1
|
Caulfield ME, Vander Werp MJ, Stancati JA, Collier TJ, Sortwell CE, Sandoval IM, Kordower JH, Manfredsson FP, Steece-Collier K. Advancing age and sex modulate antidyskinetic efficacy of striatal Ca V1.3 gene therapy in a rat model of Parkinson's disease. Neurobiol Aging 2025; 149:54-66. [PMID: 40010015 PMCID: PMC12007665 DOI: 10.1016/j.neurobiolaging.2025.02.003] [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: 09/05/2024] [Revised: 02/09/2025] [Accepted: 02/10/2025] [Indexed: 02/28/2025]
Abstract
We previously demonstrated that viral vector-mediated striatal CaV1.3 calcium channel downregulation in young adult (3mo) male parkinsonian rats provides uniform, robust protection against levodopa-induced dyskinesias (LID). Acknowledging the association of PD with aging and incidence in male and female sexes, we have expanded our studies to include rats of advancing age of both sexes. The current study directly contrasts age and sex, determining their impact on efficacy of intrastriatal AAV-CaV1.3-shRNA to prevent LID induction, removing the variable of levodopa-priming. Considering both sexes together, late-middle-aged ('aged'; 15mo) parkinsonian rats receiving AAV-CaV1.3-shRNA developed significantly less severe LID compared control AAV-scramble(SCR)-shRNA rats, however therapeutic benefit was significantly less robust than observed in young males. When considered separately, females showed significantly less therapeutic benefit than males. Furthermore, aged non-cycling/proestrous-negative female rats were refractory to LID induction, regardless of vector. This study provides novel insight into the impact of age and sex on the variable antidyskinetic responses of CaV1.3-targeted gene therapy, highlighting the importance of including clinically relevant age and sex populations in PD studies.
Collapse
Affiliation(s)
- Margaret E Caulfield
- Department of Translational Neuroscience, College of Human Medicine, Michigan State University, 400 Monroe Ave NW, Grand Rapids, MI 49503, USA
| | - Molly J Vander Werp
- Department of Translational Neuroscience, College of Human Medicine, Michigan State University, 400 Monroe Ave NW, Grand Rapids, MI 49503, USA
| | - Jennifer A Stancati
- Department of Translational Neuroscience, College of Human Medicine, Michigan State University, 400 Monroe Ave NW, Grand Rapids, MI 49503, USA
| | - Timothy J Collier
- Department of Translational Neuroscience, College of Human Medicine, Michigan State University, 400 Monroe Ave NW, Grand Rapids, MI 49503, USA
| | - Caryl E Sortwell
- Department of Translational Neuroscience, College of Human Medicine, Michigan State University, 400 Monroe Ave NW, Grand Rapids, MI 49503, USA
| | - Ivette M Sandoval
- Parkinson's Disease Research Unit, Department of Translational Neuroscience, Barrow Neurological Institute, Phoenix, AZ 85013, USA
| | - Jeffrey H Kordower
- ASU-Banner Neurodegenerative Disease Research Center (NDRC), College of Liberal Arts and Sciences, Arizona State University, Tempe, AZ 85287, USA
| | - Fredric P Manfredsson
- Parkinson's Disease Research Unit, Department of Translational Neuroscience, Barrow Neurological Institute, Phoenix, AZ 85013, USA
| | - Kathy Steece-Collier
- Department of Translational Neuroscience, College of Human Medicine, Michigan State University, 400 Monroe Ave NW, Grand Rapids, MI 49503, USA.
| |
Collapse
|
2
|
Li C, Elabi OF, Fieblinger T, Cenci MA. Structural-functional properties of direct-pathway striatal neurons at early and chronic stages of dopamine denervation. Eur J Neurosci 2024; 59:1227-1241. [PMID: 37876330 DOI: 10.1111/ejn.16166] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2023] [Revised: 09/22/2023] [Accepted: 09/25/2023] [Indexed: 10/26/2023]
Abstract
The dendritic arbour of striatal projection neurons (SPNs) is the primary anatomical site where dopamine and glutamate inputs to the basal ganglia functionally interact to control movement. These dendritic arbourisations undergo atrophic changes in Parkinson's disease. A reduction in the dendritic complexity of SPNs is found also in animal models with severe striatal dopamine denervation. Using 6-hydroxydopamine (6-OHDA) lesions of the medial forebrain bundle as a model, we set out to compare morphological and electrophysiological properties of SPNs at an early versus a chronic stage of dopaminergic degeneration. Ex vivo recordings were performed in transgenic mice where SPNs forming the direct pathway (dSPNs) express a fluorescent reporter protein. At both the time points studied (5 and 28 days following 6-OHDA lesion), there was a complete loss of dopaminergic fibres through the dorsolateral striatum. A reduction in dSPN dendritic complexity and spine density was manifest at 28, but not 5 days post-lesion. At the late time point, dSPN also exhibited a marked increase in intrinsic excitability (reduced rheobase current, increased input resistance, more evoked action potentials in response to depolarising currents), which was not present at 5 days. The increase in neuronal excitability was accompanied by a marked reduction in inward-rectifying potassium (Kir) currents (which dampen the SPN response to depolarising stimuli). Our results show that dSPNs undergo delayed coordinate changes in dendritic morphology, intrinsic excitability and Kir conductance following dopamine denervation. These changes are predicted to interfere with the dSPN capacity to produce a normal movement-related output.
Collapse
Affiliation(s)
- Chang Li
- Basal Ganglia Pathophysiology Unit, Department Experimental Medical Science, Wallenberg Neuroscience Center, Lund University, Lund, Sweden
| | - Osama F Elabi
- Basal Ganglia Pathophysiology Unit, Department Experimental Medical Science, Wallenberg Neuroscience Center, Lund University, Lund, Sweden
| | - Tim Fieblinger
- Basal Ganglia Pathophysiology Unit, Department Experimental Medical Science, Wallenberg Neuroscience Center, Lund University, Lund, Sweden
- Evotec SE, Hamburg, Germany
| | - M Angela Cenci
- Basal Ganglia Pathophysiology Unit, Department Experimental Medical Science, Wallenberg Neuroscience Center, Lund University, Lund, Sweden
| |
Collapse
|
3
|
Caulfield ME, Manfredsson FP, Steece-Collier K. The Role of Striatal Cav1.3 Calcium Channels in Therapeutics for Parkinson's Disease. Handb Exp Pharmacol 2023; 279:107-137. [PMID: 36592226 DOI: 10.1007/164_2022_629] [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: 01/03/2023]
Abstract
Parkinson's disease (PD) is a relentlessly progressive neurodegenerative disorder with typical motor symptoms that include rigidity, tremor, and akinesia/bradykinesia, in addition to a host of non-motor symptoms. Motor symptoms are caused by progressive and selective degeneration of dopamine (DA) neurons in the SN pars compacta (SNpc) and the accompanying loss of striatal DA innervation from these neurons. With the exception of monogenic forms of PD, the etiology of idiopathic PD remains unknown. While there are a number of symptomatic treatment options available to individuals with PD, these therapies do not work uniformly well in all patients, and eventually most are plagued with waning efficacy and significant side-effect liability with disease progression. The incidence of PD increases with aging, and as such the expected burden of this disease will continue to escalate as our aging population increases (Dorsey et al. Neurology 68:384-386, 2007). The daunting personal and socioeconomic burden has pressed scientists and clinicians to find improved symptomatic treatment options devoid side-effect liability and meaningful disease-modifying therapies. Federal and private sources have supported clinical investigations over the past two-plus decades; however, no trial has yet been successful in finding an effective therapy to slow progression of PD, and there is currently just one FDA approved drug to treat the antiparkinsonian side-effect known as levodopa-induced dyskinesia (LID) that impacts approximately 90% of all individuals with PD. In this review, we present biological rationale and experimental evidence on the potential therapeutic role of the L-type voltage-gated Cav1.3 calcium (Ca2+) channels in two distinct brain regions, with two distinct mechanisms of action, in impacting the lives of individuals with PD. Our primary emphasis will be on the role of Cav1.3 channels in the striatum and the compelling evidence of their involvement in LID side-effect liability. We also briefly discuss the role of these same Ca2+ channels in the SNpc and the longstanding interest in Cav1.3 in this brain region in halting or delaying progression of PD.
Collapse
Affiliation(s)
- Margaret E Caulfield
- Department of Translational Neuroscience, College of Human Medicine, Michigan State University, Grand Rapids, MI, USA
| | - Fredric P Manfredsson
- Parkinson's Disease Research Unit, Department of Translational Neuroscience, Barrow Neurological Institute, Phoenix, AZ, USA
| | - Kathy Steece-Collier
- Department of Translational Neuroscience, College of Human Medicine, Michigan State University, Grand Rapids, MI, USA.
- Hauenstein Neuroscience Center, Mercy Health Saint Mary's, Grand Rapids, MI, USA.
| |
Collapse
|
4
|
A positive allosteric modulator of mGlu4 receptors restores striatal plasticity in an animal model of l-Dopa-induced dyskinesia. Neuropharmacology 2022; 218:109205. [DOI: 10.1016/j.neuropharm.2022.109205] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2022] [Revised: 07/21/2022] [Accepted: 07/23/2022] [Indexed: 11/24/2022]
|
5
|
Jung JH, Kim YJ, Chung SJ, Yoo HS, Lee YH, Baik K, Jeong SH, Lee YG, Lee HS, Ye BS, Sohn YH, Jeong Y, Lee PH. White matter connectivity networks predict levodopa-induced dyskinesia in Parkinson's disease. J Neurol 2022; 269:2948-2960. [PMID: 34762146 DOI: 10.1007/s00415-021-10883-1] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2021] [Revised: 10/22/2021] [Accepted: 10/29/2021] [Indexed: 11/25/2022]
Abstract
BACKGROUND Although levodopa-induced dyskinesia-relevant white matter change has been evaluated, it is uncertain whether these changes may reflect the underlying predisposing conditions leading to the development of levodopa-induced dyskinesia. OBJECTIVE To elucidate the role of white matter connectivity networks in the development of levodopa-induced dyskinesia in drug-naïve Parkinson's disease. METHODS We recruited 30 patients who developed levodopa-induced dyskinesia within 5 years from MRI acquisition (vulnerable-group), 47 patients who had not developed levodopa-induced dyskinesia within 5 years (resistant-group), and 28 controls. We performed comparative analyses of whole-brain white matter integrity and connectivity using tract-based spatial and network- and degree-based statistics. We evaluated the predictability of levodopa-induced dyskinesia development and relationship with its latency, using the average connectivity strength as a predictor in Cox- and linear-regression, respectively. RESULTS Mean-diffusivity was lower mainly at the left frontal region in the vulnerable-group compared to the resistant-group. Network-based statistics identified a subnetwork consisting of the bilateral fronto-striato-pallido-thalamic and lateral parietal regions (subnetwork A) and degree-based statistics identified four subnetworks (hub-subnetwork) consisting of edges centered on the left superior frontal gyrus, left putamen, left insular, or left precentral gyrus, where the vulnerable-group had stronger connectivity compared to the resistant-group. Stronger connectivity within the subnetwork A and hub-subnetwork centered on the left superior frontal gyrus was a predictor of levodopa-induced dyskinesia development independent of known risk factors and had an inverse relationship with its latency. CONCLUSIONS Our data suggest that white matter connectivity subnetworks within corticostriatal regions play a pivotal role in the development of levodopa-induced dyskinesia.
