1
|
Alipour E, Maleki S, Razavipour N, Hajali N, Jahani S. Identification of amphetamine as a stimulant drug by pristine and doped C 70 fullerenes: a DFT/TDDFT investigation. J Mol Model 2021; 27:169. [PMID: 33991237 DOI: 10.1007/s00894-021-04788-z] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2021] [Accepted: 05/09/2021] [Indexed: 11/26/2022]
Abstract
The density functional theory (DFT) was used to examine the electronic reactivity and sensitivity of a pristine, Si, and Al-doped fullerene C70 with AM drug. AM drug has been shown to be physically absorbed by its N-head on the pristine C70 with an adsorption energy of about - 1.09 kcal/mol and to have no impact on the electric conductivity of that cluster. The atom substitution of Si and Al for C atoms at C70 significantly increases C70 fullerene reactivity, with adsorption energy predictions of approximately - 31.09 and - 45.59 kcal/mol, respectively. The energy difference of LUMO and HOMO, i.e., Eg from C70 fullerene, significantly affects AM drug. Significant LUMO destabilization in Al-C70 by adsorption of the drug AM boosts the electrical conductivity of Al-C70 while generating electric signals that are related to the environmental presence of AM drug. Hence, Al-doped C70 is demonstrated to be an effective electronic AM drug sensor. In contrast to Si-C70 fullerene, significant AM-drug adsorption effects on Fermi and Si-C70 work functions make Si-C70 an Ф-type candidate for AM drug sensor applications. The time-dependent theory of the functional density shows that the AM/Al-C70 complex is steadily situated at a maximum peak of 784.15 nm.
Collapse
Affiliation(s)
- Elham Alipour
- Department of Organic Chemistry, Faculty of Chemistry, Bu-Ali Sina University, Hamedan, Iran.
| | - Shabnam Maleki
- Department of Chemistry, College of Science, Central Tehran Branch, Islamic Azad University, Tehran, Iran
| | - Negar Razavipour
- Department of environmental engineering, Central Tehran Branch, Islamic Azad University, Tehran, Iran
| | - Narjes Hajali
- Department of Chemistry, College of Science, Central Tehran Branch, Islamic Azad University, Tehran, Iran
| | | |
Collapse
|
2
|
Reynolds LM, Yetnikoff L, Pokinko M, Wodzinski M, Epelbaum JG, Lambert LC, Cossette MP, Arvanitogiannis A, Flores C. Early Adolescence is a Critical Period for the Maturation of Inhibitory Behavior. Cereb Cortex 2020; 29:3676-3686. [PMID: 30295713 DOI: 10.1093/cercor/bhy247] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2018] [Revised: 08/28/2018] [Accepted: 09/05/2018] [Indexed: 12/29/2022] Open
Abstract
Psychiatric conditions marked by impairments in cognitive control often emerge during adolescence, when the prefrontal cortex (PFC) and its inputs undergo structural and functional maturation and are vulnerable to disruption by external events. It is not known, however, whether there exists a specific temporal window within the broad range of adolescence when the development of PFC circuitry and its related behaviors are sensitive to disruption. Here we show, in male mice, that repeated exposure to amphetamine during early adolescence leads to impaired behavioral inhibition, aberrant PFC dopamine connectivity, and reduced PFC dopamine function in adulthood. Remarkably, these deficits are not observed following exposure to the exact same amphetamine regimen at later times. These findings demonstrate that there is a critical period for the disruption of the adolescent maturation of cognitive control and PFC dopamine function and suggest that early adolescence is particularly relevant to the emergence of psychopathology in humans.
Collapse
Affiliation(s)
- Lauren M Reynolds
- Integrated Program in Neuroscience, McGill University, Montréal, QC, Canada.,Department of Psychiatry and Department of Neurology and Neurosurgery, McGill University, Douglas Mental Health University Institute, Montréal, QC, Canada
| | - Leora Yetnikoff
- Department of Psychology, College of Staten Island, City University of New York, Staten Island, NY, USA.,CUNY Neuroscience Collaborative, The Graduate Center, City University of New York, New York, NY, USA
| | - Matthew Pokinko
- Integrated Program in Neuroscience, McGill University, Montréal, QC, Canada.,Department of Psychiatry and Department of Neurology and Neurosurgery, McGill University, Douglas Mental Health University Institute, Montréal, QC, Canada
| | - Michael Wodzinski
- Department of Psychiatry and Department of Neurology and Neurosurgery, McGill University, Douglas Mental Health University Institute, Montréal, QC, Canada
| | - Julia G Epelbaum
- Department of Psychiatry and Department of Neurology and Neurosurgery, McGill University, Douglas Mental Health University Institute, Montréal, QC, Canada
| | - Laura C Lambert
- Department of Psychiatry and Department of Neurology and Neurosurgery, McGill University, Douglas Mental Health University Institute, Montréal, QC, Canada
| | - Marie-Pierre Cossette
- Department of Psychology, Center for Studies in Behavioural Neurobiology, Concordia University, Montréal, QC, Canada
| | - Andreas Arvanitogiannis
- Department of Psychology, Center for Studies in Behavioural Neurobiology, Concordia University, Montréal, QC, Canada
| | - Cecilia Flores
- Department of Psychiatry and Department of Neurology and Neurosurgery, McGill University, Douglas Mental Health University Institute, Montréal, QC, Canada
| |
Collapse
|
3
|
Cuesta S, Restrepo-Lozano JM, Popescu C, He S, Reynolds LM, Israel S, Hernandez G, Rais R, Slusher BS, Flores C. DCC-related developmental effects of abused- versus therapeutic-like amphetamine doses in adolescence. Addict Biol 2020; 25:e12791. [PMID: 31192517 PMCID: PMC8301742 DOI: 10.1111/adb.12791] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2018] [Revised: 04/11/2019] [Accepted: 05/15/2019] [Indexed: 12/24/2022]
Abstract
The guidance cue receptor DCC controls mesocortical dopamine development in adolescence. Repeated exposure to an amphetamine regimen of 4 mg/kg during early adolescence induces, in male mice, downregulation of DCC expression in dopamine neurons by recruiting the Dcc microRNA repressor, microRNA-218 (miR-218). This adolescent amphetamine regimen also disrupts mesocortical dopamine connectivity and behavioral control in adulthood. Whether low doses of amphetamine in adolescence induce similar molecular and developmental effects needs to be established. Here, we quantified plasma amphetamine concentrations in early adolescent mice following a 4 or 0.5 mg/kg dose and found peak levels corresponding to those seen in humans following recreational and therapeutic settings, respectively. In contrast to the high doses, the low amphetamine regimen does not alter Dcc mRNA or miR-218 expression; instead, it upregulates DCC protein levels. Furthermore, high, but not low, drug doses downregulate the expression of the DCC receptor ligand, Netrin-1, in the nucleus accumbens and prefrontal cortex. Exposure to the low-dose regimen did not alter the expanse of mesocortical dopamine axons or their number/density of presynaptic sites in adulthood. Strikingly, adolescent exposure to the low-dose drug regimen does not impair behavioral inhibition in adulthood; instead, it induces an overall increase in performance in a go/no-go task. These results show that developmental consequences of exposure to therapeutic- versus abused-like doses of amphetamine in adolescence have dissimilar molecular signatures and opposite behavioral effects. These findings have important clinical relevance since amphetamines are widely used for therapeutic purposes in youth.
Collapse
Affiliation(s)
- Santiago Cuesta
- Department of Psychiatry and Department of Neurology and Neurosurgery, McGill University, Douglas Mental Health University Institute, Montreal, Quebec, Canada
| | - José Maria Restrepo-Lozano
- Department of Psychiatry and Department of Neurology and Neurosurgery, McGill University, Douglas Mental Health University Institute, Montreal, Quebec, Canada
- Integrated Program in Neuroscience, McGill University, Montreal, Quebec, Canada
| | - Christina Popescu
- Department of Psychiatry and Department of Neurology and Neurosurgery, McGill University, Douglas Mental Health University Institute, Montreal, Quebec, Canada
- Integrated Program in Neuroscience, McGill University, Montreal, Quebec, Canada
| | - Susan He
- Department of Psychiatry and Department of Neurology and Neurosurgery, McGill University, Douglas Mental Health University Institute, Montreal, Quebec, Canada
| | - Lauren M. Reynolds
- Department of Psychiatry and Department of Neurology and Neurosurgery, McGill University, Douglas Mental Health University Institute, Montreal, Quebec, Canada
- Integrated Program in Neuroscience, McGill University, Montreal, Quebec, Canada
| | - Sonia Israel
- Department of Psychiatry and Department of Neurology and Neurosurgery, McGill University, Douglas Mental Health University Institute, Montreal, Quebec, Canada
| | - Giovanni Hernandez
- Department of Psychiatry and Department of Neurology and Neurosurgery, McGill University, Douglas Mental Health University Institute, Montreal, Quebec, Canada
| | - Rana Rais
- Johns Hopkins Drug Discovery, Johns Hopkins School of Medicine, Baltimore, Maryland
| | - Barbara S. Slusher
- Johns Hopkins Drug Discovery, Johns Hopkins School of Medicine, Baltimore, Maryland
| | - Cecilia Flores
- Department of Psychiatry and Department of Neurology and Neurosurgery, McGill University, Douglas Mental Health University Institute, Montreal, Quebec, Canada
| |
Collapse
|
4
|
Petković B, Kesić S, Pešić V. Critical View on the Usage of Ribavirin in Already Existing Psychostimulant-Use Disorder. Curr Pharm Des 2020; 26:466-484. [PMID: 31939725 PMCID: PMC8383468 DOI: 10.2174/1381612826666200115094642] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2019] [Accepted: 12/21/2019] [Indexed: 12/12/2022]
Abstract
Substance-use disorder represents a frequently hidden non-communicable chronic disease. Patients with intravenous drug addiction are at high risk of direct exposure to a variety of viral infections and are considered to be the largest subpopulation infected with the hepatitis C virus. Ribavirin is a synthetic nucleoside analog that has been used as an integral component of hepatitis C therapy. However, ribavirin medication is quite often associated with pronounced psychiatric adverse effects. It is not well understood to what extent ribavirin per se contributes to changes in drug-related neurobehavioral disturbances, especially in the case of psychostimulant drugs, such as amphetamine. It is now well-known that repeated amphetamine usage produces psychosis in humans and behavioral sensitization in animals. On the other hand, ribavirin has an affinity for adenosine A1 receptors that antagonistically modulate the activity of dopamine D1 receptors, which play a critical role in the development of behavioral sensitization. This review will focus on the current knowledge of neurochemical/ neurobiological changes that exist in the psychostimulant drug-addicted brain itself and the antipsychotic-like efficiency of adenosine agonists. Particular attention will be paid to the potential side effects of ribavirin therapy, and the opportunities and challenges related to its application in already existing psychostimulant-use disorder.
Collapse
Affiliation(s)
- Branka Petković
- Address correspondence to this author at the Department of Neurophysiology, Institute for Biological Research “Siniša Stanković” - National Institute of Republic of Serbia, University of Belgrade, Despota Stefana Blvd. 142, 11060, Belgrade, Serbia; Tel: +381-11-20-78-300; Fax: +381-11-27-61-433; E-mail:
| | | | | |
Collapse
|
5
|
Abstract
One of the consequences of chronic methamphetamine (Meth) abuse and Meth addiction is impaired hippocampal function which plays a critical role in enhanced propensity for relapse. This impairment is predicted by alterations in hippocampal neurogenesis, structural- and functional-plasticity of granule cell neurons (GCNs), and expression of plasticity-related proteins in the dentate gyrus. This review will elaborate on the effects of Meth in animal models during different stages of addiction-like behavior on proliferation, differentiation, maturation, and survival of newly born neural progenitor cells. We will then discuss evidence for the contribution of adult neurogenesis in context-driven Meth-seeking behavior in animal models. These findings from interdisciplinary studies suggest that a subset of newly born GCNs contribute to context-driven Meth-seeking in Meth addicted animals.
