with rAAV-D2Rs treatment, providing explicit evidence that abnormal DA release from DRN neurons can affect LID. This is the first direct evidence of dopaminergic neurotransmission in DRN neurons and its modulation with rAAV-D2Rs gene therapy confirms the serotonin hypothesis in LID, demonstrating that regulation of serotonergic neurons achieved with a gene therapy approach offers a novel and potent antidyskinetic therapy.
Collapse
|
35
|
Pathophysiology of levodopa-induced dyskinesia: Insights from multimodal imaging and immunohistochemistry in non-human primates. Neuroimage 2018; 183:132-141. [DOI: 10.1016/j.neuroimage.2018.08.016] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2018] [Revised: 07/21/2018] [Accepted: 08/09/2018] [Indexed: 12/12/2022] Open
|
36
|
Espay AJ, Morgante F, Merola A, Fasano A, Marsili L, Fox SH, Bezard E, Picconi B, Calabresi P, Lang AE. Levodopa-induced dyskinesia in Parkinson disease: Current and evolving concepts. Ann Neurol 2018; 84:797-811. [DOI: 10.1002/ana.25364] [Citation(s) in RCA: 153] [Impact Index Per Article: 25.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2018] [Revised: 10/16/2018] [Accepted: 10/17/2018] [Indexed: 01/05/2023]
Affiliation(s)
- Alberto J. Espay
- UC Gardner Neuroscience Institute and Gardner Family Center for Parkinson's Disease and Movement Disorders, Department of Neurology; University of Cincinnati; Cincinnati OH
| | - Francesca Morgante
- Institute of Molecular and Clinical Sciences; St George's University of London; London United Kingdom
| | - Aristide Merola
- UC Gardner Neuroscience Institute and Gardner Family Center for Parkinson's Disease and Movement Disorders, Department of Neurology; University of Cincinnati; Cincinnati OH
| | - Alfonso Fasano
- Edmond J. Safra Program in Parkinson's Disease, Morton and Gloria Shulman Movement Disorders Clinic, Toronto Western Hospital, University Health Network, Division of Neurology; University of Toronto; Toronto Ontario Canada
- Krembil Brain Institute; Toronto Ontario Canada
| | - Luca Marsili
- UC Gardner Neuroscience Institute and Gardner Family Center for Parkinson's Disease and Movement Disorders, Department of Neurology; University of Cincinnati; Cincinnati OH
| | - Susan H. Fox
- Edmond J. Safra Program in Parkinson's Disease, Morton and Gloria Shulman Movement Disorders Clinic, Toronto Western Hospital, University Health Network, Division of Neurology; University of Toronto; Toronto Ontario Canada
- Krembil Brain Institute; Toronto Ontario Canada
| | - Erwan Bezard
- University of Bordeaux, Institute of Neurodegenerative Diseases; Bordeaux France
- National Center for Scientific Research, Institute of Neurodegenerative Diseases; Bordeaux France
| | - Barbara Picconi
- Experimental Neurophysiology Laboratory; IRCCS San Raffaele Pisana, University San Raffaele; Rome Italy
| | - Paolo Calabresi
- Neurological Clinic; University of Perugia, Santa Maria della Misericordia Hospital; Perugia Italy
| | - Anthony E. Lang
- Edmond J. Safra Program in Parkinson's Disease, Morton and Gloria Shulman Movement Disorders Clinic, Toronto Western Hospital, University Health Network, Division of Neurology; University of Toronto; Toronto Ontario Canada
- Krembil Brain Institute; Toronto Ontario Canada
| |
Collapse
|
37
|
de Natale ER, Wilson H, Pagano G, Politis M. Imaging Transplantation in Movement Disorders. INTERNATIONAL REVIEW OF NEUROBIOLOGY 2018; 143:213-263. [PMID: 30473196 DOI: 10.1016/bs.irn.2018.10.002] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Cell replacement therapy with graft transplantation has been tested as a disease-modifying treatment in neurodegenerative diseases characterized by the damage of a predominant cell type, such as substantia nigra dopaminergic neurons in Parkinson's disease (PD) or striatal medium spiny projection neurons in Huntington's disease (HD). The results of these trials are mixed with success in preclinical and pilot open-label trials, which were not consistently reproduced in randomized controlled trials. Positron emission tomography (PET) and single photon emission computed tomography (SPECT) molecular imaging and functional magnetic resonance imaging allow the graft survival, and its relationship with the host tissues to be studied in vivo. In PD, PET with [18F]DOPA showed that graft survival does not necessarily correlate with the clinical improvement and PD patients with worse outcome had lower binding in the ventral striatum and a high serotonin ([11C]DASB PET) to dopamine ([18F]DOPA PET) ratio in the grafted neurons. In HD, PET with [11C]PK11195 showed the graft survival and the clinical responses may be related to the reactive activation of the host inflammatory/immune system. Findings from these studies have been used to refine study protocols and patient selection in current clinical trials, which includes identifying suitable candidates for transplantation using imaging markers and employing multiple and/or novel PET tracers to better assess graft functions and inflammatory responses to grafts.