Collapse
Affiliation(s)
- Jin Ho Jung
- Department of Neurology, Busan Paik Hospital, Inje University College of Medicine, Busan, South Korea
| | - Yae Ji Kim
- Program of Brain and Cognitive Engineering, Korea Advanced Institute of Science and Technology, Daejeon, South Korea
- KI for Health Science and Technology, Korea Advanced Institute of Science and Technology, Daejeon, South Korea
| | - Seok Jong Chung
- Department of Neurology, Yonsei University College of Medicine, Seoul, South Korea
- Department of Neurology, Yongin Severance Hospital, Yonsei University Health System, Yongin, South Korea
| | - Han Soo Yoo
- Department of Neurology, Yonsei University College of Medicine, Seoul, South Korea
| | - Yang Hyun Lee
- Department of Neurology, Yonsei University College of Medicine, Seoul, South Korea
| | - Kyoungwon Baik
- Department of Neurology, Yonsei University College of Medicine, Seoul, South Korea
| | - Seong Ho Jeong
- Department of Neurology, Sanggye Paik Hospital, Inje University College of Medicine, Seoul, South Korea
| | - Young Gun Lee
- Department of Neurology, Yonsei University College of Medicine, Seoul, South Korea
| | - Hye Sun Lee
- Biostatistics Collaboration Unit, Yonsei University College of Medicine, Seoul, South Korea
| | - Byoung Seok Ye
- Department of Neurology, Yonsei University College of Medicine, Seoul, South Korea
| | - Young H Sohn
- Department of Neurology, Yonsei University College of Medicine, Seoul, South Korea
| | - Yong Jeong
- Program of Brain and Cognitive Engineering, Korea Advanced Institute of Science and Technology, Daejeon, South Korea.
- KI for Health Science and Technology, Korea Advanced Institute of Science and Technology, Daejeon, South Korea.
- Department of Bio and Brain Engineering, Korea Advanced Institute of Science and Technology, Daejeon, South Korea.
| | - Phil Hyu Lee
- Department of Neurology, Yonsei University College of Medicine, Seoul, South Korea.
- Severance Biomedical Science Institute, Yonsei University College of Medicine, Seoul, South Korea.
| |
Collapse
|
6
|
Fieblinger T, Li C, Espa E, Cenci MA. Non-Apoptotic Caspase-3 Activation Mediates Early Synaptic Dysfunction of Indirect Pathway Neurons in the Parkinsonian Striatum. Int J Mol Sci 2022; 23:ijms23105470. [PMID: 35628278 PMCID: PMC9141690 DOI: 10.3390/ijms23105470] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2022] [Revised: 05/09/2022] [Accepted: 05/12/2022] [Indexed: 11/27/2022] Open
Abstract
Non-apoptotic caspase-3 activation is critically involved in dendritic spine loss and synaptic dysfunction in Alzheimer’s disease. It is, however, not known whether caspase-3 plays similar roles in other pathologies. Using a mouse model of clinically manifest Parkinson’s disease, we provide the first evidence that caspase-3 is transiently activated in the striatum shortly after the degeneration of nigrostriatal dopaminergic projections. This caspase-3 activation concurs with a rapid loss of dendritic spines and deficits in synaptic long-term depression (LTD) in striatal projection neurons forming the indirect pathway. Interestingly, systemic treatment with a caspase inhibitor prevents both the spine pruning and the deficit of indirect pathway LTD without interfering with the ongoing dopaminergic degeneration. Taken together, our data identify transient and non-apoptotic caspase activation as a critical event in the early plastic changes of indirect pathway neurons following dopamine denervation.
Collapse
Affiliation(s)
- Tim Fieblinger
- Basal Ganglia Pathophysiology Unit, Department of Experimental Medical Science, Lund University, 223 62 Lund, Sweden; (C.L.); (E.E.)
- University Medical Center Hamburg-Eppendorf, Institute for Synaptic Physiology, 20251 Hamburg, Germany
- Correspondence: (T.F.); (M.A.C.)
| | - Chang Li
- Basal Ganglia Pathophysiology Unit, Department of Experimental Medical Science, Lund University, 223 62 Lund, Sweden; (C.L.); (E.E.)
| | - Elena Espa
- Basal Ganglia Pathophysiology Unit, Department of Experimental Medical Science, Lund University, 223 62 Lund, Sweden; (C.L.); (E.E.)
| | - M. Angela Cenci
- Basal Ganglia Pathophysiology Unit, Department of Experimental Medical Science, Lund University, 223 62 Lund, Sweden; (C.L.); (E.E.)
- Correspondence: (T.F.); (M.A.C.)
| |
Collapse
|
7
|
Mohammed M, Ivica N, Bjartmarz H, Thorbergsson PT, Pettersson LME, Thelin J, Schouenborg J. Microelectrode clusters enable therapeutic deep brain stimulation without noticeable side-effects in a rodent model of Parkinson's disease. J Neurosci Methods 2022; 365:109399. [PMID: 34695455 DOI: 10.1016/j.jneumeth.2021.109399] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2021] [Revised: 10/01/2021] [Accepted: 10/19/2021] [Indexed: 10/20/2022]
Abstract
BACKGROUND Deep Brain Stimulation (DBS) is an established treatment for motor symptoms in Parkinson's disease (PD). However, side effects often limit the usefulness of the treatment. NEW METHOD To mitigate this problem, we developed a novel cluster of ultrathin platinum-iridium microelectrodes (n = 16) embedded in a needle shaped gelatin vehicle. In an established rodent PD-model (6-OHDA unilateral lesion), the clusters were implanted in the subthalamic area for up to 8 weeks. In an open field setting, combinations of microelectrodes yielding therapeutic effects were identified using statistical methods. Immunofluorescence techniques were used for histological assessments of biocompatibility. RESULTS In all rats tested (n = 5), we found subsets of 3-4 microelectrodes which, upon stimulation (160 Hz, 60 μs pulse width, 25-40 μA/microelectrode), prompted normal movements without noticeable side effects. Other microelectrode subsets often caused side effects such as rotation, dyskinesia and tremor. The threshold (per microelectrode) to elicit normal movements strongly depended on the number of activated microelectrodes in the selected subset. The histological analysis revealed viable neurons close to the electrode contacts, minor microglial and astrocytic reactions and no major changes in the vasculature, indicating high biocompatibility. COMPARISON TO EXISTING METHODS AND CONCLUSION By contrast to the continuous and relatively large stimulation fields produced by existing DBS electrodes, the developed microelectrode cluster enables a fine-tuned granular and individualized microstimulation. This granular type of stimulation pattern provided powerful and specific therapeutic effects, free of noticeable side effects, in a PD animal model.
Collapse
Affiliation(s)
| | | | - Hjalmar Bjartmarz
- Neuronano Research Center, Lund University, Lund, Sweden; Department of Neurosurgery, Lund University and Skåne University Hospital in Lund, Lund, Sweden
| | | | - Lina M E Pettersson
- Neuronano Research Center, Lund University, Lund, Sweden; NanoLund, Lund University, Lund, Sweden
| | - Jonas Thelin
- Neuronano Research Center, Lund University, Lund, Sweden
| | - Jens Schouenborg
- Neuronano Research Center, Lund University, Lund, Sweden; NanoLund, Lund University, Lund, Sweden.
| |
Collapse
|
8
|
White matter alterations in Parkinson's disease with levodopa-induced dyskinesia. Parkinsonism Relat Disord 2021; 90:8-14. [PMID: 34325387 DOI: 10.1016/j.parkreldis.2021.07.021] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/16/2020] [Revised: 06/18/2021] [Accepted: 07/20/2021] [Indexed: 11/20/2022]
Abstract
INTRODUCTION Levodopa-induced dyskinesia is a complication of levodopa therapy and negatively impacts the quality of life of patients. We aimed to elucidate white matter alterations in Parkinson's disease with levodopa-induced dyskinesia using advanced diffusion magnetic resonance imaging techniques. METHODS The enrolled subjects included 26 clinically confirmed Parkinson's disease patients without levodopa-induced dyskinesia, 25 Parkinson's disease patients with levodopa-induced dyskinesia, and 23 healthy controls. Subjects were imaged using a 3-T magnetic resonance scanner. Diffusion tensor imaging, diffusion kurtosis imaging, and neurite orientation dispersion and density imaging findings were compared between groups with a group-wise whole brain approach and a region-of-interest analysis for each white matter tract. Additionally, logistic regression analysis was used to calculate odds ratios for levodopa-induced dyskinesia. RESULTS Group-wise tract-based spatial statistical analysis revealed significant white matter differences in isotropic diffusion, complexity, or heterogeneity, and neurite density between healthy controls and Parkinson's disease patients without levodopa-induced dyskinesia and between patients with and without levodopa-induced dyskinesia. Region-of-interest analysis revealed similar alterations using a group-wise whole-brain approach in the external capsule, inferior fronto-occipital fasciculus, inferior longitudinal fasciculus, and uncinate fasciculus. These tracts had an odds ratio of approximately 2.3 for the presence of levodopa-induced dyskinesia. CONCLUSIONS Our findings suggest that Parkinson's disease with levodopa-induced dyskinesia produces less white matter microstructural disruption, especially in temporal lobe fibers, than Parkinson's disease without levodopa-induced dyskinesia. These fibers has a more than 2-fold odds ratio for the presence of levodopa-induced dyskinesia and might be associated with the pathogenesis of the sequela.
Collapse
|
9
|
Fieblinger T. Striatal Control of Movement: A Role for New Neuronal (Sub-) Populations? Front Hum Neurosci 2021; 15:697284. [PMID: 34354577 PMCID: PMC8329243 DOI: 10.3389/fnhum.2021.697284] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2021] [Accepted: 06/29/2021] [Indexed: 12/19/2022] Open
Abstract
The striatum is a very heterogenous brain area, composed of different domains and compartments, albeit lacking visible anatomical demarcations. Two populations of striatal spiny projection neurons (SPNs) build the so-called direct and indirect pathway of the basal ganglia, whose coordinated activity is essential to control locomotion. Dysfunction of striatal SPNs is part of many movement disorders, such as Parkinson’s disease (PD) and L-DOPA-induced dyskinesia. In this mini review article, I will highlight recent studies utilizing single-cell RNA sequencing to investigate the transcriptional profiles of striatal neurons. These studies discover that SPNs carry a transcriptional signature, indicating both their anatomical location and compartmental identity. Furthermore, the transcriptional profiles reveal the existence of additional distinct neuronal populations and previously unknown SPN sub-populations. In a parallel development, studies in rodent models of PD and L-DOPA-induced dyskinesia (LID) report that direct pathway SPNs do not react uniformly to L-DOPA therapy, and that only a subset of these neurons is underlying the development of abnormal movements. Together, these studies demonstrate a new level of cellular complexity for striatal (dys-) function and locomotor control.
Collapse
Affiliation(s)
- Tim Fieblinger
- Institute for Synaptic Physiology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| |
Collapse
|
10
|
Yang K, Zhao X, Wang C, Zeng C, Luo Y, Sun T. Circuit Mechanisms of L-DOPA-Induced Dyskinesia (LID). Front Neurosci 2021; 15:614412. [PMID: 33776634 PMCID: PMC7988225 DOI: 10.3389/fnins.2021.614412] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2020] [Accepted: 02/05/2021] [Indexed: 12/25/2022] Open
Abstract
L-DOPA is the criterion standard of treatment for Parkinson disease. Although it alleviates some of the Parkinsonian symptoms, long-term treatment induces L-DOPA–induced dyskinesia (LID). Several theoretical models including the firing rate model, the firing pattern model, and the ensemble model are proposed to explain the mechanisms of LID. The “firing rate model” proposes that decreasing the mean firing rates of the output nuclei of basal ganglia (BG) including the globus pallidus internal segment and substantia nigra reticulata, along the BG pathways, induces dyskinesia. The “firing pattern model” claimed that abnormal firing pattern of a single unit activity and local field potentials may disturb the information processing in the BG, resulting in dyskinesia. The “ensemble model” described that dyskinesia symptoms might represent a distributed impairment involving many brain regions, but the number of activated neurons in the striatum correlated most strongly with dyskinesia severity. Extensive evidence for circuit mechanisms in driving LID symptoms has also been presented. LID is a multisystem disease that affects wide areas of the brain. Brain regions including the striatum, the pallidal–subthalamic network, the motor cortex, the thalamus, and the cerebellum are all involved in the pathophysiology of LID. In addition, although both amantadine and deep brain stimulation help reduce LID, these approaches have complications that limit their wide use, and a novel antidyskinetic drug is strongly needed; these require us to understand the circuit mechanism of LID more deeply.
