Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 37
Filter
1.
Curr Res Neurobiol ; 4: 100088, 2023.
Article in English | MEDLINE | ID: mdl-37397817

ABSTRACT

A view continues to gain momentum that regards investigation of the cognition of great apes in captive settings as affording us a model for human cognitive evolution. Researchers from disciplines such as comparative psychology, anthropology, and even archaeology, seem eager to put their theories to the test by using great apes as their chosen experimental model. Questions addressed currently by comparative psychologists have long been the object of attention by neurophysiologists, psychobiologists and neuroscientists, who, however, often use rodents and monkeys as the species of choice. Whereas comparative psychology has been influenced greatly by ethology, much neuroscience has developed against a background of physiology and medicine. This separation of the intellectual contexts wherein they have arisen and flourished has impeded the development of fluid interaction between comparative psychologists and researchers in the other disciplines. We feel that it would be beneficial for comparative psychologists and neuroscientists to combine research endeavours far more often, in order to address common questions of interest related to cognition. We regard interdisciplinary cross-pollination to be particularly desirable, even if many comparative psychologists lack deep expertise about the workings of the brain, and even if many neuroscientists lack expert knowledge about the behaviour of different species. Furthermore, we believe that anthropology, archaeology, human evolutionary studies, and related disciplines, may well provide us with significant contextual knowledge about the physical and temporal background to the evolution in humans of specific cognitive skills. To that end, we urge researchers to dismantle methodological, conceptual and historical disciplinary boundaries, in order to strengthen cross-disciplinary cooperation in order to broaden and deepen our insights into the cognition of nonhuman and human primates.

2.
J Psychiatr Res ; 150: 246-256, 2022 06.
Article in English | MEDLINE | ID: mdl-35405409

ABSTRACT

Post-traumatic stress disorder (PTSD) develops after an exposure to a life-threatening event and is characterized by intrusive memories. According to memory reconsolidation theory retrieval of memory under certain conditions leads to its labilization and subsequent re-storage which could be disrupted by drugs. Propranolol has been the most commonly investigated drug for memory reconsolidation therapy in clinical trials. Intervention with propranolol have shown mixed results in PTSD patients with some studies showing improvement in symptoms while other failing to replicate these findings. We conducted a systematic review and meta-analysis to determine the efficacy of trauma memory disruption by propranolol on PTSD symptoms and physiological responses in PTSD patients. 3224 publications were assessed for eligibility. Seven studies on effects of propranolol on PTSD symptoms and 3 studies on effects of propranolol on physiological responses were incorporated in the meta-analyses. Overall, results indicate that propranolol did not show a beneficial effect on PTSD symptoms (standardized mean difference: 1.29; 95% CI = -2.16 - 0.17). Similarly, propranolol did not influence skin conductance (standardized mean difference: 0.77; 95% CI = -1.85 - 0.31) or EMG response (standardized mean difference: 0.16; 95% CI = -0.65 - 0.33). However, propranolol significantly reduced heart rate after trauma memory recall compared to placebo (standardized mean difference: 0.67; 95% CI = -1.27 to -0.07). This study finds a lack of evidence for the efficacy of propranolol on traumatic memory disruption, in PTSD patients, to recommend its routine clinical use. However, a high level of heterogeneity, variation in propranolol dosage and inadequate sample sizes mean that these findings require cautious interpretation.


Subject(s)
Propranolol , Stress Disorders, Post-Traumatic , Adrenergic beta-Antagonists/pharmacology , Adrenergic beta-Antagonists/therapeutic use , Fear , Humans , Memory/physiology , Propranolol/pharmacology , Propranolol/therapeutic use , Stress Disorders, Post-Traumatic/drug therapy
3.
Pharmacol Ther ; 239: 108195, 2022 11.
Article in English | MEDLINE | ID: mdl-35489438

