Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 378
Filter
Add more filters

Publication year range
1.
J Pharmacol Exp Ther ; 377(2): 232-241, 2021 05.
Article in English | MEDLINE | ID: mdl-33622770

ABSTRACT

This study evaluated a battery of pain-stimulated, pain-depressed, and pain-independent behaviors for preclinical pharmacological assessment of candidate analgesics in mice. Intraperitoneal injection of dilute lactic acid (IP acid) served as an acute visceral noxious stimulus to produce four pain-related behaviors in male and female ICR mice: stimulation of 1) stretching, 2) facial grimace, 3) depression of rearing, and 4) depression of nesting. Additionally, nesting and locomotion in the absence of the noxious stimulus were used to assess pain-independent drug effects. These six behaviors were used to compare effects of two mechanistically distinct but clinically effective positive controls (ketoprofen and oxycodone) and two negative controls that are not clinically approved as analgesics but produce either general motor depression (diazepam) or motor stimulation (amphetamine). We predicted that analgesics would alleviate all IP acid effects at doses that did not alter pain-independent behaviors, whereas negative controls would not. Consistent with this prediction, ketoprofen (0.1-32 mg/kg) produced the expected analgesic profile, whereas oxycodone (0.32-3.2 mg/kg) alleviated all IP acid effects except depression of rearing at doses lower than those that altered pain-independent behaviors. For the negative controls, diazepam (1-10 mg/kg) failed to block IP acid-induced depression of either rearing or nesting and only decreased IP acid-stimulated behaviors at doses that also decreased pain-independent behaviors. Amphetamine (0.32-3.2 mg/kg) alleviated all IP acid effects but only at doses that also stimulated locomotion. These results support utility of this model as a framework to evaluate candidate-analgesic effects in a battery of complementary pain-stimulated, pain-depressed, and pain-independent behavioral endpoints. SIGNIFICANCE STATEMENT: Preclinical assays of pain and analgesia often yield false-positive effects with candidate analgesics. This study used two positive-control analgesics (ketoprofen, oxycodone) and two active negative controls (diazepam, amphetamine) to validate a strategy for distinguishing analgesics from nonanalgesics by profiling drug effects in a battery of complementary pain-stimulated, pain-depressed, and pain-independent behaviors in male and female mice.


Subject(s)
Analgesics/toxicity , Behavior, Animal , Movement , Pain/drug therapy , Amphetamine/administration & dosage , Amphetamine/therapeutic use , Amphetamine/toxicity , Analgesics/administration & dosage , Analgesics/therapeutic use , Animals , Diazepam/administration & dosage , Diazepam/therapeutic use , Diazepam/toxicity , Drug Evaluation, Preclinical/methods , Drug Evaluation, Preclinical/standards , False Negative Reactions , Female , Ketoprofen/administration & dosage , Ketoprofen/therapeutic use , Ketoprofen/toxicity , Male , Mice , Mice, Inbred ICR , No-Observed-Adverse-Effect Level , Oxycodone/administration & dosage , Oxycodone/therapeutic use , Oxycodone/toxicity
2.
Arch Toxicol ; 95(10): 3223-3234, 2021 10.
Article in English | MEDLINE | ID: mdl-34414480

ABSTRACT

Amphetamine is widely consumed as drug of abuse due to its stimulating and cognitive enhancing effects. Since amphetamine has been on the market for quite a long time and it is one of the most commonly used stimulants worldwide, to date there is still limited information on its effects on the metabolome. In recent years, untargeted toxicometabolomics have been increasingly used to study toxicity-related pathways of such drugs of abuse to find and identify important endogenous and exogenous biomarkers. In this study, the acute effects of amphetamine intake on plasma and urinary metabolome in rats were investigated. For this purpose, samples of male Wistar rats after a single dose of amphetamine (5 mg/kg) were compared to a control group using an untargeted metabolomics approach. Analysis was performed using normal and reversed phase liquid chromatography coupled to high-resolution mass spectrometry using positive and negative ionization mode. Statistical evaluation was performed using Welch's two-sample t test, hierarchical clustering, as well as principal component analysis. The results of this study demonstrate a downregulation of amino acids in plasma samples after amphetamine exposure. Furthermore, four new potential biomarkers N-acetylamphetamine, N-acetyl-4-hydroxyamphetamine, N-acetyl-4-hydroxyamphetamine glucuronide, and amphetamine succinate were identified in urine. The present study complements previous data and shows that several studies are necessary to elucidate altered metabolic pathways associated with acute amphetamine exposure.


Subject(s)
Amphetamine/toxicity , Central Nervous System Stimulants/toxicity , Metabolome/drug effects , Metabolomics , Amino Acids/blood , Amphetamine/metabolism , Animals , Biomarkers/metabolism , Central Nervous System Stimulants/metabolism , Chromatography, Liquid , Down-Regulation/drug effects , Male , Mass Spectrometry , Principal Component Analysis , Rats , Rats, Wistar
3.
Int J Toxicol ; 40(6): 530-541, 2021 12.
Article in English | MEDLINE | ID: mdl-34610777

ABSTRACT

Respiratory monitoring, using impedance with implanted telemetry in socially housed animals, was not possible until the recent development of digital signal transmission. The objective of this study was to evaluate digital telemetry monitoring of cardiopulmonary parameters (respiratory rate, tidal volume, minute volume, electrocardiography (DII), systemic arterial blood pressure, physical activity, and body temperature) in conscious, single-housed, non-rodent species commonly used in toxicology studies following administration of positive/negative controls (saline, dexmedetomidine, morphine, amphetamine, and doxapram), and also, the effects of various social housing arrangements in untreated female and/or male cynomolgus monkeys, Beagle dogs, and Göttingen minipigs (n = 4 per species). Aggressions were observed in socially housed male minipigs, however, which prevented pair-housed assessments in this species. All tested pharmacological agents significantly altered more than one organ system, highlighting important inter-organ dependencies when analyzing functional endpoints. Stress-related physiological changes were observed with single-housing or pair-housing with a new cage mate in cynomolgus monkeys and Beagle dogs, suggesting that stable social structures are preferable to limit variability, especially around dosing. Concomitant monitoring of cardiovascular and respiratory parameters from the same animals may help reduce the number of animals (3 Rs) needed to fulfill the S7A guidelines and allows for identification of organ system functional correlations. Globally, the data support the use of social housing in non-rodents for safety pharmacology multi-organ system (heart and lungs) monitoring investigations.