Collapse
Affiliation(s)
- Kai Yang
- School of Chemistry, Chemical Engineering and Life Science, Wuhan University of Technology, Wuhan, China
| | - Xinyue Zhao
- School of Chemistry, Chemical Engineering and Life Science, Wuhan University of Technology, Wuhan, China
| | - Changcai Wang
- School of Chemistry, Chemical Engineering and Life Science, Wuhan University of Technology, Wuhan, China
| | - Cheng Zeng
- School of Chemistry, Chemical Engineering and Life Science, Wuhan University of Technology, Wuhan, China
| | - Yan Luo
- Department of Physiology, School of Basic Medical Science, Ningxia Medical University, Yinchuan, China
| | - Taolei Sun
- School of Chemistry, Chemical Engineering and Life Science, Wuhan University of Technology, Wuhan, China.,State Key Laboratory of Advanced Technology for Materials Synthesis and Processing, Wuhan University of Technology, Wuhan, China
| |
Collapse
|
11
|
Outeiro TF, Heutink P, Bezard E, Cenci AM. From iPS Cells to Rodents and Nonhuman Primates: Filling Gaps in Modeling Parkinson's Disease. Mov Disord 2020; 36:832-841. [PMID: 33200446 DOI: 10.1002/mds.28387] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2020] [Revised: 10/12/2020] [Accepted: 10/27/2020] [Indexed: 12/20/2022] Open
Abstract
Parkinson's disease (PD) is primarily known as a movement disorder because of typical clinical manifestations associated with the loss of dopaminergic neurons in the substantia nigra. However, it is now widely recognized that PD is a much more complex condition, with multiple and severe nonmotor features implicating additional brain areas and organs in the disease process. Pathologically, typical forms of PD are characterized by the accumulation of α-synuclein-rich protein inclusions known as Lewy bodies and Lewy neurites, although other types of protein inclusions are also often present in the brain. Familial forms of PD have provided a wealth of information about molecular pathways leading to neurodegeneration, but only to add to the complexity of the problem and uncover new knowledge gaps. Therefore, modeling PD in the laboratory has become increasingly challenging. Here, we discuss knowledge gaps and challenges in the use of laboratory models for the study of a disease that is clinically heterogeneous and multifactorial. We propose that the combined use of patient-derived cells and animal models, along with current technological tools, will not only expand our molecular and pathophysiological understanding of PD, but also assist in the identification of therapeutic strategies targeting relevant pathogenic pathways. © 2020 International Parkinson and Movement Disorder Society.
Collapse
Affiliation(s)
- Tiago F Outeiro
- Department of Experimental Neurodegeneration, Center for Biostructural Imaging of Neurodegeneration, University Medical Center Goettingen, Goettingen, Germany.,Max Planck Institute for Experimental Medicine, Goettingen, Germany.,Translational and Clinical Research Institute, Faculty of Medical Sciences, Newcastle University, Newcastle Upon Tyne, UK
| | - Peter Heutink
- German Center for Neurodegenerative Diseases, Tübingen, Germany
| | - Erwan Bezard
- Univ. de Bordeaux, Institut des Maladies Neurodégénératives, UMR 5293, Bordeaux, France.,CNRS, Institut des Maladies Neurodégénératives, UMR 5293, Bordeaux, France
| | - Angela M Cenci
- Department of Experimental Medical Science, Basal Ganglia Pathophysiology Unit, Lund University, Lund, Sweden
| |
Collapse
|
12
|
Fletcher EJR, Finlay CJ, Amor Lopez A, Crum WR, Vernon AC, Duty S. Neuroanatomical and Microglial Alterations in the Striatum of Levodopa-Treated, Dyskinetic Hemi-Parkinsonian Rats. Front Neurosci 2020; 14:567222. [PMID: 33041762 PMCID: PMC7522511 DOI: 10.3389/fnins.2020.567222] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2020] [Accepted: 08/17/2020] [Indexed: 02/02/2023] Open
Abstract
Dyskinesia associated with chronic levodopa treatment in Parkinson’s disease is associated with maladaptive striatal plasticity. The objective of this study was to examine whether macroscale structural changes, as captured by magnetic resonance imaging (MRI) accompany this plasticity and to identify plausible cellular contributors in a rodent model of levodopa-induced dyskinesia. Adult male Sprague-Dawley rats were rendered hemi-parkinsonian by stereotaxic injection of 6-hydroxydopamine into the left medial forebrain bundle prior to chronic treatment with saline (control) or levodopa to induce abnormal involuntary movements (AIMs), reflective of dyskinesia. Perfusion-fixed brains underwent ex vivo structural MRI before sectioning and staining for cellular markers. Chronic treatment with levodopa induced significant AIMs (p < 0.0001 versus saline). The absolute volume of the ipsilateral, lesioned striatum was increased in levodopa-treated rats resulting in a significant difference in percentage volume change when compared to saline-treated rats (p < 0.01). Moreover, a significant positive correlation was found between this volume change and AIMs scores for individual levodopa-treated rats (r = 0.96; p < 0.01). The density of Iba1+ cells was increased within the lesioned versus intact striatum (p < 0.01) with no difference between treatment groups. Conversely, Iba1+ microglia soma size was significantly increased (p < 0.01) in the lesioned striatum of levodopa-treated but not saline-treated rats. Soma size was not, however, significantly correlated with either AIMs or MRI volume change. Although GFAP+ astrocytes were elevated in the lesioned versus intact striatum (p < 0.001), there was no difference between treatment groups. No statistically significant effects of either lesion or treatment on RECA1, a marker for blood vessels, were observed. Collectively, these data suggest chronic levodopa treatment in 6-hydroxydopamine lesioned rats is associated with increased striatal volume that correlates with the development of AIMs. The accompanying increase in number and size of microglia, however, cannot alone explain this volume expansion. Further multi-modal studies are warranted to establish the brain-wide effects of chronic levodopa treatment.
Collapse
Affiliation(s)
- Edward J R Fletcher
- Wolfson Centre for Age Related Diseases, Wolfson Wing, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, United Kingdom
| | - Clare J Finlay
- Wolfson Centre for Age Related Diseases, Wolfson Wing, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, United Kingdom
| | - Ana Amor Lopez
- Department of Neuroimaging, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, United Kingdom
| | - William R Crum
- Department of Neuroimaging, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, United Kingdom
| | - Anthony C Vernon
- Department of Basic and Clinical Neuroscience, Maurice Wohl Clinical Neuroscience Institute, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, United Kingdom.,MRC Centre for Neurodevelopmental Disorders, King's College London, London, United Kingdom
| | - Susan Duty
- Wolfson Centre for Age Related Diseases, Wolfson Wing, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, United Kingdom
| |
Collapse
|
13
|
Striatal Nurr1 Facilitates the Dyskinetic State and Exacerbates Levodopa-Induced Dyskinesia in a Rat Model of Parkinson's Disease. J Neurosci 2020; 40:3675-3691. [PMID: 32238479 DOI: 10.1523/jneurosci.2936-19.2020] [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: 12/10/2019] [Revised: 03/13/2020] [Accepted: 03/19/2020] [Indexed: 12/23/2022] Open
Abstract
The transcription factor Nurr1 has been identified to be ectopically induced in the striatum of rodents expressing l-DOPA-induced dyskinesia (LID). In the present study, we sought to characterize Nurr1 as a causative factor in LID expression. We used rAAV2/5 to overexpress Nurr1 or GFP in the parkinsonian striatum of LID-resistant Lewis or LID-prone Fischer-344 (F344) male rats. In a second cohort, rats received the Nurr1 agonist amodiaquine (AQ) together with l-DOPA or ropinirole. All rats received a chronic DA agonist and were evaluated for LID severity. Finally, we performed single-unit recordings and dendritic spine analyses on striatal medium spiny neurons (MSNs) in drug-naïve rAAV-injected male parkinsonian rats. rAAV-GFP injected LID-resistant hemi-parkinsonian Lewis rats displayed mild LID and no induction of striatal Nurr1 despite receiving a high dose of l-DOPA. However, Lewis rats overexpressing Nurr1 developed severe LID. Nurr1 agonism with AQ exacerbated LID in F344 rats. We additionally determined that in l-DOPA-naïve rats striatal rAAV-Nurr1 overexpression (1) increased cortically-evoked firing in a subpopulation of identified striatonigral MSNs, and (2) altered spine density and thin-spine morphology on striatal MSNs; both phenomena mimicking changes seen in dyskinetic rats. Finally, we provide postmortem evidence of Nurr1 expression in striatal neurons of l-DOPA-treated PD patients. Our data demonstrate that ectopic induction of striatal Nurr1 is capable of inducing LID behavior and associated neuropathology, even in resistant subjects. These data support a direct role of Nurr1 in aberrant neuronal plasticity and LID induction, providing a potential novel target for therapeutic development.SIGNIFICANCE STATEMENT The transcription factor Nurr1 is ectopically induced in striatal neurons of rats exhibiting levodopa-induced dyskinesia [LID; a side-effect to dopamine replacement strategies in Parkinson's disease (PD)]. Here we asked whether Nurr1 is causing LID. Indeed, rAAV-mediated expression of Nurr1 in striatal neurons was sufficient to overcome LID-resistance, and Nurr1 agonism exacerbated LID severity in dyskinetic rats. Moreover, we found that expression of Nurr1 in l-DOPA naïve hemi-parkinsonian rats resulted in the formation of morphologic and electrophysiological signatures of maladaptive neuronal plasticity; a phenomenon associated with LID. Finally, we determined that ectopic Nurr1 expression can be found in the putamen of l-DOPA-treated PD patients. These data suggest that striatal Nurr1 is an important mediator of the formation of LID.
Collapse
|
14
|
Cenci MA, Björklund A. Animal models for preclinical Parkinson's research: An update and critical appraisal. PROGRESS IN BRAIN RESEARCH 2020; 252:27-59. [PMID: 32247366 DOI: 10.1016/bs.pbr.2020.02.003] [Citation(s) in RCA: 37] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Animal models of Parkinson's disease (PD) are essential to investigate pathogenic pathways at the whole-organism level. Moreover, they are necessary for a preclinical investigation of potential new therapies. Different pathological features of PD can be induced in a variety of invertebrate and vertebrate species using toxins, drugs, or genetic perturbations. Each model has a particular utility and range of applicability. Invertebrate PD models are particularly useful for high throughput-screening applications, whereas mammalian models are needed to explore complex motor and non-motor features of the human disease. Here, we provide a comprehensive review and critical appraisal of the most commonly used mammalian models of PD, which are produced in rats and mice. A substantial loss of nigrostriatal dopamine neurons is necessary for the animal to exhibit a hypokinetic motor phenotype responsive to dopaminergic agents, thus resembling clinical PD. This level of dopaminergic neurodegeneration can be induced using specific neurotoxins, environmental toxicants, or proteasome inhibitors. Alternatively, nigrostriatal dopamine degeneration can be induced via overexpression of α-synuclein using viral vectors or transgenic techniques. In addition, protein aggregation pathology can be triggered by inoculating preformed fibrils of α-synuclein in the substantia nigra or the striatum. Thanks to the conceptual and technical progress made in the past few years a vast repertoire of well-characterized animal models are currently available to address different aspects of PD in the laboratory.
