1
|
Kharas N, Yang PB, Robles T, Sanchez A, Dafny N. Sex differences in the intensity of cross-sensitization between methylphenidate and amphetamine in adolescent rats. Physiol Behav 2019; 202:77-86. [PMID: 30653974 DOI: 10.1016/j.physbeh.2018.12.021] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2018] [Revised: 12/17/2018] [Accepted: 12/18/2018] [Indexed: 10/27/2022]
Abstract
Chronic use of psychostimulants such as methylphenidate (MPD) and amphetamine (Amph) leads to abuse and dependence. Cross-sensitization occurs when exposure to a drug causes a significant intensified response to a different drug as compared to the effect of the drug in subjects with no previous exposure. Cross-sensitization is used as an experimental correlate for a drug's potential to elicit dependence. The present study uses male and female adolescent rats to examine whether cross-sensitization occurs with MPD, a drug not traditionally considered to elicit dependence, and Amph, a drug considered to elicit dependence. The results showed that there is cross-sensitization with MPD to Amph in adolescent rats and that there is a significant difference in male and female responses. Cross-sensitization between MPD and Amph was observed in a linear dose dependent manner in males and in an inverted U-shape pattern in females. Males treated with the highest dose of 10.0 mg/kg MPD and females treated with the mid-dose of 2.5 mg/kg MPD showed the most robust cross-sensitization. Overall, adolescent female rodents had a greater intensity of response to MPD, Amph, and cross-sensitization between MPD and Amph. This study shows that there are significant sex differences in psychostimulant cross-sensitization in adolescence, indicating the maturity of the gonadal system is not the predominant reason for differences between male and female responses to psychostimulant drugs.
Collapse
Affiliation(s)
- Natasha Kharas
- McGovern Medical School, University of Texas Health Science Center, Houston, TX 77006, United States
| | - Pamela B Yang
- Department of Biological Sciences, Chapman University, Orange, CA 92866, United States
| | - Tiffany Robles
- McGovern Medical School, University of Texas Health Science Center, Houston, TX 77006, United States
| | - Ashley Sanchez
- McGovern Medical School, University of Texas Health Science Center, Houston, TX 77006, United States
| | - Nachum Dafny
- McGovern Medical School, University of Texas Health Science Center, Houston, TX 77006, United States.
| |
Collapse
|
2
|
Intrauterine Growth Restriction Modifies the Accumbal Dopaminergic Response to Palatable Food Intake. Neuroscience 2018; 400:184-195. [PMID: 30599270 DOI: 10.1016/j.neuroscience.2018.12.036] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2018] [Revised: 12/19/2018] [Accepted: 12/20/2018] [Indexed: 11/23/2022]
Abstract
Intrauterine growth restriction (IUGR) associates with increased preference for palatable foods and altered insulin sensitivity. Insulin modulates the central dopaminergic response and changes behavioral responses to reward. We measured the release of dopamine in the accumbens during palatable food intake in IUGR rats both at baseline and in response to insulin. From pregnancy day 10 until birth, gestating Sprague-Dawley rats received either an ad libitum (Control), or a 50% food restricted (FR) diet. In adulthood, palatable food consumption and feeding behavior entropy was assessed using an electronic food intake monitor (BioDAQ®), and dopamine response to palatable food was measured by chronoamperometry recordings in the nucleus accumbens (NAcc). FR rats eat more palatable foods during the dark phase, and their eating pattern has a higher entropy compared to control rats. There was a delayed dopamine release in the FR group in response to palatable food and insulin administration reverted this delayed effect. Western blot showed a decrease in suppressor of cytokine signaling 3 protein (SOCS3) in the ventral tegmental area (VTA) and an increase in the ratio of phospho-tyrosine hydroxylase to tyrosine hydroxylase (pTH/TH) in the NAcc of FR rats. Administration of insulin also abolished this latter effect in FR rats. FR rats showed metabolic alterations and a delay in the dopaminergic response to palatable foods. This could explain the increased palatable food intake and behavioral entropy found in FR rats. IUGR may lead to binge eating, obesity and its metabolic consequences by modifying the central dopaminergic response to sweet food.
