1
|
Markowitz JS, Melchert PW. The Pharmacokinetics and Pharmacogenomics of Psychostimulants. Child Adolesc Psychiatr Clin N Am 2022; 31:393-416. [PMID: 35697392 DOI: 10.1016/j.chc.2022.03.003] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/01/2022]
Abstract
The psychostimulants-amphetamine and methylphenidate-have been in clinical use for well more than 60 years. In general, both stimulants are rapidly absorbed with relatively poor bioavailability and short half-lives. The pharmacokinetics of both stimulants are generally linear and dose proportional although substantial interindividual variability in pharmacokinetics is in evidence. Amphetamine (AMP) is highly metabolized by several oxidative enzymes forming multiple metabolites while methylphenidate (MPH) is primarily metabolized by hydrolysis to the inactive metabolite ritalinic acid. At present, pharmacogenomic testing as an aid to guide dosing and personalized treatment cannot be recommended for either agent. Few pharmacokinetically based drug-drug interactions (DDIs) have been documented for either stimulant.
Collapse
Affiliation(s)
- John S Markowitz
- Department of Pharmacotherapy and Translational Research, College of Pharmacy, University of Florida, Gainesville, FL 32610-0486, USA; Center for Pharmacogenomics and Precision Medicine, College of Pharmacy, University of Florida, Gainesville, FL 32610-0486, USA.
| | - Philip W Melchert
- Department of Pharmacotherapy and Translational Research, College of Pharmacy, University of Florida, Gainesville, FL 32610-0486, USA
| |
Collapse
|
2
|
Brown DP, Rogers DT, Pomerleau F, Siripurapu KB, Kulshrestha M, Gerhardt GA, Littleton JM. Novel multifunctional pharmacology of lobinaline, the major alkaloid from Lobelia cardinalis. Fitoterapia 2016; 111:109-23. [PMID: 27105955 PMCID: PMC5299595 DOI: 10.1016/j.fitote.2016.04.013] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2016] [Revised: 04/15/2016] [Accepted: 04/17/2016] [Indexed: 01/18/2023]
Abstract
In screening a library of plant extracts from ~1000 species native to the Southeastern United States, Lobelia cardinalis was identified as containing nicotinic acetylcholine receptor (nicAchR) binding activity which was relatively non-selective for the α4β2- and α7-nicAchR subtypes. This nicAchR binding profile is atypical for plant-derived nicAchR ligands, the majority of which are highly selective for α4β2-nicAchRs. Its potential therapeutic relevance is noteworthy since agonism of α4β2- and α7-nicAchRs is associated with anti-inflammatory and neuroprotective properties. Bioassay-guided fractionation of L. cardinalis extracts led to the identification of lobinaline, a complex binitrogenous alkaloid, as the main source of the unique nicAchR binding profile. Purified lobinaline was a potent free radical scavenger, displayed similar binding affinity at α4β2- and α7-nicAchRs, exhibited agonist activity at nicAchRs in SH-SY5Y cells, and inhibited [(3)H]-dopamine (DA) uptake in rat striatal synaptosomes. Lobinaline significantly increased fractional [(3)H] release from superfused rat striatal slices preloaded with [(3)H]-DA, an effect that was inhibited by the non-selective nicAchR antagonist mecamylamine. In vivo electrochemical studies in urethane-anesthetized rats demonstrated that lobinaline locally applied in the striatum significantly prolonged clearance of exogenous DA by the dopamine transporter (DAT). In contrast, lobeline, the most thoroughly investigated Lobelia alkaloid, is an α4β2-nicAchR antagonist, a poor free radical scavenger, and is a less potent DAT inhibitor. These previously unreported multifunctional effects of lobinaline make it of interest as a lead to develop therapeutics for neuropathological disorders that involve free radical generation, cholinergic, and dopaminergic neurotransmission. These include neurodegenerative conditions, such as Parkinson's disease, and drug abuse.
