1
|
Wang P, Bi Y, Li M, Chen J, Wang Z, Wen H, Zhou M, Luo M, Zhang W. Cortico-striatal gamma oscillations are modulated by dopamine D3 receptors in dyskinetic rats. Neural Regen Res 2025; 20:1164-1177. [PMID: 38989954 PMCID: PMC11438323 DOI: 10.4103/nrr.nrr-d-23-01240] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2023] [Accepted: 01/16/2024] [Indexed: 07/12/2024] Open
Abstract
JOURNAL/nrgr/04.03/01300535-202504000-00031/figure1/v/2024-07-06T104127Z/r/image-tiff Long-term levodopa administration can lead to the development of levodopa-induced dyskinesia. Gamma oscillations are a widely recognized hallmark of abnormal neural electrical activity in levodopa-induced dyskinesia. Currently, studies have reported increased oscillation power in cases of levodopa-induced dyskinesia. However, little is known about how the other electrophysiological parameters of gamma oscillations are altered in levodopa-induced dyskinesia. Furthermore, the role of the dopamine D3 receptor, which is implicated in levodopa-induced dyskinesia, in movement disorder-related changes in neural oscillations is unclear. We found that the cortico-striatal functional connectivity of beta oscillations was enhanced in a model of Parkinson's disease. Furthermore, levodopa application enhanced cortical gamma oscillations in cortico-striatal projections and cortical gamma aperiodic components, as well as bidirectional primary motor cortex (M1) ↔ dorsolateral striatum gamma flow. Administration of PD128907 (a selective dopamine D3 receptor agonist) induced dyskinesia and excessive gamma oscillations with a bidirectional M1 ↔ dorsolateral striatum flow. However, administration of PG01037 (a selective dopamine D3 receptor antagonist) attenuated dyskinesia, suppressed gamma oscillations and cortical gamma aperiodic components, and decreased gamma causality in the M1 → dorsolateral striatum direction. These findings suggest that the dopamine D3 receptor plays a role in dyskinesia-related oscillatory activity, and that it has potential as a therapeutic target for levodopa-induced dyskinesia.
Collapse
Affiliation(s)
- Pengfei Wang
- Neurosurgery Center, Department of Pediatric Neurosurgery, Guangdong Provincial Key Laboratory on Brain Function Repair and Regeneration, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong Province, China
| | | | | | | | | | | | | | | | | |
Collapse
|
2
|
Wang X, Zhang W. IRL790 modulated striatal D1 neurons synaptic plasticity ameliorating levodopa-induced dyskinesia in mouse. Front Aging Neurosci 2024; 16:1401991. [PMID: 38872625 PMCID: PMC11169859 DOI: 10.3389/fnagi.2024.1401991] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2024] [Accepted: 05/17/2024] [Indexed: 06/15/2024] Open
Abstract
Objective Levodopa (L-dopa) therapy is the principal pharmacological treatment for Parkinson's disease (PD). Nevertheless, prolonged use of this drug may result in different involuntary movement symptoms caused by the medication, referred to as levodopa-induced dyskinesia (LID). LID is associated with changes in synaptic plasticity of the D1 medium spiny neurons (MSNs) located in the dorsal striatum (dStr). Within the striatum, the amount of Dopamine D3 receptor (D3R) is notably increased in LID, demonstrating colocalization with D1R expression in neurons, and the level of D3R expression is directly related to the intensity of LID. IRL 790, as a D3R antagonist, can ameliorate LID. This study aims to explore if IRL 790 improves LID by regulating the synaptic plasticity of D1+ MSNs in dStr. Methods The electrophysiology and synaptic spine density of D1+ MSNs in dStr were recorded for sham mice, LID mice, and LID mice treated with IRL 790. The regulation of synaptic plasticity in LID D1+ MSNs by IRL 790 was analyzed. Behavioral tests were conducted to confirm the treatment effect of IRL 790 on LID. Results In LID D1+ MSNs, there was persistent abnormal LTP, absence of LTD, and an increase in spontaneous excitatory postsynaptic currents (sEPSCs). IRL 790 treatment restored normal LTP, LTD, and sEPSCs. Treatment with IRL 790 also restored the reduced dendritic spine density in D1+ MSNs of LID mice. IRL790 improved dyskinetic manifestations in LID mice. Conclusion IRL790 ameliorates LID by regulating the synaptic structure and functional plasticity of striatal D1+ MSNs.
Collapse
Affiliation(s)
- Xiaofei Wang
- Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Wangming Zhang
- Guangdong Provincial Key Laboratory on Brain, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| |
Collapse
|
3
|
Chagraoui A, Di Giovanni G, De Deurwaerdère P. Neurobiological and Pharmacological Perspectives of D3 Receptors in Parkinson’s Disease. Biomolecules 2022; 12:biom12020243. [PMID: 35204744 PMCID: PMC8961531 DOI: 10.3390/biom12020243] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2021] [Revised: 01/19/2022] [Accepted: 01/26/2022] [Indexed: 12/02/2022] Open
Abstract
The discovery of the D3 receptor (D3R) subtypes of dopamine (DA) has generated an understandable increase in interest in the field of neurological diseases, especially Parkinson’s disease (PD). Indeed, although DA replacement therapy with l-DOPA has provided an effective treatment for patients with PD, it is responsible for invalidating abnormal involuntary movements, known as L-DOPA-induced dyskinesia, which constitutes a serious limitation of the use of this therapy. Of particular interest is the finding that chronic l-DOPA treatment can trigger the expression of D1R–D3R heteromeric interactions in the dorsal striatum. The D3R is expressed in various tissues of the central nervous system, including the striatum. Compelling research has focused on striatal D3Rs in the context of PD and motor side effects, including dyskinesia, occurring with DA replacement therapy. Therefore, this review will briefly describe the basal ganglia (BG) and the DA transmission within these brain regions, before going into more detail with regard to the role of D3Rs in PD and their participation in the current treatments. Numerous studies have also highlighted specific interactions between D1Rs and D3Rs that could promote dyskinesia. Finally, this review will also address the possibility that D3Rs located outside of the BG may mediate some of the effects of DA replacement therapy.
Collapse
Affiliation(s)
- Abdeslam Chagraoui
- Différenciation et Communication Neuroendocrine, Endocrine et Germinale Laboratory, Institute for Research and Innovation in Biomedicine of Normandy (IRIB), University of Rouen, INSERM 1239, 76000 Rouen, France
- Department of Medical Biochemistry, Rouen University Hospital, 76000 Rouen, France
- Correspondence: ; Tel.: +33-2-35-14-83-69
| | - Giuseppe Di Giovanni
- Laboratory of Neurophysiology, Department of Physiology and Biochemistry, Faculty of Medicine and Surgery, University of Malta, 2080 Msida, Malta;
- Neuroscience Division, School of Biosciences, Cardiff University, Cardiff CF10 3AT, UK
| | - Philippe De Deurwaerdère
- Unité Mixte de Recherche (UMR) 5287, Centre National de la Recherche Scientifique (CNRS), CEDEX, 33000 Bordeaux, France;
| |
Collapse
|
4
|
Bono F, Mutti V, Tomasoni Z, Sbrini G, Missale C, Fiorentini C. Recent Advances in Dopamine D3 Receptor Heterodimers: Focus on Dopamine D3 and D1 Receptor-Receptor Interaction and Striatal Function. Curr Top Behav Neurosci 2022; 60:47-72. [PMID: 35505059 DOI: 10.1007/7854_2022_353] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
G protein-coupled receptors (GPCR) heterodimers represent new entities with unique pharmacological, signalling, and trafficking properties, with specific distribution restricted to those cells where the two interacting receptors are co-expressed. Like other GPCR, dopamine D3 receptors (D3R) directly interact with various receptors to form heterodimers: data showing the D3R physical interaction with both GPCR and non-GPCR receptors have been provided including D3R interaction with other dopamine receptors. The aim of this chapter is to summarize current knowledge of the distinct roles of heterodimers involving D3R, focusing on the D3R interaction with the dopamine D1 receptor (D1R): the D1R-D3R heteromer, in fact, has been postulated in both ventral and motor striatum. Interestingly, since both D1R and D3R have been implicated in several pathological conditions, including schizophrenia, motor dysfunctions, and substance use disorders, the D1R-D3R heteromer may represent a potential drug target for the treatment of these diseases.
Collapse
Affiliation(s)
- Federica Bono
- Division of Pharmacology, Department of Molecular and Translational Medicine, University of Brescia, Brescia, Italy
| | - Veronica Mutti
- Division of Pharmacology, Department of Molecular and Translational Medicine, University of Brescia, Brescia, Italy
| | - Zaira Tomasoni
- Division of Pharmacology, Department of Molecular and Translational Medicine, University of Brescia, Brescia, Italy
| | - Giulia Sbrini
- Division of Pharmacology, Department of Molecular and Translational Medicine, University of Brescia, Brescia, Italy
| | - Cristina Missale
- Division of Pharmacology, Department of Molecular and Translational Medicine, University of Brescia, Brescia, Italy
| | - Chiara Fiorentini
- Division of Pharmacology, Department of Molecular and Translational Medicine, University of Brescia, Brescia, Italy.
| |
Collapse
|
5
|
Lee B, Taylor M, Griffin SA, McInnis T, Sumien N, Mach RH, Luedtke RR. Evaluation of Substituted N-Phenylpiperazine Analogs as D3 vs. D2 Dopamine Receptor Subtype Selective Ligands. Molecules 2021; 26:molecules26113182. [PMID: 34073405 PMCID: PMC8198181 DOI: 10.3390/molecules26113182] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2021] [Revised: 05/19/2021] [Accepted: 05/21/2021] [Indexed: 11/20/2022] Open
Abstract
N-phenylpiperazine analogs can bind selectively to the D3 versus the D2 dopamine receptor subtype despite the fact that these two D2-like dopamine receptor subtypes exhibit substantial amino acid sequence homology. The binding for a number of these receptor subtype selective compounds was found to be consistent with their ability to bind at the D3 dopamine receptor subtype in a bitopic manner. In this study, a series of the 3-thiophenephenyl and 4-thiazolylphenyl fluoride substituted N-phenylpiperazine analogs were evaluated. Compound 6a was found to bind at the human D3 receptor with nanomolar affinity with substantial D3 vs. D2 binding selectivity (approximately 500-fold). Compound 6a was also tested for activity in two in-vivo assays: (1) a hallucinogenic-dependent head twitch response inhibition assay using DBA/2J mice and (2) an L-dopa-dependent abnormal involuntary movement (AIM) inhibition assay using unilateral 6-hydroxydopamine lesioned (hemiparkinsonian) rats. Compound 6a was found to be active in both assays. This compound could lead to a better understanding of how a bitopic D3 dopamine receptor selective ligand might lead to the development of pharmacotherapeutics for the treatment of levodopa-induced dyskinesia (LID) in patients with Parkinson’s disease.
Collapse
Affiliation(s)
- Boeun Lee
- Department of Radiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA; (B.L.); (R.H.M.)
| | - Michelle Taylor
- Department of Pharmacology and Neuroscience, University of North Texas Health Science Center-Fort Worth, Fort Worth, TX 76107, USA; (M.T.); (S.A.G.); (T.M.); (N.S.)
| | - Suzy A. Griffin
- Department of Pharmacology and Neuroscience, University of North Texas Health Science Center-Fort Worth, Fort Worth, TX 76107, USA; (M.T.); (S.A.G.); (T.M.); (N.S.)
| | - Tamara McInnis
- Department of Pharmacology and Neuroscience, University of North Texas Health Science Center-Fort Worth, Fort Worth, TX 76107, USA; (M.T.); (S.A.G.); (T.M.); (N.S.)
| | - Nathalie Sumien
- Department of Pharmacology and Neuroscience, University of North Texas Health Science Center-Fort Worth, Fort Worth, TX 76107, USA; (M.T.); (S.A.G.); (T.M.); (N.S.)
| | - Robert H. Mach
- Department of Radiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA; (B.L.); (R.H.M.)
| | - Robert R. Luedtke
- Department of Pharmacology and Neuroscience, University of North Texas Health Science Center-Fort Worth, Fort Worth, TX 76107, USA; (M.T.); (S.A.G.); (T.M.); (N.S.)
