1
|
Annawald K, Streckfuss-Bömeke K, Meyer T. Methamphetamine-induced cardiotoxicity: in search of protective transcriptional mechanisms. Herz 2024; 49:434-440. [PMID: 39455447 PMCID: PMC11602861 DOI: 10.1007/s00059-024-05279-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/30/2024] [Indexed: 10/28/2024]
Abstract
Crystalline methamphetamine hydrochloride is an illegal drug with a high addictive potential, better known by its colloquial name "ice" or "crystal meth". The abuse of this drug has led to significant health problems worldwide. Like other amphetamine-type stimulants, chronic consumption of methamphetamine leads to direct toxic effects on the central nervous system, causing cognitive impairment, depressive behavior, and other severe neurological or psychiatric symptoms. Besides its neurotoxicity, the drug exhibits numerous deleterious effects on the cardiovascular system, including hypertension, accelerated atherosclerosis, vasospasm-induced acute coronary syndromes, sudden cardiac death, and dilated cardiomyopathy with congestive heart failure and left ventricular dysfunction. The excessive release of catecholamines upon methamphetamine exposure causes vasoconstriction and vasospasm, which ultimately lead to hypertension, tachycardia, endothelial dysfunction, and cardiotoxicity. While numerous studies have focused on transcription factors expressed in the brain that cause the neurotoxic effects of the drug, much less is known about transcription factors involved in the development of methamphetamine-induced heart failure. In this article, we provide an overview of the Janus kinase-signal transducer and activator of transcription 3 (JAK-STAT3) pathway involved in ischemia/reperfusion injury in the myocardium, which may be activated by the vasospasm-inducing action of the drug. However, much more work is needed to decipher the precise role of STAT protein family members, including the potentially cardioprotective STAT3, in the pathogenesis of methamphetamine-induced cardiotoxicity.
Collapse
Affiliation(s)
- Kristin Annawald
- Department of Psychosomatic Medicine and Psychotherapy, University of Göttingen, Waldweg 33, 37073, Göttingen, Germany
| | | | - Thomas Meyer
- Department of Psychosomatic Medicine and Psychotherapy, University of Göttingen, Waldweg 33, 37073, Göttingen, Germany.
| |
Collapse
|
2
|
Bjorness TE, Greene RW. Orexin-mediated motivated arousal and reward seeking. Peptides 2024; 180:171280. [PMID: 39159833 DOI: 10.1016/j.peptides.2024.171280] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/19/2024] [Revised: 08/03/2024] [Accepted: 08/05/2024] [Indexed: 08/21/2024]
Abstract
The neuromodulator orexin has been identified as a key factor for motivated arousal including recent evidence that sleep deprivation-induced enhancement of reward behavior is modulated by orexin. While orexin is not necessary for either reward or arousal behavior, orexin neurons' broad projections, ability to sense the internal state of the animal, and high plasticity of signaling in response to natural rewards and drugs of abuse may underlie heightened drug seeking, particularly in a subset of highly motivated reward seekers. As such, orexin receptor antagonists have gained deserved attention for putative use in addiction treatments. Ongoing and future clinical trials are expected to identify individuals most likely to benefit from orexin receptor antagonist treatment to promote abstinence, such as those with concurrent sleep disorders or high craving, while attention to methodological considerations will aid interpretation of the numerous preclinical studies investigating disparate aspects of the role of orexin in reward and arousal.
Collapse
Affiliation(s)
- Theresa E Bjorness
- Research Service, VA North Texas Health Care System, Dallas, TX 75126, USA; Departments of Psychiatry University of Texas Southwestern Medical Center, Dallas, TX 75390-9111, USA.
| | - Robert W Greene
- Departments of Psychiatry University of Texas Southwestern Medical Center, Dallas, TX 75390-9111, USA; Department of Neuroscience, Peter O'Donnell Jr. Brain Institute, University of Texas Southwestern Medical Center, Dallas, TX 75390-9111, USA; International Institute for Integrative Sleep Medicine, University of Tsukuba, Tsukuba 305-8577, Japan
| |
Collapse
|
3
|
Zhang L, Meng S, Huang E, Di T, Ding Z, Huang S, Chen W, Zhang J, Zhao S, Yuwen T, Chen Y, Xue Y, Wang F, Shi J, Shi Y. High frequency deep brain stimulation of the dorsal raphe nucleus prevents methamphetamine priming-induced reinstatement of drug seeking in rats. Transl Psychiatry 2024; 14:190. [PMID: 38622130 PMCID: PMC11018621 DOI: 10.1038/s41398-024-02895-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/16/2023] [Revised: 03/23/2024] [Accepted: 03/28/2024] [Indexed: 04/17/2024] Open
Abstract
Drug addiction represents a multifaceted and recurrent brain disorder that possesses the capability to create persistent and ineradicable pathological memory. Deep brain stimulation (DBS) has shown a therapeutic potential for neuropsychological disorders, while the precise stimulation targets and therapeutic parameters for addiction remain deficient. Among the crucial brain regions implicated in drug addiction, the dorsal raphe nucleus (DRN) has been found to exert an essential role in the manifestation of addiction memory. Thus, we investigated the effects of DRN DBS in the treatment of addiction and whether it might produce side effects by a series of behavioral assessments, including methamphetamine priming-induced reinstatement of drug seeking behaviors, food-induced conditioned place preference (CPP), open field test and elevated plus-maze test, and examined brain activity and connectivity after DBS of DRN. We found that high-frequency DBS of the DRN significantly lowered the CPP scores and the number of active-nosepokes in the methamphetamine-primed CPP test and the self-administration model. Moreover, both high-frequency and sham DBS group rats were able to establish significant food-induced place preference, and no significant difference was observed in the open field test and in the elevated plus-maze test between the two groups. Immunofluorescence staining and functional magnetic resonance imaging revealed that high-frequency DBS of the DRN could alter the activity and functional connectivity of brain regions related to addiction. These results indicate that high-frequency DBS of the DRN effectively inhibits methamphetamine priming-induced relapse and seeking behaviors in rats and provides a new target for the treatment of drug addiction.
Collapse
Affiliation(s)
- Libo Zhang
- Shenzhen Key Laboratory for Drug Addiction and Medication Safety, Shenzhen Public Service Platform for Clinical Application of Medical Imaging, Department of Ultrasound, Peking University Shenzhen Hospital, Shenzhen-PKU-HKUST Medical Center, Shenzhen, China
- National Institute on Drug Dependence and Beijing Key Laboratory of Drug Dependence Research, Peking University, Beijing, China
| | - Shiqiu Meng
- National Institute on Drug Dependence and Beijing Key Laboratory of Drug Dependence Research, Peking University, Beijing, China
| | - Enze Huang
- National Institute on Drug Dependence and Beijing Key Laboratory of Drug Dependence Research, Peking University, Beijing, China
| | - Tianqi Di
- National Institute on Drug Dependence and Beijing Key Laboratory of Drug Dependence Research, Peking University, Beijing, China
| | - Zengbo Ding
- National Institute on Drug Dependence and Beijing Key Laboratory of Drug Dependence Research, Peking University, Beijing, China
| | - Shihao Huang
- National Institute on Drug Dependence and Beijing Key Laboratory of Drug Dependence Research, Peking University, Beijing, China
| | - Wenjun Chen
- National Institute on Drug Dependence and Beijing Key Laboratory of Drug Dependence Research, Peking University, Beijing, China
| | - Jiayi Zhang
- National Institute on Drug Dependence and Beijing Key Laboratory of Drug Dependence Research, Peking University, Beijing, China
| | - Shenghong Zhao
- National Institute on Drug Dependence and Beijing Key Laboratory of Drug Dependence Research, Peking University, Beijing, China
| | - Ting Yuwen
- National Institute on Drug Dependence and Beijing Key Laboratory of Drug Dependence Research, Peking University, Beijing, China
| | - Yang Chen
- National Institute on Drug Dependence and Beijing Key Laboratory of Drug Dependence Research, Peking University, Beijing, China
| | - Yanxue Xue
- National Institute on Drug Dependence and Beijing Key Laboratory of Drug Dependence Research, Peking University, Beijing, China
| | - Feng Wang
- Shenzhen Key Laboratory for Drug Addiction and Medication Safety, Shenzhen Public Service Platform for Clinical Application of Medical Imaging, Department of Ultrasound, Peking University Shenzhen Hospital, Shenzhen-PKU-HKUST Medical Center, Shenzhen, China
| | - Jie Shi
- Shenzhen Key Laboratory for Drug Addiction and Medication Safety, Shenzhen Public Service Platform for Clinical Application of Medical Imaging, Department of Ultrasound, Peking University Shenzhen Hospital, Shenzhen-PKU-HKUST Medical Center, Shenzhen, China.
- National Institute on Drug Dependence and Beijing Key Laboratory of Drug Dependence Research, Peking University, Beijing, China.
- Henan Collaborative Innovation Center of Prevention and Treatment of Mental Disorder, the Second Affiliated Hospital of Xinxiang Medical University, Xinxiang, China.
| | - Yu Shi
- Shenzhen Key Laboratory for Drug Addiction and Medication Safety, Shenzhen Public Service Platform for Clinical Application of Medical Imaging, Department of Ultrasound, Peking University Shenzhen Hospital, Shenzhen-PKU-HKUST Medical Center, Shenzhen, China.
| |
Collapse
|
4
|
Nazari-Serenjeh F, Azizbeigi R, Rashvand M, Mesgar S, Amirteymori H, Haghparast A. Distinct roles for orexin-1 and orexin-2 receptors in the dentate gyrus of the hippocampus in the methamphetamine-seeking behavior in the rats. Life Sci 2023; 312:121262. [PMID: 36470538 DOI: 10.1016/j.lfs.2022.121262] [Citation(s) in RCA: 10] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2022] [Revised: 11/26/2022] [Accepted: 11/29/2022] [Indexed: 12/11/2022]
Abstract
Because of the relapsing properties of psychostimulants such as methamphetamine (Meth), there is no established pharmacotherapy for Meth addiction. The orexinergic system is a promising target for treating psychostimulant use disorders and relapse. However, to the best of our knowledge, no investigation regarding the role of orexin receptors in the dentate gyrus (DG) region of the hippocampus has been conducted in the extinction and reinstatement of Meth-seeking behavior. Two stainless-steel guide cannulae were bilaterally implanted into the DG of the rats' brains. The unbiased conditioned place preference (CPP) procedure was conducted to induce Meth conditioning. Following the five days Meth injections (1 mg/kg; sc), animals received intra-DG microinjection of SB334867 or TCS OX2 29, as orexin 1 (OX1) or orexin 2 (OX2) receptor antagonists, respectively (without Meth administration) during extinction phase to elucidate the role of orexin receptors in the latency of the extinction period in the Meth-conditioned rats. To evaluate the role of orexin receptors in the DG region in the reinstatement of Meth-seeking behavior, the extinguished rats received SB334867 or TCS OX2 29 before injecting a priming dose of Meth (0.25 mg/kg; sc). The results indicated two distinct roles for the OX1 and OX2 receptors in the DG region. TCS OX2 29 attenuated the extinction latency, and SB334867 considerably reduced the reinstatement of Meth-seeking behavior in this region. Therefore, the DG region's orexinergic system might be a potential therapeutic target for psychostimulant use disorders.
Collapse
Affiliation(s)
| | - Ronak Azizbeigi
- Department of Basic Sciences, Sanandaj Branch, Islamic Azad University, Sanandaj, Iran
| | - Mina Rashvand
- Neurophysiology Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Somaye Mesgar
- Neurobiology Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Haleh Amirteymori
- Neuroscience Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Abbas Haghparast
- Neuroscience Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran; School of Cognitive Sciences, Institute for Research in Fundamental Sciences, Tehran, Iran.
| |
Collapse
|
5
|
Kuebler IRK, Jolton JA, Hermreck C, Hubbard NA, Wakabayashi KT. Contrasting dose-dependent effects of acute intravenous methamphetamine on lateral hypothalamic extracellular glucose dynamics in male and female rats. J Neurophysiol 2022; 128:819-836. [PMID: 36043803 PMCID: PMC9529272 DOI: 10.1152/jn.00257.2022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2022] [Revised: 08/22/2022] [Accepted: 08/24/2022] [Indexed: 11/22/2022] Open
Abstract
Glucose is the brain's primary energetic resource. The brain's use of glucose is dynamic, balancing delivery from the neurovasculature with local metabolism. Although glucose metabolism is known to differ in humans with and without methamphetamine use disorder (MUD), it is unknown how central glucose regulation changes with acute methamphetamine experience. Here, we determined how intravenous methamphetamine regulates extracellular glucose levels in a brain region implicated in MUD-like behavior, the lateral hypothalamus (LH). We measured extracellular LH glucose in awake adult male and female drug-naive Wistar rats using enzyme-linked amperometric glucose biosensors. Changes in LH glucose were monitored during a single session after: 1) natural nondrug stimuli (novel object presentation and a tail-touch), 2) increasing cumulative doses of intravenous methamphetamine (0.025, 0.05, 0.1, and 0.2 mg/kg), and 3) an injection of 60 mg of glucose. We found second-scale fluctuations in LH glucose in response to natural stimuli that differed by both stimulus type and sex. Although rapid, second-scale changes in LH glucose during methamphetamine injections were variable, slow, minute-scale changes following most injections were robust and resulted in a reduction in LH glucose levels. Dose and sex differences at this timescale indicated that female rats may be more sensitive to the impact of methamphetamine on central glucose regulation. These findings suggest that the effects of MUD on healthy brain function may be linked to how methamphetamine alters extracellular glucose regulation in the LH and point to possible mechanisms by which methamphetamine influences central glucose metabolism more broadly.NEW & NOTEWORTHY Enzyme-linked glucose biosensors were used to monitor lateral hypothalamic (LH) extracellular fluctuations during nondrug stimuli and intravenous methamphetamine injections in drug-naive awake male and female rats. Second-scale glucose changes occurred after nondrug stimuli, differing by modality and sex. Robust minute-scale decreases followed most methamphetamine injections. Sex differences at the minute-scale indicate female central glucose regulation is more sensitive to methamphetamine effects. We discuss likely mechanisms underlying these fluctuations, and their implications in methamphetamine use disorder.