Collapse
Affiliation(s)
- Dustin P Brown
- College of Medicine, Department of Anatomy & Neurobiology, University of Kentucky Chandler Medical Center, 138 Leader Avenue, Lexington, KY 40536-9983, USA
| | - Dennis T Rogers
- Naprogenix™, UK-AsTeCC, 145 Graham Avenue, Lexington, KY 40506-0286, USA.
| | - Francois Pomerleau
- College of Medicine, Department of Anatomy & Neurobiology, University of Kentucky Chandler Medical Center, 138 Leader Avenue, Lexington, KY 40536-9983, USA; College of Medicine, Parkinson's Disease Translational Research Center for Excellence, University of Kentucky Chandler Medical Center, 138 Leader Avenue, Lexington, KY 40536-9983, USA; College of Medicine, Center for Microelectrode Technology, University of Kentucky Chandler Medical Center, 138 Leader Avenue, Lexington, KY 40536-9983, USA
| | - Kirin B Siripurapu
- College of Arts and Sciences, Department of Psychology, University of Kentucky, Kastle Hall, Lexington, KY 40506-0044, USA
| | - Manish Kulshrestha
- College of Agriculture, Department of Biosystems & Agricultural Engineering, University of Kentucky, 1100 S. Limestone, Lexington, KY 40546-0091, USA
| | - Greg A Gerhardt
- College of Medicine, Department of Anatomy & Neurobiology, University of Kentucky Chandler Medical Center, 138 Leader Avenue, Lexington, KY 40536-9983, USA; College of Medicine, Department of Neurology, University of Kentucky Chandler Medical Center, 138 Leader Avenue, Lexington, KY 40536-9983, USA; College of Medicine, Department of Psychiatry, University of Kentucky Chandler Medical Center, 138 Leader Avenue, Lexington, KY 40536-9983, USA; College of Medicine, Department of Neurosurgery, University of Kentucky Chandler Medical Center, 138 Leader Avenue, Lexington, KY 40536-9983, USA; College of Medicine, Parkinson's Disease Translational Research Center for Excellence, University of Kentucky Chandler Medical Center, 138 Leader Avenue, Lexington, KY 40536-9983, USA; College of Medicine, Center for Microelectrode Technology, University of Kentucky Chandler Medical Center, 138 Leader Avenue, Lexington, KY 40536-9983, USA
| | - John M Littleton
- Naprogenix™, UK-AsTeCC, 145 Graham Avenue, Lexington, KY 40506-0286, USA; College of Arts and Sciences, Department of Psychology, University of Kentucky, Kastle Hall, Lexington, KY 40506-0044, USA
| |
Collapse
|
3
|
Antel J, Albayrak Ö, Heusch G, Banaschewski T, Hebebrand J. Assessment of potential cardiovascular risks of methylphenidate in comparison with sibutramine: do we need a SCOUT (trial)? Eur Arch Psychiatry Clin Neurosci 2015; 265:233-47. [PMID: 25149468 DOI: 10.1007/s00406-014-0522-8] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/18/2013] [Accepted: 08/01/2014] [Indexed: 12/20/2022]
Abstract
With the recent approval of methylphenidate (MPH) for treating attention-deficit/hyperactivity disorder (ADHD) in adults, the number of patients exposed will increase tremendously. The ongoing debate on the cardiovascular safety of MPH has triggered two large retrospective cohort studies in children and adolescents as well as in young to middle-aged adults. These studies looked into serious cardiovascular events (sudden cardiac death, acute myocardial infarction and stroke) as primary endpoints and concluded that MPH was safe after a mean duration of 2.1 years of follow-up in children and adolescents and mean duration of 0.33 years of current use in adults. The results are encouraging with respect to the short- and medium-term use of MPH. Without the inherent limitations of retrospective cohort studies, a prospective randomized, double-blind, placebo-controlled, multicenter trial in individuals stratified for cardiovascular risk factors would allow for an optimized risk assessment. With many millions of patients treated per year and drawing parallels to the lately discovered risks of sibutramine, another sympathomimetic with an overlapping mode of action and similar side effects on heart rate and blood pressure, we hypothesize that such a trial might be a dedicated risk mitigation strategy for public health. A critical assessment of cardiovascular side effects of MPH appears particularly warranted, because ADHD is associated with obesity, smoking and poor health in general. We summarize recent findings with the focus on cardiovascular risks of MPH in humans; we additionally analyze the limited number of rodent studies that have addressed cardiovascular risks of MPH.