Collapse
Affiliation(s)
- Yoshio Takashima
- Department of Anesthesiology, University of California San Diego, VA San Diego Healthcare System, San Diego, CA, USA
| | - Chitra D. Mandyam
- Department of Anesthesiology, University of California San Diego, VA San Diego Healthcare System, San Diego, CA, USA
| |
Collapse
|
6
|
Dopamine Development in the Mouse Orbital Prefrontal Cortex Is Protracted and Sensitive to Amphetamine in Adolescence. eNeuro 2018; 5:eN-NWR-0372-17. [PMID: 29333488 PMCID: PMC5762649 DOI: 10.1523/eneuro.0372-17.2017] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2017] [Revised: 12/14/2017] [Accepted: 12/15/2017] [Indexed: 02/04/2023] Open
Abstract
The prefrontal cortex (PFC) is divided into subregions, including the medial and orbital prefrontal cortices. Dopamine connectivity in the medial PFC (mPFC) continues to be established throughout adolescence as the result of the continuous growth of axons that innervated the nucleus accumbens (NAcc) prior to adolescence. During this period, dopamine axons remain vulnerable to environmental influences, such as drugs used recreationally by humans. The developmental trajectory of the orbital prefrontal dopamine innervation remains almost completely unstudied. Nonetheless, the orbital PFC (oPFC) is critical for some of the most complex functions of the PFC and is disrupted by drugs of abuse, both in adolescent humans and rodents. Here, we use quantitative neuroanatomy, axon-initiated viral-vector recombination, and pharmacology in mice to determine the spatiotemporal development of the dopamine innervation to the oPFC and its vulnerability to amphetamine in adolescence. We find that dopamine innervation to the oPFC also continues to increase during adolescence and that this increase is due to the growth of new dopamine axons to this region. Furthermore, amphetamine in adolescence dramatically reduces the number of presynaptic sites on oPFC dopamine axons. In contrast, dopamine innervation to the piriform cortex is not protracted across adolescence and is not impacted by amphetamine exposure during adolescence, indicating that dopamine development during adolescence is a uniquely prefrontal phenomenon. This renders these fibers, and the PFC in general, particularly vulnerable to environmental risk factors during adolescence, such as recreational drug use.
Collapse
|
7
|
Exercise protects against methamphetamine-induced aberrant neurogenesis. Sci Rep 2016; 6:34111. [PMID: 27677455 PMCID: PMC5039713 DOI: 10.1038/srep34111] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2016] [Accepted: 09/06/2016] [Indexed: 12/15/2022] Open
Abstract
While no effective therapy is available for the treatment of methamphetamine (METH)-induced neurotoxicity, aerobic exercise is being proposed to improve depressive symptoms and substance abuse outcomes. The present study focuses on the effect of exercise on METH-induced aberrant neurogenesis in the hippocampal dentate gyrus in the context of the blood-brain barrier (BBB) pathology. Mice were administered with METH or saline by i.p. injections for 5 days with an escalating dose regimen. One set of mice was sacrificed 24 h post last injection of METH, and the remaining animals were either subjected to voluntary wheel running (exercised mice) or remained in sedentary housing (sedentary mice). METH administration decreased expression of tight junction (TJ) proteins and increased BBB permeability in the hippocampus. These changes were preserved post METH administration in sedentary mice and were associated with the development of significant aberrations of neural differentiation. Exercise protected against these effects by enhancing the protein expression of TJ proteins, stabilizing the BBB integrity, and enhancing the neural differentiation. In addition, exercise protected against METH-induced systemic increase in inflammatory cytokine levels. These results suggest that exercise can attenuate METH-induced neurotoxicity by protecting against the BBB disruption and related microenvironmental changes in the hippocampus.
Collapse
|
8
|
Krasnova IN, Justinova Z, Cadet JL. Methamphetamine addiction: involvement of CREB and neuroinflammatory signaling pathways. Psychopharmacology (Berl) 2016; 233:1945-62. [PMID: 26873080 PMCID: PMC5627363 DOI: 10.1007/s00213-016-4235-8] [Citation(s) in RCA: 85] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/29/2015] [Accepted: 02/04/2016] [Indexed: 02/06/2023]
Abstract
RATIONALE AND OBJECTIVES Addiction to psychostimulant methamphetamine (METH) remains a major public health problem in the world. Animal models that use METH self-administration incorporate many features of human drug-taking behavior and are very helpful in elucidating mechanisms underlying METH addiction. These models are also helping to decipher the neurobiological substrates of associated neuropsychiatric complications. This review summarizes our work on the influence of METH self-administration on dopamine systems, transcription and immune responses in the brain. METHODS We used the rat model of METH self-administration with extended access (15 h/day for eight consecutive days) to investigate the effects of voluntary METH intake on the markers of dopamine system integrity and changes in gene expression observed in the brain at 2 h-1 month after cessation of drug exposure. RESULTS Extended access to METH self-administration caused changes in the rat brain that are consistent with clinical findings reported in neuroimaging and postmortem studies of human METH addicts. In addition, gene expression studies using striatal tissues from METH self-administering rats revealed increased expression of genes involved in cAMP response element binding protein (CREB) signaling pathway and in the activation of neuroinflammatory response in the brain. CONCLUSION These data show an association of METH exposure with activation of neuroplastic and neuroinflammatory cascades in the brain. The neuroplastic changes may be involved in promoting METH addiction. Neuroinflammatory processes in the striatum may underlie cognitive deficits, depression, and parkinsonism reported in METH addicts. Therapeutic approaches that include suppression of neuroinflammation may be beneficial to addicted patients.
Collapse
Affiliation(s)
- Irina N. Krasnova
- Molecular Neuropsychiatry Research Branch, Intramural Research Program, NIDA, NIH, DHHS, Baltimore, MD, USA,Corresponding authors: Irina N. Krasnova, Ph.D., Molecular Neuropsychiatry Research Branch, NIDA/NIH/DHHS, 251 Bayview Blvd, Baltimore, MD 21224, Tel. 443-74-2658, Fax 443-740-2856, , Jean Lud Cadet, M.D., Molecular Neuropsychiatry Research Branch, NIDA/NIH/DHHS, 251 Bayview Blvd., Baltimore, MD 21224, Tel. 443-740-2656, Fax 443-740-2856,
| | - Zuzana Justinova
- Behavioral Neuroscience Research Branch, Intramural Research Program, NIDA, NIH, DHHS Baltimore, MD 21224, USA
| | - Jean Lud Cadet
- Molecular Neuropsychiatry Research Branch, Intramural Research Program, NIDA, NIH, DHHS, 251 Bayview Blvd, Baltimore, MD, 21224, USA.
| |
Collapse
|
9
|
Somkuwar SS, Staples MC, Fannon MJ, Ghofranian A, Mandyam CD. Evaluating Exercise as a Therapeutic Intervention for Methamphetamine Addiction-Like Behavior. Brain Plast 2015; 1:63-81. [PMID: 29765835 PMCID: PMC5928557 DOI: 10.3233/bpl-150007] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Abstract
The need for effective treatments for addiction and dependence to the illicit stimulant methamphetamine in primary care settings is increasing, yet no effective medications have been FDA approved to reduce dependence [1]. This is partially attributed to the complex and dynamic neurobiology underlying the various stages of addiction [2]. Therapeutic strategies to treat methamphetamine addiction, particularly the relapse stage of addiction, could revolutionize methamphetamine addiction treatment. In this context, preclinical studies demonstrate that voluntary exercise (sustained physical activity) could be used as an intervention to reduce methamphetamine addiction. Therefore, it appears that methamphetamine disrupts normal functioning in the brain and this disruption is prevented or reduced by engaging in exercise. This review discusses animal models of methamphetamine addiction and sustained physical activity and the interactions between exercise and methamphetamine behaviors. The review highlights how methamphetamine and exercise affect neuronal plasticity and neurotoxicity in the adult mammalian striatum, hippocampus, and prefrontal cortex, and presents the emerging mechanisms of exercise in attenuating intake and in preventing relapse to methamphetamine seeking in preclinical models of methamphetamine addiction.
Collapse
Affiliation(s)
- Sucharita S Somkuwar
- Committee on the Neurobiology of Addictive Disorders, The Scripps Research Institute, La Jolla, CA, USA
| | - Miranda C Staples
- Committee on the Neurobiology of Addictive Disorders, The Scripps Research Institute, La Jolla, CA, USA
| | - McKenzie J Fannon
- Committee on the Neurobiology of Addictive Disorders, The Scripps Research Institute, La Jolla, CA, USA
| | - Atoosa Ghofranian
- Committee on the Neurobiology of Addictive Disorders, The Scripps Research Institute, La Jolla, CA, USA
| | - Chitra D Mandyam
- Committee on the Neurobiology of Addictive Disorders, The Scripps Research Institute, La Jolla, CA, USA
| |
Collapse
|
10
|
Abstract
Drug addiction is characterized by uncontrolled drug consumption and high rates of relapse to drug taking during periods of attempted abstinence. Addiction is now largely considered a disorder of experience-dependent neuroplasticity, driven by remodeling of synapses in reward and motivation relevant brain circuits in response to a history of prolonged drug intake. Alterations in gene expression play a central role in addiction-relevant neuroplasticity, but the mechanisms by which additive drugs remodel brain motivation circuits remains unclear. MicroRNAs (miRNAs) are a class of noncoding RNA that can regulate the expression of large numbers of protein-coding mRNA transcripts by binding to the 3' untranslated region (3' UTR) of target transcripts and blocking their translation into the encoded protein or triggering their destabilization and degradation. Emerging evidence has implicated miRNAs in regulating addiction-relevant neuroplasticity in the brain, and in controlling the motivational properties of cocaine and other drugs of abuse. Here, the role for miRNAs in regulating basic aspects of neuronal function is reviewed. The involvement of miRNAs in controlling the motivational properties of addictive drugs is also summarized. Finally, mechanisms by which miRNAs exert their actions on drug intake, when known, are considered.
Collapse
Affiliation(s)
- Paul J Kenny
- Laboratory of Behavioral & Molecular Neuroscience, Department of Pharmacology & Systems Therapeutics, Icahn School of Medicine at Mount Sinai, New York, USA
| |
Collapse
|
11
|
Amphetamine in adolescence disrupts the development of medial prefrontal cortex dopamine connectivity in a DCC-dependent manner. Neuropsychopharmacology 2015; 40:1101-12. [PMID: 25336209 PMCID: PMC4367452 DOI: 10.1038/npp.2014.287] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/27/2014] [Revised: 10/04/2014] [Accepted: 10/15/2014] [Indexed: 01/25/2023]
Abstract
Initiation of drug use during adolescence is a strong predictor of both the incidence and severity of addiction throughout the lifetime. Intriguingly, adolescence is a period of dynamic refinement in the organization of neuronal connectivity, in particular medial prefrontal cortex (mPFC) dopamine circuitry. The guidance cue receptor, DCC (deleted in colorectal cancer), is highly expressed by dopamine neurons and orchestrates their innervation to the mPFC during adolescence. Furthermore, we have shown that amphetamine in adolescence regulates DCC expression in dopamine neurons. Drugs in adolescence may therefore induce their enduring behavioral effects via DCC-mediated disruption in mPFC dopamine development. In this study, we investigated the impact of repeated exposure to amphetamine during adolescence on both the development of mPFC dopamine connectivity and on salience attribution to drug context in adulthood. We compare these effects to those induced by adult exposure to an identical amphetamine regimen. Finally, we determine whether DCC signaling within dopamine neurons is necessary for these events. Exposure to amphetamine in adolescence, but not in adulthood, leads to an increase in the span of dopamine innervation to the mPFC, but a reduction of presynaptic sites present on these axons. Amphetamine treatment in adolescence, but not in adulthood, also produces an increase in salience attribution to a previously drug-paired context in adulthood. Remarkably, DCC signaling within dopamine neurons is required for both of these effects. Drugs of abuse in adolescence may therefore induce their detrimental behavioral consequences by disrupting mesocortical dopamine development through alterations in the DCC signaling cascade.
Collapse
|
12
|
Jayanthi S, McCoy MT, Chen B, Britt JP, Kourrich S, Yau HJ, Ladenheim B, Krasnova IN, Bonci A, Cadet JL. Methamphetamine downregulates striatal glutamate receptors via diverse epigenetic mechanisms. Biol Psychiatry 2014; 76:47-56. [PMID: 24239129 PMCID: PMC3989474 DOI: 10.1016/j.biopsych.2013.09.034] [Citation(s) in RCA: 95] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/29/2013] [Revised: 09/27/2013] [Accepted: 09/30/2013] [Indexed: 11/16/2022]
Abstract
BACKGROUND Chronic methamphetamine (METH) exposure causes neuroadaptations at glutamatergic synapses. METHODS To identify the METH-induced epigenetic underpinnings of these neuroadaptations, we injected increasing METH doses to rats for 2 weeks and measured striatal glutamate receptor expression. We then quantified the effects of METH exposure on histone acetylation. We also measured METH-induced changes in DNA methylation and DNA hydroxymethylation. RESULTS Chronic METH decreased transcript and protein expression of GluA1 and GluA2 alpha-amino-3-hydroxy-5-methyl-4-isoxazole propionic acid receptor (AMPAR) and GluN1 N-methyl-D-aspartate receptor subunits. These changes were associated with altered electrophysiological glutamatergic responses in striatal neurons. Chromatin immunoprecipitation-polymerase chain reaction revealed that METH decreased enrichment of acetylated histone H4 on GluA1, GluA2, and GluN1 promoters. Methamphetamine exposure also increased repressor element-1 silencing transcription factor (REST) corepressor 1, methylated CpG binding protein 2, and histone deacetylase 2 enrichment, but not of sirtuin 1 or sirtuin 2, onto GluA1 and GluA2 gene sequences. Moreover, METH caused interactions of REST corepressor 1 and methylated CpG binding protein 2 with histone deacetylase 2 and of REST with histone deacetylase 1. Surprisingly, methylated DNA immunoprecipitation and hydroxymethylated DNA immunoprecipitation-polymerase chain reaction revealed METH-induced decreased enrichment of 5-methylcytosine and 5-hydroxymethylcytosine at GluA1 and GluA2 promoter sequences. Importantly, the histone deacetylase inhibitor, valproic acid, blocked METH-induced decreased expression of AMPAR and N-methyl-D-aspartate receptor subunits. Finally, valproic acid also attenuated METH-induced decrease H4K16Ac recruitment on AMPAR gene sequences. CONCLUSIONS These observations suggest that histone H4 hypoacetylation may be the main determinant of METH-induced decreased striatal glutamate receptor expression.
