1
|
Jelen LA, Young AH, Mehta MA. Opioid Mechanisms and the Treatment of Depression. Curr Top Behav Neurosci 2024; 66:67-99. [PMID: 37923934 DOI: 10.1007/7854_2023_448] [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] [Indexed: 11/06/2023]
Abstract
Opioid receptors are widely expressed in the brain, and the opioid system has a key role in modulating mood, reward processing and stress responsivity. There is mounting evidence that the endogenous opioid system may be dysregulated in depression and that drug treatments targeting mu, delta and kappa opioid receptors may show antidepressant potential. The mechanisms underlying the therapeutic effects of opioid system engagement are complex and likely multi-factorial. This chapter explores various pathways through which the modulation of the opioid system may influence depression. These include impacts on monoaminergic systems, the regulation of stress and the hypothalamic-pituitary-adrenal axis, the immune system and inflammation, brain-derived neurotrophic factors, neurogenesis and neuroplasticity, social pain and social reward, as well as expectancy and placebo effects. A greater understanding of the diverse mechanisms through which opioid system modulation may improve depressive symptoms could ultimately aid in the development of safe and effective alternative treatments for individuals with difficult-to-treat depression.
Collapse
Affiliation(s)
- Luke A Jelen
- Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, UK.
- South London and Maudsley NHS Foundation Trust, London, UK.
| | - Allan H Young
- Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, UK
- South London and Maudsley NHS Foundation Trust, London, UK
| | - Mitul A Mehta
- Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, UK
| |
Collapse
|
2
|
Cao DN, Li F, Wu N, Li J. Insights into the mechanisms underlying opioid use disorder and potential treatment strategies. Br J Pharmacol 2023; 180:862-878. [PMID: 34128238 DOI: 10.1111/bph.15592] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2020] [Revised: 05/28/2021] [Accepted: 06/02/2021] [Indexed: 12/19/2022] Open
Abstract
Opioid use disorder is a worldwide societal problem and public health burden. Strategies for treating opioid use disorder can be divided into those that target the opioid receptor system and those that target non-opioid receptor systems, including the dopamine and glutamate receptor systems. Currently, the clinical drugs used to treat opioid use disorder include the opioid receptor agonists methadone and buprenorphine, which are limited by their abuse liability, and the opioid receptor antagonist naltrexone, which is limited by poor compliance. Therefore, the development of effective medications with lower abuse liability and better potential for compliance is urgently needed. Based on recent advances in the understanding of the neurobiological mechanisms underlying opioid use disorder, potential treatment strategies and targets have emerged. This review focuses on the progress made in identifying potential targets and developing medications to treat opioid use disorder, including progress made by our laboratory, and provides insights for future medication development. LINKED ARTICLES: This article is part of a themed issue on Advances in Opioid Pharmacology at the Time of the Opioid Epidemic. To view the other articles in this section visit http://onlinelibrary.wiley.com/doi/10.1111/bph.v180.7/issuetoc.
Collapse
Affiliation(s)
- Dan-Ni Cao
- Beijing Key Laboratory of Neuropsychopharmacology, State Key Laboratory of Toxicology and Medical Countermeasures, Beijing Institute of Pharmacology and Toxicology, Beijing, China
| | - Fei Li
- Beijing Key Laboratory of Neuropsychopharmacology, State Key Laboratory of Toxicology and Medical Countermeasures, Beijing Institute of Pharmacology and Toxicology, Beijing, China
| | - Ning Wu
- Beijing Key Laboratory of Neuropsychopharmacology, State Key Laboratory of Toxicology and Medical Countermeasures, Beijing Institute of Pharmacology and Toxicology, Beijing, China
| | - Jin Li
- Beijing Key Laboratory of Neuropsychopharmacology, State Key Laboratory of Toxicology and Medical Countermeasures, Beijing Institute of Pharmacology and Toxicology, Beijing, China
| |
Collapse
|
3
|
Navaei F, Fathabadi FF, Moghaddam MH, Fathi M, Vakili K, Abdollahifar MA, Boroujeni ME, Zamani N, Zamani N, Norouzian M, Aliaghaei A. Chronic exposure to methadone impairs memory, induces microgliosis, astrogliosis and neuroinflammation in the hippocampus of adult male rats. J Chem Neuroanat 2022; 125:102139. [PMID: 35872237 DOI: 10.1016/j.jchemneu.2022.102139] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2022] [Revised: 07/18/2022] [Accepted: 07/19/2022] [Indexed: 01/15/2023]
Abstract
Methadone is a centrally-acting synthetic opioid analgesic widely used in methadone maintenance therapy (MMT) programs throughout the world. Given its neurotoxic effects, particularly on the hippocampus, this study aims to address the behavioral and histological alterations in the hippocampus associated with methadone administration. To do so, twenty-four adult male albino rats were randomized into two groups, methadone treatment and control. Methadone was administered subcutaneously (2.5-10 mg/kg) once a day for two consecutive weeks. A comparison was drawn with behavioral and structural changes recorded in the control group. The results showed that methadone administration interrupted spatial learning and memory function. Accordingly, treating rats with methadone not only induced cell death but also prompted the actuation of microgliosis, astrogliosis, and apoptotic biomarkers. Furthermore, the results demonstrated that treating rats with methadone decreased the complexity of astrocyte processes and the complexity of microglia processes. These findings suggest that methadone altered the special distribution of neurons. Also, a substantial increase was observed in the expression of TNF-α due to methadone. According to the findings, methadone administration exerts a neurodegenerative effect on the hippocampus via dysregulation of microgliosis, astrogliosis, apoptosis, and neuro-inflammation.
Collapse
Affiliation(s)
- Fatemeh Navaei
- Hearing Disorders Research Center, Loghman-Hakim Hospital, Shahid Beheshti University of Medical Sciences, Tehran, the Islamic Republic of Iran; Department of Biology and Anatomical Sciences, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, the Islamic Republic of Iran
| | - Fatemeh Fadaei Fathabadi
- Department of Biology and Anatomical Sciences, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, the Islamic Republic of Iran
| | - Meysam Hassani Moghaddam
- Department of Anatomical Sciences, Faculty of Medicine, AJA University of Medical Sciences, Tehran, the Islamic Republic of Iran
| | - Mobina Fathi
- Student Research Committee, Faculty of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, the Islamic Republic of Iran
| | - Kimia Vakili
- Student Research Committee, Faculty of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, the Islamic Republic of Iran
| | - Mohammad-Amin Abdollahifar
- Department of Biology and Anatomical Sciences, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, the Islamic Republic of Iran
| | - Mahdi Eskandarian Boroujeni
- Department of Human Molecular Genetics, Institute of Molecular Biology and Biotechnology, Faculty of Biology, Adam Mickiewicz University, Poznan, Poland
| | - Naghmeh Zamani
- Department of Biology, Tehran North Branch, Islamic Azad University, Tehran, the Islamic Republic of Iran
| | - Nasim Zamani
- Department of Clinical Toxicology, Loghman-Hakim Hospital, Shahid Beheshti University of Medical Sciences, Tehran, the Islamic Republic of Iran
| | - Mohsen Norouzian
- Department of Biology and Anatomical Sciences, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, the Islamic Republic of Iran.
| | - Abbas Aliaghaei
- Hearing Disorders Research Center, Loghman-Hakim Hospital, Shahid Beheshti University of Medical Sciences, Tehran, the Islamic Republic of Iran; Department of Biology and Anatomical Sciences, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, the Islamic Republic of Iran.
| |
Collapse
|
4
|
Prefrontal Cortex Response to Prenatal Insult and Postnatal Opioid Exposure. Genes (Basel) 2022; 13:genes13081371. [PMID: 36011282 PMCID: PMC9407090 DOI: 10.3390/genes13081371] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2022] [Revised: 07/27/2022] [Accepted: 07/28/2022] [Indexed: 12/11/2022] Open
Abstract
The influence of proinflammatory challenges, such as maternal immune activation (MIA) or postnatal exposure to drugs of abuse, on brain molecular pathways has been reported. On the other hand, the simultaneous effects of MIA and drugs of abuse have been less studied and sometimes offered inconsistent results. The effects of morphine exposure on a pig model of viral-elicited MIA were characterized in the prefrontal cortex of males and females using RNA-sequencing and gene network analysis. Interacting and main effects of morphine, MIA, and sex were detected in approximately 2000 genes (false discovery rate-adjusted p-value < 0.05). Among the enriched molecular categories (false discovery rate-adjusted p-value < 0.05 and −1.5 > normalized enrichment score > 1.5) were the cell adhesion molecule pathways associated with inflammation and neuronal development and the long-term depression pathway associated with synaptic strength. Gene networks that integrate gene connectivity and expression profiles displayed the impact of morphine-by-MIA interaction effects on the pathways. The cell adhesion molecules and long-term depression networks presented an antagonistic effect between morphine and MIA. The differential expression between the double-challenged group and the baseline saline-treated Controls was less extreme than the individual challenges. The previous findings advance the knowledge about the effects of prenatal MIA and postnatal morphine exposure on the prefrontal cortex pathways.
Collapse
|
5
|
Pourhassanali N, Zarbakhsh S, Miladi-Gorji H. Morphine dependence and withdrawal-induced changes in mouse Sertoli cell (TM4) line: Evaluation of apoptotic, inflammatory and oxidative stress biomarkers. Reprod Toxicol 2021; 105:175-183. [PMID: 34517100 DOI: 10.1016/j.reprotox.2021.09.004] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2021] [Revised: 09/02/2021] [Accepted: 09/08/2021] [Indexed: 12/01/2022]
Abstract
Chronic morphine exerts deleterious effects on testicular function through either suppression of germ cells or somatic including Sertoli cells, probably through the activation of inflammatory, oxidative, and apoptosis biomarkers. Thus, the present study aimed to investigate whether the damaging effects of morphine dependence were reversed by the spontaneous morphine withdrawal or incubation with methadone and/or naloxone in Sertoli (TM4) cells using an in- vitro cell model of morphine dependence. Morphine dependence in TM4 cells was induced by increasing daily doses of morphine for 10 days and then maintained for two weeks in 5 μM. The cAMP levels were measured for an evaluation of morphine dependence. The cell viability and inflammatory, oxidative, apoptosis biomarkers, and glial cell-derived neurotrophic factor (GDNF) were measured after the end of treatment following the incubation of cells with methadone and naloxone and spontaneous withdrawal from morphine. We found that morphine dependence decreased cell viability, GDNF level and increased the levels of pro-oxidant, pro-inflammatory, and apoptotic biomarkers in TM4 cells, while spontaneous withdrawal from morphine and by naloxone decreased the levels of the biomarkers of pro-inflammatory and apoptotic in TM4 cells. Also, despite the low levels of pro-inflammatory factors following morphine withdrawal by methadone, it increased the cleaved/pro-caspase3 ratio in TM4 cells. This study showed that morphine dependence increased apoptosis probably via oxidative stress and inflammation pathways in TM4 cells. Also, it seems likely that spontaneous and naloxone withdrawal have beneficial consequences in the treatment of morphine dependence than methadone therapy, although they may require longer incubation periods.
