201
|
G protein-coupled receptor signaling in VTA dopaminergic neurons bidirectionally regulates the acute locomotor response to amphetamine but does not affect behavioral sensitization. Neuropharmacology 2019; 161:107663. [PMID: 31173760 DOI: 10.1016/j.neuropharm.2019.06.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2019] [Revised: 05/31/2019] [Accepted: 06/03/2019] [Indexed: 11/20/2022]
Abstract
Amphetamine (AMPH) acts as a substrate of the dopamine transporter (DAT) and causes a dramatic increase in extracellular dopamine (DA). Upon entering DA neurons, AMPH promotes DA efflux via DAT through a mechanism implicating depletion of DA from vesicular stores, activation of kinase pathways and transporter phosphorylation. Despite the role of intracellular signaling for AMPH action, it remains elusive how the response to AMPH is affected in vivo by metabotropic regulation via G protein coupled receptor signaling pathways. Here, we show by employment of Designer Receptors Exclusively Activated by Designer Drugs (DREADDs) that the acute hyperlocomotor response to AMPH is bidirectionally regulated by metabotropic input to VTA DA neurons with a markedly enhanced response upon activation of a Gs-coupled pathway and a markedly decreased locomotor response upon activation of a Gi-coupled pathway. The unique mechanism of action for AMPH was underlined by the absence of an effect of Gs activation on the locomotor response to the DAT inhibitor cocaine. Regardless of the profound effect on the acute AMPH response, repeated Gs activation or Gi activation did not affect development of AMPH sensitization. Furthermore, activation of a Gs-pathway or activation of a Gi-pathway in DA neurons did not have any effect on the AMPH-induced locomotor response in the AMPH sensitized mice. This suggests induction of alterations in DA neuronal functions that overrule the stimulatory or inhibitory effect of metabotropic input seen in drug-naïve mice. The data thereby underline the remarkable strength of maladaptive changes that occur upon intake of strong psychostimulants. This article is part of the issue entitled 'Special Issue on Neurotransmitter Transporters'.
Collapse
|
202
|
Vicario-Feliciano R, Wigton RL, White TP, Shergill SS, Averbeck BB. Dopamine manipulations drive changes in information sampling in healthy volunteers. J Psychopharmacol 2019; 33:670-677. [PMID: 30644326 PMCID: PMC6996051 DOI: 10.1177/0269881118822080] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
BACKGROUND Information sampling is the cognitive process of accumulating information before committing to a decision. Patients across numerous disorders show decreased information sampling relative to controls. AIMS Here, we used the Beads and the Best Choice Tasks to study the role of dopamine signaling in information sampling. METHODS Participants were given placebo, amisulpride, or ropinirole in each session, in a double-blind cross-over design. RESULTS We found that ropinirole (agonist) increased the number of beads drawn in the Beads Task specifically when participants faced a loss, and decreased the rank of the chosen option in the Best Choice Task. CONCLUSIONS These effects are likely driven by a combination of effects at presynaptic D2 receptors, which affect dopamine release, and post-synaptic D2 receptors. Increased D2 relative to D1 receptor activation in the striatum leads to increased sampling in the loss condition in the Beads Task. It also leads to choice of a poorer ranked option in the Best Choice Task. Decreased D2 relative to D1 receptor activation leads to decreased sampling in the Beads Task in the loss condition.
Collapse
Affiliation(s)
| | - Rebekah L Wigton
- Institute of Psychiatry, Psychology and Neuroscience, London, UK
| | - Thomas P White
- Institute of Psychiatry, Psychology and Neuroscience, London, UK,School of Psychology, University of Birmingham, Birmingham, UK
| | - Sukhi S Shergill
- Institute of Psychiatry, Psychology and Neuroscience, London, UK
| | - Bruno B Averbeck
- Laboratory of Neuropsychology, National Institutes of Health, MD, USA
| |
Collapse
|
203
|
Varenicline and GZ-793A differentially decrease methamphetamine self-administration under a multiple schedule of reinforcement in rats. Behav Pharmacol 2019; 29:87-97. [PMID: 28863003 DOI: 10.1097/fbp.0000000000000340] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
Methamphetamine is a potent psychostimulant with high abuse rates. Currently, there is no Food and Drug Administration-approved pharmacotherapy for methamphetamine addiction. Ideally, a pharmacotherapy should selectively decrease methamphetamine self-administration without affecting responding for other reinforcers. One way to test this is with the use of a multiple schedule of reinforcement, in which drug and food are available in alternating components within a session. The present study evaluated GZ-793A, a vesicular monoamine transporter-2 inhibitor, and varenicline, a partial agonist at α4β2 and full agonist at α7 nicotinic acetylcholine receptors, for their ability to decrease methamphetamine and food self-administration using a multiple schedule of reinforcement. Male Sprague-Dawley rats self-administered methamphetamine (0.03 mg/kg/intravenous infusion) and food pellets under a multiple schedule of reinforcement. GZ-793A or varenicline was administered before multiple schedule sessions. GZ-793A (5 and 20 mg/kg) significantly decreased methamphetamine intake compared with saline and did not alter food-maintained responding. In contrast, varenicline decreased methamphetamine intake less specifically across time. The results suggest that vesicular monoamine transporter-2 inhibition may be a viable pharmacological target for the treatment of methamphetamine-use disorders.
Collapse
|
204
|
Su M, Zhou J, Duan Z, Zhang J. Transcriptional analysis of renal dopamine-mediated Na + homeostasis response to environmental salinity stress in Scatophagus argus. BMC Genomics 2019; 20:418. [PMID: 31126236 PMCID: PMC6534869 DOI: 10.1186/s12864-019-5795-x] [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: 11/09/2018] [Accepted: 05/10/2019] [Indexed: 02/07/2023] Open
Abstract
Background To control the osmotic pressure in the body, physiological adjustments to salinity fluctuations require the fish to regulate body fluid homeostasis in relation to environmental change via osmoregulation. Previous studies related to osmoregulation were focused primarily on the gill; however, little is known about another organ involved in osmoregulation, the kidney. The salinity adaptation of marine fish involves complex physiological traits, metabolic pathways and molecular and gene networks in osmoregulatory organs. To further explore of the salinity adaptation of marine fish with regard to the role of the kidney, the euryhaline fish Scatophagus argus was employed in the present study. Renal expression profiles of S. argus at different salinity levels were characterized using RNA-sequencing, and an integrated approach of combining molecular tools with physiological and biochemical techniques was utilized to reveal renal osmoregulatory mechanisms in vivo and in vitro. Results S. argus renal transcriptomes from the hyposaline stress (0‰, freshwater [FW]), hypersaline stress (50‰, hypersaline water [HW]) and control groups (25‰) were compared to elucidate potential osmoregulatory mechanisms. In total, 19,012 and 36,253 differentially expressed genes (DEGs) were obtained from the FW and HW groups, respectively. Based on the functional classification of DEGs, the renal dopamine system-induced Na+ transport was demonstrated to play a fundamental role in osmoregulation. In addition, for the first time in fish, many candidate genes associated with the dopamine system were identified. Furthermore, changes in environmental salinity affected renal dopamine release/reuptake by regulating the expression of genes related to dopamine reuptake (dat and nkaα1), vesicular traffic-mediated dopamine release (pink1, lrrk2, ace and apn), DAT phosphorylation (CaMKIIα and pkcβ) and internalization (akt1). The associated transcriptional regulation ensured appropriate extracellular dopamine abundance in the S. argus kidney, and fluctuations in extracellular dopamine produced a direct influence on Na+/K+-ATPase (NKA) expression and activity, which is associated with Na+ homeostasis. Conclusions These transcriptomic data provided insight into the molecular basis of renal osmoregulation in S. argus. Significantly, the results of this study revealed the mechanism of renal dopamine system-induced Na+ transport is essential in fish osmoregulation. Electronic supplementary material The online version of this article (10.1186/s12864-019-5795-x) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Maoliang Su
- Shenzhen Key Laboratory of Marine Bioresource & Eco-Environmental Science, College of Life Sciences and Oceanography, Shenzhen University, Shenzhen, 518060, China.,Key Laboratory of Optoelectronic Devices and Systems of Ministry of Education and Guangdong Province, College of Optoelectronic Engineering, Shenzhen University, Shenzhen, 518060, China
| | - Jianan Zhou
- Key Laboratory of Exploration and Utilization of Aquatic Genetic Resources, Ministry of Education, College of Fisheries and Life Sciences, Shanghai Ocean University, Shanghai, 201306, China
| | - Zhengyu Duan
- Shenzhen Key Laboratory of Marine Bioresource & Eco-Environmental Science, College of Life Sciences and Oceanography, Shenzhen University, Shenzhen, 518060, China.,Key Laboratory of Exploration and Utilization of Aquatic Genetic Resources, Ministry of Education, College of Fisheries and Life Sciences, Shanghai Ocean University, Shanghai, 201306, China
| | - Junbin Zhang
- Shenzhen Key Laboratory of Marine Bioresource & Eco-Environmental Science, College of Life Sciences and Oceanography, Shenzhen University, Shenzhen, 518060, China. .,Key Laboratory of Optoelectronic Devices and Systems of Ministry of Education and Guangdong Province, College of Optoelectronic Engineering, Shenzhen University, Shenzhen, 518060, China.
| |
Collapse
|
205
|
Ferrucci M, Limanaqi F, Ryskalin L, Biagioni F, Busceti CL, Fornai F. The Effects of Amphetamine and Methamphetamine on the Release of Norepinephrine, Dopamine and Acetylcholine From the Brainstem Reticular Formation. Front Neuroanat 2019; 13:48. [PMID: 31133823 PMCID: PMC6524618 DOI: 10.3389/fnana.2019.00048] [Citation(s) in RCA: 48] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2018] [Accepted: 04/26/2019] [Indexed: 12/22/2022] Open
Abstract
Amphetamine (AMPH) and methamphetamine (METH) are widely abused psychostimulants, which produce a variety of psychomotor, autonomic and neurotoxic effects. The behavioral and neurotoxic effects of both compounds (from now on defined as AMPHs) stem from a fair molecular and anatomical specificity for catecholamine-containing neurons, which are placed in the brainstem reticular formation (RF). In fact, the structural cross-affinity joined with the presence of shared molecular targets between AMPHs and catecholamine provides the basis for a quite selective recruitment of brainstem catecholamine neurons following AMPHs administration. A great amount of investigations, commentary manuscripts and books reported a pivotal role of mesencephalic dopamine (DA)-containing neurons in producing behavioral and neurotoxic effects of AMPHs. Instead, the present review article focuses on catecholamine reticular neurons of the low brainstem. In fact, these nuclei add on DA mesencephalic cells to mediate the effects of AMPHs. Among these, we also include two pontine cholinergic nuclei. Finally, we discuss the conundrum of a mixed neuronal population, which extends from the pons to the periaqueductal gray (PAG). In this way, a number of reticular nuclei beyond classic DA mesencephalic cells are considered to extend the scenario underlying the neurobiology of AMPHs abuse. The mechanistic approach followed here to describe the action of AMPHs within the RF is rooted on the fine anatomy of this region of the brainstem. This is exemplified by a few medullary catecholamine neurons, which play a pivotal role compared with the bulk of peripheral sympathetic neurons in sustaining most of the cardiovascular effects induced by AMPHs.
Collapse
Affiliation(s)
- Michela Ferrucci
- Human Anatomy, Department of Translational Research and New Technologies in Medicine and Surgery, University of Pisa, Pisa, Italy
| | - Fiona Limanaqi
- Human Anatomy, Department of Translational Research and New Technologies in Medicine and Surgery, University of Pisa, Pisa, Italy
| | - Larisa Ryskalin
- Human Anatomy, Department of Translational Research and New Technologies in Medicine and Surgery, University of Pisa, Pisa, Italy
| | | | | | - Francesco Fornai
- Human Anatomy, Department of Translational Research and New Technologies in Medicine and Surgery, University of Pisa, Pisa, Italy
- IRCCS Neuromed, Pozzilli, Italy
| |
Collapse
|
206
|
|
207
|
Highfill CA, Baker BM, Stevens SD, Anholt RRH, Mackay TFC. Genetics of cocaine and methamphetamine consumption and preference in Drosophila melanogaster. PLoS Genet 2019; 15:e1007834. [PMID: 31107875 PMCID: PMC6527214 DOI: 10.1371/journal.pgen.1007834] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2018] [Accepted: 03/27/2019] [Indexed: 12/11/2022] Open
Abstract
Illicit use of psychostimulants, such as cocaine and methamphetamine, constitutes a significant public health problem. Whereas neural mechanisms that mediate the effects of these drugs are well-characterized, genetic factors that account for individual variation in susceptibility to substance abuse and addiction remain largely unknown. Drosophila melanogaster can serve as a translational model for studies on substance abuse, since flies have a dopamine transporter that can bind cocaine and methamphetamine, and exposure to these compounds elicits effects similar to those observed in people, suggesting conserved evolutionary mechanisms underlying drug responses. Here, we used the D. melanogaster Genetic Reference Panel to investigate the genetic basis for variation in psychostimulant drug consumption, to determine whether similar or distinct genetic networks underlie variation in consumption of cocaine and methamphetamine, and to assess the extent of sexual dimorphism and effect of genetic context on variation in voluntary drug consumption. Quantification of natural genetic variation in voluntary consumption, preference, and change in consumption and preference over time for cocaine and methamphetamine uncovered significant genetic variation for all traits, including sex-, exposure- and drug-specific genetic variation. Genome wide association analyses identified both shared and drug-specific candidate genes, which could be integrated in genetic interaction networks. We assessed the effects of ubiquitous RNA interference (RNAi) on consumption behaviors for 34 candidate genes: all affected at least one behavior. Finally, we utilized RNAi knockdown in the nervous system to implicate dopaminergic neurons and the mushroom bodies as part of the neural circuitry underlying experience-dependent development of drug preference.
Collapse
Affiliation(s)
- Chad A. Highfill
- Department of Biological Sciences, W. M. Keck Center for Behavioral Biology, and Program in Genetics, North Carolina State University, Raleigh, NC, United States of America
| | - Brandon M. Baker
- Department of Biological Sciences, W. M. Keck Center for Behavioral Biology, and Program in Genetics, North Carolina State University, Raleigh, NC, United States of America
| | - Stephenie D. Stevens
- Department of Biological Sciences, W. M. Keck Center for Behavioral Biology, and Program in Genetics, North Carolina State University, Raleigh, NC, United States of America
| | - Robert R. H. Anholt
- Department of Biological Sciences, W. M. Keck Center for Behavioral Biology, and Program in Genetics, North Carolina State University, Raleigh, NC, United States of America
| | - Trudy F. C. Mackay
- Department of Biological Sciences, W. M. Keck Center for Behavioral Biology, and Program in Genetics, North Carolina State University, Raleigh, NC, United States of America
| |
Collapse
|
208
|
Effects of stimulant drug use on the dopaminergic system: A systematic review and meta-analysis of in vivo neuroimaging studies. Eur Psychiatry 2019; 59:15-24. [PMID: 30981746 DOI: 10.1016/j.eurpsy.2019.03.003] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/18/2019] [Revised: 03/13/2019] [Accepted: 03/17/2019] [Indexed: 12/20/2022] Open
Abstract
BACKGROUND Stimulant drugs can cause persistent changes in the brain. Imaging studies show that these changes are most apparent in dopamine transporter (DAT) or receptor availability within the striatum. METHODS This work focuses on influences of stimulant use on dopaminergic function assessed using nuclear-medicine imaging (PET/SPECT). Included are 39 studies on 655 cocaine, amphetamine, methamphetamine or nicotine users, as well as 690 healthy controls. Metaanalyses were conducted separately for D2/D3 receptors and dopamine transporters of the entire striatum, its subregions caudate and putamen respectively. RESULTS Meta-analyses results regarding nicotine did not show significant effects between smokers and nonsmokers. In cocaine users there was a significant decrease in dopamine receptor availability in all regions. The striatal DAT availability was significantly increased in cocaine users. Methamphetamine users showed a significantly decreased dopamine receptor and transporter density in all regions. Significant results also indicate a lower transporter availability in all regions. Amphetamine users showed reduced DAT availability in the striatum, as well as in the sub regions. CONCLUSION This meta-analysis provides evidence that there are ongoing changes in the dopaminergic system associated with the use of stimulants. Especially the results of cocaine, methamphetamine and amphetamine use mainly showed a downregulation. In addition, this meta-analysis is the first to include nicotine. This subset of studies showed evidence for a decreased receptor and DAT availability but no significant results were found in the metaanalyses.