Collapse
Affiliation(s)
- Isabel R K Kuebler
- Neurocircuitry of Motivated Behavior Laboratory, Department of Psychology, University of Nebraska-Lincoln, Lincoln, Nebraska
| | - Joshua A Jolton
- Neurocircuitry of Motivated Behavior Laboratory, Department of Psychology, University of Nebraska-Lincoln, Lincoln, Nebraska
| | - Chase Hermreck
- Neurocircuitry of Motivated Behavior Laboratory, Department of Psychology, University of Nebraska-Lincoln, Lincoln, Nebraska
| | - Nicholas A Hubbard
- Neurocircuitry of Motivated Behavior Laboratory, Department of Psychology, University of Nebraska-Lincoln, Lincoln, Nebraska
| | - Ken T Wakabayashi
- Neurocircuitry of Motivated Behavior Laboratory, Department of Psychology, University of Nebraska-Lincoln, Lincoln, Nebraska
| |
Collapse
|
6
|
Inactivation of the Lateral Hypothalamus Attenuates Methamphetamine-Induced Conditioned Place Preference through Regulation of Kcnq3 Expression. Int J Mol Sci 2022; 23:ijms23137305. [PMID: 35806315 PMCID: PMC9266452 DOI: 10.3390/ijms23137305] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2022] [Revised: 06/25/2022] [Accepted: 06/26/2022] [Indexed: 12/03/2022] Open
Abstract
Repeated administration of methylamphetamine (MA) induces MA addiction, which is featured by awfully unpleasant physical and emotional experiences after drug use is terminated. Neurophysiological studies show that the lateral hypothalamus (LH) is involved in reward development and addictive behaviors. Here, we show that repeated administration of MA activates the expression of c-Fos in LH neurons responding to conditioned place preference (CPP). Chemogenetic inhibition of the LH can disrupt the addiction behavior, demonstrating that the LH plays an important role in MA-induced reward processing. Critically, MA remodels the neurons of LH synaptic plasticity, increases intracellular calcium level, and enhances spontaneous current and evoked potentials of neurons compared to the saline group. Furthermore, overexpression of the potassium voltage-gated channel subfamily Q member 3 (Kcnq3) expression can reverse the CPP score and alleviate the occurrence of addictive behaviors. Together, these results unravel a new neurobiological mechanism underlying the MA-induced addiction in the lateral hypothalamus, which could pave the way toward new and effective interventions for this addiction disease.
Collapse
|
7
|
Wang X, Tong B, Hui R, Hou C, Zhang Z, Zhang L, Xie B, Ni Z, Cong B, Ma C, Wen D. The Role of Hyperthermia in Methamphetamine-Induced Depression-Like Behaviors: Protective Effects of Coral Calcium Hydride. Front Mol Neurosci 2022; 14:808807. [PMID: 35058751 PMCID: PMC8764150 DOI: 10.3389/fnmol.2021.808807] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2021] [Accepted: 12/06/2021] [Indexed: 12/15/2022] Open
Abstract
Methamphetamine (METH) abuse causes irreversible damage to the central nervous system and leads to psychiatric symptoms including depression. Notably, METH-induced hyperthermia is a crucial factor in the development of these symptoms, as it aggravates METH-induced neurotoxicity. However, the role of hyperthermia in METH-induced depression-like behaviors needs to be clarified. In the present study, we treated mice with different doses of METH under normal (NAT) or high ambient temperatures (HAT). We found that HAT promoted hyperthermia after METH treatment and played a key role in METH-induced depression-like behaviors in mice. Intriguingly, chronic METH exposure (10 mg/kg, 7 or 14 days) or administration of an escalating-dose (2 ∼ 15 mg/kg, 3 days) of METH under NAT failed to induce depression-like behaviors. However, HAT aggravated METH-induced damage of hippocampal synaptic plasticity, reaction to oxidative stress, and neuroinflammation. Molecular hydrogen acts as an antioxidant and anti-inflammatory agent and has been shown to have preventive and therapeutic applicability in a wide range of diseases. Coral calcium hydride (CCH) is a newly identified hydrogen-rich powder which produces hydrogen gas gradually when exposed to water. Herein, we found that CCH pretreatment significantly attenuated METH-induced hyperthermia, and administration of CCH after METH exposure also inhibited METH-induced depression-like behaviors and reduced the hippocampal synaptic plasticity damage. Moreover, CCH effectively reduced the activity of lactate dehydrogenase and decreased malondialdehyde, TNF-α and IL-6 generation in hippocampus. These results suggest that CCH is an efficient hydrogen-rich agent, which has a potential therapeutic applicability in the treatment of METH abusers.
Collapse
Affiliation(s)
- Xintao Wang
- College of Forensic Medicine, Hebei Medical University, Shijiazhuang, China
- Hebei Key Laboratory of Forensic Medicine, Collaborative Innovation Center of Forensic Medical Molecular Identification, Shijiazhuang, China
- Research Unit of Digestive Tract Microecosystem Pharmacology and Toxicology, Chinese Academy of Medical Sciences, Shijiazhuang, China
| | - Bonan Tong
- College of Forensic Medicine, Hebei Medical University, Shijiazhuang, China
- Hebei Key Laboratory of Forensic Medicine, Collaborative Innovation Center of Forensic Medical Molecular Identification, Shijiazhuang, China
- Research Unit of Digestive Tract Microecosystem Pharmacology and Toxicology, Chinese Academy of Medical Sciences, Shijiazhuang, China
| | - Rongji Hui
- College of Forensic Medicine, Hebei Medical University, Shijiazhuang, China
- Hebei Key Laboratory of Forensic Medicine, Collaborative Innovation Center of Forensic Medical Molecular Identification, Shijiazhuang, China
- Research Unit of Digestive Tract Microecosystem Pharmacology and Toxicology, Chinese Academy of Medical Sciences, Shijiazhuang, China
| | - Congcong Hou
- College of Forensic Medicine, Hebei Medical University, Shijiazhuang, China
- Hebei Key Laboratory of Forensic Medicine, Collaborative Innovation Center of Forensic Medical Molecular Identification, Shijiazhuang, China
- Research Unit of Digestive Tract Microecosystem Pharmacology and Toxicology, Chinese Academy of Medical Sciences, Shijiazhuang, China
| | - Zilu Zhang
- The First Clinical Medical College of Peking University Health Science Center, Peking University, Beijing, China
| | - Ludi Zhang
- College of Forensic Medicine, Hebei Medical University, Shijiazhuang, China
- Hebei Key Laboratory of Forensic Medicine, Collaborative Innovation Center of Forensic Medical Molecular Identification, Shijiazhuang, China
- Research Unit of Digestive Tract Microecosystem Pharmacology and Toxicology, Chinese Academy of Medical Sciences, Shijiazhuang, China
| | - Bing Xie
- College of Forensic Medicine, Hebei Medical University, Shijiazhuang, China
- Hebei Key Laboratory of Forensic Medicine, Collaborative Innovation Center of Forensic Medical Molecular Identification, Shijiazhuang, China
- Research Unit of Digestive Tract Microecosystem Pharmacology and Toxicology, Chinese Academy of Medical Sciences, Shijiazhuang, China
| | - Zhiyu Ni
- School of Basic Medical Sciences, Hebei University, Baoding, China
| | - Bin Cong
- College of Forensic Medicine, Hebei Medical University, Shijiazhuang, China
- Hebei Key Laboratory of Forensic Medicine, Collaborative Innovation Center of Forensic Medical Molecular Identification, Shijiazhuang, China
- Research Unit of Digestive Tract Microecosystem Pharmacology and Toxicology, Chinese Academy of Medical Sciences, Shijiazhuang, China
| | - Chunling Ma
- College of Forensic Medicine, Hebei Medical University, Shijiazhuang, China
- Hebei Key Laboratory of Forensic Medicine, Collaborative Innovation Center of Forensic Medical Molecular Identification, Shijiazhuang, China
- Research Unit of Digestive Tract Microecosystem Pharmacology and Toxicology, Chinese Academy of Medical Sciences, Shijiazhuang, China
- *Correspondence: Chunling Ma,
| | - Di Wen
- College of Forensic Medicine, Hebei Medical University, Shijiazhuang, China
- Hebei Key Laboratory of Forensic Medicine, Collaborative Innovation Center of Forensic Medical Molecular Identification, Shijiazhuang, China
- Research Unit of Digestive Tract Microecosystem Pharmacology and Toxicology, Chinese Academy of Medical Sciences, Shijiazhuang, China
- Di Wen,
| |
Collapse
|
8
|
Zlebnik NE, Holtz NA, Lepak VC, Saykao AT, Zhang Y, Carroll ME. Age-specific treatment effects of orexin/hypocretin-receptor antagonism on methamphetamine-seeking behavior. Drug Alcohol Depend 2021; 224:108719. [PMID: 33940327 PMCID: PMC8180489 DOI: 10.1016/j.drugalcdep.2021.108719] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/13/2020] [Revised: 03/09/2021] [Accepted: 03/10/2021] [Indexed: 01/02/2023]
Abstract
BACKGROUND Worldwide methamphetamine (METH) use has increased significantly over the last 10 years, and in the US, METH dependence has sky-rocketed among individuals with opioid use disorder. Of significant concern, METH use is gaining popularity among groups with susceptibility to developing severe substance use disorders, such as women and adolescents. Nevertheless, there is no established pharmacotherapy for METH addiction. Emerging evidence has identified the orexin/hypocretin system as an important modulator of reward-driven behavior and a potential target for the treatment of drug addiction and relapse. However, to date, there have been no investigations into the therapeutic efficacy of orexin/hypocretin receptor antagonists for METH-motivated behavior in adolescents or adults. In the present study, we examined the effects of selective antagonists of the orexin-1 (SB-334867, 20 mg/kg) and orexin-2 (TCS-OX2-29, 20 mg/kg) receptors on the reinstatement of METH seeking in both adolescent and adult male and female rats. METHODS Rats were trained to self-administer METH (0.05 mg/kg/inf, iv) during two 2-h sessions/day for 5 days. Following 20 sessions of extinction over 10 days, a within-subjects design was used to test for METH seeking precipitated by METH (1 mg/kg, ip) or METH cues after systemic pretreatment with SB-334867 or TCS-OX2-29. RESULTS SB-334867 reduced cue-induced reinstatement in males and females, regardless of age. Additionally, METH-induced METH seeking was attenuated by SB-334867 in adolescents and by TCS-OX2-29 in adults. CONCLUSION Selective orexin/hypocretin receptor antagonists have significant therapeutic potential for diminishing METH-seeking behavior, although their treatment efficacy may be influenced by age.
Collapse
Affiliation(s)
- Natalie E Zlebnik
- Department of Psychiatry, University of Minnesota Medical School, Minneapolis, MN, 55455, USA.
| | - Nathan A Holtz
- Department of Psychiatry, University of Minnesota Medical School, Minneapolis, MN, 55455, USA
| | - Victoria C Lepak
- Department of Psychiatry, University of Minnesota Medical School, Minneapolis, MN, 55455, USA
| | - Amy T Saykao
- Department of Psychiatry, University of Minnesota Medical School, Minneapolis, MN, 55455, USA
| | - Yanan Zhang
- Research Triangle Institute, Research Triangle Park, NC, 27709, USA
| | - Marilyn E Carroll
- Department of Psychiatry, University of Minnesota Medical School, Minneapolis, MN, 55455, USA
| |
Collapse
|
9
|
Sepulveda M, Manning EE, Gogos A, Hale M, van den Buuse M. Long-term effects of young-adult methamphetamine on dorsal raphe serotonin systems in mice: Role of brain-derived neurotrophic factor. Brain Res 2021; 1762:147428. [PMID: 33737066 DOI: 10.1016/j.brainres.2021.147428] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2020] [Revised: 02/15/2021] [Accepted: 03/08/2021] [Indexed: 01/21/2023]
Abstract
To assess the long-term effects of chronic adolescent methamphetamine (METH) treatment on the serotonin system in the brain, we used serotonin-1A receptor (5-HT1A) and serotonin transporter (SERT) autoradiography, and quantitative tryptophan-hydroxylase 2 (TPH2) immunohistochemistry in the raphe nuclei of mice. Because of the modulatory role of brain-derived neurotrophic factor (BDNF) on the serotonin system and the effects of METH, we included both BDNF heterozygous (HET) mice and wildtype (WT) controls. Male and female mice of both genotypes were treated with an escalating METH dose regimen from the age of 6-9 weeks. At least two weeks later, acute locomotor hyperactivity induced by a 5 mg/kg D-amphetamine challenge was significantly enhanced in METH-pretreated mice, showing long-term sensitisation. METH pretreatment caused a small, but significant decrease of 5-HT1A receptor binding in the dorsal raphe nucleus (DRN) of males independent of genotype, but there were no changes in the median raphe nucleus (MRN) or in SERT binding density. METH treatment reduced the number of TPH2 positive cells in ventral subregions of the rostral and medial DRN independent of genotype. METH treatment selectively reduced DRN cell counts in BDNF HET mice compared to wildtype mice in medial and caudal ventrolateral subregions previously associated with panic-like behaviour. The data increase our understanding of the long-term and selective effects of METH on brain serotonin systems. These findings could be relevant for some of the psychosis-like symptoms associated with long-term METH use.