Subject(s)
Amphetamine/toxicity , Analgesics, Opioid/toxicity , Cardiovascular System/drug effects , Dexmedetomidine/toxicity , Doxapram/toxicity , Electrocardiography/drug effects , Morphine/toxicity , Animals , Central Nervous System Stimulants/toxicity , Dogs , Electric Impedance , Macaca fascicularis , Swine , Swine, Miniature
4.
Eur J Neurosci ; 51(4): 1026-1041, 2020 02.
Article in English | MEDLINE | ID: mdl-31646669

ABSTRACT

Amphetamine-induced neuroadaptations involve vascular damage, neuroinflammation, a hypo-functioning prefrontal cortex (PFC), and cognitive alterations. Brain angiotensin II, through angiotensin type 1 receptor (AT1 -R), mediates oxidative/inflammatory responses, promoting endothelial dysfunction, neuronal oxidative damage and glial reactivity. The present work aims to unmask the role of AT1 -R in the development of amphetamine-induced changes over glial and vascular components within PFC and hippocampus. Attention deficit was evaluated as a behavioral neuroadaptation induced by amphetamine. Brain microvessels were isolated to further evaluate vascular alterations after amphetamine exposure. Male Wistar rats were administered with AT1 -R antagonist, candesartan, followed by repeated amphetamine. After one week drug-off period, animals received a saline or amphetamine challenge and were evaluated in behavioral tests. Afterward, their brains were processed for cresyl violet staining, CD11b (microglia marker), GFAP (astrocyte marker) or von Willebrand factor (vascular marker) immunohistochemistry, and oxidative/cellular stress determinations in brain microvessels. Statistical analysis was performed by using factorial ANOVA followed by Bonferroni or Tukey tests. Repeated amphetamine administration increased astroglial and microglial markers immunoreactivity, increased apoptotic cells, and promoted vascular network rearrangement at the PFC concomitantly with an attention deficit. Although the amphetamine challenge improved the attentional performance, it triggers detrimental effects probably because of the exacerbated malondialdehyde levels and increased heat shock protein 70 expression in microvessels. All observed amphetamine-induced alterations were prevented by the AT1 -R blockade. Our results support the AT1 -R involvement in the development of oxidative/inflammatory conditions triggered by amphetamine exposure, affecting cortical areas and increasing vascular susceptibility to future challenges.


Subject(s)
Amphetamine , Receptor, Angiotensin, Type 1 , Amphetamine/toxicity , Angiotensin II , Animals , Brain/metabolism , Male , Rats , Rats, Wistar , Receptor, Angiotensin, Type 1/metabolism
5.
Metab Brain Dis ; 35(2): 413-425, 2020 02.
Article in English | MEDLINE | ID: mdl-31840201

ABSTRACT

Evaluate the efficacy of folic acid (FA) as a therapeutic adjunct to lithium (Li) on the manic-like behaviors as well as parameters of oxidative stress and inflammation in an animal model of mania induced by m-amphetamine (m-AMPH). Wistar rats first received m-AMPH or saline (NaCl 0.9%, Sal) for 14 days. Between the 8th and 14th day, rats were treated with water, Li, FA or a combination of thereof drugs (Li + FA). Manic-like behaviors were assessed in the open-field test. Oxidative stress and inflammation parameters were assessed in the frontal cortex, striatum, and hippocampus. Administration of m-AMPH in rats significantly enhanced the exploratory and locomotor behaviors, as well as the risk-taking and stereotypic behaviors. Li + FA reversed these behavioral alterations elicited by m-AMPH. Administration of this psychostimulant also increased oxidative damage to lipids and proteins, whereas Li + FA reversed these oxidative damages. m-AMPH also induced an increase in the glutathione peroxidase (GPx) activity and a decrease in the glutathione reductase (GR) activity. Li + FA reversed the alteration in GR activity, but not in GPx activity. In addition, m-AMPH increased the IL-1ß and TNF-α levels in the rat brain; Li + FA combined therapy reversed the alterations on these inflammatory parameters. FA administration per se reduced the increased TNF-α content induced by m-AMPH. Present study provides evidence that FA is effective as an adjunct to Li standard therapy on manic-like behaviors, oxidative stress and inflammatory parameters in a model of mania induced by m-AMPH.


