1
|
Kolik LG, Konstantinipolsky MA, Nikolaev SV, Logvinov IO, Antipova TA, Gudasheva TA. Low-Molecular Neurotrophin-3 Mimetics with Different Patterns of Postreceptor Signaling Activation Attenuate Differentially Morphine Withdrawal in Rats. BIOCHEMISTRY. BIOKHIMIIA 2024; 89:1961-1969. [PMID: 39647825 DOI: 10.1134/s0006297924110105] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/04/2024] [Revised: 07/17/2024] [Accepted: 07/18/2024] [Indexed: 12/10/2024]
Abstract
The accumulated evidence suggests that varying levels of tyrosine kinase receptor signaling pathway activity may regulate opiate-associated neuroadaptation of noradrenergic system. Neurotrophin-3 (NT-3) interacts with tropomyosin receptor kinases (TRKs), binding mainly to TRKC receptors, which are expressed within noradrenergic neurons in the blue spot (locus coeruleus, LC). Considering the difficulties in delivering full-length neurotrophins to the CNS after systemic administration, low-molecular mimetics of loop 4 in NT-3, hexamethylenediamide bis-(N-monosuccinyl-L-asparaginyl-L-asparagine) (GTS-301), and hexamethylenediamide bis-(N-γ-oxybutyryl-L-glutamyl-L-asparagine) (GTS-302), activating TRKC and TRKB receptors, were synthesized. The aim of the study is comparative examination of the effects of NT-3 dipeptide mimetics on the signs of morphine withdrawal in outbred white rats with opiate dependence, as well as investigation of activation of postreceptor signaling pathways by the mimetics. Dipeptides GTS-301 and GTS-302 after acute administration at doses of 0.1, 1.0, and 10.0 mg/kg (i.p., intraperitoneal) had a dose-dependent effect on the specific morphine withdrawal symptoms with the most effective dose being 1.0 mg/kg. Maximum decrease in the total index of morphine withdrawal syndrome for GTS-301 was 31.3% and for GTS-302 - 41.4%. Unlike GTS-301, GTS-302 weakened mechanical allodynia induced by morphine withdrawal, reducing tactile sensitivity. When studying activation of the postreceptor signaling pathways by the NT-3 mimetics in the HT-22 hippocampal cell culture, a different pattern of postreceptor signaling was shown: GTS-302 (10-6 M), similar to NT-3, activates all three MAPK/ERK, PI3K/AKT/mTOR, and PLCγ1 pathways, while GTS-301 (10-6 M) triggers only MAPK/ERK and PLCγ1 pathways. Thus, the identified features of attenuation of the morphine withdrawal syndrome in the rats under GTS-301 and GTS-302 effects could be associated with different activation pattern of the postreceptor pathways.
Collapse
Affiliation(s)
- Larisa G Kolik
- Federal Research Center for Innovator and Emerging Biomedical and Pharmaceutical Technologies, Moscow, 125315, Russia.
| | - Mark A Konstantinipolsky
- Federal Research Center for Innovator and Emerging Biomedical and Pharmaceutical Technologies, Moscow, 125315, Russia
| | - Sergey V Nikolaev
- Federal Research Center for Innovator and Emerging Biomedical and Pharmaceutical Technologies, Moscow, 125315, Russia
| | - Ilya O Logvinov
- Federal Research Center for Innovator and Emerging Biomedical and Pharmaceutical Technologies, Moscow, 125315, Russia
| | - Tatyana A Antipova
- Federal Research Center for Innovator and Emerging Biomedical and Pharmaceutical Technologies, Moscow, 125315, Russia
| | - Tatiana A Gudasheva
- Federal Research Center for Innovator and Emerging Biomedical and Pharmaceutical Technologies, Moscow, 125315, Russia
| |
Collapse
|
2
|
Mancusi G, Miuli A, Santorelli M, Cavallotto C, Susini O, Pernaci G, Výborová E, Rosa I, d'Onofrio AM, Camardese G, Pettorruso M, Sensi SL, Martinotti G. Exploring peripheral biomarkers in psychostimulant use: A systematic review on neurotrophins, stress-related hormones, oxidative stress molecules and genetic factors. Behav Brain Res 2024; 469:115046. [PMID: 38761859 DOI: 10.1016/j.bbr.2024.115046] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2024] [Revised: 05/07/2024] [Accepted: 05/08/2024] [Indexed: 05/20/2024]
Abstract
BACKGROUND This systematic review aims to comprehensively explore the impact of psychostimulant substances on neurotrophic and inflammatory pathways, including brain-derived neurotrophic factor (BDNF), pro-BDNF, cortisol, dehydroepiandrosterone sulfate (DHEAS), thiobarbituric acid reactive substances (TBARS), interleukins, and the role of genetic factors. The study seeks to address existing gaps in the literature by providing a thorough evaluation of neurotrophic and inflammatory system alterations associated with different stages of psychostimulant dependence for a more nuanced understanding of substance use disorder (SUD) neurobiology. METHODS A systematic review was conducted in PubMed, Scopus, and Web of Science databases following the PRISMA guidelines. The research encompasses 50 studies with a participant pool totaling 6792 individuals using psychostimulant substances. RESULTS Key findings include diverse impacts of cocaine on BDNF levels, mainly consisting of their significant increase during withdrawal. In contrast, NGF showed an opposite behavior, reducing during withdrawal. Cortisol and DHEAS levels exhibited relevant increases after psychostimulant use, while TBARS showed conflicting results. Genetic investigations predominantly focused on the Val66Met polymorphism of the BDNF gene, revealing associations with susceptibility to stimulant addiction. CONCLUSIONS Neurotrophins and inflammatory molecules play a significant role in the pathophysiological mechanisms following psychostimulant use. A better understanding of their complex interplay could aid clinicians in identifying biomarkers of different disease stages. Moreover, clinical interventions designed to interfere with neurotrophic and inflammatory pathways could possibly lead to craving-modulatory strategies and reduce pathological neuronal and systemic consequences of psychostimulant use.
Collapse
Affiliation(s)
- Gianluca Mancusi
- Department of Neurosciences, Imaging and Clinical Sciences, Università degli Studi G. D'Annunzio, Chieti, Italy
| | - Andrea Miuli
- Department of Neurosciences, Imaging and Clinical Sciences, Università degli Studi G. D'Annunzio, Chieti, Italy; Department of Mental Health, ASL 2 Abruzzo Lanciano-Vasto-Chieti, Chieti, Italy.
| | - Mario Santorelli
- Department of Brain and Behavioral Science, University of Pavia, Italy
| | - Clara Cavallotto
- Department of Neurosciences, Imaging and Clinical Sciences, Università degli Studi G. D'Annunzio, Chieti, Italy
| | - Ottavia Susini
- Department of Neurosciences, Imaging and Clinical Sciences, Università degli Studi G. D'Annunzio, Chieti, Italy
| | - Giulia Pernaci
- Department of Neurosciences, Imaging and Clinical Sciences, Università degli Studi G. D'Annunzio, Chieti, Italy
| | - Eliška Výborová
- Department of Neurosciences, Imaging and Clinical Sciences, Università degli Studi G. D'Annunzio, Chieti, Italy
| | - Ilenia Rosa
- Department of Neurosciences, Imaging and Clinical Sciences, Università degli Studi G. D'Annunzio, Chieti, Italy
| | - Antonio Maria d'Onofrio
- Institute of Psychiatry and Clinical Psychology, Catholic University of Sacred Heart, Rome, Italy
| | - Giovanni Camardese
- Institute of Psychiatry and Clinical Psychology, Catholic University of Sacred Heart, Rome, Italy
| | - Mauro Pettorruso
- Department of Neurosciences, Imaging and Clinical Sciences, Università degli Studi G. D'Annunzio, Chieti, Italy; Department of Mental Health, ASL 2 Abruzzo Lanciano-Vasto-Chieti, Chieti, Italy
| | - Stefano L Sensi
- Department of Neurosciences, Imaging and Clinical Sciences, Università degli Studi G. D'Annunzio, Chieti, Italy; Center for Advanced Studies and Technology-CAST, and Institute for Advanced Biotechnology (ITAB), University G. d'Annunzio of Chieti-Pescara, Chieti 66013, Italy
| | - Giovanni Martinotti
- Department of Neurosciences, Imaging and Clinical Sciences, Università degli Studi G. D'Annunzio, Chieti, Italy; Department of Mental Health, ASL 2 Abruzzo Lanciano-Vasto-Chieti, Chieti, Italy; Psychopharmacology, Drug Misuse and Novel Psychoactive Substances Research Unit, School of Life and Medical Sciences, University of Hertfordshire, Hatfield, UK
| |
Collapse
|
3
|
Wang Y, Ke J, Li S, Kong Q, Zhang M, Li M, Gu J, Chi M. Analysis and study of the mechanism of narcotic addiction and withdrawal. Heliyon 2024; 10:e26957. [PMID: 38449641 PMCID: PMC10915384 DOI: 10.1016/j.heliyon.2024.e26957] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2023] [Revised: 02/21/2024] [Accepted: 02/22/2024] [Indexed: 03/08/2024] Open
Abstract
Narcotic drugs refer to drugs that have anesthetic effects on the central nervous system, and they easily produce physical dependence and mental dependence and can be addictive due to continuous use, abuse or unreasonable use. In this paper, bioinformatics and data analysis and mining techniques were used to analyze the methylation differences in transcriptional and clinical data of narcotic addiction in public databases, to explore the mechanism of narcotic addiction, and to mine some norepinephrine drugs. This study confirmed the possibility of using norepinephrine as an auxiliary drug for drug addiction rehabilitation. In addition, we also conducted a similar analysis on the addiction of three drugs. The results showed that the differences in the body caused by the ingestion of opiates and cocaine were significantly greater than those caused by the ingestion of methamphetamine.
Collapse
Affiliation(s)
- Yan Wang
- Department of Anesthesiology, Harbin Medical University Cancer Hospital, Harbin, 150081, China
| | - Jiawei Ke
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin, 150081, China
| | - Shanshan Li
- First Affiliated Hospital, Heilongjiang University of Chinese Medical, Harbin, 150040, China
| | - Qingling Kong
- Department of Anesthesiology, Harbin Medical University Cancer Hospital, Harbin, 150081, China
| | - Mingyue Zhang
- Department of Anesthesiology, Harbin Medical University Cancer Hospital, Harbin, 150081, China
| | - Mingming Li
- Department of Anesthesiology, Harbin Medical University Cancer Hospital, Harbin, 150081, China
| | - Jing Gu
- Department of Anesthesiology, Harbin Medical University Cancer Hospital, Harbin, 150081, China
| | - Meng Chi
- Department of Anesthesiology, Harbin Medical University Cancer Hospital, Harbin, 150081, China
| |
Collapse
|
4
|
Guo N, Zhang L, Fan W, Bai L, Zhang X, Shi Z, Bai J. Inhibition of Geranylgeranylacetone on cholecystokinin-B receptor, BDNF and dopamine D1 receptor induced by morphine. Biochem Biophys Res Commun 2022; 588:23-28. [PMID: 34942530 DOI: 10.1016/j.bbrc.2021.12.034] [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: 09/29/2021] [Revised: 11/13/2021] [Accepted: 12/13/2021] [Indexed: 11/21/2022]
Abstract
Morphine is the pain releasing and abusing drug. Morphine leads to addiction by activating dopaminergic rewarding system consisted of the ventral tegmental area (VTA) and nucleus accumbens (NAc). Cholecystokinin (CCK) is a gut-brain neuropeptide and involved in morphine dependence. Brain-derived neurotrophic factor (BDNF) is a neurotrophin and plays roles in regulating addiction. Geranylgeranylacetone (GGA) is a medicine of protecting gastric mucosal injury and protecting neurons. Our previous study showed that GGA blocked morphine-induced withdrawal and relapse through inducing thioredoxin 1(Trx1). In this study, we investigated that whether cholecystokinin-B receptor (CCKB receptor) and BDNF were related to GGA inhibition on morphine addiction. At first, we made conditioned place preference (CPP) model and confirmed again that GGA blocked the expression of morphine-CPP in present study. Then, our results showed that morphine increased the expressions of dopamine D1 receptor, tyrosine hydroxylase (TH), CCKB receptor and BDNF in the VTA and NAc in mice, which was inhibited by GGA. These results suggest that CCK and BDNF in dopaminergic systems are associated with the role of GGA blocking morphine-CPP.