Collapse
Affiliation(s)
- Nazila Pourhassanali
- Research Center of Physiology, School of Medicine, Semnan University of Medical Sciences, Semnan, Iran; Department of Physiology, School of Medicine, Semnan University of Medical Sciences, Semnan, Iran
| | - Sam Zarbakhsh
- Department of Anatomical Sciences, School of Medicine, Semnan University of Medical Sciences, Semnan, Iran
| | - Hossein Miladi-Gorji
- Research Center of Physiology, School of Medicine, Semnan University of Medical Sciences, Semnan, Iran; Department of Physiology, School of Medicine, Semnan University of Medical Sciences, Semnan, Iran.
| |
Collapse
|
6
|
Asuni GP, Speidell A, Mocchetti I. Neuronal apoptosis induced by morphine withdrawal is mediated by the p75 neurotrophin receptor. J Neurochem 2021; 158:169-181. [PMID: 33742683 PMCID: PMC10176599 DOI: 10.1111/jnc.15355] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2020] [Revised: 02/22/2021] [Accepted: 03/15/2021] [Indexed: 01/01/2023]
Abstract
Morphine withdrawal evokes neuronal apoptosis through mechanisms that are still under investigation. We have previously shown that morphine withdrawal increases the levels of pro-brain-derived neurotrophic factor (BDNF), a proneurotrophin that promotes neuronal apoptosis through the binding and activation of the pan-neurotrophin receptor p75 (p75NTR). In this work, we sought to examine whether morphine withdrawal increases p75NTR-driven signaling events. We employed a repeated morphine treatment-withdrawal paradigm in order to investigate biochemical and histological indicators of p75NTR-mediated neuronal apoptosis in mice. We found that repeated cycles of spontaneous morphine withdrawal promote an accumulation of p75NTR in hippocampal synapses. At the same time, TrkB, the receptor that is crucial for BDNF-mediated synaptic plasticity in the hippocampus, was decreased, suggesting that withdrawal alters the neurotrophin receptor environment to favor synaptic remodeling and apoptosis. Indeed, we observed evidence of neuronal apoptosis in the hippocampus, including activation of c-Jun N-terminal kinase (JNK) and increased active caspase-3. These effects were not seen in saline or morphine-treated mice which had not undergone withdrawal. To determine whether p75NTR was necessary in promoting these outcomes, we repeated these experiments in p75NTR heterozygous mice. The lack of one p75NTR allele was sufficient to prevent the increases in phosphorylated JNK and active caspase-3. Our results suggest that p75NTR participates in the neurotoxic and proinflammatory state evoked by morphine withdrawal. Because p75NTR activation negatively influences synaptic repair and promotes cell death, preventing opioid withdrawal is crucial for reducing neurotoxic mechanisms accompanying opioid use disorders.
Collapse
Affiliation(s)
- Gino P. Asuni
- Laboratory of Preclinical Neurobiology, Georgetown University Medical Center, Washington DC, USA
| | - Andrew Speidell
- Laboratory of Preclinical Neurobiology, Georgetown University Medical Center, Washington DC, USA
- Interdisciplinary Program in Neuroscience, Georgetown University Medical Center, Washington DC, USA
| | - Italo Mocchetti
- Laboratory of Preclinical Neurobiology, Georgetown University Medical Center, Washington DC, USA
- Interdisciplinary Program in Neuroscience, Georgetown University Medical Center, Washington DC, USA
| |
Collapse
|
7
|
Willner MJ, Xiao Y, Kim HS, Chen X, Xu B, Leong KW. Modeling SARS-CoV-2 infection in individuals with opioid use disorder with brain organoids. J Tissue Eng 2021; 12:2041731420985299. [PMID: 33738089 PMCID: PMC7934045 DOI: 10.1177/2041731420985299] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2020] [Accepted: 12/13/2020] [Indexed: 01/10/2023] Open
Abstract
The COVID-19 pandemic has aggravated a preexisting epidemic: the opioid crisis. Much literature has shown that the circumstances imposed by COVID-19, such as social distancing regulations, medical and financial instability, and increased mental health issues, have been detrimental to those with opioid use disorder (OUD). In addition, unexpected neurological sequelae in COVID-19 patients suggest that COVID-19 compromises neuroimmunity, induces hypoxia, and causes respiratory depression, provoking similar effects as those caused by opioid exposure. Combined conditions of COVID-19 and OUD could lead to exacerbated complications. With limited human in vivo options to study these complications, we suggest that iPSC-derived brain organoid models may serve as a useful platform to investigate the physiological connection between COVID-19 and OUD. This mini-review highlights the advances of brain organoids in other neuropsychiatric and infectious diseases and suggests their potential utility for investigating OUD and COVID-19, respectively.
Collapse
Affiliation(s)
- Moshe J Willner
- Department of Biomedical Engineering, Columbia University, New York, NY, USA
| | - Yang Xiao
- Department of Biomedical Engineering, Columbia University, New York, NY, USA
| | - Hye Sung Kim
- Department of Biomedical Engineering, Columbia University, New York, NY, USA
- Institute of Tissue Regeneration Engineering, Dankook University, Cheonan, Republic of Korea
- Department of Regenerative Dental Medicine, College of Dentistry, Dankook University, Cheonan, Republic of Korea
- Cell & Matter Institute, Dankook University, Cheonan, Republic of Korea
| | - Xuejing Chen
- Department of Biomedical Engineering, Columbia University, New York, NY, USA
- Department of Physics, Tsinghua University, Beijing, China
| | - Bin Xu
- Department of Psychiatry, Columbia University Medical Center, New York, NY, USA
| | - Kam W Leong
- Department of Biomedical Engineering, Columbia University, New York, NY, USA
- Department of Systems Biology, Columbia University Irving Medical Center, New York, NY, USA
| |
Collapse
|
8
|
Bulin SE, Simmons SJ, Richardson DR, Latchney SE, Deutsch HM, Yun S, Eisch AJ. Indices of dentate gyrus neurogenesis are unaffected immediately after or following withdrawal from morphine self-administration compared to saline self-administering control male rats. Behav Brain Res 2020; 381:112448. [PMID: 31870778 PMCID: PMC7036141 DOI: 10.1016/j.bbr.2019.112448] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2019] [Revised: 12/01/2019] [Accepted: 12/19/2019] [Indexed: 12/16/2022]
Abstract
Opiates - including morphine - are powerful analgesics with high abuse potential. In rodents, chronic opiate exposure or self-administration negatively impacts hippocampal-dependent function, an effect perhaps due in part to the well-documented opiate-induced inhibition of dentate gyrus (DG) precursor proliferation and neurogenesis. Recently, however, intravenous (i.v.) morphine self-administration (MSA) was reported to enhance the survival of new rat DG neurons. To reconcile these disparate results, we used rat i.v. MSA to assess 1) whether a slightly-higher dose MSA paradigm also increases new DG neuron survival; 2) how MSA influences cells in different stages of DG neurogenesis, particularly maturation and survival; and 3) if MSA-induced changes in DG neurogenesis persist through a period of abstinence. To label basal levels of proliferation, rats received the S-phase marker bromodeoxyuridine (BrdU, i.p.) 24 -h prior to 21 days (D) of i.v. MSA or saline self-administration (SSA). Either immediately after SA (0-D) or after 4 weeks in the home cage (28-D withdrawal), stereology was used to quantify DG proliferating precursors (or cells in cell cycle; Ki67+ cells), neuroblast/immature neurons (DCX+ cells), and surviving DG granule cells (BrdU+ cells). Analysis revealed the number of DG cells immunopositive for these neurogenesis-relevant markers was similar between MSA and SSA rats at the 0-D or 28-D timepoints. These negative data highlight the impact experimental parameters, timepoint selection, and quantification approach have on neurogenesis results, and are discussed in the context of the large literature showing the negative impact of opiates on DG neurogenesis.
Collapse
Affiliation(s)
- Sarah E Bulin
- Department of Psychiatry, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Steven J Simmons
- Department of Anesthesiology and Critical Care Medicine, Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Devon R Richardson
- Department of Psychiatry, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Sarah E Latchney
- Department of Psychiatry, University of Texas Southwestern Medical Center, Dallas, TX, USA; Department of Neurobiology, St. Mary's College of Maryland, St. Mary's City, MD, 20686-3001
| | - Hannah M Deutsch
- Department of Anesthesiology and Critical Care Medicine, Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Sanghee Yun
- Department of Anesthesiology and Critical Care Medicine, Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Amelia J Eisch
- Department of Psychiatry, University of Texas Southwestern Medical Center, Dallas, TX, USA; Department of Anesthesiology and Critical Care Medicine, Children's Hospital of Philadelphia, Philadelphia, PA, USA; Department of Neuroscience and Mahoney Institute for Neurosciences, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA.
| |
Collapse
|
9
|
Kibaly C, Xu C, Cahill CM, Evans CJ, Law PY. Non-nociceptive roles of opioids in the CNS: opioids' effects on neurogenesis, learning, memory and affect. Nat Rev Neurosci 2019; 20:5-18. [PMID: 30518959 DOI: 10.1038/s41583-018-0092-2] [Citation(s) in RCA: 46] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
Mortality due to opioid use has grown to the point where, for the first time in history, opioid-related deaths exceed those caused by car accidents in many states in the United States. Changes in the prescribing of opioids for pain and the illicit use of fentanyl (and derivatives) have contributed to the current epidemic. Less known is the impact of opioids on hippocampal neurogenesis, the functional manipulation of which may improve the deleterious effects of opioid use. We provide new insights into how the dysregulation of neurogenesis by opioids can modify learning and affect, mood and emotions, processes that have been well accepted to motivate addictive behaviours.
Collapse
Affiliation(s)
- Cherkaouia Kibaly
- Department of Psychiatry and Biobehavioral Sciences, Semel Institute for Neuroscience and Human Behavior, Shirley and Stefan Hatos Center for Neuropharmacology, University of California, Los Angeles, CA, USA.
| | - Chi Xu
- State Key Laboratory of Natural Medicines, School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing, China
| | - Catherine M Cahill
- Department of Psychiatry and Biobehavioral Sciences, Semel Institute for Neuroscience and Human Behavior, Shirley and Stefan Hatos Center for Neuropharmacology, University of California, Los Angeles, CA, USA
| | - Christopher J Evans
- Department of Psychiatry and Biobehavioral Sciences, Semel Institute for Neuroscience and Human Behavior, Shirley and Stefan Hatos Center for Neuropharmacology, University of California, Los Angeles, CA, USA
| | - Ping-Yee Law
- Department of Psychiatry and Biobehavioral Sciences, Semel Institute for Neuroscience and Human Behavior, Shirley and Stefan Hatos Center for Neuropharmacology, University of California, Los Angeles, CA, USA
| |
Collapse
|
10
|
Dentate gyrus μ-opioid receptor-mediated neurogenic processes are associated with alterations in morphine self-administration. Sci Rep 2019; 9:1471. [PMID: 30728362 PMCID: PMC6365505 DOI: 10.1038/s41598-018-37083-8] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2017] [Accepted: 11/30/2018] [Indexed: 02/06/2023] Open
Abstract
Adult hippocampal dentate gyrus (DG) neural stem cells (NSCs) continuously undergo proliferation and differentiation, producing new functional neurons that remodel existing synaptic circuits. Although proliferation of these adult DG NSCs has been implicated in opiate dependence, whether NSC neuronal differentiation and subsequent dendritogenesis are also involved in such addictive behavior remains unknown. Here, we ask whether opiate exposure alters differentiation and dendritogenesis of DG NSCs and investigate the possibility that these alterations contribute to opiate addiction. We show that rat morphine self-administration (MSA), a paradigm that effectively mimics human opiate addiction, increases NSC neuronal differentiation and promotes neuronal dendrite growth in the adult DG. Further, we demonstrate that the μ-opioid receptor (MOR) is expressed on DG NSCs and that MSA leads to a two-fold elevation of endogenous MOR levels in doublecortin expressing (DCX+) NSC progenies in the rat DG. MOR expression is also detected in the cultured rat NSCs and morphine treatment in vitro increases NSC neuronal differentiation and dendritogenesis, suggesting that MOR mediates the effect of morphine on NSC neuronal differentiation and maturation. Finally, we show that conditional overexpression of MOR in DG NSCs under a doxycycline inducible system leads to facilitation of the acquisition of MSA in rats, without affecting the extinction process. We advocate that targeting MOR selectively in the DG NSC population might offer a novel therapeutic intervention for morphine addiction.