Subject(s)
Antimanic Agents/administration & dosage , Folic Acid/administration & dosage , Inflammation Mediators/antagonists & inhibitors , Lithium/administration & dosage , Mania/drug therapy , Oxidative Stress/drug effects , Amphetamine/toxicity , Animals , Central Nervous System Stimulants/toxicity , Disease Models, Animal , Drug Therapy, Combination , Inflammation Mediators/metabolism , Male , Mania/chemically induced , Mania/metabolism , Oxidative Stress/physiology , Rats , Rats, Wistar , Treatment Outcome
6.
Int J Mol Sci ; 21(8)2020 Apr 18.
Article in English | MEDLINE | ID: mdl-32325754

ABSTRACT

Halogenation of amphetamines and methcathinones has become a common method to obtain novel psychoactive substances (NPS) also called "legal highs". The para-halogenated derivatives of amphetamine and methcathinone are available over the internet and have entered the illicit drug market but studies on their potential neurotoxic effects are rare. The primary aim of this study was to explore the neurotoxicity of amphetamine, methcathinone and their para-halogenated derivatives 4-fluoroamphetamine (4-FA), 4-chloroamphetamine (PCA), 4-fluoromethcathinone (4-FMC), and 4-chloromethcathinone (4-CMC) in undifferentiated and differentiated SH-SY5Y cells. We found that 4-FA, PCA, and 4-CMC were cytotoxic (decrease in cellular ATP and plasma membrane damage) for both cell types, whereby differentiated cells were less sensitive. IC50 values for cellular ATP depletion were in the range of 1.4 mM for 4-FA, 0.4 mM for PCA and 1.4 mM for 4-CMC. The rank of cytotoxicity observed for the para-substituents was chloride > fluoride > hydrogen for both amphetamines and cathinones. Each of 4-FA, PCA and 4-CMC decreased the mitochondrial membrane potential in both cell types, and PCA and 4-CMC impaired the function of the electron transport chain of mitochondria in SH-SY5Y cells. 4-FA, PCA, and 4-CMC increased the ROS level and PCA and 4-CMC induced apoptosis by the endogenous pathway. In conclusion, para-halogenation of amphetamine and methcathinone increases their neurotoxic properties due to the impairment of mitochondrial function and induction of apoptosis. Although the cytotoxic concentrations were higher than those needed for pharmacological activity, the current findings may be important regarding the uncontrolled recreational use of these compounds.


Subject(s)
Amphetamine/toxicity , Apoptosis/drug effects , Cell Differentiation/drug effects , Membrane Potential, Mitochondrial/drug effects , Mitochondria/drug effects , Neuroblastoma/metabolism , Adenosine Triphosphate/chemistry , Adenosine Triphosphate/metabolism , Amphetamine/chemistry , Amphetamine/metabolism , Amphetamines/metabolism , Amphetamines/toxicity , Cell Line, Tumor , Electron Transport/drug effects , Halogenation , Humans , Inhibitory Concentration 50 , Methylamines/metabolism , Methylamines/toxicity , Mitochondria/metabolism , Oxygen Consumption/drug effects , Propiophenones/metabolism , Propiophenones/toxicity , Reactive Oxygen Species/metabolism , Superoxides/metabolism
7.
Am J Emerg Med ; 37(10): 1880-1890, 2019 10.
Article in English | MEDLINE | ID: mdl-30639129

ABSTRACT

OBJECTIVE: Benzodiazepines are often recommended first-line for management of cocaine and amphetamine toxicity while antipsychotic treatment is discouraged due to the potential for lowering seizure threshold, prolonging the QT interval, and decreasing heat dissipation. We performed a systematic review including animal and human studies to elucidate the efficacy and safety of antipsychotics in managing sympathomimetic toxicity specifically evaluating the effect of treatment on mortality, seizures, hyperthermia, and cardiovascular effects. METHODS: We searched MEDLINE, Embase, BIOSIS Previews, Web of Science, Scopus, CENTRAL and gray literature from inception to 31 May 2017 to answer: Can antipsychotics be used safely and effectively to treat cocaine or amphetamine toxicity? Citations were screened by title and abstract. Additional citations were identified with citation tracking. Data were extracted from full-texts. RESULTS: 6539 citations were identified; 250 full-text articles were assessed. Citation tracking identified 2336 citations; 155 full texts were reviewed. Seventy-three papers were included in this review. In 96 subjects with cocaine toxicity treated with an antipsychotic, there were three deaths, two cardiac arrests, two seizures, and one episode of hyperthermia. In 330 subjects with amphetamine toxicity treated with an antipsychotic, there were two episodes of coma and QT prolongation and one episode of each: hypotension, NMS, cardiac arrest, and death. CONCLUSION: This systematic review represents an exhaustive compilation of the available evidence. There is neither a clear benefit of antipsychotics over benzodiazepines nor a definitive signal of harm noted. We encourage clinicians to adapt treatment based on specific circumstances and characteristics of their individual patients.


Subject(s)
Amphetamine/toxicity , Antipsychotic Agents/therapeutic use , Cocaine/toxicity , Drug Overdose/drug therapy , Illicit Drugs/toxicity , Sympathomimetics/toxicity , Animals , Humans , Treatment Outcome
8.
J Neuroinflammation ; 15(1): 246, 2018 Aug 31.
Article in English | MEDLINE | ID: mdl-30170608