Collapse
Affiliation(s)
- Subramaniam Jayanthi
- Molecular Neuropsychiatry Research Branch, US Department of Health and Human Services/National Institutes of Health/National Institute on Drug Abuse/Intramural Research Program, Baltimore, MD
| | - Michael T McCoy
- Molecular Neuropsychiatry Research Branch, US Department of Health and Human Services/National Institutes of Health/National Institute on Drug Abuse/Intramural Research Program, Baltimore, MD
| | - Billy Chen
- Synaptic Plasticity Section, US Department of Health and Human Services/National Institutes of Health/National Institute on Drug Abuse/Intramural Research Program, Baltimore, MD
| | - Jonathan P Britt
- Synaptic Plasticity Section, US Department of Health and Human Services/National Institutes of Health/National Institute on Drug Abuse/Intramural Research Program, Baltimore, MD
| | - Saїd Kourrich
- Synaptic Plasticity Section, US Department of Health and Human Services/National Institutes of Health/National Institute on Drug Abuse/Intramural Research Program, Baltimore, MD
| | - Hau-Jie Yau
- Synaptic Plasticity Section, US Department of Health and Human Services/National Institutes of Health/National Institute on Drug Abuse/Intramural Research Program, Baltimore, MD
| | - Bruce Ladenheim
- Molecular Neuropsychiatry Research Branch, US Department of Health and Human Services/National Institutes of Health/National Institute on Drug Abuse/Intramural Research Program, Baltimore, MD
| | - Irina N Krasnova
- Molecular Neuropsychiatry Research Branch, US Department of Health and Human Services/National Institutes of Health/National Institute on Drug Abuse/Intramural Research Program, Baltimore, MD
| | - Antonello Bonci
- Synaptic Plasticity Section, US Department of Health and Human Services/National Institutes of Health/National Institute on Drug Abuse/Intramural Research Program, Baltimore, MD
| | - Jean Lud Cadet
- Molecular Neuropsychiatry Research Branch, US Department of Health and Human Services/National Institutes of Health/National Institute on Drug Abuse/Intramural Research Program, Baltimore, MD.
| |
Collapse
|
13
|
Banks ML, Worst TJ, Rusyniak DE, Sprague JE. Synthetic cathinones ("bath salts"). J Emerg Med 2014; 46:632-42. [PMID: 24565885 DOI: 10.1016/j.jemermed.2013.11.104] [Citation(s) in RCA: 75] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2013] [Revised: 10/11/2013] [Accepted: 11/17/2013] [Indexed: 11/18/2022]
Abstract
BACKGROUND Synthetic cathinones are popularly referred to in the media as "bath salts." Through the direct and indirect activation of the sympathetic nervous system, smoking, snorting, or injecting synthetic cathinones can result in tachycardia, hypertension, hyperthermia, myocardial infarction, and death. OBJECTIVE The chemical structures and names of bath salts identified by the Ohio Attorney General's Bureau of Criminal Investigation are presented. Based on their common pharmacophores, we review the history, pharmacology, toxicology, detection methods, and clinical implications of synthetic cathinones. Through the integration of this information, the pharmacological basis for the management of patients using synthetic cathinones is presented. DISCUSSION Synthetic cathinones activate central serotonergic and dopaminergic systems contributing to acute psychosis and the peripheral activation of the sympathetic nervous system. The overstimulation of the sympathetic nervous system contributes to the many toxicities reported with bath salt use. The pharmacological basis for managing these patients is targeted at attenuating the activation of these systems. CONCLUSIONS Treatment of patients presenting after using bath salts should be focused on reducing agitation and psychosis and supporting renal perfusion. The majority of successfully treated synthetic cathinones cases have used benzodiazepines and antipsychotics along with general supportive care.
Collapse
Affiliation(s)
- Matthew L Banks
- Department of Pharmacology and Toxicology, Virginia Commonwealth University, Richmond, Virginia
| | - Travis J Worst
- Ohio Attorney General's Bureau of Criminal Investigation, London, Ohio
| | - Daniel E Rusyniak
- Departments of Emergency Medicine and Pharmacology and Toxicology, Indiana University School of Medicine, Indianapolis, Indiana
| | - Jon E Sprague
- Department of Pharmaceutical Sciences, College of Pharmacy, Ferris State University, Big Rapids, Michigan
| |
Collapse
|
14
|
Abstract
Stereotypies that develop spontaneously in nonhuman primates can provide an effective model for repetitive stereotyped behavior in people with neurodevelopmental or obsessive-compulsive disorders. The behaviors are similar in form, are similarly affected by environmental conditions, and are improved with similar treatment methods such as enrichment, training, and drug therapy. However, because of a greater number of commonalities in these factors, nonhuman primates may serve as a better model for stereotyped behavior in individuals with autism or intellectual disability than for compulsions in individuals with obsessive-compulsive disorder. Because animal models may not be exact in all features of the disorder being studied, it is important to investigate the strengths and weaknesses of using a nonhuman primate model for stereotyped behavior in people with psychological disorders.
Collapse
|
15
|
Cadet JL, Jayanthi S, McCoy MT, Ladenheim B, Saint-Preux F, Lehrmann E, De S, Becker KG, Brannock C. Genome-wide profiling identifies a subset of methamphetamine (METH)-induced genes associated with METH-induced increased H4K5Ac binding in the rat striatum. BMC Genomics 2013; 14:545. [PMID: 23937714 PMCID: PMC3751638 DOI: 10.1186/1471-2164-14-545] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2013] [Accepted: 07/20/2013] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND METH is an illicit drug of abuse that influences gene expression in the rat striatum. Histone modifications regulate gene transcription. METHODS We therefore used microarray analysis and genome-scale approaches to examine potential relationships between the effects of METH on gene expression and on DNA binding of histone H4 acetylated at lysine 4 (H4K5Ac) in the rat dorsal striatum of METH-naïve and METH-pretreated rats. RESULTS Acute and chronic METH administration caused differential changes in striatal gene expression. METH also increased H4K5Ac binding around the transcriptional start sites (TSSs) of genes in the rat striatum. In order to relate gene expression to histone acetylation, we binned genes of similar expression into groups of 100 genes and proceeded to relate gene expression to H4K5Ac binding. We found a positive correlation between gene expression and H4K5Ac binding in the striatum of control rats. Similar correlations were observed in METH-treated rats. Genes that showed acute METH-induced increased expression in saline-pretreated rats also showed METH-induced increased H4K5Ac binding. The acute METH injection caused similar increases in H4K5Ac binding in METH-pretreated rats, without affecting gene expression to the same degree. Finally, genes that showed METH-induced decreased expression exhibited either decreases or no changes in H4K5Ac binding. CONCLUSION Acute METH injections caused increased gene expression of genes that showed increased H4K5Ac binding near their transcription start sites.
Collapse
Affiliation(s)
- Jean Lud Cadet
- Molecular Neuropsychiatry Research Branch, DHHS/NIH/NIDA Intramural Research Program, 251 Bayview Boulevard, Baltimore, MD 21224, USA.
| | | | | | | | | | | | | | | | | |
Collapse
|
16
|
Illicit drug use and problem gambling. ISRN ADDICTION 2013; 2013:342392. [PMID: 25938114 PMCID: PMC4392972 DOI: 10.1155/2013/342392] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 06/07/2013] [Accepted: 07/18/2013] [Indexed: 11/17/2022]
Abstract
Problem gambling, substance use disorders, and their cooccurrence are serious public health concerns. We conducted a comprehensive review of the literature to understand the present state of the evidence on these coaddictions. Our main focus was illicit drug use rather than misuse of legal substances. The review covers issues related to gambling as a hidden problem in the illicit drug use community; prevalence, problem gambling, and substance use disorders as kindred afflictions; problem gambling as an addiction similar to illicit drug use; risk factors and problems associated with comorbidity, and gender issues. We end with some suggestions for future research.
Collapse
|
17
|
Jonkman S, Kenny PJ. Molecular, cellular, and structural mechanisms of cocaine addiction: a key role for microRNAs. Neuropsychopharmacology 2013; 38:198-211. [PMID: 22968819 PMCID: PMC3521966 DOI: 10.1038/npp.2012.120] [Citation(s) in RCA: 46] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
Abstract
The rewarding properties of cocaine play a key role in establishing and maintaining the drug-taking habit. However, as exposure to cocaine increases, drug use can transition from controlled to compulsive. Importantly, very little is known about the neurobiological mechanisms that control this switch in drug use that defines addiction. MicroRNAs (miRNAs) are small non-protein coding RNA transcripts that can regulate the expression of messenger RNAs that code for proteins. Because of their highly pleiotropic nature, each miRNA has the potential to regulate hundreds or even thousands of protein-coding RNA transcripts. This property of miRNAs has generated considerable interest in their potential involvement in complex psychiatric disorders such as addiction, as each miRNA could potentially influence the many different molecular and cellular adaptations that arise in response to drug use that are hypothesized to drive the emergence of addiction. Here, we review recent evidence supporting a key role for miRNAs in the ventral striatum in regulating the rewarding and reinforcing properties of cocaine in animals with limited exposure to the drug. Moreover, we discuss evidence suggesting that miRNAs in the dorsal striatum control the escalation of drug intake in rats with extended cocaine access. These findings highlight the central role for miRNAs in drug-induced neuroplasticity in brain reward systems that drive the emergence of compulsive-like drug use in animals, and suggest that a better understanding of how miRNAs control drug intake will provide new insights into the neurobiology of drug addiction.
Collapse
Affiliation(s)
- Sietse Jonkman
- Laboratory of Behavioral and Molecular Neuroscience, Departments of Molecular Therapeutics and Neuroscience, The Scripps Research Institute–Florida, Jupiter, FL, USA
| | - Paul J Kenny
- Laboratory of Behavioral and Molecular Neuroscience, Departments of Molecular Therapeutics and Neuroscience, The Scripps Research Institute–Florida, Jupiter, FL, USA,Laboratory of Behavioral and Molecular Neuroscience, Department of Molecular Therapeutics, The Scripps Research Institute—Florida, 130 Scripps Way, Jupiter, FL 33458, USA, Tel: +1 561 228 2231, Fax: +1 561 799 8961, E-mail:
| |
Collapse
|
18
|
Dean AC, Groman SM, Morales AM, London ED. An evaluation of the evidence that methamphetamine abuse causes cognitive decline in humans. Neuropsychopharmacology 2013; 38:259-74. [PMID: 22948978 PMCID: PMC3527116 DOI: 10.1038/npp.2012.179] [Citation(s) in RCA: 177] [Impact Index Per Article: 14.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Methamphetamine (MA) is one of the most commonly abused illicit substances worldwide. Among other problems, abuse of the drug has been associated with reduced cognitive function across several domains. However, much of the literature has not attempted to differentiate cognitive difficulties caused by MA abuse from preexisting cognitive difficulties that are likely caused by other factors. Here, we address this question, evaluating evidence for a priori hypotheses pertaining to six lines of research: (a) animal studies; (b) cross-sectional human studies; (c) a twin study; (d) studies of changes in cognition with abstinence from MA; (e) studies of changes in brain structure and function with abstinence from MA; and (f) studies of the relationship between the severity of MA abuse and the extent of cognitive deficits observed. Overall the findings were mixed, with some support for a causal relationship between MA abuse and cognitive decline, and other findings suggesting that there is no relationship. The preponderance of the data, however, does support the possibility that MA abuse causes cognitive decline, of unknown duration, in at least some users of the drug. When averaged across individuals, this decline is likely to be mild in early-to-middle adulthood. However, moderator variables are likely to contribute to the presence and/or severity of cognitive decline exhibited by a given individual.