ABSTRACT

Post-traumatic stress disorder (PTSD), characterized by abnormally persistent and distressing memories, is a chronic debilitating condition in need of new treatment options. Current treatment guidelines recommend psychotherapy as first line management with only two drugs, sertraline and paroxetine, approved by U.S. Food and Drug Administration (FDA) for treatment of PTSD. These drugs have limited efficacy as they only reduce symptoms related to depression and anxiety without producing permanent remission. PTSD remains a significant public health problem with high morbidity and mortality requiring major advances in therapeutics. Early evidence has emerged for the beneficial effects of psychedelics particularly in combination with psychotherapy for management of PTSD, including psilocybin, MDMA, LSD, cannabinoids, ayahuasca and ketamine. MDMA and psilocybin reduce barrier to therapy by increasing trust between therapist and patient, thus allowing for modification of trauma related memories. Furthermore, research into the memory reconsolidation mechanisms has allowed for identification of various pharmacological targets to disrupt abnormally persistent memories. A number of pre-clinical and clinical studies have investigated novel and re-purposed pharmacological agents to disrupt fear memory in PTSD. Novel therapeutic approaches like neuropeptide Y, oxytocin, cannabinoids and neuroactive steroids have also shown potential for PTSD treatment. Here, we focus on the role of fear memory in the pathophysiology of PTSD and propose that many of these new therapeutic strategies produce benefits through the effect on fear memory. Evaluation of recent research findings suggests that while a number of drugs have shown promising results in preclinical studies and pilot clinical trials, the evidence from large scale clinical trials would be needed for these drugs to be incorporated in clinical practice.


Subject(s)
Cannabinoids , N-Methyl-3,4-methylenedioxyamphetamine , Stress Disorders, Post-Traumatic , United States , Humans , Stress Disorders, Post-Traumatic/drug therapy , N-Methyl-3,4-methylenedioxyamphetamine/pharmacology , N-Methyl-3,4-methylenedioxyamphetamine/therapeutic use , Psilocybin/therapeutic use , Fear/physiology , Cannabinoids/therapeutic use
4.
Front Psychiatry ; 11: 49, 2020.
Article in English | MEDLINE | ID: mdl-32153437

ABSTRACT

Smoking is a habit that is hard to break because nicotine is highly addictive and smoking behavior is strongly linked to multiple daily activities and routines. Here, we explored the effect of gender, age, day of the week, and previous smoking on the number of cigarettes smoked on any given day. Data consisted of daily records of the number of cigarettes participants smoked over an average period of 84 days. The sample included smokers (36 men and 26 women), aged between 18 and 26 years, who smoked at least five cigarettes a day and had smoked for at least 2 years. A panel data analysis was performed by way of multilevel pooled time series modeling. Smoking on any given day was a function of the number of cigarettes smoked on the previous day, and 2, 7, 14, 21, 28, 35, 42, 49, and 56 days previously, and the day of the week. Neither gender nor age influenced this pattern, with no multilevel effects being detected, thus the behavior of all participants fitted the same smoking model. These novel findings show empirically that smoking behavior is governed by firmly established temporal dependence patterns and inform temporal parameters for the rational design of smoking cessation programs.

5.
Aging Cell ; 19(1): e13079, 2020 01.
Article in English | MEDLINE | ID: mdl-31736210

ABSTRACT

One of the hallmarks of aging is the progressive accumulation of senescent cells in organisms, which has been proposed to be a contributing factor to age-dependent organ dysfunction. We recently reported that Bruton's tyrosine kinase (BTK) is an upstream component of the p53 responses to DNA damage. BTK binds to and phosphorylates p53 and MDM2, which results in increased p53 activity. Consistent with this, blocking BTK impairs p53-induced senescence. This suggests that sustained BTK inhibition could have an effect on organismal aging by reducing the presence of senescent cells in tissues. Here, we show that ibrutinib, a clinically approved covalent inhibitor of BTK, prolonged the maximum lifespan of a Zmpste24-/- progeroid mice, which also showed a reduction in general age-related fitness loss. Importantly, we found that certain brain functions were preserved, as seen by reduced anxiety-like behaviour and better long-term spatial memory. This was concomitant to a decrease in the expression of specific markers of senescence in the brain, which confirms a lower accumulation of senescent cells after BTK inhibition. Our data show that blocking BTK has a modest increase in lifespan in Zmpste24-/- mice and protects them from a decline in brain performance. This suggests that specific inhibitors could be used in humans to treat progeroid syndromes and prevent the age-related degeneration of organs such as the brain.


Subject(s)
Agammaglobulinaemia Tyrosine Kinase/antagonists & inhibitors , Aging/drug effects , Brain/drug effects , Cognitive Dysfunction/drug therapy , Protein Kinase Inhibitors/therapeutic use , Animals , Brain/pathology , Humans , Mice , Protein Kinase Inhibitors/pharmacology
6.
PLoS One ; 14(11): e0225745, 2019.
Article in English | MEDLINE | ID: mdl-31743378

ABSTRACT

[This corrects the article DOI: 10.1371/journal.pone.0209475.].

