1
|
Fares MB, Alijevic O, Johne S, Overk C, Hashimoto M, Kondylis A, Adame A, Dulize R, Peric D, Nury C, Battey J, Guedj E, Sierro N, Mc Hugh D, Rockenstein E, Kim C, Rissman RA, Hoeng J, Peitsch MC, Masliah E, Mathis C. Nicotine-mediated effects in neuronal and mouse models of synucleinopathy. Front Neurosci 2023; 17:1239009. [PMID: 37719154 PMCID: PMC10501483 DOI: 10.3389/fnins.2023.1239009] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2023] [Accepted: 08/08/2023] [Indexed: 09/19/2023] Open
Abstract
Introduction Alpha-synuclein (α-Syn) aggregation, transmission, and contribution to neurotoxicity represent central mechanisms underlying Parkinson's disease. The plant alkaloid "nicotine" was reported to attenuate α-Syn aggregation in different models, but its precise mode of action remains unclear. Methods In this study, we investigated the effect of 2-week chronic nicotine treatment on α-Syn aggregation, neuroinflammation, neurodegeneration, and motor deficits in D-line α-Syn transgenic mice. We also established a novel humanized neuronal model of α-Syn aggregation and toxicity based on treatment of dopaminergic neurons derived from human induced pluripotent stem cells (iPSC) with α-Syn preformed fibrils (PFF) and applied this model to investigate the effects of nicotine and other compounds and their modes of action. Results and discussion Overall, our results showed that nicotine attenuated α-Syn-provoked neuropathology in both models. Moreover, when investigating the role of nicotinic acetylcholine receptor (nAChR) signaling in nicotine's neuroprotective effects in iPSC-derived dopaminergic neurons, we observed that while α4-specific antagonists reduced the nicotine-induced calcium response, α4 agonists (e.g., AZD1446 and anatabine) mediated similar neuroprotective responses against α-Syn PFF-provoked neurodegeneration. Our results show that nicotine attenuates α-Syn-provoked neuropathology in vivo and in a humanized neuronal model of synucleinopathy and that activation of α4β2 nicotinic receptors might mediate these neuroprotective effects.
Collapse
Affiliation(s)
| | - Omar Alijevic
- PMI R&D, Philip Morris Products S.A., Neuchâtel, Switzerland
| | - Stephanie Johne
- PMI R&D, Philip Morris Products S.A., Neuchâtel, Switzerland
| | - Cassia Overk
- Department of Neurosciences, University of California, San Diego, San Diego, CA, United States
| | - Makoto Hashimoto
- Department of Neurosciences, University of California, San Diego, San Diego, CA, United States
| | | | - Anthony Adame
- Department of Neurosciences, University of California, San Diego, San Diego, CA, United States
| | - Remi Dulize
- PMI R&D, Philip Morris Products S.A., Neuchâtel, Switzerland
| | - Dariusz Peric
- PMI R&D, Philip Morris Products S.A., Neuchâtel, Switzerland
| | - Catherine Nury
- PMI R&D, Philip Morris Products S.A., Neuchâtel, Switzerland
| | - James Battey
- PMI R&D, Philip Morris Products S.A., Neuchâtel, Switzerland
| | - Emmanuel Guedj
- PMI R&D, Philip Morris Products S.A., Neuchâtel, Switzerland
| | - Nicolas Sierro
- PMI R&D, Philip Morris Products S.A., Neuchâtel, Switzerland
| | - Damian Mc Hugh
- PMI R&D, Philip Morris Products S.A., Neuchâtel, Switzerland
| | - Edward Rockenstein
- Department of Neurosciences, University of California, San Diego, San Diego, CA, United States
| | - Changyoun Kim
- Department of Neurosciences, University of California, San Diego, San Diego, CA, United States
| | - Robert A. Rissman
- Department of Neurosciences, University of California, San Diego, San Diego, CA, United States
| | - Julia Hoeng
- PMI R&D, Philip Morris Products S.A., Neuchâtel, Switzerland
| | | | - Eliezer Masliah
- Department of Neurosciences, University of California, San Diego, San Diego, CA, United States
| | - Carole Mathis
- PMI R&D, Philip Morris Products S.A., Neuchâtel, Switzerland
| |
Collapse
|
2
|
Allosterism of Nicotinic Acetylcholine Receptors: Therapeutic Potential for Neuroinflammation Underlying Brain Trauma and Degenerative Disorders. Int J Mol Sci 2020; 21:ijms21144918. [PMID: 32664647 PMCID: PMC7404387 DOI: 10.3390/ijms21144918] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2020] [Revised: 07/06/2020] [Accepted: 07/10/2020] [Indexed: 12/21/2022] Open
Abstract
Inflammation is a key physiological phenomenon that can be pervasive when dysregulated. Persistent chronic inflammation precedes several pathophysiological conditions forming one of the critical cellular homeostatic checkpoints. With a steady global surge in inflammatory diseases, it is imperative to delineate underlying mechanisms and design suitable drug molecules targeting the cellular partners that mediate and regulate inflammation. Nicotinic acetylcholine receptors have a confirmed role in influencing inflammatory pathways and have been a subject of scientific scrutiny underlying drug development in recent years. Drugs designed to target allosteric sites on the nicotinic acetylcholine receptors present a unique opportunity to unravel the role of the cholinergic system in regulating and restoring inflammatory homeostasis. Such a therapeutic approach holds promise in treating several inflammatory conditions and diseases with inflammation as an underlying pathology. Here, we briefly describe the potential of cholinergic allosterism and some allosteric modulators as a promising therapeutic option for the treatment of neuroinflammation.
Collapse
|
3
|
Ionov ID, Pushinskaya II, Gorev NP, Shpilevaya LA. Synergistic anticataleptic effect of imipramine and nicotine in a rotenone-induced rat model. Psychopharmacology (Berl) 2019; 236:3125-3133. [PMID: 31069424 DOI: 10.1007/s00213-019-05261-9] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/23/2018] [Accepted: 04/26/2019] [Indexed: 12/21/2022]
Abstract
RATIONALE Some antidepressants have been previously found to produce anti-parkinsonian effect; nicotine was known to mitigate experimental neurotoxic lesions. The anticataleptic efficacy of antidepressant-nicotine co-administration is unstudied. OBJECTIVES This work aimed to evaluate anticataleptic action of imipramine-nicotine combination in rotenone model. METHODS Catalepsy was measured by the bar test. Concentrations of tyrosine hydroxylase, dopamine, and DOPAC were determined in the substantia nigra and dorsal striatum using ELISA and HPLC techniques; additionally, dopamine/DOPAC ratio was calculated for both areas. RESULTS Imipramine and nicotine alone were ineffective; however, co-administration of the drugs significantly (p < 0.01) inhibited rotenone-induced catalepsy and mitigated neurochemical changes in the nigrostriatal system. Anticataleptic effect of the combination exceeded that of levodopa, a standard drug for anti-parkinsonian treatment. CONCLUSION The combined use of imipramine and nicotine at relatively low doses inhibits neurotoxin-induced catalepsy and nigrostriatal neurochemical changes. The co-administration of these drugs might be a new approach to the treatment of extrapyramidal dysfunctions.
Collapse
Affiliation(s)
- Ilya D Ionov
- Centre on Theoretical Problems in Physical and Chemical Pharmacology, Russian Academy of Sciences, Leninsky prospect 123-4-63, Moscow, 117513, Russia.
| | | | | | | |
Collapse
|
4
|
Mouhape C, Costa G, Ferreira M, Abin-Carriquiry JA, Dajas F, Prunell G. Nicotine-Induced Neuroprotection in Rotenone In Vivo and In Vitro Models of Parkinson’s Disease: Evidences for the Involvement of the Labile Iron Pool Level as the Underlying Mechanism. Neurotox Res 2018; 35:71-82. [DOI: 10.1007/s12640-018-9931-1] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2018] [Revised: 06/18/2018] [Accepted: 07/05/2018] [Indexed: 11/29/2022]
|
5
|
Trenkwalder C, Berg D, Rascol O, Eggert K, Ceballos-Baumann A, Corvol JC, Storch A, Zhang L, Azulay JP, Broussolle E, Defebvre L, Geny C, Gostkowski M, Stocchi F, Tranchant C, Derkinderen P, Durif F, Espay AJ, Feigin A, Houeto JL, Schwarz J, Di Paolo T, Feuerbach D, Hockey HU, Jaeger J, Jakab A, Johns D, Linazasoro G, Maruff P, Rozenberg I, Sovago J, Weiss M, Gomez-Mancilla B. A Placebo-Controlled Trial of AQW051 in Patients With Moderate to Severe Levodopa-Induced Dyskinesia. Mov Disord 2016; 31:1049-54. [PMID: 26990766 DOI: 10.1002/mds.26569] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2014] [Revised: 01/12/2016] [Accepted: 01/14/2016] [Indexed: 01/22/2023] Open
Abstract
BACKGROUND This phase 2 randomized, double-blind, placebo-controlled study evaluated the efficacy and safety of the nicotinic acetylcholine receptor α7 agonist AQW051 in patients with Parkinson's disease and levodopa-induced dyskinesia. METHODS Patients with idiopathic Parkinson's disease and moderate to severe levodopa-induced dyskinesia were randomized to AQW051 10 mg (n = 24), AQW051 50 mg (n = 24), or placebo (n = 23) once daily for 28 days. Coprimary end points were change in Modified Abnormal Involuntary Movement Scale and Unified Parkinson's Disease Rating Scale part III scores. Secondary outcomes included pharmacokinetics. RESULTS In total, 67 patients completed the study. AQW051-treated patients experienced no significant improvements in Modified Abnormal Involuntary Movement Scale or Unified Parkinson's Disease Rating Scale part III scores by day 28. AQW051 was well tolerated; the most common adverse events were dyskinesia, fatigue, nausea, and falls. CONCLUSIONS AQW051 did not significantly reduce dyskinesia or parkinsonian severity. © 2016 International Parkinson and Movement Disorder Society.