- Correspondence:
| |
Collapse
|
6
|
Correlación entre la expresión del receptor dopaminérgico D2 y presencia de movimientos involuntarios anormales (MIA) en un modelo de disquinesia en ratas Wistar hemiparkinsonizadas. Neurologia 2021. [DOI: 10.1016/j.nrl.2017.12.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
|
7
|
Caro Aponte P, Otálora C, Guzmán J, Turner L, Alcázar J, Mayorga E. Correlation between dopamine receptor D2 expression and presence of abnormal involuntary movements in Wistar rats with hemiparkinsonism and dyskinesia. NEUROLOGÍA (ENGLISH EDITION) 2021. [DOI: 10.1016/j.nrleng.2017.12.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/25/2022] Open
|
8
|
Lanza K, Bishop C. Dopamine D3 Receptor Plasticity in Parkinson's Disease and L-DOPA-Induced Dyskinesia. Biomedicines 2021; 9:biomedicines9030314. [PMID: 33808538 PMCID: PMC8003204 DOI: 10.3390/biomedicines9030314] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2021] [Revised: 03/16/2021] [Accepted: 03/17/2021] [Indexed: 11/21/2022] Open
Abstract
Parkinson’s Disease (PD) is characterized by primary and secondary plasticity that occurs in response to progressive degeneration and long-term L-DOPA treatment. Some of this plasticity contributes to the detrimental side effects associated with chronic L-DOPA treatment, namely L-DOPA-induced dyskinesia (LID). The dopamine D3 receptor (D3R) has emerged as a promising target in LID management as it is upregulated in LID. This upregulation occurs primarily in the D1-receptor-bearing (D1R) cells of the striatum, which have been repeatedly implicated in LID manifestation. D3R undergoes dynamic changes both in PD and in LID, making it difficult to delineate D3R’s specific contributions, but recent genetic and pharmacologic tools have helped to clarify its role in LID. The following review will discuss these changes, recent advances to better clarify D3R in both PD and LID and potential steps for translating these findings.
Collapse
Affiliation(s)
- Kathryn Lanza
- Department of Physiology, Northwestern University, Chicago, IL 60201, USA;
| | - Christopher Bishop
- Department of Psychology, Binghamton University, Binghamton, NY 13902, USA
- Correspondence:
| |
Collapse
|
9
|
Severity of Dyskinesia and D3R Signaling Changes Induced by L-DOPA Treatment of Hemiparkinsonian Rats Are Features Inherent to the Treated Subjects. Biomolecules 2019; 9:biom9090431. [PMID: 31480516 PMCID: PMC6770442 DOI: 10.3390/biom9090431] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2019] [Revised: 08/20/2019] [Accepted: 08/29/2019] [Indexed: 12/26/2022] Open
Abstract
Extensive damage to nigrostriatal dopaminergic neurons leads to Parkinson’s disease (PD). To date, the most effective treatment has been administration of levodopa (L-DOPA) to increase dopaminergic tone. This treatment leads to responses that vary widely among patients, from predominantly beneficial effects to the induction of disabling, abnormal movements (L-DOPA induced dyskinesia (LID)). Similarly, experimental studies have shown animals with widely different degrees of LID severity. In this study, unilateral injections of 6-hydroxydopamine (6-OHDA) in the medial forebrain bundle (MFB) produced more than 90% depletion of dopamine in both the striatum and the substantia nigra reticulata (SNr) of rats. Population analysis showed that dopamine depletion levels were clustered in a single population. In contrast, analysis of abnormal involuntary movements (AIMs) induced by L-DOPA treatment of 6-OHDA-lesioned animals yielded two populations: one with mild LID, and the other with severe LID, which are also related to different therapeutic responses. We examined whether the severity of LID correlated with changes in dopamine 3 receptor (D3R) signaling because of the following: (a) D3R expression and the induction of LID are strongly correlated; and (b) dopaminergic denervation induces a qualitative change in D3R signaling in the SNr. We found that the effects of D3R activation on cAMP accumulation and depolarization-induced [3H]-gamma-aminobutyric acid ([3H]-GABA) release were switched. L-DOPA treatment normalized the denervation-induced changes in animals with mild LID. The D3R activation caused depression of both dopamine 1 receptor (D1R)-induced increases in cAMP production and depolarization-induced [3H]-GABA release, which were reversed to their pre-denervation state. In animals with severe LID, none of the denervation-induced changes were reversed. The finding that in the absence of identifiable differences in 6-OHDA and L-DOPA treatment, two populations of animals with different D3R signaling and LIDs severity implies that mechanisms intrinsic to the treated subject determine the segregation.
Collapse
|
10
|
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]
|
11
|
Signal transduction in L-DOPA-induced dyskinesia: from receptor sensitization to abnormal gene expression. J Neural Transm (Vienna) 2018; 125:1171-1186. [PMID: 29396608 PMCID: PMC6060907 DOI: 10.1007/s00702-018-1847-7] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2017] [Accepted: 01/23/2018] [Indexed: 01/06/2023]
Abstract
A large number of signaling abnormalities have been implicated in the emergence and expression of l-DOPA-induced dyskinesia (LID). The primary cause for many of these changes is the development of sensitization at dopamine receptors located on striatal projection neurons (SPN). This initial priming, which is particularly evident at the level of dopamine D1 receptors (D1R), can be viewed as a homeostatic response to dopamine depletion and is further exacerbated by chronic administration of l-DOPA, through a variety of mechanisms affecting various components of the G-protein-coupled receptor machinery. Sensitization of dopamine receptors in combination with pulsatile administration of l-DOPA leads to intermittent and coordinated hyperactivation of signal transduction cascades, ultimately resulting in long-term modifications of gene expression and protein synthesis. A detailed mapping of these pathological changes and of their involvement in LID has been produced during the last decade. According to this emerging picture, activation of sensitized D1R results in the stimulation of cAMP-dependent protein kinase and of the dopamine- and cAMP-regulated phosphoprotein of 32 kDa. This, in turn, activates the extracellular signal-regulated kinases 1 and 2 (ERK), leading to chromatin remodeling and aberrant gene transcription. Dysregulated ERK results also in the stimulation of the mammalian target of rapamycin complex 1, which promotes protein synthesis. Enhanced levels of multiple effector targets, including several transcription factors have been implicated in LID and associated changes in synaptic plasticity and morphology. This article provides an overview of the intracellular modifications occurring in SPN and associated with LID.
Collapse
|
12
|
Solís O, Garcia-Montes JR, González-Granillo A, Xu M, Moratalla R. Dopamine D3 Receptor Modulates l-DOPA-Induced Dyskinesia by Targeting D1 Receptor-Mediated Striatal Signaling. Cereb Cortex 2018; 27:435-446. [PMID: 26483399 DOI: 10.1093/cercor/bhv231] [Citation(s) in RCA: 50] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023] Open
Abstract
The dopamine D3 receptor (D3R) belongs to the dopamine D2-like receptor family and is principally located in the ventral striatum. However, previous studies reported D3R overexpression in the dorsal striatum following l-DOPA treatment in parkinsonian animals. This fact has drawn attention in the importance of D3R in l-DOPA-induced dyskinesia (LID). Here, we used D3R knockout mice to assess the role of D3R in LID and rotational sensitization in the hemiparkinsonian model. Mice lacking D3R presented a reduction in dyskinesia without interfering with the antiparkinsonian l-DOPA effect and were accompanied by a reduction in the l-DOPA-induced rotations. Interestingly, deleting D3R attenuated important molecular markers in the D1R-neurons such as FosB, extracellular signal-regulated kinase, and histone-3 (H3)-activation. Colocalization studies in D1R-tomato and D2R-green fluorescent protein BAC-transgenic mice indicated that l-DOPA-induced D3R overexpression principally occurs in D1R-containing neurons although it is also present in the D2R-neurons. Moreover, D3R pharmacological blockade with PG01037 reduced dyskinesia and the molecular markers expressed in D1R-neurons. In addition, this antagonist further reduced dyskinetic symptoms in D1R heterozygous mice, indicating a direct interaction between D1R and D3R. Together, our results demonstrate that D3R modulates the development of dyskinesia by targeting D1R-mediated intracellular signaling and suggest that decreasing D3R activity may help to ameliorate LID.
Collapse
Affiliation(s)
- Oscar Solís
- Instituto Cajal, Consejo Superior de Investigaciones Científicas, CSIC, Madrid 28002, Spain
- CIBERNED, Instituto de Salud Carlos III, Madrid, Spain
| | - Jose Ruben Garcia-Montes
- Instituto Cajal, Consejo Superior de Investigaciones Científicas, CSIC, Madrid 28002, Spain
- CIBERNED, Instituto de Salud Carlos III, Madrid, Spain
| | - Aldo González-Granillo
- Instituto Cajal, Consejo Superior de Investigaciones Científicas, CSIC, Madrid 28002, Spain
| | - Ming Xu
- Department of Anesthesia and Critical Care, The University of Chicago, Chicago, IL, USA
| | - Rosario Moratalla
- Instituto Cajal, Consejo Superior de Investigaciones Científicas, CSIC, Madrid 28002, Spain
- CIBERNED, Instituto de Salud Carlos III, Madrid, Spain
| |
Collapse
|
13
|
Kline CLB, Ralff MD, Lulla AR, Wagner JM, Abbosh PH, Dicker DT, Allen JE, El-Deiry WS. Role of Dopamine Receptors in the Anticancer Activity of ONC201. Neoplasia 2018; 20:80-91. [PMID: 29216597 PMCID: PMC5725157 DOI: 10.1016/j.neo.2017.10.002] [Citation(s) in RCA: 89] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2017] [Revised: 10/17/2017] [Accepted: 10/23/2017] [Indexed: 11/23/2022]
Abstract
ONC201/TIC10 is a first-in-class small molecule inducer of TRAIL that causes early activation of the integrated stress response. Its promising safety profile and broad-spectrum efficacy in vitro have been confirmed in Phase I/II trials in several advanced malignancies. Binding and reporter assays have shown that ONC201 is a selective antagonist of the dopamine D2-like receptors, specifically, DRD2 and DRD3. We hypothesized that ONC201's interaction with DRD2 plays a role in ONC201's anticancer effects. Using cBioportal and quantitative reverse-transcription polymerase chain reaction analyses, we confirmed that DRD2 is expressed in different cancer cell types in a cell type-specific manner. On the other hand, DRD3 was generally not detectable. Overexpressing DRD2 in cells with low DRD2 levels increased ONC201-induced PARP cleavage, which was preceded and correlated with an increase in ONC201-induced CHOP mRNA expression. On the other hand, knocking out DRD2 using CRISPR/Cas9 in three cancer cell lines was not sufficient to abrogate ONC201's anticancer effects. Although ONC201's anticancer activity was not dependent on DRD2 expression in the cancer cell types tested, we assessed the cytotoxic potential of DRD2 blockade. Transient DRD2 knockdown in HCT116 cells activated the integrated stress response and reduced cell number. Pharmacological antagonism of DRD2 significantly reduced cell viability. Thus, we demonstrate in this study that disrupting dopamine receptor expression and activity can have cytotoxic effects that may at least be in part due to the activation of the integrated stress response. On the other hand, ONC201's anticancer activity goes beyond its ability to antagonize DRD2, potentially due to ONC201's ability to activate other pathways that are independent of DRD2. Nevertheless, blocking the dopamine D1-like receptor DRD5 via siRNA or the use of a pharmacological antagonist promoted ONC201-induced anticancer activity.