ABSTRACT

BACKGROUND: Chronic persistent airway inflammation has been associated with the comorbidity of asthma and bipolar disorder (BD). However, the direct relevance between airway inflammation and BD-like psychiatric comorbidity is almost unknown. Integrin ß4 (ITGB4) is downregulated on the airway epithelial of asthma patients, which might play a critical role in the parthenogenesis of airway inflammation. So this study aimed to examine the role of ITGB4 deficiency in mediating airway inflammation and further leading to the BD-like behaviors. METHODS: ITGB4-/- mice were generated by mating ITGB4fl/fl mice with CCSP-rtTAtg/-/TetO-Cretg/tg mice. Mania-like behavior tests were performed, including hyperlocomotion, D-amphetamine-induced hyperactivity, open-field test, and elevated plus-maze test. Depressive-like behavior tests were carried out, including sucrose preference, forced swimming, and learned helplessness. Inflammatory cells (Th17, Th1, Th2) in the lung were examined by flow cytometry. Futhermore, inflammatory cytokines (IL-4, IL-13) in bronchoalveolar lavage fluid and sera were detected by ELISA. Protein expression of the IL-4Rα on choroid plexus, microglial marker (IBA1), and synapse-associated proteins (synaptophysin, SYP) in the hippocampus and prefrontal cortex were examined by western blotting. Additionally, proinflammatory cytokines (IL-1ß, IL-6, and TNF-α) in the hippocampus and prefrontal cortex were detected by immunohistochemistry. Inflammatory disorder in the lung, hippocampus, and prefrontal cortex was tested by hematoxylin and eosin (H&E) staining. And cell apoptosis in the hippocampus and prefrontal cortex was measured by TUNEL test. RESULTS: ITGB4-/- mice exhibited mania-like behavior, including hyperlocomotion, D-amphetamine-induced hyperactivity, and reduced anxiety-like behavior. While under stressful conditions, ITGB4-/- mice manifested depressive-like behavior, including anhedonia, behavioral despair, and enhanced learned helplessness. At the same time, ITGB4-/- mice mainly exerted Th2-type inflammation in periphery, like the number and major cytokines IL-4 and IL-13 of Th2-type inflammation. ITGB4-/- mice also showed a significant increase of microglia and pro-inflammatory cytokines such as IL-1ß, IL-6, and TNF-α in the hippocampus and prefrontal cortex. Additionally, neuron damage, increased neuron apoptosis, and the decrease of SYP were found in ITGB4-/- mice. CONCLUSIONS: These findings confirmed that airway inflammatory induced by ITGB4 deficiency is the important incentive for the BD-like behavior during asthma pathogenesis. The ITGB4-deficient mice provide a validated animal model for us to study the possible mechanism of BD-like psychiatric comorbidity of asthma patients.


Subject(s)
Bipolar Disorder/genetics , Bronchitis/genetics , Bronchitis/pathology , Epithelial Cells/pathology , Integrin beta4/metabolism , Amphetamine/toxicity , Animals , Anti-Bacterial Agents/pharmacology , Disease Models, Animal , Doxycycline/pharmacology , Exploratory Behavior/physiology , Gene Expression Regulation/genetics , Hyperkinesis/chemically induced , Hyperkinesis/genetics , Integrin beta4/genetics , Male , Maze Learning/physiology , Mice , Mice, Inbred C57BL , Mice, Transgenic , Microglia/pathology , T-Lymphocyte Subsets/pathology , Uteroglobin/genetics , Uteroglobin/metabolism
9.
J Pineal Res ; 64(3)2018 Apr.
Article in English | MEDLINE | ID: mdl-29149481

ABSTRACT

Chronic amphetamine (AMPH) abuse leads to damage of the hippocampus, the brain area associated with learning and memory process. Previous results have shown that AMPH-induced dopamine neurotransmitter release, reactive oxygen species formation, and degenerative protein aggregation lead to neuronal death. Melatonin, a powerful antioxidant, plays a role as a neuroprotective agent. The objective of this study was to investigate whether the protective effect of melatonin on AMPH-induced hippocampal damage in the postnatal rat acts through the dopaminergic pathway. Four-day-old postnatal rats were subcutaneously injected with 5-10 mg/kg AMPH and pretreated with 10 mg/kg melatonin prior to AMPH exposure for seven days. The results showed that melatonin decreased the AMPH-induced hippocampal neuronal degeneration in the dentate gyrus, CA1, and CA3. Melatonin attenuated the reduction in the expression of hippocampal synaptophysin, PSD-95, α-synuclein, and N-methyl-D-aspartate (NMDA) receptor protein and mRNA caused by AMPH. Melatonin attenuated the AMPH-induced reduction in dopamine transporter (DAT) protein expression in the hippocampus and the reduction in mRNA expression in the ventral tegmental area (VTA). Immunofluorescence demonstrated that melatonin not only prevented the AMPH-induced loss of DAT and NMDA receptor but also prevented AMPH-induced α-synuclein overexpression in the dentate gyrus, CA1, and CA3. Melatonin decreased the AMPH-induced reduction in the protein and mRNA of the NMDA receptor downstream signaling molecule, calcium/calmodulin-dependent protein kinase II (CaMKII), and the melatonin receptors (MT1 and MT2). This study showed that melatonin prevented AMPH-induced toxicity in the hippocampus of postnatal rats possibly via its antioxidative effect and mitochondrial protection.


Subject(s)
Amphetamine/toxicity , Central Nervous System Stimulants/toxicity , Dopaminergic Neurons/drug effects , Melatonin/pharmacology , Neuroprotective Agents/pharmacology , Animals , Dopaminergic Neurons/pathology , Hippocampus/drug effects , Hippocampus/pathology , Nerve Degeneration/chemically induced , Nerve Degeneration/pathology , Rats , Rats, Wistar
10.
Int J Toxicol ; 36(2): 113-123, 2017.
Article in English | MEDLINE | ID: mdl-28196426

ABSTRACT

A functional observational battery (FOB) is recommended as the first-tier neurotoxicity screening in the preclinical safety pharmacology testing guidelines. Minipigs have increasingly been used in regulatory toxicology studies; however, no current FOB protocol is available for neurotoxicity testing in these species. Hence, a minipig FOB instrument was developed. A complete crossover study with Sinclair minipigs was performed to evaluate physiologic, neurologic, and behavioral effects of amphetamine, ketamine, and diazepam. The treated minipigs were first observed in their home cage, were video-recorded for 10 minutes in an open field, and then went through a complete neurologic examination. Both ketamine and diazepam were shown to reduce the freezing and behavior shifts of treated minipigs, while increasing their exploratory behaviors. Both drugs also caused muscular and gait impairment. The effects of ketamine and diazepam were consistent with their roles as central nervous system (CNS) suppressants. Unique effects were also observed with ketamine and diazepam treatments, which may reflect their unique mechanisms of action. Consistent with its role as a CNS stimulant, amphetamine caused the treated minipigs to be hyperactive and to display increased freezing and behavior shifts and reduced exploring activities. These effects of amphetamine were opposite to those observed with ketamine and diazepam. Amphetamine also increased locomotion in the treated minipigs. The present effects of amphetamine, ketamine, and diazepam are in agreement with observations by others. In conclusion, the minipig is a suitable species for FOB evaluation of pharmaceuticals in preclinical safety pharmacology testing.


