1
|
Kobayashi C, Kitanaka N, Nakai M, Hall FS, Tomita K, Igarashi K, Sato T, Uhl GR, Kitanaka J. Protein phosphatase 2A inhibitors: a possible pharmacotherapy for benzodiazepine dependence. J Pharm Pharmacol 2025; 77:335-340. [PMID: 39546584 DOI: 10.1093/jpp/rgae136] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2024] [Accepted: 10/08/2024] [Indexed: 11/17/2024]
Abstract
OBJECTIVES Benzodiazepines (BZDs) activate the γ-aminobutyric acid (GABA) subtype A (GABAA) receptors, and thus are widely used medicines for the treatment of anxiety and insomnia. For chronic use, tolerance to BZDs is a major problem. Patients with chronic insomnia that develop tolerance to BZDs lose therapeutic effects but also potentially suffer from BZD dependence resulting in BZD withdrawal. The development of such treatments is important for the appropriate use of BZDs. METHODS Research articles regarding investigation of BZD dependence were searched on PubMed, Embase, and Scopus databases using keywords "benzodiazepine", "dependence", "treatment". KEY FINDINGS When BZDs are taken chronically, continuous GABAA binding results in up-regulation of α-amino-3-hydroxy-5-methyl-4-lisoxazolepropionic acid (AMPA) glutamate receptor function and release of brain-derived neurotrophic factor (BDNF). Released BDNF binds to its specific receptor tropomyosin-related kinase receptor B (TrkB). Enhanced BDNF-TrkB signaling activates protein phosphatase 2A (PP2A). Activated PP2A dephosphorylates GABAA receptors, resulting in the downregulation of the GABAA receptor function. Reduced GABAA receptor function augments long-term potentiation (LTP), AMPA-mediated glutamatergic neuroplasticity, by reducing LTP inhibition by GABAA receptor function. Augmented LTP enhances extreme anxiety, which leads to BZD dependence. CONCLUSION Therefore, iInhibiting dephosphorylation of the GABAA receptor by PP2A, PP2A inhibitors could reduce LTP and anxiety, restoring BZD effectiveness and resulting in possible therapeutic effects for BZD dependence.
Collapse
Affiliation(s)
- Chisa Kobayashi
- Laboratory of Drug Addiction and Experimental Therapeutics, Department of Pharmacy, School of Pharmacy, Hyogo Medical University, Kobe 650-8530, Japan
| | - Nobue Kitanaka
- Laboratory of Drug Addiction and Experimental Therapeutics, Department of Pharmacy, School of Pharmacy, Hyogo Medical University, Kobe 650-8530, Japan
- Department of Pharmacology, School of Medicine, Hyogo Medical University, Nishinomiya, Hyogo 663-8501, Japan
| | - Masanori Nakai
- Laboratory of Drug Addiction and Experimental Therapeutics, Department of Pharmacy, School of Pharmacy, Hyogo Medical University, Kobe 650-8530, Japan
| | - F Scott Hall
- Department of Pharmacology and Experimental Therapeutics, College of Pharmacy and Pharmaceutical Sciences, University of Toledo, Toledo, OH 43614, United States
| | - Kazuo Tomita
- Department of Applied Pharmacology, Kagoshima University Graduate School of Medical and Dental Sciences, Kagoshima 890-8544, Japan
| | - Kento Igarashi
- Department of Applied Pharmacology, Kagoshima University Graduate School of Medical and Dental Sciences, Kagoshima 890-8544, Japan
| | - Tomoaki Sato
- Department of Applied Pharmacology, Kagoshima University Graduate School of Medical and Dental Sciences, Kagoshima 890-8544, Japan
| | - George R Uhl
- Neurology, VA Maryland Healthcare System, Baltimore, Maryland 21201, United States
- Departments of Neurology and Pharmacology, University of Maryland School of Medicine, Baltimore, Maryland 21201, United States
| | - Junichi Kitanaka
- Laboratory of Drug Addiction and Experimental Therapeutics, Department of Pharmacy, School of Pharmacy, Hyogo Medical University, Kobe 650-8530, Japan
| |
Collapse
|
2
|
Shi W, Li M, Zhang T, Yang C, Zhao D, Bai J. GABA system in the prefrontal cortex involved in psychostimulant addiction. Cereb Cortex 2024; 34:bhae319. [PMID: 39098820 DOI: 10.1093/cercor/bhae319] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2024] [Revised: 07/08/2024] [Accepted: 07/17/2024] [Indexed: 08/06/2024] Open
Abstract
Drug addiction is a chronic and relapse brain disorder. Psychostimulants such as cocaine and amphetamine are highly addictive drugs. Abuse drugs target various brain areas in the nervous system. Recent studies have shown that the prefrontal cortex (PFC) plays a key role in regulating addictive behaviors. The PFC is made up of excitatory glutamatergic cells and gamma-aminobutyric acid (GABAergic) interneurons. Recently, studies showed that GABA level was related with psychostimulant addiction. In this review, we will introduce the role and mechanism of GABA and γ-aminobutyric acid receptors (GABARs) of the PFC in regulating drug addiction, especially in psychostimulant addiction.
Collapse
Affiliation(s)
- Wenjing Shi
- Faculty of Life Science and Technology, Kunming University of Science and Technology, No. 727 Jingming South Road, Kunming 650500, Yunnan, China
- Medical School, Kunming University of Science and Technology, No. 727 Jingming South Road, Kunming 650500, Yunnan, China
| | - Minyu Li
- Medical School, Kunming University of Science and Technology, No. 727 Jingming South Road, Kunming 650500, Yunnan, China
| | - Ting Zhang
- Medical School, Kunming University of Science and Technology, No. 727 Jingming South Road, Kunming 650500, Yunnan, China
| | - Chunlong Yang
- Medical School, Kunming University of Science and Technology, No. 727 Jingming South Road, Kunming 650500, Yunnan, China
| | - Dongdong Zhao
- Faculty of Life Science and Technology, Kunming University of Science and Technology, No. 727 Jingming South Road, Kunming 650500, Yunnan, China
- Medical School, Kunming University of Science and Technology, No. 727 Jingming South Road, Kunming 650500, Yunnan, China
| | - Jie Bai
- Medical School, Kunming University of Science and Technology, No. 727 Jingming South Road, Kunming 650500, Yunnan, China
| |
Collapse
|
3
|
Song H, Lu X, Du D, Peng Y, Pan W, Xu X, Fan Y, Yang X, Ge F, Guan X. Gegen-Qinlian decoction-A traditional Chinese medicine formula-Alleviates methamphetamine withdrawal induced anxiety by targeting GABAergic interneuron-pyramidal neuron pathway in mPFC. Addict Biol 2023; 28:e13314. [PMID: 37644891 DOI: 10.1111/adb.13314] [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: 02/24/2023] [Revised: 06/27/2023] [Accepted: 06/30/2023] [Indexed: 08/31/2023]
Abstract
Methamphetamine (Meth) withdrawal elicits anxiety, which is a public health concern with limited therapeutic options. Previous studies implied a strong correlation between mPFC and Meth withdrawal. Here, we examined the role of Gegen-Qinlian decoction (GQD) in Meth withdrawal anxiety and explored potential therapeutic targets in mPFC. We found that intra-gastric administration of GQD during the withdrawal period efficiently alleviated anxiety-like behaviours in Meth-withdrawn mice. Further, GQD could restore Meth withdrawal-triggered pathway of GABAergic interneurons (GABA IN)-pyramidal neurons (PN) in the mPFC of Meth-withdrawn mice, especially the prelimbic cortex (PrL) sub-region and PV-positive GABA IN. While, GQD had no obvious effects on the glial cells in the mPFC of Meth-withdrawn mice. By transcriptomic analysis and validation of several gene candidates, we found that genes in the MAPK signalling pathway, especially those related to heat shock proteins, including Hspa1a, Hspa1b and Hspb1, might be GQD-targeting genes in mPFC to treat Meth withdrawal anxiety, as indicated that these genes were up-regulated by Meth withdrawal but rescued by GQD in mPFC. Collectively, our findings identified for the first time that GQD could efficiently alleviate Meth withdrawal anxiety, partially through regulating the local GABA IN-PN pathway and transcriptomic profile of mPFC. The present study confirms that TCM, such as GQD, will be a desirable therapeutic approach in the treatment of drug addiction and related emotional deficits.
Collapse
Affiliation(s)
- Hongxiu Song
- Department of Human Anatomy and Histoembryology, Nanjing University of Chinese Medicine, Nanjing, China
| | - Xue Lu
- Department of Human Anatomy and Histoembryology, Nanjing University of Chinese Medicine, Nanjing, China
| | - Demin Du
- Department of Human Anatomy and Histoembryology, Nanjing University of Chinese Medicine, Nanjing, China
| | - Yaqin Peng
- Department of Human Anatomy and Histoembryology, Nanjing University of Chinese Medicine, Nanjing, China
| | - Weichao Pan
- Department of Human Anatomy and Histoembryology, Nanjing University of Chinese Medicine, Nanjing, China
| | - Xing Xu
- Department of Human Anatomy and Histoembryology, Nanjing University of Chinese Medicine, Nanjing, China
| | - Yu Fan
- Department of Human Anatomy and Histoembryology, Nanjing University of Chinese Medicine, Nanjing, China
| | - Xin Yang
- Department of Human Anatomy and Histoembryology, Nanjing University of Chinese Medicine, Nanjing, China
| | - Feifei Ge
- Department of Human Anatomy and Histoembryology, Nanjing University of Chinese Medicine, Nanjing, China
| | - Xiaowei Guan
- Department of Human Anatomy and Histoembryology, Nanjing University of Chinese Medicine, Nanjing, China
| |
Collapse
|
4
|
Ma J, Xiong F, Li Z, Dong G, Sun X, Yin W, Cai H. The effect of chronic alcohol exposure on spatial memory and BDNF-TrkB- PLCγ1 signaling in the hippocampus of male and female mice. Heliyon 2023; 9:e16660. [PMID: 37303582 PMCID: PMC10248118 DOI: 10.1016/j.heliyon.2023.e16660] [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: 11/16/2022] [Revised: 05/22/2023] [Accepted: 05/23/2023] [Indexed: 06/13/2023] Open
Abstract
Alcohol is a commonly used drug worldwide, and abuse of alcohol has become a serious public health problem. Alcohol consumption over time can cause cognitive deficits and memory impairment, which is thought to be associated with changes in the hippocampus. Given previously known effects of brain-derived neurotrophic factor (BDNF) in regulating synaptic plasticity and learning and memory, we investigated the effect of chronic alcohol consumption on spatial memory impairment in both sexes and changes in BDNF signaling in the hippocampus. After 4 weeks of intermittent access to 20% alcohol, memory impairment in both male and female mice was evaluated using the Morris water maze and the expression of BDNF, TrkB, phosphorylation of PLCγ1 (p-PLCγ1) and PLCγ1 in the hippocampus was examined using Western blot. As expected, females spent longer escape latencies during the training phase, and both sexes spent shorter time in the target quadrant. Furthermore, after 4 weeks 20% alcohol exposure, we found significantly decreased expression levels of BDNF in the hippocampus of female mice but increased levels in male mice. TrkB and PLCγ1 expression showed no significant change in the hippocampus of both sexes. These findings suggest that chronic alcohol exposure may induce spatial memory impairment in both sexes and opposite changes in expression of BDNF and p-PLCγ1 in the hippocampus of males and females.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Hongxing Cai
- Corresponding author. .Department of Forensic Science, Xuzhou Medical University, 209 Tongshan Road, Xuzhou, Jiangsu, China.
| |
Collapse
|
5
|
Ehinger Y, Soneja D, Phamluong K, Salvi A, Ron D. Identification of Novel BDNF-Specific Corticostriatal Circuitries. eNeuro 2023; 10:ENEURO.0238-21.2023. [PMID: 37156610 PMCID: PMC10198608 DOI: 10.1523/eneuro.0238-21.2023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2021] [Revised: 03/23/2023] [Accepted: 04/03/2023] [Indexed: 05/10/2023] Open
Abstract
Brain-derived neurotrophic factor (BDNF) is released from axon terminals originating in the cerebral cortex onto striatal neurons. Here, we characterized BDNF neurons in the corticostriatal circuitry. First, we used BDNF-Cre and Ribotag transgenic mouse lines to label BDNF-positive neurons in the cortex and detected BDNF expression in all the subregions of the prefrontal cortex (PFC). Next, we used a retrograde viral tracing strategy, in combination with BDNF-Cre knock-in mice, to map the cortical outputs of BDNF neurons in the dorsomedial and dorsolateral striatum (DMS and DLS, respectively). We found that BDNF-expressing neurons located in the medial prefrontal cortex (mPFC) project mainly to the DMS, and those located in the primary and secondary motor cortices (M1 and M2, respectively) and agranular insular cortex (AI) project mainly to the DLS. In contrast, BDNF-expressing orbitofrontal cortical (OFC) neurons differentially target the dorsal striatum (DS) depending on their mediolateral and rostrocaudal location. Specifically, the DMS is mainly innervated by the medial and ventral part of the orbitofrontal cortex (MO and VO, respectively), whereas the DLS receives projections specifically from the lateral part of the OFC (LO). Together, our study uncovers previously unknown BDNF corticostriatal circuitries. These findings could have important implications for the role of BDNF signaling in corticostriatal pathways.
Collapse
Affiliation(s)
- Yann Ehinger
- Department of Neurology, University of California, San Francisco, 94143-0663 CA
| | - Drishti Soneja
- Department of Neurology, University of California, San Francisco, 94143-0663 CA
| | - Khanhky Phamluong
- Department of Neurology, University of California, San Francisco, 94143-0663 CA
| | - Alexandra Salvi
- Department of Neurology, University of California, San Francisco, 94143-0663 CA
| | - Dorit Ron
- Department of Neurology, University of California, San Francisco, 94143-0663 CA
| |
Collapse
|
6
|
You H, Lu B. Diverse Functions of Multiple Bdnf Transcripts Driven by Distinct Bdnf Promoters. Biomolecules 2023; 13:655. [PMID: 37189402 PMCID: PMC10135494 DOI: 10.3390/biom13040655] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2023] [Revised: 04/01/2023] [Accepted: 04/04/2023] [Indexed: 05/17/2023] Open
Abstract
The gene encoding brain-derived neurotrophic factor (Bdnf) consists of nine non-coding exons driven by unique promoters, leading to the expression of nine Bdnf transcripts that play different roles in various brain regions and physiological stages. In this manuscript, we present a comprehensive overview of the molecular regulation and structural characteristics of the multiple Bdnf promoters, along with a summary of the current knowledge on the cellular and physiological functions of the distinct Bdnf transcripts produced by these promoters. Specifically, we summarized the role of Bdnf transcripts in psychiatric disorders, including schizophrenia and anxiety, as well as the cognitive functions associated with specific Bdnf promoters. Moreover, we examine the involvement of different Bdnf promoters in various aspects of metabolism. Finally, we propose future research directions that will enhance our understanding of the complex functions of Bdnf and its diverse promoters.