Collapse
MESH Headings
- Antineoplastic Agents/pharmacology
- Apoptosis/drug effects
- Cell Line, Tumor
- Cell Survival/drug effects
- Drug Resistance, Neoplasm
- Gene Expression
- Gene Expression Regulation, Neoplastic/drug effects
- Gene Knockout Techniques
- Heterocyclic Compounds, 4 or More Rings/pharmacology
- Humans
- Imidazoles
- Neoplasms/genetics
- Neoplasms/metabolism
- Pyridines
- Pyrimidines
- RNA, Small Interfering/genetics
- Receptors, Dopamine/genetics
- Receptors, Dopamine/metabolism
- Receptors, Dopamine D2/genetics
- Receptors, Dopamine D2/metabolism
- Receptors, Dopamine D3/genetics
- Receptors, Dopamine D3/metabolism
- Receptors, Dopamine D5/genetics
- Receptors, Dopamine D5/metabolism
Collapse
Affiliation(s)
- Christina Leah B Kline
- Laboratory of Translational Oncology and Experimental Cancer Therapeutics, Molecular Therapeutics Program, Department of Hematology/Oncology, Fox Chase Cancer Center, Philadelphia, PA
| | - Marie D Ralff
- Laboratory of Translational Oncology and Experimental Cancer Therapeutics, Molecular Therapeutics Program, Department of Hematology/Oncology, Fox Chase Cancer Center, Philadelphia, PA
| | - Amriti R Lulla
- Laboratory of Translational Oncology and Experimental Cancer Therapeutics, Molecular Therapeutics Program, Department of Hematology/Oncology, Fox Chase Cancer Center, Philadelphia, PA
| | - Jessica M Wagner
- Laboratory of Translational Oncology and Experimental Cancer Therapeutics, Molecular Therapeutics Program, Department of Hematology/Oncology, Fox Chase Cancer Center, Philadelphia, PA
| | - Phillip H Abbosh
- Laboratory of Translational Oncology and Experimental Cancer Therapeutics, Molecular Therapeutics Program, Department of Hematology/Oncology, Fox Chase Cancer Center, Philadelphia, PA
| | - David T Dicker
- Laboratory of Translational Oncology and Experimental Cancer Therapeutics, Molecular Therapeutics Program, Department of Hematology/Oncology, Fox Chase Cancer Center, Philadelphia, PA
| | | | - Wafik S El-Deiry
- Laboratory of Translational Oncology and Experimental Cancer Therapeutics, Molecular Therapeutics Program, Department of Hematology/Oncology, Fox Chase Cancer Center, Philadelphia, PA.
| |
Collapse
|
14
|
Nishi A, Shuto T. Potential for targeting dopamine/DARPP-32 signaling in neuropsychiatric and neurodegenerative disorders. Expert Opin Ther Targets 2017; 21:259-272. [PMID: 28052701 DOI: 10.1080/14728222.2017.1279149] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
INTRODUCTION Alterations in dopamine neurotransmission has been implicated in pathophysiology of neuropsychiatric and neurodegenerative disorders, and DARPP-32 plays a pivotal role in dopamine neurotransmission. DARPP-32 likely influences dopamine-mediated behaviors in animal models of neuropsychiatric and neurodegenerative disorders and therapeutic effects of pharmacological treatment. Areas covered: We will review animal studies on the biochemical and behavioral roles of DARPP-32 in drug addiction, schizophrenia and Parkinson's disease. In general, under physiological and pathophysiological conditions, DARPP-32 in D1 receptor expressing (D1R) -medium spiny neurons (MSNs) promotes dopamine/D1 receptor/PKA signaling, whereas DARPP-32 in D2 receptor expressing (D2R)-MSNs counteracts dopamine/D2 receptor signaling. However, the function of DARPP-32 is differentially regulated in acute and chronic phases of drug addiction; DARPP-32 enhances D1 receptor/PKA signaling in the acute phase, whereas DARPP-32 suppresses D1 receptor/PKA signaling in the chronic phase through homeostatic mechanisms. Therefore, DARPP-32 plays a bidirectional role in dopamine neurotransmission, depending on the cell type and experimental conditions, and is involved in dopamine-related behavioral abnormalities. Expert opinion: DARPP-32 differentially regulates dopamine signaling in D1R- and D2R-MSNs, and a shift of balance between D1R- and D2R-MSN function is associated with behavioral abnormalities. An adjustment of this imbalance is achieved by therapeutic approaches targeting DARPP-32-related signaling molecules.
Collapse
Affiliation(s)
- Akinori Nishi
- a Department of Pharmacology , Kurume University School of Medicine , Kurume, Fukuoka , Japan
| | - Takahide Shuto
- a Department of Pharmacology , Kurume University School of Medicine , Kurume, Fukuoka , Japan
| |
Collapse
|
15
|
Sebastianutto I, Maslava N, Hopkins CR, Cenci MA. Validation of an improved scale for rating l-DOPA-induced dyskinesia in the mouse and effects of specific dopamine receptor antagonists. Neurobiol Dis 2016; 96:156-170. [PMID: 27597526 DOI: 10.1016/j.nbd.2016.09.001] [Citation(s) in RCA: 54] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2016] [Revised: 08/24/2016] [Accepted: 09/01/2016] [Indexed: 11/17/2022] Open
Abstract
Rodent models of l-DOPA-induced dyskinesia (LID) are essential to investigate pathophysiological mechanisms and treatment options. Ratings of abnormal involuntary movements (AIMs) are used to capture both qualitative and quantitative features of dyskinetic behaviors. Thus far, validated rating scales for the mouse have anchored the definition of severity to the time during which AIMs are present. Here we have asked whether the severity of axial, limb, and orolingual AIMs can be objectively assessed with scores based on movement amplitude. Mice sustained 6-OHDA lesions in the medial forebrain bundle and were treated with l-DOPA (3-6mg/kg/day) until they developed stable AIMs scores. Two independent investigators rated AIM severity using both the validated time-based scale and a novel amplitude scale, evaluating the degree of deviation of dyskinetic body parts relative to their resting position. The amplitude scale yielded a high degree of consistency both within- and between raters. Thus, time-based scores, amplitude scores, and a combination of the two ('global AIM scores') were applied to compare antidyskinetic effects produced by amantadine and by the following subtype-specific DA receptor antagonists: SCH23390 (D1/D5), Raclopride (D2/D3), PG01037 (D3), L-745,870 (D4), and VU6004461 (D4). SCH23390 and Raclopride produced similarly robust reductions in both time-based scores and amplitude scores, while PG01037 and L-745,870 had more partial effects. Interestingly, a novel and highly brain penetrable D4 receptor antagonist (VU6004461) markedly attenuated both time-based and amplitude scores without diminishing the general motor stimulant effect of l-DOPA. In summary, our results show that a dyskinesia scale combining a time dimension with an amplitude dimension ('global AIMs') is more sensitive than unidimensional scales. Moreover, the antidyskinetic effects produced by two chemically distinct D4 antagonists identify the D4 receptor as a potential future target for the treatment of LID.
Collapse
Affiliation(s)
- Irene Sebastianutto
- Basal Ganglia Pathophysiology Unit, Dept. Exp. Medical Science, Lund University, BMC, 221 84 Lund, Sweden.
| | - Natallia Maslava
- Basal Ganglia Pathophysiology Unit, Dept. Exp. Medical Science, Lund University, BMC, 221 84 Lund, Sweden
| | - Corey R Hopkins
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Nebraska Medical Center, Omaha, NE 68198-6125, USA
| | - M Angela Cenci
- Basal Ganglia Pathophysiology Unit, Dept. Exp. Medical Science, Lund University, BMC, 221 84 Lund, Sweden.
| |
Collapse
|
16
|
Payer DE, Guttman M, Kish SJ, Tong J, Adams JR, Rusjan P, Houle S, Furukawa Y, Wilson AA, Boileau I. D3 dopamine receptor-preferring [11C]PHNO PET imaging in Parkinson patients with dyskinesia. Neurology 2015; 86:224-30. [PMID: 26718579 DOI: 10.1212/wnl.0000000000002285] [Citation(s) in RCA: 43] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2015] [Accepted: 08/10/2015] [Indexed: 01/25/2023] Open
Abstract
OBJECTIVE To investigate whether levodopa-induced dyskinesias (LID) are associated with D3 overexpression in levodopa-treated humans with Parkinson disease (PD). METHODS In this case-control study, we used PET with the D3-preferring radioligand [(11)C]-(+)-PHNO to estimate D2/3 receptor binding in patients with levodopa-treated PD with LID (n = 12) and without LID (n = 12), and healthy control subjects matched for age, sex, education, and mental status (n = 18). RESULTS Compared to nondyskinetic patients, those with LID showed heightened [(11)C]-(+)-PHNO binding in the D3-rich globus pallidus. Both PD groups also showed higher binding than controls in the sensorimotor division of the striatum. In contrast, D2/3 binding in the ventral striatum was lower in patients with LID than without, possibly reflecting higher dopamine levels. CONCLUSIONS Dopaminergic abnormalities contributing to LID may include elevated D2/3 binding in globus pallidus, perhaps reflecting D3 receptor upregulation. The findings support therapeutic strategies that target and diminish activity at D3 to prevent LID.
Collapse
Affiliation(s)
- Doris E Payer
- From the Addictions Program (D.E.P., I.B.), the Research Imaging Centre (D.E.P., S.J.K., J.T., P.R., S.H., A.A.W., I.B.), and the Human Brain Laboratory (M.G., S.J.K., Y.F.), Centre for Addiction and Mental Health, Toronto; Campbell Family Mental Health Research Institute (S.J.K., J.T., P.R., S.H., A.A.W., I.B.), Toronto; the Departments of Psychiatry (D.E.P., S.J.K., J.T., A.A.W., I.B.) and Pharmacology (S.J.K.), University of Toronto; and the Centre for Movement Disorders (M.G., J.R.A.), Markham, Canada
| | - Mark Guttman
- From the Addictions Program (D.E.P., I.B.), the Research Imaging Centre (D.E.P., S.J.K., J.T., P.R., S.H., A.A.W., I.B.), and the Human Brain Laboratory (M.G., S.J.K., Y.F.), Centre for Addiction and Mental Health, Toronto; Campbell Family Mental Health Research Institute (S.J.K., J.T., P.R., S.H., A.A.W., I.B.), Toronto; the Departments of Psychiatry (D.E.P., S.J.K., J.T., A.A.W., I.B.) and Pharmacology (S.J.K.), University of Toronto; and the Centre for Movement Disorders (M.G., J.R.A.), Markham, Canada
| | - Stephen J Kish
- From the Addictions Program (D.E.P., I.B.), the Research Imaging Centre (D.E.P., S.J.K., J.T., P.R., S.H., A.A.W., I.B.), and the Human Brain Laboratory (M.G., S.J.K., Y.F.), Centre for Addiction and Mental Health, Toronto; Campbell Family Mental Health Research Institute (S.J.K., J.T., P.R., S.H., A.A.W., I.B.), Toronto; the Departments of Psychiatry (D.E.P., S.J.K., J.T., A.A.W., I.B.) and Pharmacology (S.J.K.), University of Toronto; and the Centre for Movement Disorders (M.G., J.R.A.), Markham, Canada
| | - Junchao Tong
- From the Addictions Program (D.E.P., I.B.), the Research Imaging Centre (D.E.P., S.J.K., J.T., P.R., S.H., A.A.W., I.B.), and the Human Brain Laboratory (M.G., S.J.K., Y.F.), Centre for Addiction and Mental Health, Toronto; Campbell Family Mental Health Research Institute (S.J.K., J.T., P.R., S.H., A.A.W., I.B.), Toronto; the Departments of Psychiatry (D.E.P., S.J.K., J.T., A.A.W., I.B.) and Pharmacology (S.J.K.), University of Toronto; and the Centre for Movement Disorders (M.G., J.R.A.), Markham, Canada
| | - John R Adams
- From the Addictions Program (D.E.P., I.B.), the Research Imaging Centre (D.E.P., S.J.K., J.T., P.R., S.H., A.A.W., I.B.), and the Human Brain Laboratory (M.G., S.J.K., Y.F.), Centre for Addiction and Mental Health, Toronto; Campbell Family Mental Health Research Institute (S.J.K., J.T., P.R., S.H., A.A.W., I.B.), Toronto; the Departments of Psychiatry (D.E.P., S.J.K., J.T., A.A.W., I.B.) and Pharmacology (S.J.K.), University of Toronto; and the Centre for Movement Disorders (M.G., J.R.A.), Markham, Canada
| | - Pablo Rusjan
- From the Addictions Program (D.E.P., I.B.), the Research Imaging Centre (D.E.P., S.J.K., J.T., P.R., S.H., A.A.W., I.B.), and the Human Brain Laboratory (M.G., S.J.K., Y.F.), Centre for Addiction and Mental Health, Toronto; Campbell Family Mental Health Research Institute (S.J.K., J.T., P.R., S.H., A.A.W., I.B.), Toronto; the Departments of Psychiatry (D.E.P., S.J.K., J.T., A.A.W., I.B.) and Pharmacology (S.J.K.), University of Toronto; and the Centre for Movement Disorders (M.G., J.R.A.), Markham, Canada
| | - Sylvain Houle
- From the Addictions Program (D.E.P., I.B.), the Research Imaging Centre (D.E.P., S.J.K., J.T., P.R., S.H., A.A.W., I.B.), and the Human Brain Laboratory (M.G., S.J.K., Y.F.), Centre for Addiction and Mental Health, Toronto; Campbell Family Mental Health Research Institute (S.J.K., J.T., P.R., S.H., A.A.W., I.B.), Toronto; the Departments of Psychiatry (D.E.P., S.J.K., J.T., A.A.W., I.B.) and Pharmacology (S.J.K.), University of Toronto; and the Centre for Movement Disorders (M.G., J.R.A.), Markham, Canada
| | - Yoshiaki Furukawa
- From the Addictions Program (D.E.P., I.B.), the Research Imaging Centre (D.E.P., S.J.K., J.T., P.R., S.H., A.A.W., I.B.), and the Human Brain Laboratory (M.G., S.J.K., Y.F.), Centre for Addiction and Mental Health, Toronto; Campbell Family Mental Health Research Institute (S.J.K., J.T., P.R., S.H., A.A.W., I.B.), Toronto; the Departments of Psychiatry (D.E.P., S.J.K., J.T., A.A.W., I.B.) and Pharmacology (S.J.K.), University of Toronto; and the Centre for Movement Disorders (M.G., J.R.A.), Markham, Canada
| | - Alan A Wilson
- From the Addictions Program (D.E.P., I.B.), the Research Imaging Centre (D.E.P., S.J.K., J.T., P.R., S.H., A.A.W., I.B.), and the Human Brain Laboratory (M.G., S.J.K., Y.F.), Centre for Addiction and Mental Health, Toronto; Campbell Family Mental Health Research Institute (S.J.K., J.T., P.R., S.H., A.A.W., I.B.), Toronto; the Departments of Psychiatry (D.E.P., S.J.K., J.T., A.A.W., I.B.) and Pharmacology (S.J.K.), University of Toronto; and the Centre for Movement Disorders (M.G., J.R.A.), Markham, Canada
| | - Isabelle Boileau
- From the Addictions Program (D.E.P., I.B.), the Research Imaging Centre (D.E.P., S.J.K., J.T., P.R., S.H., A.A.W., I.B.), and the Human Brain Laboratory (M.G., S.J.K., Y.F.), Centre for Addiction and Mental Health, Toronto; Campbell Family Mental Health Research Institute (S.J.K., J.T., P.R., S.H., A.A.W., I.B.), Toronto; the Departments of Psychiatry (D.E.P., S.J.K., J.T., A.A.W., I.B.) and Pharmacology (S.J.K.), University of Toronto; and the Centre for Movement Disorders (M.G., J.R.A.), Markham, Canada.