Subject(s)
Drug Evaluation, Preclinical/methods , Neurotoxicity Syndromes/etiology , Swine, Miniature , Amphetamine/toxicity , Animals , Behavior, Animal/drug effects , Central Nervous System Depressants/toxicity , Central Nervous System Stimulants/toxicity , Cross-Over Studies , Diazepam/toxicity , Exploratory Behavior/drug effects , Ketamine/toxicity , Male , Swine
11.
J Biomed Sci ; 23: 21, 2016 Feb 03.
Article in English | MEDLINE | ID: mdl-26841904

ABSTRACT

BACKGROUND: Monoamine oxidase (MAO) enzymes play a critical role in controlling the catabolism of monoamine neurotransmitters and biogenic trace amines and behavior in humans. However, the mechanisms that regulate MAO are unclear. Several transcription factor proteins are proposed to modulate the transcription of MAO gene, but evidence supporting these hypotheses is controversial. We aimed to investigate the mechanism of gene transcription regulator proteins on amphetamine-induced behavior. We applied aptamers containing a DNA binding sequence, as well as a random sequence (without target) to study the modulation of amphetamine-induced MAO levels and hyperactivity in living mice. METHODS: We pretreated in adult male C57black6 mice (Taconic Farm, Germantown, NY) (n ≥ 3 litters at a time), 2 to 3 months of age (23 ± 2 gm body weight) with double-stranded (ds) DNA aptamers with sequence specific to activator protein-1 (5ECdsAP1), nuclear factor-kappa beta (5ECdsNF-kB), special protein-1 (5ECdsSP-1) or cyclicAMP responsive element binding (5ECdsCreB) protein binding regions, 5ECdsRan [a random sequence without target], single-stranded AP-1 (5ECssAP-1) (8 nmol DNA per kg) or saline (5 µl, intracerebroventricular [icv] injection) control before amphetamine administration (4 mg/kg, i.p.). We then measured and analyzed locomotor activities and the level of MAO-A and MAO-B activity. RESULTS: In the pathological condition of amphetamine exposure, we showed here that pretreatment with 5ECdsAP1 and 5ECdsNF-kB reversed the decrease of MAO-A activity (p < 0.05, t test), but not activity of the B isomer (MAO-B), in the ventral tegmental area (VTA) and substantia nigra (SN) of C57black6 mice. The change in MAO-A level coincided with a reversed amphetamine-induced restless behavior of mice. Pretreatments with saline, 5ECdsCreB, 5ECdsSP-1, 5ECdsRan or 5ECssAP-1 had no effect. CONCLUSION: Our data lead us to conclude that elevation of AP-1 or NF-kB indirectly decreases MAO-A protein levels which, in turn, diminishes MAO-A ability in the VTA of the mesolimbic dopaminergic pathway that has been implicated in cells under stress especially in the SN and VTA. This study has implications for design for the treatment of drug exposure and perhaps Parkinson's dementia.


Subject(s)
Amphetamine/toxicity , Aptamers, Nucleotide/pharmacology , Behavior, Animal/drug effects , Monoamine Oxidase/biosynthesis , NF-kappa B/metabolism , Transcription Factor AP-1/metabolism , Animals , Male , Mice
12.
J Neurosci ; 34(25): 8499-506, 2014 Jun 18.
Article in English | MEDLINE | ID: mdl-24948805

ABSTRACT

Drug addiction has devastating consequences on social behaviors and can lead to the impairment of social bonding. Accumulating evidence indicates that alterations in oxytocin (OT) and dopamine (DA) neurotransmission within brain reward circuitry may be involved. We investigated this possibility, as well as the therapeutic potential of OT for drug-induced social deficits, using the prairie vole (Microtus ochrogaster)-a socially monogamous rodent that forms enduring pair bonds between adult mates. We demonstrate that repeated exposure to the commonly abused psychostimulant amphetamine (AMPH) inhibits the formation of partner preferences (an index of pair bonding) in female prairie voles. AMPH exposure also altered OT and DA neurotransmission in regions that mediate partner preference formation: it decreased OT and DA D2 receptor immunoreactivity in the medial prefrontal cortex (mPFC) and nucleus accumbens (NAcc), respectively, and increased NAcc DA levels. Administration of OT directly into the mPFC of AMPH-exposed voles restored partner preferences, and altered NAcc DA levels, and this effect was dependent on OT receptor activation. Together, these data suggest that repeated AMPH exposure impairs pair bonding through an OT-mediated mechanism, and that OT and DA systems within brain reward circuitry may interact to mediate the complex relationship between drug abuse and social bonding. Further, these results provide empirical support for the idea that the central OT system may represent an important target for the treatment of social deficits in addiction.