Collapse
|
3
|
Faraone SV. The pharmacology of amphetamine and methylphenidate: Relevance to the neurobiology of attention-deficit/hyperactivity disorder and other psychiatric comorbidities. Neurosci Biobehav Rev 2018; 87:255-270. [PMID: 29428394 DOI: 10.1016/j.neubiorev.2018.02.001] [Citation(s) in RCA: 366] [Impact Index Per Article: 52.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2017] [Revised: 01/25/2018] [Accepted: 02/05/2018] [Indexed: 12/20/2022]
Abstract
Psychostimulants, including amphetamines and methylphenidate, are first-line pharmacotherapies for individuals with attention-deficit/hyperactivity disorder (ADHD). This review aims to educate physicians regarding differences in pharmacology and mechanisms of action between amphetamine and methylphenidate, thus enhancing physician understanding of psychostimulants and their use in managing individuals with ADHD who may have comorbid psychiatric conditions. A systematic literature review of PubMed was conducted in April 2017, focusing on cellular- and brain system-level effects of amphetamine and methylphenidate. The primary pharmacologic effect of both amphetamine and methylphenidate is to increase central dopamine and norepinephrine activity, which impacts executive and attentional function. Amphetamine actions include dopamine and norepinephrine transporter inhibition, vesicular monoamine transporter 2 (VMAT-2) inhibition, and monoamine oxidase activity inhibition. Methylphenidate actions include dopamine and norepinephrine transporter inhibition, agonist activity at the serotonin type 1A receptor, and redistribution of the VMAT-2. There is also evidence for interactions with glutamate and opioid systems. Clinical implications of these actions in individuals with ADHD with comorbid depression, anxiety, substance use disorder, and sleep disturbances are discussed.
Collapse
Affiliation(s)
- Stephen V Faraone
- Departments of Psychiatry and of Neuroscience and Physiology, State University of New York (SUNY) Upstate Medical University, Syracuse, NY, United States; K.G. Jebsen Centre for Research on Neuropsychiatric Disorders, University of Bergen, Bergen, Norway.
| |
Collapse
|
4
|
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
|
5
|
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
|
6
|
Shanks RA, Ross JM, Doyle HH, Helton AK, Picou BN, Schulz J, Tavares C, Bryant S, Dawson BL, Lloyd SA. Adolescent exposure to cocaine, amphetamine, and methylphenidate cross-sensitizes adults to methamphetamine with drug- and sex-specific effects. Behav Brain Res 2014; 281:116-24. [PMID: 25496784 DOI: 10.1016/j.bbr.2014.12.002] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2014] [Revised: 10/31/2014] [Accepted: 12/01/2014] [Indexed: 01/11/2023]
Abstract
The increasing availability, over-prescription, and misuse and abuse of ADHD psychostimulant medications in adolescent populations necessitates studies investigating the long-term effects of these drugs persisting into adulthood. Male and female C57Bl/6J mice were exposed to amphetamine (AMPH) (1.0 and 10 mg/kg), methylphenidate (MPD) (1.0 and 10 mg/kg), or cocaine (COC) (5.0 mg/kg) from postnatal day 22 to 31, which represents an early adolescent period. After an extended period of drug abstinence, adult mice were challenged with a subacute methamphetamine (METH) dose (0.5 mg/kg), to test the long-term effects of adolescent drug exposures on behavioral cross-sensitization using an open field chamber. There were no sex- or dose-specific effects on motor activity in adolescent, saline-treated controls. However, AMPH, MPD, and COC adolescent exposures induced cross-sensitization to a subacute METH dose in adulthood, which is a hallmark of addiction and a marker of long-lasting plastic changes in the brain. Of additional clinical importance, AMPH-exposed male mice demonstrated increased cross-sensitization to METH in contrast to the female-specific response observed in MPD-treated animals. There were no sex-specific effects after adolescent COC exposures. This study demonstrates differential drug, dose, and sex-specific alterations induced by early adolescent psychostimulant exposure, which leads to behavioral alterations that persist into adulthood.