Collapse
Affiliation(s)
- M Angela Cenci
- Department of Experimental Medical Science, Wallenberg Neuroscience Centre, Lund University, Lund, Sweden.
| | - Anders Björklund
- Department of Experimental Medical Science, Wallenberg Neuroscience Centre, Lund University, Lund, Sweden
| |
Collapse
|
15
|
Ray Chaudhuri K, Poewe W, Brooks D. Motor and Nonmotor Complications of Levodopa: Phenomenology, Risk Factors, and Imaging Features. Mov Disord 2019; 33:909-919. [PMID: 30134055 DOI: 10.1002/mds.27386] [Citation(s) in RCA: 84] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2017] [Revised: 02/16/2018] [Accepted: 02/16/2018] [Indexed: 12/14/2022] Open
Abstract
Despite enormous advances in our current understanding of PD since James Parkinson described the "shaking palsy" 200 years ago, l-dopa, in clinical use since the 1960s, remains the gold standard of treatment. Virtually every patient with PD requires varying doses of l-dopa to manage motor and some nonmotor symptoms and retain an acceptable quality of life. However, after a period of treatment with l-dopa, a number of problems emerge; the key ones are motor and nonmotor fluctuations, a range of dyskinesias, and a combination of both. Nonmotor complications can range from behavioral problems to sensory, autonomic, and cognitive issues. Even with a wealth of data, both in animal models and in vivo imaging that address the pathophysiology of l-dopa-related motor and nonmotor complications, the treatment remains challenging and is an unmet need. Although refinement in types of dopamine replacement therapy and delivery systems have improved the management of l-dopa-related complications, the search for the ideal treatment continues. © 2018 International Parkinson and Movement Disorder Society.
Collapse
Affiliation(s)
- K Ray Chaudhuri
- Institute of Psychiatry, Psychology & Neuroscience at King's College London and Parkinsons Foundation Centre of Excellence at King's College Hospital NHS Foundation Trust
| | - Werner Poewe
- Department of Neurology, Medical University Innsbruck, Innsbruck, Austria
| | - David Brooks
- Department of Medicine, Imperial College London, London, United Kingdom
| |
Collapse
|
16
|
Steece‐Collier K, Stancati JA, Collier NJ, Sandoval IM, Mercado NM, Sortwell CE, Collier TJ, Manfredsson FP. Genetic silencing of striatal CaV1.3 prevents and ameliorates levodopa dyskinesia. Mov Disord 2019; 34:697-707. [PMID: 31002755 PMCID: PMC6563183 DOI: 10.1002/mds.27695] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2018] [Revised: 03/19/2019] [Accepted: 03/21/2019] [Indexed: 01/26/2023] Open
Abstract
BACKGROUND Levodopa-induced dyskinesias are an often debilitating side effect of levodopa therapy in Parkinson's disease. Although up to 90% of individuals with PD develop this side effect, uniformly effective and well-tolerated antidyskinetic treatment remains a significant unmet need. The pathognomonic loss of striatal dopamine in PD results in dysregulation and disinhibition of striatal CaV1.3 calcium channels, leading to synaptopathology that appears to be involved in levodopa-induced dyskinesias. Although there are clinically available drugs that can inhibit CaV1.3 channels, they are not adequately potent and have only partial and transient impact on levodopa-induced dyskinesias. METHODS To provide unequivocal target validation, free of pharmacological limitations, we developed a CaV1.3 shRNA to provide high-potency, target-selective, mRNA-level silencing of striatal CaV1.3 channels and examined its ability to impact levodopa-induced dyskinesias in severely parkinsonian rats. RESULTS We demonstrate that vector-mediated silencing of striatal CaV1.3 expression in severely parkinsonian rats prior to the introduction of levodopa can uniformly and completely prevent induction of levodopa-induced dyskinesias, and this antidyskinetic benefit persists long term and with high-dose levodopa. In addition, this approach is capable of ameliorating preexisting severe levodopa-induced dyskinesias. Importantly, motoric responses to low-dose levodopa remained intact in the presence of striatal CaV1.3 silencing, indicating preservation of levodopa benefit without dyskinesia liability. DISCUSSION The current data provide some of the most profound antidyskinetic benefit reported to date and suggest that genetic silencing of striatal CaV1.3 channels has the potential to transform treatment of individuals with PD by allowing maintenance of motor benefit of levodopa in the absence of the debilitating levodopa-induced dyskinesia side effect. © 2019 The Authors. Movement Disorders published by Wiley Periodicals, Inc. on behalf of International Parkinson and Movement Disorder Society.
Collapse
Affiliation(s)
- Kathy Steece‐Collier
- Department of Translational Science & Molecular MedicineCollege of Human Medicine, Michigan State UniversityGrand RapidsMIUSA
- Hauenstein Neuroscience CenterMercy Health Saint Mary's, Grand RapidsMichiganUSA
| | - Jennifer A. Stancati
- Department of Translational Science & Molecular MedicineCollege of Human Medicine, Michigan State UniversityGrand RapidsMIUSA
| | - Nicholas J. Collier
- Department of Translational Science & Molecular MedicineCollege of Human Medicine, Michigan State UniversityGrand RapidsMIUSA
| | - Ivette M. Sandoval
- Department of Translational Science & Molecular MedicineCollege of Human Medicine, Michigan State UniversityGrand RapidsMIUSA
- Hauenstein Neuroscience CenterMercy Health Saint Mary's, Grand RapidsMichiganUSA
| | - Natosha M. Mercado
- Department of Translational Science & Molecular MedicineCollege of Human Medicine, Michigan State UniversityGrand RapidsMIUSA
| | - Caryl E. Sortwell
- Department of Translational Science & Molecular MedicineCollege of Human Medicine, Michigan State UniversityGrand RapidsMIUSA
- Hauenstein Neuroscience CenterMercy Health Saint Mary's, Grand RapidsMichiganUSA
| | - Timothy J. Collier
- Department of Translational Science & Molecular MedicineCollege of Human Medicine, Michigan State UniversityGrand RapidsMIUSA
- Hauenstein Neuroscience CenterMercy Health Saint Mary's, Grand RapidsMichiganUSA
| | - Fredric P. Manfredsson
- Department of Translational Science & Molecular MedicineCollege of Human Medicine, Michigan State UniversityGrand RapidsMIUSA
- Hauenstein Neuroscience CenterMercy Health Saint Mary's, Grand RapidsMichiganUSA
| |
Collapse
|
17
|
Eltoprazine prevents levodopa-induced dyskinesias by reducing causal interactions for theta oscillations in the dorsolateral striatum and substantia nigra pars reticulate. Neuropharmacology 2019; 148:1-10. [PMID: 30612008 DOI: 10.1016/j.neuropharm.2018.12.027] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2018] [Revised: 12/22/2018] [Accepted: 12/24/2018] [Indexed: 12/21/2022]
Abstract
Oscillatory activities within basal ganglia (BG) circuitry in L-DOPA induced dyskinesia (LID), a condition that occurs in patients with Parkinson disease (PD), are not well understood. The aims of this study were firstly to investigate oscillations in main BG input and output structures-the dorsolateral striatum (dStr) and substantia nigra pars reticulata (SNr), respectively- including the direction of oscillation information flow, and secondly to investigate the effects of 5-HT1A/B receptor agonism with eltoprazine on oscillatory activities and abnormal involuntary movements (AIMs) characteristic. To this end, we conducted local field potential (LFP) electrophysiology in the dStr and SNr of LID rats simultaneous with AIM scoring. The LFP data were submitted to power spectral density, coherence, and partial Granger causality analyses. AIM data were analyzed relative to simultaneous oscillatory activities, with and without eltoprazine. We obtained four major findings. 1) Theta band (5-8 Hz) oscillations were enhanced in the dStr and SNr of LID rats. 2) Theta power correlated with AIM scores in the 180-min period after the last LID-inducing L-DOPA injection, but not with daily summed AIM scores during LID development. 3) Oscillatory information flowed from the dStr to the SNr. 4) Chronic eltoprazine reduced BG theta activity in LID rats and normalized information flow directionality, relative to that in LID rats not given eltoprazine. These results indicate that dStr activity plays a determinative role in the causal interactions of theta oscillations and that serotonergic inhibition may suppress dyskinesia by reducing dStr-SNr theta activity and restoring theta network information flow.
Collapse
|
18
|
Plotkin JL, Goldberg JA. Thinking Outside the Box (and Arrow): Current Themes in Striatal Dysfunction in Movement Disorders. Neuroscientist 2018; 25:359-379. [PMID: 30379121 PMCID: PMC6529282 DOI: 10.1177/1073858418807887] [Citation(s) in RCA: 31] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023]
Abstract
The basal ganglia are an intricately connected assembly of subcortical nuclei, forming the core of an adaptive network connecting cortical and thalamic circuits. For nearly three decades, researchers and medical practitioners have conceptualized how the basal ganglia circuit works, and how its pathology underlies motor disorders such as Parkinson's and Huntington's diseases, using what is often referred to as the "box-and-arrow model": a circuit diagram showing the broad strokes of basal ganglia connectivity and the pathological increases and decreases in the weights of specific connections that occur in disease. While this model still has great utility and has led to groundbreaking strategies to treat motor disorders, our evolving knowledge of basal ganglia function has made it clear that this classic model has several shortcomings that severely limit its predictive and descriptive abilities. In this review, we will focus on the striatum, the main input nucleus of the basal ganglia. We describe recent advances in our understanding of the rich microcircuitry and plastic capabilities of the striatum, factors not captured by the original box-and-arrow model, and provide examples of how such advances inform our current understanding of the circuit pathologies underlying motor disorders.
Collapse
Affiliation(s)
- Joshua L Plotkin
- Department of Neurobiology and Behavior, Stony Brook University School of Medicine, Stony Brook, NY, USA
| | - Joshua A Goldberg
- Department of Medical Neurobiology, The Hebrew University of Jerusalem, Jerusalem, Israel
| |
Collapse
|
19
|
Fieblinger T, Zanetti L, Sebastianutto I, Breger LS, Quintino L, Lockowandt M, Lundberg C, Cenci MA. Striatonigral neurons divide into two distinct morphological-physiological phenotypes after chronic L-DOPA treatment in parkinsonian rats. Sci Rep 2018; 8:10068. [PMID: 29968767 PMCID: PMC6030109 DOI: 10.1038/s41598-018-28273-5] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2018] [Accepted: 06/18/2018] [Indexed: 12/12/2022] Open
Abstract
Dendritic regression of striatal spiny projection neurons (SPNs) is a pathological hallmark of Parkinson's disease (PD). Here we investigate how chronic dopamine denervation and dopamine replacement with L-DOPA affect the morphology and physiology of direct pathway SPNs (dSPNS) in the rat striatum. We used a lentiviral vector optimized for retrograde labeling (FuG-B-GFP) to identify dSPNs in rats with 6-hydroxydopamine (6-OHDA) lesions. Changes in morphology and physiology of dSPNs were assessed through a combination of patch-clamp recordings and two photon microscopy. The 6-OHDA lesion caused a significant reduction in dSPN dendritic complexity. Following chronic L-DOPA treatment, dSPNs segregated into two equal-sized clusters. One group (here called "cluster-1"), showed sustained dendritic atrophy and a partially normalized electrophysiological phenotype. The other one ("cluster-2") exhibited dendritic regrowth and a strong reduction of intrinsic excitability. Interestingly, FosB/∆FosB induction by L-DOPA treatment occurred preferentially in cluster-2 dSPNs. Our study demonstrates the feasibility of retrograde FuG-B-GFP labeling to study dSPNs in the rat and reveals, for the first time, that a subgroup of dSPNs shows dendritic sprouting in response to chronic L-DOPA treatment. Investigating the mechanisms and significance of this response will greatly improve our understanding of the adaptations induced by dopamine replacement therapy in PD.