Collapse
Affiliation(s)
- Claudia Trenkwalder
- Paracelsus-Elena Hospital, Kassel, Germany.,Department of Neurosurgery, University Medical Center, Goettingen, Germany.,German Parkinson Study Group, Marburg, Germany
| | - Daniela Berg
- German Parkinson Study Group, Marburg, Germany.,Department of Neurodegeneration, Hertie-Institute for Clinical Brain Research and German Center for Neurodegenerative Diseases, Tübingen, Germany
| | - Olivier Rascol
- Department of Clinical Pharmacology and Neurosciences, NeuroToul Excellence Center for Neurodegenerative Disorders, University UPS of Toulouse III, CIC-9302/INSERM UMR825, Hôpital Purpan - Pavillon Riser, Toulouse, France.,NS PARK/FCRIN Network, France
| | - Karla Eggert
- German Parkinson Study Group, Marburg, Germany.,Department of Neurology, Philipps-University of Marburg, Marburg, Germany
| | - Andres Ceballos-Baumann
- German Parkinson Study Group, Marburg, Germany.,Schön Klinik München-Schwabing, München, Germany
| | - Jean-Christophe Corvol
- NS PARK/FCRIN Network, France.,Sorbonne Universités, UPMC Univ Paris 06, and INSERM UMRS 1127 /CIC-1422, and CNRS UMR 7225, and AP-HP, and ICM, Hôpital Pitié-Salpêtrière, Département des maladies du système nerveux, Paris, France
| | - Alexander Storch
- German Parkinson Study Group, Marburg, Germany.,Division of Neurodegenerative Diseases, Department of Neurology, Dresden University of Technology, Dresden, Germany
| | - Lin Zhang
- Department of Neurology, UC Davis MIND Institute, Sacramento, California, USA
| | - Jean-Philippe Azulay
- NS PARK/FCRIN Network, France.,Service de Neurologie et pathologie du Mouvement, Hôpital de la Timone, Marseille Cedex, France
| | - Emmanuel Broussolle
- NS PARK/FCRIN Network, France.,Univisité Lyon 1, Hospices Civils de Lyon, Hôpital Neurologique Pierre Wertheimer, Service de Neurologie C, Lyon, France.,CNRS UMR 5229, Centre de Neurosciences Cognitives, Team Basal Ganglia, Bron, France
| | - Luc Defebvre
- NS PARK/FCRIN Network, France.,Service de Neurologie et Pathologie du movement, EA 1046, CHU de Lille, Hôpital Roger Salengro, Lille, France
| | - Christian Geny
- NS PARK/FCRIN Network, France.,Movement to Health (M2H) laboratory, Euromov, University Montpellier 1, Hôpital gui de Chauliac, Montpellier, France
| | - Michal Gostkowski
- Center for Neurological Restoration, Cleveland Clinic, Cleveland, Ohio, USA
| | - Fabrizio Stocchi
- Department of Neurology, Institute for Research and Medical Care, IROCS, Rome, Italy
| | - Christine Tranchant
- NS PARK/FCRIN Network, France.,Service de Neurologie, Hôpital de Hautepierre, Strasbourg, France
| | - Pascal Derkinderen
- NS PARK/FCRIN Network, France.,Centre Investigation Clinique Neurologie, CHU Nantes, Hôpital G&R Laennec, Nantes, France
| | - Franck Durif
- NS PARK/FCRIN Network, France.,Service de Neurologie, Hôpital Gabriel Montpied, Clermont-Ferrand, France
| | - Alberto J Espay
- Department of Neurology, James J. and Joan A. Gardner Family Center for Parkinson's Disease and Movement Disorders, University of Cincinnati Academic Health Center, Cincinnati, Ohio, USA
| | - Andrew Feigin
- Feinstein Institute for Medical Research, North Shore - LIJ Health System, Manhasset, New York, USA
| | - Jean-Luc Houeto
- NS PARK/FCRIN Network, France.,Service de Neurologie, CIC-INSERM 1402, CHU de Poitiers, Université de Poitiers, Poitiers, France
| | - Johannes Schwarz
- German Parkinson Study Group, Marburg, Germany.,Klinik Haag, Haag, Germany
| | - Thérèse Di Paolo
- Neuroscience Research Unit, Centre de recherche du CHU de Québec, Québec, Canada.,Faculty of Pharmacy, Laval University, Québec, Canada
| | | | - Hans-Ulrich Hockey
- Novartis Institutes for BioMedical Research, Basel, Switzerland.,Biometrics Matters Ltd, Hamilton, New Zealand
| | - Judith Jaeger
- Albert Einstein College of Medicine, New York, NY, USA, and CognitionMetrics, LLC, Wilmington, Delaware, USA
| | - Annamaria Jakab
- Novartis Institutes for BioMedical Research, Basel, Switzerland
| | - Donald Johns
- Novartis Pharmaceuticals Corporation, East Hanover, New Jersey, USA
| | | | | | | | - Judit Sovago
- Novartis Institutes for BioMedical Research, Basel, Switzerland
| | - Markus Weiss
- Novartis Institutes for BioMedical Research, Basel, Switzerland
| | - Baltazar Gomez-Mancilla
- Novartis Institutes for BioMedical Research, Basel, Switzerland.,Department of Neurology and Neurosurgery, McGill University, Montreal, Québec, Canada
| |
Collapse
|
6
|
Touchette JC, Breckenridge JM, Wilken GH, Macarthur H. Direct intranigral injection of dopaminochrome causes degeneration of dopamine neurons. Neurosci Lett 2015; 612:178-184. [PMID: 26704434 DOI: 10.1016/j.neulet.2015.12.028] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2015] [Revised: 12/11/2015] [Accepted: 12/12/2015] [Indexed: 12/21/2022]
Abstract
Parkinson's disease (PD) is characterized by progressive neurodegeneration of nigrastriatal dopaminergic neurons leading to clinical motor dysfunctions. Many animal models of PD have been developed using exogenous neurotoxins and pesticides. Evidence strongly indicates that the dopaminergic neurons of the substantia nigra pars compacta (SNpc) are highly susceptible to neurodegeneration due to a number of factors including oxidative stress and mitochondrial dysfunction. Oxidation of DA to a potential endogenous neurotoxin, dopaminochrome (DAC), may be a potential contributor to the vulnerability of the nigrostriatal tract to oxidative insult. In this study, we show that DAC causes slow and progressive degeneration of dopaminergic neurons in contrast to 1-methyl-4-phenylpyridinium (MPP(+)), which induces rapid lesions of the region. The DAC model may be more reflective of early stresses that initiate the progressive neurodegenerative process of PD, and may prove a useful model for future neurodegenerative studies.