Collapse
Affiliation(s)
- Ryan A Shanks
- Department of Biology, University of North Georgia, Dahlonega, GA, USA.
| | - Jordan M Ross
- Department of Psychological Science, University of North Georgia, Dahlonega, GA, USA.
| | - Hillary H Doyle
- Department of Psychological Science, University of North Georgia, Dahlonega, GA, USA.
| | - Amanda K Helton
- Department of Biology, University of North Georgia, Dahlonega, GA, USA.
| | - Brittany N Picou
- Department of Psychological Science, University of North Georgia, Dahlonega, GA, USA.
| | - Jordyn Schulz
- Department of Psychological Science, University of North Georgia, Dahlonega, GA, USA.
| | - Chris Tavares
- Department of Psychological Science, University of North Georgia, Dahlonega, GA, USA.
| | - Sarah Bryant
- Department of Biology, University of North Georgia, Dahlonega, GA, USA.
| | - Bryan L Dawson
- Department of Psychological Science, University of North Georgia, Dahlonega, GA, USA.
| | - Steven A Lloyd
- Department of Psychological Science, University of North Georgia, Dahlonega, GA, USA.
| |
Collapse
|
7
|
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
|
8
|
Daberkow DP, Brown HD, Bunner KD, Kraniotis SA, Doellman MA, Ragozzino ME, Garris PA, Roitman MF. Amphetamine paradoxically augments exocytotic dopamine release and phasic dopamine signals. J Neurosci 2013; 33:452-63. [PMID: 23303926 PMCID: PMC3711765 DOI: 10.1523/jneurosci.2136-12.2013] [Citation(s) in RCA: 120] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2012] [Revised: 10/29/2012] [Accepted: 11/08/2012] [Indexed: 11/21/2022] Open
Abstract
Drugs of abuse hijack brain-reward circuitry during the addiction process by augmenting action potential-dependent phasic dopamine release events associated with learning and goal-directed behavior. One prominent exception to this notion would appear to be amphetamine (AMPH) and related analogs, which are proposed instead to disrupt normal patterns of dopamine neurotransmission by depleting vesicular stores and promoting nonexocytotic dopamine efflux via reverse transport. This mechanism of AMPH action, though, is inconsistent with its therapeutic effects and addictive properties, which are thought to be reliant on phasic dopamine signaling. Here we used fast-scan cyclic voltammetry in freely moving rats to interrogate principal neurochemical responses to AMPH in the striatum and relate these changes to behavior. First, we showed that AMPH dose-dependently enhanced evoked dopamine responses to phasic-like current pulse trains for up to 2 h. Modeling the data revealed that AMPH inhibited dopamine uptake but also unexpectedly potentiated vesicular dopamine release. Second, we found that AMPH increased the amplitude, duration, and frequency of spontaneous dopamine transients, the naturally occurring, nonelectrically evoked, phasic increases in extracellular dopamine. Finally, using an operant sugar reward paradigm, we showed that low-dose AMPH augmented dopamine transients elicited by sugar-predictive cues. However, operant behavior failed at high-dose AMPH, which was due to phasic dopamine hyperactivity and the decoupling of dopamine transients from the reward predictive cue. These findings identify upregulation of exocytotic dopamine release as a key AMPH action in behaving animals and support a unified mechanism of abused drugs to activate phasic dopamine signaling.
Collapse
Affiliation(s)
- D P Daberkow
- School of Biological Sciences, Cell Biology, Physiology and Development Section, Illinois State University, Normal, Illinois 61790, USA
| | | | | | | | | | | | | | | |
Collapse
|
9
|
Babcock T, Dirks B, Adeyi B, Scheckner B. Efficacy of lisdexamfetamine dimesylate in adults with attention-deficit/hyperactivity disorder previously treated with amphetamines: analyses from a randomized, double-blind, multicenter, placebo-controlled titration study. BMC Pharmacol Toxicol 2012; 13:18. [PMID: 23254273 PMCID: PMC3554536 DOI: 10.1186/2050-6511-13-18] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2012] [Accepted: 12/10/2012] [Indexed: 11/21/2022] Open
Abstract