Collapse
Affiliation(s)
- He You
- School of Pharmaceutical Sciences, IDG/McGovern Institute for Brain Research, Tsinghua University, Beijing 100084, China;
- School of Life Sciences, Tsinghua University, Beijing 100084, China
| | - Bai Lu
- School of Pharmaceutical Sciences, IDG/McGovern Institute for Brain Research, Tsinghua University, Beijing 100084, China;
- Stellenbosch Institute for Advanced Study (STIAS), Wallenberg Centre, 10 Marais Street, Stellenbosch 7600, South Africa
| |
Collapse
|
7
|
Nikbakhtzadeh M, Ranjbar H, Moradbeygi K, Zahedi E, Bayat M, Soti M, Shabani M. Cross-talk between the HPA axis and addiction-related regions in stressful situations. Heliyon 2023; 9:e15525. [PMID: 37151697 PMCID: PMC10161713 DOI: 10.1016/j.heliyon.2023.e15525] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2022] [Revised: 04/04/2023] [Accepted: 04/12/2023] [Indexed: 05/09/2023] Open
Abstract
Addiction is a worldwide problem that has a negative impact on society by imposing significant costs on health care, public security, and the deactivation of the community economic cycle. Stress is an important risk factor in the development of addiction and relapse vulnerability. Here we review studies that have demonstrated the diverse roles of stress in addiction. Term searches were conducted manually in important reference journals as well as in the Google Scholar and PubMed databases, between 2010 and 2022. In each section of this narrative review, an effort has been made to use pertinent sources. First, we will provide an overview of changes in the Hypothalamus-Pituitary-Adrenal (HPA) axis component following stress, which impact reward-related regions including the ventral tegmental area (VTA) and nucleus accumbens (NAc). Then we will focus on internal factors altered by stress and their effects on drug addiction vulnerability. We conclude that alterations in neuro-inflammatory, neurotrophic, and neurotransmitter factors following stress pathways can impact related mechanisms on craving and relapse susceptibility.
Collapse
Affiliation(s)
- Marjan Nikbakhtzadeh
- Department of Physiology, School of Medicine, Tehran University of Medical Science, Tehran, Iran
| | - Hoda Ranjbar
- Neuroscience Research Center of Kerman, Institute of Neuropharmacology, Kerman University of Medical Science, Kerman, Iran
| | | | - Elham Zahedi
- Department of Physiology, School of Medicine, Tehran University of Medical Science, Tehran, Iran
| | - Mahnaz Bayat
- Clinical Neurology Research Centre, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Monavareh Soti
- Neuroscience Research Center of Kerman, Institute of Neuropharmacology, Kerman University of Medical Science, Kerman, Iran
- Corresponding author. Neuroscience Research Center, Neuropharmacology institute, Kerman University of Medical Sciences, Kerman, Postal Code: 76198-13159, Iran.
| | - Mohammad Shabani
- Neuroscience Research Center of Kerman, Institute of Neuropharmacology, Kerman University of Medical Science, Kerman, Iran
- Corresponding author. Neuroscience Research Center, Neuropharmacology institute, Kerman University of Medical Sciences, Kerman, Postal Code: 76198-13159, Iran.
| |
Collapse
|
8
|
Kida H, Kawakami R, Sakai K, Otaku H, Imamura K, Han TZ, Sakimoto Y, Mitsushima D. Motor training promotes both synaptic and intrinsic plasticity of layer V pyramidal neurons in the primary motor cortex. J Physiol 2023; 601:335-353. [PMID: 36515167 DOI: 10.1113/jp283755] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2022] [Accepted: 11/23/2022] [Indexed: 12/15/2022] Open
Abstract
Layer V neurons in the primary motor cortex (M1) are important for motor skill learning. Since pretreatment of either CNQX or APV in rat M1 layer V impaired rotor rod learning, we analysed training-induced synaptic plasticity by whole-cell patch-clamp technique in acute brain slices. Rats trained for 1 day showed a decrease in small inhibitory postsynaptic current (mIPSC) frequency and an increase in the paired-pulse ratio of evoked IPSCs, suggesting a transient decrease in presynaptic GABA release in the early phase. Rats trained for 2 days showed an increase in miniature excitatory postsynaptic current (mEPSC) amplitudes/frequency and elevated AMPA/NMDA ratios, suggesting a long-term strengthening of AMPA receptor-mediated excitatory synapses. Importantly, rotor rod performance in trained rats was correlated with the mean mEPSC amplitude and the frequency obtained from that animal. In current-clamp analysis, 1-day-trained rats transiently decreased the current-induced firing rate, while 2-day-trained rats returned to pre-training levels, suggesting dynamic changes in intrinsic properties. Furthermore, western blot analysis of layer V detected decreased phosphorylation of Ser408-409 in GABAA receptor β3 subunits in 1-day-trained rats, and increased phosphorylation of Ser831 in AMPA receptor GluA1 subunits in 2-day-trained rats. Finally, live-imaging analysis of Thy1-YFP transgenic mice showed that the training rapidly recruited a substantial number of spines for long-term plasticity in M1 layer V neurons. Taken together, these results indicate that motor training induces complex and diverse plasticity in M1 layer V pyramidal neurons. KEY POINTS: Here we examined motor training-induced synaptic and intrinsic plasticity of layer V pyramidal neurons in the primary motor cortex. The training reduced presynaptic GABA release in the early phase, but strengthened AMPA receptor-mediated excitatory synapses in the later phase: acquired motor performance after training correlated with the strength of excitatory synapses rather than inhibitory synapses. As to the intrinsic property, the training transiently decreased the firing rate in the early phase, but returned to pre-training levels in the later phase. Western blot analysis detected decreased phosphorylation of Ser408-409 in GABAA receptor β3 subunits in the acute phase, and increased phosphorylation of Ser831 in AMPA receptor GluA1 subunits in the later phase. Live-imaging analysis of Thy1-YFP transgenic mice showed rapid and long-term spine plasticity in M1 layer V neurons, suggesting training-induced increases in self-entropy per spine.
Collapse
Affiliation(s)
- H Kida
- Department of Physiology, Yamaguchi University Graduate School of Medicine, Yamaguchi, Japan
| | - R Kawakami
- Department of Molecular Medicine for Pathogenesis, Graduate School of Medicine, Ehime University, Ehime, Japan
| | - K Sakai
- Department of Physiology, Yamaguchi University Graduate School of Medicine, Yamaguchi, Japan
| | - H Otaku
- Department of Physiology, Yamaguchi University Graduate School of Medicine, Yamaguchi, Japan
| | - K Imamura
- Department of Physiology, Yamaguchi University Graduate School of Medicine, Yamaguchi, Japan
| | - Thiri-Zin Han
- Department of Physiology, Yamaguchi University Graduate School of Medicine, Yamaguchi, Japan
| | - Y Sakimoto
- Department of Physiology, Yamaguchi University Graduate School of Medicine, Yamaguchi, Japan
| | - Dai Mitsushima
- Department of Physiology, Yamaguchi University Graduate School of Medicine, Yamaguchi, Japan.,The Research Institute for Time Studies, Yamaguchi University, Yamaguchi, Japan
| |
Collapse
|
9
|
Bazzari AH, Bazzari FH. BDNF Therapeutic Mechanisms in Neuropsychiatric Disorders. Int J Mol Sci 2022; 23:ijms23158417. [PMID: 35955546 PMCID: PMC9368938 DOI: 10.3390/ijms23158417] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2022] [Revised: 07/26/2022] [Accepted: 07/28/2022] [Indexed: 11/16/2022] Open
Abstract
Brain-derived neurotrophic factor (BDNF) is the most abundant neurotrophin in the adult brain and functions as both a primary neurotrophic signal and a neuromodulator. It serves essential roles in neuronal development, maintenance, transmission, and plasticity, thereby influencing aging, cognition, and behavior. Accumulating evidence associates reduced central and peripheral BDNF levels with various neuropsychiatric disorders, supporting its potential utilization as a biomarker of central pathologies. Subsequently, extensive research has been conducted to evaluate restoring, or otherwise augmenting, BDNF transmission as a potential therapeutic approach. Promising results were indeed observed for genetic BDNF upregulation or exogenous administration using a multitude of murine models of neurological and psychiatric diseases. However, varying mechanisms have been proposed to underlie the observed therapeutic effects, and many findings indicate the engagement of disease-specific and other non-specific mechanisms. This is because BDNF essentially affects all aspects of neuronal cellular function through tropomyosin receptor kinase B (TrkB) receptor signaling, the disruptions of which vary between brain regions across different pathologies leading to diversified consequences on cognition and behavior. Herein, we review the neurophysiology of BDNF transmission and signaling and classify the converging and diverging molecular mechanisms underlying its therapeutic potentials in neuropsychiatric disorders. These include neuroprotection, synaptic maintenance, immunomodulation, plasticity facilitation, secondary neuromodulation, and preservation of neurovascular unit integrity and cellular viability. Lastly, we discuss several findings suggesting BDNF as a common mediator of the therapeutic actions of centrally acting pharmacological agents used in the treatment of neurological and psychiatric illness.
Collapse
Affiliation(s)
- Amjad H. Bazzari
- Faculty of Medicine, Arab American University, 13 Zababdeh, Jenin 240, Palestine
- Correspondence:
| | - Firas H. Bazzari
- Faculty of Pharmacy, Arab American University, 13 Zababdeh, Jenin 240, Palestine;
| |
Collapse
|
10
|
Pan HQ, Liu XX, He Y, Zhou J, Liao CZ, You WJ, Jiang SY, Qin X, Chen WB, Fei EK, Zhang WH, Pan BX. Prefrontal GABA A(δ)R Promotes Fear Extinction through Enabling the Plastic Regulation of Neuronal Intrinsic Excitability. J Neurosci 2022; 42:5755-5770. [PMID: 35705488 PMCID: PMC9302468 DOI: 10.1523/jneurosci.0689-22.2022] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2022] [Revised: 06/02/2022] [Accepted: 06/06/2022] [Indexed: 01/22/2023] Open
Abstract
Extinguishing the previously acquired fear is critical for the adaptation of an organism to the ever-changing environment, a process requiring the engagement of GABAA receptors (GABAARs). GABAARs consist of tens of structurally, pharmacologically, and functionally heterogeneous subtypes. However, the specific roles of these subtypes in fear extinction remain largely unexplored. Here, we observed that in the medial prefrontal cortex (mPFC), a core region for mood regulation, the extrasynaptically situated, δ-subunit-containing GABAARs [GABAA(δ)Rs], had a permissive role in tuning fear extinction in male mice, an effect sharply contrasting to the established but suppressive role by the whole GABAAR family. First, the fear extinction in individual mice was positively correlated with the level of GABAA(δ)R expression and function in their mPFC. Second, knockdown of GABAA(δ)R in mPFC, specifically in its infralimbic (IL) subregion, sufficed to impair the fear extinction in mice. Third, GABAA(δ)R-deficient mice also showed fear extinction deficits, and re-expressing GABAA(δ)Rs in the IL of these mice rescued the impaired extinction. Further mechanistic studies demonstrated that the permissive effect of GABAA(δ)R was associated with its role in enabling the extinction-evoked plastic regulation of neuronal excitability in IL projection neurons. By contrast, GABAA(δ)R had little influence on the extinction-evoked plasticity of glutamatergic transmission in these cells. Altogether, our findings revealed an unconventional and permissive role of extrasynaptic GABAA receptors in fear extinction through a route relying on nonsynaptic plasticity.SIGNIFICANCE STATEMENT The medial prefrontal cortex (mPFC) is one of the kernel brain regions engaged in fear extinction. Previous studies have repetitively shown that the GABAA receptor (GABAAR) family in this region act to suppress fear extinction. However, the roles of specific GABAAR subtypes in mPFC are largely unknown. We observed that the GABAAR-containing δ-subunit [GABAA(δ)R], a subtype of GABAARs exclusively situated in the extrasynaptic membrane and mediating the tonic neuronal inhibition, works oppositely to the whole GABAAR family and promotes (but does not suppress) fear extinction. More interestingly, in striking contrast to the synaptic GABAARs that suppress fear extinction by breaking the extinction-evoked plasticity of glutamatergic transmission, the GABAA(δ)R promotes fear extinction through enabling the plastic regulation of neuronal excitability in the infralimbic subregion of mPFC. Our findings thus reveal an unconventional role of GABAA(δ)R in promoting fear extinction through a route relying on nonsynaptic plasticity.
Collapse
Affiliation(s)
- Han-Qing Pan
- Laboratory of Fear and Anxiety Disorders, Institutes of Life Science, Nanchang University, Nanchang 330031, People's Republic of China
- Department of Biological Science, School of Life Sciences, Nanchang University, Nanchang 330031, People's Republic of China
| | - Xiao-Xuan Liu
- Laboratory of Fear and Anxiety Disorders, Institutes of Life Science, Nanchang University, Nanchang 330031, People's Republic of China
- Neurology Department, The Second Xiangya Hospital, Central South University, Changsha 410011, People's Republic of China
| | - Ye He
- Center for Medical Experiments, Nanchang University, Nanchang 330031, People's Republic of China
| | - Jin Zhou
- Department of Biological Science, School of Life Sciences, Nanchang University, Nanchang 330031, People's Republic of China
| | - Cai-Zhi Liao
- Laboratory of Fear and Anxiety Disorders, Institutes of Life Science, Nanchang University, Nanchang 330031, People's Republic of China
- Department of Biological Science, School of Life Sciences, Nanchang University, Nanchang 330031, People's Republic of China
| | - Wen-Jie You
- Laboratory of Fear and Anxiety Disorders, Institutes of Life Science, Nanchang University, Nanchang 330031, People's Republic of China
- Department of Biological Science, School of Life Sciences, Nanchang University, Nanchang 330031, People's Republic of China
| | - Si-Ying Jiang
- Laboratory of Fear and Anxiety Disorders, Institutes of Life Science, Nanchang University, Nanchang 330031, People's Republic of China
- Department of Biological Science, School of Life Sciences, Nanchang University, Nanchang 330031, People's Republic of China
| | - Xia Qin
- Laboratory of Fear and Anxiety Disorders, Institutes of Life Science, Nanchang University, Nanchang 330031, People's Republic of China
- Department of Biological Science, School of Life Sciences, Nanchang University, Nanchang 330031, People's Republic of China
- Jiangsu Provincial Key Laboratory of Anesthesiology, Xuzhou Medical University, Xuzhou 221004, People's Republic of China
| | - Wen-Bing Chen
- Laboratory of Fear and Anxiety Disorders, Institutes of Life Science, Nanchang University, Nanchang 330031, People's Republic of China
- Department of Biological Science, School of Life Sciences, Nanchang University, Nanchang 330031, People's Republic of China
| | - Er-Kang Fei
- Laboratory of Fear and Anxiety Disorders, Institutes of Life Science, Nanchang University, Nanchang 330031, People's Republic of China
- Department of Biological Science, School of Life Sciences, Nanchang University, Nanchang 330031, People's Republic of China
| | - Wen-Hua Zhang
- Laboratory of Fear and Anxiety Disorders, Institutes of Life Science, Nanchang University, Nanchang 330031, People's Republic of China
- Department of Biological Science, School of Life Sciences, Nanchang University, Nanchang 330031, People's Republic of China
| | - Bing-Xing Pan
- Laboratory of Fear and Anxiety Disorders, Institutes of Life Science, Nanchang University, Nanchang 330031, People's Republic of China
- Department of Biological Science, School of Life Sciences, Nanchang University, Nanchang 330031, People's Republic of China
| |
Collapse
|
11
|
Abstract
Cocaine self-administration disturbs intracellular signaling in multiple reward circuitry neurons that underlie relapse to drug seeking. Cocaine-induced deficits in prelimbic (PL) prefrontal cortex change during abstinence, resulting in different neuroadaptations during early withdrawal from cocaine self-administration than after one or more weeks of abstinence. Infusion of brain-derived neurotrophic factor (BDNF) into the PL cortex immediately following a final session of cocaine self-administration attenuates relapse to cocaine seeking for an extended period. BDNF affects local (PL) and distal subcortical target areas that mediate cocaine-induced neuroadaptations that lead to cocaine seeking. Blocking synaptic activity selectively in the PL projection to the nucleus accumbens during early withdrawal prevents BDNF from decreasing subsequent relapse. In contrast, blocking synaptic activity selectively in the PL projection to the paraventricular thalamic nucleus by itself decreases subsequent relapse and prior intra-PL BDNF infusion prevents the decrease. Infusion of BDNF into other brain structures at different timepoints after cocaine self administration differentially alters cocaine seeking. Thus, the effects of BDNF on drug seeking are different depending on the brain region, the timepoint of intervention, and the specific pathway that is affected.