Collapse
|
209
|
Sabzehzari M, Ajamgard M, Shamlouei HR. Influence of non-metallic atoms on the absorption of amphetamines on B12N12 nano-cages. Struct Chem 2019. [DOI: 10.1007/s11224-019-01316-x] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
|
210
|
Sun WL, Quizon PM, Yuan Y, Strauss MJ, McCain R, Zhan CG, Zhu J. Mutational effects of human dopamine transporter at tyrosine88, lysine92, and histidine547 on basal and HIV-1 Tat-inhibited dopamine transport. Sci Rep 2019; 9:3843. [PMID: 30846720 PMCID: PMC6405875 DOI: 10.1038/s41598-019-39872-1] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2018] [Accepted: 12/12/2018] [Indexed: 01/06/2023] Open
Abstract
Dysregulation of dopaminergic system induced by HIV-1 Tat protein-mediated direct inhibition of the dopamine transporter (DAT) has been implicated as a mediating factor of HIV-1 associated neurocognitive disorders. We have reported that single point mutations on human DAT (hDAT) at tyrosine88 (Y88F), lysine92 (K92M), and histidine547 (H547A) differentially regulate basal dopamine uptake but diminish Tat-induced inhibition of dopamine uptake by changing dopamine transport process. This study evaluated the effects of double (Y88F/H547A) and triple (Y88F/K92M/H547A) mutations on basal dopamine uptake, Tat-induced inhibition of DAT function, and dynamic transport process. Compared to wild-type hDAT, the Vmax values of [3H]Dopamine uptake were increased by 96% in Y88F/H547A but decreased by 97% in Y88F/K92M/H547A. [3H]WIN35,428 binding sites were not altered in Y88F/H547A but decreased in Y88F/K92M/H547A. Y88F/H547A mutant attenuated Tat-induced inhibition of dopamine uptake observed in wild-type hDAT. Y88F/H547A displayed an attenuation of zinc-augmented [3H]WIN35,428 binding, increased basal dopamine efflux, and reduced amphetamine-induced dopamine efflux, indicating this mutant alters transporter conformational transitions. These findings further demonstrate that both tyrosine88 and histidine547 on hDAT play a key role in stabilizing basal dopamine transport and Tat-DAT integration. This study provides mechanistic insights into developing small molecules to block multiple sites in DAT for Tat binding.
Collapse
Affiliation(s)
- Wei-Lun Sun
- Department of Drug Discovery and Biomedical Sciences, College of Pharmacy, University of South Carolina, Columbia, SC, USA
| | - Pamela M Quizon
- Department of Drug Discovery and Biomedical Sciences, College of Pharmacy, University of South Carolina, Columbia, SC, USA
| | - Yaxia Yuan
- Molecular Modeling and Biopharmaceutical Center, University of Kentucky, Lexington, KY, USA
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Kentucky, Lexington, KY, USA
| | - Matthew J Strauss
- Department of Drug Discovery and Biomedical Sciences, College of Pharmacy, University of South Carolina, Columbia, SC, USA
| | - Richard McCain
- Department of Drug Discovery and Biomedical Sciences, College of Pharmacy, University of South Carolina, Columbia, SC, USA
| | - Chang-Guo Zhan
- Molecular Modeling and Biopharmaceutical Center, University of Kentucky, Lexington, KY, USA
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Kentucky, Lexington, KY, USA
| | - Jun Zhu
- Department of Drug Discovery and Biomedical Sciences, College of Pharmacy, University of South Carolina, Columbia, SC, USA.
| |
Collapse
|
211
|
The dopamine, serotonin and norepinephrine releasing activities of a series of methcathinone analogs in male rat brain synaptosomes. Psychopharmacology (Berl) 2019; 236:915-924. [PMID: 30341459 PMCID: PMC6475490 DOI: 10.1007/s00213-018-5063-9] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/30/2018] [Accepted: 10/02/2018] [Indexed: 12/12/2022]
Abstract
RATIONALE Novel synthetic "bath salt" cathinones continue to appear on the street as abused and addictive drugs. The range of subjective experiences produced by different cathinones suggests that some compounds have primarily dopaminergic activity (possible stimulants) while others have primarily serotonergic activity (possible empathogenics). An understanding of the structure activity relationships (SARs) of these compounds will help in assessing the likely behavioral effects of future novel structures, and to define potential therapeutic strategies to reverse any reinforcing effects. OBJECTIVES A series of methcathinone analogs was systematically studied for their activity at the dopamine and serotonin transporters. Compound structures varied at the aromatic group, either by substituent or by replacement of the phenyl ring with a naphthalene or indole ring. METHODS A novel, high-yielding synthesis of methcathinone hydrochlorides was developed which avoids isolation of the unstable free bases. Neurotransmitter transporter release activity was determined in rat brain synaptosomes as previously reported. Compounds were also screened for activity at the norepinephrine transporter. RESULTS Twenty-eight methcathinone analogs were analyzed and fully characterized in dopamine and serotonin transporter release assays. Compounds substituted at the 2-position (ortho) were primarily dopaminergic. Compounds substituted at the 3-position (meta) were found to be much less dopaminergic, with some substituents favoring serotonergic activity. Compounds substituted at the 4-position (para) were found to be far more serotonergic, as were disubstituted compounds and other large aromatic groups. One exception was the fluoro-substituted analogs which seem to favor the dopamine transporter. CONCLUSIONS The dopaminergic to serotonergic ratio can be manipulated by choice of substituent and location on the aromatic ring. It is therefore likely possible to tweak the subjective and reinforcing effects of these compounds by adjusting their structure. Certain substituents like a fluoro group tend to favor the dopamine transporter, while others like a trifluoromethyl group favor the serotonin transporter.
Collapse
|
212
|
Halberstadt AL, Brandt SD, Walther D, Baumann MH. 2-Aminoindan and its ring-substituted derivatives interact with plasma membrane monoamine transporters and α 2-adrenergic receptors. Psychopharmacology (Berl) 2019; 236:989-999. [PMID: 30904940 PMCID: PMC6848746 DOI: 10.1007/s00213-019-05207-1] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/26/2018] [Accepted: 02/25/2019] [Indexed: 12/25/2022]
Abstract
RATIONALE Over the last decade, many new psychostimulant analogues have appeared on the recreational drug market and most are derivatives of amphetamine or cathinone. Another class of designer drugs is derived from the 2-aminoindan structural template. Several members of this class, including the parent compound 2-aminoindan (2-AI), have been sold as designer drugs. Another aminoindan derivative, 5-methoxy-2-aminoindan (5-MeO-AI or MEAI), is the active ingredient in a product marketed online as an alcohol substitute. METHODS Here, we tested 2-AI and its ring-substituted derivatives 5-MeO-AI, 5-methoxy-6-methyl-2-aminoindan (MMAI), and 5,6-methylenedioxy-2-aminoindan (MDAI) for their abilities to interact with plasma membrane monoamine transporters for dopamine (DAT), norepinephrine (NET) and serotonin (SERT). We also compared the binding affinities of the aminoindans at 29 receptor and transporter binding sites. RESULTS 2-AI was a selective substrate for NET and DAT. Ring substitution increased potency at SERT while reducing potency at DAT and NET. MDAI was moderately selective for SERT and NET, with tenfold weaker effects on DAT. 5-MeO-AI exhibited some selectivity for SERT, having sixfold lower potency at NET and 20-fold lower potency at DAT. MMAI was highly selective for SERT, with 100-fold lower potency at NET and DAT. The aminoindans had relatively high affinity for α2-adrenoceptor subtypes. 2-AI had particularly high affinity for α2C receptors (Ki = 41 nM) and slightly lower affinity for the α2A (Ki = 134 nM) and α2B (Ki = 211 nM) subtypes. 5-MeO-AI and MMAI also had moderate affinity for the 5-HT2B receptor. CONCLUSIONS 2-AI is predicted to have (+)-amphetamine-like effects and abuse potential whereas the ring-substituted derivatives may produce 3,4-methylenedioxymethamphetamine (MDMA)-like effects but with less abuse liability.
Collapse
Affiliation(s)
- Adam L Halberstadt
- Department of Psychiatry, University of California San Diego, 9500 Gilman Drive, La Jolla, CA, 92093-0804, USA.
- Research Service, VA San Diego Healthcare System, 3350 La Jolla Village Dr., San Diego, CA, 92161, USA.
| | - Simon D Brandt
- School of Pharmacy and Biomolecular Sciences, Liverpool John Moores University, Byrom Street, Liverpool, L3 3AF, UK
| | - Donna Walther
- Designer Drug Research Unit, Intramural Research Program, National Institute on Drug Abuse, National Institutes of Health, Baltimore, MD, 21224, USA
| | - Michael H Baumann
- Designer Drug Research Unit, Intramural Research Program, National Institute on Drug Abuse, National Institutes of Health, Baltimore, MD, 21224, USA
| |
Collapse
|
213
|
Graham-Schmidt KT, Martin-Iverson MT, Waters FAV. Setting the beat of an internal clock: Effects of dexamphetamine on different interval ranges of temporal processing in healthy volunteers. Psych J 2019; 8:90-109. [PMID: 30793518 DOI: 10.1002/pchj.274] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2018] [Revised: 12/14/2018] [Accepted: 01/09/2019] [Indexed: 12/29/2022]
Abstract
Drug studies are powerful models to investigate the neuropharmacological mechanisms underlying temporal processing in humans. This study administered dexamphetamine to 24 healthy volunteers to investigate time perception at different time scales, along with contributions from working memory. Healthy volunteers were administered 0.45 mg/kg dexamphetamine or placebo in a double-blind, crossover, placebo-controlled design. Time perception was assessed using three experimental tasks: a time-discrimination task, which asked participants to determine whether a comparison interval (1200 ± 0, 50, 100, 150, 200 ms) was shorter or longer than a standard interval (1200 ms); a retrospective time estimation task, which required participants to verbally estimate time intervals (10, 30, 60, 90 and 120 s) retrospectively; and a prospective time-production task, where participants were required to prospectively monitor the passing of time (10, 30, 60, 90 and 120 s). Working memory was assessed with the backwards digit span. On the discrimination task, there was a change in the proportion of long-to-short responses and reaction times in the dexamphetamine condition (but no association with working memory), consistent with an increase in the speed of an internal pacemaker, and an overestimation of durations in the timing of shorter intervals. There was an interaction between dexamphetamine, working memory, and performance on the estimation and production tasks, whereby increasing digit span scores were associated with decreasing interval estimates and increased produced intervals in the placebo condition, but were associated with increased interval estimates and decreased produced intervals after dexamphetamine administration. These findings indicate that the dexamphetamine-induced increase in the speed of the internal pacemaker was modulated by the basal working memory capacity of each participant. These findings in healthy humans have important implications for the role of dopamine, and its contributions to timing deficits, in models of psychiatric disorders.
Collapse
Affiliation(s)
- Kyran T Graham-Schmidt
- Faculty of Medicine, Dentistry and Health Sciences, School of Medicine and Pharmacology, University of Western Australia, Perth, Western Australia, Australia
| | - Mathew T Martin-Iverson
- Faculty of Medicine, Dentistry and Health Sciences, School of Medicine and Pharmacology, University of Western Australia, Perth, Western Australia, Australia.,Statewide Department of Neurophysiology, Clinical Research Unit, North Metro Area Mental Health, Graylands Hospital, Perth, Western Australia, Australia
| | - Flavie A V Waters
- Faculty of Medicine, Dentistry and Health Sciences, School of Psychiatry and Clinical Neurosciences, University of Western Australia, Crawley, Western Australia, Australia.,Clinical Research Centre, Graylands Health Campus, North Metropolitan Health Services - Mental Health, Mount Claremont, Western Australia, Australia
| |
Collapse
|
214
|
Matsusue A, Kubo SI, Ikeda T, Tani N, Maeda T, Kashiwagi M, Hara K, Waters B, Takayama M, Ikematsu N, Ishikawa T. VNTR polymorphism in the monoamine oxidase A promoter region and cerebrospinal fluid catecholamine concentrations in forensic autopsy cases. Neurosci Lett 2019; 701:71-76. [PMID: 30794821 DOI: 10.1016/j.neulet.2019.02.029] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2018] [Revised: 02/13/2019] [Accepted: 02/18/2019] [Indexed: 01/01/2023]
Abstract
Monoamine oxidase A (MAOA) plays important roles in the metabolism of catecholamines and modulates adrenergic, noradrenergic, and dopaminergic signaling. A polymorphic promoter variable number tandem repeat (VNTR) locus (MAOA-uVNTR) is located approximately 1.2 kb upstream from MAOA exon 1. Functional studies revealed that MAOA-uVNTR affects gene expression. In the present study, we examined the frequencies of MAOA-uVNTR alleles in Japanese autopsy cases, in which amphetamines or psychotropic drugs were not detected. In total, 87 males and 35 females were evaluated and investigated for the possible effect of MAOA-uVNTR polymorphisms on cerebrospinal fluid (CSF) catecholamine concentrations. In males, there was no significant association between MAOA-uVNTR polymorphisms and CSF adrenaline (Adr), noradrenaline (Nad), or dopamine (DA) levels. In contrast, females who were homozygous for the 3-repeat allele (i.e., 3/3 genotype carriers) had higher CSF levels of Adr (p = 0.024) and DA (p = 0.035) than individuals who were heterozygous or homozygous for the 4-repeat allele (3/4 and 4/4, respectively). We found no significant association between MAOA-uVNTR polymorphisms and CSF Nad levels in females. Thus, the results of the present study indicated that MAOA-uVNTR polymorphism influences CSF Adr and DA levels in females.
Collapse
Affiliation(s)
- Aya Matsusue
- Department of Forensic Medicine, Faculty of Medicine, Fukuoka University, 7-45-1 Nanakuma, Jonan-ku, Fukuoka 814-0180, Japan.
| | - Shin-Ichi Kubo
- Department of Forensic Medicine, Faculty of Medicine, Fukuoka University, 7-45-1 Nanakuma, Jonan-ku, Fukuoka 814-0180, Japan
| | - Tomoya Ikeda
- Department of Legal Medicine, Osaka City University Medical School, Asahi-machi 1-4-3, Abeno, Osaka 545-8585, Japan
| | - Naoto Tani
- Department of Legal Medicine, Osaka City University Medical School, Asahi-machi 1-4-3, Abeno, Osaka 545-8585, Japan
| | - Toshiki Maeda
- Department of Preventive Medicine and Public Health,Faculty of Medicine, Fukuoka University, 7-45-1 Nanakuma, Jonan-ku, Fukuoka 814-0180, Japan
| | - Masayuki Kashiwagi
- Department of Forensic Medicine, Faculty of Medicine, Fukuoka University, 7-45-1 Nanakuma, Jonan-ku, Fukuoka 814-0180, Japan
| | - Kenji Hara
- Department of Forensic Medicine, Faculty of Medicine, Fukuoka University, 7-45-1 Nanakuma, Jonan-ku, Fukuoka 814-0180, Japan
| | - Brian Waters
- Department of Forensic Medicine, Faculty of Medicine, Fukuoka University, 7-45-1 Nanakuma, Jonan-ku, Fukuoka 814-0180, Japan
| | - Mio Takayama
- Department of Forensic Medicine, Faculty of Medicine, Fukuoka University, 7-45-1 Nanakuma, Jonan-ku, Fukuoka 814-0180, Japan
| | - Natsuki Ikematsu
- Department of Forensic Medicine, Faculty of Medicine, Fukuoka University, 7-45-1 Nanakuma, Jonan-ku, Fukuoka 814-0180, Japan
| | - Takaki Ishikawa
- Department of Legal Medicine, Osaka City University Medical School, Asahi-machi 1-4-3, Abeno, Osaka 545-8585, Japan
| |
Collapse
|
215
|
Wu N, Feng Z, He X, Kwon W, Wang J, Xie XQ. Insight of Captagon Abuse by Chemogenomics Knowledgebase-guided Systems Pharmacology Target Mapping Analyses. Sci Rep 2019; 9:2268. [PMID: 30783122 PMCID: PMC6381188 DOI: 10.1038/s41598-018-35449-6] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2018] [Accepted: 09/10/2018] [Indexed: 12/26/2022] Open
Abstract
Captagon, known by its genetic name Fenethylline, is an addictive drug that complicates the War on Drugs. Captagon has a strong CNS stimulating effect than its primary metabolite, Amphetamine. However, multi-targets issues associated with the drug and metabolites as well as its underlying mechanisms have not been fully defined. In the present work, we applied our established drug-abuse chemogenomics-knowledgebase systems pharmacology approach to conduct targets/off-targets mapping (SP-Targets) investigation of Captagon and its metabolites for hallucination addiction, and also analyzed the cell signaling pathways for both Amphetamine and Theophylline with data mining of available literature. Of note, Amphetamine, an agonist for trace amine-associated receptor 1 (TAAR1) with enhancing dopamine signaling (increase of irritability, aggression, etc.), is the main cause of Captagon addiction; Theophylline, an antagonist that blocks adenosine receptors (e.g. A2aR) in the brain responsible for restlessness and painlessness, may attenuate the behavioral sensitization caused by Amphetamine. We uncovered that Theophylline's metabolism and elimination could be retarded due to competition and/or blockage of the CYP2D6 enzyme by Amphetamine; We also found that the synergies between these two metabolites cause Captagon's psychoactive effects to act faster and far more potently than those of Amphetamine alone. We carried out further molecular docking modeling and molecular dynamics simulation to explore the molecular interactions between Amphetamine and Theophylline and their important GPCRs targets, including TAAR1 and adenosine receptors. All of the systems pharmacology analyses and results will shed light insight into a better understanding of Captagon addiction and future drug abuse prevention.