Collapse
Affiliation(s)
- Mauricio Sepulveda
- Florey Institute for Neuroscience and Mental Health, University of Melbourne, Australia
| | - Elizabeth E Manning
- Florey Institute for Neuroscience and Mental Health, University of Melbourne, Australia
| | - Andrea Gogos
- Florey Institute for Neuroscience and Mental Health, University of Melbourne, Australia
| | - Matthew Hale
- School of Psychology and Public Health, La Trobe University, Melbourne, Australia
| | - Maarten van den Buuse
- School of Psychology and Public Health, La Trobe University, Melbourne, Australia; Department of Pharmacology, University of Melbourne, Australia; College of Public Health, Medical and Veterinary Sciences, James Cook University, Townsville, Australia.
| |
Collapse
|
10
|
Pittenger ST, Chou S, Murawski NJ, Barrett ST, Loh O, Duque JF, Li M, Bevins RA. Female rats display higher methamphetamine-primed reinstatement and c-Fos immunoreactivity than male rats. Pharmacol Biochem Behav 2021; 201:173089. [PMID: 33422599 PMCID: PMC9067906 DOI: 10.1016/j.pbb.2020.173089] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/19/2020] [Revised: 12/07/2020] [Accepted: 12/08/2020] [Indexed: 11/20/2022]
Abstract
Methamphetamine (meth) dependence is often characterized by persistent and chronic relapse (i.e., return to drug use). Previous work suggests females may be at greater risk to relapse. In this study, we extended this limited evidence and identified sex-dependent neural substrates related to meth-triggered reinstatement. Male and female Sprague-Dawley rats were implanted with indwelling jugular catheters. Half of the rats were then trained to self-administer meth (0.05 mg/kg/inf); the other half self-administered saline during 21 daily sessions (2 h). Rats were then given 12 extinction sessions. Twenty-four hours after the last extinction session, rats received reinstatement testing. Half of the rats received a meth-prime (0.3 mg/kg, IP) injection and the remaining rats received a saline injection. This design resulted in 4 separate groups for each sex, allowing for careful investigation of brain regions related to meth-triggered reinstatement. Brains were harvested following the reinstatement session and c-Fos immunoreactivity was measured in multiple brain regions. Meth triggered reinstatement in both sexes and this effect was more robust in females compared to males. Significant sex differences were detected. Females showed greater c-Fos immunoreactivity in the cingulate cortex area 1, lateral orbitofrontal cortex, prelimbic cortex, caudate-putamen, nucleus accumbens core and shell, and central nucleus of the amygdala following meth-primed reinstatement.
Collapse
Affiliation(s)
- Steven T Pittenger
- University of Nebraska-Lincoln, Department of Psychology, United States of America
| | - Shinnyi Chou
- University of Pittsburgh Medical Center, United States of America
| | | | - Scott T Barrett
- University of Nebraska-Lincoln, Department of Psychology, United States of America
| | - Olivia Loh
- University of Colorado-Denver, United States of America
| | - Juan F Duque
- Arcadia University, Department of Psychology, United States of America
| | - Ming Li
- University of Nebraska-Lincoln, Department of Psychology, United States of America
| | - Rick A Bevins
- University of Nebraska-Lincoln, Department of Psychology, United States of America.
| |
Collapse
|
11
|
New designer phenethylamines 2C-C and 2C-P have abuse potential and induce neurotoxicity in rodents. Arch Toxicol 2021; 95:1413-1429. [PMID: 33515270 DOI: 10.1007/s00204-021-02980-x] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2020] [Accepted: 01/04/2021] [Indexed: 12/22/2022]
Abstract
2C (2C-x) is the general name for the family of phenethylamines containing two methoxy groups at the 2 and 5 positions of the benzene ring. The abuse of 2C family drugs has grown rapidly, although the abuse potential and neurotoxic properties of 2C drugs have not yet been fully investigated. In this study, we investigated the abuse potential and neurotoxicity of 4-chloro-2,5-dimethoxyphenethylamine (2C-C) and 2,5-dimethoxy-4-propylphenethylamine (2C-P). We found that 2C-C and 2C-P produced conditioned place preference in a dose-dependent manner in mice, and increased self-administration in rats, suggesting that 2C-C and 2C-P have abuse potential. To investigate the neurotoxicity of 2C-C and 2C-P, we examined motor performance and memory impairment after high doses of 2C-C and 2C-P. High doses of 2C-C and 2C-P decreased locomotor activity, rota-rod performance, and lower Y-maze test, novel objective recognition test, and passive avoidance test scores. We also observed that 2C-C and 2C-P affected expression levels of the D1 dopamine receptor, D2 dopamine receptor, dopamine transporter, and phospho-dopamine transporter in the nucleus accumbens and the medial prefrontal cortex, and increased c-Fos immuno-positive cells in the nucleus accumbens. Moreover, high doses of 2C-C and 2C-P induced microglial activation, which is involved in the inflammatory reaction in the striatum. These results suggest that 2C-C and 2C-P have abuse potential by affecting dopaminergic signaling and induce neurotoxicity via initiating neuroinflammation at high doses.
Collapse
|
12
|
Tavakkolifard M, Vousooghi N, Mahboubi S, Golab F, Ejtemaei Mehr S, Zarrindast MR. Evaluation of the relationship between the gene expression level of orexin-1 receptor in the rat blood and prefrontal cortex, novelty-seeking, and proneness to methamphetamine dependence: A candidate biomarker. Peptides 2020; 131:170368. [PMID: 32668268 DOI: 10.1016/j.peptides.2020.170368] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/13/2019] [Revised: 06/19/2020] [Accepted: 07/06/2020] [Indexed: 01/01/2023]
Abstract
BACKGROUND previous studies have suggested that methamphetamine (METH) abuse may affect orexin regulation. However, the data regarding the relationship between the current level of orexin and the vulnerability to METH abuse are minimal. Here, we have investigated the correlation between the gene expression level of the orexin-1 receptor (OX1R) in the rat prefrontal cortex (PFC) and blood lymphocytes and susceptibility to METH dependence and its impact on novelty-seeking behavior. METHODS male Wistar rats were first examined for novelty-seeking behavior by the novel object recognition test, and the expression level of OX1R in their blood lymphocytes was evaluated by real-time PCR. Then, the susceptibility to METH abuse was investigated by voluntary METH oral consumption test. According to the amounts of METH consumption, the animals were divided into two groups of METH preferring and non-preferring. Half of the rats in each group were sacrificed, and the level of OX1R in their blood lymphocytes and PFC tissue was measured. The other half were sacrificed for the same reason after two weeks of drug abstinence. RESULTS The indexes of novelty-seeking behavior were significantly higher in the METH- preferring group compared to the non-preferring animals. Furthermore, the expression level of OX1R in the blood lymphocytes and PFC in the preferring group was considerably higher than the non-preferring group. CONCLUSION Up-regulation of the mRNA expression level of OX1R in the lymphocytes and PFC may predict vulnerability to the METH consumption and novelty-seeking, which may serve as a potential biomarker for METH abuse.
Collapse
Affiliation(s)
- Mahnoosh Tavakkolifard
- Department of Neuroscience and Addiction Studies, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Nasim Vousooghi
- Department of Neuroscience and Addiction Studies, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran, Iran; Research Center for Cognitive and Behavioral Sciences, Tehran University of Medical Sciences, Tehran, Iran; Iranian National Center for Addiction Studies (INCAS), Tehran University of Medical Sciences, Tehran, Iran.
| | - Sara Mahboubi
- Department of Neuroscience and Addiction Studies, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Fereshteh Golab
- Cellular and Molecular Research Center, Iran University of Medical Sciences, Tehran, Iran
| | - Shahram Ejtemaei Mehr
- Department of Pharmacology, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Mohammad Reza Zarrindast
- Iranian National Center for Addiction Studies (INCAS), Tehran University of Medical Sciences, Tehran, Iran; Department of Pharmacology, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran; Department of Cognitive Neuroscience, Institute for Cognitive Science Studies, Tehran, Iran.
| |
Collapse
|
13
|
Wetering R, Schenk S. Regional changes in ∆FosB expression in rat brain following MDMA self-administration predict increased sensitivity to effects of locally infused MDMA. Addict Biol 2020; 25:e12814. [PMID: 31373119 DOI: 10.1111/adb.12814] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2019] [Revised: 05/27/2019] [Accepted: 07/03/2019] [Indexed: 01/26/2023]
Abstract
Repeated exposure to drugs produces a plethora of persistent brain changes, some of which underlie the development of drug addiction. An important objective of addiction research is to identify the brain changes that might mediate the transition from drug use to drug misuse. The persistent accumulation of the transcription factor, ∆FosB, following repeated drug exposure provides a means of achieving this objective. Experiments were conducted on sexually mature male Sprague-Dawley rats. The effects of extensive 3,4-methylenedioxymethamphetamine (MDMA) self-administration on immunohistochemical measurements of ∆FosB accumulation in 12 brain regions was compared with a matched, drug-naive, control group. Other groups were pretreated with MDMA (0.0 or 10.0 mg/kg, ip, once daily for 5 days), and the locomotor-activating effect of MDMA (200 μg/side) microinjected bilaterally into brain regions selected on the basis of the ∆FosB results was subsequently determined. MDMA self-administration significantly increased ∆FosB expression in the nucleus accumbens core, ventromedial and dorsomedial caudate-putamen, anterior cingulate, prelimbic, infralimbic, and orbitofrontal cortex, and both the central and basolateral amygdala, but not in the ventrolateral or dorsolateral caudate-putamen. Increases in the nucleus accumbens shell were substantial but were not significant following statistical correction for multiple comparisons. MDMA pretreatment enhanced MDMA-produced hyperactivity only when administered into the nucleus accumbens or the medial, but not the lateral, caudate-putamen, mirroring the ∆FosB results. These data compare favorably to results following repeated exposure to other drugs of abuse and support the idea of common neuroplastic changes following repeated drug exposure.
Collapse
Affiliation(s)
- Ross Wetering
- School of PsychologyVictoria University of Wellington Wellington New Zealand
| | - Susan Schenk
- School of PsychologyVictoria University of Wellington Wellington New Zealand
| |
Collapse
|
14
|
Wen D, Hui R, Liu Y, Luo Y, Wang J, Shen X, Xie B, Yu F, Cong B, Ma C. Molecular hydrogen attenuates methamphetamine-induced behavioral sensitization and activation of ERK-ΔFosB signaling in the mouse nucleus accumbens. Prog Neuropsychopharmacol Biol Psychiatry 2020; 97:109781. [PMID: 31629777 DOI: 10.1016/j.pnpbp.2019.109781] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/30/2019] [Revised: 09/30/2019] [Accepted: 10/06/2019] [Indexed: 12/20/2022]
Abstract
Methamphetamine (METH) is one of the most prevalently used illegal psychostimulants in many countries. Continuous exposure to METH leads to behavioral sensitization in animals, which can be used as a behavioral model with many mechanisms in common with relapse in humans. Molecular hydrogen has recently gained attention for its potential as a novel healthcare product with preventive and therapeutic applicability to a wide range of pathological conditions. However, it remains unclear whether and, if so, how hydrogen regulates METH-induced behavioral abnormalities. In the present study, we investigated the roles of molecular hydrogen on the acquisition and transfer of METH-induced behavioral sensitization and the accompanying changes in ERK phosphorylation and ΔFosB activation in the nucleus accumbens (NAc) of mice. To this end, male C57BL/6 mice received METH (0.1, 0.5 and 1.0 mg/kg, i.p.) injections for 7 days followed by a METH challenge (0.1, 0.5 and 1.0 mg/kg, i.p.) after a 7-day transfer period. Molecular hydrogen, delivered through a hydrogen-rich saline (HRS) injection (10 mL/kg, i.p., 3-h interval), was administered during the acquisition and transfer periods. We found that HRS administration was able to inhibit the acquisition and transfer of 0.1 and 0.5 mg/kg METH-induced behavioral sensitization to a certain extent, thereby attenuating the expression of behavioral sensitization. The HRS injections alone did not induce any obvious changes in locomotor activity in mice. Intriguingly, the increases in pERK and ΔFosB in the NAc, which accompanied the METH-induced behavioral sensitization, were also attenuated by the HRS treatments. Due to the anti-oxidative function of molecular hydrogen, the HRS injections reduced METH-induced reactive oxygen species and malondialdehyde generation in the NAc. These results suggest that molecular hydrogen serves as an anti-oxidative agent with potentially therapeutic applicability to the treatment of METH addicts.
Collapse
Affiliation(s)
- Di Wen
- College of Forensic Medicine, Hebei Medical University, Hebei Key Laboratory of Forensic Medicine, Collaborative Innovation Center of Forensic Medical Molecular Identification, Hebei Province, Shijiazhuang 050017, PR China
| | - Rongji Hui
- College of Forensic Medicine, Hebei Medical University, Hebei Key Laboratory of Forensic Medicine, Collaborative Innovation Center of Forensic Medical Molecular Identification, Hebei Province, Shijiazhuang 050017, PR China
| | - Yi Liu
- College of Forensic Medicine, Hebei Medical University, Hebei Key Laboratory of Forensic Medicine, Collaborative Innovation Center of Forensic Medical Molecular Identification, Hebei Province, Shijiazhuang 050017, PR China
| | - Yixiao Luo
- Key Laboratory of Molecular Epidemiology of Hunan Province, School of Medicine, Hunan Normal University, Changsha 410081,China
| | - Jian Wang
- College of Forensic Medicine, Hebei Medical University, Hebei Key Laboratory of Forensic Medicine, Collaborative Innovation Center of Forensic Medical Molecular Identification, Hebei Province, Shijiazhuang 050017, PR China
| | - Xi Shen
- College of Public Health, Hebei Medical University, Hebei Province, Shijiazhuang 050017, PR China
| | - Bing Xie
- College of Forensic Medicine, Hebei Medical University, Hebei Key Laboratory of Forensic Medicine, Collaborative Innovation Center of Forensic Medical Molecular Identification, Hebei Province, Shijiazhuang 050017, PR China
| | - Feng Yu
- College of Forensic Medicine, Hebei Medical University, Hebei Key Laboratory of Forensic Medicine, Collaborative Innovation Center of Forensic Medical Molecular Identification, Hebei Province, Shijiazhuang 050017, PR China
| | - Bin Cong
- College of Forensic Medicine, Hebei Medical University, Hebei Key Laboratory of Forensic Medicine, Collaborative Innovation Center of Forensic Medical Molecular Identification, Hebei Province, Shijiazhuang 050017, PR China.
| | - Chunling Ma
- College of Forensic Medicine, Hebei Medical University, Hebei Key Laboratory of Forensic Medicine, Collaborative Innovation Center of Forensic Medical Molecular Identification, Hebei Province, Shijiazhuang 050017, PR China.
| |
Collapse
|
15
|
Tingley D, Buzsáki G. Routing of Hippocampal Ripples to Subcortical Structures via the Lateral Septum. Neuron 2020; 105:138-149.e5. [PMID: 31784288 PMCID: PMC6952543 DOI: 10.1016/j.neuron.2019.10.012] [Citation(s) in RCA: 33] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2019] [Revised: 08/06/2019] [Accepted: 10/03/2019] [Indexed: 01/10/2023]
Abstract
The mnemonic functions of hippocampal sharp wave ripples (SPW-Rs) have been studied extensively. Because hippocampal outputs affect not only cortical but also subcortical targets, we examined the impact of SPW-Rs on the firing patterns of lateral septal (LS) neurons in behaving rats. A large fraction of SPW-Rs were temporally locked to high-frequency oscillations (HFOs) (120-180 Hz) in LS, with strongest coupling during non-rapid eye movement (NREM) sleep, followed by waking immobility. However, coherence and spike-local field potential (LFP) coupling between the two structures were low, suggesting that HFOs are generated locally within the LS GABAergic population. This hypothesis was supported by optogenetic induction of HFOs in LS. Spiking of LS neurons was largely independent of the sequential order of spiking in SPW-Rs but instead correlated with the magnitude of excitatory synchrony of the hippocampal output. Thus, LS is strongly activated by SPW-Rs and may convey hippocampal population events to its hypothalamic and brainstem targets.