| |
Collapse
|
17
|
Furman CA, Roof RA, Moritz AE, Miller BN, Doyle TB, Free RB, Banala AK, Paul NM, Kumar V, Sibley CD, Newman AH, Sibley DR. Investigation of the binding and functional properties of extended length D3 dopamine receptor-selective antagonists. Eur Neuropsychopharmacol 2015; 25:1448-61. [PMID: 25583363 PMCID: PMC4449328 DOI: 10.1016/j.euroneuro.2014.11.013] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/27/2014] [Revised: 11/05/2014] [Accepted: 11/20/2014] [Indexed: 01/11/2023]
Abstract
The D3 dopamine receptor represents an important target in drug addiction in that reducing receptor activity may attenuate the self-administration of drugs and/or disrupt drug or cue-induced relapse. Medicinal chemistry efforts have led to the development of D3 preferring antagonists and partial agonists that are >100-fold selective vs. the closely related D2 receptor, as best exemplified by extended-length 4-phenylpiperazine derivatives. Based on the D3 receptor crystal structure, these molecules are known to dock to two sites on the receptor where the 4-phenylpiperazine moiety binds to the orthosteric site and an extended aryl amide moiety docks to a secondary binding pocket. The bivalent nature of the receptor binding of these compounds is believed to contribute to their D3 selectivity. In this study, we examined if such compounds might also be "bitopic" such that their aryl amide moieties act as allosteric modulators to further enhance the affinities of the full-length molecules for the receptor. First, we deconstructed several extended-length D3-selective ligands into fragments, termed "synthons", representing either orthosteric or secondary aryl amide pharmacophores and investigated their effects on D3 receptor binding and function. The orthosteric synthons were found to inhibit radioligand binding and to antagonize dopamine activation of the D3 receptor, albeit with lower affinities than the full-length compounds. Notably, the aryl amide-based synthons had no effect on the affinities or potencies of the orthosteric synthons, nor did they have any effect on receptor activation by dopamine. Additionally, pharmacological investigation of the full-length D3-selective antagonists revealed that these compounds interacted with the D3 receptor in a purely competitive manner. Our data further support that the 4-phenylpiperazine D3-selective antagonists are bivalent and that their enhanced affinity for the D3 receptor is due to binding at both the orthosteric site as well as a secondary binding pocket. Importantly, however, their interactions at the secondary site do not allosterically modulate their binding to the orthosteric site.
Collapse
Affiliation(s)
- Cheryse A Furman
- Molecular Neuropharmacology Section, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD, USA
| | - Rebecca A Roof
- Molecular Neuropharmacology Section, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD, USA
| | - Amy E Moritz
- Molecular Neuropharmacology Section, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD, USA
| | - Brittney N Miller
- Molecular Neuropharmacology Section, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD, USA
| | - Trevor B Doyle
- Molecular Neuropharmacology Section, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD, USA
| | - R Benjamin Free
- Molecular Neuropharmacology Section, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD, USA
| | - Ashwini K Banala
- Medicinal Chemistry Section, Molecular Targets and Medications Discovery Branch, National Institute on Drug Abuse, National Institutes of Health, Baltimore, MD, USA
| | - Noel M Paul
- Medicinal Chemistry Section, Molecular Targets and Medications Discovery Branch, National Institute on Drug Abuse, National Institutes of Health, Baltimore, MD, USA
| | - Vivek Kumar
- Medicinal Chemistry Section, Molecular Targets and Medications Discovery Branch, National Institute on Drug Abuse, National Institutes of Health, Baltimore, MD, USA
| | - Christopher D Sibley
- Medicinal Chemistry Section, Molecular Targets and Medications Discovery Branch, National Institute on Drug Abuse, National Institutes of Health, Baltimore, MD, USA
| | - Amy Hauck Newman
- Medicinal Chemistry Section, Molecular Targets and Medications Discovery Branch, National Institute on Drug Abuse, National Institutes of Health, Baltimore, MD, USA.
| | - David R Sibley
- Molecular Neuropharmacology Section, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD, USA.
| |
Collapse
|
18
|
Bastide MF, Meissner WG, Picconi B, Fasano S, Fernagut PO, Feyder M, Francardo V, Alcacer C, Ding Y, Brambilla R, Fisone G, Jon Stoessl A, Bourdenx M, Engeln M, Navailles S, De Deurwaerdère P, Ko WKD, Simola N, Morelli M, Groc L, Rodriguez MC, Gurevich EV, Quik M, Morari M, Mellone M, Gardoni F, Tronci E, Guehl D, Tison F, Crossman AR, Kang UJ, Steece-Collier K, Fox S, Carta M, Angela Cenci M, Bézard E. Pathophysiology of L-dopa-induced motor and non-motor complications in Parkinson's disease. Prog Neurobiol 2015. [PMID: 26209473 DOI: 10.1016/j.pneurobio.2015.07.002] [Citation(s) in RCA: 350] [Impact Index Per Article: 35.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Involuntary movements, or dyskinesia, represent a debilitating complication of levodopa (L-dopa) therapy for Parkinson's disease (PD). L-dopa-induced dyskinesia (LID) are ultimately experienced by the vast majority of patients. In addition, psychiatric conditions often manifested as compulsive behaviours, are emerging as a serious problem in the management of L-dopa therapy. The present review attempts to provide an overview of our current understanding of dyskinesia and other L-dopa-induced dysfunctions, a field that dramatically evolved in the past twenty years. In view of the extensive literature on LID, there appeared a critical need to re-frame the concepts, to highlight the most suitable models, to review the central nervous system (CNS) circuitry that may be involved, and to propose a pathophysiological framework was timely and necessary. An updated review to clarify our understanding of LID and other L-dopa-related side effects was therefore timely and necessary. This review should help in the development of novel therapeutic strategies aimed at preventing the generation of dyskinetic symptoms.
Collapse
Affiliation(s)
- Matthieu F Bastide
- Univ. de Bordeaux, Institut des Maladies Neurodégénératives, UMR 5293, 33000 Bordeaux, France; CNRS, Institut des Maladies Neurodégénératives, UMR 5293, 33000 Bordeaux, France
| | - Wassilios G Meissner
- Univ. de Bordeaux, Institut des Maladies Neurodégénératives, UMR 5293, 33000 Bordeaux, France; CNRS, Institut des Maladies Neurodégénératives, UMR 5293, 33000 Bordeaux, France; Department of Neurology, University Hospital Bordeaux, France
| | - Barbara Picconi
- Laboratory of Neurophysiology, Fondazione Santa Lucia, IRCCS, Rome, Italy
| | - Stefania Fasano
- Division of Neuroscience, Institute of Experimental Neurology, San Raffaele Scientific Institute, 20132 Milan, Italy
| | - Pierre-Olivier Fernagut
- Univ. de Bordeaux, Institut des Maladies Neurodégénératives, UMR 5293, 33000 Bordeaux, France; CNRS, Institut des Maladies Neurodégénératives, UMR 5293, 33000 Bordeaux, France
| | - Michael Feyder
- Department of Neuroscience, Karolinska Institutet, Stockholm, Sweden
| | - Veronica Francardo
- Basal Ganglia Pathophysiology Unit, Department of Experimental Medical Science, Lund University, Lund, Sweden
| | - Cristina Alcacer
- Basal Ganglia Pathophysiology Unit, Department of Experimental Medical Science, Lund University, Lund, Sweden
| | - Yunmin Ding
- Department of Neurology, Columbia University, New York, USA
| | - Riccardo Brambilla
- Division of Neuroscience, Institute of Experimental Neurology, San Raffaele Scientific Institute, 20132 Milan, Italy
| | - Gilberto Fisone
- Department of Neuroscience, Karolinska Institutet, Stockholm, Sweden
| | - A Jon Stoessl
- Pacific Parkinson's Research Centre and National Parkinson Foundation Centre of Excellence, University of British Columbia, Vancouver, Canada
| | - Mathieu Bourdenx
- Univ. de Bordeaux, Institut des Maladies Neurodégénératives, UMR 5293, 33000 Bordeaux, France; CNRS, Institut des Maladies Neurodégénératives, UMR 5293, 33000 Bordeaux, France
| | - Michel Engeln
- Univ. de Bordeaux, Institut des Maladies Neurodégénératives, UMR 5293, 33000 Bordeaux, France; CNRS, Institut des Maladies Neurodégénératives, UMR 5293, 33000 Bordeaux, France
| | - Sylvia Navailles
- Univ. de Bordeaux, Institut des Maladies Neurodégénératives, UMR 5293, 33000 Bordeaux, France; CNRS, Institut des Maladies Neurodégénératives, UMR 5293, 33000 Bordeaux, France
| | - Philippe De Deurwaerdère
- Univ. de Bordeaux, Institut des Maladies Neurodégénératives, UMR 5293, 33000 Bordeaux, France; CNRS, Institut des Maladies Neurodégénératives, UMR 5293, 33000 Bordeaux, France
| | - Wai Kin D Ko
- Univ. de Bordeaux, Institut des Maladies Neurodégénératives, UMR 5293, 33000 Bordeaux, France; CNRS, Institut des Maladies Neurodégénératives, UMR 5293, 33000 Bordeaux, France
| | - Nicola Simola
- Department of Biomedical Sciences, Section of Neuropsychopharmacology, Cagliari University, 09124 Cagliari, Italy
| | - Micaela Morelli
- Department of Biomedical Sciences, Section of Neuropsychopharmacology, Cagliari University, 09124 Cagliari, Italy
| | - Laurent Groc
- Univ. de Bordeaux, Institut Interdisciplinaire de neurosciences, UMR 5297, 33000 Bordeaux, France; CNRS, Institut Interdisciplinaire de neurosciences, UMR 5297, 33000 Bordeaux, France
| | - Maria-Cruz Rodriguez
- Department of Neurology, Hospital Universitario Donostia and Neuroscience Unit, Bio Donostia Research Institute, San Sebastian, Spain
| | - Eugenia V Gurevich
- Department of Pharmacology, Vanderbilt University Medical Center, Nashville, TN 37232, USA
| | - Maryka Quik
- Center for Health Sciences, SRI International, CA 94025, USA
| | - Michele Morari
- Department of Medical Sciences, Section of Pharmacology, University of Ferrara, Ferrara, Italy
| | - Manuela Mellone
- Dipartimento di Scienze Farmacologiche e Biomolecolari, Università degli Studi di Milano, 20133 Milano, Italy
| | - Fabrizio Gardoni
- Dipartimento di Scienze Farmacologiche e Biomolecolari, Università degli Studi di Milano, 20133 Milano, Italy
| | - Elisabetta Tronci
- Department of Biomedical Sciences, Physiology Section, Cagliari University, Cagliari, Italy
| | - Dominique Guehl
- Univ. de Bordeaux, Institut des Maladies Neurodégénératives, UMR 5293, 33000 Bordeaux, France; CNRS, Institut des Maladies Neurodégénératives, UMR 5293, 33000 Bordeaux, France
| | - François Tison
- Univ. de Bordeaux, Institut des Maladies Neurodégénératives, UMR 5293, 33000 Bordeaux, France; CNRS, Institut des Maladies Neurodégénératives, UMR 5293, 33000 Bordeaux, France; Department of Neurology, University Hospital Bordeaux, France
| | | | - Un Jung Kang
- Basal Ganglia Pathophysiology Unit, Department of Experimental Medical Science, Lund University, Lund, Sweden
| | - Kathy Steece-Collier
- Michigan State University, College of Human Medicine, Department of Translational Science and Molecular Medicine & The Udall Center of Excellence in Parkinson's Disease Research, 333 Bostwick Ave NE, Grand Rapids, MI 49503, USA
| | - Susan Fox
- Morton & Gloria Shulman Movement Disorders Center, Toronto Western Hospital, Toronto, Ontario M4T 2S8, Canada
| | - Manolo Carta
- Department of Biomedical Sciences, Physiology Section, Cagliari University, Cagliari, Italy
| | - M Angela Cenci
- Basal Ganglia Pathophysiology Unit, Department of Experimental Medical Science, Lund University, Lund, Sweden
| | - Erwan Bézard
- Univ. de Bordeaux, Institut des Maladies Neurodégénératives, UMR 5293, 33000 Bordeaux, France; CNRS, Institut des Maladies Neurodégénératives, UMR 5293, 33000 Bordeaux, France; Motac Neuroscience Ltd, Manchester, UK.