Collapse
Affiliation(s)
- Jochen Antel
- Research-Unit of the Department of Child and Adolescent Psychiatry, Psychosomatics, and Psychotherapy, LVR-Klinikum Essen, University of Duisburg-Essen, IG1 Virchowstr. 171, 45147, Essen, Germany,
| | | | | | | | | |
Collapse
|
4
|
Staller JA, Faraone SV. Targeting the dopamine system in the treatment of attention-deficit/hyperactivity disorder. Expert Rev Neurother 2014; 7:351-62. [PMID: 17425490 DOI: 10.1586/14737175.7.4.351] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023]
Abstract
Attention-deficit/hyperactivity disorder (ADHD) is a highly heritable condition that affects a significant number of children and adults worldwide. During the past 30 years, the diagnosis and treatment of ADHD has relied on clinical assessment and empirical experience with stimulant medications. More recently, advances in family genetic studies, molecular genetic studies, preclinical research, radiographic imaging techniques and neuropsychological evaluation have significantly enhanced our understanding of the neurobiology of ADHD. This review highlights the current central role of dopamine in the pathophysiology and treatment of ADHD and implications for future advances in diagnosis and treatment.
Collapse
Affiliation(s)
- Jud A Staller
- Division of Child & Adolescent Psychiatry, SUNY Upstate Medical University, Syracuse, NY 13210, USA.
| | | |
Collapse
|
5
|
Abstract
Amphetamine was discovered over 100 years ago. Since then, it has transformed from a drug that was freely available without prescription as a panacea for a broad range of disorders into a highly restricted Controlled Drug with therapeutic applications restricted to attention deficit hyperactivity disorder (ADHD) and narcolepsy. This review describes the relationship between chemical structure and pharmacology of amphetamine and its congeners. Amphetamine's diverse pharmacological actions translate not only into therapeutic efficacy, but also into the production of adverse events and liability for recreational abuse. Accordingly, the balance of benefit/risk is the key challenge for its clinical use. The review charts advances in pharmaceutical development from the introduction of once-daily formulations of amphetamine through to lisdexamfetamine, which is the first d-amphetamine prodrug approved for the management of ADHD in children, adolescents and adults. The unusual metabolic route for lisdexamfetamine to deliver d-amphetamine makes an important contribution to its pharmacology. How lisdexamfetamine's distinctive pharmacokinetic/pharmacodynamic profile translates into sustained efficacy as a treatment for ADHD and its reduced potential for recreational abuse is also discussed.