Collapse
Affiliation(s)
- Edoardo Rosario de Natale
- Neurodegeneration Imaging Group, Maurice Wohl Clinical Neuroscience Institute, Institute of Psychiatry, Psychology and Neuroscience (IoPPN), King's College London, London, United Kingdom
| | - Heather Wilson
- Neurodegeneration Imaging Group, Maurice Wohl Clinical Neuroscience Institute, Institute of Psychiatry, Psychology and Neuroscience (IoPPN), King's College London, London, United Kingdom
| | - Gennaro Pagano
- Neurodegeneration Imaging Group, Maurice Wohl Clinical Neuroscience Institute, Institute of Psychiatry, Psychology and Neuroscience (IoPPN), King's College London, London, United Kingdom
| | - Marios Politis
- Neurodegeneration Imaging Group, Maurice Wohl Clinical Neuroscience Institute, Institute of Psychiatry, Psychology and Neuroscience (IoPPN), King's College London, London, United Kingdom.
| |
Collapse
|
38
|
Strafella AP, Bohnen NI, Pavese N, Vaillancourt DE, van Eimeren T, Politis M, Tessitore A, Ghadery C, Lewis S. Imaging Markers of Progression in Parkinson's Disease. Mov Disord Clin Pract 2018; 5:586-596. [PMID: 30637278 DOI: 10.1002/mdc3.12673] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2018] [Revised: 07/22/2018] [Accepted: 07/30/2018] [Indexed: 12/12/2022] Open
Abstract
Background Parkinson's disease (PD) is the second-most common neurodegenerative disorder after Alzheimer's disease; however, to date, there is no approved treatment that stops or slows down disease progression. Over the past decades, neuroimaging studies, including molecular imaging and MRI are trying to provide insights into the mechanisms underlying PD. Methods This work utilized a literature review. Results It is now becoming clear that these imaging modalities can provide biomarkers that can objectively detect brain changes related to PD and monitor these changes as the disease progresses, and these biomarkers are required to establish a breakthrough in neuroprotective or disease-modifying therapeutics. Conclusions Here, we provide a review of recent observations deriving from PET, single-positron emission tomography, and MRI studies exploring PD and other parkinsonian disorders.