7.
PLoS One ; 14(1): e0209475, 2019.
Article in English | MEDLINE | ID: mdl-30673704

ABSTRACT

Salivary alpha-amylase (sAA) activity has been widely used in psychological and medical research as a surrogate marker of sympathetic nervous system activation, though its utility remains controversial. The aim of this work was to compare alternative intensive longitudinal models of sAA data: (a) a traditional model, where sAA is a function of hour (hr) and hr squared (sAAj,t = f(hr, hr2), and (b) an autoregressive model, where values of sAA are a function of previous values (sAAj,t = f(sAA j,t-1, sAA j,t-2, …, sAA j,t-p). Nineteen normal subjects (9 males and 10 females) participated in the experiments and measurements were performed every hr between 9:00 and 21:00 hr. Thus, a total of 13 measurements were obtained per participant. The Napierian logarithm of the enzymatic activity of sAA was analysed. Data showed that a second-order autoregressive (AR(2)) model was more parsimonious and fitted better than the traditional multilevel quadratic model. Therefore, sAA follows a process whereby, to forecast its value at any given time, sAA values one and two hr prior to that time (sAA j,t = f(SAAj,t-1, SAAj,t-2) are most predictive, thus indicating that sAA has its own inertia, with a "memory" of the two previous hr. These novel findings highlight the relevance of intensive longitudinal models in physiological data analysis and have considerable implications for physiological and biobehavioural research involving sAA measurements and other stress-related biomarkers.


Subject(s)
Circadian Rhythm/physiology , Models, Biological , Salivary alpha-Amylases/metabolism , Adult , Biomarkers/metabolism , Female , Humans , Longitudinal Studies , Male , Regression Analysis , Stress, Physiological/physiology , Sympathetic Nervous System/physiology , Young Adult
8.
Expert Opin Ther Targets ; 22(6): 513-526, 2018 06.
Article in English | MEDLINE | ID: mdl-29798691

ABSTRACT

INTRODUCTION: The trace amines, endogenous amines closely related to the biogenic amine neurotransmitters, have been known to exert physiological and neurological effects for decades. The recent identification of a trace amine-sensitive G protein-coupled receptor, trace amine-associated receptor 1 (TAAR1), and subsequent development of TAAR1-selective small-molecule ligands, has renewed research into the therapeutic possibilities of trace amine signaling. Areas covered: Recent efforts in elucidating the neuropharmacology of TAAR1, particularly in neuropsychiatric and neurodegenerative disease, addiction, and regulation of arousal state, will be discussed. Focused application of TAAR1 mutants, synthetic TAAR1 ligands, and endogenous biomolecules such as 3-iodothyronamine (T1AM) has yielded a basic functional portrait for TAAR1, despite a complex biochemistry and pharmacology. The close functional relationship between TAAR1 and dopaminergic signaling is likely to underlie many of its CNS effects. However, TAAR1's influences on serotonin and glutamate neurotransmission will also be highlighted. Expert opinion: TAAR1 holds great promise as a therapeutic target for mental illness, addiction, and sleep disorders. A combination of preclinical and translationally driven studies has solidified TAAR1 as a key node in the regulation of dopaminergic signaling. Continued focus on the mechanisms underlying TAAR1's regulation of serotonin and glutamate signaling, as well as dopamine, will yield further disease-relevant insights.


Subject(s)
Mental Disorders/drug therapy , Molecular Targeted Therapy , Receptors, G-Protein-Coupled/metabolism , Animals , Dopamine/metabolism , Drug Design , Humans , Ligands , Mental Disorders/physiopathology , Signal Transduction , Sleep Wake Disorders/drug therapy , Sleep Wake Disorders/physiopathology , Substance-Related Disorders/drug therapy , Substance-Related Disorders/physiopathology , Thyronines/metabolism
9.
Neurosci Lett ; 671: 88-92, 2018 04 03.
Article in English | MEDLINE | ID: mdl-29452175

ABSTRACT

There are currently no approved medications to effectively counteract the effects of methamphetamine (METH), reduce its abuse and prolong abstinence from it. Data accumulated in recent years have shown that a range of N-substituted benztropine (BZT) analogues possesses psychopharmacological features consistent with those of a potential replacement or "substitute" treatment for stimulant addiction. On the other hand, the evidence that antidepressant therapy may effectively prevent relapse to stimulant seeking is controversial. Here, we compared in rats the ability of the BZT analogue and high affinity dopamine (DA) reuptake inhibitor, JHW-007, and the antidepressant, trazodone, administered during extinction sessions after chronic METH self-administration, to alter METH-primed reinstatement of drug seeking. The data showed that trazodone produced paradoxical effects on lever pressing during extinction of METH self-administration, decreasing active, but increasing inactive, lever pressing. JHW-007 did not have any observable effects on extinction training. Importantly, JHW-007 significantly attenuated METH-primed reinstatement, whereas trazodone enhanced it. These findings lend support to the candidacy of selective DA uptake blockers, such as JHW-007, as potential treatments for METH addiction, but not to the use of antidepressant medication as a single therapeutic approach for relapse prevention.