Collapse
Affiliation(s)
- David Tingley
- Neuroscience Institute, New York University, New York, NY 10016, USA
| | - György Buzsáki
- Neuroscience Institute, New York University, New York, NY 10016, USA; Department of Neurology, Langone Medical Center, New York University, New York, NY 10016, USA; Center for Neural Science, New York University, New York, NY 10003, USA.
| |
Collapse
|
16
|
Eskandarian Boroujeni M, Peirouvi T, Shaerzadeh F, Ahmadiani A, Abdollahifar MA, Aliaghaei A. Differential gene expression and stereological analyses of the cerebellum following methamphetamine exposure. Addict Biol 2020; 25:e12707. [PMID: 30714656 DOI: 10.1111/adb.12707] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2018] [Revised: 11/13/2018] [Accepted: 11/21/2018] [Indexed: 12/24/2022]
Abstract
Methamphetamine (METH) is a highly addictive psychostimulant that profoundly aimed at monoaminergic systems in the brain. Despite the leading role of cerebellum in sensorimotor control as well as augmented locomotor activity under the influence of METH, there are few studies examining the effect of METH administration on gene expression profiling and structural consequences in the cerebellar region. Thus, we sought to explore the effects of METH on the cerebellum, from gene expression changes to structural alterations. In this respect, we investigated genome-wide mRNA expression using high throughput RNA-seq technology and confirmatory quantitative real-time PCR, accompanied by stereological analysis of cerebellar layers along with identification of reactive astrogliosis by glial fibrillary acidic protein and behavioral assessment following METH exposure. According to our RNA-seq data, 473 unique differentially expressed genes (DEG) were detected upon METH injections in which a large number of these genes engage basically in biological regulations and metabolic processes, chiefly located in nucleus and membrane. In addition, pathway analysis of METH-induced DEG revealed several enriched signaling cascades related largely to immune response, neurotransmission, cell growth, and death. Further, METH induced a significant reduction in volumes of cerebellar layers (molecular, granular, and Purkinje) and a decrease in the white matter volume along with a rise in astrogliosis as well as increased locomotor activity. In conclusion, considering gene expression changes combined with structural alterations of the cerebellum in response to METH, these data suggest METH-induced neurotoxicity in the cerebellar region.
Collapse
Affiliation(s)
| | - Tahmineh Peirouvi
- Department of Histology, School of MedicineUrmia University of Medical Sciences Iran
| | - Fatemeh Shaerzadeh
- Department of NeuroscienceMcKnight Brain Institute, University of Florida College of Medicine Gainesville Florida USA
| | - Abolhasan Ahmadiani
- Neurobiology Research CenterShahid Beheshti University of Medical Sciences Iran
| | - Mohammad Amin Abdollahifar
- Cell Biology and Anatomical Sciences, School of MedicineShahid Beheshti University of Medical Sciences Iran
| | - Abbas Aliaghaei
- Cell Biology and Anatomical Sciences, School of MedicineShahid Beheshti University of Medical Sciences Iran
| |
Collapse
|
17
|
Ferrucci M, Limanaqi F, Ryskalin L, Biagioni F, Busceti CL, Fornai F. The Effects of Amphetamine and Methamphetamine on the Release of Norepinephrine, Dopamine and Acetylcholine From the Brainstem Reticular Formation. Front Neuroanat 2019; 13:48. [PMID: 31133823 PMCID: PMC6524618 DOI: 10.3389/fnana.2019.00048] [Citation(s) in RCA: 50] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2018] [Accepted: 04/26/2019] [Indexed: 12/22/2022] Open
Abstract
Amphetamine (AMPH) and methamphetamine (METH) are widely abused psychostimulants, which produce a variety of psychomotor, autonomic and neurotoxic effects. The behavioral and neurotoxic effects of both compounds (from now on defined as AMPHs) stem from a fair molecular and anatomical specificity for catecholamine-containing neurons, which are placed in the brainstem reticular formation (RF). In fact, the structural cross-affinity joined with the presence of shared molecular targets between AMPHs and catecholamine provides the basis for a quite selective recruitment of brainstem catecholamine neurons following AMPHs administration. A great amount of investigations, commentary manuscripts and books reported a pivotal role of mesencephalic dopamine (DA)-containing neurons in producing behavioral and neurotoxic effects of AMPHs. Instead, the present review article focuses on catecholamine reticular neurons of the low brainstem. In fact, these nuclei add on DA mesencephalic cells to mediate the effects of AMPHs. Among these, we also include two pontine cholinergic nuclei. Finally, we discuss the conundrum of a mixed neuronal population, which extends from the pons to the periaqueductal gray (PAG). In this way, a number of reticular nuclei beyond classic DA mesencephalic cells are considered to extend the scenario underlying the neurobiology of AMPHs abuse. The mechanistic approach followed here to describe the action of AMPHs within the RF is rooted on the fine anatomy of this region of the brainstem. This is exemplified by a few medullary catecholamine neurons, which play a pivotal role compared with the bulk of peripheral sympathetic neurons in sustaining most of the cardiovascular effects induced by AMPHs.
Collapse
Affiliation(s)
- Michela Ferrucci
- Human Anatomy, Department of Translational Research and New Technologies in Medicine and Surgery, University of Pisa, Pisa, Italy
| | - Fiona Limanaqi
- Human Anatomy, Department of Translational Research and New Technologies in Medicine and Surgery, University of Pisa, Pisa, Italy
| | - Larisa Ryskalin
- Human Anatomy, Department of Translational Research and New Technologies in Medicine and Surgery, University of Pisa, Pisa, Italy
| | | | | | - Francesco Fornai
- Human Anatomy, Department of Translational Research and New Technologies in Medicine and Surgery, University of Pisa, Pisa, Italy
- IRCCS Neuromed, Pozzilli, Italy
| |
Collapse
|
18
|
Arnold MR, Williams PH, McArthur JA, Archuleta AR, O'Neill CE, Hassell JE, Smith DG, Bachtell RK, Lowry CA. Effects of chronic caffeine exposure during adolescence and subsequent acute caffeine challenge during adulthood on rat brain serotonergic systems. Neuropharmacology 2019; 148:257-271. [PMID: 30579884 PMCID: PMC6438184 DOI: 10.1016/j.neuropharm.2018.12.019] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2018] [Revised: 12/13/2018] [Accepted: 12/15/2018] [Indexed: 01/09/2023]
Abstract
Caffeine is the most commonly used drug in the world. However, animal studies suggest that chronic consumption of caffeine during adolescence can result in enhanced anxiety-like behavioral responses during adulthood. One mechanism through which chronic caffeine administration may influence subsequent anxiety-like responses is through actions on brainstem serotonergic systems. In order to explore potential effects of chronic caffeine consumption on brainstem serotonergic systems, we evaluated the effects of a 28-day exposure to chronic caffeine (0.3 g/L; postnatal day 28-56) or vehicle administration in the drinking water, followed by 24 h caffeine withdrawal, and subsequent challenge with caffeine (30 mg/kg; s.c.) or vehicle in adolescent male rats. In Experiment 1, acute caffeine challenge induced a widespread activation of serotonergic neurons throughout the dorsal raphe nucleus (DR); this effect was attenuated in rats that had been exposed to chronic caffeine consumption. In Experiment 2, acute caffeine administration profoundly decreased tph2 and slc22a3 mRNA expression throughout the DR, with no effects on htr1a or slc6a4 mRNA expression. Chronic caffeine exposure for four weeks during adolescence was sufficient to decrease tph2 mRNA expression in the DR measured 28 h after caffeine withdrawal. Chronic caffeine administration during adolescence did not impact the ability of acute caffeine to decrease tph2 or slc22a3 mRNA expression. Together, these data suggest that both chronic caffeine administration during adolescence and acute caffeine challenge during adulthood are important determinants of serotonergic function and serotonergic gene expression, effects that may contribute to chronic effects of caffeine on anxiety-like responses.
Collapse
Affiliation(s)
- M R Arnold
- Department of Integrative Physiology, University of Colorado Boulder, Boulder, CO, 80309, USA; Center for Neuroscience, University of Colorado Boulder, Boulder, CO, 80309, USA.
| | - P H Williams
- Department of Integrative Physiology, University of Colorado Boulder, Boulder, CO, 80309, USA.
| | - J A McArthur
- Department of Integrative Physiology, University of Colorado Boulder, Boulder, CO, 80309, USA.
| | - A R Archuleta
- Center for Neuroscience, University of Colorado Boulder, Boulder, CO, 80309, USA; Department of Psychology and Neuroscience, University of Colorado Boulder, Boulder, CO, 80309, USA.
| | - C E O'Neill
- Center for Neuroscience, University of Colorado Boulder, Boulder, CO, 80309, USA; Department of Psychology and Neuroscience, University of Colorado Boulder, Boulder, CO, 80309, USA.
| | - J E Hassell
- Department of Integrative Physiology, University of Colorado Boulder, Boulder, CO, 80309, USA.
| | - D G Smith
- Department of Chemistry and Biochemistry, University of Colorado Boulder, Boulder, CO, 80309, USA.
| | - R K Bachtell
- Center for Neuroscience, University of Colorado Boulder, Boulder, CO, 80309, USA; Department of Psychology and Neuroscience, University of Colorado Boulder, Boulder, CO, 80309, USA.
| | - C A Lowry
- Department of Integrative Physiology, University of Colorado Boulder, Boulder, CO, 80309, USA; Center for Neuroscience, University of Colorado Boulder, Boulder, CO, 80309, USA; Department of Physical Medicine & Rehabilitation and Center for Neuroscience, University of Colorado Anschutz Medical Campus, Aurora, CO, 80045, USA; Veterans Health Administration, Rocky Mountain Mental Illness Research Education and Clinical Center, Denver Veterans Affairs Medical Center (VAMC), Denver, CO, 80220, USA; Military and Veteran Microbiome Consortium for Research and Education (MVM-CoRE), Denver, CO, 80220, USA.
| |
Collapse
|
19
|
Klambatsen A, Nygard SK, Chang AJ, Quinones V, Jenab S. Sex differences in memory and intracellular signaling after methamphetamine binge treatment. Brain Res 2019; 1711:16-22. [PMID: 30629943 DOI: 10.1016/j.brainres.2019.01.010] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2018] [Revised: 12/21/2018] [Accepted: 01/06/2019] [Indexed: 01/21/2023]
Abstract
Methamphetamine is a neurotoxic psychostimulant known to cause cell death and terminal degradation of dopaminergic neurons in the striatum concomitant with memory deficits. However, most of the research studies have not examined the influence of sex on these changes. In this study we compared the effects of a binge regimen of methamphetamine (four injections of 4 mg/kg) on male, female, and ovariectomized (OVX) female Sprague-Dawley rats. We show that male and OVX female animals had a deficit in a novel object recognition task, while intact females did not show this deficit. Neurochemical analysis of the same animals indicated higher levels of FosB protein in caudate-putamen (CPu) and nucleus accumbens (NAc) of the male animals than intact or OVX females. Methamphetamine also increased Bcl-2 protein levels in CPu of all the cohorts. We did not find a significant effect of methamphetamine on the dopamine neuron markers tyrosine hydroxylase (TH) or dopamine transporter (DAT) 7 days after methamphetamine administrations. Our behavioral and neurochemical studies indicate that methamphetamine differentially affects male and female animals and shows sex differences in memory and molecular mechanisms in the striatum of these animals.