Collapse
|
11
|
Short term exposure to oxycodone alters the survival, proliferation and differentiation of rat embryonic neural stem cell in vitro. Brain Res Bull 2018; 143:66-72. [PMID: 30290204 DOI: 10.1016/j.brainresbull.2018.09.019] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2018] [Revised: 09/03/2018] [Accepted: 09/30/2018] [Indexed: 11/22/2022]
Abstract
As one of several opioids, oxycodone has been widely used, particularly in postoperative analgesia in children and cesarean section. However, the effect of oxycodone on developing brain still remains to be seen. Since there is a link between anesthetics exposure and long-term behavioral or cognitive dysfunction in young children, in the current study, the direct effect of oxycodone on neural stem cells (NSCs) biological behaviors was investigated. After exposed to a high dose of oxycodone (10 μg/mL) for 48 h, NSCs survival and proliferation were significantly reduced, while NSCs apoptosis and differentiation were enhanced. These effects were significantly weaker than that when exposed to same dose of morphine. No significant difference was observed regarding to above biological behaviors when exposed to lower doses (0.1 μg/mL and 1.0 μg/mL) of oxycodone. The antagonist of opioid receptor, nalmefene, successfully reversed the influence of oxycodone. Taken together, our results indicated that short term exposure to oxycodone in low dose might be allowed for developing brain.
Collapse
|
12
|
Cortical Expression of the Polysialylated Isoform of the Neural Cell Adhesion Molecule on Brain Tissue to Recognize Drug-Related Death. ACTA ACUST UNITED AC 2018; 39:8-13. [DOI: 10.1097/paf.0000000000000366] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
|
13
|
Bulin SE, Mendoza ML, Richardson DR, Song KH, Solberg TD, Yun S, Eisch AJ. Dentate gyrus neurogenesis ablation via cranial irradiation enhances morphine self-administration and locomotor sensitization. Addict Biol 2018. [PMID: 28626932 DOI: 10.1111/adb.12524] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Adult dentate gyrus (DG) neurogenesis is important for hippocampal-dependent learning and memory, but the role of new neurons in addiction-relevant learning and memory is unclear. To test the hypothesis that neurogenesis is involved in the vulnerability to morphine addiction, we ablated adult DG neurogenesis and examined morphine self-administration (MSA) and locomotor sensitization. Male Sprague-Dawley rats underwent hippocampal-focused, image-guided X-ray irradiation (IRR) to eliminate new DG neurons or sham treatment (Sham). Six weeks later, rats underwent either MSA (Sham = 16, IRR = 15) or locomotor sensitization (Sham = 12, IRR = 12). Over 21 days of MSA, IRR rats self-administered ~70 percent more morphine than Sham rats. After 28 days of withdrawal, IRR rats pressed the active lever 40 percent more than Sham during extinction. This was not a general enhancement of learning or locomotion, as IRR and Sham groups had similar operant learning and inactive lever presses. For locomotor sensitization, both IRR and Sham rats sensitized, but IRR rats sensitized faster and to a greater extent. Furthermore, dose-response revealed that IRR rats were more sensitive at a lower dose. Importantly, these increases in locomotor activity were not apparent after acute morphine administration and were not a byproduct of irradiation or post-irradiation recovery time. Therefore, these data, along with other previously published data, indicate that reduced hippocampal neurogenesis confers vulnerability for multiple classes of drugs. Thus, therapeutics to specifically increase or stabilize hippocampal neurogenesis could aid in preventing initial addiction as well as future relapse.
Collapse
Affiliation(s)
- Sarah E. Bulin
- Department of Psychiatry; University of Texas Southwestern Medical Center; Dallas TX USA
- Department of Pharmacology; University of Texas Health Science Center; San Antonio TX USA
| | - Matthew L. Mendoza
- Department of Psychiatry; University of Texas Southwestern Medical Center; Dallas TX USA
| | - Devon R. Richardson
- Department of Psychiatry; University of Texas Southwestern Medical Center; Dallas TX USA
| | - Kwang H. Song
- Department of Radiology Oncology; University of Texas Southwestern Medical Center; Dallas TX USA
- Texas Oncology PA; Fort Worth TX USA
| | - Timothy D. Solberg
- Department of Radiology Oncology; University of Texas Southwestern Medical Center; Dallas TX USA
- Department of Radiation Oncology; University of California; San Francisco CA USA
| | - Sanghee Yun
- Mahoney Institute of Neurosciences; University of Pennsylvania Perelman School of Medicine; Philadelphia PA USA
- Department of Anesthesiology and Critical Care Medicine; Children's Hospital of Philadelphia; Philadelphia PA USA
| | - Amelia J. Eisch
- Department of Psychiatry; University of Texas Southwestern Medical Center; Dallas TX USA
- Mahoney Institute of Neurosciences; University of Pennsylvania Perelman School of Medicine; Philadelphia PA USA
- Department of Anesthesiology and Critical Care Medicine; Children's Hospital of Philadelphia; Philadelphia PA USA
| |
Collapse
|
14
|
Impact of morphine on the expression of insulin receptor and protein levels of insulin/IGFs in rat neural stem cells. Neurosci Lett 2017; 660:147-154. [DOI: 10.1016/j.neulet.2017.09.035] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2017] [Revised: 09/07/2017] [Accepted: 09/15/2017] [Indexed: 12/24/2022]
|
15
|
Liu Y, Lu GY, Chen WQ, Li YF, Wu N, Li J. Agmatine inhibits chronic morphine exposure-induced impairment of hippocampal neural progenitor proliferation in adult rats. Eur J Pharmacol 2017; 818:50-56. [PMID: 29031903 DOI: 10.1016/j.ejphar.2017.10.018] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2017] [Revised: 10/09/2017] [Accepted: 10/10/2017] [Indexed: 02/09/2023]
Abstract
Our previous studies have shown that agmatine inhibited opioid dependence, yet the neural mechanism remains unclear. Growing evidence showed that opioids decrease neurogenesis in the adult hippocampal subgranular zone by inhibiting neural progenitor proliferation. However, whether agmatine affects chronic opioid exposure-induced impairment to hippocampal neural progenitor cell proliferation remains unknown. In the present study, we investigated the role of agmatine in hippocampal neural progenitors in morphine dependence rats. We found that chronic administration of morphine for 12 days induced morphine dependence in rats. This treatment not only decreased the proliferation of hippocampal neural progenitors in the granule cell layer, but also decreased the levels of hippocampal cAMP, pCREB and BDNF. However, these alterations can be restored to normal levels by co-treatment of agmatine (10mg/kg, s.c.). In vitro treatment with agmatine (10µM) for two days significantly increased proliferation of the cultured hippocampal neural progenitors. Concurrent treatment of agmatine (10µM) with morphine (10 or 50µM) reversed the supression of morphine-induced neural progenitor proliferation. In conclusion, we found that agmatine abolished chronic morphine-induced decrease in proliferation of hippocampal progenitors in vivo and in vitro, which may be due to the increase in cAMP-CREB-BDNF signaling. The enhancement of agmatine to proliferation of hippocampal progenitors may be one of the important mechanisms involved in the inhibition of morphine dependence by agmatine.
Collapse
Affiliation(s)
- Ying Liu
- Department of New Drug Evaluation, Beijing Institute of Pharmacology and Toxicology, 27 Taiping Road, Beijing 100850, PR China
| | - Guan-Yi Lu
- Department of New Drug Evaluation, Beijing Institute of Pharmacology and Toxicology, 27 Taiping Road, Beijing 100850, PR China
| | - Wen-Qiang Chen
- Department of New Drug Evaluation, Beijing Institute of Pharmacology and Toxicology, 27 Taiping Road, Beijing 100850, PR China
| | - Yun-Feng Li
- Department of New Drug Evaluation, Beijing Institute of Pharmacology and Toxicology, 27 Taiping Road, Beijing 100850, PR China
| | - Ning Wu
- Department of New Drug Evaluation, Beijing Institute of Pharmacology and Toxicology, 27 Taiping Road, Beijing 100850, PR China.
| | - Jin Li
- Department of New Drug Evaluation, Beijing Institute of Pharmacology and Toxicology, 27 Taiping Road, Beijing 100850, PR China.
| |
Collapse
|
16
|
Grilli M. Chronic pain and adult hippocampal neurogenesis: translational implications from preclinical studies. J Pain Res 2017; 10:2281-2286. [PMID: 29033604 PMCID: PMC5614764 DOI: 10.2147/jpr.s146399] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Adult hippocampal neurogenesis (ahNG) occurs in the human brain. Adult generated neurons have been proposed to functionally contribute to relevant hippocampal functions such as learning and memory, mood regulation, and stress response. Learning, environmental enrichment, and physical exercise exert positive effects on ahNG. In parallel, these proneurogenic stimuli have been shown to ameliorate cognitive performance and/or depressive-like behavior in animal models. Conversely, aging, social isolation, and chronic stress exert negative effects on ahNG. Interestingly, reduction of hippocampal neurogenesis is suggested to potentially contribute to cognitive decline and mood alterations associated with aging and several neuropsychiatric disorders. Clinical observation demonstrates that patients affected by chronic pain often exhibit increased anxiety and depression, impaired cognitive flexibility, and memory capacities. As of today, our understanding of the molecular and cellular events that may underlie the comorbidity of chronic pain, depression, and cognitive impairment is limited. Herein we review recent preclinical data suggesting that chronic pain may induce profound changes in hippocampal plasticity, including reduced ahNG. We discuss the possibility that deregulated hippocampal neurogenesis in chronic pain may, at least in part, contribute to cognitive and mood alterations. Based on this hypothesis, the mechanisms underlying chronic pain-associated changes in hippocampal neurogenesis and related functions need to be addressed experimentally. One interesting feature of ahNG is its susceptibility to pharmacological modulation. Again, based on preclinical data we discuss the possibility that, at least in principle, distinct analgesic drugs commonly used in chronic pain states (typical and atypical opiates, α2δ ligands, and acetyl-l-carnitine) may differentially impact ahNG and that this aspect could be taken into account to reduce and/or prevent the potential risk of cognitive and emotional side effects in the clinical setting.