| |
Collapse
|
19
|
Niccolini F, Rocchi L, Politis M. Molecular imaging of levodopa-induced dyskinesias. Cell Mol Life Sci 2015; 72:2107-17. [PMID: 25681866 PMCID: PMC11113208 DOI: 10.1007/s00018-015-1854-x] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2014] [Revised: 02/06/2015] [Accepted: 02/09/2015] [Indexed: 12/15/2022]
Abstract
Levodopa-induced dyskinesias (LIDs) occur in the majority of patients with Parkinson's disease (PD) following years of levodopa treatment. The pathophysiology underlying LIDs in PD is poorly understood, and current treatments generate only minor benefits for the patients. Studies with positron emission tomography (PET) molecular imaging have demonstrated that in advanced PD patients, levodopa administration induces sharp increases in striatal dopamine levels, which correlate with LIDs severity. Fluctuations in striatal dopamine levels could be the result of the attenuated buffering ability in the dopaminergically denervated striatum. Lines of evidence from PET studies indicate that serotonergic terminals could also be responsible for the development of LIDs in PD by aberrantly processing exogenous levodopa and by releasing dopamine in a dysregulated manner from the serotonergic terminals. Additionally, other downstream mechanisms involving glutamatergic, cannabinoid, opioid, cholinergic, adenosinergic, and noradrenergic systems may contribute in the development of LIDs. In this article, we review the findings from preclinical, clinical, and molecular imaging studies, which have contributed to our understanding the pathophysiology of LIDs in PD.
Collapse
Affiliation(s)
- Flavia Niccolini
- Neurodegeneration Imaging Group, Department of Basic and Clinical Neuroscience, Institute of Psychiatry, Psychology and Neuroscience (IoPPN), King’s College London, London, SE5 8AF UK
| | - Lorenzo Rocchi
- Neurodegeneration Imaging Group, Department of Basic and Clinical Neuroscience, Institute of Psychiatry, Psychology and Neuroscience (IoPPN), King’s College London, London, SE5 8AF UK
| | - Marios Politis
- Neurodegeneration Imaging Group, Department of Basic and Clinical Neuroscience, Institute of Psychiatry, Psychology and Neuroscience (IoPPN), King’s College London, London, SE5 8AF UK
| |
Collapse
|
20
|
Avalos-Fuentes A, Albarrán-Bravo S, Loya-Lopéz S, Cortés H, Recillas-Morales S, Magaña JJ, Paz-Bermúdez F, Rangel-Barajas C, Aceves J, Erlij D, Florán B. Dopaminergic denervation switches dopamine D3 receptor signaling and disrupts its Ca(2+) dependent modulation by CaMKII and calmodulin in striatonigral projections of the rat. Neurobiol Dis 2014; 74:336-46. [PMID: 25517101 DOI: 10.1016/j.nbd.2014.12.008] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2014] [Revised: 12/03/2014] [Accepted: 12/05/2014] [Indexed: 11/28/2022] Open
Abstract
In striatonigral projections activation of dopamine D3 receptors (D3Rs) potentiates the stimulation of GABA release and cAMP production caused by activation of dopamine D1 receptors (D1Rs). Cytoplasmic [Ca(2+)] in the terminals controls this response by modulating CaMKII, an enzyme that depresses D3R action. To examine the effects of dopamine deprivation on D3R signaling we investigated their function in striatonigral terminals of hemiparkinsonian rats. Denervation switched the signaling cascade initiated by D3R activation. In the non-lesioned side activation of D3R potentiated the stimulatory effects of D1R activation on cAMP production and K(+)-depolarization induced [(3)H] GABA release. In contrast, in the denervated side the stimulatory effects of both D1R activation and forskolin administration were blocked by D3R activation. In non-lesioned slices, D3R responses were inhibited by the activation of CaMKII produced by K(+)-depolarization (via increased Ca(2+) entry). The CaMKII-induced inhibition was blocked by the selective inhibitor KN-62. In denervated tissues the response to D3R stimulation was not modified either by K(+) depolarization or by blocking CaMKII with KN-62. Immunoblotting studies showed that depolarization-induced CaMKII binding to the D3 receptor and CaMKII phosphorylation were suppressed in denervated tissues. We also determined calmodulin expression with PCR and immunoblot techniques. Both techniques showed that calmodulin expression was depressed in the lesioned side. In sum, our studies show that dopaminergic denervation switches the D3R signaling cascade and depresses CaMKII signaling through a process that appears to involve reduced calmodulin levels. Since calmodulin is a major cytoplasmic Ca(2+) buffer our findings suggest that abnormal Ca(2+) buffering may be an important component of the abnormalities observed during dopaminergic denervation.
Collapse
Affiliation(s)
- Arturo Avalos-Fuentes
- Departamento de Fisiología, Biofísica y Neurociencias, Centro de Investigación y de Estudios Avanzados del Instituto Politécnico Nacional, Mexico
| | - Sacnité Albarrán-Bravo
- Departamento de Fisiología, Biofísica y Neurociencias, Centro de Investigación y de Estudios Avanzados del Instituto Politécnico Nacional, Mexico
| | - Santiago Loya-Lopéz
- Departamento de Farmacología, Centro de Investigación y de Estudios Avanzados del Instituto Politécnico Nacional, Mexico
| | - Hernán Cortés
- Laboratory of Genomic Medicine, Department of Genetics, National Rehabilitation Institute, Mexico City, Mexico
| | - Sergio Recillas-Morales
- Facultad de Medicina Veterinaria y Zootecnia, Universidad Autónoma del Estado de México, Mexico
| | - Jonathan J Magaña
- Laboratory of Genomic Medicine, Department of Genetics, National Rehabilitation Institute, Mexico City, Mexico
| | - Francisco Paz-Bermúdez
- Departamento de Fisiología, Biofísica y Neurociencias, Centro de Investigación y de Estudios Avanzados del Instituto Politécnico Nacional, Mexico
| | | | - Jorge Aceves
- Departamento de Fisiología, Biofísica y Neurociencias, Centro de Investigación y de Estudios Avanzados del Instituto Politécnico Nacional, Mexico
| | - David Erlij
- Department of Physiology, SUNY Downstate Medical Center, Brooklyn, NY 11203, USA
| | - Benjamín Florán
- Departamento de Fisiología, Biofísica y Neurociencias, Centro de Investigación y de Estudios Avanzados del Instituto Politécnico Nacional, Mexico.
| |
Collapse
|
21
|
Yan Y, Newman AH, Xu M. Dopamine D1 and D3 receptors mediate reconsolidation of cocaine memories in mouse models of drug self-administration. Neuroscience 2014; 278:154-64. [PMID: 25149631 PMCID: PMC4172503 DOI: 10.1016/j.neuroscience.2014.08.008] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2014] [Revised: 07/15/2014] [Accepted: 08/08/2014] [Indexed: 12/18/2022]
Abstract
Memories of drug experience and drug-associated environmental cues can elicit drug-seeking and taking behaviors in humans. Disruption of reconsolidation of drug memories dampens previous memories and therefore may provide a useful way to treat drug abuse. We and others previously demonstrated that dopamine D1 and D3 receptors play differential roles in acquiring cocaine-induced behaviors. Moreover, D3 receptors contribute to the reconsolidation of cocaine-induced conditioned place preference. In the present study, we examined effects of manipulating D1 or D3 receptors on reconsolidation of cocaine memories in mouse models of drug self-administration. We found that pharmacological blockade of D1 receptors or a genetic mutation of the D3 receptor gene attenuated reconsolidation that lasted for at least 1week after the memory retrieval. In contrast, with no memory retrieval, pharmacological antagonism of D1 receptors or the D3 receptor gene mutation did not significantly affect reconsolidation of cocaine memories. Pharmacological blockade of D3 receptors also attenuated reconsolidation in wild-type mice that lasted for at least 1week after the memory retrieval. These results suggest that D1 and D3 receptors and related signaling mechanisms play key roles in reconsolidation of cocaine memories in mice, and that these receptors may serve as novel targets for the treatment of cocaine abuse in humans.
Collapse
Affiliation(s)
- Y Yan
- Department of Anesthesia and Critical Care, The University of Chicago, Chicago, IL, USA
| | - A H Newman
- Medicinal Chemistry Section, National Institute on Drug Abuse-Intramural Research Program, National Institutes of Health, Baltimore, MD, USA
| | - M Xu
- Department of Anesthesia and Critical Care, The University of Chicago, Chicago, IL, USA.
| |
Collapse
|
22
|
Rangel-Barajas C, Malik M, Taylor M, Neve KA, Mach RH, Luedtke RR. Characterization of [(3) H]LS-3-134, a novel arylamide phenylpiperazine D3 dopamine receptor selective radioligand. J Neurochem 2014; 131:418-31. [PMID: 25041389 DOI: 10.1111/jnc.12825] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2014] [Revised: 07/14/2014] [Accepted: 07/15/2014] [Indexed: 12/23/2022]
Abstract
LS-3-134 is a substituted N-phenylpiperazine derivative that has been reported to exhibit: (i) high-affinity binding (Ki value 0.2 nM) at human D3 dopamine receptors, (ii) > 100-fold D3 versus D2 dopamine receptor subtype binding selectivity, and (iii) low-affinity binding (Ki > 5000 nM) at sigma 1 and sigma 2 receptors. Based upon a forskolin-dependent activation of the adenylyl cyclase inhibition assay, LS-3-134 is a weak partial agonist at both D2 and D3 dopamine receptor subtypes (29% and 35% of full agonist activity, respectively). In this study, [(3) H]-labeled LS-3-134 was prepared and evaluated to further characterize its use as a D3 dopamine receptor selective radioligand. Kinetic and equilibrium radioligand binding studies were performed. This radioligand rapidly reaches equilibrium (10-15 min at 37°C) and binds with high affinity to both human (Kd = 0.06 ± 0.01 nM) and rat (Kd = 0.2 ± 0.02 nM) D3 receptors expressed in HEK293 cells. Direct and competitive radioligand binding studies using rat caudate and nucleus accumbens tissue indicate that [(3) H]LS-3-134 selectively binds a homogeneous population of binding sites with a dopamine D3 receptor pharmacological profile. Based upon these studies, we propose that [(3) H]LS-3-134 represents a novel D3 dopamine receptor selective radioligand that can be used for studying the expression and regulation of the D3 dopamine receptor subtype.