Collapse
Affiliation(s)
- Ningning Guo
- Medical School, Kunming University of Science and Technology, Kunming, 650500, China; Faculty of Environmental Science and Engineering, Kunming University of Science and Technology, Kunming, 650500, China
| | - Le Zhang
- Medical School, Kunming University of Science and Technology, Kunming, 650500, China
| | - Wei Fan
- Medical School, Kunming University of Science and Technology, Kunming, 650500, China
| | - Liping Bai
- Medical School, Kunming University of Science and Technology, Kunming, 650500, China; Faculty of Environmental Science and Engineering, Kunming University of Science and Technology, Kunming, 650500, China
| | - Xianwen Zhang
- Medical School, Kunming University of Science and Technology, Kunming, 650500, China
| | - Zhizhou Shi
- Medical School, Kunming University of Science and Technology, Kunming, 650500, China
| | - Jie Bai
- Medical School, Kunming University of Science and Technology, Kunming, 650500, China.
| |
Collapse
|
5
|
A Role of BDNF in the Depression Pathogenesis and a Potential Target as Antidepressant: The Modulator of Stress Sensitivity "Shati/Nat8l-BDNF System" in the Dorsal Striatum. Pharmaceuticals (Basel) 2021; 14:ph14090889. [PMID: 34577589 PMCID: PMC8469819 DOI: 10.3390/ph14090889] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2021] [Revised: 08/27/2021] [Accepted: 08/27/2021] [Indexed: 12/12/2022] Open
Abstract
Depression is one of the most common mental diseases, with increasing numbers of patients globally each year. In addition, approximately 30% of patients with depression are resistant to any treatment and do not show an expected response to first-line antidepressant drugs. Therefore, novel antidepressant agents and strategies are required. Although depression is triggered by post-birth stress, while some individuals show the pathology of depression, others remain resilient. The molecular mechanisms underlying stress sensitivity remain unknown. Brain-derived neurotrophic factor (BDNF) has both pro- and anti-depressant effects, dependent on brain region. Considering the strong region-specific contribution of BDNF to depression pathogenesis, the regulation of BDNF in the whole brain is not a beneficial strategy for the treatment of depression. We reviewed a novel finding of BDNF function in the dorsal striatum, which induces vulnerability to social stress, in addition to recent research progress regarding the brain regional functions of BDNF, including the prefrontal cortex, hippocampus, and nucleus accumbens. Striatal BDNF is regulated by Shati/Nat8l, an N-acetyltransferase through epigenetic regulation. Targeting of Shati/Nat8l would allow BDNF to be striatum-specifically regulated, and the striatal Shati/Nat8l-BDNF pathway could be a promising novel therapeutic agent for the treatment of depression by modulating sensitivity to stress.
Collapse
|
6
|
Microinjection of the BDNF receptor antagonist ANA-12 into the nucleus accumbens and medial-prefrontal cortex attenuates morphine-induced reward memory, and alterations of BDNF levels and apoptotic cells in rats. Pharmacol Biochem Behav 2021; 201:173111. [PMID: 33444602 DOI: 10.1016/j.pbb.2021.173111] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/25/2020] [Revised: 12/24/2020] [Accepted: 12/28/2020] [Indexed: 01/10/2023]
Abstract
This study was designed to examine the effects of intra- nucleus accumbens (NAc) of BDNF receptor antagonist ANA-12 on the acquisition and expression and intra- medial-prefrontal cortex (mPFC) of ANA-12 on the extinction and reinstatement of morphine-induced conditioned place preference (CPP) and also BDNF levels and apoptotic neurons in the NAc and mPFC of rats. In this study, adult male Wistar rats (200-250 g) were used. Two separate cannulas were inserted bilaterally into the NAc and/or mPFC. ANA-12 (3 μg/0.5 μl/side) was injected into the NAc and/or mPFC to evaluate the rewarding effects of morphine using a CPP paradigm. Then, the levels of BDNF and apoptotic in the NAc and mPFC were assessed at the end of each treatment phase using ELISA and TUNEL methods, respectively. All of vehicle-treated rats following morphine CPP showed the increase of BDNF levels and apoptotic neurons in the NAc and mPFC. ANA-12 significantly attenuated the acquisition and expression of morphine-induced CPP, BDNF levels and apoptotic neurons in the NAc during the acquisition, but not the expression phase. Also, ANA-12 significantly facilitated the extinction, but no effect on reinstatement of morphine CPP, and decreased BDNF levels and apoptotic neurons in the mPFC during the extinction, but not the reinstatement. We conclude that blocking TrkB with ANA-12 showed therapeutic effects on morphine-associated reward memory and neuronal death in the NAc and mPFC induced by morphine CPP. Thus, the BDNF-TrkB signaling may be important in the acquisition, expression, extinction, but not the reinstatement of morphine CPP.
Collapse
|
7
|
Gold MS, Baron D, Bowirrat A, Blum K. Neurological correlates of brain reward circuitry linked to opioid use disorder (OUD): Do homo sapiens acquire or have a reward deficiency syndrome? J Neurol Sci 2020; 418:117137. [PMID: 32957037 PMCID: PMC7490287 DOI: 10.1016/j.jns.2020.117137] [Citation(s) in RCA: 36] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2020] [Revised: 08/19/2020] [Accepted: 09/08/2020] [Indexed: 12/14/2022]
Abstract
The extant literature confirms that an array of polymorphic genes related to- neurotransmitters and second messengers govern the net release of dopamine in the Nucleus Accumbens (NAc) in the mesolimbic region of the brain. They are linked predominantly to motivation, anti-stress, incentive salience (wanting), and wellbeing. Notably, in 2000 the Nobel Prize was awarded to Carlsson, Greengard, and Kandel for their work on the molecular and cellular function of dopaminergic activity at neurons. This historical psychopharmacological work involved neurotransmission of serotonin, endorphins, glutamate, and dopamine, and the seminal work of Blum, Gold, Volkow, Nestler, and others related to neurotransmitter function and related behaviors. Currently, Americans are facing their second and worst opioid epidemic, prescribed opioids, and easy access drive this epidemic of overdoses, and opioid use disorders (OUDs). Presently the clinical consensus is to treat OUD, as if it were an opioid deficiency syndrome, with long-term to life-long opioid substitution therapy. Opioid agonist administration is seen as necessary to replace missing opioids, treat OUD, and prevent overdoses, like insulin is used to treat diabetes. Treatment of OUD and addiction, in general, is similar to the endocrinopathy conceptualization in that it views opioid agonist MATs as an essential core to therapy. Is this approach logical? Other than as harm reduction, is using opioids to treat OUD therapeutic or harmful in the long term? This historical Trieste provides a molecular framework to understand the current underpinnings of endorphinergic/dopaminergic mechanisms related to opioid deficiency syndrome and generalized reward processing depletion. WC 249.
Collapse
Affiliation(s)
- Mark S Gold
- Department of Psychiatry, Washington University School of Medicine, St. Louis, MO, United States.
| | - David Baron
- Graduate School of Biomedical Sciences, Western University Health Sciences, Pomona, CA, United States
| | - Abdalla Bowirrat
- Department of Neuroscience and Genetics, Interdisciplinary Center Herzliya, Israel
| | - Kenneth Blum
- Graduate School of Biomedical Sciences, Western University Health Sciences, Pomona, CA, United States
| |
Collapse
|
8
|
Fatahi Z, Zeinaddini-Meymand A, Karimi S, Khodagholi F, Haghparast A. Impairment of cost-benefit decision making in morphine-dependent rats is partly mediated via the alteration of BDNF and p-CREB levels in the nucleus accumbens. Pharmacol Biochem Behav 2020; 194:172952. [PMID: 32428531 DOI: 10.1016/j.pbb.2020.172952] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/13/2020] [Revised: 05/12/2020] [Accepted: 05/14/2020] [Indexed: 02/09/2023]
Abstract
The ability to choose goals based on decision usefulness or the time required to reach the goals chosen are important aspects of decision making. There is considerable evidence in the literature indicating the fact that drug abuse affects different aspects of cognition. In the current study, we assessed the effects of morphine dependence and its withdrawal on cost-benefit decision making and furthermore the involvement of BDNF and p-CREB in the nucleus accumbens, a key brain area involved in decision making was measured. Different groups of male Wistar rats were trained in an effort-based and/or delay-based form of cost-benefit T-maze decision-making task. Thereafter, the animals were morphine dependent and the percentage of the high reward preference was evaluated. After behavioral tests, the BDNF level, and p-CREB/CREB ratio were measured by Western blot analysis. The results showed that during effort-based but not delay-based decision making, BDNF and p-CREB levels increased. During effort-based decision making in morphine dependent rats, BDNF decreased but there was no significant change in p-CREB. Besides, during delay-based decision making in the morphine dependent group, both BDNF and p-CREB did not show any significant change. These findings revealed that BDNF and p-CREB/CREB ratio in the NAc are essential factors for effort-based but not delay-based decision making. In addition, impairment of effort-based decision making in morphine dependent rats is related to the decrease of BDNF level but not p-CREB/CREB ratio in the NAc. However, delay-based decision making defects in morphine dependent rats did not associate with the change in BDNF and p-CREB levels in the NAc.
Collapse
Affiliation(s)
- Zahra Fatahi
- Neuroscience Research Center, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Arman Zeinaddini-Meymand
- Neuroscience Research Center, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Sara Karimi
- Neuroscience Research Center, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Fariba Khodagholi
- Neuroscience Research Center, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Abbas Haghparast
- Neuroscience Research Center, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
9
|
Puryear CB, Brooks J, Tan L, Smith K, Li Y, Cunningham J, Todtenkopf MS, Dean RL, Sanchez C. Opioid receptor modulation of neural circuits in depression: What can be learned from preclinical data? Neurosci Biobehav Rev 2020; 108:658-678. [DOI: 10.1016/j.neubiorev.2019.12.007] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2019] [Revised: 12/02/2019] [Accepted: 12/05/2019] [Indexed: 12/14/2022]
|
10
|
Koo JW, Chaudhury D, Han MH, Nestler EJ. Role of Mesolimbic Brain-Derived Neurotrophic Factor in Depression. Biol Psychiatry 2019; 86:738-748. [PMID: 31327473 PMCID: PMC6814503 DOI: 10.1016/j.biopsych.2019.05.020] [Citation(s) in RCA: 71] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/28/2019] [Revised: 05/20/2019] [Accepted: 05/24/2019] [Indexed: 11/27/2022]
Abstract
Brain-derived neurotrophic factor (BDNF) is widely accepted as being critical for neural and synaptic plasticity throughout the nervous system. Recent work has shown that BDNF in the mesolimbic dopamine (DA) circuit, originating in ventral tegmental area DA neurons that project to the nucleus accumbens, is crucial in the development of depressive-like behaviors following exposure to chronic social defeat stress in mice. Whereas BDNF modulates DA signaling in encoding responses to acute defeat stress, BDNF signaling alone appears to be responsible for the behavioral effects after chronic social defeat stress. Very different patterns are seen with another widely used chronic stress paradigm in mice, chronic mild stress (also known as chronic variable or unpredictable stress), where DA signaling, but not BDNF signaling, is primarily responsible for the behavioral effects observed. This review discusses the molecular, cellular, and circuit basis of this dramatic discrepancy, which appears to involve the nature of the stress, its severity and duration, and its effects on distinct cell types within the ventral tegmental area-to-nucleus accumbens mesolimbic circuit.
Collapse
Affiliation(s)
- Ja Wook Koo
- Department of Neural Development and Disease, Korea Brain Research Institute (KBRI), 61, Cheomdan-ro, Dong-gu, Daegu, 41068, Republic of Korea
| | - Dipesh Chaudhury
- Division of Science, New York University Abu Dhabi (NYUAD), Saadiyat Island Campus, Abu Dhabi, PO Box 129188, United Arab Emirates
| | - Ming-Hu Han
- Department of Pharmacological Sciences, Icahn School of Medicine at Mount Sinai, New York, New York; Nash Family Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, New York; Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, New York.
| | - Eric J. Nestler
- Departments of Pharmacological Sciences and of Neuroscience and Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA,Address correspondence to: Ming-Hu Han, Ph.D. and Eric J. Nestler, MD., Ph.D., Department of Neuroscience, Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; and
| |
Collapse
|
11
|
Eskandari Z, Dadashi M, Mostafavi H, Armani Kia A, Pirzeh R. Comparing the Efficacy of Anodal, Cathodal, and Sham Transcranial Direct Current Stimulation on Brain-Derived Neurotrophic Factor and Psychological Symptoms in Opioid-Addicted Patients. Basic Clin Neurosci 2019; 10:641-650. [PMID: 32477481 PMCID: PMC7253809 DOI: 10.32598/bcn.10.6.1710.1] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2019] [Revised: 03/10/2019] [Accepted: 08/31/2019] [Indexed: 12/22/2022] Open
Abstract
Introduction: Today, addiction to opioids is a serious problem all over the world. Unfortunately, the consumption of these drugs and the number of addicted people have drastically increased. This research aimed at comparing the efficacy of anodal, cathodal, and sham transcranial Direct Current Stimulation (tDCS) on the Brain-Derived Neurotrophic Factor (BDNF) and psychological symptoms in opioid-addicted patients. Methods: Thirty opioid-addicted patients were selected based on the Diagnostic and Statistical Manual of Mental Disorders, the Fifth Edition, through the convenience sampling method. They were then randomly assigned to 3 groups (10 in each group). The subjects were evaluated before and after tDCS by their serum level of BDNF, desires for drug questionnaire, and depression anxiety stress scale. The data were analyzed by the Kolmogorov-Smirnov test, one-way analysis of variance, as well as the Bonferroni test. Results: Stimulating the Dorsolateral Prefrontal Cortex (DLPFC) led to a significant change in increasing the level of BDNF (P=0.031) and reducing the degree of depression (P=0.018), anxiety (P=0.001), stress (P=0.012), and decreased the level of craving (P=0.001) in opioid-addicted patients. There was no significant difference between active stimulation groups (anodal left/cathodal right and anodal right/cathodal left). The stimulation of the right DLPFC (group B) significantly increased BDNF in comparison with the sham group (sham tDCS) and decreased anxiety and craving. Nonetheless, no change was observed in depression and stress. The stimulation of the left DLPFC (group A) significantly reduced depression, anxiety, stress, and craving compared with the sham group, while there was no change in BDNF. Conclusion: In addition to the conventional treatments of opioid-addicted patients, tDCS is an effective complementary treatment.