Collapse
Affiliation(s)
- T Fieblinger
- Basal Ganglia Pathophysiology Unit, Department of Experimental Medical Science, Lund University, Lund, Sweden. .,Wissenschaftskolleg zu Berlin, Institute for Advanced Study, Wallotstr. 19, D-14193, Berlin, Germany.
| | - L Zanetti
- Basal Ganglia Pathophysiology Unit, Department of Experimental Medical Science, Lund University, Lund, Sweden.,Institute of Pharmacy, Pharmacology and Toxicology, Center for Molecular Biosciences, University of Innsbruck, Innsbruck, Austria
| | - I Sebastianutto
- Basal Ganglia Pathophysiology Unit, Department of Experimental Medical Science, Lund University, Lund, Sweden
| | - L S Breger
- CNS Gene Therapy, Department of Experimental Medical Science, Lund University, Lund, Sweden.,CNRS, Institut des Maladies Neurodégénératives, University of Bordeaux, Bordeaux, France
| | - L Quintino
- CNS Gene Therapy, Department of Experimental Medical Science, Lund University, Lund, Sweden
| | - M Lockowandt
- CNS Gene Therapy, Department of Experimental Medical Science, Lund University, Lund, Sweden
| | - C Lundberg
- CNS Gene Therapy, Department of Experimental Medical Science, Lund University, Lund, Sweden
| | - M A Cenci
- Basal Ganglia Pathophysiology Unit, Department of Experimental Medical Science, Lund University, Lund, Sweden.
| |
Collapse
|
20
|
Rouillard C, Baillargeon J, Paquet B, St-Hilaire M, Maheux J, Lévesque C, Darlix N, Majeur S, Lévesque D. Genetic disruption of the nuclear receptor Nur77 (Nr4a1) in rat reduces dopamine cell loss and l-Dopa-induced dyskinesia in experimental Parkinson's disease. Exp Neurol 2018. [DOI: 10.1016/j.expneurol.2018.03.008] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023]
|
21
|
Cenci MA, Jörntell H, Petersson P. On the neuronal circuitry mediating L-DOPA-induced dyskinesia. J Neural Transm (Vienna) 2018; 125:1157-1169. [PMID: 29704061 PMCID: PMC6060876 DOI: 10.1007/s00702-018-1886-0] [Citation(s) in RCA: 39] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2018] [Accepted: 04/17/2018] [Indexed: 11/27/2022]
Abstract
With the advent of rodent models of l-DOPA-induced dyskinesia (LID), a growing literature has linked molecular changes in the striatum to the development and expression of abnormal involuntary movements. Changes in information processing at the striatal level are assumed to impact on the activity of downstream basal ganglia nuclei, which in turn influence brain-wide networks, but very little is actually known about systems-level mechanisms of dyskinesia. As an aid to approach this topic, we here review the anatomical and physiological organisation of cortico-basal ganglia-thalamocortical circuits, and the changes affecting these circuits in animal models of parkinsonism and LID. We then review recent findings indicating that an abnormal cerebellar compensation plays a causal role in LID, and that structures outside of the classical motor circuits are implicated too. In summarizing the available data, we also propose hypotheses and identify important knowledge gaps worthy of further investigation. In addition to informing novel therapeutic approaches, the study of LID can provide new clues about the interplay between different brain circuits in the control of movement.
Collapse
Affiliation(s)
- M Angela Cenci
- Basal Ganglia Pathophysiology Unit, Department Experimental Medical Science, Lund University, Lund, Sweden.
| | - Henrik Jörntell
- Neural Basis of Sensorimotor Control, Department Experimental Medical Science, Lund University, Lund, Sweden
| | - Per Petersson
- The Group for Integrative Neurophysiology and Neurotechnology, Neuronano Research Centre, Department Experimental Medical Science, Lund University, Lund, Sweden
- Department of Integrative Medical Biology, Umeå University, Umeå, Sweden
| |
Collapse
|
22
|
You H, Mariani LL, Mangone G, Le Febvre de Nailly D, Charbonnier-Beaupel F, Corvol JC. Molecular basis of dopamine replacement therapy and its side effects in Parkinson's disease. Cell Tissue Res 2018. [PMID: 29516217 DOI: 10.1007/s00441-018-2813-2] [Citation(s) in RCA: 57] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
There is currently no cure for Parkinson's disease. The symptomatic therapeutic strategy essentially relies on dopamine replacement whose efficacy was demonstrated more than 50 years ago following the introduction of the dopamine precursor, levodopa. The spectacular antiparkinsonian effect of levodopa is, however, balanced by major limitations including the occurrence of motor complications related to its particular pharmacokinetic and pharmacodynamic properties. Other therapeutic strategies have thus been developed to overcome these problems such as the use of dopamine receptor agonists, dopamine metabolism inhibitors and non-dopaminergic drugs. Here we review the pharmacology and molecular mechanisms of dopamine replacement therapy in Parkinson's disease, both at the presynaptic and postsynaptic levels. The perspectives in terms of novel drug development and prediction of drug response for a more personalised medicine will be discussed.
Collapse
Affiliation(s)
- Hana You
- Sorbonne Université, UPMC Univ Paris 06, UMR S 1127, ICM, Hôpital Pitié-Salpêtrière, Paris, France.,INSERM, Unit 1127, CIC 1422, NS-PARK/FCRIN, Hôpital Pitié-Salpêtrière, Paris, France.,CNRS, Unit 7225, Hôpital Pitié-Salpêtrière, Paris, France.,Assistance Publique Hôpitaux de Paris, Department of Neurology, Hôpital Pitié-Salpêtrière, Paris, France.,Department of Neurology, University Hospital (Inselspital) and University of Bern, Freiburgstrasse 18, 3010, Bern, Switzerland
| | - Louise-Laure Mariani
- Sorbonne Université, UPMC Univ Paris 06, UMR S 1127, ICM, Hôpital Pitié-Salpêtrière, Paris, France.,INSERM, Unit 1127, CIC 1422, NS-PARK/FCRIN, Hôpital Pitié-Salpêtrière, Paris, France.,CNRS, Unit 7225, Hôpital Pitié-Salpêtrière, Paris, France.,Assistance Publique Hôpitaux de Paris, Department of Neurology, Hôpital Pitié-Salpêtrière, Paris, France
| | - Graziella Mangone
- Sorbonne Université, UPMC Univ Paris 06, UMR S 1127, ICM, Hôpital Pitié-Salpêtrière, Paris, France.,INSERM, Unit 1127, CIC 1422, NS-PARK/FCRIN, Hôpital Pitié-Salpêtrière, Paris, France.,CNRS, Unit 7225, Hôpital Pitié-Salpêtrière, Paris, France.,Assistance Publique Hôpitaux de Paris, Department of Neurology, Hôpital Pitié-Salpêtrière, Paris, France
| | - Delphine Le Febvre de Nailly
- INSERM, Unit 1127, CIC 1422, NS-PARK/FCRIN, Hôpital Pitié-Salpêtrière, Paris, France.,Assistance Publique Hôpitaux de Paris, Department of Pharmacy, Hôpital Pitié-Salpêtrière, Paris, France
| | - Fanny Charbonnier-Beaupel
- Assistance Publique Hôpitaux de Paris, Department of Pharmacy, Hôpital Pitié-Salpêtrière, Paris, France
| | - Jean-Christophe Corvol
- Sorbonne Université, UPMC Univ Paris 06, UMR S 1127, ICM, Hôpital Pitié-Salpêtrière, Paris, France. .,INSERM, Unit 1127, CIC 1422, NS-PARK/FCRIN, Hôpital Pitié-Salpêtrière, Paris, France. .,CNRS, Unit 7225, Hôpital Pitié-Salpêtrière, Paris, France. .,Assistance Publique Hôpitaux de Paris, Department of Neurology, Hôpital Pitié-Salpêtrière, Paris, France. .,CIC Neurosciences, ICM building, Hôpital Pitié-Salpêtrière, 47/83 Boulevard de l'Hôpital, 75013, Paris, France.
| |
Collapse
|
23
|
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: 4.4] [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
|
24
|
Differential Synaptic Remodeling by Dopamine in Direct and Indirect Striatal Projection Neurons in Pitx3 -/- Mice, a Genetic Model of Parkinson's Disease. J Neurosci 2018; 38:3619-3630. [PMID: 29483281 DOI: 10.1523/jneurosci.3184-17.2018] [Citation(s) in RCA: 52] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2017] [Revised: 12/23/2017] [Accepted: 01/18/2018] [Indexed: 02/06/2023] Open
Abstract
In toxin-based models of Parkinson's disease (PD), striatal projection neurons (SPNs) exhibit dendritic atrophy and spine loss concurrent with an increase in excitability. Chronic l-DOPA treatment that induces dyskinesia selectively restores spine density and excitability in indirect pathway SPNs (iSPNs), whereas spine loss and hyperexcitability persist in direct pathway SPNs (dSPNs). These alterations have only been characterized in toxin-based models of PD, raising the possibility that they are an artifact of exposure to the toxin, which may engage compensatory mechanisms independent of the PD-like pathology or due to the loss of dopaminergic afferents. To test all these, we studied the synaptic remodeling in Pitx3-/- or aphakia mice, a genetic model of PD, in which most of the dopamine neurons in the substantia nigra fail to fully differentiate and to innervate the striatum. We made 3D reconstructions of the dendritic arbor and measured excitability in identified SPNs located in dorsal striatum of BAC-Pitx3-/- mice treated with saline or l-DOPA. Both dSPNs and iSPNs from BAC-Pitx3-/- mice had shorter dendritic trees, lower spine density, and more action potentials than their counterparts from WT mice. Chronic l-DOPA treatment restored spine density and firing rate in iSPNs. By contrast, in dSPNs, spine loss and hyperexcitability persisted following l-DOPA treatment, which is similar to what happens in 6-OHDA WT mice. This indicates that dopamine-mediated synaptic remodeling and plasticity is independent of dopamine innervation during SPN development and that Pitx3-/- mice are a good model because they develop the same pathology described in the toxins-based models and in human postmortem studies of advanced PD.SIGNIFICANCE STATEMENT As the only genetic model of Parkinson's disease (PD) that develops dyskinesia, Pitx3-/- mice reproduce the behavioral effects seen in humans and are a good system for studying dopamine-induced synaptic remodeling. The studies we present here establish that the structural and functional synaptic plasticity that occur in striatal projection neurons in PD and in l-DOPA-induced dyskinesia are specifically due to modulation of the neurotransmitter dopamine and are not artifacts of the use of chemical toxins in PD models. In addition, our findings provide evidence that synaptic plasticity in the Pitx3-/- mouse is similar to that seen in toxin models despite its lack of dopaminergic innervation of the striatum during development. Pitx3-/- mice reproduced the alterations described in patients with advanced PD and in well accepted toxin-based models of PD and dyskinesia. These results further consolidate the fidelity of the Pitx3-/- mouse as a PD model in which to study the morphological and physiological remodeling of striatal projection neurons by administration of l-DOPA and other drugs.