Background To examine the efficacy of lisdexamfetamine dimesylate (LDX) in adults with attention-deficit/hyperactivity disorder (ADHD) who remained symptomatic (ADHD Rating Scale IV [ADHD-RS-IV] total score >18) on amphetamine (AMPH) therapy (mixed AMPH salts and/or d-AMPH formulations) prior to enrollment in a 4-week placebo-controlled LDX trial vs the overall study population. In these post hoc analyses from a multicenter, randomized, double-blind, forced-dose titration study, clinical efficacy of LDX (30-70 mg/d) in adults with ADHD receiving AMPH treatment at screening vs the overall study population was evaluated. ADHD symptoms were assessed using the ADHD-RS-IV with adult prompts at screening, baseline (after prior treatment washout), and endpoint. Safety assessments included treatment-emergent adverse events (TEAEs), vital signs, laboratory findings, and electrocardiogram. Results Of 414 participants (62, placebo; 352, LDX) included in the overall study population, 41 were receiving AMPH therapy at screening (2, placebo; 39, LDX); mean AMPH dose was 35.0 and 34.1 mg/d for participants in placebo and all LDX groups, respectively. Of the 41 participants, 36 remained symptomatic (ADHD-RS-IV >18) at screening despite receiving AMPH. For the 36 participants in the placebo (n = 2) and LDX (n = 34) groups, respectively, at endpoint, mean change from screening ADHD-RS-IV total scores were -5.5 and -14.8 and from baseline scores were -13.5 and -17.8. For the overall study population, endpoint mean change from baseline ADHD-RS-IV total scores were -7.8 for placebo and -17.5 for LDX. In the prior AMPH subgroup, 2/2 (100.0%) in the placebo group and 22/39 (56.4%) participants in the LDX (all doses) group reported any TEAE. Events that occurred in ≥5% for LDX were dry mouth (5/39; 12.8%), headache (5/39; 12.8%), fatigue (3/39; 7.7%), insomnia (3/39; 7.7%), decreased appetite (2/39; 5.1%), and nausea (2/39; 5.1%). None of these events occurred in the 2 placebo patients with prior AMPH use. Conclusion In these post hoc analyses, adults with significant baseline ADHD symptoms despite adequate AMPH treatment dose showed similar improvements in ADHD symptoms with LDX treatment as the overall study population. Prospective studies are needed to confirm these findings. The safety profile of LDX in the overall study population was consistent with long-acting psychostimulant use. Trial registry Study to Assess the Safety and Efficacy of NRP104 in Adults With Attention-Deficit Hyperactivity Disorder (ADHD). Clinicaltrials.gov Identifier: NCT00334880
Collapse
Affiliation(s)
- Thomas Babcock
- Shire Development LLC, 725 Chesterbrook Blvd, Wayne, PA, 19087, USA.
| | | | | | | |
Collapse
|
10
|
Ford JB, Albertson TE, Owen KP, Sutter ME, McKinney WB. Acute, sustained chorea in children after supratherapeutic dosing of amphetamine-derived medications. Pediatr Neurol 2012; 47:216-8. [PMID: 22883290 DOI: 10.1016/j.pediatrneurol.2012.05.013] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/27/2012] [Accepted: 05/21/2012] [Indexed: 11/17/2022]
Abstract
Amphetamine-derived medications are being prescribed with increasing frequency to younger pediatric patients to treat attention deficit hyperactivity disorder. Although choreiform movements were reported in adults with amphetamine abuse and in those under therapeutic treatment for attention deficit hyperactivity disorder, previous literature concerning the pediatric population is spare. We describe two children who developed chorea after ingesting amphetamine-derived medications prescribed to treat attention deficit hyperactivity disorder. Patient 1, a 10-year-old boy, accidently received an extra dose of lisdexamfetamine dimesylate the night before the onset of acute chorea involving his arms, legs, and trunk. Patient 2, an 8-month-old boy, accidentally ingested his stepbrother's mixed amphetamine salts (Adderall XR) and developed acute chorea. Benzodiazepines, diphenhydramine, benztropine, and opioids did not suppress the chorea in either case. The 10-year-old received haloperidol, which significantly improved his abnormal findings, and he returned to baseline in approximately 48 hours. The 8-month-old was observed in the pediatric intensive care unit, and his signs resolved by 72 hours. Our cases demonstrate that choreiform movements of sustained duration can occur in children with acute supratherapeutic ingestions of amphetamine-derived medications.