Collapse
Affiliation(s)
- Antonio P Strafella
- Morton and Gloria Shulman Movement Disorder Unit & E.J. Safra Parkinson Disease Program, Neurology Division, Department of Medicine, Toronto Western Hospital, UHN University of Toronto Toronto Ontario Canada.,Division of Brain, Imaging and Behaviour-Systems Neuroscience, Krembil Research Institute, UHN University of Toronto Toronto Ontario Canada.,Research Imaging Centre, Campbell Family Mental Health Research Institute, Centre for Addiction and Mental Health University of Toronto Toronto Ontario Canada
| | - Nico I Bohnen
- Department of Radiology & Neurology University of Michigan Ann Arbor Michigan USA.,Veterans Administration Ann Arbor Healthcare System Ann Arbor Michigan USA.,Morris K. Udall Center of Excellence for Parkinson's Disease Research University of Michigan Ann Arbor Michigan USA
| | - Nicola Pavese
- Newcastle Magnetic Resonance Centre & Positron Emission Tomography Centre Newcastle University, Campus for Ageing & Vitality Newcastle upon Tyne United Kingdom
| | - David E Vaillancourt
- Applied Physiology and Kinesiology, Biomedical Engineering, and Neurology University of Florida Gainesville Florida USA
| | - Thilo van Eimeren
- Department of Nuclear Medicine and Department of Neurology University of Cologne Cologne Germany.,Institute for Cognitive Neuroscience, Jülich Research Centre Jülich Germany.,German Center for Neurodegenerative Diseases (DZNE) Bonn-Cologne Bonn Germany
| | - Marios Politis
- Neurodegeneration Imaging Group (NIG), Institute of Psychiatry, Psychology and Neuroscience (IoPPN), King's College London London United Kingdom
| | - Alessandro Tessitore
- Department of Medical, Surgical, Neurological, Metabolic and Aging Sciences-MRI Research Center SUN-FISM University of Campania "Luigi Vanvitelli" Naples Italy
| | - Christine Ghadery
- Morton and Gloria Shulman Movement Disorder Unit & E.J. Safra Parkinson Disease Program, Neurology Division, Department of Medicine, Toronto Western Hospital, UHN University of Toronto Toronto Ontario Canada.,Division of Brain, Imaging and Behaviour-Systems Neuroscience, Krembil Research Institute, UHN University of Toronto Toronto Ontario Canada.,Research Imaging Centre, Campbell Family Mental Health Research Institute, Centre for Addiction and Mental Health University of Toronto Toronto Ontario Canada
| | - Simon Lewis
- Parkinson's Disease Research Clinic, Brain and Mind Centre University of Sydney Sydney NSW Australia
| | | |
Collapse
|
39
|
Lane EL. L-DOPA for Parkinson's disease-a bittersweet pill. Eur J Neurosci 2018; 49:384-398. [PMID: 30118169 DOI: 10.1111/ejn.14119] [Citation(s) in RCA: 37] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2018] [Revised: 07/23/2018] [Accepted: 07/30/2018] [Indexed: 01/02/2023]
Abstract
3,4-dihydroxy-L-phenylalanine (L-DOPA) is the gold standard treatment for Parkinson's disease. It has earned that title through its highly effective treatment of some of the motor symptoms in the early stages of the disease but it is a far from perfect drug. The inevitable long-term treatment that comes with this chronic neurodegenerative condition raises the risk significantly of the development of motor fluctuations including disabling L-DOPA-induced dyskinesia. Being unsurpassed as a therapy means that understanding the mechanisms of dyskinesia priming and induction is vital to the search for therapies to treat these side effects and allow optimal use of L-DOPA. However, L-DOPA use may also have consequences (positive or negative) for the development of other interventions, such as cell transplantation, which are designed to treat or repair the ailing brain. This review looks at the issues around the use of L-DOPA with a focus on its potential impact on advanced reparative interventions.