Collapse
Affiliation(s)
| | | | | | - David J Nutt
- Department of Neuropsychopharmacology and Molecular Imaging, Division of Neuroscience & Mental Health, Imperial College London, London, UK
| |
Collapse
|
6
|
Kern CH, Smith DR. Preweaning Mn exposure leads to prolonged astrocyte activation and lasting effects on the dopaminergic system in adult male rats. Synapse 2010; 65:532-44. [PMID: 20963817 DOI: 10.1002/syn.20873] [Citation(s) in RCA: 54] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2010] [Accepted: 09/28/2010] [Indexed: 01/18/2023]
Abstract
Little is known about the effects of manganese (Mn) exposure over neurodevelopment and whether these early insults result in effects lasting into adulthood. To determine if early Mn exposure produces lasting neurobehavioral and neurochemical effects, we treated neonate rats with oral Mn (0, 25, or 50 mg Mn/kg/d over PND 1-21) and evaluated (1) behavioral performance in the open arena in the absence (PND 97) and presence (PND 98) of a d-amphetamine challenge, (2) brain dopamine D1 and D2-like receptors and dopamine transporter densities in the prefrontal cortex, striatum, and nucleus accumbens (PND 107), and (3) astrocyte marker glial fibrillary acidic protein (GFAP) levels in these same brain regions (PND 24 and 107). We found that preweaning Mn exposure did not alter locomotor activity or behavior disinhibition in adult rats, though Mn-exposed animals did exhibit an enhanced locomotor response to d-amphetamine challenge. Preweaning Mn exposure led to increased D1 and D2 receptor levels in the nucleus accumbens and prefrontal cortex, respectively, compared with controls. We also found increased GFAP expression in the prefrontal cortex in Mn-exposed PND 24 weanlings, and increased GFAP levels in prefrontal cortex, medial striatum and nucleus accumbens of adult (PND 107) rats exposed to preweaning Mn, indicating an effect of Mn exposure on astrogliosis that persisted and/or progressed to other brain regions in adult animals. These data show that preweaning Mn exposure leads to lasting molecular and functional impacts in multiple brain regions of adult animals, long after brain Mn levels returned to normal.
Collapse
Affiliation(s)
- Cynthia H Kern
- Department of Microbiology and Environmental Toxicology, University of California, Santa Cruz, California 95064, USA
| | | |
Collapse
|
7
|
Vanattou-Saïfoudine N, McNamara R, Harkin A. Mechanisms mediating the ability of caffeine to influence MDMA ('Ecstasy')-induced hyperthermia in rats. Br J Pharmacol 2010; 160:860-77. [PMID: 20590585 DOI: 10.1111/j.1476-5381.2010.00660.x] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022] Open
Abstract
BACKGROUND AND PURPOSE Caffeine exacerbates the hyperthermia associated with an acute exposure to 3,4 methylenedioxymethamphetamine (MDMA, 'Ecstasy') in rats. The present study investigated the mechanisms mediating this interaction. EXPERIMENTAL APPROACH Adult male Sprague-Dawley rats were treated with caffeine (10 mg x kg(-1); i.p.) and MDMA (15 mg x kg(-1); i.p.) alone and in combination. Core body temperatures were monitored before and after drug administration. KEY RESULTS Central catecholamine depletion blocked MDMA-induced hyperthermia and its exacerbation by caffeine. Caffeine provoked a hyperthermic response when the catecholamine releaser d-amphetamine (1 mg x kg(-1)) was combined with the 5-HT releaser D-fenfluramine (5 mg x kg(-1)) or the non-selective dopamine receptor agonist apomorphine (1 mg x kg(-1)) was combined with the 5-HT(2) receptor agonist DOI (2 mg x kg(-1)) but not following either agents alone. Pretreatment with the dopamine D(1) receptor antagonist Schering (SCH) 23390 (1 mg x kg(-1)), the 5-HT(2) receptor antagonist ketanserin (5 mg x kg(-1)) or alpha(1)-adreno- receptor antagonist prazosin (0.2 mg x kg(-1)) blocked MDMA-induced hyperthermia and its exacerbation by caffeine. Co-administration of a combination of MDMA with the PDE-4 inhibitor rolipram (0.025 mg x kg(-1)) and the adenosine A(1/2) receptor antagonist 9-chloro-2-(2-furanyl)-[1,2,4]triazolo[1,5-C]quinazolin-5-amine 15943 (10 mg x kg(-1)) or the A(2A) receptor antagonist SCH 58261 (2 mg x kg(-1)) but not the A(1) receptor antagonist DPCPX (10 mg x kg(-1)) exacerbated MDMA-induced hyperthermia. CONCLUSIONS AND IMPLICATIONS A mechanism comprising 5-HT and catecholamines is proposed to mediate MDMA-induced hyperthermia. A combination of adenosine A(2A) receptor antagonism and PDE inhibition can account for the exacerbation of MDMA-induced hyperthermia by caffeine.