Collapse
Affiliation(s)
- Jillienne C Touchette
- Department of Pharmacology and Physiology, Saint Louis University School of Medicine, 1402 S Grand Blvd, St. Louis, MO 63104, United States
| | - Julie M Breckenridge
- Department of Pharmacology and Physiology, Saint Louis University School of Medicine, 1402 S Grand Blvd, St. Louis, MO 63104, United States
| | - Gerald H Wilken
- Department of Pharmacology and Physiology, Saint Louis University School of Medicine, 1402 S Grand Blvd, St. Louis, MO 63104, United States
| | - Heather Macarthur
- Department of Pharmacology and Physiology, Saint Louis University School of Medicine, 1402 S Grand Blvd, St. Louis, MO 63104, United States.
| |
Collapse
|
7
|
Beckmann JS, Meyer AC, Pivavarchyk M, Horton DB, Zheng G, Smith AM, Wooters TE, McIntosh JM, Crooks PA, Bardo MT, Dwoskin LP. r-bPiDI, an α6β2* Nicotinic Receptor Antagonist, Decreases Nicotine-Evoked Dopamine Release and Nicotine Reinforcement. Neurochem Res 2015; 40:2121-30. [PMID: 26227997 DOI: 10.1007/s11064-015-1680-4] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2015] [Revised: 07/16/2015] [Accepted: 07/22/2015] [Indexed: 01/01/2023]
Abstract
α6β2* nicotinic acetylcholine receptors (nAChRs) expressed by dopaminergic neurons mediate nicotine-evoked dopamine (DA) release and nicotine reinforcement. α6β2* antagonists inhibit these effects of nicotine, such that α6β2* receptors serve as therapeutic targets for nicotine addiction. The present research assessed the neuropharmacology of 1,10-bis(3-methyl-5,6-dihydropyridin-1(2H)-yl)decane (r-bPiDI), a novel small-molecule, tertiary amino analog of its parent compound, N,N-decane-1,10-diyl-bis-3-picolinium diiodide (bPiDI). bPiDI was previously shown to inhibit both nicotine-evoked DA release and the reinforcing effects of nicotine. In the current study, r-bPiDI inhibition of [(3)H]nicotine and [(3)H]methyllycaconitine binding sites was evaluated to assess interaction with the recognition binding sites on α4β2* and α7* nAChRs, respectively. Further, r-bPiDI inhibition of nicotine-evoked DA release in vitro in the absence and presence of α-conotoxin MII and following chronic in vivo nicotine administration were determined. The ability of r-bPiDI to decrease nicotine self-administration and food-maintained responding was also assessed. Results show that r-bPiDI did not inhibit [(3)H]nicotine or [(3)H]methyllycaconitine binding, but potently (IC50 = 37.5 nM) inhibited nicotine-evoked DA release from superfused striatal slices obtained from either drug naïve rats or from those repeatedly treated with nicotine. r-bPiDI inhibition of nicotine-evoked DA release was not different in the absence or presence of α-conotoxin MII, indicating that r-bPiDI acts as a potent, selective α6β2* nAChR antagonist. Acute systemic administration of r-bPiDI specifically decreased nicotine self-administration by 75 %, and did not alter food-maintained responding, demonstrating greater specificity relative to bPiDI and bPiDDB, as well as the tertiary amino analog r-bPiDDB. The current work describes the discovery of r-bPiDI, a tertiary amino, α-conotoxin MII-like small molecule that acts as a potent and selective antagonist at α6β2* nAChRs to specifically decrease nicotine self-administration in rats, thus, establishing r-bPiDI as a lead compound for development as a treatment for nicotine addiction.
Collapse
Affiliation(s)
- Joshua S Beckmann
- Department of Psychology, University of Kentucky, Lexington, KY, USA
| | - Andrew C Meyer
- Department of Psychology, University of Kentucky, Lexington, KY, USA
| | - M Pivavarchyk
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Kentucky, 789 S. Limestone St., Lexington, KY, 40536-0596, USA
| | - David B Horton
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Kentucky, 789 S. Limestone St., Lexington, KY, 40536-0596, USA
| | - Guangrong Zheng
- Department of Pharmaceutical Sciences, University of Arkansas for Medical Sciences, Little Rock, AR, USA
| | - Andrew M Smith
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Kentucky, 789 S. Limestone St., Lexington, KY, 40536-0596, USA
| | - Thomas E Wooters
- Department of Psychology, University of Kentucky, Lexington, KY, USA
| | - J Michael McIntosh
- George E. Wahlen Veterans Affairs Medical Center and Departments of Psychiatry and Biology, University of Utah, Salt Lake City, UT, USA
| | - Peter A Crooks
- Department of Pharmaceutical Sciences, University of Arkansas for Medical Sciences, Little Rock, AR, USA
| | - Michael T Bardo
- Department of Psychology, University of Kentucky, Lexington, KY, USA
| | - Linda P Dwoskin
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Kentucky, 789 S. Limestone St., Lexington, KY, 40536-0596, USA.
| |
Collapse
|
8
|
Bordia T, McGregor M, Papke RL, Decker MW, McIntosh JM, Quik M. The α7 nicotinic receptor agonist ABT-107 protects against nigrostriatal damage in rats with unilateral 6-hydroxydopamine lesions. Exp Neurol 2014; 263:277-84. [PMID: 25261754 DOI: 10.1016/j.expneurol.2014.09.015] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2014] [Revised: 09/05/2014] [Accepted: 09/16/2014] [Indexed: 02/01/2023]
Abstract
The finding that smoking is inversely correlated with Parkinson's disease and that nicotine attenuates nigrostriatal damage in Parkinsonian animals supports the idea that nicotine may be neuroprotective. Nicotine is thought to exert this effect by acting at nicotinic receptors (nAChRs), including the α7 subtype. The objective of this study was twofold: first, to test the protective potential of ABT-107, an agonist with high selectivity for α7 nAChRs; and second, to investigate its cellular mechanism of action. Rats were implanted with minipumps containing ABT-107 (0.25mg/kg/d). In addition, we tested the effect of nicotine (1mg/kg/d) as a positive control, and also DMXB (2mg/kg/d) which acts primarily with α7 but also α4β2* nAChRs. Two weeks after minipump placement, the rats were lesioned by unilateral administration of 6-hydroxydopamine (6-OHDA) into the medial forebrain bundle. Lesioning alone decreased contralateral forelimb use and adjusted stepping, two measures of Parkinsonism. ABT-107 and nicotine treatment significantly improved these behaviors at all weeks tested, with variable improvement with DMXB. We next investigated the cellular mechanism involved. The striatal dopamine transporter (DAT), a marker of dopaminergic integrity, was reduced ~70% with lesioning. ABT-107 or nicotine treatment significantly increased DAT levels in lesioned striatum; these drugs did not alter DAT levels in intact striatum. ABT-107 and nicotine also significantly improved basal dopamine release from lesioned striatum, as well as nicotine-stimulated dopamine release mediated via α4β2* and α6β2* nAChRs. These data suggest that α7 nAChR agonists may improve motor behaviors associated with nigrostriatal damage by enhancing striatal dopaminergic function.
Collapse
Affiliation(s)
- Tanuja Bordia
- Center for Health Sciences, SRI International, 333 Ravenswood Ave., Menlo Park, CA 94025, USA
| | - Matthew McGregor
- Center for Health Sciences, SRI International, 333 Ravenswood Ave., Menlo Park, CA 94025, USA
| | - Roger L Papke
- Department of Pharmacology and Therapeutics, University of Florida College of Medicine, Gainesville, FL 3261, USA
| | - Michael W Decker
- AbbVie, Inc., 1 North Waukegan Road, North Chicago, IL 60064-6125, USA
| | - J Michael McIntosh
- George E. Wahlen Veterans Affairs Medical Center, Salt Lake City, UT 84148, USA; Department of Psychiatry, University of Utah, Salt Lake City, UT 84112, USA; Department of Biology, University of Utah, Salt Lake City, UT 84112, USA
| | - Maryka Quik
- Center for Health Sciences, SRI International, 333 Ravenswood Ave., Menlo Park, CA 94025, USA.
| |
Collapse
|
9
|
Michel PP, Toulorge D, Guerreiro S, Hirsch EC. Specific needs of dopamine neurons for stimulation in order to survive: implication for Parkinson disease. FASEB J 2013; 27:3414-23. [PMID: 23699175 DOI: 10.1096/fj.12-220418] [Citation(s) in RCA: 52] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Abstract
Parkinson disease (PD) is a degenerative brain disorder characterized by motor symptoms that are unequivocally associated with the loss of dopaminergic (DA) neurons in the substantia nigra (SN). Although our knowledge of the mechanisms that contribute to DA cell death in both hereditary and sporadic forms of the disease has advanced significantly, the nature of the pathogenic process remains poorly understood. In this review, we present evidence that neurodegeneration occurs when the electrical activity and excitability of these neurons is reduced. In particular, we will focus on the specific need these neurons may have for stimulation in order to survive and on the molecular and cellular mechanisms that may be compromised when this need is no longer met in PD.