Collapse
Affiliation(s)
- Anthony Klambatsen
- Department of Psychology, Hunter College, CUNY, 695 Park Avenue, New York, NY 10065, USA; Behavioral and Cognitive Neuroscience Subprogram, Graduate School and University Center, CUNY, 365 Fifth Avenue, New York, NY 10016, USA.
| | - Stephanie K Nygard
- Department of Psychology, Hunter College, CUNY, 695 Park Avenue, New York, NY 10065, USA; Behavioral and Cognitive Neuroscience Subprogram, Graduate School and University Center, CUNY, 365 Fifth Avenue, New York, NY 10016, USA
| | - Anna J Chang
- Department of Psychology, Hunter College, CUNY, 695 Park Avenue, New York, NY 10065, USA; Center for Neural Science, New York University, 4 Washington Place, Room 809, New York, NY 10003, USA.
| | - Vanya Quinones
- Department of Psychology, Hunter College, CUNY, 695 Park Avenue, New York, NY 10065, USA; Behavioral and Cognitive Neuroscience Subprogram, Graduate School and University Center, CUNY, 365 Fifth Avenue, New York, NY 10016, USA.
| | - Shirzad Jenab
- Department of Psychology, Hunter College, CUNY, 695 Park Avenue, New York, NY 10065, USA; Behavioral and Cognitive Neuroscience Subprogram, Graduate School and University Center, CUNY, 365 Fifth Avenue, New York, NY 10016, USA.
| |
Collapse
|
20
|
Epigenetic Effects Induced by Methamphetamine and Methamphetamine-Dependent Oxidative Stress. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2018; 2018:4982453. [PMID: 30140365 PMCID: PMC6081569 DOI: 10.1155/2018/4982453] [Citation(s) in RCA: 47] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/19/2018] [Accepted: 06/10/2018] [Indexed: 12/21/2022]
Abstract
Methamphetamine is a widely abused drug, which possesses neurotoxic activity and powerful addictive effects. Understanding methamphetamine toxicity is key beyond the field of drug abuse since it allows getting an insight into the molecular mechanisms which operate in a variety of neuropsychiatric disorders. In fact, key alterations produced by methamphetamine involve dopamine neurotransmission in a way, which is reminiscent of spontaneous neurodegeneration and psychiatric schizophrenia. Thus, understanding the molecular mechanisms operated by methamphetamine represents a wide window to understand both the addicted brain and a variety of neuropsychiatric disorders. This overlapping, which is already present when looking at the molecular and cellular events promoted immediately after methamphetamine intake, becomes impressive when plastic changes induced in the brain of methamphetamine-addicted patients are considered. Thus, the present manuscript is an attempt to encompass all the molecular events starting at the presynaptic dopamine terminals to reach the nucleus of postsynaptic neurons to explain how specific neurotransmitters and signaling cascades produce persistent genetic modifications, which shift neuronal phenotype and induce behavioral alterations. A special emphasis is posed on disclosing those early and delayed molecular events, which translate an altered neurotransmitter function into epigenetic events, which are derived from the translation of postsynaptic noncanonical signaling into altered gene regulation. All epigenetic effects are considered in light of their persistent changes induced in the postsynaptic neurons including sensitization and desensitization, priming, and shift of neuronal phenotype.
Collapse
|
21
|
Fakhoury M. The tail of the ventral tegmental area in behavioral processes and in the effect of psychostimulants and drugs of abuse. Prog Neuropsychopharmacol Biol Psychiatry 2018; 84:30-38. [PMID: 29421265 DOI: 10.1016/j.pnpbp.2018.02.002] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/18/2017] [Revised: 02/02/2018] [Accepted: 02/03/2018] [Indexed: 02/07/2023]
Abstract
The tail of the ventral tegmental area (tVTA) is a recently identified structure that exerts a major inhibitory drive onto midbrain dopamine (DA) neurons. Also referred to as the rostromedial tegmental nucleus (RMTg), the tVTA is a cluster of gamma-aminobutyric acid (GABA)ergic neurons that starts within the posterior end of the VTA, where it is restricted dorsolateral to the caudal part of the interpeduncular nucleus, and extends into the pons. First identified in the rat, the tVTA has been described in many species, including mice and monkeys, as a region exhibiting similar anatomical and behavioral properties; it receives strong excitatory inputs from the lateral habenula (LHb), conveys negative reward-related information, and inhibits midbrain DA neuron activity. As an important inhibitory afferent to midbrain DA neurons, the tVTA is also implicated in drug abuse and in the complex interplay between reward and aversion processes. The overarching goal of this review is to provide the current state of knowledge on the anatomy and connectivity of the tVTA and to discuss recent evidence implicating this structure in reward-related processes and in the effect of psychostimulants and drugs of abuse.
Collapse
Affiliation(s)
- Marc Fakhoury
- Department of Neurosciences, Faculty of Medicine, Université de Montréal, Montreal, Quebec, Canada.
| |
Collapse
|
22
|
Jayanthi S, Gonzalez B, McCoy MT, Ladenheim B, Bisagno V, Cadet JL. Methamphetamine Induces TET1- and TET3-Dependent DNA Hydroxymethylation of Crh and Avp Genes in the Rat Nucleus Accumbens. Mol Neurobiol 2018; 55:5154-5166. [PMID: 28842817 PMCID: PMC5948251 DOI: 10.1007/s12035-017-0750-9] [Citation(s) in RCA: 32] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2017] [Accepted: 08/15/2017] [Indexed: 12/30/2022]
Abstract
Methamphetamine (METH) addiction is a biopsychosocial disorder that is accompanied by multiple relapses even after prolonged abstinence, suggesting the possibilities of long-lasting maladaptive epigenetic changes in the brain. Here, we show that METH administration produced time-dependent increases in the expression of corticotropin-releasing hormone (Crh/Crf), arginine vasopressin (Avp), and cocaine- and amphetamine-regulated transcript prepropeptide (Cartpt) mRNAs in the rat nucleus accumbens (NAc). Chromatin immunoprecipitation (ChIP) assays revealed that METH increased the abundance of phosphorylated CREB (pCREB) at the promoter of Cartpt but not at Avp or Crh DNA sequences. In contrast, METH produced DNA hypomethylation at sites near the Crh transcription start site (TSS) and at intragenic Avp sequences. METH also increased DNA hydroxymethylation at the Crh TSS and at intragenic Avp sites. In addition, METH increased the protein expression of ten-eleven-translocation enzymes that catalyze DNA hydroxymethylation. Importantly, METH increased TET1 binding at the Crh promoter and increased TET3 binding at Avp intragenic regions. We further tested the role of TET enzymes in METH-induced changes in gene expression by using the TET inhibitor, 1,5-isoquinolinediol (IQD), and found that IQD blocked METH-induced increases in Crh and Avp mRNA expression. Together, these results indicate that METH produced changes in neuropeptide transcription by both activation of the cAMP/CREB pathway and stimulation of TET-dependent DNA hydroxymethylation. These results provide molecular evidence for epigenetic controls of METH-induced changes in the expression of neuropeptides.
Collapse
Affiliation(s)
- Subramaniam Jayanthi
- Molecular Neuropsychiatry Research Branch, NIH/NIDA Intramural Research Program, National Institutes of Health, 251 Bayview Boulevard, Baltimore, MD, USA
| | - Betina Gonzalez
- Instituto de Investigaciones Farmacológicas (Universidad de Buenos Aires-Consejo Nacional de Investigaciones Científicas y Técnicas), Ciudad Autónoma de Buenos Aires, Buenos Aires, Argentina
| | - Michael T McCoy
- Molecular Neuropsychiatry Research Branch, NIH/NIDA Intramural Research Program, National Institutes of Health, 251 Bayview Boulevard, Baltimore, MD, USA
| | - Bruce Ladenheim
- Molecular Neuropsychiatry Research Branch, NIH/NIDA Intramural Research Program, National Institutes of Health, 251 Bayview Boulevard, Baltimore, MD, USA
| | - Veronica Bisagno
- Instituto de Investigaciones Farmacológicas (Universidad de Buenos Aires-Consejo Nacional de Investigaciones Científicas y Técnicas), Ciudad Autónoma de Buenos Aires, Buenos Aires, Argentina
| | - Jean Lud Cadet
- Molecular Neuropsychiatry Research Branch, NIH/NIDA Intramural Research Program, National Institutes of Health, 251 Bayview Boulevard, Baltimore, MD, USA.
- Molecular Neuropsychiatry Research Branch, DHHS/NIH/NIDA IRP, 251 Bayview Boulevard, Baltimore, MD, 21224, USA.
| |
Collapse
|
23
|
Jacobskind JS, Rosinger ZJ, Gonzalez T, Zuloaga KL, Zuloaga DG. Chronic Methamphetamine Exposure Attenuates Neural Activation in Hypothalamic-Pituitary-Adrenal Axis-Associated Brain Regions in a Sex-specific Manner. Neuroscience 2018; 380:132-145. [PMID: 29679646 DOI: 10.1016/j.neuroscience.2018.04.010] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2018] [Revised: 03/16/2018] [Accepted: 04/09/2018] [Indexed: 12/15/2022]
Abstract
Sex differences in methamphetamine (MA) abuse and consequences of MA have been reported with females showing an increased addiction phenotype and withdrawal symptoms. One mechanism through which these effects might occur is via sex-specific alterations in the hypothalamic-pituitary-adrenal (HPA) axis and its associated brain regions. In this study, mice were administered MA (5 mg/kg) or saline for 10 consecutive days. During early withdrawal, anxiety-like behaviors were assessed in the open field, light/dark box, and elevated plus maze. At ten days of withdrawal, mice were injected with a final dose of MA (5 mg/kg) or saline. Chronic MA did not alter anxiety-like behaviors or corticosterone responses to a final dose of MA, although females showed elevated corticosterone responses compared to males. Chronic MA attenuated final MA-induced c-Fos in both sexes in the paraventricular hypothalamus (PVH), bed nucleus of the stria terminalis (BNST), cingulate cortex, central and basolateral amygdala. In CA1 and CA3 hippocampal areas, c-Fos attenuation by chronic MA occurred only in females. Within the PVH, final MA injection increased c-Fos to a greater extent in females compared to males regardless of prior MA exposure. Dual-labeling of c-Fos with glucocorticoid receptor revealed a specific attenuation of neural activation within this cell type in the PVH, central and basolateral amygdala, and BNST. Together these findings demonstrate that chronic MA can suppress subsequent activation of HPA axis-associated brain regions and cell phenotypes. Further, in select regions this reduction is sex-specific. These changes may contribute to reported sex differences in MA abuse patterns.
Collapse
Affiliation(s)
- Jason S Jacobskind
- University at Albany, Department of Psychology, Albany, NY 12222, United States
| | - Zachary J Rosinger
- University at Albany, Department of Psychology, Albany, NY 12222, United States
| | - Tiffany Gonzalez
- University at Albany, Department of Psychology, Albany, NY 12222, United States
| | - Kristen L Zuloaga
- Albany Medical College, Department of Neuroscience & Experimental Therapeutics, Albany, NY 12208, United States
| | - Damian G Zuloaga
- University at Albany, Department of Psychology, Albany, NY 12222, United States.
| |
Collapse
|
24
|
Brain Activity during Methamphetamine Anticipation in a Non-Invasive Self-Administration Paradigm in Mice. eNeuro 2018; 5:eN-NWR-0433-17. [PMID: 29632871 PMCID: PMC5889482 DOI: 10.1523/eneuro.0433-17.2018] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2017] [Revised: 02/14/2018] [Accepted: 02/21/2018] [Indexed: 12/13/2022] Open
Abstract
The ability to sense time and anticipate events is critical for survival. Learned responses that allow anticipation of the availability of food or water have been intensively studied. While anticipatory behaviors also occur prior to availability of regularly available rewards, there has been relatively little work on anticipation of drugs of abuse, specifically methamphetamine (MA). In the present study, we used a protocol that avoided possible CNS effects of stresses of handling or surgery by testing anticipation of MA availability in animals living in their home cages, with daily voluntary access to the drug at a fixed time of day. Anticipation was operationalized as the amount of wheel running prior to MA availability. Mice were divided into four groups given access to either nebulized MA or water, in early or late day. Animals with access to MA, but not water controls, showed anticipatory activity, with more anticipation in early compared to late day and significant interaction effects. Next, we explored the neural basis of the MA anticipation, using c-FOS expression, in animals euthanized at the usual time of nebulization access. In the dorsomedial hypothalamus (DMH) and orbitofrontal cortex (OFC), the pattern of c-FOS expression paralleled that of anticipatory behavior, with significant main and interaction effects of treatment and time of day. The results for the lateral septum (LS) were significant for main effects and marginally significant for interaction effects. These studies suggest that anticipation of MA is associated with activation of brain regions important in circadian timing, emotional regulation, and decision making.
Collapse
|
25
|
McGlinchey EM, Aston-Jones G. Dorsal Hippocampus Drives Context-Induced Cocaine Seeking via Inputs to Lateral Septum. Neuropsychopharmacology 2018; 43:987-1000. [PMID: 28695893 PMCID: PMC5854789 DOI: 10.1038/npp.2017.144] [Citation(s) in RCA: 68] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/18/2016] [Revised: 06/03/2017] [Accepted: 07/03/2017] [Indexed: 01/07/2023]
Abstract
Lateral septum (LS) has re-emerged as an important structure in reward and addiction; however, LS afferents that drive addiction behaviors are unknown. Here, we used a modified self-administration/reinstatement procedure combined with anatomical, pharmacological, and chemogenetic techniques to characterize LS, and hippocampal inputs to LS, in two established triggers of drug relapse-context- and cue-induced reinstatement of cocaine seeking. We found that inactivation of LS neurons attenuated both context- and cue-induced reinstatement of cocaine seeking. However, dorsal hippocampus inputs to LS showed enhanced neuronal activation (as measured by Fos expression) during context-induced, but not cue-induced reinstatement. Additionally, chemogenetic inhibition of dorsal, but not ventral, hippocampal inputs to LS specifically attenuated context-induced reinstatement. Together these findings elucidate the importance of LS in reinstatement of cocaine seeking, and indicate that dorsal hippocampal inputs to LS mediate context-, but not cue-induced, reinstatement of cocaine seeking.