Collapse
Affiliation(s)
- Nan Wu
- Department of Pharmaceutical Sciences and Computational Chemical Genomics Screening Center, School of Pharmacy, University of Pittsburgh, Pittsburgh, Pennsylvania, 15261, United States
- National Center of Excellence for Computational Drug Abuse Research, University of Pittsburgh, Pittsburgh, Pennsylvania, 15261, United States
- Drug Discovery Institute, University of Pittsburgh, Pittsburgh, Pennsylvania, 15261, United States
- Departments of Computational Biology and Structural Biology, School of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania, 15261, United States
| | - Zhiwei Feng
- Department of Pharmaceutical Sciences and Computational Chemical Genomics Screening Center, School of Pharmacy, University of Pittsburgh, Pittsburgh, Pennsylvania, 15261, United States
- National Center of Excellence for Computational Drug Abuse Research, University of Pittsburgh, Pittsburgh, Pennsylvania, 15261, United States
- Drug Discovery Institute, University of Pittsburgh, Pittsburgh, Pennsylvania, 15261, United States
- Departments of Computational Biology and Structural Biology, School of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania, 15261, United States
| | - Xibing He
- Department of Pharmaceutical Sciences and Computational Chemical Genomics Screening Center, School of Pharmacy, University of Pittsburgh, Pittsburgh, Pennsylvania, 15261, United States
- National Center of Excellence for Computational Drug Abuse Research, University of Pittsburgh, Pittsburgh, Pennsylvania, 15261, United States
- Drug Discovery Institute, University of Pittsburgh, Pittsburgh, Pennsylvania, 15261, United States
- Departments of Computational Biology and Structural Biology, School of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania, 15261, United States
| | - William Kwon
- Department of Pharmaceutical Sciences and Computational Chemical Genomics Screening Center, School of Pharmacy, University of Pittsburgh, Pittsburgh, Pennsylvania, 15261, United States
- National Center of Excellence for Computational Drug Abuse Research, University of Pittsburgh, Pittsburgh, Pennsylvania, 15261, United States
- Drug Discovery Institute, University of Pittsburgh, Pittsburgh, Pennsylvania, 15261, United States
- Departments of Computational Biology and Structural Biology, School of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania, 15261, United States
| | - Junmei Wang
- Department of Pharmaceutical Sciences and Computational Chemical Genomics Screening Center, School of Pharmacy, University of Pittsburgh, Pittsburgh, Pennsylvania, 15261, United States.
- National Center of Excellence for Computational Drug Abuse Research, University of Pittsburgh, Pittsburgh, Pennsylvania, 15261, United States.
- Drug Discovery Institute, University of Pittsburgh, Pittsburgh, Pennsylvania, 15261, United States.
- Departments of Computational Biology and Structural Biology, School of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania, 15261, United States.
| | - Xiang-Qun Xie
- Department of Pharmaceutical Sciences and Computational Chemical Genomics Screening Center, School of Pharmacy, University of Pittsburgh, Pittsburgh, Pennsylvania, 15261, United States.
- National Center of Excellence for Computational Drug Abuse Research, University of Pittsburgh, Pittsburgh, Pennsylvania, 15261, United States.
- Drug Discovery Institute, University of Pittsburgh, Pittsburgh, Pennsylvania, 15261, United States.
- Departments of Computational Biology and Structural Biology, School of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania, 15261, United States.
| |
Collapse
|
216
|
van Gaalen MM, Schlumbohm C, Folgering JH, Adhikari S, Bhattacharya C, Steinbach D, Stratford RE. Development of a Semimechanistic Pharmacokinetic-Pharmacodynamic Model Describing Dextroamphetamine Exposure and Striatal Dopamine Response in Rats and Nonhuman Primates following a Single Dose of Dextroamphetamine. J Pharmacol Exp Ther 2019; 369:107-120. [PMID: 30733244 DOI: 10.1124/jpet.118.254508] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2018] [Accepted: 01/23/2019] [Indexed: 11/22/2022] Open
Abstract
Acute central nervous system exposure to dextroamphetamine (d-amphetamine) elicits a multitude of effects, including dual action on the dopamine transporter (DAT) to increase extracellular dopamine, and induction of a negative feedback response to limit the dopamine increase. A semimechanistic pharmacokinetic and pharmacodynamic (PK/PD) model with consideration of these multiple effects as a basis was developed. Integrated pharmacokinetics of d-amphetamine in plasma, brain extracellular fluid (ECF) via microdialysis, and cerebrospinal fluid were characterized using a population approach. This PK model was then linked to an indirect-response pharmacodynamic model using as a basis the measurement of extracellular striatal dopamine, also via microdialysis. In both rats and nonhuman primates (NHPs), d-amphetamine stimulation of dopamine outflow (reverse transport) through DAT was primarily responsible for the dose-linear increase in dopamine. As well, in both species a moderator function was needed to account for loss of the dopamine response in the presence of a relatively sustained d-amphetamine ECF exposure, presumptive of an acute tolerance response. PK/PD model structure was consistent between species; however, there was a 10-fold faster return to baseline dopamine in NHPs in response to an acute d-amphetamine challenge. These results suggest preservation from rodents to NHPs regarding the mechanism by which amphetamine increases extracellular dopamine, but a faster system response in NHPs to tolerate this increase. This microdialysis-based PK/PD model suggests greater value in directing preclinical discovery of novel approaches that modify reverse transport stimulation to treat amphetamine abuse. General value regarding insertion of an NHP model in paradigm rodent-to-human translational research is also suggested.
Collapse
Affiliation(s)
- Marcel M van Gaalen
- Charles River Laboratories Germany GmbH, Göttingen, Germany (M.M.v.G., C.S.); Charles River Laboratories Den Bosch BV, 's-Hertogenbosch, The Netherlands (J.F.); Duquesne School of Pharmacy and Graduate School of Pharmaceutical Sciences, Division of Pharmaceutical, Administrative and Social Sciences, Duquesne University, Pittsburgh, Pennsylvania (S.A., C.B., D.S., R.E.S.)
| | - Christina Schlumbohm
- Charles River Laboratories Germany GmbH, Göttingen, Germany (M.M.v.G., C.S.); Charles River Laboratories Den Bosch BV, 's-Hertogenbosch, The Netherlands (J.F.); Duquesne School of Pharmacy and Graduate School of Pharmaceutical Sciences, Division of Pharmaceutical, Administrative and Social Sciences, Duquesne University, Pittsburgh, Pennsylvania (S.A., C.B., D.S., R.E.S.)
| | - Joost H Folgering
- Charles River Laboratories Germany GmbH, Göttingen, Germany (M.M.v.G., C.S.); Charles River Laboratories Den Bosch BV, 's-Hertogenbosch, The Netherlands (J.F.); Duquesne School of Pharmacy and Graduate School of Pharmaceutical Sciences, Division of Pharmaceutical, Administrative and Social Sciences, Duquesne University, Pittsburgh, Pennsylvania (S.A., C.B., D.S., R.E.S.)
| | - Saugat Adhikari
- Charles River Laboratories Germany GmbH, Göttingen, Germany (M.M.v.G., C.S.); Charles River Laboratories Den Bosch BV, 's-Hertogenbosch, The Netherlands (J.F.); Duquesne School of Pharmacy and Graduate School of Pharmaceutical Sciences, Division of Pharmaceutical, Administrative and Social Sciences, Duquesne University, Pittsburgh, Pennsylvania (S.A., C.B., D.S., R.E.S.)
| | - Chandrali Bhattacharya
- Charles River Laboratories Germany GmbH, Göttingen, Germany (M.M.v.G., C.S.); Charles River Laboratories Den Bosch BV, 's-Hertogenbosch, The Netherlands (J.F.); Duquesne School of Pharmacy and Graduate School of Pharmaceutical Sciences, Division of Pharmaceutical, Administrative and Social Sciences, Duquesne University, Pittsburgh, Pennsylvania (S.A., C.B., D.S., R.E.S.)
| | - Douglas Steinbach
- Charles River Laboratories Germany GmbH, Göttingen, Germany (M.M.v.G., C.S.); Charles River Laboratories Den Bosch BV, 's-Hertogenbosch, The Netherlands (J.F.); Duquesne School of Pharmacy and Graduate School of Pharmaceutical Sciences, Division of Pharmaceutical, Administrative and Social Sciences, Duquesne University, Pittsburgh, Pennsylvania (S.A., C.B., D.S., R.E.S.)
| | - Robert E Stratford
- Charles River Laboratories Germany GmbH, Göttingen, Germany (M.M.v.G., C.S.); Charles River Laboratories Den Bosch BV, 's-Hertogenbosch, The Netherlands (J.F.); Duquesne School of Pharmacy and Graduate School of Pharmaceutical Sciences, Division of Pharmaceutical, Administrative and Social Sciences, Duquesne University, Pittsburgh, Pennsylvania (S.A., C.B., D.S., R.E.S.)
| |
Collapse
|
217
|
Ogata M, Akita H, Ishibashi H. Behavioral responses to anxiogenic tasks in young adult rats with neonatal dopamine depletion. Physiol Behav 2019; 204:10-19. [PMID: 30738032 DOI: 10.1016/j.physbeh.2019.02.006] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2018] [Revised: 01/29/2019] [Accepted: 02/05/2019] [Indexed: 12/19/2022]
Abstract
The dopaminergic neural system plays a crucial role in motor regulation as well as regulation of anxiety-related behaviors. Although rats with neonatal dopamine depletion exhibit motor hyperactivity and have been utilized as animal models of attention deficit hyperactivity disorder, characterization of their behavior under anxiogenic conditions is lacking. In the present study, we investigated behavioral responses to anxiogenic stimuli in young adult rats with neonatal dopamine depletion using the open field (OF), elevated plus maze (EPM), and light/dark (L/B) box tests. The OF and EPM tests were performed under low-light and bright-light conditions. The ameliorative effects of pretreatment with methamphetamine (MAP) or atomoxetine (ATX) on abnormal behaviors induced by neonatal dopamine depletion were also assessed. Rats that underwent 6-hydroxydopamine treatment 4 day after birth showed significant increases in motor activity and decreases in anxiety-related behaviors in OF tests under both conditions and in EPM tests under bright-light conditions. Furthermore, rats with neonatal dopamine depletion did not show normal behavioral responsiveness to changes in the intensity of anxiogenic stimuli. Pretreatment with MAP (4 mg/kg) and ATX (1.2 mg/kg/day) ameliorated motor hyperactivity but not abnormal anxiety-related behaviors. These results suggest that the dopaminergic system plays a crucial role in the development of neural networks involved in locomotion as well as in those involved in anxiety-related behavior. The results indicate that the mechanisms underlying the abnormal anxiolytic responses partially differ from those underlying motor hyperactivity.
Collapse
Affiliation(s)
- Masanori Ogata
- Department of Physiology, School of Allied Health Sciences, Kitasato University, 1-15-1, Kitasato Minami-ku, Sagamihara, Kanagawa 252-0373, Japan.
| | - Hisanao Akita
- Department of Physiology, School of Allied Health Sciences, Kitasato University, 1-15-1, Kitasato Minami-ku, Sagamihara, Kanagawa 252-0373, Japan.
| | - Hitoshi Ishibashi
- Department of Physiology, School of Allied Health Sciences, Kitasato University, 1-15-1, Kitasato Minami-ku, Sagamihara, Kanagawa 252-0373, Japan.
| |
Collapse
|
218
|
“Polytox” synthetic cathinone abuse: A potential role for organic cation transporter 3 in combined cathinone-induced efflux. Neurochem Int 2019; 123:7-12. [DOI: 10.1016/j.neuint.2018.09.008] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2018] [Revised: 09/14/2018] [Accepted: 09/19/2018] [Indexed: 01/11/2023]
|
219
|
Arora R, Haile CN, Kosten TA, Wu Y, Ramakrishnan M, Hawkins LD, Orson FM, Kosten TR. Preclinical efficacy of an anti-methamphetamine vaccine using E6020 adjuvant. Am J Addict 2019; 28:119-126. [PMID: 30701618 DOI: 10.1111/ajad.12867] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2018] [Revised: 01/02/2019] [Accepted: 01/12/2019] [Indexed: 12/15/2022] Open
Abstract
BACKGROUND AND OBJECTIVE Methamphetamine (MA) substance use disorder (SUD) does not have an efficacious pharmacotherapy. We developed a MA vaccine and investigated its potential to attenuate MA induced responses. METHODS We examined a novel adjuvant, E6020, a Toll-like receptor-4 (TLR-4) agonist combined with tetanus-toxoid conjugated to succinyl-methamphetamine (TT-SMA) adsorbed on aluminum hydroxide (alum). Adult BALB/c female mice received the vaccine and booster injections at weeks 0, 3, and 6. The efficacy of the vaccine was assessed by the level and affinity of anti-MA antibodies elicited, its ability to attenuate MA induced locomotor activation and its reduction in the amount of MA entering the brains of vaccinated mice. RESULTS The TT-SMA vaccine containing alum and E6020 adjuvant produced anti-MA antibodies with nanomolar affinities and showed threefold greater peak titer levels than without E6020 (700 vs 250 μg/ml). These antibodies significantly decreased MA-induced locomotor activation (p < .05), and reduced the brain (p < .005) MA levels following MA administration in actively immunized mice. CONCLUSIONS Thus, this anti-MA vaccine formulated with E6020 demonstrated effective functional protection against behavioral disruptions induced by MA. SCIENTIFIC SIGNIFICANCE Together, anti-MA vaccine showing a promising improvement in the efficacy of the vaccine that could be an effective candidate vaccine for methamphetamine use disorder (MUD). Furthermore, combinations of adjuvants may be a tool to design vaccines for MA dependence in humans. (Am J Addict 2019;XX:1-8).
Collapse
Affiliation(s)
- Reetakshi Arora
- Menninger Department of Psychiatry and Behavioral Sciences, Baylor College of Medicine, Houston, Texas.,The Michael E DeBakey Veteran's Affairs Medical Center, Houston, Texas
| | - Colin N Haile
- Menninger Department of Psychiatry and Behavioral Sciences, Baylor College of Medicine, Houston, Texas.,The Michael E DeBakey Veteran's Affairs Medical Center, Houston, Texas.,Department of Psychology, University of Houston, Houston, Texas
| | - Therese A Kosten
- Menninger Department of Psychiatry and Behavioral Sciences, Baylor College of Medicine, Houston, Texas.,The Michael E DeBakey Veteran's Affairs Medical Center, Houston, Texas.,Department of Psychology, University of Houston, Houston, Texas
| | - Yan Wu
- Menninger Department of Psychiatry and Behavioral Sciences, Baylor College of Medicine, Houston, Texas.,The Michael E DeBakey Veteran's Affairs Medical Center, Houston, Texas
| | - Muthu Ramakrishnan
- The Michael E DeBakey Veteran's Affairs Medical Center, Houston, Texas.,Immunology Allergy & Rheumatology, Department of Medicine, Baylor College of Medicine, Houston, Texas
| | | | - Frank M Orson
- The Michael E DeBakey Veteran's Affairs Medical Center, Houston, Texas.,Immunology Allergy & Rheumatology, Department of Medicine, Baylor College of Medicine, Houston, Texas
| | - Thomas R Kosten
- Menninger Department of Psychiatry and Behavioral Sciences, Baylor College of Medicine, Houston, Texas.,The Michael E DeBakey Veteran's Affairs Medical Center, Houston, Texas
| |
Collapse
|
220
|
Shin EJ, Dang DK, Hwang YG, Tran HQ, Sharma N, Jeong JH, Jang CG, Nah SY, Nabeshima T, Yoneda Y, Cadet JL, Kim HC. Significance of protein kinase C in the neuropsychotoxicity induced by methamphetamine-like psychostimulants. Neurochem Int 2019; 124:162-170. [PMID: 30654115 DOI: 10.1016/j.neuint.2019.01.014] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2018] [Revised: 12/27/2018] [Accepted: 01/14/2019] [Indexed: 02/06/2023]
Abstract
The abuse of methamphetamine (MA), an amphetamine (AMPH)-type stimulant, has been demonstrated to be associated with various neuropsychotoxicity, including memory impairment, psychiatric morbidity, and dopaminergic toxicity. Compelling evidence from preclinical studies has indicated that protein kinase C (PKC), a large family of serine/threonine protein kinases, plays an important role in MA-induced neuropsychotoxicity. PKC-mediated N-terminal phosphorylation of dopamine transporter has been identified as one of the prerequisites for MA-induced synaptic dopamine release. Consistently, it has been shown that PKC is involved in MA (or AMPH)-induced memory impairment and mania-like behaviors as well as MA drug dependence. Direct or indirect regulation of factors related to neuronal plasticity seemed to be critical for these actions of PKC. In addition, PKC-mediated mitochondrial dysfunction, oxidative stress or impaired antioxidant defense system has been suggested to play a role in psychiatric and cognitive disturbance induced by MA (or AMPH). In MA-induced dopaminergic toxicity, particularly PKCδ has been shown to trigger oxidative stress, mitochondrial dysfunction, pro-apoptotic changes, and neuroinflammation. Importantly, PKCδ may be a key mediator in the positive feedback loop composed of these detrimental events to potentiate MA-induced dopaminergic toxicity. This review outlines the role of PKC and its individual isozymes in MA-induced neuropsychotoxicity. Better understanding on the molecular mechanism of PKCs might provide a great insight for the development of potential therapeutic or preventive candidates for MA (or AMPH)-associated neuropsychotoxicity.