Collapse
Affiliation(s)
- Jacqueline F McGinty
- Department of Neuroscience, Medical University of South Carolina, 173 Ashley Ave MSC 510, Charleston, SC 29425, USA
| |
Collapse
|
12
|
Addiction-induced plasticity in underlying neural circuits. Neurol Sci 2022; 43:1605-1615. [DOI: 10.1007/s10072-021-05778-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2021] [Accepted: 11/20/2021] [Indexed: 10/19/2022]
|
13
|
Significance of GABA A Receptor for Cognitive Function and Hippocampal Pathology. Int J Mol Sci 2021; 22:ijms222212456. [PMID: 34830337 PMCID: PMC8623595 DOI: 10.3390/ijms222212456] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2021] [Revised: 11/08/2021] [Accepted: 11/08/2021] [Indexed: 02/05/2023] Open
Abstract
The hippocampus is a primary area for contextual memory, known to process spatiotemporal information within a specific episode. Long-term strengthening of glutamatergic transmission is a mechanism of contextual learning in the dorsal cornu ammonis 1 (CA1) area of the hippocampus. CA1-specific immobilization or blockade of α-amino-3-hydroxyl-5-methyl-4-isoxazole-propionate (AMPA) receptor delivery can impair learning performance, indicating a causal relationship between learning and receptor delivery into the synapse. Moreover, contextual learning also strengthens GABAA (gamma-aminobutyric acid) receptor-mediated inhibitory synapses onto CA1 neurons. Recently we revealed that strengthening of GABAA receptor-mediated inhibitory synapses preceded excitatory synaptic plasticity after contextual learning, resulting in a reduced synaptic excitatory/inhibitory (E/I) input balance that returned to pretraining levels within 10 min. The faster plasticity at inhibitory synapses may allow encoding a contextual memory and prevent cognitive dysfunction in various hippocampal pathologies. In this review, we focus on the dynamic changes of GABAA receptor mediated-synaptic currents after contextual learning and the intracellular mechanism underlying rapid inhibitory synaptic plasticity. In addition, we discuss that several pathologies, such as Alzheimer’s disease, autism spectrum disorders and epilepsy are characterized by alterations in GABAA receptor trafficking, synaptic E/I imbalance and neuronal excitability.
Collapse
|
14
|
Trinoskey-Rice GE, Woon EP, Pitts EG, Gourley SL. Cocaine Elevates Calcium-Dependent Activator Protein for Secretion 2 in the Mouse Orbitofrontal Cortex. Dev Neurosci 2021; 43:376-382. [PMID: 34695821 DOI: 10.1159/000519681] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2021] [Accepted: 09/09/2021] [Indexed: 11/19/2022] Open
Abstract
Calcium-dependent activator protein for secretion 2 (CAPS2; also referred to as CADPS2) is a dense core vesicle-associated protein that promotes the activity-dependent release of neuropeptides including neurotrophins. Addictive drugs appear to prime neurotrophin release in multiple brain regions, but mechanistic factors are still being elucidated. Here, experimenters administered cocaine to adolescent mice at doses that potentiated later cocaine self-administration. Experimenter-administered cocaine elevated the CAPS2 protein content in the orbitofrontal cortex (OFC; but not striatum) multiple weeks after drug exposure. Meanwhile, proteins that are sensitive to brain-derived neurotrophic factor (BDNF) release and binding (phosphorylated protein kinase B and phosphoinositide 3-kinase, and GABAAα1 levels) did not differ between cocaine-exposed and naive mice in the OFC. This pattern is consistent with evidence that CAPS2 primes stimulated release of neurotrophins like BDNF, rather than basal levels. Thus, cocaine administered at behaviorally relevant doses elevates CAPS2 protein content in the OFC, and the effects are detected long after cocaine exposure.
Collapse
Affiliation(s)
- Gracy E Trinoskey-Rice
- Departments of Pediatrics and Psychiatry, Emory University School of Medicine, Atlanta, Georgia, USA.,Yerkes National Primate Research Center, Emory University, Atlanta, Georgia, USA
| | - Ellen P Woon
- Departments of Pediatrics and Psychiatry, Emory University School of Medicine, Atlanta, Georgia, USA.,Yerkes National Primate Research Center, Emory University, Atlanta, Georgia, USA.,Graduate Training Program in Neuroscience, Emory University, Atlanta, Georgia, USA
| | - Elizabeth G Pitts
- Departments of Pediatrics and Psychiatry, Emory University School of Medicine, Atlanta, Georgia, USA.,Yerkes National Primate Research Center, Emory University, Atlanta, Georgia, USA.,Graduate Training Program in Neuroscience, Emory University, Atlanta, Georgia, USA
| | - Shannon L Gourley
- Departments of Pediatrics and Psychiatry, Emory University School of Medicine, Atlanta, Georgia, USA.,Yerkes National Primate Research Center, Emory University, Atlanta, Georgia, USA.,Graduate Training Program in Neuroscience, Emory University, Atlanta, Georgia, USA.,Children's Healthcare of Atlanta, Atlanta, Georgia, USA
| |
Collapse
|
15
|
Cabana-Domínguez J, Martín-García E, Gallego-Roman A, Maldonado R, Fernàndez-Castillo N, Cormand B. Reduced cue-induced reinstatement of cocaine-seeking behavior in Plcb1 +/- mice. Transl Psychiatry 2021; 11:521. [PMID: 34635637 PMCID: PMC8505421 DOI: 10.1038/s41398-021-01396-6] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/19/2020] [Revised: 03/24/2021] [Accepted: 04/20/2021] [Indexed: 11/28/2022] Open
Abstract
Cocaine addiction causes serious health problems, and no effective treatment is available yet. We previously identified a genetic risk variant for cocaine addiction in the PLCB1 gene and found this gene upregulated in postmortem brains of cocaine abusers and in human dopaminergic neuron-like cells after an acute cocaine exposure. Here, we functionally tested the contribution of the PLCB1 gene to cocaine addictive properties using Plcb1+/- mice. First, we performed a general phenotypic characterization and found that Plcb1+/- mice showed normal behavior, although they had increased anxiety and impaired short-term memory. Subsequently, mice were trained for operant conditioning, self-administered cocaine for 10 days, and were tested for cocaine motivation. After extinction, we found a reduction in the cue-induced reinstatement of cocaine-seeking behavior in Plcb1+/- mice. After reinstatement, we identified transcriptomic alterations in the medial prefrontal cortex of Plcb1+/- mice, mostly related to pathways relevant to addiction like the dopaminergic synapse and long-term potentiation. To conclude, we found that heterozygous deletion of the Plcb1 gene decreases cue-induced reinstatement of cocaine-seeking, pointing at PLCB1 as a possible therapeutic target for preventing relapse and treating cocaine addiction.
Collapse
Affiliation(s)
- Judit Cabana-Domínguez
- grid.5841.80000 0004 1937 0247Department de Genètica, Microbiologia i Estadística, Facultat de Biologia, Universitat de Barcelona, Barcelona, Catalonia Spain ,grid.452372.50000 0004 1791 1185Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER), Madrid, Spain ,grid.5841.80000 0004 1937 0247Institut de Biomedicina de la Universitat de Barcelona (IBUB), Barcelona, Catalonia Spain ,grid.411160.30000 0001 0663 8628Institut de Recerca Sant Joan de Déu (IR-SJD), Barcelona, Catalonia Spain
| | - Elena Martín-García
- grid.5612.00000 0001 2172 2676Laboratory of Neuropharmacology-Neurophar, Department of Experimental and Health Sciences, Universitat Pompeu Fabra (UPF), Barcelona, Catalonia Spain ,grid.20522.370000 0004 1767 9005Hospital del Mar Medical Research Institute (IMIM), Barcelona, Catalonia Spain
| | - Ana Gallego-Roman
- grid.5612.00000 0001 2172 2676Laboratory of Neuropharmacology-Neurophar, Department of Experimental and Health Sciences, Universitat Pompeu Fabra (UPF), Barcelona, Catalonia Spain
| | - Rafael Maldonado
- grid.5612.00000 0001 2172 2676Laboratory of Neuropharmacology-Neurophar, Department of Experimental and Health Sciences, Universitat Pompeu Fabra (UPF), Barcelona, Catalonia Spain ,grid.20522.370000 0004 1767 9005Hospital del Mar Medical Research Institute (IMIM), Barcelona, Catalonia Spain
| | - Noèlia Fernàndez-Castillo
- Department de Genètica, Microbiologia i Estadística, Facultat de Biologia, Universitat de Barcelona, Barcelona, Catalonia, Spain. .,Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER), Madrid, Spain. .,Institut de Biomedicina de la Universitat de Barcelona (IBUB), Barcelona, Catalonia, Spain. .,Institut de Recerca Sant Joan de Déu (IR-SJD), Barcelona, Catalonia, Spain.
| | - Bru Cormand
- Department de Genètica, Microbiologia i Estadística, Facultat de Biologia, Universitat de Barcelona, Barcelona, Catalonia, Spain. .,Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER), Madrid, Spain. .,Institut de Biomedicina de la Universitat de Barcelona (IBUB), Barcelona, Catalonia, Spain. .,Institut de Recerca Sant Joan de Déu (IR-SJD), Barcelona, Catalonia, Spain.
| |
Collapse
|
16
|
Caffino L, Mottarlini F, Zita G, Gawliński D, Gawlińska K, Wydra K, Przegaliński E, Fumagalli F. The effects of cocaine exposure in adolescence: Behavioural effects and neuroplastic mechanisms in experimental models. Br J Pharmacol 2021; 179:4233-4253. [PMID: 33963539 PMCID: PMC9545182 DOI: 10.1111/bph.15523] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2021] [Revised: 04/15/2021] [Accepted: 05/04/2021] [Indexed: 01/23/2023] Open
Abstract
Drug addiction is a devastating disorder with a huge economic and social burden for modern society. Although an individual may slip into drug abuse throughout his/her life, adolescents are at higher risk, but, so far, only a few studies have attempted to elucidate the underlying cellular and molecular bases of such vulnerability. Indeed, preclinical evidence indicates that psychostimulants and adolescence interact and contribute to promoting a dysfunctional brain. In this review, we have focused our attention primarily on changes in neuroplasticity brought about by cocaine, taking into account that there is much less evidence from exposure to cocaine in adolescence, compared with that from adults. This review clearly shows that exposure to cocaine during adolescence, acute or chronic, as well as contingent or non‐contingent, confers a vulnerable endophenotype, primarily, by causing changes in neuroplasticity. Given the close relationship between drug abuse and psychiatric disorders, we also discuss the translational implications providing an interpretative framework for clinical studies involving addictive as well as affective or psychotic behaviours.
Collapse
Affiliation(s)
- Lucia Caffino
- Department of Pharmacological and Biomolecular Sciences, Università degli Studi di Milano, Milan, Italy
| | - Francesca Mottarlini
- Department of Pharmacological and Biomolecular Sciences, Università degli Studi di Milano, Milan, Italy
| | - Gianmaria Zita
- Dipartimento di Salute Mentale e Dipendenze, ASST Fatebenefratelli-Sacco, Milan, Italy
| | - Dawid Gawliński
- Department of Drug Addiction Pharmacology, Maj Institute of Pharmacology Polish Academy of Sciences, Kraków, Poland
| | - Kinga Gawlińska
- Department of Drug Addiction Pharmacology, Maj Institute of Pharmacology Polish Academy of Sciences, Kraków, Poland
| | - Karolina Wydra
- Department of Drug Addiction Pharmacology, Maj Institute of Pharmacology Polish Academy of Sciences, Kraków, Poland
| | - Edmund Przegaliński
- Department of Drug Addiction Pharmacology, Maj Institute of Pharmacology Polish Academy of Sciences, Kraków, Poland
| | - Fabio Fumagalli
- Department of Pharmacological and Biomolecular Sciences, Università degli Studi di Milano, Milan, Italy
| |
Collapse
|
17
|
Chan MMY, Yau SSY, Han YMY. The neurobiology of prefrontal transcranial direct current stimulation (tDCS) in promoting brain plasticity: A systematic review and meta-analyses of human and rodent studies. Neurosci Biobehav Rev 2021; 125:392-416. [PMID: 33662444 DOI: 10.1016/j.neubiorev.2021.02.035] [Citation(s) in RCA: 58] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2020] [Revised: 01/05/2021] [Accepted: 02/23/2021] [Indexed: 12/13/2022]
Abstract
The neurobiological mechanisms underlying prefrontal transcranial direct current stimulation (tDCS) remain elusive. Randomized, sham-controlled trials in humans and rodents applying in vivo prefrontal tDCS were included to explore whether prefrontal tDCS modulates resting-state and event-related functional connectivity, neural oscillation and synaptic plasticity. Fifty studies were included in the systematic review and 32 in the meta-analyses. Neuroimaging meta-analysis indicated anodal prefrontal tDCS significantly enhanced bilateral median cingulate activity [familywise error (FWE)-corrected p < .005]; meta-regression revealed a positive relationship between changes in median cingulate activity after tDCS and current density (FWE-corrected p < .005) as well as electric current strength (FWE-corrected p < .05). Meta-analyses of electroencephalography and magnetoencephalography data revealed nonsignificant changes (ps > .1) in both resting-state and event-related oscillatory power across all frequency bands. Applying anodal tDCS over the rodent hippocampus/prefrontal cortex enhanced long-term potentiation and brain-derived neurotrophic factor expression in the stimulated brain regions (ps <.005). Evidence supporting prefrontal tDCS administration is preliminary; more methodologically consistent studies evaluating its effects on cognitive function that include brain activity measurements are needed.
Collapse
Affiliation(s)
- Melody M Y Chan
- Department of Rehabilitation Sciences, The Hong Kong Polytechnic University, Hong Kong SAR, China
| | - Sonata S Y Yau
- Department of Rehabilitation Sciences, The Hong Kong Polytechnic University, Hong Kong SAR, China
| | - Yvonne M Y Han
- Department of Rehabilitation Sciences, The Hong Kong Polytechnic University, Hong Kong SAR, China.
| |
Collapse
|
18
|
Impact of Acute and Persistent Excitation of Prelimbic Pyramidal Neurons on Motor Activity and Trace Fear Learning. J Neurosci 2021; 41:960-971. [PMID: 33402420 DOI: 10.1523/jneurosci.2606-20.2020] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2020] [Revised: 12/11/2020] [Accepted: 12/16/2020] [Indexed: 11/21/2022] Open
Abstract
Drug-induced neuroadaptations in the mPFC have been implicated in addictive behaviors. Repeated cocaine exposure has been shown to increase pyramidal neuron excitability in the prelimbic (PL) region of the mouse mPFC, an adaptation attributable to a suppression of G protein-gated inwardly rectifying K+ (GIRK) channel activity. After establishing that this neuroadaptation is not seen in adjacent GABA neurons, we used viral GIRK channel ablation and complementary chemogenetic approaches to selectively enhance PL pyramidal neuron excitability in adult mice, to evaluate the impact of this form of plasticity on PL-dependent behaviors. GIRK channel ablation decreased somatodendritic GABAB receptor-dependent signaling and rheobase in PL pyramidal neurons. This manipulation also enhanced the motor-stimulatory effect of cocaine but did not impact baseline activity or trace fear learning. In contrast, selective chemogenetic excitation of PL pyramidal neurons, or chemogenetic inhibition of PL GABA neurons, increased baseline and cocaine-induced activity and disrupted trace fear learning. These effects were mirrored in male mice by selective excitation of PL pyramidal neurons projecting to the VTA, but not NAc or BLA. Collectively, these data show that manipulations enhancing the excitability of PL pyramidal neurons, and specifically those projecting to the VTA, recapitulate behavioral hallmarks of repeated cocaine exposure in mice.SIGNIFICANCE STATEMENT Prolonged exposure to drugs of abuse triggers neuroadaptations that promote core features of addiction. Understanding these neuroadaptations and their implications may suggest interventions capable of preventing or treating addiction. While previous work showed that repeated cocaine exposure increased the excitability of pyramidal neurons in the prelimbic cortex (PL), the behavioral implications of this neuroadaptation remained unclear. Here, we used neuron-specific manipulations to evaluate the impact of increased PL pyramidal neuron excitability on PL-dependent behaviors. Acute or persistent excitation of PL pyramidal neurons potentiated cocaine-induced motor activity and disrupted trace fear conditioning, effects replicated by selective excitation of the PL projection to the VTA. Our work suggests that hyperexcitability of this projection drives key behavioral hallmarks of addiction.