Collapse
Affiliation(s)
- Emma L Lane
- Cardiff School of Pharmacy and Pharmaceutical Sciences, Cardiff University, Cardiff, UK
| |
Collapse
|
40
|
Du JJ, Chen SD. Current Nondopaminergic Therapeutic Options for Motor Symptoms of Parkinson's Disease. Chin Med J (Engl) 2018; 130:1856-1866. [PMID: 28748860 PMCID: PMC5547839 DOI: 10.4103/0366-6999.211555] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
Objective: The aim of this study was to summarize recent studies on nondopaminergic options for the treatment of motor symptoms in Parkinson's disease (PD). Data Sources: Papers in English published in PubMed, Cochrane, and Ovid Nursing databases between January 1988 and November 2016 were searched using the following keywords: PD, nondopaminergic therapy, adenosine, glutamatergic, adrenergic, serotoninergic, histaminic, and iron chelator. We also reviewed the ongoing clinical trials in the website of clinicaltrials.gov. Study Selection: Articles related to the nondopaminergic treatment of motor symptoms in PD were selected for this review. Results: PD is conventionally treated with dopamine replacement strategies, which are effective in the early stages of PD. Long-term use of levodopa could result in motor complications. Recent studies revealed that nondopaminergic systems such as adenosine, glutamatergic, adrenergic, serotoninergic, histaminic, and iron chelator pathways could include potential therapeutic targets for motor symptoms, including motor fluctuations, levodopa-induced dyskinesia, and gait disorders. Some nondopaminergic drugs, such as istradefylline and amantadine, are currently used clinically, while most such drugs are in preclinical testing stages. Transitioning of these agents into clinically beneficial strategies requires reliable evaluation since several agents have failed to show consistent results despite positive findings at the preclinical level. Conclusions: Targeting nondopaminergic transmission could improve some motor symptoms in PD, especially the discomfort of dyskinesia. Although nondopaminergic treatments show great potential in PD treatment as an adjunct therapy to levodopa, further investigation is required to ensure their success.
Collapse
Affiliation(s)
- Juan-Juan Du
- Department of Neurology and Institute of Neurology, Ruijin Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Sheng-Di Chen
- Department of Neurology and Institute of Neurology, Ruijin Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| |
Collapse
|
41
|
|
42
|
Effects of the Serotonin 5-HT1A Receptor Biased Agonists, F13714 and F15599, on Striatal Neurotransmitter Levels Following l-DOPA Administration in Hemi-Parkinsonian Rats. Neurochem Res 2018; 43:1035-1046. [DOI: 10.1007/s11064-018-2514-y] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2018] [Revised: 03/15/2018] [Accepted: 03/20/2018] [Indexed: 11/27/2022]
|
43
|
Sgambato V, Tremblay L. Pathophysiology of dyskinesia and behavioral disorders in non-human primates: the role of serotonergic fibers. J Neural Transm (Vienna) 2018; 125:1145-1156. [PMID: 29502255 DOI: 10.1007/s00702-018-1871-7] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2017] [Accepted: 02/27/2018] [Indexed: 12/26/2022]
Abstract
The MPTP monkey model of Parkinson's disease (PD) has allowed huge advances regarding the understanding of the pathological mechanisms of PD and L-DOPA-induced adverse effects. Among the main findings were the imbalance between the efferent striatal pathways in opposite directions between the hypokinetic and hyperkinetic states of PD. In both normal and parkinsonian monkeys, the combination of behavioral and anatomical studies has allowed the deciphering of the cortico-basal ganglia circuits involved in both movement and behavioral disorders. A major breakthrough has then been made regarding the hypothesis of the involvement of serotonergic fibers in the conversion of L-DOPA to dopamine when dopaminergic neurons are dying and to release it, in an uncontrolled manner, as serotonergic neurons are deprived from the machinery required for buffering dopamine from the synaptic cleft. The crucial involvement of serotonergic fibers underlying L-DOPA-induced dyskinesia (LID) has been demonstrated in both rodent and monkey models of PD, in which dyskinesia induced by L-DOPA is abolished following lesion of the serotonergic system. Moreover, the role of serotonergic fibers goes well beyond dyskinesia, as lesioning of such serotonergic fibers by MDMA in the monkey also decreased other L-DOPA-induced adverse effects such as impulsive compulsive behaviors and visual hallucinations. The same pathological mechanism, i.e., an imbalance between serotonin and dopamine terminals may, therefore, favor L-DOPA-induced adverse effects according to the basal ganglia territory it inhabits. Further non-human primate studies will be needed to demonstrate the role of such a pathological mechanism in both movement and behavioral disorders driven by L-DOPA therapy but also to determine the causal link between serotonin lesions and the expression of non-motor symptoms like apathy, depression and anxiety, frequently observed in PD patients.