Collapse
Affiliation(s)
- Patrick P Michel
- Université Pierre et Marie Curie-Paris 6, Centre de Recherche de l'Institut du Cerveau et de la Moelle Epinière, Unité Mixte de Recherche (UMR) S975, Paris, France.
| | | | | | | |
Collapse
|
10
|
Gill JK, Chatzidaki A, Ursu D, Sher E, Millar NS. Contrasting properties of α7-selective orthosteric and allosteric agonists examined on native nicotinic acetylcholine receptors. PLoS One 2013; 8:e55047. [PMID: 23383051 PMCID: PMC3558472 DOI: 10.1371/journal.pone.0055047] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2012] [Accepted: 12/18/2012] [Indexed: 12/04/2022] Open
Abstract
Subtype-selective ligands are important tools for the pharmacological characterisation of neurotransmitter receptors. This is particularly the case for nicotinic acetylcholine receptors (nAChRs), given the heterogeneity of their subunit composition. In addition to agonists and antagonists that interact with the extracellular orthosteric nAChR binding site, a series of nAChR allosteric modulators have been identified that interact with a distinct transmembrane site. Here we report studies conducted with three pharmacologically distinct nicotinic ligands, an orthosteric agonist (compound B), a positive allosteric modulator (TQS) and an allosteric agonist (4BP-TQS). The primary focus of the work described in this study is to examine the suitability of these compounds for the characterisation of native neuronal receptors (both rat and human). However, initial experiments were conducted on recombinant nAChRs demonstrating the selectivity of these three compounds for α7 nAChRs. In patch-clamp recordings on rat primary hippocampal neurons we found that all these compounds displayed pharmacological properties that mimicked closely those observed on recombinant α7 nAChRs. However, it was not possible to detect functional responses with compound B, an orthosteric agonist, using a fluorescent intracellular calcium assay on either rat hippocampal neurons or with human induced pluripotent stem cell-derived neurons (iCell neurons). This is, presumably, due to the rapid desensitisation of α7 nAChR that is induced by orthosteric agonists. In contrast, clear agonist-evoked responses were observed in fluorescence-based assays with the non-desensitising allosteric agonist 4BP-TQS and also when compound B was co-applied with the non-desensitising positive allosteric modulator TQS. In summary, we have demonstrated the suitability of subtype-selective orthosteric and allosteric ligands for the pharmacological identification and characterisation of native nAChRs and the usefulness of ligands that minimise receptor desensitisation for the characterisation of α7 nAChRs in fluorescence-based assays.
Collapse
Affiliation(s)
- JasKiran K. Gill
- Department of Neuroscience, Physiology and Pharmacology, University College London, London, United Kingdom
| | - Anna Chatzidaki
- Department of Neuroscience, Physiology and Pharmacology, University College London, London, United Kingdom
| | - Daniel Ursu
- Lilly Research Centre, Eli Lilly & Co. Ltd., Windlesham, Surrey, United Kingdom
| | - Emanuele Sher
- Lilly Research Centre, Eli Lilly & Co. Ltd., Windlesham, Surrey, United Kingdom
| | - Neil S. Millar
- Department of Neuroscience, Physiology and Pharmacology, University College London, London, United Kingdom
- * E-mail:
| |
Collapse
|
11
|
Suzuki S, Kawamata J, Matsushita T, Matsumura A, Hisahara S, Takata K, Kitamura Y, Kem W, Shimohama S. 3-[(2,4-dimethoxy)benzylidene]-anabaseine dihydrochloride protects against 6-hydroxydopamine-induced parkinsonian neurodegeneration through α7 nicotinic acetylcholine receptor stimulation in rats. J Neurosci Res 2012; 91:462-71. [DOI: 10.1002/jnr.23160] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2012] [Revised: 08/28/2012] [Accepted: 09/28/2012] [Indexed: 12/12/2022]
|
12
|
Quik M, Perez XA, Bordia T. Nicotine as a potential neuroprotective agent for Parkinson's disease. Mov Disord 2012; 27:947-57. [PMID: 22693036 DOI: 10.1002/mds.25028] [Citation(s) in RCA: 171] [Impact Index Per Article: 13.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2012] [Revised: 03/13/2012] [Accepted: 04/08/2012] [Indexed: 02/06/2023] Open
Abstract
Converging research efforts suggest that nicotine and other drugs that act at nicotinic acetylcholine receptors (nAChRs) may be beneficial in the management of Parkinson's disease. This idea initially stemmed from the results of epidemiological studies that demonstrated that smoking is associated with a decreased incidence of Parkinson's disease. The subsequent finding that nicotine administration protected against nigrostriatal damage in parkinsonian animal models led to the idea that nicotine in tobacco products may contribute to this apparent protective action. Nicotine most likely exerts its effects by interacting at nAChRs. Accumulating research indicates that multiple subtypes containing nAChRs, including α4β2, α6β2, and/or α7, may be involved. Stimulation of nAChRs initially activates various intracellular transduction pathways primarily via alterations in calcium signaling. Consequent adaptations in immune responsiveness and trophic factors may ultimately mediate nicotine's ability to reduce/halt the neuronal damage that arises in Parkinson's disease. In addition to a potential neuroprotective action, nicotine also has antidepressant properties and improves attention/cognition. Altogether, these findings suggest that nicotine and nAChR drugs represent promising therapeutic agents for the management of Parkinson's disease.
Collapse
Affiliation(s)
- Maryka Quik
- Center for Health Sciences, SRI International, Menlo Park, California, USA.
| | | | | |
Collapse
|
13
|
Neurotoxin-based models of Parkinson's disease. Neuroscience 2012; 211:51-76. [DOI: 10.1016/j.neuroscience.2011.10.057] [Citation(s) in RCA: 360] [Impact Index Per Article: 27.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2011] [Revised: 10/27/2011] [Accepted: 10/28/2011] [Indexed: 12/21/2022]
|
14
|
Xiong N, Long X, Xiong J, Jia M, Chen C, Huang J, Ghoorah D, Kong X, Lin Z, Wang T. Mitochondrial complex I inhibitor rotenone-induced toxicity and its potential mechanisms in Parkinson's disease models. Crit Rev Toxicol 2012; 42:613-32. [PMID: 22574684 DOI: 10.3109/10408444.2012.680431] [Citation(s) in RCA: 121] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
The etiology of Parkinson's disease (PD) is attributed to both environmental and genetic factors. The development of PD reportedly involves mitochondrial impairment, oxidative stress, α-synuclein aggregation, dysfunctional protein degradation, glutamate toxicity, calcium overloading, inflammation and loss of neurotrophic factors. Based on a link between mitochondrial dysfunction and pesticide exposure, many laboratories, including ours, have recently developed parkinsonian models by utilization of rotenone, a well-known mitochondrial complex I inhibitor. Rotenone models for PD appear to mimic most clinical features of idiopathic PD and recapitulate the slow and progressive loss of dopaminergic (DA) neurons and the Lewy body formation in the nigral-striatal system. Notably, potential human parkinsonian pathogenetic and pathophysiological mechanisms have been revealed through these models. In this review, we summarized various rotenone-based models for PD and discussed the implied etiology of and treatment for PD.
Collapse
Affiliation(s)
- Nian Xiong
- Department of Neurology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Hubei 430022, China
| | | | | | | | | | | | | | | | | | | |
Collapse
|
15
|
Carbone M, Duty S, Rattray M. Riluzole neuroprotection in a Parkinson's disease model involves suppression of reactive astrocytosis but not GLT-1 regulation. BMC Neurosci 2012; 13:38. [PMID: 22480308 PMCID: PMC3349538 DOI: 10.1186/1471-2202-13-38] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2011] [Accepted: 04/05/2012] [Indexed: 11/10/2022] Open
Abstract
Background Riluzole is a neuroprotective drug used in the treatment of motor neurone disease. Recent evidence suggests that riluzole can up-regulate the expression and activity of the astrocyte glutamate transporter, GLT-1. Given that regulation of glutamate transport is predicted to be neuroprotective in Parkinson's disease, we tested the effect of riluzole in parkinsonian rats which had received a unilateral 6-hydroxydopamine injection into the median forebrain bundle. Results Rats were treated with intraperitoneal riluzole (4 mg/kg or 8 mg/kg), 1 hour before the lesion then once daily for seven days. Riluzole produced a modest but significant attenuation of dopamine neurone degeneration, assessed by suppression of amphetamine-induced rotations, preservation of tyrosine hydroxylase positive neuronal cell bodies in the substantia nigra pars compacta and attenuation of striatal tyrosine hydroxylase protein loss. Seven days after 6-hydroxydopamine lesion, reactive astrocytosis was observed in the striatum, as determined by increases in expression of glial fibrillary acidic protein, however the glutamate transporter, GLT-1, which is also expressed in astrocytes was not regulated by the lesion. Conclusions The results confirm that riluzole is a neuroprotective agent in a rodent model of parkinson's disease. Riluzole administration did not regulate GLT-1 levels but significantly reduced GFAP levels, in the lesioned striatum. Riluzole suppression of reactive astrocytosis is an intriguing finding which might contribute to the neuroprotective effects of this drug.