Collapse
|
19
|
Ruan H, Yao WD. Loss of mGluR1-LTD following cocaine exposure accumulates Ca 2+-permeable AMPA receptors and facilitates synaptic potentiation in the prefrontal cortex. J Neurogenet 2021; 35:358-369. [PMID: 34092163 PMCID: PMC9255266 DOI: 10.1080/01677063.2021.1931180] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Addiction results from drug-elicited alterations of synaptic plasticity mechanisms in dopaminergic reward circuits. Impaired metabotropic glutamate receptor (mGluR)-dependent long-term depression (LTD) and accumulation of synaptic Ca2+-permeable AMPA receptors (CP-AMPARs) following drug exposure have emerged as important mechanisms underlying drug craving and relapse. Here we show that repeated cocaine exposure in vivo causes transient but complete loss of mGluR1- and mTOR (mammalian target of rapamycin)-dependent LTD in layer 5 pyramidal neurons of mouse prefrontal cortex (PFC), a major dopaminergic target in the reward circuitry. This mGluR1-LTD impairment was prevented by in vivo administration of an mGluR1 positive allosteric modulator (PAM) and rescued by inhibition of dopamine D1 receptors, suggesting that impaired mGluR1 tone and excessive D1 signaling underlie this LTD deficit. Concurrently, CP-AMPARs were generated, indicated by increased sensitivity to the CP-AMPAR inhibitor Naspm and rectification of synaptic AMPAR currents, which were reversed by PAM in cocaine-exposed mice. Finally, these CP-AMPARs mediate an abnormal spike-timing-dependent long-term potentiation enabled by cocaine exposure. Our findings reveal a mechanism by which cocaine impairs LTD and remodels synaptic AMPARs to influence Hebbian plasticity in the PFC. Failure to undergo LTD may prevent the reversal of drug-potentiated brain circuits to their baseline states, perpetuating addictive behaviors.HIGHLIGHTSA mGluR1- and mTOR-dependent LTD is present in the mouse medial prefrontal cortex.Repeated cocaine exposure in vivo temporally but completely abolishes prefrontal mGluR1-LTD.Impaired mGluR1 function and excessive D1 DA signaling likely underlie cocaine impairment of mGluR1-LTD.Ca2+-permeable AMPA receptors are generated by cocaine exposure, likely resulting from mGluR1-LTD impairment, and contribute to a cocaine-induced extended spike timing LTP.
Collapse
|
20
|
Wan L, Chen L, Yu J, Wang G, Wu Z, Qian B, Liu X, Wang Y. Coordinated downregulation of KCC2 and GABA A receptor contributes to inhibitory dysfunction during seizure induction. Biochem Biophys Res Commun 2020; 532:489-495. [PMID: 32892950 DOI: 10.1016/j.bbrc.2020.08.082] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2020] [Accepted: 08/22/2020] [Indexed: 01/25/2023]
Abstract
The GABAA receptor (GABAAR) is the main inhibitory receptor in the adult mammalian brain. GABAAR function is dependent on its expression, distribution, and the chloride (Cl-) transmembrane gradient, which is determined by the potassium-chloride cotransporter 2 (KCC2) in the adult brain. KCC2 and GABAAR are downregulated in an activity-dependent manner during seizure induction. Functionally, KCC2 and GABAAR are closely related membrane proteins which modulate GABAergic inhibition. However, it remains unclear how their downregulation during seizure induction is coordinated. This study aimed to assess this interaction. Our results revealed that KCC2 and GABAAR were simultaneously downregulated in both in vivo and in vitro seizure models induced by the convulsant cyclothazide (CTZ), which was at least partly due to structural coupling in hippocampal neuronal membranes. Immunohistochemistry revealed colocalization of gephyrin with KCC2 and co-immunoprecipitation exhibited a direct coupling between GABAAR α1-subunit and KCC2 protein in hippocampal cell membranes. KCC2 specific short hairpin RNA (KCC2-shRNA) was employed to specifically reduce the expression of KCC2 in cultured hippocampal neurons. This resulted in a significant reduction in KCC2-independent GABAergic miniature inhibitory post-synaptic current (mIPSC) amplitude in shKCC2-transfected neurons. Further, pre-treatment with furosemide, a KCC2 inhibitor, during CTZ stimulation followed by washout significantly prevented convulsant stimulation-induced membrane KCC2 downregulation and significantly attenuated GABAAR downregulation concomitant with recovery of suppressed KCC2-independent GABAergic mIPSC amplitude. Our results suggest that the coordinated downregulation of KCC2 and GABAAR during seizure induction exerts a strong functional impact on GABAAR, highlighting an important regulatory mechanism in epilepsy.
Collapse
Affiliation(s)
- Li Wan
- Rehabilitation Center, Shenzhen Second People's Hospital, The First Affiliated Hospital of Shenzhen University Health Science Center, Shenzhen, 518035, China.
| | - Lulan Chen
- Department of Neurology, Institutes of Brain Science, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institute of Biological Science, Zhongshan Hospital, Fudan University, Shanghai, 200032, China
| | - Jiangning Yu
- Department of Neurology, Institutes of Brain Science, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institute of Biological Science, Zhongshan Hospital, Fudan University, Shanghai, 200032, China
| | - Guoxiang Wang
- Department of Neurology, Institutes of Brain Science, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institute of Biological Science, Zhongshan Hospital, Fudan University, Shanghai, 200032, China
| | - Zheng Wu
- Department of Neurology, Institutes of Brain Science, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institute of Biological Science, Zhongshan Hospital, Fudan University, Shanghai, 200032, China
| | - Binbin Qian
- Department of Neurology, Institutes of Brain Science, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institute of Biological Science, Zhongshan Hospital, Fudan University, Shanghai, 200032, China
| | - Xu Liu
- Department of Neurology, Institutes of Brain Science, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institute of Biological Science, Zhongshan Hospital, Fudan University, Shanghai, 200032, China.
| | - Yun Wang
- Department of Neurology, Institutes of Brain Science, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institute of Biological Science, Zhongshan Hospital, Fudan University, Shanghai, 200032, China.
| |
Collapse
|
21
|
Sakimoto Y, Mizuno J, Kida H, Kamiya Y, Ono Y, Mitsushima D. Learning Promotes Subfield-Specific Synaptic Diversity in Hippocampal CA1 Neurons. Cereb Cortex 2020; 29:2183-2195. [PMID: 30796817 PMCID: PMC6459007 DOI: 10.1093/cercor/bhz022] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2018] [Revised: 01/25/2019] [Accepted: 01/28/2019] [Indexed: 12/18/2022] Open
Abstract
The hippocampus is functionally heterogeneous between the dorsal and ventral subfields with left–right asymmetry. To determine the possible location of contextual memory, we performed an inhibitory avoidance task to analyze synaptic plasticity using slice patch-clamp technique. The training bilaterally increased the AMPA/NMDA ratio at dorsal CA3–CA1 synapses, whereas the training did not affect the ratio at ventral CA3–CA1 synapses regardless of the hemisphere. Moreover, sequential recording of miniature excitatory postsynaptic currents and miniature inhibitory postsynaptic currents from the same CA1 neuron clearly showed learning-induced synaptic plasticity. In dorsal CA1 neurons, the training dramatically strengthened both excitatory and inhibitory postsynaptic responses in both hemispheres, whereas the training did not promote the plasticity in either hemisphere in ventral CA1 neurons. Nonstationary fluctuation analysis further revealed that the training bilaterally increased the number of AMPA or GABAA receptor channels at dorsal CA1 synapses, but not at ventral CA1 synapses, suggesting functional heterogeneity of learning-induced receptor mobility. Finally, the performance clearly impaired by the bilateral microinjection of plasticity blockers in dorsal, but not ventral CA1 subfields, suggesting a crucial role for contextual learning. The quantification of synaptic diversity in specified CA1 subfields may help us to diagnose and evaluate cognitive disorders at the information level.
Collapse
Affiliation(s)
- Y Sakimoto
- Department of Physiology, Yamaguchi University Graduate School of Medicine, Yamaguchi, Japan
| | | | - H Kida
- Department of Physiology, Yamaguchi University Graduate School of Medicine, Yamaguchi, Japan
| | - Y Kamiya
- Uonuma Institute of Community Medicine, Niigata University Medical Hospital, Niigata, Japan
| | - Y Ono
- Department of Electronics and Bioinformatics, Meiji University School of Science and Technology, Tokyo, Japan
| | - D Mitsushima
- Department of Physiology, Yamaguchi University Graduate School of Medicine, Yamaguchi, Japan.,Department of Physiology and Neuroscience, Kanagawa Dental University, Kanagawa, Japan.,The Research Institute for Time Studies, Yamaguchi University, Yamaguchi, Japan
| |
Collapse
|
22
|
Li XX, Yang T, Wang N, Zhang LL, Liu X, Xu YM, Gao Q, Zhu XF, Guan YZ. 7,8-Dihydroxyflavone Attenuates Alcohol-Related Behavior in Rat Models of Alcohol Consumption via TrkB in the Ventral Tegmental Area. Front Neurosci 2020; 14:467. [PMID: 32508571 PMCID: PMC7248303 DOI: 10.3389/fnins.2020.00467] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2020] [Accepted: 04/15/2020] [Indexed: 12/22/2022] Open
Abstract
Alcohol use disorder (AUD) is a ubiquitous substance use disorder in the world, of which neural mechanisms remain unclear. Alcohol consumption induces neuro-adaptations in the dopaminergic system originating from the ventral tegmental area (VTA), an important brain region for the reward function in AUD. Endogenous brain-derived neurotrophic factor (BDNF)-TrkB implicated in the development of neuroplasticity, including long-term potentiation of GABAergic synapses (LTP GABA ). We previously found that ethanol blocks LTP GABA in the VTA, either in vivo or in vitro. 7,8-dihydroflavone (7,8-DHF), a BDNF-mimicking small compound, was recently found to penetrate the blood-brain barrier to mimic the biological role of BDNF-TrkB. In this study, we demonstrate that repeated ethanol consumption (including intermittent and continuous ethanol exposure) results in low expression of BDNF in rat VTA. The amount of ethanol intake enhances significantly in rats with intermittent ethanol exposure after 72 h abstinence. Withdrawal signs emerge in rats with continuous ethanol exposure within 3 days after abstinence. Using behavioral tests, intraperitoneal injection of 7,8-DHF can reduce excessive ethanol consumption and preference as well as withdrawal signs in rats with repeated ethanol exposure. Interestingly, microinjection of K252a, an antagonist of TrkB, into the VTA blocks the effects of 7,8-DHF on ethanol-related behaviors. Furthermore, direct microinjection of BDNF into the VTA mimics the effect of 7,8-DHF on ethanol related behaviors. Taken together, 7,8-DHF attenuates alcohol-related behaviors in rats undergoing alcohol consumption via TrkB in the VTA. Our findings suggest BDNF-TrkB in VTA is a part of regulating signals for opposing neural adaptations in AUD, and 7,8-DHF may serve as a potential candidate for treating alcoholism.
Collapse
Affiliation(s)
- Xin-Xin Li
- Department of Physiology and Neurobiology, Mudanjiang Medical University, Mudanjiang, China
| | - Tao Yang
- Department of Physiology and Neurobiology, Mudanjiang Medical University, Mudanjiang, China
| | - Na Wang
- Department of Physiology and Neurobiology, Mudanjiang Medical University, Mudanjiang, China
| | - Li-Li Zhang
- Department of Physiology and Neurobiology, Mudanjiang Medical University, Mudanjiang, China
| | - Xing Liu
- Department of Physiology and Neurobiology, Mudanjiang Medical University, Mudanjiang, China
| | - Yan-Min Xu
- Department of Physiology and Neurobiology, Mudanjiang Medical University, Mudanjiang, China
| | - Qing Gao
- Department of Physiology and Neurobiology, Mudanjiang Medical University, Mudanjiang, China
| | - Xiao-Feng Zhu
- Department of Physiology and Neurobiology, Mudanjiang Medical University, Mudanjiang, China
| | - Yan-Zhong Guan
- Department of Physiology and Neurobiology, Mudanjiang Medical University, Mudanjiang, China
| |
Collapse
|
23
|
Zilkha N, Barnea‐Ygael N, Keidar L, Zangen A. Increased relapse to cocaine-seeking in a genetic model for depression. Addict Biol 2020; 25:e12756. [PMID: 31062481 DOI: 10.1111/adb.12756] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2018] [Revised: 02/21/2019] [Accepted: 03/18/2019] [Indexed: 12/20/2022]
Abstract
The greatest difficulty in treating cocaine addiction is the enormous rates of relapse, which occur despite immense negative consequences. Relapse risks are even greater in addicts with comorbid depression, perhaps because they use drugs to alleviate depressive symptoms. Only a few preclinical studies have examined this comorbidity, mostly exploring depressive-like effects following drug exposure. We examined rats from two different depression-like models: (a) chronic-mild-stress (CMS), which respond to antidepressant medications and (b) depressed-rat-line (DRL), a genetic model of selective breeding, which is less responsive to antidepressant medications. We tested addictive behaviors in a cocaine self-administration procedure, including the "conflict model," where drug-seeking and relapse encounter adverse consequences: an electrified grid in front of the drug-delivering lever. Following behavioral testing, we explored a potential association between behavioral outcomes and protein expression of brain-derived neurotrophic factor (BDNF). We found that DRL rats self-administer more cocaine compared with both CMS and controls, while CMS and control groups did not differ significantly. Notably, DRL but not CMS rats, displayed higher rates of relapse than controls, and expressed higher levels of BDNF in the prelimbic cortex (PLC). Potential translation of these results suggest that medication-resistant depressed patients tend to consume more drugs and are more susceptible to relapse. The increase in PLC BDNF levels is consistent with previous rat models of depression, and concomitantly, with its suggested role in promoting cocaine-seeking.