Collapse
|
25
|
Solís O, Moratalla R. Dopamine receptors: homomeric and heteromeric complexes in l-DOPA-induced dyskinesia. J Neural Transm (Vienna) 2018; 125:1187-1194. [DOI: 10.1007/s00702-018-1852-x] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2017] [Accepted: 01/31/2018] [Indexed: 10/18/2022]
|
26
|
Blesa J, Trigo-Damas I, Dileone M, Del Rey NLG, Hernandez LF, Obeso JA. Compensatory mechanisms in Parkinson's disease: Circuits adaptations and role in disease modification. Exp Neurol 2017; 298:148-161. [PMID: 28987461 DOI: 10.1016/j.expneurol.2017.10.002] [Citation(s) in RCA: 178] [Impact Index Per Article: 22.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2017] [Revised: 09/27/2017] [Accepted: 10/03/2017] [Indexed: 12/21/2022]
Abstract
The motor features of Parkinson's disease (PD) are well known to manifest only when striatal dopaminergic deficit reaches 60-70%. Thus, PD has a long pre-symptomatic and pre-motor evolution during which compensatory mechanisms take place to delay the clinical onset of disabling manifestations. Classic compensatory mechanisms have been attributed to changes and adjustments in the nigro-striatal system, such as increased neuronal activity in the substantia nigra pars compacta and enhanced dopamine synthesis and release in the striatum. However, it is not so clear currently that such changes occur early enough to account for the pre-symptomatic period. Other possible mechanisms relate to changes in basal ganglia and motor cortical circuits including the cerebellum. However, data from early PD patients are difficult to obtain as most studies have been carried out once the diagnosis and treatments have been established. Likewise, putative compensatory mechanisms taking place throughout disease evolution are nearly impossible to distinguish by themselves. Here, we review the evidence for the role of the best known and other possible compensatory mechanisms in PD. We also discuss the possibility that, although beneficial in practical terms, compensation could also play a deleterious role in disease progression.
Collapse
Affiliation(s)
- Javier Blesa
- HM CINAC, Hospital Universitario HM Puerta del Sur, Móstoles, Madrid, Spain; Biomedical Research Center of Neurodegenerative Diseases (CIBERNED), Instituto Carlos III, Madrid, Spain.
| | - Inés Trigo-Damas
- HM CINAC, Hospital Universitario HM Puerta del Sur, Móstoles, Madrid, Spain; Biomedical Research Center of Neurodegenerative Diseases (CIBERNED), Instituto Carlos III, Madrid, Spain
| | - Michele Dileone
- HM CINAC, Hospital Universitario HM Puerta del Sur, Móstoles, Madrid, Spain; Biomedical Research Center of Neurodegenerative Diseases (CIBERNED), Instituto Carlos III, Madrid, Spain
| | - Natalia Lopez-Gonzalez Del Rey
- HM CINAC, Hospital Universitario HM Puerta del Sur, Móstoles, Madrid, Spain; Biomedical Research Center of Neurodegenerative Diseases (CIBERNED), Instituto Carlos III, Madrid, Spain
| | - Ledia F Hernandez
- HM CINAC, Hospital Universitario HM Puerta del Sur, Móstoles, Madrid, Spain; Biomedical Research Center of Neurodegenerative Diseases (CIBERNED), Instituto Carlos III, Madrid, Spain
| | - José A Obeso
- HM CINAC, Hospital Universitario HM Puerta del Sur, Móstoles, Madrid, Spain; Biomedical Research Center of Neurodegenerative Diseases (CIBERNED), Instituto Carlos III, Madrid, Spain.
| |
Collapse
|
27
|
Gamma Oscillations in the Hyperkinetic State Detected with Chronic Human Brain Recordings in Parkinson's Disease. J Neurosci 2017; 36:6445-58. [PMID: 27307233 DOI: 10.1523/jneurosci.1128-16.2016] [Citation(s) in RCA: 232] [Impact Index Per Article: 29.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2016] [Accepted: 05/07/2016] [Indexed: 11/21/2022] Open
Abstract
UNLABELLED Hyperkinetic states are common in human movement disorders, but their neural basis remains uncertain. One such condition is dyskinesia, a serious adverse effect of medical and surgical treatment for Parkinson's disease (PD). To study this, we used a novel, totally implanted, bidirectional neural interface to obtain multisite long-term recordings. We focus our analysis on two patients with PD who experienced frequent dyskinesia and studied them both at rest and during voluntary movement. We show that dyskinesia is associated with a narrowband gamma oscillation in motor cortex between 60 and 90 Hz, a similar, though weaker, oscillation in subthalamic nucleus, and strong phase coherence between the two. Dyskinesia-related oscillations are minimally affected by voluntary movement. When dyskinesia persists during therapeutic deep brain stimulation (DBS), the peak frequency of this signal shifts to half the stimulation frequency. These findings suggest a circuit-level mechanism for the generation of dyskinesia as well as a promising control signal for closed-loop DBS. SIGNIFICANCE STATEMENT Oscillations in brain networks link functionally related brain areas to accomplish thought and action, but this mechanism may be altered or exaggerated by disease states. Invasive recording using implanted electrodes provides a degree of spatial and temporal resolution that is ideal for analysis of network oscillations. Here we used a novel, totally implanted, bidirectional neural interface for chronic multisite brain recordings in humans with Parkinson's disease. We characterized an oscillation between cortex and subcortical modulators that is associated with a serious adverse effect of therapy for Parkinson's disease: dyskinesia. The work shows how a perturbation in oscillatory dynamics might lead to a state of excessive movement and also suggests a possible biomarker for feedback-controlled neurostimulation to treat hyperkinetic disorders.
Collapse
|
28
|
Sebastianutto I, Cenci MA, Fieblinger T. Alterations of striatal indirect pathway neurons precede motor deficits in two mouse models of Huntington's disease. Neurobiol Dis 2017; 105:117-131. [PMID: 28578004 DOI: 10.1016/j.nbd.2017.05.011] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2017] [Revised: 04/26/2017] [Accepted: 05/29/2017] [Indexed: 11/15/2022] Open
Abstract
Striatal neurons forming the indirect pathway (iSPNs) are particularly vulnerable in Huntington's disease (HD). In this study we set out to investigate morphological and physiological alterations of iSPNs in two mouse models of HD with relatively slow disease progression (long CAG repeat R6/2 and zQ175-KI). Both were crossed with a transgenic mouse line expressing eGFP in iSPNs. Using the open-field and rotarod tests, we first defined two time points in relation to the occurrence of motor deficits in each model. Then, we investigated electrophysiological and morphological properties of iSPNs at both ages. Both HD models exhibited increased iSPN excitability already before the onset of motor deficits, associated with a reduced number of primary dendrites and decreased function of Kir- and voltage-gated potassium channels. Alterations that specifically occurred at symptomatic ages included increased calcium release by back-propagating action potentials in proximal dendrites, due to enhanced engagement of intracellular calcium stores. Moreover, motorically impaired mice of both HD models showed a reduction in iSPN spine density and progressive formation of huntingtin (Htt) aggregates in the striatum. Our study therefore reports iSPN-specific alterations relative to the development of a motor phenotype in two different mouse models of HD. While some alterations occur early and are partly non-progressive, others potentially provide a pathophysiological marker of an overt disease state.
Collapse
Affiliation(s)
- Irene Sebastianutto
- Basal Ganglia Pathophysiology Unit, Dept. of Experimental Medical Science, Lund University, 22184 Lund, Sweden
| | - Maria Angela Cenci
- Basal Ganglia Pathophysiology Unit, Dept. of Experimental Medical Science, Lund University, 22184 Lund, Sweden
| | - Tim Fieblinger
- Basal Ganglia Pathophysiology Unit, Dept. of Experimental Medical Science, Lund University, 22184 Lund, Sweden.
| |
Collapse
|
29
|
Villalba RM, Smith Y. Loss and remodeling of striatal dendritic spines in Parkinson's disease: from homeostasis to maladaptive plasticity? J Neural Transm (Vienna) 2017; 125:431-447. [PMID: 28540422 DOI: 10.1007/s00702-017-1735-6] [Citation(s) in RCA: 67] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2017] [Accepted: 05/10/2017] [Indexed: 12/20/2022]
Abstract
In Parkinson's disease (PD) patients and animal models of PD, the progressive degeneration of the nigrostriatal dopamine (DA) projection leads to two major changes in the morphology of striatal projection neurons (SPNs), i.e., a profound loss of dendritic spines and the remodeling of axospinous glutamatergic synapses. Striatal spine loss is an early event tightly associated with the extent of striatal DA denervation, but not the severity of parkinsonian motor symptoms, suggesting that striatal spine pruning might be a form of homeostatic plasticity that compensates for the loss of striatal DA innervation and the resulting dysregulation of corticostriatal glutamatergic transmission. On the other hand, the remodeling of axospinous corticostriatal and thalamostriatal glutamatergic synapses might represent a form of late maladaptive plasticity that underlies changes in the strength and plastic properties of these afferents and the resulting increased firing and bursting activity of striatal SPNs in the parkinsonian state. There is also evidence that these abnormal synaptic connections might contribute to the pathophysiology of L-DOPA-induced dyskinesia. Despite the significant advances made in this field over the last thirty years, many controversial issues remain about the striatal SPN subtypes affected, the role of spine changes in the altered activity of SPNs in the parkinsonisn state, and the importance of striatal spine plasticity in the pathophysiology of L-DOPA-induced dyskinesia. In this review, we will examine the current state of knowledge of these issues, discuss the limitations of the animal models used to address some of these questions, and assess the relevance of data from animal models to the human-diseased condition.
Collapse
Affiliation(s)
- Rosa M Villalba
- Yerkes National Primate Research Center, Emory University, 954 Gatewood Rd. NE, Atlanta, GA, 30329, USA. .,UDALL Center of Excellence for Parkinson's Disease, Emory University, Atlanta, GA, USA.
| | - Yoland Smith
- Yerkes National Primate Research Center, Emory University, 954 Gatewood Rd. NE, Atlanta, GA, 30329, USA.,UDALL Center of Excellence for Parkinson's Disease, Emory University, Atlanta, GA, USA.,Department of Neurology, Emory University, Atlanta, GA, USA
| |
Collapse
|
30
|
Rajan R, Popa T, Quartarone A, Ghilardi MF, Kishore A. Cortical plasticity and levodopa-induced dyskinesias in Parkinson's disease: Connecting the dots in a multicomponent network. Clin Neurophysiol 2017; 128:992-999. [PMID: 28454042 DOI: 10.1016/j.clinph.2017.03.043] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2016] [Revised: 02/11/2017] [Accepted: 03/22/2017] [Indexed: 01/18/2023]
Abstract
Levodopa-induced dyskinesias are motor complications following long term dopaminergic therapy in Parkinson's disease (PD). Impaired brain plasticity resulting in the creation of aberrant motor maps intended to encode normal voluntary movement is proposed to result in the development of dyskinesias. Traditionally, the various nodes in the motor network like the striato-cortical and the cerebello-thalamic loops were thought to function independent of each other with little communication among them. Anatomical evidence from primates revealed the existence of reciprocal loops between the basal ganglia and the cerebellum providing an anatomical basis for communication between the motor network loops. Dyskinetic PD patients reveal impaired brain plasticity within the motor cortex which may be modulated by cortico-cortical, cerebello-cortical or striato-cortical connections. In this article, we review the evidence for altered plasticity in the multicomponent motor network in the context of levodopa induced dyskinesias in PD. Current evidence suggests a pivotal role for the cerebellum in the larger motor network with the ability to integrate sensorimotor information and independently influence multiple nodes in this network. Targeting the cerebellum seems to be a justified approach for future interventions aimed at attenuating levodopa-induced dyskinesias.