Collapse
Affiliation(s)
- N Vanattou-Saïfoudine
- Neuropsychopharmacology Research Group, School of Pharmacy and Pharmaceutical Sciences & Trinity College Institute of Neuroscience, Trinity College Dublin, Dublin, Ireland
| | | | | |
Collapse
|
8
|
Affiliation(s)
- Diane E May
- Department of Psychiatry, University of Nebraska Medical Center, Omaha, Nebraska 68198-5581, USA
| | | |
Collapse
|
9
|
Patrick KS, Straughn AB, Perkins JS, González MA. Evolution of stimulants to treat ADHD: transdermal methylphenidate. Hum Psychopharmacol 2009; 24:1-17. [PMID: 19051222 PMCID: PMC2629554 DOI: 10.1002/hup.992] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/07/2022]
Abstract
OBJECTIVE The following comprehensive review describes the evolution of stimulant drug formulations used in the treatment of attention-deficit/hyperactivity disorder (ADHD). Emphasis is placed on the basic and clinical pharmacology of the dl-methylphenidate (MPH) transdermal system (MTS). METHODS The pharmacokinetic and pharmacodynamic literature pertaining to MPH and amphetamine enantiomers was reviewed in the context of ADHD therapy and MTS as a treatment option. RESULTS MTS incorporates MPH into an adhesive monolithic matrix, using the free base form of the drug to facilitate transdermal absorption. MTS technology minimizes contact dermatitis by eliminating to need for percutaneous penetration enhancers. After a lag time of approximately 2 h, plasma concentrations of the therapeutic d-MPH isomer become detectable, then continuously rise over the course of the recommended 9 h wear time. Concentrations of l-MPH typically attain 40-50% that of d-MPH (vs. 1-2% following oral MPH). Unauthorized MTS removal poses some misuse liability and over 50% of MTS drug content remains in the discarded system. CONCLUSIONS While liquid or chewable MPH formulations overcome potential swallowing difficulties, as do sprinkled once-daily extended-release (ER) MPH products, only MTS addresses swallowing difficulties while also offering a flexible individualized MPH exposure time in a once-daily MPH regimen.
Collapse
Affiliation(s)
- Kennerly S. Patrick
- Department of Pharmaceutical and Biomedical Sciences, Medical University of South Carolina, Charleston, South Carolina, USA,Correspondence to: K. S. Patrick, 280 Calhoun St., QF221A, Medical University of South Carolina, Charleston, SC 29425−0742, USA. Tel: (843) 792−8429. Fax: (843) 792−1617. E-mail:
| | - Arthur B. Straughn
- Department of Pharmaceutical Sciences, University of Tennessee, Memphis, Tennessee, USA
| | - Jeb S. Perkins
- Department of Pharmaceutical and Biomedical Sciences, Medical University of South Carolina, Charleston, South Carolina, USA
| | | |
Collapse
|
10
|
Richardson NR, Gratton A. Changes in nucleus accumbens dopamine transmission associated with fixed- and variable-time schedule-induced feeding. Eur J Neurosci 2008; 27:2714-23. [DOI: 10.1111/j.1460-9568.2008.06236.x] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
|
11
|
Joyce BM, Glaser PEA, Gerhardt GA. Adderall produces increased striatal dopamine release and a prolonged time course compared to amphetamine isomers. Psychopharmacology (Berl) 2007; 191:669-77. [PMID: 17031708 DOI: 10.1007/s00213-006-0550-9] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/26/2006] [Accepted: 08/01/2006] [Indexed: 11/24/2022]
Abstract
RATIONALE Adderall is currently used for the treatment of Attention-Deficit Hyperactivity Disorder (ADHD) and is composed of a novel mixture of approximately 24% L-amphetamine and 76% D-amphetamine salts. There are, however, no investigations of the pharmacological effects of this combination in vivo. OBJECTIVES The technique of high-speed chronoamperometry using Nafion-coated single carbon-fiber microelectrodes was used to study amphetamine-evoked dopamine (DA) release produced by Adderall, D-amphetamine, or D,L-amphetamine in the striatum of anesthetized male Fischer 344 (F344) rats. The amphetamine solutions were locally applied from micropipettes by pressure ejection. RESULTS Local applications of Adderall resulted in significantly greater DA release signal amplitudes with prolonged time course of dopamine release and re-uptake as compared to D-amphetamine and D,L-amphetamine. CONCLUSIONS These data support the hypothesis that the combination of amphetamine enantiomers and salts in Adderall has effects on DA release, which result in increased and prolonged DA release, compared to D- and D,L-amphetamine.