Collapse
Affiliation(s)
- Zakaria Eskandari
- Department of Clinical Psychology, Faculty of Medicine, Zanjan University of Medical Sciences, Zanjan, Iran
| | - Mohsen Dadashi
- Department of Clinical Psychology, Faculty of Medicine, Social Determinants of Health Research Center, Zanjan University of Medical Sciences, Zanjan, Iran
| | - Hossin Mostafavi
- Department of Clinical Psychology, Social Determinants of Health Research Center, Zanjan University of Medical Sciences, Zanjan, Iran
| | - Alireza Armani Kia
- Department of Physiology, Faculty of Medicine, Zanjan University of Medical Sciences, Zanjan, Iran
| | - Reza Pirzeh
- Department of Physiology, Faculty of Medicine, Zanjan University of Medical Sciences, Zanjan, Iran
| |
Collapse
|
12
|
Abstract
Acupuncture is an ancient therapy with a variety of different explanatory models. A cascade of physiological effects has been reported, both in the peripheral and the central nervous system, following the insertion of a needle or light tapping of the skin. Clinical trials testing the specific claims of acupuncture have generally tried to focus on testing the efficacy of applying specific techniques and/or specified points. However, different conditions may respond differently to different modes of stimulation. Recently, it was demonstrated that both superficial and deep needling (with de qi/Hibiki) resulted in amelioration of patellofemoral pain and unpleasantness. The pleasurable aspect of the acupuncture experience has largely been ignored as it has been considered secondary to its pain alleviating effects. This aspect of acupuncture treatment is likely to be related to activation of self-appraisal and the reward system. When a patient seeks a therapist there are expectations of a specific effect. These expectations are partly based on self-relevant phenomena and self-referentia introspection and constitute the preference. Also, when asked about the effect of the treatment, processes that orientate pre-attentive anticipatory or mnemonic information and processes that mediate self-reflection and recollection are integrated together with sensory detection to enable a decision about the patient's perception of the effect of acupuncture treatment. These ‘self-appraisal’ processes are dependent on two integrated networks: a ventral medial prefrontal cortex paralimbic limbic ‘affective’ pathway and a dorsal medial prefrontal cortex cortical hippocampal ‘cognitive’ pathway. The limbic structures are implicated in the reward system and play a key role in most diseases and illness responses including chronic pain and depression, regulating mood and neuromodulatory responses (eg sensory, autonomic, and endocrine). The pleasurable and neuromodulatory aspects of acupuncture as well as ‘placebo needling’ may partly be explained by the activation or deactivation of limbic structures including the hippocampus, amygdala, and their connections with the hypothalamus. In patients with patellofemoral pain, the effects of superficial and deep needling remained for six months. These long term pain-alleviating effects have been attributed to activation of pain inhibiting systems in cortical and subcortical pathways. When considering long term effects the cortical cerebellar system needs to be taken into account. The cortical cerebellar system is probably central to the development of neural models that learn and eventually stimulate routinely executed (eg motor skills) and long term (eg pain alleviation) cognitive processes. These higher order cognitive processes are initially mediated in prefrontal cortical loci but later shift control iteratively to internal cerebellar representations of these processes. Possibly part of the long term healing effects of acupuncture may be attributed to changes in the cerebellar system thereby sparing processing load in cortical and subcortical areas. As cortical and subcortical structures are activated and/or de-activated following stimulation of receptors in the skin, disregarding site, ‘placebo or sham needling’ does not exist and conclusions drawn on the basis that it is an inert control are invalid. ‘Self’ may be seen as a shifting illusion, ceaselessly constructed and deconstructed, and the effect of acupuncture may reflect its status (as well as that of the therapist).
Collapse
Affiliation(s)
- Thomas Lundeberg
- Rehabilitation Medicine, UniversityClinic, Danderyds Hospital, Stockholm, Sweden.
| | | | | |
Collapse
|
13
|
Ferrer-Pérez C, Castro-Zavala A, Luján MÁ, Filarowska J, Ballestín R, Miñarro J, Valverde O, Rodríguez-Arias M. Oxytocin prevents the increase of cocaine-related responses produced by social defeat. Neuropharmacology 2018; 146:50-64. [PMID: 30448423 DOI: 10.1016/j.neuropharm.2018.11.011] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2018] [Revised: 11/08/2018] [Accepted: 11/09/2018] [Indexed: 12/17/2022]
Abstract
The neuropeptide oxytocin (OXT) plays a critical role in the regulation of social and emotional behaviors. OXT plays a role in stress response and in drug reward, but to date no studies have evaluated its implication in the long-lasting increase of the motivational effects of cocaine induced by repeated social defeat (RSD). During the social defeat procedure, 1 mg/kg of OXT was administered 30 min before each episode of RSD. Three weeks after the last defeat, the effects of cocaine on the conditioned place preference (CPP), locomotor sensitization and the self-administration (SA) paradigms were evaluated. The influence of OXT on the levels of BDNF in the prefrontal cortex (PFC), striatum and hippocampus was also measured. Our results confirm that raising the levels of OXT during social defeat stress can block the long-lasting effects of this type of stress. OXT counteracts the anxiety induced by social defeat and modifies BDNF levels in all the structures we have studied. Moreover, OXT prevents RSD-induced increases in the motivational effects of cocaine. Administration of OXT before each social defeat blocked the social defeat-induced increment in the conditioned rewarding effects of cocaine in the CPP, favored the extinction of cocaine-associated memories in both the CPP and SA, and decreased reinstatement of cocaine-seeking behavior in the SA. In conclusion, the long-lasting effects of RSD are counteracted by administering OXT prior to stress, and changes in BDNF expression may underlie these protective effects.
Collapse
Affiliation(s)
- Carmen Ferrer-Pérez
- Unit of Research on Psychobiology of Drug Dependence, Department of Psychobiology, Faculty of Psychology, Universitat de València, Valencia, Spain
| | - Adriana Castro-Zavala
- Neurobiology of Behavior Research Group (GReNeC-NeuroBio), Department of Health and Experimental Sciences, University Pompeu Fabra, IMIM (Hospital del Mar Medical Research Institute), Barcelona, Spain
| | - Miguel Ángel Luján
- Neurobiology of Behavior Research Group (GReNeC-NeuroBio), Department of Health and Experimental Sciences, University Pompeu Fabra, IMIM (Hospital del Mar Medical Research Institute), Barcelona, Spain
| | - Joanna Filarowska
- Department of Pharmacology and Pharmacodynamics, Medical University of Lublin, Chodzki 4a, 20-093, Lublin, Poland
| | - Raúl Ballestín
- Unit of Research on Psychobiology of Drug Dependence, Department of Psychobiology, Faculty of Psychology, Universitat de València, Valencia, Spain
| | - José Miñarro
- Unit of Research on Psychobiology of Drug Dependence, Department of Psychobiology, Faculty of Psychology, Universitat de València, Valencia, Spain
| | - Olga Valverde
- Neurobiology of Behavior Research Group (GReNeC-NeuroBio), Department of Health and Experimental Sciences, University Pompeu Fabra, IMIM (Hospital del Mar Medical Research Institute), Barcelona, Spain
| | - Marta Rodríguez-Arias
- Unit of Research on Psychobiology of Drug Dependence, Department of Psychobiology, Faculty of Psychology, Universitat de València, Valencia, Spain.
| |
Collapse
|
14
|
Moreira da Silva Santos A, Kelly JP, Doyle KM. Dose-Dependent Effects of Binge-Like Methamphetamine Dosing on Dopamine and Neurotrophin Levels in Rat Brain. Neuropsychobiology 2018; 75:63-71. [PMID: 29065400 DOI: 10.1159/000480513] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/26/2017] [Accepted: 08/21/2017] [Indexed: 01/19/2023]
Abstract
This study investigated the acute effect of a dose range of low-to-moderate binge-like methamphetamine treatments on the regional expression of neurotrophin proteins in the brain and serum 2 h after the last dose, in addition to assessing the behavioural effects and dopamine neurotransmitter changes produced. Male Sprague-Dawley rats received 4 subcutaneous doses of methamphetamine (0.5, 1, 2, and 4 mg/kg, or saline as a control) 2 h apart. Methamphetamine had a dose-dependent stimulatory effect on locomotor activity over the 8 h of observation. A significant increase in dopamine concentration was observed in the frontal cortex with the highest dose of methamphetamine (2 h after the last dose). This effect was dose- and region-specific, as no significant increase was observed with lower doses, nor was a significant change observed in any other brain region tested. A similar dose- and region-specific increase in brain-derived neurotrophic factor (BDNF) was observed in the frontal cortex with the highest-dose regimen. No significant change occurred with lower doses of methamphetamine, or in any other brain region tested. A reduction in BDNF levels in the serum was also observed with the highest concentration, but not with lower doses. Collectively, this data highlights the importance of the frontal cortex in methamphetamine-induced effects, and also the similar dose-response effect of methamphetamine on dopamine and BDNF expression.
Collapse
|
15
|
Coplan JD, Lu D, El Sehamy AM, Tang C, Jackowski AP, Abdallah CG, Nemeroff CB, Owens MJ, Mathew SJ, Gorman JM. Early Life Stress Associated With Increased Striatal N-Acetyl-Aspartate: Cerebrospinal Fluid Corticotropin-Releasing Factor Concentrations, Hippocampal Volume, Body Mass and Behavioral Correlates. ACTA ACUST UNITED AC 2018; 2. [PMID: 29963652 PMCID: PMC6020138 DOI: 10.1177/2470547018768450] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Introduction Using proton magnetic resonance spectroscopy imaging, the effects of early
life stress on nonhuman primate striatal neuronal integrity were examined as
reflected by N-acetyl aspartate (NAA) concentrations. NAA
measures were interrogated through examining their relationship to
previously documented early life stress markers—cerebrospinal fluid
corticotropin-releasing factor concentrations, hippocampal volume, body
mass, and behavioral timidity. Rodent models of depression exhibit increases
in neurotrophic effects in the nucleus accumbens. We hypothesized that
rearing under conditions of early life stress (variable foraging demand,
VFD) would produce persistent elevations of NAA concentrations (in absolute
or ratio form) in ventral striatum/caudate nucleus (VS/CN) with altered
correlation to early life stress markers. Methods Eleven bonnet macaque males reared under VFD conditions and seven age-matched
control subjects underwent proton magnetic resonance spectroscopy imaging
during young adulthood. Voxels were placed over VS/CN to capture nucleus
accumbens. Cisternal cerebrospinal fluid corticotropin-releasing factor
concentrations, hippocampal volume, body mass, and response to a human
intruder had been previously determined. Results VFD-reared monkeys exhibited significantly increased NAA/creatine
concentrations in right VS/CN in comparison to normally reared controls,
controlling for multiple comparisons. In comparison to controls, VFD
cerebrospinal fluid corticotropin-releasing factor concentrations were
directly associated with right VS/CN absolute NAA. Left hippocampal volume
was inversely associated with left VS/CN NAA/creatine in VFD reared but not
in controls. Disruption of a normative inverse correlation between left
VS/CN NAA and body mass was noted in VFD. Only non-VFD subjects exhibited a
direct relationship between timidity response to an intruder and right VS/CN
NAA. Conclusion Early life stress produced persistent increases in VS/CN NAA, which
demonstrated specific patterns of association (or lack thereof) to early
life stress markers in comparison to non-VFD subjects. The data are broadly
consistent with a stable nonhuman primate phenotype of anxiety and mood
disorder vulnerability whereby in vivo indicators of neuronal integrity,
although reduced in hippocampus, are increased in striatum. The findings may
provide a catalyst for further studies in humans and other species regarding
a reciprocal hippocampal/nucleus accumbens relationship in affective
disorders.