Collapse
Affiliation(s)
- Véronique Sgambato
- Institut des Sciences Cognitives Marc Jeannerod, UMR 5229, Univ Lyon, CNRS, 69675, Bron, France.
| | - Léon Tremblay
- Institut des Sciences Cognitives Marc Jeannerod, UMR 5229, Univ Lyon, CNRS, 69675, Bron, France
| |
Collapse
|
44
|
Carta M, Björklund A. The serotonergic system in L-DOPA-induced dyskinesia: pre-clinical evidence and clinical perspective. J Neural Transm (Vienna) 2018; 125:1195-1202. [PMID: 29480391 DOI: 10.1007/s00702-018-1865-5] [Citation(s) in RCA: 31] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2018] [Accepted: 02/19/2018] [Indexed: 11/28/2022]
Abstract
During the last decade, the serotonergic system has emerged as a key player in the appearance of L-DOPA-induced dyskinesia in animal models of Parkinson's disease. Clinical investigations, based on imaging and postmortem analyses, suggest that the serotonin neurons are also involved in the etiology of this complication of long-term L-DOPA treatment in parkinsonian patients. These findings have stimulated efforts to develop new therapies using drugs targeting the malfunctioning serotonin neurons. In this review, we summarize the experimental and clinical data obtained so far and discuss the prospects for further development of this therapeutic strategy.
Collapse
Affiliation(s)
- Manolo Carta
- Department of Biomedical Sciences, Section of Physiology, University of Cagliari, Cittadella Universitaria, SS554, Km 4.5, 09042, Monserrato, Italy.
| | - Anders Björklund
- Division of Neurobiology, Department of Experimental Medical Science, Wallenberg Neuroscience Center, Lund University, 221 84, Lund, Sweden
| |
Collapse
|
45
|
Dyskinesias and levodopa therapy: why wait? J Neural Transm (Vienna) 2018; 125:1119-1130. [PMID: 29428995 DOI: 10.1007/s00702-018-1856-6] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2017] [Accepted: 02/05/2018] [Indexed: 12/28/2022]
Abstract
Throughout the years there has been a longstanding discussion on whether levodopa therapy in Parkinson's disease should be started in early vs. later stages, in order to prevent or delay motor complications such as fluctuations and dyskinesias. This controversial topic has been extensively debated for decades, and the prevailing view today is that levodopa should not be postponed. However, there is still fear associated with its use in early stages, especially in younger patients, who are more prone to develop dyskinesias. Even though dyskinesias are linked to levodopa use in Parkinson's disease, it has been shown that starting with a different medication (such as dopamine agonists) will not significantly delay their onset once levodopa is introduced. Since levodopa provides better symptomatic control, and other drugs may be associated with notable side effects, it is our view that there is insufficient evidence to justify levodopa-sparing strategies. The physician should try to assess each patient individually, taking into account motor and non-motor demands, as well as risk factors for potential complications, finding the optimum treatment strategy for each one. The following article provides an historical narrative perspective, as well as a literature review of those intrinsic and modifiable risk factors that have been associated with levodopa-induced dyskinesias, which should be taken into consideration when choosing the therapeutic strategy in individual Parkinson's disease patients.
Collapse
|
46
|
Serotonergic targets for the treatment of L-DOPA-induced dyskinesia. J Neural Transm (Vienna) 2018; 125:1203-1216. [PMID: 29305656 DOI: 10.1007/s00702-017-1837-1] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2017] [Accepted: 12/28/2017] [Indexed: 12/31/2022]
Abstract
Dopamine (DA) replacement therapy with L-3,4-dihydroxyphenylalanine (L-DOPA) continues to be the gold-standard treatment for Parkinson's disease (PD). Despite clear symptomatic benefit, long-term L-DOPA use often results in the development of L-DOPA-induced dyskinesia (LID), significantly reducing quality of life and increasing costs for PD patients and their caregivers. Accumulated research has demonstrated that several pre- and post-synaptic mechanisms contribute to LID development and expression. In particular, raphe-striatal hyperinnervation and unregulated DA release from 5-HT terminals is postulated to play a central role in LID manifestation. As such, manipulation of the 5-HT system has garnered considerable attention. Both pre-clinical and clinical research has supported the potential of modulating the 5-HT system for LID prevention and treatment. This review discusses the rationale for continued investigation of several potential anti-dyskinetic strategies including 5-HT stimulation of 5-HT1A and 5-HT1B receptors and blockade of 5-HT2A receptors and SERT. We present the latest findings from experimental and clinical investigations evaluating these 5-HT targets with the goal of identifying those with translational promise and the challenges associated with each.