Collapse
Affiliation(s)
- Roopa Rajan
- Comprehensive Care Center for Movement Disorders, Sree Chitra Tirunal Institute for Medical Sciences and Technology, Kerala, India.
| | - Traian Popa
- National Institute of Neurological Disorders and Stroke, NIH, Bethesda, USA.
| | - Angelo Quartarone
- Department of Biomedical, Dental Sciences and Morphological and Functional Images, University of Messina, Messina, Italy; IRCCS Centro Neurolesi "Bonino Pulejo", Via Palermo, Messina, Italy.
| | - Maria Felice Ghilardi
- Department of Physiology and Pharmacology, City University of New York Medical School, New York, NY, USA.
| | - Asha Kishore
- Comprehensive Care Center for Movement Disorders, Sree Chitra Tirunal Institute for Medical Sciences and Technology, Kerala, India.
| |
Collapse
|
31
|
Gagnon D, Petryszyn S, Sanchez MG, Bories C, Beaulieu JM, De Koninck Y, Parent A, Parent M. Striatal Neurons Expressing D 1 and D 2 Receptors are Morphologically Distinct and Differently Affected by Dopamine Denervation in Mice. Sci Rep 2017; 7:41432. [PMID: 28128287 PMCID: PMC5269744 DOI: 10.1038/srep41432] [Citation(s) in RCA: 146] [Impact Index Per Article: 18.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2016] [Accepted: 12/19/2016] [Indexed: 12/11/2022] Open
Abstract
The loss of nigrostriatal dopamine neurons in Parkinson’s disease induces a reduction in the number of dendritic spines on medium spiny neurons (MSNs) of the striatum expressing D1 or D2 dopamine receptor. Consequences on MSNs expressing both receptors (D1/D2 MSNs) are currently unknown. We looked for changes induced by dopamine denervation in the density, regional distribution and morphological features of D1/D2 MSNs, by comparing 6-OHDA-lesioned double BAC transgenic mice (Drd1a-tdTomato/Drd2-EGFP) to sham-lesioned animals. D1/D2 MSNs are uniformly distributed throughout the dorsal striatum (1.9% of MSNs). In contrast, they are heterogeneously distributed and more numerous in the ventral striatum (14.6% in the shell and 7.3% in the core). Compared to D1 and D2 MSNs, D1/D2 MSNs are endowed with a smaller cell body and a less profusely arborized dendritic tree with less dendritic spines. The dendritic spine density of D1/D2 MSNs, but also of D1 and D2 MSNs, is significantly reduced in 6-OHDA-lesioned mice. In contrast to D1 and D2 MSNs, the extent of dendritic arborization of D1/D2 MSNs appears unaltered in 6-OHDA-lesioned mice. Our data indicate that D1/D2 MSNs in the mouse striatum form a distinct neuronal population that is affected differently by dopamine deafferentation that characterizes Parkinson’s disease.
Collapse
Affiliation(s)
- D Gagnon
- Centre de recherche de l'Institut universitaire en santé mentale de Québec, Department of Psychiatry and Neuroscience, Faculty of medicine, Université Laval, Quebec City, QC, Canada
| | - S Petryszyn
- Centre de recherche de l'Institut universitaire en santé mentale de Québec, Department of Psychiatry and Neuroscience, Faculty of medicine, Université Laval, Quebec City, QC, Canada
| | - M G Sanchez
- Centre de recherche de l'Institut universitaire en santé mentale de Québec, Department of Psychiatry and Neuroscience, Faculty of medicine, Université Laval, Quebec City, QC, Canada
| | - C Bories
- Centre de recherche de l'Institut universitaire en santé mentale de Québec, Department of Psychiatry and Neuroscience, Faculty of medicine, Université Laval, Quebec City, QC, Canada
| | - J M Beaulieu
- Centre de recherche de l'Institut universitaire en santé mentale de Québec, Department of Psychiatry and Neuroscience, Faculty of medicine, Université Laval, Quebec City, QC, Canada
| | - Y De Koninck
- Centre de recherche de l'Institut universitaire en santé mentale de Québec, Department of Psychiatry and Neuroscience, Faculty of medicine, Université Laval, Quebec City, QC, Canada
| | - A Parent
- Centre de recherche de l'Institut universitaire en santé mentale de Québec, Department of Psychiatry and Neuroscience, Faculty of medicine, Université Laval, Quebec City, QC, Canada
| | - M Parent
- Centre de recherche de l'Institut universitaire en santé mentale de Québec, Department of Psychiatry and Neuroscience, Faculty of medicine, Université Laval, Quebec City, QC, Canada
| |
Collapse
|
32
|
Wang Q, Zhang W. Maladaptive Synaptic Plasticity in L-DOPA-Induced Dyskinesia. Front Neural Circuits 2016; 10:105. [PMID: 28066191 PMCID: PMC5168436 DOI: 10.3389/fncir.2016.00105] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2016] [Accepted: 12/05/2016] [Indexed: 01/28/2023] Open
Abstract
The emergence of L-DOPA-induced dyskinesia (LID) in patients with Parkinson disease (PD) could be due to maladaptive plasticity of corticostriatal synapses in response to L-DOPA treatment. A series of recent studies has revealed that LID is associated with marked morphological plasticity of striatal dendritic spines, particularly cell type-specific structural plasticity of medium spiny neurons (MSNs) in the striatum. In addition, evidence demonstrating the occurrence of plastic adaptations, including aberrant morphological and functional features, in multiple components of cortico-basal ganglionic circuitry, such as primary motor cortex (M1) and basal ganglia (BG) output nuclei. These adaptations have been implicated in the pathophysiology of LID. Here, we briefly review recent studies that have addressed maladaptive plastic changes within the cortico-BG loop in dyskinetic animal models of PD and patients with PD.
Collapse
Affiliation(s)
- Qiang Wang
- The National Key Clinic Specialty, Guangdong Provincial Key Laboratory on Brain Function Repair and Regeneration, The Engineering Technology Research Center of Education Ministry of China, Department of Neurosurgery, Zhujiang Hospital, Southern Medical University Guangzhou, China
| | - Wangming Zhang
- The National Key Clinic Specialty, Guangdong Provincial Key Laboratory on Brain Function Repair and Regeneration, The Engineering Technology Research Center of Education Ministry of China, Department of Neurosurgery, Zhujiang Hospital, Southern Medical University Guangzhou, China
| |
Collapse
|
33
|
Corvol JC, Poewe W. Pharmacogenetics of Parkinson's Disease in Clinical Practice. Mov Disord Clin Pract 2016; 4:173-180. [PMID: 30363349 DOI: 10.1002/mdc3.12444] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2016] [Revised: 08/12/2016] [Accepted: 08/29/2016] [Indexed: 12/13/2022] Open
Abstract
Background Pharmacogenetics aims to identify the genetic factors participating in the heterogeneity of drug response. The ultimate goal is to provide personalized treatment by identifying responders and non-responders, individuals at risk of developing drug adverse effects, and by adjusting dosage. Several studies have been performed in Parkinson's disease (PD), to investigate drug response variability according to genetic factors for dopamine replacement therapies. Methods We performed a systematic literature search of articles related to pharmacogenetic studies in PD, and found 47 studies. Findings Motor response and adverse reactions to dopaminergic drugs were associated with genes encoding enzymes of their metabolism as well as their receptors or targets. Despite some interesting results, considerable work remains to be done to replicate and validate their clinical relevance before translation into clinical practice. Conclusions There are currently no guidelines published for pharmacogenetic factors related to PD drugs. More research is need in this field in order to improve our knowledge in drug response variability in PD. Algorithms taking into account clinical, pharmacological, and genetic factors are probably the most promising way to help for a personalized medicine in PD.
Collapse
Affiliation(s)
- Jean-Christophe Corvol
- Sorbonne Universités UPMC Univ Paris 06 UMR_S1127 ICM Paris France.,INSERM UMR_S1127 and CIC-1422 ICM Paris France.,CNRS UMR_7225 ICM Paris France.,Département des maladies du système nerveux AP-HP Hôpital Pitié-Salpêtrière Paris France
| | - Werner Poewe
- Department of Neurology Medical University Innsbruck Innsbruck Austria
| |
Collapse
|
34
|
Bez F, Francardo V, Cenci MA. Dramatic differences in susceptibility to l-DOPA-induced dyskinesia between mice that are aged before or after a nigrostriatal dopamine lesion. Neurobiol Dis 2016; 94:213-25. [DOI: 10.1016/j.nbd.2016.06.005] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2016] [Revised: 06/08/2016] [Accepted: 06/11/2016] [Indexed: 12/26/2022] Open
|
35
|
Del-Bel E, Bortolanza M, Dos-Santos-Pereira M, Bariotto K, Raisman-Vozari R. l-DOPA-induced dyskinesia in Parkinson's disease: Are neuroinflammation and astrocytes key elements? Synapse 2016; 70:479-500. [DOI: 10.1002/syn.21941] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2016] [Revised: 09/06/2016] [Accepted: 09/06/2016] [Indexed: 12/22/2022]
Affiliation(s)
- Elaine Del-Bel
- Department of MFPB-Physiology; FORP, Campus USP, University of São Paulo; Av. Café, s/no Ribeirão Preto SP 14040-904 Brazil
- Center for Interdisciplinary Research on Applied Neurosciences (NAPNA); São Paulo Brazil
- Department of Physiology; FMRP; São Paulo Brazil
- Department of Neurology and Behavioral Neuroscience; FMRP, Campus USP, University of São Paulo; Av. Bandeirantes 13400 Ribeirão Preto SP 14049-900 Brazil
| | - Mariza Bortolanza
- Department of MFPB-Physiology; FORP, Campus USP, University of São Paulo; Av. Café, s/no Ribeirão Preto SP 14040-904 Brazil
- Center for Interdisciplinary Research on Applied Neurosciences (NAPNA); São Paulo Brazil
| | - Maurício Dos-Santos-Pereira
- Department of MFPB-Physiology; FORP, Campus USP, University of São Paulo; Av. Café, s/no Ribeirão Preto SP 14040-904 Brazil
- Center for Interdisciplinary Research on Applied Neurosciences (NAPNA); São Paulo Brazil
- Department of Physiology; FMRP; São Paulo Brazil
| | - Keila Bariotto
- Department of MFPB-Physiology; FORP, Campus USP, University of São Paulo; Av. Café, s/no Ribeirão Preto SP 14040-904 Brazil
- Center for Interdisciplinary Research on Applied Neurosciences (NAPNA); São Paulo Brazil
- Department of Neurology and Behavioral Neuroscience; FMRP, Campus USP, University of São Paulo; Av. Bandeirantes 13400 Ribeirão Preto SP 14049-900 Brazil
| | - Rita Raisman-Vozari
- INSERM UMR 1127, CNRS UMR 7225, UPMC; Thérapeutique Expérimentale de la Neurodégénérescence, Hôpital de la Salpetrière-ICM (Institut du cerveau et de la moelle épinière); Paris France
| |
Collapse
|
36
|
Kamagata K, Hatano T, Aoki S. What is NODDI and what is its role in Parkinson's assessment? Expert Rev Neurother 2016; 16:241-3. [PMID: 26777076 DOI: 10.1586/14737175.2016.1142876] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Affiliation(s)
- Koji Kamagata
- b Department of Radiology , Juntendo University Graduate School of Medicine , Bunkyo-ku , Tokyo , Japan
| | - Taku Hatano
- a Department of Neurology , Juntendo University Graduate School of Medicine , Bunkyo-ku , Tokyo , Japan
| | - Shigeki Aoki
- b Department of Radiology , Juntendo University Graduate School of Medicine , Bunkyo-ku , Tokyo , Japan
| |
Collapse
|
37
|
Zhang D, McGregor M, Bordia T, Perez XA, McIntosh JM, Decker MW, Quik M. α7 nicotinic receptor agonists reduce levodopa-induced dyskinesias with severe nigrostriatal damage. Mov Disord 2015; 30:1901-1911. [PMID: 26573698 DOI: 10.1002/mds.26453] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2015] [Revised: 09/09/2015] [Accepted: 09/16/2015] [Indexed: 12/18/2022] Open
Abstract
BACKGROUND ABT-126 is a novel, safe, and well-tolerated α7 nicotinic receptor agonist in a Phase 2 Alzheimer's disease study. We tested the antidyskinetic effect of ABT-126 in 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine-treated squirrel monkeys with moderate and more severe nigrostriatal damage. METHODS Monkeys (n = 21, set 1) were lesioned with 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine 1-2×. When parkinsonian, they were gavaged with levodopa (10 mg/kg)/carbidopa (2.5 mg/kg) twice daily and dyskinesias rated. They were then given nicotine in drinking water (n = 5), or treated with vehicle (n = 6) or ABT-126 (n = 10) twice daily orally 30 min before levodopa. Set 1 was then re-lesioned 1 to 2 times for a total of 3 to 4 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine injections. The antidyskinetic effect of ABT-126, nicotine, and the β2* nicotinic receptor agonist ABT-894 was re-assessed. Another group of monkeys (n = 23, set 2) were lesioned with 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine only 1× to 2×. They were treated with levodopa/carbidopa, administered the α7 agonist ABT-107 (n = 6), ABT-894 (n = 6), nicotine (n = 5), or vehicle (n = 6) and dyskinesias evaluated. All monkeys were euthanized and the dopamine transporter measured. RESULTS With moderate nigrostriatal damage (1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine 1×-2×), ABT-126 dose-dependently decreased dyskinesias (∼60%), with similar results seen with ABT-894 (∼60%) or nicotine (∼60%). With more severe damage (1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine 3-4×), ABT-126 and nicotine reduced dyskinesias, but ABT-894 did not. The dopamine transporter was 41% and 8.9% of control, with moderate and severe nigrostriatal damage, respectively. No drug modified parkinsonism. CONCLUSION The novel α7 nicotinic receptor drug ABT-126 reduced dyskinesias in monkeys with both moderate and severe nigrostriatal damage. ABT-126 may be useful to reduce dyskinesias in both early- and later-stage Parkinson's disease.