Collapse
Affiliation(s)
- Jeremy D Coplan
- Department of Psychiatry & Behavioral Sciences, State University of New York Downstate Medical Center, Brooklyn, NY
| | - Dunyue Lu
- McLaren Behavioral Health Services, Flint Township, MI, USA
| | | | - Cheuk Tang
- Departments of Psychiatry, Neuroscience, and Radiology, Mount Sinai School of Medicine, New York, NY, USA
| | | | - Chadi G Abdallah
- Department of Psychiatry, Yale University School of Medicine, New Haven, CT, USA
| | - Charles B Nemeroff
- Department of Psychiatry and Behavioral Sciences, University of Miami Health Systems, Miami, NY, USA
| | - Michael J Owens
- Department of Psychiatry and Behavioral Sciences, Emory University School of Medicine, Atlanta GA, USA
| | - Sanjay J Mathew
- Mental Health Care Line, Michael E. Debakey VA Medical Center, Houston, TX, USA; Menninger Department of Psychiatry & Behavioral Sciences, Baylor College of Medicine, Houston, TX, USA
| | - Jack M Gorman
- Franklin Behavioral Health Care Consultants and Critica LLC, Bronx, New York, USA
| |
Collapse
|
16
|
Li Y, Xia B, Li R, Yin D, Wang Y, Liang W. Expression of brain-derived neurotrophic factors, neurotrophin-3, and neurotrophin-4 in the nucleus accumbens during heroin dependency and withdrawal. Neuroreport 2018; 28:654-660. [PMID: 28538519 PMCID: PMC5491224 DOI: 10.1097/wnr.0000000000000810] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
Neurotrophins, brain-derived neurotrophic factors (BDNF), neurotrophin-3 (NT-3), and neurotrophin-4 (NT-4), have been implicated in the modulation of heroin dependency. This study was designed to explore the expression alterations of BDNF, NT-3, and NT-4 in the context of heroin dependence and withdrawal in the rat nucleus accumbens (NAc). Heroin dependence was induced by a progressive intraperitoneal treatment of heroin. The results showed that the expression levels of BDNF and NT-4 were significantly decreased in the NAc of rats with heroin addiction in comparison with the control group, whereas there was a significant increase in BDNF and NT-4 expressions in the groups of rats with both naloxone-induced and spontaneous withdrawal. Moreover, NT-3 expression was markedly increased in the NAc of rats with heroin addiction and spontaneous withdrawal in comparison with the control group, but decreased in the NAc of rats with naloxone-induced withdrawal. These results indicated that chronic administration of heroin results in the alterations of BDNF, NT-3, and NT-4 expressions in the rat NAc. BDNF, NT-3, and NT-4 may play a critical role in the development of heroin dependency and withdrawal.
Collapse
Affiliation(s)
- Yixin Li
- Department of Histology and Embryology, Guizhou Medical University, Guiyang, China
| | | | | | | | | | | |
Collapse
|
17
|
Subedi L, Huang H, Pant A, Westgate PM, Bada HS, Bauer JA, Giannone PJ, Sithisarn T. Plasma Brain-Derived Neurotrophic Factor Levels in Newborn Infants with Neonatal Abstinence Syndrome. Front Pediatr 2017; 5:238. [PMID: 29164087 PMCID: PMC5675851 DOI: 10.3389/fped.2017.00238] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/31/2017] [Accepted: 10/20/2017] [Indexed: 12/12/2022] Open
Abstract
BACKGROUND Brain-derived neurotrophic factor (BDNF) is a type of growth factor that promotes growth and survival of neurons. Fetal exposure to opiates can lead to postnatal withdrawal syndrome, which is referred as neonatal abstinence syndrome (NAS). Preclinical and clinical studies have shown an association between opiates exposure and alteration in BDNF expression in the brain and serum levels in adult. However, to date, there are no data available on the effects of opiate exposure on BDNF levels in infant who are exposed to opiates in utero and whether BDNF level may correlate with the severity of NAS. OBJECTIVE To compare plasma BDNF levels among NAS and non-NAS infants and to determine the correlation of BDNF levels and the severity of NAS. METHODS This is a prospective cohort study with no intervention involved. Infants ≥35 weeks of gestation were enrolled. BDNF level was measured using enzyme-linked immunosorbent assay technique from blood samples drawn within 48 h of life. The severity of NAS was determined by the length of hospital stay, number of medications required to treat NAS. RESULTS 67 infants were enrolled, 34 NAS and 33 non-NAS. Mean gestational age did not differ between the two groups. Mean birth weight of NAS infants was significantly lower than the non-NAS infants (3,070 ± 523 vs. 3,340 ± 459 g, p = 0.028). Mean BDNF level in NAS group was 252.2 ± 91.6 ng/ml, significantly higher than 211.3 ± 66.3 ng/ml in the non-NAS group (p = 0.04). There were no differences in BDNF levels between NAS infants that required one medication vs. more than one medication (254 ± 91 vs. 218 ± 106 ng/ml, p = 0.47). There was no correlation between the BDNF levels and length of hospital stay (p = 0.68) among NAS infants. Overall, there were no significant correlations between BDNF levels and NAS scores except at around 15 h after admission (correlation 0.35, p = 0.045). CONCLUSION Plasma BDNF level was significantly increased in NAS infants during the first 48 h when compared to non-NAS infants. The correlations between plasma BDNF levels and the severity of NAS warrant further study. These results suggest that BDNF may play a neuromodulatory role during withdrawal after in utero opiate exposure.
Collapse
Affiliation(s)
- Lochan Subedi
- Department of Pediatrics, University of Kentucky, Lexington, KY, United States
| | - Hong Huang
- Department of Pediatrics, University of Kentucky, Lexington, KY, United States
| | - Amrita Pant
- Department of Pediatrics, University of Kentucky, Lexington, KY, United States
| | - Philip M Westgate
- Department of Biostatistics, College of Public Health, University of Kentucky, Lexington, KY, United States
| | - Henrietta S Bada
- Department of Pediatrics, University of Kentucky, Lexington, KY, United States
| | - John A Bauer
- Department of Pediatrics, University of Kentucky, Lexington, KY, United States
| | - Peter J Giannone
- Department of Pediatrics, University of Kentucky, Lexington, KY, United States
| | - Thitinart Sithisarn
- Department of Pediatrics, University of Kentucky, Lexington, KY, United States
| |
Collapse
|
18
|
Pereira PA, Millner T, Vilela M, Sousa S, Cardoso A, Madeira MD. Nerve growth factor-induced plasticity in medial prefrontal cortex interneurons of aged Wistar rats. Exp Gerontol 2016; 85:59-70. [DOI: 10.1016/j.exger.2016.09.017] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2016] [Revised: 08/04/2016] [Accepted: 09/20/2016] [Indexed: 01/03/2023]
|
19
|
Zhong X, Drgonova J, Li CY, Uhl GR. Human cell adhesion molecules: annotated functional subtypes and overrepresentation of addiction-associated genes. Ann N Y Acad Sci 2015; 1349:83-95. [PMID: 25988664 DOI: 10.1111/nyas.12776] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Human cell adhesion molecules (CAMs) are essential for proper development, modulation, and maintenance of interactions between cells and cell-to-cell (and matrix-to-cell) communication about these interactions. Despite the differential functional significance of these roles, there have been surprisingly few systematic studies to enumerate the universe of CAMs and identify specific CAMs in distinct functions. In this paper, we update and review the set of human genes likely to encode CAMs with searches of databases, literature reviews, and annotations. We describe likely CAMs and functional subclasses, including CAMs that have a primary function in information exchange (iCAMs), CAMs involved in focal adhesions, CAM gene products that are preferentially involved with stereotyped and morphologically identifiable connections between cells (e.g., adherens junctions, gap junctions), and smaller numbers of CAM genes in other classes. We discuss a novel proposed mechanism involving selective anchoring of the constituents of iCAM-containing lipid rafts in zones of close neuronal apposition to membranes expressing iCAM binding partners. We also discuss data from genetic and genomic studies of addiction in humans and mouse models to highlight the ways in which CAM variation may contribute to a specific brain-based disorder such as addiction. Specific examples include changes in CAM mRNA splicing mediated by differences in the addiction-associated splicing regulator RBFOX1/A2BP1 and CAM expression in dopamine neurons.
Collapse
Affiliation(s)
- Xiaoming Zhong
- Laboratory of Bioinformatics and Genomic Medicine, Institute of Molecular Medicine, Peking University, Beijing, China
| | - Jana Drgonova
- Molecular Neurobiology, NIH-IRP (NIDA), Baltimore, Maryland
| | - Chuan-Yun Li
- Laboratory of Bioinformatics and Genomic Medicine, Institute of Molecular Medicine, Peking University, Beijing, China
| | - George R Uhl
- Molecular Neurobiology, NIH-IRP (NIDA), Baltimore, Maryland.,Research Office, New Mexico VA Health Care System, Albuquerque, New Mexico
| |
Collapse
|
20
|
Sex differences between CRF1 receptor deficient mice following naloxone-precipitated morphine withdrawal in a conditioned place aversion paradigm: implication of HPA axis. PLoS One 2015; 10:e0121125. [PMID: 25830629 PMCID: PMC4382215 DOI: 10.1371/journal.pone.0121125] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2014] [Accepted: 01/28/2015] [Indexed: 11/19/2022] Open
Abstract
Background Extinction period of positive affective memory of drug taking and negative affective memory of drug withdrawal, as well as the different response of men and women might be important for the clinical treatment of drug addiction. We investigate the role of corticotropin releasing factor receptor type one (CRF1R) and the different response of male and female mice in the expression and extinction of the aversive memory. Methodology/Principal Finding We used genetically engineered male and female mice lacking functional CRF1R. The animals were rendered dependent on morphine by intraperitoneally injection of increasing doses of morphine (10–60 mg/kg). Negative state associated with naloxone (1 mg/kg s.c.)-precipitated morphine withdrawal was examined by using conditioned place aversion (CPA) paradigm. No sex differences for CPA expression were found in wild-type (n = 29) or CRF1R knockout (KO) mice (n = 29). However, CRF1R KO mice presented less aversion score than wild-type mice, suggesting that CRF1R KO mice were less responsive than wild-type to continuous associations between drug administration and environmental stimuli. In addition, CPA extinction was delayed in wild-type and CRF1R KO male mice compared with females of both genotypes. The genetic disruption of the CRF1R pathway decreased the period of extinction in males and females suggesting that CRF/CRF1R is implicated in the duration of aversive memory. Our results also showed that the increase in adrenocorticotropic hormone (ACTH) levels observed in wild-type (n = 11) mice after CPA expression, were attenuated in CRF1R KO mice (n = 10). In addition, ACTH returned to the baseline levels in males and females once CPA extinction was finished. Conclusion/Significance These results suggest that, at least, CPA expression is partially due to an increase in plasma ACTH levels, through activation of CRF1R, which can return when CPA extinction is finished.
Collapse
|
21
|
Epigenetic basis of opiate suppression of Bdnf gene expression in the ventral tegmental area. Nat Neurosci 2015; 18:415-22. [PMID: 25643298 PMCID: PMC4340719 DOI: 10.1038/nn.3932] [Citation(s) in RCA: 79] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2014] [Accepted: 12/22/2014] [Indexed: 12/15/2022]
Abstract
Brain-derived neurotrophic factor (BDNF) plays a crucial role in modulating neural and behavioral plasticity to drugs of abuse. Here, we demonstrate a persistent down-regulation of exon-specific Bdnf expression in the ventral tegmental area (VTA) in response to chronic opiate exposure, which is mediated by specific epigenetic modifications at the corresponding Bdnf gene promoters. Exposure to chronic morphine increases stalling of RNA polymerase II at these Bdnf promoters in VTA and alters permissive and repressive histone modifications and occupancy of their regulatory proteins at the specific promoters. Furthermore, we show that morphine suppresses binding of phospho-CREB (cAMP response element binding protein) to Bdnf promoters in VTA, which results from enrichment of trimethylated H3K27 at the promoters, and that decreased NURR1 (nuclear receptor related-1) expression also contributes to Bdnf repression and associated behavioral plasticity to morphine. These studies reveal novel epigenetic mechanisms of morphine-induced molecular and behavioral neuroadaptations.
Collapse
|
22
|
Peregud DI, Panchenko LF, Gulyaeva NV. Elevation of BDNF exon I-specific transcripts in the frontal cortex and midbrain of rat during spontaneous morphine withdrawal is accompanied by enhanced pCreb1 occupancy at the corresponding promoter. Neurochem Res 2015; 40:130-8. [PMID: 25392083 DOI: 10.1007/s11064-014-1476-y] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2014] [Revised: 11/04/2014] [Accepted: 11/05/2014] [Indexed: 01/03/2023]
Abstract
Brain-derived neurotrophic factor (BDNF) is believed to play a crucial role in the mechanisms underlying opiate dependence; however, little is known about specific features and mechanisms regulating its expression in the brain under these conditions. The aim of this study was to investigate the effects of acute morphine intoxication and withdrawal from chronic intoxication on expression of BDNF exon I-, II-, IV-, VI- and IX-containing transcripts in the rat frontal cortex and midbrain. We also have studied whether alterations of BDNF exon-specific transcripts are accompanied by changes in association of well-known transcriptional regulators of BDNF gene-phosphorylated (active form) cAMP response element binding protein (pCreb1) and methyl-CpG binding protein 2 (MeCP2) with corresponding regulatory regions of the BDNF gene. Acute morphine intoxication did not affect levels of BDNF exons in brain regions, while spontaneous morphine withdrawal in dependent rats was accompanied by an elevation of the BDNF exon I-containing mRNAs both in the frontal cortex and midbrain. During spontaneous morphine withdrawal, increased associations of pCreb1 were found with promoter of exon I in the frontal cortex and promoters of exon I, IV and VI in the midbrain. The association of MeCP2 with BDNF promoters during spontaneous morphine withdrawal did not change. Thus, BDNF exon-specific transcripts are differentially expressed in brain regions during spontaneous morphine withdrawal in dependent rats and pCreb1 may be at least partially responsible for these alterations.