Collapse
Affiliation(s)
- Ellen M McGlinchey
- Department of Neurosciences, Medical University of South Carolina, Charleston, SC, USA,Brain Health Institute, Rutgers University and Rutgers Biomedical and Health Sciences, Piscataway, NJ, USA
| | - Gary Aston-Jones
- Brain Health Institute, Rutgers University and Rutgers Biomedical and Health Sciences, Piscataway, NJ, USA,Brain Health Institute, Rutgers University and Rutgers Behavioral and Health Sciences, 683 Hoes Lane West, SPH Suite 259, Piscataway, NJ 08854, USA, Tel: +1 732 235 6077, Fax: +1 732 235 5814, E-mail:
| |
Collapse
|
26
|
Methamphetamine binge administration during late adolescence induced enduring hippocampal cell damage following prolonged withdrawal in rats. Neurotoxicology 2018; 66:1-9. [PMID: 29501631 DOI: 10.1016/j.neuro.2018.02.016] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2017] [Revised: 01/22/2018] [Accepted: 02/26/2018] [Indexed: 11/20/2022]
Abstract
A recent study from our laboratory demonstrated that binge methamphetamine induced hippocampal cell damage (i.e., impaired cell genesis) in rats when administered specifically during late adolescence (postnatal day, PND 54-57) and evaluated 24 h later (PND 58). The results also suggested a possible role for brain-derived neurotrophic factor (BDNF) regulating cell genesis and survival. This subsequent study evaluated whether these effects persisted in time as measured following prolonged withdrawal. Male Sprague-Dawley rats were treated (i.p.) with BrdU (2 × 50 mg/kg, 3 days, PND 48-50) followed by a binge paradigm (3 pulses/day, every 3 h, 4 days, PND 54-57) of methamphetamine (5 mg/kg, n = 14, M) or saline (0.9% NaCl, 1 ml/kg, n = 12, C). Following 34 days of forced withdrawal (PND 91), rats were killed 45 min after a challenge dose of saline (Sal: C-Sal, n = 6; M-Sal, n = 7) or methamphetamine (Meth: C-Meth, n = 6; M-Meth, n = 7). Neurogenesis markers (Ki-67: cell proliferation; NeuroD: early neuronal survival; BrdU: prolonged cell survival, 41-43 days old cells) were evaluated by immunohistochemistry while neuroplasticity markers (BDNF and Fos forms) were evaluated by Western blot. The main results showed that a history of methamphetamine administration (PND 54-57) induced enduring hippocampal cell damage (i.e., observed on PND 91) by decreasing cell survival (BrdU + cells) and mature-BDNF (m-BDNF) protein content, associated with neuronal survival, growth and differentiation. Interestingly, m-BDNF regulation paralleled hippocampal c-Fos protein content, indicating decreased neuronal activity, and thus reinforcing the persisting negative effects induced by methamphetamine in rat hippocampus following prolonged withdrawal.
Collapse
|
27
|
Jacobskind JS, Rosinger ZJ, Zuloaga DG. Hypothalamic-pituitary-adrenal axis responsiveness to methamphetamine is modulated by gonadectomy in males. Brain Res 2017; 1677:74-85. [PMID: 28941573 DOI: 10.1016/j.brainres.2017.09.020] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2017] [Revised: 08/18/2017] [Accepted: 09/17/2017] [Indexed: 12/14/2022]
Abstract
Sex differences in patterns of methamphetamine (MA) abuse have been reported with females (humans and rodents) showing an elevated addiction phenotype. Previous findings indicate MA-induced hypothalamic-pituitary-adrenal (HPA) axis activation is also sexually dimorphic with females exhibiting an elevated glucocorticoid release and differential neural activation patterns within HPA axis-associated brain regions. These effects may contribute to sex differences in abuse. To determine the role of gonadal hormones in mediating sex differences in MA-induced glucocorticoids, male and female C57BL/6J mice were gonadectomized or sham-operated, and following recovery, injected with MA (5mg/kg) and sacrificed 60min or 120min later. Blood was collected for corticosterone radioimmunoassay, and brains were used to assess c-Fos, and c-Fos co-localization with glucocorticoid receptor (GR). At 120min after MA injection, corticosterone levels were elevated in females compared to males and gonadectomy in males increased corticosterone to female levels. C-Fos was greater in females than males in the medial preoptic area, bed nucleus of the stria terminalis, basolateral amygdala, and central amygdala. Female gonadectomy had little effect on either corticosterone or c-Fos, while male gonadectomy elevated c-Fos in the central amygdala. Relative to sham males, gonadectomized males also showed decreased c-Fos/GR cell number in the CA3 hippocampal area compared to sham males, indicating a central site for attenuated negative feedback. Together, these findings indicate that androgens regulate MA-induced activation of the HPA axis, potentially by enhancing negative feedback. These sex and gonadal hormone effects on the HPA axis may contribute to sex differences in MA abuse patterns.
Collapse
Affiliation(s)
- Jason S Jacobskind
- University at Albany, Department of Psychology, Albany, NY 12222, United States
| | - Zachary J Rosinger
- University at Albany, Department of Psychology, Albany, NY 12222, United States
| | - Damian G Zuloaga
- University at Albany, Department of Psychology, Albany, NY 12222, United States.
| |
Collapse
|
28
|
Sánchez-Catalán MJ, Faivre F, Yalcin I, Muller MA, Massotte D, Majchrzak M, Barrot M. Response of the Tail of the Ventral Tegmental Area to Aversive Stimuli. Neuropsychopharmacology 2017; 42:638-648. [PMID: 27468916 PMCID: PMC5240171 DOI: 10.1038/npp.2016.139] [Citation(s) in RCA: 37] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/22/2016] [Revised: 07/19/2016] [Accepted: 07/22/2016] [Indexed: 12/26/2022]
Abstract
The GABAergic tail of the ventral tegmental area (tVTA), also named rostromedial tegmental nucleus (RMTg), exerts an inhibitory control on dopamine neurons of the VTA and substantia nigra. The tVTA has been implicated in avoidance behaviors, response to drugs of abuse, reward prediction error, and motor functions. Stimulation of the lateral habenula (LHb) inputs to the tVTA, or of the tVTA itself, induces avoidance behaviors, which suggests a role of the tVTA in processing aversive information. Our aim was to test the impact of aversive stimuli on the molecular recruitment of the tVTA, and the behavioral consequences of tVTA lesions. In rats, we assessed Fos response to lithium chloride (LiCl), β-carboline, naloxone, lipopolysaccharide (LPS), inflammatory pain, neuropathic pain, foot-shock, restraint stress, forced swimming, predator odor, and opiate withdrawal. We also determined the effect of tVTA bilateral ablation on physical signs of opiate withdrawal, and on LPS- and LiCl-induced conditioned taste aversion (CTA). Naloxone-precipitated opiate withdrawal induced Fos in μ-opioid receptor-positive (15%) and -negative (85%) tVTA cells, suggesting the presence of both direct and indirect mechanisms in tVTA recruitment during withdrawal. However, tVTA lesion did not impact physical signs of opiate withdrawal. Fos induction was also present with repeated, but not single, foot-shock delivery. However, such induction was mostly absent with other aversive stimuli. Moreover, tVTA ablation had no impact on CTA. Although stimulation of the tVTA favors avoidance behaviors, present findings suggest that this structure may be important to the response to some, but not all, aversive stimuli.
Collapse
Affiliation(s)
- María-José Sánchez-Catalán
- Institut des Neurosciences Cellulaires et Intégratives, Centre National de la Recherche Scientifique, Strasbourg, France,Unitat Predepartamental de Medicina, Universitat Jaume I, Castelló de la Plana, Spain,Unitat Predepartamental de Medicina, Universitat Jaume I, Avenue Vicent Sos Baynat, s/n, 13071 Castelló de la Plana, Spain, Tel: +34 964 38 74 40, Fax: +34 964 72 90 16, E-mail:
| | - Fanny Faivre
- Institut des Neurosciences Cellulaires et Intégratives, Centre National de la Recherche Scientifique, Strasbourg, France,Université de Strasbourg, Strasbourg, France
| | - Ipek Yalcin
- Institut des Neurosciences Cellulaires et Intégratives, Centre National de la Recherche Scientifique, Strasbourg, France
| | - Marc-Antoine Muller
- Université de Strasbourg, Strasbourg, France,Laboratoire de Neurosciences Cognitives et Adaptatives, Faculté de Psychologie, Centre National de la Recherche Scientifique, Strasbourg, France
| | - Dominique Massotte
- Institut des Neurosciences Cellulaires et Intégratives, Centre National de la Recherche Scientifique, Strasbourg, France
| | - Monique Majchrzak
- Université de Strasbourg, Strasbourg, France,Laboratoire de Neurosciences Cognitives et Adaptatives, Faculté de Psychologie, Centre National de la Recherche Scientifique, Strasbourg, France
| | - Michel Barrot
- Institut des Neurosciences Cellulaires et Intégratives, Centre National de la Recherche Scientifique, Strasbourg, France
| |
Collapse
|
29
|
Methamphetamine and Ovarian Steroid Responsive Cells in the Posteriodorsal Medial Amygdala are Required for Methamphetamine-enhanced Proceptive Behaviors. Sci Rep 2017; 7:39817. [PMID: 28045134 PMCID: PMC5206624 DOI: 10.1038/srep39817] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2016] [Accepted: 11/28/2016] [Indexed: 11/28/2022] Open
Abstract
Methamphetamine (Meth) is a psychomotor stimulant strongly associated with increases in sexual drive and impulse in both men and women. These changes in sexual motivation have a greater impact on women due to their likelihood of facing the greater burden of unplanned pregnancies, as well as increased risk for psychiatric co-morbidities such as depression. We have previously established a rodent model of Meth-induced increases in sexual motivation. Using this model, we have identified the posteriodorsal medial amygdala (MePD) via excitotoxic lesion studies as a necessary nucleus in Meth-facilitated female sexual motivation. While lesion studies give us insight into key nuclei that may be targets of Meth action, such an approach does not give insight into the identity of the specific MePD neurons or neural circuitry involved in Meth-induced increases in proceptive behaviors. Using the DAUN02 inactivation method, a recently established technique for removing behaviorally relevant cell populations, we present evidence that the ovarian steroid/Meth responsive cells in the MePD are necessary for Meth-induced facilitation of proceptive behaviors. These findings form the basis for future work that will allow for the classification of neuronal subtypes involved in the MePD’s modulation of proceptive behavior as well as a stronger understanding of the neurocircuitry of female sexual motivation.
Collapse
|
30
|
James MH, Mahler SV, Moorman DE, Aston-Jones G. A Decade of Orexin/Hypocretin and Addiction: Where Are We Now? Curr Top Behav Neurosci 2017; 33:247-281. [PMID: 28012090 PMCID: PMC5799809 DOI: 10.1007/7854_2016_57] [Citation(s) in RCA: 126] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
One decade ago, our laboratory provided the first direct evidence linking orexin/hypocretin signaling with drug seeking by showing that activation of these neurons promotes conditioned morphine-seeking behavior. In the years since, contributions from many investigators have revealed roles for orexins in addiction for all drugs of abuse tested, but only under select circumstances. We recently proposed that orexins play a fundamentally unified role in coordinating "motivational activation" under numerous behavioral conditions, and here we unpack this hypothesis as it applies to drug addiction. We describe evidence collected over the past 10 years that elaborates the role of orexin in drug seeking under circumstances where high levels of effort are required to obtain the drug, or when motivation for drug reward is augmented by the presence of external stimuli like drug-associated cues/contexts or stressors. Evidence from studies using traditional self-administration and reinstatement models, as well as behavioral economic analyses of drug demand elasticity, clearly delineates a role for orexin in modulating motivational, rather than the primary reinforcing aspects of drug reward. We also discuss the anatomical interconnectedness of the orexin system with wider motivation and reward circuits, with a particular focus on how orexin modulates prefrontal and other glutamatergic inputs onto ventral tegmental area dopamine neurons. Last, we look ahead to the next decade of the research in this area, highlighting the recent FDA approval of the dual orexin receptor antagonist suvorexant (Belsomra®) for the treatment of insomnia as a promising sign of the potential clinical utility of orexin-based therapies for the treatment of addiction.
Collapse
Affiliation(s)
- Morgan H James
- Brain Health Institute, Rutgers University/Rutgers Biomedical and Health Sciences, Piscataway, NJ, 08854, USA
- Florey Institute of Neuroscience and Mental Health, University of Melbourne, Parkville, VIC, 2337, Australia
| | - Stephen V Mahler
- Department of Neurobiology and Behavior, University of California, Irvine, Irvine, CA, 92967, USA
| | - David E Moorman
- Department of Psychological and Brain Sciences & Neuroscience and Behavior Graduate Program, University of Massachusetts Amherst, Amherst, MA, 01003, USA
| | - Gary Aston-Jones
- Brain Health Institute, Rutgers University/Rutgers Biomedical and Health Sciences, Piscataway, NJ, 08854, USA.
| |
Collapse
|
31
|
Li L, Qiao C, Chen G, Qian H, Hou Y, Li T, Liu X. Ifenprodil attenuates the acquisition and expression of methamphetamine-induced behavioral sensitization and activation of Ras-ERK1/2 cascade in the caudate putamen. Neuroscience 2016; 335:20-9. [DOI: 10.1016/j.neuroscience.2016.08.022] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2016] [Revised: 08/08/2016] [Accepted: 08/09/2016] [Indexed: 02/06/2023]
|
32
|
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: 76] [Impact Index Per Article: 9.5] [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
|
33
|
Abstract
Amphetamine and methamphetamine addiction is described by specific behavioral alterations, suggesting long-lasting changes in gene and protein expression within specific brain subregions involved in the reward circuitry. Given the persistence of the addiction phenotype at both behavioral and transcriptional levels, several studies have been conducted to elucidate the epigenetic landscape associated with persistent effects of drug use on the mammalian brain. This review discusses recent advances in our comprehension of epigenetic mechanisms underlying amphetamine- or methamphetamine-induced behavioral, transcriptional, and synaptic plasticity. Accumulating evidence demonstrated that drug exposure induces major epigenetic modifications-histone acetylation and methylation, DNA methylation-in a very complex manner. In rare instances, however, the regulation of a specific target gene can be correlated to both epigenetic alterations and behavioral abnormalities. Work is now needed to clarify and validate an epigenetic model of addiction to amphetamines. Investigations that include genome-wide approaches will accelerate the speed of discovery in the field of addiction.