Collapse
Affiliation(s)
- Andy C Dean
- Department of Psychiatry and Biobehavioral Sciences and Semel Institute, David Geffen School of Medicine, University of California, Los Angeles, CA 90095-1759, USA.
| | - Stephanie M Groman
- Department of Psychiatry and Biobehavioral Sciences and Semel Institute, David Geffen School of Medicine, University of California, Los Angeles, CA, USA
| | - Angelica M Morales
- Department of Psychiatry and Biobehavioral Sciences and Semel Institute, David Geffen School of Medicine, University of California, Los Angeles, CA, USA
| | - Edythe D London
- Department of Psychiatry and Biobehavioral Sciences and Semel Institute, David Geffen School of Medicine, University of California, Los Angeles, CA, USA,Department of Molecular and Medical Pharmacology, and the Brain Research Institute, David Geffen School of Medicine, University of California, Los Angeles, CA, USA
| |
Collapse
|
19
|
Possible involvement of supersensitivity in cerebral ?-adrenergic and dopaminergic receptors in the occurrence of sensitization to d-methamphetamine in mice. Neurochem Int 2012; 16:17-25. [PMID: 20504536 DOI: 10.1016/0197-0186(90)90119-e] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/1989] [Accepted: 07/11/1989] [Indexed: 11/24/2022]
Abstract
Behavioral sensitization of mice (male STD-ddy) to d-methamphetamine (MAP) and its correlation with the neurochemical alterations in cerebral catecholaminergic systems were investigated following the repeated administration of MAP (5 mg/kg, i.p. x 2/day, 5 days). MAP-pretreated mice showed an enhancement in MAP-stimulated locomotor activity following the readministration of MAP (0.625 mg/kg). The metabolic turnovers of cerebral norepinephrine (NE) and dopamine (DA) showed a significant decrease in these sensitized mice. The activities of tyrosine hydroxylase. DOPA decarboxylase, monoamine oxidase and catechol-O-methyltransferase, however, showed no change under the same experimental conditions. On the other hand, the repeated MAP pretreatment induced the enhancement of the specific bindings of [(3)H]dihydroalprenolol and [(3)H]spiperone to cerebral synaptic membrane, which resulted from the increase of B(max) values. Furthermore, pretreatments of mice with propranolol (10 mg/kg) and haloperidol (0.1 mg/kg) inhibited the development of behavioral sensitization to MAP. These results suggest that the supersensitivity in cerebral ?-adrenergic and dopaminergic receptors may be involved in behavioral sensitization to MAP.
Collapse
|
20
|
Hadamitzky M, McCunney S, Markou A, Kuczenski R. Development of stereotyped behaviors during prolonged escalation of methamphetamine self-administration in rats. Psychopharmacology (Berl) 2012; 223:259-69. [PMID: 22526541 PMCID: PMC3586274 DOI: 10.1007/s00213-012-2713-1] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/01/2011] [Accepted: 03/29/2012] [Indexed: 01/28/2023]
Abstract
RATIONALE Experimental animal studies have shown that repeated administration of psychostimulants, such as methamphetamine (METH), results in an altered behavioral response profile, which includes the sensitization of both locomotor and stereotyped behaviors. Although sensitization of these behaviors has been characterized in detail during bolus, investigator-administered drug administration, little is known about the development or expression of stereotypies during psychostimulant self-administration. OBJECTIVE/METHODS The present study investigated in rats the expression of focused stereotyped behaviors during an extended access, escalation procedure of METH self-administration. Over several weeks during stepwise-extended daily access to METH (3, 6, and 12 h) followed by exposure to 24-h "binges," rats gradually increased daily drug intake. RESULTS During the escalation procedure, the rats' behavioral response evolved from locomotor activation to progressively more focused stereotypies, culminating in continuous oral behaviors (licking, gnawing, and chewing), interrupted only by episodic lever presses. Sensitization of stereotyped behaviors was evident, particularly with regard to oral behaviors that exhibited a more rapid onset and intensification in the apparent absence of greater drug intake. CONCLUSIONS Our data demonstrate that stepwise-extended daily access to METH (3, 6, 12, and 24 h) self-administration in rats closely approximates motivational, pharmacokinetic, as well as behavioral patterns of human METH abuse. The accompanied appearance of sensitization of intense focused stereotyped behaviors, which is probably a consequence of escalation of drug intake, resembles stereotypies associated with investigator-initiated METH administration and may parallel the development of stimulant-induced psychosis seen in human abusers.
Collapse
Affiliation(s)
- Martin Hadamitzky
- Department of Psychiatry, University of California San Diego, La Jolla, CA 92093, USA.
| | | | | | | |
Collapse
|
21
|
Hodges AB, Ladenheim B, McCoy MT, Beauvais G, Cai N, Krasnova IN, Cadet JL. Long-term protective effects of methamphetamine preconditioning against single-day methamphetamine toxic challenges. Curr Neuropharmacol 2011; 9:35-9. [PMID: 21886558 PMCID: PMC3137197 DOI: 10.2174/157015911795017344] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2009] [Revised: 04/17/2010] [Accepted: 05/26/2010] [Indexed: 11/22/2022] Open
Abstract
Methamphetamine (METH) use is associated with neurotoxic effects which include decreased levels of dopamine (DA), serotonin (5-HT) and their metabolites in the brain. We have shown that escalating METH dosing can protect against METH induced neurotoxicity in rats sacrificed within 24 hours after a toxic METH challenge. The purpose of the current study was to investigate if the protective effects of METH persisted for a long period of time. We also tested if a second challenge with a toxic dose of METH would cause further damage to monoaminergic terminals. Saline-pretreated rats showed significant METH-induced decreases in striatal DA and 5-HT levels in rats sacrificed 2 weeks after the challenge. Rats that received two METH challenges showed no further decreases in striatal DA or 5-HT levels in comparison to the single METH challenge. In contrast, METH-pretreated rats showed significant protection against METH-induced striatal DA and 5-HT depletion. In addition, the METH challenge causes substantial decreases in cortical 5-HT levels which were not further potentiated by a second drug challenge. METH preconditioning provided almost complete protection against METH -induced 5-HT depletion. These results are consistent with the idea that METH pretreatment renders the brain refractory to METH-induced degeneration of brain monoaminergic systems.
Collapse
Affiliation(s)
- A B Hodges
- Molecular Neuropsychiatry Research Branch, Intramural Research Program, National Institute on Drug Abuse, NIH, DHHS, Baltimore, MD 21224, USA
| | | | | | | | | | | | | |
Collapse
|
22
|
Kitanaka N, Kitanaka J, Hall FS, Uhl GR, Watabe K, Kubo H, Takahashi H, Tatsuta T, Morita Y, Takemura M. A single administration of methamphetamine to mice early in the light period decreases running wheel activity observed during the dark period. Brain Res 2011; 1429:155-63. [PMID: 22079320 DOI: 10.1016/j.brainres.2011.10.037] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2011] [Revised: 09/28/2011] [Accepted: 10/20/2011] [Indexed: 10/15/2022]
Abstract
Repeated intermittent administration of amphetamines acutely increases appetitive and consummatory aspects of motivated behaviors as well as general activity and exploratory behavior, including voluntary running wheel activity. Subsequently, if the drug is withdrawn, the frequency of these behaviors decreases, which is thought to be indicative of dysphoric symptoms associated with amphetamine withdrawal. Such decreases may be observed after chronic treatment or even after single drug administrations. In the present study, the effect of acute methamphetamine (METH) on running wheel activity, horizontal locomotion, appetitive behavior (food access), and consummatory behavior (food and water intake) was investigated in mice. A multi-configuration behavior apparatus designed to monitor the five behaviors was developed, where combined measures were recorded simultaneously. In the first experiment, naïve male ICR mice showed gradually increasing running wheel activity over three consecutive days after exposure to a running wheel, while mice without a running wheel showed gradually decreasing horizontal locomotion, consistent with running wheel activity being a positively motivated form of natural motor activity. In experiment 2, increased horizontal locomotion and food access, and decreased food intake, were observed for the initial 3h after acute METH challenge. Subsequently, during the dark phase period decreased running wheel activity and horizontal locomotion were observed. The reductions in running wheel activity and horizontal locomotion may be indicative of reduced dopaminergic function, although it remains to be seen if these changes may be more pronounced after more prolonged METH treatments.
Collapse
Affiliation(s)
- Nobue Kitanaka
- Department of Pharmacology, Hyogo College of Medicine, Hyogo 663-8501, Japan
| | | | | | | | | | | | | | | | | | | |
Collapse
|
23
|
Krasnova IN, Ladenheim B, Hodges AB, Volkow ND, Cadet JL. Chronic methamphetamine administration causes differential regulation of transcription factors in the rat midbrain. PLoS One 2011; 6:e19179. [PMID: 21547080 PMCID: PMC3081849 DOI: 10.1371/journal.pone.0019179] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2010] [Accepted: 03/23/2011] [Indexed: 12/12/2022] Open
Abstract
Methamphetamine (METH) is an addictive and neurotoxic psychostimulant widely abused in the USA and throughout the world. When administered in large doses, METH can cause depletion of striatal dopamine terminals, with preservation of midbrain dopaminergic neurons. Because alterations in the expression of transcription factors that regulate the development of dopaminergic neurons might be involved in protecting these neurons after toxic insults, we tested the possibility that their expression might be affected by toxic doses of METH in the adult brain. Male Sprague-Dawley rats pretreated with saline or increasing doses of METH were challenged with toxic doses of the drug and euthanized two weeks later. Animals that received toxic METH challenges showed decreases in dopamine levels and reductions in tyrosine hydroxylase protein concentration in the striatum. METH pretreatment protected against loss of striatal dopamine and tyrosine hydroxylase. In contrast, METH challenges caused decreases in dopamine transporters in both saline- and METH-pretreated animals. Interestingly, METH challenges elicited increases in dopamine transporter mRNA levels in the midbrain in the presence but not in the absence of METH pretreatment. Moreover, toxic METH doses caused decreases in the expression of the dopamine developmental factors, Shh, Lmx1b, and Nurr1, but not in the levels of Otx2 and Pitx3, in saline-pretreated rats. METH pretreatment followed by METH challenges also decreased Nurr1 but increased Otx2 and Pitx3 expression in the midbrain. These findings suggest that, in adult animals, toxic doses of METH can differentially influence the expression of transcription factors involved in the developmental regulation of dopamine neurons. The combined increases in Otx2 and Pitx3 expression after METH preconditioning might represent, in part, some of the mechanisms that served to protect against METH-induced striatal dopamine depletion observed after METH preconditioning.
Collapse
Affiliation(s)
- Irina N. Krasnova
- Molecular Neuropsychiatry Research Branch, DHHS/NIH/NIDA Intramural Research Program, Bethesda, Maryland, United States of America
| | - Bruce Ladenheim
- Molecular Neuropsychiatry Research Branch, DHHS/NIH/NIDA Intramural Research Program, Bethesda, Maryland, United States of America
| | - Amber B. Hodges
- Molecular Neuropsychiatry Research Branch, DHHS/NIH/NIDA Intramural Research Program, Bethesda, Maryland, United States of America
- Department of Psychology, Morgan State University, Baltimore, Maryland, United States of America
| | - Nora D. Volkow
- National Institute on Drug Abuse (NIDA), National Institutes of Health (NIH), U.S. Department of Health and Human Services (DHHS), Bethesda, Maryland, United States of America
| | - Jean Lud Cadet
- Molecular Neuropsychiatry Research Branch, DHHS/NIH/NIDA Intramural Research Program, Bethesda, Maryland, United States of America
- * E-mail:
| |
Collapse
|
24
|
Abstract
INTRODUCTION Metamfetamine is a highly addictive amfetamine analog that acts primarily as a central nervous system (CNS) stimulant. The escalating abuse of this drug in recent years has lead to an increasing burden upon health care providers. An understanding of the drug's toxic effects and their medical treatment is therefore essential for the successful management of patients suffering this form of intoxication. AIM The aim of this review is to summarize all main aspects of metamfetamine poisoning including epidemiology, mechanisms of toxicity, toxicokinetics, clinical features, diagnosis, and management. METHODS A summary of the literature on metamfetamine was compiled by systematically searching OVID MEDLINE and ISI Web of Science. Further information was obtained from book chapters, relevant news reports, and web material. Epidemiology. Following its use in the Second World War, metamfetamine gained popularity as an illicit drug in Japan and later the United States. Its manufacture and use has now spread to include East and South-East Asia, North America, Mexico, and Australasia, and its world-wide usage, when combined with amfetamine, exceeds that of all other drugs of abuse except cannabis. Mechanisms of toxicity. Metamfetamine acts principally by stimulating the enhanced release of catecholamines from sympathetic nerve terminals, particularly of dopamine in the mesolimbic, mesocortical, and nigrostriatal pathways. The consequent elevation of intra-synaptic monoamines results in an increased activation of central and peripheral α±- and β-adrenergic postsynaptic receptors. This can cause detrimental neuropsychological, cardiovascular, and other systemic effects, and, following long-term abuse, neuronal apoptosis and nerve terminal degeneration. Toxicokinetics. Metamfetamine is rapidly absorbed and well distributed throughout the body, with extensive distribution across high lipid content tissues such as the blood-brain barrier. In humans the major metabolic pathways are aromatic hydroxylation producing 4-hydroxymetamfetamine and N-demethylation to form amfetamine. Metamfetamine is excreted predominantly in the urine and to a lesser extent by sweating and fecal excretion, with reported terminal half-lives ranging from ∼5 to 30 h. Clinical features. The clinical effects of metamfetamine poisoning can vary widely, depending on dose, route, duration, and frequency of use. They are predominantly characteristic of an acute sympathomimetic toxidrome. Common features reported include tachycardia, hypertension, chest pain, various cardiac dysrhythmias, vasculitis, headache, cerebral hemorrhage, hyperthermia, tachypnea, and violent and aggressive behaviour. Management. Emergency stabilization of vital functions and supportive care is essential. Benzodiazepines alone may adequately relieve agitation, hypertension, tachycardia, psychosis, and seizure, though other specific therapies can also be required for sympathomimetic effects and their associated complications. CONCLUSION Metamfetamine may cause severe sympathomimetic effects in the intoxicated patient. However, with appropriate, symptom-directed supportive care, patients can be expected to make a full recovery.