Collapse
Affiliation(s)
- Marica Carbone
- King's College London, Wolfson Centre for Age-Related Diseases, Guy's Campus, London SE1 1UL, UK
| | | | | |
Collapse
|
16
|
Duty S, Jenner P. Animal models of Parkinson's disease: a source of novel treatments and clues to the cause of the disease. Br J Pharmacol 2012; 164:1357-91. [PMID: 21486284 DOI: 10.1111/j.1476-5381.2011.01426.x] [Citation(s) in RCA: 510] [Impact Index Per Article: 39.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Animal models of Parkinson's disease (PD) have proved highly effective in the discovery of novel treatments for motor symptoms of PD and in the search for clues to the underlying cause of the illness. Models based on specific pathogenic mechanisms may subsequently lead to the development of neuroprotective agents for PD that stop or slow disease progression. The array of available rodent models is large and ranges from acute pharmacological models, such as the reserpine- or haloperidol-treated rats that display one or more parkinsonian signs, to models exhibiting destruction of the dopaminergic nigro-striatal pathway, such as the classical 6-hydroxydopamine (6-OHDA) rat and 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP) mouse models. All of these have provided test beds in which new molecules for treating the motor symptoms of PD can be assessed. In addition, the emergence of abnormal involuntary movements (AIMs) with repeated treatment of 6-OHDA-lesioned rats with L-DOPA has allowed for examination of the mechanisms responsible for treatment-related dyskinesia in PD, and the detection of molecules able to prevent or reverse their appearance. Other toxin-based models of nigro-striatal tract degeneration include the systemic administration of the pesticides rotenone and paraquat, but whilst providing clues to disease pathogenesis, these are not so commonly used for drug development. The MPTP-treated primate model of PD, which closely mimics the clinical features of PD and in which all currently used anti-parkinsonian medications have been shown to be effective, is undoubtedly the most clinically-relevant of all available models. The MPTP-treated primate develops clear dyskinesia when repeatedly exposed to L-DOPA, and these parkinsonian animals have shown responses to novel dopaminergic agents that are highly predictive of their effect in man. Whether non-dopaminergic drugs show the same degree of predictability of response is a matter of debate. As our understanding of the pathogenesis of PD has improved, so new rodent models produced by agents mimicking these mechanisms, including proteasome inhibitors such as PSI, lactacystin and epoximycin or inflammogens like lipopolysaccharide (LPS) have been developed. A further generation of models aimed at mimicking the genetic causes of PD has also sprung up. Whilst these newer models have provided further clues to the disease pathology, they have so far been less commonly used for drug development. There is little doubt that the availability of experimental animal models of PD has dramatically altered dopaminergic drug treatment of the illness and the prevention and reversal of drug-related side effects that emerge with disease progression and chronic medication. However, so far, we have made little progress in moving into other pharmacological areas for the treatment of PD, and we have not developed models that reflect the progressive nature of the illness and its complexity in terms of the extent of pathology and biochemical change. Only when this occurs are we likely to make progress in developing agents to stop or slow the disease progression. The overarching question that draws all of these models together in the quest for better drug treatments for PD is how well do they recapitulate the human condition and how predictive are they of successful translation of drugs into the clinic? This article aims to clarify the current position and highlight the strengths and weaknesses of available models.
Collapse
Affiliation(s)
- Susan Duty
- King's College London, Wolfson Centre for Age-Related Disease, London, UK.
| | | |
Collapse
|
17
|
García-Montes JR, Boronat-García A, Drucker-Colín R. Pharmacological strategies for Parkinson’s disease. Health (London) 2012. [DOI: 10.4236/health.2012.431174] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
|
18
|
Toulorge D, Guerreiro S, Hild A, Maskos U, Hirsch EC, Michel PP. Neuroprotection of midbrain dopamine neurons by nicotine is gated by cytoplasmic Ca
2+. FASEB J 2011; 25:2563-73. [DOI: 10.1096/fj.11-182824] [Citation(s) in RCA: 67] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Affiliation(s)
- Damien Toulorge
- Université Pierre et Marie Curie‐Paris 6Centre de Recherche de l'Institut du Cerveau et de la Moelle EpinièreUnité Mixte de Recherche‐S975ParisFrance
- Institut National de la Santé et de la Recherche Médicale U975ParisFrance
- Centre National de la Recherche ScientifiqueUnité Mixte de Recherche 7225ParisFrance
| | - Serge Guerreiro
- Université Pierre et Marie Curie‐Paris 6Centre de Recherche de l'Institut du Cerveau et de la Moelle EpinièreUnité Mixte de Recherche‐S975ParisFrance
- Institut National de la Santé et de la Recherche Médicale U975ParisFrance
- Centre National de la Recherche ScientifiqueUnité Mixte de Recherche 7225ParisFrance
| | - Audrey Hild
- Université Pierre et Marie Curie‐Paris 6Centre de Recherche de l'Institut du Cerveau et de la Moelle EpinièreUnité Mixte de Recherche‐S975ParisFrance
- Institut National de la Santé et de la Recherche Médicale U975ParisFrance
- Centre National de la Recherche ScientifiqueUnité Mixte de Recherche 7225ParisFrance
| | - Uwe Maskos
- Neurobiologie Integrative des Systèmes CholinergiquesInstitut PasteurParisFrance
| | - Etienne C. Hirsch
- Université Pierre et Marie Curie‐Paris 6Centre de Recherche de l'Institut du Cerveau et de la Moelle EpinièreUnité Mixte de Recherche‐S975ParisFrance
- Institut National de la Santé et de la Recherche Médicale U975ParisFrance
- Centre National de la Recherche ScientifiqueUnité Mixte de Recherche 7225ParisFrance
| | - Patrick P. Michel
- Université Pierre et Marie Curie‐Paris 6Centre de Recherche de l'Institut du Cerveau et de la Moelle EpinièreUnité Mixte de Recherche‐S975ParisFrance
- Institut National de la Santé et de la Recherche Médicale U975ParisFrance
- Centre National de la Recherche ScientifiqueUnité Mixte de Recherche 7225ParisFrance
| |
Collapse
|
19
|
Austin PJ, Betts MJ, Broadstock M, O'Neill MJ, Mitchell SN, Duty S. Symptomatic and neuroprotective effects following activation of nigral group III metabotropic glutamate receptors in rodent models of Parkinson's disease. Br J Pharmacol 2010; 160:1741-53. [PMID: 20649576 DOI: 10.1111/j.1476-5381.2010.00820.x] [Citation(s) in RCA: 41] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022] Open
Abstract
BACKGROUND AND PURPOSE Increased glutamatergic innervation of the substantia nigra pars reticulata (SNpr) and pars compacta (SNpc) may contribute to the motor deficits and neurodegeneration, respectively, in Parkinson's disease (PD). This study aimed to establish whether activation of pre-synaptic group III metabotropic glutamate (mGlu) receptors reduced glutamate release in the SN, and provided symptomatic or neuroprotective relief in animal models of PD. EXPERIMENTAL APPROACH Broad-spectrum group III mGlu receptor agonists, O-phospho-l-serine (l-SOP) and l-2-amino-4-phosphonobutyrate (l-AP4), were assessed for their ability to inhibit KCl-evoked [(3)H]-d-aspartate release in rat nigral prisms or inhibit KCl-evoked endogenous glutamate release in the SNpr in vivo using microdialysis. Reversal of akinesia in reserpine-treated rats was assessed following intranigral injection of l-SOP and l-AP4. Finally, the neuroprotective effect of 7 days' supra-nigral treatment with l-AP4 was examined in 6-hydroxydopamine (6-OHDA)-lesioned rats. KEY RESULTS l-SOP and l-AP4 inhibited [(3)H]-d-aspartate release by 33 and 44% respectively. These effects were blocked by the selective group III mGlu antagonist (RS)-alpha-cyclopropyl-4-phosphonophenylglycine (CPPG). l-SOP also reduced glutamate release in the SNpr in vivo by 48%. Injection of l-SOP and l-AP4 into the SNpr reversed reserpine-induced akinesia. Following administration above the SNpc, l-AP4 provided neurochemical, histological and functional protection against 6-OHDA lesion of the nigrostriatal tract. Pretreatment with CPPG inhibited these effects. CONCLUSIONS AND IMPLICATIONS These findings highlight group III mGlu receptors in the SN as potential targets for providing both symptomatic and neuroprotective relief in PD, and indicate that inhibition of glutamate release in the SN may underlie these effects.