Collapse
Affiliation(s)
- Mariagrazia Grilli
- Laboratory of Neuroplasticity, Department of Pharmaceutical Sciences, University of Piemonte Orientale, Novara, Italy
| |
Collapse
|
17
|
|
18
|
Baei F, Rajabzadeh A, Bagheri J, Jalayeri Z, Ebrahimzadeh-Bideskan A. Effect of methamphetamine exposure during pregnancy and lactation on polysialic acid-neural cell adhesion molecule expression in rat's offspring hippocampus. Metab Brain Dis 2017; 32:991-1002. [PMID: 28243847 DOI: 10.1007/s11011-017-9973-8] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/03/2016] [Accepted: 02/07/2017] [Indexed: 12/14/2022]
Abstract
Pregnant women constitute about half the users of methamphetamine (MA), in whom the consumption may continue during breastfeeding. This study aimed to evaluate the effects of MA use during pregnancy and lactation on the hippocampus of pups. 35 pregnant Wistar rats were divided into seven groups, including three experimental groups daily administered with 5 mg/kg of MA (i.p.) during the prenatal and/or postnatal period (PND1-22). In addition, three sham control groups received normal saline at the same dose, and one normal control group received no interventions since early pregnancy until the end of lactation. After the interventions, two pups (aged one and 22 days) were randomly selected from each mother and their brain tissue sections were prepared to determine the expression of PSA-NCAM molecules and sialic acids using immunohistochemical and lectinhistochemical techniques, respectively. In one-day infant rats with MA exposure during pregnancy, a significant decrease was observed in the number of PSA-NCAM positive cells in the CA1 (P = 0.047), CA3 (P = 0.05) and DG (P = 0.006) hippocampus regions compared to control and expression intensity of these molecules in all the three regions (P ≤ 0.05). Moreover, in 22-day pups with MA exposure during pregnancy and lactation, number of PSA-NCAM positive cells and expression intensity of these molecules significantly reduced in all the three regions of the hippocampus (P ≤ 0.05). Findings regarding the intensity of sialic acid expression were aligned with PSA-NCAM expression. According to our results, MA administration during pregnancy and lactation may effect on polysialic acid-neural cell adhesion molecule expression in rat's offspring hippocampus.
Collapse
Affiliation(s)
- Fariba Baei
- Department of Anatomy and cell biology, School of Medicine, Mashhad University of Medical Sciences, Azadi Sq., Vakilabad Blvd, P.O. Box 91779-48564, Mashhad, Iran
| | - Aliakbar Rajabzadeh
- Department of Anatomy and cell biology, School of Medicine, Mashhad University of Medical Sciences, Azadi Sq., Vakilabad Blvd, P.O. Box 91779-48564, Mashhad, Iran
- Microanatomy research center, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Javad Bagheri
- Department of Anatomy and cell biology, School of Medicine, Mashhad University of Medical Sciences, Azadi Sq., Vakilabad Blvd, P.O. Box 91779-48564, Mashhad, Iran
| | - Zahra Jalayeri
- Department of Anatomy and cell biology, School of Medicine, Mashhad University of Medical Sciences, Azadi Sq., Vakilabad Blvd, P.O. Box 91779-48564, Mashhad, Iran
| | - Alireza Ebrahimzadeh-Bideskan
- Department of Anatomy and cell biology, School of Medicine, Mashhad University of Medical Sciences, Azadi Sq., Vakilabad Blvd, P.O. Box 91779-48564, Mashhad, Iran.
- Microanatomy research center, Mashhad University of Medical Sciences, Mashhad, Iran.
| |
Collapse
|
19
|
Bortolotto V, Grilli M. Opiate Analgesics as Negative Modulators of Adult Hippocampal Neurogenesis: Potential Implications in Clinical Practice. Front Pharmacol 2017; 8:254. [PMID: 28536527 PMCID: PMC5422555 DOI: 10.3389/fphar.2017.00254] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2017] [Accepted: 04/24/2017] [Indexed: 12/12/2022] Open
Abstract
During the past decade, studies of the mechanisms and functional implications of adult hippocampal neurogenesis (ahNG) have significantly progressed. At present, it is proposed that adult born neurons may contribute to a variety of hippocampal-related functions, including specific cognitive aspects and mood regulation. Several groups focussed on the factors that regulate proliferation and fate determination of adult neural stem/progenitor cells (NSC/NPC), including clinically relevant drugs. Opiates were the first drugs shown to negatively impact neurogenesis in the adult mammalian hippocampus. Since that initial report, a vast array of information has been collected on the effect of opiate drugs, by either modulating proliferation of stem/progenitor cells or interfering with differentiation, maturation and survival of adult born neurons. The goal of this review is to critically revise the present state of knowledge on the effect of opiate drugs on the different developmental stages of ahNG, as well as the possible underlying mechanisms. We will also highlight the potential impact of deregulated hippocampal neurogenesis on patients undergoing chronic opiate treatment. Finally, we will discuss the differences in the negative impact on ahNG among clinically relevant opiate drugs, an aspect that may be potentially taken into account to avoid long-term deregulation of neural plasticity and its associated functions in the clinical practice.
Collapse
Affiliation(s)
- Valeria Bortolotto
- Laboratory of Neuroplasticity, Department of Pharmaceutical Sciences, University of Piemonte OrientaleNovara, Italy
| | - Mariagrazia Grilli
- Laboratory of Neuroplasticity, Department of Pharmaceutical Sciences, University of Piemonte OrientaleNovara, Italy
| |
Collapse
|
20
|
Ladrón de Guevara-Miranda D, Millón C, Rosell-Valle C, Pérez-Fernández M, Missiroli M, Serrano A, Pavón FJ, Rodríguez de Fonseca F, Martínez-Losa M, Álvarez-Dolado M, Santín LJ, Castilla-Ortega E. Long-lasting memory deficits in mice withdrawn from cocaine are concomitant with neuroadaptations in hippocampal basal activity, GABAergic interneurons and adult neurogenesis. Dis Model Mech 2017; 10:323-336. [PMID: 28138095 PMCID: PMC5374316 DOI: 10.1242/dmm.026682] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2016] [Accepted: 01/23/2017] [Indexed: 01/01/2023] Open
Abstract
Cocaine addiction disorder is notably aggravated by concomitant cognitive and emotional pathology that impedes recovery. We studied whether a persistent cognitive/emotional dysregulation in mice withdrawn from cocaine holds a neurobiological correlate within the hippocampus, a limbic region with a key role in anxiety and memory but that has been scarcely investigated in cocaine addiction research. Mice were submitted to a chronic cocaine (20 mg/kg/day for 12 days) or vehicle treatment followed by 44 drug-free days. Some mice were then assessed on a battery of emotional (elevated plus-maze, light/dark box, open field, forced swimming) and cognitive (object and place recognition memory, cocaine-induced conditioned place preference, continuous spontaneous alternation) behavioral tests, while other mice remained in their home cage. Relevant hippocampal features [basal c-Fos activity, GABA+, parvalbumin (PV)+ and neuropeptide Y (NPY)+ interneurons and adult neurogenesis (cell proliferation and immature neurons)] were immunohistochemically assessed 73 days after the chronic cocaine or vehicle protocol. The cocaine-withdrawn mice showed no remarkable exploratory or emotional alterations but were consistently impaired in all the cognitive tasks. All the cocaine-withdrawn groups, independent of whether they were submitted to behavioral assessment or not, showed enhanced basal c-Fos expression and an increased number of GABA+ cells in the dentate gyrus. Moreover, the cocaine-withdrawn mice previously submitted to behavioral training displayed a blunted experience-dependent regulation of PV+ and NPY+ neurons in the dentate gyrus, and neurogenesis in the hippocampus. Results highlight the importance of hippocampal neuroplasticity for the ingrained cognitive deficits present during chronic cocaine withdrawal.
Collapse
Affiliation(s)
- David Ladrón de Guevara-Miranda
- Departamento de Psicobiología y Metodología de las Ciencias del Comportamiento, Instituto de Investigación Biomédica de Málaga (IBIMA), Facultad de Psicología, Universidad de Málaga, 29071 Málaga, Spain
| | - Carmelo Millón
- Departamento de Fisiología, Instituto de Investigación Biomédica de Málaga (IBIMA), Facultad de Medicina, Universidad de Málaga, 29071 Málaga, Spain
| | - Cristina Rosell-Valle
- Departamento de Psicobiología y Metodología de las Ciencias del Comportamiento, Instituto de Investigación Biomédica de Málaga (IBIMA), Facultad de Psicología, Universidad de Málaga, 29071 Málaga, Spain
| | - Mercedes Pérez-Fernández
- Laboratory of Cell-based Therapy for Neuropathologies, Centro Andaluz de Biología Molecular y Medicina Regenerativa (CABIMER), 41092 Sevilla, Spain
| | - Michele Missiroli
- Laboratory of Cell-based Therapy for Neuropathologies, Centro Andaluz de Biología Molecular y Medicina Regenerativa (CABIMER), 41092 Sevilla, Spain
| | - Antonia Serrano
- Unidad de Gestión Clínica de Salud Mental, Instituto de Investigación Biomédica de Málaga (IBIMA), Hospital Regional Universitario de Málaga, 29010 Málaga, Spain
| | - Francisco J Pavón
- Unidad de Gestión Clínica de Salud Mental, Instituto de Investigación Biomédica de Málaga (IBIMA), Hospital Regional Universitario de Málaga, 29010 Málaga, Spain
| | - Fernando Rodríguez de Fonseca
- Unidad de Gestión Clínica de Salud Mental, Instituto de Investigación Biomédica de Málaga (IBIMA), Hospital Regional Universitario de Málaga, 29010 Málaga, Spain
| | - Magdalena Martínez-Losa
- Laboratory of Cell-based Therapy for Neuropathologies, Centro Andaluz de Biología Molecular y Medicina Regenerativa (CABIMER), 41092 Sevilla, Spain
| | - Manuel Álvarez-Dolado
- Laboratory of Cell-based Therapy for Neuropathologies, Centro Andaluz de Biología Molecular y Medicina Regenerativa (CABIMER), 41092 Sevilla, Spain
| | - Luis J Santín
- Departamento de Psicobiología y Metodología de las Ciencias del Comportamiento, Instituto de Investigación Biomédica de Málaga (IBIMA), Facultad de Psicología, Universidad de Málaga, 29071 Málaga, Spain
| | - Estela Castilla-Ortega
- Unidad de Gestión Clínica de Salud Mental, Instituto de Investigación Biomédica de Málaga (IBIMA), Hospital Regional Universitario de Málaga, 29010 Málaga, Spain
| |
Collapse
|
21
|
Glucocorticoid Homeostasis in the Dentate Gyrus Is Essential for Opiate Withdrawal-Associated Memories. Mol Neurobiol 2016; 54:6523-6541. [PMID: 27730515 DOI: 10.1007/s12035-016-0186-7] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2016] [Accepted: 09/30/2016] [Indexed: 12/11/2022]
Abstract
Drug-withdrawal-associated aversive memories might trigger relapse to drug-seeking behavior. However, changes in structural and synaptic plasticity, as well as epigenetic mechanisms, which may be critical for long-term aversive memory, have yet to be elucidated. We used male Wistar rats and performed conditioned-place aversion (CPA) paradigm to uncover the role of glucocorticoids (GCs) on plasticity-related processes that occur within the dentate gyrus (DG) during opiate-withdrawal conditioning (memory formation-consolidation) and after reactivation by re-exposure to the conditioned environment (memory retrieval). Rats subjected to conditioned morphine-withdrawal robustly expressed CPA, while adrenalectomy impaired naloxone-induced CPA. Importantly, while activity-regulated cytoskeletal-associated protein (Arc) expression was induced in sham- and ADX-dependent animals during the conditioning phase, Arc and early growth response 1 (Egr-1) induction was restricted to sham-dependent rats following memory retrieval. Moreover, we found a correlation between Arc induction and CPA score, and Arc was selectively expressed in the granular zone of the DG in dopaminoceptive, glutamatergic and GABAergic neurons. We further found that brain-derived neurotrophic factor was regulated in the opposite way during the test phase. Our results also suggest a role for epigenetic regulation on the expression of glucocorticoid receptors and Arc following memory retrieval. Our data provide the first evidence that GC homeostasis is important for the expression of long-term morphine-withdrawal memories. Moreover, our results support the idea that targeting Arc and Egr-1 in the DG may provide important insights into the role of these signaling cascades in withdrawal-context memory re-consolidation. Together, disrupting these processes in the DG might lead to effective treatments in drug addiction thereby rapidly and persistently reducing invasive memories and subsequent drug seeking.