Collapse
Affiliation(s)
- Eun-Joo Shin
- Neuropsychopharmacology and Toxicology Program, College of Pharmacy, Kangwon National University, Chunchon 24341, Republic of Korea
| | - Duy-Khanh Dang
- Neuropsychopharmacology and Toxicology Program, College of Pharmacy, Kangwon National University, Chunchon 24341, Republic of Korea
| | - Young Gwang Hwang
- Neuropsychopharmacology and Toxicology Program, College of Pharmacy, Kangwon National University, Chunchon 24341, Republic of Korea
| | - Hai-Quyen Tran
- Neuropsychopharmacology and Toxicology Program, College of Pharmacy, Kangwon National University, Chunchon 24341, Republic of Korea
| | - Naveen Sharma
- Neuropsychopharmacology and Toxicology Program, College of Pharmacy, Kangwon National University, Chunchon 24341, Republic of Korea
| | - Ji Hoon Jeong
- Department of Pharmacology, College of Medicine, Chung-Ang University, Seoul 06974, Republic of Korea
| | - Choon-Gon Jang
- Department of Pharmacology, School of Pharmacy, Sungkyunkwan University, Suwon 16419, Republic of Korea
| | - Seung-Yeol Nah
- Ginsentology Research Laboratory and Department of Physiology, College of Veterinary Medicine, Konkuk University, Seoul 05029, Republic of Korea
| | - Toshitaka Nabeshima
- Advanced Diagnostic System Research Laboratory, Fujita Health University Graduate School of Health Science, Toyoake 470-1192, Japan
| | - Yukio Yoneda
- Section of Prophylactic Pharmacology, Kanazawa University Venture Business Laboratory, Kanazawa, Ishikawa 920-1192, Japan
| | - Jean Lud Cadet
- NIDA Intramural Program, Molecular Neuropsychiatry Research Branch, 251 Bayview Boulevard, Baltimore, MD 21224, USA
| | - Hyoung-Chun Kim
- Neuropsychopharmacology and Toxicology Program, College of Pharmacy, Kangwon National University, Chunchon 24341, Republic of Korea.
| |
Collapse
|
221
|
González B, Torres OV, Jayanthi S, Gomez N, Sosa MH, Bernardi A, Urbano FJ, García-Rill E, Cadet JL, Bisagno V. The effects of single-dose injections of modafinil and methamphetamine on epigenetic and functional markers in the mouse medial prefrontal cortex: potential role of dopamine receptors. Prog Neuropsychopharmacol Biol Psychiatry 2019; 88:222-234. [PMID: 30056065 PMCID: PMC8424782 DOI: 10.1016/j.pnpbp.2018.07.019] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/01/2018] [Revised: 07/12/2018] [Accepted: 07/23/2018] [Indexed: 01/02/2023]
Abstract
METH use causes neuroadaptations that negatively impact the prefrontal cortex (PFC) leading to addiction and associated cognitive decline in animals and humans. In contrast, modafinil enhances cognition by increasing PFC function. Accumulated evidence indicates that psychostimulant drugs, including modafinil and METH, regulate gene expression via epigenetic modifications. In this study, we measured the effects of single-dose injections of modafinil and METH on the protein levels of acetylated histone H3 (H3ac) and H4ac, deacetylases HDAC1 and HDAC2, and of the NMDA subunit GluN1 in the medial PFC (mPFC) of mice euthanized 1 h after drug administration. To test if dopamine (DA) receptors (DRs) participate in the biochemical effects of the two drugs, we injected the D1Rs antagonist, SCH23390, or the D2Rs antagonist, raclopride, 30 min before administration of METH and modafinil. We evaluated each drug effect on glutamate synaptic transmission in D1R-expressing layer V pyramidal neurons. We also measured the enrichment of H3ac and H4ac at the promoters of several genes including DA, NE, orexin, histamine, and glutamate receptors, and their mRNA expression, since they are responsive to chronic modafinil and METH treatment. Acute modafinil and METH injections caused similar effects on total histone acetylation, increasing H3ac and decreasing H4ac, and they also increased HDAC1, HDAC2 and GluN1 protein levels in the mouse mPFC. In addition, the effects of the drugs were prevented by pre-treatment with D1Rs and D2Rs antagonists. Specifically, the changes in H4ac, HDAC2, and GluN1 were responsive to SCH23390, whereas those of H3ac and GluN1 were responsive to raclopride. Whole-cell patch clamp in transgenic BAC-Drd1a-tdTomato mice showed that METH, but not modafinil, induced paired-pulse facilitation of EPSCs, suggesting reduced presynaptic probability of glutamate release onto layer V pyramidal neurons. Analysis of histone 3/4 enrichment at specific promoters revealed: i) distinct effects of the drugs on histone 3 acetylation, with modafinil increasing H3ac at Drd1 and Adra1b promoters, but METH increasing H3ac at Adra1a; ii) distinct effects on histone 4 acetylation enrichment, with modafinil increasing H4ac at the Drd2 promoter and decreasing it at Hrh1, but METH increasing H4ac at Drd1; iii) comparable effects of both psychostimulants, increasing H3ac at Drd2, Hcrtr1, and Hrh1 promoters, decreasing H3ac at Hrh3, increasing H4ac at Hcrtr1, and decreasing H4ac at Hcrtr2, Hrh3, and Grin1 promoters. Interestingly, only METH altered mRNA levels of genes with altered histone acetylation status, inducing increased expression of Drd1a, Adra1a, Hcrtr1, and Hrh1, and decreasing Grin1. Our study suggests that although acute METH and modafinil can both increase DA neurotransmission in the mPFC, there are similar and contrasting epigenetic and transcriptional consequences that may account for their divergent clinical effects.
Collapse
Affiliation(s)
- Betina González
- Instituto de Investigaciones Farmacológicas (Universidad de Buenos Aires - Consejo Nacional de Investigaciones Científicas y Técnicas), Ciudad Autónoma de Buenos Aires, Buenos Aires, Argentina
| | - Oscar V Torres
- Department of Behavioral Sciences, San Diego Mesa College, San Diego, California, United States
| | - Subramaniam Jayanthi
- Molecular Neuropsychiatry Research Branch, NIH/NIDA Intramural Research Program, Baltimore, MD, United States
| | - Natalia Gomez
- Instituto de Investigaciones Farmacológicas (Universidad de Buenos Aires - Consejo Nacional de Investigaciones Científicas y Técnicas), Ciudad Autónoma de Buenos Aires, Buenos Aires, Argentina
| | - Máximo H Sosa
- Instituto de Investigaciones Farmacológicas (Universidad de Buenos Aires - Consejo Nacional de Investigaciones Científicas y Técnicas), Ciudad Autónoma de Buenos Aires, Buenos Aires, Argentina
| | - Alejandra Bernardi
- Instituto de Investigaciones Farmacológicas (Universidad de Buenos Aires - Consejo Nacional de Investigaciones Científicas y Técnicas), Ciudad Autónoma de Buenos Aires, Buenos Aires, Argentina
| | - Francisco J Urbano
- Laboratorio de Fisiología y Biología Molecular, Instituto de Fisiología, Biología Molecular y Neurociencias (Universidad de Buenos Aires - Consejo Nacional de Investigaciones Científicas y Técnicas), Ciudad Autónoma de Buenos Aires, Buenos Aires, Argentina
| | - Edgar García-Rill
- Center for Translational Neuroscience, Department of Neurobiology and Developmental Sciences, University of Arkansas for Medical Sciences, Little Rock, AR, United States
| | - Jean-Lud Cadet
- Molecular Neuropsychiatry Research Branch, NIH/NIDA Intramural Research Program, Baltimore, MD, United States
| | - Verónica Bisagno
- Instituto de Investigaciones Farmacológicas (Universidad de Buenos Aires - Consejo Nacional de Investigaciones Científicas y Técnicas), Ciudad Autónoma de Buenos Aires, Buenos Aires, Argentina.
| |
Collapse
|
222
|
Mulvihill KG, Brudzynski SM. Effect of microinjections of dopamine into the nucleus accumbens shell on emission of 50 kHz USV: Comparison with effects of d-amphetamine. Pharmacol Biochem Behav 2019; 176:23-32. [DOI: 10.1016/j.pbb.2018.11.006] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/04/2018] [Revised: 11/09/2018] [Accepted: 11/12/2018] [Indexed: 01/22/2023]
|
223
|
Abstract
There is a plethora of amphetamine derivatives exerting stimulant, euphoric, anti-fatigue, and hallucinogenic effects; all structural properties allowing these effects are contained within the amphetamine structure. In the first part of this review, the interaction of amphetamine with the dopamine transporter (DAT), crucially involved in its behavioral effects, is covered, as well as the role of dopamine synthesis, the vesicular monoamine transporter VMAT2, and organic cation 3 transporter (OCT3). The second part deals with requirements in amphetamine's effect on the kinases PKC, CaMKII, and ERK, whereas the third part focuses on where we are in developing anti-amphetamine therapeutics. Thus, treatments are discussed that target DAT, VMAT2, PKC, CaMKII, and OCT3. As is generally true for the development of therapeutics for substance use disorder, there are multiple preclinically promising specific compounds against (meth)amphetamine, for which further development and clinical trials are badly needed.
Collapse
Affiliation(s)
- Maarten E A Reith
- Department of Psychiatry, New York University School of Medicine, New York, NY, USA.
| | - Margaret E Gnegy
- Department of Pharmacology, University of Michigan School of Medicine, Ann Arbor, MI, USA
| |
Collapse
|
224
|
Smith CT, San Juan MD, Dang LC, Katz DT, Perkins SF, Burgess LL, Cowan RL, Manning HC, Nickels ML, Claassen DO, Samanez-Larkin GR, Zald DH. Ventral striatal dopamine transporter availability is associated with lower trait motor impulsivity in healthy adults. Transl Psychiatry 2018; 8:269. [PMID: 30531858 PMCID: PMC6286354 DOI: 10.1038/s41398-018-0328-y] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/16/2018] [Revised: 11/13/2018] [Accepted: 11/15/2018] [Indexed: 12/18/2022] Open
Abstract
Impulsivity is a transdiagnostic feature of a range of externalizing psychiatric disorders. Preclinical work links reduced ventral striatal dopamine transporter (DAT) availability with heightened impulsivity and novelty seeking. However, there is a lack of human data investigating the relationship between DAT availability, particularly in subregions of the striatum, and the personality traits of impulsivity and novelty seeking. Here we collected PET measures of DAT availability (BPND) using the tracer 18F-FE-PE2I in 47 healthy adult subjects and examined relations between BPND in striatum, including its subregions: caudate, putamen, and ventral striatum (VS), and trait impulsivity (Barratt Impulsiveness Scale: BIS-11) and novelty seeking (Tridimensional Personality Questionnaire: TPQ-NS), controlling for age and sex. DAT BPND in each striatal subregion showed nominal negative associations with total BIS-11 but not TPQ-NS. At the subscale level, VS DAT BPND was significantly associated with BIS-11 motor impulsivity (e.g., taking actions without thinking) after correction for multiple comparisons. VS DAT BPND explained 13.2% of the variance in motor impulsivity. Our data demonstrate that DAT availability in VS is negatively related to impulsivity and suggest a particular influence of DAT regulation of dopamine signaling in VS on acting without deliberation (BIS motor impulsivity). While needing replication, these data converge with models of ventral striatal functions that emphasize its role as a key interface linking motivation to action.
Collapse
Affiliation(s)
- Christopher T. Smith
- 0000 0001 2264 7217grid.152326.1Department of Psychology, PMB 407817, Vanderbilt University, 2301 Vanderbilt Place, Nashville, TN 37240-7817 USA
| | - M. Danica San Juan
- 0000 0001 2264 7217grid.152326.1Department of Psychology, PMB 407817, Vanderbilt University, 2301 Vanderbilt Place, Nashville, TN 37240-7817 USA
| | - Linh C. Dang
- 0000 0001 2264 7217grid.152326.1Department of Psychology, PMB 407817, Vanderbilt University, 2301 Vanderbilt Place, Nashville, TN 37240-7817 USA
| | - Daniel T. Katz
- 0000 0001 2264 7217grid.152326.1Department of Psychology, PMB 407817, Vanderbilt University, 2301 Vanderbilt Place, Nashville, TN 37240-7817 USA
| | - Scott F. Perkins
- 0000 0001 2264 7217grid.152326.1Department of Psychology, PMB 407817, Vanderbilt University, 2301 Vanderbilt Place, Nashville, TN 37240-7817 USA
| | - Leah L. Burgess
- 0000 0001 2264 7217grid.152326.1Department of Psychology, PMB 407817, Vanderbilt University, 2301 Vanderbilt Place, Nashville, TN 37240-7817 USA
| | - Ronald L. Cowan
- 0000 0001 2264 7217grid.152326.1Department of Psychology, PMB 407817, Vanderbilt University, 2301 Vanderbilt Place, Nashville, TN 37240-7817 USA ,0000 0004 1936 9916grid.412807.8Department of Psychiatry and Behavioral Sciences, Vanderbilt University Medical Center, 1601 23rd Avenue South, Suite 3057, Nashville, TN 37212 USA ,0000 0004 1936 9916grid.412807.8Department of Radiology and Radiological Sciences, Vanderbilt University Medical Center, Medical Center North, 1161 21st Avenue South, Nashville, TN 37232 USA
| | - H. Charles Manning
- 0000 0004 1936 9916grid.412807.8Department of Radiology and Radiological Sciences, Vanderbilt University Medical Center, Medical Center North, 1161 21st Avenue South, Nashville, TN 37232 USA ,0000 0001 2264 7217grid.152326.1Department of Chemistry, Vanderbilt University, 7330 Stevenson Center, Station B 351822, Nashville, TN 37235 USA ,0000 0001 2264 7217grid.152326.1Department of Biomedical Engineering, PMB 351826, Vanderbilt University, 2301 Vanderbilt Place, Nashville, TN 37235-1826 USA ,0000 0004 1936 9916grid.412807.8Department of Neurological Surgery, Vanderbilt University Medical Center, 1161 21st Avenue South, T4224 Medical Center North, Nashville, TN 37232-2380 USA
| | - Michael L. Nickels
- 0000 0004 1936 9916grid.412807.8Department of Radiology and Radiological Sciences, Vanderbilt University Medical Center, Medical Center North, 1161 21st Avenue South, Nashville, TN 37232 USA
| | - Daniel O. Claassen
- 0000 0004 1936 9916grid.412807.8Department of Neurology, Vanderbilt University Medical Center, 1161 21st Avenue South, A-0118, Nashville, TN 37232-2551 USA
| | - Gregory R. Samanez-Larkin
- 0000 0004 1936 7961grid.26009.3dDepartment of Psychology and Neuroscience, Duke University, 417 Chapel Drive, Durham, NC 27708 USA
| | - David H. Zald
- 0000 0001 2264 7217grid.152326.1Department of Psychology, PMB 407817, Vanderbilt University, 2301 Vanderbilt Place, Nashville, TN 37240-7817 USA ,0000 0004 1936 9916grid.412807.8Department of Psychiatry and Behavioral Sciences, Vanderbilt University Medical Center, 1601 23rd Avenue South, Suite 3057, Nashville, TN 37212 USA
| |
Collapse
|
225
|
Label-free imaging of neurotransmitters in live brain tissue by multi-photon ultraviolet microscopy. Neuronal Signal 2018; 2:NS20180132. [PMID: 32714595 PMCID: PMC7373235 DOI: 10.1042/ns20180132] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2018] [Revised: 10/20/2018] [Accepted: 10/29/2018] [Indexed: 12/20/2022] Open
Abstract
Visualizing small biomolecules in living cells remains a difficult challenge. Neurotransmitters provide one of the most frustrating examples of this difficulty, as our understanding of signaling in the brain critically depends on our ability to follow the neurotransmitter traffic. Last two decades have seen considerable progress in probing some of the neurotransmitters, e.g. by using false neurotransmitter mimics, chemical labeling techniques, or direct fluorescence imaging. Direct imaging harnesses the weak UV fluorescence of monoamines, which are some of the most important neurotransmitters controlling mood, memory, appetite, and learning. Here we describe the progress in imaging of these molecules using the least toxic direct excitation route found so far, namely multi-photon (MP) imaging. MP imaging of serotonin, and more recently that of dopamine, has allowed researchers to determine the location of the vesicles, follow their intracellular dynamics, probe their content, and monitor their release. Recent developments have even allowed ratiometric quantitation of the vesicular content. This review shows that MP ultraviolet (MP-UV) microscopy is an effective but underutilized method for imaging monoamine neurotransmitters in neurones and brain tissue.