Collapse
Affiliation(s)
- Jonathan B Ford
- Department of Emergency Medicine, School of Medicine, University of California at Davis, Sacramento, CA 95817, USA.
| | | | | | | | | |
Collapse
|
11
|
Miller EM, Pomerleau F, Huettl P, Russell VA, Gerhardt GA, Glaser PEA. The spontaneously hypertensive and Wistar Kyoto rat models of ADHD exhibit sub-regional differences in dopamine release and uptake in the striatum and nucleus accumbens. Neuropharmacology 2012; 63:1327-34. [PMID: 22960443 DOI: 10.1016/j.neuropharm.2012.08.020] [Citation(s) in RCA: 44] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2012] [Revised: 07/31/2012] [Accepted: 08/21/2012] [Indexed: 10/27/2022]
Abstract
The most widely used animal model of attention-deficit/hyperactivity disorder (ADHD) is the spontaneously hypertensive rat (SHR/NCrl), which best represents the combined subtype (ADHD-C). Recent evidence has revealed that a progenitor strain, the Wistar Kyoto from Charles River Laboratories (WKY/NCrl), is useful as a model of the inattentive subtype (ADHD-PI) and the Wistar Kyoto from Harlan Laboratories (WKY/NHsd) and the Sprague Dawley (SD) have been suggested as controls. Dopamine (DA) dysfunction in the striatum (Str) and nucleus accumbens core (NAc) is thought to play a significant role in the pathophysiology of ADHD but data obtained with the SHR is equivocal. Using high-speed chronoamperometric recordings with carbon fiber microelectrodes, we found that the SHR/NCrl displayed decreased KCl-evoked DA release versus the WKY/NCrl model of ADHD-PI in the dorsal Str. The WKY/NCrl and the WKY/NHsd control did not differ from each other; however, the control SD released less DA than the WKY/NCrl model of ADHD-PI in the dorsal Str and less than the control WKY/NHsd in the intermediate Str. The SHR/NCrl had faster DA uptake in the ventral Str and NAc versus both control strains, while the WKY/NCrl model of ADHD-PI exhibited faster DA uptake in the NAc versus the SD control. These results suggest that increased surface expression of DA transporters may explain the more rapid uptake of DA in the Str and NAc of these rodent models of ADHD.
Collapse
Affiliation(s)
- Erin M Miller
- Department of Anatomy & Neurobiology, University of Kentucky College of Medicine, 138 Leader Avenue, Lexington, KY 40506, USA
| | | | | | | | | | | |
Collapse
|
12
|
Chronic low dose Adderall XR down-regulates cfos expression in infantile and prepubertal rat striatum and cortex. Neuroscience 2010; 169:1901-12. [PMID: 20600661 DOI: 10.1016/j.neuroscience.2010.06.029] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2010] [Revised: 05/31/2010] [Accepted: 06/12/2010] [Indexed: 11/23/2022]
Abstract
We previously reported that treatment of prepubertal male rats with low, injected or oral, doses of methylphenidate stimulated cfos, fosB and arc expression in many areas of the developing brain. In the present study our objective was to determine whether the widely prescribed psychostimulant Adderall XR (ADD) exerted similar effects in infantile and prepubertal rat brain. We report here, for the first time, that low threshold doses of oral ADD, an extended-release mixture of amphetamine salts, now routinely used for the treatment of Attention Deficit Hyperactivity Disorder (ADHD), also increased cfos expression in infantile (postnatal day 10; PD10) and prepubertal (PD24) rat brain. These threshold doses were correlated with blood levels of amphetamine determined by liquid chromatography-mass spectrometry. Moreover, we observed that chronic treatment with oral ADD (1.6 mg/kg; x 14 days) not only significantly down-regulated cfos expression following a final challenge dose of ADD in prepubertal (PD24) rat striatum and cortex, quantified in terms of FOS immunoreactivity (FOS-ir), but did so at a daily dose that was without effect with methylphenidate (MPH); that is a much higher oral dose of MPH (7.5 mg/kg; x 14 days) failed to induce down-regulation of cfos expression. Similar experiments in infantile rats (PD10), but using a threshold injected dose of ADD (1.25 mg/kg sc) also significantly reduced striatal and cingulate cortical FOS-ir. An additional finding in the prepubertal rats was that oral ADD-induced FOS-ir was observed in the cerebral cortex following doses lower than the threshold dose necessary to increase FOS-ir in the striatum. This was not the case in the PD10 rats. In conclusion, our efforts to calibrate biological responses, such as immediate early gene expression, to clinically relevant blood levels of stimulants confirmed that expression of cfos is very sensitive to repeated low doses of Adderall XR. It is now feasible to examine whether other genes are also affected in these young rats and if the changes we report are reversible. The implications of such studies should be relevant to the putative effects of psychostimulant treatment of very young children.