Collapse
Affiliation(s)
- P J Austin
- Department of Anatomy and Histology, Anderson Stuart Building, University of Sydney, Sydney, NSW, Australia
| | | | | | | | | | | |
Collapse
|
20
|
Xiong N, Huang J, Zhang Z, Zhang Z, Xiong J, Liu X, Jia M, Wang F, Chen C, Cao X, Liang Z, Sun S, Lin Z, Wang T. Stereotaxical infusion of rotenone: a reliable rodent model for Parkinson's disease. PLoS One 2009; 4:e7878. [PMID: 19924288 PMCID: PMC2774159 DOI: 10.1371/journal.pone.0007878] [Citation(s) in RCA: 80] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2009] [Accepted: 10/04/2009] [Indexed: 12/21/2022] Open
Abstract
A clinically-related animal model of Parkinson's disease (PD) may enable the elucidation of the etiology of the disease and assist the development of medications. However, none of the current neurotoxin-based models recapitulates the main clinical features of the disease or the pathological hallmarks, such as dopamine (DA) neuron specificity of degeneration and Lewy body formation, which limits the use of these models in PD research. To overcome these limitations, we developed a rat model by stereotaxically (ST) infusing small doses of the mitochondrial complex-I inhibitor, rotenone, into two brain sites: the right ventral tegmental area and the substantia nigra. Four weeks after ST rotenone administration, tyrosine hydroxylase (TH) immunoreactivity in the infusion side decreased by 43.7%, in contrast to a 75.8% decrease observed in rats treated systemically with rotenone (SYS). The rotenone infusion also reduced the DA content, the glutathione and superoxide dismutase activities, and induced alpha-synuclein expression, when compared to the contralateral side. This ST model displays neither peripheral toxicity or mortality and has a high success rate. This rotenone-based ST model thus recapitulates the slow and specific loss of DA neurons and better mimics the clinical features of idiopathic PD, representing a reliable and more clinically-related model for PD research.
Collapse
Affiliation(s)
- Nian Xiong
- Department of Neurology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Hubei, China
| | - Jinsha Huang
- Department of Neurology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Hubei, China
| | - Zhentao Zhang
- Department of Neurology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Hubei, China
| | - Zhaowen Zhang
- Department of Neurology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Hubei, China
| | - Jing Xiong
- Department of Neurology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Hubei, China
| | - Xingyuan Liu
- Department of Neurology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Hubei, China
| | - Min Jia
- Department of Neurology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Hubei, China
| | - Fang Wang
- Department of Neurology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Hubei, China
| | - Chunnuan Chen
- Department of Neurology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Hubei, China
| | - Xuebing Cao
- Department of Neurology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Hubei, China
| | - Zhihou Liang
- Department of Neurology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Hubei, China
| | - Shenggang Sun
- Department of Neurology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Hubei, China
| | - Zhicheng Lin
- Department of Psychiatry, Harvard Medical School, Boston, Massachusetts, United States of America
- Mailman Research Center, McLean Hospital, Belmont, Massachusetts, United States of America
| | - Tao Wang
- Department of Neurology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Hubei, China
- * E-mail:
| |
Collapse
|
21
|
Quik M, Campos C, Parameswaran N, Langston JW, McIntosh JM, Yeluashvili M. Chronic nicotine treatment increases nAChRs and microglial expression in monkey substantia nigra after nigrostriatal damage. J Mol Neurosci 2009; 40:105-13. [PMID: 19685015 DOI: 10.1007/s12031-009-9265-9] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2009] [Accepted: 07/20/2009] [Indexed: 11/27/2022]
Abstract
Our previous work had shown that long-term nicotine administration improved dopaminergic markers and nicotinic receptors (nAChRs) in the striatum of monkeys with nigrostriatal damage. The present experiments were done to determine whether nicotine treatment also led to changes in the substantia nigra, the region containing dopaminergic cell bodies. Monkeys were chronically treated with nicotine in the drinking water for 6 months after which they were injected with low dose of 1-methyl-4-phenyl-1,2,3,6-tetrahydrophridine (MPTP) for a further 6-month period. Nicotine was administered until the monkeys were euthanized 2 months after the last MPTP injection. Nicotine treatment did not affect the dopamine transporter or the number of tyrosine hydroxylase positive cells in the substantia nigra of lesioned monkeys. However, nicotine administration did lead to a greater increase in alpha3/alpha6beta2* and alpha4beta2* nAChRs in lesioned monkeys compared to controls. Nicotine also significantly elevated microglia and reduced the number of extracellular neuromelanin deposits in the substantia nigra of MPTP-lesioned monkeys. These findings indicate that long-term nicotine treatment modulates expression of several molecular measures in monkey substantia nigra that may result in an improvement in nigral integrity and/or function. These observations may have therapeutic implications for Parkinson's disease.
Collapse
Affiliation(s)
- Maryka Quik
- The Parkinson's Institute, Sunnyvale, CA 94085, USA.
| | | | | | | | | | | |
Collapse
|
22
|
Bencherif M. Neuronal nicotinic receptors as novel targets for inflammation and neuroprotection: mechanistic considerations and clinical relevance. Acta Pharmacol Sin 2009; 30:702-14. [PMID: 19498416 PMCID: PMC4002381 DOI: 10.1038/aps.2009.37] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2009] [Accepted: 03/09/2009] [Indexed: 01/18/2023] Open
Abstract
A number of studies have confirmed the potential for neuronal nicotinic acetylcholine receptor (NNR)-mediated neuroprotection and, more recently, its anti-inflammatory effects. The mechanistic overlap between these pathways and the ubiquitous effects observed following diverse insults suggest that NNRs modulate fundamental pathways involved in cell survival. These results have wide-reaching implications for the design of experimental therapeutics that regulate inflammatory and anti-apoptotic responses through NNRs and represent an initial step toward understanding the benefits of novel therapeutic strategies for the management of central nervous system disorders that target neuronal survival and associated inflammatory processes.
Collapse
|
23
|
Multiple roles for nicotine in Parkinson's disease. Biochem Pharmacol 2009; 78:677-85. [PMID: 19433069 DOI: 10.1016/j.bcp.2009.05.003] [Citation(s) in RCA: 94] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2009] [Revised: 04/30/2009] [Accepted: 05/01/2009] [Indexed: 01/11/2023]
Abstract
There exists a remarkable diversity of neurotransmitter compounds in the striatum, a pivotal brain region in the pathology of Parkinson's disease, a movement disorder characterized by rigidity, tremor and bradykinesia. The striatal dopaminergic system, which is particularly vulnerable to neurodegeneration in this disorder, appears to be the major contributor to these motor problems. However, numerous other neurotransmitter systems in the striatum most likely also play a significant role, including the nicotinic cholinergic system. Indeed, there is an extensive anatomical overlap between dopaminergic and cholinergic neurons, and acetylcholine is well known to modulate striatal dopamine release both in vitro and in vivo. Nicotine, a drug that stimulates nicotinic acetylcholine receptors (nAChRs), influences several functions relevant to Parkinson's disease. Extensive studies in parkinsonian animals show that nicotine protects against nigrostriatal damage, findings that may explain the well-established decline in Parkinson's disease incidence with tobacco use. In addition, recent work shows that nicotine reduces l-dopa-induced abnormal involuntary movements, a debilitating complication of l-dopa therapy for Parkinson's disease. These combined observations suggest that nAChR stimulation may represent a useful treatment strategy for Parkinson's disease for neuroprotection and symptomatic treatment. Importantly, only selective nAChR subtypes are present in the striatum including the alpha4beta2*, alpha6beta2* and alpha7 nAChR populations. Treatment with nAChR ligands directed to these subtypes may thus yield optimal therapeutic benefit for Parkinson's disease, with a minimum of adverse side effects.
Collapse
|
24
|
Quik M, O'Leary K, Tanner CM. Nicotine and Parkinson's disease: implications for therapy. Mov Disord 2009; 23:1641-52. [PMID: 18683238 DOI: 10.1002/mds.21900] [Citation(s) in RCA: 88] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
Accumulating evidence suggests that nicotine, a drug that stimulates nicotinic acetylcholine receptors, may be of therapeutic value in Parkinson's disease. Beneficial effects may be several-fold. One of these is a protective action against nigrostriatal damage. This possibility stems from the results of epidemiological studies that consistently demonstrate an inverse correlation between tobacco use and Parkinson's disease. This reduced incidence of Parkinson's disease has been attributed to the nicotine in tobacco products, at least in part, based on experimental work showing a protective effect of nicotine against toxic insults. Second, several studies suggest a symptomatic effect of nicotine in Parkinson's disease, although effects are small and somewhat variable. Third, recent data in nonhuman primates show that nicotine attenuates levodopa-induced dyskinesias, a debilitating side effect that develops in the majority of patients on levodopa therapy. Collectively, these observations suggest that nicotine or CNS selective nicotinic receptor ligands hold promise for Parkinson's disease therapy to reduce disease progression, improve symptoms, and/or decrease levodopa-induced dyskinesias.