Collapse
Affiliation(s)
- Leo J Schep
- National Poisons Centre, Department of Preventive and Social Medicine, University of Otago, Dunedin, New Zealand.
| | | | | |
Collapse
|
25
|
Cadet JL, Brannock C, Ladenheim B, McCoy MT, Beauvais G, Hodges AB, Lehrmann E, Wood WH, Becker KG, Krasnova IN. Methamphetamine preconditioning causes differential changes in striatal transcriptional responses to large doses of the drug. Dose Response 2010; 9:165-81. [PMID: 21731535 DOI: 10.2203/dose-response.10-011.cadet] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2022] Open
Abstract
Methamphetamine (METH) is a toxic drug of abuse, which can cause significant decreases in the levels of monoamines in various brain regions. However, animals treated with progressively increasing doses of METH over several weeks are protected against the toxic effects of the drug. In the present study, we tested the possibility that this pattern of METH injections might be associated with transcriptional changes in the rat striatum, an area of the brain which is known to be very sensitive to METH toxicity and which is protected by METH preconditioning. We found that the presence and absence of preconditioning followed by injection of large doses of METH caused differential expression in different sets of striatal genes. Quantitative PCR confirmed METH-induced changes in some genes of interest. These include small heat shock 27 kD proteins 1 and 2 (HspB1 and HspB2), brain derived neurotrophic factor (BDNF), and heme oxygenase-1 (Hmox-1). Our observations are consistent with previous studies which have reported that ischemic or pharmacological preconditioning can cause reprogramming of gene expression after lethal ischemic insults. These studies add to the growing literature on the effects of preconditioning on the brain transcriptome.
Collapse
Affiliation(s)
- Jean Lud Cadet
- Molecular Neuropsychiatry Research Branch, Intramural Research Program, NIDA/NIH/DHHS, Baltimore, MD, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
26
|
Tipton D, Legan Z, Dabbous M. Methamphetamine cytotoxicity and effect on LPS-stimulated IL-1β production by human monocytes. Toxicol In Vitro 2010; 24:921-7. [DOI: 10.1016/j.tiv.2009.11.015] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2009] [Revised: 11/20/2009] [Accepted: 11/23/2009] [Indexed: 12/19/2022]
|
27
|
Krasnova IN, Justinova Z, Ladenheim B, Jayanthi S, McCoy MT, Barnes C, Warner JE, Goldberg SR, Cadet JL. Methamphetamine self-administration is associated with persistent biochemical alterations in striatal and cortical dopaminergic terminals in the rat. PLoS One 2010; 5:e8790. [PMID: 20098750 PMCID: PMC2808335 DOI: 10.1371/journal.pone.0008790] [Citation(s) in RCA: 102] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2009] [Accepted: 12/28/2009] [Indexed: 11/20/2022] Open
Abstract
Methamphetamine (meth) is an illicit psychostimulant that is abused throughout the world. Repeated passive injections of the drug given in a single day or over a few days cause significant and long-term depletion of dopamine and serotonin in the mammalian brain. Because meth self-administration may better mimic some aspects of human drug-taking behaviors, we examined to what extent this pattern of drug treatment might also result in damage to monoaminergic systems in the brain. Rats were allowed to intravenously self-administer meth (yoked control rats received vehicle) 15 hours per day for 8 days before being euthanized at either 24 hours or at 7 and 14 days after cessation of drug taking. Meth self-administration by the rats was associated with a progressive escalation of daily drug intake to 14 mg/kg per day. Animals that self-administered meth exhibited dose-dependent decreases in striatal dopamine levels during the period of observation. In addition, there were significant reductions in the levels of striatal dopamine transporter and tyrosine hydroxylase proteins. There were also significant decreases in the levels of dopamine, dopamine transporter, and tyrosine hydroxylase in the cortex. In contrast, meth self-administration caused only transient decreases in norepinephrine and serotonin levels in the two brain regions, with these values returning to normal at seven days after cessation of drug taking. Importantly, meth self-administration was associated with significant dose-dependent increases in glial fibrillary acidic protein in both striatum and cortex, with these changes being of greater magnitude in the striatum. These results suggest that meth self-administration by rats is associated with long-term biochemical changes that are reminiscent of those observed in post-mortem brain tissues of chronic meth abusers.
Collapse
Affiliation(s)
- Irina N. Krasnova
- National Institute on Drug Abuse, National Institutes of Health, Baltimore, Maryland, United States of America
| | - Zuzana Justinova
- National Institute on Drug Abuse, National Institutes of Health, Baltimore, Maryland, United States of America
- Department of Psychiatry, University of Maryland School of Medicine, Baltimore, Maryland, United States of America
| | - Bruce Ladenheim
- National Institute on Drug Abuse, National Institutes of Health, Baltimore, Maryland, United States of America
| | - Subramaniam Jayanthi
- National Institute on Drug Abuse, National Institutes of Health, Baltimore, Maryland, United States of America
| | - Michael T. McCoy
- National Institute on Drug Abuse, National Institutes of Health, Baltimore, Maryland, United States of America
| | - Chanel Barnes
- National Institute on Drug Abuse, National Institutes of Health, Baltimore, Maryland, United States of America
| | - John E. Warner
- National Institute on Drug Abuse, National Institutes of Health, Baltimore, Maryland, United States of America
| | - Steven R. Goldberg
- National Institute on Drug Abuse, National Institutes of Health, Baltimore, Maryland, United States of America
| | - Jean Lud Cadet
- National Institute on Drug Abuse, National Institutes of Health, Baltimore, Maryland, United States of America
- * E-mail:
| |
Collapse
|
28
|
Boikess SR, O'Dell SJ, Marshall JF. A sensitizing D-amphetamine dose regimen induces long-lasting spinophilin and VGLUT1 protein upregulation in the rat diencephalon. Neurosci Lett 2009; 469:49-54. [PMID: 19932152 DOI: 10.1016/j.neulet.2009.11.042] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2009] [Revised: 11/12/2009] [Accepted: 11/16/2009] [Indexed: 11/29/2022]
Abstract
Numerous studies in this lab and others have reported psychostimulant-induced alterations in both synaptic protein expression and synaptic density in striatum and prefrontal cortex. Recently we have shown that chronic D-amphetamine (D-AMPH) administration in rats increased synaptic protein expression in striatum and limbic brain regions including hippocampus, amygdala, septum, and paraventricular nucleus of the thalamus (PVT). Potential synaptic changes in thalamic nuclei are interesting since the thalamus serves as a gateway to cerebral cortex and a nodal point for basal ganglia influences. Therefore we sought to examine drug-induced differences in synaptic protein expression throughout the diencephalon. Rats received an escalating (1-8 mg/kg) dosing regimen of D-AMPH for five weeks and were euthanized 28 days later. Radioimmunocytochemistry (RICC) revealed significant upregulation of both spinophilin and the vesicular glutamate transporter, VGLUT1, in PVT, mediodorsal (MD), and ventromedial (VM) thalamic nuclei as well as in lateral hypothalamus (LH) and habenula. Strong positive correlations were observed between VGLUT1 and spinophilin expression in PVT, medial habenula, MD, VM and LH of D-AMPH-treated rats. No significant D-AMPH effect was seen in sensorimotor cortices for either protein. Additionally, no significant differences in the general vesicular protein synaptophysin were observed for any brain region. These findings add to evidence suggesting that long-lasting stimulant-induced synaptic alterations are widespread but not ubiquitous. Moreover, they suggest that D-AMPH-induced synaptic changes may occur preferentially in excitatory synapses.
Collapse
Affiliation(s)
- Steven R Boikess
- Department of Neurobiology and Behavior, University of California, Irvine, Irvine, CA 92697, USA
| | | | | |
Collapse
|
29
|
Cadet JL, McCoy MT, Cai NS, Krasnova IN, Ladenheim B, Beauvais G, Wilson N, Wood W, Becker KG, Hodges AB. Methamphetamine preconditioning alters midbrain transcriptional responses to methamphetamine-induced injury in the rat striatum. PLoS One 2009; 4:e7812. [PMID: 19915665 PMCID: PMC2771908 DOI: 10.1371/journal.pone.0007812] [Citation(s) in RCA: 47] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2009] [Accepted: 10/15/2009] [Indexed: 01/19/2023] Open
Abstract
Methamphetamine (METH) is an illicit drug which is neurotoxic to the mammalian brain. Numerous studies have revealed significant decreases in dopamine and serotonin levels in the brains of animals exposed to moderate-to-large METH doses given within short intervals of time. In contrast, repeated injections of small nontoxic doses of the drug followed by a challenge with toxic METH doses afford significant protection against monoamine depletion. The present study was undertaken to test the possibility that repeated injections of the drug might be accompanied by transcriptional changes involved in rendering the nigrostriatal dopaminergic system refractory to METH toxicity. Our results confirm that METH preconditioning can provide significant protection against METH-induced striatal dopamine depletion. In addition, the presence and absence of METH preconditioning were associated with substantial differences in the identity of the genes whose expression was affected by a toxic METH challenge. Quantitative PCR confirmed METH-induced changes in genes of interest and identified additional genes that were differentially impacted by the toxic METH challenge in the presence of METH preconditioning. These genes include small heat shock 27 kD 27 protein 2 (HspB2), thyrotropin-releasing hormone (TRH), brain derived neurotrophic factor (BDNF), c-fos, and some encoding antioxidant proteins including CuZn superoxide dismutase (CuZnSOD), glutathione peroxidase (GPx)-1, and heme oxygenase-1 (Hmox-1). These observations are consistent, in part, with the transcriptional alterations reported in models of lethal ischemic injuries which are preceded by ischemic or pharmacological preconditioning. Our findings suggest that multiple molecular pathways might work in tandem to protect the nigrostriatal dopaminergic pathway against the deleterious effects of the toxic psychostimulant. Further analysis of the molecular and cellular pathways regulated by these genes should help to provide some insight into the neuroadaptive potentials of the brain when repeatedly exposed to drugs of abuse.
Collapse
Affiliation(s)
- Jean Lud Cadet
- Molecular Neuropsychiatry Research Branch, DHHS/NIH/NIDA Intramural Research Program, Baltimore, MD, USA.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
30
|
Xi ZX, Kleitz HK, Deng X, Ladenheim B, Peng XQ, Li X, Gardner EL, Stein EA, Cadet JL. A single high dose of methamphetamine increases cocaine self-administration by depletion of striatal dopamine in rats. Neuroscience 2009; 161:392-402. [PMID: 19336247 PMCID: PMC3737775 DOI: 10.1016/j.neuroscience.2009.03.060] [Citation(s) in RCA: 30] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2009] [Revised: 03/20/2009] [Accepted: 03/23/2009] [Indexed: 11/24/2022]
Abstract
Psychostimulant addicts often take high doses of drugs, and high doses of psychostimulants such as methamphetamine (METH) are neurotoxic to striatal dopamine (DA) terminals. Yet, the effects of high doses of METH on drug-seeking and drug-taking behavior have not been examined. In the present study, we found that single high doses of METH in rats (10-20 mg/kg) dose-dependently increased cocaine self-administration under fixed-ratio 2 (FR2) reinforcement conditions, while higher doses (40 mg/kgx1 or 10 mg/kg/2 hx4) caused high mortality among rats maintained on daily cocaine self-administration. The increased cocaine self-administration appeared to be a compensatory response to reduced cocaine reward after METH, because the same doses of METH caused a dose-dependent reduction both in "break-point" levels for cocaine self-administration under progressive-ratio reinforcement and in nucleus accumbens DA response to acute cocaine. Further, METH (10-20 mg/kg) produced large DA release (4000%-6000% over baseline), followed by a significant reduction in striatal DA and 3,4-dihydroxyphenylacetic acid (DOPAC) contents, but without significant changes in striatal DA transporter levels. These findings suggest that the present high doses of METH caused striatal DA depletion or hypofunction without severe damage in DA terminals, which may contribute to the increased cocaine-taking behavior observed in the present study. Provided that the present doses of METH may mimic METH overdose incidents in humans, the present findings suggest that METH-induced DA depletion or neurotoxicity may lead to an increase in subsequent drug-taking and drug-seeking behavior.