Collapse
Key Words
- AMPA, α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid
- AMPH, amphetamine
- AP1, activator protein 1
- ATF2, activating transcription factor 2
- BASP1, brain abundant signal protein 1
- BDNF, brain derived neurotrophic factor
- CCR2, C‒C chemokine receptor 2
- CPP, conditioned place preference
- CREB, cAMP response element binding protein
- ChIP, chromatin immunoprecipitation
- CoREST, restrictive element 1 silencing transcription factor corepressor
- Cp60, compound 60
- DNA methylation
- DNMT, DNA methyltransferase
- FOS, Finkel–Biskis–Jinkins murine osteosarcoma viral oncogene
- GABA, γ-aminobutyric acid
- GLUA1, glutamate receptor subunit A1
- GLUA2, glutamate receptor subunit A2
- GLUN1, glutamate receptor subunit N1
- H2Bac, pan-acetylation of histone 2B
- H3, histone 3
- H3K14Ac, acetylation of histone 3 at lysine 14
- H3K18, lysine 18 of histone 3
- H3K4, lysine 4 of histone 3
- H3K4me3, trimethylation of histone 3 at lysine 4
- H3K9, lysine 9 of histone 3
- H3K9Ac, acetylation of histone 3 at lysine 9
- H3K9me3, trimethylation of histone 3 at lysine 9
- H4, histone 4
- H4Ac, pan-acetylation of histone 4
- H4K12Ac, acetylation of histone 4 at lysine 12
- H4K16, lysine 16 of histone 4
- H4K5, lysine 5 of histone 4
- H4K8, lysine 8 of histone 4
- HAT, histone acetyltransferase
- HDAC, histone deacetylase
- HDM, histone demethylase
- HMT, histone methyltransferase
- IP, intra-peritoneal
- JUN, jun proto-oncogene
- KDM, lysine demethylase
- KLF10, Kruppel-like factor 10
- KMT, lysine methyltransferase
- METH, methamphetamine
- MeCP2, methyl-CpG binding protein 2
- NAc, nucleus accumbens
- NMDA, N-methyl-D-aspartate
- NaB, sodium butyrate
- OfC, orbitofrontal cortex
- PfC, prefrontal cortex
- REST, restrictive element 1 silencing transcription factor
- RNAi, RNA interference
- Ser241, serine 241
- Sin3A, SIN3 transcription regulator family member A
- TSS, transcription start site
- VPA, valproic acid
- WT1, Wilms tumor protein 1.
- amphetamine
- histone acetylation
- histone methylation
- methamphetamine
- siRNA, silencing RNA
Collapse
Affiliation(s)
- Arthur Godino
- a Département de Biologie; École Normale Supérieure de Lyon ; Lyon , France
| | | | | |
Collapse
|
34
|
Zuloaga DG, Johnson LA, Weber S, Raber J. Immediate and lasting effects of chronic daily methamphetamine exposure on activation of cells in hypothalamic-pituitary-adrenal axis-associated brain regions. Psychopharmacology (Berl) 2016; 233:381-92. [PMID: 26525566 PMCID: PMC4815259 DOI: 10.1007/s00213-015-4114-8] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/04/2015] [Accepted: 10/11/2015] [Indexed: 10/22/2022]
Abstract
RATIONALE Chronic methamphetamine (MA) abuse leads to dependence and symptoms of withdrawal after use has ceased. Negative mood states associated with withdrawal, as well as drug reinstatement, have been linked to drug-induced disruption of the hypothalamic-pituitary-adrenal (HPA) axis. However, effects of chronic MA exposure or acute MA exposure following withdrawal on neural activation patterns within brain regions that regulate the HPA axis are unknown. OBJECTIVES In this study, neural activation patterns were assessed by quantification of c-Fos protein in mice exposed to different regimens of MA administration. METHODS (Experiment 1) Adult male mice were treated with MA (5 mg/kg) or saline once or once daily for 10 days. (Experiment 2) Mice were treated with MA or saline once daily for 10 days and following a 10-day withdrawal period were re-administered a final dose of MA or saline. c-Fos was quantified in brains after the final injection. RESULTS (Experiment 1) Compared to exposure to a single dose of MA (5 mg/kg), chronic MA exposure decreased the number of c-Fos expressing cells in the paraventricular hypothalamus, dorsomedial hypothalamus, central amygdala, basolateral amygdala, bed nucleus of the stria terminalis (BNST), and CA3 hippocampal region. (Experiment 2) Compared to mice receiving their first dose of MA, mice chronically treated with MA, withdrawn, and re-administered MA, showed decreased c-Fos expressing cells within the central and basolateral amygdala, BNST, and CA3. CONCLUSIONS HPA axis-associated amygdala, extended amygdala, and hippocampal regions endure lasting effects following chronic MA exposure and therefore may be linked to stress-related withdrawal symptoms.
Collapse
Affiliation(s)
- Damian G. Zuloaga
- Department of Behavioral Neuroscience, Oregon Health and Science University Portland, Portland, OR 97239, USA,Department of Psychology, University at Albany, Albany, NY 12222, USA
| | - Lance A. Johnson
- Department of Behavioral Neuroscience, Oregon Health and Science University Portland, Portland, OR 97239, USA
| | - Sydney Weber
- Department of Behavioral Neuroscience, Oregon Health and Science University Portland, Portland, OR 97239, USA
| | - Jacob Raber
- Department of Behavioral Neuroscience, Oregon Health and Science University Portland, Portland, OR 97239, USA,Department of Neurology, Oregon Health and Science University Portland, Portland, OR 97239, USA,Department of Radiation Medicine, Oregon Health and Science University Portland, Portland, OR 97239, USA,Division of Neuroscience, ONPRC, Oregon Health and Science University Portland, Portland, OR 97239, USA
| |
Collapse
|
35
|
Zuloaga DG, Jacobskind JS, Jacosbskind JS, Raber J. Methamphetamine and the hypothalamic-pituitary-adrenal axis. Front Neurosci 2015; 9:178. [PMID: 26074755 PMCID: PMC4444766 DOI: 10.3389/fnins.2015.00178] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2015] [Accepted: 05/03/2015] [Indexed: 01/22/2023] Open
Abstract
Psychostimulants such as methamphetamine (MA) induce significant alterations in the function of the hypothalamic-pituitary-adrenal (HPA) axis. These changes in HPA axis function are associated with altered stress-related behaviors and might contribute to addictive processes such as relapse. In this mini-review we discuss acute and chronic effects of MA (adult and developmental exposure) on the HPA axis, including effects on HPA axis associated genes/proteins, brain regions, and behaviors such as anxiety and depression. A better understanding of the mechanisms through which MA affects the HPA axis may lead to more effective treatment strategies for MA addiction.
Collapse
Affiliation(s)
| | | | | | - Jacob Raber
- Departments of Behavioral Neuroscience, Neurology, and Radiation Medicine, Oregon Health and Science University Portland Portland, OR, USA ; Division of Neuroscience, Oregon National Primate Research Center, Oregon Health and Science University Portland Portland, OR, USA
| |
Collapse
|
36
|
Shen WW, Zhang YS, Li LH, Liu Y, Huang XN, Chen LH, Zhou W. Long-term use of methamphetamine disrupts the menstrual cycles and hypothalamic-pituitary-ovarian axis. J Addict Med 2015; 8:183-8. [PMID: 24695019 DOI: 10.1097/adm.0000000000000021] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
OBJECTIVES Long-term use of methamphetamine (MA) induces the neuron damage and leads to multiple neuropsychiatric disorders. However, the effect of MA on the female reproductive functions has not yet been evaluated. The objective of this study was to determine the prevalence of abnormal menstrual cycling in female MA users. METHODS Female MA users (N = 113) were recruited from the Zhangjiang Isolated Compulsory Detoxification Center. Gynecologic history and drug use history were recorded, and serum levels of follicle-stimulating hormone, luteinizing hormone, prolactin, estrogen, progesterone, and testosterone were measured. RESULTS Long-term use of MA significantly altered the menstrual cycle, and 33.6% women suffered from abnormal uterine bleeding while using MA. Deregulation of sex hormones was observed in 73.3% of participants during abstinence. The most common patterns were simple anovular menstruation, which was caused mainly by a hypothalamic deregulation and pituitary suppression with or without ovarian suppression. Normal hormone levels were observed more frequently in participants abstinent for more than 10 months (39.5%) than in participants who were abstinent for less than 10 months (18.6%). However, no relationship was found between hormone deregulation and age or history of MA use. CONCLUSIONS The present data demonstrate that long-term use of MA results in the disruption of menstrual cycles and dysfunction of hypothalamic-pituitary-gonadal axis in women.
Collapse
Affiliation(s)
- Wen-Wen Shen
- From the Ningbo Addiction Research and Treatment Center, School of Medicine (WWS, LHL, YL, XNH, LHC, WZ), Ningbo University, Ningbo 315010, People's Republic of China; and Department of Gynecology (YSZ), the No. 1 People's Hospital of Ningbo, Ningbo 315010, People's Republic of China
| | | | | | | | | | | | | |
Collapse
|
37
|
Control of the nigrostriatal dopamine neuron activity and motor function by the tail of the ventral tegmental area. Neuropsychopharmacology 2014; 39:2788-98. [PMID: 24896615 PMCID: PMC4200489 DOI: 10.1038/npp.2014.129] [Citation(s) in RCA: 70] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/28/2014] [Revised: 05/16/2014] [Accepted: 05/29/2014] [Indexed: 01/10/2023]
Abstract
Midbrain dopamine neurons are implicated in various psychiatric and neurological disorders. The GABAergic tail of the ventral tegmental area (tVTA), also named the rostromedial tegmental nucleus (RMTg), displays dense projections to the midbrain and exerts electrophysiological control over dopamine cells of the VTA. However, the influence of the tVTA on the nigrostriatal pathway, from the substantia nigra pars compacta (SNc) to the dorsal striatum, and on related functions remains to be addressed. The present study highlights the role played by the tVTA as a GABA brake for the nigrostriatal system, demonstrating a critical influence over motor functions. Using neuroanatomical approaches with tract tracing and electron microscopy, we reveal the presence of a tVTA-SNc-dorsal striatum pathway. Using in vivo electrophysiology, we prove that the tVTA is a major inhibitory control center for SNc dopamine cells. Using behavioral approaches, we demonstrate that the tVTA controls rotation behavior, motor coordination, and motor skill learning. The motor enhancements observed after ablation of the tVTA are in this regard comparable with the performance-enhancing properties of amphetamine, a drug used in doping. These findings demonstrate that the tVTA is a major GABA brake for nigral dopamine systems and nigrostriatal functions, and they raise important questions about how the tVTA is integrated within the basal ganglia circuitry. They also warrant further research on the tVTA's role in motor and dopamine-related pathological contexts such as Parkinson's disease.
Collapse
|
38
|
Sanchez-Catalan MJ, Kaufling J, Georges F, Veinante P, Barrot M. The antero-posterior heterogeneity of the ventral tegmental area. Neuroscience 2014; 282:198-216. [PMID: 25241061 DOI: 10.1016/j.neuroscience.2014.09.025] [Citation(s) in RCA: 85] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2014] [Revised: 09/09/2014] [Accepted: 09/10/2014] [Indexed: 11/16/2022]
Abstract
The ventral tegmental area (VTA) is a brain region processing salient sensory and emotional information, controlling motivated behaviors, natural or drug-related reward, reward-related learning, mood, and participating in their associated psychopathologies. Mostly studied for its dopamine neurons, the VTA also includes functionally important GABA and glutamate cell populations. Behavioral evidence supports the presence of functional differences between the anterior VTA (aVTA) and the posterior VTA (pVTA), which is the topic of this review. This antero-posterior heterogeneity concerns locomotor activity, conditioned place preference and intracranial self-administration, and can be seen in response to ethanol, acetaldehyde, salsolinol, opioids including morphine, cholinergic agonists including nicotine, cocaine, cannabinoids and after local manipulation of GABA and serotonin receptors. It has also been observed after viral-mediated manipulation of GluR1, phospholipase Cγ (PLCγ) and cAMP response element binding protein (CREB) expression, with impact on reward and aversion-related responses, on anxiety and depression-related behaviors and on pain sensitivity. In this review, the substrates potentially underlying these aVTA/pVTA differences are discussed, including the VTA sub-nuclei and the heterogeneity in connectivity, cell types and molecular characteristics. We also review the role of the tail of the VTA (tVTA), or rostromedial tegmental nucleus (RMTg), which may also participate to the observed antero-posterior heterogeneity of the VTA. This region, partly located within the pVTA, is an inhibitory control center for dopamine activity. It controls VTA and substantia nigra dopamine cells, thus exerting a major influence on basal ganglia functions. This review highlights the need for a more comprehensive analysis of VTA heterogeneity.