Collapse
Affiliation(s)
- Maryka Quik
- The Parkinson's Institute, Sunnyvale, California, USA.
| | | | | |
Collapse
|
25
|
Nicotine induces sensitization of turning behavior in 6-hydroxydopamine lesioned rats. Neurotox Res 2009; 15:359-66. [PMID: 19384569 DOI: 10.1007/s12640-009-9041-1] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2008] [Revised: 01/16/2009] [Accepted: 03/03/2009] [Indexed: 10/20/2022]
Abstract
Nicotinic drugs have been proposed as putative drugs to treat Parkinson's disease (PD). In this study, we investigated whether nicotine can sensitize parkinsonian animals to the effect of dopaminergic drugs. Testing this hypothesis is important because nicotine has been shown to present neuroprotective and acute symptomatic effects on PD, but few studies have addressed the question of whether it may induce long-lasting effects on dopamine neurotransmission. We tested this hypothesis in the 6-hydroxydopamine (6-OHDA) rat model of PD. A pretreatment of these rats with 0.1-1.0 mg/kg nicotine induced a dose-dependent sensitization of the turning behavior when the animals were challenged with the dopamine receptor agonist apomorphine 24 h later. In agreement with previous studies, while apomorphine induced contraversive turns, nicotine, as well as amphetamine, induced ipsiversive turns in the 6-OHDA rats. This result suggests that, like amphetamine, nicotine induces turning behavior by promoting release of dopamine in the non-lesioned striatum of the rats. However, it is unlikely that the release of dopamine may also explain the nicotine-induced sensitization of turning behavior. First, the dopamine amount that could be released in the lesioned hemi-striatum by the nicotine pretreatment was minimum-less than 3%, as detected by HPLC-EC. Second, a pretreatment with amphetamine did not induce this behavioral sensitization. A pretreatment with apomorphine-induced sensitization, but it was minimal when compared to that induced by nicotine. Therefore, it is unlikely that the sensitization of the turning behavior induced by nicotine was consequent of the release of dopamine. However, the expression of such sensitization seems to depend on the activation of dopaminergic receptors, since it was seen when the nicotine-sensitized animals were challenged with apomorphine, but not with a second nicotine challenge. These findings are relevant for PD drug therapy since they suggest that the doses of dopaminergic drugs used to treat PD could be reduced if a nicotinic drug were co-administered.
Collapse
|
26
|
Huang LZ, Parameswaran N, Bordia T, Michael McIntosh J, Quik M. Nicotine is neuroprotective when administered before but not after nigrostriatal damage in rats and monkeys. J Neurochem 2009; 109:826-37. [PMID: 19250334 DOI: 10.1111/j.1471-4159.2009.06011.x] [Citation(s) in RCA: 54] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Nicotine reduces dopaminergic deficits in parkinsonian animals when administered before nigrostriatal damage. Here we tested whether nicotine is also beneficial when given to rats and monkeys with pre-existing nigrostriatal damage. Rats were administered nicotine before and after a unilateral 6-hydroxydopamine lesion of the medial forebrain bundle, and the results compared with those in which rats received nicotine only after lesioning. Nicotine pre-treatment attenuated behavioral deficits and lessened lesion-induced losses of the striatal dopamine transporter, and alpha6beta2* and alpha4beta2* nicotinic receptors (nAChRs). By contrast, nicotine administered 2 weeks after lesioning, when 6-hydroxydopamine-induced neurodegenerative effects are essentially complete, did not improve these same measures. Similar results were observed in 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine-lesioned monkeys. Nicotine did not enhance striatal markers when administered to monkeys with pre-existing nigrostriatal damage, in contrast to previous data that showed improvements when nicotine was given to monkeys before lesioning. These combined findings in two animal models suggest that nicotine is neuroprotective rather than neurorestorative against nigrostriatal damage. Receptor studies with (125)I-alpha-conotoxinMII and the alpha-conotoxinMII analog E11A were next performed to determine whether nicotine treatment pre- or post-lesioning differentially affected expression of alpha6alpha4beta2* and alpha6(nonalpha4)beta2* nAChR subtypes in striatum. The observations suggest that protection against nigrostriatal damage may be linked to striatal alpha6alpha4beta2* nAChRs.
Collapse
Affiliation(s)
- Luping Z Huang
- Basic Research Department, The Parkinson's Institute, Sunnyvale, California 94085, USA
| | | | | | | | | |
Collapse
|
27
|
Takeuchi H, Yanagida T, Inden M, Takata K, Kitamura Y, Yamakawa K, Sawada H, Izumi Y, Yamamoto N, Kihara T, Uemura K, Inoue H, Taniguchi T, Akaike A, Takahashi R, Shimohama S. Nicotinic receptor stimulation protects nigral dopaminergic neurons in rotenone-induced Parkinson's disease models. J Neurosci Res 2009; 87:576-85. [DOI: 10.1002/jnr.21869] [Citation(s) in RCA: 81] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
|
28
|
Abin-Carriquiry JA, Costa G, Urbanavicius J, Cassels BK, Rebolledo-Fuentes M, Wonnacott S, Dajas F. In vivo modulation of dopaminergic nigrostriatal pathways by cytisine derivatives: Implications for Parkinson's Disease. Eur J Pharmacol 2008; 589:80-4. [DOI: 10.1016/j.ejphar.2008.05.013] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2008] [Revised: 04/18/2008] [Accepted: 05/13/2008] [Indexed: 11/15/2022]
|
29
|
Riveles K, Huang LZ, Quik M. Cigarette smoke, nicotine and cotinine protect against 6-hydroxydopamine-induced toxicity in SH-SY5Y cells. Neurotoxicology 2008; 29:421-7. [PMID: 18359086 PMCID: PMC2486261 DOI: 10.1016/j.neuro.2008.02.001] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2007] [Revised: 01/23/2008] [Accepted: 02/05/2008] [Indexed: 11/22/2022]
Abstract
Epidemiological studies consistently demonstrate a reduced incidence of Parkinson's disease in smokers. As an approach to evaluate whether nicotine in tobacco may be involved in this apparent protective effect, we compared the effect of mainstream 1R4F cigarette smoke solutions, which contain chemicals inhaled by active smokers, and nicotine against 6-hydroxydopamine (6-OHDA)-induced toxicity in an in vitro cell culture system. For this purpose we used terminally differentiated SH-SY5Y neuroblastoma cells that exhibit a catecholaminergic phenotype and express nicotinic receptors. Cells were pre-incubated for 24 h in mainstream-cigarette smoke solutions (0.06, 0.2, or 0.6 cigarette puffs/ml) made from University of Kentucky 1R4F research brand cigarettes, followed by the addition of 6-OHDA for another 24-48 h. The 0.2, but not 0.06, puffs/ml dose, significantly protected against 6-OHDA-induced toxicity in SH-SY5Y cells. This dose yielded final nicotine concentrations of approximately 5 x 10(-7) M, which is similar to plasma smoking levels. Although the 0.6 puffs/ml dose caused significant toxicity on its own, it also appeared to protect against 6-OHDA-induced damage. We next tested the effect of nicotine, as well as its metabolite cotinine. These agents protected against the toxic effects of 6-OHDA in SH-SY5Y cells at concentrations ranging from 10(-7) to 10(-5) M. These combined results support the idea that nicotine is one of the components in cigarette smoke that has a protective effect against neurotoxic insults. These data suggest that nicotine may be of potential therapeutic value for Parkinson's disease.
Collapse
Affiliation(s)
| | | | - Maryka Quik
- The Parkinson’s Institute, Sunnyvale, CA 94085
| |
Collapse
|
30
|
Ward RJ, Lallemand F, de Witte P, Dexter DT. Neurochemical pathways involved in the protective effects of nicotine and ethanol in preventing the development of Parkinson's disease: potential targets for the development of new therapeutic agents. Prog Neurobiol 2008; 85:135-47. [PMID: 18482793 DOI: 10.1016/j.pneurobio.2008.03.003] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2007] [Revised: 01/07/2008] [Accepted: 03/18/2008] [Indexed: 12/26/2022]
Abstract
In this short review, neurochemical targets are identified where nicotine, and possibly ethanol, may interact to prevent the occurrence of Parkinson's disease. These are (a) the nicotinic acetycholine receptors present in the nigrostriatal area or on the surface of microglia, (b) monoamine oxidases and (c) inducible nitric oxide synthase. If such induced changes can be verified in clinical studies, this may help in the design of new therapeutic drugs which may be of relevance to diminish the incidence and perhaps the progression of the debilitating condition of Parkinson's disease.