Collapse
|
13
|
Affiliation(s)
- Diane E May
- Department of Psychiatry, University of Nebraska Medical Center, Omaha, Nebraska 68198-5581, USA
| | | |
Collapse
|
14
|
Hascup ER, af Bjerkén S, Hascup KN, Pomerleau F, Huettl P, Strömberg I, Gerhardt GA. Histological studies of the effects of chronic implantation of ceramic-based microelectrode arrays and microdialysis probes in rat prefrontal cortex. Brain Res 2009; 1291:12-20. [PMID: 19577548 PMCID: PMC2980830 DOI: 10.1016/j.brainres.2009.06.084] [Citation(s) in RCA: 65] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2008] [Revised: 06/23/2009] [Accepted: 06/24/2009] [Indexed: 01/31/2023]
Abstract
Chronic implantation of neurotransmitter measuring devices is essential for awake, behavioral studies occurring over multiple days. Little is known regarding the effects of long term implantation on surrounding brain parenchyma and the resulting alterations in the functional properties of this tissue. We examined the extent of tissue damage produced by chronic implantation of either ceramic microelectrode arrays (MEAs) or microdialysis probes. Histological studies were carried out on fixed tissues using stains for neurons (cresyl violet), astrocytes (GFAP), microglia (Iba1), glutamatergic nerve fibers (VGLUT1), and the blood-brain barrier (SMI-71). Nissl staining showed pronounced tissue body loss with microdialysis implants compared to MEAs. The MEAs produced mild gliosis extending 50-100 microm from the tracks, with a significant change in the affected areas starting at 3 days. By contrast, the microdialysis probes produced gliosis extending 200-300 microm from the track, which was significant at 3 and 7 days. Markers for microglia and glutamatergic fibers supported that the MEAs produce minimal damage with significant changes occurring only at 3 and 7 days that return to control levels by 1 month. SMI-71 staining supported the integrity of the blood-brain barrier out to 1 week for both the microdialysis probes and the MEAs. This data support that the ceramic MEA's small size and biocompatibility are necessary to accurately measure neurotransmitter levels in the intact brain. The minimal invasiveness of the MEAs reduce tissue loss, allowing for long term (>6 month) electrochemical and electrophysiological monitoring of brain activity.
Collapse
Affiliation(s)
- Erin R Hascup
- Anatomy and Neurobiology, Morris K. Udall Parkinson's Disease Research Center of Excellence, Center for Microelectrode Technology, University of Kentucky, College of Medicine, Lexington, KY, 40536-0098 USA.
| | | | | | | | | | | | | |
Collapse
|
15
|
Sonuga-Barke EJS, Wiersema JR, van der Meere JJ, Roeyers H. Context-dependent Dynamic Processes in Attention Deficit/Hyperactivity Disorder: Differentiating Common and Unique Effects of State Regulation Deficits and Delay Aversion. Neuropsychol Rev 2009; 20:86-102. [DOI: 10.1007/s11065-009-9115-0] [Citation(s) in RCA: 88] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2009] [Accepted: 08/17/2009] [Indexed: 11/30/2022]
|
16
|
Goodwin JS, Larson GA, Swant J, Sen N, Javitch JA, Zahniser NR, De Felice LJ, Khoshbouei H. Amphetamine and methamphetamine differentially affect dopamine transporters in vitro and in vivo. J Biol Chem 2009; 284:2978-2989. [PMID: 19047053 PMCID: PMC2631950 DOI: 10.1074/jbc.m805298200] [Citation(s) in RCA: 160] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2008] [Revised: 11/25/2008] [Indexed: 12/12/2022] Open
Abstract
The psychostimulants d-amphetamine (AMPH) and methamphetamine (METH) release excess dopamine (DA) into the synaptic clefts of dopaminergic neurons. Abnormal DA release is thought to occur by reverse transport through the DA transporter (DAT), and it is believed to underlie the severe behavioral effects of these drugs. Here we compare structurally similar AMPH and METH on DAT function in a heterologous expression system and in an animal model. In the in vitro expression system, DAT-mediated whole-cell currents were greater for METH stimulation than for AMPH. At the same voltage and concentration, METH released five times more DA than AMPH and did so at physiological membrane potentials. At maximally effective concentrations, METH released twice as much [Ca(2+)](i) from internal stores compared with AMPH. [Ca(2+)](i) responses to both drugs were independent of membrane voltage but inhibited by DAT antagonists. Intact phosphorylation sites in the N-terminal domain of DAT were required for the AMPH- and METH-induced increase in [Ca(2+)](i) and for the enhanced effects of METH on [Ca(2+)](i) elevation. Calmodulin-dependent protein kinase II and protein kinase C inhibitors alone or in combination also blocked AMPH- or METH-induced Ca(2+) responses. Finally, in the rat nucleus accumbens, in vivo voltammetry showed that systemic application of METH inhibited DAT-mediated DA clearance more efficiently than AMPH, resulting in excess external DA. Together these data demonstrate that METH has a stronger effect on DAT-mediated cell physiology than AMPH, which may contribute to the euphoric and addictive properties of METH compared with AMPH.