Collapse
Affiliation(s)
- B Matthew Joyce
- Department of Anatomy and Neurobiology, Center for Sensor Technology, The Morris K. Udall Parkinson's Disease Research Center of Excellence, University of Kentucky Chandler Medical Center, Lexington, KY, 40536-0098, USA
| | | | | |
Collapse
|
12
|
Thomas TC, Kruzich PJ, Joyce BM, Gash C, Suchland K, Surgener SP, Rutherford EC, Grandy DK, Gerhardt GA, Glaser PE. Dopamine D4 receptor knockout mice exhibit neurochemical changes consistent with decreased dopamine release. J Neurosci Methods 2007; 166:306-14. [PMID: 17449106 PMCID: PMC2699616 DOI: 10.1016/j.jneumeth.2007.03.009] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2006] [Revised: 03/08/2007] [Accepted: 03/14/2007] [Indexed: 11/17/2022]
Abstract
Dopamine D4 receptor (D4R) knockout mice (D4R-/-) provided for unique neurochemical studies designed to understand D4R contributions to dopamine (DA) regulation. In this study, post-mortem brain tissue content of DA did not differ between D4R+/+ and D4R-/- mice in the striatum (Str) or nucleus accumbens core (NAc). However, there was a significant decrease (82%) in the content of 3,4-dihydoxyphenylacetic acid (DOPAC), a major metabolite of DA, in the NAc of D4R-/- mice. Microdialysis studies performed in a region of brain spanning of the dorsal Str and NAc showed lower baseline levels of DA and a significant reduction in KCl-evoked overflow of DA in the D4R-/- mice. Baseline extracellular levels of DOPAC and homovanillic acid were also significantly lower in the D4R-/- mice. In vivo chronoamperometric recordings of KCl-evoked release of DA also showed decreased release of DA in the Str and NAc of the D4R-/- mice. These studies demonstrate a role of D4Rs in presynaptic DA regulation and support the hypothesis that alterations in D4Rs may lead to diminished DA function.