Collapse
Affiliation(s)
- Noga Zilkha
- Department of NeurobiologyWeizmann institute of Science Rehovot Israel
- Department of Life Sciences and the Zlotowski Center for NeuroscienceBen‐Gurion University Be'er Sheva Israel
| | - Noam Barnea‐Ygael
- Department of Life Sciences and the Zlotowski Center for NeuroscienceBen‐Gurion University Be'er Sheva Israel
| | - Liraz Keidar
- Department of NeurobiologyWeizmann institute of Science Rehovot Israel
| | - Abraham Zangen
- Department of Life Sciences and the Zlotowski Center for NeuroscienceBen‐Gurion University Be'er Sheva Israel
| |
Collapse
|
24
|
Cole RD, Zimmerman M, Matchanova A, Kutlu MG, Gould TJ, Parikh V. Cognitive rigidity and BDNF-mediated frontostriatal glutamate neuroadaptations during spontaneous nicotine withdrawal. Neuropsychopharmacology 2020; 45:866-876. [PMID: 31752015 PMCID: PMC7075915 DOI: 10.1038/s41386-019-0574-6] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/14/2019] [Revised: 10/13/2019] [Accepted: 11/14/2019] [Indexed: 01/12/2023]
Abstract
Cognitive flexibility is the ability to switch strategic responses adaptively in changing environments. Cognitive rigidity imposed by neural circuit adaptations during nicotine abstinence may foster maladaptive nicotine taking in addicts. We systematically examined the effects of spontaneous withdrawal in mice exposed to either nicotine (6.3 or 18 mg/kg/day) or saline for 14 days on cognitive flexibility using an operant strategy set-shifting task. Because frontostriatal circuits are critical for cognitive flexibility and brain-derived neurotrophic factor (BDNF) modulates glutamate plasticity in these circuits, we also explored the effects of nicotine withdrawal on these neurochemical substrates. Mice undergoing nicotine withdrawal required more trials to attain strategy-switching criterion. Error analysis show that animals withdrawn from both nicotine doses committed higher perseverative errors, which correlated with measures of anxiety. However, animals treated with the higher nicotine dose also displayed more strategy maintenance errors that remained independent of negative affect. BDNF mRNA expression increased in the medial prefrontal cortex (mPFC) following nicotine withdrawal. Surprisingly, BDNF protein declined in mPFC but was elevated in dorsal striatum (DS). DS BDNF protein positively correlated with perseverative and maintenance errors, suggesting mPFC-DS overflow of BDNF during withdrawal. BDNF-evoked glutamate release and synapsin phosphorylation was attenuated within DS synapses, but enhanced in the nucleus accumbens, suggesting a dichotomous role of BDNF signaling in striatal regions. Taken together, these data suggest that spontaneous nicotine withdrawal impairs distinct components of cognitive set-shifting and these deficits may be linked to BDNF-mediated alterations in glutamate signaling dynamics in discrete frontostriatal circuits.
Collapse
Affiliation(s)
- Robert D. Cole
- 0000 0001 2248 3398grid.264727.2Department of Psychology and Neuroscience Program, Temple University, Philadelphia, PA 19122 USA
| | - Matty Zimmerman
- 0000 0001 2248 3398grid.264727.2Department of Psychology and Neuroscience Program, Temple University, Philadelphia, PA 19122 USA
| | - Anastasia Matchanova
- 0000 0001 2248 3398grid.264727.2Department of Psychology and Neuroscience Program, Temple University, Philadelphia, PA 19122 USA
| | - Munir Gunes Kutlu
- 0000 0001 2097 4281grid.29857.31Department of Biobehavioral Health, Pennsylvania State University, University Park, PA 16802 USA
| | - Thomas J. Gould
- 0000 0001 2097 4281grid.29857.31Department of Biobehavioral Health, Pennsylvania State University, University Park, PA 16802 USA
| | - Vinay Parikh
- Department of Psychology and Neuroscience Program, Temple University, Philadelphia, PA, 19122, USA.
| |
Collapse
|
25
|
Salehzadeh SA, Mohammadian A, Salimi F. Effect of chronic methamphetamine injection on levels of BDNF mRNA and its CpG island methylation in prefrontal cortex of rats. Asian J Psychiatr 2020; 48:101884. [PMID: 31830601 DOI: 10.1016/j.ajp.2019.101884] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/20/2019] [Revised: 11/21/2019] [Accepted: 11/22/2019] [Indexed: 01/16/2023]
Abstract
Methamphetamine (METH) is a highly addictive psychostimulant. Its abuse causes problems in cognition, attention, or psychiatric conditions such as psychosis. Prefrontal cortex is involved in many aspects of drug addiction and in mental disorders similar to those triggered by METH. Brain-derived neurotrophic factor (BDNF), plays important roles in modulating different aspects of addiction, and is implicated in psychiatric conditions reminiscent of those suffered by METH-abusers. Male Wistar rats were intra-peritoneally injected with METH (8 mg/kg/day) for 14 days while control group received normal saline. After extraction of prefrontal cortices, expression of BDNF IV splice variant and methylation level of its CpG island were evaluated. The relative expression of BDNF IV in METH-treated group was 2.15 fold higher than the control group. Seven out of 29 CpG sites were significantly hypomethylated in the METH group, although none survived Bonferroni adjustment. However, the overall methylation level of the 29 CpGs was significantly lower in METH cases than in controls. We discuss the importance of our results and its implications in detail.
Collapse
Affiliation(s)
- Seyed Ahmad Salehzadeh
- Department of Medical Biotechnology, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran
| | - Ali Mohammadian
- Department of Medical Biotechnology, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran.
| | - Fatemeh Salimi
- Department of Medical Biotechnology, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran
| |
Collapse
|
26
|
Saitow F, Takumi T, Suzuki H. Change in serotonergic modulation contributes to the synaptic imbalance of neuronal circuit at the prefrontal cortex in the 15q11-13 duplication mouse model of autism. Neuropharmacology 2019; 165:107931. [PMID: 31901366 DOI: 10.1016/j.neuropharm.2019.107931] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2019] [Revised: 12/25/2019] [Accepted: 12/30/2019] [Indexed: 10/25/2022]
Abstract
The prefrontal cortex (PFC) has been extensively studied in autism spectrum disorder (ASD) in an attempt to understand the deficits in executive and other higher brain functions related to sociability and emotion. Disruption of the excitatory/inhibitory (E/I) balance of cortical circuits is thought to underlie the pathophysiology of ASD. Recently, we showed that 15q dup mice (a model for ASD with human chromosome 15q11-13 paternal duplication) exhibit disruption of the E/I balance in layer 2/3 pyramidal neurons of the somatosensory cortex due to a decrease in the number of inhibitory synapses. However, whether there is a pathological abnormality in E/I balance in the PFC of 15q dup mice remains unknown. In this study, we found that 15q dup facilitates the activity-induced LTP of glutamate synapses onto layer 5 pyramidal neurons by shifting the E/I balance to an excitatory state, which this was associated with differences in synaptic glutamatergic and GABAergic inputs onto GABAergic fast-spiking interneurons (FSINs). Furthermore, we found that FSIN excitability was well-modulated and regulated by the constitutive activation of 5-HT2 receptors in PFC microcircuits. These results provide new insights into the cellular mechanisms underlying maintenance of optimal E/I balance in the PFC.
Collapse
Affiliation(s)
- Fumihito Saitow
- Department of Pharmacology, Nippon Medical School, Tokyo, 113-8602, Japan.
| | - Toru Takumi
- RIKEN Center for Brain Science, Wako, Saitama, 351-0198, Japan
| | - Hidenori Suzuki
- Department of Pharmacology, Nippon Medical School, Tokyo, 113-8602, Japan
| |
Collapse
|
27
|
Noble F. [Neural circuits and neurotransmitters involved in the effects of psychoactive drugs - State of the art with a focus on cocaine]. Biol Aujourdhui 2019; 213:141-145. [PMID: 31829934 DOI: 10.1051/jbio/2019013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2019] [Indexed: 11/14/2022]
Abstract
Addiction is a chronic disease that has serious consequences, both in terms of public health and economy. Clear characteristics distinguish recreational and controlled use from addiction. Thus, today, addiction includes the notions of compulsive drug use, associated with a loss of control over consumption, leading to craving. When consumption is stopped, withdrawal symptoms may emerge: a negative emotional state, cognitive problems and physical symptoms with some products (alcohol and opiates, for example). Relapse episodes may occur during this withdrawal period, countering the negative effects of withdrawal. Relapse episodes can also be observed after long periods of abstinence. They can be precipitated by re-exposure to the context in which the drugs were taken, or by stress. Regardless of the stage of addiction (e.g., development of the addictive behavior, or relapse) changes in brain function and structure can be observed. Some brain structures are therefore modified, such as the prefrontal cortex, where several neuroadaptations have been identified. Some of these changes are described in this paper.
Collapse
Affiliation(s)
- Florence Noble
- CNRS ERL3649, Inserm U1124, Université Paris Descartes, Pharmacologie et thérapie des addictions, 45 rue des Saints-Pères, 75006 Paris, France
| |
Collapse
|
28
|
Sakimoto Y, Kida H, Mitsushima D. Temporal dynamics of learning-promoted synaptic diversity in CA1 pyramidal neurons. FASEB J 2019; 33:14382-14393. [PMID: 31689120 PMCID: PMC6894079 DOI: 10.1096/fj.201801893rrr] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
Although contextual learning requires plasticity at both excitatory and inhibitory (E/I) synapses in cornu ammonis 1 (CA1) neurons, the temporal dynamics across the neuronal population are poorly understood. Using an inhibitory avoidance task, we analyzed the dynamic changes in learning-induced E/I synaptic plasticity. The training strengthened GABAA receptor–mediated synapses within 1 min, peaked at 10 min, and lasted for over 60 min. The intracellular loop (Ser408−409) of GABAA receptor β3 subunit was also phosphorylated within 1 min of training. As the results of strengthening of α-amino-3-hydroxyl-5-methyl-4-isoxazole-propionate receptor–mediated synapses, CA1 pyramidal neurons exhibited broad diversity of E/I synaptic currents within 5 min. Moreover, presynaptic glutamate release probability at basal dendrites also increased within 5 min. To further quantify the diversified E/I synaptic currents, we calculated self-entropy (bit) for individual neurons. The neurons showed individual levels of the parameter, which rapidly increased within 1 min of training and maintained for over 60 min. These results suggest that learning-induced synaptic plasticity is critical immediately following encoding rather than during the retrieval phase of the learning. Understanding the temporal dynamics along with the quantification of synaptic diversity would be necessary to identify a failure point for learning-promoted plasticity in cognitive disorders.—Sakimoto, Y., Kida, H., Mitsushima, D. Temporal dynamics of learning-promoted synaptic diversity in CA1 pyramidal neurons.
Collapse
Affiliation(s)
- Yuya Sakimoto
- Department of Physiology, Graduate School of Medicine, Yamaguchi University, Ube, Japan
| | - Hiroyuki Kida
- Department of Physiology, Graduate School of Medicine, Yamaguchi University, Ube, Japan
| | - Dai Mitsushima
- Department of Physiology, Graduate School of Medicine, Yamaguchi University, Ube, Japan.,The Research Institute for Time Studies, Yamaguchi University, Yamaguchi, Japan
| |
Collapse
|
29
|
Abstract
The medial prefrontal cortex (mPFC) is a crucial cortical region that integrates information from numerous cortical and subcortical areas and converges updated information to output structures. It plays essential roles in the cognitive process, regulation of emotion, motivation, and sociability. Dysfunction of the mPFC has been found in various neurological and psychiatric disorders, such as depression, anxiety disorders, schizophrenia, autism spectrum disorders, Alzheimer's disease, Parkinson's disease, and addiction. In the present review, we summarize the preclinical and clinical studies to illustrate the role of the mPFC in these neurological diseases.
Collapse
Affiliation(s)
- Pan Xu
- Department of Pharmacology and Physiology, The George Washington University, Washington, District of Columbia
| | - Ai Chen
- Department of Pediatrics, Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan China
| | - Yipeng Li
- Department of Biomedical Engineering, The George Washington University, Washington, District of Columbia
| | - Xuezhi Xing
- Department of Pharmacology and Physiology, The George Washington University, Washington, District of Columbia
| | - Hui Lu
- Department of Pharmacology and Physiology, The George Washington University, Washington, District of Columbia
| |
Collapse
|
30
|
Gangarossa G, Perez S, Dembitskaya Y, Prokin I, Berry H, Venance L. BDNF Controls Bidirectional Endocannabinoid Plasticity at Corticostriatal Synapses. Cereb Cortex 2019; 30:197-214. [DOI: 10.1093/cercor/bhz081] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2019] [Revised: 03/19/2019] [Accepted: 03/20/2019] [Indexed: 12/12/2022] Open
Abstract
AbstractThe dorsal striatum exhibits bidirectional corticostriatal synaptic plasticity, NMDAR and endocannabinoids (eCB) mediated, necessary for the encoding of procedural learning. Therefore, characterizing factors controlling corticostriatal plasticity is of crucial importance. Brain-derived neurotrophic factor (BDNF) and its receptor, the tropomyosine receptor kinase-B (TrkB), shape striatal functions, and their dysfunction deeply affects basal ganglia. BDNF/TrkB signaling controls NMDAR plasticity in various brain structures including the striatum. However, despite cross-talk between BDNF and eCBs, the role of BDNF in eCB plasticity remains unknown. Here, we show that BDNF/TrkB signaling promotes eCB-plasticity (LTD and LTP) induced by rate-based (low-frequency stimulation) or spike-timing–based (spike-timing–dependent plasticity, STDP) paradigm in striatum. We show that TrkB activation is required for the expression and the scaling of both eCB-LTD and eCB-LTP. Using 2-photon imaging of dendritic spines combined with patch-clamp recordings, we show that TrkB activation prolongs intracellular calcium transients, thus increasing eCB synthesis and release. We provide a mathematical model for the dynamics of the signaling pathways involved in corticostriatal plasticity. Finally, we show that TrkB activation enlarges the domain of expression of eCB-STDP. Our results reveal a novel role for BDNF/TrkB signaling in governing eCB-plasticity expression in striatum and thus the engram of procedural learning.
Collapse
Affiliation(s)
- Giuseppe Gangarossa
- Center for Interdisciplinary Research in Biology, College de France, Centre National de la Recherche Scientifique (CNRS) UMR, Institut National de la Santé et de la Recherche (INSERM), Paris Sciences et Lettres (PSL) Research University, Paris, France
| | - Sylvie Perez
- Center for Interdisciplinary Research in Biology, College de France, Centre National de la Recherche Scientifique (CNRS) UMR, Institut National de la Santé et de la Recherche (INSERM), Paris Sciences et Lettres (PSL) Research University, Paris, France
| | - Yulia Dembitskaya
- Center for Interdisciplinary Research in Biology, College de France, Centre National de la Recherche Scientifique (CNRS) UMR, Institut National de la Santé et de la Recherche (INSERM), Paris Sciences et Lettres (PSL) Research University, Paris, France
| | - Ilya Prokin
- INRIA, Villeurbanne, France
- University of Lyon, LIRIS UMR, Villeurbanne, France
| | - Hugues Berry
- INRIA, Villeurbanne, France
- University of Lyon, LIRIS UMR, Villeurbanne, France
| | - Laurent Venance
- Center for Interdisciplinary Research in Biology, College de France, Centre National de la Recherche Scientifique (CNRS) UMR, Institut National de la Santé et de la Recherche (INSERM), Paris Sciences et Lettres (PSL) Research University, Paris, France
| |
Collapse
|
31
|
Hearing M. Prefrontal-accumbens opioid plasticity: Implications for relapse and dependence. Pharmacol Res 2018; 139:158-165. [PMID: 30465850 DOI: 10.1016/j.phrs.2018.11.012] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/06/2018] [Revised: 11/05/2018] [Accepted: 11/07/2018] [Indexed: 01/12/2023]
Abstract
In addiction, an individual's ability to inhibit drug seeking and drug taking is thought to reflect a pathological strengthening of drug-seeking behaviors or impairments in the capacity to control maladaptive behavior. These processes are not mutually exclusive and reflect drug-induced modifications within prefrontal cortical and nucleus accumbens circuits, however unlike psychostimulants such as cocaine, far less is known about the temporal, anatomical, and cellular dynamics of these changes. We discuss what is known regarding opioid-induced adaptations in intrinsic membrane physiology and pre-/postsynaptic neurotransmission in principle pyramidal and medium spiny neurons in the medial prefrontal cortex and nucleus accumbens from electrophysiological studies and explore how circuit specific adaptations may contribute to unique facets of opioid addiction.