Collapse
Affiliation(s)
- Danil I Peregud
- National Research Centre on Addictions, Ministry of Health and Social Development of the Russian Federation, Moscow, 119002, Russia
| | | | | |
Collapse
|
23
|
Li X, Wolf ME. Multiple faces of BDNF in cocaine addiction. Behav Brain Res 2014; 279:240-54. [PMID: 25449839 DOI: 10.1016/j.bbr.2014.11.018] [Citation(s) in RCA: 138] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2014] [Revised: 11/04/2014] [Accepted: 11/08/2014] [Indexed: 01/04/2023]
Abstract
Brain-derived neurotrophic factor (BDNF) has been found to play roles in many types of plasticity including drug addiction. Here, we focus on rodent studies over the past two decades that have demonstrated diverse roles of BDNF in models of cocaine addiction. First, we will provide an overview of studies showing that cocaine exposure alters (and generally increases) BDNF levels in reward-related regions including the ventral tegmental area, nucleus accumbens, prefrontal cortex, and amygdala. Then we will review evidence that BDNF contributes to behavioral changes in animal models of cocaine addiction, focusing on conditioned place preference, behavioral sensitization, maintenance and reinstatement of self-administration, and incubation of cocaine craving. Last, we will review the role of BDNF in synaptic plasticity, particularly as it relates to plasticity of AMPA receptor transmission after cocaine exposure. We conclude that BDNF regulates cocaine-induced behaviors in a highly complex manner that varies depending on the brain region (and even among different cell types within the same brain region), the nature of cocaine exposure, and the "addiction phase" examined (e.g., acquisition vs maintenance; early vs late withdrawal). These complexities make BDNF a daunting therapeutic target for treating cocaine addiction. However, recent clinical evidence suggests that the serum BDNF level may serve as a biomarker in cocaine addicts to predict future relapse, providing an alternative direction for exploring BDNF's potential relevance to treating cocaine addiction.
Collapse
Affiliation(s)
- Xuan Li
- Behavioral Neuroscience Research Branch, Intramural Research Program, NIDA/NIH/DHHS, Baltimore, MD, USA.
| | - Marina E Wolf
- Department of Neuroscience, Rosalind Franklin University of Medicine and Science, North Chicago, IL, USA
| |
Collapse
|
24
|
Raivio N, Miettinen P, Kiianmaa K. Innate BDNF expression is associated with ethanol intake in alcohol-preferring AA and alcohol-avoiding ANA rats. Brain Res 2014; 1579:74-83. [PMID: 25044407 DOI: 10.1016/j.brainres.2014.07.006] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2014] [Revised: 06/26/2014] [Accepted: 07/04/2014] [Indexed: 12/20/2022]
Abstract
We have shown recently that acute administration of ethanol modulates the expression of brain-derived neurotrophic factor (BDNF) in several rat brain areas known to be involved in the development of addiction to ethanol and other drugs of abuse, suggesting that BDNF may be a factor contributing to the neuroadaptive changes set in motion by ethanol exposure. The purpose of the present study was to further clarify the role of BDNF in reinforcement from ethanol and in the development of addiction to ethanol by specifying the effect of acute administration of ethanol (1.5 or 3.0 g/kg i.p.) on the expression profile of BDNF mRNA in the ventral tegmental area and in the terminal areas of the mesolimbic dopamine pathway in the brain of alcohol-preferring AA and alcohol-avoiding ANA rats, selected for high and low voluntary ethanol intake, respectively. The level of BDNF mRNA expression was higher in the amygdala and ventral tegmental area of AA than in those of ANA rats, and there was a trend for a higher level in the nucleus accumbens. In the amygdala and hippocampus, a biphasic change in the BDNF mRNA levels was detected: the levels were decreased at 3 and 6h but increased above the basal levels at 24h. Furthermore, there was a difference between the AA and ANA lines in the effect of ethanol, the ANA rats showing an increase in BDNF mRNA levels while such a change was not seen in AA rats. These findings suggest that the innate levels of BDNF expression may play a role in the mediation of the reinforcing effects of ethanol and in the control of ethanol intake.
Collapse
Affiliation(s)
- Noora Raivio
- Department of Alcohol, Drugs and Addiction, National Institute for Health and Welfare, POB 30, Helsinki 00271, Finland
| | - Pekka Miettinen
- Department of Alcohol, Drugs and Addiction, National Institute for Health and Welfare, POB 30, Helsinki 00271, Finland
| | - Kalervo Kiianmaa
- Department of Alcohol, Drugs and Addiction, National Institute for Health and Welfare, POB 30, Helsinki 00271, Finland.
| |
Collapse
|
25
|
Zhang J, Zhang X, Su H, Tao J, Xie Y, Han B, Lu Y, Wei Y, Sun H, Wang Y, Wu W, Zou S, Liang H, Zoghbi AW, Tang W, He J. Increased serum brain-derived neurotrophic factor levels during opiate withdrawal. Neurosci Lett 2014; 571:61-5. [DOI: 10.1016/j.neulet.2014.04.048] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2014] [Revised: 04/28/2014] [Accepted: 04/29/2014] [Indexed: 12/11/2022]
|
26
|
Nikulina EM, Johnston CE, Wang J, Hammer RP. Neurotrophins in the ventral tegmental area: Role in social stress, mood disorders and drug abuse. Neuroscience 2014; 282:122-38. [PMID: 24875178 DOI: 10.1016/j.neuroscience.2014.05.028] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2014] [Revised: 05/04/2014] [Accepted: 05/11/2014] [Indexed: 01/19/2023]
Abstract
This review discusses the impact of neurotrophins and other trophic factors, including fibroblast growth factor and glial cell line-derived neurotrophic factor, on mood disorders, weight regulation and drug abuse, with an emphasis on stress- and drug-induced changes in the ventral tegmental area (VTA). Neurotrophins, comprising nerve growth factor, brain-derived neurotrophic factor (BDNF), and neurotrophins 3 and 4/5 play important roles in neuronal plasticity and the development of different psychopathologies. In the VTA, most research has focused on the role of BDNF, because other neurotrophins are not found there in significant quantities. BDNF originating in the VTA provides trophic support to dopamine neurons. The diverse intracellular signaling pathways activated by BDNF may underlie precise physiological functions specific to the VTA. In general, VTA BDNF expression increases after psychostimulant exposures, and enhanced BDNF level in the VTA facilitates psychostimulant effects. The impact of VTA BDNF on the behavioral effects of psychostimulants relies primarily on its action within the mesocorticolimbic circuit. In the case of opiates, VTA BDNF expression and effects seem to be dependent on whether an animal is drug-naïve or has a history of drug use, only the latter of which is related to dopamine mechanisms. Social defeat stress that is continuous in mice or intermittent in rats increases VTA BDNF expression, and is associated with depressive and social avoidance behaviors. Intermittent social defeat stress induces persistent VTA BDNF expression that triggers psychostimulant cross-sensitization. Understanding the cellular and molecular substrates of neurotrophin effects may lead to novel therapeutic approaches for the prevention and treatment of substance use and mood disorders.
Collapse
Affiliation(s)
- E M Nikulina
- Department of Basic Medical Sciences, University of Arizona College of Medicine, Phoenix, AZ, USA.
| | - C E Johnston
- Department of Basic Medical Sciences, University of Arizona College of Medicine, Phoenix, AZ, USA; Interdisciplinary Neuroscience Program, Arizona State University, Tempe, AZ, USA
| | - J Wang
- Interdisciplinary Neuroscience Program, Arizona State University, Tempe, AZ, USA
| | - R P Hammer
- Department of Basic Medical Sciences, University of Arizona College of Medicine, Phoenix, AZ, USA; Interdisciplinary Neuroscience Program, Arizona State University, Tempe, AZ, USA; Department of Pharmacology and Department of Psychiatry, University of Arizona College of Medicine, Tucson, AZ, USA
| |
Collapse
|
27
|
Viola TW, Tractenberg SG, Levandowski ML, Pezzi JC, Bauer ME, Teixeira AL, Grassi-Oliveira R. Neurotrophic factors in women with crack cocaine dependence during early abstinence: the role of early life stress. J Psychiatry Neurosci 2014; 39:206-14. [PMID: 24331739 PMCID: PMC3997606 DOI: 10.1503/jpn.130027] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/15/2013] [Revised: 06/26/2013] [Accepted: 10/11/2013] [Indexed: 12/12/2022] Open
Abstract
BACKGROUND Neurotrophic factors have been investigated in the pathophysiology of alcohol and drug dependence and have been related to early life stress driving developmental programming of neuroendocrine systems. METHODS We conducted a follow-up study that aimed to assess the plasma levels of glial cell line-derived neurotrophic factor (GDNF), brain-derived neurotrophic factor (BDNF), nerve growth factor (NGF), neurotrophin-3 (NT3) and neurotrophin-4/5 (NT4/5) in crack users during 3 weeks of early abstinence in comparison with healthy controls. We performed a comprehensive clinical assessment in female inpatients with crack cocaine dependence (separated into 2 groups: participants with (CSA+) and without (CSA-) a history of childhood sexual abuse) and a group of nonuser control participants. RESULTS Our sample included 104 women with crack cocaine dependence and 22 controls; of the women who used crack cocaine, 22 had a history of childhood sexual abuse and 82 did not. The GDNF plasma levels in the CSA+ group increased dramatically during 3 weeks of detoxification. In contrast, those in the CSA- group showed lower and stable levels of GDNF under the same conditions. Compared with the control group, BDNF plasma levels remained elevated and NGF levels were reduced during early abstinence. We found no differences in NT3 and NT4/5 between the patients and controls. However, within-group analyses showed that the CSA+ group exhibited higher levels of NT4/5 than the CSA- group at the end of detoxification. LIMITATIONS Some of the participants were using neuroleptics, mood stabilizers or antidepressants; our sample included only women; memory bias could not be controlled; and we did not investigate the possible confounding effects of other forms of stress during childhood. CONCLUSION This study supports the association between early life stress and peripheral neurotrophic factor levels in crack cocaine users. During early abstinence, plasmastic GDNF and NT4/5 were the only factors to show changes associated with a history of childhood sexual abuse.