Collapse
Affiliation(s)
- M J Sanchez-Catalan
- Institut des Neurosciences Cellulaires et Intégratives, Centre National de la Recherche Scientifique, Strasbourg, France; Université de Strasbourg, Strasbourg, France
| | - J Kaufling
- Centre National de la Recherche Scientifique, Interdisciplinary Institute for Neuroscience, UMR 5297, Bordeaux, France; Université de Bordeaux, Interdisciplinary Institute for Neuroscience, UMR 5297, Bordeaux, France
| | - F Georges
- Centre National de la Recherche Scientifique, Interdisciplinary Institute for Neuroscience, UMR 5297, Bordeaux, France; Université de Bordeaux, Interdisciplinary Institute for Neuroscience, UMR 5297, Bordeaux, France
| | - P Veinante
- Institut des Neurosciences Cellulaires et Intégratives, Centre National de la Recherche Scientifique, Strasbourg, France; Université de Strasbourg, Strasbourg, France
| | - M Barrot
- Institut des Neurosciences Cellulaires et Intégratives, Centre National de la Recherche Scientifique, Strasbourg, France; Université de Strasbourg, Strasbourg, France.
| |
Collapse
|
39
|
Cadet JL, Brannock C, Jayanthi S, Krasnova IN. Transcriptional and epigenetic substrates of methamphetamine addiction and withdrawal: evidence from a long-access self-administration model in the rat. Mol Neurobiol 2014; 51:696-717. [PMID: 24939695 PMCID: PMC4359351 DOI: 10.1007/s12035-014-8776-8] [Citation(s) in RCA: 59] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2014] [Accepted: 06/01/2014] [Indexed: 01/06/2023]
Abstract
Methamphetamine use disorder is a chronic neuropsychiatric disorder characterized by recurrent binge episodes, intervals of abstinence, and relapses to drug use. Humans addicted to methamphetamine experience various degrees of cognitive deficits and other neurological abnormalities that complicate their activities of daily living and their participation in treatment programs. Importantly, models of methamphetamine addiction in rodents have shown that animals will readily learn to give themselves methamphetamine. Rats also accelerate their intake over time. Microarray studies have also shown that methamphetamine taking is associated with major transcriptional changes in the striatum measured within a short or longer time after cessation of drug taking. After a 2-h withdrawal time, there was increased expression of genes that participate in transcription regulation. These included cyclic AMP response element binding (CREB), ETS domain-containing protein (ELK1), and members of the FOS family of transcription factors. Other genes of interest include brain-derived neurotrophic factor (BDNF), tyrosine kinase receptor, type 2 (TrkB), and synaptophysin. Methamphetamine-induced transcription was found to be regulated via phosphorylated CREB-dependent events. After a 30-day withdrawal from methamphetamine self-administration, however, there was mostly decreased expression of transcription factors including junD. There was also downregulation of genes whose protein products are constituents of chromatin-remodeling complexes. Altogether, these genome-wide results show that methamphetamine abuse might be associated with altered regulation of a diversity of gene networks that impact cellular and synaptic functions. These transcriptional changes might serve as triggers for the neuropsychiatric presentations of humans who abuse this drug. Better understanding of the way that gene products interact to cause methamphetamine addiction will help to develop better pharmacological treatment of methamphetamine addicts.
Collapse
Affiliation(s)
- Jean Lud Cadet
- Molecular Neuropsychiatry Research Branch, Intramural Research Program, National Institute on Drug Abuse, NIH, DHHS, 251 Bayview Boulevard, Baltimore, MD, 21224, USA,
| | | | | | | |
Collapse
|
40
|
Liu W, Peng QX, Lin XL, Luo CH, Jiang MJ, Mo ZX, Yung KKL. Effect of rhynchophylline on the expression of p-CREB and sc-Fos in triatum and hippocampal CA1 area of methamphetamine-induced conditioned place preference rats. Fitoterapia 2013; 92:16-22. [PMID: 24140441 DOI: 10.1016/j.fitote.2013.10.002] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2013] [Revised: 09/30/2013] [Accepted: 10/05/2013] [Indexed: 02/04/2023]
Abstract
To explore the effect of rhynchophylline (Rhy) on the expression of p-CREB and c-Fos in the striatum and hippocampal CA1 area of methamphetamine-induced conditioned place preference (CPP) rat, methamphetamine (2 mg/kg) was injected to rats and the conditioned place preference was observed in these rats treated with or without Rhy. An immunohistochemistry assay was used to determine the expression of p-CREB and c-Fos in the striatum and hippocampal CA1 area. Methamphetamine induced significant behavior alteration in CPP, while after pretreatment with rhynchophylline or ketamine, the time of staying in methamphetamine-paired compartment of rats was significantly reduced. Methamphetamine also increased the number of p-CREB positive cells in the striatum and hippocampal CA1 zone, as well as p-Fos positive cells. However, the compound Rhy could attenuate the effect. These findings show that Rhy can suppress the acquisition of CPP in rats induced by methamphetamine and the action may be related with the reduced expression of p-CREB and p-Fos in the striatum and hippocampus.
Collapse
Affiliation(s)
- Wei Liu
- School of Traditional Chinese Medicine, Southern Medical University, Guangzhou 510515, China
| | - Qiu-Xian Peng
- Department of Biology, Hong Kong Baptist University, Kowloon Tong, Hong Kong
| | - Xiao-Liang Lin
- School of Traditional Chinese Medicine, Southern Medical University, Guangzhou 510515, China
| | - Chao-Hua Luo
- School of Traditional Chinese Medicine, Southern Medical University, Guangzhou 510515, China
| | - Ming-Jin Jiang
- School of Traditional Chinese Medicine, Southern Medical University, Guangzhou 510515, China
| | - Zhi-Xian Mo
- School of Traditional Chinese Medicine, Southern Medical University, Guangzhou 510515, China.
| | - Ken Kin-Lam Yung
- Department of Biology, Hong Kong Baptist University, Kowloon Tong, Hong Kong.
| |
Collapse
|
41
|
Motbey CP, Clemens KJ, Apetz N, Winstock AR, Ramsey J, Li KM, Wyatt N, Callaghan PD, Bowen MT, Cornish JL, McGregor IS. High levels of intravenous mephedrone (4-methylmethcathinone) self-administration in rats: neural consequences and comparison with methamphetamine. J Psychopharmacol 2013; 27:823-36. [PMID: 23739178 DOI: 10.1177/0269881113490325] [Citation(s) in RCA: 74] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Mephedrone (MMC) is a relatively new recreational drug that has rapidly increased in popularity in recent years. This study explored the characteristics of intravenous MMC self-administration in the rat, with methamphetamine (METH) used as a comparator drug. Male Sprague-Dawley rats were trained to nose poke for intravenous MMC or METH in daily 2 h sessions over a 10 d acquisition period. Dose-response functions were then established under fixed- and progressive-ratio (FR and PR) schedules over three subsequent weeks of testing. Brains were analyzed ex vivo for striatal serotonin (5-HT) and dopamine (DA) levels and metabolites, while autoradiography assessed changes in the regional density of 5-HT and serotonin transporter (SERT) and DA transporter (DAT) and induction of the inflammation marker translocator protein (TSPO). Results showed that MMC was readily and vigorously self-administered via the intravenous route. Under a FR1 schedule, peak responding for MMC was obtained at 0.1 mg/kg/infusion, versus 0.01 mg/kg/infusion for METH. Break points under a PR schedule peaked at 1 mg/kg/infusion MMC versus 0.3 mg/kg/infusion for METH. Final intakes of MMC were 31.3 mg/kg/d compared to 4 mg/kg/d for METH. Rats self-administering MMC, but not METH, gained weight at a slower rate than control rats. METH, but not MMC, self-administration elevated TSPO receptor density in the nucleus accumbens and hippocampus, while MMC, but not METH, self-administration decreased striatal 5-hydroxyindolacetic acid (5-HIAA) concentrations. In summary, MMC supported high levels of self-administration, matching or exceeding those previously reported with other drugs of abuse.
Collapse
Affiliation(s)
- Craig P Motbey
- School of Psychology, University of Sydney, Sydney, Australia
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
42
|
Machaalani R, Hunt NJ, Waters KA. Effects of changes in energy homeostasis and exposure of noxious insults on the expression of orexin (hypocretin) and its receptors in the brain. Brain Res 2013; 1526:102-22. [PMID: 23830852 DOI: 10.1016/j.brainres.2013.06.035] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2013] [Revised: 06/19/2013] [Accepted: 06/25/2013] [Indexed: 12/23/2022]
Abstract
This review summarizes data regarding the brain expression of the orexin (hypocretin) system including: prepro-orexin (PPO), orexin A (OxA), orexin B (OxB) and the two orexin receptors 1 and 2 (OxR1, OxR2). Clinical data is limited to OxA and OxB in cerebral spinal fluid and serum/plasma, thus necessitating the development of animal models to undertake mechanistic studies. We focus on changes in animal models that were either exposed to a regime of altered sleep, metabolic energy homeostasis, exposed to drugs and noxious insults. Many more expressional studies are available for PPO, OxA and OxB levels, compared to studies of the receptors. Interestingly, the direction and pattern of change for PPO, OxA and OxB is inconsistent amongst studies, whereas for the receptors, there tends to be increased expression for both OxR1 and OxR2 after alterations in energy homeostasis, and an increased expression after noxious insults or exposure to some drugs. The clinical implications of these results from animal models are discussed in light of the findings from human studies, and future research directions are suggested to fill knowledge gaps with regard to the orexin system, particularly during early brain development.
Collapse
Affiliation(s)
- Rita Machaalani
- Department of Medicine, Blackburn Building, DO6, University of Sydney, NSW 2006, Australia.
| | | | | |
Collapse
|
43
|
Mephedrone: Public health risk, mechanisms of action, and behavioral effects. Eur J Pharmacol 2013; 714:32-40. [PMID: 23764466 DOI: 10.1016/j.ejphar.2013.05.024] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2012] [Revised: 05/10/2013] [Accepted: 05/24/2013] [Indexed: 11/20/2022]
Abstract
The recent shortage of 3,4-methylenedioxymethamphetamine (MDMA, Ecstasy) has led to an increased demand for alternative amphetamine-like drugs such as the synthetic cathinone, 4-methylmethcathinone (mephedrone). Despite the re-classification of mephedrone as a Class B restricted substance by the United Kingdom and restrictive legislation by the United States, international policy regarding mephedrone control is still developing and interest in synthetic amphetamine-like drugs could drive the development of future mephedrone analogues. Currently, there is little literature investigating the mechanism of action and long-term effects of mephedrone. As such, we reviewed the current understanding of amphetamines, cathinones, and cocaine emphasizing the potentially translational aspects to mephedrone, as well as contrasting with the work that has been done specifically on mephedrone in order to present the current state of understanding of mephedrone in terms of its risks, mechanisms, and behavioral effects. Emerging research suggests that while there are structural and behavioral similarities of mephedrone with amphetamine-like compounds, it appears that serotonergic signaling may mediate more of mephedrone's effects unlike the more dopaminergic dependent effects observed in traditional amphetamine-like compounds. As new designer drugs are produced, current and continuing research on mephedrone and other synthetic cathinones should help inform policymakers' decisions regarding the regulation of novel 'legal highs.'
Collapse
|
44
|
Krasnova IN, Chiflikyan M, Justinova Z, McCoy MT, Ladenheim B, Jayanthi S, Quintero C, Brannock C, Barnes C, Adair JE, Lehrmann E, Kobeissy FH, Gold MS, Becker KG, Goldberg SR, Cadet JL. CREB phosphorylation regulates striatal transcriptional responses in the self-administration model of methamphetamine addiction in the rat. Neurobiol Dis 2013; 58:132-43. [PMID: 23726845 DOI: 10.1016/j.nbd.2013.05.009] [Citation(s) in RCA: 95] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2013] [Revised: 05/08/2013] [Accepted: 05/20/2013] [Indexed: 10/26/2022] Open
Abstract
Neuroplastic changes in the dorsal striatum participate in the transition from casual to habitual drug use and might play a critical role in the development of methamphetamine (METH) addiction. We examined the influence of METH self-administration on gene and protein expression that may form substrates for METH-induced neuronal plasticity in the dorsal striatum. Male Sprague-Dawley rats self-administered METH (0.1mg/kg/injection, i.v.) or received yoked saline infusions during eight 15-h sessions and were euthanized 2h, 24h, or 1month after cessation of METH exposure. Changes in gene and protein expression were assessed using microarray analysis, RT-PCR and Western blots. Chromatin immunoprecipitation (ChIP) followed by PCR was used to examine epigenetic regulation of METH-induced transcription. METH self-administration caused increases in mRNA expression of the transcription factors, c-fos and fosb, the neurotrophic factor, Bdnf, and the synaptic protein, synaptophysin (Syp) in the dorsal striatum. METH also caused changes in ΔFosB, BDNF and TrkB protein levels, with increases after 2 and 24h, but decreases after 1month of drug abstinence. Importantly, ChIP-PCR showed that METH self-administration caused enrichment of phosphorylated CREB (pCREB), but not of histone H3 trimethylated at lysine 4 (H3K4me3), on promoters of c-fos, fosb, Bdnf and Syp at 2h after cessation of drug intake. These findings show that METH-induced changes in gene expression are mediated, in part, by pCREB-dependent epigenetic phenomena. Thus, METH self-administration might trigger epigenetic changes that mediate alterations in expression of genes and proteins serving as substrates for addiction-related synaptic plasticity.
Collapse
Affiliation(s)
- Irina N Krasnova
- Intramural Research Program, National Institute on Drug Abuse, NIH, DHHS, Baltimore, MD, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
45
|
Braking dopamine systems: a new GABA master structure for mesolimbic and nigrostriatal functions. J Neurosci 2013; 32:14094-101. [PMID: 23055478 DOI: 10.1523/jneurosci.3370-12.2012] [Citation(s) in RCA: 165] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
A new mesopontine structure exerting a strong influence on dopamine systems has recently been defined: the tail of the ventral tegmental area/rostromedial tegmental nucleus (tVTA/RMTg). This review presents a neuroanatomical, physiological, and behavioral overview of some of the recent and ongoing research on this brain region and its relationship with dopamine systems. The tVTA/RMTg sends dense GABA projections to VTA and substantia nigra neurons. The inhibitory influence of tVTA/RMTg on dopamine neurons is supported by both neuroanatomical and electrophysiology data. The latter studies also reveal the tVTA/RMTg as a substrate for morphine and cannabinoid action on dopamine cells. In primates, the tVTA/RMTg has been implicated in reward prediction error signals, through a basal ganglia-lateral habenula-tVTA/RMTg-dopamine-basal ganglia circuit. In rodents, the tVTA/RMTg has been shown to play a critical role in aversive behaviors, particularly those involving behavioral inhibition, such as freezing and avoidance. These findings highlight the functional importance of the tVTA/RMTg as a major GABA brake for dopamine systems.
Collapse
|
46
|
A new control center for dopaminergic systems: pulling the VTA by the tail. Trends Neurosci 2012; 35:681-90. [DOI: 10.1016/j.tins.2012.06.007] [Citation(s) in RCA: 162] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2012] [Revised: 06/18/2012] [Accepted: 06/27/2012] [Indexed: 12/13/2022]
|