Collapse
|
22
|
McKinnon MC, Boyd JE, Frewen PA, Lanius UF, Jetly R, Richardson JD, Lanius RA. A review of the relation between dissociation, memory, executive functioning and social cognition in military members and civilians with neuropsychiatric conditions. Neuropsychologia 2016; 90:210-34. [DOI: 10.1016/j.neuropsychologia.2016.07.017] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2016] [Revised: 06/16/2016] [Accepted: 07/16/2016] [Indexed: 01/01/2023]
|
23
|
|
24
|
Effect of Opioid on Adult Hippocampal Neurogenesis. ScientificWorldJournal 2016; 2016:2601264. [PMID: 27127799 PMCID: PMC4835638 DOI: 10.1155/2016/2601264] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2015] [Accepted: 03/17/2016] [Indexed: 11/18/2022] Open
Abstract
During the past decade, the study of the mechanisms and functional implications of adult neurogenesis has significantly progressed. Many studies focus on the factors that regulate proliferation and fate determination of adult neural stem/progenitor cells, including addictive drugs such as opioid. Here, we review the most recent works on opiate drugs' effect on different developmental stages of adult hippocampal neurogenesis, as well as the possible underlying mechanisms. We conclude that opiate drugs in general cause a loss of newly born neural progenitors in the subgranular zone of dentate gyrus, by either modulating proliferation or interfering with differentiation and maturation. We also discuss the consequent impact of regulation of adult neurogenesis in animal's opioid addiction behavior. We further look into the future directions in studying the convergence between the adult neurogenesis field and opioid addiction field, since the adult-born granular cells were shown to play a role in neuroplasticity and may help to reduce the vulnerability to drug craving and relapse.
Collapse
|
25
|
Dholakiya SL, Aliberti A, Barile FA. Morphine sulfate concomitantly decreases neuronal differentiation and opioid receptor expression in mouse embryonic stem cells. Toxicol Lett 2016; 247:45-55. [DOI: 10.1016/j.toxlet.2016.01.010] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2015] [Revised: 12/21/2015] [Accepted: 01/17/2016] [Indexed: 01/20/2023]
|
26
|
Bayer R, Franke H, Ficker C, Richter M, Lessig R, Büttner A, Weber M. Alterations of neuronal precursor cells in stages of human adult neurogenesis in heroin addicts. Drug Alcohol Depend 2015; 156:139-149. [PMID: 26416695 DOI: 10.1016/j.drugalcdep.2015.09.005] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/05/2015] [Revised: 08/31/2015] [Accepted: 09/04/2015] [Indexed: 11/29/2022]
Abstract
BACKGROUND Adult neurogenesis has been shown to occur throughout life and different brain pathologies were demonstrated to be associated with altered neurogenesis. Here, an impact of heroin addiction on neurogenesis in humans is hypothesised. METHODS Post mortem hippocampal specimens of drug addicts with known heroin abuse and a group of non-addictive control subjects were analysed, using antibodies indicating different stages of neurogenesis. The subgranular zone of the dentate gyrus was examined qualitatively and quantitatively. RESULTS The data indicate (i) a decreased number of neural precursor cells, (ii) accompanied by low rates of proliferation and (iii) a marked loss of dendritic trees in targeting cells in heroin fatalities. (iv) The age-dependent increase of differentiating cells in the healthy controls was not observed in the addicts. Additionally, double immunofluorescence labelling indicated the precursor nature of Musashi-1 positive cells in the human subgranular zone of the dentate gyrus. CONCLUSIONS Present data firstly demonstrate the influence of drug addiction with known heroin abuse on different developmental stages of progenitors in the dentate gyrus. The patterns of antibody staining suggest a distinct inhibition of neurogenesis at the stage of neural precursor cells and revealed morphological changes in targeting cells in cases of heroin addicts as compared to healthy controls. These alterations could be considerable for memory and cognitive deficits as well as addictive behaviour in chronic drug abusers and may give rise to specific pro-neurogenic therapies.
Collapse
Affiliation(s)
- Ronny Bayer
- Institute of Legal Medicine, University of Leipzig, D-04103 Leipzig, Germany
| | - Heike Franke
- Rudolf-Boehm-Institute of Pharmacology and Toxicology, University of Leipzig, D-04107 Leipzig, Germany
| | - Christoph Ficker
- Rudolf-Boehm-Institute of Pharmacology and Toxicology, University of Leipzig, D-04107 Leipzig, Germany
| | - Monique Richter
- Rudolf-Boehm-Institute of Pharmacology and Toxicology, University of Leipzig, D-04107 Leipzig, Germany
| | - Rüdiger Lessig
- Institute of Legal Medicine, University of Halle-Wittenberg, D-06112 Halle (Saale), Germany
| | - Andreas Büttner
- Institute of Forensic Medicine, Rostock University Medical Centre, D-18055 Rostock, Germany
| | - Marco Weber
- Institute of Legal Medicine, University of Halle-Wittenberg, D-06112 Halle (Saale), Germany.
| |
Collapse
|
27
|
Effects of addictive drugs on adult neural stem/progenitor cells. Cell Mol Life Sci 2015; 73:327-48. [PMID: 26468052 DOI: 10.1007/s00018-015-2067-z] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2015] [Revised: 10/04/2015] [Accepted: 10/08/2015] [Indexed: 12/18/2022]
Abstract
Neural stem/progenitor cells (NSPCs) undergo a series of developmental processes before giving rise to newborn neurons, astrocytes and oligodendrocytes in adult neurogenesis. During the past decade, the role of NSPCs has been highlighted by studies on adult neurogenesis modulated by addictive drugs. It has been proven that these drugs regulate the proliferation, differentiation and survival of adult NSPCs in different manners, which results in the varying consequences of adult neurogenesis. The effects of addictive drugs on NSPCs are exerted via a variety of different mechanisms and pathways, which interact with one another and contribute to the complexity of NSPC regulation. Here, we review the effects of different addictive drugs on NSPCs, and the related experimental methods and paradigms. We also discuss the current understanding of major signaling molecules, especially the putative common mechanisms, underlying such effects. Finally, we review the future directions of research in this area.
Collapse
|
28
|
McAdams RM, McPherson RJ, Beyer RP, Bammler TK, Farin FM, Juul SE. Dose-dependent effects of morphine exposure on mRNA and microRNA (miR) expression in hippocampus of stressed neonatal mice. PLoS One 2015; 10:e0123047. [PMID: 25844808 PMCID: PMC4386824 DOI: 10.1371/journal.pone.0123047] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2014] [Accepted: 02/18/2015] [Indexed: 12/02/2022] Open
Abstract
Morphine is used to sedate critically ill infants to treat painful or stressful conditions associated with intensive care. Whether neonatal morphine exposure affects microRNA (miR) expression and thereby alters mRNA regulation is unknown. We tested the hypothesis that repeated morphine treatment in stress-exposed neonatal mice alters hippocampal mRNA and miR expression. C57BL/6 male mice were treated from postnatal day (P) 5 to P9 with morphine sulfate at 2 or 5 mg/kg ip twice daily and then exposed to stress consisting of hypoxia (100% N2 1 min and 100% O2 5 min) followed by 2h maternal separation. Control mice were untreated and dam-reared. mRNA and miR expression profiling was performed on hippocampal tissues at P9. Overall, 2 and 5 mg/kg morphine treatment altered expression of a total of 150 transcripts (>1.5 fold change, P<0.05) from which 100 unique mRNAs were recognized (21 genes were up- and 79 genes were down-regulated), and 5 mg/kg morphine affected 63 mRNAs exclusively. The most upregulated mRNAs were fidgetin, arginine vasopressin, and resistin-like alpha, and the most down-regulated were defensin beta 11, aquaporin 1, calmodulin-like 4, chloride intracellular channel 6, and claudin 2. Gene Set Enrichment Analysis revealed that morphine treatment affected pathways related to cell cycle, membrane function, signaling, metabolism, cell death, transcriptional regulation, and immune response. Morphine decreased expression of miR-204-5p, miR-455-3p, miR-448-5p, and miR-574-3p. Nine morphine-responsive mRNAs that are involved in neurodevelopment, neurotransmission, and inflammation are predicted targets of the aforementioned differentially expressed miRs. These data establish that morphine produces dose-dependent changes in both hippocampal mRNA and miR expression in stressed neonatal mice. If permanent, morphine–mediated neuroepigenetic effects may affect long-term hippocampal function, and this provides a mechanism for the neonatal morphine-related impairment of adult learning.
Collapse
Affiliation(s)
- Ryan M. McAdams
- Department of Pediatrics, Division of Neonatology, University of Washington, Seattle, Washington, United States of America
- * E-mail:
| | - Ronald J. McPherson
- Department of Pediatrics, Division of Neonatology, University of Washington, Seattle, Washington, United States of America
| | - Richard P. Beyer
- Dept of Environmental & Occupational Health Sciences, University of Washington, Seattle, Washington, United States of America
| | - Theo K. Bammler
- Dept of Environmental & Occupational Health Sciences, University of Washington, Seattle, Washington, United States of America
| | - Frederico M. Farin
- Dept of Environmental & Occupational Health Sciences, University of Washington, Seattle, Washington, United States of America
| | - Sandra E. Juul
- Department of Pediatrics, Division of Neonatology, University of Washington, Seattle, Washington, United States of America
| |
Collapse
|
29
|
Zhang Y, Brownstein AJ, Buonora M, Niikura K, Ho A, Correa da Rosa J, Kreek MJ, Ott J. Self administration of oxycodone alters synaptic plasticity gene expression in the hippocampus differentially in male adolescent and adult mice. Neuroscience 2014; 285:34-46. [PMID: 25446355 DOI: 10.1016/j.neuroscience.2014.11.013] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2014] [Revised: 10/19/2014] [Accepted: 11/04/2014] [Indexed: 11/16/2022]
Abstract
Abuse and addiction to prescription opioids such as oxycodone (a short-acting Mu opioid receptor (MOP-r) agonist) in adolescence is a pressing public health issue. We have previously shown differences in oxycodone self-administration behaviors between adolescent and adult C57BL/6J mice and expression of striatal neurotransmitter receptor genes, in areas involved in reward. In this study, we aimed to determine whether oxycodone self-administration differentially affects genes regulating synaptic plasticity in the hippocampus of adolescent compared to adult mice, since the hippocampus may be involved in learning aspects associated with chronic drug self administration. Hippocampus was isolated for mRNA analysis from mice that had self administered oxycodone (0.25 mg/kg/infusion) 2h/day for 14 consecutive days or from yoked saline controls. Gene expression was analyzed with real-time polymerase chain reaction (PCR) using a commercially available "synaptic plasticity" PCR array containing 84 genes. We found that adolescent and adult control mice significantly differed in the expression of several genes in the absence of oxycodone exposure, including those coding for mitogen-activated protein kinase, calcium/calmodulin-dependent protein kinase II gamma subunit, glutamate receptor, ionotropic AMPA2 and metabotropic 5. Chronic oxycodone self administration increased proviral integration site 1 (Pim1) and thymoma viral proto-oncogene 1 mRNA levels compared to controls in both age groups. Both Pim1 and cadherin 2 mRNAs showed a significant combined effect of Drug Condition and Age × Drug Condition. Furthermore, the mRNA levels of both cadherin 2 and cAMP response element modulators showed an experiment-wise significant difference between oxycodone and saline control in adult but not in adolescent mice. Overall, this study demonstrates for the first time that chronic oxycodone self-administration differentially alters synaptic plasticity gene expression in the hippocampus of adolescent and adult mice.