Collapse
Affiliation(s)
- Thiago Wendt Viola
- Viola, Tractenberg, Levandowski, Bauer, Grassi-Oliveira — Centre of Studies and Research in Traumatic Stress, PostGraduate Program in Psychology, Pontifical Catholic University of the Rio Grande do Sul (PUCRS), Porto Alegre, Brazil; Pezzi — Post-Graduate Program in Health Sciences, Universidade Federal de Ciências da Saúde de Porto Alegre (UFCSPA), Porto Alegre, Brazil; Bauer, Grassi-Oliveira — Laboratory of Immunosenescence, Institute of Biomedical Research, Pontifical Catholic University of the Rio Grande do Sul (PUCRS), Porto Alegre, Brazil; Teixeira — Neuroscience Program, Federal University of Minas Gerais (UFMG), Belo Horizonte, Brazil
| | - Saulo Gantes Tractenberg
- Viola, Tractenberg, Levandowski, Bauer, Grassi-Oliveira — Centre of Studies and Research in Traumatic Stress, PostGraduate Program in Psychology, Pontifical Catholic University of the Rio Grande do Sul (PUCRS), Porto Alegre, Brazil; Pezzi — Post-Graduate Program in Health Sciences, Universidade Federal de Ciências da Saúde de Porto Alegre (UFCSPA), Porto Alegre, Brazil; Bauer, Grassi-Oliveira — Laboratory of Immunosenescence, Institute of Biomedical Research, Pontifical Catholic University of the Rio Grande do Sul (PUCRS), Porto Alegre, Brazil; Teixeira — Neuroscience Program, Federal University of Minas Gerais (UFMG), Belo Horizonte, Brazil
| | - Mateus Luz Levandowski
- Viola, Tractenberg, Levandowski, Bauer, Grassi-Oliveira — Centre of Studies and Research in Traumatic Stress, PostGraduate Program in Psychology, Pontifical Catholic University of the Rio Grande do Sul (PUCRS), Porto Alegre, Brazil; Pezzi — Post-Graduate Program in Health Sciences, Universidade Federal de Ciências da Saúde de Porto Alegre (UFCSPA), Porto Alegre, Brazil; Bauer, Grassi-Oliveira — Laboratory of Immunosenescence, Institute of Biomedical Research, Pontifical Catholic University of the Rio Grande do Sul (PUCRS), Porto Alegre, Brazil; Teixeira — Neuroscience Program, Federal University of Minas Gerais (UFMG), Belo Horizonte, Brazil
| | - Júlio Carlos Pezzi
- Viola, Tractenberg, Levandowski, Bauer, Grassi-Oliveira — Centre of Studies and Research in Traumatic Stress, PostGraduate Program in Psychology, Pontifical Catholic University of the Rio Grande do Sul (PUCRS), Porto Alegre, Brazil; Pezzi — Post-Graduate Program in Health Sciences, Universidade Federal de Ciências da Saúde de Porto Alegre (UFCSPA), Porto Alegre, Brazil; Bauer, Grassi-Oliveira — Laboratory of Immunosenescence, Institute of Biomedical Research, Pontifical Catholic University of the Rio Grande do Sul (PUCRS), Porto Alegre, Brazil; Teixeira — Neuroscience Program, Federal University of Minas Gerais (UFMG), Belo Horizonte, Brazil
| | - Moisés Evandro Bauer
- Viola, Tractenberg, Levandowski, Bauer, Grassi-Oliveira — Centre of Studies and Research in Traumatic Stress, PostGraduate Program in Psychology, Pontifical Catholic University of the Rio Grande do Sul (PUCRS), Porto Alegre, Brazil; Pezzi — Post-Graduate Program in Health Sciences, Universidade Federal de Ciências da Saúde de Porto Alegre (UFCSPA), Porto Alegre, Brazil; Bauer, Grassi-Oliveira — Laboratory of Immunosenescence, Institute of Biomedical Research, Pontifical Catholic University of the Rio Grande do Sul (PUCRS), Porto Alegre, Brazil; Teixeira — Neuroscience Program, Federal University of Minas Gerais (UFMG), Belo Horizonte, Brazil
| | - Antonio Lúcio Teixeira
- Viola, Tractenberg, Levandowski, Bauer, Grassi-Oliveira — Centre of Studies and Research in Traumatic Stress, PostGraduate Program in Psychology, Pontifical Catholic University of the Rio Grande do Sul (PUCRS), Porto Alegre, Brazil; Pezzi — Post-Graduate Program in Health Sciences, Universidade Federal de Ciências da Saúde de Porto Alegre (UFCSPA), Porto Alegre, Brazil; Bauer, Grassi-Oliveira — Laboratory of Immunosenescence, Institute of Biomedical Research, Pontifical Catholic University of the Rio Grande do Sul (PUCRS), Porto Alegre, Brazil; Teixeira — Neuroscience Program, Federal University of Minas Gerais (UFMG), Belo Horizonte, Brazil
| | - Rodrigo Grassi-Oliveira
- Viola, Tractenberg, Levandowski, Bauer, Grassi-Oliveira — Centre of Studies and Research in Traumatic Stress, PostGraduate Program in Psychology, Pontifical Catholic University of the Rio Grande do Sul (PUCRS), Porto Alegre, Brazil; Pezzi — Post-Graduate Program in Health Sciences, Universidade Federal de Ciências da Saúde de Porto Alegre (UFCSPA), Porto Alegre, Brazil; Bauer, Grassi-Oliveira — Laboratory of Immunosenescence, Institute of Biomedical Research, Pontifical Catholic University of the Rio Grande do Sul (PUCRS), Porto Alegre, Brazil; Teixeira — Neuroscience Program, Federal University of Minas Gerais (UFMG), Belo Horizonte, Brazil
| |
Collapse
|
28
|
Navarro-Zaragoza J, Martínez-Laorden E, Mora L, Hidalgo J, Milanés M, Laorden M. Cardiac adverse effects of naloxone-precipitated morphine withdrawal on right ventricle: Role of corticotropin-releasing factor (CRF) 1 receptor. Toxicol Appl Pharmacol 2014; 275:28-35. [DOI: 10.1016/j.taap.2013.12.021] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2013] [Revised: 11/29/2013] [Accepted: 12/28/2013] [Indexed: 01/14/2023]
|
29
|
Ren Q, Zhang JC, Ma M, Fujita Y, Wu J, Hashimoto K. 7,8-Dihydroxyflavone, a TrkB agonist, attenuates behavioral abnormalities and neurotoxicity in mice after administration of methamphetamine. Psychopharmacology (Berl) 2014; 231:159-66. [PMID: 23934209 DOI: 10.1007/s00213-013-3221-7] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/23/2013] [Accepted: 07/18/2013] [Indexed: 01/14/2023]
Abstract
RATIONALE It is widely recognized that methamphetamine (METH) induces behavioral abnormalities and dopaminergic neurotoxicity in the brain. Several lines of evidence suggest a role for brain-derived neurotrophic factor (BDNF) and its specific receptor, tropomyosin-related kinase (TrkB), in METH-induced behavioral abnormalities. OBJECTIVE In this study, we examined whether 7,8-dihydroxyflavone (7,8-DHF), a novel potent TrkB agonist, could attenuate behavioral abnormalities and dopaminergic neurotoxicity in mice after administration of METH. RESULTS Pretreatment with 7,8-DHF (3.0, 10, or 30 mg/kg), but not the inactive TrkB compound, 5,7-dihydroxyflavone (5,7-DHF) (30 mg/kg), attenuated hyperlocomotion in mice after a single administration of METH (3.0 mg/kg), in a dose-dependent manner. The development of behavioral sensitization after repeated administration of METH (3.0 mg/kg/day, once daily for 5 days) was significantly attenuated by pretreatment with 7,8-DHF (10 mg/kg). Furthermore, pretreatment and subsequent administration of 7,8-DHF (10 mg/kg) attenuated the reduction of dopamine transporter (DAT) in the striatum after repeated administration of METH (3.0 mg/kg × 3 at 3-hourly intervals). Treatment with ANA-12 (0.5 mg/kg), a potent TrkB antagonist, blocked the protective effects of 7,8-DHF on the METH-induced reduction of DAT in the striatum. Moreover, 7,8-DHF attenuated microglial activation in the striatum after repeated administration of METH. CONCLUSIONS These findings suggest that 7,8-DHF can ameliorate behavioral abnormalities as well as dopaminergic neurotoxicity in mice after administration of METH. It is likely, therefore, that TrkB agonists such as 7,8-DHF may prove to be potential therapeutic drugs for several symptoms associated with METH abuse in humans.
Collapse
Affiliation(s)
- Qian Ren
- Division of Clinical Neuroscience, Chiba University Center for Forensic Mental Health, 1-8-1 Inohana, Chiba, 260-8670, Japan
| | | | | | | | | | | |
Collapse
|
30
|
Wade CL, Krumenacher P, Kitto KF, Peterson CD, Wilcox GL, Fairbanks CA. Effect of chronic pain on fentanyl self-administration in mice. PLoS One 2013; 8:e79239. [PMID: 24260176 PMCID: PMC3829846 DOI: 10.1371/journal.pone.0079239] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2013] [Accepted: 09/19/2013] [Indexed: 11/18/2022] Open
Abstract
The development of opioid addiction in subjects with established chronic pain is an area that is poorly understood. It is critically important to clearly understand the neurobiology associated with propensity toward conversion to addiction under conditions of chronic pain. To pose the question whether the presence of chronic pain influences motivation to self-administer opioids for reward, we applied a combination of rodent models of chronic mechanical hyperalgesia and opioid self-administration. We studied fentanyl self-administration in mice under three conditions that induce chronic mechanical hyperalgesia: inflammation, peripheral nerve injury, and repeated chemotherapeutic injections. Responding for fentanyl was compared among these conditions and their respective standard controls (naïve condition, vehicle injection or sham surgery). Acquisition of fentanyl self-administration behavior was reduced or absent in all three conditions of chronic hyperalgesia relative to control mice with normal sensory thresholds. To control for potential impairment in ability to learn the lever-pressing behavior or perform the associated motor tasks, all three groups were evaluated for acquisition of food-maintained responding. In contrast to the opioid, chronic hyperalgesia did not interfere with the reinforcing effect of food. These studies indicate that the establishment of chronic hyperalgesia is associated with reduced or ablated motivation to seek opioid reward in mice.
Collapse
Affiliation(s)
- Carrie L. Wade
- Department of Pharmacology, University of Minnesota, Minneapolis, Minnesota, United States of America
- Center for Pain Research, University of Minnesota, Minneapolis, Minnesota, United States of America
| | - Perry Krumenacher
- Department of Neuroscience, University of Minnesota, Minneapolis, Minnesota, United States of America
- Center for Pain Research, University of Minnesota, Minneapolis, Minnesota, United States of America
| | - Kelley F. Kitto
- Department of Neuroscience, University of Minnesota, Minneapolis, Minnesota, United States of America
- Center for Pain Research, University of Minnesota, Minneapolis, Minnesota, United States of America
| | - Cristina D. Peterson
- Department of Pharmaceutics, University of Minnesota, Minneapolis, Minnesota, United States of America
- Center for Pain Research, University of Minnesota, Minneapolis, Minnesota, United States of America
| | - George L. Wilcox
- Department of Pharmacology, University of Minnesota, Minneapolis, Minnesota, United States of America
- Department of Neuroscience, University of Minnesota, Minneapolis, Minnesota, United States of America
- Center for Pain Research, University of Minnesota, Minneapolis, Minnesota, United States of America
| | - Carolyn A. Fairbanks
- Department of Pharmacology, University of Minnesota, Minneapolis, Minnesota, United States of America
- Department of Neuroscience, University of Minnesota, Minneapolis, Minnesota, United States of America
- Department of Pharmaceutics, University of Minnesota, Minneapolis, Minnesota, United States of America
- Center for Pain Research, University of Minnesota, Minneapolis, Minnesota, United States of America
- * E-mail:
| |
Collapse
|
31
|
Abstract
This study examined the involvement of the brain stress system in the reinforcing effects of morphine. One group of mice was conditioned to morphine using the conditioned place preference (CPP) paradigm and the other group received morphine in a home-cage (non-conditioned). Adrenocorticotropic hormone and corticosterone levels were measured by radioimmunoassay; phospho (p) CREB expression and the number of corticotropin-releasing factor (CRF) neurons and fibres were measured by immunohistochemistry in different brain areas. We observed that the number of CRF neurons in the paraventricular nucleus (PVN) was increased after morphine-induced CPP, which was paralleled with enhanced CRF-immunoreactivity fibres in the nucleus tractus solitarius (NTS) and ventral tegmental area (VTA) vs. home-cage group injected with morphine. Morphine exposure induced an increase in CREB phosphorylated at Ser133 in the PVN and central amygdale (CeA), whereas mice exhibiting morphine CPP had higher levels of pCREB in the PVN, CeA and bed nucleus of the stria terminalis (BNST). We also found that most of the CRF-positive neurons in the PVN, CeA and BNST co-express pCREB after morphine CPP expression, suggesting that the drug-associated environmental contexts can elicit neuronal activity in the brain stress system. From the present results it is clear that exposure to a drug-associated context remains a potent activator of signalling pathways leading to CRF activation in the brain stress system.
Collapse
|
32
|
Greenwald MK, Steinmiller CL, Sliwerska E, Lundahl L, Burmeister M. BDNF Val(66)Met genotype is associated with drug-seeking phenotypes in heroin-dependent individuals: a pilot study. Addict Biol 2013; 18:836-45. [PMID: 22339949 DOI: 10.1111/j.1369-1600.2011.00431.x] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Abstract
Brain-derived neurotrophic factor (BDNF) Val(66)Met genotype has been associated with neurobehavioral deficits. To examine its relevance for addiction, we examined BDNF genotype differences in drug-seeking behavior. Heroin-dependent volunteers (n = 128) completed an interview that assessed past-month naturalistic drug-seeking/use behaviors. In African Americans (n = 74), the Met allele was uncommon (carrier frequency 6.8%); thus, analyses focused on European Americans (n = 54), in whom the Met allele was common (carrier frequency 37.0%). In their natural setting, Met carriers (n = 20) reported more time- and cost-intensive heroin-seeking and more cigarette use than Val homozygotes (n = 34). BDNF Val(66)Met genotype predicted 18.4% of variance in 'weekly heroin investment' (purchasing time × amount × frequency). These data suggest that the BDNF Met allele may confer a 'preferred drug-invested' phenotype, resistant to moderating effects of higher drug prices and non-drug reinforcement. These preliminary hypothesis-generating findings require replication, but are consistent with pre-clinical data that demonstrate neurotrophic influence in drug reinforcement. Whether this genotype is relevant to other abused substances besides opioids or nicotine, or treatment response, remains to be determined.
Collapse
Affiliation(s)
- Mark K Greenwald
- Substance Abuse Research Division, Department of Psychiatry and Behavioral Neurosciences, Wayne State University School of Medicine, Detroit, MI, USA.
| | | | | | | | | |
Collapse
|
33
|
Fernàndez-Castillo N, Roncero C, Grau-Lopez L, Barral C, Prat G, Rodriguez-Cintas L, Sánchez-Mora C, Gratacòs M, Ramos-Quiroga J, Casas M, Ribasés M, Cormand B. Association study of 37 genes related to serotonin and dopamine neurotransmission and neurotrophic factors in cocaine dependence. GENES BRAIN AND BEHAVIOR 2013; 12:39-46. [DOI: 10.1111/gbb.12013] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/02/2012] [Revised: 11/22/2012] [Accepted: 12/10/2012] [Indexed: 12/22/2022]
Affiliation(s)
| | | | | | | | - G. Prat
- Mental Health Division, Fundació Althaia; Hospital San Joan de Déu, Manresa; Catalonia; Spain
| | | | | | | | | | | | | | | |
Collapse
|
34
|
Koo JW, Mazei-Robison MS, Chaudhury D, Juarez B, LaPlant Q, Ferguson D, Feng J, Sun H, Scobie KN, Damez-Werno D, Crumiller M, Ohnishi YN, Ohnishi YH, Mouzon E, Dietz DM, Lobo MK, Neve RL, Russo SJ, Han MH, Nestler EJ. BDNF is a negative modulator of morphine action. Science 2012; 338:124-8. [PMID: 23042896 DOI: 10.1126/science.1222265] [Citation(s) in RCA: 134] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
Brain-derived neurotrophic factor (BDNF) is a key positive regulator of neural plasticity, promoting, for example, the actions of stimulant drugs of abuse such as cocaine. We discovered a surprising opposite role for BDNF in countering responses to chronic morphine exposure. The suppression of BDNF in the ventral tegmental area (VTA) enhanced the ability of morphine to increase dopamine (DA) neuron excitability and promote reward. In contrast, optical stimulation of VTA DA terminals in nucleus accumbens (NAc) completely reversed the suppressive effect of BDNF on morphine reward. Furthermore, we identified numerous genes in the NAc, a major target region of VTA DA neurons, whose regulation by BDNF in the context of chronic morphine exposure mediated this counteractive function. These findings provide insight into the molecular basis of morphine-induced neuroadaptations in the brain's reward circuitry.