Collapse
|
226
|
Mutlu U, Ikram MA, Ikram MK. Clinical interpretation of negative mediated interaction. Int J Epidemiol 2018; 48:1286-1293. [DOI: 10.1093/ije/dyy252] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/25/2018] [Indexed: 01/08/2023] Open
Abstract
AbstractBackgroundRecently, using a counterfactual framework, a causal mediation analysis has been formalized to decompose the total effect of a time-fixed exposure on an outcome into four components that can be loosely defined as being components due to mediation only, interaction only, mediated interaction and neither. The interpretation of the estimated effect sizes is challenging when these components of the total effect are of the opposite sign compared with each other. Particularly, a negative mediated interaction might be intuitively difficult to conceptualize and, so far, lacks an easy-to-understand biological or mechanical interpretation.MethodsIn this paper, we focus on negative mediated interaction, and propose an interpretation using biological examples. For negative mediated interaction to be present, the effect of interaction on the outcome and the effect of the exposure on the mediator should be in opposite directions.ResultsIn this article, we give examples of biological and biochemical processes that may exhibit negative mediated interaction, such as drug treatment in clinical practice, allosteric effects of enzymes, different adaptations in the cardiovascular system and its effect on brain health, and antibiotic drug–drug interactions.ConclusionsWe aim to make researchers realize that negative-effect estimates might reflect relevant biological processes in the mechanism under study.
Collapse
Affiliation(s)
- Unal Mutlu
- Department of Epidemiology, Erasmus Medical Center, Rotterdam, The Netherlands
| | | | - Mohammad Kamran Ikram
- Department of Epidemiology, Erasmus Medical Center, Rotterdam, The Netherlands
- Department of Neurology, Erasmus Medical Center, Rotterdam, The Netherlands
| |
Collapse
|
227
|
Mukherjee A, Dye BA, Clague J, Belin TR, Shetty V. Methamphetamine use and oral health-related quality of life. Qual Life Res 2018; 27:3179-3190. [PMID: 30076578 PMCID: PMC8346633 DOI: 10.1007/s11136-018-1957-6] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/28/2018] [Indexed: 12/26/2022]
Abstract
PURPOSE Methamphetamine (MA) is associated with adverse health effects, including the rampant tooth decay condition called "Meth Mouth." However, the impact of MA use on oral health-related quality of life (OHRQOL) is unknown. This study assessed the relationship between MA use and self-reported OHRQOL. METHODS This cross-sectional study uses information from 545 MA-using participants recruited from Los Angeles County, California. Dental examinations were performed by three calibrated dentists using National Health and Nutrition Examination Survey (NHANES) protocols. Data on socio-demographic, behavioral, and drug-use history were recorded using questionnaires. Participants were categorized as 'light' or 'moderate/heavy' users based on reported frequency of MA use in the past 30 days. Route of MA administration was categorized as 'smoking' or 'other.' Self-reported OHRQOL was based on the Oral Health Impact Profile scale. RESULTS Majority of the participants were male (80.9%). Median age was 45.0 years (IQR-13.0). Median number of days of MA use was 10.0 (IQR-12.0). Smoking was the preferred route of MA use (70.2%). Root caries in ≥ 3 teeth were reported in 78% of MA users. More than half of the participants reported having painful aching in mouth, avoidance of particular food items, feeling embarrassed, and discomfort while eating in the last 12 months. In unadjusted logistic models, moderate/heavy MA users were more likely to report an affected sense of taste [OR = 1.58, 95% CI (1.10-2.27)] and avoidance of particular foods [OR = 1.45, 95% CI (1.02-2.01)] than light users. Among individuals preferring other MA administration routes, moderate/heavy MA users were 3.09 times as likely to report an affected sense of taste than light users [OR = 3.09, 95% CI (1.52-6.27)]. CONCLUSION Oral health and OHRQOL appear to be worse among Methamphetamine users than in the US general population.
Collapse
Affiliation(s)
- Amrita Mukherjee
- NIH, National Institute of Dental and Craniofacial Research, 31 Center Drive Suite 5B55, Bethesda, MD, 20892-2190, USA
| | - Bruce A Dye
- NIH, National Institute of Dental and Craniofacial Research, 31 Center Drive Suite 5B55, Bethesda, MD, 20892-2190, USA.
- NIDCR Dental Public Health Residency Program, NIH/NIDCR, 31 Center Drive Suite 5B55, Bethesda, MD, 20892-2190, USA.
| | - Jason Clague
- UCLA, School of Dentistry, 10833 Le Conte Ave, Office 23-009CHS, Box 951668, Los Angeles, CA, 90095-1668, USA
- Department of Biostatistics, UCLA, Fielding School of Public Health, 650 Charles E. Young Dr. South, Room 51-267, Los Angeles, CA, 90095-1772, USA
| | - Thomas R Belin
- Department of Biostatistics, UCLA, Fielding School of Public Health, 650 Charles E. Young Dr. South, Room 51-267, Los Angeles, CA, 90095-1772, USA
| | - Vivek Shetty
- UCLA, School of Dentistry, 10833 Le Conte Ave, Office 23-009CHS, Box 951668, Los Angeles, CA, 90095-1668, USA
| |
Collapse
|
228
|
Lam L, Anand S, Li X, Tse ML, Zhao JX, Chan EW. Efficacy and safety of naltrexone for amfetamine and methamfetamine use disorder: a systematic review of randomized controlled trials. Clin Toxicol (Phila) 2018; 57:225-233. [PMID: 30451013 DOI: 10.1080/15563650.2018.1529317] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/27/2022]
Abstract
INTRODUCTION Amfetamine and methamfetamine abuse remains a prevalent health problem, increasing the burden on healthcare. Naltrexone, a µ-opioid receptor antagonist, has been suggested as a promising treatment for amfetamine and methamfetamine use disorder. OBJECTIVE To review the current evidence for the efficacy and safety of naltrexone as a pharmacological treatment for amfetamine and methamfetamine use disorder. The primary outcome was defined as abstinence or reduction of use. Secondary outcomes were, attenuated "positive" subjective effects (e.g., "feel good," "craving," etc.) of amfetamine or methamfetamine after naltrexone treatment, adverse events and physiological changes (e.g., blood pressure, heart rate). METHODS This systematic review was conducted according to the Preferred Reporting Items for Systematic Reviews and Meta-Analyses (PRISMA) statement. A systematic literature search was conducted on 2 April 2017, and updated on 31 March 2018. Records were retrieved from databases including PubMed, EMBASE Classic plus EMBASE 1980 via Ovid, and the databases were searched using keywords and/or headings: (naltrexone AND amfetamine AND dependence) OR (naltrexone AND amfetamine AND craving) OR (vivitrol AND amfetamine) OR (revia AND amfetamine) OR (naltrexone AND amfetamine) OR (naltrexone AND methamfetamine dependence) OR (naltrexone AND methamfetamine AND craving) OR (vivitrol AND methamfetamine) OR (revia AND methamfetamine) OR (naltrexone AND ice) OR (naltrexone AND crystal meth) OR (naltrexone AND methamfetamine). Studies investigating the effects of naltrexone on amfetamine or methamfetamine use were eligible for inclusion. All studies were rated as low risk of bias using the Cochrane tool for risk of bias. RESULTS Among 591 identified studies, there were four randomized controlled trials. Two studies investigated the effects of naltrexone on amfetamine use disorder and two on methamfetamine use. Compared to placebo, the abstinence rate was increased significantly (p < 0.05) by naltrexone in one of two amfetamine studies, whereas there was no statistical difference in the only study reporting methamfetamine use. In one out of two amfetamine studies, naltrexone significantly attenuated either craving levels or subjective effects (e.g., "want more," "like effect") relative to placebo (p < 0.05). Additionally, only in one of two methamfetamine studies did naltrexone produce a significant reduction (p < 0.05) in craving levels or attenuated subjective effects. Both amfetamine and methamfetamine studies showed good tolerability of naltrexone, with few adverse events seen. CONCLUSIONS There is presently insufficient evidence to support the use of naltrexone in amfetamine and metamfetamine use disorders. There is a compelling need for high-quality studies to further evaluate the potential use of naltrexone.
Collapse
Affiliation(s)
- Lam Lam
- a Centre for Safe Medication Practice and Research, Department of Pharmacology and Pharmacy , Li Ka Shing Faculty of Medicine, The University of Hong Kong , Hong Kong , Hong Kong
| | - Shweta Anand
- a Centre for Safe Medication Practice and Research, Department of Pharmacology and Pharmacy , Li Ka Shing Faculty of Medicine, The University of Hong Kong , Hong Kong , Hong Kong
| | - Xue Li
- a Centre for Safe Medication Practice and Research, Department of Pharmacology and Pharmacy , Li Ka Shing Faculty of Medicine, The University of Hong Kong , Hong Kong , Hong Kong
| | - M L Tse
- b Hong Kong Poison Information Centre , Hospital Authority and Clinical Toxicology Department, United Christian Hospital , Hong Kong
| | - Jia X Zhao
- a Centre for Safe Medication Practice and Research, Department of Pharmacology and Pharmacy , Li Ka Shing Faculty of Medicine, The University of Hong Kong , Hong Kong , Hong Kong
| | - Esther W Chan
- a Centre for Safe Medication Practice and Research, Department of Pharmacology and Pharmacy , Li Ka Shing Faculty of Medicine, The University of Hong Kong , Hong Kong , Hong Kong
| |
Collapse
|
229
|
Mulvihill KG. Presynaptic regulation of dopamine release: Role of the DAT and VMAT2 transporters. Neurochem Int 2018; 122:94-105. [PMID: 30465801 DOI: 10.1016/j.neuint.2018.11.004] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2018] [Revised: 10/28/2018] [Accepted: 11/08/2018] [Indexed: 01/23/2023]
Abstract
The signaling dynamics of the neurotransmitter dopamine has been established to have an important role in a variety of behavioural processes including motor control, cognition, and emotional processing. Key regulators of transmitter release and the signaling dynamics of dopamine are the plasma membrane reuptake transporter (DAT) and the vesicular monoamine transporter (VMAT2). These proteins serve to remove dopamine molecules from the extracellular and cytosolic space, respectively and both determine the amount of transmitter released from synaptic vesicles. This review provides an overview of how these transporter proteins are involved in molecular regulation and function together to govern the dynamics of vesicular release with opposing effects on the quantal size and extracellular concentration of dopamine. These transporter proteins are both focal points of convergence for a variety of regulatory molecular cascades as well as targets for many pharmacological agents. The ratio between these transporters is argued to be useful as a molecular marker for delineating dopamine functional subsystems that may differ in transmitter release patterns.
Collapse
Affiliation(s)
- Kevin G Mulvihill
- Department of Psychology, Brock University, St. Catharines, ON, L2S 3A1, Canada.
| |
Collapse
|
230
|
Sungur AÖ, Redecker TM, Andres E, Dürichen W, Schwarting RKW, Del Rey A, Wöhr M. Reduced Efficacy of d-Amphetamine and 3,4-Methylenedioxymethamphetamine in Inducing Hyperactivity in Mice Lacking the Postsynaptic Scaffolding Protein SHANK1. Front Mol Neurosci 2018; 11:419. [PMID: 30505269 PMCID: PMC6250831 DOI: 10.3389/fnmol.2018.00419] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2018] [Accepted: 10/26/2018] [Indexed: 02/02/2023] Open
Abstract
Genetic defects in the three SH3 and multiple ankyrin repeat domains (SHANK) genes (SHANK1, SHANK2, and SHANK3) are associated with multiple major neuropsychiatric disorders, including autism spectrum disorder (ASD), schizophrenia (SCZ), and bipolar disorder (BPD). Psychostimulant-induced hyperactivity is a commonly applied paradigm to assess behavioral phenotypes related to BPD and considered to be the gold standard for modeling mania-like elevated drive in mouse models. Therefore, the goal of our present study was to test whether Shank1 plays a role in the behavioral effects of psychostimulants and whether this is associated with genotype-dependent neurochemical alterations. To this aim, male and female null mutant Shank1-/- mice were treated with d-amphetamine (AMPH; 2.5 mg/kg) and 3,4-methylenedioxymethamphetamine (MDMA, commonly known as ecstasy; 20 mg/kg), and psychostimulant-induced hyperactivity was compared to heterozygous Shank1+/- and wildtype Shank1+/+ littermate controls. Results show that Shank1-/- mice display reduced psychostimulant-induced hyperactivity, although psychostimulants robustly stimulated locomotor activity in littermate controls. Shank1 deletion effects emerged throughout development, were particularly prominent in adulthood, and seen in response to both psychostimulants, i.e., AMPH and MDMA. Specifically, while AMPH-induced hyperactivity was reduced but still detectable in Shank1-/- mice, MDMA-induced hyperactivity was robustly blocked and completely absent in Shank1-/- mice. Reduced efficacy of psychostimulants to stimulate hyperactivity in Shank1-/- mice might be associated with alterations in the neurochemical architecture in prefrontal cortex, nucleus accumbens, and hypothalamus. Our observation that psychostimulant-induced hyperactivity is reduced rather than enhanced in Shank1-/- mice clearly speaks against a behavioral phenotype with relevance to BPD. Lack of BPD-like phenotype is consistent with currently available human data linking mutations in SHANK2 and SHANK3 but not SHANK1 to BPD.
Collapse
Affiliation(s)
- A Özge Sungur
- Behavioral Neuroscience, Experimental and Biological Psychology, Philipps University of Marburg, Marburg, Germany.,Center for Mind, Brain and Behavior, Philipps University of Marburg, Marburg, Germany
| | - Tobias M Redecker
- Behavioral Neuroscience, Experimental and Biological Psychology, Philipps University of Marburg, Marburg, Germany
| | - Elena Andres
- Research Group Immunophysiology, Division of Neurophysiology, Institute of Physiology and Pathophysiology, Philipps University of Marburg, Marburg, Germany
| | - Wiebke Dürichen
- Behavioral Neuroscience, Experimental and Biological Psychology, Philipps University of Marburg, Marburg, Germany
| | - Rainer K W Schwarting
- Behavioral Neuroscience, Experimental and Biological Psychology, Philipps University of Marburg, Marburg, Germany.,Center for Mind, Brain and Behavior, Philipps University of Marburg, Marburg, Germany
| | - Adriana Del Rey
- Research Group Immunophysiology, Division of Neurophysiology, Institute of Physiology and Pathophysiology, Philipps University of Marburg, Marburg, Germany
| | - Markus Wöhr
- Behavioral Neuroscience, Experimental and Biological Psychology, Philipps University of Marburg, Marburg, Germany.,Center for Mind, Brain and Behavior, Philipps University of Marburg, Marburg, Germany
| |
Collapse
|
231
|
Zheng C, Chen G, Tan Y, Zeng W, Peng Q, Wang J, Cheng C, Yang X, Nie S, Xu Y, Zhang Z, Papa SM, Ye K, Cao X. TRH Analog, Taltirelin Improves Motor Function of Hemi-PD Rats Without Inducing Dyskinesia via Sustained Dopamine Stimulating Effect. Front Cell Neurosci 2018; 12:417. [PMID: 30555300 PMCID: PMC6282053 DOI: 10.3389/fncel.2018.00417] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2018] [Accepted: 10/25/2018] [Indexed: 01/08/2023] Open
Abstract
Thyrotropin-releasing hormone (TRH) and its analogs are able to stimulate the release of the endogenic dopamine (DA) in the central nervous system. However, this effect has not been tested in the Parkinson’s disease (PD), which is characterized by the DA deficiency due to the dopaminergic neurons loss in the substantia nigra. Here, we investigated the therapeutic effect of Taltirelin, a long-acting TRH analog on 6-hydroxydopamine-lesioned hemi-Parkinsonian rat model. 1–10 mg/kg Taltirelin i.p. administration significantly improved the locomotor function and halted the electrophysiological abnormities of PD animals without inducing dyskinesia even with high-dose for 7 days treatment. Microdialysis showed that Taltirelin gently and persistently promoted DA release in the cortex and striatum, while L-DOPA induced a sharp rise of DA especially in the cortex. The DA-releasing effect of Taltirelin was alleviated by reserpine, vanoxerine (GBR12909) or AMPT, indicating a mechanism involving vesicular monoamine transporter-2 (VMAT-2), dopamine transporter (DAT) and tyrosine hydroxylase (TH). The in vivo and in vitro experiments further supported that Taltirelin affected the regulation of TH expression in striatal neurons, which was mediated by p-ERK1/2. Together, this study demonstrated that Taltirelin improved motor function of hemi-PD rats without inducing dyskinesia, thus supporting a further exploration of Taltirelin for PD treatment.