Collapse
|
47
|
Molecular Imaging of the Serotonergic System in Parkinson's Disease. INTERNATIONAL REVIEW OF NEUROBIOLOGY 2018; 141:173-210. [DOI: 10.1016/bs.irn.2018.08.002] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
|
48
|
Pagano G, Niccolini F, Politis M. The serotonergic system in Parkinson's patients with dyskinesia: evidence from imaging studies. J Neural Transm (Vienna) 2017; 125:1217-1223. [PMID: 29264660 PMCID: PMC6060863 DOI: 10.1007/s00702-017-1823-7] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2017] [Accepted: 12/05/2017] [Indexed: 12/31/2022]
Abstract
The purpose of review is to review the current status of positron emission tomography (PET) molecular imaging of serotonergic system in Parkinson’s patients who experience levodopa-induced (LIDs) and graft-induced dyskinesias (GIDs). PET imaging studies have shown that Parkinson’s disease is characterized by progressive loss of dopaminergic and serotonergic neurons. Parkinson’s patients who experienced LIDs and GIDs have an aberrant spreading of serotonergic terminals, which lead to an increased serotonergic/dopaminergic terminals ratio within the putamen. Serotonergic terminals convert exogenous levodopa into dopamine in a non-physiological manner and release an abnormal amount of dopamine without an auto-regulatory feedback. This results in higher swings in synaptic levels of dopamine, which leads to the development of LIDs and GIDs. The modulation of serotonergic terminals with 5-HT1A and 5-HT1B receptors agonists partially reduced these motor complications. In vivo PET studies confirmed that abnormal spreading of serotonergic terminals within the putamen has a pivotal role in the development of LIDs and GIDs. However, glutamatergic, adenosinergic, opioid systems, and phosphodiesterases 10A may also play a role in the development of these motor complications. An integrative multimodal imaging approach combining PET and MRI imaging techniques is needed to fully understand the mechanisms underlying the development of LIDs and GIDs.
Collapse
Affiliation(s)
- Gennaro Pagano
- Neurodegeneration Imaging Group, Maurice Wohl Clinical Neuroscience Institute, Institute of Psychiatry, Psychology and Neuroscience (IoPPN), King's College London, 125 Coldharbour Lane, Camberwell, London, SE5 9NU, UK
| | - Flavia Niccolini
- Neurodegeneration Imaging Group, Maurice Wohl Clinical Neuroscience Institute, Institute of Psychiatry, Psychology and Neuroscience (IoPPN), King's College London, 125 Coldharbour Lane, Camberwell, London, SE5 9NU, UK
| | - Marios Politis
- Neurodegeneration Imaging Group, Maurice Wohl Clinical Neuroscience Institute, Institute of Psychiatry, Psychology and Neuroscience (IoPPN), King's College London, 125 Coldharbour Lane, Camberwell, London, SE5 9NU, UK.