Subject(s)
Amphetamine-Related Disorders/metabolism , Amphetamine/toxicity , Dopamine/metabolism , Nucleus Accumbens/metabolism , Oxytocin/physiology , Pair Bond , Social Behavior , Amphetamine/antagonists & inhibitors , Amphetamine/metabolism , Animals , Arvicolinae , Female , Male , Microdialysis/methods , Oxytocin/administration & dosage
13.
J Neurochem ; 135(4): 674-85, 2015 Nov.
Article in English | MEDLINE | ID: mdl-26259827

ABSTRACT

Variants of tryptophan hydroxylase-2 (Tph2), the gene encoding enzyme responsible for the synthesis of brain serotonin (5-HT), have been associated with neuropsychiatric disorders, substance abuse and addiction. This study assessed the effect of Tph2 gene deletion on motor behavior and found that motor activity induced by 2.5 and 5 mg/kg amphetamine was enhanced in Tph2(-/-) mice. Using the in vivo microdialysis technique we found that the ability of amphetamine to stimulate noradrenaline (NA) release in the striatum was reduced by about 50% in Tph2(-/-) mice while the release of dopamine (DA) was not affected. Tph2 deletion did not affect the release of NA and DA in the prefrontal cortex. The role of endogenous 5-HT in enhancing the effect of amphetamine was confirmed showing that treatment with the 5-HT precursor 5-hydroxytryptophan (10 mg/kg) restored tissue and extracellular levels of brain 5-HT and the effects of amphetamine on striatal NA release and motor activity in Tph2(-/-) mice. Treatment with the NA precursor dihydroxyphenylserine (400 mg/kg) was sufficient to restore the effect of amphetamine on striatal NA release and motor activity in Tph2(-/-) mice. These findings indicate that amphetamine-induced hyperactivity is attenuated by endogenous 5-HT through the inhibition of striatal NA release. Tph2(-/-) mice may be a useful preclinical model to assess the role of 5-HT-dependent mechanisms in the action of psychostimulants. Acute sensitivity to the motor effects of amphetamine has been associated to increased risk of psychostimulant abuse. Here, we show that deletion of Tph2, the gene responsible for brain 5-HT synthesis, enhances the motor effect of amphetamine in mice through the inhibition of striatal NA release. This suggests that Tph2(-/-) mice is a useful preclinical model to assess the role of 5-HT-dependent mechanisms in psychostimulants action. Tph2, tryptophan hydroxylase-2.


Subject(s)
Adrenergic Uptake Inhibitors/toxicity , Amphetamine/toxicity , Corpus Striatum/metabolism , Hyperkinesis , Norepinephrine/metabolism , Serotonin/metabolism , Tryptophan Hydroxylase/deficiency , 5-Hydroxytryptophan/pharmacology , Animals , Carbidopa/pharmacology , Corpus Striatum/drug effects , Disease Models, Animal , Dopamine Agents/pharmacology , Female , Gene Expression Regulation/drug effects , Gene Expression Regulation/genetics , Hyperkinesis/chemically induced , Hyperkinesis/genetics , Hyperkinesis/pathology , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Microdialysis , Motor Activity/drug effects , Motor Activity/genetics , Time Factors , Tryptophan Hydroxylase/genetics
14.
Eur J Neurosci ; 41(12): 1553-68, 2015 Jun.
Article in English | MEDLINE | ID: mdl-25828470

ABSTRACT

Although ASIC4 is a member of the acid-sensing ion channel (ASIC) family, we have limited knowledge of its expression and physiological function in vivo. To trace the expression of this ion channel, we generated the ASIC4-knockout/CreERT(2)-knockin (Asic4(Cre) (ERT) (2)) mouse line. After tamoxifen induction in the Asic4(Cre) (ERT)(2)::CAG-STOP(floxed)-Td-tomato double transgenic mice, we mapped the expression of ASIC4 at the cellular level in the central nervous system (CNS). ASIC4 was expressed in many brain regions, including the olfactory bulb, cerebral cortex, striatum, hippocampus, amygdala, thalamus, hypothalamus, brain stem, cerebellum, spinal cord and pituitary gland. Colocalisation studies further revealed that ASIC4 was expressed mainly in three types of cells in the CNS: (i) calretinin (CR)-positive and/or vasoactive intestine peptide (VIP)-positive interneurons; (ii) neural/glial antigen 2 (NG2)-positive glia, also known as oligodendrocyte precursor cells; and (iii) cerebellar granule cells. To probe the possible role of ASIC4, we hypothesised that ASIC4 could modulate the membrane expression of ASIC1a and thus ASIC1a signaling in vivo. We conducted behavioral phenotyping of Asic4(Cre) (ERT)(2) mice by screening many of the known behavioral phenotypes found in Asic1a knockouts and found ASIC4 not involved in shock-evoked fear learning and memory, seizure termination or psychostimulant-induced locomotion/rewarding effects. In contrast, ASIC4 might play an important role in modulating the innate fear response to predator odor and anxious state because ASIC4-mutant mice showed increased freezing response to 2,4,5-trimethylthiazoline and elevated anxiety-like behavior in both the open-field and elevated-plus maze. ASIC4 may modulate fear and anxiety by counteracting ASIC1a activity in the brain.


Subject(s)
Acid Sensing Ion Channels/metabolism , Anxiety/metabolism , Fear/physiology , Acid Sensing Ion Channels/genetics , Amphetamine/toxicity , Animals , Anxiety/genetics , Body Composition/drug effects , Body Composition/genetics , Eating/drug effects , Eating/genetics , Estrogen Antagonists/pharmacology , Excitatory Amino Acid Agonists/toxicity , Fear/drug effects , Humans , Hyperkinesis/chemically induced , Kainic Acid/toxicity , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Nervous System/drug effects , Nervous System/metabolism , Nociception/drug effects , Nociception/physiology , Seizures/chemically induced , Tamoxifen/pharmacology , Time Factors
15.
Am J Addict ; 24(7): 586-9, 2015 Oct.
Article in English | MEDLINE | ID: mdl-26332037

ABSTRACT

BACKGROUND AND OBJECTIVES: We investigated transition from amphetamine-induced psychosis (AIP) to schizophrenia. METHODS: A sample of 28 individuals was identified while hospitalized for AIP. We reviewed their hospital records after six years. RESULTS: During follow-up, seven individuals (25%) died and nine (32%) had moved from the area. Of the remaining 12, four individuals (25%) were diagnosed with schizophrenia. These individuals were, at baseline, characterized by fewer hallucinatory symptoms and more homelessness. CONCLUSION AND SCIENTIFIC SIGNIFICANCE: Hospitalization for AIP was a relatively specific risk factor for schizophrenia and the mortality rate in AIP was high.