Collapse
Affiliation(s)
- Cong Zheng
- Department of Neurology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Guiqin Chen
- Department of Neurology, Renmin Hospital of Wuhan University, Wuhan, China
| | - Yang Tan
- Department of Neurology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Weiqi Zeng
- Department of Neurology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Qiwei Peng
- Department of Neurology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Ji Wang
- Department of Neurology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Chi Cheng
- Department of Neurology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Xiaoman Yang
- Department of Neurology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Shuke Nie
- Department of Neurology, Renmin Hospital of Wuhan University, Wuhan, China
| | - Yan Xu
- Department of Neurology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Zhentao Zhang
- Department of Neurology, Renmin Hospital of Wuhan University, Wuhan, China
| | - Stella M Papa
- Yerkes National Primate Research Center, Emory University School of Medicine, Atlanta, GA, United States.,Department of Neurology, Emory University School of Medicine, Atlanta, GA, United States
| | - Keqiang Ye
- Department of Pathology and Laboratory Medicine, Emory University School of Medicine, Atlanta, GA, United States
| | - Xuebing Cao
- Department of Neurology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
232
|
Mayer FP, Schmid D, Owens WA, Gould GG, Apuschkin M, Kudlacek O, Salzer I, Boehm S, Chiba P, Williams PH, Wu HH, Gether U, Koek W, Daws LC, Sitte HH. An unsuspected role for organic cation transporter 3 in the actions of amphetamine. Neuropsychopharmacology 2018; 43:2408-2417. [PMID: 29773909 PMCID: PMC6180071 DOI: 10.1038/s41386-018-0053-5] [Citation(s) in RCA: 39] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/03/2017] [Revised: 03/19/2018] [Accepted: 03/21/2018] [Indexed: 01/11/2023]
Abstract
Amphetamine abuse is a major public health concern for which there is currently no effective treatment. To develop effective treatments, the mechanisms by which amphetamine produces its abuse-related effects need to be fully understood. It is well known that amphetamine exerts its actions by targeting high-affinity transporters for monoamines, in particular the cocaine-sensitive dopamine transporter. Organic cation transporter 3 (OCT3) has recently been found to play an important role in regulating monoamine signaling. However, whether OCT3 contributes to the actions of amphetamine is unclear. We found that OCT3 is expressed in dopamine neurons. Then, applying a combination of in vivo, ex vivo, and in vitro approaches, we revealed that a substantial component of amphetamine's actions is OCT3-dependent and cocaine insensitive. Our findings support OCT3 as a new player in the actions of amphetamine and encourage investigation of this transporter as a potential new target for the treatment of psychostimulant abuse.
Collapse
Affiliation(s)
- Felix P. Mayer
- 0000 0000 9259 8492grid.22937.3dCenter for Physiology and Pharmacology, Medical University of Vienna, 1090 Vienna, Austria
| | - Diethart Schmid
- 0000 0000 9259 8492grid.22937.3dCenter for Physiology and Pharmacology, Medical University of Vienna, 1090 Vienna, Austria
| | - W. Anthony Owens
- 0000 0001 0629 5880grid.267309.9Department of Cellular and Integrative Physiology, University of Texas Health Science Center at San Antonio, San Antonio, TX 78229 USA
| | - Georgianna G. Gould
- 0000 0001 0629 5880grid.267309.9Department of Cellular and Integrative Physiology, University of Texas Health Science Center at San Antonio, San Antonio, TX 78229 USA
| | - Mia Apuschkin
- 0000 0001 0674 042Xgrid.5254.6Molecular Neuropharmacology and Genetics Laboratory, Department of Neuroscience and Pharmacology, Faculty of Health and Medical Sciences, University of Copenhagen, Panum Institute 18.6, 2200 Copenhagen N, Denmark
| | - Oliver Kudlacek
- 0000 0000 9259 8492grid.22937.3dCenter for Physiology and Pharmacology, Medical University of Vienna, 1090 Vienna, Austria
| | - Isabella Salzer
- 0000 0000 9259 8492grid.22937.3dCenter for Physiology and Pharmacology, Medical University of Vienna, 1090 Vienna, Austria
| | - Stefan Boehm
- 0000 0000 9259 8492grid.22937.3dCenter for Physiology and Pharmacology, Medical University of Vienna, 1090 Vienna, Austria
| | - Peter Chiba
- 0000 0000 9259 8492grid.22937.3dInstitute of Medical Chemistry, Medical University of Vienna, 1090 Vienna, Austria
| | - Piper H. Williams
- 0000 0001 2156 6853grid.42505.36Department of Pediatrics, The Saban Research Institute, Children’s Hospital Los Angeles, Keck School of Medicine of USC, 4661 Sunset Blvd. Rm 307, Los Angeles, CA 90027 USA
| | - Hsiao-Huei Wu
- 0000 0001 2156 6853grid.42505.36Department of Pediatrics, The Saban Research Institute, Children’s Hospital Los Angeles, Keck School of Medicine of USC, 4661 Sunset Blvd. Rm 307, Los Angeles, CA 90027 USA
| | - Ulrik Gether
- 0000 0001 0674 042Xgrid.5254.6Molecular Neuropharmacology and Genetics Laboratory, Department of Neuroscience and Pharmacology, Faculty of Health and Medical Sciences, University of Copenhagen, Panum Institute 18.6, 2200 Copenhagen N, Denmark
| | - Wouter Koek
- 0000 0001 0629 5880grid.267309.9Department of Psychiatry, University of Texas Health Science Center at San Antonio, San Antonio, TX 78229 USA ,0000 0001 0629 5880grid.267309.9Department of Pharmacology, University of Texas Health Science Center at San Antonio, San Antonio, TX 78229 USA
| | - Lynette C. Daws
- 0000 0001 0629 5880grid.267309.9Department of Cellular and Integrative Physiology, University of Texas Health Science Center at San Antonio, San Antonio, TX 78229 USA ,0000 0001 0629 5880grid.267309.9Department of Pharmacology, University of Texas Health Science Center at San Antonio, San Antonio, TX 78229 USA
| | - Harald H. Sitte
- 0000 0000 9259 8492grid.22937.3dCenter for Physiology and Pharmacology, Medical University of Vienna, 1090 Vienna, Austria ,0000 0000 9259 8492grid.22937.3dCenter for Addiction Research and Science, Medical University Vienna, Waehringerstrasse 13 A, 1090 Vienna, Austria
| |
Collapse
|
233
|
Garton DR, Ross SG, Maldonado-Hernández R, Quick M, Lasalde-Dominicci JA, Lizardi-Ortiz JE. Amphetamine enantiomers inhibit homomeric α7 nicotinic receptor through a competitive mechanism and within the intoxication levels in humans. Neuropharmacology 2018; 144:172-183. [PMID: 30359640 DOI: 10.1016/j.neuropharm.2018.10.032] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2018] [Revised: 10/10/2018] [Accepted: 10/21/2018] [Indexed: 11/28/2022]
Abstract
Amphetamine-type stimulants (ATS) are the second most consumed illicit drug worldwide and lack good treatments for associated substance use disorders, lagging behind other addictive drugs. For this reason, a deeper understanding of the pharmacodynamics of ATS is required. The present study seeks to determine amphetamine (AMPH) enantiomers' effects on the homomeric α7 nicotinic acetylcholine receptor (α7 nAChR). Here we have shown that AMPH enantiomers bind to the α7 nAChR and competitively inhibit acetylcholine responses. Our in silico docking analysis suggests that AMPH binds close to the β7 strand of the B-loop of a chimera comprising of the human α7 nAChR and the acetylcholine binding protein from Lymnaea stagnalis. This may inhibit the required movement of the C-loop for channel opening, due to steric hindrance, providing a structural mechanism for its antagonist effect. Finally, we have shown that, in α7 nAChR full knockout mice, the behavioral response to D-AMPH is attenuated, providing direct evidence for the role of α7 nAChRs on the physiological response to D-AMPH. Importantly, D-AMPH exerts these effects at concentrations predicted to be pharmacologically relevant for chronic methamphetamine users and during binges. In conclusion, our data present new findings that implicate the α7 nAChR on the pharmacodynamics of ATS, which may be important for behavioral responses to these drugs, indicating a potential role for α7 nAChRs in ATS substance-use disorders.
Collapse
Affiliation(s)
- Daniel R Garton
- Columbia College of Columbia University, New York, NY, 10027, USA
| | - Sharmaine G Ross
- Department of Biobehavioral Sciences, Teachers College Columbia University, New York, NY, 10027, USA
| | | | - Matthias Quick
- Department of Psychiatry, Molecular Therapeutics Division, Columbia University Medical Center, New York, NY, 10032, USA
| | - José A Lasalde-Dominicci
- Departments of Biology and Chemistry, University of Puerto Rico, Río Piedras Campus, San Juan, PR, 00931, USA; Molecular Sciences Research Center, University of Puerto Rico, San Juan, PR, 00927, USA
| | - José E Lizardi-Ortiz
- Molecular Sciences Research Center, University of Puerto Rico, San Juan, PR, 00927, USA; Departments of Neurology and Psychiatry, Columbia University Medical Center, New York, NY, 10032, USA.
| |
Collapse
|
234
|
Abstract
Methamphetamine has the second highest prevalence of drug abuse after cannabis, with estimates of 35 million users worldwide. The ( S)-(+)-enantiomer is the illicit drug, active neurostimulant, and eutomer, while the ( R)-(-)-enantiomer is contained in over the counter decongestants. While designated a schedule II drug in 1970, ( S)-(+)-methamphetamine is available by prescription for the treatment of attention-deficit disorder and obesity. The illicit use of ( S)-(+)-methamphetamine results in the sudden "rush" of stimulation to the motivation, movement, pleasure, and reward centers in the brain, caused by rapid release of dopamine. In this review, we will provide an overview of the synthesis, pharmacology, adverse effects, and drug metabolism of this widely abused psychostimulant that distinguish it as a DARK classic in Chemical Neuroscience.
Collapse
Affiliation(s)
- Thomas J. Abbruscato
- Department of Pharmaceutical Sciences, School of Pharmacy, Texas Tech University Health Sciences Center, Amarillo, Texas 79106, United States
| | - Paul C. Trippier
- Department of Pharmaceutical Sciences, School of Pharmacy, Texas Tech University Health Sciences Center, Amarillo, Texas 79106, United States
| |
Collapse
|
235
|
Schneier FR, Slifstein M, Whitton AE, Pizzagalli DA, Reinen J, McGrath PJ, Iosifescu DV, Abi-Dargham A. Dopamine Release in Antidepressant-Naive Major Depressive Disorder: A Multimodal [ 11C]-(+)-PHNO Positron Emission Tomography and Functional Magnetic Resonance Imaging Study. Biol Psychiatry 2018; 84:563-573. [PMID: 30041971 PMCID: PMC6347467 DOI: 10.1016/j.biopsych.2018.05.014] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/14/2018] [Revised: 05/02/2018] [Accepted: 05/15/2018] [Indexed: 12/22/2022]
Abstract
BACKGROUND Mesolimbic dopamine system dysfunction is believed to contribute to major depressive disorder (MDD), but molecular neuroimaging of striatal dopamine neurotransmission has yielded mixed results, possibly owing to limited sensitivity of antagonist radioligands used with positron emission tomography to assess dopamine release capacity. This study used an agonist radioligand with agonist challenge to assess dopamine release capacity and D2/D3 receptor availability in MDD. METHODS Twenty-six treatment-naive adults with MDD and 26 healthy comparison participants underwent functional magnetic resonance imaging during a probabilistic reinforcement task, and positron emission tomography with the D3-preferring ligand [11C]-(+)-PHNO, before and after oral dextroamphetamine. MDD participants then received pramipexole treatment for 6 weeks. RESULTS MDD participants had trend-level greater dopamine release capacity in the ventral striatum, as measured by percent change in baseline binding potential relative to nondisplaceable compartment (ΔBPND) (-34% vs. -30%; p = .072, d = 0.58) but no difference in D2/D3 receptor availability (BPND). Striatal and extrastriatal BPND and percent change in baseline BPND were not significantly associated with blood oxygen level-dependent response to reward prediction error in the ventral striatum, severity of depression and anhedonia, or antidepressant response to pramipexole (response rate = 72.7%). CONCLUSIONS [11C]-(+)-PHNO demonstrated high sensitivity to displacement by amphetamine-induced dopamine release, but dopamine release capacity and D2/D3 availability were not associated with ventral striatal activation to reward prediction error or clinical features, in this study powered to detect large effects. While a preponderance of indirect evidence implicates dopaminergic dysfunction in MDD, these findings suggest that presynaptic dopamine dysregulation may not be a feature of MDD or a prerequisite for treatment response to dopamine agonists.
Collapse
Affiliation(s)
- Franklin R Schneier
- Division of Clinical Therapeutics, New York State Psychiatric Institute, Columbia University Medical Center, New York, New York; Department of Psychiatry, Columbia University Medical Center, New York, New York.
| | - Mark Slifstein
- Division of Translational Imaging, New York State Psychiatric Institute, Columbia University Medical Center, New York, New York; Department of Psychiatry, Columbia University Medical Center, New York, New York
| | - Alexis E Whitton
- Center for Depression, Anxiety and Stress Research, McLean Hospital, Belmont; Department of Psychiatry, Harvard Medical School, Cambridge, Massachusetts
| | - Diego A Pizzagalli
- Center for Depression, Anxiety and Stress Research, McLean Hospital, Belmont; Department of Psychiatry, Harvard Medical School, Cambridge, Massachusetts
| | - Jenna Reinen
- Department of Psychology, Columbia University Medical Center, New York, New York; Department of Psychology, Yale University, New Haven, Connecticut
| | - Patrick J McGrath
- Division of Clinical Therapeutics, New York State Psychiatric Institute, Columbia University Medical Center, New York, New York; Department of Psychiatry, Columbia University Medical Center, New York, New York
| | - Dan V Iosifescu
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, New York
| | - Anissa Abi-Dargham
- Division of Translational Imaging, New York State Psychiatric Institute, Columbia University Medical Center, New York, New York; Department of Psychiatry, Columbia University Medical Center, New York, New York
| |
Collapse
|
236
|
Shahidi S, Sadeghian R, Komaki A, Asl SS. Intracerebroventricular microinjection of the 5-HT 1F receptor agonist LY 344864 inhibits methamphetamine conditioned place preference reinstatement in rats. Pharmacol Biochem Behav 2018; 173:27-35. [PMID: 30077744 DOI: 10.1016/j.pbb.2018.08.001] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/22/2018] [Revised: 07/14/2018] [Accepted: 08/01/2018] [Indexed: 01/01/2023]
Abstract
Relapse following a prolonged period of drug cessation is a key barrier in the treatment of methamphetamine (METH) addiction, for which pharmacological treatment exhibits little efficacy. Previous studies have suggested that this process involves alterations in levels of serotonin (5-HT) in the brain. Although the 5-HT1F receptor has been implicated in the reward pathway, its physiological functions remain unknown. In the present study, we examined the effect of the 5-HT1F agonist LY 344864 on the reinstatement of METH-seeking behavior in rats using a conditioned place preference (CPP) paradigm. The CPP paradigm was first used to determine the effective doses of LY and METH. Four groups were then conditioned with METH (5 mg/kg; i.p.), while the sham group received saline. METH-induced CPP was subsequently extinguished. On the 13th day of extinction, the rats received either METH (0, 1, or 2.5 mg/kg; i.p.) plus vehicle or priming METH plus LY (2 μg/5 μL; i.c.v.). On reinstatement day, preference scores were calculated as the difference in time spent in the drug-paired and vehicle-paired compartments. Rats conditioned with the lowest effective dose of METH (5 mg/kg) exhibited significant differences in pre- and post-testing preference scores. Preference scores were significantly higher in the saline + METH group than in the control group. Furthermore, preference scores were significantly higher in rats that had received priming METH treatment, and pre-treatment with LY significantly attenuated the reinstatement of METH-seeking behavior. These findings suggest that future studies should evaluate the therapeutic potential of 5-HT1F agonists for preventing relapse in individuals with METH addiction.