Collapse
Affiliation(s)
- Matthew Hearing
- Department of Biomedical Sciences, Marquette University, Milwaukee, WI, 53233, USA.
| |
Collapse
|
32
|
Singer W, Manthey M, Panford-Walsh R, Matt L, Geisler HS, Passeri E, Baj G, Tongiorgi E, Leal G, Duarte CB, Salazar IL, Eckert P, Rohbock K, Hu J, Strotmann J, Ruth P, Zimmermann U, Rüttiger L, Ott T, Schimmang T, Knipper M. BDNF-Live-Exon-Visualization (BLEV) Allows Differential Detection of BDNF Transcripts in vitro and in vivo. Front Mol Neurosci 2018; 11:325. [PMID: 30319348 PMCID: PMC6170895 DOI: 10.3389/fnmol.2018.00325] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2018] [Accepted: 08/22/2018] [Indexed: 12/16/2022] Open
Abstract
Bdnf exon-IV and exon-VI transcripts are driven by neuronal activity and are involved in pathologies related to sleep, fear or memory disorders. However, how their differential transcription translates activity changes into long-lasting network changes is elusive. Aiming to trace specifically the network controlled by exon-IV and -VI derived BDNF during activity-dependent plasticity changes, we generated a transgenic reporter mouse for B DNF- l ive- e xon- v isualization (BLEV), in which expression of Bdnf exon-IV and -VI can be visualized by co-expression of CFP and YFP. CFP and YFP expression was differentially activated and targeted in cell lines, primary cultures and BLEV reporter mice without interfering with BDNF protein synthesis. CFP and YFP expression, moreover, overlapped with BDNF protein expression in defined hippocampal neuronal, glial and vascular locations in vivo. So far, activity-dependent BDNF cannot be explicitly monitored independent of basal BDNF levels. The BLEV reporter mouse therefore provides a new model, which can be used to test whether stimulus-induced activity-dependent changes in BDNF expression are instrumental for long-lasting plasticity modifications.
Collapse
Affiliation(s)
- Wibke Singer
- Department of Otolaryngology, Tübingen Hearing Research Centre (THRC), Molecular Physiology of Hearing, University of Tübingen, Tübingen, Germany
| | - Marie Manthey
- Department of Otolaryngology, Tübingen Hearing Research Centre (THRC), Molecular Physiology of Hearing, University of Tübingen, Tübingen, Germany
| | - Rama Panford-Walsh
- Department of Otolaryngology, Tübingen Hearing Research Centre (THRC), Molecular Physiology of Hearing, University of Tübingen, Tübingen, Germany
| | - Lucas Matt
- Department of Pharmacology, Institute of Pharmacy, Toxicology and Clinical Pharmacy, University of Tübingen, Tübingen, Germany
| | - Hyun-Soon Geisler
- Department of Otolaryngology, Tübingen Hearing Research Centre (THRC), Molecular Physiology of Hearing, University of Tübingen, Tübingen, Germany
| | - Eleonora Passeri
- Department of Otolaryngology, Tübingen Hearing Research Centre (THRC), Molecular Physiology of Hearing, University of Tübingen, Tübingen, Germany
| | - Gabriele Baj
- B.R.A.I.N. Centre for Neuroscience, Department of Life Sciences, University of Trieste, Trieste, Italy
| | - Enrico Tongiorgi
- B.R.A.I.N. Centre for Neuroscience, Department of Life Sciences, University of Trieste, Trieste, Italy
| | - Graciano Leal
- Centre for Neuroscience and Cell Biology (CNC), Department of Life Sciences, University of Coimbra, Coimbra, Portugal
| | - Carlos B. Duarte
- Centre for Neuroscience and Cell Biology (CNC), Department of Life Sciences, University of Coimbra, Coimbra, Portugal
| | - Ivan L. Salazar
- Centre for Neuroscience and Cell Biology (CNC), Department of Life Sciences, University of Coimbra, Coimbra, Portugal
| | - Philipp Eckert
- Department of Otolaryngology, Tübingen Hearing Research Centre (THRC), Molecular Physiology of Hearing, University of Tübingen, Tübingen, Germany
| | - Karin Rohbock
- Department of Otolaryngology, Tübingen Hearing Research Centre (THRC), Molecular Physiology of Hearing, University of Tübingen, Tübingen, Germany
| | - Jing Hu
- Centre for Integrative Neuroscience (CIN), University of Tübingen, Tübingen, Germany
| | - Jörg Strotmann
- Department of Physiology, Institute of Physiology, University of Hohenheim, Stuttgart, Germany
| | - Peter Ruth
- Department of Pharmacology, Institute of Pharmacy, Toxicology and Clinical Pharmacy, University of Tübingen, Tübingen, Germany
| | - Ulrike Zimmermann
- Department of Otolaryngology, Tübingen Hearing Research Centre (THRC), Molecular Physiology of Hearing, University of Tübingen, Tübingen, Germany
| | - Lukas Rüttiger
- Department of Otolaryngology, Tübingen Hearing Research Centre (THRC), Molecular Physiology of Hearing, University of Tübingen, Tübingen, Germany
| | - Thomas Ott
- Transgenic Facility Tübingen, University of Tübingen, Tübingen, Germany
| | - Thomas Schimmang
- Instituto de Biologíay Genética Molecular, Universidad de Valladolid y Consejo Superior de Investigaciones Científicas, Valladolid, Spain
| | - Marlies Knipper
- Department of Otolaryngology, Tübingen Hearing Research Centre (THRC), Molecular Physiology of Hearing, University of Tübingen, Tübingen, Germany
| |
Collapse
|
33
|
Harris NA, Winder DG. Synaptic Plasticity in the Bed Nucleus of the Stria Terminalis: Underlying Mechanisms and Potential Ramifications for Reinstatement of Drug- and Alcohol-Seeking Behaviors. ACS Chem Neurosci 2018; 9:2173-2187. [PMID: 29851347 PMCID: PMC6146063 DOI: 10.1021/acschemneuro.8b00169] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023] Open
Abstract
The bed nucleus of the stria terminalis (BNST) is a component of the extended amygdala that shows significant changes in activity and plasticity through chronic exposure to drugs and stress. The region is critical for stress- and cue-induced reinstatement of drug-seeking behaviors and is thus a candidate region for the plastic changes that occur in abstinence that prime addicted patients for reinstatement behaviors. Here, we discuss the various forms of long-term potentiation (LTP) and long-term depression (LTD) in the rodent BNST and highlight the way that these changes in excitatory transmission interact with exposure to alcohol and other drugs of abuse, as well as other stressors. In addition, we highlight potential areas for future research in this area, including investigating input- and cell-specific bidirectional changes in activity. As we continue to accrue foundational knowledge in the mechanisms and effects of plasticity in the BNST, molecular targets and treatment strategies that are relevant to reinstatement behaviors will also begin to emerge. Here, we briefly discuss the effects of catecholamine receptor modulators on synaptic plasticity in the BNST due to the role of norepinephrine in LTD and dopamine on the short-term component of LTP as well as the role that signaling at these receptors plays in reinstatement of drug- and alcohol-seeking behaviors. We hope that insights gained on the specific changes in plasticity that occur within the BNST during abstinence from alcohol and other drugs of abuse will provide insight into the biological underpinnings of relapse behavior in human addicts and inform future treatment modalities for addiction that tackle this complex biological problem.
Collapse
Affiliation(s)
- Nicholas A. Harris
- Vanderbilt Center for Addiction Research
- Department of Molecular Physiology & Biophysics
| | - Danny G. Winder
- Vanderbilt Center for Addiction Research
- Department of Molecular Physiology & Biophysics
- Vanderbilt J.F. Kennedy Center for Research on Human Development
- Vanderbilt Brain Institute, Vanderbilt University School of Medicine, Nashville, Tennessee 37232, United States
| |
Collapse
|
34
|
Camarini R, Marianno P, Rae M. Social Factors in Ethanol Sensitization. INTERNATIONAL REVIEW OF NEUROBIOLOGY 2018; 140:53-80. [PMID: 30193709 DOI: 10.1016/bs.irn.2018.07.003] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Behavioral sensitization is a neuroadaptive process characterized by an increase in a particular behavior after repeated exposure to drugs or other stimuli, such as stress. Sensitization can also be extended to neurochemical and neuroendocrine sensitization. Several factors can influence sensitization to the effects of ethanol. For instance, stress is an important component in addiction that can strengthen ethanol-induced behaviors. In animal models, stressful situations can be induced by alterations in social aspects of the animal's environment, such as maternal separation, social conflicts, and housing conditions. Social conflict models involve acute, chronic or intermittent interaction of an animal to a conspecific and can occur at any stage of life, including preweaning, adolescence or adulthood. These events can influence ethanol-induced behavioral sensitization in different ways, such as increases in locomotion, drug reward, and drug-taking behaviors. On the other hand, environmental enrichment can produce a protective phenotype against drug-related behaviors. In this chapter, we discuss findings regarding consequences of social stress and environmental enrichment on sensitization to ethanol.
Collapse
Affiliation(s)
- Rosana Camarini
- Department of Pharmacology, Institute of Biomedical Sciences, University of São Paulo, SP, Brazil.
| | - Priscila Marianno
- Department of Pharmacology, Institute of Biomedical Sciences, University of São Paulo, SP, Brazil
| | - Mariana Rae
- Department of Pharmacology, Institute of Biomedical Sciences, University of São Paulo, SP, Brazil
| |
Collapse
|
35
|
Cocaine Promotes Coincidence Detection and Lowers Induction Threshold during Hebbian Associative Synaptic Potentiation in Prefrontal Cortex. J Neurosci 2017; 37:986-997. [PMID: 28123030 DOI: 10.1523/jneurosci.2257-16.2016] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2016] [Revised: 11/21/2016] [Accepted: 12/10/2016] [Indexed: 11/21/2022] Open
Abstract
Addictive drugs usurp neural plasticity mechanisms that normally serve reward-related learning and memory, primarily by evoking changes in glutamatergic synaptic strength in the mesocorticolimbic dopamine circuitry. Here, we show that repeated cocaine exposure in vivo does not alter synaptic strength in the mouse prefrontal cortex during an early period of withdrawal, but instead modifies a Hebbian quantitative synaptic learning rule by broadening the temporal window and lowers the induction threshold for spike-timing-dependent LTP (t-LTP). After repeated, but not single, daily cocaine injections, t-LTP in layer V pyramidal neurons is induced at +30 ms, a normally ineffective timing interval for t-LTP induction in saline-exposed mice. This cocaine-induced, extended-timing t-LTP lasts for ∼1 week after terminating cocaine and is accompanied by an increased susceptibility to potentiation by fewer pre-post spike pairs, indicating a reduced t-LTP induction threshold. Basal synaptic strength and the maximal attainable t-LTP magnitude remain unchanged after cocaine exposure. We further show that the cocaine facilitation of t-LTP induction is caused by sensitized D1-cAMP/protein kinase A dopamine signaling in pyramidal neurons, which then pathologically recruits voltage-gated l-type Ca2+ channels that synergize with GluN2A-containing NMDA receptors to drive t-LTP at extended timing. Our results illustrate a mechanism by which cocaine, acting on a key neuromodulation pathway, modifies the coincidence detection window during Hebbian plasticity to facilitate associative synaptic potentiation in prefrontal excitatory circuits. By modifying rules that govern activity-dependent synaptic plasticity, addictive drugs can derail the experience-driven neural circuit remodeling process important for executive control of reward and addiction. SIGNIFICANCE STATEMENT It is believed that addictive drugs often render an addict's brain reward system hypersensitive, leaving the individual more susceptible to relapse. We found that repeated cocaine exposure alters a Hebbian associative synaptic learning rule that governs activity-dependent synaptic plasticity in the mouse prefrontal cortex, characterized by a broader temporal window and a lower threshold for spike-timing-dependent LTP (t-LTP), a cellular form of learning and memory. This rule change is caused by cocaine-exacerbated D1-cAMP/protein kinase A dopamine signaling in pyramidal neurons that in turn pathologically recruits l-type Ca2+ channels to facilitate coincidence detection during t-LTP induction. Our study provides novel insights on how cocaine, even with only brief exposure, may prime neural circuits for subsequent experience-dependent remodeling that may underlie certain addictive behavior.
Collapse
|
36
|
Zhu F, Wu Q, Li J, Grycel K, Liu B, Sun X, Zhou L, Jiao R, Song R, Khan YM, Wang Q, Wang L, Xu Y, Li J, Zhang B, Zhou Z. A single dose of cocaine potentiates glutamatergic synaptic transmission onto locus coeruleus neurons. Cell Calcium 2017; 67:11-20. [PMID: 29029785 DOI: 10.1016/j.ceca.2017.07.007] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2017] [Revised: 07/11/2017] [Accepted: 07/24/2017] [Indexed: 11/15/2022]
Abstract
The brainstem locus coeruleus (LC), the primary norepinephrinergic (NE) nucleus in the brain, has been implicated in the abuse of drugs such as opioids. However, whether and how the LC-NE system is involved in cocaine addiction remains elusive. Here, we demonstrated cocaine-evoked synaptic plasticity of glutamatergic transmission onto LC neurons as one of the earliest traces occurring after a single injection of cocaine. Twenty-four hours after mice were injected intraperitoneally with cocaine, the evoked α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid receptor (AMPAR) mediated synaptic transmission onto LC neurons were strongly potentiated without major effect on N-methyl-d-aspartate receptor (NMDAR) mediated synaptic transmission. Compared with saline-pretreated mice, AMPAR-mediated excitatory postsynaptic currents (EPSCs) of cocaine-pretreated mice showed a marked inward rectification, demonstrating the insertion of GluR2-lacking AMPARs to plasma membrane. In addition, the single injection of cocaine did not affect presynaptic glutamate release probability measured by paired pulse ratio. Furthermore, we found that the cocaine-induced potentiation of AMPAR EPSCs could be blocked by prazosin, an inhibitor of α1-adrenoreceptor (AR), indicating that cocaine increases AMPAR transmission via α1-ARs. These results reveal that LC-NE serves as an initial target of drug intake.