Collapse
Affiliation(s)
- Y Zhang
- The Laboratory of the Biology of Addictive Diseases, The Rockefeller University, New York, NY 10065, USA.
| | - A J Brownstein
- The Laboratory of the Biology of Addictive Diseases, The Rockefeller University, New York, NY 10065, USA
| | - M Buonora
- The Laboratory of the Biology of Addictive Diseases, The Rockefeller University, New York, NY 10065, USA
| | - K Niikura
- The Laboratory of the Biology of Addictive Diseases, The Rockefeller University, New York, NY 10065, USA
| | - A Ho
- The Laboratory of the Biology of Addictive Diseases, The Rockefeller University, New York, NY 10065, USA
| | - J Correa da Rosa
- Laboratory of Investigative Dermatology, The Rockefeller University, New York, NY 10065, USA
| | - M J Kreek
- The Laboratory of the Biology of Addictive Diseases, The Rockefeller University, New York, NY 10065, USA
| | - J Ott
- Institute of Psychology, Chinese Academy of Sciences, Beijing, China; The Laboratory of Statistical Genetics, The Rockefeller University, New York, NY, USA
| |
Collapse
|
30
|
Loss of the mu opioid receptor induces strain-specific alterations in hippocampal neurogenesis and spatial learning. Neuroscience 2014; 278:11-9. [PMID: 25086317 DOI: 10.1016/j.neuroscience.2014.07.039] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2013] [Revised: 07/09/2014] [Accepted: 07/09/2014] [Indexed: 12/27/2022]
Abstract
Alterations in hippocampal neurogenesis affect spatial learning, though, the relative contributions of cell proliferation and cell survival on this process are poorly understood. The current study utilized mu opioid receptor (MOR-1) knockout (KO) mice on two background strains, C57BL/6 and 129S6, to assess cell survival as well as determine the impact on spatial learning using the Morris water maze. These experiments were designed to extend prior work showing that both C57BL/6 and 129S6 MOR-1 KO mice have an increased number of proliferating cells in the dentate gyrus (DG) when compared to wild-type (WT) mice. The current study indicates that newly born neurons in the DG of C57BL/6 MOR-1 KO mice exhibit enhanced survival when compared to WT mice, while new neurons in the DG of 129S6 MOR-1 KO mice do not. In addition, C57BL/6 MOR-1 KO mice have a lower number of apoptotic cells in the DG compared to WT mice while, in contrast, 129S6 MOR-1 KO mice have a higher number of apoptotic cells in this region. These alterations collectively contribute to an increase in the granule cell number in the DG of C57BL/6 MOR-1 KO mice, while the total number of granule cells in 129S6 MOR-1 KO mice is unchanged. Thus, although C57BL/6 and 129S6 MOR-1 KO mice both exhibit increased cell proliferation in the DG, the impact of the MOR-1 mutation on cell survival differs between strains. Furthermore, the decrease in DG cell survival displayed by 129S6 MOR-1 KO mice is correlated with functional deficits in spatial learning, suggesting that MOR-1-dependent alterations in the survival of new neurons in the DG, and not MOR-1-dependent changes in proliferation of progenitor cells in the DG, is important for spatial learning.
Collapse
|
31
|
Short term morphine exposure in vitro alters proliferation and differentiation of neural progenitor cells and promotes apoptosis via mu receptors. PLoS One 2014; 9:e103043. [PMID: 25072277 PMCID: PMC4114742 DOI: 10.1371/journal.pone.0103043] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2013] [Accepted: 06/26/2014] [Indexed: 11/19/2022] Open
Abstract
BACKGROUND Chronic morphine treatment inhibits neural progenitor cell (NPC) progression and negatively effects hippocampal neurogenesis. However, the effect of acute opioid treatment on cell development and its influence on NPC differentiation and proliferation in vitro is unknown. We aim to investigate the effect of a single, short term exposure of morphine on the proliferation, differentiation and apoptosis of NPCs and the mechanism involved. METHODS Cell cultures from 14-day mouse embryos were exposed to different concentrations of morphine and its antagonist naloxone for 24 hours and proliferation, differentiation and apoptosis were studied. Proliferating cells were labeled with bromodeoxyuridine (BrdU) and cell fate was studied with immunocytochemistry. RESULTS Cells treated with morphine demonstrated decreased BrdU expression with increased morphine concentrations. Analysis of double-labeled cells showed a decrease in cells co-stained for BrdU with nestin and an increase in cells co-stained with BrdU and neuron-specific class III β-tubuline (TUJ1) in a dose dependent manner. Furthermore, a significant increase in caspase-3 activity was observed in the nestin- positive cells. Addition of naloxone to morphine-treated NPCs reversed the anti-proliferative and pro-apoptotic effects of morphine. CONCLUSIONS Short term morphine exposure induced inhibition of NPC proliferation and increased active caspase-3 expression in a dose dependent manner. Morphine induces neuronal and glial differentiation and decreases the expression of nestin- positive cells. These effects were reversed with the addition of the opioid antagonist naloxone. Our results demonstrate the effects of short term morphine administration on the proliferation and differentiation of NPCs and imply a mu-receptor mechanism in the regulation of NPC survival.
Collapse
|
32
|
Meneghini V, Cuccurazzu B, Bortolotto V, Ramazzotti V, Ubezio F, Tzschentke TM, Canonico PL, Grilli M. The Noradrenergic Component in Tapentadol Action Counteracts μ-Opioid Receptor–Mediated Adverse Effects on Adult Neurogenesis. Mol Pharmacol 2014; 85:658-70. [DOI: 10.1124/mol.113.091520] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
|
33
|
Malik S, Saha R, Seth P. Involvement of extracellular signal-regulated kinase (ERK1/2)-p53-p21 axis in mediating neural stem/progenitor cell cycle arrest in co-morbid HIV-drug abuse exposure. J Neuroimmune Pharmacol 2014; 9:340-53. [PMID: 24469921 DOI: 10.1007/s11481-014-9523-7] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2013] [Accepted: 01/14/2014] [Indexed: 10/25/2022]
Abstract
Neurological complications in opioid abusing Human Immunodeficiency Virus-1 (HIV-1) patients suggest enhanced neurodegeneration as compared to non-drug abusing HIV-1 infected population. Neural precursor cells (NPCs), the multipotent cells of the mammalian brain, are susceptible to HIV-1 infection and as opiates also perturb their growth kinetics, detailed mechanistic studies for their co-morbid exposure are highly warranted. Using a well characterized in vitro model of human fetal brain-derived neural precursor cells, we investigated alterations in NPC properties at both acute and chronic durations. Chronic morphine and Tat treatment attenuated proliferation in NPCs, with cells stalled at G1-phase of the cell cycle. Furthermore HIV-Tat and morphine exposure increased activation of extracellular signal-regulated kinase-1/2 (ERK1/2), enhanced levels of p53 and p21, and decreased cyclin D1 and Akt levels in NPCs. Regulated by ERK1/2 and p53, p21 was found to be indispensible for Tat and morphine mediated cell cycle arrest. Our study elaborates on the cellular and molecular machinery in NPCs and provides significant mechanistic details into HIV-drug abuse co-morbidity that may have far reaching clinical consequences both in pediatric as well as adult neuroAIDS.
Collapse
Affiliation(s)
- Shaily Malik
- Cellular and Molecular Neuroscience, National Brain Research Centre (NBRC), NH-8, Nainwal Road, Manesar, Gurgaon, Haryana, 122051, India
| | | | | |
Collapse
|
34
|
Jahanshahi M, Shaabani R, Nikmahzar EG, Babakordi F. Female rat hippocampal cell density after conditioned place preference. Folia Biol (Praha) 2014; 60:47-51. [PMID: 24594056 DOI: 10.14712/fb2014060010047] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2024]
Abstract
The hippocampus is important for learning tasks, such as conditioned place preference (CPP), which is widely used as a model for studying the reinforcing effects of drugs with dependence liability. Long-term opiate use may produce maladaptive plasticity in the brain structures involved in learning and memory, such as the hippocampus. We investigated the phenomenon of conditioning with morphine on the cell density of female rat hippocampus. Forty-eight female Wistar rats weighing on average 200-250 g were used. Rats were distributed into eight groups. Experimental groups received morphine daily (three days) at different doses (2.5, 5, 7.5 mg/kg) and the control-saline group received normal saline (1 ml/kg), and then the CPP test was performed. Three sham groups received only different doses (2.5, 5, 7.5 mg/kg) of morphine without CPP test. Forty-eight hours after behavioural testing animals were decapitated under chloroform anaesthesia and their brains were fixed, and after tissue processing, slices were stained with cresyl violet for neurons and phosphotungstic acid haematoxylin for astrocytes. The maximum response was obtained with 5 mg/kg of morphine. The density of neurons in CA1 and CA3 areas of hippocampus after injection of morphine and CPP was decreased. The number of astrocytes in different areas of hippocampus was increased after injection of morphine and CPP. It seems that the effective dose was 5 mg/kg, as it led to the CPP. We concluded that both injection of mor phine and CPP can decrease the density of neurons and also increase the number of astrocytes in the rat hippocampus.