Collapse
Affiliation(s)
- Siamak Shahidi
- Neurophysiology Research Center, Hamadan University of Medical Sciences, Hamadan, Iran
| | - Reihaneh Sadeghian
- Neurophysiology Research Center, Hamadan University of Medical Sciences, Hamadan, Iran.
| | - Alireza Komaki
- Neurophysiology Research Center, Hamadan University of Medical Sciences, Hamadan, Iran
| | - Sara Soleimani Asl
- Anatomy Departments, School of Medicine, Hamadan University of Medical Sciences, Hamadan, Iran
| |
Collapse
|
237
|
Essmann CL, Ryan KR, Elmi M, Bryon-Dodd K, Porter A, Vaughan A, McMullan R, Nurrish S. Activation of RHO-1 in cholinergic motor neurons competes with dopamine signalling to control locomotion. PLoS One 2018; 13:e0204057. [PMID: 30240421 PMCID: PMC6150489 DOI: 10.1371/journal.pone.0204057] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2018] [Accepted: 08/31/2018] [Indexed: 12/14/2022] Open
Abstract
The small GTPase RhoA plays a crucial role in the regulation of neuronal signalling to generate behaviour. In the developing nervous system RhoA is known to regulate the actin cytoskeleton, however the effectors of RhoA-signalling in adult neurons remain largely unidentified. We have previously shown that activation of the RhoA ortholog (RHO-1) in C. elegans cholinergic motor neurons triggers hyperactivity of these neurons and loopy locomotion with exaggerated body bends. This is achieved in part through increased diacylglycerol (DAG) levels and the recruitment of the synaptic vesicle protein UNC-13 to synaptic release sites, however other pathways remain to be identified. Dopamine, which is negatively regulated by the dopamine re-uptake transporter (DAT), has a central role in modulating locomotion in both humans and C. elegans. In this study we identify a new pathway in which RHO-1 regulates locomotory behaviour by repressing dopamine signalling, via DAT-1, linking these two pathways together. We observed an upregulation of dat-1 expression when RHO-1 is activated and show that loss of DAT-1 inhibits the loopy locomotion phenotype caused by RHO-1 activation. Reducing dopamine signalling in dat-1 mutants through mutations in genes involved in dopamine synthesis or in the dopamine receptor DOP-1 restores the ability of RHO-1 to trigger loopy locomotion in dat-1 mutants. Taken together, we show that negative regulation of dopamine signalling via DAT-1 is necessary for the neuronal RHO-1 pathway to regulate locomotion.
Collapse
Affiliation(s)
- Clara L. Essmann
- MRC Laboratory for Molecular Cell Biology, University College London, London, United Kingdom
| | - Katie R. Ryan
- MRC Laboratory for Molecular Cell Biology, University College London, London, United Kingdom
| | - Muna Elmi
- MRC Laboratory for Molecular Cell Biology, University College London, London, United Kingdom
| | - Kimberley Bryon-Dodd
- MRC Laboratory for Molecular Cell Biology, University College London, London, United Kingdom
| | - Andrew Porter
- MRC Laboratory for Molecular Cell Biology, University College London, London, United Kingdom
| | - Andrew Vaughan
- MRC Laboratory for Molecular Cell Biology, University College London, London, United Kingdom
| | - Rachel McMullan
- MRC Laboratory for Molecular Cell Biology, University College London, London, United Kingdom
| | - Stephen Nurrish
- MRC Laboratory for Molecular Cell Biology, University College London, London, United Kingdom
| |
Collapse
|
238
|
Carias E, Hamilton J, Robison LS, Delis F, Eiden R, Quattrin T, Hadjiargyrou M, Komatsu D, Thanos PK. Chronic oral methylphenidate treatment increases microglial activation in rats. J Neural Transm (Vienna) 2018; 125:1867-1875. [PMID: 30238340 DOI: 10.1007/s00702-018-1931-z] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2018] [Accepted: 09/17/2018] [Indexed: 12/18/2022]
Abstract
Methylphenidate (MP) is a widely prescribed psychostimulant used to treat attention deficit hyperactivity disorder. Previously, we established a drinking paradigm to deliver MP to rats at doses that result in pharmacokinetic profiles similar to treated patients. In the present study, adolescent male rats were assigned to one of three groups: control (water), low-dose MP (LD; 4/10 mg/kg), and high dose MP (HD; 30/60 mg/kg). Following 3 months of treatment, half of the rats in each group were euthanized, and the remaining rats received only water throughout a 1-month-long abstinence phase. In vitro autoradiography using [3H] PK 11195 was performed to measure microglial activation. HD MP rats showed increased [3H] PK 11195 binding compared to control rats in several cerebral cortical areas: primary somatosensory cortex including jaw (68.6%), upper lip (80.1%), barrel field (88.9%), and trunk (78%) regions, forelimb sensorimotor area (87.3%), secondary somatosensory cortex (72.5%), motor cortices 1 (73.2%) and 2 (69.3%), insular cortex (59.9%); as well as subcortical regions including the thalamus (62.9%), globus pallidus (79.4%) and substantia nigra (22.7%). Additionally, HD MP rats showed greater binding compared to LD MP rats in the hippocampus (60.6%), thalamus (59.6%), substantia nigra (38.5%), and motor 2 cortex (55.3%). Following abstinence, HD MP rats showed no significant differences compared to water controls; however, LD MP rats showed increased binding in pre-limbic cortex (78.1%) and ventromedial caudate putamen (113.8%). These findings indicate that chronic MP results in widespread microglial activation immediately after treatment and following the cessation of treatment in some brain regions.
Collapse
Affiliation(s)
- Emily Carias
- Behavioral Neuropharmacology and Neuroimaging Laboratory on Addictions (BNNLA), Research Institute on Addictions, Department of Pharmacology and Toxicology, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, 1021 Main St., Buffalo, NY, USA
| | - John Hamilton
- Behavioral Neuropharmacology and Neuroimaging Laboratory on Addictions (BNNLA), Research Institute on Addictions, Department of Pharmacology and Toxicology, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, 1021 Main St., Buffalo, NY, USA
| | - Lisa S Robison
- Department of Neuroscience and Experimental Therapeutics, Albany Medical College, 47 New Scotland Ave., Albany, NY, USA
| | - Foteini Delis
- Department of Pharmacology, Medical School, University of Ioannina, 45110, Ioannina, Greece
| | - Rina Eiden
- Department of Psychology, University at Buffalo, 1021 Main St., Buffalo, NY, USA
| | - Teresa Quattrin
- Women and Children's Hospital of Buffalo, Department of Pediatrics, School of Medicine and Biomedical Sciences, University at Buffalo, Buffalo, NY, USA
| | - Michael Hadjiargyrou
- Department of Life Sciences, New York Institute of Technology, Northern Blvd., Old Westbury, NY, USA
| | - David Komatsu
- Department of Orthopedics, Stony Brook University, 100 Nicolls Rd., Stony Brook, NY, USA
| | - Panayotis K Thanos
- Behavioral Neuropharmacology and Neuroimaging Laboratory on Addictions (BNNLA), Research Institute on Addictions, Department of Pharmacology and Toxicology, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, 1021 Main St., Buffalo, NY, USA.
| |
Collapse
|
239
|
Schmidt U, Flössel U, Pietsch J, Dinger J, Engel A, Forberger A, Nitzsche K. Intrauterine und perinatale Todesfälle bei maternalem Methamphetaminkonsum. Rechtsmedizin (Berl) 2018. [DOI: 10.1007/s00194-018-0269-z] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
|
240
|
Manchado A, García M, Salgado MM, Díez D, Garrido NM. A novel Barton decarboxylation produces a 1,4-phenyl radical rearrangement domino reaction. Tetrahedron 2018. [DOI: 10.1016/j.tet.2018.05.043] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/16/2022]
|
241
|
Hovde MJ, Larson GH, Vaughan RA, Foster JD. Model systems for analysis of dopamine transporter function and regulation. Neurochem Int 2018; 123:13-21. [PMID: 30179648 DOI: 10.1016/j.neuint.2018.08.015] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2018] [Revised: 08/23/2018] [Accepted: 08/31/2018] [Indexed: 02/07/2023]
Abstract
The dopamine transporter (DAT) plays a critical role in dopamine (DA) homeostasis by clearing transmitter from the extraneuronal space after vesicular release. DAT serves as a site of action for a variety of addictive and therapeutic reuptake inhibitors, and transport dysfunction is associated with transmitter imbalances in disorders such as schizophrenia, attention deficit hyperactive disorder, bipolar disorder, and Parkinson disease. In this review, we describe some of the model systems that have been used for in vitro analyses of DAT structure, function and regulation, and discuss a potential relationship between transporter kinetic values and membrane cholesterol.
Collapse
Affiliation(s)
- Moriah J Hovde
- Department of Biomedical Sciences, University of North Dakota, School of Medicine and Health Sciences, Grand Forks, ND, 58202, USA
| | - Garret H Larson
- Department of Biomedical Sciences, University of North Dakota, School of Medicine and Health Sciences, Grand Forks, ND, 58202, USA
| | - Roxanne A Vaughan
- Department of Biomedical Sciences, University of North Dakota, School of Medicine and Health Sciences, Grand Forks, ND, 58202, USA
| | - James D Foster
- Department of Biomedical Sciences, University of North Dakota, School of Medicine and Health Sciences, Grand Forks, ND, 58202, USA.
| |
Collapse
|
242
|
Tomlinson MJ, Krout D, Pramod AB, Lever JR, Newman AH, Henry LK, Vaughan RA. Identification of the benztropine analog [ 125I]GA II 34 binding site on the human dopamine transporter. Neurochem Int 2018; 123:34-45. [PMID: 30125594 DOI: 10.1016/j.neuint.2018.08.008] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2018] [Revised: 08/10/2018] [Accepted: 08/14/2018] [Indexed: 12/21/2022]
Abstract
The dopamine transporter (DAT) is a neuronal membrane protein that is responsible for reuptake of dopamine (DA) from the synapse and functions as a major determinant in control of DA neurotransmission. Cocaine and many psychostimulant drugs bind to DAT and block reuptake, inducing DA overflow that forms the neurochemical basis for euphoria and addiction. Paradoxically, however, some ligands such as benztropine (BZT) bind to DAT and inhibit reuptake but do not produce these effects, and it has been hypothesized that differential mechanisms of binding may stabilize specific transporter conformations that affect downstream neurochemical or behavioral outcomes. To investigate the binding mechanisms of BZT on DAT we used the photoaffinity BZT analog [125I]N-[n-butyl-4-(4‴-azido-3‴-iodophenyl)]-4',4″-difluoro-3α-(diphenylmethoxy)tropane ([125I]GA II 34) to identify the site of cross-linking and predict the binding pose relative to that of previously-examined cocaine photoaffinity analogs. Biochemical findings show that adduction of [125I]GA II 34 occurs at residues Asp79 or Leu80 in TM1, with molecular modeling supporting adduction to Leu80 and a pharmacophore pose in the central S1 site similar to that of cocaine and cocaine analogs. Substituted cysteine accessibility method protection analyses verified these findings, but identified some differences in structural stabilization relative to cocaine that may relate to BZT neurochemical outcomes.
Collapse
Affiliation(s)
- Michael J Tomlinson
- Department of Biomedical Sciences, University of North Dakota School of Medicine and Health Sciences, 1301 North Columbia Road, Grand Forks, ND 58202, United States
| | - Danielle Krout
- Department of Biomedical Sciences, University of North Dakota School of Medicine and Health Sciences, 1301 North Columbia Road, Grand Forks, ND 58202, United States
| | - Akula Bala Pramod
- Department of Biomedical Sciences, University of North Dakota School of Medicine and Health Sciences, 1301 North Columbia Road, Grand Forks, ND 58202, United States
| | - John R Lever
- Research Service, Harry S. Truman Memorial Veterans' Hospital, Columbia, MO, 65201, United States; Department of Radiology and Radiopharmaceutical Sciences Institute, University of Missouri, Columbia, MO 65211, United States
| | - Amy Hauck Newman
- Medicinal Chemistry Section, National Institute on Drug Abuse - Intramural Research Program, Baltimore, MD 21224, United States
| | - L Keith Henry
- Department of Biomedical Sciences, University of North Dakota School of Medicine and Health Sciences, 1301 North Columbia Road, Grand Forks, ND 58202, United States.
| | - Roxanne A Vaughan
- Department of Biomedical Sciences, University of North Dakota School of Medicine and Health Sciences, 1301 North Columbia Road, Grand Forks, ND 58202, United States.
| |
Collapse
|
243
|
Castells X, Blanco‐Silvente L, Cunill R. Amphetamines for attention deficit hyperactivity disorder (ADHD) in adults. Cochrane Database Syst Rev 2018; 8:CD007813. [PMID: 30091808 PMCID: PMC6513464 DOI: 10.1002/14651858.cd007813.pub3] [Citation(s) in RCA: 47] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
BACKGROUND Attention deficit hyperactivity disorder (ADHD) is a childhood-onset disorder characterised by inattention, hyperactivity, and impulsivity. ADHD can persist into adulthood and can affects individuals' social and occupational functioning, as well as their quality of life and health. ADHD is frequently associated with other mental disorders such as substance use disorders and anxiety and affective disorders. Amphetamines are used to treat adults with ADHD, but uncertainties about their efficacy and safety remain. OBJECTIVES To examine the efficacy and safety of amphetamines for adults with ADHD. SEARCH METHODS In August 2017, we searched CENTRAL, MEDLINE, Embase, PsycINFO, 10 other databases, and two trials registers, and we ran citation searches for included studies. We also contacted the corresponding authors of all included studies, other experts in the field, and the pharmaceutical company, Shire, and we searched the reference lists of retrieved studies and reviews for other published, unpublished, or ongoing studies. For each included study, we performed a citation search in Web of Science to identify any later studies that may have cited it. SELECTION CRITERIA We searched for randomised controlled trials comparing the efficacy of amphetamines (at any dose) for ADHD in adults aged 18 years and over against placebo or an active intervention. DATA COLLECTION AND ANALYSIS Two review authors extracted data from each included study. We used the standardised mean difference (SMD) and the risk ratio (RR) to assess continuous and dichotomous outcomes, respectively. We conducted a stratified analysis to determine the influence of moderating variables. We assessed trials for risk of bias and drew a funnel plot to investigate the possibility of publication bias. We rated the quality of the evidence using the GRADE approach, which yielded high, moderate, low, or very low quality ratings based on evaluation of within-trial risk of bias, directness of evidence, heterogeneity of data; precision of effect estimates, and risk of publication bias. MAIN RESULTS We included 19 studies that investigated three types of amphetamines: dexamphetamine (10.2 mg/d to 21.8 mg/d), lisdexamfetamine (30 mg/d to 70 mg/d), and mixed amphetamine salts (MAS; 12.5 mg/d to 80 mg/d). These studies enrolled 2521 participants; most were middle-aged (35.3 years), Caucasian males (57.2%), with a combined type of ADHD (78.8%). Eighteen studies were conducted in the USA, and one study was conducted in both Canada and the USA. Ten were multi-site studies. All studies were placebo-controlled, and three also included an active comparator: guanfacine, modafinil, or paroxetine. Most studies had short-term follow-up and a mean study length of 5.3 weeks.We found no studies that had low risk of bias in all domains of the Cochrane 'Risk of bias' tool, mainly because amphetamines have powerful subjective effects that may reveal the assigned treatment, but also because we noted attrition bias, and because we could not rule out the possibility of a carry-over effect in studies that used a cross-over design.Sixteen studies were funded by the pharmaceutical industry, one study was publicly funded, and two studies did not report their funding sources.Amphetamines versus placeboSeverity of ADHD symptoms: we found low- to very low-quality evidence suggesting that amphetamines reduced the severity of ADHD symptoms as rated by clinicians (SMD -0.90, 95% confidence interval (CI) -1.04 to -0.75; 13 studies, 2028 participants) and patients (SMD -0.51, 95% CI -0.75 to -0.28; six studies, 120 participants).Retention: overall, we found low-quality evidence suggesting that amphetamines did not improve retention in treatment (risk ratio (RR) 1.06, 95% CI 0.99 to 1.13; 17 studies, 2323 participants).Adverse events: we found that amphetamines were associated with an increased proportion of patients who withdrew because of adverse events (RR 2.69, 95% CI 1.63 to 4.45; 17 studies, 2409 participants).Type of amphetamine: we found differences between amphetamines for the severity of ADHD symptoms as rated by clinicians. Both lisdexamfetamine (SMD -1.06, 95% CI -1.26 to -0.85; seven studies, 896 participants; low-quality evidence) and MAS (SMD -0.80, 95% CI -0.93 to -0.66; five studies, 1083 participants; low-quality evidence) reduced the severity of ADHD symptoms. In contrast, we found no evidence to suggest that dexamphetamine reduced the severity of ADHD symptoms (SMD -0.24, 95% CI -0.80 to 0.32; one study, 49 participants; very low-quality evidence). In addition, all amphetamines were efficacious in reducing the severity of ADHD symptoms as rated by patients (dexamphetamine: SMD -0.77, 95% CI -1.14 to -0.40; two studies, 35 participants; low-quality evidence; lisdexamfetamine: SMD -0.33, 95% CI -0.65 to -0.01; three studies, 67 participants; low-quality evidence; MAS: SMD -0.45, 95% CI -1.02 to 0.12; one study, 18 participants; very low-quality evidence).Dose at study completion: different doses of amphetamines did not appear to be associated with differences in efficacy.Type of drug-release formulation: we investigated immediate- and sustained-release formulations but found no differences between them for any outcome.Amphetamines versus other drugsWe found no evidence that amphetamines improved ADHD symptom severity compared to other drug interventions. AUTHORS' CONCLUSIONS Amphetamines improved the severity of ADHD symptoms, as assessed by clinicians or patients, in the short term but did not improve retention to treatment. Amphetamines were associated with higher attrition due to adverse events. The short duration of studies coupled with their restrictive inclusion criteria limits the external validity of these findings. Furthermore, none of the included studies had an overall low risk of bias. Overall, the evidence generated by this review is of low or very low quality.