Collapse
Affiliation(s)
- Ja Wook Koo
- Fishberg Department of Neuroscience and Friedman Brain Institute, Mount Sinai School of Medicine, New York, NY 10029, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
35
|
Almela P, Navarro-Zaragoza J, García-Carmona JA, Mora L, Hidalgo J, Milanés MV, Laorden ML. Role of corticotropin-releasing factor (CRF) receptor-1 on the catecholaminergic response to morphine withdrawal in the nucleus accumbens (NAc). PLoS One 2012; 7:e47089. [PMID: 23071721 PMCID: PMC3468529 DOI: 10.1371/journal.pone.0047089] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2012] [Accepted: 09/10/2012] [Indexed: 11/19/2022] Open
Abstract
Stress induces the release of the peptide corticotropin-releasing factor (CRF) into the ventral tegmental area (VTA), and also increases dopamine (DA) levels in brain regions receiving dense VTA input. Since the role of stress in drug addiction is well established, the present study examined the possible involvement of CRF1 receptor in the interaction between morphine withdrawal and catecholaminergic pathways in the reward system. The effects of naloxone-precipitated morphine withdrawal on signs of withdrawal, hypothalamo-pituitary-adrenocortical (HPA) axis activity, dopamine (DA) and noradrenaline (NA) turnover in the nucleus accumbens (NAc) and activation of VTA dopaminergic neurons, were investigated in rats pretreated with vehicle or CP-154,526 (selective CRF1R antagonist). CP-154,526 attenuated the increases in body weight loss and suppressed some of withdrawal signs. Pretreatment with CRF1 receptor antagonist resulted in no significant modification of the increased NA turnover at NAc or plasma corticosterone levels that were seen during morphine withdrawal. However, blockade of CRF1 receptor significantly reduced morphine withdrawal-induced increases in plasma adrenocorticotropin (ACTH) levels, DA turnover and TH phosphorylation at Ser40 in the NAc. In addition, CP-154,526 reduced the number of TH containing neurons expressing c-Fos in the VTA after naloxone-precipitated morphine withdrawal. Altogether, these results support the idea that VTA dopaminergic neurons are activated in response to naloxone-precipitated morphine withdrawal and suggest that CRF1 receptors are involved in the activation of dopaminergic pathways which project to NAc.
Collapse
Affiliation(s)
- Pilar Almela
- Departamento de Farmacología, Facultad de Medicina, Universidad de Murcia, Murcia, Spain
| | | | | | - Lucía Mora
- Departamento de Farmacología, Facultad de Medicina, Universidad de Murcia, Murcia, Spain
| | - Juana Hidalgo
- Departamento de Farmacología, Facultad de Medicina, Universidad de Murcia, Murcia, Spain
| | - María-Victoria Milanés
- Departamento de Farmacología, Facultad de Medicina, Universidad de Murcia, Murcia, Spain
| | - María-Luisa Laorden
- Departamento de Farmacología, Facultad de Medicina, Universidad de Murcia, Murcia, Spain
| |
Collapse
|
36
|
Martínez-Laorden E, Hurle MA, Milanés MV, Laorden ML, Almela P. Morphine withdrawal activates hypothalamic-pituitary-adrenal axis and heat shock protein 27 in the left ventricle: the role of extracellular signal-regulated kinase. J Pharmacol Exp Ther 2012; 342:665-75. [PMID: 22647273 DOI: 10.1124/jpet.112.193581] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
The negative affective states of withdrawal involve the recruitment of brain and peripheral stress circuitry [e.g., noradrenergic activity, induction of the hypothalamo-pituitary-adrenocortical (HPA) axis, and the expression and activation of heat shock proteins (Hsps)]. The present study investigated the role of extracellular signal-regulated protein kinase (ERK) and β-adrenoceptor on the response of stress systems to morphine withdrawal by the administration of [amino[(4-aminophenyl)thio]methylene]-2-(trifluoromethyl)benzeneacetonitrile (SL327), a selective inhibitor of ERK activation, or propranolol (a β-adrenoceptor antagonist). Dependence on morphine was induced by a 7-day subcutaneous implantation of morphine pellets. Morphine withdrawal was precipitated on day 8 by the injection of naloxone (2 mg/kg s.c.). Plasma concentrations of adrenocorticotropin and corticosterone were determined by radioimmunoassay; noradrenaline (NA) turnover in left ventricle was determined by high-performance liquid chromatography; and catechol-O-methyl transferase (COMT) and Hsp27 expression and phosphorylation at Ser82 were determined by quantitative blot immunolabeling. Morphine-withdrawn rats showed an increase of NA turnover and COMT expression in parallel with an enhancement of adrenocorticotropin and plasma corticosterone concentrations. In addition, we observed an enhancement of Hsp27 expression and phosphorylation. Pretreatment with SL327 or propranolol significantly reduced morphine withdrawal-induced increases of plasma adrenocorticotropin and Hsp27 phosphorylation at Ser82 without any changes in plasma corticosterone levels. The present findings demonstrate that morphine withdrawal is capable of inducing the activation of HPA axis in parallel with an enhancement of Hsp27 expression and Hsp27 phosphorylation at Ser82 and suggest a role for β-adrenoceptors and ERK pathways in mediating morphine-withdrawal activation of the HPA axis and cellular stress response.
Collapse
Affiliation(s)
- E Martínez-Laorden
- Department of Pharmacology, Faculty of Medicine, University of Murcia, Murcia, Spain
| | | | | | | | | |
Collapse
|
37
|
Gómez-Milanés I, Almela P, García-Carmona JA, Salud García-Gutiérrez M, Aracil-Fernández A, Manzanares J, Victoria Milanés Maquilón M, Luisa Laorden M. Accumbal dopamine, noradrenaline and serotonin activity after naloxone-conditioned place aversion in morphine-dependent mice. Neurochem Int 2012; 61:433-40. [DOI: 10.1016/j.neuint.2012.06.011] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2011] [Revised: 06/04/2012] [Accepted: 06/11/2012] [Indexed: 10/28/2022]
|
38
|
Thioredoxin-1 expression regulated by morphine in SH-SY5Y cells. Neurosci Lett 2012; 523:50-5. [DOI: 10.1016/j.neulet.2012.06.039] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2012] [Revised: 06/13/2012] [Accepted: 06/15/2012] [Indexed: 11/20/2022]
|
39
|
Raivio N, Tiraboschi E, Saarikoski ST, Castrén E, Kiianmaa K. Brain-derived neurotrophic factor expression after acute administration of ethanol. Eur J Pharmacol 2012; 687:9-13. [PMID: 22546227 DOI: 10.1016/j.ejphar.2012.04.021] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2012] [Accepted: 04/13/2012] [Indexed: 01/27/2023]
Abstract
Earlier findings suggest that, in addition to its well-known neurotrophic role, brain-derived neurotrophic factor (BDNF) is also involved in the rewarding and reinforcing effects of drugs of abuse. The purpose of the present study was to examine the effects of acute administration of ethanol (1.25 or 2.5 g/kg i.p.) on the expression profile of BDNF in the rat brain by determining the BDNF mRNA expression in the frontal cortex, nucleus accumbens, amygdala, hippocampus, and ventral tegmental area. Ethanol decreased BDNF mRNA levels dose-dependently in the hippocampus, and after the higher ethanol dose in the frontal cortex, nucleus accumbens and amygdala, while increasing them in the ventral tegmental area. Furthermore, BDNF mRNA expression was found to be regulated in a temporally different manner in all investigated brain areas. These data suggest that BDNF is involved in the acute effects of ethanol, but separate brain areas may be differentially engaged in the mediation of these effects.
Collapse
Affiliation(s)
- Noora Raivio
- Department of Alcohol, Drugs and Addiction, National Institute for Health and Welfare, Helsinki, Finland
| | | | | | | | | |
Collapse
|
40
|
Luo FC, Qi L, Lv T, Wang SD, Liu H, Nakamura H, Yodoi J, Bai J. Geranylgeranylacetone protects mice against morphine-induced hyperlocomotion, rewarding effect, and withdrawal syndrome. Free Radic Biol Med 2012; 52:1218-27. [PMID: 22285390 DOI: 10.1016/j.freeradbiomed.2012.01.004] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/22/2011] [Revised: 01/03/2012] [Accepted: 01/09/2012] [Indexed: 10/14/2022]
Abstract
There are few efficacious interventions to combat morphine dependence. Thioredoxin-1 (Trx-1) and heat shock protein 70 (Hsp70) are emerging as important modulators of neuronal function. They have been shown to be involved in cellular protective mechanisms against a variety of toxic stressors. This study was designed to investigate the effects of geranylgeranylacetone (GGA), a pharmacological inducer of Trx-1 and Hsp70, on morphine-induced hyperlocomotion, rewarding effect, and withdrawal syndrome. Trx-1 and Hsp70 expression was increased in the frontal cortex, hippocampus, ventral tegmental area, and nucleus accumbens of mice after GGA treatment. GGA administration reduced morphine-induced motor activity and inhibited conditioned place preference. GGA markedly attenuated the morphine-naloxone-induced withdrawal signs, including jumping, rearing, and forepaw tremor. Furthermore, the activation of cAMP-responsive element-binding protein and the expression of ΔFosB and cyclin-dependent kinase 5 were decreased in the nucleus accumbens by GGA treatment after morphine withdrawal. In the nucleus accumbens, GGA enhanced morphine-induced expression of Trx-1 and Hsp70 after morphine withdrawal. These results suggest that strengthening the expression of Trx-1 and Hsp70 in the brain by using noncytotoxic pharmacological inducers may provide a novel therapeutic strategy for morphine dependence. GGA could be a safe and novel therapeutic agent for morphine dependence.
Collapse
Affiliation(s)
- Fu-Cheng Luo
- Faculty of Environmental Science and Engineering, Kunming University of Science and Technology, Kunming 650500, China
| | | | | | | | | | | | | | | |
Collapse
|
41
|
Abstract
Brain derived neurotrophic factor (BDNF) is the most prevalent growth factor in the central nervous system (CNS). It is essential for the development of the CNS and for neuronal plasticity. Because BDNF plays a crucial role in development and plasticity of the brain, it is widely implicated in psychiatric diseases. This review provides a summary of clinical and preclinical evidence for the involvement of this ubiquitous growth factor in major depressive disorder, schizophrenia, addiction, Rett syndrome, as well as other psychiatric and neurodevelopmental diseases. In addition, the review includes a discussion of the role of BDNF in the mechanism of action of pharmacological therapies currently used to treat these diseases, such antidepressants and antipsychotics. The review also covers a critique of experimental therapies such as BDNF mimetics and discusses the value of BDNF as a target for future drug development.
Collapse
Affiliation(s)
- Anita E Autry
- Department of Psychiatry, The University of Texas Southwestern Medical Center, 5323 Harry Hines Blvd, Dallas, TX 75390-9070, USA
| | | |
Collapse
|
42
|
Restricted role of CRF1 receptor for the activity of brainstem catecholaminergic neurons in the negative state of morphine withdrawal. Psychopharmacology (Berl) 2012; 220:379-93. [PMID: 21947312 DOI: 10.1007/s00213-011-2478-y] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/29/2011] [Accepted: 09/01/2011] [Indexed: 01/24/2023]
Abstract
RATIONALE Evidence suggests that corticotropin-releasing factor (CRF) system is an important mediator in the negative symptoms of opioid withdrawal. OBJECTIVES We used genetically engineered mice lacking functional CRF receptor-1 (CRF1R) levels to study the role for CRF/CRF1R pathways in the negative affective states of opioid withdrawal. METHODS Wild-type and CRF1R(-/-) offspring of CRF1R(+/-) breeders were identified by PCR analysis of tail DNA and were rendered dependent on morphine via intraperitoneal injection of increasing doses of morphine (10-60 mg/kg). Negative state associated with opioid withdrawal was examined by using conditioned place aversion (CPA), TH expression and TH phosphorylation were measured in different brain regions involved in addictive behaviours using immunohistochemistry. RESULTS The weight loss in morphine withdrawn CRF1R(-/-) animals was significantly (p < 0.05) lower versus wild-type. The aversion for environmental cues paired with opioid withdrawal was lower (p < 0.001) in the CRF1R-deficient versus wild-type. Using dual immunolabeling for c-Fos, data show that naloxone-induced withdrawal increases the number of TH positive neurons phosphorylated at Ser40 or Ser31 that coexpress c-Fos in the nucleus of tractus solitarius (NTS)-A2 from wild-type and CRF(-/-) deficient mice. By contrast, the number of phospho-Ser40 or phospho-Ser31 positive neurons expressing c-Fos was lower in the ventrolateral medulla (VLM)-A1 in CRF(-/-)-deficient mice. CONCLUSION Our study demonstrates an increased activity of brainstem catecholaminergic neurons after CPA induced by morphine withdrawal suggesting that CRF1R is implicated in the activation of A1 neurons and provides evidence that this receptor is involved in the body weight loss and in the negative aversive effects of morphine withdrawal.