Subject(s)
Amphetamine/toxicity , Psychoses, Substance-Induced/epidemiology , Psychoses, Substance-Induced/mortality , Schizophrenia/epidemiology , Schizophrenia/mortality , Disease Progression , Humans , Norway/epidemiology , Prospective Studies , Risk Factors
16.
Chin J Physiol ; 58(4): 219-27, 2015 Aug 31.
Article in English | MEDLINE | ID: mdl-26211645

ABSTRACT

We investigated intermittent hypoxia (IH) on dopamine (DA) release in rat brain treated with or without amphetamine (AMPH). Rats were divided into four groups including normoxia, IH, AMPH, and AMPH + IH treatments. The cerebrospinal fluid (CSF) was collected and the DA levels were detected by high performance liquid chromatography (HPLC). The plasma prolactin (PRL) concentration was measured by radioimmunoassay (RIA). We found that IH reduced basal DA concentration in media prefrontal cortex (mPFC), but increased that in striatum, where DA level was also increased in rats treated with AMPH or AMPH + IH. Angiotensin II (Ang II) increased the DA release in mPFC and striatum and this effect was enhanced in AMPH + IH group. The stimulatory effect of IH on plasma PRL was attenuated in presence of AMPH. Tyrosine hydroxylase (TH) expression was decreased by IH, but increased by AMPH + IH in mPFC. IH or AMPH treatment decreased the expression of vesicular monoamine transporter-2 (VMAT-2) in rat brain. These data suggested that IH altered the DA release and changed the protein expression levels in different parts of rat brain treated with AMPH. IH may play a role in regulating DA metabolism in AMPH addiction.


Subject(s)
Amphetamine/toxicity , Brain/metabolism , Dopamine/metabolism , Hypoxia/metabolism , Angiotensin II/pharmacology , Animals , Male , Prolactin/blood , Rats , Rats, Sprague-Dawley
17.
Neurobiol Dis ; 62: 233-40, 2014 Feb.
Article in English | MEDLINE | ID: mdl-24135006

ABSTRACT

Graft-induced dyskinesia (GID) is a serious complication induced by dopamine (DA) cell transplantation in parkinsonian patients. We have recently shown that DA D2 receptor blockade produces striking blockade of dyskinesia induced by amphetamine in grafted 6-OHDA-lesioned rats, a model of GID. This study was designed to investigate whether blockade of DA D1 receptors could produce similar outcome, and to see whether the effect of these treatments in grafted rats was specific for dyskinesia induced by amphetamine, or could also influence L-DOPA-induced dyskinesia (LID). L-DOPA-primed rats received transplants of fetal DA neurons into the DA-denervated striatum. Beginning at 20weeks after transplantation rats were subjected to pharmacological treatments with either L-DOPA (6mg/kg) or amphetamine (1.5mg/kg) alone, or in combination with the D1 receptor antagonist SCH23390, the D2 receptor antagonist eticlopride, and the 5-HT1A agonist/D2 receptor antagonist buspirone. Grafted rats developed severe GID, while LID was reduced. Both eticlopride and SCH23390 produced near-complete suppression of GID already at very low doses (0.015 and 0.1mg/kg, respectively). Buspirone induced similar suppression at a dose as low as 0.3mg/kg, which is far lower than the dose known to affect LID in non-grafted dyskinetic rats. In agreement with our previous results, the effect of buspirone was independent from 5-HT1A receptor activation, as it was not counteracted by the selective 5-HT1A antagonist WAY100635, but likely due to D2 receptor blockade. Most interestingly, the same doses of eticlopride, SCH23390 and buspirone were found to suppress LID in grafted but not in control dyskinetic rats. Taken together, these data demonstrate that the DA cell grafts strikingly exacerbate the effect of DA D1 and D2 receptor blockade against both GID and LID, and suggest that the anti-GID effect of buspirone seen in patients may also be due to blockade of DA D2 receptors.


Subject(s)
Anti-Dyskinesia Agents/therapeutic use , Dopaminergic Neurons/transplantation , Dyskinesia, Drug-Induced/drug therapy , Parkinsonian Disorders/therapy , Receptors, Dopamine D1/antagonists & inhibitors , Amphetamine/toxicity , Animals , Antiparkinson Agents/toxicity , Benzazepines/therapeutic use , Buspirone/therapeutic use , Disease Models, Animal , Dopamine Agonists/therapeutic use , Dopamine Antagonists/therapeutic use , Dopamine D2 Receptor Antagonists , Female , Indoles/pharmacology , Levodopa/toxicity , Mesencephalon/cytology , Mesencephalon/embryology , Rats , Rats, Sprague-Dawley , Receptors, Dopamine D1/agonists , Receptors, Dopamine D2/agonists , Salicylamides/therapeutic use , Serotonin Receptor Agonists/therapeutic use
18.
Nat Rev Neurosci ; 10(4): 303-12, 2009 Apr.
Article in English | MEDLINE | ID: mdl-19277053

ABSTRACT

The effects of prenatal exposure to drugs on brain development are complex and are modulated by the timing, dose and route of drug exposure. It is difficult to assess these effects in clinical cohorts as these are beset with problems such as multiple exposures and difficulties in documenting use patterns. This can lead to misinterpretation of research findings by the general public, the media and policy makers, who may mistakenly assume that the legal status of a drug correlates with its biological impact on fetal brain development and long-term clinical outcomes. It is important to close the gap between what science tells us about the impact of prenatal drug exposure on the fetus and the mother and what we do programmatically with regard to at-risk populations.