Subject(s)
Benztropine/analogs & derivatives , Central Nervous System Stimulants/administration & dosage , Conditioning, Operant/drug effects , Dopamine Uptake Inhibitors/pharmacology , Drug-Seeking Behavior/drug effects , Extinction, Psychological/drug effects , Methamphetamine/administration & dosage , Animals , Autoantigens , Behavior, Addictive , Benztropine/pharmacology , Male , Rats , Rats, Long-Evans , Selective Serotonin Reuptake Inhibitors/pharmacology , Trazodone/pharmacology
10.
Front Hum Neurosci ; 12: 538, 2018.
Article in English | MEDLINE | ID: mdl-30687051

ABSTRACT

Attention bias modification (ABM) can decrease the selective visual attention paid to alcohol-related cues but has not been found to reliably reduce alcohol craving. Here, a cognitive intervention to decrease craving by increasing sense of control (Shamloo and Cox, 2014) was used as a complement. We investigated the effects of two such interventions administered singly or in combination. Participants were 41 binge drinkers (BDs) and 10 non-binge drinkers (NBDs). BDs received either ABM, sense of control training, both interventions, or no intervention, and were compared with NBDs who received no intervention. Groups were assessed on alcohol attention bias change including both reaction times and cue-elicited ERPs (visual dot-probe task), alcohol craving change, and alcohol consumption. BDs exhibited higher attention bias scores than NBDs. ABM had no effect on BDs' behavioral or electrophysiological markers of attention bias. Sense of control training did not increase personal sense of control but protected against decreased task accuracy and against increased craving. BDs receiving the combined intervention consumed less alcohol in a bogus taste test than participants receiving no intervention. Taken together, the results suggest that ABM procedure may reduce alcohol consumption if combined with sense of control training.

11.
Sci Rep ; 7(1): 13901, 2017 10 24.
Article in English | MEDLINE | ID: mdl-29066851

ABSTRACT

Recent evidence suggests that the trace amine-associated receptor 1 (TAAR1) plays a pivotal role in the regulation of dopamine (DA) transmission and cocaine's actions. However, the underlying mechanisms through which TAAR1 activation mediates these effects have not yet been elucidated. Here, we used fast-scan cyclic voltammetry to measure DA dynamics and explore such mechanisms. We show, first, that the full TAAR1 agonist, RO5256390, dose-dependently blocked cocaine-induced inhibition of DA clearance in slices of the nucleus accumbens. Second, subthreshold inhibition of PKA or PKC phosphorylation did not prevent TAAR1 suppression of cocaine effects whereas subeffective doses of the DA D2 receptor antagonist, L-741,626, rescued cocaine's ability to produce changes in DA uptake in the presence of full TAAR1 activation, thus indicating that TAAR1 modulation of cocaine effects requires simultaneous DA D2 receptor activation. Predictably, inhibition of glycogen synthase kinase-3 (GSK-3), which results from activation of D2/TAAR1 heterodimers, fully reproduced the inhibitory effects of TAAR1 activation on cocaine-induced changes in DA transmission. Collectively, the present observations reveal that the ability of TAAR1 to regulate cocaine effects is linked to cooperative interactions with D2 autoreceptors and associated downstream molecular targets converging on GSK-3 and suggest a new mechanism to disrupt cocaine neurochemical actions.