Collapse
Affiliation(s)
- Feipeng Zhu
- State Key Laboratory of Membrane Biology and Beijing Key Laboratory of Cardiometabolic Molecular Medicine, Institute of Molecular Medicine and Peking-Tsinghua Center for Life Sciences and PKU-IDG/McGovern Institute for Brain Research, Peking University, Beijing 100871, China.
| | - Qihui Wu
- State Key Laboratory of Membrane Biology and Beijing Key Laboratory of Cardiometabolic Molecular Medicine, Institute of Molecular Medicine and Peking-Tsinghua Center for Life Sciences and PKU-IDG/McGovern Institute for Brain Research, Peking University, Beijing 100871, China
| | - Jie Li
- State Key Laboratory of Membrane Biology and Beijing Key Laboratory of Cardiometabolic Molecular Medicine, Institute of Molecular Medicine and Peking-Tsinghua Center for Life Sciences and PKU-IDG/McGovern Institute for Brain Research, Peking University, Beijing 100871, China
| | - Katarzyna Grycel
- State Key Laboratory of Membrane Biology and Beijing Key Laboratory of Cardiometabolic Molecular Medicine, Institute of Molecular Medicine and Peking-Tsinghua Center for Life Sciences and PKU-IDG/McGovern Institute for Brain Research, Peking University, Beijing 100871, China
| | - Bing Liu
- State Key Laboratory of Membrane Biology and Beijing Key Laboratory of Cardiometabolic Molecular Medicine, Institute of Molecular Medicine and Peking-Tsinghua Center for Life Sciences and PKU-IDG/McGovern Institute for Brain Research, Peking University, Beijing 100871, China
| | - Xiaoxuan Sun
- State Key Laboratory of Membrane Biology and Beijing Key Laboratory of Cardiometabolic Molecular Medicine, Institute of Molecular Medicine and Peking-Tsinghua Center for Life Sciences and PKU-IDG/McGovern Institute for Brain Research, Peking University, Beijing 100871, China
| | - Li Zhou
- State Key Laboratory of Membrane Biology and Beijing Key Laboratory of Cardiometabolic Molecular Medicine, Institute of Molecular Medicine and Peking-Tsinghua Center for Life Sciences and PKU-IDG/McGovern Institute for Brain Research, Peking University, Beijing 100871, China
| | - Ruiyin Jiao
- State Key Laboratory of Membrane Biology and Beijing Key Laboratory of Cardiometabolic Molecular Medicine, Institute of Molecular Medicine and Peking-Tsinghua Center for Life Sciences and PKU-IDG/McGovern Institute for Brain Research, Peking University, Beijing 100871, China
| | - Rui Song
- Beijing Institute of Pharmacology and Toxicology, Beijing 100850, China
| | - Younus M Khan
- State Key Laboratory of Membrane Biology and Beijing Key Laboratory of Cardiometabolic Molecular Medicine, Institute of Molecular Medicine and Peking-Tsinghua Center for Life Sciences and PKU-IDG/McGovern Institute for Brain Research, Peking University, Beijing 100871, China
| | - Qinglong Wang
- State Key Laboratory of Membrane Biology and Beijing Key Laboratory of Cardiometabolic Molecular Medicine, Institute of Molecular Medicine and Peking-Tsinghua Center for Life Sciences and PKU-IDG/McGovern Institute for Brain Research, Peking University, Beijing 100871, China
| | - Lun Wang
- State Key Laboratory of Membrane Biology and Beijing Key Laboratory of Cardiometabolic Molecular Medicine, Institute of Molecular Medicine and Peking-Tsinghua Center for Life Sciences and PKU-IDG/McGovern Institute for Brain Research, Peking University, Beijing 100871, China
| | - Yongxin Xu
- State Key Laboratory of Membrane Biology and Beijing Key Laboratory of Cardiometabolic Molecular Medicine, Institute of Molecular Medicine and Peking-Tsinghua Center for Life Sciences and PKU-IDG/McGovern Institute for Brain Research, Peking University, Beijing 100871, China
| | - Jin Li
- Beijing Institute of Pharmacology and Toxicology, Beijing 100850, China
| | - Bo Zhang
- State Key Laboratory of Membrane Biology and Beijing Key Laboratory of Cardiometabolic Molecular Medicine, Institute of Molecular Medicine and Peking-Tsinghua Center for Life Sciences and PKU-IDG/McGovern Institute for Brain Research, Peking University, Beijing 100871, China
| | - Zhuan Zhou
- State Key Laboratory of Membrane Biology and Beijing Key Laboratory of Cardiometabolic Molecular Medicine, Institute of Molecular Medicine and Peking-Tsinghua Center for Life Sciences and PKU-IDG/McGovern Institute for Brain Research, Peking University, Beijing 100871, China.
| |
Collapse
|
37
|
Systemic Delivery of a Brain-Penetrant TrkB Antagonist Reduces Cocaine Self-Administration and Normalizes TrkB Signaling in the Nucleus Accumbens and Prefrontal Cortex. J Neurosci 2017; 36:8149-59. [PMID: 27488635 DOI: 10.1523/jneurosci.2711-14.2016] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2014] [Accepted: 06/10/2016] [Indexed: 12/17/2022] Open
Abstract
UNLABELLED Cocaine exposure alters brain-derived neurotrophic factor (BDNF) expression in the brain. BDNF signaling through TrkB receptors differentially modulates cocaine self-administration, depending on the brain regions involved. In the present study, we determined how brain-wide inhibition of TrkB signaling affects cocaine intake, the motivation for the drug, and reinstatement of drug taking after extinction. To overcome the inability of TrkB ligands to cross the blood-brain barrier, the TrkB antagonist cyclotraxin-B was fused to the nontoxic transduction domain of the tat protein from human immunodeficiency virus type 1 (tat-cyclotraxin-B). Intravenous injection of tat-cyclotraxin-B dose-dependently reduced cocaine intake, motivation for cocaine (as measured under a progressive ratio schedule of reinforcement), and reinstatement of cocaine taking in rats allowed either short or long access to cocaine self-administration. In contrast, the treatment did not affect operant responding for a highly palatable sweet solution, demonstrating that the effects of tat-cyclotraxin-B are specific for cocaine reinforcement. Cocaine self-administration increased TrkB signaling and activated the downstream Akt pathway in the nucleus accumbens, and had opposite effects in the prefrontal cortex. Pretreatment with tat-cyclotraxin-B normalized protein levels in these two dopamine-innervated brain regions. Cocaine self-administration also increased TrkB signaling in the ventral tegmental area, where the dopaminergic projections originate, but pretreatment with tat-cyclotraxin-B did not alter this effect. Altogether, our data show that systemic administration of a brain-penetrant TrkB antagonist leads to brain region-specific effects and may be a potential pharmacological strategy for the treatment of cocaine addiction. SIGNIFICANCE STATEMENT Brain-derived neurotrophic factor (BDNF) signaling through TrkB receptors plays a well established role in cocaine reinforcement. However, local manipulation of BDNF signaling yields divergent effects, depending on the brain region, thereby questioning the viability of systemic TrkB targeting for the treatment of cocaine use disorders. Our study provides first-time evidence that systemic administration of a brain-penetrant TrkB antagonist (tat-cyclotraxin-B) reduces several behavioral measures of cocaine dependence, without altering motor performance or reinforcement by a sweet palatable solution. In addition, although cocaine self-administration produced opposite effects on TrkB signaling in the nucleus accumbens and prefrontal cortex, tat-cyclotraxin-B administration normalized these cocaine-induced changes in both brain regions.
Collapse
|
38
|
The Small GTPase Rac1 Contributes to Extinction of Aversive Memories of Drug Withdrawal by Facilitating GABA A Receptor Endocytosis in the vmPFC. J Neurosci 2017. [PMID: 28630256 DOI: 10.1523/jneurosci.3859-16.2017] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023] Open
Abstract
Extinction of aversive memories has been a major concern in neuropsychiatric disorders, such as anxiety disorders and drug addiction. However, the mechanisms underlying extinction of aversive memories are not fully understood. Here, we report that extinction of conditioned place aversion (CPA) to naloxone-precipitated opiate withdrawal in male rats activates Rho GTPase Rac1 in the ventromedial prefrontal cortex (vmPFC) in a BDNF-dependent manner, which determines GABAA receptor (GABAAR) endocytosis via triggering synaptic translocation of activity-regulated cytoskeleton-associated protein (Arc) through facilitating actin polymerization. Active Rac1 is essential and sufficient for GABAAR endocytosis and CPA extinction. Knockdown of Rac1 expression within the vmPFC of rats using Rac1-shRNA suppressed GABAAR endocytosis and CPA extinction, whereas expression of a constitutively active form of Rac1 accelerated GABAAR endocytosis and CPA extinction. The crucial role of GABAAR endocytosis in the LTP induction and CPA extinction is evinced by the findings that blockade of GABAAR endocytosis by a dynamin function-blocking peptide (Myr-P4) abolishes LTP induction and CPA extinction. Thus, the present study provides first evidence that Rac1-dependent GABAAR endocytosis plays a crucial role in extinction of aversive memories and reveals the sequence of molecular events that contribute to learning experience modulation of synaptic GABAAR endocytosis.SIGNIFICANCE STATEMENT This study reveals that Rac1-dependent GABAAR endocytosis plays a crucial role in extinction of aversive memories associated with drug withdrawal and identifies Arc as a downstream effector of Rac1 regulations of synaptic plasticity as well as learning and memory, thereby suggesting therapeutic targets to promote extinction of the unwanted memories.
Collapse
|
39
|
Keralapurath MM, Briggs SB, Wagner JJ. Cocaine self-administration induces changes in synaptic transmission and plasticity in ventral hippocampus. Addict Biol 2017; 22:446-456. [PMID: 26692207 DOI: 10.1111/adb.12345] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2015] [Revised: 11/05/2015] [Accepted: 11/06/2015] [Indexed: 12/01/2022]
Abstract
Allowing rats extended access to cocaine self-administration is thought to recapitulate several key aspects of cocaine addiction in humans. Understanding the mechanisms that underlie drug-induced neuroadaptations that persist in the brain after protracted periods of abstinence is crucial towards the goal of developing therapeutic interventions for this disease state. We have employed both whole-cell voltage clamp and extracellular recording technique to assess changes in neurotransmission and long-term potentiation (LTP) in stratum radiatum of the CA1 region using the rat ventral hippocampal slice preparation. Rats allowed to self-administer cocaine daily, including 'long access' (6 hours) sessions, exhibited an increase in the α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid (AMPA)/N-methyl-d-aspartate current ratio and enhanced excitatory transmission following 3-5 weeks of abstinence. Inhibitory transmission was also significantly decreased in long-access animals, and the AMPA/N-methyl-d-aspartate ratio measured in the absence of GABAergic blockers was greatly enhanced. We also observed a significant reduction of LTP magnitude evoked in the long-access cocaine rats. These findings suggest the presence of synergistic effects of enhanced AMPA and diminished gamma-aminobutyric acid neurotransmission under physiological conditions in the CA1 region of cocaine-taking animals, supporting the conclusion that persisting enhancement of AMPA-mediated transmission and/or inhibition of gamma-aminobutyric acid-mediated transmission promoted a chronic state of potentiation that partially occluded further LTP. This increased output from the ventral hippocampus to other limbic areas would be among the drug-induced neuroadaptations that persist following abstinence from cocaine self-administration and therefore may contribute to the disease state of addiction.
Collapse
Affiliation(s)
- Madhusudhanan M. Keralapurath
- Department of Physiology & Pharmacology; The University of Georgia; Athens GA USA
- Interdisciplinary Toxicology Program; University of Georgia; Athens GA USA
| | - Sherri B. Briggs
- Department of Physiology & Pharmacology; The University of Georgia; Athens GA USA
- Neuroscience Program; University of Georgia; Athens GA USA
| | - John J. Wagner
- Department of Physiology & Pharmacology; The University of Georgia; Athens GA USA
- Interdisciplinary Toxicology Program; University of Georgia; Athens GA USA
- Neuroscience Program; University of Georgia; Athens GA USA
| |
Collapse
|
40
|
Vierci G, Pannunzio B, Bornia N, Rossi FM. H3 and H4 Lysine Acetylation Correlates with Developmental and Experimentally Induced Adult Experience-Dependent Plasticity in the Mouse Visual Cortex. J Exp Neurosci 2016; 10:49-64. [PMID: 27891053 PMCID: PMC5117113 DOI: 10.4137/jen.s39888] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2016] [Revised: 08/21/2016] [Accepted: 08/27/2016] [Indexed: 12/30/2022] Open
Abstract
Histone posttranslational modifications play a fundamental role in orchestrating gene expression. In this work, we analyzed the acetylation of H3 and H4 histones (AcH3-AcH4) and its modulation by visual experience in the mouse visual cortex (VC) during normal development and in two experimental conditions that restore juvenile-like plasticity levels in adults (fluoxetine treatment and enriched environment). We found that AcH3-AcH4 declines with age and is upregulated by treatments restoring plasticity in the adult. We also found that visual experience modulates AcH3-AcH4 in young and adult plasticity-restored mice but not in untreated ones. Finally, we showed that the transporter vGAT is downregulated in adult plasticity-restored models. In summary, we identified a dynamic regulation of AcH3-AcH4, which is associated with high plasticity levels and enhanced by visual experience. These data, along with recent ones, indicate H3-H4 acetylation as a central hub in the control of experience-dependent plasticity in the VC.
Collapse
Affiliation(s)
- Gabriela Vierci
- Laboratorio de Neurociencias "Neuroplasticity Unit", Facultad de Ciencias, Universidad de la República, Montevideo, Uruguay
| | - Bruno Pannunzio
- Laboratorio de Neurociencias "Neuroplasticity Unit", Facultad de Ciencias, Universidad de la República, Montevideo, Uruguay
| | - Natalia Bornia
- Laboratorio de Neurociencias "Neuroplasticity Unit", Facultad de Ciencias, Universidad de la República, Montevideo, Uruguay
| | - Francesco M Rossi
- Laboratorio de Neurociencias "Neuroplasticity Unit", Facultad de Ciencias, Universidad de la República, Montevideo, Uruguay
| |
Collapse
|
41
|
The Medial Orbitofrontal Cortex Regulates Sensitivity to Outcome Value. J Neurosci 2016; 36:4600-13. [PMID: 27098701 DOI: 10.1523/jneurosci.4253-15.2016] [Citation(s) in RCA: 67] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2015] [Accepted: 03/08/2016] [Indexed: 11/21/2022] Open
Abstract
UNLABELLED An essential component of goal-directed decision-making is the ability to maintain flexible responding based on the value of a given reward, or "reinforcer." The medial orbitofrontal cortex (mOFC), a subregion of the ventromedial prefrontal cortex, is uniquely positioned to regulate this process. We trained mice to nose poke for food reinforcers and then stimulated this region using CaMKII-driven Gs-coupled designer receptors exclusively activated by designer drugs (DREADDs). In other mice, we silenced the neuroplasticity-associated neurotrophin brain-derived neurotrophic factor (BDNF). Activation of Gs-DREADDs increased behavioral sensitivity to reinforcer devaluation, whereas Bdnf knockdown blocked sensitivity. These changes were accompanied by modifications in breakpoint ratios in a progressive ratio task, and they were recapitulated in Bdnf(+/-)mice. Replacement of BDNF selectively in the mOFC in Bdnf(+/-)mice rescued behavioral deficiencies, as well as phosphorylation of extracellular-signal regulated kinase 1/2 (ERK1/2). Thus, BDNF expression in the mOFC is both necessary and sufficient for the expression of typical effort allocation relative to an anticipated reinforcer. Additional experiments indicated that expression of the immediate-early gene c-fos was aberrantly elevated in the Bdnf(+/-)dorsal striatum, and BDNF replacement in the mOFC normalized expression. Also, systemic administration of an MAP kinase kinase inhibitor increased breakpoint ratios, whereas the addition of discrete cues bridging the response-outcome contingency rescued breakpoints in Bdnf(+/-)mice. We argue that BDNF-ERK1/2 in the mOFC is a key regulator of "online" goal-directed action selection. SIGNIFICANCE STATEMENT Goal-directed response selection often involves predicting the consequences of one's actions and the value of potential payoffs. Lesions or chemogenetic inactivation of the medial orbitofrontal cortex (mOFC) in rats induces failures in retrieving outcome identity memories (Bradfield et al., 2015), suggesting that the healthy mOFC serves to access outcome value information when it is not immediately observable and thereby guide goal-directed decision-making. Our findings suggest that the mOFC also bidirectionally regulates effort allocation for a given reward and that expression of the neurotrophin BDNF in the mOFC is both necessary and sufficient for mice to sustain stable representations of reinforcer value.