Collapse
Affiliation(s)
- Theresa Currier Thomas
- Department of Anatomy & Neurobiology, University of Kentucky Chandler Medical Center, Lexington, KY 40536, USA
- Center for Sensor Technology, University of Kentucky Chandler Medical Center, Lexington, KY 40536, USA
- University of Kentucky Morris K. Udall Parkinson’s Disease Research Center of Excellence, University of Kentucky Chandler Medical Center, Lexington, KY 40536, USA
| | - Paul J. Kruzich
- Departments of Physiology & Pharmacology, Oregon Health and Science University; Portland, OR 97239, USA
- Department of Physiology, Medical College of Georgia, Augusta, GA 30912, USA
| | - B. Matthew Joyce
- Department of Anatomy & Neurobiology, University of Kentucky Chandler Medical Center, Lexington, KY 40536, USA
- Center for Sensor Technology, University of Kentucky Chandler Medical Center, Lexington, KY 40536, USA
- University of Kentucky Morris K. Udall Parkinson’s Disease Research Center of Excellence, University of Kentucky Chandler Medical Center, Lexington, KY 40536, USA
| | - C.R. Gash
- Department of Psychiatry, University of Kentucky Chandler Medical Center, Lexington, KY 40536, USA
| | - Katherine Suchland
- Departments of Physiology & Pharmacology, Oregon Health and Science University; Portland, OR 97239, USA
| | - Stewart P. Surgener
- Department of Anatomy & Neurobiology, University of Kentucky Chandler Medical Center, Lexington, KY 40536, USA
- Center for Sensor Technology, University of Kentucky Chandler Medical Center, Lexington, KY 40536, USA
- University of Kentucky Morris K. Udall Parkinson’s Disease Research Center of Excellence, University of Kentucky Chandler Medical Center, Lexington, KY 40536, USA
| | - Erin C. Rutherford
- Department of Anatomy & Neurobiology, University of Kentucky Chandler Medical Center, Lexington, KY 40536, USA
- Center for Sensor Technology, University of Kentucky Chandler Medical Center, Lexington, KY 40536, USA
- University of Kentucky Morris K. Udall Parkinson’s Disease Research Center of Excellence, University of Kentucky Chandler Medical Center, Lexington, KY 40536, USA
| | - David K. Grandy
- Departments of Physiology & Pharmacology, Oregon Health and Science University; Portland, OR 97239, USA
| | - Greg A. Gerhardt
- Department of Anatomy & Neurobiology, University of Kentucky Chandler Medical Center, Lexington, KY 40536, USA
- Department of Psychiatry, University of Kentucky Chandler Medical Center, Lexington, KY 40536, USA
- Department of Neurology, University of Kentucky Chandler Medical Center, Lexington, KY 40536, USA
- Center for Sensor Technology, University of Kentucky Chandler Medical Center, Lexington, KY 40536, USA
- University of Kentucky Morris K. Udall Parkinson’s Disease Research Center of Excellence, University of Kentucky Chandler Medical Center, Lexington, KY 40536, USA
| | - Paul E.A. Glaser
- Department of Anatomy & Neurobiology, University of Kentucky Chandler Medical Center, Lexington, KY 40536, USA
- Department of Pediatrics, University of Kentucky Chandler Medical Center, Lexington, KY 40536, USA
- Department of Psychiatry, University of Kentucky Chandler Medical Center, Lexington, KY 40536, USA
- Center for Sensor Technology, University of Kentucky Chandler Medical Center, Lexington, KY 40536, USA
- University of Kentucky Morris K. Udall Parkinson’s Disease Research Center of Excellence, University of Kentucky Chandler Medical Center, Lexington, KY 40536, USA
- Address correspondence to: Paul E.A. Glaser M.D., Ph.D., University of Kentucky, 306 Whitney-Hendrickson Facility, Lexington, KY 40536, Tel.: (859) 323-4531, Fax: (859) 257-5310,
| |
Collapse
|
13
|
Doherty M, Gratton A. Differential involvement of ventral tegmental GABA(A) and GABA(B) receptors in the regulation of the nucleus accumbens dopamine response to stress. Brain Res 2007; 1150:62-8. [PMID: 17395162 DOI: 10.1016/j.brainres.2007.02.081] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2006] [Revised: 02/20/2007] [Accepted: 02/23/2007] [Indexed: 10/23/2022]
Abstract