Collapse
Affiliation(s)
- Claudia Rangel-Barajas
- Department of Pharmacology and Neuroscience, University of North Texas Health Science Center, Fort Worth, Texas, USA
| | | | | | | | | | | |
Collapse
|
23
|
Azkona G, Marcilla I, López de Maturana R, Sousa A, Pérez-Navarro E, Luquin MR, Sanchez-Pernaute R. Sustained Increase of PKA Activity in the Postcommissural Putamen of Dyskinetic Monkeys. Mol Neurobiol 2014; 50:1131-41. [DOI: 10.1007/s12035-014-8688-7] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2014] [Accepted: 03/23/2014] [Indexed: 01/14/2023]
|
24
|
Azkona G, Sagarduy A, Aristieta A, Vazquez N, Zubillaga V, Ruíz-Ortega JA, Pérez-Navarro E, Ugedo L, Sánchez-Pernaute R. Buspirone anti-dyskinetic effect is correlated with temporal normalization of dysregulated striatal DRD1 signalling in L-DOPA-treated rats. Neuropharmacology 2013; 79:726-37. [PMID: 24333147 DOI: 10.1016/j.neuropharm.2013.11.024] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2013] [Revised: 11/28/2013] [Accepted: 11/29/2013] [Indexed: 01/11/2023]
Abstract
Dopamine replacement with l-DOPA is the most effective therapy in Parkinson's disease. However, with chronic treatment, half of the patients develop an abnormal motor response including dyskinesias. The specific molecular mechanisms underlying dyskinesias are not fully understood. In this study, we used a well-characterized animal model to first establish the molecular differences between rats that did and did not develop dyskinesias. We then investigated the molecular substrates implicated in the anti-dyskinetic effect of buspirone, a 5HT1A partial agonist. Striatal protein expression profile of dyskinetic animals revealed increased levels of the dopamine receptor (DR)D3, ΔFosB and phospho (p)CREB, as well as an over-activation of the DRD1 signalling pathway, reflected by elevated ratios of phosphorylated DARPP32 and ERK2. Buspirone reduced the abnormal involuntary motor response in dyskinetic rats in a dose-dependent fashion. Buspirone (4 mg/kg) dramatically reduced the presence and severity of dyskinesias (by 83%) and normalized DARPP32 and ERK2 phosphorylation ratios, while the increases in DRD3, ΔFosB and pCREB observed in dyskinetic rats were not modified. Pharmacological experiments combining buspirone with 5HT1A and DRD3 antagonists confirmed that normalization of both pDARPP32 and pERK2 is required, but not sufficient, for blocking dyskinesias. The correlation between pDARPP32 ratio and dyskinesias was significant but not strong, pointing to the involvement of convergent factors and signalling pathways. Our results suggest that in dyskinetic rats DRD3 striatal over-expression could be instrumental in the activation of DRD1-downstream signalling and demonstrate that the anti-dyskinetic effect of buspirone in this model is correlated with DRD1 pathway normalization.
Collapse
Affiliation(s)
- Garikoitz Azkona
- Animal Model Unit, Inbiomed, Mikeletegi, 81, 20009 San Sebastian, Spain; Laboratory of Stem Cells and Neural Repair, Inbiomed, P. Mikeletegi, 81, 20009 San Sebastian, Spain.
| | - Ainhoa Sagarduy
- Department of Pharmacology, Faculty of Medicine and Dentistry, University of the Basque Country (UPV/EHU), B. Sarriena s/n, 48940 Leioa, Spain.
| | - Asier Aristieta
- Department of Pharmacology, Faculty of Medicine and Dentistry, University of the Basque Country (UPV/EHU), B. Sarriena s/n, 48940 Leioa, Spain.
| | - Nerea Vazquez
- Laboratory of Stem Cells and Neural Repair, Inbiomed, P. Mikeletegi, 81, 20009 San Sebastian, Spain.
| | - Verónica Zubillaga
- Laboratory of Stem Cells and Neural Repair, Inbiomed, P. Mikeletegi, 81, 20009 San Sebastian, Spain.
| | - José Angel Ruíz-Ortega
- Department of Pharmacology, Faculty of Medicine and Dentistry, University of the Basque Country (UPV/EHU), B. Sarriena s/n, 48940 Leioa, Spain.
| | - Esther Pérez-Navarro
- Department of Cell Biology, Immunology and Neurosciences, Faculty of Medicine, University of Barcelona, 08036 Barcelona, Spain; Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), C/ Rosselló, 149-153, 08036 Barcelona, Spain; Centro de Investigaciones Biomédicas en Red sobre Enfermedades Neurodegenerativas (CIBERNED), Casanova, 143, Barcelona, Spain.
| | - Luisa Ugedo
- Department of Pharmacology, Faculty of Medicine and Dentistry, University of the Basque Country (UPV/EHU), B. Sarriena s/n, 48940 Leioa, Spain.
| | - Rosario Sánchez-Pernaute
- Laboratory of Stem Cells and Neural Repair, Inbiomed, P. Mikeletegi, 81, 20009 San Sebastian, Spain.
| |
Collapse
|
25
|
Tu Z, Li S, Li A, Taylor M, Ho D, Malik M, Luedtke RR, Mach RH. Synthesis and in vitro pharmacological evaluation of indolyl carboxylic amide analogues as D 3 dopamine receptor selective ligands. MEDCHEMCOMM 2013; 4:1283-1289. [PMID: 24156012 PMCID: PMC3804115 DOI: 10.1039/c3md00098b] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
A series of substituted 1H-indolyl carboxylic acid amides that contain a N-(2-methoxyphenyl)piperazine or N-(2-fluoroethoxy)piperazine group were synthesized and their affinities for human dopamine D2, D3, and D4 receptors were determined. Two of these compounds, 14a and 14b, displayed high binding affinity at D3 (Ki = 0.18 and 0.4 nM, respectively), and selectivity for D3vs. D2 receptors (87-fold and 60-fold, respectively). These two compounds had low binding affinity at D4 receptors and σ receptor sites. The intrinsic activity of these compounds at D2 and D3 receptors was determined using a forskolin-dependent adenylyl cyclase inhibition assay; both 14a and 14b were found to be partial agonists. Furthermore, for compound 14a, the log D value of 2.85 suggested it has suitable lipophilicity for crossing the blood-brain-barrier.
Collapse
Affiliation(s)
- Zhude Tu
- Washington University School of Medicine, St. Louis, MO, 63110, USA., Fax: +1-314-362-8555; Tel: +1-314-362-8538
| | - Shihong Li
- Washington University School of Medicine, St. Louis, MO, 63110, USA., Fax: +1-314-362-8555; Tel: +1-314-362-8538
| | - Aixiao Li
- Washington University School of Medicine, St. Louis, MO, 63110, USA., Fax: +1-314-362-8555; Tel: +1-314-362-8538
| | - Michelle Taylor
- Department of Pharmacology and Neuroscience, University of North Texas Health Science Center, Fort Worth, TX 76107, USA
| | - David Ho
- Department of Pharmacology and Neuroscience, University of North Texas Health Science Center, Fort Worth, TX 76107, USA
| | - Maninder Malik
- Department of Pharmacology and Neuroscience, University of North Texas Health Science Center, Fort Worth, TX 76107, USA
| | - Robert R. Luedtke
- Department of Pharmacology and Neuroscience, University of North Texas Health Science Center, Fort Worth, TX 76107, USA
| | - Robert H. Mach
- Washington University School of Medicine, St. Louis, MO, 63110, USA., Fax: +1-314-362-8555; Tel: +1-314-362-8538
| |
Collapse
|
26
|
Yan Y, Kong H, Wu EJ, Newman AH, Xu M. Dopamine D3 receptors regulate reconsolidation of cocaine memory. Neuroscience 2013; 241:32-40. [PMID: 23506736 DOI: 10.1016/j.neuroscience.2013.03.005] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2013] [Revised: 03/01/2013] [Accepted: 03/07/2013] [Indexed: 12/12/2022]
Abstract
Memories of learned associations between the rewarding properties of drugs of abuse and environmental cues contribute to craving and relapse in humans. Disruption of reconsolidation dampens or even erases previous memories. Dopamine (DA) mediates the acquisition of reward memory and drugs of abuse can pathologically change related neuronal circuits in the mesolimbic DA system. Previous studies showed that DA D3 receptors are involved in cocaine-conditioned place preference (CPP) and reinstatement of cocaine-seeking behavior. However, the role of D3 receptors in reconsolidation of cocaine-induced reward memory remains unclear. In the present study, we combined genetic and pharmacological approaches to investigate the role of D3 receptors in reconsolidation of cocaine-induced CPP. We found that the mutation of the D3 receptor gene weakened reconsolidation of cocaine-induced CPP in mice triggered by a 3-min (min) retrieval. Furthermore, treatment of a selective D3 receptor antagonist PG01037 immediately following the 3-min retrieval disrupted reconsolidation of cocaine-induced CPP in wild-type mice and such disruption remained at least 1 week after the 3-min retrieval. These results suggest that D3 receptors play a key role in reconsolidation of cocaine-induced CPP in mice, and that pharmacological blockade of these receptors may be therapeutic for the treatment of cocaine craving and relapse in clinical settings.
Collapse
Affiliation(s)
- Y Yan
- Department of Anesthesia and Critical Care, The University of Chicago, Chicago, IL, USA
| | | | | | | | | |
Collapse
|
27
|
Caine SB, Thomsen M, Barrett AC, Collins GT, Grundt P, Newman AH, Butler P, Xu M. Cocaine self-administration in dopamine D₃ receptor knockout mice. Exp Clin Psychopharmacol 2012; 20:352-63. [PMID: 22867038 PMCID: PMC3587777 DOI: 10.1037/a0029135] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
The dopamine D₃ receptor has received attention over the last two decades as a target for medications development for substance abuse disorders. Results have remained mixed. Despite emergence of more D₃-selective ligands, possible attribution of observed effects to D₂ receptors remains a concern. Knockout mice may help shed light on mechanisms. Here we evaluated the effect of constitutive D₃ receptor inactivation ("knockout") on the reinforcing effects of cocaine. We tested D₃ wild-type (WT), heterozygous (D₃⁺/⁻), and knockout (D₃⁻/⁻), mice in acquisition and maintenance of intravenous self-administration across a broad range of cocaine doses, using a fixed ratio (FR) 1 and a progressive ratio (PR) schedule of reinforcement, along with parallel food-reinforced studies. Generally, D₃⁻/⁻ mice showed cocaine self-administration comparable to WT controls across assays. Moderate and nonsignificant trends toward lesser reinforcing effects of a low cocaine dose (0.32 mg/kg) were apparent in acquisition and PR studies, consistent with the idea that the D₃ receptor may play a subtle role in the reinforcing effects of low cocaine doses under low FR conditions. However, those effects with cocaine self-administration were more subtle than the lower responding of D₃ knockout mice observed with food-maintained behavior. In addition, the D₃ antagonist PG01037 failed to affect cocaine self-administration under an FR 1 schedule in WT mice. The present data do not support a necessary role for the D₃ receptor in the direct reinforcing effects of cocaine.