Collapse
Affiliation(s)
- M Jahanshahi
- Department of Anatomy, Neuroscience Research Centre, Faculty of Medicine, Golestan University of Medical Sciences, Gorgan, Iran
| | - R Shaabani
- Department of Anatomy, Neuroscience Research Centre, Faculty of Medicine, Golestan University of Medical Sciences, Gorgan, Iran
| | - E G Nikmahzar
- Department of Anatomy, Neuroscience Research Centre, Faculty of Medicine, Golestan University of Medical Sciences, Gorgan, Iran
| | - F Babakordi
- Department of Experimental and Clinical Rheumatology, Institute of Rheumatology, Prague, Czech Republic
| |
Collapse
|
35
|
Chambers RA. Adult hippocampal neurogenesis in the pathogenesis of addiction and dual diagnosis disorders. Drug Alcohol Depend 2013; 130:1-12. [PMID: 23279925 PMCID: PMC3640791 DOI: 10.1016/j.drugalcdep.2012.12.005] [Citation(s) in RCA: 71] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/28/2012] [Revised: 11/19/2012] [Accepted: 12/06/2012] [Indexed: 12/11/2022]
Abstract
BACKGROUND As knowledge deepens about how new neurons are born, differentiate, and wire into the adult mammalian brain, growing evidence depicts hippocampal neurogenesis as a special form of neuroplasticity that may be impaired across psychiatric disorders. This review provides an integrated-evidence based framework describing a neurogenic basis for addictions and addiction vulnerability in mental illness. METHODS Basic studies conducted over the last decade examining the effects of addictive drugs on adult neurogenesis and the impact of neurogenic activity on addictive behavior were compiled and integrated with relevant neurocomputational and human studies. RESULTS While suppression of hippocampal neurogenic proliferation appears to be a universal property of addictive drugs, the pathophysiology of addictions involves neuroadaptative processes within frontal-cortical-striatal motivation circuits that the neurogenic hippocampus regulates via direct projections. States of suppressed neurogenic activity may simultaneously underlie psychiatric and cognitive symptoms, but also confer or signify hippocampal dysfunction that heightens addiction vulnerability in mental illness as a basis for dual diagnosis disorders. CONCLUSIONS Research on pharmacological, behavioral and experiential strategies that enhance adaptive regulation of hippocampal neurogenesis holds potential in advancing preventative and integrative treatment strategies for addictions and dual diagnosis disorders.
Collapse
Affiliation(s)
- R Andrew Chambers
- Laboratory for Translational Neuroscience of Dual Diagnosis & Development, Department of Psychiatry, Indiana University School of Medicine, 791 Union Drive, Indianapolis, IN 46202, United States.
| |
Collapse
|
36
|
Weber M, Scherf N, Kahl T, Braumann UD, Scheibe P, Kuska JP, Bayer R, Büttner A, Franke H. Quantitative analysis of astrogliosis in drug-dependent humans. Brain Res 2013; 1500:72-87. [PMID: 23337617 DOI: 10.1016/j.brainres.2012.12.048] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2012] [Revised: 12/14/2012] [Accepted: 12/23/2012] [Indexed: 11/29/2022]
Abstract
Drug addiction is a chronic, relapsing disease caused by neurochemical and molecular changes in the brain. In this human autopsy study qualitative and quantitative changes of glial fibrillary acidic protein (GFAP)-positive astrocytes in the hippocampus of 26 lethally intoxicated drug addicts and 35 matched controls are described. The morphological characterization of these cells reflected alterations representative for astrogliosis. But, neither quantification of GFAP-positive cells nor the Western blot analysis indicated statistical significant differences between drug fatalities versus controls. However, by semi-quantitative scoring a significant shift towards higher numbers of activated astrocytes in the drug group was detected. To assess morphological changes quantitatively, graph-based representations of astrocyte morphology were obtained from single cell images captured by confocal laser scanning microscopy. Their underlying structures were used to quantify changes in astroglial fibers in an automated fashion. This morphometric analysis yielded significant differences between the investigated groups for four different measures of fiber characteristics (Euclidean distance, graph distance, number of graph elements, fiber skeleton distance), indicating that, e.g., astrocytes in drug addicts on average exhibit significant elongation of fiber structures as well as two-fold increase in GFAP-positive fibers as compared with those in controls. In conclusion, the present data show characteristic differences in morphology of hippocampal astrocytes in drug addicts versus controls and further supports the involvement of astrocytes in human pathophysiology of drug addiction. The automated quantification of astrocyte morphologies provides a novel, testable way to assess the fiber structures in a quantitative manner as opposed to standard, qualitative descriptions.
Collapse
Affiliation(s)
- Marco Weber
- Institute of Legal Medicine, University of Halle, Franzosenweg 1, 06112 Halle (Saale), Germany.
| | | | | | | | | | | | | | | | | |
Collapse
|
37
|
Therapeutic potential of histaminergic compounds in the treatment of addiction and drug-related cognitive disorders. Behav Brain Res 2013; 237:357-68. [DOI: 10.1016/j.bbr.2012.09.025] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2012] [Revised: 09/13/2012] [Accepted: 09/16/2012] [Indexed: 12/21/2022]
|
38
|
Abstract
Many tissues of the body cannot only repair themselves, but also self-renew, a property mainly due to stem cells and the various mechanisms that regulate their behavior. Stem cell biology is a relatively new field. While advances are slowly being realized, stem cells possess huge potential to ameliorate disease and counteract the aging process, causing its speculation as the next panacea. Amidst public pressure to advance rapidly to clinical trials, there is a need to understand the biology of stem cells and to support basic research programs. Without a proper comprehension of how cells and tissues are maintained during the adult life span, clinical trials are bound to fail. This review will cover the basic biology of stem cells, the various types of stem cells, their potential function, and the advantages and disadvantages to their use in medicine. We will next cover the role of G protein-coupled receptors in the regulation of stem cells and their potential in future clinical applications.
Collapse
Affiliation(s)
- VAN A. DOZE
- Department of Pharmacology, Physiology and Therapeutics, School of Medicine and Health Sciences, University of North Dakota, Grand Forks, ND 58203, USA (V.A.D.), and Department of Molecular Cardiology, Lerner Research Institute, The Cleveland Clinic Foundation, Cleveland, OH 44195, USA (D.M.P.)
| | - DIANNE M. PEREZ
- Department of Pharmacology, Physiology and Therapeutics, School of Medicine and Health Sciences, University of North Dakota, Grand Forks, ND 58203, USA (V.A.D.), and Department of Molecular Cardiology, Lerner Research Institute, The Cleveland Clinic Foundation, Cleveland, OH 44195, USA (D.M.P.)
| |
Collapse
|
39
|
Increases in doublecortin immunoreactivity in the dentate gyrus following extinction of heroin-seeking behavior. Neural Plast 2012; 2012:283829. [PMID: 23213573 PMCID: PMC3504456 DOI: 10.1155/2012/283829] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2012] [Revised: 10/03/2012] [Accepted: 10/06/2012] [Indexed: 02/02/2023] Open
Abstract
Adult-generated neurons in the dentate gyrus (DG) of the hippocampus play a role in various forms of learning and memory. However, adult born neurons in the DG, while still at an immature stage, exhibit unique electrophysiological properties and are also functionally implicated in learning and memory processes. We investigated the effects of extinction of drug-seeking behavior on the formation of immature neurons in the DG as assessed by quantification of doublecortin (DCX) immunoreactivity. Rats were allowed to self-administer heroin (0.03 mg/kg/infusion) for 12 days and then subjected either to 10 days of extinction training or forced abstinence. We also examined extinction responding patterns following heroin self-administration in glial fibrillary acidic protein thymidine kinase (GFAP-tk) transgenic mice, which have been previously demonstrated to show reduced formation of immature and mature neurons in the DG following treatment with ganciclovir (GCV). We found that extinction training increased DCX immunoreactivity in the dorsal DG as compared with animals undergoing forced abstinence, and that GCV-treated GFAP-tk mice displayed impaired extinction learning as compared to saline-treated mice. Our results suggest that extinction of drug-seeking behavior increases the formation of immature neurons in the DG and that these neurons may play a functional role in extinction learning.
Collapse
|
40
|
Abstract
The identification and functional understanding of the neurocircuitry that mediates alcohol and drug effects that are relevant for the development of addictive behavior is a fundamental challenge in addiction research. Here we introduce an assumption-free construction of a neurocircuitry that mediates acute and chronic drug effects on neurotransmitter dynamics that is solely based on rodent neuroanatomy. Two types of data were considered for constructing the neurocircuitry: (1) information on the cytoarchitecture and neurochemical connectivity of each brain region of interest obtained from different neuroanatomical techniques; (2) information on the functional relevance of each region of interest with respect to alcohol and drug effects. We used mathematical data mining and hierarchical clustering methods to achieve the highest standards in the preprocessing of these data. Using this approach, a dynamical network of high molecular and spatial resolution containing 19 brain regions and seven neurotransmitter systems was obtained. Further graph theoretical analysis suggests that the neurocircuitry is connected and cannot be separated into further components. Our analysis also reveals the existence of a principal core subcircuit comprised of nine brain regions: the prefrontal cortex, insular cortex, nucleus accumbens, hypothalamus, amygdala, thalamus, substantia nigra, ventral tegmental area and raphe nuclei. Finally, by means of algebraic criteria for synchronizability of the neurocircuitry, the suitability for in silico modeling of acute and chronic drug effects is indicated. Indeed, we introduced as an example a dynamical system for modeling the effects of acute ethanol administration in rats and obtained an increase in dopamine release in the nucleus accumbens-a hallmark of drug reinforcement-to an extent similar to that seen in numerous microdialysis studies. We conclude that the present neurocircuitry provides a structural and dynamical framework for large-scale mathematical models and will help to predict chronic drug effects on brain function.
Collapse
Affiliation(s)
- Hamid R. Noori
- Institute of Psychopharmacology, Central Institute of Mental Health, Medical Faculty Mannheim; University of Heidelberg; Mannheim; Germany
| | - Rainer Spanagel
- Institute of Psychopharmacology, Central Institute of Mental Health, Medical Faculty Mannheim; University of Heidelberg; Mannheim; Germany
| | - Anita C. Hansson
- Institute of Psychopharmacology, Central Institute of Mental Health, Medical Faculty Mannheim; University of Heidelberg; Mannheim; Germany
| |
Collapse
|
41
|
Abstract
The importance of adult neurogenesis has only recently been accepted, resulting in a completely new field of investigation within stem cell biology. The regulation and functional significance of adult neurogenesis is currently an area of highly active research. G-protein-coupled receptors (GPCRs) have emerged as potential modulators of adult neurogenesis. GPCRs represent a class of proteins with significant clinical importance, because approximately 30% of all modern therapeutic treatments target these receptors. GPCRs bind to a large class of neurotransmitters and neuromodulators such as norepinephrine, dopamine, and serotonin. Besides their typical role in cellular communication, GPCRs are expressed on adult neural stem cells and their progenitors that relay specific signals to regulate the neurogenic process. This review summarizes the field of adult neurogenesis and its methods and specifies the roles of various GPCRs and their signal transduction pathways that are involved in the regulation of adult neural stem cells and their progenitors. Current evidence supporting adult neurogenesis as a model for self-repair in neuropathologic conditions, adult neural stem cell therapeutic strategies, and potential avenues for GPCR-based therapeutics are also discussed.