Subject(s)
Cocaine/pharmacology , Receptors, Dopamine D2/metabolism , Receptors, G-Protein-Coupled/metabolism , Animals , Cyclic AMP-Dependent Protein Kinases/antagonists & inhibitors , Cyclic AMP-Dependent Protein Kinases/metabolism , Dopamine/metabolism , Dopamine D2 Receptor Antagonists/pharmacology , Enzyme Activation/drug effects , Glycogen Synthase Kinase 3/antagonists & inhibitors , Male , Neurochemistry , Protein Binding , Protein Kinase Inhibitors/pharmacology , Rats , Synaptic Transmission/drug effects
12.
Pharmacol Biochem Behav ; 157: 41-46, 2017 06.
Article in English | MEDLINE | ID: mdl-28455125

ABSTRACT

Addiction is characterised by cycles of compulsive drug taking, periods of abstinence and episodes of relapse. The extinction/reinstatement paradigm has been extensively used in rodents to model human relapse and explore underlying mechanisms and therapeutics. However, relapse to drug seeking behaviour has not been previously demonstrated in invertebrates. Here, we used a cocaine conditioned place preference (CPP) paradigm in the flatworm, planarian, followed by extinction and reinstatement of drug seeking. Once baseline preference was established for one of two distinctly textured environments (i.e. compartments with a coarse or smooth surface), planarian received pairings of cocaine (5µM) in the non-preferred, and vehicle in the most preferred, environment, and were tested for conditioning thereafter. Cocaine produced robust CPP, measured as a significant increase in the time spent in the cocaine-paired compartment. Subsequently, planarian underwent extinction training, reverting back to their original preference within three sessions. Brief exposure to cocaine (5µM) or methamphetamine (5µM) reinstated cocaine-seeking behaviour. By contrast, the high affinity dopamine transporter inhibitor, (N-(n-butyl)-3α-[bis (4-fluorophenyl) methoxy]-tropane) (JHW007), which in rodents exhibits a neurochemical and behavioural profile distinct from cocaine, was ineffective. The present findings demonstrate for the first time reinstatement of extinguished cocaine seeking in an invertebrate model and suggest that the long-term adaptations underlying drug conditioning and relapse are highly conserved through evolution.


Subject(s)
Cocaine/pharmacology , Conditioning, Operant/drug effects , Drug-Seeking Behavior/drug effects , Extinction, Psychological/drug effects , Locomotion/drug effects , Reinforcement, Psychology , Animals , Behavior, Addictive/psychology , Conditioning, Operant/physiology , Drug-Seeking Behavior/physiology , Extinction, Psychological/physiology , Invertebrates , Locomotion/physiology , Planarians , Recurrence
13.
Addict Biol ; 22(5): 1246-1256, 2017 Sep.
Article in English | MEDLINE | ID: mdl-27193165

ABSTRACT

Recent evidence suggests that the trace amine-associated receptor 1 (TAAR1) plays a pivotal role in the regulation of dopamine (DA) transmission and psychostimulant action. Several selective TAAR1 agonists have previously shown efficacy in models of cocaine addiction. However, the effects of TAAR1 activation on methamphetamine (METH)-induced behaviours are less well understood, as indeed are the underlying neurochemical mechanisms mediating potential interactions between TAAR1 and METH. Here, in a progressive ratio schedule of reinforcement the partial TAAR1 agonist, RO5263397, reduced the break-point for METH self-administration, while significantly increasing responding maintained by food reward. Following self-administration and extinction training, RO5263397 completely blocked METH-primed reinstatement of METH seeking. Moreover, when used as a substitute, unlike a low dose of METH, which sustained vigorous responding when substituting for the training dose of METH, RO5263397 was not self-administered at any dose, thus exhibiting no apparent abuse liability. Fast-scan cyclic voltammetry experiments showed that RO5263397 prevented METH-induced DA overflow in slices of the nucleus accumbens, while having no effect on DA transmission in its own right. Collectively, the present observations demonstrate that partial TAAR1 activation decreases the motivation to self-administer METH, blocks METH-primed reinstatement of METH seeking and prevents METH-induced DA elevations in the nucleus accumbens, and strongly support the candidacy of TAAR1-based medications as potential substitute treatment in METH addiction.


Subject(s)
Behavior, Animal/drug effects , Central Nervous System Stimulants/administration & dosage , Drug-Seeking Behavior/drug effects , Methamphetamine/administration & dosage , Oxazoles/pharmacology , Receptors, G-Protein-Coupled/agonists , Self Administration , Amphetamine-Related Disorders , Animals , Dopamine/metabolism , Male , Nucleus Accumbens/drug effects , Nucleus Accumbens/metabolism , Rats , Rats, Long-Evans , Reinforcement, Psychology , Reward
14.
Article in English | MEDLINE | ID: mdl-27865801