Collapse
Affiliation(s)
- J Shawn Goodwin
- Department of Cancer Biology, Meharry Medical College, Nashville, Tennessee 37208
| | - Gaynor A Larson
- Department of Pharmacology, University of Colorado Denver, Aurora, Colorado 80045
| | - Jarod Swant
- Departments of Neurobiology and Neurotoxicology, Meharry Medical College, Nashville, Tennessee 37208
| | - Namita Sen
- Departments of Psychiatry and Pharmacology, Center for Molecular Recognition, Columbia University, New York, New York 10027-6902
| | - Jonathan A Javitch
- Departments of Psychiatry and Pharmacology, Center for Molecular Recognition, Columbia University, New York, New York 10027-6902
| | - Nancy R Zahniser
- Department of Pharmacology, University of Colorado Denver, Aurora, Colorado 80045
| | - Louis J De Felice
- Center for Molecular Neuroscience, Vanderbilt University, Nashville, Tennessee 37232; Department of Pharmacology, Vanderbilt University, Nashville, Tennessee 37232
| | - Habibeh Khoshbouei
- Departments of Neurobiology and Neurotoxicology, Meharry Medical College, Nashville, Tennessee 37208
| |
Collapse
|
17
|
Migheli R, Puggioni G, Dedola S, Rocchitta G, Calia G, Bazzu G, Esposito G, Lowry JP, O'Neill RD, Desole MS, Miele E, Serra PA. Novel integrated microdialysis-amperometric system for in vitro detection of dopamine secreted from PC12 cells: design, construction, and validation. Anal Biochem 2008; 380:323-30. [PMID: 18577368 DOI: 10.1016/j.ab.2008.05.050] [Citation(s) in RCA: 16] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2008] [Accepted: 05/30/2008] [Indexed: 10/22/2022]
Abstract
A novel dual channel in vitro apparatus, derived from a previously described design, has been coupled with dopamine (DA) microsensors for the flow-through detection of DA secreted from PC12 cells. The device, including two independent microdialysis capillaries, was loaded with a solution containing PC12 cells while a constant phosphate-buffered saline (PBS) medium perfusion was carried out using a dual channel miniaturized peristaltic pump. One capillary was perfused with normal PBS, whereas extracellular calcium was removed from extracellular fluid of the second capillary. After a first period of stabilization and DA baseline recording, KCl (75 mM) was added to the perfusion fluid of both capillaries. In this manner, a simultaneous "treatment-control" experimental design was performed to detect K+-evoked calcium-dependent DA secretion. For this purpose, self-referencing DA microsensors were developed, and procedures for making, testing, and calibrating them are described in detail. The electronic circuitry was derived from previously published schematics and optimized for dual sensor constant potential amperometry applications. The microdialysis system was tested and validated in vitro under different experimental conditions, and DA secretion was confirmed by high-performance liquid chromatography with electrochemical detection (HPLC-EC). PC12 cell viability was quantified before and after each experiment. The proposed apparatus serves as a reliable model for studying the effects of different drugs on DA secretion through the direct comparison of extracellular DA increase in treatment-control experiments performed on the same initial PC12 cell population.
Collapse
Affiliation(s)
- Rossana Migheli
- Department of Neuroscience, Medical School, University of Sassari, 07100 Sassari, Italy
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|