Evidence indicates that dopamine (DA) transmission in nucleus accumbens (NAcc) is modulated by glutamate (GLUT) projections from medial prefrontal cortex (PFC) to NAcc and the ventral tegmental area (VTA). Local NMDA receptor blockade in NAcc has previously been shown to enhance the DA stress response in this region as well as in the VTA. This raises the possibility that the NAcc DA stress response is regulated by GLUT acting at NMDA receptors located on NAcc GABA output neurons that project to the VTA where GABA is known to regulate DA cell activity. Thus, in the present study, we used voltammetry to examine the effects of intra-VTA administration of GABA(A) and GABA(B) agonists and antagonists on restraint stress-induced increases in NAcc DA. The results show that local VTA GABA(B) receptor activation with baclofen (0.01, 0.1 and 1.0 nmol) dose-dependently inhibited the NAcc DA stress response whereas GABA(B) receptor blockade with phaclofen had the opposite effect, resulting in a dose-dependent potentiation of the stress response. A similar potentiation of the NAcc DA stress response was observed following VTA GABA(A) receptor blockade with bicuculline, but only at the highest dose (1.0 nmol). Interestingly, intra-VTA injection of the GABA(A) receptor agonist, muscimol, at the lowest dose (0.01 nmol) but not at the higher doses (0.1 or 1.0 nmol) also potentiated the NAcc DA stress response, suggesting an action mediated primarily at GABA(A) receptors located on non-DA neurons. These results indicate that the NAcc DA stress response is regulated by GABA afferents to VTA DA cells and that this action is differentially mediated by GABA(A) and GABA(B) receptors. The data suggest that the relevant GABA(B) receptors are located on DA neurons whereas the GABA(A) receptors are located on GABA interneurons and perhaps also on DA cells. The present findings are also consistent with the idea that the corticofugal GLUT input to NAcc indirectly regulates stress-induced DA release in this region through the GABA feedback pathway to VTA.
Collapse
Affiliation(s)
- Michael Doherty
- Douglas Hospital Research Center, Department of Psychiatry, McGill University, 6875 LaSalle Blvd, Montréal Verdun, Québec, Canada, H4H 1R3
| | | |
Collapse
|
14
|
Volz TJ, Hanson GR, Fleckenstein AE. The role of the plasmalemmal dopamine and vesicular monoamine transporters in methamphetamine‐induced dopaminergic deficits. J Neurochem 2006; 101:883-8. [PMID: 17250674 DOI: 10.1111/j.1471-4159.2006.04419.x] [Citation(s) in RCA: 62] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Amphetamine (AMPH) and methamphetamine (METH) are members of a collection of phenethylamine psychostimulants that are commonly referred to collectively as "amphetamines." Amphetamines exert their effects, in part, by affecting neuronal dopamine transport. This review thus focuses on the effects of AMPH and METH on the plasmalemmal dopamine transporter and the vesicular monoamine transporter-2 in animal models with a particular emphasis on how these effects, which may vary for the different stereoisomers, contribute to persistent dopaminergic deficits.
Collapse
Affiliation(s)
- Trent J Volz
- Department of Pharmacology and Toxicology, University of Utah, Salt Lake City, UT 84112, USA
| | | | | |
Collapse
|
15
|
Abstract
Attention-deficit hyperactivity disorder (ADHD) in girls is a topic of growing research and clinical interest. For many years, girls with ADHD have been ignored and overshadowed by hyperkinetic and impulsive boys, but they are now attracting interest in an effort to understand the similarities and differences in the prevalence, symptoms, familial risk, comorbidities and treatment of ADHD in the two sexes. A review of past and current literature finds that the symptoms of ADHD are not sex specific, but that identification of girls with ADHD is hampered by parental and teacher bias, and confusion. Girls are more likely to be inattentive without being hyperactive or impulsive, compared with boys. Girls and boys share the same familial risk patterns, as well as similar, although not identical, comorbidity or impairment patterns. The risk of non-treatment is as great in girls as it is in boys; up to 70-80% of identified children will have persistent symptoms and impairment that extends into adolescence and adulthood. Treatment modalities are equally effective in girls and boys. Stimulants, non-stimulants and behavioural modalities are the mainstays of effective treatment.
Collapse
Affiliation(s)
- Jud Staller
- SUNY Upstate Medical University, Syracuse, New York 13210, USA.
| | | |
Collapse
|