ABSTRACT

A high proportion of young methamphetamine (MA) users simultaneously consume alcohol. However, the potential neurological and behavioural alterations induced by such a drug combination have not been systematically examined. We studied in adolescent rats the long-term effects of alcohol, MA, and alcohol and MA combined on anxiety-like behaviour, memory, and neurogenesis in the adult hippocampus. Rats received saline, ethanol (ETOH, 1.5g/kg), MA (MA, 2mg/kg), or ethanol and MA combined (ETHOH-MA, 1.5g/kg ethanol plus 2mg/kg MA) via oral gavage, once daily for 5 consecutive days. Open field (OF), elevated plus maze (EPM) and radial arm maze (RAM) tests were conducted following a 15-day withdrawal period. The results showed alterations in exploratory behaviour in the OF in the MA and ETOH-MA groups, and anxiety-like effects in the EPM in all three drug treatment groups. All three drug groups exhibited reference memory deficits in the RAM, but only the combination treatment group displayed alterations in working memory. Both MA and ETOH-MA treatments increased the length of doublecortin (DCX)-void gaps in the dentate gyrus but only ETOH-MA treatment increased the number of such gaps. An increased number and length of DCX-void gaps correlated with decreased exploratory activity in the OF, and impaired working memory in the RAM was associated with an augmented number of gaps. These findings suggest that alterations in adult hippocampal neurogenesis are linked to the persistent cognitive and behavioural deficits produced by alcohol and MA exposure.


Subject(s)
Anxiety/chemically induced , Central Nervous System Depressants/toxicity , Central Nervous System Stimulants/toxicity , Cognition Disorders/chemically induced , Ethanol/toxicity , Methamphetamine/toxicity , Neurogenesis/drug effects , Animals , Disease Models, Animal , Doublecortin Domain Proteins , Doublecortin Protein , Drug Combinations , Exploratory Behavior/drug effects , Hippocampus/cytology , Hippocampus/drug effects , Immobility Response, Tonic/drug effects , Male , Maze Learning/drug effects , Microtubule-Associated Proteins/metabolism , Neuropeptides/metabolism , Rats , Rats, Long-Evans , Statistics, Nonparametric , Time
15.
Front Neurosci ; 10: 148, 2016.
Article in English | MEDLINE | ID: mdl-27092049

ABSTRACT

Biogenic amines are a collection of endogenous molecules that play pivotal roles as neurotransmitters and hormones. In addition to the "classical" biogenic amines resulting from decarboxylation of aromatic acids, including dopamine (DA), norepinephrine, epinephrine, serotonin (5-HT), and histamine, other biogenic amines, present at much lower concentrations in the central nervous system (CNS), and hence referred to as "trace" amines (TAs), are now recognized to play significant neurophysiological and behavioral functions. At the turn of the century, the discovery of the trace amine-associated receptor 1 (TAAR1), a phylogenetically conserved G protein-coupled receptor that is responsive to both TAs, such as ß-phenylethylamine, octopamine, and tyramine, and structurally-related amphetamines, unveiled mechanisms of action for TAs other than interference with aminergic pathways, laying the foundations for deciphering the functional significance of TAs and its mammalian CNS receptor, TAAR1. Although, its molecular interactions and downstream targets have not been fully elucidated, TAAR1 activation triggers accumulation of intracellular cAMP, modulates PKA and PKC signaling and interferes with the ß-arrestin2-dependent pathway via G protein-independent mechanisms. TAAR1 is uniquely positioned to exert direct control over DA and 5-HT neuronal firing and release, which has profound implications for understanding the pathophysiology of, and therefore designing more efficacious therapeutic interventions for, a range of neuropsychiatric disorders that involve aminergic dysregulation, including Parkinson's disease, schizophrenia, mood disorders, and addiction. Indeed, the recent development of novel pharmacological tools targeting TAAR1 has uncovered the remarkable potential of TAAR1-based medications as new generation pharmacotherapies in neuropsychiatry. This review summarizes recent developments in the study of TAs and TAAR1, their intricate neurochemistry and pharmacology, and their relevance for neurodegenerative and neuropsychiatric disease.