Collapse
Affiliation(s)
- Roberta J Ward
- Laboratoire de Biologie du Comportement, Université Catholique de Louvain, 1 Croix du Sud, 1348 Louvain-la-Neuve, Belgium.
| | | | | | | |
Collapse
|
31
|
The exploration of rotenone as a toxin for inducing Parkinson's disease in rats, for application in BBB transport and PK–PD experiments. J Pharmacol Toxicol Methods 2008; 57:114-30. [DOI: 10.1016/j.vascn.2007.10.003] [Citation(s) in RCA: 51] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2007] [Accepted: 10/30/2007] [Indexed: 12/21/2022]
|
32
|
The cholinergic mesopontine tegmentum is a relatively neglected nicotinic master modulator of the dopaminergic system: relevance to drugs of abuse and pathology. Br J Pharmacol 2008; 153 Suppl 1:S438-45. [PMID: 18223661 DOI: 10.1038/bjp.2008.5] [Citation(s) in RCA: 92] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
The mammalian mesopontine tegmentum (MPT) contains two cholinergic nuclei, the pedunculopontine tegmental nucleus (PPTg) and the laterodorsal tegmental nucleus (LDTg). These provide the cholinergic innervation of, among other brain areas, the dopaminergic A9 and A10 cell groups. Their axons are thus the source of endogenous acetylcholine (ACh) acting on somato-dendritic acetylcholine receptors in the substantia nigra (SN) and ventral tegmental area (VTA). The anatomy, physiology, functional and pathological implications of these interactions with the nicotinic subtype of acetylcholine receptors (nAChRs) are discussed with a view of the important role of the MPT as a master regulator of nicotinic dopaminergic signalling in the brain, including for nicotine addiction.
Collapse
|
33
|
Quik M, Bordia T, O'Leary K. Nicotinic receptors as CNS targets for Parkinson's disease. Biochem Pharmacol 2007; 74:1224-34. [PMID: 17631864 PMCID: PMC2046219 DOI: 10.1016/j.bcp.2007.06.015] [Citation(s) in RCA: 75] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2007] [Revised: 06/09/2007] [Accepted: 06/12/2007] [Indexed: 10/23/2022]
Abstract
Parkinson's disease is a debilitating neurodegenerative movement disorder characterized by damage to the nigrostriatal dopaminergic system. Current therapies are symptomatic only and may be accompanied by serious side effects. There is therefore a continual search for novel compounds for the treatment of Parkinson's disease symptoms, as well as to reduce or halt disease progression. Nicotine administration has been reported to improve motor deficits that arise with nigrostriatal damage in parkinsonian animals and in Parkinson's disease. In addition, nicotine protects against nigrostriatal damage in experimental models, findings that have led to the suggestion that the reduced incidence of Parkinson's disease in smokers may be due to the nicotine in tobacco. Altogether, these observations suggest that nicotine treatment may be beneficial in Parkinson's disease. Nicotine interacts with multiple nicotinic receptor (nAChR) subtypes in the peripheral and central nervous system, as well as in skeletal muscle. Work to identify the subtypes affected in Parkinson's disease is therefore critical for the development of targeted therapies. Results show that striatal alpha6beta2-containing nAChRs are particularly susceptible to nigrostriatal damage, with a decline in receptor levels that closely parallels losses in striatal dopamine. In contrast, alpha4beta2-containing nAChRs are decreased to a much smaller extent under the same conditions. These observations suggest that development of nAChR agonists or antagonists targeted to alpha6beta2-containing nAChRs may represent a particularly relevant target for Parkinson's disease therapeutics.
Collapse
Affiliation(s)
- Maryka Quik
- The Parkinson's Institute, Sunnyvale, CA 94089, USA.
| | | | | |
Collapse
|
34
|
Urbanavicius J, Ferreira M, Costa G, Abin-Carriquiry JA, Wonnacott S, Dajas F. Nicotine induces tyrosine hydroxylase plasticity in the neurodegenerating striatum. J Neurochem 2007; 102:723-30. [PMID: 17437548 DOI: 10.1111/j.1471-4159.2007.04560.x] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
Abstract
It has been shown that nicotine prevents the loss of dopamine (DA) in the corpus striatum (CS) after 6-hydroxydopamine injection in the substantia nigra. To study the role of the enzyme tyrosine hydroxylase (TH; EC 1.14.16.2) in this experimental paradigm, we have examined its activity by assessing the accumulation of l-3,4-dihydroxyphenylalanine after inhibiting the subsequent enzyme in the DA synthetic pathway, aromatic l-amino acid decarboxylase, with 3-hydroxybenzylhydrazine. In addition the amount of TH protein was assessed by western blotting and its distribution in the CS was examined using immunohistochemical methods. 6-hydroxydopamine injection produced a significant decrease in DA levels and l-3,4-dihydroxyphenylalanine accumulation, as well as decreases in TH protein and TH immunoreactive fibres in the CS. After nicotine treatment, the decrease in TH protein in the CS was significantly reduced, with a concomitant preservation of TH activity, but nicotine did not alter the number of TH immunoreactive fibres. The activity and amount of TH did not change in the contralateral (intact) CS. Thus, nicotine induces long lasting TH plasticity in the degenerating CS. A synergistic action of nicotine-activated and lesion-originated signals appears necessary for the expression of this neuronal molecular plasticity.
Collapse
Affiliation(s)
- Jessika Urbanavicius
- Department of Neurochemistry, Instituto de Investigaciones Biológicas Clemente Estable, Montevideo, Uruguay
| | | | | | | | | | | |
Collapse
|
35
|
Quik M, O'Neill M, Perez XA. Nicotine neuroprotection against nigrostriatal damage: importance of the animal model. Trends Pharmacol Sci 2007; 28:229-35. [PMID: 17412429 DOI: 10.1016/j.tips.2007.03.001] [Citation(s) in RCA: 58] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2006] [Revised: 01/19/2007] [Accepted: 03/19/2007] [Indexed: 10/23/2022]
Abstract
Parkinson's disease is a neurodegenerative movement disorder that is characterized by a loss of nigrostriatal dopamine-containing neurons. Unexpectedly, there is a reduced incidence of Parkinson's disease in tobacco users. This finding is important because the identification of the component(s) responsible for this effect could lead to therapeutic strategies to slow down or halt the progression of Parkinson's disease. Results from cell culture models consistently show that nicotine protects against neurotoxicity. However, data from animal models of nigrostriatal damage are conflicting, thus raising questions about a neuroprotective role of nicotine. Accumulating evidence indicates that discrepancies are observed primarily in mouse models of the disease. By contrast, reproducible protection occurs in rat models and in a nonhuman primate parkinsonian model that closely resembles the human disease. These findings highlight the need to use the appropriate animal model and treatment conditions when testing putative neuroprotective agents.
Collapse
Affiliation(s)
- Maryka Quik
- The Parkinson's Institute, 1170 Morse Avenue, Sunnyvale, CA 94089, USA.
| | | | | |
Collapse
|
36
|
Janhunen S, Ahtee L. Differential nicotinic regulation of the nigrostriatal and mesolimbic dopaminergic pathways: implications for drug development. Neurosci Biobehav Rev 2006; 31:287-314. [PMID: 17141870 DOI: 10.1016/j.neubiorev.2006.09.008] [Citation(s) in RCA: 58] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2006] [Revised: 09/10/2006] [Accepted: 09/18/2006] [Indexed: 01/21/2023]
Abstract
Neuronal nicotinic acetylcholine receptors (nAChRs) modulate dopaminergic function. Discovery of their multiplicity has lead to the search for subtype-selective nAChR agonists that might be therapeutically beneficial in diseases linked to brain dopaminergic pathways. The regulation and responses of the nigrostriatal and mesolimbic dopaminergic pathways are often similar, but some differences do exist. The cerebral distribution and characteristics of various nAChR subtypes differ between nigrostriatal and mesolimbic dopaminergic pathways. Comparison of nicotine and epibatidine, two nAChR agonists whose relative affinities for various nAChR subtypes differ, revealed differences in the nAChR-mediated regulation of dopaminergic activation between these dopamine systems. Nicotine preferentially stimulates the mesolimbic pathway, whereas epibatidine's stimulatory effect falls on the nigrostriatal pathway. Thus, it may be possible to stimulate the nigrostriatal pathway with selective nAChR agonists that do not significantly affect the mesolimbic pathway, and thus lack addictive properties. Furthermore, dopamine uptake inhibition revealed a novel inhibitory effect of epibatidine on accumbal dopamine release, which could form a basis for novel antipsychotics that could alleviate the elevated accumbal dopaminergic tone found in schizophrenia during the active psychotic state. Different regulation of nigrostriatal and mesolimbic dopaminergic pathways by nAChRs could be an important basis for developing novel drugs for treatment of Parkinson's disease and schizophrenia.
Collapse
Affiliation(s)
- Sanna Janhunen
- Division of Pharmacology and Toxicology, Faculty of Pharmacy, University of Helsinki, P.O. Box 56 (Viikinkaari 5), Helsinki, FIN-00014, Finland.
| | | |
Collapse
|