Collapse
Affiliation(s)
- Z-X Xi
- Chemical Biology Research Branch, Intramural Research Program, National Institute on Drug Abuse, 251 Bayview Boulevard, BRC Room 05A705, Baltimore, MD 21224, USA.
| | | | | | | | | | | | | | | | | |
Collapse
|
31
|
Jayanthi S, McCoy MT, Beauvais G, Ladenheim B, Gilmore K, Wood W, Becker K, Cadet JL. Methamphetamine induces dopamine D1 receptor-dependent endoplasmic reticulum stress-related molecular events in the rat striatum. PLoS One 2009; 4:e6092. [PMID: 19564919 PMCID: PMC2699544 DOI: 10.1371/journal.pone.0006092] [Citation(s) in RCA: 67] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2009] [Accepted: 05/27/2009] [Indexed: 12/25/2022] Open
Abstract
Methamphetamine (METH) is an illicit toxic psychostimulant which is widely abused. Its toxic effects depend on the release of excessive levels of dopamine (DA) that activates striatal DA receptors. Inhibition of DA-mediated neurotransmission by the DA D1 receptor antagonist, SCH23390, protects against METH-induced neuronal apoptosis. The initial purpose of the present study was to investigate, using microarray analyses, the influence of SCH23390 on transcriptional responses in the rat striatum caused by a single METH injection at 2 and 4 hours after drug administration. We identified 545 out of a total of 22,227 genes as METH-responsive. These include genes which are involved in apoptotic pathways, endoplasmic reticulum (ER) stress, and in transcription regulation, among others. Of these, a total of 172 genes showed SCH23390-induced inhibition of METH-mediated changes. Among these SCH23390-responsive genes were several genes that are regulated during ER stress, namely ATF3, HSP27, Hmox1, HSP40, and CHOP/Gadd153. The secondary goal of the study was to investigate the role of DA D1 receptor stimulation on the expression of genes that participate in ER stress-mediated molecular events. We thus used quantitative PCR to confirm changes in the METH-responsive ER genes identified by the microarray analyses. We also measured the expression of these genes and of ATF4, ATF6, BiP/GRP78, and of GADD34 over a more extended time course. SCH23390 attenuated or blocked METH-induced increases in the expression of the majority of these genes. Western blot analysis revealed METH-induced increases in the expression of the antioxidant protein, Hmox1, which lasted for about 24 hours after the METH injection. Additionally, METH caused DA D1 receptor-dependent transit of the Hmox1 regulator protein, Nrf2, from cytosolic into nuclear fractions where the protein exerts its regulatory functions. When taken together, these findings indicate that SCH23390 can provide protection against neuronal apoptosis by inhibiting METH-mediated DA D1 receptor-mediated ER stress in the rat striatum. Our data also suggest that METH-induced toxicity might be a useful model to dissect molecular mechanisms involved in ER stress-dependent events in the rodent brain.
Collapse
Affiliation(s)
- Subramaniam Jayanthi
- Molecular Neuropsychiatry Research Branch, National Institute of Drug Abuse, National Institutes of Health (NIH)/Department of Health and Human Services (DHHS), Intramural Research Program, Baltimore, Maryland, United States of America
| | - Michael T. McCoy
- Molecular Neuropsychiatry Research Branch, National Institute of Drug Abuse, National Institutes of Health (NIH)/Department of Health and Human Services (DHHS), Intramural Research Program, Baltimore, Maryland, United States of America
| | - Genevieve Beauvais
- Molecular Neuropsychiatry Research Branch, National Institute of Drug Abuse, National Institutes of Health (NIH)/Department of Health and Human Services (DHHS), Intramural Research Program, Baltimore, Maryland, United States of America
| | - Bruce Ladenheim
- Molecular Neuropsychiatry Research Branch, National Institute of Drug Abuse, National Institutes of Health (NIH)/Department of Health and Human Services (DHHS), Intramural Research Program, Baltimore, Maryland, United States of America
| | - Kristi Gilmore
- Molecular Neuropsychiatry Research Branch, National Institute of Drug Abuse, National Institutes of Health (NIH)/Department of Health and Human Services (DHHS), Intramural Research Program, Baltimore, Maryland, United States of America
| | - William Wood
- Gene Expression and Genomics Unit, National Institute of Aging, National Institutes of Health (NIH)/Department of Health and Human Services (DHHS), Intramural Research Program, Baltimore, Maryland, United States of America
| | - Kevin Becker
- Gene Expression and Genomics Unit, National Institute of Aging, National Institutes of Health (NIH)/Department of Health and Human Services (DHHS), Intramural Research Program, Baltimore, Maryland, United States of America
| | - Jean Lud Cadet
- Molecular Neuropsychiatry Research Branch, National Institute of Drug Abuse, National Institutes of Health (NIH)/Department of Health and Human Services (DHHS), Intramural Research Program, Baltimore, Maryland, United States of America
- * E-mail:
| |
Collapse
|
32
|
Kitanaka J, Kitanaka N, Tatsuta T, Hall FS, Uhl GR, Tanaka K, Nishiyama N, Morita Y, Takemura M. Sigma1 receptor antagonists determine the behavioral pattern of the methamphetamine-induced stereotypy in mice. Psychopharmacology (Berl) 2009; 203:781-92. [PMID: 19052726 PMCID: PMC3157915 DOI: 10.1007/s00213-008-1425-z] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/22/2008] [Accepted: 11/20/2008] [Indexed: 10/21/2022]
Abstract
OBJECTIVE The effects of sigma receptor antagonists on methamphetamine (METH)-induced stereotypy have not been examined. We examined the effects of sigma antagonists on METH-induced stereotypy in mice. RESULTS The administration of METH (10 mg/kg) to male ddY mice induced stereotyped behavior consisting of biting (90.1%), sniffing (4.2%), head bobbing (4.1%), and circling (1.7%) during an observation period of 1 h. Pretreatment of the mice with BMY 14802 (alpha-(4-fluorophenyl)-4-(5-fluoro-2-pyrimidinyl)-1-piperazinebutanol; 1, 5, and 10 mg/kg), a non-specific sigma receptor antagonist, significantly increased METH-induced sniffing (19.2%, 30.5%, and 43.8% of total stereotypical behavior) but decreased biting (76.6%, 66.9%, and 49.3% of total stereotypical behavior) in a dose-dependent manner. This response was completely abolished by (+)-SKF 10,047 ([2S-(2alpha,6alpha,11R)]-1,2,3,4,5,6-hexahydro-6,11-dimethyl-3-(2-propenyl)-2,6-methano-3-benzazocin-8-ol; 4 and 10 mg/kg), a putative sigma(1) receptor agonist, and partially by PB 28 (1-cyclohexyl-4-[3-(1,2,3,4-tetrahydro-5-methoxy-1-naphthalen-1-yl)-n-propyl]piperazine; 1 and 10 mg/kg), a putative sigma(2) receptor agonist. The BMY 14802 action on METH-induced stereotypy was mimicked by BD 1047 (N-[2-(3,4-dichlorophenyl)ethyl]-N-methyl-2-(dimethylamino)ethylamine; 10 mg/kg), a putative sigma(1) receptor antagonist, but not by SM-21 ((+/-)-tropanyl 2-(4-chlorophenoxy)butanoate; 1 mg/kg), a putative sigma(2) receptor antagonist. The BD 1047 effect on METH-induced stereotypy was also abolished completely by (+)-SKF 10,047 and partially by PB 28. The overall frequency of METH-induced stereotypical behavior was unchanged with these sigma receptor ligands, despite the alteration in particular behavioral patterns. The BMY 14802 action on METH-induced stereotypy was unaffected by pretreatment with centrally acting histamine H(1) receptor antagonists (pyrilamine or ketotifen, 10 mg/kg), suggesting that these effects are independent of histamine H(1) receptor signaling systems. CONCLUSION In summary, modulation of central sigma(1) receptors alters the pattern of METH-induced stereotypy, producing a shift from stereotypical biting to stereotypical sniffing, without affecting the overall frequency of stereotypical behavior.
Collapse
Affiliation(s)
- J Kitanaka
- Department of Pharmacology, Hyogo College of Medicine, 1-1 Mukogawa-cho, Nishinomiya, Hyogo, 663-8501, Japan.
| | | | | | | | | | | | | | | | | |
Collapse
|
33
|
Cadet JL, Krasnova IN, Ladenheim B, Cai NS, McCoy MT, Atianjoh FE. Methamphetamine preconditioning: differential protective effects on monoaminergic systems in the rat brain. Neurotox Res 2009; 15:252-9. [PMID: 19384598 PMCID: PMC2673017 DOI: 10.1007/s12640-009-9026-0] [Citation(s) in RCA: 31] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2008] [Revised: 09/19/2008] [Accepted: 09/19/2008] [Indexed: 01/06/2023]
Abstract
Pretreatment with methamphetamine (METH) can attenuate toxicity due to acute METH challenges. The majority of previous reports have focused mainly on the effects of the drug on the striatal dopaminergic system. In the present study, we used a regimen that involves gradual increases in METH administration to rats in order to mimic progressively larger doses of the drug used by some human METH addicts. We found that this METH preconditioning was associated with complete protection against dopamine depletion caused by a METH challenge (5 mg/kg x 6 injections given 1 h apart) in the striatum and cortex. In contrast, there was no preconditioning-mediated protection against METH-induced serotonin depletion in the striatum and hippocampus, with some protection being observed in the cortex. There was also no protection against METH-induced norepinephrine (NE) depletion in the hippocampus. These results indicate that, in contrast to the present dogmas, there might be differences in the mechanisms involved in METH toxicity on monoaminergic systems in the rodent brain. Thus, chronic injections of METH might activate programs that protect against dopamine toxicity without influencing drug-induced pathological changes in serotoninergic systems. Further studies will need to evaluate the cellular and molecular bases for these differential responses.
Collapse
Affiliation(s)
- Jean Lud Cadet
- Molecular Neuropsychiatry Branch, National Institute on Drug Abuse/NIH/DHHS, 251 Bayview Boulevard, Baltimore, MD 21224, USA.
| | | | | | | | | | | |
Collapse
|
34
|
Smith DE. Physical vs. Psychological Dependence and Tolerance in High-Dose Methamphetamine Abuse. Clin Toxicol (Phila) 2008. [DOI: 10.3109/15563656908990918] [Citation(s) in RCA: 16] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
|
35
|
Graham DL, Noailles PAH, Cadet JL. Differential neurochemical consequences of an escalating dose-binge regimen followed by single-day multiple-dose methamphetamine challenges. J Neurochem 2008; 105:1873-85. [PMID: 18248616 DOI: 10.1111/j.1471-4159.2008.05269.x] [Citation(s) in RCA: 41] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
Chronic intake of methamphetamine (METH) causes tolerance to its behavioral and subjective effects. To better mimic human patterns of drug abuse, the present study used a rodent model that took into account various facets of human drug administration and measured METH-induced effects on brain monoamine levels. Adult male Sprague-Dawley rats were injected with METH or saline according to an escalating dose schedule for 2 weeks. This was followed by a challenge regimen of either saline or one of two doses of METH (3 x 10 mg/kg every 2 h or 6 x 5 mg/kg given every hour, both given within a single day). Both challenge doses of METH caused significant degrees of depletion of dopamine in the striatum and norepinephrine and serotonin in the striatum, cortex, and hippocampus. Animals pre-treated with METH showed significant attenuation of METH-induced striatal dopamine depletion but not consistent attenuation of norepinephrine and serotonin depletion. Unexpectedly, METH pre-treated animals that received the 3 x 10 mg/kg challenge showed less increases in tympanic temperatures than saline pre-treated rats whereas METH pre-treated animals that received the 6 x 5 mg/kg METH challenge showed comparable increases in temperatures to saline pre-treated rats. Therefore, pre-treatment-induced partial protection against monoamine depletion is probably not because of attenuated METH-induced hyperthermia in those rats.