Collapse
Affiliation(s)
- Van A Doze
- Department of Molecular Cardiology, NB50, Lerner Research Institute, The Cleveland Clinic Foundation, 9500 Euclid Ave., Cleveland, OH 44195, USA
| | | |
Collapse
|
42
|
Kubesova A, Bubenikova-Valesova V, Mertlova M, Palenicek T, Horacek J. Impact of psychotropic drugs on adult hippocampal neurogenesis. Neurosci Res 2012; 73:93-8. [DOI: 10.1016/j.neures.2012.02.014] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2011] [Revised: 02/10/2012] [Accepted: 02/24/2012] [Indexed: 01/17/2023]
|
43
|
Cominski TP, Turchin CE, Hsu MS, Ansonoff MA, Pintar JE. Loss of the mu opioid receptor on different genetic backgrounds leads to increased bromodeoxyuridine labeling in the dentate gyrus only after repeated injection. Neuroscience 2012; 206:49-59. [PMID: 22280973 DOI: 10.1016/j.neuroscience.2011.12.046] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2011] [Revised: 12/14/2011] [Accepted: 12/15/2011] [Indexed: 01/14/2023]
Abstract
The endogenous opioid system is involved in various physiological processes, including neurogenesis in the dentate gyrus (DG) of the hippocampus. In the current study, we investigated the role of the mu opioid receptor (MOR-1) on DG neurogenesis and measured glucocorticoid levels following several injection paradigms to supplement the neurogenesis experiments. MOR-1 knockout (KO) mice on C57BL/6 and 129S6 backgrounds were injected with bromodeoxyuridine (BrdU) using either a single injection or two different repeated injection protocols and then sacrificed at different time points. The total number of BrdU and proliferating cell nuclear antigen (PCNA) positive cells in the DG is significantly increased in MOR-1 KO mice compared with wild type (WT) on both strains after repeated injection, but not after a single injection. Plasma corticosterone (CORT) levels increased similarly in MOR-1 KO and WT mice following both single and repeated injection, indicating that the stress response is activated following any injection protocol, but that the mechanism responsible for the increase in BrdU labeling in MOR-1 KO mice is CORT-level independent. Finally, WT 129S6 mice, independent of genotype, showed higher levels of plasma CORT compared with WT C57BL/6 mice in both noninjected controls and following injection at two separate time points; these levels were inversely correlated with low numbers of BrdU cells in the DG in 129S6 mice compared with C57BL/6 mice. In summary, these data demonstrate that loss of MOR-1 increases BrdU labeling in the DG independent of CORT levels, but only following a repeated injection, illustrating the capability of injection paradigms to influence cell-proliferative responses in a genotype-dependent manner.
Collapse
Affiliation(s)
- T P Cominski
- Department of Neuroscience, Cell Biology, University of Medicine and Dentistry of New Jersey-Robert, Wood Johnson Medical School (UMDNJ/RWJMS) 675 Hoes Lane, RWJMS-SPH, Room 352, Piscataway, NJ, USA
| | | | | | | | | |
Collapse
|
44
|
Canales JJ. Deficient plasticity in the hippocampus and the spiral of addiction: focus on adult neurogenesis. Curr Top Behav Neurosci 2012; 15:293-312. [PMID: 22976276 DOI: 10.1007/7854_2012_230] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023]
Abstract
Addiction is a complex neuropsychiatric disorder which causes disruption at multiple levels, including cognitive, emotional, and behavioral domains. Traditional biological theories of addiction have focused on the mesolimbic dopamine pathway and the nucleus accumbens as anatomical substrates mediating addictive-like behaviors. More recently, we have begun to recognize the engagement and dynamic influence of a much broader circuitry which encompasses the frontal cortex, the amygdala, and the hippocampus. In particular, neurogenesis in the adult hippocampus has become a major focus of attention due to its ability to influence memory, motivation, and affect, all of which are disrupted in addiction. First, I summarize toxicological data that reveal strongly suppressive effects of drug exposure on adult hippocampal neurogenesis. Then, I discuss the impact of deficient neurogenesis on learning and memory function, stress responsiveness and affective behavior, as they relate to addiction. Finally, I examine recent behavioral observations that implicate neurogenesis in the adult hippocampus in the emergence and maintenance of addictive behavior. The evidence reviewed here suggests that deficient neurogenesis is associated with several components of the downward spiraling loop that characterizes addiction, including elevated sensitivity to drug-induced reward and reinforcement, enhanced neurohormonal responsiveness, emergence of a negative affective state, memory impairment, and inflexible behavior.
Collapse
Affiliation(s)
- Juan J Canales
- Behavioural Neuroscience, Department of Psychology, The University of Canterbury, Private Bag 4800, 8140, Christchurch, New Zealand,
| |
Collapse
|
45
|
Drug withdrawal-induced depression: Serotonergic and plasticity changes in animal models. Neurosci Biobehav Rev 2012; 36:696-726. [DOI: 10.1016/j.neubiorev.2011.10.003] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2011] [Revised: 10/06/2011] [Accepted: 10/15/2011] [Indexed: 12/17/2022]
|
46
|
Pettit AS, Desroches R, Bennett SAL. The opiate analgesic buprenorphine decreases proliferation of adult hippocampal neuroblasts and increases survival of their progeny. Neuroscience 2011; 200:211-22. [PMID: 22079577 DOI: 10.1016/j.neuroscience.2011.10.039] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2011] [Revised: 10/15/2011] [Accepted: 10/24/2011] [Indexed: 01/10/2023]
Abstract
Although opiate drugs of abuse have been shown to decrease adult hippocampal neurogenesis, the impact of opiate analgesics has not been tested. North American regulatory boards governing the ethical treatment of experimental animals require the administration of analgesics, such as buprenorphine, following minor surgical interventions. Here, we show that two commonly used post-operative buprenorphine dosing regimes significantly inhibit the proliferation of doublecortin-positive neuroblasts but not other hippocampal stem and progenitor cell populations in adult mice. Buprenorphine, administered in schedules of three 0.05 mg/kg subcutaneous injections over a single day or seven 0.05 mg/kg injections over a 3-day period decreased the number of actively proliferating 5-iodo-2'-deoxyuridine-labeled doublecortin-positive cells for up to 6 days after opiate withdrawal. The minimal (three injection), but not standard (seven injection), analgesic paradigm also reduced basal indices of hippocampal progenitor cell apoptosis and enhanced survival of newly born cells for up to 28 days. Taken together, these data provide the first evidence that the routine administration of opiate analgesics has transient but long-lasting effects on neurogenesis and further emphasize that analgesic dosage and schedule should be reported and considered when interpreting the magnitude of neural stem and progenitor cell activation in response to in vivo intervention.
Collapse
Affiliation(s)
- A S Pettit
- Neural Regeneration Laboratory and Ottawa Institute of Systems Biology, Department of Biochemistry, Microbiology and Immunology, University of Ottawa, ON, Canada
| | | | | |
Collapse
|
47
|
Li K, He HT, Li HM, Liu JK, Fu HY, Hong M. Heroin affects purine nucleotides metabolism in rat brain. Neurochem Int 2011; 59:1104-8. [PMID: 22019714 DOI: 10.1016/j.neuint.2011.10.001] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2011] [Revised: 09/30/2011] [Accepted: 10/04/2011] [Indexed: 11/27/2022]
Abstract
OBJECTIVE To explore the effects of heroin on purine nucleotides metabolism in rat brain. METHODS Biochemical changes in association with heroin administration were compared between heroin-administered rats and non-heroin rats. HPLC method was used to detect the absolute content of purine nucleotides in brain tissues. Concentrations of uric acid (UA), blood urea nitrogen (BUN) and creatinine (Cre) in plasma were measured. Enzymatic activities of adenosine deaminase (ADA) and xanthine oxidase (XO) in brain tissue were analyzed. Real-time PCR was used to determine the relative level of transcripts of ADA, XO, adenine phosphoribosyl transferase (APRT), hypoxanthine-guaninephosphoribosyl transferase (HGPRT) and adenosine kinase (AK) in brain tissue. RESULTS Compared with those in the saline group, the content of AMP and GTP of heroin group decreased significantly; the UA concentration in plasma, ADA and XO activities and the mRNA level of ADA and XO in brain tissues in heroin group increased significantly; the mRNA level of AK, APRT and HGPRT in brain tissues in heroin group decreased significantly (P<0.01). CONCLUSION Heroin administration may enhance the catabolism and inhibit the anabolism of purine nucleotides in brain. There may be a deficiency of purine nucleotides, especially GTP and AMP in rat brain exposed to heroin. Our findings may provide a new potential approach to study the mechanism of heroin addiction.
Collapse
Affiliation(s)
- Kun Li
- School of Nursing, Jilin University, 965 Xinjiang Street, Changchun, Jilin Province, China
| | | | | | | | | | | |
Collapse
|
48
|
Shoae-Hassani A, Sharif S, Tabatabaei SAM, Verdi J. Could the endogenous opioid, morphine, prevent neural stem cell proliferation? Med Hypotheses 2011; 76:225-9. [DOI: 10.1016/j.mehy.2010.10.002] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2010] [Revised: 09/18/2010] [Accepted: 10/08/2010] [Indexed: 01/19/2023]
|
49
|
Lu XF, Li YY, Wang CG, Wei JQ, Ye Y, Zhang LC, Cao JL. Substance P in the cerebrospinal fluid-contacting nucleus contributes to morphine physical dependence in rats. Neurosci Lett 2011; 488:188-92. [DOI: 10.1016/j.neulet.2010.11.026] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2010] [Revised: 11/08/2010] [Accepted: 11/09/2010] [Indexed: 10/18/2022]
|
50
|
Reyes ARS, Levenson R, Berrettini W, Van Bockstaele EJ. Ultrastructural relationship between the mu opioid receptor and its interacting protein, GPR177, in striatal neurons. Brain Res 2010; 1358:71-80. [PMID: 20813097 PMCID: PMC2956578 DOI: 10.1016/j.brainres.2010.08.080] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2010] [Revised: 08/20/2010] [Accepted: 08/24/2010] [Indexed: 11/17/2022]
Abstract
GPR177, the mammalian ortholog of Drosophila Wntless/Evi/Sprinter, was recently identified as a novel mu-opioid receptor (MOR) interacting protein. GPR177 is a trans-membrane protein pivotal to mediating the secretion of Wnt signaling proteins. Wnt proteins, in turn, are essential in regulating neuronal development, a phenomenon inhibited upon chronic exposure to MOR agonists such as morphine and heroin. We previously showed that GPR177 and MOR are co-localized in the mouse dorsolateral striatum; however, the nature of this interaction was not fully elucidated. Therefore, in the present study, we examined cellular substrates for interactions between GPR177 and MOR using a combined immunogold-silver and peroxidase detection approach in coronal sections in the dorsolateral segment of the striatum. Semi-quantitative analysis of the ultrastructural distribution of GPR177 and MOR in striatal somata and in dendritic processes showed that, of the somata and dendritic processes exhibiting GPR177, 32% contained MOR immunolabeling while for profiles exhibiting MOR, 37% also contained GPR177 immunoreactivity. GPR177-labeled particles were localized predominantly along both the plasma membrane and within the cytoplasm of MOR-labeled dendrites. Somata and dendritic processes that contained both GPR177 and MOR more often received symmetric (inhibitory-type) synapses from unlabeled axon terminals. To further define the phenotype of GPR177 and MOR-containing cellular profiles, triple immunofluorescence detection showed that GPR177 and MOR are localized in neurons containing the opioid peptide, enkephalin, within the dorsolateral striatum. The results provide an anatomical substrate for interactions between MOR and its interacting protein, GPR177, in striatal opioid-containing neurons that may underlie the morphological alterations produced in neurons by chronic opiate use.
Collapse
Affiliation(s)
- Arith-Ruth S. Reyes
- Department of Neuroscience, Farber Institute for Neurosciences, Thomas Jefferson University, Philadelphia, Pennsylvania 19107
| | - Robert Levenson
- Department of Pharmacology, Penn State College of Medicine, Hershey, PA 17033
| | - Wade Berrettini
- Department of Psychiatry, Center for Neurobiology and Behavior, University of Pennsylvania School of Medicine, Philadelphia, PA 19104
| | - Elisabeth J. Van Bockstaele
- Department of Neuroscience, Farber Institute for Neurosciences, Thomas Jefferson University, Philadelphia, Pennsylvania 19107
| |
Collapse
|