Collapse
|
42
|
Viola TW, Wearick-Silva LE, De Azeredo LA, Centeno-Silva A, Murphy C, Marshall P, Li X, Singewald N, Garcia F, Bredy TW, Grassi-Oliveira R. Increased cocaine-induced conditioned place preference during periadolescence in maternally separated male BALB/c mice: the role of cortical BDNF, microRNA-212, and MeCP2. Psychopharmacology (Berl) 2016; 233:3279-88. [PMID: 27392631 DOI: 10.1007/s00213-016-4373-z] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/11/2016] [Accepted: 06/18/2016] [Indexed: 12/15/2022]
Abstract
RATIONALE Early life stress is a major risk factor for cocaine addiction; however, the underlying molecular mechanisms remain relatively unexplored. MicroRNA-212 (miR-212) and methyl CpG binding protein 2 (MeCP2) have recently emerged as key regulators of brain-derived neurotrophic factor (BDNF) signaling during the acquisition and maintenance of cocaine-seeking behaviors. OBJECTIVES We therefore investigated the effect of maternal separation (MS) on cocaine-induced conditioned place preference (CPP) during periadolescence and how this influences miR-212, Mecp2, and Bdnf expressions in the prefrontal cortex. METHODS Male BALB/c mice subjected to MS (3 h/day) from postnatal day 2 to 15 or normal animal facility rearing (AFR) were tested for CPP at postnatal day 45, or not exposed to experimental manipulations (drug-naïve animals). Cultured primary cortical neurons were used to determine miR-212 expression changes following depolarization by KCL treatment. RESULTS MS increased cocaine-induced CPP and decreased Bdnf exon IV expression, which correlated with higher CPP scores in such animals. An experience-dependent decrease in miR-212 expression was observed following CPP test. This effect was mimicked in primary cortical neurons in vitro, under activity-dependent conditions. In contrast, increased Mecp2 expression was found after CPP test, suggesting an opposing relationship between miR-212 and Mecp2 expression following cocaine place preference acquisition. However, these effects were not present in mice exposed to MS. CONCLUSIONS Together, our results suggest that early life stress can enhance the motivational salience for cocaine-paired cues during periadolescence, and that altered expression of miR-212, Mecp2, and Bdnf in the prefrontal cortex is involved in this process.
Collapse
Affiliation(s)
- Thiago Wendt Viola
- Postgraduate Program in Pediatrics and Child Health, Pontifícia Universidade Católica do Rio Grande do Sul (PUCRS), Porto Alegre, Brazil.,Developmental Cognitive Neuroscience Lab (DCNL), Pontifícia Universidade Católica do Rio Grande do Sul (PUCRS), Avenida Ipiranga, 6681, prédio 11, sala 928, Porto Alegre, 90619-900, RS, Brazil
| | - Luis Eduardo Wearick-Silva
- Postgraduate Program in Pediatrics and Child Health, Pontifícia Universidade Católica do Rio Grande do Sul (PUCRS), Porto Alegre, Brazil.,Developmental Cognitive Neuroscience Lab (DCNL), Pontifícia Universidade Católica do Rio Grande do Sul (PUCRS), Avenida Ipiranga, 6681, prédio 11, sala 928, Porto Alegre, 90619-900, RS, Brazil
| | - Lucas Araújo De Azeredo
- Developmental Cognitive Neuroscience Lab (DCNL), Pontifícia Universidade Católica do Rio Grande do Sul (PUCRS), Avenida Ipiranga, 6681, prédio 11, sala 928, Porto Alegre, 90619-900, RS, Brazil.,Postgraduate Program in Medicine and Health Sciences, PUCRS, Porto Alegre, RS, Brazil
| | - Anderson Centeno-Silva
- Developmental Cognitive Neuroscience Lab (DCNL), Pontifícia Universidade Católica do Rio Grande do Sul (PUCRS), Avenida Ipiranga, 6681, prédio 11, sala 928, Porto Alegre, 90619-900, RS, Brazil
| | - Conor Murphy
- Department of Pharmacology and Toxicology, Institute of Pharmacy, Center for Molecular Biosciences Innsbruck, University of Innsbruck, Innsbruck, Austria
| | - Paul Marshall
- Department of Neurobiology and Behavior, Center for the Neurobiology of Learning and Memory, University of California-Irvine, Irvine, 92697, CA, USA
| | - Xiang Li
- Department of Neurobiology and Behavior, Center for the Neurobiology of Learning and Memory, University of California-Irvine, Irvine, 92697, CA, USA
| | - Nicolas Singewald
- Department of Pharmacology and Toxicology, Institute of Pharmacy, Center for Molecular Biosciences Innsbruck, University of Innsbruck, Innsbruck, Austria
| | - Frederico Garcia
- Department of Psychiatry, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Timothy W Bredy
- Department of Neurobiology and Behavior, Center for the Neurobiology of Learning and Memory, University of California-Irvine, Irvine, 92697, CA, USA.,Queensland Brain Institute, The University of Queensland, Brisbane, QLD, Australia
| | - Rodrigo Grassi-Oliveira
- Postgraduate Program in Pediatrics and Child Health, Pontifícia Universidade Católica do Rio Grande do Sul (PUCRS), Porto Alegre, Brazil. .,Developmental Cognitive Neuroscience Lab (DCNL), Pontifícia Universidade Católica do Rio Grande do Sul (PUCRS), Avenida Ipiranga, 6681, prédio 11, sala 928, Porto Alegre, 90619-900, RS, Brazil.
| |
Collapse
|
43
|
Mulholland PJ, Chandler LJ, Kalivas PW. Signals from the Fourth Dimension Regulate Drug Relapse. Trends Neurosci 2016; 39:472-485. [PMID: 27173064 PMCID: PMC4930682 DOI: 10.1016/j.tins.2016.04.007] [Citation(s) in RCA: 52] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2016] [Revised: 04/12/2016] [Accepted: 04/14/2016] [Indexed: 12/21/2022]
Abstract
Despite the enormous societal burden of alcohol and drug addiction and abundant research describing drug-induced maladaptive synaptic plasticity, there are few effective strategies for treating substance use disorders. Recent awareness that synaptic plasticity involves astroglia and the extracellular matrix is revealing new possibilities for understanding and treating addiction. We first review constitutive corticostriatal adaptations that are elicited by and shared between all abused drugs from the perspective of tetrapartite synapses, and integrate recent discoveries regarding cell type-specificity in striatal neurons. Next, we describe recent discoveries that drug-seeking is associated with transient synaptic plasticity that requires all four synaptic elements and is shared across drug classes. Finally, we prognosticate how considering tetrapartite synapses can provide new treatment strategies for addiction.
Collapse
Affiliation(s)
- Patrick J Mulholland
- Department of Neuroscience, Medical University of South Carolina, 173 Ashley Avenue, Charleston, SC 29425, USA; Department of Psychiatry and Behavioral Sciences, Addiction Sciences Division, Medical University of South Carolina, 67 President Street, Charleston, SC, 29425, USA.
| | - L Judson Chandler
- Department of Neuroscience, Medical University of South Carolina, 173 Ashley Avenue, Charleston, SC 29425, USA; Department of Psychiatry and Behavioral Sciences, Addiction Sciences Division, Medical University of South Carolina, 67 President Street, Charleston, SC, 29425, USA
| | - Peter W Kalivas
- Department of Neuroscience, Medical University of South Carolina, 173 Ashley Avenue, Charleston, SC 29425, USA.
| |
Collapse
|
44
|
Pitts EG, Taylor JR, Gourley SL. Prefrontal cortical BDNF: A regulatory key in cocaine- and food-reinforced behaviors. Neurobiol Dis 2016; 91:326-35. [PMID: 26923993 PMCID: PMC4913044 DOI: 10.1016/j.nbd.2016.02.021] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2015] [Revised: 02/20/2016] [Accepted: 02/24/2016] [Indexed: 12/21/2022] Open
Abstract
Brain-derived neurotrophic factor (BDNF) affects synaptic plasticity and neural structure and plays key roles in learning and memory processes. Recent evidence also points to important, yet complex, roles for BDNF in rodent models of cocaine abuse and addiction. Here we examine the role of prefrontal cortical (PFC) BDNF in reward-related decision making and behavioral sensitivity to, and responding for, cocaine. We focus on BDNF within the medial and orbital PFC, its regulation by cocaine during early postnatal development and in adulthood, and how BDNF in turn influences responding for drug reinforcement, including in reinstatement models. When relevant, we draw comparisons and contrasts with experiments using natural (food) reinforcers. We also summarize findings supporting, or refuting, the possibility that BDNF in the medial and orbital PFC regulate the development and maintenance of stimulus-response habits. Further investigation could assist in the development of novel treatment approaches for cocaine use disorders.
Collapse
Affiliation(s)
- Elizabeth G Pitts
- Graduate Program in Neuroscience, Yerkes National Primate Research Center, Emory University, Atlanta, GA, United States
| | - Jane R Taylor
- Department of Psychiatry, Yale University School of Medicine, New Haven, CT, United States; Interdepartmental Neuroscience Program, Department of Psychology, Yale University, New Haven, CT, United States
| | - Shannon L Gourley
- Department of Pediatrics, Emory University School of Medicine, Atlanta, GA, United States; Department of Psychiatry and Behavioral Sciences, Emory University School of Medicine, Atlanta, GA, United States; Graduate Program in Neuroscience, Yerkes National Primate Research Center, Emory University, Atlanta, GA, United States.
| |
Collapse
|
45
|
Ju YY, Long JD, Liu Y, Liu JG. Formation of aversive memories associated with conditioned drug withdrawal requires BDNF expression in the amygdala in acute morphine-dependent rats. Acta Pharmacol Sin 2015; 36:1437-43. [PMID: 26567727 DOI: 10.1038/aps.2015.94] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/27/2015] [Accepted: 08/14/2015] [Indexed: 01/14/2023]
Abstract
AIM Brain-derived neurotrophic factor (BDNF) plays an important role in learning and memory in multiple brain areas. In the present study, we investigated the roles of BDNF in aversive memories associated with conditioned drug withdrawal in acute morphine-dependent rats. METHODS Conditioned place aversion (CPA) was induced in male SD rats exposed to a single dose of morphine (10 mg/kg, sc) followed by naloxone (0.3 mg/kg, sc). In some rats, BDNF receptor antagonist K252a (8.5 ng per side) or BDNF scavenger TrkB-FC (0.65 μg per side) was bilaterally microinjected into amygdala before naloxone injection. BDNF mRNA and protein expression levels in amygdala were detected after the behavior testing. RESULTS CPA behavior was induced in rats by the naloxone-precipitated morphine withdrawal, which was accompanied by significantly increased levels of BDNF mRNA and protein in the amygdala. Bilateral microinjection of TrkB-FC or K252a into the amygdala completely blocked CPA behavior in the rats. CONCLUSION Formation of aversive memories associated with conditioned drug withdrawal in acute morphine-dependent rats requires BDNF expression in the amygdala.
Collapse
|
46
|
Neurophysiological and Neurochemical Mechanisms Underlying Depression Disorders and Search for New Directions of Treatment. NEUROPHYSIOLOGY+ 2015. [DOI: 10.1007/s11062-015-9542-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/22/2022]
|
47
|
d-amphetamine withdrawal-induced decreases in brain-derived neurotrophic factor in sprague-dawley rats are reversed by treatment with ketamine. Neuropharmacology 2015; 97:7-17. [DOI: 10.1016/j.neuropharm.2015.04.023] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2015] [Revised: 04/24/2015] [Accepted: 04/26/2015] [Indexed: 12/16/2022]
|
48
|
BDNF interacts with endocannabinoids to regulate cocaine-induced synaptic plasticity in mouse midbrain dopamine neurons. J Neurosci 2015; 35:4469-81. [PMID: 25762688 DOI: 10.1523/jneurosci.2924-14.2015] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Brain-derived neurotrophic factor (BDNF) and endocannabinoids (eCBs) have been individually implicated in behavioral effects of cocaine. The present study examined how BDNF-eCB interaction regulates cocaine-induced synaptic plasticity in the ventral tegmental area and behavioral effects. We report that BDNF and selective tyrosine kinase receptor B (TrkB) agonist 7,8-dihydroxyflavone (DHF) activated the TrkB receptor to facilitate two forms of eCB-mediated synaptic depression, depolarization-induced suppression of inhibition (DSI), and long-term depression (I-LTD) of IPSCs in ventral tegmental area dopamine neurons in mouse midbrain slices. The facilitation appears to be mediated by an increase in eCB production via phospholipase Cγ pathway, but not by an increase in CB1 receptor responsiveness or a decrease in eCB hydrolysis. Using Cre-loxP technology to specifically delete BDNF in dopamine neurons, we showed that eCB-mediated I-LTD, cocaine-induced reduction of GABAergic inhibition, and potentiation of glutamatergic excitation remained intact in wild-type control mice, but were impaired in BDNF conditional knock-out mice. We also showed that cocaine-induced conditioned place preference was attenuated in BDNF conditional knock-out mice, in vivo pretreatments with DHF before place conditioning restored cocaine conditioned place preference in these mice, and the behavioral effect of DHF was blocked by a CB₁ receptor antagonist. Together, these results suggest that BDNF in dopamine neurons regulates eCB responses, cocaine-induced synaptic plasticity, and associative learning.
Collapse
|
49
|
Abstract
The endogenous cannabinoid (endocannabinoid) system is an important regulator of synaptic function. Endocannabinoids acutely modulate inhibitory and excitatory transmission, and also mediate long-term depression at GABAergic and glutamatergic synapses. Typically, endocannabinoid synthesis and release is stimulated by depolarization-induced calcium influx and/or activation of phospholipase-C (PLC) signaling triggered by mGluR activation. Recently it has been shown that brain-derived neurotrophic factor (BDNF) can also induce endocannabinoid release. Although there is growing evidence for cross-talk between BDNF and endocannabinoid signaling, little is known about the functional relevance of these interactions. In the present studies, we examined BDNF - endocannabinoid interactions in regulating activity-dependent long-term depression at inhibitory synapses (iLTD). We found that theta burst stimulation (TBS) in layer 2/3 of mouse somatosensory cortical slices can induce a form of endocannabinoid-mediated iLTD that is independent of metabotropic glutamate receptor (mGluR) activation. This endocannabinoid-dependent iLTD, however, requires endogenous BDNF-trkB signaling, as it is blocked by a trk tyrosine kinase inhibitor and by a trkB receptor antagonist, and also requires activation of diacylglycerol lipase (DAG-lipase, DGL). In addition, endocannabinoid-mediated iLTD can be induced by combining a subthreshold concentration of exogenous BDNF with weak TBS stimulation that by itself is insufficient to induce iLTD. Taken together, our results suggest that TBS can induce the release of endogenous BDNF, which triggers DGL-dependent endocannabinoid mobilization and cannabinoid receptor-dependent iLTD at layer 2/3 cortical synapses.
Collapse
|
50
|
GABAAα1-mediated plasticity in the orbitofrontal cortex regulates context-dependent action selection. Neuropsychopharmacology 2015; 40:1027-36. [PMID: 25348603 PMCID: PMC4330518 DOI: 10.1038/npp.2014.292] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/30/2014] [Revised: 10/21/2014] [Accepted: 10/22/2014] [Indexed: 01/04/2023]
Abstract
An essential aspect of goal-directed action selection is differentiating between behaviors that are more, or less, likely to be reinforced. Habits, by contrast, are stimulus-elicited behaviors insensitive to action-outcome contingencies and are considered an etiological factor in several neuropsychiatric disorders. Thus, isolating the neuroanatomy and neurobiology of goal-directed action selection on the one hand, and habit formation on the other, is critical. Using in vivo viral-mediated gene silencing, we knocked down Gabra1 in the orbitofrontal prefrontal cortex (oPFC) in mice, decreasing oPFC GABAAα1 expression, as well as expression of the synaptic marker PSD-95. Mice expressing Green Fluorescent Protein or Gabra1 knockdown in the adjacent M2 motor cortex served as comparison groups. Using instrumental response training followed by action-outcome contingency degradation, we then found that oPFC GABAAα1 deficiency impaired animals' ability to differentiate between actions that were more or less likely to be reinforced, though sensitivity to outcome devaluation and extinction were intact. Meanwhile, M2 GABAAα1 deficiency enhanced sensitivity to action-outcome relationships. Behavioral abnormalities following oPFC GABAAα1 knockdown were rescued by testing mice in a distinct context relative to that in which they had been initially trained. Together, our findings corroborate evidence that chronic GABAAα1 deficiency remodels cortical synapses and suggest that neuroplasticity within the healthy oPFC gates the influence of reward-related contextual stimuli. These stimuli might otherwise promote maladaptive habit-based behavioral response strategies that contribute to-or exacerbate-neuropsychiatric illness.
Collapse
|