Collapse
Affiliation(s)
- S Barak Caine
- Alcohol and Drug Abuse Research Center, McLean Hospital/Harvard Medical School, MA 02478, USA.
| | | | | | | | | | | | | | | |
Collapse
|
28
|
Paquette MA, Martinez AA, Macheda T, Meshul CK, Johnson SW, Berger SP, Giuffrida A. Anti-dyskinetic mechanisms of amantadine and dextromethorphan in the 6-OHDA rat model of Parkinson's disease: role of NMDA vs. 5-HT1A receptors. Eur J Neurosci 2012; 36:3224-34. [PMID: 22861201 DOI: 10.1111/j.1460-9568.2012.08243.x] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023]
Abstract
Amantadine and dextromethorphan suppress levodopa (L-DOPA)-induced dyskinesia (LID) in patients with Parkinson's disease (PD) and abnormal involuntary movements (AIMs) in the unilateral 6-hydroxydopamine (6-OHDA) rat model. These effects have been attributed to N-methyl-d-aspartate (NMDA) antagonism. However, amantadine and dextromethorphan are also thought to block serotonin (5-HT) uptake and cause 5-HT overflow, leading to stimulation of 5-HT(1A) receptors, which has been shown to reduce LID. We undertook a study in 6-OHDA rats to determine whether the anti-dyskinetic effects of these two compounds are mediated by NMDA antagonism and/or 5-HT(1A) agonism. In addition, we assessed the sensorimotor effects of these drugs using the Vibrissae-Stimulated Forelimb Placement and Cylinder tests. Our data show that the AIM-suppressing effect of amantadine was not affected by the 5-HT(1A) antagonist WAY-100635, but was partially reversed by the NMDA agonist d-cycloserine. Conversely, the AIM-suppressing effect of dextromethorphan was prevented by WAY-100635 but not by d-cycloserine. Neither amantadine nor dextromethorphan affected the therapeutic effects of L-DOPA in sensorimotor tests. We conclude that the anti-dyskinetic effect of amantadine is partially dependent on NMDA antagonism, while dextromethorphan suppresses AIMs via indirect 5-HT(1A) agonism. Combined with previous work from our group, our results support the investigation of 5-HT(1A) agonists as pharmacotherapies for LID in PD patients.
Collapse
Affiliation(s)
- Melanie A Paquette
- Department of Pharmacology, University of Texas Health Science Center, San Antonio, TX, USA.
| | | | | | | | | | | | | |
Collapse
|
29
|
Steece-Collier K, Rademacher DJ, Soderstrom K. Anatomy of Graft-induced Dyskinesias: Circuit Remodeling in the Parkinsonian Striatum. ACTA ACUST UNITED AC 2012; 2:15-30. [PMID: 22712056 DOI: 10.1016/j.baga.2012.01.002] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
Abstract
The goal of researchers and clinicians interested in re-instituting cell based therapies for PD is to develop an effective and safe surgical approach to replace dopamine (DA) in individuals suffering from Parkinson's disease (PD). Worldwide clinical trials involving transplantation of embryonic DA neurons into individuals with PD have been discontinued because of the often devastating post-surgical side-effect known as graft-induced dyskinesia (GID). There have been many review articles published in recent years on this subject. There has been a tendency to promote single factors in the cause of GID. In this review, we contrast the pros and cons of multiple factors that have been suggested from clinical and/or preclinical observations, as well as novel factors not yet studied that may be involved with GID. It is our intention to provide a platform that might be instrumental in examining how individual factors that correlate with GID and/or striatal pathology might interact to give rise to dysfunctional circuit remodeling and aberrant motor output.
Collapse
Affiliation(s)
- Kathy Steece-Collier
- Department of Translational Science and Molecular Medicine, Michigan State University, Grand Rapids, MI 49503
| | | | | |
Collapse
|
30
|
Feyder M, Bonito-Oliva A, Fisone G. L-DOPA-Induced Dyskinesia and Abnormal Signaling in Striatal Medium Spiny Neurons: Focus on Dopamine D1 Receptor-Mediated Transmission. Front Behav Neurosci 2011; 5:71. [PMID: 22028687 PMCID: PMC3199545 DOI: 10.3389/fnbeh.2011.00071] [Citation(s) in RCA: 132] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2011] [Accepted: 10/06/2011] [Indexed: 11/13/2022] Open
Abstract
Dyskinesia is a serious motor complication caused by prolonged administration of l-DOPA to patients affected by Parkinson's disease. Accumulating evidence indicates that l-DOPA-induced dyskinesia (LID) is primarily caused by the development of sensitized dopamine D1 receptor (D1R) transmission in the medium spiny neurons (MSNs) of the striatum. This phenomenon, combined with chronic administration of l-DOPA, leads to persistent and intermittent hyper-activation of the cAMP signaling cascade. Activation of cAMP signaling results in increased activity of the cAMP-dependent protein kinase (PKA) and of the dopamine- and cAMP-dependent phosphoprotein of 32 kDa (DARPP-32), which regulate several downstream effector targets implicated in the control of the excitability of striatal MSNs. Dyskinesia is also accompanied by augmented activity of the extracellular signal-regulated kinases (ERK) and the mammalian target of rapamycin complex 1 (mTORC1), which are involved in the control of transcriptional and translational efficiency. Pharmacological or genetic interventions aimed at reducing abnormal signal transduction at the level of these various intracellular cascades have been shown to attenuate LID in different animal models. For instance, LID is reduced in mice deficient for DARPP-32, or following inhibition of PKA. Blockade of ERK obtained genetically or using specific inhibitors is also able to attenuate dyskinetic behavior in rodents and non-human primates. Finally, administration of rapamycin, a drug which blocks mTORC1, results in a strong reduction of LID. This review focuses on the abnormalities in signaling affecting the D1R-expressing MSNs and on their potential relevance for the design of novel anti-dyskinetic therapies.
Collapse
Affiliation(s)
- Michael Feyder
- Department of Neuroscience, Karolinska Institutet Stockholm, Sweden
| | | | | |
Collapse
|
31
|
Murer MG, Moratalla R. Striatal Signaling in L-DOPA-Induced Dyskinesia: Common Mechanisms with Drug Abuse and Long Term Memory Involving D1 Dopamine Receptor Stimulation. Front Neuroanat 2011; 5:51. [PMID: 21886608 PMCID: PMC3154293 DOI: 10.3389/fnana.2011.00051] [Citation(s) in RCA: 80] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2011] [Accepted: 07/25/2011] [Indexed: 11/25/2022] Open
Abstract
Parkinson’s disease is a common neurodegenerative disorder caused by the degeneration of midbrain substantia nigra dopaminergic neurons that project to the striatum. Despite extensive investigation aimed at finding new therapeutic approaches, the dopamine precursor molecule, 3,4-dihydroxyphenyl-l-alanine (l-DOPA), remains the most effective and commonly used treatment. However, chronic treatment and disease progression lead to changes in the brain’s response to l-DOPA, resulting in decreased therapeutic effect and the appearance of dyskinesias. l-DOPA-induced dyskinesia (LID) interferes significantly with normal motor activity and persists unless l-DOPA dosages are reduced to below therapeutic levels. Thus, controlling LID is one of the major challenges in Parkinson’s disease therapy. LID is the result of intermittent stimulation of supersensitive D1 dopamine receptors located in the very severely denervated striatal neurons. Through increased coupling to Gαolf, resulting in greater stimulation of adenylyl-cyclase, D1 receptors phosphorylate DARPP-32, and other protein kinase A targets. Moreover, D1 receptor stimulation activates extracellular signal-regulated kinase and triggers a signaling pathway involving mammalian target for rapamycin and modifications of histones that results in changes in translation, chromatin modification, and gene transcription. In turn, sensitization of D1 receptor signaling causes a widespread increase in the metabolic response to D1 agonists and changes in the activity of basal ganglia neurons that correlate with the severity of LID. Importantly, different studies suggest that dyskinesias may share mechanisms with drug abuse and long term memory involving D1 receptor activation. Here we review evidence implicating D1 receptor signaling in the genesis of LID, analyze mechanisms that may translate enhanced D1 signaling into dyskinetic movements, and discuss the possibility that the mechanisms underlying LID are not unique to the Parkinson’s disease brain.
Collapse
Affiliation(s)
- Mario Gustavo Murer
- Departamento de Fisiología y Biofísica, Facultad de Medicina, Universidad de Buenos Aires Buenos Aires, Argentina
| | | |
Collapse
|
32
|
Micheli F. Recent Advances in the Development of Dopamine D3 Receptor Antagonists: a Medicinal Chemistry Perspective. ChemMedChem 2011; 6:1152-62. [DOI: 10.1002/cmdc.201000538] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2011] [Indexed: 11/08/2022]
|
33
|
Effect of cyclosporin A on the uptake of D3-selective PET radiotracers in rat brain. Nucl Med Biol 2011; 38:725-39. [PMID: 21718948 DOI: 10.1016/j.nucmedbio.2011.01.002] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2010] [Revised: 12/28/2010] [Accepted: 01/03/2011] [Indexed: 11/23/2022]
Abstract
INTRODUCTION Four benzamide analogs having a high affinity and selectivity for D(3) versus D(2) receptors were radiolabeled with (11)C or (18)F for in vivo evaluation. METHODS Precursors were synthesized, and the four D(3) selective benzamide analogs were radiolabeled. The tissue distribution and brain uptake of the four compounds were evaluated in control rats and rats pretreated with cyclosporin A, a modulator of P-glycoprotein and an inhibitor of other ABC efflux transporters that contribute to the blood brain barrier. Micro-positron emission tomographic (PET) imaging was carried out for [(11)C]6 in a control and a cyclosporin A pretreated rat. RESULTS All four compounds showed low brain uptake in control rats at 5 and 30 min post-injection; despite recently reported rat behavioral studies conducted on analogs 6 (WC-10) and 7 (WC-44). Following administration of cyclosporin A, increased brain uptake was observed with all four PET radiotracers at both 5 and 30 min post-intravenous injection. An increase in brain uptake following modulation/inhibition of the ABC transporters was also observed in the microPET study. CONCLUSIONS These data suggest that D3 selective conformationally-flexible benzamide analogs which contain a N-2-methoxyphenylpiperazine moiety are substrates for P-glycoprotein or other adenosine triphosphate (ATP)-binding cassette transporters expressed at the blood-brain barrier, and that PET radiotracers containing this pharmacophore may display low brain uptake in rodents due to the action of these efflux transporters.
Collapse
|
34
|
Tu Z, Li S, Cui J, Xu J, Taylor M, Ho D, Luedtke RR, Mach RH. Synthesis and pharmacological evaluation of fluorine-containing D₃ dopamine receptor ligands. J Med Chem 2011; 54:1555-64. [PMID: 21348515 DOI: 10.1021/jm101323b] [Citation(s) in RCA: 40] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
A series of fluorine-containing N-(2-methoxyphenyl)piperazine and N-(2-fluoroethoxy)piperazine analogues were synthesized, and their affinities for human dopamine D(2), D(3), and D(4) receptors were determined. Radioligand binding studies identified five compounds, 18a, 20a, 20c, 20e, and 21e, which bind with high affinity at D(3) (K(i) = 0.17-5 nM) and moderate to high selectivity for D(3) vs D(2) receptors (ranging from ∼25- to 163-fold). These compounds were also evaluated for intrinsic activity at D(2) and D(3) receptors using a forskolin-dependent adenylyl cyclase assay. This panel of compounds exhibits varying receptor subtype binding selectivity and intrinsic activity at D(2) vs D(3) receptors. These compounds may be useful for behavioral pharmacology studies on the role of D(2)-like dopamine receptors in neuropsychiatric and neurological disorders. Furthermore, compound 20e, which has the highest binding affinity and selectivity for the D(3) receptor (K(i) = 0.17 nM for D(3), 163-fold selectivity for D(3) vs D(2) receptors), represents a candidate fluorine-18 radiotracer for in vivo PET imaging studies on the regulation of D(3) receptor expression.
Collapse
Affiliation(s)
- Zhude Tu
- Department of Radiology, Washington University School of Medicine, St. Louis, Missouri 63110, United States
| | | | | | | | | | | | | | | |
Collapse
|
35
|
Riddle LR, Kumar R, Griffin SA, Grundt P, Newman AH, Luedtke RR. Evaluation of the D3 dopamine receptor selective agonist/partial agonist PG01042 on L-dopa dependent animal involuntary movements in rats. Neuropharmacology 2011; 60:284-94. [PMID: 20850462 PMCID: PMC3820002 DOI: 10.1016/j.neuropharm.2010.09.011] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2009] [Revised: 09/07/2010] [Accepted: 09/08/2010] [Indexed: 10/19/2022]
Abstract
The substituted 4-phenylpiperazine D3 dopamine receptor selective antagonist PG01037 ((E)-N-(4-(4-(2,3-dichlorophenyl)piperazin-1-yl)but-2-enyl)-4-(pyridin-2-yl)benzamide) was reported to attenuate L-dopa-associated abnormal involuntary movements (AIMs) in unilaterally lesioned rats, a model of L-dopa-dependent dyskinesia in patients with Parkinson's Disease (Kumar et al., 2009a). We now report that PG01042 (N-(4-(4-(2,3-dichlorophenyl)piperazin-1-yl)butyl)-4-(pyridin-3-yl)benzamide), which is a D3 dopamine receptor selective agonist for adenylyl cyclase inhibition and a partial agonist for mitogenesis, is also capable of attenuating AIMs scores. The intrinsic activity of PG01037 and PG01042 were determined using a) a forskolin-dependent adenylyl cyclase inhibition assay and b) an assay for agonist-associated mitogenesis. It was observed that the in vivo efficacy of PG01042 increased when administered by intraperitoneal (i.p.) injection simultaneously with L-dopa/benserazide (8 mg/kg each), as compared to a 60 min or 30 min pretreatment. PG01042 was found to attenuate AIM scores in these animals in a dose dependent manner. While PG01042 did not effectively inhibit SKF 81297-dependent AIMs, it inhibited apomorphine-dependent AIM scores. Rotarod studies indicate that PG01042 at a dose of 10 mg/kg did not adversely affect motor coordination of the unilaterally lesioned rats. Evaluation of lesioned rats using a cylinder test behavioral paradigm indicated that PG01042 did not dramatically attenuate the beneficial effects of L-dopa. These studies and previously published studies suggest that both D3 dopamine receptor selective antagonists, partial agonists and agonists, as defined by an adenylyl cyclase inhibition assay and a mitogenic assay, are pharmacotherapeutic candidates for the treatment of L-dopa-associated dyskinesia in patients with Parkinson's Disease.