16.
Front Psychiatry ; 7: 211, 2016.
Article in English | MEDLINE | ID: mdl-28149282

ABSTRACT

Alleviating the personal and social burden associated with substance use disorders requires the implementation of a comprehensive strategy, including outreach, education, community interventions, psychiatric treatment, and access to needle exchange programs (NEP), where peer support may be available. Given that substantial research underscores the potential benefits of peer support in psychiatric interventions, we aimed to conduct a national survey to examine key domains of mental health status in people who inject drugs (PWID) in New Zealand. PWID were recruited from 24 pharmacies and 16 dedicated peer-based needle exchanges (PBNEs) across the country. We focused on two mental health outcomes: (1) affective dysregulation, across the three emotional domains of the Depression Anxiety Stress Scale, due to its role in the maintenance of continued drug use, and (2) positive cognition and effective health- and drug-related information exchange with the provider, using the Satisfaction with Life Scale and an ad hoc questionnaire, respectively, in view of their association with improved mental health outcomes. We hypothesized that access to peer support would be associated with mental health benefits for PWIDs. Remarkably, the results of a multistep regression analysis revealed that irrespective of sex, age, ethnicity, main drug used, length of drug use, and frequency of visits to the NEP, the exclusive or preferential use of PBNEs predicted significantly lower depression and anxiety scores, greater satisfaction with life, and increased health-related information exchange with the service provider. These findings demonstrate for the first time an association between access to peer support at PBNEs and positive indices of mental health, lending strong support to the effective integration of such peer-delivered NEP services into the network of mental health services for PWID worldwide.

17.
Article in English | MEDLINE | ID: mdl-26048337

ABSTRACT

The newly discovered trace amine-associated receptor 1 (TAAR1) has emerged as a promising target for medication development in stimulant addiction due to its ability to regulate dopamine (DA) function and modulate stimulants' effects. Recent findings indicate that TAAR1 activation blocks some of the abuse-related physiological and behavioral effects of cocaine. However, findings from existing self-administration studies are inconclusive due to the very limited range of cocaine unit doses tested. Here, in order to shed light on the influence of TAAR1 on cocaine's reward and reinforcement, we studied the effects of partial and full activation of TAAR1on (1) the dose-response curve for cocaine self-administration and (2) cocaine-induced changes in intracranial self-stimulation (ICSS). In the first experiment, we examined the effects of the selective full and partial TAAR1 agonists, RO5256390 and RO5203648, on self-administration of five unit-injection doses of cocaine (0.03, 0.1, 0.2, 0.45, and 1mg/kg/infusion). Both agonists induced dose-dependent downward shifts in the cocaine dose-response curve, indicating that both partial and full TAAR1 activation decrease cocaine, reinforcing efficacy. In the second experiment, RO5256390 and the partial agonist, RO5263397, dose-dependently prevented cocaine-induced lowering of ICSS thresholds. Taken together, these data demonstrated that TAAR1 stimulation effectively suppresses the rewarding and reinforcing effects of cocaine in self-administration and ICSS models, supporting the candidacy of TAAR1 as a drug discovery target for cocaine addiction.


Subject(s)
Brain/drug effects , Cocaine/pharmacology , Dopamine Uptake Inhibitors/pharmacology , Receptors, G-Protein-Coupled/metabolism , Reward , Analysis of Variance , Animals , Conditioning, Operant/drug effects , Dose-Response Relationship, Drug , Male , Oxazoles/administration & dosage , Rats , Rats, Long-Evans , Receptors, G-Protein-Coupled/agonists , Self Administration , Ventral Tegmental Area/drug effects , Ventral Tegmental Area/metabolism
18.
Front Neurosci ; 9: 39, 2015.
Article in English | MEDLINE | ID: mdl-25762894

ABSTRACT

The newly discovered trace amine-associated receptor 1 (TAAR1) has the ability to regulate both dopamine function and psychostimulant action. Here, we tested in rats the ability of RO5203648, a selective TAAR1 partial agonist, to modulate the physiological and behavioral effects of methamphetamine (METH). In experiment 1, RO5203468 dose- and time-dependently altered METH-induced locomotor activity, manifested as an early attenuation followed by a late potentiation of METH's stimulating effects. In experiment 2, rats received a 14-day treatment regimen during which RO5203648 was co-administered with METH. RO5203648 dose-dependently attenuated METH-stimulated hyperactivity, with the effects becoming more apparent as the treatments progressed. After chronic exposure and 3-day withdrawal, rats were tested for locomotor sensitization. RO5203648 administration during the sensitizing phase prevented the development of METH sensitization. However, RO5203648, at the high dose, cross-sensitized with METH. In experiment 3, RO5203648 dose-dependently blocked METH self-administration without affecting operant responding maintained by sucrose, and exhibited lack of reinforcing efficacy when tested as a METH's substitute. Neurochemical data showed that RO5203648 did not affect METH-mediated DA efflux and uptake inhibition in striatal synaptosomes. In vivo, however, RO5203648 was able to transiently inhibit METH-induced accumulation of extracellular DA levels in the nucleus accumbens. Taken together, these data highlight the significant potential of TAAR1 to modulate METH's neurochemical and behavioral effects.