Collapse
|
43
|
Sharma HS, Ali SF, Patnaik R, Zimmermann-Meinzingen S, Sharma A, Muresanu DF. Cerebrolysin Attenuates Heat Shock Protein (HSP 72 KD) Expression in the Rat Spinal Cord Following Morphine Dependence and Withdrawal: Possible New Therapy for Pain Management. Curr Neuropharmacol 2011; 9:223-35. [PMID: 21886595 PMCID: PMC3137188 DOI: 10.2174/157015911795017100] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2009] [Revised: 04/17/2010] [Accepted: 05/26/2010] [Indexed: 12/20/2022] Open
Abstract
The possibility that pain perception and processing in the CNS results in cellular stress and may influence heat shock protein (HSP) expression was examined in a rat model of morphine dependence and withdrawal. Since activation of pain pathways result in exhaustion of growth factors, we examined the influence of cerebrolysin, a mixture of potent growth factors (BDNF, GDNF, NGF, CNTF etc,) on morphine induced HSP expression. Rats were administered morphine (10 mg/kg, s.c. /day) for 12 days and the spontaneous withdrawal symptoms were developed by cessation of the drug administration on day 13th that were prominent on day 14th and continued up to day 15th (24 to 72 h periods). In a separate group of rats, cerebrolysin was infused intravenously (5 ml/kg) once daily from day one until day 15th. In these animals, morphine dependence and withdrawal along with HSP immunoreactivity was examined using standard protocol. In untreated group mild HSP immunoreaction was observed during morphine tolerance, whereas massive upregulation of HSP was seen in CNS during withdrawal phase that correlated well with the withdrawal symptoms and neuronal damage. Pretreatment with cerebrolysin did not affect morphine tolerance but reduced the HSP expression during this phase. Furthermore, cerebrolysin reduced the withdrawal symptoms on day 14th to 15th. Taken together these observations suggest that cellular stress plays an important role in morphine induced pain pathology and exogenous supplement of growth factors, i.e. cerebrolysin attenuates HSP expression in the CNS and induce neuroprotection. This indicates a new therapeutic role of cerebrolysin in the pathophysiology of drugs of abuse, not reported earlier.
Collapse
Affiliation(s)
- Hari S Sharma
- Laboratory of Cerebrovascular Research, Department of Surgical Sciences, Anesthesiology & Intensive Care Medicine, University Hospital, Uppsala University, SE-75185 Uppsala Sweden
| | | | | | | | | | | |
Collapse
|
44
|
Martín F, Mora L, Laorden M, Milanés M. Protein kinase C phosphorylates the cAMP response element binding protein in the hypothalamic paraventricular nucleus during morphine withdrawal. Br J Pharmacol 2011; 163:857-75. [PMID: 21615389 DOI: 10.1111/j.1476-5381.2011.01287.x] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
BACKGROUND AND PURPOSE Exposure to drugs of abuse or stress results in adaptation in the brain involving changes in gene expression and transcription factors. Morphine withdrawal modulates gene expression through various second-messenger signal transduction systems. Here, we investigated changes in activation of the transcription factor, cAMP-response element binding protein (CREB), in the hypothalamic paraventricular nucleus (PVN) and the kinases that may mediate the morphine withdrawal-triggered activation of CREB and the response of the hypothalamic-pituitary-adrenocortical (HPA) axis after naloxone-induced morphine withdrawal. EXPERIMENTAL APPROACH The effects of morphine dependence and withdrawal, phosphorylated CREB (pCREB), corticotrophin-releasing factor (CRF) expression in the PVN and HPA axis activity were measured using immunoblotting, immunohistochemistry and radioimmunoassay in controls and in morphine-dependent rats, withdrawn with naloxone and pretreated with vehicle, calphostin C, chelerythrine (inhibitors of protein kinase C (PKC) or SL-327 [inhibitor of extracellular signal regulated kinase (ERK) kinase]. In addition, changes in PKCα and PKCγ immunoreactivity were measured after 60 min of withdrawal. KEY RESULTS In morphine-withdrawn rats, pCREB immunoreactivity was increased within CRF immunoreactive neurons in the PVN and plasma corticosterone levels were raised. SL-327, at doses that reduced the augmented pERK levels in the PVN, did not attenuate the rise in pCREB immunoreactivity or plasma corticosterone secretion. In contrast, PKC inhibition reduced the withdrawal-triggered rise in pCREB, pERK1/2 and corticosterone secretion. CONCLUSIONS AND IMPLICATIONS PKC mediated, in part, both CREB activation and the HPA response to morphine withdrawal. The ERK kinase/ERK pathway might not be necessary for either activation of CREB or HPA axis hyperactivity.
Collapse
Affiliation(s)
- F Martín
- Group of Cellular and Molecular Pharmacology, Department of Pharmacology, University School of Medicine, Murcia, Spain
| | | | | | | |
Collapse
|
45
|
Li ZH, Liu YF, Li KN, Duanmu HZ, Chang ZQ, Li ZQ, Zhang SZ, Xu Y. Analysis of functional and pathway association of differential co-expressed genes: a case study in drug addiction. J Biomed Inform 2011; 45:30-6. [PMID: 21907308 DOI: 10.1016/j.jbi.2011.08.014] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2011] [Revised: 08/18/2011] [Accepted: 08/24/2011] [Indexed: 10/17/2022]
Abstract
Drug addiction has been considered as a kind of chronic relapsing brain disease influenced by both genetic and environmental factors. At present, many causative genes and pathways related to diverse kinds of drug addiction have been discovered, while less attention has been paid to common mechanisms shared by different drugs underlying addiction. By applying a co-expression meta-analysis method to mRNA expression profiles of alcohol, cocaine, heroin addicted and normal samples, we identified significant gene co-expression pairs. As co-expression networks of drug group and control group constructed, associated function term pairs and pathway pairs reflected by co-expression pattern changes were discovered by integrating functional and pathway information respectively. The results indicated that respiratory electron transport chain, synaptic transmission, mitochondrial electron transport, signal transduction, locomotory behavior, response to amphetamine, negative regulation of cell migration, glucose regulation of insulin secretion, signaling by NGF, diabetes pathways, integration of energy metabolism, dopamine receptors may play an important role in drug addiction. In addition, the results can provide theory support for studies of addiction mechanisms.
Collapse
Affiliation(s)
- Zi-hui Li
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin 150081, China
| | | | | | | | | | | | | | | |
Collapse
|
46
|
Naloxone-precipitated morphine withdrawal evokes phosphorylation of heat shock protein 27 in rat heart through extracellular signal-regulated kinase. J Mol Cell Cardiol 2011; 51:129-39. [DOI: 10.1016/j.yjmcc.2011.04.002] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/31/2011] [Revised: 04/04/2011] [Accepted: 04/05/2011] [Indexed: 02/04/2023]
|
47
|
Fernandez-Espejo E, Rodriguez-Espinosa N. Psychostimulant Drugs and Neuroplasticity. Pharmaceuticals (Basel) 2011. [PMCID: PMC4058673 DOI: 10.3390/ph4070976] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Drugs of abuse induce plastic changes in the brain that seem to underlie addictive phenomena. These plastic changes can be structural (morphological) or synaptic (biochemical), and most of them take place in the mesolimbic and mesostriatal circuits. Several addiction-related changes in brain circuits (hypofrontality, sensitization, tolerance) as well as the outcome of treatment have been visualized in addicts to psychostimulants using neuroimaging techniques. Repeated exposure to psychostimulants induces morphological changes such as increase in the number of dendritic spines, changes in the morphology of dendritic spines, and altered cellular coupling through new gap junctions. Repeated exposure to psychostimulants also induces various synaptic adaptations, many of them related to sensitization and neuroplastic processes, that include up- or down-regulation of D1, D2 and D3 dopamine receptors, changes in subunits of G proteins, increased adenylyl cyclase activity, cyclic AMP and protein kinase A in the nucleus accumbens, increased tyrosine hydroxylase enzyme activity, increased calmodulin and activated CaMKII in the ventral tegmental area, and increased deltaFosB, c-Fos and AP-1 binding proteins. Most of these changes are transient, suggesting that more lasting plastic brain adaptations should take place. In this context, protein synthesis inhibitors block the development of sensitization to cocaine, indicating that rearrangement of neural networks must develop for the long-lasting plasticity required for addiction to occur. Self-administration studies indicate the importance of glutamate neurotransmission in neuroplastic changes underlying transition from use to abuse. Finally, plastic changes in the addicted brain are enhanced and aggravated by neuroinflammation and neurotrophic disbalance after repeated psychostimulants.
Collapse
Affiliation(s)
- Emilio Fernandez-Espejo
- Author to whom correspondence should be addressed; E-Mail: ; Tel.: +34-95-455-6584; Fax: +34-95-455-1769
| | | |
Collapse
|
48
|
Peregud DI, Yakovlev AA, Panchenko LF, Gulyaeva NV. Expression of the mRNA of neurotrophins in brain regions of rats after spontaneous morphine withdrawal. NEUROCHEM J+ 2011. [DOI: 10.1134/s181971241102005x] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
|
49
|
Sensitization to cocaine is inhibited after intra-accumbal GR103691 or rimonabant, but it is enhanced after co-infusion indicating functional interaction between accumbens D(3) and CB1 receptors. Psychopharmacology (Berl) 2011; 214:949-59. [PMID: 21128069 DOI: 10.1007/s00213-010-2104-4] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/26/2010] [Accepted: 11/17/2010] [Indexed: 10/18/2022]
Abstract
RATIONALE Dopamine D(3) receptors and cannabinoid CB(1) receptors are both expressed in the nucleus accumbens, and they have been involved in motor sensitization to cocaine. The objectives were: (1) to study the effects of blockade of these receptors on sensitization to repeated cocaine, by using GR103691, D(3) receptor blocker, and rimonabant, CB(1) receptor ligand, and (2) to discern if both receptors interact by co-infusing them. MATERIALS AND METHODS Cocaine (10 mg/kg) was injected daily for 3 days (induction phase) and later on day 8 (expression phase), and locomotor activity was measured during 2 h after cocaine. GR103691 and rimonabant were bilaterally injected (0.5 μl volume of each infusion) in the nucleus accumbens through cannulae (GR103691, 0, 4.85, and 9.7 μg/μl; rimonabant, 0, 0.5, and 1.5 μg/μl), before cocaine, during either induction or expression phases of sensitization. RESULTS The findings indicated that sensitizing effects of cocaine were abolished after D(3) receptor blocking during both induction and expression phases, as well as rimonabant infusion during the expression (not induction) phase. A functional interaction between both receptors was also observed, because if GR103691 was injected during induction and rimonabant during expression, sensitizing effects of cocaine were observed to be normal or further enhanced. CONCLUSION Dopamine D(3) receptors within the nucleus accumbens are critical for the development and consolidation of sensitization, and cannabinoid CB(1) receptors are critical for the expression of sensitization. Co-blockade of D(3) and CB(1) receptors exert opposite effects to blockade of these receptors separately, revealing the existence of a functional interaction between them.
Collapse
|
50
|
Ghitza UE, Zhai H, Wu P, Airavaara M, Shaham Y, Lu L. Role of BDNF and GDNF in drug reward and relapse: a review. Neurosci Biobehav Rev 2010; 35:157-71. [PMID: 19914287 PMCID: PMC2891859 DOI: 10.1016/j.neubiorev.2009.11.009] [Citation(s) in RCA: 162] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2009] [Revised: 11/09/2009] [Accepted: 11/10/2009] [Indexed: 11/23/2022]
Abstract
Brain-derived neurotrophic factor (BDNF) and glial cell line-derived neurotrophic factor (GDNF) are neurotrophic factors that are critical for the growth, survival, and differentiation of developing neurons. These neurotrophic factors also play important roles in the survival and function of adult neurons, learning and memory, and synaptic plasticity. Since the mid-1990s, investigators have studied the role of BDNF and GDNF in the behavioral effects of abused drugs and in the neuroadaptations induced by repeated exposure to drugs in the mesocorticolimbic dopamine system. Here, we review rodent studies on the role of BDNF and GDNF in drug reward, as assessed in the drug self-administration and the conditioned place preference procedures, and in drug relapse, as assessed in extinction and reinstatement procedures. Our main conclusion is that whether BDNF or GDNF would facilitate or inhibit drug-taking behaviors depends on the drug type, the brain site, the addiction phase (initiation, maintenance, or abstinence/relapse), and the time interval between site-specific BDNF or GDNF injections and the reward- and relapse-related behavioral assessments.
Collapse
Affiliation(s)
- Udi E. Ghitza
- Center for the Clinical Trials Network, NIDA, NIH, Bethesda, MD, USA
| | - Haifeng Zhai
- National Institute on Drug Dependence, Peking University, Beijing, China
| | - Ping Wu
- National Institute on Drug Dependence, Peking University, Beijing, China
| | | | - Yavin Shaham
- Intramural Research Program, NIDA, NIH, Baltimore, MD, USA
| | - Lin Lu
- National Institute on Drug Dependence, Peking University, Beijing, China
| |
Collapse
|