Subject(s)
Brain/drug effects , Drug-Related Side Effects and Adverse Reactions , Pregnancy Complications/chemically induced , Prenatal Exposure Delayed Effects , Alcohols/toxicity , Amphetamine/toxicity , Antidepressive Agents/toxicity , Brain/growth & development , Brain/physiology , Child , Crack Cocaine/toxicity , Female , Humans , Maternal-Fetal Exchange , Methamphetamine/toxicity , Nicotine/toxicity , Pregnancy , Substance-Related Disorders
19.
FASEB J ; 27(2): 712-24, 2013 Feb.
Article in English | MEDLINE | ID: mdl-23150521

ABSTRACT

How amphetamine affects the neuroglia in living brains is not well understood. In an effort to elucidate this effect, we investigated neuroglia in response to amphetamine exposure using antisense (AS) or sense (S) phosphorothioate-modified oligodeoxynucleotide (sODN) sequences that correspond to glial fibrillary acidic protein (GFAP) mRNA (AS-gfap or S-gfap, respectively) expression. The control is a random-sequence sODN (Ran). Using cyanine 5.5-superparamagnetic iron oxide nanoparticle (Cy5.5-SPION) labeling and fluorescent microscopy, we demonstrated that living neural progenitor cells (PC-12.1), as well as the cells in fresh brain slices and intact brains of male C57BL6 mice, exhibited universal uptake of all of the sODNs but rapidly excluded all sODN-Ran and most S-gfap. Moreover, transmission electron microscopy revealed electron-dense nanoparticles only in the neuroglia of normal or transgenic mice [B6;DBA-Tg(Fos-tTA, Fos-EGFP*)1MmayTg(tetO-lacZ,tTA*)1Mmay/J] that had been administered AS-gfap or Cy5.5-SPION-gfap. Subtraction R2* maps from mice with acute and chronic amphetamine exposure demonstrated, validated by postmortem immunohistochemistry, a reduction in striatal neuroglia, with gliogenesis in the subventricular zone and the somatosensory cortex in vivo. The sensitivity of our unique gene transcript targeted MRI was illustrated by a positive linear correlation (r(2)=1.0) between in vivo MRI signal changes and GFAP mRNA copy numbers determined by ex vivo quantitative RT-PCR. The study provides direct evidence for targeting neuroglia by antisense DNA-based SPION-gfap that enables in vivo MRI of inaccessible tissue with PCR sensitivity. The results enable us to conclude that amphetamine induces toxicity to neuroglia in vivo, which may cause remodeling or reconnectivity of neuroglia.


Subject(s)
Amphetamine/toxicity , Neuroglia/drug effects , Animals , Brain/drug effects , Brain/metabolism , Brain/ultrastructure , Carbocyanines/administration & dosage , Drug Delivery Systems , Glial Fibrillary Acidic Protein , Illicit Drugs/toxicity , Magnetic Resonance Imaging , Magnetite Nanoparticles/administration & dosage , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Microscopy, Electron, Transmission , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/metabolism , Neurogenesis/drug effects , Neuroglia/metabolism , Neuroglia/ultrastructure , Oligodeoxyribonucleotides, Antisense/administration & dosage , Oligodeoxyribonucleotides, Antisense/genetics , Oligodeoxyribonucleotides, Antisense/pharmacokinetics , PC12 Cells , RNA, Messenger/genetics , RNA, Messenger/metabolism , Rats
20.
Neurosciences (Riyadh) ; 19(2): 100-5, 2014 Apr.
Article in English | MEDLINE | ID: mdl-24739405

ABSTRACT

OBJECTIVE: To examine the effects of dehydroepiandrosterone (DHEA) on animal models of schizophrenia. METHODS: Seventy Swiss albino female mice (25-35 g) were divided into 4 groups: amphetamine-free (control), amphetamine, 50, and 100 mg/kg DHEA. The DHEA was administered intraperitoneally (ip) for 5 days. Amphetamine (3 mg/kg ip) induced hyper locomotion, apomorphine (1.5 mg/kg subcutaneously [sc]) induced climbing, and haloperidol (1.5 mg/kg sc) induced catalepsy tests were used as animal models of schizophrenia. The study was conducted at the Animal Experiment Laboratories, Department of Pharmacology, Medical School, Eskisehir Osmangazi University, Eskisehir, Turkey between March and May 2012. Statistical analysis was carried out using Kruskal-Wallis test for hyper locomotion, and one-way ANOVA for climbing and catalepsy tests. RESULTS: In the amphetamine-induced locomotion test, there were significant increases in all movements compared with the amphetamine-free group. Both DHEA 50 mg/kg (p<0.05), and 100 mg/kg (p<0.01) significantly decreased all movements compared with the amphetamine-induced locomotion group. There was a significant difference between groups in the haloperidol-induced catalepsy test (p<0.05). There was no significant difference between groups in terms of total climbing time in the apomorphine-induced climbing test (p>0.05). CONCLUSION: We observed that DHEA reduced locomotor activity and increased catalepsy at both doses, while it had no effect on climbing behavior. We suggest that DHEA displays typical neuroleptic-like effects, and may be used in the treatment of schizophrenia.


Subject(s)
Adjuvants, Immunologic/therapeutic use , Amphetamine/toxicity , Central Nervous System Stimulants/toxicity , Dehydroepiandrosterone/therapeutic use , Schizophrenia/chemically induced , Schizophrenia/drug therapy , Animals , Apomorphine , Catalepsy/chemically induced , Catalepsy/physiopathology , Disease Models, Animal , Dose-Response Relationship, Drug , Locomotion/drug effects , Mice , Schizophrenia/physiopathology , Statistics, Nonparametric , Time Factors
SELECTION OF CITATIONS
SEARCH DETAIL