Collapse
Affiliation(s)
- Devon L Graham
- Molecular Neuropsychiatry Branch, DHHS/NIH/NIDA Intramural Research Program, Baltimore, Maryland 21224, USA
| | | | | |
Collapse
|
36
|
Liu Y, Morgan D, Roberts DCS. Cross-sensitization of the reinforcing effects of cocaine and amphetamine in rats. Psychopharmacology (Berl) 2007; 195:369-75. [PMID: 17710385 DOI: 10.1007/s00213-007-0909-6] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/09/2007] [Accepted: 07/20/2007] [Indexed: 11/28/2022]
Abstract
RATIONALE Cross-sensitization between cocaine and amphetamine has been demonstrated in different behavioral paradigms. There is a relative paucity of studies examining whether cross-sensitization occurs between amphetamine and cocaine when both are self-administered. OBJECTIVE The current study was designed to test whether animals sensitized to the reinforcing effects of cocaine would show cross-sensitization of the reinforcing effects of amphetamine, using a self-administration paradigm. MATERIALS AND METHODS Male, Sprague-Dawley rats were trained to self-administer cocaine and given limited or high exposure to cocaine under a fixed ratio (FR) 1 procedure. After the initial exposure to cocaine, animals self-administered cocaine (1.5 mg/kg per injection) under a progressive ratio (PR) procedure. Subsequently, breakpoints on a PR schedule and rates of intake on an FR schedule maintained by different doses of amphetamine were assessed. RESULTS Animals with high initial exposure to cocaine (40 injections of 1.5 mg/kg per injection per day for 5 days) showed stable breakpoints throughout testing. Animals given limited initial cocaine exposure (20 injections of 0.75 mg/kg per injection for 1 day) produced a gradual increase in breakpoints maintained by cocaine over time (i.e., sensitization of the reinforcing effects of cocaine). When subsequently tested with amphetamine, the dose-effect curve was shifted upward in the limited-exposure group relative to the high-exposure group, suggesting cross-sensitization of the reinforcing effects of amphetamine. CONCLUSIONS Sensitization of the reinforcing effects of cocaine resulted in cross-sensitization of the reinforcing effects of amphetamine. This phenomenon occurs even when both drugs are self-administered.
Collapse
Affiliation(s)
- Yu Liu
- Department of Physiology and Pharmacology, Wake Forest University Health Sciences, Medical Center Blvd., Winston-Salem, NC 27157, USA
| | | | | |
Collapse
|
37
|
Abstract
Interest has recently been rekindled in receptors that are activated by low molecular weight, noncatecholic, biogenic amines that are typically found as trace constituents of various vertebrate and invertebrate tissues and fluids. The timing of this resurgent focus on receptors activated by the "trace amines" (TA) beta-phenylethylamine (PEA), tyramine (TYR), octopamine (OCT), synephrine (SYN), and tryptamine (TRYP) is the direct result of 2 publications that appeared in 2001 describing the cloning of a novel G protein-coupled receptor (GPCR) referred to by their discoverers Borowsky et al. as TA1 and Bunzow et al. as TA receptor 1 (TAR1). When heterologously expressed in Xenopus laevis oocytes and various eukaryotic cell lines, recombinant rodent and human TAR dose-dependently couple to the stimulation of adenosine 3',5'-monophosphate (cAMP) production. Structure-activity profiling based on this functional response has revealed that in addition to the TA, other biologically active compounds containing a 2-carbon aliphatic side chain linking an amino group to at least 1 benzene ring are potent and efficacious TA receptor agonists with amphetamine (AMPH), methamphetamine, 3-iodothyronamine, thyronamine, and dopamine (DA) among the most notable. Almost 100 years after the search for TAR began, numerous TA1/TAR1-related sequences, now called TA-associated receptors (TAAR), have been identified in the genome of every species of vertebrate examined to date. Consequently, even though heterologously expressed TAAR1 fits the pharmacological criteria established for a bona fide TAR, a major challenge for those working in the field is to discern the in vivo pharmacology and physiology of each purported member of this extended family of GPCR. Only then will it be possible to establish whether TAAR1 is the family archetype or an iconoclast.
Collapse
Affiliation(s)
- David K Grandy
- Department of Physiology and Pharmacology, L334, School of Medicine, Oregon Health and Science University, Portland, OR 97239, United States.
| |
Collapse
|
38
|
Pum M, Carey RJ, Huston JP, Müller CP. Dissociating effects of cocaine and d-amphetamine on dopamine and serotonin in the perirhinal, entorhinal, and prefrontal cortex of freely moving rats. Psychopharmacology (Berl) 2007; 193:375-90. [PMID: 17468969 DOI: 10.1007/s00213-007-0791-2] [Citation(s) in RCA: 68] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/03/2006] [Accepted: 04/03/2007] [Indexed: 02/06/2023]
Abstract
RATIONALE Neuroimaging studies with humans showed widespread activation of the cortex in response to psychostimulant drugs. However, the neurochemical nature of these brain activities is not characterized. OBJECTIVE The aim of the present study was to investigate the effects of cocaine and d-amphetamine on dopamine (DA) and serotonin (5-HT) in cortical areas of the hippocampal network in comparison to the prefrontal cortex (PFC). MATERIALS AND METHODS We conducted in vivo microdialysis experiments in behaving rats measuring DA and 5-HT in the perirhinal cortex (PRC), entorhinal cortex (EC), and PFC, after application of cocaine (0, 5, 10, 20 mg/kg; i.p.) or d-amphetamine (0, 0.5, 1.0, 2.5 mg/kg; i.p.). RESULTS Cocaine and d-amphetamine dose-dependently increased DA and 5-HT levels in the PRC, EC, and PFC. A predominant DA response to d-amphetamine was only found in the PFC, but not in the PRC and EC. Cocaine increased DA and 5-HT to an equal extent in the PFC and PRC but induced a predominant 5-HT response in the EC. When comparing the neurochemical responses between the drugs at an equal level of behavioral activation, cocaine was more potent than d-amphetamine in increasing 5-HT in the PFC, while no differences were found in the PRC or EC or in the DA responses in all three cortical areas. CONCLUSIONS We conclude that cocaine and d-amphetamine increase DA and 5-HT levels in PRC and EC largely to the same extent as in the PFC.
Collapse
Affiliation(s)
- M Pum
- Institute of Physiological Psychology and Center for Biological and Medical Research, University of Düsseldorf, Düsseldorf, Germany
| | | | | | | |
Collapse
|
39
|
Kitanaka J, Kitanaka N, Takemura M. Neurochemical consequences of dysphoric state during amphetamine withdrawal in animal models: a review. Neurochem Res 2007; 33:204-19. [PMID: 17605106 DOI: 10.1007/s11064-007-9409-7] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2007] [Accepted: 06/07/2007] [Indexed: 10/23/2022]
Abstract
Chronic abuse of amphetamines, such as d-amphetamine (AMPH) and d-methamphetamine, results in psychological dependence, a condition in which the drug produces a feeling of satisfaction and a drive that requires periodic or continuous administration of the drug to produce overwhelming pleasure or to avoid discomfort such as dysphoria. The dysphoric state of AMPH withdrawal has been recognized as depressive syndromes, such as anhedonia, depression, anxiety, and social inhibition, in early drug abstinence. Medication for treatment of the dysphoric state is important for AMPH abusers to avoid impulsive self-injurious behavior or acts that are committed with unconscious or uncontrolled suicidal ideation. However, successful treatments for AMPH withdrawal remain elusive, since the exact molecular basis of the expression of dysphoria has not been fully elucidated. This review focuses on the molecular aspects of AMPH withdrawal as indexed by neurochemical parameters under a variety of injection regimens (for example, levels of brain monoamines and their metabolites, and gamma-aminobutyric acid, expression of genes and proteins involved in neuronal activity, and monoamine metabolism and availability) in rodent models which exhibit significant phenotypic features relevant to the syndromes of AMPH withdrawal in humans.
Collapse
Affiliation(s)
- Junichi Kitanaka
- Department of Pharmacology, Hyogo College of Medicine, 1-1 Mukogawa-cho, Nishinomiya, Hyogo 663-8501, Japan.
| | | | | |
Collapse
|
40
|
Inciardi JA, Surratt HL, Kurtz SP, Cicero TJ. Mechanisms of prescription drug diversion among drug-involved club- and street-based populations. PAIN MEDICINE (MALDEN, MASS.) 2007; 8:171-83. [PMID: 17305688 PMCID: PMC2879025 DOI: 10.1111/j.1526-4637.2006.00255.x] [Citation(s) in RCA: 222] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
OBJECTIVE Prescription drug diversion involves the unlawful channeling of regulated pharmaceuticals from legal sources to the illicit marketplace, and can occur along all points in the drug delivery process, from the original manufacturing site to the wholesale distributor, the physician's office, the retail pharmacy, or the patient. However, empirical data on diversion are limited. METHOD In an attempt to develop a better understanding of how specific drug-using populations are diverting prescription opioids and other medications, or obtaining controlled drugs that have already been diverted, qualitative interviews and focus group data were collected on four separate populations of prescription drug abusers in Miami, Florida--club drug users, street-based illicit drug users, methadone maintenance patients, and HIV positive individuals who abuse and/or divert drugs. RESULTS Sources of abused prescription drugs cited by focus group participants were extremely diverse, including their physicians and pharmacists; parents and relatives; "doctor shopping"; leftover supplies following an illness or injury; personal visits to Mexico, South America and the Caribbean; prescriptions intended for the treatment of mental illness; direct sales on the street and in nightclubs; pharmacy and hospital theft; through friends or acquaintances; under-the-door apartment flyers advertising telephone numbers to call; and "stealing from grandma's medicine cabinet." CONCLUSION While doctor shoppers, physicians and the Internet receive much of the attention regarding diversion, the data reported in this paper suggest that there are numerous active street markets involving patients, Medicaid recipients and pharmacies as well. In addition, there are other data which suggest that the contributions of residential burglaries, pharmacy robberies and thefts, and "sneak thefts" to the diversion problem may be understated.
Collapse
Affiliation(s)
- James A Inciardi
- Center for Drug and Alcohol Studies, University of Delaware, Coral Gables, Florida 33134, USA.
| | | | | | | |
Collapse
|
41
|
Müller CP, Carey RJ, Huston JP, De Souza Silva MA. Serotonin and psychostimulant addiction: Focus on 5-HT1A-receptors. Prog Neurobiol 2007; 81:133-78. [PMID: 17316955 DOI: 10.1016/j.pneurobio.2007.01.001] [Citation(s) in RCA: 245] [Impact Index Per Article: 13.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2006] [Revised: 12/04/2006] [Accepted: 01/03/2007] [Indexed: 01/03/2023]
Abstract
Serotonin(1A)-receptors (5-HT(1A)-Rs) are important components of the 5-HT system in the brain. As somatodendritic autoreceptors they control the activity of 5-HT neurons, and, as postsynaptic receptors, the activity in terminal areas. Cocaine (COC), amphetamine (AMPH), methamphetamine (METH) and 3,4-methylenedioxymethamphetamine ("Ecstasy", MDMA) are psychostimulant drugs that can lead to addiction-related behavior in humans and in animals. At the neurochemical level, these psychostimulant drugs interact with monoamine transporters and increase extracellular 5-HT, dopamine and noradrenalin activity in the brain. The increase in 5-HT, which, in addition to dopamine, is a core mechanism of action for drug addiction, hyperactivates 5-HT(1A)-Rs. Here, we first review the role of the various 5-HT(1A)-R populations in spontaneous behavior to provide a background to elucidate the contribution of the 5-HT(1A)-Rs to the organization of psychostimulant-induced addiction behavior. The progress achieved in this field shows the fundamental contribution of brain 5-HT(1A)-Rs to virtually all behaviors associated with psychostimulant addiction. Importantly, the contribution of pre- and postsynaptic 5-HT(1A)-Rs can be dissociated and frequently act in opposite directions. We conclude that 5-HT(1A)-autoreceptors mainly facilitate psychostimulant addiction-related behaviors by a limitation of the 5-HT response in terminal areas. Postsynaptic 5-HT(1A)-Rs, in contrast, predominantly inhibit the expression of various addiction-related behaviors directly. In addition, they may also influence the local 5-HT response by feedback mechanisms. The reviewed findings do not only show a crucial role of 5-HT(1A)-Rs in the control of brain 5-HT activity and spontaneous behavior, but also their complex role in the regulation of the psychostimulant-induced 5-HT response and subsequent addiction-related behaviors.
Collapse
Affiliation(s)
- Christian P Müller
- Institute of Physiological Psychology I, University of Düsseldorf, Universitätsstr. 1, 40225 Düsseldorf, Germany.
| | | | | | | |
Collapse
|
42
|
Kitamura O, Wee S, Specio SE, Koob GF, Pulvirenti L. Escalation of methamphetamine self-administration in rats: a dose-effect function. Psychopharmacology (Berl) 2006; 186:48-53. [PMID: 16552556 DOI: 10.1007/s00213-006-0353-z] [Citation(s) in RCA: 168] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/07/2005] [Accepted: 02/10/2006] [Indexed: 10/24/2022]
Abstract
RATIONALE The transition from stable to escalated drug intake has been demonstrated in rats self-administrating cocaine and heroin using a single dose of drug. OBJECTIVES To investigate the prolonged exposure to methamphetamine self-administration and the effect of various training doses of methamphetamine on the changes of methamphetamine intake over a 21-day period. METHODS Two groups of rats were trained in 1-h daily sessions of methamphetamine self-administration [0.033 mg/infusion (inf); approximately 0.066 mg/kg/inf]. Methamphetamine access was increased to 6 h in one group [Long Access (LgA)] or maintained at 1 h in another [Short Access (ShA)]. The same procedure was repeated in rats exposed to different training doses of methamphetamine (0.05, 0.1, and 0.2 mg/kg/inf). RESULTS In LgA rats, total and first hour intake of methamphetamine significantly increased compared to ShA rats at various methamphetamine doses. LgA animals, at all doses in the second study, escalated intake to 8-9 mg/kg per 6-h session, with the most rapid escalation occurring at 3-5 days at a methamphetamine dose of 0.1 mg/kg/inf. CONCLUSIONS The escalation of drug intake observed with extended access is produced at multiple doses of methamphetamine. The rapidity of escalation depends on the dose. Ultimately, all doses in the dose-response study engendered self-administration of the same amount of total drug in a 6-h session in the extended-access group. Results suggest that the rapidity of escalation is dependent on dose and has an upper limit of intake over a period of 21 days.