Collapse
Affiliation(s)
- Lindsay R. Riddle
- Department of Pharmacology and Neuroscience, University of North Texas Health Science Center, 3500 Camp Bowie, Fort Worth, TX 76107
| | - Rakesh Kumar
- Department of Pharmacology and Neuroscience, University of North Texas Health Science Center, 3500 Camp Bowie, Fort Worth, TX 76107
| | - Suzy A. Griffin
- Department of Pharmacology and Neuroscience, University of North Texas Health Science Center, 3500 Camp Bowie, Fort Worth, TX 76107
| | - Peter Grundt
- Medicinal Chemistry Section, National Institute on Drug Abuse-Intramural Research Program, NIH, 333 Cassell Drive, Baltimore, MD 21224
| | - Amy Hauck Newman
- Medicinal Chemistry Section, National Institute on Drug Abuse-Intramural Research Program, NIH, 333 Cassell Drive, Baltimore, MD 21224
| | - Robert R. Luedtke
- Department of Pharmacology and Neuroscience, University of North Texas Health Science Center, 3500 Camp Bowie, Fort Worth, TX 76107
| |
Collapse
|
36
|
Higley AE, Spiller K, Grundt P, Newman AH, Kiefer SW, Xi ZX, Gardner EL. PG01037, a novel dopamine D3 receptor antagonist, inhibits the effects of methamphetamine in rats. J Psychopharmacol 2011; 25:263-73. [PMID: 20142301 PMCID: PMC3729962 DOI: 10.1177/0269881109358201] [Citation(s) in RCA: 54] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
Our previous studies have shown that the selective dopamine D(3) receptor antagonists SB-277011A or NGB 2904 significantly attenuate cocaine self-administration under a progressive-ratio reinforcement schedule and cocaine-, methamphetamine- or nicotine-enhanced brain stimulation reward. However, the poor bioavailability of SB-277011A has limited its potential use in humans. In the present study, we investigated the effects of the novel D(3) receptor antagonist PG01037 on methamphetamine self-administration, methamphetamine-associated cue-induced reinstatement of drug seeking and methamphetamine-enhanced brain stimulation reward. Rats were allowed to intravenously self-administer methamphetamine under fixed-ratio 2 and progressive-ratio reinforcement conditions, and then the effects of PG01037 on methamphetamine self-administration and cue-induced reinstatement were assessed. Additional groups of rats were trained for intracranial electrical brain stimulation reward and the effects of PG01037 and methamphetamine on brain stimulation reward were assessed. Acute intraperitoneal administration of PG01037 (3, 10, 30 mg/kg) failed to alter methamphetamine or sucrose self-administration under fixed-ratio 2 reinforcement, but significantly lowered the break-point levels for methamphetamine or sucrose self-administration under progressive-ratio reinforcement. In addition, PG01037 significantly inhibited methamphetamine-associated cue-triggered reinstatement of drug-seeking behavior and methamphetamine-enhanced brain stimulation reward. These data suggest that the novel D(3) antagonist PG01037 significantly attenuates the rewarding effects as assessed by progressive-ratio self-administration and brain stimulation reward, and inhibits methamphetamine-associated cue-induced reinstatement of drug-seeking behavior These findings support the potential use of PG01037 or other selective D(3) antagonists in the treatment of methamphetamine addiction.
Collapse
Affiliation(s)
- Amanda E Higley
- Neuropsychopharmacology Section, Intramural Research Program, National Institute on Drug Abuse, National Institute of Health, Biomedical Research Center, 251 Bayview Boulevard, Baltimore, MD 21224 USA.
| | | | | | | | | | | | | |
Collapse
|
37
|
Orio L, Wee S, Newman AH, Pulvirenti L, Koob GF. The dopamine D3 receptor partial agonist CJB090 and antagonist PG01037 decrease progressive ratio responding for methamphetamine in rats with extended-access. Addict Biol 2010; 15:312-23. [PMID: 20456290 DOI: 10.1111/j.1369-1600.2010.00211.x] [Citation(s) in RCA: 41] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
Previous work suggests a role for dopamine D3-like receptors in psychostimulant reinforcement. The development of new compounds acting selectively at dopamine D3 receptors has opened new possibilities to explore the role of these receptors in animal models of psychostimulant dependence. Here we investigated whether the dopamine D3 partial agonist CJB090 (1-10 mg/kg, i.v) and the D3 antagonist PG01037 (8-32 mg/kg, s.c.) modified methamphetamine (0.05 mg/kg/injection) intravenous self-administration under fixed- (FR) and progressive- (PR) ratio schedules in rats allowed limited (short access, ShA; 1-hour sessions 3 days/week) or extended access (long access, LgA; 6 hour sessions 6 days/week). Under a FR1 schedule, the highest dose of the D3 partial agonist CJB090 selectively reduced methamphetamine self-administration in LgA but not in ShA rats, whereas the full D3 antagonist PG01037 produced no effect in either group. Under a PR schedule of reinforcement, the D3 partial agonist CJB090 reduced the maximum number of responses performed ('breakpoint') for methamphetamine in LgA rats at the doses of 5 and 10 mg/kg, and also it produced a significant reduction in the ShA group at the highest dose. However, the D3 full antagonist PG01037 only reduced PR methamphetamine self-administration in LgA rats at the highest dose of 32 mg/kg with no effect in the ShA group. The results suggest that rats might be more sensitive to pharmacological modulation of dopamine D3 receptors following extended access to methamphetamine self-administration, opening the possibility that D3 receptors play a role in excessive methamphetamine intake.
Collapse
Affiliation(s)
- Laura Orio
- The Committee on the Neurobiology of Addictive Disorders, The Scripps Research Institute, La Jolla, CA 92037, USA.
| | | | | | | | | |
Collapse
|
38
|
Mason CW, Hassan HE, Kim KP, Cao J, Eddington ND, Newman AH, Voulalas PJ. Characterization of the transport, metabolism, and pharmacokinetics of the dopamine D3 receptor-selective fluorenyl- and 2-pyridylphenyl amides developed for treatment of psychostimulant abuse. J Pharmacol Exp Ther 2010; 333:854-64. [PMID: 20228156 PMCID: PMC2879935 DOI: 10.1124/jpet.109.165084] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2010] [Accepted: 03/11/2010] [Indexed: 11/22/2022] Open
Abstract
The recent discovery of novel high-affinity and selective dopamine D3 receptor (DA D3R) antagonists and partial agonists has provided tools with which to further elucidate the role DA D3R plays in substance abuse. The present study was conducted to evaluate the transport, metabolism, pharmacokinetics, and brain uptake of the DA D3R-selective fluorenyl amides, NGB 2904 [N-(4-(4-(2,3-dichlorophenyl)piperazin-1-yl)butyl)-9H-fluorene-2-carboxamide] fumarate) and JJC 4-077 [N-(4-(4-(2,3-dichlorophenyl)piperazin-1-yl)-3-hydroxybutyl)-9H-fluorene-2-carboxamide hydrochloride], and the 2-pyridylphenyl amides, CJB 090 [N-(4-(4-(2,3-dichlorophenyl)piperazin-1-yl)butyl)-4-(pyridine-2-yl)benzamide hydrochloride] and PG 01037 [N-(4-(4-(2,3-dichlorophenyl)piperazin-1-yl)-trans-but-2-enyl)-4-(pyridine-2-yl)benzamide hydrochloride], all of which have been studied in animal models of psychostimulant abuse. Additional screening with a panel of human and rat Supersomes was performed for NGB 2904 and PG 01037. Drug-stimulated ATPase activation assays and bidirectional transport and efflux assays were used to test for substrate specificity of NGB 2904 and PG 01037 for human and rat efflux transporters. All compounds exhibited moderate elimination half-lives, ranging from 1.49 to 3.27 h, and large volumes of distribution (5.95-14.19 l/kg). The brain-to-plasma ratios ranged from 2.93 to 11.81 and were higher than those previously reported for cocaine. Brain exposure levels of NGB 2904 and PG 01037 were significantly reduced after intraperitoneal administration compared with intravenous administration. The metabolism of these compounds was mediated primarily by CYP3A subfamilies. PG 01037 was a P-glycoprotein-transported substrate. Higher doses of these compounds are often required for in vivo action, suggesting decreased bioavailability via extravascular administration that may be attributed to high drug efflux and hepatic metabolism. These studies provide important preclinical information for optimization of next-generation D3R selective agents for the treatment of drug addiction.
Collapse
Affiliation(s)
- Clifford W Mason
- Pharmacokinetics-Biopharmaceutics Laboratory, Department of Pharmaceutical Sciences, School of Pharmacy, University of Maryland, Baltimore, Maryland 21201, USA
| | | | | | | | | | | | | |
Collapse
|
39
|
Taylor M, Grundt P, Griffin SA, Newman AH, Luedtke RR. Dopamine D3 receptor selective ligands with varying intrinsic efficacies at adenylyl cyclase inhibition and mitogenic signaling pathways. Synapse 2010; 64:251-66. [PMID: 19924694 PMCID: PMC3821045 DOI: 10.1002/syn.20725] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Abstract
A panel of structurally related substituted 4-phenylpiperazines with nanomolar affinity and selectivity at D3 dopamine receptors has been synthesized. Compounds in which a heterocyclic (2-phenyl pyridyl, 3-phenyl pyridyl, benzothiophene, or benzofuran) moiety is adjacent to the amide was varied and/or a double bond (trans-butenyl) replaced the four-carbon aliphatic chain linking the arylamide with the 4-phenylpiperazine moiety were compared for (a) affinity at human D2 and D3 dopamine receptors, (b) intrinsic efficacy using an adenylyl cyclase inhibition assay, and (c) intrinsic efficacy using a mitogenic assay. All 16 compounds were (a) more efficacious for the D3 receptor cyclase inhibition assay than for the D3 receptor mitogenic assay and (b) exhibited the same or greater efficacy at D3 compared to D2 receptor (with the exception of one compound). Although the heterocyclic amide moiety appears to be the pivotal structural element determining the intrinsic efficacy of our D3 receptor selective compounds, the magnitude of the efficacy is modulated by the (a) substituent(s) on the phenyl piperazine and (b) the saturation of the four-carbon chain that links the arylamide and the phenylpiperazine. In addition, our ligands are functionally selective, because they can have differing intrinsic efficacies for the cyclase inhibition and the mitogenic activation signaling pathways. Compounds that are essentially full agonists at the cyclase assay appear to be only partial agonists in the mitogenic assay and compounds that are partial agonists in our cyclase assay are partial agonists or antagonists in the mitogenic assay.
Collapse
Affiliation(s)
- Michelle Taylor
- Department of Pharmacology and Neuroscience, University of North Texas Health Science Center, Fort Worth, TX,76109 USA
| | - Peter Grundt
- Medicinal Chemistry Section, NIDA-IRP, NIH, Baltimore, MD, USA
| | - Suzy A. Griffin
- Department of Pharmacology and Neuroscience, University of North Texas Health Science Center, Fort Worth, TX,76109 USA
| | | | - Robert R. Luedtke
- Department of Pharmacology and Neuroscience, University of North Texas Health Science Center, Fort Worth, TX,76109 USA
| |
Collapse
|