Collapse
Affiliation(s)
- Xavier Castells
- Universitat de GironaUnit of Clinical Pharmacology, TransLab Research Group, Department of Medical SciencesEmili Grahit, 77GironaCataloniaSpain17071
| | - Lídia Blanco‐Silvente
- Universitat de GironaUnit of Clinical Pharmacology, TransLab Research Group, Department of Medical SciencesEmili Grahit, 77GironaCataloniaSpain17071
| | - Ruth Cunill
- Parc Sanitari Sant Joan de DéuParc Sanitari Sant Joan de Déu ‐ NumanciaBarcelonaCatalunyaSpain08735
| | | |
Collapse
|
244
|
Shahidi S, Komaki A, Sadeghian R, Soleimani Asl S. Effect of a 5-HT 1D receptor agonist on the reinstatement phase of the conditioned place preference test and hippocampal long-term potentiation in methamphetamine-treated rats. Brain Res 2018; 1698:151-160. [PMID: 30076792 DOI: 10.1016/j.brainres.2018.07.030] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2018] [Revised: 07/26/2018] [Accepted: 07/31/2018] [Indexed: 12/20/2022]
Abstract
Methamphetamine (METH)-seeking relapse is associated with memory and synaptic plasticity changes. Serotonin is a key neuromodulator in this process. While there is a known distribution of 5-HT1D receptors in reward and memory areas, such as the hippocampus, its physiological function is currently unknown. Here, we evaluated effect of a 5-HT1D receptor agonist, PNU142633, on the reinstatement of METH-seeking behavior and long-term potentiation. Rats were implanted with a cannula into lateral ventricle, then treated with saline or METH (5 mg/kg) during the acquisition phase of the conditioned place preference (CPP) test. On day 13 of the extinction phase, METH groups were divided into four groups: METH (0: saline, 1, or 2.5 (priming METH) mg/kg; i.p.) + vehicle (5 µl/rat) or a priming dose of METH (2.5 mg/kg; i.p.) + PNU (2 µg/5 µl; i.c.v.) and their preference scores were calculated on reinstatement day (day 14). Immediately following this, electrophysiology was performed to assay the field excitatory postsynaptic potential (fEPSP) slope and population spike (PS) amplitude between groups. The results showed that CPP induction by METH gradually declined to extinction on days 12 and 13. A priming METH treatment significantly increased preference for the METH-paired chamber when compared with other groups, but pre-treatment with PNU significantly attenuated this effect. PS amplitude and fEPSP slopes in vehicle + priming METH rats were greater when compared with other groups. Furthermore, PNU attenuated the priming METH-induced increase in PS amplitude. These findings suggest that PNU can decrease synaptic transmission and prevent METH reinstatement in rats.
Collapse
Affiliation(s)
- Siamak Shahidi
- Neurophysiology Research Center, Hamadan University of Medical Sciences, Hamadan, Iran
| | - Alireza Komaki
- Neurophysiology Research Center, Hamadan University of Medical Sciences, Hamadan, Iran
| | - Reihaneh Sadeghian
- Neurophysiology Research Center, Hamadan University of Medical Sciences, Hamadan, Iran.
| | - Sara Soleimani Asl
- Anatomy Departments, School of Medicine, Hamadan University of Medical Sciences, Hamadan, Iran
| |
Collapse
|
245
|
Chronic stress sensitizes amphetamine-elicited 50-kHz calls in the rat: Dependence on positive affective phenotype and effects of long-term fluoxetine pretreatment. Pharmacol Biochem Behav 2018; 171:10-19. [DOI: 10.1016/j.pbb.2018.05.014] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/27/2018] [Accepted: 05/23/2018] [Indexed: 11/17/2022]
|
246
|
Struntz KH, Siegel JA. Effects of methamphetamine exposure on anxiety-like behavior in the open field test, corticosterone, and hippocampal tyrosine hydroxylase in adolescent and adult mice. Behav Brain Res 2018; 348:211-218. [DOI: 10.1016/j.bbr.2018.04.019] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2018] [Revised: 04/09/2018] [Accepted: 04/14/2018] [Indexed: 12/28/2022]
|
247
|
Epigenetic Effects Induced by Methamphetamine and Methamphetamine-Dependent Oxidative Stress. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2018; 2018:4982453. [PMID: 30140365 PMCID: PMC6081569 DOI: 10.1155/2018/4982453] [Citation(s) in RCA: 47] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/19/2018] [Accepted: 06/10/2018] [Indexed: 12/21/2022]
Abstract
Methamphetamine is a widely abused drug, which possesses neurotoxic activity and powerful addictive effects. Understanding methamphetamine toxicity is key beyond the field of drug abuse since it allows getting an insight into the molecular mechanisms which operate in a variety of neuropsychiatric disorders. In fact, key alterations produced by methamphetamine involve dopamine neurotransmission in a way, which is reminiscent of spontaneous neurodegeneration and psychiatric schizophrenia. Thus, understanding the molecular mechanisms operated by methamphetamine represents a wide window to understand both the addicted brain and a variety of neuropsychiatric disorders. This overlapping, which is already present when looking at the molecular and cellular events promoted immediately after methamphetamine intake, becomes impressive when plastic changes induced in the brain of methamphetamine-addicted patients are considered. Thus, the present manuscript is an attempt to encompass all the molecular events starting at the presynaptic dopamine terminals to reach the nucleus of postsynaptic neurons to explain how specific neurotransmitters and signaling cascades produce persistent genetic modifications, which shift neuronal phenotype and induce behavioral alterations. A special emphasis is posed on disclosing those early and delayed molecular events, which translate an altered neurotransmitter function into epigenetic events, which are derived from the translation of postsynaptic noncanonical signaling into altered gene regulation. All epigenetic effects are considered in light of their persistent changes induced in the postsynaptic neurons including sensitization and desensitization, priming, and shift of neuronal phenotype.
Collapse
|
248
|
Dunn M, Henke A, Clark S, Kovalyova Y, Kempadoo KA, Karpowicz RJ, Kandel ER, Sulzer D, Sames D. Designing a norepinephrine optical tracer for imaging individual noradrenergic synapses and their activity in vivo. Nat Commun 2018; 9:2838. [PMID: 30026491 PMCID: PMC6053466 DOI: 10.1038/s41467-018-05075-x] [Citation(s) in RCA: 41] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2017] [Accepted: 05/11/2018] [Indexed: 11/14/2022] Open
Abstract
Norepinephrine is a monoamine neurotransmitter with a wide repertoire of physiological roles in the peripheral and central nervous systems. There are, however, no experimental means to study functional properties of individual noradrenergic synapses in the brain. Development of new approaches for imaging synaptic neurotransmission is of fundamental importance to study specific synaptic changes that occur during learning, behavior, and pathological processes. Here, we introduce fluorescent false neurotransmitter 270 (FFN270), a fluorescent tracer of norepinephrine. As a fluorescent substrate of the norepinephrine and vesicular monoamine transporters, FFN270 labels noradrenergic neurons and their synaptic vesicles, and enables imaging synaptic vesicle content release from specific axonal sites in living rodents. Combining FFN270 imaging and optogenetic stimulation, we find heterogeneous release properties of noradrenergic synapses in the somatosensory cortex, including low and high releasing populations. Through systemic amphetamine administration, we observe rapid release of cortical noradrenergic vesicular content, providing insight into the drug’s effect. The noradrenergic system plays numerous physiological roles but tools to study it are scarce. Here the authors develop a fluorescent analogue of norepinephrine that can be used to label noradrenergic neurons and the synaptic vesicles, and use it to measure single synaptic vesicle release sites in living mice.
Collapse
Affiliation(s)
- Matthew Dunn
- Department of Chemistry, Columbia University, New York, NY, 10027, USA
| | - Adam Henke
- Department of Chemistry, Columbia University, New York, NY, 10027, USA
| | - Samuel Clark
- Department of Neurology, Columbia University, New York, NY, 10032, USA.,Department of Psychiatry, Columbia University, New York, NY, 10032, USA.,Department of Pharmacology, Columbia University, New York, NY, 10032, USA
| | | | | | | | - Eric R Kandel
- Department of Psychiatry, Columbia University, New York, NY, 10032, USA.,Department of Neuroscience, Columbia University, New York, NY, 10032, USA.,Kavli Institute for Brain Science, New York, NY, 10032, USA.,Howard Hughes Medical Institute, New York, NY, 10032, USA
| | - David Sulzer
- Department of Neurology, Columbia University, New York, NY, 10032, USA. .,Department of Psychiatry, Columbia University, New York, NY, 10032, USA. .,Department of Pharmacology, Columbia University, New York, NY, 10032, USA.
| | - Dalibor Sames
- Department of Chemistry, Columbia University, New York, NY, 10027, USA.
| |
Collapse
|
249
|
Antipova V, Wree A, Holzmann C, Mann T, Palomero-Gallagher N, Zilles K, Schmitt O, Hawlitschka A. Unilateral Botulinum Neurotoxin-A Injection into the Striatum of C57BL/6 Mice Leads to a Different Motor Behavior Compared with Rats. Toxins (Basel) 2018; 10:E295. [PMID: 30018211 PMCID: PMC6070800 DOI: 10.3390/toxins10070295] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2018] [Revised: 07/13/2018] [Accepted: 07/15/2018] [Indexed: 12/26/2022] Open
Abstract
Different morphological changes in the caudate-putamen (CPu) of naïve rats and mice were observed after intrastriatal botulinum neurotoxin-A (BoNT-A) injection. For this purpose we here studied various motor behaviors in mice (n = 46) longitudinally up to 9 months after intrastriatal BoNT-A administration as previously reported for rats, and compared both outcomes. Apomorphine- and amphetamine-induced rotational behavior, spontaneous motor behavior, as well as lateralized neglect were studied in mice after the injection of single doses of BoNT-A into the right CPu, comparing them with sham-injected animals. Unilateral intrastriatal injection of BoNT-A in mice induced significantly increased contralateral apomorphine-induced rotations for 1 to 3 months, as well as significantly increased contralateral amphetamine-induced rotations 1 to 9 months after injection. In rats (n = 28), unilateral BoNT-A injection also induced significantly increased contralateral apomorphine-induced rotations 3 months after injection, but did not provoke amphetamine-induced rotations at all. Lateralized sensorimotor integration, forelimb preference, and forelimb stepping were significantly impaired on the left side. The differences in motor behaviors between rats and mice may be caused by different BoNT-A effects on cholinergic and catecholaminergic fibers in rat and mouse striata, interspecies differences in striatal receptor densities, and different connectomes of the basal ganglia.
Collapse
Affiliation(s)
- Veronica Antipova
- Institute of Anatomy, Rostock University Medical Center, Gertrudenstrasse 9, D-18057 Rostock, Germany.
- Gottfried Schatz Research Center for Cell Signaling, Metabolism and Aging, Macroscopic and Clinical Anatomy, Medical University of Graz, Harrachgasse 21/1, A-8010 Graz, Austria.
| | - Andreas Wree
- Institute of Anatomy, Rostock University Medical Center, Gertrudenstrasse 9, D-18057 Rostock, Germany.
| | - Carsten Holzmann
- Institute of Medical Genetics, Rostock University Medical Center, Ernst-Heydemann-Strasse 8, D-18057 Rostock, Germany.
| | - Teresa Mann
- Institute of Anatomy, Rostock University Medical Center, Gertrudenstrasse 9, D-18057 Rostock, Germany.
| | - Nicola Palomero-Gallagher
- Institute of Neuroscience and Medicine INM-1, Research Center Jülich, D-52425 Jülich, Germany.
- Department of Psychiatry, Psychotherapy and Psychosomatics, Medical Faculty, RWTH Aachen, D-52062 Aachen, Germany.
| | - Karl Zilles
- Institute of Neuroscience and Medicine INM-1, Research Center Jülich, D-52425 Jülich, Germany.
- Department of Psychiatry, Psychotherapy and Psychosomatics, Medical Faculty, RWTH Aachen, D-52062 Aachen, Germany.
- JARA-Translational Brain Medicine, D-52062 Aachen, Germany.
| | - Oliver Schmitt
- Institute of Anatomy, Rostock University Medical Center, Gertrudenstrasse 9, D-18057 Rostock, Germany.
| | - Alexander Hawlitschka
- Institute of Anatomy, Rostock University Medical Center, Gertrudenstrasse 9, D-18057 Rostock, Germany.
| |
Collapse
|
250
|
Kitanaka N, Kitanaka J, Hall FS, Kandori T, Murakami A, Muratani K, Nakano T, Uhl GR, Takemura M. Tetrabenazine, a vesicular monoamine transporter-2 inhibitor, attenuates morphine-induced hyperlocomotion in mice through alteration of dopamine and 5-hydroxytryptamine turnover in the cerebral cortex. Pharmacol Biochem Behav 2018; 172:9-16. [PMID: 30017858 DOI: 10.1016/j.pbb.2018.07.002] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/11/2018] [Revised: 07/10/2018] [Accepted: 07/11/2018] [Indexed: 10/28/2022]
Abstract
A single administration with morphine (30 mg/kg, i.p.) induced long-lasting hyperlocomotion in male ICR mice. Pretreatment of mice with a benzoquinolizine derivative tetrabenazine (TBZ; a reversible vesicular monoamine transporter-2 inhibitor) (1 mg/kg, i.p.) for 30 min significantly attenuated the hyperlocomotion induced by morphine, as compared with vehicle (saline)-pretreated mice. No significant change in locomotion was observed in mice pretreated with TBZ (1 mg/kg) alone. Mice treated with TBZ (1 mg/kg) showed an increase in immobility time in a tail suspension test, as compared with saline-treated mice. Pretreatment with TBZ (1 mg/kg) had no effect on morphine (1-30 mg/kg)-induced antinociception. TBZ at a dose of 1 mg/kg inhibited dopamine turnover (the ratio of 3,4-dihydroxyphenylacetic acid/dopamine) and 5-hydroxytryptamine turnover (the ratio of 5-hydroxyindoleacetic acid/5-hydroxytryptamine) in the cerebral cortex of mice challenged with morphine, as compared with saline-pretreated mice challenged with morphine. No stereotypic behavior was observed in mice treated with morphine (30 mg/kg) in combination with TBZ (1 mg/kg), so the reduction in observed locomotion did not result from induction of stereotypical behavior. Moreover, TBZ (1 and 2 mg/kg) pretreatment had no effect on stereotyped behaviors observed in mice challenged with 10 mg/kg methamphetamine. These data support the potential antagonistic actions of TBZ on some opiate actions, and encourage further exploration of potential effects on morphine reinforcement.
Collapse
Affiliation(s)
- Nobue Kitanaka
- Department of Pharmacology, Hyogo College of Medicine, Hyogo 663-8501, Japan
| | - Junichi Kitanaka
- Department of Pharmacology, Hyogo College of Medicine, Hyogo 663-8501, Japan.
| | - F Scott Hall
- Department of Pharmacology and Experimental Therapeutics, College of Pharmacy and Pharmaceutical Sciences, University of Toledo, Toledo, OH 43614, USA
| | - Takashi Kandori
- Department of Pharmacology, Hyogo College of Medicine, Hyogo 663-8501, Japan
| | - Ayaka Murakami
- Department of Pharmacology, Hyogo College of Medicine, Hyogo 663-8501, Japan
| | - Kazuki Muratani
- Department of Pharmacology, Hyogo College of Medicine, Hyogo 663-8501, Japan
| | - Tae Nakano
- Department of Pharmacology, Hyogo College of Medicine, Hyogo 663-8501, Japan
| | - George R Uhl
- New Mexico VA Healthcare System/BRINM, Albuquerque, NM 87108, USA
| | - Motohiko Takemura
- Department of Pharmacology, Hyogo College of Medicine, Hyogo 663-8501, Japan
| |
Collapse
|