Collapse
Affiliation(s)
- Osamu Kitamura
- Molecular and Integrative Neurosciences Department, SP30-2400, The Scripps Research Institute, 10550 North Torrey Pines Road, La Jolla, CA 92037, USA
| | | | | | | | | |
Collapse
|
43
|
Ahmed SH. Imbalance between drug and non-drug reward availability: A major risk factor for addiction. Eur J Pharmacol 2005; 526:9-20. [PMID: 16263108 DOI: 10.1016/j.ejphar.2005.09.036] [Citation(s) in RCA: 59] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2005] [Accepted: 09/23/2005] [Indexed: 11/18/2022]
Abstract
Laboratory animals self-administer most, though not all, drugs of abuse. Recent evidence shows that with increased drug availability, most laboratory rats develop all the major behavioral signs of addiction, including: 1) drug intake escalation, 2) increased motivation for the drug, 3) difficulty to abstain, 4) decreased reward function, and 5) inflexible drug use. The large prevalence of addicted rats may suggest that they are particularly vulnerable to develop compulsive drug use. I review evidence showing that this apparent vulnerability results in large part from the lack of positive (i.e., alternative non-drug rewards) and negative (i.e., costs) incentives capable of turning animals away from the pursuit of drugs. In particular, most animals seem to take drugs and eventually become addicted, not because drugs are intrinsically addictive, but more likely because drugs are the only significant sources of reward available in the laboratory. Laboratory animals would therefore represent more of a model of high-risk human groups than of the general population. Consequently, they should be more suited for searching factors that protect from, rather than predispose to, drug addiction. Reconsidering the environmental background of drug self-administration experiments in laboratory animals raises intriguing implications for understanding the initial demand for drug consumption and the transition to drug addiction, and for extrapolation from laboratory animals to humans.
Collapse
Affiliation(s)
- Serge H Ahmed
- Laboratoire de Neuropsychobiologie des Désadaptations, University Victor-Segalen Bordeaux2, CNRS-UMR 5541, 33076 Bordeaux, France.
| |
Collapse
|
44
|
Jayanthi S, Deng X, Noailles PAH, Ladenheim B, Cadet JL. Methamphetamine induces neuronal apoptosis via cross-talks between endoplasmic reticulum and mitochondria-dependent death cascades. FASEB J 2004; 18:238-51. [PMID: 14769818 DOI: 10.1096/fj.03-0295com] [Citation(s) in RCA: 222] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Methamphetamine (METH) is an illicit drug that causes neurodegenerative effects in humans. In rodents, METH induces apoptosis of striatal glutamic acid decarboxylase (GAD) -containing neurons. This paper provides evidence that METH-induced cell death occurs consequent to interactions of ER stress and mitochondrial death pathways. Specifically, injections of METH are followed by an almost immediate activation of proteases calpain and caspase-12, events consistent with drug-induced ER stress. Involvement of ER stress was further supported by observations of increases in the expression of GRP78/BiP and CHOP. Participation of the mitochondrial pathway was demonstrated by the transition of AIF, smac/DIABLO, and cytochrome c from mitochondrial into cytoplasmic fractions. These changes occur before the apoptosome-associated pro-caspase-9 cleavage. Effector caspases-3 and -6, but not -7, were cleaved with the initial time of caspase-3 activation occurring before caspase 9 cleavage; this suggests possible earlier cleavage of caspase-3 by caspase-12. These events preceded proteolysis of the caspase substrates DFF-45, lamin A, and PARP in nuclear fractions. These findings indicate that METH causes neuronal apoptosis in part via cross-talks between ER- and mitochondria-generated processes, which cause activation of both caspase-dependent and -independent pathways.
Collapse
Affiliation(s)
- Subramaniam Jayanthi
- Molecular Neuropsychiatry Branch, National Institute on Drug Abuse, Intramural Research Program, National Institute of Health, DHHS, 5500 Nathan Shock Dr., Baltimore, MD 21224, USA
| | | | | | | | | |
Collapse
|
45
|
Moszczynska A, Fitzmaurice P, Ang L, Kalasinsky KS, Schmunk GA, Peretti FJ, Aiken SS, Wickham DJ, Kish SJ. Why is parkinsonism not a feature of human methamphetamine users? Brain 2004; 127:363-70. [PMID: 14645148 DOI: 10.1093/brain/awh046] [Citation(s) in RCA: 107] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
For more than 50 years, methamphetamine has been a widely used stimulant drug taken to maintain wakefulness and performance and, in high doses, to cause intense euphoria. Animal studies show that methamphetamine can cause short-term and even persistent depletion of brain levels of the neurotransmitter dopamine. However, the clinical features of Parkinson's disease, a dopamine deficiency disorder of the brain, do not appear to be characteristic of human methamphetamine users. We compared dopamine levels in autopsied brain tissue of chronic methamphetamine users with those in patients with Parkinson's disease and in a control group. Mean dopamine levels in the methamphetamine users were reduced more in the caudate (-61%) than in the putamen (-50%), a pattern opposite to that of Parkinson's disease. Some methamphetamine users had severely decreased dopamine levels, within the parkinsonian range, in the caudate (up to 97% dopamine loss) but not in the putamen. As the putamen and caudate subserve aspects of motor and cognitive function, respectively, our data suggest that methamphetamine users are not parkinsonian because dopamine levels are not sufficiently decreased in the motor component of the striatum. However, the near-total reduction in the caudate could explain reports of cognitive disturbances, sometimes disabling, in some drug users, and suggests that treatment with dopamine substitution medication (e.g. levodopa) during drug rehabilitation might be helpful.
Collapse
Affiliation(s)
- Anna Moszczynska
- Human Neurochemical Pathology Laboratory, Centre for Addiction and Mental Health, 250 College Street, Toronto, Ontario M5T 1R8, Canada
| | | | | | | | | | | | | | | | | |
Collapse
|
46
|
Abstract
A propensity for violent behaviors to develop in chronic methamphetamine (METH) abusers has been noted. The idea that increased aggressiveness might result from chronic METH administration was tested in mice after chronic (long-term intermittent, 8 weeks) or single exposures to the drug. A single injection of METH (6 mg/kg) did not augment fighting. In contrast, chronic METH administration significantly increased the number of animals that initiated bite attacks. This regimen also shortened the latency before the first attack. Latency before the first attack was shorter at 20 h after the METH injection than at 15 min after injection. Locomotor activity was not different at 20 h after METH injection, indicating that increased fighting was not secondary to METH-induced hyperactivity. METH-induced increases in fighting were not related to the duration of persistent sniffing after the initial encounter with an intruder since the duration of this behavior was significantly increased at 15 min after METH but not at 20 h post drug. These results indicate that repeated injections of METH can increase fighting behaviors and also alter social interactions in mice. Thus, intermittent administration of METH might be useful as a pharmacological model to study the biochemical and molecular bases of aggressiveness.
Collapse
Affiliation(s)
- Boris P Sokolov
- Molecular Neuropsychiatry Branch, National Institute on Drug Abuse, NIH, DHHS, 5500 Nathan Shock Drive, Baltimore, MD 21224, USA
| | | | | |
Collapse
|
47
|
Cadet JL, Jayanthi S, Deng X. Speed kills: cellular and molecular bases of methamphetamine‐induced nerve terminal degeneration and neuronal apoptosis. FASEB J 2003; 17:1775-88. [PMID: 14519657 DOI: 10.1096/fj.03-0073rev] [Citation(s) in RCA: 228] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
Methamphetamine (METH) is a drug of abuse that has long been known to damage monoaminergic systems in the mammalian brain. Recent reports have provided conclusive evidence that METH can cause neuropathological changes in the rodent brain via apoptotic mechanisms akin to those reported in various models of neuronal death. The purpose of this review is to provide an interim account for a role of oxygen-based radicals and the participation of transcription factors and the involvement of cell death genes in METH-induced neurodegeneration. We discuss data suggesting the participation of endoplasmic reticulum and mitochondria-mediated activation of caspase-dependent and -independent cascades in the manifestation of METH-induced apoptosis. Studies that use more comprehensive approaches to gene expression profiling should allow us to draw more instructive molecular portraits of the complex plastic and degenerative effects of this drug.
Collapse
Affiliation(s)
- Jean Lud Cadet
- Molecular Neuropsychiatry Branch, NIH, NIDA, Intramural Research Program, Department of Health and Human Services, 5500 Nathan Shock Dr., Baltimore, Maryland 21224, USA.
| | | | | |
Collapse
|
48
|
Kita T, Wagner GC, Nakashima T. Current research on methamphetamine-induced neurotoxicity: animal models of monoamine disruption. J Pharmacol Sci 2003; 92:178-95. [PMID: 12890883 DOI: 10.1254/jphs.92.178] [Citation(s) in RCA: 128] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/27/2022] Open
Abstract
Methamphetamine (METH)-induced neurotoxicity is characterized by a long-lasting depletion of striatal dopamine (DA) and serotonin as well as damage to striatal dopaminergic and serotonergic nerve terminals. Several hypotheses regarding the mechanism underlying METH-induced neurotoxicity have been proposed. In particular, it is thought that endogenous DA in the striatum may play an important role in mediating METH-induced neuronal damage. This hypothesis is based on the observation of free radical formation and oxidative stress produced by auto-oxidation of DA consequent to its displacement from synaptic vesicles to cytoplasm. In addition, METH-induced neurotoxicity may be linked to the glutamate and nitric oxide systems within the striatum. Moreover, using knockout mice lacking the DA transporter, the vesicular monoamine transporter 2, c-fos, or nitric oxide synthetase, it was determined that these factors may be connected in some way to METH-induced neurotoxicity. Finally a role for apoptosis in METH-induced neurotoxicity has also been established including evidence of protection of bcl-2, expression of p53 protein, and terminal deoxynucleotidyl transferase-mediated dUTP nick-end labeling (TUNEL), activity of caspase-3. The neuronal damage induced by METH may reflect neurological disorders such as autism and Parkinson's disease.
Collapse
Affiliation(s)
- Taizo Kita
- Department of Pharmacology, Nara Medical University, Nara, Japan
| | | | | |
Collapse
|
49
|
Smith L, Yonekura ML, Wallace T, Berman N, Kuo J, Berkowitz C. Effects of prenatal methamphetamine exposure on fetal growth and drug withdrawal symptoms in infants born at term. J Dev Behav Pediatr 2003; 24:17-23. [PMID: 12584481 DOI: 10.1097/00004703-200302000-00006] [Citation(s) in RCA: 116] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
To determine fetal growth and the incidence of withdrawal symptoms in term infants exposed to methamphetamine in utero, we retrospectively identified neonates whose mothers used methamphetamine during pregnancy and matched them to unexposed newborns. Exclusion criteria included multiple and preterm gestations. Although there were no differences in infant growth parameters between the methamphetamine-exposed and methamphetamine-unexposed neonates, methamphetamine exposure throughout gestation was associated with decreased growth relative to infants exposed only for the first two trimesters. In addition, there were significantly more small for gestational age infants in the methamphetamine group compared with the unexposed group. Methamphetamine-exposed infants whose mothers smoked had significantly decreased growth relative to infants exposed to methamphetamine alone. Withdrawal symptoms (as determined by a previously reported scoring system) requiring pharmacologic intervention were observed in 4% of methamphetamine-exposed infants. These preliminary findings indicate that methamphetamine use is associated with growth restriction in infants born at term.
Collapse
Affiliation(s)
- Lynne Smith
- Department of Pediatrics, Harbor-UCLA Medical Center, University of California, Los Angeles School of Medicine, USA.
| | | | | | | | | | | |
Collapse
|
50
|
Buffum JC, Shulgin AT. Overdose of 2.3 grams of intravenous methamphetamine: case, analysis and patient perspective. J Psychoactive Drugs 2001; 33:409-12. [PMID: 11824700 DOI: 10.1080/02791072.2001.10399926] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/17/2022]
Abstract
The patient-reported toxicity of an overdose of intravenous methamphetamine is described. The authors report the case of a 34-year old man who inadvertently injected himself with approximately 2.3 grams of methamphetamine. The patient reported disorientation, hallucinations, hyperthermia, photophobia, orthostasis and extreme ataxia. He recovered in seven days without apparent sequelae. The case demonstrates the unusual, temporary neurophysiologic consequences of high-dose intravenous methamphetamine.
Collapse
Affiliation(s)
- J C Buffum
- University of California at San Francisco, USA.
| | | |
Collapse
|