Collapse
Affiliation(s)
- Danhui Zhang
- Center for Health Sciences, SRI International, 333 Ravenswood Ave, Menlo Park, CA, 94025
| | - Matthew McGregor
- Center for Health Sciences, SRI International, 333 Ravenswood Ave, Menlo Park, CA, 94025
| | - Tanuja Bordia
- Center for Health Sciences, SRI International, 333 Ravenswood Ave, Menlo Park, CA, 94025
| | - Xiomara A Perez
- Center for Health Sciences, SRI International, 333 Ravenswood Ave, Menlo Park, CA, 94025
| | - J Michael McIntosh
- George E. Wahlen Veterans Affairs Medical Center and Departments of Psychiatry and Biology, University of Utah, Salt Lake City, UT 84148
| | - Michael W Decker
- AbbVie, Inc, 1 North Waukegan Road, North Chicago, IL 60064-6125
| | - Maryka Quik
- Center for Health Sciences, SRI International, 333 Ravenswood Ave, Menlo Park, CA, 94025
| |
Collapse
|
38
|
Quik M, Zhang D, McGregor M, Bordia T. Alpha7 nicotinic receptors as therapeutic targets for Parkinson's disease. Biochem Pharmacol 2015; 97:399-407. [PMID: 26093062 PMCID: PMC4600450 DOI: 10.1016/j.bcp.2015.06.014] [Citation(s) in RCA: 86] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2015] [Accepted: 06/11/2015] [Indexed: 11/25/2022]
Abstract
Accumulating evidence suggests that CNS α7 nicotinic acetylcholine receptors (nAChRs) are important targets for the development of therapeutic approaches for Parkinson's disease. This progressive neurodegenerative disorder is characterized by debilitating motor deficits, as well as autonomic problems, cognitive declines, changes in affect and sleep disturbances. Currently l-dopa is the gold standard treatment for Parkinson's disease motor problems, particularly in the early disease stages. However, it does not improve the other symptoms, nor does it reduce the inevitable disease progression. Novel therapeutic strategies for Parkinson's disease are therefore critical. Extensive pre-clinical work using a wide variety of experimental models shows that nicotine and nAChR agonists protect against damage to nigrostriatal and other neuronal cells. This observation suggests that nicotine and/or nAChR agonists may be useful as disease modifying agents. Additionally, studies in several parkinsonian animal models including nonhuman primates show that nicotine reduces l-dopa-induced dyskinesias, a side effect of l-dopa therapy that may be as incapacitating as Parkinson's disease itself. Work with subtype selective nAChR agonists indicate that α7 nAChRs are involved in mediating both the neuroprotective and antidyskinetic effects, thus offering a targeted strategy with optimal beneficial effects and minimal adverse responses. Here, we review studies demonstrating a role for α7 nAChRs in protection against neurodegenerative effects and for the reduction of l-dopa-induced dyskinesias. Altogether, this work suggests that α7 nAChRs may be useful targets for reducing Parkinson's disease progression and for the management of the dyskinesias that arise with l-dopa therapy.
Collapse
Affiliation(s)
- Maryka Quik
- Center for Health Sciences, SRI International, 333 Ravenswood Ave, CA 94025, USA.
| | - Danhui Zhang
- Center for Health Sciences, SRI International, 333 Ravenswood Ave, CA 94025, USA
| | - Matthew McGregor
- Center for Health Sciences, SRI International, 333 Ravenswood Ave, CA 94025, USA
| | - Tanuja Bordia
- Center for Health Sciences, SRI International, 333 Ravenswood Ave, CA 94025, USA
| |
Collapse
|
39
|
Villalba RM, Mathai A, Smith Y. Morphological changes of glutamatergic synapses in animal models of Parkinson's disease. Front Neuroanat 2015; 9:117. [PMID: 26441550 PMCID: PMC4585113 DOI: 10.3389/fnana.2015.00117] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2015] [Accepted: 08/17/2015] [Indexed: 02/05/2023] Open
Abstract
The striatum and the subthalamic nucleus (STN) are the main entry doors for extrinsic inputs to reach the basal ganglia (BG) circuitry. The cerebral cortex, thalamus and brainstem are the key sources of glutamatergic inputs to these nuclei. There is anatomical, functional and neurochemical evidence that glutamatergic neurotransmission is altered in the striatum and STN of animal models of Parkinson’s disease (PD) and that these changes may contribute to aberrant network neuronal activity in the BG-thalamocortical circuitry. Postmortem studies of animal models and PD patients have revealed significant pathology of glutamatergic synapses, dendritic spines and microcircuits in the striatum of parkinsonians. More recent findings have also demonstrated a significant breakdown of the glutamatergic corticosubthalamic system in parkinsonian monkeys. In this review, we will discuss evidence for synaptic glutamatergic dysfunction and pathology of cortical and thalamic inputs to the striatum and STN in models of PD. The potential functional implication of these alterations on synaptic integration, processing and transmission of extrinsic information through the BG circuits will be considered. Finally, the significance of these pathological changes in the pathophysiology of motor and non-motor symptoms in PD will be examined.
Collapse
Affiliation(s)
- Rosa M Villalba
- Yerkes National Primate Research Center, Emory University Atlanta, GA, USA ; UDALL Center of Excellence for Parkinson's Disease, Emory University Atlanta, GA, USA
| | - Abraham Mathai
- Yerkes National Primate Research Center, Emory University Atlanta, GA, USA ; UDALL Center of Excellence for Parkinson's Disease, Emory University Atlanta, GA, USA
| | - Yoland Smith
- Yerkes National Primate Research Center, Emory University Atlanta, GA, USA ; UDALL Center of Excellence for Parkinson's Disease, Emory University Atlanta, GA, USA ; Department of Neurology, Emory University Atlanta, GA, USA
| |
Collapse
|
40
|
Dashtipour K, Chen JJ, Kani C, Bahjri K, Ghamsary M. Clinical Outcomes in Patients with Parkinson's Disease Treated with a Monoamine Oxidase Type-B inhibitor: A Cross-Sectional, Cohort Study. Pharmacotherapy 2015; 35:681-6. [PMID: 26139574 PMCID: PMC5034746 DOI: 10.1002/phar.1611] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
STUDY OBJECTIVE To evaluate the long-term risk of developing cognitive symptoms (e.g., dementia, hallucinations), dyskinesia, falls, and freezing of gait (FoG) in patients with Parkinson's disease (PD) who received monoamine oxidase type B inhibitors (MAOB-Is) compared with patients who had never received MAOB-Is. DESIGN Retrospective, cross-sectional, cohort study. SETTING Academic movement disorders clinic. PATIENTS One hundred eighty-one patients with idiopathic PD who were receiving MAOB-I therapy on a long-term basis for a minimum of 1 year (MAOB-I current-user cohort) and 121 patients with idiopathic PD who had never received MAOB-I therapy (MAOB-I never-user cohort [control group]) between January 1, 1996, and November 30, 2011. MEASUREMENTS AND MAIN RESULTS The five study outcome variables were dementia, dyskinesia, falls, FoG, and hallucinations. Baseline and outcome data were collected from medical records. Patients in the MAOB-I current-user group were included only if absence of the specified outcomes was documented at baseline. Adjusted multiple logistic regression analyses were performed to calculate the odds ratios (ORs) for MAOB-I use versus never use on clinical outcomes. MAOB-I treatment was associated with a 44.7% reduced risk of dyskinesia (adjusted OR 0.553, 95% confidence interval 0.314-0.976, p=0.041), with the greatest risk reduction observed after 2 years of treatment. No significant association was noted with MAOB-I use and development of dementia, falls, FoG, or hallucinations. CONCLUSION Long-term use of MAOB-I therapy was associated with reduced risk of dyskinesia in patients with PD.
Collapse
Affiliation(s)
| | - Jack J Chen
- School of Medicine, Loma Linda University, Loma Linda, California.,College of Pharmacy, Marshall B. Ketchum University, Fullerton, California
| | - Camellia Kani
- School of Medicine, Loma Linda University, Loma Linda, California
| | - Khaled Bahjri
- School of Public Health, Loma Linda University, Loma Linda, California
| | - Mark Ghamsary
- School of Public Health, Loma Linda University, Loma Linda, California
| |
Collapse
|
41
|
Cenci MA. Presynaptic Mechanisms of l-DOPA-Induced Dyskinesia: The Findings, the Debate, and the Therapeutic Implications. Front Neurol 2014; 5:242. [PMID: 25566170 PMCID: PMC4266027 DOI: 10.3389/fneur.2014.00242] [Citation(s) in RCA: 159] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2014] [Accepted: 11/10/2014] [Indexed: 12/24/2022] Open
Abstract
The dopamine (DA) precursor l-DOPA has been the most effective treatment for Parkinson’s disease (PD) for over 40 years. However, the response to this treatment changes with disease progression, and most patients develop dyskinesias (abnormal involuntary movements) and motor fluctuations within a few years of l-DOPA therapy. There is wide consensus that these motor complications depend on both pre- and post-synaptic disturbances of nigrostriatal DA transmission. Several presynaptic mechanisms converge to generate large DA swings in the brain concomitant with the peaks-and-troughs of plasma l-DOPA levels, while post-synaptic changes engender abnormal functional responses in dopaminoceptive neurons. While this general picture is well-accepted, the relative contribution of different factors remains a matter of debate. A particularly animated debate has been growing around putative players on the presynaptic side of the cascade. To what extent do presynaptic disturbances in DA transmission depend on deficiency/dysfunction of the DA transporter, aberrant release of DA from serotonin neurons, or gliovascular mechanisms? And does noradrenaline (which is synthetized from DA) play a role? This review article will summarize key findings, controversies, and pending questions regarding the presynaptic mechanisms of l-DOPA-induced dyskinesia. Intriguingly, the debate around these mechanisms has spurred research into previously unexplored facets of brain plasticity that have far-reaching implications to the treatment of neuropsychiatric disease.
Collapse
Affiliation(s)
- M Angela Cenci
- Basal Ganglia Pathophysiology Unit, Department of Experimental Medical Science, Lund University , Lund , Sweden
| |
Collapse
|