19.
Neuropsychopharmacology ; 39(10): 2299-308, 2014 Sep.
Article in English | MEDLINE | ID: mdl-24722355

ABSTRACT

The trace amine-associated receptor 1 (TAAR1) has emerged as a promising target for medication development in addiction because of its ability to regulate dopamine (DA) transmission. We tested in rats the efficacy of RO5203648 and RO5256390, partial and full TAAR1 agonists, respectively, in models of cocaine relapse. Using a model of context-induced relapse, both RO5203648 and RO5256390 dose-dependently suppressed cocaine seeking after a 2-week period of withdrawal from chronic cocaine self-administration. In a model of extinction-reinstatement, RO5203648 completely inhibited cocaine-primed reinstatement of cocaine seeking. At doses that effectively suppressed cocaine seeking neither RO5203648 nor RO5256390 altered responding maintained by a natural reward. Moreover, fast scan cyclic voltammetry data showed that RO5203648 prevented cocaine-induced DA overflow in the nucleus accumbens without altering DA half-life, suggesting that the partial TAAR1 agonist attenuated cocaine-stimulated DA overflow by mechanisms other than direct interference with DA uptake. Collectively, these data provide strong evidence in support of TAAR1 as a neuropharmacological target for the treatment of cocaine addiction.


Subject(s)
Central Nervous System Agents/pharmacology , Cocaine-Related Disorders/drug therapy , Drug-Seeking Behavior/drug effects , Oxazoles/pharmacology , Receptors, G-Protein-Coupled/agonists , Animals , Chronic Disease , Cocaine/administration & dosage , Cocaine-Related Disorders/physiopathology , Dopamine/metabolism , Dopamine Uptake Inhibitors/administration & dosage , Dose-Response Relationship, Drug , Drug-Seeking Behavior/physiology , Extinction, Psychological/drug effects , Extinction, Psychological/physiology , Male , Nucleus Accumbens/drug effects , Nucleus Accumbens/physiopathology , Random Allocation , Rats , Rats, Long-Evans , Receptors, G-Protein-Coupled/metabolism , Self Administration
20.
Dev Neurosci ; 36(1): 10-7, 2014.
Article in English | MEDLINE | ID: mdl-24457993

ABSTRACT

Recreational drug use among pregnant women is a source of concern due to potential harmful effects of drug exposure on prenatal and infant development. The simultaneous abuse of ecstasy [3,4-methylenedioxymethamphetamine (MDMA)] and alcohol is prevalent among young adults, including young expectant mothers. Here, we used a rat model to study the potential risks associated with exposure to alcohol and MDMA during pregnancy. Pregnant rats received alcohol, MDMA, or both alcohol and MDMA by gavage at E13 through E15 twice daily. Female offspring treated prenatally with the combination of alcohol and MDMA, but not those exposed to either drug separately, showed at 3 months of age decreased exploratory activity and impaired working memory function. Prenatal treatment with the combination of alcohol and MDMA decreased proliferation of neuronal precursors in the adult dentate gyrus of the hippocampus, as measured by 5-bromo-2-deoxyuridine labelling, and adult neurogenesis, assessed by quantifying doublecortin expression. These results provide the first evidence that the simultaneous abuse of alcohol and ecstasy during pregnancy, even for short periods of time, may cause significant abnormalities in neurocognitive development.


Subject(s)
Ethanol/pharmacology , Hippocampus/drug effects , Memory Disorders/chemically induced , N-Methyl-3,4-methylenedioxyamphetamine/pharmacology , Neurogenesis/drug effects , Prenatal Exposure Delayed Effects/physiopathology , Animals , Behavior, Animal/drug effects , Behavior, Animal/physiology , Doublecortin Protein , Ethanol/toxicity , Exploratory Behavior/drug effects , Exploratory Behavior/physiology , Female , Hippocampus/physiopathology , Memory Disorders/physiopathology , Memory Disorders/psychology , Memory, Short-Term/drug effects , Memory, Short-Term/physiology , Motor Activity/drug effects , Motor Activity/physiology , N-Methyl-3,4-methylenedioxyamphetamine/toxicity , Neurogenesis/physiology , Pregnancy , Prenatal Exposure Delayed Effects/psychology , Rats , Rats, Long-Evans
SELECTION OF CITATIONS
SEARCH DETAIL
...