| |
Collapse
|
49
|
Picconi B, Hernández LF, Obeso JA, Calabresi P. Motor complications in Parkinson's disease: Striatal molecular and electrophysiological mechanisms of dyskinesias. Mov Disord 2017; 33:867-876. [PMID: 29219207 DOI: 10.1002/mds.27261] [Citation(s) in RCA: 71] [Impact Index Per Article: 10.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2017] [Revised: 10/30/2017] [Accepted: 11/02/2017] [Indexed: 12/11/2022] Open
Abstract
Long-term levodopa (l-dopa) treatment in patients with Parkinson´s disease (PD) is associated with the development of motor complications (ie, motor fluctuations and dyskinesias). The principal etiopathogenic factors are the degree of nigro-striatal dopaminergic loss and the duration and dose of l-dopa treatment. In this review article we concentrate on analysis of the mechanisms underlying l-dopa-induced dyskinesias, a phenomenon that causes disability in a proportion of patients and that has not benefited from major therapeutic advances. Thus, we discuss the main neurotransmitters, receptors, and pathways that have been thought to play a role in l-dopa-induced dyskinesias from the perspective of basic neuroscience studies. Some important advances in deciphering the molecular pathways involved in these abnormal movements have occurred in recent years to reveal potential targets that could be used for therapeutic purposes. However, it has not been an easy road because there have been a plethora of components involved in the generation of these undesired movements, even bypassing the traditional and well-accepted dopamine receptor activation, as recently revealed by optogenetics. Here, we attempt to unify the available data with the hope of guiding and fostering future research in the field of striatal activation and abnormal movement generation. © 2017 International Parkinson and Movement Disorder Society.
Collapse
Affiliation(s)
| | - Ledia F Hernández
- HM CINAC, Hospital Universitario HM Puerta del Sur, Mostoles, Madrid, Spain.,Universidad CEU San Pablo, Madrid, Spain.,Center for Networked Biomedical Research on Neurodegenerative Diseases, Madrid, Spain
| | - Jose A Obeso
- HM CINAC, Hospital Universitario HM Puerta del Sur, Mostoles, Madrid, Spain.,Universidad CEU San Pablo, Madrid, Spain.,Center for Networked Biomedical Research on Neurodegenerative Diseases, Madrid, Spain
| | - Paolo Calabresi
- Fondazione Santa Lucia, IRCCS, Rome, Italy.,Clinica Neurologica, Università degli studi di Perugia, Ospedale Santa Maria della Misericordia, Perugia, Italy
| |
Collapse
|
50
|
Lower serotonin transporter binding in patients with cervical dystonia is associated with psychiatric symptoms. EJNMMI Res 2017; 7:87. [PMID: 29071431 PMCID: PMC5656503 DOI: 10.1186/s13550-017-0338-4] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2017] [Accepted: 10/17/2017] [Indexed: 12/12/2022] Open
Abstract
BACKGROUND Cervical dystonia (CD) is often accompanied by depressive symptoms, anxiety, and jerks/tremor. The dopamine transporter (DAT) binding is related with both depressive symptoms and jerks/tremor in CD. Serotonergic and dopaminergic systems are closely related. As serotonin is involved in the pathophysiology of psychiatric symptoms and jerks, we expected an altered serotoninergic system in CD. We hypothesized that CD is associated with reduced serotonin transporter (SERT) binding, more specific that SERT binding is lower in CD patients with psychiatric symptoms and/or jerks/tremor compared to those without, and to controls. The balance between SERT and DAT binding can be altered in different CD phenotypes. RESULTS In 23 CD patients and 14 healthy controls, SERT binding in the diencephalon/midbrain was assessed using [123I]FP-CIT SPECT, with a brain-dedicated system. The specific to non-specific binding ratio (binding potential; BPND) to SERT was the main outcome measure. There was a clear trend towards reduced SERT BPND in CD patients with psychiatric symptoms compared to those without (p = 0.05). There was no correlation between SERT binding and dystonia, jerks, or anxiety. There was a significant positive correlation between extrastriatal SERT and striatal DAT BPND in CD patients with jerks, but not in patients without jerks. CONCLUSION CD patients with psychiatric symptoms have lower SERT binding in the midbrain/diencephalon, while dystonia and jerks appear unrelated to SERT binding. The balance between extrastriatal SERT and striatal DAT binding is different in CD with and without jerks.
Collapse
|